Next Article in Journal
Post-Transcriptional Gene Silencing of Glucanase Inhibitor Protein in Phytophthora cinnamomi
Previous Article in Journal
Clarithromycin Suppresses Apple Proliferation Phytoplasma in Explant Cultures
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Why Do We Need Alternative Methods for Fungal Disease Management in Plants?

1
Agriculture and Agri-Food Canada, Kentville Research and Development Centre, Kentville, NS B4N 1J5, Canada
2
Department of Plant, Food and Environmental Sciences, Faculty of Agriculture, Dalhousie University, Truro, NS B2N 4H5, Canada
3
Department of Biology, Acadia University, Wolfville, NS B4P 2R6, Canada
4
Saint-Jean-sur-Richelieu Research Development Centre, Science and Technology Branch, Agriculture and Agri-Food Canada, Saint-Jean-sur-Richelieu, QC J3B 7B5, Canada
5
United States Department of Agriculture (USDA), Agricultural Research Service, Grain Legume Genetics and Physiology Research Unit, Prosser, WA 99350, USA
*
Author to whom correspondence should be addressed.
Plants 2023, 12(22), 3822; https://doi.org/10.3390/plants12223822
Submission received: 20 September 2023 / Revised: 3 November 2023 / Accepted: 7 November 2023 / Published: 10 November 2023
(This article belongs to the Special Issue New Methods for Disease Management of Fungal Plant Pathogens)

Abstract

:
Fungal pathogens pose a major threat to food production worldwide. Traditionally, chemical fungicides have been the primary means of controlling these pathogens, but many of these fungicides have recently come under increased scrutiny due to their negative effects on the health of humans, animals, and the environment. Furthermore, the use of chemical fungicides can result in the development of resistance in populations of phytopathogenic fungi. Therefore, new environmentally friendly alternatives that provide adequate levels of disease control are needed to replace chemical fungicides—if not completely, then at least partially. A number of alternatives to conventional chemical fungicides have been developed, including plant defence elicitors (PDEs); biological control agents (fungi, bacteria, and mycoviruses), either alone or as consortia; biochemical fungicides; natural products; RNA interference (RNAi) methods; and resistance breeding. This article reviews the conventional and alternative methods available to manage fungal pathogens, discusses their strengths and weaknesses, and identifies potential areas for future research.

1. Introduction

Plant pathogens pose a significant threat to the agricultural industry and are one of the most important factors in agricultural yield losses and food insecurity across the globe. Fungal pathogens alone may account for up to 20% of worldwide yield losses [1]. Conventional breeding techniques alone cannot provide adequate protection against fungal pathogens for many crops because fungal pathogens are able to overcome introduced genetic resistance [2]. These pathogens pose a particularly serious problem in perennial crops such as apples and other tree fruits. Orchards are expected to last 20–30 years, making it unrealistic to replace vulnerable cultivars with resistant ones, especially since resistance can be overcome long before the end of an orchard’s productive life. Considering that genetic resistance is generally not sustainable and the development of resistant cultivars takes many years, disease control has relied for decades on the application of chemical fungicides [2]. Although very effective, these fungicides are notorious for their hazardous effects on human and animal health as well as for their environmental toxicity [3,4]. Concerns over the potential environmental consequences of the uncontrolled use of these active substances has led to regulations on their use based on environmental risks assessments, and these restrictions can range from reductions in the number of applications per crop season to the outright removal of specific active substances from the market [3]. In addition, plant pathogenic fungi can develop resistance to chemical fungicides, particularly single-site fungicides, which are more likely to lead to the development of resistance in fungal pathogen populations. In the past few decades, numerous disease management strategies have been developed as an alternative to traditional chemical fungicides and breeding methods, including the use of plant defence elicitors (PDEs), biological control or biochemical fungicides, and RNA interference (RNAi). We have reached a crossroads in which these alternatives to chemical fungicides will be called on to play an increasingly important role in disease management. The present article summarizes the current state of fungal disease management strategies, discusses the strengths and weaknesses of their modes of action, and draws conclusions on the future of fungal disease management.

1.1. Chemical Fungicides

Fungicides can be broadly defined as chemical substances used to control fungal diseases by inhibiting the growth of pathogenic fungi or by eradicating them completely. Fungicides can be classified as inorganic or organic based on their chemical composition. Inorganic fungicides do not contain carbon in their structure and are typically based on sulphur or metal ions. This group of fungicides has been in use since the discovery of Bordeaux mixture (copper sulphate pentahydrate and lime) by Pierre-Marie Alexis Millardet in the in the late 19th century [5]. Centuries after this discovery, copper- and sulphur-based fungicides are still used extensively in conventional and organic agriculture [6,7,8,9]. Examples of modern inorganic fungicides include copper sulphate, copper oxychloride, and copper hydroxide [10,11]. In contrast, organic fungicides contain carbon atoms in their structure [6]. These organic synthetic compounds have become more popular, although inorganic fungicides continue to be used in modern agriculture. All synthetic inorganic or organic fungicides, regardless of their composition, can be divided into two distinct classes based on their mobility in the plant: contact (protective) fungicides, which remain on the surface of the plant, and systemic (mobile/curative) fungicides, which are absorbed into the plant.
Contact fungicides typically have a wide range of action against different fungal pathogens and are effective in preventing the infection of plant tissues. They usually achieve this by killing fungal spores or by inhibiting their germination before they penetrate and colonize the host tissues [6,12]. Contact fungicides are not effective in a curative strategy and to be effective, must be applied before the pathogen infects the plant [13]. Most contact fungicides are not absorbed by the plant and remain on the plant tissue surface. However, the frequency of application must be carefully monitored, since contact fungicides can become phytotoxic in the rare cases when they are absorbed [14]. Because contact fungicides usually remain on the plant surface, protection is temporary and can be quickly lost due to rainfall or other weather conditions [15,16]. Contact fungicides can also be eliminated from the plant surface by wind or degraded by UV radiation, and therefore, their protective action does not exceed 10–12 days [6]. It is also important to bear in mind that contact fungicides are only effective on the leaf surface present at the time of application and thus are less efficacious during heavy leaf growth. Therefore, the effectiveness of contact fungicides is particularly reliant on the proper timing of application, which generally must be performed prior to the known or predicted infection periods of the targeted pathogens. Furthermore, since these fungicides are gradually removed from the plant surface, repeated applications during the growing season are necessary for sustained protection. In some circumstances, this characteristic is beneficial to growers, since contact fungicides, unlike systemic fungicides, have the advantage of being easily removed from treated produce before it reaches the consumer [6].
Systemic fungicides are a more recent development in disease control and are considered to be more promising than contact fungicides [17]. While providing a protective effect by suppressing spore germination, these fungicides can also be absorbed into plant tissues, either locally or more broadly, and are therefore able to kill fungal pathogens after they have penetrated and infected host tissues [18]. The degree of systemic activity—which ranges from simple translaminar activity in leaf tissues to local spread from the absorption site and mobility within the xylem of the plant—is generally determined by the chemistry of the compound and can play an important role in determining the efficacy of a fungicide against specific fungal pathogens [18,19,20]. Because of the ability of systemic fungicides to be absorbed in plant tissues, using them to treat plant materials has become routine practice, and the seeds of most agricultural crops are treated with systemic fungicides to protect against both seed- and soil-borne pathogens [21]. Although systemic fungicides are highly effective, most of the compounds involved operate through a single mode of action (i.e., they generally target a single essential fungal enzyme or metabolic pathway) and, therefore, are extremely vulnerable to the development of resistance by target fungal pathogens [22].

1.2. The Disadvantages of Chemical Fungicides: Environmental Toxicity and Resistance Development

Despite their high efficacy, both contact and systemic fungicides have numerous drawbacks associated with their use. One substantial shortcoming is that, due to their lack of specificity, chemical fungicides can disrupt both beneficial and pathogenic microorganisms. For instance, the application of fungicides to mango leaves has been demonstrated to eliminate many endophytes, creating a window of opportunity for pathogens to colonize the tissues formerly that the endophytes formerly inhabited [23]. While this phenomenon has been most readily observed in foliar spray treatments, fungicidal seed treatments have also been associated with similar reductions in beneficial endophytes, and the negative consequences of fungicides on soil microbial communities are well documented [24,25,26,27,28]. Indeed, mounting evidence suggests that seeds protected with systemic fungicides may negatively impact plant health and vigour by targeting beneficial endophytes in the absence of pathogen pressure, the practice may be counterproductive [21]. Therefore, the application of chemical fungicides can have negative consequences for plant health and yield by eliminating beneficial microbes that promote growth, development, and resistance to biotic and abiotic stresses.
Fungicides can severely impact the aquatic environment, as they are able to enter aquatic ecosystems through different ways, including wastewater, runoff, and subsurface drainage [29,30], and can be toxic to a wide range of aquatic organisms, including algae, fish, and invertebrates [31,32]. Furthermore, fungicides could harm important pollinators like bees through mechanisms such as the impairment of larval and physiological development, the promotion of increased sensitivity to other pesticides, and increased mortality [33]. Exposure to fungicides can lead to acute and chronic neurotoxicity in humans, and thus significantly impact human health [34].
In addition to fungicides’ environmental toxicity, concerns have been raised over the durability of fungicide efficacy. Fungi have tremendous evolutionary potential to rapidly develop resistance against fungicides due to the intense selective pressure exerted by repeated fungicide applications [22]. Mutations in DNA sequences can arise from errors in DNA replication, damage from UV radiation, or exposure to mutagens or viral infections. Environmental stress may play a significant role in determining the rate of mutation. For example, stress from increasing temperatures accelerated the rate observed in the fungal pathogen Zymoseptoria tritici [35]. Although mutations are inherently random, those that result in enhanced resistance to fungicides will be positively selected for by the eradication of strains without resistance. Over time, this inevitably results in fungicide-resistant strains of the targeted pathogen [22].
Fungicide resistance is a stable and heritable change in an individual fungus that results in a reduction in its susceptibility to fungicides. Fungicide resistance is well documented to develop more often against single-site fungicides than those with a multi-site mode of action, making modern synthetic fungicides especially vulnerable to resistance development [22]. The threat of fungicide resistance is a major concern to growers worldwide, and numerous strategies are employed in order to prevent its emergence. The Fungicide Resistance Action Committee broadly divides fungicides by their mode of action in order to identify those with potential resistance concerns. Its recommendations include applying multiple fungicides with varying modes of action over the course of the growing season, restricting the use of the fungicides most likely to induce resistance with repeated use, and prioritizing multi-site fungicides, to which fungi are less likely to develop resistance [36]. To date, over 43 different modes of action have been identified, although the mechanisms for some of these are not yet known [36].
In conclusion, while chemical fungicides are an extremely effective tool—at least in the short term—for reducing disease incidence in the crops, they have harmful effects on beneficial plant microbiota, the health of humans and other animals, and on the environment. These factors, in addition to the rising threat of fungicide resistance, have led to increasing restrictions on the use of chemical fungicides. Given these challenges, conventional chemical fungicides must be complemented with cost-effective, eco-friendly alternatives to maintain appropriate levels of disease control with the absence or reduced usage of these vital compounds. An overview of the benefits and drawbacks of chemical fungicides and alternative disease control methods can be found in Figure 1.

2. Alternative Management of Fungal Diseases

2.1. Agronomic Practices and Cultivation Methods

Agronomic practices and cultivation methods can greatly influence the vulnerability of crops to fungal pathogens through a variety of mechanisms, including disruption of the pathogen lifecycle, improving the vegetative performance and thus the natural health status of the plant host, or the removal of sources of inoculum from the field. The main drawback of these agronomic practices is that they tend to be laborious, expensive, and less effective than conventional fungicides, and thus, they are more suited as complementary tools to more effective methods. These practices include but are not limited to: sanitation, tillage, crop rotation, pruning and thinning, the intermixing of different crops or different varieties of the same crop, and the manipulation of canopy architecture. The impacts of cultivation methods on pathogen pressures have been the subject of many thorough reviews [37,38,39], and thus, these practices will only be briefly described in this review.
Foremost among cultivation methods which can enhance the control of fungal pathogens is proper sanitation, that is, the removal of sources of inoculum from the field. For instance, the removal of crop residues, which often serve as the source of primary inoculum for Colletotrichum species, is an effective means for reducing anthracnose and black spot in guava fruit [40]. Leaf shredding is a common means of removing inoculum sources of foliar pathogens and has long been promoted as a means to control apple scab (Venturia inaequalis) [41]. However, as with many cultivation practices, sanitation practices on their own do not provide disease control comparable to chemical fungicides. In a direct comparison, the removal of senescent and necrotic leaves and the removal of unmarketable fruit from the alleys between beds of strawberries significantly reduced Botrytis fruit rot caused by Botrytis cinerea in annual strawberry compared to controls, but losses remained significantly higher than in the fungicide control [42]. Nevertheless, sanitation practices are an effective means of complementing chemical fungicides. Recently, it has been demonstrated that leaf shredding in combination with the application of demethylation inhibitor fungicides significantly influenced the fungicide sensitivity of V. inaequalis populations. V. inaequalis isolates from orchards treated with demethylation inhibitor fungicides which also underwent leaf shredding retained fungicide sensitivities close to that of unexposed populations, potentially as a result of a smaller initial effective population size [43]. Thus, the inclusion of proper sanitation practices with chemical fungicides in an integrated treatment regime may both improve disease control and delay the development of fungal resistance, though further research will be required to determine the extent of this effect.
There is conflicting evidence on the impacts of tillage on disease control, and whether specific tillage practices promote or reduce fungal diseases is reliant on the specific pathogen and macro-environmental conditions [44,45,46]. The removal of organic matter in conventional tillage practices, while consequential for soil health, also removes potential sources of primary inoculum, while conservation (or reduced) tillage can promote pathogen survival by providing residues for the pathogen between crop plantings [47]. Furthermore, conservation tillage can alter soil characteristics in ways which may either be beneficial or detrimental to individual pathogens, such as increasing soil moisture, altering soil temperatures, and failing to disrupt the soil [48]. However, compared to conventional tillage, conservation or no-tillage has been shown to improve the general disease suppressiveness of the soil by reducing tillage-induced losses in microbial biomass, and disease suppression in spring barley was improved by long-term conventional tillage and no-tillage [46]. Given the significance of the soil microbiome in the determination of the prevalence of disease, particularly in the case of soil-borne pathogens, the impact of these practices on soil and rhizosphere microbiomes is an area of intense interest [49,50].
The continuous cultivation of a particular crop results in the accumulation of pathogens and increased disease pressure [51]. As such, the rotation of crops from host to non-host species serves as an effective break to reduce pathogen inoculum [52]. Crop rotation is especially important for the control of soil-borne pathogens, and longer duration periods have been demonstrated to be more effective in preventing disease [53]. Furthermore, crop rotation may have beneficial effects on the general disease suppressiveness of the soil. Numerous studies have investigated crop rotation either alone or in combination with tillage for its potential in modulating general disease suppressiveness in the soil in crop systems such as potato, banana, and peanut [53,54,55]. Thus, ideal rotation strategies will not only serve as a break in pathogen pressure but as a means of enriching the soil’s capacity to ward off pathogens. Similar to crop rotations, the inter-cropping of different species can serve to reduce pathogen pressures [56]. In this respect, the examination of soil microbiomes as a result of multi-cropping in five organic vegetable farming systems revealed that inter-cropping was associated with a decrease in the abundance of soil-borne pathogens [57]. Thus, determining the ideal combinations for inter-cropping for each species may serve as an effective form of disease control.
Plant canopy architecture is another important factor in determining disease pressure, as the density of the canopy can significantly influence the micro-climate of the canopy as well as spore dispersal. Thus, for fungal pathogens that are dispersed over short distances by splashing rain, canopy architecture is a major factor in determining the dispersal of the pathogen [58,59,60]. On the other hand, dense canopies are also associated with a microclimate of increased humidity which favours pathogen infection, and thinning systems which promote aeration within the canopy have been shown to reduce the incidence of apple scab infection [61,62]. Thus, the determination of the optimal density for a given crop represents a significant challenge in the pursuit of enhanced disease control. Intriguingly, the uniformity of crop height in current cropping systems may be advantageous to splash dispersed fungal pathogens, as it has recently been demonstrated that growing wheat cultivars of contrasting height together enhanced the control of Septoria triciti blotch (Zymoseptoria triciti) [63]. While more research is necessary to determine whether this approach is applicable to other crop systems or pathogens, it is clear that further research into the interplay between canopy architecture and disease will be beneficial for disease management.
Whether it be through the direct removal of primary inoculum sources, the rotation of non-host crops, or the establishment of physical barriers to pathogen dispersal, cultivation methods have significant influence on the vulnerability of crops to fungal pathogens. Understanding how common cultivation practices interact with disease management will allow for the optimization of growth conditions in order to reduce disease pressure, especially as these cultivation practices can readily be combined with chemical fungicides or their alternatives for improved disease control.

2.2. Improving Plants’ Genetic Resistance through the Use of R and S Genes

Plants have evolved numerous genetic defence mechanisms to protect themselves from pathogens. Growers have long relied on the manipulation of these mechanisms, traditionally by breeding for resistance as a way to reduce crops’ susceptibility to fungal pathogens. Host plants can recognize non-specialized fungal pathogens by toll-like receptors that detect pathogen-associated molecular patterns, in turn activating the host’s downstream defense mechanisms. Specialized pathogens are able to overcome these basal defence mechanisms by secreting effector molecules into host plants [64]. However, the co-evolution of plants and fungal pathogens over millennia has provided plants with a means of defence against effector molecules: resistance (R) genes. The R-gene family is incredibly diverse and well conserved in plant species. R-genes encode for nucleotide-binding leucine-rich receptors (NLRs), which collectively recognize a broad spectrum of plant pathogens and pests, inducing an array of resistance mechanisms in response to infection or predation [65,66]. NLRs are activated by the binding and recognition of pathogen effectors but, in some cases, may detect a pathogen indirectly, generally by recognizing pathogen modified host proteins. An example of this form of recognition occurs in Arabidopsis in response to Pseudomonas syringae infection, where the effector (in this case, a protease) cleaves the Arabidopsis PBS1 kinase, triggering its recognition by the NLR RPS5 [67]. The successful recognition of effector molecules or effector-modified host proteins in plants by the associated NLR typically results in effector-triggered immunity, a localized response characterized by a hypersensitive reaction (HR) in which the plant cells near the infection undergo apoptosis [68,69]. In addition, the recognition of a pathogen by plant pattern-recognition receptors (PAMPs) or an effector protein by R proteins triggers the production of salicylic acid (SA) and the downstream induction of broad, systemic defence mechanisms against subsequent infections, triggered independently of the HR response [70,71].
The direct or indirect recognition of effector proteins by R-gene-encoded receptors involves a gene-for-gene relationship in which the R-gene receptor identifies a single effector protein (encoded by a matching avirulence [Avr] gene); therefore, a host with a given R-gene will be resistant to a pathogen with the matching Avr gene [72]. In most plant–pathogen systems, the host and pathogen species may collectively have numerous R- or Avr genes. For example, twenty R-genes have been identified in apples (Malus × domestica) that match the corresponding Avr genes identified in the pathogen Venturia inaequalis, which causes apple scab. However, it should be noted that no single cultivar or individual line will contain all these resistance genes; for example, Honeycrisp apples have Rvi19 and Rvi20 in their genomes, while Golden Delicious cultivars contain Rvi1. Therefore, while many apple cultivars have some resistance to Venturia inaequalis, these cultivars are still vulnerable to some Venturia inaequalis strains that do not have corresponding Avr genes [73].
A typical mechanism in pathogens for overcoming host resistance is Avr gene mutations to prevent the product (or activity, in the case of indirect mechanisms) from being recognized by R-gene-encoded receptors. If the effector is recognized, pathogens can also overcome resistance by interfering with the host response [74]. The presence of an R-gene in a host plant population will naturally select for pathogens in which the corresponding Avr gene has been lost or modified so that it is no longer recognized by the R-gene-encoded receptor. In turn, successful mutations in the Avr gene will induce selection pressure on host plants for R-genes which impart resistance to the mutated effector. Thus, host plants and their pathogens are continuously engaged in an evolutionary arms race and, in wild populations, the frequencies of Avr and R-genes will cycle over time [75,76]. In modern agricultural settings, the uniformity of resistance genes in a population may accelerate the selection process, leading to rapid loss of resistance in these settings [77].
The identification of R-genes and their incorporation in economically important crops is a vital pillar in the development of resistant plants. Along with the use of conventional fungicides, resistance breeding techniques have served as the most effective method of disease control for decades, particularly in annual crops [78]. Although resistance breeding is also practiced in perennial crops, its effectiveness is often limited by the lifetime of the crop. Modern tree fruit crops, for instance, are expected to have lifespans of 20–30 years, giving ample time for selective pressure from resistant cultivars to result in pathogens overcoming the associated R-genes [77]. This is particularly problematic since introducing cultivars with new resistance genes is difficult due to orchards’ long lifespans. Furthermore, plant breeding is very time-consuming and, in recent years, plant breeders have relied on transgenic tools or gene transformation to expedite resistance-breeding efforts, since they allow the faster introduction of R-genes from otherwise incompatible species as well as from compatible species [79].
Numerous strategies have been developed to reduce the ability of fungal pathogens to overcome R-genes, such as rotating R-genes in a field (most suitable for annual crops), mixing cultivars with distinct R-genes in a field or between fields, and pyramiding multiple R-genes in a single cultivar to confer more durable resistance [77]. Somewhat like multi-site fungicides, pyramiding R-genes in a single cultivar makes it more difficult for pathogens to overcome resistance despite their evolutionary potential [80]. However, pathogens are still capable of overcoming multiple R-genes in the same host plant. For example, the oomycete pathogen Phytophthora infestans can escape multiple resistance genes in potato [81]. The breakdown of resistance to rust fungi in cereal crops under different strategies was recently modelled, and it was demonstrated that, although pyramiding could provide the most effective pathogen resistance, this resistance is less durable when mutation rates in the pathogen population are high [77]. Under such conditions, mixing or rotating crops was more successful at delaying the breakdown of resistance to different R-genes. For example, in mixed populations, the breakdown of resistance to one major R-gene was correlated with increased durability of the other R-genes in the population. Rotations were particularly successful since they were modelled so that pathogens were consistently challenged with new R-genes. Consequently, the authors concluded that rotating different pyramids of R-genes was the most promising method of ensuring durable R-gene resistance [77]. However, many resistant varieties may incur yield and/or crop quality penalties when compared to their susceptible counterparts, and these costs must be carefully considered with the associated benefits [82]. Therefore, the rotation of resistant varieties may not be a feasible strategy in many cases due to economic implications. Furthermore, this strategy is unlikely to be useable or effective in perennial crops, such as apples, pears, and cherries, which have longer lifespans and a juvenile period.
Beyond the introduction of R-genes in susceptible genotypes, advances in genome editing have allowed researchers to identify other mechanisms for reducing disease severity or improving resistance, such as targeting susceptibility (S) genes [83]. S-genes are genes in the host plant required for pathogen infection. Interaction of a pathogen’s effector/toxin molecules with S-genes can assist the pathogen in a variety of ways, such as the recognition and penetration of the host, sustained compatibility between the pathogen and host, and the inhibition of immune signalling [84]. Therefore, the genetic silencing or knocking out of S-genes can improve the host plant’s resistance to the pathogen and is one of the newest frontiers in conferring durable pathogen resistance [83]. Recently, CRISPR/Cas9-mediated knockouts of three S-genes in potato, StDND1, StCHL1 and StDMR6, increased resistance to potato late blight caused by Phytophthora infestans [85]. Likewise, in apple, the expression of the MdCNGC2 gene, which encodes a cyclic nucleotide-gated ion channel, was observed to be strongly induced by Botryosphaeria dothidea infection in susceptible cultivars [86]. Improved resistance to the pathogen was observed with both virus-induced gene silencing and CRISPR/Cas9-mediated mutagenesis of MdCNGC2 [86]. To date, targeting S-genes has proven to be a successful strategy for inducing disease resistance in a number of crop systems, including cucumbers, rice and tomato [87].
Directly introducing resistance in crops is an effective disease management strategy. However, while both R-genes and S-genes can be modified or integrated in the host genome to improve disease resistance, the process is costly, laborious, and time-consuming. In addition, the rapid breakdown of resistance in the field makes resistant cultivars less effective in long-lived crops. Therefore, complementary tools are needed to help delay the breakdown of resistance in crops that cannot be rotated annually.

3. The Use of Plant Defence Elicitors

While pathogen-triggered immunity coordinates the host’s defence against specific pathogens, systemic acquired resistance (SAR) or induced systemic resistance (ISR) mediates prolonged, broad-range resistance to plant pathogens [88,89]. The induction of SAR is characterized by a local increase in levels of the phytohormone SA, which in turn results in the accumulation of reactive oxygen species (ROS) and promotes the expression of β1,3-glucanase, chitinase, and classical pathogenesis-related (PR) genes (Pr1, Pr2, and Pr5). The increased expression of these genes promotes plant defence through various downstream targets [90]. Pathogen infection is associated with a rapid increase in SA levels in apple leaf tissue, and this activity is necessary to induce SAR [91,92]. ISR, however, does not require SA accumulation and instead is associated with the accumulation of jasmonic acid (JA) and ethylene (ET). JA and ET accumulation leads to the activation of downstream targets, including chitinase, β1,3-glucanase, and an alternative set of PR genes (PDF1.2, PR3, and PR4), as well as the accumulation of ROS (like in SAR) [93]. Significant overlap and crosstalk occur between the SAR and ISR signalling pathways, and both are associated with protection against different pathogens. For example, SAR provides greater protection against biotrophic or hemi-biotrophic pathogens but may leave the plant more vulnerable to necrotrophic pathogens. Conversely, ISR provides protection against necrotrophic pathogens and chewing insects at the expense of protection against biotrophs and hemi-biotrophs [94,95]. This is a direct result of the crosstalk between these pathways, since the upregulation of SA is typically associated with a decrease in JA and vice versa, although synergism occurs between these two defence mechanisms in some cases [95].
Inducing plant defence by applying exogenous elicitors is a promising alternative to conventional fungicide applications [96]. The application of exogenous PDEs has been associated with enhanced resistance against a wide range of pathogens in different crops, including cereals, tomato, rubber tree, and apple [97,98,99,100]. Pre-treatment with SA of the leaves of the susceptible apple cultivar Gala was associated with a marked (albeit temporary) increase in the expression of the plant pathogenesis-related genes PR1, PR5 and PR8, as well as of chitinase and β1,3-glucanase, with the treated leaves demonstrating increased resistance to Glomerella leaf spot following in vitro inoculation with Glomerella cingulate, indicating the induction of SAR [100]. Similarly, exogenous applications of phenylacetic acid have been observed to induce ISR in tobacco, imparting significant resistance to the bacterial soft rot pathogen Pectobacterium carotovum [101]. Elicitor treatment may protect fruit from plant pathogens for a significantly longer period than the temporary uptick in defence-gene-related expression suggests. In this regard, field treatments of Ya Li pear with a 2.5 mM SA spray produced a remarkable decline in disease incidence and lesion diameter compared to the untreated control. Finally, activities of defence-related enzymes such as peroxidase, phenylalanine ammonia-lyase, chitinase, and β-1,3 glucanase not only increased significantly in the four days following SA application, but these increased activities were still observed after harvest [102].
Since the discovery of SA as an exogenous inducer of SAR, several synthetic PDEs that are analogues of the phytohormones SA and JA have been developed. The ones most commonly used in the past few decades are benzo(1,2,3)thiadiazole-7-carbothioic acid S-methyl ester (BTH) and 2,6-dichloro-isonicotonic acid (INA), which are preferred for their increased efficiency and reduced phytotoxicity compared to the original SA [90].
Phytohormones and their synthetic analogues are not the only methods for inducing plant defence mechanisms. Peptides, polysaccharides, and lipids isolated from plants, fungi, and bacteria can also serve as PDEs. For example, chitosan, which can be readily obtained from fungal cell walls, is well known for its ability to induce host defenses, although this compound also demonstrates direct fungicidal activity [103]. The application of ZhiNenCong (ZNC), an extract of Paecilomyces variotii, stimulated immunity in Arabidopsis thaliana to bacterial infection and in potato to Phytophthora infestans infection via SA-dependent signalling pathways. Cell wall extract from the mushroom Pleurotus ostreatus enhanced defence against Septoria and mildew in wheat and grapevine, respectively [104,105]. Similarly, extracts of giant knotweed (Reynoutria sachalinensis) improved the resistance of courgette to Podosphaera xanthii in an SA-dependent manner [106]. Extracts from the seaweeds Ascophyllum nodosum, Cystoseira myriophylloides, Laminaria digitata, and Fucus spiralis represent a significant portion of plant-based elicitors [107,108].
The application of exogenous plant defence elicitors that can stimulate SAR or ISR responses before pathogen infection is widely considered to be an eco-friendlier alternative for disease control in plants, and therefore, the identification and characterization of plant defense elicitors have been major areas of study (Table 1) [109]. However, PDEs have been shown to be significantly less effective than conventional chemical fungicides in many instances [102], which could be partially attributed to the inherent crosstalk between the SAR and ISR signalling pathways, since the induction of one pathway is often associated with the inhibition of the other. Therefore, the activation of SAR may improve a host’s defence against biotrophic and hemi-biotrophic fungal pathogens but leave the host more vulnerable to necrotrophs [95]. Furthermore, the induction of plant defences requires the significant allocation of the host plant’s resources and, thus, may be associated with a decrease in overall plant fitness [110,111]. For this reason, many PDEs are very rarely used, and therefore do not fully replace conventional chemical fungicides. Conversely, the use of elicitor products improved retention and yield, particularly in the case of seaweed extracts, which may also act as biostimulants [112,113]. These products can improve disease protection when applied in conjunction with fungicides. For example, the use of Actigard® (an SA analogue) increased the efficacy of dimethomorph, mancozeb, and azoxystrobin in preventing tobacco blue mould caused by Peronospora tabacina [114]. Consequently, despite their limitations, PDEs remain an attractive disease control tool, with their usefulness determined by the context of their use. Although PDEs are inefficient when used on their own, when incorporated in integrated disease management programs, they could potentially reduce the use of chemical fungicides, thus lessening the environmental exposure to these pesticides and potentially slowing the development of fungicide resistance in pathogens. The increased use of PDEs combined with biological control agents or biochemical fungicides could provide an economically acceptable level of control.

4. Biological Control and Biochemical Fungicides

Plant tissue is colonized by a wide range of microbes, which may be endophytic (colonizing the inner surface of the plant) or epiphytic (residing on the outer surface of the plant). In many cases endophytic and epiphytic microbes do not cause disease symptoms, However, their interaction with the host plant is not strictly mutualistic and under particular environmental conditions previously mutualistic fungi may become pathogenic [137,138,139]. Nevertheless, many plant-associated microbes play vital roles in promoting plant health, including influencing their hosts’ disease resistance [140]. These resident microbes can antagonize plant pathogens, and thus are a focus of current research on plant disease management [141]. A common mechanism for this antagonism is direct competition for resources, which reduces the availability of both physical space and nutrients in host tissues and, in turn, the opportunity for pathogen infection [142,143]. Plant-associated microbiota can also significantly impact host resistance to pathogen infection through colonization, which triggers the host’s localized defence mechanisms through ISR, resulting in a more rapid induction of the defence response when a plant pathogen subsequently colonizes the host [144]. Furthermore, plant-associated microbes can target and antagonize plant pathogens either directly, by secreting antifungal or antibacterial secondary metabolites that reduce pathogens’ growth, or indirectly, by secreting metabolites that enhance the host’s production of antifungal or antibacterial metabolites [145,146,147]. Some endophytes, such as members of the genus Trichoderma, can antagonize plant pathogens directly through mycoparasitism [148,149].
Biocontrol agents are generally plant-associated microbes that have been screened and selected for use in crop systems in order to improve plant fitness, induce plant defence mechanisms, and antagonize pathogens [129,150,151]. Microbes can be beneficial to plant health and disease control through a multitude of mechanisms, and many different biocontrol agents have been developed to protect against pathogens, particularly at the post-harvest stage. In particular, a key characteristic of effective biocontrol agents is to facilitate the exclusion of a pathogen from its ecological niche through effective competition [152]. The current literature on biocontrol focuses heavily on a small number of beneficial genera. For example, species of the soil-borne genus Trichoderma are extremely effective in controlling soil-borne and foliar pathogens and have been demonstrated to perform well against a number of pathogenic species, including Rhizoctonia solani, Fusarium oxysporum, and Bipolaris sorokinia. The antifungal properties of these soil-borne species typically arise from a combination of direct competition, mycoparasitism, and the secretion of antifungal compounds [129,149,153,154,155,156]. Members of the genus Aureobasidium, most notably Aureobasidium pullulans, show promise in controlling post-harvest pathogens, such as Botrytis cinerea, Penicillum expansum, and Diplodia seriata [157,158,159,160,161,162]. Like Trichoderma, A. pullulans antagonizes pathogens through direct competition for space and resources as well as by the secretion of antimicrobial compounds [163]. Additionally, bacterial genera such as Pseudomonas, Bacillus, and Agrobacterium have demonstrated an excellent ability to suppress economically significant plant pathogens [164,165,166].
Extensive laboratory research has resulted in commercial biocontrol agents being available in varying degrees around the world. Trichoderma spp. is the best established biocontrol agent, and several products containing various Trichoderma species have been commercialized since the 1970s [167]. Products incorporating fungi such as Aspergillus flavus and A. pullulans, yeasts such as Candida oleophila, and bacteria such as Pseudomonas, Bacillus, and Agrobacterium, have been approved in Europe and the United States [167]. Biocontrol agents are often considered to be less vulnerable to resistance development in pathogen populations, because, unlike synthetic fungicides, they often have multiple modes of action, although this is not always the case, and more research is needed to determine their modes of action [167,168]. Indeed, very few studies have been conducted on the probability of resistance development in fungal pathogens to biological control agents [168]. Despite the potential advantages of biocontrol products, they are not universally preferred to chemical fungicides because their efficacy can vary significantly. Conditions in the field are variable and difficult to predict, and microbial biocontrol agents can behave unreliably in uncontrolled conditions [169].
Mycoviruses also demonstrate remarkable promise as candidates for biocontrol, since mycoviral infections in plant pathogens are associated with a reduction in virulence as a result of the RNA silencing (RNAi) of pathogen and host genes [170]. A major advantage of this form of biocontrol is its ability to spread within a pathogen population following introduction. This spread occurs primarily through fungal hyphal anastomosis (hyphal fusion), a key process required in these pathogens for homeostasis and genetic exchange [171,172]. Upon a mycovirus’ successful infection of a pathogen, the mycovirus will be passed down to the pathogen’s progeny, thereby reducing pathogenicity across multiple generations [173]. To date, numerous mycoviruses capable of inducing hypovirulence in pathogen populations have been identified, including AsHV1, which reduces the virulence and growth of Alternaria alternata, and BcPV2, which induces hypovirulence and an absence of conidia in Botrytis cinerea [174,175].
In addition to mycoviruses, bacteriophages (viruses that target bacteria) have also been used in the biocontrol of bacterial plant pathogens. Bacteriophages have several advantages, because they are usually genus-specific and able to replicate within their hosts, but do not accumulate in the environment in their hosts’ absence [176]. In some cases, phages can degrade extracellular polysaccharides involved in virulence, as in the case of Erwinia amylovora [177]. Bacteriophage-based biocontrol products typically use a combination of different phages to increase the product’s modes of action as well as improve the range of target pathogen genotypes, and to reduce the chance of acquired pathogen resistance [178].
In contrast to biocontrol agents, biochemical fungicides are naturally occurring compounds that can be used to control fungal diseases. Biochemical fungicides can increase a plant’s disease resistance by inducing its defence responses or by directly inhibiting the pathogen. Biochemical fungicides may include compounds screened and selected based on their antimicrobial activity, many derived from biocontrol agents [179] or, alternatively, take the form of whole-cell extracts. Biochemical fungicides have been proven effective against many fungal pathogens. For example, the cell-free extracts of Pseudomonas have been shown to inhibit the mycelial growth of Alternaria alternata and Fusarium solani in vitro, while SH2, an antifungal compound derived from Streptomyces hydrogenas, was found to control Alternaria brassicicola on radish seeds in vivo [180,181]. Plant extracts are another important source of biochemical fungicides and can inhibit diverse phytopathogenic fungi such as Botrytis cinerea, Fusarium oxysporum, and Leptosphaeria sacchari (syn. Phoma sorghina) [182,183]. Similarly, natural oils can have potent antifungal activity, reducing the severity of diseases caused by numerous pathogens, such as Fusarium oxysporum, Fusarium solani, Fusarium monliforme, Thilaviopsis paradoxa, Botryodiplodia theobromae, and Rhizoctonia solani in date palm seedlings; Phytophthora parasitica var. nicotianae in tobacco; and Venturia inaequalis in apple [184,185,186].
Currently, both biocontrol agents and biochemical fungicides are viewed as eco-friendly means of disease control. These methods are typically less phytotoxic than synthetic fungicides and are considered to be a more environmentally friendly method for pathogen inhibition or eradication as they do not leave toxic residues [179,187,188]. As such the identification of biocontrol agents and biochemical fungicides is a significant area of research (Table 2). The continuation of this work will be of vital importance in the future development of disease management strategies.

5. RNAi-Based Disease Management

Small RNA-Based Fungicides

RNA silencing (or RNAi) is a highly conserved mechanism in eukaryotes that allows gene silencing at the transcriptional and post-transcriptional levels by small RNAs (sRNAs). At the transcriptional level, small, double-stranded RNA (dsRNA) is cleaved by the ribonuclease Dicer, and loaded into the RNA-induced initiation of transcriptional gene silencing complex (RITSC), which, utilizing the sRNA as a guide, binds to homologous DNA, leading to the methylation of the heterochromatin in this region and, consequently, gene silencing [224]. A similar mechanism is responsible for post-transcriptional gene regulation, in which small dsRNA, after being cleaved by Dicer, is loaded into the RNA induced-silencing complex (RISC) and guides the latter to complementary RNA targets through base pairing [225]. RISC then “slices” the homologous mRNA directly, leading to the degradation of the mRNA transcript [225,226]. The recognition of the mRNA sequence can also lead to translation repression [225,226,227], since RNAi does not require perfect complementarity. The degree of complementarity between the small RNA guide and its target is a major factor in determining the interference mechanism, with lower degrees of complementarity associated with translational repression and higher ones, with mRNA degradation, although translational repression may still occur in plants under conditions of high complementarity [228,229,230,231]. These mechanisms are originally thought to have evolved primarily as a means of protecting the host from viral infections and transposable elements; however, a body of evidence suggests that hosts and pathogens are engaged in an evolutionary arms race, with a rapid co-evolution of antiviral RNAi genes in hosts and viral RNAi suppressor genes in viruses [232].
In the last decade, RNAi has generated immense scientific interest as a way to achieve effective and eco-friendly disease control in agricultural settings (Table 3). This research can broadly be divided into two separate categories: that focusing on host-induced gene silencing (HIGS), which involves the genetic modification of a host plant to express dsRNA that targets a specific pathogen’s pathogenicity related genes, and that focusing on spray-induced gene silencing (SIGS), which involves the exogenous application of similar dsRNAs or sRNA [233,234].
i.
Host-Induced Gene Silencing
RNAi can be triggered by the presence of dsRNA, hairpin or transgenic foreign RNA, or viral dsRNA, culminating in the use of the dsRNA as a template sequence to locate and silence matching foreign dsRNA [233]. Taking advantage of this inherent disease control mechanism, researchers have developed transgenic crops that express dsRNA targeting known pathogens and pests [265]. HIGS offers significant advantages over conventional resistance breeding techniques. For instance, HIGS does not require a pool of established resistance genes. Its only limitation is that the researcher must design the dsRNA to target the relevant pathogenicity genes. The use of HIGS to target the parasitism genes of root-knot nematodes is just one example of the degree of improvement provided by HIGS in fine-tuning disease resistance. For years, efforts to combat root-knot nematode parasitism in many crops were hindered largely by the lack of effective, broad-range resistance genes [266]. However, this problem was overcome by creating transgenic Arabidopsis plants that express dsRNA for the parasitism gene 16D10, which confers resistance against a wide range of root-knot nematode species [267]. Thus, HIGS was able to provide a degree of resistance unattainable by conventional methods, and, in the future, other varieties of vulnerable crops will likely be transformed with 16D10 dsRNA [267].
HIGS techniques show significant promise in the control of fungal pathogens. After confirmation was obtained that dsRNA targeting fungal transcripts of Blumeria graminis (the causal agent of powdery mildew) drastically impedes the development of this disease in wheat and barley, transgenic plants were developed that express dsRNA to silence pathogenicity genes. Significant disruption of host–pathogen interactions was demonstrated in these transgenic plants, demonstrating HIG’s potential in controlling fungal pathogens [238]. Since then, HIGS techniques have been applied successfully against the fungal pathogens of a wide variety of crops, including Verticillium in tomato and Magnaporthe oryzae in rice [244,261].
Despite HIGS’ advantages over conventional breeding methods, this emerging technology has many of the same limitations as conventional techniques, namely that introducing new dsRNA in economically important crops and cultivars is a time-consuming process. In addition, significant opposition to genetic modifications may be present in some jurisdictions. Furthermore, although it may be more difficult for fungi to develop resistance to RNAi, there is significant evidence supporting an evolutionary arms race between fungal pathogens and host plants in regard to RNAi [232]. Therefore, there might be a need to “pyramid” dsRNA to target several relevant (and unrelated) pathogenicity genes [80]. Even with the “pyramiding” of dsRNA, the introduction of resistant cultivars remains a suboptimal option for perennial crops with lengthy life cycles. In these crops, HIGS will likely need to be complemented with conventional spray programs or the new SIGS techniques.
ii.
Using Spray-Induced Gene Silencing as the Basis for RNA-based Fungicides
SIGS techniques involve the spray application of exogenous dsRNA or sRNA to plants. The efficacy of this method was first demonstrated in barley, where the application of exogenous dsRNA targeting three ergosterol biosynthesis genes critical to the integrity of fungal cell membranes resulted in the effective inhibition of the fungal pathogen Fusarium graminearum [234]. Since then, SIGS has proved effective against a number of fungal pathogens, including Botrytis cinerea and Sclerotinia scletiorum, in a variety of crop systems, including strawberry, tomato, and Arabidopsis [250,256].
While dsRNA was originally thought to be processed directly by the fungal RNAi machinery, recent evidence suggests that SIGS requires the uptake of sprayed dsRNA by the host plant’s stomata [268]. In their study, Biekenkopf et al. demonstrated the processing of dsRNA by host plants and visualized fluorescently labelled dsRNA travelling through the vascular system of plants, indicating its long-distance spreading to distal areas [268]. Significant (~60%) gene silencing of the targeted Shp gene was reported even in aphids that fed on unsprayed distal tissues, confirming the travel of dsRNA through the vascular system to tissues [268]. Indeed, the fungal pathogen Fusarium graminearum was also impacted by the dsRNA, even in the plant roots [268]. Previously, it had been demonstrated that the use of larger dsRNAs resulted in the decreased inhibition of target mRNA when dsRNAs were applied exogenously, suggesting that the lower efficacy of larger dsRNA in SIGS must be a result of poor uptake by fungi (while this has no relation to the efficacy of HIGS) [251]. However, since more recent evidence suggests that dsRNA uptake and processing by the host plant plays a significant role in the SIGS of target pathogens, it seems that the size of dsRNA may instead inhibit the uptake of these molecules by the plant, a process required in SIGS [268]. Unfortunately, while these studies provide some insight into the uptake and dispersal of dsRNA in host plants, more research is needed to elucidate the exact mechanism for dsRNA uptake in plant tissues and, subsequently, by fungal pathogens.
While the use of SIGS in crop systems is advantageous, it should also come with the caveat that dsRNAs are not continuously transcribed by the crop, as is the case with HIGS. Until recently, this represented a major hurdle in the practical application of SIGS biopesticides, especially since dsRNAs are known to have a short half-life. However, the development of environmentally friendly, non-toxic, degradable clay nanosheets allows the sustained release of dsRNA for up to 30 days after application, drastically increasing the duration of protection offered by a single spray application [269]. Since both the clay nanosheets and dsRNA have been demonstrated to be environmentally non-toxic, SIGS offers significant environmental advantages over traditional chemical fungicides [269]. A recent RT-qPCR method for quantifying dsRNA in agricultural soils demonstrated that exogenous dsRNA dissipated to below-detectable levels within hours of application, providing further evidence that this spray technique may be far more eco-friendly than conventional fungicides, which are known to persist in the environment over a longer time scale [3,270]. In addition, because dsRNAs are designed to target specific pathogens, SIGS has the potential to avoid many of conventional fungicides’ off-target effects, which greatly alter the microbiome of crops [271]. In this regard, the specific design of the dsRNA is vital since RNAi has recently been noted to have off-target effects, such as targeting mRNA with as few as eleven contiguous nucleotides in common with the dsRNA-provided template, thus posing a substantial risk to both human health and the host plant [272,273]. Consequently, extensive research will be required to determine the off-target effects of specific dsRNA on human health, as well as on target crops and their microbiomes. Nevertheless, the potential advantages of SIGS over conventional fungicides are undeniable.

6. Future Directions

Despite recent advances in disease control, comparing a given method’s efficacy across different studies can be quite difficult due to differences in crops, cultivars, the timing of treatment, environmental conditions, and targeted pathogens. Although numerous eco-friendly options have been developed to control fungal diseases, many of these are challenging to use effectively in the field. For example, PDEs were initially considered a viable alternative to conventional pesticides, until subsequent field trials demonstrated that PDEs alone were usually less effective than conventional fungicides [274]. Direct comparisons of these control methods will be required in order to determine their relative efficacy under field conditions.
While biochemical fungicides have been a focus of research efforts for decades and are available in commercial formulations, new bio-degradable, environmentally safe products have significant potential and there is a strong need for their development [275]. Many synthetic fungicides, such as strobilurins, have been developed from natural antimicrobial compounds, and the continued identification, isolation, and production of these compounds could provide a pipeline of highly efficient antifungal agents [181,276]. Although the isolation and use of these antifungal compounds may be appealing, the resulting individual compounds are likely to be similar to modern synthetic fungicides in their reliance on a single target site and, therefore, like strobilurins, may be extremely vulnerable to the development of resistance in the targeted pathogens [36]. In contrast, biochemical fungicides produced from the whole-cell extracts of known mutualists, as well as plant extracts and essential oils, appear to be less vulnerable to resistance development due to their potential for multi-site activity. Essential oils and plant extracts may have up to six different modes of action, such as (i) the inhibition of cell wall formation; (ii) the inhibition of ergosterol synthesis; (iii) the inhibition of mitochondrial electron transport; (iv) interference with RNA/DNA and protein synthesis; (v) interference with efflux pumps; and (vi) the inhibition of cell division. On the other hand, substantial losses in the efficacy of plant extracts and essential oils in field treatments have been observed compared to experiments under controlled conditions, most likely due to a decrease in the stability of these products during storage or transport, or under field conditions [277]. However, in recent years, significant advances have been made in biopesticide formulation, and several non-liquid preparations have been developed for insecticidal purposes [278]. Similar advances in essential oil- and plant extract-based biochemical fungicides will improve their efficacy under field conditions and allow for the more widespread adoption of these important technologies, since the formulation and stability of these products remain the greatest obstacles to their effective use.
Although research on RNAi techniques is in its infancy, both SIGS and HIGS have significant potential as eco-friendly disease control methods. In addition to their high potency and reduced environmental impacts, RNAi techniques may also be less vulnerable to resistance development than modern single-site fungicides. While a single mutation can change the conformation of a target enzyme (for example, CYP51 in ergosterol synthesis) and prevent fungicide binding and activity, RNAi mechanisms are not nearly as vulnerable to evasion, since even the imperfect recognition of an mRNA transcript can still lead to translational repression and, thus, deprive the pathogen of a vital protein [228]. Owing to the relatively recent introduction of the HIGS and SIGS techniques, the rate at which fungal pathogens can develop resistance to dsRNA targeting of pathogenicity-related genes has yet to be assessed. Nevertheless, the dsRNA targeting of multiple unique pathogenicity genes, presenting the pathogen with a challenge that would require mutations in multiple loci to overcome, should obviously be investigated. However, the generation of hosts expressing dsRNA is a lengthy and laborious process and would need to be repeated with several dsRNAs unique to each targeted pathogen to ensure enduring broad-spectrum resistance. Considering the strong consumer sentiment against the genetic modification of crops, SIGS will most likely become the preferred method for controlling fungal pathogens through RNAi. Nevertheless, the development of resistant lines with HIGS and, in particular, combining the pyramiding of the expression of unique dsRNA with R-genes and the modification of susceptibility genes could significantly reduce growers’ costs—particularly labour costs—by reducing the need for pesticide applications.
The timing of fungicide applications is usually informed by plant growth stage and/or the predictive modelling of pathogen risk based on weather forecasting, resulting in greater efficiency and reduced fungicide use [279,280,281,282,283]. More recently, assays to detect inoculum levels in the field have been developed, which allow pathogen pressures throughout the growing season to be assessed and the most effective timing for fungicide application to be determined, resulting in a potential reduction in pesticide use and in the rate of fungicide resistance development [284,285]. The monitoring of inoculum levels will be particularly useful in guiding the application of SIGS, since, unlike many chemical fungicides, well-designed dsRNA should be highly specific to the targeted pathogen. The incorporation of environmentally friendly disease control methods in integrated spray programs, informed by predictive modelling and the monitoring of inoculum levels in the field, could significantly reduce our reliance on environmentally toxic chemical fungicides and delay the development of fungicide resistance in economically significant pathogens. For instance, Colletotrichum acutatum, a fungal pathogen causing bitter rot of apple, has been demonstrated to overwinter in trees by infecting the outer bud scales asymptomatically; the primary inoculum identified in orchards in early spring is mainly dispersed by the buds opening in the canopy [286]. Therefore, to manage Colletotrichum acutatum, early applications of fungicides during bud burst are critical to control the levels of primary inoculum and thus prevent secondary infections later in the growing season [286]. Subsequent to bud break treatments with systemic fungicides, biochemical fungicides or SIGS could be used to control pathogen population levels, reducing the need for additional systemic fungicide applications later in the growing season, delaying the development of fungicide resistance, and reducing the environmental impacts of fungicide use. Weather-based forecasting models for biocontrol and pathogen populations need to be developed to optimize the timing of applications.
Although PDEs and biocontrols are generally outperformed by other disease control methods in comparative assays, these methods are still beneficial in reducing disease under actual field conditions, particularly when used in combination with other techniques. For instance, the co-application of the PDE Actigard® with conventional fungicides significantly increased protection against blue mould of tobacco caused by the oomycete Peronospora tabacina compared to the use of fungicides alone [114]. Combining biocontrol agents with chemical and biological plant defence inducers and chemical fungicides has been suggested as a way to reduce fungicide use while still providing effective disease control [287]. In the case of biological PDEs and biocontrol agents, this approach requires compatibility with specific conventional fungicides. However, this is another major advantage of RNAi technologies because RNA constructs, which are designed specifically to target pathogenicity-related genes, are likely to be compatible with unrelated biocontrol agents, enabling their co-application. The successful co-application of biocontrol agents and SIGS would present pathogens with simultaneous multiple challenges, such as RNAi targeting key pathogenicity genes, direct competition for space and resources, the induction of host defences, and direct antagonism. Consequently, at a time when SIGS research is still in its infancy, the investigation of combined treatments with biocontrol agents or PDEs could clearly present a significant opportunity for improved disease management.

7. Conclusions

While chemical fungicides have been one of the most effective methods of disease control for decades, restrictions on their use are increasing due to their negative environmental impacts and consequences for animals and human health, as well as the growing threat of fungicide-resistant pathogens. However, the gap left by the elimination of these fungicides cannot be bridged by traditional breeding methods, because the introduction of new resistance genes in plant species is time- and labor-intensive, and pathogenic fungi are able to quickly overcome introduced resistance genes in the field. Consequently, for decades, researchers have been searching for effective and environmentally friendly methods for controlling fungal pathogens.
According to the literature, both biochemical fungicides and RNAi-based techniques are highly effective and have significant advantages over conventional methods in terms of environmental sustainability. The rapid breakdown of RNA in the environment suggests that RNAi techniques will be an eco-friendly alternative to chemical fungicides. In addition, owing to the specificity of RNAi, it can be more easily incorporated in integrated pest management systems than conventional fungicides, leading to combined treatments with biocontrol or PDEs. More research is required in order to determine the relative efficacy of these methods under field conditions and to develop integrated pest management systems that challenge fungal pathogens through a variety of different mechanisms. This will allow a reduction in chemical fungicide use and, therefore, prolong pathogens’ sensitivity to these vital tools while reducing environmental and human exposure. In conclusion, the successful integration of SIGS, biochemical fungicides, biocontrol, and PDEs in conventional spray programs will be beneficial not only to crop health and yield but also to the broader environment and human health, and preserve the efficacy of fungicides by challenging pests with different selection pressures.

Author Contributions

M.S.M. and S.A. conceptualized the idea; M.S.M. wrote the first draft of the manuscript, M.R. contributed to the Biological Control and Biochemical Fungicides section; M.S.M. and S.A. made a major contribution to the final draft; M.R., P.A.A., O.C., S.N.Y. and S.A. critically reviewed the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This project was funded by the Nova Scotia Fruit Growers’ Association and Agriculture and Agri-Food Canada through the Canadian Agricultural Partnership program (J-002190) and STB Annual Call for RDT Proposals (J-002366, J-002516).

Data Availability Statement

The paper will be freely available.

Conflicts of Interest

The authors declare no conflict of interest related to this study.

References

  1. Gullino, M.; Leroux, P.; Smith, C. Uses and challenges of novel compounds for plant disease control. Crop Prot. 2000, 19, 1–11. [Google Scholar] [CrossRef]
  2. Kelman, A. Introduction: The importance of research on the control of postharvest diseases of perishable food crops. Phytopathology 1989, 79, 1374. [Google Scholar]
  3. Ragsdale, N.N.; Sisler, H.D. Social and political implications of managing plant diseases with decreased availability of fungicides in the United States. Annu. Rev. Phytopathol. 1994, 32, 545–557. [Google Scholar] [CrossRef] [PubMed]
  4. Steinhauer, N.; Kulhanek, K.; Antúnez, K.; Human, H.; Chantawannakul, P.; Chauzat, M.P.; vanEngelsdorp, D. Drivers of colony losses. Curr. Opin. Insect. Sci. 2018, 26, 142–148. [Google Scholar] [CrossRef] [PubMed]
  5. Millardet, P.M.A. The Discovery of Bordeaux Mixture; The American Phytopathological Society: St. Paul, MN, USA, 2018. [Google Scholar]
  6. Baibakova, E.; Nefedjeva, E.; Suska-Malawska, M.; Wilk, M.; Sevriukova, G.; Zheltobriukhov, V. Modern Fungicides: Mechanisms of Action, Fungal Resistance and Phytotoxic Effects. Annu. Res. Rev. Biol. 2019, 32, 1–16. [Google Scholar] [CrossRef]
  7. Tamm, L.; Thuerig, B.; Apostolov, S.; Blogg, H.; Borgo, E.; Corneo, P.E.; Fittje, S.; de Palma, M.; Donko, A.; Experton, C.; et al. Use of Copper-Based Fungicides in Organic Agriculture in Twelve European Countries. Agronomy 2022, 12, 673. [Google Scholar] [CrossRef]
  8. Lamichhane, J.R.; Osdaghi, E.; Behlau, F.; Köhl, J.; Jones, J.B.; Aubertot, J.-N. Thirteen decades of antimicrobial copper compounds applied in agriculture. A review. Agron. Sustain. Dev. 2018, 38, 28. [Google Scholar] [CrossRef]
  9. Karuppuchamy, P.; Venugopal, S. Chapter 21—Integrated Pest Management. In Ecofriendly Pest Management for Food Security; Omkar, Ed.; Academic Press: San Diego, CA, USA, 2016; pp. 651–684. [Google Scholar]
  10. Anna La, T.; Valeria, I.; Federica, C. Copper in plant protection: Current situation and prospects. Phytopathol. Mediterr. 2018, 57, 201–236. [Google Scholar] [CrossRef]
  11. Oziengbe, E.O.; Osazee, J.O. Antifungal Activity of Copper Sulphate Against Colletotrichum Gloeosporioides. J. Asian Sci. Res. 2012, 2, 835–839. [Google Scholar]
  12. Oliver, R.; Hewitt, H.G. Fungicides in Crop Protection: Second Edition; CABI: Egham, UK, 2014; pp. 1–190. [Google Scholar]
  13. Dias, M. Phytotoxicity: An Overview of the Physiological Responses of Plants Exposed to Fungicides. J. Bot. 2012, 2012, 135479. [Google Scholar] [CrossRef]
  14. Petit, A.N.; Fontaine, F.; Vatsa, P.; Clément, C.; Vaillant-Gaveau, N. Fungicide impacts on photosynthesis in crop plants. Photosynth. Res. 2012, 111, 315–326. [Google Scholar] [CrossRef] [PubMed]
  15. Kromann, P.; Taipe, A.; Perez, W.; Forbes, G. Rainfall Thresholds as Support for Timing Fungicide Applications in the Control of Potato Late Blight in Ecuador and Peru. Plant Dis. 2009, 93, 142–148. [Google Scholar] [CrossRef] [PubMed]
  16. Vicent, A.; Armengol, J.; García-Jiménez, J. Rain Fastness and Persistence of Fungicides for Control of Alternaria Brown Spot of Citrus. Plant Dis. 2007, 91, 393–399. [Google Scholar] [CrossRef] [PubMed]
  17. Garcia, P.; Rivero, R.; Ruiz, J.; Romero, L. The Role of Fungicides in the Physiology of Higher Plants: Implications for Defense Responses. Bot. Rev. 2003, 69, 162–172. [Google Scholar] [CrossRef]
  18. Klittich, C.J.R. Fungicide Mobility and the Influence of Physical Properties. In Retention, Uptake, and Translocation of Agrochemicals in Plants; ACS Symposium Series; American Chemical Society: Washington, DC, USA, 2014; Volume 1171, pp. 95–109. [Google Scholar]
  19. Klittich, C.J.; Ray, S.L. Effects of physical properties on the translaminar activity of fungicides. Pestic. Biochem. Physiol. 2013, 107, 351–359. [Google Scholar] [CrossRef]
  20. Warneke, B.; Thiessen, L.; Mahaffee, W. Effect of Fungicide Mobility and Application Timing on the Management of Grape Powdery Mildew. Plant Dis. 2019, 104, 1167–1174. [Google Scholar] [CrossRef]
  21. Ayesha, M.S.; Suryanarayanan, T.S.; Nataraja, K.N.; Prasad, S.R.; Shaanker, R.U. Seed Treatment With Systemic Fungicides: Time for Review. Front. Plant Sci. 2021, 12, 654512. [Google Scholar] [CrossRef]
  22. Deising, H.; Reimann, S.; Pascholati, S. Mechanisms and significance of fungicide resistance. Braz. J. Microbiol. Publ. Braz. Soc. Microbiol. 2008, 39, 286–295. [Google Scholar] [CrossRef]
  23. Mohandoss, J.; Suryanarayanan, T. Effect of fungicide treatment on foliar fungal endophyte diversity in mango. Sydowia 2009, 61, 11–24. [Google Scholar]
  24. Leyronas, C.; Mériaux, B.; Raynal, G. Chemical Control of Neotyphodium spp. Endophytes in Perennial Ryegrass and Tall Fescue Seeds. Crop Sci. 2006, 46, 98–104. [Google Scholar] [CrossRef]
  25. Kalia, A.; Gosal, S.K. Effect of pesticide application on soil microorganisms. Arch. Agron. Soil Sci. 2011, 57, 569–596. [Google Scholar] [CrossRef]
  26. Murphy, B.R.; Doohan, F.M.; Hodkinson, T.R. A seed dressing combining fungal endophyte spores and fungicides improves seedling survival and early growth in barley and oat. Symbiosis 2017, 71, 69–76. [Google Scholar] [CrossRef]
  27. Lloyd, A.W.; Percival, D.; Yurgel, S.N. Effect of Fungicide Application on Lowbush Blueberries Soil Microbiome. Microorganisms 2021, 9, 1366. [Google Scholar] [CrossRef]
  28. Lloyd, A.W.; Percival, D.; Langille, M.G.I.; Yurgel, S.N. Changes to Soil Microbiome Resulting from Synergetic Effects of Fungistatic Compounds Pyrimethanil and Fluopyram in Lowbush Blueberry Agriculture, with Nine Fungicide Products Tested. Microorganisms 2023, 11, 410. [Google Scholar] [CrossRef] [PubMed]
  29. Kahle, M.; Buerge, I.J.; Hauser, A.; Müller, M.D.; Poiger, T. Azole Fungicides: Occurrence and Fate in Wastewater and Surface Waters. Environ. Sci. Technol. 2008, 42, 7193–7200. [Google Scholar] [CrossRef] [PubMed]
  30. Bereswill, R.; Golla, B.; Streloke, M.; Schulz, R. Entry and toxicity of organic pesticides and copper in vineyard streams: Erosion rills jeopardise the efficiency of riparian buffer strips. Agric. Ecosyst. Environ. 2012, 146, 81–92. [Google Scholar] [CrossRef]
  31. Zubrod, J.P.; Bundschuh, M.; Arts, G.; Brühl, C.A.; Imfeld, G.; Knäbel, A.; Payraudeau, S.; Rasmussen, J.J.; Rohr, J.; Scharmüller, A.; et al. Fungicides: An Overlooked Pesticide Class? Environ. Sci. Technol. 2019, 53, 3347–3365. [Google Scholar] [CrossRef]
  32. Kiaune, L.; Singhasemanon, N. Pesticidal copper (I) oxide: Environmental fate and aquatic toxicity. Rev. Env. Contam. Toxicol. 2011, 213, 1–26. [Google Scholar] [CrossRef]
  33. Belsky, J.; Joshi, N.K. Effects of Fungicide and Herbicide Chemical Exposure on Apis and Non-Apis Bees in Agricultural Landscape. Front. Environ. Sci. 2020, 8, 81. [Google Scholar] [CrossRef]
  34. Weisenburger, D.D. Human health effects of agrichemical use. Hum. Pathol. 1993, 24, 571–576. [Google Scholar] [CrossRef]
  35. Habig, M.; Lorrain, C.; Feurtey, A.; Komluski, J.; Stukenbrock, E.H. Epigenetic modifications affect the rate of spontaneous mutations in a pathogenic fungus. Nat. Commun. 2021, 12, 5869. [Google Scholar] [CrossRef] [PubMed]
  36. Hermann, D.; Stenzel, K. FRAC Mode-of-action Classification and Resistance Risk of Fungicides. In Modern Crop Protection Compounds; Wiley-VCH: Hoboken, NJ, USA, 2019; pp. 589–608. [Google Scholar]
  37. Katan, J. Cultural approaches for disease management: Present status and future prospects. J. Plant Pathol. 2010, 92, S7–S9. [Google Scholar]
  38. Conway, K.E. An overview of the influence of sustainable agricultural systems on plant diseases. Crop Prot. 1996, 15, 223–228. [Google Scholar] [CrossRef]
  39. Palti, J. Cultural Practices and Infectious Crop Diseases; Springer Science & Business Media: Berlin, Germany, 2012; Volume 9. [Google Scholar]
  40. Fischer, I.H.; Soares-Colletti, A.R.; Palharini, M.C.d.A.; Parisi, M.C.M.; Amorim, L. Temporal progress and spatial patterns of quiescent diseases in guava influenced by sanitation practices. Sci. Agric. 2017, 74, 68–76. [Google Scholar] [CrossRef]
  41. Vincent, C.; Rancourt, B.; Carisse, O. Apple leaf shredding as a non-chemical tool to manage apple scab and spotted tentiform leafminer. Agric. Ecosyst. Environ. 2004, 104, 595–604. [Google Scholar] [CrossRef]
  42. Mertely, J.C.; Chandler, C.K.; Xiao, C.L.; Legard, D.E. Comparison of Sanitation and Fungicides for Management of Botrytis Fruit Rot of Strawberry. Plant Dis. 2000, 84, 1197–1202. [Google Scholar] [CrossRef]
  43. Meitz-Hopkins, J.C.; von Diest, S.G.; Koopman, T.A.; Tobutt, K.R.; Xu, X.; Lennox, C.L. Leaf shredding as an alternative strategy for managing apple scab resistance to demethylation inhibitor fungicides. Front. Hortic. 2023, 2, 1175168. [Google Scholar] [CrossRef]
  44. Sturz, A.; Carter, M.; Johnston, H. A review of plant disease, pathogen interactions and microbial antagonism under conservation tillage in temperate humid agriculture. Soil Tillage Res. 1997, 41, 169–189. [Google Scholar] [CrossRef]
  45. Page, K.; Dang, Y.; Dalal, R. Impacts of conservation tillage on soil quality, including soil-borne crop diseases, with a focus on semi-arid grain cropping systems. Australas. Plant Pathol. 2013, 42, 363–377. [Google Scholar] [CrossRef]
  46. Palojärvi, A.; Kellock, M.; Parikka, P.; Jauhiainen, L.; Alakukku, L. Tillage System and Crop Sequence Affect Soil Disease Suppressiveness and Carbon Status in Boreal Climate. Front. Microbiol. 2020, 11, 534786. [Google Scholar] [CrossRef]
  47. Kerdraon, L.; Laval, V.; Suffert, F. Microbiomes and Pathogen Survival in Crop Residues, an Ecotone Between Plant and Soil. Phytobiomes J. 2019, 3, 246–255. [Google Scholar] [CrossRef]
  48. Bockus, W.W.; Shroyer, J.P. The impact of reduced tillage on soilborne plant pathogens. Annu. Rev. Phytopathol. 1998, 36, 485–500. [Google Scholar] [CrossRef] [PubMed]
  49. Bziuk, N.; Maccario, L.; Douchkov, D.; Lueck, S.; Babin, D.; Sørensen, S.J.; Schikora, A.; Smalla, K. Tillage shapes the soil and rhizosphere microbiome of barley—But not its susceptibility towards Blumeria graminis f. sp. hordei. FEMS Microbiol. Ecol. 2021, 97, fiab018. [Google Scholar] [CrossRef] [PubMed]
  50. Li, Y.; Wang, Z.; Li, T.; Zhao, D.; Han, J.; Liao, Y. Wheat rhizosphere fungal community is affected by tillage and plant growth. Agric. Ecosyst. Environ. 2021, 317, 107475. [Google Scholar] [CrossRef]
  51. Ma, Z.; Guan, Z.; Liu, Q.; Hu, Y.; Liu, L.; Wang, B.; Huang, L.; Li, H.; Yang, Y.; Han, M.; et al. Chapter Four—Obstacles in continuous cropping: Mechanisms and control measures. In Advances in Agronomy; Sparks, D.L., Ed.; Academic Press: Cambridge, MA, USA, 2023; Volume 179, pp. 205–256. [Google Scholar]
  52. Bennett, A.J.; Bending, G.D.; Chandler, D.; Hilton, S.; Mills, P. Meeting the demand for crop production: The challenge of yield decline in crops grown in short rotations. Biol. Rev. Camb. Philos. Soc. 2012, 87, 52–71. [Google Scholar] [CrossRef]
  53. Scholte, K. Effect of crop rotation on the incidence of soil-borne fungal diseases of potato. Neth. J. Plant Pathol. 1992, 98, 93–101. [Google Scholar] [CrossRef]
  54. Yuan, X.; Wang, B.; Hong, S.; Xiong, W.; Shen, Z.; Ruan, Y.; Li, R.; Shen, Q.; Dini-Andreote, F. Promoting soil microbial-mediated suppressiveness against Fusarium wilt disease by the enrichment of specific fungal taxa via crop rotation. Biol. Fertil. Soils 2021, 57, 1137–1153. [Google Scholar] [CrossRef]
  55. Vargas Gil, S.; Meriles, J.M.; Haro, R.; Casini, C.; March, G.J. Crop rotation and tillage systems as a proactive strategy in the control of peanut fungal soilborne diseases. BioControl 2008, 53, 685–698. [Google Scholar] [CrossRef]
  56. Lemańczyk, G.; Wilczewski, E.; Węglarz, W. Effect of catch crop and type of ploughed-in biomass on the health status of stem base and roots of spring wheatOddziaływanie międzyplonów ścierniskowych i rodzaju przyoranej biomasy na zdrowotność podstawy źdźbła i korzeni pszenicy jarej. Prog. Plant Prot. 2016, 56, 19–24. [Google Scholar]
  57. Trinchera, A.; Migliore, M.; Warren Raffa, D.; Ommeslag, S.; Debode, J.; Shanmugam, S.; Dane, S.; Babry, J.; Kivijarvi, P.; Kristensen, H.L.; et al. Can multi-cropping affect soil microbial stoichiometry and functional diversity, decreasing potential soil-borne pathogens? A study on European organic vegetable cropping systems. Front. Plant Sci. 2022, 13, 952910. [Google Scholar] [CrossRef]
  58. Schoeny, A.; Menat, J.; Darsonval, A.; Rouault, F.; Jumel, S.; Tivoli, B. Effect of pea canopy architecture on splash dispersal of Mycosphaerella pinodes conidia. Plant Pathol. 2008, 57, 1073–1085. [Google Scholar] [CrossRef]
  59. Yang, X.; TeBeest, D. Rain dispersal of Colletotrichum gloeosporioides in simulated rice field conditions. Phytopathology 1992, 82, 1219–1222. [Google Scholar] [CrossRef]
  60. Yang, X.; Madden, L.; Wilson, L.; Ellis, M. Effects of surface topography and rain intensity on splash dispersal of Colletotrichum acutatum. Phytopathology 1990, 80, 1115–1120. [Google Scholar] [CrossRef]
  61. Richard, B.; Bussière, F.; Langrume, C.; Rouault, F.; Jumel, S.; Faivre, R.; Tivoli, B. Effect of pea canopy architecture on microclimate and consequences on ascochyta blight infection under field conditions. Eur. J. Plant Pathol. 2013, 135, 509–524. [Google Scholar] [CrossRef]
  62. Simon, S.; Lauri, P.E.; Brun, L.; Defrance, H.; Sauphanor, B. Does manipulation of fruit-tree architecture affect the development of pests and pathogens? A case study in an organic apple orchard. J. Hortic. Sci. Biotechnol. 2006, 81, 765–773. [Google Scholar] [CrossRef]
  63. Vidal, T.; Boixel, A.-L.; Durand, B.; de Vallavieille-Pope, C.; Huber, L.; Saint-Jean, S. Reduction of fungal disease spread in cultivar mixtures: Impact of canopy architecture on rain-splash dispersal and on crop microclimate. Agric. For. Meteorol. 2017, 246, 154–161. [Google Scholar] [CrossRef]
  64. Bent, A.F.; Mackey, D. Elicitors, effectors, and R genes: The new paradigm and a lifetime supply of questions. Annu. Rev. Phytopathol. 2007, 45, 399–436. [Google Scholar] [CrossRef]
  65. Jones, J.D.; Vance, R.E.; Dangl, J.L. Intracellular innate immune surveillance devices in plants and animals. Science 2016, 354, aaf6395. [Google Scholar] [CrossRef]
  66. Shao, Z.-Q.; Xue, J.-Y.; Wu, P.; Zhang, Y.-M.; Wu, Y.; Hang, Y.-Y.; Wang, B.; Chen, J.-Q. Large-Scale Analyses of Angiosperm Nucleotide-Binding Site-Leucine-Rich Repeat Genes Reveal Three Anciently Diverged Classes with Distinct Evolutionary Patterns. Plant Physiol. 2016, 170, 2095–2109. [Google Scholar] [CrossRef]
  67. Shao, F.; Golstein, C.; Ade, J.; Stoutemyer, M.; Dixon, J.E.; Innes, R.W. Cleavage of Arabidopsis PBS1 by a bacterial type III effector. Science 2003, 301, 1230–1233. [Google Scholar] [CrossRef]
  68. Chisholm, S.T.; Coaker, G.; Day, B.; Staskawicz, B.J. Host-Microbe Interactions: Shaping the Evolution of the Plant Immune Response. Cell 2006, 124, 803–814. [Google Scholar] [CrossRef] [PubMed]
  69. Mur, L.A.J.; Kenton, P.; Lloyd, A.J.; Ougham, H.; Prats, E. The hypersensitive response; the centenary is upon us but how much do we know? J. Exp. Bot. 2008, 59, 501–520. [Google Scholar] [CrossRef] [PubMed]
  70. Liu, P.-P.; Yang, Y.; Pichersky, E.; Klessig, D.F. Altering Expression of Benzoic Acid/Salicylic Acid Carboxyl Methyltransferase 1 Compromises Systemic Acquired Resistance and PAMP-Triggered Immunity in Arabidopsis. Mol. Plant-Microbe Interact. 2010, 23, 82–90. [Google Scholar] [CrossRef]
  71. Tsuda, K.; Sato, M.; Stoddard, T.; Glazebrook, J.; Katagiri, F. Network properties of robust immunity in plants. PLoS Genet. 2009, 5, e1000772. [Google Scholar] [CrossRef] [PubMed]
  72. Kaur, B.; Bhatia, D.; Mavi, G.S. Eighty years of gene-for-gene relationship and its applications in identification and utilization of R genes. J. Genet. 2021, 100, 50. [Google Scholar] [CrossRef] [PubMed]
  73. Khajuria, Y.P.; Kaul, S.; Wani, A.A.; Dhar, M.K. Genetics of resistance in apple against Venturia inaequalis (Wint.) Cke. Tree Genet. Genomes 2018, 14, 16. [Google Scholar] [CrossRef]
  74. Kou, Y.; Wang, S. Broad-spectrum and durability: Understanding of quantitative disease resistance. Curr. Opin. Plant Biol. 2010, 13, 181–185. [Google Scholar] [CrossRef] [PubMed]
  75. Ashfield, T.; Ong, L.E.; Nobuta, K.; Schneider, C.M.; Innes, R.W. Convergent evolution of disease resistance gene specificity in two flowering plant families. Plant Cell 2004, 16, 309–318. [Google Scholar] [CrossRef]
  76. Stahl, E.A.; Dwyer, G.; Mauricio, R.; Kreitman, M.; Bergelson, J. Dynamics of disease resistance polymorphism at the Rpm1 locus of Arabidopsis. Nature 1999, 400, 667–671. [Google Scholar] [CrossRef]
  77. Rimbaud, L.; Papaïx, J.; Barrett, L.G.; Burdon, J.J.; Thrall, P.H. Mosaics, mixtures, rotations or pyramiding: What is the optimal strategy to deploy major gene resistance? Evol. Appl. 2018, 11, 1791–1810. [Google Scholar] [CrossRef]
  78. Mundt, C.C. Durable resistance: A key to sustainable management of pathogens and pests. Infect. Genet. Evol. 2014, 27, 446–455. [Google Scholar] [CrossRef] [PubMed]
  79. Pandolfi, V.; Neto, J.; da Silva, M.D.; Amorim, L.L.B.; Wanderley-Nogueira, A.C.; de Oliveira Silva, R.L.; Kido, E.A.; Crovella, S.; Iseppon, A.M.B. Resistance (R) Genes: Applications and Prospects for Plant Biotechnology and Breeding. Curr. Protein Pept. Sci. 2017, 18, 323–334. [Google Scholar] [CrossRef] [PubMed]
  80. Mundt, C. Pyramiding for Resistance Durability: Theory and Practice. Phytopathology 2018, 108, 792–802. [Google Scholar] [CrossRef] [PubMed]
  81. Perez, W.; Salas, A.; Raymundo, R.; Huamán, Z.; Nelson, R.; Bonierbale, M. Evaluation of Wild Potato Species for Resistance to Late Blight. CIP Program Rep. 1999, 2000, 49–62. [Google Scholar]
  82. Vyska, M.; Cunniffe, N.; Gilligan, C. Trade-off between disease resistance and crop yield: A landscape-scale mathematical modelling perspective. J. R. Soc. Interface 2016, 13, 20160451. [Google Scholar] [CrossRef]
  83. Zaidi, S.S.; Mukhtar, M.S.; Mansoor, S. Genome Editing: Targeting Susceptibility Genes for Plant Disease Resistance. Trends Biotechnol. 2018, 36, 898–906. [Google Scholar] [CrossRef] [PubMed]
  84. van Schie, C.C.; Takken, F.L. Susceptibility genes 101: How to be a good host. Annu. Rev. Phytopathol. 2014, 52, 551–581. [Google Scholar] [CrossRef]
  85. Kieu, N.P.; Lenman, M.; Wang, E.S.; Petersen, B.L.; Andreasson, E. Mutations introduced in susceptibility genes through CRISPR/Cas9 genome editing confer increased late blight resistance in potatoes. Sci. Rep. 2021, 11, 4487. [Google Scholar] [CrossRef]
  86. Zhou, H.; Bai, S.; Wang, N.; Sun, X.; Zhang, Y.; Zhu, J.; Dong, C. CRISPR/Cas9-Mediated Mutagenesis of MdCNGC2 in Apple Callus and VIGS-Mediated Silencing of MdCNGC2 in Fruits Improve Resistance to Botryosphaeria dothidea. Front. Plant Sci. 2020, 11, 575477. [Google Scholar] [CrossRef]
  87. Yin, K.; Qiu, J.L. Genome editing for plant disease resistance: Applications and perspectives. Philos. Trans. R. Soc. B 2019, 374, 20180322. [Google Scholar] [CrossRef]
  88. Sticher, L.; Mauch-Mani, B.; Métraux, J.P. Systemic acquired resistance. Annu. Rev. Phytopathol. 1997, 35, 235–270. [Google Scholar] [CrossRef]
  89. Fu, Z.Q.; Dong, X. Systemic acquired resistance: Turning local infection into global defense. Annu. Rev. Plant Biol. 2013, 64, 839–863. [Google Scholar] [CrossRef]
  90. Bektas, Y.; Eulgem, T. Synthetic plant defense elicitors. Front. Plant Sci. 2015, 5, 804. [Google Scholar] [CrossRef]
  91. Malamy, J.; Carr, J.P.; Klessig, D.F.; Raskin, I. Salicylic Acid: A likely endogenous signal in the resistance response of tobacco to viral infection. Science 1990, 250, 1002–1004. [Google Scholar] [CrossRef]
  92. Gaffney, T.; Friedrich, L.; Vernooij, B.; Negrotto, D.; Nye, G.; Uknes, S.; Ward, E.; Kessmann, H.; Ryals, J. Requirement of salicylic Acid for the induction of systemic acquired resistance. Science 1993, 261, 754–756. [Google Scholar] [CrossRef]
  93. Yu, Y.; Gui, Y.; Li, Z.; Jiang, C.; Guo, J.; Niu, D. Induced Systemic Resistance for Improving Plant Immunity by Beneficial Microbes. Plants 2022, 11, 386. [Google Scholar] [CrossRef]
  94. Thomma, B.P.; Eggermont, K.; Penninckx, I.A.; Mauch-Mani, B.; Vogelsang, R.; Cammue, B.P.; Broekaert, W.F. Separate jasmonate-dependent and salicylate-dependent defense-response pathways in Arabidopsis are essential for resistance to distinct microbial pathogens. Proc. Natl. Acad. Sci. USA 1998, 95, 15107–15111. [Google Scholar] [CrossRef]
  95. Koornneef, A.; Leon-Reyes, A.; Ritsema, T.; Verhage, A.; Den Otter, F.C.; Van Loon, L.C.; Pieterse, C.M.J. Kinetics of salicylate-mediated suppression of jasmonate signaling reveal a role for redox modulation. Plant Physiol. 2008, 147, 1358–1368. [Google Scholar] [CrossRef]
  96. Höfte, M. Induced Resistance for Plant Defence. A Sustainable Approach to Crop Protection—Edited by Dale Walters, Adrian Newton and Gary Lyon. Plant Pathol. 2007, 56, 1036–1037. [Google Scholar] [CrossRef]
  97. Wise, M.L. Plant Defense Activators: Application and Prospects in Cereal Crops. In 50 Years of Phytochemistry Research: Volume 43; Gang, D.R., Ed.; Springer International Publishing: Cham, Switzerland, 2013; pp. 55–70. [Google Scholar]
  98. Li, T.; Huang, Y.; Xu, Z.-S.; Wang, F.; Xiong, A.-S. Salicylic acid-induced differential resistance to the Tomato yellow leaf curl virus among resistant and susceptible tomato cultivars. BMC Plant. Biol. 2019, 19, 173. [Google Scholar] [CrossRef]
  99. Deenamo, N.; Kuyyogsuy, A.; Khompatara, K.; Chanwun, T.; Ekchaweng, K.; Churngchow, N. Salicylic Acid Induces Resistance in Rubber Tree against Phytophthora palmivora. Int. J. Mol. Sci. 2018, 19, 1883. [Google Scholar] [CrossRef]
  100. Zhang, Y.; Shi, X.; Li, B.; Zhang, Q.; Liang, W.; Wang, C. Salicylic acid confers enhanced resistance to Glomerella leaf spot in apple. Plant Physiol. Biochem. 2016, 106, 64–72. [Google Scholar] [CrossRef]
  101. Sumayo, M.S.; Son, J.S.; Ghim, S.Y. Exogenous application of phenylacetic acid promotes root hair growth and induces the systemic resistance of tobacco against bacterial soft-rot pathogen Pectobacterium carotovorum subsp. carotovorum. Funct. Plant Biol. 2018, 45, 1119–1127. [Google Scholar] [CrossRef]
  102. Cao, J.; Zeng, K.; Jiang, W. Enhancement of Postharvest Disease Resistance in Ya Li Pear (Pyrus bretschneideri) Fruit by Salicylic Acid Sprays on the Trees during Fruit Growth. Eur. J. Plant Pathol. 2006, 114, 363–370. [Google Scholar] [CrossRef]
  103. El Hadrami, A.; Adam, L.R.; El Hadrami, I.; Daayf, F. Chitosan in Plant Protection. Mar. Drugs 2010, 8, 968–987. [Google Scholar] [CrossRef]
  104. Lu, C.; Liu, H.; Jiang, D.; Wang, L.; Jiang, Y.; Tang, S.; Hou, X.; Han, X.; Liu, Z.; Zhang, M.; et al. Paecilomyces variotii extracts (ZNC) enhance plant immunity and promote plant growth. Plant Soil 2019, 441, 383–397. [Google Scholar] [CrossRef]
  105. Faugeron-Girard, C.; Gloaguen, V.; Koçi, R.; Célérier, J.; Raynaud, A.; Moine, C. Use of a Pleurotus ostreatus Complex Cell Wall Extract as Elicitor of Plant Defenses: From Greenhouse to Field Trial. Molecules 2020, 25, 1094. [Google Scholar] [CrossRef]
  106. Margaritopoulou, T.; Toufexi, E.; Kizis, D.; Balayiannis, G.; Anagnostopoulos, C.; Theocharis, A.; Rempelos, L.; Troyanos, Y.; Leifert, C.; Markellou, E. Reynoutria sachalinensis extract elicits SA-dependent defense responses in courgette genotypes against powdery mildew caused by Podosphaera xanthii. Sci. Rep. 2020, 10, 3354. [Google Scholar] [CrossRef]
  107. Jayaraman, J.; Wan, A.; Rahman, M.; Punja, Z. Application of seaweed extract reduces foliar fungal diseases on carrot. Can. J. Plant Pathol. 2009, 31, 137–138. [Google Scholar]
  108. Esserti, S.; Smaili, A.; Rifai, L.A.; Koussa, T.; Makroum, K.; Belfaiza, M.; Kabil, E.M.; Faize, L.; Burgos, L.; Alburquerque, N.; et al. Protective effect of three brown seaweed extracts against fungal and bacterial diseases of tomato. J. Appl. Phycol. 2016, 29, 1081–1093. [Google Scholar] [CrossRef]
  109. Wang, Z.; Jia, C.; Li, J.; Huang, S.; Xu, B.; Jin, Z. Activation of salicylic acid metabolism and signal transduction can enhance resistance to Fusarium wilt in banana (Musa acuminata L. AAA group, cv. Cavendish). Funct. Integr. Genom. 2015, 15, 47–62. [Google Scholar] [CrossRef]
  110. Heidel, A.J.; Clarke, J.D.; Antonovics, J.; Dong, X. Fitness costs of mutations affecting the systemic acquired resistance pathway in Arabidopsis thaliana. Genetics 2004, 168, 2197–2206. [Google Scholar] [CrossRef]
  111. Heil, M. The Ecological Concept of Costs of Induced Systemic Resistance (ISR). Eur. J. Plant Pathol. 2001, 107, 137–146. [Google Scholar] [CrossRef]
  112. Ngullie, C.R.; Tank, R.; Bhanderi, D. Effect of salicylic acid and humic acid on flowering, fruiting, yield and quality of mango (Mangifera indica L.) cv. KESAR. Adv. Res. J. Crop Improv. 2014, 5, 136–139. [Google Scholar] [CrossRef]
  113. Ali, O.; Ramsubhag, A.; Jayaraman, J. Biostimulant Properties of Seaweed Extracts in Plants: Implications towards Sustainable Crop Production. Plants 2021, 10, 531. [Google Scholar] [CrossRef]
  114. LaMondia, J.A. Actigard Increases Fungicide Efficacy Against Tobacco Blue Mold. Plant Dis. 2008, 92, 1463–1467. [Google Scholar] [CrossRef]
  115. Venegas-Molina, J.; Proietti, S.; Pollier, J.; Orozco-Freire, W.; Ramirez-Villacis, D.; Leon-Reyes, A. Induced tolerance to abiotic and biotic stresses of broccoli and Arabidopsis after treatment with elicitor molecules. Sci. Rep. 2020, 10, 10319. [Google Scholar] [CrossRef]
  116. Kottb, M.; Gigolashvili, T.; Großkinsky, D.K.; Piechulla, B. Trichoderma volatiles effecting Arabidopsis: From inhibition to protection against phytopathogenic fungi. Front. Microbiol. 2015, 6, 995. [Google Scholar] [CrossRef]
  117. De Britto, S.; Joshi, S.M.; Jogaiah, S. Trehalose: A mycogenic cell wall elicitor elicit resistance against leaf spot disease of broccoli and acts as a plant growth regulator. Biotechnol. Rep. 2021, 32, e00690. [Google Scholar] [CrossRef]
  118. Jayaraman, J.; Norrie, J.; Punja, Z.K. Commercial extract from the brown seaweed Ascophyllum nodosum reduces fungal diseases in greenhouse cucumber. J. Appl. Phycol. 2011, 23, 353–361. [Google Scholar] [CrossRef]
  119. Métraux, J.; Ahlgoy, P.; Staub, T.; Speich, J.; Steinemann, A.; Ryals, J.; Ward, E. Induced systemic resistance in cucumber in response to 2, 6-dichloro-isonicotinic acid and pathogens. In Advances in Molecular Genetics of Plant-Microbe Interactions Vol. 1; Springer: Berlin/Heidelberg, Germany, 1991; pp. 432–439. [Google Scholar]
  120. Jayaraj, J.; Wan, A.; Rahman, M.; Punja, Z.K. Seaweed extract reduces foliar fungal diseases on carrot. Crop Prot. 2008, 27, 1360–1366. [Google Scholar] [CrossRef]
  121. Nehela, Y.; Taha, N.A.; Elzaawely, A.A.; Xuan, T.D.; Amin, M.A.; Ahmed, M.E.; El-Nagar, A. Benzoic Acid and Its Hydroxylated Derivatives Suppress Early Blight of Tomato (Alternaria solani) via the Induction of Salicylic Acid Biosynthesis and Enzymatic and Nonenzymatic Antioxidant Defense Machinery. J. Fungi 2021, 7, 663. [Google Scholar] [CrossRef]
  122. Ali, O.; Ramsubhag, A.; Jayaraman, J. Biostimulatory activities of Ascophyllum nodosum extract in tomato and sweet pepper crops in a tropical environment. PLoS ONE 2019, 14, e0216710. [Google Scholar] [CrossRef]
  123. Mehari, Z.H.; Elad, Y.; Rav-David, D.; Graber, E.R.; Meller Harel, Y. Induced systemic resistance in tomato (Solanum lycopersicum) against Botrytis cinerea by biochar amendment involves jasmonic acid signaling. Plant Soil 2015, 395, 31–44. [Google Scholar] [CrossRef]
  124. Chalfoun, N.R.; Durman, S.B.; Budeguer, F.; Caro, M.D.P.; Bertani, R.P.; Di Peto, P.; Stenglein, S.A.; Filippone, M.P.; Moretti, E.R.; Díaz Ricci, J.C.; et al. Development of PSP1, a Biostimulant Based on the Elicitor AsES for Disease Management in Monocot and Dicot Crops. Front. Plant Sci. 2018, 9, 844. [Google Scholar] [CrossRef]
  125. Sangpueak, R.; Phansak, P.; Thumanu, K.; Siriwong, S.; Wongkaew, S.; Buensanteai, N. Effect of Salicylic AcidFormulations on Induced Plant Defense against Cassava Anthracnose Disease. Plant Pathol. J. 2021, 37, 356–364. [Google Scholar] [CrossRef]
  126. Abdel-Monaim, M.F.; Ismail, M.E.; Morsy, K.M.J.M. Induction of systemic resistance of benzothiadiazole and humic acid in soybean plants against Fusarium wilt disease. Mycobiology 2011, 39, 290–298. [Google Scholar] [CrossRef]
  127. Chávez-Arias, C.C.; Gómez-Caro, S.; Restrepo-Díaz, H.J.P. Physiological responses to the foliar application of synthetic resistance elicitors in cape gooseberry seedlings infected with Fusarium oxysporum f. sp. physali. Plants 2020, 9, 176. [Google Scholar] [CrossRef]
  128. Everts, K.L.; Egel, D.S.; Langston, D.; Zhou, X.-G. Chemical management of Fusarium wilt of watermelon. Crop Prot. 2014, 66, 114–119. [Google Scholar] [CrossRef]
  129. Acosta-González, U.; Silva-Rojas, H.V.; Fuentes-Aragón, D.; Hernández-Castrejón, J.; Romero-Bautista, A.; Rebollar-Alviter, A. Comparative Performance of Fungicides and Biocontrol Products in the Management of Fusarium Wilt of Blackberry. Plant Dis. 2022, 106, 1419–1427. [Google Scholar] [CrossRef]
  130. Olivieri, F.P.; Lobato, M.C.; González Altamiranda, E.; Daleo, G.R.; Huarte, M.; Guevara, M.G.; Andreu, A.B. BABA effects on the behaviour of potato cultivars infected by Phytophthora infestans and Fusarium solani. Eur. J. Plant Pathol. 2009, 123, 47–56. [Google Scholar] [CrossRef]
  131. Abo-Elyousr, K.; Ahmed, H.A.; Hassan, M.A.; Abd El-Fatah, B.E. Influence of foliar application of some salts, phyto-extracts and essential oils for contolling powdery mildew disease of Helianthus annuus. J. Plant Pathol. 2022, 104, 735–747. [Google Scholar] [CrossRef]
  132. Gilardi, G.; Demarchi, S.; Gullino, M.L.; Garibaldi, A.J. Management of leaf spot of wild rocket using fungicides, resistance inducers and a biocontrol agent, under greenhouse conditions. Crop Prot. 2015, 71, 39–44. [Google Scholar] [CrossRef]
  133. Elsharkawy, M.M.; Omara, R.I.; Mostafa, Y.S.; Alamri, S.A.; Hashem, M.; Alrumman, S.A.; Ahmad, A.A. Mechanism of Wheat Leaf Rust Control Using Chitosan Nanoparticles and Salicylic Acid. J. Fungi 2022, 8, 304. [Google Scholar] [CrossRef]
  134. Mejri, S.; Magnin-Robert, M.; Randoux, B.; Ghinet, A.; Halama, P.; Siah, A.; Reignault, P. Saccharin Provides Protection and Activates Defense Mechanisms in Wheat Against the Hemibiotrophic Pathogen Zymoseptoria tritici. Plant Dis. 2021, 105, 780–786. [Google Scholar] [CrossRef] [PubMed]
  135. Wang, L.; Liu, H.; Yin, Z.; Li, Y.; Lu, C.; Wang, Q.; Ding, X. A Novel Guanine Elicitor Stimulates Immunity in Arabidopsis and Rice by Ethylene and Jasmonic Acid Signaling Pathways. Front. Plant Sci. 2022, 13, 841228. [Google Scholar] [CrossRef] [PubMed]
  136. Tjosvold, S.; Koike, S.T. Evaluation of reduced risk and other biorational fungicides on the control of powdery mildew on greenhouse roses. In Proceedings of the III International Symposium on Rose Research and Cultivation 547, Herzliya, Israel, 21–26 May 2000; pp. 59–67. [Google Scholar]
  137. Álvarez-Loayza, P.; White, J.F., Jr.; Torres, M.S.; Balslev, H.; Kristiansen, T.; Svenning, J.C.; Gil, N. Light converts endosymbiotic fungus to pathogen, influencing seedling survival and niche-space filling of a common tropical tree, Iriartea deltoidea. PLoS ONE 2011, 6, e16386. [Google Scholar] [CrossRef]
  138. Kuo, H.C.; Hui, S.; Choi, J.; Asiegbu, F.O.; Valkonen, J.P.; Lee, Y.H. Secret lifestyles of Neurospora crassa. Sci. Rep. 2014, 4, 5135. [Google Scholar] [CrossRef]
  139. Mengistu, A.A. Endophytes: Colonization, Behaviour, and Their Role in Defense Mechanism. Int. J. Microbiol. 2020, 2020, 6927219. [Google Scholar] [CrossRef]
  140. Bosamia, T.C.; Barbadikar, K.M.; Modi, A. 9—Genomic insights of plant endophyte interaction: Prospective and impact on plant fitness. In Microbial Endophytes; Kumar, A., E.K, R., Eds.; Woodhead Publishing: Sawston, UK, 2020; pp. 227–249. [Google Scholar]
  141. Busby, P.E.; Ridout, M.; Newcombe, G. Fungal endophytes: Modifiers of plant disease. Plant Mol. Biol. 2016, 90, 645–655. [Google Scholar] [CrossRef]
  142. Backman, P.; Sikora, R. Endophytes: An emerging tool for biological control. Biol. Control 2008, 46, 1–3. [Google Scholar] [CrossRef]
  143. Rodriguez, R.J.; White, J.F., Jr.; Arnold, A.E.; Redman, R.S. Fungal endophytes: Diversity and functional roles. New Phytol. 2009, 182, 314–330. [Google Scholar] [CrossRef] [PubMed]
  144. Redman, R.S.; Freeman, S.; Clifton, D.R.; Morrel, J.; Brown, G.; Rodriguez, R.J. Biochemical Analysis of Plant Protection Afforded by a Nonpathogenic Endophytic Mutant of Colletotrichum magna1. Plant Physiol. 1999, 119, 795–804. [Google Scholar] [CrossRef] [PubMed]
  145. Gunatilaka, A.A. Natural products from plant-associated microorganisms: Distribution, structural diversity, bioactivity, and implications of their occurrence. J. Nat. Prod. 2006, 69, 509–526. [Google Scholar] [CrossRef] [PubMed]
  146. Kusari, S.; Hertweck, C.; Spiteller, M. Chemical Ecology of Endophytic Fungi: Origins of Secondary Metabolites. Chem. Biol. 2012, 19, 792–798. [Google Scholar] [CrossRef] [PubMed]
  147. Ludwig-Müller, J. Plants and endophytes: Equal partners in secondary metabolite production? Biotechnol. Lett. 2015, 37, 1325–1334. [Google Scholar] [CrossRef]
  148. Chen, J.-L.; Sun, S.-Z.; Miao, C.-P.; Wu, K.; Chen, Y.-W.; Xu, L.-H.; Guan, H.-L.; Zhao, L.-X. Endophytic Trichoderma gamsii YIM PH30019: A promising biocontrol agent with hyperosmolar, mycoparasitism, and antagonistic activities of induced volatile organic compounds on root-rot pathogenic fungi of Panax notoginseng. J. Ginseng Res. 2016, 40, 315–324. [Google Scholar] [CrossRef]
  149. Qualhato, T.F.; Lopes, F.A.; Steindorff, A.S.; Brandão, R.S.; Jesuino, R.S.; Ulhoa, C.J. Mycoparasitism studies of Trichoderma species against three phytopathogenic fungi: Evaluation of antagonism and hydrolytic enzyme production. Biotechnol. Lett. 2013, 35, 1461–1468. [Google Scholar] [CrossRef]
  150. Quimby, P.C.; King, L.R.; Grey, W.E. Biological control as a means of enhancing the sustainability of crop/land management systems. Agric. Ecosyst. Environ. 2002, 88, 147–152. [Google Scholar] [CrossRef]
  151. Griffin, M.R. Biocontrol and bioremediation: Two areas of endophytic research which hold great promise. In Advances in Endophytic Research; Springer: New Delhi, India, 2014; pp. 257–282. [Google Scholar]
  152. Magan, N. Importance of Ecological Windows for Efficacy of Biocontrol Agents. In How Research Can Stimulate the Development of Commercial Biological Control against Plant Diseases; De Cal, A., Melgarejo, P., Magan, N., Eds.; Springer International Publishing: Cham, Switzerland, 2020; pp. 1–14. [Google Scholar]
  153. Anees, M.; Tronsmo, A.; Edel-Hermann, V.; Hjeljord, L.G.; Héraud, C.; Steinberg, C. Characterization of field isolates of Trichoderma antagonistic against Rhizoctonia solani. Fungal Biol. 2010, 114, 691–701. [Google Scholar] [CrossRef]
  154. Ahamad, L.; Zaidi, R.K. Seed Priming Through Fungicides, Biocontrol Agents and Botanicals as Effective Method for Controlling Spot Blotch Pathogen, Bipolaris sorokiniana in Barley. Gesunde Pflanz. 2022, 74, 501–510. [Google Scholar] [CrossRef]
  155. Hermosa, R.; Cardoza, R.E.; Rubio, M.B.; Gutiérrez, S.; Monte, E. Secondary metabolism and antimicrobial metabolites of Trichoderma. In Biotechnology and biology of Trichoderma; Elsevier: Amsterdam, The Netherlands, 2014; pp. 125–137. [Google Scholar]
  156. Mukhopadhyay, R.; Kumar, D. Trichoderma: A beneficial antifungal agent and insights into its mechanism of biocontrol potential. Egypt. J. Biol. Pest Control 2020, 30, 133. [Google Scholar] [CrossRef]
  157. Iqbal, M.; Jützeler, M.; França, S.C.; Wäckers, F.; Andreasson, E.; Stenberg, J.A. Bee-Vectored Aureobasidium pullulans for Biological Control of Gray Mold in Strawberry. Phytopathology 2022, 112, 232–237. [Google Scholar] [CrossRef] [PubMed]
  158. Pinto, C.; Custódio, V.; Nunes, M.; Songy, A.; Rabenoelina, F.; Courteaux, B.; Clément, C.; Gomes, A.C.; Fontaine, F. Understand the potential role of Aureobasidium pullulans, a resident microorganism from grapevine, to prevent the infection caused by Diplodia seriata. Front. Microbiol. 2018, 9, 3047. [Google Scholar] [CrossRef]
  159. Bencheqroun, S.; Bajji, M.; Sebastien, M.; Bentata, F.; Labhilili, M.; el hassan, A.; el Jaafari, S.; Jijakli, M. Biocontrol of blue mold on apple fruits by Aureobasidium pullulans (strain Ach 1-1): In vitro and in situ evidence for the possible involvement of competition for nutrients. Commun. Agric. Appl. Biol. Sci. 2006, 71, 1151–1157. [Google Scholar]
  160. Schena, L.; Nigro, F.; Pentimone, I.; Ligorio, A.; Ippolito, A. Control of postharvest rots of sweet cherries and table grapes with endophytic isolates of Aureobasidium pullulans. Postharvest Biol. Technol. 2003, 30, 209–220. [Google Scholar] [CrossRef]
  161. Agirman, B.; Erten, H. Biocontrol ability and action mechanisms of Aureobasidium pullulans GE17 and Meyerozyma guilliermondii KL3 against Penicillium digitatum DSM2750 and Penicillium expansum DSM62841 causing postharvest diseases. Yeast 2020, 37, 437–448. [Google Scholar] [CrossRef]
  162. Di francesco, A.; Ugolini, L.; Lazzeri, L.; Mari, M. Production of volatile organic compounds by Aureobasidium pullulans as a potential mechanism of action against postharvest fruit pathogens. Biol. Control 2015, 81, 8–14. [Google Scholar] [CrossRef]
  163. Bozoudi, D.; Tsaltas, D. The Multiple and Versatile Roles of Aureobasidium pullulans in the Vitivinicultural Sector. Fermentation 2018, 4, 85. [Google Scholar] [CrossRef]
  164. Pujol, M.; Badosa, E.; Cabrefiga, J.; Montesinos, E. Development of a strain-specific quantitative method for monitoring Pseudomonas fluorescens EPS62e, a novel biocontrol agent of fire blight. FEMS Microbiol. Lett. 2005, 249, 343–352. [Google Scholar] [CrossRef]
  165. Santoyo, G.; Orozco-Mosqueda, M.d.C.; Govindappa, M. Mechanisms of biocontrol and plant growth-promoting activity in soil bacterial species of Bacillus and Pseudomonas: A review. Biocontrol Sci. Technol. 2012, 22, 855–872. [Google Scholar] [CrossRef]
  166. Vicedo, B.; Peñalver, R.; Asins, M.J.; López, M.M. Biological Control of Agrobacterium tumefaciens, Colonization, and pAgK84 Transfer with Agrobacterium radiobacter K84 and the Tra Mutant Strain K1026. Appl. Env. Microbiol. 1993, 59, 309–315. [Google Scholar] [CrossRef] [PubMed]
  167. Collinge, D.B.; Jensen, D.F.; Rabiey, M.; Sarrocco, S.; Shaw, M.W.; Shaw, R.H. Biological control of plant diseases—What has been achieved and what is the direction? Plant Pathol. 2022, 71, 1024–1047. [Google Scholar] [CrossRef]
  168. Bardin, M.; Ajouz, S.; Comby, M.; Lopez-Ferber, M.; Graillot, B.; Siegwart, M.; Nicot, P.C. Is the efficacy of biological control against plant diseases likely to be more durable than that of chemical pesticides? Front. Plant Sci. 2015, 6, 566. [Google Scholar] [CrossRef]
  169. Guetsky, R.; Shtienberg, D.; Elad, Y.; Dinoor, A. Combining biocontrol agents to reduce the variability of biological control. Phytopathology 2001, 91, 621–627. [Google Scholar] [CrossRef]
  170. García-Pedrajas, M.D.; Cañizares, M.C.; Sarmiento-Villamil, J.L.; Jacquat, A.G.; Dambolena, J.S. Mycoviruses in Biological Control: From Basic Research to Field Implementation. Phytopathology 2019, 109, 1828–1839. [Google Scholar] [CrossRef]
  171. Chagnon, P.-L. Ecological and evolutionary implications of hyphal anastomosis in arbuscular mycorrhizal fungi. FEMS Microbiol. Ecol. 2014, 88, 437–444. [Google Scholar] [CrossRef]
  172. Xie, J.; Jiang, D. New insights into mycoviruses and exploration for the biological control of crop fungal diseases. Annu. Rev. Phytopathol. 2014, 52, 45–68. [Google Scholar] [CrossRef]
  173. Pearson, M.N.; Beever, R.E.; Boine, B.; Arthur, K. Mycoviruses of filamentous fungi and their relevance to plant pathology. Mol. Plant Pathol. 2009, 10, 115–128. [Google Scholar] [CrossRef]
  174. Li, H.; Bian, R.; Liu, Q.; Yang, L.; Pang, T.; Salaipeth, L.; Andika, I.B.; Kondo, H.; Sun, L. Identification of a Novel Hypovirulence-Inducing Hypovirus From Alternaria alternata. Front. Microbiol. 2019, 10, 1076. [Google Scholar] [CrossRef]
  175. Kamaruzzaman, M.; He, G.; Wu, M.; Zhang, J.; Yang, L.; Chen, W.; Li, G. A Novel Partitivirus in the Hypovirulent Isolate QT5-19 of the Plant Pathogenic Fungus Botrytis cinerea. Viruses 2019, 11, 24. [Google Scholar] [CrossRef] [PubMed]
  176. Buttimer, C.; McAuliffe, O.; Ross, R.P.; Hill, C.; O’Mahony, J.; Coffey, A. Bacteriophages and Bacterial Plant Diseases. Front. Microbiol. 2017, 8, 34. [Google Scholar] [CrossRef] [PubMed]
  177. Born, Y.; Fieseler, L.; Klumpp, J.; Eugster, M.R.; Zurfluh, K.; Duffy, B.; Loessner, M.J. The tail-associated depolymerase of Erwinia amylovora phage L1 mediates host cell adsorption and enzymatic capsule removal, which can enhance infection by other phage. Environ. Microbiol. 2014, 16, 2168–2180. [Google Scholar] [CrossRef] [PubMed]
  178. Das, M.; Bhowmick, T.S.; Ahern, S.J.; Young, R.; Gonzalez, C.F. Control of Pierce’s Disease by Phage. PLoS ONE 2015, 10, e0128902. [Google Scholar] [CrossRef]
  179. dos Santos Gomes, A.C.; da Silva, R.R.; Moreira, S.I.; Vicentini, S.N.C.; Ceresini, P.C. Biofungicides: An Eco-Friendly Approach for Plant Disease Management. In Encyclopedia of Mycology; Zaragoza, Ó., Casadevall, A., Eds.; Elsevier: Oxford, UK, 2021; pp. 641–649. [Google Scholar]
  180. Trejo-Raya, A.B.; Rodríguez-Romero, V.M.; Bautista-Baños, S.; Quiroz-Figueroa, F.R.; Villanueva-Arce, R.; Durán-Páramo, E. Effective in vitro control of two phytopathogens of agricultural interest using cell-free extracts of pseudomonas fluorescens and chitosan. Molecules 2021, 26, 6359. [Google Scholar] [CrossRef]
  181. Kaur, T.; Kaur, A.; Sharma, V.; Manhas, R.K. Purification and Characterization of a New Antifungal Compound 10-(2, 2-dimethyl-cyclohexyl)-6, 9-dihydroxy-4, 9-dimethyl-dec-2-enoic Acid Methyl Ester from Streptomyces hydrogenans Strain DH16. Front. Microbiol. 2016, 7, 1004. [Google Scholar] [CrossRef]
  182. Polo, K.J.J.; Campos, H.L.M.; Olivera, C.C.; Nakayo, J.L.J.; Flores, J.W.V. Biofungicide for the Control of Botrytis Cinerea and Fusarium Oxysporum: A Laboratory Study. Chem. Eng. Trans. 2021, 87, 517–522. [Google Scholar]
  183. Wulff, E.; Zida, E.; Torp, J.; Lund, O. Yucca schidigera extract: A potential biofungicide against seedborne pathogens of sorghum. Plant Pathol. 2012, 61, 331–338. [Google Scholar] [CrossRef]
  184. Baraka, M.; Fatma; Shaban, M.; Arafat, K.H. Efficiency of Some Plant Extracts, Natural Oils, Biofungicides and Fungicides Against Root Rot of Date Palm. J. Biol. Chem. Environ. Sci. 2011, 6, 405–429. [Google Scholar]
  185. Lu, M.; Han, Z.; Yao, L. In vitro and in vivo antimicrobial efficacy of essential oils and individual compounds against Phytophthora parasitica var. nicotianae. J. Appl. Microbiol. 2013, 115, 187–198. [Google Scholar] [CrossRef]
  186. Géza, N.; Hochbaum, T.; Sarosi, S.; Ladanyi, M. In vitro and in planta activity of some essential oils against Venturia inaequalis (Cooke) G. Winter. Not. Bot. Horti Agrobot. Cluj-Napoca 2014, 42, 109–114. [Google Scholar]
  187. Sharma, M.; Tarafdar, A.; Ghosh, R.; Gopalakrishanan, S. Biological Control as a Tool for Eco-friendly Management of Plant Pathogens. In Advances in Soil Microbiology: Recent Trends and Future Prospects; Springer: Berlin/Heidelberg, Germany, 2017; pp. 153–188. [Google Scholar]
  188. Al-Samarrai, G.; Singh, H.; Syarhabil, M. Evaluating eco-friendly botanicals (natural plant extracts) as alternatives to synthetic fungicides. Ann. Agric. Environ. Med. 2012, 19, 4. [Google Scholar]
  189. Wang, J.; Zhang, J.; Ma, J.; Liu, L.; Li, J.; Shen, T.; Tian, Y.J. The major component of cinnamon oil as a natural substitute against Fusarium solani on Astragalus membranaceus. J. Appl. Microbiol. 2022, 132, 3125–3141. [Google Scholar] [CrossRef] [PubMed]
  190. Qiao, Y.; Xu, L.; Xu, G.; Cao, Y.; Gao, Y.; Wang, Y.; Feng, J. Efficacy and potential mechanism of hinokitiol against postharvest anthracnose of banana caused by Colletotrichum musae. LWT 2022, 161, 113334. [Google Scholar] [CrossRef]
  191. Hashem, M.; Moharam, A.M.; Saleh, F.; Alamri, S. Biocontrol efficacy of essential oils of cumin, basil and geranium against Fusarium wilt and root rot of basil. Int. J. Agric. Biol. 2018, 20, 2012–2018. [Google Scholar] [CrossRef]
  192. Khalil, N.M.; Ali, H.M.; Ibrahim, A.E. Biochemical Activity of Propolis Alcoholic Extracts against Fusarium oxysporum hm89. Egypt. J. Bot. 2022, 62, 197–212. [Google Scholar] [CrossRef]
  193. Zaki, S.A.; Ouf, S.A.; Abd-Elsalam, K.A.; Asran, A.A.; Hassan, M.M.; Kalia, A.; Albarakaty, F.M. Trichogenic Silver-Based Nanoparticles for Suppression of Fungi Involved in Damping-Off of Cotton Seedlings. Microorganisms 2022, 10, 344. [Google Scholar] [CrossRef]
  194. Han, L.; Zhao, W.; Li, A.; Zhou, B.; Zhang, J.; Wu, W. Antifungal activity of l-azetidine-2-carboxylic acid isolated from Disporopsis aspera rhizomes against Podosphaera xanthii. Pest Manag. Sci. 2022, 78, 1946–1952. [Google Scholar] [CrossRef]
  195. Safaei, M.; Jorkesh, A.; Olfati, J. Chemical and biological products for control of powdery mildew on cucumber. Int. J. Veg. Sci. 2022, 28, 233–238. [Google Scholar] [CrossRef]
  196. Tanaka, K.; Fukuda, M.; Amaki, Y.; Sakaguchi, T.; Inai, K.; Ishihara, A.; Nakajima, H. Importance of prumycin produced by Bacillus amyloliquefaciens SD-32 in biocontrol against cucumber powdery mildew disease. Pest Manag. Sci. 2017, 73, 2419–2428. [Google Scholar] [CrossRef]
  197. Abdel-Hafez, S.I.I.; Abo-Elyousr, K.A.M.; Abdel-Rahim, I.R. Fungicidal activity of extracellular products of cyanobacteria against Alternaria porri. Eur. J. Phycol. 2015, 50, 239–245. [Google Scholar] [CrossRef]
  198. Salem, M.F.; Abd-Elraoof, W.A.; Tayel, A.A.; Alzuaibr, F.M.; Abonama, O.M.J. Antifungal application of biosynthesized selenium nanoparticles with pomegranate peels and nanochitosan as edible coatings for citrus green mold protection. J. Nanobiotechnol. 2022, 20, 1–12. [Google Scholar] [CrossRef] [PubMed]
  199. Cao, L.-L.; Zhang, Y.-Y.; Liu, Y.-J.; Yang, T.-T.; Zhang, J.-L.; Zhang, Z.-G.; Shen, L.; Liu, J.-Y.; Ye, Y.-H. Anti-phytopathogenic activity of sporothriolide, a metabolite from endophyte Nodulisporium sp. A21 in Ginkgo biloba. Pestic. Biochem. Physiol. 2016, 129, 7–13. [Google Scholar] [CrossRef] [PubMed]
  200. Shahid, M.; Singh, B.N.; Verma, S.; Choudhary, P.; Das, S.; Chakdar, H.; Murugan, K.; Goswami, S.K.; Saxena, A.K. Bioactive antifungal metabolites produced by Streptomyces amritsarensis V31 help to control diverse phytopathogenic fungi. Braz. J. Microbiol. 2021, 52, 1687–1699. [Google Scholar] [CrossRef] [PubMed]
  201. El-Naggar, N.E.-A.; Saber, W.I.; Zweil, A.M.; Bashir, S.I. An innovative green synthesis approach of chitosan nanoparticles and their inhibitory activity against phytopathogenic Botrytis cinerea on strawberry leaves. Sci. Rep. 2022, 12, 3515. [Google Scholar] [CrossRef]
  202. Abdel Rahman, S.M.; Yusef, H.; Halawi, J. Biological and Chemical Control of Some Tomato Fungal Diseases. Egypt. J. Bot. 2022, 62, 45–58. [Google Scholar] [CrossRef]
  203. Ebrahimi, L.; Jalali, H.; Etebarian, H.R.; Sahebani, N. Evaluation of antifungal activity of some plant essential oils against tomato grey mould disease. J. Plant Pathol. 2022, 104, 641–650. [Google Scholar] [CrossRef]
  204. Mugao, L.; Gichimu, B.; Muturi, P.; Njoroge, E. Essential Oils as Biocontrol Agents of Early and Late Blight Diseases of Tomato under Greenhouse Conditions. Int. J. Agron. 2021, 2021, 5719091. [Google Scholar] [CrossRef]
  205. Rguez, S.; Slimene, I.B.; Abid, G.; Hammemi, M.; Kefi, A.; Elkahoui, S.; Ksouri, R.; Sellami, I.H.; Djébali, N. Tetraclinis articulata essential oil reduces Botrytis cinerea infections on tomato. Sci. Hortic. 2020, 266, 109291. [Google Scholar] [CrossRef]
  206. Mostafa, Y.S.; Alamri, S.A.; Alrumman, S.A.; Hashem, M.; Taher, M.A.; Baka, Z.A. In Vitro and In Vivo Biocontrol of Tomato Fusarium Wilt by Extracts from Brown, Red, and Green Macroalgae. Agriculture 2022, 12, 345. [Google Scholar] [CrossRef]
  207. Betz, E.C.; Punja, Z.K. Management of powdery mildew, caused by Erysiphe cruciferarum, on wasabi (Wasabia japonica) plants in British Columbia. Can. J. Plant Pathol. 2021, 43, 16–25. [Google Scholar] [CrossRef]
  208. Di Francesco, A.; Di Foggia, M.; Corbetta, M.; Baldo, D.; Ratti, C.; Baraldi, E. Biocontrol activity and plant growth promotion exerted by Aureobasidium pullulans strains. J. Plant Growth Regul. 2021, 40, 1233–1244. [Google Scholar] [CrossRef]
  209. Wang, X.; Lai, J.; Hu, H.; Yang, J.; Zang, K.; Zhao, F.; Zeng, G.; Liao, Q.; Gu, Z.; Du, Z. Infection of Nigrospora nonsegmented RNA Virus 1 Has Important Biological Impacts on a Fungal Host. Viruses 2022, 14, 795. [Google Scholar] [CrossRef] [PubMed]
  210. Liu, F.; Yang, S.; Xu, F.; Zhang, Z.; Lu, Y.; Zhang, J.; Wang, G. Characteristics of biological control and mechanisms of Pseudomonas chlororaphis zm-1 against peanut stem rot. BMC Microbiol. 2022, 22, 9. [Google Scholar] [CrossRef]
  211. Zhai, L.; Yang, M.; Zhang, M.; Hong, N.; Wang, G. Characterization of a botybirnavirus conferring hypovirulence in the phytopathogenic fungus Botryosphaeria dothidea. Viruses 2019, 11, 266. [Google Scholar] [CrossRef]
  212. Yu, L.; Sang, W.; Wu, M.-D.; Zhang, J.; Yang, L.; Zhou, Y.-J.; Chen, W.-D.; Li, G.-Q. Novel hypovirulence-associated RNA mycovirus in the plant-pathogenic fungus Botrytis cinerea: Molecular and biological characterization. Appl. Environ. Microbiol. 2015, 81, 2299–2310. [Google Scholar] [CrossRef]
  213. Yang, D.; Wu, M.; Zhang, J.; Chen, W.; Li, G.; Yang, L. Sclerotinia minor endornavirus 1, a novel pathogenicity debilitation-associated mycovirus with a wide spectrum of horizontal transmissibility. Viruses 2018, 10, 589. [Google Scholar] [CrossRef]
  214. Kunyosying, D.; To-anun, C.; Cheewangkoon, R. Control of rice blast disease using antagonistic yeasts. Int. J. Agric. Technol. 2018, 14, 83–98. [Google Scholar]
  215. Liu, B.; Stein, L.; Cochran, K.; du Toit, L.J.; Feng, C.; Correll, J.C. Three new fungal leaf spot diseases of spinach in the United States and the evaluation of fungicide efficacy for disease management. Plant Dis. 2021, 105, 316–323. [Google Scholar] [CrossRef]
  216. Farhaoui, A.; Adadi, A.; Tahiri, A.; El Alami, N.; Khayi, S.; Mentag, R.; Ezrari, S.; Radouane, N.; Mokrini, F.; Belabess, Z.; et al. Biocontrol potential of plant growth-promoting rhizobacteria (PGPR) against Sclerotiorum rolfsii diseases on sugar beet (Beta vulgaris L.). Physiol. Mol. Plant Pathol. 2022, 119, 101829. [Google Scholar] [CrossRef]
  217. Imran, M.; Abo-Elyousr, K.A.; Mousa, M.A.; Saad, M.M. Screening and biocontrol evaluation of indigenous native Trichoderma spp. against early blight disease and their field assessment to alleviate natural infection. Egypt. J. Biol. Pest Control. 2022, 32, 1–10. [Google Scholar] [CrossRef]
  218. Egel, D.; Hoagland, L.; Davis, J.; Marchino, C.; Bloomquist, M. Efficacy of organic disease control products on common foliar diseases of tomato in field and greenhouse trials. Crop Prot. 2019, 122, 90–97. [Google Scholar] [CrossRef]
  219. Di Francesco, A.; Di Foggia, M.; Zajc, J.; Gunde-Cimerman, N.; Baraldi, E. Study of the efficacy of Aureobasidium strains belonging to three different species: A. pullulans, A. subglaciale and A. melanogenum against Botrytis cinerea of tomato. Ann. Appl. Biol. 2020, 177, 266–275. [Google Scholar] [CrossRef]
  220. Sarven, M.S.; Hao, Q.; Deng, J.; Yang, F.; Wang, G.; Xiao, Y.; Xiao, X.J.P. Biological control of tomato gray mold caused by Botrytis cinerea with the entomopathogenic fungus Metarhizium anisopliae. Pathogens 2020, 9, 213. [Google Scholar] [CrossRef] [PubMed]
  221. Wang, H.; Shi, Y.; Wang, D.; Yao, Z.; Wang, Y.; Liu, J.; Zhang, S.; Wang, A. A Biocontrol Strain of Bacillus subtilis WXCDD105 Used to Control Tomato Botrytis cinerea and Cladosporium fulvum Cooke and Promote the Growth of Seedlings. Int. J. Mol. Sci. 2018, 19, 1371. [Google Scholar] [CrossRef]
  222. Rao, Y.; Zeng, L.; Jiang, H.; Mei, L.; Wang, Y. Trichoderma atroviride LZ42 releases volatile organic compounds promoting plant growth and suppressing Fusarium wilt disease in tomato seedlings. BMC Microbiol. 2022, 22, 88. [Google Scholar] [CrossRef]
  223. Akhtar, T.; Shakeel, Q.; Sarwar, G.; Muhammad, S.; Iftikhar, Y.; Ullah, M.I.; Mubeen, M.; Hannan, A. Evaluation of fungicides and biopesticides for the control of fusarium wiltof tomato. Pak. J. Bot 2017, 49, 769–774. [Google Scholar]
  224. Verdel, A.; Jia, S.; Gerber, S.; Sugiyama, T.; Gygi, S.; Grewal, S.I.; Moazed, D. RNAi-mediated targeting of heterochromatin by the RITS complex. Science 2004, 303, 672–676. [Google Scholar] [CrossRef]
  225. Pratt, A.J.; MacRae, I.J. The RNA-induced silencing complex: A versatile gene-silencing machine. J. Biol. Chem. 2009, 284, 17897–17901. [Google Scholar] [CrossRef]
  226. Tang, Y.; Yan, X.; Gu, C.; Yuan, X. Biogenesis, Trafficking, and Function of Small RNAs in Plants. Front. Plant Sci. 2022, 13, 825477. [Google Scholar] [CrossRef]
  227. Lewis, B.P.; Burge, C.B.; Bartel, D.P. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 2005, 120, 15–20. [Google Scholar] [CrossRef] [PubMed]
  228. Zeng, Y.; Yi, R.; Cullen, B.R. MicroRNAs and small interfering RNAs can inhibit mRNA expression by similar mechanisms. Proc. Natl. Acad. Sci. USA 2003, 100, 9779–9784. [Google Scholar] [CrossRef] [PubMed]
  229. López-Dolz, L.; Spada, M.; Daròs, J.A.; Carbonell, A. Fine-tune control of targeted RNAi efficacy by plant artificial small RNAs. Nucleic Acids Res. 2020, 48, 6234–6250. [Google Scholar] [CrossRef] [PubMed]
  230. Dugas, D.V.; Bartel, B. Sucrose induction of Arabidopsis miR398 represses two Cu/Zn superoxide dismutases. Plant Mol. Biol. 2008, 67, 403–417. [Google Scholar] [CrossRef] [PubMed]
  231. Liu, Q.; Wang, F.; Axtell, M.J. Analysis of complementarity requirements for plant microRNA targeting using a Nicotiana benthamiana quantitative transient assay. Plant Cell 2014, 26, 741–753. [Google Scholar] [CrossRef] [PubMed]
  232. Obbard, D.J.; Gordon, K.H.; Buck, A.H.; Jiggins, F.M. The evolution of RNAi as a defence against viruses and transposable elements. Philos. Trans. R Soc. Lond B Biol. Sci. 2009, 364, 99–115. [Google Scholar] [CrossRef]
  233. Harvey, J.J.W.; Lewsey, M.G.; Patel, K.; Westwood, J.; Heimstädt, S.; Carr, J.P.; Baulcombe, D.C. An Antiviral Defense Role of AGO2 in Plants. PLoS ONE 2011, 6, e14639. [Google Scholar] [CrossRef]
  234. Koch, A.; Biedenkopf, D.; Furch, A.; Weber, L.; Rossbach, O.; Abdellatef, E.; Linicus, L.; Johannsmeier, J.; Jelonek, L.; Goesmann, A.; et al. An RNAi-Based Control of Fusarium graminearum Infections Through Spraying of Long dsRNAs Involves a Plant Passage and Is Controlled by the Fungal Silencing Machinery. PLoS Pathog. 2016, 12, e1005901. [Google Scholar] [CrossRef]
  235. Koch, A.; Kumar, N.; Weber, L.; Keller, H.; Imani, J.; Kogel, K.H. Host-induced gene silencing of cytochrome P450 lanosterol C14α-demethylase-encoding genes confers strong resistance to Fusarium species. Proc. Natl. Acad. Sci. USA 2013, 110, 19324–19329. [Google Scholar] [CrossRef]
  236. Rana, K.; Ding, Y.; Banga, S.S.; Liao, H.; Zhao, S.; Yu, Y.; Qian, W. Sclerotinia sclerotiorum Thioredoxin1 (SsTrx1) is required for pathogenicity and oxidative stress tolerance. Mol. Plant Pathol. 2021, 22, 1413–1426. [Google Scholar] [CrossRef]
  237. Maximiano, M.R.; Santos, L.S.; Santos, C.; Aragão, F.J.L.; Dias, S.C.; Franco, O.L.; Mehta, A. Host induced gene silencing of Sclerotinia sclerotiorum effector genes for the control of white mold. Biocatal. Agric. Biotechnol. 2022, 40, 102302. [Google Scholar] [CrossRef]
  238. Nowara, D.; Gay, A.; Lacomme, C.; Shaw, J.; Ridout, C.; Douchkov, D.; Hensel, G.; Kumlehn, J.; Schweizer, P. HIGS: Host-Induced Gene Silencing in the Obligate Biotrophic Fungal Pathogen Blumeria graminis. Plant Cell 2010, 22, 3130–3141. [Google Scholar] [CrossRef] [PubMed]
  239. Zhao, M.; Wang, C.; Wan, J.; Li, Z.; Liu, D.; Yamamoto, N.; Zhou, E.; Shu, C. Functional validation of pathogenicity genes in rice sheath blight pathogen Rhizoctonia solani by a novel host-induced gene silencing system. Mol. Plant Pathol. 2021, 22, 1587–1598. [Google Scholar] [CrossRef] [PubMed]
  240. Su, X.; Lu, G.; Li, X.; Rehman, L.; Liu, W.; Sun, G.; Guo, H.; Wang, G.; Cheng, H. Host-Induced Gene Silencing of an Adenylate Kinase Gene Involved in Fungal Energy Metabolism Improves Plant Resistance to Verticillium dahliae. Biomolecules 2020, 10, 127. [Google Scholar] [CrossRef] [PubMed]
  241. Xiong, F.; Liu, M.; Zhuo, F.; Yin, H.; Deng, K.; Feng, S.; Liu, Y.; Luo, X.; Feng, L.; Zhang, S.; et al. Host-induced gene silencing of BcTOR in Botrytis cinerea enhances plant resistance to grey mould. Mol. Plant Pathol. 2019, 20, 1722–1739. [Google Scholar] [CrossRef]
  242. Zhu, L.; Zhu, J.; Liu, Z.; Wang, Z.; Zhou, C.; Wang, H. Host-Induced Gene Silencing of Rice Blast Fungus Magnaporthe oryzae Pathogenicity Genes Mediated by the Brome Mosaic Virus. Genes 2017, 8, 241. [Google Scholar] [CrossRef]
  243. Zhu, X.; Qi, T.; Yang, Q.; He, F.; Tan, C.; Ma, W.; Voegele, R.T.; Kang, Z.; Guo, J. Host-Induced Gene Silencing of the MAPKK Gene PsFUZ7 Confers Stable Resistance to Wheat Stripe Rust. Plant Physiol. 2017, 175, 1853–1863. [Google Scholar] [CrossRef]
  244. Guo, X.Y.; Li, Y.; Fan, J.; Xiong, H.; Xu, F.X.; Shi, J.; Shi, Y.; Zhao, J.Q.; Wang, Y.F.; Cao, X.L.; et al. Host-Induced Gene Silencing of MoAP1 Confers Broad-Spectrum Resistance to Magnaporthe oryzae. Front. Plant Sci. 2019, 10, 433. [Google Scholar] [CrossRef]
  245. Hu, D.; Chen, Z.Y.; Zhang, C.; Ganiger, M. Reduction of Phakopsora pachyrhizi infection on soybean through host- and spray-induced gene silencing. Mol. Plant Pathol. 2020, 21, 794–807. [Google Scholar] [CrossRef]
  246. McCaghey, M.; Shao, D.; Kurcezewski, J.; Lindstrom, A.; Ranjan, A.; Whitham, S.A.; Conley, S.P.; Williams, B.; Smith, D.L.; Kabbage, M. Host-Induced Gene Silencing of a Sclerotinia sclerotiorum oxaloacetate acetylhydrolase Using Bean Pod Mottle Virus as a Vehicle Reduces Disease on Soybean. Front. Plant Sci. 2021, 12, 677631. [Google Scholar] [CrossRef]
  247. Zhu, P.; Zhang, S.; Li, R.; Liu, C.; Fan, W.; Hu, T.; Zhao, A. Host-Induced Gene Silencing of a G Protein α Subunit Gene CsGpa1 Involved in Pathogen Appressoria Formation and Virulence Improves Tobacco Resistance to Ciboria shiraiana. J. Fungi 2021, 7, 1053. [Google Scholar] [CrossRef] [PubMed]
  248. Qi, T.; Zhu, X.; Tan, C.; Liu, P.; Guo, J.; Kang, Z.; Guo, J. Host-induced gene silencing of an important pathogenicity factor PsCPK1 in Puccinia striiformis f. sp. tritici enhances resistance of wheat to stripe rust. Plant Biotechnol. J. 2018, 16, 797–807. [Google Scholar] [CrossRef] [PubMed]
  249. Zhu, X.; Guo, J.; He, F.; Zhang, Y.; Tan, C.; Yang, Q.; Huang, C.; Kang, Z.; Guo, J. Silencing PsKPP4, a MAP kinase kinase kinase gene, reduces pathogenicity of the stripe rust fungus. Mol. Plant Pathol. 2018, 19, 2590–2602. [Google Scholar] [CrossRef] [PubMed]
  250. McLoughlin, A.G.; Wytinck, N.; Walker, P.L.; Girard, I.J.; Rashid, K.Y.; de Kievit, T.; Fernando, W.G.D.; Whyard, S.; Belmonte, M.F. Identification and application of exogenous dsRNA confers plant protection against Sclerotinia sclerotiorum and Botrytis cinerea. Sci. Rep. 2018, 8, 7320. [Google Scholar] [CrossRef]
  251. Höfle, L.; Biedenkopf, D.; Werner, B.T.; Shrestha, A.; Jelonek, L.; Koch, A. Study on the efficiency of dsRNAs with increasing length in RNA-based silencing of the Fusarium CYP51 genes. RNA Biol. 2020, 17, 463–473. [Google Scholar] [CrossRef]
  252. Werner, B.T.; Gaffar, F.Y.; Schuemann, J.; Biedenkopf, D.; Koch, A.M. RNA-Spray-Mediated Silencing of Fusarium graminearum AGO and DCL Genes Improve Barley Disease Resistance. Front. Plant Sci. 2020, 11, 476. [Google Scholar] [CrossRef]
  253. Koch, A.; Höfle, L.; Werner, B.T.; Imani, J.; Schmidt, A.; Jelonek, L.; Kogel, K.H. SIGS vs HIGS: A study on the efficacy of two dsRNA delivery strategies to silence Fusarium FgCYP51 genes in infected host and non-host plants. Mol. Plant Pathol. 2019, 20, 1636–1644. [Google Scholar] [CrossRef]
  254. Yin, C.; Zhu, H.; Jiang, Y.; Shan, Y.; Gong, L. Silencing Dicer-Like Genes Reduces Virulence and sRNA Generation in Penicillium italicum, the Cause of Citrus Blue Mold. Cells 2020, 9, 363. [Google Scholar] [CrossRef]
  255. Ruiz-Jiménez, L.; Polonio, Á.; Vielba-Fernández, A.; Pérez-García, A.; Fernández-Ortuño, D. Gene Mining for Conserved, Non-Annotated Proteins of Podosphaera xanthii Identifies Novel Target Candidates for Controlling Powdery Mildews by Spray-Induced Gene Silencing. J. Fungi 2021, 7, 735. [Google Scholar] [CrossRef]
  256. Wang, M.; Weiberg, A.; Lin, F.M.; Thomma, B.P.; Huang, H.D.; Jin, H. Bidirectional cross-kingdom RNAi and fungal uptake of external RNAs confer plant protection. Nat. Plants 2016, 2, 16151. [Google Scholar] [CrossRef]
  257. Nerva, L.; Sandrini, M.; Gambino, G.; Chitarra, W. Double-Stranded RNAs (dsRNAs) as a Sustainable Tool against Gray Mold (Botrytis cinerea) in Grapevine: Effectiveness of Different Application Methods in an Open-Air Environment. Biomolecules 2020, 10, 200. [Google Scholar] [CrossRef]
  258. Sarkar, A.; Roy-Barman, S. Spray-Induced Silencing of Pathogenicity Gene MoDES1 via Exogenous Double-Stranded RNA Can Confer Partial Resistance Against Fungal Blast in Rice. Front. Plant Sci. 2021, 12, 733129. [Google Scholar] [CrossRef] [PubMed]
  259. Spada, M.; Pugliesi, C.; Fambrini, M.; Pecchia, S. Silencing of the Slt2-Type MAP Kinase Bmp3 in Botrytis cinerea by Application of Exogenous dsRNA Affects Fungal Growth and Virulence on Lactuca sativa. Int. J. Mol. Sci. 2021, 22, 5362. [Google Scholar] [CrossRef] [PubMed]
  260. Islam, M.T.; Davis, Z.; Chen, L.; Englaender, J.; Zomorodi, S.; Frank, J.; Bartlett, K.; Somers, E.; Carballo, S.M.; Kester, M.; et al. Minicell-based fungal RNAi delivery for sustainable crop protection. Microb. Biotechnol. 2021, 14, 1847–1856. [Google Scholar] [CrossRef] [PubMed]
  261. Song, Y.; Thomma, B. Host-induced gene silencing compromises Verticillium wilt in tomato and Arabidopsis. Mol. Plant Pathol. 2018, 19, 77–89. [Google Scholar] [CrossRef] [PubMed]
  262. Song, X.S.; Gu, K.X.; Duan, X.X.; Xiao, X.M.; Hou, Y.P.; Duan, Y.B.; Wang, J.X.; Zhou, M.G. A myosin5 dsRNA that reduces the fungicide resistance and pathogenicity of Fusarium asiaticum. Pestic. Biochem. Physiol. 2018, 150, 1–9. [Google Scholar] [CrossRef]
  263. Yang, P.; Yi, S.-Y.; Nian, J.-N.; Qingsong, Y.; He, W.; Zhang, J.-B.; Liao, Y.-C. Application of Double-Strand RNAs Targeting Chitin Synthase, Glucan Synthase, and Protein Kinase Reduces Fusarium graminearum Spreading in Wheat. Front. Microbiol. 2021, 12, 660976. [Google Scholar] [CrossRef]
  264. Tretiakova, P.; Voegele, R.T.; Soloviev, A.; Link, T.I. Successful Silencing of the Mycotoxin Synthesis Gene TRI5 in Fusarium culmorum and Observation of Reduced Virulence in VIGS and SIGS Experiments. Genes 2022, 13, 395. [Google Scholar] [CrossRef]
  265. Nunes, C.C.; Dean, R.A. Host-induced gene silencing: A tool for understanding fungal host interaction and for developing novel disease control strategies. Mol. Plant Pathol. 2012, 13, 519–529. [Google Scholar] [CrossRef]
  266. Williamson, V.M.; Kumar, A. Nematode resistance in plants: The battle underground. Trends Genet. 2006, 22, 396–403. [Google Scholar] [CrossRef]
  267. Huang, G.; Allen, R.; Davis, E.L.; Baum, T.J.; Hussey, R.S. Engineering broad root-knot resistance in transgenic plants by RNAi silencing of a conserved and essential root-knot nematode parasitism gene. Proc. Natl. Acad. Sci. USA 2006, 103, 14302–14306. [Google Scholar] [CrossRef]
  268. Biedenkopf, D.; Will, T.; Knauer, T.; Jelonek, L.; Furch, A.C.U.; Busche, T.; Koch, A. Systemic spreading of exogenous applied RNA biopesticides in the crop plant Hordeum vulgare. ExRNA 2020, 2, 12. [Google Scholar] [CrossRef]
  269. Mitter, N.; Worrall, E.A.; Robinson, K.E.; Li, P.; Jain, R.G.; Taochy, C.; Fletcher, S.J.; Carroll, B.J.; Lu, G.Q.; Xu, Z.P. Clay nanosheets for topical delivery of RNAi for sustained protection against plant viruses. Nat. Plants 2017, 3, 16207. [Google Scholar] [CrossRef] [PubMed]
  270. Zhang, K.; Wei, J.; Huff Hartz, K.E.; Lydy, M.J.; Moon, T.S.; Sander, M.; Parker, K.M. Analysis of RNA Interference (RNAi) Biopesticides: Double-Stranded RNA (dsRNA) Extraction from Agricultural Soils and Quantification by RT-qPCR. Environ. Sci. Technol. 2020, 54, 4893–4902. [Google Scholar] [CrossRef] [PubMed]
  271. Abdelfattah, A.; Wisniewski, M.; Droby, S.; Schena, L. Spatial and compositional variation in the fungal communities of organic and conventionally grown apple fruit at the consumer point-of-purchase. Hortic. Res. 2016, 3, 16047. [Google Scholar] [CrossRef]
  272. Jackson, A.L.; Bartz, S.R.; Schelter, J.; Kobayashi, S.V.; Burchard, J.; Mao, M.; Li, B.; Cavet, G.; Linsley, P.S. Expression profiling reveals off-target gene regulation by RNAi. Nat. Biotechnol. 2003, 21, 635–637. [Google Scholar] [CrossRef]
  273. Neumeier, J.; Meister, G. siRNA Specificity: RNAi Mechanisms and Strategies to Reduce Off-Target Effects. Front. Plant Sci. 2020, 11, 526455. [Google Scholar] [CrossRef]
  274. Percival, G.; Noviss, K.; Haynes, I. Field evaluation of systemic inducing resistance chemicals at different growth stages for the control of apple (Venturia inaequalis) and pear (Venturia pirina) scab. Crop Prot. 2009, 28, 629–633. [Google Scholar] [CrossRef]
  275. Zaker, M. Natural Plant Products as Eco-friendly Fungicides for Plant Diseases Control—A Review. Agric. 2016, 14, 134. [Google Scholar] [CrossRef]
  276. Balba, H. Review of strobilurin fungicide chemicals. J. Environ. Sci. Health. Part B Pestic. Food Contam. Agric. Wastes 2007, 42, 441–451. [Google Scholar] [CrossRef]
  277. Raveau, R.; Fontaine, J.; Lounès-Hadj Sahraoui, A. Essential Oils as Potential Alternative Biocontrol Products against Plant Pathogens and Weeds: A Review. Foods 2020, 9, 365. [Google Scholar] [CrossRef] [PubMed]
  278. López, M.D.; Cantó-Tejero, M.; Pascual-Villalobos, M.J. New Insights into biopesticides: Solid and liquid formulations of essential oils and derivatives. Front. Agron. 2021, 3, 763530. [Google Scholar] [CrossRef]
  279. Llorente, I.; Vilardell, P.; Bugiani, R.; Gherardi, I.; Montesinos, E. Evaluation of BSPcast Disease Warning System in Reduced Fungicide Use Programs for Management of Brown Spot of Pear. Plant Dis. 2000, 84, 631–637. [Google Scholar] [CrossRef] [PubMed]
  280. Holb, I.J. Timing of first and final sprays against apple scab combined with leaf removal and pruning in organic apple production. Crop Prot. 2008, 27, 814–822. [Google Scholar] [CrossRef]
  281. de Kraker, J.; van den Ende, J.E.; Rossing, W.A.H. Control strategies with reduced fungicide input for Botrytis leaf blight in lily—A simulation analysis. Crop Prot. 2005, 24, 157–165. [Google Scholar] [CrossRef]
  282. Mertely, J.C.; MacKenzie, S.J.; Legard, D.E. Timing of Fungicide Applications for Botrytis cinerea Based on Development Stage of Strawberry Flowers and Fruit. Plant Dis. 2002, 86, 1019–1024. [Google Scholar] [CrossRef]
  283. Madden, L.; Pennypacker, S.; MacNab, A. FAST, a forecast system for Alternaria solani on tomato. Phytopathology 1978, 68, 1354–1358. [Google Scholar] [CrossRef]
  284. Dhar, N.; Mamo, B.E.; Subbarao, K.V.; Koike, S.T.; Fox, A.; Anchieta, A.; Klosterman, S.J. Measurements of Aerial Spore Load by qPCR Facilitates Lettuce Downy Mildew Risk Advisement. Plant Dis. 2020, 104, 82–93. [Google Scholar] [CrossRef]
  285. Thiessen, L.D.; Keune, J.A.; Neill, T.M.; Turechek, W.W.; Grove, G.G.; Mahaffee, W.F. Development of a grower-conducted inoculum detection assay for management of grape powdery mildew. Plant Pathol. 2016, 65, 238–249. [Google Scholar] [CrossRef]
  286. Everett, K.R.; Pushparajah, I.P.S.; Timudo, O.E.; Ah Chee, A.; Scheper, R.W.A.; Shaw, P.W.; Spiers, T.M.; Taylor, J.T.; Wallis, D.R.; Wood, P.N. Infection criteria, inoculum sources and splash dispersal pattern of Colletotrichum acutatum causing bitter rot of apple in New Zealand. Eur. J. Plant Pathol. 2018, 152, 367–383. [Google Scholar] [CrossRef]
  287. Ons, L.; Bylemans, D.; Thevissen, K.; Cammue, B.P.A. Combining Biocontrol Agents with Chemical Fungicides for Integrated Plant Fungal Disease Control. Microorganisms 2020, 8, 1930. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Methods for controlling fungal plant pathogens. The potential advantages (green) and disadvantages (red) inherent in conventional and alternative methods of disease control are also shown.
Figure 1. Methods for controlling fungal plant pathogens. The potential advantages (green) and disadvantages (red) inherent in conventional and alternative methods of disease control are also shown.
Plants 12 03822 g001
Table 1. Examples of the control of fungal pathogens by plant defense elicitors in controlled settings.
Table 1. Examples of the control of fungal pathogens by plant defense elicitors in controlled settings.
CropPathogenReference
AppleGlomerella cingulate[100]
ArabidopsisAlternaria alternata, Botrytis Cinerea, Colletotrichum brassicola[115,116]
BroccoliAlternaria brassicola[117]
CucumberAlternaria cucumerinum, Botrytis Cinerea, Colletotrichum lagenarium, Didymella applanta, Fusarium oxysporum[118,119]
CarrotAlternaria radicina, Botrytis Cinerea[120]
TomatoAlternaria solanii, Botrytis Cinerea, Verticillium dahliae[108,121,122,123]
StrawberryColletotrichum acutatum[124]
CassavaColletotrichum gloesporoides[125]
SoybeanCorynespora cassiicola, Fusarium oxysporum[124,126]
False Brome GrassFusarium graminearum[105]
GooseberryFusarium oxysporum[127]
WatermelonFusarium oxysporum[128]
BlackberryFusarium oxysporum[129]
PotatoFusarium solani[130]
SunflowerGolovinomyces chichoracearum[131]
Wild RocketPletcosphaerella cucumerina[132]
CourgettePodosphaera xanthii[106]
WheatPuccinia triticana, Zymoseptoria tritici[133,134]
Japonica RiceRhizoctonia solani[135]
Hybrid Tea RoseSphaerotheca pannoca[136]
Sweet PepperAlternaria solanii[122]
Table 2. Examples of the biological control of fungal pathogens in controlled settings.
Table 2. Examples of the biological control of fungal pathogens in controlled settings.
MethodCropPathogenReferences
Biochemical FungicideAstragalusAlternaria solani[189]
BananaColletotrichum musae[190]
BarleyBipolaris sorokinia[154]
BasilFusarium moniliforme, Fusarium oxysporum, Fusarium solani[191]
BeanFusarium oxysporum[192]
CarrotAlternaria radicina, Botrytis Cinerea[120]
CottonMacrophomina phasesolina, Fusarium fujikuroi, Rhizoctonia solani[193]
CucumberPodosphaera xanthii, Podosphaera fusca[194,195,196]
Hybrid Tea RoseSphaerotheca pannoca[136]
OnionAlternaria porri[197]
OrangePenicillum digitatum[198]
RapeseedSclerotia sclerotiorum[199]
RiceRhizoctonia solani[199,200]
StrawberryBotrytis Cinerea[201]
SunflowerGolovinomyces chichoracearum[131]
TomatoAspergillus sp., Fusarium sp., Fusarium oxysporum, Botrytis Cinerea, Alternaria solani[202,203,204,205,206]
WasabiErysiphe cruciferarum[207]
WheatPuccinia triticana[133]
Biological AgentBarleyBipolaris sorokinia[154]
BeanRhizoctonia solani[208]
BlackberryFusarium oxysporum[129]
CottonNigrospora oryzae[209]
Hybrid Tea RoseSphaerotheca pannoca[136]
PeanutSclerotium rolfsii[210]
PearBotryosphaeria dothidea[211]
RapeseedBotrytis cinerea, Sclerotinia minor[212,213]
RicePyricularia oryzae[214]
SoybeanRhizoctonia solani[208]
SpinachColletotrichum coccodes, Colletotrichum truncatum, Colletrichum spinaciae, Myrothecium verrucaria[215]
StrawberryBotrytis cinerea[157]
Sugar BeetSclerotium rolfsii[216]
TobaccoBotrytis cinerea[175]
TomatoAlternaria solani, Aspergillus sp., Fusarium sp., Fusarium oxysporum, Botrytis cinerea, Cladosporium fulvum[202,217,218,219,220,221,222,223]
WasabiErysiphe cruciferarum[207]
Wild RocketPletcosphaerella cucumerina[132]
Table 3. Examples of RNAi based control of fungal pathogens in controlled settings.
Table 3. Examples of RNAi based control of fungal pathogens in controlled settings.
MethodCropPathogenReferences
HIGSArabidopsisFusarium graminearum, Sclerotinia sclerotiorum[235,236,237]
BarleyBlumeria graminis[238]
BenthiRhizoctonia solani, Verticillium Dahliae[239,240]
PotatoBotrytis cinerea[241]
RiceMagnaporthe oryzae[242,243,244]
SoybeanPhakopsora pachyrhizi, Sclerotinia sclerotiorum[245,246]
TobaccoCiboria shiraiana[247]
Wheatpuccinia striiformis[243,248,249]
SIGSArabidopsisSclerotinia sclerotiorum[250]
BarleyFusarium graminearum[250,251,252,253]
CitrusPenicillium italicum[251,252,253,254]
CucurbitPodosphaera xanthii[254,255]
GrapeBotrytis cinerea[255,256,257]
LettuceBotrytis cinerea[256,257]
OnionBotrytis cinerea[256]
RapeseedBotrytis cinerea, Sclerotinia sclerotiorum[250,252]
RiceMagnaporthe oryzae[244,258]
RoseBotrytis cinerea[244,256,258]
SativaBotrytis cinerea[259]
SoybeanPhakopsora pachyrhizi[245]
StrawberryBotrytis cinerea, Botryitis fuckeliana[245,256,260]
TomatoBotrytis cinerea[256,260]
WheatFusarium asiaticum, Fusarium culmorum, Fusarium graminearum[261,262,263,264]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

McLaughlin, M.S.; Roy, M.; Abbasi, P.A.; Carisse, O.; Yurgel, S.N.; Ali, S. Why Do We Need Alternative Methods for Fungal Disease Management in Plants? Plants 2023, 12, 3822. https://doi.org/10.3390/plants12223822

AMA Style

McLaughlin MS, Roy M, Abbasi PA, Carisse O, Yurgel SN, Ali S. Why Do We Need Alternative Methods for Fungal Disease Management in Plants? Plants. 2023; 12(22):3822. https://doi.org/10.3390/plants12223822

Chicago/Turabian Style

McLaughlin, Michael S., Maria Roy, Pervaiz A. Abbasi, Odile Carisse, Svetlana N. Yurgel, and Shawkat Ali. 2023. "Why Do We Need Alternative Methods for Fungal Disease Management in Plants?" Plants 12, no. 22: 3822. https://doi.org/10.3390/plants12223822

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

Article Metrics

Back to TopTop