Skip to main content
Top
Published in: Inflammation 3/2020

01-06-2020 | Septicemia | Original Article

Protective Effect of Dexmedetomidine on Acute Lung Injury via the Upregulation of Tumour Necrosis Factor-α-Induced Protein-8-like 2 in Septic Mice

Authors: Qian Kong, Xiaojing Wu, Zhen Qiu, Qin Huang, Zhongyuan Xia, Xuemin Song

Published in: Inflammation | Issue 3/2020

Login to get access

Abstract

The aim of the present study was to investigate whether TIPE2 participates in the protective actions of dexmedetomidine (DEX) in a mouse model of sepsis-induced acute lung injury (ALI). We administered TIPE2 adeno-associated virus (AAV-TIPE2) intratracheally into the lungs of mice. Control mice were infected with an adeno-associated virus expressing no transgene. Three weeks later, an animal model of caecal ligation-perforation (CLP)-induced sepsis was established. DEX was administered intravenously 30 min after CLP. Twenty-four hours after sepsis, lung injury was assayed by lung histology, the ratio of polymorphonuclear leukocytes (PMNs) to total cells in the bronchoalveolar lavage fluid (BALF), myeloperoxidase (MPO) activity, BALF protein content and the lung wet-to-dry (W/D) weight ratio. Proinflammatory factor levels in the BALF of mice were measured. The protein expression levels in lung tissues were analysed by Western blotting. The results showed that DEX treatment markedly mitigated sepsis-induced lung injury, which was characterized by the deterioration of histopathology, histologic scores, the W/D weight ratio and total protein levels in the BALF. Moreover, DEX markedly attenuated sepsis-induced lung inflammation, as evidenced by the decrease in the number of PMNs in the BALF, lung MPO activity and proinflammatory cytokines in the BALF. In addition, DEX dramatically prevented sepsis-induced pulmonary cell apoptosis in mice, as reflected by decreases in the number of TUNEL-positive cells, the protein expression of cleaved caspase-9 and cleaved caspase 3 and the Bax/Bcl-2 ratio. In addition, evaluation of protein expression showed that DEX blocked sepsis-activated JNK phosphorylation and NF-κB p65 nuclear translocation. Similar results were also observed in the TIPE2 overexpression group. Our study demonstrated that DEX inhibits acute inflammation and apoptosis in a murine model of sepsis-stimulated ALI via the upregulation of TIPE2 and the suppression of the activation of the NF-κB and JNK signalling pathways.
Literature
1.
go back to reference Walkey, A.J., A.R. Kirkpatrick, and R.S. Summer. 2015. Systemic inflammatory response syndrome criteria for severe Sepsis. The New England Journal of Medicine 373 (9): 880.PubMed Walkey, A.J., A.R. Kirkpatrick, and R.S. Summer. 2015. Systemic inflammatory response syndrome criteria for severe Sepsis. The New England Journal of Medicine 373 (9): 880.PubMed
2.
go back to reference Khan, M.M., W.L. Yang, M. Brenner, A.C. Bolognese, and P. Wang. 2017. Cold- inducible RNA-binding protein (CIRP) causes sepsis-associated acute lung injury via induction of endoplasmic reticulum stress. Scientific Reports 7: 41363.PubMedPubMedCentralCrossRef Khan, M.M., W.L. Yang, M. Brenner, A.C. Bolognese, and P. Wang. 2017. Cold- inducible RNA-binding protein (CIRP) causes sepsis-associated acute lung injury via induction of endoplasmic reticulum stress. Scientific Reports 7: 41363.PubMedPubMedCentralCrossRef
3.
go back to reference Li, C., D. Yang, X. Cao, F. Wang, H. Jiang, H. Guo, et al. 2016. LFG-500, a newly synthesized flavonoid, attenuates lipopolysaccharide-induced acute lung injury and inflammation in mice. Biochemical Pharmacology 113: 57–69.PubMedCrossRef Li, C., D. Yang, X. Cao, F. Wang, H. Jiang, H. Guo, et al. 2016. LFG-500, a newly synthesized flavonoid, attenuates lipopolysaccharide-induced acute lung injury and inflammation in mice. Biochemical Pharmacology 113: 57–69.PubMedCrossRef
4.
go back to reference Zhao, Z., X. Tang, X. Zhao, M. Zhang, W. Zhang, S. Hou, et al. 2014. Tylvalosin exhibits anti-inflammatory property and attenuates acute lung injury in different models possibly through suppression of NF-kappaB activation. Biochemical Pharmacology 90 (1): 73–87.PubMedPubMedCentralCrossRef Zhao, Z., X. Tang, X. Zhao, M. Zhang, W. Zhang, S. Hou, et al. 2014. Tylvalosin exhibits anti-inflammatory property and attenuates acute lung injury in different models possibly through suppression of NF-kappaB activation. Biochemical Pharmacology 90 (1): 73–87.PubMedPubMedCentralCrossRef
5.
go back to reference Matthay, M.A., and G.A. Zimmerman. 2005. Acute lung injury and the acute respiratory distress syndrome: Four decades of inquiry into pathogenesis and rational management. American Journal of Respiratory Cell and Molecular Biology 33 (4): 319–327.PubMedPubMedCentralCrossRef Matthay, M.A., and G.A. Zimmerman. 2005. Acute lung injury and the acute respiratory distress syndrome: Four decades of inquiry into pathogenesis and rational management. American Journal of Respiratory Cell and Molecular Biology 33 (4): 319–327.PubMedPubMedCentralCrossRef
6.
go back to reference Singh, G., G. Gladdy, T.T. Chandy, and N. Sen. 2014. Incidence and outcome of acute lung injury and acute respiratory distress syndrome in the surgical intensive care unit. Indian J Crit Care Med 18 (10): 659–665.PubMedPubMedCentralCrossRef Singh, G., G. Gladdy, T.T. Chandy, and N. Sen. 2014. Incidence and outcome of acute lung injury and acute respiratory distress syndrome in the surgical intensive care unit. Indian J Crit Care Med 18 (10): 659–665.PubMedPubMedCentralCrossRef
7.
go back to reference Z'graggen, B.R., J. Tornic, B. Müller-Edenborn, L. Reyes, C. Booy, and B. Beck-Schimmer. 2010. Acute lung injury: Apoptosis in effector and target cells of the upper and lower airway compartment. Clinical and Experimental Immunology 161 (2): 324–331.PubMedPubMedCentral Z'graggen, B.R., J. Tornic, B. Müller-Edenborn, L. Reyes, C. Booy, and B. Beck-Schimmer. 2010. Acute lung injury: Apoptosis in effector and target cells of the upper and lower airway compartment. Clinical and Experimental Immunology 161 (2): 324–331.PubMedPubMedCentral
8.
go back to reference Hotchkiss, R.S., P.E. Swanson, B.D. Freeman, K.W. Tinsley, J.P. Cobb, G.M. Matuschak, et al. 1999. Apoptotic cell death in patients with sepsis, shock, and multiple organ dysfunction. Critical Care Medicine 27 (7): 1230–1251.PubMedCrossRef Hotchkiss, R.S., P.E. Swanson, B.D. Freeman, K.W. Tinsley, J.P. Cobb, G.M. Matuschak, et al. 1999. Apoptotic cell death in patients with sepsis, shock, and multiple organ dysfunction. Critical Care Medicine 27 (7): 1230–1251.PubMedCrossRef
9.
10.
go back to reference Lin, A. 2003. Activation of the JNK signaling pathway: Breaking the brake on apoptosis. Bioessays 25 (1): 17–24.PubMedCrossRef Lin, A. 2003. Activation of the JNK signaling pathway: Breaking the brake on apoptosis. Bioessays 25 (1): 17–24.PubMedCrossRef
11.
go back to reference Cho, J.S., J.K. Shim, S. Soh, M.K. Kim, and Y.L. Kwak. 2016. Perioperative dexmedetomidine reduces the incidence and severity of acute kidney injury following valvular heart surgery. Kidney International 89 (3): 693–700.PubMedCrossRef Cho, J.S., J.K. Shim, S. Soh, M.K. Kim, and Y.L. Kwak. 2016. Perioperative dexmedetomidine reduces the incidence and severity of acute kidney injury following valvular heart surgery. Kidney International 89 (3): 693–700.PubMedCrossRef
12.
go back to reference Taniguchi, T., Y. Kidani, H. Kanakura, Y. Takemoto, and K. Yamamoto. 2004. Effects of dexmedetomidine on mortality rate and inflammatory responses to endotoxin-induced shock in rats. Critical Care Medicine 32 (6): 1322–1326.PubMedCrossRef Taniguchi, T., Y. Kidani, H. Kanakura, Y. Takemoto, and K. Yamamoto. 2004. Effects of dexmedetomidine on mortality rate and inflammatory responses to endotoxin-induced shock in rats. Critical Care Medicine 32 (6): 1322–1326.PubMedCrossRef
13.
go back to reference Sanders, R.D., P. Sun, S. Patel, M. Li, M. Maze, and D. Ma. 2010. Dexmedetomidine provides cortical neuroprotection: Impact on anaesthetic-induced neuroapoptosis in the rat developing brain. Acta Anaesthesiologica Scandinavica 54 (6): 710–716.PubMedCrossRef Sanders, R.D., P. Sun, S. Patel, M. Li, M. Maze, and D. Ma. 2010. Dexmedetomidine provides cortical neuroprotection: Impact on anaesthetic-induced neuroapoptosis in the rat developing brain. Acta Anaesthesiologica Scandinavica 54 (6): 710–716.PubMedCrossRef
14.
go back to reference Zhang, Q., D. Wu, Y. Yang, T. Liu, and H. Liu. 2017. Dexmedetomidine alleviates Hyperoxia-induced acute lung injury via inhibiting NLRP3 Inflammasome activation. Cellular Physiology and Biochemistry 42 (5): 1907–1919.PubMedCrossRef Zhang, Q., D. Wu, Y. Yang, T. Liu, and H. Liu. 2017. Dexmedetomidine alleviates Hyperoxia-induced acute lung injury via inhibiting NLRP3 Inflammasome activation. Cellular Physiology and Biochemistry 42 (5): 1907–1919.PubMedCrossRef
15.
go back to reference Zhang, Y., S. Jia, T. Gao, R. Zhang, Z. Liu, and Y. Wang. 2018. Dexmedetomidine mitigate acute lung injury by inhibiting IL-17-induced inflammatory reaction. Immunobiology 223 (1): 32–37.PubMedCrossRef Zhang, Y., S. Jia, T. Gao, R. Zhang, Z. Liu, and Y. Wang. 2018. Dexmedetomidine mitigate acute lung injury by inhibiting IL-17-induced inflammatory reaction. Immunobiology 223 (1): 32–37.PubMedCrossRef
16.
go back to reference Géloën, A., C. Pichot, S. Leroy, C. Julien, M. Ghignone, C.N. May, et al. 2015. Pressor response to noradrenaline in the setting of septic shock: Anything new under the Sun-Dexmedetomidine, clonidine? A Minireview. BioMed Research International 2015: 863715.PubMedPubMedCentralCrossRef Géloën, A., C. Pichot, S. Leroy, C. Julien, M. Ghignone, C.N. May, et al. 2015. Pressor response to noradrenaline in the setting of septic shock: Anything new under the Sun-Dexmedetomidine, clonidine? A Minireview. BioMed Research International 2015: 863715.PubMedPubMedCentralCrossRef
17.
go back to reference Kumar, D., P. Gokhale, C. Broustas, D. Chakravarty, I. Ahmad, and U. Kasid. 2004. Expression of SCC-S2, an antiapoptotic molecule, correlates with enhanced proliferation and tumorigenicity of MDA-MB 435 cells. Oncogene 23 (2): 612–616.PubMedCrossRef Kumar, D., P. Gokhale, C. Broustas, D. Chakravarty, I. Ahmad, and U. Kasid. 2004. Expression of SCC-S2, an antiapoptotic molecule, correlates with enhanced proliferation and tumorigenicity of MDA-MB 435 cells. Oncogene 23 (2): 612–616.PubMedCrossRef
18.
go back to reference Sun, H., S. Gong, R.J. Carmody, A. Hilliard, L. Li, J. Sun, et al. 2008. TIPE2, a negative regulator of innate and adaptive immunity that maintains immune homeostasis. Cell 133 (3): 415–426.PubMedPubMedCentralCrossRef Sun, H., S. Gong, R.J. Carmody, A. Hilliard, L. Li, J. Sun, et al. 2008. TIPE2, a negative regulator of innate and adaptive immunity that maintains immune homeostasis. Cell 133 (3): 415–426.PubMedPubMedCentralCrossRef
19.
go back to reference Siempos, I.I., H.C. Lam, Y. Ding, M.E. Choi, A.M. Choi, and S.W. Ryter. 2014. Cecal ligation and puncture-induced sepsis as a model to study autophagy in mice. Journal of Visualized Experiments 84: e51066. Siempos, I.I., H.C. Lam, Y. Ding, M.E. Choi, A.M. Choi, and S.W. Ryter. 2014. Cecal ligation and puncture-induced sepsis as a model to study autophagy in mice. Journal of Visualized Experiments 84: e51066.
20.
go back to reference Jeremias, I.C., V.J. Victorino, H.V. Barbeiro, S.A. Kubo, C.M. Prado, T.M. Lima, et al. 2016. The role of acetylcholine in the inflammatory response in animals surviving Sepsis induced by Cecal ligation and puncture. Molecular Neurobiology 53 (10): 6635–6643.PubMedCrossRef Jeremias, I.C., V.J. Victorino, H.V. Barbeiro, S.A. Kubo, C.M. Prado, T.M. Lima, et al. 2016. The role of acetylcholine in the inflammatory response in animals surviving Sepsis induced by Cecal ligation and puncture. Molecular Neurobiology 53 (10): 6635–6643.PubMedCrossRef
21.
go back to reference Su, X., L. Wang, Y. Song, and C. Bai. 2004. Inhibition of inflammatory responses by ambroxol, a mucolytic agent, in a murine model of acute lung injury induced by lipopolysaccharide. Intensive Care Medicine 30 (1): 133–140.PubMedCrossRefPubMedCentral Su, X., L. Wang, Y. Song, and C. Bai. 2004. Inhibition of inflammatory responses by ambroxol, a mucolytic agent, in a murine model of acute lung injury induced by lipopolysaccharide. Intensive Care Medicine 30 (1): 133–140.PubMedCrossRefPubMedCentral
22.
go back to reference Johnson, E.R., and M.A. Matthay. 2010. Acute lung injury: Epidemiology, pathogenesis, and treatment. Journal of Aerosol Medicine and Pulmonary Drug Delivery 23 (4): 243–252.PubMedPubMedCentralCrossRef Johnson, E.R., and M.A. Matthay. 2010. Acute lung injury: Epidemiology, pathogenesis, and treatment. Journal of Aerosol Medicine and Pulmonary Drug Delivery 23 (4): 243–252.PubMedPubMedCentralCrossRef
23.
go back to reference Pandharipande, P.P., R.D. Sanders, T.D. Girard, S. McGrane, J.L. Thompson, A.K. Shintani, et al. 2010. Effect of dexmedetomidine versus lorazepam on outcome in patients with sepsis: An a priori-designed analysis of the MENDS randomized controlled trial. Critical Care 14 (2): R38.PubMedPubMedCentralCrossRef Pandharipande, P.P., R.D. Sanders, T.D. Girard, S. McGrane, J.L. Thompson, A.K. Shintani, et al. 2010. Effect of dexmedetomidine versus lorazepam on outcome in patients with sepsis: An a priori-designed analysis of the MENDS randomized controlled trial. Critical Care 14 (2): R38.PubMedPubMedCentralCrossRef
24.
go back to reference Hu, H., D. Shi, C. Hu, X. Yuan, J. Zhang, and H. Sun. 2017. Dexmedetomidine mitigates CLP-stimulated acute lung injury via restraining the RAGE pathway. American Journal of Translational Research 9 (12): 5245–5258.PubMedPubMedCentral Hu, H., D. Shi, C. Hu, X. Yuan, J. Zhang, and H. Sun. 2017. Dexmedetomidine mitigates CLP-stimulated acute lung injury via restraining the RAGE pathway. American Journal of Translational Research 9 (12): 5245–5258.PubMedPubMedCentral
26.
go back to reference Xi, W., Y. Hu, Y. Liu, J. Zhang, L. Wang, Y. Lou, et al. 2011. Roles of TIPE2 in hepatitis B virus-induced hepatic inflammation in humans and mice. Molecular Immunology 48 (9–10): 1203–1208.PubMedCrossRef Xi, W., Y. Hu, Y. Liu, J. Zhang, L. Wang, Y. Lou, et al. 2011. Roles of TIPE2 in hepatitis B virus-induced hepatic inflammation in humans and mice. Molecular Immunology 48 (9–10): 1203–1208.PubMedCrossRef
27.
go back to reference Li, D., L. Song, Y. Fan, X. Li, Y. Li, J. Chen, et al. 2009. Down-regulation of TIPE2 mRNA expression in peripheral blood mononuclear cells from patients with systemic lupus erythematosus. Clinical Immunology 133 (3): 422–427.PubMedCrossRef Li, D., L. Song, Y. Fan, X. Li, Y. Li, J. Chen, et al. 2009. Down-regulation of TIPE2 mRNA expression in peripheral blood mononuclear cells from patients with systemic lupus erythematosus. Clinical Immunology 133 (3): 422–427.PubMedCrossRef
28.
go back to reference Kumar, D., T.L. Whiteside, and U. Kasid. 2000. Identification of a novel tumor necrosis factor-alpha-inducible gene, SCC-S2, containing the consensus sequence of a death effector domain of fas-associated death domain-like interleukin- 1beta-converting enzyme-inhibitory protein. The Journal of Biological Chemistry 275 (4): 2973–2978.PubMedCrossRef Kumar, D., T.L. Whiteside, and U. Kasid. 2000. Identification of a novel tumor necrosis factor-alpha-inducible gene, SCC-S2, containing the consensus sequence of a death effector domain of fas-associated death domain-like interleukin- 1beta-converting enzyme-inhibitory protein. The Journal of Biological Chemistry 275 (4): 2973–2978.PubMedCrossRef
29.
go back to reference Yan, X., X. Cheng, L. Zhou, X. He, W. Zheng, and H. Chen. 2017. Dexmedetomidine alleviates lipopolysaccharide-induced lung injury in Wistar rats. Oncotarget 8 (27): 44410–44417.PubMedPubMedCentralCrossRef Yan, X., X. Cheng, L. Zhou, X. He, W. Zheng, and H. Chen. 2017. Dexmedetomidine alleviates lipopolysaccharide-induced lung injury in Wistar rats. Oncotarget 8 (27): 44410–44417.PubMedPubMedCentralCrossRef
30.
go back to reference Buras, J.A., B. Holzmann, and M. Sitkovsky. 2005. Animal models of sepsis: Setting the stage. Nature Reviews. Drug Discovery 4 (10): 854–865.PubMedCrossRef Buras, J.A., B. Holzmann, and M. Sitkovsky. 2005. Animal models of sepsis: Setting the stage. Nature Reviews. Drug Discovery 4 (10): 854–865.PubMedCrossRef
31.
go back to reference Hubbard, W.J., M. Choudhry, M.G. Schwacha, J.D. Kerby, L.W. Rue, K.I. Bland, et al. 2005. Cecal ligation and puncture. Shock 24 (Suppl 1): 52–57.PubMedCrossRef Hubbard, W.J., M. Choudhry, M.G. Schwacha, J.D. Kerby, L.W. Rue, K.I. Bland, et al. 2005. Cecal ligation and puncture. Shock 24 (Suppl 1): 52–57.PubMedCrossRef
32.
go back to reference Abraham, E., and M. Singer. 2007. Mechanisms of sepsis-induced organ dysfunction. Critical Care Medicine 35 (10): 2408–2416.PubMedCrossRef Abraham, E., and M. Singer. 2007. Mechanisms of sepsis-induced organ dysfunction. Critical Care Medicine 35 (10): 2408–2416.PubMedCrossRef
33.
34.
go back to reference Brown, K.A., S.D. Brain, J.D. Pearson, J.D. Edgeworth, S.M. Lewis, and D.F. Treacher. 2006. Neutrophils in development of multiple organ failure in sepsis. Lancet 368 (9530): 157–169.PubMedCrossRef Brown, K.A., S.D. Brain, J.D. Pearson, J.D. Edgeworth, S.M. Lewis, and D.F. Treacher. 2006. Neutrophils in development of multiple organ failure in sepsis. Lancet 368 (9530): 157–169.PubMedCrossRef
35.
go back to reference Yang, K.Y., J.J. Arcaroli, and E. Abraham. 2003. Early alterations in neutrophil activation are associated with outcome in acute lung injury. American Journal of Respiratory and Critical Care Medicine 167 (11): 1567–1574.PubMedCrossRef Yang, K.Y., J.J. Arcaroli, and E. Abraham. 2003. Early alterations in neutrophil activation are associated with outcome in acute lung injury. American Journal of Respiratory and Critical Care Medicine 167 (11): 1567–1574.PubMedCrossRef
36.
go back to reference Zhang, H.X., S.J. Liu, X.L. Tang, G.L. Duan, X. Ni, X.Y. Zhu, et al. 2016. H2S attenuates LPS-induced acute lung injury by reducing oxidative/Nitrative stress and inflammation. Cellular Physiology and Biochemistry 40 (6): 1603–1612.PubMedCrossRef Zhang, H.X., S.J. Liu, X.L. Tang, G.L. Duan, X. Ni, X.Y. Zhu, et al. 2016. H2S attenuates LPS-induced acute lung injury by reducing oxidative/Nitrative stress and inflammation. Cellular Physiology and Biochemistry 40 (6): 1603–1612.PubMedCrossRef
37.
go back to reference Grommes, J., and O. Soehnlein. 2011. Contribution of neutrophils to acute lung injury. Molecular Medicine 17 (3–4): 293–307.PubMedCrossRef Grommes, J., and O. Soehnlein. 2011. Contribution of neutrophils to acute lung injury. Molecular Medicine 17 (3–4): 293–307.PubMedCrossRef
38.
go back to reference Hirano, Y., M. Aziz, W.L. Yang, Z. Wang, M. Zhou, M. Ochani, et al. 2015. Neutralization of osteopontin attenuates neutrophil migration in sepsis-induced acute lung injury. Critical Care 19: 53.PubMedPubMedCentralCrossRef Hirano, Y., M. Aziz, W.L. Yang, Z. Wang, M. Zhou, M. Ochani, et al. 2015. Neutralization of osteopontin attenuates neutrophil migration in sepsis-induced acute lung injury. Critical Care 19: 53.PubMedPubMedCentralCrossRef
39.
go back to reference Liu, S.F., and A.B. Malik. 2006. NF-kappa B activation as a pathological mechanism of septic shock and inflammation. American Journal of Physiology. Lung Cellular and Molecular Physiology 290 (4): L622–L645.PubMedCrossRef Liu, S.F., and A.B. Malik. 2006. NF-kappa B activation as a pathological mechanism of septic shock and inflammation. American Journal of Physiology. Lung Cellular and Molecular Physiology 290 (4): L622–L645.PubMedCrossRef
40.
go back to reference Wang, H., L. Wang, N.L. Li, J.T. Li, F. Yu, Y.L. Zhao, et al. 2014. Subanesthetic isoflurane reduces zymosan-induced inflammation in murine Kupffer cells by inhibiting ROS-activated p38 MAPK/NF-kappaB signaling. Oxidative Medicine and Cellular Longevity 2014: 851692.PubMedPubMedCentral Wang, H., L. Wang, N.L. Li, J.T. Li, F. Yu, Y.L. Zhao, et al. 2014. Subanesthetic isoflurane reduces zymosan-induced inflammation in murine Kupffer cells by inhibiting ROS-activated p38 MAPK/NF-kappaB signaling. Oxidative Medicine and Cellular Longevity 2014: 851692.PubMedPubMedCentral
41.
go back to reference Blackwell, T.S., T.R. Blackwell, E.P. Holden, B.W. Christman, and J.W. 1996. Christman. In vivo antioxidant treatment suppresses nuclear factor-kappa B activation and neutrophilic lung inflammation. Journal of Immunology 157 (4): 1630–1637. Blackwell, T.S., T.R. Blackwell, E.P. Holden, B.W. Christman, and J.W. 1996. Christman. In vivo antioxidant treatment suppresses nuclear factor-kappa B activation and neutrophilic lung inflammation. Journal of Immunology 157 (4): 1630–1637.
42.
go back to reference Galani, V., E. Tatsaki, M. Bai, P. Kitsoulis, M. Lekka, G. Nakos, et al. 2010. The role of apoptosis in the pathophysiology of acute respiratory distress syndrome (ARDS): An up-to-date cell-specific review. Pathology, Research and Practice 206 (3): 145–150.PubMedCrossRef Galani, V., E. Tatsaki, M. Bai, P. Kitsoulis, M. Lekka, G. Nakos, et al. 2010. The role of apoptosis in the pathophysiology of acute respiratory distress syndrome (ARDS): An up-to-date cell-specific review. Pathology, Research and Practice 206 (3): 145–150.PubMedCrossRef
43.
go back to reference Chopra, M., J.S. Reuben, and A.C. Sharma. 2009. Acute lung injury:Apoptosis and signaling mechanisms. Experimental Biology and Medicine (Maywood, N.J.) 234 (4): 361–371.CrossRef Chopra, M., J.S. Reuben, and A.C. Sharma. 2009. Acute lung injury:Apoptosis and signaling mechanisms. Experimental Biology and Medicine (Maywood, N.J.) 234 (4): 361–371.CrossRef
44.
go back to reference Perl, M., C.S. Chung, U. Perl, R. Thakkar, J. Lomas-Neira, and A. Ayala. 2010. Therapeutic accessibility of caspase-mediated cell death as a key pathomechanism in indirect acute lung injury. Critical Care Medicine 38 (4): 1179–1186.PubMedCrossRef Perl, M., C.S. Chung, U. Perl, R. Thakkar, J. Lomas-Neira, and A. Ayala. 2010. Therapeutic accessibility of caspase-mediated cell death as a key pathomechanism in indirect acute lung injury. Critical Care Medicine 38 (4): 1179–1186.PubMedCrossRef
45.
go back to reference Porter, A.G., and R.U. Janicke. 1999. Emerging roles of caspase-3 in apoptosis. Cell Death and Differentiation 6 (2): 99–104.PubMedCrossRef Porter, A.G., and R.U. Janicke. 1999. Emerging roles of caspase-3 in apoptosis. Cell Death and Differentiation 6 (2): 99–104.PubMedCrossRef
46.
go back to reference Lee, I.T., and C.M. Yang. 2013. Inflammatory signalings involved in airway and pulmonary diseases. Mediators of Inflammation 2013: 791231.PubMedPubMedCentral Lee, I.T., and C.M. Yang. 2013. Inflammatory signalings involved in airway and pulmonary diseases. Mediators of Inflammation 2013: 791231.PubMedPubMedCentral
47.
go back to reference Kyriakis, J.M., P. Banerjee, E. Nikolakaki, T. Dai, E.A. Rubie, M.F. Ahmad, et al. 1994. The stress-activated protein kinase subfamily of c-Jun kinases. Nature 369 (6476): 156–160.PubMedCrossRef Kyriakis, J.M., P. Banerjee, E. Nikolakaki, T. Dai, E.A. Rubie, M.F. Ahmad, et al. 1994. The stress-activated protein kinase subfamily of c-Jun kinases. Nature 369 (6476): 156–160.PubMedCrossRef
48.
go back to reference Sui, X., N. Kong, L. Ye, W. Han, J. Zhou, Q. Zhang, et al. 2014. p38 and JNK MAPK pathways control the balance of apoptosis and autophagy in response to chemotherapeutic agents. Cancer Letters 344 (2): 174–179.PubMedCrossRef Sui, X., N. Kong, L. Ye, W. Han, J. Zhou, Q. Zhang, et al. 2014. p38 and JNK MAPK pathways control the balance of apoptosis and autophagy in response to chemotherapeutic agents. Cancer Letters 344 (2): 174–179.PubMedCrossRef
49.
go back to reference Kluck, R.M., E. Bossy-Wetzel, D.R. Green, and D.D. Newmeyer. 1997. The release of cytochrome c from mitochondria: A primary site for Bcl-2 regulation of apoptosis. Science 275 (5303): 1132–1136.PubMedCrossRef Kluck, R.M., E. Bossy-Wetzel, D.R. Green, and D.D. Newmeyer. 1997. The release of cytochrome c from mitochondria: A primary site for Bcl-2 regulation of apoptosis. Science 275 (5303): 1132–1136.PubMedCrossRef
50.
go back to reference Li, B., M. Zeng, W. He, X. Huang, L. Luo, H. Zhang, et al. 2015. Ghrelin protects alveolar macrophages against lipopolysaccharide-induced apoptosis through growth hormone secretagogue receptor 1a-dependent c-Jun N-terminal kinase and Wnt/beta-catenin signaling and suppresses lung inflammation. Endocrinology 156 (1): 203–217.PubMedCrossRef Li, B., M. Zeng, W. He, X. Huang, L. Luo, H. Zhang, et al. 2015. Ghrelin protects alveolar macrophages against lipopolysaccharide-induced apoptosis through growth hormone secretagogue receptor 1a-dependent c-Jun N-terminal kinase and Wnt/beta-catenin signaling and suppresses lung inflammation. Endocrinology 156 (1): 203–217.PubMedCrossRef
Metadata
Title
Protective Effect of Dexmedetomidine on Acute Lung Injury via the Upregulation of Tumour Necrosis Factor-α-Induced Protein-8-like 2 in Septic Mice
Authors
Qian Kong
Xiaojing Wu
Zhen Qiu
Qin Huang
Zhongyuan Xia
Xuemin Song
Publication date
01-06-2020
Publisher
Springer US
Published in
Inflammation / Issue 3/2020
Print ISSN: 0360-3997
Electronic ISSN: 1573-2576
DOI
https://doi.org/10.1007/s10753-019-01169-w

Other articles of this Issue 3/2020

Inflammation 3/2020 Go to the issue