Next Article in Journal
Antifungal Volatile Organic Compounds from Talaromyces purpureogenus CEF642N: Insights from One Strain Many Compounds (OSMAC) Strategy for Controlling Verticillium dahliae in Cotton
Previous Article in Journal
Morphology and Phylogeny Reveal New Species and Records of Diplodia, Dothiorella, and Phaeobotryon Associated with Tree Cankers in Xizang, China
Previous Article in Special Issue
The Gut Mycobiome for Precision Medicine
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Gut Mycobiome: Latest Findings and Current Knowledge Regarding Its Significance in Human Health and Disease

by
Bogdan Severus Gaspar
1,2,
Oana Alexandra Roşu
3,4,†,
Robert-Mihai Enache
5,†,
Monica Manciulea (Profir)
3,4,†,
Luciana Alexandra Pavelescu
3 and
Sanda Maria Creţoiu
3,*
1
Department of Surgery, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
2
Surgery Clinic, Bucharest Emergency Clinical Hospital, 014461 Bucharest, Romania
3
Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
4
Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania
5
Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
J. Fungi 2025, 11(5), 333; https://doi.org/10.3390/jof11050333
Submission received: 5 March 2025 / Revised: 16 April 2025 / Accepted: 21 April 2025 / Published: 22 April 2025
(This article belongs to the Special Issue Gut Mycobiome, 2nd Edition)

Abstract

:
The gut mycobiome, the fungal component of the gut microbiota, plays a crucial role in health and disease. Although fungi represent a small fraction of the gut ecosystem, they influence immune responses, gut homeostasis, and disease progression. The mycobiome’s composition varies with age, diet, and host factors, and its imbalance has been linked to conditions such as inflammatory bowel disease (IBD) and metabolic disorders. Advances in sequencing have expanded our understanding of gut fungi, but challenges remain due to methodological limitations and high variability between individuals. Emerging therapeutic strategies, including antifungals, probiotics, fecal microbiota transplantation, and dietary interventions, show promise but require further study. This review highlights recent discoveries on the gut mycobiome, its interactions with bacteria, its role in disease, and potential clinical applications. A deeper understanding of fungal contributions to gut health will help develop targeted microbiome-based therapies.

1. Introduction

The human gastrointestinal tract is a complex ecosystem hosting a diverse array of microorganisms, including bacteria, viruses, and fungi. While bacterial communities have long been the primary focus of gut microbiome research, the fungal component, known as the mycobiome, has gained increasing attention for its crucial roles in health and disease [1].
Recent studies using amplicon sequencing and culture-based techniques suggest that the mycobiome may develop before birth [2]. However, this remains controversial, as other studies argue that the mycobiome emerges only after birth [3]. Despite these discrepancies, there is broad agreement that the gut mycobiome evolves significantly from infancy to adulthood, exhibiting greater variability and instability between individuals compared to the bacterial microbiota [4,5]. Multiple factors influence its composition throughout life, including maternal and infant diet, delivery mode, maternal age, geography, ethnicity, lifestyle, and dietary habits. For example, increased levels of Saccharomyces cerevisiae are observed in urban populations, while specific fungal species from the Penicillium and Naumoyozyma genera are more prevalent in Chinese individuals. Likewise, dietary patterns influence mycobiome composition, with Candida spp. flourishing in carbohydrate-rich diets and showing reduced abundance in protein-enriched diets [6,7,8,9].
Although fungi constitute only 0–1% of the gut microbiota, they have profound effects on host physiology and immunity [4]. Advances in deep sequencing and microbial cultivation have led to the identification of numerous fungal species, including keystone species involved in human health and disease [4,10]. The gut mycobiome interacts dynamically with bacteria and viruses, shaping health and disease states [4]. For instance, Candida spp. influence bacterial communities by competing for nutrients, producing antimicrobial peptides, and generating secondary metabolites [11,12]. Dysbiosis-driven increases in Candida albicans levels can negatively affect dominant gut bacteria in preterm infants and reduce the efficacy of fecal microbiota transplantation (FMT) [11,12]. Another key player is Malassezia restricta, which can activate the C3 complement cascade, driving gut inflammation and exacerbating IBD and pancreatic cancer [13,14,15]. Additionally, bacterial–fungal interactions shape microbiome stability, with bacteria modulating fungal germination and hyphal growth via fatty acid production [16,17]. Notably, C. albicans has been found to protect Helicobacter pylori from stomach acidity by sequestering it within its vacuoles, demonstrating interkingdom interactions that impact colonization and survival [18].
In dysbiosis, the gut mycobiome becomes dominated by opportunistic fungal pathogens, potentially influencing immune responses and contributing to both intestinal and extra-intestinal diseases. These include inflammatory bowel disease (IBD), irritable bowel syndrome, celiac disease, metabolic disorders, neurological conditions, autoimmune diseases, and even cancer [14,19,20,21,22]. Dysbiosis can arise from bacterial or viral infections, antifungal treatments, or chronic alcohol consumption, with associated shifts in fungal composition, such as C. albicans overgrowth and bacterial microbiome alterations [12,23,24]. While the exact mechanisms remain incompletely understood, studies suggest that these interactions change over the disease course [4]. One potential mechanism is the activation of Th17 and IL-23-mediated immune responses by C. albicans cell wall components, leading to microbiota disruption and inflammation [11,25]. Increased C. albicans levels can trigger antifungal Th17 immune responses, which may reach the lungs and contribute to airway inflammation [26]. The interactions between bacteria and fungi within the gut microbiota are summarized in Figure 1.
Recent research has not only uncovered the diversity and functional significance of gut fungi but also highlighted their potential therapeutic applications [4]. Among the most researched strategies for modulating the gut mycobiome are dietary interventions, probiotics (both fungal and bacterial strains), antifungals, antibiotics, and fecal microbiota transplantation (FMT) [4]. For instance, coconut oil-rich diets can inhibit C. albicans overgrowth, while enteral nutrition in pediatric Crohn’s disease (CD) patients has been linked to reduced C. albicans abundance [27,28]. Probiotics, particularly bacterial and fungal strains, can produce antimicrobial compounds that limit fungal colonization, combat infections, and treat various diseases, such as Clostridium difficile infection (CDI) and inflammatory bowel disease [4]. Antifungal therapies, such as fluconazole, have been effective in reducing C. albicans colonization in ulcerative colitis (UC) but may disrupt microbiota balance in prolonged use [4,29]. Notably, antibiotics can have a more enduring impact on the gut fungal community than on bacterial populations [30]. Lastly, FMT has shown promise in modulating fungal overgrowth, particularly in UC (in the case of increased Candida spp. abundance in recipients before FMT and reduced Candida spp. levels after FMT), though its effectiveness may be compromised by pre-existing fungal dysbiosis [31,32].
This narrative review aims to synthesize current knowledge on the gut mycobiome, emphasizing its development, interactions with bacterial microbiota, role in health and disease, and potential therapeutic interventions.

2. Gut Mycobiome: Important Fungi Residing in the Human Gut

The human gut mycobiome is mainly composed of yeast species, with the most commonly identified genera being Candida, Saccharomyces, and Malassezia [33,34]. However, the relative abundance of these genera can vary significantly amongst individuals, being strongly influenced by factors such as diet, geography, antimicrobial use, and health status [34,35].
The composition of the gut mycobiome varies with age [4]. Recent longitudinal studies have significantly advanced our understanding of gut mycobiota colonization dynamics in early life and beyond [36]. Unlike earlier studies, which primarily reported a gradual increase in fungal diversity starting from infancy, the updated data reveal that colonization begins robustly during the first month and is rapidly influenced by factors such as the mode of delivery (vertical transmission), feeding practices (breastfeeding vs. formula), and environmental exposures [36]. During the first month of life, the gut mycobiome is predominantly composed of fungal species from the phyla Candida spp., Ascomycota (Saccharomycetales order), and Basidiomycota (Malasseziales order), with Malasseziales experiencing a significant decline by five months of age. By 5 months, fungal diversity increases (gradual increase in fungal diversity, particularly within the Saccharomycetales order), shaped by environmental exposure and dietary transition (solid foods and other dietary changes) [36,37]. These updated findings not only refine the timeline of mycobiome maturation but also underscore the impact of early-life interventions on long-term gut health [36]. As infants begin consuming a more diverse diet, the dominant fungal genera in the gut shift to include Saccharomyces cerevisiae, Cystofilobasidium spp., Ascomycota spp., and Monographella spp., reflecting the influence of solid food introduction and microbial maturation [36,38]. Over the following years, the gut mycobiome gradually transitions to resemble the adult gut mycobiome, which is characterized by the presence of Ascomycota, Basidiomycota, and Zygomycota, with Candida, Saccharomyces, and Cladosporium being the most abundant genera in healthy adults [39]. Fungal composition in adulthood is shaped by diet, lifestyle, immune status, and host genetics [36]. In elderly populations, changes in immunity, diet, and medication use may contribute to shifts in fungal composition, with an increased prevalence of Penicillium, Candida, Saccharomyces, and Aspergillus becoming more prevalent [36,40]. The changes in gut mycobiome composition throughout life are illustrated in Figure 2.
Recent longitudinal studies using TEDDY metagenomic libraries have provided valuable insights into early-life gut mycobiome dynamics and their potential implications for autoimmunity. In particular, Auchtung et al. demonstrated that children at risk for type 1 diabetes exhibit specific mycobiome signatures before clinical onset [36]. The conclusions of this work emphasize that early fungal colonization patterns—shaped by factors such as delivery mode, feeding practices, and environmental exposures—may serve as potential biomarkers for subsequent autoimmune risk. Moreover, integrating these findings with recent insights into the mechanistic roles of fungal components in immune regulation and cross-kingdom metabolic crosstalk extends our understanding of how gut fungi might actively influence disease development, as also discussed in broader reviews [41,42,43]. These recent advances not only refine our picture of the temporal dynamics of mycobiome maturation in infancy but also provide a strong rationale for further investigating the role of early-life fungal dysbiosis in the etiology of type 1 diabetes.
A comprehensive study by Hallen et al. identified the genera Cutaneotrichospon, Saccharomyces, Aspergillus, Candida, and Malassezia to be the most prevalent fungal genera in fecal samples from healthy human gut microbiota [39]. These findings align with previous research by Nash et al., which also identified Saccharomyces, Candida, and Cladosporium as dominant fungal genera in the human gut [5].
Candida species are commensal in the human gut but can become pathogenic under specific conditions such as antibiotic use, immunosuppression, and gut dysbiosis [44,45]. C. albicans, C. glabrata, C. tropicalis, C. parapsilosis, and C. lusitaniae are the species of Candida most frequently associated with infection and inflammation [46]. The excessive use of broad-spectrum antibiotics profoundly impacts fungi residing in the human gut, particularly C. albicans, leading to overgrowth and antibiotic-associated diarrhea [47]. A study by Sokol et al. identified elevated levels of Candida in patients with gastrointestinal disorders such as CD and UC [48]. C. albicans facilitates mucosal invasion by adhering to the intestinal epithelial cells and producing virulence factors [49]. In a healthy gut, Candida is typically kept in check by Lactobacillus species, which produce antimicrobial compounds that inhibit its overgrowth [50]. In addition, colonocytes play a role in bringing C. albicans back to their commensal state by signaling immune cells and activating IL-22, which stimulates the production of secretory IgA and β-defensins [34]. Charlet et al. found that in mice with induced colitis, elevated C. glabrata levels correlated with increased inflammation and mortality, primarily due to higher chitin levels in the fungal cell wall during colonization. Conversely, a deficiency in chitin synthase-3 was linked to reduced inflammatory markers [51]. Elevated levels of Candida tropicalis have been directly linked to heightened inflammation in CD in humans and colitis in mice. This occurs through mucin-degrading bacterial modifications, tight junction protein expression alterations, and increased gut permeability [52,53]. A study by Sun et al. demonstrated that Candida parapsilosis can induce diet-related obesity in fungi-free mice by increasing fungal lipase production, leading to elevated free fatty acids in the gut. This highlights the potential of targeting this fungus in obesity treatment strategies [54]. Clavispora lusitaniae (formerly Candida lusitaniae) primarily affects immunocompromised patients, such as those with hematological malignancies, causing severe infections in the digestive and urinary systems. Additionally, it has been associated with resistance to certain antifungal agents, including amphotericin B [55]. Another Candida species, Geotrichum candidum, was identified in a study by Noor-UI et al. as a potential probiotic for fish, demonstrating benefits such as enhanced feed utilization, improved immunity, and reduced disease resistance [56].
The genus Saccharomyces includes several species, with S. cerevisiae being the most well-known. S. cerevisiae is a common yeast found in dietary sources such as fermented foods and bread [57]. S. cerviaseae var. boulardii (S. boulardii) is a probiotic strain that has been shown to modulate the gut immune response, enhance the integrity of the gut barrier, and protect against traveler’s diarrhea and CDI [58,59]. According to several studies, S. boulardii has also been shown to have antineoplastic effects. One study by Chen et al. showed that S. boulardii can inactivate the EGFR pathways, reduces cell proliferation, and promotes apoptosis. In addition, it was shown to reduce intestinal tumor growth and dysplasia in mice with colon cancer [60].
Malassezia is traditionally considered a skin-dwelling fungus associated with several skin conditions, such as dermatitis and pityriasis. It is frequently identified in several other environments, including the murine and human gut [13]. Due to technological advances, specifically NGS studies, several publications have reported the presence of Malassezia in healthy gut samples in recent years, confirming that it is part of the gut microbiota [61,62,63,64]. Malassezia has been associated with several diseases, including inflammatory bowel disease, colonic polyps, intestinal cancer, pancreatic cancer, and Alzheimer’s disease [14,15,48,65,66]. The role of Malassezia in developing inflammatory bowel disease has been characterized by Sokol et al. Limon et al., who suggested that Malassezia might exacerbate inflammation in genetically susceptible individuals [15,48]. Several researchers have demonstrated the role of the gut microbiota in the development of colorectal cancer (CRC); however, the implications of Malassezia strains have only recently been demonstrated by Gao et al. and Coker et al. [65,66,67]. How Malassezia can lead to the development of CRC remains unclear. However, Malassezia species have been associated with inflammatory responses that could lead to DNA damage and tumorigenesis [68]. In addition, it can produce potent aryl hydrocarbon receptor (AhR) ligands, which may promote tumor development by modifying UV radiation carcinogenesis, altering cell cycle progression and inhibiting apoptosis [69].
Aspergillus and Penicillium are common environmental molds that can colonize the human gut primarily through dietary intake. Although they are considered transient members of the gut mycobiome, certain species within these genera produce mycotoxins that have important health consequences. Aspergillus species, particularly A. flavus and A. parasiticus, produce alfatoxins, which are hepatotoxins that have been associated with hepatocellular carcinoma [70]. These Aspergillus species are frequently found in peanuts or peanut butter, and their growth is favored by warm, humid weather [71]. Similarly, Penicillium species, such as P. verrucosum and P. nordicum, produce ochratoxin A, a nephrotoxic compound associated with kidney damage and an increased risk of renal disease, including the development of urothelial carcinoma. Ochratoxin A contamination has been reported in products like cereals, coffee, and dried fruits [72]. Table 1 synthesizes the most important key findings regarding the effects of different fungal genera on human health and disease.

3. The Balance Between Gut Fungi and Bacteria in Healthy Subjects

In healthy people, bacteria and fungi live in a delicate equilibrium, fighting for resources and available space while occasionally interacting favorably. Preventing bacterial overgrowth and preserving gut homeostasis rely on this balance [73].
Certain bacteria and fungi interact synergistically to support gut health. For example, some bacterial species produce biofilms that help stabilize fungal populations, while fungi contribute to fiber degradation, supporting bacterial fermentation [73,74]. For instance, fungi like Candida can improve their colonization by using the biofilms produced by specific gut bacteria [75,76].
A balanced gut microbiome helps regulate digestion, nutrient absorption, and immune function [77]. This harmony reduces the risk of gastrointestinal disturbances, infections, and chronic inflammation, supporting overall metabolic and immune health [78,79].
Regarding the role of bacterial metabolites in fungal regulation inside the gut microbiota of healthy individuals, many aspects of SCFAs (short-chain fatty acids) can be emphasized [80,81]. SCFAs such as butyrate, propionate, and acetate, produced by the bacterial fermentation of dietary fibers, play a critical role in regulating gut fungi. These metabolites help inhibit pathogenic fungi, maintain gut pH, and strengthen the intestinal barrier. By modifying immune responses, SCFAs promote immunological tolerance and lower systemic inflammation [82,83]. Furthermore, regarding the systemic health benefits of SCFAs, there is evidence supporting the influence SCFAs have on energy metabolism, insulin sensitivity, and lipid regulation, contributing to a healthy weight and reducing the risk of metabolic disease [84,85]. SCFAs can influence the gut–brain axis, supporting cognitive function, mood regulation, and stress resilience in healthy individuals [86,87]. Another positive aspect of gut microbiota metabolites is that butyrate strengthens the gut lining, preventing leaky gut and systemic inflammation [88,89].
The immune system is influenced and trained by the gut microbiota. The equilibrium between bacteria and fungus in healthy people guarantees the right immune responses, avoiding infections and over-inflammatory reactions [90]. Regulatory T cells (Tregs), which support immunological tolerance and avoid needless immune activation against innocuous microorganisms and environmental antigens, are developed by commensal bacteria [91]. Although TH17 cells play a crucial role in defense against fungal infections, their activity is strictly controlled in healthy people to avoid excessive inflammation, which may result in autoimmune problems [92]. A healthy immune system promotes general health and infection resistance by lowering the likelihood of allergies, autoimmunity, and chronic inflammatory diseases [93].
Although antibiotics are necessary for the treatment of bacterial illnesses, their usage may unintentionally alter the gut microbiota, which can cause fungal overgrowth in otherwise healthy people [94]. Temporary problems like bloating, diarrhea, or yeast infections may result from this imbalance. Over time, the gut microbiota of healthy people usually recovers from antibiotic-induced disturbances, especially when supportive measures like probiotic consumption and a diet high in fiber are taken [95]. Probiotic and prebiotic use, along with thoughtful antibiotic use, are some preventative measures for healthy individuals [96]. Limiting needless antibiotics helps preserve microbial diversity and resilience while introducing beneficial bacteria and encouraging their growth can assist in restoring equilibrium following an infection or antibiotic use [94,97].
By seeing direct interactions between particular bacterial and fungal species, co-culture experiments enable researchers to pinpoint the mechanisms that maintain microbial balance in healthy people [98]. The intricate ecosystem of the human gut is replicated by sophisticated in vitro models, which shed light on the interactions between bacteria and fungi to preserve health [99]. Studying the effects of particular bacteria and fungi on health in a controlled setting is made possible by germ-free mice, which have no microbiota [99]. Mice that have been colonized with the microbiota of healthy humans can reveal information about how the gut microbiota supports a range of physiological functions, from immune regulation and brain function to digestion and metabolism [100,101,102,103].
By using sequencing methods, the gut microbiome may be thoroughly examined, and patterns of bacterial and fungal diversity linked to health can be found [104].
These techniques provide a better understanding of the function that microbial communities play in preserving health by illuminating how they affect host gene expression and metabolite synthesis [105,106]. By identifying individual differences in microbiota composition, multi-omics techniques can inform individualized dietary and lifestyle recommendations to maximize health [107].

4. Gut Mycobiome Characterization in Diseases

The interaction of fungi, bacteria, the host immune system, and environmental factors influences a wide range of diseases, from metabolic, autoimmune, infectious, and neurological conditions to gastrointestinal disorders [4,108].
The gut mycobiome, which is the fungal component of the gut microbiota, constitutes approximately 0.1% of the total microbiome. Despite its small proportion, it plays a crucial role in maintaining gut homeostasis and is implicated in various pathological conditions [34,109].
Bacterial and fungal populations are dysbiotic in CD and UC, two chronic inflammatory gastrointestinal disorders [110]. Usually tied to skin disorders, Malassezia species have been discovered in CD patients’ guts and are connected to inflammation by interacting with immune receptors, including CARD9 [15,111]. Patients with IBD are frequently dominant in C. albicans. It can exacerbate inflammation by penetrating the mucosal barrier in its hyphal form [112,113,114].
Regarding intestinal fungi, antifungal secretory immunoglobulin A, and the mechanisms of pathogenesis in CD, studies show that disease activity is correlated with decreased fungal diversity and an increased abundance of pathogenic fungi, particularly Candida [115,116,117].
By forming biofilms with bacteria, fungi increase resistance to antimicrobial therapies and the immune system [118]. TH17-mediated inflammation is promoted by fungi’s β-glucans, which activate pattern recognition receptors (PRRs) like Dectin-1 [119].
The symptoms of IBS, a functional gastrointestinal illness, include bloating, changed bowel patterns, and abdominal discomfort [120]. In addition to altered fungal–bacterial ratios that might affect gut motility and immunological responses, studies have shown elevated Candida spp. in IBS patients, which may be a contributing factor to bloating and visceral hypersensitivity [121,122].
The enteric nervous system may be impacted by fungal metabolites, which might lead to symptoms including pain and discomfort [123]. Fungal dysbiosis-induced inflammation may make IBS symptoms worse [124].
Through intricate relationships between bacterial populations and host metabolism, the mycobiome plays a part in the gut microbiome’s growing recognition of its function in metabolic control [4]. Research has indicated that obese people have higher levels of Candida species and Saccharomyces cerevisiae, Mucor, and Rhodotorula, while having a lower variety of helpful fungus [125]. Insulin resistance has been associated with altered fungal profiles, such as increased levels of Candida and lower levels of helpful fungi like Pichia [126]. Due to their ability to trigger the immune system and/or generate toxic metabolites, C. albicans, Aspergillus, and Meyerozyma may be potential pathogens of metabolic disorders [125]. The overgrowth of fungi can damage the gut and cause systemic inflammation, which is a major contributing factor to metabolic syndrome [125]. SCFA synthesis, which is essential for controlling glucose and lipid metabolism, can be interfered with by fungal dysbiosis [127].
The systemic autoimmune illness known as rheumatoid arthritis (RA) is defined by persistent joint inflammation. A typical feature of this autoimmune disease is fungal dysbiosis [128]. C. albicans levels are elevated in RA patients, which may precipitate or worsen autoimmune reactions [129]. It has been noted that commensal fungi such as Saccharomyces spp. have become less diverse [130]. The mechanisms of fungal involvement, in this case, include immune modulation, where fungal components can activate dendritic cells and Th17 pathways, contributing to systemic inflammation, and molecular mimicry, where fungal antigens may resemble host tissues, triggering autoimmune responses [131,132].
Mycobiome changes are seen in multiple sclerosis (MS) or encephalomyelitis, other inflammatory diseases marked by the demyelination of neurons in the central nervous system [133]. Neuroinflammation may be exacerbated by the changed gut microbiomes of MS patients, which show higher levels of Candida and Saccharomyces spp. [134]. Two possible explanations include the gut–brain axis, which may mediate between immune signaling and fungal metabolites that impact neuroinflammatory pathways, and the fungal activation of Th17, which may worsen central nervous system inflammation in MS [135,136,137]. Airway inflammation and systemic allergy reactions may be impacted by fungal dysbiosis in the gut. Increased allergic reactions and asthma exacerbations have been linked to Aspergillus and Candida species [138,139,140]. Immune skewing, in which fungal dysbiosis may enhance Th2-mediated immune responses, thereby contributing to allergic sensitization or the potential for the gut mycobiome to impact lung immunity through the systemic circulation of immune modulators and microbial metabolites via a gut–lung axis, is the mechanism underlying these processes [141].
Opportunistic fungal infections are more common in those with weakened immune systems, such as those receiving chemotherapy or living with HIV [142,143]. Under immunocompromised conditions, opportunistic fungi like Candida may spread from the gut to other parts of the body [143,144]. Interactions between kingdoms can increase virulence and biofilm formation, making treatment more difficult and increasing the risk of recurring infections [145]. Although fungal interactions can affect the severity of the condition, CDI is a serious bacterial infection that is frequently brought on by the use of antibiotics [146,147]. Fungal overgrowth, especially that of Candida spp., can result from antibiotic-induced bacterial depletion, worsening intestinal inflammation and hindering healing. Patients with CDI may benefit from bacterial treatments combined with mycobiome modification [146,148,149,150].
The gut mycobiome may impact mood and mental health through metabolite formation and immunological regulation. Symptoms of schizophrenia, anxiety, and depression have been connected to changes in fungus variety and abundance [151]. By producing cytokines, fungal dysbiosis may exacerbate systemic inflammation, which is linked to depression [151]. Moreover, neurotransmitter production may be impacted by fungal interactions with bacteria that produce SCFAs [152].
The development and progression of CRC are influenced by the gut microbiome, which includes the mycobiome. In addition to fewer helpful fungus, CRC patients have been reported to have higher levels of C. albicans and Malassezia spp. [153,154]. While fungal metabolites can affect the proliferation and death of cancer cells, fungal-induced inflammation may also produce an environment that is conducive to tumorigenesis [155]. Chemotherapy and radiation therapy for cancer can alter the gut microbiota, which can result in fungal overgrowth and related problems. Immunosuppression makes people more vulnerable to invasive fungal infections, which makes cancer treatment results more difficult [156,157]. By controlling fungal dysbiosis using antifungal medications and microbiome modification, therapy results and consequences may be enhanced [158].
The gut mycobiome plays a significant role in liver disease through the gut–liver axis. Studies have shown that patients with primary sclerosing cholangitis, cirrhosis, and alcoholic hepatitis exhibit increased mycobiome diversity, suggesting a link between fungal dysbiosis and liver pathology [159,160,161]. Conversely, kefir has been identified as a potential therapeutic agent for alcoholic fatty liver disease by modulating the gut mycobiome [162]. These findings highlight the therapeutic potential of targeting the gut–liver axis in liver disease management [163]. Similarly, the gut–kidney axis has been implicated in fungal infections. Research in mice with Candida-disseminated infections has shown that the kidneys carry the highest fungal burden, emphasizing their role in systemic fungal circulation. However, the exact contribution of gut mycobiome imbalance to renal disease pathogenesis remains largely unexplored [164]. The interplay between the gut mycobiome and the pancreas has also been observed in metabolic and oncological conditions. Patients with type 1 and type 2 diabetes mellitus exhibit elevated C. albicans levels, suggesting a potential role of fungal dysbiosis in diabetes development [165]. Additionally, increased Malassezia spp. levels have been found in patients with pancreatic ductal adenocarcinoma, indicating a possible gut–pancreas connection via the sphincter of Oddi [14,166]. However, further research is needed to determine whether gut mycobiome dysbiosis is a driving factor in oncogenic progression or a consequence of the disease. Understanding these gut–organ axes may open new avenues for targeted fungal-based therapies in various diseases [163]. The relationship between the gut mycobiome and the main affected organs is illustrated in Figure 3.
A biofilm is a structured microbial community composed of bacteria and fungi encased in a self-produced polymeric matrix that adheres to surfaces. These biofilms can persist, reproduce, and influence human health and disease states [118]. Pathological biofilm formation is characterized by uncontrolled microbial growth, leading to infections and accounting for approximately 70% of microbial infections. These include periodontitis, chronic prostatitis, cystic fibrosis, endocarditis, and otitis media. Additionally, biofilm formation can lead to the contamination of medical devices such as prosthetic heart valves, central venous catheters, contact lenses, intrauterine devices, dental unit waterlines, and urinary catheters [167,168,169,170,171,172]. The pathogenicity of biofilms is driven by bacterial cell detachment, endotoxin production, immune evasion, and the development of a protective barrier [118,173,174]. While bacterial biofilms are well studied, fungal biofilms, particularly those involving Candida spp., play a significant role in infections and antimicrobial resistance. Candida biofilms exhibit high tolerance to antifungal treatments and can form mixed-species biofilms with bacteria, enhancing resistance and persistence [175,176]. Additionally, bacterial biofilms can impact the gut mycobiome, contributing to antimicrobial resistance against antibiotics and disinfectants [177]. However, studies suggest this resistance is lowest during the early biofilm formation stages, where antibiotic treatments and dietary supplements are most effective [178]. Studies have shown that the response to treatment also varies across mycobacterial species due to differences in metabolic states and resistance genes [118,179]. Increased antibiotic use has led to the emergence of resistant fungal biofilms, necessitating novel eradication strategies [175,176]. Approaches include surface modifications to prevent bacterial adhesion, probiotics (E. faecium and Bifidobacterium adolescentis), exopolysaccharide antagonists (e.g., N-acetylcysteine and benzimidazole), peptide-based vaccines, monoclonal antibodies, and mechanical removal methods like ultrasound or thermal shock therapy [118,180,181,182,183]. Given biofilms’ affinity for moist, nutrient-rich surfaces, enhanced preoperative and postoperative precautions and innovative biofilm-targeting therapies are crucial in combating fungal and mixed-species biofilm-associated infections [184].

5. Clinical Perspectives on the Possibilities for Calibrating the Gut Mycobiome

5.1. Antifungal Therapies

The overgrowth of fungi, especially opportunistic species like C. albicans, can lead to inflammation, systemic immunological activation, and gut dysbiosis [185]. Antifungal treatments aim to balance the gut microbiota and lessen fungal overgrowth. Antifungal drugs come in a variety of forms with a range of clinical uses [186]. Fungal cell death results from the azoles (fluconazole, itraconazole, and voriconazole) inhibiting the synthesis of ergosterol, a crucial component of fungal cell membranes [187]. Patients with severe Candida overgrowth or systemic fungal infections are treated with them [188]. For instance, fluconazole has been used to lessen inflammation in patients with fungal dysbiosis who have CD [189]. Azole use has many drawbacks, including the potential to change bacterial populations and exacerbate dysbiosis, as well as the risk of resistance if taken accordingly with the prescription method [190].
Polyenes, which bind to ergosterol and damage fungal cell membranes, are another class of antifungal agents. Examples of these include nystatin and amphotericin B [191,192]. Amphotericin B is used for severe systemic fungal infections but rarely for gut dysbiosis [125]. Nystatin is a non-absorbable antifungal utilized for gastrointestinal Candida overgrowth [193]. Nystatin has been used to treat small intestine fungal overgrowth (SIFO) and IBS to alleviate symptoms in patients suffering from fungal dysbiosis [194]. Conversely, polyenes may induce local irritation and be harsh on the intestinal lining [195,196]. Echinocandins (caspofungin and micafungin) target β-glucans to inhibit the formation of fungal cell walls [197]. They may help immunocompromised people with gut fungal overgrowth and are used for systemic fungal infections but not frequently for gut dysbiosis [198,199]. A study by Puškárová et al. investigating the antibacterial and antifungal properties of six essential oils (oregano, thyme, clove, lavender, clary sage, and arborvitae) found that they effectively inhibit mycelial growth. Their antifungal effects, whether fungicidal or fungistatic, varied depending on the fungal strain (Cladosporium cladosporoides, Alternaria alternata, Aspergillus fumigatus, Chaetomium globosum, and Penicillium chrysogenum). Additionally, the study revealed that the vapor administration of these essential oils was more effective than liquid application [200]. Research indicates that berberine possesses significant antifungal activity, notably against C. albicans. In murine models, berberine administration has been shown to reduce intestinal damage associated with fungal overgrowth and improve the balance of gut microbiota, thereby preventing dysbiosis [201]. An in vitro study conducted by Tonoue et al. examined the antimicrobial effects of Arctostaphylos uva-ursi (A. uva-ursi), a plant commonly used for urinary tract infections, on Aspergillus flavus, Aspergillus fumigatus, Aspergillus niger, C. albicans, and Escherichia coli. The results revealed that A. uva-ursi exhibited antimicrobial activity only against C. albicans and E. coli, with a stronger effect observed when using the herbal extract than the homeopathic mother tincture. However, the authors noted that their findings were inconclusive due to study limitations, including the absence of comparable research, the lack of isolation of the plant’s active ingredients, and variability in the extracts’ sources [202]. Additionally, a review by Gupta et al. emphasized the significant antibiotic properties of A. uva-ursi in treating urinary tract infections, primarily attributed to its glycoside arbutin, which is converted into hydroquinone. The authors highlighted arbutin’s multiple potential effects, including antiseptic, antibiotic, astringent, and diuretic properties, while calling for further research to better understand its therapeutic potential [203]. A study by Shi et al. investigated the effects of undecylenic acid on C. albicans skin infections and found that it serves as an effective antifungal agent. The study demonstrated that undecylenic acid inhibits C. albicans biofilm formation and prevents its transition from yeast to filamentous form, even at low treatment doses [204].
Although using antifungal drugs has many benefits, there are several drawbacks to these treatments as well. Although they lessen the fungal load, antifungals do not always return the microbial equilibrium [186,205]. Among other things, removing fungus without bacterial modulation can result in bacterial overgrowth, change gut function, and select for resistant fungal strains [158,206]. The most crucial takeaway is that fungal overgrowth could return without sustained microbiome support.
In cases of severe fungal dysbiosis, immunocompromised patients, or refractory IBD, IBS, or SIFO, antifungal medication should be evaluated on an individual basis [207,208]. Targeted antifungals that do not harm beneficial fungi should be the subject of future study.

5.2. Probiotics

Some bacteria compete with fungi for nutrition and space and have inherent antifungal qualities. The organic acids produced by Lactobacillus rhamnosus GG inhibit Candida species [209,210]. Reuterin is an antibacterial substance that works well against fungus and is produced by Lactobacillus reuteri [177,211,212]. Candida and Bifidobacterium breve compete for gut cell attachment sites [213,214]. L. rhamnosus supplementation decreased Candida colonization and enhanced gut barrier function in IBD patients [215,216,217]. Probiotics made from yeast such as Saccharomyces boulardii compete with Candida for resources, decreasing the creation of fungal biofilms and increasing the synthesis of IgA (which targets fungal infections); this non-pathogenic yeast stops fungal overgrowth [58,218,219]. It has been demonstrated that S. boulardii lowers the risk of fungal translocation in critically ill patients and is used to treat antibiotic-associated diarrhea [220]. One disadvantage of probiotics is that not all of them have antifungal qualities. Certain probiotics might not withstand stomach acid if they are not properly prepared [120]. Additionally, the overuse of probiotics in immunocompromised people may result in bacterial or fungal overgrowth. Supplementing with probiotics has potential as an adjuvant treatment, especially for fungal-associated dysbiosis, IBD, and IBS [221]. For the best results, strain selection and patient-specific characteristics must be taken into account.

5.3. Fecal Microbiota Transplantation

The goal of FMT is to restore gut microbial diversity, including fungal communities, by transferring stool from a healthy donor into a recipient [222]. Several findings about the function of FMT in mycobiome modulation indicate that it may be a focus for future research with promising outcomes in certain situations [223]. Following antibiotic treatment, FMT has been demonstrated to prevent excessive Candida overgrowth in CDI patients by restoring fungal equilibrium [146]. FMT has been proven to enhance illness outcomes and reduce fungal burden in patients with IBD and Candida-associated dysbiosis [224]. FMT may improve metabolic indicators in patients with metabolic syndrome by assisting in the recalibration of bacterial and fungal communities [225,226].
Fungal composition differs among donors, which causes variable results when it comes to the issues of FMT [227]. Over time, the transplanted mycobiome might change. There is still concern about unintentional pathogen transmission [228].
For gut dysbiosis, including fungal imbalances, FMT is a promising treatment [229]. However, before it can be widely used in conditions linked to fungi, standardization and safety procedures need to be improved.

5.4. Dietary Support

Diet has a major impact on the overall composition of the gut microbiome, which includes fungus [230]. A varied and balanced microbiome is supported in healthy people by diets high in fiber, whole grains, fruits, and vegetables [231]. Dietary fibers encourage the development of good bacteria that generate SCFAs, which support gut health and fungal population control [232]. Berries, green tea, and dark chocolate are examples of foods high in polyphenols that naturally have antifungal qualities that help maintain the delicate balance between bacteria and fungi [233]. Specific food patterns can also support a healthy microbiome [4,234]. The Mediterranean diet, which emphasizes fresh fruits, vegetables, and seafood, is, on the one hand, linked to increased microbial diversity and decreased systemic inflammation [235]. Conversely, a plant-based diet devoid of meat supports immunological and metabolic health by encouraging the diversity of beneficial bacteria and fungi [235]. Metabolic outcomes may be enhanced by dietary therapies that target both bacterial and fungal components of the microbiome [236]. Prebiotics and probiotics that promote fungal equilibrium may help control type 2 diabetes (T2D) and obesity [237].
Through microbial interactions and nutritional availability, diet has a significant impact on the gut microbiome, influencing fungal populations [238]. There are various strategies for dietary intervention to calibrate the mycobiome [239]. Restricting the intake of sugar and processed carbs can be the first step. Consuming too much sugar encourages the growth of fungi, especially Candida [240,241]. Patients with recurring infections have been treated with low-carb or ketogenic diets to lessen their fungal burden [242]. The daily consumption of meals high in fiber can be another strategy [243,244]. Beta-glucans, resistant starch, and inulin are examples of prebiotics that support the diversity of good microorganisms, including antifungal bacteria [79,245]. For instance, the Mediterranean diet, which is customarily high in fiber and polyphenols, promotes a balance of gut fungi and inhibits the growth of Candida [244,246]. Dietary modification is a foundational approach to gut mycobiome regulation, with high-fiber, low-sugar, and polyphenol-rich diets showing the most promise [239].
A comprehensive strategy that incorporates targeted antifungal medication, probiotic supplementation, FMT, and dietary interventions is needed to calibrate the gut mycobiome [34]. Antifungal medications offer short-term respite, but probiotics and dietary changes are the best ways to maintain mycobiome stability over the long run [34]. Personalized mycobiome modification techniques catered to specific medical problems should be the main emphasis of future research.
Since refined carbohydrates and rapidly digestible starches are the main energy sources for fungal development, limiting them is one of the most important dietary strategies for controlling fungal overgrowth [247].
For fungi and opportunistic bacteria, refined carbohydrates and quickly digested starches offer a readily available energy source [248]. In the digestive tract, simple sugars and quickly digested starches—such as those in white rice, pasta, potatoes, and processed grains—are hydrolyzed into glucose very fast [249]. This quick conversion raises blood sugar levels, which might upset the microbial balance and encourage fungal development [250].
Another facet of this subject is that prebiotic fibers are better for beneficial bacteria like Lactobacillus and Bifidobacterium than simple carbohydrates [251]. The overconsumption of carbohydrates can exacerbate intestinal dysbiosis by favoring harmful bacteria and yeast [252].
In addition, these carbohydrates contribute to intestinal disruption. The intestinal barrier integrity may be altered by the overabundance of fungal species, especially C. albicans, which increases the generation of mycotoxins and inflammatory chemicals [253,254]. Often called leaky gut syndrome, this illness is linked to systemic health problems, immunological dysregulation, and persistent inflammation [255].
An additional element of this subject is that prebiotic fibers, as opposed to simple carbohydrates, are what beneficial bacteria like Lactobacillus and Bifidobacterium thrive on [256]. Consuming too many carbohydrates can encourage harmful bacteria and yeast, which exacerbates intestinal dysbiosis [257].
Slow-digesting, fiber-rich carbs that support good gut bacteria and reduce fungal overgrowth should be prioritized over refined carbohydrates and quickly digested starches [257]. Legumes like lentils, chickpeas, and black beans, as well as non-starchy fiber-rich vegetables like broccoli, cauliflower, zucchini, asparagus, leafy greens, and modest amounts of carrots, are some of the suggested substitutes [258]. Other choices include seeds and nuts with prebiotic properties, such as walnuts, flaxseeds, chia seeds, and almonds, or pseudo-cereals with a lower glycemic impact, such as quinoa, buckwheat, and amaranth, when taken in moderation [259,260].
Recently, resistant starch sources that do not quickly raise blood sugar levels are becoming increasingly popular. These include cooked and cooled brown rice (which has a lower glycemic impact than white rice), cooked and cooled potatoes (after refrigeration for 12 to 24 h, as this increases resistant starch content), and unripe (green) bananas, which contain prebiotic fibers that support good gut bacteria [261,262].
Including natural antifungal chemicals in food is one of the complementary methods for antifungal support for the gut flora. These include garlic, which is high in the natural antifungal agent allicin; oregano oil, which contains the antifungal compounds carvacrol and thymol; coconut oil, which contains the antifungal compound caprylic acid; ginger and turmeric, which have anti-inflammatory and antimicrobial properties; and cloves, which contain the antifungal compound eugenol [263,264,265].
Polyphenols, which are abundant in foods like cranberries, blueberries, dark chocolate, and green tea, have been demonstrated to promote good bacteria while preventing the growth of fungi [231,266,267]. While alcohol can harm the gut lining and disturb the microbial balance, dairy products, especially those that include lactose, may encourage the growth of Candida [244,268,269]. To summarize this subchapter, Figure 4 illustrates various methods for calibrating the gut mycobiome.

6. Technological Advances in Mycobiome Research and Future Challenges

Research on the gut mycobiome faces numerous challenges, primarily due to methodological limitations [270]. Culture-dependent techniques, such as biochemical assays, microscopy, and fungal cultivation, remain widely used due to their affordability and cost-effectiveness. Recent advances in metagenomics and the development of non-culture-based methods, such as the polymerase chain reaction (PCR) and high-throughput next-generation sequencing (NGS), have significantly improved our understanding of the gut mycobiome’s composition and function [270]. However, our knowledge remains incomplete due to fungal communities’ high inter- and intra-individual variability [6,9,270].
The targeted amplicon sequencing of the internal transcribed spacer (ITS) regions is now the method of choice for profiling fungal communities. Two main markers are commonly used: ITS1 and ITS2. The ITS1 region offers high variability, allowing for fine-scale resolution at the species level. This advantage is counterbalanced by potential amplification biases, which may skew the observed community composition [271,272]. The ITS2 region, with its more conserved primer binding sites, often yields more consistent amplification across diverse fungal taxa. While ITS2 might provide slightly lower taxonomic resolution compared to ITS1, its consistency is beneficial for cross-study comparisons [273]. Choosing between ITS1 and ITS2 should be guided by the specific objectives of each study, as both have distinct advantages and limitations.
The vast amount of data generated by NGS platforms requires performant bioinformatics tools. One of the key resources in this is the UNITE database, a comprehensive and curated repository of fungal ITS sequences; UNITE improves the accuracy of taxonomic assignments and helps resolve ambiguities, particularly with poorly characterized “dark taxa”. Using tools like UNITE not only improves the reliability of the results but also makes it possible to compare findings across different studies [274,275]. Recent work in clinical mycobiome research further emphasizes that high-quality, curated reference databases like UNITE are indispensable for accurate fungal identification and for advancing diagnostic applications [276].
While the Illuma MiSeq platform has been favored for its high throughput and cot-effectiveness, newer sequencing technologies like PacBio and Oxford Nanopore are gaining attention. These platforms can generate full-length ITS amplicons, which helps in obtaining a more complete and accurate view of fungal communities. Although these technologies face challenges such as higher error rates and cost considerations, ongoing improvements promise to make them highly valuable for future mycobiome research [272]. The benefits of these advanced NGS methods extend well beyond clinical applications. For example, ecological studies have successfully applied these techniques to uncover the diversity of wood-decaying fungi in neotropical Atlantic forests. Such work highlights the versatility of NGS technologies, which are essential for understanding fungal roles in both human health and natural ecosystems [277]. By combining cutting-edge sequencing technologies with comprehensive reference databases like UNITE, researchers are paving the way for more targeted microbiome-based therapies and a deeper understanding of fungal contributions to various biological processes.
Additionally, technical challenges and host-related factors—including age, diet, immunity, and genetics—may contribute to underestimating the gut mycobiome’s significance [270,278]. Although previous reviews have extensively covered fungus–disease associations and interventions, our review distinguishes itself by incorporating recent high-impact studies. A recent landmark study involving a cohort of 12,641 Chinese individuals, using internal transcribed spacer (ITS) sequencing, revealed novel associations between specific fungal taxa and host metabolic health [279]. The findings highlight the role of fungal alpha diversity and 19 mycobiome genera in cardiometabolic diseases, as well as a notable link between Saccharomyces and type 2 diabetes. In parallel, enhanced insights into the metabolic crosstalk between gut bacteria and fungi—reviewed in part by Zhang et al., MacAlpine et al., and Deveau et al.—suggest that fungal dysbiosis may be a causative factor in disease progression rather than a secondary phenomenon [4,42,74]. Furthermore, recent advances in the understanding of immune regulation have identified specific fungal components that shape T-cell responses and modulate inflammation, thereby providing a mechanistic basis for targeted dietary, probiotic, and FMT interventions [41,125]. Future research must clarify the precise role of fungi in disease development—whether fungal dysbiosis is a cause or consequence of illness [4]. For instance, increased C. albicans levels in patients with CDI, IBD, or COVID-19 could either disrupt gut microbial equilibrium (causative factor) or result from antibiotic use (consequence) [12,24,48]. It is also essential to distinguish whether a disease is caused by pathogenic fungi from external sources (e.g., food) or by commensal fungi residing in the gut, as this distinction will directly impact treatment strategies [270]. Further studies must explore how fungal inter- and intra-individual variability affects disease progression and therapeutic responses, paving the way for targeted gut mycobiome-based therapies [24]. Additionally, future research should investigate the interactions between the gut mycobiome and other microbial communities (bacteria, viruses, and archaea) and their influence on host-specific factors such as immunity, ethnicity, and diet [280,281]. Understanding these interactions is crucial, especially given that the gut bacteriome and virome exhibit high variability across individuals [282]. Moreover, the relationship between the gut mycobiome and host metabolic pathways remains an emerging area of interest. Notably, Candida species and the gut–brain axis interplay have been implicated in disease development [283]. Encouragingly, fungal-based therapies show potential; for example, Saccharomyces boulardii supplementation improved intestinal neuromuscular anomalies in mice with IBD, while Candida kefyr administration ameliorated autoimmune encephalomyelitis in a multiple sclerosis model [284,285]. These findings highlight the therapeutic potential of gut mycobiome modulation and underscore the need for further research to harness fungal communities for disease prevention and treatment.

7. Conclusions

The gut mycobiome is an emerging and complex component of the human microbiome, playing a crucial role in maintaining health and contributing to disease pathogenesis. While recent studies have significantly advanced our understanding of its composition, interactions, and clinical relevance, many challenges remain. The balance between fungi and bacteria is essential for gut homeostasis, and disruptions in this equilibrium have been associated with conditions such as IBD, metabolic disorders, and infections. Despite advances in sequencing technologies and metagenomics, research on the gut mycobiome is still in its infancy, hindered by methodological limitations, high inter-individual variability, and the complex interplay between fungi, bacteria, and host factors. Clinical interventions targeting the gut mycobiome, including antifungal therapies, probiotics, FMT, and dietary modifications, show promising potential but require further validation through large-scale, well-controlled studies. Future research should focus on defining the precise role of gut fungi in disease progression, improving diagnostic methods, and developing targeted therapeutic strategies. Additionally, understanding how fungal communities influence host immune responses and metabolic pathways will be crucial for designing novel microbiome-based treatments. As research in this field expands, integrating fungal microbiome data with bacterial and viral microbiota studies will provide a more comprehensive view of human gut health and disease, ultimately paving the way for personalized microbiome-based medicine.

Author Contributions

Conceptualization, M.M. and S.M.C.; methodology, R.-M.E.; software, O.A.R.; validation, M.M., S.M.C. and L.A.P.; formal analysis, R.-M.E.; investigation, L.A.P.; resources, O.A.R.; data curation, S.M.C.; writing—original draft preparation, B.S.G. and M.M.; writing—review and editing, R.-M.E.; visualization, B.S.G.; supervision, S.M.C.; project administration, B.S.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
AhRAryl hydrocarbon receptor
CDCrohn’s disease
CDIClostridium difficile infection
CRCColorectal cancer
FMTFecal microbiota transplantation
IBDInflammatory bowel disease
MSMultiple sclerosis
RARheumatoid arthritis
SCFAsShort-chain fatty acids
SIFOSmall intestine fungal overgrowth
T2DType 2 diabetes
TregsRegulatory T cells
UCUlcerative colitis

References

  1. Thursby, E.; Juge, N. Introduction to the human gut microbiota. Biochem. J. 2017, 474, 1823–1836. [Google Scholar] [CrossRef]
  2. Willis, K.A.; Purvis, J.H.; Myers, E.D.; Aziz, M.M.; Karabayir, I.; Gomes, C.K.; Peters, B.M.; Akbilgic, O.; Talati, A.J.; Pierre, J.F. Fungi form interkingdom microbial communities in the primordial human gut that develop with gestational age. Faseb J. 2019, 33, 12825–12837. [Google Scholar] [CrossRef]
  3. Blaser, M.J.; Devkota, S.; McCoy, K.D.; Relman, D.A.; Yassour, M.; Young, V.B. Lessons learned from the prenatal microbiome controversy. Microbiome 2021, 9, 8. [Google Scholar] [CrossRef] [PubMed]
  4. Zhang, F.; Aschenbrenner, D.; Yoo, J.Y.; Zuo, T. The gut mycobiome in health, disease, and clinical applications in association with the gut bacterial microbiome assembly. Lancet Microbe 2022, 3, e969–e983. [Google Scholar] [CrossRef]
  5. 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.; et al. The gut mycobiome of the Human Microbiome Project healthy cohort. Microbiome 2017, 5, 153. [Google Scholar] [CrossRef]
  6. Sun, Y.; Zuo, T.; Cheung, C.P.; Gu, W.; Wan, Y.; Zhang, F.; Chen, N.; Zhan, H.; Yeoh, Y.K.; Niu, J.; et al. Population-Level Configurations of Gut Mycobiome Across 6 Ethnicities in Urban and Rural China. Gastroenterology 2021, 160, 272–286.e211. [Google Scholar] [CrossRef] [PubMed]
  7. Zhang, L.; Zhan, H.; Xu, W.; Yan, S.; Ng, S.C. The role of gut mycobiome in health and diseases. Ther. Adv. Gastroenterol. 2021, 14, 1–18. [Google Scholar] [CrossRef]
  8. Lam, S.; Zuo, T.; Ho, M.; Chan, F.K.L.; Chan, P.K.S.; Ng, S.C. Review article: Fungal alterations in inflammatory bowel diseases. Aliment. Pharmacol. Ther. 2019, 50, 1159–1171. [Google Scholar] [CrossRef] [PubMed]
  9. Li, X.V.; Leonardi, I.; Putzel, G.G.; Semon, A.; Fiers, W.D.; Kusakabe, T.; Lin, W.Y.; Gao, I.H.; Doron, I.; Gutierrez-Guerrero, A.; et al. Immune regulation by fungal strain diversity in inflammatory bowel disease. Nature 2022, 603, 672–678. [Google Scholar] [CrossRef]
  10. Hoggard, M.; Vesty, A.; Wong, G.; Montgomery, J.M.; Fourie, C.; Douglas, R.G.; Biswas, K.; Taylor, M.W. Characterizing the Human Mycobiota: A Comparison of Small Subunit rRNA, ITS1, ITS2, and Large Subunit rRNA Genomic Targets. Front. Microbiol. 2018, 9, 2208. [Google Scholar] [CrossRef]
  11. Rao, C.; Coyte, K.Z.; Bainter, W.; Geha, R.S.; Martin, C.R.; Rakoff-Nahoum, S. Multi-kingdom ecological drivers of microbiota assembly in preterm infants. Nature 2021, 591, 633–638. [Google Scholar] [CrossRef] [PubMed]
  12. Zuo, T.; Wong, S.H.; Cheung, C.P.; Lam, K.; Lui, R.; Cheung, K.; Zhang, F.; Tang, W.; Ching, J.Y.L.; Wu, J.C.Y.; et al. Gut fungal dysbiosis correlates with reduced efficacy of fecal microbiota transplantation in Clostridium difficile infection. Nat. Commun. 2018, 9, 3663. [Google Scholar] [CrossRef]
  13. Spatz, M.; Richard, M.L. Overview of the Potential Role of Malassezia in Gut Health and Disease. Front. Cell Infect. Microbiol. 2020, 10, 201. [Google Scholar] [CrossRef] [PubMed]
  14. Aykut, B.; Pushalkar, S.; Chen, R.; Li, Q.; Abengozar, R.; Kim, J.I.; Shadaloey, S.A.; Wu, D.; Preiss, P.; Verma, N.; et al. The fungal mycobiome promotes pancreatic oncogenesis via activation of MBL. Nature 2019, 574, 264–267. [Google Scholar] [CrossRef]
  15. Limon, J.J.; Tang, J.; Li, D.; Wolf, A.J.; Michelsen, K.S.; Funari, V.; Gargus, M.; Nguyen, C.; Sharma, P.; Maymi, V.I.; et al. Malassezia Is Associated with Crohn’s Disease and Exacerbates Colitis in Mouse Models. Cell Host Microbe 2019, 25, 377–388.e6. [Google Scholar] [CrossRef]
  16. Kombrink, A.; Tayyrov, A.; Essig, A.; Stöckli, M.; Micheller, S.; Hintze, J.; van Heuvel, Y.; Dürig, N.; Lin, C.W.; Kallio, P.T.; et al. Induction of antibacterial proteins and peptides in the coprophilous mushroom Coprinopsis cinerea in response to bacteria. Isme J. 2019, 13, 588–602. [Google Scholar] [CrossRef] [PubMed]
  17. Fernández de Ullivarri, M.; Arbulu, S.; Garcia-Gutierrez, E.; Cotter, P.D. Antifungal Peptides as Therapeutic Agents. Front. Cell Infect. Microbiol. 2020, 10, 105. [Google Scholar] [CrossRef]
  18. Siavoshi, F.; Heydari, S.; Shafiee, M.; Ahmadi, S.; Saniee, P.; Sarrafnejad, A.; Kolahdoozan, S. Sequestration inside the yeast vacuole may enhance Helicobacter pylori survival against stressful condition. Infect. Genet. Evol. 2019, 69, 127–133. [Google Scholar] [CrossRef]
  19. Jain, U.; Ver Heul, A.M.; Xiong, S.; Gregory, M.H.; Demers, E.G.; Kern, J.T.; Lai, C.W.; Muegge, B.D.; Barisas, D.A.G.; Leal-Ekman, J.S.; et al. Debaryomyces is enriched in Crohn’s disease intestinal tissue and impairs healing in mice. Science 2021, 371, 1154–1159. [Google Scholar] [CrossRef]
  20. Zou, R.; Wang, Y.; Duan, M.; Guo, M.; Zhang, Q.; Zheng, H. Dysbiosis of Gut Fungal Microbiota in Children with Autism Spectrum Disorders. J. Autism Dev. Disord. 2021, 51, 267–275. [Google Scholar] [CrossRef]
  21. Mar Rodríguez, M.; Pérez, D.; Javier Chaves, F.; Esteve, E.; Marin-Garcia, P.; Xifra, G.; Vendrell, J.; Jové, M.; Pamplona, R.; Ricart, W.; et al. Obesity changes the human gut mycobiome. Sci. Rep. 2015, 5, 14600. [Google Scholar] [CrossRef]
  22. Nelson, A.; Stewart, C.J.; Kennedy, N.A.; Lodge, J.K.; Tremelling, M.; Probert, C.S.; Parkes, M.; Mansfield, J.C.; Smith, D.L.; Hold, G.L.; et al. The Impact of NOD2 Genetic Variants on the Gut Mycobiota in Crohn’s Disease Patients in Remission and in Individuals Without Gastrointestinal Inflammation. J. Crohns Colitis 2021, 15, 800–812. [Google Scholar] [CrossRef] [PubMed]
  23. Lang, S.; Duan, Y.; Liu, J.; Torralba, M.G.; Kuelbs, C.; Ventura-Cots, M.; Abraldes, J.G.; Bosques-Padilla, F.; Verna, E.C.; Brown, R.S., Jr.; et al. Intestinal Fungal Dysbiosis and Systemic Immune Response to Fungi in Patients with Alcoholic Hepatitis. Hepatology 2020, 71, 522–538. [Google Scholar] [CrossRef] [PubMed]
  24. Zuo, T.; Zhan, H.; Zhang, F.; Liu, Q.; Tso, E.Y.K.; Lui, G.C.Y.; Chen, N.; Li, A.; Lu, W.; Chan, F.K.L.; et al. Alterations in Fecal Fungal Microbiome of Patients with COVID-19 During Time of Hospitalization until Discharge. Gastroenterology 2020, 159, 1302–1310.e5. [Google Scholar] [CrossRef] [PubMed]
  25. Uryu, H.; Hashimoto, D.; Kato, K.; Hayase, E.; Matsuoka, S.; Ogasawara, R.; Takahashi, S.; Maeda, Y.; Iwasaki, H.; Miyamoto, T.; et al. α-Mannan induces Th17-mediated pulmonary graft-versus-host disease in mice. Blood 2015, 125, 3014–3023. [Google Scholar] [CrossRef]
  26. Bacher, P.; Hohnstein, T.; Beerbaum, E.; Röcker, M.; Blango, M.G.; Kaufmann, S.; Röhmel, J.; Eschenhagen, P.; Grehn, C.; Seidel, K.; et al. Human Anti-fungal Th17 Immunity and Pathology Rely on Cross-Reactivity against Candida albicans. Cell 2019, 176, 1340–1355.e15. [Google Scholar] [CrossRef]
  27. Gunsalus, K.T.; Tornberg-Belanger, S.N.; Matthan, N.R.; Lichtenstein, A.H.; Kumamoto, C.A. Manipulation of Host Diet To Reduce Gastrointestinal Colonization by the Opportunistic Pathogen Candida albicans. mSphere 2015, 1, e00020-15. [Google Scholar] [CrossRef]
  28. Lewis, J.D.; Chen, E.Z.; Baldassano, R.N.; Otley, A.R.; Griffiths, A.M.; Lee, D.; Bittinger, K.; Bailey, A.; Friedman, E.S.; Hoffmann, C.; et al. Inflammation, Antibiotics, and Diet as Environmental Stressors of the Gut Microbiome in Pediatric Crohn’s Disease. Cell Host Microbe 2015, 18, 489–500. [Google Scholar] [CrossRef]
  29. van Tilburg Bernardes, E.; Pettersen, V.K.; Gutierrez, M.W.; Laforest-Lapointe, I.; Jendzjowsky, N.G.; Cavin, J.-B.; Vicentini, F.A.; Keenan, C.M.; Ramay, H.R.; Samara, J.; et al. Intestinal fungi are causally implicated in microbiome assembly and immune development in mice. Nat. Commun. 2020, 11, 2577. [Google Scholar] [CrossRef]
  30. Seelbinder, B.; Chen, J.; Brunke, S.; Vazquez-Uribe, R.; Santhaman, R.; Meyer, A.C.; de Oliveira Lino, F.S.; Chan, K.F.; Loos, D.; Imamovic, L.; et al. Antibiotics create a shift from mutualism to competition in human gut communities with a longer-lasting impact on fungi than bacteria. Microbiome 2020, 8, 133. [Google Scholar] [CrossRef] [PubMed]
  31. Leonardi, I.; Paramsothy, S.; Doron, I.; Semon, A.; Kaakoush, N.O.; Clemente, J.C.; Faith, J.J.; Borody, T.J.; Mitchell, H.M.; Colombel, J.F.; et al. Fungal Trans-kingdom Dynamics Linked to Responsiveness to Fecal Microbiota Transplantation (FMT) Therapy in Ulcerative Colitis. Cell Host Microbe 2020, 27, 823–829.e3. [Google Scholar] [CrossRef]
  32. Stewart, D.B., Sr.; Wright, J.R.; Fowler, M.; McLimans, C.J.; Tokarev, V.; Amaniera, I.; Baker, O.; Wong, H.T.; Brabec, J.; Drucker, R.; et al. Integrated Meta-omics Reveals a Fungus-Associated Bacteriome and Distinct Functional Pathways in Clostridioides difficile Infection. mSphere 2019, 4, e00454-19. [Google Scholar] [CrossRef] [PubMed]
  33. Doron, I.; Leonardi, I.; Li, X.V.; Fiers, W.D.; Semon, A.; Bialt-DeCelie, M.; Migaud, M.; Gao, I.H.; Lin, W.Y.; Kusakabe, T.; et al. Human gut mycobiota tune immunity via CARD9-dependent induction of anti-fungal IgG antibodies. Cell 2021, 184, 1017–1031.e14. [Google Scholar] [CrossRef] [PubMed]
  34. Huang, H.; Wang, Q.; Yang, Y.; Zhong, W.; He, F.; Li, J. The mycobiome as integral part of the gut microbiome: Crucial role of symbiotic fungi in health and disease. Gut Microbes 2024, 16, 2440111. [Google Scholar] [CrossRef]
  35. Mok, K.; Poolsawat, T.; Somnuk, S.; Wanikorn, B.; Patumcharoenpol, P.; Nitisinprasert, S.; Vongsangnak, W.; Nakphaichit, M. Preliminary characterization of gut mycobiome enterotypes reveals the correlation trends between host metabolic parameter and diet: A case study in the Thai Cohort. Sci. Rep. 2024, 14, 5805. [Google Scholar] [CrossRef]
  36. Auchtung, T.A.; Stewart, C.J.; Smith, D.P.; Triplett, E.W.; Agardh, D.; Hagopian, W.A.; Ziegler, A.G.; Rewers, M.J.; She, J.X.; Toppari, J.; et al. Temporal changes in gastrointestinal fungi and the risk of autoimmunity during early childhood: The TEDDY study. Nat. Commun. 2022, 13, 3151. [Google Scholar] [CrossRef] [PubMed]
  37. Fujimura, K.E.; Sitarik, A.R.; Havstad, S.; Lin, D.L.; Levan, S.; Fadrosh, D.; Panzer, A.R.; LaMere, B.; Rackaityte, E.; Lukacs, N.W.; et al. Neonatal gut microbiota associates with childhood multisensitized atopy and T cell differentiation. Nat. Med. 2016, 22, 1187–1191. [Google Scholar] [CrossRef]
  38. Schei, K.; Avershina, E.; Øien, T.; Rudi, K.; Follestad, T.; Salamati, S.; Ødegård, R.A. Early gut mycobiota and mother-offspring transfer. Microbiome 2017, 5, 107. [Google Scholar] [CrossRef]
  39. Hallen-Adams, H.E.; Kachman, S.D.; Kim, J.; Legge, R.M.; Martínez, I. Fungi inhabiting the healthy human gastrointestinal tract: A diverse and dynamic community. Fungal Ecol. 2015, 15, 9–17. [Google Scholar] [CrossRef]
  40. Wu, L.; Zeng, T.; Deligios, M.; Milanesi, L.; Langille, M.G.I.; Zinellu, A.; Rubino, S.; Carru, C.; Kelvin, D.J. Age-Related Variation of Bacterial and Fungal Communities in Different Body Habitats across the Young, Elderly, and Centenarians in Sardinia. mSphere 2020, 5, e00558-19. [Google Scholar] [CrossRef]
  41. Iliev, I.D.; Leonardi, I. Fungal dysbiosis: Immunity and interactions at mucosal barriers. Nat. Rev. Immunol. 2017, 17, 635–646. [Google Scholar] [CrossRef] [PubMed]
  42. Deveau, A.; Bonito, G.; Uehling, J.; Paoletti, M.; Becker, M.; Bindschedler, S.; Hacquard, S.; Hervé, V.; Labbé, J.; Lastovetsky, O.A.; et al. Bacterial-fungal interactions: Ecology, mechanisms and challenges. FEMS Microbiol. Rev. 2018, 42, 335–352. [Google Scholar] [CrossRef]
  43. Hill, J.H.; Round, J.L. Intestinal fungal-host interactions in promoting and maintaining health. Cell Host Microbe 2024, 32, 1668–1680. [Google Scholar] [CrossRef] [PubMed]
  44. Brown, G.D.; Denning, D.W.; Gow, N.A.; Levitz, S.M.; Netea, M.G.; White, T.C. Hidden killers: Human fungal infections. Sci. Transl. Med. 2012, 4, 165rv113. [Google Scholar] [CrossRef]
  45. Fan, D.; Coughlin, L.A.; Neubauer, M.M.; Kim, J.; Kim, M.S.; Zhan, X.; Simms-Waldrip, T.R.; Xie, Y.; Hooper, L.V.; Koh, A.Y. Activation of HIF-1α and LL-37 by commensal bacteria inhibits Candida albicans colonization. Nat. Med. 2015, 21, 808–814. [Google Scholar] [CrossRef]
  46. Pfaller, M.A.; Diekema, D.J.; Turnidge, J.D.; Castanheira, M.; Jones, R.N. Twenty Years of the SENTRY Antifungal Surveillance Program: Results for Candida Species from 1997–2016. Open Forum Infect. Dis. 2019, 6, S79–S94. [Google Scholar] [CrossRef]
  47. Li, H.; Miao, M.X.; Jia, C.L.; Cao, Y.B.; Yan, T.H.; Jiang, Y.Y.; Yang, F. Interactions between Candida albicans and the resident microbiota. Front. Microbiol. 2022, 13, 930495. [Google Scholar] [CrossRef] [PubMed]
  48. Sokol, H.; Leducq, V.; Aschard, H.; Pham, H.P.; Jegou, S.; Landman, C.; Cohen, D.; Liguori, G.; Bourrier, A.; Nion-Larmurier, I.; et al. Fungal microbiota dysbiosis in IBD. Gut 2017, 66, 1039–1048. [Google Scholar] [CrossRef]
  49. Lopes, J.P.; Lionakis, M.S. Pathogenesis and virulence of Candida albicans. Virulence 2022, 13, 89–121. [Google Scholar] [CrossRef]
  50. Kumamoto, C.A.; Gresnigt, M.S.; Hube, B. The gut, the bad and the harmless: Candida albicans as a commensal and opportunistic pathogen in the intestine. Curr. Opin. Microbiol. 2020, 56, 7–15. [Google Scholar] [CrossRef]
  51. Charlet, R.; Pruvost, Y.; Tumba, G.; Istel, F.; Poulain, D.; Kuchler, K.; Sendid, B.; Jawhara, S. Remodeling of the Candida glabrata cell wall in the gastrointestinal tract affects the gut microbiota and the immune response. Sci. Rep. 2018, 8, 3316. [Google Scholar] [CrossRef] [PubMed]
  52. Di Martino, L.; De Salvo, C.; Buela, K.A.; Hager, C.; Ghannoum, M.; Osme, A.; Buttò, L.; Bamias, G.; Pizarro, T.T.; Cominelli, F. Candida tropicalis Infection Modulates the Gut Microbiome and Confers Enhanced Susceptibility to Colitis in Mice. Cell Mol. Gastroenterol. Hepatol. 2022, 13, 901–923. [Google Scholar] [CrossRef]
  53. Hoarau, G.; Mukherjee, P.K.; Gower-Rousseau, C.; Hager, C.; Chandra, J.; Retuerto, M.A.; Neut, C.; Vermeire, S.; Clemente, J.; Colombel, J.F.; et al. Bacteriome and Mycobiome Interactions Underscore Microbial Dysbiosis in Familial Crohn’s Disease. mBio 2016, 7, e01250-16. [Google Scholar] [CrossRef]
  54. Sun, S.; Sun, L.; Wang, K.; Qiao, S.; Zhao, X.; Hu, X.; Chen, W.; Zhang, S.; Li, H.; Dai, H.; et al. The gut commensal fungus, Candida parapsilosis, promotes high fat-diet induced obesity in mice. Commun. Biol. 2021, 4, 1220. [Google Scholar] [CrossRef] [PubMed]
  55. Cooper, C.R. Chapter 2—Yeasts Pathogenic to Humans. In The Yeasts, 5th ed.; Kurtzman, C.P., Fell, J.W., Boekhout, T., Eds.; Elsevier: London, UK, 2011; pp. 9–19. [Google Scholar] [CrossRef]
  56. Noor-Ul, H.; Liu, H.; Jin, J.; Zhu, X.; Han, D.; Yang, Y.; Xie, S. Dietary supplementation of Geotrichum candidum improves growth, gut microbiota, immune-related gene expression and disease resistance in gibel carp CAS III (Carassius auratus gibelio). Fish Shellfish Immunol. 2020, 99, 144–153. [Google Scholar] [CrossRef]
  57. Farid, F.; Sideeq, O.; Khan, F.; Niaz, K. Chapter 5.1—Saccharomyces cerevisiae. In Nonvitamin and Nonmineral Nutritional Supplements; Nabavi, S.M., Silva, A.S., Eds.; Academic Press: Cambridge, MA, USA, 2019; pp. 501–508. [Google Scholar] [CrossRef]
  58. Abid, R.; Waseem, H.; Ali, J.; Ghazanfar, S.; Muhammad Ali, G.; Elasbali, A.M.; Alharethi, S.H. Probiotic Yeast Saccharomyces: Back to Nature to Improve Human Health. J. Fungi 2022, 8, 444. [Google Scholar] [CrossRef]
  59. Iancu, M.A.; Profir, M.; Roşu, O.A.; Ionescu, R.F.; Cretoiu, S.M.; Gaspar, B.S. Revisiting the Intestinal Microbiome and Its Role in Diarrhea and Constipation. Microorganisms 2023, 11, 2177. [Google Scholar] [CrossRef] [PubMed]
  60. Chen, X.; Fruehauf, J.; Goldsmith, J.D.; Xu, H.; Katchar, K.K.; Koon, H.W.; Zhao, D.; Kokkotou, E.G.; Pothoulakis, C.; Kelly, C.P. Saccharomyces boulardii inhibits EGF receptor signaling and intestinal tumor growth in Apc(min) mice. Gastroenterology 2009, 137, 914–923. [Google Scholar] [CrossRef]
  61. Chen, Y.; Chen, Z.; Guo, R.; Chen, N.; Lu, H.; Huang, S.; Wang, J.; Li, L. Correlation between gastrointestinal fungi and varying degrees of chronic hepatitis B virus infection. Diagn. Microbiol. Infect. Dis. 2011, 70, 492–498. [Google Scholar] [CrossRef]
  62. Hamad, I.; Sokhna, C.; Raoult, D.; Bittar, F. Molecular detection of eukaryotes in a single human stool sample from Senegal. PLoS ONE 2012, 7, e40888. [Google Scholar] [CrossRef]
  63. Gouba, N.; Raoult, D.; Drancourt, M. Plant and fungal diversity in gut microbiota as revealed by molecular and culture investigations. PLoS ONE 2013, 8, e59474. [Google Scholar] [CrossRef]
  64. Hallen-Adams, H.E.; Suhr, M.J. Fungi in the healthy human gastrointestinal tract. Virulence 2017, 8, 352–358. [Google Scholar] [CrossRef]
  65. 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] [PubMed]
  66. 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]
  67. Profir, M.; Roşu, O.A.; Creţoiu, S.M.; Gaspar, B.S. Friend or Foe: Exploring the Relationship between the Gut Microbiota and the Pathogenesis and Treatment of Digestive Cancers. Microorganisms 2024, 12, 955. [Google Scholar] [CrossRef] [PubMed]
  68. Yang, Q.; Ouyang, J.; Pi, D.; Feng, L.; Yang, J. Malassezia in Inflammatory Bowel Disease: Accomplice of Evoking Tumorigenesis. Front. Immunol. 2022, 13, 846469. [Google Scholar] [CrossRef] [PubMed]
  69. Gaitanis, G.; Velegraki, A.; Magiatis, P.; Pappas, P.; Bassukas, I.D. 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] [PubMed]
  70. Aflatoxins. 3 July 2024. Available online: https://www.cancer.gov/about-cancer/causes-prevention/risk/substances/aflatoxins (accessed on 23 February 2025).
  71. Norlia, M.; Jinap, S.; Nor-Khaizura, M.A.R.; Radu, S.; Samsudin, N.I.P.; Azri, F.A. Aspergillus section Flavi and Aflatoxins: Occurrence, Detection, and Identification in Raw Peanuts and Peanut-Based Products Along the Supply Chain. Front. Microbiol. 2019, 10, 2602. [Google Scholar] [CrossRef]
  72. Malir, F.; Ostry, V.; Pfohl-Leszkowicz, A.; Malir, J.; Toman, J. Ochratoxin A: 50 Years of Research. Toxins 2016, 8, 191. [Google Scholar] [CrossRef]
  73. Maas, E.; Penders, J.; Venema, K. Fungal-Bacterial Interactions in the Human Gut of Healthy Individuals. J. Fungi 2023, 9, 139. [Google Scholar] [CrossRef]
  74. MacAlpine, J.; Robbins, N.; Cowen, L.E. Bacterial-fungal interactions and their impact on microbial pathogenesis. Mol. Ecol. 2023, 32, 2565–2581. [Google Scholar] [CrossRef] [PubMed]
  75. Pawlowska, T.E. Symbioses between fungi and bacteria: From mechanisms to impacts on biodiversity. Curr. Opin. Microbiol. 2024, 80, 102496. [Google Scholar] [CrossRef] [PubMed]
  76. Wang, F.; Wang, Z.; Tang, J. The interactions of Candida albicans with gut bacteria: A new strategy to prevent and treat invasive intestinal candidiasis. Gut Pathog. 2023, 15, 30. [Google Scholar] [CrossRef]
  77. Gill, P.A.; Inniss, S.; Kumagai, T.; Rahman, F.Z.; Smith, A.M. The Role of Diet and Gut Microbiota in Regulating Gastrointestinal and Inflammatory Disease. Front. Immunol. 2022, 13, 866059. [Google Scholar] [CrossRef]
  78. Belkaid, Y.; Hand, T.W. Role of the microbiota in immunity and inflammation. Cell 2014, 157, 121–141. [Google Scholar] [CrossRef]
  79. Victoria Obayomi, O.; Folakemi Olaniran, A.; Olugbemiga Owa, S. Unveiling the role of functional foods with emphasis on prebiotics and probiotics in human health: A review. J. Funct. Foods 2024, 119, 106337. [Google Scholar] [CrossRef]
  80. Facchin, S.; Bertin, L.; Bonazzi, E.; Lorenzon, G.; De Barba, C.; Barberio, B.; Zingone, F.; Maniero, D.; Scarpa, M.; Ruffolo, C.; et al. Short-Chain Fatty Acids and Human Health: From Metabolic Pathways to Current Therapeutic Implications. Life 2024, 14, 559. [Google Scholar] [CrossRef]
  81. Liu, L.; Li, Q.; Yang, Y.; Guo, A. Biological Function of Short-Chain Fatty Acids and Its Regulation on Intestinal Health of Poultry. Front. Vet. Sci. 2021, 8, 736739. [Google Scholar] [CrossRef]
  82. Xiong, R.G.; Zhou, D.D.; Wu, S.X.; Huang, S.Y.; Saimaiti, A.; Yang, Z.J.; Shang, A.; Zhao, C.N.; Gan, R.Y.; Li, H.B. Health Benefits and Side Effects of Short-Chain Fatty Acids. Foods 2022, 11, 2863. [Google Scholar] [CrossRef]
  83. Ranjbar, R.; Vahdati, S.N.; Tavakoli, S.; Khodaie, R.; Behboudi, H. Immunomodulatory roles of microbiota-derived short-chain fatty acids in bacterial infections. Biomed. Pharmacother. 2021, 141, 111817. [Google Scholar] [CrossRef]
  84. He, J.; Zhang, P.; Shen, L.; Niu, L.; Tan, Y.; Chen, L.; Zhao, Y.; Bai, L.; Hao, X.; Li, X.; et al. Short-Chain Fatty Acids and Their Association with Signalling Pathways in Inflammation, Glucose and Lipid Metabolism. Int. J. Mol. Sci. 2020, 21, 6356. [Google Scholar] [CrossRef] [PubMed]
  85. Enache, R.M.; Profir, M.; Roşu, O.A.; Creţoiu, S.M.; Gaspar, B.S. The Role of Gut Microbiota in the Onset and Progression of Obesity and Associated Comorbidities. Int. J. Mol. Sci. 2024, 25, 12321. [Google Scholar] [CrossRef] [PubMed]
  86. Appleton, J. The Gut-Brain Axis: Influence of Microbiota on Mood and Mental Health. Integr. Med. (Encinitas) 2018, 17, 28–32. [Google Scholar] [PubMed]
  87. Fekete, M.; Lehoczki, A.; Major, D.; Fazekas-Pongor, V.; Csípő, T.; Tarantini, S.; Csizmadia, Z.; Varga, J.T. Exploring the Influence of Gut-Brain Axis Modulation on Cognitive Health: A Comprehensive Review of Prebiotics, Probiotics, and Symbiotics. Nutrients 2024, 16, 789. [Google Scholar] [CrossRef]
  88. Canani, R.B.; Costanzo, M.D.; Leone, L.; Pedata, M.; Meli, R.; Calignano, A. Potential beneficial effects of butyrate in intestinal and extraintestinal diseases. World J. Gastroenterol. 2011, 17, 1519–1528. [Google Scholar] [CrossRef]
  89. Hodgkinson, K.; El Abbar, F.; Dobranowski, P.; Manoogian, J.; Butcher, J.; Figeys, D.; Mack, D.; Stintzi, A. Butyrate’s role in human health and the current progress towards its clinical application to treat gastrointestinal disease. Clin. Nutr. 2023, 42, 61–75. [Google Scholar] [CrossRef]
  90. Wiertsema, S.P.; van Bergenhenegouwen, J.; Garssen, J.; Knippels, L.M.J. The Interplay between the Gut Microbiome and the Immune System in the Context of Infectious Diseases throughout Life and the Role of Nutrition in Optimizing Treatment Strategies. Nutrients 2021, 13, 886. [Google Scholar] [CrossRef]
  91. Nutsch, K.M.; Hsieh, C.S. T cell tolerance and immunity to commensal bacteria. Curr. Opin. Immunol. 2012, 24, 385–391. [Google Scholar] [CrossRef]
  92. Zambrano-Zaragoza, J.F.; Romo-Martínez, E.J.; Durán-Avelar Mde, J.; García-Magallanes, N.; Vibanco-Pérez, N. Th17 cells in autoimmune and infectious diseases. Int. J. Inflam. 2014, 2014, 651503. [Google Scholar] [CrossRef]
  93. Iddir, M.; Brito, A.; Dingeo, G.; Fernandez Del Campo, S.S.; Samouda, H.; La Frano, M.R.; Bohn, T. Strengthening the Immune System and Reducing Inflammation and Oxidative Stress through Diet and Nutrition: Considerations during the COVID-19 Crisis. Nutrients 2020, 12, 1562. [Google Scholar] [CrossRef]
  94. Ramirez, J.; Guarner, F.; Bustos Fernandez, L.; Maruy, A.; Sdepanian, V.L.; Cohen, H. Antibiotics as Major Disruptors of Gut Microbiota. Front. Cell Infect. Microbiol. 2020, 10, 572912. [Google Scholar] [CrossRef] [PubMed]
  95. Skoufou, M.; Tsigalou, C.; Vradelis, S.; Bezirtzoglou, E. The Networked Interaction between Probiotics and Intestine in Health and Disease: A Promising Success Story. Microorganisms 2024, 12, 194. [Google Scholar] [CrossRef] [PubMed]
  96. Dahiya, D.; Nigam, P.S. Antibiotic-Therapy-Induced Gut Dysbiosis Affecting Gut Microbiota-Brain Axis and Cognition: Restoration by Intake of Probiotics and Synbiotics. Int. J. Mol. Sci. 2023, 24, 3074. [Google Scholar] [CrossRef] [PubMed]
  97. Patangia, D.V.; Anthony Ryan, C.; Dempsey, E.; Paul Ross, R.; Stanton, C. Impact of antibiotics on the human microbiome and consequences for host health. Microbiologyopen 2022, 11, e1260. [Google Scholar] [CrossRef]
  98. Selegato, D.M.; Castro-Gamboa, I. Enhancing chemical and biological diversity by co-cultivation. Front. Microbiol. 2023, 14, 1117559. [Google Scholar] [CrossRef]
  99. Maas, E.; Penders, J.; Venema, K. Studying Fungal-Bacterial Relationships in the Human Gut Using an In Vitro Model (TIM-2). J. Fungi 2023, 9, 174. [Google Scholar] [CrossRef]
  100. Chung, H.; Pamp, S.J.; Hill, J.A.; Surana, N.K.; Edelman, S.M.; Troy, E.B.; Reading, N.C.; Villablanca, E.J.; Wang, S.; Mora, J.R.; et al. Gut immune maturation depends on colonization with a host-specific microbiota. Cell 2012, 149, 1578–1593. [Google Scholar] [CrossRef]
  101. Lundberg, R.; Toft, M.F.; Metzdorff, S.B.; Hansen, C.H.F.; Licht, T.R.; Bahl, M.I.; Hansen, A.K. Human microbiota-transplanted C57BL/6 mice and offspring display reduced establishment of key bacteria and reduced immune stimulation compared to mouse microbiota-transplantation. Sci. Rep. 2020, 10, 7805. [Google Scholar] [CrossRef]
  102. Arrieta, M.-C.; Walter, J.; Finlay, B.B. Human Microbiota-Associated Mice: A Model with Challenges. Cell Host Microbe 2016, 19, 575–578. [Google Scholar] [CrossRef]
  103. Moreno-Indias, I.; Lundberg, R.; Krych, L.; Metzdorff, S.B.; Kot, W.; Sørensen, D.B.; Nielsen, D.S.; Hansen, C.H.F.; Hansen, A.K. A Humanized Diet Profile May Facilitate Colonization and Immune Stimulation in Human Microbiota-Colonized Mice. Front. Microbiol. 2020, 11, 1336. [Google Scholar] [CrossRef]
  104. Fox, J.D.; Sims, A.; Ross, M.; Bettag, J.; Wilder, A.; Natrop, D.; Borsotti, A.; Kolli, S.; Mehta, S.; Verma, H.; et al. Bioinformatic Methodologies in Assessing Gut Microbiota. Microbiol. Res. 2024, 15, 2554–2574. [Google Scholar] [CrossRef]
  105. Hemmati, M.A.; Monemi, M.; Asli, S.; Mohammadi, S.; Foroozanmehr, B.; Haghmorad, D.; Oksenych, V.; Eslami, M. Using New Technologies to Analyze Gut Microbiota and Predict Cancer Risk. Cells 2024, 13, 1987. [Google Scholar] [CrossRef]
  106. Zhang, Y.; Chen, R.; Zhang, D.; Qi, S.; Liu, Y. Metabolite interactions between host and microbiota during health and disease: Which feeds the other? Biomed. Pharmacother. 2023, 160, 114295. [Google Scholar] [CrossRef] [PubMed]
  107. Chetty, A.; Blekhman, R. Multi-omic approaches for host-microbiome data integration. Gut Microbes 2024, 16, 2297860. [Google Scholar] [CrossRef] [PubMed]
  108. Krüger, W.; Vielreicher, S.; Kapitan, M.; Jacobsen, I.D.; Niemiec, M.J. Fungal-Bacterial Interactions in Health and Disease. Pathogens 2019, 8, 70. [Google Scholar] [CrossRef]
  109. Liu, H.-Y.; Li, S.; Ogamune, K.J.; Ahmed, A.A.; Kim, I.H.; Zhang, Y.; Cai, D. Fungi in the Gut Microbiota: Interactions, Homeostasis, and Host Physiology. Microorganisms 2025, 13, 70. [Google Scholar] [CrossRef]
  110. Santana, P.T.; Rosas, S.L.B.; Ribeiro, B.E.; Marinho, Y.; de Souza, H.S.P. Dysbiosis in Inflammatory Bowel Disease: Pathogenic Role and Potential Therapeutic Targets. Int. J. Mol. Sci. 2022, 23, 3464. [Google Scholar] [CrossRef]
  111. Ashbee, H.R.; Evans, E.G. Immunology of diseases associated with Malassezia species. Clin. Microbiol. Rev. 2002, 15, 21–57. [Google Scholar] [CrossRef]
  112. Talapko, J.; Juzbašić, M.; Matijević, T.; Pustijanac, E.; Bekić, S.; Kotris, I.; Škrlec, I. Candida albicans-The Virulence Factors and Clinical Manifestations of Infection. J. Fungi 2021, 7, 79. [Google Scholar] [CrossRef]
  113. Carlson, S.L.; Mathew, L.; Savage, M.; Kok, K.; Lindsay, J.O.; Munro, C.A.; McCarthy, N.E. Mucosal Immunity to Gut Fungi in Health and Inflammatory Bowel Disease. J. Fungi 2023, 9, 1105. [Google Scholar] [CrossRef]
  114. Sun, M.; Ju, J.; Xu, H.; Wang, Y. Intestinal fungi and antifungal secretory immunoglobulin A in Crohn’s disease. Front. Immunol. 2023, 14, 1177504. [Google Scholar] [CrossRef] [PubMed]
  115. Patnaik, S.; Durairajan, S.S.K.; Singh, A.K.; Krishnamoorthi, S.; Iyaswamy, A.; Mandavi, S.P.; Jeewon, R.; Williams, L.L. Role of Candida species in pathogenesis, immune regulation, and prognostic tools for managing ulcerative colitis and Crohn’s disease. World J. Gastroenterol. 2024, 30, 5212–5220. [Google Scholar] [CrossRef] [PubMed]
  116. Yiallouris, A.; Pana, Z.D.; Marangos, G.; Tzyrka, I.; Karanasios, S.; Georgiou, I.; Kontopyrgia, K.; Triantafyllou, E.; Seidel, D.; Cornely, O.A.; et al. Fungal diversity in the soil Mycobiome: Implications for ONE health. One Health 2024, 18, 100720. [Google Scholar] [CrossRef] [PubMed]
  117. d’Enfert, C.; Kaune, A.K.; Alaban, L.R.; Chakraborty, S.; Cole, N.; Delavy, M.; Kosmala, D.; Marsaux, B.; Fróis-Martins, R.; Morelli, M.; et al. The impact of the Fungus-Host-Microbiota interplay upon Candida albicans infections: Current knowledge and new perspectives. FEMS Microbiol. Rev. 2021, 45, fuaa060. [Google Scholar] [CrossRef]
  118. Sharma, S.; Mohler, J.; Mahajan, S.D.; Schwartz, S.A.; Bruggemann, L.; Aalinkeel, R. Microbial Biofilm: A Review on Formation, Infection, Antibiotic Resistance, Control Measures, and Innovative Treatment. Microorganisms 2023, 11, 1614. [Google Scholar] [CrossRef]
  119. Liu, H.Y.; Prentice, E.L.; Webber, M.A. Mechanisms of antimicrobial resistance in biofilms. NPJ Antimicrob. Resist. 2024, 2, 27. [Google Scholar] [CrossRef]
  120. Profir, M.; Roşu, O.A.; Ionescu, R.F.; Pavelescu, L.A.; Cretoiu, S.M. Chapter 11—Benefits and safety of probiotics in gastrointestinal diseases. In Antidotes to Toxins and Drugs; Găman, M.-A., Egbuna, C., Eds.; Elsevier: Amsterdam, The Netherlands, 2024; pp. 279–328. [Google Scholar] [CrossRef]
  121. Kreulen, I.A.M.; de Jonge, W.J.; van den Wijngaard, R.M.; van Thiel, I.A.M. Candida spp. in Human Intestinal Health and Disease: More than a Gut Feeling. Mycopathologia 2023, 188, 845–862. [Google Scholar] [CrossRef]
  122. Balderramo, D.C.; Romagnoli, P.A.; Granlund, A.V.B.; Catalan-Serra, I. Fecal Fungal Microbiota (Mycobiome) Study as a Potential Tool for Precision Medicine in Inflammatory Bowel Disease. Gut Liver 2023, 17, 505–515. [Google Scholar] [CrossRef]
  123. Lagomarsino, V.N.; Kostic, A.D.; Chiu, I.M. Mechanisms of microbial-neuronal interactions in pain and nociception. Neurobiol. Pain. 2021, 9, 100056. [Google Scholar] [CrossRef]
  124. Botschuijver, S.; Roeselers, G.; Levin, E.; Jonkers, D.M.; Welting, O.; Heinsbroek, S.E.M.; de Weerd, H.H.; Boekhout, T.; Fornai, M.; Masclee, A.A.; et al. Intestinal Fungal Dysbiosis Is Associated with Visceral Hypersensitivity in Patients with Irritable Bowel Syndrome and Rats. Gastroenterology 2017, 153, 1026–1039. [Google Scholar] [CrossRef]
  125. Wang, L.; Zhang, K.; Zeng, Y.; Luo, Y.; Peng, J.; Zhang, J.; Kuang, T.; Fan, G. Gut mycobiome and metabolic diseases: The known, the unknown, and the future. Pharmacol. Res. 2023, 193, 106807. [Google Scholar] [CrossRef]
  126. Nikolic, D.M.; Dimitrijevic-Sreckovic, V.; Ranin, L.T.; Stojanovic, M.M.; Ilic, I.D.; Gostiljac, D.M.; Soldatovic, I.A. Homeostatic microbiome disruption as a cause of insulin secretion disorders Candida albicans, a new factor in pathogenesis of diabetes: A STROBE compliant cross-sectional study. Medicine 2022, 101, e31291. [Google Scholar] [CrossRef] [PubMed]
  127. Machado, M.G.; Sencio, V.; Trottein, F. Short-Chain Fatty Acids as a Potential Treatment for Infections: A Closer Look at the Lungs. Infect. Immun. 2021, 89, e0018821. [Google Scholar] [CrossRef] [PubMed]
  128. Chauhan, K.; Jandu, J.S.; Brent, L.H.; Al-Dhahir, M.A. Rheumatoid Arthritis. Available online: https://www.ncbi.nlm.nih.gov/books/NBK441999/ (accessed on 3 March 2025).
  129. Bishu, S.; Su, E.W.; Wilkerson, E.R.; Reckley, K.A.; Jones, D.M.; McGeachy, M.J.; Gaffen, S.L.; Levesque, M.C. Rheumatoid arthritis patients exhibit impaired Candida albicans-specific Th17 responses. Arthritis Res. Ther. 2014, 16, R50. [Google Scholar] [CrossRef]
  130. Limon, J.J.; Skalski, J.H.; Underhill, D.M. Commensal Fungi in Health and Disease. Cell Host Microbe 2017, 22, 156–165. [Google Scholar] [CrossRef]
  131. Schinnerling, K.; Rosas, C.; Soto, L.; Thomas, R.; Aguillón, J.C. Humanized Mouse Models of Rheumatoid Arthritis for Studies on Immunopathogenesis and Preclinical Testing of Cell-Based Therapies. Front. Immunol. 2019, 10, 203. [Google Scholar] [CrossRef] [PubMed]
  132. Romero-Figueroa, M.D.S.; Ramírez-Durán, N.; Montiel-Jarquín, A.J.; Horta-Baas, G. Gut-joint axis: Gut dysbiosis can contribute to the onset of rheumatoid arthritis via multiple pathways. Front. Cell Infect. Microbiol. 2023, 13, 1092118. [Google Scholar] [CrossRef]
  133. Altieri, C.; Speranza, B.; Corbo, M.R.; Sinigaglia, M.; Bevilacqua, A. Gut-Microbiota, and Multiple Sclerosis: Background, Evidence, and Perspectives. Nutrients 2023, 15, 942. [Google Scholar] [CrossRef]
  134. Yadav, M.; Ali, S.; Shrode, R.L.; Shahi, S.K.; Jensen, S.N.; Hoang, J.; Cassidy, S.; Olalde, H.; Guseva, N.; Paullus, M.; et al. Multiple sclerosis patients have an altered gut mycobiome and increased fungal to bacterial richness. PLoS ONE 2022, 17, e0264556. [Google Scholar] [CrossRef]
  135. Carabotti, M.; Scirocco, A.; Maselli, M.A.; Severi, C. The gut-brain axis: Interactions between enteric microbiota, central and enteric nervous systems. Ann. Gastroenterol. 2015, 28, 203–209. [Google Scholar]
  136. Sittipo, P.; Choi, J.; Lee, S.; Lee, Y.K. The function of gut microbiota in immune-related neurological disorders: A review. J. Neuroinflammation 2022, 19, 154. [Google Scholar] [CrossRef]
  137. Buga, A.M.; Padureanu, V.; Riza, A.L.; Oancea, C.N.; Albu, C.V.; Nica, A.D. The Gut-Brain Axis as a Therapeutic Target in Multiple Sclerosis. Cells 2023, 12, 1872. [Google Scholar] [CrossRef] [PubMed]
  138. Chaudhary, N.; Marr, K.A. Impact of Aspergillus fumigatus in allergic airway diseases. Clin. Transl. Allergy 2011, 1, 4. [Google Scholar] [CrossRef]
  139. Kanj, A.N.; Skalski, J.H. Gut Mycobiome and Asthma. J. Fungi 2024, 10, 192. [Google Scholar] [CrossRef] [PubMed]
  140. van Tilburg Bernardes, E.; Gutierrez, M.W.; Arrieta, M.-C. The Fungal Microbiome and Asthma. Front. Cell. Infect. Microbiol. 2020, 10, 583418. [Google Scholar] [CrossRef]
  141. Kanj, A.N.; Kottom, T.J.; Schaefbauer, K.J.; Choudhury, M.; Limper, A.H.; Skalski, J.H. Dysbiosis of the intestinal fungal microbiota increases lung resident group 2 innate lymphoid cells and is associated with enhanced asthma severity in mice and humans. Respir. Res. 2023, 24, 144. [Google Scholar] [CrossRef] [PubMed]
  142. Kaur, R.; Dhakad, M.S.; Goyal, R.; Bhalla, P.; Dewan, R. Spectrum of Opportunistic Fungal Infections in HIV/AIDS Patients in Tertiary Care Hospital in India. Can. J. Infect. Dis. Med. Microbiol. 2016, 2016, 2373424. [Google Scholar] [CrossRef]
  143. Jerez Puebla, L.E. Fungal Infections in Immunosuppressed Patients. In Immunodeficiency; Metodiev, K., Ed.; IntechOpen: Rijeka, Croatia, 2012. [Google Scholar]
  144. Badiee, P.; Hashemizadeh, Z. Opportunistic invasive fungal infections: Diagnosis & clinical management. Indian. J. Med. Res. 2014, 139, 195–204. [Google Scholar]
  145. Anju, V.T.; Busi, S.; Imchen, M.; Kumavath, R.; Mohan, M.S.; Salim, S.A.; Subhaswaraj, P.; Dyavaiah, M. Polymicrobial Infections and Biofilms: Clinical Significance and Eradication Strategies. Antibiotics 2022, 11, 1731. [Google Scholar] [CrossRef]
  146. Wang, L.; Cao, Y.; Lou, E.; Zhao, X.; Chen, X. The role of gut fungi in Clostridioides difficile infection. Biomed. J. 2024, 47, 100686. [Google Scholar] [CrossRef]
  147. Smits, W.K.; Lyras, D.; Lacy, D.B.; Wilcox, M.H.; Kuijper, E.J. Clostridium difficile infection. Nat. Rev. Dis. Primers 2016, 2, 16020. [Google Scholar] [CrossRef]
  148. Pérez, J.C. The interplay between gut bacteria and the yeast Candida albicans. Gut Microbes 2021, 13, 1979877. [Google Scholar] [CrossRef]
  149. Vallabhaneni, S.; Almendares, O.M.; Farley, M.M.; Reno, J.; Smith, Z.; Stein, B.; Magill, S.S.; Smith, R.M.; Cleveland, A.; Lessa, F.C. Candida Co-infection Among Adults with Clostridium difficile Infection in Metropolitan Atlanta, 2009–2013. Open Forum Infect. Dis. 2015, 2 (Suppl. 1), 921. [Google Scholar] [CrossRef]
  150. van Leeuwen, P.T.; van der Peet, J.M.; Bikker, F.J.; Hoogenkamp, M.A.; Oliveira Paiva, A.M.; Kostidis, S.; Mayboroda, O.A.; Smits, W.K.; Krom, B.P. Interspecies Interactions between Clostridium difficile and Candida albicans. mSphere 2016, 1, e00187-16. [Google Scholar] [CrossRef] [PubMed]
  151. Yetgin, A. Exploring the Link Between the Gut Mycobiome and Neurological Disorders. Adv. Gut Microbiome Res. 2024, 2024, 9965893. [Google Scholar] [CrossRef]
  152. Hadrich, I.; Turki, M.; Chaari, I.; Abdelmoula, B.; Gargouri, R.; Khemakhem, N.; Elatoui, D.; Abid, F.; Kammoun, S.; Rekik, M.; et al. Gut mycobiome and neuropsychiatric disorders: Insights and therapeutic potential. Front. Cell. Neurosci. 2025, 18, 1495224. [Google Scholar] [CrossRef]
  153. Gamal, A.; Elshaer, M.; Alabdely, M.; Kadry, A.; McCormick, T.S.; Ghannoum, M. The Mycobiome: Cancer Pathogenesis, Diagnosis, and Therapy. Cancers 2022, 14, 2875. [Google Scholar] [CrossRef]
  154. Yunus, A.; Mokhtar, N.M.; Raja Ali, R.A.; Ahmad Kendong, S.M.; Ahmad, H.F. Methods for identification of the opportunistic gut mycobiome from colorectal adenocarcinoma biopsy tissues. MethodsX 2024, 12, 102623. [Google Scholar] [CrossRef]
  155. Cheng, W.; Li, F.; Gao, Y.; Yang, R. Fungi and tumors: The role of fungi in tumorigenesis (Review). Int. J. Oncol. 2024, 64, 52. [Google Scholar] [CrossRef]
  156. Prakash, V.; Singh, R.K.; Saurabh, K.; Kumar, V.; Kumari, R.; Kumar, S.; Rajpal, K.; Sinha, D.K.; Parwez, A. Spectrum of chemo-radiotherapy induced fungal infection in head and neck cancer patients at tertiary care centre of Eastern India. Oral Oncol. Rep. 2023, 6, 100039. [Google Scholar] [CrossRef]
  157. Reynolds, S. Gut Microbes May Influence How Well Radiation Therapy Works Against Cancer. Available online: https://www.cancer.gov/news-events/cancer-currents-blog/2021/cancer-fungi-gut-radiation-therapy (accessed on 23 February 2025).
  158. Cong, L.; Chen, C.; Mao, S.; Han, Z.; Zhu, Z.; Li, Y. Intestinal bacteria-a powerful weapon for fungal infections treatment. Front. Cell Infect. Microbiol. 2023, 13, 1187831. [Google Scholar] [CrossRef] [PubMed]
  159. Lemoinne, S.; Kemgang, A.; Ben Belkacem, K.; Straube, M.; Jegou, S.; Corpechot, C.; Chazouillères, O.; Housset, C.; Sokol, H. Fungi participate in the dysbiosis of gut microbiota in patients with primary sclerosing cholangitis. Gut 2020, 69, 92–102. [Google Scholar] [CrossRef]
  160. Krohn, S.; Zeller, K.; Böhm, S.; Chatzinotas, A.; Harms, H.; Hartmann, J.; Heidtmann, A.; Herber, A.; Kaiser, T.; Treuheit, M.; et al. Molecular quantification and differentiation of Candida species in biological specimens of patients with liver cirrhosis. PLoS ONE 2018, 13, e0197319. [Google Scholar] [CrossRef] [PubMed]
  161. Noel, S.; Martina-Lingua, M.N.; Bandapalle, S.; Pluznick, J.; Hamad, A.R.; Peterson, D.A.; Rabb, H. Intestinal microbiota-kidney cross talk in acute kidney injury and chronic kidney disease. Nephron Clin. Pract. 2014, 127, 139–143. [Google Scholar] [CrossRef]
  162. Kim, D.H.; Kim, H.; Jeong, D.; Kang, I.B.; Chon, J.W.; Kim, H.S.; Song, K.Y.; Seo, K.H. Kefir alleviates obesity and hepatic steatosis in high-fat diet-fed mice by modulation of gut microbiota and mycobiota: Targeted and untargeted community analysis with correlation of biomarkers. J. Nutr. Biochem. 2017, 44, 35–43. [Google Scholar] [CrossRef] [PubMed]
  163. Wu, X.; Xia, Y.; He, F.; Zhu, C.; Ren, W. Intestinal mycobiota in health and diseases: From a disrupted equilibrium to clinical opportunities. Microbiome 2021, 9, 60. [Google Scholar] [CrossRef]
  164. Fakhim, H.; Vaezi, A.; Dannaoui, E.; Chowdhary, A.; Nasiry, D.; Faeli, L.; Meis, J.F.; Badali, H. Comparative virulence of Candida auris with Candida haemulonii, Candida glabrata and Candida albicans in a murine model. Mycoses 2018, 61, 377–382. [Google Scholar] [CrossRef]
  165. Gosiewski, T.; Salamon, D.; Szopa, M.; Sroka, A.; Malecki, M.T.; Bulanda, M. Quantitative evaluation of fungi of the genus Candida in the feces of adult patients with type 1 and 2 diabetes—A pilot study. Gut Pathog. 2014, 6, 43. [Google Scholar] [CrossRef] [PubMed]
  166. Pushalkar, S.; Hundeyin, M.; Daley, D.; Zambirinis, C.P.; Kurz, E.; Mishra, A.; Mohan, N.; Aykut, B.; Usyk, M.; Torres, L.E.; et al. The Pancreatic Cancer Microbiome Promotes Oncogenesis by Induction of Innate and Adaptive Immune Suppression. Cancer Discov. 2018, 8, 403–416. [Google Scholar] [CrossRef]
  167. Yang, J.; Yao, J.L.; Wu, Z.Q.; Zeng, D.L.; Zheng, L.Y.; Chen, D.; Guo, Z.D.; Peng, L. Current opinions on the mechanism, classification, imaging diagnosis and treatment of post-traumatic osteomyelitis. Chin. J. Traumatol. 2021, 24, 320–327. [Google Scholar] [CrossRef]
  168. Cangui-Panchi, S.P.; Ñacato-Toapanta, A.L.; Enríquez-Martínez, L.J.; Reyes, J.; Garzon-Chavez, D.; Machado, A. Biofilm-forming microorganisms causing hospital-acquired infections from intravenous catheter: A systematic review. Curr. Res. Microb. Sci. 2022, 3, 100175. [Google Scholar] [CrossRef] [PubMed]
  169. Baddour, L.M.; Wilson, W.R.; Bayer, A.S.; Fowler, V.G., Jr.; Tleyjeh, I.M.; Rybak, M.J.; Barsic, B.; Lockhart, P.B.; Gewitz, M.H.; Levison, M.E.; et al. Infective Endocarditis in Adults: Diagnosis, Antimicrobial Therapy, and Management of Complications: A Scientific Statement for Healthcare Professionals from the American Heart Association. Circulation 2015, 132, 1435–1486. [Google Scholar] [CrossRef]
  170. Pruthi, V.; Al-Janabi, A.; Pereira, B.M. Characterization of biofilm formed on intrauterine devices. Indian J. Med. Microbiol. 2003, 21, 161–165. [Google Scholar] [CrossRef] [PubMed]
  171. Dahlen, G. Biofilms in Dental Unit Water Lines. Monogr. Oral Sci. 2021, 29, 12–18. [Google Scholar] [CrossRef]
  172. Stickler, D.J.; King, J.B.; Winters, C.; Morris, S.L. Blockage of urethral catheters by bacterial biofilms. J. Infect. 1993, 27, 133–135. [Google Scholar] [CrossRef]
  173. Costerton, J.W.; Stewart, P.S.; Greenberg, E.P. Bacterial Biofilms: A Common Cause of Persistent Infections. Science 1999, 284, 1318–1322. [Google Scholar] [CrossRef]
  174. Singh, S.; Datta, S.; Narayanan, K.B.; Rajnish, K.N. Bacterial exo-polysaccharides in biofilms: Role in antimicrobial resistance and treatments. J. Genet. Eng. Biotechnol. 2021, 19, 140. [Google Scholar] [CrossRef] [PubMed]
  175. Fleming, D.; Rumbaugh, K.P. Approaches to Dispersing Medical Biofilms. Microorganisms 2017, 5, 15. [Google Scholar] [CrossRef]
  176. Gallant, C.V.; Daniels, C.; Leung, J.M.; Ghosh, A.S.; Young, K.D.; Kotra, L.P.; Burrows, L.L. Common beta-lactamases inhibit bacterial biofilm formation. Mol. Microbiol. 2005, 58, 1012–1024. [Google Scholar] [CrossRef]
  177. Ortíz-Pérez, A.; Martín-de-Hijas, N.; Alonso-Rodríguez, N.; Molina-Manso, D.; Fernández-Roblas, R.; Esteban, J. Importance of antibiotic penetration in the antimicrobial resistance of biofilm formed by non-pigmented rapidly growing mycobacteria against amikacin, ciprofloxacin and clarithromycin. Enferm. Infecc. Microbiol. Clin. 2011, 29, 79–84. [Google Scholar] [CrossRef]
  178. Muñoz-Egea, M.C.; García-Pedrazuela, M.; Mahillo-Fernandez, I.; Esteban, J. Effect of Antibiotics and Antibiofilm Agents in the Ultrastructure and Development of Biofilms Developed by Nonpigmented Rapidly Growing Mycobacteria. Microb. Drug Resist. 2016, 22, 1–6. [Google Scholar] [CrossRef] [PubMed]
  179. Esteban, J.; Martín-de-Hijas, N.Z.; García-Almeida, D.; Bodas-Sánchez, A.; Gadea, I.; Fernández-Roblas, R. Prevalence of erm methylase genes in clinical isolates of non-pigmented, rapidly growing mycobacteria. Clin. Microbiol. Infect. 2009, 15, 919–923. [Google Scholar] [CrossRef]
  180. Jiang, Y.; Geng, M.; Bai, L. Targeting Biofilms Therapy: Current Research Strategies and Development Hurdles. Microorganisms 2020, 8, 1222. [Google Scholar] [CrossRef]
  181. Jiang, Q.; Jin, Z.; Sun, B. MgrA Negatively Regulates Biofilm Formation and Detachment by Repressing the Expression of psm Operons in Staphylococcus aureus. Appl. Environ. Microbiol. 2018, 84, e01008-18. [Google Scholar] [CrossRef]
  182. Sambanthamoorthy, K.; Gokhale, A.A.; Lao, W.; Parashar, V.; Neiditch, M.B.; Semmelhack, M.F.; Lee, I.; Waters, C.M. Identification of a novel benzimidazole that inhibits bacterial biofilm formation in a broad-spectrum manner. Antimicrob. Agents Chemother. 2011, 55, 4369–4378. [Google Scholar] [CrossRef]
  183. Dinicola, S.; De Grazia, S.; Carlomagno, G.; Pintucci, J.P. N-acetylcysteine as powerful molecule to destroy bacterial biofilms. A systematic review. Eur. Rev. Med. Pharmacol. Sci. 2014, 18, 2942–2948. [Google Scholar]
  184. Veerachamy, S.; Yarlagadda, T.; Manivasagam, G.; Yarlagadda, P.K. Bacterial adherence and biofilm formation on medical implants: A review. Proc. Inst. Mech. Eng. H 2014, 228, 1083–1099. [Google Scholar] [CrossRef] [PubMed]
  185. Zaongo, S.D.; Ouyang, J.; Isnard, S.; Zhou, X.; Harypursat, V.; Cui, H.; Routy, J.P.; Chen, Y. Candida albicans can foster gut dysbiosis and systemic inflammation during HIV infection. Gut Microbes 2023, 15, 2167171. [Google Scholar] [CrossRef] [PubMed]
  186. Ghannoum, M.A.; Rice, L.B. Antifungal agents: Mode of action, mechanisms of resistance, and correlation of these mechanisms with bacterial resistance. Clin. Microbiol. Rev. 1999, 12, 501–517. [Google Scholar] [CrossRef]
  187. Osset-Trénor, P.; Pascual-Ahuir, A.; Proft, M. Fungal Drug Response and Antimicrobial Resistance. J. Fungi 2023, 9, 565. [Google Scholar] [CrossRef]
  188. de Oliveira Santos, G.C.; Vasconcelos, C.C.; Lopes, A.J.O.; de Sousa Cartágenes, M.D.S.; Filho, A.; do Nascimento, F.R.F.; Ramos, R.M.; Pires, E.; de Andrade, M.S.; Rocha, F.M.G.; et al. Candida Infections and Therapeutic Strategies: Mechanisms of Action for Traditional and Alternative Agents. Front. Microbiol. 2018, 9, 1351. [Google Scholar] [CrossRef]
  189. Heng, X.; Jiang, Y.; Chu, W. Influence of Fluconazole Administration on Gut Microbiome, Intestinal Barrier, and Immune Response in Mice. Antimicrob. Agents Chemother. 2021, 65, e02552-20. [Google Scholar] [CrossRef]
  190. Hof, H. Is there a serious risk of resistance development to azoles among fungi due to the widespread use and long-term application of azole antifungals in medicine? Drug Resist. Updat. 2008, 11, 25–31. [Google Scholar] [CrossRef]
  191. Birch, M.; Sibley, G. 5.22—Antifungal Chemistry Review. In Comprehensive Medicinal Chemistry III; Chackalamannil, S., Rotella, D., Ward, S.E., Eds.; Elsevier: Oxford, UK, 2017; pp. 703–716. [Google Scholar] [CrossRef]
  192. Denning, D.W.; Hope, W.W. Therapy for fungal diseases: Opportunities and priorities. Trends Microbiol. 2010, 18, 195–204. [Google Scholar] [CrossRef]
  193. Gallagher, J.J.; Williams-Bouyer, N.; Villarreal, C.; Heggers, J.P.; Herndon, D.N. Chapter 12—Treatment of infection in burns. In Total Burn Care, 3rd ed.; Herndon, D.N., Ed.; W.B. Saunders: Edinburgh, UK, 2007; pp. 136–176. [Google Scholar] [CrossRef]
  194. Banaszak, M.; Górna, I.; Woźniak, D.; Przysławski, J.; Drzymała-Czyż, S. Association between Gut Dysbiosis and the Occurrence of SIBO, LIBO, SIFO and IMO. Microorganisms 2023, 11, 573. [Google Scholar] [CrossRef]
  195. Sousa, F.; Nascimento, C.; Ferreira, D.; Reis, S.; Costa, P. Reviving the interest in the versatile drug nystatin: A multitude of strategies to increase its potential as an effective and safe antifungal agent. Adv. Drug Deliv. Rev. 2023, 199, 114969. [Google Scholar] [CrossRef]
  196. Delaloye, J.; Calandra, T. Invasive candidiasis as a cause of sepsis in the critically ill patient. Virulence 2014, 5, 161–169. [Google Scholar] [CrossRef]
  197. Szymański, M.; Chmielewska, S.; Czyżewska, U.; Malinowska, M.; Tylicki, A. Echinocandins—Structure, mechanism of action and use in antifungal therapy. J. Enzym. Inhib. Med. Chem. 2022, 37, 876–894. [Google Scholar] [CrossRef]
  198. Lionakis, M.S.; Drummond, R.A.; Hohl, T.M. Immune responses to human fungal pathogens and therapeutic prospects. Nat. Rev. Immunol. 2023, 23, 433–452. [Google Scholar] [CrossRef]
  199. Jawhara, S. How Gut Bacterial Dysbiosis Can Promote Candida albicans Overgrowth during Colonic Inflammation. Microorganisms 2022, 10, 1014. [Google Scholar] [CrossRef]
  200. Puškárová, A.; Bučková, M.; Kraková, L.; Pangallo, D.; Kozics, K. The antibacterial and antifungal activity of six essential oils and their cyto/genotoxicity to human HEL 12469 cells. Sci. Rep. 2017, 7, 8211. [Google Scholar] [CrossRef]
  201. da Silva, A.R.; de Andrade Neto, J.B.; da Silva, C.R.; Campos Rde, S.; Costa Silva, R.A.; Freitas, D.D.; do Nascimento, F.B.; de Andrade, L.N.; Sampaio, L.S.; Grangeiro, T.B.; et al. Berberine Antifungal Activity in Fluconazole-Resistant Pathogenic Yeasts: Action Mechanism Evaluated by Flow Cytometry and Biofilm Growth Inhibition in Candida spp. Antimicrob. Agents Chemother. 2016, 60, 3551–3557. [Google Scholar] [CrossRef]
  202. Simo, L.O.T. An In-Vitro Study to Determine the Antimicrobial Properties of Artostaphylos uva-ursi on the Growth of Aspergillus, Candida albicans and Escherichia coli. Master’s Thesis, University of Johannesburg, Johannesburg, South Africa, 2018. [Google Scholar]
  203. Gupta, S. Review on Uva-Ursi—A miracle herb for urinary tract disorders. World J. Pharm. Life Sci. 2017, 3, 51–54. [Google Scholar]
  204. Shi, D.; Zhao, Y.; Yan, H.; Fu, H.; Shen, Y.; Lu, G.; Mei, H.; Qiu, Y.; Li, D.; Liu, W. Antifungal effects of undecylenic acid on the biofilm formation of Candida albicans. Int. J. Clin. Pharmacol. Ther. 2016, 54, 343–353. [Google Scholar] [CrossRef]
  205. Hossain, C.M.; Ryan, L.K.; Gera, M.; Choudhuri, S.; Lyle, N.; Ali, K.A.; Diamond, G. Antifungals and Drug Resistance. Encyclopedia 2022, 2, 1722–1737. [Google Scholar] [CrossRef]
  206. Frey-Klett, P.; Burlinson, P.; Deveau, A.; Barret, M.; Tarkka, M.; Sarniguet, A. Bacterial-fungal interactions: Hyphens between agricultural, clinical, environmental, and food microbiologists. Microbiol. Mol. Biol. Rev. 2011, 75, 583–609. [Google Scholar] [CrossRef]
  207. Stamatiades, G.A.; Ioannou, P.; Petrikkos, G.; Tsioutis, C. Fungal infections in patients with inflammatory bowel disease: A systematic review. Mycoses 2018, 61, 366–376. [Google Scholar] [CrossRef]
  208. Erdogan, A.; Rao, S.S. Small Intestinal Fungal Overgrowth. Curr. Gastroenterol. Rep. 2015, 17, 16. [Google Scholar] [CrossRef]
  209. Zangl, I.; Pap, I.J.; Aspöck, C.; Schüller, C. The role of Lactobacillus species in the control of Candida via biotrophic interactions. Microb. Cell 2019, 7, 1–14. [Google Scholar] [CrossRef]
  210. Abdel-Nasser, M.; Abdel-Maksoud, G.; Eid, A.M.; Hassan, S.E.; Abdel-Nasser, A.; Alharbi, M.; Elkelish, A.; Fouda, A. Antifungal Activity of Cell-Free Filtrate of Probiotic Bacteria Lactobacillus rhamnosus ATCC-7469 against Fungal Strains Isolated from a Historical Manuscript. Microorganisms 2023, 11, 1104. [Google Scholar] [CrossRef]
  211. Montiel, R.; Martín-Cabrejas, I.; Langa, S.; El Aouad, N.; Arqués, J.L.; Reyes, F.; Medina, M. Antimicrobial activity of reuterin produced by Lactobacillus reuteri on Listeria monocytogenes in cold-smoked salmon. Food Microbiol. 2014, 44, 1–5. [Google Scholar] [CrossRef] [PubMed]
  212. Schaefer, L.; Auchtung, T.A.; Hermans, K.E.; Whitehead, D.; Borhan, B.; Britton, R.A. The antimicrobial compound reuterin (3-hydroxypropionaldehyde) induces oxidative stress via interaction with thiol groups. Microbiology 2010, 156, 1589–1599. [Google Scholar] [CrossRef] [PubMed]
  213. Ricci, L.; Mackie, J.; Donachie, G.E.; Chapuis, A.; Mezerová, K.; Lenardon, M.D.; Brown, A.J.P.; Duncan, S.H.; Walker, A.W. Human gut bifidobacteria inhibit the growth of the opportunistic fungal pathogen Candida albicans. FEMS Microbiol. Ecol. 2022, 98, fiac095. [Google Scholar] [CrossRef]
  214. Mendonça, F.H.; Santos, S.S.; Faria Ida, S.; Gonçalves e Silva, C.R.; Jorge, A.O.; Leão, M.V. Effects of probiotic bacteria on Candida presence and IgA anti-Candida in the oral cavity of elderly. Braz. Dent. J. 2012, 23, 534–538. [Google Scholar] [CrossRef]
  215. Alonso-Roman, R.; Last, A.; Mirhakkak, M.H.; Sprague, J.L.; Möller, L.; Großmann, P.; Graf, K.; Gratz, R.; Mogavero, S.; Vylkova, S.; et al. Lactobacillus rhamnosus colonisation antagonizes Candida albicans by forcing metabolic adaptations that compromise pathogenicity. Nat. Commun. 2022, 13, 3192. [Google Scholar] [CrossRef]
  216. Guo, H.; Yu, L.; Tian, F.; Chen, W.; Zhai, Q. The Potential Therapeutic Role of Lactobacillaceae rhamnosus for Treatment of Inflammatory Bowel Disease. Foods 2023, 12, 692. [Google Scholar] [CrossRef] [PubMed]
  217. Pais, P.; Almeida, V.; Yılmaz, M.; Teixeira, M.C. Saccharomyces boulardii: What Makes It Tick as Successful Probiotic? J. Fungi 2020, 6, 78. [Google Scholar] [CrossRef]
  218. Kunyeit, L.; K A, A.A.; Rao, R.P. Application of Probiotic Yeasts on Candida Species Associated Infection. J. Fungi 2020, 6, 189. [Google Scholar] [CrossRef]
  219. Krasowska, A.; Murzyn, A.; Dyjankiewicz, A.; Łukaszewicz, M.; Dziadkowiec, D. The antagonistic effect of Saccharomyces boulardii on Candida albicans filamentation, adhesion and biofilm formation. FEMS Yeast Res. 2009, 9, 1312–1321. [Google Scholar] [CrossRef]
  220. McFarland, L.V.; Huang, Y.; Wang, L.; Malfertheiner, P. Systematic review and meta-analysis: Multi-strain probiotics as adjunct therapy for Helicobacter pylori eradication and prevention of adverse events. United Eur. Gastroenterol. J. 2016, 4, 546–561. [Google Scholar] [CrossRef]
  221. Gul, S.; Durante-Mangoni, E. Unraveling the Puzzle: Health Benefits of Probiotics—A Comprehensive Review. J. Clin. Med. 2024, 13, 1436. [Google Scholar] [CrossRef] [PubMed]
  222. Biazzo, M.; Deidda, G. Fecal Microbiota Transplantation as New Therapeutic Avenue for Human Diseases. J. Clin. Med. 2022, 11, 4119. [Google Scholar] [CrossRef] [PubMed]
  223. Sahle, Z.; Engidaye, G.; Shenkute Gebreyes, D.; Adenew, B.; Abebe, T.A. Fecal microbiota transplantation and next-generation therapies: A review on targeting dysbiosis in metabolic disorders and beyond. SAGE Open Med. 2024, 12, 20503121241257486. [Google Scholar] [CrossRef]
  224. Karimi, M.; Shirsalimi, N.; Hashempour, Z.; Salehi Omran, H.; Sedighi, E.; Beigi, F.; Mortezazadeh, M. Safety and efficacy of fecal microbiota transplantation (FMT) as a modern adjuvant therapy in various diseases and disorders: A comprehensive literature review. Front. Immunol. 2024, 15, 1439176. [Google Scholar] [CrossRef] [PubMed]
  225. de Groot, P.F.; Frissen, M.N.; de Clercq, N.C.; Nieuwdorp, M. Fecal microbiota transplantation in metabolic syndrome: History, present and future. Gut Microbes 2017, 8, 253–267. [Google Scholar] [CrossRef]
  226. Zhang, Z.; Mocanu, V.; Cai, C.; Dang, J.; Slater, L.; Deehan, E.C.; Walter, J.; Madsen, K.L. Impact of Fecal Microbiota Transplantation on Obesity and Metabolic Syndrome—A Systematic Review. Nutrients 2019, 11, 2291. [Google Scholar] [CrossRef]
  227. Chen, Q.; Fan, Y.; Zhang, B.; Yan, C.; Chen, Z.; Wang, L.; Hu, Y.; Huang, Q.; Su, J.; Ren, J.; et al. Specific fungi associated with response to capsulized fecal microbiota transplantation in patients with active ulcerative colitis. Front. Cell Infect. Microbiol. 2022, 12, 1086885. [Google Scholar] [CrossRef]
  228. Park, S.Y.; Seo, G.S. Fecal Microbiota Transplantation: Is It Safe? Clin. Endosc. 2021, 54, 157–160. [Google Scholar] [CrossRef]
  229. van Lier, Y.F.; Rolling, T.; Armijo, G.K.; Zhai, B.; Haverkate, N.J.E.; Meijer, E.; Nur, E.; Blom, B.; Peled, J.U.; van den Brink, M.R.M.; et al. Profiling the Fungal Microbiome after Fecal Microbiota Transplantation for Graft-versus-Host Disease: Insights from a Phase 1 Interventional Study. Transplant. Cell. Ther. 2023, 29, e61–e63. [Google Scholar] [CrossRef]
  230. Zhang, P. Influence of Foods and Nutrition on the Gut Microbiome and Implications for Intestinal Health. Int. J. Mol. Sci. 2022, 23, 9588. [Google Scholar] [CrossRef]
  231. Aziz, T.; Hussain, N.; Hameed, Z.; Lin, L. Elucidating the role of diet in maintaining gut health to reduce the risk of obesity, cardiovascular and other age-related inflammatory diseases: Recent challenges and future recommendations. Gut Microbes 2024, 16, 2297864. [Google Scholar] [CrossRef] [PubMed]
  232. Fu, J.; Zheng, Y.; Gao, Y.; Xu, W. Dietary Fiber Intake and Gut Microbiota in Human Health. Microorganisms 2022, 10, 2507. [Google Scholar] [CrossRef]
  233. Jawhara, S. How Do Polyphenol-Rich Foods Prevent Oxidative Stress and Maintain Gut Health? Microorganisms 2024, 12, 1570. [Google Scholar] [CrossRef]
  234. Shankar, J. Food Habit Associated Mycobiota Composition and Their Impact on Human Health. Front. Nutr. 2021, 8, 773577. [Google Scholar] [CrossRef] [PubMed]
  235. Soldán, M.; Argalášová, Ľ.; Hadvinová, L.; Galileo, B.; Babjaková, J. The Effect of Dietary Types on Gut Microbiota Composition and Development of Non-Communicable Diseases: A Narrative Review. Nutrients 2024, 16, 3134. [Google Scholar] [CrossRef] [PubMed]
  236. Medeiros, M.J.; Seo, L.; Macias, A.; Price, D.K.; Yew, J.Y. Bacterial and fungal components of the gut microbiome have distinct, sex-specific roles in Hawaiian Drosophila reproduction. bioRxiv 2023. [Google Scholar] [CrossRef]
  237. Guo, W.-L.; Deng, J.-C.; Pan, Y.-Y.; Xu, J.-X.; Hong, J.-L.; Shi, F.-F.; Liu, G.-L.; Qian, M.; Bai, W.-D.; Zhang, W.; et al. Hypoglycemic and hypolipidemic activities of Grifola frondosa polysaccharides and their relationships with the modulation of intestinal microflora in diabetic mice induced by high-fat diet and streptozotocin. Int. J. Biol. Macromol. 2020, 153, 1231–1240. [Google Scholar] [CrossRef]
  238. Singh, R.K.; Chang, H.W.; Yan, D.; Lee, K.M.; Ucmak, D.; Wong, K.; Abrouk, M.; Farahnik, B.; Nakamura, M.; Zhu, T.H.; et al. Influence of diet on the gut microbiome and implications for human health. J. Transl. Med. 2017, 15, 73. [Google Scholar] [CrossRef]
  239. Buttar, J.; Kon, E.; Lee, A.; Kaur, G.; Lunken, G. Effect of diet on the gut mycobiome and potential implications in inflammatory bowel disease. Gut Microbes 2024, 16, 2399360. [Google Scholar] [CrossRef]
  240. Van Ende, M.; Wijnants, S.; Van Dijck, P. Sugar Sensing and Signaling in Candida albicans and Candida glabrata. Front. Microbiol. 2019, 10, 99. [Google Scholar] [CrossRef]
  241. Mohammed, L.; Jha, G.; Malasevskaia, I.; Goud, H.K.; Hassan, A. The Interplay Between Sugar and Yeast Infections: Do Diabetics Have a Greater Predisposition to Develop Oral and Vulvovaginal Candidiasis? Cureus 2021, 13, e13407. [Google Scholar] [CrossRef] [PubMed]
  242. Palmucci, J.R.; Sells, B.E.; Giamberardino, C.D.; Toffaletti, D.L.; Dai, B.; Asfaw, Y.G.; Dubois, L.G.; Li, Z.; Theriot, B.; Schell, W.A.; et al. A ketogenic diet enhances fluconazole efficacy in murine models of systemic fungal infection. mBio 2024, 15, e0064924. [Google Scholar] [CrossRef]
  243. Jeziorek, M.; Frej-Mądrzak, M.; Choroszy-Król, I. The influence of diet on gastrointestinal Candida spp. colonization and the susceptibility of Candida spp. to antifungal drugs. Rocz. Panstw. Zakl. Hig. 2019, 70, 195–200. [Google Scholar] [CrossRef]
  244. Jawhara, S. Healthy Diet and Lifestyle Improve the Gut Microbiota and Help Combat Fungal Infection. Microorganisms 2023, 11, 1556. [Google Scholar] [CrossRef] [PubMed]
  245. Martyniak, A.; Medyńska-Przęczek, A.; Wędrychowicz, A.; Skoczeń, S.; Tomasik, P.J. Prebiotics, Probiotics, Synbiotics, Paraprobiotics and Postbiotic Compounds in IBD. Biomolecules 2021, 11, 1903. [Google Scholar] [CrossRef] [PubMed]
  246. Abrignani, V.; Salvo, A.; Pacinella, G.; Tuttolomondo, A. The Mediterranean Diet, Its Microbiome Connections, and Cardiovascular Health: A Narrative Review. Int. J. Mol. Sci. 2024, 25, 4942. [Google Scholar] [CrossRef]
  247. Santana, I.L.; Gonçalves, L.M.; de Vasconcellos, A.A.; da Silva, W.J.; Cury, J.A.; Del Bel Cury, A.A. Dietary carbohydrates modulate Candida albicans biofilm development on the denture surface. PLoS ONE 2013, 8, e64645. [Google Scholar] [CrossRef]
  248. Ribeiro, N.C.B.V.; Ramer-Tait, A.E.; Cazarin, C.B.B. Resistant starch: A promising ingredient and health promoter. PharmaNutrition 2022, 21, 100304. [Google Scholar] [CrossRef]
  249. Mirabelli, M.; Shehab, R. Chapter 19—Nutrition. In Clinical Men’s Health; Heidelbaugh, J.J., Ed.; W.B. Saunders: Philadelphia, PA, USA, 2008; pp. 349–385. [Google Scholar] [CrossRef]
  250. Mandal, S.M.; Mahata, D.; Migliolo, L.; Parekh, A.; Addy, P.S.; Mandal, M.; Basak, A. Glucose directly promotes antifungal resistance in the fungal pathogen, Candida spp. J. Biol. Chem. 2014, 289, 25468–25473. [Google Scholar] [CrossRef]
  251. Slavin, J. Fiber and prebiotics: Mechanisms and health benefits. Nutrients 2013, 5, 1417–1435. [Google Scholar] [CrossRef]
  252. Clemente-Suárez, V.J.; Mielgo-Ayuso, J.; Martín-Rodríguez, A.; Ramos-Campo, D.J.; Redondo-Flórez, L.; Tornero-Aguilera, J.F. The Burden of Carbohydrates in Health and Disease. Nutrients 2022, 14, 3809. [Google Scholar] [CrossRef]
  253. Peng, Z.; Tang, J. Intestinal Infection of Candida albicans: Preventing the Formation of Biofilm by C. albicans and Protecting the Intestinal Epithelial Barrier. Front. Microbiol. 2021, 12, 783010. [Google Scholar] [CrossRef] [PubMed]
  254. Basmaciyan, L.; Bon, F.; Paradis, T.; Lapaquette, P.; Dalle, F. Candida albicans Interactions with The Host: Crossing The Intestinal Epithelial Barrier. Tissue Barriers 2019, 7, 1612661. [Google Scholar] [CrossRef]
  255. Escalante, J.; Artaiz, O.; Diwakarla, S.; McQuade, R.M. Leaky gut in systemic inflammation: Exploring the link between gastrointestinal disorders and age-related diseases. GeroScience 2024, 47, 1–22. [Google Scholar] [CrossRef] [PubMed]
  256. Davani-Davari, D.; Negahdaripour, M.; Karimzadeh, I.; Seifan, M.; Mohkam, M.; Masoumi, S.J.; Berenjian, A.; Ghasemi, Y. Prebiotics: Definition, Types, Sources, Mechanisms, and Clinical Applications. Foods 2019, 8, 92. [Google Scholar] [CrossRef]
  257. Seo, Y.S.; Lee, H.B.; Kim, Y.; Park, H.Y. Dietary Carbohydrate Constituents Related to Gut Dysbiosis and Health. Microorganisms 2020, 8, 427. [Google Scholar] [CrossRef]
  258. What’s the Difference Between Starchy and Non-Starchy Vegetables? Available online: https://www.healthline.com/nutrition/starchy-vs-non-starchy-vegetables (accessed on 23 February 2025).
  259. Sugizaki, C.S.A.; Naves, M.M.V. Potential Prebiotic Properties of Nuts and Edible Seeds and Their Relationship to Obesity. Nutrients 2018, 10, 1645. [Google Scholar] [CrossRef] [PubMed]
  260. Ugural, A.; Akyol, A. Can pseudocereals modulate microbiota by functioning as probiotics or prebiotics? Crit. Rev. Food Sci. Nutr. 2022, 62, 1725–1739. [Google Scholar] [CrossRef]
  261. Sonia, S.; Witjaksono, F.; Ridwan, R. Effect of cooling of cooked white rice on resistant starch content and glycemic response. Asia Pac. J. Clin. Nutr. 2015, 24, 620–625. [Google Scholar] [CrossRef]
  262. Birt, D.F.; Boylston, T.; Hendrich, S.; Jane, J.L.; Hollis, J.; Li, L.; McClelland, J.; Moore, S.; Phillips, G.J.; Rowling, M.; et al. Resistant starch: Promise for improving human health. Adv. Nutr. 2013, 4, 587–601. [Google Scholar] [CrossRef]
  263. Argüelles, J.C.; Sánchez-Fresneda, R.; Argüelles, A.; Solano, F. Natural Substances as Valuable Alternative for Improving Conventional Antifungal Chemotherapy: Lights and Shadows. J. Fungi 2024, 10, 334. [Google Scholar] [CrossRef] [PubMed]
  264. Zhong, H.; Han, L.; Lu, R.Y.; Wang, Y. Antifungal and Immunomodulatory Ingredients from Traditional Chinese Medicine. Antibiotics 2022, 12, 48. [Google Scholar] [CrossRef]
  265. Nazzaro, F.; Fratianni, F.; Coppola, R.; Feo, V.D. Essential Oils and Antifungal Activity. Pharmaceuticals 2017, 10, 86. [Google Scholar] [CrossRef] [PubMed]
  266. Wang, X.; Qi, Y.; Zheng, H. Dietary Polyphenol, Gut Microbiota, and Health Benefits. Antioxidants 2022, 11, 1212. [Google Scholar] [CrossRef]
  267. Plamada, D.; Vodnar, D.C. Polyphenols-Gut Microbiota Interrelationship: A Transition to a New Generation of Prebiotics. Nutrients 2021, 14, 137. [Google Scholar] [CrossRef]
  268. Aslam, H.; Marx, W.; Rocks, T.; Loughman, A.; Chandrasekaran, V.; Ruusunen, A.; Dawson, S.L.; West, M.; Mullarkey, E.; Pasco, J.A.; et al. The effects of dairy and dairy derivatives on the gut microbiota: A systematic literature review. Gut Microbes 2020, 12, 1799533. [Google Scholar] [CrossRef] [PubMed]
  269. Aitzhanova, A.; Oleinikova, Y.; Mounier, J.; Hymery, N.; Leyva Salas, M.; Amangeldi, A.; Saubenova, M.; Alimzhanova, M.; Ashimuly, K.; Sadanov, A. Dairy associations for the targeted control of opportunistic Candida. World J. Microbiol. Biotechnol. 2021, 37, 143. [Google Scholar] [CrossRef]
  270. Chin, V.K.; Yong, V.C.; Chong, P.P.; Amin Nordin, S.; Basir, R.; Abdullah, M. Mycobiome in the Gut: A Multiperspective Review. Mediat. Inflamm. 2020, 2020, 9560684. [Google Scholar] [CrossRef]
  271. Schoch, C.L.; Seifert, K.A.; Huhndorf, S.; Robert, V.; Spouge, J.L.; Levesque, C.A.; Chen, W. Nuclear ribosomal internal transcribed spacer (ITS) region as a universal DNA barcode marker for Fungi. Proc. Natl. Acad. Sci. USA 2012, 109, 6241–6246. [Google Scholar] [CrossRef]
  272. Tedersoo, L.; Anslan, S.; Bahram, M.; Põlme, S.; Riit, T.; Liiv, I.; Kõljalg, U.; Kisand, V.; Nilsson, H.; Hildebrand, F.; et al. Shotgun metagenomes and multiple primer pair-barcode combinations of amplicons reveal biases in metabarcoding analyses of fungi. MycoKeys 2015, 10, 1–43. [Google Scholar] [CrossRef]
  273. Amend, A.S.; Seifert, K.A.; Samson, R.; Bruns, T.D. Indoor fungal composition is geographically patterned and more diverse in temperate zones than in the tropics. Proc. Natl. Acad. Sci. USA 2010, 107, 13748–13753. [Google Scholar] [CrossRef] [PubMed]
  274. Nilsson, R.H.; Larsson, K.H.; Taylor, A.F.S.; Bengtsson-Palme, J.; Jeppesen, T.S.; Schigel, D.; Kennedy, P.; Picard, K.; Glöckner, F.O.; Tedersoo, L.; et al. The UNITE database for molecular identification of fungi: Handling dark taxa and parallel taxonomic classifications. Nucleic Acids Res. 2019, 47, D259–D264. [Google Scholar] [CrossRef] [PubMed]
  275. Usyk, M.; Zolnik, C.P.; Patel, H.; Levi, M.H.; Burk, R.D. Novel ITS1 Fungal Primers for Characterization of the Mycobiome. mSphere 2017, 2, e00488-17. [Google Scholar] [CrossRef]
  276. Cui, L.; Morris, A.; Ghedin, E. The human mycobiome in health and disease. Genome Med. 2013, 5, 63. [Google Scholar] [CrossRef]
  277. Vaz, A.B.; Fonseca, P.L.; Leite, L.R.; Badotti, F.; Salim, A.C.; Araujo, F.M.; Cuadros-Orellana, S.; Duarte Ângelo, A.; Rosa, C.A.; Oliveira, G.; et al. USING Next-Generation Sequencing (NGS) TO UNCOVER DIVERSITY OF WOOD-DECAYING FUNGI IN NEOTROPICAL ATLANTIC FORESTS. Phytotaxa 2017, 295, 1–21. [Google Scholar] [CrossRef]
  278. Halwachs, B.; Madhusudhan, N.; Krause, R.; Nilsson, R.H.; Moissl-Eichinger, C.; Högenauer, C.; Thallinger, G.G.; Gorkiewicz, G. Critical Issues in Mycobiota Analysis. Front. Microbiol. 2017, 8, 180. [Google Scholar] [CrossRef]
  279. Gou, W.; Wang, H.; Su, C.; Fu, Y.; Wang, X.; Gao, C.; Shuai, M.; Miao, Z.; Zhang, J.; Jia, X.; et al. The temporal dynamics of the gut mycobiome and its association with cardiometabolic health in a nationwide cohort of 12,641 Chinese adults. Cell Rep. Med. 2024, 5, 101775. [Google Scholar] [CrossRef]
  280. Liang, G.; Bushman, F.D. The human virome: Assembly, composition and host interactions. Nat. Rev. Microbiol. 2021, 19, 514–527. [Google Scholar] [CrossRef]
  281. Borrel, G.; Brugère, J.F.; Gribaldo, S.; Schmitz, R.A.; Moissl-Eichinger, C. The host-associated archaeome. Nat. Rev. Microbiol. 2020, 18, 622–636. [Google Scholar] [CrossRef]
  282. Zuo, T.; Sun, Y.; Wan, Y.; Yeoh, Y.K.; Zhang, F.; Cheung, C.P.; Chen, N.; Luo, J.; Wang, W.; Sung, J.J.Y.; et al. Human-Gut-DNA Virome Variations across Geography, Ethnicity, and Urbanization. Cell Host Microbe 2020, 28, 741–751.e4. [Google Scholar] [CrossRef]
  283. Enaud, R.; Vandenborght, L.E.; Coron, N.; Bazin, T.; Prevel, R.; Schaeverbeke, T.; Berger, P.; Fayon, M.; Lamireau, T.; Delhaes, L. The Mycobiome: A Neglected Component in the Microbiota-Gut-Brain Axis. Microorganisms 2018, 6, 22. [Google Scholar] [CrossRef] [PubMed]
  284. Brun, P.; Scarpa, M.; Marchiori, C.; Sarasin, G.; Caputi, V.; Porzionato, A.; Giron, M.C.; Palù, G.; Castagliuolo, I. Saccharomyces boulardii CNCM I-745 supplementation reduces gastrointestinal dysfunction in an animal model of IBS. PLoS ONE 2017, 12, e0181863. [Google Scholar] [CrossRef]
  285. Takata, K.; Tomita, T.; Okuno, T.; Kinoshita, M.; Koda, T.; Honorat, J.A.; Takei, M.; Hagihara, K.; Sugimoto, T.; Mochizuki, H.; et al. Dietary Yeasts Reduce Inflammation in Central Nerve System via Microflora. Ann. Clin. Transl. Neurol. 2015, 2, 56–66. [Google Scholar] [CrossRef] [PubMed]
Figure 1. A balanced microbial environment and dysbiosis influence the interactions between bacteria and fungi within the gut microbiota. Under normal conditions, these microorganisms engage in cooperative relationships that support gut health. However, in dysbiosis, their interactions can become antagonistic, potentially disrupting immune regulation and contributing to disease development and progression. Created with Biorender.com (accessed on 16 February 2025).
Figure 1. A balanced microbial environment and dysbiosis influence the interactions between bacteria and fungi within the gut microbiota. Under normal conditions, these microorganisms engage in cooperative relationships that support gut health. However, in dysbiosis, their interactions can become antagonistic, potentially disrupting immune regulation and contributing to disease development and progression. Created with Biorender.com (accessed on 16 February 2025).
Jof 11 00333 g001
Figure 2. Updated dynamics of early-life gut fungal colonization. This figure illustrates the revised pattern of gut fungal colonization in infants, integrating longitudinal data from the recent studies. The updated data indicate that initial colonization occurs more rapidly than previously reported, with significant shifts in fungal diversity observed following dietary transitions and environmental exposures during the first year of life. These findings redefine our understanding of the establishment and maturation of the gut mycobiome compared to earlier studies. Created with Biorender.com (accessed on 20 April 2025).
Figure 2. Updated dynamics of early-life gut fungal colonization. This figure illustrates the revised pattern of gut fungal colonization in infants, integrating longitudinal data from the recent studies. The updated data indicate that initial colonization occurs more rapidly than previously reported, with significant shifts in fungal diversity observed following dietary transitions and environmental exposures during the first year of life. These findings redefine our understanding of the establishment and maturation of the gut mycobiome compared to earlier studies. Created with Biorender.com (accessed on 20 April 2025).
Jof 11 00333 g002
Figure 3. The relationship between the gut mycobiome and the main affected organs, highlighting the gut–brain, gut–lung, gut–liver, gut–kidney, and gut–pancreas axes. At the center of the image, the intestine is depicted as the primary reservoir of fungi, including species such as Candida, Malassezia, and Saccharomyces. Arrows indicate interactions between the gut and different organs. Created with Biorender.com (accessed on 20 April 2025).
Figure 3. The relationship between the gut mycobiome and the main affected organs, highlighting the gut–brain, gut–lung, gut–liver, gut–kidney, and gut–pancreas axes. At the center of the image, the intestine is depicted as the primary reservoir of fungi, including species such as Candida, Malassezia, and Saccharomyces. Arrows indicate interactions between the gut and different organs. Created with Biorender.com (accessed on 20 April 2025).
Jof 11 00333 g003
Figure 4. Different methods for calibrating the gut mycobiome. Most approaches target infections caused by C. albicans, while some studies suggest that certain methods are effective against both C. albicans and other fungal species or exclusively against non-Candida fungi. Created with Biorender.com (accessed on 1 March 2025). CDI = Clostridium difficile infection; IBD = inflammatory bowel disease.
Figure 4. Different methods for calibrating the gut mycobiome. Most approaches target infections caused by C. albicans, while some studies suggest that certain methods are effective against both C. albicans and other fungal species or exclusively against non-Candida fungi. Created with Biorender.com (accessed on 1 March 2025). CDI = Clostridium difficile infection; IBD = inflammatory bowel disease.
Jof 11 00333 g004
Table 1. The most commonly identified fungi genera and their effect on human health and disease.
Table 1. The most commonly identified fungi genera and their effect on human health and disease.
StudyFungiDiseaseEffectRef.
Pfaller et al.C. albicansInvasive fungal infectionExacerbates[46]
Li et al.C. albicansAntibiotic-associated diarrheaExacerbates[47]
Charlet et al.C. glabrataColitisExacerbates[51]
Martino et al.C. tropicalisColitisExacerbates[52]
Hoarau et al.C. tropicalisCDExacerbates[53]
Sun et al.C. parapsilosisDiet-related obesityExacerbates[54]
Sokol et al.C. albicansCD and UCExacerbates[48]
Chester et al.Clavispora lusitaniae (formerly Candida lusitaniae)Digestive and urinary infections
Amphotericin B resistance
Induces[55]
Noor-UI et al.Geotrichum candidum Probiotic that enhances feed utilization, improves immunity, and reduces disease resistance[56]
Abid et al.SaccharomycesTraveler’s diarrhea and CDIProtects[58]
Chen et al.SaccharomycesColon cancerAntineoplastic effects[60]
Spatz et al.MalasseziaDermatitis and pityriasisExacerbates[13]
Limon et al.MalasseziaInflammatory bowel diseaseExacerbates[15]
Sokol et al.MalasseziaInflammatory bowel diseaseExacerbates[48]
Yang et al.MalasseziaCRCUnclear—leads to tumorigenesis[68]
Norlia et al.AspergillusHepatocellular carcinomaHepatotoxic effect[71]
Malir et al.PenicilliumUrothelial carcinomaNephrotoxic effect[72]
CD = Crohn’s disease; UC = ulcerative colitis; CRC = colorectal cancer; C. albicans = Candida albicans; C. glabrata = Candida glabrata ; C. tropicalis = Candida tropicalis; C. parapsilosis = Candida parapsilosis.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Gaspar, B.S.; Roşu, O.A.; Enache, R.-M.; Manciulea, M.; Pavelescu, L.A.; Creţoiu, S.M. Gut Mycobiome: Latest Findings and Current Knowledge Regarding Its Significance in Human Health and Disease. J. Fungi 2025, 11, 333. https://doi.org/10.3390/jof11050333

AMA Style

Gaspar BS, Roşu OA, Enache R-M, Manciulea M, Pavelescu LA, Creţoiu SM. Gut Mycobiome: Latest Findings and Current Knowledge Regarding Its Significance in Human Health and Disease. Journal of Fungi. 2025; 11(5):333. https://doi.org/10.3390/jof11050333

Chicago/Turabian Style

Gaspar, Bogdan Severus, Oana Alexandra Roşu, Robert-Mihai Enache, Monica Manciulea (Profir), Luciana Alexandra Pavelescu, and Sanda Maria Creţoiu. 2025. "Gut Mycobiome: Latest Findings and Current Knowledge Regarding Its Significance in Human Health and Disease" Journal of Fungi 11, no. 5: 333. https://doi.org/10.3390/jof11050333

APA Style

Gaspar, B. S., Roşu, O. A., Enache, R.-M., Manciulea, M., Pavelescu, L. A., & Creţoiu, S. M. (2025). Gut Mycobiome: Latest Findings and Current Knowledge Regarding Its Significance in Human Health and Disease. Journal of Fungi, 11(5), 333. https://doi.org/10.3390/jof11050333

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