Skip to main content
Erschienen in: Inflammation 5/2021

19.04.2021 | Original Article

Ramelteon Ameliorates LPS-Induced Hyperpermeability of the Blood-Brain Barrier (BBB) by Activating Nrf2

verfasst von: Yonglei Liu, Lixia Wang, Ning Du, Xiaoling Yin, Hongtao Shao, Lin Yang

Erschienen in: Inflammation | Ausgabe 5/2021

Einloggen, um Zugang zu erhalten

Abstract

The blood-brain barrier (BBB) is important for protecting the brain tissue by selectively exchanging substances between the blood and brain. The integrity of the BBB can be damaged by multiple factors, including oxidative stress and inflammation. Ramelteon is an oral hypnotic drug, and in the present study, we investigated its protective effect on BBB damage, as well as the underlying mechanism. LPS was used to induce BBB damage on mice and stimulate injury on endothelial cells. Evans blue staining assay was used to measure the brain permeability. The expressions of ZO-1 and Occludin were evaluated using immunostaining and Western blot in the brain tissue and endothelial cells, respectively. qRT-PCR and ELISA were used to detect the production of IL-1β and MCP-1 in the brain vessels. TBA assay was utilized to examine the concentration of MDA in the brain tissue and endothelial cells. The expression of Nrf2 in the nucleus and NQO1 were determined using Western blot assay. The endothelial permeability of the monolayer was examined using the FITC-dextran permeation assay. Firstly, the increased brain permeability and downregulated expression of tight junction proteins in the brain tissue induced by LPS were significantly reversed by treatment with Ramelteon, accompanied by the decrease in the production of IL-1β and MCP-1 in the vessels in mice. Also, the Nrf2 signaling was activated and oxidative stress in the brain vessels was alleviated by treatment with Ramelteon. Secondly, LPS-induced increase in endothelial monolayer permeability and decrease in tight junction protein expression in bEnd.3 brain endothelial cells were significantly reversed by Ramelteon, accompanied by activated Nrf2 signaling and alleviated oxidative stress. Lastly, the protective effects of Ramelteon against LPS-induced reduction of ZO-1 and Occludin, and the increase in endothelial monolayer permeability were dramatically abolished by silencing Nrf2. Ramelteon might ameliorate LPS-induced hyperpermeability of the BBB by activating the Nrf2 signaling pathway.
Literatur
1.
Zurück zum Zitat Rosenberg, Gary A. 2012. Neurological diseases in relation to the blood-brain barrier, Neurological Diseases in Relation to the Blood–Brain Barrier. Journal of Cerebral Blood Flow and Metabolism 32 (7): 1139–1151. Rosenberg, Gary A. 2012. Neurological diseases in relation to the blood-brain barrier, Neurological Diseases in Relation to the Blood–Brain Barrier. Journal of Cerebral Blood Flow and Metabolism 32 (7): 1139–1151.
2.
Zurück zum Zitat Weiss, Nicholas, F. Miller, S. Cazaubon, and P.O. Couraud. 2009. The blood-brain barrier in brain homeostasis and neurological diseases. Biochimica et Biophysica Acta 1788 (4): 842–857.CrossRef Weiss, Nicholas, F. Miller, S. Cazaubon, and P.O. Couraud. 2009. The blood-brain barrier in brain homeostasis and neurological diseases. Biochimica et Biophysica Acta 1788 (4): 842–857.CrossRef
3.
Zurück zum Zitat Martin, Greg S., D.M. Mannino, and M. Moss. 2006. The effect of age on the development and outcome of adult sepsis. Critical Care Medicine 34 (1): 15–21.CrossRef Martin, Greg S., D.M. Mannino, and M. Moss. 2006. The effect of age on the development and outcome of adult sepsis. Critical Care Medicine 34 (1): 15–21.CrossRef
4.
Zurück zum Zitat Shukla, Preshant, G.M. Rao, G. Pandey, S. Sharma, N. Mittapelly, R. Shegokar, and P.R. Mishra. 2014. Therapeutic interventions in sepsis: current and anticipated pharmacological agents. British Journal of Pharmacology 171 (22): 5011–5031.PubMedPubMedCentral Shukla, Preshant, G.M. Rao, G. Pandey, S. Sharma, N. Mittapelly, R. Shegokar, and P.R. Mishra. 2014. Therapeutic interventions in sepsis: current and anticipated pharmacological agents. British Journal of Pharmacology 171 (22): 5011–5031.PubMedPubMedCentral
5.
Zurück zum Zitat Seok, Sun M., J.M. Kim, T.Y. Park, E.J. Baik, and S.H. Lee. 2013. Fructose-1,6-bisphosphate ameliorates lipopolysaccharide-induced dysfunction of blood-brain barrier. Archives of Pharmacal Research 36 (9): 1149–1159.CrossRef Seok, Sun M., J.M. Kim, T.Y. Park, E.J. Baik, and S.H. Lee. 2013. Fructose-1,6-bisphosphate ameliorates lipopolysaccharide-induced dysfunction of blood-brain barrier. Archives of Pharmacal Research 36 (9): 1149–1159.CrossRef
6.
Zurück zum Zitat Kahles, Timo, P. Luedike, M. Endres, H.J. Galla, H. Steinmetz, R. Busse, T. Neumann-Haefelin, and R.P. Brandes. 2007. NADPH oxidase plays a central role in blood-brain barrier damage in experimental stroke. Stroke 38 (11): 3000–3006.CrossRef Kahles, Timo, P. Luedike, M. Endres, H.J. Galla, H. Steinmetz, R. Busse, T. Neumann-Haefelin, and R.P. Brandes. 2007. NADPH oxidase plays a central role in blood-brain barrier damage in experimental stroke. Stroke 38 (11): 3000–3006.CrossRef
7.
Zurück zum Zitat Liu, Chao, J. Wu, and M.H. Zou. 2012. Activation of AMP-activated protein kinase alleviates high-glucose-induced dysfunction of brain microvascular endothelial cell tight-junction dynamics. Free Radical Biology & Medicine 53 (6): 1213–1221.CrossRef Liu, Chao, J. Wu, and M.H. Zou. 2012. Activation of AMP-activated protein kinase alleviates high-glucose-induced dysfunction of brain microvascular endothelial cell tight-junction dynamics. Free Radical Biology & Medicine 53 (6): 1213–1221.CrossRef
8.
Zurück zum Zitat Zhao, Zhihong, J. Hu, X. Gao, H. Liang, and Z. Liu. 2014. Activation of AMPK attenuates lipopolysaccharide-impaired integrity and function of blood-brain barrier in human brain microvascular endothelial cells. Experimental and Molecular Pathology 97 (3): 386–392.CrossRef Zhao, Zhihong, J. Hu, X. Gao, H. Liang, and Z. Liu. 2014. Activation of AMPK attenuates lipopolysaccharide-impaired integrity and function of blood-brain barrier in human brain microvascular endothelial cells. Experimental and Molecular Pathology 97 (3): 386–392.CrossRef
9.
Zurück zum Zitat Zhou, Xiaomei, Y. Cao, G. Ao, L. Hu, H. Liu, J. Wu, X. Wang, M. Jin, S. Zheng, X. Zhen, N.J. Alkayed, J. Jia, and J. Cheng. 2014. CaMKKbeta-dependent activation of AMP-activated protein kinase is critical to suppressive effects of hydrogen sulfide on neuroinflammation. Antioxidants & Redox Signaling 21 (12): 1741–1758.CrossRef Zhou, Xiaomei, Y. Cao, G. Ao, L. Hu, H. Liu, J. Wu, X. Wang, M. Jin, S. Zheng, X. Zhen, N.J. Alkayed, J. Jia, and J. Cheng. 2014. CaMKKbeta-dependent activation of AMP-activated protein kinase is critical to suppressive effects of hydrogen sulfide on neuroinflammation. Antioxidants & Redox Signaling 21 (12): 1741–1758.CrossRef
10.
Zurück zum Zitat Robberecht, W. 2000. Oxidative stress in amyotrophic lateral sclerosis. Journal of Neurology 247 (Suppl 1): I1–I6.CrossRef Robberecht, W. 2000. Oxidative stress in amyotrophic lateral sclerosis. Journal of Neurology 247 (Suppl 1): I1–I6.CrossRef
11.
Zurück zum Zitat van Horssen, Jack, Witte ME, Schreibelt G and de Vries HE. Radical changes in multiple sclerosis pathogenesis. 2011. Biochimica et Biophysica Acta. 1812 (2): 141-150. van Horssen, Jack, Witte ME, Schreibelt G and de Vries HE. Radical changes in multiple sclerosis pathogenesis. 2011. Biochimica et Biophysica Acta. 1812 (2): 141-150.
12.
Zurück zum Zitat Olmez, Inan, and H. Ozyurt. 2012. Reactive oxygen species and ischemic cerebrovascular disease, Neurochem. Int. 60 (2): 208–212. Olmez, Inan, and H. Ozyurt. 2012. Reactive oxygen species and ischemic cerebrovascular disease, Neurochem. Int. 60 (2): 208–212.
13.
Zurück zum Zitat Pun, B. Pamela, J. Lu, and S. Moochhala. 2009. Involvement of ROS in BBB dysfunction. Free Radical Research 43 (4): 348–364.CrossRef Pun, B. Pamela, J. Lu, and S. Moochhala. 2009. Involvement of ROS in BBB dysfunction. Free Radical Research 43 (4): 348–364.CrossRef
14.
Zurück zum Zitat Gu, Yong, G. Zheng, M. Xu, Y. Li, X. Chen, W. Zhu, Y. Tong, S.K. Chung, K.J. Liu, and J. Shen. 2012. Caveolin-1 regulates nitric oxide-mediated matrix metalloproteinases activity and blood-brain barrier permeability in focal cerebral ischemia and reperfusion injury. Journal of Neurochemistry 120 (1): 147–156.CrossRef Gu, Yong, G. Zheng, M. Xu, Y. Li, X. Chen, W. Zhu, Y. Tong, S.K. Chung, K.J. Liu, and J. Shen. 2012. Caveolin-1 regulates nitric oxide-mediated matrix metalloproteinases activity and blood-brain barrier permeability in focal cerebral ischemia and reperfusion injury. Journal of Neurochemistry 120 (1): 147–156.CrossRef
15.
Zurück zum Zitat Ramelteon. In: editors. LiverTox: Clinical and research information on drug-induced liver injury. 2012. Bethesda (MD). Ramelteon. In: editors. LiverTox: Clinical and research information on drug-induced liver injury. 2012. Bethesda (MD).
16.
Zurück zum Zitat Wang, Junming, C. Jiang, K. Zhang, X. Lan, X. Chen, W. Zang, Z. Wang, F. Guan, C. Zhu, X. Yang, H. Lu, and J. Wang. 2019. Melatonin receptor activation provides cerebral protection after traumatic brain injury by mitigating oxidative stress and inflammation via the Nrf2 signaling pathway. Free Radical Biology & Medicine 131: 345–355.CrossRef Wang, Junming, C. Jiang, K. Zhang, X. Lan, X. Chen, W. Zang, Z. Wang, F. Guan, C. Zhu, X. Yang, H. Lu, and J. Wang. 2019. Melatonin receptor activation provides cerebral protection after traumatic brain injury by mitigating oxidative stress and inflammation via the Nrf2 signaling pathway. Free Radical Biology & Medicine 131: 345–355.CrossRef
17.
Zurück zum Zitat Wang, Tao, Z. Li, S. Xia, Z. Xu, X. Chen, and H. Sun. 2020. The protective effects of ramelteon against isoflurane-induced insults and inflammatory response in brain microvascular endothelial cells. Neurotoxicity Research. Wang, Tao, Z. Li, S. Xia, Z. Xu, X. Chen, and H. Sun. 2020. The protective effects of ramelteon against isoflurane-induced insults and inflammatory response in brain microvascular endothelial cells. Neurotoxicity Research.
18.
Zurück zum Zitat Webster, Carl I., N. Caram-Salas, A.S. Haqqani, G. Thom, L. Brown, K. Rennie, A. Yogi, W. Costain, E. Brunette, and D.B. Stanimirovic. 2016. Brain penetration, target engagement, and disposition of the blood-brain barrier-crossing bispecific antibody antagonist of metabotropic glutamate receptor type 1, FASEB. J. 30 (5): 1927–1940. Webster, Carl I., N. Caram-Salas, A.S. Haqqani, G. Thom, L. Brown, K. Rennie, A. Yogi, W. Costain, E. Brunette, and D.B. Stanimirovic. 2016. Brain penetration, target engagement, and disposition of the blood-brain barrier-crossing bispecific antibody antagonist of metabotropic glutamate receptor type 1, FASEB. J. 30 (5): 1927–1940.
19.
Zurück zum Zitat Wang, Hwai-Lee, and T.W. Lai. 2014. Optimization of Evans blue quantitation in limited rat tissue samples. Scientific Reports 4: 6588.CrossRef Wang, Hwai-Lee, and T.W. Lai. 2014. Optimization of Evans blue quantitation in limited rat tissue samples. Scientific Reports 4: 6588.CrossRef
20.
Zurück zum Zitat Lin, Lianzhu, W. Deng, Y. Tian, W. Chen, J. Wang, L. Fu, D. Shi, M. Zhao, and W. Luo. 2014. Lasiodin inhibits proliferation of human nasopharyngeal carcinoma cells by simultaneous modulation of the Apaf-1/caspase, AKT/MAPK and COX-2/NF-kappaB signaling pathways. PLoS One 9 (5): e97799.CrossRef Lin, Lianzhu, W. Deng, Y. Tian, W. Chen, J. Wang, L. Fu, D. Shi, M. Zhao, and W. Luo. 2014. Lasiodin inhibits proliferation of human nasopharyngeal carcinoma cells by simultaneous modulation of the Apaf-1/caspase, AKT/MAPK and COX-2/NF-kappaB signaling pathways. PLoS One 9 (5): e97799.CrossRef
21.
Zurück zum Zitat Chen, Da, T. Ma, X.W. Liu, C. Yang, and Z. Liu. 2015. Rapamycin reverses paraquat-induced acute lung injury in a rat model through inhibition of NFkappaB activation. International Journal of Clinical and Experimental Pathology 8 (5): 4627–4638.PubMedPubMedCentral Chen, Da, T. Ma, X.W. Liu, C. Yang, and Z. Liu. 2015. Rapamycin reverses paraquat-induced acute lung injury in a rat model through inhibition of NFkappaB activation. International Journal of Clinical and Experimental Pathology 8 (5): 4627–4638.PubMedPubMedCentral
22.
Zurück zum Zitat Yamazaki, Yu, D.J. Baker, M. Tachibana, C.C. Liu, J.M. van Deursen, T.G. Brott, G. Bu, and T. Kanekiyo. 2016. Vascular cell senescence contributes to blood-brain barrier breakdown. Stroke 47 (4): 1068–1077.CrossRef Yamazaki, Yu, D.J. Baker, M. Tachibana, C.C. Liu, J.M. van Deursen, T.G. Brott, G. Bu, and T. Kanekiyo. 2016. Vascular cell senescence contributes to blood-brain barrier breakdown. Stroke 47 (4): 1068–1077.CrossRef
23.
Zurück zum Zitat Qian, Tongcheng, S.E. Maguire, S.G. Canfield, X. Bao, W.R. Olson, E.V. Shusta, and S.P. Palecek. 2017. Directed differentiation of human pluripotent stem cells to blood-brain barrier endothelial cells. Science Advances 3 (11): e1701679.CrossRef Qian, Tongcheng, S.E. Maguire, S.G. Canfield, X. Bao, W.R. Olson, E.V. Shusta, and S.P. Palecek. 2017. Directed differentiation of human pluripotent stem cells to blood-brain barrier endothelial cells. Science Advances 3 (11): e1701679.CrossRef
24.
Zurück zum Zitat Khan, and Ehsan. 2005. An examination of the blood-brain barrier in health and disease. The British Journal of Nursing 14 (9): 509–513.CrossRef Khan, and Ehsan. 2005. An examination of the blood-brain barrier in health and disease. The British Journal of Nursing 14 (9): 509–513.CrossRef
25.
Zurück zum Zitat Hawkins, A. Richard, R.L. O’Kane, I.A. Simpson, and J.R. Vina. 2006. Structure of the blood-brain barrier and its role in the transport of amino acids. The Journal of Nutrition 136 (1 Suppl): 218S–226S.CrossRef Hawkins, A. Richard, R.L. O’Kane, I.A. Simpson, and J.R. Vina. 2006. Structure of the blood-brain barrier and its role in the transport of amino acids. The Journal of Nutrition 136 (1 Suppl): 218S–226S.CrossRef
26.
Zurück zum Zitat Bauer, Hannelore, and A. Traweger. 2016. Tight junctions of the blood-brain barrier - a molecular gatekeeper. CNS & Neurological Disorders Drug Targets 15 (9): 1016–1029.CrossRef Bauer, Hannelore, and A. Traweger. 2016. Tight junctions of the blood-brain barrier - a molecular gatekeeper. CNS & Neurological Disorders Drug Targets 15 (9): 1016–1029.CrossRef
27.
Zurück zum Zitat Chern, Chang-Ming, J.F. Liao, Y.H. Wang, and Y.C. Shen. 2012. Melatonin ameliorates neural function by promoting endogenous neurogenesis through the MT2 melatonin receptor in ischemic-stroke mice. Free Radical Biology & Medicine 52 (9): 1634–1647.CrossRef Chern, Chang-Ming, J.F. Liao, Y.H. Wang, and Y.C. Shen. 2012. Melatonin ameliorates neural function by promoting endogenous neurogenesis through the MT2 melatonin receptor in ischemic-stroke mice. Free Radical Biology & Medicine 52 (9): 1634–1647.CrossRef
28.
Zurück zum Zitat Chen, Sheng, Q. Yang, G. Chen, and J.H. Zhang. 2015. An update on inflammation in the acute phase of intracerebral hemorrhage. Translational Stroke Research 6 (1): 4–8.CrossRef Chen, Sheng, Q. Yang, G. Chen, and J.H. Zhang. 2015. An update on inflammation in the acute phase of intracerebral hemorrhage. Translational Stroke Research 6 (1): 4–8.CrossRef
29.
Zurück zum Zitat Aronowski, Jaroslaw, and X. Zhao. 2011. Molecular pathophysiology of cerebral hemorrhage: secondary brain injury. Stroke. 42 (6): 1781–1786.CrossRef Aronowski, Jaroslaw, and X. Zhao. 2011. Molecular pathophysiology of cerebral hemorrhage: secondary brain injury. Stroke. 42 (6): 1781–1786.CrossRef
30.
Zurück zum Zitat Neumann, Silke, N.J. Shields, T. Balle, M. Chebib, and A.N. Clarkson. 2015. Innate immunity and inflammation post-stroke: an alpha7-nicotinic agonist perspective. International Journal of Molecular Sciences 16 (12): 29029–29046.CrossRef Neumann, Silke, N.J. Shields, T. Balle, M. Chebib, and A.N. Clarkson. 2015. Innate immunity and inflammation post-stroke: an alpha7-nicotinic agonist perspective. International Journal of Molecular Sciences 16 (12): 29029–29046.CrossRef
31.
Zurück zum Zitat Carr, Kevin R., S.L. Zuckerman, and J. Mocco. 2013. Inflammation, cerebral vasospasm, and evolving theories of delayed cerebral ischemia. Neurology Research International 2013: 506584.CrossRef Carr, Kevin R., S.L. Zuckerman, and J. Mocco. 2013. Inflammation, cerebral vasospasm, and evolving theories of delayed cerebral ischemia. Neurology Research International 2013: 506584.CrossRef
32.
Zurück zum Zitat Zhang, Hongqiao, K.J.A. Davies, and H.J. Forman. 2015. Oxidative stress response and Nrf2 signaling in aging. Free Radical Biology & Medicine 88 (Pt B): 314–336.CrossRef Zhang, Hongqiao, K.J.A. Davies, and H.J. Forman. 2015. Oxidative stress response and Nrf2 signaling in aging. Free Radical Biology & Medicine 88 (Pt B): 314–336.CrossRef
33.
Zurück zum Zitat Bellezza, Ilaria, I. Giambanco, A. Minelli, and R. Donato. 2018. Nrf2-Keap1 signaling in oxidative and reductive stress. Biochimica et Biophysica Acta, Molecular Cell Research 1865 (5): 721–733.CrossRef Bellezza, Ilaria, I. Giambanco, A. Minelli, and R. Donato. 2018. Nrf2-Keap1 signaling in oxidative and reductive stress. Biochimica et Biophysica Acta, Molecular Cell Research 1865 (5): 721–733.CrossRef
34.
Zurück zum Zitat Shrestha, Amirt K., A. Patel, R.T. Menon, W. Jiang, L. Wang, B. Moorthy, and B. Shivanna. 2017. Leflunomide induces NAD(P)H quinone dehydrogenase 1 enzyme via the aryl hydrocarbon receptor in neonatal mice. Biochemical and Biophysical Research Communications 485 (1): 195–200.CrossRef Shrestha, Amirt K., A. Patel, R.T. Menon, W. Jiang, L. Wang, B. Moorthy, and B. Shivanna. 2017. Leflunomide induces NAD(P)H quinone dehydrogenase 1 enzyme via the aryl hydrocarbon receptor in neonatal mice. Biochemical and Biophysical Research Communications 485 (1): 195–200.CrossRef
35.
Zurück zum Zitat Wu, Kai C., J.Y. Cui, and C.D. Klaassen. 2012. Effect of graded Nrf2 activation on phase-I and -II drug metabolizing enzymes and transporters in mouse liver. PLoS One 7 (7): e39006.CrossRef Wu, Kai C., J.Y. Cui, and C.D. Klaassen. 2012. Effect of graded Nrf2 activation on phase-I and -II drug metabolizing enzymes and transporters in mouse liver. PLoS One 7 (7): e39006.CrossRef
Metadaten
Titel
Ramelteon Ameliorates LPS-Induced Hyperpermeability of the Blood-Brain Barrier (BBB) by Activating Nrf2
verfasst von
Yonglei Liu
Lixia Wang
Ning Du
Xiaoling Yin
Hongtao Shao
Lin Yang
Publikationsdatum
19.04.2021
Verlag
Springer US
Erschienen in
Inflammation / Ausgabe 5/2021
Print ISSN: 0360-3997
Elektronische ISSN: 1573-2576
DOI
https://doi.org/10.1007/s10753-021-01451-w

Weitere Artikel der Ausgabe 5/2021

Inflammation 5/2021 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Blutdrucksenkung könnte Uterusmyome verhindern

Frauen mit unbehandelter oder neu auftretender Hypertonie haben ein deutlich erhöhtes Risiko für Uterusmyome. Eine Therapie mit Antihypertensiva geht hingegen mit einer verringerten Inzidenz der gutartigen Tumoren einher.

„Jeder Fall von plötzlichem Tod muss obduziert werden!“

17.05.2024 Plötzlicher Herztod Nachrichten

Ein signifikanter Anteil der Fälle von plötzlichem Herztod ist genetisch bedingt. Um ihre Verwandten vor diesem Schicksal zu bewahren, sollten jüngere Personen, die plötzlich unerwartet versterben, ausnahmslos einer Autopsie unterzogen werden.

Hirnblutung unter DOAK und VKA ähnlich bedrohlich

17.05.2024 Direkte orale Antikoagulanzien Nachrichten

Kommt es zu einer nichttraumatischen Hirnblutung, spielt es keine große Rolle, ob die Betroffenen zuvor direkt wirksame orale Antikoagulanzien oder Marcumar bekommen haben: Die Prognose ist ähnlich schlecht.

Schlechtere Vorhofflimmern-Prognose bei kleinem linken Ventrikel

17.05.2024 Vorhofflimmern Nachrichten

Nicht nur ein vergrößerter, sondern auch ein kleiner linker Ventrikel ist bei Vorhofflimmern mit einer erhöhten Komplikationsrate assoziiert. Der Zusammenhang besteht nach Daten aus China unabhängig von anderen Risikofaktoren.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.