Strongyloides ratti infection in mice: immune response and immune modulation

Strongyloides ratti is a natural parasite of wild rats and most laboratory mouse strains are also fully permissive. The infection can be divided into three distinct phases: the tissue migration of the infective third stage larvae during the first two days, the early intestinal establishment of S. ratti parasites molting to adults on days three to six and the later intestinal parasitic phase until the end of infection. Immunocompetent mice terminate the S. ratti infection after one month and are semi-resistant to a second infection. Employing the powerful tools of mouse immunology has facilitated a detailed analysis of the initiation, execution and regulation of the immune response to S. ratti. Here we review the information collected to date on the protective immune response to migrating S. ratti larvae in tissues and to adult parasites in the intestine. We show that depending on the phase of infection, a site-specific portfolio of immune effector mechanisms is required for infection control. In addition, we summarize the strategies employed by S. ratti to evade the immune system and survive long enough in its host to replicate despite an effective immune response. Selected murine studies using the closely related Strongyloides venezuelensis will be discussed. This article is part of the Theo Murphy meeting issue ‘Strongyloides: omics to worm-free populations’.

[1]  S. Lira,et al.  Neuropeptide regulation of non-redundant ILC2 responses at barrier surfaces , 2022, Nature.

[2]  D. Beule,et al.  Non-redundant functions of group 2 innate lymphoid cells , 2022, Nature.

[3]  S. Ziegler,et al.  TSLP, IL-33, and IL-25: not just for allergy and helminth infection. , 2022, The Journal of allergy and clinical immunology.

[4]  Alexandra Ehrens,et al.  Eosinophils and Neutrophils Eliminate Migrating Strongyloides ratti Larvae at the Site of Infection in the Context of Extracellular DNA Trap Formation , 2021, Frontiers in Immunology.

[5]  T. Nolan,et al.  Transgenic expression of a T cell epitope in Strongyloides ratti reveals that helminth-specific CD4+ T cells constitute both Th2 and Treg populations , 2021, PLoS pathogens.

[6]  T. Kikuchi,et al.  Venestatin from parasitic helminths interferes with receptor for advanced glycation end products (RAGE)-mediated immune responses to promote larval migration , 2021, PLoS pathogens.

[7]  K. Matsushita,et al.  Thymic stromal lymphopoietin contributes to protection of mice from Strongyloides venezuelensis infection by CD4+ T cell-dependent and -independent pathways. , 2021, Biochemical and biophysical research communications.

[8]  B. Blankenhaus,et al.  Elucidating different pattern of immunoregulation in BALB/c and C57BL/6 mice and their F1 progeny , 2021, Scientific reports.

[9]  H. McSorley,et al.  IL-33 facilitates rapid expulsion of the parasitic nematode Strongyloides ratti from the intestine via ILC2- and IL-9-driven mast cell activation , 2020, PLoS pathogens.

[10]  W. F. Gregory,et al.  A Truncated Form of HpARI Stabilizes IL-33, Amplifying Responses to the Cytokine , 2020, Frontiers in Immunology.

[11]  D. Bisanzio,et al.  The Global Prevalence of Strongyloides stercoralis Infection , 2020, Pathogens.

[12]  M. Montes,et al.  Regulatory T cell expansion resolves after effective strongyloidiasis treatment in subjects with HTLV-1 co-infection. , 2020, Parasitology international.

[13]  D. Voehringer,et al.  Basophils are dispensable for the establishment of protective adaptive immunity against primary and challenge infection with the intestinal helminth parasite Strongyloides ratti , 2018, PLoS neglected tropical diseases.

[14]  E. Liebau,et al.  Comparative characterization of two galectins excreted-secreted from intestine-dwelling parasitic versus free-living females of the soil-transmitted nematode Strongyloides. , 2018, Molecular and biochemical parasitology.

[15]  M. Breloer,et al.  Interleukin-9 promotes early mast cell-mediated expulsion of Strongyloides ratti but is dispensable for generation of protective memory , 2018, Scientific Reports.

[16]  M. Breloer,et al.  Interleukin-9 promotes early mast cell-mediated expulsion of Strongyloides ratti but is dispensable for generation of protective memory , 2018, Scientific Reports.

[17]  T. Nutman,et al.  Altered levels of memory T cell subsets and common γc cytokines in Strongyloides stercoralis infection and partial reversal following anthelmintic treatment , 2018, PLoS neglected tropical diseases.

[18]  T. Nutman,et al.  Elevated Systemic Levels of Eosinophil, Neutrophil, and Mast Cell Granular Proteins in Strongyloides Stercoralis Infection that Diminish following Treatment , 2018, Front. Immunol..

[19]  T. Le Bihan,et al.  HpARI Protein Secreted by a Helminth Parasite Suppresses Interleukin-33 , 2017, Immunity.

[20]  S. Hartmann,et al.  Th2/1 Hybrid Cells Occurring in Murine and Human Strongyloidiasis Share Effector Functions of Th1 Cells , 2017, Front. Cell. Infect. Microbiol..

[21]  M. Kubo,et al.  Differences in the Importance of Mast Cells, Basophils, IgE, and IgG versus That of CD4+ T Cells and ILC2 Cells in Primary and Secondary Immunity to Strongyloides venezuelensis , 2017, Infection and Immunity.

[22]  E. Liebau,et al.  Multifunctional Thioredoxin-Like Protein from the Gastrointestinal Parasitic Nematodes Strongyloides ratti and Trichuris suis Affects Mucosal Homeostasis , 2016, Journal of parasitology research.

[23]  D. Voehringer,et al.  Mucosal mast cells are indispensable for the timely termination of Strongyloides ratti infection , 2016, Mucosal Immunology.

[24]  E. Liebau,et al.  SPARC (secreted protein acidic and rich in cysteine) of the intestinal nematode Strongyloides ratti is involved in mucosa-associated parasite-host interaction. , 2016, Molecular and biochemical parasitology.

[25]  T. Nutman Human infection with Strongyloides stercoralis and other related Strongyloides species , 2016, Parasitology.

[26]  T. Kikuchi,et al.  Strongyloides ratti and S. venezuelensis – rodent models of Strongyloides infection , 2016, Parasitology.

[27]  D. Abraham,et al.  Strongyloides infection in rodents: immune response and immune regulation , 2016, Parasitology.

[28]  M. Eschbach,et al.  Cutting Edge: The BTLA–HVEM Regulatory Pathway Interferes with Protective Immunity to Intestinal Helminth Infection , 2015, The Journal of Immunology.

[29]  J. Girard,et al.  IL-33: an alarmin cytokine with crucial roles in innate immunity, inflammation and allergy. , 2014, Current opinion in immunology.

[30]  T. Nolan,et al.  Extracellular traps are associated with human and mouse neutrophil and macrophage mediated killing of larval Strongyloides stercoralis. , 2014, Microbes and infection.

[31]  M. Eschbach,et al.  Foxp3+ Regulatory T Cells Delay Expulsion of Intestinal Nematodes by Suppression of IL-9-Driven Mast Cell Activation in BALB/c but Not in C57BL/6 Mice , 2014, PLoS pathogens.

[32]  R. Flavell,et al.  Th9 Cells Drive Host Immunity against Gastrointestinal Worm Infection. , 2013, Immunity.

[33]  P. Vounatsou,et al.  Strongyloides stercoralis: Global Distribution and Risk Factors , 2013, PLoS neglected tropical diseases.

[34]  H. Maruyama,et al.  The role of B-cells in immunity against adult Strongyloides venezuelensis , 2013, Parasites & Vectors.

[35]  M. Muramatsu,et al.  IgG and IgE Collaboratively Accelerate Expulsion of Strongyloides venezuelensis in a Primary Infection , 2013, Infection and Immunity.

[36]  H. Rodewald,et al.  Widespread immunological functions of mast cells: fact or fiction? , 2012, Immunity.

[37]  M. Breloer,et al.  Passive immunization with a monoclonal IgM antibody specific for Strongyloides ratti HSP60 protects mice against challenge infection. , 2012, Vaccine.

[38]  Kenji Nakanishi,et al.  Contribution of IL-33–activated type II innate lymphoid cells to pulmonary eosinophilia in intestinal nematode-infected mice , 2012, Proceedings of the National Academy of Sciences.

[39]  C. Benoist,et al.  Cre-mediated cell ablation contests mast cell contribution in models of antibody- and T cell-mediated autoimmunity. , 2011, Immunity.

[40]  D. Abraham,et al.  Innate and adaptive immunity to the nematode Strongyloides stercoralis in a mouse model , 2011, Immunologic research.

[41]  M. Mitreva,et al.  Life Cycle Stage-resolved Proteomic Analysis of the Excretome/Secretome from Strongyloides ratti—Identification of Stage-specific Proteases , 2011, Molecular & Cellular Proteomics.

[42]  B. Stockinger,et al.  Interleukin 9 fate reporter reveals induction of innate IL-9 response in lung inflammation , 2011, Nature Immunology.

[43]  A. Waisman,et al.  Mast cells are key promoters of contact allergy that mediate the adjuvant effects of haptens. , 2011, Immunity.

[44]  R. Maizels,et al.  Diversity and dialogue in immunity to helminths , 2011, Nature Reviews Immunology.

[45]  C. Loddenkemper,et al.  Strongyloides ratti Infection Induces Expansion of Foxp3+ Regulatory T Cells That Interfere with Immune Response and Parasite Clearance in BALB/c Mice , 2011, The Journal of Immunology.

[46]  D. Voehringer,et al.  Basophils orchestrate chronic allergic dermatitis and protective immunity against helminths. , 2010, Immunity.

[47]  M. Eschbach,et al.  Strongyloides ratti infection induces transient nematode‐specific Th2 response and reciprocal suppression of IFN‐γ production in mice , 2010, Parasite immunology.

[48]  K. Murphy,et al.  Slow down and survive: Enigmatic immunoregulation by BTLA and HVEM. , 2010, Annual review of immunology.

[49]  J. Lake,et al.  Regulatory T Cell Expansion in HTLV-1 and Strongyloidiasis Co-infection Is Associated with Reduced IL-5 Responses to Strongyloides stercoralis Antigen , 2009, PLoS neglected tropical diseases.

[50]  D. Aronoff,et al.  Counterregulation of Th2 immunity by interleukin 12 reduces host defenses against Strongyloides venezuelensis infection. , 2009, Microbes and infection.

[51]  C. Bleay,et al.  The immune response during a Strongyloides ratti infection of rats , 2007, Parasite immunology.

[52]  C. Loddenkemper,et al.  Selective depletion of Foxp3+ regulatory T cells induces a scurfy-like disease , 2007, The Journal of experimental medicine.

[53]  M. Teixeira,et al.  Expression of IL-4 receptor on non-bone marrow-derived cells is necessary for the timely elimination of Strongyloides venezuelensis in mice, but not for intestinal IL-4 production. , 2006, International journal for parasitology.

[54]  D. Gems,et al.  Extraordinary plasticity in aging in Strongyloides ratti implies a gene‐regulatory mechanism of lifespan evolution , 2006, Aging cell.

[55]  K. Kishihara,et al.  Strongyloides ratti: the role of interleukin-5 in protection against tissue migrating larvae and intestinal adult worms. , 2003, Journal of helminthology.

[56]  S. Shimada,et al.  Intraepithelial infiltration of eosinophils and their contribution to the elimination of adult intestinal nematode, Strongyloides venezuelensis in mice. , 2003, Parasitology international.

[57]  M. Ono,et al.  Mucosal Defense against Gastrointestinal Nematodes: Responses of Mucosal Mast Cells and Mouse Mast Cell Protease 1 during PrimaryStrongyloides venezuelensis Infection in FcRγ-Knockout Mice , 2000, Infection and Immunity.

[58]  K. Kishihara,et al.  The crucial role of granulocytes in the early host defense against Strongyloides ratti infection in mice , 2000, Parasitology Research.

[59]  Young,et al.  Regulation of primary Strongyloides ratti infections in mice: a role for interleukin‐5 , 1998, Immunology.

[60]  G. Dranoff,et al.  Role for interleukin-3 in mast-cell and basophil development and in immunity to parasites , 1998, Nature.

[61]  A. Takamure Migration route of Strongyloides venezuelensis in rodents. , 1995, International journal for parasitology.

[62]  I. Taşdemir,et al.  Intestinal protection against Strongyloides ratti and mastocytosis induced by administration of interleukin-3 in mice. , 1993, Immunology.

[63]  Y. Horii,et al.  Mucosal mast cells and the expulsive mechanisms of mice against Strongyloides venezuelensis. , 1993, International journal for parasitology.

[64]  N. Yamaguchi,et al.  The role of interleukin-5 in protective immunity to Strongyloides venezuelensis infection in mice. , 1991, Immunology.

[65]  Y. Sato,et al.  Strongyloides venezuelensis infections in mice. , 1990, International journal for parasitology.

[66]  Y. Nawa,et al.  Worm expulsion and mucosal mast cell response induced by repetitive IL-3 administration in Strongyloides ratti-infected nude mice. , 1988, Immunology.

[67]  M. Kiyota,et al.  Defective protective capacity of W/Wv mice against Strongyloides vatti infection and its reconstitution with bone marrow cells , 1985, Parasite immunology.

[68]  H. Dawkins,et al.  The occurrence of Strongyloides ratti in the tissues of mice after percutaneous infection , 1982, Journal of Helminthology.

[69]  H. Dawkins,et al.  Transfer by serum and cells of resistance to infection with Strongyloides ratti in mice. , 1981, Immunology.

[70]  H. Dawkins,et al.  Histopathological appearances in primary and secondary infections with Strongyloides ratti in mice. , 1981, International journal for parasitology.

[71]  G. Mitchell,et al.  Strongyloides ratti: susceptibility to infection and resistance to reinfection in inbred strains of mice as assessed by excretion of larvae. , 1980, International journal for parasitology.

[72]  M. Viney,et al.  The biology of Strongyloides spp. , 2015, WormBook : the online review of C. elegans biology.