Elsevier

Neuroscience Research

Volume 56, Issue 3, November 2006, Pages 246-252
Neuroscience Research

Functional characterization of the brain-to-blood efflux clearance of human amyloid-β peptide (1–40) across the rat blood–brain barrier

https://doi.org/10.1016/j.neures.2006.07.006Get rights and content

Abstract

The present study sought to characterize the brain-to-blood efflux transport of human amyloid-β peptide (hAβ)(1–40) across the blood–brain barrier (BBB) in rats. We determined the apparent brain-to-blood [125I]hAβ(1–40) efflux clearance in rats and found it to be 11.0 μL/(min g brain). There were no significant gender differences in the apparent brain-to-blood [125I]hAβ(1–40) efflux clearance. The brain-to-blood [125I]hAβ(1–40) efflux transport was significantly inhibited by unlabeled hAβ(1–40) and hAβ(1–42) by 79.1% and 36.4%, respectively, but was not inhibited by hAβ(1–43) and hAβ(40–1), and was significantly facilitated by hAβ(17–40) by 16.0%, which is one of the major proteolytic fragments of hAβ(1–40) generated by the action of Aβ degradation enzymes, such as endothelin-converting enzyme. Pre-administration of human receptor-associated protein, a low-density lipoprotein receptor-related protein (LRP) antagonist, reduced the elimination of [125I]hAβ(1–40) by 20.3%, while quinidine or verapamil, P-glycoprotein (P-gp) inhibitors, did not significantly affect the elimination. Western blot analysis suggested that LRP-1 is expressed in rat brain capillary endothelial cells. In conclusion, the partial contribution of LRP-1 and the minor contribution of P-gp suggest that the hAβ(1–40) elimination from rat brain is mediated by as yet unidentified molecules.

Introduction

An abnormally elevated level of amyloid-β peptide (Aβ) in the brain is one of the prominent features of Alzheimer's disease (AD) (Hardy and Selkoe, 2002). Aβ is a 38–43 amino acid peptide derived from the proteolytic processing of amyloid precursor protein (APP), and Aβ(1–40) is the major component of amyloid plaques in brain. When [125I] human Aβ(1–40) (hAβ(1–40)) was microinjected into the brain, it was eliminated from the brain to the blood via the blood–brain barrier (BBB) (Shibata et al., 2000, Shiiki et al., 2004), which consists of brain capillary endothelial cells (Terasaki and Ohtsuki, 2005). This elimination process could play an important role in preventing the accumulation of hAβ(1–40) in the brain. On the other hand, exogenous [125I]hAβ(1–40) administered to the circulating blood or via the carotid artery was transferred into the brain (Martel et al., 1997, Wengenack et al., 2000, Deane et al., 2003), suggesting that peripheral hAβ(1–40) is also transported into the brain across the BBB. Therefore, both blood-to-brain and brain-to-blood transport determine the net flux of Aβ transport across the BBB and would play an important role in the accumulation of cerebral Aβ.

In vivo cerebral Aβ clearance has been reported to involve a specific receptor (low-density lipoprotein receptor-related protein 1 (LRP-1)) (Shibata et al., 2000) and proteases (neprilysin (NEP) (Iwata et al., 2001), insulin-degrading enzyme (IDE) (Farris et al., 2003) and endothelin-converting enzyme (ECE) (Eckman et al., 2003)). Since the involvement of each of these in cerebral hAβ(1–40) clearance has been examined in different in vivo systems, the contribution that each of these molecules makes to the clearance system is still unclear. Indeed, it has been reported that LRP-1 is involved in hAβ(1–40) efflux transport in the mouse brain using human receptor-associated protein (RAP) (Shibata et al., 2000), while our recent report failed to demonstrate a significant contribution of LRP-1 to hAβ(1–40) efflux transport in the rat brain using rat RAP (Shiiki et al., 2004).

P-glycoprotein (P-gp) is expressed on the luminal membrane of brain capillary endothelial cells (BCEC) and excretes substrates into the circulation. Recent studies have reported that hAβ(1–40) and hAβ(1–42) could be substrates of P-gp (Lam et al., 2001) and the expression levels of P-gp inversely correlate with Aβ deposition in the brain in elderly humans and P-gp knockout (multidrug resistance 1a/b−/−) mice (Vogelgesang et al., 2002, Cirrito et al., 2005). However, P-gp gene deficiency reduced the expression level of LRP-1 in brain capillaries (Cirrito et al., 2005). Furthermore, since P-gp is expressed in the liver which is the major organ responsible for systemic hAβ(1–40) clearance (Ghiso et al., 2004), P-gp gene deficiency is likely to affect systemic hAβ(1–40) clearance. Therefore, it is still unknown whether P-gp directly contributes to brain-to-blood transport of hAβ(1–40) at the BBB in vivo. It is of crucial importance to characterize in vivo BBB Aβ efflux transport and, particularly, the contribution of LRP-1 and/or P-gp as part of the efflux system preventing the accumulation of Aβ in the brain.

The purpose of this study was to clarify the brain-to-blood efflux clearance of hAβ(1–40) by combining brain efflux index (BEI) and brain slice uptake studies. We also investigated the contribution of LRP-1 and P-gp to cerebral Aβ clearance in rats by means of a BEI study.

Section snippets

Animals

Male and female Sprague–Dawley rats (7–8 weeks old) were purchased from Charles River Laboratories (Yokohama, Japan). All experiments were approved by the Animal Care Committee of the Graduate School of Pharmaceutical Sciences, Tohoku University.

Reagents

Monoiodinated and non-oxidized [125I]hAβ(1–40) (2200 Ci/mmol) was purchased from Perkin-Elmer Life Sciences (Boston, MA, USA). [3H]Dextran was obtained from American Radiolabeled Chemicals (St. Louis, MO, USA). [carboxy-14C]inulin ([14C]inulin, 1.92 

The apparent brain-to-blood efflux clearance of [125I]hAβ(1–40) in rats

The in vivo brain-to-blood efflux transport of [125I]hAβ(1–40) was determined using the BEI method with [3H]dextran as a reference compound. [125I]hAβ(1–40) was injected into rat brain at a concentration of 18.2 nM in the injectate (0.6 nM as the cerebral concentration), which was lower than the half saturation concentration of [125I]hAβ(1–40) elimination we have reported (247 nM in injectate and 8.15 nM as cerebral concentration; Shiiki et al., 2004). The microinjected [125I]hAβ(1–40) was

Discussion

The present study shows, firstly, that the apparent brain-to-blood efflux clearance of hAβ(1–40) from the male rat cortex is 11.0 μL/(min g brain) using a combination of BEI and brain slice uptake studies. The elimination of hAβ(1–40) from rat brain consists of partial LRP-1-mediated transport and P-gp does not significantly contribute to the elimination.

The microinjected [125I]hAβ(1–40) was eliminated from the rat brain in a time-dependent manner. In the BEI study, [3H]mannitol (MW 182), [14

Acknowledgements

The authors would like to thank Drs. T. Iwatsubo and T. Hashimoto for valuable discussions, and Ms. N. Funayama for secretarial assistance. This study was supported in part by a Grant-in-Aid for Scientific Research on Priority Areas 17025005 from The Ministry of Education, Culture, Sports, Science and Technology (MEXT), Japan, and a 21st Century Center of Excellence (COE) Program grant from the Japan Society for the Promotion of Science.

References (43)

  • K. Schuessel et al.

    Impaired Cu/Zn-SOD activity contributes to increased oxidative damage in APP transgenic mice

    Neurobiol. Dis.

    (2005)
  • T. Terasaki et al.

    Brain-to-blood transporters for endogenous substrates and xenobiotics at the blood-brain barrier: an overview of biology and methodology

    NeuroRx

    (2005)
  • J. Wang et al.

    Gender differences in the amount and deposition of amyloid β in APPswe and PS1 double transgenic mice

    Neurobiol. Dis.

    (2003)
  • K. Andersen et al.

    Gender differences in the incidence of AD and vascular dementia: The EURODEM Studies. EURODEM Incidence Research Group

    Neurology

    (1999)
  • T.A. Bayer et al.

    Dietary Cu stabilizes brain superoxide dismutase 1 activity and reduces amyloid Aβ production in APP23 transgenic mice

    Proc. Natl. Acad. Sci. U.S.A.

    (2003)
  • J.R. Cirrito et al.

    In vivo assessment of brain interstitial fluid with microdialysis reveals plaque-associated changes in amyloid-β metabolism and half-life

    J. Neurosci.

    (2003)
  • J.R. Cirrito et al.

    P-glycoprotein deficiency at the blood–brain barrier increases amyloid-β deposition in an Alzheimer disease mouse model

    J. Clin. Invest.

    (2005)
  • R. Deane et al.

    RAGE mediates amyloid-β peptide transport across the blood–brain barrier and accumulation in brain

    Nat. Med.

    (2003)
  • R.B. DeMattos et al.

    Plaque-associated disruption of CSF and plasma amyloid-beta (Abeta) equilibrium in a mouse model of Alzheimer's disease

    J. Neurochem.

    (2002)
  • W. Farris et al.

    Insulin-degrading enzyme regulates the levels of insulin, amyloid β protein, and the beta-amyloid precursor protein intracellular domain in vivo

    Proc. Natl. Acad. Sci. U.S.A.

    (2003)
  • L. Fratiglioni et al.

    Very old women at highest risk of dementia and Alzheimer's disease: incidence data from the Kungsholmen Project, Stockholm

    Neurology

    (1997)
  • Cited by (111)

    • The role of brain barriers in the neurokinetics and pharmacodynamics of lithium

      2021, Pharmacological Research
      Citation Excerpt :

      This is consistent with our recent work showing that 10 mM LiCl for 6 days did not increase the gene expression of P-gp (MDR1/ABCB1) in hCMEC/D3 cells [118]. Interestingly β-amyloid peptide was demonstrated as being cleared from the brain by some ABC transporters expressed in the BBB such as ABCB1, ABCG2 and ABCG4 [139–141]. Thus, Li therapy may alter β-amyloid clearance by modulating ABC transporter activity.

    View all citing articles on Scopus
    View full text