Open access

Introductory Chapter: Fusarium: Pathogenicity, Infections, Diseases, Mycotoxins and Management

Written By

Tulin Askun

Submitted: 19 July 2017 Published: 25 July 2018

DOI: 10.5772/intechopen.76507

From the Edited Volume

Fusarium - Plant Diseases, Pathogen Diversity, Genetic Diversity, Resistance and Molecular Markers

Edited by Tulin Askun

Chapter metrics overview

2,230 Chapter Downloads

View Full Metrics

1. Introduction

The genus Fusarium contains pathogens that can cause significant harm to humans, animals and plants by infecting vegetables, grains and seeds and causing diseases in humans and animals. Fusarium oxysporum, F. solani and F. fujikuroi complexes are of great importance worldwide especially as a plant, human and animal pathogen.

Identifying Fusarium species is not easy. Currently, scientists are focused on identifying Fusarium species using molecular techniques, such as genetic markers and polymerase chain reaction-restriction fragment length polymorphism, for analyzing the rDNA internal transcribed spacer region.

The aim of this book is to highlight the new information reported by numerous studies on Fusarium species. The primary aims of this book are the following:

  1. To provide an overview of historical importance and taxonomy of Fusarium species

  2. To understand the mechanism of Fusarium infections and the factors that cause the infections

  3. To discuss Fusarium species-caused diseases, pathogen diversity and host range in plants

  4. To discuss mycotoxin contaminations in cereals

  5. To discuss plant secondary metabolites as well as anti-fungal and anti-mycotoxigenic compounds

  6. To discuss plant-Fusarium interactions

  7. To discuss the antagonistic activity of Trichoderma and Fusarium species

  8. To discuss genetic diversity, genetic resistance and molecular markers to investigate population diversity

  9. To discuss the environmental conditions that enable the opportunistic growth of Fusarium

  10. To discuss the distribution and evolution of the genes responsible for mycotoxin biosynthesis

  11. To discuss the steps that can be taken to prevent toxin production

  12. To discuss suitable approaches for Fusarium species disease management

  13. To discuss development of Fusarium-resistant cultivars to reduce the diseases caused by Fusarium species on a wide scale

Advertisement

2. Plant pathogens and cereals

Fusarium attacks numerous plants and cereals that are important for human and animal nutrition. It specifically infects certain parts of them, such as grains, seedlings, heads, roots or stem, and causes various diseases, reduced commercial yield, and decrease in product quality [1]. Fusarium head blight (FHB) [2, 3], foot (FR) and root rot (RR) [4] and crown rot (CR) are among the major diseases caused by them. FHB produced by F. graminearum (teleomorph Gibberella zeae, Schwabe) causes starch and protein losses in cereals [5]. Fusarium species are saprophytic and are found commonly growing on the plants as a pathogen. F. proliferatum is a plant pathogen that is capable of infecting many important crops. F. oxysporum f.sp. cubense (FOC) causes Fusarium wilt, which is the most destructive disease of banana [6]. Many Fusarium species from the F. solani species complex (FSSC) are pathogenic and virulent. FSSC causes diseases in many agriculturally important crops, such as FR and/or RR of the infected host plant and causes necrosis. Symptoms, such as wilting, stunting and chlorosis, vary widely according to FSSC pathogenesis and the host plant species. Necrosis depends on the severity of fungal development [4]. Two of the most serious diseases of wheat known globally are Fusarium CR and Fusarium FHB.

Stephens et al. [7] investigated the CR disease in wheat infected by F. graminearum and reported that CR developed in three stages. In the first stage, the F. graminearum biomass significantly increased within 2 days after inoculation. At this stage, there was germination of spores and superficial hyphal growth on the leaf sheath. In the second stage, the fungal biomass significantly decreased over 2 weeks. At this stage, the fungus penetrated from the outer parts of the leaf sheath to the leaf sheath base. In the third stage, biomass of F. graminearum increased significantly, and this increase correlated with fungal colonization on wheat and showed that the fungal biomass was being formed as fungal colonization on wheat crown parenchyma.

Advertisement

3. Fusarium infections in humans

Fusarium species cause superficial, locally invasive and diffuse infections in humans. Although Fusarium verticillioides, including F. moniliforme and F. fujikuroi species complex [8], are opportunistic pathogens, the species in the F. solani complex include pathogenic species [9].F. solani, F. oxysporum, F. verticillioides and F. proliferatum infect the immune-compromised patients. Sidhu et al. [10] reported that prevalent meningospondylodiscitis in an elderly diabetic patient caused by F. oxysporum. F. sacchari, F. anthophilum, F. chlamydosporum and F. dimerum was also thought as related to human disease. Guendouze-Bouchefa et al. [11] reported a rare case of perinephric abscess in a child caused by F. chlamydosporum.

The members of F. solani and F. oxysporum species complexes are known to include the agents that cause human infections worldwide. F. solani can adhere to and damage the corneal membrane [12]. Some Fusarium species, such as F. dimerum, are associated with keratomycosis, particularly in the bad hygiene conditions.

Advertisement

4. Fusarium diseases in animals

Fusarium mycotoxins affect the growth, reproduction and hormonal condition of the animal. The effect of these mycotoxins on animals depends on the quantity of mycotoxin intake. After intake, these mycotoxins arrive at the gastrointestinal epithelial cell layer which is covered by the mucous secreted from goblet cells [13, 14].

Although deoxynivalenol (DON) and fumonisin-B1 (FB1) increase the permeability of intestinal epithelial cell layer in humans, animals and birds, they worsen the viability and proliferation of intestinal epithelial cells. High doses of mycotoxins may cause abdominal distress, diarrhea, cardiac insufficiency, emesis and even death in pigs and equine leukoencephalomalacia (ELEM) in horses [15]. Through in vivo and in vitro experimental studies, Cortinovis et al. [16] demonstrated that ZEN and its metabolites markedly up-regulated estrogen secretion in the reproductive organs.

ZEN is closely associated with infertility, decreased milk production and hyperestrogenism [17]. Cortinovis et al. [16] reported that ZEN directly affect ovarian cells and alter oocyte maturation under in vitro conditions; conversely, under in vivo conditions, this mycotoxin affected ovulation and puberty onset and caused morphological and functional disorders. T-2 toxin (T-2) causes cutaneous lesions in the mount and intestinal membrane and reduces egg production in poultry [18].

Advertisement

5. Mycotoxins and mycotoxin-producing conditions

Fusarium mycotoxins are very common worldwide. They exist in many plants and in various compositions. The major Fusarium mycotoxins are FB1, trichothecenes [e.g. DON, nivalenol (NIV), T-2 and ZEN] [19, 20, 21]. The most important species that is common in Europe is F. graminearum. In the past, Fusarium genus members were mostly not considered as pathogens in the field. However, F. proliferatum and F. verticillioides are of great importance as the main producers of the most dangerous Fusarium mycotoxins [22, 23]. Worldwide mycotoxin occurrence in maize and wheat/bran samples with their median and maximum levels were given in Figure 1 [24, 25].

Figure 1.

Worldwide mycotoxin occurrence (μg/kg) in maize and wheat/bran samples (A, C: Median of positive samples; B, D: Maximum levels) [24, 25].

Shi et al. [5] evaluated the mycotoxins from 20 of the most common Fusarium species and sorted them into the following three groups based on their molecular characterization (Figure 2). Group-1 comprised fusaric acid producers and was further divided into two subgroups. Subgroup-I comprised F. fujikuroi, F. solani, F. verticillioides and F. proliferatum that produce fusaric acid and fumonisins; subgroup-II comprised F. musae, F. equiseti, F. temperatum, F. subglutinans, F. tricinctum, F. oxysporum, F. concentricum, F. sacchari and F. andiyazi that produce only fusaric acid. According to the classification of Fusarium mycotoxins, type-A trichothecene producers comprising F. polyphialidicum, F. sporotrichioides and F. langsethiae formed the Group-II, and type-B trichothecene producers comprising F. meridionale, F. culmorum, F. graminearum and F. poae formed Group-III.

Figure 2.

Mycotoxin classification according main four Fusarium species (A) F. proliferatum (B) F. langsethiae (C) F. graminearum and (D) in PDA, rice and maize medium [5].

In the presence of Fusarium species in plants, the contamination with fumonisins was shown in wheat [26], garlic [27], and asparagus [28]. The most affected plants, that is, maize, beans, soybean [29], rice [30], and sorghum [31] were specifically infected by Gibberella fujikuroi species complex (F. proliferatum, F. verticillioides and F. andiyazi) [29, 32].

Guidance values for Fusarium mycotoxins were set in Commission Recommendation 2006/576/EC [33]. Recommended values for the Fusarium mycotoxins DON, ZEA and fumonisins were set in “Commission Recommendation 2006/576/EC” [33]. For T-2 and HT-2 toxin, indicative levels for cereals and cereals products were set in “Commission Recommendation 2013/165/EU” [34]. Maximum limits for DON, ZEA, fumonisins, T-2 and HT-2 toxins have been set for cereals and by-products according to the production technology used [35].

Advertisement

6. Fungal resistance

Resistance of Fusarium to antifungal drugs has been defined by many researchers. It is known that many FSSC members cause fusarial onychomycosis [36]. F. solani showed more resistance to antifungal agents than others [37]. The effect of azole antifungals used clinically is depend on a particular site, lanosterol-14α-demethylase. While imidazole or triazole rings are important for conferring the therapeutic effect in animals, epoxiconazole, propiconazole, difenoconazole, bromuconazole and tebuconazole are used for plants. Fusarium spp. are resistant to azoles [38].

Tupaki-Sreepurna et al. [39] reported that FSSC members, mainly F. falciforme and F. keratoplasticum, showed multi-drug resistance against caspofungin and azoles. Only a few antifungal agents (voriconazole, posaconazole and amphotericin B) showed in-vitro activity against F. falciforme and F. keratoplasticum [40].

Conversely, the echinocandins are lipopeptide molecules which effectively work by inhibiting 1,3-β-D-glucan synthase of the fungal membrane. If a change occurs in the amino acid residues of β-1,3-glucan synthase enzyme subunits (FKS subunits) in the treatment process, it may lead to increased drug resistance [41, 42].

Polyenes, which are fungicidal, are known as amphipathic drugs, such as nystatin and amphotericin-B. The complexes show efficacy via destroying the proton gradient, allowing for the leakage of ions and removal of ergosterol from phospholipids in the membrane, thus causing fungal cell death in the process [43, 44].

Advertisement

7. Plant disease resistance mechanism

Plants, humans and animals give instant response to the pathogen. In animals, this effect is seen as antibody production, while in plants, it is seen in the form of secretion of various proteins, such as defense-related enzymes and pathogenesis-related proteins [45]. Defense-related enzymes are of great importance in the plant disease resistance mechanism. Immunized plants have rich defense-related enzymes that prevent them from suffering large losses.

If a plant is stimulated by a pathogen, early local defense reactions (a local programmed cell death) are followed by systemic responses (signal is transmitted from infected tissue to the whole plant). At the end, overall defense gene expression gets induced. Consequently, signal perception is essential for plants to combat pathogens [46, 47].

Numerous studies have been done on the transporter genes of plants for improved resistance to Fusarium spp. A sucrose transporter gene (IbSWEET10) of the SWEET gene family obtained from the sweet potato line ND98 was tested for this purpose. This overexpression of the gene has been shown to reduce sugar levels and has a potential use to lower carbohydrate levels and increase the resistance of the plant [48].

Advertisement

8. Fungal transporters

Transporters are of great importance in protecting fungi against plant defense compounds. Transporters enable efflux of the plant-originated defense compounds. Although resveratrol (from grape) and camalexin (from Arabidopsis) transport via the transporter BcatrB of Botrytis cinerea, pisatin (from pea) transports by the NhABC1 transporter of F. solani f. sp. pisi, and rishitin (from potato) transports via the GpABC1 transporter of F. sambucinum [4, 49, 50, 51].

Transporters are divided into two major classes: the ATP-binding cassette (ABC) and the major facilitator superfamily (MFS) transporters. ABC transporters are known to be important for resistance against fungal pathogens, particularly for pleiotropic drug resistance or multidrug resistance domains [52]. Although some transporters produce specific or non-specific toxins, some of them show very specific responses to fungicide sensitivity or resistance [53].

Advertisement

9. Identification, control and management

It is possible to identify the genus Fusarium by several methods. On culturing, hyaline, banana-shaped and multicellular macroconidia are very common; however, to identify them at the species level is not easy. Therefore, molecular methods are needed. Some of the most commonly used molecular methods are the genus-specific PCR, 28 s rRNA gene sequencing, sequence-based PCR, multiplex tandem PCR and automated repetitive sequence-based PCR [54].

As a biological control, Ben Amira et al. [55] showed that when Trichoderma harzianum was co-cultured with F. solani, the former happened to have an antagonistic effect in-vitro. Then, they repeated this experiment by inoculating olive tree roots with the same T. harzianum and F. solani combination. They reported that the former showed a mycoparasitic reaction and antagonistic effects on F. solani. Therefore, mycoparasitic fungi, such as T. harzianum may be used as a biocontrol agent against Fusarium.

Notably, agricultural and chemical precautions cannot be completely successful in preventing Fusarium-related diseases in plants [56]. Therefore, synthetic fungicides are not a true approach for preventing the Fusarium-related diseases due to their harmful effects on the ecosystem and environment, and growing disease-resistant species to combat Fusarium-related diseases seems a more sustainable approach. Resolving the concern of plant diseases caused by Fusarium using biological control methods seems to be a more efficient and eco-friendly approach for agricultural products.

References

  1. 1. Lamprecht SC, Tewoldemedhin YT, Botha WJ, Calitz FJ. Fusarium graminearum species complex associated with maize crowns and roots in the KwaZulu-Natal Province of South Africa. Plant Disease. 2011;95:1153-1158. DOI: 10.1094/PDIS-02-11-0083
  2. 2. Chetouhi C, Bonhomme L, Lasserre-Zuber P, Cambon F, Pelletier S, Renou JP, et al. Transcriptome dynamics of a susceptible wheat upon Fusarium head blight reveals that molecular responses to Fusarium graminearum infection fit over the grain development processes. Functional & Integrative Genomics. 2016;16:183-201. DOI: 10.1007/s10142-016-0476-1
  3. 3. McMullen M, Bergstrom G, De Wolf E, Dill-macky R, Hershman D, Shaner G, et al. Fusarium head blight disease cycle, symptoms, and impact on grain yield and quality frequency and magnitude of epidemics since 1997. Plant Disease. 2012;96:1712-1728. DOI: 10.1094/PDIS-03-12-0291-FE©
  4. 4. Coleman JJ. The Fusarium solani species complex: Ubiquitous pathogens of agricultural importance. Molecular Plant Pathology. 2016;17(2):146-58. DOI: 10.1111/mpp.12289
  5. 5. Shi W, Tan Y, Wang S, Gardiner D, De Saeger S, Liao Y, et al. Mycotoxigenic potentials of Fusarium species in various culture matrices revealed by mycotoxin profiling. Toxins (Basel). 2016;9:1-15. DOI: 10.3390/toxins9010006
  6. 6. Thangavelu R, Palaniswami A, Velazhahan R. Mass production of Trichoderma harzianum for managing Fusarium wilt of banana. Agriculture, Ecosystems and Environment. 2004;103:259-263. DOI: 10.1016/J.AGEE.2003.09.026
  7. 7. Stephens AE, Gardiner DM, White RG, Munn AL, Manners JM. Phases of infection and gene expression of Fusarium graminearum during crown rot disease of wheat. Molecular Plant-Microbe Interactions. 2008;21:1571-1581. DOI: 10.1094/MPMI-21-12-1571
  8. 8. Austen B, Mccarthy H, Wilkins B, Smith A, Duncombe A. Fatal disseminated Fusarium infection in acute lymphoblastic leukaemia in complete remission. Journal of Clinical Pathology. 2001;54:488-490. DOI: 10.1136/jcp.54.6.488 Updated
  9. 9. Mayayo E, Pujol I, Guarro J. Experimental pathogenicity of four opportunist Fusarium species in a murine model. Journal of Medical Microbiology. 1999;48:363-366. DOI: 10.1099/00222615-48-4-363
  10. 10. Namboothiri PES, Nair S, Vijayan K, Visweswaran V. Disseminated Fusarium oxysporum neurospinal infection. Indian Journal of Orthopaedics. 2014;48(2):220-222. DOI: 10.4103/0019-5413.128773
  11. 11. Guendouze-Bouchefa N, Madani K, Chibane M, Boulekbache-Makhlouf L, Hauchard D, Kiendrebeogo M, et al. Phenolic compounds, antioxidant and antibacterial activities of three Ericaceae from Algeria. Industrial Crops and Products. 2015;70:459-466. DOI: 10.1016/j.indcrop.2015.03.053
  12. 12. Gauthier GM, Keller NP. Crossover fungal pathogens: The biology and pathogenesis of fungi capable of crossing kingdoms to infect plants and humans. Fungal Genetics and Biology. 2013;61:146-157. DOI: 10.1016/j.fgb.2013.08.016
  13. 13. Bouhet S, Oswald IP. The effects of mycotoxins, fungal food contaminants, on the intestinal epithelial cell-derived innate immune response. Veterinary Immunology and Immunopathology. 2005;108:199-209. DOI: 10.1016/j.vetimm.2005.08.010
  14. 14. Schenk M, Mueller C. The mucosal immune system at the gastrointestinal barrier. Best Practice & Research. Clinical Gastroenterology. 2008;22:391-409. DOI: 10.1016/j.bpg.2007.11.002
  15. 15. Devreese M, De Backer P, Croubels S. Overview of the most important mycotoxins for the pig and poultry husbandry. Vlaams Diergeneeskd Tijdschr 2013;82:171-180
  16. 16. Cortinovis C, Pizzo F, Spicer LJ, Caloni F. Fusarium mycotoxins: Effects on reproductive function in domestic animals—A review. Theriogenology. 2013;80:557-564. DOI: 10.1016/j.theriogenology.2013.06.018
  17. 17. Minervini F, Aquila MED. Zearalenone and reproductive function in farm animals. International Journal of Molecular Sciences. 2008;9:2570-2584. DOI: 10.3390/ijms9122570
  18. 18. Weber M, Balogh K, Fodor J, Erdélyi M, Ancsin Z, Mézes M. Effect of T-2 and HT-2 toxin during the growing period on body weight, lipid peroxide and glutathione redox status of broiler chickens. Acta Veterinaria. 2010;79:27-31. DOI: 10.2754/avb201079010027
  19. 19. Antonissen G, Martel A, Pasmans F, Ducatelle R, Verbrugghe E, Vandenbroucke V, et al. The impact of Fusarium mycotoxins on human and animal host susceptibility to infectious diseases. Toxins (Basel). 2014;6:430-452. DOI: 10.3390/toxins6020430
  20. 20. Quarta A, Mita G, Haidukowski M, Santino A, Mulè G, Visconti A. Assessment of trichothecene chemotypes of Fusarium culmorum occurring in Europe. Food Additives and Contaminants. 2005;22:309-315. DOI: 10.1080/02652030500058361
  21. 21. Tian Y, Tan Y, Liu N, Liao Y, Sun C, Wang S, et al. Functional agents to biologically control deoxynivalenol contamination in cereal grains. Frontiers in Microbiology. 2016;7:1-8.DOI: 10.3389/fmicb.2016.00395
  22. 22. Marcinkowska J. Fungi occurrence on seeds of field pea. Acta Mycologica. 2013;43:77-89. DOI: 10.5586/am.2008.010
  23. 23. Ozgonen H, Gulcu M. Determination of mycoflora of pea (Pisum sativum) seeds and the effects of Rhizobium leguminosorum on fungal pathogens of peas. African Journal of Biotechnology. 2011;10:6235-6240. DOI: 10.5897/AJB10.2691
  24. 24. Cheli F, Battaglia D, Gallo R, Dell’Orto V. EU legislation on cereal safety: An update with a focus on mycotoxins. Food Control. 2014;37:315-325. DOI: 10.1016/j.foodcont.2013.09.059
  25. 25. Binder EM, Tan LM, Chin LJ, Handl J, Richard J. Worldwide occurrence of mycotoxins in commodities, feeds and feed ingredients. Animal Feed Science and Technology. 2007;137:265-282. DOI: 10.1016/j.anifeedsci.2007.06.005
  26. 26. Chehri K, Jahromi ST, Reddy KRN, Abbasi S, Salleh B. Occurrence of Fusarium spp. and fumonisins in stored wheat grains marketed in Iran. Toxins (Basel). 2010;2:2816-2823. DOI: 10.3390/toxins2122816
  27. 27. Llamas DP, Patón LG, Díaz MG, Serna JG, Sáez SB. The effects of storage duration, temperature and cultivar on the severity of garlic clove rot caused by Fusarium proliferatum. Postharvest Biology and Technology. 2013;78:34-39. DOI: 10.1016/j.postharvbio.2012.12.003
  28. 28. Waśkiewicz A, Irzykowska L, Bocianowski J, Karolewski Z, Kostecki M, Weber Z, et al. Occurrence of Fusarium fungi and mycotoxins in marketable asparagus spears. The Polish Journal of Environmental. 2010;19:219-225
  29. 29. Hsuan HM, Salleh B, Zakaria L. Molecular identification of Fusarium species in Gibberella fujikuroi species complex from rice, sugarcane and maize from peninsular Malaysia. International Journal of Molecular Sciences. 2011;12:6722-6732. DOI: 10.3390/ijms12106722
  30. 30. Gomes LB, Ward TJ, Badiale-Furlong E, Del Ponte EM. Species composition, toxigenic potential and pathogenicity of Fusarium graminearum species complex isolates from southern Brazilian rice. Plant Pathology. 2015;64:980-987. DOI: 10.1111/ppa.12332
  31. 31. Das IK, Audilakshmi S, Annapurna A, Kannababu N, Patil JV. Relationship among seed germination and other characters associated with fusarium grain mould disease in sorghum (Sorghum bicolor L. Moench) using path coefficient analysis. Canadian Journal of Plant Pathology. 2012;34:203-212. DOI: 10.1080/07060661.2012.689260
  32. 32. Mukanga M, Derera J, Tongoona P, Laing MD. Farmers ’ perceptions and management of maize ear rots and their implications for breeding for resistance. African Journal of Agricultural Research. 2011;6:4544-4554. DOI: 10.5897/AJAR11.451
  33. 33. European Commission. Commission recommendation no 2006/576 of 17 August 2006 on the presence of deoxynivalenol, zearalenone, ochratoxin a, T-2 and HT-2 and fumonisins in products intended for animal feeding. Official Journal of the European Union, L. 2006;229:7-9
  34. 34. European Commission. Commission recommendation no 2013/165/EU of 27 March 2013 on the presence of T-2 and HT-2 toxin in cereals and cereal products. Official Journal of the European Union, L. 2013;91:12-15
  35. 35. European Commission. Commission regulation (EU) 1881/2006 of 19 December 2006 setting maximum levels for certain contaminants in foodstuffs. Official Journal of the European Union. L. 2006;364:5-24
  36. 36. van Diepeningen AD, de Hoog GS. Challenges in Fusarium, a trans-kingdom pathogen. Mycopathologia. 2016;181:161-163. DOI:10.1007/s11046-016-9993-7
  37. 37. Azor M, Gené J, Cano J, Guarro J. Universal in vitro antifungal resistance of genetic clades of the Fusarium solani species complex. Antimicrobial Agents and Chemotherapy. 2007;51:1500-1503. DOI: 10.1128/AAC.01618-06
  38. 38. Al-Hatmi AMS, Meis JF, de Hoog GS. Fusarium: Molecular diversity and ıntrinsic drug resistance. PLoS Pathogens. 2016;12:1-8. DOI:10.1371/journal.ppat.1005464
  39. 39. Tupaki-Sreepurna A, Al-Hatmi AMS, Kindo AJ, Sundaram M, de Hoog GS. Multidrug-resistant Fusarium in keratitis: A clinico-mycological study of keratitis infections in Chennai, India. Mycoses. 2017;60:230-233. DOI: 10.1111/myc.12578
  40. 40. Guevara-Suarez M, Cano-Lira JF, Cepero de García MC, Sopo L, De Bedout C, Cano LE, et al. Genotyping of Fusarium isolates from onychomycoses in Colombia: Detection of two new species within the Fusarium solani species complex and in vitro antifungal susceptibility testing. Mycopathologia. 2016;181:165-174. DOI: 10.1007/s11046-016-9983-9
  41. 41. Perlin DS. Current perspectives on echinocandin class drugs. Acta Mycologica. 2018;6:19-20. DOI: 10.2217/fmb.11.19
  42. 42. Parente-Rocha JA, Bailão AM, Amaral AC, Taborda CP, Paccez JD, Borges CL, et al. Antifungal resistance, metabolic routes as drug targets, and new antifungal agents: An overview about endemic dimorphic fungi. Mediators of Inflammation. 2017;2017: 9870679. DOI: 10.1155/2017/9870679
  43. 43. Anderson TM, Clay MC, Cioffi AG, Diaz KA, Hisao GS, Tuttle MD, et al. Amphotericin forms an extramembranous and fungicidal sterol sponge. Nature Chemical Biology. 2014;10:400-406. DOI: 10.1038/nchembio.1496
  44. 44. Gruszecki WI, Gagoś M, Hereć M, Kernen P. Organization of antibiotic amphotericin B in model lipid membranes. A mini review. Cellular & Molecular Biology Letters. 2003;8:161-170
  45. 45. Thakker JN, Patel S, Dhandhukia PC. Induction of defense-related enzymes in banana plants : Effect of live and dead pathogenic strain of Fusarium oxysporum f. Sp. Cubense. 2013;2013. DOI: 10.1016/j.agee.2003.09.026
  46. 46. Nürnberger TSD. Signal transmission in the plant immune response. Trends in Plant Science. 2018;6:372-379
  47. 47. Thakker JN, Patel S, Dhandhukia PC. Induction of defense-related enzymes in banana plants: Effect of live and dead pathogenic strain of Fusarium oxysporum f. Sp. Cubense., ISRN Biotechnology. 2013;2013:1-6. DOI: 10.5402/2013/601303
  48. 48. Li Y, Wang Y, Zhang H, Zhang Q, Zhai H, Liu Q, et al. The plasma membrane-localized sucrose transporter IbSWEET10 contributes to the resistance of sweet potato to Fusarium oxysporum. Frontiers in Plant Science. 2017;8:1-15. DOI: 10.3389/fpls.2017.00197
  49. 49. Wang Y, Lim L, DiGuistini S, Robertson G, Bohlmann J, Breuil C. A specialized ABC efflux transporter GcABC-G1 confers monoterpene resistance to Grosmannia clavigera, a bark beetle-associated fungal pathogen of pine trees. The New Phytologist. 2013;197:886-898. DOI: 10.1111/nph.12063
  50. 50. Stefanato FL, Abou-Mansour E, Buchala A, Kretschmer M, Mosbach A, Hahn M, et al. The ABC transporter BcatrB from Botrytis cinerea exports camalexin and is a virulence factor on Arabidopsis thaliana. The Plant Journal. 2009;58:499-510. DOI: 10.1111/j.1365-313X.2009.03794.x
  51. 51. Schoonbeek H, Del Sorbo G, De Waard MA. The ABC transporter BcatrB affects the sensitivity of Botrytis cinerea to the Phytoalexin resveratrol and the fungicide Fenpiclonil. Molecular Plant-Microbe Interactions. 2001;14:562-571. DOI: 10.1094/MPMI.2001.14.4.562
  52. 52. Kretschmer M, Leroch M, Mosbach A, Walker A-S, Fillinger S, Mernke D, et al. Fungicide-driven evolution and molecular basis of multidrug resistance in field populations of the grey mould fungus Botrytis cinerea. PLoS Pathogens. 2009;5:e1000696. DOI: 10.1371/journal.ppat.1000696
  53. 53. Del Sorbo G, Schoonbeek H, De Waard MA. Fungal transporters involved in efflux of natural toxic compounds and fungicides. Fungal Genetics and Biology. 2000;30:1-15. DOI: 10.1006/fgbi.2000.1206
  54. 54. Tortorano AM, Richardson M, Roilides E, van Diepeningen A, Caira M, Munoz P, et al. ESCMID and ECMM joint guidelines on diagnosis and management of hyalohyphomycosis: Fusarium spp., Scedosporium spp. and others. Clinical Microbiology and Infection. 2014;20:27-46. DOI: 10.1111/1469-0691.12465
  55. 55. Ben Amira M, Lopez D, Triki Mohamed A, Khouaja A, Chaar H, Fumanal B, et al. Beneficial effect of Trichoderma harzianum strain Ths97 in biocontrolling Fusarium solani causal agent of root rot disease in olive trees. Biological Control. 2017;110:70-78. DOI: 10.1016/j.biocontrol.2017.04.008
  56. 56. Lehoczki-Krsjak S, Szabó-Hevér Á, Tóth B, Kótai C, Bartók T, Varga M, et al. Prevention of Fusarium mycotoxin contamination by breeding and fungicide application to wheat. Food Addit Contam Part A. 2010;27:616-628. DOI: 10.1080/19440041003606144

Written By

Tulin Askun

Submitted: 19 July 2017 Published: 25 July 2018