Abstract

Delayed thrombolytic treatment with recombinant tissue plasminogen activator (tPA) may exacerbate blood-brain barrier breakdown after ischaemic stroke and lead to lethal haemorrhagic transformation. The immune system is a dynamic modulator of stroke response, and excessive immune cell accumulation in the cerebral vasculature is associated with compromised integrity of the blood-brain barrier. We previously reported that regulatory T cells, which function to suppress excessive immune responses, ameliorated blood-brain barrier damage after cerebral ischaemia. This study assessed the impact of regulatory T cells in the context of tPA-induced brain haemorrhage and investigated the underlying mechanisms of action. The number of circulating regulatory T cells in stroke patients was dramatically reduced soon after stroke onset (84 acute ischaemic stroke patients with or without intravenous tPA treatment, compared to 115 age and gender-matched healthy controls). Although stroke patients without tPA treatment gradually repopulated the numbers of circulating regulatory T cells within the first 7 days after stroke, post-ischaemic tPA treatment led to sustained suppression of regulatory T cells in the blood. We then used the murine suture and embolic middle cerebral artery occlusion models of stroke to investigate the therapeutic potential of adoptive regulatory T cell transfer against tPA-induced haemorrhagic transformation. Delayed administration of tPA (10 mg/kg) resulted in haemorrhagic transformation in the ischaemic territory 1 day after ischaemia. When regulatory T cells (2 × 106/mouse) were intravenously administered immediately after delayed tPA treatment in ischaemic mice, haemorrhagic transformation was significantly decreased, and this was associated with improved sensorimotor functions. Blood-brain barrier disruption and tight junction damages were observed in the presence of delayed tPA after stroke, but were mitigated by regulatory T cell transfer. Mechanistic studies demonstrated that regulatory T cells completely abolished the tPA-induced elevation of MMP9 and CCL2 after stroke. Using MMP9 and CCL2 knockout mice, we discovered that both molecules partially contributed to the protective actions of regulatory T cells. In an in vitro endothelial cell-based model of the blood-brain barrier, we confirmed that regulatory T cells inhibited tPA-induced endothelial expression of CCL2 and preserved blood-brain barrier integrity after an ischaemic challenge. Lentivirus-mediated CCL2 knockdown in endothelial cells completely abolished the blood-brain barrier protective effect of regulatory T cells in vitro. Altogether, our studies suggest that regulatory T cell adoptive transfer may alleviate thrombolytic treatment-induced haemorrhage in stroke victims. Furthermore, regulatory T cell-afforded protection in the tPA-treated stroke model is mediated by two inhibitory mechanisms involving CCL2 and MMP9. Thus, regulatory T cell adoptive transfer may be useful as a cell-based therapy to improve the efficacy and safety of thrombolytic treatment for ischaemic stroke.

Highlights

  • Recombinant tissue plasminogen activator is the only FDA-approved thrombolytic treatment for acute ischaemic stroke

  • Treg treatment further lessened blood–brain barrier (BBB) damage and haemorrhage in MMP9 knockout mice, suggesting that one or more MMP9 independent mechanism may contribute to the protective effect of Tregs. These results suggest that Treg treatment suppresses tissue plasminogen activator (tPA)-induced MMP9 expression, but this only partially contributes to its protective effects on BBB integrity and cerebral haemorrhage

  • TPA is an FDA-approved medicine for thrombolytic treatment in acute ischaemic stroke, the delayed application of tPA beyond the 4.5 h therapeutic window leads to an increased risk of lethal haemorrhagic transformation

Read more

Summary

Introduction

Recombinant tissue plasminogen activator (tPA) is the only FDA-approved thrombolytic treatment for acute ischaemic stroke. The use of tPA increases the risk of haemorrhagic transformation, especially when delayed beyond 4.5 h after the onset of ischaemia (Dijkhuizen et al, 2002; Yepes et al, 2003; Su et al, 2008). Delayed tPA treatment after stroke further exacerbates BBB damage and increases the risk of lethal haemorrhagic transformation (Potrovita et al, 2004; Wang et al, 2015). For MAP2 staining, free-floating sections were prepared and stained with MAP2 antibodies (1:200, Santa Cruz Biotechnology). For both TTC and MAP2 staining, infarct area was determined with NIH ImageJ. Infarct volumes (with correction for brain oedema) were calculated as the volume of the contralateral hemisphere minus the non-infarcted volume of the ipsilateral hemisphere.

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call