Skip to main content
Log in

Cav-1 Protein Levels in Serum and Infarcted Brain Correlate with Hemorrhagic Volume in a Mouse Model of Thromboembolic Stroke, Independently of rt-PA Administration

  • Published:
Molecular Neurobiology Aims and scope Submit manuscript

Abstract

Thrombolytic therapy with recombinant tissue plasminogen activator (rt-PA) is currently the only FDA-approved drug for acute ischemic stroke. However, its administration is still limited due to the associated increased risk of hemorrhagic transformation (HT). rt-PA may exacerbate blood-brain barrier (BBB) injury by several mechanisms that have not been fully elucidated. Caveolin-1 (Cav-1), a major structural protein of caveolae, has been linked to the endothelial barrier function. The effects of rt-PA on Cav-1 expression remain largely unknown. Here, Cav-1 protein expression after ischemic conditions, with or without rt-PA administration, was analyzed in a murine thromboembolic middle cerebral artery occlusion (MCAO) and in brain microvascular endothelial bEnd.3 cells subjected to oxygen/glucose deprivation (OGD). Our results show that Cav-1 is overexpressed in endothelial cells of infarcted area and in bEnd.3 cell line after ischemia but there is disagreement regarding rt-PA effects on Cav-1 expression between both experimental models. Delayed rt-PA administration significantly reduced Cav-1 total levels from 24 to 72 h after reoxygenation and increased pCav-1/Cav-1 at 72 h in the bEnd.3 cells while it did not modify Cav-1 immunoreactivity in the infarcted area at 24 h post-MCAO. Importantly, tissue Cav-1 positively correlated with Cav-1 serum levels at 24 h post-MCAO and negatively correlated with the volume of hemorrhage after infarction, the latter supporting a protective role of Cav-1 in cerebral ischemia. In addition, the negative association between baseline serum Cav-1 levels and hemorrhagic volume points to a potential usefulness of baseline serum Cav-1 levels to predict hemorrhagic volume, independently of rt-PA administration.

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

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.

Abbreviations

BBB:

Blood-brain barrier

bEnd.3:

Immortalized mouse brain endothelial cell line

BSA:

Bovine serum albumin

Cav-1:

Caveolin-1

CTR:

Control

DAB:

Diaminobenzidine

FITC-BSA:

Fluorescein isothiocyanate labelled bovine serum albumin

HT:

Hemorrhagic transformation

MCAO:

Middle cerebral artery occlusion

MTT:

3-(4,5-Dimethylthiazol)-2,5-diphenyl tetrazolium bromide

OGD:

Oxygen/glucose deprivation

pCav-1:

Phosphorylated caveolin-1

rt-PA:

Recombinant tissue plasminogen activator

References

  1. Kim J, Thayabaranathan T, Donnan GA et al (2020) Global Stroke Statistics 2019. Int J Stroke 15:819–838. https://doi.org/10.1177/1747493020909545

    Article  PubMed  Google Scholar 

  2. Pandian JD, Gall SL, Kate MP et al (2018) Prevention of stroke: a global perspective. Lancet 392:1269–1278. https://doi.org/10.1016/S0140-6736(18)31269-8

    Article  PubMed  Google Scholar 

  3. Fusco R, Scuto M, Cordaro M et al (2019) N-palmitoylethanolamide-oxazoline protects against middle cerebral artery occlusion injury in diabetic rats by regulating the SIRT1 pathway. Int J Mol Sci 20:1–22. https://doi.org/10.3390/ijms20194845

    Article  CAS  Google Scholar 

  4. Powers WJ, Rabinstein AA, Ackerson T et al (2019) Guidelines for the early management of patients with acute ischemic stroke: 2019 update to the 2018 guidelines for the early management of acute ischemic stroke a guideline for healthcare professionals from the American Heart Association/American Stroke A

  5. Aguiar de Sousa D, von Martial R, Abilleira S et al (2019) Access to and delivery of acute ischaemic stroke treatments: a survey of national scientific societies and stroke experts in 44 European countries. Eur Stroke J 4:13–28. https://doi.org/10.1177/2396987318786023

    Article  PubMed  Google Scholar 

  6. Liu C, Xie J, Sun S et al (2020) Hemorrhagic transformation after tissue plasminogen activator treatment in acute ischemic stroke. Cell Mol Neurobiol. https://doi.org/10.1007/s10571-020-00985-1

    Article  PubMed  PubMed Central  Google Scholar 

  7. Fanne RA, Nassar T, Yarovoi S et al (2010) Blood-brain barrier permeability and tPA-mediated neurotoxicity. Neuropharmacology 58:972–980. https://doi.org/10.1016/j.neuropharm.2009.12.017

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Won SJ, Tang XN, Suh SW et al (2011) Hyperglycemia promotes tissue plasminogen activator-induced hemorrhage by increasing superoxide production. Ann Neurol 70:583–590. https://doi.org/10.1002/ana.22538

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Adibhatla R, Hatcher J (2008) Tissue plasminogen activator (tPA) and matrix metalloproteinases in the pathogenesis of stroke: therapeutic strategies. CNS Neurol Disord - Drug Targets 7:243–253. https://doi.org/10.2174/187152708784936608

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Zhao YL, Song JN, Zhang M (2014) Role of caveolin-1 in the biology of the blood-brain barrier. Rev Neurosci 25:247–254. https://doi.org/10.1515/revneuro-2013-0039

    Article  CAS  PubMed  Google Scholar 

  11. Zhou M, Shi SX, Liu N et al (2021) Caveolae-mediated endothelial transcytosis across the blood-brain barrier in acute ischemic stroke. J Clin Med. https://doi.org/10.3390/jcm10173795

    Article  PubMed  PubMed Central  Google Scholar 

  12. Gu Y, Zheng G, Xu M et al (2012) Caveolin-1 regulates nitric oxide-mediated matrix metalloproteinases activity and blood-brain barrier permeability in focal cerebral ischemia and reperfusion injury. J Neurochem 120:147–156. https://doi.org/10.1111/j.1471-4159.2011.07542.x

    Article  CAS  PubMed  Google Scholar 

  13. Jasmin J-FJF, Malhotra S, Singh Dhallu M et al (2007) Caveolin-1 deficiency increases cerebral ischemic injury. Circ Res 100:721–729. https://doi.org/10.1161/01.RES.0000260180.42709.29

    Article  CAS  PubMed  Google Scholar 

  14. Nag S, Venugopalan R, Stewart DJ (2007) Increased caveolin-1 expression precedes decreased expression of occludin and claudin-5 during blood-brain barrier breakdown. Acta Neuropathol 114:459–469. https://doi.org/10.1007/s00401-007-0274-x

    Article  CAS  PubMed  Google Scholar 

  15. Song H, Cheng Y, Bi G et al (2016) Release of matrix metalloproteinases-2 and 9 by S-nitrosylated caveolin-1 contributes to degradation of extracellular matrix in tPA-treated hypoxic endothelial cells. PLoS ONE 11:1–16. https://doi.org/10.1371/journal.pone.0149269

    Article  CAS  Google Scholar 

  16. Nag S, Manias JLL, Stewart DJJ (2009) Expression of endothelial phosphorylated caveolin-1 is increased in brain injury. Neuropathol Appl Neurobiol 35:417–426. https://doi.org/10.1111/j.1365-2990.2008.01009.x

    Article  CAS  PubMed  Google Scholar 

  17. García-Yébenes I, Sobrado M, Zarruk JGG et al (2011) A mouse model of hemorrhagic transformation by delayed tissue plasminogen activator administration after in situ thromboembolic stroke. Stroke 42:196–203. https://doi.org/10.1161/STROKEAHA.110.600452

    Article  CAS  PubMed  Google Scholar 

  18. Cyrille O, Audrey LB, Anne-Laure B et al (2007) Mouse model of in situ thromboembolic stroke and reperfusion. Stroke 38:2771–2778. https://doi.org/10.1007/978-1-4939-5620-3_6

    Article  Google Scholar 

  19. Arnberg F, Grafström J, Lundberg J et al (2015) Imaging of a clinically relevant stroke model glucose hypermetabolism revisited. Stroke 46:835–842. https://doi.org/10.1161/STROKEAHA.114.008407

    Article  CAS  PubMed  Google Scholar 

  20. Gubern C, Comajoan P, Huguet G et al (2020) Evaluation of long-term rt-PA effects on bEnd.3 endothelial cells under ischemic conditions; changes in ZO-1 expression and glycosylation of the bradykinin B2 receptor. Thromb Res 187. https://doi.org/10.1016/j.thromres.2019.12.021

  21. Brown RC, Morris AP, O’Neil RG (2007) Tight junction protein expression and barrier properties of immortalized mouse brain microvessel endothelial cells. Brain Res 1130:17–30. https://doi.org/10.1016/j.brainres.2006.10.083

    Article  CAS  PubMed  Google Scholar 

  22. Comajoan P, Gubern C, Huguet G et al (2018) Evaluation of common housekeeping proteins under ischemic conditions and/or rt-PA treatment in bEnd.3 cells. J Proteomics 184. https://doi.org/10.1016/j.jprot.2018.06.011

  23. Godfrey KRKR, Tanswell P, Bates RAR et al (1998) Nonlinear pharmacokinetics of tissue-type plasminogen activator in three animal species: a comparison of mathematical models. Biopharm Drug Dispos 19:131–140. https://doi.org/10.1002/(SICI)1099-081X(199803)19:2%3c131::AID-BDD87%3e3.0.CO;2-L

    Article  CAS  PubMed  Google Scholar 

  24. Su EJ, Fredriksson L, Geyer M et al (2008) Activation of PDGF-CC by tissue plasminogen activator impairs blood-brain barrier integrity during ischemic stroke. Nat Med 14:731–737. https://doi.org/10.1038/nm1787

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Choi KH, Kim HS, Park MS et al (2016) Regulation of Caveolin-1 expression determines early brain edema after experimental focal cerebral ischemia. Stroke 47:1336–1343. https://doi.org/10.1161/STROKEAHA.116.013205

    Article  CAS  PubMed  Google Scholar 

  26. Chen S, Chen Z, Cui J et al (2018) Early abrogation of gelatinase activity extends the time window for tpa thrombolysis after embolic focal cerebral ischemia in mice. eNeuro 5. https://doi.org/10.1523/ENEURO.0391-17.2018

  27. Nag S (2003) Pathophysiology of blood-brain barrier breakdown. Methods Mol Med 89:97–119. https://doi.org/10.1385/1-59259-419-0:97

    Article  CAS  PubMed  Google Scholar 

  28. Blochet C, Buscemi L, Clément T et al (2020) Involvement of caveolin-1 in neurovascular unit remodeling after stroke: effects on neovascularization and astrogliosis. J Cereb Blood Flow Metab 40:163–176. https://doi.org/10.1177/0271678X18806893

    Article  CAS  PubMed  Google Scholar 

  29. Raeeszadeh-Sarmazdeh M, Do LD, Hritz BG (2020) Metalloproteinases and their inhibitors: potential for the development of new therapeutics. Cells 9:1–34. https://doi.org/10.3390/cells9051313

    Article  CAS  Google Scholar 

  30. Castellanos M, Van Eendenburg C, Gubern C et al (2018) Low levels of caveolin-1 predict symptomatic bleeding after thrombolytic therapy in patients with acute ischemic stroke. Stroke 49:1525–1527. https://doi.org/10.1161/STROKEAHA.118.020683

    Article  CAS  PubMed  Google Scholar 

  31. Zhang J, Zhu W, Xiao L et al (2016) Lower serum Caveolin-1 is associated with cerebral microbleeds in patients with acute ischemic stroke. Oxid Med Cell Longev. https://doi.org/10.1155/2016/9026787

    Article  PubMed  PubMed Central  Google Scholar 

  32. Watanabe T, Dohgu S, Takata F et al (2013) Paracellular barrier and tight junction protein expression in the immortalized brain endothelial cell lines bend.3, bend.5 and mouse brain endothelial cell 4. Biol Pharm Bull 36:492–495. https://doi.org/10.1248/bpb.b12-00915

    Article  CAS  PubMed  Google Scholar 

  33. Liu J, Jin X, Liu KJKJ, Liu W (2012) Matrix metalloproteinase-2-mediated occludin degradation and caveolin-1-mediated claudin-5 redistribution contribute to blood-brain barrier damage in early ischemic stroke stage. J Neurosci 32:3044–3057. https://doi.org/10.1523/JNEUROSCI.6409-11.2012

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Yang MC, Zhang HZ, Wang Z et al (2016) The molecular mechanism and effect of cannabinoid-2 receptor agonist on the blood-spinal cord barrier permeability induced by ischemia-reperfusion injury. Brain Res 1636:81–92. https://doi.org/10.1016/j.brainres.2016.01.047

    Article  CAS  PubMed  Google Scholar 

  35. Lee H, Volonte’ D, Galbiati F et al (2000) Constitutive and growth factor-regulated phosphorylation of caveolin-1 occurs at the same site (Tyr-14) in vivo: Identification of a c-Src/Cav-1/Grb7 signaling cassette. Mol Endocrinol 14:1750–1775. https://doi.org/10.1210/mend.14.11.0553

    Article  CAS  PubMed  Google Scholar 

  36. Li S, Seitz R, Lisanti MP (1996) Phosphorylation of caveolin by Src tyrosine kinases: the α-isoform of caveolin is selectively phosphorylated by v-Src in vivo. J Biol Chem 271:3863–3868. https://doi.org/10.1074/jbc.271.7.3863

    Article  CAS  PubMed  Google Scholar 

  37. Zimnicka AMAM, Husain YSYS, Shajahan ANAN et al (2016) Src-dependent phosphorylation of caveolin-1 Tyr-14 promotes swelling and release of caveolae. Mol Biol Cell 27:2090–2106. https://doi.org/10.1091/mbc.E15-11-0756

    Article  PubMed  PubMed Central  Google Scholar 

  38. Sun Y, Hu G, Zhang X, Minshall RDD (2009) Phosphorylation of caveolin-1 regulates oxidant-induced pulmonary vascular permeability via paracellular and transcellular pathways. Circ Res 105:676–685. https://doi.org/10.1161/CIRCRESAHA.109.201673

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Coelho-Santos V, Socodato R, Portugal C et al (2016) Methylphenidate-triggered ROS generation promotes caveolae-mediated transcytosis via Rac1 signaling and c-Src-dependent caveolin-1 phosphorylation in human brain endothelial cells. Cell Mol Life Sci 73:4701–4716. https://doi.org/10.1007/s00018-016-2301-3

    Article  CAS  PubMed  Google Scholar 

  40. Rui Q, Ni H, Lin X, et al (2019) Astrocyte-derived fatty acid-binding protein 7 protects blood-brain barrier integrity through a caveolin-1/MMP signaling pathway following traumatic brain injury. Exp Neurol 322. https://doi.org/10.1016/j.expneurol.2019.113044

Download references

Acknowledgements

The authors thank Dr. Maria Buxó for her support in the statistical data analysis.

Funding

This work was supported by grants from Instituto de Salud Carlos III and co-financed by the European Development Regional Fund “A Way to Achieve Europe” Health Strategic Action Program PI13/02258 and PI17/02123 (MC), PI20/00535 (IL), and Spanish Stroke Research Network RETICS RD12/0014/0010 (MC), and RD16/0019/0003 (JS), RD16/0019/0004 (MC), and RD16/0019/0009 (IL); from Regional Madrid Government B2017/BMD- 3688 (IL); from Spanish Ministry of Science and Innovation PID2019-106581RB-I00 (MAM); from Leducq Foundation for Cardiovascular Research TNE-19CVD01 (MAM); and from Fundación La Caixa HR17_00527 (MAM). P. Comajoan was a recipient of a predoctoral fellowship from the University of Girona (IF-UdG 2015).

Author information

Authors and Affiliations

Author notes

  1. Carme Gubern-Mérida and Pau Comajoan are joint first authors.

    • Elisabet Kádár and Mar Castellanos are joint last authors.

      Authors

      Contributions

      CG, PC, and EK performed in vitro procedures and Western blot, ELISA, and immunofluorescence analysis. IP helped with immunohistochemistry analysis. IG performed the mouse thromboembolic stroke model. CG, PC, EK, and GH designed the experiments and interpreted data. IL, MAM, JS, and MC contributed to data interpretation and, joint to JMS, critically revised the manuscript. CG, PC, and EK were the major contributors in writing the manuscript. All the authors read and approved the final manuscript and agreed to be accountable in ensuring appropriate answer to questions related to the accuracy and integrity of any part of the work.

      Corresponding authors

      Correspondence to Elisabet Kádár or Mar Castellanos.

      Ethics declarations

      Ethics Approval

      All procedures were performed in accordance with the European Communities Council Directive (86/609/EEC) and approved by the Ethics Committee on Animal Welfare of University Complutense (PROEX No. 016/18) and are reported according to ARRIVE (Animal Research: Reporting of In Vivo Experiments) guidelines.

      Consent to Participate

      Not applicable

      Consent to Publish

      All the authors verify that they concur with the present submission and that the material submitted has not been previously reported in any other journal.

      Competing Interests

      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.

      Rights and permissions

      Reprints and permissions

      About this article

      Check for updates. Verify currency and authenticity via CrossMark

      Cite this article

      Gubern-Mérida, C., Comajoan, P., Huguet, G. et al. Cav-1 Protein Levels in Serum and Infarcted Brain Correlate with Hemorrhagic Volume in a Mouse Model of Thromboembolic Stroke, Independently of rt-PA Administration. Mol Neurobiol 59, 1320–1332 (2022). https://doi.org/10.1007/s12035-021-02644-y

      Download citation

      • Received:

      • Accepted:

      • Published:

      • Issue Date:

      • DOI: https://doi.org/10.1007/s12035-021-02644-y

      Keywords

      Navigation