Next Article in Journal
Differential Gene Expression Analysis of Whole Blood Transcriptome Between Young and Old Border Collie Dogs
Previous Article in Journal
Effects of Supplementing Rumen-Protected Glutathione on Lactation Performance, Nutrients, Oxidative Stress, Inflammation, and Health in Dairy Cows During the Transition Period
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Gut Barrier Dysfunction and Microbiota Variations in Cryptosporidiosis: A Comprehensive Review

1
College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
2
MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
3
Department of Animal Nutrition, University of Veterinary and Animal Sciences, Lahore 54000, Pakistan
4
Department of Animal Husbandry, University of Agriculture, Faisalabad 38000, Pakistan
*
Authors to whom correspondence should be addressed.
Vet. Sci. 2025, 12(2), 85; https://doi.org/10.3390/vetsci12020085
Submission received: 26 December 2024 / Revised: 21 January 2025 / Accepted: 22 January 2025 / Published: 23 January 2025

Simple Summary

Cryptosporidium, a genus of zoonotic protozoans, poses a threat to one’s health through the disruption of gut microbiota and intestinal barrier functions. This review addresses the underlying mechanisms through which Cryptosporidium spp. impairs the gut barrier, underscoring the importance of junctional proteins (JPs), the mucosal immune response, and host microbiome–parasite interrelations. It also demonstrates how this parasitic infection causes gut microbiota alterations, focusing on the role of short-chain fatty acids (SCFAs) in alleviating infection severity. Therapeutic approaches, e.g., probiotics usage, dietary manipulations, and microbiota transplantation/restoration, are briefly discussed here, necessitating further research using advanced approaches (metagenomics or metabolomics) to control the ever-increasing global burden of cryptosporidiosis.

Abstract

Cryptosporidiosis is a zoonotic protozoan parasite-born disease, equally significant in both animals and humans, especially affecting immunocompromised individuals (e.g., AIDS patients) and neonates. The prime concerns of this review article are to demonstrate the disruption of the intestinal barrier and variations in the gut microbiome during cryptosporidiosis, and to explore host gut–parasite interactions that can lead to the development of novel therapeutics. The review concluded that the enteric barrier is particularly maintained by tight junction proteins (e.g., occludin, claudin, and ZO-1, etc.) and mucosal immunity, both of which are severely compromised during Cryptosporidium spp. infections, resulting in increased intestinal barrier permeability, inflammatory responses, diarrhea, and ultimately death in severe cases. Cryptosporidium-induced dysbiosis is characterized by reduced microbial diversity and richness, a shift from commensal to pathogenic bacteria, as evidenced by increased pro-inflammatory taxa like Proteobacteria, and reduced proportions of beneficial SCFAs producing bacteria, e.g., Firmicutes. Recent investigations have highlighted the interrelations between gut microbiota and epithelial barrier integrity, especially during cryptosporidiosis, demonstrating the modulations regarding tight junctions (TJs), immune reactions, and SCFA production, all of which are main players in alleviating this protozoal parasitic infection. This review comprehensively describes the fine details underlying these impairments, including autophagy-mediated TJs’ degradation, inflammasome activation, and gut microbiome-driven alterations in metabolic pathways, providing the latest relevant, and well-organized piece of knowledge regarding intestinal barrier alterations and microbial shifts during cryptosporidiosis. This work emphasizes the future need for longitudinal studies and advanced sequencing techniques to understand host gut microbiota–parasite interactions, aiming to formulate innovative strategies to mitigate cryptosporidiosis.

1. Introduction

Cryptosporidium, a genus of protozoan parasites, was first identified in the early 20th century [1], and later attracted significant attention due to its increasing prevalence, wide host range, zoonotic potential, and unique characteristics [2]. Cryptosporidium spp. are parasitic organisms, residing intracellularly but separated from the host cell cytoplasm due to their presence in a structure known as the parasitophorous vacuole, infecting the enteric epithelial cells in different vertebrates and resulting in cryptosporidiosis [3,4]. Cryptosporidiosis primarily infects children, newborn animals, and people with compromised immune responses. In low-income regions, cryptosporidiosis is the second leading cause of neonatal diarrhea, resulting in increased proportions of mortality in children under the age of two [5,6]. The interrelations between enterotoxic activity and diarrhea during intestinal cryptosporidiosis manifest through the secretion of an enterotoxic moiety synthesized by Cryptosporidium spp., which leads to lethal diarrhea, as evidenced by the enhanced short-circuit current (Isc) in Caco-2 cells exposed to fecal supernatant from infected individuals, demonstrating significant electrolyte secretion, especially of chloride [7]. Hence, cryptosporidiosis significantly contributes to food and waterborne illnesses, even in the developed world [8,9]. Out of more than 20 Cryptosporidium spp. and genotypes, C. hominis and C. parvum account for more than 90% of cases worldwide [10,11].
Cryptosporidium mostly affects cattle, water buffalo, camels, horses, sheep, goats, poultry, rabbits, pigs, donkeys, deer, wild mammals, and fish [12,13,14,15,16]. The prevalence of Cryptosporidium spp. infection was found to be 28.52% in cattle, 18% in buffalo calves, between 27.8 and 60.4% in pigs, 52.7% in dogs, and 29.4% in cats [17,18]. Recently, C. parvum’s presence in yaks has been reported in China, which may prove to be a serious food-safety threat, ultimately affecting public health [19,20,21]. Before weaning, C. parvum is the most common intestinal pathogen in calves [22]. One infected calf can shed up to 1 × 108 oocysts in feces, posing a risk to other susceptible hosts [23]. Losses due to this disease in the cattle industry include calf mortality, diagnosis expenses, medication and supportive care, and increased market age [24]. The global load of cryptosporidiosis in animal dung is 3.2 × 1023 oocysts per year, to which cattle is a major contributor [25,26]. Additionally, C. parvum is the leading cause of diarrhea in calves and lambs [27]. Hence, Cryptosporidium spp. have a significant zoonotic potential in human beings due to their common interactions with livestock species [28,29,30]. Two investigations in China indicated that C. andersoni showed the highest prevalent Cryptosporidium spp. found in humans [31]. Similarly, C. bovis was initially detected in a 3-year-old toddler and a 23-year-old adult from two different farms in Australia in 2012, who consumed raw milk and had intensive interaction with dairy animals [32]. Another study investigated a combined C. bovis and C. parvum infection in a diarrheic young child aged less than six years from Egypt, who had contact with cattle, resulting in mixed infections [33,34]. Therefore, the potential of cattle to transmit several species of Cryptosporidium spp., particularly C. hominis, represents a major public health hazard associated with possible interactions. Its zoonotic potential is significant worldwide in both the public health and livestock sectors, especially regarding C. parvum IIa subtypes, which are highly prevalent in dairy calves [3,35].
To prevent this protozoan parasite, there is currently no vaccine. Nitazoxanide (NTZ) is the only drug approved by the US Food and Drug Administration (FDA) for treating Cryptosporidium spp. infections [36,37,38]; however, its effectiveness is limited in immunocompromised and malnourished individuals [39,40,41,42]. Paromomycin is another drug that also shows inconsistent therapeutic efficacy in people with compromised immunity [41]. Animals such as goat kids, calves [43,44], neonatal mice in their early life stage [45], and goats are highly vulnerable to Cryptosporidium spp. infection, while in the murine model, it was observed that vulnerability to this protozoan parasite gradually decreases with age [46]. However, the underlying mechanism for lethal diarrhea induced by Cryptosporidium spp. infection still lacks clarity.
The enteric defense mechanism plays an integral role in preserving gut homeostasis and protecting it from notorious pathogens, as evidenced during cryptosporidiosis [47,48,49,50,51]. Cryptosporidium spp. disrupt the integrity of the enteric epithelial barrier by modulating the expression of integral adherens junctions (AJ) and tight junction proteins (TJPs), which result in an enhanced enteric barrier permeability [50]. Contemporary studies have shown that C. parvum infection significantly decreases the key JPs, e.g., E-cadherin and occludin, which are vital to inhibit the systemic invasion of toxins and infectious agents [50,51]. The resulting gut barrier dysfunction not only manifests as diarrhea during cryptosporidiosis, but also impairs immune reactions and overall host health, particularly in highly susceptible populations [2,51]. The exploration of these mechanisms is of great significance to develop potential therapeutics for the restoration of the gut barrier function and to mitigate serious health outcomes post infection.
Cryptosporidium spp. infections affect the gut microbe’s composition, which plays a crucial role in host survival [52,53]. It can also lead to gut dysbiosis, characterized by reduced diversity or an increase in inflammatory or potentially pathogenic bacteria [54,55]. The host may experience long-term effects, including irritable bowel syndrome [53,56]. In addition, reduced bacterial diversity in the host gut is associated with a weakened immune response and the dominance of potentially harmful bacteria [55,57]. The immune system interacts with commensal gut bacteria through nucleotide-binding oligomerization domain (NOD)-like receptors, toll-like receptors (TLRs), and pattern recognition receptors (PRRs) [58], underscoring the role of commensal bacteria in the pathogenesis of diseases [58]. Gut microbiota can influence immune responses, potentially exacerbating or misdirecting them against pathogens like C. parvum, thereby impacting disease severity [58].
This review aims to investigate the dysfunction of different intestinal barrier proteins during Cryptosporidium spp. infection to understand their integral role during cryptosporidiosis. Also, the literature regarding gut microbiota variations during cryptosporidiosis is briefly discussed here. These discussions will provide a guide for future researchers.

2. Methodology

The information presented here was obtained by searching electronic internet databases like SCOPUS, Web of Science, PubMed, and Science Direct, as well as by searching Google Scholar and by visiting specific relevant journals. Various combinations of keywords, such as the Cryptosporidium, tight junction proteins, gut microbiota variations/perturbations, Cryptosporidium and gut barrier integrity, zoonotic transmission, prevalence, and future perspectives, were used. This review mainly included peer-reviewed scientific articles from world-renowned journals written in English, focusing on the most recent and relevant data throughout the manuscript. Initially, 200 articles were downloaded, and later 34 articles were excluded due to their low impact factors, their being old, and to avoid repetition.

3. Protective Role of the Intestinal Barrier and Gut Dysfunctioning in Cryptosporidiosis

3.1. Key Components of the Intestinal Barrier and Their Roles

The enteric epithelium consists of a single layer of cells forming the gut lining, serving an essential function in nutrient absorption, maintaining barrier integrity, and initiating immune reactions. It mainly comprises enteric epithelial cells, e.g., enterocytes, enteroendocrine cells, paneth cells, and goblet cells [47,48,49]. Enterocytes are dominant cells in the intestine and are integral for nutrient absorption. Their increased surface area due to the presence of microvilli on their apical surfaces (brush border) is significant in nutrient absorption and the secretion of enzymes needed for digestion. Also, the presence of goblet cells results in mucus secretion to lubricate and protect the gut mucosal lining [59]. At the base of crypts, the presence of paneth cells ensures the secretion of antimicrobial peptides, which are important players in gut immunity [60]. Enteroendocrine cells are responsible for hormone production and are involved in different physiological processes. It has been observed that there is continuous renewal of the enteric epithelium every 3 to 5 days, linking this process to stem cells located in the crypts of Lieberkühn, guaranteeing the prompt replenishment and repair of intestinal barrier integrity [60,61].
Tight junctions are a special type of intercellular junction positioned at the apical surface of intestinal epithelial cells. They are especially involved in the maintenance of the gut barrier by checking paracellular trafficking. Tight junctions consist of several crucial membrane proteins that interact to form a selective barrier. For instance, claudins are a family of proteins that form the backbone of TJ strands. Different claudins have varying permeability properties, influencing the passage of ions and small molecules. Similarly, occludin is an integral membrane protein that plays a crucial role in the structure and function of TJs, enhancing barrier integrity [62,63,64]. Junctional adhesion molecules (JAMs) facilitate cell–cell adhesion and contribute to the stability of TJs. Tricellulin, located at tricellular junctions where three epithelial cells meet, is essential for maintaining barrier integrity in these complex areas. Additionally, zonula occluden (ZO) proteins, such as ZO-1 and ZO-2, are transmembrane proteins that link TJs to the actin cytoskeleton and various signaling pathways, further supporting the structural and functional integrity of TJs [62,63,64]. This connection is vital for maintaining cell shape and regulating TJs’ dynamics.
Tight junctions prevent the passage of pathogens, toxins, and large molecules from the intestinal lumen into the bloodstream, thus maintaining gut homeostasis [64,65]. They regulate paracellular transport by controlling ions, hence allowing the passage of small molecules. This selectivity is crucial for nutrient absorption while preventing harmful substances from entering systemic circulation [62,63]. They are involved in signaling pathways that influence cell proliferation, differentiation, and immune responses [66]. However, the integrity of TJs can be influenced by various factors, including dietary components, inflammatory cytokines (e.g., IL-1β), and microbial interactions. The disruption of TJs’ integrity has been linked to various gastrointestinal diseases, such as inflammatory bowel disease (IBD), celiac disease, and cryptosporidiosis [60,63,65].

3.2. Enteric Histopathophysiology in Cryptosporidiosis

Cryptosporidium parvum infection causes villus atrophy, with infected tissues showing significantly shorter villi than the control group. Histological studies have showed a significant reduction in intestinal villi length, with up to 50% shortening observed during peak infection in Cryptosporidium-infected animals [50,51,67]. Villi blunting results from parasites attaching to enterocytes, causing dysfunction and eventual apoptosis [51]. Unlike villus atrophy, C. parvum infection triggers crypts’ hyperplasia, with increased crypt depth as a compensatory response to the shortening of villi. This is accompanied by enhanced crypt cell proliferation observed in infected tissues [67,68]. The hyperplastic mechanism’s objective is to regain intestinal absorptive capacity; however, it often fails to fully compensate for the enteric villi architecture loss (Figure 1).
The villus-to-crypt ratio (V:C) is a key indicator of intestinal functionality and health in Cryptosporidium spp. infection; this ratio is greatly altered due to decreased villus height and enhanced crypt depth. Previous studies have demonstrated that infected animals showed a significantly lower V:C ratio than healthy controls, demonstrating compromised intestinal functioning and architecture [51,69], impairing nutrient absorption, and resulting in enhanced intestinal permeability. The process through which Cryptosporidium spp. negatively influence enteric morphology involves epithelial barrier impairment. The parasite binds to the apical surface of intestinal cells, leading to cellular stress that enhances epithelial permeability and promotes enteritis [70,71]. This disruption involves the downregulation of TJPs like E-cadherin, essential for the integrity of enteric epithelium (Figure 2) [51]. Damaged TJs allow pathogens and toxins to cross the epithelial barrier, worsening tissue damage followed by inflammatory cascades.
Cryptosporidium spp. induce strong inflammatory reactions, marked by immune cell infiltrations into the enteric mucosa. Neutrophils and released cytokines like IL-8 contribute to tissue harm and impaired epithelial regeneration [68,69]. The inflammatory mechanism promotes crypt cells’ increment while inducing apoptosis in nearby epithelial cells, playing an integral role in villi blunting [50].

3.3. Cryptosporidium and Gut Barrier Dysfunction

Cryptosporidium parvum infection significantly downregulates primary TJPs, e.g., E-cadherin, claudin-4, and occludins (Figure 2). Previous investigations using monolayers of Caco-2 cells have shown that C. parvum increased intercellular permeability because of the downregulation of TJPs, thereby impairing gut barrier functions. Particularly, claudin-4 and occludin were found to be significantly reduced, and other proteins (junctional), e.g., ZO-1 and claudin-3, also exhibited reduced levels [72]. Tight junctions’ disruption resulted in increased intestinal epithelium permeability, allowing pathogens and toxins to pass into underlying tissues. This was evidenced by an increased flux of fluorescein isothiocyanate (FITC)-dextran across infected Caco-2 cell monolayers, and decreased transepithelial electrical resistance (TEER) measurements [51,73].
Recent investigations have found that C. parvum infection positively influences autophagy in enteric epithelial cells, playing a role in the degradation of TJPs. The initiation of autophagy was associated with the reduced phosphorylation of the mammalian target of rapamycin (mTOR), clarifying the underlying mechanism where autophagy reduces the integrity of TJs via facilitating the degradation of claudin-4 and occludin [51,74]. The silencing of ATG7 (a main player in autophagy) resulted in the upregulation of TJPs, pointing out the role of autophagy in regulating this process. Additionally, the established results suggest the potential role of post-translational modifications in mediating the effects of C. parvum. For example, inhibitors such as bafilomycin-A have been shown to partially alleviate the cytopathic effects of C. parvum on occludin expression, suggesting that this protozoan parasite may trigger pathways responsible for protein degradation, resulting in decreased levels of TJ proteins [51,73].
Cryptosporidium parvum affects TJPs and AJPs, especially E-cadherin, which is integral for intercellular adhesion. During infection, the suppression of E-cadherin aggravates intestinal barrier dysfunction and results in the enhanced permeability of the intestine [51,75], which results in diarrhea during cryptosporidiosis. Additionally, after the initiation of inflammatory cascades, the parasite goes deeper into the tissues, further complicating the pathological cascades. Due to the impairment of intestinal integrity, nutrient absorption is negatively affected, resulting in malnutrition in affected individuals [51,76].

3.4. Inflammatory Cascades: Immunological Reactions Against Cryptosporidiosis

3.4.1. Role of Caspase-1 in Cryptosporidiosis

Caspase-1, a main element of the inflammasome, plays a vital role in the host’s defense against Cryptosporidium spp. infections. In an experimental model where caspase-1 was knocked out in intestinal epithelial cells, an 18-fold parasitic burden was observed compared to wild-type mice. These findings emphasize that epithelial cell intrinsic signaling via caspase-1 is crucial for early control of C. parvum replication [77,78]. In particular, intrinsic activation of caspase-1 in epithelial cells has been shown to positively influence interleukin-18 (IL-18) secretion, which is important for activating immune reactions during cryptosporidiosis [79]. Studies have shown that caspase-1 modulates the expression of the gene in a tissue-dependent fashion within the enteric environment. Especially, it has been demonstrated that caspase-1 can downregulate or upregulate a variety of genes, with special reference to the type of intestinal tissue, e.g., ileum, jejunum, and duodenum. For example, a study examining gene expression in various sections of the intestine revealed that caspase-1 influences 313 common genes’ expression across the mentioned tissues [79]. This underscores the intricate role of caspase-1 in regulating inflammatory cascades during Cryptosporidium spp. infection.
Upon activation, caspase-1 breaks pro-IL-18 and pro-IL-1β into their active forms, which play a critical role in triggering inflammatory cascades. The release of these cytokines is crucial for coordinating the immune response to cryptosporidiosis infection [79,80]. Hence, elevated levels of caspase-1 are associated with increased resistance to cryptosporidiosis [77,81]. Additionally, the external administration of IL-18 restored parasite regulation in caspase-1-deficient mice, further clarifying its role in this context [77]. However, current studies indicate that type I interferon (IFN) signaling may impair the enteric defense against C. parvum. Enteric epithelial cells infected with C. parvum have shown alterations in gene expression patterns linked to type-I IFNs reactions, potentially compromising effective immune defenses against the parasite [81]. Although caspase-1 is essential for triggering protective responses, other signaling mechanisms may complicate host immune responses.

3.4.2. Interactions Between Capsae-1 and NLRP6 in Cryptosporidiosis

Caspase-1 is crucial for the immune response to C. parvum, especially through its interaction with the NOD-like receptor family pyrin domain containing 6 (NLRP6). This interaction is vital for managing intestinal inflammation and enabling the host to control parasitic infections. Current studies have demonstrated the underlying mechanisms through which NLRP6 and caspase-1 aid the immune defense against C. parvum [77]. As an integral receptor, NLRP6 is essential for caspase-1 activation against C. parvum. Studies have shown that NLRP6 is essential for recognizing this parasite by the innate immune response, and that its lack results in enhanced vulnerability to infection. Mice lacking NLRP6 not only have elevated parasite loads, but also demonstrated impaired IL-18 production, which underscores the importance of this pathway in orchestrating an effective immune response(Table 1) [82,83,84]. The epithelial cells are primarily responsible for generating IL-18 in response to C. parvum, highlighting their role as key players in controlling infection [70]. Further studies regarding cellular processes showed that the activation of caspase-1 is especially needed within enteric epithelial cells.
The interrelation between NLRP6 and caspase-1 also impacts various other immune system elements. For example, research suggests that ASC (apoptosis-associated speck-like protein) is a key element of the inflammasome complex and is crucial in influencing susceptibility to cryptosporidiosis. Mice deficient in ASC showed an exaggerated parasite burden during acute phases of infection, but ultimately succeeded in controlling the cryptosporidiosis, indicating the possible involvement of a compensatory process, which needs to be further explored (Table 1) [78,83].

4. Gut Microbiota Alterations in Cryptosporidiosis: Impact and Implications

Cryptosporidium spp. infection in ruminants, humans, and non-human primates has been linked to reduced gut microbiome alpha diversity (Shannon Index). However, certain bacterial genera are known to increase during infection, while little is known about those that are depleted. During infection, key taxa that increased include Firmicutes [100], Pseudomonadota (formerly known as Proteobacteria) [101], different types of protozoa, and Actinomycetota [100]. In the gut ecosystem, Firmicutes are the key players in butyrate production. This phylum comprises Gram-positive bacteria such as Enterococcus, Lactobacillus, Clostridiales, and Lachnospiraceae. Increased butyrate production is linked to healthier, more diverse bacterial communities and a lower hydrogen sulfide concentration [102,103]. Firmicutes significantly increase after birth [104,105], as observed in a study of 20 neonatal calves [105]. This phylum is also considered a cornerstone of a healthy gut microbiome [106]. Another study found an increased abundance of bacteria such as Lactobacillus and members of Coriobacteriaceae in cryptosporidiosis, particularly in the small intestine. Coriobacteriaceae have been exhibited to regulate glucose metabolism [107]. Similarly, an increased level of Lactobacillus may demonstrate a gut microbiota response to compensate for the Cryptosporidium spp.-induced intestinal damage [100].
The Firmicutes/Bacteroidetes ratio is associated with gut flora homeostasis and serves as an indicator of an individual’s health [108]. An increased Firmicutes/Bacteroidetes ratio has been linked to obesity in both murine and human models [108,109], while it has not been linked to cryptosporidiosis severity, presenting a potential area for exploration. Alternatively, gut microbiota-associated metabolites such as fecal indole concentrations, as suggested by Chappell et al. (2016), could serve as biomarkers for susceptibility to this protozoal disease [110]. In this study, patients with higher fecal indole levels resisted C. parvum infection, while those with lower levels were susceptible [110]. Bacteroidota reduces fecal indole levels, indicating that the Firmicutes/Bacteroidetes ratio combined with fecal indole concentration could serve as a reliable biomarker for susceptibility to cryptosporidiosis. During cryptosporidiosis, healthy bacteria like Firmicutes decrease, while pro-inflammatory bacteria such as Proteobacteria often colonize the gut [111,112].
Intestinal infections like C. parvum cause gastroenteritis, which is worsened by pro-inflammatory bacteria colonization. High levels of Proteobacteria are found to be linked to inflammation, and metabolic disorders in bovine calves serve as markers of gut dysbiosis [67]. An increased Proteobacteria abundance during C. parvum indicates reduced gut bacterial diversity. Studies have shown that dietary modifications and probiotics can help reduce the severity of cryptosporidiosis [113,114,115]. After probiotic supplementation, Lactobacillus reduced oocyst output, whereas a mix of Bifidobacter, Streptococcus, and Lactobacillus significantly increased the oocyst output of [114,116], suggesting that outcomes depend upon host type and microbiome composition, creating further research gaps regarding host–microbiome interactions during cryptosporidiosis. Studies have shown that mice on high-fiber diets shed fewer oocysts than those on low-fiber diets, due to changes in microbiota after specific feed types, further clarifying the role of gut microbiota variations due to cryptosporidiosis [115]. Probiotics or high-fiber diets may restore the gut bacteria lost during infection, preventing pro-inflammatory bacterial colonization and alleviating cryptosporidiosis symptoms. These interventions may also support the host immune response against infection [117]. High-fiber diets may promote Firmicutes colonization in the GI tract, as this phylum is involved in converting complex fiber-based polysaccharides into their respective metabolites, such as SCFAs, supporting surrounding bacterial populations [118,119].
Fiber fermentation in the gut is a key indicator of gut health [120]. Still, a high level of fiber is unsuitable for children or neonate animals as a stimulator of beneficial bacteria to cope with cryptosporidiosis; however, probiotics could be an alternative in these cases [121]. The therapeutic effect of probiotics such as Enterococcus faecalis CECT7121 on C. parvum infection could be attributed to competition for binding sites on the gut epithelium, the acidification of the medium induced by lactic acid bacteria [122], an increase in the number of IgA-producing cells, or increased production of IgM [123]. Similarly, Cryptosporidium spp. infection in neonatal rats showed a trend where using probiotics during the infection led to more clearance of cryptosporidiosis; however, daily administration of Lactobacillus casei-containing mixtures in the neonatal rats model did not remove the protozoan [124]. Further research is needed to check the anti-Cryptosporidium effects of different probiotics.
Current studies suggest that dietary changes or probiotics may reduce cryptosporidiosis severity; however, they haven’t established guidelines for clinical use or assessed their potential as a treatment for the infection itself. Previous studies have primarily administered probiotics to the hosts before infection to reduce the severity of cryptosporidiosis. Further research is needed to determine the optimal timing for probiotic use, whether pre-infection to reduce the severity, post infection, or as a preventive measure against chronic gut diseases by rebalancing bacterial diversity [54].

4.1. Interrelations Between Crypsyposridiosis, Gut Microbiota-Derived Metabolites, and Immune Responses

The age of the host is also an important factor in the animal model, and should be considered when discussing gut microbiota variations. VanDussen et al. (2020) indicated that in the murine model, an almost fully established microbiome is formed within three weeks after birth [46]. This is significant, as the microbial alterations during cryptosporidiosis play an important role in gut metabolism. Numerous processes are involved in the Cryptosporidium–microbiome interactions [46].
Recent studies with mice by Charania et al. (2020) also demonstrated that in mice without antibiotics treatment, Cryptosporidium spp. infection increased Lactobacillus communities, whereas antibiotic pre-treatment with cloxacillin led to a decrease [125]. Lactobacillus spp. are known to act as beneficial bacteria and protect against infections, and have been reported to have a direct effect on C. parvum oocyst viability in vitro by producing antimicrobial substances [126]. Notably, the mice in this study exhibited no clinical signs of cryptosporidiosis, suggesting that they may have been asymptomatic carriers of the parasite. In contrast, studies of goats exhibited mild to severe cryptosporidiosis symptoms, e.g., hypothermia, loose feces, inhibited growth, and death, and have reported a depletion of bacterial species involved in SCFA production, disrupting SCFAs’ biosynthetic processes [127]. Short-chain fatty acids (propionate, butyrate, and acetate) are the major byproduct of gut bacteria. They break carbohydrates that are indigestible and are recognized as vital energy materials, have anti-cancer and anti-inflammatory characteristics, can lower cholesterol and fat storage, regulate the pH of the intestine, and also avoid predatory harmful germs from entering and sticking to gut surfaces [128].
Reduced levels of SCFAs in infected hosts have been linked to an altered host microbiome [100,129]. Increased concentrations of SCFAs are crucial metabolites produced by gut bacteria through the fermentation of dietary fibers, which have shown direct inhibitory effects on C. parvum. For instance, in vitro studies demonstrated that butyrate, acetate, and propionate significantly inhibited the growth of C. parvum when administered to infected human intestinal cells [130]. Notably, butyrate not only reduced parasite proliferation, but also increased apoptosis in infected enteric cells, suggesting an underlying mechanism for how increased SCFA concentration inhibits Cryptosporidium spp. infections. Additionally, SCFAs play an important role in maintaining gut epithelial integrity, as the production of mucus and strengthening of TJs between epithelial cells were found to be positively influenced by increased SCFA concentration, hence preventing the invasion of pathogens and maintaining intestinal barrier integrity [87,131].
Also, the production of SCFAs was found to be linked to increased synthesis of anti-inflammatory cytokines and modulation of immune reactions, thereby supporting local and systemic immunity against infections [130]. Short-chain fatty acids affect the activity of different immune cells, e.g., regulatory T cells (Tregs), type 3 innate lymphoid cells (ILC3s), CD4+ T cells, and Th1e ectorcells that generate IFN-γ [132]. IFN-γ is an important immune factor in controlling cryptosporidiosis [133]. It has been proved that in IFN-knockout mice or mice treated with anti-IFN-γ-antibodies, the vulnerability to C. parvum invasion was increased, and, in such mice, the parasitic shed also increased [134]. It has also been observed that mice that were lacking in B and T cells showed reduced C. parvum infection compared to IFN- γ-knockout mice, demonstrating that a part of IFNs during C. parvum infection is developed from non-T and B cells [135]. Experiments with piglets and in vitro cell lines have shown that intestinal epithelial cells secrete a large number of type-III IFNs, independent of specialized immune cells. Initially, the literature indicated that type-III IFNs had been linked with epithelial defense against viruses, but later it was proven that IFNs are involved in defense mechanisms against non-viral infection, also via TLRs [136,137]. Priming epithelial cells of the gut with recombinant IFN-λ3 results in less barrier disruption and enhanced cellular resistance against C. parvum infection [136].
Butyrate regulates gut bacterial ecology, while propionate upregulates NF-κB and IL-18 [138]. Butyrate is also a histone deacetylase inhibitor and is involved in cell cycle and cell proliferation [139]. Interestingly, butyrate, propionate, and acetate are involved in the upregulation of IL-18 [140]. The immunomodulatory effects of SCFAs include upregulating the production of anti-inflammatory mediators while suppressing pro-inflammatory cytokines, thus helping to balance immune responses during cryptosporidiosis [141]. For instance, CD4+ T cells, which are actively influenced by SCFAs, are critical players in eliciting appropriate immunological responses to cryptosporidiosis [142]. CD4+ T immunological cells play an important role in clearing Cryptosporidium spp. throughout the acute phase of an infection (which includes innate immunity) [143]. The first group of CD4+ T helper cells comprises TH17 cells that develop on antigen presentation cells (APCs) that are stimulated by pathogens, and are hence crucial during the initial phases of infection [144]. TH17 cells develop from immature CD4+ T cells in the presence of transforming growth factor beta (TGF-β) and IL-6, and IL-23 promotes TH17 cells to produce IL-17 rather than IL-4 or IFN-γ [144]. IL-17, through its involvement in chemokine and cytokine production, positively influences chemoattraction (for neutrophils) at the infection site, and IL-17 plays a role in immune reaction by facilitating innate immune response against infectious agents [144]. Additionally, TH17 cytokines IL-6, IL-17, IL-23, TNF-α, and TGF-β have been detected in increased concentrations in the GALT (gut associated lymphoid tissues) and spleen of C. parvum-infected immunodeficient BALB/c mice [145].
Lactate metabolizing bacteria actively produce D-amino acids [146], enhancing the microbial response to mitigate mucosal and epithelial damage during cryptosporidiosis [147]. D-amino acids mediate microbiome host cross-talk, [96,97] with producers like D-amino acid-producing species, such as Lachnospiraceae, Marvinbryantia spp., Lactobacillus spp., and Lachnoclostridium spp. in the mouse colon [148,149]. However, the results indicated that during cryptosporidiosis, the microbiome produces high levels of D-amino acids in the small intestine [100]. Infected mice showed increased levels of Lachnoclostridium spp., Lachnospiraceae, and Lactobacillus spp., particularly in duodenum and jejunum, suggesting their role in the production of D-amino acids, consistent with findings by Sasabe et al. (2016) [149].
It was found that during a given parasitic infection, the citrate cycle in the intestine was more active [100]. Cryptosporidium parvum lacks the citrate cycle and relies on salvaging metabolites from the host gut [150]. Research results have demonstrated that microbial carboxylase transporter proteins are implicated in promoting colonization and pathogenicity in bacteria like Salmonella enterica and Haemophilus influenzae. This process utilizes energy sources like glutamate, leading to elevated levels of dicarboxylic acids such as hexanoate and acetate [151]. In this context, Cryptosporidium spp. excystation in the duodenum has been linked to an increased expression of proteins involved in glycolysis, glutaminolysis, and the citrate cycle in the small intestine [152]. In the infected caecum, glutamate metabolism was elevated, likely due to glucose depletion, promoting Cryptosporidium and host defense cells to use glutamate as a primary carbon source. [153,154]. During cryptosporidiosis, glutamate utilization to generate α-ketoglutarate, facilitated by enzymes like glutamate kinase, glutamate-5-semialdehyde dehydrogenase, and glutamine synthetase, was observed [153]. Upregulated host and yeast transketolases, along with yeast polyubiquitin proteins, were detected, suggesting their role in catalyzing ubiquinone biosynthesis in the jejunum–ileum tract [100].
Ubiquinone biosynthesis begins with erythrose 4-phosphate (E4P) metabolism, catalyzed by glucose-6-phosphate dehydrogenase (G6PDH), during infection by trypanosomatid, C. parvum [155], and Plasmodium spp. [156]. Enzyme activities in the yeasts and host indicate a host–parasite–microbiome interaction in the small intestine. This interaction may compensate for Cryptosporidium spp.’s deficient metabolic machinery in ubiquinone (coenzyme Q) synthesis, which is essential for the electron transport chain. Similar associations supporting Cryptosporidium spp.’s multiplication have been reported in aquatic environments [157] and neonatal mice with gut dysbiosis [46]. Elevated yeast ubiquitin-related activity, mediated by ubiquitin-conjugating enzymes, was detected throughout the small intestine, particularly in the ileum. These proteins are essential for ubiquinone’s metabolism, synthesis, and transfer [153,156]. It is believed that Cryptosporidium spp. rely upon the host ubiquinone system for salvage [153].
Also, the host actin plays a crucial role in the invasion and replication of apicomplexan parasites like Cryptosporidium spp. [158]. Previous microscopy and cell culture studies have demonstrated that Cryptosporidium spp. induces the assembly and polymerization of the host actin into plaque structures, facilitating sporozoite invasion [159]. Notably, the upregulation of the actin in response to parasite infection has not been previously reported. In the context of cryptosporidiosis, the increased expression of the actin in the mouse gut is a novel finding that warrants further investigation through targeted proteomic studies.

4.2. Mechanisms Linking Microbiota to Gut Barrier Integrity in Cryptosporidiosis

The established results suggest that certain microbial populations play a vital role in maintaining TJs’ integrity. For example, studies have demonstrated that germ-free mice showed lower expression of occludin and claudin-4, increasing their vulnerability to epithelial injury [160]. Conversely, a diverse microbiome enhances mucin and antimicrobial peptide secretion, strengthening the mucus layer and supporting TJs’ stability [161]. The gut microbiota shapes mucosal immunity through interactions with epithelial cells, which is crucial for balancing immune tolerance and effector functions. For instance, certain bacterial populations boost the production of immunoglobulins (e.g., IgA), aiding in pathogen defense by blocking their adhesion to epithelial cells [162]. During cryptosporidiosis, a disrupted gut microbiome composition leads to dysbiosis, with reduced diversity and a shift in bacterial populations, exacerbating inflammation and weakening mucosal immunity [54,163]. During cryptosporidiosis, altered microbiota increased intestinal permeability and weakened immune responses, emphasizing the connection between microbial balance, health, and host defense [54,100]. Cryptosporidiosis disrupts the gut microbiome, increasing pathogenic bacteria while reducing beneficial species. This dysbiosis drives persistent inflammation and hampers recovery [100,163]. Additionally, altered SCFA production during cryptosporidiosis impacts epithelial integrity and immune response modulation, further exacerbating disease severity [100].

5. Future Research Directions

Future researchers should use high-throughput sequencing techniques to characterize the gut microbiota composition before and after Cryptosporidium spp. infection. This can lead to the identification of specific microbial communities and their relationship to health, disease severity, and outcomes. Current research indicates that increased diversity in the gut microbiome can potentially diminish clinical signs and oocyst shedding during infection [164]. Additionally, research should also explore the specific processes and micro-details through which Cryptosporidium spp. induced impairments in TJs’ integrity. Uncovering how the parasite interacts with host enteric epithelial cells and modulates inflammatory cascades can lead to the discovery of therapeutic agents. Previous studies have proven that Cryptosporidium spp. shift gut microbiota, worsening barrier dysfunctions [163].
Research has also demonstrated that infection reduces SCFA levels, contributing to dysbiosis and dysfunction of the intestinal barrier [100]. Future research could explore the effect of dietary interventions to boost SCFA production and the restoration of gut health after infection. Also, the potential of fecal microbiota transplant (FMT) to manage chronic cryptosporidiosis warrants evaluation. Clinical trials are needed to evaluate interventions for restoring microbial balance and improving outcomes during infection [100]. Chronic cryptosporidiosis in individuals with compromised immunity underscores the need for treatments targeting gut microbiome modulations [165]. Dietary modifications to enhance gut barrier functions during Cryptosporidium spp. infection offer a promising research avenue. Particular attention should be given to high-fiber diets, which have been demonstrated to improve microbial diversity and mucosal immunity [115]. Investigating how dietary components affect microbiota composition and host responses during infection could provide valuable future research directions.
Longitudinal studies in vulnerable communities like immunocompromised patients and children are crucial for understanding the long-term effects of cryptosporidiosis on microbiota variations and intestinal barrier integrity. This will aid in developing targeted interventions for chronic and recurrent infections [166].

6. Conclusions

Cryptosporidium spp. are a significant threat to both public health and the livestock sector because of severe damage to intestinal barrier integrity and reduced gut commensal microbes. This review has demonstrated the multi-dimensional impairments from cryptosporidiosis, e.g., the downregulation of TJPs, initiation of autophagy, and inflammatory mechanisms that negatively affect gut barrier functions. Also, the infection leads to variations in the gut microbiome and dysbiosis, negatively influencing the SCFA-producing bacteria (e.g., Firmicutes) and increasing the relative abundance of pathogenic bacteria, e.g., Proteobacteria. Innovative therapeutic options, e.g., probiotics supplementation, fecal microbiota transplantation, and dietary interventions, can prove promising in alleviating the hazardous effects of cryptosporidiosis. However, for their practical applications, further evidence-based protocols are needed. Future researchers should explore the intricate interrelations between the parasite and host microbiome through metabolomics and metagenomics analysis, using high-throughput sequencing to explore novel treatment options against cryptosporidiosis. Overall, addressing cryptosporidiosis from the perspective of the One Health approach, with special emphasis on interactions between animals, humans, and environmental health, is crucial to reduce the global burden of this parasitic disease.

Author Contributions

Conceptualization, K.L., Y.Q. and M.A.; methodology K.L. and M.A.; software, K.L. and M.A.; validation, K.L. and M.A.; formal analysis, K.L. and M.A.; investigation, K.L. and M.A.; resources, K.L.; data curation, K.L., M.A. and C.X.; writing—original draft preparation, K.L., Y.Q. and M.A.; writing—review and editing, M.A., C.X., M.W., Q.H. (Qazal Hina), Y.J., S.A., S.L., Q.H. (Qing He)., Y.Q. and K.L.; visualization, K.L., M.A., C.X., Q.H., S.A. and Y.J.; project administration, Y.Q. and K.L.; funding acquisition, K.L. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Natural Science Foundation of China (32102692).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study because it is a comprehensive review.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Leitch, G.J.; He, Q. Cryptosporidiosis—An Overview. J. Biomed. Res. 2012, 25, 1–16. [Google Scholar] [CrossRef] [PubMed]
  2. Gerace, E.; Presti, V.D.M.L.; Biondo, C. Cryptosporidium Infection: Epidemiology, Pathogenesis, and Differential Diagnosis. Eur. J. Microbiol. Immunol. 2019, 9, 119–123. [Google Scholar] [CrossRef] [PubMed]
  3. Guo, Y.; Ryan, U.; Feng, Y.; Xiao, L. Emergence of Zoonotic Cryptosporidium parvum in China. Trends Parasitol. 2022, 38, 335–343. [Google Scholar] [CrossRef]
  4. Checkley, W.; White, A.C.; Jaganath, D.; Arrowood, M.J.; Chalmers, R.M.; Chen, X.-M.; Fayer, R.; Griffiths, J.K.; Guerrant, R.L.; Hedstrom, L.; et al. A Review of the Global Burden, Novel Diagnostics, Therapeutics, and Vaccine Targets for Cryptosporidium. Lancet Infect. Dis. 2015, 15, 85–94. [Google Scholar] [CrossRef] [PubMed]
  5. Kotloff, K.L.; Nataro, J.P.; Blackwelder, W.C.; Nasrin, D.; Farag, T.H.; Panchalingam, S.; Wu, Y.; Sow, S.O.; Sur, D.; Breiman, R.F.; et al. Burden and Aetiology of Diarrhoeal Disease in Infants and Young Children in Developing Countries (the Global Enteric Multicenter Study, GEMS): A Prospective, Case-Control Study. Lancet 2013, 382, 209–222. [Google Scholar] [CrossRef]
  6. Moore, S.R.; Lima, N.L.; Soares, A.M.; Oriá, R.B.; Pinkerton, R.C.; Barrett, L.J.; Guerrant, R.L.; Lima, A.A.M. Prolonged Episodes of Acute Diarrhea Reduce Growth and Increase Risk of Persistent Diarrhea in Children. Gastroenterology 2010, 139, 1156–1164. [Google Scholar] [CrossRef] [PubMed]
  7. Guarino, A.; Canani, R.B.; Casola, A.; Pozio, E.; Russo, R.; Bruzzese, E.; Fontana, M.; Rubino, A. Human Intestinal Cryptosporidiosis: Secretory Diarrhea and Enterotoxic Activity in Caco-2 Cells. J. Infect. Dis. 1995, 171, 976–983. [Google Scholar] [CrossRef] [PubMed]
  8. Javed, K.; Alkheraije, K.A. Cryptosporidiosis: A Foodborne Zoonotic Disease of Farm Animals and Humans. Pak. Vet. J. 2023, 43, 213–223. [Google Scholar] [CrossRef]
  9. Ali, M.; Ji, Y.; Xu, C.; Hina, Q.; Javed, U.; Li, K. Food and Waterborne Cryptosporidiosis from a One Health Perspective: A Comprehensive Review. Animals 2024, 14, 3287. [Google Scholar] [CrossRef]
  10. Ayan, A.; Celik, B.A.; Celik, O.Y.; Akyildiz, G.; Kilinc, O.O.; Ayan, O.O.; Oguz, F.E.; Goz, Y.; Yuksek, N.; Yilmaz, A.B.; et al. First Report of Zoonotic Cryptosporidium parvum Subtype IIaA15G2R1 in Dogs in Türkiye. Pak. Vet. J. 2024, 44, 1263–1268. [Google Scholar] [CrossRef]
  11. Feng, Y.; Ryan, U.M.; Xiao, L. Genetic Diversity and Population Structure of Cryptosporidium. Trends Parasitol. 2018, 34, 997–1011. [Google Scholar] [CrossRef]
  12. Golomazou, E.; Mamedova, S.; Eslahi, A.V.; Karanis, P. Cryptosporidium and Agriculture: A Review. Sci. Total Environ. 2024, 916, 170057. [Google Scholar] [CrossRef]
  13. Puleston, R.L.; Mallaghan, C.M.; Modha, D.E.; Hunter, P.R.; Nguyen-Van-Tam, J.S.; Regan, C.M.; Nichols, G.L.; Chalmers, R.M. The First Recorded Outbreak of Cryptosporidiosis Due to Cryptosporidium cuniculus (Formerly Rabbit Genotype), Following a Water Quality Incident. J. Water Health 2014, 12, 41–50. [Google Scholar] [CrossRef] [PubMed]
  14. Wells, B.; Shaw, H.; Hotchkiss, E.; Gilray, J.; Ayton, R.; Green, J.; Katzer, F.; Wells, A.; Innes, E. Prevalence, Species Identification and Genotyping Cryptosporidium from Livestock and Deer in a Catchment in the Cairngorms with a History of a Contaminated Public Water Supply. Parasites Vectors 2015, 8, 66. [Google Scholar] [CrossRef] [PubMed]
  15. Zahedi, A.; Monis, P.; Gofton, A.W.; Oskam, C.L.; Ball, A.; Bath, A.; Bartkow, M.; Robertson, I.; Ryan, U. Cryptosporidium species and Subtypes in Animals Inhabiting Drinking Water Catchments in Three States across Australia. Water Res. 2018, 134, 327–340. [Google Scholar] [CrossRef] [PubMed]
  16. Certad, G.; Follet, J.; Gantois, N.; Hammouma-Ghelboun, O.; Guyot, K.; Benamrouz-Vanneste, S.; Fréalle, E.; Seesao, Y.; Delaire, B.; Creusy, C.; et al. Prevalence, Molecular Identification, and Risk Factors for Cryptosporidium Infection in Edible Marine Fish: A Survey Across Sea Areas Surrounding France. Front. Microbiol. 2019, 10, 1037. [Google Scholar] [CrossRef]
  17. Alarcon-Zapata, M.A.; Romero-Salas, D.; Chaparro-Gutierrez, J.J.; Gonzalez-Hernandez, M.; Ojeda-Chi, M.M.; Arturo, S.-S. Frequency of Giardia spp. and Cryptosporidium spp. in Domestic and Captive Wild Animals in the North of Veracruz, Mexico. Pak. Vet. J. 2023, 43, 814–818. [Google Scholar] [CrossRef]
  18. Dărăbuș, G.; Lupu, M.A.; Mederle, N.; Dărăbuș, R.G.; Imre, K.; Mederle, O.; Imre, M.; Paduraru, A.A.; Morariu, S.; Olariu, T.R. Epidemiology of Cryptosporidium Infection in Romania: A Review. Microorganisms 2023, 11, 1793. [Google Scholar] [CrossRef]
  19. Cidan, Y.; Lu, S.; Wang, H.; Wang, J.; Ali, M.; Fouad, D.; Ataya, F.S.; Zhu, Y.; Basang, W.; Li, K. Comparative Analysis of Microbiota in Jiani Yaks with Different Rib Structures. Life 2024, 11, 1458. [Google Scholar] [CrossRef]
  20. Peng, S.; Xu, C.; Saleem, M.U.; Babar, W.; Idrees, A.; Li, K. Epidemiological Investigation of Cryptosporidium Infection in Yaks in Chamdo, China. Pak. Vet. J. 2024, 44, 526–529. [Google Scholar] [CrossRef]
  21. Qi, M.; Cai, J.; Wang, R.; Li, J.; Jian, F.; Huang, J.; Zhou, H.; Zhang, L. Molecular Characterization of Cryptosporidium spp. and Giardia duodenalis from Yaks in the Central Western Region of China. BMC Microbiol. 2015, 15, 108. [Google Scholar] [CrossRef]
  22. Cho, Y.-I.; Han, J.-I.; Wang, C.; Cooper, V.; Schwartz, K.; Engelken, T.; Yoon, K.-J. Case–Control Study of Microbiological Etiology Associated with Calf Diarrhea. Vet. Microbiol. 2013, 166, 375–385. [Google Scholar] [CrossRef]
  23. Nydam, D.V.; Wade, S.E.; Schaaf, S.L.; Mohammed, H.O. Number of Cryptosporidium parvum Oocysts or Giardia spp. Cysts Shed by Dairy Calves after Natural Infection. Am. J. Vet. Res. 2001, 62, 1612–1615. [Google Scholar] [CrossRef]
  24. Olson, M.E.; O’Handley, R.M.; Ralston, B.J.; McAllister, T.A.; Thompson, R.C.A. Update on Cryptosporidium and Giardia Infections in Cattle. Trends Parasitol. 2004, 20, 185–191. [Google Scholar] [CrossRef] [PubMed]
  25. Morgan, U.M.; Monis, P.T.; Xiao, L.; Limor, J.R.; Sulaiman, I.M.; Raidal, S.; Donoghue, P.O.; Gasser, R.B.; Murray, A.; Fayer, R.; et al. Molecular Characterization and Phylogenetic Analysis of 18S RRNA, Gp60 and HSP70 Genes of Cryptosporidium parvum Isolated from Cattle Owners and Cattle Using Nested PCR. Pak. Vet. J. 2024, 31, 289–296. [Google Scholar] [CrossRef]
  26. Vermeulen, L.C.; Benders, J.; Medema, G.; Hofstra, N. Global Cryptosporidium Loads from Livestock Manure. Environ. Sci. Technol. 2017, 51, 8663–8671. [Google Scholar] [CrossRef]
  27. Xiao, L.; Fayer, R.; Ryan, U.; Upton, S.J. Cryptosporidium Taxonomy: Recent Advances and Implications for Public Health. Clin. Microbiol. Rev. 2004, 17, 72–97. [Google Scholar] [CrossRef] [PubMed]
  28. Zahedi, A.; Ryan, U. Cryptosporidium—An Update with an Emphasis on Foodborne and Waterborne Transmission. Res. Vet. Sci. 2020, 132, 500–512. [Google Scholar] [CrossRef]
  29. Guo, Y.; Li, N.; Ryan, U.; Feng, Y.; Xiao, L. Small Ruminants and Zoonotic Cryptosporidiosis. Parasitol. Res. 2021, 120, 4189–4198. [Google Scholar] [CrossRef] [PubMed]
  30. Santin, M. Cryptosporidium and Giardia in Ruminants. Vet. Clin. N. Am. Food Anim. Pract. 2020, 36, 223–238. [Google Scholar] [CrossRef] [PubMed]
  31. Jiang, Y.; Ren, J.; Yuan, Z.; Liu, A.; Zhao, H.; Liu, H.; Chu, L.; Pan, W.; Cao, J.; Lin, Y. Cryptosporidium andersoni as a Novel Predominant Cryptosporidium Species in Outpatients with Diarrhea in Jiangsu Province, China. BMC Infect. Dis. 2014, 14, 555. [Google Scholar] [CrossRef] [PubMed]
  32. Ng, J.S.Y.; Eastwood, K.; Walker, B.; Durrheim, D.N.; Massey, P.D.; Porigneaux, P.; Kemp, R.; McKinnon, B.; Laurie, K.; Miller, D. Evidence of Cryptosporidium Transmission Between Cattle and Humans in Northern New South Wales. Exp. Parasitol. 2012, 130, 437–441. [Google Scholar] [CrossRef]
  33. Helmy, Y.A.; Krücken, J.; Nöckler, K.; von Samson-Himmelstjerna, G.; Zessin, K.-H. Molecular Epidemiology of Cryptosporidium in Livestock Animals and Humans in the Ismailia Province of Egypt. Vet. Parasitol. 2013, 193, 15–24. [Google Scholar] [CrossRef] [PubMed]
  34. Shehata, A.A.; El-Emam, M.M.A.; Gouda, H.; El-Said, B.M.; Salman, M.B.; Abd-Elfatah, E.B. Molecular Characterization of Cryptosporidium Infection and Analysis of Hematological and Biochemical Changes in Diarrheic Pre-Weaned Calves in Egypt. Pak. Vet. J. 2024, 44, 135–140. [Google Scholar] [CrossRef]
  35. Ryan, U.M.; Feng, Y.; Fayer, R.; Xiao, L. Taxonomy and Molecular Epidemiology of Cryptosporidium and Giardia—A 50 Year Perspective (1971–2021). Int. J. Parasitol. 2021, 51, 1099–1119. [Google Scholar] [CrossRef] [PubMed]
  36. Wang, B.; Castellanos-Gonzalez, A.; White, A.C., Jr. Novel Drug Targets for Treatment of Cryptosporidiosis. Expert Opin. Ther. Targets 2020, 24, 915–922. [Google Scholar] [CrossRef]
  37. Ali, M.; Xu, C.; Wang, J.; Kulyar, M.F.-E.; Li, K. Emerging Therapeutic Avenues Against Cryptosporidium: A Comprehensive Review. Vet. Parasitol. 2024, 331, 110279. [Google Scholar] [CrossRef] [PubMed]
  38. Ali, M.; Xu, C.; Nawaz, S.; Ahmed, A.E.; Hina, Q.; Li, K. Anti-Cryptosporidial Drug-Discovery Challenges and Existing Therapeutic Avenues: A “One-Health” Concern. Life 2024, 14, 80. [Google Scholar] [CrossRef] [PubMed]
  39. Ventura, G.; Cauda, R.; Larocca, L.M.; Riccioni, M.E.; Tumbarello, M.; Lucia, M.B. Gastric Cryptosporidiosis Complicating HIV Infection: Case Report and Review of the Literature. Eur. J. Gastroenterol. Hepatol. 1997, 9, 307–310. [Google Scholar] [CrossRef]
  40. Amadi, B.; Mwiya, M.; Sianongo, S.; Payne, L.; Watuka, A.; Katubulushi, M.; Kelly, P. High Dose Prolonged Treatment with Nitazoxanide Is Not Effective for Cryptosporidiosis in HIV Positive Zambian Children: A Randomised Controlled Trial. BMC Infect. Dis. 2009, 9, 195. [Google Scholar] [CrossRef] [PubMed]
  41. Hussien, S.M.M.; Abdella, O.H.; Abuhashim, A.H.; Aboshiesha, G.A.; Taha, M.A.A.; El-Shemy, A.S.; El-Bader, M.M. Comparative Study Between the Effect of Nitazoxanide and Paromomycine in Treatment of Cryptosporidiosis in Hospitalized Children. J. Egypt. Soc. Parasitol. 2013, 43, 463–470. [Google Scholar]
  42. Sparks, H.; Nair, G.; Castellanos-Gonzalez, A.; White, A.C. Treatment of Cryptosporidium: What We Know, Gaps, and the Way Forward. Curr. Trop. Med. Rep. 2015, 2, 181–187. [Google Scholar] [CrossRef]
  43. Gormley, F.J.; Little, C.L.; Chalmers, R.M.; Rawal, N.; Adak, G.K. Zoonotic Cryptosporidiosis from Petting Farms, England and Wales, 1992–2009. Emerg. Infect. Dis. 2011, 17, 151. [Google Scholar] [CrossRef]
  44. Lee, S.; Alkathiri, B.; Kim, A.-Y.; Do, K.-H.; Seo, K.; Kim, S.; Lee, W.-K.; Kwak, D.; Lee, S.-H. Alteration of Goat Kids’ Gut Microbiota by Cryptosporidium parvum Infection. J. Biomed. Transl. Res. 2024, 25, 41–52. [Google Scholar] [CrossRef]
  45. Sherwood, D.; Angus, K.W.; Snodgrass, D.R.; Tzipori, S. Experimental Cryptosporidiosis in Laboratory Mice. Infect. Immun. 1982, 38, 471–475. [Google Scholar] [CrossRef] [PubMed]
  46. VanDussen, K.L.; Funkhouser-Jones, L.J.; Akey, M.E.; Schaefer, D.A.; Ackman, K.; Riggs, M.W.; Stappenbeck, T.S.; Sibley, L.D. Neonatal Mouse Gut Metabolites Influence Cryptosporidium parvum Infection in Intestinal Epithelial Cells. MBio 2020, 11, 10–1128. [Google Scholar] [CrossRef] [PubMed]
  47. Kong, S.; Zhang, Y.H.; Zhang, W. Regulation of Intestinal Epithelial Cells Properties and Functions by Amino Acids. Biomed Res. Int. 2018, 2018, 2819154. [Google Scholar] [CrossRef] [PubMed]
  48. Chelakkot, C.; Ghim, J.; Ryu, S.H. Mechanisms Regulating Intestinal Barrier Integrity and Its Pathological Implications. Exp. Mol. Med. 2018, 50, 1–9. [Google Scholar] [CrossRef]
  49. Ramanan, D.; Cadwell, K. Intrinsic Defense Mechanisms of the Intestinal Epithelium. Cell Host Microbe 2016, 19, 434–441. [Google Scholar] [CrossRef] [PubMed]
  50. Kumar, A.; Chatterjee, I.; Anbazhagan, A.N.; Jayawardena, D.; Priyamvada, S.; Alrefai, W.A.; Sun, J.; Borthakur, A.; Dudeja, P.K. Cryptosporidium parvum Disrupts Intestinal Epithelial Barrier Function via Altering Expression of Key Tight Junction and Adherens Junction Proteins. Cell. Microbiol. 2018, 20, e12830. [Google Scholar] [CrossRef]
  51. Luo, C.; Xu, Y.; Zhang, J.; Tian, Q.; Guo, Y.; Li, N.; Feng, Y.; Xu, R.; Xiao, L. Cryptosporidium parvum Disrupts Intestinal Epithelial Barrier in Neonatal Mice Through Downregulation of Cell Junction Molecules. PLoS Neglected Trop. Dis. 2024, 18, e0012212. [Google Scholar] [CrossRef] [PubMed]
  52. Clancy, J.L.; Bukhari, Z.; Hargy, T.M.; Bolton, J.R.; Dussert, B.W.; Marshall, M.M. Using UV to Inactivate Cryptosporidium. J.-Am. Water Work. Assoc. 2000, 92, 97–104. [Google Scholar] [CrossRef]
  53. Chi, J.; Sun, L.; Cai, L.; Fan, L.; Shao, C.; Shang, L.; Zhao, Y. Chinese Herb Microneedle Patch for Wound Healing. Bioact. Mater. 2021, 6, 3507–3514. [Google Scholar] [CrossRef] [PubMed]
  54. Hurle, G.R.; Brainard, J.; Tyler, K.M. Microbiome Diversity Is a Modifiable Virulence Factor for Cryptosporidiosis. Virulence 2023, 14, 2273004. [Google Scholar] [CrossRef]
  55. Belizário, J.E.; Faintuch, J.; Garay-Malpartida, M. Gut Microbiome Dysbiosis and Immunometabolism: New Frontiers for Treatment of Metabolic Diseases. Mediat. Inflamm. 2018, 2018, 2037838. [Google Scholar] [CrossRef] [PubMed]
  56. Grigor’eva, I.N. Gallstone Disease, Obesity and the Firmicutes/Bacteroidetes Ratio as a Possible Biomarker of Gut Dysbiosis. J. Pers. Med. 2020, 11, 13. [Google Scholar] [CrossRef]
  57. Wieërs, G.; Verbelen, V.; Van Den Driessche, M.; Melnik, E.; Vanheule, G.; Marot, J.-C.; Cani, P.D. Do Probiotics During In-Hospital Antibiotic Treatment Prevent Colonization of Gut Microbiota with Multi-Drug-Resistant Bacteria? A Randomized Placebo-Controlled Trial Comparing Saccharomyces to a Mixture of Lactobacillus, Bifidobacterium, and Saccharomyce. Front. Public Health 2021, 8, 578089. [Google Scholar] [CrossRef] [PubMed]
  58. Laurent, F.; Lacroix-Lamandé, S. Innate Immune Responses Play a Key Role in Controlling Infection of the Intestinal Epithelium by Cryptosporidium. Int. J. Parasitol. 2017, 47, 711–721. [Google Scholar] [CrossRef] [PubMed]
  59. Gustafsson, J.K.; Johansson, M.E.V. The Role of Goblet Cells and Mucus in Intestinal Homeostasis. Nat. Rev. Gastroenterol. Hepatol. 2022, 19, 785–803. [Google Scholar] [CrossRef] [PubMed]
  60. Ali, A.; Tan, H.; Kaiko, G.E. Role of the Intestinal Epithelium and Its Interaction with the Microbiota in Food Allergy. Front. Immunol. 2020, 11, 604054. [Google Scholar] [CrossRef]
  61. Régnier, M.; Van Hul, M.; Knauf, C.; Cani, P.D. Gut Microbiome, Endocrine Control of Gut Barrier Function and Metabolic Diseases. J. Endocrinol. 2021, 248, R67–R82. [Google Scholar] [CrossRef]
  62. Suzuki, T. Regulation of the Intestinal Barrier by Nutrients: The Role of Tight Junctions. Anim. Sci. J. 2020, 91, e13357. [Google Scholar] [CrossRef] [PubMed]
  63. Kaminsky, L.W.; Al-Sadi, R.; Ma, T.Y. IL-1β and the Intestinal Epithelial Tight Junction Barrier. Front. Immunol. 2021, 12, 767456. [Google Scholar] [CrossRef] [PubMed]
  64. Gimenes, G.M.; Pereira, J.N.B.; da Silva, E.B.; Dos Santos, A.A.C.; Rodrigues, T.M.; de Oliveira Santana, G.; Scervino, M.V.M.; Pithon-Curi, T.C.; Hirabara, S.M.; Gorjão, R. Intestinal Motility Dysfunction in Goto-Kakizaki Rats: Role of the Myenteric Plexus. Cells 2024, 13, 1626. [Google Scholar] [CrossRef] [PubMed]
  65. Di Tommaso, N.; Gasbarrini, A.; Ponziani, F.R. Intestinal Barrier in Human Health and Disease. Int. J. Environ. Res. Public Health 2021, 18, 12836. [Google Scholar] [CrossRef]
  66. Horowitz, A.; Chanez-Paredes, S.D.; Haest, X.; Turner, J.R. Paracellular Permeability and Tight Junction Regulation in Gut Health and Disease. Nat. Rev. Gastroenterol. Hepatol. 2023, 20, 417–432. [Google Scholar] [CrossRef] [PubMed]
  67. Brainard, J.; Hammer, C.C.; Hunter, P.R.; Katzer, F.; Hurle, G.; Tyler, K. Efficacy of Halofuginone Products to Prevent or Treat Cryptosporidiosis in Bovine Calves: A Systematic Review and Meta-Analyses. Parasitology 2021, 148, 408–419. [Google Scholar] [CrossRef]
  68. Chen, X.-M. Human Intestinal and Biliary Cryptosporidiosis. World J. Gastroenterol. 1999, 5, 424. [Google Scholar] [CrossRef]
  69. Dengler, F.; Hammon, H.M.; Liermann, W.; Görs, S.; Bachmann, L.; Helm, C.; Ulrich, R.; Delling, C. Cryptosporidium parvum Competes with the Intestinal Epithelial Cells for Glucose and Impairs Systemic Glucose Supply in Neonatal Calves. Vet. Res. 2023, 54, 40. [Google Scholar] [CrossRef]
  70. Wallbank, B.A.; Pardy, R.D.; Brodsky, I.E.; Hunter, C.A.; Striepen, B. Cryptosporidium Impacts Epithelial Turnover and Is Resistant to Induced Death of the Host Cell. MBio 2024, 15, e01720-24. [Google Scholar] [CrossRef]
  71. Abo-Mandil, M.; Alshahat, S.; El-Badry, A.; El-Sheety, A.; El-Faramawy, M.; Ismael, N. Genotypic Prevalence of Cryptosporidium in Egyptian Patients with Liver Cirrhosis. Al-Azhar Int. Med. J. 2020, 1, 225–231. [Google Scholar] [CrossRef]
  72. Schlingmann, B.; Overgaard, C.E.; Molina, S.A.; Lynn, K.S.; Mitchell, L.A.; Dorsainvil White, S.; Mattheyses, A.L.; Guidot, D.M.; Capaldo, C.T.; Koval, M. Regulation of Claudin/Zonula Occludens-1 Complexes by Hetero-Claudin Interactions. Nat. Commun. 2016, 7, 12276. [Google Scholar] [CrossRef] [PubMed]
  73. Esmat, M.; Abdel-Aal, A.A.; Shalaby, M.A.; Badawi, M.; Elaskary, H.; Yousif, A.B.; Fahmy, M.-E.A. Efficacy of Clofazimine and Nitazoxanide Combination in Treating Intestinal Cryptosporidiosis and Enhancing Intestinal Cellular Regeneration in Immunocompromised Mice. Food Waterborne Parasitol. 2022, 27, e00161. [Google Scholar] [CrossRef]
  74. Lamisere, H.; Bhalchandra, S.; Kane, A.V.; Zeng, X.-L.; Mo, D.; Adams, W.; Estes, M.K.; Ward, H.D. Differential Response to the Course of Cryptosporidium parvum Infection and Its Impact on Epithelial Integrity in Differentiated Versus Undifferentiated Human Intestinal Enteroids. Infect. Immun. 2022, 90, e00397-22. [Google Scholar] [CrossRef]
  75. Wang, L.; Cui, Z.; Li, N.; Liang, G.; Zhang, X.; Wang, Y.; Li, D.; Li, X.; Zhang, S.; Zhang, L. Comparative Proteomics Reveals Cryptosporidium parvum Infection Disrupts Cellular Barriers. J. Proteom. 2023, 287, 104969. [Google Scholar] [CrossRef] [PubMed]
  76. DiGuilio, K.M.; Rybakovsky, E.; Abdavies, R.; Chamoun, R.; Flounders, C.A.; Shepley-McTaggart, A.; Harty, R.N.; Mullin, J.M. Micronutrient Improvement of Epithelial Barrier Function in Various Disease States: A Case for Adjuvant Therapy. Int. J. Mol. Sci. 2022, 23, 2995. [Google Scholar] [CrossRef] [PubMed]
  77. Sateriale, A.; Gullicksrud, J.A.; Engiles, J.B.; McLeod, B.I.; Kugler, E.M.; Henao-Mejia, J.; Zhou, T.; Ring, A.M.; Brodsky, I.E.; Hunter, C.A. The Intestinal Parasite Cryptosporidium Is Controlled by an Enterocyte Intrinsic Inflammasome That Depends on NLRP6. Proc. Natl. Acad. Sci. USA 2021, 118, e2007807118. [Google Scholar] [CrossRef]
  78. Raupach, B.; Peuschel, S.-K.; Monack, D.M.; Zychlinsky, A. Caspase-1-Mediated Activation of Interleukin-1β (IL-1β) and IL-18 Contributes to Innate Immune Defenses Against Salmonella enterica Serovar Typhimurium Infection. Infect. Immun. 2006, 74, 4922–4926. [Google Scholar] [CrossRef] [PubMed]
  79. Li, Y.; Nanayakkara, G.; Sun, Y.; Li, X.; Wang, L.; Cueto, R.; Shao, Y.; Fu, H.; Johnson, C.; Cheng, J.; et al. Analyses of Caspase-1-Regulated Transcriptomes in Various Tissues Lead to Identification of Novel IL-1β-, IL-18- and Sirtuin-1-Independent Pathways. J. Hematol. Oncol. 2017, 10, 40. [Google Scholar] [CrossRef] [PubMed]
  80. Peng, L.; Zhu, N.; Wang, D.; Zhou, Y.; Liu, Y. Comprehensive Analysis of Prognostic Value and Immune Infiltration of NLRC4 and CASP1 in Colorectal Cancer. Int. J. Gen. Med. 2022, 15, 5425–5440. [Google Scholar] [CrossRef]
  81. Deng, S.; He, W.; Gong, A.-Y.; Li, M.; Wang, Y.; Xia, Z.; Zhang, X.-T.; Huang Pacheco, A.S.; Naqib, A.; Jenkins, M.; et al. Cryptosporidium Uses CSpV1 to Activate Host Type I Interferon and Attenuate Antiparasitic Defenses. Nat. Commun. 2023, 14, 1456. [Google Scholar] [CrossRef] [PubMed]
  82. Ali, M.; Xu, C.; Ji, Y.; Li, K. Host Immune Response to Cryptosporidium spp.: Insights and Perspectives for Vaccine Development. Anim. Zoonoses. 2025. [Google Scholar]
  83. Mead, J.R. Early Immune and Host Cell Responses to Cryptosporidium Infection. Front. Parasitol. 2023, 2, 1113950. [Google Scholar] [CrossRef]
  84. Cohn, I.S.; Henrickson, S.E.; Striepen, B.; Hunter, C.A. Immunity to Cryptosporidium: Lessons from Acquired and Primary Immunodeficiencies. J. Immunol. 2022, 209, 2261–2268. [Google Scholar] [CrossRef] [PubMed]
  85. Veshkini, A.; Kühn, C.; Dengler, F.; Bachmann, L.; Liermann, W.; Helm, C.; Ulrich, R.; Delling, C.; Hammon, H.M. Cryptosporidium parvum Infection Alters the Intestinal Mucosa Transcriptome in Neonatal Calves: Impacts on Epithelial Barriers and Transcellular Transport Systems. Front. Cell. Infect. Microbiol. 2024, 14, 1495309. [Google Scholar] [CrossRef] [PubMed]
  86. Greigert, V.; Saraav, I.; Son, J.; Zhu, Y.; Dayao, D.; Antia, A.; Tzipori, S.; Witola, W.H.; Stappenbeck, T.S.; Ding, S. Cryptosporidium Infection of Human Small Intestinal Epithelial Cells Induces Type III Interferon and Impairs Infectivity of Rotavirus. Gut Microbes 2024, 16, 2297897. [Google Scholar] [CrossRef] [PubMed]
  87. Wang, L.; Cao, L.; Chang, Y.; Fu, Y.; Wang, Y.; Zhang, K.; Zhang, S.; Zhang, L. Microbiome-Metabolomics Analysis of the Impacts of Cryptosporidium muris Infection in BALB/C Mice. Microbiol. Spectr. 2023, 11, e02175-22. [Google Scholar] [CrossRef]
  88. Zhang, X.; Gong, A.; Wang, Y.; Chen, X.; Lim, S.S.; Dolata, C.E.; Chen, X. Cryptosporidium parvum Infection Attenuates the Ex Vivo Propagation of Murine Intestinal Enteroids. Physiol. Rep. 2016, 4, e13060. [Google Scholar] [CrossRef] [PubMed]
  89. Castellanos-Gonzalez, A.; Cabada, M.M.; Nichols, J.; Gomez, G.; White, A.C., Jr. Human Primary Intestinal Epithelial Cells as an Improved In Vitro Model for Cryptosporidium parvum Infection. Infect. Immun. 2013, 81, 1996–2001. [Google Scholar] [CrossRef] [PubMed]
  90. Priyamvada, S.; Jayawardena, D.; Bhalala, J.; Kumar, A.; Anbazhagan, A.N.; Alrefai, W.A.; Borthakur, A.; Dudeja, P.K. Cryptosporidium parvum Infection Induces Autophagy in Intestinal Epithelial Cells. Cell. Microbiol. 2021, 23, e13298. [Google Scholar] [CrossRef] [PubMed]
  91. Yang, X.; Wu, X.; Huang, S.; Yao, Q.; Chen, X.; Song, J.; Fan, Y.; Zhao, G. C3a/C3aR Affects the Propagation of Cryptosporidium parvum in the Ileum Tissues of Mice by Regulating the Gut Barrier, Cell Proliferation, and CD4+ T Cell Main Effectors. Animals 2023, 13, 837. [Google Scholar] [CrossRef]
  92. Liu, J.; Enomoto, S.; Lancto, C.A.; Abrahamsen, M.S.; Rutherford, M.S. Inhibition of Apoptosis in Cryptosporidium parvum-Infected Intestinal Epithelial Cells Is Dependent on Survivin. Infect. Immun. 2008, 76, 3784–3792. [Google Scholar] [CrossRef] [PubMed]
  93. Buret, A.G.; Chin, A.C.; Scott, K.G.E. Infection of Human and Bovine Epithelial Cells with Cryptosporidium andersoni Induces Apoptosis and Disrupts Tight Junctional ZO-1: Effects of Epidermal Growth Factor. Int. J. Parasitol. 2003, 33, 1363–1371. [Google Scholar] [CrossRef]
  94. Cui, Z.; Dong, H.; Wang, R.; Jian, F.; Zhang, S.; Ning, C.; Zhang, L. A Canine Model of Experimental Infection with Cryptosporidium canis. Exp. Parasitol. 2018, 195, 19–23. [Google Scholar] [CrossRef]
  95. de Sablet, T.; Potiron, L.; Marquis, M.; Bussière, F.I.; Lacroix-Lamandé, S.; Laurent, F. Cryptosporidium parvum Increases Intestinal Permeability through Interaction with Epithelial Cells and IL-1β and TNFα Released by Inflammatory Monocytes. Cell. Microbiol. 2016, 18, 1871–1880. [Google Scholar] [CrossRef]
  96. Li, S.; Li, W.; Yang, Z.; Song, S.; Yang, J.; Gong, P.; Zhang, W.; Liu, K.; Li, J.; Zhang, G. Infection of Cattle with Cryptosporidium parvum: Mast Cell Accumulation in Small Intestine Mucosa. Vet. Pathol. 2013, 50, 842–848. [Google Scholar] [CrossRef] [PubMed]
  97. Klein, P.; Kleinová, T.; Volek, Z.; Šimůnek, J. Effect of Cryptosporidium parvum Infection on the Absorptive Capacity and Paracellular Permeability of the Small Intestine in Neonatal Calves. Vet. Parasitol. 2008, 152, 53–59. [Google Scholar] [CrossRef]
  98. Sayed, F.G.; Hamza, A.I.; Galal, L.A.; Sayed, D.M.; Gaber, M. Virulence of Geographically Different Cryptosporidium parvum Isolates in Experimental Animal Model. Ann. Parasitol. 2016, 62, 221–232. [Google Scholar]
  99. Moore, R.; Tzipori, S.; Griffiths, J.K.; Johnson, K.; de Montigny, L.; Lomakina, I. Temporal Changes in Permeability and Structure of Piglet Ileum after Site-Specific Infection by Cryptosporidium parvum. Gastroenterology 1995, 108, 1030–1039. [Google Scholar] [CrossRef] [PubMed]
  100. Karpe, A.V.; Hutton, M.L.; Mileto, S.J.; James, M.L.; Evans, C.; Shah, R.M.; Ghodke, A.B.; Hillyer, K.E.; Metcalfe, S.S.; Liu, J.-W.; et al. Cryptosporidiosis Modulates the Gut Microbiome and Metabolism in a Murine Infection Model. Metabolites 2021, 11, 380. [Google Scholar] [CrossRef]
  101. McKenney, E.A.; Greene, L.K.; Drea, C.M.; Yoder, A.D. Down for the Count: Cryptosporidium Infection Depletes the Gut Microbiome in Coquerel’s Sifakas. Microb. Ecol. Health Dis. 2017, 28, 1335165. [Google Scholar] [CrossRef] [PubMed]
  102. Bourriaud, C.; Robins, R.J.; Martin, L.; Kozlowski, F.; Tenailleau, E.; Cherbut, C.; Michel, C. Lactate Is Mainly Fermented to Butyrate by Human Intestinal Microfloras but Inter-Individual Variation Is Evident. J. Appl. Microbiol. 2005, 99, 201–212. [Google Scholar] [CrossRef] [PubMed]
  103. Stoeva, M.K.; Garcia-So, J.; Justice, N.; Myers, J.; Tyagi, S.; Nemchek, M.; McMurdie, P.J.; Kolterman, O.; Eid, J. Butyrate-Producing Human Gut Symbiont, Clostridium Butyricum, and Its Role in Health and Disease. Gut Microbes 2021, 13, 1907272. [Google Scholar] [CrossRef] [PubMed]
  104. Ichikawa-Seki, M.; Motooka, D.; Kinami, A.; Murakoshi, F.; Takahashi, Y.; Aita, J.; Hayashi, K.; Tashibu, A.; Nakamura, S.; Iida, T.; et al. Specific Increase of Fusobacterium in the Faecal Microbiota of Neonatal Calves Infected with Cryptosporidium parvum. Sci. Rep. 2019, 9, 12517. [Google Scholar] [CrossRef]
  105. Kim, E.-T.; Lee, S.-J.; Kim, T.-Y.; Lee, H.-G.; Atikur, R.M.; Gu, B.-H.; Kim, D.-H.; Park, B.-Y.; Son, J.-K.; Kim, M.-H. Dynamic Changes in Fecal Microbial Communities of Neonatal Dairy Calves by Aging and Diarrhea. Animals 2021, 11, 1113. [Google Scholar] [CrossRef]
  106. Jandhyala, S.M. Role of the Normal Gut Microbiota. World J. Gastroenterol. 2015, 21, 8787. [Google Scholar] [CrossRef]
  107. Liu, H.; Zhang, H.; Wang, X.; Yu, X.; Hu, C.; Zhang, X. The Family Coriobacteriaceae Is a Potential Contributor to the Beneficial Effects of Roux-En-Y Gastric Bypass on Type 2 Diabetes. Surg. Obes. Relat. Dis. 2018, 14, 584–593. [Google Scholar] [CrossRef] [PubMed]
  108. Magne, F.; Gotteland, M.; Gauthier, L.; Zazueta, A.; Pesoa, S.; Navarrete, P.; Balamurugan, R. The Firmicutes/Bacteroidetes Ratio: A Relevant Marker of Gut Dysbiosis in Obese Patients? Nutrients 2020, 12, 1474. [Google Scholar] [CrossRef] [PubMed]
  109. Stojanov, S.; Berlec, A.; Štrukelj, B. The Influence of Probiotics on the Firmicutes/Bacteroidetes Ratio in the Treatment of Obesity and Inflammatory Bowel Disease. Microorganisms 2020, 8, 1715. [Google Scholar] [CrossRef] [PubMed]
  110. Chappell, C.L.; Darkoh, C.; Shimmin, L.; Farhana, N.; Kim, D.-K.; Okhuysen, P.C.; Hixson, J. Fecal Indole as a Biomarker of Susceptibility to Cryptosporidium Infection. Infect. Immun. 2016, 84, 2299–2306. [Google Scholar] [CrossRef]
  111. Shin, N.-R.; Whon, T.W.; Bae, J.-W. Proteobacteria: Microbial Signature of Dysbiosis in Gut Microbiota. Trends Biotechnol. 2015, 33, 496–503. [Google Scholar] [CrossRef]
  112. Gilbert, J.A.; Quinn, R.A.; Debelius, J.; Xu, Z.Z.; Morton, J.; Garg, N.; Jansson, J.K.; Dorrestein, P.C.; Knight, R. Microbiome-Wide Association Studies Link Dynamic Microbial Consortia to Disease. Nature 2016, 535, 94–103. [Google Scholar] [CrossRef]
  113. Alak, J.I.B.; Wolf, B.W.; Mdurvwa, E.G.; Pimentel-Smith, G.E.; Adeyemo, O. Effect of Lactobacillus Reuteri on Intestinal Resistance to Cryptosporidium parvum Infection in a Murine Model of Acquired Immunodeficiency Syndrome. J. Infect. Dis. 1997, 175, 218–221. [Google Scholar] [CrossRef] [PubMed]
  114. Pickerd, N.; Tuthill, D. Resolution of Cryptosporidiosis with Probiotic Treatment. Postgrad. Med. J. 2004, 80, 112–113. [Google Scholar] [CrossRef]
  115. Oliveira, B.C.M.; Bresciani, K.D.S.; Widmer, G. Deprivation of Dietary Fiber Enhances Susceptibility of Mice to Cryptosporidiosis. PLoS Neglected Trop. Dis. 2019, 13, e0007411. [Google Scholar] [CrossRef] [PubMed]
  116. Oliveira, B.C.M.; Widmer, G. Probiotic Product Enhances Susceptibility of Mice to Cryptosporidiosis. Appl. Environ. Microbiol. 2018, 84, 1408-18. [Google Scholar] [CrossRef] [PubMed]
  117. Markowiak, P.; Śliżewska, K. Effects of Probiotics, Prebiotics, and Synbiotics on Human Health. Nutrients 2017, 9, 1021. [Google Scholar] [CrossRef] [PubMed]
  118. Hills, R.; Pontefract, B.; Mishcon, H.; Black, C.; Sutton, S.; Theberge, C. Gut Microbiome: Profound Implications for Diet and Disease. Nutrients 2019, 11, 1613. [Google Scholar] [CrossRef] [PubMed]
  119. Kelly, S.M.; Munoz-Munoz, J.; van Sinderen, D. Plant Glycan Metabolism by Bifidobacteria. Front. Microbiol. 2021, 12, 609418. [Google Scholar] [CrossRef]
  120. Hills, R.D., Jr.; Pontefract, B.A.; Mishcon, H.R.; Black, C.A.; Sutton, S.C.; Theberge, C.R. Gut Microbiome: Profound Implications for Diet and Disease. Kompass Nutr. Diet. 2022, 2, 3–18. [Google Scholar]
  121. Dang, H.T.; Tran, D.M.; Phung, T.T.B.; Bui, A.T.P.; Vu, Y.H.; Luong, M.T.; Nguyen, H.M.; Trinh, H.T.; Nguyen, T.T.; Nguyen, A.H.; et al. Promising Clinical and Immunological Efficacy of Bacillus clausii Spore Probiotics for Supportive Treatment of Persistent Diarrhea in Children. Sci. Rep. 2024, 14, 6422. [Google Scholar] [CrossRef]
  122. Del Coco, V.F. Los Microorganismos Desde Una Perspectiva de Los Beneficios Para La Salud. Rev. Argent. Microbiol. 2015, 47, 171–173. [Google Scholar] [CrossRef]
  123. Adams, C.A. The Probiotic Paradox: Live and Dead Cells Are Biological Response Modifiers. Nutr. Res. Rev. 2010, 23, 37–46. [Google Scholar] [CrossRef]
  124. Guitard, J.; Menotti, J.; Desveaux, A.; Alimardani, P.; Porcher, R.; Derouin, F.; Kapel, N. Experimental Study of the Effects of Probiotics on Cryptosporidium parvum Infection in Neonatal Rats. Parasitol. Res. 2006, 99, 522–527. [Google Scholar] [CrossRef]
  125. Charania, R.; Wade, B.E.; McNair, N.N.; Mead, J.R. Changes in the Microbiome of Cryptosporidium-Infected Mice Correlate to Differences in Susceptibility and Infection Levels. Microorganisms 2020, 8, 879. [Google Scholar] [CrossRef] [PubMed]
  126. Foster, J.C.; Glass, M.D.; Courtney, P.D.; Ward, L.A. Effect of Lactobacillus and Bifidobacterium on Cryptosporidium parvum Oocyst Viability. Food Microbiol. 2003, 20, 351–357. [Google Scholar] [CrossRef]
  127. Mammeri, M.; Obregón, D.A.; Chevillot, A.; Polack, B.; Julien, C.; Pollet, T.; Cabezas-Cruz, A.; Adjou, K.T. Cryptosporidium parvum Infection Depletes Butyrate Producer Bacteria in Goat Kid Microbiome. Front. Microbiol. 2020, 11, 548737. [Google Scholar] [CrossRef]
  128. Lee, J.; d’Aigle, J.; Atadja, L.; Quaicoe, V.; Honarpisheh, P.; Ganesh, B.P.; Hassan, A.; Graf, J.; Petrosino, J.; Putluri, N. Gut Microbiota–Derived Short-Chain Fatty Acids Promote Poststroke Recovery in Aged Mice. Circ. Res. 2020, 127, 453–465. [Google Scholar] [CrossRef] [PubMed]
  129. Dong, H.; Chen, X.; Zhao, X.; Zhao, C.; Mehmood, K.; Kulyar, M.F.-A.; Bhutta, Z.A.; Zeng, J.; Nawaz, S.; Wu, Q.; et al. Intestine Microbiota and SCFAs Response in Naturally Cryptosporidium-Infected Plateau Yaks. Front. Cell. Infect. Microbiol. 2023, 13, 1105126. [Google Scholar] [CrossRef] [PubMed]
  130. Keelaghan, A.P.; Charania, R.; Mead, J.R. The Effect of Short-Chain Fatty Acids on Growth of Cryptosporidium parvum In Vitro. Microorganisms 2022, 10, 1822. [Google Scholar] [CrossRef]
  131. He, Z.; Dong, H. The Roles of Short-Chain Fatty Acids Derived from Colonic Bacteria Fermentation of Non-Digestible Carbohydrates and Exogenous Forms in Ameliorating Intestinal Mucosal Immunity of Young Ruminants. Front. Immunol. 2023, 14, 1291846. [Google Scholar] [CrossRef]
  132. Kim, C.H. Complex Regulatory Effects of Gut Microbial Short-Chain Fatty Acids on Immune Tolerance and Autoimmunity. Cell. Mol. Immunol. 2023, 20, 341–350. [Google Scholar] [CrossRef] [PubMed]
  133. McDonald, V.; Korbel, D.S.; Barakat, F.M.; Choudhry, N.; Petry, F. Innate Immune Responses Against Cryptosporidium parvum Infection. Parasite Immunol. 2013, 35, 55–64. [Google Scholar] [CrossRef]
  134. Lacroix-Lamandé, S.; Mancassola, R.; Naciri, M.; Laurent, F. Role of Gamma Interferon in Chemokine Expression in the Ileum of Mice and in a Murine Intestinal Epithelial Cell Line After Cryptosporidium parvum Infection. Infect. Immun. 2002, 70, 2090–2099. [Google Scholar] [CrossRef]
  135. Hayward, A.R.; Chmura, K.; Cosyns, M. Interferon-γ Is Required for Innate Immunity to Cryptosporidium parvum in Mice. J. Infect. Dis. 2000, 182, 1001–1004. [Google Scholar] [CrossRef] [PubMed]
  136. Ferguson, S.H.; Foster, D.M.; Sherry, B.; Magness, S.T.; Nielsen, D.M.; Gookin, J.L. Interferon-Λ3 Promotes Epithelial Defense and Barrier Function Against Cryptosporidium parvum Infection. Cell. Mol. Gastroenterol. Hepatol. 2019, 8, 1–20. [Google Scholar] [CrossRef]
  137. Odendall, C.; Voak, A.A.; Kagan, J.C. Type III IFNs Are Commonly Induced by Bacteria-Sensing TLRs and Reinforce Epithelial Barriers During Infection. J. Immunol. 2017, 199, 3270–3279. [Google Scholar] [CrossRef] [PubMed]
  138. Zhang, Y.; Yu, K.; Chen, H.; Su, Y.; Zhu, W. Caecal Infusion of the Short-Chain Fatty Acid Propionate Affects the Microbiota and Expression of Inflammatory Cytokines in the Colon in a Fistula Pig Model. Microb. Biotechnol. 2018, 11, 859–868. [Google Scholar] [CrossRef]
  139. Mathew, O.P.; Ranganna, K.; Yatsu, F.M. Butyrate, an HDAC Inhibitor, Stimulates Interplay Between Different Posttranslational Modifications of Histone H3 and Differently Alters G1-Specific Cell Cycle Proteins in Vascular Smooth Muscle Cells. Biomed. Pharmacother. 2010, 64, 733–740. [Google Scholar] [CrossRef] [PubMed]
  140. Parada Venegas, D.; De la Fuente, M.K.; Landskron, G.; González, M.J.; Quera, R.; Dijkstra, G.; Harmsen, H.J.M.; Faber, K.N.; Hermoso, M.A. Short Chain Fatty Acids (SCFAs)-Mediated Gut Epithelial and Immune Regulation and Its Relevance for Inflammatory Bowel Diseases. Front. Immunol. 2019, 10, 277. [Google Scholar]
  141. Rowin, J.; Xia, Y.; Jung, B.; Sun, J. Gut Inflammation and Dysbiosis in Human Motor Neuron Disease. Physiol. Rep. 2017, 5, e13443. [Google Scholar] [CrossRef] [PubMed]
  142. Lemieux, M.; Sonzogni-Desautels, K.; Ndao, M. Lessons Learned from Protective Immune Responses to Optimize Vaccines Against Cryptosporidiosis. Pathogens 2017, 7, 2. [Google Scholar] [CrossRef] [PubMed]
  143. Fayer, R.; Xiao, L. Cryptosporidium and Cryptosporidiosis, 2nd ed.; CRC Press: Boca Raton, FL, USA, 2007; ISBN 1420052276. [Google Scholar] [CrossRef]
  144. Murphy, K.; Travers, P.; Walport, M. Janeway’s Immunobiology; Garland Science: New York, NY, USA, 2012; Available online: https://archive.org/details/janewaysimmunobi0000murp (accessed on 5 June 2024).
  145. Zhao, G.H.; Fang, Y.Q.; Ryan, U.; Guo, Y.X.; Wu, F.; Du, S.Z.; Chen, D.K.; Lin, Q. Dynamics of Th17 Associating Cytokines in Cryptosporidium parvum-Infected Mice. Parasitol. Res. 2016, 115, 879–887. [Google Scholar] [CrossRef]
  146. Horie, M.; Ohmiya, Y.; Ohmori, T. Analysis of D-Amino Acid in Japanese Post-Fermented Tea, Ishizuchi-Kurocha. Biosci. Microbiota Food Health 2023, 42, 254–263. [Google Scholar] [CrossRef] [PubMed]
  147. Shao, D.Y.; Bai, X.; Tong, M.W.; Zhang, Y.Y.; Liu, X.L.; Zhou, Y.H.; Li, C.; Cai, W.; Gao, X.; Liu, M. Changes to the Gut Microbiota in Mice Induced by Infection with Toxoplasma Gondii. Acta Trop. 2020, 203, 105301. [Google Scholar] [CrossRef]
  148. Matsumoto, M.; Kunisawa, A.; Hattori, T.; Kawana, S.; Kitada, Y.; Tamada, H.; Kawano, S.; Hayakawa, Y.; Iida, J.; Fukusaki, E. Free D-Amino Acids Produced by Commensal Bacteria in the Colonic Lumen. Sci. Rep. 2018, 8, 17915. [Google Scholar] [CrossRef]
  149. Sasabe, J.; Miyoshi, Y.; Rakoff-Nahoum, S.; Zhang, T.; Mita, M.; Davis, B.M.; Hamase, K.; Waldor, M.K. Interplay Between Microbial D-Amino Acids and Host d-Amino Acid Oxidase Modifies Murine Mucosal Defence and Gut Microbiota. Nat. Microbiol. 2016, 1, 16125. [Google Scholar] [CrossRef]
  150. Xu, Z.; Guo, Y.; Roellig, D.M.; Feng, Y.; Xiao, L. Comparative Analysis Reveals Conservation in Genome Organization Among Intestinal Cryptosporidium Species and Sequence Divergence in Potential Secreted Pathogenesis Determinants Among Major Human-Infecting Species. BMC Genom. 2019, 20, 406. [Google Scholar] [CrossRef]
  151. Rosa, L.T.; Bianconi, M.E.; Thomas, G.H.; Kelly, D.J. Tripartite ATP-Independent Periplasmic (TRAP) Transporters and Tripartite Tricarboxylate Transporters (TTT): From Uptake to Pathogenicity. Front. Cell. Infect. Microbiol. 2018, 8, 33. [Google Scholar] [CrossRef]
  152. Chalmers, R.M. Cryptosporidium. In Microbiology of Waterborne Diseases; Singleton Hospital: Swansea, UK, 2014; pp. 263–276. [Google Scholar] [CrossRef]
  153. Shanmugasundram, A.; Gonzalez-Galarza, F.F.; Wastling, J.M.; Vasieva, O.; Jones, A.R. Library of Apicomplexan Metabolic Pathways: A Manually Curated Database for Metabolic Pathways of Apicomplexan Parasites. Nucleic Acids Res. 2013, 41, D706–D713. [Google Scholar] [CrossRef]
  154. Wang, R.; Wang, T.; Tait, S. Metabolic Regulation of Immunity. In Kelley and Firestein’s Textbook of Rheumatology, 10th ed.; Elsevier: Amsterdam, The Netherlands, 2016; Volume 1–2, pp. 318–326. [Google Scholar] [CrossRef]
  155. Rasmussen, K.R.; Arrowood, M.J.; Healey, M.C. Effectiveness of Dehydroepiandrosterone in Reduction of Cryptosporidial Activity in Immunosuppressed Rats. Antimicrob. Agents Chemother. 1992, 36, 220–222. [Google Scholar] [CrossRef] [PubMed]
  156. Choudhary, H.H.; Srivastava, P.N.; Singh, S.; Kumar, K.A.; Mishra, S. The Shikimate Pathway Enzyme That Generates Chorismate Is Not Required for the Development of Plasmodium Berghei in the Mammalian Host nor the Mosquito Vector. Int. J. Parasitol. 2018, 48, 203–209. [Google Scholar] [CrossRef] [PubMed]
  157. Kombo Mpindou, G.O.M.; Escuder Bueno, I.; Chorda Ramon, E. Review on Emerging Waterborne Pathogens in Africa: The Case of Cryptosporidium. Water 2021, 13, 2966. [Google Scholar] [CrossRef]
  158. Chakraborty, S.; Roy, S.; Mistry, H.U.; Murthy, S.; George, N.; Bhandari, V.; Sharma, P. Potential Sabotage of Host Cell Physiology by Apicomplexan Parasites for Their Survival Benefits. Front. Immunol. 2017, 8, 1261. [Google Scholar] [CrossRef]
  159. Yu, X.; Guo, F.; Mouneimne, R.B.; Zhu, G. Cryptosporidium parvum Elongation Factor 1α Participates in the Formation of Base Structure at the Infection Site During Invasion. J. Infect. Dis. 2020, 221, 1816–1825. [Google Scholar] [CrossRef] [PubMed]
  160. Minkoff, N.Z.; Aslam, S.; Medina, M.; Tanner-Smith, E.E.; Zackular, J.P.; Acra, S.; Nicholson, M.R.; Imdad, A. Fecal Microbiota Transplantation for the Treatment of Recurrent Clostridioides difficile (Clostridium difficile). Cochrane Database Syst. Rev. 2023, 4, CD013871. [Google Scholar] [CrossRef] [PubMed]
  161. Cani, P.D.; Bibiloni, R.; Knauf, C.; Waget, A.; Neyrinck, A.M.; Delzenne, N.M.; Burcelin, R. Changes in Gut Microbiota Control Metabolic Endotoxemia-Induced Inflammation in High-Fat Diet–Induced Obesity and Diabetes in Mice. Diabetes 2008, 57, 1470–1481. [Google Scholar] [CrossRef] [PubMed]
  162. Takiishi, T.; Fenero, C.I.M.; Câmara, N.O.S. Intestinal Barrier and Gut Microbiota: Shaping Our Immune Responses Throughout Life. Tissue Barriers 2017, 5, e1373208. [Google Scholar] [CrossRef]
  163. Piazzesi, A.; Pane, S.; Russo, A.; Del Chierico, F.; Francalanci, P.; Cotugno, N.; Rossi, P.; Locatelli, F.; Palma, P.; Putignani, L. Case Report: The Impact of Severe Cryptosporidiosis on the Gut Microbiota of a Pediatric Patient with CD40L Immunodeficiency. Front. Cell. Infect. Microbiol. 2023, 13, 1281440. [Google Scholar] [CrossRef]
  164. Widmer, G.; Creasey, H.N. Fecal Microbiota Impacts Development of Cryptosporidium parvum in the Mouse. Sci. Rep. 2024, 14, 5498. [Google Scholar] [CrossRef]
  165. Prabakaran, M.; Weible, L.; Champlain, J.; Jiang, R.; Biondi, K.; Weil, A.; Van Voorhis, W.; Ojo, K. The Gut-Wrenching Effects of Cryptosporidiosis and Giardiasis in Children. Microorganisms 2023, 11, 2323. [Google Scholar] [CrossRef] [PubMed]
  166. CDC. 2024 Symptoms of Cryptosporidiosis. Available online: https://www.cdc.gov/cryptosporidium/signs-symptoms/index.html#:~:text=Key%20points%201%20Symptoms%20of%20cryptosporidiosis%20generally%20begin,of%20cryptosporidiosis%20is%20diarrhea%20lasting%20days%20to%20weeks (accessed on 5 June 2024).
Figure 1. Transmission route of Cryptosporidium spp. from a gut health perspective. The cycle begins with the ingestion of oocysts from contaminated food and water, which leads to excystation in the stomach. The parasite reproduces in the small intestine, where thin-walled oocysts cause autoinfection, resulting in diarrhea, while the thick-walled oocysts are released into the environment and infect other susceptible hosts.
Figure 1. Transmission route of Cryptosporidium spp. from a gut health perspective. The cycle begins with the ingestion of oocysts from contaminated food and water, which leads to excystation in the stomach. The parasite reproduces in the small intestine, where thin-walled oocysts cause autoinfection, resulting in diarrhea, while the thick-walled oocysts are released into the environment and infect other susceptible hosts.
Vetsci 12 00085 g001
Figure 2. Invasion of C. parvum into enterocytes triggers an inflammatory response, leading to gut dysbiosis and diarrhea. The Cryptosporidium spp. infections disrupt the intestinal barrier by downregulating key TJPs, including ZO-1, E-cadherin, occludin, and claudin. As a result of this protozoan parasitic infection, immune cell infiltration, the activation of inflammatory cascades (TLR signaling and NF-κB pathways), and the disruption of intestinal barrier functions occur.
Figure 2. Invasion of C. parvum into enterocytes triggers an inflammatory response, leading to gut dysbiosis and diarrhea. The Cryptosporidium spp. infections disrupt the intestinal barrier by downregulating key TJPs, including ZO-1, E-cadherin, occludin, and claudin. As a result of this protozoan parasitic infection, immune cell infiltration, the activation of inflammatory cascades (TLR signaling and NF-κB pathways), and the disruption of intestinal barrier functions occur.
Vetsci 12 00085 g002
Table 1. Interrelations between cryptosporidiosis, gut barrier integrity with regard to TJPs, and other pathological alterations during Cryptosporidium spp. infection.
Table 1. Interrelations between cryptosporidiosis, gut barrier integrity with regard to TJPs, and other pathological alterations during Cryptosporidium spp. infection.
Animal ModelCryptosporidium spp.Effect of Cryptosporidium spp. Infection on TJs’ Integrity Other Pathophysiological Alterations References
Neonatal miceC. parvumCell junction and adherens junction genes were downregulated at peak infection time, e.g., at 11 and 30 dpi, there was a decreased expression of E-cadherin The V:C * was significantly reduced. An exaggerated inflammatory response and increased intestinal permeability resulted in diarrhea. Cell adhesion molecules were found to be involved in modulating apoptosis[51]
CalvesC. parvumHindered cell junctions, such as AJs *, TJs *, GJs *, hemidesmosomes, and desmosomesIncreased inflammation, apoptosis, villus atrophy, and lowered V:C resulted in an increased intestinal permeability leading to diarrhea[85]
Human small intestinal epithelial cellsC. hominisDisruption of TJs and AJs, altered barrier function, or distribution of junctional proteins were detected Many inflammatory genes were upregulated and intrinsic and extrinsic apoptotic pathways were activated, resulting in a disrupted intestinal barrier and diarrhea [86]
MiceC. murisInfection altered epithelial AJs, which were mediated by E-cadherinReduced V:C and increased mucosal inflammation, mucosal thickness and villi diameter, alterations in gut bacteria composition, and production of metabolites (SCFAs) were observed[87]
MiceC. parvumInfection downregulated TJ proteins occludin and claudin, which altered TJs between intestinal epithelial cellsInfection upregulated the expression of inflammatory genes, e.g., Nos2, Mip2, and Icam1, hence boosting immune responses inducing villus atrophy and inhibiting intestinal enteroids via increased apoptosis. The release of stress signals that can inhibit intestinal stem cell function in the crypt was also detected[88]
PEC (Human Primary Intestinal Epithelial Cells) culture mediaC. parvumAffected the intestinal barrier by disrupting the assembly of TJsModulated inflammatory response, damaged microvilli, and increased epithelial permeability[89]
Human Intestinal Enteroids (HIE)C. parvumModified protein or RNA expression related to TJs and AJsHost cell nuclei were constricted or collapsed, indicating apoptosis. Polarized apical brush border microvilli displayed disrupted TJs with an increased basolateral expression of NA+/K+ ATPase[74]
MiceC. parvumIntestinal epithelial barrier dysfunctioning due to the disruption of epithelial junctional complexes was foundIncreased mucosal infiltration with neutrophils and a marker of inflammation in the ileum, epithelial cell apoptosis, shortening of ileal and jejunal villi, increased permeability, and diarrhea were observed[50]
Caco-2 cell monolayers as in vitro model of IECs *C. parvumDisruption of intestinal epithelial barrier function as a result of significant downregulation of critical epithelial TJ and AJ proteins was observedPolymorphisms in autophagy genes, impaired immune responses, and epithelial cell function led to enteritis. Autophagy in absorptive cells, e.g., Paneth and goblet cells, was observed[90]
MiceC. parvumDownregulated the mRNA expression levels of ZO-1 *, claudin 3, and occludin and compromised the integrity of the intestinal barrier by downregulating TJsPathological damage led to necrotic enteric epithelial cells, shorter villi, a lower V:C, and an increased villi diameter. Suppression of C3aR * worsened the damage, further reducing intestinal permeability[91]
Human ileal adenoma cell model HCT-8C. parvumAbsence of adherence to neighboring cells and disrupted expression of proteins such as integrins and cadherins were detectedModulated inflammatory response, cell proliferation, differentiation, apoptosis, altered gene expression, and shortened villi with an increased mitochondrial membrane permeability, causing potential anomalies and releasing apoptogenic substances like cytochrome C, were detected[92]
Human and bovine epithelial cellsC. andersoniDisruptions of ZO-1 that serves as a connection between TJ occludin and cytoskeletal F-actin were detectedParasite invasion triggered enteritis and apoptosis. These disturbances lead to cytotoxic effects on enterocytes, resulting in increased intestinal permeability and loss of the barrier function[93]
Neonatal mice and dogsC. canisThe integrity of the epithelial cells of BF * was somewhat damagedInflamed and disordered epithelial surfaces led to cell death. Additionally, the compromised microvillus border resulted in the loss of cilia and atrophied mucosa in the duodenum and jejunum, and hence, increased intestinal permeability[94]
MiceC. parvumDisrupted the AJ complex between intestinal epithelial cellsLy6C+ * inflammatory monocytes induced the production of TNF-α and IL-1β. The infection triggered apoptosis, leading to epithelial cell loss, villi blunting, and shortening[95]
CattleC. parvumDisrupted epithelial microvilli and TJs between epithelial cells Dysregulated chemokine and cytokine production exacerbated inflammation. Infection triggered apoptosis and increased mast cells in jejunal villi, leading to villi damage and increased enteric epithelial permeability[96]
Neonatal calvesC. parvumWeakened TJs between epithelial cells Enteritis, especially with concurrent bacterial or viral infections, damaged villi and microvilli led to leaky gut syndrome[97]
MiceC. parvumNegatively affected epithelial cell junctions disrupted ZO-1 in Caco-2 cellsT cell-mediated inflammation caused epithelial cell damage through parasite invasion, proliferation, and extrusion, resulting in villus atrophy, crypt hyperplasia, epithelial cell loss, and villi blunting and shortening, with increased inflammatory cell infiltration in the crypts and weakened local mucosal immunity[98]
PigletsC. parvumInfected enterocytes retained their capacity to regulate TJsInfected tissue mucosa showed increased inflammatory cells, particularly in the lamina propria, leading to epithelial cell damage through apoptosis and resulting in shortened villi and altered epithelial macromolecular permeability[99]
* in the table text indicates that the full form of the abbreviation used or an explanation regarding the given abbreviation is mentioned in the footnote. V:C: villi-to-crypt ratio; TJs: tight junctions; AJs: adherens junctions; GJ: gap junctions; IECs: intestinal epithelial cells; ZO-1: zonula occludin; C3aR: C3a (complement component 3a) receptor; BF: bursas of Fabricius; Ly6C+: Lymphocyte antigen 6 complex locus C positive.
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

Ali, M.; Xu, C.; Wang, M.; Hina, Q.; Ji, Y.; Anwar, S.; Lu, S.; He, Q.; Qiu, Y.; Li, K. Gut Barrier Dysfunction and Microbiota Variations in Cryptosporidiosis: A Comprehensive Review. Vet. Sci. 2025, 12, 85. https://doi.org/10.3390/vetsci12020085

AMA Style

Ali M, Xu C, Wang M, Hina Q, Ji Y, Anwar S, Lu S, He Q, Qiu Y, Li K. Gut Barrier Dysfunction and Microbiota Variations in Cryptosporidiosis: A Comprehensive Review. Veterinary Sciences. 2025; 12(2):85. https://doi.org/10.3390/vetsci12020085

Chicago/Turabian Style

Ali, Munwar, Chang Xu, Mingyue Wang, Qazal Hina, Yaru Ji, Subiha Anwar, Sijia Lu, Qing He, Yawei Qiu, and Kun Li. 2025. "Gut Barrier Dysfunction and Microbiota Variations in Cryptosporidiosis: A Comprehensive Review" Veterinary Sciences 12, no. 2: 85. https://doi.org/10.3390/vetsci12020085

APA Style

Ali, M., Xu, C., Wang, M., Hina, Q., Ji, Y., Anwar, S., Lu, S., He, Q., Qiu, Y., & Li, K. (2025). Gut Barrier Dysfunction and Microbiota Variations in Cryptosporidiosis: A Comprehensive Review. Veterinary Sciences, 12(2), 85. https://doi.org/10.3390/vetsci12020085

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