Skip to main content
Log in

PAI-1 production by reactive astrocytes drives tissue dysfibrinolysis in multiple sclerosis models

  • Original Article
  • Published:
Cellular and Molecular Life Sciences Aims and scope Submit manuscript

Abstract

Background

In multiple sclerosis (MS), disturbance of the plasminogen activation system (PAS) and blood brain barrier (BBB) disruption are physiopathological processes that might lead to an abnormal fibrin(ogen) extravasation into the parenchyma. Fibrin(ogen) deposits, usually degraded by the PAS, promote an autoimmune response and subsequent demyelination. However, the PAS disruption is not well understood and not fully characterized in this disorder.

Methods

Here, we characterized the expression of PAS actors during different stages of two mouse models of MS (experimental autoimmune encephalomyelitis—EAE), in the central nervous system (CNS) by quantitative RT-PCR, immunohistofluorescence and fluorescent in situ hybridization (FISH). Thanks to constitutive PAI-1 knockout mice (PAI-1 KO) and an immunotherapy using a blocking PAI-1 antibody, we evaluated the role of PAI-1 in EAE models and its impact on physiopathological processes such as fibrin(ogen) deposits, lymphocyte infiltration and demyelination.

Results

We report a striking overexpression of PAI-1 in reactive astrocytes during symptomatic phases, in two EAE mouse models of MS. This increase is concomitant with lymphocyte infiltration and fibrin(ogen) deposits in CNS parenchyma. By genetic invalidation of PAI-1 in mice and immunotherapy using a blocking PAI-1 antibody, we demonstrate that abolition of PAI-1 reduces the severity of EAE and occurrence of relapses in two EAE models. These benefits are correlated with a decrease in fibrin(ogen) deposits, infiltration of T4 lymphocytes, reactive astrogliosis, demyelination and axonal damage.

Conclusion

These results demonstrate that a deleterious overexpression of PAI-1 by reactive astrocytes leads to intra-parenchymal dysfibrinolysis in MS models and anti-PAI-1 strategies could be a new therapeutic perspective for MS.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8

Similar content being viewed by others

Availability of data and materials

The datasets of the current study are available from the corresponding author on reasonable request.

Abbreviations

BBB:

Blood brain barrier

CNS:

Central nervous system

CTCF:

Corrected total cell fluorescence

EAE:

Experimental autoimmune encephalomyelitis

KO:

Knockout

MS:

Multiple sclerosis

NSP:

Neuroserpin

PAI-1:

Type-1 plasminogen activator inhibitor

PAS:

Plasminogen activation system

PLG:

Plasminogen

PN-1:

Protease nexin 1

TAFI:

Thrombin-activatable fibrinolysis inhibitor

tPA:

Tissue-type plasminogen

WT:

Wild type

References

  1. Petersen MA, Ryu JK, Akassoglou K (2018) Fibrinogen in neurological diseases: mechanisms, imaging and therapeutics. Nat Rev Neurosci 19(5):283–301

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Gveric D, Hanemaaijer R, Newcombe J, van Lent NA, Sier CF, Cuzner ML (2001) Plasminogen activators in multiple sclerosis lesions: implications for the inflammatory response and axonal damage. Brain 124(Pt 10):1978–1988

    Article  CAS  PubMed  Google Scholar 

  3. Gveric D, Herrera B, Petzold A, Lawrence DA, Cuzner ML (2003) Impaired fibrinolysis in multiple sclerosis: a role for tissue plasminogen activator inhibitors. Brain 126(Pt 7):1590–1598

    Article  PubMed  Google Scholar 

  4. Ryu JK, Petersen MA, Murray SG, Baeten KM, Meyer-Franke A, Chan JP et al (2015) Blood coagulation protein fibrinogen promotes autoimmunity and demyelination via chemokine release and antigen presentation. Nat Commun 6:8164

    Article  PubMed  Google Scholar 

  5. Docagne F, Nicole O, Marti HH, MacKenzie ET, Buisson A, Vivien D (1999) Transforming growth factor-beta1 as a regulator of the serpins/t-PA axis in cerebral ischemia. FASEB J 13(11):1315–1324

    Article  CAS  PubMed  Google Scholar 

  6. Louessard M, Lacroix A, Martineau M, Mondielli G, Montagne A, Lesept F et al (2016) Tissue plasminogen activator expression is restricted to subsets of excitatory pyramidal glutamatergic neurons. Mol Neurobiol 53(7):5000–5012

    Article  CAS  PubMed  Google Scholar 

  7. Buisson A, Nicole O, Docagne F, Sartelet H, Mackenzie ET, Vivien D (1998) Up-regulation of a serine protease inhibitor in astrocytes mediates the neuroprotective activity of transforming growth factor beta1. FASEB J 12(15):1683–1691

    Article  CAS  PubMed  Google Scholar 

  8. Fernández-Monreal M, López-Atalaya JP, Benchenane K, Léveillé F, Cacquevel M, Plawinski L et al (2004) Is tissue-type plasminogen activator a neuromodulator? Mol Cell Neurosci 25(4):594–601

    Article  PubMed  CAS  Google Scholar 

  9. Cassé F, Bardou I, Danglot L, Briens A, Montagne A, Parcq J et al (2012) Glutamate controls tPA recycling by astrocytes, which in turn influences glutamatergic signals. J Neurosci 32(15):5186–5199

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Briens A, Bardou I, Lebas H, Miles LA, Parmer RJ, Vivien D et al (2017) Astrocytes regulate the balance between plasminogen activation and plasmin clearance via cell-surface actin. Cell Discov 3:17001

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Mehra A, Ali C, Parcq J, Vivien D, Docagne F (2016) The plasminogen activation system in neuroinflammation. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease 1862(3):395–402

    Article  CAS  Google Scholar 

  12. East E, Baker D, Pryce G, Lijnen HR, Cuzner ML, Gverić D (2005) A role for the plasminogen activator system in inflammation and neurodegeneration in the central nervous system during experimental allergic encephalomyelitis. Am J Pathol 167(2):545–554

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. East E, Gverić D, Baker D, Pryce G, Lijnen HR, Cuzner ML (2008) Chronic relapsing experimental allergic encephalomyelitis (CREAE) in plasminogen activator inhibitor-1 knockout mice: the effect of fibrinolysis during neuroinflammation. Neuropathol Appl Neurobiol 34(2):216–230

    Article  CAS  PubMed  Google Scholar 

  14. Davalos D, Ryu JK, Merlini M, Baeten KM, Le Moan N, Petersen MA et al (2012) Fibrinogen-induced perivascular microglial clustering is required for the development of axonal damage in neuroinflammation. Nat Commun 3:1227

    Article  PubMed  CAS  Google Scholar 

  15. Yates RL, Esiri MM, Palace J, Jacobs B, Perera R, DeLuca GC (2017) Fibrin(ogen) and neurodegeneration in the progressive multiple sclerosis cortex: Fibrin(ogen) and Neurodegeneration in MS Cortex. Ann Neurol août 82(2):259–270

    Article  CAS  Google Scholar 

  16. Akenami FO, Koskiniemi M, Färkkilä M, Vaheri A (1997) Cerebrospinal fluid plasminogen activator inhibitor-1 in patients with neurological disease. J Clin Pathol 50(2):157–160

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Onodera H, Nakashima I, Fujihara K, Nagata T, Itoyama Y (1999) Elevated plasma level of plasminogen activator inhibitor-1 (PAI-1) in patients with relapsing–remitting multiple sclerosis. Tohoku J Exp Med 189(4):259–265

    Article  CAS  PubMed  Google Scholar 

  18. Pérez-Martín MY, González-Platas M, Jiménez-Sosa A, Plata-Bello J, Carrillo-Padilla FJ, Franco-Maside A et al (2017) Can fibrinolytic system components explain cognitive impairment in multiple sclerosis? J Neurol Sci 382:66–72

    Article  PubMed  CAS  Google Scholar 

  19. Lundgaard I, Osório MJ, Kress BT, Sanggaard S, Nedergaard M (2014) White matter astrocytes in health and disease. Neuroscience 276:161–173

    Article  CAS  PubMed  Google Scholar 

  20. Fournier AP, Gauberti M, Quenault A, Vivien D, Macrez R, Docagne F (2019) Reduced spinal cord parenchymal cerebrospinal fluid circulation in experimental autoimmune encephalomyelitis. J Cereb Blood Flow Metab 39(7):1258–1265

    Article  PubMed  Google Scholar 

  21. Bruckert G, Vivien D, Docagne F, Roussel BD (2016) Normalization of reverse transcription quantitative PCR data during ageing in distinct cerebral structures. Mol Neurobiol 53(3):1540–1550

    Article  CAS  PubMed  Google Scholar 

  22. Watson C, Paxinos G, Kayalioglu G, Heise C (2009) Chapter 16 - Atlas of the Mouse Spinal Cord. In: Watson C, Paxinos G, Kayalioglu G (eds) The Spinal Cord [Internet]. Academic Press, San Diego, pp 308–379

    Chapter  Google Scholar 

  23. Fournier AP, Quenault A, Martinez de Lizarrondo S, Gauberti M, Defer G, Vivien D et al (2017) Prediction of disease activity in models of multiple sclerosis by molecular magnetic resonance imaging of P-selectin. Proc Natl Acad Sci USA 114(23):6116–6121

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Liddelow SA, Barres BA (2017) Reactive astrocytes: production, function, and therapeutic potential. Immunity 46(6):957–967

    Article  CAS  PubMed  Google Scholar 

  25. Van De Craen B, Scroyen I, Abdelnabi R, Brouwers E, Lijnen HR, Declerck PJ et al (2011) Characterization of a panel of monoclonal antibodies toward mouse PAI-1 that exert a significant profibrinolytic effect in vivo. Thromb Res 128(1):68–76

    Article  CAS  Google Scholar 

  26. Dietzmann K, Bossanyi Pv, Krause D, Wittig H, Mawrin Ch, Kirches E (2000) Expression of the plasminogen activator system and the inhibitors PAI-1 and PAI-2 in posttraumatic lesions of the CNS and brain injuries following dramatic circulatory arrests: an immunohistochemical study. Pathol Res Practice 196(1):15–21

    Article  CAS  Google Scholar 

  27. Liu R-M, van Groen T, Katre A, Cao D, Kadisha I, Ballinger C et al (2011) Knockout of plasminogen activator inhibitor 1 gene reduces amyloid beta peptide burden in a mouse model of Alzheimer’s disease. Neurobiol Aging 32(6):1079–1089

    Article  CAS  PubMed  Google Scholar 

  28. Boche D, Cunningham C, Docagne F, Scott H, Perry VH (2006) TGFβ1 regulates the inflammatory response during chronic neurodegeneration. Neurobiol Dis 22(3):638–650

    Article  CAS  PubMed  Google Scholar 

  29. Kroksveen AC, Opsahl JA, Guldbrandsen A, Myhr K-M, Oveland E, Torkildsen Ø et al (2015) Cerebrospinal fluid proteomics in multiple sclerosis. Biochim Biophys Acta 1854(7):746–756

    Article  CAS  PubMed  Google Scholar 

  30. Emeis JJ, Hoekzema R, de Vos AF (1995) Inhibiting interleukin-1 and tumor necrosis factor-alpha does not reduce induction of plasminogen activator inhibitor type-1 by endotoxin in rats in vivo. Blood 85(1):115–120

    Article  CAS  PubMed  Google Scholar 

  31. Zheng Z, Nakamura K, Gershbaum S, Wang X, Thomas S, Bessler M et al (2020) Interacting hepatic PAI-1/tPA gene regulatory pathways influence impaired fibrinolysis severity in obesity. J Clin Invest 130(8):4348–4359

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Lenoir S, Varangot A, Lebouvier L, Galli T, Hommet Y, Vivien D (2019) Post-synaptic release of the neuronal tissue-type plasminogen activator (tPA). Front Cell Neurosci 13:164

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Hultman K, Blomstrand F, Nilsson M, Wilhelmsson U, Malmgren K, Pekny M et al (2010) Expression of plasminogen activator inhibitor-1 and protease nexin-1 in human astrocytes: Response to injury-related factors. J Neurosci Res 88(11):2441–2449

    CAS  PubMed  Google Scholar 

  34. Ko HM, Lee SH, Kim KC, Joo SH, Choi WS, Shin CY (2014) The role of TLR4 and Fyn interaction on lipopolysaccharide-stimulated PAI-1 expression in astrocytes. Mol Neurobiol 52(1):8–25

    Article  PubMed  CAS  Google Scholar 

  35. Escartin C, Galea E, Lakatos A, O’Callaghan JP, Petzold GC, Serrano-Pozo A et al (2021) Reactive astrocyte nomenclature, definitions, and future directions. Nat Neurosci 24(3):312–325

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Groves A, Kihara Y, Jonnalagadda D, Rivera R, Kennedy G, Mayford M et al (2018) A functionally defined in vivo astrocyte population identified by c-Fos activation in a mouse model of multiple sclerosis modulated by S1P signaling: immediate-early astrocytes (ieAstrocytes). eNeuro 5(5):ENEURO.0239-18.2018

    Article  PubMed  PubMed Central  Google Scholar 

  37. Itoh N, Itoh Y, Tassoni A, Ren E, Kaito M, Ohno A et al (2018) Cell-specific and region-specific transcriptomics in the multiple sclerosis model: focus on astrocytes. Proc Natl Acad Sci USA 115(2):E302–E309

    Article  CAS  PubMed  Google Scholar 

  38. Borggrewe M, Grit C, Vainchtein ID, Brouwer N, Wesseling EM, Laman JD et al (2021) Regionally diverse astrocyte subtypes and their heterogeneous response to EAE. Glia 69(5):1140–1154

    Article  CAS  PubMed  Google Scholar 

  39. Ryu JK, Rafalski VA, Meyer-Franke A, Adams RA, Poda SB, Rios Coronado PE et al (2018) Fibrin-targeting immunotherapy protects against neuroinflammation and neurodegeneration. Nat Immunol 19(11):1212–1223

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Baumeier C, Escher F, Aleshcheva G, Pietsch H, Schultheiss H-P (2021) Plasminogen activator inhibitor-1 reduces cardiac fibrosis and promotes M2 macrophage polarization in inflammatory cardiomyopathy. Basic Res Cardiol 116(1):1

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Kubala MH, Punj V, Placencio-Hickok VR, Fang H, Fernandez GE, Sposto R et al (2018) Plasminogen activator inhibitor-1 promotes the recruitment and polarization of macrophages in cancer. Cell Rep 25(8):2177-2191.e7

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Poggi M, Paulmyer-Lacroix O, Verdier M, Peiretti F, Bastelica D, Boucraut J et al (2007) Chronic plasminogen activator inhibitor-1 (PAI-1) overexpression dampens CD25+ lymphocyte recruitment after lipopolysaccharide endotoxemia in mouse lung. J Thromb Haemost 5(12):2467–2475

    Article  CAS  PubMed  Google Scholar 

  43. Thapa B, Kim YH, Kwon H-J, Kim D-S (2014) The LRP1-independent mechanism of PAI-1-induced migration in CpG-ODN activated macrophages. Int J Biochem Cell Biol 49:17–25

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This project was supported by grants from the French Ministry of Higher Education, the INSERM (French National Institute for Health and Medical Research), the regional Council of Normandy, the ARSEP foundation and the FRM (Fondation pour la recherche médicale). We thank Dr Paul Declerck for the gift of PAI-1 blocking antibody.

Funding

This project was supported by grants from the French Ministry of Higher Education, the INSERM (French National Institute for Health and Medical Research), the regional Council of Normandy, the ARSEP foundation and the FRM (Fondation pour la recherche médicale).

Author information

Authors and Affiliations

Authors

Contributions

HL designed the experiments, performed in vitro and in vivo studies, analyzed data, and participated in manuscript preparation. SG analyzed data and participated in manuscript preparation. AP performed reactive astrogliosis and axonal damage experiments and analyzed data. AF generated P-sel+ and P-sel tissues. A-CB set up and performed FISH study. DV drafted the manuscript. MC-S gave expertise in the design of FISH study and drafted the article. FD designed the study and wrote the article. IB designed the study, analyzed data and wrote the article. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Isabelle Bardou.

Ethics declarations

Conflict of interest

The authors declare that they have no competing interests.

Ethics approval

Animal experiments were performed in accordance with French ethical laws (Decree 87/848) and European guidelines (Directive 2010/63/UE) for the care and use of laboratory animals. Experiments were undertaken in the housing and laboratories (approval #F14118001) and have been approved by the ethics committee no. 52 on animal experiments (CENOMEXA) and by the French Ministry of Research under the project license number #10799. All experiments were performed following the ARRIVE guidelines (www.nc3rs.org.uk), including randomization of treatment as well as analysis blind to the treatment.

Consent for publication

Not applicable.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Below is the link to the electronic supplementary material.

18_2022_4340_MOESM1_ESM.pdf

Supplementary file1 Additional file 1: Figure S1. PAS gene expression in forebrain, cerebellum and spinal cord during the course of MOG-induced EAE. Gene expression of a-c tPA, d-f NSP, g-i PN-1, j TAFI, k, l PAI-1 and m-o PLG in a, d, g, j, m the spinal cord, b, e, h, k, n the cerebellum and c, f, i, l, o the forebrain of mice during the course of MOG-induced EAE. RT-qPCR results expressed as mean ± SEM, n=5 per condition. Kruskal-Wallis test followed by FDR post-hoc test: * and § indicate significant difference (p < 0.05) compared to Sham and Pre-On respectively, Δ indicates other significant differences (p < 0.05) between specific stages. tPA: tissue-type plasminogen activator; NSP: Neuroserpin; PN-1: protease nexin-1; PLG: Plasminogen; TAFI: thrombin-activatable fibrinolysis inhibitor; PAI-1: type-1 plasminogen activator inhibitor. Pre-On: pre-onset; On: onset; Su: surge; Ch: chronic as depicted in Fig. 1a. (PDF 235 KB)

18_2022_4340_MOESM2_ESM.pdf

Supplementary file2 Additional file 2: Figure S2.PAS gene expression in forebrain, cerebellum and spinal cord during the course of PLP-induced EAE. Gene expression of a-c tPA, d-f NSP, g-i PN-1, j TAFI, k, l PAI-1 and m, n PLG in a, d, g, j, m the spinal cord, b, e, h, k, n the cerebellum and c, f, i, l the forebrain of mice during the course of PLP-induced EAE. RT-qPCR results expressed as mean ± SEM, n=5 per condition. Kruskal-Wallis test followed by FDR post-hoc test: *, §, #, † indicate significant difference (p < 0.05) compared to Sham, Pre-On, Rem and Rel respectively, Δ indicates other significant differences (p < 0.05) between specific stages. tPA: tissue-type plasminogen activator; NSP: Neuroserpin; PN-1: protease nexin-1; PLG: Plasminogen; TAFI: thrombin-activatable fibrinolysis inhibitor; PAI-1: type-1 plasminogen activator inhibitor. Pre-On: pre-onset; On: onset; Sc1/2/3/4/2R: score 1/2/3/4/2 remitting; Rem: remission; Rel: relapse as depicted in Fig. 1d. (PDF 295 KB)

18_2022_4340_MOESM3_ESM.pdf

Supplementary file3 Additional file 3: Figure S3: PAI-1 immunostaining quantification in white matter spinal cord sections during the course of MOG-induced EAE model. a Representative PAI-1 immunostaining (red) in cervical, high thoracic, low thoracic and lumbar sections of spinal cord from Sham and MOG-induced EAE mice for each stage of the disease (DAPI: blue). b Quantification of PAI-1 immunostaining coverage in cervical, high thoracic, low thoracic and lumbar sections of spinal cord from Sham and MOG-induced EAE mice. Histograms show mean ± SEM, n=3 per condition. Kruskal-Wallis test followed by FDR post-hoc test: * indicates significant difference (p < 0.05) compared to Sham, Δ indicates other significant differences (p < 0.05) between specific stages. Sc1/2/3/4: score 1/2/3/4. Scale bars: 50µm. (PDF 496 KB)

18_2022_4340_MOESM4_ESM.pdf

Supplementary file4 Additional file 4: Figure S4: PAI-1 immunostaining quantification in white matter spinal cord sections during the course of PLP-induced EAE model. a Representative PAI-1 immunostaining (red) in cervical, high thoracic, low thoracic and lumbar sections of spinal cord from Sham and PLP-induced EAE mice for each stage of the disease (DAPI: blue). b PAI-1 immunostaining coverage in cervical, high thoracic, low thoracic and lumbar sections of spinal cord from Sham and PLP-induced EAE mice. Histograms show mean ± SEM, n=3 per condition. Kruskal-Wallis test followed by FDR post-hoc test: *, §, #, † indicate significant difference (p < 0.05) compared to Sham, Pre-on, Rem and Rel respectively, Δ indicates other significant differences (p < 0.05) between specific stages. Sc1/2/3/4/2R: score1/2/3/4/2 remitting; Rem: remission; Rel: relapse. Scale bars: 50µm. (PDF 672 KB)

18_2022_4340_MOESM5_ESM.pdf

Supplementary file5 Additional file 5: Figure S5: SMI32 immunostaining quantification in white matter spinal cord sections during the course of EAE models. a Representative SMI-32 immunostaining (red) in cervical spinal cord from PAI-1 WT and PAI-1 KO mice at onset (14 dpi), peak (18 dpi) and late stages (45 dpi) during MOG-induced EAE (DAPI: blue). b Corresponding quantifications of axonal damage in cervical spinal cord from PAI-1 WT and PAI-1 KO mice. c Representative SMI32 immunostaining (red) in spinal cord from control and anti-PAI-1 antibody-treated mice at acute (16 dpi), remission (28 dpi) and late stages (45 dpi) during PLP-induced EAE (DAPI: blue). d Corresponding quantifications of axonal damage in spinal cord from control and anti-PAI-1 antibody-treated mice. Histograms show mean ± SEM, n=3 per condition. Mann-Whitney U test; *p < 0.05. Scale bars: 40µm. (PDF 543 KB)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lebas, H., Guérit, S., Picot, A. et al. PAI-1 production by reactive astrocytes drives tissue dysfibrinolysis in multiple sclerosis models. Cell. Mol. Life Sci. 79, 323 (2022). https://doi.org/10.1007/s00018-022-04340-z

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s00018-022-04340-z

Keywords

Navigation