You are on page 1of 8

ARTICLES

PUBLISHED ONLINE: 22 DECEMBER 2014 | DOI: 10.1038/NNANO.2014.254

Towards non-invasive diagnostic imaging


of early-stage Alzheimer’s disease
Kirsten L. Viola1†, James Sbarboro1†, Ruchi Sureka1†, Mrinmoy De2, Maíra A. Bicca1,3, Jane Wang1,
Shaleen Vasavada1, Sreyesh Satpathy4, Summer Wu4, Hrushikesh Joshi2, Pauline T. Velasco1,
Keith MacRenaris5, E. Alex Waters5, Chang Lu1, Joseph Phan1, Pascale Lacor1, Pottumarthi Prasad6,
Vinayak P. Dravid2,7* and William L. Klein1,7*

One way to image the molecular pathology in Alzheimer’s disease is by positron emission tomography using probes that
target amyloid fibrils. However, these fibrils are not closely linked to the development of the disease. It is now thought
that early-stage biomarkers that instigate memory loss are composed of Aβ oligomers. Here, we report a sensitive
molecular magnetic resonance imaging contrast probe that is specific for Aβ oligomers. We attach oligomer-specific
antibodies onto magnetic nanostructures and show that the complex is stable and binds to Aβ oligomers on cells and brain
tissues to give a magnetic resonance imaging signal. When intranasally administered to an Alzheimer’s disease mouse
model, the probe readily reached hippocampal Aβ oligomers. In isolated samples of human brain tissue, we observed a
magnetic resonance imaging signal that distinguished Alzheimer’s disease from controls. Such nanostructures that target
neurotoxic Aβ oligomers are potentially useful for evaluating the efficacy of new drugs and ultimately for early-stage
Alzheimer’s disease diagnosis and disease management.

A
lzheimer’s disease affects one in nine people over the age of including via the blood stream, spinal fluid and nasal passageways.
65 years1. Despite the great personal and economic toll, pro- Superparamagnetic MNSs generate T2* contrast on MR images,
gress in developing effective treatments remains slow. A sig- because they create local magnetic field inhomogeneities, providing
nificant factor is the lack of powerful diagnostic methods. a negative contrast compared to background16. The colloidal form
Cerebrospinal fluid (CSF) assays show promise2,3, but spinal taps of superparamagnetic MNSs also facilitates effective surface functio-
are invasive and assays of CSF analytes present challenges in nalization for targeting moieties such as antibodies and fluorescent
terms of accuracy and reliable disease-state discrimination4. A tags for multimodal imaging and cross-validation of targeting.
promising alternative diagnostic strategy is the detection of Conjugation of MNSs to target-specific antibodies has been shown
Alzheimer’s disease pathology using targeted brain imaging. The to be effective in providing T2-weighted MRI of cellular surface pro-
introduction of positron emission tomography (PET) probes for teins and localized tumours17, including those in the central nervous
amyloid plaques5 has been a great technical advance, establishing system (CNS)18.
the precedent that brain molecular imaging could become a signifi- The present work takes advantage of the AβO-specific anti-
cant tool for diagnostics and drug development6. It is known, bodies19 that have been introduced to study disease mechanisms
however, that amyloid plaques do not correlate well with (for a review see ref. 20) and that are now under development for
Alzheimer’s disease dementia7 and are not present in the earliest Alzheimer’s disease therapeutics21,22. Because a successful MRI
stages of the disease8. Probes for alternative markers, especially for probe using MNSs requires aqueous stabilization of the nanostruc-
the earliest stage of Alzheimer’s disease, are needed for effective ture and efficient antibody conjugation, we produced mono-dis-
disease intervention and management. persed, nitro-dopamine (nDOPA) and polyethylene glycol (PEG)
Studies over the past 15 years have suggested that pathogenic stabilized 12–16 nm MNSs. These carboxylate-functionalized
amyloid beta oligomers (AβOs) provide a more appropriate bio- MNSs conjugate efficiently to AβO-specific antibodies. MNS–anti-
marker than plaques. AβOs cause the synapse failure regarded as body conjugates detect Alzheimer’s disease-causing toxic oligomers
responsible for Alzheimer’s disease memory loss (reviewed in on nerve cell surfaces in vitro, and they quickly reach oligomers
refs 9–13), and they appear early in the disease8,14. Although AβOs in vivo, in a mouse model, following intranasal delivery. Prototype
are appealing targets for molecular magnetic resonance imaging imaging of the mouse model and isolated human brain tissue sub-
(MRI), a specific high-affinity contrast probe needs to be developed. stantiates the clinical potential of MRI of synaptotoxic oligomers
In this context, magnetic nanostructures (MNSs) provide an using targeted nanostructure probes.
excellent, biocompatible contrast platform for targeted MRI15.
MNSs with a diameter below ∼20 nm exhibit superparamagnetism, AβOs and synthesis of a targeted MRI probe
lack permanent magnetization at room temperature, and have AβOs are potent neurotoxins that accumulate in the CNS of humans
excellent colloidal stability for both localized and systemic delivery, with Alzheimer’s disease14,23,24 and in transgenic (Tg) rodent

1
Department of Neurobiology, Northwestern University, Evanston, Illinois 60208, USA. 2 Department of Materials Science and Engineering, Northwestern
University, Evanston, Illinois 60208, USA. 3 Department of Pharmacology, Universidade Federal de Santa Catarina, Santa Catarina 88049900 Brazil. 4 Illinois
Math & Science Academy, Aurora, llinois 60506, USA. 5 Center for Advanced Molecular Imaging, Northwestern University, Evanston, Illinois 60208, USA.
6
Department of Radiology, NorthShore University HealthSystems, Evanston, Illinois 60201, USA. 7 International Institute for Nanotechnology (IIN), Northwestern
University, Evanston, Illinois 60208, USA. †These authors contributed equally to this work. * e-mail: wklein@northwestern.edu; v-dravid@northwestern.edu

NATURE NANOTECHNOLOGY | ADVANCE ONLINE PUBLICATION | www.nature.com/naturenanotechnology 1

© 2014 Macmillan Publishers Limited. All rights reserved.


ARTICLES NATURE NANOTECHNOLOGY DOI: 10.1038/NNANO.2014.254

a
150

125

Intensity of FAM-AβOs (a.u.)


100

75
5 25 75 100 150 200 500

50

25
10 μm
0
0 60 120 180 240 300 360 420 480 540
AβOs (nM)

b 5xFAD EntCx

568-NU4 ThS Merge

5xFAD Cb wt EntCx wt Cb

25 μm

Figure 1 | Aβ oligomers (AβOs) bind to neuronal surfaces in a saturable, receptor-mediated manner and are distinct from amyloid plaques. a, Binding of
FAM-AβOs to neuronal surfaces is saturable. AβO binding to primary hippocampal cells was measured using increasing doses of FAM-AβOs (representative
cell images are shown in the inset). The numbers in the inset and along the x axis of the graph correspond to Aβ monomer equivalent concentrations
(in nM). Total fluorescence per 100 µm of neuronal process is shown as a function of AβO concentration. The data were fit to a sigmoidal dose–response
curve with an EC50 value of ∼75 nM. Saturation is consistent with receptor-mediated binding. Each point on the graph represents the mean ± s.e.m. for
counts obtained from five separate images. b, Sagittal brain sections, 50 µm thick, from eight-month-old 5xFAD and wild-type (wt) mice were probed with
568-NU4 and counterstained with thioflavin S (ThioS). Images were captured from multiple regions of the brain. Top row: 5xFAD cortical region stained with
NU4 (red), ThioS (green) and merged image (right). Bottom row: Merged images from 5xFAD cerebellum (left), wild-type cortex (centre) and wild-type
cerebellum (right). The findings demonstrate that NU4 labelling is often associated with, yet distinct from, amyloid plaques. NU4 labelling is more abundant
than the ThioS staining. The data support the notion that NU4 thus affords an excellent targeting antibody for the development of an AβO-specific MRI
probe that is distinct from currently available plaque probes.

Alzheimer’s disease models25–27 and are generally accepted as an binding of AβOs suggests their potential as a target for a suitable
early event in Alzheimer’s disease pathogenesis28–31. The usefulness MRI–antibody probe.
of targeting AβOs as an early Alzheimer’s disease biomarker is To target the MNSs we selected a monoclonal antibody (NU4)
suggested by human neuropathology studies in which AβOs that has been shown to recognize AβOs with high affinity
initially appear bound to discrete neurons, localizing to synapses in vitro19,32 as well as oligomer pathology in vivo22. To ensure that
in dendritic arbours14 through putative association with clustered the NU4 antibody specifically detects AβOs, well-differentiated hip-
cell surface receptors11 (Fig. 1a). Here, exogenous, fluorescently pocampal cells were incubated with FAM-AβOs (as shown in Fig. 1)
labelled AβOs (FAM-AβOs) were added to well-differentiated rat and probed with fluorescently tagged NU4 (633-NU4). The results
hippocampal neuron cultures, a synapse-forming model that is show a strong association of 633-NU4 with the FAM-AβOs bound
further used in characterizing the AβO MRI probe. FAM-AβOs to the hippocampal neurons (Supplementary Fig. 1). We then com-
bound at discrete sites on dendrites, showing saturable, pared the pathology detected with NU4 to that recognized by a
concentration-dependent synaptic binding. The density and common marker for amyloid plaques, thioflavin S (ThioS).
fluorescence intensity of the dendritic puncta increased as the Sagittal brain sections from the 5xFAD Tg Alzheimer’s disease
AβO concentration rose to ∼150 nM (Aβ monomer-equivalent), mouse model, and age-matched wild-type control mice were labelled
above which both measures plateaued. Curve fitting indicated a Kd with fluorescently tagged NU4 (568-NU4) followed by counterstain
value of ∼75 nM. The discrete and saturable cell-surface with ThioS. The 5xFAD mouse is a model for Alzheimer’s

2 NATURE NANOTECHNOLOGY | ADVANCE ONLINE PUBLICATION | www.nature.com/naturenanotechnology

© 2014 Macmillan Publishers Limited. All rights reserved.


NATURE NANOTECHNOLOGY DOI: 10.1038/NNANO.2014.254 ARTICLES
a H c
O N O OH
O O 80
n
O O
O NO2
60

1/T2 (s−1)
40

20

0.00 0.05 0.10 0.15 0.20


Concentration of Fe (mM)
20 nm

Figure 2 | Individual components of the NU4MNS probe. MNSs are ligand-stabilized and uniform in size. a, Structure of a carboxylate-terminated ligand-
stabilized MNS. b, TEM of the aqueous stabilized MNSs. c, Relaxivity measurements of MNSs at 1.42 T and 60 Hz. The MNSs are approximately 16 nm in
diameter and show a relaxivity, r2, of 385 s−1 mM−1. The black line and dots are the recorded values and the red line is the trendline.

disease, expressing five Alzheimer’s disease-mutated genes. This The nDOPA is used as an ‘anchoring group’ between the MNS and
model is widely used because it quickly develops Alzheimer’s PEG, leaving no free nDOPA in the solution or associated with the
disease characteristics33. In the entorhinal cortex, 568-NU4 detected probe. Moreover, nDOPA has low agonist efficacy41.
prominent diffuse pathology, sometimes presenting as a halo sur- The stabilization method involves monolayer formation of a PEG
rounding the ThioS-positive plaque cores and frequently appearing ligand on the MNS surfaces, and the ligand was terminated with a
as diffuse structures lacking a core (Fig. 1b). Differences were even carboxylate functionality for conjugation with antibodies (Fig. 2a).
more pronounced in the hippocampus. No signal was observed in The nDOPA MNSs were stable over a month in physiological
the cerebellum or in wild-type controls. Oligomer-selective NU4 buffers (Supplementary Fig. 2). No agglomeration was observed in
thus targets a pathology that is distinct from the plaque cores the stable colloidal suspension by either transmission electron
detected by ThioS. microscopy (TEM, Fig. 2b) or dynamic light scattering (DLS,
A probe for human use must offer sufficient signal sensitivity, Supplementary Fig. 2b). The zeta potential and DLS measurements
stability and biocompatibility, as well as high selectivity and specific of the 16 nm particles showed a surface potential of approximately
affinity for relevant targets of interest. Here, we use 12–16 nm Fe3O4 –40 mV and a nominal hydrodynamic radius of 30 nm, indicating
MNSs as the MRI contrast platform to which specific AβO antibodies excellent stability and colloidal dispersion in aqueous solutions
are conjugated. The MNSs were synthesized following high-tempera- (Supplementary Fig. 2). In fact, the MNS solution is sufficiently col-
ture thermal decomposition of an iron–oleate complex as reported loidal to be run through an agarose gel, similar to gold nanoparticles
earlier34. The MNSs prepared by this method exhibit very high (Supplementary Fig. 2a).
monodispersity and enhanced superparamagnetic properties. The MR relaxivity of the PEG-conjugated MNS was measured
However, as the surface is coated with hydrophobic ligands that using a Bruker minispec 60 MHz (1.41 T) relaxometer. We observed
oppose biological applications, additional modification is required. a very high r2 value of 385 s−1 mM−1, significantly higher than demon-
Previous methods have involved MNSs that were either stabilized strated by commercially available ferumoxytol (dextran-coated iron
by surface coating with polymer and inorganic layers or by micelle- oxide, 80 s−1 mM−1) and ferumoxide (silica-coated iron oxide,
like bilayer formation15,35, but maintaining the shell thickness and 120 s−1 mM−1) (Fig. 2c). We attribute the enhanced r2 to the increase
consistent functional group on the surface is difficult. Moreover, in hydrophilic surfaces with carboxylate and glycol ligand tailoring,
as the MRI efficiency depends on water diffusion in proximity to which interact strongly to influence the surrounding proton relax-
the magnetite core, the surface coating interferes with imaging36. ation39. The potential toxicity of the MNSs was assessed in primary
Similar problems also arise in the case of micelle-like bilayer stabil- hippocampal cells and four different mammalian cell lines. We
ization. Stability can also be an issue for complex biological observed that cell viability was conserved after 24 h incubation with
environments as it relies upon weak hydrophobic interactions37. a high concentration of MNS (1 mM Fe) (Supplementary Fig. 2d).
By comparison, single-layer ligand stabilization by place exchange/ The buffer-stabilized MNSs were targeted to AβOs by attaching
removal of a hydrophobic ligand from the MNS surface can enable fine- them to the NU4 antibody. Coupling was by carboxyl–amine con-
tuning of stability, functionality and overall proton diffusion closest to jugation using 1-ethyl-3-(3-dimethylaminopropyl)-carbodiimide
where its magnetic influence is strongest to enhance its magnetic reson- (EDC). The resulting probe, MNS-conjugated to NU4 (NU4MNS),
ance properties. Previous use of dopamine, phosphate and amine to was magnetically separated from free residual antibodies and
stabilize the surface ligand38 suffered from stability challenges in stored in phosphate-buffered saline (PBS) at pH 7.2. NU4MNS
complex biological environments. nDOPA has a better affinity for the remained homogeneous and colloidal in PBS for at least 3 h at
iron oxide surface and higher stability. We therefore used nDOPA as room temperature, ample time to allow administration.
the surface anchor group and PEG as a spacer. PEG improves the con- Additionally, the NU4MNS could be readily redispersed to a homo-
trast efficiency by enhancing water diffusion in close proximity to the geneous colloidal suspension with brief sonication, even several
nanocrystal core, provides appropriate biocompatibility, improves col- weeks after preparation.
loidal stability and retains desired magnetic properties such as high r2
relaxivity for T2-weighted magnetic resonance contrast39. It is expected NU4MNS probe detects AβOs in cells and human tissue
that the nDOPA will have no effect on the dopamine receptors of the To verify that the NU4MNS probe maintained the high affinity and
brain as it is sequestered in the form of a self-assembled monolayer40. specificity of the parent antibody, using fluorescence microscopy we

NATURE NANOTECHNOLOGY | ADVANCE ONLINE PUBLICATION | www.nature.com/naturenanotechnology 3

© 2014 Macmillan Publishers Limited. All rights reserved.


ARTICLES NATURE NANOTECHNOLOGY DOI: 10.1038/NNANO.2014.254

FAM-AβOs + NU4MNS Vehicle + NU4MNS


0.12

Pixel area count/length


0.08

0.04
NU4MNS
NU4
0.00
0 5 10 15 20
Concentration (μg ml−1)
40 μm

Figure 3 | NU4MNS attachment to hippocampal neurons is specific to AβOs. Attachment of NU4MNS to hippocampal neurons is stringently
AβO-dependent. Left and middle: Hippocampal cells, treated with FAM-AβOs or vehicle control, were probed with fluorescent NU4MNS. The data show that
NU4MNS is highly AβO-specific and AβO-dependent. (FAM-AβOs, green; NU4MNS, red; co-localization, yellow). Right: Dose curves of antibody (NU4) and
NU4MNS detection of AβOs shows that NU4 conjugation to MNS causes only minor loss of affinity for AβOs, if at all. Each point on the graph represents
the mean ± s.e.m. for counts obtained from five separate images.

compared AβO detection by NU4MNS with NU4. Mature cultures no binding by NU4MNS (Fig. 3, left), establishing that the high
of hippocampal cells were incubated with FAM-AβOs and probed specificity of NU4 for AβOs was retained following conjugation to
with NU4MNS (Fig. 3, left) or NU4 (Supplementary Fig. 3a) and MNSs. Experiments using immunoprecipitation further confirmed
imaged using a fluorescent secondary antibody. The efficacy of the that the NU4MNS probe retained its specificity for AβOs in solution
NU4MNS probe for binding AβOs was determined by measuring (Supplementary Fig. 3b,c). Immunoprecipitation of AβOs was not due
the percentage of co-localization between FAM-AβOs and NU4 to non-specific adsorption, as non-immune control IgGMNS showed
or NU4MNS. Quantification of fluorescence intensity established no isolation of AβOs from a 200 nM solution.
that NU4MNS showed >90% co-localization with AβOs bound to To determine the impact of the conjugation procedure on the
hippocampal neurons. Cells incubated with vehicle showed virtually affinity of the probe for AβOs, hippocampal cultures were treated
with FAM-AβOs, then probed with increasing concentrations of
Probable AD Aged control NU4 or NU4MNS (0–20 µg ml−1 antibody concentration). Both
NU4 and NU4MNS showed a high affinity for oligomers, demon-
strating saturable binding with similar doses needed for half-
maximal binding (1–2 µg ml−1) (Fig. 3, right). We next tested the
prediction that the probe would retain the specificity of the parent
NU4

NU4 for Alzheimer’s disease brain tissue relative to control brain


tissue. The first experiment compared binding of NU4MNS to cor-
100µm
100 µm tical slices from human Alzheimer’s disease and age-matched non-
demented (control) patients by immunofluorescence microscopy
(Fig. 4). Floating human brain sections were labelled using
633-NU4, 633-NU4MNS or 633-IgGMNS. Sections were imaged
at ×10 with an epifluorescent microscope. Immunofluorescent
NU4MNS

signal was detected in the probable Alzheimer’s disease slices and


minimally present in the aged control patients. 633-NU4 and
633-NU4MNS both detected diffuse plaque-like structures as well
as smaller clusters of AβOs in Alzheimer’s disease brain tissue,
consistent with 5xFAD results (Fig. 1b) and with previous studies
of AβO neuropathology14,19,24. Diffuse plaques of oligomers in
human samples have been shown previously to be distinct from
and manifest before dense-core amyloid plaques that stain with
congophilic dyes and currently used PET probes5,14,24. Controls
IgGMNS

established that these structures were not detected using MNS con-
jugated to non-specific IgG. The data demonstrate the specificity of
NU4MNS for Alzheimer’s disease neuropathology.

NU4MNS detects Alzheimer’s disease pathology by MRI


Figure 4 | NU4 and NU4MNS discriminate Alzheimer’s disease human Intranasal delivery of NU4 has previously been used to bypass the
frontal cortex sections from aged controls. Human frontal cortex sections blood–brain barrier and bind AβOs in the brain of 5xFAD mice22.
were probed with fluorescently tagged NU4, NU4MNS or control IgGMNS, To confirm that 568-NU4 could detect AβOs in living animals
and imaged. NU4 and NU4MNS detected diffuse plaque-like structures and after intranasal delivery, aged 5xFAD and wild-type littermate
smaller neuronal deposits, consistent with typical Alzheimer’s disease (AD) mice were inoculated with 568-NU4 and allowed to recover for
pathology. These structures were not seen in control brain and were not 4 h. Sagittal sections were mounted for imaging of antibody distri-
detected by the non-specific IgGMNS probe, demonstrating that NU4 is bution. Images show that the 568-NU4 antibody localized through-
capable of discriminating between Alzheimer’s disease and non-demented out the frontal and temporal cortex (not shown) and the
controls, and that this specificity is retained after MNS conjugation. hippocampus (Fig. 5a) of the 5xFAD mice, binding to diffuse

4 NATURE NANOTECHNOLOGY | ADVANCE ONLINE PUBLICATION | www.nature.com/naturenanotechnology

© 2014 Macmillan Publishers Limited. All rights reserved.


NATURE NANOTECHNOLOGY DOI: 10.1038/NNANO.2014.254 ARTICLES
a 5xFAD CA1 5xFAD CA3 wt CA1 wt CA3
SP SP
SP

SP

15 µm

b 5xFAD hipp wt hipp


Single plane Maximum projection
SP

15 µm

Figure 5 | NU4 antibody detects dendrite-bound AβOs in fixed tissue and binds its target within 4 h following intranasal inoculation. a, Mice
(eight-month-old 5xFAD and wild-type littermate controls) were intranasally inoculated with 568-NU4. The probe was allowed to distribute for 4 h before
the animals were killed and their brains removed. Sagittal sections (50 µm thick) were obtained and mounted to slides. Images were captured with an
inverted Nikon TE2000 microscope from multiple regions of the brain. The images show 568-NU4 distribution in the CA1 and CA3 hippocampal regions of
the 5xFAD mouse on the left and wild-type (wt) mouse on the right. Labelling seen in the 5xFAD mouse is punctate and appears to be more prevalently
associated with the somas of the cells. Wild-type controls showed no 568-NU4 probe distribution in the hippocampus. The data confirm that the 568-NU4
antibody readily distributes to its targets within 4 h after intranasal administration. b, Sagittal brain sections (50 µm thick) from eight-month-old 5xFAD and
wild-type mice were probed with 568-NU4. Images were captured with a confocal microscope from multiple regions of the brain. Shown, from left to right, are
a single plane from 5xFAD hippocampus, the maximum projection of all planes at the same location, and the maximum projection of the hippocampal region
from the wild-type mouse. Images show punctate labelling around the cell bodies of the pyramidal cells in the 5xFAD mouse but not the wild-type. The dotted
lines outline the pyramidale cell layer. SP, stratum pyramidale.

pathology and discrete puncta on cells. Wild-type mice showed low by MRI 96 h later to look for changes in the distribution and
levels of fluorescence in the prefrontal cortex (data not shown) com- clearance of the probe (Fig. 6b). The results show a change in the dis-
pared with the 5xFAD mice, and no labelling in the temporal or hip- tribution and a modest reduction of signal intensity in the hippo-
pocampal regions, confirming that the NU4-based probes can be campus of the 5xFAD mouse, but not a complete elimination of the
delivered by intranasal inoculation and specifically bind to the probe. Currently, the data would suggest that the probe remains in
target. For comparison, we examined the detection of AβOs the brain tissue for longer than four days, but the exact length of resi-
exposed to 568-NU4 in vitro. The pattern was similar to that seen dency is not yet certain. The clear difference between the 5xFAD
with in vivo labelling, presenting largely as dispersed puncta mouse and the control was substantiated by a second NU4MNS inocu-
around cell bodies and within the neuropil (Fig. 5b). lation and subsequent MRI (Supplementary Fig. 4c). Importantly,
Having shown that NU4MNS retained specificity for AβOs and animals were monitored for signs of infection or distress at the inocu-
that NU4 can be delivered to its targets by intranasal inoculation, lation sites and in general for more than two months after treatment.
we tested whether the NU4MNS probe offered sufficient contrast No adverse effects or signs of pain or infection from the inoculation
to identify AβO samples by MRI. In a preliminary test, we treated were detected. Overall, the data show that the NU4MNS probe, like
hippocampal neurons as above with AβOs or vehicle, with fluor- the parent antibody, quickly gains access to the hippocampus and
escence verifying the presence or absence of oligomers. Cells were that it can provide a magnetic resonance signal that distinguishes the
then probed with NU4MNS and imaged with a 7 T MRI scanner. 5xFAD mouse Alzheimer’s disease model from controls.
MRI clearly confirmed an AβO-dependent signal, exhibiting more To assess the quality of signal that eventually might be feasible
than 300% signal difference in MRI when AβOs were present under optimal conditions, we examined MRI signals using brain
(Supplementary Fig. 4b). Having confirmed the predicted magnetic sections from human and 5xFAD mice. Brain slices, 50 µm thick,
resonance signal in vitro, we next tested whether an AβO-dependent from eight-month-old 5xFAD and wild-type littermates were
signal could be obtained in vivo. Aged 5xFAD and wild-type mice, probed with 555-NU4MNS using the procedures described in
eight months old, received 5 µg (antibody concentration) Methods. Some sections were imaged for fluorescence to confirm
NU4MNS by intranasal inoculation. Animals were allowed to that the probe properly detected the pathology, while the remaining
recover and the probe to distribute for 4 h before MRI. Images slices were layered in agarose for MRI. Because of the thinness of the
from a 5xFAD mouse (Fig. 6a) and a wild-type littermate show a slices, MRI of the sections in-plane could not be readily obtained.
clear signal in the hippocampus of the mouse Alzheimer’s disease Instead, slices were layered on top of each other in agar, with
model that is absent in the wild-type mouse. Signal was also detect- about 1 cm of agar separating the 5xFAD from the wild-type
able in the cortex (not shown). Following the initial treatment and slices. Slices were imaged cross-sectionally rather than in-plane to
scans, animals were allowed to recover and were then re-examined (1) allow multiple slices to appear in the same field of view for

NATURE NANOTECHNOLOGY | ADVANCE ONLINE PUBLICATION | www.nature.com/naturenanotechnology 5

© 2014 Macmillan Publishers Limited. All rights reserved.


ARTICLES NATURE NANOTECHNOLOGY DOI: 10.1038/NNANO.2014.254

a 5xFAD wt b Day 1 96 h later

5xFAD
wt
c d
Probable AD brain

Tg 5xFAD brain slices Probable AD brain

Non-demented brain

Non-demented brain

Probable AD brain
wt brain slices MNSlgG

Non-demented brain
no treatment

Figure 6 | NU4MNS labelling of human and Tg mouse brain slices gives a pronounced, Alzheimer’s disease-dependent MRI signal that is confirmed
in vivo. a, In vivo imaging of probe distribution in live mice 4 h after intranasal inoculation shows labelling by the probe in the hippocampal region of the
5xFAD mice, but not the wild-type (wt) controls. Boxed regions are shown at higher magnification in b. b, Higher magnification images of the hippocampal
region of the 5xFAD and wild-type mice shows probe distribution 4 h after inoculation and the changes in distribution 96 h later. Although the probe
distribution has changed and decreased, there are still detectable levels of probe present. c, Sagittal sections from 5xFAD (top) and wild-type (bottom)
eight-month-old mice were probed with NU4MNS and layered in agarose for MRI. Owing to the thinness of the sections, it was necessary to capture
cross-sectional images as opposed to traditional ‘whole slice’ images. Using cross-sectional captures, NU4MNS bound to the AβOs in 5xFAD slices
(as seen in Fig. 1b) produces an MRI signal that appears as linear structures. Cross-sectional imaging allowed us to image both 5xFAD and wild-type slices
at the same time, but does not provide any specific localization information. Representative cross-sectional images were chosen for presentation, with slices
outlined by boxes for easier identification. Images show a robust signal in the 5xFAD mice brain sections that is absent from the wild-type littermate
sections. d, Human brain slices were probed with NU4MNS and examined by cross-sectional MRI as described for the mouse brain slices in c. MRI showed
that NU4MNS provided a strong signal in the Alzheimer’s disease (AD) samples. The signal was greatly reduced or absent from the age-matched non-
demented controls. NU4MNS detection of AβOs appeared as linear structures with variable signal intensities along the lines suggesting the non-uniform
distribution of AβOs across the section. No signal was detected when the slices were probed with non-specific mouse IgG attached to MNS (MNSIgG).
Representative cross-sectional images of two separate probable AD and two non-demented patients out of eight samples/condition that were imaged were
chosen for presentation. Taken together, these data provide a proof of concept for NU4MNS as an MRI probe of AβO load in human patients and 5xFAD
Alzheimer’s disease mouse models. Scale bars (a,c,d), 5 mm; (b) 1 mm.

better comparison of signal intensity and (2) address concerns about AβO-containing samples from controls in cells, mouse models
partial volume artefacts that can result from imaging samples that and isolated human tissue. Successful development of the
are thinner than the scan slice depth of the equipment. NU4MNS magnetic resonance probe may have significant value
Although this type of analysis can provide information about the for early Alzheimer’s disease diagnostics42. An AβO-directed diag-
total probe density bound to the tissue, it cannot be used to provide nostic using MRI would provide both structural information and
localization information. Images show a robust, disease-specific molecular-targeted contrast for localization of early AβO accumu-
MRI signal in the 5xFAD brain slices that is not detected in the lation. Such information would be of great value for the develop-
wild-type slices (Fig. 6c). These data further substantiate the ment of effective Alzheimer’s disease therapeutics by providing a
specificity of the probe for Alzheimer’s disease pathology. means to track the efficacy of investigational new drugs that target
We concluded by assessing the potential to obtain a disease-rel- AβOs. Moreover, previous studies with the NU4 antibody have
evant MRI signal from human tissue. Again, because the sections demonstrated a behavioural improvement following NU4
were too thin to be imaged in-plane, the sections were layered in therapy22, suggesting a therapeutic potential for the NU4MNS
agar and cross-sectional images were collected using a 7 T scanner. probe in addition to its diagnostic value. Thus, imaging of AβOs
Untreated sections were used as a control to determine the baseline with non-invasive in vivo modalities such as MRI has the potential
magnetic resonance signal from endogenous iron levels. MRI of to revolutionize Alzheimer’s disease management at its earliest stage
the human brain samples (Fig. 6d) showed that the NU4MNS for therapeutic intervention and overall disease management.
probe provided a robust, Alzheimer’s disease-dependent signal. Although a formal study of the pharmacokinetics of the probe
has not yet been completed, the data presented show that the
Conclusions probe can be introduced intranasally and cross the blood–brain
The results show that an AβO-targeted molecular MRI probe can barrier to distribute to the intended targets within 4 h, as well as
reach targets within the CNS and is capable of differentiating showing significant clearance from the brain within four days

6 NATURE NANOTECHNOLOGY | ADVANCE ONLINE PUBLICATION | www.nature.com/naturenanotechnology

© 2014 Macmillan Publishers Limited. All rights reserved.


NATURE NANOTECHNOLOGY DOI: 10.1038/NNANO.2014.254 ARTICLES
after introduction. It has been shown by Xiao et al.22 that fluorophore/NU4 was ∼1.0–2.5 mol fluorophore/mol antibody for fluorescent-
HRP-NU4 antibody is distributed throughout the brain following NU4MNS preparation.
the olfactory, rostral migratory stream and trigeminal routes with
Intranasal immunization. Mice were anaesthetized with isoflurane and then placed
a residency time in the brain of 96 h. While distribution of the on their backs with their heads positioned to maximize the residency time for the
NU4MNS probe seems to correlate with the published distribution delivered material remaining on the olfactory surface. Each naris was slowly
of HRP-NU4, NU4MNS probe residency in the brain appears to administered with 568-NU4, NU4MNS or non-immune IgGMNS (5 µl/naris) using
be longer. a sterile micropipette, over a period of 1 min, keeping the opposite naris and mouth
closed to allow complete aspiration of the delivered material.
In conclusion, a molecular MRI probe designated NU4MNS,
capable of targeting Alzheimer’s disease-related AβOs, has been MRI of brain slices. A Falcon tube containing the MNS-treated tissue samples was
synthesized by conjugating AβO-selective antibodies to superpara- placed in a Bruker PharmaScan 7 T animal MRI scanner. Samples were imaged
magnetic nanoparticles. NU4MNS is both specific and sensitive using a T⋆2 -weighted Gradient Echo (GRE) FLASH pulse sequence three-
and can distinguish Alzheimer’s disease brain tissue from non- dimensional acquisition. The total acquisition time (TA) was 6 h, 25 min. The echo
demented controls by MRI in vitro. In vivo, the probe reaches the time (TE) was 30 ms. The flip angle (FA) was 15°. The image resolution was
0.07 mm isotropic, with a field of view of 4.62 cm/2.52 cm/0.65 cm and image
brain following intranasal inoculation of mice and can provide an matrix (MTX) comprising 700 pixels/385 pixels/100 pixels. The number of averages
MRI signal that requires transgene-dependent Alzheimer’s disease (NEX) was set to 20 to improve the signal-to-noise ratio.
pathology. The development of humanized AβO-specific anti-
bodies21 substantiates the potential of this new approach for use MRI of Tg and wild-type mice in vivo. Following intranasal inoculation, the probe
in Alzheimer’s disease diagnostics and for measuring the efficacy was allowed to distribute for 4 h before MRI was performed according to the
imaging methodology described by Mundt and colleagues44. T1-, T2- and
of investigational new drugs. The nanotechnology-based probe,
T⋆2 -weighted magnetic resonance images were acquired on a Bruker BioSpec 7 T
which can be readily adapted to other targets, provides a strategic magnet, using a 25 mm RF quadrature coil. The in-plane resolution was 75 µm with
and powerful early detection advantage over current PET probes a slice thickness of 0.4 mm. T1- and T2-weighted images provided anatomical
for amyloid by targeting AβOs, toxins putatively responsible for guidance as well as some localization of the NU4MNS and were acquired with a fat
the neuron damage beginning early in Alzheimer’s disease. suppressed spin echo sequence (rapid acquisition with relaxation enhancement,
RARE) with the following parameters: T1-weighted, TR = 1,000 ms, TEeff = 13.2 ms,
RARE factor 2, number of excitations, NEX = 4; T2-weighted TR = 3,500 ms,
Methods TEeff = 58.5 ms, RARE factor 4, NEX = 4. T⋆2 -weighted imaging provides more of the
Animals. The 5xFAD Tg mouse model (B6SJL-Tg(APPSwFlLon,
localization of the NU4MNS as the iron causes local changes in the magnetic
PSEN1⋆M146L⋆L286V)6799Vas)33 (Jackson Laboratories) is bred on a non-Tg ⋆
susceptibility to which T2 -weighted images can be sensitive. A gradient echo
background (B6SJLF1). Aged Tg and wild-type littermates, 6–12 months old,
sequence was used with the following parameters (gradient echo fast imaging,
were used.
GEFI; TR = 1,200 ms, TE = 5.6 ms, flip angle = 35°, NEX = 4).
Cell lines. HeLa is a human cervical cancer cell line. NIH/3T3s is a mouse
embryonic fibroblast line. HEPG3 is from hepatocellular carcinoma. RAW264.7 is a Received 4 November 2013; accepted 8 October 2014;
mouse leukaemic monocyte macrophage cell line. published online 22 December 2014
Synthesis of MNSs. The 16 nm magnetite nanoparticles were synthesized by
decomposition of iron–oleate at 320 °C as described in an earlier report43. For the References
synthesis of iron–oleate complexes, 10.8 g of iron(III) chloride hexahydrate and 36.5 g 1. 2014 Alzheimer’s disease facts and figures. Alzheimers Dement. 10,
sodium oleate were dissolved in a mixture of 60 ml distilled water, 80 ml ethanol and e47–e92 (2014).
140 ml hexane and heated at 60 °C for 4 h. The organic layer of the biphasic mixture 2. Georganopoulou, D. G. et al. Nanoparticle-based detection in cerebral spinal
became dark, indicating phase transfer of iron(III) ions and the formation of iron– fluid of a soluble pathogenic biomarker for Alzheimer’s disease. Proc. Natl Acad.
oleate complex. The resulting dark solution was separated and washed with water Sci. USA 102, 2273–2276 (2005).
three times. 3. Toledo, J. B., Xie, S. X., Trojanowski, J. Q. & Shaw, L. M. Longitudinal change in
For the synthesis of 16 nm magnetite nanoparticles, 18 g of iron–oleate complex CSF τ and Aβ biomarkers for up to 48 months in ADNI. Acta Neuropathol. 126,
and 2.58 g of oleic acid were dissolved in 100 g octadecene at room temperature and 659–670 (2013).
heated to 320 °C at a rate of 3.3 °C min−1. The reaction mixture was kept at 320 °C 4. Slemmon, J. R. et al. Measurement of Aβ1–42 in cerebrospinal fluid is influenced
for 40 min, then cooled to room temperature. The resulting nanoparticles were by matrix effects. J. Neurochem. 120, 325–333 (2012).
separated from the solution by the addition of ethanol and ethyl acetate, followed 5. Klunk, W. E. et al. Imaging brain amyloid in Alzheimer’s disease with Pittsburgh
by centrifugation. compound-B. Annals Neurol. 55, 306–319 (2004).
6. Johnson, K. A. et al. Appropriate use criteria for amyloid PET: a report of the
Preparation of dopamine-TEG-COOH and phase transfer. To make the organic- Amyloid Imaging Task Force, the Society of Nuclear Medicine and Molecular
phase synthesized MNSs suitable for biological applications, we stabilized the MNSs Imaging, and the Alzheimer’s Association. Alzheimers Dement. 9,
using carboxylate-terminated ligand with nDOPA as an anchor. The synthesis of e1–e16 (2013).
carboxylate-terminated nDOPA ligand and functionalization of the MNSs was 7. Terry, R. D. et al. Physical basis of cognitive alterations in Alzheimer’s disease:
carried out according to the following protocol. Tetraethylene diacide, synapse loss is the major correlate of cognitive impairment. Ann. Neurol. 30,
N-hydroxysuccinimide, N,N′-dicyclohexylcarbodiimide, nDOPA hydrochloride and 572–580 (1991).
anhydrous sodium bicarbonate were dissolved in chloroform under an argon 8. Nyborg, A. C. et al. In vivo and ex vivo imaging of amyloid-beta cascade
atmosphere and stirred for 4 h. Hexane-stabilized MNSs were added and stirred for aggregates with a pronucleon peptide. J. Alzheimer’s Dis. 34, 957–967 (2013).
another 24 h. The precipitate formed was separated by magnet, dispersed in water 9. Klein, W. L., Krafft, G. A. & Finch, C. E. Targeting small Aβ oligomers: the
and purified by dialysis. solution to an Alzheimer’s disease conundrum? Trends Neurosci. 24,
219–224 (2001).
Conjugation of antibody to MNS. The conjugation of buffer-stabilized MNSs with 10. Lublin, A. L. & Gandy, S. Amyloid-β oligomers: possible roles as key neurotoxins
antibody (Ab) was performed using a conventional carboxyl–amine crosslinking in Alzheimer’s disease. Mount Sinai J. Med. New York 77, 43–49 (2010).
method. We first activated the carboxyl-terminated MNSs by sulpho-N-hydroxy 11. Ferreira, S. T. & Klein, W. L. The Aβ oligomer hypothesis for synapse failure
succinimide and EDC, followed by incubation with the corresponding antibody and memory loss in Alzheimer’s disease. Neurobiol. Learn. Mem. 96,
(NU4 or IgG1 , with or without fluorescent label) overnight. Conjugated MNSs were 529–543 (2011).
separated by magnet to remove excess reagent and antibody, then redispersed in 12. Schnabel, J. Amyloid little proteins, big clues. Nature 475, S12–S14 (2011).
working media. Conjugation efficiency was estimated using ultraviolet spectroscopy 13. Mucke, L. & Selkoe, D. J. Neurotoxicity of amyloid β-protein: synaptic and
(absorbance at 280 nm) of the magnetically separated supernatant. Conjugation network dysfunction. Cold Spring Harbor Persp. Med. 2, a006338 (2012).
efficiency was calculated according to the following equation: 14. Lacor, P. N. et al. Synaptic targeting by Alzheimer’s-related amyloid β oligomers.
J. Neurosci. 24, 10191–10200 (2004).
Ab conc. = (total mg added Ab) − (mg Ab in supernatant) 15. De, M., Chou, S. S., Joshi, H. M. & Dravid, V. P. Hybrid magnetic nanostructures
(MNS) for magnetic resonance imaging applications. Adv. Drug Deliv. Rev. 63,
Of note, because both fluorophore and MNS conjugation use carboxyl–amine 1282–1299 (2011).
crosslinking, fluorescent NU4 that has more than 2.5 mol fluorophore/mol antibody 16. Corot, C. et al. in Molecular and Cellular MR Imaging (eds Modo, M. M. J. &
does not conjugate well to the MNSs. The optimal concentration of Bulte, J. W. M.) Ch. 4, 59–84 (CRC Press, 2007).

NATURE NANOTECHNOLOGY | ADVANCE ONLINE PUBLICATION | www.nature.com/naturenanotechnology 7

© 2014 Macmillan Publishers Limited. All rights reserved.


ARTICLES NATURE NANOTECHNOLOGY DOI: 10.1038/NNANO.2014.254

17. Liu, S. et al. A novel type of dual-modality molecular probe for MR and nuclear 39. Tong, S. et al. Coating optimization of superparamagnetic iron oxide
imaging of tumor: preparation, characterization and in vivo application. Mol. nanoparticles for high T2 relaxivity. Nano Lett. 10, 4607–4613 (2010).
Pharmaceut. 6, 1074–1082 (2009). 40. Amstad, E. et al. Ultrastable iron oxide nanoparticle colloidal suspensions
18. Serres, S. et al. Molecular MRI enables early and sensitive detection of brain using dispersants with catechol-derived anchor groups. Nano Lett. 9,
metastases. Proc. Natl Acad. Sci. USA 109, 6674–6679 (2012). 4042–4048 (2009).
19. Lambert, M. P. et al. Monoclonal antibodies that target pathological assemblies 41. Palumbo, A., Napolitano, A., Barone, P. & d’Ischia, M. Nitrite- and peroxide-
of Aβ. J. Neurochem. 100, 23–35 (2007). dependent oxidation pathways of dopamine: 6-nitrodopamine and
20. Lambert, M. P., Velasco, P. T., Viola, K. L. & Klein, W. L. Targeting generation of 6-hydroxydopamine formation as potential contributory mechanisms of
antibodies specific to conformational epitopes of amyloid β-derived neurotoxins. oxidative stress- and nitric oxide induced neurotoxicity in neuronal
CNS Neurol. Disord. Drug Targets 8, 65–81 (2009). degeneration. Chem. Res. Toxicol. 12, 1213–1222 (1999).
21. Acton, P. et al. Anti-ADDL antibodies and uses thereof. US patent 42. Robinson, R. MRI probe for amyloid-β oligomers offers potential advantages for
7,811,563 (2010). detecting Alzheimer’s disease. Neurol. Today 12, 20–22 (2012).
22. Xiao, C. et al. Brain transit and ameliorative effects of intranasally delivered anti- 43. Park, J. et al. Ultra-large-scale syntheses of monodisperse nanocrystals. Nature
amyloid-β oligomer antibody in 5xFAD mice. J. Alzheimer’s Dis. 35, Mater. 3, 891–895 (2004).
777–788 (2013). 44. Mundt, A. P. et al. Targeting activated microglia in Alzheimer’s pathology by
23. Gong, Y. et al. Alzheimer’s disease-affected brain: presence of oligomeric Aβ intraventricular delivery of a phagocytosable MRI contrast agent in APP23
ligands (ADDLs) suggests a molecular basis for reversible memory loss. Proc. transgenic mice. NeuroImage 46, 367–372 (2009).
Natl Acad. Sci. USA 100, 10417–10422 (2003).
24. Kayed, R. et al. Common structure of soluble amyloid oligomers implies
common mechanism of pathogenesis. Science 300, 486–489 (2003). Acknowledgements
25. Chang, L. et al. Femtomole immunodetection of synthetic and endogenous The authors thank the staff of the Biological Imaging Facility (BIF), the Center for
amyloid-β oligomers and its application to Alzheimer’s disease drug candidate Advanced Molecular Imaging (CAMI) and Northwestern University’s Atomic and
screening. J. Mol. Neurosci. 20, 305–313 (2003). Nanoscale Characterization Experimental Center (NUANCE) at Northwestern University,
26. Lesne, S. et al. A specific amyloid-β protein assembly in the brain impairs and the Department of Radiology at NorthShore University HealthSystems. The authors
memory. Nature 440, 352–357 (2006). also thank the Northwestern Brain Bank at the Cognitive Neurology and Alzheimer’s
27. Ohno, M. et al. Temporal memory deficits in Alzheimer’s mouse models: rescue Disease Center (CNADC) for providing the human Alzheimer’s disease and control brain
by genetic deletion of BACE1. Eur. J. Neurosci. 23, 251–260 (2006). tissue. The authors acknowledge support from their funding agencies and grants. This work
28. Mucke, L. et al. High-level neuronal expression of Aβ1–42 in wild-type human was funded by the National Institutes of Health (AG022547, AG029460 and AG045637 to
amyloid protein precursor transgenic mice: synaptotoxicity without plaque W.L.K.), by Baxter Healthcare (to W.L.K.) and partially by the National Institutes of
formation. J. Neurosci. 20, 4050–4058 (2000). Health–Centers of Cancer Nanotechnology Excellence through the Nanoconstruct Core
29. Balducci, C. et al. Cognitive deficits associated with alteration of synaptic (award U54CA119341 to V.P.D., H.J. and M.D.)
metaplasticity precede plaque deposition in AβPP23 transgenic mice.
J. Alzheimer’s Dis. 21, 1367–1381 (2010).
30. Ferretti, M. T. et al. Transgenic mice as a model of pre-clinical Alzheimer’s
Author contributions
The study concept and design was provided by K.L.V., J.S., R.S., M.D., V.P.D. and W.L.K.
disease. Curr. Alzheimer Res. 8, 4–23 (2011).
The acquisition of data was performed by M.D., H.J., J.S. and S.V. (MNS development,
31. Takamura, A. et al. Extracellular and intraneuronal HMW-AβOs represent a
production, and characterization), K.L.V., J.S., R.S., M.A.B., J.W., S.V., S.S., S.W., C.L.
molecular basis of memory loss in Alzheimer’s disease model mouse. Mol.
and J.P. (antibody conjugation, cell experiments, tissue experiments and
Neurodegen. 6, 20 (2011).
immunoprecipitation experiments), K.L.V. and M.A.B. (animal experiments), K.M.
32. Velasco, P. T. et al. Synapse-binding subpopulations of Aβ oligomers sensitive to
(inductively coupled plasma mass spectrometry experiments) and E.A.W. and P.P
peptide assembly blockers and scFv antibodies. ACS Chem. Neurosci. 3,
(magnetic resonance image acquisition). All authors discussed the results and contributed
972–981 (2012).
to the analysis of the data. Critical revision of the article for intellectual content was
33. Oakley, H. et al. Intraneuronal β-amyloid aggregates, neurodegeneration, and
conducted by K.L.V., J.S., R.S., M.D., P.T.V., V.P.D. and W.L.K. Funding was obtained and
neuron loss in transgenic mice with five familial Alzheimer’s disease mutations:
studies supervised by W.L.K. and V.P.D.
potential factors in amyloid plaque formation. J. Neurosci. 26,
10129–10140 (2006).
34. Barick, K. C. et al. Nanoscale assembly of amine-functionalized colloidal iron Additional information
oxide. J. Magn. Magn. Mater. 321, 1529–1532 (2009). Supplementary information is available in the online version of the paper. Reprints and
35. Na, H. B., Song, I. C. & Hyeon, T. Inorganic nanoparticles for MRI contrast permissions information is available online at www.nature.com/reprints. Correspondence and
agents. Adv. Mater. 21, 2133–2148 (2009). requests for materials should be addressed to V.P.D. and W.L.K.
36. Pinho, S. L. C. et al. Fine tuning of the relaxometry of γ-Fe2O3@SiO2
nanoparticles by tweaking the silica coating thickness. ACS Nano. 4,
5339–5349 (2010). Competing financial interests
37. Prakash, A. et al. Bilayers as phase transfer agents for nanocrystals prepared in This work was funded, in part, by Baxter Healthcare. Northwestern University holds the
nonpolar solvents. ACS Nano. 3, 2139–2146 (2009). rights to two US and several international patents concerning antibodies that target Aβ
38. Amstad, E., Textor, M. & Reimhult, E. Stabilization and functionalization of iron oligomer assemblies. Acumen Pharmaceuticals holds the licensing rights to develop
oxide nanoparticles for biomedical applications. Nanoscale 3, 2819–2843 (2011). anti-Aβ oligomer antibodies for therapeutic use.

8 NATURE NANOTECHNOLOGY | ADVANCE ONLINE PUBLICATION | www.nature.com/naturenanotechnology

© 2014 Macmillan Publishers Limited. All rights reserved.

You might also like