Elsevier

Experimental Neurology

Volume 207, Issue 2, October 2007, Pages 227-237
Experimental Neurology

Minocycline protects the blood–brain barrier and reduces edema following intracerebral hemorrhage in the rat

https://doi.org/10.1016/j.expneurol.2007.06.025Get rights and content

Abstract

Intracerebral hemorrhage (ICH) results from rupture of a blood vessel in the brain. After ICH, the blood–brain barrier (BBB) surrounding the hematoma is disrupted, leading to cerebral edema. In both animals and humans, edema coincides with inflammation, which is characterized by production of pro-inflammatory cytokines, activation of resident brain microglia and migration of peripheral immune cells into the brain. Accordingly, inflammation is an attractive target for reducing edema following ICH. In the present study, BBB damage was assessed by quantifying intact microvessels surrounding the hematoma, monitoring extravasation of IgG and measuring brain water content 3 days after ICH induced by collagenase injection into the rat striatum. In the injured brain, the water content increased in both ipsilateral and contralateral hemispheres compared with the normal brain. Quantitative real-time RT-PCR revealed an up-regulation of inflammatory genes associated with BBB damage; IL1β, TNFα and most notably, MMP-12. Immunostaining showed MMP-12 in damaged microvessels and their subsequent loss from tissue surrounding the hematoma. MMP-12 was also observed for the first time in neurons. Dual-antibody labeling demonstrated that neutrophils were the predominant source of TNFα protein. Intraperitoneal injection of the tetracycline derivative, minocycline, beginning 6 h after ICH ameliorated the damage by reducing microvessel loss, extravasation of plasma proteins and edema; decreasing TNFα and MMP-12 expression; and reducing the numbers of TNFα-positive cells and neutrophils in the brain. Thus, minocycline, administered at a clinically relevant time, appears to target the inflammatory processes involved in edema development after ICH.

Introduction

Intracerebral hemorrhage (ICH) accounts for 10–30% of all strokes, with as much as 30–40% mortality by one month (Rincon and Mayer, 2004). After ICH, cerebral edema begins within hours, can last for weeks and is thought to contribute to neurological deterioration by increasing intracranial pressure or causing a shift in midline structures (Mayer and Rincon, 2005). Vasogenic edema is prevalent after ICH (Rincon and Mayer, 2004) and results from pathological changes in the blood–brain barrier (BBB) and consequent fluid movement from the vasculature to the extracellular space of the brain. Less important is the cytotoxic edema resulting from increased water uptake by brain cells (especially neurons). The BBB serves as the anatomical barrier between the blood and brain parenchyma and is comprised of endothelial cells, astrocytic end-feet, pericytes and a specialized basement membrane (the basal lamina) that serves as a scaffold supporting the physical integrity of the microvasculature. Although the basal lamina is clearly involved in BBB breakdown after ischemic stroke (Hamann et al., 2002), its role after ICH remains unresolved.

The inflammatory response following ICH, like other forms of brain injury, is characterized by production of several molecules that can disrupt the BBB (Wang and Dore, 2006). Moreover, because the delayed onset of both inflammation and vasogenic edema allows more time for patients to reach hospital after the onset of ICH, they are attractive therapeutic targets. In other forms of brain injury, the initial inflammatory response is orchestrated by the cytokines, TNFα and IL1β, which are up-regulated within hours (Barone and Feuerstein, 1999, Hua et al., 2006, Mayne et al., 2001b, Mayne et al., 2001a, Qureshi et al., 2001), but less is known about their temporal and spatial expression after ICH. While the ‘resting’ BBB severely limits movement of fluid, macromolecules and cells into the brain, TNFα and IL1β ‘activate’ the endothelium, increase its permeability and allow adhesion and entry of peripheral immune cells (Feuerstein et al., 1998, Middleton et al., 2002, Zhang et al., 2000). Transmigrating neutrophils in particular, produce cytotoxic molecules including inflammatory cytokines, reactive oxygen species and matrix metalloproteinases (MMPs) (Scholz et al., 2007). MMP production, also ascribed to endothelial cells, microglia/macrophages, astrocytes and neurons, can further damage the BBB by degrading the basal lamina of cerebral blood vessels (Rosenberg, 2002). Following ICH in the rat, several MMPs are up-regulated (Power et al., 2003, Wells et al., 2005), but their impact on cerebral blood vessels and BBB breakdown has yet to be assessed.

Minocycline, a semi-synthetic tetracycline derivative, is of particular therapeutic interest for CNS disorders because it has a high oral bioavailability, superior BBB penetration and is well tolerated by humans, where it has been used for decades to treat bacterial infections (Stirling et al., 2005, Yong et al., 2004). Its efficacy has been demonstrated in animal models of acute brain injury, including focal ischemia (Yrjanheikki et al., 1998, Yrjanheikki et al., 1999), ICH (Power et al., 2003), traumatic brain injury (Sanchez Mejia et al., 2001) and spinal cord injury (Wells et al., 2003), and proposed mechanisms include very diverse effects on several cell types. With respect to inflammation, it can inhibit activation and migration of inflammatory cells, and after ICH in the rat it inhibits macrophages/microglia (Power et al., 2003, Wasserman and Schlichter, 2007) and neutrophils (Wasserman and Schlichter, 2007). Minocycline has been reported to reduce specific cytokines (TNFα, IL1β) and MMPs (Elewa et al., 2006, Power et al., 2003) that have been implicated in BBB damage, and in animal models of ischemic stroke it can reduce BBB damage (Maier et al., 2006, Yenari et al., 2006).

The present work builds on two studies of collagenase-induced ICH in the rat striatum. Power and colleagues (2003) described the time course of MMP mRNA expression distal to the hematoma (2–4 mm) and effects of early treatment with minocycline (starting at 1 h) on MMP up-regulation and the neurological outcome. In the same model (Wasserman and Schlichter, 2007), we recently investigated the spatial and temporal relationship between neuron death and inflammation, and effects of minocycline treatment begun at a later, more clinically relevant time (6 h); i.e., after the hematoma had reached its maximal size. Using the same model, we now assess the effects of delayed minocycline treatment on blood vessel deterioration and edema, and on time-dependent changes in several key molecules involved in these events. BBB damage was monitored by counting the number of intact microvessels surrounding the hematoma (Hamann et al., 2002), assessing the extravasation of IgG into the brain (Tanno et al., 1992), and measuring brain water content 3 days after ICH onset. Then, because a prominent inflammatory response occurs in the tissue immediately adjacent to the hematoma, we monitored gene expression in and around the damage site with and without minocycline treatment and began at an earlier time than previously examined. Expression of IL1β, TNFα, MMP-3, MMP-9, MMP-12 and the microglia/macrophage marker, complement receptor 3 were monitored at 6 h, 1 day and 3 days after ICH, using quantitative real-time RT-PCR. Finally, for the two genes whose up-regulation was reduced by minocycline (TNFα, MMP-12), we used immunohistochemistry to determine which cell types expressed the proteins. We show that TNFα increased early, mainly in neutrophils; whereas, MMP-12 increased later in damaged microvessels surrounding the hematoma. Delayed minocycline treatment reduced: microvessel loss, plasma protein extravasation and edema; expression of TNFα and MMP-12; and the number of TNFα-positive cells and neutrophils in the brain. These findings support a model whereby immune cells produce TNFα, which leads to an increase in MMP-12 in microvessels surrounding the hematoma, causing further degradation of the basal lamina and BBB disruption.

Section snippets

Intracerebral hemorrhage and minocycline treatment

Intracerebral hemorrhage was induced in the striatum of male Sprague–Dawley rats (300–350 g, n = 40) as previously described (Rosenberg et al., 1990, Wasserman and Schlichter, 2007). All procedures were approved by the University Health Network animal care committee, in accordance with guidelines established by the Canadian Council on Animal Care. Rats were anesthetized using isoflurane (3% induction, 1.5% maintenance) and placed in a stereotaxic frame. Under aseptic conditions, a 1-mm diameter

Minocycline reduces cerebral edema and the loss of microvessels

Cerebral edema peaks at 3–4 days after ICH onset in the rat (see Discussion); thus, we measured the brain water content at 3 days (Fig. 1B). Naïve rats, not subjected to any surgical procedure, had a brain water content of 78.65 ± 0.11% (n = 6), whereas sham-operated rats (saline-injected, no collagenase; n = 3) had a brain water content of 79.2 ± 0.09% in the ipsilateral hemisphere and 78.9 ± 0.06 in the contralateral hemisphere. The cerebral hemisphere was used as the internal control, rather than the

Discussion

This work presents several novel findings. The results show for the first time that minocycline treatment, delayed to a clinically relevant starting time, protects the blood–brain barrier (BBB) after ICH. This protection was manifested by amelioration of three key processes; i.e., decreases in the number of disrupted microvessels, serum protein extravasation into the brain and cerebral edema. This is apparently the first report using collagen type IV expression in order to show reduced numbers

Acknowledgments

Excellent technical help was provided by X-P Zhu and H Yang. We are grateful for comments from Drs. J Eubanks, L Mills and EF Stanley. Supported by grants to LCS from the Heart and Stroke Foundation (HSFO; NA5158, T5546), Krembil Scientific Development Seed Fund, Canadian Stroke Network and a scholarship to JW from HSF Canada.

References (72)

  • A. Szymanska et al.

    Minocycline and intracerebral hemorrhage: influence of injury severity and delay to treatment

    Exp. Neurol.

    (2006)
  • C.M. Vos et al.

    Matrix metalloproteinase-12 is expressed in phagocytotic macrophages in active multiple sclerosis lesions

    J. Neuroimmunol.

    (2003)
  • J.K. Wasserman et al.

    Neuron death and inflammation in a rat model of intracerebral hemorrhage: Effects of delayed minocycline treatment

    Brain Res.

    (2007)
  • D. Wong et al.

    Cytokines, nitric oxide, and cGMP modulate the permeability of an in vitro model of the human blood–brain barrier

    Exp. Neurol.

    (2004)
  • G. Xi et al.

    Mechanisms of brain injury after intracerebral haemorrhage

    Lancet Neurol.

    (2006)
  • V.W. Yong et al.

    The promise of minocycline in neurology

    Lancet Neurol.

    (2004)
  • S. Abilleira et al.

    Matrix metalloproteinase-9 concentration after spontaneous intracerebral hemorrhage

    J. Neurosurg.

    (2003)
  • P. Borden et al.

    Transcriptional control of matrix metalloproteinases and the tissue inhibitors of matrix metalloproteinases

    Crit. Rev. Eukaryot. Gene Expr.

    (1997)
  • F.C. Barone et al.

    Inflammatory mediators and stroke: new opportunities for novel therapeutics

    J. Cereb. Blood Flow Metab.

    (1999)
  • G.I. Botchkina et al.

    Expression of TNF and TNF receptors (p55 and p75) in the rat brain after focal cerebral ischemia

    Mol. Med.

    (1997)
  • S.A. Bustin et al.

    Pitfalls of quantitative real-time reverse-transcription polymerase chain reaction

    J. Biomol. Tech.

    (2004)
  • J. Castillo et al.

    Molecular signatures of brain injury after intracerebral hemorrhage

    Neurology

    (2002)
  • N. Didier et al.

    Secretion of interleukin-1beta by astrocytes mediates endothelin-1 and tumour necrosis factor-alpha effects on human brain microvascular endothelial cell permeability

    J. Neurochem.

    (2003)
  • D. Dziewulska et al.

    Cellular expression of tumor necrosis factor a and its receptors in human ischemic stroke

    Clin. Neuropathol.

    (2003)
  • H.F. Elewa et al.

    Minocycline for short-term neuroprotection

    Pharmacotherapy

    (2006)
  • G.Z. Feuerstein et al.

    The role of cytokines in the neuropathology of stroke and neurotrauma

    Neuroimmunomodulation

    (1998)
  • Y. Gasche et al.

    Early appearance of activated matrix metalloproteinase-9 after focal cerebral ischemia in mice: a possible role in blood–brain barrier dysfunction

    J. Cereb. Blood Flow Metab.

    (1999)
  • R. Gregersen et al.

    Microglia and macrophages are the major source of tumor necrosis factor in permanent middle cerebral artery occlusion in mice

    J. Cereb. Blood Flow Metab.

    (2000)
  • G.F. Hamann et al.

    Microvascular basal lamina injury after experimental focal cerebral ischemia and reperfusion in the rat

    J. Cereb. Blood Flow Metab.

    (2002)
  • G.F. Hamann et al.

    Mild to moderate hypothermia prevents microvascular basal lamina antigen loss in experimental focal cerebral ischemia

    Stroke

    (2004)
  • S. Holmin et al.

    Intracerebral administration of interleukin-1beta and induction of inflammation, apoptosis, and vasogenic edema

    J. Neurosurg.

    (2000)
  • Y. Hua et al.

    Tumor necrosis factor-alpha increases in the brain after intracerebral hemorrhage and thrombin stimulation

    Neurosurgery

    (2006)
  • V. Hummel et al.

    Production of MMPs in human cerebral endothelial cells and their role in shedding adhesion molecules

    J. Neuropathol. Exp. Neurol.

    (2001)
  • V. Kaushal et al.

    The Ca2+-activated K+ channel KCNN4/KCa3.1 contributes to microglia activation and nitric oxide-dependent neurodegeneration

    J. Neurosci.

    (2007)
  • I. Klatzo

    Pathophysiological aspects of brain edema

    Acta Neuropathol. (Berl)

    (1987)
  • J.M. Knapp

    Hyperosmolar therapy in the treatment of severe head injury in children: mannitol and hypertonic saline

    AACN Clin. Issues

    (2005)
  • Cited by (129)

    • Potential application of intranasal insulin delivery for treatment of intracerebral hemorrhage: A review of the literature

      2022, Journal of Stroke and Cerebrovascular Diseases
      Citation Excerpt :

      The inflammatory response in ICH also leads to activation of inflammatory cells that produce ROS and favor secondary brain injury.59 Previously, targeting ROS production using scavengers such as deferoxamine, minocycline, and NXY-059 showed to reduce oxidative stress in ICH.29–32,60,61 In a mouse model of traumatic brain injury (TBI), intranasal insulin administration reduced microglial activation and decreased the production of monocyte chemoattractant protein-1 (MCP-1), nitric oxide, and ROS, ultimately inhibiting further microglial recruitment.4

    • Mechanisms of Damage After Cerebral Hemorrhage

      2021, Stroke: Pathophysiology, Diagnosis, and Management
    View all citing articles on Scopus
    View full text