Colchicine pre-treatment and post-treatment does not worsen bleeding or functional outcome after collagenase-induced intracerebral hemorrhage

Cassandra M Wilkinson, Aristeidis H Katsanos, Noam H Sander, Tiffany F C Kung, Frederick Colbourne, Ashkan Shoamanesh, Cassandra M Wilkinson, Aristeidis H Katsanos, Noam H Sander, Tiffany F C Kung, Frederick Colbourne, Ashkan Shoamanesh

Abstract

Patients with intracerebral hemorrhage (ICH) are at increased risk for major ischemic cardiovascular and cerebrovascular events. However, the use of preventative antithrombotic therapy can increase the risk of ICH recurrence and worsen ICH-related outcomes. Colchicine, an anti-inflammatory agent, has the potential to mitigate inflammation-related atherothrombosis and reduce the risk of ischemic vascular events. Here we investigated the safety and efficacy of colchicine when used both before and acutely after ICH. We predicted that daily colchicine administration would not impact our safety measures but would reduce brain injury and improve functional outcomes associated with inflammation reduction. To test this, 0.05 mg/kg colchicine was given orally once daily to rats either before or after they were given a collagenase-induced striatal ICH. We assessed neurological impairments, intra-parenchymal bleeding, Perls positive cells, and brain injury to gauge the therapeutic impact of colchicine on brain injury. Colchicine did not significantly affect bleeding (average = 40.7 μL) at 48 hrs, lesion volume (average = 24.5 mm3) at 14 days, or functional outcome (median neurological deficit scale score at 2 days post-ICH = 4, i.e., modest deficits) from 1-14 days after ICH. Colchicine reduced the volume of Perls positive cells in the perihematomal zone, indicating a reduction in inflammation. Safety measures (body weight, food consumption, water consumption, hydration, body temperature, activity, and pain) were not affected by colchicine. Although colchicine did not confer neuroprotection or functional benefit, it was able to reduce perihematomal inflammation after ICH without increasing bleeding. Thus, our findings suggest that colchicine treatment is safe, unlikely to worsen bleeding, and is unlikely but may reduce secondary injury after an ICH if initiated early post ICH to reduce the risk of ischemic vascular events. These results are informative for the ongoing CoVasc-ICH phase II randomized trial (NCT05159219).

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Fig 1. Experiment 1 safety measures.
Fig 1. Experiment 1 safety measures.
Body weight steadily increased in all animals (A) across experiment 1. Water (B) and food consumption (C) did not decrease by more than 20%, indicating little cause for concern. Pain (D) did not increase by more than 2 points in any animals, indicating the treatment was well tolerated. Body temperature (E) did not differ from baseline by more than 1°C in any animals, at any point in time. Dosing did not affect observed activity levels (F). Temperature and activity are graphed as difference from baseline (averaged hourly to account for circadian rhythms), where a positive reading means that animals were hotter/more active post-treatment. Lines represent individual animals.
Fig 2. Experiment 2.
Fig 2. Experiment 2.
ICH significantly worsened NDS at 48 hours post-ICH (A, p0.999). n = 10/group.
Fig 3. Experiment 2 hematoma volume and…
Fig 3. Experiment 2 hematoma volume and cell counts.
No differences in hematoma volume (A, p = 0.168), or cell density in the peri-hematoma zone (B, p = 0.071) were found at 48 hours post-ICH. C. Representative image of neurons in the PHZ stained using cresyl violet. White scale bar represents 50 μm, black scale bar represents 1 mm.
Fig 4. Experiment 3 edema and behaviour.
Fig 4. Experiment 3 edema and behaviour.
ICH significantly worsened NDS scores at 48 hours post-ICH (A, p

Fig 5. Experiment 4 behaviour and lesion…

Fig 5. Experiment 4 behaviour and lesion volume.

ICH significantly worsened NDS at all times…

Fig 5. Experiment 4 behaviour and lesion volume.
ICH significantly worsened NDS at all times post-ICH (A, p0.507) or ladder task (p = 0.791) were found between groups at any time. Lesion volume at 14 days post-stroke (C) was not affected by colchicine administration (p = 0.9848). D. Representative image of an animal from the colchicine group with a lesion volume of 21.47 mm3. This animal demonstrates ventriculomegaly and the hematoma resolution at 14 days post-ICH. Scale bar represents 1 mm.

Fig 6. Experiment 4 Perls staining.

Post-ICH…

Fig 6. Experiment 4 Perls staining.

Post-ICH colchicine administration did not affect the number of…

Fig 6. Experiment 4 Perls staining.
Post-ICH colchicine administration did not affect the number of Perls positive cells per region of interest (A, p = 0.115), but did reduce the average volume of brain containing Perls positive cells (B, p = 0.018, average area of blue tissue multiplied by section interval and number of sections) at 14 days post-ICH. C. Perls positive cells under 40 × magnification. D. Representative image of Perls staining. White scale bar represents 20 μm, black scale bar represents 1 mm.
Fig 5. Experiment 4 behaviour and lesion…
Fig 5. Experiment 4 behaviour and lesion volume.
ICH significantly worsened NDS at all times post-ICH (A, p0.507) or ladder task (p = 0.791) were found between groups at any time. Lesion volume at 14 days post-stroke (C) was not affected by colchicine administration (p = 0.9848). D. Representative image of an animal from the colchicine group with a lesion volume of 21.47 mm3. This animal demonstrates ventriculomegaly and the hematoma resolution at 14 days post-ICH. Scale bar represents 1 mm.
Fig 6. Experiment 4 Perls staining.
Fig 6. Experiment 4 Perls staining.
Post-ICH colchicine administration did not affect the number of Perls positive cells per region of interest (A, p = 0.115), but did reduce the average volume of brain containing Perls positive cells (B, p = 0.018, average area of blue tissue multiplied by section interval and number of sections) at 14 days post-ICH. C. Perls positive cells under 40 × magnification. D. Representative image of Perls staining. White scale bar represents 20 μm, black scale bar represents 1 mm.

References

    1. Li L, Poon MTC, Samarasekera NE, Perry LA, Moullaali TJ, Rodrigues MA, et al.. Risks of recurrent stroke and all serious vascular events after spontaneous intracerebral haemorrhage: pooled analyses of two population-based studies. Lancet Neurol. 2021;20: 437–447. doi: 10.1016/S1474-4422(21)00075-2
    1. Murthy SB, Zhang C, Diaz I, Levitan EB, Koton S, Bartz TM, et al.. Association between Intracerebral Hemorrhage and Subsequent Arterial Ischemic Events in Participants from 4 Population-Based Cohort Studies. JAMA Neurol. 2021;78: 809–816. doi: 10.1001/jamaneurol.2021.0925
    1. Casolla B, Moulin S, Kyheng M, Hénon H, Labreuche J, Leys D, et al.. Five-Year Risk of Major Ischemic and Hemorrhagic Events After Intracerebral Hemorrhage. Stroke. 2019;50: 1100–1107. doi: 10.1161/STROKEAHA.118.024449
    1. Kuramatsu JB, Gerner ST, Schellinger PD, Glahn J, Endres M, Sobesky J, et al.. Anticoagulant reversal, blood pressure levels, and anticoagulant resumption in patients with anticoagulation-related intracerebral hemorrhage. JAMA—J Am Med Assoc. 2015;313: 824–836. doi: 10.1001/jama.2015.0846
    1. Flaherty ML. Anticoagulant-associated intracerebral hemorrhage. Semin Neurol. 2010;30: 565–572. doi: 10.1055/s-0030-1268866
    1. Shoamanesh A, Patrice Lindsay M, Castellucci LA, Cayley A, Crowther M, de Wit K, et al.. Canadian stroke best practice recommendations: Management of Spontaneous Intracerebral Hemorrhage, 7th Edition Update 2020. Int J Stroke. 2021;16: 321–341. doi: 10.1177/1747493020968424
    1. Murthy SB, Diaz I, Wu X, Merkler AE, Iadecola C, Safford MM, et al.. Risk of Arterial Ischemic Events After Intracerebral Hemorrhage. Stroke. 2020;51: 137–142. doi: 10.1161/STROKEAHA.119.026207
    1. Xu Y, Shoamanesh A, Schulman S, Dowlatshahi D, Salman RAS, Moldovan ID, et al.. Oral anticoagulant re-initiation following intracerebral hemorrhage in non-valvular atrial fibrillation: Global survey of the practices of neurologists, neurosurgeons and thrombosis experts. PLoS One. 2018;13: e0191137. doi: 10.1371/journal.pone.0191137
    1. Kelly P, Weimar C, Lemmens R, Murphy S, Purroy F, Arsovska A, et al.. Colchicine for prevention of vascular inflammation in Non-CardioEmbolic stroke (CONVINCE)–study protocol for a randomised controlled trial. Eur Stroke J. 2021;6: 222–228. doi: 10.1177/2396987320972566
    1. Katsanos AH, Palaiodimou L, Price C, Themistocleous M, Lemmens R, Michopoulos I, et al.. An updated meta-analysis of rcts of colchicine for stroke prevention in patients with coronary artery disease. J Clin Med. 2021;10: 3110. doi: 10.3390/jcm10143110
    1. Masson W, Lobo M, Molinero G, Masson G, Lavalle-Cobo A. Role of Colchicine in Stroke Prevention: An Updated Meta-Analysis. J Stroke Cerebrovasc Dis. 2020;29: 104756. doi: 10.1016/j.jstrokecerebrovasdis.2020.104756
    1. Nidorf SM, Fiolet ATL, Mosterd A, Eikelboom JW, Schut A, Opstal TSJ, et al.. Colchicine in Patients with Chronic Coronary Disease. N Engl J Med. 2020;383: 1838–1847. doi: 10.1056/NEJMoa2021372
    1. Zhang Z, Zhang Z, Lu H, Yang Q, Wu H, Wang J. Microglial Polarization and Inflammatory Mediators After Intracerebral Hemorrhage. Molecular Neurobiology. 2017;54: 1874–1886. doi: 10.1007/s12035-016-9785-6
    1. Zhou Y, Wang Y, Wang J, Anne Stetler R, Yang QW. Inflammation in intracerebral hemorrhage: From mechanisms to clinical translation. Progress in Neurobiology. 2014;115: 25–44. doi: 10.1016/j.pneurobio.2013.11.003
    1. Wang J, Doré S. Inflammation after intracerebral hemorrhage. Journal of Cerebral Blood Flow and Metabolism. 2007;27: 894–908. doi: 10.1038/sj.jcbfm.9600403
    1. Chamorro Á, Hallenbeck J. The harms and benefits of inflammatory and immune responses in vascular disease. Stroke. 2006;37: 291–293. doi: 10.1161/01.STR.0000200561.69611.f8
    1. Cimmino G, Tarallo R, Conte S, Morello A, Pellegrino G, Loffredo FS, et al.. Colchicine reduces platelet aggregation by modulating cytoskeleton rearrangement via inhibition of cofilin and LIM domain kinase 1. Vascul Pharmacol. 2018;111: 62–70. doi: 10.1016/j.vph.2018.09.004
    1. Bouabdallaoui N, Tardif JC, Waters DD, Pinto FJ, Maggioni AP, Diaz R, et al.. Time-to-treatment initiation of colchicine and cardiovascular outcomes after myocardial infarction in the Colchicine Cardiovascular Outcomes Trial (COLCOT). Eur Heart J. 2020;41: 4092–4099. doi: 10.1093/eurheartj/ehaa659
    1. Rosenberg G a., Mun-Bryce S, Wesley M, Kornfeld M. Collagenase-induced intracerebral hemorrhage in rats. Stroke. 1990;21: 801–807. doi: 10.1161/01.str.21.5.801
    1. MacLellan CL, Silasi G, Poon CC, Edmundson CL, Buist R, Peeling J, et al.. Intracerebral hemorrhage models in rat: comparing collagenase to blood infusion. J Cereb Blood Flow Metab. 2008;28: 516–525. doi: 10.1038/sj.jcbfm.9600548
    1. Liddle LJ, Ralhan S, Ward DL, Colbourne F. Translational Intracerebral Hemorrhage Research: Has Current Neuroprotection Research ARRIVEd at a Standard for Experimental Design and Reporting? Transl Stroke Res. 2020;11: 1203–1213. doi: 10.1007/s12975-020-00824-x
    1. Brouwers HB, Chang Y, Falcone GJ, Cai X, Ayres AM, Battey TWK, et al.. Predicting hematoma expansion after primary intracerebral hemorrhage. JAMA Neurol. 2014;71: 158–164. doi: 10.1001/jamaneurol.2013.5433
    1. Del Bigio MR, Yan HJ, Buist R, Peeling J. Experimental intracerebral hemorrhage in rats: Magnetic resonance imaging and histopathological correlates. Stroke. 1996;27: 2312–2320. doi: 10.1161/01.str.27.12.2312
    1. Krafft PR, McBride DW, Lekic T, Rolland WB, Mansell CE, Ma Q, et al.. Correlation between subacute sensorimotor deficits and brain edema in two mouse models of intracerebral hemorrhage. Behav Brain Res. 2014;264: 151–160. doi: 10.1016/j.bbr.2014.01.052
    1. Xi G, Keep RF, Hoff JT. Erythrocytes and delayed brain edema formation following intracerebral hemorrhage in rats. J Neurosurg. 1998;89: 991–996. doi: 10.3171/jns.1998.89.6.0991
    1. du Sert NP, Ahluwalia A, Alam S, Avey MT, Baker M, Browne WJ, et al.. Reporting animal research: Explanation and elaboration for the arrive guidelines 2.0. PLoS Biology. 2020;18: e3000411. doi: 10.1371/journal.pbio.3000411
    1. Nair A, Jacob S. A simple practice guide for dose conversion between animals and human. J Basic Clin Pharm. 2016;7: 27–31. doi: 10.4103/0976-0105.177703
    1. Sukeishi A, Isami K, Hiyama H, Imai S, Nagayasu K, Shirakawa H, et al.. Colchicine alleviates acute postoperative pain but delays wound repair in mice: Roles of neutrophils and macrophages. Mol Pain. 2017;13: 1744806917743680. doi: 10.1177/1744806917743680
    1. Okawa H, Yamawaki-Ogata A, Narita Y, Munakata H, Hashizume R, Usui A. P3112The oral administration of colchicine prevents the progression of aortic aneurysm. Eur Heart J. 2019;40: P3112. doi: 10.1093/eurheartj/ehz745.0187
    1. Holmin S, Mathiesen T. Dexamethasone and colchicine reduce inflammation and delayed oedema following experimental brain contusion. Acta Neurochir (Wien). 1996;138: 418–424. doi: 10.1007/BF01420304
    1. Fiolet ATL, Opstal TSJ, Mosterd A, Eikelboom JW, Jolly SS, Keech AC, et al.. Efficacy and safety of low-dose colchicine in patients with coronary disease: a systematic review and meta-analysis of randomized trials. European Heart Journal. 2021;42: 2765–2775. doi: 10.1093/eurheartj/ehab115
    1. Wilkinson CM, Kalisvaart ACJ, Kung TFC, Maisey DR, Klahr AC, Dickson CT, et al.. The collagenase model of intracerebral hemorrhage in awake, freely moving animals: The effects of isoflurane. Brain Res. 2020;1728: 146593. doi: 10.1016/j.brainres.2019.146593
    1. Paxinos G, Watson C. Index of Structures. Rat Brain Stereotaxic Coord. 1982. doi: 10.1016/b978-0-12-547620-1.50008–4
    1. Sotocinal SG, Sorge RE, Zaloum A, Tuttle AH, Martin LJ, Wieskopf JS, et al.. The Rat Grimace Scale: A partially automated method for quantifying pain in the laboratory rat via facial expressions. Mol Pain. 2011;7: 1744–8069-7-55. doi: 10.1186/1744-8069-7-55
    1. MacLellan CL, Auriat AM, McGie SC, Yan RHY, Huynh HD, De Butte MF, et al.. Gauging recovery after hemorrhagic stroke in rats: Implications for cytoprotection studies. J Cereb Blood Flow Metab. 2006;26: 1031–1042. doi: 10.1038/sj.jcbfm.9600255
    1. Weber R, Wegener S, Ramos-Cabrer P, Wiedermann D, Hoehn M. MRI detection of macrophage activity after experimental stroke in rats: New indicators for late appearance of vascular degradation? Magn Reson Med. 2005;54: 59–66. doi: 10.1002/mrm.20532
    1. Wang X, Ong W, Connor J. Increase in ferric and ferrous iron in the rat hippocampus with time after kainate-induced excitotoxic injury. Exp Brain Res. 2002;143: 137–148. doi: 10.1007/s00221-001-0971-y
    1. Nakamura T, Keep RF, Hua Y, Schallert T, Hoff JT, Xi G. Deferoxamine-induced attenuation of brain edema and neurological deficits in a rat model of intracerebral hemorrhage. J Neurosurg. 2004;100: 672–678. doi: 10.3171/jns.2004.100.4.0672
    1. Williamson MR, Colbourne F. Evidence for Decreased Brain Parenchymal Volume After Large Intracerebral Hemorrhages: a Potential Mechanism Limiting Intracranial Pressure Rises. Transl Stroke Res. 2017;8: 386–396. doi: 10.1007/s12975-017-0530-x
    1. Wilkinson CM, Fedor BA, Aziz JR, Nadeau CA, Brar PS, Clark JJA, et al.. Failure of bumetanide to improve outcome after intracerebral hemorrhage in rat. PLoS One. 2019;14: 1–20. doi: 10.1371/journal.pone.0210660
    1. Oliver V, De Rantere D, Ritchie R, Chisholm J, Hecker KG, Pang DSJ. Psychometric assessment of the rat grimace scale and development of an analgesic intervention score. PLoS One. 2014;9: e97882. doi: 10.1371/journal.pone.0097882
    1. Finkelstein Y, Aks SE, Hutson JR, Juurlink DN, Nguyen P, Dubnov-Raz G, et al.. Colchicine poisoning: The dark side of an ancient drug. Clinical Toxicology. 2010:48: 407–414. doi: 10.3109/15563650.2010.495348
    1. Leung YY, Yao Hui LL, Kraus VB. Colchicine-Update on mechanisms of action and therapeutic uses. Seminars in Arthritis and Rheumatism. 2015;13: 341–350. doi: 10.1016/j.semarthrit.2015.06.013
    1. Kurup R, Galougahi KK, Figtree G, Misra A, Patel S. The Role of Colchicine in Atherosclerotic Cardiovascular Disease. Heart Lung and Circulation. 2021;6: 795–806. doi: 10.1016/j.hlc.2020.11.010
    1. Martínez GJ, Celermajer DS, Patel S. The NLRP3 inflammasome and the emerging role of colchicine to inhibit atherosclerosis-associated inflammation. Atherosclerosis. 2018;269: 262–271. doi: 10.1016/j.atherosclerosis.2017.12.027
    1. Reddel CJ, Pennings GJ, Curnow JL, Chen VM, Kritharides L. Procoagulant Effects of Low-Level Platelet Activation and Its Inhibition by Colchicine. Thromb Haemost. 2018;118: 723–733. doi: 10.1055/s-0038-1636915
    1. Shah B, Allen N, Harchandani B, Pillinger M, Katz S, Sedlis SP, et al.. Effect of Colchicine on Platelet-Platelet and Platelet-Leukocyte Interactions: a Pilot Study in Healthy Subjects. Inflammation. 2016;39: 182–189. doi: 10.1007/s10753-015-0237-7
    1. Reddel CJ, Pennings GJ, Chen VM, Gnanenthiran S, Kritharides L. Colchicine as a Modulator of Platelet Function: A Systematic Review. Semin Thromb Hemost. 2022;48: 552–567. doi: 10.1055/s-0042-1749660
    1. Tardif J-C, Kouz S, Waters DD, Bertrand OF, Diaz R, Maggioni AP, et al.. Efficacy and Safety of Low-Dose Colchicine after Myocardial Infarction. N Engl J Med. 2019;381: 2497–2505. doi: 10.1056/NEJMoa1912388
    1. John RF, Williamson MR, Dietrich K, Colbourne F. Localized Hypothermia Aggravates Bleeding in the Collagenase Model of Intracerebral Hemorrhage. Ther Hypothermia Temp Manag. 2015;5: 19–25. doi: 10.1089/ther.2014.0020
    1. Zheng H, Chen C, Zhang J, Hu Z. Mechanism and therapy of brain edema after intracerebral hemorrhage. Cerebrovascular Diseases. 2016;42: 155–169. doi: 10.1159/000445170
    1. Thiex R, Tsirka SE. Brain edema after intracerebral hemorrhage: mechanisms, treatment options, management strategies, and operative indications. Neurosurgical focus. 2007;22: 1–7. doi: 10.3171/foc.2007.22.5.7
    1. Lan X, Han X, Li Q, Yang QW, Wang J. Modulators of microglial activation and polarization after intracerebral haemorrhage. Nature Reviews Neurology. 2017;13: 420–433. doi: 10.1038/nrneurol.2017.69
    1. Zhu H, Wang Z, Yu J, Yang X, He F, Liu Z, et al.. Role and mechanisms of cytokines in the secondary brain injury after intracerebral hemorrhage. Progress in Neurobiology. 2019;78: 101610. doi: 10.1016/j.pneurobio.2019.03.003
    1. Pennlert J, Asplund K, Carlberg B, Wiklund PG, Wisten A, Åsberg S, et al.. Antithrombotic Treatment Following Intracerebral Hemorrhage in Patients With and Without Atrial Fibrillation. Stroke. 2015;46: 2094–2099. doi: 10.1161/STROKEAHA.115.009087
    1. Al-Shahi Salman R, Dennis MS, Sandercock PAG, Sudlow CLM, Wardlaw JM, Whiteley WN, et al.. Effects of antiplatelet therapy after stroke due to intracerebral haemorrhage (RESTART): a randomised, open-label trial. Lancet. 2019;393: 2613–2623. doi: 10.1016/S0140-6736(19)30840-2

Source: PubMed

3
Prenumerera