<<

Parasite Immunology, 2016, 38, 558–568 DOI: 10.1111/pim.12343

Commissioned Review Article A prospective view of animal and human

K. CWIKLINSKI,1 S. M. O’NEILL,2 S. DONNELLY3 & J. P. DALTON1

1School of Biological Sciences, Queen’s University Belfast, Belfast, UK, 2School of Biotechnology, Dublin City University, Dublin, Republic of Ireland, 3The i3 Institute & School of Medical and Molecular Biosciences, University of Technology Sydney, Sydney, NSW, Australia

SUMMARY INTRODUCTION Fasciolosis, a -borne trematodiasis, results following Among the major neglected tropical diseases (NTD) of with the parasites, hepatica and Fasciola humans are a group that result from infection with the gigantica. These trematodes greatly affect the global agri- trematode parasites mansoni, S. japonicum, cultural community, infecting millions of ruminants world- S. haematobium, S. mekongi and S. bovis (the blood wide and causing annual economic losses in excess of US $3 flukes), westermani (the fluke) and Clo- billion. Fasciolosis, an important , is classified by norchis sinensis, , WHO as a neglected tropical disease with an estimated 17 and F. gigantica (the flukes). Collectively these dis- million people infected and a further 180 million people at eases afflict >1 billion people and cause >600 million dis- risk of infection. The significant impact on agriculture and ability adjusted life years (DALYs) (1–3). Human human health together with the increasing demand for ani- are predominantly found in Africa, South mal-derived food products to support global population America, North and South Asia, China and Korea (1). growth demonstrate that fasciolosis is a major One Health Infections with the liver and lung flukes occur by inges- problem. This review details the problematic issues surround- tion, and hence, these are often classified separately as ing fasciolosis control, including drug resistance, lack of food-borne (see 1–4). In this review, we diagnosis and the threat that hybridization of the Fasciola examine the liver flukes Fasciola hepatica and F. gigantica species poses to future animal and human health. We discuss that are the most important trematodes afflicting the glo- how these parasites may mediate their long-term survival bal agricultural community. However, in the last 25 years through regulation and modulation of the immune sys- alone fasciolosis has emerged as an important zoonosis tem, by altering the host immune and/or by and NTD with an estimated 17 million people infected influencing the intestinal microbiome particularly in respect and about 180 million people living in endemic areas at to concurrent infections with other pathogens. Large gen- risk to infection (5–7). ome, transcriptome and proteomic data sets are now avail- Fasciola hepatica, usually termed the temperate fluke, is able to support an integrated One Health approach to found worldwide, on every inhabited continent, while, develop novel diagnostic and control strategies for both ani- , the tropical fluke is found in tropical mal and human disease. areas of Asia and Africa. The life cycle is essentially the same for both Fasciola species and differs only in the Keywords Fasciola hepatica, immune modulation, innate intermediate host species (8). Eggs are passed in the immunity, microbiome, serodiagnosis faeces of infected mammalian hosts and deposited into the environment, typically pastures and grazing areas near a body of water. Following a period of maturation and activation by temperature and light, the eggs hatch releas- Correspondence: Krystyna Cwiklinski, School of Biological Sciences, ing miracidia, which actively seek out the snail intermedi- Queen’s University Belfast, 97 Lisburn Rd, Belfast, BT9 7BL, UK ate host. The species of snail that act as intermediate (e-mail: [email protected]). host ( infected by F. hepatica and the Disclosures: None. African Radix natalensis and the Eurasian Radix auricu- Received: 10 February 2016 Accepted for publication: 9 June 2016 laria infected by F. gigantica) differ for each parasite

558 © 2016 The Authors. Parasite Immunology Published by John Wiley & Sons Ltd. This is an open access article under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made. Volume 38, Number 9, September 2016 Animal and human Fasciolosis species and exhibit marked differences in their geographi- although the levels of sensitivity and specificity of 87% cal distribution (9, 10). Within the snail, the parasite and 99%, respectively (19), are lower than that reported undergoes a clonal expansion through radiae and cercerial by Gonzales-Santana et al. (16). Similar ELISA tests are stages. The cercariae are released from the and available for F. gigantica using a variety of different anti- encyst on vegetation as metacercariae that can remain gens: (a) Fas1 and Fas2 cysteine proteases, resulting in viable for months. Domestic animals pick the disease up 91Á9% and 89Á1%, sensitivity and specificity, respectively by eating contaminated grass, while human infections (20); (b) sandwich ELISA using fatty acid binding protein, occur following the ingestion of infected edible aquatic resulting in 94Á7% sensitivity and 84Á62% specificities (21); salad vegetation, typically found near the infected ani- (c) 27 kDa circulating from sera of infected indi- mals, such as watercress (11, 12) or through the consump- viduals gave >93% sensitivity and specificity (22). A lateral tion of metacercariae-contaminated water (10). Upon flow test (SeroFluke) was developed by Martinez-Sernan- ingestion of infected vegetation, the parasites emerge from dez and colleagues using samples obtained from F. hepat- their cysts in the intestine, as newly excysted juveniles ica-infected patients in Spain and Portugal, for use with (NEJ) that then traverse the intestinal wall into the peri- serum or whole blood samples, which requires minimal toneal cavity before migrating to the liver capsule and training (23). However, capable of distinguishing parenchyma. Following a period of approximately 7- active infection by F. hepatica and F. gigantica are needed 8 weeks, the parasites migrate into the bile ducts where for diagnosis in areas of geographical overlap of the two they develop into sexually mature adults, releasing species, particularly if access to the adult flukes for geno- 20 000–24 000 eggs per fluke per day (13). typing is not available.

DIAGNOSIS OF HUMAN HYBRID AND/OR INTROGRESSED INFECTION POPULATIONS OF LIVER FLUKE

The exact prevalence of human fasciolosis is most likely Fasciola has the ability to self-fertilize, cross-fertilize and underestimated due to the lack of epidemiological surveys in some cases undergo parthenogenesis. These flukes are performed in potentially endemic areas. Approximately typically diploid as shown by a recent study in the UK 50% of human infections are asymptomatic and are there- (24), although triploid and mixoploid isolates have been fore not reported (14). Diagnosis of the remaining infec- observed in one animal infection in cattle in the UK tions can vary depending on the detection method used. (F. hepatica, Cullumpton strain, triploid; 25), and in Asian Faecal egg counts (FEC) are routinely used for diagnosis flukes of China, Japan, Vietnam and Korea (26–31). The of animal infections, with the FLOTAC system described triploid and mixoploid parasites are often aspermic, rely- by Cringoli and colleagues exhibiting particularly high ing on parthenogenesis for continuation of the life cycle. levels of sensitivity and accuracy (15). For human infec- In areas of Japan, Vietnam and Korea, parasites cannot tions, these methods can be inaccurate as they rely on a be classified as F. hepatica or F. gigantica using morpho- chronic infection comprising of mature adult flukes in the metrics due to the variety of intermediate forms that exist. bile ducts. Parasite burden and the sporadic nature of egg Moreover, molecular analysis of mitochondrial genes and deposition leading to miss timing of faecal sampling are intergenic genome sequences (ITS2) has found that these also weaknesses of this technique for human diagnosis. intermediate forms are hybrid species. This analysis identi- ELISA-based methods can be used as alternatives for fied individuals with nuclear DNA of one species, but the FEC, with several available that determine anti-Fasciola mitochondrial genome of the other species as well as indi- to proteins found within the parasite secretome. viduals with copies of genes derived from both species Numerous ELISAs have been reported based on the most (28–31). The ability of these species to hybridize and/or abundantly secreted groups of proteases, the cathepsin L introgress will play a role in genetic diversity within popu- cysteine proteases. Gonzales-Santana and colleagues (16) lations (32) and has the potential for crossover of anthel- have developed a recombinant cathepsin L-based ELISA minthic resistance between the two species. Furthermore, test specific for human F. hepatica infections that showed hybridization may also have serious implications for peo- 99Á9% sensitivity and 99Á9% specificity. The cathepsin L ple living in areas where the two species co-localize as this proteases are also the focus of the capture ELISA devel- could result in the emergence of more pathogenic Fasciola oped by Mezo et al. (17; MM3-SERO), which is based on isolates. A recent study by Valero and colleagues (33) a monoclonal MM3 that binds to both cathepsin showed that when directly compared in infections of L1 and L2 proteases (18). An ELISA method focussing Guirra sheep, which have similar susceptibilities to both on saposin-like protein-2 has also been developed, species, F. gigantica was found to be more pathogenic than

© 2016 The Authors. Parasite Immunology Published by John Wiley & Sons Ltd., Parasite Immunology, 38, 558–568 559 K. Cwiklinski et al. Parasite Immunology

F. hepatica. Also, a recent analysis of human infections in infections, TCBZ failure has been found in humans, with Vietnam has revealed that hybrids are also able to infect the first case being reported in 2012 in the Netherlands people (34). of a sheep farmer who became infected with F. hepatica To date, Fasciola hybrids have been identified using as a result of casual chewing of contaminated grass (43). mitochondrial genes, which act as robust markers due to Following the failed treatment with TCBZ in this case, their maternal inheritance. As mitochondrial genes are was used but with no effect, indicating that unlinked to the nuclear genome, introgression of these nitazoxanide was not effective against these potentially markers has often been shown to be greater than nuclear TCBZ-resistant parasites. Gulhan and colleagues (44) encoded markers (35). That said, these markers alone may have also reported a case of a child in Turkey that was not accurately reflect whether true hybridization/introgres- infected with F. hepatica and, did not respond to TCBZ sion occurs within Fasciola, due to different mutation rates treatment, although this report did not speculate as to across the genome. The availability of the F. hepatica gen- the mode of infection with a potentially TCBZ-resistant ome has driven the development of larger panels of mark- isolate. Similarly, Cabada and colleagues (45) have ers including SNPs (36) and microsatellites (37), which reported a group of patients, infected by ingesting water- have shown that the F. hepatica genome is highly heteroge- cress in the Cusco region of Peru that failed to respond nous. In depth analysis of these hybrids can now be car- to multiple courses of . As with the ried out across the genome to elucidate whether Winkelhagen et al. (43) case report, following TCBZ hybridization and/or introgression is occurring between treatment failure, the patients were prescribed nitazox- Asian F. hepatica and F. gigantica and at what level. Most anide, which only appeared to cure one patient, further importantly, there is a need to investigate the impact emphasizing that nitazoxanide is not effective against hybridization and/or introgression could have on human TCBZ-resistant parasites. As TCBZ resistance becomes infections, especially as more cases of human F. gigantica more prevalent in F. hepatica endemic regions, it is very infections are being reported, to determine whether the possible that more human cases of infection with TCBZ- hybrid flukes are more or less pathogenic than their resistant F. hepatica parasites will be reported which parents. poses a real problem for human treatment.

EMERGENCE OF DRUG RESISTANT IMMUNE RESPONSES AND IMMUNITY TO PARASITES: A THREAT TO ANIMALS AND LIVER FLUKE HUMANS The immune response induced during natural infection Control of Fasciola in ruminants is reliant on the use of has been well characterized in ruminants for F. hepatica drugs, particularly triclabendazole (TCBZ), although there are increasing studies on F. gigantica that which targets both the tissue damaging immature stages report similar findings (46–50). During the acute stages and mature adult stages of the parasites. TCBZ treat- of infection, cattle exhibit a mixed immune response with ment failure and/or resistance are increasingly reported elevated IL-10, TGF-b, IL-4 and IFN-c.However,as for F. hepatica infections in ruminants across Europe, infection progresses Th2/Treg immune responses become South America and Australia (38, 39) but fortunately, to more dominant (51). During the later chronic stages, date, no cases of F. gigantica TCBZ resistance have been Treg cells release cytokines that inhibit inflammatory reported. Treatment of human fasciolosis also relies on Th1/Th2 cytokines; PBMCs isolated from F. hepatica - the use of TCBZ, which was commercially available as infected cattle produced enhanced levels of IL-4 and Egaten (Novartis Pharma AG), through a joint venture IFN-c cytokines when cultured in vitro in the presence with WHO. However, it is still not licensed for use in of TGF-b and IL-10 neutralizing antibodies (51). This several countries (40), such as USA and Canada, where immune profile is similar in sheep infected with F. hepat- TCBZ is only available to US licensed physicians under ica as they also present a mixed Th1/Th2 cytokine pro- approval from the Centres for Disease Control and file in the at week 3 after infection and as Prevention (CDC) and the Food and Drug Adminstra- infection progresses enhanced gene expression of Th2 but tion (FDA) on a case-by-case basis (41). Other drugs not Th1 cytokines is observed (52). Interestingly, have also been used to treat human F. hepatica infec- although an overall systemic Th2 immune response dom- tions, including nitazoxanide, originally FDA approved inates, different cytokines are expressed at different for protozoal infections which was shown to have vary- anatomical locations; in sheep, IL-5 can be detected in ing efficacy levels depending on the study, ranging the hepatic lymph nodes, while IL-10 is primarily between 40% and 100% (42). As with the ruminant observed in the spleen (53–55), whereas in goats, IFN-c

560 © 2016 The Authors. Parasite Immunology Published by John Wiley & Sons Ltd., Parasite Immunology, 38, 558–568 Volume 38, Number 9, September 2016 Animal and human Fasciolosis and high levels of IL-4 can be detected in both the hep- were observed in CD4+ T-cell populations (81) and may atic lymph node and liver (56). explain why these cells become hyporesponsive to antigen While it has been reported that Indonesian thin-tailed stimulation in the late stages of infection (82). This anergic sheep can resist infection to F. gigantica, no natural or state, as shown by decreased cytokine responses and experimental hosts exhibit resistance to F. hepatica infec- reduced proliferative activity, could be reversed with the tion, and successful trickle infection in cattle demonstrates addition of IL-2 to cultures (81). The presence of anergic a lack of concomitant immunity (57–59). This suggests T cells is yet to be demonstrated in ruminants (and that the ability of F. hepatica to successfully infect a broad humans); however, the lack of IL-2 reported in the local spectrum of mammalian hosts across the globe (e.g. cattle, HLN of infected sheep supports such a mechanism of sheep, goats, buffaloes, kangaroos, capybara, camelids and immune inactivation (54, 55). humans, see (4, 10) can be attributed partly to the devel- opment of effective immune-modulatory mechanisms to ALTERED IMMUNE HOMEOSTASIS DURING prevent the normal protective response and thus ensure FASCIOLA INFECTION the parasites longevity. The potent suppression of host Th1 immune responses during active infection of both nat- The induction of wound-healing immune mechanisms ural hosts and experimental rodent models has been (Th2, M2 macrophages, mast cells) by F. hepatica is attributed to the development of a strong regulatory/Th2- clearly important to protect vital tissues, particularly the type immune response (51, 54, 56, 60–66). liver, from damage caused by the migratory activity of the While Th2 and regulatory T-cell cytokines are impor- parasite (83–86), while the regulation/suppression of pro- tant in downplaying host protective Th1 responses during tective pro-inflammatory type 1 immune responses is cen- infection with F. hepatica, it seems that the parasite also tral to the promotion of the host’s tolerance to the influences various cells of the innate immune response. parasite, supporting its long-term survival (87, 88). How- Firstly, in experimental mouse models, CD11c+ dendritic ever, helminth parasites are rarely the sole infecting organ- cell (DC) populations are increased during Fasciola infec- ism within a mammalian host, particularly in endemic tion displaying an immature phenotype with lower expres- regions where co-infections are common (89). Therefore, sion of co-stimulatory markers (CD40, CD80 and CD86), immune suppressive effects of F. hepatica are not confined MHC class II, increased expression of CCR5 and are to the host-parasite relationship but likely have broader hyporesponsive to TLR activation (67). These cells express implications to the induction of type 1 immune responses enhanced levels of intracellular IL-10 and ex vivo suppress necessary to mediate resistance (induced by infection or the secretion of antigen specific IL-17 and IFN-c from vaccination) to major coexisting pathogens. For example, na€ıve DO11.10 OVA TCR Tg CD4+ T cells independent of cattle infected with F. hepatica were more susceptible to IL-10 and TGF-b (68). Secondly, the induction of macro- infections with dublin and took longer to clear phages with a regulatory/M2 phenotype is common in the than nonhelminth-infected animals (90). Simi- both large animals and rodents infections (69–72). This larly, concurrent infection of mice with F. hepatica and switch occurs within the first 3 days of murine infection Bordetella pertussis resulted in a prolonged bacterial lung and similar to DCs activated by Fasciola , M2 infection (60). macrophages are hyporesponsive to TLR ligands, suggest- Importantly, epidemiological surveys indicate that ing a reduced ability to promote the differentiation of host hyperendemic regions (>10%) for infection with Fasciola Th1 immunity. In addition, it has been shown that M2 overlap with the geographical distribution of human macrophages isolated from mice during infection with microbial pathogens implying a significant impact on F. hepatica promote the polarization of Th2 cells (69, 70). human health. For example, aside from fasciolosis, the Thirdly, there is a significant increase in the number of most common childhood illnesses among the Aymara peo- mast cells observed at the site of infection and in the gut ple of the Northern Altiplano of Bolivia are upper respira- mucosa (73–77). While mast cells are critical to the expul- tory infections such as whooping and tuberculosis sion of gut helminths (78), their role in Fasciola infection (91). These children are also infected with a range of other is not clear, although we hypothesize that given that mast protozoal and helminth parasitic infections, with a signifi- cells have an important role in wound healing and tissue cant positive association for F. hepatica and Giardia remodelling (79), they are recruited to combat the exten- intestinalis co-infections (92). Fasciola is also emerging as sive tissue damage caused by migratory flukes (80). a human pathogen in communities of Iran’s Guilan Pro- It also appears that during infection of mice with vince (93, 94) that are already afflicted by malaria and F. hepatica, T cells are induced to enter an anergic state as tuberculosis (95, 96). Although there are currently no markers of anergy (GRAIL, EGR2, ICOS and ) studies on the immune status of humans co-infected with

© 2016 The Authors. Parasite Immunology Published by John Wiley & Sons Ltd., Parasite Immunology, 38, 558–568 561 K. Cwiklinski et al. Parasite Immunology

Fasciola and other pathogens, clinical studies with other through the production of immune modulating metabolites helminths (97–101) suggest that modulation of host such as short-chain fatty acids (SCFA; 129, 130). The responses by helminth infection increases susceptibility to most abundant of these are butyrate, propionate and acet- microbial pathogens and impairs efficacy. More ate produced by the bacterial fermentation of plant- extensive longitudinal human studies are required to fully derived nondigestable polysaccharides, like cellulose, in the determine the effect of Fasciola infection on the pathogen- gut (131, 132). Treatment of both murine macrophages esis of microbial pathogens within endemic populations. and dendritic cells with bacterial SCFAs is associated with Beyond this specific impact on immune response to con- a decrease in the production of pro-inflammatory cytoki- current infections, there is a growing body of evidence sug- nes such as IL-12 and TNF and increased expression of gesting that the constant presence of helminth parasites IL-10 (129, 133, 134). Similarly, human peripheral blood (particularly intestinal soil transmitted species) within the mononuclear cells exposed to SCFAs display a reduced human population since prehistoric times has strongly ability to produce TNF and IFN-c in response to treat- shaped the evolution of the human immune system (102). ment with lipopolysaccharide (135). Furthermore, SCFAs Indeed, the need to overcome the regulatory mechanisms appear to regulate production of pro-inflammatory cytoki- exerted by helminths appears to have resulted in compen- nes in nonimmune cells such as Caco-2 cells (136). In satory adjustments to immune-related genes (103, 104). addition, these SCFAs also contribute to the induction of Accordingly, in populations where parasitic infections are regulatory T cells, either directly through interaction with no longer endemic, there is an increased prevalence of the G-protein-coupled receptor GPR43 (137) or indirectly inappropriate immune responsiveness to auto-antigens and via activation of regulatory macrophages, which in turn allergens and the concomitant development of autoim- generate a population of FoxP3+ T cells (121). It is inter- mune/inflammatory diseases (105, 106). The epidemiologi- esting that parasite-associated microbiota produce an cal pattern of fasciolosis is more varied than that seen for increased amount of SCFAs, which have been shown to intestinal helminth infection. While low and stable levels mediate an increase in the proportion of lung regulatory T of Fasciola exist within small, defined populations, spo- cells and as a result attenuate allergic airway radic outbreaks are associated with climatic changes that (138). Although some studies in humans confirm that boost the life cycle of the parasite and/or intermediate there is a difference in the gut microbiome community snail host (107–109). Therefore, it is presently unclear and the production of SCFA in helminth-infected individ- whether infection with either F. hepatica or F. gigantica uals (139–141), the data are not consistent across popula- provides any such immune benefit to its host. tions, likely due to sample size, differences in diet and The possibility that helminth infection influences other yet unknown confounding factors. immune homeostasis by mediating changes to intestinal F. hepatica spends a short time in the gut during infec- microbiota has not been extensively explored, even though tion and is typically localized to the duodenum (8), which it is now clear that this commensal community profoundly suggests that this parasite may not significantly influence impacts the homeostasis of innate and adaptive immune the composition of the gut microbiota. Instead, Fasciola responses and thus the development of immune-mediated parasites take a number of weeks migrating through the disease (110–116). Changes in gut microbiota, attributed liver of its mammalian host before ultimately residing in to the use of antibiotics, particularly in childhood, have the for many years from where they can mediate been correlated with incidences of multiple sclerosis, systemic immune modulation; the adult parasite is an obli- rheumatoid arthritis, inflammatory bowel disease and gate blood feeder, an activity likely to result in the contin- (116, 117). Furthermore, experimental studies show ued release of parasite excreted/secreted (ES) products into that changes to the intestinal microbiota of mice strongly the host circulation. Like SCFAs, these parasite ES prod- influence the development of allergy, inflammatory bowel ucts actively suppress the production of pro-inflammatory disease, rheumatoid arthritis, experimental autoimmune cytokines by most cells of the innate immune system, encephalitis and type 1 diabetes (118–123). Controlled resulting in the systemic switch towards a regulatory/Th2 infections of animals have shown that the presence of immune environment (67, 69, 70, 142). intestinal parasites results in changes to the composition Additionally, the presence of the parasite appears to and abundance of intestinal microbiota species (124–127). influence the composition of bile acids (143). Bile acids However, the use of anthelminthics suggests that the con- act as signalling molecules through either TGR5 or FXR tinuing presence of the parasite may be required for sus- to regulate intestinal homeostasis via the inhibition of tained changes to the bacterial community (128). inflammation, prevention of pathogen invasion and main- A primary mechanism by which the intestinal micro- tenance of tissue integrity (144, 145). Activation of TGR5 biota regulate immune responses is proposed to be inhibits NFjB and thus reduces the production of pro-

562 © 2016 The Authors. Parasite Immunology Published by John Wiley & Sons Ltd., Parasite Immunology, 38, 558–568 Volume 38, Number 9, September 2016 Animal and human Fasciolosis inflammatory cytokines such as IL-6 and TNF by macro- potential of dramatic genetic adaptation to new environ- phages and Kupffer cells (144–146). While signalling via ments and molecules. Analysis of the draft genome has FXR also represses NFjB-dependent transcription result- clarified several gene families of interest for vaccine and ing in an anti-inflammatory response (129, 130, 144, 145, drug target development, such as the cathepsin cysteine 147), activation of this receptor also increases the expres- proteases (L & B families), the asparaginyl endopepti- sion of several genes associated with antibacterial actions dases (legumains) and the ABC transporters, which have such as IL-18 (148). Notably, infection with F. hepatica greatly expanded and diversified biochemically and func- results in significant increases in the production of bile tionally. acids (149) and IL-18 was recently identified as the most Analysis of the differential expression across the significantly up-regulated interleukin in the liver of F. hepatica life cycle has shown that the parasite under- infected sheep (52). Such preliminary observations encour- goes rapid metabolic development, particularly during age further characterization of the composition of bile the early stages of parasite migration through the host, acid during acute and chronic infection with F. hepatica in which may be notable for future drug development. animals and humans and an assessment of the ability of Increased levels of gene expression were induced as the these to regulate the functions of innate and adaptive parasite enters and migrates through the liver. This is immune cells as well as influence the microbiome of the coupled with extensive parasite growth that may possibly gut into which they are passed. be driven by neoblast-like stem cells (162, 163). This study is further complimented by the proteomic analysis ADVANCES IN -OMIC TECHNOLOGY WILL of the secretome of these life cycle stages, which has HELP FILL THE GAPS IN OUR KNOWLEDGE revealed in particular that the majority (70%) of the secretome of the early NEJ stages is represented by 10 In the past decade, there has been major advancement proteins (Cwiklinski and Dalton, unpublished). Elucidat- in the available ‘-omics’ data for trematode parasites ing the function of these secreted proteins and their rele- (150–153), which has allowed robust comparative genome vant abundance within the secretome is key to furthering analyses between these species. Data sets are now avail- our understanding of how the parasite interacts and able for both F. hepatica and F. gigantica which has manipulates its host. greatly expanded our knowledge of these parasites, par- Similarly, for F. gigantica, transcriptome data are avail- ticularly regarding life cycle stages that have been histor- able for the adult stage parasites, which when compared ically difficult to study at the molecular level due to with F. hepatica data revealed that the predicted proteins sample sizes. These recent technological developments showed between 80 and 90% homology (based on E-value have allowed more in depth analysis of these two Fasci- 1EÀ15 and E-value 1EÀ05 cut-offs; 164). These two species ola species to further our understanding of fluke biology have been consistently shown to have high levels of simi- and how they infect and persist within their hosts. Sev- larities at the morphological and life cycle level as well as eral secretome data sets are available for both species at the gene transcription level. Comparative analysis of (154–158), which for F. hepatica are being complimented the F. gigantica genome currently being sequenced (Trema- by analysis of the exosome component of the secreted tode.net, 165) with the F. hepatica genome is critical for proteins (159, 160) as well as glycan analysis of these our comprehension of these two species and their hybrids proteins (161; Ravida et al., unpublished), allowing in to define differences that may govern species-specific depth analysis of those proteins directly interacting with pathogenesis. the host. A complete set of transcriptomes from the life Similarly, more robust analysis can now be carried out cycle stages present within the definitive host, ranging for the phylum , to reveal how these parasites from metacercariae to mature adult flukes are now avail- have developed their host/tissue preferences and immune able, allowing analysis of the extensive differential evasion strategies and how this relates to their transmis- expression that occurs within this host, particularly as sion patterns and distribution. Particularly for the liver the parasite migrates through the liver (36). These tran- flukes, namely C. sinensis, O. viverrini and Fasciola spp., scriptomes have been mapped onto the recently pub- such comparative genomics could illuminate the species- lished F. hepatica draft genome (36). Together this specific strategies of surviving within the same environ- analysis has revealed that F. hepatica has one of the lar- ment (bile duct). Furthermore, these studies should unra- gest known pathogen genomes, at 1Á3 Gbp that currently vel the mode of action of the Fasciola-specific cannot be explained simply by whole genome duplication anthelmintic TCBZ and explicate why , or expansion of repeated regions. The genome also which acts on most trematodes, has no effect on shows high levels of polymorphism, allowing for the Fasciola.

© 2016 The Authors. Parasite Immunology Published by John Wiley & Sons Ltd., Parasite Immunology, 38, 558–568 563 K. Cwiklinski et al. Parasite Immunology

FASCIOLOSIS – ONE HEALTH Figure 1 Graphical representation of the REMAINING QUESTIONS remaining questions associated with Fasciolosis as a ‘One Health’ problem. Human fasciolosis Ruminant fasciolosis • Global prevalence • Suitable immunological tools • Effective diagnosis • Mechanisms of protective immunity • Immune response • Vaccine performance in the field • Microbiome • Microbiome

Fasciola hepatica vs. Fasciola gigantica • Prevalence of each species • Does true hybridization/introgression occur? • Virulence/pathogenesis of hybrids

CONCLUSION despite excellent progress, these are still some years away. Molecules actively secreted as soluble components or in Diseases caused by F. hepatica and F. gigantica in humans extracellular vesicles are critical to how Fasciola interacts have emerged as an important NTDs, mainly in poor rural and modulates the host immune response and are consid- regions in Africa and Asia. We still do not know the full ered prime vaccine candidates (reviewed in 67, 160, 166, extent or epidemiological distribution of diseases caused 167). Development in the ‘omics’ technologies is keeping by either of these two parasites, nor do we fully under- research on Fasciola abreast with the broader field of par- stand their virulence and pathogenicity in humans. Cross- asitology and pathogen biology, and complete data sets fertilization between the species threatens the spread of (genome, transcriptome, proteome, glycome) will become hybrids with unknown capacity to infect and cause dam- available to researchers in the near future. However, many age in animals and humans, and therefore, we urgently gaps in our knowledge (e.g. mechanisms of protective require new species-specific tests for diagnosis and surveil- immunity, the relevance of the microbiome in parasite lance purposes. The spread of triclabendazole-resistant resistance and protection, vaccine performance in the field parasites is also of concern because it is the only drug that etc.) and available tools (e.g. immunologicals such as cyto- targets the tissue damaging juvenile stage, and is the only kine arrays, microarrays and multiplex cytokine qPCRs) available effective treatment for human infections. While still exist and require our collective efforts to fill (Fig- immunological studies are gradually defining immune ure 1). Moreover, there is a serious dearth of information mechanisms of infection, pathogenesis and protection, regarding the global prevalence of human fasciolosis as these are predominantly confined to animal studies and well as an understanding of immune response to infection can only be assumed to apply to humans. Nevertheless, and the pathogenesis associated with the migration of the old and recent results strongly suggest that infection with parasite through the liver and its residence in the bile Fasciola species does leave the host, animal and human, duct. susceptible to co-infection with other pathogens; this affect may be mediated via immune modulation, immunosup- ACKNOWLEDGEMENTS pression, immune polarization and/or by altering the com- position of the microbiome (gut and/or bile). All individuals listed as authors contributed equally to the The relevance of fasciolosis as a ‘One Health’ food- manuscript. KC and JPD are funded by a European borne zoonosis will grow if we do not come up with new Research Council Advanced Grant (HELIVAC, 322725) control measures. for animal liver fluke disease awarded to JPD. KC, SMO’N and JPD are members of should have a major impact on human infection, but the Horizon 2020-funded Consortium PARAGONE.

564 © 2016 The Authors. Parasite Immunology Published by John Wiley & Sons Ltd., Parasite Immunology, 38, 558–568 Volume 38, Number 9, September 2016 Animal and human Fasciolosis

REFERENCES

1 Hotez PJ, Brindley PJ, Bethony JM, King 17 Mezo M, Gonzalez-Warleta M & Ubeira 30 Itagaki T, Sakaguchi K, Terasaki K, Sasaki CH, Pearce EJ & Jacobson J. Helminth FM. The use of MM3 monoclonal antibod- O, Yoshihara S & Van Dung T. Occurrence infections: the great neglected tropical dis- ies for the early immunodiagnosis of ovine of spermic diploid and aspermic triploid eases. J Clin Invest 2008; 118: 1311–1321. fascioliasis. J Parasitol 2007; 93:65–72. forms of Fasciola in Vietnam and their 2 Keiser J & Utzinger J. Food-borne tremato- 18 Muino L, Perteguer MJ, Garate T, et al. molecular characterization based on nuclear diases. Clin Microbiol Rev 2009; 22: 466– Molecular and immunological characteriza- and mitochondrial DNA. Parasitol Int 483. tion of Fasciola antigens recognized by the 2009; 58:81–85. 3 Furst T, Duthaler U, Sripa B, Utzinger J & MM3 monoclonal antibody. Mol Biochem 31 Peng M, Ichinomiya M, Ohtori M, Ichi- Keiser J. Trematode infections: liver and Parasitol 2011; 179:80–90. kawa M, Shibahara T & Itagaki T. Molecu- lung flukes. Infect Dis Clin North Am 2012; 19 Gottstein B, Schneeberger M, Boubaker G, lar characterization of Fasciola hepatica, 26: 399–419. et al. Comparative assessment of ELISAs Fasciola gigantica, and aspermic Fasciola 4 Robinson MW & Dalton JP. Zoonotic hel- using recombinant saposin-like protein 2 and sp. in China based on nuclear and mito- minth infections with particular emphasis recombinant cathepsin L-1 from Fasciola chondrial DNA. Parasitol Res 2009; 105: on fasciolosis and other trematodiases. Phi- hepatica for the serodiagnosis of human Fas- 809–815. los Trans R Soc Lond B Biol Sci 2009; 364: ciolosis. PLoS Negl Trop Dis 2014; 8: e2860. 32 King KC, Stelkens RB, Webster JP, Smith 2763–2776. 20 Rabee I, Mahana NA & Badr AM. DF & Brockhurst MA. Hybridization in 5 Mas-Coma S. of fascioliasis Immunodiagnosis of Egyptian human fasci- parasites: consequences for adaptive evolu- in human endemic areas. J Helminthol olosis gigantica using Fas1 and Fas2 cys- tion, pathogenesis, and in a 2005; 79: 207–216. teine proteinase antigens. J Egypt Soc changing world. PLoS Pathog 2015; 11: 6 Mas-Coma S, Valero MA & Bargues MD. Parasitol 2013; 43: 787–796. e1005098. Chapter 2. Fasciola, lymnaeids and 21 Allam G, Bauomy IR, Hemyeda ZM & 33 Valero MA, Bargues MD, Khoubbane M, human fascioliasis, with a global overview Sakran TF. Evaluation of a 14.5 kDa-Fas- et al. Higher physiopathogenicity by Fasci- on disease , epidemiology, ciola gigantica fatty acid binding protein as ola gigantica than by the genetically close evolutionary genetics, molecular epidemiol- a diagnostic antigen for human fascioliasis. F. hepatica: experimental long-term follow- ogy and control. Adv Parasitol 2009; 69: Parasitol Res 2012; 110: 1863–1871. up of biochemical markers. Trans R Soc 41–146. 22 Attallah AM, Bughdadi FA, El-Shazly AM Trop Med Hyg 2016; 110:55–66. 7 Gonzalez LC, Esteban JG, Bargues MD, & Ismail H. Immunodetection of Fasciola 34 Le TH, De NV, Agatsuma T, et al. Human et al. Hyperendemic human fascioliasis in gigantica circulating antigen in sera of fascioliasis and the presence of hybrid/in- Andean valleys: an altitudinal transect infected individuals for laboratory diagnosis trogressed forms of Fasciola hepatica and analysis in children of Cajamarca province, of human fascioliasis. Clin Vaccine Immunol Fasciola gigantica in Vietnam. Int J Para- Peru. Acta Trop 2011; 120: 119–129. 2013; 20: 1569–1577. sitol 2008; 38: 725–730. 8 Andrews S. The Life cycle of Fasciola hepat- 23 Martinez-Sernandez V, Muino L, Perteguer 35 Harrison RG & Larson EL. Hybridization, ica.InDaltonJP(ed):Fasciolosis.Wallingford, MJ, et al. Development and evaluation of a introgression, and the nature of species Oxon, UK, CABI Publishing, 1999: 1–29. new lateral flow immunoassay for serodiag- boundaries. J Hered 2014; 105(Suppl 1): 9 Mas-Coma S, Bargues MD & Valero MA. nosis of human fasciolosis. PLoS Negl Trop 795–809. Fascioliasis and other plant-borne trema- Dis 2011; 5: e1376. 36 Cwiklinski K, Dalton JP, Dufresne PJ, tode zoonoses. Int J Parasitol 2005; 35: 24 Beesley NJ, Cwiklinski K, Williams DJ & et al. The Fasciola hepatica genome: gene 1255–1278. Hodgkinson J. Fasciola hepatica from natu- duplication and polymorphism reveals 10 Mas-Coma S, Bargues MD & Valero MA. rally infected sheep and cattle in Great Bri- adaptation to the host environment and the Fascioliasis. In Bruschi F (ed): Helminth tain are diploid. Parasitology 2015; 142: capacity for rapid evolution. Genome Biol Infections and their Impact on Global Public 1196–1201. 2015; 16: 71. Health. Vienna, Springer, 2014: 93–122. 25 Fletcher HL, Hoey EM, Orr N, Trudgett 37 Cwiklinski K, Allen K, LaCourse J, Wil- 11 Marsden PD. Fascioliasis in man: an out- A, Fairweather I & Robinson MW. The liams DJ, Paterson S & Hodgkinson JE. break in Hampshire. Br Med J 1960; 2: occurrence and significance of triploidy in Characterisation of a novel panel of poly- 619–625. the liver fluke, Fasciola hepatica. Parasitol- morphic microsatellite loci for the liver 12 LaPook JD, Magun AM, Nickerson KG & ogy 2004; 128:69–72. fluke, Fasciola hepatica, using a next gener- Meltzer JI. Sheep, watercress, and the inter- 26 Rhee JK, Eun GS & Lee SB. Karyotype of ation sequencing approach. Infect Genet net. Lancet 2000; 356: 218. Fasciola sp. obtained from Korean cattle. Evol 2015; 32: 298–304. 13 Boray JC. Experimental fascioliasis in Aus- Kisaengchunghak Chapchi 1987; 25:37–44. 38 Fairweather I. Reducing the future threat tralia. Adv Parasitol 1969; 7:95–210. 27 Itagaki T & Tsutsumi K. Triploid form of from (liver) fluke: realistic prospect or 14 Haswell-Elkins MR & Levri E. Food-borne Fasciola in Japan: genetic relationships quixotic fantasy? Vet Parasitol 2011; 180: Trematodes. In Cook CC, Zumla A (eds): between Fasciola hepatica and Fasciola 133–143. Manson’s Tropical Diseases. London, W.B. gigantica determined by ITS-2 sequence of 39 Hodgkinson J, Cwiklinski K, Beesley Saunders, 2003: 1471–1486. nuclear rDNA. Int J Parasitol 1998; 28: NJ, Paterson S & Williams DJ. Identifi- 15 Cringoli G, Rinaldi L, Maurelli MP & 777–781. cation of putative markers of triclaben- Utzinger J. FLOTAC: new multivalent tech- 28 Itagaki T, Tsutsumi KI, Ito K & Tsutsumi dazole resistance by a genome-wide niques for qualitative and quantitative Y. Taxonomic status of the Japanese tri- analysis of genetically recombinant Fas- copromicroscopic diagnosis of parasites in ploid forms of Fasciola: comparison of ciola hepatica. Parasitology 2013; 140: animals and humans. Nat Protoc 2010; 5: mitochondrial ND1 and COI sequences 1523–1533. 503–515. with F. hepatica and F. gigantica. J Para- 40 Keiser J, Engels D, Buscher G & Utzinger 16 Gonzales Santana B, Dalton JP, Vasquez sitol 1998; 84: 445–448. J. Triclabendazole for the treatment of fasci- Camargo F, Parkinson M & Ndao M. The 29 Terasaki K, Noda Y, Shibahara T & Ita- oliasis and . Expert Opin diagnosis of human fascioliasis by enzyme- gaki T. Morphological comparisons and Investig Drugs 2005; 14: 1513–1526. linked immunosorbent assay (ELISA) using hypotheses on the origin of polyploids in 41 Centres for Disease Control and Prevention recombinant cathepsin L protease. PLoS parthenogenetic Fasciola sp. J Parasitol (CDC). Fascioliasis - Resources for health Negl Trop Dis 2013; 7: e2414. 2000; 86: 724–729. professionals. June 24, 2014; Available at:

© 2016 The Authors. Parasite Immunology Published by John Wiley & Sons Ltd., Parasite Immunology, 38, 558–568 565 K. Cwiklinski et al. Parasite Immunology

http://www.cdc.gov/parasites/fasciola/health_ Fasciola hepatica infection. Vet Parasitol 68 Walsh KP, Brady MT, Finlay CM, Boon L professionals/index.html#tx, 2016. 2011; 178: 264–272. & Mills KH. Infection with a helminth par- 42 Panic G, Duthaler U, Speich B & Keiser J. 55 Pleasance J, Wiedosari E, Raadsma HW, asite attenuates autoimmunity through Repurposing drugs for the treatment and Meeusen E & Piedrafita D. Resistance to TGF-beta-mediated suppression of Th17 control of helminth infections. Int J Para- liver fluke infection in the natural sheep and Th1 responses. J Immunol 2009; 183: sitol Drugs Drug Resist 2014; 4: 185–200. host is correlated with a type-1 cytokine 1577–1586. 43 Winkelhagen AJ, Mank T, de Vries PJ & response. Parasite Immunol 2011; 33: 495– 69 Donnelly S, O’Neill SM, Sekiya M, Mulc- Soetekouw R. Apparent triclabendazole- 505. ahy G & Dalton JP. Thioredoxin peroxidase resistant human Fasciola hepatica infection, 56 Mendes RE, Perez-Ecija RA, Zafra R, secreted by Fasciola hepatica induces the the Netherlands. Emerg Infect Dis 2012; 18: et al. Evaluation of hepatic changes and alternative activation of macrophages. Infect 1028–1029. local and systemic immune responses in Immun 2005; 73: 166–173. 44 Gulhan B, Kanik Yuksek S, Tezer H, et al. goats immunized with recombinant Perox- 70 Donnelly S, Stack CM, O’Neill SM, Sayed Partial hepatectomy for the resistant Fasci- iredoxin (Prx) and challenged with Fasciola AA, Williams DL & Dalton JP. Helminth ola hepatica infection in a child. APSP J hepatica. Vaccine 2010; 28: 2832–2840. 2-Cys peroxiredoxin drives Th2 responses Case Rep 2015; 6: 27. 57 Clery DG & Mulcahy G. Lymphocyte and through a mechanism involving alterna- 45 Cabada MM, Lopez M, Cruz M, Delgado cytokine responses of young cattle during tively activated macrophages. FASEB J JR, Hill V & White AC Jr. Treatment fail- primary infection with Fasciola hepatica. 2008; 22: 4022–4032. ure after multiple courses of triclabendazole Res Vet Sci 1998; 65: 169–171. 71 Flynn RJ, Irwin JA, Olivier M, Sekiya M, among patients with Fascioliasis in Cusco, 58 Bossaert K, Jacquinet E, Saunders J, Farnir Dalton JP & Mulcahy G. Alternative acti- Peru: a case series. PLoS Negl Trop Dis F & Losson B. Cell-mediated immune vation of ruminant macrophages by Fasci- 2016; 10: e0004361. response in calves to single-dose, trickle, ola hepatica. Vet Immunol Immunopathol 46 Ingale SL, Singh P, Raina OK, et al. Inter- and challenge infections with Fasciola hep- 2007; 120:31–40. feron-gamma and interleukin-4 expression atica. Vet Parasitol 2000; 88:17–34. 72 Hacariz O, Sayers G & Mulcahy G. A pre- during Fasciola gigantica primary infection 59 Bossaert K, Farnir F, Leclipteux T, Protz M, liminary study to understand the effect of in crossbred bovine calves as determined by Lonneux JF & Losson B. Humoral immune Fasciola hepatica tegument on naive macro- real -time PCR. Vet Parasitol 2008; 152: response in calves to single-dose, trickle and phages and humoral responses in an ovine 158–161. challenge infections with Fasciola hepatica. model. Vet Immunol Immunopathol 2011; 47 Ingale SL, Singh P, Raina OK, Verma AK, Vet Parasitol 2000; 87: 103–123. 139: 245–249. Channappanavar R & Mehra UR. Inter- 60 Brady MT, O’Neill SM, Dalton JP & Mills 73 Rahko T. The pathology of natural Fasciola leukin-2 and interleukin-10 gene expression KH. Fasciola hepatica suppresses a protec- hepatica infection in cattle. Pathol Vet 1969; in calves experimentally infected with Fasci- tive Th1 response against Bordetella pertus- 6: 244–256. ola gigantica. Livestock Sci 2010; 131: 141– sis. Infect Immun 1999; 67: 5372–5378. 74 van Milligen FJ, Cornelissen JB & Bokhout 143. 61 Everts B, Smits HH, Hokke CH & Yazdan- BA. Location of induction and expression 48 Molina EC. Serum interferon-gamma and bakhsh M. Helminths and dendritic cells: of protective immunity against Fasciola hep- interleukins-6 and -8 during infection with sensing and regulating via pattern recogni- atica at the gut level: a study using an Fasciola gigantica in cattle and buffaloes. J tion receptors, Th2 and Treg responses. Eur ex vivo infection model with ligated gut Vet Sci 2005; 6: 135–139. J Immunol 2010; 40: 1525–1537. segments. J Parasitol 1998; 84: 771–777. 49 Molina EC & Skerratt LF. Cellular and 62 O’Neill SM, Brady MT, Callanan JJ, et al. 75 Ferreras MC, Garcia-Iglesias MJ, Manga- humoral responses in liver of cattle and Fasciola hepatica infection downregulates Gonzalez MY, et al. Histopathological and buffaloes infected with a single dose of Fas- Th1 responses in mice. Parasite Immunol immunohistochemical study of lambs ciola gigantica. Vet Parasitol 2005; 131: 2000; 22: 147–155. experimentally infected with Fasciola hep- 157–163. 63 Hacariz O, Sayers G, Flynn RJ, Lejeune A atica and . J Vet Med B 50 Kumar N, Raina OK, Nagar G, Prakash V & Mulcahy G. IL-10 and TGF-beta1 are Infect Dis Vet Public Health 2000; 47: & Jacob SS. Th1 and Th2 cytokine gene associated with variations in fluke burdens 763–773. expression in primary infection and vacci- following experimental fasciolosis in sheep. 76 Vukman KV, Adams PN & O’Neill SM. nation against Fasciola gigantica in buf- Parasite Immunol 2009; 31: 613–622. Fasciola hepatica tegumental coat antigen faloes by real-time PCR. Parasitol Res 64 Zafra R, Perez-Ecija RA, Buffoni L, et al. suppresses MAPK signalling in dendritic 2013; 112: 3561–3568. Early and late peritoneal and hepatic cells and up-regulates the expression of 51 Flynn RJ & Mulcahy G. Possible role for changes in goats immunized with recombi- SOCS3. Parasite Immunol 2013; 35: 234– Toll-like receptors in interaction of Fasciola nant cathepsin L1 and infected with Fasci- 238. hepatica excretory/secretory products with ola hepatica. J Comp Pathol 2013; 148: 77 Vukman KV, Adams PN, Metz M, Maurer bovine macrophages. Infect Immun 2008; 373–384. M&O’Neill SM. Fasciola hepatica tegu- 76: 678–684. 65 Zafra R, Perez-Ecija RA, Buffoni L, mental coat impairs mast cells’ ability to 52 Alvarez Rojas CA, Ansell BR, Hall RS, et al. Early hepatic and peritoneal drive Th1 immune responses. J Immunol et al. Transcriptional analysis identifies key changes and immune response in goats 2013; 190: 2873–2879. genes involved in , /tissue vaccinated with a recombinant glu- 78 Hashimoto K, Uchikawa R, Tegoshi T, repair and the immune response against tathione transferase sigma class and chal- Takeda K, Yamada M & Arizono N. Fasciola hepatica in sheep liver. Parasit Vec- lenged with Fasciola hepatica. Res Vet Immunity-mediated regulation of fecundity tors 2015; 8: 124. Sci 2013; 94:602–609. in the Heligmosomoides poly- 53 Hacariz O, Sayers G, McCullough M, Gar- 66 Zafra R, Perez J, Buffoni L, et al. Periph- gyrus–the potential role of mast cells. Para- rett M, O’Donovan J & Mulcahy G. The eral blood lymphocyte subsets in Fasciola sitology 2010; 137: 881–887. effect of Quil A adjuvant on the course of hepatica infected and immunised goats. Vet 79 Metcalfe DD, Peavy RD & Gilfillan AM. experimental Fasciola hepatica infection in Immunol Immunopathol 2013; 155: 135–138. Mechanisms of mast cell signaling in ana- sheep. Vaccine 2009; 27:45–50. 67 Dalton JP, Robinson MW, Mulcahy G, phylaxis. J Allergy Clin Immunol 2009; 124: 54 Pleasance J, Raadsma HW, Estuningsih SE, O’Neill SM & Donnelly S. Immunomodu- 639–646; quiz 647-8. Widjajanti S, Meeusen E & Piedrafita D. latory molecules of Fasciola hepatica: candi- 80 Robinson MW, Hutchinson AT, Donnelly S Innate and adaptive resistance of Indone- dates for both vaccine and & Dalton JP. secretory molecules are sian Thin Tail sheep to liver fluke: a com- immunotherapeutic development. Vet Para- causing alarm. Trends Parasitol 2010; 26: parative analysis of Fasciola gigantica and sitol 2013; 195: 272–285. 371–372.

566 © 2016 The Authors. Parasite Immunology Published by John Wiley & Sons Ltd., Parasite Immunology, 38, 558–568 Volume 38, Number 9, September 2016 Animal and human Fasciolosis

81 Aldridge A & O’Neill SM. Fasciola hepat- 96 Jafari-Koshki T, Arsang-Jang S & Raei M. 111 Littman DR & Pamer EG. Role of the ica tegumental antigens induce anergic like Applying spatiotemporal models to study commensal microbiota in normal and T cells via dendritic cells in a mannose risk of smear-positive tuberculosis in Iran, pathogenic host immune responses. Cell receptor dependent manner. Eur J Immunol 2001–2012. Int J Tuberc Lung Dis 2015; 19: Host Microbe 2011; 10: 311–323. 2016; 46: 1180–92. 469–474. 112 Chinen T & Rudensky AY. The effects of 82 Borkow G, Leng Q, Weisman Z, et al. 97 Elias D, Mengistu G, Akuffo H & Britton commensal microbiota on immune cell sub- Chronic immune activation associated with S. Are intestinal helminths risk factors for sets and inflammatory responses. Immunol intestinal helminth infections results in developing active tuberculosis? Trop Med Rev 2012; 245:45–55. impaired signal transduction and anergy. J Int Health 2006; 11: 551–558. 113 Honda K & Littman DR. The microbiome Clin Invest 2000; 106: 1053–1060. 98 Tristao-Sa R, Ribeiro-Rodrigues R, John- in infectious disease and inflammation. 83 Reyes JL & Terrazas LI. The divergent son LT, Pereira FE & Dietze R. Intestinal Annu Rev Immunol 2012; 30: 759–795. roles of alternatively activated macrophages and pulmonary tuberculosis. 114 Hooper LV, Littman DR & Macpherson in helminthic infections. Parasite Immunol Rev Soc Bras Med Trop 2002; 35: 533– AJ. Interactions between the microbiota 2007; 29: 609–619. 535. and the immune system. Science 2012; 336: 84 Allen JE & Maizels RM. Diversity and dia- 99 Wammes LJ, Hamid F, Wiria AE, et al. 1268–1273. logue in immunity to helminths. Nat Rev Regulatory T cells in human geohelminth 115 Molloy MJ, Bouladoux N & Belkaid Y. Immunol 2011; 11: 375–388. infection suppress immune responses to Intestinal microbiota: shaping local and 85 Barron L & Wynn TA. Macrophage activa- BCG and Plasmodium falciparum. Eur J systemic immune responses. Semin Immunol tion governs -induced Immunol 2010; 40: 437–442. 2012; 24:58–66. inflammation and fibrosis. Eur J Immunol 100 Sabin EA, Araujo MI, Carvalho EM & 116 Kamada N, Seo SU, Chen GY & Nunez G. 2011; 41: 2509–2514. Pearce EJ. Impairment of tetanus toxoid- Role of the gut microbiota in immunity 86 Jenkins SJ & Allen JE. Similarity and specific Th1-like immune responses in and inflammatory disease. Nat Rev Immu- diversity in macrophage activation by humans infected with . nol 2013; 13: 321–335. nematodes, trematodes, and cestodes. J J Infect Dis 1996; 173: 269–272. 117 Zeissig S & Blumberg RS. Life at the begin- Biomed Biotechnol 2010; 2010: 262609. 101 Walson JL, Herrin BR & John-Stewart G. ning: perturbation of the microbiota by 87 Mulcahy G, O’Connor F, Clery D, et al. Deworming helminth co-infected individu- antibiotics in early life and its role in health Immune responses of cattle to experimental als for delaying HIV disease progression. and disease. Nat Immunol 2014; 15: 307–310. anti-Fasciola hepatica vaccines. Res Vet Sci Cochrane Database Syst Rev 2009; 3: 118 Belkaid Y & Hand TW. Role of the micro- 1999; 67:27–33. CD006419. biota in immunity and inflammation. Cell 88 Mulcahy G & Dalton JP. Cathepsin L pro- 102 Jackson JA, Friberg IM, Little S & Bradley 2014; 157: 121–141. teinases as vaccines against infection with JE. Review series on helminths, immune 119 Herbst T, Sichelstiel A, Schar C, et al. Dys- Fasciola hepatica (liver fluke) in ruminants. modulation and the hypothesis: regulation of allergic airway inflammation Res Vet Sci 2001; 70:83–86. immunity against helminths and immuno- in the absence of microbial colonization. 89 Salgame P, Yap GS & Gause WC. Effect of logical phenomena in modern human popu- Am J Respir Crit Care Med 2011; 184: helminth-induced immunity on infections lations: coevolutionary legacies? 198–205. with microbial pathogens. Nat Immunol Immunology 2009; 126:18–27. 120 Turnbaugh PJ, Backhed F, Fulton L & 2013; 14: 1118–1126. 103 Fumagalli M, Pozzoli U, Cagliani R, et al. Gordon JI. Diet-induced obesity is linked 90 Aitken MM, Jones PW, Hall GA, Hughes Parasites represent a major selective force to marked but reversible alterations in the DL & Brown GT. Responses of fluke- for interleukin genes and shape the genetic mouse distal gut microbiome. Cell Host infected and fluke-free cattle to experimen- predisposition to autoimmune conditions. J Microbe 2008; 3: 213–223. tal reinfection with Salmonella dublin. Res Exp Med 2009; 206: 1395–1408. 121 Sun J, Furio L, Mecheri R, et al. Pancre- Vet Sci 1981; 31: 120–126. 104 Maizels RM. Parasite immunomodulation atic beta-cells limit autoimmune diabetes 91 Diaz B, Gallegos D, Murillo F, Lenart VL, and polymorphisms of the immune system. via an immunoregulatory antimicrobial Weidman WH & Goldsmith RI. Disease J Biol 2009; 8: 62. peptide expressed under the influence of and disability among the Aymara. In Schull 105 Weinstock JV & Elliott DE. Helminth the gut microbiota. Immunity 2015; 43: WJ, Rothhammer F (eds): The Aymara: infections decrease host susceptibility to 304–317. Strategies in Human Adaptation to a Rigor- immune-mediated diseases. J Immunol 2014; 122 Wu HJ, Ivanov II, Darce J, et al. Gut-resid- ous Environment. Netherlands, Springer, 193: 3239–3247. ing segmented filamentous bacteria drive 1990: 101–131. 106 Elliott DE & Weinstock JV. Helminth-host autoimmune arthritis via T helper 17 cells. 92 Esteban JG, Flores A, Aguirre C, Strauss immunological interactions: prevention and Immunity 2010; 32: 815–827. W, Angles R & Mas-Coma S. Presence of control of immune-mediated diseases. Ann 123 Berer K, Mues M, Koutrolos M, et al. very high prevalence and intensity of infec- N Y Acad Sci 2012; 1247:83–96. Commensal microbiota and myelin tion with Fasciola hepatica among Aymara 107 Mas-Coma S, Valero MA & Bargues MD. autoantigen cooperate to trigger autoim- children from the Northern Bolivian Alti- Climate change effects on trematodiases, with mune demyelination. Nature 2011; 479: plano. Acta Trop 1997; 66:1–14. emphasis on zoonotic fascioliasis and schisto- 538–541. 93 Ashrafi K, Saadat F, O’Neill S, et al. The somiasis. Vet Parasitol 2009; 163:264–280. 124 Mutapi F. The gut microbiome in the hel- Endemicity of Human Fascioliasis in Guilan 108 van Dijk J, Sargison ND, Kenyon F & minth infected host. Trends Parasitol 2015; Province, Northern Iran: the Baseline for Skuce PJ. Climate change and infectious 31: 405–406. Implementation of Control Strategies. Iran J disease: helminthological challenges to 125 Walk ST, Blum AM, Ewing SA, Weinstock Public Health 2015; 44: 501–511. farmed ruminants in temperate regions. JV & Young VB. Alteration of the murine 94 Ashrafi K. The status of human and ani- Animal 2010; 4: 377–392. gut microbiota during infection with the mal fascioliasis in Iran: a narrative review 109 Morgan ER, Charlier J, Hendrickx G, et al. parasitic helminth Heligmosomoides poly- article. Iran J Parasitol 2015; 10: 306–328. Global change and helminth infections in gyrus. Inflamm Bowel Dis 2010; 16: 1841– 95 Vatandoost H, Ashraf H, Lak SH, Mahdi grazing ruminants in Europe: impacts, 1849. RE, Abai MR & Nazari M. Factors Trends and Sustainable Solutions. Agricul- 126 McKenney EA, Williamson L, Yoder AD, involved in the re-emergence of malaria in ture 2013; 3: 484–502. Rawls JF, Bilbo SD & Parker W. Alteration borderline of Iran, Armenia, Azerbaijan 110 Hill DA & Artis D. Intestinal bacteria and of the rat cecal microbiome during colo- and Turkey. Southeast Asian J Trop Med the regulation of immune cell homeostasis. nization with the helminth Hymenolepis Public Health 2003; 34(Suppl 2): 6–14. Annu Rev Immunol 2010; 28: 623–667. diminuta. Gut Microbes 2015; 6: 182–193.

© 2016 The Authors. Parasite Immunology Published by John Wiley & Sons Ltd., Parasite Immunology, 38, 558–568 567 K. Cwiklinski et al. Parasite Immunology

127 Wu S, Li RW, Li W, Beshah E, Dawson with alterations in the faecal microbiota. culture ex host. Mol Cell Proteomics 2007; HD & Urban JF Jr. Worm burden-depen- PLoS One 2013; 8: e76573. 6: 963–972. dent disruption of the porcine colon micro- 142 Dowling DJ, Hamilton CM, Donnelly S, 156 Robinson MW, Menon R, Donnelly SM, biota by Trichuris suis infection. PLoS One et al. Major secretory antigens of the hel- Dalton JP & Ranganathan S. An integrated 2012; 7: e35470. minth Fasciola hepatica activate a suppres- transcriptomics and proteomics analysis of 128 Houlden A, Hayes KS, Bancroft AJ, et al. sive dendritic cell phenotype that attenuates the secretome of the helminth pathogen Chronic trichuris muris infection in C57BL/ Th17 cells but fails to activate Th2 immune Fasciola hepatica: proteins associated with 6 mice causes significant changes in host responses. Infect Immun 2010; 78: 793–801. invasion and infection of the mammalian microbiota and metabolome: effects 143 Isseroff H, Tunis M & Read CP. Changes host. Mol Cell Proteomics 2009; 8: 1891– reversed by pathogen clearance. PLoS One in amino acids of bile in Fasciola hepatica 1907. 2015; 10: e0125945. infections. Comp Biochem Physiol B 1972; 157 Morphew RM, Eccleston N, Wilkinson TJ, 129 Brestoff JR & Artis D. Commensal bacteria 41: 157–163. et al. Proteomics and in silico approaches at the interface of host metabolism and the 144 Sagar NM, Cree IA, Covington JA & to extend understanding of the glutathione immune system. Nat Immunol 2013; 14: Arasaradnam RP. The interplay of the gut transferase superfamily of the tropical liver 676–684. microbiome, bile acids, and volatile organic fluke Fasciola gigantica. J Proteome Res 130 Shapiro H, Thaiss CA, Levy M & Elinav compounds. Gastroenterol Res Pract 2015; 2012; 11: 5876–5889. E. The cross talk between microbiota and 2015: 398585. 158 Hacariz O, Baykal AT, Akgun M, Kavak P, the immune system: metabolites take center 145 Jones ML, Tomaro-Duchesneau C & Pra- Sagiroglu MS & Sayers GP. Generating a stage. Curr Opin Immunol 2014; 30:54–62. kash S. The gut microbiome, probiotics, detailed protein profile of Fasciola hepatica 131 Cummings JH. Fermentation in the human bile acids axis, and human health. Trends during the chronic stage of infection in cat- large intestine: evidence and implications Microbiol 2014; 22: 306–308. tle. Proteomics 2014; 14: 1519–1530. for health. Lancet 1983; 1: 1206–1209. 146 Keitel V, Donner M, Winandy S, Kubitz R & 159 Marcilla A, Trelis M, Cortes A, et al. 132 Cummings JH & Macfarlane GT. The con- Haussinger D. Expression and function of the Extracellular vesicles from parasitic hel- trol and consequences of bacterial fermen- bile acid receptor TGR5 in Kupffer cells. Bio- minths contain specific excretory/secretory tation in the human colon. J Appl Bacteriol chem Biophys Res Commun 2008; 372:78–84. proteins and are internalized in intestinal 1991; 70: 443–459. 147 Vavassori P, Mencarelli A, Renga B, Dis- host cells. PLoS One 2012; 7: e45974. 133 Berndt BE, Zhang M, Owyang SY, et al. trutti E & Fiorucci S. The bile acid receptor 160 Cwiklinski K, de la Torre-Escudero E, Tre- Butyrate increases IL-23 production by FXR is a modulator of intestinal innate lis M, et al. The extracellular vesicles of the stimulated dendritic cells. Am J Physiol immunity. J Immunol 2009; 183: 6251–6261. helminth pathogen, Fasciola hepatica: bio- Gastrointest Liver Physiol 2012; 303: 148 Inagaki T, Moschetta A, Lee YK, et al. genesis pathways and cargo molecules G1384–G1392. Regulation of antibacterial defense in the involved in parasite pathogenesis. Mol Cell 134 Cox MA, Jackson J, Stanton M, et al. small intestine by the nuclear bile acid Proteomics 2015; 14: 3258–3273. Short-chain fatty acids act as antiinflamma- receptor. Proc Natl Acad Sci USA 2006; 161 Garcia-Campos A, Ravida A, Nguyen LD, tory mediators by regulating prostaglandin 103: 3920–3925. et al. Tegument glycoproteins and cathep- E(2) and cytokines. World J Gastroenterol 149 Lopez P, Gonzalez P, Tunon MJ & Gonza- sins of Newly Excysted Juvenile Fasciola 2009; 15: 5549–5557. lez-Gallego J. The effects of experimental hepatica carry mannosidic and pauciman- 135 Liu L, Li L, Min J, et al. Butyrate inter- fasciolosis on bilirubin metabolism in the nosidic N-glycans. PLoS Negl Trop Dis feres with the differentiation and function rat. Exp Parasitol 1994; 78: 386–393. 2016; 10: e0004688. of human monocyte-derived dendritic cells. 150 Huang Y, Chen W, Wang X, et al. The car- 162 Collins JJ 3rd, Wang B, Lambrus BG, Tharp Cell Immunol 2012; 277:66–73. cinogenic liver fluke, : ME, Iyer H & Newmark PA. Adult somatic 136 Huang N, Katz JP, Martin DR & Wu GD. new assembly, reannotation and analysis of stem cells in the Schistosoma Inhibition of IL-8 gene expression in Caco- the genome and characterization of tissue mansoni. Nature 2013; 494: 476–479. 2 cells by compounds which induce histone transcriptomes. PLoS One 2013; 8: e54732. 163 Wang B, Collins JJ 3rd & Newmark PA. hyperacetylation. Cytokine 1997; 9:27–36. 151 Gobert GN, You H & McManus DP. Gain- Functional genomic characterization of 137 Smith PM, Howitt MR, Panikov N, et al. ing biological perspectives from schisto- neoblast-like stem cells in larval Schisto- The microbial metabolites, short-chain fatty some genomes. Mol Biochem Parasitol soma mansoni. Elife 2013; 2: e00768. acids, regulate colonic Treg cell homeosta- 2014; 196:21–28. 164 Young ND, Jex AR, Cantacessi C, et al. A sis. Science 2013; 341: 569–573. 152 Young ND, Nagarajan N, Lin SJ, et al. portrait of the transcriptome of the 138 Zaiss MM, Rapin A, Lebon L, et al. The The Opisthorchis viverrini genome provides neglected trematode, Fasciola gigantica–bio- intestinal microbiota contributes to the abil- insights into life in the bile duct. Nat Com- logical and biotechnological implications. ity of helminths to modulate allergic inflam- mun 2014; 5: 4378. PLoS Negl Trop Dis 2011; 5: e1004. mation. Immunity 2015; 43: 998–1010. 153 Forrester SJ & Hall N. The revolution of 165 Martin J, Rosa BA, Ozersky P, et al. Hel- 139 Kay GL, Millard A, Sergeant MJ, et al. whole genome sequencing to study parasites. minth.net: expansions to Nematode.net and Differences in the faecal microbiome in Mol Biochem Parasitol 2014; 195:77–81. an introduction to Trematode.net. Nucleic infected children 154 Jefferies JR, Campbell AM, van Rossum Acids Res 2015; 43(Database issue): D698– vs. uninfected children. PLoS Negl Trop AJ, Barrett J & Brophy PM. Proteomic D706. Dis 2015; 9: e0003861. analysis of Fasciola hepatica excretory- 166 Toet H, Piedrafita DM & Spithill TW. 140 Lee SC, Tang MS, Lim YA, et al. Helminth secretory products. Proteomics 2001; 1: Liver fluke vaccines in ruminants: strate- colonization is associated with increased 1128–1132. gies, progress and future opportunities. Int diversity of the gut microbiota. PLoS Negl 155 Morphew RM, Wright HA, LaCourse EJ, J Parasitol 2014; 44: 915–27. Trop Dis 2014; 8: e2880. Woods DJ & Brophy PM. Comparative 167 Molina-Hernandez V, Mulcahy G, Perez J, 141 Cooper P, Walker AW, Reyes J, et al. proteomics of excretory-secretory proteins et al. Fasciola hepatica vaccine: we may not Patent human infections with the whip- released by the liver fluke Fasciola hepatica be there yet but we’re on the right road. worm, , are not associated in sheep host bile and during in vitro Vet Parasitol 2015; 208: 101–111.

568 © 2016 The Authors. Parasite Immunology Published by John Wiley & Sons Ltd., Parasite Immunology, 38, 558–568