Next Article in Journal
Targeting the PI3K/AKT/mTOR Signaling Pathway in Lung Cancer: An Update Regarding Potential Drugs and Natural Products
Previous Article in Journal
Synthesis and Evaluation of Chalcone-Quinoline Based Molecular Hybrids as Potential Anti-Malarial Agents
Previous Article in Special Issue
Anti-Influenza with Green Tea Catechins: A Systematic Review and Meta-Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Plants and Natural Products with Activity against Various Types of Coronaviruses: A Review with Focus on SARS-CoV-2

by
Susana A. Llivisaca-Contreras
1,
Jaime Naranjo-Morán
2,
Andrea Pino-Acosta
3,
Luc Pieters
4,
Wim Vanden Berghe
4,5,
Patricia Manzano
2,6,
Jeffrey Vargas-Pérez
2,
Fabian León-Tamariz
5,7,* and
Juan M. Cevallos-Cevallos
2,5,6,*
1
Escuela Superior Politécnica del Litoral, ESPOL, Centro de Agua y Desarrollo Sustentable, CADS, Campus Gustavo Galindo Km. 30.5 vía Perimetral, Guayaquil EC090112, Ecuador
2
Escuela Superior Politécnica del Litoral, ESPOL, Centro de Investigaciones Biotecnológicas del Ecuador, CIBE, Campus Gustavo Galindo Km. 30.5 vía Perimetral, Guayaquil EC090112, Ecuador
3
Escuela Superior Politécnica del Litoral, ESPOL, Facultad de Arte, Diseño y Comunicación Audiovisual, FADCOM, Bosque Protector La Prosperina, Campus Gustavo Galindo Km. 30.5 vía Perimetral, Guayaquil EC090112, Ecuador
4
Natural Products & Food Research and Analysis (NatuRA), Department of Pharmaceutical Sciences, University of Antwerp, 2610 Antwerp, Belgium
5
Red Universitaria Para la Investigación y Posgrados Vlir Network, Km. 30.5 vía Perimetral, Edificio Principal, Vicerrectorado Académico (2º Piso), Guayaquil EC090112, Ecuador
6
Escuela Superior Politécnica del Litoral, ESPOL, Facultad de Ciencias de la Vida, FCV, Centro de Investigaciones Biotecnológicas del Ecuador, CIBE, Campus Gustavo Galindo Km. 30.5 vía Perimetral, Guayaquil EC090608, Ecuador
7
Grupo de Plantas Medicinales y Productos Naturales, Departamento de Biociencias, Facultad de Ciencias Químicas, Universidad de Cuenca, Av. 12 de Abril, Cuenca EC010107, Ecuador
*
Authors to whom correspondence should be addressed.
Molecules 2021, 26(13), 4099; https://doi.org/10.3390/molecules26134099
Submission received: 22 May 2021 / Revised: 14 June 2021 / Accepted: 15 June 2021 / Published: 5 July 2021
(This article belongs to the Special Issue Antiviral Properties of Natural Products)

Abstract

:
COVID-19 is a pandemic disease caused by the SARS-CoV-2 virus, which is potentially fatal for vulnerable individuals. Disease management represents a challenge for many countries, given the shortage of medicines and hospital resources. The objective of this work was to review the medicinal plants, foods and natural products showing scientific evidence for host protection against various types of coronaviruses, with a focus on SARS-CoV-2. Natural products that mitigate the symptoms caused by various coronaviruses are also presented. Particular attention was placed on natural products that stabilize the Renin–Angiotensin–Aldosterone System (RAAS), which has been associated with the entry of the SARS-CoV-2 into human cells.

1. Introduction

The Coronavirus Disease 2019 (COVID-19), caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), was declared a pandemic on 11 March 2020 [1] and is probably the biggest challenge for public health systems in most countries given the limited knowledge about effective treatments [2].
The SARS-CoV-2 belongs to the Coronaviridae family and the Coronavirinae subfamily which has been divided into four genera: α-coronavirus, β-coronavirus, γ-coronavirus and δ-coronavirus [3]. The Human Coronavirus species HCoV (OC43, 229E, NL63 and HKU1), as well as those associated with Severe Acute Respiratory Syndrome (SARS), Middle East Respiratory Syndrome (MERS), and SARS-CoV-2, can cause respiratory tract infection but others such as the species 229E, OC43, HKU1, and NL63 usually cause the common cold [3]. Genetic characterization has shown that SARS-CoV-2 shares almost 80% of the SARS-CoV [4] and 96.2% of the bat β-coronaviruses lineage B [1] genomes. The SARS-CoV-2 belongs to the β-coronavirus group and causes milder symptoms than SARS and MERS but the transmission between people is much faster with an R0 (Basic Reproduction Number) of 3.28 [5] compared to the R0 values around 0.9 for MERS-CoV [2]. The mortality rate for SARS-CoV-2 is 3.4% compared to 9.6% and 35% for SARS-CoV and MERS respectively [6]. The incubation period for SARS is 2 to 10 days, while that of SARS-CoV-2 is 1 to 14 days (Table 1) [4]. Additionally, several studies reported that SARS-CoV-2 and SARS-CoV use the Angiotensin-Converting Enzyme 2 (ACE2) as a receptor to enter target cells, while MERS-CoV uses dipeptidyl peptidase 4 (DPP4) for the same purpose (Table 1) [2]. The alveolar lung and small intestine are potential targets for SARS-CoV-2 due to the high expression of ACE2 [1].
SARS-CoV-2 mainly affects the middle-aged and elderly, as well as people with underlying diseases such as hypertension, diabetes, obesity or with heart and kidney problems, but shows low severity in children [7] although the disease transmission in this age group is still unknown [2] and the infection rates in children are increasing with the emergence of new SARS-CoV-2 variants [9].
Home isolation and quarantine have been applied in most countries to reduce the spread of the disease. However, this measure is also leading to economic, social and political deterioration in the affected countries. Consequently, the cases of anxiety and depression due to confinement as well as the number of deaths due to these causes have increased [10]. The enormous worldwide effort to develop vaccines against COVID-19 is recognized well-known as at least 19 vaccines have entered clinical trials and some vaccines already being applied to people in several countries [11]. However, the rushed development of a vaccine is usually accompanied by numerous challenges including potentially severe side effects and the possible loss of disease protection shortly after vaccination [12]. Moreover, the rise of new virus variants can affect the effectiveness of current treatments.
Similarly, other large-scale trials are in progress for the evaluation of possible therapies, including the World Health Organization (WHO) Solidarity Trial [11]. Pharmaceutical products undergoing clinical trials as potential treatments for COVID-19 include the antiviral nucleotide analog remdesivir, systemic interferons, and monoclonal antibodies [11]. Moreover, the antiparasitic drug ivermectin has been repurposed as a potential antiviral against SARS-CoV-2 and some drugs such as hydroxychloroquine that initially seemed promising have already been discarded by conflicting results through small-scale studies [12].
The accelerated search for a cure involves questions of a bioethical nature which prompts a reflection on the Declaration of Helsinki [2013] as well as the non-maleficence and beneficence principles to enable the use of untested procedures in clinical trials under emergency conditions [13]. It is necessary to implement a sustainable program to improve the health of citizens while a cure for SARS-CoV-2 is developed. Medicinal plants and natural products have the potential for enhancing people’s health and boost the immune system [14]. Plants generally contain a combination of active ingredients or phytochemicals with different properties. Herbal medicinal formulations have been effective in treating emerging and reemerging viral diseases affecting diverse human and animal populations [14]. Plant extracts have shown specific antiviral properties in experimental animal models, which have prompted the formulation of natural products for the treatment of viral diseases [15]. Similarly, the bioactive compounds of medicinal plants can act as immunomodulators and can be combined with other therapies against viral diseases [16].
Natural products can help researchers design safe and easily accessible medical treatments [17]. For instance, plants from traditional Chinese medicine (TCM) such as Scutellaria baicalensis contain various antiviral compounds, including inhibitors of viral replication [18] and phytochemicals with anti-SARS-CoV-2 potential (Table 2). Furthermore, 125 Chinese herbs were found to contain at least 2 of 13 compounds (betulinic acid, coumaroyltyramine, cryptotanshinone, desmethoxyreserpine, dihomo-γ-linolenic acid, dihydrotanshinone I, kaempferol, lignan, moupinamide, N-cis-feruloyltyramine, quercetin, sugiol, tanshinone IIa) that can inhibit the 3C-Like protease (3CLpro) and Papain-Like protease (PLpro) as well as block the entry, replication and binding of the SARS-CoV-2 Spike protein (S protein) [19]. Similarly, a protective effect against the 229E coronavirus was observed in respiratory cell cultures pre-treated with 50 µg/mL Echinacea (Table 2) [20]. In addition, the highly pathogenic SARS and MERS coronaviruses were also inactivated in vitro (IC50 3.2 ug/mL) using the same plant. Other species such as grapefruit (Citrus × paradisi) have also been used to combat several respiratory infections [21].
The Renin–Angiotensin–Aldosterone System (RAAS) is a cascade of vasoactive peptides that regulate key processes in human physiology. SARS-CoV-1 and SARS-CoV-2 interfere with the RAAS by binding to the Angiotensin-Converting Enzyme 2 (ACE2) which serves as a receptor for both SARS viruses [66]. Overactivation of the RAAS by coronaviruses can contribute to the development of critical symptoms. Several common foods belonging to the families Alliaceae, Apiaceae, Brassicaceae, Cucurbitaceae, Rutaceae, Vitaceae, Zingiberaceae, among others have demonstrated the ability to regulate key RAAS processes [38,60] (Table 3).
Various countries such as Ecuador are considered megadiverse because of the high number of plant species. Various species from megadiverse areas have shown great potential for the treatment of respiratory conditions but have not been tested against coronaviruses (Table 4) [66]. Further research is needed to assess the effect of these species against SARS-CoV-2. The pandemic impact of the 2002 SARS epidemic that began in Foshan, China [38,67], the high mortality rate and the subsequent re-emergence of the disease one year later [60] together with the economic problems caused in Asia encouraged research efforts focused on controlling coronaviruses infections by medicinal plants [68]. The aim of this review was to summarize the available literature on medicinal plants used against various types of coronaviruses, including SARS CoV-2 [67]. Special emphasis was placed on species located in Ecuador as one of the megadiverse countries.

2. Methods

Literature Search

The PubMed, NCBI, Elsevier databases were used for searching natural compounds and medicinal plants with pharmacological activity against the SARS, MERS or SARS-CoV coronaviruses. Keywords like coronavirus; COVID-19; medicinal plants; active principle; natural compounds; inhibitor; SARS; MERS or SARS-CoV-2; Spike protein; RAAS; Angiotensin-Converting-Enzyme Inhibitors (ACEi); Angiotensin Receptor Blocker (ARB) were used to carry out the search. In addition, studies published since 2002 were reviewed, as this was the year in which SARS was reported for the first time [73]. The common name of the studied plants was determined with the help of an expert botanist, using the references “Plantas Útiles de Litoral Ecuatoriano de Flor María Valverde Vadillo” [74] and the “Enciclopedia de las Plantas Útiles del Ecuador”, and the databases “Herbario Rapid Reference” (https://plantidtools.fieldmuseum.org/es/rrc/5581) Date accessed: 19 April 2019 and “Trópicos” (https://www.tropicos.org/home) Date accessed: 14 May 2019.

3. Pathogenesis of SARS-CoV-2

SARS-CoV-2 relies on its S protein to attach to human cells having an ACE2 receptor. Studies have shown that SARS-CoV-2 has a higher ACE2 binding affinity than SARS-CoV, supporting an efficient cell entry [75]. The S protein from SARS-CoV-2 consists of subunits S1 and S2. While S1 is important for the virus attachment to the ACE2 receptor, S2 allows the fusion of the virus and cell membranes followed by the internalization of the viral genetic material. Therefore, after attachment to the ACE2 receptor, the S protein needs to be primed at the S1–S2 site by cellular proteases such as the Transmembrane Serine protease 2 (TMPRSS2) [35]. Therefore, the virus is capable of infecting human cells containing both ACE2 receptors and proteases such as the TMPRSS2, including lungs, small intestine, heart and kidney cells, as well as the nose, nasopharynx and oral mucosa [35]. Once inside the cell, the viral genetic material undergoes replication, synthesis of the S protein as well as other polyproteins. Figure 1 shows the infection process of SARS-CoV-2 in human cells.
The synthetized polyproteins are then processed by a 3C-like protease (3CLpro) also known as the main protease (MPro) and a PLpro to produce 16 nonstructural proteins (Nsp), including the Nsp13 helicase, responsible for the replication and transcription of the viral genome [18]. After cell entry and multiplication, the virus can cause inflammatory responses in the host attributed to an excessive release of cytokines. This cytokine storm has been associated with severe damage to the lungs, blood hypercoagulation, cardiac arrest and lymphocytopenia among other life-threatening conditions [76].

3.1. The Renin–Angiotensin–Aldosterone System as Affected by SARS-CoV-2

During the SARS-CoV-2 infection, the virus sequesters ACE2 causing the instability of the RAAS and contributing to various symptoms of COVID-19.
A stressed organism is usually more predisposed to infections by microorganisms [73]. Frequent or very strong episodes of stress caused by an overactivated RAAS include an excessive conversion of Angiotensin I (AI) into Angiotensin II (AII) by the ACE [77]. AII binds to the Angiotensin II Type I Receptor (AT1R), causing instability of blood pressure [78] as well as cardiovascular, renal [79] and prothrombotic issues [80]; myocardial dysfunction [81]; altered activity of the sympathetic nervous system [82]; and chronic hypertension in obese individuals [83]. AII is considered a cytokine with pro-inflammatory properties and the accumulation of this molecule can induce chemotaxis, contributing to a storm of cytokines [81,84]. To regulate the over-activated RAAS, ACE2 inactivates AII generating the harmless heptapeptide Angiotensin 1–7 (A1-7) with a powerful vasodilator function [85]. However, SARS-CoV-2 disrupts this mechanism after hijacking ACE2, causing the accumulation of AII and contributing to various symptoms of COVID-19 [66]. Therefore, the over-activation of RAAS should be prevented to reduce the severity of the infection [86]. Specific foods and plants that modulate the RAAS [60] can prevent the coronavirus entry or alleviate the COVID-19 symptoms. Figure 2 shows the effect of SARS-CoV-2 on the RAAS.

3.2. Immune System Boosting Plants and Foods

At present, different herbal plants are being subjected to studies on their ability to strengthen the immune system and cope up with the virus and some phytocompounds have already shown potential to mitigate the incidence of infection [87]. For instance, various plant polyphenols can initiate a cellular accumulation to then trigger signaling pathways and immune responses to infection. In addition, polyphenols are potent inhibitors of the COVID-19 protease (Mpro) [87].
Natural polysaccharides and terpenoids are immunomodulatory as well as adaptogenic compounds and are also recognized for their antiviral, immunomodulatory, antitumor and anticoagulant bioactivities. Similarly, giloy herbs can stimulate IgG antibody response, macrophage activation, induction of cell-regulated immunity, and humoral immunity [87]. Moreover, several plant triterpenes such as dammaradienol, dammarenediol-II, hydroxyhopanone. dammarenolic acid, hydroxymarenone-I, ursonic acid, shoic acid, eichlerianic acid and hydroxyoleanonic lactone [87] play a vital role in the modulation of cellular metabolism [88].
Sulfated polysaccharides are a structurally multifaceted class of biomolecules with diverse physicochemical characteristics well recognized in the field of medicine and pharmaceutical sciences [29]. They have immunomodulatory properties and bioactivities [89]. Furthermore, they are selective inhibitors or suppressors of enveloped viruses, e.g., HSV, HIV, human cytomegalovirus, respiratory syncytial virus, and influenza [89].
The biomolecules hispidin, lepidine E, and folic acid from Citrus sp. inhibit the 3CL hydrolase enzyme known to counteract the host’s innate immune response [90]. Similarly, Benzene 123 Triol from Nilavembu kudineer has shown immunomodulatory activity [91] while Exocarpium Citri grandis (Flavonoids and Naringin) stimulated the antiviral immune response and showed antitussive, expectorant and helped relieve pulmonary fibrosis [89]. Moreover, Allium sativum (Allicin) stimulated the activity of immune cells and inhibited the release of pro-inflammatory cytokines dependent on Necrosis Tumoral Factor alfa (TNFα) as well as the migration of neutrophilic granulocytes, a crucial process during inflammation [46]. The plant species Acacia senegal, Laportea aestuans, and Citrus spp (Hesperidin) increased antioxidant defenses, modulated the activity of the immune system, and eliminated reactive oxygen species. In addition, Curcuma longa (Curcumin) also enhanced immunity [46].
Foods containing curcumin, allicin, papain, ginsenoside, mangoosteen, chloroquine, etc., have shown a direct effect on dendritic cells, natural killer cells (NK), lymphocytes and antibodies to protect the human body from foreign particles [89].

4. Bioactive Compounds in the Mechanisms of the Virus–Host Interaction

Table 2 shows the plant species with activity against various coronaviruses.

4.1. Entry Inhibitors

Many plant bioactive compounds typically prevent the entrance of the viral particle into the host cell [87]. SARS-CoV entry inhibitors are divided into two categories: the first consists of molecules that bind to the ACE2 and TMPRSS2 receptors while the second comprises compounds that bind to the virus and prevent interaction with the cell receptors and membrane fusion [92]. The molecule [6] gingerol from Zingiber officinale inhibits the growth of the coronavirus by blocking the cell’s TMPRSS2 receptor [21].
The TCM’s Jinchai consists of plant species such as Lonicera japonica and Bupleurum chinense among others, that prevent the coronavirus entry into cells and inhibit general viral replication as well as the specific 3CLpro-mediated replication [29]. One of the main active components of Jinchai is baicalin which inhibited antiviral activity with an Effective Concentration (EC50) of 12-50 µg/mL in SARS-CoV-infected fetal rhesus monkey kidney cell line (fRHK4) and EC50 of 100 µg/mL in Vero-E6 cells [93].
Flavonoids stand out among the blockers of the ACE2 receptor, but they have also shown anti-replication activities. Similarly, compounds such as baicalin, epigallocatechin gallate, gallocatechin gallate, derivatives of kaempferol, myricetin, quercetin and scutellarein are other major constituents of TCM used to treat SARS by inhibiting the entry and replication of the virus [64].
The flavonoid hesperetin has the potential to inhibit ACE2 and block SARS-CoV-2 infection by binding to viralS protein, helicase, and protease sites of the ACE2 receptor [29].
Alternatively, computational analysis revealed that hesperidin, baicalin and kaempferol 3-O-rutinoside can block SARS-CoV-2 infection by weakening the adsorption of virus to cells [19,46]. Similarly, procyanidins and the butanol extract of Cinnamomi Cortex (bark of Cinnamomum verum) have shown antiviral effects at the RNA level, in addition to inhibiting SARS-CoV infection with an IC50 of 29.9 ± 3.3 μM (Table 2 and Table 3) [30]. Additionally, cinnamon extract inhibited wild-type SARS-CoV infection in vitro with an IC50 of 43 μM and blockage of the virus entry to the cell was suggested as the possible mechanism of action [32]. The polyphenol epigallocatechin gallate (EGCG) from Camellia sinensis (green tea) inhibited the spread of the bovine coronavirus and interfered with the viral adsorption to bovine kidney cells [94].
Among the virus-binding molecules, lectins have emerged as a new class of antivirals thanks to their ability to bind to the glycosylated molecules found on the surface of viruses such as the SARS-CoV spike glycoprotein [24]. One of the most potent molecules reported against SARS-CoV is the mannose-binding lectin isolated from leek (Allium porrum L.), with an EC50 of 0.45 μg/mL and a selectivity index >222 (Table 2 and Table 3) [29]. Specific N-acetylglucosamine lectins obtained from tobacco (Nicotiana tabacum L.) and stinging nettle (Urtica dioica L.) were also active against SARS-CoV with selectivity indexes of >77 and >59, respectively [24]. Additionally, the mannose-specific lectin from Hippeastrum striatum (Lam.) has the potential to inhibit the final step of the virus infection cycle [24,87]. Similarly, triterpenoids such as glycyrrhizin from the licorice plant, Glycyrrhiza glabra L., have been reported to have in vitro anti-SARS effects with an EC50 of 300 µg/mL [51]. These natural compounds interfere with virus–host fusion steps through the envelope of the predominant heptad repeat 2 domains in viral envelopes [89].
Emodin is a natural anthraquinone derivative and an active ingredient of medicinal plants such as rhubarb (Genus Rheum) (Table 2 and Table 3), Polygonum cuspidatum, Aloe vera, Senna obtusifolia [59] and Cassia tora L [28]. Emodin blocked SARS-CoV entry to host cells by binding to the S proteins and interfering with the 3CLpro activity of the virus, thus preventing the formation of the Nsp required for replication [27]. In trials involving SARS-CoV and OC43, emodin significantly blocked, in a dose-dependent manner, the interaction between SARS-CoV S protein and ACE2, inhibited the ion channel 3a and interrupted the release of new coronaviruses [22]. Similarly, terpenoids from medicinal plants exhibit general antiviral effects in vitro against SARS-CoV [29]. Oleanane-type saikosaponins found in medicinal plants such as Bupleurum spp. and Heteromorpha spp. prevented the entry of SARS-CoV into the cell [46].

4.2. Protease Inhibitors

Proteases are key players in the pathogenesis caused by SARS-CoV and SARS-CoV-2 as they are involved in the S protein activation and viral replication. Therefore, protease inhibitors can aid the COVID-19 treatment. Because of the good binding affinity for Mpro and S protein of eugenol and curcumin, these compounds can be considered promising anti-SARS-CoV agents [22,95]. Curcumin inhibited SARS-CoV 3CLpro with an IC50 value of 23.5 μM [22,92]. Similarly, various phenolic tea constituents, such as tannic acid, 3-isotheaflavin-3-gallate and theaflavin-3,3-digallate (Table 2 and Table 3) also inhibit SARS-CoV 3CLpro with IC50 values between 3, 7 and 9.5 μM, respectively [37]. Similarly, a cell-based study showed that sinigrin significantly blocked the cleavage process of 3CLpro with an IC50 of 752 μM. Sinigrin is a glucosinolate found in some plants of the Brassicaceae family, such as Brussels sprouts, broccoli, and black mustard seeds [29] (Table 2 and Table 3).
Scutellaria baicalensis polysaccharides, polyphenols and polyglycans can inhibit immune regulation and have shown antioxidant and antiviral activity [57]. The flavonoids scutellarein and baicalin from the same species inhibited SARS-CoV Nsp13 helicase [56], while myricetin reached an IC50 of 2.71 μM against the virus [61]. These two compounds potently inhibited Nsp13 in vitro by affecting the ATPase activity of SARS-CoV [57].

4.3. Replication Inhibitors

Inhibitors of viral replication are amongst the key molecules to fight coronavirus diseases. The phenolic compounds from Melia azedarach (cinamomo or chinaberry tree) and Camellia sinensis (green tea) have shown antiviral activity due to the inhibition of RNA polymerase or the RNA-dependent proteases involved in the replication of the coronavirus RNA [29]. Additionally, tea extracts can also affect the virus assembly and release [96]. Similarly, the consecutive application of stilbene derivatives such as resveratrol at 62.5 μM partially mitigated MERS-CoV-induced cell death and reduced the replication of infectious MERS-CoV by 10-fold [63]. Similarly, concentrations below 0.5 mg/mL of stilbene derivatives like resveratrol inhibited the replication of SARS-CoV in vitro [62]. These compounds are found in different plants, including the Vitis vinifera L. grape and berries of the genus Vaccinium (Table 2 and Table 3) [30]. Compounds in berries have been suggested to block the virus entry to cells through endocytosis [97].
In general, natural flavonoids such as quercetin, catechin, naringenin and hesperetin are the most abundant polyphenols in the human diet, as they are found in fruits and vegetables as glycosides or acylglycosides [95]. Naringenin exhibited a partial inhibition of SARS-CoV-2 replication observed at 24 h post-infection (hpi) in cells upon Two-pore channel 2 (TPC2) silencing while stronger inhibition was observed at 48 and 72 hpi [36].
The standardized extract of Pelargonium sidoides (EPS 7630), mainly containing polyphenolic compounds such as prodelphinidin, gallocatechin and its stereoisomer epigallocatechin [53,98], is an approved treatment for acute bronchitis in Germany and other countries [53]. Concentrations up to 100 μg/mL of EPS 7630 interfered with the replication of human coronavirus as well as the seasonal influenza A virus Hemagglutinin Type 1 and Neuraminidase Type 1 (H1N1, H3N2), respiratory syncytial virus, parainfluenza virus and coxsackie virus [52] and inhibited the entry and replication of 229E with EC50 of 44.50 ± 15.84 μg/mL [99].
The essential oils of Laurus nobilis and Salvia officinalis have also shown significant anti-replication activity against SARS-CoV with an Inhibitory Concentration (IC50) value of 120 μg/mL [58]. Similarly, the essential oils from Thuja orientalis (β-ocimene, 1,8-cineole, α-pinene and β-pinene) also inhibited SARS-CoV replication [58] and the aescin isolated from the horse chestnut tree also inhibited SARS-CoV replication at non-toxic concentrations [22,100].

4.4. Virucidal Activity

The inactivation of the viral particles is another strategy to combat respiratory diseases. Echinacea purpurea extracts available as the commercial product Echinaforce® showed dose-dependent inhibition of 229E infectivity in respiratory epithelial cells and this extract irreversibly inactivated the virus with an IC50 of 3.2 μg/mL [101] and 9 ± 3 μg/mL in another study [76]. The multicomponent extract non-specifically and irreversibly interfered with viral docking receptors to block the infectivity of pathogens [102]. Similarly, inhibition for MERS-CoV was observed with 10 μg/mL of Echinaforce®, reducing viral infectivity by 99.9% at 50 μg/mL [41]. Combining E. purpurea with vitamin D, vitamin C, and zinc has been suggested to reduce the risk of infection and death from SARS-CoV-2 [103]. A scientific review concluded that along with vitamin D, vitamin C and zinc, Echinacea extracts are pivotal in terms of prevention and treatment (shortening the duration and/or lessening the severity of symptoms) of common colds [104].

4.5. Immunomodulatory Agents

Generally, the viral loads observed in patients correlate with the severity of symptoms and mortality. The multisystem inflammatory syndrome, known as cytokine storm, occurring in many COVID-19 patients, is caused by an uncontrolled replication of the virus resulting in an over-activation of the immune system, including high levels of pro-inflammatory cytokines, i.e., interleukin-1β (IL-1β) and TNFα [105]. The geranylated flavonoid tomentin E from Paulownia tomentosa inhibited SARS-CoV (PLpro) in a dose-dependent manner with an IC50 between 5.0 and 14.4 μM and reduced the concentration of the pro-inflammatory cytokines IL-1β and TNFα [51]. Similarly, one study observed that chlorogenic acid, luteoloside, quercetin, and other compounds in L. japonica, exhibited anti-inflammatory, antiviral, antibacterial, and antioxidant activity and enhanced immune response. It is known that one of the main possible anti-SARS mechanisms is decreasing the expression of inflammatory mediators such as the transforming growth factor-beta (TNF-β) and IL-1β [49].
Anthocyanins are found in red to violet fruits such as berries of the genus Vaccinium, blackberry, among others (Table 2 and Table 3) [106]. Anthocyanin metabolites, such as the protocatechuic acid, were shown to weakly inhibit Nitric Oxide (NO) production and TNF-α secretion in Lipopolysaccharide-Gamma interferon-induced macrophages (LPS-INF-γ) [107]. Additionally, gallic acid decreased the secretion of the inflammatory mediators monocyte chemoattractant protein 1 (MCP-1), intercellular adhesion molecule 1 (ICAM-1), and vascular cell adhesion molecule 1 (VCAM-1) in endothelial cells [83]. However, the anthocyanins concentrations used for the anti-inflammatory activity tests cannot be achieved physiologically [107]. Similarly, Echinacea has also been proposed as a suppressor of the immunoinflammatory cascades observed in COVID-19, thanks to the plant’s ability to activate the anti-inflammatory cannabinoid-2 (CB2) receptors and peroxisome proliferator-activated receptors gamma (PPARγ) [102].

4.6. Regulators of RAAS

Table 3 summarizes medicinal plants and purified bioactives with potential benefits against SARS-CoV-2, especially those that modulate the RAAS. For example, the ethanolic extract of Thymus vulgaris (thyme), among other plants has shown the inhibitory capacity of AT1R [34].
The onions tunic extract, rich in flavonols like quercetin, has been shown to be a competitive inhibitor of ACE, comparable to pure quercetin (IC50: 0.36 ± 0.04 and 0.34 ± 0.03 μg/mL respectively). This same extract further revealed competitive ACE inhibition with the substrate, N-[3-(2-furyl) acryloyl]-L-phenylalanylglycylglycine [26]. Agrawal and collegeus reported that quercetin can interfere with various stages of the coronavirus entry and replication cycle, such as PLpro, 3CLpro and nucleoside-triphosphatase (NTPase)/helicase, showing pleiotropic activities and lack of systemic toxicity [25]. Similarly, EGCG also inhibited ACE and blocked the AII binding to AT1R in vitro, showing the potential to control the symptoms of various diseases, especially those of a respiratory nature [108]. Further research is needed to assess the potential of EGCG for the treatment of symptoms caused by coronaviruses.

4.7. Unknown Mechanisms of Action

The lycorine purified from Lycoris spp. was identified as a promising anti-SARS-CoV bioactive compound with an EC50 value of 34.5 ± 2.6 μg/mL, by poorly understood mechanisms [50]. Flavonoids, benzofurans, stilbene, polyhydroxylated alkaloids, and kuwanons from Morus spp. have shown a large variety of pharmacological activities including antiviral activity but the mechanism is also unclear [68,109]. The same is through for the compounds from Ginkgo biloba (ginkgolide, terpenic lactones, flavonoids, polyphenols, oleic acid, among others) [110]. Therefore, further research is needed to resolve their antiviral mechanism(s) of action.
Recently, two naturally occurring alkaloid-derived compounds (homoharringtonine and emetine), effectively inhibited the SARS-CoV-2 in Vero E6 cells with an estimated EC50 of 2.55 μM and 0.46 μM, respectively [111]. Similarly, emetine has been reported as an inhibitor of hCoV-OC43, hCoV-NL43, SARS-CoV MERS-CoV and MHV-A59 in vitro with EC50 at the low micromolar range. However, the study did not disclose the mechanisms by which both compounds induced anti-SARS-CoV-2 activity [112]. Emetine is a natural alkaloid isolated from Psychotria ipecacuanha and belongs to the methine class of alkaloids [113]. Similarly, homoharringtonine is a natural alkaloid derived from some species of the genus Cephalotaxus. This drug is a protein synthesis inhibitor and has been approved by the Food and Drug Administration (FDA) to treat chronic myeloid leukemia [112].

5. Risks Associated with the Incorrect Use of Natural Products

Although many of the plant species hold promise to reduce or mitigate COVID-19 symptoms, it is necessary to further validate their potential health benefits with clinical trials as well as to identify potential side effects. Despite the reported health benefits, high doses of ginkgo (Ginkgo biloba) [Table 2] can cause an increase in cerebral blood flow, and affect people with peptic ulcer and coagulation disorders [114,115].
Although no adverse effects have been reported in the consumption of ginger ([6]-gingerol) (Table 2), irritation of the gastric mucosa has sometimes been mentioned. Similarly, turmeric should not be used in case of infections or inflammation of the hepato-bile duct or jaundice [115] and only the stem of rhubarb (Table 3) can be ingested as the leaves contain a large amount of oxalic acid that causes kidney stones [30]. Moreover, the excessive use of Aloe species (Table 2) can cause damage to the epithelium and the intestinal mucosa, hemorrhagic diarrhea, and kidney damage. Doses greater than 1 g/day are not recommended for pregnant women, women during menstruating periods or people suffering from kidney disease [115]. Consequently, medical observation is recommended for people who have never consumed any of the plants mentioned in this work. People must be properly informed of the contraindications before combining medicinal plants with any treatment against the symptoms of COVID-19 [116] in order to avoid a counterproductive effect.

6. Conclusions

Scientific evidence of medicinal plants and foods that can help to mitigate the symptoms of COVID-19 has been growing since the start of the pandemic. Therefore, it is important to promote the consumption of natural products under the supervision of experts in the medical, nutritional and pharmaceutical areas as well as encouraging the generation of scientific information that promotes the manufacture of plant-based products that help to better protect the people against the SARS-CoV2.
The identification of the antiviral mechanisms of natural agents acting in different stages of the viral life cycle offers hope for future antiviral therapies [16]. In addition, the elucidation of the mechanism of action of natural compounds against COVID-19 will contribute to discover promising anti-COVID-19 natural drugs [108]. However, it is important to emphasize that medicines for the treatment of COVID-19 should not be replaced by untested natural products. Good practices of bioprospecting of medicinal plants should be fostered, in order to increase the interests of the ancestral people from developing countries [116]. This will allow the promotion of the development of new natural products that mitigate the symptoms of this COVID-19 without leaving the vital specialized medical treatment [32].

Author Contributions

S.A.L.-C.: Review of databases, writing of article, creation and elaboration of the figures. J.N.-M.: Review of databases and writing of article. A.P.-A.: Review of databases and design and elaboration of the figures. L.P.: Article review and corrections. W.V.B.: Article review and corrections. P.M.: Article review and corrections. J.V.-P.: Review of databasesand, design and elaboration of the figures. F.L.-T.: Article review and writing. J.M.C.-C.: Article review, writing and elaboration of the figures. All authors have read and agreed to the published version of the manuscript.

Funding

This research is part of an international agreement between the Flemish Interuniversitary Council (VLIR-UOS) and the Ecuadorian Interuniversitary Network (VLIR-Network Ecuador).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

This work was also supported by the Council of Flemish Universities of Belgium (VLIR) through.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Redeploying Plant Defences. Nat. Plants 2020, 6, 177. [CrossRef] [Green Version]
  2. Petersen, E.; Koopmans, M.; Go, U.; Hamer, D.H.; Petrosillo, N.; Castelli, F.; Storgaard, M.; Khalili, S.A.; Simonsen, L. Comparing SARS-CoV-2 with SARS-CoV and Influenza Pandemics. Lancet Infect. Dis. 2020, 20, 238–244. [Google Scholar] [CrossRef]
  3. Fani, M.; Teimoori, A.; Ghafari, S. Comparison of the COVID-2019 (SARS-CoV-2) Pathogenesis with SARS-CoV and MERS-CoV Infections. Future Virol. 2020, 15, 317–323. [Google Scholar] [CrossRef]
  4. Caldaria, A.; Conforti, C.; Di-Meo, N.; Dianzani, C.; Mohammad, J.; Torello, L.; Zalaudek, I.; Giuffrida, R. COVID-19 and SARS: Differences and Similarities. Dermatol. Ther. 2020, e13395. [Google Scholar] [CrossRef] [Green Version]
  5. Liu, Y.; Gayle, A.A.; Wilder-Smith, A.; Rocklöv, J. The Reproductive Number of COVID-19 is Higher Compared to SARS Coronavirus. J. Travel Med. 2020, 27, 1–4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Perlman, S. Another Decade, Another Coronavirus. N. Engl. J. Med. 2020, 382, 760–762. [Google Scholar] [CrossRef] [PubMed]
  7. 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] [PubMed] [Green Version]
  8. Petrosillo, N.; Viceconte, G.; Ergonul, O.; Ippolito, G.; Petersen, E. COVID-19, SARS and MERS: Are They Closely Related? Clin. Microbiol. Infec. 2020, 26, 729–734. [Google Scholar] [CrossRef]
  9. Lee, P.; Hu, Y.; Chen, P.; Huang, Y.; Hsueh, P. Are Children Less Susceptible to COVID-19? J. Microbiol. Immunol. Infect. 2020, 53, 371–372. [Google Scholar] [CrossRef]
  10. Guerra, E. Recorte a la Educación Superior: Una Medida que Ahondará la Crisis—Opción S. Revista S. 2020. Available online: https://opcions.ec/portal/2020/05/08/la-educacion-publica-y-el-recorte-presupuestario/ (accessed on 8 May 2020).
  11. EMA. Treatments and Vaccines for COVID-19. European Medicines Agency. 2020. Available online: https://www.ema.europa.eu/en/human-regulatory/overview/public-health-threats/coronavirus-disease-covid-19/treatments-vaccines-covid-19 (accessed on 8 November 2020).
  12. ECDC. Vaccines and Treatment of COVID-19. European Centre for Disease Prevention and Control. 2020. Available online: https://www.ecdc.europa.eu/en/covid-19/latest-evidence/treatment (accessed on 5 May 2021).
  13. Mastroleo, I. Post-trial Obligations in the Declaration of Helsinki 2013: Classification, Reconstruction and Interpretation. Dev. World Bioeth. 2016, 16, 80–90. [Google Scholar] [CrossRef]
  14. Dhama, K.; Karthik, K.; Khandia, R.; Munjal, A.; Tiwari, R.; Rana, R.; Khurana, S.; Khan, R.; Alagawany, M.; Farag, M. Medicinal and Therapeutic Potential of Herbs and Plant Metabolites/Extracts Countering Viral Pathogens—Current Knowledge and Future Prospects. Curr. Drug Metab. 2018, 19, 236–263. [Google Scholar] [CrossRef] [PubMed]
  15. Akram, M.; Tahir, I.; Shah, S.; Mahmood, Z.; Altaf, A.; Ahmad, K.; Munir, N.; Daniyal, M.; Nasir, S.; Mehboob, H. Antiviral Potential of Medicinal Plants against HIV, HSV, Influenza, Hepatitis, and Coxsackievirus: A Systematic Review. Phytother. Res. 2018, 32, 811–822. [Google Scholar] [CrossRef]
  16. Siddiqui, A.; Danciu, C.; Ashraf, S.; Moin, A.; Singh, R.; Alreshidi, M.; Patel, M.; Jahan, S.; Kumar, S.; Alkhinjar, M. Plants-Derived Biomolecules as Potent Antiviral Phytomedicines: New Insights on Ethnobotanical Evidences against Coronaviruses. Plants 2020, 9, 1244. [Google Scholar] [CrossRef] [PubMed]
  17. Khare, P.; Sahu, U.; Pandey, S.; Samant, M. Current Approaches for Target-Specific Drug Discovery Using Natural Compounds against SARS-CoV-2 Infection. Virus Res. 2020, 290, 198169. [Google Scholar] [CrossRef]
  18. Tahir, M.; Alqahtani, S.; Alamri, M.; Chen, L. Structural Basis of SARS-CoV-2 3CLpro and Anti-COVID-19 Drug Discovery from Medicinal Plants. J. Pharm. Anal. 2020, 1, 313–319. [Google Scholar] [CrossRef]
  19. Zhang, D.; Wu, K.; Zhang, X.; Deng, S.; Peng, B. In Silico Screening of Chinese Herbal Medicines with the Potential to Directly Inhibit 2019 Novel Coronavirus. J. Integr. Med. 2020, 18, 152–158. [Google Scholar] [CrossRef]
  20. Signer, J.; Jonsdottir, H.; Albrich, W.; Strasser, M.; Züst, R.; Ryter, S.; Ackermann, R.; Lenz, N.; Siegrist, D.; Suter, A. In Vitro Antiviral Activity of Echinaforce®, an Echinacea purpurea Preparation, against Common Cold Coronavirus 229E and Highly Pathogenic MERS-CoV and SARS-CoV. Virol. J. 2020, 10, 2. [Google Scholar]
  21. Tallei, T.; Tumilaar, S.; Niode, N.; Fatimawali, K.; Johnson, B.; Idroes, R.; Effendi, Y.; Sakib, S.; Emran, T. Potential of Plant Bioactive Compounds as SARS-CoV-2 Main Protease (Mpro) and Spike (S) Glycoprotein Inhibitors: A Molecular Docking Study. Scientifica 2020. [Google Scholar] [CrossRef] [PubMed]
  22. Yang, Y.; Islam, M.; Wang, J.; Li, Y.; Chen, X. Traditional Chinese Medicine in the Treatment of Patients Infected with 2019-New Coronavirus (SARS-CoV-2): A Review and Perspective. Int. J. Biol. Sci. 2020, 16, 1708–1717. [Google Scholar] [CrossRef]
  23. Lee, C. Griffithsin, a Highly Potent Broad-Spectrum Antiviral Lectin from Red Algae: From Discovery to Clinical Application. Mar. Drugs 2019, 17, 567. [Google Scholar] [CrossRef] [Green Version]
  24. 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] [PubMed]
  25. Agrawal, P.; Agrawal, C.; Blunden, G. Quercetin: Antiviral Significance and Possible COVID-19 Integrative Considerations. Nat. Prod. Commun. 2020, 15, 1–10. [Google Scholar]
  26. Olayeriju, O.; Crown, O.; Akinmoladun, A.; Kolawole, A.; Olaleye, M.; Akindahunsi, A. Onions tunic: A Flavonol Rich Competitive Inhibitor of Key Enzyme (Angiotensin-1 Converting Enzyme) Linked Hypertension. Int. J. Sci. Eng. Res. 2017, 8, 2229–5518. [Google Scholar]
  27. Lin, C.; Tsai, F.; Tsai, C.; Lai, C.; Wan, L.; Ho, T.; Hsieh, C.; Chao, P. 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]
  28. Hyun, S.; Lee, H.; Kang, S.; Chung, H.; Choi, J. Inhibitory Activities of Cassia tora and its Anthraquinone Constituents on Angiotensin—Converting Enzyme. Phyther. Res. 2009, 23, 178–184. [Google Scholar] [CrossRef] [PubMed]
  29. Chinsembu, K. Coronaviruses and Nature’s Pharmacy for the Relief of Coronavirus Disease 2019. Rev. Bras. Farmacogn. 2020, 30, 603–621. [Google Scholar] [CrossRef]
  30. Zhuang, M.; Jiang, H.; Suzuki, Y.; Li, X.; Xiao, P.; Tanaka, T.; Ling, H.; Yang, B.; Saitoh, H.; Zhang, L. Procyanidins and Butanol Extract of Cinnamomi Cortex Inhibit SARS-CoV Infection. Antivir. Res. 2009, 82, 73–81. [Google Scholar] [CrossRef]
  31. Polansky, H.; Lori, G. Coronavirus Disease 2019 (COVID-19): First Indication of Efficacy of Gene-Eden-VIR/Novirin in SARS-CoV-2 Infection. Int. J. Antimicrob. Agents 2020, 55, 105971. [Google Scholar] [CrossRef]
  32. Temitope, A.; Eleojo, C.; Abiodun, I.; Ayokunnun, A.; Saheed, S. Phytotherapeutic Evidence against Coronaviruses and Prospects for COVID-19. Pharmacogn. J. 2020, 12, 1252–1267. [Google Scholar]
  33. Shetty, R.; Ghosh, A.; Honavar, S.; Khamar, P.; Sethu, S. Therapeutic Opportunities to Manage COVID-19/SARS-CoV-2 Infection: Present and Future. Indian J. Ophthalmol. 2020, 68, 693–702. [Google Scholar]
  34. Oladele, J.O.; Ajayi, E.I.; Oyeleke, O.M.; Oladele, O.T.; Olowookere, B.D.; Adeniyi, B.M.; Oyewole, O.I.; Oladiji, A.T. A Systematic Review on COVID-19 Pandemic with Special Emphasis on Curative Potentials of Nigeria Based Medicinal Plants. Heliyon 2020, 6, e04897. [Google Scholar] [CrossRef]
  35. Li, H.; Liu, S.-M.; Yu, X.-H.; Tang, S.-L.; Tang, C.-K. Coronavirus Disease 2019 (COVID-19): Current Status and Future Perspectives. Int. J. Antimicrob. Agents 2020, 55, 105951. [Google Scholar] [CrossRef]
  36. Clementi, N.; Scagnolari, C.; D’Amore, A.; Palombi, F.; Criscuolo, E.; Frasca, F.; Pierangeli, A.; Mancini, N.; Antonelli, G.; Clementi, M.; et al. Naringenin is a Powerful Inhibitor of SARS-CoV-2 Infection In vitro. Pharmacol. Res. 2021, 163, 105255. [Google Scholar] [CrossRef]
  37. Bansal, S.; Choudhary, S.; Sharma, M.; Kumar, S.; Lohan, S.; Bhardwaj, V.; Syan, N.; Jyoti, S. Tea: A Native Nource of Antimicrobial Agents. Food Res. Int. 2013, 53, 568–584. [Google Scholar] [CrossRef]
  38. Patten, G.S.; Abeywardena, M.Y.; Head, R.J.; Bennett, L.E. Processed Dietary Plants Demonstrate Broad Capacity for Angiotensin Converting Enzyme and Angiotensin II Receptor Binding Inhibition In Vitro. J. Funct. Foods 2012, 4, 851–863. [Google Scholar] [CrossRef]
  39. Kim, H.; Shin, H.; Park, H.; Kim, Y.; Yun, Y.; Park, S.; Shin, H.; Kim, K. In Vitro Inhibition of Coronavirus Replications by the Traditionally Used Medicinal Herbal Extracts, Cimicifuga rhizoma, Meliae cortex, Coptidis rhizoma, and Phellodendron cortex. J. Clin. Virol. 2008, 41, 122–128. [Google Scholar] [CrossRef] [PubMed]
  40. Schapowal, A. Use of Echinaforce to Prevent Coronavirus Infections. Switzerland. 2020. Available online: https://www.who.int/emergencies/ (accessed on 31 December 2019).
  41. Engler, O.; Strasser, M.; Signer, J.; Schoop, R. Neutralizing Activity of Echinacea purpurea on Coronaviruses Including Highly Pathogenic Middle-East-Respiratory Syndrome Virus (MERS-CoV). Planta Med. Int. Open 2017, 4, 1–202. Available online: http://www.thieme-connect.de/DOI/DOI?10.1055/s-0037-1608557 (accessed on 24 October 2017).
  42. Banyeres, M. Herbario Virtual de Banyeres de Mariola y Alicante. 2010. Available online: http://herbariovirtualbanyeres.blogspot.com/2010/05/morus-alba-morera-morer.html (accessed on 26 May 2020).
  43. Cinatl, J.; Morgenstern, B.; Bauer, G.; Chandra, P.; Rabenau, H.; Doerr, H. Glycyrrhizin, an Active Component of Liquorice Roots, and Replication of SARS-Associated Coronavirus. Lancet 2003, 361, 2045–2046. [Google Scholar] [CrossRef] [Green Version]
  44. Baltina, L.; Zarubaev, V.; Baltina, L.; Orshanskaya, I.; Fairushina, A.; Kiselev, O.; Yunusov, M. Glycyrrhizic Acid Derivatives as Influenza A/H1N1 Virus Inhibitors. Bioorganic Med. Chem. Lett. 2015, 25, 1742–1746. [Google Scholar] [CrossRef]
  45. Cheng, P.; Ng, L.; Chiang, L.; Lin, C. Antiviral Effects of Saikosaponins on Human Coronavirus 229E In vitro. Clin. Exp. Pharmacol. Physiol. 2006, 33, 612–616. [Google Scholar] [CrossRef]
  46. Chen, F.; Chan, K.; Jiang, Y.; Kao, R.; Lu, H.; Fan, K.; Cheng, V.; Tsui, W.; Hung, I.; Lee, T. In Vitro Susceptibility of 10 Clinical Isolates of SARS Coronavirus to Selected Antiviral Compounds. J. Clin. Virol. 2004, 31, 69–75. [Google Scholar] [CrossRef] [PubMed]
  47. Kumaki, Y.; Wandersee, M.; Smith, A.; Zhou, Y.; Simmons, G.; Nelson, N.; Bailey, K.; Vest, Z.; Li, J.; Chan, P. Inhibition of Severe Acute Respiratory Syndrome Coronavirus Replication in a Lethal SARS-CoV BALB/c Mouse Model by Stinging Nettle Lectin, Urtica dioica Agglutinin. Antivir. Res. 2011, 90, 22–32. [Google Scholar] [CrossRef] [PubMed]
  48. Wang, Q.; Quan, Q.; Zhou, X.; Zhu, Y.; Lan, Y.; Li, S.; Yu, Y.; Cheng, Z. A Comparative Study of Lonicera japonica with Related Species: Morphological Characteristics, ITS Sequences and Active Compounds. Biochem. Syst. Ecol. 2014, 54, 198–207. [Google Scholar] [CrossRef] [PubMed]
  49. Shang, X.; Pan, H.; Li, M.; Miao, X.; Ding, H. Lonicera japonica Thunb.: Ethnopharmacology, Phytochemistry and Pharmacology of an Important Traditional Chinese Medicine. J. Ethnopharmacol. 2011, 138, 1–21. [Google Scholar] [CrossRef]
  50. Li, S.; Chen, C.; Zhang, H.; Guo, H.; Wang, H.; Wang, L.; Zhang, X.; Hua, S.; Yu, J.; Xiao, P. Identification of Natural Compounds with Antiviral Activities against SARS-Associated Aoronavirus. Antivir. Res. 2005, 67, 18–23. [Google Scholar] [CrossRef]
  51. Cho, J.; Curtis-Long, M.; Lee, K.; Kim, D.; Ryu, H.; Yuk, H.; Park, K. Geranylated Flavonoids Displaying SARS-CoV Papain-Like Protease Inhibition from the Fruits of Paulownia tomentosa. Bioorganic Med. Chem. 2013, 21, 3051–3057. [Google Scholar] [CrossRef] [PubMed]
  52. Michaelis, M.; Doerr, H.; Cinatl, J. Investigation of the Influence of EPs® 7630, a Herbal Drug Preparation from Pelargonium sidoides, on Replication of a Broad Panel of Respiratory Viruses. Phytomedicine 2011, 18, 384–386. [Google Scholar] [CrossRef]
  53. Theisen, L.L.; Muller, C.P. EPs® 7630 (Umckaloabo®), an Extract from Pelargonium sidoides Roots, Exerts Anti-influenza Virus Activity In Vitro and In Vivo. Antivir. Res. 2012, 94, 147–156. [Google Scholar] [CrossRef] [PubMed]
  54. Moyo, M.; Van, J. Medicinal Properties and Conservation of Pelargonium sidoides DC. J. Ethnopharmacol. 2014, 152, 243–255. [Google Scholar] [CrossRef] [PubMed]
  55. Lin, L.; Hsu, W.; Lin, C. Antiviral Natural Products and Herbal Medicines. J. Tradit. Complement. Med. 2014, 4, 24. [Google Scholar] [CrossRef] [Green Version]
  56. 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]
  57. 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] [PubMed] [Green Version]
  58. Loizzo, M.; Saab, A.; Tundis, R.; Statti, G.; Menichimi, F.; Lampronti, D.; Gambari, R.; Cinatl, J.; Doerr, H. Phytochemical Analysis and In Vitro Antiviral Activities of the Essential Oils of Seven Lebanon Species. Chem. Biodivers. 2008, 5, 461–470. [Google Scholar] [CrossRef] [PubMed]
  59. Dong, X.; Fu, J.; Yin, X.; Cao, S.; Li, X.; Lin, L.; Ni, J. Emodin: A Review of its Pharmacology, Toxicity and Pharmacokinetics. Phytother. Res. 2016, 30, 1207–1218. [Google Scholar] [CrossRef]
  60. Patten, G.S.; Abeywardena, M.Y.; Bennett, L.E. Inhibition of Angiotensin Converting Enzyme, Angiotensin II Receptor Blocking, and Blood Pressure Lowering Bioactivity across Plant Families. Crit. Rev. Food Sci. Nutr. 2016, 56, 181–214. [Google Scholar] [CrossRef] [PubMed]
  61. Semwal, D.; Semwal, R.; Combrinck, S.; Viljoen, A. Myricetin: A Dietary Molecule with Diverse Biological Activities. Nutrients 2016, 8, 90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Li, Y.; Li, Z.; Zhao, W.; Wen, R.; Meng, Q.; Zeng, Y. Synthesis of Stilbene Derivatives with Inhibition of SARS Coronavirus Replication. Eur. J. Med. Chem. 2006, 41, 1084–1089. [Google Scholar] [CrossRef] [PubMed]
  63. Lin, S.; Ho, C.; Chuo, W.; Li, S.; Wang, T.; Lin, C. Effective Inhibition of MERS-CoV Infection by Resveratrol. BMC Infect. Dis. 2017, 17, 144. [Google Scholar] [CrossRef] [Green Version]
  64. Maurya, V.K.; Kumar, S.; Bhatt, M.L.; Saxena, S.K. Therapeutic Development and Drugs for the Treatment of COVID-19. In Coronavirus Disease 2019 (COVID-19); Nature Publishing Group: Berlin, Germany, 2020; pp. 109–126. [Google Scholar]
  65. Palit, P.; Chattopadhyay, D.; Thomas, S.; Kundu, A.; Kim, H.; Rezaei, N. Phytopharmaceuticals Mediated Furin and TMPRSS2 Receptor Blocking: Can It Be a Potential Therapeutic Option for Covid-19? Phytomedicine 2020, 85, 153396. [Google Scholar] [CrossRef]
  66. Vaduganathan, M.; Vardeny, O.; Michel, T.; McMurray, J.J.; Pfeffer, M.A. Solomon SD. Renin–Angiotensin–Aldosterone System Inhibitors in Patients with Covid-19. N. Engl. J. Med. 2020, 382, 1653–1659. [Google Scholar] [CrossRef]
  67. Christy, M.; Uekusa, Y.; Gerwick, L.; Gerwick, W. Natural Products with Potential to Treat RNA Virus Pathogens Including SARS-CoV-2. J. Nat. Prod. 2021, 84, 161–182. [Google Scholar] [CrossRef] [PubMed]
  68. Manzano, P.; Peñarreta, J.; Chóez, I.; Barragán, A.; Orellana, A.; Rastrelli, L. Potential Bioactive Compounds of Medicinal Plants against New Coronavirus (SARS-CoV-2): A Review. Bionatura 2020. [Google Scholar] [CrossRef]
  69. Balslev, H.; Navarrete, H.; Torre, L.; Macía, M. Enciclopedia de Plantas Útiles del Ecuador; Herbario QCA de la Escuela de Ciencias Biológicas de la Pontificia, Universidad Católica del Ecuador: Quito, Ecuador, 2008; pp. 1–323. [Google Scholar]
  70. León, S.; Valencia, R.; Pitman, N.; Endara, L.; Ulloa, H.; Navarrete, C. Libro Rojo de las Plantas Endémicas del Ecuador, 2nd ed.; Pontifica Universidad Católica del Ecuador: Quito, Ecuador, 2011; pp. 1–440. [Google Scholar]
  71. Sut, S.; Maggi, F.; Dall’Acqua, S. Bioactive Secondary Metabolites from Orchids (Orchidaceae). Chem. Biodivers. 2017, 14, e1700172. [Google Scholar] [CrossRef] [PubMed]
  72. Naranjo, P.; Escaleras, R. La Medicina Tradicional en el Ecuador: Memorias de las Primeras Jornadas Ecuatorianas de Etnomedicina Andina; Universidad Andina Simón Bolívar: Quito, Ecuador, 2002; pp. 1–192. [Google Scholar]
  73. Selmi, C.; Ansari, A.; Invernizzi, P.; Podda, M.; Gershwin, E. The Search for a Practical Approach to Emerging Diseases: The Case of Severe Acute Respiratory Syndrome (SARS). Dev. Immunol. 2002, 9, 113–117. [Google Scholar] [CrossRef] [Green Version]
  74. Zhong, N. Management and Prevention of SARS in China. R. Soc. 2004, 359, 1115–1116. [Google Scholar] [CrossRef] [PubMed]
  75. Cheng, V.; Lau, S.; Woo, P.; Yuen, K. Severe Acute Respiratory Syndrome Coronavirus as an Agent of Emerging and Reemerging Infection. Clin. Microbiol. Rev. 2007, 20, 660–694. [Google Scholar] [CrossRef] [Green Version]
  76. Ruiz, M.; Ruperez, M.; Lorenzo, O.; Esteban, V.; Blanco, J.; Mezzano, S.; Egido, J. Angiotensin II Regulates the Synthesis of Proinflammatory Cytokines and Chemokines in the Kidney. Kidney Int. Suppl. 2002, 62, 12–22. [Google Scholar] [CrossRef] [Green Version]
  77. Carlson, S.H.; Wyss, J.M. Mechanisms Underlying Hypertension and Obesity. Hypertension 2011, 57, 375–376. [Google Scholar] [CrossRef] [Green Version]
  78. Valverde, F.D.M. Plantas Utiles del Litoral Ecuatoriano; Fundación Ecuatoriana de Estudios Ecológicos: Quito, Ecuador, 1998. [Google Scholar]
  79. Shang, J.; Wan, Y.; Luo, C.; Ye, G.; Geng, Q.; Auerbach, A.; Li, F. Cell Entry Mechanisms of SARS-CoV-2. Proc. Natl. Acad. Sci. USA 2020, 117, 11727–11734. [Google Scholar] [CrossRef]
  80. Sawalha, A.; Zhao, M.; Coit, P.; Lu, Q. Epigenetic Dysregulation of ACE2 and Interferon-regulated Genes Might Suggest Increased COVID-19 Susceptibility and Severity in Lupus Patients. Clin. Immunol. 2020, 215, 108410. [Google Scholar] [CrossRef]
  81. Remkova, A.; Remko, M. The Renin-Angiotensin-Aldosterone System and Prothrombotic State in Arterial Hypertension. Salud Cienc. 2011, 18, 220–224. [Google Scholar]
  82. Ayada, C.; Toru, Ü.; Korkut, Y. The Relationship of Stress and Blood Pressure Effectors. Hippokratia 2015, 19, 99. [Google Scholar] [PubMed]
  83. Gurwitz, D. Angiotensin Receptor Blockers as Tentative SARS-CoV-2 Therapeutics. Drug Dev. Res. 2020, 81, 537–540. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Arnold, A.C.; Sakima, A.; Kasper, S.O.; Vinsant, S.; Garcia, M.A.; Diz, D.I. The Brain Renin-Angiotensin System and Cardiovascular Responses to Stress: Insights from Transgenic Rats with Low Brain Angiotensinogen. J. Appl. Physiol. 2012, 113, 1929–1936. [Google Scholar] [CrossRef] [Green Version]
  85. Guo, F.; Chen, X.L.; Wang, F.; Liang, X.; Sun, Y.X.; Wang, Y.J. Role of Angiotensin II Type 1 Receptor in Angiotensin II-Induced Cytokine Production in Macrophages. J. Interferon Cytokine Res. 2011, 31, 351–361. [Google Scholar] [CrossRef]
  86. Engeli, S.; Schling, P.; Gorzelniak, K.; Boschmann, M.; Janke, J.; Ailhaud, G.; Teboul, M.; Massiéra, F.; Sharma, A. The Adipose-Tissue Renin-Angiotensin-Aldosterone System: Role in the Metabolic Syndrome? Int. J. Biochem. Cell Biol. 2003, 35, 807–825. [Google Scholar] [CrossRef]
  87. Khanna, K.; Kohli, S.; Kaur, R.; Bhardwaj, A.; Bhardwaj, V.; Ohri, P.; Sharma, A.; Ahmad, A.; Bhardwaj, R.; Ahmad, P. Herbal Immune-Boosters: Substantial Warriors of Pandemic Covid-19 Battle. Phytomedicine 2021, 85, 153361. [Google Scholar] [CrossRef]
  88. Malinowska, M.; Sikora, E.; Ogonowski, J. Production of Triterpenoids with Cell and Tissue Cultures. Acta. Biochim. Pol. 2013, 60, 731–735. [Google Scholar] [CrossRef] [Green Version]
  89. Teissier, E.; Penin, F.; Pécheur, E. Targeting Cell Entry of Enveloped Viruses as an Antiviral Strategy. Molecules 2011, 16, 221–250. [Google Scholar] [CrossRef]
  90. Fedoung, E.F.; Biwole, A.B.; Biyegue, C.F.N.; Tounkam, M.N.; Ntonga, P.A.; Nguiamba, V.P.; Essono, D.M.; Funwi, P.F.; Tonga, C.; Nguenang, G.M.; et al. A Review of Cameroonian Medicinal Plants with Potentials for the Management of the COVID-19 Pandemic. Adv. Tradit. Med. 2021, 1–26. [Google Scholar] [CrossRef]
  91. Walter, T.; Justinraj, S.; Justinraj, C.; Nandini, V. Effect of Nilavembu Kudineer in the Prevention and Management of COVID-19 by Inhibiting ACE2 Receptor. Siddha Pap. 2020. Available online: www.siddhapapers.org (accessed on 21 December 2020).
  92. Wu, C.; Liu, Y.; Yang, Y.; Zhang, P.; Zhong, W.; Wang, Y.; Wang, Q.; Xu, Y.; Li, M.; Li, X. Analysis of Therapeutic Targets for SARS-CoV-2 and Discovery of Potential Drugs by Computational Methods. Acta Pharm. Sin. B 2020, 10, 766–788. [Google Scholar] [CrossRef]
  93. 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]
  94. Maiti, S.; Banerjee, A. Epigallocatechin Gallate and Theaflavin Gallate Interaction in SARS-CoV-2 Spike-protein Central Channel with Reference to the Hydroxychloroquine Interaction: Bioinformatics and Molecular Docking Study. Drug Dev. Res. 2021, 82, 86–96. [Google Scholar] [CrossRef]
  95. Ngwa, W.; Kumar, R.; Thompson, D.; Lyerly, W.; Moore, R.; Reid, T.E.; Lowe, H.; Toyang, N. Potential of Flavonoid-Inspired Phytomedicines against COVID-19. Molecules 2020, 25, 2707. [Google Scholar] [CrossRef]
  96. Chen, C.; Lin, C.; Huang, K.; Chen, W.; Hsieh, H.; Liang, P.; Hsu, J. Inhibition of SARS-CoV 3C-Like Protease Activity by Theaflavin-3,3′- Digallate (TF3). Evid. Based Complementary Altern. Med. 2005, 2, 209–215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Musarra, M.; Pennisi, R.; Ben, I.; Mandalari, G.; Sciortino, M.T. Antiviral Activity Exerted by Natural Products against Human Viruses. Viruses 2021, 13, 828. [Google Scholar] [CrossRef] [PubMed]
  98. Schötz, K.; Nöldner, M. Mass Spectroscopic Characterisation of Oligomeric Proanthocyanidins Derived from an Extract of Pelargonium sidoides Roots (EPs® 7630) and Pharmacological Screening in CNS Models. Phytomedicine 2007, 14, 32–39. [Google Scholar] [CrossRef] [PubMed]
  99. Verma, S.; Twilley, D.; Esmear, T.; Oosthuizen, C.; Reid, A.M.; Nel, M.; Lall, N. Anti-SARS-CoV Natural Products with the Potential to Inhibit SARS-CoV-2 (COVID-19). Front. Pharmacol. 2020, 11, 1514. [Google Scholar] [CrossRef]
  100. Orhan, I.E.; Senol, F.S. Natural Products as Potential Leads Against Coronaviruses: Could They be Encouraging Structural Models Against SARS-CoV-2? Nat. Prod. Bioprospecting 2020, 10, 171–186. [Google Scholar] [CrossRef]
  101. Signer, J.; Jonsdottir, H.R.; Albrich, W.C.; Strasser, M.; Züst, R.; Ryter, S.; Ackermann, R.; Lenz, N.; Siegrist, D.; Suter, A. In Vitro Virucidal Activity of Echinaforce®, an Echinacea purpurea Preparation, against Coronaviruses, Including Common Cold Coronavirus 229E and SARS-CoV-2. Virol. J. 2020, 17, 136. [Google Scholar] [CrossRef]
  102. Meeran, M.N.; Javed, H.; Sharma, C.; Goyal, S.N.; Kumar, S.; Jha, N.K.; Ojha, S. Can Echinacea be a Potential Candidate to Target Immunity, Inflammation, and Infection—The Trinity of Coronavirus Disease 2019. Heliyon 2021, 7, e05990. [Google Scholar] [CrossRef]
  103. Grant, W.; Lahore, H.; McDonnell, S.; Baggerly, C.; French, C.; Aliano, J.; Bhattoa, H. Evidence that Vitamin D Supplementation Could Reduce Risk of Influenza and COVID-19 Infections and Deaths. Nutrients 2020, 12, 988. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Rondanelli, M.; Miccono, A.; Lamburghini, S.; Avanzato, I.; Riva, A.; Allegrini, P.; Faliva, M.; Peroni, G.; Nichetti, M.; Perna, S. Self-Care for Common Colds: The Pivotal Role of Vitamin D, Vitamin C, Zinc, and Echinacea in Three Main Immune Interactive Clusters (Physical Barriers, Innate and Adaptive Immunity) Involved During an Episode of Common Colds—Practical Advice on Dosages. Evid. Based Complementary Altern. Med. 2018, 2018, 5813095. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Wong, S.S.; Yuen, K.Y. The Management of Coronavirus Infections with Particular Reference to SARS. J. Antimicrob. Chemother. 2008, 62, 437–441. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Llivisaca, S.; Manzano, P.; Ruales, J.; Flores, J.; Mendoza, J.; Peralta, E.; Cevallos-Cevallos, J.M. Chemical, Antimicrobial, and Molecular Characterization of Mortiño (Vaccinium floribundum Kunth) Fruits and Leaves. Food Sci. Nutr. 2018, 6, 934–942. [Google Scholar] [CrossRef]
  107. Hidalgo, M.; Martin, S.; Recio, I.; Sanchez, C.; De Pascual, B.; Rimbach, G.; De Pascual, S. Potential Anti-inflammatory, Anti-adhesive, Anti/Estrogenic, and Angiotensin-Converting Enzyme Inhibitory Activities of Anthocyanins and their Gut Metabolites. Genes Nutr. 2012, 7, 295–306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Muchtaridi, M.; Fauzi, M.; Khairul Ikram, N.K.; Mohd Gazzali, A.; Wahab, H.A. Natural Flavonoids as Potential Angiotensin-Converting Enzyme 2 Inhibitors for Anti-SARS-CoV-2. Molecules 2020, 25, 3980. [Google Scholar] [CrossRef]
  109. Thabti, I.; Albert, Q.; Philippot, S.; Dupire, F.; Westerhuis, B.; Fontanay, S.; Risler, A.; Kassab, T.; Elfalleh, W.; Aferchichi, A.; et al. Advances on Antiviral Activity of Morus spp. Plant Extracts: Human Coronavirus and Virus-related Respiratory Tract Infections in the Spotlight. Molecules 2020, 25, 1876. [Google Scholar] [CrossRef]
  110. Xian, Y.; Zhang, J.; Bian, Z.; Zhou, H.; Zhang, Z.; Lin, Z.; Xu, H. Bioactive Natural Compounds against Human Coronaviruses: A Review and Perspective. Acta Pharm. Sin. B 2020, 10, 1163–1174. [Google Scholar] [CrossRef] [PubMed]
  111. Choy, K.; Wong, A.; Kaewpreedee, P.; Sia, S.; Chen, D.; Hui, K.; Chu, D.; Chan, M.; Cheung, P.; Huang, X. Remdesivir, Lopinavir, Emetine, and Homoharringtonine Inhibit SARS-CoV-2 Replication In vitro. Antivir. Res. 2020, 178, 104786. [Google Scholar] [CrossRef] [PubMed]
  112. Hassan, S. Shedding Light on the Effect of Natural Anti-Herpesvirus Alkaloids on SARS-CoV-2: A Treatment Option for COVID-19. Viruses 2020, 12, 476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Valadão, A.L.; Abreu, C.M.; Días, J.Z.; Arantes, P.; Verli, H.; Tanuri, A.; De Aguiar, R.S. Natural Plant Alkaloid (Emetine) Inhibits HIV-1 Replication by Interfering with Reverse Transcriptase Activity. Molecules 2015, 20, 11474–11489. [Google Scholar] [CrossRef] [Green Version]
  114. Bian, Y.; An, G.J.; Kim, K.; Ngo, T.; Shin, S.; Bae, O.N.; Lim, K.M.; Chung, J.H. Ginsenoside Rg3, a Component of Ginseng, Induces Pro-thrombotic Activity of Erythrocytes Via Hemolysis-associated Phosphatidylserine Exposure. Food Chem. Toxicol. 2019, 131, 110553. [Google Scholar] [CrossRef] [PubMed]
  115. Gafner, S. Herbal Drugs and Phytopharmaceuticals, 3rd ed.; American Chemical Society: London, UK, 2004; pp. 1774–1775. [Google Scholar]
  116. Efferth, T.; Banerjee, M.; Paul, N.; Abdelfatah, S.; Arend, J.; Elhassan, G.; Hamdoun, S.; Hamm, R.; Hong, C.; Kadioglu, O. Biopiracy of Natural Products and Good Bioprospecting Practice. Phytomedicine 2016, 23, 166–173. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Various active principles and their mechanism of action. The infection cycle of SARS-CoV-2 in human cells. The SARS-CoV-2 spike (S) protein binds to ACE2 in host cells followed by priming of protein S by transmembrane protease serine 2 protease (TMPRSS2). Then, the virus produces the polyproteins pp1a and pp1ab, which are processed by viral proteases (3CLpro/Mpro, PLpro) to non-structural proteins (nsps), including RNA-dependent RNA polymerase (RdRp). Viral RdRp synthesizes a full-length complementary negative-strand RNA as a template for the production of the positive strand genome of the virus. Subgenomic mRNAs are then translated into structural proteins in the rough endoplasmic reticulum or in the cytosol. The viral genomic RNA is encapsulated by the nucleocapsid protein N and, finally, the virus is released by exocytosis. The blunt arrows indicate the possible targets of the active principles of medicinal plants. Irreversibly interference with viral docking receptors: Caphtharic acid (o), cichoric acid and echinacoside from Echinacea purpurea (p), vitamins D, C and Zn (q). Entry locks: Emodin (a), lectins (b), quercetin (c), catechin (d), naringenin (e), hesperetin (f), baicalin (g), epigallocatechin (h), gallocatechin gallate (i), prodelphinidin (af), gallocatechin (ag), saikosaponins derivatives of oleanane from Heteromorpha arborescens and Bupleurum spp. (aj), glycyrrhizine (al), Licorice (am), desmethoxyreserpine (ao), dihydrotanshinone I (ay). ACE2 receptor blocking: Emodin (a), hesperetin (f), kaempferol (j), anthocyanins (t), phenolic compounds: tannic acid (aa), 3-isotheaflavin-3-gallate (ab) and theaflavin-3,3′-digallate (ac) from Camellia sinensis. TMPRSS2 receptor blocking: [6]-gingerol (ak). Block the entry of cells through endocytosis: Butanol extract (v) and procyanidins (ai) from Cinnamomum verum. Inhibit 3CLpro: Quercetin (c), kaempferol (j), curcumin (y), sinigrin (z), eugenol (an), betulinic acid (ap), coumaroyltyramine (aq), cryptotanshinone (ar), desmethoxyreserpine (ao), Dihomo-γ-linolenic acid (au), lignan (as), sugiol (at), N-cis-feruloyltyramine (av), Tanshinone IIa (aw). Inhibit PLpro: Quercetin (c), baicalin (g), kaempferol (j), myricetin (n), scutellarein (l), eugenol (an), coumaroyltyramine (aq), cryptotanshinone (ar), N-cis-feruloyltyramine (av), Tanshinone IIa (aw), moupinamide (ax). Affinity with S protein: Eugenol (an), dihydrotanshinone I (ay). Viral replication: Aescin (k), kaempferol (j), resveratrol (s), prodelphinidin (af), gallocatechin (ag), epigallocatechin isomer (ah) from Pelargonium sidoides, essential oils: β-ocimene, 1,8-cineole, α-pinene and β-pinene (ad), phenolic compounds: tannic acid (aa), 3-isotheaflavin-3-gallate (ab) and theaflavin-3,3′-digallate (ac), betulinic acid (ap), desmethoxyreserpine (ao), lignan (as), sugiol (at). Affects the release or assembly of the virus: Phenolic compounds: tannic acid (aa), 3-isotheaflavin-3-galalate (ab) and theaflavin-3,3′-digallate (ac), lectin agglutinin (w) from Hippeastrum striatum. TNF-β, IL-1β expressions: Quercetin (c), luteoloside (m), chlorogenic acid (x) geranylated flavonoids (tomebrin A, B, D and E) (r), resveratrol (s), anthocyanins (t), gallic acid (u), prodelphinidin (af), gallocatechin (ag), epigallocatechin isomer (ah).
Figure 1. Various active principles and their mechanism of action. The infection cycle of SARS-CoV-2 in human cells. The SARS-CoV-2 spike (S) protein binds to ACE2 in host cells followed by priming of protein S by transmembrane protease serine 2 protease (TMPRSS2). Then, the virus produces the polyproteins pp1a and pp1ab, which are processed by viral proteases (3CLpro/Mpro, PLpro) to non-structural proteins (nsps), including RNA-dependent RNA polymerase (RdRp). Viral RdRp synthesizes a full-length complementary negative-strand RNA as a template for the production of the positive strand genome of the virus. Subgenomic mRNAs are then translated into structural proteins in the rough endoplasmic reticulum or in the cytosol. The viral genomic RNA is encapsulated by the nucleocapsid protein N and, finally, the virus is released by exocytosis. The blunt arrows indicate the possible targets of the active principles of medicinal plants. Irreversibly interference with viral docking receptors: Caphtharic acid (o), cichoric acid and echinacoside from Echinacea purpurea (p), vitamins D, C and Zn (q). Entry locks: Emodin (a), lectins (b), quercetin (c), catechin (d), naringenin (e), hesperetin (f), baicalin (g), epigallocatechin (h), gallocatechin gallate (i), prodelphinidin (af), gallocatechin (ag), saikosaponins derivatives of oleanane from Heteromorpha arborescens and Bupleurum spp. (aj), glycyrrhizine (al), Licorice (am), desmethoxyreserpine (ao), dihydrotanshinone I (ay). ACE2 receptor blocking: Emodin (a), hesperetin (f), kaempferol (j), anthocyanins (t), phenolic compounds: tannic acid (aa), 3-isotheaflavin-3-gallate (ab) and theaflavin-3,3′-digallate (ac) from Camellia sinensis. TMPRSS2 receptor blocking: [6]-gingerol (ak). Block the entry of cells through endocytosis: Butanol extract (v) and procyanidins (ai) from Cinnamomum verum. Inhibit 3CLpro: Quercetin (c), kaempferol (j), curcumin (y), sinigrin (z), eugenol (an), betulinic acid (ap), coumaroyltyramine (aq), cryptotanshinone (ar), desmethoxyreserpine (ao), Dihomo-γ-linolenic acid (au), lignan (as), sugiol (at), N-cis-feruloyltyramine (av), Tanshinone IIa (aw). Inhibit PLpro: Quercetin (c), baicalin (g), kaempferol (j), myricetin (n), scutellarein (l), eugenol (an), coumaroyltyramine (aq), cryptotanshinone (ar), N-cis-feruloyltyramine (av), Tanshinone IIa (aw), moupinamide (ax). Affinity with S protein: Eugenol (an), dihydrotanshinone I (ay). Viral replication: Aescin (k), kaempferol (j), resveratrol (s), prodelphinidin (af), gallocatechin (ag), epigallocatechin isomer (ah) from Pelargonium sidoides, essential oils: β-ocimene, 1,8-cineole, α-pinene and β-pinene (ad), phenolic compounds: tannic acid (aa), 3-isotheaflavin-3-gallate (ab) and theaflavin-3,3′-digallate (ac), betulinic acid (ap), desmethoxyreserpine (ao), lignan (as), sugiol (at). Affects the release or assembly of the virus: Phenolic compounds: tannic acid (aa), 3-isotheaflavin-3-galalate (ab) and theaflavin-3,3′-digallate (ac), lectin agglutinin (w) from Hippeastrum striatum. TNF-β, IL-1β expressions: Quercetin (c), luteoloside (m), chlorogenic acid (x) geranylated flavonoids (tomebrin A, B, D and E) (r), resveratrol (s), anthocyanins (t), gallic acid (u), prodelphinidin (af), gallocatechin (ag), epigallocatechin isomer (ah).
Molecules 26 04099 g001
Figure 2. Mechanism of action of SARS-CoV-2 on the Renin–Angiotensin–Aldosterone System (RAAS) and its possible regulation by the Angiotensin converting enzyme inhibitors (ACEi), Angiotensin receptor blockers ARBs or Angiotensin converting enzyme (ACE2) that converts AI to A1-7 to restore the RAAS.
Figure 2. Mechanism of action of SARS-CoV-2 on the Renin–Angiotensin–Aldosterone System (RAAS) and its possible regulation by the Angiotensin converting enzyme inhibitors (ACEi), Angiotensin receptor blockers ARBs or Angiotensin converting enzyme (ACE2) that converts AI to A1-7 to restore the RAAS.
Molecules 26 04099 g002
Table 1. Pathogenetic and epidemiological characteristics of SARS-CoV-2, SARS-CoV and MERS-CoV.
Table 1. Pathogenetic and epidemiological characteristics of SARS-CoV-2, SARS-CoV and MERS-CoV.
SpeciesReceptorIncubation PeriodROCase Fatality RateReferences
SARS-CoV-2ACE21 to 14 days3.28 3.4[3,5,7]
SARS-CoVACE22 to 10 days1.7–1.99.6[4,8]
MERS-CoVDPP40.935[2]
Table 2. Medicinal plants and natural products with inhibitory activity against various types of coronaviruses.
Table 2. Medicinal plants and natural products with inhibitory activity against various types of coronaviruses.
Scientific/Common NameActive PrincipleVirus/ Antiviral ActivityReference
Aesculus hippocastanum CN: Horse-chestnutAescin (k)SARS-CoV/Inhibits viral replication [22]
Allium ampeloprasum Var. porrum J. Gay
CN: Leek
Mannose-binding specific lectin (b)SARS-CoV/Ability to bind to the glycosylated molecules found on the surface of viruses, including the spike glycoprotein [23]
[24]
Allium cepa L.
CN: Onion
Flavonols: quercetin, quercetinglycosides (isoquercitrin, quercitrin and rutin) (c) and kaempferol (j)SARS-CoV2/Interfere with various stages of the coronavirus entry and replication cycle such as PLpro, 3CLpro, and NTPase/helicase; Inhibits ACE by competing with the substrate, N-[3-(2-furyl) acryloyl]-L-phenylalanylglycylglycine[25]
[26]
Brassica oleracea L.
CN: Broccoli
Glucosinolate type sinigrin (z)SARS-CoV/blocks the cleavage process of 3CLpro[27]
[28]
Bupleurum spp.
CN: Bupleurum
Oleanane-type saikosaponins (aj)SARS-CoV/Inhibit human coronavirus entry into cells, general replication, and specific 3CLpro mediated replication [29]
Cassia tora L.
CN:
Anthraquinone derived emodin (a)Inhibitory activities on angiotensin-converting enzyme.[28]
Cinnamomumverum J. Presl
CN: Cinnamon (cortex)
Butanol (v), procyanidins (ai)SARS-CoV/Possibly blocks the entry of cells through endocytosis[30]
[31]
[32]
Curcuma spp.
CN: Turmeric
Curcumin (y),
Eugenol (an)
SARS-CoV/Inhibits 3CLpro (y); Good binding affinity with Mpro and S protein (an)[33]
[34]
Citrus spp.
CN: Three main species in the country: Citrus maxima (Rumph. ex Burm.) Merr; Citrus medica L.; Citrus reticulata Blanco.
Hesperetin (f) and naringenin (e)SARS-CoV-2/(f) Inhibits ACE2 and inhibit the entry of virus into cells host by binding to S protein, helicase, and protease sites on the ACE receptor
HCoV229E/(e) Partial inhibition of 229E replication in cells silenced for TPC2 by siRNA
[35]
[36]
Camellia sinensis Kuntze
CN: Green tea
Phenolic compounds:
Tannic acid (aa), 3-isotheaflavin-3-galalate (ab) and theaflavin-3,3′-digallate (ac)
Coronavirus in general/Possibly inhibition of RNA polymerase or RNA-dependent proteases; They can also affect the release or assembly of the virus; inhibits ECA and blocking AII receptor binding in vitro, avoiding symptoms of various diseases, especially those of a respiratory nature[37]
[38]
Melia azedarach L.
CN: Cinamomo
[39]
[37]
Echinacea purpurea Moench
CN: Echinaceae®
Caphtharic acid (o), cichoric acid (p) and echinacoside (p)MERS-CoV, 229E/The extract non-specifically and irreversibly interferes with viral docking receptors (eg, influenza) to block infectivity of pathogens[40]
[41]
Ginkgo biloba L.
CN: Ginkgo
Ginkgolide, terpenic lactones, flavonoids, polyphenols, oleic acid, among others.SARS-CoV/Antiviral mechanism is unclear[19]
[42]
Glycyrrhiza glabra L.
CN: Licorice (root)
Licorice (am) y glycyrrhizin (al)SARS-CoV/Modulate some virus-host fusion functions through the envelope of the repetition domain 2 of the predominant heptad in viral envelopes; Improvement of the function of upper respiratory mucosal immune system; Inhibit viral adsorption and penetration[29]
[22]
[43]
[44]
Heteromorpha arborescens Cham.
CN: Parsley tree
Oleanane-type saikosaponins (aj) SARS-CoV/Prevent the entry of SARS-CoV into the cell[45]
[46]
Hippeastrum striatum Lam
CN: Lily
Lectin agglutinin (w)SARS-CoV/Inhibits the end of the virus cycle infection[29]
[47]
Lonicera japonica Thunb
CN: Madreselva
Eriobotrya japonica Thunb
CN: Níspero
Quercetin (c), luteoloside (m), chlorogenic acid (x)SARS-CoV, RSV, HIV, HSV, PRV and NDV/This mechanism possibly is due to diminishing the inflammation mediators and TNF-β, IL-1β expression. Anti-inflammatory, antiviral, antibacterial, antioxidant activity. Enhances the immune response.[48]
[49]
Lycoris spp.
CN: hurricane lilies or cluster amaryllis
LycorineSARS-CoV/Compound with extensive antiviral activities. However, the antiviral mechanism of this molecule is unclear[50]
Morus alba L.
CN: Tree mulberry
Aliphatic, aromatic phenolic, heterocyclic and aliphatic cyclic compoundsSARS-CoV and MERS-CoV/Antiviral mechanism is unclear[19]
[42]
Nicotiana tabacum L.
CN: Tobacco
N-acetylglucosamine specific lectins (b)SARS-CoV/Ability to bind to the glycosylated molecules found on the surface of viruses, including the spike glycoprotein. [23]
[29]
Paulownia tomentosa Steud
CN: Kiri
Flavonoids: (quercetin (c), catechin (d) and naringenin (e) and geranilated flavonoids (tomentin A, tomentin B, tomentin C, tomentin D, tomentin E) (r)SARS-CoV/Inhibits SARS-CoV (PLpro) by reducing the concentration of pro-inflammatory cytokines (IL-1β) and TNFα[51]
Pelargonium sidoides D.C.
CN: Geranium
Prodelphinidin (af), gallocatechin (ag) and their epigallocatechin stereoisomer (ah)H1N1, H3N2, HCoV-229E/inhibits the entry and replication of 229E; Also is immunomodulatory and cytoprotective effects, inhibition of the interaction between bacteria and host cells; Inhibits viral hemagglutination and Neuraminidase (NA) activity[52]
[53]
[54]
[55]
Psidium guajava
CN: Guava
Eugenol (an)SARS-CoV/Good binding affinity with Mpro and S protein[34]
[21]
Scutellaria baicalensis Georgi.
CN: Skullcap
Baicalin (g) and scutellarein (l)SARS-CoV/Inhibits nsP13 in vitro by affecting ATPase activity[56]
[57]
[46]
Thuja orientalis L.
CN: Tree of life
Essential oils:
b-ocimene, 1,8-cineole, a-pinene and b-pinene mainly (ad)
SARS-CoV, HSV-1/Inhibitory activity against viral replication in vitro by visually scoring of the virus-induced cytopathogenic effect post-infection[58]
[29]
Laurus nobilis L.
CN: Laurel
Salvia officinalis L.
CN: Sage
Urtica dioica L.
CN: Nettle
Lectin agglutinin (w)SARS-CoV/Inhibits the end of the virus cycle infection[29]
[47]
Polygonum cuspidatum L.
CN: Japanese knotty grass
Anthraquinone derived emodin (a)SARS-CoV, HCoV-OC43/inhibits by blocking viral entry by binding to the S protein and interfering with the 3CLpro activity of the SARS-CoV and prevented the formation of the Nsp required for viral replication; Blocked the interaction between SARS-CoV S protein and ACE2, inhibited ion channel 3a and interrupted the release of new coronaviruses[59]
[28]
Senna obtusifolia L.
CN: Abejorra
Emodin (a)
Rheum spp.
CN: Rhubarb
Aloe spp.
CN: Aloe
Aloe emodin (a)[27]
Vaccinium spp.
CN: Blueberry, mortiño, Agráz, among others.
Anthocyanins (t), myricetin (n), gallic acid (u), stilbenoid resveratrol (s) and procyanidins (ai)SARS-CoV, MERS-CoV/(t) inhibits the production of NO and the secretion of TNF-α in macrophages induced by LPS-INF-γ caused by protocatechic acid, also show ACE inhibitory activity; (n) inhibits the coronavirus helicase protein by affecting the ATPase activity in vitro; Gallic acid decreases the secretion of MCP-1, ICAM-1, and VCAM-1 in endothelial cells; (s) partially mitigates induced cell death and reduces infectious viral replication; (v) possibly blocks the entry of cells through endocytosis[60]
[61]
[62]
[63]
Vitis vinifera L.
CN: Red grape
Zingiber officinale Rosc.
CN: Ginger
[6]-gingerol (ak)SARS-CoV-2/TMPRSS2 receptor blocking[64]
[65]
Table 3. Studies based on food for human consumption ACEi activity () and inhibition of AII to AT1R binding activity (ATRi). Individual results are given (ACE-%; ATR-%), based on studies by Patten et al., (2012) y Patten et al., (2016) [38,60].
Table 3. Studies based on food for human consumption ACEi activity () and inhibition of AII to AT1R binding activity (ATRi). Individual results are given (ACE-%; ATR-%), based on studies by Patten et al., (2012) y Patten et al., (2016) [38,60].
FamilyCommon Name of Plant with ACE and AT1R Inhibition Activities (%, %)
ActinidiaceaeGold kiwi (−0.2; 20.5), green kiwi (16.6; 2.5)
AgaricaceaeButton mushroom (12.5; 0.3)
AlliaceaeChives (23.2; 28.4), garlic (6.8; 27.4), leek (2.8; 42.7), onion (−1.2; 34.2), shallot (0.9; 11.5), red onion (−4.0; 31.8), spring onion (6.4; 53.3), white onion (−1.2; 18.8)
AmaranthaceaeSpinach (−0.7; 29.6)
ApiaceaeBlack carrot juice (91.1; 31.0), carrot (0.7; 5.0), coriander leaf (37.4; 56.6),
coriander seed (11.7; 16.4), fennel (−2.1; 15.2), parsley (8.2; 41.3)
ArecaceaeCoconut (11.8; −18.0)
AsparagaceaeAsparagus (35.1; 27.7)
AsteraceaeRadicchio (56; 43.5), red coral lettuce (31.5; 15.8), tarragon (32.1; 30.7)
AuricularaceaeWood Ear mushroom (13.1; 33.4)
BetulaceaeHazelnut (−9.8; 25.1)
BrassicaceaeBok choi (7.1; 30.4), broccoli (6.1; 0.2), brussel sprout (10.3; 1.2), Chinese broccoli (21.9; 38.7), Chinese cabbage (6.5; 28.8), choi sum (21.8; 2.6), red cabbage (24.6; 6.0), savoy cabbage (2.2; 52.1), watercress (18.7; 27.9), yellow mustard seed (5.2; −1.8)
ChenopodiaceaSilver beet (−1.0; 31.7), rainbow silver beet (−3.2; 10.2), beetroot (0.8; 6.2)
CombretaceaeKakadu plum (48.7; 0.0)
ConvolvulaceaeRed sweet potato (8.6; 16.5), sweet potato (4.9; 26.0)
CucurbitaceaeChoko (5.2; 3.4), choko skin (53.2; 14.0), cucumber (14.6; 40.8), pumpkin (3.3; 1.1), squash (4.3; 46.0), zucchini (16.0; 11.8)
Ericaceae Blueberry (−0.1; 43.3)
FabaceaeGreen bean (10.7; 27.2), green pea (−7.2; 9.3), lupin (−15.4; 12.1), Parafield lupin (−24.3; 7.6), peanut (1.4; −16.7)
FagaceaeChestnut (61.7; −5.6)
JuglandaceaePecan nut (0; 7.8), walnut (−10.9; 2.4)
LamiaceaeGreen basil (37.9; 26.4), purple basil (46.3; 11.0), Thai basil (69.5; 36.5), oregano (67.5; 55.7), rosemary (91.0; 55.7), sage (89.3; 68.2), thyme (87.4; 42.1)
LauraceaeAvocado (6.2; 43.4), bay leaf (34.9; 37.3), cinnamon (100.0; 54.4), Indian bay leaf (28.7; 0.4)
LythraceaePomegranate flesh (−6.2; 10.7)
MarasmiaceaeEnoki mushroom (4.8; −3.7), Shiitake mushroom (26.4; 11.8)
MeriplaceaeMaitake mushroom (67.0; 32.1)
MyrtaceaeClove (66.1; 30.8), cedar Bay cherry (63.8; 2.1), riberry (11.3; −12.1)
PoaceaeCorn (0; 27.8), lemongrass (5.0; 7.2)
PodacarpaceaeIllawarra plum (100; 7.0)
PolygonaceaeRhubarb (16.3; 8.5)
RosaceaeQuince (12.3; 11.1), raspberry (6.2; 6.2), strawberry (20.3; 3.5), red delicious apple (6.8; 1.5)
RubiaceaeColumbian dark coffee bean (63.416.0), Mocha coffee bean (56.7; 21.5)
RutaceaeDesert lemon (6.1; −0.6), green finger lime (11.5; 15.8), red finger lime (−6.3; 13.7), green citrus (14.8; 21.1), lemon skin (12.4; 7.9), lime (−16.4; 6.2), lime skin (47.1; 33.8), mandarin (0.2; 3.6), navel oranges (6.5; −3.9), orange skin (46.1; 7.8), red citrus (2.9; 40.1), red citrus skin (11.8; 17.4), ruby grapefruit (6.6; 14.9), Valencia orange (1.5; 5.4), yellow citrus (5.1; 18.6), yellow citrus skin (10.5; 7.3)
SaccharomycetaceaeBrewer’s yeast (31.8; −19.3)
SantalumQuandong (40.6; 8.5)
SolanaceaePotato (1.6; 16.6)
SterculiaceaeCocoa bean (81.2; 10.5)
TheaceaeEnglish breakfast black tea (88.8; 27.1), green tea (41.1; 12.4), Japanese green tea (100; 41.6), Madura black tea (100; 30.5)
PleurotaceaeOyster mushroom (35.9; 16.1), Honey Brown mushroom (14.6; 8.6)
VitaceaeMuscat grape (59.0; −2.8), white grape seed (100; 0.0), red grape skin (92.7; 14.4), Chambourcin grape (58.2; 10.6), Muscat Hamburg grape (73.5; -7.9), Cabinet Sauvignon grape (72.3; 0.0), Sun Muscat grape (59.0; −1.0), Concord grape (49.3; −3.3),
ZingiberaceaeCardamom (7.4; 1.2), ginger (9.9; 38.0), tumeric (15.1; −1.4)
Table 4. Plant species with potential for the bioprospecting of secondary metabolites located in Ecuador.
Table 4. Plant species with potential for the bioprospecting of secondary metabolites located in Ecuador.
FamilyPotential Species OriginRegionPotential Anti-Sars EffectReferences
BetulaceaeBirches (Betula spp.)IntroducedSierra regionAnticoagulants and antirheumatic[69]
BurseraceaePalo santo (Bursera graveolens Triana and Planch)NativeCoast and Sierra regionsAnti-inflammatory and antioxidant[70]
EricaceaeMortiño (Vaccinium floribundum Kunth)EndemicSierra regionAntioxidant[29]
EuphorbiaceaeCroto de monte (Croton rivinifolius Kunth)EndemicCoast regionAnticarcinogenic and antiviral[70]
Dog tongue (Euphorbia neriifolia L.)IntroducedCoast regiónAntitussive, antifungal and antitumor[69]
FabaceaeFrijolillo (Cassia tora L.)NativeCoast regiónAnticoagulants and anti-inflammatory[70]
White rain (Gliricidia brenningii Harms)NativeCoast regiónAntiherpetic and anticarcinogenic[29]
OrchidaceaeOrchid (Dendrobium spp.)IntroducedCoast, Sierra y Amazon regionsAntiviral[71]
Guayaquil Orchid (Encyclia angustiloba Schltr)EndemicCoast regiónAntiviral[71]
PolygonaceaeBloodroot (Polygonum arenastrum Boreau)IntroducedCoast y Amazon regionsAntiviral[59]
RubiaceaeCascarilla (Cinchona pubescens Vahl)NativeSierra regionFebrifuge, antiviral[72]
Cat’s claw (Uncaria tomentosa D. C.)NativeSierra and Amazon regionsAnti-inflammatory[72]
Colorado (Simira ecuadoriensis Standl)EndemicCoast regionFebrifuge and antiviral[72]
Crucita (Rosenbergiodendron formosum Fagerl.)NativeCoast regionFebrifuge and antiviral[72]
ScrophulariaceaeEscrofularia (Scrophularia spp.)IntroducedCoast regiónAnti-inflammatory and antimicrobial[69]
UrticaceaeNettle (Urtica urens L.)IntroducedSierra regionAntiviral [72]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Llivisaca-Contreras, S.A.; Naranjo-Morán, J.; Pino-Acosta, A.; Pieters, L.; Vanden Berghe, W.; Manzano, P.; Vargas-Pérez, J.; León-Tamariz, F.; Cevallos-Cevallos, J.M. Plants and Natural Products with Activity against Various Types of Coronaviruses: A Review with Focus on SARS-CoV-2. Molecules 2021, 26, 4099. https://doi.org/10.3390/molecules26134099

AMA Style

Llivisaca-Contreras SA, Naranjo-Morán J, Pino-Acosta A, Pieters L, Vanden Berghe W, Manzano P, Vargas-Pérez J, León-Tamariz F, Cevallos-Cevallos JM. Plants and Natural Products with Activity against Various Types of Coronaviruses: A Review with Focus on SARS-CoV-2. Molecules. 2021; 26(13):4099. https://doi.org/10.3390/molecules26134099

Chicago/Turabian Style

Llivisaca-Contreras, Susana A., Jaime Naranjo-Morán, Andrea Pino-Acosta, Luc Pieters, Wim Vanden Berghe, Patricia Manzano, Jeffrey Vargas-Pérez, Fabian León-Tamariz, and Juan M. Cevallos-Cevallos. 2021. "Plants and Natural Products with Activity against Various Types of Coronaviruses: A Review with Focus on SARS-CoV-2" Molecules 26, no. 13: 4099. https://doi.org/10.3390/molecules26134099

Article Metrics

Back to TopTop