Please use this identifier to cite or link to this item: http://hdl.handle.net/1942/37272
Full metadata record
DC FieldValueLanguage
dc.contributor.advisorRigo, Jean-Michel-
dc.contributor.advisorChabwine, Joëlle Nsimire-
dc.contributor.authorBAGUMA, Marius-
dc.date.accessioned2022-05-06T08:10:17Z-
dc.date.available2022-05-06T08:10:17Z-
dc.date.issued2022-
dc.date.submitted2022-04-12T17:44:49Z-
dc.identifier.urihttp://hdl.handle.net/1942/37272-
dc.description.abstractKonzo is an upper motor neuron disease characterized by a sudden onset of symmetrical, non-progressive, and irreversible spastic paraparesis. This crippling disease mostly affects children from 2 years of age and childbearing women, causing permanent gait difficulties. It occurs only in food-deprived communities exposed to dietary cyanide from insufficiently processed toxic cassava (Manihot esculenta Crantz). Although protein malnutrition and dietary cyanide poisoning are undeniably identified as major risk factors of konzo, the relative importance of each of these two risk factors and the underlying mechanisms of konzo remain largely unknown. In this thesis, using a multi-disciplinary approach, we aimed at a better understanding of konzo risk factors, determinants, and etiopathogenesis. The Democratic Republic of the Congo (DRC) holds the highest prevalence of konzo, with over a half of all reported cases documented in two provinces of the DRC: the former Bandundu Province, in the western part of the country, and South-Kivu province, in the eastern DRC. Strikingly, in South-Kivu, all konzo cases were recorded in Burhinyi and its immediate neighborhoods, while cassava constitutes the staple food for all inhabitants of this province, and cassava-derived cyanide poisoning was documented as a contributive factor to a high prevalence of endemic goiter in Idjwi, another rural area of South-Kivu since the 1970s, with no konzo case reported so far. By comparing data on nutritional status and level of cyanide poisoning of subjects from the two areas of South-Kivu (Burhinyi and Idjwi), our findings showed a similar profile of healthy subjects from Burhinyi and those from Idjwi, with a low prevalence of wasting (3.3% and 6.5%, respectively), intermediate prevalence of stunting (26.7% and 23.9%, respectively) and a high prevalence of dietary cassava-derived cyanide poisoning (50.0% and 63.0%, respectively). As well, konzo patients (from Burhinyi) had a high prevalence cyanide poisoning (81.8%) but, unlike healthy subjects from both areas, had also a high prevalence of both wasting (54.5%) and stunting (72.7%). Thus, a compromised nutritional status was the only factor differentiating konzo patients from healthy individuals exposed to cyanide poisoning. This further suggests that, besides the known role of cyanide poisoning in the pathogenesis of konzo, protein malnutrition may be a key factor for the disease occurrence. In addition, our results showed a seasonal variation in the two above-mentioned konzo risk factors, with acute nutritional deterioration in Burhinyi and higher cyanide intoxication both in Burhinyi and in Idjwi during the dry season. Investigations on local cassava varieties showed high cyanide content in fresh cassava roots both of bitter and sweet taste, but the usual processing methods (maintained during the raining season) proved to be effective enough to reduce cyanide content in cassava roots till safe cyanide levels (< 10 ppm) in obtained flour. In the dry season, however, the cyanide content in cassava flour significantly increased, following a shortening of cassava-processing. Skipping heap-fermentation during cassava processing, as well as a preferential consumption of bitter cassava roots, were the two factors independently associated with very high urinary thiocyanate levels (witnessing a high cyanide poisoning) in participants from Idjwi and Burhinyi during the dry season. Thus, restoring the traditional processing methods (in particular a sufficiently long heap-fermentation) should ensure safe consumption of cassava. Finally, in vitro investigations on the neurotoxic effects of the two major cyanide metabolites (namely cyanate and thiocyanate) confirmed the neurotoxicity of cyanate in two neuron-like cell lines (mouse Neuro-2A neuroblastoma cell line and human SH-SY5Y neuroblastoma cell line) as it induced a drastic drop in cell viability from relatively low concentrations (0.3 mM in Neuro-2A and 3 mM in SH-SY5Y), contrary to thiocyanate which required to reach extremely high concentrations (30 mM), well above physiologically encountered magnitudes, to induce a modest cell death in cell lines. Cyanate neurotoxicity would act simultaneously on several mechanisms, including apoptosis related to caspases (3, 8, and 9), oxidative stress, and excitotoxicity (through AMPA and NMDA receptors activation, probably following a presynaptic release of glutamate). In conclusion, the occurrence of konzo is likely a consequence of the interplay between several factors. An acute worsening of the nutritional status appears to be a key risk factor, favoring an impairment of cyanide detoxification process in subject undergoing high cassava-related cyanide poisoning, leading to an unusual excessive production of minor metabolite (especially cyanate) which, in fine, are responsible for an upper motor neuron damage through various mechanisms (including apoptosis, oxidative stress, and excitotoxicity). Geo-environmental, cultural, and behavioral factors, as well as other factors of individual susceptibility (not yet fully identified) seems to also play a role in the appearance of konzo, although their exact contribution still needs to be elucidated.-
dc.language.isoen-
dc.titleKonzo risk factors, determinants, and etiopathogenesis: Review and new perspectives-
dc.typeTheses and Dissertations-
local.format.pages176-
local.bibliographicCitation.jcatT1-
dc.relation.referencesAdamolekun, B., 2011. Neurological disorders associated with cassava diet: A review of putative etiological mechanisms. Metab. Brain Dis. 26, 79–85. https://doi.org/10.1007/s11011-011-9237-y Adamolekun, B., 2010. Etiology of Konzo, epidemic spastic paraparesis associated with cyanogenic glycosides in cassava: Role of thiamine deficiency? J. Neurol. Sci. 296, 30–33. https://doi.org/10.1016/j.jns.2010.06.016 Ahmed, W., Lingner, J., 2018. Impact of oxidative stress on telomere biology. Differentiation 99, 21–27. https://doi.org/10.1016/j.diff.2017.12.002 Ali Ekangu, R., Kambale Kikandau, J., Vumbi Lelo, G., Kalala Lunganza, R., Yandju Marie-Claire, M., Takaisi Kikuni, P., Okitundu Luwa E-Andjafono, D., Mumba Ngoyi, D., Boivin, J.M., Tshala- Katumbay, D., 2015. Analysis of motor pathway involvement in Konzo disease. J. Neurol. Sci. 357, e496–e497. https://doi.org/10.1016/j.jns.2015.09.287 Allen, T.J., 2010. Konzo in Angola. Cassava Cyanide Dis. Neurolathyrism Netw. 3–4. https://doi.org/10.1371/journal.pntd.0000487 Alter, B.P., Kan, Y.W., Nathan, D.G., 1974. Toxic Effects of High-Dose Cyanate Administration in Rodents. Blood 43, 69–77. https://doi.org/10.1182/blood-2015-06-651331 Anonymous, 2005. Konzo Count. CCDN News 6, 3. Aranguri-llerena, G., Siche, R., 2020. Superior Plants with Significant Amounts of Cyanide and Their Toxicological Implications. Rev. Agric. Sci. 8, 354–366. Baguma, M., Malembaka, E.B., Bahizire, E., Mudumbi, G.Z., Shamamba, D.B., Matabaro, A.-N., Rigo, J.-M., Njamnshi, A.K., Chabwine, J.N., 2021a. Revisiting Konzo Risk Factors in Three Areas Differently Affected by Spastic Paraparesis in Eastern Democratic Republic of the Congo Discloses a Prominent Role of the Nutritional Status—A Comparative Cross-Sectional Study. Nutrients 13. https://doi.org/10.3390/nu13082628 Baguma, M., Nzabara, F., Balemba, G.M., Malembaka, E.B., Migabo, C., Mudumbi, G., Bito, V., Cliff, J., Rigo, J.-M., Chabwine, J.N., 2021b. Konzo risk factors, determinants and etiopathogenesis: what is new? A systematic review. Neurotoxicology 85, 54–67. https://doi.org/10.1016/j.neuro.2021.05.001 Baguma, M., Nzabara, F., Bwenge, E., Mudumbi, G., Maheshe, G., Cliff, J., Rigo, J.M., Chabwine, J.N., 2019. Seasonal variation but not processing methods differently affect cassava-derived cyanide exposure in two areas with different konzo prevalence in south-kivu (D.R. Congo). IBRO Reports 7, 50. https://doi.org/https://doi.org/10.1016/j.ibror.2019.09.099 Banea, J.P., Bikangi, N., Nahimana, G., Nunga, M., Tylleskar, T., Rosling, H., 1992a. High prevalence of konzo associated with a food shortage crisis in the Bandundu region of zaire. Ann. Soc. Belg. Med. Trop. (1920). 72, 295–309. Banea, J.P., Bradbury, J.H., Mandombi, C., Nahimana, D., Denton, I.C., Foster, M.P., Kuwa, N., Tshala Katumbay, D., 2015a. Konzo prevention in six villages in the DRC and the dependence of konzo prevalence on cyanide intake and malnutrition. Toxicol. Reports 2, 609–616. https://doi.org/10.1016/ j.toxrep.2015.03.014 Banea, J.P., Bradbury, J.H., Mandombi, C., Nahimana, D., Denton, I.C., Kuwa, N., Tshala Katumbay, D., 2014. Effectiveness of wetting method for control of konzo and reduction of cyanide poisoning by removal of cyanogens from cassava flour. Food Nutr. Bull. 35, 28–32. Banea, J.P., Bradbury, J.H., Mandombi, C., Nahimana, D., Denton, I.C., Kuwa, N., Tshala Katumbay, D., 2013. Control of konzo by detoxification of cassava flour in three villages in the Democratic Republic of Congo. Food Chem. Toxicol. 60, 506–513. https://doi.org/10.1016/j.fct.2013.08.012 Banea, J.P., Bradbury, J.H., Nahimana, D., Denton, I.C., Foster, M.P., Mekob, N., Kuwa, N., Bokundabi, G., Foley, W.J., 2016. Health factors associated with persistent konzo in four villages in the Democratic Republic of Congo (DRC). African J. Food Sci. 10, 1–6. https://doi.org/10.5897/AJFS2015. Banea, J.P., Bradbury, J.H., Nahimana, D., Denton, I.C., Kuwa, N., 2015b. Survey of the konzo prevalence of village people and their nutrition in Kwilu District, Bandundu Province, DRC. African J. Food Sci. 9, 43–50. https://doi.org/ 10.5897/AJFS2014.1206 Banea, J.P., Poulter, N.H., Rosling, H., 1992b. Shortcuts in cassava processing and risk of dietary cyanide exposure in Zaire. Food Nutr. Bull. 14, 137–143. Banea, J.P., Tylleskar, T., Gitebo, N., Matadi, N., Gebre-Medhin, M., Rosling, H., 1997a. Geographical and seasonal association between linamarin and cyanide exposure from cassava and the upper motor neurone disease konzo in former Zaire. Trop. Med. Int. Health 2, 1143–1151. Banea, J.P., Tylleskär, T., Rosling, H., 1997b. Konzo and Ebola in Bandundu region of Zaire. Lancet 349, 621. https://doi.org/10.1016/S0140-6736(05)61569-3 Bangham, C.R.M., Araujo, A., Yamano, Y., Taylor, G.P., 2015. HTLV-1-associated myelopathy/tropical spastic paraparesis. Nat. Rev. Dis. Prim. 1. https://doi.org/10.1038/nrdp.2015.12 Barclay, D. V., Mauron, J., Blondela, A., Cavadini, C., Verwilghen, A.M., Van Geert, C., Dirren, H., 2003. Micronutrient intake and status in rural Democratic Republic of Congo. Nutr. Res. 23, 659–671. https://doi.org/10.1016/S0271-5317(03)00027-7 Bhattacharya, R., Flora, S.J.S., 2015. Chapter 23 - Cyanide Toxicity and its Treatment, in: Gupta, R.C.B.T.-H. of T. of C.W.A. (Second E. (Ed.), Handbook of Toxicology of Chemical Warfare Agents (Second Edition). Academic Press, Boston, pp. 301–314. https://doi.org/10.1016/B978-0-12-800159-2.00023-3 Blagbrough, I.S., Bayoumi, S.A.L., Rowan, M.G., Beeching, J.R., 2010. Cassava: An appraisal of its phytochemistry and its biotechnological prospects. Phytochemistry 71, 1940–1951. https://doi.org/10.1016/j.phytochem.2010. 09.001 Boivin, M.J., Okitundu, D., Makila-Mabe, B., Sombo, M.T., Mumba, D., Sikorskii, A., Mayambu, B., Tshala-Katumbay, D., 2017. Cognitive and motor performance in Congolese children with konzo during 4 years of follow-up: a longitudinal analysis. Lancet Glob. Heal. 5, e936–e947. https://doi.org/ 10.1016/S2214-109X(17)30267-X Boivin, M.J., Okitundu, D., Makila-Mabe Bumoko, G., Sombo, M.-T., Mumba, D., Tylleskar, T., Page, C.F., Tamfum Muyembe, J.-J., Tshala-Katumbay, D., 2013. Neuropsychological effects of konzo: a neuromotor disease associated with poorly processed cassava. Pediatrics 131, e1231-9. https://doi.org/10.1542/ peds.2012-3011 Bonmarin, I., Nunga, M., Perea, W.A., 2002. Konzo outbreak, in the south-west of the Democratic Republic of Congo, 1996. J. Trop. Pediatr. 48, 234–238. Bradbury, J.H., Cliff, J., Denton, I.C., 2011. Uptake of wetting method in Africa to reduce cyanide poisoning and konzo from cassava. Food Chem. Toxicol. 49, 539–542. https://doi.org/10.1016/j.fct.2010.04.049 Bradbury, J.H., Denton, I.C., 2014. Mild method for removal of cyanogens from cassava leaves with retention of vitamins and protein. Food Chem. 158, 417–420. https://doi.org/10.1016/j.foodchem.2014.02.132 Bradbury, M.G., Egan, S. V, Bradbury, J.H., 1999. Picrate paper kits for determination of total cyanogens in cassava roots and all forms of cyanogens in cassava products. J. Sci. Food Agric. 79, 593–601. https://doi.org/ 10.1002/(SICI)1097-0010(19990315)79:4<593::AID-JSFA222>3.0.CO;2-2 Bramble, M.S., Vashist, N., Ko, A., Priya, S., Musasa, C., Mathieu, A., Spencer, D.A., Lupamba Kasendue, M., Mamona Dilufwasayo, P., Karume, K., Nsibu, J., Manya, H., Uy, M.N.A., Colwell, B., Boivin, M., Mayambu, J.P.B., Okitundu, D., Droit, A., Mumba Ngoyi, D., Blekhman, R., Tshala-Katumbay, D., Vilain, E., 2021. The gut microbiome in konzo. Nat. Commun. 12, 5371. https://doi.org/ 10.1038/s41467-021-25694-1 Brosnan, J.T., Brosnan, M.E., 2006. The Sulfur-Containing Amino Acids: An Overview. J. Nutr. 136, 1636S-1640S. https://doi.org/10.1093/jn/ 136.6.1636S Bumoko, G.M., Sombo, M.T., Okitundu, L.D., Mumba, D.N., Kazadi, K.T., Tamfum-Muyembe, J.J., Lasarev, M.R., Boivin, M.J., Banea, J.P., Tshala-Katumbay, D.D., 2014. Determinants of cognitive performance in children relying on cyanogenic cassava as staple food. Metab. Brain Dis. 29, 359–366. https://doi.org/10.1007/s11011-014-9492-9 Bumoko, G.M.M., Sadiki, N.H., Rwatambuga, A., Kayembe, K.P., Okitundu, D.L., Mumba Ngoyi, D., Muyembe, J.T.J.T., Banea, J.P., Boivin, M.J., Tshala-katumbay, D., Ngoyi, D.M., Muyembe, J.T.J.T., Banea, J.P., Boivin, M.J., Tshala-katumbay, D., 2015. Lower serum levels of selenium, copper, and zinc are related to neuromotor impairments in children with konzo. J. Neurol. Sci. 349, 149–153. https://doi.org/10.1016/j.jns.2015.01.007 Burke, T.G., Mutnick, A.H., 1994. Treatment of Cyanide and Thiocyanate Toxicity Secondary to Sodium Nitroprusside. J. Pharm. Technol. 10, 207–209. Cardoso, A.P., Ernesto, M., Nicala, D., Mirione, E., Chavane, L., N’zwalo, H., Chikumba, S., Cliff, J., Mabota, A.P., Haque, M.R., Bradbury, J.H., 2004. Combination of cassava flour cyanide and urinary thiocyanate measurements of school children in Mozambique. Int. J. Food Sci. Nutr. 55, 183–190. https://doi.org/10.1080/09637480410001725265 Cardoso, A.P., Mirione, E., Ernesto, M., Massaza, F., Cliff, J., Haque, M.R., Bradbury, J.H., 2005. Processing of cassava roots to remove cyanogens 18, 451–460. https://doi.org/10.1016/j.jfca.2004.04.002 Carlsson, L., Mlingi, N., Juma, A., Ronquist, G., Rosling, H., 1999. Metabolic fates in humans of linamarin in cassava flour ingested as stiff porridge. Food Chem. Toxicol. 37, 307–312. https://doi.org/10.1016/S0278-6915(99)00015-0 Carter, S.E., Jones, P.G., 1993. A model of the distribution of cassava in Africa. Appl. Geogr. 13, 353–371. https://doi.org/10.1016/0143-6228(93)90037-2 Carton, H., Kayembe, K., Kabeya, Odio, Billiau, A., Maertens, K., 1986. Epidemic spastic paraparesis in Bandundu (Zaire). J. Neurol. Neurosurg. Psychiatry 49, 620–627. https://doi.org/10.1136/jnnp.49.6.620 Casadei, E., Cliff, J., Neves, J., 1990. Surveillance of urinary thiocyanate concentration after epidemic spastic paraparesis in Mozambique. J. Trop. Med. Hyg. 93, 257–261. Casseb, J., de Oliveira, A.C.P., Vergara, M.P.P., Montanheiro, P., Bonasser, F., Meilman Ferreira, C., Smid, J., Duarte, A.J. da S., 2008. Presence of tropical spastic paraparesis/human T-cell lymphotropic virus type 1-associated myelopathy (TSP/HAM)-like among HIV-1-infected patients. J. Med. Virol. 80, 392–398. https://doi.org/10.1002/jmv.21111 Ceballos, H., Iglesias, C.A., Pérez, J.C., Dixon, A.G.O., 2004. Cassava breeding: opportunities and challenges. Plant Mol. Biol. 56, 503–516. https://doi.org/ 10.1007/s11103-004-5010-5 Chabwine, J., Masheka, C., Balol’ebwami, Z., Maheshe, B., Balegamire, S., Rutega, B., Wa Lola, M., Mutendela, K., Bonnet, M.-J., Shangalume, O., Balegamire, J.M., Nemery, B., 2011. Appearance of konzo in South-Kivu, a wartorn area in the Democratic Republic of Congo. Food Chem. Toxicol. 49, 644–649. https://doi.org/10.1016/j.fct.2010.07.050 Chandler, J.D., Min, E., Huang, J., Nichols, D.P., Day, B.J., 2013. Nebulized thiocyanate improves lung infection. Br. J. Pharmacol. 169, 1166–1177. https://doi.org/10.1111/bph.12206 Chandra, A.K., 2010. Goitrogen in Food: Cyanogenic and Flavonoids Containing Plant Foods in the Development of Goiter, in: Watson, R.R., Preedy, V.R.B.T.-B.F. in P.H. (Eds.), Bioactive Foods in Promoting Health. Academic Press, San Diego, pp. 691–716. https://doi.org/https://doi.org/10.1016/B978-0-12-374628-3.00042-6 Charlier, C., Gougnard, T., Lamiable, D., Levillain, P., Plomteux, G., 2000. Cyanures et thiocyanates en toxicologie hospitalière. Ann. Toxicol. Anal. 12, 131–136. https://doi.org/10.1051/ata/2000019 Chauhan, A., Srivastva, N., Bubber, P., 2018. Thiamine Deficiency Induced Dietary Disparity Promotes Oxidative Stress and Neurodegeneration. Indian J. Clin. Biochem. 33, 422–428. https://doi.org/10.1007/s12291-017-0690-1 Chen, D., Chen, H.W., 2013. Using the Köppen classification to quantify climate variation and change: An example for 1901-2010. Environ. Dev. 6, 69–79. https://doi.org/10.1016/j.envdev.2013.03.007 Chuma, G.B., Mondo, J.M., Ndeko, A.B., Mugumaarhahama, Y., Bagula, E.M., Blaise, M., Valérie, M., Jacques, K., Karume, K., Mushagalusa, G.N., 2021. Forest cover affects gully expansion at the tropical watershed scale: Case study of Luzinzi in Eastern DR Congo. Trees, For. People 4, 100083. https://doi.org/ 10.1016/j.tfp.2021.100083 Ciglenečki, I., Eyema, R., Kabanda, C., Taafo, F., Mekaoui, H., Urbaniak, V., 2011. Konzo outbreak among refugees from Central African Republic in Eastern region, Cameroon. Food Chem. Toxicol. 49, 579–582. https://doi.org/ 10.1016/j.fct.2010.05.081 Cliff, J., Lundqvist, P., Martensson, J., Rosling, H., Sorbo, B., 1985. Association of high cyanide and low sulphur intake in cassava-induced spastic paraparesis. Lancet (London, England) 2, 1211–1213. Cliff, J., Muquingue, H., Nhassico, D., Nzwalo, H., Bradbury, J.H., 2011. Konzo and continuing cyanide intoxication from cassava in Mozambique. Food Chem. Toxicol. 49, 631–635. https://doi.org/10.1016/j.fct.2010.06.056 Cliff, J., Nicala, D., Saute, F., Givragy, R., Azambuja, G., Taela, A., Chavane, L., Gani, A., 1999. Ankle Clonus and Thiocyanate, Linamarin, and Inorganic Sulphate Excretion in School Children in Communities with Konzo, Mozambique. J. Trop. Pediatr. 45, 139–142. Cliff, J., Nicala, D., Saute, F., Givragy, R., Azambuja, G., Taela, A., Chavane, L., Howarth, J., 1997. Konzo associated with war in Mozambique. Trop. Med. Int. Health 2, 1068–1074. https://doi.org/10.1046/j.1365-3156.1997.d01-178.x Cluskey, S., Ramsden, D.B., 2001. Mechanisms of neurodegeneration in amyotrophic lateral sclerosis. J. Clin. Pathol. - Mol. Pathol. 54, 386–392. https://doi.org/10.1136/mp.54.6.386 Coentrão, L., Moura, D., 2011. Acute cyanide poisoning among jewelry and textile industry workers. Am. J. Emerg. Med. 29, 78–81. https://doi.org/10.1016/ j.ajem.2009.09.014 Coghlan, B., Brennan, R.J., Ngoy, P., Dofara, D., Otto, B., Clements, M., Stewart, T., 2006. Mortality in the Democratic Republic of Congo: a nationwide survey. Lancet 367, 44–51. https://doi.org/10.1016/S0140-6736(06)67923-3 Cole, D.E., Evrovski, J., 2000. The clinical chemistry of inorganic sulfate. Crit. Rev. Clin. Lab. Sci. 37, 299–344. https://doi.org/10.1080/10408360091174231 Congo Research Group, 2019. Congo, Forgotten: The Numbers Behind Africa’s Longest Humanitarian Crisis. Cumbana, A., Mirione, E., Cliff, J., Bradbury, J.H., 2007. Reduction of cyanide content of cassava flour in Mozambique by the wetting method. Food Chem. 101, 894–897. https://doi.org/10.1016/j.foodchem.2006.02.062 De-The, G., Giordano, C., Gassain, A., Howlett, W., Sonan, T., Akani, F., Rosling, H., Carton, H., Mouanga, Y., Caudie, C., Stenger, F., Malone, G., 1989. Human retroviruses HTLV-I, HIV-1, and HIV-2 and neurological diseases in some equatorial areas of Africa. J. Acquir. Immune Defic. Syndr. 2, 550-556. De Moura, F.F., Moursi, M., Lubowa, A., Ha, B., Boy, E., Oguntona, B., Sanusi, R.A., Maziya-Dixon, B., 2015. Cassava Intake and Vitamin A Status among Women and Preschool Children in Akwa-Ibom, Nigeria. PLoS One 10, e0129436. https://doi.org/10.1371/journal.pone.0129436 de Onis, M., Onyango, A.W., Borghi, E., Siyam, A., Nishida, C., Siekmann, J., 2007. Development of a WHO growth reference for school-aged children and adolescents. Bull. World Health Organ. 85, 660–667. https://doi.org/10.2471/blt.07.043497 Delange, F., Ermans, A.M., Vis, H.L., Stanbury, J.B., 1972. Endemic cretinism in Idjwi Island (Kivu Lake, Republic of the Congo). J. Clin. Endocrinol. Metab. 34, 1059–1066. https://doi.org/10.1210/jcem-34-6-1059 Diasolua Ngudi, D., 2005. Konzo and cassava toxicity: a study of associated nutritional factors in the Popokabaka District, Democratic Republic of Congo. Ph D. thesis. Universiteit Gent, Belgium. Diasolua Ngudi, D., Banea-Mayambu, J.P., Lambein, F., Kolsteren, P., 2011. Konzo and dietary pattern in cassava-consuming populations of Popokabaka, Democratic Republic of Congo. Food Chem. Toxicol. 49, 613–619. https://doi.org/10.1016/j.fct.2010.06.053 Donaghy, M., 1999. Classification and clinical features of motor neurone diseases and motor neuropathies in adults. J. Neurol. 246, 331–333. https://doi.org/ 10.1007/s004150050358 Egekeze, J.O., Oehme, F.W., 1980. Cyanides and their toxicity: a literature review. Tijdschr. Diergeneeskd. 105. https://doi.org/10.1080/ 01652176.1980.9693766 Ermans, A.M., Delange, F., Van der Velden, M., Kinthaert, J., 1973. Goitrogenic action of cyanogenic glucosides present in cassava: a possible etiologic factor of endemic goiter in the Idjwi island. Acta Endocrinol. Suppl. (Copenh). 179, 31. Ernesto, M., Cardoso, A.P., Nicala, D., Mirione, E., Massaza, F., Cliff, J., Haque, M.R., Bradbury, J.H., 2002. Persistent konzo and cyanogen toxicity from cassava in northern Mozambique. Acta Trop. 82, 357–362. https://doi.org/ 10.1016/S0001-706X(02)00042-6 Essers, A.J.A., Alsen, P., Rosling, H., 1992. Insufficient processing of cassava induced acute intoxications and the paralytic disease konzo in a rural area of mozambique. Ecol. Food Nutr. 27, 17–27. https://doi.org/10.1080/ 03670244.1992.9991222 Essers, A.J.A., Van der Grift, R.M., Voragen, A.G.J., 1996. Cyanogen removal from cassava roots during. Food Chem. 55, 319–325. FAO, 2019. FAOSTAT. URL http://www.fao.org/faostat/en/#data/QC (accessed 12.1.20). Flahaut, M., 2015. Le cyanure dans l’histoire et intoxications actuelles. Médecine humaine et pathologie. Université de Bordeaux 2. UFR des Sciences Médicales. 109. Food and Agriculture Organization, 2013. Save and Grow : Cassava - A guide to sustainable production intensification. Roma. Food and Agriculture Organization of the United Nations, 2017. FAOSTAT. URL http://www.fao.org/faostat/en/#data (accessed 11.22.19). Fulda, S., Debatin, K.M., 2006. Extrinsic versus intrinsic apoptosis pathways in anticancer chemotherapy. Oncogene 25, 4798–4811. https://doi.org/10.1038/ sj.onc.1209608 German, W.F., Messinger, E., Herman, M., 1949. Toxicity of thiocyanates used in treatment of hypertension. Ann. Intern. Med. 30, 1054–1059. https://doi.org/ 10.7326/0003-4819-30-5-1054 Gessain, A., Mahieux, R., 2012. Tropical spastic paraparesis and HTLV-1 associated myelopathy: Clinical, epidemiological, virological and therapeutic aspects. Rev. Neurol. (Paris). 168, 257–269. https://doi.org/10.1016/j.neurol. 2011.12.006 Getahun, H., Lambein, F., Vanhoorne, M., Van der Stuyft, P., 2002. Pattern and associated factors of the neurolathyrism epidemic in Ethiopia. Trop. Med. Int. Heal. 7, 118–124. https://doi.org/10.1046/j.1365-3156.2002.00836.x Ginting, E., Widodo, Y., 2013. Cyanide Reduction in Cassava Root Products Through Processing and Selection of Cultivars in Relation to Food Safety. Bul. Palawija 25, 25–36. https://doi.org/10.21082/bulpalawija.v0n25. 2013.p Gratton, S.M., Lam, B.L., 2014. Visual loss and optic nerve head swelling in thiamine deficiency without prolonged dietary deficiency. Clin. Ophthalmol. 8, 1021–1024. https://doi.org/10.2147/OPTH.S64228 Gyamfi, O.A., Bortey-Sam, N., Mahon, S.B., Brenner, M., Rockwood, G.A., Logue, B.A., 2019. Metabolism of Cyanide by Glutathione To Produce the Novel Cyanide Metabolite 2-Aminothiazoline-4-oxoaminoethanoic Acid. Chem. Res. Toxicol. 32, 718–726. https://doi.org/10.1021/acs.chemrestox.8b00384 Hall, L., Guo, C., Tandy, S., Broadhouse, K., Dona, A.C., Malle, E., Bartels, E.D., Christoffersen, C., Grieve, S.M., Figtree, G., Hawkins, C.L., Davies, M.J., 2021. Oral pre-treatment with thiocyanate (SCN−) protects against myocardial ischaemia–reperfusion injury in rats. Sci. Rep. 11, 12712. https://doi.org/ 10.1038/s41598-021-92142-x Haque, M.R., Bradbury, J.H., 1999. Simple method for determination of thiocyanate in urine. Clin. Chem. 45, 1459–1464. Harvey-Leeson, S., Karakochuk, C.D., Hawes, M., Tugirimana, P.L., Bahizire, E., Akilimali, P.Z., Michaux, K.D., Lynd, L.D., Whitfield, K.C., Moursi, M., Boy, E., Foley, J., McLean, J., Houghton, L.A., Gibson, R.S., Green, T.J., 2016. Anemia and Micronutrient Status of Women of Childbearing Age and Children 6-59 Months in the Democratic Republic of the Congo. Nutrients 8, 98. https://doi.org/10.3390/nu8020098 Hawkinson, J.E., Espitia, S.A., 1997. Effects of thiocyanate and AMPA receptor ligands on (S) -5-fluorowillardiine, (S) -AMPA and (R,S) -AMPA binding. Eur. J. Pharmacol. 329, 213–221. https://doi.org/10.1016/S0014-2999(97)89182-0 Heath, P.R., Shaw, P.J., 2002. Update on the glutamatergic neurotransmitter system and the role of excitotoxicity in amyotrophic lateral sclerosis. Muscle Nerve 26, 438–458. https://doi.org/10.1002/mus.10186 Hollenberg, S.M., 2007. Vasodilators in acute heart failure. Heart Fail. Rev. 12, 143–147. https://doi.org/10.1007/s10741-007-9017-2 Howlett, W.P., Brubaker, G., Mlingi, N., Rosling, H., 1992. A geographical cluster of konzo in Tanzania. J. Trop. Geogr. Neurol. 2, 102–108. Howlett, W.P., Brubaker, G.R., Mlingi, N., Rosling, H., 1990. Konzo, an epidemic upper motor neuron disease studied in Tanzania. Brain 113 ( Pt 1, 223–235. Hu, L., Tian, K., Zhang, T., Fan, C.-H., Zhou, P., Zeng, D., Zhao, S., Li, L.-S., Smith, H.S., Li, J., Ran, J.-H., 2019. Cyanate Induces Oxidative Stress Injury and Abnormal Lipid Metabolism in Liver through Nrf2/HO-1. Molecules 24. https://doi.org/10.3390/molecules24183231 Imakumbili, M.L.E., Semu, E., Semoka, J.M.R., Abass, A., Mkamilo, G., 2019. Farmers’ perceptions on the causes of cassava root bitterness: A case of konzo-affected mtwara region, Tanzania. PLoS One 14, 1–14. https://doi.org/ 10.1371/journal.pone.0215527 Ismaila, A.R., Alakali, J.S., Atume, T.G., 2018. Effect of Local Processing Techniques on the Nutrients and Anti-Nutrients Content of Bitter Cassava (Manihot Esculenta Crantz). Am. J. Food Sci. Technol. 6, 92–97. Ivory, S.J., McGlue, M.M., Peterman, C., Baldwin, P., Lucas, J., Cohen, A., Russell, J., Saroni, J., Msaky, E., Kimirei, I., Soreghan, M., 2021. Climate, vegetation, and weathering across space and time in Lake Tanganyika (tropical eastern Africa). Quat. Sci. Adv. 3, 100023. https://doi.org/10.1016/j.qsa.2021. 100023 John Arnfield, A., 2020. Köppen climate classification. Encycl. Br. URL https://www.britannica.com/science/Koppen-climate-classification%0A (accessed 11.22.20). Johnson, J.M., Fox, V., 2018. Beyond Thiamine: Treatment for Cognitive Impairment in Korsakoff’s Syndrome. Psychosomatics 59, 311–317. https://doi.org/10.1016/j.psym.2018.03.011 Joint FAO/WHO Food Standards Programme. Codex Alimentarius Commission, 2009. Codex Committee On Contaminants In Foods. Third Session. Discussion Paper On Cyanogenic Glycosides. Kambale, K.J., Ali, E.R., Sadiki, N.H., Kayembe, K.P., Mvumbi, L.G., Yandju, D.L., Boivin, M.J., Boss, G.R., Stadler, D.D., Lambert, W.E., Lasarev, M.R., Okitundu, L.A., Mumba Ngoyi, D., Banea, J.P., Tshala-Katumbay, D.D., 2017. Lower sulfurtransferase detoxification rates of cyanide in konzo—A tropical spastic paralysis linked to cassava cyanogenic poisoning. Neurotoxicology 59, 256–262. https://doi.org/10.1016/j.neuro.2016.05.016 Kannan, K., Jain, S.K., 2000. Oxidative stress and apoptosis. Pathophysiology 7, 153–163. https://doi.org/10.1016/s0928-4680(00)00053-5 Kashala-Abotnes, E., Okitundu, D., Mumba, D., Boivin, M.J., Tylleskär, T., Tshala-Katumbay, D., 2018. Konzo: a distinct neurological disease associated with food (cassava) cyanogenic poisoning. Brain Res. Bull. 0–1. https://doi.org/10.1016/j.brainresbull.2018.07.001 Kasonde, J.M., 2015. Ministerial Statement On Konzo - Presented to parliament by the Minister of Health. Kassa, R.M., Kasensa, N.L., Monterroso, V.H., Kayton, R.J., Klimek, J.E., David, L.L., Lunganza, K.R., Kayembe, K.T., Bentivoglio, M., Juliano, S.L., Tshala-Katumbay, D.D., 2011. On the biomarkers and mechanisms of konzo, a distinct upper motor neuron disease associated with food (cassava) cyanogenic exposure. Food Chem. Toxicol. 49, 571–578. https://doi.org/10.1016/ j.fct.2010.05.080 Kassubek, J., Ludolph, A.C., Muller, H.P., 2012. Neuroimaging of motor neuron diseases. Ther. Adv. Neurol. Disord. 5, 119–127. https://doi.org/10.1177/ 1756285612437562 Kimani, S., Moterroso, V., Lasarev, M., Kipruto, S., Bukachi, F., Maitai, C., David, L., Tshala-Katumbay, D., 2013. Carbamoylation correlates of cyanate neuropathy and cyanide poisoning: Relevance to the biomarkers of cassava cyanogenesis and motor system toxicity. Springerplus 2, 1–8. https://doi.org/ 10.1186/2193-1801-2-647 Kimani, S., Moterroso, V., Morales, P., Wagner, J., Kipruto, S., Bukachi, F., Maitai, C., Tshala-Katumbay, D., 2014a. Cross-species and tissue variations in cyanide detoxification rates in rodents and non-human primates on protein-restricted diet. Food Chem. Toxicol. 66, 203–209. https://doi.org/10.1016/ j.fct.2014.01.047 Kimani, S., Sinei, K., Bukachi, F., Tshala-Katumbay, D., Maitai, C., 2014b. Memory deficits associated with sublethal cyanide poisoning relative to cyanate toxicity in rodents. Metab. Brain Dis. 29, 105–112. https://doi.org/10.1007/ s11011-013-9459-2 Kimani, S.T., 2011. Neurotoxicity of cassava cyanogens in rodents and non-human primates. University of Nairobi. Kuvibidila, S., Vuvu, M., 2009. Unusual low plasma levels of zinc in non-pregnant Congolese women. Br. J. Nutr. 101, 1783–1786. https://doi.org/10.1017/ S0007114508147390 Lambein, F., Travella, S., Kuo, Y.H., Van Montagu, M., Heijde, M., 2019. Grass pea (Lathyrus sativus L.): orphan crop, nutraceutical or just plain food? Planta 250, 821–838. https://doi.org/10.1007/s00425-018-03084-0 Lantum, H., 1998. Spastic paraparesis konzo in the Garoua Boulai Health District, East Province - Cameroon: a hidden endemic disease. Monograph 85. Lavigne, J., Roy, L., Lefebvre, L.F., 2004. Section B - 1 : Les cyanures, in: Guide Toxicologique Pour Les Urgences En Santé Environnementale. p. 25. LePage, K.T., Dickey, R.W., Gerwick, W.H., Jester, E.L., Murray, T.F., 2005. On the use of neuro-2a neuroblastoma cells versus intact neurons in primary culture for neurotoxicity studies. Crit. Rev. Neurobiol. 17, 27–50. https://doi.org/10.1615/critrevneurobiol.v17.i1.20 Lim, S.-W., Loh, H.-S., Ting, K.-N., Bradshaw, T.D., Allaudin, Z.N., 2015. Reduction of MTT to Purple Formazan by Vitamin E Isomers in the Absence of Cells. Trop. life Sci. Res. 26, 111–120. Liu, D., Ke, Z., Luo, J., 2017. Thiamine Deficiency and Neurodegeneration: the Interplay Among Oxidative Stress, Endoplasmic Reticulum Stress, and Autophagy. Mol. Neurobiol. 54, 5440–5448. https://doi.org/10.1007/s12035-016-0079-9 Lucasse, C., 1952. Le Kitondji : une paralysie spastique. Ann Soc Belge Med Trop 33, 391–401. Ludolph, A.C., Spencer, P.S., 1996. Toxic models of upper motor neuron disease. J. Neurol. Sci. 139 Suppl, 53–59. Lundquist, P., Gedal, B.K., Nilsson, L., 1995a. An Improved Method for Determination of Thiocyanate in Plasma and Urine 1 ). Eur. J. Clin. Chem. Clin. Biochem. 33, 343–349. https://doi.org/10.1515/cclm.1995.33.6.343 Lundquist, P., Kågedal, B., Nilsson, L., Rosling, H., 1995b. Analysis of the cyanide metabolite 2-aminothiazoline-4-carboxylic acid in urine by high-performance liquid chromatography. Anal. Biochem. https://doi.org/10.1006/ abio.1995.1310 Madiyal, A., Ajila, V., Babu, S.G., Hegde, S., Kumari, S., Madi, M., Achalli, S., Alva, P., Ullal, H., 2018. Status of thiocyanate levels in the serum and saliva of non-smokers, ex-smokers and smokers. Afr. Health Sci. 18, 727–736. https://doi.org/10.4314/ahs.v18i3.31 Maiorka, P.C., Go, S.L., 2002. Neuropathologic study of long term cyanide administration to goats 40, 1693–1698. Makene, W.J., Wilson, J., 1972. Biochemical studies in Tanzanian patients with ataxic tropical neuropathy. J. Neurol. Neurosurg. Psychiatry 35, 31–33. Malouff, J.M., Schutte, N.S., 2017. A meta-analysis of the relationship between anxiety and telomere length. Anxiety, Stress Coping 30, 264–272. https://doi.org/10.1080/10615806.2016.1261286 Martel, J.L., Kerndt, C.C., Franklin, D.S., 2020. Vitamin B1 (Thiamine), in: StatPearls. Treasure Island (FL). Mbanjo, E.G.N., Rabbi, I.Y., Ferguson, M.E., Kayondo, S.I., Eng, N.H., Tripathi, L., Kulakow, P., Egesi, C., 2021. Technological Innovations for Improving Cassava Production in Sub-Saharan Africa. Front. Genet. 11, 623736. https://doi.org/10.3389/fgene.2020.623736 Mbelesso, P., Yogo, M.-L., Yangatimbi, E., Paul-Senekian, V. de, Nali, N.M., Preux, P.-M., 2009. Outbreak of konzo disease in health region No. 2 of the Central African Republic. Rev. Neurol. (Paris). 165, 466–470. https://doi.org/10.1016/ j.neurol.2008.10.020 Mcmahon, J.M., White, W.L.B., Say, R.T., 1995. Cyanogenesis in cassava ( Manihot esculenta Crantz ). J. Exp. Bot. 46, 731–741. https://doi.org/10.1093/ jxb/46.7.731 Ministry of health Mozambique, 1984. Mantakassa: an epidemic of spastic paraparesis associated with chronic cyanide intoxication in a cassava staple area of Mozambique. 1. Epidemiology and clinical and laboratory findings in patients. Ministry of Health, Mozambique. Bull. World Health Organ. 62, 477–484. Mlingi, N., Kimatta, S., Rosling, H., 1991. Konzo, a paralytic disease observed in southern Tanzania. Trop Doct 21, 24–25. https://doi.org/10.1177/ 004947559102100110 Mlingi, N., Nkya, S., Tatala, S., Rashid, S., Bradbury, H., 2011. Recurrence of konzo in southern Tanzania: rehabilitation and prevention using the wetting method. Food Chem. Toxicol. 49, 673–677. https://doi.org/10.1016/ j.fct.2010.09.017 Mlingi, N., Poulter, N., Rosling, H., 1992. An outbreak of acute intoxications from consumption of insufficiently processed cassava in Tanzania. Nutr. Res. 12, 677–687. Mlingi, N. V., Assey, V.D., Swai, A.B.M., Mclarty, D.G., Karlen, H., Rosling, H., 1993. Determinants of cyanide exposure from cassava in a konzo-affected population in northern Tanzania. Int. J. Food Sci. Nutr. 44, 137–144. https://doi.org/10.3109/09637489309017432 Mlingi, N.L. V, Bainbridge, Z.A., Poulter, N.H., Rosling, H., 1995. Critical stages in cyanogen removal during cassava processing in southern Tanzania. Food Chem. 53, 29–33. https://doi.org/10.1016/0308-8146(95)95782-2 Moher, D., Liberati, A., Tetzlaff, J., Altman, D.G., 2009. Preferred reporting items for systematic reviews and meta- analyses: the PRISMA statement. Ann. Intern. Med. 151, 264–269. Montagnac, J.A., Davis, C.R., Tanumihardjo, S.A., 2009a. Nutritional value of cassava for use as a staple food and recent advances for improvement. Compr. Rev. Food Sci. Food Saf. 8, 181–194. https://doi.org/10.1111/j.1541-4337. 2009.00077.x Montagnac, J.A., Davis, C.R., Tanumihardjo, S.A., 2009b. Processing techniques to reduce toxicity and antinutrients of Cassava for use as a staple food. Compr. Rev. Food Sci. Food Saf. 8, 17–27. https://doi.org/10.1111/j.1541-4337.2008. 00064.x Morgan, P.E., Laura, R.P., Maki, R.A., Reynolds, W.F., Davies, M.J., 2015. Thiocyanate supplementation decreases atherosclerotic plaque in mice expressing human myeloperoxidase. Free Radic. Res. 49, 743–749. https://doi.org/10.3109/10715762.2015.1019347 Murphy, D.E., Snowhill, E.W., Williams, M., 1987. Characterization of quisqualate recognition sites in rat brain tissue using Dl-[3H]α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) and a filtration assay. Neurochem. Res. 12, 775–781. https://doi.org/10.1007/BF00971514 Mwanza, J.-C., Lysebo, D.E., Kayembe, D.L., Tshala-Katumbay, D., Nyamabo, L.K., Tylleskar, T., Plant, G.T., 2003. Visual evoked potentials in konzo, a spastic paraparesis of acute onset in Africa. Ophthalmol. J. Int. d’ophtalmologie. Int. J. Ophthalmol. 217, 381–386. https://doi.org/10.1159/ 000073066. Nambisan, B., 2011. Strategies for elimination of cyanogens from cassava for reducing toxicity and improving food safety. Food Chem. Toxicol. 49, 690–693. https://doi.org/10.1016/j.fct.2010.10.035 Nambisan, B., 1994. Evaluation of the Effect of Various Processing Techniques on Cyanogen Content Reduction in Cassava. Acta Hortic. https://doi.org/ 10.17660/ActaHortic.1994.375.17 Nambisan, B., Sundaresan, S., 1985. Effect of processing on the cyanoglucoside content of cassava. J. Sci. Food Agric. 36, 1197–1203. https://doi.org/ 10.1002/jsfa.2740361126 National Research Council (US) Committee on Acute Exposure Guideline Levels; National Research Council (US) Committee on Toxicology, 2009. Acute Exposure Guideline Levels for Selected Airborne Chemicals: Volume 7. Nedoboy, P.E., Morgan, P.E., Mocatta, T.J., Richards, A.M., Winterbourn, C.C., Davies, M.J., 2014. High plasma thiocyanate levels are associated with enhanced myeloperoxidase-induced thiol oxidation and long-term survival in subjects following a fi rst myocardial infarction. Free Radic. Res. 48, 1256–1266. https://doi.org/10.3109/10715762.2014.947286 Ngudi, D.D., Kuo, Y.-H., Lambein, F., 2003. Cassava cyanogens and free amino acids in raw and cooked leaves. Food Chem. Toxicol. 41, 1193–1197. https://doi.org/10.1016/S0278-6915(03)00111-X Ngudi, D.D., Kuo, Y.-H., Van Montagu, M., Lambein, F., 2012. Research on motor neuron diseases konzo and neurolathyrism: trends from 1990 to 2010. PLoS Negl. Trop. Dis. 6, e1759. https://doi.org/10.1371/journal.pntd.0001759 Ngudi, D.D., Kuo, Y.H., Lambein, F., 2002. Food safety and amino acid balance in processed cassava “Cossettes.” J. Agric. Food Chem. 50, 3042–3049. https://doi.org/10.1021/jf011441k Nhassico, D., Bradbury, J.H., Cliff, J., Majonda, R., Cuambe, C., Denton, I.C., Foster, M.P., Martins, A., Cumbane, A., Sitoe, L., Pedro, J., Muquingue, H., 2016. Use of the wetting method on cassava flour in three konzo villages in Mozambique reduces cyanide intake and may prevent konzo in future droughts. Food Sci. Nutr. 4, 555–561. https://doi.org/10.1002/fsn3.317 Nimni, M.E., Han, B., Cordoba, F., 2007. Are we getting enough sulfur in our diet? Nutr. Metab. 4, 24. https://doi.org/10.1186/1743-7075-4-24 Njankouo, Y., Mounjouenpou, P., Kansci, G., Josiane, M., Priscile, M., Meguia, F., Natacha, N.S., Eyenga, N., Mikhaïl, M., Nyegue, A., 2019. Influence of cultivars and processing methods on the cyanide contents of cassava ( Manihot esculenta Crantz ) and its traditional food products. Sci. African 5, e00119. https://doi.org/10.1016/j.sciaf.2019.e00119 Nunn, P.B., Lyddiard, J.R.A., Christopher Perera, K.P.W., 2011. Brain glutathione as a target for aetiological factors in neurolathyrism and konzo. Food Chem. Toxicol. 49, 662–667. https://doi.org/10.1016/j.fct.2010.08.037 Nzwalo, H., Cliff, J., 2011. Konzo: from poverty, cassava, and cyanogen intake to toxico-nutritional neurological disease. PLoS Negl. Trop. Dis. 5, e1051. https://doi.org/10.1371/journal.pntd.0001051 Okitundu, Luwa E-Andjofono Daniel, Ayanne, M.-T.S., Makila-Mabe, G.B., Banea, J.-P.M., Ngoy, D.M., Boivin, M., Tamfum-Muyembe, J.-J., Tshala-Katumbay, D., 2018. Konzo global neurological index: a clinical marker of susceptibility and severity of neurocognitive deficits in children living in konzo-affected areas. African J. Neurol. Sci. 37, 51–62. Okitundu, Luwa E-Andjafono Daniel, Ayanne, M.T.S.S., Makila-Mabe, G.B., Mayambu, J.P.B., Ngoyi, D.M., Boivin, M., Tamfum-Muyembe, J.J., Tshala-Katumbay, D., 2018. Socioemotional disorders in children living in Konzo-affected areas, an epidemic paralytic disease associated with cyanide poisoning from food in sub-Saharan Africa. Pan Afr. Med. J. 31, 1–11. https://doi.org/ 10.11604/pamj.2018.31.118.11640 Okitundu, L.E.-A.D., Bumoko, M.-M.G., Sombo, M.T.S.A., Kambale, J.K., Mashukano, N., Kazadi, K.T., Mumba, N.D., Boivin, M.J., Tamfum-Muyembe, J.-J., Banea Mayambu, J.-P., Tshala-Katumbay, D., 2014. Persistence of konzo epidemics in Kahemba, Democratic Republic of Congo. phenomenological and socio-economic aspects. Pan Afr. Med. J. 18, 213. https://doi.org/10.11604/ pamj.2014.18.213.4572 Oluwole, O.S.A., 2015. Cyclical konzo epidemics and climate variability. Ann. Neurol. 77, 371–380. https://doi.org/10.1002/ana.24334 Oluwole, O.S.A., Oludiran, A., 2013a. Normative concentrations of urine thiocyanate in cassava eating communities in Nigeria. Int. J. Food Sci. Nutr. 64, 1036–1041. https://doi.org/10.3109/09637486.2013.825697 Oluwole, O.S.A., Oludiran, A., 2013b. Geospatial association of endemicity of ataxic polyneuropathy and highly cyanogenic cassava cultivars. Int. J. Health Geogr. 12, 41. https://doi.org/10.1186/1476-072X-12-41 Osuntokun, B.O., Durowoju, J.E., McFarlane, H., Wilson, J., 1968. Plasma Amino-acids in the Nigerian Nutritional Ataxic Neuropathy. Br. Med. J. 3, 647–649. https://doi.org/10.1136/bmj.3.5619.647 Padmaja, G., 1995. Cyanide Detoxification in Cassava for Food and Feed Uses. Crit. Rev. Food Sci. Nutr. 35, 299–339. https://doi.org/10.1080/ 10408399509527703 Peterson, C.M., Tsairis, P., Onishi, A., Lu, Y.S., Grady, R., 1974. Sodium cyanate induced polyneuropathy in patients with sickle-cell disease. Ann. Intern. Med. 81, 152–158. https://doi.org/10.7326/0003-4819-81-2-152 Pimenta, E., Calhoun, D.A., Oparil, S., 2010. Hypertensive Emergencies, Second Edi. ed, Cardiac Intensive Care. Elsevier Inc. https://doi.org/10.1016/B978-1-4160-3773-6.10028-X Posada-Vergara, M.P., Montanheiro, P., Fukumori, L.M.I., Bonasser, F., Duarte, A.J.D.S., Penalva De Oliveira, A.C., Casseb, J., 2006. Clinical and epidemiological aspects of HTLV-II infection in São Paulo, Brazil: Presence of tropical spastic paraparesis/HTLV-associated myelopathy (TSP/HAM) simile diagnosis in HIV-1-co-infected subjects. Rev. Inst. Med. Trop. Sao Paulo 48, 207–210. Pourhassan, M., Angersbach, B., Lueg, G., Klimek, C.N., Wirth, R., 2019. Blood Thiamine Level and Cognitive Function in Older Hospitalized Patients. J. Geriatr. Psychiatry Neurol. 32, 90–96. https://doi.org/10.1177/ 0891988718819862 Rivadeneyra-Domínguez, E., Rodríguez-Landa, J.F., 2020. Preclinical and clinical research on the toxic and neurological effects of cassava (Manihot esculenta Crantz) consumption. Metab. Brain Dis. 35, 65–74. https://doi.org/10.1007/ s11011-019-00522-0 Rivadeneyra-Dominguez, E., Rodriguez-Landa, J.F., Article, O., 2016. Motor impairments induced by microinjection of linamarin in the dorsal hippocampus of Wistar rats. Neurologia 31, 516–522. https://doi.org/10.1016/ j.nrl.2014.10.018 Rivadeneyra-Domínguez, E., Vázquez-Luna, a., Díaz-Sobac, R., Eduardo Briones-Céspedes, E., Rodríguez-Landa, J.F., 2015. Contribution of hippocampal area CA1 to acetone cyanohydrin-induced loss of motor coordination in rats. Neurologia 32, 1–6. https://doi.org/10.1016/j.nrl.2015.11.010 Rivadeneyra-Domínguez, E., Vázquez-Luna, A., Rodríguez-Landa, J.F., Díaz-Sobac, R., 2013. Neurotoxic effect of linamarin in rats associated with cassava (Manihot esculenta Crantz) consumption. Food Chem. Toxicol. 59, 230–235. https://doi.org/10.1016/j.fct.2013.06.004 Rocha, J.A., Reis, C., Simões, F., Fonseca, J., Mendes Ribeiro, J., 2005. Diagnostic investigation and multidisciplinary management in motor neuron disease. J. Neurol. 252, 1435–1447. https://doi.org/10.1007/s00415-005-0007-9 Rosling, H., Gessain, A., de Thé, G., Ebondo, N., Banea, M., Bikangi, N., Kinjanja, K., Nunga, K., 1988. Tropical and epidemic spastic paraparesis are different. Lancet MAY 28, 1222–1223. Ross, S.M., Roy, D.N., Spencer, P.S., 1989. β‐N‐Oxalylamino‐L‐Alanine Action on Glutamate Receptors. J. Neurochem. 53, 710–715. https://doi.org/10.1111/ j.1471-4159.1989.tb11762.x Sanders, J.L., Newman, A.B., 2013. Telomere length in epidemiology: A biomarker of aging, age-related disease, both, or neither? Epidemiol. Rev. 35, 112–131. https://doi.org/10.1093/epirev/mxs008 Santisopasri, V., Kurotjanawong, K., Chotineeranat, S., Piyachomkwan, K., Sriroth, K., Oates, C.G., 2001. Impact of water stress on yield and quality of cassava starch. Ind. Crops Prod. 13, 115–129. https://doi.org/10.1016/ S0926-6690(00)00058-3 Sawalha, K., Gonzalez-Toledo, E., Hussein, O., 2019. Role of Magnetic Resonance Imaging in Diagnosis of Motor Neuron Disease: Literature Review and Two Case Illustrations. Perm. J. 23, 1–7. https://doi.org/10.7812/TPP/18-131 Sayre, R., Beeching, J.R., Cahoon, E.B., Egesi, C., Fauquet, C., Fellman, J., Fregene, M., Gruissem, W., Mallowa, S., Manary, M., Maziya-Dixon, B., Mbanaso, A., Schachtman, D.P., Siritunga, D., Taylor, N., Vanderschuren, H., Zhang, P., 2011. The BioCassava plus program: biofortification of cassava for sub-Saharan Africa. Annu. Rev. Plant Biol. 62, 251–272. https://doi.org/ 10.1146/annurev-arplant-042110-103751 Schulz, V., 1984. Clinical Pharmacokinetics of Nitroprusside, Cyanide, Thiosulphate and Thiocyanate. Clin. Pharmacokinet. 9, 239–251. https://doi.org/10.2165/00003088-198409030-00005 Shaw, C., Papayannopoulou, T., Stamatoyannopoulos, G., 1974. Neuropathology of Cyanate Toxicity in Rhesus Monkeys 176, 166–176. Shaw, P.J., 2005. Molecular and cellular pathways of neurodegeneration in motor neurone disease. J. Neurol. Neurosurg. Psychiatry 76, 1046–1057. https://doi.org/10.1136/jnnp.2004.048652 Shi, Y., 2002. Mechanisms of caspase activation and inhibition during apoptosis. Mol. Cell 9, 459–470. https://doi.org/10.1016/S1097-2765(02)00482-3 Siddiqi, O.K., Kapina, M., Kumar, R., Ngomah Moraes, A., Kabwe, P., Mazaba, M.L., Hachaambwa, L., Ng’uni, N.M., Chikoti, P.C., Morel-Espinosa, M., Jarrett, J.M., Baggett, H.C., Chizema-Kawesha, E., 2020. Konzo outbreak in the Western Province of Zambia. Neurology 94, E1495–E1501. https://doi.org/ 10.1212/WNL.0000000000009017 Smith, R.P., 1973. Cyanate and Thiocyanate : Acute Toxicity. Exp. Biol. Med. 142, 1041–1044. https://doi.org/10.3181/00379727-142-37171 Sokołowska, M., Lorenc-Koci, E., Bilska, A., Iciek, M., 2013. The effect of lipoic acid on cyanate toxicity in different structures of the rat brain. Neurotox. Res. 24, 345–357. https://doi.org/10.1007/s12640-013-9395-2 Soler-martín, C., Riera, J., Seoane, A., Cutillas, B., Ambrosio, S., Boadas-vaello, P., Llorens, J., 2010. The targets of acetone cyanohydrin neurotoxicity in the rat are not the ones expected in an animal model of konzo ☆. Neurotoxicol. Teratol. 32, 289–294. https://doi.org/10.1016/j.ntt.2009.11.001 Spencer, P.S., 1999. Food toxins, ampa receptors, and motor neuron diseases. Drug Metab. Rev. 31, 561–587. https://doi.org/10.1081/DMR-100101936 Spencer, P.S., Palmer, V.S., 2012. Interrelationships of undernutrition and neurotoxicity: Food for thought and research attention. Neurotoxicology 33, 605–616. https://doi.org/10.1016/j.neuro.2012.02.015 Spencer, P.S., Roy, D.N., Ludolph, A., Hugon, J., Dwivedi, M.P., Schaumburg, H.H., 1986. Lathyrism: evidence for role of the neuroexcitatory aminoacid BOAA. Lancet 2, 1066–1067. https://doi.org/10.1016/s0140-6736(86)90468-x Sreeja, V.G., Nagahara, N., Li, Q., Minami, M., 2003. New aspects in pathogenesis of konzo: neural cell damage directly caused by linamarin contained in cassava (Manihot esculenta Crantz). Br. J. Nutr. 90, 467–472. https://doi.org/10.1079/ BJN2003902 Srinivasula, S.M., Ahmad, M., Fernandes-Alnemri, T., Alnemri, E.S., 1998. Autoactivation of procaspase-9 by Apaf-1-mediated oligomerization. Mol. Cell 1, 949–957. https://doi.org/10.1016/S1097-2765(00)80095-7 Stanke, C., Kerac, M., Prudhomme, C., Medlock, J., Murray, V., 2013. Health Effects of Drought: A Systematic Review of the Evidence. PLoS Curr. 1–36. https://doi.org/10.1371/currents.dis.7a2cee9e980f91ad7697b570bcc4b004 Statland, J.M., Barohn, R.J., Mcvey, A.L., Katz, J., Dimachkie, M.M., City, K., Pacific, C., Francisco, S., 2016. Patterns of Weakness, Classification of Motor Neuron Disease, and Clinical Diagnosis of Sporadic Amyotrophic Lateral Sclerosis 33, 735–748. https://doi.org/10.1016/j.ncl.2015.07.006.Patterns Swenne, I., Eriksson, U.J., Christoffersson, R., Kagedal, B., Lundquist, P., Nilsson, L., Tylleskar, T., Rosling, H., 1996. Cyanide detoxification in rats exposed to acetonitrile and fed a low protein diet. Fundam. Appl. Toxicol. 32, 66–71. Szabo, E.A., Jansson, E., Miles, D., Hambridge, T., Stanley, G., Baines, J., Brent, P., 2010. Responding to Incidents of Low Level Chemical Contamination in Food, First Edit. ed, Ensuring Global Food Safety. Elsevier Inc. https://doi.org/10.1016/B978-0-12-374845-4.00024-2 Tan, S.L., 1995. Factors affecting cyanide content in cassava (Manihot esculenta Crantz). J. Trop. Agric. Food Sci. 23, 121–131. Teles, F.F.F., 2002. Chronic poisoning by hydrogen cyanide in cassava and its prevention in Africa and Latin America. Food Nutr. Bull. 23, 407–412. Tor-Agbidye, J, Palmer, V.S., Lasarev, M.R., Craig, A.M., Blythe, L.L., Sabri, M.I., Spencer, P.S., 1999. Bioactivation of cyanide to cyanate in sulfur amino acid deficiency: relevance to neurological disease in humans subsisting on cassava. Toxicol. Sci. 50, 228–235. Tor-Agbidye, J., Palmer, V.S., Sabri, M.I., Craig, A.M., Blythe, L.L., Spencer, P.S., 1998. Dietary deficiency of cystine and methionine in rats alters thiol homeostasis required for cyanide detoxification. J. Toxicol. Environ. Heal. - Part A 55, 583–595. https://doi.org/10.1080/009841098158269 Tor-Agbidye, John, Palmer, V.S., Spencer, P.S., Craig, A.M., Blythe, L.L., Sabri, M.I., 1999. Sodium cyanate alters glutathione homeostasis in rodent brain: Relationship to neurodegenerative diseases in protein-deficient malnourished populations in Africa. Brain Res. 820, 12–19. https://doi.org/10.1016/S0006-8993(98)01343-2 Tshala-Katumbay, D, Edebol Eeg-Olofsson, K., Kazadi-Kayembe, T., Fällmar, P., Tylleskär, T., Kayembe-Kalula, T., 2002. Abnormalities of somatosensory evoked potentials in konzo--an upper motor neuron disorder. Clin. Neurophysiol. 113, 10–15. Tshala-Katumbay, Desire, Eeg-Olofsson, K.E., Kazadi-Kayembe, T., Tylleskär, T., Fällmar, P., 2002. Analysis of motor pathway involvement in konzo using transcranial electrical and magnetic stimulation. Muscle and Nerve 25, 230–235. https://doi.org/10.1002/mus.10029 Tshala-Katumbay, D., Eeg-Olofsson, K.E., Tylleskar, T., Kazadi-Kayembe, T., 2001. Impairments, disabilities and handicap pattern in konzo--a non-progressive spastic para/tetraparesis of acute onset. Disabil. Rehabil. 23, 731–736. Tshala-Katumbay, D., Lukusa, V.M., Eeg-Olofsson, K.E., 2000. EEG Findings in Konzo: A Spastic Para/Tetraparesis of Acute Onset. Clin. EEG Neurosci. 31, 196–200. https://doi.org/10.1177/155005940003100408 Tshala-katumbay, D.D., Ngombe, N.N., Okitundu, D., David, L., Westaway, S.K., Boivin, M.J., Mumba, N.D., Banea, J., 2016. Cyanide and the human brain : perspectives from a model of food ( cassava ) poisoning 1378, 50–57. https://doi.org/10.1111/nyas.13159 Tshala-katumbay, D.D., Spencer, P.S., 2007. Toxic disorders of the upper motor neuron system, in: Handbook of Clinical Neurology. pp. 361–370. Tylleskar, T., 1994. The Causation of Konzo. Studies on a Paralytic Disease in Africa. Uppsala University. Tylleskar, T., Banea, M., Bikangi, N., Fresco, L., Persson, L.A., Rosling, H., 1991. Epidemiological evidence from Zaire for a dietary etiology of konzo, an upper motor neuron disease. Bull. World Health Organ. 69, 581–589. Tylleskar, T., Banea, M., Bikangi, N., Nahimana, G., Persson, L.Å., Rosling, H., 1995. Dietary determinants of a non-progressive spastic paraparesis (konzo): A case-referent study in a high incidence area of Zaire. Int. J. Epidemiol. 24, 949–956. https://doi.org/10.1093/ije/24.5.949 Tylleskar, T., Banea, M., Bottiger, B., Thorstensson, R., Biberfeld, G., Rosling, H., 1996. Konzo, an epidemic spastic paraparesis in Africa, is not associated with antibodies to HTLV-I, HIV, or HIV gag-encoded proteins. J. Acquir. Immune Defic. Syndr. Hum. Retrovirology 12, 317–318. Tylleskar, T., Howlett, W.P., Rwiza, H.T., Aquilonius, S.M., Stalberg, E., Linden, B., Mandahl, A., Larsen, H.C., Brubaker, G.R., Rosling, H., Tylleskär, T., Howlett, W.P., Rwiza, H.T., Aquilonius, S.M., Stâlberg, E., Linden, B., Mandahi, A., Larsen, H.C., Brubaker, G.R., Rosling, H., 1993. Konzo: a distinct disease entity with selective upper motor neuron damage. J. Neurol. Neurosurg. Psychiatry 56, 638–643. https://doi.org/10.1136/jnnp.56.6.638 Tylleskar, T., Legue, F.D., Kpizingui, E., 1994. Konzo in the Central African Republic. Neurology 44, 959–961. Tylleskar, T., Rosling, H., Banea, M., Bikangi, N., Cooke, R.D., Poulter, N.H., 1992. Cassava cyanogens and konzo, an upper motoneuron disease found in Africa. Lancet 339, 208–211. https://doi.org/10.1016/0140-6736(92)90006-O UCL – LTAP, n.d. Thiocyanates: urine. Louvain Cent. Toxicol. Appl. Pharmacol. URL https://www.toxi.ucl.ac.be/documents/thiocyanate_u.htm (accessed 9.29.18). Umoh, I.B., Ogunkoya, F.O., Maduagwu, E.N., Oke, O.L., 1985. Effect of thiamin status on the metabolism of linamarin in rats. Ann. Nutr. Metab. 29, 319–324. Van Damme, P., Braeken, D., Callewaert, G., Robberecht, W., Van Den Bosch, L., 2005a. GluR2 deficiency accelerates motor neuron degeneration in a mouse model of amyotrophic lateral sclerosis. J Neuropathol Exp Neurol 64, 605–612. Van Damme, P., Dewil, M., Robberecht, W., Van Den Bosch, L., 2005b. Excitotoxicity and amyotrophic lateral sclerosis. Neurodegener. Dis. 2, 147–159. https://doi.org/10.1159/000089620 Van Moorhem, Marijke, Decrock, E., De Vuyst, E., De Bock, M., Wang, N., Lambein, F., Van Den Bosch, L., Leybaert, L., 2011. L-β-N-oxalyl-α,β-diaminopropionic acid toxicity in motor neurons. Neuroreport 22, 131–135. https://doi.org/10.1097/WNR.0b013e3283433027 Van Moorhem, M., Lambein, F., Leybaert, L., 2011. Unraveling the mechanism of β-N-oxalyl-α,β-diaminopropionic acid (β-ODAP) induced excitotoxicity and oxidative stress, relevance for neurolathyrism prevention. Food Chem. Toxicol. 49, 550–555. https://doi.org/10.1016/j.fct.2010.03.054 Veldink, J.H., Van den Berg, L.H., Wokke, J.H.J., 2004. The future of motor neuron disease: the challenge is in the genes. J. Neurol. 251, 491–500. https://doi.org/10.1007/s00415-004-0322-6 Wachsmann, P., Lamprecht, A., 2012. Polymeric nanoparticles for the selective therapy of inflammatory bowel disease. Methods Enzymol. 508, 377–397. https://doi.org/10.1016/B978-0-12-391860-4.00019-7 Wang, X., Sundquist, K., Hedelius, A., Palmér, K., Memon, A.A., Sundquist, J., 2017. Leukocyte telomere length and depression, anxiety and stress and adjustment disorders in primary health care patients. BMC Psychiatry 17, 1–10. https://doi.org/10.1186/s12888-017-1308-0 Wang, Y., Qin, Z.H., 2010. Molecular and cellular mechanisms of excitotoxic neuronal death. Apoptosis 15, 1382–1402. https://doi.org/10.1007/s10495-010-0481-0 Weyns, Y., Hoex, L., Matthysen, K., 2016. Analysis of the interactive map of artisanal mining areas in eastern DR Congo: 2015 update. Antwerp. Woldeamanuel, Y.W., Hassan, A., Zenebe, G., 2012. Neurolathyrism: two Ethiopian case reports and review of the literature. J. Neurol. 259, 1263–1268. https://doi.org/10.1007/s00415-011-6306-4 World Health Organization, 2010. First WHO report on neglected tropical diseases: working to overcome the global impact of neglected tropical diseases, World Health Organization. https://doi.org/10.1177/1757913912449575 World Health Organization, 2007. Growth reference data for 5-19 years (WHO 2007). World Health Organization. URL https://www.who.int/growthref/en/ (accessed 1.15.19). World Health Organization, 1996. Konzo - a distinct type of upper motoneuron disease. Wkly. Epidemiol. Rec. 71, 225–228. WorldData.info, 2021. Climate of South Kivu (Democratic Republic of the Congo). URL https://www.worlddata.info/africa/congo-kinshasa/climate-south-kivu. php (accessed 11.10.21). Xicoy, H., Wieringa, B., Martens, G.J.M., 2017. The SH-SY5Y cell line in Parkinson’s disease research: a systematic review. Mol. Neurodegener. 12, 10. https://doi.org/10.1186/s13024-017-0149-0 Yang, X., Le Corronc, H., Legendre, P., Triller, A., Specht, C.G., 2021. Differential regulation of glycinergic and GABAergic nanocolumns at mixed inhibitory synapses. EMBO Rep. 22, e52154. https://doi.org/10.15252/embr.202052154 Zacarias, C.H., Esteban, C., Rodrigues, G.L., Nascimento, E. de S., 2017. Occupational exposure to hydrogen cyanide during large-scale cassava processing, in Alagoas State, Brazil. Cad. Saude Publica 33, e00073416. https://doi.org/10.1590/0102-311X00073416 Zaninovic, V., 1999. On the etiology of tropical spastic paraparesis and human T-cell lymphotropic virus I - Associated myelopathy. Int. J. Infect. Dis. 3, 168–177. Zietzer, A., Niepmann, S.T., Camara, B., Lenart, M.A., Jansen, F., Becher, M.U., Andrié, R., Nickenig, G., Tiyerili, V., 2019. Sodium thiocyanate treatment attenuates atherosclerotic plaque formation and improves endothelial regeneration in mice. PLoS One 14, e0214476.-
local.type.refereedRefereed-
local.type.specifiedPhd thesis-
local.uhasselt.internationalno-
item.fulltextWith Fulltext-
item.contributorBAGUMA, Marius-
item.fullcitationBAGUMA, Marius (2022) Konzo risk factors, determinants, and etiopathogenesis: Review and new perspectives.-
item.accessRightsEmbargoed Access-
item.embargoEndDate2027-04-27-
Appears in Collections:Research publications
Files in This Item:
File Description SizeFormat 
Konzo risk factors, determinants, and etiopathogenesis: Review and new perspectives
  Until 2027-04-27
3.93 MBAdobe PDFView/Open    Request a copy
Show simple item record

Page view(s)

112
checked on Sep 6, 2022

Download(s)

32
checked on Sep 6, 2022

Google ScholarTM

Check


Items in DSpace are protected by copyright, with all rights reserved, unless otherwise indicated.