The Cholinergic Drug Galantamine Alleviates Oxidative Stress Alongside Anti-inflammatory and Cardio-Metabolic Effects in Subjects With the Metabolic Syndrome in a Randomized Trial
Journal: 2021/March - Frontiers in Immunology
Abstract:
Background: The metabolic syndrome (MetS) is an obesity-associated disorder of pandemic proportions and limited treatment options. Oxidative stress, low-grade inflammation and altered neural autonomic regulation, are important components and drivers of pathogenesis. Galantamine, an acetylcholinesterase inhibitor and a cholinergic drug that is clinically-approved (for Alzheimer's disease) has been implicated in neural cholinergic regulation of inflammation in several conditions characterized with immune and metabolic derangements. Here we examined the effects of galantamine on oxidative stress in parallel with inflammatory and cardio-metabolic parameters in subjects with MetS. Trial Design and Methods: The effects of galantamine treatment, 8 mg daily for 4 weeks or placebo, followed by 16 mg daily for 8 weeks or placebo were studied in randomly assigned subjects with MetS (n = 22 per group) of both genders. Oxidative stress, including superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase activities, lipid and protein peroxidation, and nitrite levels were analyzed before and at the end of the treatment. In addition, plasma cytokine and adipokine levels, insulin resistance (HOMA-IR) and other relevant cardio-metabolic indices were analyzed. Autonomic regulation was also examined by heart rate variability (HRV) before treatment, and at every 4 weeks of treatment. Results: Galantamine treatment significantly increased antioxidant enzyme activities, including SOD [+1.65 USOD/mg protein, [95% CI 0.39-2.92], P = 0.004] and CAT [+0.93 nmol/mg, [95% CI 0.34-1.51], P = 0.01], decreased lipid peroxidation [thiobarbituric acid reactive substances [log scale 0.72 pmol/mg, [95% CI 0.46-1.07], P = 0.05], and systemic nitrite levels [log scale 0.83 μmol/mg protein, [95% CI 0.57-1.20], P = 0.04] compared with placebo. In addition, galantamine significantly alleviated the inflammatory state and insulin resistance, and decreased the low frequency/high frequency ratio of HRV, following 8 and 12 weeks of drug treatment. Conclusion: Low-dose galantamine alleviates oxidative stress, alongside beneficial anti-inflammatory, and metabolic effects, and modulates neural autonomic regulation in subjects with MetS. These findings are of considerable interest for further studies with the cholinergic drug galantamine to ameliorate MetS.
Keywords: autonomic modulation; cholinergic; galantamine; heart rate variability; inflammation; metabolic syndrome; oxidative stress.
Relations:
Content
Citations
(2)
References
(68)
Diseases
(3)
Conditions
(1)
Drugs
(1)
Chemicals
(7)
Processes
(2)
Similar articles
Articles by the same authors
Discussion board

The Cholinergic Drug Galantamine Alleviates Oxidative Stress Alongside Anti-inflammatory and Cardio-Metabolic Effects in Subjects With the Metabolic Syndrome in a Randomized Trial

+6 authors
Click here for additional data file.(103K, TIF)
Hypertension Unit, University of São Paulo (USP), São Paulo, Brazil
Postgraduate Program in Health Science, Midwestern State University (UNICENTRO), Paraná, Brazil
Nursing Department Graduate Program in Nanosciences and Biosciences, Nove de Julho University (UNINOVE), São Paulo, Brazil
Department of Physiology, Federal University of São Paulo (UNIFESP), São Paulo, Brazil
Biomedical Sciences Institute Department of Physiology and Biophysics, University of São Paulo (USP), São Paulo, Brazil
Laboratory of Immunobiology, Department of Medicine, Center for Bioelectronic Medicine, Karolinska Institutet, Stockholm, Sweden
The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
Edited by: Niccolo Terrando, Duke University, United States
Reviewed by: Robert A. Whittington, Columbia University, United States; Bruno Bonaz, Center Hospitalier Universitaire de Grenoble, France; Annette Bruchfeld, Karolinska Institutet (KI), Sweden
*Correspondence: Valentin A. Pavlov ude.llewhtron@volvapv
Fernanda Marciano Consolim Colombo rb.evoninu@milosnoc.adnanref
This article was submitted to Inflammation, a section of the journal Frontiers in Immunology
†These authors have contributed equally to this work
Edited by: Niccolo Terrando, Duke University, United States
Reviewed by: Robert A. Whittington, Columbia University, United States; Bruno Bonaz, Center Hospitalier Universitaire de Grenoble, France; Annette Bruchfeld, Karolinska Institutet (KI), Sweden
Received 2020 Oct 28; Accepted 2021 Feb 8.
This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

Abstract

Background: The metabolic syndrome (MetS) is an obesity-associated disorder of pandemic proportions and limited treatment options. Oxidative stress, low-grade inflammation and altered neural autonomic regulation, are important components and drivers of pathogenesis. Galantamine, an acetylcholinesterase inhibitor and a cholinergic drug that is clinically-approved (for Alzheimer's disease) has been implicated in neural cholinergic regulation of inflammation in several conditions characterized with immune and metabolic derangements. Here we examined the effects of galantamine on oxidative stress in parallel with inflammatory and cardio-metabolic parameters in subjects with MetS.

Trial Design and Methods: The effects of galantamine treatment, 8 mg daily for 4 weeks or placebo, followed by 16 mg daily for 8 weeks or placebo were studied in randomly assigned subjects with MetS (n = 22 per group) of both genders. Oxidative stress, including superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase activities, lipid and protein peroxidation, and nitrite levels were analyzed before and at the end of the treatment. In addition, plasma cytokine and adipokine levels, insulin resistance (HOMA-IR) and other relevant cardio-metabolic indices were analyzed. Autonomic regulation was also examined by heart rate variability (HRV) before treatment, and at every 4 weeks of treatment.

Results: Galantamine treatment significantly increased antioxidant enzyme activities, including SOD [+1.65 USOD/mg protein, [95% CI 0.39–2.92], P = 0.004] and CAT [+0.93 nmol/mg, [95% CI 0.34–1.51], P = 0.01], decreased lipid peroxidation [thiobarbituric acid reactive substances [log scale 0.72 pmol/mg, [95% CI 0.46–1.07], P = 0.05], and systemic nitrite levels [log scale 0.83 μmol/mg protein, [95% CI 0.57–1.20], P = 0.04] compared with placebo. In addition, galantamine significantly alleviated the inflammatory state and insulin resistance, and decreased the low frequency/high frequency ratio of HRV, following 8 and 12 weeks of drug treatment.

Conclusion: Low-dose galantamine alleviates oxidative stress, alongside beneficial anti-inflammatory, and metabolic effects, and modulates neural autonomic regulation in subjects with MetS. These findings are of considerable interest for further studies with the cholinergic drug galantamine to ameliorate MetS.

Keywords: metabolic syndrome, galantamine, cholinergic, oxidative stress, heart rate variability, autonomic modulation, inflammation
Abstract

Footnotes

Funding. This study was supported by Fundação de Amparo a Pesquisa do Estado de São Paulo (FAPESP), Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq), and by the National Institutes of Health (NIH) Grants NIH-NIGMS: R01GM128008 to VP, and R35GM118182-01 to KT. The funders of this study had no role in study design, data collection, data analysis, data interpretation, or manuscript writing. The corresponding author had full access to all the data of the study and had the final responsibility for the manuscript submission. This manuscript has been released as a pre-print at medRxiv (70).

Footnotes
Click here for additional data file.(103K, TIF)

References

  • 1. Grundy SM. Metabolic syndrome pandemic. Arterioscl Thromb Vascu Biol. (2008) 28:629–36. 10.1161/ATVBAHA.107.151092 [] [[PubMed]
  • 2. Eckel RH, Grundy SM, Zimmet PZ. The metabolic syndrome. Lancet (London, England). (2005) 365:1415–28. 10.1016/S0140-6736(05)66378-7 [] [[PubMed]
  • 3. Pavlov VA. The evolving obesity challenge: targeting the vagus nerve and the inflammatory reflex in the response. Pharmacol Therap. (2021) 222:107794. 10.1016/j.pharmthera.2020.107794 [] [[PubMed]
  • 4. Roberts CK, Sindhu KK. Oxidative stress and metabolic syndrome. Life Sci. (2009) 84:705–712. 10.1016/j.lfs.2009.02.026 [] [[PubMed]
  • 5. Bonomini F, Rodella LF, Rezzani R. Metabolic syndrome, aging and involvement of oxidative stress. Aging Dis. (2015) 6:109–20. 10.14336/AD.2014.0305 ] [
  • 6. Van Guilder GP, Hoetzer GL, Greiner JJ, Stauffer BL, Desouza CA. Influence of metabolic syndrome on biomarkers of oxidative stress and inflammation in obese adults. Obesity. (2006) 14:2127–31. 10.1038/oby.2006.248 [] [[PubMed]
  • 7. Sutherland JP, McKinley B, Eckel RH. The metabolic syndrome and inflammation. Metab Syndr Related Disord. (2004) 2:82–104. 10.1089/met.2004.2.82 [] [[PubMed]
  • 8. Mancia G, Bousquet P, Elghozi JL, Esler M, Grassi G, Julius S, et al. . The sympathetic nervous system and the metabolic syndrome. J Hypert. (2007) 25:909–20. 10.1097/HJH.0b013e328048d004 [] [[PubMed]
  • 9. Tentolouris N, Liatis S, Katsilambros N. Sympathetic system activity in obesity and metabolic syndrome. Ann N Y Acad Sci. (2006) 1083:129–52. 10.1196/annals.1367.010 [] [[PubMed]
  • 10. Pavlov VA, Tracey KJ. The vagus nerve and the inflammatory reflex–linking immunity and metabolism. Nat Rev Endocrinol. (2012) 8:743–54. 10.1038/nrendo.2012.189 ] [
  • 11. Koskinen T, Kahonen M, Jula A, Mattsson N, Laitinen T, Keltikangas-Jarvinen L, et al. . Metabolic syndrome and short-term heart rate variability in young adults. The cardiovascular risk in young Finns study. Diabetic Med J Br Diab Assoc. (2009) 26:354–61. 10.1111/j.1464-5491.2009.02686.x [] [[PubMed]
  • 12. Schlaich M, Straznicky N, Lambert E, Lambert G. Metabolic syndrome: a sympathetic disease?Lancet Diab Endocrinol. (2015) 3:148–57. 10.1016/S2213-8587(14)70033-6 [] [[PubMed]
  • 13. Conti FF, Brito Jde O, Bernardes N, Dias Dda S, Sanches IC, Malfitano C, et al. . Cardiovascular autonomic dysfunction and oxidative stress induced by fructose overload in an experimental model of hypertension and menopause. BMC Cardiovasc Disord. (2014) 14:185. 10.1186/1471-2261-14-185 ] [
  • 14. Gerritsen J, Dekker JM, TenVoorde BJ, Kostense PJ, Heine RJ, Bouter LM, et al. . Impaired autonomic function is associated with increased mortality, especially in subjects with diabetes, hypertension, or a history of cardiovascular disease: the Hoorn Study. Diab Care. (2001) 24:1793–798. 10.2337/diacare.24.10.1793 [] [[PubMed]
  • 15. Carrier A. Metabolic syndrome and oxidative stress: a complex relationship. Antioxid Redox Signal. (2017) 26:429–31. 10.1089/ars.2016.6929 [] [[PubMed]
  • 16. de Melo LGP, Nunes SOV, Anderson G, Vargas HO, Barbosa DS, Galecki P, et al. . Shared metabolic and immune-inflammatory, oxidative and nitrosative stress pathways in the metabolic syndrome and mood disorders. Progress Neuro-Psychopharm Biol Psych. (2017) 78:34–50. 10.1016/j.pnpbp.2017.04.027 [] [[PubMed]
  • 17. Esser N, Paquot N, Scheen AJ. Anti-inflammatory agents to treat or prevent type 2 diabetes, metabolic syndrome and cardiovascular disease. Expert Opinion Invest Drugs. (2015) 24:283–307. 10.1517/13543784.2015.974804 [] [[PubMed]
  • 18. Chang EH, Chavan SS, Pavlov VA. Cholinergic control of inflammation, metabolic dysfunction, and cognitive impairment in obesity-associated disorders: mechanisms and novel therapeutic opportunities. Front Neurosci. (2019) 13:263. 10.3389/fnins.2019.00263 ] [
  • 19. Tracey KJ. The inflammatory reflex. Nature. (2002) 420:853–859. 10.1038/nature01321 [] [[PubMed]
  • 20. Pavlov VA, Tracey KJ. Neural regulation of immunity: molecular mechanisms and clinical translation. Nat Neurosci. (2017) 20:156–66. 10.1038/nn.4477 [] [[PubMed]
  • 21. Pavlov VA, Ochani M, Gallowitsch-Puerta M, Ochani K, Huston JM, Czura CJ, et al. . Central muscarinic cholinergic regulation of the systemic inflammatory response during endotoxemia. Proc Natl Acad Sci US A. (2006) 103:5219–23. 10.1073/pnas.0600506103 ] [
  • 22. Pavlov VA, Parrish WR, Rosas-Ballina M, Ochani M, Puerta M, Ochani K, et al. . Brain acetylcholinesterase activity controls systemic cytokine levels through the cholinergic anti-inflammatory pathway. Brain Behav Immun. (2009) 23:41–5. 10.1016/j.bbi.2008.06.011 ] [
  • 23. Ji H, Rabbi MF, Labis B, Pavlov VA, Tracey KJ, Ghia JE. Central cholinergic activation of a vagus nerve-to-spleen circuit alleviates experimental colitis. Mucosal Immunol. (2014) 7:335–47. 10.1038/mi.2013.52 ] [
  • 24. Munyaka P, Rabbi MF, Pavlov VA, Tracey KJ, Khafipour E, Ghia JE. Central muscarinic cholinergic activation alters interaction between splenic dendritic cell and CD4+CD25- T cells in experimental colitis. PLoS ONE. (2014) 9:e109272. 10.1371/journal.pone.0109272 ] [
  • 25. Pavlov VA, Tracey KJ. Neural circuitry and immunity. Immunol Res. (2015) 63:38–57. 10.1007/s12026-015-8718-1 ] [
  • 26. Birks J. Cholinesterase inhibitors for Alzheimer's disease. Cochr Database Syst Rev. (2006) 2006:Cd005593. 10.1002/14651858.CD005593 [] [[PubMed]
  • 27. Prvulovic D, Hampel H, Pantel J. Galantamine for Alzheimer's disease. Expert Opin Drug Metab Toxicol. (2010) 6:345–54. 10.1517/17425251003592137 [] [[PubMed]
  • 28. Kaushik V, Smith ST, Mikobi E, Raji MA. Acetylcholinesterase inhibitors: beneficial effects on comorbidities in patients with Alzheimer's disease. Am J Alzheimer's Dis Other Dem. (2018) 33:73–85. 10.1177/1533317517734352 [] [[PubMed]
  • 29. Metz CN, Pavlov VA. Treating disorders across the lifespan by modulating cholinergic signaling with galantamine. J Neurochem. (2020). 10.1111/jnc.15243. [Epub ahead of print]. [] [[PubMed]
  • 30. Satapathy SK, Ochani M, Dancho M, Hudson LK, Rosas-Ballina M, Valdes-Ferrer SI, et al. . Galantamine alleviates inflammation and other obesity-associated complications in high-fat diet-fed mice. Mol Med. (2011) 17:599–606. 10.2119/molmed.2011.00083 ] [
  • 31. Hanes WM, Olofsson PS, Kwan K, Hudson LK, Chavan SS, Pavlov VA, et al. . Galantamine attenuates type 1 diabetes and inhibits anti-insulin antibodies in non-obese diabetic mice. Mol Med. (2015) 21:702–8. 10.2119/molmed.2015.00142 ] [
  • 32. Pham GS, Wang LA, Mathis KW. Pharmacological potentiation of the efferent vagus nerve attenuates blood pressure and renal injury in a murine model of systemic lupus erythematosus. Am J Physiol Regul Integr Comp Physiol. (2018) 315:R1261–71. 10.1152/ajpregu.00362.2017 ] [
  • 33. Gowayed MA, Refaat R, Ahmed WM, El-Abhar HS. Effect of galantamine on adjuvant-induced arthritis in rats. Europ J Pharm. (2015) 764:547–53. 10.1016/j.ejphar.2015.07.038 [] [[PubMed]
  • 34. Waldburger JM, Boyle DL, Edgar M, Sorkin LS, Levine YA, Pavlov VA, et al. . Spinal p38 MAP kinase regulates peripheral cholinergic outflow. Arthr Rheum. (2008) 58:2919–21. 10.1002/art.23807 ] [
  • 35. Consolim-Colombo FM, Sangaleti CT, Costa FO, Morais TL, Lopes HF, Motta JM, et al. . Galantamine alleviates inflammation and insulin resistance in patients with metabolic syndrome in a randomized trial. JCI Insight. (2017) 2:1–13. 10.1172/jci.insight.93340 ] [
  • 36. Netzer N, Gatterer H, Faulhaber M, Burtscher M, Pramsohler S, Pesta D. Hypoxia, oxidative stress and fat. Biomolecules. (2015) 5:1143–50. 10.3390/biom5021143 ] [
  • 37. Whaley-Connell A, McCullough PA, Sowers JR. The role of oxidative stress in the metabolic syndrome. Rev Cardiovasc Med. (2011) 12:21–9. 10.3909/ricm0555 [] [[PubMed]
  • 38. Soares R, Costa C. Oxidative Stress, Inflammation and Angiogenesis in the Metabolic Syndrome. Porto: Springer; (2009). 10.1007/978-1-4020-9701-0 [[PubMed]
  • 39. Fernández-García JC, Cardona F, Tinahones FJ. Inflammation, oxidative stress and metabolic syndrome: dietary modulation. Curr Vasc Pharm. (2013) 11:906–19. 10.2174/15701611113116660175 [] [[PubMed]
  • 40. Marklund SL. Superoxide dismutase isoenzymes in tissues and plasma from New Zealand black mice, nude mice and normal BALB/c mice. Mutation Res. (1985) 148:129–34. 10.1016/0027-5107(85)90216-7 [] [[PubMed]
  • 41. Buege JA, Aust SD. Microsomal lipid peroxidation. Methods Enzymol. (1978) 52:302–10. 10.1016/S0076-6879(78)52032-6 [] [[PubMed]
  • 42. Aebi H. Catalase in vitro. Methods Enzymol. (1984) 105:121–6. 10.1016/S0076-6879(84)05016-3 [] [[PubMed]
  • 43. Reznick AZ, Packer L. Oxidative damage to proteins: spectrophotometric method for carbonyl assay. Methods Enzymol. (1994) 233:357–63. 10.1016/S0076-6879(94)33041-7 [] [[PubMed]
  • 44. Heart rate variability: standards of measurement physiological interpretation and clinical use . Task force of the european society of cardiology and the north american society of pacing and electrophysiology. Circulation. (1996) 93:1043–65. 10.1161/01.CIR.93.5.1043 [] [[PubMed]
  • 45. Samaha FF, Szapary PO, Iqbal N, Williams MM, Bloedon LT, Kochar A, et al. . Effects of rosiglitazone on lipids, adipokines, and inflammatory markers in nondiabetic patients with low high-density lipoprotein cholesterol and metabolic syndrome. Arterioscl Thromb Vasc Biol. (2006) 26:624–30. 10.1161/01.ATV.0000200136.56716.30 [] [[PubMed]
  • 46. Risérus U, Arner P, Brismar K, Vessby B. Treatment with dietary trans10cis12 conjugated linoleic acid causes isomer-specific insulin resistance in obese men with the metabolic syndrome. Diabetes Care. (2002) 25:1516–21. 10.2337/diacare.25.9.1516 [] [[PubMed]
  • 47. Laborde S, Mosley E, Mertgen A. A unifying conceptual framework of factors associated to cardiac vagal control. Heliyon. (2018) 4:e01002. 10.1016/j.heliyon.2018.e01002 ] [
  • 48. Xydakis AM, Case CC, Jones PH, Hoogeveen RC, Liu M-Y, Smith EOB, et al. . Adiponectin, inflammation, and the expression of the metabolic syndrome in obese individuals: the impact of rapid weight loss through caloric restriction. J Clin Endocrinol Metab. (2004) 89:2697–703. 10.1210/jc.2003-031826 [] [[PubMed]
  • 49. Sureda A, Bibiloni MD, Martorell M, Buil-Cosiales P, Marti A, Pons A, et al. . Mediterranean diets supplemented with virgin olive oil and nuts enhance plasmatic antioxidant capabilities and decrease xanthine oxidase activity in people with metabolic syndrome: the PREDIMED study. Mol Nutr Food Res. (2016) 60:2654–64. 10.1002/mnfr.201600450 [] [[PubMed]
  • 50. Kozirog M, Poliwczak AR, Duchnowicz P, Koter-Michalak M, Sikora J, Broncel M. Melatonin treatment improves blood pressure, lipid profile, and parameters of oxidative stress in patients with metabolic syndrome. J Pineal Res. (2011) 50:261–6. 10.1111/j.1600-079X.2010.00835.x [] [[PubMed]
  • 51. Tabrizi R Tamtaji OR Lankarani KB Mirhosseini N Akbari M Dadgostar E . The effects of resveratrol supplementation on biomarkers of inflammation and oxidative stress among patients with metabolic syndrome and related disorders: a systematic review and meta-analysis of randomized controlled trials. Food Funct. (2018) 9:6116–28. 10.1039/C8FO01259H [] [[PubMed]
  • 52. Diaz-Flores M, Cruz M, Duran-Reyes G, Munguia-Miranda C, Loza-Rodriguez H, Pulido-Casas E, et al. . Oral supplementation with glycine reduces oxidative stress in patients with metabolic syndrome, improving their systolic blood pressure. Canad J Physiol Pharm. (2013) 91:855–60. 10.1139/cjpp-2012-0341 [] [[PubMed]
  • 53. Wang Q, Sun Y, Ma A, Li Y, Han X, Liang H. Effects of vitamin E on plasma lipid status and oxidative stress in Chinese women with metabolic syndrome. Int J Vitamin Nutr Res. (2010) 80:178–87. 10.1024/0300-9831/a000015 [] [[PubMed]
  • 54. Tilg H, Moschen AR. Adipocytokines: mediators linking adipose tissue, inflammation and immunity. Nat Rev Immunol. (2006) 6:772–83. 10.1038/nri1937 [] [[PubMed]
  • 55. Ouchi N, Parker JL, Lugus JJ, Walsh K. Adipokines in inflammation and metabolic disease. Nat Rev Immunol. (2011) 11:85–97. 10.1038/nri2921 ] [
  • 56. Pavlov VA. Collateral benefits of studying the vagus nerve in bioelectronic medicine. Bioelectr Med. (2019) 5:5. 10.1186/s42234-019-0021-3 ] [
  • 57. Lehner KR, Silverman HA, Addorisio ME, Roy A, Al-Onaizi MA, Levine Y, et al. . Forebrain cholinergic signaling regulates innate immune responses and inflammation. Front Immunol. (2019) 10:585. 10.3389/fimmu.2019.00585 ] [
  • 58. Bonaz B, Sinniger V, Hoffmann D, Clarencon D, Mathieu N, Dantzer C, et al. . Chronic vagus nerve stimulation in Crohn's disease: a 6-month follow-up pilot study. Neurogastroenterol Motility. (2016) 28:948–953. 10.1111/nmo.12792 [] [[PubMed]
  • 59. Levine YA, Koopman FA, Faltys M, Caravaca A, Bendele A, Zitnik R, et al. . Neurostimulation of the cholinergic anti-inflammatory pathway ameliorates disease in rat collagen-induced arthritis. PLoS ONE. (2014) 9:e104530. 10.1371/journal.pone.0104530 ] [
  • 60. Koopman FA, Chavan SS, Miljko S, Grazio S, Sokolovic S, Schuurman PR, et al. . Vagus nerve stimulation inhibits cytokine production and attenuates disease severity in rheumatoid arthritis. Proc Natl Acad Sci US A. (2016) 113:8284–9. 10.1073/pnas.1605635113 ] [
  • 61. Pavlov VA, Chavan SS, Tracey KJ. Bioelectronic medicine: from preclinical studies on the inflammatory reflex to new approaches in disease diagnosis and treatment. Cold Spring Harb Perspect Med. (2020) 10:a034140. 10.1101/cshperspect.a034140 ] [
  • 62. Taché Y. The vagal anti-inflammatory pathways to the viscera: from basic understanding to therapeutic implications. J Physiol. (2016) 594:5769–70. 10.1113/JP271540 ] [
  • 63. Pavlov VA, Chavan SS, Tracey KJ. Molecular and functional neuroscience in immunity. Ann Rev Immunol. (2018) 36:783–812. 10.1146/annurev-immunol-042617-053158 ] [
  • 64. Nordstrom P, Religa D, Wimo A, Winblad B, Eriksdotter M. The use of cholinesterase inhibitors and the risk of myocardial infarction and death: a nationwide cohort study in subjects with Alzheimer's disease. Europ Heart J. (2013) 34:2585–91. 10.1093/eurheartj/eht182 [] [[PubMed]
  • 65. Wu PH, Lin YT, Hsu PC, Yang YH, Lin TH, Huang CT. Impact of acetylcholinesterase inhibitors on the occurrence of acute coronary syndrome in patients with dementia. Sci Rep. (2015) 5:15451. 10.1038/srep15451 ] [
  • 66. Isik AT, Soysal P, Stubbs B, Solmi M, Basso C, Maggi S, et al. . Cardiovascular outcomes of cholinesterase inhibitors in individuals with dementia: a meta-analysis and systematic review. J Am Geriatr Soc. (2018) 66:1805–811. 10.1111/jgs.15415 [] [[PubMed]
  • 67. Secnik J, Schwertner E, Alvarsson M, Hammar N, Fastbom J, Winblad B, et al. . Cholinesterase inhibitors in patients with diabetes mellitus and dementia: an open-cohort study of ~23 000 patients from the Swedish Dementia Registry. BMJ Open Diabetes Res Care. (2020) 8:e000833. 10.1136/bmjdrc-2019-000833 ] [
  • 68. Isik AT, Bozoglu E, Naharci MI, Kilic S. Evaluation of the effects of galantamine on cardiac function in elderly patients with Alzheimer's disease. Am J Geriatr Pharmacoth. (2010) 8:454. 10.1016/j.amjopharm.2010.09.001 [] [[PubMed]
  • 69. Metz CN, Pavlov VA. Vagus nerve cholinergic circuitry to the liver and the gastrointestinal tract in the neuroimmune communicatome. Am J Physiol Gastrointestinal Liver Physiol. (2018) 315:G651–8. 10.1152/ajpgi.00195.2018 ] [
  • 70. Sangaleti CT, Katayama KY, De Angelis K, de Moraes TL, Araujo AA, Lopes HF, et al. . Galantamine alleviates oxidative stress alongside anti-inflammatory and cardio-metabolic effects in subjects with the metabolic syndrome in a randomized trial. medRxiv. (2020) 2020:2019.2012.2030.19016105. 10.1101/2019.12.30.19016105 [[PubMed]
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.