Open access peer-reviewed chapter

Mycovirus Containing Aspergillus flavus and Acute Lymphoblastic Leukemia: Carcinogenesis beyond Mycotoxin Production

Written By

Cameron K. Tebbi, Ioly Kotta-Loizou and Robert H.A. Coutts

Submitted: 29 May 2021 Reviewed: 14 June 2021 Published: 20 July 2021

DOI: 10.5772/intechopen.98897

From the Edited Volume

The Genus Aspergillus - Pathogenicity, Mycotoxin Production and Industrial Applications

Edited by Mehdi Razzaghi-Abyaneh and Mahendra Rai

Chapter metrics overview

331 Chapter Downloads

View Full Metrics

Abstract

Carcinogenic effects of Aspergillus spp. have been well established and generally attributed to a variety of mycotoxin productions, particularly aflatoxins. It is known that most carcinogenic mycotoxins, with the exception of fumonisins, are genotoxic and mutagenic, causing chromosomal aberrations, micronuclei, DNA single-strand breaks, sister chromatid exchange, unscheduled DNA synthesis etc. Some Aspergillus spp. are infected with mycoviruses which can result in loss of aflatoxin production. The effects of mycovirus containing Aspergillus on human health have not been fully evaluated. Recent studies in patients with acute lymphoblastic leukemia, in full remission, have revealed the existence of antibody to the products of a certain Aspergillus flavus isolate which harbored an unknown mycovirus. Exposure of blood mononuclear cells from these patients, but not controls, to the products of this organism had reproduced cell surface phenotypes and genetic markers, characteristic of acute lymphoblastic leukemia. Carcinogenic effects of Aspergillus spp. may not always be mycotoxin related and this requires further investigation.

Keywords

  • Acute lymphoblastic leukemia
  • Mycovirus
  • Aspergillus
  • Cancer
  • Etiology
  • Leukemogenesis
  • Carcinogenesis
  • Virus
  • Mycotoxin

1. Introduction

With a worldwide distribution and a significant level of genetic diversity, fungi are of importance in both medical and agricultural fields and represent major health and commercial concerns. Medically, fungal organisms can be a part of the normal flora of humans and animals. However, these also have the potential to cause mild to severe life-threatening invasive infections or toxicities. The immune response to fungal agents is variable and complex, ranging from lack of recognition to severe inflammatory reactions resulting in significant morbidity and mortality [1, 2, 3, 4, 5, 6].

There is a broad and diverse spectrum of human and animal diseases attributed to fungi. Major effects of fungal agents in human health include, but are not limited to, organ-specific and systemic infections, especially in immunocompromised individuals, toxicity emanating from fungal products, carcinogenicity, mutagenicity, growth impairment and stimulation of allergic reactions. Common and usually non-life-threatening infections caused by fungal agents affecting humans are well recognized and often localized on nails, skin, oral cavity, throat and vagina. Severe and fatal infections, however, can be caused by a variety of fungi including Aspergillus, Blastomyces, Candida, Coccidioides, Cryptococcus, Histoplasma, Mucoromycetes, Pneumocystis, Talaromyces, etc. Despite the significance of fungal infections an understanding of their pathophysiology has lagged behind other human pathogens. While the immune system of healthy individuals, in general, can effectively prevent some fungal infections, this is not the case in immunosuppressed patients [7, 8].

In addition to causing direct infections, the products of some fungal organisms can be toxic to animals and humans. Also, the mycobiome has been implicated in the pathogenesis of various types of cancers. An example is the link between Malassezia spp. and development of pancreatic ductal adenocarcinoma (PDA) [9]. Based on a reported murine experiment, fungal migration from the intestinal lumen to the pancreas initiates the pathogenesis of PDA by driving the complement cascade through the activation of mannose-binding lectin (MBL) [10]. Another example is the carcinogenic potential of Candida spp. Some findings indicate that Candida albicans is capable of promoting cancer by several mechanisms, including production of carcinogenic byproducts, inflammation, induction of T helper type 17 (Th17) cell response and molecular imitations [10, 11, 12]. As will be discussed later in this article, possible relationships between fungal agents and hematological malignancies have been explored.

In light of the above, here the well-established significance of mycotoxins in carcinogenesis is discussed and novel findings illustrating that mycovirus infections may also play a role in human diseases is highlighted. In particular, focus is placed on a mycovirus containing Aspergillus flavus and its effects on leukemogenesis.

Advertisement

2. Mycotoxins

The toxicity, mutagenic and carcinogenic effects of some fungi is often attributed to their production of mycotoxins. Mycotoxins are low molecular weight metabolites produced by yeasts and filamentous fungi. These metabolites are heterogeneous chemicals, toxic to vertebrates, including humans. Several mycotoxins also have toxicities to invertebrates, plants, and other microorganisms [13, 14].

Currently, there are over 450 known mycotoxins, which along with their secondary metabolites, can produce varying degrees of toxicity ranging from mild gastrointestinal symptoms to cancer. A large number of common mycotoxins have been identified that are of major concern to human health, among which are aflatoxins, fumonisins, ochratoxins, patulin, zearalenone and nivalenol/deoxynivalenol. Some organisms can produce several different mycotoxins, and many different species may produce the same mycotoxins. Mycotoxin producing fungi are usually found in improperly saved edibles and agricultural commodities. They can enter and contaminate human and animal food supplies. Animals fed contaminated foods can pass aflatoxins through their eggs, milk, and meats, thus indirectly transmitting aflatoxins to humans [15, 16]. While toxicity in humans is often due to ingestion of large doses of mycotoxins, these can also permeate through the skin [17].

Many mycotoxins are cytotoxic and suppress the functions of lymphocytes, granulocytes, and monocytes. Exposure to some mycotoxins inhibits interferon gamma producing Th1 cells and results in decreased number of these cells. Mycotoxins may lead to T cell polarization toward the Th2 phenotype and is a risk factor for the development of allergies [18, 19, 20, 21, 22, 23]. The principal function of Th1 cells is cell-mediated immunity and inflammation. In normal conditions, there is a balance between Th1 and Th2 cells. A shift of such a balance results in various disorders. Th1 cells play an important role in the functions of immunity related cells such as macrophages, B cells, and cytotoxic CD8+ T lymphocytes (CTLs). The latter stimulate cellular immune response, participate in the inhibition of the activation of macrophages and invigorate B cells to produce IgM and IgG1. For instance, it is found that T cells of children exposed to Aspergillus have significantly lower Th1 cytokines, including tumor necrosis factors (TNFs), interferon-γ, interleukin-2 and -10. These cytokines are involved in the development of CTLs and natural killer (NK) cells which are responsible for the cell-mediated immune response against viruses and detection and removal of tumor cells. Thus, exposure to fungal agents may significantly change cellular composition and cytokine production and immune function [24, 25].

Exposure to aflatoxins can lead to life threatening acute poisoning (aflatoxicosis) [26]. In turn, acute aflatoxicosis can result in acute hepatic necrosis often manifested by symptoms of liver failure [27]. This eventually may cause development of cirrhosis in the liver and hepatic carcinoma. Chronic low-level exposure to mycotoxins, particularly aflatoxins and especially aflatoxin B1, is known to be associated with increased risk of hepatic damage, liver and gallbladder cancer and impaired immune activity [27, 28, 29]. Several studies have documented liver and gallbladder toxicity and carcinogenicity related to mycotoxins. Other organs, including bones, kidneys, pancreas, bladder, viscera and central nervous system, can be subject to carcinogenesis [30].

A variety of mycotoxins have carcinogenic potential in animals and humans [16, 17, 26, 28, 31, 32, 33, 34, 35]. Certain mycotoxins, especially aflatoxins, produced by genetically diverse Aspergillus spp. including A. fumigatus, A. parasiticus and A. flavus can be genotoxic with damage to DNA, which is attributed to the development of cancer in animals and humans. The effects of aflatoxins B1, B2, G1 and G2 and their metabolites such as aflatoxins M1, M2a, P1, Q1, Q2a, R0, H1; B2a, M2; GM1, GM2a, parasiticol (B3) and GM2, produced by the Aspergillus spp., are well recognized [35].

The carcinogenesis of mycotoxins is reported to be due to the intercalation of aflatoxin metabolites into DNA which alkylate the bases through epoxide moiety. This can be as a result of the mutations in the p53 gene or signaling apoptosis. The third base of codon 249 of the p53 gene is reported to be more susceptible to aflatoxin-mediated mutations. For example, in hepatocellular carcinoma, upon exposure to aflatoxin, mutation of p53 gene is fixed at codon 249 third base and take the form of G to T transversion [36, 37].

In one report, using a mammalian cell line, the mutagenicity of various mycotoxins and the efficiency of mutagenic mycotoxins in producing DNA single strand breaks and chromosome aberrations were investigated. These experiments revealed that aflatoxin B1, mycophenolic acid, patulin, penicillic acid, and sterigmatocystin induce 8-azaguanine-resistant mutations. At higher concentrations, aflatoxin B1, mycophenolic acid, and sterigmatocystin were found to have minimal effects on single-stranded DNA. In contrast, treatment with patulin and penicillic acid at higher concentrations had resulted in severe breaks. Chaetoglobosin B, fusarenon X, luteoskyrin, and ochratoxin A had not induced 8-azaguanine-resistant mutations [38].

Overall, the mutagenicity of mycotoxins varies significantly and depends on their efficiency in causing DNA single-strand breaks, resulting in chromosomal aberrations. Adults are believed to have a higher tolerance to mycotoxins but exposure of children, while controversial and not uniformly accepted, can lead to delayed development and stunted growth [16, 31, 32, 33].

In addition to laboratory-based experiments, reports regarding isolation of mycotoxin producing strains of fungi, including that of A. flavus, from the residences of leukemia patients are available [39, 40, 41, 42]. In many reports, except for recent publications, fungal carcinogenesis is attributed to mycotoxins and their immunosuppressive effects. One report describes examination of sera from 36 cancer patients against an aflatoxin producing A. flavus which was isolated from the home of a patient with leukemia. A modified microimmunodiffiusion technique was used for this immunological evaluation. This study had found that 30% of cancer patients, 15 of whom had leukemia or lymphoid malignancy, and 6% of controls had shown a precipitation band indicating positive results [39]. Another published article reports four leukemic patients, from three families, in a residence where a mycotoxin producing fungus was isolated. The leukemogenesis was attributed to the immune depressive effects of mycotoxins [41]. In a house where a husband and wife had developed acute myelomonocytic and undifferentiated leukemia, respectively, fungal surveyance of the residence had been performed. Three fungal isolates were found, an extract of which had shown a depressive effect on a phytohemagglutinin skin test in guinea pigs as compared to negative findings using extracts isolated from a control residence [40]. As described below, a significant amount of data regarding the correlation of a mycovirus containing A. flavus, isolated from the home of a patient with acute lymphoblastic leukemia, has been recently published.

Advertisement

3. Viruses and human cancer

A vast amount of data on several viruses and their possible association with cancer development has been published [43, 44, 45, 46, 47, 48, 49, 50, 51, 52]. While not the focus of this article, a brief review of the subject reveals the importance of the study of viral agents and their relation to occurrence of malignant disorders. Both DNA and RNA viruses are capable of causing cancer in humans. Some of the known DNA viruses that are capable of causing human cancers are Epstein-Barr (EB) virus, human papilloma virus, hepatitis B virus, and human herpes virus 8. The relationship of EB virus to the development of Burkitt’s lymphoma and nasopharyngeal carcinoma is well established [53, 54, 55, 56, 57, 58, 59]. Likewise, the relation of human papilloma virus and the development of cervical cancer and retention of HPV viral oncoproteins E6 and E7 for their continued expression and proliferation has been demonstrated [60, 61, 62, 63]. Human T lymphotropic virus type 1, human immunodeficiency virus (HIV) and hepatitis C viruse are some of the RNA viruses that contribute to human cancers. It appears that viruses have diverse biological pathways to malignant disorders. The presence of viral gene products in cancer and precancerous cells are known. Despite the well-known carcinogenic role of viruses, little data regarding any possible health effects of mycoviruses alone, or in conjunction with their host, are available. This area needs to be further explored.

Advertisement

4. Mycoviruses

Viruses that infect fungi, also known as mycoviruses (myco = ‘fungus’ in Greek), are widespread geographically and are expected to infect all fungal taxa, from early divergent lineages to the most well-studied ascomycetes (sac fungi) and basidiomycetes (mushrooms). Mycovirus infection is persistent but does not result in disease or death of the host fungus, and often does not lead to obvious alterations in its phenotype under controlled laboratory conditions; therefore, mycovirology is an underappreciated and understudied field, similar to all non-disease associated virology [64].

Mycoviruses are currently classified in 22 taxa (21 families and one genus) by the International Committee on Taxonomy of Viruses (ICTV; https://talk.ictvonline.org/) (Figure 1). Some of these taxa exclusively accommodate viruses infecting fungi, such as the families Hypoviridae and Polymycoviridae. Other taxa also accommodate viruses infecting protozoa, plants, insects and mammals, such as the families Botourmiaviridae, Chrysoviridae, Partitiviridae, Reoviridae and Totiviridae. Members of the DNA-containing Genomoviridae family have been discovered in sequencing data from a variety of samples, including plant and insect tissue, animal blood, serum and feces, human blood, plasma, cerebrospinal fluid, cervical biopsies, and feces, and sewage [65]. Mycoviruses may be closely related to viruses pathogenic for humans. For instance, family Mymonaviridae belongs to the order Mononegavirales, as are viruses that cause Ebola, measles, mumps, rabies and respiratory diseases. Families Metaviridae and Pseudoviridae belong to order Ortervirales, together with human immunodeficiency virus (HIV), cause of acquired immunodeficiency syndrome (AIDS), and other retroviruses.

Figure 1.

Current classification of mycoviruses according to the International Committee on Taxonomy of Viruses. The realms Riboviria and Monodnaviria accommodate viruses with respectively RNA and DNA genomes. Underlying family names accommodate mycoviruses known to infect Aspergillus spp. Next to family/genus names, (+)SS, (−)SS and DS indicate respectively, positive-sense single-stranded, negative-sense single-stranded and double-stranded genomes; hexagons indicate the presence of true virions, either isometric, bacilliform of filamentous.

Classification of exemplar mycoviruses known to infect Aspergillus spp is shown in Figure 2.

Figure 2.

Classification of exemplar mycoviruses known to infect Aspergillus spp. Not all known mycoviruses found in Aspergillus spp. are officially assigned to recognized taxa. The phenotypes and effects of the majority of these mycoviruses on their Aspergillus host is unknown.

Almost all known mycoviruses have double stranded (ds) RNA genomes or single stranded (ss) RNA genomes, either positive sense or negative sense, with one family of mycoviruses having circular ssDNA genomes. Virions are often proteinaceous in nature, composed of virus capsid proteins and their structure may range from spherical, to bacilliform in the case of barnaviruses, to filamentous in the case of flexiviruses and mymonaviruses. The absence of true virions is also common: narnaviruses and mitoviruses exist as naked RNA molecules respectively in the cytoplasm and mitochondria, hypoviruses are encapsulated in host derived lipid vesicles, polymycoviruses are non-conventionally encapsidated by a viral protein [66, 67]. Mycoviruses move intracellularly within the infected fungus and spread in mycelia during cell division and growth. Almost all known mycoviruses lack an extracellular phase in their replication cycle; they are transmitted vertically during asexual and/or sexual spore production and horizontally between fungal strains following cell fusion. The absence of an extracellular phase explains the general lack of lipid envelopes in virions.

Early reports focused on the mycovirus-mediated alterations on fungal phenotype, including morphology, pigmentation, asexual and sexual sporulation, and growth. Production of viral toxins conferring a competitive advantage to the fungal host [68], clearly illustrate that viral infection can be beneficial to the host and viruses are undeserving of their name, derived from the Latin word for ‘poison’ or ‘venom’. These killer yeast systems have been primarily studied in the eukaryotic model organism Saccharomyces cerevisiae [69], extensively used in biotechnological applications such as baking, brewing and winemaking. However, interest in mycoviruses stems mainly from their effects on the interaction between their host fungus and the plant, insect or mammalian/human host of the fungus.

An increasing number of studies clearly illustrate the importance of mycoviruses in host-microbe interactions. The discovery of ‘transmissible hypovirulence’, i.e., mycovirus-mediated decrease in fungal pathogenicity represents a major advance in the field and the first mycovirus-based biological control application to combat chestnut blight caused by the plant pathogen Cryphonectria parasitica [70, 71]. The opposite phenomenon called hypervirulence, i.e., mycovirus-mediated increase in fungal pathogenicity, has also been noted. For instance, two variants of Aspergillus fumigatus polymycovirus 1 (AfuPmV-1), the first virus demonstrated to be infectious as dsRNA [66], respectively cause hypovirulence in an immunosuppressed mouse infection model [72] and hypervirulence in the greater wax moth G. mellonella infection model [73]. Additionally, AfuPmV-1 renders its fungal host more sensitive to the bacterium Pseudomonas aeruginosa [74]. Furthermore, partitivirus infection of Talaromyces marneffei leads to hypervirulence in a BALB/c mouse model [75]. Mycoviruses dsRNA genomes or replication intermediates are recognized by Toll-like receptor 3 (TLR-3) [76] and may induce an interferon immune response in a TLR-3 dependent or independent manner, as illustrated with totivirus infected Malassezia [77, 78]. A link between azole resistance and mycovirus infection has been noted in Penicillium digitatum [79]. Finally, mycovirus infection is known to be responsible for modulation of fungal toxins and this phenomenon has been studied mainly in Aspergillus spp [80]. Carcinogenic aflatoxin production may be repressed by the presence of a mycovirus in A. flavus [81, 82, 83, 84], while ochratoxin A is enhanced by the presence of a partitivirus in A. ochraceus [85].

Currently most mycovirus studies are focused on economically important phytopathogenic fungi, while scant data regarding fungi containing mycoviruses and human disorders are available. Since mycoviruses do exist in fungi, and humans are exposed to them, further research on these organisms may expand our knowledge of their possible role and effects of their interaction with humans.

Advertisement

5. Studies of mycovirus containing Aspergillus flavus

A report describing plasma of patients with acute lymphoblastic leukemia (ALL) having a positive reaction to an A. flavus isolate containing an unknown mycovirus is available [86]. Exposure of the peripheral blood mononuclear cells (PBMCs) obtained from a group of ALL patients who were in a complete remission to the culture of this organism was reported to reproduce genetic and cell surface phenotypes, characteristic of active ALL [87]. Conversely, this was not observed in the control group of patients [87]. To describe these findings (which are patented) in more detail, in a series of experiments, a mycovirus infected A. flavus separated from the home of a patient with B-cell ALL was found to contain unknown mycovirus particles. These mycovirus particles were found within the body of the organism and culture supernatant. Chemical analysis of the isolated mycovirus containing A. flavus had revealed a lack of aflatoxin production [86]. The latter may be due to the influence of the unknown mycovirus which may have caused suppression of the production of aflatoxin as described previously [80, 81, 82, 83, 84]. Utilizing fast protein liquid chromatography (FPLC) for the analysis of the supernatant of the culture of this isolate, three separate peaks were identified. As noted above, in controlled experiments using plasma of patients with ALL in complete remission, with no evidence of the disease, using crude supernatant of the culture of the mycovirus containing A. flavus and enzyme-linked immunosorbent assay (ELISA) for the detection of antibodies, plasma of patients with ALL had reacted positively. The plasma obtained from three separate groups of controls, including normal individuals, patients with sickle cell disease and individuals with various solid tumors, had been negative. In a separate study evaluating peaks obtained by fractionation using FPLC, of the three peaks which were found, peak 1 had the strongest positive effect [86]. The authors suggest that this technique can be used for screening for ALL or a test to identify patients who have had this disease [86].

As noted before, in a related publication, exposure of PBMCs obtained from ALL patients in complete remission, and long-term survivors of this disease, to the supernatant of the culture of the mycovirus containing A. flavus resulted in the re-development of the genetic and cell surface phenotypes, characteristic of ALL. The cell surface phenotypes examined were CD10/CD19, CD19/CD34 and CD34/CD117. The redevelopment of the ALL cell surface phenotypes was reported to be gradual, completed in 24 hours, and remained stable thereafter. Following exposure to the supernatant of the mycovirus containing A. flavus, alterations in gene expression were evaluated using microarray technique. Some of these alterations were reported to be upregulation of JAK1 (12.87-fold), JAK2 (1.5-fold), JAK3 (2.73-fold), IKZF1 (10.12-fold), MCL1 (59.37-fold), MYC (14.19-fold), HDAC1 (26.39-fold) and downregulation of PAX5 (3.05-fold). Following incubation, a significant and robust activation of transcription factor NF-ϰB p65 was reported by immunoblotting in ALL patients without any changes in the controls. The supernatant of the culture of Mycocladus corymbifer, which was used as a negative control, was reported to have no effects on PBMCs either from the ALL or control patients [87]. The above studies suggest a possible role for the mycovirus containing A. flavus in the process of leukemogenesis and opens a venue for vaccination and prevention of this disease.

Advertisement

6. Conclusion

It is apparent that fungal spp. are important in human and animal health. The mechanism of the effects of fungal agents in the development of human diseases appears to be multifaceted. Fungi are widespread in nature and inevitably, humans encounter these organisms. Many fungi contain mycoviruses. Although a significant amount of data regarding the carcinogenic effects of mycotoxins in the development of malignant disorders are available, possible pathogenicity and role of the mycoviruses in fungi, if any, in human and animal health, including malignant disorders, are not known. Recent reports describing in vitro effects of a mycovirus containing A. flavus isolate in redeveloping characteristic ALL cell surface and genetic phenotypes in the PMBCs of acute lymphoblastic leukemia patients in complete remission is of interest. The existence of antibody to this organism in plasma of these patients is intriguing and further indicates its possible role in leukemogenesis. This area needs to be further investigated.

References

  1. 1. Jovanovic, S, Felder-Kennel, A, Gabrio, T, Kouros, B, Link, B, Maisner, V, Piechotowski, I, Schick, K-H, Schrimpf, M, Ursula Weidner, M, Zöllner, I, Schwenk, M. Indoor fungi levels in homes of children with and without allergy history. Int J Hyg Environ Health207:369-378, 2004
  2. 2. Van Burik, J, Magee, PT. Aspects of fungal pathogenesis in humans. Annu Rev Microbiol55:743-772, 2001
  3. 3. McGinnis, MR, Sigler L, Rinaldi MG. Some medically important fungi and their common synonyms and names of uncertain application. Clin Infect Dis29:728-730, 1999
  4. 4. Sternberg, S. The emerging fungal threat. Science266:1632-1634,1994
  5. 5. Barrios, N, Tebbi, CK, Rotstein, C, Siddiqui, S, Humbert, JR. Brainstem invasion by Aspergillus fumigatus in a child with leukemia. NY State J Med88:656-658, 1988
  6. 6. Rotstein, C, Tebbi, CK, Brass, C. Viral, bacterial and fungal infections in adolescent oncology. In Adolescent Oncology, Tebbi CK (Editor), Futura Publishing Company, Mt. Kisco, NY, 1987, 429-506
  7. 7. Brown, GD, Denning, DW, Gow, NA, Levitz, SM, Netea, MG, White, TC. Hidden killers: human fungal infections. Sci Transl Med4:165rv13, 2012
  8. 8. Types of Fungal Diseases. Centers for Disease Control and Prevention, National Center for Emerging and Zoonotic Infectious Diseases (NCEZID), Division of Foodborne, Waterborne, and Environmental Diseases (DFWED), USA, May 6, 2019
  9. 9. Aykut, B, Pushalkar, S, Chen, R, Li Q, Abengozar, R, Kim, JI, Shadaloey, SA, Wu, D, Preiss, P, Verma, N, Guo, Y, Saxena, A, Vardhan, M, Diskin, B, Wang, W, Leinwand, J, Kurz, E, Kochen Rossi, JA, Hundeyin, M, Zambrinis, C, Li, X, Saxena, D, Miller, G. The fungal mycobiome promotes pancreatic oncogenesis via activation of MBL. Nature574:264-267, 2019
  10. 10. Ramirez-Garcia, A, Rementeria, A, Aguirre-Urizar, JM, Moragues, MD, Antoran A, Pellon, A, Abad-Diaz-de-Cerio, A, Hernando, FL. Candida albicans and cancer: Can this yeast induce cancer development or progression? Crit Rev Microbiol42:181-93, 2016
  11. 11. Sankari, SL, Gayathri, K, Balachander, N, Malathi, L. Candida in potentially malignant oral disorders. J Pharm Bioallied Sci7:S162-164, 2015
  12. 12. Nørgaard, M, Thomsen, RW, Farkas, DK, Mogensen, MF, Sørensen, HT. Candida infection and cancer risk: a Danish nationwide cohort study. Eur J Intern Med24:451-455, 2013
  13. 13. Bennett, JW, Klich, M. Mycotoxins. Clin Microbiol Rev16:497-516, 2003
  14. 14. Bennett, JW. Mycotoxins, mycotoxicoses, mycotoxicology and mycopathologia. Mycopathologia100:3-5, 1987
  15. 15. Iqbal, SZ, Nisar, S, Asi, MR, Jinap, S. Natural incidence of aflatoxins, ochratoxin A and zearalenone in chicken meat and eggs. Food Control, 43:98-103, 2014
  16. 16. Khlangwiset P, Shephard GS, Wu F. Aflatoxins and growth impairment: a review. Crit Rev Toxicol41:740-55, 2011
  17. 17. Boonen J, Malysheva SV, Taevernier L, Diana Di Mavungu J, De Saeger S, De Spiegeleer B. Human skin penetration of selected model mycotoxins. Toxicology301:21-32, 2012
  18. 18. Njoroge, SMC, Matumba, L, Kanenga, K, Siambi, M, Waliyar, F, Maruwo, J, Machinjiri, N, Monyo, ES. Aflatoxin B1 levels in groundnut products from local markets in Zambia. Mycotoxin Res 33:113-119, 2017
  19. 19. Lioi, M, Santoro, A, Barbieri, R, Salzano,S, Ursini, M. Ochratoxin A and zearalenone: a comparative study on genotoxic effects and cell death induced in bovine lymphocytes. Mutat Res557:19-27, 2004
  20. 20. Muller, G, Burkert, B, Moller, U, et al. Ochratoxin A and some of its derivatives modulate radical formation of porcine blood monocytes and granulocytes. Toxicology199:251-259, 2004
  21. 21. Muller, G, Rosner, H, Rohrmann, B et al. Effects of the mycotoxin ochratoxin A and some of its metabolites on the human cell line THP-1. Toxicology184: 69-82, 2003
  22. 22. Nielsen, KF, Smedsgaard, J. Fungal metabolite screening: database of 474 mycotoxins and fungal metabolites for dereplication by standardised liquid chromatography–UV–mass spectrometry methodology. J Chromatogr A1002:111-136, 2003
  23. 23. Müller, A, Lehmann, I, Seiffart, A, Diez, U, Wetzig, H, Borte, M, Herbarth, O. Increased incidence of allergic sensitization and respiratory diseases due to mold exposure: Results of the Leipzig Allergy Risk children Study (LARS). Int J Hyg Environ Health204:363-365, 2002
  24. 24. Romagnani S. Lymphokine production by human T cells in disease states. Annu Rev Immunol12:227-257, 2003
  25. 25. Nutt, SL, Huntington, ND. Cytotoxic T lymphocytes and natural killer cells. In Clinical Immunology: Principles and Practice, Rich RR, Fleisher TA, Shearer WT, Schroeder HW, Frew AJ, Weyand CM (Editors), Fifth Edition, Elsevier Publications 2019, 247-259
  26. 26. Barrett JR. Mycotoxins: of molds and maladies. Environ Health Perspect108:A20-A23, 2000
  27. 27. Dhakal, A, Sbar, E. Aflatoxin toxicity. In StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2021
  28. 28. Nogueira, L, Foerster, C, Groopman, J, Egner, P, Koshiol, J, Ferreccio, C. Association of aflatoxin with gallbladder cancer in Chile. JAMA 313:2075-2077, 2015.
  29. 29. Barrett, JR. Liver cancer and aflatoxin: New information from the Kenyan outbreak. Environ Health Perspect113:A837-A838, 2005
  30. 30. Benkerroum, N. Chronic and acute toxicities of aflatoxins: Mechanisms of action. Int. J. Environ. Res. Public Health17:423, 2020
  31. 31. Chen, C, Mitchell, NJ, Gratz, J, Houpt, ER, Gong, Y, Egner, PA, Groopman, JD, Riley, RT, Showker, JL, Svensen, E, Mduma, ER, Patil, CL, Wu, F. Exposure to aflatoxin and fumonisin in children at risk for growth impairment in rural Tanzania. Environ Int 115:29-37, 2018
  32. 32. Mitchell NJ, Hsu HH, Chandyo RK, Shrestha B, Bodhidatta L, Tu YK, Gong YY, Egner PA, Ulak M, Groopman JD, Wu F. Aflatoxin exposure during the first 36 months of life was not associated with impaired growth in Nepalese children: An extension of the MAL-ED study. PLOS ONE 12:e0172124, 2017.
  33. 33. Turner PC, Collinson AC, Cheung YB, Gong Y, Hall AJ, Prentice AM, Wild CP. Aflatoxin exposure in utero causes growth faltering in Gambian infants. Int J Epidemiol 36:1119-1125, 2007.
  34. 34. Williams, JH, Phillips, TD, Jolly, PE, Stiles, JK, Jolly, CM, Aggarwal, D. Human aflatoxicosis in developing countries: a review of toxicology, exposure, potential health consequences, and interventions. Am J Clin Nutr 80:1106-1122, 2004.
  35. 35. Squire, RA. Ranking animal carcinogens: a proposed regulatory approach. Science214:877-880, 1981
  36. 36. Deng ZL, Ma Y. Aflatoxin sufferer and p53 gene mutation in hepatocellular carcinoma. World J Gastroenterol4:28-29, 1998
  37. 37. Aguilar F, Hussain SP, Cerutti P. Aflatoxin B1 induces the transversion of G-->T in codon 249 of the p53 tumor suppressor gene in human hepatocytes. Proc Natl Acad Sci USA90:8586-8590, 1993
  38. 38. Umeda M, Tsutsui T, Saito M. Mutagenicity and inducibility of DNA single-strand breaks and chromosome aberrations by various mycotoxins. Gan68:619-625, 1977
  39. 39. Wray, BB, Harmon, CA, Rushing, EJ, Cole, RJ. Precipitins to an aflatoxin-producing strain of Aspergillus flavus in patients with malignancy. J Cancer Res Clin Oncol103:181-185, 1982
  40. 40. Wray, BB, Rushing, EJ, Boyd, RC, Schindel, AM Suppression of phytohemagglutinin response by fungi from a "leukemia" house. Arch Environ Health34:350-353, 1979
  41. 41. Wray, BB, O’Steen, KG Mycotoxin-producing fungi from house associated with leukemia. Arch Environ Health30:571-573, 1975
  42. 42. McPhedran, P, Heath, CW. Multiple cases of leukemia associated with one house. JAMA209:2021-2025, 1969
  43. 43. Mesri EA, Feitelson MA, Munger K. Human viral oncogenesis: a cancer hallmarks analysis. Cell Host Microbe15:266-282, 2014
  44. 44. Momin B, Richardson L. An analysis of content in comprehensive cancer control clans that address chronic hepatitis B and C virus infections as major risk factors for liver cancer. J Community Health37:912-916, 2012
  45. 45. Snow AN, Laudadio J. Human papilloma virus detection in head and neck squamous cell carcinomas. Adv Anat Pathol17:394-403, 2010
  46. 46. Liao JB. Viruses and human cancer. Yale J Biol Med79:115-122, 2006
  47. 47. Montaner, S, Sodhi, A, Ramsdell, AK, Martin, D, Hu, J, Sawai, ET, Gutkind, JS. The Kaposi's sarcoma-associated herpesvirus G protein-coupled receptor as a therapeutic target for the treatment of Kaposi's sarcoma. Cancer Res66:168-174, 2006
  48. 48. Lehtinen M, Koskela P, Ogmundsdottir HM, Bloigu A, Dillner J, Gudnadottir M, Hakulinen T, Kjartansdottir A, Kvarnung M, Pukkala E, Tulinius H, Lehtinen T. Maternal herpesvirus infections and risk of acute lymphoblastic leukemia in the offspring. Am J Epidemiol 158:207-213, 2003
  49. 49. Sarid R, Olsen SJ, Moore PS. Kaposi's sarcoma-associated herpesvirus: epidemiology, virology, and molecular biology. Adv Virus Res52:139-232, 1999
  50. 50. Flore, O, Rafii, S, Ely, S, O'Leary, JJ, Hyjek, EM, Cesarman, E. Transformation of primary human endothelial cells by Kaposi's sarcoma-associated herpesvirus. Nature394:588-592, 1998
  51. 51. Chang, Y, Cesarman, E, Pessin, MS, Lee, F, Culpepper, J, Knowles, DM, Moore, PS. Identification of herpesvirus-like DNA sequences in AIDS-associated Kaposi's sarcoma. Science266:1865-1869, 1994
  52. 52. Gold, JE, Castella,A, Zalusky, R. B-cell acute lymphoblastic leukemia in HIV antibody-positive patients. J Hematol32:200-204, 1989
  53. 53. Tebbi, CK. Etiology of acute leukemia: A review. Cancers13:2256, 2021
  54. 54. Rowe, M, Fitzsimmons, L, Bell, AI. Epstein-Barr virus and Burkitt lymphoma. Chin J Cancer33:609-619, 2014
  55. 55. Haque T, Wilkie GM, Taylor C, et al. Treatment of Epstein-Barr-virus-positive post-transplantation lymphoproliferative disease with partly HLA-matched allogeneic cytotoxic T cells. Lancet360:436-442, 2002
  56. 56. Gottschalk S, Gottschalk S, Ng CY, Perez M, Smith CA, Sample C, Brenner MK, Heslop HE, Rooney CM. An Epstein-Barr virus deletion mutant associated with fatal lymphoproliferative disease unresponsive to therapy with virus-specific CTLs. Blood 97:835-843, 2001
  57. 57. Papadopoulos EB, Ladanyi, M, Emanuel, D et al. Infusions of donor leukocytes to treat Epstein-Barr virus-associated lymphoproliferative disorders after allogeneic bone marrow transplantation. N Engl J Med 330:1185-1191, 1994
  58. 58. Thorley-Lawson DA, Poodry CA. Identification and isolation of the main component (gp350-gp220) of Epstein-Barr virus responsible for generating neutralizing antibodies in vivo. J Virol43:730-736, 1982
  59. 59. Ho JH. An epidemiologic and clinical study of nasopharyngeal carcinoma. Int J Radiat Oncol Biol Phys4:182-198, 1978
  60. 60. Kaufmann, AM, Stern, PL, Rankin, EM, Sommer, H, Nuessler, V, Schneider, A, Adams, M, Onon, TS, Bauknecht, T, Wagner, U, Kroon, K, Hickling, J, Boswell, CM, Stacey SN, Kitchener, HC, Gillard, J, Wanders, J, Roberts, JS, Zwierzina, H. Safety and immunogenicity of TA-HPV, a recombinant vaccinia virus expressing modified human papillomavirus (HPV)-16 and HPV-18 E6 and E7 genes, in women with progressive cervical cancer. Clin Cancer Res8:3676-3685, 2002
  61. 61. Wallin, KL, Wiklund, F, Angström, T, Bergman, F, Stendahl, U, Wadell, G, Hallmans, G, Dillner, J.Type-specific persistence of human papillomavirus DNA before the development of invasive cervical cancer. N Engl J Med341:1633-1638, 1999
  62. 62. von Knebel Doeberitz, M, Oltersdorf, T, Schwarz, E, Gissmann, L. Correlation of modified human papilloma virus early gene expression with altered growth properties in C4-1 cervical carcinoma cells. Cancer Res48:3780-3786, 1988
  63. 63. Halpert, R, Fruchter, RG, Sedlis, A, Butt, K, Boyce, JG, Sillman, FH. Human papillomavirus and lower genital neoplasia in renal transplant patients. Obstet Gynecol68:251-258, 1986
  64. 64. Kotta-Loizou, I. Mycoviruses: past, present, and future. Viruses11:361, 2019
  65. 65. Krupovic, M, Ghabrial, SA, Jiang, D, Varsani, A. Genomoviridae: a new family of widespread single-stranded DNA viruses. Arch Virol161:2633-2643, 2016
  66. 66. Kanhayuwa, L, Kotta-Loizou, I, Özkan, S, Gunning, AP, Coutts, RHA. A novel mycovirus from Aspergillus fumigatus contains four unique dsRNAs as its genome and is infectious as dsRNA. Proc Natl Acad Sci U S A112:9100-9105, 2015
  67. 67. Kotta-Loizou I, Coutts, RHA. Studies on the virome of the entomopathogenic fungus Beauveria bassiana reveal novel dsRNA elements and mild hypervirulence. PLoS Pathog13:e1006183, 2017
  68. 68. Drinnenberg, IA, Fink, GR, Bartel, DP. Compatibility with killer explains the rise of RNAi-deficient fungi. Science333:1592, 2011
  69. 69. Schmitt, MJ, Breinig, F. Yeast viral killer toxins: lethality and self-protection. Nat Rev Microbiol4:212-221, 2006
  70. 70. Van Alfen, NK, Jaynes, RA, Anagnostakis, SL, Day, PR. Chestnut blight: biological control by transmissible hypovirulence in Endothia parasitica. Science189:890-891, 1975
  71. 71. Anagnostakis, SL. Biological control of chestnut blight. Science215:466-471, 1982
  72. 72. Takahashi-Nakaguchi, A, Shishido, E, Yahara, M, Urayama, SI, Ninomiya, A, Chiba, Y, Sakai, K, Hagiwara, D, Chibana, H, Moriyama, H, Gonoi, T. Phenotypic and molecular biological analysis of polymycovirus AfuPmV-1M from Aspergillus fumigatus: reduced fungal virulence in a mouse infection model. Front Microbiol11:607795, 2020
  73. 73. Özkan, S, Coutts, RHA. Aspergillus fumigatus mycovirus causes mild hypervirulent effect on pathogenicity when tested on Galleria mellonella. Fungal Genet Biol76:20-26, 2015
  74. 74. Nazik, H, Kotta-Loizou, I, Sass, G, Coutts, RHA, Stevens, DA. Virus infection of Aspergillus fumigatus compromises the fungus in intermicrobial competition. Viruses13:686, 2021
  75. 75. Lau, SKP, Lo, GCS, Chow, FWN, Fan, RYY, Cai, JJ, Yuen, KY, Woo, PCY. Novel partitivirus enhances virulence of and causes aberrant gene expression in Talaromyces marneffei. mBio9:e00947-18, 2018
  76. 76. Ives, A, Ronet, C, Prevel, F, Ruzzante, G, Fuertes-Marraco, S, Schutz, F, Zangger, H, Revaz-Breton, M, Lye, LF, Hickerson, SM, Beverley, SM, Acha-Orbea, H, Launois, P, Fasel, N, Masina, S. Leishmania RNA virus controls the severity of mucocutaneous leishmaniasis. Science331:775-778, 2011
  77. 77. Park, M, Cho, YJ, Kim, D, Yang, CS, Lee, SM, Dawson, TL Jr, Nakamizo, S, Kabashima, K, Lee, YW, Jung, WH. A novel virus alters gene expression and vacuolar morphology in Malassezia cells and induces a TLR3-mediated inflammatory immune response. mBio11:e01521-20, 2020
  78. 78. Applen Clancey, S, Ruchti, F, LeibundGut-Landmann, S, Heitman, J, Ianiri, G. A novel mycovirus evokes transcriptional rewiring in the fungus Malassezia and stimulates beta interferon production in macrophages. mBio11:e01534-20, 2020
  79. 79. Niu, Y, Yuan, Y, Mao, J, Yang, Z, Cao, Q, Zhang, T, Wang, S, Liu, D. Characterization of two novel mycoviruses from Penicillium digitatum and the related fungicide resistance analysis. Sci Rep8:5513, 2018
  80. 80. Kotta-Loizou, I, Coutts, RHA. Mycoviruses in Aspergilli: A comprehensive review. Front Microbiol8:1699-1714, 2017
  81. 81. Schmidt, FR. The RNA interference-virus interplay: tools of nature for gene modulation, morphogenesis, evolution and a possible mean for aflatoxin control. Appl Microbiol Biotechnol83:611-615, 2009
  82. 82. Silva, VN, Durigon, EL, de Fátima Costa Pires, M, Lourenço, A, de Faria, MJ, Corrêa, B. Time course of virus-like particles (VLPs) double-stranded RNA accumulation in toxigenic and non-toxigenic strains of Aspergillus flavus. Braz J Microbiol32:56-60, 2001
  83. 83. Schmidt, FR, Lemke, PA, and Esser, K. Viral influences on aflatoxin formation by Aspergillus flavus. Appl Microbiol Biotechnol24:248-252, 1986
  84. 84. Schmidt, FR, Davis, ND, Diener, UL and Lemke, PA. Cycloheximide induction of aflatoxin synthesis in a nontoxigenic strain of Aspergillus flavus. BioTechnology 1:794-795, 1983
  85. 85. Nerva, L, Chitarra, W, Siciliano, I, Gaiotti, F, Ciuffo, M, Forgia, M, Varese, GC, Turina, M. Mycoviruses mediate mycotoxin regulation in Aspergillus ochraceus. Environ Microbiol21:1957-1968, 2019
  86. 86. Tebbi, CK, Badiga, A, Sahakian, E, Arora, AI, Nair, S, Powers, JJ, Achille, AN, Jaglal, MV, Patel, S, Migone, F. Plasma of acute lymphoblastic leukemia patients react to the culture of a mycovirus containing Aspergillus flavus. J Pediatr Hematol Oncol42:350-358, 2020
  87. 87. Tebbi, CK, Badiga, A, Sahakian, E, Powers, JJ, Achille, AN, Patel, S, Migone, F. Exposure to a mycovirus containing Aspergillus flavus reproduces acute lymphoblastic leukemia cell surface and genetic markers in cells from patients in remission and not controls. Cancer Treat Res Commun26:100279, 2020

Written By

Cameron K. Tebbi, Ioly Kotta-Loizou and Robert H.A. Coutts

Submitted: 29 May 2021 Reviewed: 14 June 2021 Published: 20 July 2021