Next Article in Journal
Enhanced Photocatalytic Removal of Cyanotoxins by Al-Doped ZnO Nanoparticles with Visible-LED Irradiation
Next Article in Special Issue
Impact of Fusarium-Derived Mycoestrogens on Female Reproduction: A Systematic Review
Previous Article in Journal
Switching Shiga Toxin (Stx) Type from Stx2d to Stx2a but Not Stx2c Alters Virulence of Stx-Producing Escherichia coli (STEC) Strain B2F1 in Streptomycin (Str)-Treated Mice
Previous Article in Special Issue
Mycotoxin Deoxynivalenol Has Different Impacts on Intestinal Barrier and Stem Cells by Its Route of Exposure
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Toxic Effect of Aflatoxins in Dogs Fed Contaminated Commercial Dry Feed: A Review

1
Centro de Ciencias Agropecuarias, Universidad Autonoma de Aguascalientes, Aguascalientes 20131, Mexico
2
Centro de Ciencias Básicas, Universidad Autonoma de Aguascalientes, Aguascalientes 20130, Mexico
*
Author to whom correspondence should be addressed.
Submission received: 16 December 2020 / Revised: 8 January 2021 / Accepted: 12 January 2021 / Published: 15 January 2021
(This article belongs to the Special Issue Toxic Effect of Mycotoxins)

Abstract

:
Since its first patent (1897), commercial dry feed (CDF) for dogs has diversified its formulation to meet the nutritional needs of different breeds, age, or special conditions and establish a foundation for integration of these pets into urban lifestyles. The risk of aflatoxicosis in dogs has increased because the ingredients used to formulate CDF have also proliferated, making it difficult to ensure the quality required of each to achieve the safety of the entire CDF. This review contains a description of the fungi and aflatoxins detected in CDF and the ingredients commonly used for their formulation. The mechanisms of action and pathogenic effects of aflatoxins are outlined; as well as the clinical findings, and macroscopic and microscopic lesions found in aflatoxicosis in dogs. In addition, alternatives for diagnosis, treatment, and control of aflatoxins (AF) in CDF are analyzed, such as biomarkers of effect, improvement of blood coagulation, rate of elimination of AF, control of secondary infection, protection of gastric mucosa, reduction of oxidative stress, use of chemo-protectors, sequestrants, grain-free CDF, biocontrol, and maximum permitted limits, are also included.
Key Contribution: This review study highlights the extensive presence of Aspergillus flavus and its aflatoxins (AF) in commercial dry food (CDF) for dogs; unfortunately, information on mycotoxins in dogs is scarce and scattered. Various agro-industrial by-products used as ingredients of CDF show high levels of AF, which reveals the exposure of dogs to AF in their diet, causing clinically observable damage from low doses (60 µg/kg). Treatment and control strategies for contamination due to AF in CDF are also outlined. Therefore, this study serves as the foundation for the knowledge of the risk that dogs integrated into urban life have, in which the CDF contaminated by AF is ingested until all the feed contained within each bag is finished, suggesting an urgent need for quality control mechanisms.

1. Introduction

Commercial dry feed (CDF) for dogs was first patented in 1860 by James Spratt, but it was not until 1957 that it started to be sold commercially. The diversity in the CDF formula increases due to the rise in the knowledge of the nutritional needs of dogs depending on their breeds, ages, and activities carried out. Another aspect that drives the supply of CDF is the increasing availability of agro-industrial ingredients of diverse bromatological composition, which guides the diversification of suitable feed formulas for dogs with differences in their digestive needs, nutrient profiles, and metabolism [1]. Another aspect that drives the demand for CDF is the integration of the dog into the urban lifestyle, as well as the strengthening of the human–pet bond in which dog owners give equal priority to the healthy diet of their pets as well as their own [2].
The CDF offer includes dry derivatives of all types of meat and guts from chicken, beef, pork, and other species; quail, pheasant, and ostrich meat are even incorporated in the making of these feeds [3]. CDF also adds various cereals, such as corn, rice, wheat, barley, and sorghum due to their low cost and acceptable nutritional value; besides this, it does not affect the palatability and digestibility of the nutrients, although the quality and safety of cereals is sometimes objectionable [4]. These ingredients are widely used as a source of energy, as well as a supply of some vitamins, minerals, fibers, and fats [5]. In addition, protein ingredients from soybeans and soybean paste are incorporated as well as some high-fiber ingredients such as alfalfa and oatmeal [6].
Products may be contaminated with Aspergillus spp. fungus, aflatoxins (AF), or some of their secondary metabolites as well as other mycotoxins; contamination occurs at various stages of ingredient production, such as flowering, harvest, processing, or storage of cereals as well as metabolic residues in meat, dairy, and egg products [7,8,9]. Aspergillus species are mainly found in tropical and subtropical regions around the world. However, due to the global import of food materials and climate change, it is likely that soon also regions with temperate climates (such as Europe) will be as affected as developing countries [10].
As presented later in this review, contamination by Aspergillus spp. or its metabolites is an important factor because it reduces the nutritional value of the substrate and produces a great variety of toxic effects; consequently, regulations have been established throughout the world on the maximum permissible levels of aflatoxins in food and feed. Reports of outbreaks of clinical forms of aflatoxicosis in dogs are scarce, but their geographic distribution is very diverse: North America, Latin America, Asia, and Africa. This coincides with a worldwide distribution of aflatoxigenic fungi and their aflatoxins in both complete dog feed and ingredients [11]. Therefore, this work aims to review the presence of aflatoxigenic fungi and their metabolites in commercial dry feed for dogs. The mechanisms of action of AF and its repercussions on animal health are examined. Viable alternatives for the diagnosis and therapy of aflatoxicosis are analyzed; in addition, the control of CDF contamination by aflatoxins is highlighted.

2. Methods

This review was carried out by establishing a research objective, search strategies and relevant research articles; a selection of articles, data extraction, data mapping and summary of results were carried out [12]. The literature for this review was identified by searching online databases (Google scholar, PubMed, and Web of Science). Scientific publications were searched from 2000 to 2020. The search terms were ‘aflatoxin’, ‘dog’, ‘food or feed’, and ‘flavus’. All relevant scientific publications were included in the review, but other kind of information was excluded from the analysis. Two researchers independently searched the literature. Then, the two sets of literature were compared; disagreements about the inclusion of the literature were resolved through group discussion to make the decision. Data on the design, objectives, sample size, setting, instrumental methodology, and main findings were extracted. The articles were classified in the following research areas: ‘Aspergillus spp.’, ‘Aflatoxins’, ‘Feed ingredients’, ‘Commercial dry feed’, ‘Clinical findings’, ‘Lesions’, ‘Therapeutic strategies’ and ‘Control’. All findings and statements in this review are based on published information as indicated in the references.

3. Presence of Aspergillus spp. in Commercial Dry Feed for Dogs

The presence of toxigenic Aspergillus spp. in cereals or other dog feed ingredients incorporated into CDF formulations deteriorates the quality and affects the safety of the whole feed [13]. Cereals and other ingredients are suitable substrates for the growth of fungal microflora, both in the pre-harvest stage and in storage [14]. When the raw extruded material for CDF formulation presents an excess of relative humidity content (20–25%) in the initial stage of the process, and it is reduced by drying (8–10%), only the growth of the vegetative forms of the fungal microflora is inhibited, but the spores and mycotoxins produced within the processed material remain stable. It has been reported that the optimal temperature and water activity for regulatory genes of the AF biosynthesis pathway to reach their maximum expression are 28–30 °C and 0.96–0.99, respectively, although the AF production range is very wide [15,16,17]. Therefore, if the substrate is rehydrated during improper storage conditions, AF concentrations may increase.
Some studies report that the most frequent fungal contaminants present in CDF for dogs are Aspergillus spp., Mucor spp., Penicillium spp., and Rhizopus spp. Several of the genera and species isolated and identified are mycotoxigenic, which produces a risk to dogs’ health [11,18,19,20] (Table 1). Most of the samples included in each study were contaminated by Aspergillus spp. and by some other type of toxigenic fungal microbiota. Although information regarding the presence of Aspergillus spp. and other fungal microflora in various ingredients of human food is extensive, studies of this contamination in CDF are scarce, despite being made with similar ingredients. Furthermore, these reports refer to contamination of whole feed, but no results of toxigenic microflora in CDF ingredients have been reported [18,19,20,21].

4. Aflatoxins and Their Biotransformation Products

The AF are difuranocoumarin compounds produced as secondary metabolites of fungi of the genus Aspergillus spp. following a polyketide path; A. flavus is the main species of fungus involved in AF production [9,11,22]. Four AF naturally present in agricultural products are described (AFB1, AFB2, AFG1, AFG2); other forms of AF are derived from the metabolic process of these primary forms within the human or animal body [28]. AF are not destroyed by boiling nor do they confer, color, aroma, or flavor to contaminated ingredients, so they usually go unnoticed by both the owner and the dog [13]. When dogs ingest CDF made with ingredients contaminated with AF, the mycotoxin are absorbed in the duodenum and bind to plasma albumin and proceed to be transported through the bloodstream [29].
In various tissues, especially in the liver and kidneys, AF are biotransformed and bioactivated by isoenzymes of the multiple function oxidase system or cytochromes (CyP450), giving rise to highly active electrophilic forms called 8,9 endo-epoxide and 8, 9 exo-epoxide, which bind and exert an electrophilic attack on subcellular structures [30]. When AF are metabolized (hydrolysis, demethylation, or ketoreduction), they form less toxic intermediate compounds with greater solubility in water (AFM1, AFM2, AFQ1, AFQ2, AFP1, AFP2, and aflatoxicol); therefore, they are eliminated through feces, urine, milk, or egg [31,32]. The most common ways of elimination of metabolites in the urine of dogs is AFM1, as well as traces of AFQ1 [33,34]. When the epoxide binds to DNA, then an AF-DNA adduct (dihydro-8-(N7-guanyl)-9hydroxy-1 AF-N7–guanidine) is formed, which rearranges as AF-formaminopyrimidine (AF-FAPy), or it is excreted through urine as AF-N7guanine, which is considered a biomarker of genotoxic damage from AF [31,32,33,34]. In general, all compounds derived from natural forms of AF, due to the activity of the enzymes that participate in the detoxification process, are considered biomarkers of exposure and damage [35].
An important detoxification mechanism in many animal species is the involvement of a group of enzymes called Glutathione S-transferases (GST) [36]. The function of GST is to bind the epoxide with the reduced glutathione tripeptide (GSH), which loses two amino acids (glycine and glutamate) to be eliminated as a cysteine residue linked to AF, called mercapturic acid or N-acetylcysteine-AF, which is eliminated through bile or urine [37]. Dogs have a reduced GST activity, which makes them especially susceptible to AF damage; in addition, a deficiency of GSH or its three precursor amino acids facilitates the occurrence of the most extensive oxidative injury [38,39,40].

5. Contamination of Feed Ingredients by Aflatoxins

Cereals are usually integrated into dog feeds, especially corn, sorghum, rice, wheat, oats, barley, and millet; they are a good source of carbohydrates, fiber, protein, fat, minerals, and vitamins [41,42]. However, cereals present an important risk for the health of dogs because they are vulnerable to contamination by A. flavus both in the field and in storage [43,44,45]. In some CDF formulations for dogs, pumpkin seeds, chia, quinoa, and even some legumes such as lentils are included among the ingredients due to their high protein and mineral content. However, these ingredients can also be contaminated by some forms of AF [46,47,48]. Furthermore, the incorporation of both potatoes and sweet potatoes in the manufacturing of premium types of dog feeds is used as a source of carbohydrates and fiber. It is reported that the presence of A. flavus and high concentrations of AF can be found in potato tubers [49,50].
Fruits in CDF are used as a natural source of fiber; papaya is one of the main fruits that are included, however, it may have A. flavus in the postharvest, which has effects on its nutritional value, and it may also have AF concentrations [51]. Blueberries are used as antioxidants, although there are reports of A. flavus contamination and concentrations of AFB1, AFB2, y AFG1 [52]. Orange and coconut are other ingredients that are included in some dog feeds, but they can also be contaminated by FA-producing fungal microflora [53,54,55].
Dairy and meat products as well as eggs are added to dog feed as an important source of proteins and fats. However, secondary AF metabolites such as AFM1, AFM2, AFP1 may be found, which can also contaminate these feed ingredients [56]. The AF residues can be located in by-products of animal origin used in the manufacturing of CDF, such as liver, kidneys, muscle, meat, milk, and egg. The residual compounds in eggs, milk and meat are derived from the biotransformation of the original AF ingested in the feed of animals used for food and remain in the dog that ingests the CDF [57].

6. Aflatoxin Contamination in Commercial Dry Feed for Dogs

Natural forms of AFs and its metabolites can be found in the ingredients used to make CDF for dogs (Table 2). Cereals used in the formulation of CDF may contain high levels of AF contamination (0.48–1.081 μg/kg), making them the most likely sources of aflatoxin contamination [58,59]. AFB1 is the most abundant form in open sampling CDF trials, with values that vary widely (<0.5 and 4.946 μg/kg) [8,60]. CDF is classified into economic, premium, or super premium types of dog feeds according to the nutritional quality of the ingredients, but this classification does not guarantee that it is an AF-free product because they are found in all types of CDF [20,61,62]. Therefore, AF present in the CDF are a health risk, which is especially important because CDF is used as the sole or main component of the diet during most of a dog’s life; in addition, all the feed contained on each bag is usually eaten until it is exhausted, suggesting that prolonged ingestion of feed contaminated with these mycotoxins, even at low doses, can have adverse health effects [63].

7. Clinical Findings in Dog Aflatoxicosis

The median lethal dose (LD50) of AFB1 for the dog is of 0.5–1.5 mg/kg body weight; clinical manifestations are even observed at doses greater than 60 µg/kg of AF in feed [73]. As can be seen in Table 3, higher doses are associated with acute forms of aflatoxicosis. In addition, studies of poisoning outbreaks in dogs have found very high AFB1 values (<5.0 mg/kg) [66].
The clinical signs shown by dogs with aflatoxicosis are identified as digestive, hemodynamic, and nervous alterations. Digestive findings include vomiting, anorexia, hematemesis, hematochezia, and melena. The hemodynamic changes reported are ascites, peripheral edema, jaundice, dehydration, decreased blood pressure, hemorrhagic diathesis, and petechiae in the mucous membranes [38,75,76,77]. Clinical findings related to central nervous system disturbances are attributable to hepatic encephalopathy, manifested by depression, vocalization, stupor, seizures, and coma [38,77].
The clinical findings observed in cases of aflatoxicosis are also related to some variables of the blood biochemistry and coagulation tests (Table 4); the changes that occur are the increase in the specific activity of the liver enzymes: alanine aminotransferase, aspartate aminotransferase, and alkaline phosphatase (ALT, AST, and ALP). The increased activity of these hepatic membrane enzymes is a biomarker of damage due to injury induced by the epoxides generated via AF metabolism. Coagulation tests in cases of aflatoxicosis show a decrease in the ability of blood to clot, evidenced mainly by an increase in prothrombin time and activated partial thromboplastin time (PT and aPTT) as well as a decrease in antithrombin in plasma, protein C activity, and coagulation factor VII (FVII: C) [78]. A decrease in fibrinogen and platelets is also observed. The hemorrhagic effects of AF are attributed to its chemical structure that contains a coumarin ring with an anticoagulant effect; therefore, a delay in coagulation occurs and secondarily induces disseminated intravascular coagulation (DIC) with depletion of the coagulation factors [38,74].
In aflatoxicosis, hyperbilirubinemia is also seen, which is associated with liver failure or inability to conjugate bilirubin generated in the spleen in higher-than-normal amounts, resulting in an increase in total bilirubin [66]. In acute AF poisonings, a decrease in total proteins and albumin is observed. These changes are related to impaired liver function, as well as protein loss from enteric hemorrhage, ascites, and edema [80]. The decrease in cholesterol concentrations in AF poisonings is due to cholestasis, which arises from fibrosis of the bile ducts [81]. Therefore, the digestive and nervous clinical signs that are present simultaneously with the enzymatic and hematic changes specified, suggest the presence of aflatoxicosis, which must be corroborated with clinical history data and with the presence of AF in the feed and in the dog’s stomach. In general, biochemical analyses and coagulation tests are used for the early detection of the effects of exposure to AF; therefore, they constitute biomarkers of exposure and damage by AF [82].

8. Macroscopic and Microscopic Lesions in Aflatoxicosis in Dogs

When dogs ingest CDF contaminated by AF, bioactive compounds are generated that attack subcellular structures and cause damage [30]. For this reason, various macroscopic and microscopic lesions are presented that can be used as a suggestive diagnostic element. The most reported findings are restricted to the liver; however, localized lesions in other organs are also reported, which are related to direct damage from AF, liver failure, coagulation abnormalities, or disseminated intravascular coagulation (Table 5) [66,75,76].

9. Therapeutic Strategies

When a diagnosis of aflatoxicosis has been established in dogs, it is suggested that therapy include, in addition to symptomatic treatment, hemostatic stabilization, increasing the rate of AF elimination and hepatic–renal protection (Table 6) [33,38]. Because AF have an anticoagulant effect like coumarin, the provision of vitamin K1 is proposed to promote the activation of coagulation factors II, VII, IX, X, protein C, and protein S [73,82,88], as well as the provision of intravenous plasma to provide procoagulant proteins [88]. To increase the rate of elimination of AF metabolites, intravenous fluid therapy is administered [33], which is also indicated to correct dehydration and hypovolemia that occur in these cases. As symptomatic treatment, antiemetics, such as metoclopramide or ondansetron; protectors of the gastroenteric mucosa, such as famotidine and sucralfate; and broad-spectrum antibiotics are administered. The latter are used to protect against systemic infection caused by AF-mediated immunosuppression.
The use of N-acetylcysteine (NAC) in dogs with AF poisoning is successful [91] because this compound provides an essential amino acid (L-cysteine) for the intracellular synthesis of GSH; this tripeptide is bound to reactive AF epoxides by the interaction of the GST enzyme, and the combined AF-GSH compound is eliminated as mercapturic acid in feces and urine [92]. Furthermore, NAC functions by itself as a free radical scavenger and has anti-inflammatory properties [91]. The use of silymarin is also proposed [84]; this flavonoid derived from milk thistle could increase GST activity and promote GSH’s synthesis [88]. When the rate of elimination of reactive AF metabolites is increased, the attack of subcellular structures is reduced or inhibited and therefore, cellular integrity is protected, especially of the liver and kidney as targeted organs of attack by AF [89]. Vitamin E is another liver protector used in CDF to prevent lipid peroxidation by AF epoxides and to prevent damage to cell membranes. In the same sense, the use of L-carnitine reduces oxidative damage; this amino acid transports fatty acids from the cytosol to the mitochondria for their β-oxidation and energy generation, thereby reducing intracellular lipid deposition and protecting the cell membrane against the epoxide-induced lipid peroxidation processes of AF [93].

10. Methods Used to Control Aflatoxins in Commercial Dry Feed

The dog feed industry uses various physicochemical and biological methods to reduce AF contamination. Physical processing techniques of CDF ingredients are successfully used, including sieving and pearling of cereals, which are used to separate damaged grains and abrasion of the outer portions of the seeds; both methods decrease the growth of fungal microflora and reduce the content of AF [94]. The use of washing techniques for cereal grains is suggested; although the product moistened by this method requires being completely dried before storage, which generates an additional cost [95]. The proper extrusion of the grains eliminates fungal spores present in the mixture of raw materials because they cannot survive at the temperature and pressure used (150 °C, 37 atm) [96].
They also use fungal microbiota inhibitors based on a wide variety of chemical compounds. Benzoic, acetic, sorbic, and propionic acids inhibit the growth of fungi by acidifying their cytoplasmic content, which is why it is a method used to prevent the formation of AF [94]. The use of ozone gas (O3) in DCF is proposed as an oxidizing agent because O3 alters the structure of cell membranes and induces alterations in cell permeability and destruction of the fungal microflora. Furthermore, when O3 is applied to feed, a reaction occurs with the double bond C8 = C9 to form a vinyl ether in the furan terminal ring of AF; then, an intermediate compound called AF-ozonide is formed; finally, the AF-ozonide is easily degraded into less toxic compounds (carboxylic acid, aldehyde, ketone, and carbon dioxide) [96,97,98,99].
The bioavailability of AF can be decreased by using compounds that reduce its gastrointestinal absorption [31]. These sequestering compounds are widely used because they bind to AF within the digestive tract of dogs through the chemisorption of β-dicarbonyl from AF, thereby reducing their intestinal absorption. The most widely used mineral sequestrants are some phyllosilicates, such as hydrated calcium and sodium aluminosilicates, bentonite, and tectosilicates or zeolites [100]. Bentonite is administered to absorb AF through cation exchange and carbonyl groups, as well as ion-dipole interaction [100]. The adsorption of the zeolite arises from the interaction of the AF with Ca2+ existing on its surface [101]. Furthermore, the use of organic compounds derived from yeast and other microorganisms is a method of adsorption of AFs present in CDF for dogs. Saccharomyces cerevisiae cell walls contain large proportions of mannoproteins and β-D-glucomannan, which exhibit high activity to adsorb AF. Lactic acid bacteria also could reduce the bioavailability of AF through their adsorption by peptidoglycans from their cell wall [102].
The methods of reducing the bioavailability of AF in feed have a wide range of efficacy, but they do not eliminate the risk of poisoning at high concentrations of AF, or long-time ingestion, and they do not prevent the development of fungi and an increased AF concentration when CDF is improperly stored [63]. Even AF contamination has not been able to be eliminated in the food supply chain, even if good agricultural practices are adhered to or optimal storage conditions are maintained. Although the best conditions of agro-industrial production of CDF are able to ensure that the concentration of AF is innocuous and it is distributed within airtight sacks, as soon as the sack is exposed to the environment, the hygroscopicity increases the relative humidity of the CDF (>17%) and the conidia can germinate and begin the production of AF to toxic levels. This risk may be greater with increasing ambient temperature, relative humidity, and time of consumption of the sack content [16,17,103].
One of the most common strategies to control AF contamination is to set the maximum residue level (MRL) or the action levels for AF, which are the maximum concentrations permitted of AF in food or feed [104]. The MRL for AF in feedstuffs varies widely among different countries (0.0–50.0 µg/kg) and is indicated for any animal feed. The European Union has fixed the MRL for AFB1 at 10.0 μg/kg in complete feed and twice as much for feed materials; China, Japan, and Korea apply the same MRL (10.0 μg/kg) for animal feed, especially in dairy feed. However, in many countries in both North America and Latin America, the MRL for AF (aflatoxin B1 + B2 + G1 and G2) are set at 20 µg/kg for AF in feedstuffs because it is assumed that animals like dogs are susceptible to the toxic effects of AF at higher doses [73,105]; although, many countries of Asia and Oceania, like India, Nepal and Senegal have higher MRL (>20.0 μg/kg) for AF in feedstuffs, including in the dog feed [63,95,104,106]. On other hand, these regulations are intended to protect animal health and prevent AF toxicity; but there is no evidence of the effect that prolonged exposure to AF below the MRL could have on dog health, especially when AF-contaminated CDF is ingested until all the feed contained within each bag is finished, suggesting an urgent need for quality control mechanisms.

11. Conclusions and Recommendations

Aflatoxins and the fungi that produce them, particularly Aspergillus flavus, are common in the main ingredients used to make commercial dry feed for dogs (grains, meat and bone meals, viscera, tubers, fruits, etc.). These ingredients are agro-industrial by-products that are used to satisfy the specific nutritional requirements of the dog (age, weight, activity, etc.) and to allow its incorporation into the urban lifestyle. Therefore, the presence of mycotoxigenic fungi and their toxins could be considered a serious problem for the health of the dog, to develop its zootechnical function as a companion, guardian, or as a sports animal. In addition, dangerous levels of aflatoxins are potentiated because the dog must ingest large amounts of the CDF until all the content found in each bag that its owner purchases is finished off. Although studies on the impact of exposure to AF on the health of dogs are limited, these studies are adequate to demonstrate its impact on clinical manifestations, macroscopic and microscopic lesions, and hematological and enzymatic alterations. Therefore, to address these problems, the use of therapeutic and control strategies that mitigate the impact developed by aflatoxins is recommended. In addition, the establishment of maximum permissible levels of AF specifically for CDF and research on prolonged exposure to low concentrations of aflatoxins should be encouraged.

Author Contributions

Conceptualization, L.M.-M. and A.G.V.-F.; methodology, E.J.R.-M.; data curation, E.J.R.-M.; writing—original draft preparation, L.M.-M. and A.G.V.-F.; writing—review and editing, A.G.V.-F., A.L.G.-B., T.Q.-T., and R.O.-M.; All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Science and Technology National Council (CONACyT, grant number 738906 and Universidad Autónoma de Aguascalientes (project number: PIP/SA 19-1).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

We are grateful to Adrian Ugarte, Keith MacMillan, and Abby Greazel for their participation for researching information and translating the draft manuscript into the English language.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the man-uscript, or in the decision to publish the results.

References

  1. Case, L.; Daristotle, L.; Hayek, M.; Raasch, M. History and regulation of pet food. In Canine and Feline Nutrition; Case, L., Daristotle, L., Hayek, M., Raasch, M., Eds.; Mosby: Saint Louis, MO, USA, 2011; pp. 121–129. [Google Scholar] [CrossRef]
  2. Schleicher, M.; Cash, S.B.; Freeman, L.M. Determinants of pet food purchasing decisions. Can. Vet. J. 2019, 60, 644–650. [Google Scholar] [PubMed]
  3. Rojas, M.; González, I.; De la Cruz, S.; Hernández, P.E.; García, T.; Martín, R. Application of species-specific polymerase chain reaction assays to verify the labeling of quail (Coturnix coturnix), pheasant (Phasianus colchicus) and ostrich (Struthio camelus) in pet foods. Anim. Feed Sci. Tech. 2011, 169, 128–133. [Google Scholar] [CrossRef]
  4. Tegzes, J.H.; Oakley, B.B.; Brennan, G. Comparison of mycotoxin concentrations in grain versus grain-free dry and wet commercial dog foods. Toxicol. Commun. 2019, 3, 61–66. [Google Scholar] [CrossRef] [Green Version]
  5. Beloshapka, A.; Buff, P.P.; Fahey Jr, G.C.; Swanson, K.S. Compositional analysis of whole grains, processed grains, grain co-products, and other carbohydrate sources with applicability to pet animal nutrition. Foods 2016, 5, 23. [Google Scholar] [CrossRef] [PubMed]
  6. Yathavamoorthi, R.; Nithin, C.T.; Ananthanarayanana, T.R.; Mathew, S.; Bindu, J.; Anandana, R.; Gopal, T.K. Tuna red meat as a novel ingredient in pet food for dogs. J. Aquat. Food Prod. Technol. 2020, 29, 1–10. [Google Scholar] [CrossRef]
  7. Ott, L.C.; Appleton, H.J.; Shi, H.; Keener, K.; Mellata, M. High voltage atmospheric cold plasma treatment inactivates Aspergillus flavus spores and deoxynivalenol toxin. Food Microbiol. 2020, in press. [Google Scholar] [CrossRef] [PubMed]
  8. Fuentes, S.; Carvajal, M.; Ruiz, S.; Martínez, N.C.; Gómez, A.A.; Rojo, F. Presence of mutagens and carcinogens, called aflatoxins, and their hydroxylated metabolites in industrialized food for dogs. J. Microb. Biochem. Technol. 2018, 10, 76–86. [Google Scholar] [CrossRef]
  9. Alshannaq, A.; Yu, J. Occurrence, toxicity, and analysis of major mycotoxins in food. Int. J. Environ. Res. Pub. Health 2017, 14, 632. [Google Scholar] [CrossRef] [Green Version]
  10. Mahato, D.K.; Lee, K.E.; Kamle, M.; Devi, S.; Dewangan, K.N.; Kumar, P.; Kang, S.G. Aflatoxins in food and feed: An overview on prevalence, detection and control strategies. Front. Microbiol. 2019, 10, 2266. [Google Scholar] [CrossRef]
  11. Bueno, D.J.; Silva, J.O.; Oliver, G. Mycoflora in commercial pet foods. J. Food Protect. 2001, 64, 741–743. [Google Scholar] [CrossRef]
  12. Arksey, H.; O’Malley, L. Scoping studies: Towards a methodological framework. Int. J. Soc. Res. Methodol. 2007, 8, 19–32. [Google Scholar] [CrossRef] [Green Version]
  13. Aquino, S.; Corrêa, B. Aflatoxins in Pet Foods: A risk to special consumers. In Aflatoxins—Detection, Measurement and Control; Torres-Pacheco, I., Ed.; InTech: Rijeka, Croatia, 2011; pp. 53–74. [Google Scholar] [CrossRef]
  14. Bennett, J.M.; Klich, M. Mycotoxins. Clin. Microbiol. Rev. 2003, 16, 497–516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Zicker, S.C. Evaluating pet foods: How confident are you when you recommend a commercial pet food? Top Companion Anim. Med. 2008, 23, 121–126. [Google Scholar] [CrossRef] [PubMed]
  16. Gallo, A.; Solfrizzo, M.; Epifani, F.; Panzarini, G.; Perrone, G. Effect of temperature and water activity on gene expression and aflatoxin biosynthesis in Aspergillus flavus on almond medium. Int. J. Food Microbiol. 2015, 217, 162–169. [Google Scholar] [CrossRef]
  17. Chiewchan, N.; Mujumdar, A.; Devahastin, S. Application of drying technology to control aflatoxins in foods and feeds: A Review. Dry Technol. 2015, 33, 1700–1707. [Google Scholar] [CrossRef]
  18. Witaszak, N.; Stepién, Ł.; Bocianowski, J.; Wáskiewicz, A. Fusarium species and mycotoxins contaminating veterinary diets for dogs and cats. Microorganisms 2019, 7, 26. [Google Scholar] [CrossRef] [Green Version]
  19. Błajet-Kosicka, A.; Kosicki, R.; Twarużek, M.; Grajewski, J. Determination of moulds and mycotoxins in dry dog and cat food using liquid chromatography with mass spectrometry and fluorescence detection. Food Addit. Contam. B Part 2014, 7, 302–308. [Google Scholar] [CrossRef]
  20. Singh, S.D.; Chuturgoon, A.A. A comparative analysis of mycotoxin contamination. J. S. Afr. Vet. Assoc. 2017, 88, 1–6. [Google Scholar] [CrossRef]
  21. Hołda, K.; Wiczuk, W.; Hać-szymańczuk, E.; Głogowski, R. Comprehensive microbiological evaluation of dry foods for growing dogs marketed in Poland. Anim. Sci. 2017, 56, 81–89. [Google Scholar] [CrossRef] [Green Version]
  22. Campos, S.G.; Keller, L.M.; Cavaglieri, L.R.; Krüger, C.; Fernández, M.G.; Dalcero, A.M.; Magnoli, C.E.; Rosa, C.A. Aflatoxigenic fungi and aflatoxin B1 in commercial pet food in Brazil. World Mycotoxin J. 2009, 2, 85–90. [Google Scholar] [CrossRef]
  23. Copetti, M.V.; Santurio, J.M.; Cavalheiro, A.S.; Alves, S.H.; Ferreiro, L. Comparison of different culture media for mycological evaluation of commercial pet food. Acta Sci. Vet. 2009, 37, 329–335. [Google Scholar] [CrossRef]
  24. Girio, T.M.S.; Filho, A.N.; Junior, O.D.R.; Amaral, L.A.; Girio, R.J.S. Microbiological quality of dog feed sold in sealed packages and in bulk. Ars. Vet. Jaboticabal. SP 2012, 28, 36–40. [Google Scholar]
  25. Martins, M.L.; Martins, H.M.; Bernardo, F. Fungal flora and mycotoxins detection in commercial pet food. Rev. Port. Ciênc. Vet. 2003, 98, 179–183. [Google Scholar]
  26. Scudamore, K.A.; Hetmanski, M.T.; Nawaz, S.; Naylor, J.; Rainbird, S. Determination of mycotoxins in pet foods sold for domestic pets and wild birds using linked-column immunoassay clean-up and HPLC. Food Addit. Contam. Part A Chem. Anal. Control Expo. Risk Assess 2009, 14, 175–186. [Google Scholar] [CrossRef] [PubMed]
  27. Muñoz, D.J.; Rodríguez, R.; Mota, J.J.; Suarez, L.R. Isolation and identification of phylamentous fungi in feed for domestic pets (dogs and cats). Rev. Cientif. FCV-LUZ 2015, 25, 432–438. [Google Scholar]
  28. Klich, M. Aspergillus flavus: The major producer of aflatoxin. Mol. Plant Pathol. 2007, 8, 713–722. [Google Scholar] [CrossRef]
  29. Tessari, E.N.; Kobashigawa, E.; Cardoso, A.L.; Ledoux, D.R. Effects of Aflatoxin B1 and Fumonisin B1 on blood biochemical. Toxins 2010, 2, 453–460. [Google Scholar] [CrossRef] [Green Version]
  30. Tulayakul, P.; Sakuda, S.; Dong, K.S.; Kumagai, S. Comparative activities of glutathione-S-transferase and dialdehyde reductase toward aflatoxin B1 in livers of experimental and farm animals. Toxicon 2005, 46, 204–209. [Google Scholar] [CrossRef]
  31. Yiannikouris, A.; Jouany, J. Mycotoxins in feeds and their fate in animals: A review. Anim. Res. 2002, 51, 81–99. [Google Scholar] [CrossRef] [Green Version]
  32. Galtier, P. Biotransformation and Fate of Mycotoxins. J. Toxicol. 1999, 18, 295–312. [Google Scholar] [CrossRef]
  33. Bingham, A.; Huebner, H.; Phillips, T.; Bauer, J. Identification and reduction of urinary aflatoxin metabolites in dogs. Food Chem. Toxicol. 2004, 42, 1851–1858. [Google Scholar] [CrossRef] [PubMed]
  34. Wu, Q.; Jezkova, A.; Yuan, Z.; Pavlikova, L.; Dohnal, V.; Kuca, K. Biological degradation of aflatoxins. Drug Metab. Rev. 2009, 41, 1–7. [Google Scholar] [CrossRef] [PubMed]
  35. Benkerroum, N. Aflatoxins: Producing-molds, structure, health issues and incidence in Southeast Asian and Sub-Saharan African countries. Int. J. Environ. Res. Pub. Health 2020, 17, 1215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Uribe-Yunda, D.F.; Navas, M.C. Mecanismos moleculares involucrados en la mutagenicidad inducida por aflatoxina B1. Rev. Cienc. Salud. 2012, 10, 403–419. [Google Scholar]
  37. Rawal, S.; Kim, J.E.; Coulombe, R., Jr. Aflatoxin B1 in poultry: Toxicology, metabolism and prevention. Res. Vet. Sci. 2010, 89, 325–331. [Google Scholar] [CrossRef]
  38. Bruchim, Y.; Segev, G.; Sela, U.; Bdolah, T.; Salomon, A.; Aroch, I. Accidental fatal aflatoxicosis due to contaminated commercial diet in 50 dogs. Res. Vet. Sci. 2012, 93, 279–287. [Google Scholar] [CrossRef]
  39. Mehrzad, J.; Fazel, F.; Pouyamehr, N.; Hosseinkhanil, S.; Dehghani, H. Naturally occurring level of aflatoxin B1 injures human, canine and bovine leukocytes through ATP depletion and caspase activation. Int. J. Toxicol. 2019, 141, 16–25. [Google Scholar] [CrossRef]
  40. Towner, R.A.; Qian, S.Y.; Kadiiska, M.B.; Mason, R.P. In vivo identification of aflatoxin-induced free radicals in rat bile. Free Radic. Biomed. 2003, 35, 1330–1340. [Google Scholar] [CrossRef]
  41. Macías, A.; Rial, C.; Acosta, A.; Henríquez, L.A.; Almeida, M.; Rodríguez, Á.; Zumbado, M.; Boada, L.D.; Zaccaroni, A.; Luzardo, O.P. Risk assessment of the exposure to mycotoxins in dogs and cats through the consumption of commercial dry food. Sci. Total Environ. 2020, 15, 134592. [Google Scholar] [CrossRef]
  42. Peterson, D.M. Oat antioxidants. J. Cereal. Sci. 2001, 33, 115–129. [Google Scholar] [CrossRef]
  43. Wan, J.; Chen, B.; Rao, J.J. Occurrence and preventive strategies to control mycotoxins in cereal-based food. Compr. Rev. Food Sci. F 2019, 19, 1–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Xu, L.; Zhang, Z.; Zhang, Q.; Zhang, W.; Yu, L.; Wang, D.; Li, H.; Li, P. An on-site simultaneous semi-quantification of aflatoxin B1, zearalenone, and T-2 toxin in maize- and cereal-based feed via multicolor immunochromatographic Assay. Toxins 2018, 10, 87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Kamala, A.; Ortiz, J.; Kimanya, M.; Haesaert, G.; Donoso, S.; Tiisekwa, B.; De Meulenaer, B. Multiple mycotoxin co-occurrence in maize grown in three agro-ecological zones of Tanzania. Food Control 2015, 54, 208–215. [Google Scholar] [CrossRef]
  46. Herrera, M.; Bervis, N.; Carramiñana, J.J.; Juan, T.; Herrera, A.; Ariño, A.; Lorán, S. Occurrence and exposure assessment of aflatoxins and deoxynivalenol in cereal-based baby foods for infants. Toxins 2019, 11, 150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Hacıbekiroğlu, I.; Kolak, U. Aflatoxins in various food from Istanbul, Turkey. Food Addit. Contam. B 2013, 6, 260–264. [Google Scholar] [CrossRef] [PubMed]
  48. Nazir, A.; Kalim, I.; Sajjad, M.; Usman, M.; Iqbal, M. Prevalence of aflatoxin contamination in pulses and spices in different regions of Punjab. Chem. Int. 2019, 5, 274–280. [Google Scholar] [CrossRef]
  49. Zöngür, A. Investigation of DON and aflatoxin content in Agria and Hermes chips potato cultivars grown in Aksaray, Sivas, Kayseri, Niğde. J. Plant Dis. Protect 2020, 127, 521–527. [Google Scholar] [CrossRef]
  50. Amri, E.; Lenoi, S. Aflatoxin and fumonisin contamination of sun-dried sweet potato (Ipomoea batatas L.) chips in Kahama district, Tanzania. Appl. Environ. Microbiol. 2016, 4, 55–62. [Google Scholar] [CrossRef]
  51. Bagwan, N.B. Aflatoxin B1 contamination in papaya fruits (Carica papaya L.) during post harvest pathogenesis. Indian Phytopath. 2011, 64, 48–50. [Google Scholar]
  52. Munitz, M.S.; Resnik, S.L.; Pacin, A.; Salas, P.; Gonzalez, H.L.; Montti, M.L.; Drunday, V.; Guillin, E. Mycotoxigenic potential of fungi isolated from freshly harvested Argentinean blueberries. Mycotoxin Res. 2014, 30, 221–229. [Google Scholar] [CrossRef]
  53. Drusch, S.; Ragab, W. Mycotoxins in fruits, fruit juices, and dried fruits. J. Food Protect 2003, 66, 1514–1527. [Google Scholar] [CrossRef] [PubMed]
  54. AboDalam, T.H.; Amra, H.; Sultan, Y.; Magan, N.; Carlobos, A.L.; Cumagun, C.J.; Yli, T. New genotypes of aflatoxigenic fungi from Egypt and the Philippines. Fungal. Biol. 2020, 10, 142–155. [Google Scholar] [CrossRef]
  55. Granados, F.; Redondo, M.; Jaikel, D. Mycotoxin contamination of beverages obtained from tropical crops. Beverages 2018, 4, 83. [Google Scholar] [CrossRef] [Green Version]
  56. Pour, S.H.; Mahmoudi, S.; Masoumi, S.; Rezaie, S.; Barac, A.; Ranjbaran, M.; Oliya, S.; Mehravar, F.; Sasani, E.; Noorbakhsh, F.; et al. Aflatoxin M1 contamination level in Iranian milk and dairy products: A systematic review and meta-analysis. World Mycotoxin J. 2020, 13, 67–82. [Google Scholar] [CrossRef]
  57. Cavus, S.; Tornuk, F.; Sarioglu, K.; Yetim, H. Determination of mold contamination and aflatoxin levels of the meat products/ingredients collected from Turkey market. J. Food Saf. 2018, 38, 1–7. [Google Scholar] [CrossRef]
  58. Pitt, J.I.; Taniwaki, M.H.; Cole, M.B. Mycotoxin production in major crops as influenced by growing, harvesting, storage and processing, with emphasis on the achievement of food safety objectives. Food Control 2013, 32, 205–215. [Google Scholar] [CrossRef]
  59. Rodríguez, M.; Ramos, A.J.; Prim, M.; Sanchis, V.; Marín, S. Usefulness of the analytical control of aflatoxins in feedstuffs for dairy cows for the prevention of aflatoxin M1 in milk. Mycotoxin Res. 2019, 36, 11–22. [Google Scholar] [CrossRef]
  60. Algahtani, F.D.; Morshdy, A.E.; Hussein, M.; Abouelkheir, E.S.; Adeboye, A.; Valentine, A.; Elabbasy, M.T. Biogenic amines and aflatoxins in some imported meat products: Incidence, occurrence, and public health impacts. J. Food Qual. 2020, 8718179. [Google Scholar] [CrossRef]
  61. Teixeira, E.M.; Frehse, M.S.; Freire, R.L.; Ono, M.A.; Bordini, J.G.; Hirozawa, M.T.; Ono, E.Y. Safety of low and high cost dry feed intended for dogs in Brazil concerning fumonisins, zearalenone and aflatoxins. World Mycotoxin J. 2017, 10, 273–283. [Google Scholar] [CrossRef]
  62. Gazzotti, T.; Biagi, G.; Pagliuca, G.; Pinna, C.; Scardilli, M.; Grandi, M.; Zaghini, G. Occurrence of mycotoxins in extruded commercial dog food. Anim. Feed Sci. Tech. 2015, 202, 81–89. [Google Scholar] [CrossRef]
  63. Hernandez, E.; Valdivia, A.; Cruz, C.; Saldaña, M.; Quezada, T.; Rangel, E.; Ortiz, R.; Medina, L.; Jaramillo. Diagnosis of subclinical aflatoxicosis by biochemical changes in dairy cows under field conditions. Pak. Vet. J. 2020, 1–6. [Google Scholar] [CrossRef]
  64. Maia, P.P.; de Siqueira, M.E.P.B. Occurrence of aflatoxins B1, B2, G1 and G2 in some Brazilian pet foods. Food Addit. Contam. 2002, 19, 1180–1183. [Google Scholar] [CrossRef] [PubMed]
  65. Shao, M.; Li, L.; Gu, Z.; Yao, M.; Xu, D.; Fan, W. Mycotoxins in commercial dry pet food in China. Food Addit. Contamb. 2018, 11, 237–245. [Google Scholar] [CrossRef]
  66. Newman, S.J.; Smith, J.R.; Stenske, K.A.; Newman, L.B.; Dunlap, J.R.; Imerman, P.M.; Kirk, C.A. Aflatoxicosis in nine dogs after exposure to contaminated commercial dog food. J. Vet. Diagn. Investig. 2007, 19, 168–175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Castaldo, L.; Graziani, G.; Gaspari, A.; Izzo, L.; Tolosa, J.; Rodríguez, Y.; Ritieni, A. Target analysis and retrospective screening of multiple mycotoxins in pet food using UHPLC-Q-Orbitrap HRMS. Toxins 2019, 11, 434. [Google Scholar] [CrossRef] [Green Version]
  68. Sharma, M.; Marquez, C. Determination of aflatoxins in domestic pet foods (dog and cat) using immunoaffinity column and HPLC. Anim. Feed Sci. Technol. 2001, 93, 109–114. [Google Scholar] [CrossRef]
  69. Akinrinmade, J.F.; Akinrinde, A.S. Aflatoxin status of some commercial dry dog foods in Ibadan, Nigeria. Afr. J. Biotechnol. 2012, 11, 11463–11467. [Google Scholar] [CrossRef]
  70. Basalan, M.; Hismiogullari, S.E.; Hismiogullari, A.A.; Filazi, A. Fungi and aflatoxin B1 in horse and dog feeds in western Turkey. Revue Méd. Vét. 2004, 156, 248–252. [Google Scholar]
  71. Gunsen, U.; Yaroglu, T. Aflatoxin in dog and horse feeds in Turkey. Vet. Hum. Toxicol. 2002, 44, 113–114. [Google Scholar]
  72. Reis-Gomes, A.; Marcolongo-Pereira, C.; Sallis, E.S.V.; Pereira, D.I.B.; Schild, A.L.; Faria, R.O.; Meireles, M.C.A. Aflatoxicosis in dogs in Southern Rio Grande do Sul. Pesq. Vet. Bras. 2014, 34, 162–166. [Google Scholar] [CrossRef] [Green Version]
  73. Stenske, K.; Smith, J.; Newman, S.; Newman, L.; Kirk, C. Aflatoxicosis in dogs and dealing with suspected contaminated commercial foods. J. Am. Vet. Med. 2006, 228, 1686–1691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Hagiwara, M.; Kogika, M.; Malucelli, B. Disseminated intravascular coagulation in dog with aflatoxicosis. J. Small Anim. Pract. 1990, 31, 239–243. [Google Scholar] [CrossRef]
  75. Bastianello, S.S.; Nesbit, J.W.; Williams, M.C.; Lange, A.L. Pathological findings in a natural outbreak of aflatoxicosis in dogs. Onderstepoort J. Vet. Res. 1987, 54, 635–640. [Google Scholar] [PubMed]
  76. Arnot, L.; Duncan, N.; Coetzer, H.; Botha, C. An outbreak of canine aflatoxicosis in Gauteng province, South Africa. J. S. Afr. Vet. Assoc. 2012, 83, 1–4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Dereszynski, D.; Center, S.; Randolph, J.; Brooks, M.; Hadden, A.; Palyada, K.; McDonough, S.; Messick, J.; Stokol, T.; Bischoff, K.; et al. Clinical and clinicopathologic features of dogs that consumed foodborne hepatotoxic aflatoxins: 72 cases (2005–2006). JAVMA J. Am. Vet. Med. A 2008, 232, 1329–1337. [Google Scholar] [CrossRef]
  78. Prins, M.; Schellens, C.J.; van Leeuwen, M.W.; Rothuizen, J.; Teske, E. Coagulation disorders in dogs with hepatic disease. Vet. J. 2010, 185, 163–168. [Google Scholar] [CrossRef] [PubMed]
  79. Donahue, S.M.; Brooks, M.; Otto, C.M. Examination of hemostatic parameters to detect hypercoagulability in dogs with severe protein-losing nephropathy. J. Vet. Emerg. Crit. Care 2011, 21, 346–355. [Google Scholar] [CrossRef]
  80. Ensley, S.M.; Radke, S.L. Mycotoxins in grains and feeds. In Diseases of Swine; Zimmerman, J.J., Karriker, L.A., Ramirez, A., Schwartz, K.J., Stevenson, G.W., Zhang, J., Eds.; Wiley-Blackwell: Hoboken, NJ, USA, 2019; pp. 1055–1071. [Google Scholar] [CrossRef]
  81. Rauber, R.H.; Dilkin, P.; Giacomini, L.Z.; de Almeida, C.A.A.; Mallmann, C.A. Performance of Turkey poults fed different doses of aflatoxins in the diet. Poult. Sci. 2007, 86, 1620–1624. [Google Scholar] [CrossRef]
  82. Wada, H.; Usui, M.; Sakuragawa, N. Hemostatic Abnormalities and Liver Diseases; Favaloro, J.E., Ed.; Seminars in Thrombosis and Hemostasis: New York, NY, USA, 2008. [Google Scholar]
  83. Newberne, P.; Russo, R.; Wogan, G. Acute toxicity of aflatoxin B1 in the dog. Pathol. Vet. 1966, 3, 331–340. [Google Scholar] [CrossRef]
  84. Bischoff, K.; Rumbeiha, W. Pet food recalls and pet food contaminants in small animals: An update. Vet. Clin. Small Anim. 2018, 48, 917–931. [Google Scholar] [CrossRef]
  85. Eroksuz, Y.; Kaya, E.; Issi, M.; Baydar, E.; Cevik, A.; Eroksuz, H.; Asci, Z.; Timurkan, O. Subacute aflatoxicosis due to moldy bread consumption in a dog. Revue Méd. Vét. 2015, 166, 259–265. [Google Scholar]
  86. Ural, K.; Ulutas, B.; Tunca, R.; Kum, C.; Avci, H.; Boyacioglu, M.; Gultekin, M.; Atasoy, A. Aflatoxicosis in rottweilers after eating moldy bread: Clinicopathological features and effective tetrasulphate therapy. Vet. Arhiv. 2013, 83, 403–412. [Google Scholar]
  87. Wouters, A.T.; Casagrande, R.A.; Wouters, F.; Watanabe, T.T.; Boabaid, F.M.; Cruz, C.E.; Driemeier, D. An outbreak of aflatoxin poisoning in dogs associated with aflatoxin B1–contaminated maize products. J. Vet. Diagn. Investig. 2013, 25, 282–287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Weingarten, M.A.; Sande, A.A. Acute liver failure in dogs and cats. J. Vet. Emerg. Crit Care 2015, 25, 455–473. [Google Scholar] [CrossRef] [PubMed]
  89. Griebsch, C.; Whitney, J.; Angles, J.; Bennett, P. Acute liver failure in two dogs following ingestion of cheese tree (Glochidion ferdinandi) roots. J. Vet. Emerg. Crit. Care 2018, 1, 1–11. [Google Scholar] [CrossRef] [PubMed]
  90. Sanderson, S.L. Taurine and carnitine in canine cardiomyopathy. Vet. Clin. Small Anim. 2006, 36, 1325–1343. [Google Scholar] [CrossRef]
  91. Viviano, K.R.; Wielen, B.V. Effect of N-Acetylcysteine supplementation on intracellular glutathione, urine isoprostanes, clinical score, and survival in hospitalized Ill dogs. J. Vet. Intern. Med. 2013, 27, 250–258. [Google Scholar] [CrossRef]
  92. Wild, C.P.; Turner, P.C. The toxicology of aflatoxins as a basis for public health decisions. Mutagenesis 2002, 17, 471–481. [Google Scholar] [CrossRef]
  93. Citil, M.; Gunes, V.; Atakisi, O.; Ozcan, A.; Tuzcua, M.; Dogan, A. Protective effect of L-carnitine against oxidative damage caused by experimental chronic aflatoxicosis in quail (Coturnix coturnix). Acta Vet. Hung. 2005, 53, 319–324. [Google Scholar] [CrossRef] [Green Version]
  94. Atungulu, G.G.; Mohammadi-Shad, Z.; Wilson, S. Mycotoxin issues in pet food. Food Feed Saf. Syst. Anal. 2018, 1, 25–44. [Google Scholar] [CrossRef]
  95. Leung, M.C.K.; Díaz-Llano, G.; Smith, T.K. Mycotoxins in pet food: A review on worldwide prevalence and preventative strategies. J. Agric. Food Chem. 2006, 54, 9623–9635. [Google Scholar] [CrossRef] [PubMed]
  96. Silva, J.; Pereira, M.N.; Scussel, V.M. Ozone gas antifungal effect on extruded dog food contaminated with Aspergillus flavus. Ozone Sci. 2018, 7, 1–8. [Google Scholar] [CrossRef]
  97. Lambert, R.; Stratford, M. Weak-acid preservatives: Modelling microbial inhibition and response. J. Appl. Microbiol. 1999, 86, 157–164. [Google Scholar] [CrossRef] [PubMed]
  98. Christ, D.; Savi, G.D.; Scussel, V.M. Effectiveness of ozone gas in raw and processed food for fungi and mycotoxin decontamination-A review. J. Chem. Biol. Phys. Sci. 2016, 6, 326–348. [Google Scholar] [CrossRef]
  99. Diao, E.; Hou, H.; Chen, B.; Shan, C.; Dong, H. Ozonolysis efficiency and safety evaluation of aflatoxi B1 in peanuts. Food Chem. Toxicol. 2013, 55, 519–525. [Google Scholar] [CrossRef]
  100. Di Gregorio, M.C.; de Neeff, D.V.; Jager, A.V.; Corassin, C.H.; de Pinho, A.C.; de Albuquerque, R.; de Azevedo, A.C.; Fernandes, C.A. Mineral adsorbents for prevention of mycotoxins in animal feeds. Toxin Rev. 2014, 33, 125–135. [Google Scholar] [CrossRef]
  101. Tomašević-Čanović, M.; Daković, A.; Rottinghaus, G.; Matijašević, S.; Đuričić, M. Surfactant modified zeolites–new efficient adsorbents for mycotoxins. Microporous Mat. 2003, 61, 173–180. [Google Scholar] [CrossRef]
  102. Lahtinen, S.J.; Haskard, C.A.; Ouwehand, A.C.; Salminen, S.J.; Ahokas, J.T. Binding of aflatoxin B1 to cell wall components of Lactobacillus rhamnosus strain GG. Food Addit. Contam. 2004, 21, 158–164. [Google Scholar] [CrossRef]
  103. De Luna-López, M.C.; Valdivia-Flores, A.G.; Jaramillo-Juárez, F.; Reyes, J.L.; Ortiz-Martínez, R.; Quezada-Tristán, T. Association between Aspergillus flavus colonization and aflatoxins production in immature grains of maize genotypes. J. Food Sci. Eng. 2013, 3, 688–698. [Google Scholar] [CrossRef]
  104. FAO (Food and Agriculture Organization). Worldwide Regulations for mycotoxins in food and feed 2003. FAO Food Nutr. Pap. 2003, 81, 59. [Google Scholar]
  105. Witaszak, N.; Waśkiewicz, A.; Bocianowski, J.; Stępień, Ł. Contamination of pet food with mycobiota and Fusarium mycotoxins-focus on dogs and cats. Toxins 2020, 12, 130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. EU (European Union). Commission Directive 2003/100/EC of 31 October 2003 amending Annex I to Directive 2002/32/EC of the European Parliament and of the Council on undesirable substances in animal feed. Off. J. Eur. Communities 2003, 46, 33–37. [Google Scholar]
Table 1. Fungal microflora detected in commercial dry feed for dogs.
Table 1. Fungal microflora detected in commercial dry feed for dogs.
LocationNumber of Samples (n)Major Fungi IdentifiedPositive Samples (%)Citation
Argentina12Aspergillus flavus, A. niger, Mucor globosus, M. plumbeus, M. racemosus, Rhizopus spp.100[11]
Brazil180A. flavus, A. candidus, A. flavipes, A. fumigatus, A. niger, A. ochraceus, A. parasiticus, Cladosporium spp., Fusarium spp., Mucor spp., Penicillium spp.100[22]
Brazil34Absidia spp., Aureobasidium spp., Alternaria spp., Aspergillus spp., Chrysonilia spp., Cladosporium spp., Emericella spp., Eurotium spp., Fusarium spp., Geotrichum spp., Monascus spp., Mucor spp., Olyptrichum spp., Paecilomyces spp., Penicillium spp., Phoma spp., Rhodotorula spp., Rhizopus spp., Scapulariopsis spp., Syncephalostrum spp., Tilletiopsis spp., Trichoderma spp., Wallemia spp., Yeasts74[23]
Brazil60Aspergillus spp., Fusarium spp., Penicillium spp., Rhizopus spp.53.3[24]
Poland25Aspergillus spp., Mucor spp., Penicillium spp.52[19]
Poland20Aspergillus spp., Penicillium spp., Rhizopus spp.25[21]
Poland25Aspergillus spp., Alternaria spp., Cladosporium spp., Fusarium spp., F. verticillioides., F. proliferatum100[18]
Portugal 20A. niger, Mucor spp., Penicillium spp.100[25]
South Africa20A. flavus, A. fumigatus, A. parasiticus, F. graminearum, F. verticilloides, Penicillium spp.100[20]
United Kingdom5Absidia spp., Acremonium spp., Alternaria spp., Aspergillus spp., Cladosporium spp., Eurotium spp., Mucor spp., Pénicillium spp., Rhizopus spp., Syncephalastrum spp., Wallemia spp., yeasts.100[26]
Venezuela4Acremonium charticola, A. flavus, A. fumigatus, A. terreus, C. herbarum, F. poae, P. citrinum, P. expansum.94[27]
Table 2. Presence of aflatoxins in commercial dry feed for dogs.
Table 2. Presence of aflatoxins in commercial dry feed for dogs.
LocationNumber of Sample (n)TestMean AF (μg/kg)Positive Samples (%)Citation
Brazil45TLCAFB1 (19.0)AFB1 (6.7)[64]
Brazil180HPLCAFB1 (7.0)AFB1 (100)[22]
Brazil(AE) 49 (AP) 25 (ASP) 13HPLC(SF) AF (1.2)
(PF) AF (0.4)
(SPF) AF (0.5)
AF (95.4)[61]
China32LC-MS/MSAFB1 (47.7)AFB1 (87.5)[65]
United States9ELISA, TLC, HPLCAFB1 (530)
AFB2 (19.0)
AFB1 (88.8)
AFB2 (77.7)
[66]
Italy(AE) 24 (AP) 24LC-MS, PLC-MS/MSAFB1 y AFG1 (<0.5)
AFB2 (5.7)
AFG2 (15.8)
AF (12.0)[62]
Italy55UHPLC-Q-Orbitrap HRMSAFB1 (4.3)AFB1 (25.8)[67]
Mexico19HPLCAFB1 (5.0), AFB2(0.07), AFG1 (0.05), AFG2 (0.03), AFM1 (2.0) AFM2 (0.1) AFP1 (1.1), AFL (0.3)AFB1 (79.0), AFB2 (26.0), AFG1 (63.0), AFG2 (21.0), AFM1 (63.0), AFM2 (89.0), AFP1 (58.0), AFL (47.0)[68]
Mexico29HPLC-FLAFB1 (1.6), AFB2(0.1), AFG1 (28.2), AFG2 (1.3), AFM1 (1.8), AFM2 (0.2), AFP1 (1.7), AFL (28.6)AFB1 (76.0), AFB2 (4.0), AFG1 (86.0), AFG2 (93.0), AFM1 (48.0), AFM2 (21.0), AFP1 (100), AFL (100)[8]
Nigeria30HPLCAF (9.6)AF (100)[69]
Poland25HPLC-FLDAF (0.2)AF (4.0)[19]
South Africa(AE)10 (AP)10TLC, HPLC-FLD(SF) AFB1 (44.1)
(PF) AFB1 (20.1)
AFB1 (100)[20]
Turkey21ELISAAFB1 (6.6)AFB1 (100)[70]
Turkey18ELISAAF 1.75 a 20AF (16.7)[71]
BrazilRetrospective studyHPLCAFB1–AFG1 (89.0–191)-[72]
Definitions: AF: Total aflatoxins; AFB1: Aflatoxin B1; AFB2: Aflatoxin B2; AFG1: Aflatoxin G1; AFG2: Aflatoxin G2; AFL: Aflatoxicol; AFM1: Aflatoxin M1; AFM2: Aflatoxin M2; AFP1: Aflatoxin P1; ELISA: Enzyme Linked Immunosorbent Assay; HPLC: High Performance Liquid Chromatography; LC-MS/MS: Liquid Chromatography-Tandem Mass Spectrometry; LC-MS: Liquid chromatography coupled to mass spectrometry; PLC-MS/MS: Ultra performance liquid chromatography coupled to tandem mass spectrometry; UHPLC-Q-Orbitrap HRMS: ultra-high performance liquid chromatography coupled to high resolution mass spectrometry; HPLC-FL: Fluorescence High Performance Liquid Chromatography; HPLC-FLD with fluorescence detection (FLD); SF: standard feed; PF: premium feed; SPF: super premium feed.
Table 3. Acute poisoning in dogs due to ingestion of aflatoxins in commercial dry feed (CDF).
Table 3. Acute poisoning in dogs due to ingestion of aflatoxins in commercial dry feed (CDF).
LocationNumber of Intoxicated Dogs (n)Mortality (%)Range of AF in CDF (µg/kg)Citation
Brazil4100AFB1–AFB2 (89.0–191)[72]
Brazil2100AFB1 (83.2–150)[74]
Israel5068.0AF (80–300)[38]
South Africa10100AF (100–300)[75]
South Africa10096.0AF (<5–4946)[76]
United States9100AFB1 (223–579)[66]
United States7236.1AF (48–800)[77]
Table 4. Blood biochemistry and coagulation tests in aflatoxicosis in dogs *.
Table 4. Blood biochemistry and coagulation tests in aflatoxicosis in dogs *.
AnalyteA. IntoxicationB. Reference ValuesComparison (A/B)
Blood biochemistry
Total bilirubin (µM/L)130 (1.71–428)2.7 (0.00–5.1)48.1 (0.00–83.9)
ALT (U/L)598 (6.0–2278)59.5 (5.0–106)10.1 (1.20–21.5)
AST (U/L)178 (15.0–748)31.8 (9.0–56.0)5.6 (1.7–13.4)
ALP (U/L)284 (10.0–3477)67.3 (4.0–140)4.2 (2.5–24.8)
TP (s)41.4 (4.5–71)12.0 (6.0–18.0)3.4 (0.75–3.9)
APTT (s)34.5 (9.6–241)15.7 (10.0–23.8)2.2 (0.96–10.1)
GGT (U/L)10.4 (0.00–44.4)7.8 (0.00–19.0)1.34 (0.00–2.3)
Total protein (g/dL)4.9 (1.10–7.9)6.3 (5.4–7.1)0.78 (0.20–1.11)
Albumin (g/dL)2.5 (0.50–3.9)3.5 (2.8–4.1)0.70 (0.18–0.95)
Coagulation tests
FVII:C (% activity)32.0 (1.40–67.0)125 (50–200)0.26 (0.03–0.34)
Platelets (X109/L)156 (8.0–432)347 (143–700)0.45 (0.06–0.62)
Protein C (% activity)18.0 (4.0–55.0)105 (75–135)0.17 (0.05–0.41)
Cholesterol (mmol/L)1.26 (0.00–7.9)5.2 (2.6–8.6)0.24 (0.00–0.91)
Antithrombin (% activity)11.0 (0.00–147)105 (65–145)0.10 (0.00–1.01)
Fibrinogen (mg/dL)37.0 (11.0–344)305 (100–510)0.12 (0.11–0.67)
Definitions: ALT: Alanine aminotransferase; AST: Aspartate aminotransferase; ALP: alkaline phosphatase; PT: prothrombin time; APTT: active partial thromboplastin time; GGT: γ-glutamyltransferase; FVII: C: Coagulation factor VII. * Mean and range values adapted from: [38,66,77,79].
Table 5. Macroscopic and microscopic lesions in aflatoxin poisoning in dogs *.
Table 5. Macroscopic and microscopic lesions in aflatoxin poisoning in dogs *.
LocationMacroscopic LesionsMicroscopic Lesions
GeneralizedHemorrhagic diathesis, jaundice, and ascites.
HepaticHepatomegaly, uneven surface, pale yellowish discoloration, enhanced lobular pattern, cholestasis, and gallbladder edema.Hepatocytes with micro and macrovesicular steatosis, cytomegaly, pyknosis, karyorexis, and necrosis. Centrilobular areas with hemorrhage, reticulin, and collagen. Hyperplasia and proliferation of bile ducts.
PulmonaryAtelectasis, congestion, pleural effusion, hydrothorax, and petechiae.Alveoli with hemorrhage and perivascular edema.
CardiacEcchymosis and petechiae in the endocardium and epicardium. Hydropericardium.
DigestiveEdema and congestion in the gastrointestinal lumen, mesenteric lymph nodes and pancreas.Necrosis with mononuclear infiltration in the mucosa.
SplenicSplenomegalyDiffuse perivascular edema and red pulp with hemorrhages and erythrophagocytosis.
RenalDark red coloration with subcapsular depressions and multifocal hemorrhages.Fluid accumulation in Bowman’s space and glomerular basement membrane thickening. Multifocal vascular congestion in the interstitial tissue of the renal medulla and pelvis. Degeneration, ectasia, and necrosis of the proximal and distal tubular epithelium.
* Adapted from [66,74,75,76,83,84,85,86,87].
Table 6. Suggested therapy for aflatoxicosis treatment in dogs *.
Table 6. Suggested therapy for aflatoxicosis treatment in dogs *.
DrugDoseAdministration viaUsual Interval (h/d)Therapeutic Indications
Hemostatic Stabilization
Vitamin K12.0 mg/kgSC24/5Synthesis of coagulation factors
Intravenous plasma10.0 mL/kgIVUntil TP & TTPa are restored Correction of coagulopathy
Elimination of AF and Hepato-Renal Protection
Hartman solution40.0–60.0 mL/kg/dIVcbpIncreased glomerular filtration rate and restoration of water/electrolyte balance
N-acetylcysteine70.0 mg/kgIV12/15GSH synthesis and AF binding
Silymarin20 mg/kgPO24/30Counter AF epoxides
Vitamin E10.0 U/kgPO24/30Counter AF epoxides
L-carnitine50.0–100 mg/kgPO8/30Decreased liver lipidosis and counter epoxides
Symptomatic Treatment
Metoclopramide0.40 mg/kgSC8/3Antiemetic
Ondansetron0.15 mg/kgIV12/3Antiemetic
Famotidine0.5 mg/kgIV12/30Decreased gastric secretion
Sucralfate0.5–1.0 g/dogPO8/30Gastric cytoprotection
Ampicillin25.0 mg/kgIV8/7Broad spectrum bactericide
Enrofloxacin5.0–20.0 mg/kgIV12/7 díasBroad spectrum bactericide
Definitions: TP: prothrombin time; TTPa: activated partial thromboplastin time. * Adapted from [38,77,88,89,90].
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Martínez-Martínez, L.; Valdivia-Flores, A.G.; Guerrero-Barrera, A.L.; Quezada-Tristán, T.; Rangel-Muñoz, E.J.; Ortiz-Martínez, R. Toxic Effect of Aflatoxins in Dogs Fed Contaminated Commercial Dry Feed: A Review. Toxins 2021, 13, 65. https://0-doi-org.brum.beds.ac.uk/10.3390/toxins13010065

AMA Style

Martínez-Martínez L, Valdivia-Flores AG, Guerrero-Barrera AL, Quezada-Tristán T, Rangel-Muñoz EJ, Ortiz-Martínez R. Toxic Effect of Aflatoxins in Dogs Fed Contaminated Commercial Dry Feed: A Review. Toxins. 2021; 13(1):65. https://0-doi-org.brum.beds.ac.uk/10.3390/toxins13010065

Chicago/Turabian Style

Martínez-Martínez, Lizbeth, Arturo G. Valdivia-Flores, Alma Lilian Guerrero-Barrera, Teódulo Quezada-Tristán, Erika Janet Rangel-Muñoz, and Raúl Ortiz-Martínez. 2021. "Toxic Effect of Aflatoxins in Dogs Fed Contaminated Commercial Dry Feed: A Review" Toxins 13, no. 1: 65. https://0-doi-org.brum.beds.ac.uk/10.3390/toxins13010065

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