Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 Medicinal plants and their natural bioactive compounds provide significant and powerful resources, displaying diverse antiviral properties. Their biodiversity serves as an excellent source of new antiviral drugs, revealing new chemical structures + 12323 word(s) 12323 2020-10-07 18:43:34 |
2 format correct -5881 word(s) 6442 2020-11-30 11:42:45 | |
3 format correct + 3 word(s) 6445 2020-12-01 05:32:05 | |
4 format correct Meta information modification 6445 2020-12-01 07:29:17 | |
5 format correct Meta information modification 6445 2020-12-01 07:38:23 | |
6 format correct Meta information modification 6445 2020-12-01 07:39:15 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Siddiqui, A.J.; Danciu, C.; Ashraf, S.A.; Moin, A.; Singh, R.; Alreshidi, M.; Patel, M.; Jahan, S.; Kumar, S.; Alkhinjar, M.I.M.; et al. Medicinal Plants against SARS-CoV. Encyclopedia. Available online: https://encyclopedia.pub/entry/3277 (accessed on 29 March 2024).
Siddiqui AJ, Danciu C, Ashraf SA, Moin A, Singh R, Alreshidi M, et al. Medicinal Plants against SARS-CoV. Encyclopedia. Available at: https://encyclopedia.pub/entry/3277. Accessed March 29, 2024.
Siddiqui, Arif Jamal, Corina Danciu, Syed Amir Ashraf, Afrasim Moin, Ritu Singh, Mousa Alreshidi, Mitesh Patel, Sadaf Jahan, Sanjeev Kumar, Mulfi I. M. Alkhinjar, et al. "Medicinal Plants against SARS-CoV" Encyclopedia, https://encyclopedia.pub/entry/3277 (accessed March 29, 2024).
Siddiqui, A.J., Danciu, C., Ashraf, S.A., Moin, A., Singh, R., Alreshidi, M., Patel, M., Jahan, S., Kumar, S., Alkhinjar, M.I.M., Badraoui, R., Snoussi, M., & Adnan, M. (2020, November 30). Medicinal Plants against SARS-CoV. In Encyclopedia. https://encyclopedia.pub/entry/3277
Siddiqui, Arif Jamal, et al. "Medicinal Plants against SARS-CoV." Encyclopedia. Web. 30 November, 2020.
Medicinal Plants against SARS-CoV
Edit

SARS-CoV-2 infection (COVID-19) is in focus over all known human diseases, because it is destroying the world economy and social life, with increased mortality rate each day. To date, there is no specific medicine or vaccine available against this pandemic disease. However, the presence of medicinal plants and their bioactive molecules with antiviral properties might also be a successful strategy in order to develop therapeutic agents against SARS-CoV-2 infection. The identification of novel antiviral compounds is of critical significance, and medicinal plant based natural compounds are a good source for such discoveries. In depth search and analysis revealed several medicinal plants with excellent efficacy against SARS-CoV-1 and MERS-CoV, which are well-known to act on ACE-2 receptor, 3CLpro and other viral protein targets. It has consolidated the data of several medicinal plants and their natural bioactive metabolites, which have promising antiviral activities against coronaviruses with detailed modes of action/mechanism. It is concluded that this topics will be useful for researchers worldwide and highly recommended for the development of naturally safe and effective therapeutic drugs/agents against SARS-CoV-2 infection, which might be used in therapeutic protocols alone or in combination with chemically synthetized drugs.

SARS-CoV COVID-19 coronavirus medicinal plant phytomedicine ethnobotany antiviral natural products bioactive compounds

1. Introduction 

There are various medicinal plants which are known to have an inhibitory effect against SARS-CoV, HCoV-22E9, MERS-CoV and other viral infections. They were chosen specifically due to their mode of action and potency, and have been used and researched with ethnobotanical evidence against coronaviruses or other viruses (HIV, Influenza, etc.). Coronaviruses belong to positive sense RNA viruses and mostly use the ACE-2 (Angiotensin-converting enzyme-2) receptor, 3CLpro (3 Chymotrypsin-like protease), PLpro (Papain-like protease), RdRp (RNA-dependent RNA polymerase) enzyme and other known factors to gain entry into the human cell and complete the life cycle. Thereby, all these selected plants have been tested by various researchers globally to act on these specific target proteins and receptors, and, moreover, inhibit RNA replication in the other viruses too. This was the chief rationale in selecting these plants, which are described in detail below with their mode of action, which may also possibly be considered as a therapeutic choice against SARS-CoV-2. This presentation is designed in order to open new pathways towards the management of highly contagious diseases with the help of natural compounds.

2. Bupleurum Species

Bupleurum plant species are extensively dispersed in the northern hemisphere and are used as one of the oldest phytomedicines in China. Many reports have identified the activity of this herbal plant in the treatment of HCoV-22E9 and other viral infections [1][2]. Generally, Radix bupleuri (R. bupleuri) is derived from the dried roots of Bupleurum species and used for the treatment of various diseases [3]. It has great pharmacologically significant activities, the main ones reported in the literature being: antiviral, anti-inflammatory, anti-tumor, neuro-modulation and immunoregulation [3][4]. Approximately 7% of naturally occurring saikosaponins (triterpene saponin glycosides) are present in R. bupleuri, which is the main component of this medicinal plant with potent effects. Four types of saikosaponins (SS) are found; SSa, SSb2, SSc and SSd, which are responsible for the most pharmacological activities in this medicinal plant [5]. The SSa, SSb2 and SSd have potential to inhibit the effects against coronavirus 229E, SARS-CoV and influenza A virus [4]. Moreover, the mechanism of action of these SS employing antiviral activity interrupts the early stage of viral replication inside the host cells [4]. In addition, these SS also attenuate pro-inflammatory cytokines production, inhibiting viral replication through down-regulating NF-kB signaling, caspase 3-dependent virus ribonucleoprotein nuclear export, lung neutrophil and monocytes recruitment in an experimental in vivo mice model [4].

3. Lycoris radiate (L’Hér.) Herb.

Lycoris radiata (L. radiata) belongs to the Amaryllidaceae family and originally it was found in China, Korea, Japan and Nepal [6]. This medicinal plant has wide-ranging biological activities comprising: antiviral, anticancer [6], anti-malarial [7], anti-inflammatory [8] and induction of nausea and emesis [9]. Additionally, and most importantly, L. radiata has been known to have antiviral effects on SARS-CoV [10][11], poliovirus, human immunodeficiency virus (HIV), measles virus, herpes simplex virus and coxsackie virus [11][12]. Its potent bioactive compound is lycorine, which is extracted from the flower and stem cortex of L. radiata plants. Currently, this plant is in use for the treatment of various diseases due to its broad-spectrum biological activities. It has also been recommended as a promising medicinal plant for the development of potential drugs against SARS-CoV infection [11][12]. The antiviral mechanism of action of this plant is by inhibiting virus replication in the cells through inhibiting autophagy [12]. Moreover, JNK/MAPK signaling pathway is closely connected to autophagy, and through this signaling pathway, the plant extract inhibits the process of autophagy due to reduced JNK phosphorylation induced by viral replication [12][13].

4. Artemisia annua L.

This Chinese medicinal plant has been used for a long time to treat various diseases such as bronchitis and hemorrhoids, and is potentially effective for its anti-malarial, antiviral, anticancer, etc., properties [14][15][16]. However, Artemisia annua (A. annua) has been known to possess antiviral activity and currently is in use for the treatment of Poliovirus, HIV, RSV, HSV1, hepatitis C, type 2 dengue virus and human cytomegalovirus [17][18]. A. annua contains quercetine, flavonoid, polyphenols, triterpenes, sterols, saponins, polysaccharides, dicaffeoylquinic acid and other molecules [18]. Due to the presence of these molecules, A. annua extracts (whole plant) have shown an important role, being assigned with immunomodulator, antiviral, antioxidant and anti-inflammatory properties. Moreover, these compounds/molecules have been known to inhibit the enzyme activity of 3CLPro [19][20]. Previously, this medicinal plant has been used to treat SARS-CoV and MERS infections [20], and is currently being used against novel SARS-CoV-2 infection [19]. The mechanism of action of A. annua is to inhibit the enzymatic activity of 3CLPro, which is also produced by SARS-CoV-2, and increase the production of pro-inflammatory cytokines prostaglandin E2 (PGE2), IL-6, TNF-a, IFN-γ and enhance the genesis of CD4+ and CD8+ T cell populations [19][20][21].

5. Pyrrosia lingua (Thunb.) Farw.

Pyrrosia lingua (P. lingua) belongs to the Polypodiaceae family and mostly occurs in China, Japan, Korea and other Asian regions [22]. P. lingua is known for its antiviral, antioxidant, antibacterial and anticancer activities; it even stops the formation of urinary calculi [22][23]. Furthermore, it contains several bioactive components, such as flavonoids, chlorogenic acid, mangiferin, isomangiferin, astragalin and trifolin [24]. The extract of P. lingua leaves has been used by many researchers for the treatment of HIV, SARS and other viral infections [20][22][23]. In the case of SARS-CoV-1, this plant has shown the ability to inhibit viral infection, but the mechanism of action is still not clear [18][20].

6. Isatis indigotica Fortune ex Lindl.

Isatis indigotica (I. indigotica) is a very old Chinese herbal plant belonging to the Cruciferae family. It is mostly found in China, Hong Kong, Taiwan and other regions of Asia [25]. According to Lin et al., I. indigotica has the potential to inhibit/block SARS-CoV-1 entry and replication in its host [18]. However, the research group used Radix isatidis (dried root) of I. indigotica for extracting potent compounds for the treatment of SARS-CoV-1-infected patients. Furthermore, its root contains indirubin, indican, indigo, sinigrin, β-sitosterol, hesperetin, aloe-emodin and many more bioactive compounds [25][26]. According to one in vitro study, all these extracted compounds were used against SARS-CoV-1 infection, and it was found that indigo, sinigrin, aloe-emodin and hesperetin were able to inhibit the virus entry and replication by inhibiting the SARS-CoV-1 3CLpro [27]. We know that coronavirus 3CLpro mediates the proteolytic processing of replicase polypeptides into the functional proteins and plays a key role in viral replication [28]. Therefore, I. indigotica can also be considered as a potential therapeutic choice against SARS-CoV-2.

7. Torreya nucifera L.

This plant is mostly found in snowy areas near the Sea of Jeju Island in Korea, and is considered as a traditional medicinal plant. Its leaves are mostly used for the treatment of stomachache, hemorrhoids and rheumatoid arthritis [29][30]. During SARS-CoV-1 infection, Young Bae Ryu et al. used Torreya nucifera (T. nucifera) plant leaves for in vitro experiments, and the results showed a potential inhibitory effect [18][31]. Ryu et al. isolated 12 phytochemical compounds from the ethanol extract of the T. nucifera leaves. Only the biflavonoid amentoflavone showed efficacy against SARS-CoV-1 [31]. This biflavonoid of T. nucifera has the potential to block the activity of 3CLpro of the coronavirus and can inhibit the viral replication [31].

8. Houttuynia cordata Thunb.

This Southeast Asian plant belongs to the family of Saururaceae, which is traditionally used for the treatment of lung disorders such as cough, lung abscess, phlegm, and dyspnea. Houttuynia cordata Thorn (HCT) is a Chinese herbal plant well-known for its potent effects in the treatment of pneumonia, refractory hemoptysis, and SARS-CoV-1 infection [32][33]. It has anti-inflammatory, anti-allergic, antioxidant and anticancer properties [32]. The bioactive compounds present in HCT are comprised of rutin, hyperin, isoquercitrin, quercetin, afzelin, reyoutrin, kalium sulfuricum, cordarine, decanoyl acetaldehyde, lauric aldehyde, myrcene, α-pinene, methyl nonyl ketone, d-limonene, linoleic acid, aspartic acid, palmitic acid, water-soluble polysaccharides, amino acids, vitamins, manganese, potassium, zinc, iron and copper [34][35][36]. During the SARS-CoV-1 infection, the leaves of this Chinese medicinal plant were used to treat patients and showed good efficacy against SARS-CoV-1 [18][27]. Lau et al. conducted one experiment using HCT against SARS-CoV-1. Results showed that HCT can inhibit SARS-CoV-1 activity including an immunomodulatory effect [33]. However, the mode of action of HCT is to inhibit the 3CLpro activity of SARS-CoV-1 and obstruct the activity of RdRp [33]. Hence, it can block the entry of the virus and impede viral replication [33]. This inhibitory mechanism makes HCT a good choice to be used against SARS-CoV-2 infections.

9. Lindera aggregate (Sims) Kosterm.

Lindera aggregate(L. aggregate) is a traditional Chinese medicinal plant belonging to the Lauraceae family and mostly found in China and Japan [37]. The root of this plant is mostly used to treat chest pain, inflammation, indigestion, cold hernia and other diseases. It contains several bioactive components, such as flavonoids, isoquinoline alkaloids, sesquiterpene lactones and tannins [38][39]. Moreover, L. aggregate has also showed other biological activities such as antiviral, anti-tumor, anti-inflammatory, antimicrobial and anti-diabetic activities [18][39][40]. L. aggregate leaves can also be used to drink as tea, due to their protective effect against oxidative stress [40]. In 2005, Shi-you Li et al. investigated the effect and efficacy of L. aggregate roots against SARS-CoV-1 [20]. An in vitro study showed that L. aggregate is able to inhibit SARS-CoV-1 with EC50 value of 88.2 ± 7.7 µg/mL [20]. However, the mode of action is still not clear, but it was suggested that L. aggregate roots can possibly inhibit the viral replication and block the entry of virus [20].

10. Rheum palmatum L.

This herbal plant belongs to the family of Polygonaceae. It is mostly found in mountainous regions with high elevations, such as the Sichuan, Gansu and Shaanxi regions of China [41]. It is effectively used as a laxative or astringent for the treatment of stomachache, hemorrhoids, liver bile disease or gastroenteritis [42]. It contains some potent bioactive compounds including emodin, physcion, chrysophanol, rhein and aloe-emodin [43][44]. Known biological activities are antiviral, anti-pyretic, anti-neoplastic, anti-spasmolytic, antibacterial, laxative, hemostatic and anti-spasmodic [45][41][44][46]. It was also used against SARS-CoV-1 infection, due to its potential efficacy for acting on the ACE-2 receptor, leading to blockage of viral entry into cells and replication of the CoVs [47][43]. An in vitro study conducted by Ho et al. 2007 showed the potential of Radix et Rhizoma Rhei (root tubers of Rheum palmatum L.) in blocking the entry of SARS-CoV-1 to inhibition sites such as the ACE-2 receptor [47].

Furthermore, the major active component of this plant is emodin, which is responsible for blocking the binding of SARS-CoV-1 S protein to ACE-2 receptor [47][43]. Therefore, the use of emodin extracted from Radix et Rhizoma Rhei can be considered for the possible therapeutic management of COVID-19. This will possibly provide us with new insight into therapy against SARS-CoV-2.

11. Polygonum multiflorum Thunb.

Polygonum mulitflorum Thunb (PMT) is mostly found in China, Korea and Japan, belonging to Polygonaceae family [48]. Radix Polygoni multiflori (root tubers of PMT) is mostly used in treating many kinds of diseases, such as rubella, scrofula, waist and knee pain, paralysis, vaginal discharge, hypercholesterolemia (liver and kidney), malaria, and various other diseases, possessing neuro-protective, antioxidation, immunomodulation, anti-hyperlipidemia, anticancer, heap-toprotection, anti-inflammation, and anti-CoV functions [49][50][51]. The potent bioactive compounds present in PMT which are responsible for the therapeutic effects against various diseases are listed in Table 1 [52][53][54][55]. However, Ho et al. found that emodin is the most effective compound against SARS-CoV-1. The data were published to show the potential and efficacy of PMT in blocking the entry of SARS-CoV-1 by acting on the ACE-2 receptor [47]. The mode of action of PMT is similar to Rheum palmatum L and the major active constituent is found to be emodin in both plants.

Therefore, it is highly recommended to focus on emodin for possible and effective management of SARS-CoV-2 infection, combination with other therapeutic approaches.

12. Cerasus avium (L.) Moench.

This Persian medicinal plant belongs to the Rosaceae family and is mostly used as an antioxidant, antimicrobial and antiviral [56]. Its stem contains polyphenols, carotenoids, vitamins, minerals and many other bioactive components [57][58]. However, this plant has strong potential to act on the ACE-2 receptor and block the further processing of the viruses [47][59][60]. According to Ziai et al.’s 2009 in vitro study, this plant showed very good potential to inhibit or completely block the ACE-2 receptor [60]. Subsequently, Heidary et al., 2020 recently suggested that this plant has good potency against SARS-CoV-2 and must be used for the treatment of its infection [61].

13.  Alcea digitata (Boiss.) Alef.

Alcea digitata (A. digitate) is a Persian medicinal plant belonging to the Malvaceae family with antiviral, antioxidant, anti-inflammatory, antimicrobial, anti-tussive, expectorant and laxative therapeutic effects [62][63]. The flowers of A. digitata have been used for lung and respiratory disorders, head and neck cancer, and lubrication of the throat [63]. According to one published report [62], A. digitata is known to have good potential to block or inhibit the ACE-2 receptor. Recently, Heidary et al., 2020 suggested that A. digitata can possibly show good inhibitory effects against SARS-CoV-2 infection [61].

14. Citrus aurantium L.

Citrus aurantium (C. aurantium) belongs to the family of Rutaceae and is generally known as bitter orange [64]. This plant is known to have many essential components with biological effects [65][66], such as phenolics (flavanone glycosides, hydroxycinnamic acids), vitamin C, and carotenoids [99,249,250]. However, C. aurantium fruit extract is manly used for the treatment of anxiety, lung related diseases, obesity, gastrointestinal disorders and prostate cancer [64][67], buts
has potential to inhibit or block the ACE-2 receptor. Some in vitro studies have shown its efficacy in inhibiting ACE-2 receptors [61].

15. Rubia tinctorum L.

Rubia tinctorum (R. tinctorum) is mostly found in Southern Europe, Western Asia and North Africa and belongs to the family of Rubiaceae [68]. R. tinctorum is mostly used to treat kidney and bladder stones, and menstrual and urinary disorders [69][70]. Furthermore, the root of R. tinctorum contains red color due to the presence of anthraquinone, alizarin and pseudopurpurin, which is also used for dyeing purposes [71]. On the other hand, R. tinctorum has shown potential to inhibit or block the ACE-2 receptor [61]. in vitro studies revealed the efficient use of R. tinctorum to inhibit ACE-2
receptors [61].

16. Allium sativum L.

The common name of Allium sativum (A. sativum) is garlic, and it belongs to the Amaryllidaceae family. A. sativum use for human welfare has been reported for thousands of years in the form of a spice [72]. It is an aromatic herbaceous plant and is consumed worldwide as a food as well as a remedy for different diseases [72]. A. sativum is reported to have numerous
biological properties, such as antibacterial, antifungal, anti-carcinogenic, antioxidant, anti-diabetic, reno-protective, anti-atherosclerotic, and anti-hypertensive effects. Cloves of this traditional medicinal plant contain several potent components, such as alliin, allicin, ajoenes, vinyldithiins, and flavonoids [72][73][74][75][76], due to which it is mostly used for treatment of various disorders [77][78][72][73][79]. On the other hand, an in vitro study conducted by Ziai et al., 2009 on A. sativum and its potential efficacy to inhibit the ACE-2 receptor reported some effective results [60].

17. Quercus infectoria G. Olivier.

Quercus infectoria (Q. infectoria) is commonly known as gall oak and belongs to the family of Fagaceae [80]. This medicinal plant is traditionally used for the treatment of diarrhea, menorrhagia, dysentery, gonorrhea, tonsillitis, impetigo and internal hemorrhages [80][81]. Bioactive constituents of Q. infectoria gall extract include phenolic compounds (catechol, p hydroxybenzoic acid, caffeine, catechin, pyrogallol, e-vanillic acid, 3-hydroxytyrosol cinnamic, p-coumaric, gallic acids and resveratrol), flavonoids (naringin, rutin, 7-hydrohyflavone and hispertin) [82][83][84] with biological activities such as antiviral, antifungal, antibacterial, antioxidant, anti-inflammatory, anti-diabetic, anti-parasitic, anti-venom, etc. [80][85][86]. Q. infectoria has also shown strong potential to completely block the ACE-2 receptors due to the presence of many potent and tannin active components in vitro [87]. Similarly, this medicinal plant can also be considered for combinational therapeutic approaches in controlling the COVID-19 pandemic directly or indirectly.

18. Onopordum acanthium L.

Onopordum acanthium (O. acanthium) basically belongs to a family of Asteraceae and is commonly known as Scotch thistle [88]. It is found all over the world [89]. The biological activities of O. acanthium include antiviral, anti-tumor, anti-inflammatory and antioxidant effects. Extracts from the leaf, flower, stem and root of O. acanthium are also used as cardiotonic agents. O. acanthium contains many bioactive components, such as flavonoids, triterpenoids, lignans, phenylpropanoids, sesquiterpene lactones, and sterols [90][91][89]. Moreover, O. acanthium has shown efficacy to completely inhibit the activity of ACE-2 due to the presence of tannin bioactive components, as demonstrated by Sharifi et al., 2013 in his in vitro study. This makes it a considerable choice to test against SARS-CoV-2.

19. Berberis integerrima Bunge.

Berberis integerrima (B. integerrima) belongs to the family of Berberidaceae, with different parts of the plant showing different colors [92]. It is mostly found in Iran and contains many types of alkaloids [93]. Bioactive components extracted from the root of B. integerrima include berbamine, berberuin, palmatine, oxyacanthine, malic acid, ascorbic acid, caffeic acid, ursolic acid, coumarin, beta-carotene and tannin [94][93]. B. integerrima possesses many bioactive properties, such as antiviral, anti-inflammatory, anti-hyperglycemic, anti-hyperlipidemic, anticancer, and antioxidant effects, as well as being a liver protective agent [94][92][93]. Moreover, this medicinal plant was tested by Sharifi et al., 2013 in vitro. His team showed that the usage of a 330 µg/mL concentration of B. integerrima was able to inhibit the ACE-2 receptor due to 88.2 ± 1.7 IC50 [87]. Therefore, B. integerrima can be further investigated for its potent medicinal values and may provide fruitful results against SARS-CoV-2.

20. Crataegus microphylla C. Koch

This medicinal plant belongs to the family of Rosaceae and almost all parts of the plant are used for remedial purposes [95]. It is widely used for the treatment of many diseases, including heart muscle cells activation, coronary dilation, regulated blood flow, use as an antioxidant and anti-diabetic, and many others [96][95]. It contains flavonoids (phenols, phenolic acids, procyanidins, flavonoids, triterpenes, polysaccharides, catecho-lamines) which help in controlling/regulating various diseases [94][97][96][95]. Furthermore, it has also showed efficacy to inhibit the ACE-2 receptor and prohibit the entry of virus into the cell [87]. in vitro results suggested the use of a 330 µg/mL concentration of Crataegus microphylla was able to inhibit the virus binding to the ACE-2 receptor, and their IC50 was observed as 80.9 ± 1.3 [87]. The occurrence of some potential bioactive compounds in this medicinal plant and their efficacy against SARS-CoV-2 must be tested for better drug therapy to manage COVID-19.

21. Alnus japonica (Thunb.) Steud.

Alnus japonica (A. japonica) belongs to Betulaceae family and originally it was found in Japan, Korea, China and Russia [98]. This medicinal plant has wide range of biological activities comprising antiviral, anticancer, anti-inflammatory, and antioxidant effects, as well as the induction of lymphatic and gastroenteric disorders [98][99]. It is mostly used for the treatment of various diseases such as fever, cancer, and blood, lymphatic and gastroenteric disorders [100]. Additionally, and most importantly, A. japonica has been known to have an antiviral effect on SARS-CoV, and its potent bioactive compounds include hirsutenone, oregonin, rubranoside rubranoside B, rubranol, and hirsutanonol, which are extracted from the bark of the A. japonica plant [99]. These bioactive components have also been recommended as a promising medicinal plant for the development of potential drugs against SARS-CoV PLpro. In 2012, Park et al. investigated the effect and efficacy of A. japonica bark against SARS-CoV.

An in vitro study showed that A. japonica is able to inhibit the SARS-CoV PLpro with IC50 value ranging 21 of 41 from 3 to 44.5 µM of these compounds (hirsutenone, oregonin, rubranoside rubranoside B, rubranol, and hirsutanonol) [99]. However, the mode of action suggested that A. japonica bark can possibly inhibit the SARS-CoV PLpro activity.

22. Psoralea corylifolia L.

Psoralea corylifolia (P. corylifolia) belongs to the Leguminosae family and mostly occurs in India, China, Bangladesh, Indonesia, Malaysia, Sri Lanka and other Asian countries. P. corylifolia is known for its antiviral, antioxidant, antibacterial and anti-depressant activities [101][102]. Furthermore, it contains several potent bioactive components such as neoba-vaisoflavone, isobavachalcone, Bavachinin, 40–O-methyl bavachalcone, corylifol A, and psoralidin [103]. In 2014, Kim et al. investigated the effect of P. corylifolia seed extract and showed an imperative inhibitory effect of SARS-CoV PLpro, and their IC50 was 15 µg/mL [104]. Furthermore, all these bioactive components were tested by Kim et al., and the IC50 of these components against SARS-CoV PLpro was estimated to range between 4.2 to 38.4 µM. In addition, psoralidin and isobavachalcone showed the highest inhibitory activity against SARS-CoV PLpro, with IC50 of 4.2 ± 1.0 µM and 7.3± 0.8 µM, respectively [104].

23. Paulownia tomentosa (Thunb.) Steud.

Paulownia tomentosa (P. tomentosa) is an old Chinese medicinal plant belonging to the Scrophulariaceae family. It is mostly found in central and western China, Taiwan and Korea. P. tomentosa has wide-ranging biological activities comprising antiviral, antioxidant and antibacterial effects [105][106]. It is mostly used for the treatment of various diseases, such as inflammatory bronchitis, upper respiratory tract infection, asthma, tonsillitis, gonorrhea, traumatic bleeding, enteritis, bacteriological diarrhea, erysipelas, swelling, bronchopneumonia, conjunctivitis, and hemorrhoids [107][108]. Furthermore, P. tomentosa has been known to have an antiviral effect on SARS-CoV PLpro. P. tomentosa fruit contains many bioactive components such as tomentin A, tomentin B, tomentin C, tomentin D, tomentin E, geranylated flavonones and others [109].

In 2013, Cho et al. examined the effect and efficacy of P. tomentosa fruit-extracted bioactive components against SARS-CoV. An in vitro study showed that P. tomentosa is able to inhibit SARS-CoV PLpro activity with an IC50 value ranging from 5.0 to 14.4 µM [109]. Out of all those studied, Tomentin E showed the most promising and highest inhibitory effect against SARS-CoV, with the lowest IC50 5.0 ± 0.06 µM [109].

24. Tribulus terrestris L.

Tribulus terrestris (T. terrestris) is mostly found in China, India, Pakistan, South Americas, Bulgaria, Mexico and Spain, and is considered as a traditional medicinal plant. T. terrestris belongs to the Zygophyllaceae family and possesses several biological activities such as antiviral, anti-inflammatory, antioxidant, anti-tumor, anti-diabetic and anti-urolithic properties [110][111]. It contains several bioactive compounds, mainly flavonoids and alkaloids [112][113]. In 2014, Song et al. studied the effect of T. terrestris fruit extract (six cinnamic amides), and showed significant inhibitory effects against SARS-CoV PLpro [112]. Furthermore, all bioactive components tested by Song et al. against SARS-CoV PLpro were estimated to have an IC50 in a range between 15.8 and 70.1 µM [112]. However, terrestrimine[(E)-N-(1-hydroxy-2-(4-hydroxyphenyl)-2-oxoethyl)-3-(4-hydroxy3-methoxypheny) acrylamide] showed the utmost inhibitory activity against SARS-CoV PLpro with an IC50 of 15.8 ± 0.6 µM [112].

25. Sambucus nigra L.

This plant belongs to the family of Caprrifoliaceae and is mostly used in the treatment of common cold, HIV, HSV-1, influenza, urinary tract infection, edema and other rheumatic diseases [114][115]. It contains several active components extracted from the leaves, flower and fruit parts of the plant, such as flavonoids, lectins, anthocyanin, etc., which have been found to increase the immunity and inhibit the viral activity [116][117][118][119]. However, in case of the H1N1 influenza virus, this plant has shown great potential to block or impede the entry of the virus into the host cells [114][120][121]. Furthermore, the presence of lectins in this plant is responsible for controlling the symptoms or pathogenesis of the influenza virus [114][122]. It also has immunomodulating activity due to the presence of peptic polysaccharides, polyphenolic compounds and flavonoids [114]. Due to several significant and antiviral relevant properties of this plant, it can possibly be used against SARS-CoV-2.

26. Eleutherococcus senticosus (Rupr. & Maxim.) Maxim.

Eleutherococcus senticosus (E. senticosus) belongs to the family of Araliaceae and is mostly found in China, Japan and Korea [123]E. senticosus is used for the treatment of chronic coughing, ischemic heart disease, diabetes, cancer, altitude sickness, neurodegenerative disorders, and chronic fatigue [114][124][123][125]. Moreover, its leaves are used as food in the form of tea, wine, soups and many others [126]E. senticosus leaves have the potential efficacy to inhibit bacterial glucosidase activity, reported in in vitro results by many researchers. The nature of the component of E. senticosus responsible for its antiviral activity remains to be determined, and is currently under investigation together with the characterization of the target molecules and the molecular basis of the antiviral efficacy of E. senticosus. However, its extract is able to inhibit the replication of the influenza virus, and viral replication is common in all kinds of viruses [127][128][124][129]. Several potent bioactive components are known to be present in the roots of this medicinal plant, such as phenols, lignans, coumarins, phenylpropanoids, flavonoids, hyperin, rutin, afzelin, quercetin, kaempferol, phenolic acids, triterpenic acids, and anthocyanin, etc. Due to the presence of these bioactive compounds, in vitro results showed some antiviral activity too, by blocking the replication of influenza virus in the cells [114][130]. Therefore, it is a possible recommendation that plant should go further investigation and may be helpful in directly or indirectly controlling SARS-CoV-2.

27. Salvia miltiorrhiza Bunge

This plant belongs to the family of Lamiaceae and is commonly known as red sage [131]. Its bioactive components are extracted from the root, including lipophilic diterpenoids, flavonoids, triterpenoids and hydrophilic phenolic compounds [132][131][133]. It is also used for the treatment of various diseases, such as removing blood stasis, improving blood circulation, atherosclerosis, thrombosis, angina pectoris, other cardiovascular diseases and antiviral activity of HIV-1 and Enterovirus by inhibiting RdRp enzyme activity [134][135][132][136].

28. Acacia arabica (Lam.) Willd.

Acacia arabica (A. arabica) belongs to the family of Fabaceae and is widely distributed in Asian regions [137]. It is basically used for the treatment of various diseases, such as Newcastle disease, vaccinia virus, bursal disease virus, H9N2 influenza disease, skin diseases, and possesses many biological properties including antimicrobial, anti-diabetic, and antioxidant effects. The mechanism of action of A. arabica is known. It specifically inhibits the stage of viral intracellular multiplication [114][137][138][139]. Furthermore, A. arabica contains several bioactive components extracted from the leaves of the plant which are responsible for its bioactivity, such as flavonoids, methyl 3,4,5 tri hydroxyl benzoate, p-coumaroyl glucoside, p-coumaroylquinic acid, ferulic acid, isoferulic acid, epicatechin-3-gallate, ascorbic acid, quercetin 3-O-(4′-O-acetyl)-rhamnopyranoside, oleic acid, myristic acid, palmitic acid and steroidal sapogenin aglycone [140][137][138][141]. A. arabica also has the potential to inhibit the viral replication against HIV infection [114][139]. Due to its antiviral nature, it is highly recommended to use A. arabica for controlling/managing SARS-CoV-2 infection.

29. Ocimum sanctum L.

Ocimum sanctum (O. sanctum) belongs to the family of Lamiaceae and is commonly known as tulsi [137][142]. This aromatic plant is basically found in all Asian countries. It is used for the treatment of diseases such as cough, anxiety, arthritis, dysentery, diarrhea, asthma, fever, skin and eye disorders, otalgia, gastrointestinal disorders, cardiac and genitourinary disorders, back pain, snake, insect and scorpion bites, malaria, and H9N2 influenza disease [143][137][138][142][144]. The leaves of O. sanctum contain several bioactive compounds, such as alkanoids, saponins, tannins, flavonoids, phenols, anthocyanins and triterpenoids [137][145][142]. However, this medicinal plant has the potential to block the activity of different pathogens and can act as a potent antiviral, antifungal, anti-protozoan, anti-malarial, anti-helminthic, antibacterial, mosquito repellent, etc.; its other clinical activities are detailed in Table 1 [137][146][145][142][147][148][149][150]. Ghoke et al. showed that treatment with the crude extract derived from the leaves of O. sanctum leads to significant H9N2 virus reduction in assessing all three—virucidal, therapeutic and prophylactic—activities using an in vivo model. They suggested that the crude extract of O. sanctum could be a promising extract for developing safe and efficacious antiviral compounds against the H9N2 virus. The protecting effectiveness of the crude extract of O. sanctum might be ascribed to multiple mechanisms of action (specific inhibition of viral intracellular multiplication stage and non-specific interference with virus–cell interactions such as masking/blocking the HA glycoprotein [137]. Due to these vast known biological properties, it would be of great importance to study the potential particular active ingredient or combinations, which are responsible for its broader antiviral activity, further. Therefore, O. sanctum might be helpful for the treatment of COVID-19, and can potentially block the entry of virus as well as its replication.

30. Ocimum basilicum L.

This medicinal herb belongs to the family of Lamiaceae and it is also known as sweet basil [114][151]. It is mostly used in industries as food, perfumes and cosmetics [151] due to its potent antiviral, anti-inflammatory, antioxidant and antibacterial activities [152][151][153][154]. Moreover, it contains several bioactive components such as phenolic compounds, flavonoids and anthocyanin extracted from the whole plant of O. basilicum [155][153][154]. This herb has been used for HIV treatment and showed very good potential to inhibit the replication of the HIV virus, and blocks further viral processing [114][151].

31. Theobroma cacao L.

Theobroma cacao (T. cacao) belongs to the family of Sterculiaceae. The seeds of this cocoa plant are commonly used in food industries [156]. It contains several types of bioactive compounds, such as polyphenol, theobromine and flavonoids [157], which are responsible for its antioxidant, antiviral, anti-inflammatory and many other biological activities [158][159][160]. However, some studies reported the anti-influenza activity of T. cacao, due to presence of flavonoids, theobromine, lignin, dietary fiber, free fatty acid, and minerals (zinc, copper, iron) [158][161][159]. Kamei et al., 2014 investigated the effect of T. cacao against the influenza virus and found that it enhances the antibody response due to stimulatory effect [161]. Furthermore, it has also been observed that, T. cacao helps in developing acquired immunity and activates the NK cells against the influenza virus [161]. Further investigation may lead to the use of T. cacao against SARS-CoV-2, and can help in boosting immunity.

32. Pelargonium sidoides DC.

Pelargonium sidoides (P. sidoides) belongs to the family of Geraniaceae and is commonly known as Umckaloabo [162]. It is found all over the world and roots of this plant are traditionally used for remedial purposes against tuberculosis, respiratory diseases, cough, gastrointestinal infection, viral diseases and others [162][163]P. sidoides roots are known to have some potent compounds, such as methoxycoumarin, proanthocyanidins and prodelphinidins [164]. Furthermore, its roots are also used for the production of herbal drugs known as EPs 7639 by ethanolic extract, which have been approved for the treatment of respiratory tract infections [162][165]. According to Theisen et al., 2012, P. sidoides also has the potential to inhibit the viral entry of the influenza virus [165]. Therefore, it is suggested that roots of P. sidoides should be further investigated for the treatment of COVID-19.

33. Taraxacum officinale (L.) WEB. ex WIGG.

This medicinal plant belongs to the family of Asteraceae and it is commonly known as dandelion [18]. It is traditionally used for the treatment of various diseases such as kidney diseases, lung diseases, breast tumor, diabetes, uterus infections, digestive system related abnormalities, etc. [166][167]. Pharmacological research has proven the efficacy of this medicinal plant as antiviral, antibacterial, choleretic, anti-diabetic, anti-inflammatory, antioxidant, hepato-protective, diuretic and antifungal [166]. It contains several bioactive components, extracted from the aerial parts and roots, such as terpenes, flavonoids, phenolic compounds, terpenoids, triterpenoids, steroids, coumarins, phenols, saponins, flavones, flavonols, chalcones, phlobatannins, and cardiac glycosides [168][169][170]. Han et al., 2011 found the potential of this medicinal plant to inhibit the viral replication of HIV [171]. Similarly, Lee et al., 2012 also suggested its potential to enhance pro-inflammatory cytokines and improve the immune system [172]. Furthermore, it is also known to inhibit the influenza virus’ entry into cells [18][173]. Therefore, due to its vast and significant antiviral properties, it is highly recommended to conduct further investigation on this medicinal plant for the discovery of potent drugs against COVID-19.

34. Illicium oligandrum Merr & Chun

Illicium oligandrum (I. oligandrum) belongs to the family of Magnoliaceae, being a rich source of seco-prezizaane type sesquiterpenes [174]. It is known to have antiviral activity against herpes simplex virus type 2, coxsackie virus and influenza virus [175][176]. It has some potent bioactive compounds, such as sesquiterpene lactones, neolignan glycosides, phenolic diglycosides and prenylated compounds which are responsible for its antiviral activities [175][176]. However, this medicinal plant is also used for the treatment of rheumatoid arthritis, and neurotoxic and neuro-trophic effects [177]. Ma et al., 2013 reported the ethanolic extraction of spirooliganones A 1 & B from the roots of I. oligandrum and showed its potential to inhibit the activity of influenza virus (H3N2) (IC50 3.70–33.33 µM) and coxsackie virus B3 [175].

35. Glycyrrhiza glabra L.

Glycyrrhiza glabra (Liquorice) belongs to the family of Fabaceae and is among the most ancient medicinal plants [178]. It has several very well-known biological activities, such as antiviral (HIV, SARS-CoV), anti-inflammatory, antimicrobial, antioxidant, anti-tumorigenic and anti-ulcer properties [179][180]. The root of Liquorice is known to have many bioactive components, including flavonoids, glycyrrhizic acid, triterpenoid, saponins, etc. [180][178]. Few studies showed that chalcones extracted from Liquorice have the ability to block or inhibit the activity of influenza virus [18][181]. Therefore, there is a possibility that this plant might be useful against SARS-CoV-2 due to its antiviral properties.

36. Angelica keiskei (Miq.) Koidz.

Angelica keiskei (A. keiskei) belongs to the family of Umbelliferae, and its leaves are basically used for remedial purposes [182]. Its bioactive components include chalcones, flavanones and coumarins, coumarins phenolic, acetylenes, sesquiterpene, diterpene, and triterpenes [183][184]A. keiskei is known and considered to be antiviral, anticancer, anti-inflammatory, anti-obesity, anti-oxidative, anti-coagulant, anti-tumor, anti-mutagenic, anti-diabetic, antibacterial and hepato-protective [183][185][184]. Park et al., 2016 extracted bioactive components (9 alkylated chalcones and 4 coumarins) from A. keiskei plant [184], and revealed that the extracted chalcones were able to significantly block the entry of coronavirus (SARS-CoV-1) by inhibiting the chymotrypsin-like protease (75% inhibition using 30 µg/mL dose) and a papain-like protease (88% inhibition using 30 µg/mL dose) [184]. In addition, the IC50 of this chalcone and chalcone 6 are 11.4 and 1.2 µM, respectively [184]. Therefore, due to this very specific inhibition property of A. keiskei deserves further investigation for the development of potent antiviral agents against COVID-19.

37. Polygala karensium Kurz

Polygala karensium (P. karensium) is a medicinal plant belonging to the family of Polygalaceae and can mostly be found in China, Myanmar, Thailand, and Vietnam [186]. It has important and potent bioactive compounds, i.e., xanthones, which have shown many biological activities such as antiviral, antimicrobial, antioxidant, cytotoxicity, etc. [187][186][188]. In addition, it is also used to treat various ailments such as cough, bronchitis, neurasthenia, inflammation and amnesia [186]. However, Dao et al., 2012 conducted one in vitro study on H1N1, H9N2, and novel H1N1 (WT) strains using ethanol-extracted xanthones from the root of P. karensium, and found that xanthones have the potential to completely inhibit influenza virus activity [187]. Therefore, xanthones from P. karensium can also be one of the choices worth investigation for the further development of phytomedicine against SARS-CoV-2.

38. Calophyllum brasiliense Cambess.

Calophyllum brasiliense (C. brasiliense) is a medicinal plant, and basically belongs to the family of Clusiaceae, mostly found in South America, Central America and the Caribbean region [189]. It is used as a remedy to treat several diseases, such as parasitic, viral, bacterial and fungal diseases [189][190]. Its potent biological activities include antiviral, antibacterial, anti-protozoal and antifungal effects [189][191]. However, Kudo et al., 2013 investigated the role of this medicinal plant in HIV disease, firstly by extracting tricyclic coumarin from the leaves of C. brasiliense and testing in vitro [190]. Hence, they revealed that tricyclic coumarin from C. Brasiliense possesses great potential to inhibit viral replication by blocking the NFkB pathway [190].

39. Cimicifuga foetida L.

Cimicifuga foetida (C. foetida) belongs to the family of Ranunculaceae and it is also known as Shengma. It is abundantly distributed in Asian region [192]. It is basically used to treat various ailments, such as fever, headache, sore throat, toothache, uterine prolapse and inflammation [193]. It contains several bioactive compounds extracted from rhizomes, including cycloartane triterpenoids and glycosides with antiviral, anti-tumor, anti-inflammatory activities [192]. Wang et al., 2012 investigated the role of C. foetida, especially the cimicifugin component of it, against Respiratory Syncytial Virus, and found that the plant has a strong potential to inhibit viral attachment and internalization [194][193]. Moreover, cimicifugin was also able to stimulate epithelial cells and initiate the secretion of cytokines such as IFN-β, to clear the viral infection/load [195]. Furthermore, another in vitro study conducted by Dai et al., 2016 observed the potential of C. foetida in inhibiting the hepatitis B virus transcription and replication by producing pro-inflammatory cytokines [196]. Due to the capacity of producing strong pro-inflammatory cytokines and immunomodulatory properties, C. foetida can be used to treat COVID-19 disease.

40. Boerhavia diffusa L.

Boerhavia diffusa (B. diffusa) belongs to the family of Nyctaginaceae and is commonly known as punarnava [197]. It is mostly found in Asian countries and is basically used for the treatment of various diseases, such as abdominal pain, jaundice, dyspepsia, stress, spleen enlargement and liver diseases [198]B. diffusa bioactive components extracted from leaf, stem and root include flavonoids, triterpenoids, alkaloids, hypoxanthine, steroids, lipids, lignin, proteins, ursolic acid, boeravinone, punarnavoside, etc. [199][200]. However, Bose et al., 2017 suggested that B. diffusa has a strong potential to inhibit the entry of hepatitis C virus and its major compound (boeravinone H component) were able to block the initial phase of HCV entry through acting directly on the viral particles [198]. Moreover, Manu et al., 2007 also showed that its second major bioactive compound (Punarnavine) was also able to enhance the immune response, especially IFN-γ and interleukin-2 cytokines [201]. This categorizes B. diffusa as a therapeutically important plant to be considered under the current circumstances of the COVID-19 pandemic and worth further investigation.

41. Terminalia chebula Retz

Terminalia chebula (T. chebula) belongs to the family of Combretaceae and is mostly found in the Asian region [202]. It is one of the most important medicinal plants due to the presence of a huge number of different kinds of phytoconstituents [203]. It is customarily used as a household remedy and also in modern, Ayurveda, Unani and Homoeopathic medicines [203][202]. Its bioactive components extracted from the leaves, bark and fruit of the plant include flavonoids, polyphenols, terpenes, anthocyanins, glycosides, gallic acid, chebulagic acid, punicalagin, chebulanin, corilagin, neochebulinic acid, ellagic acid, chebulinic acid, alkaloids and many more [203][204][205]. It is also known to be used as a cure for irregular fevers, urinary diseases, diabetes, skin diseases, heart diseases, constipation, ulcers, vomiting, colic pain, hemorrhoids, digestive diseases, and others [203][206][204]. However, T. chebula has many pharmacological activities such as antiviral, antioxidant, antibacterial, antifungal, anti-protozoal, anti-carcinogenic, anti-mutagenic, radio-protective, chemo-preventive, hepato-protective, cardio-protective, cyto-protective, anti-diabetic, reno-protective, anti-inflammatory, anti-arthritic, adaptogenic, anti-anaphylactic, hypolipidemic, hypocholesterolemic, anti-caries, wound healing, anti-allergic, immunomodulatory, anti-ulcer, anti-spasmodic and gastrointestinal motility properties [203][207][206][208][204]. Lin et al., 2013 conducted an in vitro study and found that chebulagic acid and punicalagin from the fruit of T. chebula have the potential to inhibit the activity of different viruses, such as human cytomegalovirus, HCV, dengue virus, measles virus, and respiratory syncytial virus [209]. Due to its strong biological properties, it is highly recommended to study T. chebula as a possible remedy against SARS-CoV-2.

42. Caesalpinia sappan L.

Caesalpinia sappan (C. sappan) belongs to the family of Caesalpiniaceae and it is usually known as Brazil or Sappan wood [191]. It is mostly found in Southeast Asian regions and is traditionally used for the treatment of various diseases such as tuberculosis, diarrhea, dysentery, skin infections, anemia, etc. [191][210]C. sappan is effectively considered as an antiviral, anti-inflammatory, antioxidant, antibacterial, antifungal, and anti-complementary [191][210][211]. Its bioactive constituents include xanthone, coumarin, chalcones, flavones, homoisoflavonoids, and brazilin [210]. Tewtrakul et al., 2015, extracted nine compounds from the roots of C. sappan. The results showed that, out of those nine, sappanchalcone (IC50 2.3 µM) and protosappanin A (IC50 12.6 µM) presented the strongest effect against HIV-1 IN [210]. On the other hand, Liu et al., 2009 also investigated the role of this medicinal plant against influenza virus. The in vitro study showed that 3-deoxysappanchalcone and sappanchalcone component isolated from C. sappan exhibited the highest activity against influenza virus (H3N2), with IC50 1.06 and 2.06 μg/mL, respectively [212]. Therefore, sappanchalcone from C. sappan should also be considered for further examination against SARS-CoV-2.

References

  1. Yao, R.-Y.; Zou, Y.-F.; Chen, X.-F. Traditional use, pharmacology, toxicology, and quality control of species in genus Bupleurum L. Chin. Herb. Med. 2013, 5, 245–255.
  2. Cheng, P.-W.; Ng, L.-T.; Chiang, L.-C.; Lin, C.-C.; Ng, L.-T. Antiviral effects of saikosaponins on human coronavirus 229E in vitro. Clin. Exp. Pharmacol. Physiol. 2006, 33, 612–616.
  3. Yang, F.; Dong, X.; Yin, X.; Wang, W.; You, L.; Ni, J. Radix bupleuri: A review of traditional uses, botany, phytochemistry, pharmacology, and toxicology. BioMed Res. Int. 2017, 2017, 1–22.
  4. Chen, J.; Duan, M.; Zhao, Y.; Ling, F.; Xiao, K.; Li, Q.; Li, B.; Lu, C.; Qi, W.; Zeng, Z.; et al. Saikosaponin A inhibits influenza A virus replication and lung immunopathology. Oncotarget 2015, 6, 42541–42556.
  5. Tykheev, Z.A.; Taraskin, V.V.; Radnaeva, L.D.; Zhang, F.Q.; Chen, S.L. Total saikosaponin content in some species of Bupleurum L. IOP Conf. Ser. Earth Environ. Sci. 2019, 320, 012055.
  6. Lamoral-Theys, D.; Decaestecker, C.; Mathieu, V.; Dubois, J.; Kornienko, A.; Kiss, R.; Evidente, A.; Pottier, L. Lycorine and its derivatives for anticancer drug design. Mini Rev. Med. Chem. 2010, 10, 41–50.
  7. Cedrón, J.C.; Gutiérrez, D.G.; Flores, N.; Ravelo, Á.G.; Estévez-Braun, A. Synthesis and antiplasmodial activity of lycorine derivatives. Bioorg. Med. Chem. 2010, 18, 4694–4701.
  8. Mikami, M.; Kitahara, M.; Kitano, M.; Ariki, Y.; Mimaki, Y.; Sashida, Y.; Yamazaki, M.; Yui, S. Suppressive activity of lycoricidinol (narciclasine) against cytotoxicity of neutrophil-derived calprotectin, and its suppressive effect on rat adjuvant arthritis model. Biol. Pharm. Bull. 1999, 22, 674–678.
  9. Kretzing, S.; Abraham, G.; Seiwert, B.; Ungemach, F.R.; Krügel, U.; Regenthal, R. Dose-dependent emetic effects of the Amaryllidaceous alkaloid lycorine in beagle dogs. Toxicon 2011, 57, 117–124.
  10. Ieven, M.; Berghe, D.A.V.D.; Vlietinck, A.J. Plant antiviral agents. IV. Influence of lycorine on growth pattern of three animal viruses. Planta Med. 1983, 49, 109–114.
  11. Liu, J.-N.; Yang, Y.; Xu, Y.; Ma, C.; Qin, C.; Zhang, L. Lycorine reduces mortality of human enterovirus 71-infected mice by inhibiting virus replication. Virol. J. 2011, 8, 483.
  12. Wang, H.; Guo, T.; Yang, Y.; Yu, L.; Pan, X.; Li, Y.-H. Lycorine derivative LY-55 inhibits EV71 and CVA16 replication through downregulating autophagy. Front. Microbiol. 2019, 9, 277.
  13. Mukhtar, M.; Arshad, M.; Ahmad, M.; Pomerantz, R.J.; Wigdahl, B.; Parveen, Z. Antiviral potentials of medicinal plants. Virus Res. 2008, 131, 111–120.
  14. Efferth, T.; Romero, M.R.; Wolf, D.G.; Stamminger, T.; Marin, J.J.G.; Marschall, M. The antiviral activities of artemisinin and artesunate. Clin. Infect. Dis. 2008, 47, 804–811.
  15. AleSaeidi, S.; Miraj, S. A systematic review of anti-malarial properties, immunosuppressive properties, anti-inflammatory properties, and anti-cancer properties of artemisia annua. Electron. Physician 2016, 8, 3150–3155.
  16. Ho, W.E.; Peh, H.Y.; Chan, T.K.; Wong, W.F. Artemisinins: Pharmacological actions beyond anti-malarial. Pharmacol. Ther. 2014, 142, 126–139.
  17. Arunkumar, G.; Mudgal, P.P.; Maity, H.; Dowarha, D.; Devadiga, S.; Nag, S.; Arunkumar, G. Herbal plants and plant preparations as remedial approach for viral diseases. Virusdisease 2015, 26, 225–236.
  18. Lin, L.-T.; Hsu, W.-C.; Lin, C.-C. Antiviral natural products and herbal medicines. J. Tradit. Complement. Med. 2014, 4, 24–35.
  19. Law, S.; Leung, A.W.; Xu, C. Is the traditional Chinese herb “Artemisia annua” possible to fight against COVID-19? Integr. Med. Res. 2020, 9, 100474.
  20. Li, S.-Y.; Chen, C.; Zhang, H.-Q.; Guo, H.-Y.; Wang, H.; Wang, L.; Zhang, X.; Hua, S.-N.; Yu, J.; Xiao, P.-G.; et al. Identification of natural compounds with antiviral activities against SARS-associated coronavirus. Antivir. Res. 2005, 67, 18–23.
  21. Benatouil, C.P.; Reanimator, A. Action of Artemisia Annua on Adaptive Immunity in COVID-19 Infections. Available online: https://lavierebelle.org/action-de-l-artemisia-annua-sur-l?lang=en (accessed on 22 June 2020).
  22. Gao, D.; Fan, Y.; Feng, H.; Liu, L.; Zhang, Y.; Xin, X. Chemical components and antibacterial activity of the essential oil of six pyrrosia species. Chem. Biodivers. 2020, 10, 1–10.
  23. Zheng, M. Experimental study of 472 herbs with antiviral action against the herpes simplex virus. Chin. J. Mod. Dev. Tradit. Med. 1990, 10, 39–41.
  24. Xiao, W.; Peng, Y.; Tan, Z.; Lv, Q.; Chan, C.-O.; Yang, J.; Chen, S. Comparative evaluation of chemical profiles of pyrrosiae folium originating from three pyrrosia species by HPLC-DAD combined with multivariate statistical analysis. Molecules 2017, 22, 2122.
  25. Chen, Y.; Fan, C.-L.; Wang, Y.; Zhang, X.-Q.; Huang, X.-J.; Ye, W.-C. Chemical constituents from roots of Isatis indigotica. China J. Chin. Mater. Med. 2018, 43, 2091–2096.
  26. Zhang, D.; Shi, Y.; Xu, R.; Du, K.; Guo, F.; Chen, K.; Li, Y.; Wang, R. Alkaloid enantiomers from the roots of isatis indigotica. Molecules 2019, 24, 3140.
  27. Lin, C.-W.; Tsai, F.-J.; Tsai, C.-H.; Lai, C.-C.; Wan, L.; Ho, T.-Y.; Hsieh, C.-C.; Chao, P.-D.L. Anti-SARS coronavirus 3C-like protease effects of Isatis indigotica root and plant-derived phenolic compounds. Antivir. Res. 2005, 68, 36–42.
  28. Chang, S.-J.; Chang, Y.-C.; Lu, K.-Z.; Tsou, Y.-Y.; Lin, C.-W. Antiviral activity of isatis indigotica extract and its derived indirubin against japanese encephalitis virus. Evid. Based Complement. Altern. Med. 2012, 2012, 1–7.
  29. Oh, J.; Rho, H.S.; Yang, Y.; Yoon, J.Y.; Lee, J.; Hong, Y.D.; Kim, H.C.; Choi, S.S.; Kim, T.W.; Shin, S.S.; et al. Extracellular signal-regulated kinase is a direct target of the anti-inflammatory compound amentoflavone derived from Torreya nucifera. Mediat. Inflamm. 2013, 2013, 1–11.
  30. Endo, Y.; Osada, Y.; Kimura, F.; Shirakawa, H.; Fujimoto, K. Effects of Japanese Torreya (Torreya nucifera) seed oil on the activities and mRNA expression of lipid metabolism-related enzymes in rats. Biosci. Biotechnol. Biochem. 2007, 71, 231–233.
  31. Ryu, Y.B.; Jeong, H.J.; Kim, J.H.; Kim, Y.M.; Park, J.-Y.; Kim, D.; Naguyen, T.T.H.; Park, S.-J.; Chang, J.S.; Park, K.H.; et al. Biflavonoids from Torreya nucifera displaying SARS-CoV 3CLpro inhibition. Bioorg. Med. Chem. 2010, 18, 7940–7947.
  32. Shingnaisui, K.; Dey, T.; Manna, P.; Kalita, J. Therapeutic potentials of Houttuynia cordata Thunb. against inflammation and oxidative stress: A review. J. Ethnopharmacol. 2018, 220, 35–43.
  33. Lau, K.-M.; Lee, K.-M.; Koon, C.-M.; Cheung, C.S.-F.; Lau, C.-P.; Ho, H.-M.; Lee, M.Y.-H.; Au, S.W.-N.; Cheng, C.H.K.; Lau, C.B.; et al. Immunomodulatory and anti-SARS activities of Houttuynia cordata. J. Ethnopharmacol. 2008, 118, 79–85.
  34. Chiow, K.; Phoon, M.; Putti, T.; Tan, B.K.; Chow, V.T.K. Evaluation of antiviral activities of Houttuynia cordata Thunb. extract, quercetin, quercetrin and cinanserin on murine coronavirus and dengue virus infection. Asian Pac. J. Trop. Med. 2016, 9, 1–7.
  35. Cheng, D.; Sun, L.; Zou, S.; Chen, J.; Mao, H.-Y.; Zhang, Y.; Liao, N.; Zhang, R.-H. Antiviral effects of Houttuynia cordata polysaccharide extract on Murine Norovirus-1 (MNV-1)—A human norovirus surrogate. Molecules 2019, 24, 1835.
  36. Chen, M.-Y.; Li, H.; Lu, X.-X.; Ling, L.-J.; Weng, H.-B.; Sun, W.; Chen, D.-F.; Zhang, Y.-Y. Houttuynia cordata polysaccharide alleviated intestinal injury and modulated intestinal microbiota in H1N1 virus infected mice. Chin. J. Nat. Med. 2019, 17, 187–197.
  37. Xu, C.; Yang, B.-X.; Zhu, W.; Li, X.; Tian, J.; Zhang, L. Characterisation of polyphenol constituents of Linderae aggregate leaves using HPLC fingerprint analysis and their antioxidant activities. Food Chem. 2015, 186, 83–89.
  38. Wei, G.; Chen, H.; Nie, F.; Ma, X.; Jiang, H. 1, 3, 6-trihydroxy-7-methyl-9, 10-anthracenedione isolated from genus lindera with anti-cancer activity. Anticancer Agents Med. Chem. 2017, 17, 1604–1607.
  39. Xiao, M.; Cao, N.; Fan, J.-J.; Shen, Y.; Xu, Q. Studies on flavonoids from the leaves of Lindera aggregata. J. Chin. Med. Mater. 2011, 34, 62–64.
  40. Jung, S.-H.; Han, J.-H.; Park, H.-S.; Lee, J.-J.; Yang, S.Y.; Kim, Y.; Heo, K.-S.; Myung, C.-S. Inhibition of collagen-induced platelet aggregation by the secobutanolide secolincomolide a from lindera obtusiloba blume. Front. Pharmacol. 2017, 8, 560.
  41. Zhao, M.-J.; Chao, J.; Dai, Y.-T.; Chen, S.-L.; Li, Q.; Fan, Z.-Q.; Wang, D.-D. Quality evaluation of Rhei Radix et Rhizoma decoction. China J. Chin. Mater. Med. 2018, 43, 861–867.
  42. Su, B.; Li, X.-B. Advance in studies on effect of Glycyrrhizae Radix et Rhizoma in relieving purgative activity of Rhei Radix et Rhizoma. China J. Chin. Mater. Med. 2015, 40, 577–581.
  43. Yang, K.L.; Gao, Y.; Yang, F.W.; Liu, M.; Shi, S.Z.; Chen, Y.M.; Zhang, J.H.; Tian, J.H. Analysis of traditional Chinese medicine from patent information sharing platform of coronavirus disease 2019 (COVID-19). China J. Chin. Mater. Med. 2020, 45, 3001–3006.
  44. Zheng, L.; Chen, S.; Cao, Y.; Zhao, L.; Gao, Y.; Ding, X.; Wang, X.; Gu, Y.; Wang, S.; Zhu, Z.; et al. Combination of comprehensive two-dimensional prostate cancer cell membrane chromatographic system and network pharmacology for characterizing membrane binding active components from Radix et Rhizoma Rhei and their targets. J. Chromatogr. A 2018, 1564, 145–154.
  45. Lee, J.-C.; Tseng, C.-K.; Wu, S.-F.; Chang, F.-R.; Chiu, C.-C.; Wu, Y.-C. San-Huang-Xie-Xin-Tang extract suppresses hepatitis C virus replication and virus-induced cyclooxygenase-2 expression. J. Viral Hepat. 2011, 18, e315–e324.
  46. Wei, Y.; Liu, M.; Liu, J.; Li, H. Influence factors on the hepatotoxicity of polygoni multiflori Radix. Evid. Based Complement. Altern. Med. 2019, 2019, 5482896.
  47. Ho, T.-Y.; Wu, S.-L.; Chen, J.-C.; Li, C.-C.; Hsiang, C.-Y. Emodin blocks the SARS coronavirus spike protein and angiotensin-converting enzyme 2 interaction. Antivir. Res. 2007, 74, 92–101.
  48. Kim, Y.-J.; Lee, J.Y.; Kim, H.-J.; Kim, D.-H.; Lee, T.H.; Kang, M.S.; Choi, Y.-K.; Lee, H.L.; Kim, J.; An, H.-J.; et al. Inhibitory effect of emodin on raw 264.7 activated with double stranded rna analogue poly I:C. Afr. J. Tradit. Complement. Altern. Med. 2017, 14, 157–166.
  49. Lee, B.-J.; Lee, K. Discrimination and proper use of polygoni multiflori radix, cynanchi wilfordii radix, and cynanchi auriculati radix in Korea: A descriptive review. Evid. Based Complement. Altern. Med. 2015, 2015, 1–7.
  50. Liang, Z.T.; Chen, H.; Yu, Z.-L.; Zhao, Z.-Z. Comparison of raw and processed Radix Polygoni Multiflori (Heshouwu) by high performance liquid chromatography and mass spectrometry. Chin. Med. 2010, 5, 29.
  51. Liang, L.; Xu, J.; Liang, Z.-T.; Dong, X.-P.; Chen, H.; Zhao, Z.-Z. Tissue-specific analysis of secondary metabolites creates a reliable morphological criterion for quality grading of polygoni multiflori Radix. Molecules 2018, 23, 1115.
  52. Cheng, W.; Li, Y.; Yang, W.; Wu, S.; Wei, M.; Gao, Y.; Kang, C.; Zhang, S.; Li, Y. Simultaneous determination of 13 constituents of radix polygoni multiflori in rat plasma and its application in a pharmacokinetic study. Int. J. Anal. Chem. 2020, 2020, 4508374.
  53. He, Q.; Tu, C.; Wang, J.-B.; Liu, Z.-J.; Sha, M.-C.; Zhang, L.; Li, C.-Y.; Xiao, X.-H. Antiplatelet aggregation bioactivity of Polygoni Multiflori Radix with chemical fingerprints and spectrum-effect correlation analysis. China J. Chin. Mater. Med. 2017, 42, 1679–1684.
  54. Lin, L.; Ni, B.; Lin, H.; Zhang, M.; Yan, L.; Qu, C.; Ni, J. Simultaneous determination of 14 constituents of Radix polygoni multiflori from different geographical areas by liquid chromatography-tandem mass spectrometry. Biomed. Chromatogr. 2014, 29, 1048–1055.
  55. Budak, N.H. Bioactive components of Prunus avium L. black gold (red cherry) and Prunus avium L. stark gold (white cherry) juices, wines and vinegars. J. Food Sci. Technol. 2016, 54, 62–70.
  56. Serteser, A.; Kargioglu, M.; Gök, V.; Bağci, Y.; Özcan, M.M.; Arslan, D. Determination of antioxidant effects of some plant species wild growing in Turkey. Int. J. Food Sci. Nutr. 2008, 59, 643–651.
  57. Console, L.; Giangregorio, N.; Cellamare, S.; Bolognino, I.; Palasciano, M.; Indiveri, C.; Incampo, G.; Campana, S.; Tonazzi, A.; Lara, C.; et al. Human mitochondrial carnitine acylcarnitine carrier: Molecular target of dietary bioactive polyphenols from sweet cherry (Prunus avium L.). Chem. Interact. 2019, 307, 179–185.
  58. Shen, C.-Y.; Jiang, J.-G.; Zhu, W.; Ou-Yang, Q. Anti-inflammatory effect of essential oil from Citrus aurantium L. var. amara Engl. J. Agric. Food Chem. 2017, 65, 8586–8594.
  59. Kouchmeshky, A.; Jameie, S.B.; Amin, G.; Ziai, S.A. Investigation of angiotensin-convertings enzyme inhibitory effects of medicinal plants used in traditional persian medicine for treatment of hypertension: Screening study. Thrita Stud. J. Med. Sci. 2012, 1, 13–23.
  60. Ziai, S.A.; Heidari, M.R.; Amin, G.H.; Koochemeshki, A.; Heidari, M. Inhibitory effects of germinal angiotensin converting enzyme by medicinal plants used in iranian traditional medicine as antihypertensive. J. Kerman Univ. Med Sci. 2009, 16, 134.
  61. Heidary, F.; Varnaseri, M.; Gharebaghi, R. The potential use of persian herbal medicines against COVID-19 through angiotensin-converting enzyme. Arch. Clin. Infect. Dis. 2020, 15, e102838.
  62. Ameri, A.; Heydarirad, G.; Rezaeizadeh, H.; Choopani, R.; Ghobadi, A.; Gachkar, L. Evaluation of efficacy of an herbal compound on dry mouth in patients with head and neck cancers. J. Evid. Based Integr. Med. 2015, 21, 30–33.
  63. Nasser, R.; Jafari, F.; Rezaeizadeh, H.; Nasseri, M.; Kamalinejad, M.; Ghobadi, A.; Shamsipour, M.; Zargaran, A.; Ameri, A. Efficacy of a persian medicine herbal compound (alcea digitataalefandmalva sylvestrisl.) on prevention of radiation induced acutemucositis in patients with head and neck cancer: A pilot study. Int. J. Cancer Manag. 2017, 10, e8642.
  64. Stohs, S.J. Safety, efficacy, and mechanistic studies regarding citrus aurantium (bitter orange) extract andp-synephrine. Phytother. Res. 2017, 31, 1463–1474.
  65. Süntar, I.; Khan, H.; Patel, S.; Celano, R.; Rastrelli, L. An overview on Citrus aurantium L.: Its functions as food ingredient and therapeutic agent. Oxidative Med. Cell. Longev. 2018, 2018, 1–12.
  66. Zhao, H.-Y.; Yang, L.; Wei, J.; Huang, M.; Jiang, J.-G. Bioactivity evaluations of ingredients extracted from the flowers of Citrus aurantium L. var. amara Engl. Food Chem. 2012, 135, 2175–2181.
  67. Marhoume, F.Z.; Laaradia, M.A.; Zaid, Y.; Laadraoui, J.; Oufquir, S.; Aboufatima, R.; Chait, A.; Bagri, A.; Zaid, Y.; Oufkir, S. Anti-aggregant effect of butanolic extract of Rubia tinctorum L. on platelets in vitro and ex vivo. J. Ethnopharmacol. 2019, 241, 111971.
  68. Xiong, Y.; Yang, Y.; Xiong, W.; Yao, Y.; Wu, H.; Zhang, M. Network pharmacology-based research on the active component and mechanism of the antihepatoma effect of Rubia cordifolia L. J. Cell. Biochem. 2019, 120, 12461–12472.
  69. Nejad, H.E. Ahmad esalat nejad. Rubia tinctorum L. (Rubiaceae) or madder as one of the living color to dyeing wool. Int. J. Adv. Biol. Biomed. Res. 2013, 1, 1315–1319.
  70. Shang, A.; Cao, S.-Y.; Xu, X.-Y.; Gan, R.-Y.; Tang, G.-Y.; Corke, H.; Mavumengwana, V.; Li, H.-B. Bioactive compounds and biological functions of garlic (Allium sativum L.). Foods 2019, 8, 246.
  71. Lajkó, E.; Bányai, P.; Zámbó, Z.; Kursinszki, L.; Szőke, É.; Kohidai, L. Targeted tumor therapy by Rubia tinctorum L.: Analytical characterization of hydroxyanthraquinones and investigation of their selective cytotoxic, adhesion and migration modulator effects on melanoma cell lines (A2058 and HT168-M1). Cancer Cell Int. 2015, 15, 1–15.
  72. Batiha, G.E.-S.; Beshbishy, A.M.; Wasef, L.; Elewa, Y.H.A.; Abdel-Daim, M.; El-Hack, M.; Taha, A.E.; Abd-Elhakim, Y.M.; Devkota, H.P. Chemical constituents and pharmacological activities of garlic (Allium sativum L.): A review. Nutrients 2020, 12, 872.
  73. Phan, A.D.; Netzel, G.; Chhim, P.; Netzel, M.E.; Sultanbawa, Y. Phytochemical characteristics and antimicrobial activity of australian grown garlic (Allium sativum L.) cultivars. Foods 2019, 8, 358.
  74. Martins, N.; Petropoulos, S.A.; Ferreira, I.C. Chemical composition and bioactive compounds of garlic (Allium sativum L.) as affected by pre- and post-harvest conditions: A review. Food Chem. 2016, 211, 41–50.
  75. Kim, S.; Kim, D.-B.; Jin, W.; Park, J.; Yoon, W.; Lee, Y.; Kim, S.; Lee, S.; Kim, S.; Lee, O.-H.; et al. Comparative studies of bioactive organosulphur compounds and antioxidant activities in garlic (Allium sativum L.), elephant garlic (Allium ampeloprasum L.) and onion (Allium cepa L.). Nat. Prod. Res. 2017, 32, 1193–1197.
  76. Chavan, R.D.; Shinde, P.; Girkar, K.; Madage, R.; Chowdhary, A. Assessment of anti-influenza activity and hemagglutination inhibition of plumbago indica and Allium sativum extracts. Pharmacogn. Res. 2016, 8, 105–111.
  77. Baek, S.C.; Nam, K.H.; Yi, S.; Jo, M.S.; Lee, K.H.; Lee, Y.H.; Lee, J.; Kim, K.H. Anti-adipogenic effect of β-carboline alkaloids from garlic (Allium sativum). Foods 2019, 8, 673.
  78. Burian, J.P.; Carlos, I.Z.; Sacramento, L.V.S. Fungal infection control by garlic extracts (Allium sativum L.) and modulation of peritoneal macrophages activity in murine model of sporotrichosis. Braz. J. Biol. 2017, 77, 848–855.
  79. Tayel, A.A.; El-Sedfy, M.A.; Ibrahim, A.I.; Moussa, S.H. Application of Quercus infectoria extract as a natural antimicrobial agent for chicken egg decontamination. Rev. Argent. Microbiol. 2018, 50, 391–397.
  80. Chusri, S.; Phatthalung, P.N.; Voravuthikunchai, S. Anti-biofilm activity of Quercus infectoria G. Olivier against methicillin-resistant Staphylococcus aureus. Lett. Appl. Microbiol. 2012, 54, 511–517.
  81. Ahmed, A.; Salih, F.A. Quercus infectoria gall extracts reduce quorum sensing-controlled virulence factors production and biofilm formation in Pseudomonas aeruginosa recovered from burn wounds. BMC Complement. Altern. Med. 2019, 19, 177.
  82. Chokpaisarn, J.; Urao, N.; Voravuthikunchai, S.P.; Koh, T.J. Quercus infectoria inhibits Set7/NF-κB inflammatory pathway in macrophages exposed to a diabetic environment. Cytokine 2017, 94, 29–36.
  83. Motamedi, H.; Azizi, A.; Ahmadi, M. Nutritive value of treated Quercus infectoria and Quercus libani leaves with the tannin-degrading bacterium Klebsiella pneumoniae for ruminant feeding in vitro. J. Appl. Microbiol. 2019, 127, 1339–1348.
  84. Kheirandish, F.; Delfan, B.; Mahmoudvand, H.; Moradi, N.; Ezatpour, B.; Ebrahimzadeh, F.; Rashidipour, M. Antileishmanial, antioxidant, and cytotoxic activities of Quercus infectoria Olivier extract. Biomed. Pharmacother. 2016, 82, 208–215.
  85. Shrestha, S.; Kaushik, V.S.; Eshwarappa, R.S.B.; Subaramaihha, S.R.; Ramanna, L.M.; Lakkappa, D.B.; Prasad, S.B.B. Pharmacognostic studies of insect gall of Quercus infectoria Olivier (Fagaceae). Asian Pac. J. Trop. Biomed. 2014, 4, 35–39.
  86. Qaderi, M.M.; Cavers, P.B.; Bernards, M.A. Isolation and structural characterization of a water-soluble germination inhibitor from Scotch thistle (Onopordum acanthium) cypselas. J. Chem. Ecol. 2003, 29, 2425–2438.
  87. Sharifi, N.; Souri, E.; Ziai, S.A.; Amin, G.; Amanlou, M. Discovery of new angiotensin converting enzyme (ACE) inhibitors from medicinal plants to treat hypertension using an in vitro assay. Daru 2013, 21, 74.
  88. Robertovna, G.E.; Alexeevich, K.D.; Alexeevich, S.A.; Petrovna, G.M.; Kenzhebaevna, O.K.; Garsiya, E.R.; Konovalov, D.A.; Шамилoв, A.A.; Glushko, M.P.; Орынбасарoва, K.K. A traditional medicine plant, Onopordum acanthium L. (asteraceae): Chemical composition and pharmacological research. Plants 2019, 8, 40.
  89. Tung, N.H.; Kwon, H.-J.; Kim, J.-H.; Ra, J.C.; Ding, Y.; Kim, J.A.; Kim, Y. Anti-influenza diarylheptanoids from the bark of Alnus japonica. Bioorg. Med. Chem. Lett. 2010, 20, 1000–1003.
  90. Abusamra, Y.A.-K.; Scuruchi, M.; Habibatni, S.; Maammeri, Z.; Benayache, S.; D’Ascola, A.; Avenoso, A.; Campo, G.M.; Spina, E. Evaluation of putative cytotoxic activity of crude extracts from Onopordum acanthium leaves and Spartium junceum flowers against the U-373 glioblastoma cell line. Pak. J. Pharm. Sci. 2015, 28, 1225–1232.
  91. Csupor-Löffler, B.; Zupkó, I.; Molnár, J.; Forgo, P.; Hohmann, J. Bioactivity-guided isolation of antiproliferative compounds from the roots of Onopordum acanthium. Nat. Prod. Commun. 2014, 9, 337–340.
  92. Rafiee, F.; Nejati, V.; Heidari, R.; Ashraf, H. Department of biology, faculty of science, urmia university, urmia, iran protective effect of methanolic extract of berberis integerrima bunge. root on carbon tetrachloride-induced testicular injury in wistar rats. Int. J. Reprod. Biomed. 2016, 14, 133–140.
  93. Sabahi, Z.; Farmani, F.; Soltani, F.; Moein, M. DNA protection, antioxidant and xanthine oxidase inhibition activities of polyphenol-enriched fraction of Berberis integerrima Bunge fruits. Iran. J. Basic Med. Sci 2018, 21, 411–416.
  94. Kooch, Y.; Noghre, N. The effect of shrubland and grassland vegetation types on soil fauna and flora activities in a mountainous semi-arid landscape of Iran. Sci. Total. Environ. 2020, 703, 135497.
  95. Renda, G.; Ozel, A.; Barut, B.; Korkmaz, B.; Yaylı, N. The in vitro protection by Crataegus microphylla extracts against oxidative damage and enzyme inhibition effects. Turk. J. Pharm. Sci. 2018, 15, 77–84.
  96. Melikoğlu, G.; Bitis, L.; Meriçli, A.H. Flavonoids of crataegus microphylla. Nat. Prod. Res. 2004, 18, 211–213.
  97. Hosseinimehr, S.J.; Mahmoudzadeh, A.; Azadbakht, M.; Akhlaghpoor, S.; Azadbakht, M. Radioprotective effects of hawthorn against genotoxicity induced by gamma irradiation in human blood lymphocytes. Radiat. Environ. Biophys. 2008, 48, 95–98.
  98. Tung, N.H.; Kwon, H.-J.; Kim, J.-H.; Ra, J.C.; Kim, J.A.; Kim, Y. An anti-influenza component of the bark of Alnus japonica. Arch. Pharmacal Res. 2010, 33, 363–367.
  99. Park, J.-Y.; Jeong, H.J.; Kim, J.H.; Kim, Y.M.; Park, S.-J.; Kim, M.; Park, K.H.; Lee, W.S.; Ryu, Y.B. Diarylheptanoids from Alnus japonica inhibit papain-like protease of severe acute respiratory syndrome coronavirus. Biol. Pharm. Bull. 2012, 35, 2036–2042.
  100. Won, T.H.; Song, I.-H.; Kim, K.-H.; Yang, W.-Y.; Lee, S.K.; Oh, D.-C.; Oh, W.K.; Oh, K.-B.; Shin, J. Bioactive metabolites from the fruits of psoralea corylifolia. J. Nat. Prod. 2015, 78, 666–673.
  101. Alam, F.; Khan, G.N.; Bin Asad, M.H.H. Psoralea corylifolia L: Ethnobotanical, biological, and chemical aspects: A review. Phytother. Res. 2017, 32, 597–615.
  102. Chopra, B.; Dhingra, A.K.; Dhar, K.L. Psoralea corylifolia L. (Buguchi)—Folklore to modern evidence: Review. Fitoterapia 2013, 90, 44–56.
  103. Schneiderová, K.; Šlapetová, T.; Hrabal, R.; Dvorakova, H.; Prochazkova, P.; Novotna, J.; Urbanova, M.; Cvačka, J.; Smejkal, K. Tomentomimulol and mimulone B: Two new C geranylated flavonoids from Paulownia tomentosa fruits. Nat. Prod. Res. 2013, 27, 613–618.
  104. Kim, D.W.; Seo, K.H.; Curtis-Long, M.J.; Oh, K.Y.; Oh, J.-W.; Cho, J.K.; Lee, K.H.; Park, K.H. Phenolic phytochemical displaying SARS-CoV papain-like protease inhibition from the seeds of Psoralea corylifolia. J. Enzym. Inhib. Med. Chem. 2013, 29, 59–63.
  105. Ali, S.A.; Ibrahim, N.A.; Mohammed, M.M.; El-Hawary, S.; Refaat, E.A. The potential chemo preventive effect of ursolic acid isolated from Paulownia tomentosa, against N-diethylnitrosamine: Initiated and promoted hepatocarcinogenesis. Heliyon 2019, 5, 01769.
  106. Schneiderová, K.; Smejkal, K. Phytochemical profile of Paulownia tomentosa (Thunb). steud. Phytochem. Rev. 2014, 14, 799–833.
  107. Zima, A.; Hošek, J.; Treml, J.; Muselík, J.; Suchy, P.; Pražanová, G.; Lopes, A.; Žemlička, M. Antiradical and cytoprotective activities of several C-geranyl-substituted flavanones from Paulownia tomentosa fruit. Molecules 2010, 15, 6035–6049.
  108. Tian, C.; Zhang, Z.; Wang, H.; Guo, Y.; Zhao, J.; Liu, M. Extraction technology, component analysis, and in vitro antioxidant and antibacterial activities of total flavonoids and fatty acids from Tribulus terrestris L. fruits. Biomed. Chromatogr. 2019, 33, e4474.
  109. Cho, J.K.; Curtis-Long, M.J.; Lee, K.H.; Kim, D.W.; Ryu, H.W.; Yuk, H.J.; Park, K.H. Geranylated flavonoids displaying SARS-CoV papain-like protease inhibition from the fruits of Paulownia tomentosa. Bioorg. Med. Chem. 2013, 21, 3051–3057.
  110. Reshma, P.L.; Lekshmi, V.S.; Sankar, V.; Raghu, K.G. Tribulus terrestris (Linn.) attenuates cellular alterations induced by ischemia in H9c2 cells via antioxidant potential. Phytother. Res. 2015, 29, 933–943.
  111. Stefanescu, R.; Tero-Vescan, A.; Negroiu, A.; Aurică, E.; Vari, C. A comprehensive review of the phytochemical, pharmacological, and toxicological properties of Tribulus terrestris L. Biomolecules 2020, 10, 752.
  112. Song, Y.H.; Kim, D.W.; Curtis-Long, M.J.; Yuk, H.J.; Wang, Y.; Zhuang, N.; Lee, K.H.; Jeon, K.S.; Park, K.H. Papain-Like Protease (PLpro) inhibitory effects of cinnamic amides from Tribulus terrestris fruits. Biol. Pharm. Bull. 2014, 37, 1021–1028.
  113. Hawkins, J.; Baker, C.; Cherry, L.; Dunne, E. Black elderberry (Sambucus nigra) supplementation effectively treats upper respiratory symptoms: A meta-analysis of randomized, controlled clinical trials. Complement. Ther. Med. 2019, 42, 361–365.
  114. Akram, M.; Tahir, I.M.; Shah, S.M.A.; Mahmood, Z.; Altaf, A.; Ahmad, K.; Munir, N.; Daniyal, M.; Nasir, S.; Mehboob, H. Antiviral potential of medicinal plants against HIV, HSV, influenza, hepatitis, and coxsackievirus: A systematic review. Phytother. Res. 2018, 32, 811–822.
  115. Shahsavandi, S.; Ebrahimi, M.M.; Farahani, A.H. Interfering with lipid raft association: A mechanism to control influenza virus infection by Sambucus nigra. Iran. J. Pharm. Res. 2017, 16, 1147–1154.
  116. Chen, C.; Zuckerman, D.M.; Brantley, S.E.; Sharpe, M.; Childress, K.O.; Hoiczyk, E.; Pendleton, A.R. Sambucus nigra extracts inhibit infectious bronchitis virus at an early point during replication. BMC Vet. Res. 2014, 10, 24.
  117. Młynarczyk, K.; Walkowiak-Tomczak, D.; Łysiak, G. Bioactive properties of Sambucus nigra L. as a functional ingredient for food and pharmaceutical industry. J. Funct. Foods 2018, 40, 377–390.
  118. Porter, R.S.; Bode, R.F. A review of the antiviral properties of black elder (Sambucus nigra L.) products. Phytother. Res. 2017, 31, 533–554.
  119. Ulbricht, C.; Basch, E.; Cheung, L.; Goldberg, H.; Hammerness, P.; Isaac, R.; Khalsa, K.P.S.; Romm, A.; Rychlik, I.; Varghese, M.; et al. An evidence-based systematic review of elderberry and elderflower (Sambucus nigra) by the natural standard research collaboration. J. Diet. Suppl. 2014, 11, 80–120.
  120. Zakay-Rones, Z.; Varsano, N.; Zlotnik, M.; Manor, O.; Regev, L.; Schlesinger, M.; Mumcuoglu, M. Inhibition of several strains of influenza virus in vitro and reduction of symptoms by an elderberry extract (Sambucus nigra L.) during an outbreak of influenza B panama. J. Altern. Complement. Med. 1995, 1, 361–369.
  121. Zhang, S.; Guo, S.-L.; Wang, Q.-B.; Liu, Y.; Shen, H.-W.; Wang, Z.-Y. Effects of fungi fraction on growth and anti-oxidative activity of Eleutherococcus senticosus. China J. Chin. Mater. Med. 2019, 44, 1517–1523.
  122. Della, V.A.; Ricci, G.; Ralli, M.; Gambacorta, V.; De Lucia, A.; Minni, A.; Pirozzi, C.; Paccone, M.; Pastore, V.; Di Stadio, A. The effects of oral supplements with Sambucus nigra, Zinc, Tyndallized Lactobacillus acidophilus (HA122), Arabinogalactans, vitamin D, vitamin E and vitamin C in otitis media with effusion in children: A randomized controlled trial. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 6360–6370.
  123. Li, T.; Ferns, K.; Yan, Z.-Q.; Yin, S.-Y.; Kou, J.-J.; Li, D.; Zeng, Z.; Yin, L.; Wang, X.; Bao, H.-X.; et al. Acanthopanax senticosus: Photochemistry and anticancer potential. Am. J. Chin. Med. 2016, 44, 1543–1558.
  124. Yamauchi, Y.; Ge, Y.-W.; Yoshimatsu, K.; Komatsu, K.; Kuboyama, T.; Yang, X.; Tohda, C.; Komastu, K. Memory enhancement by oral administration of extract of Eleutherococcus senticosus leaves and active compounds transferred in the brain. Nutrients 2019, 11, 1142.
  125. Jin, L.; Schmiech, M.; El Gaafary, M.; Zhang, X.; Syrovets, T.; Simmet, T. A comparative study on root and bark extracts of Eleutherococcus senticosus and their effects on human macrophages. Phytomedicine 2020, 68, 153181.
  126. Zhou, H.; Xing, J.; Liu, S.; Song, F.; Caib, Z.; Pi, Z.; Liu, Z.; Liu, S. Screening and determination for potential α-glucosidase inhibitors from leaves of acanthopanax senticosus harms by using UF-LC/MS and ESI-MSn. Phytochem. Anal. 2011, 23, 315–323.
  127. Lee, S.; Shin, N.-S.; Oh, K.-B.; Shin, K.H. Antibacterial compounds from the leaves of Acanthopanax senticosus. Arch. Pharm. Res. 2003, 26, 40–42.
  128. Wang, Z.; Jiang, H.; Xia, Y.-G.; Yang, B.-Y.; Kuang, H.-X. α-glucosidase inhibitory constituents from acanthopanax senticosus harm leaves. Molecules 2012, 17, 6269–6276.
  129. Wang, L.; Zhang, R.-M.; Liu, G.-Y.; Wei, B.-L.; Wang, Y.; Cai, H.-Y.; Li, F.-S.; Xu, Y.-L.; Zheng, S.-P.; Wang, G. Chinese herbs in treatment of influenza: A randomized, double-blind, placebo-controlled trial. Respir. Med. 2010, 104, 1362–1369.
  130. Jia, Q.; Zhu, R.; Tian, Y.; Chen, B.; Li, R.; Li, L.; Wang, L.; Che, Y.; Zhao, D.; Mo, F.; et al. Salvia miltiorrhiza in diabetes: A review of its pharmacology, phytochemistry, and safety. Phytomedicine 2019, 58, 152871.
  131. Wang, L.; Ma, R.; Liu, C.; Liu, H.; Zhu, R.; Guo, S.; Tang, M.; Li, Y.; Niu, J.; Fu, M.; et al. Salvia miltiorrhiza: A potential red light to the development of cardiovascular diseases. Curr. Pharm. Des. 2017, 23, 1077–1097.
  132. Yin, Z.-K.; Feng, Z.-M.; Jiang, J.-S.; Zhang, X.; Zhang, P.-C.; Yang, Y.-N. Two new tanshinone derivatives from the rhizomes of Salvia miltiorrhiza and their antiviral activities. J. Asian Nat. Prod. Res. 2019, 22, 24–29.
  133. Rahuman, A.A.; Bagavan, A.; Kamaraj, C.; Vadivelu, M.; Zahir, A.A.; Elango, G.; Pandiyan, G.; Kamaraj, C. Evaluation of indigenous plant extracts against larvae of culex quinquefasciatus say (diptera: Culicidae). Parasitol. Res. 2008, 104, 637–643.
  134. Shao, F.; Lu, S. Identification, molecular cloning and expression analysis of five RNA-dependent RNA polymerase genes in salvia miltiorrhiza. PLoS ONE 2014, 9, e95117.
  135. Wu, B.-W.; Pan, T.-L.; Leu, Y.; Chang, Y.-K.; Tai, P.-J.; Lin, K.-H.; Horng, J.-T. Antiviral effects of Salvia miltiorrhiza (danshen) against enterovirus. Am. J. Chin. Med. 2007, 35, 153–168.
  136. Zhang, D.; Guo, J.; Zhang, M.; Liu, X.; Ba, M.; Tao, X.; Yu, L.; Guo, Y.; Dai, J.-G. Oxazole-containing diterpenoids from cell cultures of Salvia miltiorrhiza and their anti-HIV-1 activities. J. Nat. Prod. 2017, 80, 3241–3246.
  137. Ghoke, S.S.; Sood, R.; Kumar, N.; Pateriya, A.K.; Bhatia, S.; Mishra, A.; Dixit, R.; Singh, V.K.; Desai, D.; Kulkarni, D.D.; et al. Evaluation of antiviral activity of Ocimum sanctum and Acacia arabica leaves extracts against H9N2 virus using embryonated chicken egg model. BMC Complement. Altern. Med. 2018, 18, 174.
  138. Hegazy, G.A.; Alnoury, A.M.; Gad, H.G. The role of Acacia Arabica extract as an antidiabetic, antihyperlipidemic, and antioxidant in streptozotocin-induced diabetic rats. Saudi Med. J. 2013, 34, 727–733.
  139. Nutan, N.; Modi, M.; Dezzutti, C.S.; Kulshreshtha, S.; Rawat, A.K.S.; Srivastava, S.K.; Malhotra, S.; Verma, A.; Ranga, U.; Gupta, S.K. Extracts from Acacia catechu suppress HIV-1 replication by inhibiting the activities of the viral protease and Tat. Virol. J. 2013, 10, 309.
  140. El Gendy, A.E.-N.G.; Al-Mahdy, D.A.M.; El Dine, R.S.; Fahmy, S.; Yassin, A.; Porzel, A.; Brandt, W. Structure activity relationships of antimicrobial gallic acid derivatives from pomegranate and acacia fruit extracts against potato bacterial wilt pathogen. Chem. Biodivers. 2015, 12, 955–962.
  141. Vlachojannis, J.; Erne, P.; Zimmermann, B.; Chrubasik-Hausmann, S. The impact of cocoa flavanols on cardiovascular health. Phytother. Res. 2016, 30, 1641–1657.
  142. Cohen, M.; Cohen, M. Tulsi—Ocimum sanctum: A herb for all reasons. J. Ayurveda Integr. Med. 2014, 5, 251–259.
  143. Prakash, K.; Goyal, M.; Soni, A.; Siddiqui, A.J.; Bhardwaj, J.; Puri, S.K. Molecular cloning and biochemical characterization of iron superoxide dismutase from the rodent malaria parasite Plasmodium vinckei. Parasitol. Int. 2014, 63, 817–825.
  144. Prakash, P.; Gupta, N. Therapeutic uses of Ocimum sanctum Linn (Tulsi) with a note on eugenol and its pharmacological actions: A short review. Indian J. Physiol. Pharmacol. 2005, 49, 125–131.
  145. Baliga, M.S.; Jimmy, R.; Thilakchand, K.R.; Sunitha, V.; Bhat, N.R.; Saldanha, E.; Rao, S.; Rao, P.; Arora, R.B.; Palatty, P.L. Ocimum sanctum L (holy basil or tulsi) and its phytochemicals in the prevention and treatment of cancer. Nutr. Cancer 2013, 65, 26–35.
  146. Ahirwar, P.; Shashikiran, N.D.; Sundarraj, R.K.; Singhla, S.; Thakur, R.A.; Maran, S. A clinical trial comparing antimicrobial efficacy of “essential oil of Ocimum sanctum” with triple antibiotic paste as an intracanal medicament in primary molars. J. Indian Soc. Pedod. Prev. Dent. 2018, 36, 191–197.
  147. Kamyab, A.A.; Eshraghian, A. Anti-inflammatory, gastrointestinal and hepatoprotective effects of Ocimum sanctum Linn: An ancient remedy with new application. Inflamm. Allergy Drug Targets 2013, 12, 378–384.
  148. Mondal, S.; Mirdha, B.R.; Mahapatra, S.C. The science behind sacredness of Tulsi (Ocimum sanctum Linn.). Indian J. Physiol. Pharmacol. 2010, 53, 291–306.
  149. Pattanayak, P.; Behera, P.; Das, D.; Panda, S.K. Ocimum sanctum Linn. A reservoir plant for therapeutic applications: An overview. Pharmacogn. Rev. 2010, 4, 95–105.
  150. Penmetsa, G.S.; Pitta, S.R. Efficacy of Ocimum sanctum, Aloe vera and chlorhexidine mouthwash on gingivitis: A randomized controlled comparative clinical study. An. Int. Q. J. Res. Ayurveda 2019, 40, 23–26.
  151. Ayuob, N.; El Wahab, M.G.A.; Ali, S.S.; Abdel-Tawab, H.S. Ocimum basilicum improve chronic stress-induced neurodegenerative changes in mice hippocampus. Metab. Brain Dis. 2018, 33, 795–804.
  152. Alegría-Herrera, E.; Herrera-Ruiz, M.; Román-Ramos, R.; Zamilpa, A.; Santillán-Urquiza, M.A.; Aguilar, M.I.; Avilés-Flores, M.; Fuentes-Mata, M.; Jiménez-Ferrer, E. Effect of Ocimum basilicum, Ocimum selloi, and rosmarinic acid on cerebral vascular damage in a chronic hypertension model. Biol. Pharm. Bull. 2019, 42, 201–211.
  153. Sestili, P.; Ismail, T.; Calcabrini, C.; Guescini, M.; Catanzaro, E.; Turrini, E.; Layla, A.; Akhtar, S.; Fimognari, C. The potential effects of Ocimum basilicum on health: A review of pharmacological and toxicological studies. Expert Opin. Drug Metab. Toxicol. 2018, 14, 679–692.
  154. Singh, P.; Chakraborty, P.; He, D.-H.; Mergia, A. Extract prepared from the leaves of Ocimum basilicum inhibits the entry of Zika virus. Acta Virol. 2019, 63, 316–321.
  155. Rashidian, A.; Roohi, P.; Mehrzadi, S.; Ghannadi, A.R.; Minaiyan, M. Protective effect of Ocimum basilicum essential oil against acetic acid–induced colitis in rats. J. Evid. Based Integr. Med. 2016, 21, NP36–NP42.
  156. Wickramasuriya, A.M.; Dunwell, J.M. Cacao biotechnology: Current status and future prospects. Plant Biotechnol. J. 2017, 16, 4–17.
  157. Wirngo, F.E.; Lambert, M.N.; Jeppesen, P.B. The physiological effects of dandelion (Taraxacum officinale) in type 2 diabetes. Rev. Diabet. Stud. 2016, 13, 113–131.
  158. Goya, L.; Martin, A.E.; Sarriá, B.; Ramos, S.; Mateos, R.; Bravo-Clemente, L. Effect of cocoa and its flavonoids on biomarkers of inflammation: Studies of cell culture, animals and humans. Nutrients 2016, 8, 212.
  159. Latif, R. Chocolate/cocoa and human health: A review. Neth. J. Med. 2013, 71, 63–68.
  160. Oyeleke, S.A.; Ajayi, A.M.; Umukoro, S.; Aderibigbe, A.; Ademowo, O.G. Anti-inflammatory activity of Theobroma cacao L. stem bark ethanol extract and its fractions in experimental models. J. Ethnopharmacol. 2018, 222, 239–248.
  161. Kamei, M.; Nishimura, H.; Takahashi, T.; Takahashi, N.; Inokuchi, K.; Mato, T.; Takahashi, K. Anti-influenza virus effects of cocoa. J. Sci. Food Agric. 2015, 96, 1150–1158.
  162. Careddu, D.; Pettenazzo, A. Pelargonium sidoides extract EPs 7630: A review of its clinical efficacy and safety for treating acute respiratory tract infections in children. Int. J. Gen. Med. 2018, 11, 91–98.
  163. Moyo, M.; Van Staden, J. Medicinal properties and conservation of Pelargonium sidoides DC. J. Ethnopharmacol. 2014, 152, 243–255.
  164. Moyo, M.; Aremu, A.O.; Gruz, J.; Šubrtová, M.; Szüčová, L.; Doležal, K.; Van Staden, J. Conservation strategy for Pelargonium sidoides DC: Phenolic profile and pharmacological activity of acclimatized plants derived from tissue culture. J. Ethnopharmacol. 2013, 149, 557–561.
  165. Theisen, L.L.; Muller, C.P. EPs® 7630 (Umckaloabo®), an extract from Pelargonium sidoides roots, exerts anti-influenza virus activity in vitro and in vivo. Antivir. Res. 2012, 94, 147–156.
  166. Flores-Ocelotl, M.R.; Rosas-Murrieta, N.H.; Moreno, D.A.; Vallejo-Ruiz, V.; Reyes-Leyva, J.; Domínguez, F.; Santos-López, G. Taraxacum officinale and Urtica dioica extracts inhibit dengue virus serotype 2 replication in vitro. BMC Complement. Altern. Med. 2018, 18, 95.
  167. Schütz, K.; Carle, R.; Schieber, A. Taraxacum—A review on its phytochemical and pharmacological profile. J. Ethnopharmacol. 2006, 107, 313–323.
  168. Abdel-Magied, N.; Fattah, S.M.A.; Elkady, A.A. Differential effect of Taraxacum officinale L. (dandelion) root extract on hepatic and testicular tissues of rats exposed to ionizing radiation. Mol. Biol. Rep. 2019, 46, 4893–4907.
  169. Choi, J.; Yoon, K.D.; Kim, J. Chemical constituents from Taraxacum officinale and their α-glucosidase inhibitory activities. Bioorg. Med. Chem. Lett. 2018, 28, 476–481.
  170. Wang, K.C.; Chang, J.S.; Chiang, L.C.; Lin, C.C. Cimicifuga foetida L. inhibited human respiratory syncytial virus in HEp-2 and A549 cell lines. Am. J. Chin. Med. 2012, 40, 151–162.
  171. Han, H.; He, W.; Wang, W.; Gao, B. Inhibitory effect of aqueous dandelion extract on HIV-1 replication and reverse transcriptase activity. BMC Complement. Altern. Med. 2011, 11, 112.
  172. Lee, B.-R.; Lee, J.-H.; An, H.-J. Effects of Taraxacum officinale on fatigue and immunological parameters in mice. Molecules 2012, 17, 13253–13265.
  173. He, W.; Han, H.; Wang, W.; Gao, B. Anti-influenza virus effect of aqueous extracts from dandelion. Virol. J. 2011, 8, 538.
  174. Lü, H.-N.; Ma, S.-G.; Liu, Y.-B.; Qu, J.; Li, Y.; Xu, S.; Zhu, H.; Yu, S.-S. Sesquiterpenes from the roots of Illicium oligandrum. J. Asian Nat. Prod. Res. 2015, 17, 430–438.
  175. Ma, S.-G.; Gao, R.-M.; Li, Y.-H.; Jiang, J.-D.; Gong, N.-B.; Li, L.; Lu, Y.; Tang, W.-Z.; Liu, Y.-B.; Qu, J.; et al. Antiviral spirooliganones A and B with unprecedented skeletons from the roots of Illicium oligandrum. Org. Lett. 2013, 15, 4450–4453.
  176. Zhu, Q.; Tang, C.-P.; Ke, C.-Q.; Wang, W.; Zhang, H.-Y.; Ye, Y. Sesquiterpenoids and phenylpropanoids from pericarps of Illicium oligandrum. J. Nat. Prod. 2009, 72, 238–242.
  177. Tang, W.-Z.; Liu, Y.; Yu, S.-S.; Qu, J.; Su, D.-M. New sesquiterpene lactone and neolignan glycosides with antioxidant and anti-inflammatory activities from the fruits of Illicium oligandrum. Planta Med. 2007, 73, 484–490.
  178. Kew Science. Glycyrrhiza glabra L. Available online: http://www.plantsoftheworldonline.org/taxon/urn:lsid:ipni.org:names:496941-1 (accessed on 23 August 2020).
  179. Dastagir, G.; Rizvi, M.A. Review—Glycyrrhiza glabra L. (Liquorice). Pak. J. Pharm. Sci. 2016, 29, 1727–1733.
  180. Harding, V.; Stebbing, J. Liquorice: A treatment for all sorts? Lancet Oncol. 2017, 18, 1155.
  181. Dao, T.T.; Nguyen, P.H.; Lee, H.S.; Kim, E.; Park, J.; Lim, S.I.; Oh, W. Chalcones as novel influenza A (H1N1) neuraminidase inhibitors from Glycyrrhiza inflata. Bioorg. Med. Chem. Lett. 2011, 21, 294–298.
  182. Zhang, T.; Wang, Q.; Fredimoses, M.; Gao, G.; Wang, K.; Chen, H.; Wang, T.; Oi, N.; Zykova, T.A.; Reddy, K.; et al. The Ashitaba (Angelica keiskei) chalcones 4-hydroxyderricin and xanthoangelol suppress melanomagenesis by targeting BRAF and PI3-K. Cancer Prev. Res. 2018, 11, 607–620.
  183. Kil, Y.-S.; Pham, S.T.; Seo, E.K.; Jafari, M. Angelica keiskei, an emerging medicinal herb with various bioactive constituents and biological activities. Arch. Pharm. Res. 2017, 40, 655–675.
  184. Park, J.-Y.; Ko, J.-A.; Kim, D.W.; Kim, Y.M.; Kwon, H.-J.; Jeong, H.J.; Kim, C.Y.; Park, K.H.; Lee, W.-S.; Ryu, Y.B. Chalcones isolated from Angelica keiskeiinhibit cysteine proteases of SARS-CoV. J. Enzym. Inhib. Med. Chem. 2015, 31, 23–30.
  185. Kweon, M.; Lee, H.; Park, C.; Choi, Y.H.; Ryu, J.-H. A chalcone from Ashitaba (Angelica keiskei) stimulates myoblast differentiation and inhibits dexamethasone-induced muscle atrophy. Nutrients 2019, 11, 2419.
  186. Panda, S.; Chand, M.; Sakhuja, R.; Jain, S. Xanthones as potential antioxidants. Curr. Med. Chem. 2013, 20, 4481–4507.
  187. Le Pogam, P.; Boustie, J. Xanthones of lichen source: A 2016 update. Molecules 2016, 21, 294.
  188. EOL. Polygala Karensium Kurz. Available online: https://eol.org/pages/2886028 (accessed on 23 August 2020).
  189. Jeong, H.J.; Kim, Y.M.; Kim, J.H.; Kim, J.Y.; Park, J.-Y.; Park, S.-J.; Ryu, Y.B.; Lee, W.S. Homoisoflavonoids from Caesalpinia sappan displaying viral neuraminidases inhibition. Biol. Pharm. Bull. 2012, 35, 786–790.
  190. Wikipedia. Calophyllum Brasiliense. Available online: https://en.wikipedia.org/wiki/Calophyllum_brasiliense (accessed on 24 August 2020).
  191. Kudo, E.; Taura, M.; Matsuda, K.; Shimamoto, M.; Kariya, R.; Goto, H.; Hattori, S.; Kimura, S.; Okada, S. Inhibition of HIV-1 replication by a tricyclic coumarin GUT-70 in acutely and chronically infected cells. Bioorg. Med. Chem. Lett. 2013, 23, 606–609.
  192. Efloras. Cimicifuga Foetida. Available online: http://www.efloras.org/object_page.aspx?object_id=108347&flora_id=800 (accessed on 24 August 2020).
  193. Zhu, G.-L.; Zhu, D.-F.; Wan, L.-S.; Peng, X.-R.; Bao, N.-M.; Zhang, Z.-R.; Zhou, L.; Qiu, M.-H. Six new 9,19-cycloartane triterpenoids from Cimicifuga foetida L. Nat. Prod. Bioprospect. 2016, 6, 187–193.
  194. Wang, K.-C.; Chang, J.-S.; Lin, L.-T.; Chiang, L.-C.; Lin, C.-C. Antiviral effect of cimicifugin from cimicifuga foetida against human respiratory syncytial virus. Am. J. Chin. Med. 2012, 40, 1033–1045.
  195. Thomford, N.E.; Awortwe, C.; Dzobo, K.; Adu, F.; Chopera, D.; Wonkam, A.; Skelton, M.; Blackhurst, D.; Dandara, C. Inhibition of CYP2B6 by medicinal plant extracts: Implication for use of efavirenz and nevirapine-based Highly Active Anti-Retroviral Therapy (HAART) in resource-limited settings. Molecules 2016, 21, 211.
  196. Gai, Y.-Y.; Liu, W.; Sha, C.-J.; Wang, Y.-L.; Sun, Y.-T.; Li, X.-J.; Fawcett, J.P.; Gu, J. Pharmacokinetics and bioavailability of cimicifugosides after oral administration of Cimicifuga foetida L. extract to rats. J. Ethnopharmacol. 2012, 143, 249–255.
  197. Mandeville, A.; Cock, I.E. Terminalia chebula Retz. fruit extracts inhibit bacterial triggers of some autoimmune diseases and potentiate the activity of tetracycline. Indian J. Microbiol. 2018, 58, 496–506.
  198. Manu, K.; Kuttan, G. Effect of punarnavine, an alkaloid from boerhaavia diffusa, on cell-mediated immune responses and TIMP-1 in B16F-10 metastatic melanoma-bearing mice. Immunopharmacol. Immunotoxicol. 2007, 29, 569–586.
  199. Bose, M.; Kamra, M.; Mullick, R.; Bhattacharya, S.; Das, S.; Karande, A.A. A plant-derived dehydrorotenoid: A new inhibitor of hepatitis C virus entry. FEBS Lett. 2017, 591, 1305–1317.
  200. Wikipedia. Boerhavia Diffusa. Available online: https://en.wikipedia.org/wiki/Boerhavia_diffusa (accessed on 24 August 2020).
  201. Mishra, S.; Aeri, V.; Gaur, P.K.; Jachak, S.M. Phytochemical, therapeutic, and ethnopharmacological overview for a traditionally important herb: Boerhavia diffusaLinn. BioMed Res. Int. 2014, 2014, 1–19.
  202. Li, K.; Han, X.; Li, R.; Xu, Z.; Pan, T.; Liu, J.; Li, B.; Wang, S.; Diao, Y.; Liu, X. Composition, antivirulence activity, and active property distribution of the fruit of terminalia chebula Retz. J. Food Sci. 2019, 84, 1721–1729.
  203. Kesharwani, A.; Polachira, S.K.; Nair, R.; Agarwal, A.; Mishra, N.N.; Gupta, S.K. Anti-HSV-2 activity of Terminalia chebula Retz extract and its constituents, chebulagic and chebulinic acids. BMC Complement. Altern. Med. 2017, 17, 110.
  204. Promila, P.; Madan, V.K. Therapeutic & phytochemical profiling of Terminalia chebula Retz. (harad): A review. J. Med. Plants Stud. 2018, 6, 25–31.
  205. El Sayed, K.A. Natural products as antiviral agents. Stud. Nat. Prod. Chem. 2000, 24, 473–572.
  206. Sheng, Z.; Zhao, J.; Muhammad, I.; Zhang, Y. Optimization of total phenolic content from Terminalia chebula Retz. fruits using response surface methodology and evaluation of their antioxidant activities. PLoS ONE 2018, 13, e0202368.
  207. Lin, L.-T.; Chen, T.-Y.; Lin, S.-C.; Chung, C.-Y.; Lin, T.-C.; Wang, G.-H.; Anderson, R.; Lin, C.-C.; Richardson, C.D. Broad-spectrum antiviral activity of chebulagic acid and punicalagin against viruses that use glycosaminoglycans for entry. BMC Microbiol. 2013, 13, 187.
  208. Zhang, X.; He, L.; Lu, Q.; Li, D. Pharmacological activity of Terminalia chebula. China J. Chin. Mater. Med. 2016, 41, 619–623.
  209. Nigam, M.; Mishra, A.P.; Adhikari-Devkota, A.; Dirar, A.I.; Hassan, M.; Adhikari, A.; Belwal, T.; Devkota, H.P. Fruits of Terminalia chebula Retz.: A review on traditional uses, bioactive chemical constituents and pharmacological activities. Phytother. Res. 2020, 10, 1–9.
  210. Yang, F.; Zhou, W.-L.; Liu, A.; Qin, H.-L.; Lee, S.M.; Wang, Y.-T.; Du, G. The protective effect of 3-deoxysappanchalcone on in vitro influenza virus-induced apoptosis and inflammation. Planta Med. 2012, 78, 968–973.
  211. Natures Beauty Creations. Caesalpinia sappan L. Available online: https://www.asia-medicinalplants.info/caesalpinia-sappan-l/ (accessed on 23 August 2020).
  212. Tewtrakul, S.; Chaniad, P.; Pianwanit, S.; Karalai, C.; Ponglimanont, C.; Yodsaoue, O. Anti-HIV-1 integrase activity and molecular docking study of compounds from Caesalpinia sappan L. Phytother. Res. 2015, 29, 724–729.
More
Information
Subjects: Plant Sciences
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , , , , , , , ,
View Times: 1.2K
Revisions: 6 times (View History)
Update Date: 01 Dec 2020
1000/1000