Next Article in Journal
In Vitro Antifungal Susceptibility Profile of Miltefosine against a Collection of Azole and Echinocandins Resistant Fusarium Strains
Previous Article in Journal
Correction: Yang et al. A Kinesin Vdkin2 Required for Vacuole Formation, Mycelium Growth, and Penetration Structure Formation of Verticillium dahliae. J. Fungi 2022, 8, 391
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Malassezia: Zoonotic Implications, Parallels and Differences in Colonization and Disease in Humans and Animals

1
Department of Veterinary Clinical Sciences, Jockey Club College of Veterinary Medicine and Life Sciences, City University, Tat Chee Avenue, Kowloon, Hong Kong, China
2
Department of Veterinary Medicine, University of Bari, Str. prov. per Casamassima Km 3, Valenzano, (Bari), 70010, Italy
3
Centre for Animal Health and Welfare, City University of Hong Kong, Kowloon Tong, Hong Kong, China
*
Authors to whom correspondence should be addressed.
J. Fungi 2022, 8(7), 708; https://doi.org/10.3390/jof8070708
Submission received: 15 June 2022 / Revised: 29 June 2022 / Accepted: 30 June 2022 / Published: 4 July 2022

Abstract

:
Malassezia spp. are commensals of the skin, oral/sinonasal cavity, lower respiratory and gastrointestinal tract. Eighteen species have been recovered from humans, other mammals and birds. They can also be isolated from diverse environments, suggesting an evolutionary trajectory of adaption from an ecological niche in plants and soil to the mucocutaneous ecosystem of warm-blooded vertebrates. In humans, dogs and cats, Malassezia-associated dermatological conditions share some commonalities. Otomycosis is common in companion animals but is rare in humans. Systemic infections, which are increasingly reported in humans, have yet to be recognized in animals. Malassezia species have also been identified as pathogenetic contributors to some chronic human diseases. While Malassezia species are host-adapted, some species are zoophilic and can cause fungemia, with outbreaks in neonatal intensive care wards associated with temporary colonization of healthcare worker’s hands from contact with their pets. Although standardization is lacking, susceptibility testing is usually performed using a modified broth microdilution method. Antifungal susceptibility can vary depending on Malassezia species, body location, infection type, disease duration, presence of co-morbidities and immunosuppression. Antifungal resistance mechanisms include biofilm formation, mutations or overexpression of ERG11, overexpression of efflux pumps and gene rearrangements or overexpression in chromosome 4.

Graphical Abstract

1. Introduction

Malassezia are small thick-walled ovoid, ellipsoid or cylindrical commensal yeasts of warm-blooded vertebrates. Their genome of approximately 10 Mb is almost half the size of Cryptococcus, another basidiomycete of medical and veterinary importance [1,2]. The mycelial phase of Malassezia spp. has been observed naturally in some skin lesions and induced in specialized culture media incubated at 30 °C [2,3,4,5,6]. Malassezia species reproduce asexually by unipolar broad-based budding. The sexual form has not been detected, although the mating-type locus region has been identified [7].
An important characteristic of all Malassezia is their dependence on lipids for growth due to an absent fatty-acid synthetase gene and consequent inability to synthesize long-chain fatty acids. Although one species, M. pachydermatis, can readily grow on Sabauraud’s dextrose agar (SDA), a medium without lipid supplementation, it is still lipid dependent and its growth in this medium is due to the use of lipid fractions within the peptone, a component of SDA [2,8,9,10,11].
M. furfur was first identified on human skin in 1846 [12], but recently the genus has received more attention, not only because of its association with dermatological diseases in animals (dermatitis, otitis externa) and humans (pityriasis versicolor, atopic dermatitis, Malassezia folliculitis, seborrheic dermatitis) [2,13,14], but also due to its increased detection in systemic infections, especially in neonates and immunocompromised patients [15,16,17,18].
M. pachydermatis, originally thought to be part of the mycobiome in dogs and cats only, has now also been isolated from humans, production animals and from multiple exotic and wildlife species such as the sea lion, scarlet macaw, brown bear, American black bear, Eurasian badger, big anteater, common wombat, Mangaliza pig, wide-mouthed rhinoceros, Indian elephant, red fox, porcupine and coyote [19,20,21,22].
In this review, we use the One-Health paradigm to explore similarities and differences regarding carriage of Malassezia species in humans and companion animals, antifungal susceptibility, resistance mechanisms, Malassezia-associated diseases and treatment. The available evidence for transmission between animals and humans, directionality of transmission, and clinical relevance are also discussed.

2. Classification of Malassezia Yeasts

Malassezia yeasts belong to the family Malasseziaceae, order Malasseziales and class Malasseziomycetes. They are included in the morphologically highly diverse subdivision of Ustilaginomycotina, and due to their filament (hyphae) and reproduction characteristics, they are contained in the division of Basidiomycota [23,24,25,26].
Thus far, 18 Malassezia species have been identified from a variety of mammalian hosts and birds (Table 1) and further expansion of the genus is likely [27]. For species differentiation, locus analysis of specific ribosomal gene sequences, such as ITS, D1/D2, ß-tubulin, chitin synthetase 2 and large subunit polymerase 2, is used. For phylogenetic stem evaluation and species delimitation, whole genome sequencing (WGS) is necessary [1,27,28,29,30,31,32,33,34,35].
Recently, after the WGS of 28 representative isolates from 15 Malassezia species, concatenated protein sequences of 254 conserved orthologues were included in a phylogenetic analysis to resolve the taxonomy of the genus [27]. Similar to previous analyses [1,26], all species fell into three distinct clades [27] (Table 1).

3. Malassezia Species in the Environment and Possible Vectors

Although first isolated from the skin of humans, followed by other warm-blooded vertebrates, recent data have shown that Malassezia species have a much broader spectrum of ecological diversity than originally thought [36,37,38]. These yeasts have now been isolated from a range of environments, including marine water, anoxic oceans, hydrothermal vents, deep-sea to high arctic marine sediment and Antarctic soil [36,39,40,41,42,43,44,45,46,47,48,49,50,51,52]. Malassezia species also dominate the mycobiome of marine invertebrates, such as sponges and corals, and have been identified in healthy and diseased marine algae [36,53,54]. In addition, Malassezia species have been isolated from soil nematodes, cone-snails, olive fruit-flies and orchid roots [55,56,57,58]. A potential role for nematodes and flies as vectors for Malassezia has been speculated [55,58,59,60].
It is now apparent that Malassezia species are among the most widespread fungi on Earth [36,37,38]. Their evolutionary trajectory involves adaptation from an ecological niche in plants and soil to the mucocutaneous ecosystem of animals [36,37,38]. This has been facilitated by the loss of complex carbohydrate metabolism genes (glycosyl hydrolase encoding) and a genus-wide gain of lipid hydrolases including lipases, phospholipases and acid sphingomyelinases that are required to degrade and use skin- or mucosa-associated lipids [1,36,37,38].

4. Malassezia Species and Their Role as Commensals in Humans

Twelve Malassezia species have been isolated from human skin [6,16,34,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75] (Table 1). Malassezia arunalokei is the only species isolated from humans that has not been isolated from animals, with the exception of dogs [74,76].
Malassezia species colonization of the skin starts directly after birth, increases until around 12 months of age, and then remains relatively static until puberty, when another significant quantitative increase in colonization occurs, associated with increased sebaceous gland activity and changes in the lipid composition of the skin [6,77]. After puberty, Malassezia species comprise 50 to 80% of the human mycobiome [78,79,80,100]. The limited data currently available about cutaneous mycobiomes in preterm and term neonates shows that Malassezia species distribution on the skin of neonates and children varies between studies, but M. globosa, M. furfur, M. sympodialis and M. restricta are the most prevalent species described [14].
In contrast, M. restricta and M. globosa dominate the mycobiome of both healthy and diseased skin in adult humans, followed by M. sympodialis, albeit at a much lower frequency than the former two [1,62,69,81,82,83,84,85,86,87,88,89,90,91]. M. furfur can be common at certain body sites (e.g., toe-web space) in healthy individuals but is not a dominating species overall. Instead, this species is more frequently isolated from skin diseases, such as psoriasis vulgaris and pityriasis versicolor [1,5,73,89,90,91,92,93].
Climate and ethnicity also impact the carriage of Malassezia species [94,95]. In a study by Leong et al. in 2019, people in Singapore of four different ethnicities (Chinese, Malay, Indian and Caucasian) carried a higher number of Malassezia species and showed greater species diversity and evenness than Caucasians in Switzerland. The predominant species (isolated by culture from the skin of the side of the nose) in the latter were M. restricta and M. sympodialis, while M. globosa was absent. In contrast, sampling from the same site in the four ethnic groups in Singapore showed M. globosa, M. furfur and M. restricta were the dominant species. Caucasians from the two locations showed different species distributions, with M. restricta being twice as common among those in Singapore, while M. globosa was absent in Swiss Caucasians. The same study associated the lower temperature and humidity of Switzerland compared to Singapore with a lower positive culture rate and lower species diversity [95] (Figure 1). From other studies, it can be concluded that M. restricta plays a dominant role as a skin commensal in Europe, whereas M. globosa comparatively dominates in Asia [13,96].
Several studies have shown that sex and body site also influence the species of Malassezia species present on the skin and their abundance [1,88,91,97,98,99,100]. Site-specific species include M. restricta, which favors colonization of the external ear canals, retroauricular crease and forehead and M. globosa, which is most commonly isolated from the back, occiput and groin [100,101].
A Japanese study in 2010 quantified Malassezia colonization of the skin of the cheek using real-time PCR and determined associations with gender and age in 770 healthy individuals [99]. Total Malassezia DNA in males stayed constant from age 0 until around 9 years of age, with a progressive increase each year thereafter until the age of 16 to 18. In females, total Malassezia DNA increased until the age of 12, decreased between the ages of 19 and 22, and then increased again between the ages of 30 and 39. In both genders, there was a gradual decrease in Malassezia species abundance over the course of life. Overall, males tended to have more abundant Malassezia DNA than females, and M. globosa and M. restricta were the dominant species for both for all ages.
Malassezia species carriage at different skin locations was investigated using culture-based methods. No significant differences between the genders were found. While M. restricta dominated the scalp and M. sympodialis dominated the trunk, M. globosa was about equally common at both locations [102].
Other factors that may influence the colonization of Malassezia species include host factors (immune response, body secretion, skin occlusion), other skin inhabitants (e.g. parasites, other microbes) and environmental parameters, including exposure to ultraviolet light [96]. Even the birth process itself has a significant impact. If a baby is born via natural delivery, its skin microbiota resembles the mother’s vaginal communities, but if delivered via caesarian section, it represents the mother’s skin surface population [103,104,105,106,107]. In addition, vaginal birth is associated with a higher abundance of Malassezia [108,109].
Malassezia species were previously thought to be commensals of the skin only. Although the skin is the primary ecological niche, more recent data demonstrate that these yeasts also colonize the mucosa of the sinonasal and oral cavities, as well as the gastrointestinal and lower respiratory tract [110,111,112,113,114,115]. Malassezia species are dominant members of the mycobiome of the sinuses, with M. restricta and M. sympodialis most frequently detected [116]. Malassezia also comprise 30% of the gastrointestinal mycobiome, with three species detected—M. globosa, M. restricta and M. pachydermatis [117]. The fungal burden in the lungs of healthy people is relatively low. In one study, using a metagenomic approach, the lung mycobiome was characterized by a high proportion of basidiomycetes, including M. restricta and M. globosa [118], while in another ascomycetes, including Candida species, were most abundant [119] (Figure 1).

5. Malassezia Species and Their Role as Commensals in Companion Animals

Using culture-based techniques, Malassezia species have been identified as the most common yeast colonizing healthy canine skin [120,121]. Metagenomic approaches reveal that, in contrast to humans, Ascomycota, especially Alternaria and Cladosporium species, are the most abundant fungal species on the skin of healthy dogs and cats [122,123].
Overall, eleven Malassezia species have been isolated from cats and seven from dogs [2,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142] (Table 1). Culture-based studies clearly favor M. pachydermatis as the dominant species colonizing the skin of dogs and cats [124,125,126,128,135,143,144,145,146]. In one recent study using metagenomics and quantitative PCR (qPCR), M. restricta and M. globose, but not M. pachydermatis, were identified as the dominant species colonizing healthy feline skin [139].
Malassezia abundance and species diversity are influenced by body site, genetic predispositions and concurrent diseases [2,135,136,137,140,141,146,147,148]. M. pachydermatis is more frequently isolated from dogs from perioral and interdigital skin than from the back or ventral body sites, such as the axillae or groin [2] (Figure 2).
In cats, the external ear canal is most commonly colonized by M. pachydermatis, followed by other species such as M. furfur, M. globosa, M. sympodialis, M. obtusa and M. nana [134,142,149,150,151,152]. M. nana is the most common skin and ear isolate after M. pachydermatis, with one specific genotype dominating [136,137]. Claw folds of cats are a particular niche for M. slooffiae [135,136,137] (Figure 2).
Two specific feline breeds, the Devon Rex and Sphynx, harbor high Malassezia species loads, with a dominance of M. pachydermatis [135,146,147]. Cats with otitis externa also have a higher abundance of Malassezia species in the ear canals compared to healthy individuals. The most prevalent Malassezia species were M. pachydermatis (57.7–62.62%), followed by M. globosa (11.4–22.2%), M. furfur (2.4–17.2%), M. obtusa (15.4%), M. slooffiae (7.3%), M. sympodialis (1–4.1%) and M. restricta (1.6%) [134,152].
In dogs, similar to cats, M. pachydermatis dominates the healthy ear canal, although other species, such as M. sympodialis and M. obtusa, can also be detected [142,149,150,153] (Figure 2). In diseased canine ears, the colonization rate increases, but M. pachydermatis remains most prevalent, followed by M. sympodialis, M. furfur, M. obtusa, M. globosa and M. restricta. This has mostly been shown by cultural and biochemical evaluation methods [142,150,153,154].
Allergic dogs have a higher abundance of Malassezia species, especially of M. pachydermatis, compared to healthy dogs [140,148] as well as a decreased overall diversity [155].
In particular, in dogs with atopic dermatitis, the isolation of M. sympodialis associated with M. pachydermatis and/or M. furfur has also been reported [156]. The coexistence of M. pachydermatis with other Malassezia species makes the pathogenic role determination of a single Malassezia species challenging.
Other body sites where Malassezia species are commensals have not been well characterized in dogs and cats. Low numbers of M. pachydermatis are present on the corneal surfaces of healthy dogs [120].
In a recent study, the mycobiome of the healthy canine oral cavity was found to be dominated by Malassezia species and Cladosporium species. Malassezia arunalokei, M. restricta, M. pachydermatis and M. globosa have all been detected in the oral cavity of healthy and diseased dogs (Figure 2), although no correlation was found between these Malassezia species and periodontal disease [76].
In the gastrointestinal tract of dogs, Ascomycota and Basidiomycota are the most numerous, with Candida as a major genus [157,158,159,160]. Malassezia species have not yet been detected. In cats, even less data are available, but Ascomycota seem to be dominant [158].
The urinary mycobiome of healthy dogs was recently characterized, and reads of several fungal genera in very low abundance were detected, including M. restricta [161].

6. Malassezia Species in Other Animals

Malassezia species have been isolated from multiple mammalian and avian species (Table 1). In pigs, Malassezia species, especially M. sympodialis and M. slooffiae, were isolated from 73% of healthy ear samples but not from multiple skin sites [162]. M. pachydermatis has been isolated from pigs with otitis externa and from the skin of healthy farmed pigs [163,164]. One study [165] compared Malassezia species detection rates and species from healthy porcine ears among different porcine breed and age groups, showing that, similar to humans and companion animals, genetic factors and age can impact Malassezia abundance and diversity [2,85,95,141,145,152,166]. Overall, Malassezia were isolated from 22.5% of sampled pigs, including M. pachydermatis, M. furfur and M. sympodialis. M. pachydermatis was found in all breeds but not in adults of large breeds, whereas M. furfur and M. sympodialis were only present in adult pigs of large breeds [165].
Several Malassezia species were isolated from multiple skin sites of 12 to 60% of healthy cattle using culture-based techniques [149,167] (Table 1). One study showed a clear difference in the species isolated in cases of otitis externa depending on the time of the year, with thermotolerant M. sympodialis dominating in summer and less thermotolerant M. globosa being predominant in winter [168].
A variety of Malassezia species have been isolated from the skin of horses, sheep and goats (Table 1). In goats, skin infections have been associated with M. pachydermatis and M. slooffiae [169,170].
Among rabbits, M. cuniculi is commonly detected in healthy skin and ears [30,171]. In one study, rabbits bred for meat consumption were more frequently colonized with Malassezia species compared to pet rabbits [171]. In contrast to humans, Malassezia species were more commonly present in young rabbits (<3 months of age), and diet impacted Malassezia species prevalence.
Among different bird species, Malassezia species have been isolated from healthy and diseased sites, including beak (M. brasiliensis, M. psittaci), feathers and wings (M. pachydermatis, M. furfur), oropharynx (M. pachydermatis, M. furfur, M. brasiliensis, M. psittaci), and feces (M. pachydermatis, M. furfur) [20,125,172]. M. sympodialis has also been commonly found in diseased combs of adult chickens [173].

7. Zoonotic and Reverse Zoonotic Transmission of Malassezia Species

There is now ample evidence that different Malassezia species are shared between humans and animals (Table 1). However, some genotypes within a species might be host adapted or linked to a particular host site location or skin disorder [27,30,31,74,110,174]. In particular, sequence analyses of the LSU rDNA showed distinct Malassezia species subtypes on different host species [110]. Sequence analysis of IGS1 distinguished specific M. globosa, M. restricta, and M. pachydermatis variants in seborrheic dermatitis and atopic eczema and on the healthy skin of humans and animals [85,174]. Among M. pachydermatis, eight IGS1 subtypes were identified and subtype 3D was mainly associated with skin lesions [175]. Additionally, M. pachydermatis, frequently isolated from cats and dogs [176,177,178,179,180], but rarely from human skin [62,181,182] was known to cause fungemia in people, especially in neonates [34,63,183,184,185,186,187,188,189,190,191]. However, newborn babies have skin colonization by M. sympodialis and M. globosa, but not by M. pachydermatis [78,79,80,100]. Thus, the ease with which these yeasts can be transmitted from one body site to another [192] or between animals and their owners [182] makes us hypothesize that zoonotic and reverse zoonotic transmission of these yeast species can occur.
In particular, the carriage of M. pachydermatis in healthy and diseased dogs with allergic dermatitis or otitis externa was compared to healthy human owners [182]. M. pachydermatis DNA was identified on the palms of over 90% of pet owners, regardless of the disease state of their dogs. Based on culture results indicating the relative abundance of Malassezia species, owners of affected dogs were 11 times more likely to be culture positive than owners of healthy dogs [182].
The zoophilic potential of M. pachydermatis was first postulated by Dr. Gueho [193] but was clearly confirmed ten years later when an outbreak of neonatal fungemia caused by M. pachydermatis was investigated [184]. The strain implicated in the outbreak was isolated from a health care worker’s hands, from contaminated equipment and from dogs belonging to three health care workers working in the involved intensive care nursery unit. One or several healthcare workers likely contaminated the nursery environment and their patients after transient colonization of their hands by the organism. After optimizing hand hygiene, no further cases were reported and all cultures from staff members tested negative [184].
Other studies have also demonstrated that hospitalized infants can be colonized by Malassezia species, especially M. pachydermatis and M. furfur, via contact with their parents or healthcare workers or indirectly via incubator surfaces [16,77,183,184,194,195,196]. Healthcare workers can then further transmit the organism from one infant to another via their hands. Through this mechanism, several Malassezia species outbreaks have occurred in the past [184,197,198].
Carriage of M. pachydermatis in humans was detected in low numbers on the scalp and palms of 12% of healthy individuals in one study [181], and on the skin of 5% of healthy medical students in another [62]. In other studies it was not detected at all in healthy individuals, and overall appears to be a rare, transient colonizer of human skin [6,102]. Similarly, other studies have found no causal associations between M. pachydermatis and human Malassezia-associated skin conditions, including seborrheic dermatitis and pityriasis versicolor [62,199].
While there is evidence that M. pachydermatis can be transmitted between dogs and humans, further investigations into the genotypes involved, and the strain characteristics are warranted [184,188,189,190,200]. The relatively recent discovery of M. pachydermatis as a commensal of the human gut introduces another potential reservoir of infection in humans by this species [117].
There is phenotypic and phylogenetic evidence that species with high host diversity, such as M. furfur, are undergoing diversification to enable successful adaptation to different hosts [201]. Strains from different animal species remain closely genetically related, but the extent and frequency of zoonotic or reverse zoonotic transmission have not been investigated.

8. Superficial Malassezia-Associated Diseases in Humans and Animals

8.1. Malassezia-Associated Dermatological Diseases in Humans

The most common Malassezia-associated skin diseases in human patients are pityriasis versicolor, seborrheic dermatitis, Malassezia folliculitis and atopic eczema [6,13,38,202,203,204]. The skin sites and species involved in these diseases are shown in Figure 3.

8.1.1. Pityriasis Versicolor

Pityriasis versicolor, sometimes called tinea versicolor, is a common disease worldwide, with a prevalence of up to 50% in hot and humid regions. There is no gender or ethnic predisposition [205,206]. The disease is most commonly seen in young adult to adult patients, correlating with increased sebaceous gland activity and altered lipid composition of the skin around this time. The disease is clearly associated with Malassezia species, especially M. furfur, M. globosa and M. sympodialis. A combination of factors including genetics, warm and humid environment, immunodeficiency, pregnancy, oily skin or application of oily topical substances lead to a transformation of resident Malassezia species into a pathogenic filamentous form. Most patients have multiple affected areas characterized by well-demarcated, oval, hyper- or hypopigmented macules with a fine scaly surface. These lesions are variably pruritic and the neck, trunk and proximal extremities are commonly involved [207,208,209] (Figure 3). Diagnosis is usually made clinically, but in individual cases, Wood’s lamp examination (coppery-orange fluorescence) or microscopic examination of fungal elements may be needed [6,210,211].

8.1.2. Seborrheic Dermatitis

Seborrheic dermatitis also occurs worldwide, with ‘normal’ and dandruff forms affecting around 5% and up to 50% of the population, respectively. There is also an HIV-associated form. There is no ethnic predisposition, but males are clearly predisposed. Disease is mainly seen in infants and adults [212,213,214]. The etiology is not completely clear but involves an interplay of skin flora, lipid composition on the skin surface, skin barrier integrity, immune response to Malassezia species and individual host factors. Increased sebaceous gland activity, immunodeficiency, neurological and psychological diseases, certain drugs and environmental factors such as low humidity and temperature are risk factors for seborrheic dermatitis [213,215,216]. M. restricta or M. globosa are typically isolated from active lesions and antifungal treatment usually leads to significant clinical improvement. Other species can be isolated, including M. furfur, M. sympodialis, M. obtusa and M. slooffiae [38,217,218,219]. The scalp, face and chest are most commonly affected, although in infants, the diaper area, neck and axillae may also be involved (Figure 3). Skin lesions are often inflamed, pruritic and present at one or multiple locations. They include poorly defined follicular papules and plaques, fine white scales, and yellow crusts. In the mild dandruff form, no inflammation but a fine, mild scaling on the scalp and beard dominates [220,221].

8.1.3. Malassezia Folliculitis

Malassezia folliculitis is another common worldwide disease with a prevalence of 1 to 17%. It occurs more commonly in young to middle-aged adult males [222,223,224]. Follicular occlusion or a disturbance of the normal cutaneous flora leads to an abnormal proliferation of Malassezia species and the development of disease. Common associated species include M. globosa, M. restricta and M. sympodialis [6,202,224,225,226,227,228]. Predisposing factors include hot and humid climate, excessive sweating, non-breathable clothing, application of make-up or sunscreens, certain drugs (antibiotics, glucocorticoids) and immunosuppression [6,224,229,230]. The disease typically involves the face, upper back, extensor surfaces of the arms, chest and neck (Figure 3). In almost 75% of cases, more than one location is affected. Lesions include small but pruritic follicular papules and pustules. This presentation is often mistaken for acne or bacterial folliculitis [223,224,231,232].

8.1.4. Atopic Dermatitis (Head and Neck Dermatitis)

Atopic dermatitis (AD) is a common, chronic, inflammatory and pruritic disease, affecting 10 to 25% of children and 1 to 2% of adults. Head and neck dermatitis (HND), a subtype of AD, mostly occurs in adolescence and adulthood in individuals with a history of IgE-mediated AD. There is no gender or ethnic predisposition [233,234,235,236]. The etiology is incompletely understood, but it is clear that Malassezia species play an important role in disease pathogenesis. The high activity of sebaceous glands at affected sites, together with the skin barrier disruption of the atopic disease, allow Malassezia species to proliferate, leading to increased exposure to the immune system, triggering a humoral and cell-mediated immune response [237,238,239,240,241,242,243]. Some involved Malassezia antigens have been well characterized (M. globosa—MGL_1304; M. sympodialis—Mala s 8; M. restricta—Mala r 8) and have been identified in the sweat of patients, leading to aggravated clinical signs, especially after intense sweating [244,245]. These antigens have also shown variable histamine-releasing properties [246]. Malassezia species isolated from disease-associated sites have included M. furfur, M. obtusa, M. globosa, M. restricta and M. sympodialis, but there was no significant difference in isolation compared to healthy individuals [240,247]. Others found a higher colonization rate by M. furfur, as well as a lower colonization rate by M. globosa and M. sympodialis, in affected AD patients [62]. Specific genotypes of M. globosa and M. restricta have also been identified as colonizing AD skin [65,174].
HND patients have erythema and erythematous plaques on the forehead, eyelids, perioral, neck and upper trunk together with variable pruritus (Figure 3). In severe cases, the whole face may be involved, leading to the term “red face”. With the chronicity of the disease, lichenification and scaling can occur [245,248]. Wheal-like, edematous changes have also been described [245].

8.2. Malassezia Dermatitis and Otitis Externa in Animals

In dogs and cats, Malassezia dermatitis and otitis externa are commonly encountered in daily practice [141,156,249] but they can also be seen in farm animals, especially horses and goats. The prevalence of Malassezia-associated skin diseases in farm animals may be underestimated [169,170,250,251,252,253,254,255,256]. Malassezia dermatitis and otitis externa have also been reported in many other animals, including sea lions, fennecs, okapi, dromedaries, rhinoceros, canaries and pinnipeds [21,163,257,258,259,260,261,262].
Concurrent Malassezia dermatitis and sarcoptic or demodectic mange are occasionally seen in lagomorphs or hamsters, respectively [263,264,265]. Some specific dog and cat breeds have a higher risk of Malassezia dermatitis [135,145,266,267,268,269] (Table 2).
In veterinary Malassezia-associated dermatitis, typical cutaneous manifestations include alopecia, erythema, scaling, crusts and accumulation of greasy, malodorous, brown to black keratosebaceous debris. In chronic infections, lichenification and hyperpigmentation may also be present. The intensity of pruritus is variable [2,141,179,254,255,256,260,265,266,270,271].
In canine patients, an infection or overgrowth with Malassezia species is most commonly associated with allergic diseases (flea bite hypersensitivity, food allergy, atopic dermatitis), ectoparasitic infestations, superficial pyoderma, occasionally with endocrinopathies (hypothyroidism, hyperadrenocorticism, diabetes mellitus), keratinization disorders and rarely with autoimmune diseases [272,273,274,275]. Common involved areas include the external ear canal, pinnae, lips, muzzle, ventral neck, ventral body sites, medial hind limbs, perianal site and paws [270,276] (Figure 4).
In dogs with environmental allergies, the clinical signs of Malassezia dermatitis often mimic, or even worsen, those of atopic disease [270]. It has been shown that affected patients show elevated levels of Malassezia-specific IgG and IgE in their serum [277]. In addition, immediate hypersensitivity reactions were observed in canine atopic patients in which M. pachydermatis extracts were intradermally injected or after passive transfer of atopic serum to healthy recipient dogs using the Prausnitz–Küstner (P-K) technique [278,279]. Together with the frequent isolation and higher colonization rate of Malassezia species on and from the skin of these patients, their relevance and contribution, especially M. pachydermatis, to disease pathogenesis has been demonstrated [140]. Four major allergens of M. pachydermatis with a size of 45, 52, 56 and 63 kDa were detected in more than 50% of atopic dogs in a study by Chen et al. in 2002 [280].
While Malassezia dermatitis in cats can be associated with similar diseases in dogs, especially skin fold dermatitis and hypersensitivities [2,141,281,282] there are also more exclusive presentations, such as idiopathic facial dermatitis [141,270,283,284,285], acne [2,141,286,287], paraneoplastic alopecia [2,141,288,289,290,291,292,293,294], thymoma-associated exfoliative dermatitis [2,141,295,296], FIV-associated dermatitis [2,141,297], Feline leukemia virus or Feline immunodeficiency virus infection [298] and superficial necrolytic dermatitis [2,141,299]. In most cats, common affected body regions include the pinnae, face, chin, neck, limbs and abdomen, while in Devon Rex and Sphynx cats, the ventral neck, axillae, groins and paws dominate [2,145,166] (Figure 4).
Malassezia-associated otitis externa in animals can be unilateral or bilateral and is associated with ear scratching, head shaking and brown to black, often malodorous discharge. The pinnae, especially near the orifice of the ear canal, are often also affected [2,141,252,300]. In a recent canine study, a painful, erosive to ulcerative form of otitis externa with a watery brown to black discharge caused by Malassezia species was described [301]. In contrast, otomycosis due to Malassezia spp. is considered rare in humans [302,303].

8.3. Miscellaneous Forms of Superficial Malassezia-Associated Diseases

Occasionally, Malassezia species can also infect the nails of humans and the claws of animals. In companion animals, paronychia with erythema, swelling and a waxy brown to black discharge is common, while in humans, subungual hyperkeratosis and onycholysis can be seen [85,141,304,305,306].
Another potential site of superficial Malassezia infection is the cornea. There are sparse reports of keratomycosis in humans and dogs associated with M. furfur and M. restricta in humans and M. pachydermatis in dogs [307,308,309]. Interestingly, one affected dog [309] and a human patient [308] both had diabetes mellitus and in all described cases immunomodulatory or antibiotic drugs were used. These predisposing factors could have facilitated Malassezia species overgrowth. The burden of corneal colonization by Malassezia species significantly increases in cases of corneal ulceration [120,310]. Whether Malassezia species could have a primary pathogenic role in some cases of corneal ulceration requires further investigation.

9. Systemic Infections and Chronic Malassezia-Associated Diseases in Humans and Animals

9.1. Fungemia and Systemic Infections

Of the 18 Malassezia species, only three are known to cause fungemia—M. furfur, M. pachydermatis and M. sympodialis. In the former two, one specific genotype is involved [14,198,311,312]. In fungemic patients, M. furfur is most frequently isolated, followed by M. pachdermatis and M. sympodialis [14,16,313].
Since the first report of systemic infection by an unspecified Malassezia species in 1979 [314], systemic infections have been described with increasing frequency [14,15,16,18,198], likely due to growing recognition of the pathogenic potential of Malassezia species, as well as improved detection methods [14,110].
The skin plays a significant role in the development of fungemia as both a reservoir of Malassezia species and a portal of entry into the bloodstream by Malassezia species when it is compromised [14]. Predisposing factors for fungemia include premature birth, hospitalization and duration of stay in a neonatal intensive care unit, immunosuppression, peritoneal dialysis, presence of central venous catheter, total parenteral nutrition with lipid supplementation (especially in neonates), invasive surgical procedures, long-term or broad-spectrum antimicrobial administration, chronic illnesses and topical application of soybean oil containing products [16,184,185,315]. Parenteral lipids are not only favorable for Malassezia species growth but can also reduce the immune response of a patient by the generation of reactive oxygen species, which decrease neutrophil phagocytosis [183,184,316,317].
The pathogenesis of Malassezia fungemia is not fully understood. Since only particular genotypes of M. furfur or M. pachydermatis are associated with fungemia, pathogen virulence factors are likely important determinants of systemic infection [198,311,312]. Malassezia species possess a number of virulence factors, including lipases, phospholipases, metabolites (indirubin, indole carbazole, pityriacitrin and others), nanovesicles, cell membrane µ-opioid receptors, hydrophobicity, adherence and the ability to form biofilm [38,318,319,320,321,322,323,324,325]. Of these, increased phospholipase activity and the release of allergen-enriched nanovesicles are often related to more severe disease and fungemia [312,318,321,322,326].
Pathogenic Malassezia strains associated with fungemia are either already present colonizing the patient’s skin or are transmitted to the skin through interactions with healthcare worker’s hands or contaminated medical devices, materials and/or parenteral solutions [14,16,77,183,184,194,196].
Severe illness, the administration of immunosuppressive, antifungal or broad-spectrum antimicrobial drugs or parenteral lipids, poor anatomic conformation and/or premature age lead to an impaired immune state. Different combinations of such factors enable invasion of the body at an entrance point, such as a surgical wound or an intravenous catheter site [14,38,194,327,328].
Hematogenous dissemination of Malassezia species can involve infection of the heart, lungs and, less commonly, the kidneys, pancreas, liver, spleen, brain and skin (multiple cutaneous pustules) [183,184,316,317]. Biofilm formation facilitates local replication and further shedding of the organism into the blood system [317,329,330,331].
Systemic infections with Malassezia species include a broad range of presentations, from single-organ infection to fungemia, and can be fatal. Single-site infections include meningitis [332,333], endocardial mass [334], pneumonia [335,336], peritonitis [314,337,338], osteomyelitis [339], septic arthritis [339], sinusitis [340] and mastitis [341].
Clinical signs of systemic Malassezia species infection in infants include fever, respiratory distress from pneumonia or bronchopneumonia, lethargy, bradycardia, seizures and cyanosis. Infected infants often show hepato- and splenomegaly. The main hematological findings are leukocytosis or leukopenia and thrombocytopenia [183,184,185,342,343,344,345].
Infections in children and adults are characterized by fever, chills, myalgia, nausea, vomiting and respiratory distress. Haematological findings include leukopenia (rarely leukocytosis) and thrombocytosis [329,346,347,348].
The diagnosis of Malassezia-associated fungemia is challenging due to its special needs for growth including lipid dependency. It is recommended to directly culture blood or central venous catheter tips on lipid-rich culture media via blood culture specimen tubes and not to use an automated blood culture system [14,313,349]. In addition, since human blood can have inhibitory and toxic effects on yeasts, the addition of 3% palmitic acid may favor positive detection [350].
Thus far, Malassezia-associated fungemia has not been reported in animals.

9.2. Chronic Diseases in Humans and Animals

In patients with HIV infection, the burden of the Malassezia species in the gut and on the skin of individuals with seborrheic dermatitis is significantly increased, associated with low numbers of CD4+ helper cells/Th17 cells. This overgrowth of Malassezia species is a risk factor for fungemia and other Malassezia-associated infections, including HIV-associated seborrheic dermatitis [115,351,352,353].
In patients with inflammatory bowel disease (IBD), including Crohn’s disease, Malassezia species dominate the gastrointestinal mycobiome [354,355,356]. M. restricta colonization, especially in the sigmoid colon, can increase disease severity by intensifying the inflammatory response [356]. This effect is strongly associated with the presence of the Crohn’s disease risk allele altered caspase recruitment domain 9 (CARD9 S12N). CARD9 is an adapter protein of the CARD-CC family that mediates pattern recognition signaling and is essential for fungal defense [115,354,355,356,357,358,359]. In mice models, the same authors showed the capability of M. restricta to cause significant changes to the colon, including colon shortening, mucosal erosion and crypt destruction [356].
It has been speculated that Malassezia species could have a pathogenic role in the development or progression of colorectal cancer since affected individuals have gastrointestinal mycobiome dysbiosis with a higher burden of Malassezia species compared to healthy individuals [115,360,361,362]. However, whether this is an effect or a cause of cancer remains to be proven. Malassezia species have been found to play a causal role in pancreatic ductal adenocarcinoma (PDA) associated with migration from the gut to the pancreas [361]. In human and murine PDA, cancerous pancreatic tissue contained a 3000-fold higher burden of fungi compared to healthy pancreatic tissue and was specifically enriched for the Malassezia species. The oncogenic pathway was also identified as the activation of mannose-binding lectin, which drives the complement cascade and promotes oncogenesis [361].
A role for Malassezia species in neurodegenerative diseases, such as Alzheimer’s disease and Parkinson’s disease, has been speculated due to their frequent detection in affected areas of brain tissue [363,364,365,366]. The source of Malassezia species is fungemia due to breaches of the cutaneous or gastrointestinal barriers. However, whether their presence reflects opportunistic colonization of damaged tissue or is causal has not been determined. Similar studies in veterinary medicine are lacking.

10. Antifungal Susceptibility Testing

10.1. Methodology

Usually, established testing concentrations are used as a reference for systemically applied drugs at their recommended doses [367,368]. Topically, much higher concentrations can be reached, important for topical therapy and thereby susceptibility testing methods would need to be adjusted [369,370,371]. Since Malassezia species are involved in common diseases and can potentially cause deep infections, fungemia or even death, susceptibility testing becomes a necessary and very important tool [14,15,16,18,110]. Even if there are standard proposed guidelines for testing the susceptibility profile of filamentous fungi and yeasts, it is difficult to implement these methods with Malassezia species due to their special needs and growth characteristics [372,373]. As a consequence, and due to the lack of standardization, different procedures were proposed with culture medium, inoculum size, incubation time, and criteria used to determine MIC endpoints largely vary among the studies, thus making it difficult to interpret the data in the literature [374]. The susceptibility of Malassezia species to antifungal compounds has been tested using different methods, including a modified Clinical and Laboratory Standards Institute (CLSI) broth microdilution protocol [375,376,377,378] and agar-based diffusion methods (Disk Diffusion – DD and the E test—ET) [379,380,381,382,383]. However, the agreement analysis between agar-based diffusion methods and modified CLSI standard reference procedures still needs to be better investigated. Overall, DD might not represent a valid alternative for determining the susceptibility of Malassezia yeasts to azoles and amphotericin B (AmB), and ET should be used with specific media and longer reading times and only for specific drugs [374].
A completely different approach has been described via corneofungimetry. Stratum corneum cells coated with olive oil form the basement of this testing process, mimicking an in vivo situation [384,385,386]. There is no comparison of this principle with commonly used methods.
Overall, clear international standard guidelines for susceptibility testing of Malassezia are urgently warranted to effectively compare and analyze data, but the authors consider the broth microdilution method the most suitable one and regard this as the gold standard.

10.2. Patterns of Antifungal Susceptibility

For clinical usability, not only the MIC distribution but also other factors such as serum concentration of the drug, pharmacodynamics, resistance mechanisms and clinical efficacy need to be considered [387,388,389]. These are encompassed by clinical breakpoint values established by the CLSI and EUCAST [372,388,389,390]. These breakpoints are regularly updated and if not available, usually the ones for Candida, including C. krusei, C. parapsilosis, C. tropicalis and C. albicans, are considered [391].
Nevertheless, the final proof of resistance is through the detection of the underlying mechanism. For Malassezia yeasts, clinical breakpoint values are still not established, but proof of the underlying mechanisms of resistance has been verified for some Malassezia species (see below).
Overall, Malassezia species antifungal susceptibility profiles against azoles, AmB and terbinafine (TER) vary between species or intraspecies, regardless of the media or other conditions employed [374]. M. sympodialis and M. pachydermatis are reported to have lower MICs of antifungals AmB, TER and azoles, in general compared to M. furfur and M. globosa [38,195,378,392,393,394].
MIC variation can also be seen within a given species, as shown for M. sympodialis, M. globosa and M. furfur [376,378,393,395]. Similar results are reflected in a canine study involving M. pachydermatis, indicating less variation within the same patient but more dissimilarity between different patients [394,396].
Malassezia spp. bloodstream isolates have higher MICs for the same antifungal drug compared to skin-origin isolates [392,393,397,398,399,400]. Accordingly, the disease status can affect the MIC, as shown in dogs [394,401,402,403]. Patients with prior antifungal exposure showed higher values than healthy individuals. In an in vitro evaluation, strains from diseased dogs showed higher MIC values across several azole drugs, including fluconazole (FCZ), ketoconazole (KZ), miconazole (MIZ), itraconazole (ITZ), voriconazole (VCZ) and posaconazole (PSZ), compared to strains from healthy individuals [404]. Weiler and colleagues found M. pachydermatis isolates from diseased animals to be less susceptible to AmB, nystatin, FCZ, clotrimazole (CL) and MIZ [402]. In an Asian study, high MIC values for KZ and ITZ were found among isolates of atopic dogs compared to their healthy counterparts [403].
Not surprisingly, the duration of a disease influences the MIC, as reflected in a canine study on otitis externa, in which patients with chronic disease had higher MIC values associated with MIZ and CL than those with an acute form [394,405]. This could also be related to prior antifungal exposure.
Studies focusing on fungemia have shown a better efficacy of AmB against M. pachydermatis than against M. furfur [393,395,406]. For M. furfur, better effects can be achieved when using the liposomal version of the drug or when combined with FCZ [393,407]. TER works better for M. pachydermatis and M. sympodialis than for M. furfur [395,396,397]. Considering Malassezia species overall, ITZ and KZ are reported to be more effective than FCZ, VCZ or AmB [195,392,393,395].
Nevertheless, looking at various reports, it can be concluded that for M. pachydermatis, ITZ and PSZ show the highest activity compared to other antifungals, with an MIC 90 of mostly less than 0.5 μg/mL. On the other hand, CL (up to 16 μg/mL) and thiabendazole (up to 32 μg/mL) show relatively high values [382,393,396,408,409]. However, from a clinical perspective, MIC 90 (values at which the growth of 90% of the tested isolates is inhibited) warrants careful interpretation since tissue concentrations are not included in the calculation.

11. Resistance Mechanisms

Antifungal resistance can be primary (intrinsic) or secondary (acquired) [410]. The former occurs naturally without previous exposure to antifungal drugs. Acquired resistance takes place after or during interactions with antimicrobials [410].
An early study in 1994 showed that resistant-induced mutant strains of M. pachydermatis exhibited significantly decreased levels of membrane sterols but increased amounts of fecosterol, indicating a possible evasion mechanism of polyene antifungals by replacement of sterol with a precursor product [411]. Mutations in the gene ERG11 (CYP51), encoding for lanosterol-14α-demethylase, which converts lanosterol to ergosterol, have been detected for induced KZ-resistant M. pachydermatis and for clinically resistant M. globosa strains. These mutations include missense mutations, amino acid alterations and tandem quadruplication and confer azole resistance [412,413]. Chromosomal rearrangements and gene overexpression, leading to tandem quadruplication of genes within chromosome 4, have been identified in some mutant-resistant strains. Since this region carries genes, including ERG 4 and 11, affecting ergosterol synthesis, azole resistance was conferred by this resistance mechanism [413].
Overexpression of ERG11 can also lead to resistance due to the overwhelming presence of the target protein, which has been demonstrated in clinical isolates of M. pachydermatis and M. restricta [413,414].
A different resistance mechanism affecting azole drugs involves efflux pumps. These overexpressed proteins can actively transport accumulated intracellular antifungal drugs out of fungal cells. Around 30 different proteins have been described either belonging to the ATP-binding cassette (such as CDR1, CDR2 or PDR10) or the major facilitator (such as MDR1) superfamily. Such mechanisms have been detected among isolates of M. pachydermatis, M. furfur and M. restricta (Pdr5) [393,414,415,416]. Mitochondrial dysfunction in M. restricta strains involving ATM1, an iron-sulfur transporter, leading to the activation of the pleiotropic drug resistance (PDR) pathway, resulting in an increased expression of efflux pump transporters, has also been described [414]. Interestingly, by using a Malassezia species broth microdilution chequerboard analysis testing the in vitro efficacy of azoles in combination with drug efflux pump modulators (i.e., haloperidol—HAL, promethazine—PTZ, and cyclosporine), FCZ MIC = 128 µg/mL for M. furfur, FCZ MIC = 64 µg/mL for M. pachydermatis and VOR MIC = 4 µg/mL for both Malassezia species were proposed as cut-off values to discriminate suscep2tible and resistant strains [415].
Finally, biofilm formation can also significantly decrease antifungal sensitivity, as shown in studies of M. pachydermatis [321,394,409].

12. Treatment of Malassezia-Related Diseases

12.1. Treatment of Malassezia-Associated Skin Diseases

12.1.1. Treatment in Animals

For topical therapy, preparations of chlorhexidine alone or in combination with an azole antifungal are mostly used [2,232,374]. For severe Malassezia-associated skin diseases or cases that do not respond to topical therapy alone, oral KZ or ITZ are favored in dogs [2,374,417,418,419,420] and ITZ in cats [2,282,374,421,422]. Due to its high concentration and persistence within the stratum corneum, pulse therapy of ITZ is used with 7 days on, 7 days off, 7 days on, or twice weekly administration [421,423]. Terbinafine [423,424,425,426] and FCZ [427] have been prescribed in single case reports and clinical trials are warranted before treatment recommendations can be made. Even if clinical evidence indicates the efficacy of azole for the control of skin infections, the common recurrence of skin disorders requires the recognition of underlying diseases or the use of prophylaxis systems for the management of these infections in animals [2]. As maintenance therapy, plant-based compounds (i.e., essential oils and phenolic compounds) and peptides have achieved interesting results, but future studies need to be done in order to propose them for clinical use [374].

12.1.2. Treatment in Humans

Pityriasis versicolor—A combination of keratomodulating (sulfur, salicylic acid, selenium sulfide, zinc pyrithione) and antifungal (azoles, ciclopirox olamine, TER) shampoos, sprays or solutions is usually effective, but in widespread, severe, refractory or recurrent cases, systemic antifungal therapy with ITZ or FCZ may be required. Terbinafine is not effective [428,429]. Relapses are common, even after successful initial treatment and long-term management can be challenging [429,430,431] (Figure 3).
Seborrheic dermatitis—Topical treatment with a combination of keratomodulating (pine, tar, salicylic acid, sulfur), antifungal (KZ, ciclopirox, zinc pyrithione) and anti-inflammatory drugs (glucocorticoids, calcineurin inhibitors) together with brushing to remove and soften keratinous material is usually the first choice [219,232,432,433,434,435,436,437]. In severe, widespread and refractory cases, systemic antifungal drugs including ITZ, FCZ, TER and rarely KZ are considered. In addition, it is always important to address the underlying disease if it is present [219,232,432,433,434,435,436,437] (Figure 3).
Malassezia folliculitis—There is some evidence that systemic treatment is the most efficient method, considering the location of the disease within the hair follicles [438]. Itraconazole and FCZ show good efficacy [222,224,439,440,441]. Topical treatment (azoles, selenium sulfide and propylene glycol 50%) may be better used as a preventive measurement or for patients where systemic treatment is contraindicated [439,440,441,442]. Photodynamic therapy as an alternative treatment has also been mentioned [443,444] (Figure 3).
Atopic dermatitis (head and neck dermatitis, HND)—HND patients respond best to systemic antifungal treatment, especially when using ITZ or KZ [243,445,446,447,448,449,450]. Affected individuals are often treated daily for one to two months and then twice weekly for maintenance [448]. Fluconazole can also be used, although some studies report that it would not be as effective as the latter two mentioned drugs [448,451]. Limited data exist for systemic TER [452]. Topical antifungal treatment has not been very promising, although ciclopirox olamine twice daily may be an option for selected cases [453] (Figure 3).
With increased recognition of azole resistance in Malassezia species, there has also been an expansion in the investigation of alternative treatment approaches, including photodynamic therapy, natural products, antifungal peptides and proteinase inhibitors [443,454,455,456,457,458,459].

12.2. Treatment of Systemic Malassezia Infections

For systemic infections in humans, rapid organism identification, together with an aggressive systemic treatment approach, is essential [14,110,196,460]. Intravenous therapy with AmB is effective in infants and adults [14,16,18,38,72,187,188,191,315,347]. FCZ, PSZ and VCZ have been administered, but careful considerations are necessary since failure of the first two drugs are reported, especially due to reported or suspected reduced susceptibility [18,34,187,188,191,404,461,462,463]. Flucytosine or echinocandins have no efficacy against Malassezia and should be avoided [18,185,191,464]. In addition to antifungal therapy, it is of fundamental importance to remove any indwelling devices, such as catheters and to temporarily stop parenteral lipid supplementation [14,18,110,191,196,229,460].

13. Conclusions

Malassezia species are among the most widespread fungi on our planet and it is expected that new species and hosts will be discovered. While some Malassezia species are host adapted, many are shared between animals and humans. There is evidence of zoonotic transmission, especially for M. pachydermatis, but more longitudinal data are needed for further elucidation. Malassezia species can be associated with many different skin diseases in companion, production, avian and exotic animals as well as in humans. In people, Malassezia fungemia and internal infections are increasingly recognized, especially in immunocompromised individuals. In addition, these yeasts are associated with certain chronic diseases, such as Crohn’s disease, but also with some cancers, such as pancreatic ductal adenocarcinoma. Malassezia species need special culture media to grow and international standardization for susceptibility testing is urgently needed. In both human and veterinary medicine, topical treatment is preferred unless the type, severity or refractory state of the disease doesn’t allow it. For systemic Malassezia species infections, AmB is typically used, while for other diseases, azole preparations dominate.

Author Contributions

Conceptualization, S.H. and V.R.B.; methodology, S.H. and V.R.B.; validation, S.H. and V.R.B.; formal analysis, S.H. and V.R.B.; investigation, S.H. and V.R.B.; resources, S.H., V.R.B., V.R., and C.C.; data curation, S.H. and V.R.B.; writing—original draft preparation, S.H. and V.R.B.; writing—review and editing, S.H., V.R.B., V.R., and C.C.; visualization, S.H. and V.R.B.; supervision, V.R.B. and C.C.; project administration, S.H. and V.R.B.; funding acquisition, S.H. All authors have read and agreed to the published version of the manuscript.

Funding

The authors thank the City University of Hong Kong for financial support through the UGC Block Grant.

Institutional Review Board Statement

This review does not require ethical approval.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Wu, G.; Zhao, H.; Li, C.; Rajapakse, M.P.; Wong, W.C.; Xu, J.; Saunders, C.W.; Reeder, N.L.; Reilman, R.A.; Scheynius, A.; et al. Genus-Wide Comparative Genomics of Malassezia Delineates Its Phylogeny, Physiology, and Niche Adaptation on Human Skin. PLoS Genet. 2015, 11, e1005614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Bond, R.; Morris, D.O.; Guillot, J.; Bensignor, E.J.; Robson, D.; Mason, K.V.; Kano, R.; Hill, P.B. Biology, diagnosis and treatment of Malassezia dermatitis in dogs and cats Clinical Consensus Guidelines of the World Association for Veterinary Dermatology. Vet. Dermatol. 2020, 31, 28–74. [Google Scholar] [CrossRef] [PubMed]
  3. Saadatzadeh, M.R. The Immunology of the Mycelial Phase of Malassezia; University of Leeds: Leeds, UK, 1998. [Google Scholar]
  4. Saadatzadeh, M.; Ashbee, H.; Holland, K.; Ingham, E. Production of the mycelial phase of Malassezia in vitro. Sabouraudia 2001, 39, 487–493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Saadatzadeh, M.; Ashbee, H.; Cunliffe, W.; Ingham, E. Cell-mediated immunity to the mycelial phase of Malassezia spp. in patients with pityriasis versicolor and controls. Br. J. Dermatol. 2001, 144, 77–84. [Google Scholar] [CrossRef] [PubMed]
  6. Prohic, A.; Jovovic Sadikovic, T.; Krupalija-Fazlic, M.; Kuskunovic-Vlahovljak, S. Malassezia species in healthy skin and in dermatological conditions. Int. J. Dermatol. 2016, 55, 494–504. [Google Scholar] [CrossRef]
  7. Gioti, A.; Nystedt, B.R.; Li, W.; Xu, J.; Andersson, A.; Averette, A.F.; MŘnch, K.; Wang, X.; Kappauf, C.; Kingsbury, J.M. Genomic insights into the atopic eczema-associated skin commensal yeast Malassezia sympodialis. MBio 2013, 4, e00572-12. [Google Scholar] [CrossRef] [Green Version]
  8. Shifrine, M.; Marr, A. The requirement of fatty acids by Pityrosporum ovale. Microbiology 1963, 32, 263–270. [Google Scholar] [CrossRef] [Green Version]
  9. Brunke, S.; Hube, B. MfLIP1, a gene encoding an extracellular lipase of the lipid-dependent fungus Malassezia furfur. Microbiology 2006, 152, 547–554. [Google Scholar] [CrossRef] [Green Version]
  10. Juntachai, W.; Oura, T.; Murayama, S.Y.; Kajiwara, S. The lipolytic enzymes activities of Malassezia species. Sabouraudia 2009, 47, 477–484. [Google Scholar] [CrossRef] [Green Version]
  11. Puig, L.; Bragulat, M.R.; Castella, G.; Cabanes, F.J. Characterization of the species Malassezia pachydermatis and re-evaluation of its lipid dependence using a synthetic agar medium. PLoS ONE 2017, 12, e0179148. [Google Scholar] [CrossRef]
  12. Vest, B.E.; Krauland, K. Malassezia Furfur. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2020. [Google Scholar]
  13. Saunte, D.M.L.; Gaitanis, G.; Hay, R.J. Malassezia-Associated Skin Diseases, the Use of Diagnostics and Treatment. Front. Cell. Infect. Microbiol. 2020, 10, 112. [Google Scholar] [CrossRef] [PubMed]
  14. Rhimi, W.; Theelen, B.; Boekhout, T.; Otranto, D.; Cafarchia, C. Malassezia spp. Yeasts of Emerging Concern in Fungemia. Front. Cell Infect. Microbiol. 2020, 10, 370. [Google Scholar] [CrossRef] [PubMed]
  15. Miceli, M.H.; Díaz, J.A.; Lee, S.A. Emerging opportunistic yeast infections. Lancet Infect. Dis. 2011, 11, 142–151. [Google Scholar] [CrossRef]
  16. Iatta, R.; Cafarchia, C.; Cuna, T.; Montagna, O.; Laforgia, N.; Gentile, O.; Rizzo, A.; Boekhout, T.; Otranto, D.; Montagna, M.T. Bloodstream infections by Malassezia and Candida species in critical care patients. Med. Mycol. 2014, 52, 264–269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Ilahi, A.; Hadrich, I.; Neji, S.; Trabelsi, H.; Makni, F.; Ayadi, A. Real-Time PCR Identification of Six Malassezia Species. Curr. Microbiol. 2017, 74, 671–677. [Google Scholar] [CrossRef]
  18. Pedrosa, A.F.; Lisboa, C.; Rodrigues, A.G. Malassezia infections with systemic involvement: Figures and facts. J. Dermatol. 2018, 45, 1278–1282. [Google Scholar] [CrossRef]
  19. Salkin, I.; Stone, W.; Gordon, M. Association of Malassezia (Pityrosporum) pachydermatis with sarcoptic mange in New York State. J. Wildl. Dis. 1980, 16, 509–514. [Google Scholar] [CrossRef]
  20. Breuer-Strosberg, R.; Hochleithner, M.; Kuttin, E. Malassezia pachydermatis isolation from a scarlet macaw. Mycoses 1990, 33, 247–250. [Google Scholar] [CrossRef]
  21. Guillot, J.; Petit, T.; Degorce-Rubiales, F.; Guého, E.; Chermette, R. Dermatitis caused by Malassezia pachydermatis in a California sea lion (Zalophus californianus). Vet. Rec. 1998, 142, 311–312. [Google Scholar] [CrossRef]
  22. Nakagaki, K.; Hata, K.; Iwata, E.; Takeo, K. Malassezia pachydermatis isolated from a South American sea lion (Otaria byronia) with dermatitis. J. Vet. Med. Sci. 2000, 62, 901–903. [Google Scholar] [CrossRef] [Green Version]
  23. Begerow, D.; Bauer, R.; Boekhout, T. Phylogenetic placements of ustilaginomycetous anamorphs as deduced from nuclear LSU rDNA sequences. Mycol. Res. 2000, 104, 53–60. [Google Scholar] [CrossRef] [Green Version]
  24. Fell, J.W.; Boekhout, T.; Fonseca, A.; Scorzetti, G.; Statzell-Tallman, A. Biodiversity and systematics of basidiomycetous yeasts as determined by large-subunit rDNA D1/D2 domain sequence analysis. Int. J. Syst. Evol. Microbiol. 2000, 50, 1351–1371. [Google Scholar] [CrossRef] [PubMed]
  25. Hibbett, D.S.; Binder, M.; Bischoff, J.F.; Blackwell, M.; Cannon, P.F.; Eriksson, O.E.; Huhndorf, S.; James, T.; Kirk, P.M.; Lücking, R. A higher-level phylogenetic classification of the Fungi. Mycol. Res. 2007, 111, 509–547. [Google Scholar] [CrossRef] [PubMed]
  26. Wang, Q.-M.; Theelen, B.; Groenewald, M.; Bai, F.-Y.; Boekhout, T. Moniliellomycetes and Malasseziomycetes, two new classes in Ustilaginomycotina. Pers. Mol. Phylogeny Evol. Fungi 2014, 33, 41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Lorch, J.M.; Palmer, J.M.; Vanderwolf, K.J.; Schmidt, K.Z.; Verant, M.L.; Weller, T.J.; Blehert, D.S. Malassezia vespertilionis sp. nov.: A new cold-tolerant species of yeast isolated from bats. Persoonia 2018, 41, 56–70. [Google Scholar] [CrossRef]
  28. Castella, G.; Coutinho, S.D.; Cabanes, F.J. Phylogenetic relationships of Malassezia species based on multilocus sequence analysis. Med. Mycol. 2014, 52, 99–105. [Google Scholar] [PubMed] [Green Version]
  29. Cabanes, F.J.; Theelen, B.; Castella, G.; Boekhout, T. Two new lipid-dependent Malassezia species from domestic animals. FEMS Yeast Res. 2007, 7, 1064–1076. [Google Scholar] [PubMed] [Green Version]
  30. Cabañes, F.; Vega, S.; Castellá, G. Malassezia cuniculi sp. nov. a novel yeast species isolated from rabbit skin. Med. Mycol. 2011, 49, 40–48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Cabanes, F.J.; Coutinho, S.D.; Puig, L.; Bragulat, M.R.; Castella, G. New lipid-dependent Malassezia species from parrots. Rev. Iberoam. Micol. 2016, 33, 92–99. [Google Scholar] [CrossRef]
  32. Park, M.; Cho, Y.J.; Lee, Y.W.; Jung, W.H. Whole genome sequencing analysis of the cutaneous pathogenic yeast Malassezia restricta and identification of the major lipase expressed on the scalp of patients with dandruff. Mycoses 2017, 60, 188–197. [Google Scholar] [CrossRef] [PubMed]
  33. Morand, S.C.; Bertignac, M.; Iltis, A.; Kolder, I.; Pirovano, W.; Jourdain, R.; Clavaud, C. Complete Genome Sequence of Malassezia restricta CBS 7877, an Opportunist Pathogen Involved in Dandruff and Seborrheic Dermatitis. Microbiol. Resour. Announc. 2019, 8, e01543-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Chow, N.A.; Chinn, R.; Pong, A.; Schultz, K.; Kim, J.; Gade, L.; Jackson, B.R.; Beer, K.D.; Litvintseva, A.P. Use of whole-genome sequencing to detect an outbreak of Malassezia pachydermatis infection and colonization in a neonatal intensive care unit—California, 2015–2016. Infect. Control. Hosp. Epidemiol. 2020, 41, 851–853. [Google Scholar] [CrossRef] [PubMed]
  35. D’Andreano, S.; Viñes, J.; Francino, O. Whole-Genome Sequencing and De Novo Assembly of Malassezia pachydermatis Isolated from the Ear Canal of a Dog with Otitis. Microbiol. Resour. Announc. 2021, 10, e00205-21. [Google Scholar] [CrossRef] [PubMed]
  36. Amend, A. From dandruff to deep-sea vents: Malassezia-like fungi are ecologically hyper-diverse. PLoS Pathog. 2014, 10, e1004277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Amend, A.; Burgaud, G.; Cunliffe, M.; Edgcomb, V.P.; Ettinger, C.L.; Gutiérrez, M.; Heitman, J.; Hom, E.F.; Ianiri, G.; Jones, A.C. Fungi in the marine environment: Open questions and unsolved problems. MBio 2019, 10, e01189-18. [Google Scholar] [CrossRef] [Green Version]
  38. Theelen, B.; Cafarchia, C.; Gaitanis, G.; Bassukas, I.D.; Boekhout, T.; Dawson, T.L., Jr. Malassezia ecology, pathophysiology, and treatment. Med. Mycol. 2018, 56 (Suppl. S1), S10–S25. [Google Scholar] [CrossRef] [Green Version]
  39. Arenz, B.E.; Held, B.W.; Jurgens, J.A.; Farrell, R.L.; Blanchette, R.A. Fungal diversity in soils and historic wood from the Ross Sea Region of Antarctica. Soil Biol. Biochem. 2006, 38, 3057–3064. [Google Scholar] [CrossRef]
  40. Fell, J.W.; Scorzetti, G.; Connell, L.; Craig, S. Biodiversity of micro-eukaryotes in Antarctic Dry Valley soils with<5% soil moisture. Soil Biol. Biochem. 2006, 38, 3107–3119. [Google Scholar]
  41. Bass, D.; Howe, A.; Brown, N.; Barton, H.; Demidova, M.; Michelle, H.; Li, L.; Sanders, H.; Watkinson, S.C.; Willcock, S.; et al. Yeast forms dominate fungal diversity in the deep oceans. Proc. Biol. Sci. 2007, 274, 3069–3077. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Lai, X.; Cao, L.; Tan, H.; Fang, S.; Huang, Y.; Zhou, S. Fungal communities from methane hydrate-bearing deep-sea marine sediments in South China Sea. ISME J. 2007, 1, 756–762. [Google Scholar] [CrossRef] [PubMed]
  43. Le Calvez, T.; Burgaud, G.; Mahe, S.; Barbier, G.; Vandenkoornhuyse, P. Fungal diversity in deep-sea hydrothermal ecosystems. Appl. Environ. Microbiol. 2009, 75, 6415–6421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Gao, Z.; Johnson, Z.I.; Wang, G. Molecular characterization of the spatial diversity and novel lineages of mycoplankton in Hawaiian coastal waters. ISME J. 2010, 4, 111–120. [Google Scholar] [CrossRef] [PubMed]
  45. Roy, M.; Watthana, S.; Stier, A.; Richard, F.; Vessabutr, S.; Selosse, M.A. Two mycoheterotrophic orchids from Thailand tropical dipterocarpacean forests associate with a broad diversity of ectomycorrhizal fungi. BMC Biol. 2009, 7, 51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Jebaraj, C.S.; Raghukumar, C.; Behnke, A.; Stoeck, T. Fungal diversity in oxygen-depleted regions of the Arabian Sea revealed by targeted environmental sequencing combined with cultivation. FEMS Microbiol. Ecol. 2010, 71, 399–412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Edgcomb, V.P.; Beaudoin, D.; Gast, R.; Biddle, J.F.; Teske, A. Marine subsurface eukaryotes: The fungal majority. Environ. Microbiol. 2011, 13, 172–183. [Google Scholar] [CrossRef]
  48. Singh, P.; Raghukumar, C.; Verma, P.; Shouche, Y. Fungal community analysis in the deep-sea sediments of the Central Indian Basin by culture-independent approach. Microb. Ecol. 2011, 61, 507–517. [Google Scholar] [CrossRef] [PubMed]
  49. Richards, T.A.; Jones, M.D.; Leonard, G.; Bass, D. Marine fungi: Their ecology and molecular diversity. Ann. Rev. Mar. Sci. 2012, 4, 495–522. [Google Scholar] [CrossRef]
  50. Orsi, W.; Biddle, J.F.; Edgcomb, V. Deep sequencing of subseafloor eukaryotic rRNA reveals active Fungi across marine subsurface provinces. PLoS ONE 2013, 8, e56335. [Google Scholar]
  51. Zhang, T.; Wang, N.F.; Zhang, Y.Q.; Liu, H.Y.; Yu, L.Y. Diversity and distribution of fungal communities in the marine sediments of Kongsfjorden, Svalbard (High Arctic). Sci. Rep. 2015, 5, 14524. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Pang, K.L.; Guo, S.Y.; Chen, I.A.; Burgaud, G.; Luo, Z.H.; Dahms, H.U.; Hwang, J.S.; Lin, Y.L.; Huang, J.S.; Ho, T.W.; et al. Insights into fungal diversity of a shallow-water hydrothermal vent field at Kueishan Island, Taiwan by culture-based and metabarcoding analyses. PLoS ONE 2019, 14, e0226616. [Google Scholar] [CrossRef] [Green Version]
  53. Gao, Z.; Li, B.; Zheng, C.; Wang, G. Molecular detection of fungal communities in the Hawaiian marine sponges Suberites zeteki and Mycale armata. Appl. Environ. Microbiol. 2008, 74, 6091–6101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Amend, A.S.; Barshis, D.J.; Oliver, T.A. Coral-associated marine fungi form novel lineages and heterogeneous assemblages. ISME J. 2012, 6, 1291–1301. [Google Scholar] [CrossRef] [Green Version]
  55. Renker, C.; Alphei, J.; Buscot, F. Soil nematodes associated with the mammal pathogenic fungal genus Malassezia (Basidiomycota: Ustilaginomycetes) in Central European forests. Biol. Fertil. Soils 2003, 37, 70–72. [Google Scholar] [CrossRef]
  56. Tondello, A.; Vendramin, E.; Villani, M.; Baldan, B.; Squartini, A. Fungi associated with the southern Eurasian orchid Spiranthes spiralis (L.) Chevall. Fungal Biol. 2012, 116, 543–549. [Google Scholar] [CrossRef] [PubMed]
  57. Quandt, A.; Glasco, A.; James, T. Intestinal Mycobiome Variation Across Geography and Phylogeny in the Snail Genus Conus. Genetics Society of America. In Proceedings of the 29th Fungal Genetics Conference Asilomar, Asilomar, CA, USA, 14–19 March 2017; Volume 20. [Google Scholar]
  58. Malacrinò, A.; Schena, L.; Campolo, O.; Laudani, F.; Mosca, S.; Giunti, G.; Strano, C.P.; Palmeri, V. A metabarcoding survey on the fungal microbiota associated to the olive fruit fly. Microb. Ecol. 2017, 73, 677–684. [Google Scholar] [CrossRef] [PubMed]
  59. Duarte, E.; Resende, J.; Rosa, C.; Hamdan, J. Prevalence of yeasts and mycelial fungi in bovine parasitic otitis in the State of Minas Gerais, Brazil. J. Vet. Med. Ser. B 2001, 48, 631–635. [Google Scholar] [CrossRef] [PubMed]
  60. Elhady, A.; Gine, A.; Topalovic, O.; Jacquiod, S.; Sorensen, S.J.; Sorribas, F.J.; Heuer, H. Microbiomes associated with infective stages of root-knot and lesion nematodes in soil. PLoS ONE 2017, 12, e0177145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Simmons, R.B.; Gueho, E. A new species of Malassezia. Mycol. Res. 1990, 94, 1146–1149. [Google Scholar] [CrossRef]
  62. Nakabayashi, A.; Sei, Y.; Guillot, J. Identification of Malassezia species isolated from patients with seborrhoeic dermatitis, atopic dermatitis, pityriasis versicolor and normal subjects. Med. Mycol. 2000, 38, 337–341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Bernier, V.; Weill, F.X.; Hirigoyen, V.; Elleau, C.; Feyler, A.; Labrèze, C.; Sarlangue, J.; Chène, G.; Couprie, B.; Taïeb, A. Skin colonization by Malassezia species in neonates: A prospective study and relationship with neonatal cephalic pustulosis. Arch. Dermatol. 2002, 138, 215–218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Sugita, T.; Takashima, M.; Shinoda, T.; Suto, H.; Unno, T.; Tsuboi, R.; Ogawa, H.; Nishikawa, A. New yeast species, Malassezia dermatis, isolated from patients with atopic dermatitis. J. Clin. Microbiol. 2002, 40, 1363–1367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Sugita, T.; Takashima, M.; Kodama, M.; Tsuboi, R.; Nishikawa, A. Description of a new yeast species, Malassezia japonica, and its detection in patients with atopic dermatitis and healthy subjects. J. Clin. Microbiol. 2003, 41, 4695–4699. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Sugita, T.; Tajima, M.; Takashima, M.; Amaya, M.; Saito, M.; Tsuboi, R.; Nishikawa, A. A new yeast, Malassezia yamatoensis, isolated from a patient with seborrheic dermatitis, and its distribution in patients and healthy subjects. Microbiol. Immunol. 2004, 48, 579–583. [Google Scholar] [CrossRef]
  67. Gupta, A.K.; Boekhout, T.; Theelen, B.; Summerbell, R.; Batra, R. Identification and typing of Malassezia species by amplified fragment length polymorphism and sequence analyses of the internal transcribed spacer and large-subunit regions of ribosomal DNA. J. Clin. Microbiol. 2004, 42, 4253–4260. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Batra, R.; Boekhout, T.; Guého, E.; Cabanes, F.J.; Dawson, T.L., Jr.; Gupta, A.K. Malassezia Baillon, emerging clinical yeasts. FEMS Yeast Res. 2005, 5, 1101–1113. [Google Scholar] [CrossRef] [Green Version]
  69. Boekhout, T.; Guého-Kellermann, E.; Mayser, P.; Velegraki, A. Malassezia and the Skin: Science and Clinical Practice; Springer Science & Business Media: Berlin/Heidelberg, Germany, 2010. [Google Scholar]
  70. Gaitanis, G.; Velegraki, A.; Mayser, P.; Bassukas, I.D. Skin diseases associated with Malassezia yeasts: Facts and controversies. Clin. Dermatol. 2013, 31, 455–463. [Google Scholar] [CrossRef]
  71. Cabañes, F. Malassezia Yeasts: How Many Species Infect Humans and Animals? PLoS Pathog. 2014, 10, e1003892. [Google Scholar] [CrossRef] [Green Version]
  72. Aguirre, C.; Euliarte, C.; Finquelievich, J.; de los Ángeles Sosa, M.; Giusiano, G. Fungemia and interstitial lung compromise caused by Malassezia sympodialis in a pediatric patient. Rev. Iberoam. Micol. 2015, 32, 118–121. [Google Scholar] [CrossRef]
  73. Honnavar, P.; Chakrabarti, A.; Dogra, S.; Handa, S.; Rudramurthy, S.M. Phenotypic and molecular characterization of Malassezia japonica isolated from psoriasis vulgaris patients. J. Med. Microbiol. 2015, 64, 232–236. [Google Scholar] [CrossRef] [Green Version]
  74. Honnavar, P.; Prasad, G.S.; Ghosh, A.; Dogra, S.; Handa, S.; Rudramurthy, S.M. Malassezia arunalokei sp. nov. a novel yeast species isolated from seborrheic dermatitis patients and healthy individuals from India. J. Clin. Microbiol. 2016, 54, 1826–1834. [Google Scholar] [CrossRef] [Green Version]
  75. Patron, R.L. A 34-year-old man with cough, lung nodules, fever, and eosinophilia. Clin. Infect. Dis. 2016, 63, 1525–1526. [Google Scholar] [PubMed]
  76. Niemiec, B.A.; Gawor, J.; Tang, S.; Prem, A.; Krumbeck, J.A. The mycobiome of the oral cavity in healthy dogs and dogs with periodontal disease. Am. J. Vet. Res. 2021, 83, 42–49. [Google Scholar] [CrossRef] [PubMed]
  77. Nagata, R.; Nagano, H.; Ogishima, D.; Nakamura, Y.; Hiruma, M.; Sugita, T. Transmission of the major skin microbiota, Malassezia, from mother to neonate. Pediatrics Int. 2012, 54, 350–355. [Google Scholar] [CrossRef] [PubMed]
  78. Grice, E.A.; Segre, J.A. The skin microbiome. Nat. Rev. Microbiol. 2011, 9, 244–253. [Google Scholar] [CrossRef]
  79. Jo, J.-H.; Deming, C.; Kennedy, E.A.; Conlan, S.; Polley, E.C.; Ng, W.-I.; Segre, J.A.; Kong, H.H.; Program, N.C.S. Diverse human skin fungal communities in children converge in adulthood. J. Investig. Dermatol. 2016, 136, 2356–2363. [Google Scholar] [CrossRef] [Green Version]
  80. Ward, T.L.; Dominguez-Bello, M.G.; Heisel, T.; Al-Ghalith, G.; Knights, D.; Gale, C.A. Development of the human mycobiome over the first month of life and across body sites. MSystems 2018, 3, e00140-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Gueho, E.; Midgley, G.; Guillot, J. The genus Malassezia with description of four new species. Antonie Van Leeuwenhoek 1996, 69, 337–355. [Google Scholar] [CrossRef]
  82. Aspiroz, C.; Moreno, L.-A.; Rezusta, A.; Rubio, C. Differentiation of three biotypes of Malassezia species on human normal skin. Correspondence with M. globosa, M. sympodialis and M. restricta. Mycopathologia 1999, 145, 69–74. [Google Scholar] [CrossRef]
  83. Gupta, A.; Kohli, Y.; Summerbell, R.; Faergemann, J. Quantitative culture of Malassezia species from different body sites of individuals with or without dermatoses. Med. Mycol. 2001, 39, 243–251. [Google Scholar] [CrossRef] [Green Version]
  84. Gemmer, C.M.; DeAngelis, Y.M.; Theelen, B.; Boekhout, T.; Dawson, T.L., Jr. Fast, noninvasive method for molecular detection and differentiation of Malassezia yeast species on human skin and application of the method to dandruff microbiology. J. Clin. Microbiol. 2002, 40, 3350–3357. [Google Scholar] [CrossRef] [Green Version]
  85. Gupta, A.K.; Batra, R.; Bluhm, R.; Boekhout, T.; Dawson, T.L., Jr. Skin diseases associated with Malassezia species. J. Am. Acad. Dermatol. 2004, 51, 785–798. [Google Scholar] [CrossRef] [PubMed]
  86. Gaitanis, G.; Velegraki, A.; Alexopoulos, E.; Chasapi, V.; Tsigonia, A.; Katsambas, A. Distribution of Malassezia species in pityriasis versicolor and seborrhoeic dermatitis in Greece. Typing of the major pityriasis versicolor isolate M. globosa. Br. J. Dermatol. 2006, 154, 854–859. [Google Scholar] [CrossRef] [PubMed]
  87. Sugita, T.; Tajima, M.; Tsubuku, H.; Tsuboi, R.; Nishikawa, A. Quantitative analysis of cutaneous Malassezia in atopic dermatitis patients using real-time PCR. Microbiol. Immunol. 2006, 50, 549–552. [Google Scholar] [CrossRef] [PubMed]
  88. Akaza, N.; Akamatsu, H.; Sasaki, Y.; Takeoka, S.; Kishi, M.; Mizutani, H.; Sano, A.; Hirokawa, K.; Nakata, S.; Matsunaga, K. Cutaneous Malassezia microbiota of healthy subjects differ by sex, body part and season. J. Dermatol. 2010, 37, 786–792. [Google Scholar] [CrossRef] [PubMed]
  89. Clavaud, C.; Jourdain, R.; Bar-Hen, A.; Tichit, M.; Bouchier, C.; Pouradier, F.; El Rawadi, C.; Guillot, J.; Ménard-Szczebara, F.; Breton, L. Dandruff is associated with disequilibrium in the proportion of the major bacterial and fungal populations colonizing the scalp. PLoS ONE 2013, 8, e58203. [Google Scholar] [CrossRef]
  90. Tanaka, A.; Cho, O.; Saito, M.; Tsuboi, R.; Kurakado, S.; Sugita, T. Molecular characterization of the skin fungal microbiota in patients with seborrheic dermatitis. J. Clin. Exp. Dermatol. Res. 2014, 5, 239. [Google Scholar]
  91. Leung, M.H.; Chan, K.C.; Lee, P.K. Skin fungal community and its correlation with bacterial community of urban Chinese individuals. Microbiome 2016, 4, 1–15. [Google Scholar] [CrossRef] [Green Version]
  92. Aydogan, K.; Tore, O.; Akcaglar, S.; Oral, B.; Ener, B.; Tunalı, S.; Saricaoglu, H. Effects of Malassezia yeasts on serum Th1 and Th2 cytokines in patients with guttate psoriasis. Int. J. Dermatol. 2013, 52, 46–52. [Google Scholar]
  93. Gomez-Moyano, E.; Crespo-Erchiga, V.; Martínez-Pilar, L.; Diaz, D.G.; Martínez-García, S.; Navarro, M.L.; Casaño, A.V. Do Malassezia species play a role in exacerbation of scalp psoriasis? J. Mycol. Med. 2014, 24, 87–92. [Google Scholar] [CrossRef]
  94. Zhang, H.; Zhang, R.; Ran, Y.; Dai, Y.; Lu, Y.; Wang, P. Genetic polymorphism of Malassezia furfur isolates from Han and Tibetan ethnic groups in China using DNA fingerprinting. Med. Mycol. 2010, 48, 1034–1038. [Google Scholar] [CrossRef] [Green Version]
  95. Leong, C.; Schmid, B.; Toi, M.J.; Wang, J.; Irudayaswamy, A.S.; Goh, J.P.Z.; Bosshard, P.P.; Glatz, M.; Dawson, T.L., Jr. Geographical and ethnic differences influence culturable commensal yeast diversity on healthy skin. Front. Microbiol. 2019, 10, 1891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Grice, E.A.; Dawson, T.L. Host–microbe interactions: Malassezia and human skin. Curr. Opin. Microbiol. 2017, 40, 81–87. [Google Scholar] [CrossRef] [PubMed]
  97. Gaitanis, G.; Velegraki, A.; Alexopoulos, E.C.; Kapsanaki-Gotsi, E.; Zisova, L.; Ran, Y.; Zhang, H.; Arsenis, G.; Bassukas, I.D.; Faergemann, J. Malassezia furfur fingerprints as possible markers for human phylogeography. ISME J. 2009, 3, 498–502. [Google Scholar] [CrossRef] [Green Version]
  98. Giusiano, G.; de los Angeles Sosa, M.; Rojas, F.; Vanacore, S.T.; Mangiaterra, M. Prevalence of Malassezia species in pityriasis versicolor lesions in northeast Argentina. Rev. Iberoam. Micol. 2010, 27, 71–74. [Google Scholar] [CrossRef] [PubMed]
  99. Sugita, T.; Suzuki, M.; Goto, S.; Nishikawa, A.; Hiruma, M.; Yamazaki, T.; Makimura, K. Quantitative analysis of the cutaneous Malassezia microbiota in 770 healthy Japanese by age and gender using a real-time PCR assay. Med. Mycol. 2010, 48, 229–233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Findley, K.; Oh, J.; Yang, J.; Conlan, S.; Deming, C.; Meyer, J.A.; Schoenfeld, D.; Nomicos, E.; Park, M.; Kong, H.H. Topographic diversity of fungal and bacterial communities in human skin. Nature 2013, 498, 367–370. [Google Scholar] [CrossRef]
  101. Oh, J.; Byrd, A.L.; Deming, C.; Conlan, S.; Kong, H.H.; Segre, J.A. Biogeography and individuality shape function in the human skin metagenome. Nature 2014, 514, 59–64. [Google Scholar] [CrossRef] [Green Version]
  102. Prohic, A.; Simic, D.; Sadikovic, T.J.; Krupalija-Fazlic, M. Distribution of Malassezia species on healthy human skin in Bosnia and Herzegovina: Correlation with body part, age and gender. Iran J. Microbiol. 2014, 6, 253–262. [Google Scholar]
  103. Dominguez-Bello, M.G.; Costello, E.K.; Contreras, M.; Magris, M.; Hidalgo, G.; Fierer, N.; Knight, R. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc. Natl. Acad. Sci. USA 2010, 107, 11971–11975. [Google Scholar] [CrossRef] [Green Version]
  104. Oh, K.J.; Lee, S.E.; Jung, H.; Kim, G.; Romero, R.; Yoon, B.H. Detection of ureaplasmas by the polymerase chain reaction in the amniotic fluid of patients with cervical insufficiency. J. Perinat. Med. 2010, 38, 261–268. [Google Scholar] [CrossRef] [Green Version]
  105. Aagaard, K.; Ma, J.; Antony, K.M.; Ganu, R.; Petrosino, J.; Versalovic, J. The placenta harbors a unique microbiome. Sci. Transl. Med. 2014, 6, 237ra65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Dunn, A.B.; Jordan, S.; Baker, B.J.; Carlson, N.S. The maternal infant microbiome: Considerations for labor and birth. MCN. Am. J. Matern. Child Nurs. 2017, 42, 318. [Google Scholar] [CrossRef] [PubMed]
  107. Georgountzou, A.; Papadopoulos, N.G. Postnatal innate immune development: From birth to adulthood. Front. Immunol. 2017, 8, 957. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Bellemain, E.; Carlsen, T.; Brochmann, C.; Coissac, E.; Taberlet, P.; Kauserud, H. ITS as an environmental DNA barcode for fungi: An in silico approach reveals potential PCR biases. BMC Microbiol. 2010, 10, 1–9. [Google Scholar] [CrossRef] [Green Version]
  109. Bazzicalupo, A.L.; Bálint, M.; Schmitt, I. Comparison of ITS1 and ITS2 rDNA in 454 sequencing of hyperdiverse fungal communities. Fungal Ecol. 2013, 6, 102–109. [Google Scholar] [CrossRef]
  110. Gaitanis, G.; Magiatis, P.; Hantschke, M.; Bassukas, I.D.; Velegraki, A. The Malassezia genus in skin and systemic diseases. Clin. Microbiol. Rev. 2012, 25, 106–141. [Google Scholar] [CrossRef] [Green Version]
  111. Dupuy, A.K.; David, M.S.; Li, L.; Heider, T.N.; Peterson, J.D.; Montano, E.A.; Dongari-Bagtzoglou, A.; Diaz, P.I.; Strausbaugh, L.D. Redefining the human oral mycobiome with improved practices in amplicon-based taxonomy: Discovery of Malassezia as a prominent commensal. PLoS ONE 2014, 9, e90899. [Google Scholar] [CrossRef] [Green Version]
  112. Nash, A.K.; Auchtung, T.A.; Wong, M.C.; Smith, D.P.; Gesell, J.R.; Ross, M.C.; Stewart, C.J.; Metcalf, G.A.; Muzny, D.M.; Gibbs, R.A. The gut mycobiome of the Human Microbiome Project healthy cohort. Microbiome 2017, 5, 1–13. [Google Scholar] [CrossRef]
  113. Hoggard, M.; Zoing, M.; Biswas, K.; Taylor, M.W.; Douglas, R.G. The sinonasal mycobiota in chronic rhinosinusitis and control patients. Rhinology 2019, 57, 190–199. [Google Scholar] [CrossRef]
  114. Pisa, D.; Alonso, R.; Carrasco, L. Parkinson’s Disease: A Comprehensive Analysis of Fungi and Bacteria in Brain Tissue. Int. J. Biol. Sci. 2020, 16, 1135. [Google Scholar] [CrossRef]
  115. Abdillah, A.; Ranque, S. Chronic Diseases Associated with Malassezia Yeast. J. Fungi 2021, 7, 855. [Google Scholar] [CrossRef] [PubMed]
  116. Lee, K.; Zhang, I.; Kyman, S.; Kask, O.; Cope, E.K. Co-infection of Malassezia sympodialis with bacterial pathobionts Pseudomonas aeruginosa or Staphylococcus aureus leads to distinct sinonasal inflammatory responses in a murine acute sinusitis model. Front. Cell. Infect. Microbiol. 2020, 10, 472. [Google Scholar] [CrossRef] [PubMed]
  117. Hallen-Adams, H.E.; Suhr, M.J. Fungi in the healthy human gastrointestinal tract. Virulence 2017, 8, 352–358. [Google Scholar] [CrossRef] [PubMed]
  118. Fraczek, M.G.; Chishimba, L.; Niven, R.M.; Bromley, M.; Simpson, A.; Smyth, L.; Denning, D.W.; Bowyer, P. Corticosteroid treatment is associated with increased filamentous fungal burden in allergic fungal disease. J. Allergy Clin. Immunol. 2018, 142, 407–414. [Google Scholar] [CrossRef] [Green Version]
  119. Martinsen, E.M.; Eagan, T.M.; Leiten, E.O.; Haaland, I.; Husebø, G.R.; Knudsen, K.S.; Drengenes, C.; Sanseverino, W.; Paytuví-Gallart, A.; Nielsen, R. The pulmonary mycobiome—A study of subjects with and without chronic obstructive pulmonary disease. PLoS ONE 2021, 16, e0248967. [Google Scholar] [CrossRef]
  120. Prado, M.R.; Brilhante, R.S.; Cordeiro, R.A.; Monteiro, A.J.; Sidrim, J.J.; Rocha, M.F. Frequency of yeasts and dermatophytes from healthy and diseased dogs. J. Vet. Diagn. Investig. 2008, 20, 197–202. [Google Scholar] [CrossRef] [Green Version]
  121. Brito, E.H.; Fontenelle, R.O.; Brilhante, R.S.; Cordeiro, R.A.; Monteiro, A.J.; Sidrim, J.J.; Rocha, M.F. The anatomical distribution and antimicrobial susceptibility of yeast species isolated from healthy dogs. Vet. J. 2009, 182, 320–326. [Google Scholar] [CrossRef]
  122. Meason-Smith, C.; Diesel, A.; Patterson, A.P.; Older, C.E.; Mansell, J.M.; Suchodolski, J.S.; Rodrigues Hoffmann, A. What is living on your dog’s skin? Characterization of the canine cutaneous mycobiota and fungal dysbiosis in canine allergic dermatitis. FEMS Microbiol. Ecol. 2015, 91, fiv139. [Google Scholar] [CrossRef] [Green Version]
  123. Meason-Smith, C.; Diesel, A.; Patterson, A.P.; Older, C.E.; Johnson, T.J.; Mansell, J.M.; Suchodolski, J.S.; Hoffmann, A.R. Characterization of the cutaneous mycobiota in healthy and allergic cats using next generation sequencing. Adv. Vet. Dermatol. 2017, 8, 84–94. [Google Scholar]
  124. Gustafson, B.A. Otitis Externa in the Dog. A Bacteriological and Experimental Study; Gernandts Boktryckeri: Stockholm, Sweden, 1955; p. 117. [Google Scholar]
  125. Dufait, R. Presence of Malassezia pachydermatis (syn. Pityrosporum canis) on the hair and feathers of domestic animals. Bull. Soc. Franc. Mycol. Med. 1985, 14, 19–22. [Google Scholar]
  126. Hajsig, M.; Tadic, V.; Lukman, P. Malassezia pachydermatis in dogs: Significance of its location. Vet Arhiv 1985, 55, 259–266. [Google Scholar]
  127. Bond, R.; Anthony, R.; Dodd, M.; Lloyd, D. Isolation of Malassezia sympodialis from feline skin. J. Med. Vet. Mycol. 1996, 34, 145–147. [Google Scholar] [CrossRef] [PubMed]
  128. Bond, R.; Howell, S.; Haywood, P.; Lloyd, D. Isolation of Malassezia sympodialis and Malassezia globosa from healthy pet cats. Vet. Rec. 1997, 141, 200–201. [Google Scholar] [CrossRef] [PubMed]
  129. Bond, R.; Lloyd, D. Skin and mucosal populations of Malassezia pachydermatis in healthy and seborrhoeic basset hounds. Vet. Dermatol. 1997, 8, 101–106. [Google Scholar] [CrossRef]
  130. Raabe, P.; Mayser, P.; Weiss, R. Demonstration of Malassezia furfur and M. sympodialis together with M. pachydermatis in veterinary specimens: Nachweis von Malassezia furfur und M. sympodialis in veterinärmedizinischem Untersuchungsgut. Mycoses 1998, 41, 493–500. [Google Scholar] [CrossRef]
  131. Crespo, M.; Abarca, M.; Cabanes, F. Isolation of Malassezia furfur from a cat. J. Clin. Microbiol. 1999, 37, 1573–1574. [Google Scholar] [CrossRef] [Green Version]
  132. Crespo, M.; Abarca, M.; Cabanes, F. Atypical lipid-dependent Malassezia species isolated from dogs with otitis externa. J. Clin. Microbiol. 2000, 38, 2383–2385. [Google Scholar] [CrossRef]
  133. Crespo, M.; Abarca, M.; Cabanes, F. Otitis externa associated with Malassezia sympodialis in two cats. J. Clin. Microbiol. 2000, 38, 1263–1266. [Google Scholar] [CrossRef] [Green Version]
  134. Nardoni, S.; Mancianti, F.; Rum, A.; Corazza, M. Isolation of Malassezia species from healthy cats and cats with otitis. J. Feline Med. Surg. 2005, 7, 141–145. [Google Scholar] [CrossRef]
  135. Åhman, S.; Perrins, N.; Bond, R. Carriage of Malassezia spp. yeasts in healthy and seborrhoeic Devon Rex cats. Sabouraudia 2007, 45, 449–455. [Google Scholar] [CrossRef] [Green Version]
  136. Bellis, F.D.; Castellá, G.; Cabañes, F.J.; Bond, R. Absence of DNA sequence diversity of the intergenic spacer 1 region in Malassezia nana isolates from cats. Med. Mycol. 2010, 48, 427–429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Castellá, G.; de Bellis, F.; Bond, R.; Cabañes, F.J. Molecular characterization of Malassezia nana isolates from cats. Vet. Microbiol. 2011, 148, 363–367. [Google Scholar] [CrossRef] [PubMed]
  138. Sihelská, Z.; Čonková, E.; Váczi, P.; Harčárová, M.; Böhmová, E. Occurrence of Malassezia yeasts In dermatologically diseased dogs. Folia Vet. 2017, 61, 17–21. [Google Scholar] [CrossRef] [Green Version]
  139. Older, C.E.; Diesel, A.B.; Lawhon, S.D.; Queiroz, C.R.; Henker, L.C.; Rodrigues Hoffmann, A. The feline cutaneous and oral microbiota are influenced by breed and environment. PLoS ONE 2019, 14, e0220463. [Google Scholar] [CrossRef] [Green Version]
  140. Meason-Smith, C.; Olivry, T.; Lawhon, S.D.; Hoffmann, A.R. Malassezia species dysbiosis in natural and allergen-induced atopic dermatitis in dogs. Med. Mycol. 2020, 58, 756–765. [Google Scholar] [CrossRef]
  141. Guillot, J.; Bond, R. Malassezia Yeasts in Veterinary Dermatology: An Updated Overview. Front. Cell Infect. Microbiol. 2020, 10, 79. [Google Scholar] [CrossRef]
  142. Crespo, M.; Abarca, M.; Cabañes, F.J. Occurrence of Malassezia spp. in the external ear canals of dogs and cats with and without otitis externa. Med. Mycol. 2002, 40, 115–121. [Google Scholar] [CrossRef] [Green Version]
  143. Hajsig, D.; Hajsig, M.; Svoboda Vukovic, D. Malassezia pachydermatis in healthy cats. Vet Arhiv 1990, 60, 69–73. [Google Scholar]
  144. Hirai, A.; Kano, R.; Makimura, K.; Duarte, E.R.; Hamdan, J.S.; Lachance, M.-A.; Yamaguchi, H.; Hasegawa, A. Malassezia nana sp. nov. a novel lipid-dependent yeast species isolated from animals. Int. J. Syst. Evol. Microbiol. 2004, 54, 623–627. [Google Scholar] [CrossRef]
  145. Ahman, S.E.; Bergstrom, K.E. Cutaneous carriage of Malassezia species in healthy and seborrhoeic Sphynx cats and a comparison to carriage in Devon Rex cats. J. Feline Med. Surg. 2009, 11, 970–976. [Google Scholar] [CrossRef]
  146. Volk, A.V.; Belyavin, C.E.; Varjonen, K.; Cadiergues, M.-C.; Stevens, K.B.; Bond, R. Malassezia pachydermatis and M nana predominate amongst the cutaneous mycobiota of Sphynx cats. J. Feline Med. Surg. 2010, 12, 917–922. [Google Scholar] [CrossRef] [PubMed]
  147. Bond, R.; Stevens, K.; Perrins, N.; Åhman, S. Carriage of Malassezia spp. yeasts in Cornish Rex, Devon Rex and Domestic short-haired cats: A cross-sectional survey. Vet. Dermatol. 2008, 19, 299–304. [Google Scholar] [CrossRef]
  148. Korbelik, J.; Singh, A.; Rousseau, J.; Weese, J.S. Analysis of the otic mycobiota in dogs with otitis externa compared to healthy individuals. Vet. Dermatol. 2018, 29, 417-e138. [Google Scholar] [CrossRef] [PubMed]
  149. Crespo, M.; Abarca, M.; Cabanes, F. Occurrence of Malassezia spp. in horses and domestic ruminants. Mycoses 2002, 45, 333–337. [Google Scholar] [CrossRef] [PubMed]
  150. Cafarchia, C.; Gallo, S.; Capelli, G.; Otranto, D. Occurrence and population size of Malassezia spp. in the external ear canal of dogs and cats both healthy and with otitis. Mycopathologia 2005, 160, 143–149. [Google Scholar] [CrossRef]
  151. Dizotti, C.; Coutinho, S. Isolation of Malassezia pachydermatis and M. sympodialis from the external ear canal of cats with and without otitis externa. Acta Vet. Hung. 2007, 55, 471–477. [Google Scholar] [CrossRef] [Green Version]
  152. Shokri, H.; Khosravi, A.; Rad, M.; Jamshidi, S. Occurrence of Malassezia species in Persian and domestic short hair cats with and without otitis externa. J. Vet. Med. Sci. 2010, 72, 293–296. [Google Scholar] [CrossRef] [Green Version]
  153. Eidi, S.; Khosravi, A.R.; Jamshidi, S. A comparison of different kinds of Malassezia species in healthy dogs and dogs with otitis externa and skin lesions. Turk. J. Vet. Anim. Sci. 2011, 35, 345–350. [Google Scholar] [CrossRef]
  154. Bradley, C.W.; Lee, F.F.; Rankin, S.C.; Kalan, L.R.; Horwinski, J.; Morris, D.O.; Grice, E.A.; Cain, C.L. The otic microbiota and mycobiota in a referral population of dogs in eastern USA with otitis externa. Vet. Dermatol. 2020, 31, 225-e49. [Google Scholar] [CrossRef]
  155. Bradley, C.W.; Morris, D.O.; Rankin, S.C.; Cain, C.L.; Misic, A.M.; Houser, T.; Mauldin, E.A.; Grice, E.A. Longitudinal evaluation of the skin microbiome and association with microenvironment and treatment in canine atopic dermatitis. J. Investig. Dermatol. 2016, 136, 1182–1190. [Google Scholar] [CrossRef] [Green Version]
  156. Nardoni, S.; Mancianti, F.; Corazza, M.; Rum, A. Occurrence of Malassezia species in healthy and dermatologically diseased dogs. Mycopathologia 2004, 157, 383–388. [Google Scholar] [CrossRef] [PubMed]
  157. Suchodolski, J.S.; Morris, E.K.; Allenspach, K.; Jergens, A.E.; Harmoinen, J.A.; Westermarck, E.; Steiner, J.M. Prevalence and identification of fungal DNA in the small intestine of healthy dogs and dogs with chronic enteropathies. Vet. Microbiol. 2008, 132, 379–388. [Google Scholar] [CrossRef] [PubMed]
  158. Handl, S.; Dowd, S.E.; Garcia-Mazcorro, J.F.; Steiner, J.M.; Suchodolski, J.S. Massive parallel 16S rRNA gene pyrosequencing reveals highly diverse fecal bacterial and fungal communities in healthy dogs and cats. FEMS Microbiol. Ecol. 2011, 76, 301–310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Swanson, K.S.; Dowd, S.E.; Suchodolski, J.S.; Middelbos, I.S.; Vester, B.M.; Barry, K.A.; Nelson, K.E.; Torralba, M.; Henrissat, B.; Coutinho, P.M. Phylogenetic and gene-centric metagenomics of the canine intestinal microbiome reveals similarities with humans and mice. ISME J. 2011, 5, 639–649. [Google Scholar] [CrossRef] [PubMed]
  160. Foster, M.L.; Dowd, S.E.; Stephenson, C.; Steiner, J.M.; Suchodolski, J.S. Characterization of the fungal microbiome (mycobiome) in fecal samples from dogs. Vet. Med. Int. 2013, 2013, 658373. [Google Scholar] [CrossRef] [Green Version]
  161. Melgarejo, T.; Oakley, B.B.; Krumbeck, J.A.; Tang, S.; Krantz, A.; Linde, A. Assessment of bacterial and fungal populations in urine from clinically healthy dogs using next-generation sequencing. J. Vet. Intern. Med. 2021, 35, 1416–1426. [Google Scholar] [CrossRef]
  162. Garau, M.; del Palacio, A.; Garcia, J. Prevalence of Malassezia spp. in healthy pigs. Mycoses 2005, 48, 17–20. [Google Scholar] [CrossRef]
  163. Kuttin, E.; Glas, I. Mycotic otitis externa in animals. Mycoses 1985, 28, 61–68. [Google Scholar] [CrossRef]
  164. Pinter, L.; Anthony, R.M.; Glumac, N.; Hajsig, D.; Pogacnik, M.; Drobnic-Kosorok, M. Apparent cross-infection with a single strain of Malassezia pachydermatis on a pig farm. Acta Vet. Hung. 2002, 50, 151–156. [Google Scholar] [CrossRef]
  165. Nardoni, S.; Merildi, V.; Frangioni, S.; Ariti, G.; Verin, R.; Vannucci, P.; Mancianti, F. Isolation and characterization of Malassezia spp. in healthy swine of different breeds. Vet. Microbiol. 2010, 141, 155–158. [Google Scholar] [CrossRef]
  166. Colombo, S.; Nardoni, S.; Cornegliani, L.; Mancianti, F. Prevalence of Malassezia spp. yeasts in feline nail folds: A cytological and mycological study. Vet. Dermatol. 2007, 18, 278–283. [Google Scholar] [CrossRef] [PubMed]
  167. Zia, M.; Mirhendi, H.; Toghyani, M. Detection and identification of Malassezia species in domestic animals and aquatic birds by PCR-RFLP. Iran J. Vet. Res. 2015, 16, 36–41. [Google Scholar] [PubMed]
  168. Duarte, E.; Batista, R.; Hahn, R.; Hamdan, J. Factors associated with the prevalence of Malassezia species in the external ears of cattle from the state of Minas Gerais, Brazil. Med. Mycol. 2003, 41, 137–142. [Google Scholar] [CrossRef] [PubMed]
  169. Pin, D. Seborrhoeic dermatitis in a goat due to Malassezia pachydermatis. Vet. Dermatol. 2004, 15, 53–56. [Google Scholar] [CrossRef]
  170. Uzal, F.A.; Paulson, D.; Eigenheer, A.L.; Walker, R.L. Malassezia slooffiae-associated dermatitis in a goat. Vet. Dermatol. 2007, 18, 348–352. [Google Scholar] [CrossRef]
  171. Galuppi, R.; Morandi, B.; Agostini, S.; Dalla Torre, S.; Caffara, M. Survey on the Presence of Malassezia spp. in Healthy Rabbit Ear Canals. Pathogens 2020, 9, 696. [Google Scholar] [CrossRef]
  172. Mendes, J.F.; Albano, A.P.N.; Coimbra, M.A.A.; Ferreira, G.F.D.; Gonçalves, C.L.; Nascente, P.d.S.; Mello, J.R.B.d. Fungi isolated from the excreta of wild birds in screening centers in Pelotas, RS, Brazil. Rev. Do Inst. Med. Trop. São Paulo 2014, 56, 525–528. [Google Scholar] [CrossRef] [Green Version]
  173. Gründer, S.; Mayser, P.; Redmann, T.; Kaleta, E. Mycological examinations on the fungal flora of the chicken comb. Mycoses 2005, 48, 114–119. [Google Scholar] [CrossRef]
  174. Sugita, T.; Tajima, M.; Amaya, M.; Tsuboi, R.; Nishikawa, A. Genotype analysis of Malassezia restricta as the major cutaneous flora in patients with atopic dermatitis and healthy subjects. Microbiol. Immunol. 2004, 48, 755–759. [Google Scholar] [CrossRef]
  175. Kobayashi, T.; Kano, R.; Nagata, M.; Hasegawa, A.; Kamata, H. Genotyping of Malassezia pachydermatis isolates from canine healthy skin and atopic dermatitis by internal spacer 1 (IGS1) region analysis. Vet. Dermatol. 2011, 22, 401–405. [Google Scholar] [CrossRef]
  176. Åkerstedt, J.; Vollset, I. Malassezia pachydermatis with special referenceto canine skin disease. Br. Vet. J. 1996, 152, 269–281. [Google Scholar] [CrossRef]
  177. Guillot, J.; Bond, R. Malassezia pachydermatis: A review. Med. Mycol. 1999, 37, 295–306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Outerbridge, C.A. Mycologic disorders of the skin. Clin. Tech. Small Anim. Pract. 2006, 21, 128–134. [Google Scholar] [CrossRef]
  179. Bajwa, J. Canine Malassezia dermatitis. Can. Vet. J. 2017, 58, 1119–1121. [Google Scholar] [PubMed]
  180. Dawson, T.L., Jr. Malassezia: The forbidden kingdom opens. Cell Host Microbe 2019, 25, 345–347. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Bandhaya, M. The distribution of Malassezia furfur and Malassezia pachydermatis on normal human skin. Southeast Asian J. Trop. Med. Public Health 1993, 24, 343–346. [Google Scholar]
  182. Morris, D.O.; O’Shea, K.; Shofer, F.S.; Rankin, S. Malassezia pachydermatis carriage in dog owners. Emerg. Infect. Dis. 2005, 11, 83. [Google Scholar] [CrossRef]
  183. Welbel, S.F.; McNeil, M.M.; Pramanik, A.; Silberman, R.; Oberle, A.D.; Midgley, G.; Crow, S.; Jarvis, W.R. Nosocomial Malassezia pachydermatis bloodstream infections in a neonatal intensive care unit. Pediatric Infect. Dis. J. 1994, 13, 104–108. [Google Scholar] [CrossRef]
  184. Chang, H.J.; Miller, H.L.; Watkins, N.; Arduino, M.J.; Ashford, D.A.; Midgley, G.; Aguero, S.M.; Pinto-Powell, R.; von Reyn, C.F.; Edwards, W. An epidemic of Malassezia pachydermatis in an intensive care nursery associated with colonization of health care workers’ pet dogs. N. Engl. J. Med. 1998, 338, 706–711. [Google Scholar] [CrossRef]
  185. Chryssanthou, E.; Broberger, U.; Petrini, B. Malassezia pachydermatis fungaemia in a neonatal intensive care unit. Acta Paediatr. 2001, 90, 323–327. [Google Scholar] [CrossRef]
  186. Ayhan, M.; Sancak, B.; Karaduman, A.; Arıkan, S.; Şahin, S. Colonization of neonate skin by Malassezia species: Relationship with neonatal cephalic pustulosis. J. Am. Acad. Dermatol. 2007, 57, 1012–1018. [Google Scholar] [CrossRef] [PubMed]
  187. Al-Sweih, N.; Ahmad, S.; Joseph, L.; Khan, S.; Khan, Z. Malassezia pachydermatis fungemia in a preterm neonate resistant to fluconazole and flucytosine. Med. Mycol. Case Rep. 2014, 5, 9–11. [Google Scholar] [CrossRef] [PubMed]
  188. Choudhury, S.; Marte, R.L. Malassezia pachydermatis fungaemia in an adult on posaconazole prophylaxis for acute myeloid leukaemia. Pathol. J. RCPA 2014, 46, 466–467. [Google Scholar] [CrossRef] [PubMed]
  189. Roman, J.; Bagla, P.; Ren, P.; Blanton, L.S.; Berman, M.A. Malassezia pachydermatis fungemia in an adult with multibacillary leprosy. Med. Mycol. Case Rep. 2016, 12, 1–3. [Google Scholar] [CrossRef] [PubMed]
  190. Lee, J.; Cho, Y.G.; Im Choi, S.; Lee, H.S. First case of catheter-related Malassezia pachydermatis fungemia in an adult. Ann. Lab. Med. 2019, 39, 99–101. [Google Scholar] [CrossRef] [Green Version]
  191. Huang, C.-Y.; Peng, C.-C.; Hsu, C.-H.; Chang, J.-H.; Chiu, N.-C.; Chi, H. Systemic infection caused by Malassezia pachydermatis in infants: Case series and review of the literature. Pediatric Infect. Dis. J. 2020, 39, 444–448. [Google Scholar] [CrossRef]
  192. Cafarchia, C.; Gallo, S.; Romito, D.; Capelli, G.; Chermette, R.; Guillot, J.; Otranto, D. Frequency, body distribution, and population size of Malassezia species in healthy dogs and in dogs with localized cutaneous lesions. J. Vet. Diagn. Investig. 2005, 17, 316–322. [Google Scholar] [CrossRef] [Green Version]
  193. Gueho, E.; Simmons, R.; Pruitt, W.; Meyer, S.; Ahearn, D. Association of Malassezia pachydermatis with systemic infections of humans. J. Clin. Microbiol. 1987, 25, 1789–1790. [Google Scholar] [CrossRef] [Green Version]
  194. Van Belkum, A.; Boekhout, T.; Bosboom, R. Monitoring spread of Malassezia infections in a neonatal intensive care unit by PCR-mediated genetic typing. J. Clin. Microbiol. 1994, 32, 2528–2532. [Google Scholar] [CrossRef] [Green Version]
  195. Iatta, R.; Figueredo, L.A.; Montagna, M.T.; Otranto, D.; Cafarchia, C. In vitro antifungal susceptibility of Malassezia furfur from bloodstream infections. J. Med. Microbiol. 2014, 63, 1467–1473. [Google Scholar] [CrossRef] [Green Version]
  196. Velegraki, A.; Cafarchia, C.; Gaitanis, G.; Iatta, R.; Boekhout, T. Malassezia infections in humans and animals: Pathophysiology, detection, and treatment. PLoS Pathog. 2015, 11, e1004523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Archer-Dubon, C.; Icaza-Chivez, M.E.; Orozco-Topete, R.; Reyes, E.; Baez-Martinez, R.; Ponce de Leon, S. An epidemic outbreak of Malassezia folliculitis in three adult patients in an intensive care unit: A previously unrecognized nosocomial infection. Int. J. Dermatol. 1999, 38, 453–456. [Google Scholar] [CrossRef] [PubMed]
  198. Ilahi, A.; Hadrich, I.; Goudjil, S.; Kongolo, G.; Chazal, C.; Léké, A.; Ayadi, A.; Chouaki, T.; Ranque, S. Molecular epidemiology of a Malassezia pachydermatis neonatal unit outbreak. Med. Mycol. 2018, 56, 69–77. [Google Scholar] [CrossRef] [PubMed]
  199. Prohic, A.; Kasumagic-Halilovic, E. Identification of Malassezia pachydermatis from healthy and diseased human skin. Med. Arh. 2009, 63, 317–319. [Google Scholar]
  200. Fan, Y.-M.; Huang, W.-M.; Li, S.-F.; Wu, G.-F.; Lai, K.; Chen, R.-Y. Granulomatous skin infection caused by Malassezia pachydermatis in a dog owner. Arch. Dermatol. 2006, 142, 1181–1184. [Google Scholar] [CrossRef] [Green Version]
  201. Puig, L.; Bragulat, M.R.; Castella, G.; Cabanes, F.J. Phenotypic and genetic diversity of Malassezia furfur from domestic and zoo animals. Med. Mycol. 2018, 56, 941–949. [Google Scholar] [CrossRef]
  202. Rubenstein, R.M.; Malerich, S.A. Malassezia (pityrosporum) folliculitis. J. Clin. Aesthetic Dermatol. 2014, 7, 37. [Google Scholar]
  203. Tucker, D.; Masood, S. Seborrheic Dermatitis. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2020. [Google Scholar]
  204. Karray, M.; McKinney, W.P. Tinea Versicolor. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2021. [Google Scholar]
  205. Alvarado, Z.; Pereira, C. Fungal diseases in children and adolescents in a referral centre in Bogota, Colombia. Mycoses 2018, 61, 543–548. [Google Scholar] [CrossRef]
  206. De Luca, D.A.; Maianski, Z.; Averbukh, M. A study of skin disease spectrum occurring in Angola phototype V–VI population in Luanda. Int. J. Dermatol. 2018, 57, 849–855. [Google Scholar] [CrossRef]
  207. Diongue, K.; Kébé, O.; Faye, M.; Samb, D.; Diallo, M.; Ndiaye, M.; Seck, M.; Badiane, A.; Ranque, S.; Ndiaye, D. MALDI-TOF MS identification of Malassezia species isolated from patients with pityriasis versicolor at the Seafarers’ Medical Service in Dakar, Senegal. J. Mycol. Médicale 2018, 28, 590–593. [Google Scholar] [CrossRef]
  208. Brandi, N.; Starace, M.; Alessandrini, A.; Piraccini, B.M. Tinea versicolor of the neck as side effect of topical steroids for alopecia areata. J. Dermatol. Treat. 2019, 30, 757–759. [Google Scholar] [CrossRef] [PubMed]
  209. Choi, F.D.; Juhasz, M.L.; Mesinkovska, N.A. Topical ketoconazole: A systematic review of current dermatological applications and future developments. J. Dermatol. Treat. 2019, 30, 760–771. [Google Scholar] [CrossRef] [PubMed]
  210. Errichetti, E.; Stinco, G. Dermoscopy in general dermatology: A practical overview. Dermatol. Ther. 2016, 6, 471–507. [Google Scholar] [CrossRef] [Green Version]
  211. Rosen, T. Mycological Considerations in the Topical Treatment of Superficial Fungal Infections. J. Drugs Dermatol. JDD 2016, 15 (Suppl. 2), s49–s55. [Google Scholar] [PubMed]
  212. Palamaras, I.; Kyriakis, K.; Stavrianeas, N. Seborrheic dermatitis: Lifetime detection rates. J. Eur. Acad. Dermatol. Venereol. 2012, 26, 524–526. [Google Scholar] [CrossRef] [PubMed]
  213. Scognamiglio, P.; Chiaradia, G.; de Carli, G.; Giuliani, M.; Mastroianni, C.M.; Barbacci, S.A.; Buonomini, A.R.; Grisetti, S.; Sampaolesi, A.; Corpolongo, A. The potential impact of routine testing of individuals with HIV indicator diseases in order to prevent late HIV diagnosis. BMC Infect. Dis. 2013, 13, 473. [Google Scholar] [CrossRef] [Green Version]
  214. Sanders, M.; Pardo, L.; Franco, O.; Ginger, R.; Nijsten, T. Prevalence and determinants of seborrhoeic dermatitis in a middle-aged and elderly population: The Rotterdam Study. Br. J. Dermatol. 2018, 178, 148–153. [Google Scholar] [CrossRef]
  215. Lally, A.; Casabonne, D.; Imko-Walczuk, B.; Newton, R.; Wojnarowska, F. Prevalence of benign cutaneous disease among Oxford renal transplant recipients. J. Eur. Acad. Dermatol. Venereol. 2011, 25, 462–470. [Google Scholar] [CrossRef]
  216. Dessinioti, C.; Katsambas, A. Seborrheic dermatitis: Etiology, risk factors, and treatments: Facts and controversies. Clin. Dermatol. 2013, 31, 343–351. [Google Scholar] [CrossRef]
  217. Harada, K.; Saito, M.; Sugita, T.; Tsuboi, R. Malassezia species and their associated skin diseases. J. Dermatol. 2015, 42, 250–257. [Google Scholar] [CrossRef]
  218. Celis, A.; Wösten, H.; Triana, S.; Restrepo, S.; de Cock, H. Malassezia spp. beyond the mycobiota. SM Dermatol. J. 2017, 3, 1–10. [Google Scholar]
  219. Kamamoto, C.; Nishikaku, A.; Gompertz, O.; Melo, A.; Hassun, K.; Bagatin, E. Cutaneous fungal microbiome: Malassezia yeasts in seborrheic dermatitis scalp in a randomized, comparative and therapeutic trial. Derm. Endocrinol. 2017, 9, e1361573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  220. Peyri, J.; Lleonart, M. Clinical and therapeutic profile and quality of life of patients with seborrheic dermatitis. Actas Dermo-Sifiliográficas 2007, 98, 476–482. [Google Scholar] [CrossRef]
  221. Schwartz, J.R.; Messenger, A.G.; Tosti, A.; Todd, G.; Hordinsky, M.; Hay, R.; Wang, X.; Zachariae, C.; Kerr, K.M.; Henry, J.P. A comprehensive pathophysiology of dandruff and seborrheic dermatitis: Towards a more precise definition of scalp health. Acta Derm. Venereol. 2013, 93, 131–137. [Google Scholar] [CrossRef]
  222. Parsad, D.; Saini, R.; Negi, K. Short-term treatment of pityrosporum folliculitis: A double blind placebo-controlled study. J. Eur. Acad. Dermatol. Venereol. 1998, 11, 188–190. [Google Scholar] [CrossRef]
  223. Poli, F. Differential diagnosis of facial acne on black skin. Int. J. Dermatol. 2012, 51, 24–26. [Google Scholar] [CrossRef]
  224. Durdu, M.; Güran, M.; Ilkit, M. Epidemiological characteristics of Malassezia folliculitis and use of the May-Grünwald-Giemsa stain to diagnose the infection. Diagn. Microbiol. Infect. Dis. 2013, 76, 450–457. [Google Scholar] [CrossRef]
  225. Hill, M.K.; Goodfield, M.J.; Rodgers, F.G.; Crowley, J.L.; Saihan, E.M. Skin surface electron microscopy in Pityrosporum folliculitis: The role of follicular occlusion in disease and the response to oral ketoconazole. Arch. Dermatol. 1990, 126, 1071–1074. [Google Scholar] [CrossRef]
  226. Erchiga, V.C.; Florencio, V.D. Malassezia species in skin diseases. Curr. Opin. Infect. Dis. 2002, 15, 133–142. [Google Scholar] [CrossRef]
  227. Akaza, N.; Akamatsu, H.; Sasaki, Y.; Kishi, M.; Mizutani, H.; Sano, A.; Hirokawa, K.; Nakata, S.; Nishijima, S.; Matsunaga, K. Malassezia folliculitis is caused by cutaneous resident Malassezia species. Med. Mycol. 2009, 47, 618–624. [Google Scholar] [CrossRef] [Green Version]
  228. Ko, J.H.; Lee, Y.W.; Choe, Y.B.; Ahn, K.J. Epidemiologic study of Malassezia yeasts in patients with Malassezia folliculitis by 26S rDNA PCR-RFLP analysis. Ann. Dermatol. 2011, 23, 177–184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  229. Tragiannidis, A.; Bisping, G.; Koehler, G.; Groll, A.H. Minireview: Malassezia infections in immunocompromised patients. Mycoses 2010, 53, 187–195. [Google Scholar] [CrossRef] [PubMed]
  230. Alves, E.V.; Martins, J.E.C.; de Ribeiro, E.B.O.; Sotto, M.N. Pityrosporum folliculitis: Renal transplantation case report. J. Dermatol. 2000, 27, 49–51. [Google Scholar] [CrossRef] [PubMed]
  231. Potter, B.S.; Burgoon, C.F.; Johnson, W.C. Pityrosporum folliculitis: Report of seven cases and review of the Pityrosporum organism relative to cutaneous disease. Arch. Dermatol. 1973, 107, 388–391. [Google Scholar] [CrossRef]
  232. Hald, M.; Arendrup, M.C.; Svejgaard, E.L.; Lindskov, R.; Foged, E.K.; Saunte, D.M.L. Evidence-based Danish guidelines for the treatment of Malassezia-related skin diseases. Acta Derm. Venereol. 2015, 95, 12–19. [Google Scholar] [CrossRef] [Green Version]
  233. Bieber, T. Atopic Dermatitis. N. Engl. J. Med. 2008, 358, 1483–1494. [Google Scholar] [CrossRef]
  234. Katsarou, A.; Armenaka, M. Atopic dermatitis in older patients: Particular points. J. Eur. Acad. Dermatol. Venereol. 2011, 25, 12–18. [Google Scholar] [CrossRef]
  235. Baron, S.; Cohen, S.; Archer, C. British Association of Dermatologists and Royal College of General Practitioners. Guidance on the diagnosis and clinical management of atopic eczema. Clin. Exp. Dermatol. 2012, 37 (Suppl. S1), 7–12. [Google Scholar] [CrossRef]
  236. Weidinger, S.; Novak, N. Atopic dermatitis. Lancet 2016, 387, 1109–1122. [Google Scholar] [CrossRef]
  237. Kim, T.; Jang, I.; Park, Y.; Kim, H.; Kim, C. Head and neck dermatitis: The role of Malassezia furfur, topical steroid use and environmental factors in its causation. Clin. Exp. Dermatol. 1999, 24, 226–231. [Google Scholar] [CrossRef]
  238. Brehler, R.; Luger, T. Atopic dermatitis: The role of Pityrosporum ovale. J. Eur. Acad. Dermatol. Venereol. 2001, 15, 5–6. [Google Scholar] [CrossRef] [PubMed]
  239. Savolainen, J.; Lintu, P.; Kosonen, J.; Kortekangas-Savolainen, O.; Viander, M.; Pene, J.; Kalimo, K.; Terho, E.; Bousquet, J. Pityrosporum and Candida specific and non-specific humoral, cellular and cytokine responses in atopic dermatitis patients. Clin. Exp. Allergy 2001, 31, 125–134. [Google Scholar] [PubMed]
  240. Faergemann, J. Atopic dermatitis and fungi. Clin. Microbiol. Rev. 2002, 15, 545–563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  241. Zhang, E.; Tanaka, T.; Tajima, M.; Tsuboi, R.; Kato, H.; Nishikawa, A.; Sugita, T. Anti-Malassezia-specific IgE antibodies production in Japanese patients with head and neck atopic dermatitis: Relationship between the level of specific IgE antibody and the colonization frequency of cutaneous Malassezia species and clinical severity. J. Allergy 2011, 2011, 645670. [Google Scholar] [CrossRef] [Green Version]
  242. Reguiaï, Z. In Atopic dermatitis of the adult: Clinical presentation, complications and comorbidities. Ann. Dermatol. Venereol. 2017, 144, VS15–VS22. [Google Scholar] [CrossRef]
  243. Silvestre Salvador, J.; Romero-Pérez, D.; Encabo-Durán, B. Atopic dermatitis in adults: A diagnostic challenge. J. Investig. Allergol. Clin. Immunol. 2017, 27, 78–88. [Google Scholar] [CrossRef]
  244. Hiragun, T.; Ishii, K.; Hiragun, M.; Suzuki, H.; Kan, T.; Mihara, S.; Yanase, Y.; Bartels, J.; Schröder, J.-M.; Hide, M. Fungal protein MGL_1304 in sweat is an allergen for atopic dermatitis patients. J. Allergy Clin. Immunol. 2013, 132, 608–615.e4. [Google Scholar] [CrossRef]
  245. Maarouf, M.; Saberian, C.; Lio, P.A.; Shi, V.Y. Head-and-neck dermatitis: Diagnostic difficulties and management pearls. Pediatric Dermatol. 2018, 35, 748–753. [Google Scholar] [CrossRef]
  246. Kohsaka, T.; Hiragun, T.; Ishii, K.; Hiragun, M.; Kamegashira, A.; Hide, M. Different hypersensitivities against homologous proteins of MGL_1304 in patients with atopic dermatitis. Allergol. Int. 2018, 67, 103–108. [Google Scholar] [CrossRef]
  247. Sugita, T.; Suto, H.; Unno, T.; Tsuboi, R.; Ogawa, H.; Shinoda, T.; Nishikawa, A. Molecular analysis of Malassezia microflora on the skin of atopic dermatitis patients and healthy subjects. J. Clin. Microbiol. 2001, 39, 3486–3490. [Google Scholar] [CrossRef] [Green Version]
  248. Guglielmo, A.; Sechi, A.; Patrizi, A.; Gurioli, C.; Neri, I. Head and neck dermatitis, a subtype of atopic dermatitis induced by Malassezia spp: Clinical aspects and treatment outcomes in adolescent and adult patients. Pediatric Dermatol. 2021, 38, 109–114. [Google Scholar] [CrossRef] [PubMed]
  249. Moraru, R.; Chermette, R.; Guillot, J. Superficial mycoses in dogs and cats. In Recent Trends in Human and Animal Mycology; Springer: Berlin/Heidelberg, Germany, 2019; pp. 27–45. [Google Scholar]
  250. Guillot, J.; Guého, E.; Lesourd, M.; Midgley, G.; Chévrier, G.; Dupont, B. Identification of Malassezia species. A practical approach. J. Mycol. Med. 1996, 6, 103–110. [Google Scholar]
  251. Guillot, J.; Poujade, A.; Boulouha, L.; Chermette, R. Could Malassezia Dermatitis be Diagnosed in Animals Other than Pet Carnivores. In Advances in Vetinary Dermatology, 4th ed.; Kusochka, K.W., Willemse, T., von Tscharner, C., Eds.; Butterworth Heinemann: Oxford, UK, 2000. [Google Scholar]
  252. Duarte, E.; Melo, M.; Hahn, R.; Hamdan, J. Prevalence of Malassezia spp. in the ears of asymptomatic cattle and cattle with otitis in Brazil. Med. Mycol. 1999, 37, 159–162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  253. Nell, A.; Herrtage, M.; James, S.; Bond, C.; Hunt, B. Identification and distribution of a novel Malassezia species yeast on normal equine skin. Vet. Rec. 2002, 150, 395–398. [Google Scholar] [CrossRef] [PubMed]
  254. White, S.D.; Vandenabeele, S.; Drazenovich, N.; Foley, J.E. Malassezia species isolated from the intermammary and preputial fossa areas of horses. J. Vet. Intern. Med. 2006, 20, 395–398. [Google Scholar] [CrossRef] [PubMed]
  255. Eguchi-Coe, Y.; Valentine, B.A.; Gorman, E.; Villarroel, A. Putative Malassezia dermatitis in six goats. Vet. Dermatol. 2011, 22, 497–501. [Google Scholar] [CrossRef]
  256. Kim, D.Y.; Johnson, P.J.; Senter, D. Severe bilaterally symmetrical alopecia in a horse. Vet. Pathol. 2011, 48, 1216–1220. [Google Scholar] [CrossRef]
  257. Weidman, F. Exfoliative Dermatitis in the Indian Rhinoceros (Rhinoceros unicornis), with Description of a New Species: Pityrosporum pachydermatis; Zoological Society of Philadelphia: Philadelphia, PA, USA, 1925; pp. 36–44. [Google Scholar]
  258. Midgley, G.; Clayton, Y. The yeast flora of birds and mammals in captivity. Antonie Van Leeuwenhoek 1969, 35, E23–E24. [Google Scholar]
  259. Guillot, J.; Gueho, E. The diversity of Malassezia yeasts confirmed by rRNA sequence and nuclear DNA comparisons. Antonie Van Leeuwenhoek 1995, 67, 297–314. [Google Scholar] [CrossRef]
  260. Dinsdale, J.; Rest, J. Yeast infection in ferrets. Vet. Rec. 1995, 137, 647–648. [Google Scholar]
  261. Pier, A.; Cabañes, F.; Chermette, R.; Ferreiro, L.; Guillot, J.; Jensen, H.; Santurio, J. Prominent animal mycoses from various regions of the world. Sabouraudia 2000, 38 (Suppl. S1), 47–58. [Google Scholar] [CrossRef] [Green Version]
  262. Pollock, C.G.; Rohrbach, B.; Ramsay, E.C. Fungal dermatitis in captive pinnipeds. J. Zoo Wildl. Med. 2000, 31, 374–378. [Google Scholar]
  263. Radi, Z.A. Outbreak of sarcoptic mange and malasseziasis in rabbits (Oryctolagus cuniculus). Comp. Med. 2004, 54, 434–437. [Google Scholar]
  264. Tani, K.; Iwanaga, T.; Sonoda, K.; Hayashiya, S.; Hayashiya, M.; Taura, Y. Ivermectin treatment of demodicosis in 56 hamsters. J. Vet. Med. Sci. 2001, 63, 1245–1247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  265. Shaheena, A.S. Combined infection of malasseziosis and demodicosis in golden hamster: A case report. J. Entomol. Zool. Stud. 2020, 8, 947–948. [Google Scholar]
  266. Mason, K.; Evans, A. Dermatitis associated with Malassezia pachydermatis in 11 dogs. J. Am. Anim. Hosp. Assoc. 1991, 27, 1–13. [Google Scholar]
  267. Plant, J.; Rosenkrantz, W.; Griffin, C. Factors associated with and prevalence of high Malassezia pachydermatis numbers on dog skin. J. Am. Vet. Med. Assoc. 1992, 201, 879–882. [Google Scholar]
  268. Bond, R.; Ferguson, E.; Curtis, C.; Craig, J.; Lloyd, D. Factors associated with elevated cutaneous Malassezia pachydermatis populations in dogs with Pruritic skin isease. J. Small Anim. Pract. 1996, 37, 103–107. [Google Scholar] [CrossRef]
  269. Mauldin, E.A.; Scott, D.W.; Miller, W.H.; Smith, C.A. Malassezia dermatitis in the dog: A retrospective histopathological and immunopathological study of 86 cases (1990–1995). Vet. Dermatol. 1997, 8, 191–202. [Google Scholar] [CrossRef]
  270. Bond, R.; Guillot, J.; Cabañes, F.J. Malassezia Yeasts in Animal Disease. In Malassezia and the Skin; Springer: Berlin/Heidelberg, Germany, 2010; pp. 271–299. [Google Scholar]
  271. Seemanthini, R.; Tresamol, P.; Vinodkumar, K.; Suchithra, S.; Sreenesh, S. Malassezial dermatitis in a guinea pig–a case report. Indian J. Vet. Sci. Biotechnol. 2016, 11, 55–56. [Google Scholar]
  272. Bond, R. Malassezia pachydermatis Colonisation and Infection of Canine Skin; Royal Veterinary College (University of London): London, UK, 1996. [Google Scholar]
  273. Morris, D.O. Malassezia dermatitis and otitis. Vet. Clin. N. Am. Small Anim. Pract. 1999, 29, 1303–1310. [Google Scholar] [CrossRef]
  274. Matousek, J.L.; Campbell, K.L. Malassezia dermatitis. Compend. Contin. Educ. Pract. Vet. 2002, 24, 224–232. [Google Scholar]
  275. CHEN, T.; Hill, P.B. The biology of Malassezia organisms and their ability to induce immune responses and skin disease. Vet. Dermatol. 2005, 16, 4–26. [Google Scholar] [CrossRef] [PubMed]
  276. Nardoni, S.; Dini, M.; Taccini, F.; Mancianti, F. Occurrence, distribution and population size of Malassezia pachydermatis on skin and mucosae of atopic dogs. Vet. Microbiol. 2007, 122, 172–177. [Google Scholar] [CrossRef]
  277. Nuttall, T.J.; Halliwell, R.E. Serum antibodies to Malassezia yeasts in canine atopic dermatitis. Vet. Dermatol. 2001, 12, 327–332. [Google Scholar] [CrossRef] [Green Version]
  278. Morris, D.; Olivier, N.; Rosser, E. Type-1 hypersensitivity reactions to Malassezia pachydermatis extracts in atopic dogs. Am. J. Vet. Res. 1998, 59, 836–841. [Google Scholar]
  279. Morris, D.O.; DeBoer, D.J. Evaluation of serum obtained from atopic dogs with dermatitis attributable to Malassezia pachydermatis for passive transfer of immediate hypersensitivity to that organism. Am. J. Vet. Res. 2003, 64, 262–266. [Google Scholar] [CrossRef]
  280. Chen, T.A.; Halliwell, R.E.; Pemberton, A.D.; Hill, P.B. Identification of major allergens of Malassezia pachydermatis in dogs with atopic dermatitis and Malassezia overgrowth. Vet. Dermatol. 2002, 13, 141–150. [Google Scholar] [CrossRef]
  281. Bensignor, E. Malassezia dermatitis in cats: 15 cases treated with itraconazole. Vet. Rec. 2010, 167, 1011–1012. [Google Scholar] [CrossRef]
  282. Ordeix, L.; Galeotti, F.; Scarampella, F.; Dedola, C.; Bardagi, M.; Romano, E.; Fondati, A. Malassezia spp. overgrowth in allergic cats. Vet. Dermatol. 2007, 18, 316–323. [Google Scholar] [CrossRef]
  283. Bond, R.; Curtis, C.; Ferguson, E.; Mason, I.; Rest, J. An idiopathic facial dermatitis of Persian cats. Vet. Dermatol. 2000, 11, 35–41. [Google Scholar] [CrossRef] [PubMed]
  284. Fontaine, J.; Heimann, M. P-70 Idiopathic facial dermatitis of the Persian cat: Three cases controlled with cyclosporine. Vet. Dermatol. 2004, 15, 64. [Google Scholar] [CrossRef]
  285. Chung, T.H.; Ryu, M.H.; Kim, D.Y.; Yoon, H.Y.; Hwang, C.Y. Topical tacrolimus (FK506) for the treatment of feline idiopathic facial dermatitis. Aust. Vet. J. 2009, 87, 417–420. [Google Scholar] [CrossRef] [PubMed]
  286. White, S.D.; Bordeau, P.B.; Blumstein, P.; Ibisch, C.; GuaguÈre, E.; Denerolle, P.; Carlotti, D.N.; Scott, K.V. Feline acne and results of treatment with mupirocin in an open clinical trial: 25 cases (1994–1996). Vet. Dermatol. 1997, 8, 157–164. [Google Scholar] [CrossRef] [PubMed]
  287. Jazic, E.; Coyner, K.; Loeffler, D.; Lewis, T. An evaluation of the clinical, cytological, infectious and histopathological features of feline acne. Vet. Dermatol. 2006, 17, 134–140. [Google Scholar] [CrossRef]
  288. Pascal-Tenorio, A.; Olivry, T.; Gross, T.L.; Atlee, B.A.; Ihrke, P.J. Paraneoplastic alopecia associated with internal malignancies in the cat. Vet. Dermatol. 1997, 8, 47–52. [Google Scholar] [CrossRef]
  289. Godfrey, D. A case of feline paraneoplastic alopecia with secondary Malassezia-associated dermatitis. J. Small Anim. Pract. 1998, 39, 394–396. [Google Scholar] [CrossRef]
  290. Tasker, S.; Griffon, D.; Nuttall, T.; Hill, P. Resolution of paraneoplastic alopecia following surgical removal of a pancreatic carcinoma in a cat. J. Small Anim. Pract. 1999, 40, 16–19. [Google Scholar] [CrossRef]
  291. Roccabianca, P.; Rondena, M.; Paltrinieri, S.; Pocacqua, V.; Scarpa, P.; Faverzani, S.; Scanziani, E.; Caniatti, M. Multiple endocrine neoplasia type-I-like syndrome in two cats. Vet. Pathol. 2006, 43, 345–352. [Google Scholar] [CrossRef] [Green Version]
  292. Marconato, L.; Albanese, F.; Viacava, P.; Marchetti, V.; Abramo, F. Paraneoplastic alopecia associated with hepatocellular carcinoma in a cat. Vet. Dermatol. 2007, 18, 267–271. [Google Scholar] [CrossRef]
  293. Grandt, L.-M.; Roethig, A.; Schroeder, S.; Koehler, K.; Langenstein, J.; Thom, N.; Neiger, R. Feline paraneoplastic alopecia associated with metastasising intestinal carcinoma. J. Feline Med. Surg. Open Rep. 2015, 1, 2055116915621582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  294. Caporali, C.; Albanese, F.; Binanti, D.; Abramo, F. Two cases of feline paraneoplastic alopecia associated with a neuroendocrine pancreatic neoplasia and a hepatosplenic plasma cell tumour. Vet. Dermatol. 2016, 27, 508-e137. [Google Scholar] [CrossRef] [PubMed]
  295. HLJJFTE, M.F.V.; Curtis, C.; White, R. Resolution of exfoliative dermatitis and Malassezia pachydermatis overgrowth in a cat after surgical thymoma resection. J. Small Anim. Pract. 1997, 38, 451–454. [Google Scholar] [CrossRef] [PubMed]
  296. Rottenberg, S.; Von Tscharner, C.; Roosje, P. Thymoma-associated exfoliative dermatitis in cats. Vet. Pathol. 2004, 41, 429–433. [Google Scholar] [CrossRef] [PubMed]
  297. Reche, A., Jr.; Daniel, A.G.; Strauss, T.C.L.; Taborda, C.P.; Vieira Marques, S.A.; Haipek, K.; Oliveira, L.J.; Monteiro, J.M.; Kfoury, J.R., Jr. Cutaneous mycoflora and CD4: CD8 ratio of cats infected with feline immunodeficiency virus. J. Feline Med. Surg. 2010, 12, 355–358. [Google Scholar] [CrossRef]
  298. Sierra, P.; Guillot, J.; Jacob, H.; Bussiéras, S.; Chermette, R. Fungal flora on cutaneous and mucosal surfaces of cats infected with feline immunodeficiency virus or feline leukemia virus. Am. J. Vet. Res. 2000, 61, 158–161. [Google Scholar] [CrossRef]
  299. Mauldin, E.A.; Morris, D.O.; Goldschmidt, M.H. Retrospective study: The presence of Malassezia in feline skin biopsies. A clinicopathological study. Vet. Dermatol. 2002, 13, 7–14. [Google Scholar] [CrossRef]
  300. Han, J.-I.; Na, K.-J. Otitis externa caused by Malassezia furfur in a miniature pig. J. Vet. Clin. 2009, 26, 303–305. [Google Scholar]
  301. Nunes Rodrigues, T.C.; Vandenabeele, S.I. Pilot study of dogs with suppurative and non-suppurative Malassezia otitis: A case series. BMC Vet. Res. 2021, 17, 353. [Google Scholar] [CrossRef]
  302. Shiota, R.; Kaneko, T.; Yano, H.; Takeshita, K.; Nishioka, K.; Makimura, K. A Study of Otitis Externa Associated with Malassezia. Jpn. J. Med. Mycol. 2009, 50, 109–116. [Google Scholar] [CrossRef] [Green Version]
  303. Latha, R.; Sasikala, R.; Muruganandam, N. Chronic otomycosis due to Malassezia spp. J. Glob. Infect. Dis. 2010, 2, 189. [Google Scholar] [CrossRef] [PubMed]
  304. Crosier, W.J.; Wise, K.A. Onychomycosis due to Pityrosporum. Australas. J. Dermatol. 1993, 34, 109–112. [Google Scholar] [CrossRef] [PubMed]
  305. Silva, V.; Moreno, G.A.; Zaror, L.; de-Oliveira, E.; Fischman, O. Isolation of Malassezia furfur from patients with onychomycosis. J. Med. Vet. Mycol. 1997, 35, 73–74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  306. Chowdhary, A.; Randhawa, H.; Sharma, S.; Brandt, M.E.; Kumar, S. Malassezia furfur in a case of onychomycosis: Colonizer or etiologic agent? Med. Mycol. 2005, 43, 87–90. [Google Scholar] [CrossRef] [Green Version]
  307. Roodhooft, J.; van Rens, G.; Bogaerts, M.; Vermander, J. Infectious crystalline keratopathy: A case report. Bull. Société Belg. D’Ophtalmologie 1998, 268, 121–126. [Google Scholar]
  308. Suzuki, T.; Hori, N.; Miyake, T.; Hori, Y.; Mochizuki, K. Keratitis caused by a rare fungus, Malassezia restricta. Jpn. J. Ophthalmol. 2007, 51, 292–294. [Google Scholar] [CrossRef]
  309. Ledbetter, E.C.; Starr, J.K. Malassezia pachydermatis keratomycosis in a dog. Med. Mycol. Case Rep. 2015, 10, 24–26. [Google Scholar] [CrossRef]
  310. Prado, M.; Brito, E.; Girão, M.; Monteiro, A.; Sidrim, J.; Rocha, M. Higher incidence of Malassezia pachydermatis in the eyes of dogs with corneal ulcer than in healthy dogs. Vet. Microbiol. 2004, 100, 115–120. [Google Scholar] [CrossRef]
  311. Theelen, B.; Silvestri, M.; Guého, E.; van Belkum, A.; Boekhout, T. Identification and typing of Malassezia yeasts using amplified fragment length polymorphism (AFLPTm), random amplified polymorphic DNA (RAPD) and denaturing gradient gel electrophoresis (DGGE). FEMS Yeast Res. 2001, 1, 79–86. [Google Scholar] [CrossRef]
  312. Kaneko, T.; Murotani, M.; Ohkusu, K.; Sugita, T.; Makimura, K. Genetic and biological features of catheter-associated Malassezia furfur from hospitalized adults. Med. Mycol. 2012, 50, 74–80. [Google Scholar] [CrossRef]
  313. Iatta, R.; Battista, M.; Miragliotta, G.; Boekhout, T.; Otranto, D.; Cafarchia, C. Blood culture procedures and diagnosis of Malassezia furfur bloodstream infections: Strength and weakness. Med. Mycol. 2018, 56, 828–833. [Google Scholar] [CrossRef] [Green Version]
  314. Wallace, M.; Bagnall, H.; Glen, D.; Averill, S. Isolation of lipophilic yeast in “sterile” peritonitis. Lancet 1979, 2, 956. [Google Scholar] [CrossRef]
  315. Oliveri, S.; Trovato, L.; Betta, P.; Romeo, M.; Nicoletti, G. Malassezia furfur fungaemia in a neonatal patient detected by lysis-centrifugation blood culture method: First case reported in Italy. Mycoses 2011, 54, e638–e640. [Google Scholar] [CrossRef] [PubMed]
  316. Marcon, M.J.; Powell, D.A. Human infections due to Malassezia spp. Clin. Microbiol. Rev. 1992, 5, 101–119. [Google Scholar] [CrossRef] [PubMed]
  317. Ashbee, H.R.; Evans, E.G.V. Immunology of diseases associated with Malassezia species. Clin. Microbiol. Rev. 2002, 15, 21–57. [Google Scholar] [CrossRef] [Green Version]
  318. Cafarchia, C.; Otranto, D. Association between phospholipase production by Malassezia pachydermatis and skin lesions. J. Clin. Microbiol. 2004, 42, 4868–4869. [Google Scholar] [CrossRef] [Green Version]
  319. Cafarchia, C.; Dell’Aquila, M.; Traversa, D.; Albrizio, M.; Guaricci, A.; de Santis, T.; Otranto, D. Expression of the μ-opioid receptor on Malassezia pachydermatis and its effect in modulating phospholipase production. Med. Mycol. 2010, 48, 73–78. [Google Scholar] [CrossRef] [Green Version]
  320. Gaitanis, G.; Velegraki, A.; Magiatis, P.; Pappas, P.; Bassukas, I. Could Malassezia yeasts be implicated in skin carcinogenesis through the production of aryl-hydrocarbon receptor ligands? Med. Hypotheses 2011, 77, 47–51. [Google Scholar] [CrossRef]
  321. Figueredo, L.A.; Cafarchia, C.; Otranto, D. Antifungal susceptibility of Malassezia pachydermatis biofilm. Med. Mycol. 2013, 51, 863–867. [Google Scholar] [CrossRef] [Green Version]
  322. Vlachos, C.; Gaitanis, G.; Alexopoulos, E.; Papadopoulou, C.; Bassukas, I. Phospholipase activity after β-endorphin exposure discriminates Malassezia strains isolated from healthy and seborrhoeic dermatitis skin. J. Eur. Acad. Dermatol. Venereol. 2013, 27, 1575–1578. [Google Scholar] [CrossRef]
  323. Angiolella, L.; Carradori, S.; Maccallini, C.; Giusiano, G.; Supuran, C.T. Targeting Malassezia species for novel synthetic and natural antidandruff agents. Curr. Med. Chem. 2017, 24, 2392–2412. [Google Scholar] [CrossRef] [PubMed]
  324. Johansson, H.J.; Vallhov, H.; Holm, T.; Gehrmann, U.; Andersson, A.; Johansson, C.; Blom, H.; Carroni, M.; Lehtiö, J.; Scheynius, A. Extracellular nanovesicles released from the commensal yeast Malassezia sympodialis are enriched in allergens and interact with cells in human skin. Sci. Rep. 2018, 8, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  325. Zhang, Y.-J.; Han, Y.; Sun, Y.-Z.; Jiang, H.-H.; Liu, M.; Qi, R.-Q.; Gao, X.-H. Extracellular vesicles derived from Malassezia furfur stimulate IL-6 production in keratinocytes as demonstrated in in vitro and in vivo models. J. Dermatol. Sci. 2019, 93, 168–175. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  326. Pedrosa, A.F.; Lisboa, C.; Faria-Ramos, I.; Silva, R.; Ricardo, E.; Teixeira-Santos, R.; Miranda, I.; Rodrigues, A.G. Epidemiology and susceptibility profile to classic antifungals and over-the-counter products of Malassezia clinical isolates from a Portuguese University Hospital: A prospective study. J. Med. Microbiol. 2019, 68, 778–784. [Google Scholar] [CrossRef]
  327. Findley, K.; Grice, E.A. The skin microbiome: A focus on pathogens and their association with skin disease. PLoS Pathog. 2014, 10, e1004436. [Google Scholar] [CrossRef]
  328. Thayikkannu, A.B.; Kindo, A.J.; Veeraraghavan, M. Malassezia—Can it be ignored? Indian J. Dermatol. 2015, 60, 332. [Google Scholar]
  329. Marcon, M.J.; Powell, D.A. Epidemiology, diagnosis, and management of Malassezia furfur systemic infection. Diagn. Microbiol. Infect. Dis. 1987, 7, 161–175. [Google Scholar] [CrossRef]
  330. Powell, D.A.; Marcon, M.J.; Durrell, D.E.; Pfister, R.M. Scanning electron microscopy of Malassezia furfur attachment to Broviac catheters. Hum. Pathol. 1987, 18, 740–745. [Google Scholar] [CrossRef]
  331. Cannizzo, F.T.; Eraso, E.; Ezkurra, P.A.; Villar-Vidal, M.; Bollo, E.; Castellá, G.; Javier Cabañes, F.; Vidotto, V.; Quindós, G. Biofilm development by clinical isolates of Malassezia pachydermatis. Med. Mycol. 2007, 45, 357–361. [Google Scholar] [CrossRef] [Green Version]
  332. Aoba, S.; Komiyama, A.; Hasegawa, O. Fungal meningitis caused by a Malassezia species masquerading as painful ophthalmoplegia. Rinsho Shinkeigaku Clin. Neurol. 1993, 33, 462–464. [Google Scholar]
  333. Rosales, C.M.; Jackson, M.A.; Zwick, D. Malassezia furfur meningitis associated with total parenteral nutrition subdural effusion. Pediatric Dev. Pathol. 2004, 7, 86–90. [Google Scholar] [CrossRef] [PubMed]
  334. Schleman, K.A.; Tullis, G.; Blum, R. Intracardiac mass complicating Malassezia furfur fungemia. Chest 2000, 118, 1828–1829. [Google Scholar] [CrossRef] [PubMed]
  335. Shparago, C.N.I.; Bruno, L.P.P. Systemic Malassezia furfur infection in an adult receiving total parenteral nutrition. J. Osteopath. Med. 1995, 95, 375. [Google Scholar] [CrossRef] [Green Version]
  336. Chu, C.; Lai, R. Malassezia furfur fungaemia in a ventilator-dependent patient without known risk factors. Hong Kong Med. J. 2002, 8, 212–215. [Google Scholar]
  337. Fine, A.; Churchill, D.; Gault, H.; Fardy, P. Pityrosporum Pachydermatis Peritonitis in a CAPD Patient on Longterm Intraperitoneal Antibiotics. Perit. Dial. Int. 1983, 3, 108. [Google Scholar] [CrossRef]
  338. Johnson, A.; Bailey, E.; Wright, P.; Solomon, L. Malassezia furfur: A possible cause of culture-negative CAPD peritonitis. Perit. Dial. Int. 1996, 16, 187. [Google Scholar] [CrossRef]
  339. Wurtz, R.M.; Knospe, W.N. Malassezia furfur fungemia in a patient without the usual risk factors. Ann. Intern. Med. 1988, 109, 432–433. [Google Scholar] [CrossRef]
  340. Oberle, A.D.; Fowler, M.; Grafton, W.D. Pityrosporum isolate from the upper respiratory tract. Am. J. Clin. Pathol. 1981, 76, 112–116. [Google Scholar] [CrossRef] [Green Version]
  341. Bertini, B.; Kuttin, E.; Beemer, A. Cytopathology of nipple discharge due to Pityrosporum orbiculare and cocci in an elderly woman. Acta Cytol. 1975, 19, 38–42. [Google Scholar]
  342. Alpert, G.; Bell, L.M.; Campos, J.M. Malassezia furfur fungemia in infancy. Clin. Pediatrics 1987, 26, 528–531. [Google Scholar] [CrossRef]
  343. Shek, Y.H.; Tucker, M.C.; Viciana, A.L.; Manz, H.J.; Connor, D.H. Malassezia furfur—disseminated infection in premature infants. Am. J. Clin. Pathol. 1989, 92, 595–603. [Google Scholar] [CrossRef] [PubMed]
  344. Devlin, R.K. Invasive fungal infections caused by Candida and Malassezia species in the neonatal intensive care unit. Adv. Neonatal Care 2006, 6, 68–77. [Google Scholar] [CrossRef] [PubMed]
  345. Zomorodain, K.; Mirhendi, H.; Tarazooie, B.; Kordbacheh, P.; Zeraati, H.; Nayeri, F. Molecular analysis of Malassezia species isolated from hospitalized neonates. Pediatric Dermatol. 2008, 25, 312–316. [Google Scholar] [CrossRef] [PubMed]
  346. Dankner, W.M.; Spector, S.A.; Fierer, J.; Davis, C.E. Malassezia fungemia in neonates and adults: Complication of hyperalimentation. Rev. Infect. Dis. 1987, 9, 743–753. [Google Scholar] [CrossRef]
  347. Barber, G.R.; Brown, A.E.; Kiehn, T.E.; Edwards, F.F.; Armstrong, D. Catheter-related Malassezia furfur fungemia in immunocompromised patients. Am. J. Med. 1993, 95, 365–370. [Google Scholar] [CrossRef]
  348. Morrison, V.; Weisdorf, D. The spectrum of Malassezia infections in the bone marrow transplant population. Bone Marrow Transplant. 2000, 26, 645–648. [Google Scholar] [CrossRef] [Green Version]
  349. Campigotto, A.; Richardson, S.E.; Sebert, M.; McElvania TeKippe, E.; Chakravarty, A.; Doern, C.D. Low utility of pediatric isolator blood culture system for detection of fungemia in children: A 10-year review. J. Clin. Microbiol. 2016, 54, 2284–2287. [Google Scholar] [CrossRef] [Green Version]
  350. Nelson, S.C.; Yau, Y.; Richardson, S.E.; Matlow, A.G. Improved detection of Malassezia species in lipid-supplemented Peds Plus blood culture bottles. J. Clin. Microbiol. 1995, 33, 1005–1007. [Google Scholar] [CrossRef] [Green Version]
  351. Schechtman, R.; Midgley, G.; Hay, R. HIV disease and Malassezia yeasts: A quantitative study of patients presenting with seborrhoeic dermatitis. Br. J. Dermatol. 1995, 133, 694–698. [Google Scholar] [CrossRef]
  352. Moreno-Coutiño, G.; Sánchez-Cárdenas, C.D.; Bello-Hernández, Y.; Fernández-Martínez, R.; Arroyo-Escalante, S.; Arenas, R. Isolation of Malassezia spp. in HIV-positive patients with and without seborrheic dermatitis. An. Bras. Dermatol. 2019, 94, 527–531. [Google Scholar] [CrossRef]
  353. Krzyściak, P.; Bakuła, Z.; Gniadek, A.; Garlicki, A.; Tarnowski, M.; Wichowski, M.; Jagielski, T. Prevalence of Malassezia species on the skin of HIV-seropositive patients. Sci. Rep. 2020, 10, 1–13. [Google Scholar] [CrossRef] [PubMed]
  354. Liguori, G.; Lamas, B.; Richard, M.L.; Brandi, G.; Da Costa, G.; Hoffmann, T.W.; Di Simone, M.P.; Calabrese, C.; Poggioli, G.; Langella, P. Fungal dysbiosis in mucosa-associated microbiota of Crohn’s disease patients. J. Crohn’s Colitis 2016, 10, 296–305. [Google Scholar] [CrossRef] [PubMed]
  355. Sokol, H.; Leducq, V.; Aschard, H.; Pham, H.-P.; Jegou, S.; Landman, C.; Cohen, D.; Liguori, G.; Bourrier, A.; Nion-Larmurier, I. Fungal microbiota dysbiosis in IBD. Gut 2017, 66, 1039–1048. [Google Scholar] [CrossRef] [Green Version]
  356. Limon, J.J.; Tang, J.; Li, D.; Wolf, A.J.; Michelsen, K.S.; Funari, V.; Gargus, M.; Nguyen, C.; Sharma, P.; Maymi, V.I. Malassezia is associated with Crohn’s disease and exacerbates colitis in mouse models. Cell Host Microbe 2019, 25, 377–388.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  357. Jawhara, S.; Thuru, X.; Standaert-Vitse, A.; Jouault, T.; Mordon, S.; Sendid, B.; Desreumaux, P.; Poulain, D. Colonization of mice by Candida albicans is promoted by chemically induced colitis and augments inflammatory responses through galectin-3. J. Infect. Dis. 2008, 197, 972–980. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  358. Chiaro, T.R.; Soto, R.; Zac Stephens, W.; Kubinak, J.L.; Petersen, C.; Gogokhia, L.; Bell, R.; Delgado, J.C.; Cox, J.; Voth, W. A member of the gut mycobiota modulates host purine metabolism exacerbating colitis in mice. Sci. Transl. Med. 2017, 9, eaaf9044. [Google Scholar] [CrossRef]
  359. Drummond, R.A.; Franco, L.M.; Lionakis, M.S. Human CARD9: A critical molecule of fungal immune surveillance. Front. Immunol. 2018, 9, 1836. [Google Scholar] [CrossRef] [Green Version]
  360. Gao, R.; Kong, C.; Li, H.; Huang, L.; Qu, X.; Qin, N.; Qin, H. Dysbiosis signature of mycobiota in colon polyp and colorectal cancer. Eur. J. Clin. Microbiol. Infect. Dis. 2017, 36, 2457–2468. [Google Scholar] [CrossRef]
  361. Aykut, B.; Pushalkar, S.; Chen, R.; Li, Q.; Abengozar, R.; Kim, J.I.; Shadaloey, S.A.; Wu, D.; Preiss, P.; Verma, N. The fungal mycobiome promotes pancreatic oncogenesis via activation of MBL. Nature 2019, 574, 264–267. [Google Scholar] [CrossRef]
  362. Coker, O.O.; Nakatsu, G.; Dai, R.Z.; Wu, W.K.K.; Wong, S.H.; Ng, S.C.; Chan, F.K.L.; Sung, J.J.Y.; Yu, J. Enteric fungal microbiota dysbiosis and ecological alterations in colorectal cancer. Gut 2019, 68, 654–662. [Google Scholar] [CrossRef]
  363. Alonso, R.; Pisa, D.; Fernández-Fernández, A.M.; Rábano, A.; Carrasco, L. Fungal infection in neural tissue of patients with amyotrophic lateral sclerosis. Neurobiol. Dis. 2017, 108, 249–260. [Google Scholar] [CrossRef] [PubMed]
  364. Alonso, R.; Pisa, D.; Aguado, B.; Carrasco, L. Identification of fungal species in brain tissue from Alzheimer’s disease by next-generation sequencing. J. Alzheimer’s Dis. 2017, 58, 55–67. [Google Scholar] [CrossRef] [PubMed]
  365. Alonso, R.; Pisa, D.; Fernández-Fernández, A.M.; Carrasco, L. Infection of fungi and bacteria in brain tissue from elderly persons and patients with Alzheimer’s disease. Front. Aging Neurosci. 2018, 10, 159. [Google Scholar] [CrossRef] [Green Version]
  366. Alonso, R.; Fernández-Fernández, A.M.; Pisa, D.; Carrasco, L. Multiple sclerosis and mixed microbial infections. Direct identification of fungi and bacteria in nervous tissue. Neurobiol. Dis. 2018, 117, 42–61. [Google Scholar] [CrossRef] [PubMed]
  367. Rex, J.H.; Pfaller, M.A. Has antifungal susceptibility testing come of age? Clin. Infect. Dis. 2002, 35, 982–989. [Google Scholar] [CrossRef] [Green Version]
  368. Eschenauer, G.A.; Carver, P.L. The evolving role of antifungal susceptibility testing. Pharmacother. J. Hum. Pharmacol. Drug Ther. 2013, 33, 465–475. [Google Scholar] [CrossRef] [Green Version]
  369. Lipsky, B.A.; Hoey, C. Topical antimicrobial therapy for treating chronic wounds. Clin. Infect. Dis. 2009, 49, 1541–1549. [Google Scholar] [CrossRef] [Green Version]
  370. Olivry, T.; DeBoer, D.J.; Favrot, C.; Jackson, H.A.; Mueller, R.S.; Nuttall, T.; Prélaud, P. Treatment of canine atopic dermatitis: 2010 clinical practice guidelines from the International Task Force on Canine Atopic Dermatitis. Vet. Dermatol. 2010, 21, 233–248. [Google Scholar] [CrossRef]
  371. Ray, P.; Singh, S.; Gupta, S. Topical antimicrobial therapy: Current status and challenges. Indian J. Med. Microbiol. 2019, 37, 299–308. [Google Scholar] [CrossRef]
  372. Wayne, P. Reference Method for Broth Dilution Antifungal Susceptibility Testing of filamentous Fungi; Clinical and Laboratory Standards Institute: Wayne, PA, USA, 2008. [Google Scholar]
  373. Peano, A.; Pasquetti, M.; Tizzani, P.; Chiavassa, E.; Guillot, J.; Johnson, E. Methodological issues in antifungal susceptibility testing of Malassezia pachydermatis. J. Fungi 2017, 3, 37. [Google Scholar] [CrossRef]
  374. Rhimi, W.; Theelen, B.; Boekhout, T.; Aneke, C.I.; Otranto, D.; Cafarchia, C. Conventional therapy and new antifungal drugs against Malassezia infections. Med. Mycol. 2021, 59, 215–234. [Google Scholar] [CrossRef] [PubMed]
  375. Romald, P.N.; Kindo, A.J.; Mahalakshmi, V.; Umadevi, U. Epidemiological pattern of Malassezia, its phenotypic identification and antifungal susceptibility profile to azoles by broth microdilution method. Indian J. Med. Microbiol. 2020, 38, 351–356. [Google Scholar] [CrossRef] [PubMed]
  376. Leong, C.; Buttafuoco, A.; Glatz, M.; Bosshard, P.P. Antifungal susceptibility testing of Malassezia spp. with an optimized colorimetric broth microdilution method. J. Clin. Microbiol. 2017, 55, 1883–1893. [Google Scholar] [CrossRef] [Green Version]
  377. Sharma, A. In Vitro Susceptibility of Malassezia Furfur to Azoles. Int. J. Health Sci. Res. 2015, 5, 158–164. [Google Scholar]
  378. Rojas, F.D.; Sosa, M.d.l.A.; Fernandez, M.S.; Cattana, M.E.; Cordoba, S.B.; Giusiano, G.E. Antifungal susceptibility of Malassezia furfur, Malassezia sympodialis, and Malassezia globosa to azole drugs and amphotericin B evaluated using a broth microdilution method. Sabouraudia 2014, 52, 641–646. [Google Scholar] [CrossRef] [Green Version]
  379. Gupta, A.K.; Kohli, Y.; Summerbell, R.C. Molecular differentiation of seven Malassezia species. J. Clin. Microbiol. 2000, 38, 1869–1875. [Google Scholar] [CrossRef] [Green Version]
  380. Brito, E.H.; Fontenelle, R.O.; Brilhante, R.S.; Cordeiro, R.A.; Júnior, F.A.S.; Monteiro, A.J.; Sidrim, J.J.; Rocha, M.F. Phenotypic characterization and in vitro antifungal sensitivity of Candida spp. and Malassezia pachydermatis strains from dogs. Vet. J. 2007, 174, 147–153. [Google Scholar] [CrossRef]
  381. Sugita, T.; Tajima, M.; Ito, T.; Saito, M.; Tsuboi, R.; Nishikawa, A. Antifungal activities of tacrolimus and azole agents against the eleven currently accepted Malassezia species. J. Clin. Microbiol. 2005, 43, 2824–2829. [Google Scholar] [CrossRef] [Green Version]
  382. Pasquetti, M.; Chiavassa, E.; Tizzani, P.; Danesi, P.; Peano, A. Agar diffusion procedures for susceptibility testing of Malassezia pachydermatis: Evaluation of Mueller-Hinton Agar Plus 2% glucose and 0.5 µg/ml methylene blue as the test medium. Mycopathologia 2015, 180, 153–158. [Google Scholar] [CrossRef]
  383. Muslimin, V.R.C.; Yogiswara, W.D.; Septiningrum, A.; Budiastuti, A.; Kustarini, S.I. In vitro antifungal susceptibility of Malassezia spp. to azole drugs. JPAD-J. Pak. Assoc. Dermatol. 2018, 28, 502–506. [Google Scholar]
  384. Arrese, J.; Fogouang, L.; Piérard-Franchimont, C.; Piérard, G. Euclidean and fractal computer-assisted corneofungimetry: A comparison of 2% ketoconazole and 1% terbinafine topical formulations. Dermatology 2002, 204, 222–227. [Google Scholar] [CrossRef] [PubMed]
  385. Piérard-Franchimont, C.; Vroome, V.; Cauwenbergh, G.; Piérard, G. Corneofungimetry bioassay on Malassezia spp. under ketoconazole and desonide influences. Ski. Pharmacol. Physiol. 2005, 18, 98–102. [Google Scholar] [CrossRef] [PubMed]
  386. Piérard-Franchimont, C.; Ausma, J.; Wouters, L.; Vroome, V.; Vandeplassche, L.; Borgers, M.; Cauwenbergh, G.; Piérard, G. Activity of the triazole antifungal R126638 as assessed by corneofungimetry. Ski. Pharmacol. Physiol. 2006, 19, 50–56. [Google Scholar] [CrossRef]
  387. Johnson, E.M. Issues in antifungal susceptibility testing. J. Antimicrob. Chemother. 2008, 61 (Suppl. S1), i13–i18. [Google Scholar] [CrossRef] [Green Version]
  388. Cantón, E.; Espinel-Ingroff, A.; Pemán, J. Trends in antifungal susceptibility testing using CLSI reference and commercial methods. Expert Rev. Anti-Infect. Ther. 2009, 7, 107–119. [Google Scholar] [CrossRef] [PubMed]
  389. Pfaller, M.A. Antifungal drug resistance: Mechanisms, epidemiology, and consequences for treatment. Am. J. Med. 2012, 125, S3–S13. [Google Scholar] [CrossRef]
  390. Rex, J.H.; Pfaller, M.A.; Galgiani, J.N.; Bartlett, M.S.; Espinel-Ingroff, A.; Ghannoum, M.A.; Lancaster, M.; Odds, F.C.; Rinaldi, M.G.; Walsh, T.J. Development of interpretive breakpoints for antifungal susceptibility testing: Conceptual framework and analysis of in vitro-in vivo correlation data for fluconazole, itraconazole, and Candida infections. Clin. Infect. Dis. 1997, 24, 235–247. [Google Scholar] [CrossRef]
  391. Willinger, B.; Apfalter, P.; Hirschl, A.M.; Makristathis, A.; Rotter, M.; Seibold, M. Susceptibility testing of Candida species: Comparison of NCCLS microdilution method with Fungitest®. Diagn. Microbiol. Infect. Dis. 2000, 38, 11–15. [Google Scholar] [CrossRef]
  392. Carrillo-Muñoz, A.J.; Rojas, F.; Tur-Tur, C.; de los Ángeles Sosa, M.; Diez, G.O.; Espada, C.M.; Payá, M.J.; Giusiano, G. In vitro antifungal activity of topical and systemic antifungal drugs against Malassezia species. Mycoses 2013, 56, 571–575. [Google Scholar] [CrossRef]
  393. Cafarchia, C.; Iatta, R.; Immediato, D.; Puttilli, M.R.; Otranto, D. Azole susceptibility of Malassezia pachydermatis and Malassezia furfur and tentative epidemiological cut-off values. Med. Mycol. 2015, 53, 743–748. [Google Scholar] [CrossRef] [Green Version]
  394. Peano, A.; Johnson, E.; Chiavassa, E.; Tizzani, P.; Guillot, J.; Pasquetti, M. Antifungal resistance regarding Malassezia pachydermatis: Where are we now? J. Fungi 2020, 6, 93. [Google Scholar] [CrossRef] [PubMed]
  395. Rojas, F.D.; Córdoba, S.B.; de los Ángeles Sosa, M.; Zalazar, L.C.; Fernández, M.S.; Cattana, M.E.; Alegre, L.R.; Carrillo-Muñoz, A.J.; Giusiano, G.E. Antifungal susceptibility testing of Malassezia yeast: Comparison of two different methodologies. Mycoses 2017, 60, 104–111. [Google Scholar] [CrossRef] [PubMed]
  396. Álvarez-Pérez, S.; García, M.E.; Peláez, T.; Blanco, J.L. Genotyping and antifungal susceptibility testing of multiple Malassezia pachydermatis isolates from otitis and dermatitis cases in pets: Is it really worth the effort? Med. Mycol. 2016, 54, 72–79. [Google Scholar] [PubMed] [Green Version]
  397. Velegraki, A.; Alexopoulos, E.C.; Kritikou, S.; Gaitanis, G. Use of fatty acid RPMI 1640 media for testing susceptibilities of eight Malassezia species to the new triazole posaconazole and to six established antifungal agents by a modified NCCLS M27-A2 microdilution method and Etest. J. Clin. Microbiol. 2004, 42, 3589–3593. [Google Scholar] [CrossRef] [Green Version]
  398. Rincón, S.; Cepero de García, M.; Espinel-Ingroff, A. A modified Christensen’s urea and CLSI broth microdilution method for testing susceptibilities of six Malassezia species to voriconazole, itraconazole, and ketoconazole. J. Clin. Microbiol. 2006, 44, 3429–3431. [Google Scholar] [CrossRef] [Green Version]
  399. Miranda, K.C.; de Araujo, C.R.; Costa, C.R.; Passos, X.S.; Fernandes, O.d.F.L.; Silva, M.d.R.R. Antifungal activities of azole agents against the Malassezia species. Int. J. Antimicrob. Agents 2007, 29, 281–284. [Google Scholar] [CrossRef]
  400. Yurayart, C.; Nuchnoul, N.; Moolkum, P.; Jirasuksiri, S.; Niyomtham, W.; Chindamporn, A.; Kajiwara, S.; Prapasarakul, N. Antifungal agent susceptibilities and interpretation of Malassezia pachydermatis and Candida parapsilosis isolated from dogs with and without seborrheic dermatitis skin. Med. Mycol. 2013, 51, 721–730. [Google Scholar] [CrossRef] [Green Version]
  401. Cafarchia, C.; Figueredo, L.A.; Iatta, R.; Montagna, M.T.; Otranto, D. In vitro antifungal susceptibility of Malassezia pachydermatis from dogs with and without skin lesions. Vet. Microbiol. 2012, 155, 395–398. [Google Scholar] [CrossRef]
  402. Weiler, C.B.; Jesus, F.P.K.D.; Nardi, G.H.; Loreto, É.S.; Santurio, J.M.; Coutinho, S.D.A.; Alves, S.H. Susceptibility variation of Malassezia pachydermatis to antifungal agents according to isolate source. Braz. J. Microbiol. 2013, 44, 175–178. [Google Scholar] [CrossRef]
  403. Watanabe, S.; Koike, A.; Kano, R.; Nagata, M.; Chen, C.; Hwang, C.-Y.; Hasegawa, A.; Kamata, H. In vitro susceptibility of Malassezia pachydermatis isolates from canine skin with atopic dermatitis to ketoconazole and itraconazole in East Asia. J. Vet. Med. Sci. 2014, 76, 579–581. [Google Scholar] [CrossRef] [Green Version]
  404. Cafarchia, C.; Figueredo, L.A.; Iatta, R.; Colao, V.; Montagna, M.T.; Otranto, D. In vitro evaluation of Malassezia pachydermatis susceptibility to azole compounds using E-test and CLSI microdilution methods. Med. Mycol. 2012, 50, 795–801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  405. Chiavassa, E.; Tizzani, P.; Peano, A. In vitro antifungal susceptibility of Malassezia pachydermatis strains isolated from dogs with chronic and acute otitis externa. Mycopathologia 2014, 178, 315–319. [Google Scholar] [CrossRef] [PubMed]
  406. Álvarez-Pérez, S.; Blanco, J.L.; Peláez, T.; Cutuli, M.; García, M.E. In vitro amphotericin B susceptibility of Malassezia pachydermatis determined by the CLSI broth microdilution method and Etest using lipid-enriched media. Antimicrob. Agents Chemother. 2014, 58, 4203–4206. [Google Scholar] [CrossRef] [Green Version]
  407. Iatta, R.; Immediato, D.; Montagna, M.T.; Otranto, D.; Cafarchia, C. In vitro activity of two amphotericin B formulations against Malassezia furfur strains recovered from patients with bloodstream infections. Med. Mycol. 2015, 53, 269–274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  408. Brilhante, R.S.N.; da Rocha, M.G.; de Melo Guedes, G.M.; de Oliveira, J.S.; dos Santos Araújo, G.; España, J.D.A.; Sales, J.A.; de Aguiar, L.; Paiva, M.d.A.N.; de Aguiar Cordeiro, R. Malassezia pachydermatis from animals: Planktonic and biofilm antifungal susceptibility and its virulence arsenal. Vet. Microbiol. 2018, 220, 47–52. [Google Scholar] [CrossRef] [PubMed]
  409. Bumroongthai, K.; Chetanachan, P.; Niyomtham, W.; Yurayart, C.; Prapasarakul, N. Biofilm production and antifungal susceptibility of co-cultured Malassezia pachydermatis and Candida parapsilosis isolated from canine seborrheic dermatitis. Sabouraudia 2016, 54, 544–549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  410. Kanafani, Z.A.; Perfect, J.R. Resistance to antifungal agents: Mechanisms and clinical impact. Clin. Infect. Dis. 2008, 46, 120–128. [Google Scholar] [CrossRef] [Green Version]
  411. Uchida, Y.; Onodera, S.; Nakade, T.; Otomo, K. Sterol composition in polyene antibiotic-sensitive and resistant strains of Malassezia pachydermatis. Vet. Res. Commun. 1994, 18, 183–187. [Google Scholar] [CrossRef]
  412. Kim, D.; Lim, Y.-R.; Ohk, S.O.; Kim, B.J.; Chun, Y.-J. Functional expression and characterization of CYP51 from dandruff-causing Malassezia globosa. FEMS Yeast Res. 2011, 11, 80–87. [Google Scholar] [CrossRef] [Green Version]
  413. Kim, M.; Cho, Y.-J.; Park, M.; Choi, Y.; Hwang, S.Y.; Jung, W.H. Genomic tandem quadruplication is associated with ketoconazole resistance in Malassezia pachydermatis. J. Microbiol. Biotechnol. 2018, 28, 1937–1945. [Google Scholar] [CrossRef] [Green Version]
  414. Park, M.; Cho, Y.J.; Lee, Y.W.; Jung, W.H. Genomic Multiplication and Drug Efflux Influence Ketoconazole Resistance in Malassezia restricta. Front. Cell. Infect. Microbiol. 2020, 10, 191. [Google Scholar] [CrossRef]
  415. Iatta, R.; Puttilli, M.R.; Immediato, D.; Otranto, D.; Cafarchia, C. The role of drug efflux pumps in Malassezia pachydermatis and Malassezia furfur defence against azoles. Mycoses 2017, 60, 178–182. [Google Scholar] [CrossRef] [PubMed]
  416. Ianiri, G.; Dagotto, G.; Sun, S.; Heitman, J. Advancing functional genetics through Agrobacterium-mediated insertional mutagenesis and CRISPR/Cas9 in the commensal and pathogenic yeast Malassezia. Genetics 2019, 212, 1163–1179. [Google Scholar] [CrossRef] [PubMed]
  417. Kumar, A.; Singh, K.; Sharma, A. Treatment of dermatitis in dogs associated with Malassezia pachydermatis. Indian Vet. J. 2002, 79, 730–732. [Google Scholar]
  418. Pinchbeck, L.R.; Hillier, A.; Kowalski, J.J.; Kwochka, K.W. Comparison of pulse administration versus once daily administration of itraconazole for the treatment of Malassezia pachydermatis dermatitis and otitis in dogs. J. Am. Vet. Med. Assoc. 2002, 220, 1807–1812. [Google Scholar] [CrossRef]
  419. Bensignor, E. Oral itraconazole as a pulse therapy for the treatment of canine Malassezia dermatitis: A randomised, blinded, comparative trial. Eur. J. Companion Anim. Pract. 2008, 18, 69–72. [Google Scholar]
  420. Negre, A.; Bensignor, E.; Guillot, J. Evidence-based veterinary dermatology: A systematic review of interventions for Malassezia dermatitis in dogs. Vet. Dermatol. 2009, 20, 1–12. [Google Scholar] [CrossRef]
  421. Åhman, S.; Perrins, N.; Bond, R. Treatment of Malassezia pachydermatis-associated seborrhoeic dermatitis in Devon Rex cats with itraconazole–a pilot study. Vet. Dermatol. 2007, 18, 171–174. [Google Scholar] [CrossRef]
  422. Bensignor, E. Treatment of Malassezia overgrowth with itraconazole in 15 cats. Vet. Rec. 2010, 167, 1011. [Google Scholar] [CrossRef]
  423. Berger, D.J.; Lewis, T.P.; Schick, A.E.; Stone, R.T. Comparison of once-daily versus twice-weekly terbinafine administration for the treatment of canine Malassezia dermatitis–a pilot study. Vet. Dermatol. 2012, 23, 418-e79. [Google Scholar] [CrossRef]
  424. Guillot, J.; Bensignor, E.; Jankowski, F.; Seewald, W.; Chermette, R.; Steffan, J. Comparative efficacies of oral ketoconazole and terbinafine for reducing Malassezia population sizes on the skin of Basset Hounds. Vet. Dermatol. 2003, 14, 153–157. [Google Scholar] [CrossRef] [PubMed]
  425. Rosales, M.S.; Marsella, R.; Kunkle, G.; Harris, B.L.; Nicklin, C.F.; Lopez, J. Comparison of the clinical efficacy of oral terbinafine and ketoconazole combined with cephalexin in the treatment of Malassezia dermatitis in dogs–a pilot study. Vet. Dermatol. 2005, 16, 171–176. [Google Scholar] [CrossRef] [PubMed]
  426. Gimmler, J.R.; White, A.G.; Kennis, R.A.; Cruz-Espindola, C.; Boothe, D.M. Determining canine skin concentrations of terbinafine to guide the treatment of Malassezia dermatitis. Vet. Dermatol. 2015, 26, 411-e96. [Google Scholar] [CrossRef] [PubMed]
  427. Sickafoose, L.; Hosgood, G.; Snook, T.; Westermeyer, R.; Merchant, S. A noninferiority clinical trial comparing fluconazole and ketoconazole in combination with cephalexin for the treatment of dogs with Malassezia dermatitis. Vet. Ther. Res. Appl. Vet. Med. 2010, 11, E1–E13. [Google Scholar]
  428. Villars, V.; Jones, T. Clinical efficacy and tolerability of terbinafine (Lamisil)—a new topical and systemic fungicidal drug for treatment of dermatomycoses. Clin. Exp. Dermatol. 1989, 14, 124–127. [Google Scholar] [CrossRef]
  429. Gupta, A.K.; Foley, K.A. Antifungal treatment for pityriasis versicolor. J. Fungi 2015, 1, 13–29. [Google Scholar] [CrossRef]
  430. Gupta, A.K.; Lyons, D.C. Pityriasis versicolor: An update on pharmacological treatment options. Expert Opin. Pharmacother. 2014, 15, 1707–1713. [Google Scholar] [CrossRef]
  431. Hawkins, D.M.; Smidt, A.C. Superficial fungal infections in children. Pediatric Clin. 2014, 61, 443–455. [Google Scholar] [CrossRef]
  432. Cheong, W.K.; Yeung, C.K.; Torsekar, R.G.; Suh, D.H.; Ungpakorn, R.; Widaty, S.; Azizan, N.Z.; Gabriel, M.T.; Tran, H.K.; Chong, W.S. Treatment of seborrhoeic dermatitis in Asia: A consensus guide. Ski. Appendage Disord. 2015, 1, 187–196. [Google Scholar] [CrossRef] [Green Version]
  433. Das, A.; Panda, S. Use of topical corticosteroids in dermatology: An evidence-based approach. Indian J. Dermatol. 2017, 62, 237. [Google Scholar]
  434. Skorvanek, M.; Bhatia, K.P. The skin and Parkinson’s disease: Review of clinical, diagnostic, and therapeutic issues. Mov. Disord. Clin. Pract. 2017, 4, 21–31. [Google Scholar] [CrossRef] [PubMed]
  435. Augustin, M.; Kirsten, N.; Körber, A.; Wilsmann-Theis, D.; Itschert, G.; Staubach-Renz, P.; Maul, J.T.; Zander, N. Prevalence, predictors and comorbidity of dry skin in the general population. J. Eur. Acad. Dermatol. Venereol. 2019, 33, 147–150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  436. Victoire, A.; Magin, P.; Coughlan, J.; van Driel, M.L. Interventions for infantile seborrhoeic dermatitis (including cradle cap). In Cochrane Database of Systematic Reviews; John Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2019. [Google Scholar]
  437. Abbas, Z.; Ghodsi, S.Z.; Abedeni, R. Effect of itraconazole on the quality of life in patients with moderate to severe seborrheic dermatitis: A randomized, placebo-controlled trial. Dermatol. Pract. Concept. 2016, 6, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  438. Lévy, A.; Feuilhade de Chauvin, M.; Dubertret, L.; Morel, P.; Flageul, B. In Malassezia folliculitis: Characteristics and therapeutic response in 26 patients. Ann. Dermatol. Vénéréologie 2007, 11, 823–828. [Google Scholar] [CrossRef]
  439. Suzuki, C.; Hase, M.; Shimoyama, H.; Sei, Y. Treatment outcomes for malassezia folliculitis in theDermatology department of a university hospital in Japan. Med. Mycol. J. 2016, 57, E63–E66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  440. Tsai, Y.-C.; Wang, J.-Y.; Wu, Y.-H.; Wang, Y.-J. Clinical differences in pediatric and adult Malassezia folliculitis: Retrospective analysis of 321 cases over 9 years. J. Am. Acad. Dermatol. 2019, 81, 278–280. [Google Scholar] [CrossRef] [Green Version]
  441. Rhie, S.; Turcios, R.; Buckley, H.; Suh, B. Clinical features and treatment of Malassezia folliculitis with fluconazole in orthotopic heart transplant recipients. J. Heart Lung Transplant. 2000, 19, 215–219. [Google Scholar] [CrossRef]
  442. Prindaville, B.; Belazarian, L.; Levin, N.A.; Wiss, K. Pityrosporum folliculitis: A retrospective review of 110 cases. J. Am. Acad. Dermatol. 2018, 78, 511–514. [Google Scholar] [CrossRef]
  443. Lee, J.W.; Kim, B.J.; Kim, M.N. Photodynamic therapy: New treatment for recalcitrant Malassezia folliculitis. Lasers Surg. Med. Off. J. Am. Soc. Laser Med. Surg. 2010, 42, 192–196. [Google Scholar] [CrossRef]
  444. Lee, J.W.; Lee, H.I.; Kim, M.N.; Kim, B.J.; Chun, Y.J.; Kim, D. Topical photodynamic therapy with methyl aminolevulinate may be an alternative therapeutic option for the recalcitrant Malassezia folliculitis. Int. J. Dermatol. 2011, 50, 488–490. [Google Scholar] [CrossRef]
  445. Buentke, E.; Zargari, A.; Heffler, L.; Avila-Carino, J.; Savolainen, J.; Scheynius, A. Uptake of the yeast Malassezia furfur and its allergenic components by human immature CD1a+ dendritic cells. Clin. Exp. Allergy 2000, 30, 1759–1770. [Google Scholar] [CrossRef]
  446. Selander, C.; Zargari, A.; Möllby, R.; Rasool, O.; Scheynius, A. Higher pH level, corresponding to that on the skin of patients with atopic eczema, stimulates the release of Malassezia sympodialis allergens. Allergy 2006, 61, 1002–1008. [Google Scholar] [CrossRef] [PubMed]
  447. Vickery, B.P. Skin barrier function in atopic dermatitis. Curr. Opin. Pediatrics 2007, 19, 89–93. [Google Scholar] [CrossRef] [PubMed]
  448. Darabi, K.; Hostetler, S.G.; Bechtel, M.A.; Zirwas, M. The role of Malassezia in atopic dermatitis affecting the head and neck of adults. J. Am. Acad. Dermatol. 2009, 60, 125–136. [Google Scholar] [CrossRef] [PubMed]
  449. Brodská, P.; Panzner, P.; Pizinger, K.; Schmid-Grendelmeier, P. IgE-mediated sensitization to Malassezia in atopic dermatitis: More common in male patients and in head and neck type. Dermatitis 2014, 25, 120–126. [Google Scholar] [CrossRef] [PubMed]
  450. Kaffenberger, B.H.; Mathis, J.; Zirwas, M.J. A retrospective descriptive study of oral azole antifungal agents in patients with patch test–negative head and neck predominant atopic dermatitis. J. Am. Acad. Dermatol. 2014, 71, 480–483. [Google Scholar] [CrossRef] [PubMed]
  451. Kolmer, H.L.; Taketomi, E.A.; Hazen, K.C.; Hughs, E.; Wilson, B.B.; Platts-Mills, T.A. Effect of combined antibacterial and antifungal treatment in severe atopic dermatitis. J. Allergy Clin. Immunol. 1996, 98, 702–707. [Google Scholar] [CrossRef]
  452. Ruiz-Villaverde, R.; Sanchez-Cano, D.; Lopez-Delgado, D. In Head and neck dermatitis: Successful response to terbinafine. J. Am. Acad. Dermatol. 2018, 79, AB150. [Google Scholar]
  453. Mayser, P.; Kupfer, J.; Nemetz, D.; Schäfer, U.; Nilles, M.; Hort, W.; Gieler, U. Treatment of head and neck dermatitis with ciclopiroxolamine cream–results of a double-blind, placebo-controlled study. Ski. Pharmacol. Physiol. 2006, 19, 153–158. [Google Scholar] [CrossRef]
  454. Mehra, T.; Köberle, M.; Braunsdorf, C.; Mailänder-Sanchez, D.; Borelli, C.; Schaller, M. Alternative approaches to antifungal therapies. Exp. Dermatol. 2012, 21, 778–782. [Google Scholar] [CrossRef] [Green Version]
  455. Khosravi, A.; Shokri, H.; Fahimirad, S. Efficacy of medicinal essential oils against pathogenic Malassezia sp. isolates. J. Mycol. Médicale 2016, 26, 28–34. [Google Scholar] [CrossRef]
  456. Gupta, A.K.; Versteeg, S.G. Topical treatment of facial seborrheic dermatitis: A systematic review. Am. J. Clin. Dermatol. 2017, 18, 193–213. [Google Scholar] [CrossRef]
  457. Oliveira, A.M.; Devesa, J.S.; Hill, P.B. In vitro efficacy of a honey-based gel against canine clinical isolates of Staphylococcus pseudintermedius and Malassezia pachydermatis. Vet. Dermatol. 2018, 29, 180-e65. [Google Scholar] [CrossRef] [PubMed]
  458. Sjöström, Y.; Mellor, P.; Bergvall, K. A novel non-azole topical treatment reduces Malassezia numbers and associated dermatitis: A short term prospective, randomized, blinded and placebo-controlled trial in naturally infected dogs. Vet. Dermatol. 2018, 29, 14-e7. [Google Scholar] [CrossRef] [PubMed]
  459. Sastoque, A.; Triana, S.; Ehemann, K.; Suarez, L.; Restrepo, S.; Wösten, H.; de Cock, H.; Fernández-Niño, M.; González Barrios, A.F.; Celis Ramírez, A.M. New Therapeutic Candidates for the Treatment of Malassezia pachydermatis -Associated Infections. Sci. Rep. 2020, 10, 4860. [Google Scholar] [CrossRef]
  460. Arendrup, M.C.; Boekhout, T.; Akova, M.; Meis, J.F.; Cornely, O.A.; Lortholary, O.; Group, E.E.S. ECMM, ESCMID and ECMM joint clinical guidelines for the diagnosis and management of rare invasive yeast infections. Clin. Microbiol. Infect. 2014, 20, 76–98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  461. Mickelsen, P.A.; Viano-Paulson, M.C.; Stevens, D.A.; Diaz, P.S. Clinical and microbiological features of infection with Malassezia pachydermatis in high-risk infants. J. Infect. Dis. 1988, 157, 1163–1168. [Google Scholar] [CrossRef] [PubMed]
  462. Jesus, F.; Lautert, C.; Zanette, R.; Mahl, D.; Azevedo, M.; Machado, M.; Dutra, V.; Botton, S.; Alves, S.; Santurio, J. In vitro susceptibility of fluconazole-susceptible and-resistant isolates of Malassezia pachydermatis against azoles. Vet. Microbiol. 2011, 152, 161–164. [Google Scholar] [CrossRef]
  463. Chen, I.-T.; Chen, C.-C.; Huang, H.-C.; Kuo, K.-C. Malassezia furfur emergence and candidemia trends in a neonatal intensive care unit during 10 years: The experience of fluconazole prophylaxis in a single hospital. Adv. Neonatal Care 2020, 20, E3. [Google Scholar] [CrossRef] [PubMed]
  464. Ashbee, H. Update on the genus Malassezia. Med. Mycol. 2007, 45, 287–303. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Predominant Malassezia species found as commensals in the sinonasal cavity (A), on the skin of the forehead and external ear canal (B), in the lungs and gastrointestinal tract (C) and on the skin of the occiput, back and groin (D). The relative diversity of Malassezia species found in healthy skin is shown in (E,F). The predominant species isolated from the culture of the skin on the side of the nose of Caucasians in Switzerland were M. restricta and M. sympodialis, while M. globosa was absent. In contrast, sampling from the skin of the noses of 4 ethnic groups (Chinese, Malay, Indian and Caucasian) in Singapore overall revealed M. globosa, M. furfur and M. restricta to be the dominant species [95].
Figure 1. Predominant Malassezia species found as commensals in the sinonasal cavity (A), on the skin of the forehead and external ear canal (B), in the lungs and gastrointestinal tract (C) and on the skin of the occiput, back and groin (D). The relative diversity of Malassezia species found in healthy skin is shown in (E,F). The predominant species isolated from the culture of the skin on the side of the nose of Caucasians in Switzerland were M. restricta and M. sympodialis, while M. globosa was absent. In contrast, sampling from the skin of the noses of 4 ethnic groups (Chinese, Malay, Indian and Caucasian) in Singapore overall revealed M. globosa, M. furfur and M. restricta to be the dominant species [95].
Jof 08 00708 g001
Figure 2. Malassezia species colonization in healthy dogs (A) and cats (B).
Figure 2. Malassezia species colonization in healthy dogs (A) and cats (B).
Jof 08 00708 g002
Figure 3. Overview of Malassezia-associated skin diseases in humans. Typical affected areas for pityriasis versicolor (A), seborrheic dermatitis (B), Malassezia-associated folliculitis (C) and head and neck dermatitis (D), as well as commonly involved Malassezia species, are shown.
Figure 3. Overview of Malassezia-associated skin diseases in humans. Typical affected areas for pityriasis versicolor (A), seborrheic dermatitis (B), Malassezia-associated folliculitis (C) and head and neck dermatitis (D), as well as commonly involved Malassezia species, are shown.
Jof 08 00708 g003
Figure 4. Common affected skin sites in dogs and cats with Malassezia-associated dermatitis and otitis externa. In cats, generalized diseases can also be seen.
Figure 4. Common affected skin sites in dogs and cats with Malassezia-associated dermatitis and otitis externa. In cats, generalized diseases can also be seen.
Jof 08 00708 g004
Table 1. Classification of Malassezia by species, reference strain, hosts and phylogenetic clades.
Table 1. Classification of Malassezia by species, reference strain, hosts and phylogenetic clades.
SpeciesReference Strain/GenBank Accession Genome NumberDescribed HostsClade
M. furfurCBS 14141, GCA_009938135Human, Cat, Dog, Cattle, Pig, Goat, Elk, Horse, Sheep, Elephant, Monkey, Ostrich, PelicanA
M. brasiliensis *MA 1455ParrotA
M. yamatoensisMY9725, GCA_001264885Human, CatA
M. psittaci *MA 1454 ParrotA
M. obtusaCBS 7876, GCA_001264985Human, Cat, Dog, Goat, HorseA
M. japonicaCBS 9431, GCA_001264785Human, CatA
M. vespertilionisCBS 15041, GCA_002818225BatA
M. globosaCBS 7966, GCA_001264805Human, Cat, Dog, Cattle, Goat, Horse, Sheep, CheetahB
M. restrictaCBS 7877, GCA_001264765Human, Cat, Dog, Cattle, Goat, Horse, SheepB
M. arunalokeiCBS 13387, GCA_020085095Human, DogB
M. sympodialisATCC 42132, GCA_001264925Human, Dog, Cat, Pig, Cattle, Goat, Horse, Sheep, ChickenB
M. dermatisCBS 9169, GCA_001264665Human, CatB
M. capraeCBS 10434, GCA_001264625Goat, Horse, HumanB
M. equinaCBS 9969, GCA_001264685Horse, CattleB
M. nanaJCM 12085, GCA_001600835Cat, Dog, Cattle, HorseB
M. pachydermatisCBS 1879, GCA_001264975Human, Dog, Cat, Pig, Goat, Rabbit, Various exotic and wild mammals, Birds (Thraupidae, Macaw)B
M. cuniculiCBS 11721, GCA_001264635RabbitC
M. slooffiaeCBS 7956, GCA_001264965Human, Cat Cattle, Sheep, Pig, Goat, HorseC
* = whole genome not available.
Table 2. Breed predisposition for Malassezia dermatitis in companion animals.
Table 2. Breed predisposition for Malassezia dermatitis in companion animals.
Dog BreedsCat Breeds
West Highland White TerrierDevon Rex
English SetterSphynx
Basset Hound
Boxer
American Cocker Spaniel
Poodle
Dachshund
Australian Silky Terrier
Shih Tzu
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Hobi, S.; Cafarchia, C.; Romano, V.; Barrs, V.R. Malassezia: Zoonotic Implications, Parallels and Differences in Colonization and Disease in Humans and Animals. J. Fungi 2022, 8, 708. https://doi.org/10.3390/jof8070708

AMA Style

Hobi S, Cafarchia C, Romano V, Barrs VR. Malassezia: Zoonotic Implications, Parallels and Differences in Colonization and Disease in Humans and Animals. Journal of Fungi. 2022; 8(7):708. https://doi.org/10.3390/jof8070708

Chicago/Turabian Style

Hobi, Stefan, Claudia Cafarchia, Valentina Romano, and Vanessa R. Barrs. 2022. "Malassezia: Zoonotic Implications, Parallels and Differences in Colonization and Disease in Humans and Animals" Journal of Fungi 8, no. 7: 708. https://doi.org/10.3390/jof8070708

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