Skip to main content
Top
Published in: Neurocritical Care 2/2022

Open Access 18-05-2022 | Subarachnoid Hemorrhage | Original work

Minocycline Attenuates Microglia/Macrophage Phagocytic Activity and Inhibits SAH-Induced Neuronal Cell Death and Inflammation

Authors: Kinga G. Blecharz-Lang, Victor Patsouris, Melina Nieminen-Kelhä, Stefanie Seiffert, Ulf C. Schneider, Peter Vajkoczy

Published in: Neurocritical Care | Issue 2/2022

Login to get access

Abstract

Background

Neuroprotective treatment strategies aiming at interfering with either inflammation or cell death indicate the importance of these mechanisms in the development of brain injury after subarachnoid hemorrhage (SAH). This study was undertaken to evaluate the influence of minocycline on microglia/macrophage cell activity and its neuroprotective and anti-inflammatory impact 14 days after aneurismal SAH in mice.

Methods

Endovascular filament perforation was used to induce SAH in mice. SAH + vehicle-operated mice were used as controls for SAH vehicle-treated mice and SAH + minocycline-treated mice. The drug administration started 4 h after SAH induction and was daily repeated until day 7 post SAH and continued until day 14 every second day. Brain cryosections were immunolabeled for Iba1 to detect microglia/macrophages and NeuN to visualize neurons. Phagocytosis assay was performed to determine the microglia/macrophage activity status. Apoptotic cells were stained using terminal deoxyuridine triphosphate nick end labeling. Real-time quantitative polymerase chain reaction was used to estimate cytokine gene expression.

Results

We observed a significantly reduced phagocytic activity of microglia/macrophages accompanied by a lowered spatial interaction with neurons and reduced neuronal apoptosis achieved by minocycline administration after SAH. Moreover, the SAH-induced overexpression of pro-inflammatory cytokines and neuronal cell death was markedly attenuated by the compound.

Conclusions

Minocycline treatment may be implicated as a therapeutic approach with long-term benefits in the management of secondary brain injury after SAH in a clinically relevant time window.
Appendix
Available only for authorised users
Literature
1.
go back to reference Macdonald RL, Kassell NF, Mayer S, et al. Clazosentan to overcome neurological ischemia and infarction occurring after subarachnoid hemorrhage (CONSCIOUS-1): randomized, double-blind, placebo-controlled phase 2 dose-finding trial. Stroke. 2008;39(11):3015–21.PubMedCrossRef Macdonald RL, Kassell NF, Mayer S, et al. Clazosentan to overcome neurological ischemia and infarction occurring after subarachnoid hemorrhage (CONSCIOUS-1): randomized, double-blind, placebo-controlled phase 2 dose-finding trial. Stroke. 2008;39(11):3015–21.PubMedCrossRef
2.
go back to reference Macdonald RL, Higashida RT, Keller E, et al. Preventing vasospasm improves outcome after aneurysmal subarachnoid hemorrhage: rationale and design of CONSCIOUS-2 and CONSCIOUS-3 trials. Neurocrit Care. 2010;13(3):416–24.PubMedCrossRef Macdonald RL, Higashida RT, Keller E, et al. Preventing vasospasm improves outcome after aneurysmal subarachnoid hemorrhage: rationale and design of CONSCIOUS-2 and CONSCIOUS-3 trials. Neurocrit Care. 2010;13(3):416–24.PubMedCrossRef
3.
go back to reference Alaraj A, Charbel FT, Amin-Hanjani S. Peri-operative measures for treatment and prevention of cerebral vasospasm following subarachnoid hemorrhage. Neurol Res. 2009;31(6):651–9.PubMedCrossRef Alaraj A, Charbel FT, Amin-Hanjani S. Peri-operative measures for treatment and prevention of cerebral vasospasm following subarachnoid hemorrhage. Neurol Res. 2009;31(6):651–9.PubMedCrossRef
4.
go back to reference Wilkins RH, Alexander JA, Odom GL. Intracranial arterial spasm: a clinical analysis. J Neurosurg. 1968;29(2):121–34.PubMedCrossRef Wilkins RH, Alexander JA, Odom GL. Intracranial arterial spasm: a clinical analysis. J Neurosurg. 1968;29(2):121–34.PubMedCrossRef
5.
go back to reference Sehba FA, Pluta RM, Zhang JH. Metamorphosis of subarachnoid hemorrhage research: from delayed vasospasm to early brain injury. Mol Neurobiol. 2011;43(1):27–40.PubMedCrossRef Sehba FA, Pluta RM, Zhang JH. Metamorphosis of subarachnoid hemorrhage research: from delayed vasospasm to early brain injury. Mol Neurobiol. 2011;43(1):27–40.PubMedCrossRef
6.
go back to reference Hansen-Schwartz J, Vajkoczy P, Macdonald RL, Pluta RM, Zhang JH. Cerebral vasospasm: looking beyond vasoconstriction. Trends Pharmacol Sci. 2007;28(6):252–6.PubMedCrossRef Hansen-Schwartz J, Vajkoczy P, Macdonald RL, Pluta RM, Zhang JH. Cerebral vasospasm: looking beyond vasoconstriction. Trends Pharmacol Sci. 2007;28(6):252–6.PubMedCrossRef
7.
go back to reference Pluta RM, Hansen-Schwartz J, Dreier J, et al. Cerebral vasospasm following subarachnoid hemorrhage: time for a new world of thought. Neurol Res. 2009;31(2):151–8.PubMedPubMedCentralCrossRef Pluta RM, Hansen-Schwartz J, Dreier J, et al. Cerebral vasospasm following subarachnoid hemorrhage: time for a new world of thought. Neurol Res. 2009;31(2):151–8.PubMedPubMedCentralCrossRef
8.
go back to reference Schneider UC, Davids AM, Brandenburg S, et al. Microglia inflict delayed brain injury after subarachnoid hemorrhage. Acta Neuropathol. 2015;130(2):215–31.PubMedCrossRef Schneider UC, Davids AM, Brandenburg S, et al. Microglia inflict delayed brain injury after subarachnoid hemorrhage. Acta Neuropathol. 2015;130(2):215–31.PubMedCrossRef
9.
go back to reference Heppner FL, Greter M, Marino D, et al. Experimental autoimmune encephalomyelitis repressed by microglial paralysis. Nat Med. 2005;11(2):146–52.PubMedCrossRef Heppner FL, Greter M, Marino D, et al. Experimental autoimmune encephalomyelitis repressed by microglial paralysis. Nat Med. 2005;11(2):146–52.PubMedCrossRef
10.
go back to reference Sarrafzadeh A, Copin JC, Bengualid DJ, et al. Matrix metalloproteinase-9 concentration in the cerebral extracellular fluid of patients during the acute phase of aneurysmal subarachnoid hemorrhage. Neurol Res. 2012;34(5):455–61.PubMedCrossRef Sarrafzadeh A, Copin JC, Bengualid DJ, et al. Matrix metalloproteinase-9 concentration in the cerebral extracellular fluid of patients during the acute phase of aneurysmal subarachnoid hemorrhage. Neurol Res. 2012;34(5):455–61.PubMedCrossRef
11.
go back to reference Fassbender K, Hodapp B, Rossol S, et al. Inflammatory cytokines in subarachnoid haemorrhage: association with abnormal blood flow velocities in basal cerebral arteries. J Neurol Neurosurg Psych. 2001;70(4):534–7.CrossRef Fassbender K, Hodapp B, Rossol S, et al. Inflammatory cytokines in subarachnoid haemorrhage: association with abnormal blood flow velocities in basal cerebral arteries. J Neurol Neurosurg Psych. 2001;70(4):534–7.CrossRef
12.
go back to reference Schneider UC, Schiffler J, Hakiy N, Horn P, Vajkoczy P. Functional analysis of Pro-inflammatory properties within the cerebrospinal fluid after subarachnoid hemorrhage in vivo and in vitro. J Neuroinflammation. 2012;9:28.PubMedPubMedCentralCrossRef Schneider UC, Schiffler J, Hakiy N, Horn P, Vajkoczy P. Functional analysis of Pro-inflammatory properties within the cerebrospinal fluid after subarachnoid hemorrhage in vivo and in vitro. J Neuroinflammation. 2012;9:28.PubMedPubMedCentralCrossRef
13.
go back to reference Ishikawa M, Kusaka G, Yamaguchi N, et al. Platelet and leukocyte adhesion in the microvasculature at the cerebral surface immediately after subarachnoid hemorrhage. Neurosurgery. 2009;64(3):546–53 (discussion 53-4).PubMedCrossRef Ishikawa M, Kusaka G, Yamaguchi N, et al. Platelet and leukocyte adhesion in the microvasculature at the cerebral surface immediately after subarachnoid hemorrhage. Neurosurgery. 2009;64(3):546–53 (discussion 53-4).PubMedCrossRef
14.
go back to reference Atangana E, Schneider UC, Blecharz K, et al. Intravascular inflammation triggers intracerebral activated microglia and contributes to secondary brain injury after experimental subarachnoid hemorrhage (eSAH). Transl Stroke Res. 2017;8(2):144–56.PubMedCrossRef Atangana E, Schneider UC, Blecharz K, et al. Intravascular inflammation triggers intracerebral activated microglia and contributes to secondary brain injury after experimental subarachnoid hemorrhage (eSAH). Transl Stroke Res. 2017;8(2):144–56.PubMedCrossRef
15.
go back to reference Rifkin BR, Vernillo AT, Golub LM, Ramamurthy NS. Modulation of bone resorption by tetracyclines. Ann N Y Acad Sci. 1994;732:165–80.PubMedCrossRef Rifkin BR, Vernillo AT, Golub LM, Ramamurthy NS. Modulation of bone resorption by tetracyclines. Ann N Y Acad Sci. 1994;732:165–80.PubMedCrossRef
16.
go back to reference Amin AR, Attur MG, Thakker GD, et al. A novel mechanism of action of tetracyclines: effects on nitric oxide synthases. Proc Natl Acad Sci U S A. 1996;93(24):14014–9.PubMedPubMedCentralCrossRef Amin AR, Attur MG, Thakker GD, et al. A novel mechanism of action of tetracyclines: effects on nitric oxide synthases. Proc Natl Acad Sci U S A. 1996;93(24):14014–9.PubMedPubMedCentralCrossRef
17.
go back to reference Wang Z, Meng CJ, Shen XM, et al. Potential contribution of hypoxia-inducible factor-1α, aquaporin-4, and matrix metalloproteinase-9 to blood-brain barrier disruption and brain edema after experimental subarachnoid hemorrhage. J Mol Neurosci. 2012;48(1):273–80.PubMedCrossRef Wang Z, Meng CJ, Shen XM, et al. Potential contribution of hypoxia-inducible factor-1α, aquaporin-4, and matrix metalloproteinase-9 to blood-brain barrier disruption and brain edema after experimental subarachnoid hemorrhage. J Mol Neurosci. 2012;48(1):273–80.PubMedCrossRef
18.
go back to reference Pi R, Li W, Lee NT, et al. Minocycline prevents glutamate-induced apoptosis of cerebellar granule neurons by differential regulation of p38 and Akt pathways. J Neurochem. 2004;91(5):1219–30.PubMedCrossRef Pi R, Li W, Lee NT, et al. Minocycline prevents glutamate-induced apoptosis of cerebellar granule neurons by differential regulation of p38 and Akt pathways. J Neurochem. 2004;91(5):1219–30.PubMedCrossRef
19.
20.
go back to reference Suk K. Minocycline suppresses hypoxic activation of rodent microglia in culture. Neurosci Lett. 2004;366(2):167–71.PubMedCrossRef Suk K. Minocycline suppresses hypoxic activation of rodent microglia in culture. Neurosci Lett. 2004;366(2):167–71.PubMedCrossRef
21.
go back to reference Guo ZD, Wu HT, Sun XC, Zhang XD, Zhang JH. Protection of minocycline on early brain injury after subarachnoid hemorrhage in rats. Acta Neurochir Suppl. 2011;110(Pt 1):71–4.PubMed Guo ZD, Wu HT, Sun XC, Zhang XD, Zhang JH. Protection of minocycline on early brain injury after subarachnoid hemorrhage in rats. Acta Neurochir Suppl. 2011;110(Pt 1):71–4.PubMed
22.
go back to reference Sherchan P, Lekic T, Suzuki H, et al. Minocycline improves functional outcomes, memory deficits, and histopathology after endovascular perforation-induced subarachnoid hemorrhage in rats. J Neurotrauma. 2011;28(12):2503–12.PubMedPubMedCentralCrossRef Sherchan P, Lekic T, Suzuki H, et al. Minocycline improves functional outcomes, memory deficits, and histopathology after endovascular perforation-induced subarachnoid hemorrhage in rats. J Neurotrauma. 2011;28(12):2503–12.PubMedPubMedCentralCrossRef
23.
go back to reference Li J, Chen J, Mo H, et al. Minocycline protects against NLRP3 inflammasome-induced inflammation and P53-associated apoptosis in early brain injury after subarachnoid hemorrhage. Mol Neurobiol. 2016;53(4):2668–78.PubMedCrossRef Li J, Chen J, Mo H, et al. Minocycline protects against NLRP3 inflammasome-induced inflammation and P53-associated apoptosis in early brain injury after subarachnoid hemorrhage. Mol Neurobiol. 2016;53(4):2668–78.PubMedCrossRef
24.
go back to reference Kreutzberg GW. Microglia: a sensor for pathological events in the CNS. Trends Neurosci. 1996;19(8):312–8.PubMedCrossRef Kreutzberg GW. Microglia: a sensor for pathological events in the CNS. Trends Neurosci. 1996;19(8):312–8.PubMedCrossRef
25.
go back to reference McGeer EG, McGeer PL. Brain inflammation in Alzheimer disease and the therapeutic implications. Curr Pharm Des. 1999;5(10):821–36.PubMed McGeer EG, McGeer PL. Brain inflammation in Alzheimer disease and the therapeutic implications. Curr Pharm Des. 1999;5(10):821–36.PubMed
26.
go back to reference Beal MF. Aging, energy, and oxidative stress in neurodegenerative diseases. Ann Neurol. 1995;38(3):357–66.PubMedCrossRef Beal MF. Aging, energy, and oxidative stress in neurodegenerative diseases. Ann Neurol. 1995;38(3):357–66.PubMedCrossRef
27.
go back to reference Hanisch UK, Kettenmann H. Microglia: active sensor and versatile effector cells in the normal and pathologic brain. Nat Neurosci. 2007;10(11):1387–94.PubMedCrossRef Hanisch UK, Kettenmann H. Microglia: active sensor and versatile effector cells in the normal and pathologic brain. Nat Neurosci. 2007;10(11):1387–94.PubMedCrossRef
28.
go back to reference Szalay G, Martinecz B, Lénárt N, et al. Microglia protect against brain injury and their selective elimination dysregulates neuronal network activity after stroke. Nat Commun. 2016;7:11499.PubMedPubMedCentralCrossRef Szalay G, Martinecz B, Lénárt N, et al. Microglia protect against brain injury and their selective elimination dysregulates neuronal network activity after stroke. Nat Commun. 2016;7:11499.PubMedPubMedCentralCrossRef
30.
go back to reference de Beer MC, Zhao Z, Webb NR, van der Westhuyzen DR, de Villiers WJ. Lack of a direct role for macrosialin in oxidized LDL metabolism. J Lipid Res. 2003;44(4):674–85.PubMedCrossRef de Beer MC, Zhao Z, Webb NR, van der Westhuyzen DR, de Villiers WJ. Lack of a direct role for macrosialin in oxidized LDL metabolism. J Lipid Res. 2003;44(4):674–85.PubMedCrossRef
31.
go back to reference Huang CY, Chen YL, Li AH, Lu JC, Wang HL. Minocycline, a microglial inhibitor, blocks spinal CCL2-induced heat hyperalgesia and augmentation of glutamatergic transmission in substantia gelatinosa neurons. J Neuroinflammation. 2014;11:7.PubMedPubMedCentralCrossRef Huang CY, Chen YL, Li AH, Lu JC, Wang HL. Minocycline, a microglial inhibitor, blocks spinal CCL2-induced heat hyperalgesia and augmentation of glutamatergic transmission in substantia gelatinosa neurons. J Neuroinflammation. 2014;11:7.PubMedPubMedCentralCrossRef
32.
go back to reference Zhao P, Waxman SG, Hains BC. Extracellular signal-regulated kinase-regulated microglia-neuron signaling by prostaglandin E2 contributes to pain after spinal cord injury. J Neurosci. 2007;27(9):2357–68.PubMedPubMedCentralCrossRef Zhao P, Waxman SG, Hains BC. Extracellular signal-regulated kinase-regulated microglia-neuron signaling by prostaglandin E2 contributes to pain after spinal cord injury. J Neurosci. 2007;27(9):2357–68.PubMedPubMedCentralCrossRef
33.
go back to reference Ledeboer A, Sloane EM, Milligan ED, et al. Minocycline attenuates mechanical allodynia and proinflammatory cytokine expression in rat models of pain facilitation. Pain. 2005;115(1–2):71–83.PubMedCrossRef Ledeboer A, Sloane EM, Milligan ED, et al. Minocycline attenuates mechanical allodynia and proinflammatory cytokine expression in rat models of pain facilitation. Pain. 2005;115(1–2):71–83.PubMedCrossRef
34.
go back to reference Yrjänheikki J, Tikka T, Keinänen R, et al. 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(23):13496–500.PubMedPubMedCentralCrossRef Yrjänheikki J, Tikka T, Keinänen R, et al. 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(23):13496–500.PubMedPubMedCentralCrossRef
36.
go back to reference Patel RN, Attur MG, Dave MN, et al. A novel mechanism of action of chemically modified tetracyclines: inhibition of COX-2-mediated prostaglandin E2 production. J Immunol. 1999;163(6):3459–67.PubMed Patel RN, Attur MG, Dave MN, et al. A novel mechanism of action of chemically modified tetracyclines: inhibition of COX-2-mediated prostaglandin E2 production. J Immunol. 1999;163(6):3459–67.PubMed
37.
go back to reference Gong K, Zou X, Fuchs PN, Lin Q. Minocycline inhibits neurogenic inflammation by blocking the effects of tumor necrosis factor-α. Clin Exp Pharmacol Physiol. 2015;42:940–9.PubMedCrossRef Gong K, Zou X, Fuchs PN, Lin Q. Minocycline inhibits neurogenic inflammation by blocking the effects of tumor necrosis factor-α. Clin Exp Pharmacol Physiol. 2015;42:940–9.PubMedCrossRef
38.
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.PubMedCrossRef Vezzani A, Viviani B. Neuromodulatory properties of inflammatory cytokines and their impact on neuronal excitability. Neuropharmacology. 2015;96(Pt A):70–82.PubMedCrossRef
39.
go back to reference Pugazhenthi S, Zhang Y, Bouchard R, Mahaffey G. Induction of an inflammatory loop by interleukin-1β and tumor necrosis factor-α involves NF-kB and STAT-1 in differentiated human neuroprogenitor cells. PLoS ONE. 2013;8(7): e69585.PubMedPubMedCentralCrossRef Pugazhenthi S, Zhang Y, Bouchard R, Mahaffey G. Induction of an inflammatory loop by interleukin-1β and tumor necrosis factor-α involves NF-kB and STAT-1 in differentiated human neuroprogenitor cells. PLoS ONE. 2013;8(7): e69585.PubMedPubMedCentralCrossRef
40.
go back to reference Snow WM, Stoesz BM, Kelly DM, Albensi BC. Roles for NF-κB and gene targets of NF-κB in synaptic plasticity, memory, and navigation. Mol Neurobiol. 2014;49(2):757–70.PubMedCrossRef Snow WM, Stoesz BM, Kelly DM, Albensi BC. Roles for NF-κB and gene targets of NF-κB in synaptic plasticity, memory, and navigation. Mol Neurobiol. 2014;49(2):757–70.PubMedCrossRef
41.
go back to reference Liaury K, Miyaoka T, Tsumori T, et al. Morphological features of microglial cells in the hippocampal dentate gyrus of Gunn rat: a possible schizophrenia animal model. J Neuroinflammation. 2012;9:56.PubMedPubMedCentralCrossRef Liaury K, Miyaoka T, Tsumori T, et al. Morphological features of microglial cells in the hippocampal dentate gyrus of Gunn rat: a possible schizophrenia animal model. J Neuroinflammation. 2012;9:56.PubMedPubMedCentralCrossRef
42.
go back to reference Tikka T, Fiebich BL, Goldsteins G, Keinanen R, Koistinaho J. Minocycline, a tetracycline derivative, is neuroprotective against excitotoxicity by inhibiting activation and proliferation of microglia. J Neurosci. 2001;21(8):2580–8.PubMedPubMedCentralCrossRef Tikka T, Fiebich BL, Goldsteins G, Keinanen R, Koistinaho J. Minocycline, a tetracycline derivative, is neuroprotective against excitotoxicity by inhibiting activation and proliferation of microglia. J Neurosci. 2001;21(8):2580–8.PubMedPubMedCentralCrossRef
43.
go back to reference Gomes JA, Selim M, Cotleur A, et al. Brain iron metabolism and brain injury following subarachnoid hemorrhage: iCeFISH-pilot (CSF iron in SAH). Neurocrit Care. 2014;21(2):285–93.PubMedPubMedCentralCrossRef Gomes JA, Selim M, Cotleur A, et al. Brain iron metabolism and brain injury following subarachnoid hemorrhage: iCeFISH-pilot (CSF iron in SAH). Neurocrit Care. 2014;21(2):285–93.PubMedPubMedCentralCrossRef
44.
go back to reference Vellimana AK, Zhou ML, Singh I, et al. Minocycline protects against delayed cerebral ischemia after subarachnoid hemorrhage via matrix metalloproteinase-9 inhibition. Ann Clin Transl Neurol. 2017;4(12):865–76.PubMedPubMedCentralCrossRef Vellimana AK, Zhou ML, Singh I, et al. Minocycline protects against delayed cerebral ischemia after subarachnoid hemorrhage via matrix metalloproteinase-9 inhibition. Ann Clin Transl Neurol. 2017;4(12):865–76.PubMedPubMedCentralCrossRef
45.
go back to reference Li J, McCullough LD. Sex differences in minocycline-induced neuroprotection after experimental stroke. J Cereb Blood Flow Metab. 2009;29(4):670–4.PubMedCrossRef Li J, McCullough LD. Sex differences in minocycline-induced neuroprotection after experimental stroke. J Cereb Blood Flow Metab. 2009;29(4):670–4.PubMedCrossRef
Metadata
Title
Minocycline Attenuates Microglia/Macrophage Phagocytic Activity and Inhibits SAH-Induced Neuronal Cell Death and Inflammation
Authors
Kinga G. Blecharz-Lang
Victor Patsouris
Melina Nieminen-Kelhä
Stefanie Seiffert
Ulf C. Schneider
Peter Vajkoczy
Publication date
18-05-2022
Publisher
Springer US
Published in
Neurocritical Care / Issue 2/2022
Print ISSN: 1541-6933
Electronic ISSN: 1556-0961
DOI
https://doi.org/10.1007/s12028-022-01511-5

Other articles of this Issue 2/2022

Neurocritical Care 2/2022 Go to the issue