Skip to main content

Mechanism-based biomarkers for the quality control of Dangkwisoo-san: a scoping review

Abstract

Dangkwisoo-san (DS) is a traditional Korean herbal medicine used to treat traumatic diseases, including pulmonary contusions, traumatic pneumothorax, bruising, and ankle sprain. Quality control (QC) biomarkers for DS can help ensure its safety and efficacy. Although chemical quality assessments are performed to ensure consistent efficacy of DS, the identity and quantity of the compounds contained within a given natural product is a frequent complication. We conducted a literature review to identify biological assays that support the chemical QC of DS. The results of our investigation confirmed that in vitro experiments with aqueous and alcoholic extracts of DS exhibited positive effects on many aspects of treatment. With 80% EtOH extraction, a low concentration of DS (1 μg/ml) significantly diminished the expression of inflammatory factors, such as nitric oxide (NO), TNF-α, IL-1β, and IL-6, in the Raw264.7 cell line. MeOH extracts activated NRF2 and antioxidant activities in response to the inflammatory inducer LPS, and water extracts of DS remarkably reduced proinflammatory cytokine levels compared to dexamethasone and cyclosporin treatments. Aqueous extracts of DS at a moderate dose of 125 μg/ml supported bone regeneration, recovered ischemic injury in an eNOS-dependent manner, and prevented metabolic disorders (TRPM7 channel inhibition). Cytokines, NO, and immunoglobulins are potential biological QC biomarkers to assess the anti-inflammation and immune response to DS. Future quality evaluation studies of herbal medicines (herbal prescriptions) should aim to select the mechanism-based in vitro efficacy evaluation methods that can estimate consistent clinical effects.

Introduction

The use of complementary and alternative medicines, including herbal medicines, has been increasing worldwide [1, 2]. Distinguished from synthetic drugs, which are targeted toward specific acute disease symptoms and pathologies, botanical drugs target a wide range of physiological processes for long-term therapies [3, 4]. Many traditional therapy systems have recorded theoretical knowledge and practical applications, such as prevention, diagnosis, and management of physical and mental disorders. Overall, 80% of the world’s population residing in developing countries relies on herbal products as a primary source of pharmaceutical drugs [5,6,7].

Although significantly consumed worldwide, herbal medicines are facing difficulties to reach the new drug application (NDA) step of the Food and Drug Administration (FDA) [8]. To put this into perspective, the key issue of botanical products is to guarantee “therapeutic consistency” and present clinical evidence consistent with standard treatments in phase III clinical trials [9]. Since the FDA approval of Veregen, a green tea-derived drug, for genital wart treatment [8], few botanical drugs have met these criteria. Traditional herbal medicines contain certain extracts and compounds that affect multiple targets [10, 11], and many of them may not be identified. Typical pharmacokinetic studies cannot cover the active combinations and conflicts among them. To address this difficulty, the co-treatment of herbal medicines with commercialized drugs has been established. Recently, the combination of nelfinavir, an FDA-approved drug that effectively inhibits SARS-CoV-2, and the natural origin drug cepharanthine showed enhanced efficacy [12, 13]. Another line of research is the study of the active components of herbal remedies to eliminate potential risks. Exelon and Razadyne are herb-derived compounds that are FDA-approved for clinical trials against Alzheimer’s disease [14].

For the widespread use of natural origin prescriptions, the mode of action of these drugs demands supporting scientific rationale. Unlike the vertebrate immune system, plants use phytochemical metabolites as a defense mechanism. They are rich in phenolic compounds, such as alkaloids, coumarins, flavonoids, polyphenols, and tannins, which exhibit efficacy against health disorders and pathogenic organisms [15]. These herbal substances stimulate secondary messenger systems, resulting in the activation of transcription factors of certain genes in response to specific stimulants against illness or diseases [16]. Secondary messengers, including cyclic nucleotides, lipid derivatives, ions, and intracellular signals, are small and rapidly broadcasting molecules present during signal transduction [17]. Among them, the agonist–G-protein-coupled receptor (GPCR) complex activates the adenosine 3′,5′-cyclic monophosphate (cAMP) pathway [18] and leads to the formation of phosphatidylinositol 3,4,5-trisphosphate (PIP3) in response to growth factors and receptor tyrosine kinase (RTK) binding [19], forming a notable messenger system that amplifies the signal following intracellular and extracellular stimulation.

Chemical quality control (QC) refers to the assessment of the chemical composition and purity of a drug or herbal medicine. Although chemical QC is important for ensuring the safety and efficacy of drugs, it has some limitations [20]. Chemical analysis does not always reflect biological activity: a compound may be present in high concentrations in a sample, but it may not be the active ingredient responsible for the biological effects of the drug. Therefore, relying solely on chemical analysis may not provide an accurate assessment of the biological activity of a drug. Chemical analysis may miss important components; some active ingredients or biomolecules may be present in small quantities or may not be amenable to detection by chemical methods. Therefore, relying solely on chemical analysis may miss the important components of the drug that contribute to its biological activity [20, 21]. Chemical analysis may not detect contaminants or adulterants, particularly if they are structurally similar to the active ingredients of the drug. Therefore, relying solely on chemical analysis may not provide an accurate assessment of drug safety and quality.

To overcome these limitations, biological QC can be performed in addition to chemical QC. Biological QC refers to the assessment of the biological activity of a drug or herbal medicine using in vitro or in vivo methods [22]. It can provide a more accurate assessment of the biological activity of a drug and can help detect potential contaminants or adulterants that may not be detected by chemical analysis. Therefore, the mechanism-based biological QC is an important complement to chemical QC for ensuring the safety and efficacy of drugs and herbal medicines.

Dangkwisoo-san (DS) is a herbal medicine used to treat various pathological conditions, such as qi movement stagnation and static blood primarily caused by trauma, such as contusions or falls [23]. This prescription was first described in Introduction to Medicine by Li Chan, a Confucian scholar of the Ming Dynasty of China. According to the composition of the prescription, Angelicae Gigantis Radix Palva had the highest dose among the nine herbs, followed by Paeoniae Radix, Linderae Radix, Cyperi Rhizoma, and Sappan Lignum in equal amounts, and Carthami Flos, Persicae Semen, Cinnamomi Cortex, and Glycyrrhizae Radix et Rhizoma. Clinically, these herbs are boiled with water and medicated wine at a ratio of 1:1 and provided in the form of decoction preparations; however, they are also used in various formulations, such as powder preparations [24]. DS exerts various pharmacological effects on the cardiovascular system [25], inflammatory diseases [26, 27] and cancers [28, 29]. Regarding the potential benefits and safety of natural origin medicines, including DS, in human and animal diseases, bioactive components have been verified, followed by probable modes of action and adverse reactions. To eliminate the undesired effects of the extracts, chemical modifications are employed to optimize and expand their scope of therapeutic use. In addition, dereplication methods are applied to serve proper biological functions and exclude the known active constituents of complex natural products [30].

Because obtaining a renewable supply of active compounds from biological sources can be challenging, we need a general and complete knowledge of DS component properties. This scoping review aimed to confirm how the effects of DS were evaluated in various diseases in addition to extending the knowledge of DS component properties. This was achieved through the process of identifying and comprehensively summarizing and analyzing in vitro, in vivo, and ex vivo experiments and clinical studies on DS through a systematic methodology. Our research can be expected to suggest biological assays to assess biomarkers that support chemical QC.

Search strategy and selection criteria

This review introduces the current state of cell-, animal-based, and clinical studies on target diseases and efficacy of DS by collecting, identifying, and classifying the literature with a pilot search-based search strategy. Considering that DS prescription is known by various scientific names, the following search terms were set through related reviews and databases: “dangguisoo,” “dangguisu,” “dangguixu,” “danggwisoo,” “danggwisu,” “dangkisoo,” “dangkisu,” “dangkwisoo,” “dangkwisu,” “当帰須散,” and “当归须散.” A literature search was conducted on six databases: EMBASE, CNKI, ScienceON, KISS, RISS, and OASIS up to August 8, 2022, and there were no restrictions on language during the search process. These classification criteria were included in the review of clinical and basic laboratory studies evaluating effects based on the original regimen of DS only without other components. Studies were excluded for the following reasons. (1) research unrelated to traditional East Asian medicine; (2) studies not related to DS; (3) studies in which it is difficult to confirm the effect of DS alone (e.g., case reports in which DS and other interventions were applied simultaneously); (4) studies that included only chemical analysis without data on the effectiveness of DS; (5) non-original research (e.g. conference abstracts, editorials, letters). As for the search strategy, researchers independently reviewed and evaluated the title, abstract, and full text qualifications of the thesis, extracted data on certain items from the literature, and other researchers independently verified the results. Disagreements that occurred in the process of literature selection and data extraction were resolved through discussions among researchers. Afterward, the extracted data was summarized and presented in a table with a narrative description. Among a total of 298 articles, 35 articles (6 clinical studies, 2 ex vivo studies, 15 in vivo studies, 10 in vitro studies, and 2 in vivo and in vitro studies) were selected and included in the analysis using mentioned search strategy (Fig. 1).

Fig. 1
figure 1

Flow chart for study selection. The flow diagram indicates the retrieval process of the study

Clinical studies on DS

Six clinical studies [27, 31,32,33,34,35] on DS were analyzed, including three randomized controlled trials (RCTs) [27, 31, 35] and three case reports [32,33,34] (Table 1). DS was used as a treatment for traumatic diseases, including pulmonary contusion, traumatic pneumothorax, bruising, and ankle sprain, in all studies except for one case series of peptic ulcers. In the RCT for pulmonary contusion [31], the use of DS in the form of a decoction in addition to usual care ameliorated inflammation-related indicators and reduced hospitalization times significantly compared to the control group. In two RCTs on ankle sprains, DS powder capsules were [27, 35] more effective in alleviating pain, functional abnormalities, and edema and improving quality of life than the placebo control group [27].

Table 1 Summary of clinical trials on DS

The preparation form of DS used in the six clinical studies was primarily a decoction of medicinal herbs (n = 3) or powder encapsulation (n = 2), and one study did not describe the form of the prepared medicine [34]. In the case of decoction, only one case mentioned preparing the herbal decoction using alcohol and water together as in the original prescription [33]. In another study using decoction [31, 32] and two studies using commercialized powdered capsules, water extraction was performed [27, 35].

The composition of the DS prescriptions used in the six clinical studies was slightly different. The content of Angelicae Gigantis Radix Palva was the highest, consistent with the composition of the original prescription. However, the proportions of the remaining herbs, including the four herbs listed as the second-highest proportions in the original prescription (Paeoniae Radix, Linderae Radix, Sappan Lignum, and Cyperi Rhizoma), were heterogeneous in the literature. When the content of Angelicae Gigantis Radix Palva in the prescription was set as 100%, the proportion of other herbs in each study and the original prescription were calculated and are presented in Table 2.

Table 2 Ratio of each herb to Angelicae Gigantis Radix Palva in the Dangkwisoo-san prescription used in the analyzed clinical studies

In vivo and ex vivo studies on DS

Between 1982 and 2021, 18 studies on the efficacy of DS have been published. The efficacy of DS has been studied for various target diseases, such as bruises, hematomas, toxin coagulation, blood pressure control, fractures, wound healing and inflammation, brain damage, and liver toxicity. The details of in vivo and ex vivo studies on DS are summarized in Tables 3 and 4.

Table 3 Summary of in vivo studies on DS
Table 4 Summary of ex vivo studies on DS

Two articles on bruises were published in 2002, and the same author evaluated the efficacy of DS by measuring the levels of lactate dehydrogenase (LDH) in the blood and a water maze test for studying the recovery of endurance exercise capacity and dehydration enzymes [36]. In another study [37], the researchers evaluated the effect of DS on blood enzyme activity in bruised rats by measuring blood levels of glutamic pyruvic transaminase (GPT), glutamic oxaloacetic transaminase (GOT), leucine aminopeptidase (LAP), and alkaline phosphatase (ALP).

Six articles were published on the efficacy of DS for hemostasis, hematomas, toxin-induced coagulation, and blood pressure. Lee et al. [38] measured the circumference and temperature of the thigh and the levels of platelets, fibrinogen, fibrinogen degradation production (FDP), and prothrombin time, which are factors related to blood coagulation, to evaluate efficacy. In another study on hemostasis [39], the researchers measured the blood levels of white blood cells (WBC), red blood cells (RBC), ALP, LDH, LAP, GOP, and GPT to confirm efficacy. An efficacy study [40] on treating subcutaneous hematomas was histologically analyzed by H&E staining, and it confirmed the efficacy of DS. Efficacy studies on endotoxin-induced coagulation [41, 42] focused on the effect of DS after coagulation induction by measuring the FDP, platelet count, and prothrombin time. In the ex vivo study [43] for blood pressure regulation, New Zealand white rabbit veins were collected, and the efficacy of DS was evaluated by comparing the degree of vascular contraction with that of vessels cultured in atropine, pheniramine, and propranolol.

Four studies evaluated the efficacy of DS in treating fractures. The researchers [44, 45] evaluated the efficacy of DS in SD rats by tissue staining and determination of blood levels of total-alanine transaminase (ALT), ALP, aspartate transaminase (AST), and bone formation-related factors osteocalcin and transforming growth factor-beta 1 (TGF-β1) in addition to recent studies [46, 47]. The same research team used C57BL/6 J mice to evaluate the efficacy of DS by measuring blood concentrations of bone morphogenetic protein 2 (BMP2), cyclooxygenase 2 (COX2), collagen type II alpha 1 chain (Col2a1), SRY-box transcription factor 9 (Sox9), runt-related transcription factor 2 (Runx2), and osterix and histological analysis.

Three studies confirming the efficacy of DS for wound healing and inflammation-related diseases have been published. First, wound healing efficacy was evaluated based on the levels of inflammatory factors TNF-α, IL-1β, and IL-6 and wound repair-related factors MMP-1, MMP-2, and MMP-9 [48]. In another study on acute lung inflammation [26], the researchers focused on body weight change, bronchoscopy, and bronchoalveolar lavage (BAL) levels. In a study on analgesia, anti-inflammation, and muscle relaxation [49], the efficacy of DS was confirmed in response to stimuli, such as mouse paw licking time, escape time, foot swelling, body temperature, and muscle relaxation.

Three studies on the efficacy of DS for brain injury and liver toxicity have been published. In a study on traumatic brain injury [50], the efficacy of DS was confirmed by evaluating exercise capacity, including the beam walking test, grip strength test, and NOR test, and measuring the degree of brain damage. In ischemic brain injury research [51], the efficacy of DS was evaluated by measuring blood pressure; resting cerebral blood flow (CBF); damage measurement through tissue staining; levels of inflammatory factors, such as eNOS, AKT, iNOS, nNOS; and exercise capacity. In addition, the efficacy of DS for the contraction and relaxation of vessels was evaluated through an ex vivo experiment using blood vessel collection. In this vascular relaxation study [52], the effect of DS was confirmed using a New Zealand white rabbit aorta. Studies on liver toxicity [53] confirmed the efficacy of DS against carbon tetrachloride-induced hepatotoxicity, and its efficacy was evaluated based on the levels of ALP, LDH, GOT, GPT, LAP, and α-hydroxybutyric dehydrogenase in the blood.

The sample used for evaluating the efficacy of DS was hot water extraction in this study, except for two studies [48, 49]. The difference between decoction and extract is that decoction refers to the state of hot water extraction with herbal medicines, and extracts can be weighed by processing the state after hot water extraction. The oral volume was measured when administering decoction, whereas, in the case of extract, the dry powder was weighed to determine the dose. After preparation, the extract was administered orally.

In vitro studies on DS

Twelve articles investigated the effect of DS using in vitro model. The details of in vitro studies on DS are summarized in Table 5. DS reportedly possesses anti-inflammatory, antioxidant, anticancer, and antimicrobial activities; prevents ischemic injury; and helps in treating metabolic syndromes, such as cardiovascular disease. Studies on DS have primarily focused on anti-inflammatory activity against LPS, an inflammatory inducer in RAW264.7, and DS was primarily extracted with water (92.3%). Generally, DS prescription was boiled with 1L of distilled water by an herb extractor for 2 h, yielding final 200 ml of DS extract. The supernatant was then evaporated and lyophilized under reduced pressure at low temperature. For MeOH extraction, DS was mixed with 3L of methanol at ambient temperature for 24 h. After centrifugation in a sterile condition, the supernatant was collected and lyophilized through evaporation under reduced pressure at − 80 ℃. Another method is using 80% EtOH where DS was boiled in 1L of 80% EtOH for 3 h and then concentrated by rotary vacuum evaporator.

Table 5 Summary of in vitro study designs on DS

DS exerts anti-inflammatory effects by regulating inflammatory mediators and cytokines. Lyu et al. [26] demonstrated that DS, which was extracted with distilled water, activated the anti-inflammatory factor Nrf2 (nuclear factor erythroid 2-related factor 2) and the expression of Nrf2-related genes, such as glutamate–cysteine ligase catalytic subunit (GCLC), heme oxygenase (HO-1), and NAD(P)H quinone oxidoreductase 1 (NQO-1) in RAW264.7cells. In addition, pre-treatment of NF-κB reporter cells derived from RAW264.7 with DS, followed by LPS treatment, regulated NF-κB, which was confirmed using HEK293 cells transfected with inhibitor of kappa B (IκB) kinase- β (Iκκ-β) and NF-κB. DS treatment suppressed the levels of pro-inflammatory cytokines, including TNF, MCP-1, and IL-6. In addition, analysis of reactive oxygen species (ROS) induction by DS using flow cytometry confirmed that DS did not produce ROS.

Two studies on the anti-inflammatory effects of DS used extraction solvents other than water and revealed significant effects at low DS concentrations. Bak et al. [48] investigated the anti-inflammatory effects of DS extracted using 80% ethanol. DS reduced the production of NO, IL-1β, IL-6, and TNF-α in the LPS-induced RAW264.7 cell line in a dose-dependent manner (1, 10, and 100 μg/ml). Ryu et al. [54] compared the anti-inflammatory effects of methanol (mDS) and water extracts (wDS) of DS. mDS activated Nrf-2 and its dependent protein HO-1 and expressed Nrf-2-related genes, GCLC, HO-1, and NQO1, in RAW264.7cells. The results confirmed that 50 μg/ml mDS was more effective in increasing Nrf2 than wDS, with sulforaphane (SFN; 5 μM), an Nrf-2 activator, as a control. However, mDS was less effective in decreasing NF-κB than wDS in RAW264.7 stably transfected with NF-κB. NF-κB activation by mDS was weakened 30 min after LPS treatment, and the degradation of IκB-α was not suppressed 15 min after LPS induction in murine macrophages. Pro-inflammatory cytokines TNF-α and IL-1β were reduced by mDS. These findings indicated that DS attenuates inflammation.

In a study of the effect of 9 components of DS on Nrf2 and NF-κB, the activation of Nrf2 at 50 μg/ml of DS in LPS-induced RAW264.7cells was confirmed [22]; however, each constituent of DS showed cytotoxicity at 50 μg/ml. Therefore, DS was used at 25 μg/ml, and most components induced the activation of Nrf2 in the Nrf2 reporter cell line derived from RAW264.7, particularly Carthamus tinctorius L. (CT). In addition, nine components of DS regulated NF-κB in the NF-κB luciferase reporter cell line using the same method as for Nrf2, particularly Carthamus tinctorius L. (CT) and Cyperus rotundus L. (CR). These findings suggested that two constituent herbal medicines, CT and CR, in DS serve as the main anti-inflammatory agents.

Jeon et al. [55] examined the anti-inflammatory activity of DS-dry extract (DS-DE) with DS-mix extract powder (DS-MEP) and used dexamethasone (20 µM) as a positive control. DS-DE and DS-MEP had a greater inhibitory effect than dexamethasone (20 μM) on NO production. DS-DE and DS-MEP suppressed the protein and mRNA expression of iNOS, PGE2, and COX-2 in a dose-dependent manner (50, 100, and 200 μg/ml) in LPS-stimulated RAW264.7cells. The secretion of TNF-α, IL-1β, and IL-6 was significantly suppressed by DS-DE and DS-MEP, coinciding with transcriptional levels.

In a study conducted by Jo et al. [56], 100 µg/ml DS with hot aqueous extraction inhibited the production of NO and PGE and suppressed the levels of IL-1β, IL-6, and TNF-α in LPS-activated RAW264.7. In addition, 100 µg/ml DS had more than 50% free-radical scavenging ability, as measured using the DPPH (2,2-diphenyl-1-picrylhydrazy) radical scavenging method. Further, the effect of DS was evaluated for bone repair in osteoblast-like MG63cell line and RAW264.7 macrophages [46]. DS (500 μg/ml)-treated MG63 cells demonstrated increased gene expression of osteocalcin and Runx2, with calcitriol as a positive control. DS increased the secretion of TNF-α in LPS-induced RAW264.7 cells in a dose-dependent manner compared to cyclosporine A (as a positive control), and 250 and 500 g/ml of DS yielded statistically significant results.

Hwang et al. [28] reported the anticancer efficacy of DS. Levels of caspase-3, caspase 9, and subG1 and mitochondrial depolarization increased in AGS gastric adenocarcinoma cells treated with varying concentrations (50, 100, 200, 300, and 400 μg/ml) of DS. Moreover, DS decreased transient receptor potential melastatin (TRPM7) in AGS and TRPM7-transfected HEK293 cells, determined using the patch-clamp technique. These results highlighted that the MAPK (p38 and JNK) signaling pathway is associated with DS through a JNK II inhibitor or SB203580 (p38 MAPK inhibitor).

Another study [57] on TRPM7 focused on the action of DS in HEK293 cells that stably overexpressed TRPM7 using the whole-cell patch clamp technique. DS suppressed the overexpression of TRPM7 current in a dose-dependent manner (100, 300, and 500 μg/ml), thus alleviating metabolic syndromes, such as cardiovascular disease.

Lee et al. revealed the antimicrobial activity of DS [58] Ten pathogenic microorganisms (Bacillus cereus, Staphylococcus aureus, Listeria monocytogens, Vibrio parahaemolytixus, Escherichia coli DH5α, E. coli O157, Salmonella nteritidis, Yersinia enterocolitica, Shigella flexneri, and Helicobacter pylori) were screened for the antimicrobial effects of 51 herbal formulae used in traditional Korean medical prescriptions. Of these, DS showed antibacterial effects against B. cereus and antimicrobial activity against L. monocytogenes (5, 10, 15, and 20 mg/disc) and V. parahaemolyticus (15 and 20 mg/disc).

Kim et al. [25] suggested that human brain microvascular endothelial cells (HBMEC) treated with 30 μg/ml DS exhibited stimulated NO production, with acetylcholine as a positive control. Therefore, DS in ischemic injury regulates vascular function through an eNOS-dependent mechanism.

DS causes pacemaker depolarization of the interstitial cells of Cajal (ICCs) through the M3 receptor. In addition, G-protein, external Na+, and non-selective cation channels are mediated by DS-activated pacemaker depolarization [59]. Moreover, external or internal Ca2+ plays an important role in regulating pacemaker potentials in ICCs, and DS elevates Ca2+ in ICCs.

In summary, DS appears to have effects on inflammatory factor, bone regeneration, ischemic injury repair and metabolic disorders. Since a small number of in vitro experimental reports on the metabolic disorders, bone regeneration, and research on the recovery from ischemic injury have been published, there are advance studies should be conducted to generate reproducibility of findings. However, we figured out mouse macrophage RAW 246.7 cell model was used for most of the examinations on effects of DS on inflammatory response, while LPS was used as a stimulant in all of experiments. In addition, the concentration range from 1 to 200 μg/mL was applied for evaluating the DS efficacy. It was confirmed that the effective dose range of DS was homogenous. Although there is only two publishes, it was apparently confirmed that the difference in therapeutic efficacy depending on DS extraction methods. Using an organic solvent extracting method achieved better curing effect even at a lower concentration than that of the hot water extracts.

Although chemical quality assessments are being performed to ensure consistent efficacy of DS, the identity and quantity of the compounds contained within a given natural product is a frequent complication of studies. Even when the composition is relatively well understood, the lack of simple and effective analytical methods hampers the ability to extrapolate results beyond quality studies. We conducted a literature review to identify biological assays that support chemical QC.

Implications that could be obtained from the current clinical research of DS are as follows. First, although the use of DS prescriptions in clinical practice is extensive, relatively few clinical studies have been conducted. In particular, based on the insights obtained during the search phase for clinical studies, DS has been used as a representative prescription for trauma in Korea [60] also recently published in the clinical practice guidelines of Korean medicine for traffic injuries and acute ankle sprains (recommendation level/evidence level: C/Very Low, respectively) [61, 62]. DS has been used to cure diverse symptoms in China; however, it is difficult to consider it a representative prescription, and it has rarely been investigated in Japan. The tendency toward DS utilization in clinical practice was not sufficiently reflected in the results of this scoping review because of the small number of clinical studies. Second, it is necessary to discuss standardization of the herb composition ratio in DS prescriptions. We excluded the modified formula to control heterogeneity among the DS prescriptions. The herbs constituting DS prescriptions were the same; however, there were differences in the proportion of herbs used in each study. The DS prescription used in Korean studies was the same as the original prescription; however, in the two Chinese studies on thoracic injuries, the content ratio of Linderae Radix, Cyperi Rhizoma, Paeoniae Radix, Carthami Flos, and Persicae Semen to Angelicae Gigantis Radix Palva was higher than that of the original prescription. Third, there were also rare cases where the “decoction form by boiling herbs with water and medicinal wine” mentioned in the original prescription was used identically. We need to understand the reasons for the extraction of the original recipe through additional research. In addition, alcohol extraction at a certain ratio may be more efficient than water extraction, which is primarily used in clinical studies [63].

Based on in vivo and ex vivo studies, a preclinical study on DS reported efficacy verification and methods for various diseases, such as blood coagulation, hepatotoxicity, fractures, brain disease, inflammation-related disease, and bruises. These results could be valid for evaluating efficacy by a simple method that can be used in clinical applications. As the approval of the FDA for herbal medicine for post-marketing is rather laborious, effective clinical outcome demands attention. In fact, in vivo and in vitro preclinical assessments are the initial stages of the evaluation of later steps. However, preclinical research has certain difficulties, such as ethical problems, excessive experimental period, requirement of high-quality human resources, and high cost. These problems make it difficult to maintain efficacy consistency in the same experiment. In addition, it hinders the use of many in vitro methods and their clinical and preclinical relevance. Therefore, the evaluation and consistency of efficacy of various herbal medicines and prescriptions, including DS, should be subjected to evaluation methods by carefully selecting clinical and preclinical related assays. In this study, we aimed to develop a scoping review of DS as an alternative therapy, particularly for in vitro research.

Inflammation is a representative expression of the secondary defense mechanism of the body in response to foreign stimuli. Many reports on aqueous DS extract treatments have shown effective anti-inflammatory effects. Using an LPS-stimulated Raw264.7 cell model, water-extracted DS reduced the expression level of certain proinflammatory cytokines, such as TNF-α, IL-1β, and IL-6, as well as the NF-kB signaling pathway [26, 54, 56, 64]. Carthamus tinctorius L. (CT) and Cyperus rotundus L. (CR) are two active components of DS that have shown anti-inflammatory effects at lower doses than dexamethasone [55]. Moreover, according to Ryu [54] and Bak et al. [48], MeOH and EtOH extraction of DS notably suppress inflammation of stimulated Raw 264.7 cells at concentrations lower than 50 μg/ml. Accounting for 92.3% of the DS extracts, water-extracted DS was effectively involved in several metabolic mechanisms, including cancer, bone regeneration, intestinal phasic contraction, and cell membrane potential change. For instance, anticancer activity was recorded with the induction of certain apoptotic markers (caspase 9 and caspase 3) by Hwang et al. [28] while inhibiting TRPM7 channel activity. Moreover, Lee et al. [58] demonstrated the antimicrobial activity of aqueous DS extract at less than 20 mg/disc. Sung et al. [59] reported DS-induced changes in the membrane Ca2+ potential of ICCs, resulting in pacemaker depolarization.

We concluded that DS in vitro was the most effective anti-inflammatory agent at concentrations of 50 and 100 μg/ml, and in vitro reduction of biochemical markers, such as TNF, IL-1β, IL-6, NO, Nrf2, and NF-ĸB, effectively demonstrated the potential role of DS in the inhibition of inflammation (Table 4). In addition, DS extracts are extracted by hot water as traditional method. However, using 80% EtOH or MeOH as extraction solvents, the treatment of DS showed more effective results at lower doses in vitro. The efficacy might be associated with the higher concentration of active component because of extraction with organic solvents. DS is composed of nine herbal medicines: Paeoniae Radix, Angelicae Gigantis Radix, Sappan Lignum, Linderae Radix, Persicae Semen, Cyperi Rhizoma, Carthami Flos, Glycyrrhizae Radix et Rhizoma, and Cinnamomi Cortex. Rhizoma. However, the identification of active compounds in DS will help understand their molecular mechanisms and targets. However, multiple herbal medicines for multi-target treatment may produce synergistic effects. Although DS is known to be commonly used to relieve headaches, contusions, and external and cerebral ischemic injuries, in vitro tests for DS are limited. High DS concentration has effects similar to those of other natural compounds. Experimental studies can provide insights into maximizing therapeutic efficacy by inhibiting inflammatory mediators and cytokines.

As introduced in mechanism based QC study showing in vivo activities of irinotecan [65], suitable bioassays to identify the biomarkers related to mechanism of action of the clinical efficacy would be a good example of biological QC. For these biomarkers, standard operating procedures (SOPs) should be prepared for the generation of reproducible and accurate results. Therefore, the mechanism-based QC using biomarkers would be useful for the prediction and confirmation of consistent efficacy of DS. Quality control of herbal preparations including DS should be different depending on the pharmacological usages. As chemical evaluation methods are limited in their ability to distinguish irrelevant chemicals, QCs evaluating specific uses and efficacy should be developed concise and appropriate to biological assays based on in vitro mechanisms of action.

The above discussion suggests that future quality evaluation studies of herbal medicines (herbal medicine prescriptions) should be aimed at selecting an optimal in vitro efficacy evaluation method capable of evaluating the mechanism of action for the assessment of consistent clinical effects. This method should evaluate the mechanism of action of the clinical effect, use appropriate positive controls and statistical methods, and prepare reproducible SOPs.

Availability of data and materials

All data generated or analysed during this study are included in this published article [and its Additional files].

Abbreviations

DS:

Dangkwisoo-san

EtOH:

Ethanol

MeOH:

Methanol

RCT:

Randomized controlled trial

eNOS:

Endothelial nitric oxide synthase

iNOS:

Inducible nitric oxide synthase

nNOS:

Neuronal nitric oxide synthase

BMP2:

Bone morphogenetic protein 2

COX2:

Cyclooxygenase-2

Col2a1:

Collagen type II alpha 1 chain

Sox9:

SRY-box transcription factor 9

Runx2:

Runt-related transcription factor 2

GPT:

Glutamic pyruvic transaminase

ALT:

Alanine aminotransferase

GOT:

Glutamic oxaloacetic transaminase

AST:

Aspartate aminotransferase

LAP:

Leukocyte alkaline phosphatase

ALP:

Alkaline phosphatase

LDH:

Lactate dehydrogenase

FDP:

Fibrinogen degradation production

MMPs:

Matrix metalloproteinases

4-DAMP:

4-Diphenylacetoxy-n-methyl-piperidine methiodide

GDP-β-S:

A non-hydrolysable guanosine 5′-diphosphate analogue

ILs:

Interleukin

TNF:

Tumor necrosis factor

Runx:

Runt-related transcription factor

Nrf-2:

Nuclear factor erythroid-2-related factor 2

GCLC:

Glutamate–cysteine ligase catalytic subunit

HO-1:

Heme oxygenase-1

NQO-1:

Quinone oxidoreductase 1

ROS:

Reactive oxygen species

MCP-1:

Monocyte chemoattractant protein-1

TRPM7:

Transient receptor potential cation channel subfamily M member 7

References

  1. Sorokina M, Steinbeck C (2020) Review on natural products databases: where to find data in 2020. J Cheminf 12:20

    CAS  Google Scholar 

  2. Ekor M (2014) The growing use of herbal medicines: issues relating to adverse reactions and challenges in monitoring safety. Front Pharmacol 4:177

    PubMed  PubMed Central  Google Scholar 

  3. Ventegodt S, Morad M, Hyam E, Merrick J (2004) Clinical holistic medicine: use and limitations of the biomedical paradigm. Sci World J 4:295–306

    Google Scholar 

  4. Karimi A, Majlesi M, Rafieian-Kopaei M (2015) Herbal versus synthetic drugs; beliefs and facts. J Nephropharmacol 4:27

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Mukherjee PK (2002) Quality control of herbal drugs: an approach to evaluation of botanicals. Business Horizon’s Publishers, New Delhi, pp 380–421

    Google Scholar 

  6. Ahmad I, Aqil F, Owais M (2006) Modern phytomedicine: turning medicinal plants into drugs. John Wiley & Sons, India

    Google Scholar 

  7. Bodeker G, Ong CK, Grundy G, Burford G, Shein K (2005) WHO global atlas of traditional, complementary and alternative medicine. World Health Organization, Japan

    Google Scholar 

  8. Ahn K (2017) The worldwide trend of using botanical drugs and strategies for developing global drugs. BMB Rep 50:111

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Ministry of Food and Drug Safety. (2016) Botanical drug development guidance for industry. Korea.

  10. Qiu J (2007) ’Back to the future’for Chinese herbal medicines. Nat Rev Drug Dis 6:506

    CAS  Google Scholar 

  11. Wang L, Zhou GB, Liu P, Song JH, Liang Y, Yan XJ, Xu F, Wang BS, Mao JH, Shen ZX (2008) Dissection of mechanisms of Chinese medicinal formula Realgar-Indigo naturalis as an effective treatment for promyelocytic leukemia. Proc Natl Acad Sci 105:4826–4831

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Jan JT, Cheng TJR, Juang YP, Ma HH, Wu YT, Yang WB, Cheng CW, Chen X, Chou TH, Shie JJ, Cheng WC, Chein RJ, Mao SS, Liang PH, Ma C, Hunag SC, Wong CH (2021) Identification of existing pharmaceuticals and herbal medicines as inhibitors of SARS-CoV-2 infection. Proc Natl Acad Sci 118:e2021579118

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Ohashi H, Watashi K, Saso W, Shionoya K, Iwanami S, Hirokawa T, Shirai T, Kanaya S, Ito Y, Kim KS, Nomura T, Suzuki T, Nishioka K, Ando S, Ejima K, Koizumi Y, Tanaka T, Aoki S, Kuramochi K, Suzuki T, Hashiguchi T, Maenaka K, Matano T, Muramatsu M, Saijo M, Aihara K, Iwami S, Takeda M, Mckeating J, Wakita T (2021) Potential anti-COVID-19 agents, cepharanthine and nelfinavir, and their usage for combination treatment. Iscience 24:102367

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Kumar, A., Singh, A., and Aggarwal, A. (2017) Therapeutic potentials of herbal drugs for Alzheimer’s disease—An overview.

  15. Wink M (2015) Modes of action of herbal medicines and plant secondary metabolites. Medicines 2:251–286

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Newton AC, Bootman MD, Scott JD (2016) Second messengers. Cold Spring Harb Perspect Biol 8:a005926

    PubMed  PubMed Central  Google Scholar 

  17. Heldin C-H, Lu B, Evans R, Gutkind JS (2016) Signals and receptors. Cold Spring Harb Perspect Biol 8:a005900

    PubMed  PubMed Central  Google Scholar 

  18. Yan K, Gao LN, Cui YL, Zhang Y, Zhou X (2016) The cyclic AMP signaling pathway: exploring targets for successful drug discovery. Mol Med Rep 13:3715–3723

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Thul R, Coombes S, Roderick HL, Bootman MD (2012) Subcellular calcium dynamics in a whole-cell model of an atrial myocyte. Proc Natl Acad Sci 109:2150–2155

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Guantai E, Chibale K (2011) How can natural products serve as a viable source of lead compounds for the development of new/novel anti-malarials? Malar J 10:1–8

    Google Scholar 

  21. Hur M, Campbell AA, Almeida-de-Macedo M, Li L, Ransom N, Jose A, Crispin M, Nikolau BJ, Wurtele ES (2013) A global approach to analysis and interpretation of metabolic data for plant natural product discovery. Nat Prod Rep 30:565–583

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Kumar K, Waldmann H (2009) Synthesis of natural product inspired compound collections. Angew Chem Int Ed 48:3224–3242

    CAS  Google Scholar 

  23. Oasis. (2016) 당귀수산 , 當歸鬚散, Dangguisu-san. https://oasis.kiom.re.kr/oasis/pres/prdetailView.jsp?idx=28&selectname=%EB%28B%B29%EA%B27%80%EC%88%98%EC%82%B20&srch_menu_nix=22Z36BIeh#view03

  24. Ministry of Food and Drug Safety. (2005) Standardization of herbal medicine preparation. Korea.

  25. Kim JH, Park SH, Kim YW, Ha JM, Bae SS, Lee GS, Cho SI, Choi BT, Shin HK (2011) The traditional herbal medicine, Dangkwisoo-San, prevents cerebral ischemic injury through nitric oxide-dependent mechanisms. Evid Based Complement Alt Med 2011:718302

    Google Scholar 

  26. Lyu JH, Kim KH, Kim HW, Cho SI, Ha KT, Choi JY, Han CW, Jeong HS, Lee HK, Ahn KS, Oh SR, Sadikot RT, Christman JW, Joo M (2012) Dangkwisoo-san, an herbal medicinal formula, ameliorates acute lung inflammation via activation of Nrf2 and suppression of NF-κB. J Ethnopharmacol 140:107–116

    PubMed  PubMed Central  Google Scholar 

  27. Kim JH, Lee CK, Lee EY, Cho MR, Lee YS, Lee JS (2021) Effects of Dangguixu-san in patients with acute lateral ankle sprain: a randomized controlled trial. Trials 22:1–12

    Google Scholar 

  28. Hwang MW, Kim BJ (2014) Apoptotic effects and involvement of TRPM7 channels of the traditional herbal medicine, Dangkwisoo-san in gastric cancer cells. Int J Pharmacol 10:398–405

    CAS  Google Scholar 

  29. Cho, J. H., Park, I. J., Jung, Y. H., Choi, G. H., Yoon, W. J., Park, S. H., Sin, S. H., Yun, J, H., and Baik, S. O. (2016) Composition for preventing and treating inflammatory disease, containing oil of litsea japonica as active ingredient, and method for preparing same. U.S. Patent Application

  30. Atanasov AG, Zotchev SB, Dirsch VM, Supuran CT (2021) Natural products in drug discovery: advances and opportunities. Nat Rev Drug Discovery 20:200–216

    CAS  PubMed  Google Scholar 

  31. Wei CH, Fang SY, Liu JQ, Deng P, Lu JL, Tan YM (2014) Analysis of the efficacy of Dangguixusan in the treatment of acute pulmonary contusion. Chinese Journal of Trauma 30:423–425

    Google Scholar 

  32. Chen M (1993) Dangguixusan treatment of traumatic hemopneumothorax in 16 cases Summary. J Tradit Chin Orthop Traumatol 5:32

    Google Scholar 

  33. Shao, Y. F. (2000) Dangguixusan for peptic ulcer 60 cases. Jilin Journal of Chinese Medicine

  34. (1987) Use Dangguixusan to treat bruise after a bump. Anthology of Medicine, 206–207. Anthology of Medicine, 206–207

  35. Hwang, S. J. (2021) Economic evaluation of acupuncture-Taping and acupuncture-Danggui-Susan products combined for single acupuncture. Dissertation, University of Kyung Hee.

  36. Yeo NH, Lee HY (2002) The effect of dang kwisoosan on the restoration of endurable exercise ability. Excise Sci 11:17–24

    Google Scholar 

  37. Yeo NH, Lee HY (2002) The effect of Dangkwisoo-San on blood enzyme activity in brusied rats. Korean J Phys Educ 41:333–340

    Google Scholar 

  38. Lee GH, Ahn KS, Choi SH (1999) Study on the effects of dangguixusan on experimental blood stasis model induced by compression. J KyungHee Oriental Med Coll 22:22–38

    Google Scholar 

  39. Lee, H. Y. (1990) Experimental studies on the effect of Dangkwisoo-san on the elimination of the hematostasis and restoration of endurable exercise ability. Dissertation, University of Dong-A

  40. Kim KH (1985) Effects of Dangkwisoosan on subcutaneous hematoma. J KyungHee Oriental Med Coll 8:23–31

    Google Scholar 

  41. Nam SS, Ahn BC, Park DS (1993) Effect of tangguisusan aqua-acupuncture and acupuncture on the intravascular coagulation induced by endotoxin in rats. J Korean Acupunct Moxib Soc 10:133–149

    Google Scholar 

  42. Kim TS, Ahn KS (1988) Effect of Dangkwisoosan and Dodamtang on the intravascular coagulation induced by endotoxin in rat. J Korean Orient Med Pathol 3:91–98

    Google Scholar 

  43. Lew, J. H. (1983) Depressor response of Dangkwisoo-san water extract on the arterial blood pressure in the rabbits. Dissertation, University of Wonkwang.

  44. Jung, I. M. (2006) Effect of Dangkwisoo-san(danffuixu-san) and native copper on TGF-β1 expression in fractured rats. Dissertation, University of Dongshin.

  45. Ahn HL, Shin MS, Kim SJ, Choi JB (2007) Effects of neutral Eohyeol (Yuxue) herbal acupuncture and Dangkisoo-san (Dangguixu-san) on fracture healing in the early stage in rats. Korea J Herbol 29:55–63

    Google Scholar 

  46. Jeon DH, Oh MS (2021) Healing effect of Danggwisu-san (Dangguixu-san) on femur fractured mice. J Korean Med Rehabilit 31:1–16

    Google Scholar 

  47. Jeon, D. H. (2006) Healing effect of danggwisu-san (Dangguixu-san) on femur fractured mice. Dissertation, University of Daejeon.

  48. Bak JW, Sim BY, Kim DH (2014) The effects of Danggwisusan on restoration ability in wound induced animal models. Korea J Herbol 29:55–63

    Google Scholar 

  49. Ko, W. S. (1991) Effects of Dangkwiwoosan on analgesic, anti-inflammatory, muscle relaxation and change of body temperature through experimental animals. Dissertation, University of Wonkwang

  50. Jung JY, Joo H, Bae J, Bang Y, Lee BJ, Cho JH, Park JW, Lee K, Bu Y (2018) Effects of Dangguisusan, a prescription of Korean medicine on controlled cortical impact-induced traumatic brain injury mouse model. J Ethnopharmacol 225:198–201

    PubMed  Google Scholar 

  51. Kim JH, Park SH, Kim YW, Ha JM, Bae SS, Lee GS, Cho SI, Choi BT, Shin HK (2011) The traditional herbal medicine, Dangkwisoo-San, prevents cerebral ischemic injury through nitric oxide-dependent mechanisms. Evid Based Complement Alternat Med 2011:718302

    PubMed  PubMed Central  Google Scholar 

  52. Ko H, Shin SM, Park SY (2019) Endothelium-dependent vasorelaxation effects of DangGuiSu-San, SamHwangSaSim-Tang extract on rabbit carotid artery. J Physiol Pathol Korean Med 33:198–206

    Google Scholar 

  53. Lim JG, Moon JJ (1982) Experimental studies on the effect of dangkwisoo-san on the CCl4-induced liver damage in albino-rats. J KyungHee Oriental Med Coll 5:191–208

    Google Scholar 

  54. Ryu JH, Kim H, Cho SI, Joo M (2017) Comparative study of the methanol and water extracts of dangguisoo-san in suppressing inflammatory reaction. J Physiol Pathol Korean Med 31:75–81

    Google Scholar 

  55. Jeon YH, Nam WH, Leem HH, Kim SJ, Yu BW, Son SM, Kim MJ, Choi HM, Kwon HS, Kim, J.- O. (2021) Anti-inflammatory effect and analysis of functional constituents of dangguisu-san by processing methods. Korean J Pharm 52:192–201

    CAS  Google Scholar 

  56. Jo NY (2018) Anti-inflammatory and anti-oxidative effects of danggwisusan on macrophages. J Acupunct Res 35:41–45

    Google Scholar 

  57. Kim BJ (2018) Effects of Dangkwisoo-San, ginger and curcumin on transient receptor potential melastatin 7 channels. J Korean Med Obesity Res 18:10–18

    Google Scholar 

  58. Lee N, Shin HK, Ha H, Choi SY (2019) Antimicrobial activities of 51 herbal formulae on pathogenic microorganisms. Herbal Formula Sci 27:257–267

    Google Scholar 

  59. Sung SK, Kim SJ, Ahn TS, Hong NR, Park HS, Kwon YK, Kim BJ (2015) Effects of Dangkwisoo-san, a traditional herbal medicine for treating pain and blood stagnation, on the pacemaker activities of cultured interstitial cells of Cajal. Mol Med Rep 12:6370–6376

    CAS  PubMed  Google Scholar 

  60. Kim HK, Kim JI, Kim YI (2021) Retrospective statistical analysis on patients admitted to a korean medicine hospital by traffic accident. J Korean Med 42:26–45

    Google Scholar 

  61. Kim, N. G. (2020) National Institute for Korean Medicine Development. Clinical practice guideline (CPG) of Korean medicine: ankle sprain. National Institute for Korean Medicine Develement, Korea

  62. Kim, N. G. (2021) National Institute for Korean Medicine Development. Clinical practice guideline (CPG) of Korean medicine: traffic injuries. National Institute for Korean Medicine Develement, Korea

  63. Lee DY, Lee HS, Jo JH, Yi YW, Kim SJ, Kang K, Kwon TW, Yang SG, Lee IH (2020) Comparison of ingredients and activities of Danggwisoo-san and Jakyakgamcho-tang by extraction method. J Korean Med Rehabilit 30:31–39

    Google Scholar 

  64. Kim KH, Jeong JH, Jeong HS, Ha KT, Joo MS (2012) Identification of the constituents for Nrf2 activation and NF-κB suppression in Dangguisoo-san. J Physiol Pathol Korean Med 26:344–350

    Google Scholar 

  65. Lam W, Ren Y, Guan F, Jiang Z, Cheng W, Xu CH, Liu SH, Cheng YC (2018) Mechanism based quality control (MBQC) of herbal products: a case study YIV-906 (PHY906). Front Pharmacol 9:1324

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This study was supported by a Grant (21173MFDS56-3) from the Ministry of Food and Drug Safety, 2023.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: S-YK and KSK; methodology, JHL, SJ, MJL and NKT; investigation, JHL and Y-JK; writing—original draft preparation, JHL and SJ; writing—review and editing, KSK; supervision, S-YK and KSK; project administration, KSK. All authors have read and agreed to the published version of the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Song-Yi Kim or Ki Sung Kang.

Ethics declarations

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lee, J.H., Jin, S., Lee, M.J. et al. Mechanism-based biomarkers for the quality control of Dangkwisoo-san: a scoping review. Appl Biol Chem 66, 52 (2023). https://doi.org/10.1186/s13765-023-00802-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13765-023-00802-5

Keywords