Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2110 2023-01-06 11:35:35 |
2 format correct Meta information modification 2110 2023-01-09 06:29:02 | |
3 format correct Meta information modification 2110 2023-01-09 06:42:52 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Łuc, M.;  Woźniak, M.;  Rymaszewska, J. Tumor Necrosis Factor Antagonists in Neuroinflammation for Dementia. Encyclopedia. Available online: https://encyclopedia.pub/entry/39838 (accessed on 17 May 2024).
Łuc M,  Woźniak M,  Rymaszewska J. Tumor Necrosis Factor Antagonists in Neuroinflammation for Dementia. Encyclopedia. Available at: https://encyclopedia.pub/entry/39838. Accessed May 17, 2024.
Łuc, Mateusz, Marta Woźniak, Joanna Rymaszewska. "Tumor Necrosis Factor Antagonists in Neuroinflammation for Dementia" Encyclopedia, https://encyclopedia.pub/entry/39838 (accessed May 17, 2024).
Łuc, M.,  Woźniak, M., & Rymaszewska, J. (2023, January 06). Tumor Necrosis Factor Antagonists in Neuroinflammation for Dementia. In Encyclopedia. https://encyclopedia.pub/entry/39838
Łuc, Mateusz, et al. "Tumor Necrosis Factor Antagonists in Neuroinflammation for Dementia." Encyclopedia. Web. 06 January, 2023.
Tumor Necrosis Factor Antagonists in Neuroinflammation for Dementia
Edit

Prolonged activation of pro-inflammatory responses in Alzheimer’s disease (AD) alters function of glial cells and in turn, further accelerates neuroinflammation. Subsequent synaptic dysfunction and loss of neurons are responsible for clinical symptoms of the disease. Additionally, factors such as insufficient sleep length and subsequent reduction in amyloid clearance via the glymphatic system lead to amyloid accumulation, while simultaneously aggravating systemic inflammatory response. Inflammation in vascular dementia (VaD) contributes to the three-hit hypothesis, along with hypertension and hypoxia. Vasculitis is responsible for restricted blood circulation in microvessels and leads to decreased oxygen supply and regional glial activation favoring neuroinflammation in the central nervous system (CNS). Peripheral immune response and increased cytokine production, including an early surge in TNF and IL-1β concentrations activate glia, leading to aggravation of neuroinflammation and dysfunction of neurons during COVID-19. Lifestyle factors, such as diet, have a large impact on future cognitive outcomes and should be included as a crucial intervention in dementia prevention.

TNF COVID-19 neuroinflammation

1. Tumor Necrosis Factor Triggers Dementia Pathology

Among various cytokines involved in the immune response, tumor necrosis factor (TNF) is considered to play a significant upstream role in dementia pathology on a molecular level. Its pleiotropic effects vary from physiological neuroprotective and repair activities to pathological neuronal loss occurring in neurodegenerative and autoimmune conditions which are dependent on TNF form and activated receptor [1]. TNF receptor type 1 (TNFR1) is commonly expressed and can be bound by both transmembrane and soluble TNF forms, while TNF receptor type 2 (TNFR2), expressed by myeloid and endothelial cells but also by CNS-residing glia and neurons, is mainly bound by transmembrane TNF [2]. Protective versus deleterious outcomes of receptor activation depend on various factors including TNF concentration, activation of other signaling pathways or cell susceptibility resulting from cell type and age-related priming [3][4]. TNF signaling activates intracellular pathways with transcription factors such as NFκB (nuclear factor kappa-light-chain-enhancer of activated B cells) leading to pro-inflammatory cytokine production and, in conditions of prolonged signaling, aggravation of inflammation [5][6].
Exposure to TNF in an in vitro AD model has been shown to result in aggregation of extracellular proteins which are considered characteristic traits of AD and LBD pathology [7]. Interestingly, exposure to TNF does not need to be sustained in order to maintain increased secretion and aggregation of amyloid-β or α-synuclein in such models. This mechanism may demonstrate profound effects of TNF signaling on pro-inflammatory activation and cytokine production of astrocytes and microglia, leading to prolonged neuroinflammation [8]. This phenomenon has been reflected in a cohort study by Lindbergh et al., in which TNF plasma concentration of participants were assessed annually. Increased systemic TNF resulted in reduction in grey matter volumes in further assessments in a curvilinear correlation, with initially-increased TNF correlating with following loss of volume [9]. Additionally, within-person increases in TNF correlated with lower scores obtained in neuropsychological evaluation with the use of Mini Mental State Examination.

2. Methods for Reduction of Pro-Inflammatory Activation

Since TNF signaling exerts a triggering effect in the development of cognitive decline [7][9], the question arises, whether interventions focused on reduction in TNF concentrations or inhibition of its signaling pathway may play a role in dementia prevention or treatment. There are several methods of reducing an inflammatory state in the human organism, including dietary and pharmacological interventions.
Diet has been implicated in pathology of numerous neurodegenerative diseases, including dementia. A well-balanced diet provides all nutritional ingredients necessary for maintaining healthy and functional neurons [10]. On the other hand, it is believed that diet largely impacts inflammatory activation and immune response, which in turn influences regional inflammatory response in the brain. Various studies, including large cohorts, have associated inflammatory dietary patterns with faster cognitive decline and subsequent cognitive impairment [11][12][13]. The correlation between inflammatory diet and cognition is especially apparent in regard to several cognitive functions, such as episodic memory, semantic-based memory, executive functions and working memory [14].
The Western diet which comprises highly-processed food rich in fructose and saturated fat is known to increase TNF concentration in animal models [15]. Mice immunized against Klebsiella pneumoniae were shown to have lower levels of inflammasome-related inflammation, providing yet another link between gut microbiota and inflammation. This phenomenon was mediated by the presence of apolipoprotein E and was not observed in ApoE −/− animals [15]. Although individually tailored diets are not yet within reach and require further exploration, some conclusions regarding the influence of particular diets on neurodegenerative processes can be made based on existing studies [16]. For example, use of Dietary Inflammatory index (DII), which takes into account individual’s dietary composition and characteristics, allows researchers to indicate the general influence of one’s diet on systemic cytokine levels and inflammatory activation [17]. Higher scores of DII were shown to strongly correlate with worse cognitive performance [18]. It stands to reason, that the influence of environmental factors such as diet in preserving cognition cannot be underestimated. On the other hand, it seems that the inflammatory potential of the consumed food may be related not only to the specific products but also to the gut microbiota composition resulting from the daily diet. A summary of findings associated with specific diets can be found in Table 1 [19][20][21][22][23][24][25][26][27][28].
Table 1. Summary of findings associated with diet and inflammation.
Study (Type) Outcomes Diet/Intervention Group Key Findings
Ostan et al., 2015 [19]
(cohort study)
Inflammatory and metabolic parameters RISTOMED diet (personalized and balanced)
+/− nutraceutics
125 participants RISTOMED diet alone or with each nutraceutical supplementation significantly decreased erythrocyte sedimentation rate
Kim et al., 2022 [20]
(non-randomized intervention study)
Inflammatory parameters
Insulin sensitivity
Short-term ketogenic diet (3 days) 15 participants Short-term Ketogenic diet resulted in lower IL-1β and TNF secretion;
Improved insulin sensitivity
Al-Abauidy et al., 2021 [21]
(randomized clinical trial)
Oxidative stress and inflammatory parameters Mediterranean diet (12 weeks) 19 participants Mediterranean diet reduced IL-6 levels by 49% and levels of oxidative stress marker, 8-OHdG, by 32.4%
Georgoulis et al., 2021 [22]
(randomized clinical trial)
Oxidative stress and inflammatory parameters Mediterranean diet (6 months) 187 patients with obstructive sleep apnea Mediterranean diet reduced hs-CRP levels in patients
Casas et al., 2017 [25]
(randomized clinical trial)
Cytokine levels Mediterranean diet +/− extra virgin olive oil (5 years) 66 participants Mediterranean diet reduced IL-6, IL-8, MCP-1, and MIP-1β levels. Addition of extra virgin olive oil reduced IL-1β, IL-5, IL-7, IL-12p70, IL-18, TNF-α, IFN-γ, GCSF, GMCSF, and ENA78
Omorogieva et al., 2021 [23]
(meta-analysis)
Lipid profiles, LPS, BMI, inflammatory markers Diet rich in fiber 10 studies included in meta-analysis Dietary fiber reduces total cholesterol, BMI and CRP, but no significant changes were observed for IL-6 and TNF
Shivappa et al., 2016 [24]
(cross-sectional study)
Inflammatory markers - 532 adolescents Higher dietary inflammatory index scores were associated with increased levels of various inflammatory markers: TNF-α, IL-1, 2, IFN-γ and VCAM
Mazzoli et al., 2020 [26]
(animal study)
Inflammatory markers, insulin sensitivity, BDNF Western diet (4 weeks) 16 rats Western diet increased TNF levels in white adipose tissue and hippocampus of rats; brain BDNF and synaptotagmin I were decreased, while PSD-95 was increased.
Jena et al., 2020 [27]
(animal study)
Interleukin-17, PD-95, BDNF High sugar and high fat diet (FPC diet) for 3 months, and 5 months +/− inulin supplementation 12 mice FPC diet elevated RORγ and IL-17A signaling. Accompanied by microglia activation and reduced hippocampal long-term potentiation, FPC diet intake also reduced postsynaptic density-95 and brain derived neurotrophic factor.
Godfrey et al., 2020 [28]
(animal study)
CRP levels, CSF dopamine concentrations
Functional connectivity
12 months of obesogenic diet 34 female rhesus monkeys CSF dopamine concentrations decreased, and CRP concentrations increased. Resting-state magnetic resonance neuroimaging showed that higher CRP concentrations were associated with decreased functional connectivity.
TNF antagonists offer yet another interesting approach to reducing inflammatory activation in humans. These agents are commonly used in autoimmune diseases, such as rheumatoid arthritis, psoriasis or inflammatory bowel diseases [40]. Their effects in controlling excessive immune response are mediated by binding TNF but the exact mechanisms and affinity to soluble and transmembrane TNF vary, hence their clinical use can also differ [4][41][42]. Moreover, some other mechanisms of action have been described, such as lymphotoxin-α blocking by etanercept. Additionally, infliximab has been proven to reduce expression of GM-CSF (granulocyte-macrophage colony stimulating factor) [43][44], while infliximab and adalimumab are able to induce production of immunosuppressive IL-10 by macrophages in vitro [45]. Psoriasis patients treated with etanercept had decreased expression of IL-1 and IL-8 genes which correlated with reduction in total pro-inflammatory immune response [46]. Importantly, although molecular weight and properties do not allow for free entry of TNF antagonists into the CNS, evidence indicates that these drugs may have protective influence against brain aging.
It seems that both BBB-nonpenetrating and modified, BBB-penetrating, etanercept reduce tauopathy, microgliosis and therefore, neuronal loss in a mouse model of AD. Additionally, they increase PSD95 protein levels indicating synaptic health. This phenomenon may be related to the peripheral effects of the drug and underlines the importance of tackling chronic inflammatory activation in order to maintain physiological neuronal function [47]. Similar observations had been made previously, in a study by Chang et al. [48]. Administration of BBB-penetrating TNF-inhibitor, cTfRMAb-TNFR, resulted in a significant decrease in neuroinflammatory markers, amyloid burden and BBB disruption in an AD mouse model. These results were comparable to those obtained with the use of etanercept in regard to amyloid burden and BBB integrity but not for neuroinflammation portrayed with ICAM-1 concentration. The cognitive performance of tested mice was highest in a group treated with cTfRMAb-TNFR highlighting the crucial role of neuroinflammation in cognitive decline [48]. Use of BBB-penetrating agents remains especially interesting in regard to FTD pathology, in which cytokine production seems to take place mainly in the brain [49].
An experiment conducted in an animal model of metabolic syndrome proved that intraperitoneal administration of infliximab improved lipid profiles in rats—i.e., decreased triglycerides and increased HDL. Additionally, the study group had lower adiponectin concentrations compared to the control, impacting low-grade inflammation. Noteworthy, not all metabolic aspects were normalized and cognitive tests were not included in the study protocol [50]. It may be hypothesized that an up-stream role of TNF in obesity-induced pathology, limits the potential protective effects of TNF inhibition in conditions of already present metabolic syndrome. On the other hand, intracerebroventricular administration of infliximab in a transgenic mouse model of AD resulted in a significant decrease in brain TNF levels, reduction in amyloid burden and tau pathology [51]. Another study conducted in a rat model of VaD, revealed a therapeutic effect of adalimumab administration in treating cognitive deficits resulting from cerebral hypoperfusion. This finding was associated with a reduction in neuronal loss and of microglial activation mediated by NFκB suppression [52].
A systematic literature review of TNF antagonist effects on AD revealed beneficial influence of TNF inhibition on cognition [5]. Most commonly studied agents, etanercept, infliximab and adalimumab, used in rheumatoid arthritis coincided with up to 60–70% reduction in AD incidence in large epidemiological analyzes of rheumatoid arthritis patients, whereas other methods of treatment did not affect AD incidence [53]. Similar results were obtained in patients with psoriasis who were treated with etanercept, infliximab or adalimumab [54]. Rheumatoid arthritis and psoriasis, among other autoimmune diseases, are known risk factors of AD due to the occurrence of persistent inflammatory activation which may constitute a confounding factor for generalized conclusions [55][56][57]. However, these observations warrant further research in other populations in order to establish a protective role of TNF inhibition on development and progression of various types of dementia. Updates on current clinical trials can be found in Table 2.

References

  1. Probert, L. TNF and its receptors in the CNS: The essential, the desirable and the deleterious effects. Neuroscience 2015, 302, 2–22.
  2. Decourt, B.; Lahiri, D.K.; Sabbagh, M.N. Targeting Tumor Necrosis Factor Alpha for Alzheimer’s Disease. Curr. Alzheimer Res. 2016, 14, 412–425.
  3. Montgomery, S.L.; Bowers, W.J. Tumor necrosis factor-alpha and the roles it plays in homeostatic and degenerative processes within the central nervous system. J. Neuroimmune Pharmacol. 2012, 7, 42–59.
  4. Tracey, D.; Klareskog, L.; Sasso, E.H.; Salfeld, J.G.; Tak, P.P. Tumor necrosis factor antagonist mechanisms of action: A comprehensive review. Pharmacol. Ther. 2008, 117, 244–279.
  5. Torres-Acosta, N.; O’Keefe, J.H.; O’Keefe, E.L.; Isaacson, R.; Small, G. The rapeutic Potential of TNF-α Inhibition for Alzheimer’s Disease Prevention. J. Alzheimer’s Dis. 2020, 78, 619–626.
  6. Brás, J.P.; Bravo, J.; Freitas, J.; Barbosa, M.A.; Santos, S.G.; Summavielle, T.; Almeida, M.I. TNF-alpha-induced microglia activation requires miR-342: Impact on NF-kB signaling and neurotoxicity. Cell Death Dis. 2020, 11, 415.
  7. Whiten, D.R.; Brownjohn, P.W.; Moore, S.; De, S.; Strano, A.; Zuo, Y.; Haneklaus, M.; Klenerman, D.; Livesey, F.J. Tumour necrosis factor induces increased production of extracellular amyloid-β- and α-synuclein-containing aggregates by human Alzheimer’s disease neurons. Brain Commun. 2020, 2, fcaa146.
  8. Raffaele, S.; Lombardi, M.; Verderio, C.; Fumagalli, M. TNF Production and Release from Microglia via Extracellular Vesicles: Impact on Brain Functions. Cells 2020, 9, 2145.
  9. Lindbergh, C.A.; Casaletto, K.B.; Staffaroni, A.M.; Elahi, F.; Walters, S.M.; You, M.; Neuhaus, J.; Contreras, W.R.; Wang, P.; Karydas, A.; et al. Systemic tumor necrosis factor-alpha trajectories relate to brain health in typically aging older adults. J. Gerontol Ser. A Biol. Sci. Med. Sci. 2020, 75, 1558–1565.
  10. Vasefi, M.; Hudson, M.; Ghaboolian-Zare, E. Diet Associated with Inflammation and Alzheimer’s Disease. J. Alzheimer’s Dis. Rep. 2019, 3, 299–309.
  11. Ozawa, M.; Shipley, M.; Kivimaki, M.; Singh-Manoux, A.; Brunner, E.J. Dietary pattern, inflammation and cognitive decline: The Whitehall II prospective cohort study. Clin. Nutr. 2017, 36, 506–512.
  12. Shin, D.; Kwon, S.C.; Kim, M.H.; Lee, K.W.; Choi, S.Y.; Shivappa, N.; Hébert, J.R.; Chung, H.-K. Inflammatory potential of diet is associated with cognitive function in an older adult Korean population. Nutrition 2018, 55–56, 56–62.
  13. Hayden, K.M.; Beavers, D.P.; Steck, S.E.; Hebert, J.R.; Tabung, F.K.; Shivappa, N.; Casanova, R.; Manson, J.E.; Padula, C.B.; Salmoirago-Blotcher, E.; et al. The association between an inflammatory diet and global cognitive function and incident dementia in older women: The Women’s Health Initiative Memory Study. Alzheimer’s Dement. 2017, 13, 1187–1196.
  14. Frith, E.; Shivappa, N.; Mann, J.R.; Hébert, J.R.; Wirth, M.D.; Loprinzi, P.D. Dietary inflammatory index and memory function: Population-based national sample of elderly Americans. Br. J. Nutr. 2018, 119, 552–558.
  15. Saita, D.; Ferrarese, R.; Foglieni, C.; Esposito, A.; Canu, T.; Perani, L.; Ceresola, E.R.; Visconti, L.; Burioni, R.; Clementi, M.; et al. Adaptive immunity against gut microbiota enhances apoE-mediated immune regulation and reduces atherosclerosis and western-diet-related inflammation. Sci. Rep. 2016, 6, 29353.
  16. Milošević, M.; Arsić, A.; Cvetković, Z.; Vučić, V. Memorable Food: Fighting Age-Related Neurodegeneration by Precision Nutrition. Front. Nutr. 2021, 8, 688086.
  17. Shivappa, N.; Steck, S.E.; Hurley, T.G.; Hussey, J.R.H.J. Dietary Inflammatory Index. Public Health Nutr. 2014, 17, 1689–1696.
  18. Kesse-Guyot, E.; Assmann, K.E.; Andreeva, V.A.; Touvier, M.; Neufcourt, L.; Shivappa, N.; Hébert, J.R.; Wirth, M.D.; Hercberg, S.; Galan, P.; et al. Long-term association between the dietary inflammatory index and cognitive functioning: Findings from the SU.VI.MAX study. Eur. J. Nutr. 2017, 56, 1647–1655.
  19. Ostan, R.; Béné, M.C.; Spazzafumo, L.; Pinto, A.; Donini, L.M.; Pryen, F.; Charrouf, Z.; Valentini, L.; Lochs, H.; Bourdel-Marchasson, I.; et al. Impact of diet and nutraceutical supplementation on inflammation in elderly people. Results from the RISTOMED study, an open-label randomized control trial. Clin. Nutr. 2016, 35, 812–818.
  20. Kim, E.R.; Kim, S.R.; Cho, W.; Lee, S.G.; Kim, S.H.; Kim, J.H.; Choi, E.; Kim, J.-H.; Yu, J.-W.; Lee, B.-W.; et al. Short Term Isocaloric Ketogenic Diet Modulates NLRP3 Inflammasome Via B-hydroxybutyrate and Fibroblast Growth Factor 21. Front. Immunol. 2022, 13, 843520.
  21. Al-Aubaidy, H.A.; Dayan, A.; Deseo, M.A.; Itsiopoulos, C.; Jamil, D.; Hadi, N.R.; Thomas, C. Twelve-week mediterranean diet intervention increases citrus bioflavonoid levels and reduces inflammation in people with type 2 diabetes mellitus. Nutrients 2021, 13, 1133.
  22. Georgoulis, M.; Yiannakouris, N.; Tenta, R.; Fragopoulou, E.; Kechribari, I.; Lamprou, K.; Perraki, E.; Vagiakis, E.; Kontogianni, M.D. A weight-loss Mediterranean diet/lifestyle intervention ameliorates inflammation and oxidative stress in patients with obstructive sleep apnea: Results of the “MIMOSA” randomized clinical trial. Eur. J. Nutr. 2021, 60, 3799–3810.
  23. Ojo, O.; Ojo, O.O.; Zand, N.; Wang, X. The Effect of Dietary Fibre on Gut Microbiota, Lipid Profile, and Inflammatory Markers in Patients with Type 2 Diabetes: A Systematic Review and Meta-Analysis of Randomised Controlled Trials. Nutrients 2021, 13, 1805.
  24. Shivappa, N.; Hebert, J.R.; Marcos, A.; Diaz, L.E.; Gomez, S.; Nova, E.; Michels, N.; Arouca, A.; González-Gil, E.; Frederic, G.; et al. Association between dietary inflammatory index and inflammatory markers in the HELENA study. Mol. Nutr. Food Res. 2017, 61, 1–23.
  25. Casas, R.; Urpi-Sardà, M.; Sacanella, E.; Arranz, S.; Corella, D.; Castañer, O.; Lamuela-Raventós, R.-M.; Salas-Salvadó, J.; Lapetra, J.; Portillo, M.P.; et al. Anti-Inflammatory Effects of the Mediterranean Diet in the Early and Late Stages of Atheroma Plaque Development. Mediat. Inflamm. 2017, 2017, 3674390.
  26. Mazzoli, A.; Spagnuolo, M.S.; Gatto, C.; Nazzaro, M.; Cancelliere, R.; Crescenzo, R.; Iossa, S.; Cigliano, L. Adipose tissue and brain metabolic responses to western diet—is there a similarity between the two? Int. J. Mol. Sci. 2020, 21, 786.
  27. Jena, P.K.; Sheng, L.; Nguyen, M.; Di Lucente, J.; Hu, Y.; Li, Y.; Maezawa, I.; Jin, L.-W.; Wan, Y.-J.Y. Dysregulated bile acid receptor-mediated signaling and IL-17A induction are implicated in diet-associated hepatic health and cognitive function. Biomark. Res. 2020, 8, 59.
  28. Godfrey, J.R.; Pincus, M.; Kovacs-Balint, Z.; Feczko, E.; Earl, E.; Miranda-Dominguez, O.; Fair, D.A.; Jones, S.R.; Locke, J.; Sanchez, M.M.; et al. Obesogenic diet-associated C-reactive protein predicts reduced central dopamine and corticostriatal functional connectivity in female rhesus monkeys. Brain Behav. Immun. 2020, 88, 166–173.
  29. Weng, J.; Zhao, G.; Weng, L.; Guan, J. Aspirin using was associated with slower cognitive decline in patients with Alzheimer’s disease. PLoS ONE 2021, 16, e0252969.
  30. Sekiyama, K.; Fujita, M.; Sekigawa, A.; Takamatsu, Y.; Waragai, M.; Takenouchi, T.; Sugama, S.; Hashimoto, M. Ibuprofen ameliorates protein aggregation and astrocytic gliosis, but not cognitive dysfunction, in a transgenic mouse expressing dementia with Lewy bodies-linked P123H β-synuclein. Neurosci. Lett. 2012, 515, 97–101.
  31. Szekely, C.A.; Breitner, J.C.S.; Fitzpatrick, A.L.; Rea, T.D.; Psaty, B.M.; Kuller, L.H.; Zandi, P.P. NSAID use and dementia risk in the Cardiovascular Health Study: Role of APOE and NSAID type. Neurology 2008, 70, 17–24.
  32. Knopman, D.S.; Petersen, R.C. The quest for dementia prevention does not include an aspirin a day. Neurology 2020, 95, 105–106.
  33. Tabet, N.; Feldman, H. Ibuprofen for Alzheimer’s disease. Cochrane Database Syst. Rev. 2003, CD004031.
  34. Ryan, J.; Storey, E.; Murray, A.M.; Woods, R.L.; Wolfe, R.; Reid, C.M.; Nelson, M.R.; Chong, T.T.; Williamson, J.D.; Ward, S.A.; et al. Randomized placebo-controlled trial of the effects of aspirin on dementia and cognitive decline. Neurology 2020, 95, E320–E331.
  35. Li, H.; Li, W.; Zhang, X.; Ma, X.C.; Zhang, R.W. Aspirin Use on Incident Dementia and Mild Cognitive Decline: A Systematic Review and Meta-Analysis. Front. Aging Neurosci. 2021, 12, 578071.
  36. Jordan, F.; Quinn, T.J.; McGuinness, B.; Passmore, P.; Kelly, J.P.; Tudur Smith, C.; Murphy, K.; Devane, D. Aspirin and other non-steroidal anti-inflammatory drugs for the prevention of dementia. Cochrane Database Syst. Rev. 2020, CD011459.
  37. Rands, G.; Orrell, M. Aspirin for vascular dementia. Cochrane Database Syst. Rev. 2000, 2012, CD001296.
  38. Jaturapatporn, D.; Mgekn, I.; Mccleery, J.; Tabet, N. Aspirin, steroidal and non-steroidal anti-inflammatory drugs for the treatment of Alzheimer’s disease. Cochrane Database Syst. Rev. 2012, 2, CD006378.
  39. Veronese, N.; Stubbs, B.; Maggi, S.; Thompson, T.; Schofield, P.; Muller, C.; Tseng, P.; Lin, P.; Carvalho, A.F.; Solmi, M. Low-Dose Aspirin Use and Cognitive Function in Older Age: A Systematic Review and Meta-analysis. J. Am. Geriatr. Soc. 2017, 65, 1763–1768.
  40. Gottlieb, A.B. Tumor necrosis factor blockade: Mechanism of action. J. Investig. Dermatol. Symp. Proc. 2007, 12, 1–4.
  41. Levin, A.D.; Wildenberg, M.E.; van den Brink, G.R. Mechanism of Action of Anti-TNF Therapy in Inflammatory Bowel Disease. J. Crohn’s Colitis 2016, 10, 989–997.
  42. Mitoma, H.; Horiuchi, T.; Tsukamoto, H.; Ueda, N. Molecular mechanisms of action of anti-TNF-α agents—Comparison among therapeutic TNF-α antagonists. Cytokine 2018, 101, 56–63.
  43. Kirman, I.; Whelan, R.L.; Nielsen, O.H. Infliximab: Mechanism of action beyond TNF-α neutralization in inflammatory bowel disease. Eur. J. Gastroenterol. Hepatol. 2004, 16, 639–641.
  44. Agnholt, J.; Kelsen, J.; Brandsborg, B.; Jakobsen, N.O.; Dahlerup, J.F. Increased production of granulocyte-macrophage colony-stimulating factor in Crohn’s disease—A possible target for infliximab treatment. Eur. J. Gastroenterol. Hepatol. 2004, 16, 649–655.
  45. Vos, A.C.W.; Wildenberg, M.E.; Duijvestein, M.; Verhaar, A.P.; Van Den Brink, G.R.; Hommes, D.W. AntiTumor necrosis factor-α antibodies induce regulatory macrophages in an Fc region-dependent manner. Gastroenterology 2011, 140, 221–230.e3.
  46. Gottlieb, A.B.; Chamian, F.; Masud, S.; Cardinale, I.; Abello, M.V.; Lowes, M.A.; Chen, F.; Magliocco, M.; Krueger, J.G. TNF Inhibition Rapidly Down-Regulates Multiple Proinflammatory Pathways in Psoriasis Plaques. J. Immunol. 2005, 175, 2721–2729.
  47. Ou, W.; Yang, J.; Simanauskaite, J.; Choi, M.; Castellanos, D.M.; Chang, R.; Sun, J.; Jagadeesan, N.; Parfitt, K.D.; Cribbs, D.H.; et al. Biologic TNF-α inhibitors reduce microgliosis, neuronal loss, and tau phosphorylation in a transgenic mouse model of tauopathy. J. Neuroinflamm. 2021, 18, 312.
  48. Chang, R.; Knox, J.; Chang, J.; Derbedrossian, A.; Vasilevko, V.; Cribbs, D.; Boado, R.J.; Pardridge, W.M.; Sumbria, R.K. Blood-Brain Barrier Penetrating Biologic TNF-α Inhibitor for Alzheimer’s Disease. Mol. Pharm. 2017, 14, 2340–2349.
  49. Sjogren, M. Increased intrathecal inflammatory activity in frontotemporal dementia: Pathophysiological implications. J. Neurol. Neurosurg. Psychiatry 2004, 75, 1107–1111.
  50. Abdelhamid, Y.A.; Elyamany, M.F.; Al-Shorbagy, M.Y.; Badary, O.A. Effects of TNF-α antagonist infliximab on fructose-induced metabolic syndrome in rats. Hum. Exp. Toxicol. 2021, 40, 801–811.
  51. Shi, J.Q.; Shen, W.; Chen, J.; Wang, B.R.; Zhong, L.L.; Zhu, Y.W.; Zhu, H.-Q.; Zhang, Q.-Q.; Zhang, Y.-D.; Xu, J. Anti-TNF-α reduces amyloid plaques and tau phosphorylation and induces CD11c-positive dendritic-like cell in the APP/PS1 transgenic mouse brains. Brain Res. 2011, 1368, 239–247.
  52. Xu, J.J.; Guo, S.; Xue, R.; Xiao, L.; Kou, J.N.; Liu, Y.Q.; Han, J.-Y.; Fu, J.-J.; Wei, N. Adalimumab ameliorates memory impairments and neuroinflammation in chronic cerebral hypoperfusion rats. Aging 2021, 13, 14001–14014.
  53. Chou, R.C.; Kane, M.; Ghimire, S.; Gautam, S.; Gui, J. Treatment for Rheumatoid Arthritis and Risk of Alzheimer’s Disease: A Nested Case-Control Analysis. CNS Drugs 2016, 30, 1111–1120.
  54. Zhou, M.; Xu, R.; Kaelber, D.C.; Gurney, M.E. Tumor Necrosis Factor (TNF) blocking agents are associated with lower risk for Alzheimer’s disease in patients with rheumatoid arthritis and psoriasis. PLoS ONE 2020, 15, e0229819.
  55. Zhao, J.; Li, T.; Wang, J. Association between psoriasis and dementia: A systematic review. Neurologia 2021. Online ahead of print.
  56. Trzeciak, P.; Herbet, M.; Dudka, J. Common Factors of Alzheimer’s Disease and Rheumatoid Arthritis—Pathomechanism and Treatment. Molecules 2021, 26, 6038.
  57. Lin, T.M.; Chen, W.S.; Sheu, J.J.; Chen, Y.H.; Chen, J.H.; Chang, C.C. Autoimmune rheumatic diseases increase dementia risk in middle-aged patients: A nationwide cohort study. PLoS ONE 2018, 13, e0186475.
  58. Jin, L.W.; Lucente, J.D.; Nguyen, H.M.; Singh, V.; Singh, L.; Chavez, M.; Bushong, T.; Wulff, H.; Maezawa, I. Repurposing the KCa3.1 inhibitor senicapoc for Alzheimer’s disease. Ann. Clin. Transl. Neurol. 2019, 6, 723–738.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 294
Entry Collection: Neurodegeneration
Revisions: 3 times (View History)
Update Date: 09 Jan 2023
1000/1000