Review

Parasitology and Covid-19: epidemiological, immunological, pathological, and therapeutic aspects

  • Received: 07 June 2022 Revised: 22 December 2022 Accepted: 02 February 2023 Published: 03 March 2023
  • Studies suggest that there is a complex interaction between parasitic infections, human microbiota, and host immunity. Reports have shown that there is the prevalence of viral diseases have inverse correlations with their severities (as is the case for Covid-19), their resulting mortalities, and helminth infections in endemic areas. This review study was conducted to discover the possible association between parasitic infections and Covid-19 epidemics from immunological, pathological, and therapeutic aspects. Our studies were conducted by reviewing texts, reports, and articles on reputable websites such as PubMed, Science Direct, medRxvi, Google Scholar, and bioRxiv published by 2022 07 April for keywords such as a parasite, helminth, radioactive, COVID-19 or SARS-CoV-2. In particular, reports of co-infection with helminths with complications and severity of Covid-19 in endemic areas were considered. The findings indicate that parasitic helminths can regulate host immune responses associated with a viral infection. For example, intestinal parasitic infections may be effective in reducing the symptoms of SARS-CoV-2 and the complications of Covid-19. Infected hosts can induce an innate and Th2-compatible immune response to CD4+ T cells, eosinophils, and interleukins (IL-4, IL-5, and IL-10). Chronic helminth infections prevent strong immune responses by altering the host response to T helper 2 (Th2). Interestingly, some antimalarial drugs, such as Artemisinin-based combination therapies (ACTs), may inhibit SARS-CoV-2-induced severe acute respiratory syndrome (SARS). Parasitic infections may alter the host's immune response to SARS-CoV-2 with potentially beneficial or detrimental effects. However, more large-scale epidemiological studies are needed to uncover the links between parasitic infections and COVID-19 and to clarify existing ambiguities.

    Citation: Kamyar Nasiri, Zohreh Mortezania, Sanaz Oftadehbalani, Mohammad Khosousi Sani, Amin Daemi, Seyyedeh Touran Hosseini, Yusuf Döğüş, Zafer Yönden. Parasitology and Covid-19: epidemiological, immunological, pathological, and therapeutic aspects[J]. AIMS Allergy and Immunology, 2023, 7(1): 92-103. doi: 10.3934/Allergy.2023007

    Related Papers:

  • Studies suggest that there is a complex interaction between parasitic infections, human microbiota, and host immunity. Reports have shown that there is the prevalence of viral diseases have inverse correlations with their severities (as is the case for Covid-19), their resulting mortalities, and helminth infections in endemic areas. This review study was conducted to discover the possible association between parasitic infections and Covid-19 epidemics from immunological, pathological, and therapeutic aspects. Our studies were conducted by reviewing texts, reports, and articles on reputable websites such as PubMed, Science Direct, medRxvi, Google Scholar, and bioRxiv published by 2022 07 April for keywords such as a parasite, helminth, radioactive, COVID-19 or SARS-CoV-2. In particular, reports of co-infection with helminths with complications and severity of Covid-19 in endemic areas were considered. The findings indicate that parasitic helminths can regulate host immune responses associated with a viral infection. For example, intestinal parasitic infections may be effective in reducing the symptoms of SARS-CoV-2 and the complications of Covid-19. Infected hosts can induce an innate and Th2-compatible immune response to CD4+ T cells, eosinophils, and interleukins (IL-4, IL-5, and IL-10). Chronic helminth infections prevent strong immune responses by altering the host response to T helper 2 (Th2). Interestingly, some antimalarial drugs, such as Artemisinin-based combination therapies (ACTs), may inhibit SARS-CoV-2-induced severe acute respiratory syndrome (SARS). Parasitic infections may alter the host's immune response to SARS-CoV-2 with potentially beneficial or detrimental effects. However, more large-scale epidemiological studies are needed to uncover the links between parasitic infections and COVID-19 and to clarify existing ambiguities.



    加载中


    Conflict of interest



    The authors declare there is no conflict of interest.

    [1] Ramírez JD, Sordillo EM, Gotuzzo E, et al. (2020) SARS-CoV-2 in the Amazon region: a harbinger of doom for Amerindians. PLoS Negl Trop Dis 14: e0008686. https://doi.org/10.1371/journal.pntd.0008686
    [2] Paniz-Mondolfi AE, Ramírez JD, Delgado-Noguera LA, et al. (2021) Covid-19 and helminth infection: beyond the Th1/Th2 paradigm. PLoS Negl Trop Dis 15: e0009402. https://doi.org/10.1371/journal.pntd.0009402
    [3] Brosschot TP, Reynolds LA (2018) The impact of a helminth-modified microbiome on host immunity. Mol Immunol 11: 1039-1046. https://doi.org/10.1038/s41385-018-0008-5
    [4] Abdoli A (2020) Helminths and COVID-19 co-infections: a neglected critical challenge. ACS Pharmacol Transl Sci 3: 1039-1041. https://doi.org/10.1021/acsptsci.0c00141
    [5] Wolday D, Gebrecherkos T, Arefaine ZG, et al. (2021) Effect of co-infection with intestinal parasites on COVID-19 severity: A prospective observational cohort study. EClinicalMedicine 39: 101054. https://doi.org/10.1016/j.eclinm.2021.101054
    [6] Gutman JR, Lucchi NW, Cantey PT, et al. (2020) Malaria and parasitic neglected tropical diseases: potential syndemics with COVID-19?. Am J Trop Med Hyg 103: 572-577. https://doi.org/10.4269/ajtmh.20-0516
    [7] Osborne LC, Monticelli LA, Nice TJ, et al. (2014) Virus-helminth coinfection reveals a microbiota-independent mechanism of immunomodulation. Science 345: 578-582. https://doi.org/10.1126/science.1256942
    [8] Yang X, Yu Y, Xu J, et al. (2020) Clinical course and outcomes of critically ill patients with SARS-CoV-2 pneumonia in Wuhan, China: a single-centered, retrospective, observational study. Lancet Respir Med 8: 475-481. https://doi.org/10.1016/S2213-2600(20)30079-5
    [9] White MPJ, McManus CM, Maizels RM (2020) Regulatory T-cells in helminth infection: induction, function and therapeutic potential. Immunology 160: 248-260. https://doi.org/10.1111/imm.13190
    [10] Cepon-Robins TJ, Gildner TE (2020) Old friends meet a new foe: a potential role for immune-priming parasites in mitigating Covid-19 morbidity and mortality. Evol Med Public Health 2020: 234-248. https://doi.org/10.1093/emph/eoaa037
    [11] Sinha P, Matthay MA (2020) Is a “cytokine storm” relevant to COVID-19. JAMA Intern Med 180: 1152-1154. https://doi.org/10.1001/jamainternmed.2020.3313
    [12] Wolday D, Tasew G, Amogne W, et al. (2021) Interrogating the impact of intestinal parasite-microbiome on the pathogenesis of COVID-19 in Sub-Saharan Africa. Front Microbiol 12: 614522. https://doi.org/10.3389/fmicb.2021.614522
    [13] Siles-Lucas M, González-Miguel J, Geller R, et al. (2021) Potential influence of helminth molecules on COVID-19 pathology. Trends Parasitol 37: 11-14. https://doi.org/10.1016/j.pt.2020.10.002
    [14] Farsalinos K, Barbouni A, Niaura R (2020) Smoking, vaping and hospitalization for COVID-19. Qeios 2020: 1-10. https://doi.org/10.32388/Z69O8A.12
    [15] Rodriguez C, Veciana C (2020) Asthma and Covid-19: The eosinophilic link. Qeios 2020: 1-14. https://doi.org/10.32388/5IY4IF
    [16] Tanaka A, Allam VSRR, Simpson J, et al. (2018) The parasitic 68-mer peptide FhHDM-1 inhibits mixed granulocytic inflammation and airway hyperreactivity in experimental asthma. J Allergy Clin Immunol 141: 2316-2319.
    [17] Ramos-Benitez MJ, Ruiz-Jimenez C, Rosado-Franco JJ, et al. (2018) The parasitic 68-mer peptide FhHDM-1 inhibits mixed granulocytic inflammation and airway hyperreactivity in experimental asthma. J Allergy Clin Immunol 141: 2316-2319. https://doi.org/10.1016/j.jaci.2018.01.050
    [18] Oliveira SC, Figueiredo BC, Cardoso LS, et al. (2016) A double edged sword: Schistosoma mansoni Sm29 regulates both Th1 and Th2 responses in inflammatory mucosal diseases. Mucosal Immunol 9: 1366-1371. https://doi.org/10.1038/mi.2016.69
    [19] Zakeri A, Hansen EP, Andersen SD, et al. (2018) Immunomodulation by helminths: intracellular pathways and extracellular vesicles. Front Immunol 9: 2349. https://doi.org/10.3389/fimmu.2018.02349
    [20] Hosseini A, Hashemi V, Shomali N, et al. (2020) Innate and adaptive immune responses against coronavirus. Biomed Pharmacother 132: 110859. https://doi.org/10.1016/j.biopha.2020.110859
    [21] Shibabaw T, Molla MD, Teferi B, et al. (2020) Role of IFN and complements system: innate immunity in SARS-CoV-2. J Inflamm Res 13: 507-518. http://doi.org/10.2147/JIR
    [22] Ragab D, Salah Eldin H, Taeimah M, et al. (2020) The COVID-19 cytokine storm; what we know so far. Front Immunol 11: 1446. https://doi.org/10.3389/fimmu.2020.01446
    [23] Akelew Y, Andualem H, Ebrahim E, et al. (2022) Immunomodulation of COVID-19 severity by helminth co-infection: Implications for COVID-19 vaccine efficacy. Immun Inflamm Dis 10: e573. https://doi.org/10.1002/iid3.573
    [24] Orecchioni M, Ghosheh Y, Pramod AB, et al. (2019) Macrophage polarization: different gene signatures in M1 (LPS+) vs classically and M2 (LPS–) vs. alternatively activated macrophages. Front Immunol 10: 1084. https://doi.org/10.3389/fimmu.2019.01084
    [25] Stafford S, Lowell C, Sur S, et al. (2002) Lyn tyrosine kinase is important for IL-5-stimulated eosinophil differentiation. J Immunol 168: 1978-1983. https://doi.org/10.4049/jimmunol.168.4.1978
    [26] Rosenberg HF, Dyer KD, Domachowske JB (2008) Eosinophils and their interactions with respiratory virus pathogens. Immunol Res 43: 128-137. https://doi.org/10.1007/s12026-008-8058-5
    [27] Lee JJ, Helene FR (2012) Eosinophils in Health and Disease. Cambridge: Academic Press.
    [28] Giannis D, Ziogas IA, Gianni P (2020) Coagulation disorders in coronavirus infected patients: COVID-19, SARS-CoV-1, MERS-CoV and lessons from the past. J Clin Virol 127: 104362. https://doi.org/10.1016/j.jcv.2020.104362
    [29] Tang N, Li D, Wang X, et al. (2020) Abnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia. J Thromb Haemost 18: 844-847. https://doi.org/10.1111/jth.14768
    [30] Guan W, Ni Z, Hu Y, et al. (2020) Clinical characteristics of coronavirus disease 2019 in China. N Engl J Med 382: 1708-1720. https://doi/full/10.1056/neJMoa2002032
    [31] Klok FA, Kruip M, Van der Meer NJM, et al. (2020) Incidence of thrombotic complications in critically ill ICU patients with COVID-19. Thromb Res 191: 145-147. https://doi.org/10.1016/j.thromres.2020.04.013
    [32] Srichaikul T (1993) Hemostatic alterations in malaria. Southeast Asian J Trop Med Public Health 24: 86-91.
    [33] Maizels RM, McSorley HJ (2016) Regulation of the host immune system by helminth parasites. J Allergy Clin Immunol 138: 666-675. https://doi.org/10.1016/j.jaci.2016.07.007
    [34] Bashi T, Bizzaro G, Shor DBA, et al. (2015) The mechanisms behind helminth's immunomodulation in autoimmunity. Autoimmun Rev 14: 98-104. https://doi.org/10.1016/j.autrev.2014.10.004
    [35] Jackson JA, Friberg IM, Little S, et al. (2009) Review series on helminths, immune modulation and the hygiene hypothesis: immunity against helminths and immunological phenomena in modern human populations: coevolutionary legacies?. Immunology 126: 18-27. https://doi.org/10.1111/j.1365-2567.2008.03010.x
    [36] Rajasekaran S, Anuradha R, Bethunaickan R (2017) TLR specific immune responses against helminth infections. J Parasitol Res 2017: 1-9. https://doi.org/10.1155/2017/6865789
    [37] Monie TP (2017) Section 6–The Innate Immune System in Health and Disease. The Innate Immune System. Cambridge: Academic Press: 189-207.
    [38] Motran CC, Silvane L, Chiapello LS, et al. (2018) Helminth infections: recognition and modulation of the immune response by innate immune cells. Front Immunol 9: 664. https://doi.org/10.3389/fimmu.2018.00664
    [39] Maizels RM, Smits HH, McSorley HJ (2018) Modulation of host immunity by helminths: the expanding repertoire of parasite effector molecules. Immunity 49: 801-818. https://doi.org/10.1016/j.immuni.2018.10.016
    [40] Zhu J (2015) T helper 2 (Th2) cell differentiation, type 2 innate lymphoid cell (ILC2) development and regulation of interleukin-4 (IL-4) and IL-13 production. Cytokine 75: 14-24. https://doi.org/10.1016/j.cyto.2015.05.010
    [41] Rodriguez C (2020) The global helminth belt and Covid-19: the new eosinophilic link. Qeios 2020: 1-18. https://doi.org/10.32388/IWKQH9.2
    [42] Eichenberger RM, Ryan S, Jones L, et al. (2018) Hookworm secreted extracellular vesicles interact with host cells and prevent inducible colitis in mice. Front Immunol 9: 850. https://doi.org/10.3389/fimmu.2018.00850
    [43] Al-Riyami L, Harnett W (2012) Immunomodulatory properties of ES-62, a phosphorylcholine-containing glycoprotein secreted by Acanthocheilonema viteae. Endocr Metab Immune Disord Drug Targets 12: 45-52. https://doi.org/10.2174/187153012799278893
    [44] Chacon N, Chacin-Bonilla L, Cesari IM (2021) Implications of helminth immunomodulation on Covid-19 co-infections. Life Res 4: 26. https://doi.org/10.53388/life2021-0502-309
    [45] Lokken KL, Stull-Lane AR, Poels K, et al. (2018) Malaria parasite-mediated alteration of macrophage function and increased iron availability predispose to disseminated nontyphoidal Salmonella infection. Infect Immun 86: e00301-e00318. https://doi.org/10.1128/IAI.00301-18
    [46] Edwards CL, Zhang V, Werder RB, et al. (2015) Coinfection with blood-stage Plasmodium promotes systemic type I interferon production during pneumovirus infection but impairs inflammation and viral control in the lung. Clin Vaccine Immunol 22: 477-483. https://doi.org/10.1128/CVI.00051-15
    [47] Thakur N, Rai N (2016) Alarmingly high incidence of eosinophilia in barabanki and neighboring districts of eastern uttar pradesh: A prospective hospital-based study. J Trop Pediatr 65: 500-502. https://doi.org/10.1093/tropej/fmw035
    [48] Drake MG, Bivins-Smith ER, Proskocil BJ, et al. (2016) Human and mouse eosinophils have antiviral activity against parainfluenza virus. Am J Respir Cell Mol Biol 55: 387-394. https://doi.org/10.1165/rcmb.2015-0405oc
    [49] Domachowske JB, Bonville CA, Dyer KD, et al. (2000) Pulmonary eosinophilia and production of MIP-1α are prominent responses to infection with pneumonia virus of mice. Cellu Immun 200: 98-104. https://doi.org/10.1006/cimm.2000.1620
    [50] Couper KN, Blount DG, Riley EM (2008) IL-10: the master regulator of immunity to infection. J Immunol 180: 5771-5777. https://doi.org/10.4049/jimmunol.180.9.5771
    [51] Logan J, Navarro S, Loukas A, et al. (2018) Helminth-induced regulatory T cells and suppression of allergic responses. Curr Opin Immunol 54: 1-6. https://doi.org/10.1016/j.coi.2018.05.007
    [52] Anuradha R, Munisankar S, Bhootra Y, et al. (2016) IL-10-and TGFβ-mediated Th9 Responses in a human helminth infection. PLoS Negl Trop Dis 10: e0004317. https://doi.org/10.1371/journal.pntd.0004317
    [53] Xu X, Han M, Li T, et al. (2020) Effective treatment of severe COVID-19 patients with tocilizumab. Proc Natl Acad Sci USA 117: 10970-10975. https://doi.org/10.1073/pnas.2005615117
  • Reader Comments
  • © 2023 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(1333) PDF downloads(80) Cited by(0)

Article outline

Figures and Tables

Figures(2)

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog