Skip to main content
Top
Published in: Molecular Brain 1/2021

Open Access 01-12-2021 | Central Nervous System Trauma | Research

iTRAQ-based proteomic profiling reveals protein alterations after traumatic brain injury and supports thyroxine as a potential treatment

Authors: Zhongxiang Zhang, Jiangtao Yu, Pengcheng Wang, Lian Lin, Ruining Liu, Rong Zeng, Haoli Ma, Yan Zhao

Published in: Molecular Brain | Issue 1/2021

Login to get access

Abstract

Traumatic brain injury (TBI) is a primary cause of disability and death across the world. Previously, RNA analysis was widely used to study the pathophysiological mechanisms underlying TBI; however, the relatively low correlation between the transcriptome and proteome revealed that RNA transcription abundance does not reliably predict protein abundance, which led to the emergence of proteomic research. In this study, an iTRAQ proteomics approach was applied to detect protein alterations after TBI on a large scale. A total of 3937 proteins were identified, and 146 proteins were significantly changed after TBI. Moreover, 23 upregulated proteins were verified by parallel reaction monitoring (PRM), and fold changes in 16 proteins were consistent with iTRAQ outcomes. Transthyretin (Ttr) upregulation has been demonstrated at the transcriptional level, and this study further confirmed this at the protein level. After treatment with thyroxine (T4), which is transported by Ttr, the effects of T4 on neuronal histopathology and behavioral performance were determined in vivo (TBI + T4 group). Brain edema was alleviated, and the integrity of the blood brain barrier (BBB) improved. Escape latency in the Morris water maze (MWM) declined significantly compared with the group without T4 treatment. Modified neurological severity scores (mNSS) of the TBI + T4 group decreased from day 1 to day 7 post-TBI compared with the TBI + saline group. These results indicate that T4 treatment has potential to alleviate pathologic and behavioral abnormalities post-TBI. Protein alterations after T4 treatment were also detected by iTRAQ proteomics. Upregulation of proteins like Lgals3, Gfap and Apoe after TBI were reversed by T4 treatment. GO enrichment showed T4 mainly affected intermediate filament organization, cholesterol transportation and axonal regeneration. In summary, iTRAQ proteomics provides information about the impact of TBI on protein alterations and yields insight into underlying mechanisms and pathways involved in TBI and T4 treatment. Finally, Ttr and other proteins identified by iTRAQ may become potential novel treatment targets post-TBI.
Appendix
Available only for authorised users
Literature
1.
go back to reference Schiller JS, Lucas JW, Ward BW, et al. Summary health statistics for U.S. adults: National Health Interview Survey, 2010. Vital Health Stat 10. 2012:1–207. Schiller JS, Lucas JW, Ward BW, et al. Summary health statistics for U.S. adults: National Health Interview Survey, 2010. Vital Health Stat 10. 2012:1–207.
2.
go back to reference Cheng P, Yin P, Ning P, et al. Trends in traumatic brain injury mortality in China, 2006–2013: a population-based longitudinal study. PLoS Med. 2017;14:e1002332.PubMedPubMedCentralCrossRef Cheng P, Yin P, Ning P, et al. Trends in traumatic brain injury mortality in China, 2006–2013: a population-based longitudinal study. PLoS Med. 2017;14:e1002332.PubMedPubMedCentralCrossRef
3.
go back to reference Roozenbeek B, Maas AI, Menon DK. Changing patterns in the epidemiology of traumatic brain injury. Nat Rev Neurol. 2013;9:231–6.PubMedCrossRef Roozenbeek B, Maas AI, Menon DK. Changing patterns in the epidemiology of traumatic brain injury. Nat Rev Neurol. 2013;9:231–6.PubMedCrossRef
4.
go back to reference Marin JR, Weaver MD, Mannix RC. Burden of USA hospital charges for traumatic brain injury. Brain Inj. 2017;31:24–31.PubMedCrossRef Marin JR, Weaver MD, Mannix RC. Burden of USA hospital charges for traumatic brain injury. Brain Inj. 2017;31:24–31.PubMedCrossRef
5.
go back to reference Leonard J, Garrett RE, Salottolo K, et al. Cerebral salt wasting after traumatic brain injury: a review of the literature. Scand J Trauma Resusc Emerg Med. 2015;23:98.PubMedPubMedCentralCrossRef Leonard J, Garrett RE, Salottolo K, et al. Cerebral salt wasting after traumatic brain injury: a review of the literature. Scand J Trauma Resusc Emerg Med. 2015;23:98.PubMedPubMedCentralCrossRef
6.
go back to reference Cheng G, Kong RH, Zhang LM, et al. Mitochondria in traumatic brain injury and mitochondrial-targeted multipotential therapeutic strategies. Br J Pharmacol. 2012;167:699–719.PubMedPubMedCentralCrossRef Cheng G, Kong RH, Zhang LM, et al. Mitochondria in traumatic brain injury and mitochondrial-targeted multipotential therapeutic strategies. Br J Pharmacol. 2012;167:699–719.PubMedPubMedCentralCrossRef
7.
go back to reference Campos-Pires R, Hirnet T, Valeo F, et al. Xenon improves long-term cognitive function, reduces neuronal loss and chronic neuroinflammation, and improves survival after traumatic brain injury in mice. Br J Anaesth. 2019;123:60–73.PubMedPubMedCentralCrossRef Campos-Pires R, Hirnet T, Valeo F, et al. Xenon improves long-term cognitive function, reduces neuronal loss and chronic neuroinflammation, and improves survival after traumatic brain injury in mice. Br J Anaesth. 2019;123:60–73.PubMedPubMedCentralCrossRef
8.
go back to reference Tucker B, Aston J, Dines M, et al. Early brain edema is a predictor of in-hospital mortality in traumatic brain injury. J Emerg Med. 2017;53:18–29.PubMedCrossRef Tucker B, Aston J, Dines M, et al. Early brain edema is a predictor of in-hospital mortality in traumatic brain injury. J Emerg Med. 2017;53:18–29.PubMedCrossRef
9.
go back to reference Kenney K, Amyot F, Haber M, et al. Cerebral vascular injury in traumatic brain injury. Exp Neurol. 2016;275(Pt 3):353–66.PubMedCrossRef Kenney K, Amyot F, Haber M, et al. Cerebral vascular injury in traumatic brain injury. Exp Neurol. 2016;275(Pt 3):353–66.PubMedCrossRef
10.
go back to reference Carney N, Totten AM, O’Reilly C, et al. Guidelines for the management of severe traumatic brain injury, fourth edition. Neurosurgery. 2017;80:6–15.PubMedCrossRef Carney N, Totten AM, O’Reilly C, et al. Guidelines for the management of severe traumatic brain injury, fourth edition. Neurosurgery. 2017;80:6–15.PubMedCrossRef
11.
go back to reference Ding H, Wang X, Wang H, et al. Nrf2-ARE signaling provides neuroprotection in traumatic brain injury via modulation of the ubiquitin proteasome system. Neurochem Int. 2017;111:32–44.PubMedCrossRef Ding H, Wang X, Wang H, et al. Nrf2-ARE signaling provides neuroprotection in traumatic brain injury via modulation of the ubiquitin proteasome system. Neurochem Int. 2017;111:32–44.PubMedCrossRef
12.
go back to reference Chandran R, Kim T, Mehta SL, et al. A combination antioxidant therapy to inhibit NOX2 and activate Nrf2 decreases secondary brain damage and improves functional recovery after traumatic brain injury. J Cereb Blood Flow Metab. 2018;38:1818–27.PubMedCrossRef Chandran R, Kim T, Mehta SL, et al. A combination antioxidant therapy to inhibit NOX2 and activate Nrf2 decreases secondary brain damage and improves functional recovery after traumatic brain injury. J Cereb Blood Flow Metab. 2018;38:1818–27.PubMedCrossRef
13.
go back to reference Cheng T, Wang W, Li Q, et al. Cerebroprotection of flavanol (-)-epicatechin after traumatic brain injury via Nrf2-dependent and -independent pathways. Free Radic Biol Med. 2016;92:15–28.PubMedCrossRef Cheng T, Wang W, Li Q, et al. Cerebroprotection of flavanol (-)-epicatechin after traumatic brain injury via Nrf2-dependent and -independent pathways. Free Radic Biol Med. 2016;92:15–28.PubMedCrossRef
14.
go back to reference Dong W, Yang B, Wang L, et al. Curcumin plays neuroprotective roles against traumatic brain injury partly via Nrf2 signaling. Toxicol Appl Pharmacol. 2018;346:28–36.PubMedCrossRef Dong W, Yang B, Wang L, et al. Curcumin plays neuroprotective roles against traumatic brain injury partly via Nrf2 signaling. Toxicol Appl Pharmacol. 2018;346:28–36.PubMedCrossRef
15.
go back to reference Chen X, Chen C, Fan S, et al. Omega-3 polyunsaturated fatty acid attenuates the inflammatory response by modulating microglia polarization through SIRT1-mediated deacetylation of the HMGB1/NF-κB pathway following experimental traumatic brain injury. J Neuroinflamm. 2018;15:116.CrossRef Chen X, Chen C, Fan S, et al. Omega-3 polyunsaturated fatty acid attenuates the inflammatory response by modulating microglia polarization through SIRT1-mediated deacetylation of the HMGB1/NF-κB pathway following experimental traumatic brain injury. J Neuroinflamm. 2018;15:116.CrossRef
17.
go back to reference Tang R, Lin YM, Liu HX, et al. Neuroprotective effect of docosahexaenoic acid in rat traumatic brain injury model via regulation of TLR4/NF-Kappa B signaling pathway. Int J Biochem Cell Biol. 2018;99:64–71.PubMedCrossRef Tang R, Lin YM, Liu HX, et al. Neuroprotective effect of docosahexaenoic acid in rat traumatic brain injury model via regulation of TLR4/NF-Kappa B signaling pathway. Int J Biochem Cell Biol. 2018;99:64–71.PubMedCrossRef
18.
go back to reference Yuan F, Xu ZM, Lu LY, et al. SIRT2 inhibition exacerbates neuroinflammation and blood-brain barrier disruption in experimental traumatic brain injury by enhancing NF-κB p65 acetylation and activation. J Neurochem. 2016;136:581–93.CrossRefPubMed Yuan F, Xu ZM, Lu LY, et al. SIRT2 inhibition exacerbates neuroinflammation and blood-brain barrier disruption in experimental traumatic brain injury by enhancing NF-κB p65 acetylation and activation. J Neurochem. 2016;136:581–93.CrossRefPubMed
19.
go back to reference Xu K, Wu F, Xu K, et al. NaHS restores mitochondrial function and inhibits autophagy by activating the PI3K/Akt/mTOR signalling pathway to improve functional recovery after traumatic brain injury. Chem Biol Interact. 2018;286:96–105.PubMedCrossRef Xu K, Wu F, Xu K, et al. NaHS restores mitochondrial function and inhibits autophagy by activating the PI3K/Akt/mTOR signalling pathway to improve functional recovery after traumatic brain injury. Chem Biol Interact. 2018;286:96–105.PubMedCrossRef
20.
go back to reference Fang J, Wang H, Zhou J, et al. Baicalin provides neuroprotection in traumatic brain injury mice model through Akt/Nrf2 pathway. Drug Des Dev Ther. 2018;12:2497–508.CrossRef Fang J, Wang H, Zhou J, et al. Baicalin provides neuroprotection in traumatic brain injury mice model through Akt/Nrf2 pathway. Drug Des Dev Ther. 2018;12:2497–508.CrossRef
21.
go back to reference Shen M, Wang S, Wen X, et al. Dexmedetomidine exerts neuroprotective effect via the activation of the PI3K/Akt/mTOR signaling pathway in rats with traumatic brain injury. Biomed Pharmacother. 2017;95:885–93.PubMedCrossRef Shen M, Wang S, Wen X, et al. Dexmedetomidine exerts neuroprotective effect via the activation of the PI3K/Akt/mTOR signaling pathway in rats with traumatic brain injury. Biomed Pharmacother. 2017;95:885–93.PubMedCrossRef
22.
go back to reference Raible DJ, Frey LC, Del Angel YC, et al. JAK/STAT pathway regulation of GABAA receptor expression after differing severities of experimental TBI. Exp Neurol. 2015;271:445–56.PubMedPubMedCentralCrossRef Raible DJ, Frey LC, Del Angel YC, et al. JAK/STAT pathway regulation of GABAA receptor expression after differing severities of experimental TBI. Exp Neurol. 2015;271:445–56.PubMedPubMedCentralCrossRef
23.
go back to reference Morganti JM, Goulding DS, Van Eldik LJ. Deletion of p38α MAPK in microglia blunts trauma-induced inflammatory responses in mice. J Neuroinflamm. 2019;16:98.CrossRef Morganti JM, Goulding DS, Van Eldik LJ. Deletion of p38α MAPK in microglia blunts trauma-induced inflammatory responses in mice. J Neuroinflamm. 2019;16:98.CrossRef
24.
go back to reference Bhowmick S, D’Mello V, Abdul-Muneer PM. Synergistic inhibition of ERK1/2 and JNK, not p38, phosphorylation ameliorates neuronal damages after traumatic brain injury. Mol Neurobiol. 2019;56:1124–36.PubMedCrossRef Bhowmick S, D’Mello V, Abdul-Muneer PM. Synergistic inhibition of ERK1/2 and JNK, not p38, phosphorylation ameliorates neuronal damages after traumatic brain injury. Mol Neurobiol. 2019;56:1124–36.PubMedCrossRef
25.
go back to reference Shim SS, Stutzmann GE. Inhibition of glycogen synthase kinase-3: an emerging target in the treatment of traumatic brain injury. J Neurotrauma. 2016;33:2065–76.PubMedCrossRef Shim SS, Stutzmann GE. Inhibition of glycogen synthase kinase-3: an emerging target in the treatment of traumatic brain injury. J Neurotrauma. 2016;33:2065–76.PubMedCrossRef
26.
go back to reference Lin CJ, Chen TH, Yang LY, et al. Resveratrol protects astrocytes against traumatic brain injury through inhibiting apoptotic and autophagic cell death. Cell Death Dis. 2014;5:e1147.PubMedPubMedCentralCrossRef Lin CJ, Chen TH, Yang LY, et al. Resveratrol protects astrocytes against traumatic brain injury through inhibiting apoptotic and autophagic cell death. Cell Death Dis. 2014;5:e1147.PubMedPubMedCentralCrossRef
27.
go back to reference Wang G, Shi Y, Jiang X, et al. HDAC inhibition prevents white matter injury by modulating microglia/macrophage polarization through the GSK3β/PTEN/Akt axis. Proc Natl Acad Sci USA. 2015;112:2853–8.PubMedCrossRefPubMedCentral Wang G, Shi Y, Jiang X, et al. HDAC inhibition prevents white matter injury by modulating microglia/macrophage polarization through the GSK3β/PTEN/Akt axis. Proc Natl Acad Sci USA. 2015;112:2853–8.PubMedCrossRefPubMedCentral
28.
go back to reference Hill JL, Kobori N, Zhao J, et al. Traumatic brain injury decreases AMP-activated protein kinase activity and pharmacological enhancement of its activity improves cognitive outcome. J Neurochem. 2016;139:106–19.PubMedPubMedCentralCrossRef Hill JL, Kobori N, Zhao J, et al. Traumatic brain injury decreases AMP-activated protein kinase activity and pharmacological enhancement of its activity improves cognitive outcome. J Neurochem. 2016;139:106–19.PubMedPubMedCentralCrossRef
29.
go back to reference Cifani P, Kentsis A. Towards comprehensive and quantitative proteomics for diagnosis and therapy of human disease. Proteomics. 2017;17:1600079.CrossRef Cifani P, Kentsis A. Towards comprehensive and quantitative proteomics for diagnosis and therapy of human disease. Proteomics. 2017;17:1600079.CrossRef
30.
go back to reference Smith JG, Gerszten RE. Emerging affinity-based proteomic technologies for large-scale plasma profiling in cardiovascular disease. Circulation. 2017;135:1651–64.PubMedPubMedCentralCrossRef Smith JG, Gerszten RE. Emerging affinity-based proteomic technologies for large-scale plasma profiling in cardiovascular disease. Circulation. 2017;135:1651–64.PubMedPubMedCentralCrossRef
31.
33.
go back to reference Liu X, Dai R, Ke M, et al. Differential proteomic analysis of dimethylnitrosamine (DMN)-induced liver fibrosis. Proteomics. 2017;17:1700267.CrossRef Liu X, Dai R, Ke M, et al. Differential proteomic analysis of dimethylnitrosamine (DMN)-induced liver fibrosis. Proteomics. 2017;17:1700267.CrossRef
34.
go back to reference Rayner SL, Morsch M, Molloy MP, et al. Using proteomics to identify ubiquitin ligase-substrate pairs: how novel methods may unveil therapeutic targets for neurodegenerative diseases. Cell Mol Life Sci. 2019;76:2499–510.PubMedCrossRef Rayner SL, Morsch M, Molloy MP, et al. Using proteomics to identify ubiquitin ligase-substrate pairs: how novel methods may unveil therapeutic targets for neurodegenerative diseases. Cell Mol Life Sci. 2019;76:2499–510.PubMedCrossRef
35.
go back to reference Drummond E, Nayak S, Faustin A, et al. Proteomic differences in amyloid plaques in rapidly progressive and sporadic Alzheimer’s disease. Acta Neuropathol. 2017;133:933–54.PubMedPubMedCentralCrossRef Drummond E, Nayak S, Faustin A, et al. Proteomic differences in amyloid plaques in rapidly progressive and sporadic Alzheimer’s disease. Acta Neuropathol. 2017;133:933–54.PubMedPubMedCentralCrossRef
36.
go back to reference Hosp F, Gutiérrez-Ángel S, Schaefer MH, et al. Spatiotemporal proteomic profiling of Huntington’s disease inclusions reveals widespread loss of protein function. Cell Rep. 2017;21:2291–303.PubMedPubMedCentralCrossRef Hosp F, Gutiérrez-Ángel S, Schaefer MH, et al. Spatiotemporal proteomic profiling of Huntington’s disease inclusions reveals widespread loss of protein function. Cell Rep. 2017;21:2291–303.PubMedPubMedCentralCrossRef
37.
go back to reference Zhang P, Zhang L, Li Y, et al. Quantitative proteomic analysis to identify differentially expressed proteins in myocardium of epilepsy using iTRAQ coupled with Nano-LC-MS/MS. J Proteome Res. 2018;17:305–14.PubMedCrossRef Zhang P, Zhang L, Li Y, et al. Quantitative proteomic analysis to identify differentially expressed proteins in myocardium of epilepsy using iTRAQ coupled with Nano-LC-MS/MS. J Proteome Res. 2018;17:305–14.PubMedCrossRef
38.
go back to reference Azzam S, Schlatzer D, Nethery D, et al. Proteomic profiling of the hypothalamus in two mouse models of narcolepsy. Proteomics. 2017;17:1600478.CrossRef Azzam S, Schlatzer D, Nethery D, et al. Proteomic profiling of the hypothalamus in two mouse models of narcolepsy. Proteomics. 2017;17:1600478.CrossRef
39.
go back to reference Hori M, Nakamachi T, Rakwal R, et al. Transcriptomics and proteomics analyses of the PACAP38 influenced ischemic brain in permanent middle cerebral artery occlusion model mice. J Neuroinflamm. 2012;9:256.CrossRef Hori M, Nakamachi T, Rakwal R, et al. Transcriptomics and proteomics analyses of the PACAP38 influenced ischemic brain in permanent middle cerebral artery occlusion model mice. J Neuroinflamm. 2012;9:256.CrossRef
40.
go back to reference Chen JH, Kuo HC, Lee KF, et al. Global proteomic analysis of brain tissues in transient ischemia brain damage in rats. Int J Mol Sci. 2015;16:11873–91.PubMedPubMedCentralCrossRef Chen JH, Kuo HC, Lee KF, et al. Global proteomic analysis of brain tissues in transient ischemia brain damage in rats. Int J Mol Sci. 2015;16:11873–91.PubMedPubMedCentralCrossRef
41.
go back to reference Huie JR, Diaz-Arrastia R, Yue JK, et al. Testing a multivariate proteomic panel for traumatic brain injury biomarker discovery: a TRACK-TBI pilot study. J Neurotrauma. 2019;36:100–10.PubMedCrossRef Huie JR, Diaz-Arrastia R, Yue JK, et al. Testing a multivariate proteomic panel for traumatic brain injury biomarker discovery: a TRACK-TBI pilot study. J Neurotrauma. 2019;36:100–10.PubMedCrossRef
42.
go back to reference Guingab-Cagmat JD, Newsom K, Vakulenko A, et al. In vitro MS-based proteomic analysis and absolute quantification of neuronal-glial injury biomarkers in cell culture system. Electrophoresis. 2012;33:3786–97.PubMedCrossRef Guingab-Cagmat JD, Newsom K, Vakulenko A, et al. In vitro MS-based proteomic analysis and absolute quantification of neuronal-glial injury biomarkers in cell culture system. Electrophoresis. 2012;33:3786–97.PubMedCrossRef
43.
go back to reference Cheng SX, Xu ZW, Yi TL, et al. iTRAQ-based quantitative proteomics reveals the new evidence base for traumatic brain injury treated with targeted temperature management. Neurotherapeutics. 2018;15:216–32.PubMedCrossRef Cheng SX, Xu ZW, Yi TL, et al. iTRAQ-based quantitative proteomics reveals the new evidence base for traumatic brain injury treated with targeted temperature management. Neurotherapeutics. 2018;15:216–32.PubMedCrossRef
44.
go back to reference Wu P, Zhao Y, Haidacher SJ, et al. Detection of structural and metabolic changes in traumatically injured hippocampus by quantitative differential proteomics. J Neurotrauma. 2013;30:775–88.PubMedPubMedCentralCrossRef Wu P, Zhao Y, Haidacher SJ, et al. Detection of structural and metabolic changes in traumatically injured hippocampus by quantitative differential proteomics. J Neurotrauma. 2013;30:775–88.PubMedPubMedCentralCrossRef
45.
go back to reference Song H, Fang S, Gao J, et al. Quantitative proteomic study reveals up-regulation of cAMP signaling pathway-related proteins in mild traumatic brain injury. J Proteome Res. 2018;17:858–69.PubMedCrossRef Song H, Fang S, Gao J, et al. Quantitative proteomic study reveals up-regulation of cAMP signaling pathway-related proteins in mild traumatic brain injury. J Proteome Res. 2018;17:858–69.PubMedCrossRef
46.
go back to reference Zhao L, Liu Q, Ma S, et al. TPEN attenuates neural autophagy induced by synaptically-released zinc translocation and improves histological outcomes after traumatic brain injury in rats. Ann Clin Lab Sci. 2018;48:446–52.PubMed Zhao L, Liu Q, Ma S, et al. TPEN attenuates neural autophagy induced by synaptically-released zinc translocation and improves histological outcomes after traumatic brain injury in rats. Ann Clin Lab Sci. 2018;48:446–52.PubMed
47.
go back to reference Büchele F, Morawska MM, Schreglmann SR, et al. Novel rat model of weight drop-induced closed diffuse traumatic brain injury compatible with electrophysiological recordings of vigilance states. J Neurotrauma. 2016;33:1171–80.PubMedCrossRef Büchele F, Morawska MM, Schreglmann SR, et al. Novel rat model of weight drop-induced closed diffuse traumatic brain injury compatible with electrophysiological recordings of vigilance states. J Neurotrauma. 2016;33:1171–80.PubMedCrossRef
48.
go back to reference Wiśniewski JR, Zougman A, Nagaraj N, et al. Universal sample preparation method for proteome analysis. Nat Methods. 2009;6:359–62.PubMedCrossRef Wiśniewski JR, Zougman A, Nagaraj N, et al. Universal sample preparation method for proteome analysis. Nat Methods. 2009;6:359–62.PubMedCrossRef
49.
go back to reference Li J, Donangelo I, Abe K, et al. Thyroid hormone treatment activates protective pathways in both in vivo and in vitro models of neuronal injury. Mol Cell Endocrinol. 2017;452:120–30.PubMedCrossRef Li J, Donangelo I, Abe K, et al. Thyroid hormone treatment activates protective pathways in both in vivo and in vitro models of neuronal injury. Mol Cell Endocrinol. 2017;452:120–30.PubMedCrossRef
50.
go back to reference Sullivan PG, Sebastian AH, Hall ED. Therapeutic window analysis of the neuroprotective effects of cyclosporine A after traumatic brain injury. J Neurotrauma. 2011;28:311–8.PubMedPubMedCentralCrossRef Sullivan PG, Sebastian AH, Hall ED. Therapeutic window analysis of the neuroprotective effects of cyclosporine A after traumatic brain injury. J Neurotrauma. 2011;28:311–8.PubMedPubMedCentralCrossRef
51.
go back to reference Crupi R, Paterniti I, Campolo M, et al. Exogenous T3 administration provides neuroprotection in a murine model of traumatic brain injury. Pharmacol Res. 2013;70:80–9.PubMedCrossRef Crupi R, Paterniti I, Campolo M, et al. Exogenous T3 administration provides neuroprotection in a murine model of traumatic brain injury. Pharmacol Res. 2013;70:80–9.PubMedCrossRef
52.
go back to reference Bareyre F, Wahl F, McIntosh TK, et al. Time course of cerebral edema after traumatic brain injury in rats: effects of riluzole and mannitol. J Neurotrauma. 1997;14:839–49.PubMedCrossRef Bareyre F, Wahl F, McIntosh TK, et al. Time course of cerebral edema after traumatic brain injury in rats: effects of riluzole and mannitol. J Neurotrauma. 1997;14:839–49.PubMedCrossRef
53.
go back to reference Vigil FA, Giese KP. Calcium/calmodulin-dependent kinase II and memory destabilization: a new role in memory maintenance. J Neurochem. 2018;147:12–23.PubMedPubMedCentralCrossRef Vigil FA, Giese KP. Calcium/calmodulin-dependent kinase II and memory destabilization: a new role in memory maintenance. J Neurochem. 2018;147:12–23.PubMedPubMedCentralCrossRef
54.
go back to reference Zhao J, Wang T, Lv Q, et al. Expression of heat shock protein 70 and Annexin A1 in serum of patients with acutely severe traumatic brain injury. Exp Ther Med. 2020;19:1896–902.PubMed Zhao J, Wang T, Lv Q, et al. Expression of heat shock protein 70 and Annexin A1 in serum of patients with acutely severe traumatic brain injury. Exp Ther Med. 2020;19:1896–902.PubMed
55.
go back to reference Wang T, Yu DR, Huang J, et al. Multimodal rehabilitation program promotes motor function recovery of rats after ischemic stroke by upregulating expressions of GAP-43, SYN, HSP70, and C-MYC. J Stroke Cerebrovasc Dis. 2018;27:2829–39.PubMedCrossRef Wang T, Yu DR, Huang J, et al. Multimodal rehabilitation program promotes motor function recovery of rats after ischemic stroke by upregulating expressions of GAP-43, SYN, HSP70, and C-MYC. J Stroke Cerebrovasc Dis. 2018;27:2829–39.PubMedCrossRef
56.
go back to reference Gorter RP, Nutma E, Jahrei MC, et al. Heat shock proteins are differentially expressed in brain and spinal cord: implications for multiple sclerosis. Clin Exp Immunol. 2018;194:137–52.PubMedPubMedCentralCrossRef Gorter RP, Nutma E, Jahrei MC, et al. Heat shock proteins are differentially expressed in brain and spinal cord: implications for multiple sclerosis. Clin Exp Immunol. 2018;194:137–52.PubMedPubMedCentralCrossRef
57.
go back to reference Zhu Z, Reiser G. The small heat shock proteins, especially HspB4 and HspB5 are promising protectants in neurodegenerative diseases. Neurochem Int. 2018;115:69–79.PubMedCrossRef Zhu Z, Reiser G. The small heat shock proteins, especially HspB4 and HspB5 are promising protectants in neurodegenerative diseases. Neurochem Int. 2018;115:69–79.PubMedCrossRef
58.
go back to reference Kim JY, Kim JW, Yenari MA. Heat shock protein signaling in brain ischemia and injury. Neurosci Lett. 2020;715:134642.PubMedCrossRef Kim JY, Kim JW, Yenari MA. Heat shock protein signaling in brain ischemia and injury. Neurosci Lett. 2020;715:134642.PubMedCrossRef
59.
go back to reference Qiu X, Ping S, Kyle M, et al. S100 calcium-binding protein A9 knockout contributes to neuroprotection and functional improvement after traumatic brain injury. J Neurotrauma. 2020;37:950–65.PubMedCrossRef Qiu X, Ping S, Kyle M, et al. S100 calcium-binding protein A9 knockout contributes to neuroprotection and functional improvement after traumatic brain injury. J Neurotrauma. 2020;37:950–65.PubMedCrossRef
60.
go back to reference Yang Y, Dong B, Lu J, et al. Hemopexin reduces blood–brain barrier injury and protects synaptic plasticity in cerebral ischemic rats by promoting EPCs through the HO-1 pathway. Brain Res. 2018;1699:177–85.PubMedCrossRef Yang Y, Dong B, Lu J, et al. Hemopexin reduces blood–brain barrier injury and protects synaptic plasticity in cerebral ischemic rats by promoting EPCs through the HO-1 pathway. Brain Res. 2018;1699:177–85.PubMedCrossRef
61.
go back to reference Toman E, Bishop JR, Davies DJ, et al. Vitamin D deficiency in traumatic brain injury and its relationship with severity of injury and quality of life: a prospective, observational study. J Neurotrauma. 2017;34:1448–56.PubMedCrossRef Toman E, Bishop JR, Davies DJ, et al. Vitamin D deficiency in traumatic brain injury and its relationship with severity of injury and quality of life: a prospective, observational study. J Neurotrauma. 2017;34:1448–56.PubMedCrossRef
62.
go back to reference Gao JB, Tang WD, Wang X, et al. Prognostic value of neuropeptide proenkephalin A in patients with severe traumatic brain injury. Peptides. 2014;58:42–6.PubMedCrossRef Gao JB, Tang WD, Wang X, et al. Prognostic value of neuropeptide proenkephalin A in patients with severe traumatic brain injury. Peptides. 2014;58:42–6.PubMedCrossRef
63.
go back to reference Meidahl AC, Klukinov M, Tzabazis AZ, et al. Nasal application of HSV encoding human preproenkephalin blocks craniofacial pain in a rat model of traumatic brain injury. Gene Ther. 2017;24:482–6.PubMedCrossRef Meidahl AC, Klukinov M, Tzabazis AZ, et al. Nasal application of HSV encoding human preproenkephalin blocks craniofacial pain in a rat model of traumatic brain injury. Gene Ther. 2017;24:482–6.PubMedCrossRef
64.
65.
go back to reference Merkel SF, Razmpour R, Lutton EM, et al. Adolescent traumatic brain injury induces chronic mesolimbic neuroinflammation with concurrent enhancement in the rewarding effects of cocaine in mice during adulthood. J Neurotrauma. 2017;34:165–81.PubMedPubMedCentralCrossRef Merkel SF, Razmpour R, Lutton EM, et al. Adolescent traumatic brain injury induces chronic mesolimbic neuroinflammation with concurrent enhancement in the rewarding effects of cocaine in mice during adulthood. J Neurotrauma. 2017;34:165–81.PubMedPubMedCentralCrossRef
66.
67.
go back to reference Volkow ND, Wise RA, Baler R. The dopamine motive system: implications for drug and food addiction. Nat Rev Neurosci. 2017;18:741–52.PubMedCrossRef Volkow ND, Wise RA, Baler R. The dopamine motive system: implications for drug and food addiction. Nat Rev Neurosci. 2017;18:741–52.PubMedCrossRef
68.
go back to reference Lu K, Liang CL, Li PC, et al. Risk factors for myocardial dysfunction after traumatic brain injury: a one-year follow-up study. Injury. 2017;48:1794–800.PubMedCrossRef Lu K, Liang CL, Li PC, et al. Risk factors for myocardial dysfunction after traumatic brain injury: a one-year follow-up study. Injury. 2017;48:1794–800.PubMedCrossRef
69.
go back to reference El-Menyar A, Goyal A, Latifi R, et al. Brain–heart interactions in traumatic brain injury. Cardiol Rev. 2017;25:279–88.PubMedCrossRef El-Menyar A, Goyal A, Latifi R, et al. Brain–heart interactions in traumatic brain injury. Cardiol Rev. 2017;25:279–88.PubMedCrossRef
70.
go back to reference Liu YY, Brent GA. Thyroid hormone and the brain: mechisms of action in development and role in protection and promotion of recovery after brain injury. Pharmacol Ther. 2018;186:176–85.PubMedPubMedCentralCrossRef Liu YY, Brent GA. Thyroid hormone and the brain: mechisms of action in development and role in protection and promotion of recovery after brain injury. Pharmacol Ther. 2018;186:176–85.PubMedPubMedCentralCrossRef
71.
go back to reference Richardson SJ, Wijayagunaratne RC, D’Souza DG, et al. Transport of thyroid hormones via the choroid plexus into the brain: the roles of transthyretin and thyroid hormone transmembrane transporters. Front Neurosci. 2015;9:66.PubMedPubMedCentralCrossRef Richardson SJ, Wijayagunaratne RC, D’Souza DG, et al. Transport of thyroid hormones via the choroid plexus into the brain: the roles of transthyretin and thyroid hormone transmembrane transporters. Front Neurosci. 2015;9:66.PubMedPubMedCentralCrossRef
72.
go back to reference Arneson D, Zhang G, Ying Z, et al. Single cell molecular alterations reveal target cells and pathways of concussive brain injury. Nat Commun. 2018;9:3894.PubMedPubMedCentralCrossRef Arneson D, Zhang G, Ying Z, et al. Single cell molecular alterations reveal target cells and pathways of concussive brain injury. Nat Commun. 2018;9:3894.PubMedPubMedCentralCrossRef
73.
go back to reference Boza-Serrano A, Ruiz R, Sanchez-Varo R, et al. Galectin-3, a novel endogenous TREM2 ligand, detrimentally regulates inflammatory response in Alzheimer’s disease. Acta Neuropathol. 2019;138:251–73.PubMedPubMedCentralCrossRef Boza-Serrano A, Ruiz R, Sanchez-Varo R, et al. Galectin-3, a novel endogenous TREM2 ligand, detrimentally regulates inflammatory response in Alzheimer’s disease. Acta Neuropathol. 2019;138:251–73.PubMedPubMedCentralCrossRef
74.
go back to reference Siew JJ, Chen HM, Chen HY, et al. Galectin-3 is required for the microglia-mediated brain inflammation in a model of Huntington’s disease. Nat Commun. 2019;10:3473.PubMedPubMedCentralCrossRef Siew JJ, Chen HM, Chen HY, et al. Galectin-3 is required for the microglia-mediated brain inflammation in a model of Huntington’s disease. Nat Commun. 2019;10:3473.PubMedPubMedCentralCrossRef
75.
go back to reference Nishikawa H, Nakano F, Liu L, et al. The role of galectin-3 in subarachnoid hemorrhage: a preliminary study. Acta Neurochir Suppl. 2020;127:65–8.PubMedCrossRef Nishikawa H, Nakano F, Liu L, et al. The role of galectin-3 in subarachnoid hemorrhage: a preliminary study. Acta Neurochir Suppl. 2020;127:65–8.PubMedCrossRef
76.
go back to reference Zhai W, Chen D, Shen H, et al. A1 adenosine receptor attenuates intracerebral hemorrhage-induced secondary brain injury in rats by activating the P38-MAPKAP2-Hsp27 pathway. Mol Brain. 2016;9:66.PubMedPubMedCentralCrossRef Zhai W, Chen D, Shen H, et al. A1 adenosine receptor attenuates intracerebral hemorrhage-induced secondary brain injury in rats by activating the P38-MAPKAP2-Hsp27 pathway. Mol Brain. 2016;9:66.PubMedPubMedCentralCrossRef
77.
go back to reference Shi Y, Jiang X, Zhang L, et al. Endothelium-targeted overexpression of heat shock protein 27 ameliorates blood-brain barrier disruption after ischemic brain injury. Proc Natl Acad Sci USA. 2017;114:E1243-e1252.PubMedCrossRefPubMedCentral Shi Y, Jiang X, Zhang L, et al. Endothelium-targeted overexpression of heat shock protein 27 ameliorates blood-brain barrier disruption after ischemic brain injury. Proc Natl Acad Sci USA. 2017;114:E1243-e1252.PubMedCrossRefPubMedCentral
78.
go back to reference Wilhelmsson U, Pozo-Rodrigalvarez A, Kalm M, et al. The role of GFAP and vimentin in learning and memory. Biol Chem. 2019;400:1147–56.PubMedCrossRef Wilhelmsson U, Pozo-Rodrigalvarez A, Kalm M, et al. The role of GFAP and vimentin in learning and memory. Biol Chem. 2019;400:1147–56.PubMedCrossRef
79.
go back to reference Yue JK, Yuh EL, Korley FK, et al. Association between plasma GFAP concentrations and MRI abnormalities in patients with CT-negative traumatic brain injury in the TRACK-TBI cohort: a prospective multicentre study. Lancet Neurol. 2019;18:953–61.PubMedCrossRef Yue JK, Yuh EL, Korley FK, et al. Association between plasma GFAP concentrations and MRI abnormalities in patients with CT-negative traumatic brain injury in the TRACK-TBI cohort: a prospective multicentre study. Lancet Neurol. 2019;18:953–61.PubMedCrossRef
80.
go back to reference Driessen TM, Zhao C, Saenz M, et al. Down-regulation of fatty acid binding protein 7 (Fabp7) is a hallmark of the postpartum brain. J Chem Neuroanat. 2018;92:92–101.PubMedPubMedCentralCrossRef Driessen TM, Zhao C, Saenz M, et al. Down-regulation of fatty acid binding protein 7 (Fabp7) is a hallmark of the postpartum brain. J Chem Neuroanat. 2018;92:92–101.PubMedPubMedCentralCrossRef
81.
go back to reference Rui Q, Ni H, Lin X, et al. Astrocyte-derived fatty acid-binding protein 7 protects blood-brain barrier integrity through a caveolin-1/MMP signaling pathway following traumatic brain injury. Exp Neurol. 2019;322:113044.PubMedCrossRef Rui Q, Ni H, Lin X, et al. Astrocyte-derived fatty acid-binding protein 7 protects blood-brain barrier integrity through a caveolin-1/MMP signaling pathway following traumatic brain injury. Exp Neurol. 2019;322:113044.PubMedCrossRef
82.
go back to reference Chirackal Manavalan AP, Kober A, Metso J, et al. Phospholipid transfer protein is expressed in cerebrovascular endothelial cells and involved in high density lipoprotein biogenesis and remodeling at the blood-brain barrier. J Biol Chem. 2014;289:4683–98.PubMedCrossRef Chirackal Manavalan AP, Kober A, Metso J, et al. Phospholipid transfer protein is expressed in cerebrovascular endothelial cells and involved in high density lipoprotein biogenesis and remodeling at the blood-brain barrier. J Biol Chem. 2014;289:4683–98.PubMedCrossRef
83.
go back to reference Hoshi Y, Uchida Y, Kuroda T, et al. Distinct roles of ezrin, radixin and moesin in maintaining the plasma membrane localizations and functions of human blood-brain barrier transporters. J Cereb Blood Flow Metab. 2020;40:1533–45.PubMedCrossRef Hoshi Y, Uchida Y, Kuroda T, et al. Distinct roles of ezrin, radixin and moesin in maintaining the plasma membrane localizations and functions of human blood-brain barrier transporters. J Cereb Blood Flow Metab. 2020;40:1533–45.PubMedCrossRef
84.
go back to reference Cui X, Chopp M, Zhang Z, et al. ABCA1/ApoE/HDL pathway mediates GW3965-induced neurorestoration after stroke. Stroke. 2017;48:459–67.PubMedCrossRef Cui X, Chopp M, Zhang Z, et al. ABCA1/ApoE/HDL pathway mediates GW3965-induced neurorestoration after stroke. Stroke. 2017;48:459–67.PubMedCrossRef
85.
go back to reference Rak M, Bénit P, Chrétien D, et al. Mitochondrial cytochrome c oxidase deficiency. Clin Sci (Lond). 2016;130:393–407.CrossRef Rak M, Bénit P, Chrétien D, et al. Mitochondrial cytochrome c oxidase deficiency. Clin Sci (Lond). 2016;130:393–407.CrossRef
Metadata
Title
iTRAQ-based proteomic profiling reveals protein alterations after traumatic brain injury and supports thyroxine as a potential treatment
Authors
Zhongxiang Zhang
Jiangtao Yu
Pengcheng Wang
Lian Lin
Ruining Liu
Rong Zeng
Haoli Ma
Yan Zhao
Publication date
01-12-2021
Publisher
BioMed Central
Published in
Molecular Brain / Issue 1/2021
Electronic ISSN: 1756-6606
DOI
https://doi.org/10.1186/s13041-021-00739-0

Other articles of this Issue 1/2021

Molecular Brain 1/2021 Go to the issue