Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2021

Open Access 01-12-2021 | Etanercept | Research

Biologic TNF-α inhibitors reduce microgliosis, neuronal loss, and tau phosphorylation in a transgenic mouse model of tauopathy

Authors: Weijun Ou, Joshua Yang, Juste Simanauskaite, Matthew Choi, Demi M. Castellanos, Rudy Chang, Jiahong Sun, Nataraj Jagadeesan, Karen D. Parfitt, David H. Cribbs, Rachita K. Sumbria

Published in: Journal of Neuroinflammation | Issue 1/2021

Login to get access

Abstract

Background

Tumor necrosis factor-α (TNF-α) plays a central role in Alzheimer’s disease (AD) pathology, making biologic TNF-α inhibitors (TNFIs), including etanercept, viable therapeutics for AD. The protective effects of biologic TNFIs on AD hallmark pathology (Aβ deposition and tau pathology) have been demonstrated. However, the effects of biologic TNFIs on Aβ-independent tau pathology have not been reported. Existing biologic TNFIs do not cross the blood–brain barrier (BBB), therefore we engineered a BBB-penetrating biologic TNFI by fusing the extracellular domain of the type-II human TNF-α receptor (TNFR) to a transferrin receptor antibody (TfRMAb) that ferries the TNFR into the brain via receptor-mediated transcytosis. The present study aimed to investigate the effects of TfRMAb-TNFR (BBB-penetrating TNFI) and etanercept (non-BBB-penetrating TNFI) in the PS19 transgenic mouse model of tauopathy.

Methods

Six-month-old male and female PS19 mice were injected intraperitoneally with saline (n = 12), TfRMAb-TNFR (1.75 mg/kg, n = 10) or etanercept (0.875 mg/kg, equimolar dose of TNFR, n = 10) 3 days/week for 8 weeks. Age-matched littermate wild-type mice served as additional controls. Blood was collected at baseline and 8 weeks for a complete blood count. Locomotion hyperactivity was assessed by the open-field paradigm. Brains were examined for phosphorylated tau lesions (Ser202, Thr205), microgliosis, and neuronal health. The plasma pharmacokinetics were evaluated following a single intraperitoneal injection of 0.875 mg/kg etanercept or 1.75 mg/kg TfRMAb-TNFR or 1.75 mg/kg chronic TfRMAb-TNFR dosing for 4 weeks.

Results

Etanercept significantly reduced phosphorylated tau and microgliosis in the PS19 mouse brains of both sexes, while TfRMAb-TNFR significantly reduced these parameters in the female PS19 mice. Both TfRMAb-TNFR and etanercept treatment improved neuronal health by significantly increasing PSD95 expression and attenuating hippocampal neuron loss in the PS19 mice. The locomotion hyperactivity in the male PS19 mice was suppressed by chronic etanercept treatment. Equimolar dosing resulted in eightfold lower plasma exposure of the TfRMAb-TNFR compared with etanercept. The hematological profiles remained largely stable following chronic biologic TNFI dosing except for a significant increase in platelets with etanercept.

Conclusion

Both TfRMAb-TNFR (BBB-penetrating) and non-BBB-penetrating (etanercept) biologic TNFIs showed therapeutic effects in the PS19 mouse model of tauopathy.
Appendix
Available only for authorised users
Literature
3.
go back to reference Spillantini MG, Goedert M. Tau pathology and neurodegeneration. Lancet Neurol. 2013;12(6):609–22.PubMedCrossRef Spillantini MG, Goedert M. Tau pathology and neurodegeneration. Lancet Neurol. 2013;12(6):609–22.PubMedCrossRef
4.
go back to reference Parbo P, Ismail R, Sommerauer M, Stokholm MG, Hansen AK, Hansen KV, et al. Does inflammation precede tau aggregation in early Alzheimer’s disease? A PET study. Neurobiol Dis. 2018;117:211–6.PubMedCrossRef Parbo P, Ismail R, Sommerauer M, Stokholm MG, Hansen AK, Hansen KV, et al. Does inflammation precede tau aggregation in early Alzheimer’s disease? A PET study. Neurobiol Dis. 2018;117:211–6.PubMedCrossRef
5.
go back to reference Wyss-Coray T. Inflammation in Alzheimer disease: driving force, bystander or beneficial response? Nat Med. 2006;12(9):1005–15.PubMed Wyss-Coray T. Inflammation in Alzheimer disease: driving force, bystander or beneficial response? Nat Med. 2006;12(9):1005–15.PubMed
6.
go back to reference Tarkowski E, Andreasen N, Tarkowski A, Blennow K. Intrathecal inflammation precedes development of Alzheimer’s disease. J Neurol Neurosurg Psychiatry. 2003;74(9):1200–5.PubMedPubMedCentralCrossRef Tarkowski E, Andreasen N, Tarkowski A, Blennow K. Intrathecal inflammation precedes development of Alzheimer’s disease. J Neurol Neurosurg Psychiatry. 2003;74(9):1200–5.PubMedPubMedCentralCrossRef
7.
go back to reference Zelova H, Hosek J. TNF-alpha signalling and inflammation: interactions between old acquaintances. Inflamm Res. 2013;62(7):641–51.PubMedCrossRef Zelova H, Hosek J. TNF-alpha signalling and inflammation: interactions between old acquaintances. Inflamm Res. 2013;62(7):641–51.PubMedCrossRef
8.
go back to reference Alvarez A, Cacabelos R, Sanpedro C, Garcia-Fantini M, Aleixandre M. Serum TNF-alpha levels are increased and correlate negatively with free IGF-I in Alzheimer disease. Neurobiol Aging. 2007;28(4):533–6.PubMedCrossRef Alvarez A, Cacabelos R, Sanpedro C, Garcia-Fantini M, Aleixandre M. Serum TNF-alpha levels are increased and correlate negatively with free IGF-I in Alzheimer disease. Neurobiol Aging. 2007;28(4):533–6.PubMedCrossRef
9.
go back to reference Paganelli R, Di Iorio A, Patricelli L, Ripani F, Sparvieri E, Faricelli R, et al. Proinflammatory cytokines in sera of elderly patients with dementia: levels in vascular injury are higher than those of mild-moderate Alzheimer’s disease patients. Exp Gerontol. 2002;37(2–3):257–63.PubMedCrossRef Paganelli R, Di Iorio A, Patricelli L, Ripani F, Sparvieri E, Faricelli R, et al. Proinflammatory cytokines in sera of elderly patients with dementia: levels in vascular injury are higher than those of mild-moderate Alzheimer’s disease patients. Exp Gerontol. 2002;37(2–3):257–63.PubMedCrossRef
10.
11.
go back to reference Patel NS, Paris D, Mathura V, Quadros AN, Crawford FC, Mullan MJ. Inflammatory cytokine levels correlate with amyloid load in transgenic mouse models of Alzheimer’s disease. J Neuroinflammation. 2005;2(1):9.PubMedPubMedCentralCrossRef Patel NS, Paris D, Mathura V, Quadros AN, Crawford FC, Mullan MJ. Inflammatory cytokine levels correlate with amyloid load in transgenic mouse models of Alzheimer’s disease. J Neuroinflammation. 2005;2(1):9.PubMedPubMedCentralCrossRef
12.
go back to reference He P, Zhong Z, Lindholm K, Berning L, Lee W, Lemere C, et al. Deletion of tumor necrosis factor death receptor inhibits amyloid beta generation and prevents learning and memory deficits in Alzheimer’s mice. J Cell Biol. 2007;178(5):829–41.PubMedPubMedCentralCrossRef He P, Zhong Z, Lindholm K, Berning L, Lee W, Lemere C, et al. Deletion of tumor necrosis factor death receptor inhibits amyloid beta generation and prevents learning and memory deficits in Alzheimer’s mice. J Cell Biol. 2007;178(5):829–41.PubMedPubMedCentralCrossRef
13.
go back to reference Yamamoto M, Kiyota T, Horiba M, Buescher JL, Walsh SM, Gendelman HE, et al. Interferon-gamma and tumor necrosis factor-alpha regulate amyloid-beta plaque deposition and beta-secretase expression in Swedish mutant APP transgenic mice. Am J Pathol. 2007;170(2):680–92.PubMedPubMedCentralCrossRef Yamamoto M, Kiyota T, Horiba M, Buescher JL, Walsh SM, Gendelman HE, et al. Interferon-gamma and tumor necrosis factor-alpha regulate amyloid-beta plaque deposition and beta-secretase expression in Swedish mutant APP transgenic mice. Am J Pathol. 2007;170(2):680–92.PubMedPubMedCentralCrossRef
14.
go back to reference Liao YF, Wang BJ, Cheng HT, Kuo LH, Wolfe MS. Tumor necrosis factor-alpha, interleukin-1beta, and interferon-gamma stimulate gamma-secretase-mediated cleavage of amyloid precursor protein through a JNK-dependent MAPK pathway. J Biol Chem. 2004;279(47):49523–32.PubMedCrossRef Liao YF, Wang BJ, Cheng HT, Kuo LH, Wolfe MS. Tumor necrosis factor-alpha, interleukin-1beta, and interferon-gamma stimulate gamma-secretase-mediated cleavage of amyloid precursor protein through a JNK-dependent MAPK pathway. J Biol Chem. 2004;279(47):49523–32.PubMedCrossRef
15.
go back to reference Janelsins MC, Mastrangelo MA, Park KM, Sudol KL, Narrow WC, Oddo S, et al. Chronic neuron-specific tumor necrosis factor-alpha expression enhances the local inflammatory environment ultimately leading to neuronal death in 3xTg-AD mice. Am J Pathol. 2008;173(6):1768–82.PubMedPubMedCentralCrossRef Janelsins MC, Mastrangelo MA, Park KM, Sudol KL, Narrow WC, Oddo S, et al. Chronic neuron-specific tumor necrosis factor-alpha expression enhances the local inflammatory environment ultimately leading to neuronal death in 3xTg-AD mice. Am J Pathol. 2008;173(6):1768–82.PubMedPubMedCentralCrossRef
16.
go back to reference Ralay Ranaivo H, Craft JM, Hu W, Guo L, Wing LK, Van Eldik LJ, et al. Glia as a therapeutic target: selective suppression of human amyloid-beta-induced upregulation of brain proinflammatory cytokine production attenuates neurodegeneration. J Neurosci. 2006;26(2):662–70.PubMedCrossRef Ralay Ranaivo H, Craft JM, Hu W, Guo L, Wing LK, Van Eldik LJ, et al. Glia as a therapeutic target: selective suppression of human amyloid-beta-induced upregulation of brain proinflammatory cytokine production attenuates neurodegeneration. J Neurosci. 2006;26(2):662–70.PubMedCrossRef
17.
go back to reference Rosenberg PB. Clinical aspects of inflammation in Alzheimer’s disease. Int Rev Psychiatry. 2005;17(6):503–14.PubMedCrossRef Rosenberg PB. Clinical aspects of inflammation in Alzheimer’s disease. Int Rev Psychiatry. 2005;17(6):503–14.PubMedCrossRef
18.
go back to reference McGeer EG, McGeer PL. Inflammatory processes in Alzheimer’s disease. Prog Neuropsychopharmacol Biol Psychiatry. 2003;27(5):741–9.PubMedCrossRef McGeer EG, McGeer PL. Inflammatory processes in Alzheimer’s disease. Prog Neuropsychopharmacol Biol Psychiatry. 2003;27(5):741–9.PubMedCrossRef
19.
go back to reference Paouri E, Tzara O, Kartalou GI, Zenelak S, Georgopoulos S. Peripheral tumor necrosis factor-alpha (TNF-α) modulates amyloid pathology by regulating blood-derived immune cells and glial response in the brain of AD/TNF transgenic mice. J Neurosci. 2017;37(20):5155–71.PubMedPubMedCentralCrossRef Paouri E, Tzara O, Kartalou GI, Zenelak S, Georgopoulos S. Peripheral tumor necrosis factor-alpha (TNF-α) modulates amyloid pathology by regulating blood-derived immune cells and glial response in the brain of AD/TNF transgenic mice. J Neurosci. 2017;37(20):5155–71.PubMedPubMedCentralCrossRef
20.
go back to reference Kalovyrna N, Apokotou O, Boulekou S, Paouri E, Boutou A, Georgopoulos S. A 3’UTR modification of the TNF-α mouse gene increases peripheral TNF-α and modulates the Alzheimer-like phenotype in 5XFAD mice. Sci Rep. 2020;10(1):8670.PubMedPubMedCentralCrossRef Kalovyrna N, Apokotou O, Boulekou S, Paouri E, Boutou A, Georgopoulos S. A 3’UTR modification of the TNF-α mouse gene increases peripheral TNF-α and modulates the Alzheimer-like phenotype in 5XFAD mice. Sci Rep. 2020;10(1):8670.PubMedPubMedCentralCrossRef
21.
go back to reference Collins JS, Perry RT, Watson B Jr, Harrell LE, Acton RT, Blacker D, et al. Association of a haplotype for tumor necrosis factor in siblings with late-onset Alzheimer disease: the NIMH Alzheimer disease genetics initiative. Am J Med Genet. 2000;96(6):823–30.PubMedCrossRef Collins JS, Perry RT, Watson B Jr, Harrell LE, Acton RT, Blacker D, et al. Association of a haplotype for tumor necrosis factor in siblings with late-onset Alzheimer disease: the NIMH Alzheimer disease genetics initiative. Am J Med Genet. 2000;96(6):823–30.PubMedCrossRef
22.
go back to reference Shi JQ, Shen W, Chen J, Wang BR, Zhong LL, Zhu YW, et al. 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;1368:239–47.CrossRef Shi JQ, Shen W, Chen J, Wang BR, Zhong LL, Zhu YW, et al. 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;1368:239–47.CrossRef
23.
go back to reference Shi JQ, Wang BR, Jiang WW, Chen J, Zhu YW, Zhong LL, et al. Cognitive improvement with intrathecal administration of infliximab in a woman with Alzheimer’s disease. J Am Geriatr Soc. 2011;59(6):1142–4.PubMedCrossRef Shi JQ, Wang BR, Jiang WW, Chen J, Zhu YW, Zhong LL, et al. Cognitive improvement with intrathecal administration of infliximab in a woman with Alzheimer’s disease. J Am Geriatr Soc. 2011;59(6):1142–4.PubMedCrossRef
24.
go back to reference Kim DH, Choi SM, Jho J, Park MS, Kang J, Park SJ, et al. Infliximab ameliorates AD-associated object recognition memory impairment. Behav Brain Res. 2016;311:384–91.PubMedCrossRef Kim DH, Choi SM, Jho J, Park MS, Kang J, Park SJ, et al. Infliximab ameliorates AD-associated object recognition memory impairment. Behav Brain Res. 2016;311:384–91.PubMedCrossRef
25.
go back to reference Tobinick E, Gross H, Weinberger A, Cohen H. TNF-alpha modulation for treatment of Alzheimer’s disease: a 6-month pilot study. MedGenMed. 2006;8(2):25.PubMedPubMedCentral Tobinick E, Gross H, Weinberger A, Cohen H. TNF-alpha modulation for treatment of Alzheimer’s disease: a 6-month pilot study. MedGenMed. 2006;8(2):25.PubMedPubMedCentral
26.
go back to reference McAlpine FE, Lee JK, Harms AS, Ruhn KA, Blurton-Jones M, Hong J, et al. Inhibition of soluble TNF signaling in a mouse model of Alzheimer’s disease prevents pre-plaque amyloid-associated neuropathology. Neurobiol Dis. 2009;34(1):163–77.PubMedPubMedCentralCrossRef McAlpine FE, Lee JK, Harms AS, Ruhn KA, Blurton-Jones M, Hong J, et al. Inhibition of soluble TNF signaling in a mouse model of Alzheimer’s disease prevents pre-plaque amyloid-associated neuropathology. Neurobiol Dis. 2009;34(1):163–77.PubMedPubMedCentralCrossRef
27.
go back to reference Steeland S, Gorlé N, Vandendriessche C, Balusu S, Brkic M, Van Cauwenberghe C, et al. Counteracting the effects of TNF receptor-1 has therapeutic potential in Alzheimer's disease. EMBO Mol Med. 2018;10(4). Steeland S, Gorlé N, Vandendriessche C, Balusu S, Brkic M, Van Cauwenberghe C, et al. Counteracting the effects of TNF receptor-1 has therapeutic potential in Alzheimer's disease. EMBO Mol Med. 2018;10(4).
28.
go back to reference Boado RJ, Hui EK, Lu JZ, Zhou QH, Pardridge WM. Selective targeting of a TNFR decoy receptor pharmaceutical to the primate brain as a receptor-specific IgG fusion protein. J Biotechnol. 2010;146(1–2):84–91.PubMedPubMedCentralCrossRef Boado RJ, Hui EK, Lu JZ, Zhou QH, Pardridge WM. Selective targeting of a TNFR decoy receptor pharmaceutical to the primate brain as a receptor-specific IgG fusion protein. J Biotechnol. 2010;146(1–2):84–91.PubMedPubMedCentralCrossRef
29.
30.
31.
go back to reference Yoshiyama Y, Higuchi M, Zhang B, Huang SM, Iwata N, Saido TC, et al. Synapse loss and microglial activation precede tangles in a P301S tauopathy mouse model. Neuron. 2007;53(3):337–51.PubMedCrossRef Yoshiyama Y, Higuchi M, Zhang B, Huang SM, Iwata N, Saido TC, et al. Synapse loss and microglial activation precede tangles in a P301S tauopathy mouse model. Neuron. 2007;53(3):337–51.PubMedCrossRef
32.
go back to reference Maphis N, Xu G, Kokiko-Cochran ON, Jiang S, Cardona A, Ransohoff RM, et al. Reactive microglia drive tau pathology and contribute to the spreading of pathological tau in the brain. Brain. 2015;138(Pt 6):1738–55.PubMedPubMedCentralCrossRef Maphis N, Xu G, Kokiko-Cochran ON, Jiang S, Cardona A, Ransohoff RM, et al. Reactive microglia drive tau pathology and contribute to the spreading of pathological tau in the brain. Brain. 2015;138(Pt 6):1738–55.PubMedPubMedCentralCrossRef
33.
34.
go back to reference Yoshiyama Y, Kojima A, Ishikawa C, Arai K. Anti-inflammatory action of donepezil ameliorates tau pathology, synaptic loss, and neurodegeneration in a tauopathy mouse model. J Alzheimers Dis. 2010;22(1):295–306.PubMedCrossRef Yoshiyama Y, Kojima A, Ishikawa C, Arai K. Anti-inflammatory action of donepezil ameliorates tau pathology, synaptic loss, and neurodegeneration in a tauopathy mouse model. J Alzheimers Dis. 2010;22(1):295–306.PubMedCrossRef
35.
go back to reference Sumbria RK, Zhou QH, Hui EK, Lu JZ, Boado RJ, Pardridge WM. Pharmacokinetics and brain uptake of an IgG-TNF decoy receptor fusion protein following intravenous, intraperitoneal, and subcutaneous administration in mice. Mol Pharm. 2013;10(4):1425–31.PubMedPubMedCentralCrossRef Sumbria RK, Zhou QH, Hui EK, Lu JZ, Boado RJ, Pardridge WM. Pharmacokinetics and brain uptake of an IgG-TNF decoy receptor fusion protein following intravenous, intraperitoneal, and subcutaneous administration in mice. Mol Pharm. 2013;10(4):1425–31.PubMedPubMedCentralCrossRef
36.
go back to reference Zhou QH, Boado RJ, Hui EK, Lu JZ, Pardridge WM. Brain-penetrating tumor necrosis factor decoy receptor in the mouse. Drug Metab Dispos. 2011;39(1):71–6.PubMedPubMedCentralCrossRef Zhou QH, Boado RJ, Hui EK, Lu JZ, Pardridge WM. Brain-penetrating tumor necrosis factor decoy receptor in the mouse. Drug Metab Dispos. 2011;39(1):71–6.PubMedPubMedCentralCrossRef
37.
go back to reference Chang R, Knox J, Chang J, Derbedrossian A, Vasilevko V, Cribbs D, et al. Blood–brain barrier penetrating biologic TNF-α inhibitor for Alzheimer’s disease. Mol Pharm. 2017;14(7):2340–9.PubMedCrossRef Chang R, Knox J, Chang J, Derbedrossian A, Vasilevko V, Cribbs D, et al. Blood–brain barrier penetrating biologic TNF-α inhibitor for Alzheimer’s disease. Mol Pharm. 2017;14(7):2340–9.PubMedCrossRef
38.
go back to reference Hassett B, McMillen S, Fitzpatrick B. Characterization and comparison of commercially available TNF receptor 2-Fc fusion protein products: letter to the editor. MAbs. 2013;5(5):624–5.PubMedPubMedCentralCrossRef Hassett B, McMillen S, Fitzpatrick B. Characterization and comparison of commercially available TNF receptor 2-Fc fusion protein products: letter to the editor. MAbs. 2013;5(5):624–5.PubMedPubMedCentralCrossRef
39.
go back to reference Dumont M, Stack C, Elipenahli C, Jainuddin S, Gerges M, Starkova NN, et al. Behavioral deficit, oxidative stress, and mitochondrial dysfunction precede tau pathology in P301S transgenic mice. Faseb j. 2011;25(11):4063–72.PubMedPubMedCentralCrossRef Dumont M, Stack C, Elipenahli C, Jainuddin S, Gerges M, Starkova NN, et al. Behavioral deficit, oxidative stress, and mitochondrial dysfunction precede tau pathology in P301S transgenic mice. Faseb j. 2011;25(11):4063–72.PubMedPubMedCentralCrossRef
40.
go back to reference Takeuchi H, Iba M, Inoue H, Higuchi M, Takao K, Tsukita K, et al. P301S mutant human tau transgenic mice manifest early symptoms of human tauopathies with dementia and altered sensorimotor gating. PLoS ONE. 2011;6(6):e21050.PubMedPubMedCentralCrossRef Takeuchi H, Iba M, Inoue H, Higuchi M, Takao K, Tsukita K, et al. P301S mutant human tau transgenic mice manifest early symptoms of human tauopathies with dementia and altered sensorimotor gating. PLoS ONE. 2011;6(6):e21050.PubMedPubMedCentralCrossRef
41.
go back to reference Patel H, Martinez P, Perkins A, Taylor X, Jury N, McKinzie D, et al. Pathological tau and reactive astrogliosis are associated with distinct functional deficits in a mouse model of tauopathy. Neurobiol Aging. 2022;109:52–63.PubMedCrossRef Patel H, Martinez P, Perkins A, Taylor X, Jury N, McKinzie D, et al. Pathological tau and reactive astrogliosis are associated with distinct functional deficits in a mouse model of tauopathy. Neurobiol Aging. 2022;109:52–63.PubMedCrossRef
42.
go back to reference Chang R, Knox J, Chang J, Derbedrossian A, Vasilevko V, Cribbs D, et al. Blood–brain barrier penetrating biologic TNF-alpha inhibitor for Alzheimer’s disease. Mol Pharm. 2017;14(7):2340–9.PubMedCrossRef Chang R, Knox J, Chang J, Derbedrossian A, Vasilevko V, Cribbs D, et al. Blood–brain barrier penetrating biologic TNF-alpha inhibitor for Alzheimer’s disease. Mol Pharm. 2017;14(7):2340–9.PubMedCrossRef
43.
go back to reference Chang R, Al Maghribi A, Vanderpoel V, Vasilevko V, Cribbs DH, Boado R, et al. Brain penetrating bifunctional erythropoietin-transferrin receptor antibody fusion protein for Alzheimer’s disease. Mol Pharm. 2018;15(11):4963–73.PubMedPubMedCentralCrossRef Chang R, Al Maghribi A, Vanderpoel V, Vasilevko V, Cribbs DH, Boado R, et al. Brain penetrating bifunctional erythropoietin-transferrin receptor antibody fusion protein for Alzheimer’s disease. Mol Pharm. 2018;15(11):4963–73.PubMedPubMedCentralCrossRef
44.
go back to reference Sun J, Yang J, Whitman K, Zhu C, Cribbs DH, Boado RJ, et al. Hematologic safety of chronic brain-penetrating erythropoietin dosing in APP/PS1 mice. Alzheimers Dement (N Y). 2019;5:627–36.CrossRef Sun J, Yang J, Whitman K, Zhu C, Cribbs DH, Boado RJ, et al. Hematologic safety of chronic brain-penetrating erythropoietin dosing in APP/PS1 mice. Alzheimers Dement (N Y). 2019;5:627–36.CrossRef
45.
go back to reference Pan B, Zhang H, Cui T, Wang X. TFEB activation protects against cardiac proteotoxicity via increasing autophagic flux. J Mol Cell Cardiol. 2017;113:51–62.PubMedPubMedCentralCrossRef Pan B, Zhang H, Cui T, Wang X. TFEB activation protects against cardiac proteotoxicity via increasing autophagic flux. J Mol Cell Cardiol. 2017;113:51–62.PubMedPubMedCentralCrossRef
46.
go back to reference Pan B, Li J, Parajuli N, Tian Z, Wu P, Lewno MT, et al. The calcineurin-TFEB-p62 pathway mediates the activation of cardiac macroautophagy by proteasomal malfunction. Circ Res. 2020;127(4):502–18.PubMedPubMedCentralCrossRef Pan B, Li J, Parajuli N, Tian Z, Wu P, Lewno MT, et al. The calcineurin-TFEB-p62 pathway mediates the activation of cardiac macroautophagy by proteasomal malfunction. Circ Res. 2020;127(4):502–18.PubMedPubMedCentralCrossRef
47.
go back to reference Castellanos DM, Sun J, Yang J, Ou W, Zambon AC, Pardridge WM, et al. Acute and chronic dosing of a high-affinity rat/mouse chimeric transferrin receptor antibody in mice. Pharmaceutics. 2020;12(9):852.PubMedCentralCrossRef Castellanos DM, Sun J, Yang J, Ou W, Zambon AC, Pardridge WM, et al. Acute and chronic dosing of a high-affinity rat/mouse chimeric transferrin receptor antibody in mice. Pharmaceutics. 2020;12(9):852.PubMedCentralCrossRef
48.
go back to reference Yang J, Sun J, Castellanos DM, Pardridge WM, Sumbria RK. Eliminating Fc N-linked glycosylation and its impact on dosing consideration for a transferrin receptor antibody-erythropoietin fusion protein in mice. Mol Pharm. 2020;17(8):2831–9.PubMedPubMedCentralCrossRef Yang J, Sun J, Castellanos DM, Pardridge WM, Sumbria RK. Eliminating Fc N-linked glycosylation and its impact on dosing consideration for a transferrin receptor antibody-erythropoietin fusion protein in mice. Mol Pharm. 2020;17(8):2831–9.PubMedPubMedCentralCrossRef
49.
go back to reference Sun J, Boado RJ, Pardridge WM, Sumbria RK. Plasma pharmacokinetics of high-affinity transferrin receptor antibody-erythropoietin fusion protein is a function of effector attenuation in mice. Mol Pharm. 2019;16(8):3534–43.PubMedPubMedCentralCrossRef Sun J, Boado RJ, Pardridge WM, Sumbria RK. Plasma pharmacokinetics of high-affinity transferrin receptor antibody-erythropoietin fusion protein is a function of effector attenuation in mice. Mol Pharm. 2019;16(8):3534–43.PubMedPubMedCentralCrossRef
50.
go back to reference Pan B, Lewno MT, Wu P, Wang X. Highly dynamic changes in the activity and regulation of macroautophagy in hearts subjected to increased proteotoxic stress. Front Physiol. 2019;10:758.PubMedPubMedCentralCrossRef Pan B, Lewno MT, Wu P, Wang X. Highly dynamic changes in the activity and regulation of macroautophagy in hearts subjected to increased proteotoxic stress. Front Physiol. 2019;10:758.PubMedPubMedCentralCrossRef
51.
go back to reference Sheffield LG, Marquis JG, Berman NE. Regional distribution of cortical microglia parallels that of neurofibrillary tangles in Alzheimer’s disease. Neurosci Lett. 2000;285(3):165–8.PubMedCrossRef Sheffield LG, Marquis JG, Berman NE. Regional distribution of cortical microglia parallels that of neurofibrillary tangles in Alzheimer’s disease. Neurosci Lett. 2000;285(3):165–8.PubMedCrossRef
52.
go back to reference Ishizawa K, Dickson DW. Microglial activation parallels system degeneration in progressive supranuclear palsy and corticobasal degeneration. J Neuropathol Exp Neurol. 2001;60(6):647–57.PubMedCrossRef Ishizawa K, Dickson DW. Microglial activation parallels system degeneration in progressive supranuclear palsy and corticobasal degeneration. J Neuropathol Exp Neurol. 2001;60(6):647–57.PubMedCrossRef
54.
go back to reference Zilka N, Kazmerova Z, Jadhav S, Neradil P, Madari A, Obetkova D, et al. Who fans the flames of Alzheimer’s disease brains? Misfolded tau on the crossroad of neurodegenerative and inflammatory pathways. J Neuroinflammation. 2012;9:47.PubMedPubMedCentralCrossRef Zilka N, Kazmerova Z, Jadhav S, Neradil P, Madari A, Obetkova D, et al. Who fans the flames of Alzheimer’s disease brains? Misfolded tau on the crossroad of neurodegenerative and inflammatory pathways. J Neuroinflammation. 2012;9:47.PubMedPubMedCentralCrossRef
55.
go back to reference Quintanilla RA, Orellana DI, Gonzalez-Billault C, Maccioni RB. Interleukin-6 induces Alzheimer-type phosphorylation of tau protein by deregulating the cdk5/p35 pathway. Exp Cell Res. 2004;295(1):245–57.PubMedCrossRef Quintanilla RA, Orellana DI, Gonzalez-Billault C, Maccioni RB. Interleukin-6 induces Alzheimer-type phosphorylation of tau protein by deregulating the cdk5/p35 pathway. Exp Cell Res. 2004;295(1):245–57.PubMedCrossRef
56.
go back to reference Li Y, Liu L, Barger SW, Griffin WS. Interleukin-1 mediates pathological effects of microglia on tau phosphorylation and on synaptophysin synthesis in cortical neurons through a p38-MAPK pathway. J Neurosci. 2003;23(5):1605–11.PubMedPubMedCentralCrossRef Li Y, Liu L, Barger SW, Griffin WS. Interleukin-1 mediates pathological effects of microglia on tau phosphorylation and on synaptophysin synthesis in cortical neurons through a p38-MAPK pathway. J Neurosci. 2003;23(5):1605–11.PubMedPubMedCentralCrossRef
57.
go back to reference Sheng JG, Zhu SG, Jones RA, Griffin WS, Mrak RE. Interleukin-1 promotes expression and phosphorylation of neurofilament and tau proteins in vivo. Exp Neurol. 2000;163(2):388–91.PubMedCrossRef Sheng JG, Zhu SG, Jones RA, Griffin WS, Mrak RE. Interleukin-1 promotes expression and phosphorylation of neurofilament and tau proteins in vivo. Exp Neurol. 2000;163(2):388–91.PubMedCrossRef
58.
go back to reference Gabbita SP, Johnson MF, Kobritz N, Eslami P, Poteshkina A, Varadarajan S, et al. Oral TNFα modulation alters neutrophil infiltration, improves cognition and diminishes tau and amyloid pathology in the 3xTgAD mouse model. PLoS ONE. 2015;10(10):e0137305.PubMedPubMedCentralCrossRef Gabbita SP, Johnson MF, Kobritz N, Eslami P, Poteshkina A, Varadarajan S, et al. Oral TNFα modulation alters neutrophil infiltration, improves cognition and diminishes tau and amyloid pathology in the 3xTgAD mouse model. PLoS ONE. 2015;10(10):e0137305.PubMedPubMedCentralCrossRef
59.
go back to reference Mancuso R, Fryatt G, Cleal M, Obst J, Pipi E, Monzon-Sandoval J, et al. CSF1R inhibitor JNJ-40346527 attenuates microglial proliferation and neurodegeneration in P301S mice. Brain. 2019;142(10):3243–64.PubMedPubMedCentralCrossRef Mancuso R, Fryatt G, Cleal M, Obst J, Pipi E, Monzon-Sandoval J, et al. CSF1R inhibitor JNJ-40346527 attenuates microglial proliferation and neurodegeneration in P301S mice. Brain. 2019;142(10):3243–64.PubMedPubMedCentralCrossRef
60.
go back to reference DiPatre PL, Gelman BB. Microglial cell activation in aging and Alzheimer disease: partial linkage with neurofibrillary tangle burden in the hippocampus. J Neuropathol Exp Neurol. 1997;56(2):143–9.PubMedCrossRef DiPatre PL, Gelman BB. Microglial cell activation in aging and Alzheimer disease: partial linkage with neurofibrillary tangle burden in the hippocampus. J Neuropathol Exp Neurol. 1997;56(2):143–9.PubMedCrossRef
61.
go back to reference Bellucci A, Westwood AJ, Ingram E, Casamenti F, Goedert M, Spillantini MG. Induction of inflammatory mediators and microglial activation in mice transgenic for mutant human P301S tau protein. Am J Pathol. 2004;165(5):1643–52.PubMedPubMedCentralCrossRef Bellucci A, Westwood AJ, Ingram E, Casamenti F, Goedert M, Spillantini MG. Induction of inflammatory mediators and microglial activation in mice transgenic for mutant human P301S tau protein. Am J Pathol. 2004;165(5):1643–52.PubMedPubMedCentralCrossRef
62.
go back to reference Paasila PJ, Davies DS, Kril JJ, Goldsbury C, Sutherland GT. The relationship between the morphological subtypes of microglia and Alzheimer’s disease neuropathology. Brain Pathol. 2019;29(6):726–40.PubMedPubMedCentralCrossRef Paasila PJ, Davies DS, Kril JJ, Goldsbury C, Sutherland GT. The relationship between the morphological subtypes of microglia and Alzheimer’s disease neuropathology. Brain Pathol. 2019;29(6):726–40.PubMedPubMedCentralCrossRef
63.
64.
go back to reference Wang J, Zhao D, Pan B, Fu Y, Shi F, Kouadir M, et al. Toll-like receptor 2 deficiency shifts PrP106-126-induced microglial activation from a neurotoxic to a neuroprotective phenotype. J Mol Neurosci. 2015;55(4):880–90.PubMedCrossRef Wang J, Zhao D, Pan B, Fu Y, Shi F, Kouadir M, et al. Toll-like receptor 2 deficiency shifts PrP106-126-induced microglial activation from a neurotoxic to a neuroprotective phenotype. J Mol Neurosci. 2015;55(4):880–90.PubMedCrossRef
65.
go back to reference Kozlowski C, Weimer RM. An automated method to quantify microglia morphology and application to monitor activation state longitudinally in vivo. PLoS ONE. 2012;7(2):e31814.PubMedPubMedCentralCrossRef Kozlowski C, Weimer RM. An automated method to quantify microglia morphology and application to monitor activation state longitudinally in vivo. PLoS ONE. 2012;7(2):e31814.PubMedPubMedCentralCrossRef
66.
go back to reference Deczkowska A, Keren-Shaul H, Weiner A, Colonna M, Schwartz M, Amit I. Disease-associated microglia: a universal immune sensor of neurodegeneration. Cell. 2018;173(5):1073–81.PubMedCrossRef Deczkowska A, Keren-Shaul H, Weiner A, Colonna M, Schwartz M, Amit I. Disease-associated microglia: a universal immune sensor of neurodegeneration. Cell. 2018;173(5):1073–81.PubMedCrossRef
67.
go back to reference Nelson PT, Alafuzoff I, Bigio EH, Bouras C, Braak H, Cairns NJ, et al. Correlation of Alzheimer disease neuropathologic changes with cognitive status: a review of the literature. J Neuropathol Exp Neurol. 2012;71(5):362–81.PubMedCrossRef Nelson PT, Alafuzoff I, Bigio EH, Bouras C, Braak H, Cairns NJ, et al. Correlation of Alzheimer disease neuropathologic changes with cognitive status: a review of the literature. J Neuropathol Exp Neurol. 2012;71(5):362–81.PubMedCrossRef
68.
go back to reference Malpetti M, Kievit RA, Passamonti L, Jones PS, Tsvetanov KA, Rittman T, et al. Microglial activation and tau burden predict cognitive decline in Alzheimer’s disease. Brain. 2020;143(5):1588–602.PubMedPubMedCentralCrossRef Malpetti M, Kievit RA, Passamonti L, Jones PS, Tsvetanov KA, Rittman T, et al. Microglial activation and tau burden predict cognitive decline in Alzheimer’s disease. Brain. 2020;143(5):1588–602.PubMedPubMedCentralCrossRef
70.
go back to reference Bien-Ly N, Boswell CA, Jeet S, Beach TG, Hoyte K, Luk W, et al. Lack of widespread BBB disruption in Alzheimer’s disease models: focus on therapeutic antibodies. Neuron. 2015;88(2):289–97.PubMedCrossRef Bien-Ly N, Boswell CA, Jeet S, Beach TG, Hoyte K, Luk W, et al. Lack of widespread BBB disruption in Alzheimer’s disease models: focus on therapeutic antibodies. Neuron. 2015;88(2):289–97.PubMedCrossRef
71.
go back to reference Iwai K. Diverse ubiquitin signaling in NF-kappaB activation. Trends Cell Biol. 2012;22(7):355–64.PubMedCrossRef Iwai K. Diverse ubiquitin signaling in NF-kappaB activation. Trends Cell Biol. 2012;22(7):355–64.PubMedCrossRef
72.
go back to reference Zhou QH, Boado RJ, Hui EK, Lu JZ, Pardridge WM. Chronic dosing of mice with a transferrin receptor monoclonal antibody-glial-derived neurotrophic factor fusion protein. Drug Metab Dispos. 2011;39(7):1149–54.PubMedPubMedCentralCrossRef Zhou QH, Boado RJ, Hui EK, Lu JZ, Pardridge WM. Chronic dosing of mice with a transferrin receptor monoclonal antibody-glial-derived neurotrophic factor fusion protein. Drug Metab Dispos. 2011;39(7):1149–54.PubMedPubMedCentralCrossRef
73.
go back to reference Zhou H. Clinical pharmacokinetics of etanercept: a fully humanized soluble recombinant tumor necrosis factor receptor fusion protein. J Clin Pharmacol. 2005;45(5):490–7.PubMedCrossRef Zhou H. Clinical pharmacokinetics of etanercept: a fully humanized soluble recombinant tumor necrosis factor receptor fusion protein. J Clin Pharmacol. 2005;45(5):490–7.PubMedCrossRef
74.
go back to reference Korth-Bradley JM, Rubin AS, Hanna RK, Simcoe DK, Lebsack ME. The pharmacokinetics of etanercept in healthy volunteers. Ann Pharmacother. 2000;34(2):161–4.PubMedCrossRef Korth-Bradley JM, Rubin AS, Hanna RK, Simcoe DK, Lebsack ME. The pharmacokinetics of etanercept in healthy volunteers. Ann Pharmacother. 2000;34(2):161–4.PubMedCrossRef
75.
go back to reference Couch JA, Yu YJ, Zhang Y, Tarrant JM, Fuji RN, Meilandt WJ, et al. Addressing safety liabilities of TfR bispecific antibodies that cross the blood-brain barrier. Sci Transl Med. 2013;5(183):183ra57 (1–12).PubMedCrossRef Couch JA, Yu YJ, Zhang Y, Tarrant JM, Fuji RN, Meilandt WJ, et al. Addressing safety liabilities of TfR bispecific antibodies that cross the blood-brain barrier. Sci Transl Med. 2013;5(183):183ra57 (1–12).PubMedCrossRef
76.
go back to reference Hyrich KL, Silman AJ, Watson KD, Symmons DP. Anti-tumour necrosis factor alpha therapy in rheumatoid arthritis: an update on safety. Ann Rheum Dis. 2004;63(12):1538–43.PubMedPubMedCentralCrossRef Hyrich KL, Silman AJ, Watson KD, Symmons DP. Anti-tumour necrosis factor alpha therapy in rheumatoid arthritis: an update on safety. Ann Rheum Dis. 2004;63(12):1538–43.PubMedPubMedCentralCrossRef
77.
go back to reference Pathare SK, Heycock C, Hamilton J. TNFalpha blocker-induced thrombocytopenia. Rheumatology (Oxford). 2006;45(10):1313–4.CrossRef Pathare SK, Heycock C, Hamilton J. TNFalpha blocker-induced thrombocytopenia. Rheumatology (Oxford). 2006;45(10):1313–4.CrossRef
78.
go back to reference Propson NE, Roy ER, Litvinchuk A, Köhl J, Zheng H. Endothelial C3a receptor mediates vascular inflammation and blood–brain barrier permeability during aging. J Clin Invest. 2021;131(1). Propson NE, Roy ER, Litvinchuk A, Köhl J, Zheng H. Endothelial C3a receptor mediates vascular inflammation and blood–brain barrier permeability during aging. J Clin Invest. 2021;131(1).
79.
go back to reference Iba M, Guo JL, McBride JD, Zhang B, Trojanowski JQ, Lee VM. Synthetic tau fibrils mediate transmission of neurofibrillary tangles in a transgenic mouse model of Alzheimer’s-like tauopathy. J Neurosci. 2013;33(3):1024–37.PubMedPubMedCentralCrossRef Iba M, Guo JL, McBride JD, Zhang B, Trojanowski JQ, Lee VM. Synthetic tau fibrils mediate transmission of neurofibrillary tangles in a transgenic mouse model of Alzheimer’s-like tauopathy. J Neurosci. 2013;33(3):1024–37.PubMedPubMedCentralCrossRef
80.
go back to reference Pascoal TA, Benedet AL, Ashton NJ, Kang MS, Therriault J, Chamoun M, et al. Microglial activation and tau propagate jointly across Braak stages. Nat Med. 2021;27(9):1592–9.PubMedCrossRef Pascoal TA, Benedet AL, Ashton NJ, Kang MS, Therriault J, Chamoun M, et al. Microglial activation and tau propagate jointly across Braak stages. Nat Med. 2021;27(9):1592–9.PubMedCrossRef
Metadata
Title
Biologic TNF-α inhibitors reduce microgliosis, neuronal loss, and tau phosphorylation in a transgenic mouse model of tauopathy
Authors
Weijun Ou
Joshua Yang
Juste Simanauskaite
Matthew Choi
Demi M. Castellanos
Rudy Chang
Jiahong Sun
Nataraj Jagadeesan
Karen D. Parfitt
David H. Cribbs
Rachita K. Sumbria
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2021
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-021-02332-7

Other articles of this Issue 1/2021

Journal of Neuroinflammation 1/2021 Go to the issue