Next Article in Journal
Mosquitoes from Europe Are Able to Transmit Snowshoe Hare Virus
Previous Article in Journal
Examining Chronic Inflammation, Immune Metabolism, and T Cell Dysfunction in HIV Infection
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Medicinal Plants against Viral Infections: A Review of Metabolomics Evidence for the Antiviral Properties and Potentials in Plant Sources

by
Wilson Bamise Adeosun
* and
Du Toit Loots
Human Metabolomics, North-West University, Private Bag X6001, Box 269, Potchefstroom 2531, South Africa
*
Author to whom correspondence should be addressed.
Submission received: 12 December 2023 / Revised: 22 January 2024 / Accepted: 29 January 2024 / Published: 31 January 2024

Abstract

:
Most plants have developed unique mechanisms to cope with harsh environmental conditions to compensate for their lack of mobility. A key part of their coping mechanisms is the synthesis of secondary metabolites. In addition to their role in plants’ defense against pathogens, they also possess therapeutic properties against diseases, and their use by humans predates written history. Viruses are a unique class of submicroscopic agents, incapable of independent existence outside a living host. Pathogenic viruses continue to pose a significant threat to global health, leading to innumerable fatalities on a yearly basis. The use of medicinal plants as a natural source of antiviral agents has been widely reported in literature in the past decades. Metabolomics is a powerful research tool for the identification of plant metabolites with antiviral potentials. It can be used to isolate compounds with antiviral capacities in plants and study the biosynthetic pathways involved in viral disease progression. This review discusses the use of medicinal plants as antiviral agents, with a special focus on the metabolomics evidence supporting their efficacy. Suggestions are made for the optimization of various metabolomics methods of characterizing the bioactive compounds in plants and subsequently understanding the mechanisms of their operation.

1. Introduction

The continuous emergence of viral diseases as a major challengeto global health warrants an intensified effort from a combined team of researchers and healthcare professionals. Continuous or regularly occurring pathogenic viral infections of pandemic proportions account for millions of deaths annually [1]. Although the development of conventional antiviral drugs is a significant stride toward curbing this trend, in the last decade, limitations have become increasingly conspicuous. The limitations include issues such as drug resistance [2], a narrow spectrum of efficacy [3], costs [4], and adverse side effects [5]. This has led to a resurgence of interest in the exploration of alternative treatment modalities from health stakeholders. Numerous chemical compounds found in natural sources are exclusive to plants, microbes, and marine life [6]. They provide important leads for drug discoveries and can potentially contribute to antiviral treatment/drug development [7,8,9]. Compounds derived from natural sources play a crucial role in drug discovery and the development of new antiviral treatments [7,10]. Medicinal plants have emerged as a promising frontier in the search for innovative antiviral therapies [11].
Throughout history, different cultures have sought healing in the potency of plants for the alleviation of pain and diseases. Their use in traditional medicine systems globally underscores their potential as a source of valuable antiviral compounds.
Plants respond to biotic and abiotic stress by synthesizing a large array of secondary metabolites with complex chemical combinations [12,13,14]. Although numerous studies have reported the activities and potentials of plants against viruses [15,16], it is important to validate traditional remedies through clinical trials [17,18].
The application of advanced analytical techniques to modern scientific research presents an opportunity for the exploration of plant-based compounds for their therapeutic effects. Metabolomics has emerged as an indispensable tool for the identification of different classes of antiviral secondary metabolites from plants over the last decade. These include various flavonoids, terpenoids, alkaloids, and polyphenols, each of which exhibit various therapeutic effects during different stages of the viral cycle (e.g., viral attachment, entry, viral replication, and release) [19]. These identified antiviral plant metabolites account for about a quarter of all drugs developed and used today [20,21].
The interplay between medicinal plants, viral infections, and metabolomics offers a multifaceted view of the potentials inherent in plants for the development of new antiviral therapeutics. This review highlights the role of metabolomics in the discovery and development of new antiviral bioactive compounds from medicinal plants. It also identifies the obvious gaps in current knowledge and offers comprehensive suggestions to future research directions.

2. Overview of Viral Infections

Viruses have been part of life since time immemorial. They are the smallest known agents of human infections, with a diameter that ranges between 20–200 nanometers [22]. Viruses are ubiquitous and can be found in animals, plants, humans, and other living organisms. All viruses are referred to as obligate intracellular pathogens since they cannot complete their life cycle without a living host [23]. Many of them are benign, non-pathogenic, and can even save lives. Bacteriophages, for instance, have been used as natural predators for therapeutic purposes, either exclusively in cases of failed conventional antibiotic therapy or in conjunction with antibiotics with satisfactory outcomes [24,25,26]. Some, however, do contribute to a considerable number of infections in humans [27,28]. Viral infections present a significant public health concern. Over the last few decades, many new viruses have emerged, with a significant number of them having deleterious effects on human health [29]. There are reportedly 26 virus families implicated in human diseases, with each exhibiting a different genomic structure, physiochemical properties, molecular processes, and morphology [30].
Viral infections are diverse and include: (1) sexually transmitted infections, including hepatitis B, HIV, herpes simplex virus (HSV), and human papillomavirus (HPV); (2) gastrointestinal infections, which lead to gastroenteritis caused by noroviruses, rotaviruses, adenoviruses, and sapoviruses; (3) zoonotic infections, caused by viruses that can be hosted by both animals and humans, e.g., Ebola, rabies, and hantaviruses; (4) hepatic infections, which can result in hepatitis, e.g., hepatitis A, B, C, D, and E and others including yellow fever and the Epstein-Barr virus; (5) Respiratory infections such as the common cold (caused by rhinoviruses), flu (caused by influenza viruses), COVID-19 (caused by the SARS-CoV-2 virus), and the respiratory syncytial virus (RSV) [30,31,32].
The most common of these viral diseases are those that affect the respiratory system [33]. A collaboration between researchers from several institutions and countries reported that respiratory diseases were the third leading cause of death worldwide between the years 1990 and 2019, beneath only cancer and cardiovascular diseases [34]. The influenza virus, which is the causative agent for the acute respiratory infection seasonal flu, remains one of the biggest threats to public health according to the World Health Organization (WHO). The organization gave an estimate of 290,000–650,000 flu-related deaths annually [35]. SARS-CoV-2 accounted for at least 14.9 million deaths directly or indirectly at the height of COVID-19 pandemic (between the years 2020 and 2021) according to WHO [36].
Viral infections are risk factors for other medical conditions, since they have the potential to weaken the immune system and induce an inflammatory response from the host cell [29,37]. Recent studies confirmed that oncogenic viruses implicated in the development of some cancers account for about 10% of all global cancer burden [38], and may also be responsible for long-term persistent infections [39]. Viral infections can also result in secondary bacterial infections, a condition in which infected patients are predisposed to health complications from bacterial sources due to a weakened immune system [40,41,42]. In the past, cardiovascular diseases have also been linked to viral infections [43]. In 1932, [44] reported that the peak period of an influenza pandemic in the United States was in direct proportion to an increase in heart diseases. Also, in the wake of the COVID-19 pandemic, there were many reports of heightened cases of cardiovascular diseases linked to the SARS-CoV-2 virus [45,46].
The unique nature of viruses and their ability to rapidly mutate, leading to an emerging pathogenesis and drug resistance, makes the current range of therapeutic and prophylactic options available for treating viral infections increasingly smaller [2,47,48,49]. Plants have been used for centuries as part of traditional medicine in the treatment of a variety of diseases, including various viral infections. Since the first attempt to screen over 200 plants for anti-influenza activity some seven decades ago [50], many additional studies have shown the vast potential of an enormous array of various medicinal plants across diverse geographical locations. Plants display antiviral activities and potentials for antiviral drug development, which can be used as a standalone treatment or as complementary therapeutic agents to conventional antiviral medicines [51,52,53,54,55,56].
Metabolomics, which is the study of all metabolites present in a biological system at a given time, has become a valuable tool for the identification and quantification of possible new therapeutic compounds from plants. A combination of techniques including Nuclear Magnetic Resonance (NMR) and advanced hyphenated mass spectrometry, are now central to metabolomics studies and are routinely applied to new biomarker identification.

3. A Brief History of the Use of Medicinal Plants against Viral Infections

Medicinal plant use against a variety of viral infections across various cultures dates to the dawn of human civilization. Traditional Chinese medicine, the Eber papyrus of Ancient Egypt, and the Ayurveda of India are among the oldest cultures practicing medicinal plant use with well documented manuscripts available for such [57,58,59]. Ancient traditional Chinese medicine has a history of about 3000 years or more. Writings based on this practice describing the use of plants for healing purposes are among the oldest medical writings of any culture [60]. Examples of plants used to treat viral infections in ancient China include ephedra (Ephedra sinica) for the treatment of the common cold [61], Andrographis paniculata for treating coughs, colds, and influenza [62], Camellia sinensis (green tea) which has confirmed efficacy against herpes, hepatitis B and C, and Epstein-Barr viruses [63,64,65]. The Egyptian papyrus contains descriptions of plant and natural product preparations against a plethora of diseases, including various viral infections [66]. One of the plants discussed is garlic (Allium sativum) [67], used for the treatment of respiratory catarrh, influenza, and recurring colds [68]. Echinacea (Echinacea purpurea), also described in the papyrus, has been reported to have efficacy against respiratory viral infections [69]. Ayurveda, a natural system of medicine with historical roots in India dating back three millennia, describes the plants Aegle marmelos, Ficus religiosa, and Azadirachta indica, among others, to have efficacy against a variety of viruses [57,70,71,72]. European herbal medicine and traditional African medicine also have long histories of using plants to combat various illnesses, including viral infections. Elderberry (Sambucus nigra), for instance, has been used in Europe for many years as a remedy for colds and flu [73], while plants like Sutherlandia frutescens and Artemisia afra, native to Africa, have reported potency against various viral infections [74,75]. The last few decades have seen a resurgence in the use of medicinal plants as an alternative source of treatments for various viral infections [76]. This is due in part to the rise in the prevalence of viral infections and growing concern about antibiotic resistance [2,77].

4. Mechanism of Actions of Antiviral Secondary Metabolites in Medicinal Plants

Plants’ secondary metabolites are organic compounds that are not directly involved in the growth, development, or reproduction of the plants. They are produced as a survival strategy against adverse conditions in the surrounding environment and to carry out important physiological tasks [12]. There are various criteria used in determining the classification of secondary metabolites in plants. and include chemical structure, composition of constituent elements, and how soluble they are in water or organic solvents. The most commonly accepted criterion however is their biosynthetic pathway [78]. Based on these pathways, three classes of secondary metabolites have been identified in medicinal plants: (1) alkaloids, (2) terpenoids and (3) phenolic compounds [79]. Each exhibits different phytochemical constituents and pharmacological effects against various viral agents [78,80]. Commonly reported mechanisms of action of plant-derived secondary metabolites against viruses include: (1) virus entry attachment [81,82], (2) inhibition of viral replication [19,80], (3) protein synthesis inhibition [83,84,85], (4) modulation of the host’s immune system [86], (5) modulation of cellular signaling pathways [87], and (6) direct virucidal activity [87,88].

4.1. Phenolics

Phenolic compounds are a diverse group of plant-derived organic molecules structurally characterized by at least one phenol group. They are the most widely distributed secondary metabolites in plants [89] and are synthesized as an adaptive response to unfavorable conditions [90]. Phenols are commonly found in all plant organs and are rich constituents of fruits, vegetables, beverages, cereals, and legumes [91]. Some well-known phenolic compounds with reported antiviral activities include flavonoids, tannins, and phenolic acids [92,93,94]. The specific mechanism of action of phenols against viruses depends on the type of phenolic compound and the virus targeted.
Phenols are well-known to inhibit viral infection of a target host cell in a variety of ways [95,96,97,98,99], including the disruption of the virus lipid bilayer envelopes [100,101] and preventing viral attachment to the host cell [97,102]. Furthermore, the inhibition of various viral protein activities results in the disruption of the viral life cycle, which ultimately prevents the multiplication and release of the virus [85,102,103].
Various phenols have also been shown to interfere with viral replication by binding to and subsequently inactivating various viral proteins or enzymes, and subsequently halt disease progression in the host cells [102,104,105]. Specific examples of compounds reported to interfere with viral replication include the tannin epigallocatechin-3-gallate (EGCG), a catechin derived from green tea [106]. EGCG demonstrates inhibition of the M2 protein of the influenza A virus by increasing IFN-λ2 expression in human lung epithelial BEAS-2B cells through the p38 mitogen-activated protein kinase signaling pathway [107]. EGCG has also been reported to inhibit the binding of HIV-1 envelope glycoprotein gp120 to the glycoprotein CD4 receptor found on an immune cell’s surface [108,109]. Four flavonoids, namely quercetin, baicalein, myricetin, and quercetagetin, have also previously been reported to inhibit HIV-1 enzyme reverse transcriptase and cellular DNA and RNA polymerases [110].
Sodium ferulate, another phenolic acid found abundantly in plants [111], inhibits the replication of influenza viruses via the activation of the toll-like receptors TLR7 and TLR9, and subsequently promotes transcription factor IRF7’s translocation into the host nucleus and the production of type-1 interferons [112]. Chicoric acid such as (caffeic acid derivative) and a number of dicaffeoylquinic acids have been reported to interfere with the enzyme integrase of HIV, an enzyme that is crucial to the integration of viral DNA into the host genome [113]. Gallic acid has also been shown to possess anti-HSV-2 properties, primarily through the inhibition of the virus’s attachment to the host cell, subsequently limiting the spread of the virus [114]. Furthermore, multiple antiviral defense mechanisms, including the inhibition of viral replication and cell to cell movement have been reportedly activated by salicylic acid [115]. The immunomodulatory effects of various phenolic compounds have also been previously shown to activate the prouction of various host cytokines and chemokines [116,117]. Lastly, a number of plant-derived phenolic compounds have been shown to induce apoptosis in an infected host, limiting the spread of viral infections [102,118,119,120].
Figure 1 schematically summarizes the antiretroviral effects of plant-derived phenolic compounds.

4.2. Alkaloids

Alkaloids, primarily but not exclusively present in plants, are a class of naturally occurring organic compounds containing at least one nitrogen atom. They can be found in many plant structures and have been reported to have pharmacological effects against various microbial diseases and viruses. Alkaloids have in particular been highlighted for their broad-spectrum activities against both DNA and RNA viruses [121,122]. Due to their importance, they have also been identified as the largest class of plants’ secondary metabolites investigated to date [122]. The most well-known plant alkaloids found in nature include cocaine, morphine, and quinine.
Alkaloids have been identified as important inhibitors of the flow of genetic information (from the DNA/RNA viral particle to protein synthesis) necessary to ensure the lifecycle of the virus. The antiviral activities of alkaloids, as proven by experimental evidence, primarily involve the inhibition of: (1) DNA and RNA replication, (2) RNA translation, (3) protein synthesis, (4) DNA intercalation, (5) enzymatic activities, (6) the translocation of the ribonucleoprotein complex, (7) DNA synthesis, and (8) protein synthesis. Many studies have reported the use of alkaloids for the treatment and prevention of viral infections [122,123,124]. Alkaloids can also regulate human immune mechanisms toward viral resistance by mediating the humoral immune response [125].
Figure 2 below is a schematic representation of the antiviral effects of alkaloids.

4.3. Terpenoids

Terpenoids are a large group of diverse organic products that are ubiquitous in nature. They exist in six categories namely: hemiterpenes, monoterpenes, sesqui-terpenes, diterpenes, sesterpenes, triterpenes, and tetra-terpenoids [126]. As an essential component of all living cells, they are products of both primary and secondary cellular metabolism [127]. Terpenoids are mostly present in the leaves and fruits of higher plants, where they sometimes contribute to their vibrant colors. They are also highly volatile and combustible organic compounds [128]. As aromatic metabolites, they are largely responsible for the flavor and fragrance of plants [129]. Terpenoids are of special interest to medical chemists because of their significant pharmacological potential [130]. Common terpenoids include citral, menthol camphor, and salvinorin A.
Various terpenoids have been shown to possess promising antimicrobial and antiviral properties [131]. Those with reported antiviral properties include glycyrrhizin, an important antiviral chemical compound found in the roots of the licorice plant (Glycyrrhiza glabra) [132]. Historical sources from China [133], India [134], and parts of Europe [135] make references to the use of glycyrrhizin in the treatment of viral respiratory tract and liver infections caused by hepatitis. Ref. [136] reported the immunomodulatory activities of glycyrrhizin due to the induction of interferon gamma, and [137] described its anti-inflammatory mechanisms. Glycyrrhizin furthermore inhibits replication of severe acute respiratory syndrome associated with corona virus (SARS-Chikungunya virus (CV)) infection and prevents the adsorption and penetration of the virus [138]. Isoborneol, a monoterpene present in a variety of different essential oils, totally inhibits replication of the herpes simplex virus (HSV-1), and that at a concentration of only 0.06% [139]. Additionally, β-pinene and limonene monoterpenes present in various essential oils, showed high anti-HSV-1 activity, and function by reducing the viral infectivity by 100%, by directly interfering with free viral particles [140]. Many studies have also indicated that celastrol, a pentacyclic triterpenoid, inhibits the replication of the dengue virus [141], human immunodeficiency virus [142], and hepatitis C virus [143].
Ref. [144] reported that the extracted pigment of carotenoids of Natrialba sp. M6, a tetraterpene isolated from Wadi El-Natrun in Egypt, exhibited antiviral potency against Hepatitis C Virus (HCV) and HBV. The RNA and DNA polymerase enzymes responsible for the amplification of the virus in both HCV and HBV were inhibited, thereby suppressing the viruses’ replication. Molecular docking simulation was also used to predict the antiviral effects of two marine carotenoids (specifically fucoxanthin and siphonaxanthin) against SARS-CoV-2. The effects were later confirmed by in vitro studies [145].
Figure 3 shows a schematic summary of the antiviral activities of terpenoids.

5. Applications of Metabolomics in Plant Antiviral Research

Metabolomics involves the comprehensive study of small molecules known as metabolites in a living system or biological sample. It can give broad insight into the identity and detailed information on the chemical fingerprints of metabolites and the metabolic processes that occur within a biological system [146]. Metabolomics is broadly categorized into two types, (1) targeted and (2) untargeted metabolomics. In the targeted approach, the goal is not the identification of all metabolites, but the quantitative measurement of a specific metabolite or metabolite group that has been previously identified and characterized. Untargeted metabolomics deals with a comprehensive identification and quantification of all detectable metabolites present in a biological sample [147].
Advances in metabolomic techniques now play a significant role in the discovery of antiviral compounds in plants, as evidenced by relevant recent publications. The possibilities for the comprehensive analysis of the complete set of compounds in plants using advanced analytical techniques are endless. Many studies have reported the use of metabolomics as an essential tool for the targeted screening of plant secondary metabolites [148,149]. The many advantages that metabolomics presents make it an essential and indispensable technique in current plant antiviral research.
Arguably the most important application of metabolomics in plant antiviral research is in the identification and characterization of the plant metabolome. Metabolomics, when used for this, can provide a detailed snapshot of a plant’s metabolomic profile, with information on both the identification and quantification of the metabolites present within the metabolome. There is an incredible diversity of secondary metabolites exhibited by plants, many of which have bioactive compounds [150,151,152]. The use of metabolomics has additionally assisted in identifying a wide range of metabolites in plants across different geographical locations and seasons, including those with antiviral potential. The specific bioactive compounds responsible for the antiviral activity can then be isolated and further studied for potential therapeutic use.
Metabolomics can also be used for mechanistic or toxicology studies of the isolated plant antiviral compounds in vivo [153,154]. Since metabolites are downstream products of cellular metabolism [153], the identification, quantification, and characterization of these compounds in vivo, in the presence of the isolated plant metabolite/drug show the metabolic pathways that are altered in response to the plant compound/drug’s antiviral activity in the culture or host.
There are many complex metabolic interactions that occur within the plant system. Antiviral compounds can either act alone or in synergy with other compounds to carry out their activities. In studying the synergistic effect of more than one compound on microbes, stronger bioactivities have been reported using metabolomics [155,156,157,158]. The metabolomics approach is therefore promising, as a useful strategy for the study of complex interactions between plant metabolites [159,160].
Metabolomics has also been applied to investigating the effect of the different seasons on the chemical profiles and biological activities of plants [161,162]. Ref. [163] used metabolomics to study the influence of seasonal change on the anti-HSV1 properties of Helichrysum aureonitens and reported a correlation between plants harvested in spring and better antiviral activity. A targeted metabolomics study conducted to determine the effect of seasonal change on the chlorogenic acids content of H. aureonitens further revealed an association between water availability and the production of different isomers of chlorogenic acids [163]. This knowledge is important in determining the optimal season for plant collection to yield better antiviral efficacy.
Metabolomics can also be used for studying those metabolic pathways involved in plant-pathogen interaction [159,164,165]. These adapted metabolic pathways can also be determined by analyzing those changes in metabolome during a viral infection. This is important in understanding the mechanism by which plants respond to viral infections and can provide insight into the identification of useful metabolites that may be developed as therapeutic candidates for antiviral intervention.
A summary of such studies is given in Table 1 below.

6. Steps for the Discovery of Antiviral Compounds in Plant Metabolomics Studies

The discovery of compounds with antiviral activities in plants using metabolomics follows a systematic approach, combining different steps and methodologies. The choice of plant selected for metabolomics studies is often predicated upon prior knowledge of the plant’s antiviral properties, either from oral tradition or previous studies. Different plant parts such as stems, roots, leaves, and flowers, are often collected from the wild or from a cultivated environment. Due to the different distribution or constituency of various bioactive compounds in different parts of the plants [178,179,180], a thorough knowledge from literature or indigenous knowledge systems is required to determine the specific plant parts which need to be harvested for the specific application/investigation.
Post-harvest, sample preparation often involves cleaning, drying, and pulverizing/homogenizing the plant materials, with the subsequent extraction of the bioactive metabolites using various solvents of varying polarities depending on the compound of interest. Phytochemical investigations typically use a solvent mixture for the extraction of both polar and non-polar compounds, which comprises a mixture of alcohols (methanol or ethanol) and water [181,182,183,184,185,186]. Extraction may be followed by sample preparation for NMR, GC-MS, or LC-MS analysis, depending on the aim of the studies and the nature of the targeted metabolites.
Sample preparation for subsequent mass spectrometry analyses is generally more complex than that required for NMR, often requiring prior derivatization of the extracted compounds to improve detection of a wider range of analytes [187]. However, the sensitivity of hyphenated mass spectrometry techniques far surpasses that of NMR. NMR sample preparation generally involves the addition of deuterated methanol (CH3OH-d4), potassium dihydrogen sulfate (which acts as a buffer in a deuterium water (D2O) solvent) and trimethyl silane propionic acid (TSP) as an internal reference to detect and correct possible chemical shift during the analyses of multiple samples [188,189]. Data when using either approach are generated in the form of NMR spectra and mass spectrometer chromatograms.
There are a number of software choices for multivariate data analysis and data processing, including MestReNova [190], MetaboLab [191], the Soft independent modeling by class analogy (SIMCA) [192], Metaboanalyst [193], and Chenomx [194]. Software exclusively designed for mass spectrometry include XCMS (one of the most popular software for MS) [195], MZmine [196], Automated Data Analysis Pipeline (ADAP) [196], Mass Spectrometry-Data Independent Analysis software (MS-DIAL) [197] and METLIN [198].
Multivariate data analysis and data processing are essential steps and precursors to the characterization of bioactive compounds in medicinal plants. The chemical structure of potential antiviral bioactive principles can then be further and more accurately annotated by advanced techniques such as 2D NMR. This is a versatile technique that provides more information about a compound. When used in tandem with relevant databases such as plant metabolome database and reference libraries [199,200], it is helpful in the elucidation of small organic molecules’ structures.
Subsequently, a bioguided isolation study to determine the antiviral activity of compounds of interest is carried out, increasing the probability of isolating compounds with high bioactive activity [201,202]. A knowledge of natural product chemistry is also invaluable in investigating small organic molecules with medicinal properties from plant sources. Thin-layer chromatography (TLC) and column chromatography are popular choices for compound isolation owing to their convenience and relatively low cost of operation [203]. TLC (or its more sophisticated and improved form, called high-performance thin-layer chromatography) are commonly used techniques that provides a fast and comprehensive overview of most of the compounds present in a plant extract, supports the identification of target compounds in a mixture, and is effective in the isolation of analytes in a sample mixture [204,205]. The basic principle involves the movement of a compound mixture along the mobile phase (solvent), through the stationary phase, where separation occurs depending on the adsorption capacities of the compounds in the mixture. The aforementioned column chromatography (or its highly-improved automated form-high-performance liquid chromatography) is a technique similar to TLC. It works on the same principle, except that instead of the stationary phase on a thin layer in TLC, the solid in the stationary phase is loaded into a long glass column [204,206,207,208].
When determining the antiviral activities of plants, it is important to conduct an assay to evaluate the antiviral potential of the plant of interest to inhibit viral replication, inhibit viral binding to host cell receptors, or interfere with the viral life cycle. An assay commonly used for this purpose is the cytopathic effect inhibition assay (CPE). This involves infecting a host cell culture with the virus, then treating the cells with the plant extract. The cytopathic effect on the cell (cell damage caused by the viral infection) can then be determined [209,210]. Another often-used assay is the plaque reduction assay. Here, an assessment is made of a plant extract’s ability to inhibit viral replication. Cultured cells are seeded to a surface to form a confluent monolayer and infected with a known number of viral particles. The viruses replicate and form plaques while plant extracts are added at different concentrations. Fewer plaques suggest antiviral activity of the plant extract [211,212,213].
Preclinical trials are studies conducted on animal models to evaluate the effect of a test compound or drug for its therapeutic potential [214]. Such trials can be performed in vivo, in vitro, ex vivo, or in silico to gather information regarding the safety and efficacy of the test compound before performing clinical trials on humans [215], which determines the effect of drugs or bioactive compounds on human health outcomes [7,216,217,218]. These medical studies follow specific protocols, including the recruitment of healthy volunteers and patients [219]. With respect to the clinical trials of plant-based antiviral compounds, the trials pass through a number of phases, including initially testing the plant-derived compound/drug on healthy volunteers to determine possible side effects and other safety concerns [220], before testing these on infected or disease patients for drug efficacy.

7. Future Directions for the Application of Metabolomics to the Development of Antiviral Therapies from Plant Sources

In the just about 20 years since the introduction of metabolomics [221], some key advancements and significant progress have been made in the field of medicinal plant and natural product research. There remains an immense potential for the full exploration of metabolomics in medicinal plant research and in the search and development of new therapeutic agents. As [222] noted, although many studies have been conducted on plants to determine their effects on viruses, most studies do not isolate and identify active compounds responsible for the antiviral potentials of the tested plants. This is further confirmed by the limited number of studies that reached the level of compound identification and isolation in the last couple of years, as listed in Table 1 above. Some studies reported good plant activities against viruses and even went as far as isolating compounds, but did not elucidate the compound names [177,223]. This presents difficulties in characterizing the bioactive principles responsible for the biological activity of the plant. Taking advantage of advances in metabolomics techniques to identify, isolate, and characterize compounds that have both pharmacological effects and good biological activity will prove beneficial in the development of antiviral leads as part of an effort toward viral eradication. Furthermore, a detailed search of many scientific databases reveals a lack of information on the biosynthetic pathways responsible for producing the plant bioactive compounds. Future studies should harness metabolomics for elucidating the biosynthetic pathways responsible for the formation of identified antiviral metabolites by these plants [224]. This is crucial in optimizing plant cultivation and incorporating genetic engineering strategies for the enhancement of compound synthesis with antiviral bioactivity [221,225].
The emergence of systems biology integrates high throughput data generation and analysis from many platforms to understand complex interactions between different levels of organization in a biological system [226,227]. Omics technology, an indispensable molecular technique in systems biology, incorporates genomics, transcriptomics, proteomics, and metabolomics techniques to foster a better understanding of biological processes [224,228]. The application of a systems biology paradigm in plant studies will provide a more holistic view of the antiviral potentials in medicinal plants.

Author Contributions

W.B.A.: Idea conceptualization and write-up. D.T.L.: Supervision, review, and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

No statement from the institutional review board was required for the review.

Informed Consent Statement

Not applicable.

Data Availability Statement

No research data were generated in this review.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Baker, R.E.; Mahmud, A.S.; Miller, I.F.; Rajeev, M.; Rasambainarivo, F.; Rice, B.L.; Takahashi, S.; Tatem, A.J.; Wagner, C.E.; Wang, L.F.; et al. Infectious Disease in an Era of Global Change. Nat. Rev. Microbiol. 2022, 20, 193–205. [Google Scholar] [CrossRef]
  2. Strasfeld, L.; Chou, S. Antiviral Drug Resistance: Mechanisms and Clinical Implications. Infect. Dis. Clin. N. Am. 2010, 24, 413–437. [Google Scholar] [CrossRef]
  3. Chitalia, V.C.; Munawar, A.H. A Painful Lesson from the COVID-19 Pandemic: The Need for Broad-Spectrum, Host-Directed Antivirals. J. Transl. Med. 2020, 18, 390. [Google Scholar] [CrossRef]
  4. Fleßa, S.; Marschall, P. Socio-Economic Impact of Antiviral Intervention; Springer: Berlin/Heidelberg, Germany, 2009. [Google Scholar]
  5. Morris, D.J. Adverse Effects and Drug Interactions of Clinical Importance with Antiviral Drugs. Drug Saf. 1994, 10, 281–291. [Google Scholar] [CrossRef]
  6. Ntie-Kang, F.; Svozil, D. An Enumeration of Natural Products from Microbial, Marine and Terrestrial Sources. Phys. Sci. Rev. 2020, 5, 20180121. [Google Scholar] [CrossRef]
  7. Atanasov, A.G.; Zotchev, S.B.; Dirsch, V.M.; Orhan, I.E.; Banach, M.; Rollinger, J.M.; Barreca, D.; Weckwerth, W.; Bauer, R.; Bayer, E.A.; et al. Natural Products in Drug Discovery: Advances and Opportunities. Nat. Rev. Drug Discov. 2021, 20, 200–216. [Google Scholar] [CrossRef]
  8. Ji, H.-F.; Li, X.-J.; Zhang, H.-Y. Natural Products and Drug Discovery Can Thousands of Years of Ancient Medical Knowledge Lead Us to New and Powerful Drug Combinations in the Fight against Cancer and Dementia? EMBO Rep. 2009, 10, 194–200. [Google Scholar] [CrossRef] [PubMed]
  9. Dias, D.A.; Urban, S.; Roessner, U. A Historical Overview of Natural Products in Drug Discovery. Metabolites 2012, 2, 303–336. [Google Scholar] [CrossRef] [PubMed]
  10. Lin, L.T.; Hsu, W.C.; Lin, C.C. Antiviral Natural Products and Herbal Medicines. J. Tradit. Complement. Med. 2014, 4, 24–35. [Google Scholar] [CrossRef] [PubMed]
  11. Kumar Shakya, A.; Arvind Kumar Shakya, C. Medicinal Plants: Future Source of New Drugs. Int. J. Herb. Med. 2016, 4, 59–64. [Google Scholar] [CrossRef]
  12. Guerriero, G.; Berni, R.; Muñoz-Sanchez, J.A.; Apone, F.; Abdel-Salam, E.M.; Qahtan, A.A.; Alatar, A.A.; Cantini, C.; Cai, G.; Hausman, J.F.; et al. Production of Plant Secondary Metabolites: Examples, Tips and Suggestions for Biotechnologists. Genes 2018, 9, 309. [Google Scholar] [CrossRef]
  13. Isah, T. Stress and Defense Responses in Plant Secondary Metabolites Production. Biol. Res. 2019, 52, 39. [Google Scholar] [CrossRef]
  14. Ramakrishna, A.; Ravishankar, G.A. Influence of Abiotic Stress Signals on Secondary Metabolites in Plants. Plant Signal Behav. 2011, 6, 1720–1731. [Google Scholar]
  15. TePaske, M.R.; Gloer, J.B. Tubingensin A: An Antiviral Carbazole Alkaloid from the Sclerotia of Aspergillus Tubingensis. J. Org. Chem. 1989, 54, 4743–4746. [Google Scholar] [CrossRef]
  16. Verpoorte, R. Exploration of Nature’s: The Role of secondary Metabolites as leads in Drug Development. Drug Discov. Today 1998, 5, 232–238. [Google Scholar] [CrossRef]
  17. Parveen, A.; Parveen, B.; Parveen, R.; Ahmad, S. Challenges and Guidelines for Clinical Trial of Herbal Drugs. J. Pharm. Bioallied Sci. 2015, 7, 329–333. [Google Scholar] [PubMed]
  18. Owen, L.; Laird, K.; Shivkumar, M. Antiviral Plant-Derived Natural Products to Combat RNA Viruses: Targets throughout the Viral Life Cycle. Lett. Appl. Microbiol. 2022, 75, 476–499. [Google Scholar] [CrossRef] [PubMed]
  19. Ghildiyal, R.; Prakash, V.; Chaudhary, V.K.; Gupta, V.; Gabrani, R. Phytochemicals as Antiviral Agents: Recent Updates. In Plant-Derived Bioactives: Production, Properties and Therapeutic Applications; Springer: Singapore, 2020; pp. 279–295. ISBN 9789811517617. [Google Scholar]
  20. Yeshi, K.; Crayn, D.; Ritmejerytė, E.; Wangchuk, P. Plant Secondary Metabolites Produced in Response to Abiotic Stresses Has Potential Application in Pharmaceutical Product Development. Molecules 2022, 27, 313. [Google Scholar] [CrossRef]
  21. Hussein, R.A.; El-Anssary, A.A. Plants Secondary Metabolites: The Key Drivers of the Pharmacological Actions of Medicinal Plants. In Herbal Medicine; IntechOpen: London, UK, 2019. [Google Scholar]
  22. Louten, J. Virus Structure and Classification. In Essential Human Virology; Elsevier: Amsterdam, The Netherlands, 2016; pp. 19–29. [Google Scholar]
  23. Gelderblom, H.R. Structure and Classification of Viruses; University of Texas Medical Branch at Galveston: Galveston, TX, USA, 1996. [Google Scholar]
  24. Hatfull, G.F.; Dedrick, R.M.; Schooley, R.T. Annual Review of Medicine Phage Therapy for Antibiotic-Resistant Bacterial Infections. Annu. Rev. Med. 2022, 73, 197–211. [Google Scholar] [CrossRef] [PubMed]
  25. Schooley, R.T.; Biswas, B.; Gill, J.J.; Hernandez-Morales, A.; Lancaster, J.; Lessor, L.; Barr, J.J.; Reed, S.L.; Rohwer, F.; Benler, S.; et al. Development and Use of Personalized Bacteriophage-Based Therapeutic Cocktails to Treat a Patient with a Disseminated Resistant Acinetobacter Baumannii Infection. Antimicrob. Agents Chemother. 2017, 61, e00954-17. [Google Scholar] [CrossRef]
  26. Chan, B.K.; Turner, P.E.; Kim, S.; Mojibian, H.R.; Elefteriades, J.A.; Narayan, D. Phage Treatment of an Aortic Graft Infected with Pseudomonas aeruginosa. Evol. Med. Public Health 2018, 2018, 60–66. [Google Scholar] [CrossRef]
  27. Janeway, C.A.; Medzhitov, R. Innate Immune Recognition. Annu. Rev. Immunol. 2002, 20, 197–216. [Google Scholar] [CrossRef]
  28. Cohen, F.S. How Viruses Invade Cells. Biophys. J. 2016, 110, 1028–1032. [Google Scholar] [CrossRef]
  29. Sudhan, S.S.; Sharma, P. Human Viruses: Emergence and Evolution. In Emerging and Reemerging Viral Pathogens: Volume 1: Fundamental and Basic Virology Aspects of Human, Animal and Plant Pathogens; Elsevier: Amsterdam, The Netherlands, 2019; pp. 53–68. ISBN 9780128194003. [Google Scholar]
  30. Siegel, R.D. Classification of Human Viruses. In Principles and Practice of Pediatric Infectious Diseases; Elsevier Inc.: Amsterdam, The Netherlands, 2017; pp. 1044–1048. ISBN 9780323401814. [Google Scholar]
  31. Siegel, D.; Hui, H.C.; Doerffler, E.; Clarke, M.O.; Chun, K.; Zhang, L.; Neville, S.; Carra, E.; Lew, W.; Ross, B.; et al. Discovery and Synthesis of a Phosphoramidate Prodrug of a Pyrrolo[2,1-f][Triazin-4-Amino] Adenine C-Nucleoside (GS-5734) for the Treatment of Ebola and Emerging Viruses. J. Med. Chem. 2017, 60, 1648–1661. [Google Scholar] [CrossRef]
  32. Mathekga, A.D.M.; Meyer, J.J.M.; Horn, M.M.; Drewes, S.E. An Acylated Phloroglucinol with Antimicrobial Properties from Helichrysum Caespititium. Phytochemistry 2000, 53, 93–96. [Google Scholar] [CrossRef]
  33. Boncristiani, H.F. Respiratory Viruses Defining Statement Introduction Human Respiratory Syncytial Virus Human Parainfluenza Viruses Human Metapneumovirus Rhinovirus Respiratory Adenoviruses Human Coronaviruses Unrelated to SARS SARS Coronavirus Human Bocavirus Further Reading, 3rd ed.; Book-Encyclopedia of Microbiology; Academic Press: Cambridge, MA, USA, 2009; pp. 500–518. [Google Scholar]
  34. Dattani, S.; Spooner, F.; Ritchie, H.; Roser, M. Causes of Death; Global Change Data Lab: Oxford, UK, 2018. [Google Scholar]
  35. WHO. WHO Launches New Global Influenza Strategy; WHO: Geneva, Switzerland, 2019. [Google Scholar]
  36. WHO. Methods for Estimating the Excess Mortality Associated with the COVID-19 Pandemic; WHO: Geneva, Switzerland, 2023. [Google Scholar]
  37. Pawlotsky, J.M. Pathophysiology of Hepatitis C Virus Infection and Related Liver Disease. Trends Microbiol. 2004, 12, 96–102. [Google Scholar] [CrossRef] [PubMed]
  38. Plummer, M.; de Martel, C.; Vignat, J.; Ferlay, J.; Bray, F.; Franceschi, S. Global Burden of Cancers Attributable to Infections in 2012: A Synthetic Analysis. Lancet Glob. Health 2016, 4, e609–e616. [Google Scholar] [CrossRef] [PubMed]
  39. Green, M. Oncogenic Viruses. Annu. Rev. Biochem. 1970, 39, 701–756. [Google Scholar] [CrossRef]
  40. MacIntyre, C.R.; Chughtai, A.A.; Barnes, M.; Ridda, I.; Seale, H.; Toms, R.; Heywood, A. The Role of Pneumonia and Secondary Bacterial Infection in Fatal and Serious Outcomes of Pandemic Influenza a(H1N1)Pdm09. BMC Infect. Dis. 2018, 18, 637. [Google Scholar] [CrossRef]
  41. Morris, D.E.; Cleary, D.W.; Clarke, S.C. Secondary Bacterial Infections Associated with Influenza Pandemics. Front. Microbiol. 2017, 8, 1041. [Google Scholar] [CrossRef] [PubMed]
  42. Dunn, C.; Brunetto, M.; Reynolds, G.; Christophides, T.; Kennedy, P.T.; Lampertico, P.; Das, A.; Lopes, A.R.; Borrow, P.; Williams, K.; et al. Cytokines Induced during Chronic Hepatitis B Virus Infection Promote a Pathway for NK Cell-Mediated Liver Damage. J. Exp. Med. 2007, 204, 667–680. [Google Scholar] [CrossRef] [PubMed]
  43. Kawana, R. Viral and Other Myocarditis Cardiovascular Diseases Due to Viruses. Heart Vessel. 1985, 1, 91–96. [Google Scholar] [CrossRef] [PubMed]
  44. Collins, S.D. Excess Mortality from Causes Other than Influenza and Pneumonia during Influenza Epidemics. Public Health Rep. 1932, 47, 2159–2179. [Google Scholar] [CrossRef]
  45. Bekkering, S.; Burgner, D. Viruses and Cardiovascular Disease: From Bad to Worse. Nat. Cardiovasc. Res. 2022, 1, 601–602. [Google Scholar] [CrossRef]
  46. Schultheiss, H.P.; Baumeier, C.; Pietsch, H.; Bock, C.T.; Poller, W.; Escher, F. Cardiovascular Consequences of Viral Infections: From COVID to Other Viral Diseases. Cardiovasc. Res. 2021, 117, 2610–2623. [Google Scholar] [CrossRef]
  47. Rouse, B.T.; Lukacher, A.E. Some Unmet Challenges in the Immunology of Viral Infections. Discov. Med. 2010, 10, 363. [Google Scholar] [PubMed]
  48. Wong, J.P.; Viswanathan, S.; Wang, M.; Sun, L.Q.; Clark, G.C.; D’elia, R.V. Current and Future Developments in the Treatment of Virus-Induced Hypercytokinemia. Future Med. Chem. 2017, 9, 169–178. [Google Scholar] [CrossRef]
  49. Moghadas, S.M.; Bowman, C.S.; Röst, G.; Wu, J. Population-Wide Emergence of Antiviral Resistance during Pandemic Influenza. PLoS ONE 2008, 3, e1839. [Google Scholar] [CrossRef]
  50. H Chantrill, B.B.; Coulthard, C.E.; Dickinson, L.; Inkley, G.W.; Morris, W.; Pyle, A.H. The Action. of Plant Extracts on a Bacteriophage of Pseudornonas Pyocyanea and on Influenza A Virus. Microbiology 1952, 6, 74–84. [Google Scholar]
  51. Thomas, E.; Stewart, L.E.; Darley, B.A.; Pham, A.M.; Esteban, I.; Panda, S.S. Plant-Based Natural Products and Extracts: Potential Source to Develop New Antiviral Drug Candidates. Molecules 2021, 26, 6197. [Google Scholar] [CrossRef]
  52. Welz, A.N.; Emberger-Klein, A.; Menrad, K. Why People Use Herbal Medicine: Insights from a Focus-Group Study in Germany. BMC Complement. Altern. Med. 2018, 18, 92. [Google Scholar] [CrossRef]
  53. Chattopadhyay, D.; Mukherjee, H.; Bag, P.; Ghosh, S.; Samanta, A.; Sekhar, C. Ethnomedicines in Antiviral Drug Discovery. Int. J. Biomed. Pharm. Sci. 2009, 3, 1–25. [Google Scholar]
  54. Mukhtar, M.; Arshad, M.; Ahmad, M.; Pomerantz, R.J.; Wigdahl, B.; Parveen, Z. Antiviral Potentials of Medicinal Plants. Virus Res. 2008, 131, 111–120. [Google Scholar] [CrossRef]
  55. Behl, T.; Rocchetti, G.; Chadha, S.; Zengin, G.; Bungau, S.; Kumar, A.; Mehta, V.; Uddin, M.S.; Khullar, G.; Setia, D.; et al. Phytochemicals from Plant Foods as Potential Source of Antiviral Agents: An Overview. Pharmaceuticals 2021, 14, 381. [Google Scholar] [CrossRef]
  56. Bachar, S.C.; Mazumder, K.; Bachar, R.; Aktar, A.; Al Mahtab, M. A Review of Medicinal Plants with Antiviral Activity Available in Bangladesh and Mechanistic Insight into Their Bioactive Metabolites on SARS-CoV-2, HIV and HBV. Front. Pharmacol. 2021, 12, 732891. [Google Scholar] [CrossRef]
  57. Mukherjee, A.; Banerjee, M.; Mandal, V.; Shukla, A.C.; Mandal, S.C. Modernization of Ayurveda: A Brief. Overview of Indian. Initiatives. Nat. Prod. Commun. 2013, 9, 1934578X1400900239. [Google Scholar] [CrossRef]
  58. Stiefel, M.; Shaner, A.; Schaefer, S.D. The Edwin Smith Papyrus: The Birth of Analytical Thinking in Medicine and Otolaryngology. Laryngoscope 2006, 116, 182–188. [Google Scholar] [CrossRef] [PubMed]
  59. Chung, V.C.H.; Ma, P.H.X.; Lau, C.H.; Wong, S.Y.S.; Yeoh, E.K.; Griffiths, S.M. Views on Traditional Chinese Medicine amongst Chinese Population: A Systematic Review of Qualitative and Quantitative Studies. Health Expect. 2014, 17, 622–636. [Google Scholar] [CrossRef] [PubMed]
  60. Reid, D.P. The Shambhala Guide to Traditional Chinese Medicine, 1st ed.; Shambhala: Boulder, CO, USA, 1996. [Google Scholar]
  61. Bensky, D.; Clavey, S.; Stõger, E.; Lai, L. Materia Medica 3rd Edition Compiled and Translated By; Eastland Press Publisher: Seattle, DC, USA, 2004. [Google Scholar]
  62. Fashner, J.; Ericson, K.; Werner, S.; Joseph, S. Treatment of the Common. Cold in Children and Adults. Am. Fam. Phys. 2012, 86, 153–159. [Google Scholar]
  63. Song, J.M.; Seong, B.L. Tea Catechins as a Potential Alternative Anti-Infectious Agent. Expert. Rev. Anti Infect. Ther. 2007, 5, 497–506. [Google Scholar] [CrossRef] [PubMed]
  64. Xu, J.; Wang, J.; Deng, F.; Hu, Z.; Wang, H. Green Tea Extract and Its Major Component Epigallocatechin Gallate Inhibits Hepatitis B Virus In Vitro. Antivir. Res. 2008, 78, 242–249. [Google Scholar] [CrossRef]
  65. Tran, J. Green Tea: A Potential Alternative Anti-Infectious Agent Catechins and Viral Infections. Adv. Anthropol. 2013, 3, 198–202. [Google Scholar] [CrossRef]
  66. Aboelsoud, N.H. Herbal Medicine in Ancient Egypt. J. Med. Plants Res. 2010, 4, 082–086. [Google Scholar]
  67. Rivlin, R.S. Recent Advances on the Nutritional Effects Associated with the Use of Garlic as a Supplement Historical Perspective on the Use of Garlic 1,2. J. Nutr. 2001, 131, 951S–954S. [Google Scholar] [CrossRef]
  68. Anadón, A.; Martínez-Larrañaga, M.R.; Ares, I.; Martínez, M.A. Interactions between Nutraceuticals/Nutrients and Therapeutic Drugs. In Nutraceuticals: Efficacy, Safety and Toxicity; Elsevier Inc.: Amsterdam, The Netherlands, 2016; pp. 855–874. ISBN 9780128021477. [Google Scholar]
  69. Hudson, J.; Vimalanathan, S. Echinacea-A Source of Potent Antivirals for Respiratory Virus Infections. Pharmaceuticals 2011, 4, 1019–1031. [Google Scholar] [CrossRef]
  70. Singh, D.; Singh, B.; Goel, R.K. Traditional Uses, Phytochemistry and Pharmacology of Ficus Religiosa: A Review. J. Ethnopharmacol. 2011, 134, 565–583. [Google Scholar] [CrossRef] [PubMed]
  71. Alzohairy, M.A. Therapeutics Role of Azadirachta Indica (Neem) and Their Active Constituents in Diseases Prevention and Treatment. Evid.-Based Complement. Altern. Med. 2016, 2016, 7382506. [Google Scholar] [CrossRef] [PubMed]
  72. Manandhar, B.; Paudel, K.R.; Sharma, B.; Karki, R. Phytochemical Profile and Pharmacological Activity of Aegle Marmelos Linn. J. Integr. Med. 2018, 16, 153–163. [Google Scholar] [CrossRef] [PubMed]
  73. Helman, C.G. Feed a Cold, Starve a Fever—Folk Models of Infection in an English Suburban Community, and Their Relation to Medical Treatment. Cult. Med. Psychiatry 1978, 2, 107–137. [Google Scholar] [CrossRef] [PubMed]
  74. Harnett, S.M.; Oosthuizen, V.; Van De Venter, M. Anti-HIV Activities of Organic and Aqueous Extracts of Sutherlandia Frutescens and Lobostemon Trigonus. J. Ethnopharmacol. 2005, 96, 113–119. [Google Scholar] [CrossRef] [PubMed]
  75. Liu, N.Q.; Van der Kooy, F.; Verpoorte, R. Artemisia Afra: A Potential Flagship for African Medicinal Plants? S. Afr. J. Bot. 2009, 75, 185–195. [Google Scholar] [CrossRef]
  76. Sagaya Jansi, R.; Khusro, A.; Agastian, P.; Alfarhan, A.; Al-Dhabi, N.A.; Arasu, M.V.; Rajagopal, R.; Barcelo, D.; Al-Tamimi, A. Emerging Paradigms of Viral Diseases and Paramount Role of Natural Resources as Antiviral Agents. Sci. Total Environ. 2021, 759, 143539. [Google Scholar] [CrossRef]
  77. Wang, Y.; Dong, T.; Qi, G.; Qu, L.; Liang, W.; Qi, B.; Zhang, Z.; Shang, L.; Gao, H.; Du, X.; et al. Prevalence of Common Respiratory Viral Infections and Identification of Adenovirus in Hospitalized Adults in Harbin, China 2014 to 2017. Front. Microbiol. 2018, 9, 2919. [Google Scholar] [CrossRef] [PubMed]
  78. Mera, I.F.G.; Falconí, D.E.G.; Córdova, V.M. Secondary Metabolites in Plants: Main Classes, Phytochemical Analysis and Pharmacological Activities. Bionatura 2019, 4, 1000–1009. [Google Scholar] [CrossRef]
  79. García, A.Á.; Carril, E.P.U. Metabolismo Secundario de Plantas. Reduca (Biol.) Ser. Fisiol. Veg. 2009, 2, 119–145. [Google Scholar]
  80. Ebenezer, K.S.; Manivannan, R.; Punniyamoorthy, A.; Tamilselvan, C. Plant Secondary Metabolites of Antiviral Properties a Rich Medicinal Source for Drug Discovery: A Mini Review. J. Drug Deliv. Ther. 2019, 9, 161–167. [Google Scholar] [CrossRef]
  81. Helfer, M.; Koppensteiner, H.; Schneider, M.; Rebensburg, S.; Forcisi, S.; Müller, C.; Schmitt-Kopplin, P.; Schindler, M.; Brack-Werner, R. The Root Extract of the Medicinal Plant Pelargonium Sidoides Is a Potent HIV-1 Attachment Inhibitor. PLoS ONE 2014, 9, e87487. [Google Scholar] [CrossRef] [PubMed]
  82. Derksen, A.; Kühn, J.; Hafezi, W.; Sendker, J.; Ehrhardt, C.; Ludwig, S.; Hensel, A. Antiviral Activity of Hydroalcoholic Extract from Eupatorium perfoliatum L. Against the Attachment of Influenza A Virus. J. Ethnopharmacol. 2016, 188, 144–152. [Google Scholar] [CrossRef]
  83. Mansouri, S.; Kutky, M.; Hudak, K.A. Pokeweed Antiviral Protein Increases HIV-1 Particle Infectivity by Activating the Cellular Mitogen Activated Protein Kinase Pathway. PLoS ONE 2012, 7, e36369. [Google Scholar] [CrossRef]
  84. Oloche, J.J.; Oluremi, B.B.; Aruwa, C.E.; Sabiu, S. Molecular Modeling Identification of Key Secondary Metabolites from Xylopia Aethiopica as Promising Therapeutics Targeting Essential Measles Viral Proteins. Evid. Based Complement. Altern. Med. 2023, 2023, 1575358. [Google Scholar] [CrossRef] [PubMed]
  85. Wink, M. Potential of DNA Intercalating Alkaloids and Other Plant Secondary Metabolites against SARS-CoV-2 Causing COVID-19. Diversity 2020, 12, 175. [Google Scholar] [CrossRef]
  86. Alhazmi, H.A.; Najmi, A.; Javed, S.A.; Sultana, S.; Al Bratty, M.; Makeen, H.A.; Meraya, A.M.; Ahsan, W.; Mohan, S.; Taha, M.M.E.; et al. Medicinal Plants and Isolated Molecules Demonstrating Immunomodulation Activity as Potential Alternative Therapies for Viral Diseases Including COVID-19. Front. Immunol. 2021, 12, 637553. [Google Scholar] [CrossRef]
  87. Bhuiyan, F.R.; Howlader, S.; Raihan, T.; Hasan, M. Plants Metabolites: Possibility of Natural Therapeutics against the COVID-19 Pandemic. Front. Med. 2020, 7, 444. [Google Scholar] [CrossRef]
  88. Perez, G.R.M. Antiviral Activity of Compounds Isolated from Plants. Pharm. Biol. 2003, 41, 107–157. [Google Scholar] [CrossRef]
  89. Lin, D.; Xiao, M.; Zhao, J.; Li, Z.; Xing, B.; Li, X.; Kong, M.; Li, L.; Zhang, Q.; Liu, Y.; et al. An Overview of Plant Phenolic Compounds and Their Importance in Human Nutrition and Management of Type 2 Diabetes. Molecules 2016, 21, 1374. [Google Scholar] [CrossRef] [PubMed]
  90. Kumar, K.; Debnath, P.; Singh, S.; Kumar, N. An Overview of Plant Phenolics and Their Involvement in Abiotic Stress Tolerance. Stresses 2023, 3, 570–585. [Google Scholar] [CrossRef]
  91. Dai, J.; Mumper, R.J. Plant Phenolics: Extraction, Analysis and Their Antioxidant and Anticancer Properties. Molecules 2010, 15, 7313–7352. [Google Scholar] [CrossRef] [PubMed]
  92. Badshah, S.L.; Faisal, S.; Muhammad, A.; Poulson, B.G.; Emwas, A.H.; Jaremko, M. Antiviral Activities of Flavonoids. Biomed. Pharmacother. 2021, 140, 111596. [Google Scholar] [CrossRef] [PubMed]
  93. Srinivasan, V.; Brognaro, H.; Prabhu, P.R.; de Souza, E.E.; Günther, S.; Reinke, P.Y.A.; Lane, T.J.; Ginn, H.; Han, H.; Ewert, W.; et al. Antiviral Activity of Natural Phenolic Compounds in Complex at an Allosteric Site of SARS-CoV-2 Papain-like Protease. Commun. Biol. 2022, 5, 805. [Google Scholar] [CrossRef]
  94. Vilhelmova-Ilieva, N.; Galabov, A.S.; Mileva, M. Tannins as Antiviral Agents. In Tannins—Structural Properties, Biological Properties and Current Knowledge; IntechOpen: London, UK, 2020. [Google Scholar]
  95. Chen, T.Y.; Chen, D.Y.; Wen, H.W.; Ou, J.L.; Chiou, S.S.; Chen, J.M.; Wong, M.L.; Hsu, W.L. Inhibition of Enveloped Viruses Infectivity by Curcumin. PLoS ONE 2013, 8, e62482. [Google Scholar] [CrossRef] [PubMed]
  96. Orfali, R.; Rateb, M.E.; Hassan, H.M.; Alonazi, M.; Gomaa, M.R.; Mahrous, N.; Gaballah, M.; Kandeil, A.; Perveen, S.; Abdelmohsen, U.R.; et al. Antibiotics Sinapic Acid Suppresses SARS-CoV-2 Replication by Targeting Its Envelope Protein. Antibiotics 2021, 10, 25. [Google Scholar] [CrossRef]
  97. Goc, A.; Sumera, W.; Rath, M.; Niedzwiecki, A. Phenolic Compounds Disrupt Spike-Mediated Receptor-Binding and Entry of SARS-CoV-2 Pseudo-Virions. PLoS ONE 2021, 16, e0253489. [Google Scholar] [CrossRef]
  98. Yamada, K.; Ogawa, H.; Hara, A.; Yoshida, Y.; Yonezawa, Y.; Karibe, K.; Nghia, V.B.; Yoshimura, H.; Yamamoto, Y.; Yamada, M.; et al. Mechanism of the Antiviral Effect of Hydroxytyrosol on Influenza Virus Appears to Involve Morphological Change of the Virus. Antivir. Res. 2009, 83, 35–44. [Google Scholar] [CrossRef]
  99. 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]
  100. Münch, J.; Sanchez-Garcia, E.; Shorter, J.; Schrader, T.; Weil, T.; Diger Groß, R.; Röcker, A.; Bravo-Rodriguez, K.; Heid, C.; Sowislok, A.; et al. Supramolecular Mechanism of Viral Envelope Disruption by Molecular Tweezers. J. Am. Chem. Soc. 2020, 142, 17024–17038. [Google Scholar] [CrossRef]
  101. Teissier, E.; Penin, F.; Pécheur, E.I. Targeting Cell Entry of Enveloped Viruses as an Antiviral Strategy. Molecules 2011, 16, 221–250. [Google Scholar] [CrossRef] [PubMed]
  102. Tirado-Kulieva, V.A.; Hernández-Martínez, E.; Choque-Rivera, T.J. Phenolic Compounds versus SARS-CoV-2: An Update on the Main Findings against COVID-19. Heliyon 2022, 8, e10702. [Google Scholar] [CrossRef] [PubMed]
  103. Montenegro-Landívar, M.F.; Tapia-Quirós, P.; Vecino, X.; Reig, M.; Valderrama, C.; Granados, M.; Cortina, J.L.; Saurina, J. Polyphenols and Their Potential Role to Fight Viral Diseases: An Overview. Sci. Total Environ. 2021, 801, 149719. [Google Scholar] [CrossRef]
  104. Steinmann, J.; Buer, J.; Pietschmann, T.; Steinmann, E. Anti-Infective Properties of Epigallocatechin-3-Gallate (EGCG), a Component of Green Tea. Br. J. Pharmacol. 2013, 168, 1059–1073. [Google Scholar] [CrossRef] [PubMed]
  105. Wu, Y.; Crich, D.; Pegan, S.D.; Lou, L.; Hansen, M.C.; Booth, C.; Desrochers, E.; Mullininx, L.N.; Starling, E.B.; Chang, K.Y.; et al. Polyphenols as Potential Inhibitors of SARS-CoV-2 Rna Dependent Rna Polymerase (Rdrp). Molecules 2021, 26, 7438. [Google Scholar] [CrossRef] [PubMed]
  106. Uchida, S.; Ozaki, M.; Akashi, T.; Yamashita, K.; Niwat, M.; Taniyamat, K. Effects of (-)-Epigallocatechin-3-O-Gallate (Green Tea Tannin) on The Life Span of Stroke-Prone Spontaneously Hypertensive Rats. Clin. Exp. Pharmacol. Physiol. Suppl. 1993, 22, S302–S303. [Google Scholar] [CrossRef]
  107. Zhu, J.; Ou, L.; Zhou, Y.; Yang, Z.; Bie, M. (-)-Epigallocatechin-3-Gallate Induces Interferon-Λ2 Expression to Anti-Influenza A Virus in Human Bronchial Epithelial Cells (BEAS-2B) through P38 MAPK Signaling Pathway. J. Thorac. Dis. 2020, 12, 989–997. [Google Scholar] [CrossRef]
  108. More, G.K. Metabolomics and Cell-Based Assay Analysis of Selected South African Plants to Determine Anti-Rift Valley Fever Virus Activity. Ph.D. Thesis, University of South Africa, Johannesburg, South Africa, 2021. [Google Scholar]
  109. Kawai, K.; Tsuno, N.H.; Kitayama, J.; Okaji, Y.; Yazawa, K.; Asakage, M.; Hori, N.; Watanabe, T.; Takahashi, K.; Nagawa, H. Basic and Clinical Immunology Basic and Clinical Immunology Epigallocatechin Gallate, the Main Component of Tea Polyphenol, Binds to CD4 and Interferes with Gp120 Binding. J. Allergy Clin. Immunol. 2003, 112, 951–957. [Google Scholar] [CrossRef]
  110. Ono, K.; Nakane, H.; Fukushima, M.; Chermann, J.-C.; Barré-Sinoussi, F. Differential Inhibitory Effects of Various Flavonoids on the Activities of Reverse Transcriptase and Cellular DNA and RNA Polymerases. Eur. J. Biochem. 1990, 190, 469–476. [Google Scholar] [CrossRef] [PubMed]
  111. Qiao, Y.; He, H.; Zhang, Z.; Liao, Z.; Yin, D.; Liu, D.; Yi, B.; He, M. Long-Term Sodium Ferulate Supplementation Scavenges Oxygen Radicals and Reverses Liver Damage Induced by Iron Overloading. Molecules 2016, 21, 1219. [Google Scholar] [CrossRef] [PubMed]
  112. Zhu, Y.; Shao, Y.; Qu, X.; Guo, J.; Yang, J.; Zhou, Z.; Wang, S. Sodium Ferulate Protects against Influenza Virus Infection by Activation of the TLR7/9-MyD88-IRF7 Signaling Pathway and Inhibition of the NF-ΚB Signaling Pathway. Biochem. Biophys. Res. Commun. 2019, 512, 793–798. [Google Scholar] [CrossRef] [PubMed]
  113. Robinson, W.E.; Reinecket, M.G.; Abdel-Malekt, S.; Jiat, Q.I.; Chow, S.A.; Dickerson, R.E. Inhibitors of HIV-1 Replication That Inhibit HIV Integrase (AIDS/Therapeutics/Natural Products/Chicoric Acid) Communicated By. Proc. Natl. Acad. Sci. USA 1996, 93, 6326–6331. [Google Scholar] [CrossRef]
  114. Kratz, J.M.; Andrighetti-Fröhner, C.R.; Leal, P.C.; Nunes, R.J.; Yunes, R.A.; Trybala, E.; Simões, C.M.O. Evaluation of Anti-HSV-2 Activity of Gallic Acid and Pentyl Gallate. Biol. Pharm. Bull. 2008, 31, 903–907. [Google Scholar] [CrossRef] [PubMed]
  115. Singh, D.P.; Moore, C.A.; Gilliland, A.; Carr, J.P. Activation of Multiple Antiviral Defence Mechanisms by Salicylic Acid. Mol. Plant Pathol. 2004, 5, 57–63. [Google Scholar] [CrossRef]
  116. Wang, K.; Ping, S.; Huang, S.; Hu, L.; Xuan, H.; Zhang, C.; Hu, F. Molecular Mechanisms Underlying the In Vitro Anti-Inflammatory Effects of a Flavonoid-Rich Ethanol Extract from Chinese Propolis (Poplar Type). Evid. Based Complement. Altern. Med. 2013, 2013, 127672. [Google Scholar] [CrossRef]
  117. Giovinazzo, G.; Gerardi, C.; Uberti-Foppa, C.; Lopalco, L. Can Natural Polyphenols Help in Reducing Cytokine Storm in COVID-19 Patients? Molecules 2020, 25, 5888. [Google Scholar] [CrossRef] [PubMed]
  118. Dejani, N.N.; Elshabrawy, H.A.; da Bezerra Filho, C.S.M.; de Sousa, D.P. Anticoronavirus and Immunomodulatory Phenolic Compounds: Opportunities and Pharmacotherapeutic Perspectives. Biomolecules 2021, 11, 1254. [Google Scholar] [CrossRef] [PubMed]
  119. Wang, B.; Ding, Y.; Zhao, P.; Li, W.; Li, M.; Zhu, J.; Ye, S. Systems Pharmacology-Based Drug Discovery and Active Mechanism of Natural Products for Coronavirus Pneumonia (COVID-19): An Example Using Flavonoids. Comput. Biol. Med. 2022, 143, 105241. [Google Scholar] [CrossRef] [PubMed]
  120. Upton, J.W.; Chan, F.K.M. Staying Alive: Cell Death in Antiviral Immunity. Mol. Cell 2014, 54, 273–280. [Google Scholar] [CrossRef] [PubMed]
  121. Moradi, M.T.; Karimi, A.; Lorigooini, Z. Alkaloids as the Natural Anti-Influenza Virus Agents: A Systematic Review. Toxin Rev. 2018, 37, 11–18. [Google Scholar] [CrossRef]
  122. Abookleesh, F.L.; Al-Anzi, B.S.; Ullah, A. Potential Antiviral Action of Alkaloids. Molecules 2022, 27, 903. [Google Scholar] [CrossRef] [PubMed]
  123. Gürü, M.; Gürü, S.; Yılmaz Aydın, D. Effect of Alkaloids on SARS-CoV-2. In Naturengs Mtu Journal of Engineering and Natural Sciences; Malatya Turgut Ozal University: Malatya, Turkey, 2020. [Google Scholar] [CrossRef]
  124. Peng, J.; Hu, J.F.; Kazi, A.B.; Li, Z.; Avery, M.; Peraud, O.; Hill, R.T.; Franzblau, S.G.; Zhang, F.; Schinazi, R.F.; et al. Manadomanzamines A and B: A Novel Alkaloid Ring System with Potent Activity against Mycobacteria and HIV-1. J. Am. Chem. Soc. 2003, 125, 13382–13386. [Google Scholar] [CrossRef] [PubMed]
  125. Amasheh, M.; Fromm, A.; Krug, S.M.; Amasheh, S.; Andres, S.; Zeitz, M.; Fromm, M.; Schulzke, J.D. TNFα-Induced and Berberine-Antagonized Tight Junction Barrier Impairment via Tyrosine Kinase, Akt and NFκB Signaling. J. Cell Sci. 2010, 123, 4145–4155. [Google Scholar] [CrossRef]
  126. Abdallah, I.I.; Quax, W.J. A Glimpse into the Biosynthesis of Terpenoids. KnE Life Sci. 2017, 3, 81. [Google Scholar] [CrossRef]
  127. Rodriguez-Concepcion, M.; Avalos, J.; Bonet, M.L.; Boronat, A.; Gomez-Gomez, L.; Hornero-Mendez, D.; Limon, M.C.; Meléndez-Martínez, A.J.; Olmedilla-Alonso, B.; Palou, A.; et al. A Global Perspective on Carotenoids: Metabolism, Biotechnology, and Benefits for Nutrition and Health. Prog. Lipid Res. 2018, 70, 62–93. [Google Scholar] [CrossRef]
  128. Yazaki, K.; Arimura, G.I.; Ohnishi, T. “Hidden” Terpenoids in Plants: Their Biosynthesis, Localization and Ecological Roles. Plant Cell Physiol. 2017, 58, 1615–1621. [Google Scholar] [CrossRef] [PubMed]
  129. Huang, Y.; Xie, F.J.; Cao, X.; Li, M.Y. Research Progress in Biosynthesis and Regulation of Plant Terpenoids. Biotechnol. Biotechnol. Equip. 2021, 35, 1800–1809. [Google Scholar] [CrossRef]
  130. Ashour, M.; Wink, M.; Gershenzon, J. Biochemistry of Terpenoids: Monoterpenes, Sesquiterpenes and Diterpenes. In Biochemistry of Plant Secondary Metabolism, 2nd ed.; Wiley Blackwell: Hoboken, NJ, USA, 2010; Volume 40, pp. 258–303. ISBN 9781444320503. [Google Scholar]
  131. Hassan, S.T.S.; Masarčíková, R.; Berchová, K. Bioactive Natural Products with Anti-Herpes Simplex Virus Properties. J. Pharm. Pharmacol. 2015, 67, 1325–1336. [Google Scholar] [CrossRef] [PubMed]
  132. Rizzato, G.; Scalabrin, E.; Radaelli, M.; Capodaglio, G.; Piccolo, O. A New Exploration of Licorice Metabolome. Food Chem. 2017, 221, 959–968. [Google Scholar] [CrossRef] [PubMed]
  133. Wang, L.; Yang, R.; Yuan, B.; Liu, Y.; Liu, C. The Antiviral and Antimicrobial Activities of Licorice, a Widely-Used Chinese Herb. Acta Pharm. Sin. B 2015, 5, 310–315. [Google Scholar] [CrossRef]
  134. Thyagarajan, S.P.; Jayaram, S.; Gopalakrishnan, V.; Hari, R.; And, J.; Sripathi, M.S. Conference Proceedings Herbal Medicines for Liver Diseases in India. J. Gastroenterol. Hepatol. 2002, 17, 370–376. [Google Scholar] [CrossRef]
  135. Marisa, R.; Assessor, D.; Calapai, G.; Delbò, M. Assessment Report on Glycyrrhiza glabra L. and/or Glycyrrhiza inflata Bat. and/or Glycyrrhiza uralensis Fisch, Radix; Committee on Herbal Medicinal Products (HMPC): Basel, Switzerland, 2013. [Google Scholar]
  136. Fiore, C.; Eisenhut, M.; Krausse, R.; Ragazzi, E.; Pellati, D.; Armanini, D.; Bielenberg, J. Antiviral Effects of Glycyrrhiza Species 141 Antiviral Effects of Glycyrrhiza Species. Phytother. Res. 2008, 22, 141–148. [Google Scholar] [CrossRef]
  137. Richard, S.A. Exploring the Pivotal Immunomodulatory and Anti-Inflammatory Potentials of Glycyrrhizic and Glycyrrhetinic Acids. Mediat. Inflamm. 2021, 2021, 6699560. [Google Scholar] [CrossRef]
  138. Schmidt, R.; Enzinger, C.; Ropele, S.; Schmidt, H.; Fazekas, F.; Cinatl, J.; Morgenstern, B.; Bauer, G.; Chandra, P.; Rabenau, H.; et al. Glycyrrhizin, an Active Component of Liquorice Roots, and Replication of SARS-Associated Coronavirus. Lancet 1997, 71, 2045–2046. [Google Scholar]
  139. Armaka, M.; Papanikolaou, E.; Sivropoulou, A.; Arsenakis, M. Antiviral Properties of Isoborneol, a Potent Inhibitor of Herpes Simplex Virus Type 1. Antivir. Res. 1999, 43, 79–92. [Google Scholar] [CrossRef]
  140. Astani, A.; Schnitzler, P. Antiviral Activity of Monoterpenes Beta-Pinene and Limonene against Herpes Simplex Virus In Vitro. Iran. J. Microbiol. 2014, 6, 149. [Google Scholar]
  141. Yu, J.S.; Tseng, C.K.; Lin, C.K.; Hsu, Y.C.; Wu, Y.H.; Hsieh, C.L.; Lee, J.C. Celastrol Inhibits Dengue Virus Replication via Up-Regulating Type I Interferon and Downstream Interferon-Stimulated Responses. Antivir. Res. 2017, 137, 49–57. [Google Scholar] [CrossRef]
  142. Narayan, V.; Ravindra, K.C.; Chiaro, C.; Cary, D.; Aggarwal, B.B.; Henderson, A.J.; Prabhu, K.S. Celastrol Inhibits Tat-Mediated Human Immunodeficiency Virus (HIV) Transcription and Replication. J. Mol. Biol. 2011, 410, 972–983. [Google Scholar] [CrossRef] [PubMed]
  143. Tseng, C.K.; Hsu, S.P.; Lin, C.K.; Wu, Y.H.; Lee, J.C.; Young, K.C. Celastrol Inhibits Hepatitis C Virus Replication by Upregulating Heme Oxygenase-1 via the JNK MAPK/Nrf2 Pathway in Human Hepatoma Cells. Antivir. Res. 2017, 146, 191–200. [Google Scholar] [CrossRef]
  144. Hegazy, G.E.; Abu-Serie, M.M.; Abo-Elela, G.M.; Ghozlan, H.; Sabry, S.A.; Soliman, N.A.; Abdel-Fattah, Y.R. In Vitro Dual (Anticancer and Antiviral) Activity of the Carotenoids Produced by Haloalkaliphilic Archaeon Natrialba Sp. M6. Sci. Rep. 2020, 10, 5986. [Google Scholar] [CrossRef]
  145. Yim, S.K.; Kim, I.; Warren, B.; Kim, J.; Jung, K.; Ku, B. Antiviral Activity of Two Marine Carotenoids against SARS-CoV-2 Virus Entry in Silico and In Vitro. Int. J. Mol. Sci. 2021, 22, 6481. [Google Scholar] [CrossRef]
  146. Krastanov, A. Metabolomics—The State of Art. Biotechnol. Biotechnol. Equip. 2010, 24, 1537–1543. [Google Scholar] [CrossRef]
  147. Roberts, L.D.; Souza, A.L.; Gerszten, R.E.; Clish, C.B. Targeted Metabolomics. Curr. Protoc. Mol. Biol. 2012, 1. [Google Scholar] [CrossRef] [PubMed]
  148. Steinfath, M.; Strehmel, N.; Peters, R.; Schauer, N.; Groth, D.; Hummel, J.; Steup, M.; Selbig, J.; Kopka, J.; Geigenberger, P.; et al. Discovering Plant Metabolic Biomarkers for Phenotype Prediction Using an Untargeted Approach. Plant Biotechnol. J. 2010, 8, 900–911. [Google Scholar] [CrossRef]
  149. Nanusha, M.Y.; Krauss, M.; Schönsee, C.D.; Günthardt, B.F.; Bucheli, T.D.; Brack, W. Target Screening of Plant Secondary Metabolites in River Waters by Liquid Chromatography Coupled to High-Resolution Mass Spectrometry (LC–HRMS). Environ. Sci. Eur. 2020, 32, 142. [Google Scholar] [CrossRef]
  150. Ehrlich, P.R.; Raven, P.H. Butterflies and Plants: A Study in Coevolution. Evolution 1964, 18, 586–608. [Google Scholar] [CrossRef]
  151. Jones, C.G.; Firn, R.D. On the Evolution of Plant Secondary Chemical Diversity. Philos. Trans. R. Soc. Lond. B 1991, 333, 273–280. [Google Scholar] [CrossRef]
  152. Lee, S.; Oh, D.G.; Singh, D.; Lee, J.S.; Lee, S.; Lee, C.H. Exploring the Metabolomic Diversity of Plant Species across Spatial (Leaf and Stem) Components and Phylogenic Groups. BMC Plant Biol. 2020, 20, 39. [Google Scholar] [CrossRef]
  153. Halouska, S.; Fenton, R.J.; Barletta, R.G.; Powers, R. Predicting the in Vivo Mechanism of Action for Drug Leads Using NMR Metabolomics. ACS Chem. Biol. 2012, 7, 166–171. [Google Scholar] [CrossRef] [PubMed]
  154. Shahid, M.; Singh, U.B.; Khan, M.S. Metabolomics-Based Mechanistic Insights into Revealing the Adverse Effects of Pesticides on Plants: An Interactive Review. Metabolites 2023, 13, 246. [Google Scholar] [CrossRef] [PubMed]
  155. Hussein, M.; Han, M.-L.; Zhu, Y.; Zhou, Q.; Lin, Y.-W.; Hancock, R.E.W.; Hoyer, D.; Creek, D.J.; Li, J.; Velkov, T. Metabolomics Study of the Synergistic Killing of Polymyxin B in Combination with Amikacin against Polymyxin-Susceptible and-Resistant Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 2019, 64, 10–1128. [Google Scholar] [CrossRef]
  156. Liu, L.W.; Shi, Y.Y.; Li, Z.L.; Zuo, L.H.; Tang, M.; Jing, Z.W.; Zhao, H.Y.; Xue, P.; Zhou, L.; Du, Q.Z.; et al. Metabolomic Insights into the Synergistic Effect of Biapenem in Combination with Xuebijing Injection Against Sepsis. Front. Pharmacol. 2020, 11, 502. [Google Scholar] [CrossRef] [PubMed]
  157. Vidar, W.S.; Baumeister, T.U.H.; Caesar, L.K.; Kellogg, J.J.; Todd, D.A.; Linington, R.G.; Kvalheim, O.M.; Cech, N.B. Interaction Metabolomics to Discover Synergists in Natural Product Mixtures. J. Nat. Prod. 2023, 86, 655–671. [Google Scholar] [CrossRef] [PubMed]
  158. Zhu, S.; Yue, J.; Wang, X.; Zhang, J.; Yu, M.; Zhan, Y.; Zhu, Y.; Sy, S.K.B.; Lv, Z. Metabolomics Revealed Mechanism for the Synergistic Effect of Sulbactam, Polymyxin-B and Amikacin Combination against Acinetobacter Baumannii. Front. Microbiol. 2023, 14, 1217270. [Google Scholar] [CrossRef] [PubMed]
  159. Gupta, S.; Schillaci, M.; Roessner, U. Metabolomics as an Emerging Tool to Study Plant-Microbe Interactions. Emerg. Top. Life Sci. 2022, 6, 175–183. [Google Scholar] [CrossRef] [PubMed]
  160. Serag, A.; Salem, M.A.; Gong, S.; Wu, J.-L.; Farag, M.A. Decoding Metabolic Reprogramming in Plants under Pathogen Attacks, a Comprehensive Review of Emerging Metabolomics Technologies to Maximize Their Applications. Metabolites 2023, 13, 424. [Google Scholar] [CrossRef] [PubMed]
  161. Sampaio, B.L.; Edrada-Ebel, R.; Da Costa, F.B. Effect of the Environment on the Secondary Metabolic Profile of Tithonia Diversifolia: A Model for Environmental Metabolomics of Plants. Sci. Rep. 2016, 6, 29265. [Google Scholar] [CrossRef] [PubMed]
  162. Prinsloo, G.; Nogemane, N. The Effects of Season and Water Availability on Chemical Composition, Secondary Metabolites and Biological Activity in Plants. Phytochem. Rev. 2018, 17, 889–902. [Google Scholar] [CrossRef]
  163. Adeosun, W.B.; More, G.K.; Steenkamp, P.; Prinsloo, G. Influence of Seasonal and Geographic Variation on the Anti-HSV-1 Properties and Chlorogenic Acids Content of Helichrysum aureonitens Sch. Bip. Front. Mol. Biosci. 2022, 9, 961859. [Google Scholar] [CrossRef] [PubMed]
  164. Manchester, M.; Anand, A. Metabolomics: Strategies to Define the Role of Metabolism in Virus Infection and Pathogenesis. In Advances in Virus Research; Academic Press Inc.: Cambridge, MA, USA, 2017; Volume 98, pp. 57–81. [Google Scholar]
  165. Ren, Z.; Fang, M.; Muhae-Ud-Din, G.; Gao, H.; Yang, Y.; Liu, T.; Chen, W.; Gao, L. Metabolomics Analysis of Grains of Wheat Infected and Noninfected with Tilletia Controversa Kühn. Sci. Rep. 2021, 11, 18876. [Google Scholar] [CrossRef] [PubMed]
  166. Nothias-Scaglia, L.F.; Retailleau, P.; Paolini, J.; Pannecouque, C.; Neyts, J.; Dumontet, V.; Roussi, F.; Leyssen, P.; Costa, J.; Litaudon, M. Jatrophane Diterpenes as Inhibitors of Chikungunya Virus Replication: Structure-Activity Relationship and Discovery of a Potent Lead. J. Nat. Prod. 2014, 77, 1505–1512. [Google Scholar] [CrossRef]
  167. Ferraz, A.C.; Gomes, P.W.P.; da Menegatto, M.B.S.; Lima, R.L.S.; Guimarães, P.H.; Reis, J.D.E.; Carvalho, A.R.V.; Pamplona, S. das G.S.R.; Muribeca, A. de J.B.; de Magalhães, J.C.; et al. Exploring the Antiviral Potential of Justicidin B and Four Glycosylated Lignans from Phyllanthus brasiliensis against Zika Virus: A Promising Pharmacological Approach. Phytomedicine 2024, 123, 155197. [Google Scholar] [CrossRef]
  168. Haggag, E.G.; Elshamy, A.M.; Rabeh, M.A.; Gabr, N.M.; Salem, M.; Youssif, K.A.; Samir, A.; Bin Muhsinah, A.; Alsayari, A.; Abdelmohsen, U.R. Antiviral Potential of Green Synthesized Silver Nanoparticles of Lampranthus coccineus and Malephora lutea. Int. J. Nanomed. 2019, 14, 6217–6229. [Google Scholar] [CrossRef]
  169. Takeda, Y.; Okuyama, Y.; Nakano, H.; Yaoita, Y.; Machida, K.; Ogawa, H.; Imai, K. Antiviral Activities of Hibiscus sabdariffa L. Tea Extract Against Human Influenza A Virus Rely Largely on Acidic PH but Partially on a Low-PH-Independent Mechanism. Food Environ. Virol. 2020, 12, 9–19. [Google Scholar] [CrossRef]
  170. Aati, H.Y.; Ismail, A.; Rateb, M.E.; AboulMagd, A.M.; Hassan, H.M.; Hetta, M.H. Garcinia Cambogia Phenolics as Potent Anti-COVID-19 Agents: Phytochemical Profiling, Biological Activities, and Molecular Docking. Plants 2022, 11, 2521. [Google Scholar] [CrossRef]
  171. Cock, I.E.; Matthews, B. Metabolomic Profiling of Antiviral Scaevola spinescens Extracts by High Resolution Tandem Mass Spectrometry. Acta Hortic. 2016, 1125, 1–18. [Google Scholar] [CrossRef]
  172. Nagai, T.; Shimizu, Y.; Shirahata, T.; Sunazuka, T.; Kiyohara, H.; Omura, S.; Yamada, H. Oral Adjuvant Activity for Nasal Influenza Vaccines Caused by Combination of Two Trihydroxy Fatty Acid Stereoisomers from the Tuber of Pinellia ternata. Int. Immunopharmacol. 2010, 10, 655–661. [Google Scholar] [CrossRef] [PubMed]
  173. Mei, J.; Zhou, Y.; Yang, X.; Zhang, F.; Liu, X.; Yu, B. Active Components in Ephedra Sinica Stapf Disrupt the Interaction between ACE2 and SARS-CoV-2 RBD: Potent COVID-19 Therapeutic Agents. J. Ethnopharmacol. 2021, 278, 114303. [Google Scholar] [CrossRef]
  174. Ngoc, T.M.; Phuong, N.T.T.; Khoi, N.M.; Park, S.J.; Kwak, H.J.; Nhiem, N.X.; Trang, B.T.T.; Tai, B.H.; Song, J.H.; Ko, H.J.; et al. A New Naphthoquinone Analogue and Antiviral Constituents from the Root of Rhinacanthus Nasutus. Nat. Prod. Res. 2019, 33, 360–366. [Google Scholar] [CrossRef] [PubMed]
  175. Chung, C.Y.; Liu, C.H.; Burnouf, T.; Wang, G.H.; Chang, S.P.; Jassey, A.; Tai, C.J.; Tai, C.J.; Huang, C.J.; Richardson, C.D.; et al. Activity-Based and Fraction-Guided Analysis of Phyllanthus urinaria Identifies Loliolide as a Potent Inhibitor of Hepatitis C Virus Entry. Antivir. Res. 2016, 130, 58–68. [Google Scholar] [CrossRef] [PubMed]
  176. Wu, S.F.; Lin, C.K.; Chuang, Y.S.; Chang, F.R.; Tseng, C.K.; Wu, Y.C.; Lee, J.C. Anti-Hepatitis C Virus Activity of 3-Hydroxy Caruilignan C from Swietenia Macrophylla Stems. J. Viral Hepat. 2012, 19, 364–370. [Google Scholar] [CrossRef]
  177. Gómez-Calderón, C.; Mesa-Castro, C.; Robledo, S.; Gómez, S.; Bolivar-Avila, S.; Diaz-Castillo, F.; Martínez-Gutierrez, M. Antiviral Effect of Compounds Derived from the Seeds of Mammea Americana and Tabernaemontana Cymosa on Dengue and Chikungunya Virus Infections. BMC Complement. Altern. Med. 2017, 17, 57. [Google Scholar] [CrossRef]
  178. Abubakar, A.R.; Haque, M. Preparation of Medicinal Plants: Basic Extraction and Fractionation Procedures for Experimental Purposes. J. Pharm. Bioallied Sci. 2020, 12, 1. [Google Scholar] [CrossRef]
  179. Savina, T.; Lisun, V.; Feduraev, P.; Skrypnik, L. Variation in Phenolic Compounds, Antioxidant and Antibacterial Activities of Extracts from Different Plant Organs of Meadowsweet (Filipendula ulmaria (L.) Maxim.). Molecules 2023, 28, 3512. [Google Scholar] [CrossRef] [PubMed]
  180. Wojdyło, A.; Nowicka, P.; Turkiewicz, I.P.; Tkacz, K.; Hernandez, F. Comparison of Bioactive Compounds and Health Promoting Properties of Fruits and Leaves of Apple, Pear and Quince. Sci. Rep. 2021, 11, 20253. [Google Scholar] [CrossRef] [PubMed]
  181. Herrera-Ruiz, M.; Román-Ramos, R.; Zamilpa, A.; Tortoriello, J.; Jiménez-Ferrer, J.E. Flavonoids from Tilia Americana with Anxiolytic Activity in Plus-Maze Test. J. Ethnopharmacol. 2008, 118, 312–317. [Google Scholar] [CrossRef] [PubMed]
  182. Goulas, V.; Manganaris, G.A. Towards an Efficient Protocol for the Determination of Triterpenic Acids in Olive Fruit: A Comparative Study of Drying and Extraction Methods. Phytochem. Anal. 2012, 23, 444–449. [Google Scholar] [CrossRef] [PubMed]
  183. Kim, K.H.; Moon, E.; Kim, S.Y.; Choi, S.U.; Lee, K.R. Lignan Constituents of Tilia Amurensis and Their Biological Evaluation on Antitumor and Anti-Inflammatory Activities. Food Chem. Toxicol. 2012, 50, 3680–3686. [Google Scholar] [CrossRef]
  184. Oniszczuk, A.; Podgórski, R.; Oniszczuk, T.; Zukiewicz-Sobczak, W.; Nowak, R.; Waksmundzka-Hajnos, M. Extraction Methods for the Determination of Phenolic Compounds from Equisetum arvense L. Herb. Ind. Crops Prod. 2014, 61, 377–381. [Google Scholar] [CrossRef]
  185. Noguerón-Merino, M.C.; Jiménez-Ferrer, E.; Román-Ramos, R.; Zamilpa, A.; Tortoriello, J.; Herrera-Ruiz, M. Interactions of a Standardized Flavonoid Fraction from Tilia Americana with Serotoninergic Drugs in Elevated plus Maze. J. Ethnopharmacol. 2015, 164, 319–327. [Google Scholar] [CrossRef]
  186. Zhang, A.; Sun, H.; Wang, X. Emerging Role and Recent Applications of Metabolomics Biomarkers in Obesity Disease Research. RSC Adv. 2017, 7, 14966–14973. [Google Scholar] [CrossRef]
  187. Halket, J.M.; Zaikin, V.G. Derivatization in Mass Spectrometry—7. On-Line Derivatization/Degradation. Eur. J. Mass. Spectrom. 2006, 12, 1–13. [Google Scholar] [CrossRef] [PubMed]
  188. More, G.K.; Vervoort, J.; Steenkamp, P.A.; Prinsloo, G. Metabolomic Profile of Medicinal Plants with Anti-RVFV Activity. Heliyon 2022, 8, e08936. [Google Scholar] [CrossRef]
  189. Tugizimana, F.; Piater, L.; Dubery, I. Plant Metabolomics: A New Frontier in Phytochemical Analysis. S. Afr. J. Sci. 2013, 109, 11. [Google Scholar] [CrossRef]
  190. Markley, J.L.; Dashti, H.; Wedell, J.R.; Westler, W.M.; Eghbalnia, H.R. Tools for Enhanced NMR-Based Metabolomics Analysis. In Methods in Molecular Biology; Humana Press Inc.: Totowa, NJ, USA, 2019; Volume 2037, pp. 413–427. [Google Scholar]
  191. Ludwig, C.; Günther, U.L. MetaboLab—Advanced NMR Data Processing and Analysis for Metabolomics. BMC Bioinform. 2011, 12, 366. [Google Scholar] [CrossRef]
  192. Tsugawa, H.; Tsujimoto, Y.; Arita, M.; Bamba, T.; Fukusaki, E. GC/MS Based Metabolomics: Development of a Data Mining System for Metabolite Identification by Using Soft Independent Modeling of Class Analogy (SIMCA). BMC Bioinform. 2011, 12, 131. [Google Scholar] [CrossRef]
  193. Xia, J.; Psychogios, N.; Young, N.; Wishart, D.S. MetaboAnalyst: A Web Server for Metabolomic Data Analysis and Interpretation. Nucleic Acids Res. 2009, 37, W652–W660. [Google Scholar] [CrossRef]
  194. Kılınc, H.; D’Urso, G.; Paolillo, A.; Alankus, O.; Piacente, S.; Masullo, M. LC-MS and NMR Based Plant Metabolomics: A Comprehensive Phytochemical Investigation of Symphytum Anatolicum. Metabolites 2023, 13, 1051. [Google Scholar] [CrossRef]
  195. Smith, C.A.; Want, E.J.; O’Maille, G.; Abagyan, R.; Siuzdak, G. XCMS: Processing Mass Spectrometry Data for Metabolite Profiling Using Nonlinear Peak Alignment, Matching, and Identification. Anal. Chem. 2006, 78, 779–787. [Google Scholar] [CrossRef]
  196. Du, X.; Smirnov, A.; Pluskal, T.; Jia, W.; Sumner, S. Metabolomics Data Preprocessing Using ADAP and MZmine 2. In Methods in Molecular Biology; Humana Press Inc.: Totowa, NJ, USA, 2020; Volume 2104, pp. 25–48. [Google Scholar]
  197. Tsugawa, H.; Cajka, T.; Kind, T.; Ma, Y.; Higgins, B.; Ikeda, K.; Kanazawa, M.; Vandergheynst, J.; Fiehn, O.; Arita, M. MS-DIAL: Data-Independent MS/MS Deconvolution for Comprehensive Metabolome Analysis. Nat. Methods 2015, 12, 523–526. [Google Scholar] [CrossRef] [PubMed]
  198. Guijas, C.; Montenegro-Burke, J.R.; Domingo-Almenara, X.; Palermo, A.; Warth, B.; Hermann, G.; Koellensperger, G.; Huan, T.; Uritboonthai, W.; Aisporna, A.E.; et al. METLIN: A Technology Platform for Identifying Knowns and Unknowns. Anal. Chem. 2018, 90, 3156–3164. [Google Scholar] [CrossRef] [PubMed]
  199. Mahrous, E.A.; Farag, M.A. Two Dimensional NMR Spectroscopic Approaches for Exploring Plant Metabolome: A Review. J. Adv. Res. 2015, 6, 3–15. [Google Scholar] [CrossRef] [PubMed]
  200. Udayakumar, M.; Chandar, D.P.; Arun, N.; Mathangi, J.; Hemavathi, K.; Seenivasagam, R. PMDB: Plant Metabolome Database-A Metabolomic Approach. Med. Chem. Res. 2012, 21, 47–52. [Google Scholar] [CrossRef]
  201. Pieters, L.; Vlietinck, A.J. Bioguided Isolation of Pharmacologically Active Plant Components, Still a Valuable Strategy for the Finding of New Lead Compounds? J. Ethnopharmacol. 2005, 100, 57–60. [Google Scholar] [CrossRef]
  202. Thissera, B.; Sayed, A.M.; Hassan, M.H.A.; Abdelwahab, S.F.; Amaeze, N.; Semler, V.T.; Alenezi, F.N.; Yaseen, M.; Alhadrami, H.A.; Belbahri, L.; et al. Bioguided Isolation of Cyclopenin Analogues as Potential SARS-CoV-2 Mpro Inhibitors from Penicillium citrinum Tdpef34. Biomolecules 2021, 11, 1366. [Google Scholar] [CrossRef]
  203. Altemimi, A.; Lakhssassi, N.; Baharlouei, A.; Watson, D.G.; Lightfoot, D.A. Phytochemicals: Extraction, Isolation, and Identification of Bioactive Compounds from Plant Extracts. Plants 2017, 6, 42. [Google Scholar] [CrossRef]
  204. Sasidharan, S.; Chen, Y.; Saravanan, D.; Sundram, K.M.; Latha, L.Y. Extraction, Isolation and Characterization of Bioactive Compounds from Plants’ Extracts. Afr. J. Tradit. Complement. Altern. Med. 2011, 8, 1–10. [Google Scholar] [CrossRef] [PubMed]
  205. Zhang, M.; Yu, Q.; Guo, J.; Wu, B.; Kong, X. Review of Thin-Layer Chromatography Tandem with Surface-Enhanced Raman Spectroscopy for Detection of Analytes in Mixture Samples. Biosensors 2022, 12, 937. [Google Scholar] [CrossRef] [PubMed]
  206. Czaplicki, S. Chromatography in Bioactivity Analysis of Compounds. In Column Chromatography; InTech: London, UK, 2013. [Google Scholar]
  207. Medini, F.; Megdiche, W.; Mshvildadze, V.; Pichette, A.; Legault, J.; St-Gelais, A.; Ksouri, R. Antiviral-Guided Fractionation and Isolation of Phenolic Compounds from Limonium Densiflorum Hydroalcoholic Extract. Comptes Rendus Chim. 2016, 19, 726–732. [Google Scholar] [CrossRef]
  208. Royani, A.; Hanafi, M.; Dewi, N.; Lotulung, P.; Eka Prastya, M.; Verma, C.; Manaf, A.; Alfantazi, A. Isolation and Identification of Bioactive Compounds from Tinospora Cordifolia Stem Extracts as Antibacterial Materials in Seawater Environments. Arab. J. Chem. 2023, 16, 105014. [Google Scholar] [CrossRef]
  209. Cecílio, A.B.; De Faria, D.B.; Oliveira, P.D.C.; Caldas, S.; De Oliveira, D.A.; Sobral, M.E.G.; Duarte, M.G.R.; Moreira, C.P.D.S.; Silva, C.G.; Almeida, V.L. De Screening of Brazilian Medicinal Plants for Antiviral Activity against Rotavirus. J. Ethnopharmacol. 2012, 141, 975–981. [Google Scholar] [CrossRef]
  210. Visintini Jaime, M.F.; Redko, F.; Muschietti, L.V.; Campos, R.H.; Martino, V.S.; Cavallaro, L.V. In Vitro Antiviral Activity of Plant Extracts from Asteraceae Medicinal Plants. Virol. J. 2013, 10, 245. [Google Scholar] [CrossRef]
  211. Beuscher, N.; Bodinet, C.; Neumann-Haefelin, D.; Marston, A.; Hostettmann, K. Antiviral Activity of African Medicinal Plants; Elsevier: Amsterdam, The Netherlands, 1994; Volume 42. [Google Scholar]
  212. Mohamed, A.-K.; Shier, W.T. A Simplified Plaque Reduction Assay for Antiviral Agents from Plants. Demonstration of Frequent Occurrence of Antiviral Activity in Higher Plants. J. Nat. Prod. 1990, 5, 340–344. [Google Scholar]
  213. Zhang, C.J.; Li, W.; Li, H.Y.; Wang, Y.L.; Yun, T.; Song, Z.P.; Song, Y.; Zhao, X.W. In Vivo and In Vitro Antiviral Activity of Five Tibetan Medicinal Plant Extracts against Herpes Simplex Virus Type 2 Infection. Pharm. Biol. 2009, 47, 598–607. [Google Scholar] [CrossRef]
  214. Huang, W.; Percie du Sert, N.; Vollert, J.; Rice, A.S.C. General Principles of Preclinical Study Design. In Handbook of Experimental Pharmacology; Springer: Berlin/Heidelberg, Germany, 2020; Volume 257, pp. 55–69. [Google Scholar]
  215. Shegokar, R. Preclinical Testing—Understanding the Basics First. In Drug Delivery Aspects: Volume 4: Expectations and Realities of Multifunctional Drug Delivery Systems; Elsevier: Amsterdam, The Netherlands, 2020; pp. 19–32. ISBN 9780128212226. [Google Scholar]
  216. England, C.; TrejoMartinez, J.; PerezSanchez, P.; Karki, U.; Xu, J. Plants as Biofactories for Therapeutic Proteins and Antiviral Compounds to Combat COVID-19. Life 2023, 13, 617. [Google Scholar] [CrossRef]
  217. Trivedi, P.; Abbas, A.; Lehmann, C.; Rupasinghe, H.P.V. Antiviral and Anti-Inflammatory Plant-Derived Bioactive Compounds and Their Potential Use in the Treatment of COVID-19-Related Pathologies. J. Xenobiotics 2022, 12, 289–306. [Google Scholar] [CrossRef]
  218. Venturella, G.; Ferraro, V.; Cirlincione, F.; Gargano, M.L. Medicinal Mushrooms: Bioactive Compounds, Use, and Clinical Trials. Int. J. Mol. Sci. 2021, 22, 634. [Google Scholar] [CrossRef]
  219. Ward, B.J.; Makarkov, A.; Séguin, A.; Pillet, S.; Trépanier, S.; Dhaliwall, J.; Libman, M.D.; Vesikari, T.; Landry, N. Efficacy, Immunogenicity, and Safety of a Plant-Derived, Quadrivalent, Virus-like Particle Influenza Vaccine in Adults (18–64 Years) and Older Adults (≥65 Years): Two Multicentre, Randomised Phase 3 Trials. Lancet 2020, 396, 1491–1503. [Google Scholar] [CrossRef]
  220. Pandey, A.; Khan, M.K.; Hamurcu, M.; Gezgin, S. Natural Plant Products: A Less Focused Aspect for the COVID-19 Viral Outbreak. Front. Plant Sci. 2020, 11, 568890. [Google Scholar] [CrossRef]
  221. Kell, D.B.; Oliver, S.G. The Metabolome 18 Years on: A Concept Comes of Age. Metabolomics 2016, 12, 148. [Google Scholar] [CrossRef]
  222. Thompson, K.D. Lead Molecules from Natural Products: Discovery and New Trends, 1st ed.; Khan, M.T.H., Ather, A., Eds.; Elsevier: Amsterdam, The Netherlands, 2006; Volume 2. [Google Scholar]
  223. Bang, S.; Quy Ha, T.K.; Lee, C.; Li, W.; Oh, W.K.; Shim, S.H. Antiviral Activities of Compounds from Aerial Parts of Salvia Plebeia R. Br. J. Ethnopharmacol. 2016, 192, 398–405. [Google Scholar] [CrossRef] [PubMed]
  224. Yadav, A.K.; Banerjee, S.K.; Das, B.; Chaudhary, K. Editorial: Systems Biology and Omics Approaches for Understanding Complex Disease Biology. Front. Genet. 2022, 13, 896818. [Google Scholar] [CrossRef] [PubMed]
  225. Melini, F.; Luziatelli, F.; Bonini, P.; Ficca, A.G.; Melini, V.; Ruzzi, M. Optimization of the Growth Conditions through Response Surface Methodology and Metabolomics for Maximizing the Auxin Production by Pantoea Agglomerans C1. Front. Microbiol. 2023, 14, 1022248. [Google Scholar] [CrossRef]
  226. Ferrell, J.E. Q&A: Systems Biology. J. Biol. 2009, 8, 2. [Google Scholar]
  227. Greene, C.S.; Troyanskaya, O.G. Integrative Systems Biology for Data-Driven Knowledge Discovery. Semin. Nephrol. 2010, 30, 443–454. [Google Scholar] [CrossRef] [PubMed]
  228. Debnath, M.; Prasad, G.B.K.S.; Bisen, P.S. Omics Technology. In Molecular Diagnostics: Promises and Possibilities; Springer: Berlin/Heidelberg, Germany, 2010; pp. 11–31. [Google Scholar]
Figure 1. A schematic representation showing the mechanisms of action of the antiviral properties of phenolic compounds.
Figure 1. A schematic representation showing the mechanisms of action of the antiviral properties of phenolic compounds.
Viruses 16 00218 g001
Figure 2. A schematic diagram showing the mechanism of the antiviral activities of alkaloids.
Figure 2. A schematic diagram showing the mechanism of the antiviral activities of alkaloids.
Viruses 16 00218 g002
Figure 3. A schematic diagram showing the mechanism of the antiviral activities of terpenoids.
Figure 3. A schematic diagram showing the mechanism of the antiviral activities of terpenoids.
Viruses 16 00218 g003
Table 1. Metabolomics Studies on Antiviral Compounds from Selected Plants.
Table 1. Metabolomics Studies on Antiviral Compounds from Selected Plants.
Plant SpeciesCompound/
Compound
Class
Metabolomics
Technique Used
Targeted
Virus (es)
Study OutcomesReference
Helichrysum aureonitensChlorogenic
acids
UPLC-qTOF-MSHerpes simplex virus-1 (HSV-1)Significant reduction of HSV-1 titre values[163]
Euphorbia amygdaloides ssp. semiperfoliataJatrophene esters
(terpenes)
UPLC-MS, 1H and 13C NMRChikungunya virus, HIV-1, and HIV-2Inhibition of viral replication via activation of protein kinase C[166]
Phyllanthus brasiliensisJusticidin B (polyphenols)HPLC and 13C NMRZika virusPost-infection intracellular reduction of viral load[167]
Lampranthus coccineus and Malephora luteaGreen synthesized silver nanoparticle
(AgNPs)
UPLC-MSHSV-1, HAV-10 virus, and Coxsackie B4Prevents viral entry into the host cell by binding to the viral envelope glycoproteins[168]
Hibiscus sabdariffaProtocatechuic acid (hydroxybenzoic acid derivatives)GC-MSHuman Influenza A VirusAcid-dependent virus inactivation[169]
Elaeodendron croceum, Artemisia afra, and Adansonia digitata13-Hydroxy-9Z,11E-octadecadienoic acid (fatty acid), 13S-Hydroxy-9Z,11E,15Z-octadecatrienoic acid (fatty acids)NMRRift valley fever virusInterferes with binding of the virion to the cellular receptors, therefore inhibiting viral entry and replication[108]
Garcinia cambogiaNaringin (flavonoid)LC-HRESIMSCOVID-19Inhibition of virus replication in the lung cells pre-infection[170]
Scaevola spinescensAmmarin (phenolic compounds), nodakenetin
(psoralens)
HPLC-MS/MS Q-TOFMS2 bacteriophageBacteriophage MS2 plaque reduction[171]
Pinellia ternataPinellic acid
(long-chain fatty acids)
HPLCNasal influenzaDemonstrated potency via activation of antiviral of IgA
antibody (Ab) and antiviral IgG1 Ab
[172]
Ephedra sinica4,6-dihydroxyquinoline-2-carboxylic acid, 4-hydroxyquinoline-2-carboxylic acid, and 4-hydroxy-6-methoxyquinoline-2-carboxylic acid
(quinoline carboxylic acids)
HPLC-Q-TOF-MS/MS, NMRCOVID-19Inhibited the infec
tivity rate of SARS-CoV-2 S protein-pseudoviruses
[173]
Rhinacanthus nasutusRhinacanthins C, D, N, Q, and E (naphthoquinones)NMR, mass spectrometryRhinovirus and coxsackievirusObserved cytopathic effect confirmed antiviral action[174]
Bombax ceibaBombasinol A
(polyphenols)
NMRHepatitis B VirusInhibits growth and replication of HepG2 2.2.15 cell lines[116]
Phyllanthus urinariaLoliolide
(benzofurans)
NMR,
ESI-LCMS
Hepatitis C Virus (HCV)Inhibits HCV host cell entry[175]
Swietenia macrophylla3-hydroxy caruilignan C (polyphenolic compound)NMR
LC-MS
Hepatitis C Virus (HCV)Inhibits
HCV via IFN-stimulated response and IFN-dependent antiviral gene expression
[176]
Tabernaemontana cymosaCoumarin A and B
(benzopyrone)
NMR, mass spectrometryChikungunya virus (CV)Inhibits CV host cell infection[177]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Adeosun, W.B.; Loots, D.T. Medicinal Plants against Viral Infections: A Review of Metabolomics Evidence for the Antiviral Properties and Potentials in Plant Sources. Viruses 2024, 16, 218. https://0-doi-org.brum.beds.ac.uk/10.3390/v16020218

AMA Style

Adeosun WB, Loots DT. Medicinal Plants against Viral Infections: A Review of Metabolomics Evidence for the Antiviral Properties and Potentials in Plant Sources. Viruses. 2024; 16(2):218. https://0-doi-org.brum.beds.ac.uk/10.3390/v16020218

Chicago/Turabian Style

Adeosun, Wilson Bamise, and Du Toit Loots. 2024. "Medicinal Plants against Viral Infections: A Review of Metabolomics Evidence for the Antiviral Properties and Potentials in Plant Sources" Viruses 16, no. 2: 218. https://0-doi-org.brum.beds.ac.uk/10.3390/v16020218

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop