Active infection with <i>Onchocerca volvulus</i> and the linkage to epilepsy/nodding syndrome

Citation: Arndts K, Kegele J, Ritter M, Prazeres da Costa C, Hoerauf A, Winkler AS (2024) Active infection with Onchocerca volvulus and the linkage to epilepsy/nodding syndrome. PLoS Negl Trop Dis 18(5): e0012076. https://doi.org/10.1371/journal.pntd.0012076

Editor: Uwem Friday Ekpo, Federal University of Agriculture Abeokuta, NIGERIA

Received: December 22, 2023; Accepted: March 16, 2024; Published: May 9, 2024

Copyright: © 2024 Arndts et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Funding: JK was funded by the Deutsche Gesellschaft für Epileptologie (DGFE). ASW, CP and AH were co-applicants of the DZIF Verbundprojekt: TI 07.001. In addition, AH and CP are members of DZIF. AH is supported by the BMBF [01KA1611, 01KA2027 and 01KA2113A] and additionally funded by the Deutsche Forschungsgemeinschaft (DFG) under Germany’s Excellence Strategy – EXC2151 – 390873048. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Competing interests: The authors have declared that no competing interests exist.

We appreciate the comment “Onchocerciasis-associated epilepsy and biomarkers” from Robert Colebunders and colleagues published in PLOS NTD 2024 [1] as response to our original publication “Epilepsy and nodding syndrome in association with an Onchocerca volvulus infection drive distinct immune profile patterns”.

In our study [2], we focused on the presence of an active Onchocerca volvulus (OV) infection, determined by polymerase chain reaction (PCR) from skin snips, in the context of epilepsy/nodding syndrome (NS) and not on the immune profiles of onchocerciasis-associated epilepsy (OAE) patients. Therefore, we did not present the OAE criteria. OAE is still a matter of debate since a causal relationship between OV infection and epilepsy/NS has not been established so far. We would like to emphasize that a positive OV16 test result confirming IgG4 antibodies to the recombinant antigen OV-16 [3], which is part of the OAE core criteria, only indicates exposure/previous infection but does not confirm an active infection [4], in contrast to the highly specific and sensitive PCR [5]. Indeed, we observed during our African field studies that most of the individuals, including controls, were antibody-positive in the OV16 test. This was also confirmed by Edridge et al. [6].This is not surprising in a previously OV high-endemic area like Mahenge. Thus, this test cannot not discriminate between individuals who developed epilepsy, NS or remained healthy. Contrary to the application of the OAE criteria [717], studies investigating epilepsy/NS in context of an active OV infection are scarce. It has been shown that immunity and metabolism are strongly modulated in individuals who are actively infected with filarial nematodes (or helminths) in comparison to individuals that have cleared the infection [1820]. To find out more about the possible ways of immunomodulation of OV and its effect on neurological signs/symptoms, we also suggested in our discussion that future studies should take into account the presence of patent and latent infections, which unfortunately was not possible in our study due to the small group sizes.

We fully agree with the suggestion of Colebunders et al. [1] to include an additional control group, namely OV-infected individuals without any epilepsy, in future studies to confirm our findings. Indeed, we clearly mentioned this as a limitation of our study. Most of the individuals with epilepsy/NS were already known and registered patients at the Mahenge Epilepsy Clinic, where they received medication. Nevertheless, we initially had five individuals without epilepsy/NS (caregivers, family members of patients) who were positive for OV determined by PCR from their skin snips. Unfortunately, they did not provide urine and blood samples and were therefore excluded from the study.

The overall conclusion from our publication was that biomarkers might be useful to discriminate epilepsy from NS in individuals with or without an OV infection, which was met with criticism by Colebunders et al. [1]. One of the potential candidates could be N-acetyltyramine-O, β-glucuronide (NATOG), which has been previously described as a potential non-invasive biomarker of active O. volvulus infections and which was significantly elevated in NS patients with OV infection compared to epilepsy patients without OV infection and to controls in our study. We drew our conclusions carefully and did not state that NATOG had discriminative power pointing to further limitations (e.g., small sample size, lacking OV-infected individuals without epilepsy/NS, further studies needed to proof that NATOG might be a potential biomarker) of our study. We are fully aware that the described method of NATOG measurement is not suitable so far for the application in the field due its technical complexity and thus argued that a simplified test version, e.g., a rapid test, could support a diagnosis of active OV infection in the field, particularly in children that are reluctant to undergo invasive methods. Colebunders et al. [1] continued to argue that “While determining NATOG levels may be useful in persons with epilepsy in high OV transmission zones, the NATOG test will have low discriminating power to differentiate between NS and other forms of epilepsy because most other forms of epilepsy in such areas will be OAE.” This statement is incorrect as epilepsy has many different causes, also in endemic areas where the OV infection rate is high.

In summary, our study focused on active OV infection in people with epilepsy/NS and not on people with OAE. We cautiously stated our conclusions regarding potential biomarkers to differentiate people with epilepsy and those with NS with or without an active OV infection and call for further studies. Whether or not OV is the causal agent for epilepsy or NS in OV-endemic areas is not yet conclusively established and may require prospective studies comparing different areas with long-term follow-up of healthy individuals and those who eventually develop epilepsy/NS.

1. Colebunders R HA, Njamnshi AK, Mmbando BP, Kamoen O, Siewe Fodjo JN. Onchocerciasis-associated epilepsy and biomarkers. PLOS Negl Trop Dis. 2024. 2. Arndts K, Kegele J, Massarani AS, Ritter M, Wagner T, Pfarr K, et al. Epilepsy and nodding syndrome in association with an Onchocerca volvulus infection drive distinct immune profile patterns. PLoS Negl Trop Dis. 2023;17(8):e0011503. pmid:37535695 3. Weil GJ, Steel C, Liftis F, Li BW, Mearns G, Lobos E, et al. A rapid-format antibody card test for diagnosis of onchocerciasis. J Infect Dis. 2000;182(6):1796–9. pmid:11069258 4. Unnasch TR, Golden A, Cama V, Cantey PT. Diagnostics for onchocerciasis in the era of elimination. Int Health. 2018;10(suppl_1):i20–i6. pmid:29471336 5. Fischer P, Hoerauf A., Weil G.J. The Filariases. Manson’s Tropical Diseases. 2023;24rd edition. 6. Edridge AWD, Abd-Elfarag G, Deijs M, Broeks MH, Cristella C, Sie B, et al. Parasitic, bacterial, viral, immune-mediated, metabolic and nutritional factors associated with nodding syndrome. Brain Commun. 2023;5(5):fcad223. pmid:37731906 7. Colebunders R, Njamnshi AK, van Oijen M, Mukendi D, Kashama JM, Mandro M, et al. Onchocerciasis-associated epilepsy: From recent epidemiological and clinical findings to policy implications. Epilepsia Open. 2017;2(2):145–52. pmid:29588943 8. Colebunders R, Nelson Siewe FJ, Hotterbeekx A. Onchocerciasis-Associated Epilepsy, an Additional Reason for Strengthening Onchocerciasis Elimination Programs. Trends Parasitol. 2018;34(3):208–16. pmid:29288080 9. Siewe JFN, Ngarka L, Tatah G, Mengnjo MK, Nfor LN, Chokote ES, et al. Clinical presentations of onchocerciasis-associated epilepsy (OAE) in Cameroon. Epilepsy Behav. 2019;90:70–8. pmid:30513438 10. Vinkeles Melchers NVS, Mollenkopf S, Colebunders R, Edlinger M, Coffeng LE, Irani J, et al. Burden of onchocerciasis-associated epilepsy: first estimates and research priorities. Infect Dis Poverty. 2018;7(1):101. pmid:30253788 11. Hotterbeekx A, Raimon S, Abd-Elfarag G, Carter JY, Sebit W, Suliman A, et al. Onchocerca volvulus is not detected in the cerebrospinal fluid of persons with onchocerciasis-associated epilepsy. Int J Infect Dis. 2020;91:119–23. pmid:31786246 12. Hotterbeekx A, Vieri MK, Ramberger M, Jozefzoon-Aghai A, Mandro M, Tepage F, et al. No Evidence for the Involvement of Leiomodin-1 Antibodies in the Pathogenesis of Onchocerciasis-Associated Epilepsy. Pathogens. 2021;10(7). pmid:34357995 13. Vieri MK, Hotterbeekx A, Mandro M, Siewe Fodjo JN, Dusabimana A, Nyisi F, et al. Serotonin Levels in the Serum of Persons with Onchocerciasis-Associated Epilepsy: A Case-Control Study. Pathogens. 2021;10(6). pmid:34201076 14. Vieri MK, Hotterbeekx A, Raimon S, Abd-Elfarag G, Mukendi D, Carter JY, et al. Cytokines and Onchocerciasis-Associated Epilepsy, a Pilot Study and Review of the Literature. Pathogens. 2021;10(3). pmid:33799934 15. Dusabimana A, Siewe Fodjo JN, Ndahura MM, Mmbando BP, Jada SR, Boven A, et al. Surveillance for Onchocerciasis-Associated Epilepsy and OV16 IgG4 Testing of Children 6–10 Years Old Should Be Used to Identify Areas Where Onchocerciasis Elimination Programs Need Strengthening. Pathogens. 2022;11(3). 16. Jada SR, Dusabimana A, Abd-Elfarag G, Okaro S, Brusselaers N, Carter JY, et al. The Prevalence of Onchocerciasis-Associated Epilepsy in Mundri West and East Counties, South Sudan: A Door-to-Door Survey. Pathogens. 2022;11(4). pmid:35456071 17. Bhattacharyya S, Vinkeles Melchers NVS, Siewe Fodjo JN, Vutha A, Coffeng LE, Logora MY, et al. Onchocerciasis-associated epilepsy in Maridi, South Sudan: Modelling and exploring the impact of control measures against river blindness. PLoS Negl Trop Dis. 2023;17(5):e0011320. pmid:37235598 18. Ritter M, Osei-Mensah J, Debrah LB, Kwarteng A, Mubarik Y, Debrah AY, et al. Wuchereria bancrofti-infected individuals harbor distinct IL-10-producing regulatory B and T cell subsets which are affected by anti-filarial treatment. PLoS Negl Trop Dis. 2019;13(5):e0007436. pmid:31120872 19. Arndts K, Specht S, Debrah AY, Tamarozzi F, Klarmann Schulz U, Mand S, et al. Immunoepidemiological profiling of onchocerciasis patients reveals associations with microfilaria loads and ivermectin intake on both individual and community levels. PLoS Negl Trop Dis. 2014;8(2):e2679. pmid:24587458 20. Arndts K, Deininger S, Specht S, Klarmann U, Mand S, Adjobimey T, et al. Elevated Adaptive Immune Responses Are Associated with Latent Infections of Wuchereria bancrofti. PLoS Negl Trop Dis. 2012;6(4):e1611. pmid:22509424

留言 (0)

沒有登入
gif