Skip to main content
Top
Published in: Journal of Translational Medicine 1/2018

Open Access 01-12-2018 | Research

The treatment value of IL-1β monoclonal antibody under the targeting location of alpha-methyl-l-tryptophan and superparamagnetic iron oxide nanoparticles in an acute temporal lobe epilepsy model

Authors: Yanli Wang, Yanling Wang, Ran Sun, Xingrao Wu, Xu Chu, Shuhu Zhou, Xibin Hu, Lingyun Gao, Qingxia Kong

Published in: Journal of Translational Medicine | Issue 1/2018

Login to get access

Abstract

Background

Temporal lobe epilepsy (TLE) is a common and often refractory brain disease that is closely correlated with inflammation. Alpha-methyl-l-tryptophan (AMT) is recognized as a surrogate marker for epilepsy, characterized by high uptake in the epileptic focus. There are many advantages of using the magnetic targeting drug delivery system of superparamagnetic iron oxide nanoparticles (SPIONs) to treat many diseases, including epilepsy. We hypothesized that AMT and an IL-1β monoclonal antibody (anti-IL-1β mAb) chelated to SPIONs would utilize the unique advantages of SPIONs and AMT to deliver the anti-IL-1β mAb across the blood–brain barrier (BBB) as a targeted therapy.

Methods

Acute TLE was induced in 30 rats via treatment with lithium-chloride pilocarpine. The effects of plain-SPIONs, anti-IL-1β-mAb-SPIONs, or AMT-anti-IL-1β-mAb-SPIONs on seizure onset were assessed 48 h later. Perl’s iron staining, Nissl staining, immunofluorescence staining and western blotting were performed after magnetic resonance imaging examination.

Results

The imaging and histopathology in combination with the molecular biology findings showed that AMT-anti-IL-1β-mAb-SPIONs were more likely to penetrate the BBB in the acute TLE model to reach the targeting location and deliver a therapeutic effect than plain-SPIONs and anti-IL-1β-mAb-SPIONs.

Conclusions

This study demonstrated the significance of anti-IL-1β-mAb treatment in acute TLE with respect to the unique advantages of SPIONs and the active location-targeting characteristic of AMT.
Literature
1.
go back to reference Espinosa-Jovel CA, Sobrino-Mejía FE. Drug resistant epilepsy. Clinical and neurobiological concept. Revista De Neurologia. 2015;61(4):159.PubMed Espinosa-Jovel CA, Sobrino-Mejía FE. Drug resistant epilepsy. Clinical and neurobiological concept. Revista De Neurologia. 2015;61(4):159.PubMed
2.
go back to reference Fisher RS, et al. ILAE official report: a practical clinical definition of epilepsy. Epilepsia. 2014;55(4):475–82.CrossRefPubMed Fisher RS, et al. ILAE official report: a practical clinical definition of epilepsy. Epilepsia. 2014;55(4):475–82.CrossRefPubMed
4.
go back to reference Vezzani A, Friedman A. Brain inflammation as a biomarker in epilepsy. Biomark. 2011;5(5):607–14. Vezzani A, Friedman A. Brain inflammation as a biomarker in epilepsy. Biomark. 2011;5(5):607–14.
6.
go back to reference Mattia M, et al. Interleukin-1beta biosynthesis inhibition reduces acute seizures and drug resistant chronic epileptic activity in mice. Neurotherapeutic. 2011;8(2):304–15.CrossRef Mattia M, et al. Interleukin-1beta biosynthesis inhibition reduces acute seizures and drug resistant chronic epileptic activity in mice. Neurotherapeutic. 2011;8(2):304–15.CrossRef
7.
go back to reference Juillerrat-Jaannerert L. The targeted delivery of cancer drugs across the blood-brain barrier: chemical modifications of drugs or drug-nanoparticles? Drug Discov Today. 2008;13(23–24):1099–106.CrossRef Juillerrat-Jaannerert L. The targeted delivery of cancer drugs across the blood-brain barrier: chemical modifications of drugs or drug-nanoparticles? Drug Discov Today. 2008;13(23–24):1099–106.CrossRef
8.
go back to reference Corot C, et al. Recent advances in iron oxide nanocrystal technology for medical imaging. Adv Drug Deliv Rev. 2006;58(14):1471–504.CrossRefPubMed Corot C, et al. Recent advances in iron oxide nanocrystal technology for medical imaging. Adv Drug Deliv Rev. 2006;58(14):1471–504.CrossRefPubMed
9.
go back to reference Zhang JL, et al. Core-shell magnetite nanoparticles surface encapsulated with smart stimuli-responsive polymer: synthesis, characterization, and LCST of viable drug-targeting delivery system. Langmuir. 2007;23(11):6342–51.CrossRefPubMed Zhang JL, et al. Core-shell magnetite nanoparticles surface encapsulated with smart stimuli-responsive polymer: synthesis, characterization, and LCST of viable drug-targeting delivery system. Langmuir. 2007;23(11):6342–51.CrossRefPubMed
10.
11.
go back to reference Lukyanov AN, et al. Tumor-targeted liposomes: doxorubicin-loaded long-circulating liposomes modified with anti-cancer antibody. Control Release. 2004;100(1):135–44.CrossRef Lukyanov AN, et al. Tumor-targeted liposomes: doxorubicin-loaded long-circulating liposomes modified with anti-cancer antibody. Control Release. 2004;100(1):135–44.CrossRef
12.
go back to reference Xu Z, et al. In vitro and in vivo evaluation of actively targetable nanoparticles for paclitaxel delivery. Int J Pharm. 2005;288(2):361–8.CrossRefPubMed Xu Z, et al. In vitro and in vivo evaluation of actively targetable nanoparticles for paclitaxel delivery. Int J Pharm. 2005;288(2):361–8.CrossRefPubMed
13.
go back to reference Shen Z, et al. A galactosamine-mediated drug delivery carrier for targeted liver cancer therapy. Pharmacol Res. 2011;64(4):410–9.CrossRefPubMed Shen Z, et al. A galactosamine-mediated drug delivery carrier for targeted liver cancer therapy. Pharmacol Res. 2011;64(4):410–9.CrossRefPubMed
14.
go back to reference Chugani DC, et al. Imaging epileptogenic tubers in children with tuberous sclerosis complex using alpha-[11C]methyl-l-tryptophan positron emission tomography. Ann Neurol. 1998;44(6):858–66.CrossRefPubMed Chugani DC, et al. Imaging epileptogenic tubers in children with tuberous sclerosis complex using alpha-[11C]methyl-l-tryptophan positron emission tomography. Ann Neurol. 1998;44(6):858–66.CrossRefPubMed
15.
go back to reference Akhtari Massoud, Bragin Anatol, Cohen Mark, et al. Functionalized magnetona noparticles for MRI diagnosis and localization in epilepsy. Epilepsia. 2008;49(8):1419–30.CrossRefPubMedPubMedCentral Akhtari Massoud, Bragin Anatol, Cohen Mark, et al. Functionalized magnetona noparticles for MRI diagnosis and localization in epilepsy. Epilepsia. 2008;49(8):1419–30.CrossRefPubMedPubMedCentral
16.
go back to reference Kumar A, et al. α-[11C]- methyl-l-tryptophan PET for tracer localization of epileptogenic brain regions: clinical studies. Biomarkers Med. 2011;5(5):577–84.CrossRef Kumar A, et al. α-[11C]- methyl-l-tryptophan PET for tracer localization of epileptogenic brain regions: clinical studies. Biomarkers Med. 2011;5(5):577–84.CrossRef
17.
go back to reference Rubí S, et al. Positron emission tomography with α-[11C] methyl-l-tryptophan in tuberous sclerosis complex-related epilepsy. Epilepsia. 2013;54(12):2143–50.CrossRefPubMed Rubí S, et al. Positron emission tomography with α-[11C] methyl-l-tryptophan in tuberous sclerosis complex-related epilepsy. Epilepsia. 2013;54(12):2143–50.CrossRefPubMed
18.
go back to reference Ergün EL, et al. SPECT-PET in epilepsy and clinical approach in evaluation. Semin Nucl Med. 2016;46(4):294–307.CrossRefPubMed Ergün EL, et al. SPECT-PET in epilepsy and clinical approach in evaluation. Semin Nucl Med. 2016;46(4):294–307.CrossRefPubMed
19.
go back to reference Kaqawa K, et al. Epilepsy surgery outcome in children with tuberous sclerosis complex evaluated with alpha- [11C] methyl-L-trypotophan position emission tomograpgy (PET). Child Neurol. 2005;20(5):429–38.CrossRef Kaqawa K, et al. Epilepsy surgery outcome in children with tuberous sclerosis complex evaluated with alpha- [11C] methyl-L-trypotophan position emission tomograpgy (PET). Child Neurol. 2005;20(5):429–38.CrossRef
20.
go back to reference Fedi M, et al. Localizing value of alpha-methyl-l-tryptophan PET in intractable epilepsy of neocortical origin. Neurology. 2001;57(9):1629–36.CrossRefPubMed Fedi M, et al. Localizing value of alpha-methyl-l-tryptophan PET in intractable epilepsy of neocortical origin. Neurology. 2001;57(9):1629–36.CrossRefPubMed
21.
go back to reference Juhasz C, et al. Imaging the epileptic brain with positron emission tomography. Neuroimag Clin N Am. 2003;4:705–16.CrossRef Juhasz C, et al. Imaging the epileptic brain with positron emission tomography. Neuroimag Clin N Am. 2003;4:705–16.CrossRef
22.
go back to reference Natsume J, et al. Alpha-[11C] methyl-l-tryptophan and glucose metabolism in patients with temporal lobe epilepsy. Neurology. 2003;60(5):756–61.CrossRefPubMed Natsume J, et al. Alpha-[11C] methyl-l-tryptophan and glucose metabolism in patients with temporal lobe epilepsy. Neurology. 2003;60(5):756–61.CrossRefPubMed
23.
go back to reference Kaqawa K, et al. Epilepsy surgery outcome in children with tuberous sclerosis complex evaluated with alpha- [11C] methyl-l-trypotophan position emission tomograpgy (PET). ChildNeurol. 2005;20(5):429–38. Kaqawa K, et al. Epilepsy surgery outcome in children with tuberous sclerosis complex evaluated with alpha- [11C] methyl-l-trypotophan position emission tomograpgy (PET). ChildNeurol. 2005;20(5):429–38.
24.
go back to reference Akhtari M, et al. Imaging brain neuronal activity using functionalized magnetonanoparticles and MRI. Brain Topogr. 2012;25(4):374–88.CrossRefPubMed Akhtari M, et al. Imaging brain neuronal activity using functionalized magnetonanoparticles and MRI. Brain Topogr. 2012;25(4):374–88.CrossRefPubMed
25.
go back to reference Fu T, Kong Q, et al. Value of functionalized superparamagnetic iron oxide nanoparticles in the diagnosis and treatment of acute temporal lobe epilepsy on MRI. Neural Plast. 2016;2016:2412958.PubMedPubMedCentral Fu T, Kong Q, et al. Value of functionalized superparamagnetic iron oxide nanoparticles in the diagnosis and treatment of acute temporal lobe epilepsy on MRI. Neural Plast. 2016;2016:2412958.PubMedPubMedCentral
26.
go back to reference Johnson MR, et al. Systems genetics identifies Sestrin 3 as a regulator of a proconvulsant gene network in human epileptic hippocampus. Nat Commun. 2015;23(6):6031.CrossRef Johnson MR, et al. Systems genetics identifies Sestrin 3 as a regulator of a proconvulsant gene network in human epileptic hippocampus. Nat Commun. 2015;23(6):6031.CrossRef
27.
go back to reference Vezzani A, et al. ICE/caspase 1 inhibitors and IL-1beta receptor antagonists as potential therapeutics in epilepsy. Curr Opin Investig Drugs. 2010;11(1):43–50.PubMed Vezzani A, et al. ICE/caspase 1 inhibitors and IL-1beta receptor antagonists as potential therapeutics in epilepsy. Curr Opin Investig Drugs. 2010;11(1):43–50.PubMed
28.
go back to reference Maroso M, et al. Interleukin-1beta biosynthesis inhibition reduces acute seizures and drug resistant chronic epileptic activity in mice. Neurotherapeutics. 2011;8(2):304–15.CrossRefPubMedPubMedCentral Maroso M, et al. Interleukin-1beta biosynthesis inhibition reduces acute seizures and drug resistant chronic epileptic activity in mice. Neurotherapeutics. 2011;8(2):304–15.CrossRefPubMedPubMedCentral
29.
go back to reference Akin D, et al. IL-1beta is induced in reactive astrocytes in the somatosensory cortex of rats with genetic absence epilepsy at the onset of spike-and-wave discharges, and contributes to their occurrence. Neurobiol Dis. 2011;44(3):259–69.CrossRefPubMed Akin D, et al. IL-1beta is induced in reactive astrocytes in the somatosensory cortex of rats with genetic absence epilepsy at the onset of spike-and-wave discharges, and contributes to their occurrence. Neurobiol Dis. 2011;44(3):259–69.CrossRefPubMed
30.
go back to reference Jarvela JT, et al. Temporal profiles of age-dependent changes in cytokine mRNA expression and glial cell activation after status epilepticus in postnatal rat hippocampus. J Neuroinflamm. 2011;8(8):29.CrossRef Jarvela JT, et al. Temporal profiles of age-dependent changes in cytokine mRNA expression and glial cell activation after status epilepticus in postnatal rat hippocampus. J Neuroinflamm. 2011;8(8):29.CrossRef
32.
go back to reference Vezzani A, Viviani B. Neuromodulatory properties of inflammatory cytokines and their impact on neuronal excitability. Neuropharmacology. 2015;96(Pt A):70–82.CrossRefPubMed Vezzani A, Viviani B. Neuromodulatory properties of inflammatory cytokines and their impact on neuronal excitability. Neuropharmacology. 2015;96(Pt A):70–82.CrossRefPubMed
34.
go back to reference Pedrazzi M, et al. Potentiation of NMDA receptor-dependent cell responses by extracellular high mobility group box 1 protein. PLoS ONE. 2012;7(8):e44518.CrossRefPubMedPubMedCentral Pedrazzi M, et al. Potentiation of NMDA receptor-dependent cell responses by extracellular high mobility group box 1 protein. PLoS ONE. 2012;7(8):e44518.CrossRefPubMedPubMedCentral
35.
go back to reference Roseti C, et al. GABAA currents are decreased by IL-1β in epileptogenic tissue of patients with temporal lobe epilepsy: implications for ictogenesis. Neurobiol Dis. 2015;82:311–20.CrossRefPubMed Roseti C, et al. GABAA currents are decreased by IL-1β in epileptogenic tissue of patients with temporal lobe epilepsy: implications for ictogenesis. Neurobiol Dis. 2015;82:311–20.CrossRefPubMed
36.
go back to reference Iori V, et al. Receptor for advanced glycation endproducts is upregulated in temporal lobe epilepsy and contributes to experimental seizures. Neurobiol Dis. 2013;58:102–14.CrossRefPubMed Iori V, et al. Receptor for advanced glycation endproducts is upregulated in temporal lobe epilepsy and contributes to experimental seizures. Neurobiol Dis. 2013;58:102–14.CrossRefPubMed
37.
go back to reference Balosso S, et al. Disulfide-containing high mobility group box-1 promotes N-methyl-d-aspartate receptor function and excitotoxicity by activating toll-like receptor 4-dependent signaling in hippocampal neurons. Antioxid Redox Signal. 2014;21:1726–40.CrossRefPubMed Balosso S, et al. Disulfide-containing high mobility group box-1 promotes N-methyl-d-aspartate receptor function and excitotoxicity by activating toll-like receptor 4-dependent signaling in hippocampal neurons. Antioxid Redox Signal. 2014;21:1726–40.CrossRefPubMed
38.
go back to reference Xiao Z, et al. Lobe epilepsy through the PI3K/Akt/mTOR signaling pathway in hippocampal neurons. J Neuroimmunol. 2015;282:110–7.CrossRefPubMed Xiao Z, et al. Lobe epilepsy through the PI3K/Akt/mTOR signaling pathway in hippocampal neurons. J Neuroimmunol. 2015;282:110–7.CrossRefPubMed
39.
go back to reference Dantzer R, O’Connor JC, Lawson MA, et al. Inflammation-associated depression: from serotonin to kynurenine. Psychoneuroendocrinology. 2011;36(3):426–36.CrossRefPubMed Dantzer R, O’Connor JC, Lawson MA, et al. Inflammation-associated depression: from serotonin to kynurenine. Psychoneuroendocrinology. 2011;36(3):426–36.CrossRefPubMed
40.
go back to reference Zunszain PA, Anacker C, Cattaneo A, et al. Interleukin-1β: a new regulator of the kynurenine pathway affecting human hippocampal neurogenesis. Neuropsychopharmacology. 2012;37(4):939.CrossRefPubMed Zunszain PA, Anacker C, Cattaneo A, et al. Interleukin-1β: a new regulator of the kynurenine pathway affecting human hippocampal neurogenesis. Neuropsychopharmacology. 2012;37(4):939.CrossRefPubMed
41.
go back to reference Xie W, et al. Activation of brain indoleamine 2,3-dioxygenase contributes to epilepsy-associated depressive-like behavior in rats with chronic temporal lobe epilepsy. J Neuroinflamm. 2014;11:41.CrossRef Xie W, et al. Activation of brain indoleamine 2,3-dioxygenase contributes to epilepsy-associated depressive-like behavior in rats with chronic temporal lobe epilepsy. J Neuroinflamm. 2014;11:41.CrossRef
42.
go back to reference Colín-González AL, Paz-Loyola AL, Serratos I, et al. Toxic synergism between quinolinic acid and organic acids accumulating in glutaric acidemia type I and in disorders of propionate metabolism in rat brain synaptosomes: relevance for metabolic acidemias. Neuroscience. 2015;308:64–74.CrossRefPubMed Colín-González AL, Paz-Loyola AL, Serratos I, et al. Toxic synergism between quinolinic acid and organic acids accumulating in glutaric acidemia type I and in disorders of propionate metabolism in rat brain synaptosomes: relevance for metabolic acidemias. Neuroscience. 2015;308:64–74.CrossRefPubMed
43.
go back to reference Muller F, Song W, Jang Y, et al. Denervation-induced skeletal muscle atrophy is associated with increased mitochondrial ROS production. Am J Physiol Regul Integr Comp Physiol. 2007;293:R1159–68.CrossRefPubMed Muller F, Song W, Jang Y, et al. Denervation-induced skeletal muscle atrophy is associated with increased mitochondrial ROS production. Am J Physiol Regul Integr Comp Physiol. 2007;293:R1159–68.CrossRefPubMed
44.
go back to reference Santamaría A, Galván-Arzate S, Lisý V, et al. Quinolinic acid induces oxidative stress in rat brain synaptosomes. Neuroreport. 2001;12:871–4.CrossRefPubMed Santamaría A, Galván-Arzate S, Lisý V, et al. Quinolinic acid induces oxidative stress in rat brain synaptosomes. Neuroreport. 2001;12:871–4.CrossRefPubMed
45.
go back to reference Pierozan P, Zamoner A, Soska Â, et al. Acute intrastriatal administration of quinolinic acid provokes hyperphosphorylation of cytoskeletal intermediate filament proteins in astrocytes and neurons of rats. Exp Neurol. 2010;224:188–96.CrossRefPubMed Pierozan P, Zamoner A, Soska Â, et al. Acute intrastriatal administration of quinolinic acid provokes hyperphosphorylation of cytoskeletal intermediate filament proteins in astrocytes and neurons of rats. Exp Neurol. 2010;224:188–96.CrossRefPubMed
46.
47.
go back to reference Zhao M, et al. Magnetic paclitaxel nanoparticles inhibit glioma growth and improve the survival of rats bearing glioma xenografts. Anticancer Res. 2010;30(6):2217–23.PubMed Zhao M, et al. Magnetic paclitaxel nanoparticles inhibit glioma growth and improve the survival of rats bearing glioma xenografts. Anticancer Res. 2010;30(6):2217–23.PubMed
48.
go back to reference Michalak Z, Sano T, Engel T, et al. Spatio-temporally restricted blood brain barrier disruption after intra-amy-gdala kainic acid-induced status epilepticus in mice. Epilepsy Res. 2013;103(2/3):167–79.CrossRefPubMed Michalak Z, Sano T, Engel T, et al. Spatio-temporally restricted blood brain barrier disruption after intra-amy-gdala kainic acid-induced status epilepticus in mice. Epilepsy Res. 2013;103(2/3):167–79.CrossRefPubMed
49.
50.
go back to reference Jun YW, et al. Chemical design of nanoparticle probea for high-peformance magnetic resonance imaging. Angew Chem Int Ed. 2008;47(28):5122–35.CrossRef Jun YW, et al. Chemical design of nanoparticle probea for high-peformance magnetic resonance imaging. Angew Chem Int Ed. 2008;47(28):5122–35.CrossRef
51.
go back to reference Weinstein JS, et al. Superparamagnetic iron oxide nanop articles: diagnostic magnetic resonance imaging and potential therapeutic applications in neurooncology and central nervous system inflammatory pathologies, a review. J Cereb Blood Flow Metab. 2010;30(1):15–35.CrossRefPubMed Weinstein JS, et al. Superparamagnetic iron oxide nanop articles: diagnostic magnetic resonance imaging and potential therapeutic applications in neurooncology and central nervous system inflammatory pathologies, a review. J Cereb Blood Flow Metab. 2010;30(1):15–35.CrossRefPubMed
52.
go back to reference Islam T, Harisinghani MG. Overview of nanoparticle use in cancer imaging. Cancer Biomark. 2009;5(2):61–7.CrossRefPubMed Islam T, Harisinghani MG. Overview of nanoparticle use in cancer imaging. Cancer Biomark. 2009;5(2):61–7.CrossRefPubMed
53.
go back to reference Ndong C, Toraya-Brown S, Kekalo K, et al. Antibody-mediated targeting of iron oxide nanoparticles to the folate receptor alpha increases tumor cell association in vitro and in vivo. Int J Nanomed. 2015;1(10):2595–617. Ndong C, Toraya-Brown S, Kekalo K, et al. Antibody-mediated targeting of iron oxide nanoparticles to the folate receptor alpha increases tumor cell association in vitro and in vivo. Int J Nanomed. 2015;1(10):2595–617.
54.
55.
go back to reference Chugani DC, Heyes MP, Kuhn DM, Chugani HT. Evidence that α[11C] methyl-l-tryptophan PET traces tryptophan metabolism via the kynurenine pathway in tuberous sclerosis complex. Soc Neurosci Abstracts. 1998;24:1757. Chugani DC, Heyes MP, Kuhn DM, Chugani HT. Evidence that α[11C] methyl-l-tryptophan PET traces tryptophan metabolism via the kynurenine pathway in tuberous sclerosis complex. Soc Neurosci Abstracts. 1998;24:1757.
56.
go back to reference Chuqani DC, et al. Alpha[C-ll]methyl-l-tryptophan PET maps brain serotonin synthesis and kynurenine pathway metabolism. Cereb Blood Flow Metab. 2000;20(1):2–9.CrossRef Chuqani DC, et al. Alpha[C-ll]methyl-l-tryptophan PET maps brain serotonin synthesis and kynurenine pathway metabolism. Cereb Blood Flow Metab. 2000;20(1):2–9.CrossRef
58.
go back to reference Gan N, et al. Myoloid-related protein 8, an endogenous ligand of Toll-like receptor 4, is 2 involved in epileptogenesis of mesial temporal lobe epilepsy via activation of the nuclear factor-kappaB pathway in astrocytes. Mol Neurobiol. 2014;49:337–51.CrossRefPubMed Gan N, et al. Myoloid-related protein 8, an endogenous ligand of Toll-like receptor 4, is 2 involved in epileptogenesis of mesial temporal lobe epilepsy via activation of the nuclear factor-kappaB pathway in astrocytes. Mol Neurobiol. 2014;49:337–51.CrossRefPubMed
61.
go back to reference Deprez F, et al. Adoptive transfer of T lymphocytes in immunodeficient mice influences epileptogenesis and neurodegeneration in a model of temporal lobe epilepsy. Neurobiol Dis. 2011;44(2):174–84.CrossRefPubMed Deprez F, et al. Adoptive transfer of T lymphocytes in immunodeficient mice influences epileptogenesis and neurodegeneration in a model of temporal lobe epilepsy. Neurobiol Dis. 2011;44(2):174–84.CrossRefPubMed
62.
go back to reference Pasti L, et al. Intracellular calcium oscillations in astrocytes: a highly plastic, bidirectional form of communication between neurons and astrocytes in situ. J Neurosci. 1997;17(20):7817–30.CrossRefPubMed Pasti L, et al. Intracellular calcium oscillations in astrocytes: a highly plastic, bidirectional form of communication between neurons and astrocytes in situ. J Neurosci. 1997;17(20):7817–30.CrossRefPubMed
63.
go back to reference Fellin T, et al. Neuronal synchrony mediated by astrocytic glutamate through activation of extrasynaptic NMDA receptors. Neuron. 2004;43(5):729–43.CrossRefPubMed Fellin T, et al. Neuronal synchrony mediated by astrocytic glutamate through activation of extrasynaptic NMDA receptors. Neuron. 2004;43(5):729–43.CrossRefPubMed
64.
go back to reference Losi G, et al. A new experimental model of focal seizures in the entorhinal cortex. Epilepsia. 2010;51(8):1493–502.CrossRefPubMed Losi G, et al. A new experimental model of focal seizures in the entorhinal cortex. Epilepsia. 2010;51(8):1493–502.CrossRefPubMed
65.
66.
go back to reference Nedergaard M. Direct signaling from astrocytes to neurons in cultures of mammalian brain cells. Science. 1994;263(5154):1768–71.CrossRefPubMed Nedergaard M. Direct signaling from astrocytes to neurons in cultures of mammalian brain cells. Science. 1994;263(5154):1768–71.CrossRefPubMed
67.
68.
go back to reference Block ML, et al. Microglia-mediated neurotoxicity: uncovering the molecular mechanisms. Nat Rev Neurosci. 2007;8(1):57–69.CrossRefPubMed Block ML, et al. Microglia-mediated neurotoxicity: uncovering the molecular mechanisms. Nat Rev Neurosci. 2007;8(1):57–69.CrossRefPubMed
69.
go back to reference Salter MW, Beggs S. Sublime microglia: expanding roles for the guardians of the CNS. Cell. 2014;158(1):15–24.CrossRefPubMed Salter MW, Beggs S. Sublime microglia: expanding roles for the guardians of the CNS. Cell. 2014;158(1):15–24.CrossRefPubMed
Metadata
Title
The treatment value of IL-1β monoclonal antibody under the targeting location of alpha-methyl-l-tryptophan and superparamagnetic iron oxide nanoparticles in an acute temporal lobe epilepsy model
Authors
Yanli Wang
Yanling Wang
Ran Sun
Xingrao Wu
Xu Chu
Shuhu Zhou
Xibin Hu
Lingyun Gao
Qingxia Kong
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Journal of Translational Medicine / Issue 1/2018
Electronic ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-018-1712-3

Other articles of this Issue 1/2018

Journal of Translational Medicine 1/2018 Go to the issue