Next Article in Journal
Cytoplasmic Expression of TP53INP2 Modulated by Demethylase FTO and Mutant NPM1 Promotes Autophagy in Leukemia Cells
Next Article in Special Issue
Quercetin 3-O-Galactoside Isolated from Limonium tetragonum Inhibits Melanogenesis by Regulating PKA/MITF Signaling and ERK Activation
Previous Article in Journal
Genome-Wide Identification of the Odorant Receptor Gene Family and Revealing Key Genes Involved in Sexual Communication in Anoplophora glabripennis
Previous Article in Special Issue
Silibinin Suppresses the Hyperlipidemic Effects of the ALK-Tyrosine Kinase Inhibitor Lorlatinib in Hepatic Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Corydalis saxicola Bunting: A Review of Its Traditional Uses, Phytochemistry, Pharmacology, and Clinical Applications

1
State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin 541004, China
2
Ministry of Education Key Laboratory for Ecology of Rare and Endangered Species and Environmental Protection, School of Life Sciences, Guangxi Normal University, Guilin 541004, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(2), 1626; https://doi.org/10.3390/ijms24021626
Submission received: 5 September 2022 / Revised: 5 December 2022 / Accepted: 10 December 2022 / Published: 13 January 2023

Abstract

:
Corydalis saxicola Bunting (CSB), whose common name in Chinese is Yanhuanglian, is a herb in the family Papaveraceae. When applied in traditional Chinese medicine, it is used to treat various diseases including hepatitis, abdominal pain, and bleeding haemorrhoids. In addition, Corydalis saxicola Bunting injection (CSBI) is widely used against acute and chronic hepatitis. This review aims to provide up-to-date information on the botanical distribution, description, traditional uses, phytochemistry, pharmacology, and clinical applications of CSB. A comprehensive review was implemented on studies about CSB from several scientific databases, such as SciFinder, Elsevier, Springer, ACS Publications, Baidu Scholar, CNKI, and Wanfang Data. Phytochemical studies showed that 81 chemical constituents have been isolated and identified from CSB, most of which are alkaloids. This situation indicates that these alkaloids would be the main bioactive substances and that they have antitumour, liver protective, antiviral, and antibacterial pharmacological activities. CSBI can not only treat hepatitis and liver cancer but can also be used in combination with other drugs. However, the relationships between the traditional uses and modern pharmacological actions, the action mechanisms, quality standards, and the material basis need to be implemented in the future. Moreover, the pharmacokinetics of CSBI in vivo and the toxicology should be further investigated.

1. Introduction

Corydalis saxicola Bunting (CSB), which is called Yanhuanglian in Chinese, is a species of Corydalis DC., is a genus in the family Papaveraceae. CSB is mainly distributed on rocky cliffs or in alpine caves in southwestern China, including Guangxi, Guizhou, Hubei, Shanxi, Sichuan, Yunnan, Zhejiang [1]. CSB is a traditional Chinese folk medicine in southwestern China, where the bitter-tasting and cool-natured whole plant can be used as a medicine that has the effects of heat clearing and detoxification, damp elimination, pain alleviation, and haemostasis [2]. In tradition, the whole plant of CSB, namely Yan-huang-lian is used to cure diseases in Chinese folk. On the basis of traditional Chinese medicine theories, its use spectrum contains acute conjunctivitis, corneal pannus, acute abdominal pain, hemorrhoidal bleeding, haematochezia, swelling, hepatitis, cirrhosis, and liver cancer [3,4,5]. Until now, pharmacological studies have provided evidence that this ethnomedicine can treat liver diseases and show that it also has significant pharmacological properties such as anticancer, anti-inflammatory, antibacterial, anti-oxidative, analgesic, and hepatoprotective effects [6,7,8]. Clinically, Corydalis saxicola Bunting injection (CSBI) was approved for production in Guangxi in 1982 [1] and is the exclusive variety in production in China. In 2002, the national medicine standard (Z20026725) was obtained by landmark upgrading and national standard rectification. CSBI is clinically used for patients with acute and chronic hepatitis.
Although a large number of studies have been carried out on CSB, many aspects of CSB remain unclear, such as monomer component exploration, the establishment of quality standards, the mechanism of CSB therapeutic effects, and combination mechanism. Additionally, toxicological studies need to be strengthened to support its therapeutic safety. In this paper, the botanical properties, traditional uses, phytochemistry, pharmacological activities, and clinical applications of CSB are reviewed. This review can provide a reference for deep research and application of this plant.

2. Database Search Method

This review of CBS on its botanical distribution and description, traditional uses, phytochemistry, pharmacological activity, clinical applications, and toxicity assessment is based on a variety of reliable databases such as Scifinder, PubMed, ScienceDirect, Wiley, ACS, CNKI, Springer, Google Scholar, Web of Science, and Baidu Scholar, as well as other published materials (Ph.D. and M.Sc. dissertations and books). The literature was searched and accessed using the keywords “Corydalis saxicola Bunting”, “Corydalis saxicola Bunting total alkaloids”, and “dehydrocavidine” which are related to the present review.

3. Botanical Distribution and Description

CSB can be found in southwestern China with a wide distribution in Chongqing (Chengkou), Guangxi (Debao, Fengshan, Jingxi), Guizhou (Dushan, Weng’an, Zunyi), Hubei (Yichang), Shanxi (Mianxian), Sichuan, Yunnan (Xichou), and Zhejiang (Ningbo), among other areas [9,10]. Notably, it is mainly distributed in western and northwestern Guangxi, especially Donglan, Bama, Du’an, Jingxi, and Debao. It has stringent requirements for environmental conditions and is produced in extremely low yield. The distribution of this plant is limited to limestone mountain areas and is a stone mountain endemic species. It grows primarily on rocky cliffs or in alpine caves at elevations of 600–1690 m, reaching 2800–3900 m in southwestern Sichuan, with a growth temperature of 15–25 °C [10,11,12].
CSB is a light green soft herb that can grow up to 30–40 cm high and has thick main roots and single to multi-headed rhizomes. The stems can be branched or unbranched. The branches are opposite to the leaves and are scape-shaped. The basal leaves are approximately 10–15 cm long with a long stalk, the leaves are approximately the same length as the petioles with two or one pinnate cleft, and the last pinnate is wedge-shaped to obovate, approximately 2–4 cm long and 2–3 cm wide, with unequal 2–3 splits or thick round teeth at the edges. The raceme is approximately 7–15 cm long, with many flowers that are first dense, then alienated. The bracts are elliptic to lanceolate, whole, and the lower part is approximately 1.5 cm long and 1 cm wide while the upper part narrows, all of which are longer than the pedicels. The length of the pedicel is approximately 5 mm. The flowers are golden yellow and flat. The sepals are like triangles, and in their entirety are approximately 2 mm long. The outer petals are wider and acuminate, and the coronal process is limited to the keel process but does not reach the top. The upper petals are approximately 2.5 cm long with the distance accounting for approximately 1/4 of the full length of the petals, slightly bent down, and the end is cystic. The lower petals are approximately 1.8 cm long, and the base looks like a small tumour protrusion. The inner petals are approximately 1.5 cm long, with a thick crown protruding from the top. The stamen bundles are lanceolate, tapering above the middle. The stigma is two-forked, with two cleft papillae at the top of each branch. The capsule is linear and recurved, approximately 2.5 cm long, with 1 row of seeds [10]. (see Figure 1).

4. Traditional Uses

As a type of folk medicinal herb in South China, various classical ancient Chinese books, including “Guizhou folk medicine”, “Traditional Chinese Medicine Dictionary”, “Chinese Materia Medica”, “Flora of Guangxi”, and “Flora of Yunnan”, have recorded the traditional medicinal uses of CSB. Based on the theory of traditional Chinese medicine, CSB has been widely used in the treatment of various diseases for a long time, such as heat clearance and detoxification, damp elimination, pain alleviation, haemostasis, erosion of the mucous membranes in the oral cavity, dysentery, eyes of fire, eye shade, diarrhoea, stomachache, bleeding haemorrhoids, hepatitis, jaundice, ascites, cirrhosis, and hepatoma [13,14,15,16]. For example, direct oral administration of 6 g of CSB is used to treat acute abdominal pain. For the treatment of pile ale bleeding and red dysentery, 15 g of CSB soaked in 50 g of steamed liquor is taken orally. In addition, CSB can be mixed with other medicines to treat diseases. For example, for the treatment of eyes of fire and eye shade, CSB (3 g), Gentiana scabra Bunge (3 g), and “Shang Mei Pian” (1.5 g) are ground together into powder, steamed in a porcelain cup, and finally dipped into the eye with a lantern. An injection containing the total alkaloids of CSB has a good effect on hepatitis, cirrhosis, and hepatoma [12,17]. CSB is mainly used orally (decoction, 3–15 g) and externally (put on the affected area after grinding in the appropriate amount). When using CSB, dry and spicy foods should be avoided. Due to diverse traditional and folk uses of it, CSB has been extensively studied by numerous chemists and biologists.

5. Phytochemistry

A research team from Guangxi in China reported on CSB in 1980. To date, 81 chemical components including alkaloids, steroids, and flavonoids, have been separated and identified from CSB (Table 1). The chemical structures of these 81 compounds are shown in Figure 2.

5.1. Alkaloids

Alkaloids are key secondary metabolites of CSB. Due to the diversity of their structures and biological activities, an increasing number of studies on alkaloids have been conducted in China in recent decades. To date, 67 kinds of alkaloids have been identified and reported in CSB, such as berberines, protoberberines, benzophenanthridines, protoberberines, benzyltetrahydroisoquinolines, aporphines, protopines, and lignin amides (167) (Table 1). Six alkaloids with high purity were obtained from the crude n-butanol extract of CSB by high-speed counter-current chromatography: (−)-scoulerine (30), (+)-isocorydine (50), dehydrocheilanthifoline (8), dehydrocavidine (1), palmatine (9), and berberine (4) [24]. To investigate the phytochemical composition of CSB, the whole plant is usually selected as the source. As a rapid separation strategy, high-speed counter-current chromatography is the major majority approach to the isolation of the main components of it. The results basically show that alkaloids are the main components of CSB and the content of dehydrocavidine is the highest one.

5.2. Others

With the exception of alkaloids, 14 non-alkaloids were isolated and identified from CSB, including 9 steroids (6876), 2 flavonoids (7778), 2 alkanes (7980), and 1 uracil (81) [22,23]. The research on other types of natural products is rarely performed apart from the alkaloids. Therefore, it is urgent to explore novel leading compound with biological activity in future discovery.

6. Pharmacology

In recent decades, modern pharmacological studies have shown that CSB has a wide range of pharmacological activities, such as anticancer, hepatoprotective, anti-hepatitis B virus (HBV), central nervous system, immune function enhancement, anti-inflammatory, analgesic, antioxidant, antibacterial, and choleretic effects. A large number of studies have reported that the crude extracts and chemical constituents of CSB have strong biological activity both in vivo and in vitro. In addition, research on the activity of CSB has mainly focused on alkaloids. The pharmacological effects of CSB reported in recent years are summarized in Table 2.

6.1. Anticancer Activity

6.1.1. Crude Extracts

Cancer is still the leading cause of mortality among humans. Many parts of the body can trigger the growth of cancer cells, and the emergence of cancer poses a very large threat to human health. Recent studies have provided evidence that the anticancer activity of CSB is mainly related to its alkaloid compounds. Corydalis saxicola Bunting total alkaloids (CSBTA) has shown cytotoxicity against five cancer cell lines: CNE-1 (IC50 = 112.41 µg/mL), CNE-2 (IC50 = 123.46 µg/mL), A2780 (IC50 = 148.40 µg/mL), SKOV3 (IC50 = 128.51 µg/mL), and PM2 (IC50 = 166.66 µg/mL) [23].
CSB extracts can increase actin filament disassembly and cofilin-1 activity, and decrease Cdc42 protein expression in A549 cells to reduce the migration ability of cells [27]. Additionally, CSBTA inhibits migration of A549 cells by suppressing cell division cycle 42 or vav guanine nucleotide exchange factor 1 [8]. Another study found that the effects of CSBTA on the proliferation of A549 lung cancer cells displayed a time–dose relationship; with increasing concentration of CSBTA, the apoptosis rate of A549 cells increased, the expression of caspase-3 mRNA was upregulated, and the expression of survivin mRNA was downregulated [28]. Moreover, CSBTA combined with cisplatin can enhance the inhibition of proliferation of A549 cells [29].
A study confirmed that CSBTA inhibited the growth of A549 cell-transplanted tumours by downregulating survivin in nude mice, and it can also reduce the degree of bone destruction in bone metastases [26]. Furthermore, CSBTA attenuate Walker 256-induced bone pain and osteoporosis by suppressing the RANKL-induced NF-κB and c-Fos/NFATc1 pathways in rats [25].
The above five studies reported the activity of CSBTA on A549 cells, while there have also been studies on CSBTA in Tca8113 cells. CSBTA can significantly inhibit the proliferation and apoptosis of human tongue squamous cell carcinoma Tca8113 cells in a time-dependent and dose-dependent manner [31]. Additionally, [30] found that CSBTA could promote apoptosis of Tca8113 cells by inhibiting the expression of Bcl-2 at the mRNA and protein levels. [32]. Overall, as a leading compound targeting tumour pathway, CSBTA is promising for developing into a novel anticancer drug.
CSBI has been reported to exert a certain inhibitory effect on mouse S180 sarcoma and Ehrlich ascites tumours, with an average inhibition rate of more than 30%. In addition, the combination of CSBI and phagocytic acid can enhance the antitumour effects, with the average tumour inhibition rate reaching over 50% [33]. The antitumour activity of CSBTA was further evaluated through in vitro, semi-in vivo, in vivo, methylene blue reduction, and cell staining methods, and the results showed that CSBTA had a certain inhibitory effect on sarcoma 180 Ehrlich ascites carcinoma in mice and sarcoma 256 in rats [17,34,35]. Lu’s test reported that CSBTA could inhibit respiratory metabolism in S180, HAC, and EAC tumour cells [34]. The water extract of CSB can inhibit the proliferation and migration of HepG2 liver cancer cells, and its mechanism may be related to the upregulation of NF-κB p65 expression [25]. Therefore, the water extract of CSB and CSBTA has excellent antitumour activity against a variety of tumour cells. However, what specific components take effect in the water extract is still unclear and needs to be further explored.

6.1.2. Isolated Phytochemicals

Eight lignanamides (corydalisin A (58), corydalisin B (59), corydalisin C (61), cannabisin F (60), cannabisin E (64), cannabisin D (62), 1,2-dihydro-6,8-dimethoxy-7-hydroxy-1-(3,5-dimethoxy-4-hydroxyphenyl)-N1,N2-bis-[2-(4-hydroxyphenyl)ethyl]-2,3-naphthalene dicarboxamide (63), and grossamide (65)) exhibited anticancer activity against five tumour cell lines (MGC-803, HepG2, T24, NCI-H460, and Spca-2; IC50 > 8.81 ± 2.05 μM). Meanwhile, corydalisin C may induce apoptosis via both the intrinsic and extrinsic apoptosis pathways and downregulation of Bcl-2 and FasL expression in a time-dependent manner [1]. Dehydrocavidine (1) and palmatine (9) can inhibit the proliferation of hepatocellular carcinoma SMMC-7721 cells [37]. Dehydrocavidine (1) significantly inhibited the proliferation of human tongue squamous cell carcinoma Tca8113 cells and decreased the expression of NF-κB, telomerase activity and the expression of hTERT [38,39]. (−)-Pallidine (51) and (−)-scoulerine (30) have strong inhibitory effects on human DNA topoisomerase I [19]. Chelerythrine (15) and (−)-13β-hydroxystylopine (23) have certain anticancer effects [21]. Corysaxicolaine A (67) displayed obvious inhibitory effects on the tested human cancer cells (T24, A549, HepG2, MGC-803, SKOV3), with IC50 values of 7.63, 13.32, 12.39, 9.98, and 12.36 μM, respectively [7]. However, there is little research on the mechanisms of these individual compounds or their anticancer activity in vivo. Elucidating the anticancer activity and mechanism of CSBTA and its individual compounds should be the focus of future research.

6.2. Hepatoprotective Effects

6.2.1. Crude Extracts

A large number of studies have reported that CSB plays a pivotal role in the treatment of acute and chronic liver injury, liver fibrosis, and liver cirrhosis. Some researchers found that the expression levels of ALT, AST, MDA, and ALP in serum were significantly decreased, and ALB, SOD, and GSH was significantly increased in liver tissue of rats with acute and chronic liver injury after CSBTA intervention [20,39,40,41,45,46,48,49,50,51]. Moreover, CSBTA has an obvious protective effect on acute liver injury caused by CCl4 and can also inhibit the formation of chronic liver fibrosis in rats. The potential mechanisms for this protective effect may be related to anti-lipid peroxidation, inhibition of collagen synthesis, and the promotion of extracellular matrix degradation [48]. However, Bi [46] showed that 0.78 mg/kg and 2.34 mg/kg CSBTA could significantly reduce hepatocyte degeneration, necrosis, inflammatory cell infiltration, and the activity of serum ALT but had no obvious effect on the activity of AST.
Some studies found that CSBTA has hepatoprotective and anti-fibrotic effects on rats with chronic liver fibrosis [47], which may promote the reversal of liver fibrosis by inhibiting the expression of TGF-β1 and MMP-9 [49]. The effects of CSBTA on chronic liver injury induced by CCl4 may include the regulation of tricarboxylic acid (TCA) circulation, intestinal microbial metabolism, and taurine and hypotaurine metabolism disorder [43]. Furthermore, researchers reported that dehydrocavidine (1), palmatine (9), and berberine (4) were shown to induce apoptosis and autophagy in HSC-T6 cells, and suggested that these three components may be the components in CSBTA that are effective against liver fibrosis [41]. Through the biotransformation mediation of CSBTA by CYP450s in the liver, CSBTA-drug interactions might occur through CYP450s inhibition, particularly CYP1A and CYP2D. [42].
This CSB extract could significantly reduce the degree of liver damage, exert a protective effect on acute liver injury caused by acetaminophen [54] and CCl4 [53], respectively. Meanwhile, the therapeutic effects of this CSB extract on acute liver injury induced by CCl4 may be related to regulation of alanine, aspartic acid, glutamic acid, and glycerol metabolic dysfunction [52,55].
Betulinic acid (71), β-amyrin acetate (73), and (−)-pallidine (51) in a CSB extract may play an anti-fibrotic role by regulating the targets FXR, COX-2, and MMP-1 [78]. 1H NMR was used to study the changes in the sera of rats with liver fibrosis induced by CCl4 treated with this CSB extract, and partial least squares discriminant analysis showed that metabolic disorders were reduced after CSB treatment [51]. In summary, the protective efficiency and anti-fibrosis effects on acute and chronic liver injury of CSBTA provide the scientific basis for the utilization of CSB resources. However, the discrepancy of action mechanism between monomer compounds and extracts needs further explanation.

6.2.2. Isolated Phytochemicals

Through the study of group–effect relationships, Lu [56] reported for the first time that dehydrocavidine (1), palmatine (9), and berberine (4) could significantly inhibit the proliferation of HSC-T6 cells and induce their apoptosis in vitro without obvious cytotoxic effects at effective concentrations. Dehydrocavidine (1) can significantly reduce ALT, AST, and total bilirubin (TBIL) in mice with acute liver injury and effectively reduce CCl4-induced hepatocyte degeneration, necrosis, inflammatory cell infiltration, and ultrastructural destruction [59]. In a rat model of liver fibrosis induced by CCl4, dehydrocavidine (1) could reduce the degree of liver injury and the formation of interstitial fibrous tissue, and its mechanism of liver protection and liver fibrosis inhibition may be related to the extracellular matrix and antioxidant stress [57,58]. To date, most studies have focused on the effects of CSBTA on liver injury and fibrosis, while few studies have focused on its contained individual compounds. More studies should be undertaken that focus on this field.

6.3. Anti-HBV Activity

6.3.1. Crude Extract

HBV causes the disease hepatitis B, and it is also infectious to a certain extent. CSB has been reported to have very good anti-HBV activity. Wang’s experiment proved that the extract of CSB has anti-duck hepatitis B virus (DHBV) effects in vivo, as the level of serum DHBV DNA in all CSB groups decreased significantly after treatment. Pathological examination showed that the CSB extract had a protective effect on liver injury induced by DHBV [60].

6.3.2. Isolated Phytochemicals

Zeng et al. [3] isolated dehydrocheilanthifoline (8) from CSB and showed its anti-HBV activity for the first time. The inhibition of HBsAg (IC50 17.12 μM) and HBeAg (IC50 15.58 μM) by dehydrocheilanthifoline was dose- and time-dependent, with obvious anti-HBV activity in vitro. Moreover, the anti-HBV activities of certain compounds isolated from CSB were evaluated, and dihydrochelerythrine (18) and (−)-salutaridine (54) were found to have strong inhibitory effects on HBsAg and HBeAg with IC50 values of <0.02, <0.02 mg/mL for dihydrochelerythrine (18), respectively, and 0.09, 0.15 mg/mL for (−)-salutaridine (54), respectively [61]. Dehydrocavidine (1), dehydroapocavidine (2), and dehydroisoapocavidine (3) had obvious inhibitory effects on HBsAg and HBeAg and showed no toxicity in 2.2.15 cells [15]. Furthermore, the anti-HBV activities of 10 major alkaloids were tested. Among the tested compounds, dihydrochelerythrine (18) exhibited the most potent activity against HBsAg and HBeAg secretion, with IC50 values < 0.05 μM and selectivity index (SI) values > 3.5 [18]. The research mentioned above showed the application value in the prevention and treatment of HBV of CSB. The practical values related to prevention and treatment of HBV indicate that the prevention of HBV utilizing dihydrochelerythrine (18) demands further studies on its mechanism of action.

6.4. Enhancement of Immune Function

Crude Extract

Immune function is the body’s resistance to diseases. The immune function of the human body is mainly manifested in three aspects: immunoligic defence, immunoligic homeostasis, and immunoligic surveillance. A study showed that CSBTA enhanced the haemolytic plaque value and delayed-type hypersensitivity in mice in vivo, enhanced the mixed culture response of allotypic mice splenocytes in vitro, and enhanced the proliferation response of splenocytes stimulated by mitogen. In addition, CSBTA increased the levels of IFN-γ and IL-2 produced by T cells in a dose-dependent manner, indicating that CSBTA is an enhancer of immune regulation [62]. It can be seen from the literature results that CSBTA has a certain effect on enhancing immune function.

6.5. Antioxidant Activity

6.5.1. Crude Extract

The IC50 refers to the measured semi-inhibitory concentration of an antagonist 2,2-Diphenyl-1-picrylhydrazyl radical (DPPH) is a relatively stable free radical and which is frequently used as a reactivity model of reactive oxygen species. In a study, when the DPPH radical was eliminated, the absorbance A at the maximum absorption wavelength of 519 nm decreased. CSBTA enhanced the antioxidative ability of rat livers by reducing the content of MDA and increasing the activity of SOD in a dose-dependent manner, which indicated that CSBTA can directly or indirectly alleviate liver cell injury and inflammation through antioxidation, thus playing a very good role in protecting the liver [63].

6.5.2. Isolated Phytochemicals

The IC50 of the free radical scavenging activity of nine alkaloids derived from CSB in vitro ranged from 0.25 to 16.51 mg/mL [2]. The structure–activity relationship study of the antioxidant activity showed that the antioxidant activities of compounds with basic tertiary amines were higher than those of their corresponding basic quaternary amine compounds [2]. The kinetics results showed that the compounds with the highest activity, (−)-pallidine (51) and (+)-cheilanthifoline (36), had non-linear DPPH radical scavenging activity, while the other alkaloids displayed linear DPPH radical scavenging activity, indicating that their DPPH radical scavenging activity was dose-dependent [2]. Despite its strong antioxidant activity, many unknown mechanisms of alkaloids exist, which demands further investigation.

6.6. Effects on the Central Nervous System

6.6.1. Crude Extract

The central nervous system is a group of neurons that regulate a specific physiological function, such as a respiratory center, a thermoregulation center, or a language center. There is increasing evidence showing how phytochemicals influence the central nervous system. They may take effect as anticonvulsants, antidepressants, hypnotic sedation, and anti-senile dementia, and have other pharmacological effects. Some studies have reported that CSB has effects on the central nervous system. A study demonstrated that CSBTA (50, 100 mg/kg) significantly decreased the contents of 3,4-dihydroxyphenylacetic acid (DOPAC), homovanillic acid (HVA), 5-hydroxytryptamine (5-HT), and 5-hydroxyinolacetic acid (5-HIAA) but had no significant effect on the level of dopamine (DA). However, the DA/DOPAC and DA/HVA ratios increased, and the 5-HT/5-HIAA ratio of the limbic system also increased. Thus, it has been suggested that CSBTA can inhibit the metabolism of DA and 5-HT in certain brain regions [64]. Furthermore, CSBTA significantly inhibited caffeine-induced excitatory activity in mice. In general, CSBTA had a calming effect on monkeys and cats. Additionally, in some animals, CSBTA produced catalepsy. The irritation response induced by electrical stimulation was found to be significantly inhibited in mice. The conditioned response of the rats was blocked, but CSBTA showed little effect on the unconditioned response. In a study of the central inhibition mechanism, it was found that CSBTA decreased the levels of 5-HT and 5-HIAA in the striatum [66].

6.6.2. Isolated Phytochemicals

Dehydrocavidine (1), isolated from CSB, can increase the contents of DA and 3-dihydroxyphenylacetic acid in the striatum of normal rats and decrease the contents of DA and HVA in the striatum of model rats [67]. The results suggested that dehydrocavidine (1) can significantly block central DA function in rats. In short, dehydrocavidine (1) is the major contributor of CSB’s effects on the central nervous system.

6.7. Anti-Inflammatory Activity

Crude Extracts

Inflammation is a body’s protective response to potentially harmful stimuli. The stimuli can initiate the immune system to provide protection, while the protective mechanism depends on neutrophils, macrophages, dendritic cells, and monocytes. Those cells produce various inflammatory mediators (IL-1β, IL-6, TNF-α, IL-8, chemokines, eicosanoids, histamine, etc.), which results in acute inflammation. Notably, most Chinese herbal medicines have anti-inflammatory properties. A low dose of CSBTA (0.4375 mg/kg) could significantly inhibit the formation of cotton ball granuloma in mice and reduce the chronic inflammatory response [70]. Furthermore, intraperitoneal injection of 50 mg/kg CSBTA could reduce the degree of foot swelling in rats with Danqing arthritis, but the same dose of subcutaneous radiation had no effect on formalin arthritis in rats [66]. CSBTA ameliorates diet-induced non-alcoholic steatohepatitis by regulating hepatic PI3K/Akt and TLR4/NF-κB pathways in mice [4]. It has been reported that C. saxicola rectal suppository had obvious inhibitory effects on ear swelling induced by croton oil, on the increase in capillary permeability induced by acetic acid, and on the writhing reaction induced by peritoneal injection of acetic acid [72]. In addition, CSBTA also was shown to effectively suppress M1 polarization of THP-1-derived Mφs, which may improve the inflammatory environment [69]. Some studies have shown that C. saxicola suppository has a certain therapeutic effect on rats with chronic pelvic inflammatory disease, shows good anti-inflammatory effects, and regulates immunity. Its mechanism may be related to the regulation of the expression of the inflammatory factors TNF-α and IL-6 and the immune proteins IgG and IgM [71]. Future research should identify which chemical constituents of CSB are responsible for these anti-inflammatory effects.

6.8. Analgesic Effect

6.8.1. Crude Extract

Pain is a kind of disease with complex pathogenesis and serious harm to human physical and mental health. Traditional medicines have long been used in the treatment of pain, and numerous medicinal herbs have been reported to be able to relieve pain effectively. Two studies have proven that CSBTA has good analgesic effects. Subcutaneous injection of CSBTA (50 mg/kg, 100 mg/kg) had an obvious inhibitory effect in the writhing reaction of mice, and CSBTA (10 mg/kg) also improved the pain threshold to heat stimulation in rats in the tail flicking test [66]. In another report, Kuai et al. [73] believed that the therapeutic effects of CSBTA were a result of blocking the activation of TRPV1 by improving neuronal damage, improving the loss of intraepidermal nerve fibers (IENFs), and inhibiting inflammation-induced p38 phosphorylation. Therefore, the analgesic effect of CSBTA initially reported on chemotherapy-induced peripheral neuropathy has proposed a novel strategy for the clinical treatment of this disease.

6.8.2. Isolated Phytochemicals

CSB is a Chinese herbal medicine with anti-inflammatory and analgesic effects. The main component of CSB, dehydrocavidine (1), has shown sedative and analgesic effects [21,68]. In the future, the chemical composition of the components that produce analgesia need to be further explored and identified.

6.9. Antibacterial Activity

6.9.1. Crude Extract

Infections caused by Gram-positive and Gram-negative bacteria are one of the foremost causes of morbidity and mortality globally. However, natural products are the main sources of antimicrobials used in clinical practice, serving as a rich reservoir for the discovery of new antibiotics. As two good indicators for evaluating antimicrobial agents, the minimum inhibitory concentration (MIC) refers to the minimum drug concentration that can inhibit the growth of bacteria in a certain medium, which is the minimum inhibitory concentration. The minimum bactericidal concentration (MBC) is the minimum concentration required to kill 99.9% of bacteria. CSBTA had inhibitory and bactericidal effects against nine common Gram-positive and Gram-negative bacteria with MIC values ranging from 16.8–130 mg/mL [74]. Additionally, CSBTA exhibited antimicrobial activity with an MIC value of 20 mg/mL against Staphylococcus aureus. Notably, CSBTA combined with penicillin, cefradine, and levofloxacin effectively inhibited S. aureus [75]. The results above indicated that CSB and penicillin, cefradine, and levofloxacin had a synergistic antibacterial effect.

6.9.2. Isolated Phytochemicals

The content of dehydrocavidine (1) from CSB is high, approximately 0.1–0.2%. Dehydrocavidine (1) has inhibitory effects on S. aureus, Streptococcus β-haemolyticus, diphtheria bacilli, and β-streptococcus, as well as penicillin-resistant Staphylococcus albicans and S. aureus [21,68]. In addition, in vitro antibacterial tests further showed that dehydrocavidine (1) had certain inhibitory effects on Gram-positive strains at a minimum concentration of 0.078 mg/mL. However, it had no inhibitory effects on Gram-negative bacteria [76]. Palmatine (9) has a strong inhibitory effect on some common pyogenic cocci and intestinal pathogenic bacteria [81]. The potential mechanism of the antibacterial activity of these compounds needs to be studied in the future.

6.10. Choleretic Effects

Crude Extract

The impairment of bile flow generally results from a decreased function of the liver and gallbladder. Many traditional Chinese medicines are considered to have good choleretic effects. A researcher found that intravenous injection of CSBTA could increase bile excretion in normal Sprague–Dawley (SD) rats [77], which suggested that CSBTA has a regulating protection for the function gallbladder. However, intravenous injection of CSBTA (20 mg/kg) had no cholagogic effect on anesthetized guinea pigs [66]. Its mechanism needs further elaboration.

6.11. Other Activities

Without affecting the normal physiological state of rats, CSBTA can significantly reduce the levels of plasma total cholesterol and low-density lipoprotein cholesterol in rats, regulate the level of blood lipids in rats fed a high-fat diet, and protect against fatty liver diseases caused by a high-fat diet [79]. Based on 16S rRNA gene sequencing and untargeted metabolomics analyses, CSBTA is an effective and reliable compound for use in co-metabolism and intestinal flora intervention methods in rats with antibiotic-induced intestinal flora imbalance [78]. Regarding the intestinal mucosal transport of dehydrocavidine (1), it had the ability to be effluxed by transporters, but it showed the characteristics of passive transport at a higher concentration range [80]. Apart from the mentioned activities, we suggest that we should further explore other pharmacological properties of CSB.

7. Clinical Applications

7.1. Icteric Hepatitis

Icteric hepatitis is an injury to the hepatocytes caused by various factors. The destruction of liver tissue leads to a decrease in bilirubin uptake and a decrease in the binding functions of hepatocytes, which leads to an increase in bilirubin in the blood [82,83,84]. The common causes of icteric hepatitis are viral infection, drug injury, alcohol injury, autoimmune factors, and so on. A large number of studies have reported that CSBI has a high total effective rate in the treatment of icteric hepatitis (Table 3).
After treating some patients with chronic liver disease and jaundice with CSBI, the symptoms of fatigue, abdominal distension, liver pain, and anorexia were significantly improved [84], and the decreases in transaminase and bilirubin were more obvious [82]. However, after CSBI injection, a patient with acute icteric hepatitis developed symptoms of pruritus, palpitation, chills, and fever, and immediately discontinued the use of CSBI. After discontinuation, the patient did not develop these symptoms again, which were considered to be an allergic reaction to CSBI [83]. All in all, CSBI plays a significant role in acute and chronic hepatitis with a higher total effective rate, but a few adverse reactions occur. Additionally, how to solve those adverse reactions is a potential research point.
In the clinical symptoms and the liver function indexes of some icteric hepatitis, CSBI injection is found to be superior to potassium magnesium aspartate [12,88,89,118], “Yinzhihuang” injection (including baicalin and the extracts of Artemisiacapillaris Thunb, Gardenia jasminoides Ellis, and Lonicera japonica Thunb.) [87]. Meanwhile, the curative effect of CSBI plus sugar once a day was significantly higher than that of 40 mL of Xilikang plus sugar once a day [85]. CSBI can repair liver damage after interventional therapy for advanced liver cancer with hepatocellular jaundice without obvious adverse reactions [86].
In some cases, CSB combined with other drugs for the treatment of icteric hepatitis showed high total effective rate, such as “Yinzhihuang” injection [119], compound glycyrrhizin [120], “Danshen” (Salvia miltiorrhiza Bge), and “Yinzhihuang” [90,91]. In addition, Artemisiae Scopariae Herba (30 g), CSB (30 g), Lysimachia christinae Hance (30 g), Serissa japonica (30 g), Pteris multifida poir (10 g), and Ardisia japonica (10 g) were used in the treatment of acute icteric hepatitis, The contents of TB and ALT decreased compared with those before treatment. Taking these results into account, the combined drug therapy with CSBI has a high efficacy in the treatment of icteric hepatitis in spite of the unknown metabolic interactions and mechanisms of action.

7.2. Viral Hepatitis

Viral hepatitis is a group of infectious diseases caused by hepatitis viruses that mainly damage the liver. The clinical manifestations of various hepatitis diseases are similar, as most patients may have digestive tract symptoms, systemic symptoms, jaundice, right upper abdominal pain, and so on.
In some clinical studies, after being treated with CSBI, ALT, and TBIL, levels were significantly decreased in patients with viral hepatitis [93] and chronic viral hepatitis B [92]. Compared with “Yinzhihuang” injection, CSBI has a better effect on patients with acute and chronic viral hepatitis and significantly decreased the levels of ALT, AST, and TBIL [94]; the degrees of reduction in TBIL, DBIL, and ALT were also significantly different [95]. Additionally, compared with diammonium glycyrrhizinate injection, CSBI has more obvious efficacy in the symptoms of jaundice. However, two patients reported that they had local vascular pain during the CSBI infusion, but the pain disappeared after the infusion was completed [96].
CSBI was shown to effectively improve the clinical symptoms of patients with acute and chronic viral hepatitis, and decrease the levels of serum total bilirubin (STB), 1-min bilirubin, ALT, and AST in a short period of time [97]. The effect of CSBI on STB was significantly better than that of potassium magnesium aspartate after 4 weeks of treatment [121]. In addition, “Danshen” injection combined with CSBI was used to treat severe jaundice stemming from viral hepatitis with a total effective rate of 96.14%. There was one mild rash in the treatment group and another in the control group [98]. From the perspective of enzymes, CSBI treats acute and chronic viral hepatitis by reducing the expression of ALT, AST, TBIL, and DBIL.

7.3. Acute and Chronic Hepatitis

Hepatitis is inflammation of the liver caused by various factors and is divided into acute and chronic categories. Acute hepatitis is an abnormal liver function that occurs for the first time.
In a study, the levels of serum TBIL and ALT in patients with chronic hepatitis B treated with CSBI were found to be significantly lower than those in the control group, and symptoms such as fatigue, nausea, vomiting, and greasiness were improved [92,122]. In addition, injection of CSBI at the “Zusanli” acupoint can not only treat patients with acute hepatitis but also play the role of acupuncture in dredging meridians, regulating visceral function, strengthening body resistance, and eliminating toxins. Therefore, the combination of the two treatments has a good therapeutic effect; this provides an innovative perspective for the application of CSBI.
Magnesium isoglycyrrhizinate combined with CSBI was used to treat patients with chronic cholestatic hepatitis B [99]. Moreover, CSBI was combined with telbivudine for the treatment of chronic severe hepatitis B [123]. CSBI combined with breviscapine injection for the treatment of chronic severe hepatitis showed that the clinical efficacy in the treatment group was significantly better than that in the control group [100]. Furthermore, it has also been reported that CSBI can be combined with “Danshen” injection or atomolan for the treatment of chronic hepatitis B [101,124]. The above CSBI in combination with other drugs has a significant effect on the treatment of hepatitis B. Hence, assessing the contribution of combined drugs would optimize the combination and facilitate the development of CSBI-related drugs.

7.4. Liver Cancer

Liver cancer refers to malignant tumours occurring in the liver, usually due to primary liver cancer, but including both primary and metastatic liver disease. Primary carcinoma in liver cancer refers to carcinomas occurring in liver cells or intrahepatic bile duct cells. The etiology of primary liver cancer is not completely clear and may be the result of synergistic effects from multiple factors.
The effect of CSBI combined with interventional radiotherapy on liver function and quality of life (QOL) score in the observation group of a study were significantly higher than those in the control group after treatment [102,103]. Additionally, CSBI combined with interventional radiotherapy can effectively improve the clinical efficacy of middle and advanced liver cancer [104,105,107,108], and improve the quality of life of patients with advanced liver cancer [109]. In addition, CSBI had an obvious liver reparative effect after interventional therapy for advanced liver cancer with hepatocellular jaundice with fewer adverse reactions [86,106]. Hence, the combination therapies with CSBI and interventional radiotherapy have been shown to effectively treat liver cancer which encourages medicinal chemistry to further develop and improve therapies.
CSBI combined with hepatic arterial chemoembolization (TACE) significantly improved the immune function and quality of life of patients with advanced liver cancer [110]. CSBI can effectively prevent and treat liver damage after TACE treatment [112]. Moreover, CSBI combined with interventional therapy has a significant effect on relieving clinical symptoms and improving liver function, especially with respect to increasing serum albumin levels in patients with liver cancer [111]. Octreotide combined with CSBI had a significant effect on advanced liver cancer [113], No adverse reactions were observed except for local pain during subcutaneous injection of octreotide, nausea, vomiting, and local vascular prickling when CSBI was added to the treatment [125].

7.5. Hyperbilirubinemia

Bilirubin is a bile pigment that is not only the main pigment in human bile but also an important index of liver function and an important basis for the diagnosis of jaundice [126]. Jaundice may cause the patient’s skin, mucous membranes, and sclera to display yellow staining that may also be accompanied by abdominal distension, abdominal pain, loss of appetite, and other symptoms, some of which are even life-threatening in severe cases [114].
It has been reported that 54 cases of hyperbilirubinemia treated by CSBI showed significant clinical effects, with a total effective rate of 91.47% [126]. CSBI treatment can significantly reduce the degree of neonatal jaundice and shorten its duration, and adverse reactions such as rash and diarrhea were significantly lower in the CSBI treatment group than in the control group [114]. Furthermore, CSBI combined with reduced glutathione is effective in the treatment of hyperbilirubinemia.

7.6. Others

In addition to treating hepatitis and liver cancer, CSBI can also treat a number of other diseases. For example, CSBI has a good clinical effect in the treatment of advanced rectal cancer [115]. When ribavirin and CSBI were combined for the treatment of hemorrhagic fever with renal syndrome, the recovery of AST, ALT, LDH, and BUN levels was more significant [116]. Both CSBI and “Shengmai” injections (the main ingredients include Radix ginseng rubra, Ophiopogon japonicus (Linn. f.) Ker-Gawl. and Schisandra chinensis) had good curative effects in alleviating the symptoms of liver cirrhosis, protecting the liver, and enhancing the gallbladder. However, “Shengmai” injection was notably superior to CSBI in alleviating discomfort in the liver area and yellow skin staining, and especially in reducing serum transaminase and bilirubin [117]. Therefore, in order to fully utilize CSBI, researchers should further explore CSBI alone or in combination with other drugs to treat other diseases.

8. Toxicity Assessment

To date, research on the toxicology and safety of CSB is very limited from both a traditional and modern standpoint.
There are some studies on the toxicity of CSBTA [54,68,83,91], and CSBTA (20 mg/kg/day) may have an inhibitory effect on the growth of rats, which was more obvious after three weeks of administration. In addition, there were no significant changes in SGPT, NPN, or haemogram in dogs after administration of CSBTA (3 mg/kg/day) for 2 or 4 weeks. The animals in both the treatment and control groups showed different degrees of sinus arrhythmias before and after administration [66]. The LD50 of CSBTA administered subcutaneously to mice was determined to be 233 mg/kg [65]. In another study, after oral administration of the CSB extract (560, 450, 360, 290, and 230 mg/kg), most of the mice died from poisoning within 2 days. The LD50 of this CSB extract was 298.5 mg/kg, and the 95% confidence limit was 257.2–346.5 mg/kg [54]. Therefore, this CSBI extract can be considered safe for use in a certain dose range.
There has been only one report on the toxicity of the individual CSB compounds. Acute toxicity from dehydrocavidine manifests as lethargy, weakness, paralysis, and death. The LD50 was determined to be 71.6 ± 2.92 mg/kg. A subtoxicity test administered this compound at dosages of 10 and 5 mg/kg/day. [68]. On the basis of the literature, toxicity tests of pure substances in CSB can be rarely found, which means it is urgent to perform a more toxicological experiment on it to subsequently guide its clinical application.
In several clinical studies using CSBI, adverse reactions have occurred. When CSBI was used in a clinical study for the treatment of jaundice hepatitis, one patient developed pruritus, palpitations, chills, and fever [83]. In another report, three patients had pain at the injection site after intravenous administration of CSBI, but the symptoms disappeared after deceleration [91]. Moreover, diarrhoea and rash occurred during the treatment of hyperbilirubinemia [114].
When CSBI was used to treat viral hepatitis, some studies showed adverse reactions. One patient developed a low fever within a few hours after the first treatment, but there was no fever after the second treatment when the infusion speed was slowed down [94]. Two patients reported local vascular pain during CSBI infusion, with the pain disappearing after the infusion [96]. Additionally, one patient in the CSBI treatment group had a mild rash [98]. The adverse reaction related to the allergic reaction is caused by the specific constitution of the patient, suggesting that the doctor would pay attention to it.
Finally, there are adverse symptoms associated with the use of CSBI for the treatment of liver cancer. In one study, fever, trembling, and itchy skin occurred in the CSBI injection group (6.7%) [86,106]. In this study group, adverse reactions occurred in five patients, with an incidence rate of 10.42% [113]. In the treatment of patients with advanced liver cancer, the adverse effect rates in the treatment groups were 20.8%, 5.2%, and 5.2% [108,113,125].

9. Conclusion and Future Perspectives

In summary, the botanical distribution and description, traditional usages, phytochemistry, pharmacological activities, and clinical applications of CSB are presented in the current review. Although the related quality control standards of CSB have not been included in the Chinese Pharmacopoeia, according to ancient Chinese herbal writings, CSB has been widely applied for treating hepatitis, liver cancer, clearing away heat, and toxic materials, relieving pain, and stopping bleeding. Alkaloids, the abundant active ingredients of CSB based on the current phytochemical, pharmacological studies, and clinical applications, have the fruitful effects of protecting the liver, being an anti-hepatitis virus, and having anti-tumour and analgesic effects, which expand traditional Chinese medicine applications.
At present, studies on the chemical constituents, pharmacological activities, and clinical applications of CSB have made substantial development whereas the scientific research gap in the mechanism of action remains. Therefore, to improve the safety and efficacy of CSB, further exploration and discussion of relative studies on CSB from the following aspects in the future are required.
Firstly, only 80 different secondary metabolites have been reported with 50% alkaloids, when the compressive structural activity relationship between the structural features and bioactivities is highly required. In addition, most major studies have been focused on the exploration of alkaloids from CSB, and studies on other bioactive components are limited which may lead to the loss of significant discoveries.
Secondly, diverse pharmacological effects of CSB have been reported whereas limited studies have been conducted on its pharmacological mechanism of action. Thus, the pharmacological mechanism should be further elucidated. Moreover, the preliminary pharmacokinetic parameters of CBSTA have been accomplished by performing analytical studies of in vivo experiments. The clarified process in vivo and the reasonably modified dosage form of CSB would improve its usage of it with more wide and safe use in clinical practice.
Thirdly, combination drug developments with CSBI have been adopted in the clinical treatment of acute and chronic diseases, resulting in a vastly enriched the diversity of drug ratio between CSBI and drugs. However, the mechanism of the pharmacological action of combination and the contribution of each active component are remaining which require more effort and endeavors from chemists and biologists.
Fourthly, the main chemical alkaloid constituent of CSB has been determined to be dehydrocavidine. CSBI and tablets can be applied to clinical practices; however, clinical application of dehydrocavidine has not yet been reported.
Finally, non-adequate toxicological properties of CSB suggest that research should be performed assessing adverse effects and exploring potential toxicity components of CSB, which encourages that the safety evaluation should be required in each pharmacological study as a supplement in the future.

Author Contributions

F.Q. collected literature data and wrote the manuscript; Y.C. and F.-F.W. retrieved relevant literature arranged the tables. S.-Q.T. corrected the botanical distribution and description, Y.-L.F. gave important suggestions and provided financial support for this work. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the National Natural Science Foundation of China (NO. 81760626, NO. 21977021).

Institutional Review Board Statement

Not Applicable.

Informed Consent Statement

Not Applicable.

Data Availability Statement

Not Applicable.

Conflicts of Interest

The authors confirm that this article content has no conflict of interest.

Abbreviations

ALTalanine transaminase
ASTaspartate transaminase
Bcl-2B cell lymphoma-2
BCGbacillus calmette-guerin
BUNblood urea nitrogen
Cdc42Cell division cycle 42
CYPcytochrome
CSBCorydalis saxicola Bunting
CSBICorydalis saxicola Bunting injection
CSBTACorydalis saxicola Bunting total alkaloids
CCl4carbontetrachloride
CD86cluster of differentiation 86
COX-2cyclooxygenase-2
DAdopamine
DPPH2,2-diphenyl-1-picrylhydrazyl
DBiLdirect bilirubin
DBdirect bilirubin
DHBVduck hepatitis B virus
DOPAC3,4-Dihydroxyphenylacetic acid
EC50concentration for 50% of maximal effect
FasLFas ligand
HThydroxytryptamine
HIAAhydroxyindolacetic acid
HBVhepatitis B virus
HBsAghepatitis B surface antigen
HBeAghepatitis B e antigen
hTERTtelomerase reverse transcriptase
HVAhomovanillic acid
GTglutamyltransferase
i.pintraperitoneally injected
iNOSinducible nitric oxide synthase
IC50half maximal inhibitory concentration
IFN-γinterferon-γ
ILinterleukin
IENFintraepidermal nerve fiber
Kiinhibition constant
LD50median lethal dose
LDHlactate dehydrogenase
MDAmalondialdehyde
MAPKsmitogen-activated protein kinases
MMPmatrix metal loprotease
MTT3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
MICminimal inhibitory concentration
NPNnon-protein nitrogen
NF-κBnuclear transcription factor-κB
NMRnuclear magnetic resonance
NFATc1nuclear factor of activated T-cells c1
OPGosteoprotegerin
QOLquality of life
RANKLreceptor activator of nuclear factor- κB ligand
SODsuperoxide dismutase
SGPTserum glutamate pyruvate transaminase
SBserum total bilirubin
TNF-αtumour necrosis factor-α
TCAtricarboxylic acid
TGF-β1transforming growth factor-β1
TBILtotalbilirubin
THP-1leukemia
TRPV1transient receptor potential vanilloid 1
TBiLtotal bilirubin
TACEhepatic artery chemoembolization

References

  1. Zhang, B.; Huang, R.Z.; Hua, J.; Liang, H.; Pan, Y.M.; Dai, L.M.; Liang, D.; Wang, H.S. Antitumor lignanamides from the aerial parts of Corydalis saxicola. Phytomedicine 2016, 23, 1599–1609. [Google Scholar] [CrossRef]
  2. He, Z.C.; Wang, D.M.; Li, G.C.; Wu, J.Y. Study on alkaloids from Corydalis saxicola and their anti-oxidative activities. Chin. Tradit. Herb. Drugs 2014, 45, 1526–1531. [Google Scholar]
  3. Zeng, F.L.; Xiang, Y.F.; Liang, Z.R.; Wang, X.; Huang, D.E.; Zhu, S.N.; Li, M.M.; Yang, D.P.; Wang, D.M.; Wang, Y.F. Anti-hepatitis B virus effects of dehydrocheilanthifoline from Corydalis saxicola. Am. J. Chin. Med. 2013, 41, 119–130. [Google Scholar] [CrossRef]
  4. Wu, J.J.; Chen, P.; Ju, L.J.; Gao, R.H.; Li, S.L.; Huang, Z.Q.; Cheng, Y.Q.; Gui, S.Q.; Qiu, Z.X.; Cheng, J.; et al. Corydalis saxicola Bunting total alkaloids ameliorate diet-induced non-alcoholic steatohepatitis by regulating hepatic PI3K/Akt and TLR4/NF-κB pathways in mice. Biomed. Pharmacother. 2022, 151, 113132. [Google Scholar] [CrossRef]
  5. Xie, G.Y.; Jin, S.Y.; Li, H.T.; Ai, M.K.; Han, F.; Dai, Y.Q.; Tao, W.; Zhu, Y.; Zhao, Y.C.; Qin, M.J. Chemical constituents and antioxidative, anti-inflammatory and anti-proliferative activities of wild and cultivated Corydalis saxicola. Ind. Crop. Prod. 2021, 169, 113647. [Google Scholar] [CrossRef]
  6. Huang, Q.Q.; Bi, J.L.; Sun, Q.Y.; Yang, F.M.; Wang, Y.H.; Tang, G.H.; Zhao, F.W.; Wang, H.; Xu, J.J.; Kennelly, E.J.; et al. Bioactive isoquinoline alkaloids from Corydalis saxicola. Planta Med. 2012, 78, 65–70. [Google Scholar] [CrossRef] [Green Version]
  7. Qin, F.; Dai, L.M.; Zhang, B.; Huang, R.Z.; Wang, F.F.; Qin, J.K.; Liang, D.; Wang, H.S. (±)-Corysaxicolaine A: A pair of antitumor enantiomeric alkaloid dimers from Corydalis saxicola. Org. Biomol. Chem. 2022, 20, 1396–1400. [Google Scholar] [CrossRef] [PubMed]
  8. Li, M.M.; Wang, J.Y.; Mo, B.W.; Zeng, J.R.; Yao, D. Total alkaloids of Corydalis saxicola Bunting inhibits migration of A549 cells by suppressing Cdc42 or Vav1. Oncol. Lett. 2018, 15, 475–482. [Google Scholar] [CrossRef]
  9. Jiang, S.Y.; Hu, X.H.; Zhao, R.F. Study on the introduction and cultivation of Corydalis saxicola Bunting. Guihaia 2002, 22, 469–473. [Google Scholar]
  10. Institute of Botany, Chinese Academy of Sciences. Flora of China; Institute of Botany, Chinese Academy of Sciences: Beijing, China, 1999; Volume 32, p. 418. [Google Scholar]
  11. Wen, H.Q.; Xu, Z.R.; Villa-Lobos, J.; Skog, L.E. A list of threatened limestone plants in south China. Guihaia 1993, 13, 110–127. [Google Scholar]
  12. Wei, M.K.; Lu, J.X. High-yield cultivation techniques of Corydalis saxicola Bunting in karst mountains. Guangxi Agric. Sci. 2004, 35, 21. [Google Scholar]
  13. Wu, Y.; Lu, T.L.; Ji, D.; Zhou, Y.; Mao, C.Q. Isolation and structural identification of alkaloids from Corydalis saxicola. J. Nanjing Univ. Tradit. Chin. Med. 2015, 31, 81–83. [Google Scholar]
  14. Li, H.L.; Zhang, W.D.; Zhang, W.; Zhang, C.; Liu, R.H. A new nitro alkaloid from Corydalis saxicola Bunting. Chin. Chem. Lett. 2005, 16, 367–368. [Google Scholar]
  15. Li, H.L.; Han, T.; Liu, R.H.; Zhang, C.; Chen, H.S.; Zhang, W.D. Alkaloids from Corydalis saxicola and their anti-hepatitis B virus activity. Chem. Biodivers. 2008, 5, 777–783. [Google Scholar] [CrossRef] [PubMed]
  16. Li, H.L.; Zhang, W.D.; Han, T.; Zhang, C.; Liu, R.H.; Chen, H.S. Tetrahydroprotoberberines alkaloids from Corydalis saxicola. Chem. Nat. Comp. 2007, 43, 173–175. [Google Scholar] [CrossRef]
  17. Ke, M.M.; Zhang, X.D.; Wu, L.Z.; Zhao, Y.; Zhu, D.Y.; Song, C.Q.; Xu, R.S. Studies on the active principles of Corydalis saxicola Bunting. Chin. Pharm. J. 1980, 15, 41. [Google Scholar]
  18. Wu, Y.R.; Ma, Y.B.; Zhao, Y.X.; Yao, S.Y.; Chen, J.J. Two new quaternary alkaloids and anti-hepatitis B virus active constituents from Corydalis saxicola. Planta Med. 2007, 73, 787–791. [Google Scholar] [CrossRef]
  19. Cheng, X.X.; Wang, D.; Jiang, L.; Yang, D.P. DNA topoisomerase I inhibitory alkaloids from Corydalis saxicola. Chem. Biodivers. 2010, 5, 1335–1344. [Google Scholar] [CrossRef]
  20. Wang, Q.Z.; Liang, J.Y.; Feng, X. A new alkaloid from the herb of Corydalis saxicola. Chin. J. Nat. Medicines. 2009, 7, 0414–0416. [Google Scholar] [CrossRef]
  21. Ke, M.M.; Zhang, X.D.; Wu, L.Z.; Zhao, Y.; Zhu, D.Y.; Song, C.Q.; Xu, R.S. Studies on the active principles of Corydalis saxicola Bunting. Acta Bot. Sin. 1982, 24, 289. [Google Scholar]
  22. Wang, Q.Z.; Liang, J.Y.; Yuan, Y. Chemical constituents of Corydalis saxicola. Chin. J. Nat. Medicines. 2007, 5, 31–34. [Google Scholar]
  23. Mao, Y.A. Chemical Constituents and Activity of Corydalis saxicola Bunting; Guangxi Medical University: Nanning, China, 2006. [Google Scholar]
  24. Cheng, X.X.; Yang, D.P.; Wang, D.M.; Jiang, M. Isolation of alkaloids from Corydalis saxicola by high-speed counter-current chromatography. J. Chin. Med. Mater. 2011, 34, 1062–1064. [Google Scholar]
  25. Ju, J.N.; Ming, Y.Z.; Dai, Q.K.; Li, K.Z.; Huang, S.; He, J.B.; Wu, G.B.; Chen, C. Effect of Corydalis saxicola Bunting aqueous extract on proliferation and migration of hepatocellular carcinoma HepG2 cells and its possible mechanism. Chin. J. Oncol. Prev. Treat. 2018, 10, 434–438. [Google Scholar]
  26. Sang, Y. Research of Corydalis saxicola Bunting Total Alkaloids on Nude Mouse with Subcutaneous Transplantation Tumor and Bone Metastasis in Lung Cancer A549; Guilin Medical University: Guilin, China, 2017. [Google Scholar]
  27. Du, Y.J. Corydalis saxicola Bunting Extractive inhibit F-actin Polymerization in A549 Cells and Its Possible Mechanism; Guilin Medical University: Guilin, China, 2017. [Google Scholar]
  28. Li, J.H.; Wang, J.Y.; Zeng, J.R.; Gao, Y.; Li, M.M.; Yu, Y.Y. Effect of Corydalis Saxicola total alkaloids on human A549 cell proliferation, apoptosis and expressions of caspase surviving. Chin. J. Exp. Tradit. Med. Formul. 2015, 21, 165–169. [Google Scholar]
  29. An, R. Study on Mechanisms and Inhibitory Effects of Saxicola on Human A549 Cell Proliferation; Guilin Medical University: Guilin, China, 2014. [Google Scholar]
  30. Zhu, Y.; Liao, J.X. Effects of Yanhuanglian total alkalions on Bcl-2 activity of oral squamous carcinoma cell lines. J. Oral. Maxil. Surg. 2011, 21, 96–98. [Google Scholar]
  31. Yin, J.K.; Liao, J.X. Effects of Corydalis saxicola Bunting total alkaloids on Tca8113 cell proliferation and apoptosis. J. Oral. Maxil. Surg. 2010, 20, 245–248. [Google Scholar]
  32. Li, J.F.; Liao, J.X.; Li, H.L.; Zhang, W.D. Inhibition of Yanhuanglian total alkalions on cell proliferation and telomerase activity of Tca8113 cell lines. J. Oral. Maxil. Surg. 2007, 17, 32–35. [Google Scholar]
  33. Xie, P.S.; Li, A.Y.; Zhou, F.; Zhao, Y. Experimental study on anti-tumor screening of chinese herbal medicine. Shizhen J. Tradit. Chin. Med. Res. 1995, 7, 19–20. [Google Scholar]
  34. Lu, G.X. Experimental report on the effect of corydalis saxicola on the respiration of tumor cells. Guangxi J. Tradit. Chin. Med. 1979, 4, 11–14. [Google Scholar]
  35. Zhao, Y.; Li, A.Y.; Zhou, F.; Xie, P.S.; Tan, W.J.; Lu, G.X. Experimental study on antitumor effect of Corydalis saxicola bunge. Guangxi J. Tradit. Chin. Med. 1979, 5, 27. [Google Scholar]
  36. Tan, W.Z.; Xie, P.S.; Li, X.J.; Zhou, F.; Li, A.Y. Effect of Corydalis saxicola on tumor cells-experimental summary of methylene blue reduction and cell staining. Guangxi J. Tradit. Chin. Med. 1979, 3, 31–35. [Google Scholar]
  37. Tang, C.L.; Zheng, H.; Wang, J.; Song, H.; Lu, S.Y.; Cheng, B.; Wu, F.; Zhang, H.Y.; Ruan, J.X.; Liang, Y.H. Identification of the active components of Corydalis saxicola extracted from zhuang medicine rock against the inhibition of human hepatocarcinoma cell line SMMC-7721 based on group-activity relationship. Lishizhen Med. Mater. Med. Res. 2016, 27, 2372–2375. [Google Scholar]
  38. Xu, R.; Liao, J.X. Effects of Yanhuanglian total alkaloids and dehydroapocavidine on NF-kappa B activity of oral carcinoma cell lines. J. Oral. Maxil. Surg. 2010, 20, 241–244. [Google Scholar]
  39. Lei, J.; Liao, J.X. Tetradehydroscoulerine on cell proliferation and hTERT expression of Tca8113 cell lines. J. Oral. Maxil. Surg. 2008, 18, 173–177. [Google Scholar]
  40. Wu, F. The Study of the Hepatoprotective Effect of Total Alkaloids from Corydalis saxicola Bunting on Metabonomics and the Metabolism of Its Active Components; Guangxi Medical University: Nanning, China, 2018. [Google Scholar]
  41. Meng, C.; Lu, S.Y.; Liang, Y.; Mo, X.M.; Li, W.D.; Huang, Y.; Cheng, X.; Su, Z.H.; Zheng, H. Electron microscopy study on the apoptosis and autophagy of the hepatic stellate cells induced by total alkaloids. J. Guangxi Norm. Univ. (Nat. Sci. Ed.). 2018, 36, 76–79. [Google Scholar]
  42. Yu, J.J.; Liu, Q.; Lu, X.; Li, X.; Li, N.; Liu, B.; Fang, H.; Qiu, Z. Inhibitory and inductive effects of Corydalis saxicola Bunting total alkaloids (CSBTA) on cytochrome P450s in rats. Phytother. Res. 2018, 32, 1818–1827. [Google Scholar] [CrossRef]
  43. Wu, F.; Zheng, H.; Yang, Z.T.; Cheng, B.; Wu, J.X.; Liu, X.W.; Tang, C.L.; Lu, S.Y.; Chen, Z.N.; Song, F.M. Urinary metabonomics study of the hepatoprotective effects of total alkaloids from Corydalis saxicola Bunting on carbon tetrachloride-induced chronic hepatotoxicity in rats using 1H NMR analysis. J. Pharmaceut. Biomed. 2017, 140, 199–209. [Google Scholar] [CrossRef]
  44. Zhou, J.G. Protective effect of total alkaloids of Corydalis saxicola on chronic liver injury in rats. Shandong Med. J. 2010, 50, 3. [Google Scholar]
  45. Zhou, J.G. Study of hepatoprotective effect of the total alkaloids from Corydalis saxicola Bunting of the acute chemical hepatic injury model mice. China Mod. Med. 2010, 17, 29–30. [Google Scholar]
  46. Bi, M.G.; Zhou, J.; Xu, Y.; Sun, H.; Ji, Y.B. Improving effect of total alkaloids extract from Corydalis thalictrifolia Franch. on immune hepatic injury in mice. Chin. J. Pharmacol. Toxicol. 2009, 23, 39–44. [Google Scholar]
  47. Xi, G.L. Protective Effects of Total Alkaloids in Corydalis saxicola Bunting on Liver Injuries and Meta-Analysis of Telmisartan on Blood Pressure Control; The Second Military Medical University: Shanghai, China, 2009. [Google Scholar]
  48. Jia. J. Protective Effects and Mechanisms of Corydalis saxicola Bunting Total Alkaloids on Experimental Liver Injury; Guangxi Medical University: Nanning, China, 2009. [Google Scholar]
  49. Liang, Y.H.; Gu, J.; Peter, S.S.; Mao, Y.A.; Qin, C.J. Protective effect of Corydalis saxicola Alkaloids (CSA) on level of TGF-β1, MMP-9 in rats with liver fibrosis. Lishizhen Med. Mater. Med. Res. 2008, 19, 2620–2622. [Google Scholar]
  50. Liang, Y.H.; Gu, J.; Peter, S.S.; Mao, Y.A.; Qin, C.J. Protective effect of Corydalis saxicola alkaloids (CSA) on CCl4-induced acute liver injury in mice. Lishizhen Med. Mater. Med. Res. 2008, 19, 2922–2923. [Google Scholar]
  51. Liu, X.W.; Tang, C.L.; Hua, Z.; Wu, J.X.; Fang, W.; Mo, Y.Y.; Xi, L.; Zhu, H.J.; Yin, C.L.; Bang, C. Investigation of the hepatoprotective effect of Corydalis saxicola Bunting on carbon tetrachloride-induced liver fibrosis in rats by 1H-NMR-based metabonomics and network pharmacology approaches. J. Pharmaceut. Biomed. 2018, 159, 252–261. [Google Scholar] [CrossRef] [PubMed]
  52. Liang, Y.H.; Tang, C.L.; Lu, S.Y.; Cheng, B.; Wu, F.; Chen, Z.N.; Song, F.; Ruan, J.X.; Zhang, H.Y.; Song, H. Serum metabonomics study of the hepatoprotective effect of Corydalis saxicola Bunting on carbon tetrachloride-induced acute hepatotoxicity in rats by 1H NMR analysis. J. Pharmaceut. Biomed. 2016, 129, 70–79. [Google Scholar] [CrossRef]
  53. Meng, T.X.; Xie, L.S.; Huang, M.C. Protective effect of Corydalis saxicola Buning extract on liver injury induced by CCl4 in mice. Shanghai J. Tradit. Chin. Med. 2013, 47, 89–91. [Google Scholar]
  54. Huang, X.Z.; Liu, X.P.; Huang, M.; Huang, Z.Q.; Li, D.; Jiang, W.Z. Study on main pharmacodynamics and acute toxicity of herba Corydalis saxicola extract. Chin. J. Hosp. Pharm. 2007, 27, 146. [Google Scholar]
  55. Jiang, D.P. Protective effects of Corydalis saxicola Bunting on acute liver injury in rats. Health Vocat. Educ. 2007, 25, 119–120. [Google Scholar]
  56. Lu, S.Y.; Zheng, H.; Cheng, B.; Fang, W.U.; Wu, J.X.; Liu, X.W.; Tang, C.L.; Liang, Y.H.; Zhang, H.Y.; Xiang, R.J. Discrimination of proliferation inhibiting ingredients in Corydalis soxicola on rat hepatic stellate cell-T6 based on composition-activity relationship. Chin. Tradit. Herb. Drugs 2017, 48, 1354–1361. [Google Scholar]
  57. Rafiee, F.; Nejati, V.; Heidari, R.; Ashraf, H. Protective effect of methanolic extract of Berberis integerrima Bunge root on carbon tetrachlorideinduced testicular injury in wistar rats. Iran. J. Reprod. Med. 2016, 14, 133–140. [Google Scholar]
  58. Wang, T.; Sun, N.L.; Zhang, W.D.; Li, H.L.; Lu, G.C.; Yuan, B.J.; Hua, J.; She, J.H.; Zhang, C. Protective effects of dehydrocavidine on carbon tetrachloride-induced acute hepatotoxicity in rats. J. Ethnopharmacol. 2008, 117, 300–308. [Google Scholar] [CrossRef]
  59. Sun, N.L.; Wang, T.; Yuan, B.L.; Li, H.L.; Lu, G.C.; Zhang, W.D.; Zhang, C.; Yuan, B.J. Protective effects of dehydrocavidine on carbon tetrachloride induced acute liver injury in mice. J. Pharm. Pract. 2008, 26, 23–27. [Google Scholar]
  60. Wang, J.; Zhang, S.J.; Wu, D.H.; Jiang, W.Z. Inhibitory effect of extract from Corydalis saxicola Bunting on HBV in vivo. China Pharm. 2009, 18, 7–9. [Google Scholar]
  61. Wu, Y.R.; Ma, Y.B.; Zhao, Y.X.; Yao, S.Y.; Zhou, J.; Gong, Q.F.; Chen, J.J. Anti-hepatitis virus constituents from Corydalis saxicola. Chin. Tradit. Herb. Drugs 2012, 43, 32–37. [Google Scholar]
  62. Tong, K.; Wu, L.Z.; Liang, Y.Y. Effect of total alkaloids of Corydalis saxicola on immune function of mice. Immunol. J. 1995, 11, 238–241. [Google Scholar]
  63. Deng, W.; Zhu, L.P.; Liu, J.; Zhou, G.L.; Yao, Q.S. Effect of yanhuanglian alkaloids on serum biochemical parameters and liver tissue antioxidant capacity in long-term feeding SD rats. J. Health Toxicol. 2010, 24, 228–230. [Google Scholar]
  64. Wu, C.F.; Liu, W.; Li, F.L. Effect of total alkaloids of corydalis saxicola on monoamine neurotransmitters in rat brain. J. Shenyang Pharm. Univ. 1994, 000, 101–104. [Google Scholar]
  65. Huang, X.N.; Liu, G.X.; Zhang, Y. Stability of total alkaloids from Corydalis saxicola. Acta Pharmacol. Sin. 1981, 2, 156–159. [Google Scholar]
  66. Huang, X.N.; Liu, G.X.; Zhang, X.D. Analgesic, anti-inflammatory and cholagogic effects of total alkaloids from Corydalis saxicola Bunge. Acta Acad. Med. Zunyi. 1981, 4, 22–25. [Google Scholar]
  67. Peng, X.C. Effects of Dehydrocavidine on Behavior and Dopamine in Striatum of Rats; Zunyi Medical University: Zunyi, China, 2008; pp. 18–22. [Google Scholar]
  68. Chen, C.Y.; Zhao, Y. The pharmacological study of the main component dehydrocavitin in the traditional Chinese medicine Corydalis thalictrifolia. China J. Chin. Meteria Med. 1982, 7, 31–34. [Google Scholar]
  69. Feng, C.; Yong, X.Z.; Jiang, Q.Z.; Wu, T.T.; Jiang, L.L.; Su, Z.H.; Tao, R.C. Inhibitory effects of Corydalis saxicola Bunting total alkaloid on M1 macrophages polarization. J. Guangxi. Med. Univ. 2020, 37, 2103–2110. [Google Scholar]
  70. Li, L. Experimental study on anti-inflammatory effect of corydalis saxicola. Chin. J. Ethnomed. Ethnopharm. 2009, 18, 20–21. [Google Scholar]
  71. Xiao, P.; Lin, C.X.; Pan, B.J.; Zhu, G.M.L.; Huang, Y.; Zeng, D.Y. Pharmacodynamics of Yanhuanglian suppository for rats with chronic pelvic inflammatory diseases. Cent. South. Pharm. 2019, 17, 2052–2058. [Google Scholar]
  72. Zhu, G.M.L.; Jiang, W.Z.; Xiao, P.; Huang, X.Y. Experimental study on anti-inflammatory and analgesic effects of Corydalis saxicola rectal suppository. Chin. J. Ethnomed. Ethnopharm. 2019, 28, 11–13. [Google Scholar]
  73. Kuai, C.P.; Lin, J.J.; Hu, P.P.; Huang, F. Corydalis saxicola alkaloids attenuate cisplatin-induced neuropathic pain by reducing loss of IENF and blocking TRPV1 activation. Am. J. Chin. Med. 2020, 48, 407–428. [Google Scholar] [CrossRef]
  74. Qiu, Q.Q.; Wei, Z.P.; Wu, L.M.; Liu, Y.W.; Meng, T.X. In vitro antibacterial activity of total alkaloids from Corydalis saxicola Bunting. Sci. Technol. Innov. 2020, 9, 43–44. [Google Scholar]
  75. Sun, T.T.; Jiang, P.; Zhong, Z.M.; Fang, Y.; Yang, Y.Q.; Dan, H.H.; Zuo, R.H. Bacteriostasis of Corydalis Saxicola combined with antibiotics on Staphylococcus Aureus in vitro. J. Jinggangshan Univ. (Nat. Sci. Ed.) 2020, 41, 111–114. [Google Scholar]
  76. Ye, Q.L.; Wu, L.Z.; Li, H.; Zhang, J. Antibacterial experiment of dehydrocavidine, the main component of Corydalis saxicola Bunting. Guangxi J. Tradit. Chin. Med. 1984, 7, 48–49. [Google Scholar]
  77. Li, H.L. Studies on Bio-Active Constituents of Corydalis saxicola Bunting and Toxic Constituents of Veratrum nigrum Linn; The Second Military Medical University: Shanghai, China, 2006. [Google Scholar]
  78. Liu, X.; Zheng, H.; Lu, R.; Huang, H.; Zhu, H.; Yin, C.; Mo, Y.; Wu, J.; Liu, X.; Deng, M. Intervening effects of total alkaloids of Corydalis saxicola Bunting on rats with antibiotic-induced gut microbiota dysbiosis based on 16S rRNA gene sequencing and untargeted metabolomics analyses. Front. Pharmacol. 2019, 10, 1151. [Google Scholar] [CrossRef]
  79. Guo, Z.M. Studies on Antihyperlipidemic and Antioxidative Activities of the Total Alkaloids Prepared from Corydalis saxicola in Rats; Sun Yat-sen University: Guangzhou, China, 2009; pp. 35–41. [Google Scholar]
  80. Shi, Y.J.; Xie, H.; Mao, C.Q.; Zhang, X.D.; Zheng, X.P. Intestinal transport characteristics of dehydrocavidine. Chin. Tradit. Pat. Med. 2013, 35, 256–260. [Google Scholar]
  81. Institue of Materia Medica Chinese Academy of Medical Science. Chinese Medicinal Materials; People’s Medical Publishing: Beijing, China, 1979; Volume 253. [Google Scholar]
  82. Bian, X.X.; Zhang, G.X.; Gao, X.L. Clinical observation of Corydalis thalictrifolia injection intreating chronic hepatic jaundice. J. Pract. Med. 2008, 24, 241. [Google Scholar]
  83. Liu, S.P.; Qu, T. Allergic reaction caused by Corydalis saxicola injection: A case report. Tradit. Chin. Drug. Res. Clin. Pharmacol. 2004, 15, 434. [Google Scholar]
  84. Li, J.Y.; Huang, J.J. Curative effect analysis of 464 cases of hepatitis treated with corydalis saxicola injection. Chin. Tradit. Pat. Med. 1987, 2, 19. [Google Scholar]
  85. Cong, C.Y. Study on the effect of Corydalis saxicola in treating jaundice hepatitis. In Proceedings of the Ninth National Conference of Infectious Diseases of Chinese Medical Association, Chengdu, Sichuan, 12–16 October 2006; p. 362. [Google Scholar]
  86. Wang, Y. Clinical observation of corydalis saxicola injection combined with interventional therapy for advanced liver cancer with hepatocellular jaundice. Jilin Med. J. 2010, 31, 1737–1738. [Google Scholar]
  87. Zhao, Y.F. Therapeutic effects of Yanhuanglian injection on acute cholestatic hepatitis. Chin. J. Clin. Ration. Drug Use 2009, 2, 20–21. [Google Scholar]
  88. Shao, X.J. Observation on clinical curative effect of Corydalis saxicolae injection in that treatment of icteric hepatitis b and nurse care. J. Qiqihua Med. Coll. 2006, 27, 112–113. [Google Scholar]
  89. Huang, W.Q.; Song, M.N.; Min, F.; Zhang, L.; Fan, R.H.; Luo, M.L.; Wu, W.B. Clinical study of Corydalis saxicola for icterohepatitis. Mod. Med. Health 2005, 21, 389. [Google Scholar]
  90. Zhong, H.X. Observation and nursing of triple treatment of chronic hepatitis B jaundice with chinese patent medicine. Mod. Nurs. 2005, 11, 1253–1254. [Google Scholar]
  91. Zhong, H.X.; Guo, X. Effect of treatment with combined traditional chinese and western medicine in chronic B hepatitis with jaundice. J. Nurs. Sci. 2005, 20, 29–30. [Google Scholar]
  92. Zheng, Z.X. Curative effect observation on 93 cases of chronic hepatitis B treated with Corydalis saxicola injection. Guide China Med. 2013, 5, 256–257. [Google Scholar]
  93. Li, S.X. Clinical Observation of corydalis saxicola in Treating Viral Hepatitis. China Foreign Med. Treat. 2010, 29, 104. [Google Scholar]
  94. Zhang, X.S.; Jiang, X.R. Clinical observation of Corydalis saxicola injection on acute and chronic viral hepatitis. China Healthc. Innov. 2009, 4, 10–17. [Google Scholar]
  95. Jiang, H.M. Analysis of therapeutic effect of Corydalis saxicola injection on 180 cases of chronic hepatitis B. World Health Dig. Med. Theory Praltice 2008, 4, 78. [Google Scholar]
  96. Zhao, S.H. Clinical observation on treatment of viral hepatitis with Corydalis saxicola Bunting. Sichuan Med. J. 2003, 24, 59. [Google Scholar]
  97. Ren, Z.X. Efficacy of injection of Corydalis saxicolae on 33 patients with viral hepatitis. Clin. Focus 2003, 18, 94–95. [Google Scholar]
  98. Deng, S.R.; Long, M.; Jiang, T.; Huang, Y.N. Treatment of severe jaundice of viral hepatitis with compound salvia miltiorrhiza combined with Corydalis saxicola injection. Chin. J. Primary. Med. Pharm. 2005, 12, 1630–1631. [Google Scholar]
  99. Wang, S.P.; Wang, X.H.; Dong, Y.Q.; Li, F. Clinical observation on treatment of 65 case of severe cholestasis of chronic hepatitis b with magnesium isoglycyrrhizinate and Corydalis saxicola Bunting. Hubei J. Tradit. Chin. Med. 2011, 33, 12–13. [Google Scholar]
  100. Zhang, W.W.; Yuan, X.H.; Zhu, L.; Tan, H.B.; Zhan, G.Q.; Li, R.G. Chronic severe hepatitis B in the treatment of telbivudine combined Yanhuanglian. Shaanxi. J. Tradit. Chin. Med. 2010, 31, 20–21. [Google Scholar]
  101. Yin, H. Effect of Corydalis saxicola injection combined with salvia miltiorrhiza injection on hepatic fibrosis in patients with chronic hepatitis B. J. Proactical Med. 2001, 17, 782–783. [Google Scholar]
  102. Zhou, Y.F.; Feng, Z.T.; Jiang, C.X. Effect of Corydalis saxicola decumbentis combined with radiointervention on liver function of patients with advanced liver cancer. Spec. Health 2020, 7, 112. [Google Scholar]
  103. Zhou, Y.F.; Feng, Z.T.; Jiang, C.X. Effect of Corydalis saxicola decumbentis combined with radiointervention on QOL score of patients with advanced liver cancer. Health Must-Read Mag. 2020, 33, 105. [Google Scholar]
  104. Feng, Z.T.; Jiang, C.X.; Zhou, Y.F.; Qin, P.; Qiu, A.Y. Effect of Corydalis saxicola decumbentis combined with radiointervention on levels of serum IL-2, IFN-γ, and TNF-α in patients with advanced hepatocellular carcinoma. Spec. Health 2020, 31, 74–75. [Google Scholar]
  105. Feng, Z.T.; Jiang, C.X.; Zhou, Y.F. Effect of Corydalis saxicola decumbentis combined with radiointervention on serum Th1/Th2 cytokines in patients with advanced liver cancer. Health Must-Read Mag. 2020, 33, 45. [Google Scholar]
  106. Qin, P. Efficacy of Corydalis saxicola injection in the repair of liver damage after interventional therapy for advanced hepatocellular carcinoma with hepatocellular jaundice. Health Must-Read Mag. 2020, 32, 218. [Google Scholar]
  107. Jiang, C.X.; Feng, Z.T.; Zhou, Y.F.; Qin, P.; Qiu, A.Y. Effect of Corydalis saxicola decumbentis combined with radiointervention on immune function of patients with moderate and advanced stage liver cancer. Spec. Health 2020, 8, 86. [Google Scholar]
  108. Jiang, C.X.; Feng, Z.T.; Zhou, Y.F. Effect of Corydalis saxicola bunge combined with radiointervention on adverse reactions in patients with advanced liver cancer. Spec. Health 2020, 31, 84. [Google Scholar]
  109. Qin, P.; Feng, Z.T.; Jiang, C.X.; Zhou, Y.F.; Qiu, A.Y. Effect of Corydalis saxicola combined with radiotherapy on improving quality of life of patients with advanced hepatocellular carcinoma. Sci. Regimen 2020, 9, 246. [Google Scholar]
  110. Zhang, Z.H.; Li, S.R.; Ma, G.Y.; Kong, X.D.; Chen, W.L.; Liu, Y.; Jia, L.L. Effects of Corydalis saxicola Bunting injection combined with transcatheter arterial chemoembolization on the immune function and life quality of patients with advanced liver cancer. Hebei Med. J. 2019, 41, 2902–2905. [Google Scholar]
  111. Sun, Z.C. Curative effect observation and nursing care of primary liver cancer treated with Corydalis saxicolae injection combined with interventional therapy. Qinghai Med. J. 2003, 33, 39–40. [Google Scholar]
  112. Yuan, W.P.; Hong, J.S.; Zhao, Y.N.; Chen, J.S. Treatment of liver damage after interventional chemoembolization of hepatocellular carcinoma with Corydalis saxicola Bunting. J. Guangxi Med. Univ. 2002, 19, 257. [Google Scholar]
  113. Qin, P.; Feng, Z.T.; Jiang, C.X.; Zhou, Y.F.; Qiu, A.Y. Efficacy and adverse reactions of octreotide combined with Corydalis saxicola injection in the treatment of advanced liver cancer. Spec. Health 2020, 23, 28. [Google Scholar]
  114. Zhang, X.H. 63 cases of neonatal hyperbilirubinemia treated with Corydalis saxicola Bunting. Shaanxi J. Tradit. Chin. Med. 2013, 34, 786–787. [Google Scholar]
  115. Gu, F.L.; Xu, S.S.; Shi, C.G. Therapeutic effects of Corydalis saxicola Bunting injection on advanced rectal cancer. China Mod. Doctor. 2020, 58, 107–109. [Google Scholar]
  116. Tai, Y.R.; Yin, M.S.; Pu, H.X.; Han, X.J. Observation of 60 hfrs cases cured with radix Corydalis Theaictrifoliae. Chin. J. Pract. Chin. Mod. Med. 2004, 4, 2746–2747. [Google Scholar]
  117. Wang, D.; Wang, Y.K. Comparison of therapeutic effects of shengmai and corydalis saxicola on liver cirrhosis. RenShen Yan Jiu 2002, 14, 28–29. [Google Scholar]
  118. Fang, S.X.; Dong, X.F.; Jie, S.H.; Ye, J. 40 cases of acute icteric hepatitis treated by Corydalis saxicola injection. Her. Med. 2001, 20, 567. [Google Scholar]
  119. Tang, Y.L. Clinical analysis of Corydalis saxicola combined with Yinzhihuang in treating icteric hepatitis. Med. Inf. 2009, 22, 2452–2453. [Google Scholar]
  120. Jiang, S.; You, S.G. Treatment of 33 case of icteric viral hepatitis with Corydalis saxicola Bunting and compound glycyrrhizin. Her. Med. 2008, 27, 403–404. [Google Scholar]
  121. Liu, L.; Zhang, G.H. Treatment of 31 cases of viral hepatitis and hyperbilirubinemia with Corydalis saxicola Bunting. Chin. J. Pharmacoepidemiol. 2001, 10, 182. [Google Scholar]
  122. Su, G.Q.; Wang, B.C.; Liu, S.F.; Li, J.; Ma, L.W.; Liu, L. Short-term therapeutic effect of Corydalis saxicola injection on chronic hepatitisb. Chin. J. Curr. Tradit. West. Med. 2004, 2, 117–118. [Google Scholar]
  123. Zhang, W.W.; Qin, M.H. 43 cases of chronic severe hepatitis treated with Corydalis saxicola Bunting and breviscapine injection. Shaanxi. J. Tradit. Chin. Med. 2006, 27, 21–23. [Google Scholar]
  124. Wan, S.C.; Gao, H.M. Treatment over 50 cases of chronic hepatitis B with GSH and Yanhuanglian. Pract. Clin. J. Integr. Tradit. Chin. West. Med. 2002, 2, 13. [Google Scholar]
  125. Xiong, L.G.; Li, L.L. Clinical observation of Octreodide combined with Corydalis in the treatment of advanced primary liver cancer. Sichuan J. Cancer Control 2005, 18, 232–234. [Google Scholar]
  126. Li, X.Y.; Tian, L.T. Treatment of 54 cases of hyperbilirubinemia with corydalis saxicola injection. Mod. Tradit. Chin. Med. 2003, 5, 30. [Google Scholar]
Figure 1. The pictures of Corydalis saxicola Bunting.
Figure 1. The pictures of Corydalis saxicola Bunting.
Ijms 24 01626 g001
Figure 2. Chemical structures isolated from Corydalis saxicola Bunting.
Figure 2. Chemical structures isolated from Corydalis saxicola Bunting.
Ijms 24 01626 g002aIjms 24 01626 g002bIjms 24 01626 g002cIjms 24 01626 g002d
Table 1. The phytoconstituents isolated from Corydalis saxicola Bunting.
Table 1. The phytoconstituents isolated from Corydalis saxicola Bunting.
NO.NameMolecular FormulaMolecular WeightPlant PartsReferences
1dehydrocavidineC21H20NO4350.14Whole plant[17]
2dehydroapocavidineC20H18NO4336.12Root[18]
3dehydroisoapocavidineC20H18NO4336.12Whole plant[15]
4berberineC20H18NO4336.12Whole plant[17]
5dehydroisocorypalmineC20H20NO4338.14Whole plant[15]
6coptisineC19H14NO4320.09Root[18]
7tetradehydroscoulerineC19H18NO4324.12Whole plant[15]
8dehydrocheilanthifolineC19H16NO4322.11Whole plant[19]
9palmatineC21H22NO4352.15Root[18]
10dehydrodiscretamineC19H18NO4324.12Whole plant[19]
11thalifaurineC19H16NO4322.11Root[18]
12jatrorrhizineC20H20NO4338.14Whole plant[13]
13berberrubineC19H16NO4322.11Whole plant[20]
14sanguinarineC20H14NO4332.09Whole plant[6]
15chelerythrineC21H18NO4348.12Whole plant[17]
166-acetonyl-5,6-dihydrosanguinarineC23H19NO5389.13Root[18]
17dihydrosanguinarineC20H15NO4333.10Root[18]
18dihydrochelerythrineC21H19NO4349.13Root[18]
198-acetonyldihydrochelerythrineC24H23NO5405.16Whole plant[6]
201-nitro-apocavidineC20H20N2O6384.13Whole plant[14]
212, 9, 10-thrihydroxy-3-methoxytetrahydroprotoberberine C18H19NO4313.13Whole plant[16]
22sinactineC20H21NO4339.15Whole plant[16]
23(−)-13β-hydroxystylopineC19H17NO5339.34Whole plant[17]
24tetrahydrocolumbamineC20H23NO4341.16Whole plant[17]
25canadineC20H21NO4339.15Whole plant[2]
26tetrahydropalmatrubineC20H23NO4341.16Whole plant[16]
27(−)-2,9-dihydroxyl-3,11-dimethoxy-1,10-dinitrotetrahydroprotoberberineC19H19N3O8417.12Whole plant[6]
28(+)-tetrahydropalmatineC21H25NO4355.15Whole plant[17]
29(−)-corynoxidineC21H25NO5371.17Whole plant[6]
30(−)-scoulerineC19H21NO4327.15Whole plant[17]
31(−)-cavidineC21H23NO4353.16Root[18]
32(+)-4-nitroisoapocavidineC20H20N2O6384.13Whole plant[6]
33(+)-1-nitroapocavidineC20H20N2O6384.13Whole plant[6]
34(±)-cavidineC21H23NO4353.16Whole plant[17]
35(+)-thalictrifolineC21H23NO4353.16Whole plant[21]
36(+)-cheilanthifolineC19H19NO4325.13Whole plant[16]
37corydalineC22H27NO4369.19Root[18]
38stylopineC19H17NO4323.12Root[18]
39mesotetrahydrocorysamineC20H19NO4337.13Whole plant[16]
40apocavidineC20H21NO4339.15Whole plant[16]
4113-β-hydroxystylopineC19H17NO5339.11Whole plant[16]
42(−)-tetrahydropalmatineC21H25NO4355.18Whole plant[19]
43protopineC20H19NO5353.13Whole plant[17]
44allocryptopineC21H23NO5369.16Whole plant[20]
45berbiniumC19H16NO4+322.11Whole plant[15]
461-formyl-5-methoxy-6-methylindolineC11H13NO2191.09Whole plant[15]
471-formyl-2-hydroxy-5-methoxy-6-methylindolineC11H13NO3207.09Whole plant[15]
48saxicolaline AC20H22NO5356.15Root[18]
49(+)-magnoflorineC20H24NO4342.17Root[18]
50(+)-isocorydineC20H23NO4341.16Whole plant[19]
51(−)-pallidineC19H21NO4327.15Whole plant[19]
52N-methylnarceimicineC22H22NO8428.13Root[18]
53adlumidineC20H17NO6367.11Root[18]
54(−)-salutaridineC19H21NO4327.15Root[18]
55cavidilinineC19H13NO4319.08Whole plant[20]
56corypallineC11H15NO2193.11Whole plant[19]
57oxyacanthineC37H40N2O6608.29Whole plant[22]
58corydalisin AC37H38N2O9654.26aerial parts[1]
59corydalisin BC38H40N2O10684.27aerial parts[1]
60cannabisin FC36H36N2O8624.25aerial parts[1]
61corydalisin CC38H40N2O10684.27aerial parts[1]
62cannabisin DC36H36N2O8624.25aerial parts[1]
631,2-dihydro-6,8-dimethoxy-7-hydroxyl-1-(3,5-dimethoxy-4-hydroxyphenyl)-N1, N2-bis[2-(4-hydroxyphenyl)ethyl]-2,3-naphthalene dicarboxamideC38H40N2O10684.27aerial parts[1]
64cannabisin EC36H38N2O9642.26aerial parts[1]
65grossamideC36H36N2O8624.25aerial parts[1]
66feruloylagmatineC15H22N4O3306.17Whole plant[19]
67corysaxicolaine AC39H30N2O8654.20aerial parts[7]
68β-sitosterolC29H50O414.39Whole plant[22]
69daucosterolC35H60O6576.44Whole plant[22]
70betulin C30H50O2484.43Whole plant[22]
71betulinic acidC30H48O3498.41Whole plant[22]
72β-amyrinC30H50O426.39Whole plant[20]
73β-amyrin acetateC32H52O2468.40Whole plant[22]
74(+)-oleanolic acidC30H48O3454.38Whole plant[22]
75cholesterolC27H46O386.35Whole plant[23]
76cycloeucalenolC30H50O426.39Whole plant[22]
775-hydroxy-3′, 4′, 6, 7-tetramethoxyflavoneC19H18O7358.11Whole plant[20]
78quercetin-3-O-β-D-galactosideC21H20O12461.10Whole plant[20]
79tetra-amino-27 alkaneC27H57N395.45Whole plant[23]
80tetra-amino-28 alkaneC28H59N409.46Whole plant[23]
81uracilC4H4N2O2112.03Whole plant[20]
Table 2. Pharmacological activities of Corydalis saxicola Bunting.
Table 2. Pharmacological activities of Corydalis saxicola Bunting.
Tested SubstanceAssay, Organism, or Cell LineBiological ResultsReferences
Anticancer activityCSBTAWalker 256 induced bone pain and osteoporosis in rats, Breast Cancer Cells, RAW 264.7 macrophage cells, Walker 256 cellsImproved bone pain and osteoporosis in rats, suppressed expression of Rankl, down regulated the ratio of RANKL/OPG, inhibited pathways of NF-κB and c-Fos/NFATc1 to suppressed osteoclast formation[25]
CSBTAA549 cellsInhibition migration of A549 cells by suppressing Cdc42 or Vav1[8]
CSBTAA549 cellsInhibited tumor growth by down-regulating Survivin, reduced the degree of bone destruction[26]
CSBTAA549 cellsInhibited the migration ability of A549 cells, decreased the expression of Cdc42 protein[27]
CSBTAA549 cellsInhibition proliferation, induced apoptosis and up regulation of caspase and down of survivin [28]
CSBTA and cis-platinumA549 cellsInhibition proliferation[29]
CSBTATca8113 cellsInduced apoptosis and suppression of Bcl-2[30]
CSBTATca8113 cellsInhibition proliferation, induced apoptosis[31]
CSBTATca8113 cellsInhibition proliferation and telomerase activity[32]
CSBTACNE-1112.41 µg/mL (IC50)[23]
CSBTACNE-2123.46 µg/mL (IC50)[23]
CSBTAA2780148.40 µg/mL (IC50)[23]
CSBTASKOV3128.51 µg/mL (IC50)[23]
CSBTAPM2166.66 µg/mL (IC50)[23]
CSB injectionmouse sarcoma S180The average tumor inhibition rate was more than 30%[33]
CSB injectionehrlich ascites tumorThe average tumor inhibition rate was more than 30%[33]
CSB injection and aristolochic acidmouse sarcoma S180The average tumor inhibition rate was more than 50%[33]
CSB injection and aristolochic acidehrlich ascites tumorThe average tumor inhibition rate was more than 50%[33]
CSB injectionmouse sarcoma S180Some inhibition and inhibited respiration[17]
CSB injectionehrlich ascites tumorSome inhibition and inhibited respiration[17]
CSB injectionliver cancerSome inhibition and inhibited respiration[17]
CSB injectionascites cancerSome inhibition and inhibited respiration[17]
CSB injectionrat sarcoma 256Some inhibition and inhibited respiration[17]
CSB injectionmouse peritoneal macrophagesSignificantly enhanced phagocytosis[17]
CSB injectionS180Inhibition of respiratory metabolism[34]
CSB injectionHACInhibition of respiratory metabolism[34]
CSB injectionEACInhibition of respiratory metabolism[34]
CSB injectionS180, HAC, EAC, W256, ratSignificantly inhibition[35]
CSB injectionS180, HAC, EAC, W256Killing effect[36]
aqueous extractHepG2Inhibition proliferation and migration[25]
Corydalisin AMGC-80383.56 ±1.89 μM (IC50)[1]
Corydalisin AHepG2> 100 μM (IC50)[1]
Corydalisin AT24> 100 μM (IC50)[1]
Corydalisin ANCI-H460> 100 μM (IC50)[1]
Corydalisin ASpca-2> 100 μM (IC50) [1]
Corydalisin BMGC-803> 100 μM (IC50)[1]
Corydalisin BHepG2> 100 μM (IC50)[1]
Corydalisin BT24> 100 μM (IC50)[1]
Corydalisin BNCI-H460> 100 μM (IC50)[1]
Corydalisin BSpca-2> 100 μM (IC50)[1]
Corydalisin CMGC-8038.81 ±2.05 μM (IC50)[1]
Corydalisin CHepG222.23 ±1.85 μM (IC50)[1]
Corydalisin CT249.62 ±1.46 μM (IC50)[1]
Corydalisin CNCI-H46025.79 ±1.04 μM (IC50)[1]
Corydalisin CSpca-217.28 ±1.29 μM (IC50)[1]
Cannabisin FMGC-80310.10 ±1.15 μM (IC50)[1]
Cannabisin FHepG238.93 ±1.07 μM (IC50)[1]
Cannabisin FT2411.54 ±1.49 μM (IC50)[1]
Cannabisin FNCI-H46030.96 ±1.27 μM (IC50)[1]
Cannabisin FSpca-222.23 ±1.44 μM (IC50)[1]
Cannabisin EMGC-803> 100 μM (IC50)[1]
Cannabisin EHepG2> 100 μM (IC50)[1]
Cannabisin ET2446.54 ±1.62 μM (IC50)[1]
Cannabisin ENCI-H460> 100 μM (IC50)[1]
Cannabisin ESpca-2> 100 μM (IC50)[1]
Cannabisin DMGC-803> 100 μM (IC50)[1]
Cannabisin DHepG2> 100 μM (IC50)[1]
Cannabisin DT24> 100 μM (IC50)[1]
Cannabisin DNCI-H460> 100 μM (IC50)[1]
Cannabisin DSpca-2> 100 μM (IC50)[1]
1,2-dihydro-6,8-dimethoxy-7-hydroxy-1-(3,5-dimethoxy-4- hydroxyphenyl)-N 1, N 2 -bis [2-(4-hydroxyphenyl) ethyl]-2,3-naphthalene dicarboxamideMGC-803;
HepG2;T24;
NCI-H460;
Spca-2
55.16 ±0.78 μM (IC50);
> 100 μM (IC50);
48.15 ±1.09 μM (IC50);
> 100 μM (IC50);
43.89 ±1.57 μM (IC50)
[1]
grossamideMGC-80326.95 ±1.24 μM (IC50)[1]
grossamideHepG240.75 ±0.88 μM (IC50)[1]
grossamideT2421.19 ±1.53 μM (IC50)[1]
grossamideNCI-H46036.38 ±1.39 μM (IC50)[1]
grossamideSpca-227.22 ±1.72 μM (IC50)[1]
DehydrocavidineSMMC-7721Significantly inhibition[37]
palmatineSMMC-7721Significantly inhibition[37]
DehydrocavidineTca8113Significantly inhibition, suppression of NF-kappa B, P50 and P60[38]
CSBTATca8113Significantly inhibition, suppression of NF-kappa B, P50 and P60[38]
DehydrocavidineTca8113Inhibition proliferation, telomerase activity and the expression of hTERT[39]
PallidineDNA topoisomerase IStrong inhibitory effect on human DNA topoisomerase I[19]
scoulerineDNA topoisomerase IStrong inhibitory effect on human DNA topoisomerase I[19]
chelerythrineunknownHave certain anticancer effect[21]
(−)-13β-hydroxystylopineunknownHave certain anticancer effect[21]
Corysaxicolaine AT247.63 μM (IC50)[7]
Corysaxicolaine AA54913.32 μM (IC50)[7]
Corysaxicolaine AHepG212.39 μM (IC50)[7]
Corysaxicolaine AMGC-8039.98 μM (IC50)[7]
Corysaxicolaine ASKOV312.36 μM (IC50)[7]
Hepatoprotective effectsCSBTAratsInterventional treatment of chronic liver injury[40]
CSBTAHSC-T6Induced apoptosis and autophagy[41]
CSBTACYP450s in ratsCYP1A2 (IC50, 38.08 μg/mL; Ki, 14.3 μg/mL), CYP2D1 (IC50, 20.89 μg/mL; Ki, 9.34 μg/mL), CYP2C6/11 (IC50 for diclofenac and S-mephenytoin, 56.98 and 31.59 μg/mL; Ki, 39.0 and 23.8 μg/mL), CYP2B1 (IC50, 48.49 μg/mL; )Ki, 36.3 μg/mL)[42]
CSBTAchronic hepatotoxicity in ratsRestored the levels of 2-oxoglutarate, citrate, hippurateand taurine[43]
CSBTAacute hepatic injury ratsSignificantly reduced the content of AST, ALT [44]
chronic hepatic injury ratsSignificantly increased the level of serum TP, reduced the content of AST, ALT, AKP, LN and HA[45]
CSBTAimmune hepatic injury ratReduced serum GOT activity, IL-4, increased the rate of IFN-γ/IL-4[46]
CSBTAratsHave certain preventive and therapeutic effect on acute liver injury and on chronic liver fibrosis[47]
CSBTAratsObvious protective effect on acute liver injury, inhibited the formation of chronic liver fibrosis[48]
CSBTAhepatic fibrosis ratsInhibited the expression of TGF-β1 and MMP-9[49]
CSBTAacute hepatic injury ratsIncreased the content of AST, ALT and SOD,
reduced MDA
[50]
aqueous extractliver fibrosis in ratsRegulated the level of some amino acids, identified 157 potential targets of CS and265 targets of liver fibrosis[51]
aqueous extractacute hepatic injury ratsImproved deviations of metabolites (soleucine, alanine, glutamine, phosphocholine and glycerol)[52]
aqueous extractacute hepatic injury ratsIncreased the content of AST, ALT and SOD,
reduced MDA
[53]
aqueous extractacute hepatic injury ratsReduced the contents of AST and ALTpromote the production of mouse hemolysin antibody
LD50 = 298.5 mg·kg-1
[54]
aqueous extractacute hepatic injury ratsReduced the content of AST and ALT[55]
DehydrocavidineHSC-T6Inhibition proliferation, induced apoptosis[56]
palmatineHSC-T6Inhibition proliferation, induced apoptosis[56]
berberineHSC-T6Inhibition proliferation, induced apoptosis[56]
Dehydrocavidinehepatic fibrosis ratsReduced hepatic hydroxyproline,
increases urinary hydroxyproline
[57]
Dehydrocavidineliver injury in ratsDown regulated EPHX2, HYOU1, GSTM3, Sult1a2 and P450, reduce free radical, lose weight, MDA, ALT, AST, ALP and TBIL[58]
Dehydrocavidineliver injury in ratsIncreased ALT, AST and TBIL, Reduces the inflammatory cell infiltration of cell degeneration and necrosis and damages the ultrastructure of liver cells[59]
Anti-HBV activityextractDuck hepatitis B virusReduced DHBV-DNA[60]
total extract of rootHBsAg0.17 mg/mL (IC50)[18]
total extract of rootHBeAg<0.04 mg/mL (IC50)[18]
Saxicolalines AHBsAg2.19 μM (IC50)[18]
Saxicolalines AHBeAg>2.81μM (IC50)[18]
N-methylnarceimicineHBsAg1.22 μM (IC50)[18]
N-methylnarceimicineHBeAg1.84 μM (IC50)[18]
6-acetonyl-5,6-dihydrosanguinarineHBsAg6.55 μM (IC50)[18]
6-acetonyl-5,6-dihydrosanguinarineHBeAg>2.54 μM (IC50)[18]
dihydrochelerythrineHBsAg<0.05 μM (IC50)[18]
dihydrochelerythrineHBeAg<0.05 μM (IC50)[18]
adlumidineHBsAg1.35 μM (IC50)[18]
adlumidineHBeAg>2.73 μM (IC50)[18]
(−)-salutaridineHBsAg0.26 μM (IC50)[18]
(−)-salutaridineHBeAg0.43 μM (IC50)[18]
palmatineHBsAg>4.26 μM (IC50)[18]
palmatineHBeAg>4.26 μM (IC50)[18]
protopineHBsAg2.61 μM (IC50)[18]
protopineHBeAg>4.25 μM (IC50)[18]
coptisineHBsAg2.74 μM (IC50)[18]
coptisineHBeAg3.19 μM (IC50)[18]
(+)-magnoflorineHBsAg>4.39 μM (IC50)[18]
(+)-magnoflorineHBeAg>4.39 μM (IC50)[18]
dehydrocheilanthifolineHepG2.2.15115.95 μM (CC50)[3]
dehydrocheilanthifolineHBsAg15.84 ± 0.36 μM (IC50)[3]
dehydrocheilanthifolineHBeAg17.12 ± 0.45 μM (IC50)[3]
dehydrocheilanthifolineExtracellular DNA15.08 ± 0.66 μM (IC50)[3]
dehydrocheilanthifolineIntracellular DNA7.62 ± 0.24 μM (IC50)[3]
dehydrocheilanthifolineIntracellular cccDNA8.25 ± 0.43 μM (IC50)[3]
Crude extractHBsAg0.16 mg/mL (IC50)[61]
Crude extractHBeAg< 0.04 mg/mL (IC50)[61]
6-acetonyl-5, 6-dihydrosanguinarineHBsAg0.65 mg/mL (IC50)[61]
6-acetonyl-5, 6-dihydrosanguinarineHBeAg>1.00 mg/mL (IC50)[61]
dihydrochelerythrineHBsAg<0.02 mg/mL (IC50)[61]
dihydrochelerythrineHBeAg<0.02 mg/mL (IC50)[61]
adlumidineHBsAg0.50 mg/mL (IC50)[61]
adlumidineHBeAg>1.00 mg/mL (IC50)[61]
(−)-salutaridineHBsAg0.09 mg/mL (IC50)[61]
(−)-salutaridineHBeAg0.15 mg/mL (IC50)[61]
palmatineHBsAg>1.50 mg/mL (IC50)[61]
palmatineHBeAg>1.50 mg/mL (IC50)[61]
protopineHBsAg0.92 mg/mL (IC50)[61]
protopineHBeAg>1.50 mg/mL (IC50)[61]
coptisineHBsAg0.88 mg/mL (IC50)[61]
coptisineHBeAg>1.02 mg/mL (IC50)[61]
(+)-magnoflorineHBsAg>1.50 mg/mL (IC50)[61]
(+)-magnoflorineHBeAg1.50 mg/mL (IC50)[61]
dehydrocavidineHBsAg33% inhibition [62.5 μg/mL][15]
dehydrocavidineHBeAg22% inhibition [62.5 μg/mL][15]
dehydroapocavidineHBsAg39% inhibition [62.5 μg/mL][15]
dehydroapocavidineHBeAg24% inhibition [62.5 μg/mL][15]
dehydroisoapocavidineHBsAg29% inhibition [62.5 μg/mL][15]
dehydroisoapocavidineHBeAg23% inhibition [62.5 μg/mL][15]
berberineHBsAg8% inhibition [62.5 μg/mL][15]
berberineHBeAg7% inhibition [62.5 μg/mL][15]
dehydroisocorypalmineHBsAg6% inhibition [62.5 μg/mL][15]
dehydroisocorypalmineHBeAg6% inhibition [62.5 μg/mL][15]
coptisineHBsAg6% inhibition [62.5 μg/mL][15]
coptisineHBeAg9% inhibition [62.5 μg/mL][15]
tetradehydroscoulerineHBsAg7% inhibition [62.5 μg/mL][15]
tetradehydroscoulerineHBeAg6% inhibition [62.5 μg/mL][15]
berbiniumHBsAg9% inhibition [62.5 μg/mL][15]
berbiniumHBeAg6% inhibition [62.5 μg/mL][15]
1-formyl-5-methoxy-6-methylindoline HBsAg2% inhibition [62.5 μg/mL][15]
1-formyl-5-methoxy-6-methylindoline HBeAg7% inhibition [62.5 μg/mL][15]
1-formyl-2-hydroxy-5-methoxy-6-methylindolineHBsAg5% inhibition [62.5 μg/mL][15]
1-formyl-2-hydroxy-5-methoxy-6-methylindolineHBeAg3% inhibition [62.5 μg/mL][15]
Enhancement of immune functionCSBTAratsCSBTA (40 μg/mL) began to enhance, enhanced the levels of T cell production of IFN-γ and IL-2 [62]
Antioxidant activityCSBTAratsReduced the of content MDA and increase SOD activity, enhance the antioxidant capacity of rat liver[63]
cavidineDPPH assay6.85 mg/mL (IC50)[2]
cheilanthifolineDPPH assay0.25 mg/mL (IC50)[2]
tetrahydropalmatineDPPH assay3.79 mg/mL (IC50)[2]
stylopineDPPH assay2.56 mg/mL (IC50)[2]
canadineDPPH assay2.18 mg/mL (IC50)[2]
dehydrocavidineDPPH assay16.51 mg/mL (IC50)[2]
dehydrocheilanthifolineDPPH assay1.63 mg/mL (IC50)[2]
berberineDPPH assay7.40 mg/mL (IC50)[2]
pallidineDPPH assay1.00 mg/mL (IC50)[2]
Effects on the central nervous systemCSBTAratsReduced the content of DOPAC, HVA, 5-HT and 5-HIAA, the level of DA has no effect (50, 100 mg/kg CSBTA)[64]
CSBTAratsReduced activity (25 mg/kg CSBTA)[65]
CSBTAmonkeyReduced activity (12 mg/kg CSBTA)[65]
CSBTAcatsReduced activity (10-15 mg/kg CSBTA)[65]
CSBTAratsReduced irritated response (50 mg/kg CSBTA)[65]
CSBTArats77% suppressed conditional emission (50 mg/kg CSBTA)[65]
CSBTAratsIncreased the hypnotic time of pentobarbital sodium by more than 2 to 4 times (25 mg/kg CSBTA)[65]
CSBTArabbitActivity slow down (20-30 mg/kg CSBTA)[65]
CSBTAratsLD50 = 223 mg/kg[65]
CSBTAratsReduced the arthritis (50 mg/kg CSBTA)[66]
DehydrocavidineratsReduced the content of DA and HVA[67]
DohydrocyaidineratsReduced spontaneous activity[68]
DohydrocyaidineratsSynergistic effect with barbiturates[68]
Anti-inflammatory activityCSBTAM1 macrophages Obvious toxic effect on the activity of M1-Mφ, significantly reduced the mRNA level of IL-6, TNF-α, CD86, IL-1β[69]
CSBTAratsSignificantly inhibited the addition of capillary permeability, and suppressed exudation, edema and connective tissue hyperplasia[70]
Corydalis saxicola suppositorychronic pelvic inflammatory disease model ratsSignificantly inhibited the uterine swelling, significantly reduced the spleen index, hemameba, neutrophil, TNF-α, IL-6 and MDA, improved thymus index, ovary index, lgG, lgM and SOD[71]
Corydalis saxicola rectal suppositoryratsObvious inhibited ear swelling, the addition of capillary permeability, and writhing reaction in rat[72]
Analgesic effectCSBTAratsReduced the level of proinflammatory cytokines, such as TNF-α, IL-1β and PGE2. inhibited the overexpression level of DRG, TG, p-p38 and TRPV1[73]
CSBTAratsInhibited the "writhing reaction" in rat (50 mg/kg CSBTA), improve the "pain closure" of rats to heat stimulation (100 mg/kg CSBTA)[66]
DohydrocyaidineratsThe effects of sedative, analgesic, and spasmolysis, LD50 = 71.6±2.92 mg/kg[68]
deheydrocavidineunknownHave certain sedative and analgesic effects[21]
Antibacterial activityCSBTAstaphylococcus aureus17.8 mg/mL (MIC) [74]
CSBTAstaphylococcus aureus70.0 mg/mL (MBC) [74]
CSBTAstreptococcus pyogenes20.5 mg/mL (MIC) [74]
CSBTAstreptococcus pyogenes70.0 mg/mL (MBC) [74]
CSBTAstreptococcus faecalis17.8 mg/mL (MIC) [74]
CSBTA 70.0 mg/mL (MBC) [74]
CSBTAescherichia coli35.5 mg/mL (MIC) [74]
CSBTA 70.0 mg/mL (MBC) [74]
CSBTApseudomonas aeruginosa70.0 mg/mL (MIC) [74]
CSBTA >300 mg/mL (MBC) [74]
CSBTAshigella flexneri16.8 mg/mL (MIC) [74]
CSBTA 35.5 mg/mL (MBC) [74]
CSBTAsalmonella typhi35.5 mg/mL (MIC) [74]
CSBTA 70.0 mg/mL (MBC) [74]
CSBTAsalmonella enteritidis16.8 mg/mL (MIC) [74]
CSBTA 70.0 mg/mL (MBC) [74]
CSBTAklebsiella pneumoniae70.0 mg/mL (MIC) [74]
CSBTA 130.0 mg/mL (MBC) [74]
CSBTAproteus70.0 mg/mL (MIC) [74]
CSBTA 130.0 mg/mL (MBC) [74]
CSBTAcandida albicans130.0 mg/mL (MIC) [74]
CSBTA > 300 mg/mL (MBC) [74]
extractstaphylococcus aureus20.0 mg/mL (MIC) [75]
extract +Cefradinestaphylococcus aureus0.375 (FIC) synergistic effect[75]
extract + Levofloxacinstaphylococcus aureus0.5 (FIC) synergistic effect[75]
extract + Fosfomycinstaphylococcus aureus1.5 (FIC) irrelevant effect[75]
extract + Penicillinstaphylococcus aureus0.375 (FIC) synergistic effect[75]
dehydrocarvidinegram-positive strains;
gram-negative bacterium
Have certain inhibitory effect on gram-positive strains, minimum concentration is 0.078 mg/mL, and has no inhibitory effect on gram-negative bacteria[76]
deheydrocavidinestaphylococcus aureus;
beta hemolytic streptococcus;
corynebacterium diphtheriae;
penicillin-resistant white staphylococcus aureus
Have an inhibiting effect[21]
dohydrocyaidineratsAntibacterial effect[68]
ChOleretic effectsCSBTAguinea pigBile secretion is temporarily reduced[66]
extractratsIncreased the amount of bile excretion[77]
Other activitiesCSBTAratsIntervention of host co-metabolism and intestinal flora in rats with intestinal flora imbalance[78]
CSBTAratsSignificantly decreased the levels of plasma total cholesterol and low-density lipoprotein cholesterol in rats, regulated blood lipid levels in high-fat diet rats[79]
DehydrocarvidineratsThe transport of dehydrocavidine was carried out in vitro at different intestine segments[80]
DehydrocarvidineCaco-2 cellsThe bi-directional transport of dehydrocavidine in Caco-2 monolayer model showed significant difference[80]
Table 3. The clinical application of Corydalis saxicola Bunting.
Table 3. The clinical application of Corydalis saxicola Bunting.
ClassDrugsCasesResultAdverse ReactionReferences
Icteric hepatitisCSBIunknowThe total effective rate of the treatment group was 87.5%, the control group was 62.5% (p < 0.01) [85]
CSBI42Significantly decreased contents of ALT, AST, γ-GT and TBIL (p < 0.05). [86]
CSBI60The clinical symptoms and liver function in the treatment group were better than those in the control group (p < 0.05) [87]
CSBI82Significantly improved symptoms of fatigue, abdominal distension, hepatalgia and poor appetite; obvious decrease of transaminase and bilirubin [82]
CSBI98Improvement rate of poor appetite, hepatalgia, fatigue and abdominal distension was 85.7%, 84.4%, 76.8% and 87.8%, respectively, summary improvement is 83.4%. Significantly decreased contents of ALT, TBIL [88]
CSBI29The total effective rate of the treatment group was 93.1%, the control group was 71.0% (p < 0.05) [89]
CSBI1Caused allergic reactionsItching, palpitating, chills, fever[83]
Salvia miltiorrhiza Bge & Yinzhihuang & CSB90The total effective rate of the treatment group was 96.0%, the control group was 82.5% (p < 0.05) [90]
Salvia miltiorrhiza Bge & Yinzhihuang & CSB90The total effective rate of the treatment group was 96.0%, the control group was 82.5% (p < 0.05)Precardiac discomfort, urticaria, skin itching, pain at the injection site[91]
Viral hepatitisCSBI93The total effective rate of the treatment group was 91.7%, the control group was 68.9% (p < 0.03)There were no adverse reactions[92]
CSBI50Significantly decreased ALT and TBIL (p < 0.05), the total effective rate of the treatment group was 94.0% [93]
CSBI208TBil of treatment group dropped by 71.22%, the control group was 44.30% (p < 0.01); Dbil of treatment group dropped by 67.53% (p < 0.01)low-grade fever[94]
CSBI360Significant difference in improvement rate of hepatalgia and poor appetite (p < 0.01), decreased the levels of T-BILI, D-BILI and ALT [95]
CSBI60The total effective rate of the treatment group was 88.23%, the control group was 76.92% (p < 0.05)Local vascular pain[96]
CSBI33Decreased the contents of ALT and AST in a short time, improved protein metabolism [97]
Compound Danshen & CSB100The total effective rate of the treatment group was 96.14%, the control group was 64.58% (p < 0.01)Mild rash[98]
Acute and chronic hepatitisCSBI93The total effective rate of the treatment group was 91.7%, the control group was 68.9% (p < 0.03)There were no adverse reactions[92]
Magnesium isoglycyrrhizinate & CSB65The total effective rate of the treatment group was 89.23%, the control group was 70.14% (p < 0.05) [99]
CSB & Telbivudine80The total effective rate of the treatment group was 72.5%, the control group was 50% (p < 0.05) [100]
CSB & Danshen injection70Significantly decreased ALT and TBIL, increased the ratio of A/G [101]
Liver cancerCSBI96The total effective rate of the treatment group was 83.3%, the control group was 72.9% (p < 0.05), Significantly decreased ALT and AST [102]
CSBI96The total effective rate of the treatment group was 83.3%, the control group was 72.9% (p < 0.05), the QOL of the treatment group (68.6±7.2) more than the control group (60.5±6.1) after treatment (p < 0.05) [103]
CSBI96The total effective rate of the treatment group was 79.2%, the control group was 50.0% (p < 0.05), effectively improve the levels of serum IL-2, IFN-γ and TNF-α, better than the control group [104]
CSBI96The total effective rate of the treatment group was 81.3%, the control group was 70.8% (p < 0.05), after treatment, the INF-γ, IL-4 and the ratio of INF-γ/IL-4 in the observation group were significantly better than those in the control group (p < 0.05) [105]
CSBI120After treatment, the liver function recovery effect of the injection group was better than that of the control group (p < 0.05)fever, skin itch;
the injection group (6.7%), the control group (8.3%)
[106]
CSBI96The total effective rate of the treatment group was 72.9%, the control group was 52.1% (p < 0.05) [107]
CSBI96The total effective rate of the treatment group was 83.3%, the control group was 64.4% (p < 0.05)The adverse effects rate of the treatment group was 20.8%, the control group was 39.6% (p < 0.05)[108]
CSBI96The total effective rate of the treatment group was 97.92%, the control group was 87.50% (p < 0.05)The adverse effects rate of the treatment group was 10.42%, the control group was 39.6% (p < 0.05)[109]
CSBI110Quality of life improvement rate in the treatment group was 81.82%, the control group was 52.73% (p < 0.05) [110]
CSBI42Significantly decreased the levels of ALT, AST, TBIL and γ-GTFever, shiver, skin itch[86]
CSBI60Significantly decreased the level of AFP, the white blood cell count goes up [111]
CSBI46Significantly increased the level of ALT and AST (p < 0.05), decreased TBIL (p > 0.05) [112]
CSBI & Octreotide116The total effective rate of the treatment group was 96.4%, the control group was 92.9% (p < 0.05)The adverse effects rate of the treatment group was 5.2%, the control group was 17.2% (p < 0.05)[113]
HyperbilirubinemiaCSBI126The total effective rate of the treatment group was 95.3%, the control group was 93.6% (p > 0.05)Diarrhea, rash [114]
Others
Rectal cancerCSBI68 The total effective rate of the treatment group was 38.24%, the control group was 5.88% (p < 0.05) [115]
Hemorrhagic fever with renal syndromeCSBI & Ribavirin60Significantly decreased the duration of fever period and oliguria period, obvious the recovery of AST, ALT, LDH and BUN than the control group (p < 0.05) [116]
Liver cirrhosisCSBI 60Significantly effect in relieving symptoms, protecting liver and gallbladder [117]
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

Qin, F.; Chen, Y.; Wang, F.-F.; Tang, S.-Q.; Fang, Y.-L. Corydalis saxicola Bunting: A Review of Its Traditional Uses, Phytochemistry, Pharmacology, and Clinical Applications. Int. J. Mol. Sci. 2023, 24, 1626. https://doi.org/10.3390/ijms24021626

AMA Style

Qin F, Chen Y, Wang F-F, Tang S-Q, Fang Y-L. Corydalis saxicola Bunting: A Review of Its Traditional Uses, Phytochemistry, Pharmacology, and Clinical Applications. International Journal of Molecular Sciences. 2023; 24(2):1626. https://doi.org/10.3390/ijms24021626

Chicago/Turabian Style

Qin, Feng, Yao Chen, Fan-Fan Wang, Shao-Qing Tang, and Yi-Lin Fang. 2023. "Corydalis saxicola Bunting: A Review of Its Traditional Uses, Phytochemistry, Pharmacology, and Clinical Applications" International Journal of Molecular Sciences 24, no. 2: 1626. https://doi.org/10.3390/ijms24021626

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop