TNF-α knockout and minocycline treatment attenuates blood–brain barrier leakage in MPTP-treated mice

https://doi.org/10.1016/j.nbd.2006.11.012Get rights and content

Abstract

Following intraparenchymal injection of the dopamine (DA) neurotoxin 6-hydroxydopamine, we previously demonstrated passage of fluoresceinisothiocyanate-labeled albumin (FITC-LA) from blood into the substantia nigra (SN) and striatum suggesting damage to the blood–brain barrier (BBB). The factors contributing to the BBB leakage could have included neuroinflammation, loss of DA neuron control of barrier function, or a combination of both. In order to determine which factor(s) was responsible, we assessed BBB integrity using the FITC-LA technique in wild-type (WT), tumor necrosis factor alpha (TNF-α) knockout (KO), and minocycline (an inhibitor of microglia activation) treated mice 72 h following treatment with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Compared with WT mice, TNF-α KO mice treated with MPTP showed reduced FITC-LA leakage, decreased numbers of activated microglia, and reduced proinflammatory cytokines (TNF-α and interleukin 1β) associated with significant MPTP-induced DA neuron loss. In contrast, minocycline treated animals did not exhibit significant MPTP-induced DA neuron loss although their FITC-LA leakage, numbers of activated microglia, and MPTP-induced cytokines were markedly attenuated. Since both TNF-α KO and minocycline treatment attenuated MPTP-induced BBB dysfunction, microglial activation, and cytokine increases, but had differential effects on DA neuron loss, it appears that neuroinflammation and not DA neuron loss was responsible for disrupting the blood–brain barrier integrity.

Introduction

Parkinson’s disease (PD) is marked by the progressive loss of dopamine (DA) neurons in the substantia nigra (SN) (Hastings and Zigmond, 1997). While the etiology of PD remains unclear, both genetic factors and environmental toxins have been proposed in its pathogenesis (Ladeby et al., 2005, McGeer and McGeer, 2004). Regardless of the underlying etiology, neuroinflammation [the some total of cellular changes (e.g., microglial activation) and secreted factors (e.g., proinflammatory cytokines or free radicals) that accompany an inflammatory response within the CNS] is thought to contribute to the loss of DA neurons seen in patients with PD (McGeer and McGeer, 2004, Ladeby et al., 2005). Neuroinflammation is also present in trauma, stroke, multiple sclerosis, epilepsy and bacterial meningitis (Mennicken et al., 1999, Phillis et al., 2006) in which damage to the blood–brain barrier (BBB) has been reported (Huber et al., 2001). More recent studies point to microvascular changes in the SN and alterations in several markers of normal BBB integrity in PD patients (Barcia et al., 2004, Faucheux et al., 1999, Kortekaas et al., 2005). Whether actual changes in permeability, functionality, or physical damage of the BBB occur in PD is currently unknown.

We recently demonstrated that the DA neurotoxin, 6-hydroxydopamine (6-OHDA), compromised BBB integrity producing apparent leakage of both fluoresceinisothiocyanate-labeled albumin (FITC-LA) and horseradish peroxidase into the SN and striatum. This leakage was accompanied by loss of DA neurons, activation of microglia, up-regulation of p-glycoprotein and β-integrin on the endothelial cells that comprise the BBB, and attenuation of a DA-mediated behavior by domperidone, a DA antagonist that normally does not cross the BBB (Carvey et al., 2005). Since neuroinflammation including microglia activation and increased levels of proinflammatory cytokines including tumor necrosis factor-alpha (TNF-α) are present in patients with PD and in animal models of the disease (Aschner, 1998, Hirsch et al., 2005, Nagatsu and Sawada, 2005) and both are well known to affect the integrity of the BBB (Ryu and McLarnon, 2006, Tsao et al., 2001, Yenari et al., 2006), it is quite possible that 6-OHDA-induced neuroinflammation was responsible for the breakdown in barrier integrity. However, neurons containing biogenic amines are found in close proximity to brain capillaries (Rennels and Nelson, 1975, DiCarlo et al., 1984, Kapadia and de Lanerolle, 1984), endothelial cells express both noradrenergic and serotoninergic transporters (Wakayama et al., 2002) and receptors (Wakayama et al., 2002, Kobayashi et al., 1985), and stimulation of the locus coeruleus increases BBB permeability (Raichle et al., 1975) suggesting that neurotransmitters may regulate BBB function as well. The BBB leakage we observed in rats treated with 6-OHDA could be the result of neuroinflammation, DA neuron loss, or a combination of both. Therefore, we designed a set of experiments to determine the relative contribution of each to the leakage of the BBB.

Section snippets

Animals

A total of 88 male mice, 8 weeks of age and weighing 22–25 g at the start of the study, were used. The TNF-α KO mice (B6;129S6-TnftmlGkl/J; n = 22), WT background control mice (B6;129S6; n = 22), and C57BL/6 (n = 44) mice were purchased from Jackson Laboratory (Bar Harbor, ME). All mice were acclimated to the animal facility for at least 2 weeks prior to the start of the study. One day prior to MPTP treatment, the mice were moved to a controlled ventilated room and housed in ventilation chambers

MPTP increased FITC-LA leakage into the SN and striatum

FITC-LA was injected into the common carotid artery 72 h following treatment with MPTP (a time at which active DA neurodegeneration was occurring (Sugama et al., 2003)), and the brains were assessed using confocal microscopy to determine if this large molecular weight marker (MW range = 69–70 kDa) entered brain parenchyma. FITC-LA was confined exclusively to blood vessels in the SN and striatum in the control animals of both genotypes [WT/Sal (Figs. 1a, c); TNF-α KO/Sal (Fig. 1b, d)] and the

Discussion

The current study confirms and extends our previous work in rats in which injection of the DA neurotoxin 6-hydroxydopamine into the striatum or medial forebrain bundle produced similar patchy areas of leakage of FITC-LA and horseradish peroxidase as well as increases in β3 integrin expression (a marker of angiogenesis) in the SN and striatum (Carvey et al., 2005). In the present study, the DA neurotoxin (MPTP) was injected systemically so that the barrier dysfunction seen could not be a

Acknowledgments

This work was supported by NINDS NS045316, NIEHS 012307, W81XWH-04-01-0365 and the Michael J. Fox Foundation.

References (60)

  • S.E. Kapadia et al.

    Immunohistochemical and electron microscopic demonstration of vascular innervation in the mammalian brainstem

    Brain Res.

    (1984)
  • I. Kurkowska-Jastrzebska et al.

    The inflammatory reaction following 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine intoxication in mouse

    Exp. Neurol.

    (1999)
  • R. Ladeby et al.

    Microglial cell population dynamics in the injured adult central nervous system

    Brain Res. Brain Res. Rev.

    (2005)
  • A. Leng et al.

    Tumor necrosis factor-alpha receptor ablation in a chronic MPTP mouse model of Parkinson’s disease

    Neurosci. Lett.

    (2005)
  • Z. Ling et al.

    Rotenone potentiates dopamine neuron loss in animals exposed to lipopolysaccharide prenatally

    Exp. Neurol.

    (2004)
  • N.J. Lynch et al.

    Microglial activation and increased synthesis of complement component C1q precedes blood–brain barrier dysfunction in rats

    Mol. Immunol.

    (2004)
  • P.L. McGeer et al.

    Inflammation and neurodegeneration in Parkinson’s disease

    Parkinsonism Relat. Disord.

    (2004)
  • F. Mennicken et al.

    Chemokines and chemokine receptors in the CNS: a possible role in neuroinflammation and patterning

    Trends Pharmacol. Sci.

    (1999)
  • J.P. O’callaghan et al.

    Characterization of the origins of astrocyte response to injury using the dopaminergic neurotoxicant, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine

    Brain Res.

    (1990)
  • C.F. Orr et al.

    An inflammatory review of Parkinson’s disease

    Prog. Neurobiol.

    (2002)
  • I. Petrache et al.

    Central involvement of Rho family GTPases in TNF-alpha-mediated bovine pulmonary endothelial cell apoptosis

    Biochem. Biophys. Res. Commun.

    (2003)
  • J.W. Phillis et al.

    Cyclooxygenases, lipoxygenases, and epoxygenases in CNS: their role and involvement in neurological disorders

    Brain Res. Brain Res. Rev.

    (2006)
  • E. Rousselet et al.

    Role of TNF-alpha receptors in mice intoxicated with the parkinsonian toxin MPTP

    Exp. Neurol.

    (2002)
  • J.K. Ryu et al.

    Minocycline or iNOS inhibition block 3-nitrotyrosine increases and blood–brain barrier leakiness in amyloid beta-peptide-injected rat hippocampus

    Exp. Neurol.

    (2006)
  • S. Sugama et al.

    Age-related microglial activation in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced dopaminergic neurodegeneration in C57BL/6 mice

    Brain Res.

    (2003)
  • K. Wakayama et al.

    Localization of norepinephrine and serotonin transporter in mouse brain capillary endothelial cells

    Neurosci. Res.

    (2002)
  • 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)
  • N.J. Abbott

    Inflammatory mediators and modulation of blood–brain barrier permeability

    Cell. Mol. Neurobiol.

    (2000)
  • M. Aschner

    Astrocytes as mediators of immune and inflammatory responses in the CNS

    Neurotoxicology

    (1998)
  • C. Barcia et al.

    Blood vessels and parkinsonism

    Front. Biosci.

    (2004)
  • Cited by (0)

    View full text