Warning: file_put_contents(/opt/frankenphp/design.onmedianet.com/storage/proxy/cache/d9f736eac81949816dc21f3ceb47983b.html): Failed to open stream: No space left on device in /opt/frankenphp/design.onmedianet.com/app/src/Arsae/CacheManager.php on line 36

Warning: http_response_code(): Cannot set response code - headers already sent (output started at /opt/frankenphp/design.onmedianet.com/app/src/Arsae/CacheManager.php:36) in /opt/frankenphp/design.onmedianet.com/app/src/Models/Response.php on line 17

Warning: Cannot modify header information - headers already sent by (output started at /opt/frankenphp/design.onmedianet.com/app/src/Arsae/CacheManager.php:36) in /opt/frankenphp/design.onmedianet.com/app/src/Models/Response.php on line 20
Advances in understanding Giardia: determinants and mechanisms of chronic sequelae - PMC Skip to main content
F1000Prime Reports logoLink to F1000Prime Reports
. 2015 May 26;7:62. doi: 10.12703/P7-62

Advances in understanding Giardia: determinants and mechanisms of chronic sequelae

Luther A Bartelt 1,, R Balfour Sartor 2
PMCID: PMC4447054  PMID: 26097735

Abstract

Giardia lamblia is a flagellated protozoan that is the most common cause of intestinal parasitic infection in children living in resource-limited settings. The pathogenicity of Giardia has been debated since the parasite was first identified, and clinical outcomes vary across studies. Among recent perplexing findings are diametrically opposed associations between Giardia and acute versus persistent diarrhea and a poorly understood potential for long-term sequelae, including impaired child growth and cognitive development. The mechanisms driving these protean clinical outcomes remain elusive, but recent advances suggest that variability in Giardia strains, host nutritional status, the composition of microbiota, co-infecting enteropathogens, host genetically determined mucosal immune responses, and immune modulation by Giardia are all relevant factors influencing disease manifestations after Giardia infection.

Introduction

Giardia lamblia (also known as G. duodenalis and G. intestinalis) is the most common and oftentimes earliest parasitic infection in children and the most common cause of diarrhea in travelers presenting to clinics [14]. Owing to an estimated 280 million infections every year [5,6], nearly universal infection among children living in resource-limited regions worldwide [6,7], the potential for persistent and recurrent infections with an average duration of Giardia carriage of 6.3 months among children in some populations [2], and limited affordable effective therapeutic options [8,9], the World Health Organization (WHO) considers Giardia a neglected tropical disease [10,11]. Because the parasite is environmentally ubiquitous, can persist for prolonged periods in the environment as hardy cysts, is capable of propagating through both anthroponotic [12] and zoonotic [1319] reservoirs, and has an infectious dose as low as 10 cysts [20], Giardia exposures can also occur in resource-abundant settings [2125]. In the United States, for example, infection is characterized by seasonal and recreational waterborne transmission [22,23] and clustered daycare outbreaks [26]. Transmission through food sources, such as leafy green vegetables and culinary bivalves harboring G. lamblia shed from coastal and marine life [2729], is also possible.

Anton van Leeuwenhoek first identified Giardia in his own diarrheal stool in 1681, but the parasite did not become an officially WHO-recognized pathogen until 1981. Even after Theodore Nash and colleagues [30] fulfilled Robert Koch's postulates in human volunteers in 1987 [20], disease attributable to Giardia was debated. Complicating this clinical equipoise is the predominance of apparently asymptomatic hosts and as-of-yet elusive mechanisms explaining how some individuals develop the characteristic giardiasis syndrome: abdominal cramping and bloating, malabsorptive diarrhea (steatorrhea), and weight loss [18]. Furthermore, for unclear reasons, chronic sequelae, including post-infectious irritable bowel syndrome, chronic fatigue [31], malnutrition [32], cognitive impairment [33], and extra-intestinal manifestations (such as food allergy, urticaria, reactive arthritis, and inflammatory ocular manifestations), can develop and possibly persist beyond detectable parasite shedding (Figure 1).

Figure 1. The clinical spectrum of Giardia infection.

Figure 1.

The majority of individuals infected with Giardia are asymptomatic. Within the diarrhea spectrum of disease, Giardia paradoxically associates with both protection against acute diarrhea and persistent diarrhea with or without malabsorption and enteropathy. Hypothesized determinants of how disease will manifest on this spectrum include the virulence of the infecting strain(s) of Giardia, host nutritional intake, co-infecting enteropathogens, the composition and function of resident microbiota, immune modulation by Giardia, and host genetics and immunity. Changes in these dynamic variables may further skew disease manifestation. Histological changes vary from normal histopathology (bottom left) to villus shortening and chronic inflammatory infiltrate (enteropathy) (bottom right). Chronic sequelae associated with giardiasis include irritable bowel syndrome, chronic fatigue, childhood growth faltering, failure to thrive, cognitive impairment, and extra-intestinal manifestations presumed to be related to immunologic phenomena (reactive arthritis, inflammatory ocular manifestations, and urticaria) that are not necessarily dependent on severity of diarrheal manifestations and may persist even beyond detection of the parasite. Giardia trophozoite image courtesy of Joel Mancuso and Scott Dawson. Images of small-bowel biopsies from patients with giardiasis showing normal (left) and abnormal (right) histopathology courtesy of Leana Guerin (left) and Jörg-Dieter Schulzke (right).

Global efforts to improve child health in resource-limited settings situate Giardia infections amidst the broader context of childhood malnutrition, a multitude of diverse and frequent enteric pathogen exposures [34], and new understandings of the role of environmental enteropathy (a form of chronic gastrointestinal dysfunction and inflammation possibly inclusive of the previously termed clinical condition ‘tropical sprue‘) [11,35] and microbiota [36] on childhood development and growth. The most recent analyses, equipped with more rigorous surveillance methods and more sensitive molecular diagnostics [3739], have not only reiterated the wide range of variability in pediatric Giardia infection outcomes seen in prior studies [3] but unveiled a much greater burden of exposure than estimates based on microscopic techniques [39,40]. These and other studies have raised provocative considerations, including the potential for Giardia to be protective against acute diarrhea [4143].

In light of the incompletely understood pathology attributable to Giardia infection, recent advances in parasite and gastrointestinal biology hope to clarify how and when Giardia causes disease. These advances address fundamental questions regarding how Giardia interacts within the complex ecology of microbial, nutritional, and host factors within the small intestine, the determinants of host susceptibility, and whether and how host inflammation contributes to pathogenesis (Figure 2).

Figure 2. Proposed determinants and mechanisms of Giardia infection outcomes.

Figure 2.

Complex interactions between microbiota, nutrients, Giardia strain, co-enteropathogens, and host molecular responses in the luminal and mucosal environment likely influence Giardia infectivity and disease outcomes. (Left) Resident microbiota maintain resiliency to colonization. Giardia uses, and potentially sequesters, nutrients such as bile, arginine, and zinc in order to survive, replicate, and evade microbiota and host defenses. Flagella and the ventral disc are structures of trophozoites that aid attachment and adherence to intestinal epithelial cells (IECs). Giardia uses functional virulence factors to evade host inflammatory responses through antioxidant production, cleavage of interleukin-8 (IL-8), arginine depletion via arginine deiminase (ADI), and shifts in variant surface protein (VSP) expression. Effects of Giardia on epithelial cells (that is cell-cycle arrest, impaired proliferation, tight-junction disruption, and apoptosis) may be strain dependent and either direct or indirect. Subsequent changes in nutrient availability, microbiota composition, inflammatory defenses, and epithelial cell pathogen attachment sites may secondarily alter disease manifestations of co-infecting enteropathogens. (Right) Redundant mucosal immune responses promote Giardia clearance early (epithelial cell nutrient uptake for host fitness, barrier function maintenance, and pro-inflammatory molecules; IL-6 derived from dendritic cells and mast cells; and CD4+ and CD8+ T cells) and late in disease (CD4+ T-cell memory and B cells). CD4+ T cells induce memory responses but also contribute to chronic inflammation and may promote disaccharidase deficiency. CD8+ T cells mediate apoptosis, microvillus shortening, and disaccharidase deficiency. Epithelial cell damage may persist beyond parasite clearance, allowing sustained translocation of microbiota and microbial products. The altered mucosal homeostasis and inflammation (enteropathy) and microbiota composition may further impede nutrient uptake and contribute to prolonged sequelae, including impaired growth and cognitive development.

Endemic pediatric giardiasis equipoise

In 1971, after charting weights of children in Guatemala, Leonardo J. Mata reported an association between arrested growth and frequent episodes of diarrhea [44]. A follow-up analysis of these children associated increased duration and frequency of Giardia episodes with a greater detrimental growth impact [45]. Since then, several studies have attempted to demonstrate how early Giardia infections promote diarrhea and developmental shortfalls. A 2012 meta-analysis found that, although Giardia infection was associated with persistent diarrhea (>14 days; odds ratio [OR] 3.2, 95% confidence interval [CI] 1.50-6.76; P <0.001), there was a negative association with moderate-severe acute diarrhea (OR 0.6, 95% CI 0.38-0.95; P <0.03) [3]. Subsequently, the Global Enteric Multicenter Study identified a similar paradox [41]. This finding was consistent regardless of diagnostic modality and quantitative burden in stool specimens [39]. Other prospective cohort studies in Tanzania [42] and Iran [43] also demonstrated a delay in first episode of diarrhea in children who were positive for Giardia at enrollment. Elsewhere, Giardia is positively associated with acute diarrhea [46], and younger children may be at greater risk for acute diarrhea with Giardia infection [3].

Associations between Giardia and childhood growth and developmental shortfalls have also been variable. Stunting (low height-for-age z score) has been documented in Brazil [47], Colombia [48], and Ecuador [49]; poor intestinal permeability has been seen in Nepali children [50], growth impairment in Rwandan children [51], wasting in Malaysia [32] and India [52], and low weight-for-age and height-for-age in Brazilian children with persistent symptoms [53,54]. In addition, cognitive impairment has been documented in children up to seven years after Giardia infection independent of physical growth [33]. Other equally valid studies, however, have shown no significant influence on developmental outcome or nutritional status [3,5557]. The Malnutrition and Enteric Disease Study, to date the most comprehensive longitudinal cohort study aimed at determining environmental, nutritional, and enteric pathogenic influences on childhood development, is ongoing and will offer the most complete opportunity to ascribe associations between Giardia, growth, and gastrointestinal dysfunction [58]. There have been no randomized controlled trials evaluating the developmental impact of specific anti-giardial therapies in malnourished children. However, bovine fecal lactoferrin supplementation simultaneously decreases Giardia colonization and promotes child growth [59].

Whether Giardia infection initiates or promotes childhood malnutrition has major implications for the nearly 20% of undernourished children worldwide [58]. Malnutrition substantially increases mortality and morbidities to both infectious and non-infectious conditions, and undernourished children may not demonstrate expected restorative gains with nutritional supplementation. Indeed, protein energy malnutrition [60], zinc deficiency, and vitamin A deficiency [61] may increase susceptibility to Giardia. Conversely, Giardia infection may reduce supplemental zinc uptake [6264] and influence dose response to vitamin A supplementation [65]. Further confounding nutritional determinants in Giardia outcomes are the findings in a study of multi-nutrient therapy for childhood diarrhea that Giardia-infected subjects in the placebo arm were protected against diarrhea but that Giardia-infected subjects in the multi-nutrient intervention arm were not [42]. Proposed mechanisms explaining such paradoxical findings suggest that specific nutrients may promote Giardia growth or otherwise directly mediate Giardia pathogenicity, alter host microbiota and other defenses, or interfere with the parasite's influence on co-enteropathogens and host immune responses.

Determining how Giardia causes disease

Infection with Giardia occurs after ingestion of the environmental cyst stage of the parasite, which upon gastric passage excysts to release two trophozoites into the less acidic and bile-enriched environment of the upper small bowel. Trophozoites adhere to epithelial cells and replicate but typically do not invade the intestinal mucosa or cause ulceration [6,66]. If symptoms develop, onset typically begins after a 6- to 15-day incubation period that oftentimes precedes detection of parasites in stool [5]. The type of diarrhea experienced in giardiasis is classically greasy and foul-smelling (steatorrhea) with findings of malabsorption of vitamins A and B12, d-xylose, iron, and zinc [3,6769] as well as lactase deficiency in 20-40% of symptomatic cases [70]. Histological biopsies of small-bowel mucosa are often without any apparent abnormality [71]. However, increased lymphocytic infiltrates and decreased villus-to-crypt ratios have been described in some children [72], and inflammation has been reported in some adults with either acute [73] or chronic [74] infection.

Experimental as well as clinical observations indicate that the primary cause of diarrhea in giardiasis is malabsorption and that hypersecretion during chronic infection possibly contributes [74,75]. Malabsorption is presumed to contribute to growth impairment but has not been causally linked to it. Multiple investigations suggest both direct and indirect mechanisms of disease, including luminal competition for nutrients, Giardia-induced epithelial cell damage (apoptosis [76,77], arrested proliferation [78], and tight-junction abnormalities [76,77,79]); T cell-mediated epithelial cell injury [80,81], microvilli loss [75,8082], and disaccharidase deficiency [83,84]; nutrient-dependent growth impairment and decreased villus-to-crypt ratios [85]; and translocation of microbiota across the mucosa [86] (Figure 2).

Unscrambling strain virulence and pathogenicity

Of at least six recognized species of Giardia, only G. lamblia causes infections in humans. G. lamblia isolates are further divided into eight assemblages, designated A to H. Among the assemblages, only assemblages A and B and their respective subtypes cause human infection. The relative proportion of assemblage A to assemblage B infection varies both temporally and spatially, with a predilection for more assemblage B infections in endemic settings [8790].

Whole-genome sequencing has revealed that assemblage A and assemblage B laboratory isolates are quite dissimilar and may be better categorized as separate species [91]. Despite this genetic divergence, attempts to ascribe clinical variability and pathogenicity based on assemblage designation have been inconclusive. In separate studies, either assemblage A [92,93] or assemblage B [94,95] more strongly associates with diarrhea. A limitation in assessing assemblage-specific pathogenicity in naturally acquired infection is that targets used to differentiate between assemblages A and B—β-giardin (bg), glutamate dehydrogenase (gdh), triose phosphate isomerase (tpi), and the small subunit 18S rRNA genes—are not known virulence factors. Furthermore, studies that incorporate multilocus genotyping demonstrate a substantial proportion of specimens with mixed results [96,97], raising the possibility of heterologous infections or even inter-assemblage recombination events [98,99]. Expanded comparative genomics studies investigating several assemblage A and assemblage B isolates may further clarify strain-specific pathogenicity.

In experimental conditions, Giardia strain is the strongest predictor of disease outcome. In initial volunteer studies, the axenized assemblage B isolate GS/M caused more symptoms than other isolates, a finding that was reproduced in murine models [30,100]. Laboratory assemblage A and assemblage B isolates have different growth characteristics in standard media conditions, and the former are more readily recoverable; however, assemblage B isolates are more infectious [84,85]. Historically, there has been significant experimental heterogeneity in laboratory studies [101], and this limits comparisons between investigators. When tested under identical conditions, however, there are apparent assemblage-dependent differences in induction or disruption of interleukin-8 (IL-8) from epithelial cells in vitro [102,103] and ex vivo biopsies from patients with inflammatory bowel disease [104] and immune-mediated disaccharidase deficiency in murine models [84]. Furthermore, in vitro models demonstrate that strain-dependent changes occur in epithelial cells [77] and that mixed assemblage co-cultivation, or cultivation with a clone expressing mixed assemblage traits, induces more epithelial cell apoptosis and tight-junction disruption than mono-infection with either assemblage A or B [105]. Finally, proximity to a natural source may also be relevant given that recently human-passaged strains were more infectious in an animal model than laboratory strains were [106] and that gerbil-passaged cysts achieve prolonged infections [85].

Advances in Giardia biology and gene regulation using transcriptomic and proteomic approaches aim to identify new virulence traits that could be confirmed in human infections. For example, as a possible means of evading host humoral and innate defenses, Giardia has a repertoire of 20 to 200 kDa cysteine-rich proteins that densely coat the surface of trophozoites, termed variant surface proteins (VSPs). Although only a single VSP is expressed on an individual trophozoite, the breadth of potential VSPs (73 to 270 or more) [91,107] in a given strain is associated with enhanced virulence [108]. How VSP switching occurs every 6-13 generations and what events trigger post-transcriptional VSP regulation [107,109] and trafficking may help unravel parasite determinants of transient colonization opposed to chronic infection. The role of Giardia cathepsins as disease determinants also warrants further consideration [102,103].

Nutritional determinants of disease

It is increasingly recognized that, within the luminal ecology of the human intestine, resident microbiota modify ingested nutrients and their metabolites, influencing nutrient availability and uptake. Giardia lacks fundamental enzymes necessary for generating critical biomolecules such as cholesterol and therefore must rely on acquisition of these materials from the luminal environment [110112]. The parasite acquires nutrients primarily via bulk-phase uptake in endocytic vacuoles localized to the exposed dorsal (non-adherent) side [113], adheres to epithelial cells using the ventral disc [114,115], and may direct fluid flow toward adherent parasites via coordinated flagellar motion [116]. Arginine utilization represents an example of how shunting of key nutrients may provide an advantage to the parasite at a detriment to the host. Stadelmann and colleagues [117] demonstrated that Giardia isolates expressing arginine deiminase rapidly deplete arginine, resulting in epithelial cell cell-cycle arrest. Since both Giardia and epithelial cells use arginine for growth, an arginine-depleting mechanism could explain why, under conditions of arginine scarcity such as severe protein malnutrition, there is increased susceptibility to villus shortening during Giardia infection [85,118]. For the host, decreased availability of arginine also impairs inducible nitric oxide production by epithelial cells and may alter cellular immune responses [119,120], whereas Giardia uses the sequestered arginine primarily for energy production via the arginine dehydrolase pathway. Host genetically determined pathways that facilitate arginine acquisition or utilization despite Giardia infection may be important for maintaining host resiliency during infection. For example, cellular arginine uptake occurs through the cationic transporter (CAT), and CAT expression is increased in some cell types in the presence of APO-E 4/4 allelotype [121]. The APO-E 4/4 allelotype confers protection against diarrhea in children [122,123] and Cryptosporidium infection in mice [121], and may also associate with better cognitive outcomes in Giardia-infected children [124].

There is mounting recognition of an intricate relationship between host nutrition and mucosal immune responses. Malnourished children, for example, have diminished seroconversion to some components of the oral polio vaccine [125]. In experimental models, malnutrition confers enhanced susceptibility to Cryptosporidium [126128] and enteroaggregative Escherichia coli (EAEC) [129]. Zinc deficiency enhances susceptibility to Giardia [130] and diminishes inflammatory responses to EAEC [131]. In a chronic giardiasis model, protein malnutrition led to decreased expression of IL-4 and IL-5 and diminished B-cell populations in intestinal tissues, villus blunting, and growth failure despite a parasite burden similar to that of well-nourished infected controls [85]. These findings suggest that, in children with chronic giardiasis, nutritional status could similarly determine disease severity.

Microbiota and co-pathogen interactions

There is increasing recognition of the role of the nearly 1014 resident bacteria in regulating host metabolism during states of both obesity and undernutrition [36,132] as well as immunity. In experimental giardiasis, the microbiota composition was shown to have more influence over susceptibility to infection than the absence of CD4+ T cells [133]. Thus, several experimental models rely on manipulating host microbiota through the use of antibiotic-containing water to promote infection [84,85,103]. Though sometimes necessary, the depletion of microbiota in these models may exclude important Giardia effects on microbiota that indirectly influence pathogenesis. Although attempts to identify small-bowel bacterial overgrowth in children with giardiasis have failed to show an association [134], microbiota from duodenal contents of Giardia-infected patients caused more inflammation than axenized Giardia trophozoites in germ-free mice [135]. Chen and colleagues [86] also observed that diverse populations of mucosally associated bacteria, including Lactobacillus, Streptococcus, and Staphylococcus species, are increased during experimental Giardia infection and after clearance of the parasite. Associated increases in the pro-inflammatory cytokines interferon-gamma (IFN-γ), tumor necrosis factor-alpha (TNF-α), and myeloperoxidase were also seen [86]. The persistent translocation of mucosally associated bacterial microbiota has been proposed to explain post-infection irritable bowel syndrome and chronic fatigue [31], sequelae present in up to 5% of individuals with prior giardiasis [136], particularly in those whose initial course was more protracted [137]. Mechanisms by which Giardia could influence microbiota composition include alterations in the luminal nutritional environment as previously stated, as well as dampening the epithelial cell inflammatory responses through post-translational reductions in IL-8 and decreased nitric oxide synthesis [102,120,138140].

Whether resident microbiota in the small intestine are protective against Giardia or promote chronic gastrointestinal disease may be conditional on their composition. When used as probiotics in experimental models, Lactobacillus species (L. johnsonii, L. casei, and L. rahmnosus GG) have been shown to promote Giardia clearance [141]. Concurrently, these Lactobacillus species enhance pro-inflammatory responses and improve histopathological parameters [142144]. Furthermore, bacteriocins produced by L. acidophilus (P106) and L. plantarum (P164) reduced parasite adhesion [145,146]. The fermented milk product kefir, which contains a combination of lactic acid bacteria (lactobacillus, lactococci, and Leuconostoc), acetic acid bacteria, and yeast, protected mice against G. lamblia trophozoite infection and enhanced pro-inflammatory responses, including IFN-γ and TNF-α, with a decrease in CD4+ T-cell expansion [147]. Interestingly, activation of Toll-like receptor 2 (TLR2) signaling in murine dendritic cells, as occurs in response to some Lactobacillus species, in combination with TLR1 or TLR6, leads to increased IL-12/23p40, IL-23, and IL-10 secretion in the presence of Giardia lysate [148], whereas Giardia lysates decrease IL-12p70 and IL-23 but increase IL-10 responses to TLR4 agonists [148]. The down-regulatory effect of Giardia lysate on dendritic cell responses to TLR4 agonists has been shown to be dependent on phosphoinositide 3-kinase inhibition and may depend on Giardia arginine deiminase [78,149]. In contrast, Giardia lysates have no regulatory effect on bovine monocyte-derived dendritic cells [150].

The influence of Giardia-mediated changes in the luminal ecology may also alter susceptibility to other co-infecting enteropathogens, which are now recognized to be more common than mono-infections in children in resource-limited settings [34]. Furthermore, only a narrow margin separates the mean number of co-enteropathogens found in non-diarrheal specimens (4.3 ± 0.1) from the number in diarrheal specimens (5.6 ± 0.1) [34]. In some populations, up to 75% of Giardia infections include co-enteropathogens, most commonly with Vibrio cholerae and rotavirus [151] and elsewhere with norovirus and enteropathogenic E. coli [46]. Cases of co-infections with Tropheryma whipplei have also been reported [152]. In rural Ecuador, the OR for diarrhea with rotavirus infection was 6-14.8, but G. lamblia with rotavirus increased the OR to 11-24.13. However, Giardia co-infection with enterotoxigenic Escherichia coli, Campylobacter, Cryptosporidium, and enteroinvasive Escherichia coli did not demonstrate an increased risk of diarrhea, suggesting specificity to Giardia co-pathogen interactions [153]. On the other hand, associations between decreased risk of acute diarrhea in Giardia-infected cohorts raise questions that require additional investigation of whether Giardia decreases susceptibility to some pathogens or attenuates their severity. Cotton and colleagues [103], for example, demonstrated that Giardia infection diminished neutrophil chemotaxis and inflammation in a model of Clostridium difficile toxin-induced colitis.

Host responses and disease pathogenesis

The duality of host immune response in Giardia pathogenesis is complex and has been reviewed extensively [83,154]. Increasingly, redundant mechanisms of immunologic control are being identified, including chemokines (CCL2, CCL20, CXCL1, CXCL2, and CXCL3) from epithelial cells [155], IL-6 derived from dendritic cells and possibly mast cells [156158], TNF-α [159], α-defensins activated by matrix metalloprotease 7 (Mmp7) [159], and nitric oxide generated from either nitric oxide synthase 1 (NOS1) or NOS2 [159161]. Data from isolated outbreaks and experimental models demonstrate a mixed immune response consisting of both antibody production [162,163] and mucosal and systemic Th1-type CD4+ T-cell responses [84,164,165]. Studies in populations with serial outbreaks demonstrate that exposure to Giardia can promote protective immunity [166], which is characterized by a predominance of circulating T cells with a Th1-memory phenotype (CD25+CD26bright cells that produce IFN-γ) [165]. Also, the absence of CD4+ T cells leads to more prolonged infection in experimental models [84].

Despite evidence for the role of T cells in protective immunity, clinical observations suggest that humoral responses are critical for immunity. These studies demonstrate greater prevalence of Giardia cysts and more symptomatology in patients with hypogammaglobulinemia (that is, x-linked agammaglobulinemia [Bruton's disease] and common variable immunodeficiency) [167169] than those with T-cell deficits (advanced AIDS or thymic aplasia). Decreased secretory IgA variably associates with increased risk [170] and impaired IgA responses are seen in children with persistent Giardia infection [171]. In animal models, the majority of genes expressed in experimental murine infections are related to antibody production [159], and IgA-deficient knockout mice have difficulty clearing Giardia [172]. The polymeric Ig receptor, which transports IgA and IgM across the epithelium, is essential for eliminating G. muris from murine hosts, further supporting possible dependence on mucosal secretory antibody responses for eradication of infection [173].

In addition to facilitating parasite clearance, the secondary host immune response to Giardia may promote pathologic changes [84,174]. CD8+ T cells may contribute to villus [85] and microvillus [175] shortening in G. lamblia infection as demonstrated in models of murine giardiasis (G. muris) [80,81,84]. In addition, brush border actin cytoskeletal changes in ezrin and villin and disaccharidase deficiency after G. lamblia infection are dependent on T-cell responses [174]. Clinical findings also demonstrate that subsets of infected symptomatic adults [74] and some children show increased lymphocytic infiltrates as well as decreased villus-to-crypt ratios [72] on histological biopsy. Decreased villus surface area in patients with chronic giardiasis coincides with increased T-cell populations and epithelial cell apoptosis [74], and may similarly suggest immune-mediated diffuse microvillus shortening seen in some patients with giardiasis [82]. A murine model using freshly acquired gerbil-passaged G. lamblia cysts in combination with antibiotic-containing water to reduce resident microbiota similarly demonstrated persistent parasitism, with increased mucosal T cells and epithelial cell apoptosis through 9 weeks in immunocompetent hosts, suggesting that some Giardia strains that evade host responses can promote chronic mucosal inflammation [85].

Such chronic and repeated Giardia exposures are common in children in low-income settings who also have a high prevalence of malnutrition and an increasingly recognized gut dysfunction with chronic intestinal inflammation, increased gut permeability, and reduced villus length, termed environmental enteropathy [11]. Few studies have characterized immune responses to Giardia in these children. Kohli and colleagues [87] identified inflammatory diarrhea by using fecal lactoferrin during the first case (but not subsequent cases) of Giardia, and Long and colleagues [176] demonstrated increased IL-4, IL-5, monocyte chemoattractant protein-1 (MCP-1), and IFN-γ but decreased IL-8 in children with more prolonged episodes of Giardia. Thus, relationships between intestinal inflammatory response and Giardia infection may influence disease outcomes in these children.

Elucidation of immune-protective responses in contrast to those that might directly promote enteropathy in these children could also help guide vaccine development. Currently, the only licensed vaccine, GiardiaVax, is for use only in canines and has modest efficacy [177]. Attempts to identify correlates of protection in children in low-resource endemic settings are inconclusive. For example, data from Bangladesh demonstrated that, although human breast milk has direct anti-Giardia properties and may contain high concentrations of anti-Giardia antibodies [18], there is no correlation between the presence of Giardia-specific antibodies in breast milk and incidence, severity, or recurrence of Giardia in these children [178]. Potential approaches to vaccine development include targeting the repertoire of G. lamblia VSPs, since purified VSPs can protect against primary infection in experimental models [179]. Another vaccine candidate, α1-giardin expressed in an attenuated Salmonella vector, showed efficacy in mice, but α-enolase and ornithine carbamoyl transferase did not [180]. Challenging field vaccine application is the recognition that some mucosal vaccines are less effective in children in low-resource settings, with an as-of-yet unidentified but hypothesized immunologic hurdle in the setting of malnutrition [125], adding another layer of complexity to this preventive strategy.

Conclusions

G. lamblia remains an enigmatic parasite with potential influence over health and development in children in endemic resource-limited settings where infection is nearly universal. One conclusion explaining the variable outcomes in both field studies and experimental models is that Giardia may not conform to traditional concepts of the ‘commensal‘ or ‘pathogenic‘ microbe and at times may promote states of either mucosal protection or disease. Therefore, discerning the direct effects of various Giardia strains from indirect effects mediated through the parasite's influence on a complex and dynamic luminal ecology and host mucosal homeostasis will be a critical step toward translating basic laboratory findings to outcomes in human disease. Such understanding could lead to improved interventions for chronic giardiasis, as well as environmental enteropathy and childhood malnutrition, while also identifying properties of the organism that could be exploited for the promotion of human health.

Acknowledgments

We thank Richard L. Guerrant, Cirle A. Warren, Leana Guerin, Scott Dawson, and Jörg-Dieter Schulzke for their insights and assistance with figure preparation. Support was received from National Institutes of Health/National Institute of Allergy and Infectious Diseases grant K08-AI108730-01 to Luther A. Bartelt.

Abbreviations

APO-E

apolipoprotein E

CAT

cationic transporter

CI

confidence interval

EAEC

enteroaggregative Escherichia coli

IFN-γ

interferon-gamma

IL

interleukin

NOS

nitric oxide synthase

OR

odds ratio

TLR

Toll-like receptor

TNF-α

tumor necrosis factor-alpha

VSP

variant surface protein

WHO

World Health Organization

Disclosures

The authors declare that they have no disclosures.

The electronic version of this article is the complete one and can be found at: http://f1000.com/prime/reports/b/7/62

References

  • 1.Ryan ET, Wilson ME, Kain KC. Illness after international travel. N Engl J Med. 2002;347:505–16. doi: 10.1056/NEJMra020118. [DOI] [PubMed] [Google Scholar]
  • 2.Fraser D, Dagan R, Naggan L, Greene V, El-On J, Abu-Rbiah Y, Decklebaum R. Natural history of Giardia lamblia and Cryptosporidium infections in a cohort of Israeli Bedouin infants: a study of a population in transition. Am J of Trop Med and Hyg. 1997;57:5449. doi: 10.4269/ajtmh.1997.57.544. [DOI] [PubMed] [Google Scholar]
  • 3.Muhsen K, Levine MM. A systematic review and meta-analysis of the association between Giardia lamblia and endemic pediatric diarrhea in developing countries. Clin Infect Dis. 2012;55(Suppl 4):S271–93. doi: 10.1093/cid/cis762. [DOI] [PMC free article] [PubMed] [Google Scholar]; http://f1000.com/prime/717969213
  • 4.Ross AG, Olds GR, Cripps AW, Farrar JJ, McManus DP. Enteropathogens and chronic illness in returning travelers. N Engl J Med. 2013;368:1817–25. doi: 10.1056/NEJMra1207777. [DOI] [PubMed] [Google Scholar]
  • 5.Ankarklev J, Jerlström-Hultqvist J, Ringqvist E, Troell K, Svärd SG. Behind the smile: cell biology and disease mechanisms of Giardia species. Nat Rev Microbiol. 2010;8:413–22. doi: 10.1038/nrmicro2317. [DOI] [PubMed] [Google Scholar]; http://f1000.com/prime/720772355
  • 6.Hill DR, Nash TE. Intestinal flagellate and ciliate infections. In: Guerrant RL, Walker DH, Weller PF, editors. Tropical infectious diseases: Principles, Pathogens, & Practice. 3. Philadelphia: Churchill Livingtson; 2011. pp. 623–32. [DOI] [Google Scholar]
  • 7.Haque R, Mondal D, Karim A, Molla IH, Rahim A, Faruque ASG, Ahmad N, Kirkpatrick BD, Houpt E, Snider C, Petri WA. Prospective case-control study of the association between common enteric protozoal parasites and diarrhea in Bangladesh. Clin Infect Dis. 2009;48:1191–7. doi: 10.1086/597580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Solaymani-Mohammadi S, Genkinger JM, Loffredo CA, Singer SM. A meta-analysis of the effectiveness of albendazole compared with metronidazole as treatments for infections with Giardia duodenalis. PLoS Negl Trop Dis. 2010;4:e682. doi: 10.1371/journal.pntd.0000682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Rossignol JF. Cryptosporidium and Giardia: treatment options and prospects for new drugs. Exp Parasitol. 2010;124:45–53. doi: 10.1016/j.exppara.2009.07.005. [DOI] [PubMed] [Google Scholar]
  • 10.Savioli L, Smith H, Thompson A. Giardia and Cryptosporidium join the ‘Neglected Diseases Initiative’. Trends Parasitol. 2006;22:203–8. doi: 10.1016/j.pt.2006.02.015. [DOI] [PubMed] [Google Scholar]
  • 11.Bartelt LA, Lima AA, Kosek M, Peñataro Yori P, Lee G, Guerrant RL. ‘Barriers’ to child development and human potential: the case for including the ‘neglected enteric protozoa’ (NEP) and other enteropathy-associated pathogens in the NTDs. PLoS Negl Trop Dis. 2013;7:e2125. doi: 10.1371/journal.pntd.0002125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Thompson RC, Monis P. Giardia—from genome to proteome. Adv Parasitol. 2012;78:57–95. doi: 10.1016/b978-0-12-394303-3.00003-7. [DOI] [PubMed] [Google Scholar]
  • 13.Baruch AC, Isaac-Renton J, Adam RD. The molecular epidemiology of Giardia lamblia: a sequence-based approach. J of Infect Dis. 1996;174:233–6. doi: 10.1093/infdis/174.1.233. [DOI] [PubMed] [Google Scholar]
  • 14.Hogan JN, Daniels ME, Watson FG, Conrad PA, Oates SC, Miller MA, Hardin D, Byrne BA, Cominik C, Melli A, Jessup DA, Miller WA. Longitudinal Poisson regression to evaluate the epidemiology of Cryptosporidium, Giardia, and fecal indicator bacteria in coastal California wetlands. Appl Environ Microbiol. 2012;78:3606–13. doi: 10.1128/AEM.00578-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Gomez-Puerta LA, Lopez-Urbina MT, Alarcon V, Cama V, Gonzalez AE, Xiao L. Occurrence of Giardia duodenalis assemblages in alpacas in the Andean region. Parasitol Int. 2014;63:31–4. doi: 10.1016/j.parint.2013.10.003. [DOI] [PubMed] [Google Scholar]
  • 16.García-Presedo I, Pedraza-Díaz S, González-Warleta M, Mezo M, Gómez-Bautista M, Ortega-Mora LM, Castro-Hermida JA. The first report of Cryptosporidium bovis, C. ryanae and Giardia duodenalis sub-assemblage A-II in roe deer (Capreolus capreolus) in Spain. Vet Parasitol. 2013;197:658–64. doi: 10.1016/j.vetpar.2013.07.002. [DOI] [PubMed] [Google Scholar]
  • 17.Feng Y, Xiao L. Zoonotic potential and molecular epidemiology of Giardia species and giardiasis. Clin Microbiol Rev. 2011;24:110–40. doi: 10.1128/CMR.00033-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Hill DR, Nash TE. Giardiais. In: Bennett JE, Dolin R, Blaser MJ, editors. Mandell, Douglas, and Bennet's Principles and Practice of Infectious Disease. Vol. 281. Saunders: Elsevier; 2015. pp. 3154–60.e3. [Google Scholar]
  • 19.Traub RJ, Monis PT, Robertson I, Irwin P, Mencke N, Thompson RC. Epidemiological and molecular evidence supports the zoonotic transmission of Giardia among humans and dogs living in the same community. Parasitiology. 2004;128:253–62. doi: 10.1017/S0031182003004505. [DOI] [PubMed] [Google Scholar]
  • 20.Lipoldová M. Giardia and Vilém Dušan Lambl. PLoS Negl Trop Dis. 2014;8:e2686. doi: 10.1371/journal.pntd.0002686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Mahmoudi MR, Kazemi B, Mohammadiha A, Mirzaei A, Karanis P. Detection of Cryptosporidium and Giardia (oo)cysts by IFA, PCR, and LAMP in surface water from Rasht, Iran. Trans R Soc Trop Med Hyg. 2013;107:511–7. doi: 10.1093/trstmh/trt042. [DOI] [PubMed] [Google Scholar]
  • 22.Mohamed AS, Levine M, Camp JW, Lund E, Yoder JS, Glickman LT, Moore GE. Temporal patterns of human and canine Giardia infection in the United States: 2003-2009. Prev Vet Med. 2014;113:249–56. doi: 10.1016/j.prevetmed.2013.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Kitajima M, Haramoto E, Iker BC, Gerba CP. Occurrence of Cryptosporidium, Giardia, and Cyclospora in influent and effluent water at wastewater treatment plants in Arizona. Sci Total Environ. 2014;484:129–36. doi: 10.1016/j.scitotenv.2014.03.036. [DOI] [PubMed] [Google Scholar]
  • 24.Sroka J, Stojecki K, Zdybel J, Karamon J, Cencek T, Dutkiewicz J. Occurrence of Cryptosporidium oocysts and Giardia cysts in effluent from sewage treatment plant from eastern Poland. Ann Agric Environ Med. 2013;1:57–62. [PubMed] [Google Scholar]
  • 25.ElBakri A, Samie A, Bessong P, Potgieter N, Odeh RA. Detection and molecular characterization of Giardia lamblia genotypes in Sharjah, United Arab Emirates. Trans R Soc Trop Med Hyg. 2014;108:466–73. doi: 10.1093/trstmh/tru083. [DOI] [PubMed] [Google Scholar]
  • 26.Enserink R, van den Wijngaard C, Bruijning-Verhagen P, van Asten L, Mughini-Gras L, Duizer E, Kortbeek T, Scholts R, Nagelkerke N, Smit HA, Kooistra-Smid M, van Pelt W. Gastroenteritis attributable to 16 Enteropathogens in children attending day care. Significant effects of rotavirus, norovirus, astrovirus, Cryptosporidium and Giardia. Pediatr Infect Dis J. 2014 doi: 10.1097/INF.0000000000000472. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  • 27.Adell AD, Smith WA, Shapiro K, Melli A, Conrad PA. Molecular Epidemiology of Cryptosporidium spp. and Giardia spp. in Mussels (Mytilus californianus) and California Sea Lions (Zalophus californianus) from Central California. Appl Environ Microbiol. 2014;80:7732–40. doi: 10.1128/AEM.02922-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Giangaspero A, Papini R, Marangi M, Koehler AV, Gasser RB. Cryptosporidium parvum genotype IIa and Giardia duodenalis assemblage A in Mytilus galloprovincialis on sale at local food markets. Int J Food Microbiol. 2014;171:62–7. doi: 10.1016/j.ijfoodmicro.2013.11.022. [DOI] [PubMed] [Google Scholar]
  • 29.Schets FM, van den Berg HH, de Roda Husman AM. Determination of the recovery efficiency of Cryptosporidium oocysts and Giardia cysts from seeded bivalve mollusks. J Food Prot. 2013;76:93–8. doi: 10.4315/0362-028X.JFP-12-326. [DOI] [PubMed] [Google Scholar]
  • 30.Nash TE, Herrington DA, Losonsky GA, Levine MM. Experimental human infections with Giardia lamblia. J Infect Dis. 1987;156:974–84. doi: 10.1093/infdis/156.6.974. [DOI] [PubMed] [Google Scholar]; http://f1000.com/prime/725450152
  • 31.Hanevik K, Wensaas KA, Rortveit G, Eide GE, Mørch K, Langeland N. Irritable bowel syndrome and chronic fatigue 6 years after Giardia infection: a controlled prospective cohort study. Clin Infect Dis. 2014;59:1394–400. doi: 10.1093/cid/ciu629. [DOI] [PMC free article] [PubMed] [Google Scholar]; http://f1000.com/prime/718530391
  • 32.Al-Mekhlafi MS, Azlin M, Nor Aini U, Shaik A, Sa'iah A, Fatmah MS, Ismail MG, Ahmad Firdaus MS, Asiah MY, Rozlida AR, Morhayati M. Giardiasis as a predictor of childhood malnutrition in Orang Asli children in Malaysia. Trans R Soc Trop Med Hyg. 2005;99:686–91. doi: 10.1016/j.trstmh.2005.02.006. [DOI] [PubMed] [Google Scholar]; http://f1000.com/prime/1026970
  • 33.Berkman DS, Lescano AG, Gilman RH, Lopez SL, Black MM. Effects of stunting, diarrhoeal disease, and parasitic infection during infancy on cognition in late childhood: a follow-up study. Lancet. 2002;359:564–71. doi: 10.1016/S0140-6736(02)07744-9. [DOI] [PubMed] [Google Scholar]; http://f1000.com/prime/1004822
  • 34.Taniuchi M, Sobuz SU, Begum S, Platts-Mills JA, Liu J, Yang Z, Wang Xin-Qun, Petri WA, Haque R, Houpt ER. Etiology of diarrhea in Bangladeshi infants in the first year of life analyzed using molecular methods. J Infect Dis. 2013;208:1794–802. doi: 10.1093/infdis/jit507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Korpe PS, Petri WA. Environmental enteropathy: critical implications of a poorly understood condition. Trends Mol Med. 2012;18:328–36. doi: 10.1016/j.molmed.2012.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Smith MI, Yatsunenko T, Manary MJ, Trehan I, Mkakosya R, Chang J, Kau AL, Rich SS, Concannon P, Mychaleckyj JC, Liu J, Houpt E, Li JV, Holmes E, Nicholson J, Knights D, Ursell LK, Knight R, Gordon JI. Gut microbiomes of Malawian twin pairs discordant for kwashiorkor. Science. 2013;339:548–54. doi: 10.1126/science.1229000. [DOI] [PMC free article] [PubMed] [Google Scholar]; http://f1000.com/prime/717976764
  • 37.Boadi S, Polley SD, Kilburn S, Mills GA, Chiodini PL. A critical assessment of two real-time PCR assays targeting the (SSU) rRNA and gdh genes for the molecular identification of Giardia intestinalis in a clinical laboratory. J Clin Pathol. 2014;67:811–6. doi: 10.1136/jclinpath-2014-202224. [DOI] [PubMed] [Google Scholar]
  • 38.Heyworth MF. Diagnostic testing for Giardia infections. Trans R Soc Trop Med Hyg. 2014;108:123–5. doi: 10.1093/trstmh/tru005. [DOI] [PubMed] [Google Scholar]
  • 39.Liu J, Kabir F, Manneh J, Lertsethtakarn P, Begum S, Gratz J, Becker SM, Operario DJ, Taniuchi M, Janaki L, Platts-Mills JA, Haverstick DM, Kabir M, Sobuz SU, Nakjarung K, Sakpaisal P, Silapong S, Bodhidatta L, Qureshi S, Kalam A, Saidi Q, Swai N, Mujaga B, Maro A, Kwambana B, Dione M, Antonio M, Kibiki G, Mason CJ, Haque R, et al. Development and assessment of molecular diagnostic tests for 15 enteropathogens causing childhood diarrhoea: a multicentre study. Lancet Infect Dis. 2014;14:716–24. doi: 10.1016/S1473-3099(14)70808-4. [DOI] [PubMed] [Google Scholar]
  • 40.Mejia R, Vicuña Y, Broncano N, Sandoval C, Vaca M, Chico M, Cooper PJ, Nutman TB. A novel, multi-parallel, real-time polymerase chain reaction approach for eight gastrointestinal parasites provides improved diagnostic capabilities to resource-limited at-risk populations. Am J Trop Med Hyg. 2013;88:1041–7. doi: 10.4269/ajtmh.12-0726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Kotloff KL, Nataro JP, Blackwelder WC, Blackwelder WC, Nasrin D, Farag TH, Panchalingam S, Wu Y, Sow S, Sur D, Breiman RF, Faruque AS, Zaidi AK, Saha D, Alonso PL, Tamboura B, Sanogo D, Onwuchekwa U, Manna B, Ramamurthy T, Kanungo S, Ochieng JB, Omore R, Oundo JO, Hossain A, Das SK, Ahmed S, Qureshi S, Quadri F, Adegbola RA, 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;382:209–22. doi: 10.1016/S0140-6736(13)60844-2. [DOI] [PubMed] [Google Scholar]; http://f1000.com/prime/718203197
  • 42.Veenemans J, Schouten LR, Ottenhof MJ, Mank TG, Uges DRA, Mbugi EV, Demir AY, Kraaijenhagen RJ, Savelkoul HFJ, Verhoef H. Effect of preventive supplementation with zinc and other micronutrients on non-malarial morbidity in Tanzanian pre-school children: a randomized trial. PLoS One. 2012;7:e41630. doi: 10.1371/journal.pone.0041630. [DOI] [PMC free article] [PubMed] [Google Scholar]; http://f1000.com/prime/717954589
  • 43.Muhsen K, Cohen D, Levine MM. Can Giardia lamblia infection lower the risk of acute diarrhea among preschool children? J Trop Pediatr. 2014;60:99–103. doi: 10.1093/tropej/fmt085. [DOI] [PubMed] [Google Scholar]
  • 44.Mata LJ, Urrutia JJ, Lechtig A. Infection and nutrition of children of a low socioeconomic rural community. Am J Clin Nutr. 1971;24:249–59. doi: 10.1093/ajcn/24.2.249. [DOI] [PubMed] [Google Scholar]
  • 45.Farthing MJ, Mata L, Urrutia JJ, Kronmal RA. Natural history of Giardia infection of infants and children in rural Guatemala and its impact on physical growth. Am J Clin Nutr. 1986;43:395–405. doi: 10.1093/ajcn/43.3.395. [DOI] [PubMed] [Google Scholar]
  • 46.Becker-Dreps S, Bucardo F, Vilchez S, Zambrana LE, Liu L, Weber DJ, Rodolfo P, Barclay L, Vinjé J, Hudgens MG, Nordgren J, Svensson L, Morgan DR, Espinoza F, Paniagua M. Etiology of childhood diarrhea after rotavirus vaccine introduction: A prospective, population-based study in Nicaragua. Pediatr Infect Dis J. 2014;33:1156–63. doi: 10.1097/INF.0000000000000427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Prado MS, Cairncross S, Strina A, Barreto ML, Oliveira-Assis AM, Rego S. Asymptomatic giardiasis and growth in young children; a longitudinal study in Salvador, Brazil. Parasitology. 2005;131:51–6. doi: 10.1017/S0031182005007353. [DOI] [PubMed] [Google Scholar]
  • 48.Boeke CE, Mora-Plazas M, Forero Y, Villamor E. Intestinal protozoan infections in relation to nutritional status and gastrointestinal morbidity in Colombian school children. J Trop Pediatr. 2010;56:299–306. doi: 10.1093/tropej/fmp136. [DOI] [PubMed] [Google Scholar]
  • 49.Sackey ME, Weigel MM, Armijos RX. Predictors and nutritional consequences of intestinal parasitic infections in rural Ecuadorian children. J Trop Pediatr. 2003;49:17–23. doi: 10.1093/tropej/49.1.17. [DOI] [PubMed] [Google Scholar]
  • 50.Goto R, Panter-Brick C, Northrop-Clewes CA, Manahdhar R, Tuladhar NR. Poor intestinal permeability in mildly stunted Nepali children: associations with weaning practices and Giardia lamblia infection. Br J Nutr. 2002;88:141–9. doi: 10.1079/BJN2002599. [DOI] [PubMed] [Google Scholar]
  • 51.Ignatius R, Gahutu JB, Klotz C, Steininger C, Shyirambere C, Lyng M, Musemakweri A, Aebischer T, Martus P, Gundel H, Mockenhaupt FP. High prevalence of Giardia duodenalis Assemblage B infection and association with underweight in Rwandan children. PLoS Negl Trop Dis. 2012;6:e1677. doi: 10.1371/journal.pntd.0001677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Ajjampur SSR, Koshy B, Venkataramani M, Sarkar R, Joseph AA, Jacob KS, Ward H, Kang G. Effect of cryptosporidial and giardial diarrhoea on social maturity, intelligence and physical growth in children in a semi-urban slum in south India. Ann Trop Paediatr. 2011;31:205–12. doi: 10.1179/1465328111Y.0000000003. [DOI] [PubMed] [Google Scholar]
  • 53.Newman RD, Moore SR, Lima AA, Nataro JP, Guerrant RL, Sears CL. A longitudinal study of Giardia lamblia infection in north-east Brazilian children. Trop Med Int Health. 2001;6:624–34. doi: 10.1046/j.1365-3156.2001.00757.x. [DOI] [PubMed] [Google Scholar]
  • 54.Botero-Garcés JH, García-Montoya GM, Grisales-Patiño D, Aguirre-Acevedo DC, Alvarez-Uribe MC. Giardia intestinalis and nutritional status in children participating in the complementary nutrition program, Antioquia, Colombia, May to October 2006. Rev Inst Med Trop Sao Paulo. 2009;51:155–62. doi: 10.1590/S0036-46652009000300006. [DOI] [PubMed] [Google Scholar]
  • 55.Ish-Horowicz M, Korman SH, Shapiro M, Har-Even U, Tamir I, Strauss N, Deckelbaum RJ. Asymptomatic giardiasis in children. Pediatr Infect Dis J. 1989;8:773–9. doi: 10.1097/00006454-198911000-00009. [DOI] [PubMed] [Google Scholar]
  • 56.Moya-Camarena SY, Sotelo N, Valencia ME. Effects of asymptomatic Giardia intestinalis infection on carbohydrate absorption in well-nourished Mexican children. Am J Trop Med Hyg. 2002;66:255–9. doi: 10.4269/ajtmh.2002.66.255. [DOI] [PubMed] [Google Scholar]
  • 57.Hollm-Delgado M-G, Gilman RH, Bern C, Cabrera L, Sterling CR, Black RE, Checkley W. Lack of an adverse effect of Giardia intestinalis infection on the health of Peruvian children. Am J Epidemiol. 2008;168:647–55. doi: 10.1093/aje/kwn177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.MAL-ED Network Investigators The MAL-ED study: a multinational and multidisciplinary approach to understand the relationship between enteric pathogens, malnutrition, gut physiology, physical growth, cognitive development, and immune responses in infants and children up to 2 years of age in resource-poor environments. Clin Infect Dis. 2014;59(Suppl 4):S193–206. doi: 10.1093/cid/ciu653. [DOI] [PubMed] [Google Scholar]
  • 59.Ochoa TJ, Chea-Woo E, Campos M, Pecho I, Prada A, McMahon RJ, Cleary TG. Impact of lactoferrin supplementation on growth and prevalence of Giardia colonization in children. Clin Infect Dis. 2008;46:1881–3. doi: 10.1086/588476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Muniz-Junqueira MI, Queiroz EF. Relationship between protein-energy malnutrition, vitamin A, and parasitoses in living in Brasília. Rev Soc Bras Med Trop. 2002;35:133–41. doi: 10.1590/S0037-86822002000200002. [DOI] [PubMed] [Google Scholar]
  • 61.Quihui-Cota L, Astiazarán-García H, Valencia ME, Morales-Figueroa GG, Lopez-Mata MA, Vazquez Ortiz F. Impact of Giardia intestinalis on vitamin a status in schoolchildren from northwest Mexico. Int J Vitam Nutr Res. 2008;78:51–6. doi: 10.1024/0300-9831.78.2.51. [DOI] [PubMed] [Google Scholar]
  • 62.Ertan P, Yereli K, Kurt O, Balcioğlu IC, Onağ A. Serological levels of zinc, copper and iron elements among Giardia lamblia infected children in Turkey. Pediatr Int. 2002;44:2868. doi: 10.1046/j.1442-200X.2002.01550.x. [DOI] [PubMed] [Google Scholar]
  • 63.Abou-Shady O, El Raziky MS, Zaki MM, Mohamed RK. Impact of Giardia lamblia on growth, serum levels of zinc, copper, and iron in Egyptian children. Biol Trace Elem Res. 2011;140:1–6. doi: 10.1007/s12011-010-8673-6. [DOI] [PubMed] [Google Scholar]
  • 64.Rosado JL, Caamaño MC, Montoya YA, de Lourdes Solano M, Santos JI, Long KZ. Interaction of zinc or vitamin A supplementation and specific parasite infections on Mexican infants' growth: a randomized clinical trial. Eur J Clin Nutr. 2009;63:1176–84. doi: 10.1038/ejcn.2009.53. [DOI] [PubMed] [Google Scholar]
  • 65.Astiazaran-Garcia H, Lopez-Teros V, Valencia ME, Vazquez-Ortiz F, Sotelo-Cruz N, Quihui-Cota L. Giardia lamblia infection and its implications for vitamin A liver stores in school children. Ann Nutr Metab. 2010;57:228–33. doi: 10.1159/000321682. [DOI] [PubMed] [Google Scholar]
  • 66.Martinez-Gordillo M, Gonzalez-Maciel A, Reynoso-Robles R, Montijo-Barrios E, Ponce-Macotela M. Intraepithelial Giardia intestinalis: a case report and literature review. Medicine. 2014;93:e277. doi: 10.1097/MD.0000000000000277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Hjelt K, Paerregaard A, Krasilnikoff PA. Giardiasis causing chronic diarrhoea in suburban Copenhagen: incidence, physical growth, clinical symptoms and small intestinal abnormality. Acta Paediatr. 1992;81:881–6. doi: 10.1111/j.1651-2227.1992.tb12128.x. [DOI] [PubMed] [Google Scholar]
  • 68.Behera B, Mirdha BR, Makharia GK, Bhatnagar S, Dattagupta S, Samantaray JC. Parasites in patients with malabsorption syndrome: a clinical study in children and adults. Dig Dis Sci. 2008;53:672–9. doi: 10.1007/s10620-007-9927-9. [DOI] [PubMed] [Google Scholar]
  • 69.Solomons NW. Giardiasis: nutritional implications. Rev Infect Dis. 1982;4:859–69. doi: 10.1093/4.4.859. [DOI] [PubMed] [Google Scholar]
  • 70.Rana SV, Bhasin DK, Vinayak VK. Lactose hydrogen breath test in Giardia lamblia-positive patients. Dig Dis Sci. 2005;50:259–61. doi: 10.1007/s10620-005-1592-2. [DOI] [PubMed] [Google Scholar]
  • 71.Oberhuber G, Kastner N, Stolte M. Giardiasis: a histologic analysis of 567 cases. Scand J Gastroenterol. 1997;32:48–51. doi: 10.3109/00365529709025062. [DOI] [PubMed] [Google Scholar]
  • 72.Koot BG, ten Kate FJ, Juffrie M, Rosalina I, Taminiau JJ, Benninga MA. Does Giardia lamblia cause villous atrophy in children?: A retrospective cohort study of the histological abnormalities in giardiasis. J Pediatr Gastroenterol Nutr. 2009;49:304–8. doi: 10.1097/MPG.0b013e31818de3c4. [DOI] [PubMed] [Google Scholar]
  • 73.Hanevik K, Hausken T, Morken MH, Strand EA, Mørch K, Coll P, Helgeland L, Langeland N. Persisting symptoms and duodenal inflammation related to Giardia duodenalis infection. J Infect. 2007;55:524–30. doi: 10.1016/j.jinf.2007.09.004. [DOI] [PubMed] [Google Scholar]
  • 74.Troeger H, Epple HJ, Schneider T, Wahnschaffe U, Reiner U, Burchard G-D, Jelinek T, Zeitz M, Fromm M, Schulzke J-D. Effect of chronic Giardia lamblia infection on epithelial transport and barrier function in human duodenum. Gut. 2007;56:328–35. doi: 10.1136/gut.2006.100198. [DOI] [PMC free article] [PubMed] [Google Scholar]; http://f1000.com/prime/1071915
  • 75.Buret A, Hardin JA, Olson ME, Gall DG. Pathophysiology of small intestinal malabsorption in gerbils infected with Giardia lamblia. Gastroenterology. 1992;103:506–13. doi: 10.1016/0016-5085(92)90840-u. [DOI] [PubMed] [Google Scholar]
  • 76.Panaro MA, Cianciulli A, Mitolo V, Mitolo CI, Acquafredda A, Brandonisio O, Cavallo P. Caspase-dependent apoptosis of the HCT-8 epithelial cell line induced by the parasite Giardia intestinalis. FEMS Immunol Med Microbiol. 2007;51:302–9. doi: 10.1111/j.1574-695X.2007.00304.x. [DOI] [PubMed] [Google Scholar]
  • 77.Chin AC, Teoh DA, Scott KG, Meddings JB, Macnaughton WK, Buret AG. Strain-dependent induction of enterocyte apoptosis by Giardia lamblia disrupts epithelial barrier function in a caspase-3-dependent manner. Infect Immun. 2002;7:3673–80. doi: 10.1128/IAI.70.7.3673-3680.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Banik S, Renner Viveros P, Seeber F, Klotz C, Ignatius R, Aebischer T. Giardia duodenalis arginine deiminase modulates the phenotype and cytokine secretion of human dendritic cells by depletion of arginine and formation of ammonia. Infect Immun. 2013;81:2309–17. doi: 10.1128/IAI.00004-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Teoh DA, Kamieniecki D, Pang G, Buret AG. Giardia lamblia rearranges F-actin and alpha-actinin in human colonic and duodenal monolayers and reduces transepithelial electrical resistance. J Parasitol. 2000;86:800–6. doi: 10.1645/0022-3395(2000)086[0800:GLRFAA]2.0.CO;2. [DOI] [PubMed] [Google Scholar]
  • 80.Scott KG, Logan MR, Klammer GM, Teoh DA, Buret AG. Jejunal brush border microvillous alterations in Giardia muris-infected mice: role of T lymphocytes and interleukin-6. Infect Immun. 2000;68:3412–8. doi: 10.1128/IAI.68.6.3412-3418.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Scott KG, Yu LC, Buret AG. Role of CD8+ and CD4+ T lymphocytes in jejunal mucosal injury during murine giardiasis. Infect Immun. 2004;72:3536–42. doi: 10.1128/IAI.72.6.3536-3542.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Barbieri D, de Brito T, Hoshino S, Nascimento Fa OB, Martins Campos J, Quarentei G, Marcondes E. Giardiasis in Childhood: Absorption Tests and Biochemistry, Histochemistry, Light, and Electron Microscopy of Jejunal Mucosa. Arch Dis Child. 1970;45:466–72. doi: 10.1136/adc.45.242.466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Solaymani-Mohammadi S, Singer SM. Giardia duodenalis: the double-edged sword of immune responses in giardiasis. Exp Parasitol. 2010;126:292–7. doi: 10.1016/j.exppara.2010.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]; http://f1000.com/prime/725450153
  • 84.Solaymani-Mohammadi S, Singer SM. Host immunity and pathogen strain contribute to intestinal disaccharidase impairment following gut infection. J of Immunol. 2011;187:3769–75. doi: 10.4049/jimmunol.1100606. [DOI] [PMC free article] [PubMed] [Google Scholar]; http://f1000.com/prime/719454306
  • 85.Bartelt LA, Roche J, Kolling G, Bolick D, Noronha F, Naylor C, Hoffman P, Warren C, Singer S, Guerrant R. Persistent, G. lamblia impairs growth in a murine malnutrition model. J Clin Invest. 2013;123:2672–84. doi: 10.1172/JCI67294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Chen TL, Chen S, Wu HW, Lee TC, Lu YZ, Wu LL, Ni YH, Sun CH, Yu WH, Buret AG, Yu LCH. Persistent gut barrier damage and commensal bacterial influx following eradication of Giardia infection in mice. Gut Pathog. 2013;5:26. doi: 10.1186/1757-4749-5-26. [DOI] [PMC free article] [PubMed] [Google Scholar]; http://f1000.com/prime/725450154
  • 87.Kohli A, Bushen OY, Pinkerton RC, Houpt E, Newman RD, Sears CL, Lima AAM, Guerrant RL. Giardia duodenalis assemblage, clinical presentation and markers of intestinal inflammation in Brazilian children. Trans R Soc Trop Med Hyg. 2008;102:718–25. doi: 10.1016/j.trstmh.2008.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Asher AJ, Holt DC, Andrews RM, Power ML. Distribution of Giardia duodenalis Assemblages, A and B among Children Living in a Remote Indigenous Community of the Northern Territory, Australia. PLoS One. 2014;9:e112058. doi: 10.1371/journal.pone.0112058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Helmy YA, Klotz C, Wilking H, Jürgen K, Nöckler K, Von Samson-Himmelstjerna G, Zessin KH, Aebischer T. Epidemiology of Giardia duodenalis infection in ruminant livestock and children in the Ismailia province of Egypt: insights by genetic characterization. Parasit Vectors. 2014;7:321. doi: 10.1186/1756-3305-7-321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Choy SH, Al-Mekhlafi HM, Mahdy MA, Nasr NN, Sulaiman M, Lim YAL, Surin J. Prevalence and associated risk factors of Giardia infection among indigenous communities in rural Malaysia. Sci Rep. 2014;4:6909. doi: 10.1038/srep06909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Adam RD, Dahlstrom EW, Martens CA, Bruno DP, Barbia KD, Ricklefs SM, Hernandez MM, Narla NP, Patel RB, Porcella SF, Nash TE. Genome sequencing of Giardia lamblia genotypes A2 and B isolates and comparative analysis with the genomes of genotypes A1 and E (WB and Pig) Genome Biol Evol. 2013;5:2498–511. doi: 10.1093/gbe/evt197. [DOI] [PMC free article] [PubMed] [Google Scholar]; http://f1000.com/prime/718199716
  • 92.Haque R, Roy S, Kabir M, Stroup SE, Mondal D, Houpt ER. Giardia assemblage A infection and diarrhea in Bangladesh. J Infect Dis. 2005;192:2171–3. doi: 10.1086/498169. [DOI] [PubMed] [Google Scholar]
  • 93.Pestechian N, Rasekh H, Rostami-Nejad M, Yousofi HA, Hosseini-Safa A. Molecular identification of Giardia lamblia; is there any correlation between diarrhea and genotyping in Iranian population? Gastroenterol Hepatol Bed Bench. 2014;7:168–72. [PMC free article] [PubMed] [Google Scholar]
  • 94.ElBakri A, Samie A, Bessong P, Potgieter N, Odeh RA. Detection and molecular characterization of Giardia lamblia genotypes in Sharjah, United Arab Emirates. Trans R Soc Trop Med Hyg. 2014;108:466–73. doi: 10.1093/trstmh/tru083. [DOI] [PubMed] [Google Scholar]
  • 95.Puebla LJ, Núñez FA, Fernández YA, Fraga J, Rivero LR, Millán IA, Valdés, Silva Im. Correlation of Giardia duodenalis assemblages with clinical and epidemiological data in Cuban children. Infect Genet Evol. 2014;23:7–12. doi: 10.1016/j.meegid.2014.01.007. [DOI] [PubMed] [Google Scholar]
  • 96.Laishram S, Kannan A, Rajendran P, Kang G, Ajjampur SS. Mixed Giardia duodenalis assemblage infections in children and adults in South India. Epidemiol Infect. 2012;140:2023–7. doi: 10.1017/S0950268811002767. [DOI] [PubMed] [Google Scholar]
  • 97.Broglia A, Weitzel T, Harms G, Cacció SM, Nöckler K. Molecular typing of Giardia duodenalis isolates from German travellers. Parasitol Res. 2013;112:3449–56. doi: 10.1007/s00436-013-3524-y. [DOI] [PubMed] [Google Scholar]
  • 98.Liu A, Yang F, Shen Y, Zhang W, Wang R, Zhao W, Zhang L, Ling H, Cao J. Genetic analysis of the Gdh and Bg genes of animal-derived Giardia duodenalis isolates in Northeastern China and evaluation of zoonotic transmission potential. PLoS One. 2014;9:e95291. doi: 10.1371/journal.pone.0095291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Wang H, Zhao G, Chen G, Jian F, Zhang S, Feng C, Wang R, Zhu J, Dong H, Hua J, Wang M, Zhang L. Multilocus genotyping of Giardia duodenalis in dairy cattle in Henan, China. PLoS One. 2014;9:e100453. doi: 10.1371/journal.pone.0100453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Byrd LG, Conrad JT, Nash TE. Giardia lamblia infections in adult mice. Infect Immun. 1994;62:3583–5. doi: 10.1128/iai.62.8.3583-3585.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Adell AD, Miller WA, Harvey DJ, Van Wormer E, Wuertz S, Conrad PA. Individual subject meta-analysis of parameters for Giardia duodenalis shedding in animal experimental models. Biomed Res Int. 2014;2014:476142. doi: 10.1155/2014/476142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Lee HY, Hyung S, Lee NY, Yong TS, Han SH, Park SJ. Excretory-secretory products of Giardia lamblia induce interleukin-8 production in human colonic cells via activation of p38, ERK1/2, NF-κB and AP-1. Parasite Immunol. 2012;34:183–98. doi: 10.1111/j.1365-3024.2012.01354.x. [DOI] [PubMed] [Google Scholar]
  • 103.Cotton JA, Bhargava A, Ferraz JG, Yates RM, Beck PL, Buret AG. Giardia duodenalis cathepsin B proteases degrade intestinal epithelial interleukin-8 and attenuate interleukin-8-induced neutrophil chemotaxis. Infect Immun. 2014;82:2772–87. doi: 10.1128/IAI.01771-14. [DOI] [PMC free article] [PubMed] [Google Scholar]; http://f1000.com/prime/718351096
  • 104.Cotton JA, Motta JP, Schenck LP, Hirota SA, Beck PL, Buret AG. Giardia duodenalis infection reduces granulocyte infiltration in an in vivo model of bacterial toxin-Induced colitis and attenuates inflammation in human intestinal tissue. PLoS One. 2014;9:e109087. doi: 10.1371/journal.pone.0109087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Koh WH, Geurden T, Paget T, O'Handley R, Steuar RF, Thompson RCA, Buret AG. Giardia duodenalis assemblage-specific induction of apoptosis and tight junction disruption in human intestinal epithelial cells: effects of mixed infections. J Parasitol. 2013;99:353–8. doi: 10.1645/GE-3021.1. [DOI] [PubMed] [Google Scholar]
  • 106.Tysnes KR, Robertson LJ. Investigation of effects of Giardia duodenalis on transcellular and paracellular transport in enterocytes using in vitro Ussing chamber experiments. Parasitology. 2015;142:691–7. doi: 10.1017/S0031182014001772. [DOI] [PubMed] [Google Scholar]
  • 107.Li W, Saraiya AA, Wang CC. Experimental verification of the identity of variant-specific surface proteins in Giardia lamblia trophozoites. MBio. 2013;4:e00321–13. doi: 10.1128/mBio.00321-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Emery SJ, van Sluyter S, Haynes PA. Proteomic analysis in Giardia duodenalis yields insights into strain virulence and antigenic variation. Proteomics. 2014;14:2523–34. doi: 10.1002/pmic.201400144. [DOI] [PubMed] [Google Scholar]
  • 109.Touz MC, Rópolo AS, Rivero MR, Vranych CV, Conrad JT, Svard SG, Nash TE. Arginine deiminase has multiple regulatory roles in the biology of Giardia lamblia. J Cell Sci. 2008;121:2930–8. doi: 10.1242/jcs.026963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Das S, Schteingart CD, Hofmann AF, Reiner DS, Aley SB, Gillin FD. Giardia lamblia: evidence for carrier-mediated uptake and release of conjugated bile acids. Exp Parasitol. 1997;87:133–41. doi: 10.1006/expr.1997.4197. [DOI] [PubMed] [Google Scholar]
  • 111.Erlandsen S. Reduction in fecal excretion of Giardia cysts: effect of cholestasis and diet. J Parasitol. 2005;91:1482–4. doi: 10.1645/GE-3497RN.1. [DOI] [PubMed] [Google Scholar]
  • 112.de la Guardia RD, Lopez MB, Burgos M, Osuna A. Purification and characterization of a protein capable of binding to fatty acids and bile salts in Giardia lamblia. J Parasitol. 2011;97:642–47. doi: 10.1645/GE-2469.1. [DOI] [PubMed] [Google Scholar]
  • 113.Wampfler PB, Tosevski V, Nanni P, Spycher C, Hehl AB. Proteomics of secretory and endocytic organelles in Giardia lamblia. PLoS One. 2014;9:e94089. doi: 10.1371/journal.pone.0094089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Woessner DJ, Dawson SC. The Giardia median body protein is a ventral disc protein that is critical for maintaining a domed disc conformation during attachment. Eukaryot Cell. 2012;11:292–301. doi: 10.1128/EC.05262-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Schwartz CL, Heumann JM, Dawson SC, Hoenger A. A detailed, hierarchical study of Giardia lamblia's ventral disc reveals novel microtubule-associated protein complexes. PLoS One. 2012;7:e43783. doi: 10.1371/journal.pone.0043783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Lenaghan SC, Davis CA, Henson WR, Zhang Z, Zhang M. High-speed microscopic imaging of flagella motility and swimming in Giardia lamblia trophozoites. Proc Nat Acad Sci. 2011;108:E551. doi: 10.1073/pnas.1106904108. [DOI] [PMC free article] [PubMed] [Google Scholar]; http://f1000.com/prime/723903174
  • 117.Stadelmann B, Merino MC, Persson L, Svärd SG. Arginine consumption by the intestinal parasite Giardia intestinalis reduces proliferation of intestinal epithelial cells. PLoS One. 2012;7:e45325. doi: 10.1371/journal.pone.0045325. [DOI] [PMC free article] [PubMed] [Google Scholar]; http://f1000.com/prime/724152297
  • 118.Ventura LL, Oliveira DR, Viana JC, Santos JF, Caliari MV, Gomes MA. Impact of protein malnutrition on histological parameters of experimentally infected animals with Giardia lamblia. Exp Parasitol. 2013;133:391–5. doi: 10.1016/j.exppara.2013.01.007. [DOI] [PubMed] [Google Scholar]
  • 119.Eckmann L, Laurent F, Langford TD, Hetsko ML, Smith JR, Kagnoff MF, Gillin FD. Nitric oxide production by human intestinal epithelial cells and competition for arginine as potential determinants of host defense against the lumen-dwelling pathogen Giardia lamblia. J Immunol. 2000;164:1478–87. doi: 10.4049/jimmunol.164.3.1478. [DOI] [PubMed] [Google Scholar]
  • 120.Stadelmann B, Hanevik K, Andersson MK, Bruserud O, Svärd SG. The role of arginine and arginine-metabolizing enzymes during Giardia - host cell interactions in vitro. BMC Microbiol. 2013;13:256. doi: 10.1186/1471-2180-13-256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Azevedo OG, Bolick DT, Roche JK, Pinkerton RF, Lima AAM, Vitek MP, Warren CA, Oriá RB, Guerrant RL. Apolipoprotein E plays a key role against cryptosporidial infection in transgenic undernourished mice. PLoS One. 2014;9:e89562. doi: 10.1371/journal.pone.0089562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Oriá RB, Patrick PD, Oriá MO, Lorntz B, Thompson MR, Azevedo OGR, Lobo RNB, Pinkerton RF, Guerrant RL, Lima AAM. ApoE polymorphisms and diarrheal outcomes in Brazilian shanty town children. Braz J Med Biol Res. 2010;43:249–56. doi: 10.1590/S0100-879X2010007500003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Mitter SS, Oriá RB, Kvalsund MP, Pamplona P, Joventino ES, Mota RMS, Gonçalves DC, Patrick PD, Guerrant RL, Lima AA. Apolipoprotein E4 influences growth and cognitive responses to micronutrient supplementation in shantytown children from northeast Brazil. Clinics (Sao Paulo) 2012;67:11–8. doi: 10.6061/clinics/2012(01)03. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Yahya RS, Awad SI, Elgayar EK, Elborii H, Ragab A, Al-Sawah GA. Impact of apolipoprotein E4 on development and cognitive function in Giardia-infected children. Internet J Parasit Dis. 2009;4 [Google Scholar]
  • 125.Haque R, Snider C, Liu Y, Ma JZ, Liu L, Nayak U, Mychaleckyj JC, Korpe P, Mondal D, Kabir M, Alam M, Pallansch M, Oberste MS, Weldon W, Kirkpatrick BD, Petri WA. Oral polio vaccine response in breast fed infants with malnutrition and diarrhea. Vaccine. 2014;32:478–82. doi: 10.1016/j.vaccine.2013.11.056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Costa LB, Noronha FJ, Roche JK, Sevilleja JE, Warren CA, Oriá RB, Lima A, Guerrant RL. Novel in vitro and in vivo models and potential new therapeutics to break the vicious cycle of Cryptosporidium infection and malnutrition. J Infect Dis. 2012;205:1464–71. doi: 10.1093/infdis/jis216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Coutinho BP, Oriá RB, Vieira CM, Sevilleja JEAD, Warren CA, Maciel JG, Thompson MR, Pinkerton RC, Lima AAM, Guerrant RL. Cryptosporidium infection causes undernutrition and, conversely, weanling undernutrition intensifies infection. J Parasitol. 2008;94:1225–32. doi: 10.1645/GE-1411.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Costa LB, JohnBull EA, Reeves JT, Sevilleja JE, Freire RS, Hoffman PS, Lima AAM, Oriá RB, Roche JK, Guerrant RL, Warren CA. Cryptosporidium-malnutrition interactions: mucosal disruption, cytokines, and TLR signaling in a weaned murine model. J Parasitol. 2011;97:1113–20. doi: 10.1645/GE-2848.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Bolick DT, Roche JK, Hontecillas R, Bassaganya-Riera J, Nataro JP, Guerrant RL. Enteroaggregative Escherichia coli strain in a novel weaned mouse model: exacerbation by malnutrition, biofilm as a virulence factor and treatment by nitazoxanide. J Med Microbiol. 2013;62:896–905. doi: 10.1099/jmm.0.046300-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Iñigo-Figueroa G, Méndez-Estrada RO, Quihui-Cota L, Velásquez-Contreras CA, Garibay-Escobar A, Canett-Romero R, Astiazarán-García Effects of dietary zinc manipulation on growth performance, zinc status and immune response during Giardia lamblia infection: a study in CD-1 mice. Nutrients. 2013;5:3447–60. doi: 10.3390/nu5093447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Bolick DT, Kolling GL, Moore JH, de Oliveira LA, Tung K, Philipson C, Viladomiu M, Hontecillas R, Bassaganya-Riera J, Guerrant RL. Zinc deficiency alters host response and pathogen virulence in a mouse model of enteroaggregative Escherichia coli-induced diarrhea. Gut Microbes. 2014;5:618–27. doi: 10.4161/19490976.2014.969642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Subramanian S, Huq S, Yatsunenko T, Haque R, Mahfuz M, Alam MA, Benezra A, DeStefano J, Meier M, Muegge BD, Barratt MJ, VanArendonk LG, Zhang Q, Province MA, Petri WA, Ahmed T, Gordon JI. Persistent gut microbiota immaturity in malnourished Bangladeshi children. Nature. 2014;510:417–21. doi: 10.1038/nature13421. [DOI] [PMC free article] [PubMed] [Google Scholar]; http://f1000.com/prime/718433659
  • 133.Singer SM, Nash TE. The role of normal flora in Giardia lamblia infections in mice. J Infect Dis. 2000;181:1510–2. doi: 10.1086/315409. [DOI] [PubMed] [Google Scholar]; http://f1000.com/prime/725450155
  • 134.Morken MH, Nysaeter G, Strand EA, Hausken T, Berstad A. Lactulose breath test results in patients with persistent abdominal symptoms following Giardia lamblia infection. Scand J Gastroenterol. 2008;43:141–5. doi: 10.1080/00365520701673960. [DOI] [PubMed] [Google Scholar]
  • 135.Torres MF, Uetanabaro AP, Costa AF, Alves CA, Faria LM, Bambirra EA, Penna FJ, Vieira EC, Nicoli JR. Influence of bacteria from the duodenal microbiota of patients with symptomatic giardiasis on the pathogenicity of Giardia duodenalis in gnotoxenic mice. J Med Microbiol. 2000;49:209–15. doi: 10.1099/0022-1317-49-3-209. [DOI] [PubMed] [Google Scholar]
  • 136.Naess H, Nyland M, Hausken T, Follestad I, Nyland HI. Chronic fatigue syndrome after Giardia enteritis: clinical characteristics, disability and long-term sickness absence. BMC Gastroenterol. 2012;12:13. doi: 10.1186/1471-230X-12-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Mørch K, Hanevik K, Rortveit G, Wensaas KA, Eide GE, Hausken T, Langeland N. Severity of Giardia infection associated with post-infectious fatigue and abdominal symptoms two years after. BMC Infect Dis. 2009;9:206. doi: 10.1186/1471-2334-9-206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Fisher BS, Estraño CE, Cole JA. Modeling long-term host cell-Giardia lamblia interactions in an in vitro co-culture system. PLoS One. 2013;8:e81104. doi: 10.1371/journal.pone.0081104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Mastronicola D, Falabella M, Testa F, Pucillo LP, Teixeira M, Sarti P, Saraiva LM, Giuffré A. Functional characterization of peroxiredoxins from the human protozoan parasite Giardia intestinalis. PLoS Negl Trop Dis. 2014;8:e2631. doi: 10.1371/journal.pntd.0002631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Ferella M, Davids BJ, Cipriano MJ, Birkeland SR, Palm D, Gillin FD, McCarthur AG, Svärd S. Gene expression changes during Giardia-host cell interactions in serum-free medium. Mol Biochem Parasitol. 2014;197:21–3. doi: 10.1016/j.molbiopara.2014.09.007. [DOI] [PubMed] [Google Scholar]
  • 141.Humen MA, De Antoni GL, Benyacoub J, Costas ME, Cardozo MI, Kozubsky L, Saudan KY, Boenzli-Bruand A, Blum S, Schiffrin EJ, Pérez PF. Lactobacillus johnsonii La1 antagonizes Giardia intestinalis in vivo. Infect Immun. 2005;73:1265–9. doi: 10.1128/IAI.73.2.1265-1269.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]; http://f1000.com/prime/1024442
  • 142.Shukla G, Sidhu RK, Verma A. Restoration of anthropometric, biochemical and histopathological alterations by Lactobacillus casei supplementation in Giardia intestinalis infected renourished BALB/c mice. Antonie Van Leeuwenhoek. 2012;102:61–72. doi: 10.1007/s10482-012-9713-3. [DOI] [PubMed] [Google Scholar]
  • 143.Goyal N, Shukla G. Probiotic Lactobacillus rhamnosus GG modulates the mucosal immune response in Giardia intestinalis-infected BALB/c mice. Dig Dis Sci. 2013;58:1218–25. doi: 10.1007/s10620-012-2503-y. [DOI] [PubMed] [Google Scholar]
  • 144.Goyal N, Rishi P, Shukla G. Lactobacillus rhamnosus GG antagonizes Giardia intestinalis induced oxidative stress and intestinal disaccharidases: an experimental study. World J Microbiol Biotechnol. 2013;29:1049–57. doi: 10.1007/s11274-013-1268-6. [DOI] [PubMed] [Google Scholar]
  • 145.Pérez PF, Minnaard J, Rouvet M, Knabenhans C, Brassart D, De Antoni GL, Schiffrin EJ. Inhibition of Giardia intestinalis by extracellular factors from Lactobacilli: an in vitro study. Appl Environ Microbiol. 2001;67:5037–42. doi: 10.1128/AEM.67.11.5037-5042.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Amer EI, Mossallam SF, Mahrous H. Therapeutic enhancement of newly derived bacteriocins against Giardia lamblia. Exp Parasitol. 2014;146:52–63. doi: 10.1016/j.exppara.2014.09.005. [DOI] [PubMed] [Google Scholar]
  • 147.Franco MC, Golowczyc MA, De Antoni GL, Pérez PF, Humen M, Serradell Mde L. Administration of kefir-fermented milk protects mice against Giardia intestinalis infection. J Med Microbiol. 2013;62:1815–22. doi: 10.1099/jmm.0.068064-0. [DOI] [PubMed] [Google Scholar]
  • 148.Obendorf J, Renner Viveros P, Fehlings M, Klotz C, Aebischer T, Ignatius R. Increased expression of CD25, CD83, and CD86, and secretion of IL-12, IL-23, and IL-10 by human dendritic cells incubated in the presence of Toll-like receptor 2 ligands and Giardia duodenalis. Parasit Vectors. 2013;6:317. doi: 10.1186/1756-3305-6-317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Kamda JD, Singer SM. Phosphoinositide 3-kinase-dependent inhibition of dendritic cell interleukin-12 production by Giardia lamblia. Infect Immun. 2009;77:685–93. doi: 10.1128/IAI.00718-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Grit GH, Devriendt B, Van Coppernolle S, Geurden T, Hope J, Vercruysse J, Cox E, Geldhof P, Claerebout E. Giardia duodenalis stimulates partial maturation of bovine dendritic cells associated with altered cytokine secretion and induction of T-cell proliferation. Parasite Immunol. 2014;36:157–69. doi: 10.1111/pim.12095. [DOI] [PubMed] [Google Scholar]
  • 151.Mukherjee AK, Chowdhury P, Rajendran K, Nozaki T, Ganguly S. Association between Giardia duodenalis and coinfection with other diarrhea-causing pathogens in India. Biomed Res Int. 2014;2014:786480. doi: 10.1155/2014/786480. [DOI] [PMC free article] [PubMed] [Google Scholar]; http://f1000.com/prime/725450156
  • 152.Sulis G, Villanacci V, Missale G, Salemme M, Castelli F, Caligaris S. Whipple's disease concomitant with candida esophagitis and subsequent Giardia lamblia coinfection. Eur J Gastroenterol Hepatol. 2014;26:1181–5. doi: 10.1097/MEG.0000000000000154. [DOI] [PubMed] [Google Scholar]
  • 153.Vasco G, Trueba G, Atherton R, Calvopiña M, Cevallos W, Andrade T, Eguiguren M, Eisenberg JNS. Identifying etiological agents causing diarrhea in low income Ecuadorian communities. Am J Trop Med Hyg. 2014;91:563–9. doi: 10.4269/ajtmh.13-0744. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Cotton JA, Beatty JK, Buret AG. Host parasite interactions and pathophysiology in Giardia infections. Int J Parasitol. 2011;41:925–33. doi: 10.1016/j.ijpara.2011.05.002. [DOI] [PubMed] [Google Scholar]; http://f1000.com/prime/725450157
  • 155.Roxström-Lindquist K, Ringqvist E, Palm D, Svärd S. Giardia lamblia-induced changes in gene expression in differentiated Caco-2 human intestinal epithelial cells. Infect Immun. 2005;73:8204–8. doi: 10.1128/IAI.73.12.8204-8208.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Kamda JD, Nash TE, Singer SM. Giardia duodenalis: dendritic cell defects in IL-6 deficient mice contribute to susceptibility to intestinal infection. Exp Parasitol. 2012;130:288–91. doi: 10.1016/j.exppara.2012.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Zhou P, Li E, Zhu N, Robertson J, Nash T, Singer SM. Role of interleukin-6 in the control of acute and chronic Giardia lamblia infections in mice. Infect Immun. 2003;71:1566–8. doi: 10.1128/IAI.71.3.1566-1568.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Li E, Zhou P, Petrin Z, Singer SM. Mast cell-dependent control of Giardia lamblia infections in mice. Infect Immun. 2004;72:6642–9. doi: 10.1128/IAI.72.11.6642-6649.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Tako EA, Hassimi MF, Li E, Singer SM. Transcriptomic analysis of the host response to Giardia duodenalis infection reveals redundant mechanisms for parasite control. MBio. 2013;4:e00660–13. doi: 10.1128/mBio.00660-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Li E, Zhou P, Singer SM. Neuronal nitric oxide synthase is necessary for elimination of Giardia lamblia infections in mice. J Immunol. 2006;176:516–21. doi: 10.4049/jimmunol.176.1.516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Andersen YS, Gillin FD, Eckmann L. Adaptive immunity-dependent intestinal hypermotility contributes to host defense against Giardia spp. Infect Immun. 2006;74:2473–6. doi: 10.1128/IAI.74.4.2473-2476.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Ljungström I, Castor B. Immune response to Giardia lamblia in a water-borne outbreak of giardiasis in Sweden. J Med Microbiol. 1992;36:347–52. doi: 10.1099/00222615-36-5-347. [DOI] [PubMed] [Google Scholar]
  • 163.Jiménez JC, Fontaine J, Creusy C, Fleurisse L, Grzych JM, Capron M, Dei-Cas E. Antibody and cytokine responses to Giardia excretory/secretory proteins in Giardia intestinalis-infected BALB/c mice. Parasitol Res. 2014;113:2709–18. doi: 10.1007/s00436-014-3927-4. [DOI] [PubMed] [Google Scholar]
  • 164.Ebert EC. Giardia induces proliferation and interferon gamma production by intestinal lymphocytes. Gut. 1999;44:342–6. doi: 10.1136/gut.44.3.342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Hanevik K, Kristoffersen E, Svard S, Bruserud O, Ringqvist E, Sørnes S, Langeland N. Human cellular immune response against Giardia lamblia 5 years after acute giardiasis. J Infect Dis. 2011;204:1779–86. doi: 10.1093/infdis/jir639. [DOI] [PubMed] [Google Scholar]; http://f1000.com/prime/725450158
  • 166.Isaac-Renton JL, Lewis LF, Ong CS, Nulsen MF. A second community outbreak of waterborne giardiasis in Canada and serological investigation of patients. Trans R Soc Trop Med Hyg. 1994;88:395–9. doi: 10.1016/0035-9203(94)90397-2. [DOI] [PubMed] [Google Scholar]
  • 167.Ament ME, Rubin CE. Relation of giardiasis to abnormal intestinal structure and function in gastrointestinal immunodeficiency syndromes. Gastroenterology. 1972;62:216–26. [PubMed] [Google Scholar]
  • 168.Perlmutter DH, Leichtner AM, Goldman H, Winter HS. Chronic diarrhea associated with hypogammaglobulinemia and enteropathy in infants and children. Dig Dis Sci. 1985;30:1149–55. doi: 10.1007/BF01314049. [DOI] [PubMed] [Google Scholar]
  • 169.Oksenhendler E, Gérard L, Fieschi C, Malphettes M, Mouillot G, Jaussaud R, Viallard JF, Gardembas M, Galicier L, Schleinitz N, Suarez F, Soulas-Sprauel P, Hachulla E, Jaccard A, Gardeur A, Théodorou I, Rabian C. Infections in 252 patients with common variable immunodeficiency. Clin Infect Dis. 2008;15:1547–54. doi: 10.1086/587669. [DOI] [PubMed] [Google Scholar]
  • 170.Zinneman HH, Kaplan AP. The association of giardiasis with reduced intestinal secretory immunoglobulin A. Am J Dig Dis. 1972;17:793–7. doi: 10.1007/BF02231148. [DOI] [PubMed] [Google Scholar]
  • 171.Char S, Cevallos AM, Yamson P, Sullivan PB, Neale G, Farthing MJ. Impaired IgA response to Giardia heat shock antigen in children with persistent diarrhoea and giardiasis. Gut. 1993;34:38–40. doi: 10.1136/gut.34.1.38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Langford TD, Housley MP, Boes M, Chen J, Kagnoff MF, Gillin FD, Eckmann L. Central Importance of Immunoglobulin A in Host Defense against Giardia spp. Infect Immun. 2002;70:11–18. doi: 10.1128/IAI.70.1.11-18.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]; http://f1000.com/prime/1004758
  • 173.Davids BJ, Palm JE, Housley MP, Smith JR, Andersen YS, Martin MG, Hendrickson BA, Johansen FE, Svärd SG, Gillin FD, Eckmann L. Polymeric immunoglobulin receptor in intestinal immune defense against the lumen-dwelling protozoan parasite Giardia. J Immunol. 2006;177:6281–90. doi: 10.4049/jimmunol.177.9.6281. [DOI] [PubMed] [Google Scholar]
  • 174.Solaymani-Mohammadi S, Singer S. Regulation of Intestinal Epithelial Cell Cytoskeletal Remodeling by Cellular Immunity Following Gut Infection. Mucosal Immunol. 2013;6:369–78. doi: 10.1038/mi.2012.80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Buret A, Gall DG, Olson ME. Growth, activities of enzymes in the small intestine, and ultrastructure of microvillous border in gerbils infected with Giardia duodenalis. Parasitol Res. 1991;77:109–14. doi: 10.1007/BF00935423. [DOI] [PubMed] [Google Scholar]
  • 176.Long KZ, Rosado JL, Santos JI, Haas M, Mamun AA, DuPont HL, Nanthakumar NN, Estrada-Garcia T. Associations between mucosal innate and adaptive immune responses and resolution of diarrheal pathogen infections. Infect Immun. 2010;78:1221–8. doi: 10.1128/IAI.00767-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Anderson KA, Brooks AS, Morrison AL, Reid-Smith RJ, Martin SW, Benn DM, Peregrine AS. Impact of Giardia vaccination on asymptomatic Giardia infections in dogs at a research facility. Can Vet J. 2004;45:924–30. [PMC free article] [PubMed] [Google Scholar]
  • 178.Korpe PS, Liu Y, Siddique A, Kabir M, Ralston K, Ma JZ, Haque R, Petri WA. Breast milk parasite-specific antibodies and protection from amebiasis and cryptosporidiosis in Bangladeshi infants: a prospective cohort study. Clin Infect Dis. 2013;56:988–92. doi: 10.1093/cid/cis1044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Rivero FD, Saura A, Prucca CG, Carranza PG, Torri A, Lujan HD. Disruption of antigenic variation is crucial for effective parasite vaccine. Nature Medicine. 2010;16:551–7. doi: 10.1038/nm.2141. [DOI] [PubMed] [Google Scholar]; http://f1000.com/prime/3175959
  • 180.Jenikova G, Hruz P, Andersson KM, Tejman-Yarden N, Ferreira PCD, Andersen YS, Davids BJ, Gillin FD, Svärd SG, Curtiss R, Eckmann L. α1-giardin based live heterologous vaccine protects against Giardia lamblia infection in a murine model. Vaccine. 2011;29:9529–37. doi: 10.1016/j.vaccine.2011.09.126. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from F1000Prime Reports are provided here courtesy of Faculty of 1000 Ltd

RESOURCES