Skip to main content

REVIEW article

Front. Microbiol., 02 October 2018
Sec. Virology

Antiviral Agents From Fungi: Diversity, Mechanisms and Potential Applications

  • 1Natural Resources Institute Finland (Luke), Helsinki, Finland
  • 2Division of Cell and Molecular Biology, Department of Biological and Environmental Science, Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
  • 3Natural Resources Institute Finland (Luke), Joensuu, Finland

Viral infections are amongst the most common diseases affecting people worldwide. New viruses emerge all the time and presently we have limited number of vaccines and only few antivirals to combat viral diseases. Fungi represent a vast source of bioactive molecules, which could potentially be used as antivirals in the future. Here, we have summarized the current knowledge of fungi as producers of antiviral compounds and discuss their potential applications. In particular, we have investigated how the antiviral action has been assessed and what is known about the molecular mechanisms and actual targets. Furthermore, we highlight the importance of accurate fungal species identification on antiviral and other natural products studies.

Introduction

Viruses cause serious outbreaks in all continents leading to difficult symptoms and mortality, and enormous economic burden for society. In addition, the constant emergence of new serotypes in virus groups that have a high mutation rate and low fidelity for viral replication adds challenges in combatting against these viruses.

Viruses can be divided into those containing a lipid envelope and those whose genome is only covered by a protein shell. Enveloped viruses are less stable and more prone to degradation when treated with lipid solvents. Their infection mechanisms are usually based on the presence of fusogenic peptides in the lipid envelope leading to a merge of viral and cellular membranes. The non-enveloped viruses are much more stable and may stay active in wastewaters and on surfaces from several weeks to months. The non-enveloped viruses such as Noro viruses and enteroviruses are therefore causing outbreaks that are difficult to handle. In addition, they show little sensitivity to chemical disinfectants (Wutzler and Sauerbrei, 2004; Chan and Abu Bakar, 2005). Thus, there is a need for both vaccines and antivirals to encounter viral infections. However, the development of vaccines against a wide range of newly emerging virus serotypes is challenging, and currently vaccines are available only against a handful of viruses. In addition, vaccination cannot help if the infection is already present in the system.

The antiviral drugs inhibit the virus infection either by specifically targeting the viral proteins or the host cellular factors that the viruses exploit for their reproduction (Clercq, 2002). However, the problem in using viral proteins as drug targets is the high rate at which viruses produce mutant resistant strains against them (De Palma et al., 2008). Cellular factors exploited by viruses also serve as potential drug targets. However, they cannot be considered automatically as reliable targets, since viruses may deviate from their original pathway and still cause an effective infection (Van der Linden et al., 2015). Also, targeting cellular factors might have an adverse effect on normal functioning of the host cells. Furthermore, the mechanisms of non-enveloped viruses to break the host cell membrane barrier is less well known, which forms an additional challenge in developing strategies against these viruses.

An antiviral drug has to fulfill a set of prerequisites when undergoing preclinical and clinical trials. A vital requirement is that the drug should be effective in inhibiting the virus infection without causing any cytotoxicity and with minimal side effects to the host cells. In addition, a drug should be able to completely inhibit the virus infection, partial inhibition leads to the generation of drug resistant mutant strains. Due to these prerequisites, only a handful of synthetic antiviral drugs have made it past the clinical phase. Until today, the successful ‘one bug–one drug’ approach has been used for antiviral drug development. However, today the focus has shifted toward designing broad-spectrum antivirals, which can act on multiple viruses by targeting a common but essential viral function (Vigant et al., 2015). Combinatorial chemistry is nowadays a preferred approach adapted by major drug companies for discovering pharmacologically significant compounds (Strobel and Daisy, 2003). Although combinatorial chemistry approach has proven successful in optimizing structures of drug compounds, only one de novo new chemical entity (NCE) has been approved as a drug [sorafenib (Nexavar) acting as anti-tumor] in these 25 plus years from this method (Cragg and Newman, 2007).

On the other hand, bioactive compounds isolated from natural biological sources offer a vast and unexplored diversity of chemical structures, unmatched by even the biggest combinatorial databases (Strobel and Daisy, 2003). Since thousands of years, natural products have served as traditional medicine and still provide the most affordable treatment for diseases in many developing countries (Amzat and Razum, 2018). Around 40% of modern drugs and 49% of new chemical products registered by the United States Food and Drug Administration (FDA) are based on natural products or their derivatives (Brewer, 2000). Bioactive compounds are naturally derived metabolites and/or by-products from microorganisms, plants, or animals (Baker et al., 2000). Since the past 25 years, bioactive compounds from many traditional medicinal plants have been screened for their antiviral activity by various research groups in Asia, Far East, Europe, and America (Jassim and Naji, 2003).

Particular importance for novel drug discoveries has been bioactive molecules of fungal origin. Especially fungi growing in unique environments such as endophytic and marine fungi are being constantly explored for their antibacterial and antifungal potential. During the past decade, many novel bioactive natural products possessing cytotoxic, anticancer, antibacterial or antifungal activities have been discovered from marine fungi (Mayer et al., 2013; Cheung et al., 2014; Singh et al., 2015). Fungi potentially contain and/or produce several effective molecules that could also be used as antivirals for other hosts. The discovery and characterization of fungal compounds having antiviral activities is an emerging field of research, and several compounds have already been identified as promising. In this review, we go through the present knowledge of fungi-derived extracts and other bioactive agents against viral infection. We especially focus on how the antiviral action has been assessed and how much is known about the mechanisms of action and actual targets.

Fungi as a Source of Antiviral Agents - an Overview

The kingdom Fungi represents a rich source of various biologically active compounds. During the past decades, thousands of compounds with diverse biological activities have been recognized and continue to be investigated. Fungal compounds with antiviral activities are less extensively studied, but also number of these investigations is on the increase. We have compiled a list of fungal orders with reported positive antiviral activities (Table 1) and also mapped this information on illustrative phylogenetic trees (Figures 13). Fungal species with reported antiviral activities are given in Supplementary Table S1. These demonstrate that the previous studies have focused on the late-diverging fungal phyla (Ascomycota and Basidiomycota) and on rather limited taxonomic groups, while several remaining completely uninvestigated.

TABLE 1
www.frontiersin.org

TABLE 1. Fungal orders with positive antiviral activities.

FIGURE 1
www.frontiersin.org

FIGURE 1. A tree illustrating the larger phylogeny of Fungi shows that the origin of presently known fungal-derived antiviral agents (highlighted) is restricted to the late-diverging fungal phyla (Ascomycota and Basidiomycota). The figure is constructed based on phylogenetic relationships of Fungi on Tree of Life Web Project (http://tolweb.org). This tree is illustrative and does not represent real phylogenetic data. Dashed lines: The group may not be monophyletic, or phylogenetic position of the group is uncertain.

FIGURE 2
www.frontiersin.org

FIGURE 2. In phylum Ascomycota, antiviral agents have been mainly identified from endophytes and other microfungi restricted to limited number of orders. Higher red color intensity indicates higher number of reports in literature. The figure is constructed based on phylogenetic relationships of Fungi on Tree of Life Web Project (http://tolweb.org). This tree is illustrative and does not represent real phylogenetic data. IA, indole alkaloids; NRPS, non-ribosomal peptides; PKS, polyketides; NRPS-PKS, hybrids; T, terpenoids; N/A, information not available. Dashed lines: The group may not be monophyletic, or phylogenetic position of the group is uncertain.

FIGURE 3
www.frontiersin.org

FIGURE 3. Antiviral agents reported from the phylum Basidiomycota. Higher red color intensity indicates higher number of reports in literature. The figure is constructed based on phylogenetic relationships of Fungi on Tree of Life Web Project (http://tolweb.org). This tree is illustrative and does not represent real phylogenetic data. L, lignin derivative; PS, polysaccharides; P, proteins; C, polysaccharide-protein/amino acid complex; NRPS, non-ribosomal peptides; PKS, polyketides; T, terpenoids; N/A, information not available. Dashed lines: The group may not be monophyletic, or phylogenetic position of the group is uncertain.

Particularly well-studied for their biologically active compounds, including antivirals, are edible and medicinal mushrooms. Another group of fungi that has been a focus of interest are endophytic fungi, particularly those that grow in marine habitats. The biologically active compounds can be roughly divided into two major groups of molecules; the high-molecular weight compounds present in the extracts and products derived from the fruiting bodies of edible and medicinal mushrooms, and the small organic molecules (secondary metabolites) excreted by the endophytic and other fungi in a liquid culturing (fermentation) setups.

Further rough division can be made when considering the repertoire of antiviral compounds found from different fungal taxonomic groups. Mapping the antiviral compounds on the larger phylogeny of Fungi (Figure 1) demonstrates that all the currently known secondary metabolites have been identified from Ascomycota and Basidiomycota. Ascomycota with antiviral activities includes endophytes and other microfungi restricted to limited number of orders (Figure 2), while the edible and medicinal mushrooms in the Agaricales and Polyporales (Basidiomycota) (Figure 3) are recognized as a source of high-molecular weight compounds. The increasing number of published fungal genome data combined with the traditional bioactivity screening methods has provided novel insights into the true capacity of fungi as producers of bioactive compounds (Bergmann et al., 2007; Khaldi et al., 2010; Brakhage, 2013; Clevanger et al., 2017). These studies indicate that differences exist between these two phyla in a number of secondary metabolites biosynthetic gene clusters and their dominance in their genomes; basidiomycetes typically having fewer compared to ascomycetes (Brakhage, 2013). However, the reported differences between Ascomycota and Basidiomycota reflect also to the bias from the different methods that have been commonly used in screening their biologically active compounds, not differences in their true arsenals of bioactive compounds.

The most recent estimates predicting fungal species diversity indicate that only 3–8% of existing fungal species are discovered and described (Hawksworth and Lücking, 2017). Therefore, the fungi investigated and found to have potential positive antiviral activities thus far represent only a minute fraction of these organisms and their potential.

Edible and Medicinal Mushrooms

Mushrooms have been an important part of our diet for centuries due to their nutritional properties. Their rich content in proteins, carbohydrates, minerals, vitamins, unsaturated fatty acids and low values of fat and energy content makes them a valuable food source (Barros et al., 2007, 2008; Çağlarırmak, 2007; Kalač, 2009; Ouzouni et al., 2009; Reis et al., 2012).

Some species producing conspicuous fruiting bodies have a long history of medicinal use. Bioactive compounds of the fungal genera which have had an important role in traditional medicine, such as Ganoderma, have been subject to extensive research. However, there is a broad number of other edible and medicinal species from different genera considered to be potential antiviral precursors (Supplementary Table S1 and Figure 3). The antiviral activity of these mushrooms is associated mainly to the presence of polysaccharides in mycelium and fruiting bodies, and synthesis of triterpenoid secondary metabolites (Chen et al., 2012; Rincão et al., 2012). However, large number of other potentially bioactive compounds and/or genes involved in their synthesis has been reported (Shiao, 2003; Chen et al., 2012), indicating that the full potential of mushroom and medicinal fungi as a source of bioactive compounds remains only partially understood. Previous study has reported considerable differences in the contents of bioactive compounds produced at different stages of fungal life cycle (Chen et al., 2012), implying that antiviral studies need to take into account the phenotypic variation and growth conditions of the fungal material.

Endophytes, Marine Fungi and Plant Pathogens

Endophytic fungi that inhabit above-ground tissues of healthy plant at least part of their life cycle are highly diverse in terms of species richness. These primarily ascomycetous (Ascomycota) fungi common in all terrestrial habitats are considered to have important ecological roles in the terrestrial plant communities. Their interactions with host plants and cross-talk with other endophytic microorganisms colonizing the same plant are complex and dynamic (Kusari et al., 2012). Endophytic fungi have been recognized as a rich source of secondary metabolites, which role in the natural habitat likely include chemical signaling, defense against other microorganism, and establishment of symbiosis with host plant (Schulz and Boyle, 2005; Yim et al., 2007; Khaldi et al., 2010). Some also mimic plant defense compounds, and can, therefore, protect host plants against herbivores and pathogens (Kusari et al., 2012). These secondary metabolites are known to have great chemical variety and numerous biological activities with pharmaceutical and biotechnological potential.

It has been hypothesized that extreme habitats harbor greater changes for novel drug discovery (Thatoi et al., 2013; Chávez et al., 2015). Interestingly, rich fungal species diversity inhabits extreme environments such as deep-sea sediments and mangrove ecosystems (Kumaresan and Suryanarayanan, 2001; Mahé et al., 2013). Many of ascomycetous species found in these habitats have been discovered having antiviral and other biological activities (Desmukh et al., 2018). The extreme conditions are thought to shape the secondary metabolite patterns of fungi, and these fungi are recognized as a particularly promising source of diverse and structurally unprecedented novel compounds, which some have already been structurally characterized and several been discovered to constitute of novel carbon skeletons (Saleem et al., 2007).

However, also already relatively well-known fungi should not be overlooked. Less intensively investigated fungi for their bioactivities include tree-pathogens that also seem promising source of antiviral agents. A previous study has detected a number of plant pathogenic fungi with various ecological roles (white-rot fungi, soft-rot fungi, blue-stain fungi and insect-symbionts) having antiviral activities (Mlinaric et al., 2005).

Antiviral Research and Fungal Taxonomy

Accurate organism identification is the basis for any biological research and its applications. This is particularly important for bioactive compounds aimed for pharmaceutical products. When the physical material used is reported with a misapplied name, the reproducibility of the study is very low. Unfortunately, in the literature on bioactivity and mechanisms of action of isolated compounds or crude extracts of fungal origin, reporting on the methods used to identify fungal materials reveals insensitivity to the relevant taxonomic discussion. Methodologically, only a minority of studies have included a combination of morphological and molecular methods for species identification (Raja et al., 2017). Given the factual diversity of kingdom Fungi, and the resulting difficulties in delimitating species and genera, as well as constant discoveries of species new to science (Hawksworth and Lücking, 2017), transparency in this matter is paramount. Long lasting debates among taxonomists, whether to accept new names, splitting of an old species into many new, or combinations of old names are an everyday affair in the field. This has in some cases resulted in considerable nomenclatural stratification, highlighting the need to engage taxonomists also in the study of applications.

To illustrate this problem, we evaluated literature on one of the most commonly reported name appearing in fungal antiviral research, ‘Ganoderma lucidum,’ as well as other species in the genus Ganoderma Karst. The poroid, saprotrophic fungal species G. lucidum (W. Curt. : Fr.) Karst is an concise example of the broader issue. The traditional medicinal use of Ganoderma spp. in East Asia, South-East Asia, and Africa has promoted interest in studying the bioactivity of these fungi, with ‘G. lucidum’ often cited as the species of the material. However, exact delimitation of the species concept for G. lucidum, with a European type locality, has been difficult due to lack of a holotype specimen (Steyaert, 1972; Moncalvo and Ryvarden, 1997). After morphological and molecular phylogenetic studies on the diversity of the genus in the past decades (Moncalvo et al., 1995; Cao et al., 2012; Zhou et al., 2015), the consensus in the taxonomic literature is that the industrially cultivated “Linghzi” and “Reishi” do not represent the G. lucidum s. str, but in fact other species (Wang et al., 2009; Cao et al., 2012). Therefore, careful consideration is required when identifying such samples under this name. Here, we listed the reported methods of acquisition and identification used in each antiviral study on Ganoderma (Supplementary Table S2). As a summary, out of the 13 studies, only four used material that we can safely assume to represent the species declared, as a fungal taxonomist was being consulted. In eight cases it seems unlikely given the sourcing of the materials, but could in principle be verified to the contrary, assuming access to the original material in herbaria. In one case, the experimental set-up is likely not reproducible due to vague description of the material used, and apparent lack of any preserved specimens. No studies reported sequence data accession numbers, nor morphological criteria used for species determination. Various forms of authorship, including outdated and erroneous, were present with the name G. lucidum.

Whether fungal material is in fact correctly identified, has consequences to the independent reproducibility of the study, and reflects also to understanding the species characteristics (i.e., requirements and phenotypic variation in artificial cultivation settings). There is yet a limited amount of comparative work on the differences between species and strains of the composition in the bioactive compounds within Ganoderma. The publications available at the moment indicate that differences may be considerable (Welti et al., 2015; Hennicke et al., 2016), though assessments into the extent of occurrence of compounds of interest within the genus is again convoluted by the non-transparent reporting of materials (Richter et al., 2015). In conclusion, given the likelihood of misapplied names in the literature, citing studies not reporting identification criteria as evidence on the antiviral potential of G. lucidum s. str. needs to take this ambiguity into account.

The misidentification of species and even genera is even more likely with microscopic fungi (such as endophytes) containing minute and overlapping morphological characteristics, and of which taxonomy and diversity remains widely uninvestigated. Therefore, we highlight the importance for transparency in reporting of used nomenclature, physical fungal material and method of identification, which is paramount to the advancement of research on antivirals from fungi. Furthermore, we encourage the natural product research community adopting the recently suggested set of standardized procedures for the identification of fungi (Raja et al., 2017).

Overview of Methods Assessing Antiviral Activity

The most widely used methods for the initial screening of fungal extracts to evaluate their antiviral activity are the plaque reduction assay (Zhu et al., 2004; Faccin et al., 2007; Rincão et al., 2012), cytopathic effect (CPE) assay (Liu et al., 2004; Zhang et al., 2011) and immunofluorescence assay (Faccin et al., 2007) (Table 2). In addition, various commercially available viability assays monitoring for, e.g., the cellular ATP levels have also been used. These assays are also used for performing the time of addition studies and investigating the direct virucidal activity of the fungal extracts (Liu et al., 2004; Faccin et al., 2007).

TABLE 2
www.frontiersin.org

TABLE 2. Methods used to evaluate antiviral effects.

All of these methods calculate in different ways the viability of the cells after virus action, and the antiviral activity is monitored as the rescue of the cells from the viral infection. The read out for the plaque reduction assay is the visual counting of the number of plaques formed [plaques forming unit (PFU)/ml] i.e., number of unstained “holes” in the culture plate after crystal violet staining of the cells that still adhere on the plate. This number is then used to calculate the percentage of viral inhibition (% V.I.) (Zhu et al., 2004). In immunofluorescence assay, the cells are observed under microscope and typically several hundreds of cells are scored. First, the number of infected cells is calculated from the number of cells showing high abundance of viral capsid proteins produced in the cell cytoplasm (Marjomäki et al., 2002). Then, from the obtained number, V.I. is calculated with respect to untreated infected cells (Faccin et al., 2007). In the case of CPE assay, the read out is based on the spectrophotometric absorbance reading of the stained viable cells, which is used to calculate the % V.I. (Liu et al., 2004). Typically, the viable cells left on the bottom of the culture plate and stained with crystal violet, are dissolved in the lysis buffer to provide a homogenous blue suspension that is easy to measure in the spectrophotometer (Schmidtke et al., 2001). The linear regression analysis of the plots of % V.I. is used to determine the 50% inhibitory concentration (IC50) which is used further to calculate the selectivity index (SI) (Rincão et al., 2012). The calculations are also given here as formulas:

% VI calculated from the plaque reduction assay read-out=[1(number of plaques in test/number of plaques in virus control)]×100(Rincão et al., 2012)% VI calculated from the CPE assay read-out= [(ODt)v - (ODc)v]/[(ODc)mock - (ODc)v] × 100(Liu et al., 2004)

where (ODt)v is the optical density (OD) of the cell, treated with virus and bioextract (test), (ODc)v is the OD of the cell, treated with virus (virus control) and (ODc)mock is the OD of the mock infected cell (cell control).

SI = CC50/IC50

Where CC50 is 50% cytotoxic concentration, i.e., the concentration which caused a 50% reduction in the number of viable cells or in the optical density and IC50 is 50% inhibitory concentration, i.e., the concentration capable of reducing 50% PFU in relation to the controls.

These above-mentioned methods only affirm the antiviral potential of bioactive compounds and do not reveal any information regarding their mechanism of action. Only few papers have progressed to evaluate the actual molecular targets. In order to study various viral or cellular targets of drug action, several approaches could be used. To study the direct effect on the virus, there are several methods that could be employed. First of all, perhaps the easiest way to see gross effect on the virus particle is to negatively stain the virus samples and observe them under transmission electron microscope (TEM) (Myllynen et al., 2016). There is a characteristic feature to distinguish between intact viruses from empty particles in TEM imaging. The staining dye, e.g., 2% Uranyl acetate or 1% phosphotungstic acid cannot enter the capsid of intact viruses because of which the intact particles appear bright, i.e., unstained, in TEM images (due to the contrast of the dye). However, in case of empty viruses, since the capsid is open, the dye enters the capsid and stains the insides of the virus thus giving a dark appearance for empty virus particles in TEM images. Density gradient centrifugation of either radioactively labeled or non-labeled virus is also insightful in revealing the direct effect of the extract on the virus (Marjomäki et al., 2002; Myllynen et al., 2016). The read out of radioactive gradient fractionation is the radioactivity [counts per minute (CPM)] of various fractions from different densities showing peaks of more dense intact virus and less dense empty viruses or even smaller products like pentamers. Direct effects of bioactive agents should show clear changes in the fraction of intact versus empty viruses.

The effect on the virus attachment on cellular receptors has been studied using binding assays. Binding is most sensitively studied using radioactively labeled virus and by performing binding assays in cold, hence eliminating the virus entry inside the cells by endocytosis (Marjomäki et al., 2002). Specific effects of molecules interfering with receptor binding have been also performed in silico by using molecular docking studies (Zhang et al., 2014). Whether the drug targets the virus uncoating in vitro or while the virus is inside cellular compartments, can be evaluated using real-time spectroscopy by using RNA/DNA binding fluorescent dyes (Myllynen et al., 2016) and using radioactive gradient fractionation studies, respectively. Radioactively labeled virus may be isolated from the cells for gradient fractionation which may reveal if there is a block in the viral genome release, thus leaving the virus as intact for longer periods. In order to assess the effect of bioextract on the efficiency of replication and viral translation, immunofluorescent labeling may be performed that reveals production of virus capsid proteins and specific replication intermediates, such as, e.g., dsRNA (Martikainen et al., 2015). Furthermore, qPCR to reveal new viral RNA production may be used.

There are also more specific assays that have been used to test the bioactive agents against virus-specific proteins such as proteases. Those assays have been developed directly against specific viruses such as HIV or human cytomegalovirus (hCMV) (Guo et al., 2000; Singh et al., 2004). Those assays are usually in vitro assays relying on purified proteases.

Bioactive Agents Acting as Antivirals

Small-molecule fungal secondary metabolites have been a source of various drugs, and the same classes of secondary metabolites seem promising also against viruses (Table 3). Other bioactive compounds with potential antiviral activities include high molecular weight compounds, such as polysaccharides, proteins and lignin-derivatives.

TABLE 3
www.frontiersin.org

TABLE 3. Fungal bioactive agents with reported antiviral activities.

Small Organic Molecules (Secondary Metabolites)

Fungal secondary metabolites are low-molecular-weight compounds, which in contrast to primary metabolites, are not directly required for the growth of the organism. Their ecological function in nature remains widely unknown. It has been hypothesized that secondary metabolites contribute to chemical communication with and competition against other organisms (Yim et al., 2007; Khaldi et al., 2010). Some also mimic plant defense compounds, and can, therefore, protect host plants against herbivores and pathogens (Kusari et al., 2012). A majority of known secondary metabolites have been identified from ascomycetes in traditional culture-based screening approaches, particular interest have been marine and plant endophytic fungi (Strobel and Daisy, 2003; Saleem et al., 2007; Kusari et al., 2012; Thatoi et al., 2013; Stierle and Stierle, 2015; Desmukh et al., 2018).

The production of secondary metabolites has been most commonly studied in in vitro setups, where the compounds secreted by hyphal cells to a culture medium are studied. Although these studies have formed an important basis for the discovery of fungal bioactive metabolites, it is likely that the true potential of fungi as producers of secondary metabolites has been underestimated. Genome-based projects have provided novel insights and demonstrated that many cryptic gene clusters involved in secondary metabolite biosynthesis are silent or not well expressed in standard cultivation/fermentation conditions traditionally used for screening for the secondary metabolites (Bergmann et al., 2007; Khaldi et al., 2010; Brakhage, 2013; Clevanger et al., 2017). To activate silent biosynthetic gene clusters, altering the growth conditions (such as carbon and nitrogen sources, temperature, light, pH and aeration) have been used as stimuli. However, regulation of secondary metabolism biosynthesis pathway is complex, and these are not universally relevant stimuli for most gene clusters and fungal species (Brakhage, 2013). Genome mining provides novel possibilities for understanding the genetic basis of secondary metabolite production and developing strategies for activation of the silent metabolic pathways (Bergmann et al., 2007; Andersen et al., 2012; Ochi and Hosaka, 2013).

The genome of each fungus contains a remarkable capacity of biosynthetic gene clusters encoding the production of diverse secondary metabolites (Khaldi et al., 2010; Chen et al., 2012; Inglis et al., 2013; Han et al., 2016). Although the secondary metabolites are structurally highly diverse, they are produced by a few common biosynthetic pathways (Keller et al., 2005). Previous studies which have applied genome predictions have identified non-ribosomal peptide synthase (NRPSs) and polyketide synthase (PKSs) gene clusters being the most abundant, while also hybrid NRPS-PKS enzymes, prenyltransferases (DMATSs), terpene cyclases (TCs) are commonly present in fungi (Bergmann et al., 2007; Khaldi et al., 2010; Andersen et al., 2012; Han et al., 2016). These “backbone” enzymes are responsible for the synthesis of the secondary metabolite core structures which include non-ribosomal peptides, polyketides, NRPS-PKS hybrids, indole alkaloids and terpenoids, respectively (Hoffmeister and Keller, 2007). The synthesized core structures and product intermediates are typically further modified by tailoring enzymes before the final product is transported outside the fungal cell (Andersen et al., 2012; Brakhage, 2013).

High Molecular Weight Compounds

The fungal cell wall is an essential structure component that protects the cells against the environment and other organisms. The fungal cell wall allows the selective exchange of compounds with other cells and with their surroundings. Apart from that, it also provides of shape and strength to the fungal cell. The composition of the cell wall varies between fungal species and within the same species or strains (e.g., growth stage, growth conditions, environmental factors). Despite the variability of the composition, the main components that can be commonly found in mushrooms are proteins and polysaccharides (Bowman and Free, 2006). High molecular weight polysaccharides (such as glucan, chitin, mannan, PSK or lentinan) extracted from fruiting bodies and fungal mycelia have been reported to present antiviral activities (Tochikura et al., 1987, 1988; Cardozo et al., 2011; Rincão et al., 2012).

To study the mechanism of action of polysaccharides against a determinate virus, its chemical composition must be understood. The analysis of the structure of polysaccharides is a complex task that requires several isolation steps. When polysaccharides are extracted from a fungal sample, the determination of the purity becomes a priority in order to understand the chemical structure. Knowledge on the monosaccharide composition, the linkage positions between glycosidic linkages, the distinction of furanosidic and pyranosidic rings, the anomeric configuration, the sequences of monosaccharide residues and repeating units, the substitutions and the molecular weight including its distribution are essential to define the structure of a certain polysaccharide (Cui, 2005).

The presence of proteins in the cell wall has also a protective function since they are the responsible of stimuli perception (Geoghegan et al., 2017). Hence, proteins are involved in the production and regulation of secondary metabolites (Bok and Keller, 2016). Moreover, their presence in the cell wall in combination with polysaccharide complexes allows the interaction with the environment, helping to the fungal cell to the transport of substances in and out of the fungal cell.

Several reports refer to replication inhibition for several virus types, suggesting that both polysaccharides and proteins act at the first phases of viral replication system (Tochikura et al., 1988; Collins and Ng, 1997; Eo et al., 1999a,b, 2000; Piraino and Brandt, 1999; Kim et al., 2000; Wang and Ng, 2000; Ngai and Ng, 2003; Gu et al., 2007; Cardozo et al., 2011; Yamamoto et al., 2013). However, the interaction of proteins and polysaccharides with the viral replication system is not completely understood.

Antiviral Mechanisms

Possibility to Act at Different Stages of Virus Life Cycle

There are several possibilities to interfere viral infection (Figure 4). Viruses can be directly attacked outside cells in order to destroy the viral particles before their attachment on cellular receptors. Such agents could irreversibly modify viral particles on different surfaces, or, if being non-toxic, also in human body. For human enteroviruses, several molecules have been designed to fit in to the special hydrophobic pocket, thus replacing the aliphatic fatty acid normally housed in the virus particle (De Colibus et al., 2014). The hope in this strategy is to stabilize the virus particle and prevent virus uncoating. This pocket is also close to the receptor binding area and thus molecules targeted to the pocket could potentially inhibit receptor binding. These molecules have shown some potency in their antiviral effect. However, binding to these pockets is usually dynamic, and the effects in long-term studies have not been successful. However, during short time periods, these molecules have shown efficacy.

FIGURE 4
www.frontiersin.org

FIGURE 4. A schematic representation of the life-cycle of a non-enveloped, positive sense single-stranded RNA. The infection stages of the virus that include attachment, entry, uncoating, genome release, genomic replication, translation, assembly and exit serve as potential targets for antivirals.

Inhibiting the receptor binding is another possibility to prevent virus infection. When several virus groups use similar receptors, this strategy offers a nice possibility to prevent viral infection, e.g., in the case of heparan sulfate binding (Cagno et al., 2018). Most viruses use cytoplasmic endosomes as their portal for cellular entry. This may be considered as a true rate limiting step for many viruses, especially those of non-enveloped viruses. By using fusogenic peptides that normally act in low pH, enveloped viruses have developed means to fuse with the limiting membrane of the endosomes, thus releasing their contents to the cytoplasm. Thus, several molecules that prevent the lowering of the endosomal pH quite effectively inhibit viral infection. However, as the low pH is such a crucial event for normal nutrient uptake and signaling through endosomes, such strategies are not really feasible. Non-enveloped viruses do not usually rely on low acidity. We have shown recently that enteroviruses in general do not use low pH in their strategy to infect cells, but rather accumulate in endosomes with higher acidity (Marjomäki et al., 2015). There is very little information yet available on the strategies of non-enveloped viruses to break the endosomal barrier.

RNA-based viruses start their translation and transcription usually in the cytoplasm. Those events are a good target for several antivirals as many of the mechanisms could target a larger amount of virus groups. DNA-viruses travel to the nucleus and start their replication there. The replication for several viruses take advantage of various membranous organelles as usually the replication occurs on the surface of membranous structures. Indeed, virus infection usually strongly perturbs the functioning of various endosomes, ER and Golgi for the profit of viruses. In addition, often cholesterol and some more rare forms of lipids are being utilized for viral replication, and may serve as targets for combatting viral infection.

During translation and replication, in addition to the viral structural proteins, several non-structural viral proteins are being synthetized in the cytoplasm. Several present strategies against viral infection target the viral proteases or viral polymerases and may prove successful in preventing viral infection.

Virus assembly occurs in the cytoplasm for several non-enveloped viruses that, in the end, causes lytic disruption of the cell and spread of the progeny viruses to neighboring cells. The lytic events are often preceded by activation of caspases to promote apoptosis. Viruses are master manipulators of anti-apoptotic growth factor signaling and proapoptotic caspase pathways. Usually viruses try to prevent apoptosis during the early infection but may boost apoptotic processes later to facilitate an efficient spread to the cell surroundings. Therefore, the strategies to manipulate apoptosis may be complicated. However, we showed previously that targeting BCl-molecules, thus boosting apoptosis, facilitated killing of virus infection early, and prevented possibilities for influenza and HSV to develop difficult symptoms usually encountered with virus infection (Bulanova et al., 2017). Thus, maybe in combination with other antivirals, this strategy could perhaps be used for antiviral action.

Enveloped viruses take their envelope usually from the plasma membrane and use some components of the cellular machinery, ESCRTS, to facilitate the topologically outbound formation of viral particles, as in the case of HIV viruses budding from cells. Attacking those ESCRT components could potentially prevent virus spread but also would target the elementary aspects of multivesicular structure formation of endosomes.

Mechanisms Found So Far

For mechanistic studies, rather limited numbers of viruses have been studied so far. Herpes simplex virus (HSV) has been most thoroughly tested against some purified and unpurified fungal products (Table 4). In addition to HSV, also influenza viruses (IF) have been tested against some purified fungal products. Both HSV and IF are enveloped viruses that are in general suspected to be more prone to degradation and destabilization. In contrast, non-enveloped viruses are considered more robust and may keep stabile even in the harsh conditions for long time periods. Maybe therefore, less hits have been discovered from fungal products. However, poliovirus, a member of non-enveloped enteroviruses, has been shown to be affected by Lentinula edodes and Agaricus subrufescens -derived products (Table 4). In addition, triterpenoids from G. lucidum have been shown to effectively reduce the infectivity of enterovirus 71 (Table 4).

TABLE 4
www.frontiersin.org

TABLE 4. Antiviral mechanisms.

There are several published antiviral studies especially with edible mushrooms and with their aqueous and ethanol/methanol extracts. Most studies on antiviral action have been performed using standard plaque assay or CPE assay, measuring the amount of infective particles after the treatment. With such assays, the inhibitory action may have occurred during any step of the viral infection, starting from direct action on the virus particle itself. More information on the inhibitory effect has been acquired from time of addition studies, where inhibitory molecules were added at different stages along viral infection (Faccin et al., 2007; Yamamoto et al., 2013). These studies have pinpointed several extracts and isolated molecules that showed inhibitory action directly on the virus particles or on the adsorption of the virus on cells (see Table 4). More detailed studies with Ganoderma triterpenoids using molecular docking tools showed affinity to the hydrophobic pocket of enterovirus 71 suggesting that either uncoating or binding to the cellular receptor could be affected (Zhang et al., 2014). These triterpenoids showed best efficacy when they were first mixed with the virus before adding on cells, confirming that either uncoating or binding on cells indeed were targeted. However, without further analysis addressing those steps with specific binding assays or uncoating assays the actual mechanism remains unknown.

Some studies showed preferential inhibition still some hours p.i. suggesting that the inhibitory action was probably in the viral protein translation or replication. More direct studies have been done with assays that specifically target viral proteins in vitro. These studies have been performed most heavily with HIV proteases and reverse transcriptases. Such studies have pinpointed ganoderic acid and triterpenoids, as well as adenosine, velutin and a novel 4.5 kDa agent to directly act on HIV proteins (Sato et al., 2009). In many cases the bioactive compound is chemically modified to increase its antiviral potency. Cardozo et al. (2011), produced a sulfated derivative of a polysaccharide, isolated from Agaricus brasiliensis and found that the sulfated polysaccharide showed increased antiviral activity against HSV I.

Future Perspectives

Currently, numerous fungal-derived metabolites such as lovastatins, antibiotics and antifungal agent griseofulvin are present on the drug markets. Fungal-derived compounds have not been approved for antiviral treatment. However, as numerous previous studies have found many of them exhibiting potential antiviral efficacy agents (Tables 1, 4), it is probably only a matter of time before some molecules will be taken for clinical testing. The effective antiviral fungal compounds showing the best ADME (pharmacokinetic characteristics adsorption, distribution, metabolism and excretion) in vitro will be taken for animal testing in vivo. However, thus far there are very few well-designed, high-quality clinical trials on treatments with fungal-derived standardized pharmaceuticals (Zhou et al., 2005; Gargano et al., 2017).

Standardizing the biosynthesis of biologically active compounds for trials, as well as up-scaling to industrial scale has to deal with complexity of fungal biology and ecology. The interspecies interactions are known to influence the fungal metabolism in the organism’s native environment, but their importance in biotechnological applications remains an underexplored issue (Kusari et al., 2014a,b). Intraspecies genetic and morphological variation complicates the optimization of cultivation conditions (Posch et al., 2013). Still, increasing number of fungal genome sequences in combination with metabolomics provide novel possibilities for understanding the regulation of secondary metabolism, enhancing the yields of target compounds, as well as providing a platform for novel drug discoveries (Harvey et al., 2015).

Alongside liquid cultivation, potential fungal antivirals have been extracted from harvested sporocarps, especially in studies on basidiomycetes. Domestication of various such sporocarp-producing species has been successful within industrial symbiosis built on easily obtainable lignocellulosic waste from agriculture and forestry. This approach has been supported in some cases by the observed difficulties in obtaining particular metabolites otherwise (Chen et al., 2012), though there are known issues of economic costs and quality control (Hu et al., 2017; Wu et al., 2017).

Despite these numerous challenges with species that have been studied in some cases for decades, it is also important to continue investigating fungal species diversity, as only a small number of the known fungi have been investigated for antiviral activity. Whereas biodiversity hot-spots and little-explored habitats are particularly important for finding unrecognized fungal species, cryptic species represent considerable genetic reserve also in long studied ecosystems (Hawksworth and Lücking, 2017). Multidisciplinary engagement between virologists and fungal taxonomists is particularly pressing in this case.

Conclusion

As fungi are a rich source of bioactive agents, the accumulation of know how on the actual bioactive molecules enriched, and their detailed targets in virus families will probably increase in the near future. Presently, there is a rather limited understanding of the antiviral mechanisms of fungal products on virus infection. Thus, more detailed knowledge on the actual molecular targets is crucial in order to develop these molecules further to efficiently combat virus infections in the future. Laboratory assays targeting directly various steps along virus infection are needed to understand in detail the mechanisms of action.

Author Contributions

All authors actively taken part in developing the idea of this review article. The literature review and manuscript writing were performed by RL, DR, PV, MC-E, and VM. HV arranged the required resources and commented on the manuscript. All authors contributed to the finalizing of the manuscript and approved it for publication.

Funding

This study was financially supported by the LUKE Leads MushValue-project, Natural Resources Institute Finland (LUKE). DR was financially supported by the Jane and Aatos Erkko Foundation and MC-E by the Niemi Foundation.

Conflict of Interest Statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Supplementary Material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fmicb.2018.02325/full#supplementary-material

References

Amoros, M., Boustie, J., Py, M.-L., Hervé, V., and Robin, V. (1997). Antiviral activity of homobasidiomycetes: evaluation of 121 basidiomycetes extracts on four viruses. Int. J. Pharmacogn. 35, 255–260. doi: 10.1076/phbi.35.4.255.13308

CrossRef Full Text | Google Scholar

Amzat, J., and Razum, O. (eds). (2018). “Traditional medicine in Africa,” in Towards a Sociology of Health Discourse in Africa, (Cham: Springer). doi: 10.1007/978-3-319-61672-8

CrossRef Full Text | Google Scholar

Andersen, M. R., Nielsen, J. B., Klitgaard, A., Petersen, L. M., Zachariasen, M., Hansen, T. J., et al. (2012). Accurate prediction of secondary metabolite gene clusters in filamentous fungi. Proc. Natl. Acad. Sci. U.S.A. 110, E99–E107. doi: 10.1073/pnas.1205532110

PubMed Abstract | CrossRef Full Text | Google Scholar

Awadh Ali, N. A., Mothana, R. A., Lesnau, A., Pilgrim, H., and Lindequist, U. (2003). Antiviral activity of Inonotus hispidus. Fitoterapia 74, 483–485. doi: 10.1016/S0367-326X(03)00119-9

CrossRef Full Text | Google Scholar

Baker, D., Mocek, U., and Garr, C. (2000). “Natural products vs. combinatorials: a case study,” in Biodiversity: New Leads for Pharmaceutical and Agrochemical Industries, eds S. K. Wrigley, M. A. Hayes, R. Thomas, E. J. T. Chrystal, and N. Nicholson (Cambridge: The Royal Society of Chemistry), 66–72.

Google Scholar

Barros, L., Baptista, P., Correia, D. M., Casal, S., Oliveira, B., and Ferreira, I. C. F. R. (2007). Fatty acid and sugar composition, and nutritional value of five wild edible mushrooms from Northeast Portugal. Food Chem. 105, 140–145. doi: 10.1016/j.foodchem.2007.03.052

CrossRef Full Text | Google Scholar

Barros, L., Cruz, T., Baptista, P., Estevinho, L. M., and Ferreira, I. C. F. R. (2008). Wild and edible mushroom as source of nutrients and nutraceuticals. Food Chem. Toxicol. 46, 2742–2747. doi: 10.1016/j.fct.2008.04.030

PubMed Abstract | CrossRef Full Text | Google Scholar

Bashyal, B. P., Wellensiek, B. P., Ramakrishan, R., Faeth, S. H., Ahmad, N., and Gunatilaka, A. A. (2014). Altertoxins with potent anti-HIV activity from Alternaria tenuissima QUE1Se, a fungal endophyte of Quercus emoryi. Bioorgan. Med. Chem. 2, 6112–6116. doi: 10.1016/j.bmc.2014.08.039

PubMed Abstract | CrossRef Full Text | Google Scholar

Bergmann, S., Schümann, J., Sherlach, K., Lange, C., Brakhage, A. A., and Hertweck, C. (2007). Genomics-driven discovery of PKS-NRPS hybrid metabolites from Aspergillus nidulans. Nat. Chem. Biol. 3, 213–217. doi: 10.1038/nchembio869

PubMed Abstract | CrossRef Full Text | Google Scholar

Bok, J. W., and Keller, N. P. (2016). “Insight into fungal secondary metabolism from ten years of LaeA research,” in Biochemistry and Molecular Biology, ed. D. Hoffmeister (Cham: Springer International Publishing Switzerland), 21–30.

Google Scholar

Bowman, S. M., and Free, S. J. (2006). The structure and synthesis of the fungal cell wall. BioEssays 28, 799–808. doi: 10.1002/bies.20441

PubMed Abstract | CrossRef Full Text | Google Scholar

Brakhage, A. A. (2013). Regulation of fungal secondary metabolism. Nat. Rev. Microbiol. 11, 21–32. doi: 10.1038/nrmicro2916

PubMed Abstract | CrossRef Full Text | Google Scholar

Brewer, S. (2000). “The relationship between natural products and synthetic chemistry in the discovery process,” in Biodiversity: New Leads for Pharmaceutical and Agrochemical Industries, eds S. K. Wrigley, M. A. Hayes, R. Thomas, E. J. T. Chrystal, and N. Nicholson (Cambridge: The Royal Society of Chemistry), 59–65.

Google Scholar

Bruggemann, R., Orlandi, J. M., Benati, F. J., Faccin, L. C., Mantovani, M. S., Nozawa, C., et al. (2006). Antiviral activity of Agaricus blazei Murrill ss. Heinem extract against human and bovine herpesviruses in cell culture. Braz. J. Biol. 37, 561–565.

Google Scholar

Bulanova, D., Ianevski, A., Bugai, A., Akimov, Y., Kuivanen, S., Paavilainen, H., et al. (2017). Antiviral properties of chemical inhibitors of cellular anti-apoptotic Bcl-2 proteins. Viruses 9:E271. doi: 10.3390/v9100271

PubMed Abstract | CrossRef Full Text | Google Scholar

Bunyapaiboonsri, T., Yoiprommarat, S., Srikitikuchai, P., Srichomthong, K., and Lumyong, S. (2010). Oblongolides from the endophytic fungus Phomopsis sp. BCC 9789. J. Nat. Prod. 73, 55–59. doi: 10.1021/np900650c

PubMed Abstract | CrossRef Full Text | Google Scholar

Çağlarırmak, N. (2007). The nutrients of exotic mushrooms (Lentinula edodes and Pleurotus species) and an estimated approach to the volatile compounds. Food Chem. 105, 1188–1194. doi: 10.1016/j.foodchem.2007.02.021

CrossRef Full Text | Google Scholar

Cagno, V., Androzzi, P., D’Alicarnasso, M., Silva, P. J., Mueller, M., Galloux, M., et al. (2018). Broad-spectrum non-toxic antiviral nanoparticles with a virucidal inhibition mechanism. Nat. Mater. 17, 195–203. doi: 10.1038/nmat5053

PubMed Abstract | CrossRef Full Text | Google Scholar

Cao, Y., Wu, S., and Dai, Y. (2012). Species clarification of the prize medicinal Ganoderma mushroom “Lingzhi”. Fungal Divers. 56, 49–62. doi: 10.1007/s13225-012-0178-5

CrossRef Full Text | Google Scholar

Cardozo, F. T., Camelini, C. M., Leal, P. C., Kratz, J. M., Nunes, R. J., Mendonça, M. M., et al. (2014). Antiherpetic mechanism of a sulfated derivative of Agaricus brasiliensis fruiting bodies polysaccharide. Intervirology 57, 375–383. doi: 10.1159/000365194

PubMed Abstract | CrossRef Full Text | Google Scholar

Cardozo, F. T., Camelini, C. M., Mascarello, A., Rossi, M. J., Barardi, C. R., de Mendonça, M. M., et al. (2011). Antiherpetic activity of a sulfated polysaccharide from Agaricus brasiliensis mycelia. Antiviral Res. 92, 108–114. doi: 10.1016/j.antiviral.2011.07.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Chan, Y. F., and Abu Bakar, S. (2005). Virucidal activity of Virkon S on human Enterovirus 71. Med. J. Malaysia 60, 246–248.

PubMed Abstract | Google Scholar

Chávez, R., Fierro, F., García-Rico, R. O., and Vaca, I. (2015). Filamentous fungi from extreme environments as a promising source of novel bioactive secondary metabolites. Front. Microbiol. 6:903. doi: 10.3389/fmicb.2015.00903

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, L., Shao, H. J., and Su, Y. B. (2004). Coimmunization of Agaricus blazei Murill extract with hepatitis B virus core protein through DNA vaccine enhances cellular and humoral immune responses. Int. Immunopharmacol. 4, 403–409. doi: 10.1016/j.intimp.2003.12.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, S., Xu, J., Liu, C., Zhu, Y., Nelson, D. R., Zhou, S., et al. (2012). Genome sequence of the model medicinal mushroom Ganoderma lucidum. Nat. Commun. 3:913. doi: 10.1038/ncomms1923

PubMed Abstract | CrossRef Full Text | Google Scholar

Cheung, R. C., Wong, J. H., Pan, W. L., Chan, Y. S., Yin, C. M., Dan, X. L., et al. (2014). Antifungal and antiviral products of marine organisms. Appl. Microbiol. Biotechnol. 98, 3475–3494. doi: 10.1007/s00253-014-5575-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Clercq, E. D. (2002). Strategies in the design of antiviral drugs. Nat. Rev. 1, 13–25.

Google Scholar

Clevanger, K. D., Bok, J. W., Ye, R., Miley, G. P., Verdan, M. H., Velk, T., et al. (2017). A scalable platform to identify fungal secondary metabolites and their gene clusters. Nat. Chem. Biol. 13, 895–905. doi: 10.1038/nchembio.2408

PubMed Abstract | CrossRef Full Text | Google Scholar

Collins, R. A., and Ng, T. B. (1997). Polysaccharopeptide from Coriolus versicolor has potential for use against human immunodeficiency virus type 1 infection. Life Sci. 60, 383–387. doi: 10.1016/S0024-3205(97)00294-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Cragg, G. M., and Newman, D. J. (2007). Natural products as sources of new drugs over the last 25 Years. J. Nat. Prod. 70, 461–477. doi: 10.1021/np068054v

PubMed Abstract | CrossRef Full Text | Google Scholar

Cui, S. W. (2005). “Structural analysis of polysaccharides,” in Food Carbohydrates, ed. S. W. Cui (Boca Raton, FL: CRC Press), 105–160.

Google Scholar

De Colibus, L. D., Wang, X., Spyrou, J. A. B., Kelly, J., Ren, J., Grimes, J., et al. (2014). More-powerful virus inhibitors from structure-based analysis of HEV71 capsid-binding molecules. Nat. Struct. Mol. Biol. 21, 282–288. doi: 10.1038/nsmb.2769

PubMed Abstract | CrossRef Full Text | Google Scholar

De Palma, A. M., Vliegen, I., De Clercq, E., and Neyts, J. (2008). Selective inhibitors of picornavirus replication. Med. Res. Rev. 28, 823–884. doi: 10.1002/med.20125

PubMed Abstract | CrossRef Full Text | Google Scholar

Desmukh, S. K., Prakash, V., and Ranjan, N. (2018). Marine fungi: a source of potential anticancer compounds. Front. Microbiol. 8:2536. doi: 10.3389/fmicb.2017.02536

CrossRef Full Text | Google Scholar

El Dine, R. S., El Halawany, A. M., Ma, C.-M., and Hattori, M. (2008). Anti-HIV-1 protease activity of lanostane triterpenes from the Vietnamese mushroom Ganoderma colossum. J. Nat. Prod. 71, 1022–1026. doi: 10.1021/np8001139

PubMed Abstract | CrossRef Full Text | Google Scholar

El-Mekkawy, S., Meselhy, M. R., Nakamura, N., Tezuka, Y., Hattori, M., Kakiuchi, N., et al. (1998). Anti-HIV-1 and anti-HIV-1-protease substances from Ganoderma lucidum. Phytochemistry 49, 1651–1657. doi: 10.1016/S0031-9422(98)00254-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Eo, S.-K., Kim, Y. S., Lee, C. K., and Han, S. S. (1999a). Antiherpetic activities of various protein bound polysaccharides isolated from Ganoderma lucidum. J. Ethnopharmacol. 68, 175–181. doi: 10.1016/S0378-8741(99)00086-0

CrossRef Full Text | Google Scholar

Eo, S.-K., Kim, Y.-O., Lee, C.-K., and Han, S.-S. (1999b). Antiviral activities of various water and methanol soluble substances isolated from Ganoderma lucidum. J. Ethnopharmacol. 68, 129–136.

PubMed Abstract | Google Scholar

Eo, S.-K., Kim, Y. S., Lee, C. K., and Han, S. S. (2000). Possible mode of antiviral activity of acidic protein bound polysaccharide isolated from Ganoderma lucidum on herpes simplex virus. J. Ethnopharmacol. 72, 475–481. doi: 10.1016/S0378-8741(00)00266-X

PubMed Abstract | CrossRef Full Text | Google Scholar

Faccin, L. C., Benati, F., Rincão, V. P., Mantovani, M. S., Soares, S. A., Gonzaga, M. L., et al. (2007). Antiviral activity of aqueous and ethanol extracts and of an isolated polysaccharide from Agaricus brasiliensis against poliovirus type 1. Lett. Appl. Microbiol. 45, 24–28. doi: 10.1111/j.1472-765X.2007.02153.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Fang, W., Lin, X., Zhou, X., Wan, J., Lu, X., Yang, B., et al. (2014). Cytotoxic and antiviral nitrobenzyl sesquiterpenoids from the marine-derived fungus Aspergillus ochraceus Jcma1F17. Med. Chem. Communn. 5, 701–705. doi: 10.1039/C3MD00371J

CrossRef Full Text | Google Scholar

Gao, H., Guo, W., Wang, Q., Zhang, L., Zhu, M., Zhu, T., et al. (2013a). Aspulvinones from a mangrove rhizosphere soil-derived fungus Aspergillus terreus Wwq-48 with anti-influenza A viral (H1N1) activity. Bioorgan. Med. Chem. 23, 1776–1778. doi: 10.1016/j.bmcl.2013.01.051

PubMed Abstract | CrossRef Full Text | Google Scholar

Gao, W., Sun, Y., Chen, S., Zhang, J., Kang, J., Wang, Y., et al. (2013b). Mushroom lectin enhanced immunogenicity of HBV DNA cavvine in C57BL/6 and HbsAg-transgenic mice. Vaccine 31, 2273–2280. doi: 10.1016/j.vaccine.2013.02.062

PubMed Abstract | CrossRef Full Text | Google Scholar

Gargano, M., van Griensven, L., Isikhuemhen, O., Lindequist, U., Venturella, G., Wasser, S. P., et al. (2017). Medicinal mushrooms: valuable biological resources of high exploitation potential. Plant Biosyst. 151, 548–565. doi: 10.1080/11263504.2017.1301590

CrossRef Full Text | Google Scholar

Geoghegan, I., Steinberg, G., and Gurr, S. (2017). The role of fungal cell wall in the infection of plants. Trends Microbiol. 25, 957–967. doi: 10.1016/j.tim.2017.05.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Grinde, B., Hetland, G., and Johnson, E. (2006). Effects on gene expression and viral load of medicinal extract from Agaricus blazei in patients with chronic hepatitis C infection. Int. Immunopharmacol. 6, 1311–1314. doi: 10.1016/j.intimp.2006.04.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Gu, C.-Q., Li, J.-W., Chao, F., Jin, M., Wang, X.-W., and Shen, Z.-Q. (2007). Isolation, identification and function of a novel anti-HSV-1 protein from Grifola frondosa. Antiviral Res. 75, 250–257. doi: 10.1016/j.antiviral.2007.03.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Guo, B., Dai, J. R., Ng, S., Huang, Y., Leong, C., Ong, W., et al. (2000). Cytonic acids A and B: novel tridepside inhibitors of hCMV protease from the endophytic fungus Cytonaema species. J. Nat. Prod. 63, 602–604. doi: 10.1021/np990467r

PubMed Abstract | CrossRef Full Text | Google Scholar

Guo, H., Sun, B., Gao, H., Chen, X., Liu, S., Yao, X., et al. (2009). Diketopiperazines from the Cordyceps-colonizing fungus Epicoccum nigrum. J. Nat. Prod. 72, 2115–2119. doi: 10.1021/np900654a

PubMed Abstract | CrossRef Full Text | Google Scholar

Han, X., Chakrabortti, A., Zhu, J., Liang, Z. X., and Li, J. (2016). Sequencing and functional annotation of the whole genome of the filamentous fungus Aspergillus westerdiijkiae. BMC Genomics 17:633. doi: 10.1186/s12864-016-2974-x

PubMed Abstract | CrossRef Full Text

Harvey, A. L., Edrada-Ebel, R., and Quinn, R. J. (2015). The re-emergence of natural products for drug discovery in the genomics era. Nat. Rev. Drug Discov. 14, 111–129. doi: 10.1038/nrd4510

PubMed Abstract | CrossRef Full Text | Google Scholar

Hawksworth, D., and Lücking, R. (2017). Fungal diversity revisited: 2.2 to 3.8 million species. Microbiol. Spectr. 5, FUNK-0052-2016. doi: 10.1128/microbiolspec.FUNK-0052-2016

PubMed Abstract | CrossRef Full Text | Google Scholar

Hazuda, D., Blau, C. U., Felock, P., Hastings, J., Pramanik, B., Wolfe, A., et al. (1999). Isolation and characterization of novel human immunodeficiency virus integrase inhibitors from fungal metabolites. Antivir. Chem. Chemother. 10, 63–70. doi: 10.1177/095632029901000202

PubMed Abstract | CrossRef Full Text | Google Scholar

He, F., Bao, J., Zhang, X.-Y., Tu, Z.-C., Shi, Y.-M., and Qi, S.-H. (2013). Asperterrestide A, a cytotoxic cyclic tetrapeptide from the marine-derived fungus Aspergillus terreus SCSGAF0162. J. Nat. Prod. 76, 1182–1186. doi: 10.1021/np300897v

PubMed Abstract | CrossRef Full Text | Google Scholar

Hennicke, F., Cheikh-Ali, Z., Liebisch, T., Maciá-Vicente, J. G., Bode, H. B., and Piepenbring, M. (2016). Distinguishing commercially grown Ganoderma lucidum from Ganoderma lingzhi from Europe and East Asia on the basis of morphology, molecular phylogeny, and triterpenic acid profiles. Phytochemistry 127, 29–37. doi: 10.1016/j.phytochem.2016.03.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Hewat, E. A., and Blaas, D. (2004). Cryoelectron microscopy analysis of the structural changes associated with human rhinovirus type 14 uncoating. J. Virol. 78, 2935–2942. doi: 10.1128/JVI.78.6.2935-2942.2004

PubMed Abstract | CrossRef Full Text | Google Scholar

Hirose, K., Hakozaki, M., Kakuchi, J., Matsunaga, K., Yoshikumi, C., Takahashi, M., et al. (1987). A biological response modifier, PSK, inhibits reverse transcriptase in vitro. Biochem. Biophys. Res. Commun. 149, 562–567. doi: 10.1016/0006-291X(87)90404-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Hoffmeister, D., and Keller, N. P. (2007). Natural products of filamentous fungi: enzymes, genes, and their regulation. Nat. Prod. Rep. 24, 393–416. doi: 10.1039/B603084J

PubMed Abstract | CrossRef Full Text | Google Scholar

Hu, Y., Ahmed, S., Li, J., Luo, B., Gao, Z., Zhang, Q., et al. (2017). Improved ganoderic acids production in Ganoderma lucidum by wood decaying components. Sci. Rep. 7:46623. doi: 10.1038/srep46623

PubMed Abstract | CrossRef Full Text | Google Scholar

Ichimura, T., Watanabe, O., and Muruyama, S. (1998). Inhibition of HIV-1 protease by water-soluble lignin-like substance from an edible mushroom, Fuscoporia oblique. Biosci. Biotechnol. Biochem. 62, 575–577. doi: 10.1271/bbb.62.575

PubMed Abstract | CrossRef Full Text | Google Scholar

Inglis, D. O., Binkley, J., Skrzypek, M. S., Arnaud, M. B., Cerqueira, G. C., and Shah, P. (2013). Comprehensive annotation of secondary metabolite biosynthetic genes and gene clusters in Aspergillus nidulans, A. fumigatus, A. niger and A. oryzae. BMC Microbiol. 13:91. doi: 10.1186/1471-2180-13-91

PubMed Abstract | CrossRef Full Text | Google Scholar

Isaka, M., Berkaew, P., Intereya, K., Komwijit, S., and Sathitkunanon, T. (2007). Antiplasmodial and antiviral cyclohexadepsipeptides from the endophytic fungus Pullularia sp. BCC 8613. Tetrahedron 29, 6855–6860. doi: 10.1016/j.tet.2007.04.062

CrossRef Full Text | Google Scholar

Iwatsuki, K., Akhisa, T., Tokuda, H., Ukiya, M., Oshikubo, M., Kimura, Y., et al. (2003). Lucidenic acids P and Q, methyl lucidenate P, and other triterpenoids from the fungus Ganoderma lucidum and their inhibitory effects of Epstein-Barr virus activation. J. Nat. Prod. 66, 1582–1585. doi: 10.1021/np0302293

PubMed Abstract | CrossRef Full Text | Google Scholar

Jassim, S. A., and Naji, M. A. (2003). Novel antiviral agents: a medicinal plant perspective. J. Appl. Microbiol. 95, 412–427. doi: 10.1046/j.1365-2672.2003.02026.x

CrossRef Full Text | Google Scholar

Jayasuriya, H., Guan, Z., Polishook, J. D., Dombrowski, A. W., Felock, P. J., Hazuda, D. J., et al. (2003). Isolation, structure, and HIV-1 integrase inhibitory activity of cytosporic acid, a fungal metabolite produced by Cytospora sp. J. Nat. Prod. 66, 551–553. doi: 10.1021/np020533g

PubMed Abstract | CrossRef Full Text | Google Scholar

Jia, Y.-L., Guan, F.-F., Ma, J., Wang, C.-Y., and Shao, C.-L. (2015). Pestalotiolide A, a new antiviral phthalide derivative from a soft coral-derived fungus Pestalotiopsis sp. Nat. Prod. Sci. 21, 227–230. doi: 10.20307/nps.2015.21.4.227

CrossRef Full Text | Google Scholar

Jiang, Y., Wong, J. H., Ng, T. B., Liu, Z. K., Wang, C. R., Li, N., et al. (2011). Isolation of adenosine, iso-sinensetin and dimethylguanosine with antioxidant and HIV-1 protease inhibiting activities from fruiting bodies of Cordyceps militaris. Phytomedicine 18, 189–193. doi: 10.1016/j.phymed.2010.04.010

PubMed Abstract | CrossRef Full Text | Google Scholar

Kalač, P. (2009). Chemical composition and nutritional value of European species of wild growing mushrooms: a review. Food Chem. 113, 9–16. doi: 10.1080/10590500802350086

PubMed Abstract | CrossRef Full Text | Google Scholar

Kandefer-Szerszeń, M., Kawechi, Z., Sałata, B., and Witek, M. (1980). Mushrooms as a source of substances with antiviral activity. Acta Mycol. 16, 215–220. doi: 10.5586/am.1980.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Kanokmedhakul, S., Kanokmedhakul, K., Prajuabsuk, T., Soytong, K., Kongsaeree, P., and Suksamrarn, A. (2003). A bioactive triterpenoid and vulpinic acid derivatives from the mushroom Scleroderma citrinum. Planta Med. 69, 568–571. doi: 10.1055/s-2003-40639

PubMed Abstract | CrossRef Full Text | Google Scholar

Keller, N. P., Turner, G., and Bennett, J. W. (2005). Fungal secondary metabolism - from biochemistry to genomics. Nat. Rev. Microbiol. 3, 937–947. doi: 10.1038/nrmicro1286

PubMed Abstract | CrossRef Full Text | Google Scholar

Khaldi, N., Seifuddin, F. T., Turner, G., Haft, D., Nierman, W. C., Wolfe, K. H., et al. (2010). SMURF: genomic mapping of fungal secondary metabolites. Fungal Genet. Microbiol. 47, 736–741. doi: 10.1016/j.fgb.2010.06.003

PubMed Abstract | CrossRef Full Text

Kim, Y.-S., Eo, S.-K., Oh, K.-W., Lee, C.-K., and Han, S.-S. (2000). Antiherpetic activities of acidic protein bound polysaccharide isolated from Ganoderma lucidum alone and in combinations with interferons. J. Ethnopharmacol. 72, 451–458. doi: 10.1016/S0378-8741(00)00263-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Krawczyk, E., Łuczak, M., Kobus, M., Bańka, D., and Daniewski, W. (2003). Antiviral activity of ?-benzoylphenylisoserinates of Lactarius sesquiterpenoid alcohols in vitro. Planta Med. 69, 552–554. doi: 10.1055/s-2003-40649

PubMed Abstract | CrossRef Full Text | Google Scholar

Krohn, K., Bahramsari, R., Flörke, U., Ludewig, K., Kliche-Spory, C., Michel, A., et al. (1997). Dihydrocoumarins from fungi: isolation, structure elucidation, circular dichroism and biological activity. Phytochemistry 45, 313–320. doi: 10.1016/S0031-9422(96)00854-0

CrossRef Full Text | Google Scholar

Krupodorova, T., Rybalko, S., and Barshteyn, V. (2014). Antiviral activity of Basidiomycete mycelia against influenza type A (serotype H1N1) and herpes simplex virus type 2 in cell culture. Virol. Sin. 29, 284–290. doi: 10.1007/s12250-014-3486-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Kumaresan, V., and Suryanarayanan, T. S. (2001). Occurrence and distribution of endophytic fungi in a mangrove community. Mycol. Res. 105, 1388–1391. doi: 10.1017/S0953756201004841

CrossRef Full Text | Google Scholar

Kusari, S., Hertweck, C., and Spiteller, M. (2012). Chemical ecology of endophytic fungi: origins of secondary metabolites. Chem. Biol. 19, 792–798. doi: 10.1016/j.chembiol.2012.06.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Kusari, S., Singh, S., and Jayabaskaran, C. (2014a). Biotechnological potential of plant-associated endophytic fungi: hope versus hype. Trends Biotechnol. 32, 297–303. doi: 10.1016/j.tibtech.2014.03.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Kusari, S., Singh, S., and Jayabaskaran, C. (2014b). Rethinking production of Taxol(R) (paclitaxel) using endophyte biotechnology. Trends Biotehnol. 32, 303–311. doi: 10.1016/j.tibtech.2014.03.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Lehmann, V. K. B., Huan, A., Ibanez-Calero, S., Wilson, G. R., and Rinehart, K. L. (2003). Illudin S, the sole antiviral compound in mature fruiting bodies of Omphalotus illudens. J. Nat. Prod. 66, 1257–1258. doi: 10.1021/np030205w

PubMed Abstract | CrossRef Full Text | Google Scholar

Levy, H. C., Bostina, M., Filman, D. J., and Hogle, J. M. (2010). Catching a virus in the act of RNA release: a novel poliovirus uncoating intermediate characterized by cryo-electron microscopy. J. Virol. 84, 4426–4441. doi: 10.1128/JVI.02393-09

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, E., Tian, R., Liu, S., Chen, X., Guo, L., and Che, Y. (2008). Pestalotheols A-D, bioactive metabolites from the plant endophytic fungus Pestalotiopsis theae. J. Nat. Prod. 71, 664–668. doi: 10.1021/np700744t

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, Y., Liu, D., Cen, S., Proksch, P., and Lin, W. (2014). Isoindolinone-type alkaloids from the sponge-derived fungus Stachybotrys chartarum. Tetrahedron 70, 7010–7015. doi: 10.1016/j.tet.2014.07.047

CrossRef Full Text | Google Scholar

Liu, J., Yang, F., Ye, L. B., Yang, X. J., Timani, K. A., Zheng, Y., et al. (2004). Possible mode of action of antiherpetic activities of a proteoglycan isolated from the mycelia of Ganoderma lucidum in vitro. J. Ethnopharmacol. 95, 265–272. doi: 10.1016/j.jep.2004.07.010

PubMed Abstract | CrossRef Full Text | Google Scholar

Ma, X., Zhu, T., Ba, M., Li, G., Gu, Q., Guo, Y., et al. (2013). Phenylspirodrimanes with anti-HIV activity from the sponge-derived fungus Stachybotrys chartarum MXH-X73. J. Nat. Prod. 76, 2298–2306. doi: 10.1021/np400683h

PubMed Abstract | CrossRef Full Text | Google Scholar

Mahé, S., Rédou, V., Calvez, T. L., Vandenkoornhuyse, P., and Burgaud, G. (2013). “Fungi in deep-sea environments and metagenomics,” in The Ecological Genomics of Fungi, ed. F. Martin (Hoboken, NJ: John Wiley & Sons Inc), 325–354. doi: 10.1002/9781118735893.ch15

CrossRef Full Text | Google Scholar

Marjomäki, V., Pietiäinen, V., Matilaien, H., Upla, P., Ivaska, J., Nissinen, L., et al. (2002). Internalization of echovirus 1 in caveolae. J. Virol. 76, 1856–1865. doi: 10.1128/JVI.76.4.1856-1865.2002

CrossRef Full Text | Google Scholar

Marjomäki, V., Turkki, P., and Huttunen, M. (2015). Infectious entry pathway of Enterovirus B species. Viruses 7, 6387–6399. doi: 10.3390/v7122945

PubMed Abstract | CrossRef Full Text | Google Scholar

Martikainen, M., Salorinne, K., Lahtinen, T., Malola, S., Permi, P., Häkkinen, H., et al. (2015). Hydrophobic pocket targeting probes for Enteroviruses. Nanoscale 7, 17457–17467. doi: 10.1039/c5nr04139b

PubMed Abstract | CrossRef Full Text | Google Scholar

Matsuhisa, K., Yamane, S., Okamoto, T., Atari, A., Kondoh, M., Matsuura, Y., et al. (2015). Anti-HVC effect of Lentinula edodes mycelia solid culture extracts an low-molecular with lignin. Biochem. Biophys. Res. Communn. 462, 52–57. doi: 10.1016/j.bbrc.2015.04.104

PubMed Abstract | CrossRef Full Text | Google Scholar

Matsuzaki, K., Ikeda, H., Masuma, R., Tanaka, H., and Omura, S. (1995). Isochromophilones I and II, novel inhibitors agains gp120-CD4 binding produced by Penicillium multicolor FO-2338. J. Antibiot. 48, 703–707. doi: 10.7164/antibiotics.48.703

CrossRef Full Text | Google Scholar

Mayer, A. M., Rodriguez, A. D., Taglialatela-Scafati, O., and Fusetani, N. (2013). Marine pharmacology in 2009-2011: marine compounds with antibacterial, antidiabetic, antifungal, anti-inflammatory, antiprotozoal, antituberculosis, and antiviral activities; affecting the immune and nervous systems, and other miscellaneous mechanisms of action. Mar. Drugs 11, 2510–2573. doi: 10.3390/md11072510

PubMed Abstract | CrossRef Full Text | Google Scholar

Min, B.-S., Nakamura, N., Miyashiro, H., Bae, K.-W., and Hattori, M. (1998). Triterpenes from the spores of Ganoderma lucidum and their inhibitory against HIV-1 protease. Chem. Pharm. Bull. 46, 1607–1612. doi: 10.1248/cpb.46.1607

PubMed Abstract | CrossRef Full Text | Google Scholar

Minagawa, K., Kouzoki, S., Yoshimoto, J., Kawamura, Y., Tani, H., Iwata, T., et al. (2002). Stachyflin and acetylstachyflin, novel anti-influenza A virus substances, produced by Stachybotrys sp. RF-7260. I. Isolation, structure elucidation and biological activities. J. Antibiot. 55, 155–164. doi: 10.7164/antibiotics.55.155

PubMed Abstract | CrossRef Full Text | Google Scholar

Mizerska-Dudka, M., Jaszek, M., Błachowicz, A., Rejczak, T. P., Maturszewska, A., Osińska-Jaroszuk, M., et al. (2015). Fungus Cerrena unicolor as an effective source of new antiviral, immunomodulatory, and anticancer compounds. Int. J. Biol. Macromol. 79, 459–468. doi: 10.1016/j.ijbiomac.2015.05.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Mlinaric, A., Kac, J., and Pohleven, F. (2005). Screening of selected wood-damaging fungi for the HIV-1 reverse transcriptase inhibitors. Acta Pharm. 55, 69–79.

PubMed Abstract | Google Scholar

Moncalvo, J. M., and Ryvarden, L. (1997). A Nomenclatural Study of the Ganodermataceae Donk, Synopsis Fungorum 11. Oslo: Fungiflora.

Moncalvo, J. M., Wang, H. H., and Hseu, R. S. (1995). Gene phylogeny of the Ganoderma lucidum complex based on ribosomal DNA sequences. Comparison with traditional taxonomic characters. Mycol. Res. 99, 1489–1499. doi: 10.1016/S0953-7562(09)80798-3

CrossRef Full Text | Google Scholar

Mothana, R. A., Awadh Ali, N. A., Jansen, R., Wegner, U., Mentel, R., and Lindequist, U. (2003). Antiviral lanostanoid triterpenes from the fungus Ganoderma pfeifferi. Fitoterapia 74, 177–180. doi: 10.1016/S0367-326X(02)00305-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Myllynen, M., Kazmertsuk, A., and Marjomäki, V. (2016). A novel open and infectious form of Echovirus 1. J. Virol. 90, 6759–6770. doi: 10.1128/JVI.00342-16

PubMed Abstract | CrossRef Full Text | Google Scholar

Ngai, P. H., and Ng, T. B. (2003). Lentin, a novel and potent antifungal protein from shitake mushroom with inhibitory effects on activity of human immunodeficiency virus-1 reverse transcriptase and proliferation of leukemia cells. Life Sci. 73, 3363–3374. doi: 10.1016/j.lfs.2003.06.023

PubMed Abstract | CrossRef Full Text | Google Scholar

Niedermeyer, T. H., Lindequist, U., Mentel, R., Gördes, D., Schmidt, E., Thurow, K., et al. (2005). Antiviral terpenoids constituents of Ganoderma pfeifferi. J. Nat. Prod. 68, 1728–1731. doi: 10.1021/np0501886

PubMed Abstract | CrossRef Full Text | Google Scholar

Ochi, K., and Hosaka, T. (2013). New strategies for drug discovery: activation of silent or weakly expressed microbial gene clusters. Appl. Microbiol. Biotechnol. 97, 87–98. doi: 10.1007/s00253-012-4551-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Okada, M., and Minamishima, Y. (1987). The effect of biological response modifiers on chronic and latent murine cytomegalovirus infections. Microbial. Immunol. 31, 435–447. doi: 10.1111/j.1348-0421.1987.tb03106.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Omura, S., Tanaka, H., Matsuzaki, K., Ikeda, H., and Masuma, R. (1993). Isochromophilones I and II, novel inhibitors against gp120-CD4 binding from Penicillium sp. J. Antibiot. 46, 1908–1911. doi: 10.7164/antibiotics.46.1908

PubMed Abstract | CrossRef Full Text | Google Scholar

Ondeyka, J. G., Zink, D., Dombrowski, A. W., Polishhook, J. D., Felock, P. J., Hazuda, D. J., et al. (2003). Isolation, structure and HIV-1 integrase inhibitory activity of exophilic acid, a novel fungal metabolites from Exophiala pisciphila. J. Antibiot. 56, 1018–1023. doi: 10.7164/antibiotics.56.1018

CrossRef Full Text | Google Scholar

Ouzouni, P. K., Petridis, D., Koller, W.-D., and Riganakos, K. A. (2009). Nutritional value and metal content of wild edible mushrooms collected from West Macedonia and Epirus, Greece. Food Chem. 115, 1575–1580. doi: 10.1016/j.foodchem.2009.02.014

CrossRef Full Text | Google Scholar

Pang, X., Lin, X., Tian, Y., Liang, R., Wang, J., and Yang, B. (2018). Three new polyketides from the marine sponge-derived fungus Trichoderma sp. SCSIO41004. Nat. Prod. Res. 32, 105–111. doi: 10.1080/14786419.2017.1338286

PubMed Abstract | CrossRef Full Text | Google Scholar

Peng, J., Lin, T., Wang, W., Xin, Z., Zhu, T., Gu, Q., et al. (2013). Antiviral alkaloids produced by the mangrove-derived fungus Cladosporium sp. PJX-41. J. Nat. Prod. 76, 1133–1140. doi: 10.1021/np400200k

PubMed Abstract | CrossRef Full Text | Google Scholar

Peng, J., Zhang, X., Du, L., Wang, W., Zhu, T., Gu, Q., et al. (2014). Sorbicatechols A and B, antiviral sorbicillinoids from the marine-derived fungus Penicillium chrysogenum PJX-17. J. Nat. Prod. 77, 424–428. doi: 10.1021/np400977e

PubMed Abstract | CrossRef Full Text | Google Scholar

Pérez, M., Soler-Torronteras, R., Collado, J. A., Limones, C. G., Hellsten, R., Johansson, M., et al. (2014). The fungal metabolite galiellalactone interferes with the nuclear import of NF-κB and inhibits HIV-replication. Chem. Biol. Interact. 214, 69–75. doi: 10.1016/j.cbi.2014.02.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Piraino, F., and Brandt, C. R. (1999). Isolation and partial characterization of an antiviral, RC-183, from the edible mushroom Rozites caperata. Antivir. Res. 43, 67–78. doi: 10.1016/S0166-3542(99)00035-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Pittayakhajonwut, P., Suvannakad, R., Thienhirun, S., Prabpai, S., Kongsaeree, P., and Tanticharoen, M. (2005). An anti-herpes simplex virus-type 1 agent from Xylaria mellissii (BCC 1005). Tetrahedron Lett. 46, 1341–1344. doi: 10.1016/j.tetlet.2004.12.110

CrossRef Full Text | Google Scholar

Posch, A. E., Herwig, C., and Spadiut, O. (2013). Science-based bioprocess design for filamentous fungi. Trends Biotechnol. 31, 37–44. doi: 10.1016/j.tibtech.2012.10.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Raekiansyah, M., Mori, M., Nonaka, K., Agoh, M., Shiomi, K., Matsumoto, A., et al. (2017). Identification of novel antiviral of fungus-derived brefeldin A against dengue virus. Trop. Med. Health 45:32. doi: 10.1186/s41182-017-0072-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Raja, H. A., Miller, A. N., Pearce, C. J., and Oberlies, N. H. (2017). Fungal identification using molecular tools: a primer for the natural products research community. J. Nat. Prod. 80, 757–770. doi: 10.1021/acs.jnatprod.6b01085

PubMed Abstract | CrossRef Full Text | Google Scholar

Razumov, I. A., Kosogova, T. A., Kazachinskaia, E. I., Puchkova, L. I., Shcherbakova, N. S., Gorbunova, I. A., et al. (2010). Antiviral activity of aqueous extracts and polysaccharide fractions from mycelium and fruit bodies of higher fungi. Antibiot. Khimioter. 55, 14–18.

PubMed Abstract | Google Scholar

Reis, F. S., Barros, L., Martins, A., and Ferreira, I. C. (2012). Chemical composition and nutritional value of the most widely appreciated cultivated mushrooms: an inter-species comparative study. Food. Chem. Toxicol. 50, 191–197. doi: 10.1016/j.fct.2011.10.056

PubMed Abstract | CrossRef Full Text | Google Scholar

Richter, C., Wittstein, C., Kirk, P. M., and Stadler, M. (2015). An assessment of the taxonomy and chemotaxonomy of Ganoderma. Fungal Divers. 71, 1–15. doi: 10.1007/s13225-014-0313-6

CrossRef Full Text | Google Scholar

Rincão, V. P., Yamamoto, K. A., Ricardo, N. M., Soares, S. A., Meirelles, L. D., Nozawa, C., et al. (2012). Polysaccharides and extracts from Lentinula edodes: structural features and antiviral activity. Virol. J. 15, 37. doi: 10.1186/1743-422X-9-37

PubMed Abstract | CrossRef Full Text

Rowley, D. C., Kelly, S., Kauffman, C. A., Jensen, P. R., and Fenical, W. (2003). Halovirs A-E, new antiviral agents from marine-derive fungus of the genus Scytalidium. Bioorgan. Med. Chem. 11, 4263–4274. doi: 10.1016/S0968-0896(03)00395-X

PubMed Abstract | CrossRef Full Text | Google Scholar

Saboulard, D., Gaspar, A., Roussel, B., and Villard, J. (1998). New antiherpetic nucleoside from a Basidiomycete. C. R. Acad. Sci. 321, 585–591. doi: 10.1016/S0764-4469(98)80461-7

CrossRef Full Text | Google Scholar

Sacramento, C. Q., Marttorelli, A., Fintelman-Rodrigues, N., de Freitas, C. S., de Melo, G. R., Rocha, M. E., et al. (2015). Aureonitol, a fungi-derived tetrahydrofuran, inhibits influenza replication by targeting its surface glycoprotein hemagglutin. PLoS One 10:e0139236. doi: 10.1371/journal.pone.0139236

PubMed Abstract | CrossRef Full Text | Google Scholar

Saleem, M., Ali, M. S., Hussain, S., Jabbar, A., Ashraf, M., and Lee, Y. S. (2007). Marine natural products of fungal origin. Nat. Prod. Rep. 24, 1142–1152. doi: 10.1039/b607254m

PubMed Abstract | CrossRef Full Text | Google Scholar

Sarkar, S., Koga, J., Whitley, R. J., and Chatterjee, S. (1993). Antiviral effect of the extract of culture medium of Lentinus edodes mycelia on the replication of herpex simplex virus type I. Antiviral Res. 20, 293–303. doi: 10.1016/0166-3542(93)90073-R

PubMed Abstract | CrossRef Full Text | Google Scholar

Sato, N., Zhang, Q., Ma, C. M., and Hattori, M. (2009). Anti-human immunodeficiency virus-1 protease activity of new lanostane-type triterpenoids from Ganoderma sinense. Chem. Pharm. Bull. 57, 1076–1080. doi: 10.1248/cpb.57.1076

PubMed Abstract | CrossRef Full Text | Google Scholar

Sawadjoon, S., Kittakoop, P., Isaka, M., Madla, S., and Thebtaranonth, Y. (2004). Antiviral and antiplasmodial spirodihydrobenzofuran terpenes from the fungus Stachybotrys nephospora. Planta Med. 70, 1085–1087. doi: 10.1055/s-2004-832652

PubMed Abstract | CrossRef Full Text | Google Scholar

Schmidtke, M., Schnittler, U., Jahn, B., Dahse, H., and Stelzner, A. (2001). A rapid assay for evaluation of antiviral activity against coxsackie virus B3, influenza virus A, and herpes simplex virus type 1. J. Virol. Methods 95, 133–143. doi: 10.1016/S0166-0934(01)00305-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Schulz, B., and Boyle, C. (2005). The endophytic continuum. Mycol. Res. 109, 661–686. doi: 10.1017/S095375620500273X

CrossRef Full Text | Google Scholar

Sebastian, L., Madhusudana, S. N., Ravi, V., and Desai, A. (2011). Mycophenolic acid inhibits replication of Japanese encephalitis virus. Chemotherapy 57, 56–61. doi: 10.1159/000321483

PubMed Abstract | CrossRef Full Text | Google Scholar

Shiao, M. S. (2003). Natural products of the medicinal fungus Ganoderma lucidum: occurrence, biological activities, and pharmacological functions. Chem. Res. 3, 172–180.

PubMed Abstract | Google Scholar

Shiomi, K., Matsui, R., Isozaki, M., Chiba, H., Sugai, T., Yamguchi, Y., et al. (2005). Fungal phenalenones inhibit HIV-1 integrase. J. Antibiot. 58, 65–68. doi: 10.1038/ja.2005.8

PubMed Abstract | CrossRef Full Text | Google Scholar

Shushni, M. A., Singh, R., Mentel, R., and Lindequist, U. (2011). Balticolid: a new 12-membered macrolide with antiviral activity from an Ascomycetous fungus of marine origin. Mar. Drugs 9, 844–851. doi: 10.3390/md9050844

PubMed Abstract | CrossRef Full Text | Google Scholar

Singh, R. P., Kumari, P., and Reddy, C. R. (2015). Antimicrobial compounds from seaweeds-associated bacteria and fungi. Appl. Microbiol. Biotechnol. 99, 1571–1586. doi: 10.1007/s00253-014-6334-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Singh, S. B., Ondeyka, J. G., Tsipouras, N., Ruby, C., Sardana, V., Schulman, M., et al. (2004). Hinnuliquinone, a C2-symmetric dimeric non-peptide fungal metabolite inhibitor of HIV-1 protease. Biochem. Biophys. Res. Commun. 324, 108–113. doi: 10.1016/j.bbrc.2004.08.234

PubMed Abstract | CrossRef Full Text | Google Scholar

Singh, S. B., Zink, D. B., Quamina, D. S., Pelaez, F., Teran, A., Felock, P., et al. (2002). Integrastatins: structure and HIV-1 integrase inhibitory activities of two novel racemic tetracyclic aromatic heterocycles produced by two fungal species. Tetrahedron Lett. 43, 2351–2354. doi: 10.1016/S0040-4039(02)00265-4

CrossRef Full Text | Google Scholar

Singh, S. B., Jayasuriya, H., Dewey, R., Polishook, J. D., Dombrowski, A. W., Zink, D. L., et al. (2003a). Isolation, structure, and HIV-1-integrase inhibitory activity of structurally diverse fungal metabolites. J. Ind. Microbiol. Biotechnol. 30, 721–731.

PubMed Abstract | Google Scholar

Singh, S. B., Zink, D. L., Dombrowski, A. W., Polishook, J. D., Ondeyka, J. G., Hirshfield, J., et al. (2003b). Integracides: tetracyclic triterpenoid inhibitors of HIV-1 integrase produced by Fusarium sp. Bioorg. Med. Chem. 11, 1577–1582.

PubMed Abstract | Google Scholar

Sorimachi, K., Ikehara, Y., Maezato, G., Okubo, A., Yamazaki, S., Akimoto, K., et al. (2001). Inhibition by Agaricus blazei Murill fractions of cytopathic effect induced by Western Equine Encephalitis (WEE) virus on VERO cells in vitro. Bioschi. Biotechnol. Biochem. 65, 1645–1647. doi: 10.1271/bbb.65.1645

PubMed Abstract | CrossRef Full Text | Google Scholar

Sorimachi, K., Niwa, A., Yamazaki, A., Toda, S., and Yasumura, Y. (1990). Anti-viral activity of water-solubilized lignin derivatives in vitro. Agric. Biol. Chem. 54, 1337–1339. doi: 10.1007/s00253-014-6334-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Steyaert, R. L. (1972). Species of Ganoderma and related genera mainly of the Bogor and Leiden Herbaria. Persoonia 7, 55–118.

Google Scholar

Stierle, A. A., and Stierle, D. B. (2015). Bioactive secondary metabolites produced by the fungal endophytes of conifers. Nat. Prod. Communn. 10, 1671–1682.

Google Scholar

Strobel, G., and Daisy, B. (2003). Bioprospecting for microbial endophytes and their natural products. Microbiol. Mol. Biol. Rev. 67, 491–502. doi: 10.1128/MMBR.67.4.491-502.2003

CrossRef Full Text | Google Scholar

Suzuki, H., Okubo, A., Yamazuki, S., Suzuki, K., Mitsuya, H., and Toda, S. (1989). Inhibition of the infectivity and cytopathic effect of human immunodeficiency virus by water-soluble lignin in an extract of the culture medium of Lentinus edodes mycelia (LEM). Biochem. Biophys. Res. Commun. 160, 367–373. doi: 10.1016/0006-291X(89)91665-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Teplyakova, T. V., Psurtseva, N. V., Kosogova, N. V., Mazurkova, T. A., Khanin, V. A., and Viasenko, V. A. (2012). Antiviral activity of polyporoid mushrooms (higher Basidiomycetes) from Altai Mountains (Russia). Int. J. Med. Mushrooms 14, 37–45. doi: 10.1615/IntJMedMushr.v14.i1.40

PubMed Abstract | CrossRef Full Text | Google Scholar

Thatoi, H., Behera, B. C., and Mishra, R. R. (2013). Ecological role and biotechnological potential of mangrove fungi: a review. Mycology 4, 54–71.

Google Scholar

Tochikura, T. S., Nakashima, H., Hirose, K., and Yamamoto, N. (1987). A biological response modifier, PSK, inhibits human immunodeficiency virus infection in vitro. Biochem. Biophys. Res. Commun. 2, 726–733. doi: 10.1016/0006-291X(87)90936-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Tochikura, T. S., Nakashima, H., Ohashi, Y., and Yamamoto, N. (1988). Inhibition (in vitro) of replication and of the cytopathic effect of human immunodeficiency virus by an extract of the culture medium of Lentinus edodes mycelia. Med. Microbiol. Immunol. 177, 235–244. doi: 10.1007/BF00189409

PubMed Abstract | CrossRef Full Text | Google Scholar

Van der Linden, L., Wolthers, K. C., and van Kuppeveld, F. J. (2015). Replication and inhibitors of Enteroviruses and parechoviruses. Viruses 7, 4529–4562. doi: 10.3390/v7082832

PubMed Abstract | CrossRef Full Text | Google Scholar

Vigant, F., Santos, N. C., and Lee, B. (2015). Broad-spectrum antivirals against viral fusion. Nat. Rev. Microbiol. 13, 426–437. doi: 10.1038/nrmicro3475

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, D.-M., Wu, S.-H., Su, C.-H., Peng, J.-T., Shih, Y.-H., and Chen, L.-C. (2009). Ganoderma multipileum, the correct name for ‘G. lucidum’ in tropical Asia. Bot. Stud 50, 451–458.

Google Scholar

Wang, H., and Ng, T. B. (2000). Isolation of a novel ubiquitin-like protein from Pleurotus ostreatus mushroom with anti-human immunode?ciency virus, translation-inhibitory, and ribonuclease activities. Biochem. Biophys. Res. Commun. 276, 587–593. doi: 10.1006/bbrc.2000.3540

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, H., and Ng, T. B. (2001). Isolation and characterization of velutin, a novel low-molecular-weight ribosome-inactivating protein from winter mushroom (Flammulina velutipes) fruiting bodies. Life Sci. 68, 2151–2158. doi: 10.1016/S0024-3205(01)01023-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, J., Wang, H. X., and Ng, T. B. (2007). A peptide with HIV-1 reverse transcriptase inhibitory activity from the medicinal mushroom Russula paludosa. Peptides 28, 560–565. doi: 10.1016/j.peptides.2006.10.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, J., Wei, X., Lu, X., Lu, X., Wan, J., Lin, X., et al. (2014). Eight new polyketide metabolites from the fungus Pestalotiopsis vaccinii endogenous with the mangrove plant Kandelia candel (L.) Druce. Tetrahedron 70, 9695–9701. doi: 10.1016/j.tet.2014.10.056

CrossRef Full Text | Google Scholar

Wang, J.-F., Lin, X.-P., Qin, C., Liao, S.-R., Wan, J.-T., Zhang, T. Y., et al. (2014). Antimicrobial and antiviral sesquiterpenes from sponge-derived fungus, Aspergillus sydowii ZSDS1-F6. J. Antibiot. 67, 581–583. doi: 10.1038/ja.2014.97

PubMed Abstract | CrossRef Full Text | Google Scholar

Welti, S., Moreau, P.-A., Decock, C., Danel, C., Duhal, N., Favel, A., et al. (2015). Oxygenated lanostane-type triterpenes profiling in laccate Ganoderma chemotaxonomy. Mycol. Prog. 14:45. doi: 10.1007/s11557-015-1066-7

CrossRef Full Text | Google Scholar

Wu, D.-T., Deng, Y., Chen, L.-X., Zhao, J., Bzhelyansky, A., and Li, S.-P. (2017). Evaluation on quality consistency of Ganoderma lucidum dietary supplements collected in the United States. Sci. Rep. 7:7792. doi: 10.1038/s41598-017-06336-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, G., Sun, X., Yu, G., Wang, W., Zhu, T., Gu, Q., et al. (2014). Cladosins A-E, hybrid polyketides from a deep-sea-derived fungus, Cladosporium sphaerospermum. J. Nat. Prod. 77, 270–275. doi: 10.1021/np400833x

PubMed Abstract | CrossRef Full Text | Google Scholar

Wutzler, P., and Sauerbrei, A. (2004). Virucidal activity of the new disinfectant monopercitric acid. Lett. Appl. Microbiol. 39, 194–198. doi: 10.1111/j.1472-765X.2004.01561.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Yamamoto, K. A., Galhardi, L. C., Rincão, V. P., Soares Sde, A., Ricardo, N. M., Nozawa, C., et al. (2013). Antiherpetic activity of an Agaricus brasiliensis polysaccharide, its sulfated derivative and fractions. Int. J. Biol. Macromol. 52, 9–13. doi: 10.1016/j.ijbiomac.2012.09.029

PubMed Abstract | CrossRef Full Text | Google Scholar

Yim, G., Wang, H. H., and Davies, J. (2007). Antibiotics as signaling molecules. Philos. Trans. Soc. B 362, 1195–1200. doi: 10.1098/rstb.2007.2044

PubMed Abstract | CrossRef Full Text | Google Scholar

Yoshimoto, J., Kakui, M., Iwasaki, H., Fujiwara, T., Sugimoto, H., and Hattori, N. (1999). Identification of a novel HA conformational change inhibitor of human influenza virus. Arch. Virol. 144, 865–878. doi: 10.1007/s007050050552

CrossRef Full Text | Google Scholar

Yu, G., Zhou, G., Zhu, M., Wang, W., Gu, Q., and Li, D. (2016). Neosartoryadins A and B, fumiquinazoline alkaloids from a mangrove-derived fungus Neosartorya udagawae HDN13-313. Org. Lett. 18, 244–247. doi: 10.1021/acs.orglett.5b02964

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, D., Tao, X., Chen, R., Liu, J., Fang, X., Yu, L., et al. (2015). Pericoannosin A, a polyketide synthase-nonribosomal peptide synthetase hybrid metabolite with new carbon skeleton from the endophytic fungus Periconia sp. Org. Lett. 17, 4304–4307. doi: 10.1021/acs.orglett.5b02123

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, G., Sun, S., Zhu, T., Lin, Z., Gu, J., Li, D., et al. (2011). Antiviral isoindolone derivatives from an endophytic fungus Emericella sp. associated with Aegiceras corniculatum. Phytochemistry 72, 1436–1442. doi: 10.1016/j.phytochem.2011.04.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, S. P., Huang, R., Li, F. F., Wei, H. X., Fang, X. W., Xie, X. S., et al. (2016). Antiviral anthraquinones and azaphilones produced by an endophytic fungus Nigrospora sp. from Aconitum carmichaeli. Fitotherapia 112, 85–89. doi: 10.1016/j.fitote.2016.05.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, W., Tao, J., Yang, X., Zhang, J., Lu, H., Wu, K., et al. (2014). Antiviral effects of two Ganoderma lucidum triterpenoids against enterovirus 71 infection. Biochem. Biophys. Communn. 449, 307–312. doi: 10.1016/j.bbrc.2014.05.019

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhao, J., Lu, J., Shen, Y., Tan, Z., Zhang, M., Chen, R., et al. (2017). Stachybotrysams A-E, prenylated isoindolinone derivatives with anti-HIV activity from the fungus Stachybotrys chartarum. Phytochem. Lett. 20, 289–294. doi: 10.1016/j.phytol.2017.04.031

CrossRef Full Text | Google Scholar

Zhou, L.-W., Cao, Y., Wu, S.-H., Vlasák, J., Li, D. W., Li, M. J., et al. (2015). Global diversity of the Ganoderma lucidum complex (Ganodermataceae, Polyporales) inferred from morphology and multilocus phylogeny. Phytochemistry 114, 7–15. doi: 10.1016/j.phytochem.2014.09.023

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, S., Gao, Y., and Chan, E. (2005). Clinical trials for medicinal mushrooms: experience with Ganoderma lucidum (W.Curt.:Fr.) Lloyd (Lingzhi Mushroom). Int. J. Med. Mushrooms 7, 111–118. doi: 10.1615/IntJMedMushr.v7.i12.110

CrossRef Full Text | Google Scholar

Zhu, W., Chiu, L., Ooi, V. E., Chan, P. K., and Ang, P. O. Jr. (2004). Antiviral property and mode of action of a sulphated polysaccharide from Sargassum patens against herpes simplex virus type 2. Int. J. Antimicrob. Agents 24, 81–85. doi: 10.1016/j.ijantimicag.2004.02.022

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhu, Y.-C., Wang, G., Yang, X.-L., Luo, D.-Q., Zhu, Q.-C., Peng, T., et al. (2010). Agrocybone, a novel bis-sesquiterpene with a spirodienone structures from basidiomycetes Agrocybe salicacola. Tetrahedron Lett. 51, 3443–3445. doi: 10.1016/j.tetlet.2010.04.128

CrossRef Full Text | Google Scholar

Keywords: antiviral agents, antiviral mechanisms, endophytes, fungal secondary metabolites, medicinal mushrooms, natural products

Citation: Linnakoski R, Reshamwala D, Veteli P, Cortina-Escribano M, Vanhanen H and Marjomäki V (2018) Antiviral Agents From Fungi: Diversity, Mechanisms and Potential Applications. Front. Microbiol. 9:2325. doi: 10.3389/fmicb.2018.02325

Received: 03 July 2018; Accepted: 11 September 2018;
Published: 02 October 2018.

Edited by:

Juan-Carlos Saiz, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Spain

Reviewed by:

Ulrike Lindequist, University of Greifswald, Germany
Taisuke Izumi, Henry M. Jackson Foundation, United States

Copyright © 2018 Linnakoski, Reshamwala, Veteli, Cortina-Escribano, Vanhanen and Marjomäki. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Riikka Linnakoski, riikka.linnakoski@luke.fi

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.