Next Article in Journal
Estimation of Air Pollutant Emissions by Tractor Utilization in Korea
Next Article in Special Issue
The Effectiveness of Synthetic and Inorganic Substances in Different Apple Scab Control Strategies
Previous Article in Journal
An Overview of the Impacts of Climate Change on Vineyard Ecosystems in Niagara, Canada
Previous Article in Special Issue
Evaluation of Fungicides and Fungicide Application Methods to Manage Phytophthora Blight of Pigeonpea
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Fusarium Fungi Pathogens, Identification, Adverse Effects, Disease Management, and Global Food Security: A Review of the Latest Research

by
Theodora Ijeoma Ekwomadu
1,* and
Mulunda Mwanza
1,2
1
Food Security and Food Safety Niche Area, Northwest University, Private Bag X2046, Mmabatho 2735, South Africa
2
Department of Animal Health, Faculty of Natural and Agriculture Sciences, Northwest University, Private Bag X2046, Mmabatho 2735, South Africa
*
Author to whom correspondence should be addressed.
Agriculture 2023, 13(9), 1810; https://doi.org/10.3390/agriculture13091810
Submission received: 21 August 2023 / Revised: 9 September 2023 / Accepted: 11 September 2023 / Published: 14 September 2023
(This article belongs to the Special Issue Integrated Management of Fungal Diseases in Crops)

Abstract

:
Fusarium pathogens are ubiquitous and mainly associated with diseases in plants. They are the subject of great economic concern in agriculture due to crop losses to contamination of cereal grains with mycotoxins. Fusarium species are also considered agents of human and animal mycotic infections, having a wide-ranging spectrum of clinical manifestations in immunocompromised patients. Fusarium phytopathogens infect a wide variety of plants and cause symptoms ranging from stunted growth, fruit or seed decay, yellowing, and wilting of the leaves and cankers to root or stem decay. The identification of these fungi is difficult due to their pleomorphic tendency and the presence of both homothallic and heterothallic strains in the same species, and so is identifying them at species level because of variation among isolates. However, molecular tools have so far been very powerful in species identification and phylogeny, as the great diversity of the Fusarium genus has compelled scientists to continuously revise previous taxons. Mostly, Fusarium diseases are difficult to control, as fungi easily overcome host resistance to various methods of control. We present an overview of the recent research on Fusarium fungi, its adverse effects, and its impacts on food security. We further elucidate various methods of identifying them to encourage much-needed research on integrated management of this unavoidable food contaminant to achieve sustainable global food security.

1. Introduction

More than 70% of plant diseases can be traced to fungi or fungus-like pathogens that threaten food security [1]. In addition, many fungal pathogens including Fusarium spp. also produce mycotoxins, that further threaten human and livestock health [2,3]. Fusarium spp. are also important to food safety in crop production and especially to the production of cereals. The genus Fusarium is one of the most economically important fungi, containing many agronomically important plant pathogens, mycotoxin producers, and opportunistic human pathogens [4]. This large and diverse fungi family breeds these mycotoxins mostly in the field before harvest. Fusarium species adapt to a variety of habitats and, although they have a notable affinity for moderate climates, they contaminate crops all around the world [5]. Consumption of mycotoxin-contaminated grain by humans or animals can cause acute or chronic illness and, in some cases, death. The most toxic and prevalent Fusarium toxins of economic importance include trichothecenes, fumonisins, and zearalenone.
Many Fusarium species are pathogenic to several important crops; two of them, namely, F. graminearum and F. oxysporum, are listed among the ten most important fungal pathogens in plant pathology [6]. Possible disease symptoms caused by members of Fusarium include root or stem rots, cankers, vascular wilts, fruit or seed rots, and leaf diseases. Most fusarium species can cause a wide range of plant diseases that affect many crops, including major food and cash crops such as wheat, barley, maize, bananas, and cotton, often with devastating socio-economic impact [7].
Cereal crops provide a major food source for humans and animals, yet they are heavily attacked by fungal pathogens. These diseases are often insidious, without obvious symptoms at first, and are extremely difficult to control [7]. Their success as plant pathogens can be attributed to wide host ranges, endophytic infection, and varied modes of survival and dispersal. Representatives occur in virtually all bioclimatic regions of the world in agricultural and natural ecosystems [7]. Also, the emergence of novel fungal pathogens can threaten food production and/or ecological environments. Hence, the successful control of fungal diseases is challenging, as fungi can easily overcome host resistance [8]. Biological control of fungal diseases is one such approach that offers much toward increasing world crop production.

2. Fusarium: Overview and Taxonomy

The taxonomy of Fusarium started in 1809 when the genus Fusarium was first described by Link [9]. It comprises naturally ubiquitous species [10,11]. Fusarium is a large group of filamentous fungi that occur predominantly in the air and soil and which usually associate with plants and occasionally with humans. Some of the most important plant pathogenic fungal species known today are members of this genus [12]. Worldwide, it is a concern that a large number of economically important plant species are susceptible to at least one or more Fusarium spp. [12]. Fungi now included in the genus Fusarium were originally described and defined as Fusisporium based on the type Fusisporium Roseum described by Link in 1809 [13]. Wollenweber and Reinking [14] reclassified the two F. roseum type specimens as F. sambucinum and F. graminearum and currently accept F. sambucinum as the type species for the genus. Although the taxonomy of Fusarium continues to undergo major changes, mainly based on molecular classifications, the Wollenweber and Reinking classification system continues to form the foundation on which species are described [12].
Members of the genus Fusarium are characterized by having septate, hyaline, delicately curved, and elongate macroconidia [12,15]. Mycelia and spore masses are generally brightly colored [16]. In some species, smaller, 0- to 1-septate microconidia and chlamydospores are common, while some authors recognize a third conidial type known as mesoconidium. The Fusarium genus comprises around 70 recognized species, identified using a polyphasic approach, and about 300 putative species. Following phylogenetic species concepts, many putative species do not yet have formal names [17].

3. Identification of Phytopathogenic Fusarium Fungal Species

3.1. Morphological Characters for Identifying Phytopathogenic Fusarium Species

Morphological characters are considered the most traditional criteria used to identify any fungal species. Fusarium produces a range of mycelia that are cottony with shades of pink, yellow, and purple. Some species produce either macroconidia or microconidia as asexual reproductive structures, while other species produce both macroconidia and microconidia [18]. The morphology of microscopic characteristics, i.e., the general shape and dimensions of the macroconidia, the generation of microconidia, chlamydospores, sclerotia, sexual stages, and pigmentation, are the primary means used for the identification of Fusarium species. Members of the genus are variable in cultural characteristics because changes in the environment in which they grow can result in morphological changes both in culture and in conidia [15]. However, the description of these characteristics is necessary under very specific environmental conditions because of the distinctive cultural variability of morphological traits.

3.1.1. Universally Found Characters

The genus Fusarium is categorized by the production of septate, hyaline, delicately curved, elongate macroconidia and chlamydospores along with other secondary characteristics like mycelial growth and pigmentation [15].
(i)
The macroconidium is the single most important cultural characteristic for the identification of a culture of Fusarium species. The most distinctive characteristic of the macroconidia is its shape, followed by the size and number of septa, and finally, the nature of apical, basal, or foot cells [12]. Regarding shape, most Fusarium produce sickle-shaped macroconidia that can be characterized into three types: (1) straight macroconidia, which can appear virtually needle-like if they are thin, e.g., F. avenacum; (2) microconidia having dorsivental curvature along all or a portion of the spore (these spores are almost of the same width along their entire length, e.g., F. equiseti); and (3) microconidia in which the dorsal side is more curved than the ventral side, e.g., F. crookwellence (Figure 1). Macroconidia can be long (F. armeniacum) or short (F. culmorum), but usually spore size is a relatively constant character and major variations indicate improper culture conditions. Typically, Fusarium macroconidia are 3–5-septate. The number of septa ought to be determined depending on the range and the average number of septa per spore [12].
Another important macroconidium characteristic is the apical and basal cell forms. There are four common forms of apical cells: (1) blunt, e.g., F. culmorum; (2) papillate, e.g., F. sambucinum; (3) hooked, e.g., F. lateritium; and (4) Tapering, e.g., F. equiseti (Figure 1).
The apical cell length also can vary amongst species, but it is usually constant within a species. The main diagnostic features of apical cells are the degree of curvature, relative length, and general form. Microconidia are not produced by all Fusarium species; therefore, their presence is a potential diagnostic characteristic for Fusarium identification. The major characteristics regarding microconidia are the microconidia shape, the endogenous cells where they are born, and their arrangement on or around the conidiogenous cell [12].
(ii)
Chlamydospores are verrucose (rough) or smooth-walled structures produced individually, e.g., F. solani; as doubles or pairs, e.g., F. compactum; as clumps, e.g., F. scirpi; or as chains, e.g., F. compactum. Chlamydospores are produced rarely and take a longer time (more than 6 weeks) when compared to macro- or microconidia. The presence of chlamydospores in the aerial mycelia or embedded on the agar surface is another important criterion used in the identification of Fusarium species [12].

3.1.2. Other Important Characteristics

The characteristics discussed above are universally found in almost all Fusarium species. Some other characteristics are restricted to only a few species of Fusarium, which serve as an important delimiting factor in their identification; for example, mesoconidia teleomorphism and other characteristics like circulated (coiled) hyphae in the case of F. circinatum. In addition, the formation of sclerotia-like structures (compact masses of hardened mycelium with stored reserves of food material), etc., are some other relevant characteristics that help in the primary identification process [12].
However, the most important and diagnostically relevant secondary characteristic is pigmentation [12]. The different Fusarium species produce colors ranging from yellow to orange to carmine red [19]. The pattern of pigmentation is detectable on PDA and a 12:12 h light:dark cycle is usually preferred. Pigments produced by these fungi may be sensitive to light or pH and may be diffusible or non-diffusible into the growth media, and most of the evaluations are carried out a week after incubation. Another important characteristic is the growth rate of the species, usually measured as colony diameter from PDA plates incubated with a single spore culture and incubated at 25 or 30 degrees for 3 days. There are slow-growing species like F. lateritium and F. merismoides and fast-growing species like F. culmorum, F. graminearum, etc. [12]. These characteristics, if not properly analyzed, may not be clear and are not usually preferred for the identification of species. Furthermore, morphological identification is time-consuming and could easily result in misidentifications, especially for phylogenetically related species.
Secondary metabolites and mycotoxins are also characteristic features, which may influence a particular order in the culture and serve as specific secondary characteristics. The chemical background of the metabolites or mycotoxins can be used to primarily group the fungi, which can be further analyzed to finally assign the fungi to particular species. Fusarium is known to produce many toxins, which can be effectively used for their specific identification (Table 1).

3.2. Molecular Tools for Identifying Phytopathogenic Fusarium Species Based on Genetic Diversity

Although morphological identification alone might be problematic, it is still helpful in practice and frequently used in combination with molecular methods. Different molecular tools, for example, PCR-based techniques, aid in the description of differences amongst species according to genetic diversity. Some examples such as random amplified polymorphic DNA (RAPD) [20], restriction fragment length polymorphism (RFLP) [21], amplified fragment length polymorphism (AFLP) [22,23], DNA sequences of intergenic spacers (IGS) [24], β-tubulin (tub2) [25], translation elongation factor-1 alpha (TEF-1α) [26], and internal transcribed spacers (ITS) [27] have been used to differentiate and diagnose fungal strains. Analyses of other genes such as calmodulin, topoisomerase II and cell biohydrolase-C have also been used for the identification of Fusarium [28,29]. In addition, the MALDI-TOF MS technique is also used in agriculture and food safety for the identification of Fusarium species (Wigmann et al., 2019) [30].
(i)
Random Amplified Polymorphic DNA (RAPD)
This is a PCR technique where primers (usually 10-20 base-pair (bp) in length) randomly bind to complementary sequences of the genomic DNA of a given organism, which leads to the generation of consensus sequence patterns that serve as fingerprints for the organism [31]. This technique works in such a way that nucleotide sequence variation due to insertions, additions, or base substitutions, inversion of priming site, conformational changes in the template DNA, etc., in the PCR priming regions, especially at the 3′ ends, prevent primer annealing. This results in different-sized PCR fragments that are highly specific for a particular species [32]. The RAPD assays have been effectively used for genome analyses of different bacteria and fungi [33,34]. In addition, RAPD has been used for the identification of other Fusarium species such as F. oxysporum, F. avenaceum, F. poae, F. solani, and F. moniliforme [35,36,37,38]. Despite the advantages of RAPD, it has been criticized due to poor reproducibility of results, affecting its use in fungal taxonomy, and there is a need for fastidious PCR conditions [39,40].
(ii)
Restriction fragment length polymorphism (RFLP)
This technique is based on restriction enzyme digestion of the pathogen DNA, and afterward, separation of the fragments by electrophoresis in agarose or polyacrylamide gels to identify differences in the sizes of DNA fragments [41]. Polymorphisms within the restriction enzyme cleavage sites are meant to differentiate fungal species. Although the DNA restriction profile can be directly noticed by staining the gels, Southern blot analysis is usually needed. The DNA is transferred to appropriate membranes and hybridized with an appropriate probe [40]. RFLPs have been largely used for the study of the diversity of mycorrhizal and soil fungal populations/communities [21,42,43]. Although meant for the differentiation of pathogenic fungi [44], this early technique has been increasingly supplanted by other fingerprint techniques based on PCR. RFLP combines the amplification of a target region with the further digestion of the PCR products obtained.
(iii)
Amplified fragment length polymorphism (AFLP)
The AFLP technology [45] is a modified version of RAPD, which is based on the use of restriction enzymes to digest total genomic DNA followed by ligation of restriction half site-specific adaptors to all restriction fragments. Then, selective amplification of these restriction fragments is achieved with PCR primers that have in their 3′ end the corresponding adaptor sequence and selective bases. The bands of the amplified fragments are visualized on denaturing polyacrylamide gels. The AFLP technology can amplify between 50 and 100 fragments at one time and detect various polymorphisms in different genomic regions simultaneously. It is also very sensitive and reproducible. The disadvantages of AFLPs are that they need high-molecular-weight DNA and more technical expertise than RAPDs (ligations, restriction enzyme digestions, and polyacrylamide gels) and that AFLP analyses suffer the same analytical limitations as RAPDs [46]. AFLP has been used to differentiate fungal isolates at several taxonomic levels, e.g., to differentiate Monilinia laxa that infect apple trees from isolates infecting other host plants [47] and to separate non-pathogenic strains of Fusarium oxysporum from those of F. commune [48]. AFLP profiles have also been widely used for the phylogenetic analysis of the Fusarium oxysporum complex [49,50,51]. Leissner et al. [22] also used AFLP to differentiate between isolates of F. graminearum.
(iv)
Inter-genic Spacers (IGS)
These are regions that separate nuclear ribosomal DNA repeat units, which consist of highly conserved genes and more variable spacer regions [52]. The number of ribosomal DNA repeat units varies among different species, and this results in variations in the length and restriction sites of IGS [53]. IGS-RFLP has been used for the analysis of genetic variation within and between closely related species or communities [54,55]. Analysis of the IGS region with the RFLP technique has been effectively used for phylogenetic analysis of closely related species of Fusarium, such as F. langsethiae vs. F. sporotrichioides and F. poae vs. F. kyushuense. The result showed clear differentiation between the two species [24].
(v)
β-tubulin
The β-tubulin gene sequences have been widely used for phylogenetic investigations in various fungi [56,57]. It has also been used in the phylogenetic investigations of F. xylarioides [58]. Schmidt et al. [23] also used DNA sequences of β-tubulin along with other marker genes for the taxonomic study of F. langsethiae, F. poae, and F. sporotrichioides.
(vi)
Translation elongation factors
TEF-1a has been widely used for Fusarium classification because it is highly informative at the species level in Fusarium [58]. Also, universal primers have been designed that work across the genus [58] to amplify a ~700 bp region of TEF, including three introns. These introns cover over half of the amplicon’s length in all known Fusarium species [59]. The TEF gene is a single copy in Fusarium and its sequence shows high variability among closely related species.
(vii)
Internal Transcribed Spacers (ITS)
Internal Transcribed Spacer (ITS) regions are useful tools for the identification of different fungal species [60]. Internal Transcribed Spacers ITS1 and ITS2 undergo more variations even within closely related species and hence are widely used for identification processes and for studying evolutionary events. The highly conserved priming site of the ITS region makes it easy to be amplified from practically all fungal species. The stretches of DNA between 18S, 5.8S, and 28S rRNA regions make up the ITS regions [27]. The growing ITS sequence data is also an added advantage that helps in the identification of various fungi. This information can also be used for the development of species-specific primers for the detection of some fungi in a much-reduced time instead of via the morphology method [29]. For the identification of Fusarium species, ITS-RFLPs has been extensively used [61]. Variations occurring in ITS1 and ITS2 sequences have been used to study the genetic relationship between different Fusarium species [61].
The ITS region is the perfect region for species identification since even closely related species have sequence variations [27]. In addition, these variable regions are flanked by conserved ribosomal RNAs, which provide the option to use primers for PCR amplification of the variable regions that are recognized by the majority of fungi and fungal-like organisms. For fungal identification, most people use the ITS1/ITS4 primer pair [27] for amplification of the ITS region, which leads to another fact in favor of ITS as a barcode marker: there are huge databases consisting of ITS sequences from the majority of known fungi. Though perfect for species identification, the ITS region is too variable to determine the phylogeny of higher ranks.
This region contains highly conserved areas adequate for genera- or species-consensus primer designing (RNA ribosomal genes), alternate with highly variable areas that allow discrimination over a wide range of taxonomic levels (ITS region) [27]. The ITS region is ubiquitous and found in all eukaryotes. In addition, the high copy numbers of rRNA genes in the fungal genome enable a highly sensitive PCR amplification. Furthermore, large numbers of ribosomal sequences are publicly available in databases, facilitating the validation and reliability of the detection assays. Traditionally, molecular identification of plant pathogenic fungi is accomplished by PCR amplification of the ITS region followed by either restriction analysis [62] or direct sequencing and BLAST searching against GenBank or other databases [27].
(viii)
The matrix-assisted laser desorption/ionization time-of-flight mass spectrometry
This technique can also be applied for the identification and differentiation of fusarium species, e.g., F. fujikuroi and F. solani. Besides the medically important fungi, reliable identification at the species level using MALDI-TOF MS has been reported for environmental fungi and other food-spoilage fungi [63,64]. Unlike the PCR-based methods, this technique is an advanced tool for rapid and accurate identification of fungi and other microbes using either intact cells or cell extracts. It is based on the molecular mass spectral readout from an ionized protein mixture. The cell culture assay is carried out based on the mass spectral pattern, which can be taken as the unique fingerprint to identify fungi among closely related species [65].

4. The Pathogen—Fusarium

The genus Fusarium comprises numerous toxigenic species that are pathogenic to plants and/or humans. They are capable of colonizing various environments on earth [17]. Fusarium species as versatile fungi are found everywhere, such as in air, water, soil, plants, and on organic substrates. Fusarium’s widespread distribution is attributable to its ability to withstand a wide range of conditions and grow on a broad range of substrates, as well as their efficient mechanisms for dispersal [66]. Often regarded as soil-borne fungi, since they are abundant in soil and frequently associated with plant roots, they are also present in water as parts of water biofilms [67]. Fusarium species have been isolated from public swimming pools, shower drainage pipes, and hospital water systems [68].
These field fungi require high moisture levels to colonize and contaminate grain [69,70]. Aside from their ability to act as plant pathogens, Fusarium species have been linked to a wide range of diseases and infections, directly or indirectly in humans and animals [71]. Fusarium is one of the most economically important fungal genera because of yield loss due to plant pathogenic activity. Mycotoxin contamination of food and feed products, which often renders them unacceptable for marketing, also adds to the huge economic losses of the agricultural industry and poses a great threat to human and animal health due to consumption of mycotoxins [71,72,73]. Management of plant Fusarium diseases is difficult as they are both seed-borne and soil-borne, and many strains have been reported as etiologic instruments of infections in humans [74]. It is therefore important to gain insight into the processes involved in the development of diseases in the host of this transkingdom pathogen.

4.1. Fusarium as a Plant Pathogen

Fusarium is known to contain a range of plant pathogenic fungal species, with a report of devastating infections (Table 2) and has been in existence for the past two centuries [12]. As common invaders of aerial plant parts, they can either be part of the normal mycoflora or act as plant pathogens on horticultural crops and cereal grains, such as maize, where they render them unfit for consumption [75,76]. Fusarium spp. can cause seedling, root, and crown rot as well as stalk and ear rot at any stage of plant development [77,78,79]. For fusarium infection of plants to be successful, different highly regulated processes are involved, starting from initial infection to development of disease in the host [80].
(i)
Adhesion
Fungal infection begins after the recognition of roots through unknown host signals, followed by infectious hyphae adhering to the host root surface [81]. This adhesion process of fungi to the host surface is not specific, as they may adhere to the surface of either hosts or non-hosts [82]. Site-specific binding may be vital in the attachment of the propagules at the root surface, after which other processes required for colonization can continue [83].
(ii)
Entry
During pathogenesis, the fungus penetrates the complex physical defense barriers of the host plant cell walls [84,85]. Gaining entrance to plant cells requires hydrolytic degradation of physical host barriers such as the cell-wall endodermis, whereby fungi secrete a mixture of hydrolytic enzymes including cutinases, cellulases, pectinases, and proteases [86]. This is for it to reach the vascular tissues where it lodges.
After penetration in the vascular tissues, it has to adapt to the hostile plant environment and tolerate plant antifungal compounds. The fungus tries suppressing and inactivating host defense responses, usually by secreting toxins or plant-hormone-like compounds that manipulate the plant’s physiology to the benefit of the pathogen [86]. This quite often is achieved through the production of phytotoxins with varying degrees of specificity toward different plants [87].
(iii)
Colonization, Adaptation, and Propagation
During colonization, the mycelium spreads intracellularly through the root cortex until it reaches the xylem vessels and enters them through the pits. The fungus then remains solely within the xylem vessels, using them to colonize the host [88]. Fungal colonization of the host’s vascular system is often fast and frequently facilitated by the formation of microconidia inside the xylem vessel elements [89] that are detached and taken upward in the sap stream [88]. As soon as the perforation plates stop the spores, they ultimately germinate, and germ tubes pierce the perforation plates. Subsequently, hyphae, conidiophores, and conidia are formed [89,90].
(iv)
Disease development
Wilting is usually triggered by various pathogen activities. These include a buildup of fungal mycelium in the xylem tissue and/or production of toxins; host defense responses comprising the production of gels, gums, and tyloses; and vessel crushing by multiplication of adjacent parenchyma cells [91]. The wilting symptoms seem to be a result of serious water stress, mainly due to vessel occlusion. Symptoms are somewhat variable but involve combinations of vein clearing, leaf epinasty, wilting, chlorosis, necrosis, and abscission. Harshly infected plants may wilt and die, while plants affected to a lesser extent may become stunted and unproductive [92].
In other words, the parasitic phase in the Fusarium disease development involves the fungus’ penetration of host plant roots, colonization of the root cortex and endodermis, movement to and colonization of the xylem vessels of stems/leaves, development of the symptoms, and finally, death of the host plant. Meanwhile, the saprophytic stage of Fusarium disease development involves the formation of resting structures in the dead host [93]. With fungal penetration of the host, the management of the multiplication rate of the fungus can be very challenging. During the dormant stage of the Fusarium, the germination of mycelia, chlamydospores, microconidia, and macroconidia (fungal propagules) that are present in infected soil is suppressed because of microbiostasis or mycostasis [94].
Table 2. Phytopathogenic Fusarium fungal species, host plants, and infections caused.
Table 2. Phytopathogenic Fusarium fungal species, host plants, and infections caused.
PathogenHost PlantInfectionReference
F. avenaceumWheatFusarium head blight (FHB)[95]
F. oxysporiumOriental lilium plantRoot and bulb disease[96]
F. oxysporumPotatoStem-end rot [97]
F. oxysporumBananaFusarium wilt[98]
F. oxysporiumPineappleDieback[99]
F. oxysporiumAvocadoStem-end rot[100]
F. fujikuroiRiceBakane[101]
F. graminearumOil palm Fusarium wilt[102]
F. graminearumBananaCrown rot[103]
F. graminearumWheat and barleyFusarium head blight (FHB)[104]
F. proliferatumOriental lilium plantRoot and bulb disease[96]
F. proliferatumMangoLeaf spot[105]
F. proliferatumPineappleFruitlet core rot[106]
F. proliferatumChilli pepper (Capsicum annuum L.)Fruit rot[107]
F. solaniPaprikaFusarium rot[108]
F. solaniAvocadoStem-end rot[109]
F. solaniPapayaRoot rot[110]
F. verticilloidesMaizeFusarium ear rot[111]
F. verticilloidesMangoLeaf spot[112]
F. verticilloidesBananaCrown rot[113]
F. communeChinese water plant (Eleocharis dulcis)Fusarium wilt[114]
F. equisetiPapayaFruit rot[115]
F. equisetiAvocadoStem-end rot[116]
F. sporotrichioidesAvocadoStem-end rot[117]

4.2. Fusarium as Human and Animal Pathogen

Conventionally, Fusarium has been more of an agronomic threat than a medical one, but over the last thirty years, due to a variety of contributing factors, this scenario has undergone a radical change. This made Fusarium spp. emerge as a major opportunistic human pathogen, causing an expansive range of infections with high morbidity and mortality [66,118,119]. Infections caused by Fusarium species are generally referred to as fusariosis, which is largely dependent on the immune status of the host and the route of entry of the infection [71,120]. Among immunocompetent hosts, the common Fusarium infections are keratitis and onychomycosis with other less common conditions such as sinusitis, pneumonia, thrombophlebitis, and fungemia [71].
It is not all species of the genus that possess the ability to induce disease or infection; only a few cause infections in humans and animals. Fusarium human pathogens of growing importance include F. oxysporum, F. moniliforme, and F. solani, although infections by F. proliferatum and F. napiforme have also been reported recently [66,121]. According to Sullivan and Moran, as well as Gnat et al. [122,123], the types of diseases found in animals, especially pets, are often the same as those that are found in people. Although not much is known as regards fungal pathogenesis, it involves a complex interplay of many factors [124].
The disease mechanisms of Fusarium human/animal pathogens include the following:
(i)
Adhesion
Fungal hyphae must adhere to the host surfaces both as a commensal to avoid being washed out of the various niches and during the onset of infections [125]. Fungal pathogens can adhere to host cells by way of specialized cell-wall glycoproteins.
(ii)
Entry
A major factor in the pathogenesis of invasive fusariosis involves the disruption of the mucosa or cutaneous barrier of the host cell. Fungi hardly cause disease in healthy, immunocompetent hosts, although humans are constantly exposed to infectious propagules. It is only when fungi accidentally penetrate barriers such as intact skin and mucous membrane linings, or once immunologic deficiencies and other devastating conditions occur in the host, that fungi can colonize and grow [124]. Hence, the ability of fungi to penetrate host cells is crucial for the progression of infection in the setting of intact skin or gut barriers [125]. Fusarium from soil or water enters the body, making contact with minute breaks in the skin or mucous membranes, causing infections. These sites serve these organisms as cutaneous portals of systemic entry during periods of immunosuppression, allowing for the dissemination of infection [126]. Infectious agents may also enter the body because of extensive skin breakdowns, such as in burns and wounds, where even airborne conidia may be the source [127], or due to the presence of foreign bodies, such as keratitis in contact lens wearers or peritonitis in patients receiving continuous ambulatory peritoneal dialysis (CAPD).
(iii)
Colonization, Adaptation, and Propagation
To effectively colonize the host, these organisms must be able to survive at the elevated body temperature and either evade phagocytosis, counteract the hostility they come across, or adapt in a way that will make them multiply. Several factors contribute to the infection and pathogenesis of these organisms. The ability to secrete enzymes, e.g., keratinase, their ability to grow at 37 °C, dimorphism, and other yet-undefined factors contribute to fungal pathogenesis, which involves a complex interplay of many fungal and host factors. Fungi often develop both virulence mechanisms that facilitate their multiplication within the host [124].
(iv)
Disease development, Dissemination
Propagation of fungi in the body shows a breach or paucity of host defenses. Endocrinopathies or immune conditions may cause such a breach, or it may be by iatrogenic induction [124]. Effective infection may result in disease, defined as an abnormality or interruption of the normal structure or function of body parts, organs, or systems (or combinations thereof) that is marked by a characteristic set of symptoms and signs and whose etiology, pathology, and prognosis are known or unknown [124].

5. Plant Pathologies Caused by Fusarium Species

The Fusarium species cause most plant diseases, infect all plant parts, and induce cell death. While growing on a host plant, Fusarium produces an array of toxins that are both phytotoxic and/or mycotoxic. During infection processes in plants, these toxins modulate plant physiology and favor Fusarium colonization and multiplication [128].
These toxins remain on the infected agriculture products and enter the food chain, where they cause mycotoxicosis in animals, livestock, and humans. The Fusarium genome is compartmentalized into two regions: the core genome responsible for primary metabolism and reproduction, and the adaptive genome responsible for pathogen virulence, host specialization, and probably other functions [4]. In some hosts, for instance, tomato plants, genes involved in virulence and host specialization are located on pathogenicity chromosomes within pathogenic strains [4]. Genome comparison studies revealed that the transfer of Fusarium pathogenicity chromosomes into a nonpathogen transformed the non-pathogen into a plant pathogen. Therefore, horizontal transfer may explain the polyphyletic origins of host specificity within the Fusarium genus [4].

6. Host–Pathogen Interaction in Fusarium Infections

The interactions between the plant and the fungus are diverse and complex, and can alter the physiology and morphology of both partners. Fusarium establishes the colony and destroys plant tissues by overriding the plant defense mechanisms and also by producing host-specific toxins (Figure 2). It is facilitated by the secondary metabolites produced by the fungus, for which the host plant initiates a defense response termed effector-triggered immunity or pathogen-associated molecular pattern (PAMP)-triggered immunity [129]. Penetration into the plant cell wall is achieved by the action of several cell-wall-degrading enzymes like cellulases, pectinases, lipases, and xylanases [130,131]. So, the fungal pathogenic factors which act as pathogens to plants include the following:
(i)
Host-specific toxins in Fusarium infections
Most phytopathogenic fungi produce toxins (Table 1) that can cause plant diseases [132]. Notably, many fungal toxins are low-molecular-weight secondary metabolites that show obvious symptoms such as wilting, chlorosis, necrosis, growth inhibition, and leaf spotting [133] in host plants. Fungal toxins can increase the permeability of the host plant cell membrane, cause leakage of cell electrolytes, and damage the membrane system, thereby causing it to lose normal physiological functions and potentially die [134]. Toxins can also damage the inner membrane of the chloroplasts in host plant leaves, and progressively cause the disintegration of the basal lamella, causing the chloroplasts to form vesicles, thus triggering severe poisoning effects or death to the host plants. Toxins that act on the mitochondria of the host plant cause damage to the mitochondrial membrane structure, swelling of the cristae, vacuolization, decrease in the mitochondrial matrix and cristae number, or make the cristae disappear [132].
(ii)
Effector Proteins
Phytopathogenic fungi secrete proteins that can interact with host plants during the infection process. These effector proteins, e.g., Fg62 and Avr2, play crucial roles in plant cells and therefore affect the interaction between pathogens and their hosts. Research on plant-pathogenic fungal effectors has often focused on model fungi and those that had their whole genome sequenced [135,136]. So far, Fusarium spp., e.g., Fusarium oxysporum f.sp. lycopersici are among the major cloned pathogenic fungal effectors. The virulence effects and transport molecular mechanisms of effector proteins are thus far in the early stages of research. It has not yet been identified which effector proteins can modify plant metabolism to meet the nutritional needs of the infecting fungi, or which plant signal transduction pathways control the expression of effector protein genes [132]. Through comparative analysis of fungal effector molecules, common motifs of fungal effectors may be found. Such knowledge would lead to better explanations of the interactions between pathogenic fungi and host plants and would indicate the pathogenic mechanisms of pathogenic fungi and the disease-resistance mechanisms of host plants [132].
(iii)
Cell-Wall-Degrading Enzymes
These enzymes break the cell wall and cuticle of the plant and enhance the invasion, colonization, and proliferation of pathogenic fungi [137,138]. The cell-wall-degrading enzymes of these fungi and their interaction with host plants during the infection process is increasingly being studied using molecular biology and proteomics. The main cell-wall-degrading enzymes are pectinase, chitinase, cellulase, and protease [139]. In living plant tissues, cellulases from pathogens play a role in the softening and disintegration of cell wall material [139,140]. Fusarium graminearum secretes cell-wall-degrading enzymes such as cellulase, pectinase, and xylanase during infection. The secreted enzymes cause the decomposition of plant cell-wall components and aid the penetration and growth of the pathogen in the host tissue [141]. Specifically, the cell-wall-degrading enzyme β-galactosidase can promote the degradation of lactose in the cell wall, which precedes the production of galactose and glucose and enhances fruit softening [142]. Ma et al. [143] suggested that there is an abundance of β-galactosidase in the early stages of fruit softening which contributes to the breakdown of cell-wall galactosyl bonds, preceding the reduction of cell-wall integrity. Some enzymes, including protease, hemicellulase, amylase, and phospholipase, which can degrade protein, hemicellulose, starch, and lipids, respectively, also play specific roles in the infective process [142]. Fungal infection of plants and disease production is not only caused by cell-wall-degrading enzymes, but also by hormones, toxins, and other factors. Simultaneously, pathogen infection can also activate plant defense enzyme systems and induce plants to generate antifungal substances to inhibit the pathogens’ cell-wall-degrading enzymes and achieve disease resistance. Therefore, the infection of plants by pathogenic fungal species is a complex biochemical process [132].
Fusarium spp., for instance, F. oxysporum, exhibit a high degree of host specificity [4]. A complex network comprising interconnected and overlapping signaling pathways is activated once Fusarium recognizes a host in its vicinity. These pathways include mitogen-activated protein (MAP) kinase signaling pathways [144], Ras proteins, G-protein signaling components, [145,146,147] components of the velvet complex [148], and cAMP pathways [146] (Figure 2).
Therefore, effective management of Fusarium wilt diseases will require thorough knowledge and understanding of the molecular mechanisms involved in the pathogenesis.
Figure 2. Fusarium pathogen and plant host defense mechanisms (molecular interactions). Adapted from Ma et al. [4].
Figure 2. Fusarium pathogen and plant host defense mechanisms (molecular interactions). Adapted from Ma et al. [4].
Agriculture 13 01810 g002

7. Control and Management of Phytopathogenic Fusarium spp.

Fusarium is difficult to control in crops, as methods such as the physical, chemical, and cultural ways of control are not only ineffective but also expensive. Some of the reasons why Fusarium species are widely distributed include the following: (1) they can grow and develop on a wide variety of substrates and (2) their means of spore dispersal are very efficient [68]. Fusarium is a soilborne phytopathogen; hence, it can survive in soil and plant debris for many years. The infected soil could be redistributed to different places by tools used in the fields and by animals, which can lead to outbreaks in other new areas. This fungus can survive in a wide range of environmental conditions. However, one of the less common routes for fungal transmission include insects acting as vectors for plant pathogens, and recent studies have shown that beetles can be effectual vectors for the Fusarium solani species [149]. The amount and distribution of the fungal inoculum in the field and the stage of plant growth at which the Fusarium disease occurs determine the extent of yield reduction [150]. Mostly dry and warm growing conditions favor the growth of Fusarium. This poses a great problem in parts of the world and in areas where crops are grown in such seasons [151]. One of the challenges of fusarium elimination in agricultural practice is the constant presence of Fusarium propagules on all organs of living plants, on plant residues, and in the soil [152]. A dangerous feature of Fusarium infection of plants is the possible absence of external symptoms, which does not always mean that the pathogen and mycotoxins are absent [153].
Wilts caused by Fusarium are commonly assumed to be monocyclic, which means that the disease does not spread from one plant to another during the season [154]. This is because the propagules that can disseminate the disease to other plants to cause secondary infections only form very late during the crop season. So, managing Fusarium’s dispersal and growing population is a very difficult task at the field level.
Plants are most susceptible to Fusarium infection and sporulation during the warm moist seasons and the flowering phase, but the probability of infection continues throughout the growing season [155]. Since Fusarium diseases are difficult to control, as fungi easily overcome host resistance to various methods of control, biological control of fungal diseases has increasingly received attention as one of the alternative means of control [1]. Biological control is also usually perceived as natural and thus more environmentally friendly than chemical control since no completely novel molecule is being introduced to the environment.

8. Phytopathogenic Fusarium spp. and Global Food Security

Fusarium is regarded as the most devastating fungi to crop plants in most parts of the world, especially in Asia, Africa, the USA, Europe, and Australia [156]. It damages the host plant severely, causing a decrease in quality and productivity and affecting plant development.
Fusarium diseases of crops have been increasing in severity since the last century and now pose a serious threat to global food security [157,158]. Recently, it has become evident that Fusarium pathogens pose the greatest biotic challenge to many of the world’s staple crops. They destroy both the essential calorie crops, such as rice, wheat, maize, and soybean, and the commodity crops, such as bananas, coffee, and barley [159,160]. Fusarium infection of crops also causes significant socio-economic and international trade implications for global food security by its ability to reduce crop yields and contaminate plant products with mycotoxins. Additionally, the loss of commodity crops to fungal disease destabilizes the economies of developing countries that rely on export revenues generated by the global trade of these crops to import food from elsewhere [160], thereby increasing the dimension of the food insecurity threat.
Moreover, modern agricultural practices of monoculture planting of uniform crops have accelerated the emergence of new virulent fusarium strains [161]. Climate change, on the other hand, also heightens the problem, as we observe altered disease demographics as the pathogens move towards the warmer region [162]. In addition, the trade and transport of plants and plant products have further dispersed plant pathogens to new hosts in previously unaffected areas of the world [160]. However, modern intensive agriculture has become the mainstay of global food security and generates the calories needed to feed the increasing global population. Nevertheless, we face a changing world plagued by fungal pathogens and new crop diseases. Therefore, to protect our future harvests, there is a need for better disease surveillance and the adoption of new disease intervention strategies.

9. Conclusions

The Fusarium spp. is one of the most important fungal groups and is significant in the fields of agriculture, medicine, and veterinary science [163]. They have emerged over the past decades as an important cause of diverse plant diseases, opportunistic human and animal pathogens, and mycotoxin producers. Many of the world’s staple food crops are prone to fungal plant pathogens [164]. Furthermore, the loss of economically important crops to these fungal diseases can destabilize the economies of developing nations who depend on export revenues generated to import food grown somewhere else in the world. However, successful disease management strategies are challenging, since fungi can easily develop resistance to various methods of control. Therefore, biological control of fungal diseases has emerged as one of the alternative methods of control and promises much toward increasing world crop production. Thus, we need better disease surveillance, more accurate predictive forecasting, and new disease interventions in order to achieve better disease control and protect our future harvests, thus achieving sustained global food security.

Author Contributions

T.I.E. conceptualized the topic and designed, wrote, and performed revision of the manuscript. M.M. is the head of the lab. All authors have read and agreed to the published version of the manuscript.

Funding

This review was part of research funded by the National Research Foundation of South Africa (NRF), grant number 116643, and North-West University, South Africa.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We have a lot of published articles on Fusarium and the approaches to its control and management. Nevertheless, it was not possible to cite all of them. I, therefore, want to thank those researchers whose works were not cited for their understanding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Tian, B.; Xie, J.; Fu, Y.; Cheng, J.; Li, B.; Chen, T.; Zhao, Y.; Gao, Z.; Yang, P.; Barbetti, M.J.; et al. A cosmopolitan fungal pathogen of dicots adopts an endophytic lifestyle on cereal crops and protects them from major fungal diseases. ISME J. 2020, 14, 3120–3135. [Google Scholar] [CrossRef] [PubMed]
  2. Wu, F. Perspective: Time to face the fungal threat. Nature 2014, 516, S7. [Google Scholar] [CrossRef] [PubMed]
  3. Ekwomadu, T.I.; Gopane, R.E.; Mwanza, M. Occurrence of filamentous fungi in maize destined for human consumption in South Africa. Food Sci. Nutr. 2018, 6, 884–890. [Google Scholar] [CrossRef] [PubMed]
  4. Ma, L.J.; Geiser, D.M.; Proctor, R.H.; Rooney, A.P.; O’Donnell, K.; Trail, F.; Gardiner, D.M.; Manners, J.M.; Kazan, K. Fusarium pathogenomics. Annu. Rev. Microbiol. 2013, 67, 399–416. [Google Scholar] [CrossRef]
  5. Ekwomadu, T.I. Occurrence and Variation of Fusarium Free and Masked Mycotoxins in Maize from Agriculture Regions of South Africa. Doctoral Dissertation, North-West University, Potchefstroom, South Africa, 2019. [Google Scholar]
  6. Dean, R.; van Kan, J.A.L.; Pretorius, Z.A.; Hammond-Kosack, K.E.; Di Pietro, A.; Spanu, P.D.; Rudd, J.J.; Dickman, M.; Kahmann, R.; Ellis, J.; et al. The top 10 fungal pathogens in molecular plant pathology. Mol. Plant. Pathol. 2012, 13, 414–430. [Google Scholar] [CrossRef]
  7. Burgess, L.W.; Bryden, W.L. Fusarium: A ubiquitous fungus of global significance. Microbiol. Aust. 2012, 33, 22–25. [Google Scholar] [CrossRef]
  8. Rampersad, S.N. Pathogenomics and Management of Fusarium Diseases in Plants. Pathogens 2020, 9, 340. [Google Scholar] [CrossRef]
  9. Nelson, P.E.; Toussoun, T.A.; Cook, R.J. Fusarium: Diseases, Biology, and Taxonomy; The Pennsylvania State University: State College, PA, USA, 1981. [Google Scholar]
  10. Nelson, P.E.; Toussoun, T.A.; Marasas, W.F.O. Fusarium Species: An Illustrated Manual for Identification; Pennsylvania State University Press: State College, PA, USA, 1983. [Google Scholar]
  11. Logrieco, A.; Bottalico, A.; Mule, G.; Moretti, A.; Perrone, G. Epidemiology of toxigenic fungi and their associated mycotoxins for some Mediterranean crops. Eur. J. Plant Pathol. 2003, 109, 645–667. [Google Scholar] [CrossRef]
  12. Leslie, J.F.; Summerell, B.A. The Fusarium Laboratory Manual; Blackwell Publishing: Hoboken, NJ, USA, 2006. [Google Scholar]
  13. Summerell, B.A.; Laurence, M.H.; Liew, E.C.Y.; Leslie, J.F. Biogeography and phylogeography of Fusarium: A review. Fungal Divers. 2010, 44, 3–13. [Google Scholar] [CrossRef]
  14. Wollenweber, H.W.; Reinking, O.A. Die Usarien, HhreBeschreibung, Schadwirkung, und Bekämpfung; Verlag Paul Parey: Berlin, Germany, 1983. [Google Scholar]
  15. Moss, M.O.; Thrane, U. Fusarium taxonomy with relation to trichothecene formation. Toxicol. Lett. 2004, 153, 23–28. [Google Scholar] [CrossRef]
  16. Booth, C. The Genus Fusarium; Commonwealth Mycological Institute, The Eastern Press Limited: London, UK, 1971. [Google Scholar]
  17. Munkvold, G. Fusarium Species and their Associated Mycotoxins. In Mycotoxigenic Fungi; Moretti, A., Susca, A., Eds.; Springer: New York, NY, USA, 2017; pp. 51–106. ISBN 978-1-4939-6705-6. [Google Scholar]
  18. Jay, J.M. Modern Food Microbiology, 3rd ed.; CBS Publishers and Distributors: New Delhi, India, 1987; pp. 541–551. [Google Scholar]
  19. Joffe, A.Z. A modern system of Fusarium taxonomy. Mycopathologia 1974, 53, 201–228. [Google Scholar] [CrossRef] [PubMed]
  20. Arici, S.E.; Koc, N.K. RAPD–PCR analysis of genetic variation among isolates of Fusarium graminearum and Fusarium culmorum from wheat in Adana Turkey. Pak. J. Biol. Sci. 2010, 13, 138–142. [Google Scholar] [PubMed]
  21. Martínez-García, L.B.; Armas, C.; Miranda, J.D.; Padilla, F.M.; Pugnaire, F.I. Shrubs influence arbuscular mycor-rhizal fungi communities in a semi-arid environment. Soil Biol. Biochem. 2011, 43, 682–689. [Google Scholar] [CrossRef]
  22. Leissner, C.E.W.; Niessen, M.L.; Vogel, R.F. Use of the AFLP technique for the identification and discrimination of Fusarium graminearum. Cereal Res. Commun. 1997, 25, 555–556. [Google Scholar] [CrossRef]
  23. Schmidt, H.; Adler, A.; Holst-Jensen, A.; Klemsdal, S.; Logrieco, A.; Mach, R.; Nirenberg, H.; Thrane, U.; Torp, M.; Vogel, R.; et al. An integrated taxonomic study of Fusarium langsethiae, Fusarium poae and Fusarium sporotrichioides based on the use of composite datasets. Int. J. Food Microbiol. 2004, 95, 341–349. [Google Scholar] [CrossRef]
  24. Konstantinova, P.; Yli-Mattila, T. IGS-RFLP analysis and development of molecular markers for identification of Fusarium poae, F. langsethiae, F. sporotrichioides and F. kyushuense. Int. J. Food Microbiol. 2004, 95, 321–331. [Google Scholar] [CrossRef]
  25. Yli-Mattila, T.; Mach, R.; Alekhina, I.; Bulat, S.; Koskinen, S.; Kullnig-Gradinger, C.; Kubicek, C.; Klemsdal, S. Phylogenetic relationship of Fusarium langsethiae to Fusarium poae and Fusarium sporotrichioides as inferred by IGS, ITS, β-tubulin sequences and UP-PCR hybridization analysis. Int. J. Food Microbiol. 2004, 95, 267–285. [Google Scholar] [CrossRef]
  26. Knutsen, A.; Torp, M.; Holst-Jensen, A. Phylogenetic analyses of the Fusarium poae, Fusarium sporotrichioides and Fusarium langsethiae species complex based on partial sequences of the translation elongation factor-1 alpha gene. Int. J. Food Microbiol. 2004, 95, 287–295. [Google Scholar] [CrossRef]
  27. White, T.J.; Bruns, T.; Lee, S.; Taylor, J. Amplification and direct sequencing of fungal ribosomal RNA genes for phylogenetics. In PCR Protocols: A Guide to Methods and Applications; Innis, M.A., Gelfand, D.H., Sninsky, J.J., White, T.J., Eds.; Academic Press: New York, NY, USA, 1990; pp. 315–322. [Google Scholar]
  28. Hatsch, D.; Phalip, V.; Jeltsch, J.M. Use of genes encoding cellobiohydrolase-C and topoisomerase II as targets for phylogenetic analyses and identification of Fusarium. Res. Microbiol. 2004, 155, 290–296. [Google Scholar] [CrossRef]
  29. Mulè, G.; Susca, A.; Stea, G.; Moretti, A. Specific detection of the toxigenic species Fusarium proliferatum and F. oxysporum from asparagus plants using primers based on calmodulin gene sequences. FEMS Microbiol. Lett. 2004, 230, 235–240. [Google Scholar] [CrossRef]
  30. Wigmann, F.; Behr, J.; Vogel, R.F.; Niessen, L. MALDI-TOF MS fingerprinting for identification and differentiation of species within the Fusarium fujikuroi species complex. Appl. Microbiol. Biotechnol. 2019, 103, 5323–5337. [Google Scholar] [CrossRef] [PubMed]
  31. Dassanayake, R.S.; Samaranayake, L.P. Amplification-Based Nucleic Acid Scanning Techniques to Assess Genetic Polymorphism in Candida. Crit. Rev. Microbiol. 2003, 29, 1–24. [Google Scholar] [CrossRef] [PubMed]
  32. Kumar, N.S.; Gurusubramanian, G. Random amplified polymorphic DNA (RAPD) markers and its applications. Sci. Vis. 2011, 3, 116–124. [Google Scholar]
  33. Nicholson, P.; Simpson, D.R.; Weston, G.; Rezanoor, H.N.; Lees, A.K.; Parry, D.W.; Joyce, D. Detection and quantification of Fusarium culmorum and Fusarium graminearum in cereals using PCR assays. Physiol. Mol. Plant Pathol. 1998, 53, 17–37. [Google Scholar] [CrossRef]
  34. Fungaro, M.H.P.; Magnani, M.; Vilas-Boas, L.A.; Vissotto, P.C.; Furlaneto, M.C.; Vieira, M.L.C.; Taniwaki, M.H. Genetic relationships among Brazilian strains of Aspergillus ochraceus based on RAPD and ITS sequences. Can. J. Microbiol. 2004, 50, 985–988. [Google Scholar] [CrossRef]
  35. Yli-Mattila, T.; Paavanen, S.; Hannukkala, A.; Parikka, P.; Tahvonen, R.; Karjalainen, R. Isozyme and RAPD-PCR analyses of Fusarium avenaceum strains from Finland. Plant Pathol. 1996, 45, 126–134. [Google Scholar] [CrossRef]
  36. Kernnyi, Z.; Tabarhegyi, E.; Pomazi, A.; Hornok, L. Variability among strains of Fusarium Poae assessed by vege-tative compatibility and RAPD polymorphism. Plant Pathol. 1997, 46, 882–889. [Google Scholar] [CrossRef]
  37. Hue, F.-X.; Huerre, M.; Rouffault, M.A.; de Bievre, C. Specific Detection of Fusarium Species in Blood and Tissues by a PCR Technique. J. Clin. Microbiol. 1999, 37, 2434–2438. [Google Scholar] [CrossRef]
  38. Paavanen-Huhtala, S.; Hyvönen, J.; Bulat, S.; Yli-Mattila, T. RAPD-PCR, isozyme, rDNA RFLP and rDNA sequence analyses in identification of Finnish Fusarium oxysporum isolates. Mycol. Res. 1999, 103, 625–634. [Google Scholar] [CrossRef]
  39. Ellsworth, D.L.; Rittenhouse, K.D.; Honeycutt, R.L. Artifactual variation in randomly amplified polymorphic DNA banding patterns. BioTechniques 1993, 14, 214–217. [Google Scholar]
  40. Khandka, D.K.; Tuna, M.; Tal, M.; Nejidat, A.; Golan-Goldhirsh, A. Variability in the pattern of random amplified polymorphic DNA. Electrophoresis 1997, 18, 2852–2856. [Google Scholar] [CrossRef]
  41. Capote, N.; Pastrana, A.M.; Aguado, A.; Sánchez-Torres, P. Molecular Tools for Detection of Plant Pathogenic Fungi and Fungicide Resistance. In Plant Pathology; Cumagun, C.J., Ed.; InTechOpen: London, UK, 2012; ISBN 978-953-51-0489-6. [Google Scholar]
  42. Thies, J.E. Soil Microbial Community Analysis using Terminal Restriction Fragment Length Polymorphisms. Soil Sci. Soc. Am. J. 2007, 71, 579–591. [Google Scholar] [CrossRef]
  43. Kim, Y.T.; Cho, M.; Jeong, J.Y.; Lee, H.B.; Kim, S.B. Application of Terminal Restriction Fragment Length Polymorphism (T-RFLP) analysis to monitor effect of biocontrol agents on rhizosphere microbial community of hot pepper (Capsicum annuum L.). J. Microbiol. 2010, 48, 566–572. [Google Scholar] [CrossRef]
  44. Hyakumachi, M.; Priyatmojo, A.; Kubota, M.; Fukui, H. New anastomosis groups, AG-T and AG-U, of binucleate Rhizoctonia spp. causing root and stem rot of cutflower and miniature roses. Phytopathology 2005, 95, 784–792. [Google Scholar] [CrossRef]
  45. Vos, P.; Hogers, R.; Bleeker, M.; Reijans, M.; Van De Lee, T.; Hornes, M.; Friters, A.; Pot, J.; Paleman, J.; Kuiper, M.; et al. AFLP: A new technique for DNA fingerprinting. Nucleic Acids Res. 1995, 21, 4407–4414. [Google Scholar] [CrossRef]
  46. McDonald, B.A.; Kimunye, J.N.; Muzhinji, N.; Mostert, D.; Viljoen, A.; der Merwe, A.E.B.-V.; Everhart, S.; Gambhir, N.; Stam, R.; Wyman, C.R.; et al. The Population Genetics of Fungi: Tools and Techniques. Phytopathology 1997, 87, 448–453. [Google Scholar] [CrossRef]
  47. Gril, T.; Celar, F.; Munda, A.; Javornik, B.; Jakse, J. AFLP Analysis of Intraspecific Variation Between Monilinia laxa Isolates from Different Hosts. Plant Dis. 2008, 92, 1616–1624. [Google Scholar] [CrossRef]
  48. Stewart, J.E.; Kim, M.-S.; James, R.L.; Dumroese, R.K.; Klopfenstein, N.B.; Hassan, O.; Lim, T.-H.; Chang, T.; Zhu, Z.; Zheng, L.; et al. Molecular Characterization of Fusarium oxysporum and Fusarium commune Isolates from a Conifer Nursery. Phytopathology 2006, 96, 1124–1133. [Google Scholar] [CrossRef]
  49. Baayen, R.P.; O’Donnell, K.; Bonants, P.J.M.; Cigelnik, E.; Kroon, L.P.N.M.; Roebroeck, E.J.A.; Waalwijk, C. Gene genealogies and AFLP analyses in the Fusarium oxysporum complex identify monophyletic and nonmonophyletic formae speciales causing wilt and rot disease. Phytopathology 2000, 90, 891–900. [Google Scholar] [CrossRef]
  50. Groenewald, S.; Van Den Berg, N.; Marasas, W.F.; Viljoen, A. The application of high-throughput AFLP’s in assessing genetic diversity in Fusarium oxysporum f. sp. cubense. Mycol. Res. 2006, 110, 297–305. [Google Scholar] [CrossRef]
  51. Fourie, G.; Steenkamp, E.T.; Ploetz, R.C.; Gordon, T.R.; Viljoen, A. Current status of the taxonomic position of Fusarium oxysporum formae specialis cubense within the Fusarium oxysporum complex. Infect. Genet. Evol. 2011, 11, 533–542. [Google Scholar] [CrossRef]
  52. Taylor, J.W.; Jacobson, D.J.; Kroken, S.; Kasuga, T.; Geiser, D.M.; Hibbett, D.S.; Fisher, M.C. Phylogenetic Species Recognition and Species Concepts in Fungi. Fungal Genet. Biol. 2000, 31, 21–32. [Google Scholar] [CrossRef] [PubMed]
  53. Hillis, D.M.; Dixon, M.T. Ribosomal DNA: Molecular Evolution and Phylogenetic Inference. Q. Rev. Biol. 1991, 66, 411–453. [Google Scholar] [CrossRef] [PubMed]
  54. Mishra, P.K.; Tewari, J.P.; Clear, R.M.; Turkington, T.K. Genetic diversity and recombination within populations of Fusarium pseudograminearum from western Canada. Int. Microbiol. 2006, 9, 65–68. [Google Scholar] [PubMed]
  55. Mbofung, G.C.Y.; Hong, S.G.; Pryor, B.M.; Herrero, M.L.; Nagy, N.E.; Solheim, H.; Hafez, M.; Abdelmagid, A.; Aboukhaddour, R.; Adam, L.R.; et al. Phylogeny of Fusarium oxysporum f. sp. lactucae Inferred from Mitochondrial Small Subunit, Elongation Factor 1-α, and Nuclear Ribosomal Intergenic Spacer Sequence Data. Phytopathology 2007, 97, 87–98. [Google Scholar] [CrossRef]
  56. Samson, R.A.; Seifert, K.A.; Kuijpers, A.F.A.; Houbraken, J.A.M.P.; Frisvad, J.C. Phylogenetic analysis of Penicillium subgenus Penicillium using partial B-tubulin sequences. Stud. Mycol. 2004, 49, 175–200. [Google Scholar]
  57. Amrani, L.; Corio-Costet, M.-F. A single nucleotide in the β-tubulin gene distinguishing two genotypes of Erysiphe necator expressing different symptoms on grapevine. Plant Pathol. 2006, 55, 505–512. [Google Scholar] [CrossRef]
  58. Geiser, D.M.; Jiménez-Gasco, M.d.M.; Kang, S.; Makalowska, I.; Veeraraghavan, N.; Ward, T.J.; Zhang, N.; Kuldau, G.A.; O’Donnell, K. FUSARIUM-ID v. 1.0: A DNA Sequence Database for Identifying Fusarium. Eur. J. Plant Pathol. 2004, 110, 473–479. [Google Scholar] [CrossRef]
  59. O’Donnell, K.; Kistler, H.C.; Cigelnik, E.; Ploetz, R.C. Multiple evolutionary origins of the fungus causing Panama disease of banana: Concordant evidence from nuclear and mitochondrial gene genealogies. Proc. Natl. Acad. Sci. USA 1998, 9, 2044–2049. [Google Scholar] [CrossRef]
  60. Bruns, T.D.; Shefferson, R.P. Evolutionary studies of ectomycorrhizal fungi: Recent advances and future directions. Can. J. Bot. 2004, 82, 1122–1132. [Google Scholar] [CrossRef]
  61. Young-Mi, L.; Choi, Y.-K.; Min, B.-R. PCR-RFLP and sequence analysis of the rDNA ITS region in the Fusarium spp. J. Microbiol. 2000, 38, 66–73. [Google Scholar]
  62. Durán, A.; Gryzenhout, M.; Drenth, A.; Slippers, B.; Ahumada, R.; Wingfield, B.D.; Wingfield, M.J. AFLP analysis reveals a clonal population of Phytophthora pinifolia in Chile. Fungal Biol. 2010, 114, 746–752. [Google Scholar] [CrossRef]
  63. Sanitá-Lima, M.; de Lucas, R.C.; Lima, N.; Polizeli, M.L.T.; Santos, C. Fungal community ecology using MALDI-TOF MS demands curated mass spectral databases. Front. Microbiol. 2019, 10, 315. [Google Scholar] [CrossRef]
  64. Quéro, L.; Girard, V.; Pawtowski, A.; Tréguer, S.; Weill, A.; Arend, S.; Cellière, B.; Polsinelli, S.; Monnin, V.; van Belkum, A.; et al. Development and application of MALDI-TOF MS for identification of food spoilage fungi. Food Microbiol. 2019, 81, 76–88. [Google Scholar] [CrossRef] [PubMed]
  65. Huschek, D.; Witzel, K. Rapid dereplication of microbial isolates using matrix-assisted laser desorption ionization time-of-flight mass spectrometry: A mini-review. J. Adv. Res. 2019, 19, 99–104. [Google Scholar] [CrossRef] [PubMed]
  66. Tupaki-Sreepurna, A.; Kindo, A.J. Fusarium The versatile pathogen. Indian J. Med. Microbiol. 2018, 36, 8–17. [Google Scholar] [CrossRef] [PubMed]
  67. Elvers, K.T.; Leeming, K.; Moore, C.P.; Lappin-Scott, H.M. Bacterial-fungal biofilms in flowing water pho-to-processing tanks. J. Appl. Microbiol. 1998, 84, 607–618. [Google Scholar] [CrossRef] [PubMed]
  68. Williams, M.M.; Armbruster, C.R.; Arduino, M.J. Plumbing of hospital premises is a reservoir for opportunistically pathogenic microorganisms: A review. Biofouling 2013, 29, 147–162. [Google Scholar] [CrossRef]
  69. Placinta, C.M.; D’Mello, J.P.F.; Macdonald, A.M.C. A review of worldwide contamination of cereal grains and animal feed with Fusarium mycotoxins. Anim. Feed Sci. Technol. 1999, 78, 21–37. [Google Scholar] [CrossRef]
  70. Gale, L.R.; Chen, L.F.; Hernick, C.A.; Takamura, K.; Kistler, H.C. Population Analysis of Fusarium graminearum from Wheat Fields in Eastern China. Phytopathology 2002, 92, 1315–1322. [Google Scholar] [CrossRef]
  71. Nucci, M.; Anaissie, E. Fusarium Infections in Immunocompromised Patients. Clin. Microbiol. Rev. 2007, 20, 695–704. [Google Scholar] [CrossRef] [PubMed]
  72. Nelson, P.E.; Dignani, M.C.; Anaissie, E.J. Taxonomy, biology, and clinical aspects of Fusarium species. Clin. Microbiol. Rev. 1994, 7, 479–504. [Google Scholar] [CrossRef] [PubMed]
  73. Guarro, J. Fusariosis, a complex infection caused by a high diversity of fungal species refractory to treatment. Eur. J. Clin. Microbiol. Infect. Dis. 2013, 32, 1491–1500. [Google Scholar] [CrossRef] [PubMed]
  74. Richardson, M.; Lass-Flörl, C. Changing epidemiology of systemic fungal infections. Clin. Microbiol. Infect. 2008, 14, 5–24. [Google Scholar] [CrossRef]
  75. Lysøe, E.; Klemsdal, S.S.; Bone, K.R.; Frandsen, R.J.N.; Johansen, T.; Thrane, U.; Giese, H. The PKS4 Gene of Fusarium graminearum Is Essential for Zearalenone Production. Appl. Environ. Microbiol. 2006, 72, 3924–3932. [Google Scholar] [CrossRef]
  76. Schollenberger, M.; Muller, H.M.; Rufle, M.; Suchy, S.; Drochner, W. Redistribution of 16 Fusarium toxins during comer cial dry milling of maize. Cereal Chem. 2008, 85, 557–560. [Google Scholar] [CrossRef]
  77. Marasas, W.F.O.; Wehner, F.C.; van Rensburg, S.J.; van Schalkwyk, D.J. Mycoflora of corn produced in human esoph ageal cancer areas in Transkei, Southern Africa. Phytopathology 1981, 71, 792–796. [Google Scholar] [CrossRef]
  78. Rheeder, J.P.; Marasas, W.F.O.; Thiel, P.G.; Sydenham, E.W.; Shephard, G.S.; van Schalkwyk, D.J. Fusarium moniliform and fumonisins in corn in relation to human oesophageal cancer in Transkei. Phytopathology 1992, 82, 353–357. [Google Scholar] [CrossRef]
  79. Cotten, T.K.; Munkvold, G.P. Survival of Fusarium moniliforme, F. proliferatum and F. subglutinans in maize stalk residue. Ecol. Popul. Biol. 1998, 88, 550–555. [Google Scholar] [CrossRef]
  80. Lucas, J. Plant Diseases. In Plant Pathology and Plant Pathogens; Blackwell Publishing: Malden, MA, USA, 1998. [Google Scholar]
  81. Bishop, C.; Cooper, R. An ultrastructural study of root invasion in three vascular wilt diseases. Physiol. Plant Pathol. 1983, 22, 15-IN13. [Google Scholar] [CrossRef]
  82. Vidhyasekaran, P. Fungal Pathogenesis in Plants and Crops. In Molecular Biology and Host Defense Mechanisms; Marcel Dekker Inc.: New York, NY, USA, 1997; p. 553. [Google Scholar]
  83. Recorbet, G.; Alabouvette, C. Adhesion of Fusarium oxysporum conidia to tomato roots. Lett. Appl. Microbiol. 1997, 25, 375–379. [Google Scholar] [CrossRef]
  84. Mendgen, K.; Hahn, M.; Deising, H. Morphogenesis and mechanisms of penetration by plant pathogenic fungi. Annu. Rev. Phytopathol. 1996, 34, 367–386. [Google Scholar] [CrossRef]
  85. Koretsky, L.S. The influence of Fusarium oxysporum infection and low temperatures on the activity of soybean esterase and PR proteins. Búvisindi Icel. Agric. Sci. 2001, 14, 67–73. [Google Scholar]
  86. Knogge, W. Fungal Infection of Plants. Plant Cell 1996, 8, 1711–1722. [Google Scholar] [CrossRef] [PubMed]
  87. Walton, J.D. Deconstructing the cell wall. Plant Physiol. 1994, 104, 191–196. [Google Scholar] [CrossRef] [PubMed]
  88. Bishop, C.; Cooper, R. An ultrastructural study of vascular colonization in three vascular wilt diseases I. Colonization of susceptible cultivars. Physiol. Plant Pathol. 1983, 23, 323–343. [Google Scholar] [CrossRef]
  89. Beckman, C.H.; Mace, M.E.; Halmos, S.; McGahan, M.W. Physical barriers associated with resistance in Fusarium wilt of bananas. Phytopathology 1961, 51, 507–515. [Google Scholar]
  90. Beckman, C.H.; Halmos, S.; Mace, M.E. The interaction of host, pathogen and soil temperature in relation to sus-ceptibility to Fusarium wilt of bananas. Phytopathology 1962, 52, 134–140. [Google Scholar]
  91. Beckman, C.H. The Nature of Wilt Diseases of Plants; American Phytopathological Society: St. Paul, MN, USA, 1987; p. 175. [Google Scholar]
  92. MacHardy, W.E.; Beckman, C.H. Vascular wilt Fusaria: Infections and Pathogenesis. In Fusarium: Diseases, Biology and Taxonomy; Nelson, P.E., Toussoun, T.A., Cook, R.J., Eds.; The Pennysylvania State University Press: State College, PA, USA, 1981; pp. 365–390. [Google Scholar]
  93. Schnathorst, W.C. Life cycle and epidemiology of Verticillium. In Fungal Wilt Diseases of Plants; Mace, M.E., Ed.; Academic Press, Inc.: New York, NY, USA, 1981; pp. 81–111. [Google Scholar]
  94. Huisman, O.C. Interactions of root growth dynamics to epidemiology of root invading fungi. Annu. Rev. Phytopathol. 1982, 20, 303–327. [Google Scholar] [CrossRef]
  95. Moradi, M.; Oerke, E.-C.; Steiner, U.; Tesfaye, D.; Schellander, K.; Dehne, H.-W. Microbiological and Sybr® Green real-time PCR detection of major Fusarium head blight pathogens on wheat ears. Microbiology 2010, 79, 646–654. [Google Scholar] [CrossRef]
  96. Prados-Ligero, A.M.; Basallote-Ureba, M.J.; Melero-Vara, J.M. First report of Fusarium oxysporum f. sp. lilii and F. proliferatum affecting Lilium crops in Spain. Trop. Plant Pathol. 2008, 33, 235–236. [Google Scholar] [CrossRef]
  97. Aktaruzzaman, M.; Xu, S.J.; Kim, J.Y. First report of potato stem-end rot caused by Fusarium oxysporum in Korea. Mycobiology 2014, 2, 206–209. [Google Scholar] [CrossRef] [PubMed]
  98. Viljoen, A. The status of Fusarium wilt (Panama disease) of banana in South Africa. S. Afr. J. Sci. 2002, 98, 341–344. [Google Scholar]
  99. Green, J.; Nelson, S. Heart and root rots of pineapples. Plant Dis. 2015, 106, 1–7. [Google Scholar]
  100. Nilmini, R.K.; Panapitiya, D.; Abeywickrama, K.; Kuruppu, M. Morphological and molecular identification of fungal species associated with postharvest stem-end rot disease of avocado in Sri Lanka. Sri Lanka J. Food Agric. 2020, 6, 47–56. [Google Scholar] [CrossRef]
  101. Wulff, E.G.; Sørensen, J.L.; Lübeck, M.; Nielsen, K.F.; Thrane, U.; Torp, J. Fusarium spp. associated with rice Bakanae: Ecology, genetic diversity, pathogenicity and toxigenicity. Environ. Microbiol. 2010, 12, 649–657. [Google Scholar] [CrossRef]
  102. Flood, J.A. Review of Fusarium Wilt of Oil Palm Caused by Fusarium oxysporum f. sp. Elaeidis. Phytopathology 2006, 6, 660–662. [Google Scholar] [CrossRef]
  103. Umaña-Rojas, G.; García, J. Frequency of organisms associated with crown rot of bananas in integrated and organic production systems. Acta Hortic. 2011, 906, 211–217. [Google Scholar] [CrossRef]
  104. Brandfass, C.; Karlovsky, P. Simultaneous detection of Fusarium culmorum and F. graminearum in plant material by duplex PCR with melting curve analysis. BMC Microbiol. 2006, 6, 4. [Google Scholar] [CrossRef]
  105. Omar, N.H.; Mohd, M.; Nor, N.M.I.M.; Zakaria, L. Characterization and pathogenicity of Fusarium species associated with leaf spot of mango (Mangifera indica L.). Microb. Pathog. 2018, 114, 362–368. [Google Scholar] [CrossRef]
  106. Barral, B.; Chillet, M.; Doizy, A.; Grassi, M.; Ragot, L.; Léchaudel, M.; Durand, N.; Rose, L.J.; Viljoen, A.; Schorr-Galindo, S. Diversity and Toxigenicity of Fungi that Cause Pineapple Fruitlet Core Rot. Toxins 2020, 12, 339. [Google Scholar] [CrossRef] [PubMed]
  107. Rampersad, S.N.; Teelucksingh, L.D. First report of Fusarium proliferatum infecting pimento chili peppers in Trinidad. Plant Dis. 2011, 5, 13. [Google Scholar] [CrossRef] [PubMed]
  108. Jee, H.J.; Ryu, K.Y.; Shim, C.K.; Nam, K.W. Occurrence of stem and fruit rot paprika caused by Nectria haemato-cocca. Plant Pathol. J. 2005, 21, 317–321. [Google Scholar] [CrossRef]
  109. Wanjiku, E.K.; Waceke, J.W.; Wanjala, B.W.; Mbaka, J.N. Identification and Pathogenicity of Fungal Pathogens Associated with Stem End Rots of Avocado Fruits in Kenya. Int. J. Microbiol. 2020, 2020, 4063697. [Google Scholar] [CrossRef]
  110. Singh, S.K.; Kumar, R. Etiology, symptoms and molecular characterization of papaya root rot—A new and serious threat. Indian Phytopathol. 2015, 68, 348–349. [Google Scholar]
  111. Boutigny, A.-L.; Beukes, I.; Small, I.; Zühlke, S.; Spiteller, M.; Van Rensburg, B.J.; Flett, B.; Viljoen, A. Quantitative detection of Fusarium pathogens and their mycotoxins in South African maize. Plant Pathol. 2012, 61, 522–531. [Google Scholar] [CrossRef]
  112. Guo, Z.; Yu, Z.; Li, Q.; Tang, L.; Guo, T.; Huang, S.; Mo, J.; Hsiang, T.; Luo, S. Fusarium species associated with leaf spots of mango in China. Microb. Pathog. 2021, 150, 104736. [Google Scholar] [CrossRef]
  113. Marin, D.H.; Sutton, T.B.; Blankenship, S.M.; Swallow, W.H. Pathogenicity of fungi associated with crown rot of bananas in Latin America on Grande Naine and disease-resistant hybrid bananas. Plant Dis. 1996, 80, 525–528. [Google Scholar] [CrossRef]
  114. Zhu, Z.; Zheng, L.; Pan, L.; Hsiang, T.; Huang, J. Identification and characterization of Fusarium species associated with wilt of Eleocharis dulcis (Chinese water chestnut) in China. Plant Dis. 2014, 98, 977–987. [Google Scholar] [CrossRef]
  115. Sharddha, G.; Lal, A.A. Eco-friendly management of post-harvest fungal pathogen causing Fusarium rot of papaya (Carica papaya L.) in Allahabad. Natl. Acad. Sci. Lett. 2010, 33, 227–233. [Google Scholar]
  116. Suratos, S.C.M. Interaction of molds associated with stem-end rot in avocado (Persea Americana Mill.) fruit. CLSU Sci. J. 2005, 25, 63–64. [Google Scholar]
  117. Darvas, J.M.; Kotze, J.M. Post-harvest diseases of avocados. S. Afr. Avocado Grow. Assoc. Yearb. 1981, 4, 63–66. [Google Scholar]
  118. Flückiger, U.; Marchetti, O.; Bille, J.; Eggimann, P.; Zimmerli, S.; Imhof, A.; Garbino, J.; Ruef, C.; Pittet, D.; Täuber, M.; et al. Treatment options of invasive fungal infections in adults. Swiss Med. Wkly. 2006, 136, 447–463. [Google Scholar]
  119. Stempel, J.M.; Hammond, S.P.; Sutton, D.A.; Weiser, L.M.; Marty, F.M. Invasive Fusariosis in the Voriconazole Era: Single-Center 13-Year Experience. Open Forum Infect. Dis. 2015, 2, ofv099. [Google Scholar] [CrossRef] [PubMed]
  120. Nucci, M.; Anaissie, E. Cutaneous Infection by Fusarium Species in Healthy and Immunocompromised Hosts: Implications for Diagnosis and Management. Clin. Infect. Dis. 2002, 35, 909–920. [Google Scholar] [CrossRef]
  121. Pontón, J.; Rüchel, R.; Clemons, K.V.; Coleman, D.C.; Grillot, R.; Guarro, J.; Aldebert, D.; Ambroise-Thomas, P.; Cano, J.; Carrillo-Muñoz, A.J.; et al. Emerging pathogens. MedicalMycology 2000, 38, 225–236. [Google Scholar]
  122. Sullivan, D.J.; Moran, G.P. (Eds.) Human Pathogenic Fungi: Molecular Biology and Pathogenic Mechanisms; Caister Academic Press: Norfolk, UK, 2014. [Google Scholar]
  123. Gnat, S.; Łagowski, D.; Nowakiewicz, A.; Dyląg, M. A global view on fungal infections in humans and animals: Opportunistic infections and microsporidioses. J. Appl. Microbiol. 2021, 131, 2095–2113. [Google Scholar] [CrossRef]
  124. Kobayashi, G.S. Disease of Mechanisms of Fungi. In Medical Microbiology, 4th ed.; Baron, S., Ed.; University of Texas Medical Branch: Galveston, TX, USA, 1996; Chapter 74. [Google Scholar]
  125. Van Burik, J.A.; Magee, P.T. Aspects of fungal pathogenesis in humans. Annu. Rev. Microbiol. 2001, 55, 743–772. [Google Scholar] [CrossRef]
  126. Vennewald, I.; Wollina, U. Cutaneous infections due to opportunistic molds: Uncommon presentations. Clin. Dermatol. 2005, 23, 565–571. [Google Scholar] [CrossRef]
  127. Lukaszuk, C.R.; Kułak, W. Effects of fungal air pollution on human health. Prog. Health Sci. 2011, 1, 156–164. [Google Scholar]
  128. Gurdaswani, V.; Ghag, S.B. Chapter 2—Toxins from Fusarium Species and their Role in Animal and Plant Diseases. In New and Future Developments in Microbial Biotechnology and Bioengineering; Singh, J., Gehlot, P., Eds.; Elsevier: Amsterdam, The Netherlands, 2020; pp. 7–27. ISBN 9780128210062. [Google Scholar]
  129. Perincherry, L.; Lalak-Kańczugowska, J.; Stępień, Ł. Fusarium-Produced Mycotoxins in Plant-Pathogen Interactions. Toxins 2019, 11, 664. [Google Scholar] [CrossRef] [PubMed]
  130. Kang, Z.; Buchenauer, H. Ultrastructural and Cytochemical Studies on Cellulose, Xylan and Pectin Degradation in Wheat Spikes Infected by Fusarium culmorum. J. Phytopathol. 2000, 148, 263–275. [Google Scholar] [CrossRef]
  131. Kang, Z.; Zingen-Sell, I.; Buchenauer, H. Infection of wheat spikes by Fusarium avenaceum and alterations of cell wall components in the infected tissue. Eur. J. Plant Pathol. 2005, 111, 19–28. [Google Scholar] [CrossRef]
  132. Peng, Y.; Li, S.J.; Yan, J.; Tang, Y.; Cheng, J.P.; Gao, A.J.; Yao, X.; Ruan, J.J.; Xu, B.L. Research Progress on Phytopathogenic Fungi and Their Role as Biocontrol Agents. Front. Microbiol. 2021, 12, 670135. [Google Scholar] [CrossRef]
  133. Jajić, I.; Dudaš, T.; Krstović, S.; Krska, R.; Sulyok, M.; Bagi, F.; Savić, Z.; Guljaš, D.; Stankov, A. Emerging Fusarium Mycotoxins Fusaproliferin, Beauvericin, Enniatins, and Moniliformin in Serbian Maize. Toxins 2019, 11, 357. [Google Scholar] [CrossRef]
  134. Lyu, X.; Shen, C.; Fu, Y.; Xie, J.; Jiang, D.; Li, G.; Cheng, J. Comparative genomic and transcriptional analyses of the car-bohydrate-active enzymes and secretomes of phytopathogenic fungi reveal their significant roles during infection and development. Sci. Rep. 2015, 5, 15565. [Google Scholar] [CrossRef]
  135. De Guillen, K.; Ortiz-Vallejo, D.; Gracy, J.; Fournier, E.; Kroj, T.; Padilla, A. Structure Analysis Uncovers a Highly Diverse but Structurally Conserved Effector Family in Phytopathogenic Fungi. PLoS Pathog. 2015, 11, e1005228. [Google Scholar] [CrossRef]
  136. Tan, K.-C.; Oliver, R.P. Regulation of proteinaceous effector expression in phytopathogenic fungi. PLoS Pathog. 2017, 13, e1006241. [Google Scholar] [CrossRef]
  137. Zhao, Z.; Liu, H.; Wang, C.; Xu, J.-R. Comparative analysis of fungal genomes reveals different plant cell wall degrading capacity in fungi. BMC Genom. 2013, 14, 274. [Google Scholar] [CrossRef]
  138. Guerriero, G.; Hausman, J.-F.; Strauss, J.; Ertan, H.; Siddiqui, K.S. Destructuring plant biomass: Focus on fungal and extremophilic cell wall hydrolases. Plant Sci. 2015, 234, 180–193. [Google Scholar] [CrossRef]
  139. Lara-Márquez, A.; Zavala-Páramo, M.G.; López-Romero, E.; Calderón-Cortés, N.; López-Gómez, R.; Conejo-Saucedo, U.; Cano-Camacho, H. Cloning and characterization of a pectin lyase gene from Colletotrichum lindemuthianum and comparative phyloge-netic/structural analyses with genes from phytopathogenic and saprophytic/opportunistic microorganisms. BMC Microbiol. 2011, 11, 260. [Google Scholar] [CrossRef]
  140. Janusz, G.; Pawlik, A.; Sulej, J.; Świderska-Burek, U.; Jarosz-Wilkołazka, A.; Paszczyński, A. Lignin degradation: Microorganisms, enzymes involved, genomes analysis and evolution. FEMS Microbiol. Rev. 2017, 41, 941–962. [Google Scholar] [CrossRef]
  141. Beliën, T.; Van Campenhout, S.; Van Acker, M.; Robben, J.; Courtin, C.M.; Delcour, J.A.; Volckaert, G. Mutational analysis of endoxylanases XylA and XylB from the phytopathogen Fusarium graminearum reveals comprehensive insights into their inhibitor insensitivity. Appl. Environ. Microbiol. 2007, 73, 4602–4608. [Google Scholar] [CrossRef]
  142. Félix, C.; Libório, S.; Nunes, M.; Félix, R.; Duarte, A.S.; Alves, A.; Esteves, A.C. Lasiodiplodia theobromae as a Producer of Biotechnologically Relevant Enzymes. Int. J. Mol. Sci. 2018, 19, 29. [Google Scholar] [CrossRef] [PubMed]
  143. Ma, H.; Zhang, B.; Gai, Y.; Sun, X.; Chung, K.R.; Li, H. Cellwall-degrading enzymes required for virulence in the host selective toxin producing necrotroph alternaria alternata of citrus. Front. Microbiol. 2019, 10, 2514. [Google Scholar] [CrossRef]
  144. Di Pietro, A.; García-Maceira, F.I.; Méglecz, E.; Roncero, M.I.G. A MAP kinase of the vascular wilt fungus Fusarium oxysporum is essential for root penetration and pathogenesis. Mol. Microbiol. 2001, 39, 1140–1152. [Google Scholar] [CrossRef] [PubMed]
  145. Jain, S.; Akiyama, K.; Mae, K.; Ohguchi, T.; Takata, R. Targeteddisruption of a G protein a subunit gene results in reduced pathogenicityin Fusarium oxysporum. Curr. Genet. 2002, 41, 407–413. [Google Scholar] [CrossRef] [PubMed]
  146. Jain, S.; Akiyama, K.; Takata, R.; Ohguchi, T. Signaling via the G-protein subunit FGA2 is necessary for pathogenesis in Fusarium oxysporum. FEMS Microbiol. Lett. 2005, 243, 165–172. [Google Scholar] [CrossRef]
  147. Martínez-Rocha, A.L.; Roncero, M.I.G.; López-Ramirez, A.; Mariné, M.; Guarro, J.; Martínez-Cadena, G.; Di Pietro, A. Rho1 has, distinct functions in morphogenesis, cell wall biosynthesis and virulence of Fusarium oxysporum. Cell. Microbiol. 2008, 10, 1339–1351. [Google Scholar] [CrossRef]
  148. López-Berges, M.S.; Hera, C.; Sulyok, M.; Schäfer, K.; Capilla, J.; Guarro, J.; Di Pietro, A. The velvet complex governs mycotoxin production and virulence of Fusarium oxysporum on plant and mammalian hosts. Mol. Microbiol. 2013, 87, 49–65. [Google Scholar] [CrossRef]
  149. Carreras-Villaseñor, N.; Rodríguez-Haas, J.B.; Martínez-Rodríguez, L.A.; Pérez-Lira, A.J.; Ibarra-Laclette, E.; Villafán, E.; Castillo-Díaz, A.P.; Ibarra-Juárez, L.A.; Carrillo-Hernández, E.D.; Sánchez-Rangel, D. Characterization of Two Fusarium solani Species Complex Isolates from the Ambrosia Beetle Xylosandrus morigerus. J. Fungi 2022, 8, 231. [Google Scholar] [CrossRef] [PubMed]
  150. Muimba-Kankolongo, A. Chapter 10—Leguminous Crops Food Crop Production by Smallholder Farmers in Southern Africa; Academic Press: Cambridge, MA, USA, 2018; pp. 173–203. [Google Scholar]
  151. Knights, E.J.; Hobson, K.B. Chickpea Overview. In Reference Module in Food Science, 2nd ed.; Elsevier: Amsterdam, The Netherlands, 2016; Volume 1. [Google Scholar] [CrossRef]
  152. Nikitin, D.A.; Ivanova, E.A.; Semenov, M.V.; Zhelezova, A.D.; Ksenofontova, N.A.; Tkhakakhova, A.K.; Kholodov, V.A. Diversity, Ecological Characteristics and Identification of Some Problematic Phytopathogenic Fusarium in Soil: A Review. Diversity 2023, 15, 49. [Google Scholar] [CrossRef]
  153. Goncharov, A.A.; Glebova, A.A.; Tiunov, A.V. Trophic interactions between Fusarium species and soil fauna: A meta-analysis of experimental studies. Appl. Soil Ecol. 2020, 145, 103–302. [Google Scholar] [CrossRef]
  154. Egel, D.S.; Martyn, R.D. Fusarium wilt of watermelon and other cucurbits. Plant Health Instr. 2007, 10, 1094. [Google Scholar] [CrossRef]
  155. Shah, L.; Ali, A.; Yahya, M.; Zhu, Y.; Wang, S.; Si, H.; Rahman, H.; Ma, C. Integrated control of fusarium head blight and deoxynivalenol mycotoxin in wheat. Plant Pathol. 2018, 67, 532–548. [Google Scholar] [CrossRef]
  156. Khokhar, M.; Hooda, K.; Sharma, S.; Singh, V.; Saini, A. Fusarium stalk rot: A major threat to maize production in India. Maize J. 2013, 1, 1–6. [Google Scholar]
  157. Fisher, M.C.; Henk, D.A.; Briggs, C.J.; Brownstein, J.S.; Madoff, L.C.; McCraw, S.L.; Gurr, S.J. Emerging fungal threats to animal, plant and ecosystem health. Nature 2012, 484, 186–194. [Google Scholar] [CrossRef]
  158. Savary, S.; Willocquet, L.; Pethybridge, S.J.; Esker, P.; McRoberts, N.; Nelson, A. The global burden of pathogens and pests on major food crops. Nat. Ecol. Evol. 2019, 3, 430–439. [Google Scholar] [CrossRef]
  159. Gordon, T.R.; Martyn, R.D. The Evolutionary Biology of Fusarium oxysporum. Annu. Rev. Phytopathol. 1997, 35, 111–128. [Google Scholar] [CrossRef]
  160. Fones, H.N.; Bebber, D.P.; Chaloner, T.M.; Kay, W.T.; Steinberg, G.; Gurr, S.J. Threats to global food security from emerging fungal and oomycete crop pathogens. Nat. Food 2020, 1, 332–342. [Google Scholar] [CrossRef]
  161. Fisher, M.C.; Hawkins, N.J.; Sanglard, D.; Gurr, S.J. Worldwide emergence of resistance to antifungal drugs challenges human health and food security. Science 2018, 360, 739–742. [Google Scholar] [CrossRef] [PubMed]
  162. Bebber, D.P.; Ramotowski, M.A.T.; Gurr, S.J. Crop pests and pathogens move polewards in a warming world. Nat. Clim. Chang. 2013, 3, 985–988. [Google Scholar] [CrossRef]
  163. Lević, J. Species of Genus FUSARIUM in the Fields of Agriculture, Veterinary and Human Medicine; Monograph, Maize Research Institute Zemun Polje and Serbian Genetic Society: Belgrade, Serbia, 2008. [Google Scholar]
  164. Spadaro, D.; Gullino, M.L. Improving the efficacy of biocontrol agents against soilborne pathogens. Crop. Prot. 2005, 24, 601–613. [Google Scholar] [CrossRef]
Figure 1. Spore morphological characteristics of Fusarium spp. (AD): Macroconidia shapes; (EH): macroconidia apical cell shapes; (IL): macroconidia basal cell shapes; (M): microconidia shapes; (N): phialide morphology; (O): microconidial chains. Adapted from [12].
Figure 1. Spore morphological characteristics of Fusarium spp. (AD): Macroconidia shapes; (EH): macroconidia apical cell shapes; (IL): macroconidia basal cell shapes; (M): microconidia shapes; (N): phialide morphology; (O): microconidial chains. Adapted from [12].
Agriculture 13 01810 g001
Table 1. Fusarium species and their major mycotoxins.
Table 1. Fusarium species and their major mycotoxins.
Fusarium SpeciesMajor Mycotoxins Produced
F. cerealisNIV, FX, ZEA
F. culmoriumDON, 3-AcDON, 15-AcDON, NIV, FX, ZEA
F. equisetiZEA, DAS
F. gramineariumDON, 15-AcDON, NIV, FX, ZEA
F. oxysporiumMoniliformin, fusaric acid
F. poaeT-2 toxin, HT-2 toxin, NIV, DAS, FX
F. ploriferatumFumonisin, fusarin C, moniliformin
F. solaniFusaric acid, solaniol
F. sporotrichioidesT-2 toxin, HT-2 toxin, NEO, DAS, FX, ZEA
F. verticiloidesFumonisin, fusarin C, moniliformin
F. sambucinumSambutoxin, DON, DAS, T-2 toxin
Deoxynivalenol (DON), 3-acetyl DON (3-AcDON), 15-acetyl DON (15-AcDON), Nivalenol (NIV), Zearalenone (ZEA), Fusarenon (FX), Neosolaniol (NEO), Diacetoxyscirpinol (DAS).
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

Ekwomadu, T.I.; Mwanza, M. Fusarium Fungi Pathogens, Identification, Adverse Effects, Disease Management, and Global Food Security: A Review of the Latest Research. Agriculture 2023, 13, 1810. https://doi.org/10.3390/agriculture13091810

AMA Style

Ekwomadu TI, Mwanza M. Fusarium Fungi Pathogens, Identification, Adverse Effects, Disease Management, and Global Food Security: A Review of the Latest Research. Agriculture. 2023; 13(9):1810. https://doi.org/10.3390/agriculture13091810

Chicago/Turabian Style

Ekwomadu, Theodora Ijeoma, and Mulunda Mwanza. 2023. "Fusarium Fungi Pathogens, Identification, Adverse Effects, Disease Management, and Global Food Security: A Review of the Latest Research" Agriculture 13, no. 9: 1810. https://doi.org/10.3390/agriculture13091810

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