Elsevier

Experimental Neurology

Volume 248, October 2013, Pages 16-29
Experimental Neurology

Amyloid-β plaque reduction, endogenous antibody delivery and glial activation by brain-targeted, transcranial focused ultrasound

https://doi.org/10.1016/j.expneurol.2013.05.008Get rights and content

Highlights

  • Focused ultrasound treatment reduces amyloid-β plaque pathology within 4 days.

  • Host immunoglobulins enter the ultrasound-treated cortex where they bind to plaques.

  • Microglia and astrocyte markers are increased in the ultrasound-treated cortex.

  • Volumes of microglia and astrocytes are increased in the ultrasound-treated cortex.

  • Aβ within microglia and astrocytes is increased in the ultrasound-treated cortex.

Abstract

Noninvasive, targeted drug delivery to the brain can be achieved using transcranial focused ultrasound (FUS), which transiently increases the permeability of the blood–brain barrier (BBB) for localized delivery of therapeutics from the blood to the brain. Previously, we have demonstrated that FUS can deliver intravenously-administered antibodies to the brain of a mouse model of Alzheimer's disease (AD) and rapidly reduce plaques composed of amyloid-β peptides (Aβ). Here, we investigated two potential effects of transcranial FUS itself that could contribute to a reduction of plaque pathology, namely the delivery of endogenous antibodies to the brain and the activation of glial cells.

We demonstrate that transcranial FUS application leads to a significant reduction in plaque burden four days after a single treatment in the TgCRND8 mouse model of AD and that endogenous antibodies are found bound to Aβ plaques. Immunohistochemical and western blot analyses showed an increase in endogenous immunoglobulins within the FUS-targeted cortex. Subsequently, microglia and astrocytes in FUS-treated cortical regions show signs of activation through increases in protein expression and changes in glial size, without changes in glial cell numbers. Enhanced activation of glia correlated with increased internalization of Aβ in microglia and astrocytes.

Together these data demonstrate that FUS improved the bioavailability of endogenous antibodies and led to a temporal activation of glial cells, providing evidence towards antibody- and glia-dependent mechanisms of FUS-mediated plaque reduction.

Introduction

The blood–brain barrier (BBB) poses a challenge for the delivery of therapeutics to the brain for treatment of neurological diseases. Chemicals administered intravenously can facilitate the passage of therapeutics from the blood to the brain but produce variability in the extent and duration of BBB opening (Joshi et al., 2010, Patel et al., 2009, Salahuddin et al., 1988). Ideally, only the brain areas affected by disease would be targeted for treatment, minimizing BBB disruption in other brain regions.

In Alzheimer's disease (AD), amyloid-β peptides (Aβ) aggregate and form extracellular plaques. Animal studies delivering anti-Aβ antibodies directly to the cortex have demonstrated a rapid therapeutic response but employed invasive surgical techniques (Kotilinek et al., 2002, Wilcock et al., 2003). The use of transcranial focused ultrasound (FUS) guided by magnetic resonance imaging (MRI) to locally increase the permeability of the BBB has several advantages, including non-surgical application, targeting of specific brain regions, and control of the extent of BBB opening without damaging surrounding tissues when combined with an intravenous injection of microbubbles (Hynynen et al., 2001, Hynynen et al., 2005, McDannold et al., 2005, Sheikov et al., 2004). We previously demonstrated that MRI-guided FUS (MRIgFUS) efficiently delivered systemically administered anti-Aβ antibodies to targeted brain regions of the TgCRND8 mouse model of AD, reducing plaque load within 4 days (Jordão et al., 2010).

Here, we hypothesize that MRIgFUS alone reduces Aβ pathology, considering that it promotes the entry of monomeric endogenous antibodies (Raymond et al., 2008, Sheikov et al., 2004). Previous studies have shown that endogenous antibodies present in the blood can bind to and disaggregate Aβ fibrils (Dodel et al., 2002, Du et al., 2003, Hyman et al., 2001). We first detected the entry of endogenous antibodies at the site of cortical Aβ plaques in MRIgFUS-treated TgCRND8 mice. Then, we investigated whether MRIgFUS allowed pentameric endogenous immunoglobulin M (IgM, ~ 900 kDa), in addition to monomeric immunoglobulin G (IgG, ~ 150 kDa), to pass from the blood to the brain in TgCRND8 and non-transgenic (non-Tg) mice. Finally, we evaluated whether MRI detection of FUS-mediated changes in BBB permeability can predict the amount of endogenous immunoglobulin entering the brain.

In addition, glia have been implicated in the mechanism of antibody-mediated Aβ clearance (Bard et al., 2000, Koenigsknecht-Talboo et al., 2008, Kraft, 2013, Magga et al., 2010, Nicoll et al., 2006, Wilcock et al., 2003, Wilcock et al., 2004). Therefore, we investigated whether MRIgFUS-enhanced BBB permeability activates microglia and astrocytes, and whether these glia contain Aβ, which would suggest their contribution to Aβ internalization and clearance. Glial activation can be characterized by an increase in the expression of certain proteins, such as ionized calcium-binding adaptor molecule 1 (Iba1) in phagocytic microglia (Ito et al., 1998, Ito et al., 2001) and glial fibrillary acidic protein (GFAP) in astrocytes (Pekny and Nilsson, 2005). Activated glial cells undergo morphological changes, including increased volume and surface area. We aimed to establish the glial activation response to MRIgFUS, and the potential role of glia in MRIgFUS-mediated plaque reduction. The temporal response of glial activation following MRIgFUS was characterized using Iba1 and GFAP expression in the cortex of TgCRND8 and non-Tg mice. Changes in glial volume and area, in addition to Aβ internalization by glia were also investigated.

MRIgFUS technology represents a major advance in the field of non-invasive drug delivery to the brain. For validation of this delivery technique for a wide range of applications, establishing the effects of MRIgFUS in animal models under normal and diseased conditions is important. Here, we show that under disease conditions, MRIgFUS alone reduces Aβ plaque load in the targeted cortex of TgCRND8 mice. Additionally, in both TgCRND8 and non-Tg mice, MRIgFUS delivers endogenous antibodies to the brain and activates glial cells.

Section snippets

Animals

Four month-old male and female non-Tg and TgCRND8 mice (Chishti et al., 2001) were used in this study. Mice at this age were chosen because they exhibit abundant plaque load and match our previous study (Jordão et al., 2010), for comparison purposes. Tissue for western blot analyses was collected from mice sacrificed at 4 h (n = 7 for non-Tg; n = 6 for TgCRND8 mice), 4 days (n = 6 for non-Tg; n = 7 for TgCRND8 mice) and 15 days (n = 7 for non-Tg and n = 7 for TgCRND8 mice).

A separate cohort of mice was

Plaque burden is reduced in cortical brain regions targeted with focused ultrasound

TgCRND8 mice treated with MRIgFUS along the right cortex were sacrificed at 4 days post-treatment. Brains were sectioned and stained for plaques with an anti-Aβ antibody specific for the N-terminus (residues 8–17). Contours were drawn outlining the MRIgFUS-targeted cortical region (Fig. 2A, right) and an equivalent region on the contralateral side (Fig. 2A, left). Plaques were quantified at 40 × magnification within each of these contours. After a single treatment, plaque size (Fig. 2B) and total

Discussion

MRIgFUS is a promising strategy to enhance the delivery of therapeutics to the brain for treatment of neurological disorders (Burgess et al., 2011b, Burgess et al., 2012, Huang et al., 2012, Jordão et al., 2010, Kinoshita et al., 2006b, Raymond et al., 2008, Thévenot et al., 2012, Treat et al., 2007). Although disrupting the BBB can allow potentially harmful cells or toxins into the brain (Miller, 2002), unlike osmotic or chemical BBB disruption, MRIgFUS can target permeabilization to

Conclusions

Our study demonstrates that Aβ plaque size is significantly reduced within 4 days of treating TgCRND8 mice, using MRIgFUS as the only external therapeutic intervention. At the same time, MRIgFUS increases endogenous immunoglobulin levels and glial activation. The decoration of Aβ plaques with IgG and IgM in the MRIgFUS-treated cortex, when plaque load was reduced, suggests a potential role for endogenous antibodies in the mechanism of MRIgFUS-mediated amelioration of Aβ pathology. Furthermore,

Disclosure statement

Dr. Kullervo Hynynen has stock in FUS Instruments, from which he receives non-study related support.

Acknowledgments

We thank Drs. Paul Fraser, David Westaway, and Peter St George-Hyslop for their contributions in creating the TgCRND8 mice. The authors are grateful to Dr. Stefanovic for the use of Imaris software for the glial analysis. We are also grateful to Nicholas Ellens and Ping Wu for their technical assistance with MRIgFUS experiments. Shawna Rideout-Gros, Alex Garces, and Stephanie Bell helped with the animal care. We thank Rosemary Ahrens and Mary Hill for the genotyping and animal care. Funding was

References (62)

  • M.A. O'Reilly et al.

    Focused-ultrasound disruption of the blood–brain barrier using closely-timed short pulses: influence of sonication parameters and injection rate

    Ultrasound Med. Biol.

    (2011)
  • N. Sheikov et al.

    Cellular mechanisms of the blood–brain barrier opening induced by ultrasound in presence of microbubbles

    Ultrasound Med. Biol.

    (2004)
  • N. Sheikov et al.

    Effect of focused ultrasound applied with an ultrasound contrast agent on the tight junctional integrity of the brain microvascular endothelium

    Ultrasound Med. Biol.

    (2008)
  • R.A. Adams

    The fibrin-derived gamma377-395 peptide inhibits microglia activation and suppresses relapsing paralysis in central nervous system autoimmune disease

    J. Exp. Med.

    (2007)
  • B.J. Bacskai

    Imaging of amyloid-beta deposits in brains of living mice permits direct observation of clearance of plaques with immunotherapy

    Nat. Med.

    (2001)
  • F. Bard

    Peripherally administered antibodies against amyloid beta-peptide enter the central nervous system and reduce pathology in a mouse model of Alzheimer disease

    Nat. Med.

    (2000)
  • A. Burgess et al.

    Focused ultrasound: crossing barriers to treat Alzheimer's disease

    Ther. Deliv.

    (2011)
  • A. Burgess et al.

    Targeted delivery of neural stem cells to the brain using MRI-guided focused ultrasound to disrupt the blood–brain barrier

    PLoS One

    (2011)
  • K.S. Chen

    Neurodegenerative Alzheimer-like pathology in PDAPP 717V→F transgenic mice

    Prog. Brain Res

    (1998)
  • E.E. Cho et al.

    Two-photon fluorescence microscopy study of cerebrovascular dynamics in ultrasound-induced blood–brain barrier opening

    J. Cereb. Blood Flow Metab.

    (2011)
  • J.J. Choi et al.

    Noninvasive and localized blood–brain barrier disruption using focused ultrasound can be achieved at short pulse lengths and low pulse repetition frequencies

    J. Cereb. Blood Flow Metab.

    (2011)
  • R. Chopra et al.

    An MRI-compatible system for focused ultrasound experiments in small animal models

    Med. Phys.

    (2009)
  • R. Deane

    IgG-assisted age-dependent clearance of Alzheimer's amyloid beta peptide by the blood–brain barrier neonatal Fc receptor

    J. Neurosci.

    (2005)
  • R.B. DeMattos et al.

    Peripheral anti-A beta antibody alters CNS and plasma A beta clearance and decreases brain A beta burden in a mouse model of Alzheimer's disease

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

    (2001)
  • J. Deng

    The role of caveolin-1 in blood–brain barrier disruption induced by focused ultrasound combined with microbubbles

    J. Mol. Neurosci.

    (2012)
  • R. Dodel

    Human antibodies against amyloid beta peptide: a potential treatment for Alzheimer's disease

    Ann. Neurol.

    (2002)
  • Y. Du

    Human anti-beta-amyloid antibodies block beta-amyloid fibril formation and prevent beta-amyloid-induced neurotoxicity

    Brain

    (2003)
  • S.A. Frautschy et al.

    Microglial response to amyloid plaques in APPsw transgenic mice

    Am. J. Pathol.

    (1998)
  • C.A. Hawkes et al.

    In vivo uptake of beta-amyloid by non-plaque associated microglia

    Curr. Alzheimer Res.

    (2012)
  • B.T. Hyman et al.

    Autoantibodies to amyloid-beta and Alzheimer's disease

    Ann. Neurol.

    (2001)
  • K. Hynynen et al.

    Noninvasive MR imaging-guided focal opening of the blood–brain barrier in rabbits

    Radiology

    (2001)
  • Cited by (251)

    View all citing articles on Scopus
    View full text