• Users Online: 1039
  • Print this page
  • Email this page

 
Table of Contents
REVIEW ARTICLE
Year : 2022  |  Volume : 8  |  Issue : 1  |  Page : 1-20

Forsythosides as essential components of Forsythia-based traditional chinese medicines used to treat inflammatory diseases and COVID-19


OncoWitan, Scientific Consulting Office, Lille (Wasquehal, 59290), France

Date of Submission07-Dec-2020
Date of Acceptance22-Feb-2021
Date of Web Publication16-Sep-2021

Correspondence Address:
Dr. Christian Bailly
OncoWitan, Lille (Wasquehal), 59290
France
Login to access the Email id

Source of Support: None, Conflict of Interest: None


DOI: 10.4103/wjtcm.WJTCM_36_21

Rights and Permissions
  Abstract 


The dried fruits of the plant Forsythia suspensa (Forsythia Fructus: Lianqiao in Chinese) are used in many herbal preparations to treat various diseases or the associated symptoms. Forsythia extracts contain phenylethanoid glycosides (PhGs) such as the forsythosides (Fst A-to-P). The leading products, Fst-A,-B and-F (arenarioside), can be found also in >90 other plants inventoried here. The pharmacological properties of Fst are reviewed, with emphasis on their anticancer, antiviral, and antibacterial activities, which essentially derive from their anti-inflammatory and antioxidant effects. Fst-B functions as a potential binder of the repressor protein Kelch-like ECH-association protein 1 (Keap 1), thus promoting the nuclear translocation of the transcription factor Nuclear factor erythroid 2-related factor 2 (Nrf2) implicated in the subsequent activation of the production of antioxidant enzymes and repression of the oxidative stress. The regulation of the Nrf2/Heme oxygenase-1 pathway is the central piece of the multifaceted mechanism of action of Fst-A/B. Their prominent antioxidant and anti-inflammatory effects support the use of these compounds in different inflammation-related diseases and conditions, from sepsis to neuroprotection and many other pathologies discussed here. In addition, these properties contribute to the antiviral action of the compounds. Fst-A/B displays activities against the influenza A virus and different Fst-containing traditional Chinese medicinal (TCMs) have revealed beneficial effects to combat the current COVID-19 pandemic. The mechanisms whereby Fst-A/B could inhibit viral multiplication are discussed. PhGs likely contribute to the anti-COVID-19 activities reported with several TCM such as Shuang-Huang-Lian oral liquid, Lianhua-Qingwen capsules, and others. This review highlights the pharmacological profile of Fst and illustrates health benefits associated with the use of Forsythia Fructus.

Keywords: Chinese medicine; COVID-19; forsythia; forsythoside; mechanism of action; natural products; phenylethanoid glycosides


How to cite this article:
Bailly C. Forsythosides as essential components of Forsythia-based traditional chinese medicines used to treat inflammatory diseases and COVID-19. World J Tradit Chin Med 2022;8:1-20

How to cite this URL:
Bailly C. Forsythosides as essential components of Forsythia-based traditional chinese medicines used to treat inflammatory diseases and COVID-19. World J Tradit Chin Med [serial online] 2022 [cited 2023 Jun 2];8:1-20. Available from: https://www.wjtcm.net/text.asp?2022/8/1/1/336830




  Introduction Top


The forsythosides (Fst) represent a small group of natural products found in several traditional Chinese medicinal (TCM) preparations. The subgroup includes 16 members, named Fst A-to-P, initially isolated from Forsythia species. The compounds are listed in [Table 1] together with their characteristics. The best-known members are Fst-A and forsythoside B (Fst-B) isolated from the plant Forsythia suspensa (Thunberg) Vahl. (weeping forsythia) originating from China. The plant has hollow, pendulous stems about 3 m long and golden-yellow flowers [Figure 1]. Many medicinal preparations include either the plant F. suspensa or the dried fruits of the plant commonly referred to as Forsythia fructus (Lianqiao in Chinese). Both the greenish fruits (Qingqiao) and the yellow fruits (Laoqiao) are used for medical applications.[1],[2],[3],[4] Aqueous extracts of Forsythia Fructus (Lianqiao) are considered as fundamental components in TCM, being among the top ten of the most potent anti-inflammatory herbs[5]
Table 1: Plants containing forsythosides

Click here to view
Figure 1: Illustrations of the plant Forsythia suspensa (Thunberg) Vahl. (flowers and leaves) and its fruits called Forsythia Fructus (known as Lianqiao in Chinese, and Rengyo in Japanese) from which large quantities of forsythoside A can be isolated. The compound is also known as forsythiaside A (3,4-dihydroxy-β-phenethyl-O-α-L-rhamnopyranosyl-(1 → 6)-4-O-caffeoyl-β-d-glucopyranoside)

Click here to view


F. suspensa is truly a reservoir of natural products. The plant and the fruits contain diverse flavonoids, alkaloids, and phenylethanoid or phenylpropanoid glycosides.[3],[6] The Fst represent one of the main subgroups of phenylethanoid glycosides (PhGs) found in F. suspensa, but the compounds are also well represented in different species of Phlomis, Callicarpa, Marrubium, Verbascum, and others. Fst compounds could be identified in about 90 plants inventoried in [Table 1]. However, the major medicinal material containing Fst is F. suspensa (and Forsythia Fructus) commonly used to prepare several TCMs. Nineteen TCM preparations made from F. suspensa can be identified [Table 2]. They are used for the treatment of respiratory diseases and infections (such as Siji-kangbingdu mixture, Shufeng Jiedu capsule, Tianreqing injection, and Shuang Huang Lian injection), while others are used to treat liver diseases (such as Dahuang Zhechong pill and Li-Dan-He-Ji granules).[7],[8],[9],[10],[11],[12],[13],[14],[15],[16],[17],[18],[19],[20],[21],[22],[23],[24],[25],[26],[27],[28],[29],[30],[31],[32],[33],[34],[35],[36],[37],[38],[39],[40],[41],[42],[43],[44],[45],[46],[47],[48],[49],[50],[51],[52],[53],[54],[55],[56],[57],[58],[59],[60],[61],[62],[63],[64],[65],[66],[67],[68],[69],[70],[71],[72],[73],[74],[75],[76],[77],[78],[79],[80],[81],[82],[83],[84],[85],[86],[87],[88],[89],[90],[91],[92]
Table 2: Traditional chinese medicine preparations containing forsythosides

Click here to view


Interestingly, several recent studies have highlighted the use of Forsythia Fructus-containing TCM to treat the current COVID-19 pandemic. This is the case of Lianhuaqingwen capsule and Shufeng Jiedu capsule, which are both proposed to alleviate the symptoms of the infectious viral disease. Lianhuaqingwen has shown efficacy in COVID-19 patients to reduce the symptoms associated with the coronavirus pneumonia.[93],[94],[95],[96],[97],[98],[99] Fst-A and Fst-I would contribute importantly to the anti-COVID-19 activity.[100] Tianreqing injection is also a medication tested with some success in patients suffering from viral pneumonia,[101],[102],[103],[104] and the preparation contains significant quantities of Fst-A and Fst-E.[105],[106] Similarly, different studies have pointed out the efficacy of Shufeng Jiedu capsules to treat COVID-19[107],[108],[109],[110],[111] and here again, Fst-A seems to play a role in the antiviral and anti-inflammatory effects of the herbal preparation.[112],[113],[114],[115],[116],[117],[118],[119],[120],[121],[122],[123],[124],[125],[126],[127],[128],[129],[130],[131],[132],[133],[134],[135],[136],[137],[138],[139],[140],[141],[142],[143]

For these reasons, it is timely to review the pharmacological properties of the Fst and their potential therapeutic uses, but also the limitations associated with the use of these compounds. The present review starts with a brief survey of the molecular diversity of the Fst and then provides a comprehensive analysis of the pharmacological properties of the best known-members in the series, mainly Fst-A and Fst-B (also known as forsythiasides A and B, respectively).


  Molecular Diversity of the Forsythosides Top


here are 16 Fst, designated Fst A-to-P, but one of them (Fst-L) could not be identified. The structures of the 15 other Fst compounds are presented in [Figure 2]. Fst-A was first isolated in 1981 in Japan, from the fruits (Forsythia fructus called “rengyo” in Japanese) and the leaves of the plant F. suspensa.[144] The compound is composed of a central disaccharide unit (L-rhamnose-(1→6)-D-glucose) substituted with a caffeoyl group at position C-4 on the glucose unit and a 3,4-hydroxyphenylethanol (hydroxytyrosol) unit at position C-1 of the glucose residue [Figure 2]. It is an isomer of verbascoside (also known as acteoside or kusaginin) which bears the rhamnose unit at position C-3. Fst-B was isolated from F. suspensa a few months later by the same group in Japan.[145] The molecule is characterized by the presence of an atypical β-D-apiosyl unit at position C-6 on the central glucose unit. It is a trisaccharide with β-D-apiosyl-(1→6), α-L-rhamnosyl-(1→3)-β-D-glucosyl unit. Fst-B (verbascoside 6'-0-β-D-apiofuranoside) is abundant in the stalk of Forsythiae Fructus but not found in the seeds and pericarp, whereas Fst-A can be found at a relatively high level in each segment.[146] It was rapidly followed with the discovery of Fst-C and Fst-D in 1982, again by the same Japanese research group.[143] They both include a phenylethane-1,2-diol moiety instead of the phenylethanol unit of Fst-A and Fst-B. Fst-C is a hydroxyl derivative of Fst-A, whereas Fst-D lacks the caffeoyl unit, as it is the case for Fst-E [Figure 2]. These four compounds, Fst-A/B/C/D, exhibit a very mild antibacterial activity.[144],[145],[146],[147]
Figure 2: Chemical structures of the forsythosides and the corresponding discovery dates. forsythoside-A (PubChem CID#5281773, C29H36O15), Fst-B (CID# 23928102, C34H44O19), Fst-C (C29H36O16), Fst-D (C20H30O13), Fst-E (CID# 69634125, C20H30O12), Fst-F (CID# 6442994, C34H44O19), Fst-G (CID# 101231533, C35H46O19), Fst-H (CID# 129449684, C29H35O15), Fst-I (CID# 23958169, C29H35O15), Fst-J (C28H33O15), Fst-K (C30H38O16), Fst-M (C22H26O10), Fst-N (C22H26O10), Fst-O (C24H28O11) and Fst-P (C29H36O14). Note that the structure of forsythoside-D cited in PubMed (CID# 24721571) is incorrect. The structure indicated presents a furofuranyl core corresponding to phyllirin, not forsythoside-D. The structure shown here corresponds to the original structure determined for forsythoside-D in 1982[144]

Click here to view


Fst-A and Fst-B are very abundant in the plant kingdom and have been isolated from many plants [Table 1]. F. suspensa produces usually a large quantity of fruits and the plant can be cultivated. Nevertheless, another method to produce Fst-B consists in transforming a root culture of a Fst-producing plant with the bacteria Agrobacterium rhizogenes. The method has been described to produce verbascoside and Fst-B from transformed root culture of Verbascum xanthophoeniceum.[148] It can be applied to produce other PhGs.[149],[150] Callus and cell suspension cultures of F. suspensa, Forsythia viridissima, and Forsythia koreana can be made as well, to produce phenylethanoid derivatives, such as cornoside, vervascoside, and Fst-F (arenarioside).[151]

The three Forsythia species, F. suspensa, F. viridissima, and F. koreana, have been used as herbal medicines in China, Japan, and Korea for centuries and they are known to be rich sources of phenylpropanoid glycosides.[33] Fst-E (decaffeoyl- Fst A) was first isolated in 1984 from the fruits of F. suspensa.[152] Fst-F and-G were isolated 6 years later from the stems of F. viridissima.[34] Fst-F presents a trisaccharide core (α-rhamnosyl-(1 → 3)-glucosyl-(6 → 1)-β-xylosyl) and Fst-G bears an unusual D-2-O-methylapiose unit (α-rhamnosyl-(1 → 3)-glucosyl-(6 → 1)-β-(2-O-methyl) apiosyl). In the literature, Fst-F is often cited as arenarioside [Table 1], initially isolated from the plant Orobanche arenaria.[153] This compound can be found in different Orobanche species.[154] Fst-F is one of the rare PhGs, for which the total organic synthesis of the trisaccharide moiety and a synthetic benzyl protected derivative have been described.[155],[156] Several multisteps total syntheses of different natural PhGs have been reported but not for the Fst.[157],[158],[159],[160]

Fst-H,-I and-J were isolated more recently in 2009, again from the plant F. suspensa.[161] They possess all three a disaccharide core, a caffeoyl group, and a dihydroxyphenylethyl moiety but they differ by the nature of the sugar units and/or the linkage. Fst-H and Fst-I bear the same α-L-rhamnopyranosyl-(1→6)-β-D-glucopyranoside unit, but the position of the caffeoyl group is different for the two compounds [Figure 2]. In contrast, Fst-J has a β-D-xylopyranosyl-(1→6)-β-D-glucopyranoside unit. It is worth noting here that these compounds Fst-E,-I and-H can be generated in biological milieu by isomerization and/or degradation of Fst-A.[162] Fst-K (7R-suspensaside methyl ether) was isolated in 2016 from an ethanolic extract of F. suspensa.[163] As indicated above, Fst-L could not be found.

The isolation of Fst-M,-N,-O and-P was reported 2017.[164] Starting from 80 kg of the dried fruits of F. suspensa, the authors isolated about 5 mg of each compound. Fst-M,-N and-O possess a single β-D-glucose unit differently substituted. A 4-hydroxyphenylethanol group is common to Fst-M and-N, whereas Fst-O has a 2,5-dihydroxyphenylethanol group connected to the C2-position of the sugar through an ether linkage. On the C6-osition, an ester linkage connects the sugar to 4-hydroxyl-3-methoxylbenzoyl group for Fst-M, whereas Fst-O has a classical feruloyl unit. Fst-P bears a disaccharide unit α-L-rhamnose-(1→6)-β-D-glucose.[164]

Other phenylpropanoid glycosides and lignan glycosides have been isolated from F. suspensa, such as the forsythialensides A-D,[165] forsythenethosides A and B,[166] forsythoneosides,[167] forsythiaside and suspensaside,[168],[169],[170],[171] forsyphensides A-C,[172] susaroysides A-E,[173] suspensanosides A-C,[174] Lianqiaoxinsides A and B,[175],[176] forsythensides,[165],[177] and other phenylethanoid and lignan glycosides.[178],[179],[180]


  Pharmacological Properties of the Forsythosides Top


Anticancer activities

Different types of Forsythia Fructus extracts have revealed anticancer properties. An aqueous extract was shown to reduce markedly the growth of B16-F10 murine melanoma cells both in vitro and in vivo. The effects were attributed to the anti-inflammatory and anti-oxidative activities of the extract, leading to inhibition of tumor cell proliferation and angiogenesis.[181] This extract upregulated the expression of several proteins, notably the antioxidant proteins Nuclear factor erythroid 2-related factor 2 (Nrf-2) and heme oxygenase-1 (HO-1), the tumor suppressor proteins P53, and phospho-PTEN (phosphatase and tensin homolog deleted on chromosome 1) in the tumor tissue.[181] A parallel study indicated that the antimelanoma effect was linked to a modulation of glycerophospholipid metabolism in cancer cells induced by the Forsythia Fructus extract.[182] The mechanism is likely even more complex because the same extract was also found to activate different anti-inflammatory factors but also to increase the expression of the ubiquitin-regulator protein A20 which inhibits signaling cascades of endotoxin or cytokines in RAW 264.7 murine macrophage cells.[183] Whatever the molecular mechanism, it is interesting to note that water extracts made from the green fruits of F. suspensa displayed much more pronounced anti-melanoma effects in vitro and in vivo than extracts made from the ripe Forsythia fruits. Importantly, the Fst content (Fst-A,-E,-H,-I and-J were detected) was much higher in the green Forsythia extracts compared to the ripe Forsythia extracts.[88],[184] The authors showed that Fst-A,-E and-I (more abundant in the green fruits) were among the cytotoxic components of the aqueous extract and could serve as molecular markers to analyze the quality of extracts and their anticancer potential.[185] However, the anticancer activity of F. Fructus is not solely due to the presence of Fst compounds in the extract; other natural products also contribute to the activity such as betulinic acid and arctigenin.[186],[187]

A marked anticancer activity has been reported also with extracts of the fruits and leaves of the plant F. koreana, which showed antimetastatic effects by blocking invasion of breast cancer cells. The plant extract inhibited osteoclast formation and osteoclast-mediated bone resorption by reducing the activities of matrix metalloproteinases and cathepsin K.[188] As mentioned above, the three species F. suspensa, F. viridissima, and F. koreana, used as herbal medicines in Asia, possess common genes associated with the biosynthesis of acteoside and Fst-A (33), but the antitumor activity of the plant extracts could derive also in part from the presence of anticancer dammarane-type terpenoids.[189],[190]

Antiproliferative and anti-inflammatory activities have been observed with nonpolar fractions of F. fructus, suggesting that components other than the Fst can contribute to the global effects of fruit extracts.[191],[192] A marked antitumor activity was observed in vivo in a TE-13 esophageal cancer cell xenograft murine model, using an ethanolic root extract of F. suspensa.[193] In a study comparing the anticancer activity of 130 crude extracts used in Kampo medicine, an extract of Rengyo (Forsythia fruit) was found among the most potent extracts at regulating autophagic cell death in HepG2 hepatocellular carcinoma cells.[194] Fst-A and-B contribute to the anticancer activity, together with other natural products contained in the extract, notably tannic acid which displays a potent antiangiogenic activity through inhibition of the SDF-1/CXCR4 pathway.[195],[196]

In brief, Forsythia Fructus extracts display anticancer activities and several forsythosides (Fst-A/E/H/I/J) likely contribute to the anticancer activity, most likely through anti-inflammatory and antioxidative effects [Figure 3]. Other natural products included in the extract also contribute the antitumor action.
Figure 3: Antitumor activity of Forsythia Fructus extracts. Ethanolic extracts made from the fruits of Forsythia species contain numerous natural products, including diverse flavonoids, alkaloids, terpenoids, and phenylethanoid glycosides such as the forsythosides. The extracts inhibit cancer cell proliferation, invasion, and tumor angiogenesis, leading to the observed anticancer effects. The forsythosides contribute to the antitumor action through their antioxidant and anti-inflammatory activities

Click here to view


Antiviral activities

A large number of patents (about 400) cover the extraction, purification, characterization, formulations of Fst natural products from plants and their various medicinal uses. Several patents are focused on the antiviral activities of the compounds [Table 3]. The antiviral activity of Fst-A was first demonstrated using the avian infectious bronchitis virus (IBV) infecting primary chicken embryo kidney cells. Fst-A showed a direct virucidal effect, reducing cell infection by the IBV in vitro, but had no effect on IBV-infected cells.[197] A recent study has confirmed that Fst-A can markedly reduce viral infection in chicken exposed to the avian IBV IBV-M41. The compound was also efficient for the treatment of IBV-M41-infected chicken, through an improvement of CD3+, CD4+, and CD8+ T-lymphocytes in infected chickens and a sustained production of interleukin-2 (IL-2) and interferon-α (IFN-α).[198]
Table 3: Patents related to the antiviral activities of forsythosides*

Click here to view


Fst-A has shown activities against different viruses including influenza viruses which infect the respiratory epithelium in human and many vertebrates. However, the bioavailability of Fst-A is very limited, even when the compound is included in traditional medicines such as the TCM antiviral preparation Shuang-Huang-Lian used to treat various bacterial, fungal, and viral infections [Table 2].[199],[200],[201] All major PhGs, including compounds such as acteoside and salidroside, show a poor oral bioavailability.[202] However, the bioavailability of Fst-A and its antiviral activity can be enhanced through the use of absorption enhancers, such as water-soluble chitosan, that contributes to limit the influenza virus propagation.[203] Chito-oligosaccharides have been shown to facilitate the intestinal absorption of various PhGs in F. fructus extracts.[204] Fst-B also contributes to the anti-influenza A virus efficacy of Shuang-Huang-Lian recipes.[205] In recent years, the molecular mechanism of action against the influenza virus has been investigated. Fst-A was shown to control influenza A virus (IAV, H1N1) infection by inhibiting the virus replication and downregulating different pro-viral factors such as TLR7 (Toll-like receptor 7), MyD88 (myeloid differentiating factor 88), and nuclear factor kappa-B (NFκB) p65 mRNA.[206] In particular, the down-regulation of the TLR7 signaling pathway induced by Fst-A is key to control IAV infection[207] and the parallel blockade of the TLR4/NFκB axis leads to an essential immunomodulatory action [Figure 4].[208] Fst-A downregulates the expression of the influenza matrix protein 1 (M1), one of the most abundant protein in influenza virions,[125] and the compound also reduces the inflammatory response caused by influenza A virus (FM1 strain) in mouse lungs by affecting the retinoic acid-inducible gene-I-like receptors signaling pathway.[209] Fst-B also inhibits the TLR4/NFκB axis, contributing to an anti-inflammatory action.[210] The different TCM preparations containing Fst-A and-B thus benefit from the simultaneous attenuation of inflammation and reduction of viral replication in the infected lungs.[211]
Figure 4: Scheme summarizing the activity of forsythoside-A/B against the Influenza A virus. The compounds inhibit virus-induced inflammatory pathways and the replication of the virus through the indicated signaling route, leading to downregulation of several proteins implicated in virus infection

Click here to view


The antiviral activity of Fst-A and-B is not limited to the influenza virus. Fst-A protects peripheral blood mononuclear cells (PBMC) from infection by the bovine viral diarrhea virus (BVDV, a virus at the origin of viral diarrhea and mucosal disease in cattle). The compound inhibits the replication of BVDV, reduces apoptosis of PBMC (through a drug-induced over-secretion of IFN-γ and enhanced secretion of IL-2), and promotes activation of T cells.[211] Moreover, Fst-A promoted proliferation of bovine PBMCs and T cells activation, thereby counteracting the BVDV-induced overproduction of IFN-γ to maintain immune homeostasis.[211]

There is little scientific evidence today that the Fst per se can be active against the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) responsible for the current COVID-19 pandemic. However, several TCM preparations containing Fst compounds have revealed beneficial activities against the disease, either to reduce the viral charge or to alleviate the associated respiratory symptoms. Preparation like Shuang-Huang-Lian oral liquid and Tanreqing injection, alone or combined with Western medicine, are claimed to be active against severe pneumonia.[114],[137],[120],[201],[212] Other TCM preparations have revealed activities against COVID-19, such as those indicated in [Figure 5]. The contribution of the Fst to these effects is unclear at present, but preliminary information suggests that they play a role in the antiviral activity, through their anti-inflammatory action. Notably, the herbal product Lianhuaqingwen (LHQW) capsule can contribute to accelerate the recovery of patients with moderate COVID-19.[132] The treatment helps to reduce fever, fatigue, and coughing in COVID-19 patients and to improve other clinical symptoms such as diarrhea in patients with ordinary or mild COVID-19.[129],[131],[132] This phytochemical preparation is known to reduce the proliferation of influenza viruses, through the suppression of NF-B activation and reduction of pro-inflammatory cytokines production (IL-6, IL-8, TNFα, and MCP-1),[213] and it showed the same effect when using SARS-CoV-2-infected Vero E6 cells in vitro.[129] The anti-inflammatory action of Lianhuaqingwen is well established, but the preparation is complex (11 herbal constituents) and more than 60 active compounds have been identified.[214] Among them, 15 effective components have been characterized for their contribution to the anti-inflammatory and antiviral effects, and Fst-A belongs to this group of active compounds (as well as two compounds called “isomers of Fst-A” but not specified).[214] In a recent study, Fst-A,-B and-I were all three identified in Lianhuaqingwen capsules and found to bind to and to inhibit the angiotensin-converting enzyme-2 (ACE2), whereas Fst-E showed little or no effect.[102] Their affinities for the enzyme are modest (KD = 15.8, 61.0, and 18.7 μM for Fst-A,-B and-I, respectively, compared to prunasin and glycyrrhizin, KD = 0.27 and 4.4 μM), but the Fst-compounds could form stable complexes with the enzyme, according to a modeling analysis.[102] This binding is important because the virus SARS-CoV-2 utilizes the ACE2 receptor system for cell entry.[215] This multifunctional transmembrane protein – an essential component of the renin–angiotensin system – is overexpressed in human airway epithelial cells.[216] Drugs which could interfere with the interaction of ACE2 with the SARS-CoV-2 spike protein can be useful to combat COVID-19.[217],[218] Fst-A and its isomer Fst-I could thus play a marked role in the activity of the TCM preparations aforementioned.
Figure 5: Several forsythoside-containing traditional Chinese medicinal preparations used to combat Influenza (KBD, YQS, SHL, and T) or severe acute respiratory syndrome coronavirus-2 (SHL, T, SFJDC, JQG, QBD, and LHQW) viral infections. The plant and forsythoside-content of these traditional Chinese medicinal is indicated in Table 2

Click here to view


Perhaps, another indirect action of Fst against the virus is through the metabolites caffeic acid and hydroxytyrosol which can both inhibit coronavirus in humans.[219],[220],[221] On the basis of molecular modeling studies, it has been proposed that caffeic acid binds to the cell-surface heat shock protein A5 (HSPA5) so as to compete for recognition by the viral spike protein [Figure 6]. By this mechanism, caffeic acid could inhibit the cell attachment of the virus.[222] Different caffeic acid derivatives have been proposed as anti-COVID-19 agents.[223],[224] A spray containing hydroxytyrosol combined with α-cyclodextrin (called “Endovir Stop”) is being evaluated in COVID-19 patients, after the recent demonstration of its safety on healthy volunteers.[225] Preliminary clinical data indicated that this spray could improve defenses against SARS-CoV-2.[226] Therefore, the two aglycone constituents of Fst-A/B could also potentially contribute to the antiviral activity. They are also metabolites of the main PhG verbascoside which is viewed as an inhibitor of the SARS-CoV-2 main protease Mpro (also known as 3C-like proteinase, 3CLpro).[227]
Figure 6: Phenylethanoid glycosides and COVID-19. Most forsythosides are built with a variable glycosidic core substituted with a caffeoyl unit and a hydroxytyrosol unit. The forsythoside natural products and their metabolites are believed to contribute to the antiviral activity. Molecular modeling studies have suggested that forsythoside-B interact with the angiotensin-converting enzyme-2 implicated in the cell entry of the severe acute respiratory syndrome coronavirus-2 virus, whereas caffeic acid and hydroxytyrosol would bind to the cell-surface protein heat shock protein A5 implicated in the interaction with the viral spike protein

Click here to view


Antioxidative and anti-inflammatory activities

Recently, the anti-inflammatory activity of Fst-B isolated from the TCM herb Callicarpa kwangtungensis was thoroughly investigated.[15] Fst-B showed the strongest anti-inflammatory activity among six PhGs tested. The compound inhibited the release of TNF-α, IL-6, and nitric oxide in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages [Figure 5]. Fst-B was found to upregulate the expression of HO-1 and quinone oxidoreductase 1 (NQO1), through an inhibition of the interaction between the nuclear transcription factor Nrf2 and its repressor protein Kelch-like ECH-association protein 1 (Keap1). Fst-B also induced the accumulation of Nrf2 in cell nuclei. A potential direct binding of Fst-B to Keap1 was proposed, based on a molecular modeling analysis[15] and a similar physical interaction with Keap1 has been proposed for other structurally related PhGs such as acteoside and echinacoside.[228] Fst-A also activates Nrf2 and reduces endoplasmic reticulum stress. This effect has been evidenced in a model of cerebral ischemic injury induced by middle cerebral artery occlusion in rats. In this case, Fst-A markedly increased the expression levels of Nrf2, superoxide dismutase, and glutathione and reduced malonaldehyde expression, leading to a drug-induced reduction of cell apoptosis.[229] It has been demonstrated also in a model of hydrogen peroxide-induced oxidative stress in PC12 cells.[230]

The regulation of the Nrf2/HO-1 pathway is thus at the origin of the anti-inflammatory action of both Fst-A and Fst-B. This mechanism of action is shared with several other PhGs, such as acteoside, alyssonoside, paraboside B, and others.[231],[232],[233],[234] Fst-B was found to behave as a strong activator of Nrf2 and inducer of HO-1 expression in human keratinocyte cells, providing a long-lasting skin protection.[235] The compound is a potent inhibitor of gene expression and de novo synthesis of all chemokines.[236] It is important to underline that the anti-inflammation action of Fst-B has been well characterized both in vitro but in vivo as well, in a mouse model of acute lung injury. Fst-B was shown to markedly inhibit activation of the TLR4/NFκB signaling pathway in this model, thus reducing the infiltration of inflammatory cell in the inflamed lung.[210] Another good demonstration of the antioxidant activity of Fst-B refers to the capacity of the product to scavenge reactive oxygen species (ROS) liberated by polymorphonuclear neutrophils during inflammatory disorders or when stimulated by chemicals.[237] Other studies have reaffirmed the anti-inflammatory role of Fst-A or-B through an activation of Nrf2 and inhibition of NFκB.[238] This is really the central action of the natural products [Figure 7].
Figure 7: Antioxidant and anti-inflammatory effects of forsythoside-A,-B. (a) Upon entering macrophages, the compounds have the capacity to inhibit the interaction between the transcription factor Nrf2 and its repressor Keap1, through a selective binding to Keap1. Nrf2 thus released is phosphorylated and can accumulate into cell nuclei to activate transcription of specific genes, through its interaction with antioxidant response elements. The production of several mRNA is enhanced (HO-1, NQO1, and Nrf2) or repressed (COX-2, TNFα, and iNOS) leading to a reduced production of pro-inflammatory cytokines and enzymes which act by reducing the cell oxidative stress and free radicals. (b) The proposed interaction of forsythoside-B with Keap1[15]

Click here to view


The anti-inflammatory and antioxidant actions occur in parallel, in a cooperative manner. Fst-A and B are potent antioxidant compounds, showing marked free-radical-scavenging effects.[60],[239] The apiosyl moiety of Fst-B seems to improve the radical scavenging property of the compound because the analog acteoside (vervascoside) lacking the apiosyl unit showed a lower antioxidative capacity compared to Fst-B.[240] In fact, the antioxidant effect and drug-induced regulation of the Nrf2/HO-1 pathway lead to a general cytoprotective effect which can be evidenced at different levels and in different tissues, in particular to protect the cardiovascular and neuronal systems, as evoked hereafter, but also to protect from allergic airway inflammation and asthma.[241] Both Fst-A and Fst-B can reduce the oxidative damages and inflammation induced by CuSO4 in zebrafish larvae, through the modulation of multiple metabolomic pathways.[242]

Neuroprotection

Interestingly, an aqueous extract of F. suspensa not only exhibited direct anticancer effects, but it contributed also to reduce the unwanted side effects of chemotherapy. The extract was found to reduce oxaliplatin-induced peripheral neuropathy, without affecting the anticancer activity of the platin drug in human cancer cells. The effect was attributed, at least in part, to Fst-A abundantly present in the extract and which has shown neuroprotective effects in vivo.[243] An aqueous extract of F. viridissima has revealed similar neuroprotective effects on oxaliplatin-induced peripheral neuropathy[244] and Fst-A was found to reduce side effects induced by cisplatin in the cochlea of guinea pig.[245] In fact, the drug can reduce neuronal damages and inflammation induced by a variety of stimuli. In another study using organotypic hippocampal slices, Fst-A and an ethanol extract of F. suspensa leaves have shown neuroprotective effects, associated to an upregulation of the endogenous endocannabinoid 2-arachidonoylglycerol and a marked inhibition of cyclooxygenase-2 (COX-2). Moreover, a potential direct interaction between Fst-A and COX-2 enzyme was postulated, based on molecular modeling.[246]

A neuroprotective effect has been evidenced with Fst-B in a model of cerebral ischemia and reperfusion injury. The compound attenuated the blood–brain barrier breakdown and reduced inflammation.[247] Fst-B is a potent agent to reduce neuro-inflammation and to attenuate memory-impairment, suggesting a beneficial action of the molecule (and TCM preparations containing Fst-B) for the treatment of Alzheimer disease.[248] Indeed, Fst-B was found to restore cognitive function in neurodeficient (APP/PS1) mice, through an improvement of amyloid-β (Aβ25–35) peptide deposition and tau protein phosphorylation. It also reduced microglia-mediated neurotoxicity in BV-2 microglial cells and HT22 cells. Similar anti-inflammatory effects in LPS-stimulated BV-2 microglia cells have been reported with Fst-A.[249] The anti-neuroinflammatory action of Fst-B could be useful to limit the progression of the Alzheimer disease.[248] Fst-B inhibits glutamate-induced neurotoxicity.[12] Similarly, Fst-A exhibits protective effects on Aβ-induced apoptosis in PC12 cells by downregulating acetylcholinesterase.[250] The compound can help to improve memory deficit. This property has been demonstrated using a murine model of age-dependent neurodegenerative disorders (senescence-accelerated mouse prone, or SAMP mice). An oral treatment of 8-month-old (SAMP8) mice for 45 days was found to improve the cognitive functions, and the effect was associated with the drug-induced suppression of oxidative stress and decreased production of inflammatory cytokines. The study suggested that Fst-A may be a useful treatment against amnesia.[251] Different structurally related phenylpropanoids, such as tenuifoliside A and arctigenin, have been found to exert similar cognitive enhancing property and to improve memory deficit in mice.[252],[253] It is likely the caffeic acid moiety of Fst-A which is responsible for this effect because this unit (and related units such as p-methoxycinnamic acid, ethyl-p-methoxycinnamate and ferulic acid [4-hydroxy-3-methoxycinnamic acid]), has been shown to improve memory deficit in rats.[254],[255],[256] Caffeic acid esters (widely found in propolis extract and plants) are known for their neurogenesis and neuroprotection function.[257],[258] Moreover, ferulic acid derivatives are being designed as multitarget-directed ligands for the treatment of Alzheimer's Disease.[259] Fst-A/B fall into this category and their mechanism of action is complex, as it implicates also a drug-induced upregulation of the levels of the endogenous endocannabinoid 2-arachidonoylglycerol (2-AG), mediated through the cannabinoid receptor 1 (CB1R)-dependent NFκB signaling pathway.[260] This mechanism, as well as the reported binding of Fst-A to acetylcholinesterase, could contribute to reduce the evolution of the Alzheimer disease.[240],[261]

Cardioprotection

The capacity of Fst-A and-B to reduce tissue inflammation has been evidenced in different pathological situations and with different experimental models. In particular, Fst-B was found to rescue cardiac function from ischemia-reperfusion injury by limiting inflammation response and antioxidative damages in a rat model.[262] Mechanistically, Fst-B is able to limit the secretion of the potent vasoconstrictor peptide endothelin-1 in endothelial cells treated with chemically oxidized low-density lipoproteins (LDLs). Given the important role of endothelin-1 in atherosclerosis development, the use of phyto-preparations containing Fst-B and-J (and related inhibitors of lipid peroxidation) could be useful to prevent or treat atherosclerosis development.[263],[264],[265] Phenylpropanoid glycosides are strong inhibitors of copper-induced LDL oxidation.[254] At this point, it is worth mentioning that a molecular modeling study has suggested that Fst-C, which is not frequently studied, would be able to interact with the enzyme squalene synthase, thereby reducing cholesterol synthesis.[266] However, the proposed lipid-lowering effect of Fst-C has not been validated experimentally; it remains a hypothesis at this stage.

Hepatoprotection

The accumulation of cytotoxic bile acids can lead to liver damages and ultimately to serious liver cirrhosis or liver failure. The TCM prescription Dan-He-Ji (LDHJ), which contains Fst-A, has been developed to treat infantile cholestatic hepatopathy. The phyto-preparation antagonizes calcium-sensing receptor and regulates hepatocyte apoptosis in cholestasis through the mitogen-activated protein kinase pathway. As an active substance of LDHJ, Fst-A contributes to reduce the levels of calcium and ROS and thus the level of apoptosis in cholestatic hepatocytes.[101] A marked hepatoprotective effect has been reported also when using water extract of Forsythia Fructus to protect from liver fibrosis in mice. The plant extract protected against CCl4-induced liver injury by inhibiting hepatic stellate cells activation, reducing hepatic extracellular matrix disposition, and reversing epithelial–mesenchymal transition.[267] The effects can be attributed to different compounds included in the extract, notably the lignan pinoresinol,[268] forsythin, and Fst-A.[256] Fst-A has a direct effect of drug-induced liver injury by inhibiting toxin-induced NFκB activation and serum/hepatic TNFα levels in a mice model of acute liver injury.[269]

Fst-A/B exhibits protective effects against damages to the central nervous system, the heart, the liver, and other organs, such as the kidneys. Indeed, Fst-A was shown to alleviate renal damage in adriamycin-induced nephropathy in rats,[270] and a F. suspensa extract can protect from kidney damages induced by diquat, a commonly used pesticide.[230] As cellular protective agents, Fst-A/B can prevent cellular damages and cell death in many different situations, including in cases of alopecia, reducing apoptosis of hair cells, and retarding their entry into the catagen phase.[271]

Protection from other inflammatory diseases and damages

The anti-inflammatory action of Fst-A and Fst-B can be useful to prevent or reduce other diseases with an inflammatory component such as peritonitis, sepsis, asthma, and others [Figure 8]. Fst-A was found to regulate the production of inflammatory cytokines and chemokine in zymosan-stimulated RAW 264.7 macrophages in vitro. In vivo, the compound decreased the number of neutrophils and reduced the level of TNFα, IL-6, and the monocyte chemoattractant protein-1 (MCP-1, also called CCL-2) in the peritoneal cavity, in a mouse model of zymosan-induced acute peritonitis.[272] Fst-A-induced inhibition of the production of MCP-1 was observed in LPS-stimulated alveolar epithelial MLE-12 cells. In this cell model, Fst-A decreased the adhesion and migration of monocytes to MLE-12 cells, through an upregulation of the microRNA miR-124, and a subsequent inhibition of the expression of MCP-1.[273],[274] However, the mechanism of action of Fst-A is likely multifactorial, implicating several targets. The compound was shown to inhibit the secretion of IL-8 and prostaglandin E2, and calcium influx in the cells expressing the Transient receptor potential vanilloid 1 (TRPV1) channel implicated in the thermoregulation, in a model of yeast-induced pyrexia mice.[275]
Figure 8: The antioxidant and anti-inflammatory activities of forsythoside-A and forsythoside-B can be useful to combat a variety of diseases and conditions. The two natural products reduce oxidative stress and the inflammatory component of these diseases

Click here to view


As mentioned above, the anti-endotoxin effects of Fst-A seem to rely, at least in part, on the capacity of the compound to inhibit the TLR4/MyD88/NFκB signaling pathway.[208] Similarly, Fst-B reduced the production levels of TNFα, IL-6, and HMGB1 in RAW 264.7 macrophages and markedly reduced the lethality in a rat mode of sepsis, induced by cecal ligation and puncture and also in a mouse model of sepsis.[210],[276] The anti-inflammatory action of Fst-A can also be useful for the treatment of bovine mastitis, generally caused by the Gram-positive bacterial pathogen Staphylococcus aureus. Fst-A was found to down-regulate the expressions of TNFα, IL-1 β, and IL-6 and to suppress NFκB and MAPKs activation in S. aureus-stimulated primary bovine mammary epithelial cells.[277]

Interestingly, a recent study has highlighted the activity of an extract of the plant Callicarpa japonica against allergic airway inflammation. The extract contained several phenylpropanoids (in particular verbascoside and Fst-B) and showed a protective activity against ovalbumin-induced airway inflammation, through downregulation of NFκB activation and upregulation of HO-1 (20). Fst-B can be useful to treat allergic inflammation and other skin inflammatory diseases such as prurit and dermatitis. Through its capacity to inhibit the production of chemokines in TNFα/IFNγ-activated human keratinocytes, Fst-A can be considered also for treating allergic skin inflammatory disorders.[278] Again recently, the two compounds verbascoside and Fst-B were found to function as selective inhibitors of the temperature-sensitive and calcium-permeable transient receptor potential vanilloid 3 (TRPV3), which is frequently overactive in chronic pruritus, skin allergy, and other inflammation-related skin diseases.[279],[280] Fst-B dose dependently inhibited human TRPV3 (but not hTRPV1, hTRPV4, and hTRPA1) and reduced cell death of HaCaT keratinocytes expressing a gain-of-function TRPV3 G573S mutant. The compound may bind to TRPV3 and could block the channel, as a putative binding pocket for Fst-B has been identified in the central cavity of mouse TRPV3, based on a molecular modeling analysis. By blocking the channel, the compound could restrict the entry of calcium and calcium overload in cells, responsible for keratinocyte cell death and keratinization, itching, or skin damages.[279] However, the link between this mechanism and the capacity of Fst-B to inhibit the production of pro-inflammatory chemokines at transcriptional and translational levels in keratinocytes remain to be determined.

Antibacterial activity

F. suspensa extracts (FSE) exhibit bacteriostatic activity against Escherichia coli K88, staphylococcus aureus, and salmonella strains. The use of such an extract as a substitute for antibiotic has been proposed, to enhance the antioxidant status and anti-inflammatory function in broiler chicken.[281],[282] FSE is also proposed as a dietary supplementation in sows and newborn piglets.[283] Relatively modest antibacterial effects were initially reported with different Fst compounds, but the bactericidal activities were often relatively weak. However, Fst-B has shown a noticeable activity against different strains of Staphylococcus aureus, notably against the multiple drug-resistant strain S. aureus 1199B (which overexpresses the norA gene encoding a multidrug-resistant efflux pump).[61] The antibacterial effect is more pronounced when using the crude extract of the fruits from F. suspensa, in particular the raw fruits (qingqiao) which contain a higher level of PhGs than the ripe fruits (laoqiao) and display more pronounced antibacterial effects against different bacterial strains.[88] As regard Fst-A, the combination of a bacteriostatic effect and an anti-inflammatory effect can be useful to prevent or treat periprosthetic infection. Fst-A can reduce Staphylococcus aureus and methicillin-resistant S. aureus adhesion and biofilm formation on the surface of titanium alloy (a recurrent problem encountered with orthopedic joint prostheses). Fst-A reduces bacterial adhesion and colony formation on the surface of the titanium disc and attenuates Ti-induced activation of NFκB and cytokines production in macrophages.[284] More globally, Fst-A reduces the inflammatory response stimulated on infection with S. aureus.[277]


  Metabolism of Forsythosides-A Top


Upon oral administration, the intestinal absorption of the Fst-A mainly occurs by passive diffusion across the different segments of the intestine, notably in the upper part of small intestine, at least in rat.[285],[286],[287] The intestinal absorption of Fst-A is limited but the other constituents of Forsythia fructus help to increase the bioavailability of the natural product, as well as the use of a hydrogel delivery system.[288],[289] In rat liver microsomes, Fst-A provides a good substrate for many cytochromes P450 (CYP3A4, CYP2C9, CYP1A2, UGT1A6, UGT1A3, UGT1A1, and UGT1A9) and has an inductive effect on the activities of CYP1A2 and CYP2C11.[290],[291] The compound is a substrate for different transporters such as the efflux pumps P-glycoprotein (P-gp) and the multidrug-resistance-protein, and uptake transporters (OATP), thus explaining the low oral bioavailability of the product.[203],[204],[292],[293] The low intrinsic bioavailability of Fst-A is largely promoted by other natural products found in the different plant extracts which are combined in the TCM preparations. In particular, the combined use of Flos Lonicerae and Forsythia Fructus (two herbs included in different TCM preparations such as Shuang-Huang-Lian tablet, Yin-Qiao-Jie-Du tablet, and Fufang Qin-Lan oral liquid) is beneficial to improve the absorption of Fst-A.[294],[295]

After an oral administration, Fst-A is largely metabolized. In rats, up to 43 metabolites have been identified in biological samples, mostly in the urine (42 metabolites), plasma (22 metabolites), and feces (42 metabolites). The numerous drug transformations include combination of mono-and di-methylation, mono- and di-glucuronidation, sulfation, and cysteine conjugation.[296] The intestinal metabolites of Fst-A in Human include the two products caffeic acid and hydroxytyrosol formed upon hydrolytic deglycosylation of the natural product, presumably by human intestinal bacteria.[297] In dogs, Fst-A showed a linear pharmacokinetic profile; after intravenous administration of Fst-A, the highest Cmax value reached 54.5 μg/mL, but the drug is also rapidly eliminated, with a T1/2 value >1.5 h in dogs.[298] The toxicology data available for Fst-A and-B are limited. A more precise evaluation of the benefit/risk ratio of Fst-A would be useful.


  Discussion Top


PhGs are naturally occurring water-soluble compounds, widely distributed in plants. They are endowed with remarkable biological properties.[299],[300],[301],[302] A recent exhaustive survey has identified more than 570 PhGs which display a wide range of pharmacological properties: antibacterial, anticancer, antidiabetic, anti-inflammatory, anti-obesity, antioxidant, antiviral, and neuroprotective properties.[233] The best-known member of the PhGs are arguably acteoside/verbascoside (>1000 references in PubMed Central), echinacoside (250 references), more frequently studied than the Fst (140 references). Nevertheless, the Fst now emerge as a group of natural products with a marked medicinal interest. These compounds are well represented in many traditional Chinese medicine preparations, as reported here. The Fst subgroup includes 16 members (Fst-A-to-P) but one of them (Fst-L) could not be identified, despite an extensive search through the literature, with several search engines. Several of these compounds, in particular the most recent ones, have been isolated and chemically described only. Their structures are known but not their biological properties. On the opposite, Fst-A and-B have been extensively studied over the past 40 years and they clearly emerge as potent cell-protective natural products. They are both potent anti-inflammatory and antioxidant agents, acting mainly through a drug-induced activation of the Nrf2/HO-1 pathway in different cell types, possibly through the sequestration of the Nrf2-suppressor Keap1. This property is shared with other natural products and phenolic compounds qualified as direct or indirect Nrf2 activators, protecting cells from oxidative stress. Upon binding of Fst-B to Keap1, Nrf2 is liberated from the Nrf2-Keap1 complex, translocated into the nucleus, and bound to the antioxidant response element in association with other factors, resulting in the expression of detoxifying enzymes, such as NQO1 and HO-1 [Figure 7]. The mechanism of action of Fst-B is apparently complex and multifactorial, but inflammation is also a multifaceted biological system. Medicinal plants and the natural products that they contain are well suited to reduce inflammation.[303]

Fst-A/B has the capacity to block the expression and/or production of pro-inflammatory cytokines. It is therefore not totally surprising to observe that the two compounds have been proposed as remedies for a large number of inflammatory situations and pathologies [Figure 8]. Inflammation is a general trait associated with many diseases. The molecular pharmacology of inflammation implicates multiple biological targets and mechanisms, depending on the cellular context and the organ considered. For a long time, medicinal plants have been exploited to treat inflammatory symptoms and to reduce associated damages. In this context, the extensive use of the plant F. suspensa (Thunberg) Vahl. is exemplary. The plant and its fruits Forsythia fructus contain numerous anti-inflammatory agents, notably different PhGs such as Fst-A/B which are abundant in the green fruits (laoqiao). Most of the biological and therapeutic effects reported with Fst-A/B derive from their anti-inflammatory and antioxidant effects. This is the foundation of their medicinal activities.

Could Fst-A/B be exploited for the design of new drugs? Probably not the compounds sensu stricto, because they are relatively unstable biologically, with a short half-life in biological media and they show a low bioavailability, at least for Fst-A (pharmacokinetic data are more limited for Fst-B). Designing drugs based on the skeleton of Fst-A would be difficult. Nevertheless, the two compounds can be useful when combined with other products, in the form of plant extracts (confere the antitumor activities reported with different forsythia extracts) and medicinal phyto-preparations. It is interesting to underline the diversity of TCM preparations including F. suspensa or Forsythia fructus. They are used to treat a variety of diseases, mostly some gastrointestinal diseases and lung diseases. Interestingly, several of the Fst-containing TCM exhibit antiviral activities and are being evaluated clinically for the treatment of the current COVID-19 pandemic. As discussed above, the anti-inflammatory and antioxidative activities of compounds such as Fst-A,-B and-I likely contribute positively to the antiviral effects, to reduce the viral charge and the pulmonary symptoms. Different TCM preparations look promising against the symptoms of COVID-19, such as Jinhua Qinggan granules, Qingwen Baidu decoction, Shufeng Jiedu capsules, and a few others [Figure 5]. Among the six TCM preparation containing Fst useful to combat COVID-19, the case of Shuanghuanglian Injection (SHLI) deserves a mention because cases of adverse reactions affecting the skin, mucosa, and/or the digestive system have been observed when using this preparation, with an incidence of about 5%.[304] A pseudo-allergic action of F. suspensa included in the preparation has been noted in different studies.[305],[306] The pseudo-allergic reaction apparently implicates a SHLI-induced reorganization of actin cytoskeleton, an activation of the RhoA/ROCK signaling pathway, and a disruption of the endothelial barrier.[307] A recent study indicated that the effect could be linked to the action of Fst-A and-B (but not Fst-E), which would disrupt the endothelial barrier, inducing an hyperpermeability and thus causing a vascular leakage (with activation the RhoA/ROCK signaling pathway).[113] For this reason, the injectable form of Shuanghuanglian (SHLI) should be used with caution, especially in children.[308] The oral liquid form should be preferred as it is also active against COVID-19 symptoms.[114],[309] The flavonoids baicalin and baicalein are considered as the main bioactive components of the Shuanghuanglian preparation,[114] but the potential beneficial contribution of Fst-A/B cannot be excluded.

Chinese herbal formulae represent an alternative approach to the prevention or treatment of COVID-19. TCMs containing Forsythia Fructus (Lianqiao) are among the most popular and the apparently the most beneficial, at least to alleviate the associated symptoms of the disease.[310] It clearly helps to reduce fever, cough, and fatigue.[311] The contribution of the Fst to these effects is unclear at present, but nevertheless, compounds like Fst-A,-B,-H, and-I stand as potent anti-inflammatory and antioxidant agents that can be useful for other inflammatory situations and diseases.

Acknowledgments

The author thanks Dr Chang Li (Department of Medicinal Chemistry and Natural Medicine Chemistry, College of Pharmacy, Harbin Medical University, Harbin, China) who kindly helped to identify the structure of Fst-K from a publication in Chinese.

Financial support and sponsorship

Nil.

Conflicts of interest

There are no conflicts of interest.



 
  References Top

1.
da Silva CC, Vandresen F, de Oliveira CMA, Kato L, Tanaka CMA, Ferreira HD. Chemical composition of Aloysia gratissima (Gill. et Hook) Tronc. (Verbenaceae). Biochem System Ecol. 2006; 34,593-595.  Back to cited text no. 1
    
2.
Marchetti L, Pellati F, Graziosi R, Brighenti V, Pinetti D, Bertelli D. Identification and determination of bioactive phenylpropanoid glycosides of Aloysia polystachya (Griseb. et Moldenke) by HPLC-MS. J Pharm Biomed Anal. 2019;166:364-370.   Back to cited text no. 2
    
3.
de Oliveira CMA, da Silva CC, Ferreira HD, Lemes GF, Schmitt E. Kauranes, phenylethanoids and flavone from Aloysia virgata. Biochem System Ecol. 2005;33,1191-1193.  Back to cited text no. 3
    
4.
Tóth E, Tóth G, Máthé I, Blunden G. Martynoside, forsythoside B, ladanein and 7a-acetoxyroyleanone from Ballota nigra L. Biochem System Ecol. 2007;35:894-897.  Back to cited text no. 4
    
5.
Vrchovská V, Spilková J, Valentão P, Sousa C, Andrade PB, Seabra RM. Antioxidative properties and phytochemical composition of Ballota nigra infusion. Food Chem. 2007;105:1396-1403.  Back to cited text no. 5
    
6.
Siciliano T, Bader A, Vassallo A, Braca A, Morelli I, Pizza C, De Tommasi N. Secondary metabolites from Ballota undulata (Lamiaceae). Biochem System Ecol. 2005;33:341-351.  Back to cited text no. 6
    
7.
Kanchanapoom T, Kasai R, Yamasaki K. Iridoid glucosides from Barleria lupulina. Phytochemistry. 2001;58:337-341.   Back to cited text no. 7
    
8.
Bankova V, Koeva-Todorovskab J, Stam bolijskab T, Ignatova-G rocevab MD, Todorova D, Popov S. Polyphenols in Stachys and Betonica Species (Lamiaceae). Z. Naturforsch. 1999;54:876-880.  Back to cited text no. 8
    
9.
Kong LD, Wolfender JL, Cheng CH, Hostettmann K, Tan RX. Xanthine oxidase inhibitors from Brandisia hancei. Planta Med. 1999;65:744-746.  Back to cited text no. 9
    
10.
Liao YH, Houghton PJ, Hoult JR. Novel and known constituents from Buddleja species and their activity against leukocyte eicosanoid generation. J Nat Prod. 1999;62:1241-1245.  Back to cited text no. 10
    
11.
Nicoletti M, Galeffi C, Multari G, Garbarino JA, Gambaro V. Polar Constituents of Calceolaria ascendens. Planta Med. 1988;54:347-348.   Back to cited text no. 11
    
12.
Koo KA, Sung SH, Park JH, Kim SH, Lee KY, Kim YC. In vitro neuroprotective activities of phenylethanoid glycosides from Callicarpa dichotoma. Planta Med. 2005;71:778-780.   Back to cited text no. 12
    
13.
Liu J, Gu Z, Yao S, Zhang Z, Chen B. Rapid analysis of Callicarpa L. using direct spray ionization mass spectrometry. J Pharm Biomed Anal. 2016;124:93-103.   Back to cited text no. 13
    
14.
Kim SM, Ryu HW, Kwon OK, Hwang D, Kim MG, Min JH, et al. Callicarpa japonica Thunb. ameliorates allergic airway inflammation by suppressing NF-κB activation and upregulating HO-1 expression. J Ethnopharmacol. 2021;267:113523.  Back to cited text no. 14
    
15.
Wu A, Yang Z, Huang Y, Yuan H, Lin C, Wang T, et al. Natural phenylethanoid glycosides isolated from Callicarpa kwangtungensis suppressed lipopolysaccharide-mediated inflammatory response via activating Keap1/Nrf2/HO-1 pathway in RAW 264.7 macrophages cell. J Ethnopharmacol. 2020;258:112857.   Back to cited text no. 15
    
16.
Hu X, Li L, Yang YF, Huang CY, Huang GL. [Caffeoyl phenylethanoid glycosides from Callicarpa kwangtungensis]. Zhongguo Zhong Yao Za Zhi. 2014;39:1630-1634.  Back to cited text no. 16
    
17.
Niu C, Li Q, Yang LP, Zhang ZZ, Zhang WK, Liu ZQ, et al. Phenylethanoid glycosides from Callicarpa macrophylla Vahl. Phytochemistry Lett. 2020;38:65-69  Back to cited text no. 17
    
18.
Ado MA, Abas F, Leong SW, Shaari K, Ismail IS, Ghazali HM, et al. Chemical constituents and biological activities of Callicarpa maingayi leaves. South Afric J Botany 2016;104:98-104  Back to cited text no. 18
    
19.
Ma YC, Zhang M, Xu WT, Feng SX, Lei M, Yi B. [Chemical constituents from Callicarpa nudiflora and their cytotoxic activities]. Zhongguo Zhong Yao Za Zhi. 2014;39:3094-3101.  Back to cited text no. 19
    
20.
Wu YS, Shi L, Liu XG, Li W, Wang R, Huang S, et al. Chemical profiling of Callicarpa nudiflora and its effective compounds identification by compound-target network analysis. J Pharm Biomed Anal. 2020;182:113110.   Back to cited text no. 20
    
21.
Wu AZ, Zhai YJ, Zhao ZX, Zhang CX, Lin CZ, Zhu CC. Phenylethanoid glycosides from the stems of Callicarpa peii (hemostatic drug). Fitoterapia. 2013;84:237-241.   Back to cited text no. 21
    
22.
Morikawa T, Pan Y, Ninomiya K, Imura K, Matsuda H, Yoshikawa M, et al. Acylated phenylethanoid oligoglycosides with hepatoprotective activity from the desert plant Cistanche tubulosa. Bioorg Med Chem. 2010;18:1882-1890.   Back to cited text no. 22
    
23.
Gong X, Wang J, Zhang M, Wang P, Wang C, Shi R, et al. Bioactivity, Compounds Isolated, Chemical Qualitative, and Quantitative Analysis of Cymbaria daurica Extracts. Front Pharmacol. 2020;11:48.   Back to cited text no. 23
    
24.
Delazar A, Gibbons S, Kumarasamy Y, Nahar L, Shoeb M, Sarker SD. Antioxidant phenylethanoid glycosides from the rhizomes of Eremostachys glabra (lamiaceae). Biochem. Syst. Ecol. 2005;33:87-90.  Back to cited text no. 24
    
25.
Delazar A, Sarker SD, Shoeb M, Kumarasamy Y, Nahar L, Nazemyieh H. Three antioxidant phenylethanoid glycosides from the rhizomes of Eremostachys pulvinaris (family: Labiatae). Iran J Pharm Res. 2005;3:23-24.  Back to cited text no. 25
    
26.
Ukaegbu CI, Shah SR, Hazrulrizawati AH, Alara OR. Acetone extract of Flammulina velutipes caps: A promising source of antioxidant and anticancer agents. Beni-Suef Univ J Basic Applied Sci. 2018;7:675-682.  Back to cited text no. 26
    
27.
Sun L, Rai A, Rai M, Nakamura M, Kawano N, Yoshimatsu K, et al. Comparative transcriptome analyses of three medicinal Forsythia species and prediction of candidate genes involved in secondary metabolisms. J Nat Med. 2018;72:867-881.   Back to cited text no. 27
    
28.
Wang Z, Xia Q, Liu X, Liu W, Huang W, Mei X, et al. Phytochemistry, pharmacology, quality control and future research of Forsythia suspensa (Thunb.) Vahl: A review. J Ethnopharmacol. 2018;210:318-339.   Back to cited text no. 28
    
29.
Endo K, Takahashi K. Constitutions of forsythosides F and G, new phenol glycosides of Forsythia viridissima stems. Heterocycles. 1990;30:291-294.  Back to cited text no. 29
    
30.
Es-Safi NE, Khlifi S, Kerhoas L, Kollmann A, El Abbouyi A, Ducrot PH. Antioxidant constituents of the aerial parts of Globularia alypum growing in Morocco. J Nat Prod. 2005;68:1293-1296.   Back to cited text no. 30
    
31.
Caliş I, Kirmizibekmez H, Sticher O. Iridoid glycosides from Globularia trichosantha. J Nat Prod. 2001;64:60-64.   Back to cited text no. 31
    
32.
Šliumpaitė I, Venskutonis PR, Murkovic M, Pukalskas A. Antioxidant properties and polyphenolics composition of common hedge hyssop (Gratiola officinalis L.). J Function Food. 2013;5:1927-1937.  Back to cited text no. 32
    
33.
Andary C, Tahrouch S, Marion C, Wylde R, Heitz A. Caffeic glycoside esters from Jasminum nudiflorum and some related species. Phytochemistry. 1992;31:885-886.   Back to cited text no. 33
    
34.
Zan K, Jiao XP, Guo LN, Zheng J, Ma SC. [HPLC specific chromatogram of Lamiophlomis Herba and its counterfeit and determination of four effective components]. Zhongguo Zhong Yao Za Zhi. 2016;41:2284-2290.   Back to cited text no. 34
    
35.
Yue HL, Zhao XH, Mei LJ, Shao Y. Separation and purification of five phenylpropanoid glycosides from Lamiophlomis rotata (Benth.) Kudo by a macroporous resin column combined with high-speed counter-current chromatography. J Sep Sci. 2013;36:3123-3129.   Back to cited text no. 35
    
36.
Wu L, Xu L, Li L, Wang JY, Ding N, Yu D, et al. [Diverse Solvent Effect on Extraction of Chemical Components from Lamiophlomis rotate]. Zhong Yao Cai. 2016;39:571-574.  Back to cited text no. 36
    
37.
Sena Filho JG, Nimmo SL, Xavier HS, Barbosa-Filho JM, Cichewicz RH. Phenylethanoid and lignan glycosides from polar extracts of Lantana, a genus of verbenaceous plants widely used in traditional herbal therapies. J Nat Prod. 2009;72:1344-1347.   Back to cited text no. 37
    
38.
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. 2020;193:113667.   Back to cited text no. 38
    
39.
Abe F, Nagao T, Okabe H. Antiproliferative constituents in plants 9. Aerial parts of Lippia dulcis and Lippia canescens. Biol Pharm Bull. 2002;25:920-922.   Back to cited text no. 39
    
40.
Sánchez-Marzo N, Lozano-Sánchez J, Cádiz-Gurrea ML, Herranz-López M, Micol V, Segura-Carretero A. Relationships Between Chemical Structure and Antioxidant Activity of Isolated Phytocompounds from Lemon Verbena. Antioxidants (Basel). 2019;8:324.   Back to cited text no. 40
    
41.
Cheng LC, Murugaiyah V, Chan KL. In vitro Xanthine Oxidase Inhibitory Studies of Lippia nodiflora and Isolated Flavonoids and Phenylethanoid Glycosides as Potential Uric Acid-lowering Agents. Nat Prod Commun. 2015;10:945-948.  Back to cited text no. 41
    
42.
Cheng LC, Murugaiyah V, Chan KL. Flavonoids and phenylethanoid glycosides from Lippia nodiflora as promising antihyperuricemic agents and elucidation of their mechanism of action. J Ethnopharmacol. 2015;176:485-493.   Back to cited text no. 42
    
43.
Martins GR, da Fonseca TS, Martínez-Fructuoso L, Simas RC, Silva FT, Salimena FRG, Alviano DS, Alviano CS, Leitão GG, Pereda-Miranda R, Leitão SG. Antifungal Phenylpropanoid Glycosides from Lippia rubella. J Nat Prod. 2019;82:566-572.   Back to cited text no. 43
    
44.
Funari CS, Passalacqua TG, Rinaldo D, Napolitano A, Festa M, Capasso A, et al. Interconverting flavanone glucosides and other phenolic compounds in Lippia salviaefolia Cham. Ethanol extracts. Phytochemistry. 2011;72:2052-2061.   Back to cited text no. 44
    
45.
Zhang FX, Li ZT, Li M, Yuan YL, Cui SS, Chen JX, et al. Dissection of the potential anti-influenza materials and mechanism of Lonicerae japonicae flos based on in vivo substances profiling and network pharmacology. J Pharm Biomed Anal. 2020;193:113721.   Back to cited text no. 45
    
46.
Zhang J, Yi P, Xiong Y, Du C, Zhang Y, Yuan C, Huang L, Gu W, Hao X. A new acorane sesquiterpenes of Lysionotus pauciflorus maxim. Form Guizhou province, China. Biochem System Ecol. 2020;93: 104165.  Back to cited text no. 46
    
47.
Xue Z, Lai C, Kang L, Kotani A, Hakamata H, Jing Z, et al. Profiling and isomer recognition of phenylethanoid glycosides from Magnolia officinalis based on diagnostic/holistic fragment ions analysis coupled with chemometrics. J Chromatogr A. 2020;1611:460583.   Back to cited text no. 47
    
48.
Çaliş I, Hosny M, Khalifa T, Rüedi P. Phenylpropanoid glycosides from Marrubium alysson. Phytochem. 1992;31:3624-3626.  Back to cited text no. 48
    
49.
Karioti A, Protopappa A, Megoulas N, Skaltsa H. Identification of tyrosinase inhibitors from Marrubium velutinum and Marrubium cylleneum. Bioorg Med Chem. 2007;15:2708-2714.   Back to cited text no. 49
    
50.
Zaabat N, Hay AE, Michalet S, Darbour N, Bayet C, Skandrani I, et al. Antioxidant and antigenotoxic properties of compounds isolated from Marrubium deserti de Noé. Food Chem Toxicol. 2011;49:3328-3335.   Back to cited text no. 50
    
51.
Sahpaz S, Garbacki N, Tits M, Bailleul F. Isolation and pharmacological activity of phenylpropanoid esters from Marrubium vulgare. J Ethnopharmacol. 2002;79:389-392.   Back to cited text no. 51
    
52.
Pukalskas A, Venskutonis PR, Salido S, de Waard P, van Beek TA. Isolation, identification and activity of natural antioxidants from horehound (Marrubium vulgare L.) cultivated in Lithuania. Food Chem. 2012;130:695-701.  Back to cited text no. 52
    
53.
Zhou F, Peng J, Zhao Y, Huang W, Jiang Y, Li M, et al. Varietal classification and antioxidant activity prediction of Osmanthus fragrans Lour. Flowers using UPLC-PDA/QTOF-MS and multivariable analysis. Food Chem. 2017;217:490-497.   Back to cited text no. 53
    
54.
Zhang L, Yue HL, Zhao XH, Li J, Shao Y. Separation of Four Phenylpropanoid Glycosides from a Chinese Herb by HSCCC. J Chrom Sci. 2015;53:860-865  Back to cited text no. 54
    
55.
Saracoglu I, Inoue M, Calis I, Ogihara Y. Studies on constituents with cytotoxic and cytostatic activity of two Turkish medicinal plants Phlomis armeniaca and Scutellaria salviifolia. Biol Pharm Bull. 1995;18:1396-1400.   Back to cited text no. 55
    
56.
Sarikurkcu C, Uren MC, Tepe B, Cengiz M, Kocak MS. Phlomis armeniaca: Phenolic compounds, enzyme inhibitory and antioxidant activities. Indus Crops Prod. 2015;78:95-101.  Back to cited text no. 56
    
57.
Aboutabl EA, Meselhy MR, Afifi MS. Iridoids from Phlomis aurea Decne growing in Egypt. Pharmazie. 2002;57:646-647.  Back to cited text no. 57
    
58.
Khitri W, Smati D, Mitaine-Offer AC, Paululat T, Lacaille-Dubois MA. Chemical constituents from Phlomis bovei Noë and their chemotaxonomic significance. Biochem System Ecol. 2020;91:104054.  Back to cited text no. 58
    
59.
Kirmizibekmez H, Calis I, Perozzo R, Brun R, Dönmez AA, Linden A, et al. Inhibiting activities of the secondary metabolites of Phlomis brunneogaleata against parasitic protozoa and plasmodial enoyl-ACP Reductase, a crucial enzyme in fatty acid biosynthesis. Planta Med. 2004;70:711-717.   Back to cited text no. 59
    
60.
Delazar A, Sabzevari A, Mojarrab M, Nazemiyeh H, Esnaashari S, Nahar L, et al. Free-radical-scavenging principles from Phlomis caucasica. J Nat Med. 2008;62:464-466.   Back to cited text no. 60
    
61.
Kirmizibekmez H, Montoro P, Piacente S, Pizza C, Dönmez A, Caliş I. Identification by HPLC-PAD-MS and quantification by HPLC-PAD of phenylethanoid glycosides of five Phlomis species. Phytochem Anal. 2005;16:1-6.   Back to cited text no. 61
    
62.
Nazemiyeh H, Rahman MM, Gibbons S, Nahar L, Delazar A, Ghahramani MA, Talebpour AH, Sarker SD. Assessment of the antibacterial activity of phenylethanoid glycosides from Phlomis lanceolata against multiple-drug-resistant strains of Staphylococcus aureus. J Nat Med. 2008;62:91-95.  Back to cited text no. 62
    
63.
Ersöz T, Schühly W, Popov S, Handjieva N, Sticher O, Caliş I. Iridoid and phenylethanoid glycosides from Phlomis longifolia var. longifolia. Nat Prod Lett. 2001;15:345-351.   Back to cited text no. 63
    
64.
Ersöz T, Alipieva KI, Yalçin FN, Akbay P, Handjieva N, Dönmez AA, et al. Physocalycoside, a new phenylethanoid glycoside from Phlomis physocalyx Hub.-Mor. Z Naturforsch C J Biosci. 2003;58:471-476.   Back to cited text no. 64
    
65.
Ersöz T, Ivancheva S, Akbay P, Sticher O, Caliş I. Iridoid and phenylethanoid glycosides from Phlomis tuberosa L. Z Naturforsch C J Biosci. 2001;56:695-698.   Back to cited text no. 65
    
66.
Zhang LN, Deng RX, Wang Y, Liu MM, Liu P. Chemical Constituents from Rhizome of Phlomis umbrosa Turcz var. latibracteata Sun ex C H Hu. Chinese Pharm J. 2019;54:450-456.  Back to cited text no. 66
    
67.
Calis I, Kirmizibekmez H, Beutler JA, Donmez A, Yalçin FN, Kiliç E, et al. Secondary Metabolites of Phlomis viscosa and Their Biological Activities. Turk J Chem. 2005;29:71-81  Back to cited text no. 67
    
68.
Martin F, Hay AE, Corno L, Gupta MP, Hostettmann K. Iridoid glycosides from the stems of Pithecoctenium crucigerum (Bignoniaceae). Phytochemistry. 2007;68:1307-1311.  Back to cited text no. 68
    
69.
Guragac Dereli FT, Genc Y, Saracoglu I, Kupeli Akkol E. Enzyme inhibitory assessment of the isolated constituents from Plantago holosteum Scop. Z Naturforsch C J Biosci. 2020;75:121-128.   Back to cited text no. 69
    
70.
Simamora A, Santoso AW, Timotius KH, Rahayu I. Antioxidant Activity, Enzyme Inhibition Potentials, and Phytochemical Profiling of Premna serratifolia L. Leaf Extracts. Int J Food Sci. 2020;2020:3436940.   Back to cited text no. 70
    
71.
Phakeovilay C, Disadee W, Sahakitpichan P, Sitthimonchai S, Kittakoop P, Ruchirawat S, Kanchanapoom T. Phenylethanoid and flavone glycosides from Ruellia tuberosa L. J Nat Med. 2013;67:228-233.   Back to cited text no. 71
    
72.
Xu HT, Zhang CG, He YQ, Shi SS, Wang YL, Chou GX. Phenylethanoid glycosides from the Schnabelia nepetifolia (Benth.) P.D.Cantino promote the proliferation of osteoblasts. Phytochemistry. 2019;164:111-121.   Back to cited text no. 72
    
73.
Zengin G, Uğurlu A, Baloglu MC, Diuzheva A, Jekő J, Cziáky Z, et al. Chemical fingerprints, antioxidant, enzyme inhibitory, and cell assays of three extracts obtained from Sideritis ozturkii Aytac & Aksoy: An endemic plant from Turkey. J Pharm Biomed Anal. 2019;171:118-125.   Back to cited text no. 73
    
74.
Todorova M, Trendafilova A. Sideritis scardica Griseb., an endemic species of Balkan peninsula: traditional uses, cultivation, chemical composition, biological activity. J Ethnopharmacol. 2014;152:256-265.   Back to cited text no. 74
    
75.
Miyase T, Yamamoto R, Ueno A. Phenylethanoid glycosides from Stachys officinalis. Phytochemistry. 1996;43:475-479.   Back to cited text no. 75
    
76.
Machida K, Ohkawa N, Ohsawa A, Kikuchi M. Two new phenolic glycosides from Syringa reticulata. J Nat Med. 2009;63:192-194.   Back to cited text no. 76
    
77.
Filipek A, Wyszomierska J, Michalak B, Kiss AK. Syringa vulgaris bark as a source of compounds affecting the release of inflammatory mediators from human neutrophils and monocytes/macrophages. Phytochem Lett. 2019;30:309-313.  Back to cited text no. 77
    
78.
Frezza C, Venditti A, Matrone G, Serafini I, Foddai S, Bianco A, Serafini M. Iridoid glycosides and polyphenolic compounds from Teucrium chamaedrys L. Nat Prod Res. 2018;32:1583-1589.   Back to cited text no. 78
    
79.
Milutinović MG, Maksimović VM, Cvetković DM, Nikodijević DD, Stanković MS, Pešić M, et al. Potential of Teucrium chamaedrys L. to modulate apoptosis and biotransformation in colorectal carcinoma cells. J Ethnopharmacol. 2019;240:111951.   Back to cited text no. 79
    
80.
Klimek B. 6'-0-apiosyl-verbascoside in the flowers of mullein (Verbascum species). Acta Pol Pharm. 1996;53:137-140.  Back to cited text no. 80
    
81.
Georgiev MI, Ali K, Alipieva K, Verpoorte R, Choi YH. Metabolic differentiations and classification of Verbascum species by NMR-based metabolomics. Phytochemistry. 2011;72:2045-2051.   Back to cited text no. 81
    
82.
Dimitrova P, Alipieva K, Grozdanova T, Simova S, Bankova V, Georgiev MI, Popova MP. New iridoids from Verbascum nobile and their effect on lectin-induced T cell activation and proliferation. Food Chem Toxicol. 2018;111:605-615.   Back to cited text no. 82
    
83.
Klimek B. Hydroxycinnamoyl ester glycosides and saponins from flowers of Verbascum phlomoides. Phytochemistry 1996;43:1281-1284.  Back to cited text no. 83
    
84.
Tatli II, Akdemir ZS, Yesilada E, Küpeli E. Anti-inflammatory and antinociceptive potential of major phenolics from Verbascum salviifolium Boiss. Z Naturforsch C J Biosci. 2008;63:196-202.   Back to cited text no. 84
    
85.
Warashina T, Miyase T, Ueno A. Phenylethanoid and lignan glycosides from Verbascum thapsus. Phytochemistry. 1992;31:961-965.  Back to cited text no. 85
    
86.
Georgiev M, Alipieva K, Orhan I, Abrashev R, Denev P, Angelova M. Antioxidant and cholinesterases inhibitory activities of Verbascum xanthophoeniceum Griseb. and its phenylethanoid glycosides. Food Chem. 2011;128:100-105.   Back to cited text no. 86
    
87.
Georgiev M, Pastore S, Lulli D, Alipieva K, Kostyuk V, Potapovich A, et al. Verbascum xanthophoeniceum-derived phenylethanoid glycosides are potent inhibitors of inflammatory chemokines in dormant and interferon-gamma-stimulated human keratinocytes. J Ethnopharmacol. 2012;144:754-760.   Back to cited text no. 87
    
88.
Qu J, Yan X, Li C, Wen J, Lu C, Ren J, et al. Comparative Evaluation of Raw and Ripe Fruits of Forsythia suspensa by HPLC-ESI-MS/MS Analysis and Anti-Microbial Assay. J Chromatogr Sci. 2017;55:451-458.   Back to cited text no. 88
    
89.
Dong Z, Lu X, Tong X, Dong Y, Tang L, Liu M. Forsythiae Fructus: A Review on its Phytochemistry, Quality Control, Pharmacology and Pharmacokinetics. Molecules. 2017;22:1466.   Back to cited text no. 89
    
90.
Wu MH, Shi SM, Cao H. [Herbalogical study on merit rating of Forsythiae Fructus based on near-mature fruit and hyper-mature fruit]. Zhongguo Zhong Yao Za Zhi. 2019;44:5508-5512.   Back to cited text no. 90
    
91.
Chen CL, Zhang DD. Anti-inflammatory effects of 81 chinese herb extracts and their correlation with the characteristics of traditional chinese medicine. Evid Based Complement Alternat Med. 2014;2014:985176.   Back to cited text no. 91
    
92.
Wei Q, Zhang R, Wang Q, Yan XJ, Yu QW, Yan FX, Li C, Pei YH. Iridoid, phenylethanoid and flavonoid glycosides from Forsythia suspensa. Nat Prod Res. 2020;34:1320-1325.   Back to cited text no. 92
    
93.
Mao Q, Shi L, Wang ZG, Luo YH, Wang YY, Li X, et al. Chemical profiles and pharmacological activities of Chang-Kang-Fang, a multi-herb Chinese medicinal formula, for treating irritable bowel syndrome. J Ethnopharmacol. 2017;201:123-135.   Back to cited text no. 93
    
94.
Wu L, Du S, Yang F, Ni Z, Chen Z, Liu X, Wang Y, Zhou Q, Li W, Qin K. Simultaneous determination of nineteen compounds of Dahuang zhechong pill in rat plasma by UHPLC-MS/MS and its application in a pharmacokinetic study. J Chromatogr B Analyt Technol Biomed Life Sci. 2020;1151:122200.   Back to cited text no. 94
    
95.
Ni ZH, Wu L, Cao KX, Zhang XQ, Wang DY, Zeng YW, et al. Investigation of the pharmacodynamic substances in dahuang zhechong pill that inhibit energy metabolism. J Ethnopharmacol. 2020;251:112332.   Back to cited text no. 95
    
96.
Dong Q, Qiu LL, Zhang CE, Chen LH, Feng WW, Ma LN, et al. Identification of compounds in an anti-fibrosis Chinese medicine (Fufang Biejia Ruangan Pill) and its absorbed components in rat biofluids and liver by UPLC-MS. J Chromatogr B Analyt Technol Biomed Life Sci. 2016;1026:145-151.  Back to cited text no. 96
    
97.
Zhang Y, Mao X, Chen W, Guo X, Yu L, Jiang F, et al. A Discovery of Clinically Approved Formula FBRP for Repositioning to Treat HCC by Inhibiting PI3K/AKT/NF-κB Activation. Mol Ther Nucleic Acids. 2020;19:890-904.   Back to cited text no. 97
    
98.
Ren Y, Yin ZH, Dai JX, Yang Z, Ye BB, Ma YS, et al. Evidence- Based Complementary and Alternative Medicine Exploring Active Components and Mechanism of Jinhua Qinggan Granules in Treatment of COVID-19 Based on Virus- Host Interaction. Nat Prod. Commun. 2020;15:1-11.  Back to cited text no. 98
    
99.
Wang J, Qi F. Traditional Chinese medicine to treat COVID-19: the importance of evidence-based research. Drug Discov Ther. 2020;14:149-150.   Back to cited text no. 99
    
100.
Chen H, Jie C, Tang LP, Meng H, Li XB, Li YB, et al. New insights into the effects and mechanism of a classic traditional Chinese medicinal formula on influenza prevention. Phytomedicine. 2017;27:52-62.   Back to cited text no. 100
    
101.
Liao Y, Hou Y, Zhong Y, Chen H, Xu C, Tsunoda M, et al. One-step ionic liquid-based ultrasound-assisted dispersive liquid-liquid microextraction coupled with high-performance liquid chromatography for the determination of pyrethroids in traditional Chinese medicine oral liquid preparations. BMC Chem. 2019;13:61.   Back to cited text no. 101
    
102.
Chen X, Wu Y, Chen C, Gu Y, Zhu C, Wang S, et al. Identifying potential anti-COVID-19 pharmacological components of traditional Chinese medicine Lianhuaqingwen capsule based on human exposure and ACE2 biochromatography screening. Acta Pharm Sin B. 2021;11:222-236.  Back to cited text no. 102
    
103.
Jia W, Wang C, Wang Y, Pan G, Jiang M, Li Z, et al. Qualitative and quantitative analysis of the major constituents in Chinese medical preparation Lianhua-Qingwen capsule by UPLC-DAD-QTOF-MS. ScientificWorldJournal. 2015;2015:731765.   Back to cited text no. 103
    
104.
Qin H, Zhang LL, Xiong XL, Jiang ZX, Xiao CP, Zhang LL, et al. Li-Dan-He-Ji Improves Infantile Cholestasis Hepatopathy Through Inhibiting Calcium-Sensing Receptor-Mediated Hepatocyte Apoptosis. Front Pharmacol. 2020;11:156.   Back to cited text no. 104
    
105.
Cui HR, Xu GH, Wu MQ, Jiang WY, Han JJ, Wang JB, et al. Simultaneous Determination of Eight Active Components in Liuwei Wuling Tablet Using HPLC. Chinese Herb Med. 2016;8:331-336.  Back to cited text no. 105
    
106.
Zhou XJ, Li YF, Chen Y, Chen XH, Qu Y, Ren XD, et al. Rapid establishment of Q-Marker database for Qingreling Granules with UPLC coupled with hybrid quadrupole-orbitrap mass spectrometry. Chinese Tradit Herbal Drugs 2017;48:67-74.  Back to cited text no. 106
    
107.
Zheng Y, Liu S, Fan C, Zeng H, Huang H, Tian C, et al. Holistic quality evaluation of Qingwen Baidu Decoction and its anti-inflammatory effects. J Ethnopharmacol. 2020;263:113145.  Back to cited text no. 107
    
108.
Wen J, Wang R, Liu H, Tong Y, Wei S, Zhou X, et al. Potential therapeutic effect of Qingwen Baidu Decoction against Corona Virus Disease 2019: a mini review. Chin Med. 2020;15:48.   Back to cited text no. 108
    
109.
Wang P, Huang H, Zhong J, Cai H, Huang Y, Chen D, et al. Qinwen Baidu decoction for sepsis: A protocol for a systematic review and meta-analysis. Medicine (Baltimore). 2019;98:e14761.   Back to cited text no. 109
    
110.
Yang P, Gao R, Liu Z, Qu Q, Yang C, Shi X, et al. Analysis of chemical constituents and six compounds in Qu-feng-sheng-shi Granules via HPLC-ESI-Q/TOF-MSn and HPLC-UV technique. Biomed Chromatogr. 2020;34:e4829.   Back to cited text no. 110
    
111.
Ji S, Liu ZZ, Wu J, Du Y, Su ZY, Wang TY, et al. Chemical Profiling and Comparison of Sangju Ganmao Tablet and Its Component Herbs Using Two-Dimensional Liquid Chromatography to Explore Compatibility Mechanism of Herbs. Front Pharmacol. 2018;9:1167.   Back to cited text no. 111
    
112.
Shi Y, Xu J, Qiao Y, Zhang W, Liu D, Qin M, et al, Dong M. Effects of shuanghuanglian injection on the activities of CYP1A2, 2C11, 2D1 and 3A1/2 in rats in vivo and in vitro. Xenobiotica. 2019;49:905-911.   Back to cited text no. 112
    
113.
Han J, Zhang Y, Pan C, Xian Z, Pan C, Zhao Y, et al. Forsythoside A and Forsythoside B Contribute to Shuanghuanglian Injection-Induced Pseudoallergic Reactions through the RhoA/ROCK Signaling Pathway. Int J Mol Sci. 2019;20:6266.   Back to cited text no. 113
    
114.
Su HX, Yao S, Zhao WF, Li MJ, Liu J, Shang WJ, et al. Anti-SARS-CoV-2 activities in vitro of Shuanghuanglian preparations and bioactive ingredients. Acta Pharmacol Sin. 2020;41:1167-1177.   Back to cited text no. 114
    
115.
Liu X, Zhang H, Xu J, Gong S, Han Y, Zhang T, et al. Identification of absorbed components and their metabolites in rat plasma after oral administration of Shufeng Jiedu capsule using ultra-performance liquid chromatography/quadrupole time-of-flight mass spectrometry. Rapid Commun Mass Spectrom. 2019;33:1494-1501.   Back to cited text no. 115
    
116.
Han YQ, Cao Y, Dong YN, Li XY, Wu Q, Xu J, et al. Spectrum-effect relationship of anti-inflammatory activity of Shufeng Jiedu Capsule based on neural network analysis. Chinese Tradit Herbal Drugs 2019;50:3526-353.  Back to cited text no. 116
    
117.
Xia RY, Hu XY, Fei YT, Willcox M, Wen LZ, Yu MK, et al. Shufeng Jiedu capsules for treating acute exacerbations of chronic obstructive pulmonary disease: a systematic review and meta-analysis. BMC Complement Med Ther. 2020;20:151.  Back to cited text no. 117
    
118.
Shan YQ, Cao Y, Dong YN, Wu Q, Xu J, et al. Spectrum-effect relationship of Shufeng Jiedu Capsules in dispersing wind and releasing exterior. Chinese Tradit Herbal Drugs 2019;50:3534-3540.  Back to cited text no. 118
    
119.
Yao Z, Yu J, Tang Z, Liu H, Ruan K, Song Z, et al. Multi-Evaluating Strategy for Siji-kangbingdu Mixture: Chemical Profiling, Fingerprint Characterization, and Quantitative Analysis. Molecules. 2019;24:3545.   Back to cited text no. 119
    
120.
Liu H, Ding XF, Guo R, Zhao MF, Deng D, Hao Y, et al. Effects and safety of tanreqing injection on viral pneumonia: A protocol for systematic review and meta-analysis. Medicine (Baltimore). 2020;99:e21808.   Back to cited text no. 120
    
121.
Jian LH, Hu Q, Zhong JQ, Meng Q, Wang K, Ji S. Determination of forsythoside E in Tanreqing injection by LC-MS/MS. Chinese J Pharm Anal. 2013;33:435-438.  Back to cited text no. 121
    
122.
Feng SX, Li XH, Wang MM, Hao R, Li MM, Zhang L, et al. A sensitive HPLC-MS method for simultaneous determination of thirteen components in rat plasma and its application to pharmacokinetic study of Tanreqing injection. J Pharm Biomed Anal. 2018;148:205-213.   Back to cited text no. 122
    
123.
Tian G, Yang XF, Li C, Wu F, Liu X, Feng L, et al. [Simultaneous determination of 9 components in Xiao'er Chiqiao Qingre granules by UPLC]. Zhongguo Zhong Yao Za Zhi. 2018;43:2081-2085.   Back to cited text no. 123
    
124.
Xu T, Li X, Huang M, Wang Q, Li C, Tian G, et al. A Preferable Approach for the Quality Control of Xiaoer Chiqiao Qingre Granules Based on the Combination of Chromatographic Fingerprints and Chemometrics. J Anal Methods Chem. 2020;2020:6836981.   Back to cited text no. 124
    
125.
Law AH, Yang CL, Lau AS, Chan GC. Antiviral effect of forsythoside A from Forsythia suspensa (Thunb.) Vahl fruit against influenza A virus through reduction of viral M1 protein. J Ethnopharmacol. 2017;209:236-247.   Back to cited text no. 125
    
126.
Lee H, Kang B, Hong M, Lee HL, Choi JY, Lee JA. Eunkyosan for the common cold: A PRISMA-compliment systematic review of randomised, controlled trials. Medicine (Baltimore). 2020;99:e21415.   Back to cited text no. 126
    
127.
Sang Q, Jia Q, Zhang H, Lin C, Zhao X, Zhang M, et al. Chemical profiling and quality evaluation of Zhishi-Xiebai-Guizhi Decoction by UPLC-Q-TOF-MS and UPLC fingerprint. J Pharm Biomed Anal. 2021;194:113771.  Back to cited text no. 127
    
128.
Tang Y, Cai H, Zhan Z, Luo Y, Huang Y, Chen D, Chen B. Herbal medicine (zhishi xiebai guizhi decoction) for unstable angina: Protocol for a systematic review and meta-analysis. Medicine (Baltimore). 2018;97:e13965.   Back to cited text no. 128
    
129.
Runfeng L, Yunlong H, Jicheng H, Weiqi P, Qinhai M, Yongxia S, et al. Lianhuaqingwen exerts anti-viral and anti-inflammatory activity against novel coronavirus (SARS-CoV-2). Pharmacol Res. 2020;156:104761.   Back to cited text no. 129
    
130.
Zhang Q, Cao F, Ji G, Xu X, Sun Y, Li J, et al. The efficacy and safety of Lianhua Qingwen (LHQW) for coronavirus disease 2019 (COVID-19): A protocol for systematic review and meta analysis. Medicine (Baltimore). 2020;99:e20979.  Back to cited text no. 130
    
131.
Hu K, Guan WJ, Bi Y, Zhang W, Li L, Zhang B, Liu Q, Song Y, Li X, Duan Z, Zheng Q, Yang Z, Liang J, Han M, Ruan L, Wu C, Zhang Y, Jia ZH, Zhong NS. Efficacy and safety of Lianhuaqingwen capsules, a repurposed Chinese herb, in patients with coronavirus disease 2019: A multicenter, prospective, randomized controlled trial. Phytomedicine. 2020:153242.   Back to cited text no. 131
    
132.
Fang J, Li H, Du W, Yu P, Guan YY, Ma SY, et al. Efficacy of Early Combination Therapy With Lianhuaqingwen and Arbidol in Moderate and Severe COVID-19 Patients: A Retrospective Cohort Study. Front Pharmacol. 2020;11:560209.   Back to cited text no. 132
    
133.
Zeng M, Li L, Wu Z. Traditional Chinese medicine Lianhua Qingwen treating corona virus disease 2019(COVID-19): Meta-analysis of randomized controlled trials. PLoS One. 2020;15:e0238828.   Back to cited text no. 133
    
134.
Liu N, Zhang T, Ma L, Wang H, Cao Y, Yang Y, et al. Efficacy and safety of Lianhua Qingwen in the treatment of patients with moderate COVID-19 infection: A protocol for systematic review and meta analysis. Medicine (Baltimore). 2020;99:e21614.   Back to cited text no. 134
    
135.
Liu M, Gao Y, Yuan Y, Yang K, Shi S, Tian J, Zhang J. Efficacy and safety of herbal medicine (Lianhuaqingwen) for treating COVID-19: A systematic review and meta-analysis. Integr Med Res. 2021;10:100644.   Back to cited text no. 135
    
136.
Qiu Y, Pan X, Su L, Lui H, Li YD. Effects and safety of Tanreqing injection on viral pneumonia: A protocol for systematic review and meta-analysis. Medicine (Baltimore). 2020;99:e22022.  Back to cited text no. 136
    
137.
Wang L, Fan Y, Xu J, Deng H, Geng C, Jia B. The efficacy and safety of Tanreqing injection combined with western medicine for severe pneumonia: A protocol for systematic review and meta-analysis. Medicine (Baltimore). 2020;99:e22010.   Back to cited text no. 137
    
138.
Zhuang W, Fan Z, Chu Y, Wang H, Yang Y, Wu L, et al. Chinese Patent Medicines in the Treatment of Coronavirus Disease 2019 (COVID-19) in China. Front Pharmacol. 2020;11:1066.  Back to cited text no. 138
    
139.
Xia L, Shi Y, Su J, Friedemann T, Tao Z, Lu Y, et al. Shufeng Jiedu, a promising herbal therapy for moderate COVID-19:Antiviral and anti-inflammatory properties, pathways of bioactive compounds, and a clinical real-world pragmatic study. Phytomedicine. 2020:153390.   Back to cited text no. 139
    
140.
Tao Z, Zhang L, Friedemann T, Yang G, Li J, Wen Y, et al. Systematic analyses on the potential immune and anti-inflammatory mechanisms of Shufeng Jiedu Capsule against Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2)-caused pneumonia. J Funct Foods. 2020;75:104243.   Back to cited text no. 140
    
141.
Chen X, Yin YH, Zhang MY, Liu JY, Li R, Qu YQ. Investigating the mechanism of ShuFeng JieDu capsule for the treatment of novel Coronavirus pneumonia (COVID-19) based on network pharmacology. Int J Med Sci. 2020;17:2511-2530.   Back to cited text no. 141
    
142.
Chen J, Lin S, Niu C, Xiao Q. Clinical evaluation of Shufeng Jiedu Capsules combined with umifenovir (Arbidol) in the treatment of common-type COVID-19: a retrospective study. Expert Rev Respir Med. 2020:1-9.   Back to cited text no. 142
    
143.
Li R, Li Y, Li B, Sun H, Liu X, Ge X, et al. Efficacy and safety of Shufeng Jiedu capsule for coronavirus disease 2019 (COVID-19): A protocol for systematic review and meta-analysis. Medicine (Baltimore). 2020;99:e21615.  Back to cited text no. 143
    
144.
Endo K, Hikino H. Structure forsythoside C and D, an antibacterial principles of Forsythia suspensa fruits. Heterocycles 1982;19:2033-2036.  Back to cited text no. 144
    
145.
Endo K, Takahashi K, Abe T, Hikino H. Structure of forsythoside A, an antibacterial principle of Forsythia suspensa leaves. Heterocycles 1981;16:1311-1314.  Back to cited text no. 145
    
146.
Endo K, Takahashi K, Abe T, Hikino H. Structure of forsythoside B, an antibacterial principle of Forsythia koreana stems. Heterocycles 1981;19:261-264.  Back to cited text no. 146
    
147.
Wei L, Mei Y, Zou L, Chen J, Tan M, Wang C, et al. Distribution Patterns for Bioactive Constituents in Pericarp, Stalk and Seed of Forsythiae Fructus. Molecules. 2020;25:340.   Back to cited text no. 147
    
148.
Georgiev MI, Ludwig-Müller J, Alipieva K, Lippert A. Sonication-assisted Agrobacterium rhizogenes-mediated transformation of Verbascum xanthophoeniceum Griseb. for bioactive metabolite accumulation. Plant Cell Rep. 2011;30:859-866.   Back to cited text no. 148
    
149.
Wysokińska H, Lisowska K, Floryanowicz-Czekalska K. Transformation of Catalpa ovata by Agrobacterium rhizogenes and phenylethanoid glycosides production in transformed root cultures. Z Naturforsch C J Biosci. 2001;56:375-381.   Back to cited text no. 149
    
150.
Wilczańska-Barska A, Królicka A, Głód D, Majdan M, Kawiak A, Krauze-Baranowska M. Enhanced accumulation of secondary metabolites in hairy root cultures of Scutellaria lateriflora following elicitation. Biotechnol Lett. 2012;34:1757-1763.   Back to cited text no. 150
    
151.
Yamamoto H, Yoshida K, Kondo Y, Inoue K. Production of cornoside in Abeliophyllum distichum cell suspension cultures. Phytochemitry 1998;48:273-277.  Back to cited text no. 151
    
152.
Endo K, Hikino H. Structures of rengyol, rengyoxide, and rengyolone, new cyclohexylethane derivatives from Forsythia suspensa fruits. Can. J. Chem. 1984;62:2011-2014.  Back to cited text no. 152
    
153.
Andary C, Privat G, Wylde R, Heitz A. Pheliposide et arenarioside, deux nouveaux esters heterosidiques de l'acide cafeique isoles de Orobanche arenaria. J Nat Prod. 1985;48:778-783.  Back to cited text no. 153
    
154.
Jedrejek D, Pawelec S, Piwowarczyk R, Pecio Ł Stochmal A. Identification and occurrence of phenylethanoid and iridoid glycosides in six Polish broomrapes (Orobanche spp. and Phelipanche spp., Orobanchaceae). Phytochemistry. 2020;170:112189.   Back to cited text no. 154
    
155.
Li ZJ, Huang HQ, Cai MS. Facile synthesis of the sugar cores from phenylpropanoid glycosides. Carbohydr Res. 1994;265:227-236.  Back to cited text no. 155
    
156.
Zhou FY, She J, Wang YG. Synthesis of a benzyl-protected analog of arenarioside, a trisaccharide phenylpropanoid glycoside. Carbohydr Res. 2006;341:2469-2477.   Back to cited text no. 156
    
157.
Liu YG, Li X, Xiong DC, Yu B, Pu X, Ye XS. Synthetic phenylethanoid glycoside derivatives as potent neuroprotective agents. Eur J Med Chem. 2015;95:313-323.  Back to cited text no. 157
    
158.
Zhao Y, Zeng J, Liu Y, Xiao X, Sun G, Sun J, Shu P, Fu D, Meng L, Wan Q. Collective syntheses of phenylethanoid glycosides by interrupted Pummerer reaction mediated glycosylations. J Carbohydrate Chem. 2018;37:471-497.  Back to cited text no. 158
    
159.
Hu Z, Silipo A, Li W, Molinaro A, Yu B. Synthesis of Forsythenethoside A, a Neuroprotective Macrocyclic Phenylethanoid Glycoside, and NMR Analysis of Conformers. J Org Chem. 2019;84:13733-13743.   Back to cited text no. 159
    
160.
Shu P, Zhang L, Liu A, Li J, Liu Q, Sun N, Zhang Y, Wei X, Cui M, Ju Z, Xu Z. Six Natural Phenylethanoid Glycosides: Total Synthesis, Antioxidant and Tyrosinase Inhibitory Activities. ChemistrySelect 2020;5:10817-10820.  Back to cited text no. 160
    
161.
Wang FN, Ma ZQ, Liu Y, Guo YZ, Gu ZW. New phenylethanoid glycosides from the fruits of forsythia suspense (thunb.) vahl. Molecules. 2009;14:1324-1331.   Back to cited text no. 161
    
162.
Qi M, Zhao S, Zhou B, Zhang M, Zhang H, Wang Y, Hu P. Probing the degradation mechanism of forsythiaside A and simultaneous determination of three forsythiasides in Forsythia preparations by a single marker. J Sep Sci. 2019;42:3503-3511.   Back to cited text no. 162
    
163.
Yan XJ, Xiang Z, Wen J, Zhang WJ, Yang B, Qu ZY, Xu BL, Li WL. Phenylethanoid glycosides from fruits of Forsythia Suspensa. Chinese Tradit Herbal Drug 2016;47:3262-3265.  Back to cited text no. 163
    
164.
Shao SY, Feng ZM, Yang YN, Jiang JS, Zhang PC. Eight new phenylethanoid glycoside derivatives possessing potential hepatoprotective activities from the fruits of Forsythia suspensa. Fitoterapia. 2017;122:132-137.   Back to cited text no. 164
    
165.
Li C, Dai Y, Zhang SX, Duan YH, Liu ML, Chen LY, Yao XS. Quinoid glycosides from Forsythia suspensa. Phytochemistry. 2014;104:105-113.   Back to cited text no. 165
    
166.
Shao SY, Feng ZM, Yang YN, Jiang JS, Zhang PC. Forsythenethosides A and B: two new phenylethanoid glycosides with a 15-membered ring from Forsythia suspensa. Org Biomol Chem. 2017;15:7034-7039.   Back to cited text no. 166
    
167.
Zhang F, Yang YN, Feng ZM, Jiang JS, Zhang PC. Four new phenylethanoid and flavonoid glycoside dimers from the fruits of Forsythia suspensa and their neuroprotective activities. RSC Adv. 2017;7:24963.  Back to cited text no. 167
    
168.
Nishibe S, Okabe K, Tsukamoto H, Sakushima A., Hisada S. The Structure of Forsythiaside isolated from Forsythia suspensa. Chem Pharm Bull 1982;30:1048-1050.  Back to cited text no. 168
    
169.
Nishibe S, Okabe K, Tsukamoto H, Sakushima A., Hisada S., Baba H, Akisada T. Studies on the Chinese Crude Drug “Forsythiae Fructus.” VI. The Structure and Antibacterial Activity of Suspensaside isolated from Forsythia suspensa. Chem Pharm Bull. 1982;30:4548-4553.  Back to cited text no. 169
    
170.
Kitagawa S, Tsukamoto H, Hisada S, Nishibe S. Studies on the Chinese crude drug “forsythiae fructus”. VII. A new caffeoyl glycoside from Forsythia viridissima. Chem Pharm Bull. 1984;32:1209-1213.  Back to cited text no. 170
    
171.
Ming DS, Yu DQ, Yu SS. Two new caffeyol glycosides from Forsythia suspensa. J Asian Nat Prod Res. 1999;1:327-335.   Back to cited text no. 171
    
172.
Shao S, Yang Y, Feng Z, Jiang J, Zhang P. New triacetic acid lactone glycosides from the fruits of Forsythia suspensa and their nitric oxide production inhibitory activity. Carbohydr Res. 2020;488:107908.   Back to cited text no. 172
    
173.
Shao SY, Yang YN, Feng ZM, Jiang JS, Zhang PC. Anti-inflammatory phenylpropanoid glycosides from the fruits of Forsythia suspensa. Bioorg Med Chem Lett. 2019;29:126635.   Back to cited text no. 173
    
174.
Ge Y, Wang Y, Chen P, Wang Y, Hou C, Wu Y, et al. Polyhydroxytriterpenoids and Phenolic Constituents from Forsythia suspensa (Thunb.) Vahl Leaves. J Agric Food Chem. 2016;64:125-131.   Back to cited text no. 174
    
175.
Kuang HX, Xia YG, Yang BY, Liang J, Zhang QB, Li GY. A New Caffeoyl Phenylethanoid Glycoside from the Unripe Fruits of Forsythia suspensa. Chinese J Nat Med. 2009;7:278-282.  Back to cited text no. 175
    
176.
Kuang HX, Xia YG, Liang J, Yang BY, Wang QH. Lianqiaoxinoside B, a novel caffeoyl phenylethanoid glycoside from Forsythia suspensa. Molecules. 2011;16:5674-5681.   Back to cited text no. 176
    
177.
Ming DS, Yu DQ, Yu SS. New Quinoid Glycosides from Forsythia suspensa. J Nat Prod. 1998;61:377-379.   Back to cited text no. 177
    
178.
Liu D.-L, Zhang Y, Xu S-X, Xu Y, Wang Z.-X. Phenylethanoid glycosides from Forsythia suspensa Vahl. J. Chin. Pharm. Sci. 1998;7:103-105.  Back to cited text no. 178
    
179.
Guo H, Liu AH, Ye M, Yang M, Guo DA. Characterization of phenolic compounds in the fruits of Forsythia suspensa by high-performance liquid chromatography coupled with electrospray ionization tandem mass spectrometry. Rapid Commun Mass Spectrom. 2007;21:715-729.   Back to cited text no. 179
    
180.
Li C, Dai Y, Duan YH, Liu ML, Yao XS. A new lignan glycoside from Forsythia suspensa. Chin J Nat Med. 2014;12:697-699.   Back to cited text no. 180
    
181.
Bao J, Ding R, Zou L, Zhang C, Wang K, Liu F, et al. Forsythiae Fructus Inhibits B16 Melanoma Growth Involving MAPKs/Nrf2/HO-1 Mediated Anti-Oxidation and Anti-Inflammation. Am J Chin Med. 2016;44:1043-1061.   Back to cited text no. 181
    
182.
Bao J, Liu F, Zhang C, Wang K, Jia X, Wang X, et al. Anti-melanoma activity of Forsythiae Fructus aqueous extract in mice involves regulation of glycerophospholipid metabolisms by UPLC/Q-TOF MS-based metabolomics study. Sci Rep. 2016;6:39415.  Back to cited text no. 182
    
183.
Lee JJ, Kim KH, Kim EJ, Choi JY, Kim SJ, Jeong SI, et al. Anti-inflammatory activity of the decoction of Forsythia suspensa (Thunb.) Vahl is related to Nrf2 and A20. J Ethnopharmacol. 2018;227:97-104.   Back to cited text no. 183
    
184.
Jia J, Zhang F, Li Z, Qin X, Zhang L. Comparison of Fruits of Forsythia suspensa at Two Different Maturation Stages by NMR-Based Metabolomics. Molecules. 2015;20:10065-10081.   Back to cited text no. 184
    
185.
Bao J, Ding RB, Liang Y, Liu F, Wang K, Jia X, et al. Differences in Chemical Component and Anticancer Activity of Green and Ripe Forsythiae Fructus. Am J Chin Med. 2017;45:1513-1536.   Back to cited text no. 185
    
186.
Bao J, Ding RB, Jia X, Liang Y, Liu F, Wang K, et al. Fast identification of anticancer constituents in Forsythiae Fructus based on metabolomics approaches. J Pharm Biomed Anal. 2018;154:312-320.   Back to cited text no. 186
    
187.
Okubo S, Ohta T, Shoyama Y, Uto T. Arctigenin suppresses cell proliferation via autophagy inhibition in hepatocellular carcinoma cells. J Nat Med. 2020;74:525-532.   Back to cited text no. 187
    
188.
Kim YL, Lee SK, Park KK, Chung WY. The Inhibitory Effects of Forsythia Koreana Extracts on the Metastatic Ability of Breast Cancer Cells and Bone Resorption by Osteoclasts. J Cancer Prev. 2016;21:88-94.   Back to cited text no. 188
    
189.
Rouf AS, Ozaki Y, Rashid MA, Rui J. Dammarane derivatives from the dried fruits of Forsythia suspensa. Phytochemistry. 2001;56:815-818.   Back to cited text no. 189
    
190.
Hawas UW, Gamal-Eldeen AM, El-Desouky SK, Kim YK, Huefner A, Saf R. Induction of caspase-8 and death receptors by a new dammarane skeleton from the dried fruits of Forsythia koreana. Z Naturforsch C J Biosci. 2013;68:29-38.  Back to cited text no. 190
    
191.
Lee SE, Lim C, Kim H, Cho S. A study of the anti-inflammatory effects of the ethyl acetate fraction of the methanol extract of Forsythiae fructus. Afr J Tradit Complement Altern Med. 2016;13:102-113.   Back to cited text no. 191
    
192.
Lee SE, Lim C, Ahn SC, Cho S. A Study of the Anti-Cancer Effects of the Hexane Fraction of the Methanol Extract of Forsythiae Fructus. Pharmacogn Mag. 2017;13:719-724.   Back to cited text no. 192
    
193.
Zhao L, Yan X, Shi J, Ren F, Liu L, Sun S, Shan B. Ethanol extract of Forsythia suspensa root induces apoptosis of esophageal carcinoma cells via the mitochondrial apoptotic pathway. Mol Med Rep. 2015;11:871-880.   Back to cited text no. 193
    
194.
Okubo S, Komori H, Kuwahara A, Ohta T, Shoyama Y, Uto T. Screening of Crude Drugs Used in Japanese Kampo Formulas for Autophagy-Mediated Cell Survival of the Human Hepatocellular Carcinoma Cell Line. Medicines (Basel). 2019;6:63.   Back to cited text no. 194
    
195.
Cheng-Dong N, Xin-Jia Y, Jing W, Wen-Lan LI, Yu F, Yuan-Yuan J, et al. [Study on molecular of anti-tumor mechanism of Forsythia suspensa based on molecular docking and network pharmacology]. Zhongguo Zhong Yao Za Zhi. 2020;45:4455-4465.  Back to cited text no. 195
    
196.
Chen X, Beutler JA, McCloud TG, Loehfelm A, Yang L, Dong HF, et al. Tannic acid is an inhibitor of CXCL12 (SDF-1alpha)/CXCR4 with antiangiogenic activity. Clin Cancer Res. 2003;9:3115-3123.  Back to cited text no. 196
    
197.
Li H, Wu J, Zhang Z, Ma Y, Liao F, Zhang Y, Wu G. Forsythoside a inhibits the avian infectious bronchitis virus in cell culture. Phytother Res. 2011;25:338-342.   Back to cited text no. 197
    
198.
Wang X, Li X, Wang X, Chen L, Ning E, Fan Y, et al. Experimental study of Forsythoside A on prevention and treatment of avian infectious bronchitis. Res Vet Sci 2020; in press (https://doi.org/10.1016/j.rvsc.2020.11.009).  Back to cited text no. 198
    
199.
Tang Y, Wang Z, Huo C, Guo X, Yang G, Wang M, Tian H, Hu Y, et al. Antiviral effects of Shuanghuanglian injection powder against influenza A virus H5N1 in vitro and in vivo. Microb Pathog. 2018;121:318-324.   Back to cited text no. 199
    
200.
Zhou L, Sun H, Song S, Liu J, Xia Z, Sun Y, Lyu Y. H3N2 canine influenza virus and Enterococcus faecalis coinfection in dogs in China. BMC Vet Res. 2019;15:113.   Back to cited text no. 200
    
201.
Shi L, Wu QG, Zhang JC, Yang GM, Liu W, Wang ZF. Mechanism of Shuang Huang Lian oral liquid for Treatment of Mycoplasma Pneumonia in Children on Network Pharmacology. Comb Chem High Throughput Screen. 2020;23:955-971.  Back to cited text no. 201
    
202.
Zhou F, Huang W, Li M, Zhong Y, Wang M, Lu B. Bioaccessibility and Absorption Mechanism of Phenylethanoid Glycosides Using Simulated Digestion/Caco-2 Intestinal Cell Models. J Agric Food Chem. 2018;66:4630-4637.   Back to cited text no. 202
    
203.
Zhou W, Zhu XX, Yin AL, Cai BC, Wang HD, Di L, et al. Effect of various absorption enhancers based on tight junctions on the intestinal absorption of forsythoside A in Shuang-Huang-Lian, application to its antivirus activity. Pharmacogn Mag. 2014;10:9-17.   Back to cited text no. 203
    
204.
Zhou W, Tan X, Shan J, Liu T, Cai B, Di L. Effect of chito-oligosaccharide on the intestinal absorptions of phenylethanoid glycosides in Fructus Forsythiae extract. Phytomedicine. 2014;21:1549-1558.   Back to cited text no. 204
    
205.
Liu T, Wang HD, Di LQ, Kang A, Zhao XL, Zhu XX, et al. [HPLC specific chromatogram spectrum-effect relationship for Shuanghuanglian on MDCK cell injury induced by influenza A virus (H1N1)]. Zhongguo Zhong Yao Za Zhi. 2015;40:4194-4199.  Back to cited text no. 205
    
206.
Wen W, He FY, Liu WL, Zhou YQ, Liu HY. [Screening of Shuanghuanglian Injection allergenic ingredients based on immune fingerprint]. Zhongguo Zhong Yao Za Zhi. 2019;44:1588-1595.   Back to cited text no. 206
    
207.
Deng L, Pang P, Zheng K, Nie J, Xu H, Wu S, et al. Forsythoside A Controls Influenza A Virus Infection and Improves the Prognosis by Inhibiting Virus Replication in Mice. Molecules. 2016;21:524.   Back to cited text no. 207
    
208.
Zeng XY, Yuan W, Zhou L, Wang SX, Xie Y, Fu YJ. Forsythoside A exerts an anti-endotoxin effect by blocking the LPS/TLR4 signaling pathway and inhibiting Tregs in vitro. Int J Mol Med. 2017;40:243-250.   Back to cited text no. 208
    
209.
Zheng X, Fu Y, Shi SS, Wu S, Yan Y, Xu L, et al. Effect of Forsythiaside A on the RLRs Signaling Pathway in the Lungs of Mice Infected with the Influenza A Virus FM1 Strain. Molecules. 2019;24:4219.   Back to cited text no. 209
    
210.
Liu JX, Li X, Yan FG, Pan QJ, Yang C, Wu MY, et al. Protective effect of forsythoside B against lipopolysaccharide-induced acute lung injury by attenuating the TLR4/NF-kappaB pathway. Int Immunopharmacol. 2019;66:336-346.   Back to cited text no. 210
    
211.
Song QJ, Weng XG, Cai DJ, Zhang W, Wang JF. Forsythoside A Inhibits BVDV Replication via TRAF2-Dependent CD28-4-1BB Signaling in Bovine PBMCs. PLoS One. 2016;11:e0162791.  Back to cited text no. 211
    
212.
Qu CC, Zhao GZ, Wang XP, Xu XL, Li B, Guo YH, et al. Tanreqing Injection for Patients with Influenza: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Chin J Integr Med. 2020;26:936-942.  Back to cited text no. 212
    
213.
Ding Y, Zeng L, Li R, Chen Q, Zhou B, Chen Q, et al. The Chinese prescription lianhuaqingwen capsule exerts anti-influenza activity through the inhibition of viral propagation and impacts immune function. BMC Complement Altern Med. 2017;17:130.   Back to cited text no. 213
    
214.
Wang CH, Zhong Y, Zhang Y, Liu JP, Wang YF, Jia WN, et al. A network analysis of the Chinese medicine Lianhua-Qingwen formula to identify its main effective components. Mol Biosyst. 2016;12:606-613.   Back to cited text no. 214
    
215.
Oz M, Lorke DE, Kabbani N. A comprehensive guide to the pharmacologic regulation of angiotensin converting enzyme 2 (ACE2), the SARS-CoV-2 entry receptor. Pharmacol Ther. 2020; Available online 1 December 2020, 107750 (https://doi.org/10.1016/j.pharmthera.2020.107750).  Back to cited text no. 215
    
216.
Yamamoto K, Takeshita H, Rakugi H. ACE2, angiotensin 1-7 and skeletal muscle: review in the era of COVID-19. Clin Sci (Lond). 2020;134:3047-3062.   Back to cited text no. 216
    
217.
Choi Y, Shin B, Kang K, Park S, Beck BR. Target-Centered Drug Repurposing Predictions of Human Angiotensin-Converting Enzyme 2 (ACE2) and Transmembrane Protease Serine Subtype 2 (TMPRSS2) Interacting Approved Drugs for Coronavirus Disease 2019 (COVID-19) Treatment through a Drug-Target Interaction Deep Learning Model. Viruses. 2020;12:1325.   Back to cited text no. 217
    
218.
Saponaro F, Rutigliano G, Sestito S, Bandini L, Storti B, Bizzarri R, Zucchi R. ACE2 in the Era of SARS-CoV-2: Controversies and Novel Perspectives. Front Mol Biosci. 2020;7:588618.   Back to cited text no. 218
    
219.
Mani JS, Johnson JB, Steel JC, Broszczak DA, Neilsen PM, Walsh KB, Naiker M. Natural product-derived phytochemicals as potential agents against coronaviruses: A review. Virus Res. 2020;284:197989.   Back to cited text no. 219
    
220.
Bhowmik D, Nandi R, Jagadeesan R, Kumar N, Prakash A, Kumar D. Identification of potential inhibitors against SARS-CoV-2 by targeting proteins responsible for envelope formation and virion assembly using docking based virtual screening, and pharmacokinetics approaches. Infect Genet Evol. 2020;84:104451.   Back to cited text no. 220
    
221.
Wijayasinghe YS, Bhansali P, Viola RE, Kamal MA, Poddar NK. Natural Products: A Rich Source of Antiviral Drug Lead Candidates for the Management of COVID-19. Curr Pharm Des. 2020. doi: 10.2174/1381612826666201118111151. Online ahead of print.  Back to cited text no. 221
    
222.
Elfiky AA. Natural products may interfere with SARS-CoV-2 attachment to the host cell. J Biomol Struct Dyn. 2020:1-10.   Back to cited text no. 222
    
223.
Adem Ş, Eyupoglu V, Sarfraz I, Rasul A, Zahoor AF, Ali M, et al. Caffeic acid derivatives (CAFDs) as inhibitors of SARS-CoV-2: CAFDs-based functional foods as a potential alternative approach to combat COVID-19. Phytomedicine. 2020:153310.   Back to cited text no. 223
    
224.
Attia YA, Alagawany MM, Farag MR, Alkhatib FM, Khafaga AF, Abdel-Moneim AE, et al. Phytogenic Products and Phytochemicals as a Candidate Strategy to Improve Tolerance to Coronavirus. Front Vet Sci. 2020;7:573159.   Back to cited text no. 224
    
225.
Paolacci S, Ceccarini MR, Codini M, Manara E, Tezzele S, Percio M, et al. Pilot study for the evaluation of safety profile of a potential inhibitor of SARS-CoV-2 endocytosis. Acta Biomed. 2020;91:e2020009.   Back to cited text no. 225
    
226.
Ergoren MC, Paolacci S, Manara E, Dautaj A, Dhuli K, Anpilogov K, et al. A pilot study on the preventative potential of alpha-cyclodextrin and hydroxytyrosol against SARS-CoV-2 transmission. Acta Biomed. 2020;91:e2020022.   Back to cited text no. 226
    
227.
Kallingal A, Thachan Kundil V, Ayyolath A, Karlapudi AP, Muringayil Joseph T, E JV. Molecular modeling study of tectoquinone and acteoside from Tectona grandis linn: a new SARS-CoV-2 main protease inhibitor against COVID-19. J Biomol Struct Dyn. 2020:1-12. doi: 10.1080/07391102.2020.1832580. Online ahead of print.  Back to cited text no. 227
    
228.
Li M, Xu T, Zhou F, Wang M, Song H, Xiao X, Lu B. Neuroprotective Effects of Four Phenylethanoid Glycosides on H2O2-Induced Apoptosis on PC12 Cells via the Nrf2/ARE Pathway. Int J Mol Sci. 2018;19:1135.   Back to cited text no. 228
    
229.
Ma T, Shi YL, Wang YL. Forsythiaside A protects against focal cerebral ischemic injury by mediating the activation of the Nrf2 and endoplasmic reticulum stress pathways. Mol Med Rep. 2019;20:1313-1320.   Back to cited text no. 229
    
230.
Huang C, Lin Y, Su H, Ye D. Forsythiaside protects against hydrogen peroxide-induced oxidative stress and apoptosis in PC12 cell. Neurochem Res. 2015;40:27-35.   Back to cited text no. 230
    
231.
Wang HQ, Xu YX, Zhu CQ. Upregulation of heme oxygenase-1 by acteoside through ERK and PI3 K/Akt pathway confer neuroprotection against beta-amyloid-induced neurotoxicity. Neurotox Res. 2012;21:368-378.   Back to cited text no. 231
    
232.
Seo ES, Oh BK, Pak JH, Yim SH, Gurunathan S, Kim YP, et al. Acteoside improves survival in cecal ligation and puncture-induced septic mice via blocking of high mobility group box 1 release. Mol Cells. 2013;35:348-354.   Back to cited text no. 232
    
233.
Wu L, Georgiev MI, Cao H, Nahar L, El-Seedi HR, Sarker SD, et al. Therapeutic potential of phenylethanoid glycosides: A systematic review. Med Res Rev. 2020;40:2605-2649.   Back to cited text no. 233
    
234.
Gong X, Xu Y, Ren K, Bai X, Zhang C, Li M. Phenylethanoid glycosides from Paraboea martinii protect rat pheochromocytoma (PC12) cells from hydrogen peroxide-induced cell injury. Biosci Biotechnol Biochem. 2019;83:2202-2212.   Back to cited text no. 234
    
235.
Sgarbossa A, Dal Bosco M, Pressi G, Cuzzocrea S, Dal Toso R, Menegazzi M. Phenylpropanoid glycosides from plant cell cultures induce heme oxygenase 1 gene expression in a human keratinocyte cell line by affecting the balance of NRF2 and BACH1 transcription factors. Chem Biol Interact. 2012;199:87-95.   Back to cited text no. 235
    
236.
Georgiev M, Pastore S, Lulli D, Alipieva K, Kostyuk V, Potapovich A, et al. Verbascum xanthophoeniceum-derived phenylethanoid glycosides are potent inhibitors of inflammatory chemokines in dormant and interferon-gamma-stimulated human keratinocytes. J Ethnopharmacol. 2012;144:754-760.   Back to cited text no. 236
    
237.
Daels-Rakotoarison DA, Seidel V, Gressier B, Brunet C, Tillequin F, Bailleul F, Luyckx M, et al. Neurosedative and antioxidant activities of phenylpropanoids from ballota nigra. Arzneimittelforschung. 2000;50:16-23.   Back to cited text no. 237
    
238.
Cheng L, Li F, Ma R, Hu X. Forsythiaside inhibits cigarette smoke-induced lung inflammation by activation of Nrf2 and inhibition of NF-kappaB. Int Immunopharmacol. 2015;28:494-499.   Back to cited text no. 238
    
239.
Lu T, Piao XL, Zhang Q, Wang D, Piao XS, Kim SW. Protective effects of Forsythia suspensa extract against oxidative stress induced by diquat in rats. Food Chem Toxicol. 2010;48:764-770.   Back to cited text no. 239
    
240.
Li X, Xie Y, Li K, Wu A, Xie H, Guo Q, et al. Antioxidation and Cytoprotection of Acteoside and Its Derivatives: Comparison and Mechanistic Chemistry. Molecules. 2018;23:498.   Back to cited text no. 240
    
241.
Qian J, Ma X, Xun Y, Pan L. Protective effect of forsythiaside A on OVA-induced asthma in mice. Eur J Pharmacol. 2017;812:250-255.   Back to cited text no. 241
    
242.
Gong L, Yu L, Gong X, Wang C, Hu N, Dai X, et al. Exploration of anti-inflammatory mechanism of forsythiaside A and forsythiaside B in CuSO(4)-induced inflammation in zebrafish by metabolomic and proteomic analyses. J Neuroinflammation. 2020;17:173.   Back to cited text no. 242
    
243.
Yi JM, Shin S, Kim NS, Bang OS. Ameliorative effects of aqueous extract of Forsythiae suspensa fruits on oxaliplatin-induced neurotoxicity in vitro and in vivo. BMC Complement Altern Med. 2019;19:339.   Back to cited text no. 243
    
244.
Yi JM, Shin S, Kim NS, Bang OS. Neuroprotective Effects of an Aqueous Extract of Forsythia viridissima and Its Major Constituents on Oxaliplatin-Induced Peripheral Neuropathy. Molecules. 2019;24:1177.   Back to cited text no. 244
    
245.
An N, Shi S. [The effect of forsythiaside on the expression of c-jun induced by cisplatin in the cochlea of guinea pig]. Lin Chung Er Bi Yan Hou Tou Jing Wai Ke Za Zhi. 2014;28:731-734.  Back to cited text no. 245
    
246.
Chen L, Lin L, Dong Z, Zhang L, Du H. Comparison of neuroprotective effect of Forsythia suspensa leaf extract and forsythiaside, one of its metabolites. Nat Prod Res. 2018;32:2705-2708.  Back to cited text no. 246
    
247.
Jiang WL, Tian JW, Fu FH, Zhu HB, Hou J. Neuroprotective efficacy and therapeutic window of Forsythoside B: in a rat model of cerebral ischemia and reperfusion injury. Eur J Pharmacol. 2010;640:75-81.   Back to cited text no. 247
    
248.
Kong F, Jiang X, Wang R, Zhai S, Zhang Y, Wang D. Forsythoside B attenuates memory impairment and neuroinflammation via inhibition on NF-kappaB signaling in Alzheimer's disease. J Neuroinflammation. 2020;17:305.   Back to cited text no. 248
    
249.
Wang Y, Zhao H, Lin C, Ren J, Zhang S. Forsythiaside A Exhibits Anti-inflammatory Effects in LPS-Stimulated BV2 Microglia Cells Through Activation of Nrf2/HO-1 Signaling Pathway. Neurochem Res. 2016;41:659-665.   Back to cited text no. 249
    
250.
Yan X, Chen T, Zhang L, Du H. Protective effects of Forsythoside A on amyloid beta-induced apoptosis in PC12 cells by downregulating acetylcholinesterase. Eur J Pharmacol. 2017;810:141-148.   Back to cited text no. 250
    
251.
Wang HM, Wang LW, Liu XM, Li CL, Xu SP, Farooq AD. Neuroprotective effects of forsythiaside on learning and memory deficits in senescence-accelerated mouse prone (SAMP8) mice. Pharmacol Biochem Behav. 2013;105:134-141.   Back to cited text no. 251
    
252.
Kim KS, Lee DS, Bae GS, Park SJ, Kang DG, Lee HS, et al. The inhibition of JNK MAPK and NF-κB signaling by tenuifoliside A isolated from Polygala tenuifolia in lipopolysaccharide-induced macrophages is associated with its anti-inflammatory effect. Eur J Pharmacol. 2013;721:267-276.  Back to cited text no. 252
    
253.
Qi Y, Dou DQ, Jiang H, Zhang BB, Qin WY, Kang K, et al. Arctigenin Attenuates Learning and Memory Deficits through PI3k/Akt/GSK-3beta Pathway Reducing Tau Hyperphosphorylation in Abeta-Induced AD Mice. Planta Med. 2017;83:51-56.   Back to cited text no. 253
    
254.
Kim SR, Kang SY, Lee KY, Kim SH, Markelonis GJ, Oh TH, et al. Anti-amnestic activity of E-p-methoxycinnamic acid from Scrophularia buergeriana. Brain Res Cogn Brain Res. 2003;17:454-461.   Back to cited text no. 254
    
255.
Kim MJ, Choi SJ, Lim ST, Kim HK, Heo HJ, Kim EK, et al. Ferulic acid supplementation prevents trimethyltin-induced cognitive deficits in mice. Biosci Biotechnol Biochem. 2007;71:1063-1068.   Back to cited text no. 255
    
256.
Rijal S, Changdar N, Kinra M, Kumar A, Nampoothiri M, Arora D, et al. Neuromodulatory potential of phenylpropanoids; para-methoxycinnamic acid and ethyl-p-methoxycinnamate on aluminum-induced memory deficit in rats. Toxicol Mech Methods. 2019;29:334-343.   Back to cited text no. 256
    
257.
Konar A, Kalra RS, Chaudhary A, Nayak A, Guruprasad KP, Satyamoorthy K, et al. Identification of Caffeic Acid Phenethyl Ester (CAPE) as a Potent Neurodifferentiating Natural Compound That Improves Cognitive and Physiological Functions in Animal Models of Neurodegenerative Diseases. Front Aging Neurosci. 2020;12:561925.   Back to cited text no. 257
    
258.
Luo C, Zou L, Sun H, Peng J, Gao C, Bao L, Ji R, Jin Y, Sun S. A Review of the Anti-Inflammatory Effects of Rosmarinic Acid on Inflammatory Diseases. Front Pharmacol. 2020;11:153.   Back to cited text no. 258
    
259.
Sang Z, Wang K, Han X, Cao M, Tan Z, Liu W. Design, Synthesis, and Evaluation of Novel Ferulic Acid Derivatives as Multi-Target-Directed Ligands for the Treatment of Alzheimer's Disease. ACS Chem Neurosci. 2019;10:1008-1024.   Back to cited text no. 259
    
260.
Chen L, Yan Y, Chen T, Zhang L, Gao X, Du C, et al. Forsythiaside prevents beta-amyloid-induced hippocampal slice injury by upregulating 2-arachidonoylglycerol via cannabinoid receptor 1-dependent NF-kappaB pathway. Neurochem Int. 2019;125:57-66.   Back to cited text no. 260
    
261.
Yan X, Chen T, Zhang L, Du H. Study of the interactions of forsythiaside and rutin with acetylcholinesterase (AChE). Int J Biol Macromol. 2018;119:1344-1352.   Back to cited text no. 261
    
262.
Jiang WL, Fu FH, Xu BM, Tian JW, Zhu HB, Jian-Hou. Cardioprotection with forsythoside B in rat myocardial ischemia-reperfusion injury: relation to inflammation response. Phytomedicine. 2010;17:635-639.   Back to cited text no. 262
    
263.
Martin-Nizard F, Sahpaz S, Furman C, Fruchart JC, Duriez P, Bailleul F. Natural phenylpropanoids protect endothelial cells against oxidized LDL-induced cytotoxicity. Planta Med. 2003;69:207-211.  Back to cited text no. 263
    
264.
Martin-Nizard F, Sahpaz S, Kandoussi A, Carpentier M, Fruchart JC, Duriez P, Bailleul F. Natural phenylpropanoids inhibit lipoprotein-induced endothelin-1 secretion by endothelial cells. J Pharm Pharmacol. 2004;56:1607-1611.   Back to cited text no. 264
    
265.
Seidel V, Verholle M, Malard Y, Tillequin F, Fruchart JC, Duriez P, et al. Phenylpropanoids from Ballota nigra L. inhibit in vitro LDL peroxidation. Phytother Res. 2000;14:93-98.   Back to cited text no. 265
    
266.
Huo X, Lu F, Qiao L, Li G, Zhang Y. A Component Formula of Chinese Medicine for Hypercholesterolemia Based on Virtual Screening and Biology Network. Evid Based Complement Alternat Med. 2018;2018:1854972.   Back to cited text no. 266
    
267.
Zhang Y, Miao H, Yan H, Sheng Y, Ji L. Hepatoprotective effect of Forsythiae Fructus water extract against carbon tetrachloride-induced liver fibrosis in mice. J Ethnopharmacol. 2018;218:27-34.   Back to cited text no. 267
    
268.
Kim HY, Kim JK, Choi JH, Jung JY, Oh WY, Kim DC, et al. Hepatoprotective effect of pinoresinol on carbon tetrachloride-induced hepatic damage in mice. J Pharmacol Sci. 2010;112:105-112.   Back to cited text no. 268
    
269.
Pan CW, Zhou GY, Chen WL, Zhuge L, Jin LX, Zheng Y, et al. Protective effect of forsythiaside A on lipopolysaccharide/d-galactosamine-induced liver injury. Int Immunopharmacol. 2015;26:80-85.   Back to cited text no. 269
    
270.
Lu C, Zheng SF, Liu J. Forsythiaside A alleviates renal damage in adriamycin-induced nephropathy. Front Biosci (Landmark Ed). 2020;25:526-535.  Back to cited text no. 270
    
271.
Shin HS, Park SY, Song HG, Hwang E, Lee DG, Yi TH. The Androgenic Alopecia Protective Effects of Forsythiaside-A and the Molecular Regulation in a Mouse Model. Phytother Res. 2015;29:870-876.   Back to cited text no. 271
    
272.
Zhang XT, Ding Y, Kang P, Zhang XY, Zhang T. Forsythoside A Modulates Zymosan-Induced Peritonitis in Mice. Molecules. 2018;23:593.   Back to cited text no. 272
    
273.
Lu Z, Yang H, Cao H, Huo C, Chen Y, Liu D, et al. Forsythoside A protects against lipopolysaccharide-induced acute lung injury through up-regulating microRNA-124. Clin Sci (Lond). 2020;134:2549-2563.   Back to cited text no. 273
    
274.
Lu ZB, Liu SH, Ou JY, Cao HH, Shi LZ, Liu DY, et al. Forsythoside A inhibits adhesion and migration of monocytes to type II alveolar epithelial cells in lipopolysaccharide-induced acute lung injury through upregulating miR-124. Toxicol Appl Pharmacol. 2020 Nov 15;407:115252. Corrigendum in Toxicol Appl Pharmacol. 2020:115328.  Back to cited text no. 274
    
275.
Liu C, Su H, Wan H, Qin Q, Wu X, Kong X, Lin N. Forsythoside A exerts antipyretic effect on yeast-induced pyrexia mice via inhibiting transient receptor potential vanilloid 1 function. Int J Biol Sci. 2017;13:65-75.   Back to cited text no. 275
    
276.
Jiang WL, Yong-Xu, Zhang SP, Zhu HB, Jian-Hou. Forsythoside B protects against experimental sepsis by modulating inflammatory factors. Phytother Res. 2012;26:981-987.   Back to cited text no. 276
    
277.
Zhang J, Zhang Y, Huang H, Zhang H, Lu W, Fu G, et al. Forsythoside A inhibited S. aureus stimulated inflammatory response in primary bovine mammary epithelial cells. Microb Pathog. 2018;116:158-163.   Back to cited text no. 277
    
278.
Sung YY, Yoon T, Jang S, Kim HK. Forsythia suspensa Suppresses House Dust Mite Extract-Induced Atopic Dermatitis in NC/Nga Mice. PLoS One. 2016;11:e0167687.   Back to cited text no. 278
    
279.
Zhang H, Sun X, Qi H, Ma Q, Zhou Q, Wang W, et al. Pharmacological Inhibition of the Temperature-Sensitive and Ca(2+)-Permeable Transient Receptor Potential Vanilloid TRPV3 Channel by Natural Forsythoside B Attenuates Pruritus and Cytotoxicity of Keratinocytes. J Pharmacol Exp Ther. 2019;368:21-31.   Back to cited text no. 279
    
280.
Sun X, Qi H, Wu H, Qu Y, Wang K. Anti-pruritic and anti-inflammatory effects of natural verbascoside through selective inhibition of temperature-sensitive Ca2+-permeable TRPV3 channel. J Dermatol Sci. 2020;97:229-231.   Back to cited text no. 280
    
281.
Han X, Piao XS, Zhang HY, Li PF, Yi JQ, Zhang Q, et al. Forsythia suspensa Extract Has the Potential to Substitute Antibiotic in Broiler Chicken. Asian-Aust J Anim Sci. 2012;25:569-576.  Back to cited text no. 281
    
282.
Long SF, He TF, Wu D, Yang M, Piao XS. Forsythia suspensa extract enhances performance via the improvement of nutrient digestibility, antioxidant status, anti-inflammatory function, and gut morphology in broilers. Poultry Sci. 2020;99:4217-4226.  Back to cited text no. 282
    
283.
Long SF, Wu D, He TF, Piao HX. Dietary supplementation with Forsythia suspensa extract during late gestation improves reproductive performance, colostrum composition, antioxidant status, immunoglobulin, and inflammatory cytokines in sows and newborn piglets. Animal Feed Sci Technol. 2021;271:114700.  Back to cited text no. 283
    
284.
Li H, Tang D, Qi C, Zhao X, Wang G, Zhang Y, Yu T. Forsythiaside inhibits bacterial adhesion on titanium alloy and attenuates Ti-induced activation of nuclear factor-kappaB signaling-mediated macrophage inflammation. J Orthop Surg Res. 2018;13:139.   Back to cited text no. 284
    
285.
Zhou W, Di LQ, Bi XL, Chen LT, Du Q. [Intestinal absorption of forsythoside A by rat circulation in situ]. Yao Xue Xue Bao. 2010;45:1373-1378.  Back to cited text no. 285
    
286.
Zhou W, Di L, Bi X, Chen L, Du Q. [Study on in situ intestinal absorption of active ingredients in Shuanghuanglian oral liquid in rats]. Zhongguo Zhong Yao Za Zhi. 2011;36:1733-1738.  Back to cited text no. 286
    
287.
Zhou W, Di LQ, Wang J, Shan JJ, Liu SJ, Ju WZ, Cai BC. Intestinal absorption of forsythoside A in in situ single-pass intestinal perfusion and in vitro Caco-2 cell models. Acta Pharmacol Sin. 2012;33:1069-1079.   Back to cited text no. 287
    
288.
Zhou W, Di LQ, Shan JJ, Bi XL, Chen LT, Wang LC. Intestinal absorption of forsythoside A in different compositions of Shuang-Huang-Lian. Fitoterapia. 2011;82:375-382.   Back to cited text no. 288
    
289.
Wu YT, Cai MT, Chang CW, Yen CC, Hsu MC. Bioanalytical Method Development Using Liquid Chromatography with Amperometric Detection for the Pharmacokinetic Evaluation of Forsythiaside in Rats. Molecules. 2016;21:1384.   Back to cited text no. 289
    
290.
Zhou W, Di LQ, Shan JJ, Bi XL, Chen LT, Wang LC, et al. In vitro metabolism in Sprague-Dawley rat liver microsomes of forsythoside A in different compositions of Shuang-Huang-Lian. Fitoterapia. 2011;82:1222-1230.   Back to cited text no. 290
    
291.
Cheng Y, Liang X, Feng L, Liu D, Qin M, Liu S, Liu G, Dong M. Effects of phillyrin and forsythoside A on rat cytochrome P450 activities in vivo and in vitro. Xenobiotica. 2017;47:297-303.   Back to cited text no. 291
    
292.
Zhou W, Qin KM, Shan JJ, Ju WZ, Liu SJ, Cai BC, Di LQ. Improvement of intestinal absorption of forsythoside A in weeping forsythia extract by various absorption enhancers based on tight junctions. Phytomedicine. 2012;20:47-58.   Back to cited text no. 292
    
293.
Zhou W, Wang H, Zhu X, Shan J, Yin A, Cai B, Di L. Improvement of intestinal absorption of forsythoside A and chlorogenic acid by different carboxymethyl chitosan and chito-oligosaccharide, application to Flos Lonicerae-Fructus Forsythiae herb couple preparations. PLoS One. 2013;8:e63348.   Back to cited text no. 293
    
294.
Zhou W, Tan X, Shan J, Wang S, Yin A, Cai B, Di L. Study on the main components interaction from Flos Lonicerae and Fructus Forsythiae and their dissolution in vitro and intestinal absorption in rats. PLoS One. 2014;9:e109619.   Back to cited text no. 294
    
295.
Zhou W, Tam KY, Meng M, Shan J, Wang S, Ju W, et al. Pharmacokinetics screening for multi-components absorbed in the rat plasma after oral administration of traditional Chinese medicine Flos Lonicerae Japonicae-Fructus Forsythiae herb couple by sequential negative and positive ionization ultra-high-performance liquid chromatography/tandem triple quadrupole mass spectrometric detection. J Chromatogr A. 2015;1376:84-97.   Back to cited text no. 295
    
296.
Wang F, Cao GS, Li Y, Xu LL, Wang ZB, Liu Y, et al. Characterization of forsythoside A metabolites in rats by a combination of UHPLC-LTQ-Orbitrap mass spectrometer with multiple data processing techniques. Biomed Chromatogr. 2018;32:e4164.   Back to cited text no. 296
    
297.
Xing S, Peng Y, Wang M, Chen D, Li X. In vitro human fecal microbial metabolism of Forsythoside A and biological activities of its metabolites. Fitoterapia. 2014;99:159-165.   Back to cited text no. 297
    
298.
Shi R, Xuan Z, Ma Y, Liu Y, Lu H, Sun T. Pharmacokinetics of forsythoside after intravenous administration in beagle dogs. Eur J Drug Metab Pharmacokinet. 2009;34:101-105.   Back to cited text no. 298
    
299.
Jiménez C, Riguera R. Phenylethanoid glycosides in plants: structure and biological activity. Nat Prod Rep. 1994;11:591-606.   Back to cited text no. 299
    
300.
Fu G, Pang H, Wong YH. Naturally occurring phenylethanoid glycosides: potential leads for new therapeutics. Curr Med Chem. 2008;15:2592-2613.  Back to cited text no. 300
    
301.
Xue Z, Yang B. Phenylethanoid Glycosides: Research Advances in Their Phytochemistry, Pharmacological Activity and Pharmacokinetics. Molecules. 2016;21:991.  Back to cited text no. 301
    
302.
Tian XY, Li MX, Lin T, Qiu Y, Zhu YT, Li XL, et al. A review on the structure and pharmacological activity of phenylethanoid glycosides. Eur J Med Chem. 2020;209:112563.   Back to cited text no. 302
    
303.
Tasneem S, Liu B, Li B, Choudhary MI, Wang W. Molecular pharmacology of inflammation: Medicinal plants as anti-inflammatory agents. Pharmacol Res. 2019;139:126-140.   Back to cited text no. 303
    
304.
Tang W, Shi QP, Ma T, Jiang XD, Liu SX, Wang YX. [Meta-analysis on incidence of adverse drug reaction induced by Shuanghuanglian injection]. Zhongguo Zhong Yao Za Zhi. 2016;41:2732-2742.   Back to cited text no. 304
    
305.
Yi Y, Zhang YS, Li CY, Zhao HY, Xiao HB, Li GQ, et al. [Study of screening pseudoallergenic substances of Shuanghuanglian injection]. Zhongguo Zhong Yao Za Zhi. 2015;40:2727-2731.  Back to cited text no. 305
    
306.
Yi Y, Liang AH, Li CY, Zhang YS, Zhao Y, Han JY, et al. [Influence of solvent and drug preparation time on Shuanghuanglian injections induce pseudo-allergic reaction]. Zhongguo Zhong Yao Za Zhi. 2015;40:2723-2726.  Back to cited text no. 306
    
307.
Han J, Zhao Y, Zhang Y, Li C, Yi Y, Pan C, et al. RhoA/ROCK Signaling Pathway Mediates Shuanghuanglian Injection-Induced Pseudo-allergic Reactions. Front Pharmacol. 2018;9:87.   Back to cited text no. 307
    
308.
Tan L, Li M, Lin Y. Safety Concerns of Traditional Chinese Medicine Injections Used in Chinese Children. Evid Based Complement Alternat Med. 2019;2019:8310368.   Back to cited text no. 308
    
309.
Ni L, Zhou L, Zhou M, Zhao J, Wang DW. Combination of western medicine and Chinese traditional patent medicine in treating a family case of COVID-19. Front Med. 2020;14:210-214.   Back to cited text no. 309
    
310.
Luo H, Tang QL, Shang YX, Liang SB, Yang M, Robinson N, Liu JP. Can Chinese Medicine Be Used for Prevention of Corona Virus Disease 2019 (COVID-19)? A Review of Historical Classics, Research Evidence and Current Prevention Programs. Chin J Integr Med. 2020;26:243-250.   Back to cited text no. 310
    
311.
Xiong X, Wang P, Su K, Cho WC, Xing Y. Chinese herbal medicine for coronavirus disease 2019: A systematic review and meta-analysis. Pharmacol Res. 2020;160:105056.  Back to cited text no. 311
    


    Figures

  [Figure 1], [Figure 2], [Figure 3], [Figure 4], [Figure 5], [Figure 6], [Figure 7], [Figure 8]
 
 
    Tables

  [Table 1], [Table 2], [Table 3]


This article has been cited by
1 Mucoadhesive chitosan/polyvinylpyrrolidone-co-poly (2-acrylamide-2-methylpropane sulphonic acid) based hydrogels of captopril with adjustable properties as sustained release carrier: Formulation design and toxicological evaluation
Rubina Qaiser, Fahad Pervaiz, Hina Shoukat, Haya Yasin, Hanasul Hanan, Ghulam Murtaza
Journal of Drug Delivery Science and Technology. 2023; : 104291
[Pubmed] | [DOI]
2 Virtual screening and molecular dynamics simulation analysis of Forsythoside A as a plant-derived inhibitor of SARS-CoV-2 3CLpro
Shabana Bibi, Muhammad Saad Khan, Sherif Aly El-Kafrawy, Thamir A. Alandijany, Mai Mohamed El-Daly, Qudsia Yousafi, Dua Fatima, Arwa A. Faizo, Leena H. Bajrai, Esam Ibraheem Azhar
Saudi Pharmaceutical Journal. 2022;
[Pubmed] | [DOI]
3 Synergistic deciphering of bioenergy production and electron transport characteristics to screen traditional Chinese medicine (TCM) for COVID-19 drug development
Po-Wei Tsai, Cheng-Yang Hsieh, Jasmine U. Ting, Yi-Ru Ciou, Chia-Jung Lee, Chieh-Lun Hsieh, Tzu-Kuan Lien, Chung-Chuan Hsueh, Bor-Yann Chen
Journal of the Taiwan Institute of Chemical Engineers. 2022; 135: 104365
[Pubmed] | [DOI]



 

Top
 
  Search
 
    Similar in PUBMED
   Search Pubmed for
   Search in Google Scholar for
 Related articles
    Access Statistics
    Email Alert *
    Add to My List *
* Registration required (free)  

 
  In this article
Abstract
Introduction
Molecular Divers...
Pharmacological ...
Metabolism of Fo...
Discussion
References
Article Figures
Article Tables

 Article Access Statistics
    Viewed4040    
    Printed126    
    Emailed0    
    PDF Downloaded465    
    Comments [Add]    
    Cited by others 3    

Recommend this journal