Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2016

Open Access 01-12-2016 | Research

Heme oxygenase-1 promotes neuron survival through down-regulation of neuronal NLRP1 expression after spinal cord injury

Authors: Wen-Ping Lin, Gong-Peng Xiong, Qing Lin, Xuan-Wei Chen, Li-Qun Zhang, Jin-Xing Shi, Qing-Feng Ke, Jian-Hua Lin

Published in: Journal of Neuroinflammation | Issue 1/2016

Login to get access

Abstract

Background

Understanding the mechanisms underlying neuronal death in spinal cord injury (SCI) and developing novel therapeutic approaches for SCI-induced damage are critical for functional recovery. Here we investigated the role of heme oxygenase-1 (HO-1) in neuroprotection after SCI.

Methods

Adeno-associated virus expressing HO-1 was prepared and injected into rat spinal cords before SCI model was performed. HO-1 expression, inflammasome activation, and the presence of inflammatory cytokines were determined by quantitative polymerase chain reaction, immunohistological staining, immunoblot, and immunoprecipitation. Neuronal apoptosis was assessed by terminal deoxynucleotidyl transferase dUTP nick end labeling. The hindlimb locomotor function was evaluated for extent of neurologic damage. In an in vitro model, hydrogen peroxide was used to induce similar inflammasome activation in cultured primary spinal cord neurons, followed by evaluation of above parameters with or without transduction of HO-1-expressing adeno-associated virus.

Results

Endogenous HO-1 expression was found in spinal cord neurons after SCI in vivo, in association with the expression of Nod-like receptor protein 1 (NLRP1) and the formation of NLRP1 inflammasomes. Administration of HO-1-expressing adeno-associated virus effectively decreased expression of NLRP1, therefore alleviating NLRP1 inflammasome-induced neuronal death and improving functional recovery. In the in vitro model, exogenous HO-1 expression protected neurons from hydrogen peroxide-induced neuronal death by inhibiting NLRP1 expression. In addition, HO-1 inhibited expression of activating transcription factor 4 (ATF4), which is a transcription factor regulating NLRP1 expression.

Conclusions

HO-1 protects spinal cord neurons after SCI through inhibiting NLRP1 inflammasome formation.
Appendix
Available only for authorised users
Literature
1.
go back to reference Zhang N, Yin Y, Xu SJ, Wu YP, Chen WS. Inflammation & apoptosis in spinal cord injury. Indian J Med Res. 2012;135:287–96.PubMedPubMedCentral Zhang N, Yin Y, Xu SJ, Wu YP, Chen WS. Inflammation & apoptosis in spinal cord injury. Indian J Med Res. 2012;135:287–96.PubMedPubMedCentral
2.
go back to reference Liu NK, Zhang YP, Titsworth WL, Jiang X, Han S, Lu PH, et al. A novel role of phospholipase A2 in mediating spinal cord secondary injury. Ann Neurol. 2006;59:606–19.CrossRefPubMed Liu NK, Zhang YP, Titsworth WL, Jiang X, Han S, Lu PH, et al. A novel role of phospholipase A2 in mediating spinal cord secondary injury. Ann Neurol. 2006;59:606–19.CrossRefPubMed
3.
go back to reference Ren Y, Young W. Managing inflammation after spinal cord injury through manipulation of macrophage function. Neural Plast. 2013;2013:945034.PubMedPubMedCentral Ren Y, Young W. Managing inflammation after spinal cord injury through manipulation of macrophage function. Neural Plast. 2013;2013:945034.PubMedPubMedCentral
4.
go back to reference Liu XZ, Xu XM, Hu R, Du C, Zhang SX, McDonald JW, et al. Neuronal and glial apoptosis after traumatic spinal cord injury. J Neurosci. 1997;17:5395–406.PubMed Liu XZ, Xu XM, Hu R, Du C, Zhang SX, McDonald JW, et al. Neuronal and glial apoptosis after traumatic spinal cord injury. J Neurosci. 1997;17:5395–406.PubMed
5.
go back to reference Di Giovanni S, Knoblach SM, Brandoli C, Aden SA, Hoffman EP, Faden AI. Gene profiling in spinal cord injury shows role of cell cycle in neuronal death. Ann Neurol. 2003;53:454–68.CrossRefPubMed Di Giovanni S, Knoblach SM, Brandoli C, Aden SA, Hoffman EP, Faden AI. Gene profiling in spinal cord injury shows role of cell cycle in neuronal death. Ann Neurol. 2003;53:454–68.CrossRefPubMed
6.
go back to reference Beattie MS, Hermann GE, Rogers RC, Bresnahan JC. Cell death in models of spinal cord injury. Prog Brain Res. 2002;137:37–47.CrossRefPubMed Beattie MS, Hermann GE, Rogers RC, Bresnahan JC. Cell death in models of spinal cord injury. Prog Brain Res. 2002;137:37–47.CrossRefPubMed
7.
go back to reference Liu S, Sarkar C, Dinizo M, Faden AI, Koh EY, Lipinski MM, et al. Disrupted autophagy after spinal cord injury is associated with ER stress and neuronal cell death. Cell Death Dis. 2015;6, e1582.CrossRefPubMedPubMedCentral Liu S, Sarkar C, Dinizo M, Faden AI, Koh EY, Lipinski MM, et al. Disrupted autophagy after spinal cord injury is associated with ER stress and neuronal cell death. Cell Death Dis. 2015;6, e1582.CrossRefPubMedPubMedCentral
8.
go back to reference Park E, Velumian AA, Fehlings MG. The role of excitotoxicity in secondary mechanisms of spinal cord injury: a review with an emphasis on the implications for white matter degeneration. J Neurotrauma. 2004;21:754–74.CrossRefPubMed Park E, Velumian AA, Fehlings MG. The role of excitotoxicity in secondary mechanisms of spinal cord injury: a review with an emphasis on the implications for white matter degeneration. J Neurotrauma. 2004;21:754–74.CrossRefPubMed
9.
go back to reference Kaushal V, Dye R, Pakavathkumar P, Foveau B, Flores J, Hyman B, et al. Neuronal NLRP1 inflammasome activation of Caspase-1 coordinately regulates inflammatory interleukin-1-beta production and axonal degeneration-associated Caspase-6 activation. Cell Death Differ. 2015;22:1676–86.CrossRefPubMed Kaushal V, Dye R, Pakavathkumar P, Foveau B, Flores J, Hyman B, et al. Neuronal NLRP1 inflammasome activation of Caspase-1 coordinately regulates inflammatory interleukin-1-beta production and axonal degeneration-associated Caspase-6 activation. Cell Death Differ. 2015;22:1676–86.CrossRefPubMed
10.
go back to reference Adamczak SE, de Rivero Vaccari JP, Dale G, Brand 3rd FJ, Nonner D, Bullock MR, et al. Pyroptotic neuronal cell death mediated by the AIM2 inflammasome. J Cereb Blood Flow Metab. 2014;34:621–9.CrossRefPubMedPubMedCentral Adamczak SE, de Rivero Vaccari JP, Dale G, Brand 3rd FJ, Nonner D, Bullock MR, et al. Pyroptotic neuronal cell death mediated by the AIM2 inflammasome. J Cereb Blood Flow Metab. 2014;34:621–9.CrossRefPubMedPubMedCentral
11.
go back to reference Fann DY, Lee SY, Manzanero S, Tang SC, Gelderblom M, Chunduri P, et al. Intravenous immunoglobulin suppresses NLRP1 and NLRP3 inflammasome-mediated neuronal death in ischemic stroke. Cell Death Dis. 2013;4, e790.CrossRefPubMed Fann DY, Lee SY, Manzanero S, Tang SC, Gelderblom M, Chunduri P, et al. Intravenous immunoglobulin suppresses NLRP1 and NLRP3 inflammasome-mediated neuronal death in ischemic stroke. Cell Death Dis. 2013;4, e790.CrossRefPubMed
12.
go back to reference Tan CC, Zhang JG, Tan MS, Chen H, Meng DW, Jiang T, et al. NLRP1 inflammasome is activated in patients with medial temporal lobe epilepsy and contributes to neuronal pyroptosis in amygdala kindling-induced rat model. J Neuroinflammation. 2015;12:18.CrossRefPubMedPubMedCentral Tan CC, Zhang JG, Tan MS, Chen H, Meng DW, Jiang T, et al. NLRP1 inflammasome is activated in patients with medial temporal lobe epilepsy and contributes to neuronal pyroptosis in amygdala kindling-induced rat model. J Neuroinflammation. 2015;12:18.CrossRefPubMedPubMedCentral
13.
go back to reference Jorgensen I, Miao EA. Pyroptotic cell death defends against intracellular pathogens. Immunol Rev. 2015;265:130–42.CrossRefPubMed Jorgensen I, Miao EA. Pyroptotic cell death defends against intracellular pathogens. Immunol Rev. 2015;265:130–42.CrossRefPubMed
14.
go back to reference Martinon F, Burns K, Tschopp J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell. 2002;10:417–26.CrossRefPubMed Martinon F, Burns K, Tschopp J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell. 2002;10:417–26.CrossRefPubMed
16.
go back to reference Latz E, Xiao TS, Stutz A. Activation and regulation of the inflammasomes. Nat Rev Immunol. 2013;13:397–411.CrossRefPubMed Latz E, Xiao TS, Stutz A. Activation and regulation of the inflammasomes. Nat Rev Immunol. 2013;13:397–411.CrossRefPubMed
17.
go back to reference Tringali G, Mancuso C, Mirtella A, Pozzoli G, Parente L, Preziosi P, et al. Evidence for the neuronal origin of immunoreactive interleukin-1 beta released by rat hypothalamic explants. Neurosci Lett. 1996;219:143–6.CrossRefPubMed Tringali G, Mancuso C, Mirtella A, Pozzoli G, Parente L, Preziosi P, et al. Evidence for the neuronal origin of immunoreactive interleukin-1 beta released by rat hypothalamic explants. Neurosci Lett. 1996;219:143–6.CrossRefPubMed
18.
go back to reference Tringali G, Mirtella A, Mancuso C, Guerriero G, Preziosi P, Navarra P. The release of immunoreactive interleukin-1 beta from rat hypothalamic explants is modulated by neurotransmitters and corticotropin-releasing hormone. Pharmacol Res. 1997;36:269–73.CrossRefPubMed Tringali G, Mirtella A, Mancuso C, Guerriero G, Preziosi P, Navarra P. The release of immunoreactive interleukin-1 beta from rat hypothalamic explants is modulated by neurotransmitters and corticotropin-releasing hormone. Pharmacol Res. 1997;36:269–73.CrossRefPubMed
19.
go back to reference Abulafia DP, de Rivero Vaccari JP, Lozano JD, Lotocki G, Keane RW, Dietrich WD. Inhibition of the inflammasome complex reduces the inflammatory response after thromboembolic stroke in mice. J Cereb Blood Flow Metab. 2009;29:534–44.CrossRefPubMed Abulafia DP, de Rivero Vaccari JP, Lozano JD, Lotocki G, Keane RW, Dietrich WD. Inhibition of the inflammasome complex reduces the inflammatory response after thromboembolic stroke in mice. J Cereb Blood Flow Metab. 2009;29:534–44.CrossRefPubMed
20.
go back to reference de Rivero Vaccari JP, Lotocki G, Alonso OF, Bramlett HM, Dietrich WD, Keane RW. Therapeutic neutralization of the NLRP1 inflammasome reduces the innate immune response and improves histopathology after traumatic brain injury. J Cereb Blood Flow Metab. 2009;29:1251–61.CrossRefPubMedPubMedCentral de Rivero Vaccari JP, Lotocki G, Alonso OF, Bramlett HM, Dietrich WD, Keane RW. Therapeutic neutralization of the NLRP1 inflammasome reduces the innate immune response and improves histopathology after traumatic brain injury. J Cereb Blood Flow Metab. 2009;29:1251–61.CrossRefPubMedPubMedCentral
21.
go back to reference de Rivero Vaccari JP, Brand F, 3rd, Adamczak S, Lee SW, Barcena JP, Wang MY, Bullock MR, Dietrich WD, Keane RW. Exosome-mediated inflammasome signaling after central nervous system injury. J Neurochem. 2015;21(Suppl 1):39-48. de Rivero Vaccari JP, Brand F, 3rd, Adamczak S, Lee SW, Barcena JP, Wang MY, Bullock MR, Dietrich WD, Keane RW. Exosome-mediated inflammasome signaling after central nervous system injury. J Neurochem. 2015;21(Suppl 1):39-48.
22.
go back to reference Paine A, Eiz-Vesper B, Blasczyk R, Immenschuh S. Signaling to heme oxygenase-1 and its anti-inflammatory therapeutic potential. Biochem Pharmacol. 2010;80:1895–903.CrossRefPubMed Paine A, Eiz-Vesper B, Blasczyk R, Immenschuh S. Signaling to heme oxygenase-1 and its anti-inflammatory therapeutic potential. Biochem Pharmacol. 2010;80:1895–903.CrossRefPubMed
23.
go back to reference Mancuso C, Barone E. The heme oxygenase/biliverdin reductase pathway in drug research and development. Curr Drug Metab. 2009;10:579–94.CrossRefPubMed Mancuso C, Barone E. The heme oxygenase/biliverdin reductase pathway in drug research and development. Curr Drug Metab. 2009;10:579–94.CrossRefPubMed
24.
go back to reference Raju VS, McCoubrey Jr WK, Maines MD. Regulation of heme oxygenase-2 by glucocorticoids in neonatal rat brain: characterization of a functional glucocorticoid response element. Biochim Biophys Acta. 1997;1351:89–104.CrossRefPubMed Raju VS, McCoubrey Jr WK, Maines MD. Regulation of heme oxygenase-2 by glucocorticoids in neonatal rat brain: characterization of a functional glucocorticoid response element. Biochim Biophys Acta. 1997;1351:89–104.CrossRefPubMed
25.
go back to reference Maines MD. The heme oxygenase system: a regulator of second messenger gases. Annu Rev Pharmacol Toxicol. 1997;37:517–54.CrossRefPubMed Maines MD. The heme oxygenase system: a regulator of second messenger gases. Annu Rev Pharmacol Toxicol. 1997;37:517–54.CrossRefPubMed
26.
go back to reference Okinaga S, Takahashi K, Takeda K, Yoshizawa M, Fujita H, Sasaki H, et al. Regulation of human heme oxygenase-1 gene expression under thermal stress. Blood. 1996;87:5074–84.PubMed Okinaga S, Takahashi K, Takeda K, Yoshizawa M, Fujita H, Sasaki H, et al. Regulation of human heme oxygenase-1 gene expression under thermal stress. Blood. 1996;87:5074–84.PubMed
27.
go back to reference Cantoni L, Rossi C, Rizzardini M, Gadina M, Ghezzi P. Interleukin-1 and tumour necrosis factor induce hepatic haem oxygenase. Feedback regulation by glucocorticoids. Biochem J. 1991;279(Pt 3):891–4.CrossRefPubMedPubMedCentral Cantoni L, Rossi C, Rizzardini M, Gadina M, Ghezzi P. Interleukin-1 and tumour necrosis factor induce hepatic haem oxygenase. Feedback regulation by glucocorticoids. Biochem J. 1991;279(Pt 3):891–4.CrossRefPubMedPubMedCentral
28.
go back to reference Gozzelino R, Jeney V, Soares MP. Mechanisms of cell protection by heme oxygenase-1. Annu Rev Pharmacol Toxicol. 2010;50:323–54.CrossRefPubMed Gozzelino R, Jeney V, Soares MP. Mechanisms of cell protection by heme oxygenase-1. Annu Rev Pharmacol Toxicol. 2010;50:323–54.CrossRefPubMed
29.
go back to reference Mancuso C, Preziosi P, Grossman AB, Navarra P. The role of carbon monoxide in the regulation of neuroendocrine function. Neuroimmunomodulation. 1997;4:225–9.PubMed Mancuso C, Preziosi P, Grossman AB, Navarra P. The role of carbon monoxide in the regulation of neuroendocrine function. Neuroimmunomodulation. 1997;4:225–9.PubMed
30.
go back to reference Mancuso C, Ragazzoni E, Tringali G, Liberale I, Preziosi P, Grossman A, et al. Inhibition of heme oxygenase in the central nervous system potentiates endotoxin-induced vasopressin release in the rat. J Neuroimmunol. 1999;99:189–94.CrossRefPubMed Mancuso C, Ragazzoni E, Tringali G, Liberale I, Preziosi P, Grossman A, et al. Inhibition of heme oxygenase in the central nervous system potentiates endotoxin-induced vasopressin release in the rat. J Neuroimmunol. 1999;99:189–94.CrossRefPubMed
31.
go back to reference Panahian N, Maines MD. Site of injury-directed induction of heme oxygenase-1 and -2 in experimental spinal cord injury: differential functions in neuronal defense mechanisms? J Neurochem. 2001;76:539–54.CrossRefPubMed Panahian N, Maines MD. Site of injury-directed induction of heme oxygenase-1 and -2 in experimental spinal cord injury: differential functions in neuronal defense mechanisms? J Neurochem. 2001;76:539–54.CrossRefPubMed
32.
go back to reference Yamada K, Tanaka N, Nakanishi K, Kamei N, Ishikawa M, Mizuno T, et al. Modulation of the secondary injury process after spinal cord injury in Bach1-deficient mice by heme oxygenase-1. J Neurosurg Spine. 2008;9:611–20.CrossRefPubMed Yamada K, Tanaka N, Nakanishi K, Kamei N, Ishikawa M, Mizuno T, et al. Modulation of the secondary injury process after spinal cord injury in Bach1-deficient mice by heme oxygenase-1. J Neurosurg Spine. 2008;9:611–20.CrossRefPubMed
33.
go back to reference Lin Y, Vreman HJ, Wong RJ, Tjoa T, Yamauchi T, Noble-Haeusslein LJ. Heme oxygenase-1 stabilizes the blood-spinal cord barrier and limits oxidative stress and white matter damage in the acutely injured murine spinal cord. J Cereb Blood Flow Metab. 2007;27:1010–21.PubMed Lin Y, Vreman HJ, Wong RJ, Tjoa T, Yamauchi T, Noble-Haeusslein LJ. Heme oxygenase-1 stabilizes the blood-spinal cord barrier and limits oxidative stress and white matter damage in the acutely injured murine spinal cord. J Cereb Blood Flow Metab. 2007;27:1010–21.PubMed
34.
go back to reference Hall JC, Priestley JV, Perry VH, Michael-Titus AT. Docosahexaenoic acid, but not eicosapentaenoic acid, reduces the early inflammatory response following compression spinal cord injury in the rat. J Neurochem. 2012;121:738–50.CrossRefPubMed Hall JC, Priestley JV, Perry VH, Michael-Titus AT. Docosahexaenoic acid, but not eicosapentaenoic acid, reduces the early inflammatory response following compression spinal cord injury in the rat. J Neurochem. 2012;121:738–50.CrossRefPubMed
35.
go back to reference Kehl LJ, Fairbanks CA, Laughlin TM, Wilcox GL. Neurogenesis in postnatal rat spinal cord: a study in primary culture. Science. 1997;276:586–9.CrossRefPubMed Kehl LJ, Fairbanks CA, Laughlin TM, Wilcox GL. Neurogenesis in postnatal rat spinal cord: a study in primary culture. Science. 1997;276:586–9.CrossRefPubMed
36.
go back to reference Anderson KN, Potter AC, Piccenna LG, Quah AK, Davies KE, Cheema SS. Isolation and culture of motor neurons from the newborn mouse spinal cord. Brain Res Brain Res Protoc. 2004;12:132–6.CrossRefPubMed Anderson KN, Potter AC, Piccenna LG, Quah AK, Davies KE, Cheema SS. Isolation and culture of motor neurons from the newborn mouse spinal cord. Brain Res Brain Res Protoc. 2004;12:132–6.CrossRefPubMed
37.
go back to reference Basso DM, Beattie MS, Bresnahan JC. A sensitive and reliable locomotor rating scale for open field testing in rats. J Neurotrauma. 1995;12:1–21.CrossRefPubMed Basso DM, Beattie MS, Bresnahan JC. A sensitive and reliable locomotor rating scale for open field testing in rats. J Neurotrauma. 1995;12:1–21.CrossRefPubMed
38.
39.
go back to reference D'Osualdo A, Anania VG, Yu K, Lill JR, Kaufman RJ, Matsuzawa S, et al. Transcription factor ATF4 induces NLRP1 inflammasome expression during endoplasmic reticulum stress. PLoS One. 2015;10, e0130635.CrossRefPubMedPubMedCentral D'Osualdo A, Anania VG, Yu K, Lill JR, Kaufman RJ, Matsuzawa S, et al. Transcription factor ATF4 induces NLRP1 inflammasome expression during endoplasmic reticulum stress. PLoS One. 2015;10, e0130635.CrossRefPubMedPubMedCentral
40.
go back to reference Matsuyama D, Watanabe M, Suyama K, Kuroiwa M, Mochida J. Endoplasmic reticulum stress response in the rat contusive spinal cord injury model-susceptibility in specific cell types. Spinal Cord. 2014;52:9–16.CrossRefPubMed Matsuyama D, Watanabe M, Suyama K, Kuroiwa M, Mochida J. Endoplasmic reticulum stress response in the rat contusive spinal cord injury model-susceptibility in specific cell types. Spinal Cord. 2014;52:9–16.CrossRefPubMed
41.
go back to reference Byrnes KR, Stoica BA, Fricke S, Di Giovanni S, Faden AI. Cell cycle activation contributes to post-mitotic cell death and secondary damage after spinal cord injury. Brain. 2007;130:2977–92.CrossRefPubMed Byrnes KR, Stoica BA, Fricke S, Di Giovanni S, Faden AI. Cell cycle activation contributes to post-mitotic cell death and secondary damage after spinal cord injury. Brain. 2007;130:2977–92.CrossRefPubMed
42.
go back to reference Mizuno Y, Mochizuki H, Sugita Y, Goto K. Apoptosis in neurodegenerative disorders. Intern Med. 1998;37:192–3.CrossRefPubMed Mizuno Y, Mochizuki H, Sugita Y, Goto K. Apoptosis in neurodegenerative disorders. Intern Med. 1998;37:192–3.CrossRefPubMed
43.
go back to reference Meng XF, Wang XL, Tian XJ, Yang ZH, Chu GP, Zhang J, et al. Nod-like receptor protein 1 inflammasome mediates neuron injury under high glucose. Mol Neurobiol. 2014;49:673–84.CrossRefPubMed Meng XF, Wang XL, Tian XJ, Yang ZH, Chu GP, Zhang J, et al. Nod-like receptor protein 1 inflammasome mediates neuron injury under high glucose. Mol Neurobiol. 2014;49:673–84.CrossRefPubMed
44.
go back to reference Silverman WR, de Rivero Vaccari JP, Locovei S, Qiu F, Carlsson SK, Scemes E, et al. The pannexin 1 channel activates the inflammasome in neurons and astrocytes. J Biol Chem. 2009;284:18143–51.CrossRefPubMedPubMedCentral Silverman WR, de Rivero Vaccari JP, Locovei S, Qiu F, Carlsson SK, Scemes E, et al. The pannexin 1 channel activates the inflammasome in neurons and astrocytes. J Biol Chem. 2009;284:18143–51.CrossRefPubMedPubMedCentral
45.
go back to reference de Rivero Vaccari JP, Bastien D, Yurcisin G, Pineau I, Dietrich WD, De Koninck Y, et al. P2X4 receptors influence inflammasome activation after spinal cord injury. J Neurosci. 2012;32:3058–66.CrossRefPubMed de Rivero Vaccari JP, Bastien D, Yurcisin G, Pineau I, Dietrich WD, De Koninck Y, et al. P2X4 receptors influence inflammasome activation after spinal cord injury. J Neurosci. 2012;32:3058–66.CrossRefPubMed
46.
go back to reference Zendedel A, Johann S, Mehrabi S, Joghataei MT, Hassanzadeh G, Kipp M, Beyer C. Activation and regulation of NLRP3 inflammasome by intrathecal application of SDF-1a in a spinal cord injury model. Mol Neurobiol. 2015. Epub ahead of print. Zendedel A, Johann S, Mehrabi S, Joghataei MT, Hassanzadeh G, Kipp M, Beyer C. Activation and regulation of NLRP3 inflammasome by intrathecal application of SDF-1a in a spinal cord injury model. Mol Neurobiol. 2015. Epub ahead of print.
49.
50.
go back to reference Gold M, El Khoury J. Beta-amyloid, microglia, and the inflammasome in Alzheimer’s disease. Semin Immunopathol. 2015;37:607–11.CrossRefPubMed Gold M, El Khoury J. Beta-amyloid, microglia, and the inflammasome in Alzheimer’s disease. Semin Immunopathol. 2015;37:607–11.CrossRefPubMed
51.
go back to reference Schnaars M, Beckert H, Halle A. Assessing beta-amyloid-induced NLRP3 inflammasome activation in primary microglia. Methods Mol Biol. 2013;1040:1–8.CrossRefPubMed Schnaars M, Beckert H, Halle A. Assessing beta-amyloid-induced NLRP3 inflammasome activation in primary microglia. Methods Mol Biol. 2013;1040:1–8.CrossRefPubMed
52.
go back to reference Wang X, Arcuino G, Takano T, Lin J, Peng WG, Wan P, et al. P2X7 receptor inhibition improves recovery after spinal cord injury. Nat Med. 2004;10:821–7.CrossRefPubMed Wang X, Arcuino G, Takano T, Lin J, Peng WG, Wan P, et al. P2X7 receptor inhibition improves recovery after spinal cord injury. Nat Med. 2004;10:821–7.CrossRefPubMed
53.
go back to reference Peng W, Cotrina ML, Han X, Yu H, Bekar L, Blum L, et al. Systemic administration of an antagonist of the ATP-sensitive receptor P2X7 improves recovery after spinal cord injury. Proc Natl Acad Sci U S A. 2009;106:12489–93.CrossRefPubMedPubMedCentral Peng W, Cotrina ML, Han X, Yu H, Bekar L, Blum L, et al. Systemic administration of an antagonist of the ATP-sensitive receptor P2X7 improves recovery after spinal cord injury. Proc Natl Acad Sci U S A. 2009;106:12489–93.CrossRefPubMedPubMedCentral
54.
go back to reference Lafon M, Megret F, Lafage M, Prehaud C. The innate immune facet of brain: human neurons express TLR-3 and sense viral dsRNA. J Mol Neurosci. 2006;29:185–94.CrossRefPubMed Lafon M, Megret F, Lafage M, Prehaud C. The innate immune facet of brain: human neurons express TLR-3 and sense viral dsRNA. J Mol Neurosci. 2006;29:185–94.CrossRefPubMed
55.
go back to reference Tang SC, Arumugam TV, Xu X, Cheng A, Mughal MR, Jo DG, et al. Pivotal role for neuronal Toll-like receptors in ischemic brain injury and functional deficits. Proc Natl Acad Sci U S A. 2007;104:13798–803.CrossRefPubMedPubMedCentral Tang SC, Arumugam TV, Xu X, Cheng A, Mughal MR, Jo DG, et al. Pivotal role for neuronal Toll-like receptors in ischemic brain injury and functional deficits. Proc Natl Acad Sci U S A. 2007;104:13798–803.CrossRefPubMedPubMedCentral
56.
go back to reference Peltier DC, Simms A, Farmer JR, Miller DJ. Human neuronal cells possess functional cytoplasmic and TLR-mediated innate immune pathways influenced by phosphatidylinositol-3 kinase signaling. J Immunol. 2010;184:7010–21.CrossRefPubMedPubMedCentral Peltier DC, Simms A, Farmer JR, Miller DJ. Human neuronal cells possess functional cytoplasmic and TLR-mediated innate immune pathways influenced by phosphatidylinositol-3 kinase signaling. J Immunol. 2010;184:7010–21.CrossRefPubMedPubMedCentral
57.
go back to reference Leow-Dyke S, Allen C, Denes A, Nilsson O, Maysami S, Bowie AG, et al. Neuronal Toll-like receptor 4 signaling induces brain endothelial activation and neutrophil transmigration in vitro. J Neuroinflammation. 2012;9:230.CrossRefPubMedPubMedCentral Leow-Dyke S, Allen C, Denes A, Nilsson O, Maysami S, Bowie AG, et al. Neuronal Toll-like receptor 4 signaling induces brain endothelial activation and neutrophil transmigration in vitro. J Neuroinflammation. 2012;9:230.CrossRefPubMedPubMedCentral
58.
go back to reference Yamauchi T, Lin Y, Sharp FR, Noble-Haeusslein LJ. Hemin induces heme oxygenase-1 in spinal cord vasculature and attenuates barrier disruption and neutrophil infiltration in the injured murine spinal cord. J Neurotrauma. 2004;21:1017–30.CrossRefPubMed Yamauchi T, Lin Y, Sharp FR, Noble-Haeusslein LJ. Hemin induces heme oxygenase-1 in spinal cord vasculature and attenuates barrier disruption and neutrophil infiltration in the injured murine spinal cord. J Neurotrauma. 2004;21:1017–30.CrossRefPubMed
59.
go back to reference Dang TN, Robinson SR, Dringen R, Bishop GM. Uptake, metabolism and toxicity of hemin in cultured neurons. Neurochem Int. 2011;58:804–11.CrossRefPubMed Dang TN, Robinson SR, Dringen R, Bishop GM. Uptake, metabolism and toxicity of hemin in cultured neurons. Neurochem Int. 2011;58:804–11.CrossRefPubMed
60.
go back to reference Choi AM, Alam J. Heme oxygenase-1: function, regulation, and implication of a novel stress-inducible protein in oxidant-induced lung injury. Am J Respir Cell Mol Biol. 1996;15:9–19.CrossRefPubMed Choi AM, Alam J. Heme oxygenase-1: function, regulation, and implication of a novel stress-inducible protein in oxidant-induced lung injury. Am J Respir Cell Mol Biol. 1996;15:9–19.CrossRefPubMed
61.
go back to reference Palozza P, Serini S, Curro D, Calviello G, Igarashi K, Mancuso C. beta-Carotene and cigarette smoke condensate regulate heme oxygenase-1 and its repressor factor Bach1: relationship with cell growth. Antioxid Redox Signal. 2006;8:1069–80.CrossRefPubMed Palozza P, Serini S, Curro D, Calviello G, Igarashi K, Mancuso C. beta-Carotene and cigarette smoke condensate regulate heme oxygenase-1 and its repressor factor Bach1: relationship with cell growth. Antioxid Redox Signal. 2006;8:1069–80.CrossRefPubMed
62.
go back to reference Yano Y, Ozono R, Oishi Y, Kambe M, Yoshizumi M, Ishida T, et al. Genetic ablation of the transcription repressor Bach1 leads to myocardial protection against ischemia/reperfusion in mice. Genes Cells. 2006;11:791–803.CrossRefPubMed Yano Y, Ozono R, Oishi Y, Kambe M, Yoshizumi M, Ishida T, et al. Genetic ablation of the transcription repressor Bach1 leads to myocardial protection against ischemia/reperfusion in mice. Genes Cells. 2006;11:791–803.CrossRefPubMed
63.
go back to reference Kanno H, Ozawa H, Dohi Y, Sekiguchi A, Igarashi K, Itoi E. Genetic ablation of transcription repressor Bach1 reduces neural tissue damage and improves locomotor function after spinal cord injury in mice. J Neurotrauma. 2009;26:31–9.CrossRefPubMed Kanno H, Ozawa H, Dohi Y, Sekiguchi A, Igarashi K, Itoi E. Genetic ablation of transcription repressor Bach1 reduces neural tissue damage and improves locomotor function after spinal cord injury in mice. J Neurotrauma. 2009;26:31–9.CrossRefPubMed
64.
go back to reference Perez-de-Puig I, Martin A, Gorina R, de la Rosa X, Martinez E, Planas AM. Induction of hemeoxygenase-1 expression after inhibition of hemeoxygenase activity promotes inflammation and worsens ischemic brain damage in mice. Neuroscience. 2013;243:22–32.CrossRefPubMed Perez-de-Puig I, Martin A, Gorina R, de la Rosa X, Martinez E, Planas AM. Induction of hemeoxygenase-1 expression after inhibition of hemeoxygenase activity promotes inflammation and worsens ischemic brain damage in mice. Neuroscience. 2013;243:22–32.CrossRefPubMed
65.
go back to reference Xiaowei H, Ninghui Z, Wei X, Yiping T, Linfeng X. The experimental study of hypoxia-inducible factor-1alpha and its target genes in spinal cord injury. Spinal Cord. 2006;44:35–43.CrossRefPubMed Xiaowei H, Ninghui Z, Wei X, Yiping T, Linfeng X. The experimental study of hypoxia-inducible factor-1alpha and its target genes in spinal cord injury. Spinal Cord. 2006;44:35–43.CrossRefPubMed
66.
go back to reference Florczyk U, Czauderna S, Stachurska A, Tertil M, Nowak W, Kozakowska M, et al. Opposite effects of HIF-1alpha and HIF-2alpha on the regulation of IL-8 expression in endothelial cells. Free Radic Biol Med. 2011;51:1882–92.CrossRefPubMedPubMedCentral Florczyk U, Czauderna S, Stachurska A, Tertil M, Nowak W, Kozakowska M, et al. Opposite effects of HIF-1alpha and HIF-2alpha on the regulation of IL-8 expression in endothelial cells. Free Radic Biol Med. 2011;51:1882–92.CrossRefPubMedPubMedCentral
67.
go back to reference Loboda A, Stachurska A, Florczyk U, Rudnicka D, Jazwa A, Wegrzyn J, et al. HIF-1 induction attenuates Nrf2-dependent IL-8 expression in human endothelial cells. Antioxid Redox Signal. 2009;11:1501–17.CrossRefPubMed Loboda A, Stachurska A, Florczyk U, Rudnicka D, Jazwa A, Wegrzyn J, et al. HIF-1 induction attenuates Nrf2-dependent IL-8 expression in human endothelial cells. Antioxid Redox Signal. 2009;11:1501–17.CrossRefPubMed
68.
go back to reference Luo YP, Jiang L, Kang K, Fei DS, Meng XL, Nan CC, et al. Hemin inhibits NLRP3 inflammasome activation in sepsis-induced acute lung injury, involving heme oxygenase-1. Int Immunopharmacol. 2014;20:24–32.CrossRefPubMed Luo YP, Jiang L, Kang K, Fei DS, Meng XL, Nan CC, et al. Hemin inhibits NLRP3 inflammasome activation in sepsis-induced acute lung injury, involving heme oxygenase-1. Int Immunopharmacol. 2014;20:24–32.CrossRefPubMed
69.
go back to reference Li H, Zhou X, Zhang J. Induction of heme oxygenase-1 attenuates lipopolysaccharide-induced inflammasome activation in human gingival epithelial cells. Int J Mol Med. 2014;34:1039–44.PubMed Li H, Zhou X, Zhang J. Induction of heme oxygenase-1 attenuates lipopolysaccharide-induced inflammasome activation in human gingival epithelial cells. Int J Mol Med. 2014;34:1039–44.PubMed
70.
go back to reference Navarra P, Dello Russo C, Mancuso C, Preziosi P, Grossman A. Gaseous neuromodulators in the control of neuroendocrine stress axis. Ann N Y Acad Sci. 2000;917:638–46.CrossRefPubMed Navarra P, Dello Russo C, Mancuso C, Preziosi P, Grossman A. Gaseous neuromodulators in the control of neuroendocrine stress axis. Ann N Y Acad Sci. 2000;917:638–46.CrossRefPubMed
71.
go back to reference Barone E, Di Domenico F, Sultana R, Coccia R, Mancuso C, Perluigi M, et al. Heme oxygenase-1 posttranslational modifications in the brain of subjects with Alzheimer disease and mild cognitive impairment. Free Radic Biol Med. 2012;52:2292–301.CrossRefPubMedPubMedCentral Barone E, Di Domenico F, Sultana R, Coccia R, Mancuso C, Perluigi M, et al. Heme oxygenase-1 posttranslational modifications in the brain of subjects with Alzheimer disease and mild cognitive impairment. Free Radic Biol Med. 2012;52:2292–301.CrossRefPubMedPubMedCentral
72.
go back to reference Salinas M, Wang J, Rosa de Sagarra M, Martin D, Rojo AI, Martin-Perez J, et al. Protein kinase Akt/PKB phosphorylates heme oxygenase-1 in vitro and in vivo. FEBS Lett. 2004;578:90–4.CrossRefPubMed Salinas M, Wang J, Rosa de Sagarra M, Martin D, Rojo AI, Martin-Perez J, et al. Protein kinase Akt/PKB phosphorylates heme oxygenase-1 in vitro and in vivo. FEBS Lett. 2004;578:90–4.CrossRefPubMed
73.
go back to reference He CH, Gong P, Hu B, Stewart D, Choi ME, Choi AM, et al. Identification of activating transcription factor 4 (ATF4) as an Nrf2-interacting protein. Implication for heme oxygenase-1 gene regulation. J Biol Chem. 2001;276:20858–65.CrossRefPubMed He CH, Gong P, Hu B, Stewart D, Choi ME, Choi AM, et al. Identification of activating transcription factor 4 (ATF4) as an Nrf2-interacting protein. Implication for heme oxygenase-1 gene regulation. J Biol Chem. 2001;276:20858–65.CrossRefPubMed
74.
go back to reference Dey S, Sayers CM, Verginadis II, Lehman SL, Cheng Y, Cerniglia GJ, et al. ATF4-dependent induction of heme oxygenase 1 prevents anoikis and promotes metastasis. J Clin Invest. 2015;125:2592–608.CrossRefPubMedPubMedCentral Dey S, Sayers CM, Verginadis II, Lehman SL, Cheng Y, Cerniglia GJ, et al. ATF4-dependent induction of heme oxygenase 1 prevents anoikis and promotes metastasis. J Clin Invest. 2015;125:2592–608.CrossRefPubMedPubMedCentral
75.
go back to reference Biswas C, Shah N, Muthu M, La P, Fernando AP, Sengupta S, et al. Nuclear heme oxygenase-1 (HO-1) modulates subcellular distribution and activation of Nrf2, impacting metabolic and anti-oxidant defenses. J Biol Chem. 2014;289:26882–94.CrossRefPubMedPubMedCentral Biswas C, Shah N, Muthu M, La P, Fernando AP, Sengupta S, et al. Nuclear heme oxygenase-1 (HO-1) modulates subcellular distribution and activation of Nrf2, impacting metabolic and anti-oxidant defenses. J Biol Chem. 2014;289:26882–94.CrossRefPubMedPubMedCentral
76.
go back to reference Gandini NA, Fermento ME, Salomon DG, Blasco J, Patel V, Gutkind JS, et al. Nuclear localization of heme oxygenase-1 is associated with tumor progression of head and neck squamous cell carcinomas. Exp Mol Pathol. 2012;93:237–45.CrossRefPubMed Gandini NA, Fermento ME, Salomon DG, Blasco J, Patel V, Gutkind JS, et al. Nuclear localization of heme oxygenase-1 is associated with tumor progression of head and neck squamous cell carcinomas. Exp Mol Pathol. 2012;93:237–45.CrossRefPubMed
77.
go back to reference Tibullo D, Barbagallo I, Giallongo C, La Cava P, Parrinello N, Vanella L, et al. Nuclear translocation of heme oxygenase-1 confers resistance to imatinib in chronic myeloid leukemia cells. Curr Pharm Des. 2013;19:2765–70.CrossRefPubMed Tibullo D, Barbagallo I, Giallongo C, La Cava P, Parrinello N, Vanella L, et al. Nuclear translocation of heme oxygenase-1 confers resistance to imatinib in chronic myeloid leukemia cells. Curr Pharm Des. 2013;19:2765–70.CrossRefPubMed
78.
go back to reference Sagulenko V, Thygesen SJ, Sester DP, Idris A, Cridland JA, Vajjhala PR, et al. AIM2 and NLRP3 inflammasomes activate both apoptotic and pyroptotic death pathways via ASC. Cell Death Differ. 2013;20:1149–60.CrossRefPubMedPubMedCentral Sagulenko V, Thygesen SJ, Sester DP, Idris A, Cridland JA, Vajjhala PR, et al. AIM2 and NLRP3 inflammasomes activate both apoptotic and pyroptotic death pathways via ASC. Cell Death Differ. 2013;20:1149–60.CrossRefPubMedPubMedCentral
79.
go back to reference Chen HG, Xie KL, Han HZ, Wang WN, Liu DQ, Wang GL, et al. Heme oxygenase-1 mediates the anti-inflammatory effect of molecular hydrogen in LPS-stimulated RAW 264.7 macrophages. Int J Surg. 2013;11:1060–6.CrossRefPubMed Chen HG, Xie KL, Han HZ, Wang WN, Liu DQ, Wang GL, et al. Heme oxygenase-1 mediates the anti-inflammatory effect of molecular hydrogen in LPS-stimulated RAW 264.7 macrophages. Int J Surg. 2013;11:1060–6.CrossRefPubMed
80.
go back to reference Lee TS, Chau LY. Heme oxygenase-1 mediates the anti-inflammatory effect of interleukin-10 in mice. Nat Med. 2002;8:240–6.CrossRefPubMed Lee TS, Chau LY. Heme oxygenase-1 mediates the anti-inflammatory effect of interleukin-10 in mice. Nat Med. 2002;8:240–6.CrossRefPubMed
Metadata
Title
Heme oxygenase-1 promotes neuron survival through down-regulation of neuronal NLRP1 expression after spinal cord injury
Authors
Wen-Ping Lin
Gong-Peng Xiong
Qing Lin
Xuan-Wei Chen
Li-Qun Zhang
Jin-Xing Shi
Qing-Feng Ke
Jian-Hua Lin
Publication date
01-12-2016
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2016
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-016-0521-y

Other articles of this Issue 1/2016

Journal of Neuroinflammation 1/2016 Go to the issue