Вы находитесь на странице: 1из 11

Neuron

NeuroResource

Direct Targeted Quantitative Molecular Imaging


of Neurotransmitters in Brain Tissue Sections
Mohammadreza Shariatgorji,1 Anna Nilsson,1 Richard J.A. Goodwin,1,2 Patrik Källback,1 Nicoletta Schintu,3
Xiaoqun Zhang,3 Alan R. Crossman,4 Erwan Bezard,5 Per Svenningsson,3 and Per E. Andren1,*
1Biomolecular Imaging and Proteomics, National Center for Mass Spectrometry Imaging, Department of Pharmaceutical Biosciences,

Uppsala University, P.O. Box 591 BMC, 75124 Uppsala, Sweden


2AstraZeneca R&D, Alderley Park, Macclesfield, Cheshire SK10 4TF, UK
3Center for Molecular Medicine, Department of Neurology and Clinical Neuroscience, Karolinska Institutet and Karolinska University Hospital,

17176 Stockholm, Sweden


4Faculty of Life Sciences, University of Manchester, Manchester M13 9PL, UK
5Université de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293 Bordeaux, France

*Correspondence: per.andren@farmbio.uu.se
http://dx.doi.org/10.1016/j.neuron.2014.10.011

SUMMARY Nuclear medicine imaging is a widely used tool for indirect


visualization of the distribution, abundance, and activity of
Current neuroimaging techniques have very limited neurotransmitters in the brain, relying on radiolabeled tracers
abilities to directly identify and quantify neurotrans- that emit gamma rays, which are detected using positron emis-
mitters from brain sections. We have developed a sion tomography (PET) or single photon emission computed
molecular-specific approach for the simultaneous tomography scanners (Pimlott and Sutherland, 2011). Devel-
imaging and quantitation of multiple neurotransmit- oping appropriate labeled compounds is often complicated,
particularly for studying endogenous chemical messengers
ters, precursors, and metabolites, such as tyrosine,
(Badgaiyan, 2011). There is often an inability to discriminate
tryptamine, tyramine, phenethylamine, dopamine,
between the labeled parent compound and a metabolite retain-
3-methoxytyramine, serotonin, GABA, glutamate, ing the label. Imaging techniques that can directly visualize the
acetylcholine, and L-alpha-glycerylphosphorylcho- localization of neurotransmitters and simultaneously quantitate
line, in histological tissue sections at high spatial them without relying on a label would thus represent a scientific
resolutions. The method is employed to directly breakthrough. Another significant advance in the molecular
measure changes in the absolute and relative levels analysis of neurological tissues would be the development of
of neurotransmitters in specific brain structures in a multiplex method that enables the simultaneous detection of
animal disease models and in response to drug treat- multiple endogenous compounds and pharmaceutical agents.
ments, demonstrating the power of mass spectrom- MALDI mass spectrometry (MS) imaging has been used
etry imaging in neuroscience. for direct molecule-specific compound detection, distribution
mapping, and identifying molecular species in tissue sections
(Caprioli et al., 1997; Cornett et al., 2007). It can generate multi-
INTRODUCTION plex full mass spectrum data at near-cellular spatial resolution.
Crucially, it provides a powerful tool for in situ visualization of
Small-molecule neurotransmitters such as the catecholamine target compounds and for determining their abundance and
dopamine (DA), the amino acids g-amino butyric acid (GABA) spatial distribution without requiring a great deal of a priori infor-
and glutamate (Glu), and acetylcholine (ACh) are critical chemi- mation. Moreover, the data gathered during such experiments
cal messengers that transmit signals between neurons. Changes can be used to simultaneously identify unknown endogenous
in their concentrations are associated with numerous normal molecular changes due to disease or degeneration. MALDI-MS
neuronal processes, such as sleep and aging, but also several can be used for the direct analysis of targets, ranging from small
disease states, including Alzheimer’s disease, Parkinson’s molecules to large proteins (Schwamborn and Caprioli, 2010).
disease (PD), depression, drug addiction, and attention deficit However, despite some notable successes, it is relatively insen-
hyperactivity disorder. A better understanding of their relative sitive toward certain compound classes and sometimes suffers
abundance and distribution would provide insights into these from signal interference caused by matrix clusters/fragments
complex neurological processes and disorders. At present, re- (Sugiura et al., 2012; Ye et al., 2013). To date, it has found only
searchers rely on indirect histochemical, immunohistochemical limited applications in the study of neurotransmitters due to
(IHC), and ligand-based assays to detect these small-molecule their poor ionization efficiencies. As such, the development of
transmitter substances (de Jong et al., 2005; Falck et al., 1962; a reagent that would enable selective neurotransmitter derivati-
Jones and Beaudet, 1987). The antibodies used in IHC often zation while also facilitating their detection by MS and acting
have major limitations, such as an inability to distinguish be- as an efficient matrix to assist their laser-induced desorption
tween different transmitters (Keenan and Koopowitz, 1981). and ionization would be a significant breakthrough.

Neuron 84, 697–707, November 19, 2014 ª2014 Elsevier Inc. 697
Neuron
Quantitative Molecular Imaging of Neurotransmitters

Here we describe a unique approach for the direct and In addition to showing the distributions of known derivatives,
absolute quantitative imaging of classical and common neuro- these images can be used to characterize unidentified ions
transmitters in histological brain tissue sections. We report whose concentrations and localizations may be affected by
an example of the simultaneous mapping of biogenic amines neurological events and diseases. We demonstrated this by
and amino acid chemical messengers following in situ derivati- determining the localization of selected neurotransmitters in
zation in brain tissue sections. The method is employed to brain tissue samples derivatized using DPP-TFB and also
simultaneously measure changes in the levels of neurotransmit- 2,4,6-trimethyl-pyranylium tetrafluoroborate (TMP-TFB) as a
ters, including tyrosine, tryptamine, tyramine, phenethylamine, confirmatory reagent. The identities of the signals for the neuro-
DA, 3-methoxytyramine (3-MT), serotonin (5-HT), GABA, and transmitter derivatives were verified by performing tandem
Glu, in specific brain structures of primates and rodents with MS (MS/MS) experiments using derivatized synthetic standards
and without DA depletion and L-3,4-dihydroxyphenylalanine alongside the endogenous neurotransmitter. In all cases, the
(L-DOPA) treatment. In addition, we have developed a method product ions observed for the standards were identical to those
that can be used to image and quantitate ACh and an ACh for the endogenous species. For example, both exogenous and
precursor, L-alpha-glycerylphosphoryl-choline (alpha-GPC) in standard GABA generated a peak at m/z 208.1 and similar MS/
brain sections, as well as to study activity within cholinergic MS product ions when derivatized with TMP-TFB and m/z
pathways by measuring changes in ACh concentrations, imaged 318.1 and similar MS/MS product ions when derivatized with
at a 15 mm spatial resolution. Together, these experiments DPP-TFB (Figure S1). Similarly, both endogenous and standard
demonstrate a significant advancement in the molecular imaging DA yielded peaks at m/z 136 and m/z 232, respectively, after
arsenal available to neuroscientists. derivatization with these reagents (Figure S1). In addition, the
elemental compositions obtained for the target ions from accu-
RESULTS AND DISCUSSION rate mass Fourier transform ion cyclotron resonance (FT-ICR)-
MS measurements were in good agreement with the expected
Without modification, most neurotransmitters are not directly structures of the derivatized neurotransmitters (Tables S1 and
detectable by MALDI-MS due to their poor ionization efficiency S2). Deuterated neurotransmitters were used as calibration stan-
and the overlapping signals of isobaric compounds. To address dards to determine the absolute concentrations of the endoge-
the limited ionization and desorption of neurotransmitters, we nous neurotransmitters directly in brain tissue sections.
developed a method for their in situ chemical derivatization.
Once modified, the neurotransmitters are readily ionized and de- MALDI-MS Imaging of Neurotransmitters in Control
tected, enabling their identification and the quantitative mapping Rodent Brain Sections
of their distributions. Our strategy uses pyrylium salts such The in situ neurotransmitter derivatization and imaging proce-
as 2,4-diphenyl-pyranylium tetrafluoroborate (DPP-TFB), which dure was initially demonstrated using coronal and sagittal mouse
react selectively with primary amines to produce N-alkyl- or and rat brain sections (Figures 1, S1, and S2). The distribution
N-aryl-pyridinium derivatives (Figure S1, available online). The of the pyrylium salt formed by derivatizing GABA with DPP-
reaction proceeds under very mild conditions and occurs rapidly TFB (m/z 318.1) in coronal rat brain tissue sections showed
at ambient temperature and pressure without any need for that GABA is most abundant in the medial septum/diagonal
stirring or agitation. Such conditions are required to preserve band region (MSDB) of the brain (Figure S2), which is consistent
the localization of endogenous compounds. The resulting with previous reports (Alreja et al., 2000). The MSDB provides a
charged derivatives have laser desorption and ionization major GABAergic input to the hippocampus (Alreja et al., 2000;
efficiencies sufficient to enable the detection of endogenous Qin and Luo, 2009). Imaging of sagittal rat brain sections (Fig-
small-molecule primary amines (Johannesen et al., 2012; Quirke ure 1B) revealed structures with high GABA concentrations,
et al., 1994), such as tyrosine, tryptamine, tyramine, phenethyl- including the hypothalamus, midbrain, and basal forebrain (Fig-
amine, DA, 3-MT, 5-HT, GABA, and Glu, simultaneously in a sin- ure 1C). GABA is a dominant neurotransmitter in the hypothala-
gle experiment. Importantly, the selective derivatization of the mus and plays an important role in hypothalamic inhibitory
primary amines separates their signals from those of overlapping circuits (Decavel and Van den Pol, 1990). Moreover, the basal
isobaric compounds. A MALDI-MS analysis of a representative forebrain and its substructures are reported to provide major
pyrylium derivatization reaction clearly showed that all of the GABAergic inputs to the hippocampus (Alreja et al., 2000). The
desired pyridinium ions were formed in high yields (Figure S1). importance of midbrain neurons in GABA recycling and the
Furthermore, the DPP-TFB derivatives of endogenous primary corelease of GABA from the dopaminergic midbrain neurons
amines, including neurotransmitters and their metabolites and via plasma membrane uptake (Tritsch et al., 2014) may explain
precursors, undergo self-assisted laser desorption ionization. the high GABA concentration in this region. Deuterated GABA
As such, they are amenable to MALDI-MS imaging without (D6-GABA) was used to determine absolute GABA concentra-
needing to be assisted by a matrix such as a-cyano-4-hydroxy- tions in rat brain tissue sections. The absolute concentration of
cinnamic acid (CHCA). GABA in the coronal brain section as a whole was 5 nmol/mg,
The combination of pyrylium derivatization and multiplex data while that in the MSDB region was 10 nmol/mg (Figure S2). These
acquisition generates hundreds of ion signals in each MALDI-MS values are consistent with previous results (Schaaf et al., 1985).
imaging experiment. This makes it possible to obtain images Other primary amine neurotransmitters, such as tyrosine,
showing the distribution of every individual ion within an m/z tryptamine, tyramine, phenethylamine, DA, 3-MT, 5-HT, GABA,
window of 200 to 450 in the sample being studied (Figure 1A). and Glu, were detected and imaged in the same brain tissue

698 Neuron 84, 697–707, November 19, 2014 ª2014 Elsevier Inc.
Neuron
Quantitative Molecular Imaging of Neurotransmitters

Figure 1. MALDI-MS Imaging Data Display-


ing GABA, DA, and 3-MT and Distributions
in Rat Brain Tissue Sections
(A–I) The mass spectrum generated in a MALDI-
MS imaging experiment (A) contains hundreds of
ion signals associated with each individual pixel.
These can be used to create images showing the
distributions of each individual ion in the spectrum.
A sagittal tissue section from a rat brain (B) and the
associated relative abundances and distributions
of GABA (C), DA (D), and 3-MT (E). GABA was
found in all structures of the brain but was most
abundant in HY, BF and its substructures, and MB.
High DA concentrations were observed in SN,
CP, VS, and AON. The distribution of 3-MT
mirrored that of DA. DA imaging in a rat brain
coronal tissue section (F) revealed that it was most
abundant in the striatal region (G). By rescaling the
signal intensity of the latter image, it was possible
to detect DA in other parts of the brain, notably
in the A24, Pir, MS, and VDB. However, the
DA concentrations in these substructures were all
much lower than that in striatal region (H). The
intensities of the DA signals in each region of the
brain were compared to enable its relative quan-
titation. The inset bars show the rescaled DA signal
intensities for the Pir, MS and VDB, and A24
regions (I). MS data were acquired using either
a MALDI-TOF/TOF (A–E) or a MALDI-FT-ICR
mass spectrometer (G–I). Data are shown using a
rainbow scale, normalized against the total ion
count. Scale bar, 2 and 5 mm. Images were ac-
quired at spatial resolutions of 100 mm for sagittal
sections and 150 mm for coronal sections. Sub-
stantia nigra, SN; caudate-putamen, CP; ventral
striatum, STRv; anterior olfactory nucleus, AON;
cingulate cortex (area 24), A24; piriform cortex, Pir;
medial septal nucleus, MS; nucleus of the vertical
limb of the diagonal band, VDB; accumbens
nucleus, ACB; hypothalamus, HY; thalamus, TH;
hippocampus, HIPP; corpus callosum, cc; cere-
bral cortex, CTX; midbrain, MB; basal forebrain,
BF. See also Figure S1.

sections that were treated with DPP-TFB to produce 2,4-diphe- metabolite, but recent studies suggest that it is a neuromodula-
nylpyridinium derivatives. As before, this substantially increased tor that acts as an agonist of the trace amine-associated recep-
the neurotransmitter’s MALDI desorption/ionization efficiency tor 1 (Sotnikova et al., 2010). It is worth noting that distinguishing
and generated a detectable signal at m/z 362.1 (Figure S1). between an endogenous compound and its metabolites is
Glu was found in most structures of the brain but was mainly very difficult when using techniques based on radiolabeled
localized in the striatal and cortex regions (Figure S2), which is compounds. In the coronal sections (Figure 1F), DA was mostly
consistent with previous reports on the distribution of Glu trans- localized in the striatal regions (Figure 1G), but it was also
porters (Bressan and Pilowsky, 2000; El Mestikawy et al., 2011; present at lower concentrations in the cingulate cortex, piriform
Herzog et al., 2004). The identity of the Glu peak was verified by cortex, medial septal nucleus, and the nucleus of the vertical
MS/MS fragmentation of derivatized synthetic standards limb of the diagonal band (Figure 1H). The intensities of the DA
(deuterated and nondeuterated) and the endogenous signal signals for selected regions (Figure 1F) were used to quantify
(Figure S1). their DA levels in relative terms (Figure 1I), revealing that the
Analyses of rat brain sagittal sections revealed high DA con- average DA concentration in the striatal region is about 40 times
centrations in the substantia nigra, striatum, and ventral pallidum higher than that in the cingulate cortex region.
structures due to the nigrostriatal and mesolimbic DAergic path-
ways (Figure 1D). 3-MT, a methylated DA metabolite formed Neurotransmitter MALDI-MS Imaging of Parkinson’s
by the action of the enzyme catechol-O-methyl transferase, Disease Models
was also imaged and found to have the same distribution as To further demonstrate the potential of the derivatization/MALDI-
DA (Figure 1E). 3-MT was initially reported to be an inactive DA MS imaging methodology, it was used to analyze the distribution

Neuron 84, 697–707, November 19, 2014 ª2014 Elsevier Inc. 699
Neuron
Quantitative Molecular Imaging of Neurotransmitters

Figure 2. MALDI-MS Images of Neurotrans-


mitters in Coronal Rat PD Model Tissue
Sections
(A–U) Imaging experiments were conducted
on brain tissue sections from unilateral sham-
lesioned-, unilateral 6-OHDA-lesioned, and uni-
lateral 6-OHDA-lesioned animals that were treated
with subchronic L-DOPA for 4 weeks, with the
final dose being given as deuterated (D3)-L-DOPA.
The images show the distributions of DA (A–F)
in sham-lesioned (A), 6-OHDA-lesioned (B),
and 6-OHDA-lesioned L-DOPA-treated animals.
Rescaling images (D–F) (with new max intensity
corresponding to 5% of the previous) made it
possible to determine the distribution of DA in
the 6-OHDA-lesioned side of the brain and in
structures with low DA concentrations, such as the
cortex. (D) Rescaled image from (A). (E) Rescaled
image from (B). (F) Rescaled image from (C).
Distribution of D3-DA (formed by the in vivo
breakdown of D3-L-DOPA is imaged in sham-
lesioned (G), 6-OHDA-lesioned (H), and 6-OHDA-
lesioned L-DOPA-treated animals (I). Distribution
of endogenous 3-MT is imaged in sham-lesioned
(J), 6-OHDA-lesioned (K), and 6-OHDA-lesioned
L-DOPA-treated animals (L), and distribution of
D3-3-MT (derived from D3-L-DOPA) is imaged
in sham-lesioned (M), 6-OHDA-lesioned (N), and
6-OHDA-lesioned L-DOPA-treated animals (O).
Distribution of GABA (P–R) is imaged in sham-
lesioned (P), 6-OHDA-lesioned (Q), and 6-OHDA-
lesioned L-DOPA-treated animals (R), and tyrosine
is imaged in sham-lesioned (S), 6-OHDA-lesioned
(T), and 6-OHDA-lesioned L-DOPA-treated ani-
mals (U). There are clear differences in the
concentrations of the studied amines in different
regions of the brain and between treatments. MS
images were acquired using a MALDI-FT-ICR
mass spectrometer. Data are shown using a
rainbow scale, normalized against the total ion
count. Scale bar, 2 mm; spatial resolution =
150 mm. See also Figure S2.

of neurotransmitters in experimental models of PD. When study- neously distributed in the striatum; in PD, it is mainly the
ing neurodegeneration processes in PD models, it is desirable to dorsolateral regions that exhibit reduced DA levels. Important
assess the integrity and function of the nigrostriatal DAergic information might therefore be lost if the anatomical distribution
pathway. Various techniques have been used for this purpose, of DA is not determined. Our method allows the simultaneous
including the quantitation of dopaminergic neurons and termi- direct imaging and quantitation of primary amine neurotransmit-
nals in the substantia nigra pars compacta and striatum by ters in tissue sections from animal models of PD.
immunostaining using an antibody against tyrosine hydroxylase The PD models used in our experiments were generated
(TH), and the measurement of TH or DA transporters (DAT) in by injecting the neurotoxin 6-hydroxydopamine (6-OHDA) into
striatal tissue extracts by immunoblotting (Ciliax et al., 1995; rodents’ medial forebrain bundles (Zhang et al., 2008) or by
Huot et al., 2007). Alternatively, the dopamine neurons can be intravenous administration of 1-methyl-4-phenyl-1,2,3,6-tetra-
quantified by using autoradiography to measure the levels of hydropyridine (MPTP) to primates. Pyrylium derivatization
a radiolabeled DAT ligand (Boja et al., 1991). The best way to and MALDI-MS imaging were then used to quantify neurotrans-
quantitate DA is to use high-performance liquid chromatography mitter levels in rat brain sections by depositing known con-
coupled with an electrochemical detector. However, this centrations of deuterated standards onto vehicle control tissue
approach provides no information on the neurotransmitter’s sections. The DA concentrations in the intact and 6-OHDA-
anatomical localization. Dopaminergic terminals are heteroge- lesioned sides of rat striata were determined to be 124 pmol/mg

700 Neuron 84, 697–707, November 19, 2014 ª2014 Elsevier Inc.
Neuron
Quantitative Molecular Imaging of Neurotransmitters

and 84 pmol/mg, respectively (Figure S2); these values are used to study the distribution of DA and the effects of L-DOPA in
consistent with previous results obtained by HPLC (Zhao and PD models, we were able to map the distribution of 5-HT in con-
Suo, 2008). trol (Figure 3G) and MPTP-lesioned (Figure 3H) brain tissue sec-
Pyrylium derivatization and MS imaging were used to evaluate tions. 5-HT was localized to the nucleus basalis (Meynert), the
the neurochemical changes that occur in 6-OHDA PD models hypothalamic regions, and the substantia nigra. MPTP lesioning
with and without L-3,4-dihydroxyphenylalanine (L-DOPA) treat- increased its concentration in these regions by >40% (Figure S3).
ment. Some of the model animals were treated with deuterated Increases in 5-HT concentrations following MPTP-induced DA
L-DOPA (D3-L-DOPA), which is converted into deuterated denervation have previously been demonstrated by in vivo
dopamine (D3-DA) in vivo. The derivatization/MALDI-MS imag- microdialysis; it was suggested that they are due to the brain’s
ing protocol successfully distinguished endogenous DA from compensatory processes and recovery mechanisms (Boulet
both the administered D3-L-DOPA and D3-DA. Sham-lesioned et al., 2008).
rat brain tissue sections had a normal DA distribution in their Increased levels of striatal GABA have been reported in
striatal regions, but the intensity of the DA signal was signifi- humans with PD, 6-OHDA-treated rats, and MPTP-treated pri-
cantly lower in the lesioned side of the brain, with or without mates. Striatal GABA levels increased by 70% in MPTP-lesioned
D3-L-DOPA treatment (Figures 2A–2C). The distributions of brains (Figures 3I, 3J, and S3) (Boulet et al., 2008); interestingly,
DA in the lesioned side of the brain and structures with low its concentration also rose in the hypothalamic regions. In addi-
DA levels, such as the cortex, were determined by rescaling tion to DA, 5-HT, and GABA, high mass accuracy FT-ICR imag-
the intensities of the MS-derived images (Figures 2D–2F). ing maps were also generated for phenethylamine, tyramine, and
No interfering signal for D3-DA was detected in sham- or tryptamine (Table S2) in control and MPTP-treated primate
6-OHDA-lesioned brain tissue sections (Figures 2G and 2H), brains (Figure S3).
and the signal corresponding to D3-DA (derived from D3-L- It is important to recall that while the derivatization/MALDI-MS
DOPA) was only observed in the nonlesioned sides of brains imaging technique makes it possible to simultaneously deter-
treated with D3-L-DOPA (Figure 2I). This is presumably due to mine the distributions of multiple known species by interrogating
the destruction of dopaminergic neurons, which are essential the data set from a single experiment in a targeted manner, it
for the uptake and conversion of L-DOPA to DA. The distribu- can also provide information on as-yet unidentified molecular
tion images for 3-MT showed that its concentration was high species.
in the intact sides of the brain but lower in the lesioned sides
(Figures 2J–2L). The average concentration of 3-MT in the non- ACh Imaging and Analysis of the Distribution of
lesioned sides of 6-OHDA-treated brains (Figures 2K and 2L) Cholinergic Neurons Using a Deuterated MALDI Matrix
was higher than that in sham-lesioned brains (Figure 2J). The While pyrylium derivatization provides a powerful tool for
metabolic conversion of D3-L-DOPA to D3-3-MT was traced studying primary amine neurotransmitters, some important
by measuring the concentration of D3-3-MT, which was highest neurotransmitters, such as ACh and alpha-GPC do not contain
in the nonlesioned side of the brain (Figure 2O); no interfering primary amines and so do not react with pyrylium cations. We
signal was detected in untreated brains (Figures 2M and 2N). therefore developed an alternative method for their imaging.
This experiment also showed that D3-L-DOPA treatment MALDI-MS analysis generally requires the homogeneous appli-
increased striatal GABA levels in the lesioned side of the brain cation of a matrix to the sample surface to facilitate the laser
(Figure 2R) relative to those in sham- and 6-OHDA-lesioned desorption ionization process. One of the most widely used
brain sections (Figures 2P and 2Q, respectively). Changes in matrices for MALDI-MS imaging of small molecules is CHCA.
GABAergic signaling throughout the basal ganglia (including Unfortunately, all matrices generate numerous readily detected
the striatum and the substantia nigra pars reticulate) after the fragments and cluster ions in the low molecular weight region
destruction of dopaminergic neurons and L-DOPA treatment of the mass spectrum, which can mask signals from target
have been extensively documented (Galeffi et al., 2003; Wang analytes such as ACh (m/z 146.1). One way of avoiding such
et al., 2007). Tyrosine is the biosynthetic precursor of the target mass interference is to use a deuterated CHCA (D4-
catecholamines and exhibited the same distribution as 3-MT CHCA) matrix (Shariatgorji et al., 2012), which shifts the inter-
and DA (Figures 2S–2U). fering signals by a minimum of 4 m/z units. This method was
Accurate mass FT-ICR-MS imaging was also performed on used to facilitate the imaging of molecular ACh and alpha-GPC
selected regions of coronal brain sections, including the caudate (m/z 258.1) and thereby monitor changes in their abundance
and putamen, of control and MPTP-lesioned primates (Figures in different parts of the brain during neurological events that
3A and 3B). The measured m/z values for selected neurotrans- affect the cholinergic system. ACh and alpha-GPC were identi-
mitters matched the corresponding theoretical values with fied by MS/MS fragmentation (Figure S4).
sub-ppm deviations (Table S2). DA (Figure 3C) and 3-MT (Fig- Cholinergic neurons are anatomically ubiquitous. However, it
ure 3E) were localized in the caudate and putamen regions of has been difficult to study their distribution in the CNS based
the brain. However, the concentrations of DA (Figure 3D) and on the presence of ACh because of difficulties in detecting and
3-MT (Figure 3F) in the MPTP-lesioned brain sections were quantifying ACh in situ. Instead, cholinergic neuron distributions
around 30 and 100 times lower, respectively, than that in the have been studied by examining markers such as acetylcholin-
controls (Figure S3). esterase (the enzyme that degrades ACh), muscarinic and nico-
Several reports have discussed the varied roles of 5-HT in PD tinic ACh receptors, and choline acetyltransferase (the enzyme
(Fox et al., 2009). By reanalyzing the data from the experiments that catalyzes ACh synthesis). By using a deuterated CHCA

Neuron 84, 697–707, November 19, 2014 ª2014 Elsevier Inc. 701
Neuron
Quantitative Molecular Imaging of Neurotransmitters

Figure 3. MALDI-MS Images of Neurotrans-


mitters in Coronal Primate PD Model Brain
Tissue Sections
(A–J) MALDI-MS images showing changes in
neurotransmitter levels in one hemisphere of a
coronal brain tissue sections from a control animal
(A) and an MPTP-lesioned animal (B). MALDI-FT-
ICR imaging of DA (C) and 3-MT (E) in control
brains showed that these compounds are most
abundant in the caudate and putamen structures.
Conversely, both DA (D) and 3-MT (F) are almost
absent in these regions after the destruction of
their dopaminergic neurons by MPTP-lesioning.
5-HT is mostly localized in the nucleus basalis
(Meynert) of the brains, and its concentration is
significantly lower in control brains (G) than in
MPTP-lesioned tissue sections (H). The 5-HT-rich
hypothalamic structure was also imaged in control
and MPTP-lesioned brain sections, revealing
that lesioning increased its 5-HT concentration.
The concentrations of GABA in the putamen and
caudate were significantly higher in the MPTP-
lesioned brain (J) than the control (I). All MS images
were acquired using a MALDI-FT-ICR mass
spectrometer. Data are shown using a rainbow
scale over a single range, normalized against the
total ion count. Scale bar, 10 mm; spatial resolu-
tion = 200 mm. Caudate, cd; putamen, Put. See
also Figure S3.

were also enriched in ACh, notably the


granule cell layer of the dentate gyrus
(DG-sg) and the pyramidal layer (sp) of
CA1 (Ammon’s horn) (Figures 4C and
4F). These subregions of the hippocam-
pus are delineated in Figure 4B, which
shows a MALDI-MS image generated
using data for a lipid with an m/z value
of 721.2.
The power of high spatial resolution MS
imaging was further demonstrated by
matrix, we were able to directly investigate the cholinergic studying the distribution of ACh in the basal ganglia of the brain.
neuron distribution in the brain based on the concentrations of In the striatum, ACh serves as the transmitter of the large aspiny
ACh in its various regions (Figure 4A). Imaging of rodent sagittal interneurons, which have diameters of 20–60 mm (Kawaguchi,
sections taken from different levels of the brain revealed high 1992) and comprise 1%–2% of the total striatal cell population
ACh concentrations in the frontal cortex, olfactory bulb, striatum, (Aosaki et al., 1998). While the cell bodies of the cholinergic inter-
hippocampus, medial habenula, tectum, thalamus, ventral neurons are relatively few in number, they densely innervate the
tegmental area, interpeduncular nucleus, pedunculopontine striatum with their extensive axonal arborizations (Holt et al.,
tegmental nucleus, and the laterodorsal tegmental nucleus. 2005). As shown in Figure 4D, the striatal ACh concentrations
This is consistent with previous findings (Hammond et al., 1994). can be measured by MALDI-MS imaging with a spatial resolution
of 15 mm.
High-Resolution MALDI-MS Imaging of ACh It is anticipated that the continuing development of MALDI-
High spatial resolution MALDI-MS imaging makes it possible to MS imaging technologies will improve to a point that enables
map the distributions of neurotransmitters within substructures the analysis of small sample areas of tissue sections at sub-
of the brain with high spatial resolution. Using our methodology, cellular resolution. Recently, it was demonstrated that MALDI-
we determined the relative abundance of ACh in various sub- MS instrumentation capable of imaging at resolutions < 1 mm
structures of the hippocampus, cerebellum, and striatum at a is technically feasible (Zavalin et al., 2012). Therefore, imaging
spatial resolution of 15 mm (Figures 4C–4E). The concentration neurotransmitters at subcellular spatial resolution in the near
of ACh was greater in the gray matter of cerebellum than in the future would lead to further biological insights and make the
white matter (Figure 4E). Certain subregions of the hippocampus method even more compelling.

702 Neuron 84, 697–707, November 19, 2014 ª2014 Elsevier Inc.
Neuron
Quantitative Molecular Imaging of Neurotransmitters

Figure 4. High-Resolution MALDI-MS Im-


ages of ACh in a Sagittal Rat Brain Section
(A–F) MALDI-MS images of ACh in a rodent brain
sagittal tissue section. The relative distribution
and abundance of ACh determined by MS imaging
are consistent with existing data on the major
cholinergic pathways in the brain (A). ACh-rich
structures such as the HIPP, CP, CTX, and even
small structures such as the nuclei of the facial
nerves and pons are readily apparent in the
MALDI-MS image. High-resolution MALDI-MS
images (15 mm spatial resolution) of various
structures in consecutive brain tissue sections
were also acquired. Substructures of the hippo-
campus were delineated by high-resolution im-
aging of the distribution of a lipid with m/z 721.2
(B). The relative abundance and distribution of
ACh in the HIPP (C), a subregion of the CP indi-
cated by a dashed rectangular border in (A) (D),
and CB (E) were determined by high-resolution
imaging. The concentration of ACh in the gray
matter of CB is around five times greater than that
in the white matter. In the HIPP, high ACh con-
centrations are apparent in DG-sg and sp regions
(F). MS images were acquired using a MALDI-
TOF/TOF mass spectrometer. Data are shown
using a rainbow scale and are normalized against
the total ion counts; scale bars, 2 mm, 1 mm, and
500 mm; spatial resolutions = 100 mm and 15 mm.
Caudate-putamen, CP; hippocampus, HIPP; cerebellum, CB; granule cell layer of the dentate gyrus, DG-sg; pyramidal layer of CA1, sp; cerebral cortex, CTX;
piriform area, PIR; striatum (ventral region), STRv; thalamus, TH; subiculum, SUB; stratum lacunosum-moleculare, Slm; stratum radiatum, Sr; stratum oriens, So;
cornu ammonis (field 2), CA2; cornu ammonis (field 3), CA3; polymorph layer of dentate gyrus, PO. See also Figure S4.

Imaging of ACh and Alpha-GPC in Brain Sections of extend the reach of MALDI-MS imaging, a technique with excel-
Tacrine-Administered Rodents lent mass accuracy and resolving power, allowing it to be used in
As a final illustration of the power of the deuterated matrix tech- the characterization of compounds that were previously prob-
nique, it was used to investigate the distributions of ACh (Figures lematic. Pyrylium derivatization converts low molecular weight
5A and 5B) and alpha-GPC (Figures 5C and 5D) in sagittal sec- endogenous primary amines into readily ionized species of rela-
tions of mouse brains from animals that had been treated with tively high molecular weight. This makes it possible to simulta-
10 mg/kg intraperitoneal tacrine (a centrally acting cholines- neously image tyrosine, tryptamine, tyramine, phenethylamine,
terase inhibitor) and untreated control animals. The concentra- DA, 3-MT, 5-HT, GABA, and Glu and other endogenous primary
tion of ACh in the tacrine-treated brains was approximately amines in a single MALDI-MS experiment. The use of a deuter-
seven times greater than that in the control brains, and the con- ated matrix allows one to sidestep problems caused by the
centrations of alpha-GPC within brain structures decreased as masking effect of matrix-derived ions in MALDI-MS analyses of
their ACh levels increased. In both the control (Figure 5A) and low molecular weight species, such as ACh, that lack a primary
tacrine-treated (Figure 5B) brain sections, the ACh concentration amine group. Because MALDI-MS imaging can be performed at
was determined for selected brain structures using a standard a spatial resolution as low as 15 mm, these technologies make
calibration curve created by spotting known quantities of deuter- it possible to measure the distributions of a wide range of neuro-
ated ACh (D9-ACh) onto consecutive control brain tissue sec- transmitters with high spatial resolution. The concentrations
tions (Figure S5). Absolute quantities of ACh (Figure 5E) and rela- of neurotransmitters in different subregions of the brain were
tive concentrations of alpha-GPC (Figure 5F) were determined quantified by comparing the intensities of their signals to those
for the whole brain, hippocampus, caudate putamen, thalamus, generated by known quantities of deuterated calibration stan-
and cerebral cortex regions in both control and tacrine-treated dards. The techniques presented herein permit the simultaneous
brains. The average measured concentration of ACh was consis- quantitation and specific molecular imaging of multiple neuro-
tent with previous results (Stavinoha and Weintraub, 1974). transmitters in situ. As such, they should find diverse and imme-
diate applications in fields such as neuroscience, pharmacology,
drug discovery, neurochemistry, and pathology.
Conclusions
The methods presented herein represent powerful tools for the
EXPERIMENTAL PROCEDURES
quantitative imaging of neurotransmitters, including biogenic
amines, amino acids, and ACh in histological brain tissue sec- All chemicals and reagents were purchased from Sigma-Aldrich unless other-
tions. Pyrylium derivatization and deuterated CHCA matrices wise stated and were used without further purification. Ketamine was obtained

Neuron 84, 697–707, November 19, 2014 ª2014 Elsevier Inc. 703
Neuron
Quantitative Molecular Imaging of Neurotransmitters

Figure 5. MALDI-MS Images of ACh and


Alpha-GPC Concentrations in Sagittal
Mouse Brain Sections from a Control Animal
and after Administration of 10 mg/kg of the
Cholinesterase Inhibitor Tacrine
(A–D) The absolute concentration of ACh in the
control brain section (A) is about seven times lower
than that observed following the injection of tacrine
(B). Conversely, the concentration of the ACh
precursor alpha-GPC is higher in the control
sample (C) than in the tacrine-treated brain (D). The
overall levels of alpha-GPC are reduced following
tacrine treatment. The absolute concentrations of
ACh in the control (A) and tacrine-treated (B)
mouse brains were quantified using a calibration
standard curve.
(E and F) Bar graphs showing the absolute con-
centration of ACh (E) and relative concentration of
alpha-GPC (F) with (black bars) and without (white
bars) administration of tacrine in whole brains and
selected structures. MS images were acquired
using a MALDI-TOF/TOF mass spectrometer. Data
are shown using a rainbow scale over a single
range, normalized against the total ion count. Scale
bar, 2 mm; spatial resolution = 100 mm. Caudate-
putamen, CP; hippocampus, HIPP; cerebral cor-
tex, CTX; thalamus, TH. See also Figure S5.

For D3-L-DOPA treatment experiments, 4 weeks


after the surgery, rats were treated with L-DOPA
(10 mg/kg, i.p.) and benserazide (7.5 mg/kg, i.p.)
once daily for 3 weeks. Animals were sacrificed
by decapitation 30 min after the last i.p. injection,
which was deuterated (D3)-L-DOPA (10 mg/kg)
and benserazide (7.5 mg/kg). Handling and storing
the brains was performed as described above.
from Parke-Davis, and xylazine was obtained from Bayer. Deuterated analogs Primate experiments were conducted using tissue from a previously pub-
of neurotransmitters and phenol-2,3,4,5,6-d5 (D5-phenol) were purchased lished brain bank (Fernagut et al., 2010; Porras et al., 2012; Santini et al.,
from CDN Isotopes. Water, methanol, and trifluoroacetic acid (TFA) were ob- 2010). Experiments were carried out in accordance with European Commu-
tained from Merck. nities Council Directive of November 24, 1986 (86/609/EEC) for care of labora-
tory animals in an AAALAC-accredited facility following acceptance of study
Animal Experiments design by the Institute of Lab Animal Science (Chinese Academy of Science,
Male Sprague-Dawley rats (approximately 150 g; Scanbur) and C57BL/6J Beijing, China) IACUC. Briefly, two female rhesus monkeys (Macaca mulatta,
male mice (3 months old, Charles River Laboratories) were housed in separate Xierxing; mean age = 5 ± 1 years; mean weight = 5.3 ± 0.8 kg) were used.
air-conditioned rooms (with a 12 hr dark/light cycle) at 20 C and a humidity of One animal was kept untreated as control. The other animal received daily
53%. Experiments were performed in agreement with the European Commu- MPTP (0.2 mg/kg, i.v.) according to previously published protocol (Bezard
nities Council Directive of November 24, 1986 (86/609/EEC) on the ethical use et al., 1997, 2001a, 2001b). Following stabilization of the MPTP-induced syn-
of animals and were approved by the local ethical committee at the Karolinska drome, animal behavior was assessed as previously published (Ahmed et al.,
Institute (N350/08). 2010; Fasano et al., 2010; Fernagut et al., 2010; Muñoz et al., 2008; Porras
Unilateral 6-OHDA lesioning of nigral dopaminergic axons was performed as et al., 2012). The degree of parkinsonism was assessed using a validated
described previously (Zhang et al., 2008). Briefly, rats were anesthetized with macaque clinical scale (Imbert et al., 2000). Animals were killed by sodium
ketamine (80 mg/kg, i.p.) and xylazine (10 mg/kg, i.p.) and pretreated with desi- pentobarbital overdose (150 mg/kg, i.v.), and brains were removed quickly
pramine (25 mg/kg, i.p.) and pargyline (5 mg/kg, i.p.). They were placed in a after death and immediately frozen by immersion in isopentane ( 45 C) and
stereotaxic instrument and injected with 6-OHDA (2.5 ml of a 5 mg/ml solution) stored at 80 C.
in the medial forebrain bundle (MFB) of the right hemisphere (AP = 2.8 mm, For the ACh experiments, mice that were 3 months old were injected with
ML = 2.0 mm, and V = 9.0 mm). saline or tacrine (10 mg/kg i.p.) and then sacrificed after 30 min according to
To determine the success of nigrostriatal denervation following unilateral the procedure described above.
6-OHDA lesioning, a single injection of apomorphine (1 mg/kg, i.p.) was admin-
istered to the rodents 2 weeks after their treatment with 6-OHDA, and their Tissue Section Preparation
resulting contralateral rotations were measured over 30 min. Only rats that The frozen brain tissues were cut using a cryostat-microtome (Leica
performed more than 100 turns were used in subsequent studies. They were CM3050S, Leica Microsystems). Sagittal and coronal brain sections were
killed by decapitation 4 weeks after the surgery, after which their brains cut at a thickness of 14 mm. Tissue sections were transferred by thaw mounting
were removed, frozen in dry ice-cooled isopentane, and stored at 80 C until onto conductive indium tin oxide (ITO) glass slides (Bruker Daltonics) and
needed. then stored at 80 C. Sections were desiccated at room temperature for

704 Neuron 84, 697–707, November 19, 2014 ª2014 Elsevier Inc.
Neuron
Quantitative Molecular Imaging of Neurotransmitters

15 min prior to spotting of calibration standards, after which they were imaged set according to the manufacturer’s recommendations for optimal acquisi-
optically using a photo scanner (Epson perfection V500). The samples were tion performance. The laser spot size was selected to yield intermediate
then coated with matrix for ACh experiments or derivatized with or without and sharp levels of focus (40 mm and 10 mm laser spot diameters) for
matrix coating for primary amine imaging experiments. To enable the quanti- low and high spatial resolution analyses. The laser power was optimized at
tation of individual neurotransmitters, deuterated ACh, DA, or GABA dissolved the start of each run and then held constant during the MALDI-MS imaging
in 50% methanol was spotted onto the control tissue sections to serve as experiment. On-tissue and on-plate MS/MS fragmentation experiments
calibration standards prior to derivatization and matrix application. were performed in positive ion mode using the MALDI-TOF/TOF according
to the manufacturer’s recommended settings. Tissue sections were analyzed
Derivatization of Calibration Standards and Endogenous in a random order to prevent any possible bias due to factors such as matrix
Primary Amines degradation or variation in mass spectrometer sensitivity. MS imaging
DPP-TFB and TMP-TFB were dissolved in methanol to prepare 1 mg/ml stock data were visualized using FlexImaging (Bruker Daltonics), version 3.0 for
solutions. Derivatization was performed using solutions containing 150 ml of TOF/TOF data and version 4.0 for FT-ICR data. Mass spectra analysis and
the stock solution of DPP-TFB or TMP-TFB in 1.5 ml of 50% methanol, chemical formula calculations were performed in Data Analysis, version 4.2
buffered by 1 ml triethylamine (TEA). The derivatization solution was applied (Bruker Daltonics). An in-house-developed software package was used for
to the tissue sections using an automatic sprayer (ImagePrep, Bruker Dalton- data processing, normalization, and quantitation. Regions of interest were
ics). To facilitate the derivatization reaction and to avoid delocalization of the manually defined in the analysis software using both the optical image and
endogenous compounds, reagents were added using a three-step process MS imaging data (Källback et al., 2012). The concentration of the target
involving spraying, incubation, and drying, which lasted for 2.5, 15, and 50 s, compounds in defined regions of interest were calculated by correlating their
respectively. The prepared slides were then incubated for 60 min in a chamber signal intensities to a calibration curve derived from deuterated analogs of
saturated with the vapor arising from a 50% methanol solution. To acidify the the analytes spotted on control tissue sections considering the brain density
tissue sections, 1 ml of a solution containing acetic acid, water, and methanol as 1.0 g/cm3 (Barber et al., 1970).
(10:45:45) was sprayed over the tissue using the same setup as above. The
treated tissue sections were then incubated once more in the methanol
Autoradiographic Studies
vapor-saturated chamber for 30 min. Primate and D3-L-DOPA-treated rat
Sections that were used to study the distribution of DAT (a dopamine trans-
tissue sections were analyzed using a high concentration of DPP-TFB as reac-
porter) were preincubated in 50 mM Tris-HCl and 120 mM NaCl (pH 7.5) for
tive matrix. DPP-TFB was dissolved in 1.2 ml of methanol to prepare 8 mg/ml
20 min and then incubated for 1 hr in the same buffer supplemented with
of the stock solution. This solution was diluted in 6 ml of 70% methanol con-
50 pM [125I] RTI-55 in the presence of 1 mM fluoxetine (a selective serotonin
taining 3.5 ml of TEA. An automated pneumatic sprayer (TM-Sprayer, HTX
reuptake inhibitor). The slides were washed twice for 10 s each in ice-cold
Technologies) was used to spray warm reagent over the dried samples. The
binding buffer, rapidly dipped in deionized water, and dried. The sections
nozzle temperature was set at 80 C, and the reagent was sprayed (30 passes)
were then exposed to Kodak BioMax MR films for 2 days.
over the tissue sections at a linear velocity of 110 cm/min with a flow rate of
about 80 ml/min. Samples were then incubated for 15 min (dried by nitrogen
flow every 5 min) in a chamber saturated with the vapor arising from a 50% SUPPLEMENTAL INFORMATION
methanol solution. Stock solutions of DPP-TFB and TMP-TFB (2 mg/ml in
methanol) and standards of DA, GABA, and Glu (1 mg/ml in methanol) were Supplemental Information includes five figures and two tables and can be
used for in-solution derivatization experiments. Stock solutions of neuro- found with this article online at http://dx.doi.org/10.1016/j.neuron.2014.10.011.
transmitters (2 ml) and derivatization reagents (10 ml) were added to the solvent
containing 90 ml of 50% methanol, buffered by 0.1 ml TEA. The mixture was
AUTHOR CONTRIBUTIONS
incubated for 30 min at room temperature, followed by addition of 1 ml of acetic
acid. Samples were spotted (0.5 ml) to dry on stainless steel plates.
M.S. conceived the project and its essential ideas, assisted with the
MALDI-MS experiments and data evaluation, and contributed to discus-
Matrix Coating and MALDI-MS Imaging Data Acquisition
sions. A.N. assisted with the MALDI-MS experiments and data evaluation
CHCA (0.5 ml of 10 mg/ml in 50% ACN, 0.2% TFA) was spotted over dried
and contributed to discussions. N.S. conducted animal (mouse) and DAT
derivatized neurotransmitters on stainless steel plates. D4-CHCA was pre-
experiments and contributed to discussions. R.J.A.G. contributed to data
pared as reported previously (Shariatgorji et al., 2012). The MALDI-MS matrix
evaluation and discussions. X.Z. conducted animal (rat) experiments and
was applied to the tissue sections as follows. A 10 ml solution of 10 mg/ml
contributed to discussions. P.K. developed the quantitation software and
CHCA (for analysis of tissues with low concentration of derivatization re-
contributed to discussions. A.R.C and E.B. supervised the primate experi-
agent) or D4-CHCA (for analysis of ACh) in 50% ACN containing 0.2% TFA
ments and contributed to discussions. P.S. supervised the rodent experi-
was sprayed onto the tissues sections using a TLC sprayer. The sprayer
ments and contributed to discussions. P.E.A. supervised the overall
was maintained at a distance of 30 cm from the target during matrix coating,
research, conceived the project and essential ideas, assisted with the
and sections were allowed to dry between coating cycles. For high spatial
data evaluations, and contributed to discussions. All authors contributed
resolution (HSR) MALDI-MS imaging of ACh and low spatial resolution
to the writing of the manuscript.
(LSR) matrix-assisted imaging experiments, D4-CHCA and CHCA (5 mg/ml
dissolved in 50% ACN containing 0.2% TFA) were applied using an auto-
matic sprayer (TM-Sprayer, HTX Technologies). All parameters were opti- ACKNOWLEDGMENTS
mized in order to reach the best sensitivity and minimize delocalization of
target compounds. The nozzle temperature was set at 98 C (for HSR exper- The authors acknowledge Dr. Matthias Witt and Patrik Ek (Bruker Daltonics) for
iments) and 90 C (for LSR experiments). The matrix was sprayed (eight assisting with FT-ICR-MS imaging and Dr. Andras Szabo and members of
passes for HSR and six passes for LSR experiments) over the tissue sections Ubichem laboratory for synthesis of deuterated matrix. This work was
at a linear velocity of 110 cm/min with a flow rate of about 70 ml/min. The supported by the Swedish Research Council (Medicine and Health 2013-
quantity of matrix solution applied in this way was measured using an optical 3105, Natural and Engineering Science 2010-5421, and Research Infrastruc-
sensor present in the automatic sprayer (ImagePrep, Bruker Daltonics) to ture 2009-6050 to P.E.A.), AstraZeneca R&D, UK (to P.E.A. and A.N.),
ensure that the same amount of matrix was applied to each slide. All Agence Nationale de la Recherche (ANR-12-BSV4-0001-01 to E.B.), and the
MALDI-MS imaging experiments were performed using either MALDI-TOF/ Michael J. Fox Foundation (to E.B.).
TOF (Ultraflextreme, Bruker Daltonics) or MALDI-FT-ICR (Solarix XR 12T,
Bruker Daltonics) mass spectrometer in positive ion mode using a Smart- Accepted: September 29, 2014
beam II 2 kHz laser. The mass spectrometer’s operating parameters were Published: November 6, 2014

Neuron 84, 697–707, November 19, 2014 ª2014 Elsevier Inc. 705
Neuron
Quantitative Molecular Imaging of Neurotransmitters

REFERENCES reverts motor symptoms associated with L-dopa-induced dyskinesia. Proc.


Natl. Acad. Sci. USA 107, 21824–21829.
Ahmed, M.R., Berthet, A., Bychkov, E., Porras, G., Li, Q., Bioulac, B.H., Carl, Fernagut, P.O., Li, Q., Dovero, S., Chan, P., Wu, T., Ravenscroft, P., Hill, M.,
Y.T., Bloch, B., Kook, S., Aubert, I., et al. (2010). Lentiviral overexpression Chen, Z.W., and Bezard, E. (2010). Dopamine transporter binding is unaffected
of GRK6 alleviates L-dopa-induced dyskinesia in experimental Parkinson’s by L-DOPA administration in normal and MPTP-treated monkeys. PLoS ONE
disease. Sci. Transl. Med. 2, 28ra28. 5, e14053.
Alreja, M., Shanabrough, M., Liu, W.M., and Leranth, C. (2000). Opioids sup- Fox, S.H., Chuang, R., and Brotchie, J.M. (2009). Serotonin and Parkinson’s
press IPSCs in neurons of the rat medial septum/diagonal band of Broca: disease: On movement, mood, and madness. Mov. Disord. 24, 1255–1266.
involvement of mu-opioid receptors and septohippocampal GABAergic
Galeffi, F., Bianchi, L., Bolam, J.P., and Della Corte, L. (2003). The effect of
neurons. J. Neurosci. 20, 1179–1189.
6-hydroxydopamine lesions on the release of amino acids in the direct and
Aosaki, T., Kiuchi, K., and Kawaguchi, Y. (1998). Dopamine D1-like receptor
indirect pathways of the basal ganglia: a dual microdialysis probe analysis.
activation excites rat striatal large aspiny neurons in vitro. J. Neurosci. 18,
Eur. J. Neurosci. 18, 856–868.
5180–5190.
Hammond, P., Rao, R., Koenigsberger, C., and Brimijoin, S. (1994). Regional
Badgaiyan, R.D. (2011). Neurotransmitter Imaging: Basic Concepts and
variation in expression of acetylcholinesterase mRNA in adult rat brain
Future Perspectives. Curr Med Imaging Rev 7, 98–103.
analyzed by in situ hybridization. Proc. Natl. Acad. Sci. USA 91, 10933–10937.
Barber, T.W., Brockway, J.A., and Higgins, L.S. (1970). The density of tissues
Herzog, E., Gilchrist, J., Gras, C., Muzerelle, A., Ravassard, P., Giros, B.,
in and about the head. Acta Neurol. Scand. 46, 85–92.
Gaspar, P., and El Mestikawy, S. (2004). Localization of VGLUT3, the vesicular
Bezard, E., Imbert, C., Deloire, X., Bioulac, B., and Gross, C.E. (1997). glutamate transporter type 3, in the rat brain. Neuroscience 123, 983–1002.
A chronic MPTP model reproducing the slow evolution of Parkinson’s disease:
Holt, D.J., Bachus, S.E., Hyde, T.M., Wittie, M., Herman, M.M., Vangel, M.,
evolution of motor symptoms in the monkey. Brain Res. 766, 107–112.
Saper, C.B., and Kleinman, J.E. (2005). Reduced density of cholinergic inter-
Bezard, E., Dovero, S., Prunier, C., Ravenscroft, P., Chalon, S., Guilloteau, neurons in the ventral striatum in schizophrenia: an in situ hybridization study.
D., Crossman, A.R., Bioulac, B., Brotchie, J.M., and Gross, C.E. (2001a). Biol. Psychiatry 58, 408–416.
Relationship between the appearance of symptoms and the level of nigros-
Huot, P., Lévesque, M., and Parent, A. (2007). The fate of striatal dopami-
triatal degeneration in a progressive 1-methyl-4-phenyl-1,2,3,6-tetrahydro-
nergic neurons in Parkinson’s disease and Huntington’s chorea. Brain 130,
pyridine-lesioned macaque model of Parkinson’s disease. J. Neurosci. 21,
222–232.
6853–6861.
Bezard, E., Ravenscroft, P., Gross, C.E., Crossman, A.R., and Brotchie, J.M. Imbert, C., Bezard, E., Guitraud, S., Boraud, T., and Gross, C.E. (2000).
(2001b). Upregulation of striatal preproenkephalin gene expression occurs Comparison of eight clinical rating scales used for the assessment of
before the appearance of parkinsonian signs in 1-methyl-4-phenyl- 1,2,3,6- MPTP-induced parkinsonism in the Macaque monkey. J. Neurosci. Methods
tetrahydropyridine monkeys. Neurobiol. Dis. 8, 343–350. 96, 71–76.

Boja, J.W., Patel, A., Carroll, F.I., Rahman, M.A., Philip, A., Lewin, A.H., Johannesen, S.A., Beeren, S.R., Blank, D., Yang, B.Y., Geyer, R., and
Kopajtic, T.A., and Kuhar, M.J. (1991). [125I]RTI-55: a potent ligand for dopa- Hindsgaul, O. (2012). Glycan analysis via derivatization with a fluorogenic
mine transporters. Eur. J. Pharmacol. 194, 133–134. pyrylium dye. Carbohydr. Res. 352, 94–100.

Boulet, S., Mounayar, S., Poupard, A., Bertrand, A., Jan, C., Pessiglione, M., Jones, B.E., and Beaudet, A. (1987). Distribution of acetylcholine and cate-
Hirsch, E.C., Feuerstein, C., François, C., Féger, J., et al. (2008). Behavioral cholamine neurons in the cat brainstem: a choline acetyltransferase and
recovery in MPTP-treated monkeys: neurochemical mechanisms studied by tyrosine hydroxylase immunohistochemical study. J. Comp. Neurol. 261,
intrastriatal microdialysis. J. Neurosci. 28, 9575–9584. 15–32.
Bressan, R.A., and Pilowsky, L.S. (2000). Imaging the glutamatergic system Källback, P., Shariatgorji, M., Nilsson, A., and Andrén, P.E. (2012). Novel mass
in vivo—relevance to schizophrenia. Eur. J. Nucl. Med. 27, 1723–1731. spectrometry imaging software assisting labeled normalization and quantita-
tion of drugs and neuropeptides directly in tissue sections. J. Proteomics
Caprioli, R.M., Farmer, T.B., and Gile, J. (1997). Molecular imaging of biological
75, 4941–4951.
samples: localization of peptides and proteins using MALDI-TOF MS. Anal.
Chem. 69, 4751–4760. Kawaguchi, Y. (1992). Large aspiny cells in the matrix of the rat neostriatum
Ciliax, B.J., Heilman, C., Demchyshyn, L.L., Pristupa, Z.B., Ince, E., Hersch, in vitro: physiological identification, relation to the compartments and excit-
S.M., Niznik, H.B., and Levey, A.I. (1995). The dopamine transporter: immuno- atory postsynaptic currents. J. Neurophysiol. 67, 1669–1682.
chemical characterization and localization in brain. J. Neurosci. 15, 1714– Keenan, C.L., and Koopowitz, H. (1981). Limitations in identifying neurotrans-
1723. mitters within neurons by fluorescent histochemistry techniques. Science 214,
Cornett, D.S., Reyzer, M.L., Chaurand, P., and Caprioli, R.M. (2007). MALDI 1151–1152.
imaging mass spectrometry: molecular snapshots of biochemical systems. Muñoz, A., Li, Q., Gardoni, F., Marcello, E., Qin, C., Carlsson, T., Kirik, D., Di
Nat. Methods 4, 828–833. Luca, M., Björklund, A., Bezard, E., and Carta, M. (2008). Combined 5-HT1A
de Jong, L.A., Uges, D.R., Franke, J.P., and Bischoff, R. (2005). Receptor- and 5-HT1B receptor agonists for the treatment of L-DOPA-induced dyski-
ligand binding assays: technologies and applications. J. Chromatogr. B nesia. Brain 131, 3380–3394.
Analyt. Technol. Biomed. Life Sci. 829, 1–25. Pimlott, S.L., and Sutherland, A. (2011). Molecular tracers for the PET and
Decavel, C., and Van den Pol, A.N. (1990). GABA: a dominant neurotransmitter SPECT imaging of disease. Chem. Soc. Rev. 40, 149–162.
in the hypothalamus. J. Comp. Neurol. 302, 1019–1037. Porras, G., Berthet, A., Dehay, B., Li, Q., Ladepeche, L., Normand, E., Dovero,
El Mestikawy, S., Wallén-Mackenzie, A., Fortin, G.M., Descarries, L., and S., Martinez, A., Doudnikoff, E., Martin-Négrier, M.L., et al. (2012). PSD-95
Trudeau, L.E. (2011). From glutamate co-release to vesicular synergy: vesicu- expression controls L-DOPA dyskinesia through dopamine D1 receptor
lar glutamate transporters. Nat. Rev. Neurosci. 12, 204–216. trafficking. J. Clin. Invest. 122, 3977–3989.
Falck, B., Thieme, G., Hillarp, N.A., and Torp, A. (1962). Fluorescence of Qin, C., and Luo, M. (2009). Neurochemical phenotypes of the afferent and
catechol amines and related compounds condensed with formaldehyde. efferent projections of the mouse medial habenula. Neuroscience 161,
J. Histochem. Cytochem. 10, 348–354. 827–837.
Fasano, S., Bezard, E., D’Antoni, A., Francardo, V., Indrigo, M., Qin, L., Doveró, Quirke, J.M.E., Adams, C.L., and Vanberkel, G.J. (1994). Chemical
S., Cerovic, M., Cenci, M.A., and Brambilla, R. (2010). Inhibition of Ras- Derivatization for Electrospray-Ionization Mass-Spectrometry 0.1. Alkyl-
guanine nucleotide-releasing factor 1 (Ras-GRF1) signaling in the striatum Halides, Alcohols, Phenols, Thiols, and Amines. Anal. Chem. 66, 1302–1315.

706 Neuron 84, 697–707, November 19, 2014 ª2014 Elsevier Inc.
Neuron
Quantitative Molecular Imaging of Neurotransmitters

Santini, E., Sgambato-Faure, V., Li, Q., Savasta, M., Dovero, S., Fisone, G., Tritsch, N.X., Oh, W.J., Gu, C., and Sabatini, B.L. (2014). Midbrain dopamine
and Bezard, E. (2010). Distinct changes in cAMP and extracellular signal-regu- neurons sustain inhibitory transmission using plasma membrane uptake of
lated protein kinase signalling in L-DOPA-induced dyskinesia. PLoS ONE 5, GABA, not synthesis. Elife 3, e01936.
e12322. Wang, H., Katz, J., Dagostino, P., and Soghomonian, J.J. (2007). Unilateral 6-
Schaaf, J.M., Teelken, A.W., Muskiet, F.A., Nagel, G., and Wolthers, B.G. hydroxydopamine lesion of dopamine neurons and subchronic L-DOPA
(1985). A specific and sensitive determination of gamma aminobutyric acid administration in the adult rat alters the expression of the vesicular GABA
in CSF and brain tissue by gas chromatography-mass spectrometry. transporter in different subsets of striatal neurons and in the substantia nigra,
J. Neurosci. Methods 13, 257–265. pars reticulata. Neuroscience 145, 727–737.
Schwamborn, K., and Caprioli, R.M. (2010). Molecular imaging by mass Ye, H., Wang, J., Greer, T., Strupat, K., and Li, L. (2013). Visualizing neurotrans-
spectrometry—looking beyond classical histology. Nat. Rev. Cancer 10, mitters and metabolites in the central nervous system by high resolution and
639–646. high accuracy mass spectrometric imaging. ACS Chem Neurosci 4, 1049–
Shariatgorji, M., Nilsson, A., Goodwin, R.J., Svenningsson, P., Schintu, N., 1056.
Banka, Z., Kladni, L., Hasko, T., Szabo, A., and Andren, P.E. (2012). Zavalin, A., Todd, E.M., Rawhouser, P.D., Yang, J., Norris, J.L., and Caprioli,
Deuterated matrix-assisted laser desorption ionization matrix uncovers R.M. (2012). Direct imaging of single cells and tissue at sub-cellular spatial res-
masked mass spectrometry imaging signals of small molecules. Anal. olution using transmission geometry MALDI MS. J. Mass Spectrom. 47, 1473–
Chem. 84, 7152–7157. 1481.
Sotnikova, T.D., Beaulieu, J.M., Espinoza, S., Masri, B., Zhang, X.D., Zhang, X., Andren, P.E., Chergui, K., and Svenningsson, P. (2008). Neurokinin
Salahpour, A., Barak, L.S., Caron, M.G., and Gainetdinov, R.R. (2010). The B/NK3 receptors exert feedback inhibition on L-DOPA actions in the
dopamine metabolite 3-methoxytyramine is a neuromodulator. PLoS ONE 5, 6-OHDA lesion rat model of Parkinson’s disease. Neuropharmacology 54,
e13452. 1143–1152.
Stavinoha, W.B., and Weintraub, S.T. (1974). Choline content of rat brain. Zhao, X.E., and Suo, Y.R. (2008). Simultaneous determination of mono-
Science 183, 964–965. amine and amino acid neurotransmitters in rat endbrain tissues by pre-
Sugiura, Y., Zaima, N., Setou, M., Ito, S., and Yao, I. (2012). Visualization of column derivatization with high-performance liquid chromatographic
acetylcholine distribution in central nervous system tissue sections by tandem fluorescence detection and mass spectrometric identification. Talanta 76,
imaging mass spectrometry. Anal. Bioanal. Chem. 403, 1851–1861. 690–697.

Neuron 84, 697–707, November 19, 2014 ª2014 Elsevier Inc. 707

Вам также может понравиться