Skip to main content

Interpreting the efficacy enhancement mechanism of Chinese medicine processing from a biopharmaceutic perspective

Abstract

Chinese medicine processing (CMP) is a unique pharmaceutical technology that distinguishes it from natural medicines. Current research primarily focuses on changes in chemical components to understand the mechanisms behind efficacy enhancement in processing. However, this paper presents a novel perspective on the biopharmaceutics of CMP. It provides a comprehensive overview of the current research, emphasizing two crucial aspects: the role of ‘heat’ during processing and the utilization of processing adjuvants. The paper highlights the generation of easily absorbed components through the hydrolysis of glycosides by ‘heat’, as well as the facilitation of dissolution, absorption, and targeted distribution of active components through the utilization of processing adjuvants. From a biopharmaceutic perspective, this paper provides a lucid comprehension of the scientific foundation for augmenting the efficacy of CMP. Moreover, it proposes a three-dimensional research framework encompassing chemical reactions, phase transitions, and biopharmaceutical properties to further investigate the mechanisms involved in enhancing the efficacy of CMP.

Graphical Abstract

Introduction

Chinese medicinal materials cannot be used directly in clinical practice but must undergo processing according to Chinese medicine processing theory to become decoction pieces that are suitable for use in clinical prescriptions. Processing is a unique pharmaceutical technology in China and involves various methods, such as stir-frying, steaming, and boiling [1,2,3,4]. Furthermore, common adjuvants like vinegar, honey, wine, salt solution, ginger juice, bran, and rice are frequently employed during processing [5,6,7,8]. Over millennia of clinical practice, the scope of clinical application for Chinese medicine has expanded, producing various decoction pieces that undergo processing with adjuvants [9, 10]. The Chinese Pharmacopeia (2020 edition) lists decoction pieces and their related processing methods as specific items of Chinese medicines.

While the classical processing theory has proven reliable in long-term clinical practice, the scientific principles underlying it are not fully understood to date. Gaining a thorough understanding of the chemical reactions and changes in chemical components during processing is crucial for uncovering alterations in clinical efficacy before and after CMP [11, 12]. Currently, the explanation of the CMP mechanism mainly focuses on the changes in chemical components. Although recent research has shed light on some of these changes, the modifications in components often do not align with pharmacological effects. In other words, the changes in component content following processing cannot fully elucidate the scientific significance of the CMP. This suggests that the efficacy of Chinese medicine is not only influenced by changical components but is also closely related to biopharmaceutical processes within the body.

The low bioavailability of active components in Chinese medicine is a critical factor that affects its effectiveness. This is closely associated with the biopharmaceutical properties of these active components, such as poor solubility, low dissolution rate, low mucosal permeability, and limited absorption in the gastrointestinal tract [13,14,15]. The processes of absorption, distribution, metabolism, and excretion of active components in the gastrointestinal tract significantly influence their biological effects, with absorption playing a vital role in determining efficacy [16, 17]. The poor solubility and low permeability of most active components in Chinese medicine hinder their absorption in the gastrointestinal tract [18,19,20]. Furthermore, studies have demonstrated that the combination of adjuvants and active components can form unique morphologies, which can alter the solubility, permeability, and intestinal absorption of the active components, ultimately enhancing their therapeutic effectiveness. Therefore, to comprehensively understand the mechanisms of CMP, it is crucial to not only investigate the changes in chemical components during processing in vitro but also gain insights into the changes that occur in vivo. This involves elucidating the solubility, permeability, gastrointestinal absorption, and tissue distribution of active components.

Based on the overview of chemical reactions and changes in active components during processing, our research team has proposed an innovative approach to investigating the mechanism of CMP from a biopharmaceutical perspective. Our emphasis is on improving the biopharmaceutical properties of active components under the dual influence of heat and adjuvants. In addition to the hydrolysis reaction that occurs under the heating effect, resulting in the formation of easily absorbed components, we propose that the special formulation characteristics of adjuvants can establish a specific form with active components. This alteration in form can subsequently modify the solubility and permeability of the active components, thereby enhancing their in vivo biopharmaceutical behavior. This paper provides a clearer understanding of the scientific basis for enhancing the efficacy of CMP from a biopharmaceutic perspective, providing a novel perspective for revealing the scientific significance of CMP.

Formation of absorbable components during processing

For active components to be effective, they must be absorbed into the bloodstream (except for direct intestinal action and external application). Although Chinese medicine contains numerous components, it is commonly believed that only those components that are absorbed into the bloodstream can exert their effects [21, 22]. When evaluating the CMP from a biopharmaceutic perspective, it becomes apparent that the processing transforms these components, thereby enhancing their biopharmaceutical properties. This transformation enhances the absorption of these components into the bloodstream, resulting in an increased content of active components in the body. This could be a significant contributing factor to the observed improvement in efficacy during processing.

Hydrolysis of glycoside to produce easily absorbed components by “heat”

Hydrolysis is a frequently occurring chemical reaction during processing. Active components, such as flavonoid glycosides, saponins, iridoid glycosides, and polysaccharides, undergo hydrolysis reactions. These reactions can effectively reduce the number of glycosyl groups present in these components, consequently increasing their permeability in the body.

Flavonoid glycosides

Flavonoid glycosides are widely present in various types of Chinese medicine, such as Epimedh Folium (Yinyanghuo in Chinese), and Glycyrrhizae Radix et Rhizoma (Gancao in Chinese). Epimedh Folium, contains three-glycosyl flavonoid glycosides (epimedin A, epimedin B, epimedin C, 3‴-carbonyl-2″-β-l-quinovosyl icariin), two-glycosyl flavonoid glycosides (sagittatoside A, sagittatoside B, 2-O-Rhamnosylicariside II, Icariin) and one-glycosyl flavonoid glycosides (baohuoside I), which are considered the primary active components. Research has shown that one- or two-glycosyl flavonoid glycosides are more easily absorbed than those with three-glycosyl, as they not only ensure suitable solubility but also exhibit better permeability in the body [23, 24]. When Epimedh Folium is stir-fried with mutton tallow, hydrolysis reactions occur, converting epimedin A to sagittatoside A, epimedin B to sagittatoside B, epimedin C to 2″-O-Rhamnosylicariside II, and 3‴-carbonyl-2″-β-l-quinovosyl icariin to icariin (Fig. 1A–D) [25,26,27]. This indicates that the heat employed during processing can reduce the number of glycosyl groups on flavonoid glycosides in Epimedh Folium through hydrolysis reactions. Consequently, this process enhances their permeability, promotes absorption, increases their concentration in the body, and overall enhances the effectiveness of Epimedh Folium.

Liquiritin apioside and isoliquiritin apioside are flavonoid glycosides commonly found in Glycyrrhizae Radix et Rhizoma. However, for them to be absorbed into the bloodstream, they require the assistance of digestive tract microorganisms to convert them into liquiritigenin and isoliquiritigenin, respectively [28]. Alternatively, when Glycyrrhizae Radix et Rhizoma is stir-fried under heat, liquiritin apioside and isoliquiritin apioside into liquiritigenin and isoliquiritigenin (Fig. 1E, F) [29, 30]. This process facilitates the absorption of active flavonoids into the bloodstream, leading to an increase in their concentration in the body. Therefore, processing plays a crucial role in enhancing the effectiveness of Glycyrrhizae Radix et Rhizoma after processing.

Fig. 1
figure 1

Hydrolysis reaction of flavonoid glycoside components during processing. A Epimedin A is hydrolyzed into sagittatoside A; B Epimedin B is hydrolyzed into sagittatoside B; C Epimedin C is hydrolyzed into 2″-O-rhamnosylicariside II; D 3‴-carbonyl-2″-β-l-quinovosyl icariin is hydrolyzed into icariin; E liquiritin apioside is hydrolyzed into liquiritigenin; F isoliquiritin apioside is hydrolyzed into isoliquiritigenin

Saponins

Saponins can be classified into two categories based on their aglycone structures: triterpenoid saponins and steroidal saponins. These components are abundant in Chinese medicine, with Glycyrrhizae Radix et Rhizoma, Rhizoma Anemarrhenae (Zhimu in Chinese), and Ginseng Radix et Rhizoma (Renshen in Chinese) being rich sources of saponins. During processing with heat, saponins may undergo hydrolysis, leading to the removal of their glycosyl groups.

Glycyrrhizae Radix et Rhizoma is known for its ability to invigorate the spleen and Qi, relieve urgency and pain, and harmonize with other medicines. Its primary component, glycyrrhizic acid, is a typical triterpenoid saponin [31]. However, studies have indicated that the oral bioavailability of glycyrrhizic acid is low, at only 4.0%. It is mainly absorbed through direct or stepwise removal of two molecules of glucuronic acid by intestinal microbial hydrolysis, which generates glycyrrhetinic acid. Glycyrrhetinic acid has a high bioavailability of up to 90% and is the primary form of glycyrrhizic acid that is absorbed into the bloodstream and exerts its effects [32]. Several factors influence the transport of active components across membranes. These factors include the expression and activity of efflux transporters, the strength of membrane permeability, and the state of tight junctions between cells. When triterpenoid saponins undergo deglycosylation and turn into sapogenins, their polarity decreases while their fat solubility increases. As a result, they become more easily absorbed in the intestine. In vivo, glycyrrhetinic acid demonstrates excellent absorption properties. It can further enhance its absorption in the gastrointestinal tract by inhibiting efflux transporters such as P-gp, MRP, and BCRP [33, 34]. During the stir-frying of Glycyrrhizae Radix et Rhizoma, glycyrrhizic acid undergoes hydrolysis due to heat, resulting in the formation of the easily absorbed active component glycyrrhetinic acid (Fig. 2A) [35].

Rhizoma Anemarrhenae possesses medicinal properties such as heat-clearing, fire-purging, yin-nourishing, and dryness-moistening. Its steroidal saponins are considered the main active components, which can be classified into two categories based on the aglycone structure: spirostanol saponins and furostanol saponins. Among these, Timosaponin BII is the most abundant component, accounting for over 70% of the total steroidal saponins in Rhizoma Anemarrhenae [36]. However, the absolute bioavailability of Timosaponin BII in rats was found to be only 0.26%. Nevertheless, research has indicated that processing Rhizoma Anemarrhenae (by str-frying with salt solution) can hydrolyze timosaponin BII into timosaponin AIII under the influence of heat, as illustrated in Fig. 2B [37]. This suggests that processing can enhance the efficacy of Rhizoma Anemarrhenae by promoting the transformation of steroidal saponins into easily absorbable components, such as timosaponin AIII, timosaponin AI, and sarsasapogenin. These are the primary forms that are absorbed into the bloodstream [38, 39].

Ginsenosides, the primary active components of Ginseng Radix et Rhizoma, mainly consist of dammarane-type ginsenosides. These include ginsenosides Rb1, Rc, Rb2, Rd, Re, and Rg1, which are the most abundant [32, 40]. However, due to their high relative molecular mass and structural characteristics, they have poor membrane permeability, low water solubility, and are difficult to absorb in the gastrointestinal tract. Consequently, the oral bioavailability of almost all ginsenosides is low. For instance, the oral bioavailability of ginsenosides Rb1 and Rb2 is only 0.78% and 0.08%, respectively [41]. The presence of glycosyl groups is the main factor contributing to the differences in absorption among various ginsenosides. The higher the number of glycosyl groups, the more hydrogen bonds are formed, resulting in reduced membrane permeability and decreased absorption of most ginsenosides. Therefore, ginsenosides with multiple glycosides are challenging to absorb in vivo [42].

Numerous studies have demonstrated that ginsenosides are not hydrolyzed in the stomach when orally administered. Only a small portion of ginsenosides are absorbed in their original form directly through the small intestine. The primary forms of ginsenosides that are absorbed into the bloodstream and exhibit their medicinal effects are the rare saponins (Rg3, Rg5, Rh2, Rh3, Rk1, Rk2), and the glycosides produced after hydrolysis [43,44,45]. During the steaming process of Ginseng Radix et Rhizoma, ginsenosides containing more glycosyl groups can undergo hydrolysis, resulting in the removal of glycosyl groups and the formation of ginsenosides with fewer glycosyl groups. Specifically, during steaming, the protopanaxatriol-type saponin ginsenoside Rd can undergo deglycosylation at the C20, producing ginsenoside Rg3. Furthermore, ginsenoside Rg3 can undergo further deglycosylation at the C3 position, leading to the production of ginsenoside Rh2 [46,47,48]. Studies have shown that newly formed ginsenosides in the form of mono-glycoside or aglycone are more easily absorbed in the gastrointestinal tract compared to primary saponins that contain more glycosyl groups, such as Ginsenoside Rd [49].

Iridoid glycosides

Iridoids can be categorized into two primary chemical structures: cyclopentane iridoids and cyclopentane-cracked secoiridoids. They are predominantly present as iridoid glycosides and are commonly found in Chinese medicine derived from the Scrophulariaceae, Rubiaceae, and Oleaceae families, such as Rehmanniae Radix (Dihuang in Chinese), Gardeniae Fructus (Zhizi in Chinese) and Ligustri Lucidi Fructus (Nvzhenzi in Chinese). However, iridoid glycosides are susceptible to hydrolysis and thus exhibit instability. During processing, the glycosidic bonds of iridoid glycosides can be cleaved under the influence of heat, resulting in the loss of glycosyl groups through hydrolysis. For example, during the steaming process of Rehmanniae Radix into Rehmanniae Radix Praeparata (Shudihuang in Chinese), iridoid glycosides undergo various degrees of hydrolysis reactions [50]. The extent of hydrolysis is correlated with the number of glycosyl groups, with monoglycosides iridoid glycosides like catalpol being the most prone to hydrolysation (Fig. 2D).

Ligustri Lucidi Fructus is composed mainly of cyclopentane-cracked secoiridoid glycosides [51, 52]. These glycosides are structurally linked to salidroside, tyrosol, or hydroxytyrosol through an ester bond on the cracked cyclopentane. For instance, specnuezhenide and ligustroside G13 are connected to salidroside, while oleuropein and ligustroside are associated with hydroxytyrosol and tyrosol via the ester bond. During processing (steaming), hydrolysis reactions can occur in the iridoid glycosides. This leads to a decrease in the levels of specnuezhenide, ligustroside G13, oleuropein, and ligustroside, while the levels of salidroside, tyrosol, and hydroxytyrosol increase. It has been observed that specnuezhenide can be hydrolyzed to salidroside under heat (Fig. 2E) [53, 54]. The wine steaming of Ligustri Lucidi Fructus enhances its renal protective function by hydrolyzing its iridoid glycosides [55]. This hydrolysis causes changes in the components from large to small molecular weight, facilitating their absorption in the intestinal tract [56].

Gardeniae Fructus contains geniposide, a cyclopentane iridoid glycoside known for its beneficial effects. Although geniposide’s bioavailability is limited due to its poor lipid solubility, despite being highly soluble in water. In rats, oral administration of geniposide resulted in only 4.23% absolute bioavailability compared to intravenous administration. The absorption of geniposide is rapid, occurring within 30 min [57]. When orally administered, most geniposide undergoes deglycosylation by intestinal bacteria and is absorbed into the bloodstream as genipin, its aglycone form [58]. It is commonly believed that the prototype components contained in Chinese medicine undergo metabolism and transformation in vivo, leading to enhanced effects. Processing techniques can facilitate the conversion of difficult-to-absorb components into easily absorbed active components. For example, during processing, geniposide can be hydrolyzed to genipin through heat (Fig. 2F) [59], which increases the content of active components in the body.

Polysaccharides/oligosaccharide

Polysaccharides have garnered considerable attention in Chinese medicine research due to their widespread presence and potential therapeutic effects [60,61,62]. For instance, Ganoderma lucidum polysaccharides have demonstrated promising anti-tumor activity, while Poria cocos polysaccharides have immune-enhancing effects [63, 64]. Additionally, ginseng polysaccharides, lentinan, fucoidan, pachman, and Coriolus versicolor polysaccharides are already being used as polysaccharide drugs in both domestic and foreign markets [65]. Polysaccharides are complex carbohydrates composed of more than 10 monosaccharides [66]. They are abundantly found in the cell walls of botanical Chinese medicine and the cell membrane of animal Chinese medicine. The biological activities of polysaccharides are closely related to their physicochemical properties, such as monosaccharide composition, glycosidic linkage features, molecular weights, and chain conformations. The solubility of polysaccharides directly affects their hydrolysis, absorption, and subsequent biological effects. Generally, polysaccharides are believed to have low oral bioavailability due to their high molecular weight and low stability in the gastrointestinal tract, leading to their direct excretion through urine in the body [67, 68]. There are three potential ways for the absorption of oral polysaccharides: direct absorption [69,70,71], transformation by intestinal microflora [72, 73], and absorption by the intestinal Peyer’s aggregated lymph node [74]. Although progress has been made in understanding the in vivo behavior of polysaccharides, the exact mode of oral absorption is still a subject of debate. However, evidence increasingly suggests that transformation by intestinal microflora plays a significant role. In most cases, polysaccharides are absorbed in the form of oligosaccharides. Upon oral ingestion, polysaccharides are hydrolyzed by the intestinal microflora into various monosaccharides and oligosaccharides. These breakdown products are then transported across the intestinal epithelium as monosaccharides or oligosaccharides and subsequently absorbed.

In addition to hydrolysis by intestinal microflora, polysaccharides can also undergo hydrolysis during processing under the influence of heat. This hydrolysis leads to the production of polysaccharides with slightly lower molecular weight, oligosaccharides, and monosaccharides [75]. Polygoni Multiflori Radix and its processed products have been widely used as herbal preparations for medicinal and health products in China and many other East Asian countries for centuries. Polysaccharides are the main components in Polygoni Multiflori Radix, with the weight of the raw product (660.7 kDa) significantly higher than that of the processed product (344.4 kDa) [76]. Honey-processed Astragali Radix (Huangqi in Chinese), which is Astragali Radix processed with honey, exhibits enhanced efficacy in tonifying Qi compared to raw Astragali Radix. Polysaccharides are the main water-soluble active components in honey-processed Astragali Radix. After processing, Astragali Radix polysaccharide (APS3a) undergoes a hydrolysis reaction, converting the 1,4-β-Galpa and 1,6-α-GlcpA aldehyde acid residues into the corresponding neutral residue in honey-processed Astragali Radix polysaccharide (HAPS3a), resulting in a change in molecular weight from 3373.2 to 2463.5 kDa (Fig. 2G) [77]. However, the oral absorption characteristics of processed HAPS3a, such as whether it is absorbed in its prototype or degraded form, and the transport mode involved in its absorption process, have not been extensively researched and remain unclear. Further studies are needed to address these questions.

The effect of heat during steaming on carbohydrate components can be illustrated and explained using oligosaccharides as an example. Rehmanniae Radix is rich in oligosaccharides, including stachyose and raffinose, with stachyose being the most abundant. Stachyose, an oligosaccharide, is rapidly absorbed but less effectively through oral administration, with a bioavailability of less than 4%. Rehmanniae Radix contains a significant amount of stachyose [78, 79]. However, under the influence of heat during processing, the content of stachyose gradually decreases, while the content of fructose, glucose, galactose, and mannose significantly increases [80,81,82]. One molecule of stachyose can be hydrolyzed into two molecules of galactose, one molecule of fructose, and one molecule of glucose (Fig. 2H) [83]. The hydrolysis of oligosaccharides into monosaccharides during processing (steaming) also contributes to the formation mechanism of Rehmanniae Radix Praeparata, which is described as having a ‘sweet as jelly’ taste.

Fig. 2
figure 2

Hydrolysis reaction of saponins, iridoid glycosides, polysaccharides/oligosaccharide during processing. A Glycyrrhizic acid is hydrolyzed into glycyrrhetinic acid; B timosaponin BII is hydrolyzed into timosaponin AIII; C ginsenoside Rd undergoes deglycosylation at the C20 position to yield ginsenoside Rg3, which further hydrolyzes into generate Rh3; D monoglycosides iridoid glycosides catalpol is hydrolysed; E specnuezhenide is hydrolyzed into salidroside; F geniposide is hydrolyzed into genipin; G APS3a undergoes a hydrolysis reaction, converting the 1,4-β-Galpa and 1,6-α-GlcpA aldehyde acid residues into HAPS3a; H stachyose is hydrolyzed into two molecules of galactose, one molecule of fructose, and one molecule of glucose

Maillard reaction improves protein solubility

The Maillard reaction, also known as glycosylation, is a condensation reaction that occurs between amino-containing components (such as proteins, peptides, or amino acids) and carbonyl-containing components (primarily reducing sugars). Chinese medicine contains significant amounts of proteins, polysaccharides, unsaturated fatty acids, and other components, which makes them prone to undergoing the Maillard reaction during processing. This reaction leads to the formation of Maillard reaction products, which can encapsulate active components and form covalent complexes. The formation of these complexes can offer various benefits, such as improved solubility, emulsification, and stability of the active components. Ultimately, the covalent complexes formed through the Maillard reaction contribute to enhancing the overall quality and efficacy of Chinese medicine.

The Maillard reaction is a complex process that occurs in three stages. In the first stage, a condensation reaction takes place between the free amino group in amino acids or proteins and the carbonyl group in reducing sugars, resulting in the formation of a Schiff base. The Schiff base then undergoes cyclization, leading to the production of N-substituted glucosamine. Through Amadori rearrangement, N-substituted glucosamine transforms into 1-amino-1-deoxy-2-ketose [84, 85]. One notable characteristic of this stage is the conversion of aldose to ketose derivatives. The second stage of the Maillard reaction is still not fully understood, and the principles behind polymer generation in this stage remain unclear. However, certain small-molecule components are known to be produced, including furan rings, nitrogen-containing heterocyclic components, and γ-pyrones.

Recent research has highlighted the pronounced occurrence of the Maillard reaction during the processing (steaming) of red Ginseng Radix et Rhizoma, Polygoni Multiflori Radix (Heshouwu in Chinese) and Rehmanniae Radix [86]. Specifically, during the processing (steaming) of Rehmanniae Radix, there is a rapid decrease in the content of amino acids, especially lysine and arginine. This leads to the formation of various components, including Maltol, 2-ethtylpyrrole, 2,3-dihydro-3,5-dihydroxy-6-methyl-4H-pyran-4-one (DDMP), and 5-HMF [87]. One noteworthy finding is that the Maillard reaction can enhance protein solubility. During this reaction, the free amino groups in amino acid residues, particularly lysine and arginine, react with the reducing carbonyl groups of sugar, resulting in covalent cross-linking. This introduces hydrophilic groups, primarily hydroxyl groups, from the sugar, which reduces the isoelectric point of the glycosylated proteins and improves their solubility. The difference in effectiveness between Rehmanniae Radix and Rehmanniae Radix Praeparata may be attributed to the occurrence of the Maillard reaction during processing [88].

Adjuvants promote the dissolution, absorption, and targeted distribution of active components

To optimize the efficacy of herbs, various liquid adjuvants such as yellow rice wine, vinegar, and honey are commonly employed during processing. Currently, the liquid adjuvants used in processing primarily include wine, vinegar, honey, salt solution, ginger juice, licorice juice, and others.

These adjuvants not only possess therapeutic properties but also enhance the therapeutic effect through compatibility [89]. For example, honey, with its antitussive effects, can synergistically enhance the cough-relieving properties of Ephedrae Herba, leading to prominent effects in relieving cough and asthma [90]. Another common processing method is ginger juice, which enhances the anti-vomiting effect of Bamboo Shavings [91, 92]. In addition to their synergistic effects, adjuvants also play a crucial role in promoting the dissolution, absorption, and targeted distribution of active components in Chinese medicine. These adjuvants help enhance the bioavailability and therapeutic effects of the active components by improving solubility and facilitating absorption in the body.

Adjuvants promote the dissolution of active components

Wine, vinegar, and honey are commonly employed as solvents to enhance the solubility of various active components in Chinese medicine. When heated, adjuvants such as vinegar, wine, and honey help active components dissolve more easily from complex textures, resulting in improved efficacy. The solubility of active components is influenced by multiple factors, including their structure, solvents, co-existing components, and environmental conditions. Adjuvants can exert various mechanisms to enhance solubility. They can form complexes with the active components, altering their chemical properties and improving their solubility in the chosen solvent. Adjuvants may also act as solubilizers by disrupting the intermolecular interactions within the components, allowing them to dissolve more readily. Overall, the use of adjuvants significantly enhances the dissolution of active components, ensuring their availability and efficacy in Chinese medicine.

Vinegar increases the dissolution of active components

Processing with vinegar is a traditional technique that effectively enhances the dissolution of active components by altering the solvent. Corydalis is renowned in Chinese medicine for its ability to promote blood circulation and relieve pain. Processing Corydalis Rhizoma (Yanhusuo in Chinese) with vinegar has been shown to significantly enhance its effects of invigorating the circulation of blood, tonifying Qi, and relieving pain, as evidenced by the clinical practice of Chinese medicine.

Recent research has revealed the mechanisms by which component solubility is enhanced through processing with vinegar. Acetic acid, a key component of vinegar, interacts with the alkaloid present in Corydalis Rhizoma, forming acetates and thereby increasing the solubility of these components in water [93, 94] (Fig. 3A). Furthermore, processing with vinegar has been found to induce changes in the physical properties of certain active components. For instance, in the case of Olibanum (Ruxiang in Chinese), processing with vinegar alters the surface morphology, reduces particle size and polydispersity index, and decreases viscosity. These alterations contribute to an increased dissolution rate of boswellic acid [95]. Vinegar quenching, a common processing method for mineral and crustacean drugs, is known to modify their crystal form and facilitate decoction. This process also generates acetate, an electrolyte with high solubility. For example, Pyritum (Zirantong in Chinese), which is used for its functions of removing blood stasis, relieving pain, and connecting bones and tendons, contains FeS2. Through calcination and vinegar quenching, Pyritum is converted into Fe2O3 and also produces ferrous acetate, thereby enhancing the solubility of the drugs in decoction [96]. This process also forms ferrous acetate, which increases the solubility of drugs in decoction. These findings highlight the scientific basis behind processing with vinegar, emphasizing its role in enhancing the solubility of active components and improving their therapeutic effects.

Wine affects the dissolution of active components

Wine is commonly used as an adjuvant in the CMP. Wine processing involves infusing a certain amount of wine into Chinese medicine by various methods, including stir-frying, stewing, steaming, tempering, and quenching. Ethanol, the main component of wine, is a semi-polar organic solvent that dissolves many active components like flavonoids, phenylpropanoids, terpenes, and steroids, which are easily soluble in ethanol. Additionally, ethanol can also dissolve volatile oils, resins, gums, and other insoluble components [97]. As a result, wine has a better extraction effect on low polar components and can enhance the solubility of active components, thereby increasing their extraction and leaching from Chinese medicine.

Achyranthis Bidentatae Radix (Niuxi in Chinese) is a well-known Chinese medicine that is recognized for its various medicinal properties, including expelling blood stasis, clearing the meridians, tonifying the liver and kidneys, strengthening the tendons and bones, inducing diuresis, and draining blood downstream. The wine-processed Achyranthis Bidentatae Radix is a commonly processed product. Researchers have employed chemometrics combined with quantitative analysis using UHPLC-MS/MS to analyze the components of the wine-processed extract. The results showed that wine processing significantly increases the content of certain components in Achyranthis Bidentatae Radix, such as β-ecdysterone, 25-R inokosterone, 25-S inokosterone, ginsenoside R0, and chikusetsusaponin IVa, compared to raw Achyranthis Bidentatae Radix [98] (Fig. 3B). For instance, wine treatment has been shown to increase the levels of 15 out of 18 inorganic elements in Corni Fructus (Shanzhuyu in Chinese) [99]. These findings suggest that wine processing can improve the extraction and dissolution of certain insoluble components.

Gentianae Radix et Rhizoma (Longdan in Chinese) is traditionally used in Chinese medicine for clearing heat and drying dampness, as well as relieving fire in the liver and gall bladder. Studies have demonstrated that the content of active components, such as gentiakochianoside, 1-O-glucosyl corymbiferin, macrophylloside A, and oleanolic acid, significantly increases in Gentianae Radix et Rhizoma after being stir-fried with wine. These findings suggest that wine processing can improve the extraction and dissolution of certain insoluble components.

Honey promotes the dissolution and absorption of active components

Honey is widely used in CMP to enhance therapeutic effects, particularly in terms of Qi-nourishing and lung moistening [100, 101]. It primarily consists of saccharides and water, with saccharides making up around 60–80% of its composition. The main saccharides in honey are glucose and fructose, with a small amount of sucrose. These saccharides play a significant role in honey’s physical and chemical properties [102].

Studies have shown that honey forms a supramolecular structure due to the strong hydrogen bond between saccharides and water, giving it similar characteristics to natural ionic liquids and deep eutectic solvents (NADES) [103, 104]. NADES are composed of hydrogen bond donors and acceptors with low vapor pressure [103]. They offer several advantages, including strong solubility, low volatility, and the ability to maintain the stability of solute molecules. Recent research has utilized NADES to improve the solubility and stability of active components (such as saponins, flavonoids, polysaccharides, quinones, alkaloids, phenolic acids, volatile oils, etc.) [105,106,107]. Both domestic and international studies have confirmed that NADES significantly improve the solubility and extraction rate of phenols, flavonoids, and other components, as well as promote their absorption [108,109,110,111].

Astragali Radix is traditionally used for tonifying Qi and raising Yang, consolidating the surface and stopping sweating, inducing diuresis, reducing swelling, and nourishing blood. Recent studies have shown that Astragali Radix possesses various pharmacological activities, including immunomodulatory, anti-inflammatory, antioxidant, and antitumor effects [112, 113]. In clinical practice, honey-processed Astragali Radix is commonly used as a Qi-tonifying and immunomodulating Chinese medicine, exhibiting enhanced tonic effects compared to raw Astragali Radix [114, 115]. Researchers have investigated the mechanism underlying the strengthened tonic effect of honey-processed Astragali Radix, and have reported that honey, along with the influence of heat on chemical components during processing, plays a significant role. Honey processing helps maintain the stability of astragaloside II, increases the water-soluble extract of Astragali Radix, and significantly enhances the content of isoflavone glycosides (e.g., calycosin-7-O-β-d-glucoside and ononin, etc.) in Astragali Radix [104] (Fig. 3C). These findings explain the superior Qi-nourishing effect of honey-processed Astragali Radix.

Fig. 3
figure 3

Adjuvants (vinegar, wine, honey) promotes the dissolution of active components. A Acetic acid interacts with alkaloids to form acetate salts, increasing their solubility in water; B wine processing increases the content of active components in Achyranthis Bidentatae Radix; C honey processing increases the water-soluble extract of Astragali Radix

Phase transition induced by adjuvants to increase absorption

Adjuvants can physically change the existing form of active components, which may improve their absorption, and consequently affect their pharmacological effects. This alteration is achieved through non-covalent bonding interactions, including van der Waals forces, hydrogen bonds, π–π superposition, halogen bonds, cation–π interactions, ionic bonds, and CH–π interactions. These interactions facilitate the formation of supramolecular complexes via self-assembly. The formation of such complexes can significantly impact the solubility of insoluble components [116]. Chinese medicine is known for its diverse range of complex components, including flavonoids, saponins, alkaloids, amino acids, and polysaccharides. These components can form aggregates with adjuvants via non-covalent bonding interactions, resulting in the creation of various forms like gels, micelles, spiral bands, and vesicles. These forms serve as solubilizers or permeation enhancers [117].

The self-assembly of nano micelle of mutton tallow

Mutton tallow, obtained from goats or sheep, is a known adjuvant with tonifying and wind-dispelling properties. Epimedh Folium, a Chinese medicine used for osteoporosis, cardiovascular issues, and impotence, has a long history of use. When processed together with mutton tallow, it can enhance its tonifying deficiency and Yang [118,119,120]. The main active components of Epimedh Folium are flavonoids, which have poor solubility and low bioavailability, thereby impacting clinical efficacy [121,122,123]. However, by processing Epimedh Folium with mutton tallow, the solubility of icariin can be increased, improving its intestinal absorption and addressing the issue of poor absorption of active flavonoids [124, 125].

Mutton tallow is known to contain fatty acids such as stearic acid, palmitic acids, and oleic acids, which have long fatty chains and exhibit surface activity. Research has found that these fatty acids in mutton tallow can form mixed micelles with sodium taurocholate, an endogenous bile acid. These micelles act as carriers to enhance the absorption of flavonoids [124]. In the case of Baohuoside I, a representative component of Epimedh Folium, it has been observed that the addition of mutton tallow to Baohuoside I-bile salt micelles results in a more stable system. Baohuoside I and bile were able to form micellar particles with an average particle size of (911.2 ± 15.0) nm and zeta potential of (− 11.7 ± 0.5) mV. However, these particles had large polydispersity coefficients and an uneven distribution of particle sizes. The addition of mutton tallow resulted in a significant decrease in the micellar particle size, a more even distribution of particle sizes compared to the Baohuoside I-bile salt micelles, and a significant increase in the zeta potential [126].

After oral administration of mutton tallow-processed Epimedh Folium, the insoluble active flavonoids can self-assemble with endogenous sodium deoxycholate to form nano micelles. The presence of mutton tallow, which contains stearic acid, palmitic acid, and oleic acid, further promotes the formation of self-assembled micelles, resulting in more stable nano micelles. These nano micelles can then further self-assemble with the action of amphiphilic phospholipids in the body, forming phospholipid-encapsulated nano micelles that play a crucial role in intestinal absorption. This process increases permeability and enhances bioavailability, thereby boosting their synergistic effects. The dynamic self-assembly process significantly improves the solubility and enhances the intestinal absorption of active flavonoids, such as icariin and baohuoside I, ultimately improving their therapeutic efficacy (Fig. 4) [124].

Fig. 4
figure 4

The self-assembly of nano micelle after oral administration of mutton tallow-processed Epimedh Folium

The self-assembly of nano micelle of bile

Bile, a secretion produced by the liver cells of vertebrates, is commonly used in CMP. Fresh bile derived from cows, pigs, and sheep is known for its heat-clearing, restlessness-relieving, gallbladder-benefiting, detoxifying, liver-clearing, and vision-improving properties. Bile is rich in bile acids and bile salts, which are the main components of bile. These components possess a unique structure with a concave hydrophilic face and a convex hydrophobic face [127]. This structural characteristic allows them to form self-assembled supramolecular nano micelles or micelles [128,129,130]. Researchers have demonstrated that bile acid or bile salts can self-assemble with lipids or surfactants to form mixed micelles, which exhibit different solubilization capabilities. These mixed micelles act as carriers, enhancing the absorption rate of insoluble components [131, 132].

Coptidis Rhizoma (Huanglian in Chinese) is renowned for its heat-clearing, dampness-drying, fire-dipping, and toxin-removing properties [133]. The main active components of this herb are protoberberine-type alkaloids, including palmatine, coptisine, and jatrorrhizine [134]. Bile-processed Coptidis Rhizoma is a commonly used processed form of this herb, but the mechanism behind its processing with bile has not yet reached a clear consensus. Pharmacokinetic studies have shown that the maximum plasma concentration (Cmax) of berberine and palmatine doubles in bile-processed Coptidis Rhizoma compared to regular Coptidis Rhizoma, suggesting that bile processing enhances alkaloid absorption [135, 136]. Although the mechanism by which bile improves alkaloid absorption in Coptidis Rhizoma requires further investigation, our research team believes that the self-assembly characteristics of bile acids or bile salts may play a key role. The self-assembly properties of bile acids or bile salts, which can form mixed micelles, may contribute to the enhanced absorption of alkaloids in bile-processed Coptidis Rhizoma. However, further research is necessary to fully comprehend the underlying mechanism and elucidate the specific role of bile in improving the absorption of alkaloids from Coptidis Rhizoma.

Several studies have demonstrated the capacity of bile acids and bile salts to dissolve substances that are typically insoluble. Moreover, phospholipids, which are commonly present in cell membranes, can affect the solubility and stability of these insoluble substances in the intestinal tract. Phospholipids, being amphiphilic, can interact with surfactants like bile salts, resulting in the formation of micelles and lipid aggregates. These structures effectively increase the apparent solubility of insoluble components [137]. The concentration of phospholipids and bile salts in the small intestine has a significant role in shaping the intestinal environment. Research has shown that phospholipids affect the phase behavior of bile components after they are secreted into the duodenum. It has been observed that when the mass fraction of phospholipids is low, the particles formed have a diameter ranging from 3 to 12 nm, but this diameter gradually increases as the amount of phospholipids increases [138]. Therefore, it is believed that the self-assembly properties of bile’s main components (bile salts, bile acids) and the resulting dissolution and absorption of insoluble components, induced by their interaction with phospholipids through phase transition behavior in the intestinal lumen, may contribute to the enhanced potency of the concoction with bile as an adjuvant (Fig. 5). In summary, from a biopharmaceutical perspective, the analysis of the in vivo absorption behavior of insoluble components under bile intervention offers a novel perspective to uncover the scientific basis of CMP.

Fig. 5
figure 5

The self-assembly of nano micelle after oral administration of mutton bile-processed Coptidis Rhizoma

The self-assembly of nano micelle of licorice juice

Herbal juices, such as licorice juice, ginger juice, and black bean juice, are commonly used as adjuvants in CMP. Glycyrrhizae Radix et Rhizoma, one of the oldest and most commonly used Chinese medicines, is documented in the pharmacopeias of China, Japan, the US, and Europe. It plays a crucial role in harmonizing and modifying other herbs in a prescription. Licorice juice is an extract obtained through the decoction of Glycyrrhizae Radix et Rhizoma, and its main components include glycyrrhiza polysaccharide, triterpene saponins, glycyrrhiza protein, flavonoids, etc. [139,140,141,142,143]. .

Glycyrrhiza polysaccharides are key bioactive components found in licorice juice. In recent decades, there has been a growing interest in studying the extraction, separation, and structural characterization of these polysaccharides. The molecular characteristics of polysaccharides play a significant role in their functionality and behavior. They can form nanoparticles with hydrophobic cores and hydrophilic shells in specific solvents, driven by various internal forces such as hydrophobic interactions, van der Waals forces, hydrogen bonds, and electrostatic forces [144]. Glycyrrhiza polysaccharides are known to have complex branched structures and triple helical conformations [145, 146]. These structures can form complex arrangements and encapsulate active small molecules, resulting in the formation of spherical aggregates [147,148,149]. Moreover, glycyrrhiza polysaccharides can interact with metal ions through ion interactions, leading to the formation of hydrogels with three-dimensional networks and hydrophilic porous structures (Fig. 6A) [150, 151]. Previous studies have demonstrated that glycyrrhiza polysaccharides have the potential to enhance the stability and solubility of aconitine, hypaconitine, and benzoylmesaconine in vitro. Furthermore, glycyrrhiza polysaccharides have been found to improve the bioavailability and increase the elimination rate of aconitine, thereby allowing for a reduction in the clinical dosage and mitigating its toxicity. Additionally, glycyrrhiza polysaccharides have been observed to prolong the presence of hypaconitine in the body, thereby extending its therapeutic effect, and enhancing the bioavailability of benzoylmesaconine [152].

Moreover, licorice juice also contains triterpene saponins, which are amphiphilic components consisting of non-polar saponins and water-soluble side chains. During the decoction process, these triterpene saponins can form nanoparticles, thereby increasing the solubility of insoluble components [153]. Glycyrrhizic acid, a typical triterpene saponin found in licorice juice, consists of a hydrophobic triterpene and a hydrophilic sugar chain containing two glucuronides and one carboxyl group [154]. This amphiphilic nature allows glycyrrhizic acid to self-assemble and form non-covalent complexes through hydrophobic interaction [155] (Fig. 6B). Dimeric complexes of glycyrrhizic acid can be observed in solution at concentrations ranging from 0.01 to 1 mmol/L, while at concentrations exceeding 1 mmol/L, large micelle-like aggregates can be formed [156, 157]. At a critical micelle concentration, hydrophobic chain segments (glycoside) form the inner core, while hydrophilic chain segments (sugar chains) form the outer shell, driven by a non-covalent bonding force. This self-assembled micelle effectively enhances the solubility of insoluble components. For instance, the glycyrrhizic acid dimer, formed through the self-assembly of glycyrrhizic acid, can bind hydrophobic molecules on its ring surface, creating a “host-guest complex” that enhances the solubility of hydrophobic components [158]. Studies have demonstrated that glycyrrhizic acid can form a ‘micellar phase’ with puerarin, berberine, and baicalin in Ge-Gen-Qin-Lian-Tang decoction, increasing solubility and absorption [159]. Current research on the solubilization mechanism of glycyrrhizic acid primarily focuses on its supramolecular self-assembly properties, which may explain its solubilization mechanism for certain hydrophobic active components.

Research on the chemical and structural characteristics of nanoparticles indicates that macromolecular components, such as proteins and polysaccharides, often form the primary framework [160, 161]. Proteins, which play a fundamental role in living organisms, exert their biological functions mainly through supramolecular self-assembly [162]. Extensive studies have been conducted on glycyrrhiza proteins, which are rich in tyrosine and tryptophan, revealing their self-assembly behavior during the decoction process to form spherical nanoparticles [163]. It is important to note that proteins achieve their effects through self-assembly via non-covalent interactions. Amino acid residues, acting as weak electrolytes, are distributed on the protein surface, resulting in charge variations at different pH values. This property enables the attraction of electrostatic interactions and facilitates large-scale protein self-assembly [164]. Additionally, glycyrrhiza protein has been observed to encapsulate insoluble components through weak bonding, such as hydrophobic interactions and electrostatic interactions between amide groups and quaternary ammonium ions. This leads to the formation of a subspherical shape [165] (Fig. 6C). The berberine-licorice protein conjugate exhibits a significantly different microscopic morphology compared to the two monomers. It presents a clear globular-like structure with an average particle size of approximately 185.5 nm and a polydispersity index of 0.285, indicating a large-scale self-assembly behavior [165]. The existing body of evidence suggests that glycyrrhiza polysaccharide, triterpene saponins, and glycyrrhiza protein found in licorice juice have the potential to enhance the solubility and absorption of active components. This is achieved by forming aggregates, hydrogel, or micelles via self-assembly. Consequently, their self-assembly characteristics and their ability to improve the solubility and permeability of active components may play a crucial role in their utilization as adjuvants in processing.

Fig. 6
figure 6

The self-assembly of nano micelle polysaccharides, glycyrrhizic acid and protein in licorice juice. A Glycyrrhiza polysaccharides form micellar aggregates and polysaccharide-based hydrogels; B glycyrrhizic acids form nano-micelles; C glycyrrhiza protein form protein nanoparticles

Adjuvants promote the targeted distribution of active components

During the processing of Chinese medicine, adjuvants are carefully chosen based on their specific properties and effects. Wine is used to promote upward movement and cleanse upper-energizer heat, vinegar enhances liver-soothing and analgesic effects, honey increases Qi-nourishing and lung-moistening effects, and salt solution facilitates the delivery of drugs to the kidneys. The concept of meridian tropism in adjuvant processing shares similarities with targeted preparations in pharmacology, as both aim to modify drug behavior in the body to increase their effectiveness. In recent years, extensive research has focused on the scientific implications of the high-concentration distribution of active components in target organs and their potentiation under the influence of adjuvants.

Salt solution promotes active component distribution in the kidney

Salt mainly contains NaCl and trace amounts of MgCl2, CaCl2, KCl, NaI, and other components. Recent research has shed light on the vital physiological roles of sodium ions (Na+) in salt. These ions are essential for maintaining osmotic pressure, facilitating nerve and muscle cell excitation, and aiding nutrient absorption in the gastrointestinal tract [166]. Highly concentrated salt solution is commonly used in processing [167]. Salt solution-frying and salt solution-steaming are the primary techniques mentioned in the Chinese Pharmacopeia (2020 edition) and regional processing norms [168]. According to Chinese medicine theory, processing with salt solution can enhance the transportation of active components to the kidney meridian, thereby enhancing their therapeutic effects on lower jiao disorders [169, 170].

Psoraleae Fructus (Buguzhi in Chinese) is well-known for its ability to ‘warm and invigorate the Kidney-Yang, gather the spirit, and enrich the bone marrow’, as described in the Compendium of Materia Medica. In clinical practice, salt solution-processed Psoraleae Fructus is commonly used in Chinese medicine, and has been shown to enhance its kidney-warming and yang-tonifying effects. This enhancement may be attributed to the effects of salt solution-frying on the absorption behavior of the major components [171,172,173]. Among these components, psoralen and isopsoralen are the two isomers and main active components within Psoraleae Fructus [174]. Processing with the salt solution significantly increases the area under the curve (AUC) for psoralen and isopsoralen and promotes their distribution in the kidneys and related reproductive organs [175, 176]. The use of salt treatment has been shown to enhance the uptake of psoralen and isopsoralen, leading to their increased distribution in various organs such as the heart, liver, spleen, lungs, kidneys, ovaries, and testes, with the reproductive organs experiencing the greatest increase. A comparison of the AUC value revealed that the distribution of isopsoralen in the ovary and testis, as well as the distribution of psoralen in the ovary more than doubled [175].

Morindae Officinalis Radix (Bajitian in Chinese) is renowned for its ability to tonify kidney yang, alleviate rheumatism, and strengthen muscles and bones. Raw Morindae Officinalis Radix is known to dispel wind and dampness, while salt solution-processed Morindae Officinalis Radix enhances its kidney-tonifying and Yang-strengthening effects. The main active components of Morindae Officinalis Radix are Iridoid glycosides, with monotropein being the most abundant [177, 178]. Researchers have investigated the impact of salt solution stir-frying on the distribution of monotropein in Morindae Officinalis Radix and have found that salt solution processing can improve the distribution of monotropein and promote its presence in kidney tissue. These findings align with the traditional theory of ‘salt solution-processing entering the kidney meridian’ [179].

Extensive research has revealed that after undergoing salt processing, the active components are distributed specifically in the kidneys and related reproductive organs, resulting in kidney-tonifying effects. However, the exact mechanisms behind this phenomenon are still not fully understood. The kidney plays a pivotal role in maintaining fluid and electrolyte balance in the body [180]. The renal reabsorption of Na+ is a crucial physiological process carried out by the kidneys [181]. In clinical practice, a significant number of active components in salt-processed medications are absorbed by the kidneys through Na+ reabsorption. Therefore, the targeted distribution of active components following salt processing may be attributed to their absorption by the kidneys via Na+ reabsorption.

Wine affects the upward distribution of active components

According to historical records from the Yuan Dynasty, it was mentioned that when a disease affects the head, face, or surface of limb muscle, Chinese medicine should be stir-fried with wine to allow it to ascend via the power of wine. This indicates that wine can alter the nature of Chinese medicine by promoting an upward direction and cleaning upper-energizer heat. Recent studies have shown that wine facilitates the entry of more active components into the bloodstream, thereby promoting an upward direction and enhancing its ascending efficacy. Scutellariae Radix (Huangqin in Chinese) is a widely used Chinese medicine known for its ability to clear heat, and dry dampness, relieve fire, and detoxify the body [182]. Wine-processed Scutellariae Radix is a specific product derived from Scutellariae Radix and is commonly used in clinical practice to clear upper-energizer heat [183]. The main active components of Scutellariae Radix are flavonoids [184, 185]. Pharmacokinetic studies have revealed that wine processing significantly increases the Cmax and area under the curve (AUC0−t) of certain flavonoids in upper-energizer tissues such as the lungs and heart, while significantly decreasing in middle-energizer and lower-energizer tissues such as the spleen, liver, and kidney [186]. The distribution of flavonoids in the tissues is consistent with the ascending and descending theory, indicating that wine has an ‘induce medicine upward’ effect and concentrates active components in the upper-energizer tissues.

The modern research system for the synergistic mechanism of CMP

The paper proposes a modern research system to understand the synergistic mechanism of CMP, which encompasses three key aspects: (1) the changes in chemical components that occur during processing; (2) the formation of different phase transitions (micelles, aggregates, etc.) under the intervention of the components themselves or adjuvants used in the preparation process, like decoction; (3) the absorption, distribution and metabolic characteristics of active components in vivo, influenced by the adjuvants in the biopharmaceutical process (Fig. 7).

Previous research on the mechanism of CMP has predominantly focused on changes in the ‘content’ of components caused by heat and adjuvants during processing. The increase in the concentration of active components has been considered an important factor contributing to the synergistic effect of CMP. However, recent studies have highlighted that the conversion of chemical components during processing can result in alterations in biopharmaceutical properties, such as solubility and permeability. The active components that exhibit enhanced absorption (with appropriate solubility and permeability) after processing are closely linked to their synergistic effect. Therefore, investigating the conversion of chemical components during processing and its impact on improving the biopharmaceutical properties of active components represents a significant aspect of research into the synergistic mechanism of CMP.

The preparation process, which serves as an intermediate step between CMP and the clinical administration of Chinese medicine, has often been overlooked in previous research on the mechanism of CMP. However, it is crucial to consider that active components within Chinese medicine can form nano aggregates through non-covalent bonds. For example, sugar components are rich in hydrophilic groups, allowing them to self-assemble with other structural units through hydrophobic or hydrogen bonding [187]. Triterpene components, with their rigid skeleton and multiple chiral centers, can easily fold into different forms, forming self-assembled nanoparticles in various media. Current research has discovered that self-assembly behavior can occur within the same component or between different components of Chinese medicine [188, 189]. During the processing, preparation, and biopharmaceutical processes of Chinese medicine, the phenomenon of multi-component self-assembly is observed. Furthermore, adjuvants also possess certain special components that can form a unique form with the active components, altering their solubility and permeability, and thereby influencing their intestinal absorption behavior. Therefore, studying the formation of different phase transitions, such as micelles and aggregates, under the influence of the components themselves or adjuvants during the preparation process (decoction), and their impact on the intestinal absorption behavior of active components, represents an important aspect of research into the synergistic mechanism of CMP.

Oral administration is the primary method of clinical use for Chinese medicine. The absorption of Chinese medicine through oral administration mainly occurs in the small intestine. The chemical components of Chinese medicine may interact with each other within the intestinal absorption barrier network, thereby before and after preparation, influencing their absorption. Therefore, understanding the biopharmaceutical processes of absorption and transit of orally administered Chinese medicine in the gastrointestinal tract is crucial to comprehend their biological effects. By investigating the changes in chemical components in vitro, analyzing the absorption barrier network of the gastrointestinal tract, and exploring the impact of intestinal flora on the absorption, distribution, and metabolism of chemical components, we can delve into the scientific connotations of the CMP from various aspects. We propose establishing a three-dimensional research system, encompassing ‘chemical reaction—phase transition—biopharmaceutical properties’. This system integrates the changes in chemical components during the CMP process, the existing state of active components during preparation, and the absorption, distribution, and metabolism of components during the biopharmaceutical process. Only through such an approach can we provide a comprehensive and multi-level research system to investigate the mechanism of CMP, comprehensively explain how CMP enhances efficiency, and avoid the paradox where the increase or decrease of components after processing contradicts their efficacy.

Fig. 7
figure 7

The modern research system for the synergistic mechanism of CMP

Summary and outlook

The efficacy of Chinese medicine is not only dependent on its active components but also on its physical state and biopharmaceutical properties. Analyzing the physical state and self-assembly behavior of active components during different stages, including processing, boiling, and in vivo absorption, provides a comprehensive understanding of the CMP mechanism. The self-assembly behavior between adjuvants and active components has become a research focus. In most cases, adjuvants can synergize with active components to enhance efficacy and increase solubility. This paper provides a biopharmaceutical perspective on the self-assembly behavior between adjuvants and active components, emphasizing their positive role in component synergy and the enhancement of efficacy by increasing the solubility of insoluble or poorly soluble components.

The absorption of active components is influenced by changes in their physical properties and increased solubility. Biopharmaceutics and pharmacokinetics theories and methods are commonly used to better understand the transmission mechanism between the existing forms of active components and their biopharmaceutical properties in vivo. These theories and methods help investigate how active components are absorbed, distributed, and metabolized in different forms. In recent years, research on the in vivo absorption, distribution, and metabolism of active components has become essential in comprehending the mechanism of CMP. Additionally, there is a growing interest in understanding the processing mechanism from the perspective of transporters, metabolic enzymes, and other in vivo aspects.

In the study of the mechanism of CMP, researchers should adhere to the principle that Chinese medicine originates from clinical practice and serves the clinic, fully considering the clinical needs of Chinese medicine and its application form. In addition to focusing on the active components, researchers should also analyze the impact of the gastrointestinal absorption barrier network and intestinal bacterial flora on the absorption, transportation, metabolism, and changes of the active components within the body. The proposed three levels of “chemical reaction—phase transition—biopharmaceutical properties” in this paper present a comprehensive and integrated research strategy for exploring the mechanism of CMP. The interpretation of the enhanced efficacy of CMP from a biopharmaceutic perspective not only provides a novel viewpoint but also offers a valuable research strategy. This strategy can greatly contribute to guiding the study of CMP mechanisms, quality control of Chinese medicine, quality standardization of decoction pieces, and the development of new medicines in Chinese medicine.

Availability of data and materials

Not applicable.

Abbreviations

CMP:

Chinese medicine processing

APS3a:

Astragali Radix polysaccharide

HAPS3a:

Honey-processed Astragali Radix polysaccharide

DDMP:

3,5-dihydroxy-6-methyl-4 H-pyran-4-one

NADES:

Natural deep eutectic solvents

AUC:

The area under the curve

References

  1. Gu W, Wang H, Su M, Wang Y, Xu F, Hu Q, Cai X, et al. Investigation of tannins transformation in Sanguisorbae Radix over carbonizing by stir-frying. Front Mol Biosci. 2022;9:762224.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Xiong Y, Chen L, Man J, Hu Y, Cui X. Chemical and bioactive comparison of Panax notoginseng root and rhizome in raw and steamed forms. J Ginseng Res. 2019;43:385–93.

    Article  PubMed  Google Scholar 

  3. Qu Z, Bing Y, Zhang T, Zheng Y, Wu S, Ji C, et al. Screening of Q-markers for the wine-steamed Schisandra chinensis decoction pieces in improving allergic asthma. Chin Med. 2023;18:10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Wu F, Li Y, Liu W, Xiao R, Yao B, Gao M, et al. Comparative investigation of raw and processed radix polygoni multiflori on the treatment of vascular dementia by liquid chromatograph–mass spectrometry based metabolomic approach. Metabolites. 2022;12:1297.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Chen Z, Ye SY, Zhu RG. The extraordinary transformation of traditional Chinese medicine: processing with liquid excipients. Pharm Biol. 2020;58:561–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Wang X, Yu Y, Pei L, Gao H. Comparison of the pharmacokinetics of timosaponin AIII, timosaponin BIII, and mangiferin extracted from crude and salt-processed Anemarrhenae Rhizoma by UPLC-MS/MS. RSC Adv. 2023;13:11919–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Hu T, Zhu Y, Zhu J, Yang M, Wang Y, Zheng Q. Wine-processed radix scutellariae alleviates ARDS by regulating tryptophan metabolism through gut microbiota. Front Pharmacol. 2023;13: 1104280.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Yin H, Ni H, Zhang L, Wu W, Wu X, Zhang Z, et al. Untargeted metabolomics coupled with chemometric analysis deducing robust markers for discrimination of processing procedures: wine-processed angelica sinensis as a case study. J Sep Sci. 2021;44:4092–110.

    Article  CAS  PubMed  Google Scholar 

  9. Wu L, Yang Y, Mao Z, Wu J, Ren D, Zhu B, et al. Processing and compatibility of Corydalis yanhusuo: phytochemistry, pharmacology, pharmacokinetics, and safety. Evid Based Complement Alternat Med. 2021;2021:1271953.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Zhang XY, Xu JD, Wang Y, Wu CY, Zhou J, Shen H, et al. Comparing steamed and wine-stewed Rehmanniae radix in terms of Yin-nourishing effects via metabolomics and microbiome analysis. J Ethnopharmacol. 2023;311: 116424.

    Article  CAS  PubMed  Google Scholar 

  11. Li RL, Zhang Q, Liu J, He LY, Huang QW, Peng W, et al. Processing methods and mechanisms for alkaloid-rich Chinese herbal medicines: a review. J Integr Med. 2021;19:89–103.

    Article  CAS  PubMed  Google Scholar 

  12. Liu X, Huang Z, Zhang J, Zhou Y, Zhang Y, Wu M, et al. Comparisons of the anti-inflammatory, antiviral, and hemostatic activities and chemical profiles of raw and charred Schizonepetae Spica. J Ethnopharmacol. 2021;278: 114275.

    Article  CAS  PubMed  Google Scholar 

  13. Abramson A, Caffarel-Salvador E, Khang M, Dellal D, Silverstein D, Gao Y, et al. An ingestible self-orienting system for oral delivery of macromolecules. Science. 2019;363:611–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Guan D, Xuan B, Wang C, Long R, Jiang Y, Mao L, et al. Improving the physicochemical and biopharmaceutical properties of active pharmaceutical ingredients derived from traditional Chinese medicine through cocrystal engineering. Pharmaceutics. 2021;13(12): 2160.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Keck CM, Specht D, Brüßler J. Influence of lipid matrix composition on biopharmaceutical properties of lipid nanoparticles. J Control Release. 2021;338:149–63.

    Article  CAS  PubMed  Google Scholar 

  16. Ahire D, Kruger L, Sharma S, Mettu VS, Basit A, Prasad B. Quantitative proteomics in translational absorption, distribution, metabolism, and excretion and precision medicine. Pharmacol Rev. 2022;74:769–96.

    Article  PubMed  Google Scholar 

  17. Borges G, Fong RY, Ensunsa JL, Kimball J, Medici V, Ottaviani JI, et al. Absorption, distribution, metabolism and excretion of apigenin and its glycosides in healthy male adults. Free Radic Biol Med. 2022;185:90–6.

    Article  CAS  PubMed  Google Scholar 

  18. Boyd BJ, Bergström CAS, Vinarov Z, Kuentz M, Brouwers J, Augustijns P, et al. Successful oral delivery of poorly water-soluble drugs both depends on the intraluminal behavior of drugs and of appropriate advanced drug delivery systems. Eur J Pharm Sci. 2019;137: 104967.

    Article  CAS  PubMed  Google Scholar 

  19. Spleis H, Sandmeier M, Claus V, Bernkop-Schnürch A. Surface design of nanocarriers: key to more efficient oral drug delivery systems. Adv Colloid Interface Sci. 2023;313: 102848.

    Article  CAS  PubMed  Google Scholar 

  20. Rocha B, de Morais LA, Viana MC, Carneiro G. Promising strategies for improving oral bioavailability of poor water-soluble drugs. Expert Opin Drug Discov. 2023;18:615–27.

    Article  PubMed  Google Scholar 

  21. Li K, Liu X, Hou R, Zhao H, Zhao P, Tian Y, et al. Uncovering mechanisms of Baojin Chenfei formula treatment for silicosis by inhibiting inflammation and fibrosis based on serum pharmacochemistry and network analysis. Ecotoxicol Environ Saf. 2023;260: 115082.

    Article  CAS  PubMed  Google Scholar 

  22. Shao D, Liu X, Wu J, Zhang A, Bai Y, Zhao P, et al. Identification of the active compounds and functional mechanisms of Jinshui Huanxian formula in pulmonary fibrosis by integrating serum pharmacochemistry with network pharmacology. Phytomedicine. 2022;102: 154177.

    Article  CAS  PubMed  Google Scholar 

  23. Zhou J, Chen Y, Wang Y, Gao X, Qu D, Liu C. A comparative study on the metabolism of epimedium koreanum nakai-prenylated flavonoids in rats by an intestinal enzyme (lactase phlorizin hydrolase) and intestinal flora. Molecules. 2013;19:177–203.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Sun X, Li Q, Zhang J, Zheng W, Ding Q, Yang J, et al. The reason leading to the increase of icariin in Herba Epimedii by heating process. J Pharm Biomed Anal. 2018;149:525–31.

    Article  CAS  PubMed  Google Scholar 

  25. Chen Y, Wang JY, Jia XB, Tan XB, Hu M. Role of intestinal hydrolase in the absorption of prenylated flavonoids present in Yinyanghuo. Molecules. 2011;16(2):1336–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Shen Y, Lu Y, Gao J, Zhu Y, Wang M, Jing S, et al. Efficient preparation of rare sagittatoside A from epimedin A, by recyclable aqueous organic two-phase enzymatic hydrolysis. Nat Prod Res. 2019;33:3095–102.

    Article  CAS  PubMed  Google Scholar 

  27. Wang Z, Liu C, Yu H, Wu B, Huai B, Zhuang Z, et al. Icaritin preparation from icariin by a special epimedium flavonoid-glycosidase from aspergillus sp.y848 strain. J Microbiol Biotechnol. 2022;32:437–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Qiao X, Ye M, Xiang C, Wang Q, Liu CF, Miao WJ, et al. Analytical strategy to reveal the in vivo process of multi-component herbal medicine: a pharmacokinetic study of licorice using liquid chromatography coupled with triple quadrupole mass spectrometry. J Chromatogr A. 2012;1258:84–93.

    Article  CAS  PubMed  Google Scholar 

  29. Kong S, Li P, Verpoorte R, Li M, Dai Y. Chemical and pharmacological difference between honey-fried licorice and fried licorice. J Ethnopharmacol. 2023;302(Pt A):115841.

    Article  CAS  PubMed  Google Scholar 

  30. Ota M, Makino T. History and the immunostimulatory effects of heat-processed licorice root products with or without honey. J Ethnopharmacol. 2022;292: 115108.

    Article  CAS  PubMed  Google Scholar 

  31. Ding Y, Brand E, Wang W, Zhao Z. Licorice: resources, applications in ancient and modern times. J Ethnopharmacol. 2022;298: 115594.

    Article  CAS  PubMed  Google Scholar 

  32. Wang Y, Pan JY, Xiao XY, Lin RC, Cheng YY. Simultaneous determination of ginsenosides in Panax ginseng with different growth ages using high-performance liquid chromatography-mass spectrometry. Phytochem Anal. 2006;17:424–30.

    Article  CAS  PubMed  Google Scholar 

  33. Koyama M, Shirahata T, Hirashima R, Kobayashi Y, Itoh T, Fujiwara R. Inhibition of UDP-glucuronosyltransferase (UGT)-mediated glycyrrhetinic acid 3-O-glucuronidation by polyphenols and triterpenoids. Drug Metab Pharmacokinet. 2017;32:218–23.

    Article  CAS  PubMed  Google Scholar 

  34. Chen Q, Chen H, Wang W, Liu J, Liu W, Ni P, et al. Glycyrrhetic acid, but not glycyrrhizic acid, strengthened entecavir activity by promoting its subcellular distribution in the liver via efflux inhibition. Eur J Pharm Sci. 2017;106:313–27.

    Article  CAS  PubMed  Google Scholar 

  35. Sung MW, Li PC. Chemical analysis of raw, dry-roasted, and honey-roasted licorice by capillary electrophoresis. Electrophoresis. 2004;25:3434–40.

    Article  CAS  PubMed  Google Scholar 

  36. Sun Y, Liu L, Peng Y, Liu B, Lin D, Li L, et al. Metabolites characterization of timosaponin AIII in vivo and in vitro by using liquid chromatography-mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci. 2015;997:236–43.

    Article  CAS  PubMed  Google Scholar 

  37. Lu L, Liu Y, Ding Y, Hou J, Zhang Y, Xue H, et al. Preparation of highly purified timosaponin AIII from rhizoma anemarrhenae through an enzymatic method combined with preparative liquid chromatography. Nat Prod Res. 2016;30:2364–7.

    Article  CAS  PubMed  Google Scholar 

  38. Dong GM, Yu H, Pan LB, Ma SR, Xu H, Zhang ZW, et al. Biotransformation of timosaponin BII into seven characteristic metabolites by the gut microbiota. Molecules. 2021;26:3861.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Liu Z, Dong X, Ding X, Chen X, Lv L, Li Y, et al. Comparative pharmacokinetics of timosaponin B-II and timosaponin A-III after oral administration of Zhimu-Baihe herb-pair, Zhimu extract, free timosaponin B-II and free timosaponin A-III to rats. J Chromatogr B Analyt Technol Biomed Life Sci. 2013;926:28–35.

    Article  CAS  PubMed  Google Scholar 

  40. Hou M, Wang R, Zhao S, Wang Z. Ginsenosides in Panax genus and their biosynthesis. Acta Pharm Sin B. 2021;11:1813–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Zhao J, Su C, Yang C, Liu M, Tang L, Su W, et al. Determination of ginsenosides Rb1, Rb2, and Rb3 in rat plasma by a rapid and sensitive liquid chromatography tandem mass spectrometry method: application in a pharmacokinetic study. J Pharm Biomed Anal. 2012;64–65:94–7.

    Article  PubMed  Google Scholar 

  42. Zheng Y, Feng G, Sun Y, Liu S, Pi Z, Song F, et al. Study on the compatibility interactions of formula Ding-Zhi-Xiao-Wan based on their main components transport characteristics across Caco-2 monolayers model. J Pharm Biomed Anal. 2018;159:179–85.

    Article  CAS  PubMed  Google Scholar 

  43. Liu H, Yang J, Du F, Gao X, Ma X, Huang Y, et al. Absorption and disposition of ginsenosides after oral administration of Panax notoginseng extract to rats. Drug Metab Dispos. 2009;37:2290–8.

    Article  CAS  PubMed  Google Scholar 

  44. Ye XW, Li CS, Zhang HX, Li Q, Cheng SQ, Wen J, et al. Saponins of ginseng products: a review of their transformation in processing. Front Pharmacol. 2023;14:1177819.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Ding K, Tabuchi Y, Makino T. Effect of steam-processing of the Panax ginseng root on its inducible activity on granulocyte-colony stimulating factor secretion in intestinal epithelial cells in vitro. J Ethnopharmacol. 2022;287:114927.

    Article  CAS  PubMed  Google Scholar 

  46. Lee SM, Bae BS, Park HW, Ahn NG, Cho BG, Cho YL, et al. Characterization of Korean Red Ginseng (Panax ginseng Meyer): history, preparation method, and chemical composition. J Ginseng Res. 2015;39:384–91.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Lee SM. Thermal conversion pathways of ginsenoside in red ginseng processing. Nat Prod Sci. 2014;20:119–25.

    CAS  Google Scholar 

  48. Xie YY, Luo D, Cheng YJ, Ma JF, Wang YM, Liang QL, et al. Steaming-induced chemical transformations and holistic quality assessment of red ginseng derived from Panax ginseng by means of HPLC-ESI-MS/MS(n)-based multicomponent quantification fingerprint. J Agric Food Chem. 2012;60(33):8213–24.

    Article  CAS  PubMed  Google Scholar 

  49. Pan WL, Xue BL, Yang CL, Miao LL, Zhou LL, Chen QY, et al. Biopharmaceutical characters and bioavailability improving strategies of ginsenosides. Fitoterapia. 2018;129:272–82.

    Article  CAS  PubMed  Google Scholar 

  50. Qin KM, Shu YC, Cao G, Li WD, Cai H, Li JS, et al. Thoughts and methods of Chinese materia medica processing-taking research on Rehmanniae Radix processing as an example. Chin Tradit Herb Drugs. 2013;44(11):1363–70.

    CAS  Google Scholar 

  51. He F, Chen L, Liu Q, Wang X, Li J, Yu J. Preparative separation of phenylethanoid and secoiridoid glycosides from Ligustri Lucidi Fructus by high-speed counter-current chromatography coupled with ultrahigh pressure extraction. Molecules. 2018;23:3353.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Gao L, Li C, Wang Z, Liu X, You Y, Wei H, et al. Ligustri lucidi fructus as a traditional Chinese medicine: a review of its phytochemistry and pharmacology. Nat Prod Res. 2015;29(6):493–510.

    Article  CAS  PubMed  Google Scholar 

  53. Li M, Wang X, Han L, Jia L, Liu E, Li Z, et al. Integration of multicomponent characterization, untargeted metabolomics and mass spectrometry imaging to unveil the holistic chemical transformations and key markers associated with wine steaming of Ligustri Lucidi Fructus. J Chromatogr A. 2020;1624:461228.

    Article  CAS  PubMed  Google Scholar 

  54. Shang Z, Xu L, Zhang Y, Ye M, Qiao X. An integrated approach to reveal the chemical changes of Ligustri Lucidi Fructus during wine steaming processing. J Pharm Biomed Anal. 2021;193:113667.

    Article  CAS  PubMed  Google Scholar 

  55. Luan RQ, Zhao P, Zhang XL, Li QQ, Chen XF, Wang L. Pharmacodynamics, pharmacokinetics, and kidney distribution of raw and wine-steamed Ligustri Lucidi Fructus extracts in diabetic nephropathy rats. Molecules. 2023;28(2):791.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Zhang DJ, Sun LL, Li HF, Cui YL, Liu S, Wu P, et al. Pharmacokinetic comparison of nine bioactive components in rat plasma following oral administration of raw and wine-processed Ligustri Lucidi Fructus by ultra-high-performance liquid chromatography coupled with triple quadrupole mass spectrometry. J Sep Sci. 2020;43(21):3995–4005.

    Article  CAS  PubMed  Google Scholar 

  57. Cheng S, Lin LC, Lin CH, Tsai TH. Comparative oral bioavailability of geniposide following oral administration of geniposide, Gardenia jasminoides Ellis fruits extracts and Gardenia herbal formulation in rats. J Pharm Pharmacol. 2014;66:705–12.

    Article  CAS  PubMed  Google Scholar 

  58. Han H, Yang L, Xu Y, Ding Y, Bligh SW, Zhang T, et al. Identification of metabolites of geniposide in rat urine using ultra-performance liquid chromatography combined with electrospray ionization quadrupole time-of-flight tandem mass spectrometry. Rapid Commun Mass Spectrom. 2011;25:3339–50.

    Article  CAS  PubMed  Google Scholar 

  59. Lei L, Wang Y, Huo ZP, Song ZH, Liu LF, He Y. Variations of chemical constituents in Gardeniae Fructus before and after stir-frying by LCMS-IT-TOF. Chin J Exp Tradit Med Formulae. 2019;25(17):88–97.

    Google Scholar 

  60. Hou S, Tan M, Chang S, Zhu Y, Rong G, Wei G, et al. Effects of different processing (Paozhi) on structural characterization and antioxidant activities of polysaccharides from Cistanche deserticola. Int J Biol Macromol. 2023;245:125507.

    Article  CAS  PubMed  Google Scholar 

  61. Jiang T, Wu T, Gao P, Wang L, Yang X, Chen X, et al. Research on processing-induced chemical variations in polygonatum Cyrtonema Rhizome by integrating metabolomics and glycomics. Molecules. 2022;27:5869.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Zhang WJ, Wang S, Kang CZ, Lv CG, Zhou L, Huang LQ, et al. Pharmacodynamic material basis of traditional Chinese medicine based on biomacromolecules: a review. Plant Methods. 2020;16:26.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Wen L, Sheng Z, Wang J, Jiang Y, Yang B. Structure of water-soluble polysaccharides in spore of Ganoderma lucidum and their anti-inflammatory activity. Food Chem. 2022;373(Pt A):131374.

    Article  CAS  PubMed  Google Scholar 

  64. Li YR, Liu ST, Gan Q, Zhang J, Chen N, Han CF, et al. Four polysaccharides isolated from Poria cocos mycelium and fermentation broth supernatant possess different activities on regulating immune response. Int J Biol Macromol. 2023;226:935–45.

    Article  CAS  PubMed  Google Scholar 

  65. Yu Y, Shen M, Song Q, Xie J. Biological activities and pharmaceutical applications of polysaccharide from natural resources: a review. Carbohydr Polym. 2018;183:91–101.

    Article  CAS  PubMed  Google Scholar 

  66. Prajapati VD, Jani GK, Moradiya NG, Randeria NP, Nagar BJ, Naikwadi NN, et al. Galactomannan: a versatile biodegradable seed polysaccharide. Int J Biol Macromol. 2013;60:83–92.

    Article  CAS  PubMed  Google Scholar 

  67. Zheng Z, Pan X, Luo L, Zhang Q, Huang X, Liu Y, et al. Advances in oral absorption of polysaccharides: mechanism, affecting factors, and improvement strategies. Carbohydr Polym. 2022;282: 119110.

    Article  CAS  PubMed  Google Scholar 

  68. Koropatkin NM, Cameron EA, Martens EC. How glycan metabolism shapes the human gut microbiota. Nat Rev Microbiol. 2012;10(5):323.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Xie SZ, Ge JC, Li F, Yang J, Pan LH, Zha XQ, et al. Digestive behavior of Dendrobium huoshanense polysaccharides in the gastrointestinal tracts of mice. Int J Biol Macromol. 2018;107(Pt A):825–32.

    Article  CAS  PubMed  Google Scholar 

  70. Wang K, Cheng F, Pan X, Zhou T, Liu X, Zheng Z, et al. Investigation of the transport and absorption of Angelica sinensis polysaccharide through gastrointestinal tract both in vitro and in vivo. Drug Deliv. 2017;24:1360–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Wang Z, Zhang H, Shen Y, Zhao X, Wang X, Wang J, et al. Characterization of a novel polysaccharide from Ganoderma lucidum and its absorption mechanism in Caco-2 cells and mice model. Int J Biol Macromol. 2018;118(Pt A):320–6.

    Article  CAS  PubMed  Google Scholar 

  72. Flint HJ, Bayer EA, Rincon MT, Lamed R, White BA. Polysaccharide utilization by gut bacteria: potential for new insights from genomic analysis. Nat Rev Microbiol. 2008;6:121–31.

    Article  CAS  PubMed  Google Scholar 

  73. Asano I, Hamaguchi K, Fujii S, Iino H. In vitro digestibility and fermentation of mannooligosaccharides from coffee mannan. Food Sci Technol Res. 2003;9:62–6.

    Article  CAS  Google Scholar 

  74. Han QB. Critical problems stalling progress in natural bioactive polysaccharide research and development. J Agric Food Chem. 2018;66:4581–3.

    Article  CAS  PubMed  Google Scholar 

  75. Sha X, Xu X, Liao S, Chen H, Rui W. Evidence of immunogenic cancer cell death induced by honey-processed Astragalus polysaccharides in vitro and in vivo. Exp Cell Res. 2022;410: 112948.

    Article  CAS  PubMed  Google Scholar 

  76. Wang Y, Yang J, Jin H, Gu D, Wang Q, Liu Y, et al. Comparisons of physicochemical features and hepatoprotective potentials of unprocessed and processed polysaccharides from Polygonum multiflorum Thunb. Int J Biol Macromol. 2023;235:123901.

    Article  CAS  PubMed  Google Scholar 

  77. Wu J, Li C, Bai L, Wu J, Bo R, Ye M, et al. Structural differences of polysaccharides from Astragalus before and after honey processing and their effects on colitis mice. Int J Biol Macromol. 2021;182:815–24.

    Article  CAS  PubMed  Google Scholar 

  78. Li M, Jiang H, Hao Y, Du K, Du H, Ma C, et al. A systematic review on botany, processing, application, phytochemistry and pharmacological action of Radix Rehmnniae. J Ethnopharmacol. 2022;285: 114820.

    Article  CAS  PubMed  Google Scholar 

  79. Ota M, Nakazaki J, Tabuchi Y, Ono T, Makino T. Historical and pharmacological studies on rehmannia root processing-trends in usage and comparison of the immunostimulatory effects of its products with or without steam processing and pretreatment with liquor. J Ethnopharmacol. 2019;242:112059.

    Article  CAS  PubMed  Google Scholar 

  80. Xue S, Wang L, Chen S, Cheng Y. Simultaneous analysis of saccharides between fresh and processed Radix Rehmanniae by HPLC and UHPLC-LTQ-Orbitrap-MS with multivariate statistical analysis. Molecules. 2018;23: 541.

    Article  PubMed  PubMed Central  Google Scholar 

  81. Liu Z, Lou Z, Ding X, Li X, Qi Y, Zhu Z, et al. Global characterization of neutral saccharides in crude and processed Radix Rehmanniae by hydrophilic interaction liquid chromatography tandem electrospray ionization time-of-flight mass spectrometry. Food Chem. 2013;141:2833–40.

    Article  CAS  PubMed  Google Scholar 

  82. Gong PY, Guo YJ, Tian YS, Gu LF, Qi J, Yu BY. Reverse tracing anti-thrombotic active ingredients from dried Rehmannia Radix based on multidimensional spectrum-effect relationship analysis of steaming and drying for nine cycles. J Ethnopharmacol. 2021;276: 114177.

    Article  CAS  PubMed  Google Scholar 

  83. Yu L, Sun SQ, Fan KF, Zhou Q, Noda I. Research on processing medicinal herbs with multi-steps infrared macro-fingerprint method. Spectrochim Acta A Mol Biomol Spectrosc. 2005;62:22–9.

    Article  PubMed  Google Scholar 

  84. Garcia-Amezquita EL, Martinez-Alvarenga SM, Olivas IG, Zamudio-Flores BP, Acosta-Muniz HC. Effect of Maillard reaction conditions on the degree of glycation and functional properties of whey protein isolate-maltodextrin conjugates. Food Hydrocolloid. 2014;38:110–8.

    Article  Google Scholar 

  85. Nooshkam M, Madadlou A. Microwave-assisted isomerisation of lactose to lactulose and Maillard conjugation of lactulose and lactose with whey proteins and peptides. Food Chem. 2016;200:1–9.

    Article  CAS  PubMed  Google Scholar 

  86. Liu Z, Chao Z, Liu Y, Song Z, Lu A. Maillard reaction involved in the steaming process of the root of Polygonum multiflorum. Planta Med. 2009;75:84–8.

    Article  CAS  PubMed  Google Scholar 

  87. Xing Y, Yu Z, Hu X, Yin J, Fan T, Fu Z, et al. Characterization of volatile organic compounds in Polygonum multiflorum and two of its processed products based on multivariate statistical analysis for processing technology monitoring. J Chromatogr A. 2022;1680:463431.

    Article  CAS  PubMed  Google Scholar 

  88. Xia F, Liu C, Wan JB. Characterization of the cold and hot natures of raw and processed Rehmanniae Radix by integrated metabolomics and network pharmacology. Phytomedicine. 2020;74: 153071.

    Article  CAS  PubMed  Google Scholar 

  89. Fukuda M, Kobayashi K, Hirono Y, Miyagawa M, Ishida T, Ejiogu EC, et al. Jungle honey enhances immune function and antitumor activity. Evid Based Complement Alternat Med. 2011;2011:908743.

    Article  PubMed  Google Scholar 

  90. Paul IM, Beiler J, McMonagle A, Shaffer ML, Duda L, Berlin CM Jr. Effect of honey, dextromethorphan, and no treatment on nocturnal cough and sleep quality for coughing children and their parents. Arch Pediatr Adolesc Med. 2007;161:1140–6.

    Article  PubMed  Google Scholar 

  91. Gong J, Huang J, Xiao G, Chen F, Lee B, Ge Q, et al. Antioxidant capacities of fractions of bamboo shaving extract and their antioxidant components. Molecules. 2016;21: 996.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Marx W, Ried K, McCarthy AL, Vitetta L, Sali A, McKavanagh D, et al. Ginger-mechanism of action in chemotherapy-induced nausea and vomiting: a review. Crit Rev Food Sci Nutr. 2017;57:141–6.

    Article  CAS  PubMed  Google Scholar 

  93. Tao Y, Huang S, Yan J, Cai B. Establishment of a rapid and sensitive UPLC-MS/MS method for pharmacokinetic determination of nine alkaloids of crude and processed Corydalis turtschaninovii Besser aqueous extracts in rat plasma. J Chromatogr B Analyt Technol Biomed Life Sci. 2019;1124:218–25.

    Article  CAS  PubMed  Google Scholar 

  94. Wu H, Waldbauer K, Tang L, Xie L, McKinnon R, Zehl M, et al. Influence of vinegar and wine processing on the alkaloid content and composition of the traditional Chinese medicine Corydalis Rhizoma (Yanhusuo). Molecules. 2014;19:11487–504.

    Article  PubMed  PubMed Central  Google Scholar 

  95. Liang D, Ning Z, Song Z, Wang C, Liu Y, Wan X, et al. The effects of vinegar processing on the changes in the physical properties of frankincense related to the absorption of the main boswellic acids. Molecules. 2019;24: 3453.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Zhao GH, Wang ZB, Guo QQ, Chen ZP, Cai BC, Li WD. Dynamic phase variation rule of Pyritum during calcining process. Chin J Exp Tradit Med Formulae. 2015;21(18):1–4.

    Google Scholar 

  97. Ning Y, Pei K, Cao G, Cai H, Liu X, Cao L, et al. Comparative study on pharmacokinetics of four active compounds in rat plasma after oral administration of raw and wine processed Chuanxiong Rhizoma. Molecules. 2019;25: 93.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Yang L, Jiang H, Yan M, Xing X, Guo X, Yang B, et al. UHPLC-MS/MS quantification combined with chemometrics for comparative analysis of different batches of raw, wine-processed, and salt-processed Radix Achyranthis Bidentatae. Molecules. 2018;23: 758.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Lv X, Sun JZ, Xu SZ, Cai Q, Liu YQ. Rapid characterization and identification of chemical constituents in Gentiana Radix before and after wine-processed by UHPLC-LTQ-Orbitrap MSn. Molecules. 2018;23: 3222.

    Article  PubMed  PubMed Central  Google Scholar 

  100. Bogdanov S, Jurendic T, Sieber R, Gallmann P. Honey for nutrition and health: a review. J Am Coll Nutr. 2008;27:677–89.

    Article  CAS  PubMed  Google Scholar 

  101. Huang L, Ye M, Wu J, Liu W, Chen H, Rui W. A metabonomics and lipidomics based network pharmacology study of qi-tonifying effects of honey-processed Astragalus on spleen qi deficiency rats. J Chromatogr B Analyt Technol Biomed Life Sci. 2020;1146: 122102.

    Article  CAS  PubMed  Google Scholar 

  102. Bobis O, Moise AR, Ballesteros I, Reyes ES, Durán SS, Sánchez-Sánchez J, et al. Eucalyptus honey: quality parameters, chemical composition and health-promoting properties. Food Chem. 2020;325: 126870.

    Article  CAS  PubMed  Google Scholar 

  103. Dai Y, Van Spronsen J, Witkamp GJ, Verpoorte R, Choi YH. Natural deep eutectic solvents as new potential media for green technology. Anal Chim Acta. 2013;766:61–8.

    Article  CAS  PubMed  Google Scholar 

  104. Dai Y, Jin R, Verpoorte R, Lam W, Cheng YC, Xiao Y, et al. Natural deep eutectic characteristics of honey improve the bioactivity and safety of traditional medicines. J Ethnopharmacol. 2020;250: 112460.

    Article  CAS  PubMed  Google Scholar 

  105. Li D. Natural deep eutectic solvents in phytonutrient extraction and other applications. Front Plant Sci. 2022;13: 1004332.

    Article  PubMed  PubMed Central  Google Scholar 

  106. Cannavacciuolo C, Pagliari S, Frigerio J, Giustra CM, Labra M, Campone L. Natural deep eutectic solvents (NADESs) combined with sustainable extraction techniques: a review of the green chemistry approach in food analysis. Foods. 2022;12: 56.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Hansen BB, Spittle S, Chen B, Poe D, Zhang Y, Klein JM, et al. Deep eutectic solvents: a review of fundamentals and applications. Chem Rev. 2021;121:1232–85.

    Article  CAS  PubMed  Google Scholar 

  108. Hikmawanti NPE, Ramadon D, Jantan I, Mun’im A. Natural deep eutectic solvents (NADES): phytochemical extraction performance enhancer for pharmaceutical and nutraceutical product development. Plants. 2021;10:2091.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Cvjetko Bubalo M, Ćurko N, Tomašević M, Kovačević Ganić K. Radojčić Redovniković I. Green extraction of grape skin phenolics by using deep eutectic solvents. Food Chem. 2016;200:159–66.

    Article  CAS  PubMed  Google Scholar 

  110. Dai Y, Rozema E, Verpoorte R, Choi YH. Application of natural deep eutectic solvents to the extraction of anthocyanins from Catharanthus roseus with high extractability and stability replacing conventional organic solvents. J Chromatogr A. 2016;1434:50–6.

    Article  CAS  PubMed  Google Scholar 

  111. Huang J, Chen H, Li C, Liu W, Ma W, Rui W. Screening and identification of the metabolites of water extracts of raw and honey-processed Astragalus in rat urine based on UHPLC/ESI-Q-TOF-MS and multivariate statistical analysis. J Am Soc Mass Spectrom. 2018;29:1919–35.

    Article  CAS  PubMed  Google Scholar 

  112. Li Z, Qi J, Guo T, Li J. Research progress of Astragalus membranaceus in treating peritoneal metastatic cancer. J Ethnopharmacol. 2023;305:116086.

    Article  CAS  PubMed  Google Scholar 

  113. Ren C, Zhao X, Liu K, Wang L, Chen Q, Jiang H, et al. Research progress of natural medicine Astragalus mongholicus Bunge in treatment of myocardial fibrosis. J Ethnopharmacol. 2023;305:116128.

    Article  CAS  PubMed  Google Scholar 

  114. Shi J, Zheng L, Lin Z, Hou C, Liu W, Yan T, et al. Study of pharmacokinetic profiles and characteristics of active components and their metabolites in rat plasma following oral administration of the water extract of Astragali radix using UPLC-MS/MS. J Ethnopharmacol. 2015;169:183–94.

    Article  CAS  PubMed  Google Scholar 

  115. Liu W, Li C, Huang J, Liao J, Liao S, Ma W, et al. Application of pathways activity profiling to urine metabolomics for screening Qi-tonifying biomarkers and metabolic pathways of honey-processed Astragalus. J Sep Sci. 2018;41:2661–71.

    Article  CAS  PubMed  Google Scholar 

  116. Liu Y, Wang Q, Li F, Ling D. Nature-inspired supramolecular assemblies for precise biomedical imaging and therapy. Acta Pharm Sin B. 2022;12:4008–10.

    Article  PubMed  PubMed Central  Google Scholar 

  117. Liu Y, Wang S, Qin Y, Wang Y, Yang J, Zhang L, et al. Enhanced TSG stability through co-assembly with C3G: the mechanism behind processing Polygonum multiflorum Thunb with black beans via supramolecular analysis. Food Funct. 2023;14:4204–12.

    Article  CAS  PubMed  Google Scholar 

  118. Wang SW, Ren BX, Qian F, Luo XZ, Tang X, Peng XC, et al. Radioprotective effect of epimedium on neurogenesis and cognition after acute radiation exposure. Neurosci Res. 2019;145:46–53.

    Article  CAS  PubMed  Google Scholar 

  119. Kim JY, Shim SH. Epimedium Koreanum extract and its flavonoids reduced atherosclerotic risk via suppressing modification of human HDL. Nutrients. 2019;11: 1110.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Sun E, Huang R, Ding K, Wang L, Hou J, Tan X, et al. Integrating strategies of metabolomics, network pharmacology, and experiment validation to investigate the processing mechanism of Epimedium fried with suet oil to warm kidney and enhance Yang. Front Pharmacol. 2023;14: 1113213.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. He F, Li M, He Y, Dong Z, Cao J, Dai Z, et al. Authentication of processed Epimedii folium by EA-IRMS. J Anal Methods Chem. 2020;2020:8920380.

    Article  PubMed  PubMed Central  Google Scholar 

  122. Zhang H, Wang H, Wei J, Chen X, Sun M, Ouyang H, et al. Comparison of the active compositions between raw and processed Epimedium from different species. Molecules. 2018;23: 1656.

    Article  PubMed  PubMed Central  Google Scholar 

  123. Zhou J, Ma YH, Zhou Z, Chen Y, Wang Y, et al. Intestinal absorption and metabolism of Epimedium flavonoids in osteoporosis rats. Drug Metab Dispos. 2015;43:1590–600.

    Article  CAS  PubMed  Google Scholar 

  124. Cui L, Sun E, Zhang ZH, Tan XB, Wei YJ, Jin X, et al. Enhancement of epimedium fried with suet oil based on in vivo formation of self-assembled flavonoid compound nanomicelles. Molecules. 2012;17:12984–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Chen Y, Zhao YH, Jia XB, Hu M. Intestinal absorption mechanisms of prenylated flavonoids present in the heat-processed Epimedium Koreanum Nakai (Yin Yanghuo). Pharm Res. 2008;25:2190–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Gu HM, Sun E, Li J, Hou J, Jia XB. Effect of processing excipient suet oil on formation and absorption of baohuoside I-bile salt self-assembled micelles. Chin J Mater Med. 2019;2019(44):5144–50.

    Google Scholar 

  127. Tint GS, Dayal B, Batta AK, Shefer S, Joanen T, McNease L, et al. Biliary bile acids, bile alcohols, and sterols of Alligator mississippiensis. J Lipid Res. 1980;21:110–7.

    Article  CAS  PubMed  Google Scholar 

  128. Tian R, Chen J, Niu R. The development of low-molecular weight hydrogels for applications in cancer therapy. Nanoscale. 2014;6:3474–82.

    Article  CAS  PubMed  Google Scholar 

  129. Hofmann AF, Roda A. Physicochemical properties of bile acids and their relationship to biological properties: an overview of the problem. J Lipid Res. 1984;25:1477–89.

    Article  CAS  PubMed  Google Scholar 

  130. Lv X, Zhang S, Ma H, Dong P, Ma X, Xu M, et al. In situ monitoring of the structural change of microemulsions in simulated gastrointestinal conditions by SAXS and FRET. Acta Pharm Sin B. 2018;8:655–65.

    Article  PubMed  PubMed Central  Google Scholar 

  131. Parekh PY, Patel VI, Khimani MR, Bahadur P. Self-assembly of bile salts and their mixed aggregates as building blocks for smart aggregates. Adv Colloid Interface Sci. 2023;312: 102846.

    Article  CAS  PubMed  Google Scholar 

  132. Macierzanka A, Torcello-Gómez A, Jungnickel C, Maldonado-Valderrama J. Bile salts in digestion and transport of lipids. Adv Colloid Interface Sci. 2019;274: 102045.

    Article  CAS  PubMed  Google Scholar 

  133. Zhou W, Dai Y, Meng J, Wang P, Wu Y, Dai L, et al. Network pharmacology integrated with molecular docking reveals the common experiment-validated antipyretic mechanism of bitter-cold herbs. J Ethnopharmacol. 2021;274: 114042.

    Article  CAS  PubMed  Google Scholar 

  134. Wang J, Wang L, Lou GH, Zeng HR, Hu J, Huang QW, et al. Coptidis Rhizoma: a comprehensive review of its traditional uses, botany, phytochemistry, pharmacology and toxicology. Pharm Biol. 2019;57:193–225.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Zi-Min Y, Yue C, Hui G, Jia L, Gui-Rong C, Wang J. Comparative pharmacokinetic profiles of three protoberberine-type alkaloids from raw and bile-processed Rhizoma coptidis in heat syndrome rats. Pharmacogn Mag. 2017;13:51–7.

    PubMed  PubMed Central  Google Scholar 

  136. Zhou N, Wang Y, Zhang Z, Liu T, Zhang J, Cao Y, et al. Exploring the efficacy mechanism and material basis of three processed Coptidis Rhizoma via metabolomics strategy. J Pharm Biomed Anal. 2023;232: 115450.

    Article  CAS  PubMed  Google Scholar 

  137. Söderlind E, Karlsson E, Carlsson A, Kong R, Lenz A, Lindborg S, et al. Simulating fasted human intestinal fluids: understanding the roles of lecithin and bile acids. Mol Pharm. 2010;7:1498–507.

    Article  PubMed  Google Scholar 

  138. Birru WA, Warren DB, Ibrahim A, Williams HD, Benameur H, Porter CJ, et al. Digestion of phospholipids after secretion of bile into the duodenum changes the phase behavior of bile components. Mol Pharm. 2014;11:2825–34.

    Article  CAS  PubMed  Google Scholar 

  139. Shan L, Yang N, Zhao Y, Sheng X, Yang S, Li Y. A rapid classification and identification method applied to the analysis of glycosides in Bupleuri radix and liquorice by ultra high performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry. J Sep Sci. 2018;41:3791–805.

    Article  CAS  PubMed  Google Scholar 

  140. Zhang Y, Wang Z, Ma X, Yang S, Hu X, Tao J, et al. Glycyrrhetinic acid binds to the conserved P-loop region and interferes with the interaction of RAS-effector proteins. Acta Pharm Sin B. 2019;9:294–303.

    Article  PubMed  Google Scholar 

  141. Selyutina OY, Polyakov NE. Glycyrrhizic acid as a multifunctional drug carrier—from physicochemical properties to biomedical applications: a modern insight on the ancient drug. Int J Pharm. 2019;559:271–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Fan S, Gu K, Wu Y, Luo H, Wang Y, Zhang T, et al. Liquiritinapioside—a mineralocorticoid-like substance from liquorice. Food Chem. 2019;289:419–25.

    Article  CAS  PubMed  Google Scholar 

  143. Ren K, Zhang C, Liu M, Gao H, Ren S, Wang D, et al. The attenuation effect of licorice on the hepatotoxicity of Euodiae Fructus by inhibiting the formation of protein conjugates and GSH depletion. J Ethnopharmacol. 2023;308: 116307.

    Article  CAS  PubMed  Google Scholar 

  144. Simayi Z, Rozi P, Yang X, Ababaikeri G, Maimaitituoheti W, Bao X, et al. Isolation, structural characterization, biological activity, and application of Glycyrrhiza polysaccharides: systematic review. Int J Biol Macromol. 2021;183:387–98.

    Article  CAS  PubMed  Google Scholar 

  145. Pan LC, Zhu YM, Zhu ZY, Xue W, Liu CY, Sun HQ, et al. Chemical structure and effects of antioxidation and against α-glucosidase of natural polysaccharide from Glycyrrhiza inflata Batalin. Int J Biol Macromol. 2020;155:560–71.

    Article  CAS  PubMed  Google Scholar 

  146. Ain NU, Wu S, Li X, Li D, Zhang Z. Isolation, characterization, pharmacology and biopolymer applications of licorice polysaccharides: review. Materials. 2022;15(10):3654.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Wu Y, Zhou H, Wei K, Zhang T, Che Y, Nguyễn AD, et al. Structure of a new glycyrrhiza polysaccharide and its immunomodulatory activity. Front Immunol. 2022;13: 1007186.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Mutaillifu P, Bobakulov K, Abuduwaili A, Huojiaaihemaiti H, Nuerxiati R, Aisa HA, et al. Structural characterization and antioxidant activities of a water soluble polysaccharide isolated from Glycyrrhiza glabra. Int J Biol Macromol. 2020;144:751–9.

    Article  CAS  PubMed  Google Scholar 

  149. Wang Y, Li Y, Ma X, Fan W, Leng F, Yang M, et al. Extraction, purification, and bioactivities analyses of polysaccharides from Glycyrrhiza uralensis. Ind Crop Prod. 2018;122:596–608.

    Article  CAS  Google Scholar 

  150. Gao W, Zhang Y, Zhang Q, Zhang L. Nanoparticle-hydrogel: a hybrid biomaterial system for localized drug delivery. Ann Biomed Eng. 2016;44(6):2049–61.

    Article  PubMed  PubMed Central  Google Scholar 

  151. Moura MJ, Faneca H, Lima MP, Gil MH, Figueiredo MM. In situ forming chitosan hydrogels prepared via ionic/covalent co-cross-linking. Biomacromol. 2011;12(9):3275–84.

    Article  CAS  Google Scholar 

  152. Sun L, You G, Zheng F, Wang M, Ren X, Deng Y. In vitro and in vivo evaluation of the influences of polysaccharides derived from Glycyrrhiza uralensis on three alkaloids and potential interaction mechanisms. Int J Biol Macromol. 2020;157:452–60.

    Article  CAS  PubMed  Google Scholar 

  153. Mesgarzadeh I, Akbarzadeh AR, Rahimi R. Correction to surface-active properties of solvent-extracted Panax ginseng saponin-based surfactants. J Surfact Deterg. 2017;20:609–14.

    Article  CAS  Google Scholar 

  154. Selyutina OY, Polyakov NE, Korneev DV, Zaitsev BN. Influence of glycyrrhizin on permeability and elasticity of cell membrane: perspectives for drugs delivery. Drug Deliv. 2016;23:858–65.

    Article  PubMed  Google Scholar 

  155. Zelikman MV, Kim AV, Medvedev NN, Selyutina OY, Polyakov NE. Structure of dimers of glycyrrhizic acid in water and their complexes with cholesterol: molecular dynamics simulation. J Struct Chem. 2015;56:67–76.

    Article  CAS  Google Scholar 

  156. Polyakov NE. Glycyrrhizic acid as a novel drug delivery vector: synergy of drug transport and efficacy. Open Conf Proc J. 2011;2:64–72.

    Article  CAS  Google Scholar 

  157. Zhao X, Zhang H, Gao Y, Lin Y, Hu J. A simple injectable moldable hydrogel assembled from natural glycyrrhizic acid with inherent antibacterial activity. ACS Appl Bio Mater. 2020;3:648–53.

    Article  CAS  PubMed  Google Scholar 

  158. Polyakov NE, Leshina TV, Salakhutdinov NF, Kispert LD. Host-guest complexes of carotenoids with beta-glycyrrhizic acid. J Phys Chem B. 2006;110:6991–8.

    Article  CAS  PubMed  Google Scholar 

  159. Lin D, Du Q, Wang H, Gao G, Zhou J, Ke L, et al. Antidiabetic micro-/nanoaggregates from Ge-Gen-Qin-Lian-Tang decoction increase absorption of baicalin and cellular antioxidant activity in vitro. Biomed Res Int. 2017;2017:9217912.

    Article  PubMed  PubMed Central  Google Scholar 

  160. Zhou J, Liu J, Lin D, Gao G, Wang H, Guo J, et al. Boiling-induced nanoparticles and their constitutive proteins from Isatis indigotica Fort. Root decoction: purification and identification. J Tradit Complement Med. 2016;7(2):178–87.

    Article  PubMed  PubMed Central  Google Scholar 

  161. Weng Q, Cai X, Zhang F, Wang S. Fabrication of self-assembled Radix Pseudostellariae protein nanoparticles and the entrapment of curcumin. Food Chem. 2019;274:796–802.

    Article  CAS  PubMed  Google Scholar 

  162. Bai Y, Luo Q, Liu J. Protein self-assembly via supramolecular strategies. Chem Soc Rev. 2016;45(10):2756–67.

    Article  CAS  PubMed  Google Scholar 

  163. Zhou J, Zhang J, Gao G, Wang H, He X, Chen T, et al. Boiling licorice produces self-assembled protein nanoparticles: a novel source of bioactive nanomaterials. J Agric Food Chem. 2019;67:9354–61.

    Article  CAS  PubMed  Google Scholar 

  164. Tanaka S, Kerfeld CA, Sawaya MR, Cai F, Heinhorst S, Cannon GC, et al. Atomic-level models of the bacterial carboxysome shell. Science. 2008;319(5866):1083–6.

    Article  CAS  PubMed  Google Scholar 

  165. Li W, Wang ZJ, Liu XJ, Han NN, Li T, Lei HM, et al. Based on weak bond chemistry, the interaction mechanism between glycyrrhiza protein and berberine in water decocting process of Rhizoma Coptidis and Liquorice was investigated. Acta Pharm Sin. 2021;56:8.

    Google Scholar 

  166. Itoh N, Tsuya A, Togashi H, Kimura H, Konta T, Nemoto K, et al. Increased salt intake is associated with diabetes and characteristic dietary habits: a community-based cross-sectional study in Japan. J Clin Biochem Nutr. 2022;71:143–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Chen LL, Verpoorte R, Yen HR, Peng WH, Cheng YC, Chao J, et al. Effects of processing adjuvants on traditional Chinese herbs. J Food Drug Anal. 2018;26:96–114.

    Article  Google Scholar 

  168. Ji D, Su X, Huang Z, Wang Q, Lu T. A novel ultra high-performance liquid chromatography-tandem mass spectrometry method for the simultaneous determination of xanthones and steroidal saponins in crude and salt-processed Anemarrhenae Rhizoma aqueous extracts. J Sep Sci. 2018;41:2310–20.

    Article  CAS  PubMed  Google Scholar 

  169. Hou A, Lv J, Zhang S, Zhang J, Yang L, Jiang H, et al. Salt processing: a unique and classic technology for Chinese medicine processing. Front Pharmacol. 2023;14: 1116047.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Du W, Lv Y, Wu H, Li Y, Tang R, Zhao M, et al. Research on the effect of Dipsaci Radix before and after salt-processed on kidney Yang deficiency syndrome rats and the preliminary mechanism study through the BMP-Smad signaling pathway. J Ethnopharmacol. 2023;312: 116480.

    Article  CAS  PubMed  Google Scholar 

  171. Li K, Zhou N, Zheng XK, Feng WS, Li F, Zhang ZL, et al. Quantitative analysis, pharmacokinetics and metabolomics study for the comprehensive characterization of the salt-processing mechanism of Psoraleae Fructus. Sci Rep. 2019;9:661.

    Article  PubMed  PubMed Central  Google Scholar 

  172. Lu J, Liu L, Zhu X, Wu L, Chen Z, Xu Z, et al. Evaluation of the absorption behavior of main component compounds of salt-fried herb ingredients in Qing’e pills by using Caco-2 cell model. Molecules. 2018;23: 3321.

    Article  PubMed  PubMed Central  Google Scholar 

  173. Lu J, Liu L, Zhu X, Wu L, Chen Z, Xu Z, et al. Evaluation of the absorption behavior of main component compounds of salt-fried herb ingredients in Qing’e pills by using Caco-2 cell model. Molecules. 2018;23(12):3321.

    Article  PubMed  PubMed Central  Google Scholar 

  174. Yang J, Yang J, Du J, Feng Y, Chai X, Xiao M, et al. General survey of Fructus Psoraleae from the different origins and chemical identification of the roasted from raw Fructus Psoraleae. J Food Drug Anal. 2018;26:807–14.

    Article  CAS  PubMed  Google Scholar 

  175. Zhao GH, Yan CP, Xu ZS, Gao QQ, Chen ZP, Li WD. The effect of salt-processed Psoralea corylifolia on generative organ targeting. J Anal Methods Chem. 2016;2016:7484202.

    Article  PubMed  PubMed Central  Google Scholar 

  176. Hou J, Lin S, Lu J, Wu Y, Wu L, Chen Z, et al. Establishment of a UPLC-MS/MS method for studying the effect of salt-processing on tissue distribution of twelve major bioactive components of Qing’e pills in rats. J Anal Methods Chem. 2020;2020:8832736.

    Article  PubMed  PubMed Central  Google Scholar 

  177. Zhang JH, Xin HL, Xu YM, Shen Y, He YQ, Hsien-Yeh, et al. Morinda officinalis How.—a comprehensive review of traditional uses, phytochemistry and pharmacology. J Ethnopharmacol. 2018;213:230–55.

    Article  CAS  PubMed  Google Scholar 

  178. Kang L, Zhang Y, Zhou L, Yang J, He Y, Yang S, et al. Structural characterization and discrimination of Morinda officinalis and processing Morinda officinalis based on metabolite profiling analysis. Front Chem. 2022;9:803550.

    Article  PubMed  PubMed Central  Google Scholar 

  179. Shi J, Jing HY, Huang YQ, Fan YN, Jia TZ. Effects of monotropein in different processing products of Morindae officinalis Radix on plasma concentration and tissue distribution in rats. Chin J Inf Tradit Chin Med. 2017;24:76–81.

    Google Scholar 

  180. Gonzalez-Vicente A, Saez F, Monzon CM, Asirwatham J, Garvin JL. Thick ascending limb sodium transport in the pathogenesis of hypertension. Physiol Rev. 2019;99(1):235–309.

    Article  CAS  PubMed  Google Scholar 

  181. Minegishi S, Ishigami T, Kino T, Chen L, Nakashima-Sasaki R, Araki N, Yatsu K, et al. An isoform of Nedd4-2 is critically involved in the renal adaptation to high salt intake in mice. Sci Rep. 2016;6: 27137.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Zhao Q, Chen XY, Martin C. Scutellaria baicalensis, the golden herb from the garden of Chinese medicinal plants. Sci Bull. 2016;61:1391–8.

    Article  CAS  Google Scholar 

  183. Cui CL, He X, Dong CL, Song ZJ, Ji J, Wang X, et al. The enhancement mechanism of wine-processed Radix Scutellaria on NTG-induced migraine rats. Biomed Pharmacother. 2017;91:138–46.

    Article  CAS  PubMed  Google Scholar 

  184. Li C, Lin G, Zuo Z. Pharmacological effects and pharmacokinetics properties of Radix Scutellariae and its bioactive flavones. Biopharm Drug Dispos. 2011;32:427–45.

    Article  PubMed  Google Scholar 

  185. Wang F, Wang B, Wang L, Xiong ZY, Gao W, Li P, et al. Discovery of discriminatory quality control markers for Chinese herbal medicines and related processed products by combination of chromatographic analysis and chemometrics methods: Radix Scutellariae as a case study. J Pharm Biomed Anal. 2017;138:70–9.

    Article  CAS  PubMed  Google Scholar 

  186. Huang P, Tan S, Zhang YX, Li JS, Chai C, Li JJ, et al. The effects of wine-processing on ascending and descending: the distribution of flavonoids in rat tissues after oral administration of crude and wine-processed Radix scutellariae. J Ethnopharmacol. 2014;155:649–64.

    Article  CAS  PubMed  Google Scholar 

  187. Datta S, Bhattacharya S. Multifarious facets of sugar-derived molecular gels: molecular features, mechanisms of self-assembly and emerging applications. Chem Soc Rev. 2015;44:5596–637.

    Article  CAS  PubMed  Google Scholar 

  188. Huang X, Wang P, Li T, Tian X, Guo W, Xu B, et al. Self-assemblies based on traditional medicine berberine and cinnamic acid for adhesion-induced inhibition multidrug-resistant staphylococcus aureus. ACS Appl Mater Interfaces. 2020;12:227–37.

    Article  CAS  PubMed  Google Scholar 

  189. Li T, Wang P, Guo W, Huang X, Tian X, Wu G, et al. Natural berberine-based Chinese herb medicine assembled nanostructures with modified antibacterial application. ACS Nano. 2019;13:6770–81.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Thank you to my co-authors for their support in this work.

Funding

This work is supported by the National Natural Science Foundation of China (82204626, 82230117) and Jiangsu Funding Program for Excellent Postdoctoral Talent (2022ZB317).

Author information

Authors and Affiliations

Authors

Contributions

BY and XBJ conceived and designed this review. BY, ZBZ, JJS, THQ, and JQZ analyzed the literature and summarized the results. BY and ZBZ reviewed and edited this review. LF revised this review. All of the authors have read and approved the final manuscript.

Corresponding authors

Correspondence to Liang Feng or Xiaobin Jia.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

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/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yang, B., Zhang, Z., Song, J. et al. Interpreting the efficacy enhancement mechanism of Chinese medicine processing from a biopharmaceutic perspective. Chin Med 19, 14 (2024). https://doi.org/10.1186/s13020-024-00887-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13020-024-00887-0

Keywords