Разнонаправленное влияние гельминтов на аллергические заболевания и иммунитет
https://doi.org/10.31146/1682-8658-ecg-231-11-157-163
Аннотация
Об авторах
Е. В. НадейРоссия
Е. С. Лепехина
Россия
Е. А. Лялюкова
Россия
Е. В. Усачева
Россия
Список литературы
1. Hossny E., Ebisawa M., El-Gamal Y. et al. Challenges of managing food allergy in the developing world. World Allergy Organ J. 2019;12(11):100089. doi: 10.1016/j.waojou.2019.100089.
2. Potaczek D.P., Hard H., Michel S. et al. Epigenetics and allergy: from basic mechanisms to clinical applications. Epigenomics. 2017;9(4):539-571. doi: 10.2217/epi-2016-0162.
3. Figueiredo C.A., Barreto M.L., Alcantara-Neves N.M. et al. Coassociations between IL10 polymorphisms, IL-10 production, helminth infection, and asthma/wheeze in an urban tropical population in Brazil. Allergy Clin Immunol. 2013;131(6):1683-90. doi: 10.1016/j.jaci.2012.10.043.
4. Zaiss M.M., Harris N.L.Interactions between the intestinal microbiome and helminth parasites. Parasite Immunol. 2016;38(1):5-11. doi: 10.1111/pim.12274.
5. Navarro S., Ferreira I., Loukas A. The hookworm pharmacopoeia for inflammatory diseases.International Journal for Parasitology. 2013;43(3-4), 225-231. doi: 10.1016/j.ijpara.2012.11.005.
6. Sroka-Tomaszewska J., Trzeciak M. Molecular Mechanisms of Atopic Dermatitis Pathogenesis.Int J Mol Sci. 2021;22(8):4130. doi: 10.3390/ijms22084130.
7. Hammad H., Lambrecht B.N. The basic immunology of asthma. Cell. 2021;184(6):1469-1485. doi: 10.1016/j.cell.2021.02.016.
8. Wise S.K., Damask C., Roland L.T. et al.International consensus statement on allergy and rhinology: Allergic rhinitis - 2023.Int Forum Allergy Rhinol. 2023;13(4):293-859. doi: 10.1002/alr.23090.
9. Peters R.L., Krawiec M., Koplin J.J., Santos A.F. Update on food allergy. Pediatr Allergy Immunol. 2021;32(4):647-657. doi: 10.1111/pai.13443.
10. Grencis R.K. Immunity to helminths: resistance, regulation, and susceptibility to gastrointestinal nematodes. Annu Rev Immunol. 2015;33:201-25. doi: 10.1146/annurev-immunol-032713-120218.
11. Medzhitov R., Schneider D.S., Soares M.P. Disease tolerance as a defense strategy. Science. 2012;335(6071):936-41. doi: 10.1126/science.1214935.
12. Maizels R.M, Yazdanbakhsh M. Immune regulation by helminth parasites: cellular and molecular mechanisms. Nat Rev Immunol. 2003;3(9):733-44. doi: 10.1038/nri1183.
13. McSorley H.J., Maizels R.M. Helminth infections and host immune regulation. Clin Microbiol Rev. 2012;25(4):585-608. doi: 10.1128/CMR.05040-11.
14. Mbow M., Larkin B.M., Meurs L. et al. T-helper 17 cells are associated with pathology in human schistosomiasis. J Infect Dis. 2013;207(1):186-95. doi: 10.1093/infdis/jis654.
15. Nogueira D.S., Gazzinelli-Guimarães P.H., Barbosa F.S. et al. Multiple Exposures to Ascaris suum Induce Tissue Injury and Mixed Th2/Th17 Immune Response in Mice. PLoS Negl Trop Dis. 2016;10(1): e0004382. doi: 10.1371/journal.pntd.0004382.
16. Babu S., Bhat S.Q., Pavan Kumar N. et al. Filarial lymphedema is characterized by antigen-specific Th1 and th17 proinflammatory responses and a lack of regulatory T cells. PLoS Negl Trop Dis. 2009;3(4): e420. doi: 10.1371/journal.pntd.0000420.
17. Turner J.E., Morrison P.J., Wilhelm C. et al. IL-9-mediated survival of type 2 innate lymphoid cells promotes damage control in helminth-induced lung inflammation. J Exp Med. 2013;210(13):2951-65. doi: 10.1084/jem.20130071.
18. Van Tong H., Brindley P.J., Meyer C.G., Velavan T.P. Parasite Infection, Carcinogenesis and Human Malignancy. EBioMedicine. 2017;15:12-23. doi: 10.1016/j.ebiom.2016.11.034.
19. Smout M.J., Sotillo J., Laha T. et al. Carcinogenic Parasite Secretes Growth Factor That Accelerates Wound Healing and Potentially Promotes Neoplasia. PLoS Pathog. 2015;11(10): e1005209. doi: 10.1371/journal.ppat.1005209.
20. Tsai M., Starkl P., Marichal T., Galli S.J. Testing the ‘toxin hypothesis of allergy’: mast cells, IgE, and innate and acquired immune responses to venoms. Curr Opin Immunol. 2015;36:80-7. doi: 10.1016/j.coi.2015.07.001.
21. Torow N., Marsland B.J., Hornef M.W., Gollwitzer E.S. Neonatal mucosal immunology. Mucosal Immunol. 2017;10(1):5-17. doi: 10.1038/mi.2016.81.
22. Saluzzo S., Gorki A.D., Rana B.M.J., Martins R. et al. First-Breath-Induced Type 2 Pathways Shape the Lung Immune Environment. Cell Rep. 2017;18(8):1893-1905. doi: 10.1016/j.celrep.2017.01.071.
23. Odegaard J.I., Lee M.W., Sogawa Y. et al. Perinatal Licensing of Thermogenesis by IL-33 and ST2. Cell. 2016;166(4):841-854. doi: 10.1016/j.cell.2016.06.040.
24. Knipper J.A., Willenborg S., Brinckmann J. et al.Interleukin-4 Receptor α Signaling in Myeloid Cells Controls Collagen Fibril Assembly in Skin Repair. Immunity. 2015;43(4):803-16. doi: 10.1016/j.immuni.2015.09.005.
25. Heredia J.E., Mukundan L., Chen F.M. et al. Type 2 innate signals stimulate fibro/adipogenic progenitors to facilitate muscle regeneration. Cell. 2013;153(2):376-88. doi: 10.1016/j.cell.2013.02.053.
26. Goh Y.P., Henderson N.C., Heredia J.E. et al. Eosinophils secrete IL-4 to facilitate liver regeneration. Proc Natl Acad Sci U S A. 2013;110(24):9914-9. doi: 10.1073/pnas.1304046110.
27. Blériot C., Dupuis T., Jouvion G. et al. Liver-resident macrophage necroptosis orchestrates type 1 microbicidal inflammation and type-2-mediated tissue repair during bacterial infection. Immunity. 2015;42(1):145-58. doi: 10.1016/j.immuni.2014.12.020.
28. Strachan D.P. Hay fever, hygiene, and household size. BMJ. 1989;299(6710):1259-60. doi: 10.1136/bmj.299. 6710.1259.
29. Rook G.A., Adams V., Hunt J. et al. Mycobacteria and other environmental organisms as immunomodulators for immunoregulatory disorders. Springer Semin Immunopathol. 2004;25(3-4):237-55. doi: 10.1007/s00281-003-0148-9.
30. Haahtela T. A biodiversity hypothesis. Allergy. 2019;74(8):1445-1456. doi: 10.1111/all.13763.
31. Santiago H.C., Nutman T.B. Human Helminths and Allergic Disease: The Hygiene Hypothesis and Beyond. Am J Trop Med Hyg. 2016;95(4):746-753. doi: 10.4269/ajtmh.16-0348.
32. Leonardi-Bee J., Pritchard D., Britton J. Asthma and current intestinal parasite infection: systematic review and meta-analysis. Am J Respir Crit Care Med. 2006;174(5):514-23. doi: 10.1164/rccm.200603-331OC.
33. Feary J., Britton J., Leonardi-Bee J. Atopy and current intestinal parasite infection: a systematic review and meta-analysis. Allergy. 2011;66(4):569-78. doi: 10.1111/j.1398-9995.2010.02512.x.
34. Li L., Gao W., Yang X. et al. Asthma and toxocariasis. Ann Allergy Asthma Immunol. 2014;113(2):187-92. doi: 10.1016/j.anai.2014.05.016.
35. Mohammadzadeh I., Riahi S.M., Saber V. et al. The relationship between Toxocara species seropositivity and allergic skin disorders: a systematic review and meta-analysis. Trans R Soc Trop Med Hyg. 2018;112(12):529-537. doi: 10.1093/trstmh/try094.
36. Arrais M., Maricoto T., Nwaru B.I. et al. Helminth infections and allergic diseases: Systematic review and meta-analysis of the global literature. J Allergy Clin Immunol. 2022;149(6):2139-2152. doi: 10.1016/j.jaci.2021.12.777.
37. Burbank A.J., Sood A.K., Kesic M.J. et al. Environmental determinants of allergy and asthma in early life. J Allergy Clin Immunol. 2017;140(1):1-12. doi: 10.1016/j.jaci.2017.05.010.
38. Jackson D.J., Makrinioti H., Rana B.M. et al. IL-33-dependent type 2 inflammation during rhinovirus-induced asthma exacerbations in vivo. Am J Respir Crit Care Med. 2014;190(12):1373-82. doi: 10.1164/rccm.201406-1039OC.
39. Osbourn M., Soares D.C., Vacca F. et al. HpARI Protein Secreted by a Helminth Parasite Suppresses Interleukin-33. Immunity. 2017;47(4):739-751.e5. doi: 10.1016/j.immuni.2017.09.015.
40. McFarlane A.J., McSorley H.J., Davidson D.J. et al. Enteric helminth-induced type I interferon signaling protects against pulmonary virus infection through interaction with the microbiota. J Allergy Clin Immunol. 2017;140(4):1068-1078.e6. doi: 10.1016/j.jaci.2017.01.016.
41. Scheer S., Krempl C., Kallfass C. et al. S. mansoni bolsters anti-viral immunity in the murine respiratory tract. PLoS One. 2014;9(11): e112469. doi: 10.1371/journal.pone.0112469.
42. Zimmermann P., Messina N., Mohn W.W. et al. Association between the intestinal microbiota and allergic sensitization, eczema, and asthma: A systematic review. J Allergy Clin Immunol. 2019;143(2):467-485. doi: 10.1016/j.jaci.2018.09.025.
43. Gollwitzer E.S., Saglani S., Trompette A. et al. Lung microbiota promotes tolerance to allergens in neonates via PD-L1. Nat Med. 2014;20(6):642-7. doi: 10.1038/nm.3568.
44. Ramanan D., Bowcutt R., Lee S.C. et al. Helminth infection promotes colonization resistance via type 2 immunity. Science. 2016;352(6285):608-12. doi: 10.1126/science.aaf3229.
45. Jenkins T.P., Rathnayaka Y., Perera P.K. et al. Infections by human gastrointestinal helminths are associated with changes in faecal microbiota diversity and composition. PLoS One. 2017;12(9): e0184719. doi: 10.1371/journal.pone.0184719.
46. Zaiss M.M., Harris N.L.Interactions between the intestinal microbiome and helminth parasites. Parasite Immunol. 2016;38(1):5-11. doi: 10.1111/pim.12274.
47. Peachey L.E., Jenkins T.P., Cantacessi C. This Gut Ain’t Big Enough for Both of Us. Or Is It? Helminth-Microbiota Interactions in Veterinary Species. Trends Parasitol. 2017;33(8):619-632. doi: 10.1016/j.pt.2017.04.004.
48. Reynolds L.A., Finlay B.B. Early life factors that affect allergy development. Nat Rev Immunol. 2017;17(8):518-528. doi: 10.1038/nri.2017.39.
49. Lee S.C., Tang M.S., Lim Y.A. et al. Helminth colonization is associated with increased diversity of the gut microbiota. PLoS Negl Trop Dis. 2014;8(5): e2880. doi: 10.1371/journal.pntd.0002880.
50. Ramanan D., Bowcutt R., Lee S.C. et al. Helminth infection promotes colonization resistance via type 2 immunity. Science. 2016;352(6285):608-12. doi: 10.1126/science.aaf3229.
51. Rausch S., Midha A., Kuhring M. et al. Parasitic Nematodes Exert Antimicrobial Activity and Benefit From Microbiota-Driven Support for Host Immune Regulation. Front Immunol. 2018;9:2282. doi: 10.3389/fimmu.2018.02282.
52. Robinson M.W., Hutchinson A.T., Dalton J.P., Donnelly S. Peroxiredoxin: a central player in immune modulation. Parasite Immunol. 2010;32(5):305-13. doi: 10.1111/j.1365-3024.2010.01201.x.
53. Zaiss M.M., Harris N.L.Interactions between the intestinal microbiome and helminth parasites. Parasite Immunol. 2016;38(1):5-11. doi: 10.1111/pim.12274.
54. Moyat M., Lebon L., Perdijk O. et al. Microbial regulation of intestinal motility provides resistance against helminth infection. Mucosal Immunol. 2022;15(6):1283-1295. doi: 10.1038/s41385-022-00498-8.
55. Jin X., Liu Y., Wang J. et al. β-Glucan-triggered Akkermansia muciniphila expansion facilitates the expulsion of intestinal helminth via TLR2 in mice. Carbohydr Polym. 2022;275:118719. doi: 10.1016/j.carbpol.2021.118719.
56. Rosa B.A., Supali T., Gankpala L. et al. Differential human gut microbiome assemblages during soil-transmitted helminth infections in Indonesia and Liberia. Microbiome. 2018;6(1):33. doi: 10.1186/s40168-018-0416-5.
57. Sakaguchi S., Yamaguchi T., Nomura T., Ono M. Regulatory T cells and immune tolerance. Cell. 2008;133(5):775-87. doi: 10.1016/j.cell.2008.05.009.
58. Belkaid Y., Tarbell K. Regulatory T cells in the control of host-microorganism interactions. Annu Rev Immunol. 2009;27:551-89. doi: 10.1146/annurev.immunol.021908.132723.
59. Belkaid Y. Regulatory T cells and infection: a dangerous necessity. Nat Rev Immunol. 2007;7(11):875-88. doi: 10.1038/nri2189.
60. Abdoli A., Mirzaian Ardakani H. Potential application of helminth therapy for resolution of neuroinflammation in neuropsychiatric disorders. Metab Brain Dis. 2020;35(1):95-110. doi: 10.1007/s11011-019-00466-5.
61. Shimokawa C., Kato T., Takeuchi T. et al. CD8+ regulatory T cells are critical in prevention of autoimmune-mediated diabetes. Nat Commun. 2020;11(1):1922. doi: 10.1038/s41467-020-15857-x.
62. Xu J., Liu M., Yu P. Et al. Effect of recombinant Trichinella spiralis cysteine proteinase inhibitor on TNBS-induced experimental inflammatory bowel disease in mice.Int Immunopharmacol. 2019;66:28-40. doi: 10.1016/j.intimp.2018.10.043.
63. Summers R.W., Elliott D.E., Urban J.F. et al. Trichuris suis therapy for active ulcerative colitis: a randomized controlled trial. Gastroenterology. 2005;128(4): 825-32. doi: 10.1053/j.gastro.2005.01.005.
64. Tong H., Brindley P.J., Meyer C.G., Velavan T.P. Parasite Infection, Carcinogenesis and Human Malignancy. EBioMedicine. 2017;15:12-23.doi: 10.1016/j.ebiom.2016.11.034.
65. Fleming J.O., Isaak A., Lee J.E. et al. Probiotic helminth administration in relapsing-remitting multiple sclerosis: a phase 1 study. Mult Scler 2011;17:743-54. doi:10.1177/1352458511398054.
66. Wammes L.J., Mpairwe H., Elliott A.M. et al. Helminth therapy or elimination: epidemiological, immunological, and clinical considerations. Lancet Infect Dis. 2014;14(11):1150-1162. doi: 10.1016/S1473-3099(14)70771-6.
67. Croese J., Giacomin P., Navarro S. et al. Experimental hookworm infection and gluten microchallenge promote tolerance in celiac disease. J Allergy Clin Immunol. 2015;135(2):508-16. doi: 10.1016/j.jaci.2014.07.022.
68. Daveson A.J., Jones D.M., Gaze S. et al. Effect of hookworm infection on wheat challenge in celiac disease - a randomised double-blinded placebo-controlled trial. PLoS One. 2011;6(3): e17366. doi: 10.1371/journal.pone.0017366.
69. Navarro S., Pickering D.A., Ferreira I.B. et al. Hookworm recombinant protein promotes regulatory T cell responses that suppress experimental asthma. Sci Transl Med. 2016;8(362):362ra143. doi: 10.1126/scitranslmed.aaf8807.
Рецензия
Для цитирования:
Надей Е.В., Лепехина Е.С., Лялюкова Е.А., Усачева Е.В. Разнонаправленное влияние гельминтов на аллергические заболевания и иммунитет. Экспериментальная и клиническая гастроэнтерология. 2024;(11):157-163. https://doi.org/10.31146/1682-8658-ecg-231-11-157-163
For citation:
Nadey E.V., Lepehina E.S., Lyalyukova E.A., Usacheva E.V. Multidirectional influence of helminths on allergic diseases and immunity. Experimental and Clinical Gastroenterology. 2024;(11):157-163. (In Russ.) https://doi.org/10.31146/1682-8658-ecg-231-11-157-163