Hostname: page-component-848d4c4894-wzw2p Total loading time: 0 Render date: 2024-06-07T06:11:49.144Z Has data issue: false hasContentIssue false

Lessons from studying roundworm and whipworm in the mouse: common themes and unique features

Published online by Cambridge University Press:  11 August 2021

C. V. Holland*
Affiliation:
Department of Zoology, School of Natural Sciences, Trinity College, Dublin 2, Ireland
K. J. Else*
Affiliation:
Faculty of Biology, Medicine and Health, Lydia Becker Institute of Immunology and Inflammation, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, ManchesterM13 9PT, UK
*
Authors for correspondence: C. V. Holland, E-mail: cholland@tcd.ie; K. J. Else, E-mail: Kathryn.Else@manchester.ac.uk
Authors for correspondence: C. V. Holland, E-mail: cholland@tcd.ie; K. J. Else, E-mail: Kathryn.Else@manchester.ac.uk

Abstract

Ascaris lumbricoides, the roundworm, and Trichuris trichiura, the whipworm, are human intestinal nematode parasites; both are soil-transmitted helminths, are often placed together in an epidemiological context and both remain neglected despite high prevalence. Our understanding of parasitic disease continues to be enhanced through animal models. Despite the similarities between whipworm and roundworm, there are key differences between the two species and these have influenced the application of their respective animal models. In the case of T. trichiura, the fact that a murine equivalent, T. muris completes its life cycle in a mouse model has greatly enhanced our knowledge of whipworm biology, pathogenicity and immunology. In contrast, A. lumbricoides and its porcine equivalent, Ascaris suum, lack a rodent model in which the life cycle is completed. However, evidence continues to accumulate demonstrating that mice represent useful models of early Ascaris infection, a key stage of the life cycle. The use of mouse models for both Ascaris and Trichuris has a long history with early pioneers discovering fundamental aspects of each parasite's biology. Novel technologies and perspectives, as outlined in this special issue, demonstrate how through the prism of mouse models, we can continue to explore the similarities and differences between roundworms and whipworms.

Type
Editorial
Copyright
Copyright © The Author(s), 2021. Published by Cambridge University Press

Introduction

Why study parasite infections in mice? The house mouse, Mus musculus, is a powerful model organism given its similar anatomy, physiology and immunology to humans, its short breeding cycle and the availability of a whole ‘toolbox’ of reagents, transgenic mice and advanced methodologies. In the context of parasitic infection, one of the key research imperatives is a desire to alleviate the ill-health caused by infection in humans and their domestic animals, and research into the soil-transmitted helminth parasites is no exception to this. A variety of gut helminth mouse models exist, including the rodent hookworms Heligomosomoides polygyrus and Nippostrongylus brasiliensis, the whipworm Trichuris muris and the roundworm Ascaris spp. This special edition of Parasitology focuses on the latter two parasites with articles focused upon immunology, immunoregulation, co-morbidities, the microbiome and vaccine development.

Table 1. Mouse models of Trichuris and Ascaris infections: a selection of the early pioneers

Despite whipworms and roundworms sharing certain commonalities, for example, an association with humans for several thousand years and an ability to trigger a similar quality of immune response (reviewed in Else et al., Reference Else, Keiser, Holland, Grencis, Sattelle, Fujiwara, Bueno, Asaolu, Sowemimo and Cooper2020), these two nematode species also have many differences. Ascaris worms are large; up to 35 cm in length whilst whipworms are small (up to 5 cm in length); whipworms live in the large intestine whilst Ascaris inhabits the small intestine. More than 70 spp. of Trichuris are described, including T. muris in the mouse, whilst in the genus Ascaris only two species are known, A. suum in pigs and the human parasite A. lumbricoides. Notably, whipworms are entirely enteric in their life style whilst Ascaris parasites have a migratory larval phase that contributes to pathology in the liver and lungs in addition to the intestine (Holland, Reference Holland2021). In the context of mouse models, a key difference between the two parasites is that the mouse is a natural host of whipworm infection but not of Ascaris infections, and this difference plays out in some of the ways that mouse models have been used to understand whipworm and roundworm infections, as described within this special collection. Collectively, the articles in this special issue explore the application of these two important nematode parasites, in both basic and applied research, and describe some of the recent advances made through the use of the mouse model.

Historical aspects

Trichuris muris in the mouse has been used as a model system by parasitologists for well over half a century with Shikhobalova reporting studies in ‘white mice' as long ago as 1937 (Shikhobalova, Reference Shikhobalova1937). Detailed studies of the whipworm life cycle in the mouse were published in 1954 (Fahmy, Reference Fahmy1954) and at that time there was a growing desire to understand the host–parasite interaction better. Thus, a number of studies reporting strain variation in the ability to carry a chronic infection were published, including Keeling (Reference Keeling1961) and Worley et al. (Reference Worley, Meisenhelder, Sheffield and Thompson1962), leading to a debate as to whether the inability to carry an infection through to the adult stage represented an incompatibility between the host environment and the parasite, or immune-mediated resistance (or both). Important work by Campbell in 1963 discussed the concept of immune-mediated worm expulsion which was then built upon by numerous elegant studies by Wakelin in the 1960s and 70s (see Wakelin, Reference Wakelin1967, Reference Wakelin1970a, Reference Wakelin1970b). It is interesting, however, to reflect on the early debate around a host environment incompatibility vs immunity, given our new knowledge of the important contribution of the host microbiome to the success of the parasite, a topic discussed by Lawson et al. (Reference Lawson, Roberts and Grencis2021) in this volume, and the fact that Pike had noted changes in the bacterial flora in the caeca of Trichuris-infected mice as long ago as 1976 (Pike, Reference Pike1976) (Table 1).

In contrast to Trichuris, Ascaris does not have a species that infects a natural rodent host where the life cycle is completed. Animal models in which Ascaris does not complete its life cycle but mimic the all important hepato-tracheal migration are described as abnormal hosts. Larval stages of Ascaris do not return successfully to the small intestine to mature into adult worms (Holland, Reference Holland2021). This has undoubtedly hindered research into what is regarded by some as the most neglected of the neglected tropical diseases (Hotez, Reference Hotez and Holland2013). However, it is clear that this phase of infection is likely to be crucial in the manifestation of susceptibility and resistance and successful adult worm establishment (Nogueira et al., Reference Nogueira, Gazzinelli-Guimarães, Barbosa, Resende, Silva, de Oliveira, Amorim, Oliveira, Mattos, Kraemer, Caliari, Gaze, Bueno, Russo and Fujiwara2016) and this is one of the reasons why mice serve as a useful model for the assessment of vaccine candidates (see Gazzinelli-Guimaraes et al., Reference Gazzinelli-Guimaraes, Gazzinelli-Guimaraes and Weatherhead2021).

As early as 1916, investigators used A. suum-infected mice to make novel and important observations concerning the biology of the ascarid (Stewart, Reference Stewart1916; Holland, Reference Holland2021) (Table 1). Since then, mouse models of early Ascaris infection have been used for such diverse investigations as the intestinal hepatic migratory pattern (e.g. Ransom and Foster, Reference Ransom and Foster1920; Slotved et al., Reference Slotved, Eriksen, Murrell and Nansen1998; Dold et al., Reference Dold, Cassidy, Stafford, Behnke and Holland2010). Of particular note are the findings of Slotved et al. (Reference Slotved, Eriksen, Murrell and Nansen1998) who demonstrated, in important comparative work, that the migratory pattern of A. suum is similar in murine and porcine hosts; the hepatic-pulmonary migratory pattern (e.g. Sprent, Reference Sprent1952; Douvres and Tromba, Reference Douvres and Tromba1971; Song et al., Reference Song, Kim, Min and Lee1985; Lewis et al., Reference Lewis, Behnke, Stafford and Holland2006); hepatic pathology (e.g. Bindseil, Reference Bindseil1969; Dold et al., Reference Dold, Cassidy, Stafford, Behnke and Holland2010; Deslyper et al., Reference Deslyper, Colgan, Holland and Carolan2016, Reference Deslyper, Holland, Colgan and Carolan2019); immunological responses (Mitchell et al., Reference Mitchell, Hogarth-Scott, Edwards, Lewers, Cousins and Moore1976; Kennedy et al., Reference Kennedy, Gordon, Tomlinson and Qureshi1987; Gazzinelli-Guimaraes et al., Reference Gazzinelli-Guimaraes, Gazzinelli-Guimaraes, Silva, Mati, de Carvalho Dhom-Lemos, Barbosa, Passos, Gaze, Carneiro, Bartholomeu, Bueno and Fujiwara2013; Nogueira et al., Reference Nogueira, Gazzinelli-Guimarães, Barbosa, Resende, Silva, de Oliveira, Amorim, Oliveira, Mattos, Kraemer, Caliari, Gaze, Bueno, Russo and Fujiwara2016) and host–parasite genetics (e.g. Mitchell et al., Reference Mitchell, Hogarth-Scott, Edwards, Lewers, Cousins and Moore1976; Nejsum et al., Reference Nejsum, Roepstorff, Anderson, Jorgensen, Fredholm and Thamsborg2008; Dold et al., Reference Dold, Pemberton, Stafford, Holland and Behnke2011; Peng et al., Reference Peng, Yuan, Peng, Dai, Yuan, Hu and Hu2012). Furthermore, Gazzinelli-Guimaraes et al. (Reference Gazzinelli-Guimaraes, Gazzinelli-Guimaraes and Weatherhead2021) describe the mouse model as the primary in vivo animal system for the evaluation of vaccine candidates for A. lumbricoides and provide a detailed historical perspective on the studies performed from 1957 to 2021.

Wild immunology

Whilst the early drivers to study T. muris in the mouse were largely driven by curiosity, the majority of the articles in this collection focus on the use of mouse models of infection to further our understanding of human disease, and the article by Mair et al. (Reference Mair, Else and Forman2021) is no exception covering the most recent immunological knowledge gained from studying T. muris in the laboratory mouse. However, in addition, Mair et al. move on to extend the beneficiaries of whipworm research beyond the medical community, highlighting contributions made by mouse whipworm research to the field of host–parasite co-evolutionary relationships, ecology and parasite genetic diversity, enabled by the fact that, unlike Ascaris spp., T. muris is a natural parasite of wild mice. Of course the study of parasites in wild populations is not new and one early pioneer in the context of whipworm research was Behnke who assessed the epidemiology of T. muris in its natural host, the wild house mouse (Behnke and Wakelin, Reference Behnke and Wakelin1973). The new and growing field of Eco-Immunology or ‘Wild Immunology’ is also enabled by the fact that, as mentioned above, T. muris naturally infects mice. Eco-immunologists aim to study immunity to infection in real-world settings rather than the highly controlled laboratory environments where most studies on the immune response to infection are conducted. A number of elegant mouse model systems have recently been developed which move from fully wild settings, to the semi-wild enclosure-based systems pioneered by Graham (reviewed by Graham, Reference Graham2021) and a system where inbred strains of mice carry a ‘wild’ microbiome (Rosshart et al., Reference Rosshart, Herz, Vassallo, Hunter, Wall, Badger, McCulloch, Anastasakis, Sarshad, Leonardi, Collins, Blatter, Han, Tamoutounour, Potapova, Foster St Claire, Yuan, Sen, Dreier, Hild, Hafner, Wang, Iliev, Belkaid, Trinchieri and Rehermann2019). Gut-dwelling helminth parasites live in intimate association with their host and with a large diverse microbial community with which they share their niche. The intricate relationship between the host, its parasites and the gut microbiota is extremely well illustrated by studies using T. muris infection of mice, and these complex relationships are described by Lawson et al. (Reference Lawson, Roberts and Grencis2021). Indeed research in this area has illuminated a dependency of the parasite on the host microbiome for maintenance of its life cycle, with T. muris parasites evolving to respond to microbial cues within the host gut as well as harbouring their own unique microbiome.

Vaccine development

Given the persistently high prevalence of A. lumbricoides and Trichuris trichiura globally, research focussing on vaccine development is a priority. A historical perspective by Gazzinelli-Guimaraes et al. (Reference Gazzinelli-Guimaraes, Gazzinelli-Guimaraes and Weatherhead2021) highlights the challenges associated with the development of a vaccine against a complex helminth infection like Ascaris that produces resistant eggs into the environment, promotes rapid re-infection and has the potential to induce anthelmintic resistance. This is despite the clear imperative to identify immunogenic Ascaris antigens that can enhance larval killing and/or larval expulsion, in order to prevent maturation and the successful establishment of adult worms that induce both chronic and acute ascariasis. Promising recombinant Ascaris antigens such as As14, As16 and As37 (Tsuji et al., Reference Tsuji, Suzuki, Kasuga-Aoki, Matsumoto, Arakawa, Ishiwata and Isobe2001, Reference Tsuji, Kasuga-Aoki, Isobe, Arakawa and Matsumoto2002) have been identified and a recent study by de Castro et al. (Reference De Castro, De Almeida, Cardoso, Oliveira, Nogueira, Reis-Cunha, Magalhaes, Zhan, Bottazzi, Hotez, Bueno, Bartholomeu and Fujiwara2021) outlines the use of a more intricate vaccine target, an adjuvanted chimeric protein derived from the three recombinants with an efficacy of 73.54% in BALB/c mice. Our continued paucity of knowledge of the totality of the Ascaris immune responses and the possibility that other undiscovered proteins may exist and act as better vaccine targets remains an ongoing challenge.

Research advances in whipworm antigen discovery are reviewed in Hayon et al. (Reference Hayon, Weatherhead, Hotez, Bottazzi and Zhan2021) which ends with a view of the global health policy needs if we are to develop and implement vaccine delivery in the field. Whilst a case can be made for anti-whipworm vaccine research based on the impact the parasite itself has on human health, whipworm infections also have substantial, widespread systemic effects despite being localized within the large intestine.

Co-morbidities, immunomodulation and co-infection

Hayes and Grencis (Reference Hayes and Grencis2021) highlight these body-wide influences of Trichuris infection which, in addition to affecting the progression of bowel inflammation, can also worsen stroke outcome, suppress lung inflammation and inhibit anti-tumour immunity. Given the far-reaching effects whipworm has on disease progression at sites distal from the site of infection, unsurprisingly Trichuris parasites are a source of immunoregulatory molecules, in keeping with many other helminth parasites; our current understanding of these molecules is reviewed by Bancroft and Grencis (Reference Bancroft and Grencis2021) including the quantitatively dominant single novel protein T. muris p43.

Mouse models have also provided an opportunity to understand the role of Ascaris infection in the exacerbation of other conditions such as pulmonary fibrosis and pulmonary allergic inflammation as discussed by Magalhães et al. (Reference Magalhães, Nogueira, Gazzinelli-Guimaraes, Oliveira, Kraemer, Gazzinelli-Guimaraes, Vieira-Santos, Fujiwara and Bueno2021). Evidence from a mouse model of both larval ascariasis and lung fibrosis revealed an exacerbation of lung damage and an associated diminishment of pulmonary physiological parameters (Oliveira et al., Reference Oliveira, da Paixão Matias, Kraemer, Gazzinelli-Guimarães, Santos, Amorim, Nogueira, Freitas, Caliari, Bartholomeu, Bueno, Russo and Fujiwara2019). Furthermore, allergic airway disease can be observed in Ascaris-infected mice experiencing tissue damage in the lungs (Weatherhead et al., Reference Weatherhead, Porter, Coffey, Haydel, Versteeg, Zhan, Gazzinelli Guimarães, Fujiwara, Jaramillo, Bottazzi, Hotez, Corry and Beaumier2018). As with whipworm infection, an infection with roundworm can exacerbate lung disease. However, the effects of Ascaris on diseases of the lung do not necessarily evidence a systemic reach, unlike whipworm, given that Ascaris migrates through the lung whilst whipworm is entirely enteric. The role of Ascaris infection in liver inflammation lags behind that of the lungs as outlined by Holland (Reference Holland2021). In contrast to other animal models of ascariasis, mice are the only model for which the basis of resistance and susceptibility has been defined (Holland et al., Reference Holland, Behnke, Dold and Holland2013). Furthermore, two inbred strains of mice with highly consistent and diverging larval burdens in their lungs represent the extremes of the host phenotype displayed in the aggregated distribution of Ascaris adult worm burdens in humans (Holland et al., Reference Holland, Asaolu, Crompton, Stoddart, MacDonald and Torimiro1989; Holland, Reference Holland2009). The establishment of this model provided an opportunity to explore the mechanistic basis that confers resistance and predisposition to light and heavy Ascaris infection with a particular emphasis on the liver. In resistant mice, the most pronounced inflammatory response was observed on day 4 post-infection, a day that coincides with the migration of larvae from the liver to the lungs whereas in susceptible mice occurred later on day 6 post-infection when the majority of the larvae are known to have successfully migrated to the lungs (Dold et al., Reference Dold, Cassidy, Stafford, Behnke and Holland2010). These observations led to several proteomic analyses of hepatic tissues in this mouse model that demonstrated intrinsic differences between the two strains, suggesting that resistance might be associated with the oxidative phosphorylation pathway and reactive oxygen species production (Deslyper et al., Reference Deslyper, Colgan, Holland and Carolan2016) and differential expression of components of the complement system (Deslyper et al., Reference Deslyper, Holland, Colgan and Carolan2019). There is obviously a clear need for further investigation of the role of Ascaris infection in tissue damage to key organs such as the liver and the lungs, a pathology which sets Ascaris apart from any of the other soil-transmitted helminths.

The role of helminths including Ascaris in co-infection and the possible perturbation of other infections particularly microparasites has received considerable attention and conflicting observations (Kirwan et al., Reference Kirwan, Jackson, Asaolu, Molloy, Abiona, Bruce, Ranford-Cartwright, O'Neill and Holland2010; Vaumourin et al., Reference Vaumourin, Vourc'h, Gasqui and Vayssier-Taussat2015). A recent paper by Vieira-Santos et al. (Reference Vieira-Santos, Leal-Silva, de Lima Silva Padrao, Ruas, Nogueira, Kraemer, Oliveira, Caliari, Russo, Fujiwara and Bueno2021) explored concomitant infections of Plasmodium berghei and A. suum in a mouse model and found that co-infection exacerbated reduced respiratory function. As with Trichuris, the immunomodulatory properties of Ascaris parasites have received attention given their potential development as therapeutic tools. Caraballo et al. (Reference Caraballo, Zakzuk and Acevedo2021) provide a review of the cystatins as cysteine protein inhibitors with a particular emphasis on the A1-CPI nematode type 2 cystatin in A. lumbricoides. The production of a recombinant form of A1-CPI has been found to be safe in mice and to manifest intestinal anti-inflammatory properties (Coronado et al., Reference Coronado, Barrios, Zakzuk, Regino, Ahumada, Franco, Ocampo and Caraballo2017) and anti-inflammatory properties in the context of an allergic airway inflammatory mouse model (Coronado et al., Reference Coronado, Zakzuk, Regino, Ahumada, Benedetti, Angelina, Palomares and Caraballo2019). The authors conclude that A1-CPI may prove useful in the immunotherapy of asthma (Caraballo et al., Reference Caraballo, Zakzuk and Acevedo2021).

Conclusion

To conclude, as outlined above, the use of mouse models to understand the complex interplay between two important soil-transmitted helminths – A. lumbricoides and T. trichiura and their human hosts – has a long history. However, novel technologies and perspectives, as illuminated in this volume, demonstrate the continued relevance of such animal model systems and how they can help us to fill some of the gaps in our knowledge. We hope that this special edition will stimulate and enhance scientific interest in two parasites that remain among the neglected tropical diseases.

References

Bancroft, AJ and Grencis, RK (2021) Immunoregulatory molecules secreted by Trichuris muris. Parasitology, 17. doi: 10.1017/S0031182021000846Google ScholarPubMed
Behnke, JM and Wakelin, D (1973) The survival of Trichuris muris in wild populations of its natural hosts. Parasitology 67, 157164.10.1017/S0031182000046382CrossRefGoogle ScholarPubMed
Bindseil, E (1969) Immunity to Ascaris suum 2. Investigations of the fate of larvae in immune and non-immune mice. Acta Pathologica Microbiologica Scandinavia 77, 223234.10.1111/j.1699-0463.1969.tb04227.xCrossRefGoogle ScholarPubMed
Campbell, WC (1963) Spontaneous cure in Trichuris muris infections in albino mice and its suppression by cortisone. The Journal of Parasitology 49, 628632.10.2307/3275772CrossRefGoogle ScholarPubMed
Caraballo, L, Zakzuk, J and Acevedo, N (2021) Helminth-derived cystatins: the immunomodulatory properties of an Ascaris lumbricoides cystatin. Parasitology, 113. doi: 10.1017/S0031182021000214Google ScholarPubMed
Coronado, S, Barrios, L, Zakzuk, J, Regino, R, Ahumada, V, Franco, L, Ocampo, Y and Caraballo, L (2017) A recombinant cystatin from Ascaris lumbricoides attenuates inflammation of DSS-induced colitis. Parasite Immunology 39, 4.10.1111/pim.12425CrossRefGoogle ScholarPubMed
Coronado, S, Zakzuk, J, Regino, R, Ahumada, V, Benedetti, I, Angelina, A, Palomares, O and Caraballo, L (2019) Ascaris lumbricoides cystatin prevents development of allergic airway inflammation in a mouse model. Frontiers in Immunology 10, 2280.10.3389/fimmu.2019.02280CrossRefGoogle ScholarPubMed
De Castro, JC, De Almeida, LV, Cardoso, MS, Oliveira, SFM, Nogueira, DS, Reis-Cunha, JL, Magalhaes, LMD, Zhan, B, Bottazzi, ME, Hotez, PJ, Bueno, LL, Bartholomeu, DC and Fujiwara, RT (2021) Vaccination with chimeric protein induces protection in murine model against ascariasis. Vaccine 39(2), 394401.10.1016/j.vaccine.2020.11.046CrossRefGoogle ScholarPubMed
Deslyper, G, Colgan, TC, Holland, CV and Carolan, JC (2016) A proteomic investigation of hepatic resistance to Ascaris in a murine model. PLoS Neglected Tropical Diseases 10, e0004837.10.1371/journal.pntd.0004837CrossRefGoogle ScholarPubMed
Deslyper, G, Holland, CV, Colgan, TC and Carolan, JC (2019) The liver proteome in a mouse model for Ascaris suum resistance and susceptibility: evidence for an altered innate immune response. Parasites and Vectors 12, 402.10.1186/s13071-019-3655-9CrossRefGoogle Scholar
Dold, C, Cassidy, JP, Stafford, P, Behnke, JM and Holland, CV (2010) Genetic influence on the kinetics and associated pathology of the early stage (intestinal-hepatic) migration of Ascaris suum in mice. Parasitology 137, 173185.10.1017/S0031182009990850CrossRefGoogle ScholarPubMed
Dold, C, Pemberton, A, Stafford, P, Holland, CV and Behnke, JMB (2011) The role of intelectin-2 in resistance to Ascaris suum lung larval burdens in susceptible and resistant mouse strains. Parasitology 138, 660669.10.1017/S0031182011000084CrossRefGoogle ScholarPubMed
Douvres, FW and Tromba, FG (1971) Comparative development of Ascaris suum in rabbits, guinea pigs, mice and swine in 11 days. Proceedings of the Helminthological Society of Washington 38, 246252.Google Scholar
Else, KJ, Keiser, J, Holland, CV, Grencis, RK, Sattelle, DB, Fujiwara, RT, Bueno, LL, Asaolu, SO, Sowemimo, OA and Cooper, PJ (2020) Whipworm and roundworm infections. Nature Reviews Disease Primers 6, 44.10.1038/s41572-020-0171-3CrossRefGoogle ScholarPubMed
Fahmy, MA (1954) An investigation on the life cycle of Trichuris muris. Parasitology 44, 5057.10.1017/S003118200001876XCrossRefGoogle ScholarPubMed
Gazzinelli-Guimaraes, PH, Gazzinelli-Guimaraes, AC, Silva, FN, Mati, VLT, de Carvalho Dhom-Lemos, L, Barbosa, FS, Passos, LSA, Gaze, S, Carneiro, CM, Bartholomeu, DC, Bueno, LL and Fujiwara, RT (2013) Parasitological and immunological aspects of early Ascaris spp. infection in mice. International Journal for Parasitology 43, 697706.10.1016/j.ijpara.2013.02.009CrossRefGoogle ScholarPubMed
Gazzinelli-Guimaraes, AC, Gazzinelli-Guimaraes, PH and Weatherhead, J (2021) A historical and systematic overview of Ascaris vaccine development. Parasitology, 142. doi: 10.1017/S0031182021001347Google Scholar
Graham, AL (2021) Naturalizing mouse models for immunology. Nature Immunology 22, 111117.10.1038/s41590-020-00857-2CrossRefGoogle ScholarPubMed
Hayes, KS and Grencis, RK (2021) Trichuris muris and comorbidities – within a mouse model context. Parasitology, 19. doi: 10.1017/S0031182021000883Google ScholarPubMed
Hayon, J, Weatherhead, J, Hotez, PJ, Bottazzi, ME and Zhan, B (2021) Advances in vaccine development for human trichuriasis. Parasitology, 112. doi: 10.1017/S0031182021000500Google ScholarPubMed
Holland, CV (2009) Predisposition to ascariasis: patterns, mechanisms and implications. Parasitology 136, 15371547.10.1017/S0031182009005952CrossRefGoogle ScholarPubMed
Holland, CV (2021) The long and winding road of Ascaris larval migration: the role of mouse models. Parasitology, 19. doi: 10.1017/S0031182021000366Google Scholar
Holland, CV, Asaolu, SO, Crompton, DWT, Stoddart, R, MacDonald, R and Torimiro, SEA (1989) The epidemiology of Ascaris lumbricoides and other soil-transmitted helminths in primary school children from Ile-Ife. Nigeria. Parasitology 99, 275285.10.1017/S003118200005873XCrossRefGoogle ScholarPubMed
Holland, CV, Behnke, JM and Dold, C (2013) Larval ascariasis: impact, significance, and model organisms. In Holland, CV (ed.), Ascaris the Neglected Parasite. London, UK/Waltham, MA/San Diego, CA: Academic Press is an imprint of Elsevier, pp. 107125.10.1016/B978-0-12-396978-1.00005-7CrossRefGoogle Scholar
Hotez, P (2013) Foreword. In Holland, CV (ed.), Ascaris the Neglected Parasite. London, UK/Waltham, MA/San Diego, CA: Academic Press is an imprint of Elsevier, pp. xiiixxiv.10.1016/B978-0-12-396978-1.06001-8CrossRefGoogle Scholar
Keeling, JED (1961) Experimental trichuriasis. I. Antagonism between Trichuris muris and Aspiculuris tetraptera in the albino mouse. The Journal of Parasitology 47, 641646.10.2307/3275076CrossRefGoogle ScholarPubMed
Kennedy, MW, Gordon, AM, Tomlinson, LA and Qureshi, F (1987) Genetic (major histocompatibility complex) control of the antibody repertoire to the secreted antigens of Ascaris. Parasite Immunology 9, 269273.10.1111/j.1365-3024.1987.tb00506.xCrossRefGoogle Scholar
Kirwan, P, Jackson, AL, Asaolu, SO, Molloy, SF, Abiona, TC, Bruce, MC, Ranford-Cartwright, L, O'Neill, SM and Holland, CV (2010) Impact of repeated four-monthly anthelmintic treatment on Plasmodium infection in preschool children: a double-blind placebo-controlled randomized trial. BMC Infectious Diseases 10, 277.10.1186/1471-2334-10-277CrossRefGoogle ScholarPubMed
Lawson, MAE, Roberts, IS and Grencis, RK (2021) The interplay between Trichuris and the microbiota. Parasitology, 18. doi: 10.1017/S0031182021000834Google ScholarPubMed
Lewis, R, Behnke, JM, Stafford, P and Holland, CV (2006) The development of a mouse model to explore resistance and susceptibility to early Ascaris suum infection. Parasitology 132, 289300.10.1017/S0031182005008978CrossRefGoogle ScholarPubMed
Magalhães, L, Nogueira, D, Gazzinelli-Guimaraes, P, Oliveira, F, Kraemer, L, Gazzinelli-Guimaraes, AC, Vieira-Santos, F, Fujiwara, R and Bueno, L (2021) Immunological underpinnings of Ascaris infection, reinfection and co-infection and their associated comorbidities. Parasitology, 110. doi: 10.1017/S0031182021000627Google Scholar
Mair, I, Else, KJ and Forman, R (2021) Trichuris muris as a tool for holistic discovery research: from translational research to environmental bio-tagging. Parasitology, 113. doi: 10.1017/S003118202100069XGoogle ScholarPubMed
Mitchell, GE, Hogarth-Scott, RS, Edwards, RD, Lewers, HM, Cousins, G and Moore, T (1976) Studies on immune response to parasite antigens in mice. 1. Ascaris suum larvae numbers and antiphosphorylcholine responses in infected mice of various strains and in hypothymic nu/nu mice. International Archives of Allergy and Applied Immunity 52, 6478.10.1159/000231669CrossRefGoogle Scholar
Nejsum, P, Roepstorff, A, Anderson, TJC, Jorgensen, C, Fredholm, M and Thamsborg, SM (2008) The dynamics of genetically marked Ascaris suum infections in pigs. Parasitology 136, 193201.10.1017/S0031182008005349CrossRefGoogle ScholarPubMed
Nogueira, DS, Gazzinelli-Guimarães, PH, Barbosa, FS, Resende, NM, Silva, CC, de Oliveira, LM, Amorim, CCO, Oliveira, FMC, Mattos, MS, Kraemer, LR, Caliari, MV, Gaze, S, Bueno, LL, Russo, RC and Fujiwara, RT (2016) Multiple exposures to Ascaris suum induce tissue injury and mixed Th2/Th17 immune response in mice. PLoS Neglected Tropical Diseases 10, e0004382.10.1371/journal.pntd.0004382CrossRefGoogle ScholarPubMed
Oliveira, FMS, da Paixão Matias, PH, Kraemer, L, Gazzinelli-Guimarães, AC, Santos, FV, Amorim, CCO, Nogueira, DS, Freitas, CS, Caliari, MV, Bartholomeu, DC, Bueno, LL, Russo, RC and Fujiwara, RT (2019) Comorbidity associated to Ascaris suum infection during pulmonary fibrosis exacerbates chronic lung and liver inflammation and dysfunction but not affect the parasite cycle in mice. PLoS Neglected Tropical Diseases 13, e0007896.10.1371/journal.pntd.0007896CrossRefGoogle Scholar
Peng, W, Yuan, K, Peng, G, Dai, Z, Yuan, F, Hu, Y and Hu, N (2012) Ascaris: development of selected genotypes in mice. Experimental Parasitology 131, 6974.10.1016/j.exppara.2012.03.006CrossRefGoogle ScholarPubMed
Pike, EH (1976) Changes in the bacterial cecal flora of mice infected with Trichuris muris (Schrank, 1788). Revista de Biologia Tropical 24, 251259.Google Scholar
Ransom, BH and Foster, WD (1920) Observations on the life history of Ascaris lumbricoides. United States Department of Agriculture Bulletin No. 818, Washington D.C. pp. 46.10.5962/bhl.title.64926CrossRefGoogle Scholar
Rosshart, SP, Herz, J, Vassallo, BG, Hunter, A, Wall, MK, Badger, JH, McCulloch, JA, Anastasakis, DG, Sarshad, AA, Leonardi, I, Collins, N, Blatter, JA, Han, SJ, Tamoutounour, S, Potapova, S, Foster St Claire, MB, Yuan, W, Sen, SK, Dreier, MS, Hild, B, Hafner, M, Wang, D, Iliev, ID, Belkaid, Y, Trinchieri, G and Rehermann, B (2019) Laboratory mice born to wild mice have natural microbiota and model human immune responses. Science 365(6452), eaaw4361.10.1126/science.aaw4361CrossRefGoogle ScholarPubMed
Shikhobalova, NP (1937) Experimental study of the chemotherapy of trichocephalosis. I. Trichocephalosis of white mice. Medskya Parazit 6, 389400.Google Scholar
Slotved, H-C, Eriksen, L, Murrell, KD and Nansen, P (1998) Early Ascaris suum migration in mice as a model for pigs. The Journal of Parasitology 84, 1618.10.2307/3284520CrossRefGoogle ScholarPubMed
Song, JS, Kim, JJ, Min, DY and Lee, KT (1985) Studies on the comparative migration patterns of Ascaris suum larvae between primary and re-infected mice. Korean Journal of Parasitology 23, 247252.10.3347/kjp.1985.23.2.247CrossRefGoogle ScholarPubMed
Sprent, JFA (1952) Migratory behaviour of the larvae of various Ascaris species in white mice. I. Distribution of larvae in the tissues. Journal of Infectious Diseases 90, 165176.10.1093/infdis/90.2.165CrossRefGoogle Scholar
Stewart, FH (1916) On the life history of Ascaris lumbricoides. Preliminary note. British Medical Journal 2896, 57.10.1136/bmj.2.2896.5CrossRefGoogle Scholar
Stewart, FH (1917) On the development of Ascaris lumbricoides Lin. and Ascaris suilla Duj. in the rat and mouse. Parasitology 9, 213227.10.1017/S0031182000006053CrossRefGoogle Scholar
Tsuji, N, Suzuki, K, Kasuga-Aoki, H, Matsumoto, Y, Arakawa, T, Ishiwata, K and Isobe, T (2001) Intranasal immunization with recombinant Ascaris suum 14-kilodalton antigen coupled with cholera toxin B subunit induces protective immunity to A. suum infection in mice. Infection and Immunity 69, 72857292.10.1128/IAI.69.12.7285-7292.2001CrossRefGoogle ScholarPubMed
Tsuji, N, Kasuga-Aoki, H, Isobe, T, Arakawa, T and Matsumoto, Y (2002) Cloning and characterisation of a highly immunoreactive 37 kDa antigen with multiimmunoglobulin domains from the swine roundworm Ascaris suum. International Journal for Parasitology 32, 17391746.10.1016/S0020-7519(02)00179-0CrossRefGoogle ScholarPubMed
Vaumourin, E, Vourc'h, G, Gasqui, P and Vayssier-Taussat, M (2015) The importance of multiparasitism: examining the consequences of coinfections for human and animal health. Parasites & Vectors 8, 545.10.1186/s13071-015-1167-9CrossRefGoogle ScholarPubMed
Vieira-Santos, F, Leal-Silva, T, de Lima Silva Padrao, L, Ruas, ACL, Nogueira, DS, Kraemer, L, Oliveira, FMS, Caliari, MV, Russo, RC, Fujiwara, RC and Bueno, LL (2021) Concomitant experimental coinfection by Plasmodium berghei NK65-NY and Ascaris suum downregulates the Ascaris-specific immune response and potentiates Ascaris-associated lung pathology. Malaria Journal 20, 296.10.1186/s12936-021-03824-wCrossRefGoogle ScholarPubMed
Wakelin, D (1967) Acquired immunity to Trichuris muris in the albino laboratory mouse. Parasitology 57, 515524.10.1017/S0031182000072395CrossRefGoogle ScholarPubMed
Wakelin, D (1970a) The stimulation of immunity and the induction of unresponsiveness to Trichuris muris in various strains of laboratory mice. Zeitschrift Fur Parasitenkunde 35, 162168.10.1007/BF00259993CrossRefGoogle Scholar
Wakelin, D (1970b) Studies on the immunity of albino mice to Trichuris muris. Suppression of immunity by cortisone acetate. Parasitology 60, 229237.10.1017/S0031182000078070CrossRefGoogle Scholar
Weatherhead, JE, Porter, P, Coffey, A, Haydel, D, Versteeg, L, Zhan, B, Gazzinelli Guimarães, AC, Fujiwara, R, Jaramillo, AM, Bottazzi, ME, Hotez, PJ, Corry, DB and Beaumier, CM (2018) Ascaris larval infection and lung invasion directly induce severe allergic airway disease in mice. Infection and Immunity 86, e00533–18.10.1128/IAI.00533-18CrossRefGoogle ScholarPubMed
Worley, DE, Meisenhelder, JE, Sheffield, HG and Thompson, PE (1962) Experimental studies on Trichuris muris in mice with an appraisal of its use for evaluating anthelmintics. The Journal of Parasitology 48, 433437.10.2307/3275209CrossRefGoogle ScholarPubMed
Figure 0

Table 1. Mouse models of Trichuris and Ascaris infections: a selection of the early pioneers