Elsevier

Brain Research

Volume 1646, 1 September 2016, Pages 494-503
Brain Research

Research report
Cilostazol reduces blood brain barrier dysfunction, white matter lesion formation and motor deficits following chronic cerebral hypoperfusion

https://doi.org/10.1016/j.brainres.2016.06.036Get rights and content

Highlights

  • BBB permeability is significantly elevated for 2 weeks following permanent BCCAO in the rat.

  • BCCAO was found to impair gait in the tapered beam test.

  • Cilostazol, a type III phosphodiesterase inhibitor, reduced Evans blue dye extravasation and gait disturbance in vivo.

  • Cilostazol reduces white matter rarefaction and gliosis in the optic tract.

  • Cilostazol reduced plasma toxicity in vitro.

Abstract

Cerebral small vessel disease (CSVD) is a pathological process leading to lacunar infarcts, leukoaraiosis and cerebral microbleeds. Dysfunction of the blood brain barrier (BBB) has been proposed as a mechanism in the progression cerebral small vessel disease. A rodent model commonly used to study some aspects of CSVD is bilateral common carotid artery occlusion (BCCAO) in the rat. In the present study it was determined that gait impairment, as determined by a tapered beam test, and BBB permeability increased following BCCAO. Cilostazol, a type III phosphodiesterase inhibitor, has been shown to have anti-apoptotic effects and prevent white matter vacuolation and rarefaction induced by BCCAO in rats. In this study the protective effect of cilostazol administration on the increase BBB permeability following BCCAO was determined as well as the effect on plasma levels of circulating microparticles (MPs), cerebral white matter rarefaction, glial activation and gait disturbance. The effect of cilostazol on in vitro endothelial barriers was also evaluated. Cilostazol treatment improved BBB permeability and reduced gait disturbance, visual impairment and microglial activation in optic tract following BCCAO in vivo. It also reduced the degree of cell death and the reduction in trans-endothelial electrical resistance (TEER) in artificial endothelial barriers in vitro induced by MP treatment of in vitro barriers.

Introduction

Cerebral small vessel disease (CSVD) is now thought to be the most common cause of vascular cognitive impairment (VCI) and accounts for more than 40% of all VCI (Bowler, 2005). CSVD is associated with chronic cerebral hypoperfusion and the reduction in blood flow in the perforating arteries resulting in ischemia in the white matter and deep gray matter nuclei. The microangiopathy induced by CSVD is characterized radiologically by the presence of lacunes (microinfarcts of the central gray matter) and leukoaraiosis in various regions of the brain (Schmidtke and Hüll, 2005). CSVD is a salient risk factor for stroke (Wardlaw et al., 2013). CSVD represents a serious health care challenge, as lacunar stroke may occur with an incidence of up to five-times that of large vessel stroke (Vermeer et al., 2007). CSVD may result in cognitive impairment, especially affecting executive function, and mild motor impairments such as gait disturbance (de Laat et al., 2010). Disruption of white matter tracts that connect motor regions is thought to be the mechanism underlying the gait disturbance noted in some patient with CSVD (Chui, 2007), and WM lesions in certain brain regions such as subcortical areas and periventricular frontal lobe are related to lower gait velocity, shorter stride length and broader stride width (Silbert et al., 2008).

The endothelium plays a crucial role in processes such as the autoregulation of blood flow, reactive hyperemia and formation and function of the blood-brain barrier. A failure of the arteriolar endothelium due to partial ischemia has been proposed as an early event in the pathogenesis of CSVD (Pantoni and Garcia, 1997). Endothelial injury also leads to BBB leakage, which has been considered as the starting point of CSVD (Schreiber et al., 2013). This derangement in the blood brain barrier may initiate some years before the first symptoms of CSVD and leads to structural, vascular and perivascular changes in brain small vessels such as edema, enlarged perivascular spaces and tissue damage (Wardlaw, 2005, Wardlaw, 2010).

MPs are small vesicles, 0.1–1.0 µm in size, that bud off from plasma membrane of various cell types in response to shear stress, physiological agonizts such as collagen (Morel et al., 2011), and pro-apoptotic stimuli (György et al., 2011). MPs may also be released spontaneously from cells under basal condition (Italiano et al., 2010). MPs transfer information from the cell of origin to target cells by cell-to-cell contact or through secretion of soluble mediators (Mause and Weber, 2010). It has been proposed that MPs play a role in pathogenesis of cerebral malaria and multiple sclerosis through induction of brain endothelial dysfunction and alterations in BBB permeability (Combes et al., 2006, Minagar and Alexander, 2003). Circulating MPs are markers of early stages of multiple sclerosis and increase endothelial permeability (Marcos-Ramiro et al., 2014).

BCCAO is one of most widely used models for the study of CVSD (Hainsworth and Markus, 2008, Hainsworth et al., 2012). BCCAO in rats induces a chronic reduction in cerebral blood flow in frontal cortex for 8 weeks to 3 months after surgery (Ohta et al., 1997, Ueno et al., 2002, Farkas et al., 2007). Lesions are mainly in white matter (WM), and appear 7 days after occlusion and are persistent (Wakita et al., 1995; Ueno et al., 2002; Farkas et al., 2004). They are preceded by temporary BBB opening in white matter areas with collagen deposition in vessel walls (Ueno et al., 2002).

Cilostazol, a type III phosphodiesterase inhibitor, has been reported to be neuroprotective against apoptotic white matter damage and cognitive impairment in rats that received BCCAO (Lee et al., 2006, Watanabe et al., 2006, Lee et al., 2007, Kwak et al., 2012). It has been recently acknowledged as one of potential pharmacologic interventions for treatment of CSVD (Bath and Wardlaw, 2015).

It has been previously shown that the induction of chronic cerebral hypoperfusion in rats causes an increase in the number of MPs circulating in the plasma, and these MPs are able to induce cell death when they are applied to rat brain microvascular endothelial cells (Schock et al., 2014). MPs isolated from plasma of rats which received BCCAO also increase rat brain microvascular endothelial cell (RBMVECs) permeability when they are applied to in vitro artificial barriers (Edrissi et al., 2016).

The purpose of this study was to assess the role of increased BBB permeability in the progression of small vessel disease using this rat model of chronic cerebral hypoperfusion. The specific objectives were to study the extent and time course of changes in BBB permeability and motor impairment and to determine the effects of cilostazol administration on these processes. Cilostazol treatment reduced the effect of chronic ischemia on the BBB, improved gait performance in the tapered beam test and reduced the severity of lesions in vulnerable white matter regions (optic tract and corpus callosum) following BCCAO. Plasma toxicity was measured at 2 weeks following BCCAO in order to determine the association between the numbers of circulating MPs and the pathological consequences of cerebral ischemia.

Section snippets

Increased BBB permeability following BCCAO

Post hoc measurement of Evans Blue leakage in the frontal cortex showed that 2VO rats had a significant increase in dye extravasation that began as early as 24 h and lasted up to 2 weeks post-surgery. There was no difference between sham and 2VO groups at 4 weeks after surgery (Fig. 1A, group effect: F (1, 50)=38.575, P<0.0001, time effect: F (4, 50)=4.48 (P<0.01) time*group effect: F (1, 50)=2.04 (p<0.05)2). EB Leakage appeared as a red fluorescence in brain sections obtained 3 days after

Discussion

The BBB is relatively vulnerable to ischemia as it becomes permeable to large molecules under severe ischemic conditions. However, there is little evidence in the literature to date of BBB disruption in the 2VO rat model of chronic cerebral hypoperfusion (Farkas et al., 2007). Some studies have indicated altered vascular permeability to horse radish peroxidase in paramedian portion of corpus callosum (Ueno et al., 2002) or extravasation of Evans Blue bound to albumin at 3 days following 2VO

Animals and surgical procedure

All experiments conformed to the guidelines set forth by the Canadian Council for the Use and Care of Animals in Research (CCAC) with approval from the Ottawa Health Research Institute Animal Care Facility. Male Long Evans rats weighing 200–225 g (Charles River) received sham or 2VO (two vessel occlusion) surgery and survived for different periods of time following surgery. A group of rats did not receive sham or 2VO surgery and was designated as un-operated control. The surgery performed as

References (58)

  • Y.J. Lee et al.

    Elevated platelet microparticles in transient ischemic attacks, lacunar infarcts, and multiinfarct dementias

    Thromb. Res.

    (1993)
  • H. Ohta et al.

    Chronic cerebral hypoperfusion by permanent internal carotid ligation produces learning impairment without brain damage in rats

    Neuroscience

    (1997)
  • C. Sarti et al.

    Persistent impairment of gait performances and working memory after bilateral common carotid artery occlusion in the adult Wistar rat

    Behav. Brain Res.

    (2002)
  • K. Schmidtke et al.

    Cerebral small vessel disease: how does it progress?

    J. Neurol. Sci.

    (2005)
  • S.C. Schock et al.

    Microparticles generated during chronic cerebral ischemia deliver proapoptotic signals to cultured endothelial cells

    Biochem. Biophys. Res. Commun.

    (2014)
  • J.S. Shin et al.

    Chronic hypoperfusion increases claudin-3 immunoreactivity in rat brain

    Neurosci. Lett.

    (2008)
  • J. Simak et al.

    Circulating endothelial microparticles in acute ischemic stroke: a link to severity, lesion volume and outcome

    J. Thromb. Haemost.

    (2006)
  • H. Torii et al.

    Cilostazol inhibits the redistribution of the actin cytoskeleton and junctional proteins on the blood-brain barrier under hypoxia/reoxygenation

    Pharmacol. Res.

    (2007)
  • S.E. Vermeer et al.

    Silent brain infarcts: a systematic review

    Lancet Neurol.

    (2007)
  • J.M. Wardlaw

    Blood-brain barrier and cerebral small vessel disease

    J. Neurol. Sci.

    (2010)
  • J.M. Wardlaw et al.

    Mechanisms of sporadic cerebral small vessel disease: insights from neuroimaging

    Lancet Neurol.

    (2013)
  • H. Yamamoto et al.

    Complex neurodegeneration in retina following moderate ischemia induced by bilateral common carotid artery occlusion in Wistar rats

    Exp. Eye Res.

    (2006)
  • P.M. Bath et al.

    Pharmacological treatment and prevention of cerebral small vessel disease: a review of potential interventions

    Int. J. Stroke

    (2015)
  • F. Bianco et al.

    Acid sphingomyelinase activity triggers microparticle release from glial cells

    EMBO J.

    (2009)
  • J.V. Bowler

    Vascular cognitive impairment

    J. Neurol. Neurosurg. Psychiatry

    (2005)
  • S. Chahed et al.

    Increased vitreous shedding of microparticles in proliferative diabetic retinopathy stimulates endothelial proliferation

    Diabetes

    (2010)
  • P. Cherian et al.

    Endothelial and platelet activation in acute ischemic stroke and its etiological subtypes

    Stroke

    (2003)
  • A. Corbett et al.

    A method for reliable voluntary oral administration of a fixed dosage (mg/kg) of chronic daily medication to rats

    Lab. Anim.

    (2012)
  • L. Doeuvre et al.

    Cell-derived microparticles: a new challenge in neuroscience

    J. Neurochem.

    (2009)
  • Cited by (36)

    • Post-stroke treatment with K-134, a phosphodiesterase 3 inhibitor, improves stroke outcomes in the stroke-prone spontaneously hypertensive rat model–A comparative evaluation of antiplatelet drugs

      2022, Journal of Pharmacological Sciences
      Citation Excerpt :

      Both cAMP pathways and PDE3 have well-described key roles in the homeostasis of the blood–brain barrier.40 Indeed, previous studies using in vivo animal models other than SHRSP showed that cilostazol ameliorates blood–brain barrier dysfunction induced by bilateral common carotid artery occlusion and ischemia-reperfusion41,42 and reduces hemorrhagic injury induced by collagenase, warfarin, and tissue-type plasminogen activator.43–47 Furthermore, K-134,48 as well as cilostazol,49,50 have pleiotropic effects, including attenuation of inflammatory responses, suppression of oxidative stress, and reduction of matrix metalloproteinases (MMPs) activity.

    • Salvianolic acid A relieves cognitive disorder after chronic cerebral ischemia: Involvement of Drd2/Cryab/NF-κB pathway

      2022, Pharmacological Research
      Citation Excerpt :

      Its main early clinical manifestations were memory decline and progressive cognitive disorder, which seriously affect the daily work and life of patients. CCI is involved in the development of various neurological diseases, such as Alzheimer's disease and vascular dementia [1,2]. The protracted course of the disease results in a series of brain damage, such as blood-brain barrier destruction, abnormal glial cells activation, and neuronal degeneration and loss [3,4].

    • Epimedium flavonoids improve cognitive impairment and white matter lesions induced by chronic cerebral hypoperfusion through inhibiting the Lingo-1/Fyn/ROCK pathway and activating the BDNF/NRG1/PI3K pathway in rats

      2020, Brain Research
      Citation Excerpt :

      All protocols were approved by the Experimental Animal Center of the Capital Medical University. Permanent bilateral common carotid artery ligation (2VO) surgery in rats was performed as previously described (Edrissi et al., 2016). Briefly, rats were anesthetized with chloral hydrate (400 mg/kg) under 12 h fasting conditions.

    View all citing articles on Scopus
    View full text