Skip to content
Licensed Unlicensed Requires Authentication Published by De Gruyter April 15, 2017

Garcinia kola seeds may prevent cognitive and motor dysfunctions in a type 1 diabetes mellitus rat model partly by mitigating neuroinflammation

  • Paul F. Seke Etet EMAIL logo , Mohammed Farahna , Gwiria M.H. Satti , Yahia M. Bushara , Ahmed El-Tahir , Muaawia A. Hamza , Sayed Y. Osman , Ambrose C. Dibia and Lorella Vecchio

Abstract

Background

We reported recently that extracts of seeds of Garcinia kola, a plant with established hypoglycemic properties, prevented the loss of inflammation-sensible neuronal populations like Purkinje cells in a rat model of type 1 diabetes mellitus (T1DM). Here, we assessed G. kola extract ability to prevent the early cognitive and motor dysfunctions observed in this model.

Methods

Rats made diabetic by single injection of streptozotocin were treated daily with either vehicle solution (diabetic control group), insulin, or G. kola extract from the first to the 6th week post-injection. Then, cognitive and motor functions were assessed using holeboard and vertical pole behavioral tests, and animals were sacrificed. Brains were dissected out, cut, and processed for Nissl staining and immunohistochemistry.

Results

Hyperglycemia (209.26 %), body weight loss (–12.37 %), and T1DM-like cognitive and motor dysfunctions revealed behavioral tests in diabetic control animals were not observed in insulin and extract-treated animals. Similar, expressions of inflammation markers tumor necrosis factor (TNF), iba1 (CD68), and Glial fibrillary acidic protein (GFAP), as well as decreases of neuronal density in regions involved in cognitive and motor functions (–49.56 % motor cortex, –33.24 % medial septal nucleus, –41.8 % /–37.34 % cerebellar Purkinje /granular cell layers) were observed in diabetic controls but not in animals treated with insulin or G. kola.

Conclusions

Our results indicate that T1DM-like functional alterations are mediated, at least partly, by neuroinflammation and neuronal loss in this model. The prevention of the development of such alterations by early treatment with G. kola confirms the neuroprotective properties of the plant and warrant further mechanistic studies, considering the potential for human disease.

Acknowledgments

The authors thank the colleagues of their respective institutions for their support and for manuscript proofreading.

  1. Author contributions: All the authors have accepted responsibility for the entire content of this submitted manuscript and approved submission.

  2. Research funding: The present study was supported by King Fahad Medical City (Riyadh, Saudi Arabia) and was performed by collaboration between King Fahad Medical City (Riyadh, Saudi Arabia) and the Department of Basic Health Sciences, Qassim University (Buraydah, Saudi Arabia).

  3. Employment or leadership: None declared.

  4. Honorarium: None declared.

  5. Competing interests: The funding organization(s) played no role in the study design; in the collection, analysis, and interpretation of data; in the writing of the report; or in the decision to submit the report for publication.

References

1. Rodrigues VV, De Castro Ruiz MA, Schamber CR, Bazotte RB. Hypoglycemia induced by insulin as a triggering factor of cognitive deficit in diabetic children. Scientific World Journal. 2014;2014:616534.10.1155/2014/616534Search in Google Scholar PubMed PubMed Central

2. Bialo SR, Agrawal S, Boney CM, Quintos JB. Rare complications of pediatric diabetic ketoacidosis. World J Diabetes. 2015;6:167–174.10.4239/wjd.v6.i1.167Search in Google Scholar PubMed PubMed Central

3. Litchfield AB, Hayes RM, Shuler FD, Flesher SL. Type I diabetes in children and vitamin D. WV Med J. 2015;111:32–37.Search in Google Scholar

4. Ghorbani A, Shafiee-Nick R. Pathological consequences of C-peptide deficiency in insulin-dependent diabetes mellitus. World J Diabetes. 2015;6:145–150.10.4239/wjd.v6.i1.145Search in Google Scholar PubMed PubMed Central

5. Wherrett DK, Chiang JL, Delamater AM, DiMeglio LA, Gitelman SE, Gottlieb PA, et al. Defining pathways for development of disease-modifying therapies in children with type 1 diabetes: A consensus report. Diabetes Care. 2015;38:1975–1985.10.2337/dc15-1429Search in Google Scholar PubMed PubMed Central

6. Bryden KS, Dunger DB, Mayou RA, Peveler RC, Neil HA. Poor prognosis of young adults with type 1 diabetes: A longitudinal study. Diabetes Care. 2003;26:1052–1057.10.2337/diacare.26.4.1052Search in Google Scholar PubMed

7. Raymond J. Updates in behavioural and psychosocial literature in adolescents with type 1 diabetes. Curr Opin Endocrinol Diabetes Obes. 2015;22:265–269.10.1097/MED.0000000000000167Search in Google Scholar PubMed

8. Hagger V, Hendrieckx C, Sturt J, Skinner TC, Speight J. Diabetes distress among adolescents with type 1 diabetes: A systematic review. Curr Diab Rep. 2016;16:9.10.1007/s11892-015-0694-2Search in Google Scholar PubMed

9. Menzin J, Korn JR, Cohen J, Lobo F, Zhang B, Friedman M, et al. Relationship between glycemic control and diabetes-related hospital costs in patients with type 1 or type 2 diabetes mellitus. J Manag Care Pharm. 2010;16:264–275.10.18553/jmcp.2010.16.4.264Search in Google Scholar PubMed

10. Lind M, Svensson AM, Kosiborod M, Gudbjornsdottir S, Pivodic A, Wedel H, et al. Glycemic control and excess mortality in type 1 diabetes. N Engl J Med. 2014;371:1972–1982.10.1056/NEJMoa1408214Search in Google Scholar PubMed

11. Van De Ven KC, Van Der Graaf M, Tack CJ, Heerschap A, De Galan BE. Steady-state brain glucose concentrations during hypoglycemia in healthy humans and patients with type 1 diabetes. Diabetes. 2012;61:1974–1977.10.2337/db11-1778Search in Google Scholar PubMed PubMed Central

12. Mayhan WG, Scott JP, Arrick DM. Influence of type 1 diabetes on basal and agonist-induced permeability of the blood-brain barrier. Physiol Rep. 2015;3(12):pii: e12653 .10.14814/phy2.12653Search in Google Scholar PubMed PubMed Central

13. De Senna PN, Xavier LL, Bagatini PB, Saur L, Galland F, Zanotto C, et al. Physical training improves non-spatial memory, locomotor skills and the blood brain barrier in diabetic rats. Brain Res. 2015;1618:75–82.10.1016/j.brainres.2015.05.026Search in Google Scholar PubMed

14. Duarte JM. Metabolic alterations associated to brain dysfunction in diabetes. Aging Dis. 2015;6:304–321.10.14336/ad.2014.1104Search in Google Scholar

15. Cameron FJ. The impact of diabetes on brain function in childhood and adolescence. Pediatr Clin North Am. 2015;62:911–927.10.1016/j.pcl.2015.04.003Search in Google Scholar PubMed

16. Seaquist ER. The impact of diabetes on cerebral structure and function. Psychosom Med. 2015;77:616–621.10.1097/PSY.0000000000000207Search in Google Scholar PubMed PubMed Central

17. Moheet A, Mangia S, Seaquist ER. Impact of diabetes on cognitive function and brain structure. Ann NY Acad Sci. 2015;1353:60–71.10.1111/nyas.12807Search in Google Scholar PubMed PubMed Central

18. Olsson Y, Save-Soderbergh J, Sourander P, Angervall L. A patho-anatomical study of the central and peripheral nervous system in diabetes of early onset and long duration. Pathol Eur. 1968;3:62–79.Search in Google Scholar

19. Nagayach A, Patro N, Patro I. Experimentally induced diabetes causes glial activation, glutamate toxicity and cellular damage leading to changes in motor function. Front Cell Neurosci. 2014;8:355.10.3389/fncel.2014.00355Search in Google Scholar PubMed PubMed Central

20. Yang S, Xia C, Li S, Du L, Zhang L, Hu Y. Mitochondrial dysfunction driven by the LRRK2-mediated pathway is associated with loss of Purkinje cells and motor coordination deficits in diabetic rat model. Cell Death Dis. 2014;5:1217.10.1038/cddis.2014.184Search in Google Scholar PubMed PubMed Central

21. Farahna M, Seke Etet PF, Osman SY, Yurt KK, Amir N, Vecchio L, et al. Garcinia kola aqueous suspension prevents cerebellar neurodegeneration in long-term diabetic rat - a type 1 diabetes mellitus model. J Ethnopharmacol. 2016;195:159–165 .10.1016/j.jep.2016.11.001Search in Google Scholar PubMed

22. Baluchnejadmojarad T, Kiasalari Z, Afshin-Majd S, Ghasemi Z, Roghani M. S-allyl cysteine ameliorates cognitive deficits in streptozotocin-diabetic rats via suppression of oxidative stress, inflammation, and acetylcholinesterase. Eur J Pharmacol. 2016;794:69–76.10.1016/j.ejphar.2016.11.033Search in Google Scholar PubMed

23. Ahshin-Majd S, Zamani S, Kiamari T, Kiasalari Z, Baluchnejadmojarad T, Roghani M. Carnosine ameliorates cognitive deficits in streptozotocin-induced diabetic rats: Possible involved mechanisms. Peptides. 2016;86:102–111.10.1016/j.peptides.2016.10.008Search in Google Scholar PubMed

24. Ma LY, Lv YL, Huo K, Liu J, Shang SH, Fei YL, et al. Autophagy-lysosome dysfunction is involved in Abeta deposition in STZ-induced diabetic rats. Behav Brain Res. 2016;320:484–493 .10.1016/j.bbr.2016.10.031Search in Google Scholar PubMed

25. Gan MJ, Albanese-O’Neill A, Haller MJ. Type 1 diabetes: Current concepts in epidemiology, pathophysiology, clinical care, and research. Curr Probl Pediatr Adolesc Health Care. 2012;42:269–291.10.1016/j.cppeds.2012.07.002Search in Google Scholar PubMed

26. Kanungo A, Jhingan A, Sahay RK, Muruganathan A, Das AK. Consensus evidence-based guidelines for insulin therapy in patients with type 1 diabetes mellitus as per Indian clinical practice. J Assoc Physicians India. 2014;62:26–33.Search in Google Scholar

27. Ogunkunle AT, Oyelakin TM, Enitan AO, Oyewole FE. A quantitative documentation of the composition of two powdered herbal formulations (antimalarial and haematinic) using ethnomedicinal information from Ogbomoso, Nigeria. Evid Based Complement Alternat Med. 2014;2014:751291.10.1155/2014/751291Search in Google Scholar PubMed PubMed Central

28. Adedara IA, Awogbindin IO, Anamelechi JP, Farombi EO. Garcinia kola seed ameliorates renal, hepatic, and testicular oxidative damage in streptozotocin-induced diabetic rats. Pharm Biol. 2015;53:695–704.10.3109/13880209.2014.937504Search in Google Scholar PubMed

29. Farombi EO, Owoeye O. Antioxidative and chemopreventive properties of Vernonia amygdalina and Garcinia biflavonoid. Int J Environ Res Public Health. 2011;8:2533–2555.10.3390/ijerph8062533Search in Google Scholar PubMed PubMed Central

30. Ayepola OR, Brooks NL, Oguntibeju OO. Kolaviron improved resistance to oxidative stress and inflammation in the blood (erythrocyte, serum, and plasma) of streptozotocin-induced diabetic rats. ScientificWorldJournal. 2014;2014:921080.10.1155/2014/921080Search in Google Scholar PubMed PubMed Central

31. Adaramoye OA. Antidiabetic effect of kolaviron, a biflavonoid complex isolated from Garcinia kola seeds, in Wistar rats. Afr Health Sci. 2012;12:498–506.10.4314/ahs.v12i4.16Search in Google Scholar PubMed PubMed Central

32. Farombi EO, Adedara IA, Oyenihi AB, Ekakitie E, Kehinde S. Hepatic, testicular and spermatozoa antioxidant status in rats chronically treated with Garcinia kolaseed. J Ethnopharmacol. 2013;146:536–542.10.1016/j.jep.2013.01.018Search in Google Scholar PubMed

33. Adedara IA, Farombi EO. Influence of kolaviron and vitamin E on ethylene glycol monoethyl ether-induced haematotoxicity and renal apoptosis in rats. Cell Biochem Funct. 2013;32:31–38.10.1002/cbf.2968Search in Google Scholar PubMed

34. Ijomone OM, Obi AU. Kolaviron, isolated from Garcinia kola, inhibits acetylcholinesterase activities in the hippocampus and striatum of Wistar rats. Ann Neurosci. 2013;20:42–46.10.5214/ans.0972.7531.200203Search in Google Scholar PubMed PubMed Central

35. Abarikwu SO. Kolaviron, a natural flavonoid from the seeds of Garcinia kola, reduces LPS-induced inflammation in macrophages by combined inhibition of IL-6 secretion, and inflammatory transcription factors, ERK1/2, NF-kappaB, p38, Akt, p-c-JUN and JNK. Biochim Biophys Acta. 2014;1840:2373–2381.10.1016/j.bbagen.2014.03.006Search in Google Scholar PubMed

36. Akinmoladun AC, Akinrinola BL, Olaleye MT, Farombi EO. Kolaviron, a Garcinia kola biflavonoid complex, protects against ischemia/reperfusion injury: Pertinent mechanistic insights from biochemical and physical evaluations in rat brain. Neurochem Res. 2015;40:777–787.10.1007/s11064-015-1527-zSearch in Google Scholar PubMed

37. Kaur C, Rathnasamy G, Ling EA. Roles of activated microglia in hypoxia induced neuroinflammation in the developing brain and the retina. J Neuroimmune Pharmacol. 2013;8:66–78.10.1007/s11481-012-9347-2Search in Google Scholar PubMed

38. Jarius S, Wildemann B. ‘Medusa head ataxia’: The expanding spectrum of Purkinje cell antibodies in autoimmune cerebellar ataxia. Part 3: Anti-Yo/CDR2, anti-Nb/AP3B2, PCA-2, anti-Tr/DNER, other antibodies, diagnostic pitfalls, summary and outlook. J Neuroinflammation. 2015;12:168.10.1186/s12974-015-0358-9Search in Google Scholar PubMed PubMed Central

39. Motawi TK, Darwish HA, Hamed MA, El-Rigal NS, Naser AF. A Therapeutic insight of niacin and coenzyme Q10 against diabetic encephalopathy in rats. Mol Neurobiol. 2016;54(3):1601–1611 .10.1007/s12035-016-9765-xSearch in Google Scholar PubMed

40. Sima AA, Zhang W, Li ZG, Kamiya H. The effects of C-peptide on type 1 diabetic polyneuropathies and encephalopathy in the BB/Wor-rat. Exp Diabetes Res. 2008;2008:230458.10.1155/2008/230458Search in Google Scholar PubMed PubMed Central

41. Gandhi GK, Ball KK, Cruz NF, Dienel GA. Hyperglycaemia and diabetes impair gap junctional communication among astrocytes. ASN Neuro. 2010;2:00030.10.1042/AN20090048Search in Google Scholar PubMed PubMed Central

42. Adamec RE, Sayin U, Brown A. The effects of corticotrophin releasing factor (CRF) and handling stress on behavior in the elevated plus-maze test of anxiety. J Psychopharmacol. 1991;5:175–186.10.1177/026988119100500301Search in Google Scholar PubMed

43. Brown GR, Nemes C. The exploratory behaviour of rats in the hole-board apparatus: Is head-dipping a valid measure of neophilia?. Behav Processes. 2008;78:442–448.10.1016/j.beproc.2008.02.019Search in Google Scholar PubMed PubMed Central

44. Paylor R, Nguyen M, Crawley JN, Patrick J, Beaudet A, Orr-Urtreger A. Alpha7 nicotinic receptor subunits are not necessary for hippocampal-dependent learning or sensorimotor gating: A behavioral characterization of Acra7-deficient mice. Learn Mem. 1998;5:302–316.10.1101/lm.5.4.302Search in Google Scholar

45. Bouet V, Freret T, Toutain J, Divoux D, Boulouard M, Schumann-Bard P. Sensorimotor and cognitive deficits after transient middle cerebral artery occlusion in the mouse. Exp Neurol. 2007;203:555–567.10.1016/j.expneurol.2006.09.006Search in Google Scholar

46. Paxinos G, Charles Watson C. The rat brain in stereotaxic coordinates: Hard cover edition, 6th Cambridge, Massachusetts, USA: Academic Press. ISBN: 9780125476126 2006 .Search in Google Scholar

47. Adaramoye OA. Protective effect of kolaviron, a biflavonoid from Garcinia kola seeds, in brain of Wistar albino rats exposed to gamma-radiation. Biol Pharm Bull. 2010;33:260–266.10.1248/bpb.33.260Search in Google Scholar

48. Atiase Y, Farni K, Plange-Rhule J, Luke A, Bovet P, Forrester TG, et al. A comparison of indices of glucose metabolism in five black populations: Data from modeling the epidemiologic transition study (METS). BMC Public Health. 2015;15:895.10.1186/s12889-015-2233-0Search in Google Scholar

49. Ojji DB, Libhaber E, Atherton JJ, Abdullahi B, Nwankwo A, Sliwa K. Risk-factor profile and comorbidities in 2398 patients with newly diagnosed hypertension from the abuja heart study. Medicine (Baltimore). 2015;94:1660.10.1097/MD.0000000000001660Search in Google Scholar

50. Millogo GR, Yameogo C, Samandoulougou A, Yameogo NV, Kologo KJ, Toguyeni JY, et al. Diabetes in urban setting in Ouagadougou, Burkina Faso: Epidemiological profile and level of perception in the adult population. Pan Afr Med J. 2015;20:146.Search in Google Scholar

51. NCD-RisC. Worldwide trends in diabetes since 1980: A pooled analysis of 751 population-based studies with 4.4 million participants. Lancet. 2016;387:1513–1530.10.1016/S0140-6736(16)00618-8Search in Google Scholar

52. Simon P, Dupuis R, Costentin J. Thigmotaxis as an index of anxiety in mice. Influence of dopaminergic transmissions. Behav Brain Res. 1994;61:59–64.10.1016/0166-4328(94)90008-6Search in Google Scholar

53. Lamprea MR, Cardenas FP, Setem J, Morato S. Thigmotactic responses in an open-field. Braz J Med Biol Res. 2008;41:135–140.10.1590/S0100-879X2008000200010Search in Google Scholar PubMed

54. Loseva EV, Sarkisova KY, Loginova NA, Kudrin VS. Depressive behavior and monoamine contents in brain structures of rats during chronic overcrowding. Bull Exp Biol Med. 2015;159:327–330.10.1007/s10517-015-2953-2Search in Google Scholar PubMed

55. Yan W, Kang J, Zhang G, Li S, Kang Y, Wang L, et al. The effects of gonadectomy and binge-like ethanol exposure during adolescence on open field behaviour in adult male rats. Neurosci Lett. 2015;604:52–57.10.1016/j.neulet.2015.07.039Search in Google Scholar PubMed

56. Soultanov V, Fedotova J, Nikitina T, Roschin V, Ordyan N, Hritcu L. Antidepressant-like effect of Ropren® in β-amyloid-(25–35) rat model of Alzheimer’s disease with altered levels of androgens. Mol. Neurobiol. 2016 3 19. . DOI:10.1007/s12035-016-9848-8.Search in Google Scholar PubMed

57. Stefanello N, Schmatz R, Pereira LB, Rubin MA, Da Rocha JB, Facco G, et al. Effects of chlorogenic acid, caffeine, and coffee on behavioral and biochemical parameters of diabetic rats. Mol Cell Biochem. 2014;388:277–286.10.1007/s11010-013-1919-9Search in Google Scholar PubMed

58. Gambeta E, De Souza CP, De Morais H, Zanoveli JM. Reestablishment of the hyperglycemia to the normal levels seems not to be essential to the anxiolytic-like effect induced by insulin. Metab Brain Dis. 2016;31:563–571.10.1007/s11011-015-9770-1Search in Google Scholar PubMed

59. Knight S, Northam E, Donath S, Gardner A, Harkin N, Taplin C, et al. Improvements in cognition, mood and behaviour following commencement of continuous subcutaneous insulin infusion therapy in children with type 1 diabetes mellitus: A pilot study. Diabetologia. 2009;52:193–198.10.1007/s00125-008-1197-3Search in Google Scholar PubMed

60. Phillips AG, Vacca G, Ahn S. A top-down perspective on dopamine, motivation and memory. Pharmacol Biochem Behav. 2008;90:236–249.10.1016/j.pbb.2007.10.014Search in Google Scholar PubMed

61. Merz CJ, Wolf OT, Schweckendiek J, Klucken T, Vaitl D, Stark R. Stress differentially affects fear conditioning in men and women. Psychoneuroendocrinology. 2013;38:2529–2541.10.1016/j.psyneuen.2013.05.015Search in Google Scholar PubMed

62. MacLaren DA, Santini JA, Russell AL, Markovic T, Clark SD. Deficits in motor performance after pedunculopontine lesions in rats–impairment depends on demands of task. Eur J Neurosci. 2014;40:3224–3236.10.1111/ejn.12666Search in Google Scholar PubMed

63. Jimenez-Martin J, Blanco-Lezcano L, Gonzalez-Fraguela ME, Diaz-Hung ML, Serrano-Sanchez T, Almenares JL, et al. Effect of neurotoxic lesion of pedunculopontine nucleus in nigral and striatal redox balance and motor performance in rats. Neuroscience. 2015;289:300–314.10.1016/j.neuroscience.2014.12.056Search in Google Scholar PubMed

64. Frokjaer JB, Brock C, Softeland E, Dimcevski G, Gregersen H, Simren M, et al. Macrostructural brain changes in patients with longstanding type 1 diabetes mellitus - a cortical thickness analysis study. Exp Clin Endocrinol Diabetes. 2013;121:354–360.10.1055/s-0033-1345120Search in Google Scholar PubMed

65. Biessels GJ, Reijmer YD. Brain changes underlying cognitive dysfunction in diabetes: What can we learn from MRI?. Diabetes. 2014;63:2244–2252.10.2337/db14-0348Search in Google Scholar PubMed

66. Toprak H, Yetis H, Alkan A, Filiz M, Kurtcan S, Aralasmak A, et al. Relationships of DTI findings with neurocognitive dysfunction in children with Type 1 diabetes mellitus. Br J Radiol. 2016;89:20150680.10.1259/bjr.20150680Search in Google Scholar PubMed PubMed Central

67. Hideyama T, Aono G, Uesaka Y, Kunimoto M, Nasu M. A 95-year-old female with autopsy-proven cerebral necrosis due to candidiasis who developed stroke-like manifestations. Rinsho Shinkeigaku. 2005;45:230–234.Search in Google Scholar

68. Bolo NR, Musen G, Simonson DC, Nickerson LD, Flores VL, Siracusa T, et al. Functional connectivity of insula, basal ganglia, and prefrontal executive control networks during hypoglycemia in Type 1 diabetes. J Neurosci. 2015;35:11012–11023.10.1523/JNEUROSCI.0319-15.2015Search in Google Scholar PubMed PubMed Central

69. Rooijackers HM, Wiegers EC, Tack CJ, Van Der Graaf M, De Galan BE. Brain glucose metabolism during hypoglycemia in type 1 diabetes: Insights from functional and metabolic neuroimaging studies. Cell Mol Life Sci. 2016;73:705–722.10.1007/s00018-015-2079-8Search in Google Scholar PubMed PubMed Central

70. Yang C, DeVisser A, Martinez JA, Poliakov I, Rosales-Hernandez A, Ayer A, et al. Differential impact of diabetes and hypertension in the brain: Adverse effects in white matter. Neurobiol Dis. 2011;42:446–458.10.1016/j.nbd.2011.02.007Search in Google Scholar PubMed

71. Palomba M, Seke Etet PF, Veronesi C. Effect of inflammatory challenge on hypothalamic neurons expressing orexinergic and melanin-concentrating hormone. Neurosci Lett. 2014;570:47–52.10.1016/j.neulet.2014.03.069Search in Google Scholar PubMed

72. Chen J, Li Y, Tian Y, Huang C, Li D, Zhong Q, et al. Interaction between microbes and host intestinal health: Modulation by dietary nutrients and gut-brain-endocrine-immune axis. Curr Protein Pept Sci. 2015;16:592–603.10.2174/1389203716666150630135720Search in Google Scholar PubMed

73. Miklossy J, McGeer PL. Common mechanisms involved in Alzheimer’s disease and type 2 diabetes: A key role of chronic bacterial infection and inflammation. Aging (Albany NY). 2016;8:575–588.10.18632/aging.100921Search in Google Scholar PubMed PubMed Central

Received: 2016-12-22
Accepted: 2017-2-21
Published Online: 2017-4-15

© 2017 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 6.5.2024 from https://www.degruyter.com/document/doi/10.1515/jcim-2016-0167/html
Scroll to top button