Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2024

Open Access 01-12-2024 | Central Nervous System Trauma | Research

Fibrin promotes oxidative stress and neuronal loss in traumatic brain injury via innate immune activation

Authors: Terry Dean, Andrew S. Mendiola, Zhaoqi Yan, Rosa Meza-Acevedo, Belinda Cabriga, Katerina Akassoglou, Jae Kyu Ryu

Published in: Journal of Neuroinflammation | Issue 1/2024

Login to get access

Abstract

Background

Traumatic brain injury (TBI) causes significant blood-brain barrier (BBB) breakdown, resulting in the extravasation of blood proteins into the brain. The impact of blood proteins, especially fibrinogen, on inflammation and neurodegeneration post-TBI is not fully understood, highlighting a critical gap in our comprehension of TBI pathology and its connection to innate immune activation.

Methods

We combined vascular casting with 3D imaging of solvent-cleared organs (uDISCO) to study the spatial distribution of the blood coagulation protein fibrinogen in large, intact brain volumes and assessed the temporal regulation of the fibrin(ogen) deposition by immunohistochemistry in a murine model of TBI. Fibrin(ogen) deposition and innate immune cell markers were co-localized by immunohistochemistry in mouse and human brains after TBI. We assessed the role of fibrinogen in TBI using unbiased transcriptomics, flow cytometry and immunohistochemistry for innate immune and neuronal markers in Fggγ390–396A knock-in mice, which express a mutant fibrinogen that retains normal clotting function, but lacks the γ390–396 binding motif to CD11b/CD18 integrin receptor.

Results

We show that cerebral fibrinogen deposits were associated with activated innate immune cells in both human and murine TBI. Genetic elimination of fibrin-CD11b interaction reduced peripheral monocyte recruitment and the activation of inflammatory and reactive oxygen species (ROS) gene pathways in microglia and macrophages after TBI. Blockade of the fibrin-CD11b interaction was also protective from oxidative stress damage and cortical loss after TBI.

Conclusions

These data suggest that fibrinogen is a regulator of innate immune activation and neurodegeneration in TBI. Abrogating post-injury neuroinflammation by selective blockade of fibrin’s inflammatory functions may have implications for long-term neurologic recovery following brain trauma.
Appendix
Available only for authorised users
Literature
1.
go back to reference Dewan MC et al. Estimating the global incidence of traumatic brain injury. J Neurosurg, 2018: p. 1–18. Dewan MC et al. Estimating the global incidence of traumatic brain injury. J Neurosurg, 2018: p. 1–18.
3.
go back to reference Algattas H, Huang JH. Traumatic brain Injury pathophysiology and treatments: early, intermediate, and late phases post-injury. Int J Mol Sci. 2013;15(1):309–41.PubMedPubMedCentralCrossRef Algattas H, Huang JH. Traumatic brain Injury pathophysiology and treatments: early, intermediate, and late phases post-injury. Int J Mol Sci. 2013;15(1):309–41.PubMedPubMedCentralCrossRef
4.
go back to reference Cash A, Theus MH. Mechanisms of blood-brain barrier dysfunction in traumatic brain Injury. Int J Mol Sci, 2020. 21(9). Cash A, Theus MH. Mechanisms of blood-brain barrier dysfunction in traumatic brain Injury. Int J Mol Sci, 2020. 21(9).
5.
go back to reference Hay JR, et al. Blood-brain barrier disruption is an early event that May Persist for many years after traumatic brain Injury in humans. J Neuropathol Exp Neurol. 2015;74(12):1147–57.PubMed Hay JR, et al. Blood-brain barrier disruption is an early event that May Persist for many years after traumatic brain Injury in humans. J Neuropathol Exp Neurol. 2015;74(12):1147–57.PubMed
6.
7.
go back to reference Flick MJ, et al. Leukocyte engagement of fibrin(ogen) via the integrin receptor alphaMbeta2/Mac-1 is critical for host inflammatory response in vivo. J Clin Invest. 2004;113(11):1596–606.PubMedPubMedCentralCrossRef Flick MJ, et al. Leukocyte engagement of fibrin(ogen) via the integrin receptor alphaMbeta2/Mac-1 is critical for host inflammatory response in vivo. J Clin Invest. 2004;113(11):1596–606.PubMedPubMedCentralCrossRef
8.
go back to reference Adams RA, et al. The fibrin-derived gamma377-395 peptide inhibits microglia activation and suppresses relapsing paralysis in central nervous system autoimmune disease. J Exp Med. 2007;204(3):571–82.PubMedPubMedCentralCrossRef Adams RA, et al. The fibrin-derived gamma377-395 peptide inhibits microglia activation and suppresses relapsing paralysis in central nervous system autoimmune disease. J Exp Med. 2007;204(3):571–82.PubMedPubMedCentralCrossRef
9.
go back to reference Davalos D, et al. Fibrinogen-induced perivascular microglial clustering is required for the development of axonal damage in neuroinflammation. Nat Commun. 2012;3:1227.PubMedCrossRef Davalos D, et al. Fibrinogen-induced perivascular microglial clustering is required for the development of axonal damage in neuroinflammation. Nat Commun. 2012;3:1227.PubMedCrossRef
10.
go back to reference Merlini M, et al. Fibrinogen induces microglia-mediated spine elimination and cognitive impairment in an Alzheimer’s Disease Model. Neuron. 2019;101(6):1099–e11086.PubMedPubMedCentralCrossRef Merlini M, et al. Fibrinogen induces microglia-mediated spine elimination and cognitive impairment in an Alzheimer’s Disease Model. Neuron. 2019;101(6):1099–e11086.PubMedPubMedCentralCrossRef
11.
12.
13.
14.
go back to reference Jenkins DR et al. The contribution of fibrinogen to inflammation and neuronal density in human traumatic brain Injury. J Neurotrauma, 2018. Jenkins DR et al. The contribution of fibrinogen to inflammation and neuronal density in human traumatic brain Injury. J Neurotrauma, 2018.
15.
go back to reference Witcher KG, et al. Traumatic brain Injury causes chronic cortical inflammation and neuronal dysfunction mediated by Microglia. J Neurosci. 2021;41(7):1597–616.PubMedPubMedCentralCrossRef Witcher KG, et al. Traumatic brain Injury causes chronic cortical inflammation and neuronal dysfunction mediated by Microglia. J Neurosci. 2021;41(7):1597–616.PubMedPubMedCentralCrossRef
16.
go back to reference Morganti JM, et al. CCR2 antagonism alters brain macrophage polarization and ameliorates cognitive dysfunction induced by traumatic brain injury. J Neurosci. 2015;35(2):748–60.PubMedPubMedCentralCrossRef Morganti JM, et al. CCR2 antagonism alters brain macrophage polarization and ameliorates cognitive dysfunction induced by traumatic brain injury. J Neurosci. 2015;35(2):748–60.PubMedPubMedCentralCrossRef
17.
go back to reference Jung S, et al. Analysis of Fractalkine receptor CX3CR1 function by targeted deletion and green fluorescent protein reporter Gene Insertion. Mol Cell Biol. 2000;20(11):4106–14.PubMedPubMedCentralCrossRef Jung S, et al. Analysis of Fractalkine receptor CX3CR1 function by targeted deletion and green fluorescent protein reporter Gene Insertion. Mol Cell Biol. 2000;20(11):4106–14.PubMedPubMedCentralCrossRef
18.
go back to reference Saederup N, et al. Selective chemokine receptor usage by central nervous system myeloid cells in CCR2-red fluorescent protein knock-in mice. PLoS ONE. 2010;5(10):e13693.PubMedPubMedCentralCrossRef Saederup N, et al. Selective chemokine receptor usage by central nervous system myeloid cells in CCR2-red fluorescent protein knock-in mice. PLoS ONE. 2010;5(10):e13693.PubMedPubMedCentralCrossRef
19.
go back to reference Petersen MA, et al. Fibrinogen activates BMP Signaling in Oligodendrocyte Progenitor cells and inhibits remyelination after vascular damage. Neuron. 2017;96(5):1003–e10127.PubMedPubMedCentralCrossRef Petersen MA, et al. Fibrinogen activates BMP Signaling in Oligodendrocyte Progenitor cells and inhibits remyelination after vascular damage. Neuron. 2017;96(5):1003–e10127.PubMedPubMedCentralCrossRef
20.
21.
go back to reference Pan C, et al. Shrinkage-mediated imaging of entire organs and organisms using uDISCO. Nat Methods. 2016;13(10):859–67.PubMedCrossRef Pan C, et al. Shrinkage-mediated imaging of entire organs and organisms using uDISCO. Nat Methods. 2016;13(10):859–67.PubMedCrossRef
22.
go back to reference Petersen MA, et al. BMP receptor blockade overcomes extrinsic inhibition of remyelination and restores neurovascular homeostasis. Brain. 2021;144(8):2291–301.PubMedPubMedCentralCrossRef Petersen MA, et al. BMP receptor blockade overcomes extrinsic inhibition of remyelination and restores neurovascular homeostasis. Brain. 2021;144(8):2291–301.PubMedPubMedCentralCrossRef
23.
go back to reference Ewels PA, et al. The nf-core framework for community-curated bioinformatics pipelines. Nat Biotechnol. 2020;38(3):276–8.PubMedCrossRef Ewels PA, et al. The nf-core framework for community-curated bioinformatics pipelines. Nat Biotechnol. 2020;38(3):276–8.PubMedCrossRef
24.
go back to reference Dunham J, et al. Oxidative Injury and Iron Redistribution are pathological hallmarks of Marmoset Experimental Autoimmune Encephalomyelitis. J Neuropathol Exp Neurol. 2017;76(6):467–78.PubMedCrossRef Dunham J, et al. Oxidative Injury and Iron Redistribution are pathological hallmarks of Marmoset Experimental Autoimmune Encephalomyelitis. J Neuropathol Exp Neurol. 2017;76(6):467–78.PubMedCrossRef
25.
go back to reference Akiyama H, et al. Expression of MRP14, 27E10, interferon-alpha and leukocyte common antigen by reactive microglia in postmortem human brain tissue. J Neuroimmunol. 1994;50(2):195–201.PubMedCrossRef Akiyama H, et al. Expression of MRP14, 27E10, interferon-alpha and leukocyte common antigen by reactive microglia in postmortem human brain tissue. J Neuroimmunol. 1994;50(2):195–201.PubMedCrossRef
26.
go back to reference Gay D, Esiri M. Blood-brain barrier damage in acute multiple sclerosis plaques. An immunocytological study. Brain. 1991;114:557–72.PubMedCrossRef Gay D, Esiri M. Blood-brain barrier damage in acute multiple sclerosis plaques. An immunocytological study. Brain. 1991;114:557–72.PubMedCrossRef
27.
go back to reference Tsai PS, et al. Correlations of neuronal and microvascular densities in murine cortex revealed by direct counting and colocalization of nuclei and vessels. J Neurosci. 2009;29(46):14553–70.PubMedPubMedCentralCrossRef Tsai PS, et al. Correlations of neuronal and microvascular densities in murine cortex revealed by direct counting and colocalization of nuclei and vessels. J Neurosci. 2009;29(46):14553–70.PubMedPubMedCentralCrossRef
28.
go back to reference Baskaya MK, et al. The biphasic opening of the blood-brain barrier in the cortex and hippocampus after traumatic brain injury in rats. Neurosci Lett. 1997;226(1):33–6.PubMedCrossRef Baskaya MK, et al. The biphasic opening of the blood-brain barrier in the cortex and hippocampus after traumatic brain injury in rats. Neurosci Lett. 1997;226(1):33–6.PubMedCrossRef
29.
go back to reference Silva LM, et al. Fibrin is a critical regulator of neutrophil effector function at the oral mucosal barrier. Science. 2021;374(6575):eabl5450.PubMedCrossRef Silva LM, et al. Fibrin is a critical regulator of neutrophil effector function at the oral mucosal barrier. Science. 2021;374(6575):eabl5450.PubMedCrossRef
30.
go back to reference Fibrin induces neurotoxic microglia gene programs in neurodegeneration. Nat Immunol, 2023. 24(7): p. 1062–3. Fibrin induces neurotoxic microglia gene programs in neurodegeneration. Nat Immunol, 2023. 24(7): p. 1062–3.
32.
go back to reference Swanson CA, Burns JC, Peterson BM. Low plasma D-dimer concentration predicts the absence of traumatic brain injury in children. J Trauma. 2010;68(5):1072–7.PubMedPubMedCentral Swanson CA, Burns JC, Peterson BM. Low plasma D-dimer concentration predicts the absence of traumatic brain injury in children. J Trauma. 2010;68(5):1072–7.PubMedPubMedCentral
33.
go back to reference DeFazio MV, et al. The potential utility of blood-derived biochemical markers as indicators of early clinical trends following severe traumatic brain injury. World Neurosurg. 2014;81(1):151–8.PubMedCrossRef DeFazio MV, et al. The potential utility of blood-derived biochemical markers as indicators of early clinical trends following severe traumatic brain injury. World Neurosurg. 2014;81(1):151–8.PubMedCrossRef
34.
go back to reference Lv K, et al. Impact of fibrinogen level on the prognosis of patients with traumatic brain injury: a single-center analysis of 2570 patients. World J Emerg Surg. 2020;15(1):54.PubMedPubMedCentralCrossRef Lv K, et al. Impact of fibrinogen level on the prognosis of patients with traumatic brain injury: a single-center analysis of 2570 patients. World J Emerg Surg. 2020;15(1):54.PubMedPubMedCentralCrossRef
35.
go back to reference Ito H, et al. Association of Extravascular Leakage on computed tomography angiography with fibrinogen levels at admission in patients with traumatic brain Injury. Neurotrauma Rep. 2022;4(1):3–13.PubMedPubMedCentralCrossRef Ito H, et al. Association of Extravascular Leakage on computed tomography angiography with fibrinogen levels at admission in patients with traumatic brain Injury. Neurotrauma Rep. 2022;4(1):3–13.PubMedPubMedCentralCrossRef
36.
go back to reference Sabouri M, et al. Efficacy and safety of fibrinogen administration in acute post-traumatic hypofibrinogenemia in isolated severe traumatic brain injury: a randomized clinical trial. J Clin Neurosci. 2022;101:204–11.PubMedCrossRef Sabouri M, et al. Efficacy and safety of fibrinogen administration in acute post-traumatic hypofibrinogenemia in isolated severe traumatic brain injury: a randomized clinical trial. J Clin Neurosci. 2022;101:204–11.PubMedCrossRef
37.
go back to reference Kirk J, et al. Tight junctional abnormality in multiple sclerosis white matter affects all calibres of vessel and is associated with blood-brain barrier leakage and active demyelination. J Pathol. 2003;201(2):319–27.PubMedCrossRef Kirk J, et al. Tight junctional abnormality in multiple sclerosis white matter affects all calibres of vessel and is associated with blood-brain barrier leakage and active demyelination. J Pathol. 2003;201(2):319–27.PubMedCrossRef
38.
go back to reference Ryu JK, McLarnon JG. A leaky blood-brain barrier, fibrinogen infiltration and microglial reactivity in inflamed Alzheimer’s disease brain. J Cell Mol Med. 2009;13(9A):2911–25.PubMedCrossRef Ryu JK, McLarnon JG. A leaky blood-brain barrier, fibrinogen infiltration and microglial reactivity in inflamed Alzheimer’s disease brain. J Cell Mol Med. 2009;13(9A):2911–25.PubMedCrossRef
39.
go back to reference Massberg S, et al. Fibrinogen deposition at the postischemic vessel wall promotes platelet adhesion during ischemia-reperfusion in vivo. Blood. 1999;94(11):3829–38.PubMedCrossRef Massberg S, et al. Fibrinogen deposition at the postischemic vessel wall promotes platelet adhesion during ischemia-reperfusion in vivo. Blood. 1999;94(11):3829–38.PubMedCrossRef
40.
go back to reference Davalos D, Akassoglou K. Fibrinogen as a key regulator of inflammation in disease. Semin Immunopathol. 2012;34(1):43–62.PubMedCrossRef Davalos D, Akassoglou K. Fibrinogen as a key regulator of inflammation in disease. Semin Immunopathol. 2012;34(1):43–62.PubMedCrossRef
41.
go back to reference van Vliet EA, et al. Long-lasting blood-brain barrier dysfunction and neuroinflammation after traumatic brain injury. Neurobiol Dis. 2020;145:105080.PubMedCrossRef van Vliet EA, et al. Long-lasting blood-brain barrier dysfunction and neuroinflammation after traumatic brain injury. Neurobiol Dis. 2020;145:105080.PubMedCrossRef
43.
go back to reference Fleminger S, et al. Head injury as a risk factor for Alzheimer’s disease: the evidence 10 years on; a partial replication. J Neurol Neurosurg Psychiatry. 2003;74(7):857–62.PubMedPubMedCentralCrossRef Fleminger S, et al. Head injury as a risk factor for Alzheimer’s disease: the evidence 10 years on; a partial replication. J Neurol Neurosurg Psychiatry. 2003;74(7):857–62.PubMedPubMedCentralCrossRef
44.
go back to reference Mortimer JA, et al. Head trauma as a risk factor for Alzheimer’s disease: a collaborative re-analysis of case-control studies. EURODEM risk Factors Research Group. Int J Epidemiol. 1991;20(Suppl 2):S28–35.PubMedCrossRef Mortimer JA, et al. Head trauma as a risk factor for Alzheimer’s disease: a collaborative re-analysis of case-control studies. EURODEM risk Factors Research Group. Int J Epidemiol. 1991;20(Suppl 2):S28–35.PubMedCrossRef
45.
go back to reference Graham NSN, et al. Distinct patterns of neurodegeneration after TBI and in Alzheimer’s disease. Alzheimers Dement. 2023;19(7):3065–77.PubMedCrossRef Graham NSN, et al. Distinct patterns of neurodegeneration after TBI and in Alzheimer’s disease. Alzheimers Dement. 2023;19(7):3065–77.PubMedCrossRef
46.
47.
go back to reference Hsieh CL, et al. CCR2 deficiency impairs macrophage infiltration and improves cognitive function after traumatic brain injury. J Neurotrauma. 2014;31(20):1677–88.PubMedPubMedCentralCrossRef Hsieh CL, et al. CCR2 deficiency impairs macrophage infiltration and improves cognitive function after traumatic brain injury. J Neurotrauma. 2014;31(20):1677–88.PubMedPubMedCentralCrossRef
49.
go back to reference Zanier ER, et al. Fractalkine receptor Deficiency is Associated with Early Protection but late worsening of Outcome following Brain Trauma in mice. J Neurotrauma. 2016;33(11):1060–72.PubMedPubMedCentralCrossRef Zanier ER, et al. Fractalkine receptor Deficiency is Associated with Early Protection but late worsening of Outcome following Brain Trauma in mice. J Neurotrauma. 2016;33(11):1060–72.PubMedPubMedCentralCrossRef
50.
go back to reference Kochanek PM, et al. Paths to successful translation of New therapies for severe traumatic brain Injury in the Golden Age of traumatic brain Injury Research: a Pittsburgh Vision. J Neurotrauma. 2020;37(22):2353–71.PubMedPubMedCentralCrossRef Kochanek PM, et al. Paths to successful translation of New therapies for severe traumatic brain Injury in the Golden Age of traumatic brain Injury Research: a Pittsburgh Vision. J Neurotrauma. 2020;37(22):2353–71.PubMedPubMedCentralCrossRef
53.
go back to reference Roberts I, et al. Effect of intravenous corticosteroids on death within 14 days in 10008 adults with clinically significant head injury (MRC CRASH trial): randomised placebo-controlled trial. Lancet. 2004;364(9442):1321–8.PubMedCrossRef Roberts I, et al. Effect of intravenous corticosteroids on death within 14 days in 10008 adults with clinically significant head injury (MRC CRASH trial): randomised placebo-controlled trial. Lancet. 2004;364(9442):1321–8.PubMedCrossRef
54.
go back to reference Merlini M, et al. Fibrinogen induces microglia-mediated spine elimination and cognitive impairment in Alzheimer’s Disease. Neuron. 2019;101(6):1099–108.PubMedPubMedCentralCrossRef Merlini M, et al. Fibrinogen induces microglia-mediated spine elimination and cognitive impairment in Alzheimer’s Disease. Neuron. 2019;101(6):1099–108.PubMedPubMedCentralCrossRef
55.
go back to reference Flick MJ, et al. Fibrin(ogen) exacerbates inflammatory joint disease through a mechanism linked to the integrin alphaMbeta2 binding motif. J Clin Invest. 2007;117(11):3224–35.PubMedPubMedCentralCrossRef Flick MJ, et al. Fibrin(ogen) exacerbates inflammatory joint disease through a mechanism linked to the integrin alphaMbeta2 binding motif. J Clin Invest. 2007;117(11):3224–35.PubMedPubMedCentralCrossRef
56.
go back to reference Steinbrecher KA, et al. Colitis-associated cancer is dependent on the interplay between the hemostatic and inflammatory systems and supported by integrin alpha(M)beta(2) engagement of fibrinogen. Cancer Res. 2010;70(7):2634–43.PubMedPubMedCentralCrossRef Steinbrecher KA, et al. Colitis-associated cancer is dependent on the interplay between the hemostatic and inflammatory systems and supported by integrin alpha(M)beta(2) engagement of fibrinogen. Cancer Res. 2010;70(7):2634–43.PubMedPubMedCentralCrossRef
57.
go back to reference Gveric D, et al. Impaired fibrinolysis in multiple sclerosis: a role for tissue plasminogen activator inhibitors. Brain. 2003;126(Pt 7):1590–8.PubMedCrossRef Gveric D, et al. Impaired fibrinolysis in multiple sclerosis: a role for tissue plasminogen activator inhibitors. Brain. 2003;126(Pt 7):1590–8.PubMedCrossRef
58.
go back to reference Cortes-Canteli M, et al. Fibrinogen and beta-amyloid association alters thrombosis and fibrinolysis: a possible contributing factor to Alzheimer’s disease. Neuron. 2010;66(5):695–709.PubMedPubMedCentralCrossRef Cortes-Canteli M, et al. Fibrinogen and beta-amyloid association alters thrombosis and fibrinolysis: a possible contributing factor to Alzheimer’s disease. Neuron. 2010;66(5):695–709.PubMedPubMedCentralCrossRef
61.
go back to reference Doran SJ, et al. Sex differences in Acute Neuroinflammation after Experimental Traumatic Brain Injury are mediated by infiltrating myeloid cells. J Neurotrauma. 2019;36(7):1040–53.PubMedPubMedCentralCrossRef Doran SJ, et al. Sex differences in Acute Neuroinflammation after Experimental Traumatic Brain Injury are mediated by infiltrating myeloid cells. J Neurotrauma. 2019;36(7):1040–53.PubMedPubMedCentralCrossRef
62.
go back to reference Bardehle S, Rafalski VA, Akassoglou K. Breaking boundaries-coagulation and fibrinolysis at the neurovascular interface. Front Cell Neurosci. 2015;9:354.PubMedPubMedCentralCrossRef Bardehle S, Rafalski VA, Akassoglou K. Breaking boundaries-coagulation and fibrinolysis at the neurovascular interface. Front Cell Neurosci. 2015;9:354.PubMedPubMedCentralCrossRef
63.
go back to reference Draxler DF, et al. Tranexamic acid modulates the cellular immune profile after traumatic brain injury in mice without hyperfibrinolysis. J Thromb Haemost. 2019;17(12):2174–87.PubMedCrossRef Draxler DF, et al. Tranexamic acid modulates the cellular immune profile after traumatic brain injury in mice without hyperfibrinolysis. J Thromb Haemost. 2019;17(12):2174–87.PubMedCrossRef
64.
go back to reference Duque P, et al. Pathophysiology of Trauma-Induced Coagulopathy. Transfus Med Rev. 2021;35(4):80–6.PubMedCrossRef Duque P, et al. Pathophysiology of Trauma-Induced Coagulopathy. Transfus Med Rev. 2021;35(4):80–6.PubMedCrossRef
Metadata
Title
Fibrin promotes oxidative stress and neuronal loss in traumatic brain injury via innate immune activation
Authors
Terry Dean
Andrew S. Mendiola
Zhaoqi Yan
Rosa Meza-Acevedo
Belinda Cabriga
Katerina Akassoglou
Jae Kyu Ryu
Publication date
01-12-2024
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2024
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-024-03092-w

Other articles of this Issue 1/2024

Journal of Neuroinflammation 1/2024 Go to the issue