Skip to main content

Advertisement

Log in

Perlecan Improves Blood Spinal Cord Barrier Repair Through the Integrin β1/ROCK/MLC Pathway After Spinal Cord Injury

  • Published:
Molecular Neurobiology Aims and scope Submit manuscript

Abstract

Spinal cord injury (SCI) can lead to the destruction of the blood-spinal cord barrier (BSCB), causing various inflammatory cytokines, neutrophils, and macrophages to infiltrate the lesion area, resulting in secondary injury. Basement membranes (BMs) are maintained by all types of cells in the BSCB and contribute to BSCB maintenance. Perlecan is an important constituent of vascular BMs, maintaining vascular integrity and neuroprotection. However, it is not clear whether Perlecan is involved in BSCB repair after SCI. In this study, we found that Perlecan was specifically expressed in the BMs in the spinal cord and underwent degradation/remodeling after SCI. Subsequently, a CRISPR/Cas9-based SAM system was used to overexpress Perlecan in the injured spinal cord, resulting in significantly enhanced locomotor recovery and neural regeneration. Overexpression of Perlecan reduced BSCB permeability along with the neuroinflammatory response. Interestingly, Perlecan inhibited stress fiber formation by interacting with integrin β1 and inhibiting downstream ROCK/MLC signaling, resulting in reduced tight junctions (TJs) disassembly and improved BSCB integrity. Furthermore, the integrin receptor antagonist GRGDSP abolished the effects of Perlecan overexpression on BSCB permeability and TJs integrity. Overall, our findings suggest that Perlecan reduces BSCB permeability and the neuroinflammatory response by interacting with integrin β1 and inhibiting the downstream ROCK/MLC pathway to promote neurological recovery after SCI.

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

Data Availability

The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.

References

  1. Xue W et al (2020) Epidermal growth factor receptor-extracellular-regulated kinase blockade upregulates TRIM32 signaling cascade and promotes neurogenesis after spinal cord injury. Stem Cells 38(1):118–133

    Article  CAS  Google Scholar 

  2. Kumar H et al (2017) Propitious therapeutic modulators to prevent blood-spinal cord barrier disruption in spinal cord injury. Mol Neurobiol 54(5):3578–3590

    Article  CAS  Google Scholar 

  3. Sofroniew MV (2018) Dissecting spinal cord regeneration. Nature 557(7705):343–350

    Article  CAS  Google Scholar 

  4. Hu J et al (2016) Targeting the blood-spinal cord barrier: a therapeutic approach to spinal cord protection against ischemia-reperfusion injury. Life Sci 158:1–6

    Article  CAS  Google Scholar 

  5. Nakamura K et al (2019) Perlecan regulates pericyte dynamics in the maintenance and repair of the blood-brain barrier. J Cell Biol 218(10):3506–3525

    Article  CAS  Google Scholar 

  6. Thomsen MS, Routhe LJ, Moos T (2017) The vascular basement membrane in the healthy and pathological brain. J Cereb Blood Flow Metab 37(10):3300–3317

    Article  Google Scholar 

  7. Takigawa T et al (2010) Separation of the perivascular basement membrane provides a conduit for inflammatory cells in a mouse spinal cord injury model. J Neurotrauma 27(4):739–751

    Article  Google Scholar 

  8. Roberts J, Kahle MP, Bix GJ (2012) Perlecan and the blood-brain barrier: beneficial proteolysis? Front Pharmacol 3:155

    Article  Google Scholar 

  9. Fukuda S et al (2004) Focal cerebral ischemia induces active proteases that degrade microvascular matrix. Stroke 35(4):998–1004

    Article  CAS  Google Scholar 

  10. Kerever A et al (2014) Perlecan is required for FGF-2 signaling in the neural stem cell niche. Stem Cell Res 12(2):492–505

    Article  CAS  Google Scholar 

  11. Martinez JR, Dhawan A, Farach-Carson MC (2018) Modular proteoglycan Perlecan/HSPG2: mutations, phenotypes, and functions. Genes (Basel) 9(11):556

    Article  Google Scholar 

  12. Xu L, Nirwane A, Yao Y (2019) Basement membrane and blood-brain barrier. Stroke Vasc Neurol 4(2):78–82

    Article  Google Scholar 

  13. Lee B et al (2011) Perlecan domain V is neuroprotective and proangiogenic following ischemic stroke in rodents. J Clin Invest 121(8):3005–3023

    Article  CAS  Google Scholar 

  14. Kahle MP et al (2012) Perlecan domain V is upregulated in human brain arteriovenous malformation and could mediate the vascular endothelial growth factor effect in lesional tissue. NeuroReport 23(10):627–630

    Article  CAS  Google Scholar 

  15. Trout AL et al (2021) Perlecan domain-V enhances neurogenic brain repair after stroke in mice. Transl Stroke Res 12(1):72–86

    Article  Google Scholar 

  16. van Horssen J et al (2001) Heparan sulfate proteoglycan expression in cerebrovascular amyloid beta deposits in Alzheimer’s disease and hereditary cerebral hemorrhage with amyloidosis (Dutch) brains. Acta Neuropathol 102(6):604–614

    Article  Google Scholar 

  17. Marques SA et al (2009) A simple, inexpensive and easily reproducible model of spinal cord injury in mice: morphological and functional assessment. J Neurosci Methods 177(1):183–193

    Article  Google Scholar 

  18. Xie C et al (2020) Astrocytic YAP promotes the formation of glia scars and neural regeneration after spinal cord injury. J Neurosci 40(13):2644–2662

    Article  CAS  Google Scholar 

  19. Joshi M, Fehlings MG (2002) Development and characterization of a novel, graded model of clip compressive spinal cord injury in the mouse: Part 1. Clip design, behavioral outcomes, and histopathology. J Neurotrauma 19(2):175–90

    Article  Google Scholar 

  20. Yang L et al (2021) Low expression of TRAF3IP2-AS1 promotes progression of NONO-TFE3 translocation renal cell carcinoma by stimulating N(6)-methyladenosine of PARP1 mRNA and downregulating PTEN. J Hematol Oncol 14(1):46

    Article  CAS  Google Scholar 

  21. Konermann S et al (2015) Genome-scale transcriptional activation by an engineered CRISPR-Cas9 complex. Nature 517(7536):583–588

    Article  CAS  Google Scholar 

  22. Wu J et al (2016) Recombinant osteopontin stabilizes smooth muscle cell phenotype via integrin receptor/integrin-linked kinase/Rac-1 pathway after subarachnoid hemorrhage in rats. Stroke 47(5):1319–1327

    Article  CAS  Google Scholar 

  23. Suzuki H et al (2010) Mechanisms of osteopontin-induced stabilization of blood-brain barrier disruption after subarachnoid hemorrhage in rats. Stroke 41(8):1783–1790

    Article  CAS  Google Scholar 

  24. Gu Y et al (2019) Conditional ablation of reactive astrocytes to dissect their roles in spinal cord injury and repair. Brain Behav Immun 80:394–405

    Article  Google Scholar 

  25. Demjen D et al (2004) Neutralization of CD95 ligand promotes regeneration and functional recovery after spinal cord injury. Nat Med 10(4):389–395

    Article  CAS  Google Scholar 

  26. Xu N et al (2015) A sensitive and reliable test instrument to assess swimming in rats with spinal cord injury. Behav Brain Res 291:172–183

    Article  Google Scholar 

  27. Ma M et al (2001) Behavioral and histological outcomes following graded spinal cord contusion injury in the C57Bl/6 mouse. Exp Neurol 169(2):239–254

    Article  CAS  Google Scholar 

  28. Hill RL et al (2009) Anatomical and functional outcomes following a precise, graded, dorsal laceration spinal cord injury in C57BL/6 mice. J Neurotrauma 26(1):1–15

    Article  Google Scholar 

  29. Ge X et al (2021) Exosomal miR-155 from M1-polarized macrophages promotes EndoMT and impairs mitochondrial function via activating NF-κB signaling pathway in vascular endothelial cells after traumatic spinal cord injury. Redox Biol 41:101932

    Article  CAS  Google Scholar 

  30. Joshi HP et al (2020) CORM-2-solid lipid nanoparticles maintain integrity of blood-spinal cord barrier after spinal cord injury in rats. Mol Neurobiol 57(6):2671–2689

    Article  CAS  Google Scholar 

  31. Ying X et al (2020) Water treadmill training attenuates blood-spinal cord barrier disruption in rats by promoting angiogenesis and inhibiting matrix metalloproteinase-2/9 expression following spinal cord injury. Fluids Barriers CNS 17(1):70

    Article  CAS  Google Scholar 

  32. Xu J et al (2017) Ultrastructural features of neurovascular units in a rat model of chronic compressive spinal cord injury. Front Neuroanat 11:136

    Article  Google Scholar 

  33. Freria CM et al (2017) Deletion of the fractalkine receptor, CX3CR1, improves endogenous repair, axon sprouting, and synaptogenesis after spinal cord injury in mice. J Neurosci 37(13):3568–3587

    Article  CAS  Google Scholar 

  34. Yao Y (2019) Basement membrane and stroke. J Cereb Blood Flow Metab 39(1):3–19

    Article  CAS  Google Scholar 

  35. Kaneko S et al (2006) A selective Sema3A inhibitor enhances regenerative responses and functional recovery of the injured spinal cord. Nat Med 12(12):1380–1389

    Article  CAS  Google Scholar 

  36. Ren ZW et al (2019) Effect of exosomes derived from MiR-133b-modified ADSCs on the recovery of neurological function after SCI. Eur Rev Med Pharmacol Sci 23(1):52–60

    Google Scholar 

  37. Orr MB, Gensel JC (2018) Spinal cord injury scarring and inflammation: therapies targeting glial and inflammatory responses. Neurotherapeutics 15(3):541–553

    Article  CAS  Google Scholar 

  38. Bartanusz V et al (2011) The blood-spinal cord barrier: morphology and clinical implications. Ann Neurol 70(2):194–206

    Article  Google Scholar 

  39. Reinhold AK, Rittner HL (2017) Barrier function in the peripheral and central nervous system-a review. Pflugers Arch 469(1):123–134

    Article  CAS  Google Scholar 

  40. Nag S et al (2019) Increased Expression of vascular endothelial growth factor-D following brain injury. Int J Mol Sci 20(7):1594

    Article  CAS  Google Scholar 

  41. Nourhaghighi N et al (2003) Altered expression of angiopoietins during blood-brain barrier breakdown and angiogenesis. Lab Invest 83(8):1211–1222

    Article  CAS  Google Scholar 

  42. Prasain N, Stevens T (2009) The actin cytoskeleton in endothelial cell phenotypes. Microvasc Res 77(1):53–63

    Article  CAS  Google Scholar 

  43. Shi Y et al (2016) Rapid endothelial cytoskeletal reorganization enables early blood-brain barrier disruption and long-term ischaemic reperfusion brain injury. Nat Commun 7:10523

    Article  CAS  Google Scholar 

  44. dos Remedios CG et al (2003) Actin binding proteins: regulation of cytoskeletal microfilaments. Physiol Rev 83(2):433–473

    Article  Google Scholar 

  45. Baeten KM, Akassoglou K (2011) Extracellular matrix and matrix receptors in blood-brain barrier formation and stroke. Dev Neurobiol 71(11):1018–1039

    Article  CAS  Google Scholar 

  46. Engelhardt B (2011) β1-integrin/matrix interactions support blood-brain barrier integrity. J Cereb Blood Flow Metab 31(10):1969–1971

    Article  CAS  Google Scholar 

  47. Izawa Y et al (2018) β1-integrin-matrix interactions modulate cerebral microvessel endothelial cell tight junction expression and permeability. J Cereb Blood Flow Metab 38(4):641–658

    Article  CAS  Google Scholar 

  48. Deguchi Y et al (2002) Internalization of basic fibroblast growth factor at the mouse blood-brain barrier involves perlecan, a heparan sulfate proteoglycan. J Neurochem 83(2):381–389

    Article  CAS  Google Scholar 

  49. Miller JD et al (1997) Localization of perlecan (or a perlecan-related macromolecule) to isolated microglia in vitro and to microglia/macrophages following infusion of beta-amyloid protein into rodent hippocampus. Glia 21(2):228–243

    Article  CAS  Google Scholar 

  50. Grindel BJ et al (2014) Matrilysin/matrix metalloproteinase-7(MMP7) cleavage of perlecan/HSPG2 creates a molecular switch to alter prostate cancer cell behavior. Matrix Biol 36:64–76

    Article  CAS  Google Scholar 

  51. Bix GJ, Gowing EK, Clarkson AN (2013) Perlecan domain V is neuroprotective and affords functional improvement in a photothrombotic stroke model in young and aged mice. Transl Stroke Res 4(5):515–523

    Article  CAS  Google Scholar 

  52. Marcelo A, Bix G (2015) The potential role of perlecan domain V as novel therapy in vascular dementia. Metab Brain Dis 30(1):1–5

    Article  CAS  Google Scholar 

  53. Daley WP, Peters SB, Larsen M (2008) Extracellular matrix dynamics in development and regenerative medicine. J Cell Sci 121(Pt 3):255–264

    Article  CAS  Google Scholar 

  54. Kyprianou C et al (2020) Basement membrane remodelling regulates mouse embryogenesis. Nature 582(7811):253–258

    Article  CAS  Google Scholar 

  55. Gasche Y et al (2006) Matrix metalloproteinases and diseases of the central nervous system with a special emphasis on ischemic brain. Front Biosci 11:1289–1301

    Article  CAS  Google Scholar 

  56. Rust R, Kaiser J (2017) Insights into the dual role of inflammation after spinal cord injury. J Neurosci 37(18):4658–4660

    Article  CAS  Google Scholar 

  57. Liu WY et al (2015) Increasing the permeability of the blood-brain barrier in three different models in vivo. CNS Neurosci Ther 21(7):568–574

    Article  CAS  Google Scholar 

  58. Dejana E, Tournier-Lasserve E, Weinstein BM (2009) The control of vascular integrity by endothelial cell junctions: molecular basis and pathological implications. Dev Cell 16(2):209–221

    Article  CAS  Google Scholar 

  59. Nieuwenhuis B et al (2018) Integrins promote axonal regeneration after injury of the nervous system. Biol Rev Camb Philos Soc 93(3):1339–1362

    Article  Google Scholar 

  60. Tang J et al (2020) TIMP1 preserves the blood-brain barrier through interacting with CD63/integrin β 1 complex and regulating downstream FAK/RhoA signaling. Acta Pharm Sin B 10(6):987–1003

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank Home for Researchers editorial team (www.home-for-researchers.com) for language editing service.

Funding

This work is supported by the Natural Science Foundation of Guangdong Province (2017A030312009, 2017A030313111), the National Natural Science Foundation (82071386, 81974329, 81672140), Key Research & Development Program of Guangzhou Regenerative Medicine and Health Guangdong Laboratory (2018GZR110104008), and Research Grant of Guangdong Province Key Laboratory of Psychiatric Disorders (N201904).

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to the study conception and design. Lixin Zhu and Jiasong Guo designed the study. Material preparation, data collection and analysis were performed by Changnan Xie, Yihan Wang, Jinfeng Wang, Yizhou Xu and Haining Liu. The first draft of the manuscript was written by Changnan Xie and all authors commented on previous versions of the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Jiasong Guo or Lixin Zhu.

Ethics declarations

Ethics Approval

All animal experiments were performed in accordance with Institutional Animal Care and Use committee guidelines and approved protocols at the Southern Medical University (ethics Number: LAEC-2022–064).

Consent to Participate

Not applicable.

Consent for Publication

All data generated or analyzed during this study are included in this published article.

Conflict of Interest

The authors declare no competing interests.

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.

Supplemental Fig. 1

The expression effect of Perlecan by SAM system in injured spinal cords A and B Real time PCR analysis showed the relative mRNA level of Perlecan in injured spinal cords from SCI, LV-GFP and LV-Perlecan groups at 7 and 14 days after SCI (n = 3 animal per group). C Immunostaining of Perlecan in spinal cords from SCI, LV-GFP and LV-Perlecan groups at 14 days after SCI. White dashed lines indicated the injury sites. LC indicated lesion core. Quantitative data in (A and B) were analyzed using one-way ANOVA with Dunnett’s multiple comparison test. **p<0.01, compared with LV-Perlecan group. Data were mean ± SEM. Scale bars, 20 μm (PNG 3.26 MB)

Supplementary file1 (TIF 7152 KB)

Rights and permissions

Springer Nature or its licensor holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xie, C., Wang, Y., Wang, J. et al. Perlecan Improves Blood Spinal Cord Barrier Repair Through the Integrin β1/ROCK/MLC Pathway After Spinal Cord Injury. Mol Neurobiol 60, 51–67 (2023). https://doi.org/10.1007/s12035-022-03041-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12035-022-03041-9

Keywords

Navigation