Next Article in Journal
Biosynthesized ZnO Nanoparticles Using Albizia lebbeck Extract Induced Biochemical and Morphological Alterations in Wistar Rats
Next Article in Special Issue
Pimenta dioica (L.) Merr. Bioactive Constituents Exert Anti-SARS-CoV-2 and Anti-Inflammatory Activities: Molecular Docking and Dynamics, In Vitro, and In Vivo Studies
Previous Article in Journal
Actinidia arguta Leaf as a Donor of Potentially Healthful Bioactive Compounds: Implications of Cultivar, Time of Sampling and Soil N Level
Previous Article in Special Issue
Targeting Multiple Signal Transduction Pathways of SARS-CoV-2: Approaches to COVID-19 Therapeutic Candidates
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Brief Overview of Potential Treatments for Viral Diseases Using Natural Plant Compounds: The Case of SARS-Cov

by
Rambod Abiri
1,
Hazandy Abdul-Hamid
1,2,*,
Oksana Sytar
3,4,
Ramin Abiri
5,6,
Eduardo Bezerra de Almeida, Jr.
7,
Surender K. Sharma
8,
Victor P. Bulgakov
9,*,
Randolph R. J. Arroo
10 and
Sonia Malik
11,12,*
1
Department of Forestry Science and Biodiversity, Faculty of Forestry and Environment, Universiti Putra Malaysia, Serdang 43400, Malaysia
2
Laboratory of Bioresource Management, Institute of Tropical Forestry and Forest Products (INTROP), Universiti Putra Malaysia, Serdang 43400, Malaysia
3
Educational and Scientific Center “Institute of Biology and Medicine”, Department of Plant Biology, Taras Shevchenko National University of Kyiv, Volodymyrska 60, 01033 Kyiv, Ukraine
4
Department of Plant Physiology, Slovak University of Agriculture Nitra, A. Hlinku 2, 94976 Nitra, Slovakia
5
Department of Microbiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah 6718773654, Iran
6
Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6718773654, Iran
7
Biological and Health Sciences Centre, Laboratory of Botanical Studies, Department of Biology, Federal University of Maranhão, São Luís 65080-805, MA, Brazil
8
Department of Physics, Central University of Punjab, Bathinda 151401, India
9
Department of Biotechnology, Federal Scientific Center of the East Asia Terrestrial Biodiversity (Institute of Biology and Soil Science), Far Eastern Branch of the Russian Academy of Sciences, 159 Stoletija Str., 690022 Vladivostok, Russia
10
Leicester School of Pharmacy, De Montfort University, The Gateway, Leicester LE1 9BH, UK
11
Health Sciences Graduate Program, Biological & Health Sciences Centre, Federal University of Maranhão, São Luís 65080-805, MA, Brazil
12
Laboratoire de Biologie des Ligneux et des Grandes Cultures (LBLGC), University of Orléans, 1 Rue de Chartres-BP 6759, 45067 Orleans, France
*
Authors to whom correspondence should be addressed.
Molecules 2021, 26(13), 3868; https://doi.org/10.3390/molecules26133868
Submission received: 5 March 2021 / Revised: 2 June 2021 / Accepted: 3 June 2021 / Published: 24 June 2021
(This article belongs to the Special Issue Natural Products for COVID-19: Prevention and Treatment)

Abstract

:
The COVID-19 pandemic, as well as the more general global increase in viral diseases, has led researchers to look to the plant kingdom as a potential source for antiviral compounds. Since ancient times, herbal medicines have been extensively applied in the treatment and prevention of various infectious diseases in different traditional systems. The purpose of this review is to highlight the potential antiviral activity of plant compounds as effective and reliable agents against viral infections, especially by viruses from the coronavirus group. Various antiviral mechanisms shown by crude plant extracts and plant-derived bioactive compounds are discussed. The understanding of the action mechanisms of complex plant extract and isolated plant-derived compounds will help pave the way towards the combat of this life-threatening disease. Further, molecular docking studies, in silico analyses of extracted compounds, and future prospects are included. The in vitro production of antiviral chemical compounds from plants using molecular pharming is also considered. Notably, hairy root cultures represent a promising and sustainable way to obtain a range of biologically active compounds that may be applied in the development of novel antiviral agents.

1. Introduction

Bronchitis is a respiratory disease caused by bacterial infections, viral infections, or irritant particles [1]. In response to infection, the bronchial tubes become inflamed and swollen, which may eventually result in acute respiratory arrest. Coronavirus disease-19 (Covid-19), which recently emerged as a pandemic, is an infectious respiratory disease caused by a newly discovered coronavirus. The first report of this novel coronavirus was traced back to the Huanan wholesale seafood market in Wuhan city, China, in December 2019, where a group of patients exhibited a mysterious kind of viral pneumonia [2]. Nowadays, viral pneumonia is diagnosed through analyzing a sample of bronchoalveolar lavage fluid using PCR, cell cultures, and whole-genome sequencing [3]. The virus was isolated from infected individuals and recognized as genus beta-coronavirus, placing it alongside other viruses causing Severe Acute Respiratory Syndrome (SARS) and Middle East Respiratory Syndrome (MERS) [4]. Previously, SARS had been reported in Southern China during 2002–2003, and its outbreak has been reported in 29 countries, with almost 8000 infected cases and around 700 mortalities (http://www.who.int/csr/sars/en/, the accessed date: 7 February 2021). A decade after the appearance of SARS, MERS was reported during 2012–2014 as a second coronavirus generation that caused a global pandemic. MERS affected people in more than 27 countries with over 2000 (32.97%) case-fatality. Coronavirus is an enveloped, positive-sense, and single-stranded RNA or (+) ssRNA virus with a large genome of approximately 30 kb.
Covid-19 is a life-threatening illness with a tremendous rate of spreading in humans due to its high level of infectiousness [5]. The treatment of this disease is a great challenge due to several reasons, including the rapid emergence of mutant strains, the consequent high rate of virus adaptation, and the development of resistance to antiviral medicines. Another factor is that of unwanted side effects and the high cost of synthetic antiviral drugs. There has been a significant global interest in developing safe and effective Covid-19 vaccines since 2020. Forman and his colleagues provided an overview of current phase II/III, III, and IV COVID-19 vaccine candidates (20 different vaccines) [6]. The standard approach for viral infections comprises antiviral medicines that do not cause damage to the human host but can help shorten viral infection, inhibit virus expansion, and help in reducing/blocking complications [1]. In parallel with the generation of vaccines in 2020, studies on the antiviral properties of natural compounds have also been performed via the molecular docking methodology [7,8,9,10]. The potency of marine natural products has been confirmed to target SARS-CoV-2 main protease (Mpro) [11].
Medicinal plants have been identified as reliable resource against several diseases for millennia. More than 70% of the global population still depends on herbal medicines due to their relatively low cost and better compatibility with the human body compared to synthetic drugs [12]. During the pandemic period, studies were performed using databases of scientific literature to screen and identify the potential of herbal plants to act as anti-coronavirus medication [13]. It has been reported that water and ethanol plant extracts contain biologically active substances with antiviral activity [14].
A wide range of compounds identified in several plant species have demonstrated antiviral activities, including alkaloids, flavonoids, triterpenes, anthraquinones, and lignans. Interestingly, plant selection based on ethnomedical concerns provides a higher hit rate than screening plants or general synthetic products [15]. For instance, lycorine extracted from Lycoris radiata L. and glycyrrhizin from Glycyrrhiza uralensis Fisch showed anti-SARS-CoV activities [16]. Some known pharmacophore structures of bioactive substances may be useful in the creation of new anti-Covid-19 drugs. Natural compounds such as betulinic acid, indigo, aloe emodin, luteolin, quinomethyl triterpenoids, quercetin, and gallates have been reported to inhibit viral proteases with the potential to develop antiviral drugs [14]. In addition, plants have also been introduced as a safe and reliable bioreactor for the production of recombinant virus proteins that can be used in vaccine development [17], e.g., nuclear transformed tomatoes and tobacco-expressing antigens have been reported to induce immunogenic responses against SARS-CoV [18].
The main objective of the current review is to provide the complete overview of the ethnomedicinal uses of herbs employed to treat respiratory diseases. We address questions regarding the potential of plant-derived compounds in inhibiting virus propagation, thus providing relief for viral-induced pathogenesis. We also discuss how biotechnology may help solve the challenge of rapidly obtaining pure antiviral compounds. Furthermore, this review discusses the current state of the art regarding the possible antiviral activities of herbal medicine and makes an effort to tackle the gaps in scientific knowledge that may lead to the advancement of innovative treatments for the welfare of people and against the spread of viral diseases, especially SAR-CoV.

2. A Brief Model of Viral Entering/Replication in Host Cells

The life and replication cycle of a virus is dependent on the cell processes of its host (Figure 1). The reproduction cycle of viruses causes significant structural (cytopathic) and biochemical damage that, in severe conditions, may ultimately result in the death of the host organism [19]. Viruses can enter host cells via different biological mechanisms including phagocytosis, pinocytosis, and endocytosis. To enter and spread to the cells of a living organism, the virus fuses at the plasma membrane in the first phase, and then it infects the other cells of the host organism via cell-to-cell syncytia or fusion in the second phase. The respiratory system is the main site of entrance for viruses into host organisms, and severe infection of the respiratory tract may cause life-threatening damage to the lungs [20]. Generally, viral replication is completed within several hours [21] and involves various phases including attachment, penetration, un-coating, replication, assembly, and release [22].

3. General Model of SARS-CoV Pathogenicity

Coronaviruses (CoVs) are considered as a subfamily of Orthocoronavirinae and belongs to a cluster of vertebrate animal and human viruses that affect the respiratory tract, liver, central nervous systems, and digestive systems [24]. Currently, no specific anti-viral treatment has been recommended for COVID-19, and investigations have focused on vaccine development [25]. This human pathogen is a positive-strand RNA virus (positive-sense (5′-to-3′) viral RNA) enveloped by spike glycoproteins (S). These proteins bind to their receptor on the surface of the host cells. The spike proteins possess two compartments, namely envelop proteins (E) and membrane proteins (M), which play important roles in pathogenesis. Some spike glycoproteins have envelope-associated hemagglutinin-esterases (HEs) [26]. CoVs can bind to angiotensin-converting enzyme (ACE2) receptors through the receptor-binding domain (RBD) and then enter cells. The virus has two different shapes, L and S. The “L” from is assumed to be more virulent and adaptable than the “S” form to interact with the [27]. Specifically, the S1 domain of Covid-19’s spike glycoprotein interacts with an immunoregulatory factor for virulence and hijacking, the human CD26 [28]. CD26 is linked to inflammation; it plays a vital function in T-cell activation and is expressed in plasma and on the cell surface of several non-immune and immune cell types. It has been reported that high-fiber diets and citrus flavonoids that ameliorate the effects of type 2 diabetes mellitus (T2DM) are also inhibitors of CD26 [20].
Furthermore, the epigenetic dysregulation of ACE2 and interferon-regulated genes may suggest increased Covid-19 susceptibility and severity in lupus patients [29]. The S protein has two functional domains: (a) the receptor-binding domain and (b) sequences that mediate the fusion of viral and cell membranes. S-glycoprotein must be cleaved by cellular proteases to provide access to fusion sequences and is required for cell entry. The nature of the cell protease that cleaves the S glycoprotein varies according to the coronavirus type. For instance, the MERS-CoV S glycoprotein contains a furin protease cleavage site and is processed by these intracellular proteases during exit from the cell. This protein processing by the host’s furin protease is necessary for the development of the virus and allows it to enter the next cell. In contrast, SARS-CoV S glycoprotein is uncleaved upon release of virus from cells. It is probably cleaved during virus entry into a cell. Furin proteases may therefore be targeted for therapeutic uses [20] (Figure 2).

4. Replication Inhibitors of SARS-CoV

Previous investigations have demonstrated that the development of proteases is an ideal goal to be tackled for the inhibition of CoV replication. In silico analysis demonstrated a 96% similarity between 2019-CoV main protease and the SARS-CoV Mpro, and no mutation was reported in the active sites in both proteins [31]. Though the protease activity disruption causes various diseases, host proteases are considered reliable therapeutic targets. For several different viruses, protease activity represents a vital factor in replication; thus, proteases are frequently targeted as protein candidates during antiviral therapeutics studies [32]. Lopinavir and nelfinavir are in the category of medications named protease inhibitors with a high level of cytotoxicity recommended for the treatment of cells infected with MERS, SARS, and HIV [33]. Some preliminary investigations have examined the potential of protease inhibitor ritonavir/lopinavir, which is regularly implemented to cure human immunodeficiency virus (HIV)/acquired immunodeficiency syndrome cases for the therapy of Covid-19-infected patients. In addition, other studies on the antiviral treatment of pathogenic forms of human CoV have reported on lamivudine (3TC), tenofovir disoproxil (TDF), umifenovir (arbidol), remdesivir, neuraminidase inhibitors, and nucleoside analogues, among others [34]. Moreover, another effort demonstrated that nelfinavir was the best potential inhibitor candidate over praziquantel, perampanel, and pitavastatin against COVID-19 Mpro. The efficiency of nelfinavir has been reported due to the binding free energy calculations using the molecular mechanics with generalized Born and surface area solvation (MM/GBSA) model and solvated interaction energy (SIE) methods. In this regard, the crystal structures of protease/chymotrypsin-like protease (3CLpro) [27] from Covid-19 patients have shown that this protease could have the capability to inhibit CoV replication [35]. On the other hand, molecular docking investigations revealed that epicatechin-gallate, catechin, curcumin, oleuropein, apigenin-7-glucoside, demethoxycurcumin, and luteolin-7-glucoside may have the potential to inhibit Covid-19 Mpro. Additionally, an in silico analysis showed that the mentioned phenolic compounds share a pharmacophore with nelfinavir [36]. Thus far, different flavonoid compounds derived from medicinal plants have shown antiviral bioactivities for inhibiting protease.

5. Inhibitors of Assembly and Packaging of SARS-CoV

During the virus’ life cycle, the assembly and release of infectious particles is the final phase [37]. In this phase, viral structural proteins (often mentioned as pre-structural proteins such as P1 of enterovirus 71) mature until they are assembled into viral capsids. In this phase, the SARS-CoV genomes are actively tightly packaged into pre-formed viral protein capsids for enveloped intracellular cargo transport and then released [38]. Notwithstanding the absolute necessity for virus infection, so far, no antiviral drugs/agents have tackled this phase [39]. However, the results of studies have shown that some medicinal plants can interfere with the viral packaging and assembly mechanisms [23].

6. Evidence Supporting the Antiviral Efficacy of Medicinal Plants

The use of therapeutic plants against viral infection can be traced back to the dawn of civilization; however, BOOTS Pure Drug Co., Ltd., Nottingham (England) made the first systematic effort to screen plants against influenza [40]. Later on, the inhibitory effect of medicinal plants on the replication of viruses was studied on severe acute respiratory syndrome (SARS) virus, emerging viral infections linked with poxvirus, hepatitis B virus (HBV), HIV, and herpes simplex virus type 2 (HSV-2) [41,42,43,44,45,46] (Table 1). Many studies have applied either alcoholic or aqueous extracts of medicinal plants. However, only a few investigations have been conducted to study active natural compounds presenting antiviral effects. It has been demonstrated that molecular mechanisms linked to the antiviral effects of medicinal plant extracts vary among various types of viruses. Nonetheless, medicinal plant extracts potentially improve the inherent antiviral defense mechanisms of the human body, which involve a complicated system and might use several concurrent pathways. Thus far, some investigations have discovered immunostimulatory properties of medicinal plant extracts possessing antiviral activity [47]. Many plants including Vitex negundo, Solanum nigrum, Scharicum guerke, Ocimum kilim, Sambucus ebulus, Ocimum sanctum, Euphorbia granulate, Eugenia jambolana, and Acacia nilotica have been revealed to target reverse transcriptase activity and have shown inhibitory actions towards HIV proteases [48,49,50,51,52,53]. The root extracts of Heracleum maximum Bartr. (Apiaceae) enhanced interleukin 6 (IL-6) in a macrophage activation test, thus proving antiviral effects linked with the immunostimulatory characteristics [47]. Likewise, Plantago asiatica Linn. (Plantaginaceae) and Plantago major Linn. are frequently used as folk medicinal plants in Taiwan for the treatment of different infectious diseases, and both were found to induce the secretion of interferon-gamma (IFN-γ) and lymphocyte proliferation at low concentrations. Both the secretion of interferon-gamma (IFN-γ) and the induction of lymphocyte proliferation activity are reliable indicators of cell-mediated immune response modulation [47,54].

7. Plant-Derived Immunomodulators

The phagocyte–microbe interactions in the immune system comprise a defense reaction that, under more harmful circumstances, may take part in the advancement of various immune and non-immune chronic inflammatory diseases. The immune system of a healthy organism manages the homeostasis of the body. Agents that express a capacity to modulate and normalize pathophysiological processes are named immunomodulators [116]. Most of the well-known immunostimulants and immunosuppressants used in clinical practice are cytotoxic drugs, which can have severe side effects. Therefore, plant-derived compounds and extracts have been studied regarding their immunomodulatory potential in humans due to their lower cytotoxicity and high bioavailability [117,118]. Plant-derived immunomodulators can also be used for a long period [119] (Table 2).
Some plant-derived compounds, e.g., curcumin, genistein, fisetin, quercetin, resveratrol, epigallocatechin-3-gallate, andrographolide, and colchicine, have immunomodulatory effects [120,121,122,123,124,125,126]. These compounds can downregulate the production of proinflammatory cytokines induced by some viroidal agents [122]. Andrographolide and other natural immunomodulators can enhance the activity of cytotoxic T cells, phagocytosis, natural killer (NK) cells, and antibody-dependent cell-mediated cytotoxicity [125]. The use of quercetin in combination with highly active compounds such as psoralen, baccatin III, embelin, and menisdaurin increased its anti-hepatitis B activity up to 10% [127].
At the same time, analyses of other medicinal plant extracts with antiviral properties have shown that Gymnema sylvestre [121], Stephania tetrandra S Moore roots [128,129,130], and Vitex trifolia extracts [107] possess immunomodulating activity. Naser et al. [131] found immunomodulation potential and antiviral activities against acute common cold in leaf extracts of Thuja occidentalis. The anti-SARS and immunomodulatory activity of water extracts of Houttuynia cordata have been reported via the stimulation of lymphocyte proliferation together with enhancing the proportion of CD4+ and CD8+T cells [81] (Table 3).
Table 2. Plant sources of polyphenolic compounds with anti-protease activity.
Table 2. Plant sources of polyphenolic compounds with anti-protease activity.
Plant SpeciesSourcesCompounds Molecular FormulaLipinski’s Rule of FiveReference
PropertiesValue
Spinacia oleracea,
Brassica oleracea, Anethum graveolens, Brassica rapa, Sauropus androgynus
Spinach
Cabbage
Dill
Chinese cabbage Katuk
KaempferolC15H10O6Molecular weight (<500 Da)286.24[132,133]
LogP (<5)1.58
H-bond donor (5)4
H-bond acceptor (<10)6
Violations0
Anethum graveolens, Foeniculum vulgare, Allium cepa,
Oregano vulgare, Capsicum annum
Dill
Fennel leaves
Onion
Oregano
Chili pepper
QuercetinC15H10O7Molecular weight (<500 Da)302.24[132]
LogP (<5)1.23
H-bond donor (5)5
H-bond acceptor (<10)7
Violations0
Olea europaea, Averrhoa belimbi, Capsicum annum, Allium fistulosumOlive
Star fruit
Chili pepper
Welsh onion/Leek
Luteolin-7-glucosideC21H20O11Molecular weight (<500 Da)448.38[134]
LogP (<5)0.16
H-bond donor (5)7
H-bond acceptor (<10)11
Violations2
Curcuma longa,
Curcuma xanthorriza
Turmeric
Curcuma
DemethoxycurcumineC20H18O5Molecular weight (<500 Da)338.35[135,136]
LogP (<5)3
H-bond donor (5)2
H-bond acceptor (<10)5
Violations0
Citrus sinensisCitrus fruitNaringeninC15H12O5Molecular weight (<500 Da)567.78[137]
LogP (<5)4.33
H-bond donor (5)4
H-bond acceptor (<10)5
Violations1
Averrhoa belimbi, Lycium chinese, Apium graveolens, Olea EuropaeaStar fruit
Goji berries
Celery
Olive
Apigenine-7-glucosideC21H20O10Molecular weight (<500 Da)432.34[138,139,140]
LogP (<5)0.55
H-bond donor (5)6
H-bond acceptor (<10)10
Violations1
Olea EuropaeaOliveOleuropeinC19H22O8Molecular weight (<500 Da)378.37[138]
LogP (<5)1.57
H-bond donor (5)3
H-bond acceptor (<10)8
Violations0
Camellia sinesisGreen teaCatechinC15H14O6Molecular weight (<500 Da)290.27[141,142]
LogP (<5)0.85
H-bond donor (5)5
H-bond acceptor (<10)6
Violations0
Curcuma xanthorriza, Curcuma longaTurmeric
Curcuma
CurcuminC21H20O6Molecular weight (<500 Da)368.38[135,136]
LogP (<5)3.03
H-bond donor (5)2
H-bond acceptor (<10)6
Violations0
Zingiber officialeGingerZingerolC11H16O3Molecular weight (<500 Da)196.24[36,143,144]
LogP (<5)1.86
H-bond donor (5)2
H-bond acceptor (<10)3
Violations0
Zingiber officialeGingerGingerolC17H26O4Molecular weight (<500 Da)294.39[36,144,145]
LogP (<5)3.13
H-bond donor (5)2
H-bond acceptor (<10)4
Violations0
Allium sativumGarlicAllicinC6H10OS2Molecular weight (<500 Da)162.27[36]
LogP (<5)1.61
H-bond donor (5)0
H-bond acceptor (<10)1
Violations0
Camellia sinesisGreen teaEpicatechin gallateC22H18O10Molecular weight (<500 Da)442.37[139]
LogP (<5)1.23
H-bond donor (5)7
H-bond acceptor (<10)10
Violations1
Table 3. The mode of action against viruses and methods of active compound extraction from medicinal plants.
Table 3. The mode of action against viruses and methods of active compound extraction from medicinal plants.
Plant Species and
Plant Part
Active CompoundsCoumarinsExtractModel OrganismMode of Action/ActivityRef
Terpenes
Terpenoids
Flavonoids
Flavones
AlkaloidsStilbenes
Méntha piperíta
(whole plant)
Lamiaceae
α-pinene
β-pinene
β-caryophyllene
L-Limonene
Menthol
Eriocitrin
Hesperidin
Kaempferol 7-O-rutinoside
Luteolin and its derivatives
n/aTrans-resveratroln/aEthanolVero cell culturesHigh antiviral activity[145,146,147]
Thymus vulgaris
(whole plant)
Lamiaceae
Thymol
p-cymene
g-erpinene
γ-Terpinene
Linalool
Rutin
Quercetin
n/an/an/aEthanolVero cell culturesHigh antiviral activity and
antioxidant effects
[145,148,149]
Desmodium canadense
(whole plant)
Fabaceae
Sandosaponin B and its derivativesSoyasaponin I Soyasaponin VIHomoorientin
Orientin
2-vicenin
Vitexin
Isovitexin
Rutin
Desmodin
Homoadonivernite
Indole-3-alkylamine phenylethylamine
alkaloids,
pyrrolidine
alkaloids
n/an/aEthanolVero cell culturesHigh antiviral activity[145,150,151,152,153]
Camellia japonica
(whole plant, flowers)
Theaceae
Oleanane triterpenes
3β,18β-dihydroxy-28-norolean-12-en-16-one
18β-hydroxy-28-norolean-12-ene-3,16-dione
Quercetin
Kaempferol
Apigenin
Do not produce purine alkaloidsn/an/aEthanolVero cells (African green monkey kidney cell line; ATCC CCR-81)High antiviral activity on PEDV corona virus
Inhibitory effects on key gene and protein synthesis during PEDV replication
[154,155,156,157,158,159]
Saposhnikovia divaricate
(whole plant)
Apiaceae
n/an/an/an/acis-3′-Isovaleryl4′-acetylkhellactone
Praeruptorin F
Praeruptorin B
(−)-cis-khellactone
EthanolVero cells (African green monkey kidney cell line; ATCC CCR-81)High antiviral activity on PEDV corona virus[155]
Quercus ilex L.
(Leaves)
Fagaceae
kaempferol glycosides (juglanin, kaempferol-3-O-α-L-arabinofuranoside, and afzelin, kaempferol-3-O-α-L-rhamnosiden/an/an/aDMSOXenopus oocytesInhibits 3a channel protein of coronavirus[156,157]
Bupleurum sp.
(whole plant)
Apiaceae
Triterpenoid saponins
Saikosaponins 2″-O-Acetylsaikosaponins
Prosaikogenins
Bupleurosides
Etc.
Quercetin
Isorhamnetin
Narcissin
Rutin
Eugenin
Saikochrome A
n/an/an/aDMSOHuman fetal lung fibroblasts (MRC-5; ATCC CCL-171)Saikosaponins attenuate viral attachment and penetration[160,161]
Houttuynia cordata
(whole plant)
(Saururaceae)
Cycloart-25-ene-3b,24-diolQuercetin 7-rhamnoside
Hyperin
Quercetin
Afzelin
Rutin
Arisolactams
Piperolactam A
Caldensin
n/an/aWaterBALB/c miceDecreases the viral SARS-3CLpro activity
Stops viral t RNA polymerase activity (RdRp)
Increases the secretion of interleukin (IL)-2 and (IL)-10
[88,162]
Isatis tinctoria
(Roots extracts)
Brasicaceae
n/aHesperetin Quercetin
Isoorientin
Isovitexin
Indigo
Indirubin
Indican
Sinigrinn/aWaterVero cellsCleavage of the activity of SARS-3CLpro enzyme decreased[163,164]
Lycoris radiata
(Bulbs)
Amaryllidaceae
β-Myrcene
A-terpineol
Eucalyptol
β-cyclocitral
n/aLycorine
Amaryllidaceae alkaloids
Lycoranines
n/aEthanolVero E6 cellsExhibits anti-SARS-CoV activity[16,165,166,167]
Litchi chinensis
(seeds)
Sapindaceae
3-Oxotrirucalla-7,24-dien-21-oic acidHerbacetin Rhoifolin
Pectolinarin Quercetin Epigallocatechin gallate Gallocatechin gallate
Litchitannins
Kaemferol derivatives
Epicatechin
Cinnamtannin
n/an/an/aWaterOn model with SARS-CoV 3CLproInhibits SARS-3CLpro activity[168,169,170,171]
Stephania tetrandra S Moore
(Roots)
Menispermaceae
n/an/aTetrandrine Fangchinoline, Cepharanthinen/an/aDMSOHuman cell line MRC-5 cellsInhibits the expression of HCoV-OC43 spike and nucleocapsid protein.
Immunomodulation/
[129,172]
Scutellaria baicalensis
(Roots)
Lamiaceae
DodecanedioxinsScutellarein
Baicalin
Wogonin
Wogonoside
n/an/an/aDMSOModel with SARS-CoV helicase, and nsP13Inhibits nsP13 by affecting the ATPase activity[61,173]
Allium sativum
(Bulbs)
Alliaceae
Nerolidol Phytol
Squalene
α-pinene Terpinolene Limonene
1,8-cineole
γ-terpinene
Catechin
Epicatechin
Allicin
Ajoene
Alliin
Diallyl disulfide
Diallyl trisulfide
n/an/aAquaporinChicken embryosInhibitory effects on avian coronavirus[174,175,176,177]
Artemisia sp.
Artemisia absentium
(whole plants)
Asteraceae
Absinthin
Artemisin
Scopoletin
Artamarin
Rutin
Glycosides of quercetin
Artamarin Artamaridin, Artamaridinin, Artamarinin QuebrachitolArtemitinn/an/aWaterDelayed brain tumor cellsReduces coronavirus replication[178,179]
Juniperus communis
(Fruits)
Cupressaceae
Sugiol
α-pinene
β-pinene
Rutin
Scutellarein
Quercetin-3-O-rhamnoside quercitrin
n/an/aUmbelliferonen/aProtein-molecular docking with network pharmacology analysisInhibits the replication, 3CLpro[180,181]
Ecklonia cava
(whole plant)
Lessoniaceae
n/aQuercetinn/an/an/an/aprotein-molecular docking with network pharmacology analysisPLpro and 3CLpro[182]

7.1. Lectins

Lectins are a special type of natural proteins (split into seven different classes of evolutionarily- and structurally-related proteins) found in higher plants that bind to the sugar moieties of a wide range of glycoproteins [180]. Plant lectins can inhibit virus replication by preventing the adsorption and fusion of HIV in lymphocyte cell cultures [181,182,183,184,185,186,187,188,189]. Furthermore, the antiviral effect of agglutinins specific for N-acetylglucosamine and mannose on HIV has been reported. The inhibitory effect of these plant lectins has been shown in vitro on infection with influenza A virus, respiratory syncytial virus, and cytomegalovirus [188,189,190]. The SARS-CoV spike protein contains 23 putative N-glycosylation sites and is heavily glycosylated. Among the putative N-glycosylation sites, 12 have been defined to be glycosylated [191]. It can be expected that the infectivity of the coronavirus will be suppressed by those lectins that are specific to the glycans present in the spike glycoprotein. The antiviral effect of mannose-specific plant lectins has been reported against coronavirus.

7.2. Quercetin

Quercetin is a plant-derived flavonol (pigment) that is commonly found in vegetables and fruits. It is known to possess an antioxidant, antiviral, anti-inflammatory, and anti-carcinogenic effects, which may reduce risk of infection and improve physical or mental performance. Moreover, quercetin facilitates the ability to stimulate mitochondrial biogenesis and inhibit capillary permeability, platelet aggregation, and lipid peroxidation [20]. Quercetin is widely found in leaves, flowers, barks, nuts, and seeds of a variety of plants such as tomatoes, tea, shallots, grapes, capers, Brassica vegetables, berries, apples, Sambucus canadensis, Hypericum perforatum, and Ginkgo biloba [192]. The recommended consumption of this plant-derived flavonol has been stated to be not less than 4.37 mg/day. Though the highest concentration of quercetin is reported in capers (234 mg of flavonol per 100 g of edible portion), the main plant source for quercetin glycosides is apples, with 7.4% 13 mg/100 g fruit. During the digestion of food, quercetin and its conjugated metabolites can be converted into a range of metabolites (phenolic acids) by enteric enzymes and bacteria in intestinal mucosal epithelial cells (IMECs) [191]. Quercetin also inhibits the senescence-associated pro-inflammatory response and suppresses stress-induced senescent cells [193]. Numerous in vitro investigations have confirmed the inhibitory impact of quercetin on interleukin 8 (IL-8) and tumor-necrosis factor (TNF-α) production in cells. Additionally, several studies have shown the protective function of this flavonol against inflammation in human umbilical vein endothelial cells (HUVECs), as well as mediation via the downregulation of vascular cell adhesion molecule 1 (VCAM-1) and CD80 expression [20,193]. Quercetin considerably induces the production of derived interferon (IFN) and T helper type 1 (Th-1), and it consequently downregulates Th-2-derived interleukin 4 (IL-4) by normal peripheral blood mononuclear cells. Quercetin has been found to suppress the infection caused by a wide spectrum of influenza strains including A/Puerto Rico/8/34 (H1N1), A/FM-1/47/1 (H1N1), and A/Aichi/2/68 (H3N2) with half-maximal inhibitory concentrations (IC50) of 7.76, 6.22, and 2.74 μg/mL, respectively [20,194,195]. Investigations into influenza mechanisms have shown the positive interactions between the viral HA2 subunit (a mark for antiviral vaccines) and quercetin. This discovery may determine the antiviral potential of quercetin in the early stages of influenza. Furthermore, this anti-viral compound could prevent H5N1 virus entry into the cell [20,194,196].

7.3. Sulforaphane

Sulforaphane is an isothiocyanate (isothiocyanate sulforaphane (SFN)) that has been stated to be an antiviral agent. It has been reported that the osteoblast supporting transcription factor Runx2 is essential for the long-term perseverance of antiviral CD8+ memory T cells [197,198]. An addition, SFN-rich broccoli homogenate attenuated granzyme B production in NK cells that was induced by influenza virus and granzyme B production in NK cells, and granzyme B levels appeared to have negatively interacted with influenza RNA levels in nasal lavage fluid cells [199]. Nasal influenza infection can induce complex cascades of changes in peripheral blood NK cell activation. SFN increases as a result of virus-induced peripheral blood NK cell granzyme B production, which may enhance antiviral defense mechanisms [20,199].

7.4. Resveratrol

Resveratrol is a natural polyphenol found in grapes, mulberry, and peanuts. It is known to have antiviral properties against a variety of viral pathogens in vitro and in vivo [200]. Resveratrol is available in trans- and cis-isomeric forms. The cis-resveratrol isomer is unstable and can be easily transformed into the trans form when it reacts with light. It was demonstrated that resveratrol substantially inhibited MERS-CoV replication in vitro through the inhibition of RNA production, as well as other pleiotropic effects. Studies have shown that indomethacin and resveratrol can act as adjuncts for SARS-CoV-2/COVID-19 [201,202]. Medina-Bolivar et al. developed the hairy root lines from Arachis hypogaea (peanut) for the sustained and reproducible production of resveratrol and resveratrol derivatives [201].

7.5. Baicalin

Baicalin (baicalein glucuronide) accumulates in the roots of Scutellaria baicalensis [202]. Baicalin has been reported as an antioxidant possessing anti-apoptotic properties, and it has been used for pulmonary arterial hypertension treatment [203]. This flavone glucuronide has been reported to have anti-SARS-CoV inhibitory effects comparable to those of interferon-beta 1a, interferon-alpha, and glycyrrhizin. At the same time, this compound has a low toxicity in human cell lines [204]. Baicalin showed considerable anti-viral properties on lipopolysaccharide-activated cells, while the oral application of baicalin expressively increased the survival rate of influenza A virus-infected mice [57]. The in silico analysis of the inhibitory effect of baicalin showed that this flavone inhibits ACE2 in the case of COVID-19 disease. It has been revealed that baicalin can inhibit 3CLPro activity of the SARS-Cov2 virus in vitro [205].

7.6. Glycyrrhizin

Glycyrrhizin (a triterpene saponin) is one of the most important phytochemical components of the Glycyrrhiza glabra (licorice) root [206]. Glycyrrhizin has anti-inflammatory and antioxidant properties used for treatments of different diseases such as jaundice, bronchitis, and gastritis [204]. Glycyrrhizin could block the SARS-Cov virus attachment to the host cells, especially during the initial stage of the viral life cycle [207]. An in silico analysis of glycyrrhizin behavior showed the inhibitory effect of this compound on SARS-Cov2 [208].

7.7. Narcissoside

Narcissoside (synonym: narcissin) is a phytochemical belonging to the group of mono-methoxyflavones. This isorhamnetin-3-O-rutinoside flavonoid is extracted from leaves of various folk plants such as Atriplex halimus L., Gynura divaricate, Caragana spinose, and Manihot escylenta. An in silico analysis demonstrated that narcissoside has inhibitory potential for the viral COVID 19 protein 6W63 [209].

7.8. Curcumin

Curcumin is diarylheptanoid that possesses inflammatory and antioxidant properties and is mainly extracted from Alpinia galanga, Curcuma longa, and Caesalpinia sappan [210]. This active compound has been applied in the treatment of hyperlipidemia, anxiety, arthritis, and metabolic syndrome [211]. Additionally, curcumin displays antibacterial and antiviral properties against Pseudomonas, Streptococcus, and Staphylococcus strains, as well as HIV, hepatitis C, and the influenza virus. Moreover, antiviral properties of this compound have been reported against chikungunya virus (CHIKV), human papillomavirus (HPV), HIV-1 and HIV-2 proteases, emerging arboviruses like the Zika virus (ZIKV), influenza viruses, HIV, HSV-2, and hepatitis viruses [212]. However, due to its rapid elimination, rapid metabolism, and poor absorption, curcumin has poor bioavailability, which reduces its therapeutic effect [213]. It has been reported that the combination of this diarylheptanoid with other chemical compounds like piperine can increase bioavailability (by up to 2000%) and provide multiple benefits to human health [214,215]. This compound can diminish various forms of free radicals, such as reactive nitrogen and oxygen species, and modulate the function of SOD [216], catalase, and GSH enzymes in the neutralization of free radicals [217]. Furthermore, curcumin can block ROS-generating enzyme activities such as xanthine oxidase/hydrogenase and cyclooxygenase/lipoxygenase [20]. It has been reported that this plant-derived compound can inhibit the NF-κB activation caused by numerous inflammatory stimuli such as markers of soluble vascular cell adhesion molecule 1 (sVCAM-1), IL-1 beta, IL-6, and inflammation (soluble CD40 ligand (sCD40L)). The results of studies have shown that curcumin can inhibit SARS-CoV through binding to three different protein receptors: SARS-CoV-2 protease (PDB ID:6LU7), PD-ACE2 (PDB ID: 6VW1), and RBD-S (PDB ID:6LXT) [218].

7.9. Epigallocatechin Gallate

Epigallocatechin gallate or epigallocatechin-3-gallate is the ester of gallic acid and epigallocatechin [219]. This compound possesses activity against neurological diseases, premature aging, metabolic diseases, and inflammation [220]. Epigallocatechin gallate has shown anti-inflammatory and antioxidative properties, facilitated DNA repair and stability, ensured the modifications of miRNAs, and modulated the epigenetic methylation of histones [20,221]. The antiviral activity of this compound has been reported against a broad spectrum of viruses such as hepatitis B virus (HBV; Hepadnaviridae), human papillomavirus (HPV; Papovaviridae), adenovirus (Adenoviridae), and herpes simplex virus (HSV; Herpesviridae). It has been observed that epigallocatechin gallate can inhibit (+)-RNA viruses such as chikungunya virus (CHIKV; Togaviridae), West Nile viruses (WNV; Flaviviridae), dengue virus (DENV; Flaviviridae), Zika virus (ZIKV; Flaviviridae), and hepatitis C virus (HCV; Flaviviridae). On the other hand, it can inhibit (−)-RNA viruses such as influenza virus (Orthomyxoviridae), Ebola virus (EBOV; Filoviridae), and HIV (Retroviridae) [13]. Epigallocatechin gallate inhibits the early stage of infections, such as attachment, entry, and membrane fusion, by interfering with viral membrane proteins [222]. The anti-viral mechanism of this compound may be generated from interactions with helicase, ACE-2, and DNMTs [20].

8. Laboratory Evidence Supporting Application of Medicinal Plants Against Respiratory Disorders

Some investigations have identified the antiviral properties of medicinal plants against bronchitis. For example, it has been demonstrated that Verbascum thapsus, Justicia adhatoda, and Hyoscyamus niger can reduce infections risks associated with influenza viruses [108]. Polyphenol, extracted from Cistus incanus (a Mediterranean plant), possesses anti-influenza activity in MDCK and A549 cell cultures infected with human influenza strains and different subtypes of the avian virus [223]. Similarly, sambucol, extracted from Sambucus nigra, showed positive activity against influenza by boosting immune responses through the secretion of inflammatory cytokines (IL-1 beta, TNF-alpha, IL-6, and IL-8) [224].

8.1. Artemisia annua

Artemisia annua (commonly known as sweet wormwood) is a traditional Chinese medicinal herb. It is a source of the sesquiterpene lactone artemisinin, which is used for the manufacturing of the antimalarial drugs artemether and artesunate [225]. The results of an investigation demonstrated that artemisinin is a reliable source to act as an antiviral compound [226]. Furthermore, sterols extracted from A. annua presented inhibitory potential against viruses [227]. An in vitro investigation on the antiviral properties of A. annua against SARS-CoV showed positive feedback by using an ethanolic extract with a 50% effective concentration (EC50) value of 34.5 ± 2.6 μg/mL and 5a 0% cytotoxic concentration (CC50) of 1053 ± 92.8 μg/mL. This result suggested the possibility of applying A. annua against SARS-CoV infectious diseases [16]. An analysis of antiviral activity of methanolic extracts of A. annua against herpes simplex virus type 1 showed the antiviral properties of the aerial parts of plant. It has been suggested that high levels of bioactive compounds are the main reasons for antiviral activities, which has led to the use of this plant as a potential candidate against viruses [226]. Pulmonary fibrosis (lungs become scarred) is caused by the infection of SARS-CoV with spiked severity, which mediated by interleukin-1 [227]. It has been shown that the consumption of natural antioxidants like polyphenolic compounds [228] is effective in the treatment of lung fibrosis associated with oxidative stress [229]. The positive effect of artesunate, an artemisinin-based drug, to treat pulmonary fibrosis was confirmed due to it inhibiting pro-fibrotic molecules linked to pulmonary fibrosis [230].

8.2. Allium cepa

Allium cepa (commonly known as onion) is a common raw material served in salad and is rich in natural resources of organosulfur and flavonoids with antioxidant activity [231]. Reportedly, quercetin and isorhamnetin (two potential therapeutic agents in onion) can reduce blood pressure and prevent angiotensin-II-induced endothelial dysfunction. Furthermore, superoxide production was increased and subsequently and led to a high nitric oxide bioavailability [232]. It has been reported that the chemical compounds of onion-like flavonoids or even prolines possess antiviral properties against respiratory viruses by stimulating proinflammatory cytokines [233].

8.3. Andrographis paniculata

Andrographis paniculata (which is commonly known as King of Bitters) is extensively applied in the treatment of several ailments like liver disorders, viral fever, cold, and cough [231]. Andrographis paniculata has shown a strong therapeutic effect against viral respiratory infections [234,235,236,237] by suppressing interleukin-1β molecules and increasing NOD-like receptor protein 3 (NLRP3) and caspase-1, which are widely known to play roles in SARS-CoV and likely SARS-CoV-2 pathogenesis [238,239]. Andrographolide (a diterpenoid), is the main bioactive compound extracted from the leaves and stem of this plant and possesses anti-inflammatory capacity. Notably, andrographolide has antiviral potential against different viruses diseases such as chikungunya virus, human immunodeficiency virus, human papillomavirus, Epstein-Barr virus, herpes simplex virus, hepatitis C and B, and influenza A virus (H1N1, H5N1, and H9N2) [125].

8.4. Aloe vera

Aloe vera is a kind of medicinal plant that possesses antiviral activity against different viruses including human papillomavirus, cytomegalovirus, poliovirus, influenza virus, human immunodeficiency virus, varicella-zoster virus, herpes simplex virus type 2, herpes simplex virus type 1, and hemorrhagic viral rhabdovirus septicemia. Molecular investigations have shown the effectiveness of this plant against other viruses by different action mechanisms such as the breakdown of the viral envelope and interactions with virus enzymes. The presence of some minerals like zinc, copper (Cu), iron (Fe), potassium (K), sodium (Na), magnesium (Mg), and calcium (Ca) make Aloe vera a suitable candidate against SARS-CoV-1 [240]. Reports have shown that Zn2+ blocks arterivirus RNA polymerase and SARS-CoV activity and inhibits SARS-CoV replication in cell lines [241] (Figure 3).

8.5. Nigella sativa

Nigella sativa (black seed) is a kind of medicinal plant used for the treatment of a variety of diseases, disorders, and conditions pertaining to the respiratory system, immune systems and cardiovascular, liver, kidneys, and digestive tract. Recently, the result of studies showed that the oil extract of N. sativa reduced the H9N2 avian influenza virus (which is fundamentally associated to SARS-CoV-2 pathogenicity in chicken), and consequently supported the immune response. The most important active compounds which have been identified in N. sativa are monoterpenes, e.g., t-anethol, 4-terpineol, carvacrol, p-cymene, thymohydroquinone, and thymoquinone or dimers thereof like dithymoquinone [20].

8.6. Salvia officinalis

Salvia officinalis (sage) is a medicinal plant with antiviral, antibacterial, antimalarial and antifungal effects. Reportedly, the antiviral and antifungal properties of this plant are most probably facilitated by sageone, safficinolide, and diterpenoids [242].

8.7. Toona sinensis

The crude oil of the tender leaves of Toona sinensis Roem induced apoptosis in A549 lung cancer cells but also improved the lipolysis of differentiated 3T3-L1 adipocytes [243,244]. Further, the leaf extract of T. sinensis Roem was reported to relieve hyperglycemia via modifying adipose glucose transporter 4 [245]. Studies of purified compounds of T. sinensis leaves have shown several compounds include toosendanin, phytol, stigmasterol glucoside, beta-sitosterol-glucoside, stigmasterol, beta-sitosterol, (−)-epicatechin, (+)-catechin, kaempferol-D-glucoside, rutin, quercitrin, quercetin, kaempferol, gallic acid, and methyl gallate [246]. Additionally, it has been reported that, quercetin, one of T. sinensis leaves’ compounds, has an antiviral impact against HIV-luc/SARS pseudotyped virus [65].

8.8. Eckolina cava

Eckolina cava (Laminariaceae) is a type of brown alga with anti-viral activity against influenza virus neuraminidase and HIV-1 reverse transcriptase 11 phlorotannin chemotype (a diphenyl ether-linked dieckol) extracted from E. cava (IC50s = 2.7 and 68.1 µM) that was found to highly block the cleavage of SARS-CoV 3CLpro in a cell-based test with no toxicity [182].

8.9. Isatis indigotica

Isatis indigotica (I. indigotica root Radix isatidis) is a Chinese medicinal plant belonging to the family of Cruciferae, with a high phenolic content in the root. The root of this plant was frequently applied during the outbreak of SARS in Taiwan, Hong Kong, and China. Additionally, the antiviral effects of different compounds of this plant such as sinigrin, γ-sitosterol, indican (indoxyl-β-D-glucoside), β-sitosterol, indirubin, and indigo have been reported against Japanese encephalitis, hepatitis A, and influenza [247,248]. Indirubin and indigo were recognized as inhibitors of promiscuous chymotrypsin [249]. Moreover, the antiviral effects of naringenin, quercetin, hesperetin, phenolics, and aloe emodin derived from I. indigotica have been accredited against parainfluenza virus, sindbis virus, herpes simplex virus types 1 and 2, vesicular stomatitis virus, poliovirus, and vaccinia virus [250,251]. It has been well-demonstrated that the 3C-like protease (3CLpro) can mediate the proteolytic processing of polypeptides 1a and 1ab into functional proteins, which is an ideal target for the development of drugs against SARS-coronavirus. In a cell-based assay, the investigation of seven different phenolic compounds derived from I. indigotica revealed that only two polyphenols, namely hesperetin (8.3 µM) and emodin (366 µM), could inhibit the cleavage activity of the 3CLpro in a dose-dependent manner [163].

8.10. Azadirachta indica

Azadirachta indica (commonly known as neem) is a kind of biological antiviral agent against duck plague virus, herpes simplex virus type-1, bovine herpesvirus type-1 (BoHV-1), poliovirus type 1, group B coxsackieviruses, polio, and dengue virus type-2, as well as infectious bursal diseases like viral infections, Newcastle disease, and highly pathogenic avian influenza virus (H5N1) [252]. It has been shown that different extracts of this neem’s explants have potential against common clinical symptoms of Covid-19 [253,254]. In this regard, crude leaf extracts of neem could be effective against malarial and normal fever [246], as well as a gastrointestinal disorders [255]. The leaf extract of this plant possesses strong antioxidant potential by directly scavenging the hydroxyl radical and preventing hydroxyl radical-mediated oxidative damage in the rat model [256].

8.11. Other Medicinal Herbs

Studies on some medicinal plants including Evolvulus alsinoides, Pergularia daemi, Clerodendrum inerme Gaertn, Clitoria ternatea, Sphaeranthus indicus, Cassia alata, Leucas aspera, Abutilon indicum, Gymnema sylvestre, Vitex trifolia, and Indigofera tinctoria (AO) have shown anti-mouse coronaviral activity [107]. C. inerme Gaertn was observed as a promising medicinal plant having potential to inactivate the viral ribosome [104]. Additionally, anti-inflammatory cytokines are significantly reduced by Sphaeranthus indicus and Vitex trifolia when using the nuclear factor kappa-light-chain-enhancer of activated B (NF-kB) signaling cascade, associated with acute respiratory distress syndrome [152] in SARS-CoV [256,257]. Furthermore, C. ternatea (Asian pigeonwings) has been reported as an inhibitor of metallopeptidase domain 17 (ADAM17), a metalloproteinase involved in angiotensin being converted for enzyme shredding, and can be targeted with Clitoria ternatea. ACE-2 shredding is associated with the spike formation of viruses [105]. Likewise, Strobilanthes cusia was found to inhibit viral RNA genome synthesis and to induce papain like protease (PLpro) activity targeting the human coronavirus OC43 [1,113]. Allium sativum and Glycyrrhiza glabra have been identified to inhibit the viral replication of SARS-CoV. Inhibitory effects on the Ca2+ channel were observed when Hyoscyamus niger was applied as a bronchodilator [108]. This plant is able to target the orf3a Ca2+ channels that attack downstream pathways upon viral infection. The inhibitory effect of other medicinal plants including Embelia ribes, Cassia occidentalis, Punica granatum, Coscinium fenestratum, Cynara scolymus, Boerhaavia diffusa, and Coriandrum sativum have been identified against ACE [107]. However, Punica granatum has been shown to exhibit a competitive style of action against virus infection [102,103]. Salacia oblonga was found to exhibit suppressive impacts on angiotensin II, an AT1 signal, which was related to lung damage [109] (Table 4).

9. In Silico Analysis of Medicinal Plants Role Against SARS-CoV

The in silico analysis of 18 extracted compounds of 11 Indian herbal plants demonstrated different inhibitory properties against Covid-19. Based on the data achieved through log S and log P, as well as the binding affinity (Table 5), the potential inhibitory effect of plants were in the following order: Nyctanthes arbortristis (harsingar) > Aloe barbadensis miller (aloe vera) > Tinospora cordifolia (giloy) > Curcuma longa (turmeric) > Azadirachta indica (neem) > Withania somnifera (ashwagandha) > Allium cepa (red onion) > Ocimum sanctum (tulsi) > Cannabis sativa (cannabis) > Piper nigrum (black pepper). However, the results of this investigation confirmed that harsingar, Aloe vera, and giloy are more reliable natural resources for future investigations [305]. The in silico analysis of compounds with anti-viral, anti-malaria, or other similar activities are presented in Table 5.
On the other hand, an in silico molecular docking study on thirty-six phytochemical compounds showed high binding affinities for betulinic acid (−10.0 Kcal/mol), silibinin (−9.13 Kcal/mol), oleanolic acid (−9.08 Kcal/mol), and epigallocatechin-3-gallate (−8.51 Kcal/mol). These results suggested that the medicinal plants containing the mentioned compounds are potential candidates against Covid-19 [306].

10. Biotechnological Production of Vaccines

At time of writing of this document, no antiviral medicines have been proven to be effective against Covid-19 [307]. As mentioned earlier, remdesivir and hydroxychloroquine have been documented as the most reliable candidates against SARS-CoV [308]. On the other hand, a lot of efforts have been made to develop monoclonal antibodies and to evaluate receptor blockers [309]. Additionally, the use of transfusions by employing the plasma of recovered donor from the SARS-CoV virus infection is under assessment [310]. Over the last few decades, therapeutic vaccines have been revealed as the most effective tool against infectious diseases [311]. Due to the high transmissibility of SARS-CoV, the discovery of novel vaccines is an urgent target to tackle this pathogen [312]. A traditional way to generate vaccine is the method of inactivated vaccines, which can be made with SARS-CoV-2 virions previously inactivated by physical or chemical treatments [313]. Currently, an attenuated vaccine that is generated by reducing the microbial virulence of a pathogen is known [314]. Biotechnology is mostly applied to generate vaccines in three ways: the separation of a pure antigen using a specific monoclonal antibody, the synthesis of an antigen with the assistance of a cloned gene, and the synthesis of peptides to be used as vaccines. Furthermore, producing active pharmaceutical chemical compounds in genetically modified organisms is a novel application of biotechnology achieved by molecular pharming or hairy root technologies.

10.1. Molecular Pharming: A Mature Technology to Produce Plant-Derived Pharmaceutical Products

Molecular pharming is one of the biotechnology tools that involves the application of various plant species for the production of recombinant proteins, which include enzymes, hormones, vaccines, and antibodies [315] (Figure 4).
Over the last few decades, a lot of efforts have been made to produce different bioactive compounds and proteins in high yield through plant genetic engineering approaches [316]. The molecular pharming platform has several advantages over other transgenic systems such as a low overall cost, a high scale-up capacity, safety, high product quality, and the ability of post-translational modifications [317]. In 2005, Pogrebnyak and colleagues [17] tried to develop a recombinant vaccine against SARS by expressing the N-terminal fragment of the SARS-CoV S protein (S1) in tomatoes and low-nicotine tobacco plants. The results of their experiment showed the high expression of recombinant S1 protein in transgenic lines. The production of plant-derived antigens significantly enhanced the levels of SARS-CoV-specific IgA after the oral ingestion of tomato fruits expressing the S1 protein. Sera of mice parenterally primed with tobacco-derived S1 protein revealed the presence of SARS-CoV-specific IgG as observed with Western blot and ELISA analysis. Two companies have announced their effort to produce plant-derived antibodies and vaccines against SARS-CoV-2. Soon after reporting the SARS-CoV-2 virus S protein, Medicago Inc. (www.medicago.com/en/pipeline/, the accessed date: 2 December 2020) announced VLP production in a transient gene expression system. Based on estimation, this company should be able to produce an attractive production capacity of VLP based vaccines (10 million doses). This amount of plant-derived vaccine can immunize approximately 10 million US adults who are at severe risk of critical illness or death if infected with SARSCoV-2. Furthermore, developing epitope-based vaccines is reported to reduce disease possibility. Kentucky Bioprocessing (www.kentuckybioprocessing.com, the accessed date: 2 December 2020), protalix (www.protalix.com, the accessed date: 2 December 2020), Greenovation Biopharmaceuticals (www.greenovation.com, the accessed date: 2 December 2020), Ventria (www.ventria.com, the accessed date: 2 December 2020), Nomad www.nomadbioscience.com, the accessed date: 2 December 2020), and iBio Inc. (www.ibioinc.com/pipeline, the accessed date: 2 December 2020) are other companies that are making efforts to produce a VLP vaccine. On the other hand, academic institutions such as the Infectious Disease Research Centre at Laval University and Medicago have joined hands together to develop plant-derived pipelines. Simultaneously, several institutes and universities from different countries including Thailand, Mexico, South Korea, South Africa, the UK, Germany, and the US are working on the production of plant-derived products through molecular farming technology against the Covid-19. It is believed that producing a stable plant-derived product through molecular farming takes a long time, and a major concern is that these products are not as approachable as expected in response to the pandemic. However, applying transient systems should be a reliable, fast, and efficient tool to overcome these barriers. Due to the high adaptability and potential of the SARS-CoV-2 virus, it seems that oral vaccines like plant molecular farming products are the most reliable weapons against the SARS-CoV-2 virus [318].

10.2. Hairy Root Culture: A Reliable Method of Producing Pharmaceutical Products

Hairy roots, which are induced by Rhizobium rhizogenes (syn. Agrobacterium rhizogenes), a Gram-negative bacterium, comprise an important biotechnological system to induce bioactive compounds from plants. Hairy root cultures employ organs instead of undifferentiated cells, thereby providing better yields than cell culture or natural plant roots [318,319]. Other advantages include biochemical and genetic stability, ease of preservation, and rapid growth on hormone-free media. Different Agrobacterium strains, which are divergent in terms of virulence, the growth rate of transformed cells, their morphology, and secondary metabolite production, are generally employed to induce hairy roots from explants in different plant species. It has been recognized that Agrobacterium strains have different abilities to promote the production of bioactive compounds in hairy root cultures [302,320,321]. This is mainly due to the differential expression and integration of T-DNA genes into the plant genome [302,321]. Therefore, the selection of an effective Agrobacterium strain for hairy root induction and secondary compound accumulation is noticeably dependent on plant species and must be empirically figured out [302,322] (Figure 5).
Mentha spicata, commonly known as spearmint or garden mint, is known to possess antiviral properties due to the presence of high amounts of phenolic compounds such as rosmarinic acid, chlorogenic acid, caffeic acid, lithospermic acid B, and cinnamic acid. The production of phenolic acids in M. spicata was compared among hairy roots induced from five different A. rhizogenes strains: ATCC15834, 9534, A13, A4, and R318. It was found that A. rhizogenes strain 9534 efficiently produced caffeic acid, lithospermic acid B, cinnamic acid, rosmarinic acid, and chlorogenic acid (106.76, 60.22, 44.02, 20.08, and 13.53 μg g−1 DW, respectively), but hairy root induction was effective with A13 and R318 [323,324,325,326].
It has been shown in many plant species that transformation by A. rhizogenes strains leads to the modification of metabolic pathways. As a result, transformed roots differ in their chemical profiles from normal roots, which shows that Ri T-DNA interferes with the biosynthesis of secondary metabolites [324,325,326].
The effects of different media (Schenk and Hildebrandt (SH), Woody plant medium (WPM), and Gamborg (B5)) and lighting conditions (light or dark) on biomass accumulation and secondary metabolite production in hairy root line (RC3) were examined in Rhaponticum carthamoides [326]. The WPM supported the highest biomass (93 g L−1 of the fresh weight after 35 days) under periodic light. Additionally, the higher production of caffeoylquinic acids and their derivatives was observed in hairy roots grown in the light as compared to untransformed roots. The biosynthesis of flavonoid glycosides such as quercetagetin, quercetin, luteolin, and patuletin hexoside from transformed roots was also found in light conditions [326]. Balasubramanian et al. [327] showed that time, number of co-cultivation days, acetosyringone concentration, media type, media strength, and sucrose concentration affect hairy root production for the improvement of quercetin content in Raphanus sativus (radish). Explants infected with an A. rhizogenes MTCC 2364 suspension for 10 min and co-cultivated in a 1/2 MS medium containing acetosyringone (100 µM) for two days displayed a maximum percentage of hairy root induction (77.6%). Hairy roots were found to produce higher amounts of quercetin (114.8 mg g−1) compared to the auxin-induced roots of non-transformed radish. Similarly, higher amounts of phenolic compounds including pyrogallol, hesperidin, naringenin, and formononetin were observed in Polygonum multiflorum hairy roots compared to untransformed roots [328].
Because it has been found that both endogenous and exogenous factors affect resveratrol production in the hairy root culture of grapes and increase resveratrol production, this strategy could be useful [329]. Different features like the type of explants, seedling age, concentration of the bacterial inoculum, and inoculation time have been studied to improve the efficiency of hairy root formation and resveratrol production. Hairy roots induced by A. rhizogenes strain A4 from internodes of Vitis vinifera subsp. sylvesteris (W16) were found to produce the highest amount of resveratrol (31-fold higher than that of control root) [329]. In Scutellaria baicalensis, hairy roots grown in a full-strength MS medium produced a 2.5-fold higher amount of resveratrol than those grown in a half-strength B5 medium. The addition of auxin (indole acetic acid (IAA)) at 0.1 mg/L into the medium resulted in the highest accumulation of resveratrol [323,330].
The elicitation of hairy root cultures by biotic/abiotic stressors or chemicals for the exploitation of genetic engineering manipulation can significantly increase the amount of the desired secondary compound [331]. Different elicitors such as UV radiation, methyl jasmonate (MeJA), jasmonic acid, gibberellic acid, sodium acetate, salicylic acid, acetic acid, ammonium nitrate, chitosan, and cyclodextrins have been employed to stimulate the production of secondary metabolites in hairy roots [332]. Chitosan, methyl jasmonate, and yeast extract have been employed to enhance the glycyrrhizin contents in Glycyrrhiza species. Glycyrrhizin, an active component of licorice roots, is known to be effective against several viral diseases including HIV and SARS [104]. It has been reported that glycyrrhizin also inhibits the adsorption and penetration of the virus-early steps of the replicative cycle in addition to the inhibition of virus replication [104]. Elicitation with 100 μM MeJA enhanced glycyrrhizin content (5.7 times higher than the control) after five days of treatment in G. inflata hairy roots [333]. MeJA (at 100 μM concentration) was found to be the most effective elicitor for increasing glycyrrhizin production up to 108.9 ± 1.15 μg g−1 DW after five days of elicitation [334].
Elicitation with sodium acetate for 24 h resulted in the 60-fold induction and secretion of trans-resveratrol into a peanut hairy root culture medium [201]. The authors [335] studied the effect of different concentrations of abiotic elicitors including MeJA, sodium acetate, acetic acid, and ammonium nitrate. Their results showed that treatment of hairy roots with 3 mM acetic acid or 50 μM methyl jasmonate resulted in the highest or lowest amounts of hairy root biomass and resveratrol content, respectively.

11. Conclusions

The current review provides an overview of employing ethnomedicinal herbs to treat respiratory diseases. It has been shown that plant biodiversity can be a source of bioactive compounds of different natures, such as terpenes, stilbenes, coumarins, flavone glycosides, and alkaloids. The most common of these are lectins, quercetin, resveratrol, glycyrrhizin, and curcumin. It has been found that qualitative and quantitative biochemical specification can vary between representatives of the same family. Such a literature analysis could be used to choose an appropriate plant model for a specific region with specific needs. With the intervention of biotechnological tools such as hairy root transformation, it is possible to obtain compounds of interest in a higher amount. Plant-based production systems comprise another alternative for molecular farming technology. This review not only provides a reference point for the screening of plants against viral diseases but can also be useful for practical implications and applications.

12. Future Directions

The present review describes the potential of plant or plant-based compounds for treating antiviral diseases, especially emphasizing SARS-CoV. The urgency of this work is that new viruses may appear in the near future. There is a gap between information regarding the complete understanding of the mechanism of action of plant-based compounds and their execution as treatments of viral diseases. Both empirical and rational approaches are required to gain insight into the phytochemical evaluation and validation of plant-based compounds. Critical knowledge of genetic, molecular, and biochemical processes under in situ conditions could be helpful to better control the accumulation of natural products. Future studies may be targeted at understanding the mechanisms of action of complex plant extracts and isolated plant-derived compounds from different biosynthetic pathways. Molecular docking studies and in silico analyses of extracted compounds from different herbal plants including aloe vera and giloy could be beneficial for further investigations. By employing biotechnological tools like hairy root cultures and molecular pharming, it is quite possible to enhance the production of the compounds of interest. No doubt, careful preclinical and clinical procedures need to be followed before coming to the validation of plant-based drugs or vaccines against these viral diseases. This would help to pave the way and take important steps to combat this life-threatening and deadly disease.

Author Contributions

Conceptualization, R.A. (Rambod Abiri), and S.M.; methodology, R.A. (Rambod Abiri), O.S. and R.A. (Ramin Abiri); investigation, S.M., R.A. (Rambod Abiri), H.A.-H., S.M. and R.A. (Ramin Abiri); resources, H.A.-H.; writing—original draft preparation, R.A. (Rambod Abiri), O.S., S.M., R.A. (Ramin Abiri), E.B.d.A.J., S.K.S. and R.R.J.A.; writing—review and editing, E.B.d.A.J.; S.K.S., V.P.B. and S.M.; visualization, R.A. (Ramin Abiri), S.M. and V.P.B.; supervision, H.A.-H., V.P.B. and S.M. All authors have read and agreed to the published version of the manuscript.

Funding

This review paper received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank Universiti Putra Malaysia (UPM) and Federal University of Maranhão for supporting the present study.

Conflicts of Interest

The authors report no declarations of interest.

References

  1. Amber, R.; Adnan, M.; Tariq, A.; Mussarat, S. A review on antiviral activity of the Himalayan medicinal plants traditionally used to treat bronchitis and related symptoms. J. Pharm. Pharmacol. 2017, 69, 109–122. [Google Scholar] [CrossRef] [PubMed]
  2. Zhu, N.; Zhang, D.; Wang, W.; Li, X.; Yang, B.; Song, J.; Zhao, X.; Huang, B.; Shi, W.; Lu, R.; et al. A Novel Coronavirus from Patients with Pneumonia in China, 2019. N. Engl. J. Med. 2020, 382, 727–733. [Google Scholar] [CrossRef] [PubMed]
  3. Peeri, N.C.; Shrestha, N.; Rahman, S.; Zaki, R.; Tan, Z.; Bibi, S.; Baghbanzadeh, M.; Aghamohammadi, N.; Zhang, W.; Haque, U. The SARS, MERS and novel coronavirus (COVID-19) epidemics, the newest and biggest global health threats: What lessons have we learned? Int. J. Epidemiol. 2020, 49, 717–726. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Li, J.; Liu, W. Puzzle of highly pathogenic human coronaviruses (2019-nCoV). Protein Cell 2020, 11, 235–238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Sathya, B.; Velpandian, V.; Ramani, M.; Kumar, M.P. A primitive approach on review of Siddha herbs, herbo-mineral formulation exhibiting antiviral activity. Int. J. Pharm. Bio. Sci. 2014, 5, 138–147. [Google Scholar]
  6. Forman, R.; Shah, S.; Jeurissen, P.; Jit, M.; Mossialos, E. COVID-19 vaccine challenges: What have we learned so far and what remains to be done? Health Policy 2021, 125, 553–567. [Google Scholar] [CrossRef] [PubMed]
  7. Khan, A.; Khan, M.; Saleem, S.; Babar, Z.; Ali, A.; Khan, A.A.; Sardar, Z.; Hamayun, F.; Ali, S.S.; Wei, D.-Q. Phylogenetic Analysis and Structural Perspectives of RNA-Dependent RNA-Polymerase Inhibition from SARs-CoV-2 with Natural Products. Interdiscip. Sci. Comput. Life Sci. 2020, 12, 335–348. [Google Scholar] [CrossRef]
  8. Khan, A.; Tahir Khan, M.; Saleem, S.; Junaid, M.; Ali, A.; Shujait Ali, S.; Khan, M.; Wei, D.Q. Structural insights into the mechanism of RNA recognition by the N-terminal RNA-binding domain of the SARS-CoV-2 nucleocapsid phosphoprotein. Comput. Struct. Biotechnol. J. 2020, 18, 2174–2184. [Google Scholar] [CrossRef]
  9. Alrasheid, A.A.; Babiker, M.Y.; Awad, T.A. Evaluation of certain medicinal plants compounds as new potential inhibitors of novel corona virus (COVID-19) using molecular docking analysis. Silico Pharmacol. 2021, 9, 10. [Google Scholar] [CrossRef]
  10. Khan, M.T.; Zeb, M.T.; Ahsan, H.; Ahmed, A.; Ali, A.; Akhtar, K.; Malik, S.I.; Cui, Z.; Ali, S.; Khan, A.S.; et al. SARS-CoV-2 nucleocapsid and Nsp3 binding: An in silico study. Arch. Microbiol. 2021, 203, 59–66. [Google Scholar] [CrossRef]
  11. Khan, M.T.; Ali, A.; Wang, Q.; Irfan, M.; Khan, A.; Zeb, M.T.; Zhang, Y.-J.; Chinnasamy, S.; Wei, D.-Q. Marine natural compounds as potents inhibitors against the main protease of SARS-CoV-2—A molecular dynamic study. J. Biomol. Struct. Dyn. 2020, 1–11. [Google Scholar] [CrossRef]
  12. Wang, X.; Liu, Z. Prevention and treatment of viral respiratory infections by traditional Chinese herbs. Chin. Med. J. 2014, 127, 1344–1350. [Google Scholar]
  13. Remali, J.; Aizat, W.M. A Review on Plant Bioactive Compounds and Their Modes of Action Against Coronavirus Infection. Front. Pharmacol. 2021, 11, 589044. [Google Scholar] [CrossRef]
  14. Chojnacka, K.; Witek-Krowiak, A.; Skrzypczak, D.; Mikula, K.; Młynarz, P. Phytochemicals containing biologically active polyphenols as an effective agent against Covid-19-inducing coronavirus. J. Funct. Foods 2020, 73, 104146. [Google Scholar] [CrossRef]
  15. Siddiqui, M.H.; Alamri, S.; Al-Whaibi, M.H.; Hussain, Z.; Ali, H.M.; El-Zaidy, M.E. A mini-review of anti-hepatitis B virus activity of medicinal plants. Biotechnol. Biotechnol. Equip. 2016, 31, 9–15. [Google Scholar] [CrossRef] [Green Version]
  16. 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. Identification of natural compounds with antiviral activities against SARS-associated coronavirus. Antivir. Res. 2005, 67, 18–23. [Google Scholar] [CrossRef]
  17. Pogrebnyak, N.; Golovkin, M.; Andrianov, V.; Spitsin, S.; Smirnov, Y.; Egolf, R.; Koprowski, H. Severe acute respiratory syndrome (SARS) S protein production in plants: Development of recombinant vaccine. Proc. Natl. Acad. Sci. USA 2005, 102, 9062–9067. [Google Scholar] [CrossRef] [Green Version]
  18. Li, H.-Y.; Ramalingam, S.; Chye, M.-L. Accumulation of recombinant SARS-CoV spike protein in plant cytosol and chloroplasts indicate potential for development of plant-derived oral vaccines. Exp. Biol. Med. 2006, 231, 1346–1352. [Google Scholar] [CrossRef]
  19. Vignuzzi, M.; López, C.B. Defective viral genomes are key drivers of the virus–host interaction. Nat. Microbiol. 2019, 4, 1075–1087. [Google Scholar] [CrossRef]
  20. Haslberger, A.; Jacob, U.; Hippe, B.; Karlic, H. Mechanisms of selected functional foods against viral infections with a view on COVID-19: Mini review. Funct. Foods Heal. Dis. 2020, 10, 195. [Google Scholar] [CrossRef]
  21. Leung, A.K.; Hon, K.; Leong, K.; Sergi, C. Measles: A disease often forgotten but not gone. Hong Kong Med. J. 2018, 24, 512–520. [Google Scholar] [CrossRef] [Green Version]
  22. Roy, P. Bluetongue Virus Proteins and Particles and Their Role In Virus Entry, Assembly, And Release. Int. Rev. Cytol. 2005, 64, 69–123. [Google Scholar] [CrossRef]
  23. Li, T.; Peng, T. Traditional Chinese herbal medicine as a source of molecules with antiviral activity. Antivir. Res. 2013, 97, 1–9. [Google Scholar] [CrossRef]
  24. Xu, Z.; Peng, C.; Shi, Y.; Zhu, Z.; Mu, K.; Wang, X.; Zhu, W. Nelfinavir was predicted to be a potential inhibitor of 2019-nCov main protease by an integrative approach combining homology modelling, molecular docking and binding free energy calculation. bioRxiv 2020. [Google Scholar] [CrossRef] [Green Version]
  25. Rodríguez-Morales, A.J.; MacGregor, K.; Kanagarajah, S.; Patel, D.; Schlagenhauf, P. Going global—Travel and the 2019 novel coronavirus. Travel Med. Infect. Dis. 2020, 33, 101578. [Google Scholar] [CrossRef]
  26. Gould Maule, G. Investigation of Host Factors Required for Lymphocytic Choriomeningitis Virus Entry. Master’s Thesis, University of Ottawa, Ottawa, ON, Canada, 2019. [Google Scholar] [CrossRef]
  27. Coutard, B.; Valle, C.; De Lamballerie, X.; Canard, B.; Seidah, N.; Decroly, E. The spike glycoprotein of the new coronavirus 2019-nCoV contains a furin-like cleavage site absent in CoV of the same clade. Antivir. Res. 2020, 176, 104742. [Google Scholar] [CrossRef]
  28. Vankadari, N.; Wilce, J.A. Emerging COVID-19 coronavirus: Glycan shield and structure prediction of spike glycoprotein and its interaction with human CD26. Emerg. Microbes Infect. 2020, 9, 601–604. [Google Scholar] [CrossRef] [PubMed]
  29. Sargiacomo, C.; Sotgia, F.; Lisanti, M.P. COVID-19 and chronological aging: Senolytics and other anti-aging drugs for the treatment or prevention of corona virus infection? Aging 2020, 12, 6511–6517. [Google Scholar] [CrossRef] [PubMed]
  30. Ortiz-Prado, E.; Simbaña-Rivera, K.; Gómez-Barreno, L.; Rubio-Neira, M.; Guaman, L.P.; Kyriakidis, N.C.; López-Cortés, A. Clinical, molecular and epidemiological characterization of the SARS-CoV2 virus and the Coronavirus disease 2019 (COVID-19), a comprehensive literature review. Diagn. Microbiol. Infect. Dis. 2020, 98, 115094. [Google Scholar] [CrossRef] [PubMed]
  31. Liu, X.; Wang, X.-J. Potential inhibitors against 2019-nCoV coronavirus M protease from clinically approved medicines. J. Genet. Genom. 2020, 47, 119–121. [Google Scholar] [CrossRef]
  32. Chang, K.-O.; Kim, Y.; Lovell, S.; Rathnayake, A.D.; Groutas, W.C. Antiviral Drug Discovery: Norovirus Proteases and Development of Inhibitors. Viruses 2019, 11, 197. [Google Scholar] [CrossRef] [Green Version]
  33. Li, J.-Y.; You, Z.; Wang, Q.; Zhou, Z.-J.; Qiu, Y.; Luo, R.; Ge, X.-Y. The epidemic of 2019-novel-coronavirus (2019-nCoV) pneumonia and insights for emerging infectious diseases in the future. Microbes Infect. 2020, 22, 80–85. [Google Scholar] [CrossRef]
  34. Wang, Z.; Chen, X.; Lu, Y.; Chen, F.; Zhang, W. Clinical characteristics and therapeutic procedure for four cases with 2019 novel coronavirus pneumonia receiving combined Chinese and Western medicine treatment. Biosci. Trends 2020, 14, 64–68. [Google Scholar] [CrossRef] [Green Version]
  35. Loizzo, M.R.; Saab, A.M.; Tundis, R.; Statti, G.A.; Menichini, F.; Lampronti, I.; Gambari, R.; Cinatl, J.; Doerr, H.W. Phytochemical Analysis andin vitro Antiviral Activities of the Essential Oils of Seven Lebanon Species. Chem. Biodivers. 2008, 5, 461–470. [Google Scholar] [CrossRef]
  36. Khaerunnisa, S.; Kurniawan, H.; Awaluddin, R.; Suhartati, S.; Soetjipto, S. Potential inhibitor of COVID-19 main protease (Mpro) from several medicinal plant compounds by molecular docking study. Preprints 2020. [Google Scholar] [CrossRef] [Green Version]
  37. Zimmerman, A.E.; Howard-Varona, C.; Needham, D.M.; John, S.G.; Worden, A.Z.; Sullivan, M.B.; Waldbauer, J.R.; Coleman, M.L. Metabolic and biogeochemical consequences of viral infection in aquatic ecosystems. Nat. Rev. Genet. 2020, 18, 21–34. [Google Scholar] [CrossRef]
  38. Feng, Y.; Gao, G.F. Towards our understanding of SARS-CoV, an emerging and devastating but quickly conquered virus. Comp. Immunol. Microbiol. Infect. Dis. 2007, 30, 309–327. [Google Scholar] [CrossRef]
  39. Kpewou, D.E. Antioxidant Micronutrients Intake in People Living with HIV: Implications on Serum Levels and Liver Function. Master’s Thesis, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana, October 2017. [Google Scholar]
  40. Chantrill, B.H.; Coulthard, C.E.; Dickinson, L.; Inkley, G.W.; Morris, W.; Pyle, A.H. The Action of Plant Extracts on a Bacteriophage of Pseudomonas pyocyanea and on Influenza A Virus. J. Gen. Microbiol. 1952, 6, 74–84. [Google Scholar] [CrossRef] [Green Version]
  41. Debiaggi, M.; Pagani, L.; Cereda, P.M.; Landini, P.; Romero, E. Antiviral activity of Chamaecyparis lawsoniana extract: Study with herpes simplex virus type 2. Microbiology 1988, 11, 55–61. [Google Scholar]
  42. Vermani, K.; Garg, S. Herbal medicines for sexually transmitted diseases and AIDS. J. Ethnopharmacol. 2002, 80, 49–66. [Google Scholar] [CrossRef]
  43. Asres, K.; Bucar, F. Anti-HIV activity against immunodeficiency virus type 1 (HIV-I) and type II (HIV-II) of compounds isolated from the stem bark of Combretum molle. Ethiop. Med. J. 2005, 43, 15–20. [Google Scholar] [PubMed]
  44. Kotwal, G.J.; Kaczmarek, J.N.; Leivers, S.; Ghebremariam, Y.T.; Kulkarni, A.P.; Bauer, G.; De Beer, C.; Preiser, W.; Mohamed, A.R. Anti-HIV, Anti-Poxvirus, and Anti-SARS Activity of a Nontoxic, Acidic Plant Extract from the Trifollium Species Secomet-V/anti-Vac Suggests That It Contains a Novel Broad-Spectrum Antiviral. Ann. N. Y. Acad. Sci. 2005, 1056, 293–302. [Google Scholar] [CrossRef] [PubMed]
  45. Kwon, D.H.; Kwon, H.Y.; Kim, H.J.; Chang, E.J.; Kim, M.B.; Yoon, S.K.; Song, E.Y.; Yoon, D.Y.; Lee, Y.H.; Choi, I.S.; et al. Inhibition of hepatitis B virus by an aqueous extract of Agrimonia eupatoria L. Phytotherapy Res. 2005, 19, 355–358. [Google Scholar] [CrossRef] [PubMed]
  46. Huang, K.-L.; Lai, Y.-K.; Lin, C.-C.; Chang, J.-M. Inhibition of hepatitis B virus production byBoehmeria nivearoot extract in HepG2 2.2.15 cells. World J. Gastroenterol. 2006, 12, 5721–5725. [Google Scholar] [CrossRef]
  47. Webster, D.; Taschereau, P.; Lee, T.D.; Jurgens, T. Immunostimulant properties of Heracleum maximum Bartr. J. Ethnopharmacol. 2006, 106, 360–363. [Google Scholar] [CrossRef]
  48. Shanti, B.M. Perspective of Potential Plants for Medicine from Rajasthan, India. Int. J. Pharm. Res. 2016, 7, 1–6. [Google Scholar]
  49. Otake, T.; Mori, H.; Morimoto, M.; Ueba, N.; Sutardjo, S.; Kusumoto, I.T.; Hattori, M.; Namba, T. Screening of Indonesian plant extracts for anti-human immunodeficiency virus—Type 1 (HIV-1) activity. Phytotherapy Res. 1995, 9, 6–10. [Google Scholar] [CrossRef]
  50. Rege, A.; Chowdhary, A.S. Evaluation of Ocimum sanctum and Tinospora cordifolia as probable HIV protease inhibitors. Int. J. of Pharm. Sci. Rev. Res. 2014, 25, 315–318. [Google Scholar]
  51. Thayil Seema, M.; Thyagarajan, S. Methanol and aqueous extracts of Ocimum kilimandscharicum (Karpuratulasi) inhibits HIV-1 reverse transcriptase in vitro. Int. J. Pharmacogn. Phytochem. Res. 2016, 8, 1099–1103. [Google Scholar]
  52. Yu, Y.-B. The extracts of Solanum nigrum L. for inhibitory effects on HIV-1 and its essential enzymes. Korean J. Orient Med. Prescr. 2004, 10, 119–126. [Google Scholar]
  53. Nair, R. HIV-1 reverse transcriptase inhibition by Vitex negundo L. leaf extract and quantification of flavonoids in relation to anti-HIV activity. J. Cell. Mol. Biol. 2012, 10, 53–59. [Google Scholar]
  54. Castro, F.; Cardoso, A.; Gonçalves, R.M.; Serre, K.; Oliveira, M.J. Interferon-Gamma at the Crossroads of Tumor Immune Surveillance or Evasion. Front. Immunol. 2018, 9, 847. [Google Scholar] [CrossRef] [Green Version]
  55. 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. Biflavonoids from Torreya nucifera displaying SARS-CoV 3CLpro inhibition. Bioorg. Med. Chem. 2010, 18, 7940–7947. [Google Scholar] [CrossRef]
  56. 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 fromAngelica keiskeiinhibit cysteine proteases of SARS-CoV. J. Enzym. Inhib. Med. Chem. 2016, 31, 23–30. [Google Scholar] [CrossRef] [Green Version]
  57. Chen, Z.; Ye, S.-Y.; Yang, Y.; Li, Z.-Y. A review on charred traditional Chinese herbs: Carbonization to yield a haemostatic effect. Pharm. Biol. 2019, 57, 498–506. [Google Scholar] [CrossRef] [Green Version]
  58. Wen, C.-C.; Shyur, L.-F.; Jan, J.-T.; Liang, P.-H.; Kuo, C.-J.; Arulselvan, P.; Wu, J.-B.; Kuo, S.-C.; Yang, N.-S. Traditional Chinese medicine herbal extracts of Cibotium barometz, Gentiana scabra, Dioscorea batatas, Cassia tora, and Taxillus chinensis inhibit SARS-CoV replication. J. Tradit. Complement. Med. 2011, 1, 41–50. [Google Scholar] [CrossRef] [Green Version]
  59. 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. [Google Scholar] [CrossRef] [Green Version]
  60. Zhen, H.; Fang, F.; Ye, D.-Y.; Shu, S.-N.; Zhou, Y.-F.; Dong, Y.-S.; Nie, X.-C.; Li, G. Experimental study on the action of allitridin against human cytomegalovirus in vitro: Inhibitory effects on immediate-early genes. Antivir. Res. 2006, 72, 68–74. [Google Scholar] [CrossRef]
  61. Yu, M.-S.; Lee, J.; Lee, J.M.; Kim, Y.; Chin, Y.-W.; Jee, J.-G.; Keum, Y.-S.; Jeong, Y.-J. Identification of myricetin and scutellarein as novel chemical inhibitors of the SARS coronavirus helicase, nsP13. Bioorg. Med. Chem. Lett. 2012, 22, 4049–4054. [Google Scholar] [CrossRef]
  62. Zhu, Q.-C.; Wang, Y.; Liu, Y.-P.; Zhang, R.-Q.; Li, X.; Su, W.-H.; Long, F.; Luo, X.-D.; Peng, T. Inhibition of enterovirus 71 replication by chrysosplenetin and penduletin. Eur. J. Pharm. Sci. 2011, 44, 392–398. [Google Scholar] [CrossRef]
  63. Wen, C.-C.; Kuo, Y.-H.; Jan, J.-T.; Liang, P.-H.; Wang, S.-Y.; Liu, H.-G.; Lee, C.-K.; Chang, S.-T.; Kuo, C.-J.; Lee, S.-S.; et al. Specific Plant Terpenoids and Lignoids Possess Potent Antiviral Activities against Severe Acute Respiratory Syndrome Coronavirus. J. Med. Chem. 2007, 50, 4087–4095. [Google Scholar] [CrossRef] [Green Version]
  64. Gao, Y.; Dong, X.; Kang, T.G. Activity of in vitro anti-influenza virus of arctigenin. Chin. Tradit. Herb. Drugs 2002, 33, 724–725. [Google Scholar]
  65. Yi, L.; Li, Z.; Yuan, K.; Qu, X.; Chen, J.; Wang, G.; Zhang, H.; Luo, H.; Zhu, L.; Jiang, P.; et al. Small Molecules Blocking the Entry of Severe Acute Respiratory Syndrome Coronavirus into Host Cells. J. Virol. 2004, 78, 11334–11339. [Google Scholar] [CrossRef] [Green Version]
  66. Zuo, G.-Y.; Li, Z.-Q.; Chen, L.-R.; Xu, X.-J. In Vitro Anti-HCV Activities of Saxifraga Melanocentra and its Related Polyphenolic Compounds. Antivir. Chem. Chemother. 2005, 16, 393–398. [Google Scholar] [CrossRef] [Green Version]
  67. Zuo, G.; Li, Z.; Chen, L.; Xu, X. Activity of compounds from Chinese herbal medicine Rhodiola kirilowii (Regel) Maxim against HCV NS3 serine protease. Antivir. Res. 2007, 76, 86–92. [Google Scholar] [CrossRef]
  68. Guo, Q.; Zhao, L.; You, Q.; Yang, Y.; Gu, H.; Song, G.; Lu, N.; Xin, J. Anti-hepatitis B virus activity of wogonin in vitro and in vivo. Antivir. Res. 2007, 74, 16–24. [Google Scholar] [CrossRef]
  69. Zhou, Z.; Zhang, Y.; Ding, X.-R.; Chen, S.-H.; Yang, J.; Wang, X.-J.; Jia, G.-L.; Chen, H.-S.; Bo, X.-C.; Wang, S.-Q. Protocatechuic aldehyde inhibits hepatitis B virus replication both in vitro and in vivo. Antivir. Res. 2007, 74, 59–64. [Google Scholar] [CrossRef] [PubMed]
  70. Wang, Y.-P.; Zhao, W.; Xue, R.; Zhou, Z.-X.; Liu, F.; Han, Y.-X.; Ren, G.; Peng, Z.-G.; Cen, S.; Chen, H.-S.; et al. Oxymatrine inhibits hepatitis B infection with an advantage of overcoming drug-resistance. Antivir. Res. 2011, 89, 227–231. [Google Scholar] [CrossRef] [PubMed]
  71. Zhuang, M.; Jiang, H.; Suzuki, Y.; Li, X.; Xiao, P.; Tanaka, T.; Ling, H.; Yang, B.; Saitoh, H.; Zhang, L.; et al. Procyanidins and butanol extract of Cinnamomi Cortex inhibit SARS-CoV infection. Antivir. Res. 2009, 82, 73–81. [Google Scholar] [CrossRef] [PubMed]
  72. Park, J.-Y.; Jeong, H.J.; Kim, J.H.; Kim, Y.M.; Park, S.-J.; Kim, D.; 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. [Google Scholar] [CrossRef] [Green Version]
  73. Chin, L.W.; Cheng, Y.-W.; Lin, S.-S.; Lai, Y.-Y.; Lin, L.-Y.; Chou, M.-Y.; Yang, C.-C.; Chou, M.-C. Anti-herpes simplex virus effects of berberine from Coptidis rhizoma, a major component of a Chinese herbal medicine, Ching-Wei-San. Arch. Virol. 2010, 155, 1933–1941. [Google Scholar] [CrossRef]
  74. Lee, J.S.; Kim, H.J.; Lee, Y.S. A new Anti-HIV Flavonoid Glucuronide fromChrysanthemum morifolium. Planta Medica 2003, 69, 859–861. [Google Scholar] [CrossRef]
  75. Zhang, H.; Tan, G.T.; Hoang, V.D.; Van Hung, N.; Cuong, N.M.; Soejarto, D.D.; Pezzuto, J.M.; Fong, H.H.S. Natural Anti-HIV Agents. Part IV. Anti-HIV Constituents fromVaticacinerea1. J. Nat. Prod. 2003, 66, 263–268. [Google Scholar] [CrossRef]
  76. Yang, X.-W.; Zhao, J.; Cui, Y.-X.; Liu, X.-H.; Ma, C.-M.; Hattori, M.; Zhang, L.-H. Anti-HIV-1 Protease Triterpenoid Saponins from the Seeds of Aesculuschinensis. J. Nat. Prod. 1999, 62, 1510–1513. [Google Scholar] [CrossRef]
  77. Kuo, Y.H.; Li, S.Y.; Huang, R.L.; Wu, M.D.; Huang, H.C.; Lee, K.H. Schizarin B, C, D, and E, Four New Lignans from Kadsuram atsudai and Their Antihepatitis Activities. J. Nat. Prod. 2001, 64, 487–490. [Google Scholar] [CrossRef]
  78. Kuo, Y.-C.; Kuo, Y.-H.; Lin, Y.-L.; Tsai, W.-J. Yatein from Chamaecyparis obtusa suppresses herpes simplex virus type 1 replication in HeLa cells by interruption the immediate-early gene expression. Antivir. Res. 2006, 70, 112–120. [Google Scholar] [CrossRef]
  79. Cheng, H.-Y.; Lin, T.-C.; Yang, C.-M.; Wang, K.-C.; Lin, L.-T.; Lin, C.-C. Putranjivain A from Euphorbia jolkini inhibits both virus entry and late stage replication of herpes simplex virus type 2 in vitro. J. Antimicrob. Chemother. 2004, 53, 577–583. [Google Scholar] [CrossRef] [Green Version]
  80. Kuo, Y.-C.; Lin, L.-C.; Tsai, W.-J.; Chou, C.-J.; Kung, S.-H.; Ho, Y.-H. Samarangenin B from Limonium sinense Suppresses Herpes Simplex Virus Type 1 Replication in Vero Cells by Regulation of Viral Macromolecular Synthesis. Antimicrob. Agents Chemother. 2002, 46, 2854–2864. [Google Scholar] [CrossRef] [Green Version]
  81. Lin, L.-C.; Kuo, Y.-C.; Chou, C.-J. Anti-Herpes Simplex Virus Type-1 Flavonoids and a New Flavanone from the Root of Limonium sinense. Planta Medica 2000, 66, 333–336. [Google Scholar] [CrossRef]
  82. Parida, M.; Upadhyay, C.; Pandya, G.; Jana, A. Inhibitory potential of neem (Azadirachta indica Juss) leaves on Dengue virus type-2 replication. J. Ethnopharmacol. 2002, 79, 273–278. [Google Scholar] [CrossRef]
  83. Eze, D.C.; Okwor, E.C.; Ehirim, C.H.; Ibu, J.O.; Shoyinka, S.V.O. Comparative evaluation of Moringa oleifera and vacci-boost immuno-modulators in chickens experimentally infected with Newcastle Disease Virus (Kudu 113 Strain). South Asian J. Exp. Biol. 2014, 4, 42–47. [Google Scholar]
  84. Murakami, A.; Kitazono, Y.; Jiwajinda, S.; Koshimizu, K.; Ohigashi, H. Niaziminin, a thiocarbamate from the leaves of Moringa oleifera, holds a strict structural requirement for inhibition of tumor-promotor-induced Epstein-Barr virus activation. Planta Med. 1998, 64, 319–323. [Google Scholar] [CrossRef] [PubMed]
  85. Jayasundar, R.; Ghatak, S.; Makhdoomi, M.A.; Luthra, K.; Singh, A.; Velpandian, T. Challenges in integrating component level technology and system level information from Ayurveda: Insights from NMR phytometabolomics and anti-HIV potential of select Ayurvedic medicinal plants. J. Ayurveda Integr. Med. 2019, 10, 94–101. [Google Scholar] [CrossRef] [PubMed]
  86. Vickers, N.J. Animal Communication: When I’m Calling You, Will You Answer Too? Curr. Biol. 2017, 27, R713–R715. [Google Scholar] [CrossRef] [PubMed]
  87. Beula, J.M.; Gnanadesigan, M.; Rajkumar, P.B.; Ravikumar, S.; Anand, M. Antiviral, antioxidant and toxicological evaluation of mangrove plant from South East coast of India. Asian Pac. J. Trop. Biomed. 2012, 2, S352–S357. [Google Scholar] [CrossRef]
  88. Lau, K.M.; Lee, K.M.; Koon, C.M.; Cheung, C.S.; Lau, C.P.; Ho, H.M.; Lee, M.Y.; Au, S.W.; Cheng, C.H.; Lau, C.B.; et al. Immunomodulatory and anti-SARS activities of Houttuynia cordata. J. Ethnopharmacol. 2008, 118, 79–85. [Google Scholar] [CrossRef] [PubMed]
  89. Tolo, F.M.; Rukunga, G.M.; Muli, F.W.; Njagi, E.N.; Njue, W.; Kumon, K.; Mungai, G.M.; Muthaura, C.N.; Muli, J.M.; Keter, L.K.; et al. Anti-viral activity of the extracts of a Kenyan medicinal plant Carissa edulis against herpes simplex virus. J. Ethnopharmacol. 2006, 104, 92–99. [Google Scholar] [CrossRef]
  90. Demurtas, O.C.; Massa, S.; Illiano, E.; De Martinis, D.; Chan, P.K.; Di Bonito, P.; Franconi, R. Antigen production in plant to tackle infectious diseases flare up: The case of SARS. Front. Plant. Sci. 2016, 7, 54. [Google Scholar] [CrossRef] [Green Version]
  91. Van Der Meer, F.J.U.M.; De Haan, C.A.M.; Schuurman, N.M.P.; Haijema, B.J.; Verheije, M.H.; Bosch, B.J.; Balzarini, J.; Egberink, H.F. The carbohydrate-binding plant lectins and the non-peptidic antibiotic pradimicin A target the glycans of the coronavirus envelope glycoproteins. J. Antimicrob. Chemother. 2007, 60, 741–749. [Google Scholar] [CrossRef] [Green Version]
  92. Notka, F.; Meier, G.; Wagner, R. Concerted inhibitory activities of Phyllanthus amarus on HIV replication in vitro and ex vivo. Antivir. Res. 2004, 64, 93–102. [Google Scholar] [CrossRef]
  93. Das Mahapatra, A.; Bhowmik, P.; Banerjee, A.; Das, A.; Ojha, D.; Chattopadhyay, D. Ethnomedicinal Wisdom. In New Look to Phytomedicine; Elsevier: Amsterdam, The Netherlands, 2019; pp. 35–61. [Google Scholar]
  94. Mukherjee, H.; Ojha, D.; Bag, P.; Chandel, H.S.; Bhattacharyya, S.; Chatterjee, T.K.; Mukherjee, P.K.; Chakraborti, S.; Chattopadhyay, D. Anti-herpes virus activities of Achyranthes aspera: An Indian ethnomedicine, and its triterpene acid. Microbiol. Res. 2013, 168, 238–244. [Google Scholar] [CrossRef]
  95. Chen, C.-N.; Lin, C.P.C.; Huang, K.-K.; Chen, W.-C.; Hsieh, H.-P.; Liang, P.-H.; Hsu, J.T.-A. Inhibition of SARS-CoV 3C-like Protease Activity by Theaflavin-3,3′-digallate (TF3). Evid. Based Complement. Altern. Med. 2005, 2, 209–215. [Google Scholar] [CrossRef] [Green Version]
  96. Arthanari, S.K.; Vanitha, J.; Ganesh, M.; Venkateshwaran, K.; Clercq, D. Evaluation of antiviral and cytotoxic activities of methanolic extract of S. grandiflora (Fabaceae) flowers. Asian Pac. J. Trop. Biomed. 2012, 2, S855–S858. [Google Scholar] [CrossRef]
  97. Cagno, V.; Civra, A.; Kumar, R.; Pradhan, S.; Donalisio, M.; Sinha, B.N.; Ghosh, M.; Lembo, D. Ficus religiosa L. bark extracts inhibit human rhinovirus and respiratory syncytial virus infection in vitro. J. Ethnopharmacol. 2015, 176, 252–257. [Google Scholar] [CrossRef]
  98. Jain, M.; Ganju, L.; Katiyal, A.; Padwad, Y.; Mishra, K.; Chanda, S.; Karan, D.; Yogendra, K.; Sawhney, R. Effect of Hippophae rhamnoides leaf extract against Dengue virus infection in human blood-derived macrophages. Phytomedicine 2008, 15, 793–799. [Google Scholar] [CrossRef]
  99. Yamai, M.; Tsumura, K.; Kimura, M.; Fukuda, S.; Murakami, T.; Kimura, Y. Antiviral Activity of a Hot Water Extract of Black Soybean against a Human Respiratory Illness Virus. Biosci. Biotechnol. Biochem. 2003, 67, 1071–1079. [Google Scholar] [CrossRef] [Green Version]
  100. Mishra, S.; Aeri, V.; Gaur, P.K.; Jachak, S.M. Phytochemical, Therapeutic, and Ethnopharmacological Overview for a Traditionally Important Herb: Boerhavia diffusa Linn. BioMed. Res. Int. 2014, 2014, 808302. [Google Scholar] [CrossRef] [Green Version]
  101. Keyaerts, E.; Vijgen, L.; Pannecouque, C.; Van Damme, E.; Peumans, W.; Egberink, H.; Balzarini, J.; Van Ranst, M. Plant lectins are potent inhibitors of coronaviruses by interfering with two targets in the viral replication cycle. Antivir. Res. 2007, 75, 179–187. [Google Scholar] [CrossRef]
  102. Prathapan, A.; Vineetha, V.P.; Abhilash, P.A.; Raghu, K.G. Boerhaaviadiffusa, L. attenuates angiotensin II-induced hypertrophy in H9c2 cardiac myoblast cells via modulating oxidative stress and down-regulating NF-κβ and transforming growth factor β1. Br. J. Nutr. 2013, 110, 1201–1210. [Google Scholar] [CrossRef] [Green Version]
  103. Khan, M.Y.; Kumar, V. Mechanism & inhibition kinetics of bioassay-guided fractions of Indian medicinal plants and foods as ACE inhibitors. J. Tradit. Complement. Med. 2019, 9, 73–84. [Google Scholar] [CrossRef]
  104. Olivieri, F.; Prasad, V.; Valbonesi, P.; Srivastava, S.; Ghosal-Chowdhury, P.; Barbieri, L.; Bolognesi, A.; Stirpe, F. A systemic antiviral resistance-inducing protein isolated fromClerodendrum inermeGaertn. is a polynucleotide:adenosine glycosidase (ribosome-inactivating protein). FEBS Lett. 1996, 396, 132–134. [Google Scholar] [CrossRef] [Green Version]
  105. Mukherjee, P.K.; Maity, N.; Nema, N.K.; Sarkar, B.K. Standardized Clitoria ternatea leaf extract as hyaluronidase, elastase and matrix-metalloproteinase-1 inhibitor. Indian J. Pharmacol. 2012, 44, 584–587. [Google Scholar] [CrossRef] [Green Version]
  106. Bigoniya, P.; Patel, K.K.; Pandey, A.; Raj, V. Pharmacological screening of Coriandrum sativum Linn. for hepatoprotective activity. J. Pharm. Bioallied Sci. 2011, 3, 435–441. [Google Scholar] [CrossRef]
  107. Vimalanathan, S.; Ignacimuthu, S.; Hudson, J.B. Medicinal plants of Tamil Nadu (Southern India) are a rich source of antiviral activities. Pharm. Biol. 2009, 47, 422–429. [Google Scholar] [CrossRef]
  108. Gilani, A.H.; Khan, A.-U.; Raoof, M.; Ghayur, M.N.; Siddiqui, B.; Vohra, W.; Begum, S. Gastrointestinal, selective airways and urinary bladder relaxant effects of Hyoscyamus niger are mediated through dual blockade of muscarinic receptors and Ca2+channels. Fundam. Clin. Pharmacol. 2008, 22, 87–99. [Google Scholar] [CrossRef]
  109. He, L.; Qi, Y.; Rong, X.; Jiang, J.; Yang, Q.; Yamahara, J.; Murray, M.; Li, Y. The Ayurvedic MedicineSalacia oblongaAttenuates Diabetic Renal Fibrosis in Rats: Suppression of Angiotensin II/AT1 Signaling. Evid. Based Complement. Altern. Med. 2011, 2011, 807451. [Google Scholar] [CrossRef] [Green Version]
  110. Ganjhu, R.K.; Mudgal, P.P.; Maity, H.; Dowarha, D.; Devadiga, S.; Nag, S.; Arunkumar, G. Herbal plants and plant preparations as remedial approach for viral diseases. Virus Dis. 2015, 26, 225–236. [Google Scholar] [CrossRef] [PubMed]
  111. Galani, V.J.; Patel, B.G.; Rana, D.G. Sphaeranthus indicus Linn.: A phytopharmacological review. Int. J. Ayurveda Res. 2010, 1, 247–253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Tiwari, B.K.; Khosa, R.L. Hepatoprotective and antioxidant effect of Sphaeranthus indicus against acetaminophen-induced hepatotoxicity in rats. J. Pharm Sci Res. 2009, 1, 26–30. [Google Scholar]
  113. Tsai, Y.-C.; Lee, C.-L.; Yen, H.-R.; Chang, Y.-S.; Lin, Y.-P.; Huang, S.-H.; Lin, C.-W. Antiviral Action of Tryptanthrin Isolated from Strobilanthes cusia Leaf against Human Coronavirus NL63. Biomolecules 2020, 10, 366. [Google Scholar] [CrossRef] [Green Version]
  114. Kumar, K.P.S.; Bhowmik, D.; Dutta, A.; Yadav, A.P.; Paswan, S.; Srivastava, S.; Deb, L. Recent trends in potential traditional Indian herbs Emblica officinalis and its medicinal importance. J. Pharmacogn. Phytochem. 2012, 1, 18–28. [Google Scholar]
  115. Edai, Z.-C.; Fu, W.; Ewan, L.-Y.; Ecai, H.-H.; Ewang, N.; Qi, S.-S.; Du, D.-L. Different Growth Promoting Effects of Endophytic Bacteria on Invasive and Native Clonal Plants. Front. Plant Sci. 2016, 7, 706. [Google Scholar] [CrossRef] [Green Version]
  116. Puri, A.; Saxena, R.P.; Saxena, K.; Srivastava, V.; Tandon, J. Immunostimulant activity of Nyctanthes arbor-tristis L. J. Ethnopharmacol. 1994, 42, 31–37. [Google Scholar] [CrossRef]
  117. Ejantan, I.; Eahmad, W.; Ebukhari, S.N.A. Plant-derived immunomodulators: An insight on their preclinical evaluation and clinical trials. Front. Plant Sci. 2015, 6, 655. [Google Scholar] [CrossRef]
  118. Mohammadi, S.; Jafari, B.; Asgharian, P.; Martorell, M.; Sharifi-Rad, J. Medicinal plants used in the treatment of Malaria: A key emphasis to Artemisia, Cinchona, Cryptolepis and Tabebuia genera. Phytother. Res. 2020, 34, 1556–1569. [Google Scholar] [CrossRef]
  119. Brattig, N.; Diao, G.-J.; Berg, P. Immunoenhancing effect of flavonoid compounds on lymphocyte proliferation and immunoglobulin synthesis. Int. J. Immunopharmacol. 1984, 6, 205–215. [Google Scholar] [CrossRef]
  120. Yadav, V.S.; Mishra, K.P.; Singh, D.P.; Mehrotra, S.; Singh, V.K. Immunomodulatory Effects of Curcumin. Immunopharmacol. Immunotoxicol. 2005, 27, 485–497. [Google Scholar] [CrossRef]
  121. Fürst, R.; Zündorf, I. Plant-Derived Anti-Inflammatory Compounds: Hopes and Disappointments regarding the Translation of Preclinical Knowledge into Clinical Progress. Mediat. Inflamm. 2014, 2014, 146832. [Google Scholar] [CrossRef] [Green Version]
  122. Ložienė, K.; Švedienė, J.; Paškevičius, A.; Raudonienė, V.; Sytar, O.; Kosyan, A. Influence of plant origin natural α-pinene with different enantiomeric composition on bacteria, yeasts and fungi. Fitoterapia 2018, 127, 20–24. [Google Scholar] [CrossRef]
  123. Lecher, J.C.; Diep, N.; Krug, P.W.; Hilliard, J.K. Genistein Has Antiviral Activity against Herpes B Virus and Acts Synergistically with Antiviral Treatments to Reduce Effective Dose. Viruses 2019, 11, 499. [Google Scholar] [CrossRef] [Green Version]
  124. Malaguarnera, L. Influence of Resveratrol on the Immune Response. Nutrients 2019, 11, 946. [Google Scholar] [CrossRef] [Green Version]
  125. Gupta, S.; Mishra, K.P.; Ganju, L. Broad-spectrum antiviral properties of andrographolide. Arch. Virol. 2017, 162, 611–623. [Google Scholar] [CrossRef]
  126. Perricone, C.; Triggianese, P.; Bartoloni, E.; Cafaro, G.; Bonifacio, A.F.; Bursi, R.; Perricone, R.; Gerli, R. The anti-viral facet of anti-rheumatic drugs: Lessons from COVID-19. J. Autoimmun. 2020, 111, 102468. [Google Scholar] [CrossRef]
  127. Parvez, M.K.; Rehman, T.; Alam, P.; Al-Dosari, M.S.; Alqasoumi, S.I.; Alajmi, M.F. Plant-derived antiviral drugs as novel hepatitis B virus inhibitors: Cell culture and molecular docking study. Saudi Pharm. J. 2019, 27, 389–400. [Google Scholar] [CrossRef] [PubMed]
  128. Khan, F.; Sarker, M.R.; Ming, L.C.; Mohamed, I.N.; Zhao, C.; Sheikh, B.Y.; Tsong, H.F.; Rashid, M.A. Comprehensive Review on Phytochemicals, Pharmacological and Clinical Potentials of Gymnema sylvestre. Front. Pharmacol. 2019, 10, 1223. [Google Scholar] [CrossRef] [PubMed]
  129. Liu, T.; Liu, X.; Li, W. Tetrandrine, a Chinese plant-derived alkaloid, is a potential candidate for cancer chemotherapy. Oncotarget 2016, 7, 40800–40815. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Kim, E.; Kwak, J. Antiviral phenolic compounds from the whole plants of Zostera marina against influenza A virus. Planta Medica 2015, 81, PW_06. [Google Scholar] [CrossRef]
  131. Naser, B.; Bodinet, C.; Tegtmeier, M.; Lindequist, U. Thuja occidentalis(Arbor vitae): A Review of its Pharmaceutical, Pharmacological and Clinical Properties. Evid. Based Complement. Altern. Med. 2005, 2, 69–78. [Google Scholar] [CrossRef] [Green Version]
  132. Dabeek, W.M.; Marra, M.V. Dietary Quercetin and Kaempferol: Bioavailability and Potential Cardiovascular-Related Bioactivity in Humans. Nutrients 2019, 11, 2288. [Google Scholar] [CrossRef] [Green Version]
  133. Andarwulan, N.; Batari, R.; Sandrasari, D.A.; Bolling, B.; Wijaya, H. Flavonoid content and antioxidant activity of vegetables from Indonesia. Food Chem. 2010, 121, 1231–1235. [Google Scholar] [CrossRef] [Green Version]
  134. Lama-Muñoz, A.; Contreras, M.D.M.; Espínola, F.; Moya, M.; Romero, I.; Castro, E. Optimization of Oleuropein and Luteolin-7-O-Glucoside Extraction from Olive Leaves by Ultrasound-Assisted Technology. Energies 2019, 12, 2486. [Google Scholar] [CrossRef] [Green Version]
  135. Cahyono, B.; Ariani, J.; Failasufa, H.; Suzery, M.; Susanti, S.; Hadiyanto, H. Extraction of homologous compounds of curcuminoid isolated from temulawak (Curcuma xanthorriza Roxb.) plant. Rasayan J. Chem. 2019, 12, 7–13. [Google Scholar] [CrossRef]
  136. Rosidi, A.; Khomsan, A.; Setiawan, B.I.; Riyadi, H.; Briawan, D. Antioxidant Potential of Temulawak (Curcuma xanthorrhiza roxb). Pak. J. Nutr. 2016, 15, 556–560. [Google Scholar] [CrossRef] [Green Version]
  137. Salehi, B.; Fokou, P.V.T.; Sharifi-Rad, M.; Zucca, P.; Pezzani, R.; Martins, N.; Sharifi-Rad, J. The Therapeutic Potential of Naringenin: A Review of Clinical Trials. Pharmaceuticals 2019, 12, 11. [Google Scholar] [CrossRef] [Green Version]
  138. Meirinhos, J.; Silva, B.M.; Valentão, P.; Seabra, R.M.; Pereira, J.A.; Dias, A.; Andrade, P.B.; Ferreres, F. Analysis and quantification of flavonoidic compounds from Portuguese olive (Olea Europaea L.) leaf cultivars. Nat. Prod. Res. 2005, 19, 189–195. [Google Scholar] [CrossRef]
  139. Nicolì, F.; Negro, C.; Vergine, M.; Aprile, A.; Nutricati, E.; Sabella, E.; Miceli, A.; Luvisi, A.; De Bellis, L. Evaluation of Phytochemical and Antioxidant Properties of 15 Italian Olea europaea L. Cultivar Leaves. Molecules 2019, 24, 1998. [Google Scholar] [CrossRef] [Green Version]
  140. Miean, K.H.; Mohamed, S. Flavonoid (Myricetin, Quercetin, Kaempferol, Luteolin, and Apigenin) Content of Edible Tropical Plants. J. Agric. Food Chem. 2001, 49, 3106–3112. [Google Scholar] [CrossRef]
  141. De Maat, M.; Pijl, H.; Kluft, C.; Princen, H. Consumption of black and green tea has no effect on inflammation, haemostasis and endothelial markers in smoking healthy individuals. Eur. J. Clin. Nutr. 2000, 54, 757–763. [Google Scholar] [CrossRef] [Green Version]
  142. De Oliveira, R.M.M. Quantification of catechins and caffeine from green tea (Camellia sinensis) infusions, extract, and ready-to-drink beverages. Food Sci. Technol. 2012, 32, 163–166. [Google Scholar] [CrossRef] [Green Version]
  143. Sanwal, S.K.; Yadav, R.K.; Singh, P.K.; Buragohain, J.; Verma, M.R. Gingerol content of different genotypes of ginger (Zingiber officinale). Indian J. Agric. Sci. 2010, 80, 258. [Google Scholar]
  144. Li, L.-L.; Cui, Y.; Guo, X.-H.; Ma, K.; Tian, P.; Feng, J.; Wang, J.-M. Pharmacokinetics and Tissue Distribution of Gingerols and Shogaols from Ginger (Zingiber officinale Rosc.) in Rats by UPLC–Q-Exactive–HRMS. Molecules 2019, 24, 512. [Google Scholar] [CrossRef] [Green Version]
  145. Lelešius, R.; Karpovaitė, A.; Mickienė, R.; Drevinskas, T.; Tiso, N.; Ragažinskienė, O.; Kubilienė, L.; Maruška, A.; Šalomskas, A. In vitro antiviral activity of fifteen plant extracts against avian infectious bronchitis virus. BMC Vet. Res. 2019, 15, 178. [Google Scholar] [CrossRef] [Green Version]
  146. Dolzhenko, Y.; Bertea, C.M.; Occhipinti, A.; Bossi, S.; Maffei, M. UV-B modulates the interplay between terpenoids and flavonoids in peppermint (Mentha piperita L.). J. Photochem. Photobiol. B Biol. 2010, 100, 67–75. [Google Scholar] [CrossRef]
  147. Fatih, B.; Madani, K.; Chibane, M.; Duez, P.; Brahmi, F.; Khodir, M.; Mohamed, C.; Pierre, D. Chemical Composition and Biological Activities of Mentha Species. In Aromatic and Medicinal Plants—Back to Nature; IntechOpen: London, UK, 2017. [Google Scholar] [CrossRef] [Green Version]
  148. Porte, A.; Godoy, R. Chemical composition of Thymus vulgaris L. (Thyme) essential oil from the Rio de Janeiro state, Brazil. J. Serb. Chem. Soc. 2008, 73, 307–310. [Google Scholar] [CrossRef]
  149. Gedikoğlu, A.; Sökmen, M.; Çivit, A. Evaluation of Thymus vulgaris and Thymbra spicata essential oils and plant extracts for chemical composition, antioxidant, and antimicrobial properties. Food Sci. Nutr. 2019, 7, 1704–1714. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Puodziuniene, G.; Kairyte, V.; Janulis, V.; Razukas, A.; Barsteigiene, Z.; Ragažinskienė, O. Quantitative hplc estimation of flavonoids in showy tick trefoil (Desmodium canadense) herbs. Pharm. Chem. J. 2011, 45, 88–90. [Google Scholar] [CrossRef]
  151. Batyuk, V.S.V.; Vasil’eva, L.N.; Chernobrovaya, N.V.; Komissarenko, N.F. Flavonoids of Desmodium canadense and their analgesic effect. Khim. Farm. Zh. 1987, 21, 63–67. [Google Scholar]
  152. Taylor, W.G.; Sutherland, D.H.; Richards, K.W. Soyasaponins and Related Glycosides of Desmodium canadense and Desmodium illinoense. Open Nat. Prod. J. 2009, 2, 59–67. [Google Scholar] [CrossRef]
  153. Ma, X.; Zheng, C.; Hu, C.; Rahman, K.; Qin, L. The genus Desmodium (Fabaceae)-traditional uses in Chinese medicine, phytochemistry and pharmacology. J. Ethnopharmacol. 2011, 138, 314–332. [Google Scholar] [CrossRef]
  154. Yang, J.-L.; Ha, T.-K.-Q.; Dhodary, B.; Pyo, E.; Nguyen-Ngoc, H.; Cho, H.; Kim, E.; Oh, W.K. Oleanane Triterpenes from the Flowers ofCamellia japonicaInhibit Porcine Epidemic Diarrhea Virus (PEDV) Replication. J. Med. Chem. 2015, 58, 1268–1280. [Google Scholar] [CrossRef]
  155. Yang, J.-L.; Ha, T.K.Q.; Oh, W.K. Discovery of inhibitory materials against PEDV corona virus from medicinal plants. Jpn. J. Vet. Res. 2016, 64, S53–S63. [Google Scholar]
  156. Azuma, C.M.; Dos Santos, F.C.S.; Lago, J.H.G. Flavonoids and fatty acids of Camellia japonica leaves extract. Rev. Bras. Farm. 2011, 21, 1159–1162. [Google Scholar] [CrossRef] [Green Version]
  157. Itokawa, H.; Nakajima, H.; Ikuta, A.; Iitaka, Y. Two triterpenes from the flowers of Camellia japonica. Phytochemistry 1981, 20, 2539–2542. [Google Scholar] [CrossRef]
  158. Kato, M.; Ashihara, H. Biosynthesis and Catabolism of Purine Alkaloids in Camellia Plants. Nat. Prod. Commun. 2008, 3, 1934578 0800300907. [Google Scholar] [CrossRef] [Green Version]
  159. Karioti, A.; Bilia, A.R.; Skaltsa, H. Quercus ilex L.: A rich source of polyacylated flavonoid glucosides. Food Chem. 2010, 123, 131–142. [Google Scholar] [CrossRef]
  160. Cheng, P.-W.; Huang, L.-T.; Chiang, L.-C.; Lin, C.-C. Antiviral effects of saikosaponins on human coronavirus 229e in vitro. Clin. Exp. Pharmacol. Physiol. 2006, 33, 612–616. [Google Scholar] [CrossRef]
  161. 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, 7597596. [Google Scholar] [CrossRef] [Green Version]
  162. Hemalatha, S.; Kumar, M.; Prasad, S. A current update on the phytopharmacological aspects of Houttuynia cordata Thunb. Pharmacogn. Rev. 2014, 8, 22–35. [Google Scholar] [CrossRef] [Green Version]
  163. 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. [Google Scholar] [CrossRef]
  164. Speranza, J.; Miceli, N.; Taviano, M.F.; Ragusa, S.; Kwiecień, I.; Szopa, A.; Ekiert, H. Isatis tinctoria L. (Woad): A Review of Its Botany, Ethnobotanical Uses, Phytochemistry, Biological Activities, and Biotechnological Studies. Plants 2020, 9, 298. [Google Scholar] [CrossRef] [Green Version]
  165. Wang, L.; Zhang, X.-Q.; Yin, Z.-Q.; Wang, Y.; Ye, W.-C. Two New Amaryllidaceae Alkaloids from the Bulbs of Lycoris radiata. Chem. Pharm. Bull. 2009, 57, 610–611. [Google Scholar] [CrossRef] [Green Version]
  166. Tian, Y.; Zhang, C.; Guo, M. Comparative Analysis of Amaryllidaceae Alkaloids from Three Lycoris Species. Molecules 2015, 20, 21854–21869. [Google Scholar] [CrossRef] [Green Version]
  167. Shi, T.; Yue, Y.; Shi, M.; Chen, M.; Yang, X.; Wang, L. Exploration of Floral Volatile Organic Compounds in Six Typical Lycoris taxa by GC-MS. Plants 2019, 8, 422. [Google Scholar] [CrossRef] [Green Version]
  168. Gong, S.J.; Su, X.J.; Yu, H.P.; Li, J.; Qin, Y.J.; Xu, Q.; Luo, W.-S. A study on anti-SARS-CoV 3CL protein of flavonoids from litchi chinensis sonn core. Chin. Pharmacol. 2008, 24, 699–700. [Google Scholar]
  169. Jo, S.; Kim, S.; Shin, D.H.; Kim, M.-S. Inhibition of SARS-CoV 3CL protease by flavonoids. J. Enzym. Inhib. Med. Chem. 2020, 35, 145–151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Nimmanpipug, P.; Lee, V.S.; Wolschann, P.; Hannongbua, S. Litchi chinensis-derived terpenoid as anti-HIV-1 protease agent: Structural design from molecular dynamics simulations. Mol. Simul. 2009, 35, 673–680. [Google Scholar] [CrossRef]
  171. Ibrahim, S.R.; Mohamed, G.A. Litchi chinensis: Medicinal uses, phytochemistry, and pharmacology. J. Ethnopharmacol. 2015, 174, 492–513. [Google Scholar] [CrossRef] [PubMed]
  172. Kim, D.E.; Min, J.S.; Jang, M.S.; Lee, J.Y.; Shin, Y.S.; Park, C.M.; Song, J.H.; Kim, H.R.; Kim, S.; Jin, Y.-H.; et al. Natural Bis-Benzylisoquinoline Alkaloids-Tetrandrine, Fangchinoline, and Cepharanthine, Inhibit Human Coronavirus OC43 Infection of MRC-5 Human Lung Cells. Biomolecules 2019, 9, 696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Zhao, T.; Tang, H.; Xie, L.; Zheng, Y.; Ma, Z.; Sun, Q.; Li, X. Scutellaria baicalensis Georgi. (Lamiaceae): A review of its traditional uses, botany, phytochemistry, pharmacology and toxicology. J. Pharm. Pharmacol. 2019, 71, 1353–1369. [Google Scholar] [CrossRef] [Green Version]
  174. Shojai, T.M.; Langeroudi, A.G.; Karimi, V.; Barin, A.; Sadri, N. The effect of Allium sativum (Garlic) extract on infectious bronchitis virus in specific pathogen free embryonic egg. Avicenna J. Phytomed. 2016, 6, 458. [Google Scholar]
  175. Weber, N.D.; Andersen, D.O.; North, J.A.; Murray, B.K.; Lawson, L.D.; Hughes, B.G. In VitroVirucidal Effects of Allium sativum (Garlic) Extract and Compounds. Planta Med. 1992, 58, 417–423. [Google Scholar] [CrossRef]
  176. Szychowski, K.A.; Binduga, U.E.; Rybczyńska-Tkaczyk, K.; Leja, M.L.; Gmiński, J. Cytotoxic effects of two extracts from garlic (Allium sativum L.) cultivars on the human squamous carcinoma cell line SCC-15. Saudi J. Biol. Sci. 2018, 25, 1703–1712. [Google Scholar] [CrossRef] [Green Version]
  177. Pontin, M.; Bottini, R.; Burba, J.L.; Piccoli, P. Allium sativum produces terpenes with fungistatic properties in response to infection with Sclerotium cepivorum. Phytochemistry 2015, 115, 152–160. [Google Scholar] [CrossRef]
  178. Shin, S.-W. In vitro Effects of Essential Oils from the Aerial Parts of Artemisia annua L. Against Antibiotic-Susceptible and-Resistant Strains of Salmenella typhimurium. Yakhak Hoeji 2007, 51, 355–360. [Google Scholar]
  179. Nigam, M.; Atanassova, M.; Mishra, A.P.; Pezzani, R.; Devkota, H.P.; Plygun, S.; Salehi, B.; Setzer, W.N.; Sharifi-Rad, J. Bioactive Compounds and Health Benefits of Artemisia Species. Nat. Prod. Commun. 2019, 14, 19850354. [Google Scholar] [CrossRef] [Green Version]
  180. Li, W.; Moore, M.J.; Vasilieva, N.; Sui, J.; Wong, S.K.; Berne, M.A.; Somasundaran, M.; Sullivan, J.L.; Luzuriaga, K.; Greenough, T.C.; et al. Angiotensin-converting enzyme 2 is a functional receptor for the SARS coronavirus. Nature 2003, 426, 450–454. [Google Scholar] [CrossRef] [Green Version]
  181. Bais, S.; Gill, N.S.; Rana, N.; Shandil, S. A Phytopharmacological Review on a Medicinal Plant: Juniperus communis. Int. Sch. Res. Not. 2014, 2014, 634723. [Google Scholar] [CrossRef] [Green Version]
  182. Park, J.Y.; Kim, J.H.; Kwon, J.M.; Kwon, H.J.; Jeong, H.J.; Kim, Y.M. Dieckol, a SARS-CoV 3CL inhibitor, isolated from the edible brown algae Ecklonia cava. Phytochemistry 2013, 20, 2539–2542. [Google Scholar]
  183. Van Damme, E.J.M.; Peumans, W.J.; Barre, A.; Rougé, P. Plant Lectins: A Composite of Several Distinct Families of Structurally and Evolutionary Related Proteins with Diverse Biological Roles. Crit. Rev. Plant Sci. 1998, 17, 575–692. [Google Scholar] [CrossRef]
  184. Müller, W.E.; Renneisen, K.; Kreuter, M.H.; Schröder, H.C.; Winkler, I. The D-mannose-specific lectin from Gerardia savaglia blocks binding of human immunodeficiency virus type I to H9 cells and human lymphocytes in vitro. J. Acquir. Immune Defic. Syndr. 1988, 1, 453–458. [Google Scholar]
  185. Hammar, L.; Eriksson, S.; Morein, B. Human Immunodeficiency Virus Glycoproteins: Lectin Binding Properties. AIDS Res. Hum. Retrovir. 1989, 5, 495–506. [Google Scholar] [CrossRef]
  186. Hansen, J.-E.S.; Nielsen, C.; Heegaard, P.; Mathiesen, L.R.; Nielsen, J.O.; Nielsen, C. Correlation between carbohydrate structures on the envelope glycoprotein gp120 of.HIV-1 and HIV-2 and syncytium inhibition with lectins. AIDS 1989, 3, 635–642. [Google Scholar] [CrossRef]
  187. Matsui, T.; Kobayashi, S.; Yoshida, O.; Ishii, S.-I.; Abe, Y.; Yamamoto, N. Effects of succinylated concanavalin A on infectivity and syncytial formation of human immunodeficiency virus. Med. Microbiol. Immunol. 1990, 179, 225–235. [Google Scholar] [CrossRef]
  188. Balzarini, J.; Schols, D.; Neyts, J.; Van Damme, E.; Peumans, W.; De Clercq, E. Alpha-(1-3)- and alpha-(1-6)-D-mannose-specific plant lectins are markedly inhibitory to human immunodeficiency virus and cytomegalovirus infections in vitro. Antimicrob. Agents Chemother. 1991, 35, 410–416. [Google Scholar] [CrossRef] [Green Version]
  189. Balzarini, J.; Neyts, J.; Schols, D.; Hosoya, M.; Van Damme, E.; Peumans, W.; De Clercq, E. The mannose-specific plant lectins from Cymbidium hybrid and Epipactis helleborine and the (N-acetylglucosamine)n-specific plant lectin from Urtica dioica are potent and selective inhibitors of human immunodeficiency virus and cytomegalovirus replication in vitro. Antivir. Res. 1992, 18, 191–207. [Google Scholar] [CrossRef]
  190. Balzarini, J.; Hatse, S.; Vermeire, K.; Princen, K.; Aquaro, S.; Perno, C.-F.; De Clercq, E.; Egberink, H.; Mooter, G.V.; Peumans, W.; et al. Mannose-Specific Plant Lectins from the Amaryllidaceae Family Qualify as Efficient Microbicides for Prevention of Human Immunodeficiency Virus Infection. Antimicrob. Agents Chemother. 2004, 48, 3858–3870. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  191. Krokhin, O.; Li, Y.; Andonov, A.; Feldmann, H.; Flick, R.; Jones, S.; Stroeher, U.; Bastien, N.; Dasuri, K.V.N.; Cheng, K.; et al. Mass Spectrometric Characterization of Proteins from the SARS Virus. Mol. Cell. Proteom. 2003, 2, 346–356. [Google Scholar] [CrossRef]
  192. Salehi, B.; Machin, L.; Monzote, L.; Sharifi-Rad, J.; Ezzat, S.M.; Salem, M.A.; Merghany, R.M.; El Mahdy, N.M.; Kılıç, C.S.; Sytar, O.; et al. Therapeutic Potential of Quercetin: New Insights and Perspectives for Human Health. ACS Omega 2020, 5, 11849–11872. [Google Scholar] [CrossRef] [PubMed]
  193. Coppé, J.-P.; Desprez, P.-Y.; Krtolica, A.; Campisi, J. The Senescence-Associated Secretory Phenotype: The Dark Side of Tumor Suppression. Annu. Rev. Pathol. Mech. Dis. 2010, 5, 99–118. [Google Scholar] [CrossRef] [Green Version]
  194. Guerrero, L.; Castillo, J.; Quiñones, M.; Garcia-Vallvé, S.; Arola, L.; Pujadas, G.; Muguerza, B. Inhibition of Angiotensin-Converting Enzyme Activity by Flavonoids: Structure-Activity Relationship Studies. PLoS ONE 2012, 7, e49493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Wu, W.; Li, R.; Li, X.; He, J.; Jiang, S.; Liu, S.; Yang, J. Quercetin as an Antiviral Agent Inhibits Influenza A Virus (IAV) Entry. Viruses 2015, 8, 6. [Google Scholar] [CrossRef]
  196. Häckl, L.; Cuttle, G.; Dovichi, S.S.; Lima-Landman, M.; Nicolau, M. Inhibition of Angiotensin-Converting Enzyme by Quercetin Alters the Vascular Response to Bradykinin and Angiotensin I. Pharmacology 2002, 65, 182–186. [Google Scholar] [CrossRef]
  197. Thaler, R.; Maurizi, A.; Roschger, P.; Sturmlechner, I.; Khani, F.; Spitzer, S.; Rumpler, M.; Zwerina, J.; Karlic, H.; Dudakovic, A.; et al. Anabolic and Antiresorptive Modulation of Bone Homeostasis by the Epigenetic Modulator Sulforaphane, a Naturally Occurring Isothiocyanate. J. Biol. Chem. 2016, 291, 6754–6771. [Google Scholar] [CrossRef] [Green Version]
  198. Olesin, E.; Nayar, R.; Saikumar-Lakshmi, P.; Berg, L.J. The Transcription Factor Runx2 Is Required for Long-Term Persistence of Antiviral CD8+ Memory T Cells. ImmunoHorizons 2018, 2, 251–261. [Google Scholar] [CrossRef] [Green Version]
  199. Müller, L.; Meyer, M.; Bauer, R.N.; Zhou, H.; Zhang, H.; Jones, S.; Robinette, C.; Noah, T.L.; Jaspers, I. Effect of Broccoli Sprouts and Live Attenuated Influenza Virus on Peripheral Blood Natural Killer Cells: A Randomized, Double-Blind Study. PLoS ONE 2016, 11, e0147742. [Google Scholar] [CrossRef] [Green Version]
  200. Marinella, M.A. Indomethacin and resveratrol as potential treatment adjuncts for SARS-CoV-2/COVID-19. Int. J. Clin. Pract. 2020, 74, e13535. [Google Scholar] [CrossRef]
  201. Medina-Bolivar, F.; Condori, J.; Rimando, A.M.; Hubstenberger, J.; Shelton, K.; O’Keefe, S.F.; Bennett, S.; Dolan, M.C. Production and secretion of resveratrol in hairy root cultures of peanut. Phytochemistry 2007, 68, 1992–2003. [Google Scholar] [CrossRef]
  202. Zhao, Q.; Chen, X.; Martin, C. Scutellaria baicalensis, the golden herb from the garden of Chinese medicinal plants. Sci. Bull. 2016, 61, 1391–1398. [Google Scholar] [CrossRef] [Green Version]
  203. Sowndhararajan, K.; Deepa, P.; Kim, M.; Park, S.J.; Kim, S. Neuroprotective and Cognitive Enhancement Potentials of Baicalin: A Review. Brain Sci. 2018, 8, 104. [Google Scholar] [CrossRef] [Green Version]
  204. Yonesi, M.; Rezazadeh, A. Plants as a prospective source of natural anti-viral compounds and oral vaccines against COVID-19 coronavirus. Preprints 2020. [Google Scholar] [CrossRef]
  205. Liu, H.; Ye, F.; Sun, Q.; Liang, H.; Li, C.; Li, S.; Lu, R.; Huang, B.; Tan, W.; Lai, L. Scutellaria baicalensis extract and baicalein inhibit replication of SARS-CoV-2 and its 3C-like protease in vitro. J. Enzym. Inhib. Med. Chem. 2021, 36, 497–503. [Google Scholar] [CrossRef] [PubMed]
  206. Shah, S.L.; Wahid, F.; Khan, N.; Farooq, U.; Shah, A.J.; Tareen, S.; Ahmad, F.; Khan, T. Inhibitory Effects ofGlycyrrhiza glabraand Its Major Constituent Glycyrrhizin on Inflammation-Associated Corneal Neovascularization. Evid. Based Complement. Altern. Med. 2018, 2018, 8438101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  207. Pilcher, H. Liquorice may tackle SARS. Nature 2003. [Google Scholar] [CrossRef]
  208. Chen, H.; Du, Q. Potential natural compounds for preventing SARS-CoV-2 (2019-nCoV) infection. Preprints 2020. [Google Scholar] [CrossRef]
  209. Dubey, K.; Dubey, R. Computation screening of narcissoside a glycosyloxyflavone for potential novel coronavirus 2019 (COVID-19) inhibitor. Biomed. J. 2020, 43, 363–367. [Google Scholar] [CrossRef]
  210. Akram, M.; Shahab-Uddin, A.A.; Usmanghani, K.; Hannan, A.; Mohiuddin, E.; Asif, M. Curcuma longa and curcumin: A review article. Rom. J. Biol Plant. Biol 2010, 55, 65–70. [Google Scholar]
  211. Hewlings, S.J.; Kalman, D.S. Curcumin: A Review of Its Effects on Human Health. Foods 2017, 6, 92. [Google Scholar] [CrossRef]
  212. Praditya, D.; Kirchhoff, L.; Brüning, J.; Rachmawati, H.; Steinmann, J.; Steinmann, E. Anti-infective Properties of the Golden Spice Curcumin. Front. Microbiol. 2019, 10, 912. [Google Scholar] [CrossRef] [Green Version]
  213. Rein, M.J.; Renouf, M.; Cruz-Hernandez, C.; Actis-Goretta, L.; Thakkar, S.K.; Da Silva Pinto, M. Bioavailability of bioactive food compounds: A challenging journey to bioefficacy. Br. J. Clin. Pharmacol. 2013, 75, 588–602. [Google Scholar] [CrossRef] [Green Version]
  214. Bansal, T.; Awasthi, A.; Jaggi, M.; Khar, R.K.; Talegaonkar, S. Pre-clinical evidence for altered absorption and biliary excretion of irinotecan (CPT-11) in combination with quercetin: Possible contribution of P-glycoprotein. Life Sci. 2008, 83, 250–259. [Google Scholar] [CrossRef]
  215. Shoba, G.; Joy, D.; Joseph, T.; Majeed, M.; Rajendran, R.; Srinivas, P.S.S.R. Influence of Piperine on the Pharmacokinetics of Curcumin in Animals and Human Volunteers. Planta Med. 1998, 64, 353–356. [Google Scholar] [CrossRef] [Green Version]
  216. Basu, P.; Dutta, S.; Begum, R.; Mittal, S.; Das Dutta, P.; Bharti, A.C.; Panda, C.K.; Biswas, J.; Dey, B.; Talwar, G.P.; et al. Clearance of cervical human papillomavirus infection by topical application of curcumin and curcumin containing polyherbal cream: A phase II randomized controlled study. Asian Pac. J. Cancer Prev. 2013, 14, 5753–5759. [Google Scholar] [CrossRef] [Green Version]
  217. Lobo, V.; Patil, A.; Phatak, A.; Chandra, N. Free radicals, antioxidants and functional foods: Impact on human health. Pharmacogn. Rev. 2010, 4, 118–126. [Google Scholar] [CrossRef] [Green Version]
  218. Utomo, R.Y.; Meiyanto, E. Revealing the potency of citrus and galangal constituents to halt SARS-CoV-2 infection. Preprints 2020. [Google Scholar] [CrossRef] [Green Version]
  219. Del Rio, D.; Stewart, A.J.; Mullen, W.; Burns, J.; Lean, M.E.J.; Brighenti, A.F.; Crozier, A. HPLC-MSnAnalysis of Phenolic Compounds and Purine Alkaloids in Green and Black Tea. J. Agric. Food Chem. 2004, 52, 2807–2815. [Google Scholar] [CrossRef]
  220. Lucas, L.; Russell, A.; Keast, R. Molecular Mechanisms of Inflammation. Anti-Inflammatory Benefits of Virgin Olive Oil and the Phenolic Compound Oleocanthal. Curr. Pharm. Des. 2011, 17, 754–768. [Google Scholar] [CrossRef]
  221. Remely, M.; Ferk, F.; Sterneder, S.; Setayesh, T.; Kepcija, T.; Roth, S.; Noorizadeh, R.; Greunz, M.; Rebhan, I.; Wagner, K.-H.; et al. Vitamin E Modifies High-Fat Diet-Induced Increase of DNA Strand Breaks, and Changes in Expression and DNA Methylation of Dnmt1 and MLH1 in C57BL/6J Male Mice. Nutrients 2017, 9, 607. [Google Scholar] [CrossRef] [Green Version]
  222. Kaihatsu, K.; Yamabe, M.; Ebara, Y. Antiviral Mechanism of Action of Epigallocatechin-3-O-gallate and Its Fatty Acid Esters. Molecules 2018, 23, 2475. [Google Scholar] [CrossRef] [Green Version]
  223. Ehrhardt, C.; Hrincius, E.R.; Korte, V.; Mazur, I.; Droebner, K.; Poetter, A.; Dreschers, S.; Schmolke, M.; Planz, O.; Ludwig, S. A polyphenol rich plant extract, CYSTUS052, exerts anti influenza virus activity in cell culture without toxic side effects or the tendency to induce viral resistance. Antivir. Res. 2007, 76, 38–47. [Google Scholar] [CrossRef]
  224. Barak, V.; Halperin, T.; Kalickman, I. The effect of Sambucol, a black elderberry-based, natural product, on the production of human cytokines: I. Inflammatory cytokines. Eur. Cytokine Netw. 2001, 12, 290–296. [Google Scholar]
  225. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Karamoddini, M.K.; Emami, S.A.; Ghannad, M.S.; Sani, E.A.; Sahebkar, A. Antiviral activities of aerial subsets of Artemisia species against Herpes Simplex virus type 1 (HSV1) in vitro. Asian Biomed. 2011, 5, 63–68. [Google Scholar] [CrossRef]
  227. Khan, M.A.A.; Jain, D.; Bhakuni, R.; Zaim, M.; Thakur, R. Occurrence of some antiviral sterols in Artemisia annua. Plant Sci. 1991, 75, 161–165. [Google Scholar] [CrossRef]
  228. Ferreira, J.F.; Luthria, D.L.; Sasaki, T.; Heyerick, A. Flavonoids from Artemisia annua L. as Antioxidants and Their Potential Synergism with Artemisinin against Malaria and Cancer. Molecules 2010, 15, 3135–3170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  229. Day, B.J. Antioxidants as Potential Therapeutics for Lung Fibrosis. Antioxid. Redox Signal. 2008, 10, 355–370. [Google Scholar] [CrossRef] [Green Version]
  230. Wang, C.; Xuan, X.; Yao, W.; Huang, G.; Jin, J. Anti-profibrotic effects of artesunate on bleomycin-induced pulmonary fibrosis in Sprague Dawley rats. Mol. Med. Rep. 2012, 12, 1291–1297. [Google Scholar] [CrossRef] [Green Version]
  231. Roberfroid, M.B. Inulin-Type Fructans: Functional Food Ingredients. J. Nutr. 2007, 137, 2493S–2502S. [Google Scholar] [CrossRef] [Green Version]
  232. Sanchez, M.; Lodi, F.; Vera, R.; Villar, I.C.; Cogolludo, A.; Jimenez, R.; Moreno, L.; Romero, M.; Tamargo, J.; Perez-Vizcaino, F.; et al. Quercetin and Isorhamnetin Prevent Endothelial Dysfunction, Superoxide Production, and Overexpression of p47phox Induced by Angiotensin II in Rat Aorta. J. Nutr. 2007, 137, 910–915. [Google Scholar] [CrossRef] [Green Version]
  233. Chen, L.; Li, J.; Luo, C.; Liu, H.; Xu, W.; Chen, G.; Liew, O.W.; Zhu, W.; Puah, C.M.; Shen, X.; et al. Binding interaction of quercetin-3-β-galactoside and its synthetic derivatives with SARS-CoV 3CLpro: Structure–activity relationship studies reveal salient pharmacophore features. Bioorg. Med. Chem. 2006, 14, 8295–8306. [Google Scholar] [CrossRef]
  234. Okhuaroboa, A.; Falodun, J.E.; Erharuyicd, O.; Imiejec, V.; Faloduncde, A.; Langerc, P. Harnessing the medicinal properties of Andrographis paniculata for diseases and beyond: A review of its phytochemistry and pharmacology. Asian Pac. J. Trop. Dis. 2014, 4, 213–222. [Google Scholar] [CrossRef]
  235. Yarnell, E. Herbs for Viral Respiratory Infections. Altern. Complement. Ther. 2018, 24, 35–43. [Google Scholar] [CrossRef]
  236. Arora, R.; Chawla, R.; Marwah, R.; Arora, P.; Sharma, R.K.; Kaushik, V.; Goel, R.; Kaur, A.; Silambarasan, M.; Tripathi, R.P.; et al. Potential of Complementary and Alternative Medicine in Preventive Management of Novel H1N1 Flu (Swine Flu) Pandemic: Thwarting Potential Disasters in the Bud. Evid. Based Complement. Altern. Med. 2010, 2011, 586506. [Google Scholar] [CrossRef]
  237. Coon, J.T.; Ernst, E. Andrographis paniculata in the treatment and prevention of upper respiratory tract infection (URTI): A systematic review of safety and efficacy. Focus Altern. Complement. Ther. 2010, 8, 160–161. [Google Scholar] [CrossRef]
  238. Liu, Y.-T.; Chen, H.-W.; Lii, C.-K.; Jhuang, J.-H.; Huang, C.-S.; Li, M.-L.; Yao, H.-T. A Diterpenoid, 14-Deoxy-11, 12-Didehydroandrographolide, in Andrographis paniculata Reduces Steatohepatitis and Liver Injury in Mice Fed a High-Fat and High-Cholesterol Diet. Nutrients 2020, 12, 523. [Google Scholar] [CrossRef] [Green Version]
  239. Liu, Z.; Xiao, X.; Wei, X.; Li, J.; Yang, J.; Tan, H.; Zhu, J.; Zhang, Q.; Wu, J.; Liu, L. Composition and divergence of coronavirus spike proteins and host ACE2 receptors predict potential intermediate hosts of SARS-CoV-2. J. Med Virol. 2020, 92, 595–601. [Google Scholar] [CrossRef] [Green Version]
  240. Mpiana, P.T.; Ngbolua, K.-T.-N.; Tshibangu, D.S.T.; Kilembe, J.T.; Gbolo, B.Z.; Mwanangombo, D.T.; Inkoto, C.L.; Lengbiye, E.M.; Mbadiko, C.M.; Matondo, A.; et al. Aloe vera (L.) Burm. F. as a Potential Anti-COVID-19 Plant: A Mini-review of Its Antiviral Activity. Eur. J. Med. Plants 2020, 86–93. [Google Scholar] [CrossRef]
  241. Velthuis, A.J.W.T.; Worm, S.H.E.V.D.; Sims, A.C.; Baric, R.S.; Snijder, E.J.; Van Hemert, M.J. Zn2+ Inhibits Coronavirus and Arterivirus RNA Polymerase Activity In Vitro and Zinc Ionophores Block the Replication of These Viruses in Cell Culture. PLoS Pathog. 2010, 6, e1001176. [Google Scholar] [CrossRef]
  242. Mohammadi, E.; Golchin, M. High protection of mice against Brucella abortus by oral immunization with recombinant probiotic Lactobacillus casei vector vaccine, expressing the outer membrane protein OMP19 of Brucella species. Comp. Immunol. Microbiol. Infect. Dis. 2020, 70, 101470. [Google Scholar] [CrossRef]
  243. Chang, H.-C.; Hung, W.-C.; Huang, M.-S.; Hsu, H.-K. Extract from the Leaves of Toona sinensis Roemor Exerts Potent Antiproliferative Effect on Human Lung Cancer Cells. Am. J. Chin. Med. 2002, 30, 307–314. [Google Scholar] [CrossRef]
  244. Yang, H.-L.; Chang, W.-H.; Chia, Y.-C.; Huang, C.-J.; Lu, F.-J.; Hsu, H.-K.; Hseu, Y.-C. Toona sinensis extracts induces apoptosis via reactive oxygen species in human premyelocytic leukemia cells. Food Chem. Toxicol. 2006, 44, 1978–1988. [Google Scholar] [CrossRef]
  245. Wang, P.-H.; Tsai, M.-J.; Hsu, C.-Y.; Wang, C.-Y.; Hsu, H.-K.; Weng, C.-F. Toona sinensis Roem (Meliaceae) leaf extract alleviates hyperglycemia via altering adipose glucose transporter 4. Food Chem. Toxicol. 2008, 46, 2554–2560. [Google Scholar] [CrossRef]
  246. Chen, C.-J.; Michaelis, M.; Hsu, H.-K.; Tsai, C.-C.; Yang, K.D.; Wu, Y.-C.; Cinatl, J.; Doerr, H.W. Toona sinensis Roem tender leaf extract inhibits SARS coronavirus replication. J. Ethnopharmacol. 2008, 120, 108–111. [Google Scholar] [CrossRef]
  247. Qin, G.-W.; Xu, R.-S. Recent advances on bioactive natural products from Chinese medicinal plants. Med. Res. Rev. 1998, 18, 375–382. [Google Scholar] [CrossRef]
  248. Wu, X.; Qin, G.; Cheung, K.K.; Cheng, K.F. New alkaloids from Isatis indigotica. Tetrahedron 1997, 53, 13323–13328. [Google Scholar] [CrossRef]
  249. McGovern, S.L.; Shoichet, B.K. Information Decay in Molecular Docking Screens against Holo, Apo, and Modeled Conformations of Enzymes. J. Med. Chem. 2003, 46, 2895–2907. [Google Scholar] [CrossRef] [PubMed]
  250. Andersen, D.O.; Weber, N.D.; Wood, S.G.; Hughes, B.G.; Murray, B.K.; North, J.A. In vitro virucidal activity of selected anthraquinones and anthraquinone derivatives. Antivir. Res. 1991, 16, 185–196. [Google Scholar] [CrossRef]
  251. Mak, N.-K.; Leung, C.-Y.; Wei, X.-Y.; Shen, X.-L.; Wong, R.N.-S.; Leung, K.-N.; Fung, M.-C. Inhibition of RANTES expression by indirubin in influenza virus-infected human bronchial epithelial cells. Biochem. Pharmacol. 2004, 67, 167–174. [Google Scholar] [CrossRef]
  252. Roy, S.; Bhattacharyya, P. Possible role of traditional medicinal plant Neem (Azadirachta indica) for the management of COVID-19 infection. Int. J. Res. Pharm. Sci. 2020, 11, 122–125. [Google Scholar] [CrossRef]
  253. Xu, Z.; Shi, L.; Wang, Y.; Zhang, J.; Huang, L.; Zhang, C.; Liu, S.; Zhao, P.; Liu, H.; Zhu, L.; et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir. Med. 2020, 8, 420–422. [Google Scholar] [CrossRef]
  254. Sujarwo, W.; Keim, A.P.; Caneva, G.; Toniolo, C.; Nicoletti, M. Ethnobotanical uses of neem (Azadirachta indica A. Juss.; Meliaceae) leaves in Bali (Indonesia) and the Indian subcontinent in relation with historical background and phytochemical properties. J. Ethnopharmacol. 2016, 189, 186–193. [Google Scholar] [CrossRef]
  255. Thakurta, P.; Bhowmik, P.; Mukherjee, S.; Hajra, T.K.; Patra, A.; Bag, P.K. Antibacterial, antisecretory and antihemorrhagic activity of Azadirachta indica used to treat cholera and diarrhea in India. J. Ethnopharmacol. 2007, 111, 607–612. [Google Scholar] [CrossRef] [PubMed]
  256. Bandyopadhyay, U.; Biswas, K.; Chatterjee, R.; Bandyopadhyay, D.; Chattopadhyay, I.; Ganguly, C.K.; Chakraborty, T.; Bhattacharya, K.; Banerjee, R.K. Gastroprotective effect of Neem (Azadirachta indica) bark extract: Possible involvement of H+-K+-ATPase inhibition and scavenging of hydroxyl radical. Life Sci. 2002, 71, 2845–2865. [Google Scholar] [CrossRef]
  257. Alam, G.; Wahyuono, S.; Ganjar, I.G.; Hakim, L.; Timmerman, H.; Verpoorte, R. Tracheospasmolytic Activity of Viteosin-A and Vitexicarpin Isolated fromVitex trifolia. Planta Med. 2002, 68, 1047–1049. [Google Scholar] [CrossRef] [PubMed]
  258. Vellingiri, B.; Jayaramayya, K.; Iyer, M.; Narayanasamy, A.; Govindasamy, V.; Giridharan, B.; Ganesan, S.; Venugopal, A.; Venkatesan, D.; Ganesan, H.; et al. COVID-19: A promising cure for the global panic. Sci. Total. Environ. 2020, 725, 138277. [Google Scholar] [CrossRef]
  259. Dashputre, N.L.; Naikwade, N.S. Immunomodulatory activity of Abutilon indicum linn on albino mice. Int J. Pharm Sci Res. 2010, 1, 178–184. [Google Scholar]
  260. Grover, A.; Agrawal, V.; Shandilya, A.; Bisaria, V.S.; Sundar, D. Non-nucleosidic inhibition of Herpes simplex virus DNA polymerase: Mechanistic insights into the anti-herpetic mode of action of herbal drug withaferin A. BMC Bioinform. 2011, 12, S22. [Google Scholar] [CrossRef] [Green Version]
  261. Mehta, P.; McAuley, D.F.; Brown, M.; Sanchez, E.; Tattersall, R.S.; Manson, J.J. COVID-19: Consider cytokine storm syndromes and immunosuppression. Lancet 2020, 395, 1033–1034. [Google Scholar] [CrossRef]
  262. Mohan, C.; Dinakar, S.; Anand, T.; Elayaraja, R.; SathiyaPriya, B. Phytochemical, GC-MS analysis and Antibacterial activity of a Medicinal Plant Acalypha indica. Int. J. Pharm. Tech. Res. 2012, 4, 1050–1054. [Google Scholar]
  263. Dhaarani, V.; Sarvalingam, A.; Rajendran, A. Medicinal uses of psammophytic plants in Tranquebar regions of Tamil Nadu, India. J. Herbs Spices Med. Plants 2018, 24, 282–292. [Google Scholar] [CrossRef]
  264. Premila, M.; Conboy, L. Ayurvedic Herbs: A Clinical Guide to the Healing Plants of Traditional Indian Medicine. J. Altern. Complement. Med. 2007, 13, 841–842. [Google Scholar] [CrossRef]
  265. Vaghasiya, Y.; Chanda, S. Screening of some traditionally used Indian plants for antibacterial activity against Klebsiella pneumonia. J. Herb Med. Toxicol. 2009, 3, 161–164. [Google Scholar]
  266. Kundu, S.; Roy, S.; Lyndem, L.M. Cassia alata L.: Potential role as anthelmintic agent against Hymenolepis diminuta. Parasitol. Res. 2012, 111, 1187–1192. [Google Scholar] [CrossRef]
  267. Singh, R.P.; Jain, D.A. Evaluation of antimicrobial activity of alcoholic and aqueous extracts of five plants used in traditional medicine in North India. Int. J. Pharm. Tech. Res. 2011, 3, 376–380. [Google Scholar]
  268. Nayak, V.; Jadhav, V.; Sajjanshetty, M.R. Traditional medicine in the management of Recurrent Tonsillitis—An Ayurvedic Perspective. J. Ayurveda Integr. Med Sci. 2017, 2, 98–106. [Google Scholar] [CrossRef] [Green Version]
  269. Harwansh, R.K.; Pareta, S.K.; Patra, K.C.; Rahman, M.A. Preliminary phytochemical screening and anthelmintic activity of Chloroxylon swietenia root extract. Int. J. Phytomed. 2010, 2, 255–259. [Google Scholar]
  270. Jacob, L.; Latha, M.S. In vitro antioxidant activity of Clitoria ternatea Linn. Int. J. Res. Phytochem. Pharmacol. 2013, 3, 35–39. [Google Scholar]
  271. Dharmananda, S. Tibetan herbal medicine. Int. J. Orient. Med. 2011, 26, 1–13. [Google Scholar]
  272. Boothapandi, M.; Ramanibai, R. Immunomodulatory activity of Indigofera tinctoria leaf extract on in vitro macrophage responses and lymphocyte proliferation. Int. J. Pharm. Pharm. Sci. 2016, 8, 58–63. [Google Scholar]
  273. Afzal, U.; Gulfraz, M.; Hussain, S.; Malik, F.; Maqsood, S.; Shah, I.; Mahmood, S. Hepatoprotective effects of Justicia adhatoda L. against carbon tetrachloride (CCl4) induced liver injury in Swiss albino mice. Afr. J. Pharm. Pharmacol. 2013, 7, 8–14. [Google Scholar] [CrossRef] [Green Version]
  274. John, K.M.; Enkhtaivan, G.; Ayyanar, M.; Jin, K.; Yeon, J.B.; Kim, D.H. Screening of ethnic medicinal plants of South India against influenza (H1N1) and their antioxidant activity. Saudi J. Biol. Sci. 2015, 22, 191–197. [Google Scholar] [CrossRef] [Green Version]
  275. Kaur, A.; Katoch, D.; Singh, B.; Arora, S. Seclusion of vasicine-and quinazoline alkaloid from bioactive fraction of Justicia adhathoda and its antioxidant, antimutagenic and anticancerous activities. J. Glob. Biosci. 2016, 5, 3836–3850. [Google Scholar]
  276. Ravindran, R.; Juliet, S.; Sunil, A.; Kumar, K.A.; Nair, S.N.; Amithamol, K.; Shynu, M.; Rawat, A.K.S.; Ghosh, S. Eclosion blocking effect of ethanolic extract of Leucas aspera (Lamiaceae) on Rhipicephalus (Boophilus) annulatus. Vet. Parasitol. 2011, 179, 287–290. [Google Scholar] [CrossRef]
  277. Thenmozhi, M.; Dhanalakshmi, M.; Devi, K.M.; Sushila, K.; Thenmozhi, S. Evaluation of hepatoprotective activity of Leucas aspera hydroalcoholic leaf extract during exposure to lead acetate in male albino Wistar rats. Asian J. Pharm Clin. Res. 2013, 6, 78–81. [Google Scholar]
  278. Kalita, N.; Kalita, M.C. Ethnomedicinal plants of Assam, India as an alternative source of future medicine for treatment of Pneumonia. Int. Res. J. 2014, 3, 76–82. [Google Scholar]
  279. Sureshkumar, S.; Mishra, S. Hepatoprotective effect of extracts from Pergularia daemia Forsk. J. Ethnopharmacol. 2006, 107, 164–168. [Google Scholar] [CrossRef] [PubMed]
  280. Naika, R.; Prasanna, K.P.; Ganapathy, P.S. Antibacterial activity of piperlongumine an alkaloid isolated from methanolic root extract of Piper Longum, L. Pharmacophore 2010, 1, 141–148. [Google Scholar]
  281. Vadakkan, K.; Hemapriya, J.; Selvaraj, V. Quorum quenching intervened in vivo attenuation and immunological clearance enhancement by Solanum torvum root extract against Pseudomonas aeruginosa instigated pneumonia in Sprague Dawley rats. Clin. Phytosci. 2019, 5, 24. [Google Scholar] [CrossRef] [Green Version]
  282. Abdel-Rahman, M.K.; Abd El-Megeid, A.A. Hepatoprotective effect of soapworts (Saponaria officinalis), pomegranate peel (Punica granatum L) and cloves (Syzygium aromaticum linn) on mice with CCl4 hepatic intoxication. World J. Chem. 2006, 1, 41–46. [Google Scholar]
  283. Mishra, A.; Kumar, S.; Pandey, A.K. Scientific Validation of the Medicinal Efficacy of Tinospora cordifolia. Sci. World J. 2013, 2013, 292934. [Google Scholar] [CrossRef] [Green Version]
  284. Tharakan, B.; Dhanasekaran, M.; Manyam, B.V. Antioxidant and DNA protecting properties of anti-fatigue herb Trichopus zeylanicus. Phytotherapy Res. 2005, 19, 669–673. [Google Scholar] [CrossRef] [PubMed]
  285. More, D.; Shetty, V.; Shah, A.; Mungekar, S.; Kamat, S.K.; Rege, N.N. Effects of polyherbal ayurvedic formulations on non-specific immunity: An experimental study. Indian J. Pharmacol. 2008, 40, S160–S171. [Google Scholar]
  286. Koh, E.M.; Kim, H.J.; Kim, S.; Choi, W.H.; Choi, Y.H.; Ryu, S.Y.; Kim, Y.S.; Koh, W.S.; Park, S.-Y. Modulation of Macrophage Functions by Compounds Isolated from Zingiber officinale. Planta Med. 2008, 75, 148–151. [Google Scholar] [CrossRef] [Green Version]
  287. Chang, J.S.; Wang, K.C.; Yeh, C.F.; Shieh, D.E.; Chiang, L.C. Fresh ginger (Zingiber officinale) has anti-viral activity against human respiratory syncytial virus in human respiratory tract cell lines. J. Ethnopharmacol. 2013, 145, 146–151. [Google Scholar] [CrossRef]
  288. Akoachere, J.-F.T.K.; Ndip, R.N.; Chenwi, E.B.; Ndip, L.M.; Njock, T.E.; Anong, D.N. Antibacterial effect of Zingiber officinale and Garcinia kola on respiratory tract pathogens. East Afr. Med. J. 2002, 79, 588–592. [Google Scholar] [CrossRef] [Green Version]
  289. Ali, T.; Shahbaz, B.; Suleri, A. Analysis of myths and realities of deforestation in Northwest Pakistan: Implications for forestry extension. Int. J. Agric. Biol. 2006, 8, 107–110. [Google Scholar]
  290. Abbasi, A.M.; Iqbal, J.; Khan, M.A.; Shah, M.H. Health risk assessment and multivariate apportionment of trace metals in wild leafy vegetables from Lesser Himalayas, Pakistan. Ecotoxicol. Environ. Saf. 2013, 92, 237–244. [Google Scholar] [CrossRef]
  291. Singh, G.D.; Kaiser, P.; Youssouf, M.S.; Singh, S.; Khajuria, A.; Koul, A.; Bani, S.; Kapahi, B.K.; Satti, N.K.; Suri, K.A.; et al. Inhibition of early and late phase allergic reactions by Euphorbia hirta L. Phytotherapy Res. 2006, 20, 316–321. [Google Scholar] [CrossRef]
  292. Ko, F.-N.; Huang, T.-F.; Teng, C.-M. Vasodilatory action mechanisms of apigenin isolated from Apium graveolens in rat thoracic aorta. Biochim. Biophys. Acta Gen. Subj. 1991, 1115, 69–74. [Google Scholar] [CrossRef]
  293. Khare, C.P. Indian Medicinal Plants: An Illustrated Dictionary; Springer: Berlin/Heidelberg, Germany, 2008. [Google Scholar]
  294. Walter, T.M. Review of Acalypha indica Linn. in Traditional Siddha Medicine. 2007. Available online: https://www.academia.edu/1410079/Review_of_Review_of_Acalypha_indica_Linn_in_Traditional_Siddha_Acalypha_indica_Linn_in_Traditional_Siddha_Medicine (accessed on 5 March 2021).
  295. Hosseinzadeh, H.; Ghenaati, J. Evaluation of the antitussive effect of stigma and petals of saffron (Crocus sativus) and its components, safranal and crocin in guinea pigs. Fitoterapia 2006, 77, 446–448. [Google Scholar] [CrossRef]
  296. Joshi, V.K.; Joshi, A.; Dhiman, K.S. The Ayurvedic Pharmacopoeia of India, development and perspectives. J. Ethnopharmacol. 2017, 197, 32–38. [Google Scholar] [CrossRef]
  297. Hirata, N.; Naruto, S.; Inaba, K.; Itoh, K.; Tokunaga, M.; Iinuma, M.; Matsuda, H. Histamine Release Inhibitory Activity of Piper nigrum Leaf. Biol. Pharm. Bull. 2008, 31, 1973–1976. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  298. Naseri, M.K.; Yahyavi, H. Antispasmodic effect of Piper nigrum fruit hot water extract on rat ileum. Pak. J. Biol. Sci 2008, 11, 1492–1496. [Google Scholar] [PubMed]
  299. Bharali, R.; Azad, M.R.H.; Tabassum, J. Chemopreventive Action of Boerhaavia Diffusa on DMDA-induced Skin Carcinogenesis in Mice. Indian J. Physiol. Pharmacol. 2003, 47, 459–464. [Google Scholar] [PubMed]
  300. Simin, K.; Khaliq-Uz-Zaman, S.M.; Ahmad, V.U. Antimicrobial activity of seed extracts and bondenolide from Caesalpinia bonduc (L.) Roxb. Phytother. Res. 2001, 15, 437–440. [Google Scholar] [CrossRef]
  301. Shinwari, Z.K. Medicinal plants research in Pakistan. J. Med. Plant. Res. 2010, 4, 161–176. [Google Scholar] [CrossRef]
  302. Thwe, A.; Arasu, M.V.; Li, X.; Park, C.H.; Kim, S.J.; Al-Dhabi, N.A.; Park, S.U. Effect of Different Agrobacterium rhizogenes Strains on Hairy Root Induction and Phenylpropanoid Biosynthesis in Tartary Buckwheat (Fagopyrum tataricum Gaertn). Front. Microbiol. 2016, 7, 318. [Google Scholar] [CrossRef]
  303. Shanmugavelu, M. Noikaluku Siddha parikaram. Part I; Indian Medicine-Homeopathy Department, Government of India: New Delhi, India, 2004.
  304. Moineddin, R.; Nie, J.X.; Domb, G.; Leong, A.M.; Upshur, R.E.G. Seasonality of primary care utilization for respiratory diseases in Ontario: A time-series analysis. BMC Health Serv. Res. 2008, 8, 160. [Google Scholar] [CrossRef] [Green Version]
  305. Srivastava, A.K.; Kumar, A.; Misra, N. On the Inhibition of COVID-19 Protease by Indian Herbal Plants: An in Silico Investigation. arXiv 2020, arXiv:2004.03411 2020. [Google Scholar]
  306. Alsaffar, D.F. In Silico Molecular Docking Studies of Medicinal Arabic Plant-Based Bioactive Compounds as a Promising Drug Candidate against COVID-19. Int. J. Innov. Sci. Res. Technol. 2020, 5, 876–896. [Google Scholar]
  307. Gautret, P.; Lagier, J.-C.; Parola, P.; Hoang, V.T.; Meddeb, L.; Sevestre, J.; Mailhe, M.; Doudier, B.; Aubry, C.; Amrane, S.; et al. Clinical and microbiological effect of a combination of hydroxychloroquine and azithromycin in 80 COVID-19 patients with at least a six-day follow up: A pilot observational study. Travel Med. Infect. Dis. 2020, 34, 101663. [Google Scholar] [CrossRef]
  308. Juurlink, D.N. Safety considerations with chloroquine, hydroxychloroquine and azithromycin in the management of SARS-CoV-2 infection. Can. Med. Assoc. J. 2020, 192, E450–E453. [Google Scholar] [CrossRef] [Green Version]
  309. Gurwitz, D. Angiotensin receptor blockers as tentative SARS-CoV-2 therapeutics. Drug Dev. Res. 2020, 81, 537–540. [Google Scholar] [CrossRef] [Green Version]
  310. Shen, C.; Wang, Z.; Zhao, F.; Yang, Y.; Li, J.; Yuan, J.; Wang, F.; Li, D.; Yang, M.; Xing, L.; et al. Treatment of 5 Critically Ill Patients With COVID-19 With Convalescent Plasma. JAMA 2020, 323, 1582. [Google Scholar] [CrossRef]
  311. Dhama, K.; Sharun, K.; Tiwari, R.; Dadar, M.; Malik, Y.S.; Singh, K.P.; Chaicumpa, W. COVID-19, an emerging coronavirus infection: Advances and prospects in designing and developing vaccines, immunotherapeutics, and therapeutics. Hum. Vaccines Immunother. 2020, 16, 1232–1238. [Google Scholar] [CrossRef] [Green Version]
  312. Uddin, M.; Mustafa, F.; Rizvi, T.A.; Loney, T.; Al Suwaidi, H.; Al-Marzouqi, A.H.H.; Eldin, A.K.; Alsabeeha, N.; Adrian, T.E.; Stefanini, C.; et al. SARS-CoV-2/COVID-19: Viral Genomics, Epidemiology, Vaccines, and Therapeutic Interventions. Viruses 2020, 12, 526. [Google Scholar] [CrossRef]
  313. Sanders, J.M.; Monogue, M.L.; Jodlowski, T.Z.; Cutrell, J.B. Pharmacologic Treatments for Coronavirus Disease 2019 (COVID-19). JAMA 2020, 323, 1824–1836. [Google Scholar] [CrossRef]
  314. Menachery, V.D.; Gralinski, L.E.; Mitchell, H.D.; Dinnon, K.H.; Leist, S.R.; Yount, B.L.; McAnarney, E.T.; Graham, R.L.; Waters, K.M.; Baric, R.S. Combination Attenuation Offers Strategy for Live Attenuated Coronavirus Vaccines. J. Virol. 2018, 92. [Google Scholar] [CrossRef] [Green Version]
  315. Fischer, R.; Buyel, J. Molecular farming—The slope of enlightenment. Biotechnol. Adv. 2020, 40, 107519. [Google Scholar] [CrossRef]
  316. Gleba, Y.; Klimyuk, V.; Marillonnet, S. Magnifection?a new platform for expressing recombinant vaccines in plants. Vaccine 2005, 23, 2042–2048. [Google Scholar] [CrossRef]
  317. A Critical Review of the Concept of Transgenic Plants: Insights into Pharmaceutical Biotechnology and Molecular Farming. Curr. Issues Mol. Biol. 2016, 18, 21–42. [CrossRef] [Green Version]
  318. Rosales-Mendoza, S. Will plant-made biopharmaceuticals play a role in the fight against COVID-19? Expert Opin. Biol. Ther. 2020, 20, 545–548. [Google Scholar] [CrossRef] [Green Version]
  319. Malik, S. Production of Plant Derived Natural Compounds through Hairy Root Culture; Springer: Berlin/Heidelberg, Germany, 2017. [Google Scholar]
  320. Grzegorczyk, I.; Królicka, A.; Wysokińska, H. Establishment of Salvia officinalis L. hairy root cultures for the production of rosmarinic acid. Z. Naturforschung C 2006, 61, 351–356. [Google Scholar] [CrossRef]
  321. Tavassoli, P.; Afshar, A.S. Influence of different Agrobacterium rhizogenes strains on hairy root induction and analysis of phenolic and flavonoid compounds in marshmallow (Althaea officinalis L.). 3 Biotech 2018, 8, 351. [Google Scholar] [CrossRef]
  322. Park, C.H.; Zhao, S.; Yeo, H.J.; Park, Y.E.; Baska, T.B.; Arasu, M.V.; Al-Dhabi, N.A.; Park, S.U. Comparison of Different Strains of Agrobacterium rhizogenes for Hairy Root Induction and Betulin and Betulinic Acid Production in Morus alba. Nat. Prod. Commun. 2017, 12, 1934578X1701200403. [Google Scholar] [CrossRef] [Green Version]
  323. Yousefian, S.; Lohrasebi, T.; Farhadpour, M.; Haghbeen, K. Production of phenolic acids in hairy root cultures of medicinal plant Mentha spicata L. in response to elicitors. Mol. Biol. Res. Commun. 2020, 9, 23–34. [Google Scholar]
  324. Gabr, A.; Sytar, O.; Ahmed, A.R.; Smetanska, I. Production of phenolic acid and antioxidant activity in transformed hairy root cultures of common buckwheat (Fagopyrum Esculentum M.). Aust. J. Basic Appl. Sci. 2012, 6, 577–586. [Google Scholar]
  325. Zhou, Y.; Hirotani, M.; Yoshikawa, T.; Furuya, T. Flavonoids and phenylethanoids from hairy root cultures of Scutellaria baicalensis. Phytochemistry 1997, 44, 83–87. [Google Scholar] [CrossRef]
  326. Skala, E.K.A.; Olszewska, M.A.; Kiss, A.K.; WysokiNska, H. Establishment of Hairy Root Cultures of Rhaponticum carthamoides (Willd.) Iljin for the Production of Biomass and Caffeic Acid Derivatives. BioMed Res. Int. 2015. [Google Scholar] [CrossRef] [Green Version]
  327. Balasubramanian, M.; Anbumegala, M.; Surendran, R.; Arun, M.; Shanmugam, G. Elite hairy roots of Raphanus sativus (L.) as a source of antioxidants and flavonoids. 3 Biotech 2018, 8, 128. [Google Scholar] [CrossRef]
  328. Thiruvengadam, M.; Praveen, N.; Kim, E.-H.; Kim, S.-H.; Chung, I.-M. Production of anthraquinones, phenolic compounds and biological activities from hairy root cultures of Polygonum multiflorum Thunb. Protoplasma 2014, 251, 555–566. [Google Scholar] [CrossRef]
  329. Hosseini, S.M.; Bahramnejad, B.; Baneh, H.D.; Emamifar, A.; Goodwin, P.H. Hairy root culture optimization and resveratrol production from Vitis vinifera subsp. sylvesteris. World J. Microbiol. Biotechnol. 2017, 33, 67. [Google Scholar] [CrossRef] [PubMed]
  330. Lee, S.-W.; Kim, Y.S.; Uddin, R.; Kwon, D.Y.; Kim, Y.B.; Lee, M.Y.; Kim, S.-J.; Park, S.U. Resveratrol Production from Hairy Root Cultures of Scutellaria baicalensis. Nat. Prod. Commun. 2013, 8, 1934578x1300800517. [Google Scholar] [CrossRef] [Green Version]
  331. Ludwig-Müller, J.; Jahn, L.; Lippert, A.; Püschel, J.; Walter, A. Improvement of hairy root cultures and plants by changing biosynthetic pathways leading to pharmaceutical metabolites: Strategies and applications. Biotechnol. Adv. 2014, 32, 1168–1179. [Google Scholar] [CrossRef]
  332. Halder, M.; Sarkar, S.; Jha, S. Elicitation: A biotechnological tool for enhanced production of secondary metabolites in hairy root cultures. Eng. Life Sci. 2019, 19, 880–895. [Google Scholar] [CrossRef] [Green Version]
  333. Vasconsuelo, A.; Boland, R. Molecular aspects of the early stages of elicitation of secondary metabolites in plants. Plant Sci. 2007, 172, 861–875. [Google Scholar] [CrossRef]
  334. Putalun, W. Technology of compact MAb and its application for medicinal plant breeding named as missile type molecular breeding. Curr. Drug Discov. Technol. 2011, 8, 24–31. [Google Scholar] [CrossRef]
  335. Hoseinpanahi, B.; Bahramnejad, B.; Majdi, M.; Dastan, D.; Ashengroph, M. The effect of different elicitors on hairy root biomass and resveratrol production in wild Vitis vinifera. Appl. Biotechnol. Rep. 2020, 7, 25–31. [Google Scholar] [CrossRef]
Figure 1. The most important viral replication chain includes virus attack to the host cell (Steps 1–3); entry using receptor bindings (Steps 1 and 4–6); and mRNA transcription/replication, and protein translation, and assembly and budding of progeny virus particles (Steps 1, 7, and 8). These steps are the most important goals for viral polymerase inhibition, replication (e.g., protease inhibitors, inhibitors of entry, and integrate inhibitors, among others), budding, and assembly [23].
Figure 1. The most important viral replication chain includes virus attack to the host cell (Steps 1–3); entry using receptor bindings (Steps 1 and 4–6); and mRNA transcription/replication, and protein translation, and assembly and budding of progeny virus particles (Steps 1, 7, and 8). These steps are the most important goals for viral polymerase inhibition, replication (e.g., protease inhibitors, inhibitors of entry, and integrate inhibitors, among others), budding, and assembly [23].
Molecules 26 03868 g001
Figure 2. The replication cycle of SARS-CoV-2 and its inhibitors. The binding of the spike (S) protein to the host cell receptor initiates SARS-CoV-2 infection. SARS-CoV-2 has so far been linked to two cellular receptors: angiotensin-converting enzyme 2 (ACE2) and CD147. The cleavage of the S protein by the cell surface-associated enzyme occurs after receptor engagement. The viral genomic RNA is translated through ribosomal frameshifting to produce the polyproteins pp1a and pp1ab, which are co-translationally proteolytically processed into the 15 non-structural proteins (nsp1–nsp10 and nsp12–nsp16) that make up the replication-transcription complex (RTC). The RTC is involved in the replication of genomic RNA and the transcription of a series of nested subgenomic mRNAs that are essential for the expression of structural and accessory protein genes. New virions are formed by budding into the intracellular membranes of the ER–Golgi intermediate compartment membranes and then being released via exocytosis. In addition, blue denotes the extensive host-based therapy choices, and pink denotes specific viral-based treatment possibilities [30].
Figure 2. The replication cycle of SARS-CoV-2 and its inhibitors. The binding of the spike (S) protein to the host cell receptor initiates SARS-CoV-2 infection. SARS-CoV-2 has so far been linked to two cellular receptors: angiotensin-converting enzyme 2 (ACE2) and CD147. The cleavage of the S protein by the cell surface-associated enzyme occurs after receptor engagement. The viral genomic RNA is translated through ribosomal frameshifting to produce the polyproteins pp1a and pp1ab, which are co-translationally proteolytically processed into the 15 non-structural proteins (nsp1–nsp10 and nsp12–nsp16) that make up the replication-transcription complex (RTC). The RTC is involved in the replication of genomic RNA and the transcription of a series of nested subgenomic mRNAs that are essential for the expression of structural and accessory protein genes. New virions are formed by budding into the intracellular membranes of the ER–Golgi intermediate compartment membranes and then being released via exocytosis. In addition, blue denotes the extensive host-based therapy choices, and pink denotes specific viral-based treatment possibilities [30].
Molecules 26 03868 g002
Figure 3. Antioxidant properties of Aloe vera.
Figure 3. Antioxidant properties of Aloe vera.
Molecules 26 03868 g003
Figure 4. Plant molecular pharming products in different plants.
Figure 4. Plant molecular pharming products in different plants.
Molecules 26 03868 g004
Figure 5. Hairy root cultures used to produce recombinant proteins. The final products of hairy root systems have been used to produce cytokines, vaccines, antibodies, enzymes, and other therapeutic proteins. These systems have various pros and cons, and these points should be considered when attempting to choose the best platforms.
Figure 5. Hairy root cultures used to produce recombinant proteins. The final products of hairy root systems have been used to produce cytokines, vaccines, antibodies, enzymes, and other therapeutic proteins. These systems have various pros and cons, and these points should be considered when attempting to choose the best platforms.
Molecules 26 03868 g005
Table 1. Example of biological active agents extracted from medicinal plants against SARS-CoV and other virus infections.
Table 1. Example of biological active agents extracted from medicinal plants against SARS-CoV and other virus infections.
Plant Species or Plant OrganCommon NameActive AgainstMode of ActionCompound(s) Isolated/TargetReference
Torreya nuciferaConiferous treeSARS-CoV3CLpro inhibitorPlant extract[55]
Thuja orientalisOriental ArborvitaeSARS-CoVViral growth inhibitorPlant extract[35]
Angelica keiskeiTomorrow’s leafSARS-CoV3CLpro inhibitorChalcones[56]
Radix glycyrrhizaeLicorice rootSARS-CoVInhibits viral replicationGlycyrrhizin[57]
Dioscoreae RhizomaYam RhizomeSARS-CoVViral growth inhibitorPlant extract[58]
Psoralea corylifoliaBabchiSARS-CoVPLpro inhibitorPlant extract[59]
Allium sativumGarlicHCMVInhibits viral replication in earlier stages of viral cycle before viral DNA synthesisAllitridin[60]
Myrica fayaFire treeSARS-CoVHelicase inhibitorMyricetin[61]
Laggera
pterodonta
Curly BlumeaEV71Inhibits viral RNA replicationChrysosplenetin and penduletin[62]
Cassiae semenn/aSARS-CoVViral growth inhibitorPlant extract[58]
Cryptomeria japonicaSugiSARS-CoVViral
growth
inhibitor
Hydroxy-deoxycryptojaponol[63]
Triterygium regeliiRegel’s threewingnutSARS-CoV3CLpro inhibitorCelastrol
Pristimerin
Tingenone
Iguesterin
[55]
Gentianae RadixGentian RootSARS-CoVViral growth inhibitorPlant extract[58]
Pterocarpus santalinusRed sandalwoodSARS-CoV3CLpro inhibitorSavinin[63]
Betula pubescensDowny birchSARS-CoV3CLpro inhibitorBetulinic acid[63]
Fructus arctiiBurdockFluInhibits viral replicationArctigenin[64]
Galla chinensisNutgall treeSARS-CoVViral spike protein and
Human ACE2 receptors
inhibitor
Tetra-O-galloyl-
β-d-glucose
[65]
Saxifraga melanocentra
Rhodiola kirilowii
Saxifrages
Golden root
HCVInhibits viral NS3 serine proteasePolyphenolic compounds
(−)-Epicatechin-3-O-gallate, 3,30-digalloylproprodelphinidin B2, 3,30-digalloylprocyanidin B2, and (−)-epigallocatechin-3-O-gallate
[66,67]
Paulownia
tomentosa
Princess treeSARS-CoVPLpro
inhibitor
Flavonoids[67]
Radix scutellariae
Salvia miltiorrhiza
Ranunculus sieboldii
Ranunculus sceleratus
Radix sophorae
Radix bupleuri
Huang Qin
Red sage
Buttercup
Celery-leaved buttercup
Shrubby sophora
Thorowax Root
HBVInhibits viral DNA polymerase
Inhibits viral replication
Inhibits viral replication
Inhibits viral replication
Inhibits viral DNA replication and
HBeAg production
Wogonin
Protocatechuic aldehyde/Tanshinones
Apigenin 40-O-a-rhamnopyranoside,
apigenin 7-O-b-glucopyranosyl-40-O-a-rhamnopyranoside,
tricin 7-O-b-glucopyranoside, tricin,
isoscopoletin
Apigenin 4′-O-α-rhamnopyranoside,
apigenin 7-O-β-glucopyranosyl-4′-O-a-rhamnopyranoside,
tricin 7-O-β-glucopyranoside, tricin,
isoscopoletin
Saikosaponin C
[16,68,69,70]
Scutellaria laterifloraBlue skullcapSARS-CoVHelicase inhibitorScutellarein[71]
Loranthi ramusMulberrySARS-CoVViral growth inhibitorPlant extract[58]
Stephania cepharanthan/aSARS-CoVViral growth inhibitorBiscoclaurine[35]
Cinnamomum cassiaChinese cassiaSARS-CoV3CLpro inhibitorPlant extract[71]
Linum usitatissimumFlaxSARS-CoV3CLpro inhibitorHerbacetin[55]
Alnus japonicaEast Asian alderSARS-CoVPLpro inhibitorPlant extract[72]
Laurus nobilisBay laurel.SARS-CoVViral growth inhibitorPlant Extract[35]
Rhizoma coptidis
Chrysanthemum
Morifolium
Vatica cinerea
Aesculus chinensis
Kadsura matsudai
Huang Lian
Florist’s daisy
Resak Laut
Chinese horse chestnut
Kadsura
HIV/SARS-CoVInhibits viral DNA synthesis/3CLpro inhibitor
Inhibits viral integrase
Inhibits viral replication
Inhibits viral protease
Inhibits viral replication
Berberine
Apigenin-7-O-b-D-g-lucopyranoside
Vaticinone (23E)-27-nor-3-hydroxycycloart-23-en-25-one
Triterpenoid saponins
Schizanrin B, C, D, and E
[73,74,75,76,77]
Chamaecyparis obtuse
Euphorbia jolkini
Limonium sinense
Ranunculus sieboldii
Ranunculus sceleratus
Limonium sinense
Hinoki cypress
Spurge
Girard
Buttercup
Celery-leaved buttercup
Girard
HSVInhibits HSV-1 ICP0, ICP4 expression,
and as viral DNA synthesis
Affects the late stage of HSV-2
Replication
Inhibits viral replication
Inhibits viral replication
Inhibits viral replication
Yatein
Putranjivain A
Samarangenin B
Protocatechuyl aldehyde
Isodihydrosyringetin, (−)-epigallocatechin 3-O-gallate,
samarangenin B, myricetin, myricetin 3-α-arhamnopyranoside,
quercetin 3-O-α-rhamnopyranoside, (−)-epigallocatechin,
gallic acid, N-trans-caffeoyltyramine, N-transferuloyltyramine
[16,78,79,80,81]
Azadirachta indicaNeem treeDengue virusn/aLeaf extract (Aqueous)
inhibits DEN-2 in vivo
[82]
Moringa oleiferaHorseradish treeHIV/Epstein-Barr virus
(EBV)
n/aLeaves used to inhibit
viral replication/leaves and seeds inhibits
activity against EBV
activation
[83,84]
Terminalia bellericaMyrobalanHIV-1
Pseudo viruses
n/aPlant extract against
HIV-1
[85]
Rheum palmatumChinese rhubarbSARS-CoVViral spike protein andEmodin[86]
Avicennia marinaGrey mangroveHepatitis B virusInhibits HBV antigenn/a[87]
Litchi chinensislitchiSARS-CoV3CLpro inhibitorFlavonoids extract[52]
Multiflora TuberTuber fleeceflowerSARS-CoVViral spike protein and
Human ACE2 receptors inhibitor
Emodin[86]
Canthium coromandelicumAlstonHIV-Leaf extract controls HIV infection[82]
Houttuynia cordataFish mintSARS-CoV3CLpro inhibitor
and RNA-dependent
RNA polymerase (RdRp) inhibitor
Plant extract[88]
Veronica linariifoliaSpeedwellSARS-CoVViral growth inhibitorLuteolin[62]
Carissa edulisConkerberryHerpes simplex virusExhibits anti-HSV-1 and -2 properties in vitro and in vivo stronglyn/a[89]
Nicotiana benthamianaBenthSARS-CoVViral growth inhibitorNICTABA Lectin[90]
Urtica dioicaCommon nettleSARS-CoVViral spike protein inhibitorUrtica dioica agglutinin[91]
Phyllanthus amarusIndian gooseberryHuman
immunodeficiency virus/hepatitis B virus
Inhibits HIV replicationPlant extract had lost
HBV antigen surface
[92]
Guazuma ulmifolia LamWest Indian elmPolio virusExtracts inhibits polio
replications
n/a[93]
Achyranthus asperaChaff-flowerHerpes
simplex virus
Inhibits earlier stages of
HSV multiplications
n/a[94]
CamelliasinensisTea treeSARS-CoV3CLpro inhibitorTannic acid/3-isotheaflavin-3-gallate[95]
Sesbania grandifloraVegetable hummingbirdHerpes simplex virusn/aExtract possesses strong
antiviral activity against HSV
[96]
Ficus religiosaBo treeHuman rhino virus
(HRV) and Respiratory
syncytial virus (RSV)
n/aBark extract endowed
with antivirus activity
against HRV and RSV
[97]
Hippophae rhamnoidesDengue virusSignificant anti-dengue
activity
Leaf extract[98]
Glycine max(black)SoybeanHuman adenovirus
(type 1)
Inhibits human ADV-1 in
dose-dependent manner
n/a[99]
Acacia niloticaGum arabic treeHIV-PRInhibitionn/a[100]
Allium sativumGarlicSARSProteolytic and hemagglutinating activity and
viral replication
n/a[101]
Andrographispaniculata
Boerhaavia diffusa
Green chireta
Punarnava
SARS-COV and likely SARS-CoV-2Suppression
Inhibition
NLRP3, capase-1, and IL-1β
ACE
[31,102,103]
Clerodendrum inermeThe glory bowerSARS-CoV-2InactivationRibosome[104]
Clitoria ternateaButterfly pean/aMetalloproteinase inhibitorADAM17[105]
Coriandrum sativumCoriandern/aInhibitionACE[106]
Cynara scolymus
Cassia occidentalis
Scolymusn/aInhibitionACE[102,103]
Embelia ribesWhite-flowered Embelian/aInhibitionACE[102,103]
Eugenia jambolanaBlack Plumn/aInhibitionProtease[49]
Euphorbia granulataAsthma-plantHIV-1 PRInhibition-[100]
Glycyrrhiza glabra
Gymnema sylvestre
Licorice
Gurmar
SARS; HIV-1Inhibition of viral replication; modulation of membrane fluidity
Inhibition of viral DNA synthesis
Glycyrrhizin[57,107]
Hyoscyamus nigerblack henbanen/aInhibition and BronchodilatorCa2+[108]
Ocimum
kilimandscharicum
Camphor basilHIV-1Inhibitionn/a[51]
Ocimum sanctumHoly basilHIV-1Inhibitionn/a[50]
Punica granatum PomegranateHuman herpes virus-3InhibitionACE/Phytochemical extract
exhibits potential antiviral activity
[102,103]
Salacia oblongaOblong leaf salacian/aSuppressionAngiotensin II and AT1[109]
Sambucus ebulusDanewortEnveloped virusInhibitionn/a[110]
Solanum nigrumEuropean black nightshadeHIV-1n/an/a[52]
Sphaeranthus indicusEast Indian globeMouse corona virus and
Herpes virus
Inhibitionn/a[111,112]
Strobilanthes callosaPlietesialsHCoV-NL63Blockingn/a[107,113]
Strobilanthes cusiaKuntzeHCoV-NL63Blockingn/a[113]
Vitex negundoFive-leaved chaste treeHIV-1Inhibitionn/a[52,53,113,114]
Emblica officinalisEmblicInfluenza A virusPrevention of virus adsorption and suppression of virus releasePentagalloyl glucose[114]
Vitex trifoliaSimpleleaf ChastetreeSARS-COVReductionn/a[115]
Table 4. List of commonly used medicinal herbs involved in treatment of respiratory disorders.
Table 4. List of commonly used medicinal herbs involved in treatment of respiratory disorders.
Scintific NameCommon NamesMode of ActionReferences
Tinospora cordifoliaHeart-leaved moonseedChronic fever[257,258]
Andrograhis paniculataCreat or green chiretaFever and cold
Cydonia oblongaQuinceAntioxidant, immune-modulatory, anti-allergic, smooth muscle relaxant, and anti-influenza activity
Zizyphus jujubeJujube
Cordia myxalasura
Agastya haritakiAgastya RasayanaUpper respiratory infections
Anu Thailam-Respiratory infections
Fever
Adathoda siddhaMalabar nutFever
Bryonia albaWhite bryonyReduce lung inflammation
Rhus toxicodendronEastern Poison OakViral infections
Atropa
belladonna
Deadly nightshadeAsthma and chronic lung diseases
Bignonia
sempervirens
Yellow jessamineAsthma
Eupatorium perfoliatumAgueweedRespiratory symptoms
Kabasura Kudineer-Fever, cough, sore throat, shortness of
breath
Abutilon indicumMallowImmunomodulatory function[107,259]
Withania somniferaAshwagandhaGeneral tonic and to boost immunity/against herpes simplex virus[260]
Hydrastis canadensisGoldenseal
root
Reduces plasma TNF-α, INF-γ, and
NO levels; inhibits the T helper-type 2
cytokine profile.
[261]
Acalypha indicaIndian AcalyphaAnthelmintic[262]
Achyranthes asperaPrickly chaff flowerAnti-viral activity[263]
Momordica charantiabitter melonInhibits the release of TNF-α,
NO and proliferation of spleen cells
induced by PHA and Con A.
[261]
Adhatoda vasicaAdulsaAnti-asthmatic, anti-allergic and anti-tussive activity[264]
Nigella sativaBlack cuminReduces the pancreatic ductal adenocarcinoma
cell (PDA) synthesis of monocyte
chemoattractant protein-1 (MCP-1), TNF-
α, IL-1β, and cyclooxygenase (COX)-2,
as well as inhibiting the polymorphonuclear leukocyte
functions.
[261]
Alangium salvifoliumSage-leaved alangiumAnti-rheumatoid[265]
Urtica dioicaCommon NettleReduction of TNF-α and other
inflammatory cytokines
[261]
Cassia alataCandle BushAnti-helmintic activity[107,266]
Cassia fistulaGolden ShowerAntibacterial activity[267]
Cayratia pedataBirdfoot GrapeAnti-inflammatory activity[268]
Chloroxylon swieteniaEast Indian satinwoodAnti-helmintic activity[269]
Clitoria ternatea linn.Asian pigeonwingsAnti-oxidant activity[270]
Eugenia singampattianaJungle GuavaAnti-inflammatory activity[107,268]
Hippophae rhamnoidesSea-buckthornEliminating phlegm, stopping coughing, improving digestion, and treating lung diseases[271]
Indigofera tinctoriaTrue IndigoImmunomodulatory function[107,272]
Justicia adhatodaMalabar NutAnti-oxidant and anti-mutagenic activity; hepatoprotective activity[1,236,273,274,275]
Leucas asperaThumbaiHepatoprotective activity, acaricidal properties[107,276,277]
Mucuna pruriens
Xanthium strumanium
Monkey Tamarind
Common Cocklebur
Antibacterial drugs against pneumonia[278]
Pergularia daemiaPergulariaHepatoprotective effect[107,279]
Piper longumLong pepperAnti-pneumonia drug[280]
Salacia reticulataMeharimulaAnti-inflammatory activity[268]
Santalum albumWhite SandalwoodAnti-inflammation of the lungs, blood, and pus in the sputum[271]
Solanum tornumTurkey BerryAnti-pneumonia drug[281]
Solanum suratensSurattense NightshadeAnti-viral activity[263]
Solanum xanthocarpumYellow-fruit nightshadeAnti-asthmatic[264,271]
Strychnos minorSnakewoodAnti-inflammatory activity[268]
Strychnosnux vomicaStrychnine treeAnti-inflammatory activity[268]
Syzygium aromaticumCaryophyllusHepatoprotective properties[282]
Tinospora cordifoliaMoonseedAnti-pneumonia drug[283]
Trichopus zeylanicusArogyapachaAnti-oxidant and anti-fatigue activity[284]
Tylophora indicaIndian IpecacAnti-asthmatic[264]
Verbascum thapsusGreat mulleinEnhancing peroxidase, phenolics, and antioxidant activity[1]
Vitex altissimaPeacock Chaste TreeActs against acute inflammation[285]
Vitex trifoliaHand-of-MaryTracheospasmolytic activity[107,257]
Wrightia tinctoriaPala IndigoAnti-inflammatory activity[268]
Yuthog’s BambooBambooReduction of pain and anti-inflammation of the lungs and respiratory tract[271]
Zingiber officinaleCanton GingerModulation of macrophage functions, phagocytic properties, anti-viral activity, and bronchial infections[1,286,287]
Garcinia KolaBitter KolaAnti-bacterial drug against respiratory pathogens[288]
Cymbopogon citratusWest Indian lemon grassAnti-viral infections[289]
Camellia sinensisTea plantAnti-infective activity[290]
Achillea mellefoliumYarrowProtects upper respiratory tract from viral infections[291]
Apium graveolensCeleryAnti-bacterial and anti-viral agent[292]
Borassus flabelliferPalmyra PalmProtects from pulmonary infections; anti-bacterial and anti-viral activity[293]
Caesalpinia bonducGrey NickerTreatment for asthma (anti-bacterial and anti-viral agent)[294]
Calotropis giganteaCrown flowerAnti-bacterial and anti-viral agent[293]
Crocus sativusSaffron CrocusTreatment for asthma and cough[295]
Euphorbia hirtaAsthma-PlantAnti-bacterial and anti-viral agent[291]
Piper nigrumBlack pepperAnti-viral agent[296]
Strychnos potatorumClearing-Nut TreeTreatment for bronchitis[297]
Terminalia bellirica RoxbBeleric MyrobalanEffective for asthma[298]
Tylophora indica MerrillAntamulTreatment for bronchitis and asthma[299]
Tussilago farfaraColtsfootTreatment for cough and asthma[300]
Thymus linearisHimalayan Wild ThymeAnti-viral activity[290]
Senecio chrysanthemoidesSenecioTreatment for lung diseases[301]
Portulaca oleraceaDuckweedAnti-inflammatory and anti-asthma properties[294]
Papaver somniferumOpium PoppyTreatment for respiratory diseases[293]
Morus laevigata Wall. ex BrandisWhite MulberryTreatment for cough[302]
Ephedra gerardiana Wall. ex StapfGerard’s JointfirTreatment for cough and asthma[303]
Geranium wallichianumSylvia’s SurpriseTreatment for cough[292]
Micromeria bifloraLemon Scented ThymeTreatment for cough[295]
Picrorhiza kurroa Royle ex. BenthKutkiTreatment for asthma and bronchitis[289]
Primula denticulata Sm.Drumstick PrimulaTreatment for cough and bronchitis[304]
Table 5. In silico analysis of phenolic compounds extracted form medicinal plants.
Table 5. In silico analysis of phenolic compounds extracted form medicinal plants.
Plant PartsScientific NameThe Major Phenolic CompoundFormulaActive Sites/Binding Residue/H-Bond Length (Å)
Leaves of harsingarNyctanthes arbortristisNictoflorinC27H30O15N-H--O/GLY-143/2.311
AstragalinC21H20O11O--H/PHE-140/2.197
LupeolC25H26O4N-H--O/THR-26/2.027
GiloyTinospora cordifoliaBerberine
Sitosterol
C28H18NO4
C29H50O
N-H--O/GLY-143/2.540
N-H--O/GLY-143/2.577
O--H/PHE-166/2.080
Aloe vera leavesAloe barbadensisAloenin
Aloesin
C19H22O10
C19H22O9
O--H/PHE-140/2.151
N-H--O/GLY-143/2.016
N-H--O/GLU-166/2.297
The dried ground rhizome of the turmericCurcuma longaCurcuminC21H20O6N-H--O/GLY-143/2.243
The oil of neemAzadirachta indicaNimbinC30H36O9N-H--O/GLY-143/2.161
Steroidal constituents of ashwagandhaWithanolide
Withaferin A
C28H38O6
C28H38O6
O--H/GLU-166/1.991
N-H--O/GLU-166/2.110
N-H--O/GLY-143/2.577
Constituents of pungent ketones, which result in the strong aroma of gingerZingiber officinaleGingerol
Shogaol
C17H26O4
C17H24O3
O--H/THR-190/2.026
N-H--O/GLY-143/2.289
N-H--O/THR-26/2.398
O--H/THR-24/2.345
Red onionAllium cepaQuercetinC15H10O7O--H/THR-26/1.936
Tulsi leavesOcimum sanctumUrsolic acid
Apigenin
C30H48O3
C15H10O5
N-H--O/GLY-143/2.330
O--H/THR-26/1.994
Cannabis extractsCannabis sativaCannabidiolC21H30O2N-H--O/GLY-143/2.325
Isolated from the plants of both the black and white pepper grainsPiper nigrumPiperineC17H19NO3N-H--O/THR-26/2.529
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Abiri, R.; Abdul-Hamid, H.; Sytar, O.; Abiri, R.; Bezerra de Almeida, E., Jr.; Sharma, S.K.; Bulgakov, V.P.; Arroo, R.R.J.; Malik, S. A Brief Overview of Potential Treatments for Viral Diseases Using Natural Plant Compounds: The Case of SARS-Cov. Molecules 2021, 26, 3868. https://doi.org/10.3390/molecules26133868

AMA Style

Abiri R, Abdul-Hamid H, Sytar O, Abiri R, Bezerra de Almeida E Jr., Sharma SK, Bulgakov VP, Arroo RRJ, Malik S. A Brief Overview of Potential Treatments for Viral Diseases Using Natural Plant Compounds: The Case of SARS-Cov. Molecules. 2021; 26(13):3868. https://doi.org/10.3390/molecules26133868

Chicago/Turabian Style

Abiri, Rambod, Hazandy Abdul-Hamid, Oksana Sytar, Ramin Abiri, Eduardo Bezerra de Almeida, Jr., Surender K. Sharma, Victor P. Bulgakov, Randolph R. J. Arroo, and Sonia Malik. 2021. "A Brief Overview of Potential Treatments for Viral Diseases Using Natural Plant Compounds: The Case of SARS-Cov" Molecules 26, no. 13: 3868. https://doi.org/10.3390/molecules26133868

Article Metrics

Back to TopTop