Minocycline fails to improve neurologic and histologic outcome after ventricular fibrillation cardiac arrest in rats

Andreas Janata, Ingrid Am Magnet, Kristin L Schreiber, Caleb D Wilson, Jason P Stezoski, Keri Janesko-Feldman, Patrick M Kochanek, Tomas Drabek, Andreas Janata, Ingrid Am Magnet, Kristin L Schreiber, Caleb D Wilson, Jason P Stezoski, Keri Janesko-Feldman, Patrick M Kochanek, Tomas Drabek

Abstract

Background: Prolonged cardiac arrest (CA) produces extensive neuronal death and microglial proliferation and activation resulting in neuro-cognitive disabilities. Among other potential mechanisms, microglia have been implicated as triggers of neuronal death after hypoxic-ischemic insults. Minocycline is neuroprotective in some brain ischemia models, either by blunting the microglial response or by a direct effect on neurons.

Aim: To improve survival, attenuate neurologic deficits, neuroinflammation, and histological damage after ventricular fibrillation (VF) CA in rats.

Methods: Adult male isoflurane-anesthetized rats were subjected to 6 min VF CA followed by 2 min resuscitation including chest compression, epinephrine, bicarbonate, and defibrillation. After return of spontaneous circulation (ROSC), rats were randomized to two groups: (1) Minocycline 90 mg/kg intraperitoneally (i.p.) at 15 min ROSC, followed by 22.5 mg/kg i.p. every 12 h for 72 h; and (2) Controls, receiving the same volume of vehicle (phosphate-buffered saline). The rats were kept normothermic during the postoperative course. Neurologic injury was assessed daily using Overall Performance Category (OPC; 1 = normal, 5 = dead) and Neurologic Deficit Score (NDS; 0% = normal, 100% = dead). Rats were sacrificed at 72 h. Neuronal degeneration (Fluoro-Jade C staining) and microglia proliferation (anti-Iba-1 staining) were quantified in four selectively vulnerable brain regions (hippocampus, striatum, cerebellum, cortex) by three independent reviewers masked to the group assignment.

Results: In the minocycline group, 8 out of 14 rats survived to 72 h compared to 8 out of 19 rats in the control group (P = 0.46). The degree of neurologic deficit at 72 h [median, (interquartile range)] was not different between survivors in minocycline vs controls: OPC 1.5 (1-2.75) vs 2 (1.25-3), P = 0.442; NDS 12 (2-20) vs 17 (7-51), P = 0.328) or between all studied rats. The number of degenerating neurons (minocycline vs controls, mean ± SEM: Hippocampus 58 ± 8 vs 76 ± 8; striatum 121 ± 43 vs 153 ± 32; cerebellum 20 ± 7 vs 22 ± 8; cortex 0 ± 0 vs 0 ± 0) or proliferating microglia (hippocampus 157 ± 15 vs 193 cortex 0 ± 0 vs 0 ± 0; 16; striatum 150 ± 22 vs 161 ± 23; cerebellum 20 ± 7 vs 22 ± 8; cortex 26 ± 6 vs 31 ± 7) was not different between groups in any region (all P > 0.05). Numerically, there were approximately 20% less degenerating neurons and proliferating microglia in the hippocampus and striatum in the minocycline group, with a consistent pattern of histological damage across the individual regions of interest.

Conclusion: Minocycline did not improve survival and failed to confer substantial benefits on neurologic function, neuronal loss or microglial proliferation across multiple brain regions in our model of rat VF CA.

Keywords: Cardiopulmonary resuscitation; Heart arrest/pathology; Microglia/drug effects; Minocycline/pharmacology; Neurons/drug effects; Survival rate.

©The Author(s) 2019. Published by Baishideng Publishing Group Inc. All rights reserved.

Figures

Figure 1
Figure 1
A flow-chart of the study. Please refer to the text for details on randomization protocol. ROSC: Return of spontaneous circulation.
Figure 2
Figure 2
Overall performance categories after cardiac arrest. Each dot represents one rat. No difference between groups. OPC: Overall performance category.
Figure 3
Figure 3
Neurologic deficit score at 72 h in all rats studied (left panel) and in survivors only (right panel). Boxes represent interquartile ranges. The line across each box indicates the median, and the whiskers are the highest and lowest values. No differences between groups. C: Control group; M: Minocycline group; NDS: Neurologic deficit score.
Figure 4
Figure 4
Kaplan-Maier survival plot. No differences between groups.
Figure 5
Figure 5
Regional neuronal degeneration (top) and microglial proliferation (bottom) after cardiac arrest. Regional neuronal loss and microglial proliferation after cardiac arrest in controls and rats treated with minocycline were not different between groups in any region at 72 h. HIP: Hippocampus; STRI: Striatum; CEREB: Cerebellum; CTX: Cortex. Mean ± SEM values are displayed.
Figure 6
Figure 6
Representative samples of neuronal degeneration after 6 min ventricular fibrillation cardiac arrest at 72 h. Blue staining is 4',6-diamidino-2-phenylindole, visualizing neurons, and green staining is Fluoro-Jade C, visualizing degenerating neurons. Hippocampal neuronal loss is visible in the cardiac arrest 1 sector and in hilar region of the dentate gyrus. The inset shows the mid-section of cardiac arrest 1 in closer detail. Marked neuronal degeneration of the medium spiny neurons is seen in the striatum. Selectively vulnerable neuronal loss of Purkinje neurons visualized in the cerebellum. No neurodegeneration is observed in the cortex. Magnification × 10 except the panoramic view of the hippocampus, magnification × 4. The scale bars in the far left and far right lower panels represent 10 µm.
Figure 7
Figure 7
Representative samples of microglial activation and proliferation after 6 min ventricular fibrillation cardiac arrest at 72 h. Sections are stained with hematoxylin. Brown staining is anti-Iba-1 staining, visualizing microglia, counterstained with diaminobenzamide. Magnification × 10. The scale bars in the far left and far right lower panels represent 10 µm.

References

    1. Janata A, Drabek T, Magnet IA, Stezoski JP, Janesko-Feldman K, Popp E, Garman RH, Tisherman SA, Kochanek PM. Extracorporeal versus conventional cardiopulmonary resuscitation after ventricular fibrillation cardiac arrest in rats: a feasibility trial. Crit Care Med. 2013;41:e211–e222.
    1. Drabek T, Tisherman SA, Beuke L, Stezoski J, Janesko-Feldman K, Lahoud-Rahme M, Kochanek PM. Deep hypothermia attenuates microglial proliferation independent of neuronal death after prolonged cardiac arrest in rats. Anesth Analg. 2009;109:914–923.
    1. Uray T, Lamade A, Elmer J, Drabek T, Stezoski JP, Missé A, Janesko-Feldman K, Garman RH, Chen N, Kochanek PM, Dezfulian C, Callaway CW, Doshi AA, Frisch A, Guyette FX, Reynolds JC, Rittenberger JC University of Pittsburgh Post-Cardiac Arrest Service. Phenotyping Cardiac Arrest: Bench and Bedside Characterization of Brain and Heart Injury Based on Etiology. Crit Care Med. 2018;46:e508–e515.
    1. Neigh GN, Karelina K, Glasper ER, Bowers SL, Zhang N, Popovich PG, DeVries AC. Anxiety after cardiac arrest/cardiopulmonary resuscitation: exacerbated by stress and prevented by minocycline. Stroke. 2009;40:3601–3607.
    1. Keilhoff G, Schweizer H, John R, Langnaese K, Ebmeyer U. Minocycline neuroprotection in a rat model of asphyxial cardiac arrest is limited. Resuscitation. 2011;82:341–349.
    1. Wang W, Lu R, Feng DY, Liang LR, Liu B, Zhang H. Inhibition of microglial activation contributes to propofol-induced protection against post-cardiac arrest brain injury in rats. J Neurochem. 2015;134:892–903.
    1. Tang M, Alexander H, Clark RS, Kochanek PM, Kagan VE, Bayir H. Minocycline reduces neuronal death and attenuates microglial response after pediatric asphyxial cardiac arrest. J Cereb Blood Flow Metab. 2010;30:119–129.
    1. Wang QY, Sun P, Zhang Q, Yao SL. Minocycline attenuates microglial response and reduces neuronal death after cardiac arrest and cardiopulmonary resuscitation in mice. J Huazhong Univ Sci Technolog Med Sci. 2015;35:225–229.
    1. Hewlett KA, Corbett D. Delayed minocycline treatment reduces long-term functional deficits and histological injury in a rodent model of focal ischemia. Neuroscience. 2006;141:27–33.
    1. Drabek T, Janata A, Wilson CD, Stezoski J, Janesko-Feldman K, Tisherman SA, Foley LM, Verrier JD, Kochanek PM. Minocycline attenuates brain tissue levels of TNF-α produced by neurons after prolonged hypothermic cardiac arrest in rats. Resuscitation. 2014;85:284–291.
    1. Carrillo P, Takasu A, Safar P, Tisherman S, Stezoski SW, Stolz G, Dixon CE, Radovsky A. Prolonged severe hemorrhagic shock and resuscitation in rats does not cause subtle brain damage. J Trauma. 1998;45:239–248; discussion 248-249.
    1. Neumar RW, Bircher NG, Sim KM, Xiao F, Zadach KS, Radovsky A, Katz L, Ebmeyer E, Safar P. Epinephrine and sodium bicarbonate during CPR following asphyxial cardiac arrest in rats. Resuscitation. 1995;29:249–263.
    1. Cho KO, Kim SK, Cho YJ, Sung KW, Kim SY. Regional differences in the neuroprotective effect of minocycline in a mouse model of global forebrain ischemia. Life Sci. 2007;80:2030–2035.
    1. Schmued LC, Hopkins KJ. Fluoro-Jade B: a high affinity fluorescent marker for the localization of neuronal degeneration. Brain Res. 2000;874:123–130.
    1. Ito D, Imai Y, Ohsawa K, Nakajima K, Fukuuchi Y, Kohsaka S. Microglia-specific localisation of a novel calcium binding protein, Iba1. Brain Res Mol Brain Res. 1998;57:1–9.
    1. Koshinaga M, Suma T, Fukushima M, Tsuboi I, Aizawa S, Katayama Y. Rapid microglial activation induced by traumatic brain injury is independent of blood brain barrier disruption. Histol Histopathol. 2007;22:129–135.
    1. Naderi Y, Sabetkasaei M, Parvardeh S, Moini Zanjani T. Neuroprotective effects of pretreatment with minocycline on memory impairment following cerebral ischemia in rats. Behav Pharmacol. 2017;28:214–222.
    1. Naderi Y, Sabetkasaei M, Parvardeh S, Zanjani TM. Neuroprotective effect of minocycline on cognitive impairments induced by transient cerebral ischemia/reperfusion through its anti-inflammatory and anti-oxidant properties in male rat. Brain Res Bull. 2017;131:207–213.
    1. Gehrmann J, Banati RB, Wiessner C, Hossmann KA, Kreutzberg GW. Reactive microglia in cerebral ischaemia: an early mediator of tissue damage? Neuropathol Appl Neurobiol. 1995;21:277–289.
    1. Barakat R, Redzic Z. The Role of Activated Microglia and Resident Macrophages in the Neurovascular Unit during Cerebral Ischemia: Is the Jury Still Out? Med Princ Pract. 2016;25 Suppl 1:3–14.
    1. Fan LW, Lin S, Pang Y, Rhodes PG, Cai Z. Minocycline attenuates hypoxia-ischemia-induced neurological dysfunction and brain injury in the juvenile rat. Eur J Neurosci. 2006;24:341–350.
    1. Yrjänheikki J, Keinänen R, Pellikka M, Hökfelt T, Koistinaho J. Tetracyclines inhibit microglial activation and are neuroprotective in global brain ischemia. Proc Natl Acad Sci U S A. 1998;95:15769–15774.
    1. Arvin KL, Han BH, Du Y, Lin SZ, Paul SM, Holtzman DM. Minocycline markedly protects the neonatal brain against hypoxic-ischemic injury. Ann Neurol. 2002;52:54–61.
    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 U S A. 1999;96:13496–13500.
    1. Liu Z, Fan Y, Won SJ, Neumann M, Hu D, Zhou L, Weinstein PR, Liu J. Chronic treatment with minocycline preserves adult new neurons and reduces functional impairment after focal cerebral ischemia. Stroke. 2007;38:146–152.
    1. Wang CX, Yang T, Shuaib A. Effects of minocycline alone and in combination with mild hypothermia in embolic stroke. Brain Res. 2003;963:327–329.
    1. Wang CX, Yang T, Noor R, Shuaib A. Delayed minocycline but not delayed mild hypothermia protects against embolic stroke. BMC Neurol. 2002;2:2.
    1. Sanchez Mejia RO, Ona VO, Li M, Friedlander RM. Minocycline reduces traumatic brain injury-mediated caspase-1 activation, tissue damage, and neurological dysfunction. Neurosurgery. 2001;48:1393–9; discussion 1399-401.
    1. Bye N, Habgood MD, Callaway JK, Malakooti N, Potter A, Kossmann T, Morganti-Kossmann MC. Transient neuroprotection by minocycline following traumatic brain injury is associated with attenuated microglial activation but no changes in cell apoptosis or neutrophil infiltration. Exp Neurol. 2007;204:220–233.
    1. Stirling DP, Khodarahmi K, Liu J, McPhail LT, McBride CB, Steeves JD, Ramer MS, Tetzlaff W. Minocycline treatment reduces delayed oligodendrocyte death, attenuates axonal dieback, and improves functional outcome after spinal cord injury. J Neurosci. 2004;24:2182–2190.
    1. Festoff BW, Ameenuddin S, Arnold PM, Wong A, Santacruz KS, Citron BA. Minocycline neuroprotects, reduces microgliosis, and inhibits caspase protease expression early after spinal cord injury. J Neurochem. 2006;97:1314–1326.
    1. Power C, Henry S, Del Bigio MR, Larsen PH, Corbett D, Imai Y, Yong VW, Peeling J. Intracerebral hemorrhage induces macrophage activation and matrix metalloproteinases. Ann Neurol. 2003;53:731–742.
    1. Lampl Y, Boaz M, Gilad R, Lorberboym M, Dabby R, Rapoport A, Anca-Hershkowitz M, Sadeh M. Minocycline treatment in acute stroke: an open-label, evaluator-blinded study. Neurology. 2007;69:1404–1410.
    1. Saivin S, Houin G. Clinical pharmacokinetics of doxycycline and minocycline. Clin Pharmacokinet. 1988;15:355–366.
    1. Yenari MA, Xu L, Tang XN, Qiao Y, Giffard RG. Microglia potentiate damage to blood-brain barrier constituents: improvement by minocycline in vivo and in vitro. Stroke. 2006;37:1087–1093.
    1. Golub LM, Ramamurthy NS, McNamara TF, Greenwald RA, Rifkin BR. Tetracyclines inhibit connective tissue breakdown: new therapeutic implications for an old family of drugs. Crit Rev Oral Biol Med. 1991;2:297–321.
    1. Morimoto N, Shimazawa M, Yamashima T, Nagai H, Hara H. Minocycline inhibits oxidative stress and decreases in vitro and in vivo ischemic neuronal damage. Brain Res. 2005;1044:8–15.
    1. Lin S, Wei X, Xu Y, Yan C, Dodel R, Zhang Y, Liu J, Klaunig JE, Farlow M, Du Y. Minocycline blocks 6-hydroxydopamine-induced neurotoxicity and free radical production in rat cerebellar granule neurons. Life Sci. 2003;72:1635–1641.
    1. Du Y, Ma Z, Lin S, Dodel RC, Gao F, Bales KR, Triarhou LC, Chernet E, Perry KW, Nelson DL, Luecke S, Phebus LA, Bymaster FP, Paul SM. Minocycline prevents nigrostriatal dopaminergic neurodegeneration in the MPTP model of Parkinson's disease. Proc Natl Acad Sci USA. 2001;98:14669–14674.
    1. Lin S, Zhang Y, Dodel R, Farlow MR, Paul SM, Du Y. Minocycline blocks nitric oxide-induced neurotoxicity by inhibition p38 MAP kinase in rat cerebellar granule neurons. Neurosci Lett. 2001;315:61–64.
    1. Tomás-Camardiel M, Rite I, Herrera AJ, de Pablos RM, Cano J, Machado A, Venero JL. Minocycline reduces the lipopolysaccharide-induced inflammatory reaction, peroxynitrite-mediated nitration of proteins, disruption of the blood-brain barrier, and damage in the nigral dopaminergic system. Neurobiol Dis. 2004;16:190–201.
    1. Ryu JK, McLarnon JG. Minocycline or iNOS inhibition block 3-nitrotyrosine increases and blood-brain barrier leakiness in amyloid beta-peptide-injected rat hippocampus. Exp Neurol. 2006;198:552–557.
    1. Attur MG, Patel RN, Patel PD, Abramson SB, Amin AR. Tetracycline up-regulates COX-2 expression and prostaglandin E2 production independent of its effect on nitric oxide. J Immunol. 1999;162:3160–3167.
    1. Chen M, Ona VO, Li M, Ferrante RJ, Fink KB, Zhu S, Bian J, Guo L, Farrell LA, Hersch SM, Hobbs W, Vonsattel JP, Cha JH, Friedlander RM. Minocycline inhibits caspase-1 and caspase-3 expression and delays mortality in a transgenic mouse model of Huntington disease. Nat Med. 2000;6:797–801.
    1. Teng YD, Choi H, Onario RC, Zhu S, Desilets FC, Lan S, Woodard EJ, Snyder EY, Eichler ME, Friedlander RM. Minocycline inhibits contusion-triggered mitochondrial cytochrome c release and mitigates functional deficits after spinal cord injury. Proc Natl Acad Sci U S A. 2004;101:3071–3076.
    1. Kelly KJ, Sutton TA, Weathered N, Ray N, Caldwell EJ, Plotkin Z, Dagher PC. Minocycline inhibits apoptosis and inflammation in a rat model of ischemic renal injury. Am J Physiol Renal Physiol. 2004;287:F760–F766.
    1. Wang X, Zhu S, Drozda M, Zhang W, Stavrovskaya IG, Cattaneo E, Ferrante RJ, Kristal BS, Friedlander RM. Minocycline inhibits caspase-independent and -dependent mitochondrial cell death pathways in models of Huntington's disease. Proc Natl Acad Sci U S A. 2003;100:10483–10487.
    1. Sato Y, Laskowitz DT, Bennett ER, Newman MF, Warner DS, Grocott HP. Differential cerebral gene expression during cardiopulmonary bypass in the rat: evidence for apoptosis? Anesth Analg. 2002;94:1389–1394, table of contents.
    1. Hindman BJ, Moore SA, Cutkomp J, Smith T, Ross-Barta SE, Dexter F, Brian JE., Jr Brain expression of inducible cyclooxygenase 2 messenger RNA in rats undergoing cardiopulmonary bypass. Anesthesiology. 2001;95:1380–1388.
    1. Zhang TJ, Hang J, Wen DX, Hang YN, Sieber FE. Hippocampus bcl-2 and bax expression and neuronal apoptosis after moderate hypothermic cardiopulmonary bypass in rats. Anesth Analg. 2006;102:1018–1025.
    1. Fan LW, Pang Y, Lin S, Rhodes PG, Cai Z. Minocycline attenuates lipopolysaccharide-induced white matter injury in the neonatal rat brain. Neuroscience. 2005;133:159–168.
    1. Fox C, Dingman A, Derugin N, Wendland MF, Manabat C, Ji S, Ferriero DM, Vexler ZS. Minocycline confers early but transient protection in the immature brain following focal cerebral ischemia-reperfusion. J Cereb Blood Flow Metab. 2005;25:1138–1149.
    1. Sriram K, Miller DB, O'Callaghan JP. Minocycline attenuates microglial activation but fails to mitigate striatal dopaminergic neurotoxicity: role of tumor necrosis factor-alpha. J Neurochem. 2006;96:706–718.
    1. Cornet S, Spinnewyn B, Delaflotte S, Charnet C, Roubert V, Favre C, Hider H, Chabrier PE, Auguet M. Lack of evidence of direct mitochondrial involvement in the neuroprotective effect of minocycline. Eur J Pharmacol. 2004;505:111–119.
    1. Elewa HF, Hilali H, Hess DC, Machado LS, Fagan SC. Minocycline for short-term neuroprotection. Pharmacotherapy. 2006;26:515–521.
    1. Carty ML, Wixey JA, Colditz PB, Buller KM. Post-insult minocycline treatment attenuates hypoxia-ischemia-induced neuroinflammation and white matter injury in the neonatal rat: a comparison of two different dose regimens. Int J Dev Neurosci. 2008;26:477–485.
    1. Buller KM, Carty ML, Reinebrant HE, Wixey JA. Minocycline: a neuroprotective agent for hypoxic-ischemic brain injury in the neonate? J Neurosci Res. 2009;87:599–608.
    1. Cikla U, Chanana V, Kintner DB, Covert L, Dewall T, Waldman A, Rowley P, Cengiz P, Ferrazzano P. Suppression of microglia activation after hypoxia-ischemia results in age-dependent improvements in neurologic injury. J Neuroimmunol. 2016;291:18–27.
    1. Tsuji M, Wilson MA, Lange MS, Johnston MV. Minocycline worsens hypoxic-ischemic brain injury in a neonatal mouse model. Exp Neurol. 2004;189:58–65.
    1. Yu IC, Kuo PC, Yen JH, Paraiso HC, Curfman ET, Hong-Goka BC, Sweazey RD, Chang FL. A Combination of Three Repurposed Drugs Administered at Reperfusion as a Promising Therapy for Postischemic Brain Injury. Transl Stroke Res. 2017;8:560–577.
    1. Drabek T, Wilson CD, Janata A, Stezoski JP, Janesko-Feldman K, Garman RH, Tisherman SA, Kochanek PM. Unique brain region-dependent cytokine signatures after prolonged hypothermic cardiac arrest in rats. Ther Hypothermia Temp Manag. 2015;5:26–39.
    1. Janata A, Magnet IA, Uray T, Stezoski JP, Janesko-Feldman K, Tisherman SA, Kochanek PM, Drabek T. Regional TNFα mapping in the brain reveals the striatum as a neuroinflammatory target after ventricular fibrillation cardiac arrest in rats. Resuscitation. 2014;85:694–701.
    1. Liu T, Clark RK, McDonnell PC, Young PR, White RF, Barone FC, Feuerstein GZ. Tumor necrosis factor-alpha expression in ischemic neurons. Stroke. 1994;25:1481–1488.
    1. González JC, Egea J, Del Carmen Godino M, Fernandez-Gomez FJ, Sánchez-Prieto J, Gandía L, García AG, Jordán J, Hernández-Guijo JM. Neuroprotectant minocycline depresses glutamatergic neurotransmission and Ca(2+) signalling in hippocampal neurons. Eur J Neurosci. 2007;26:2481–2495.
    1. Huang WC, Qiao Y, Xu L, Kacimi R, Sun X, Giffard RG, Yenari MA. Direct protection of cultured neurons from ischemia-like injury by minocycline. Anat Cell Biol. 2010;43:325–331.
    1. Liu J, Bartels M, Lu A, Sharp FR. Microglia/macrophages proliferate in striatum and neocortex but not in hippocampus after brief global ischemia that produces ischemic tolerance in gerbil brain. J Cereb Blood Flow Metab. 2001;21:361–373.

Source: PubMed

3
S'abonner