Neuroprotective effects of minocycline and KML29, a potent inhibitor of monoacylglycerol lipase, in an experimental stroke model: a small-animal positron emission tomography study

Tomoteru Yamasaki, Akiko Hatori, Yiding Zhang, Wakana Mori, Yusuke Kurihara, Masanao Ogawa, Hidekatsu Wakizaka, Jian Rong, Lu Wang, Steven Liang, Ming-Rong Zhang, Tomoteru Yamasaki, Akiko Hatori, Yiding Zhang, Wakana Mori, Yusuke Kurihara, Masanao Ogawa, Hidekatsu Wakizaka, Jian Rong, Lu Wang, Steven Liang, Ming-Rong Zhang

Abstract

Hypoxia caused by ischemia induces acidosis and neuroexcitotoxicity, resulting in neuronal death in the central nervous system (CNS). Monoacylglycerol lipase (MAGL) is a modulator of 2-arachidonoylglycerol (2-AG), which is involved in retrograde inhibition of glutamate release in the endocannabinoid system. In the present study, we used positron emission tomography (PET) to monitor MAGL-positive neurons and neuroinflammation in the brains of ischemic rats. Additionally, we performed PET imaging to evaluate the neuroprotective effects of an MAGL inhibitor in an ischemic injury model. Methods: Ischemic-injury rat models were induced by intraluminal right middle cerebral artery occlusion (MCAO). PET studies of the brains of the ischemic rats were performed at several experimental time points (pre-occlusion, days 2, 4, and 7 after the MCAO surgery) using [11C]SAR127303 for MAGL and [18F]FEBMP for 18 kDa translocator protein (TSPO, a hall-mark of neuroinflammation). Medication using minocycline (a well-known neuroprotective agent) or KML29 (a potent MAGL inhibitor) was given immediately after the MCAO surgery and then daily over the subsequent three days. Results: PET imaging of the ischemic rats using [11C]SAR127303 showed an acute decline of radioactive accumulation in the ipsilateral side at two days after MCAO surgery (ratio of the area under the curve between the ipsilateral and contralateral sides: 0.49 ± 0.04 in the cortex and 0.73 ± 0.02 in the striatum). PET imaging with [18F]FEBMP, however, showed a moderate increase in accumulation of radioactivity in the ipsilateral hemisphere on day 2 (1.36 ± 0.11), and further increases on day 4 (1.72 ± 0.15) and day 7 (1.99 ± 0.06). Treatment with minocycline or KML29 eased the decline in radioactive accumulation of [11C]SAR127303 for MAGL (minocycline-treated group: 0.82 ± 0.06 in the cortex and 0.81 ± 0.05 in the striatum; KML29-treated group: 0.72 ± 0.07 in the cortex and 0.88 ± 0.04 in the striatum) and increased uptake of [18F]FEBMP for TSPO (minocycline-treated group: 1.52 ± 0.21 in the cortex and 1.56 ± 0.11 in the striatum; KML29-treated group: 1.63 ± 0.09 in the cortex and 1.50 ± 0.17 in the striatum). In MCAO rats, minocycline treatment showed a neuroprotective effect in the sensorimotor cortex suffering from severe hypoxic injury, whereas KML29 treatment saved neurons in the striatum, including bundles of myelinated axons. Conclusions: PET imaging allowed visualization of the different neuroprotective effects of minocycline and KML29, and indicated that combination pharmacotherapy using these drugs may be an effective therapy in acute ischemia.

Keywords: KML29; MAGL; Minocycline; PET; TSPO.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interest exists.

© The author(s).

Figures

Figure 1
Figure 1
PET imaging for MAGL and TSPO in the brain of MCAO rats. A: Representative 0-60 minute summed PET images of [11C]SAR127303 (upper) and [18F]FEBMP (lower) in the brain pre-occlusion and days 2, 4, and 7 after MCAO surgery. The pseudo color bar represents the level of radioactive accumulation (SUV) in the brain. B-E: TACs of [11C]SAR127303 (B, C) and [18F]FEBMP (D, E) in the contralateral (B, D) and ipsilateral (C, E) cerebral hemisphere (n = 3 for each group). F and G: The ratio of AUC0-60 min in the ipsilateral to contralateral cerebral hemisphere.
Figure 2
Figure 2
Immunofluorescence staining of rat brain sections on day 2 after MCAO surgery. Representative immunofluorescence images in the whole hemisphere (×4 magnification) are shown for MAGL (A; green), NeuN (B; red), and MBP (C; pink). D shows merged images of A-C. All images (A-D) on the top include yellow squares indicating the positions of the four close-ups. E-H: Close-up images (×20) for MAGL, NeuN, MBP, and merged stains in the cortex area of the contralateral hemisphere. I-L: Close-up images of the striatum area in the contralateral hemisphere. M-P: Close-up images of the cortex area in the ipsilateral hemisphere. Q-T: Close-up images of the striatum area in the ipsilateral hemisphere. Scale bar: 1 mm in the top images (A-D) and 50 μm in the middle and bottom images (E-T). Yellow arrows indicate a remarkable decline in MAGL signals overlapping with decreased MBP signals (T).
Figure 3
Figure 3
PET imaging for MAGL in the brains of MCAO rats treated with minocycline or KML-29, or without medication. PET images were averaged between 0 and 60 minutes after injection of [11C]SAR127303 and exhibited as coronal and horizontal views for healthy control, ischemia, minocycline-treated, and KML29-treated rats. The AUC0-60 min values in regions of interests (cortex: B; striatum: C) were calculated from time-activity curves of the contralateral and ipsilateral sides in each rat. The ratio of AUC0-60 min was calculated by dividing the AUC0-60 min value of the ipsilateral side by the AUC0-60 min of the contralateral side in each region of interest (D). Radioactivity in PET images is expressed as SUV. Values are presented are presented as the mean ± SEM. *P < 0.05, **P < 0.01, and ***P < 0.001.
Figure 4
Figure 4
PET imaging for TSPO in the brain of MCAO rats with or without medication treatment. PET images were summed between 0 and 60 minutes after injection of [18F]FEBMP and are shown as coronal and horizontal slices for healthy control, ischemia, minocycline-treated, and KML29-treated rats. The AUC0-60 min values in regions of interests (cortex: B; striatum: C) were calculated from time-activity curves of the contralateral and ipsilateral sides in each rat. The ratio of AUC0-60 min was calculated by dividing the AUC0-60 min value of the ipsilateral side by the AUC0-60 min of the contralateral side in each region of interest (D). Radioactivity in PET images is expressed as SUV. Values are presented are presented as the mean ± SEM. *P < 0.05, **P < 0.01, and ***P < 0.001.
Figure 5
Figure 5
Histological analysis of the neuroprotective effect of medication. A-D: Immunofluorescence staining for MAGL in brain sections of control, ischemia, minocycline-treated, and KML-29-treated rats. E-H: Immunofluorescence staining for TSPO in brain sections of each rat. Cresyl violet I-L: staining for neurons in coronal brain sections of each rat. Yellow indicates neuronal injured areas. Scale bar: 1 mm.
Figure 6
Figure 6
Our proposal for combination pharmacotherapy against ischemia. Ischemia induces two main factors, oxidative stress and neuroexcitotoxicity, which both lead to neuroinflammation. Therefore, neuroprotective effects should be enhanced by preventing both factors by using an anti-oxidative agent and an MAGL inhibitor.

References

    1. Bouzat P, Oddo M. Lactate and the injured brain: friend or foe? Curr Opin Crit Care. 2014;20:133–40.
    1. Hermans E, Challiss RA. Structural, signalling and regulatory properties of the group I metabotropic glutamate receptors: prototypic family C G-protein-coupled receptors. The Biochem J. 2001;359:465–84.
    1. Leng T, Shi Y, Xiong ZG, Sun D. Proton-sensitive cation channels and ion exchangers in ischemic brain injury: new therapeutic targets for stroke? Prog Neurobiol. 2014;115:189–209.
    1. Osuna-Zazuetal MA, Ponce-Gómez JA, Pérez-Neri I. Neuroprotective mechanisms of cannabinoids in brain ischemia and neurodegenerative disorders. Invest Clin. 2015;56:188–200.
    1. Hou L, Rong J, Haider A, Ogasawara D, Varlow C, Schafroth MA. et al. Positron emission tomography imaging of the endocannabinoid system: opportunities and challenges in radiotracer development. J Med Chem. 2021;64:123–49.
    1. Zou S, Kumar U. Cannabinoid receptors and the endocannabinoid system: signaling and function in the central nervous system. Int J Mol Sci. 2018;19:833.
    1. Buczynski MW, Parsons LH. Quantification of brain endocannabinoid levels: methods, interpretations and pitfalls. Br J Pharmacol. 2010;160:423–42.
    1. Pellegrini-Giampietro DE, Mannaioni G, Bagetta G. Post-ischemic brain damage: the endocannabinoid system in the mechanisms of neuronal death. FEBS J. 2009;276:2–12.
    1. Di Marzo V, Stella N, Zimmer A. Endocannabinoid signalling and the deteriorating brain. Nat Rev Neurosci. 2015;16:30–42.
    1. Viader A, Blankman JL, Zhong P, Liu X, Schlosburg JE, Joslyn CM. et al. Metabolic interplay between astrocytes and neurons regulates endocannabinoid action. Cell Rep. 2015;12:798–808.
    1. Carloni S, Alonso-Alconada D, Girelli S, Duranti A, Tontini A, Piomelli D. et al. Pretreatment with the monoacylglycerol lipase inhibitor URB602 protects from the long-term consequences of neonatal hypoxic-ischemic brain injury in rats. Pediatr Res. 2012;72:400–6.
    1. Choi SH, Arai AL, Mou Y, Kang B, Yen CC, Hallenbeck J. et al. Neuroprotective effects of MAGL (monoacylglycerol lipase) inhibitors in experimental ischemic stroke. Stroke. 2018;49:718–26.
    1. Kim JY, Kim N, Yenari MA. Mechanisms and potential therapeutic applications of microglial activation after brain injury. CNS Neurosci Ther. 2015;21:309–19.
    1. Butler CR, Beck EM, Harris A, Huang Z, McAllister LA, Am Ende CW. et al. Azetidine and piperidine carbamates as efficient, covalent inhibitors of monoacylglycerol lipase. J Med Chem. 2017;60:9860–73.
    1. McAllister LA, Butler CR, Mente S, O'Neil SV, Fonseca KR, Piro JR. et al. Discovery of trifluoromethyl glycol carbamates as potent and selective covalent monoacylglycerol lipase (MAGL) inhibitors for treatment of neuroinflammation. J Med Chem. 2018;61:3008–26.
    1. Wang L, Mori W, Cheng R, Yui J, Hatori A, Ma L. et al. Synthesis and preclinical evaluation of sulfonamido-based [11C-carbonyl]-carbamates and ureas for imaging monoacylglycerol lipase. Theranostics. 2016;6:1145–59.
    1. Yamasaki T, Mori W, Zhang Y, Hatori A, Fujinaga M, Wakizaka H. et al. First demonstration of in vivo mapping for regional brain monoacylglycerol lipase using PET with [11C]SAR127303. Neuroimage. 2018;176:313–20.
    1. Tiwari AK, Ji B, Yui J, Fujinaga M, Yamasaki T, Xie L. et al. [18F]FEBMP: Positron emission tomography imaging of TSPO in a model of neuroinflammation in rats, and in vitro autoradiograms of the human brain. Theranostics. 2015;5:961–9.
    1. Papadopoulos V, Baraldi M, Guilarte TR, Knudsen TB, Lacapère JJ, Lindemann P. et al. Translocator protein (18kDa): new nomenclature for the peripheral-type benzodiazepine receptor based on its structure and molecular function. Trends Pharmacol Sci. 2006;27:402–9.
    1. Chen MK, Guilarte TR. Translocator protein 18 kDa (TSPO): molecular sensor of brain injury and repair. Pharmacol Ther. 2008;118:1–17.
    1. Lavisse S, Guillermier M, Hérard AS, Petit F, Delahaye M, Van Camp N. et al. Reactive astrocytes overexpress TSPO and are detected by TSPO positron emission tomography imaging. J Neurosci. 2012;32:10809–18.
    1. Pulsinelli WA, Brierley JB. A new model of bilateral hemispheric ischemia in the unanesthetized rat. Stroke. 1979;10:267–72.
    1. Ji B, Maeda J, Sawada M, Ono M, Okauchi T, Inaji M. et al. Imaging of peripheral benzodiazepine receptor expression as biomarkers of detrimental versus beneficial glial responses in mouse models of Alzheimer's and other CNS pathologies. J Neurosci. 2008;28:12255–67.
    1. Yui J, Xie L, Fujinaga M, Yamasaki T, Hatori A, Kumata K. et al. Monitoring neuroprotective effects using positron emission tomography with [11C]ITMM, a radiotracer for metabotropic glutamate 1 receptor. Stroke. 2013;44:2567–72.
    1. Pulagam KR, Colás L, Padro D, Plaza-García S, Gómez-Vallejo V, Higuchi M. et al. Evaluation of the novel TSPO radiotracer [18F]VUIIS1008 in a preclinical model of cerebral ischemia in rats. EJNMMI Res. 2017;7:93.
    1. Signaling at the synapse. Cell. 1993; 72 Suppl: 1-149.
    1. Wolf HK, Buslei R, Schmidt-Kastner R, Schmidt-Kastner PK, Pietsch T, Wiestler OD. et al. NeuN: a useful neuronal marker for diagnostic histopathology. J Histochem Cytochem. 1996;44:1167–71.
    1. Best L, Ghadery C, Pavese N, Tai YF, Strafella AP. New and Old TSPO PET radioligands for imaging brain microglial activation in neurodegenerative disease. Curr Neurol Neurosci Rep. 2019;19:24.
    1. Zhang H, Song LC, Jia CH, Lu YL. Effects of ATP sensitive potassium channel opener on the mRNA and protein expressions of caspase-12 after cerebral ischemia-reperfusion in rats. Neurosci Bull. 2008;24:7–12.
    1. Meng C, Zhang J, Zhang L, Wang Y, Li Z, Zhao J. Effects of NLRP6 in Cerebral Ischemia/Reperfusion (I/R) Injury in Rats. J Mol Neurosci. 2019;69:411–8.
    1. Wang X, Cheng JL, Ran YC, Zhang Y, Yang L, Lin YN. Expression of RGMb in brain tissue of MCAO rats and its relationship with axonal regeneration. J Neurol Sci. 2017;383:79–86.
    1. Martín A, Boisgard R, Thézé B, Van Camp N, Kuhnast B, Damont A. et al. Evaluation of the PBR/TSPO radioligand [18F]DPA-714 in a rat model of focal cerebral ischemia. J Cereb Blood Flow Metab. 2010;30:230–41.
    1. Boorman E, Zajkwska Z, Ahmed R, Pariante CM, Zunszain PA. Crosstalk between endocannabinoid and immune systems: a potential dysregulation in depression? Psychopharmacology (Berl) 2016;233:1591–604.
    1. Walter L, Dinh T, Stella N. ATP induces a rapid and pronounced increase in 2-arachidonoylglycerol production by astrocytes, a response limited by monoacylglycerol lipase. J Neurosci. 2004;24:8068–74.
    1. Yrjänheikki J, Tikka T, Keinänen R, Goldsteins G, Chan PH, Koistinaho J. A tetracycline derivative, minocycline, reduces inflammation and protects against focal cerebral ischemia with a wide therapeutic window. Proc Natl Acad Sci USA. 1999;96:13496–500.
    1. Kim JY, Kawabori M, Yenari MA. Innate inflammatory responses in stroke: mechanisms and potential therapeutic targets. Curr Med Chem. 2014;21:2076–97.
    1. Matsukawa N, Yasuhara T, Hara K, Xu L, Maki M, Yu G. et al. Therapeutic targets and limits of minocycline neuroprotection in experimental ischemic stroke. BMC Neurosci. 2009;10:126.
    1. Naderi Y, Panahi Y, Barreto GE, Sahebkar A. Neuroprotective effects of minocycline on focal cerebral ischemia injury: a systematic review. Neural Regen Res. 2020;15:773–82.
    1. Ignatowska-Jankowska BM, Ghosh S, Crowe MS, Kinsey SG, Niphakis MJ, Abdullah RA. et al. In vivo characterization of the highly selective monoacylglycerol lipase inhibitor KML29: antinociceptive activity without cannabimimetic side effects. Br J Pharmacol. 2014;171:1392–407.
    1. Gerfen CR. Synaptic organization of the striatum. J Electron Microsc Tech. 1988;10:265–81.
    1. Mengler L, Khmelinskii A, Diedenhofen M, Po C, Staring M, Lelieveldt BP. et al. Brain maturation of the adolescent rat cortex and striatum: changes in volume and myelination. Neuroimage. 2014;84:35–44.
    1. Wu CS, Zhu J, Wager-Miller J, Wang S, O'Leary D, Monory K. et al. Requirement of cannabinoid CB1 receptors in cortical pyramidal neurons for appropriate development of corticothalamic and thalamocortical projections. Eur J Neurosci. 2010;32:693–706.
    1. Matamales M, Bertran-Gonzalez J, Salomon L, Degos B, Deniau JM, Valjent E. et al. Striatal medium-sized spiny neurons: identification by nuclear staining and study of neuronal subpopulations in BAC transgenic mice. PLoS One. 2009;4:e4770.
    1. Uchigashima M, Narushima M, Fukaya M, Katona I, Kano M, Watanabe M. Subcellular arrangement of molecules for 2-arachidonoyl-glycerol-mediated retrograde signaling and its physiological contribution to synaptic modulation in the striatum. J Neurosci. 2007;27:3663–76.
    1. Su SH, Wang YQ, Wu YF, Wang DP, Lin Q, Hai J. Cannabinoid receptor agonist WIN55,212-2 and fatty acid amide hydrolase inhibitor URB597 may protect against cognitive impairment in rats of chronic cerebral hypoperfusion via PI3K/AKT signaling. Behav Brain Res. 2016;313:334–44.
    1. Ding D, Moskowitz SI, Li R, Lee SB, Esteban M, Tomaselli K. et al. Acidosis induces necrosis and apoptosis of cultured hippocampal neurons. Exp Neurol. 2000;162:1–12.
    1. Wu MY, Yiang GT, Liao WT, Tsai AP, Cheng YL, Cheng PW. et al. Current Mechanistic Concepts in Ischemia and Reperfusion Injury. Cell Physiol Biochem. 2018;46:1650–67.

Source: PubMed

3
구독하다