ArticlePDF Available

Chemogenetic stimulation of tonic locus coeruleus activity strengthens the default mode network

Authors:

Abstract and Figures

The default mode network (DMN) of the brain is functionally associated with a wide range of behaviors. In this study, we used functional magnetic resonance imaging (fMRI), positron emission tomography (PET), and spectral fiber photometry to investigate the selective neuromodulatory effect of norepinephrine (NE)-releasing noradrenergic neurons in the locus coeruleus (LC) on the mouse DMN. Chemogenetic-induced tonic LC activity decreased cerebral blood volume (CBV) and glucose uptake and increased synchronous low-frequency fMRI activity within the frontal cortices of the DMN. Fiber photometry results corroborated these findings, showing that LC-NE activation induced NE release, enhanced calcium-weighted neuronal spiking, and reduced CBV in the anterior cingulate cortex. These data suggest that LC-NE alters conventional coupling between neuronal activity and CBV in the frontal DMN. We also demonstrated that chemogenetic activation of LC-NE neurons strengthened functional connectivity within the frontal DMN, and this effect was causally mediated by reduced modulatory inputs from retrosplenial and hippocampal regions to the association cortices of the DMN.
Content may be subject to copyright.
Oyarzabal et al., Sci. Adv. 8, eabm9898 (2022) 29 April 2022
SCIENCE ADVANCES | RESEARCH ARTICLE
1 of 12
NEUROSCIENCE
Chemogenetic stimulation of tonic locus coeruleus
activity strengthens the default mode network
Esteban A. Oyarzabal1,2,3,4†, Li-Ming Hsu1,2,3†, Manasmita Das1,2,3†,
Tzu-Hao Harry Chao1,2,3, Jingheng Zhou5, Sheng Song1,2,3, Weiting Zhang1,2,3,
Kathleen G. Smith6, Natale R. Sciolino6, Irina Y. Evsyukova6, Hong Yuan2, Sung-Ho Lee1,2,3,
Guohong Cui5, Patricia Jensen6, Yen-Yu Ian Shih1,2,3*
The default mode network (DMN) of the brain is functionally associated with a wide range of behaviors. In this
study, we used functional magnetic resonance imaging (fMRI), positron emission tomography (PET), and spectral
fiber photometry to investigate the selective neuromodulatory effect of norepinephrine (NE)–releasing noradrenergic
neurons in the locus coeruleus (LC) on the mouse DMN. Chemogenetic-induced tonic LC activity decreased cere-
bral blood volume (CBV) and glucose uptake and increased synchronous low-frequency fMRI activity within the
frontal cortices of the DMN. Fiber photometry results corroborated these findings, showing that LC-NE activation
induced NE release, enhanced calcium-weighted neuronal spiking, and reduced CBV in the anterior cingulate
cortex. These data suggest that LC-NE alters conventional coupling between neuronal activity and CBV in the
frontal DMN. We also demonstrated that chemogenetic activation of LC-NE neurons strengthened functional con-
nectivity within the frontal DMN, and this effect was causally mediated by reduced modulatory inputs from retro-
splenial and hippocampal regions to the association cortices of the DMN.
INTRODUCTION
Functional magnetic resonance imaging (fMRI) has been widely
used to demonstrate the presence of spatiotemporally consistent in-
trinsic functional brain networks during resting state. The default
mode network (DMN), composed of the prefrontal, orbitofrontal,
prelimbic, cingulate, retrosplenial, posterior parietal, and temporal
association cortices as well as the dorsal hippocampus, is among the
most robust intrinsic networks because of its highly synchronized
activity in the absence of cognitive tasks or saliency (1). The DMN
is vulnerable in several neurological and neuropsychiatric disorders
(2), is functionally associated with a wide range of behaviors (3),
integrates interoceptive and exteroceptive information from multiple
brain networks (4), and maintains the brain in a semivigilant state
(5). To make causal interpretations of behaviorally relevant DMN
changes and design network-based interventions for disorders that
afflict DMN activity, identifying the modulatory mechanisms con-
trolling the DMN is of paramount importance.
The locus coeruleus (LC), a small nucleus within the pons, is a
potential DMN modulator (6,7). A large portion of the neuro-
modulator norepinephrine (NE) originates from the LC and is re-
leased in the brain regions that are considered DMN nodes (8,9).
Accumulating evidence suggests that LC-NE may be essential for
DMN modulation because (i) NE receptors are prominently expresse d
in DMN-related brain structures (9); (ii) LC-NE can bidirectionally
modulate attention reorientation in a dose-dependent manner (8);
(iii) LC-NE neuron degeneration and DMN disruption are coincidently
found in depression (10), traumatic brain injury (11), Parkinson’s
disease (12), Alzheimer’s disease (13), and aging (14); and (iv)
pharmacological treatment of pathological LC-NE levels reduces
attentional lapses (15) and restores DMN integrity in attention defi-
cit hyperactivity disorder (ADHD) patients (16). Despite these findings ,
the modulatory association between LC-NE and the DMN remains
circumstantial because pharmacological interventions using NE-
related agents inherently result in nonselective binding on dopaminergic,
cholinergic, and serotonergic receptors (7,17,18). Furthermore,
systemic administration of these agents indiscriminately targets all
NE-producing neurons in the brain and sympathetic nervous system,
making it difficult to determine the role of LC-NE in modulating the
DMN. Although selective manipulation of LC-NE while imaging
the DMN is currently impossible in humans due to technical and
ethical constraints (7,19), such studies are feasible in rodent models
because structural and functional homologs of the human DMN have
been identified in mice (2028).
In this study, we used an established data-driven approach to
identify DMN modules (29) and an intersectional chemogenetic
strategy to selectively and reproducibly induce tonic LC-NE activity
in mice (30,31). To reveal potential confounders that could affect
our interpretations of LC-NE influence on the DMN, we measured
changes in several fMRI metrics, neuronal calcium activity, and glu-
cose uptake across different spatial and temporal scales. Through
modeling the signal dynamics, we revealed the circuit mechanism
by which LC-NE activation modulates the DMN. Our findings should
pave the way toward a better understanding of how large-scale
brain networks are mediated by a specific neuromodulatory system.
RESULTS
To selectively and reproducibly activate LC-NE neurons, we used
an intersectional chemogenetic approach in which the excitatory G
protein (heterotrimeric GTP-binding protein)–coupled receptor
hM3Dq, fused to mCherry, is expressed in 99.6% of the anatomically
1Center for Animal MRI, University of North Carolina, Chapel Hill, NC, USA. 2Biomedical
Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA.
3Department of Neurology, University of North Carolina, Chapel Hill, NC, USA.
4Curriculum in Neurobiology, University of North Carolina, Chapel Hill, NC, USA. 5In
Vivo Neurobiology Group, Neurobiology Laboratory, NIEHS/NIH, Research Triangle
Park, NC, USA. 6Developmental Neurobiology Group, Neurobiology Laboratory,
NIEHS/NIH, Research Triangle Park, NC, USA.
*Corresponding author. Email: shihy@unc.edu
†These authors contributed equally to this work.
Copyright © 2022
The Authors, some
rights reserved;
exclusive licensee
American Association
for the Advancement
of Science. No claim to
original U.S. Government
Works. Distributed
under a Creative
Commons Attribution
NonCommercial
License 4.0 (CC BY-NC).
Oyarzabal et al., Sci. Adv. 8, eabm9898 (2022) 29 April 2022
SCIENCE ADVANCES | RESEARCH ARTICLE
2 of 12
defined LC located within the central gray and a small portion of the
dorsal subcoeruleus immediately adjacent to and continuous with the
LC (LC-NE/hM3Dq) (Fig.1) (31). This population of NE neurons
robustly innervates canonical DMN regions including cingulate 1
(Cg1) and retrosplenial (RSC) cortices (fig. S1) (32).
To functionally delineate the DMN and study selective NE mod-
ulatory effects, we performed invivo cerebral blood volume (CBV)–
weighted fMRI scans on LC-NE/hM3Dq mice and littermate controls
(n=9 and 12, respectively) under light isoflurane (~1%) anesthesia
using a previously described isotropic echo planar imaging (EPI)
protocol (33). Although medetomidine with low-dose isoflurane is
considered the preferred sedative for rodent fMRI (34), we avoided
its usage because it suppresses NE release (35). We collected a
10-min resting-state baseline scan before administering clozapine-
n- oxide (CNO; 1 mg/kg, intraperitoneally) (Fig.2A). CNO was
selected because it was previously used for behavior studies of the
same mouse line (30,31). This protocol has been shown to activate
LC-NE neurons at tonic frequency and to suppress locomotion in
LC-NE/hM3Dq mice (30,31). Subsequent comparisons were made
against littermate controls to account for off-target effects of CNO
and/or the back-metabolized clozapine (36). We spatially warped each
imaging dataset into the Allen Mouse Common Coordinate Framework
(fig. S2), functionally parcellated the baseline fMRI data from all
subjects (n = 21) by performing a 100-component independent
component analysis (ICA) (Fig.2, Band C), and verified their re-
producibility (fig. S3). We identified 17 DMN-related independent
components (ICs) (fig. S4A) according to previous rodent DMN
studies (22,24,25,28,29,37). The areas showing significant tempo-
ral correlation associated with the 17 identified DMN ICs were re-
constructed using dual regression (DR), and a one-sample two-sided
t test was performed to generate the group-level maps representing
the connectivity of these ICs. These maps showed high spatial simi-
larity with an RSC seed-based connectivity map commonly used to
depict DMN (fig. S4, B to D). Louvain community modularity anal-
ysis clustered the 17 DMN ICs into three distinct modules (Q=0.10,
P<0.01; Fig. 2D): a Frontal module composed of prelimbic/
infralimbic (PrL/IL), lateral orbital (LO), Cg1, and anterior cingu-
late 2 (aCg2) cortices; an RSC-HIPP module composed of the dorsal
hippocampus (HIPP) and posterior Cg2 (pCg2), medial parietal
association (MPtA), retrosplenial granular (RSG), and retrosplenial
dysgranual/visual (RSD/Vis) cortices; and an Association module
composed of posterior parietal (PPtC) and auditory (Aud) cortices.
No significant difference among the connectivity of DMN ICs was
found in the pre-CNO baseline data between LC-NE/hM3Dq and
control groups (PFDR-corrected>0.05; fig. S4E).
Using the Frontal, RSC-HIPP, and Association module regions
of interest (one-sample two-sided t test, P<0.0001) and striatum as
a reference region due to sparse innervation from LC-NE neurons,
we examined how NE release from LC modulates fMRI-derived CBV,
regional homogeneity (ReHo), and amplitude of low-frequency
fluctuation (ALFF) changes. We compared the 10-min pre-CNO
baseline fMRI data against data acquired between 20- and 30-min
post-CNO. We selected a later phase of the CNO response to avoid
the transition time period that has been shown to have greater
intrasubject variability and weaker behavioral effects in designer
receptor exclusively activated by designer drugs induced activity
kinetics as observed by fMRI (6,3842) and behavior studies
(30,31). CNO-evoked LC-NE activation significantly decreased CBV
from pre-CNO baseline to post-CNO in all DMN modules in LC-NE/
hM3Dq. These changes are significant (two-sided two-sample t test,
PFDR-corrected<0.005) compared to control mice (Fig.3A). LC-NE
activation induced robust CBV changes in the striatum despite sparse
innervation from LC-NE neurons (6), possibly because of NE-induced
vasoconstriction at watershed arteries upstream of the striatum (43).
While CBV changes appeared less specific, ReHo (Fig.3B) and ALFF
(Fig.3C) changes were more localized, and the increase of these
signals contradicted the intuitive interpretation of CBV, suggesting
a possible increase of synchronous, low-frequency activity in the
Frontal and RSC-HIPP DMN modules by LC-NE. Specifically,
LC-NE activation enhanced ALFF changes within 0.01- and 0.05-Hz
band (unpaired t test, PFDR-corrected<0.05; fig. S5A).
To validate these fMRI findings, we used spectral fiber photometry
(Fig.4A) in LC-NE/hM3Dq (n =5) and control mice (n =4). We
virally expressed a genetically engineered NE2.1 sensor (44) and a
red-shifted jRGECO1a calcium activity sensor (45) under the pan-
neuronal human Synapsin-1 (hSyn) promoter in the Cg1 of the
Frontal DMN module (Fig.4B), where the effects of LC-NE activa-
tion were most robust, and intravenously administered a CY5-
conjugated dextran far-red fluorescent dye to measure CBV (fig. S6).
Collectively, this allowed us to simultaneously detect changes in syn-
aptic NE release, neuronal activity–mediated calcium influx, and
CBV in Cg1 (Fig.4C). After CNO administration, we observed sig-
nificant increases in NE release (unpaired t test, PFDR-corrected<0.05;
Fig. 1. Intersectional chemogenetic strategy to selectively activate LC-NE neu-
rons. (A) Schematic illustration of the intersectional genetic strategy. (B) A sagittal
schematic diagram of the hindbrain compressed along the mediolateral axis illus-
trates the approximate position of NE neurons. Recombination of the RC::FL-hM3Dq
allele by the noradrenergic-specific driver DbhFlpo and En1cre results in expression
of the excitatory G protein–coupled receptor hM3Dq fused to mCherry in LC-NE
neurons (magenta cells in schematic). Expression of DbhFlp by all remaining NE
neurons results in expression of green fluorescent protein (GFP) (green neurons in
schematic). (C) Immunofluorescent labeling of sections from the adult brainstem
of LC-NE/hM3Dq mice reveals hM3Dq-mCherry–expressing NE neurons in the LC
(magenta) and GFP-expressing NE neurons (green) in the SubC, A5, C2/A2, and C1/
A1 nuclei. Scale bars, 50 m.
Oyarzabal et al., Sci. Adv. 8, eabm9898 (2022) 29 April 2022
SCIENCE ADVANCES | RESEARCH ARTICLE
3 of 12
Fig. 4D), decreases in CBV (unpaired t test, PFDR-corrected <0.05;
Fig.4E), and increases in the neuronal activity (unpaired t test,
PFDR-corrected<0.05; Fig.4F) of LC-NE/hM3Dq mice compared to
controls. Notably, the number of calcium spikes also increased signifi-
cantly following CNO administration (unpaired t test, PFDR-corrected<
0.05; Fig.4G), a hallmark of NE that tunes the signal-to-noise ratio
of downstream neuronal firing (46). These findings corroborate well
with our fMRI results (Fig.3) and indicate that driving LC-NE re-
lease can concurrently induce regional vasoconstriction while enhancing
neuronal excitability in Cg1. Given that fMRI does not directly
measure neuronal activity, these results highlight the importance to
cautiously interpret fMRI-derived DMN results when NE is involved,
as inferring neuronal activity by direct fMRI signal changes in this
case may be erroneous.
We also examined the effect of NE on brain glucose uptake in a sub-
set of LC-NE/hM3Dq (n=5) and control (n=5) mice using an
established 18F-fluorodeoxyglucose (FDG) positron emission tomog-
raphy (PET) protocol (47). We found that CNO-induced LC-NE acti-
vation significantly decreased glucose uptake in all three DMN
modules (paired t test, PFDR-corrected<0.001), but not in control subjects
(Fig.5 and fig. S7). Although there are mixed findings on how NE alters
glucose uptake (4853), many studies using pharmacology to enhance
NE release have also shown glucose uptake suppression (4853) but
are often difficult to disambiguate. One possible mechanism
governing this effect is a shift in metabolic pathway by astrocytes to
preferentially use glycogen reserves (51,52). Together, these find-
ings suggest that additional caution needs to be considered for FDG
PET data interpretation when triggering NE release.
To access how selective NE release modulates DMN connectivity
and network properties, we examined functional connectivity (FC)
changes within and between Frontal, RSC-HIPP, and Association
DMN modules in LC-NE/hM3Dq (n=9) and control mice (n=12).
We found that CNO significantly enhanced FC within Frontal
(t=2.89, PFDR-corrected< 0.05) and between Frontal and Association
modules (t=2.63, PFDR-corrected<0.05) of the DMN in LC-NE/hM3Dq,
but not in control mice (Fig.6, Aand B). Notably, the high intra-
modular connectivity characterized by within-module degree (WD),
which quantifies the level of node connectivity within a module, indi-
cated that Cg1 and RSC serve as provincial hubs for the Frontal and
RSC-HIPP modules of the DMN, respectively (fig. S8, A and B). The
high intermodular connectivity characterized by the partition coef-
ficient (PC), which estimates the level of interaction with nodes of
other modules, indicated that the aCg2 of the Frontal module serves
as a connector hub throughout the entire DMN and may control the
FC between Frontal and other modules (fig. S8, A and B). Given
the putative causal control of the anterior insular (AI) cortex on the
Fig. 2. Experimental design and identification of mouse DMN modules. (A) CBV fMRI experimental design includes a 10-min baseline scan before CNO administration
and 30 min of scans after CNO. i.p., intraperitoneally. (B) One hundred ICs were derived from group ICA of baseline scans among all subjects. Specific IC masks were de-
termined by a winner-take-all strategy by comparing mean z values from all ICs on a voxel basis and then color-coded. DMN-related ICs were then identified according to
rodent DMN topology in the literature. (C) Correlation among the 100 ICs was plotted with a threshold Fischer |z| > 0.3. DMN ICs were labeled in red. (D) Modularity anal-
ysis of DMN ICs showing that the mouse DMN is composed of Frontal (yellow), RSC-HIPP (pink), and Association modules (blue).
Oyarzabal et al., Sci. Adv. 8, eabm9898 (2022) 29 April 2022
SCIENCE ADVANCES | RESEARCH ARTICLE
4 of 12
Frontal module of the DMN (26,54), we also examined the effects of
LC-NE activation on their FC changes and found that CNO-evoked
activation significantly enhanced anticorrelation between AI and
DMN frontal module (fig. S8, C and D).
To further unravel the causal influences on FC in DMN modules by
LC-NE activation, we conducted a dynamic causal modeling (DCM)
analysis and found that LC-NE activation significantly reduced effec-
tive connectivity (EC) from RSC-HIPP to the Association module
(paired two-sided t test, PFDR-corrected<0.05; Fig.7A), whereas no
change was detected in the littermate controls (fig. S9A). Coactivation
pattern (CAP) (55) analyses may support these findings, as the CAP
representing distinct RSC-HIPP and Association states was also sup-
pressed following LC-NE activation (fig. S9, B to D). Together with
the robust NE modulatory effects in the Frontal module, these findings
prompted us to conduct a mediation analysis to determine the origin
of the FC changes in the Frontal module. A moderation analysis model
was constructed using the structural equation modeling method in
AMOS 17.0. We first demonstrated that the reduction in EC observed
from RSC-HIPP to the Association module causally manipulates the
FC changes within the Frontal module (coefficient=−0.53±0.13,
P< 0.001; Fig.7B). Then, when we incorporated the FC increases
between the Frontal and Association modules as a mediator, we ob-
served a full mediation effect of the reduced EC to the Frontal module
FC changes, while the direct relationship between the reduced EC
from RSC-HIPP to the Association module and the increased FC
within the Frontal module became insignificant (coefficient= −0.26±
0.13, no significance). The Sobel test further indicated the significant
mediation effect (Sobel z=−2.02, P<0.05; Fig.7B). In contrast, we
did not observe the reduced EC from the RSC-HIPP to Association
module being mediated by the FC increases between Frontal and Asso-
ciation modules (fig. S10A). Collectively, these findings indicate that
LC-NE activation modulates the DMN by (i) strengthening FC within
the Frontal module, (ii) strengthening FC between Frontal and Asso-
ciation modules, and (iii) reducing RSC-HIPP control over the Asso-
ciation module, which causally alters Frontal module FC, with the FC
between Frontal and Association modules serving as a key mediator.
Fig. 3. CBV, ReHo, and ALFF changes in DMN modules following activation of LC-NE neurons. (A) CBV decreased (post-CNO, baseline) significantly in LC-NE/hM3Dq
following LC-NE activation across all DMN modules compared to controls. LC-NE activation significantly increased (B) ReHo change in the Frontal DMN module and
(C) ALFF changes in Frontal and RSC-HIPP modules compared to controls. *PFDR-corrected < 0.005; horizontal dotted lines represent means, and error bars represent ±SD. The
brain maps indicate the significant difference of CBV, ReHo, and ALFF changes between LC-NE/hM3Dq and controls (P3dClustSim-corrected < 0.05).
Oyarzabal et al., Sci. Adv. 8, eabm9898 (2022) 29 April 2022
SCIENCE ADVANCES | RESEARCH ARTICLE
5 of 12
Fig. 4. Triple-spectral fiber photometry measuring NE release, CBV, and calcium-weighted neuronal activity in Cg1. (A) Experimental setup of the fiber photometry
system with 488-, 561-, and 640-nm lasers simultaneously used to detect NE release (NE2.1), neuronal calcium activity (jRGECO1a), and CBV (CY5-dextran dye) changes,
respectively. i.v., intravenously. (B) Cg1 neurons were confirmed to be transfected to express NE2.1 (green) and jRGECO1a (red) sensors. (C) Spectral profiles of NE neuronal
activity and CBV sensor emissions used to resolve signals via an established spectral unmined approach. (D to F) Respective effects of CNO on NE release, CBV, and neu-
ronal activity in Cg1 from representative subjects and group level bar graphs. Subjects were continuously recorded for 40 min with a dose of CNO (1 mg/kg) administered
via an intraperitoneal catheter at 10 min (t = 0) after scan onset. (G) CNO-induced LC-NE activation altered postsynaptic calcium spiking patterns, resulting in a significant
increase in spike counts. Red dots represent the Ca2+ spikes (z score > 1.96). *P < 0.05 and **P < 0.01; horizontal lines represent means, and error bars represent ±SD.
Oyarzabal et al., Sci. Adv. 8, eabm9898 (2022) 29 April 2022
SCIENCE ADVANCES | RESEARCH ARTICLE
6 of 12
DISCUSSION
The mouse DMN is well documented in the literature (2025,28),
and its structural foundation has been recently established by a piv-
otal study by Whitesell etal. (25). Most studies analyzing the mouse
DMN using ICA or seed-based connectivity analyses report consist-
ent DMN architectures that are homologous to the human DMN. A
multicenter study compiled by Grandjean etal. (22) analyzed several
resting-state mouse fMRI datasets acquired under various condi-
tions (e.g., magnetic field strengths, coils, imaging parameters, and
anesthesia protocols) and generated a group ICA atlas delineating
mouse brain connectivity. The results pulled three DMN modules
that include prefrontal, cingulate/retrosplenial, and temporal asso-
ciation areas. Similar to those findings, our data analysis revealed
three modules including Frontal, RSC-HIPP, and Association modules,
where the Frontal and RSC-HIPP modules included prefrontal and
posterior cingulate components, respectively (Fig.2C). Note that
the involvement of the hippocampus in the mouse DMN remains
disputed because of the lack of direct anatomical projections
(22,24,25). This study includes hippocampus as part of the DMN
because we followed established studies identifying the DMN con-
stituents across rodents (29,37) and humans (4). In addition, sever-
al unbiased hierarchical clustering analyses (24,56), including our
own (29,33), functionally classified the hippocampus as part of the
DMN and lend further support to the validity of the DMN regions
used for the analysis in this study. Aside from hippocampus, note
that thalamus has been recently shown to alter large-scale cortical
rhythms and affects fMRI-derived functional connectivity in the
cortex, including several putative DMN regions (57). Given the robust
Fig. 5. Glucose uptake changes in DMN modules following activation of LC-NE neurons. (A) Activation of LC-NE neurons significantly decreased FDG uptake com-
pared to saline-treated sham and littermate controls receiving CNO. (B) LC-NE activation significantly reduced standardized uptake values (SUVs) in all DMN regions. SUV
changes were derived from each subject that underwent two scans (vehicle and CNO administration) following co-registration. Comparisons were made against litter-
mate controls. *P < 0.001 and **P < 0.0001; horizontal dotted line represents the mean, and error bars represent ±SD.
Fig. 6. FC changes among DMN modules following activation of LC-NE neurons. (A) Within and between DMN module FC changes. (B) Network-based sta-
tisti cal analysis showing significant differences in FC among edges in LC-NE/hM3Dq > Control. *PFDR-corrected < 0.05; horizontal dotted lines represent means, and error
bars represent ±SD.
Oyarzabal et al., Sci. Adv. 8, eabm9898 (2022) 29 April 2022
SCIENCE ADVANCES | RESEARCH ARTICLE
7 of 12
LC innervations into thalamus (32), it is crucial for future studies to
examine network interactions among LC, thalamus, and DMN.
Several research groups have pioneered chemogenetic fMRI
(6,3842) and PET (47) approaches to selectively map the influence
of neurotransmitters on brain networks in rodents. Our experimen-
tal design using an intersectional chemogenetic mouse line presented
a unique opportunity to selectively investigate functional DMN
modulation. It is our hope that through dissemination of raw data,
this work, together with a seminal study by Zerbi etal. (6), will help
set the foundation for future studies examining other network sys-
tems manipulated by LC-NE.
We demonstrated that chemogenetic activation of LC-NE neu-
rons significantly reduced CBV and glucose uptake among all three
DMN modules compared to littermate controls (Figs.3and5).
While CBV measures may suffer from systemic effects from LC-NE
activation on sympathetic outputs (58), our study used an array of
multimodal techniques to help interpret the influence of LC-NE ac-
tivation on DMN. We found that directional CBV changes are not
a proper metric to explain network activity or connectivity changes.
Future studies should incorporate central and peripheral NE levels
as confounders of hemodynamic-based brain mapping techniques.
Both fMRI and fiber photometry corroborated the findings of
CBV reduction in the Frontal DMN module following activation of
LC-NE neurons. Concurrently, we observed robust increases in
synchronous low-frequency activity as measured by ReHo, ALFF,
and photometry-derived calcium activity in our experimental con-
dition (Fig.4,FandG). The most straightforward interpretation of
the fMRI data based on well-documented neurovascular coupling
rules (59) does not apply, likely because of the potent vasoconstric-
tive properties of LC-NE (60). It is not surprising that the neuro-
modulatory effect induced by NE release appears inconsistent in the
literature owing to various brain states and basal firing rate examined,
as well as the distinct experimental approaches used to promote/
benchmark NE release (6,6163). The literature also shows mixed
findings regarding the effects of NE on both cerebral hemodynamics
and metabolism that are difficult to disambiguate because common
pharmacological agents used to induce NE release suffer from dif-
ferential actions on NE receptor subtypes and nonselective binding
(64) that can either increase (65) or decrease perfusion (60,66) and
increase (48,53) or decrease glucose metabolism (49,50). One possible
mechanism governing the observed changes in glucose metabolism
following LC-NE stimulation could also be attributed to a dose-
dependent metabolic shift in astrocytes to preferentially use glyco-
gen reserves (51,52). Unlike many studies that use pharmacological
manipulations of NE release, our chemogenetic approach coupled
with multimodal measurement of NE, neuronal activity, and CBV
revealed an effect of NE in increasing synchronized low-frequency
activity, strengthening neuronal firing, and decreasing CBV. This has
substantial implications when using fMRI to interpret DMN neuro-
nal activity, as “deactivation” of raw fMRI signal caused by LC-NE
activation may not necessarily represent reduced DMN neuronal act ivity.
Recent animal and human fMRI studies support the role of NE
in brain network reorganization (6,62,63,67). NE release has been
shown to promote topological integration within the network (68,69).
This aligns well with our findings showing enhanced ReHo and FC
within the Frontal DMN module following CNO-evoked LC-NE
activation. In addition, our ALFF results show that LC-NE activa-
tion enhanced low-frequency power of the Frontal DMN that was
restricted to a frequency band ranging from 0.01 to 0.05 Hz—a
range associated with robust changes in human DMN (70). Alter-
ations in the power within this subfrequency band of ALFF also
plays a vital role in attention reorientation during visual-motor
attentional tasks (70), which has also been linked to changes in cor-
tical NE levels (71).
Fig. 7. DCM and mediation analysis among DMN modules. (A) DCM analysis among DMN modules found that RSC-HIPP reduced its causal modulation to the Association
module upon LC-NE activation (PFDR-corrected < 0.05; horizontal dotted lines represent means, and error bars represent ±SD). (B) Mediation analysis was performed with
direct effect (a, b, and c) and with a mediator (c′). A Sobel test was also performed (red dotted line) to evaluate the significance of mediation effect (Sobel z value = −2.02,
P < 0.05). *P < 0.05, **P < 0.01, and ***P < 0.001; n.s., no significance.
Oyarzabal et al., Sci. Adv. 8, eabm9898 (2022) 29 April 2022
SCIENCE ADVANCES | RESEARCH ARTICLE
8 of 12
LC-NE enhanced FC within the Frontal module and between
Frontal and Association modules. In agreement with our finding, a
seminal study virally transfecting hM3Dq via the Dbh promotor
into the LC found significant FC enhancement in the Cg1, Cg2, and
RSC following chemogenetic stimulation (6). Abnormally height-
ened FC in Frontal DMN regions has been associated with anxiety
and depression (72). Anxiety-like behavioral phenotypes such as
reduced locomotor activity in a novel environment and anhedonia
have also been found following chemogenetic-evoked LC-NE acti-
vation in awake mice (30). Such a behavior response is similar to
optogenetic-induced LC-NE neuronal activation at tonic frequencies
(73). The intersectional chemogenetic strategy used in the current
study has been shown to result in approximately 2Hz of tonic firing
(31), comparable to that in (74). Unlike phasic LC bursting that in-
duces arousal by desynchronizing cortical electroencephalography
(EEG) states (75), the low tonic LC firing induced by CNO as seen
in our experimental condition has been shown to strengthen theta
and suppress delta power (74) and does not trigger sufficient arousal
activity under anesthesia (76). Together with our data showing
tonically elevated NE release (Fig.4D) and strengthened DMN con-
nectivity (Fig.6), these results suggest that tonic firing of LC-NE
neurons shifts the brain toward a DMN-dominated state and there-
fore facilitates DMN-associated behaviors (77).
As we did not observe any FC decreases in the DMN and only
identified strengthening FC within and between modules, our re-
sults support the functional integration theory of NE proposed by
Shine (68), complemented by the results of Zerbi etal. (6), and
suggest that LC-NE–induced functional integration could occur at
a rather focal, subnetwork level within the DMN. In addition, we
found that FC strengthened anticorrelated coupling between the
Frontal DMN module and AI, a key node of the salience network
(SN) that may causally suppress the DMN (fig. S8, C and D) (54).
This also indicates that integration between the DMN and SN is
greater following LC-NE activation under our experimental condi-
tion. This finding may suggest a putative role of tonic LC-NE activity
in improving efficient cognitive control and reducing behavioral
variability by fostering SN-DMN anticorrelation (78). In addition,
our findings point to the possibility that tonic and phasic outputs
from LC-NE neurons may preferentially drive DMN and SN, re-
spectively. Furthermore, the enhanced anticorrelation between net-
works also supports the plausible mechanism by which NE-targeting
pharmacological agents like atomoxetine, clonidine, and guanfacine
are effective in treating ADHD patients because they strengthen the
FC within the Frontal DMN module and result in improved atten-
tion (79).
Our DCM and mediation analyses show that enhanced Frontal
module connectivity in the DMN was causally manipulated by re-
ducing RSC-HIPP control of the Association module, with the con-
nectivity between the Association and Frontal modules serving as a
key mediator. These findings reveal a new understanding of how
LC-NE activation controls the signaling cascades within DMN modules
and achieves its control of the Frontal cortical regions, which are
among the most well-studied projection targets of LC-NE because
of their importance in shaping multiple behaviors (80). LC-NE neu-
ronal loss and the subsequent depletion of cortical NE levels are
widely considered to be among the first sites of neurodegeneration
in Parkinson’s and Alzheimer’s diseases (81), resulting in behavioral
pathologies linked to alterations in DMN such as delayed attention
shifting (82), enhanced mind-wandering (83), and reduced cognitive
and emotional processing of sensory information (82,83). Optogenetic-
induced LC-NE activation at lower tonic frequencies into prefrontal
and orbitofrontal cortices enhances stimulus and goal-directed
attention with decreased impulsivity (84). Conversely, the suppres-
sion of LC-NE activation exacerbates distractibility and impulsivity
(84), similar to that observed in Parkinson’s and Alzheimer’s dis-
ease patients (83). Together, alterations within Frontal and between
Frontal and RSC-HIPP DMN modules have potential to serve as
early biomarkers for pathophysiological changes in LC-NE neurons.
Our circuit-level findings could also pave the way toward novel tar-
gets to causally control the Frontal DMN via the RSC-HIPP module
when LC neurons have degenerated such that the behavioral traits
relevant to the Frontal DMN may be restored when endogenous NE
is pathologically diminished.
MATERIALS AND METHODS
Animals
All animal procedures were performed in strict compliance with
ethical regulations for animal research and approved by the Institu-
tional Animal Care and Use Committee of the University of North
Carolina at Chapel Hill. En1cre (85), DbhFlpo (32), and RC::FL-hM3Dq
(31) mouse colonies are maintained on a C57BL/6J background.
Male and female triple-transgenic animals were generated at the
National Institute of Environmental Health Sciences by crossing
En1cre mice to double-transgenic DbhFlpo;RC::FL-hM3Dq mice. Single-
and double-transgenic littermates served as controls essential for rigor
in chemogenetic studies (86) because off-target binding of CNO or
clozapine through reverse metabolism of CNO may occur (36). All
animals were maintained on a 12-hour/12-hour light-dark cycle
with access to food and water ad libitum.
CBV-fMRI acquisition
For fMRI studies, LC-NE (n=9) and control mice (n =12) were
initially anesthetized using 2 to 3% isoflurane and maintained un-
der light anesthesia (1% isoflurane) while preserving physiological
homeostasis. All MRI experiments were performed on a Bruker
BioSpec 9.4-T, 30-cm bore system (Bruker BioSpin Corp., Billerica,
MA) with ParaVision 6.0.1 on an AVANCE II console. An RRI BFG
150/90 gradient insert (Resonance Research Inc., Billerica, MA) paired
with a Copley C700 gradient amplifier (Copley Controls Corp., Canton,
MA) was used for all experiments. A 72-mm-volume coil was used
as the transmitter, and a quadrature mouse brain coil was used as
the receiver (Bruker BioSpin Corp., Billerica, MA). Magnetic field
homogeneity was optimized first by global shimming, followed by
local second-order shims using a MAPSHIM protocol. All CBV-fMRI
data were acquired using a two-dimensional (2D) multislice, single-
shot, gradient-echo EPI sequence: TR (repetition time)=3000 ms,
TE (echo time)=7.9 ms, bandwidth=250 kHz, flip angle=70°, FOV
(field of view)=19.2mm × 19.2 mm, matrix size=64 × 64, slice
number=26, slice thickness=0.3 mm, resulting in an isotropic
voxel size of 0.3mm3. Subjects were continuously recorded for
40min with a dose of CNO (1 mg/kg) administered via an intraper-
itoneal catheter 10min after scan onset. CBV-weighted fMRI was
achieved by a bolus dose of an in-house–developed iron oxide
nanoparticle (30mg of Fe/kg, intravenously) (87). Rectal body tem-
peratures were continuously maintained at 37°±0.5°C with a tem-
perature controller (Oakton Temp9500, Cole-Parmer, Vernon Hills,
IL, USA) coupled to a circulating water bath (Haake S13, Thermo
Oyarzabal et al., Sci. Adv. 8, eabm9898 (2022) 29 April 2022
SCIENCE ADVANCES | RESEARCH ARTICLE
9 of 12
Fisher Scientific, Waltham, MA, USA) that heats the MRI mouse
cradle. Respiration was monitored through a pneumatic pillow
(Respiration/EEG Monitor, SA Instruments, Stony Brook, NY, USA)
and maintained between 90 and 110 breaths/min through fine ad-
justments in the inhaled isoflurane concentration.
Physiological parameters were monitored during fMRI. The only
change observed following CNO administration was an increase in
respiratory rate, which rapidly recovered within a few seconds and
did not follow the sustained CBV changes measured by fMRI and
photometry. Moreover, these changes were observed in both LC-NE/
hM3Dq and control groups, indicating that it is likely because of the
well-known injection volume stress.
fMRI data analysis
Preprocessing of images
All fMRI data were corrected for slice timing and motion using Anal-
ysis of Functional NeuroImages. Brain data were isolated using a
U-Net deep-learning skull stripping tool and spatially normalized
to our EPI template using Advanced Normalization Tools. In addi-
tion, despiking, ICA denoising, and nuisance variable regression of
the six motion parameters estimated from motion correction and the
cerebrospinal fluid signal extracted using a mask of the major brain
ventricles were performed. Datasets were then smoothed using a
Gaussian kernel with full width at half maximum (FWHM) at 0.6 mm,
detrended, and temporally filtered by applying a high-pass filter
between >0.01Hz. Datasets underwent quality control by measur-
ing frame-wise displacement (FD), temporal signal-to-noise ratio
(tSNR), and DVARS (temporal derivative of the root mean square
variance over voxels). A detailed description of the preprocessing
pipeline (56) can be found in Supplementary Methods.
ICA and modularity analyses
MRI data were decomposed into 100 functional components using
baseline data from all subjects via a group-level ICA (FSL MELODIC).
Functional modules of the identified 17 DMN components were
parcellated using the Louvain community detection algorithm.
Within- and between-module connectivity was then defined as the
average of FC across node pairs within or between the identified DMN
modules. The ICA and modularity analysis method are detailed in
Supplementary Methods.
DCM and mediation analysis
We specified a DCM model with full connectivity consisting of
three modules from DMN to estimate pairwise EC among the DMN
modules and constructed a directed and weighted graph (representing
an EC network) for each subject. We applied serial multiple media-
tion analysis model in AMOS 17.0 (SPSS Inc., Chicago, IL, USA) to
uncover underlying functional pathways within DMN. Specifically,
we first estimated the direct relationships between dependent vari-
able (EC from RSC-HIPP module to Association module) and inde-
pendent variable (FC within Frontal module). Then, in a mediation
model, the FC between Association and Frontal module was added
as a mediator. In this context, full mediation occurs when the rela-
tionship between the independent variable and the dependent variable
is no longer significant with the inclusion of a mediator variable.
Detailed data analysis is further described in Supplementary Methods.
FDG PET procedure
Mice were fasted 12hours before undergoing 18F-FDG PET scans to
reduce variability in blood glucose levels that could alter 18F-FDG
uptake (47). Static PET scans were collected on the same cohort of
animal over two scan sessions using LC-NE/hM3Dq (n=5) and
control (n=6) mice to represent sham-treated baseline or CNO-
treated condition. Mice were briefly anesthetized under 1 to 3% iso-
flurane and injected with either a saline + dimethyl sulfoxide
(DMSO) vehicle or CNO dissolved in DMSO (1 mg/kg, intraperito-
neally) and subsequently received an intravenous injection of
0.2 mCi of 18F-FDG after 5min. Mice were recovered in their home
cages for a 45-min uptake period. Mice were subsequently anesthe-
tized with isoflurane (2%) and underwent a 10-min computed to-
mography (CT) and 20-min PET scan on a small-animal PET/CT
scanner (Argus-2R, Sedecal, Madrid, Spain). PET data were recon-
structed using the 2D ordered subset expectation maximization
(OSEM) algorithm expressed in standardized uptake values (SUVs)
and normalized using arm muscle uptake of 18F-FDG using PMOD
(PMOD Technologies LLC, Zurich, Switzerland). Data were repre-
sented as % changes in SUV between vehicle and CNO scans.
Fiber photometry procedure
Surgical adeno-associated virus (AAV) microinjection and
fiber implantation
LC-NE (n=5) and control (n =5) mice were microinjected
with 0.3 l of AAV5-hSyn-NE2.1 (h-N01, WZ Biosciences) and
0.5 l of AAV9-hSyn-jRGECO1a (100854, Addgene) to the left Cg1
(Anterior- Posterior=2.2 mm, Medial-Lateral=0.2 mm, Dorsal-
Ventral =−1.6 mm). NE2.1 (a green fluorescent NE sensor) and
jRGECO1a (a red-shifted intracellular calcium sensor) were used
for determining the NE release and neuronal activity, respectively.
An optic fiber was implanted 0.3mm above the injection site
and imbedded to the skull using cement (C&B Metabond,
S380, Parkell).
Fiber photometry recording
All recordings began at least 4 weeks after surgery. CY5-conjugated
dextran fluorescent dye (20 mg/kg; R-FN-006, RuixiBio) was injected
intravenously for CBV measurements. Animals were prepared and
maintained under the same conditions as fMRI experiments. A spectral
fiber photometry system capable of recording NE2.1, jRGECO1a,
and CY5 signals was simultaneously recorded during the experiments.
Detailed methods can be found in Supplementary Methods.
Immunohistology procedure
Mice were deeply anesthetized with sodium pentobarbital and tran-
scardially perfused with 0.1M phosphate-buffered saline (PBS)
followed by 4% paraformaldehyde (PFA). Brains were postfixed
overnight by immersion in 4% PFA at 4°C. Following a rinse in
PBS, brains were cryoprotected in 30% sucrose in PBS and embedded
in Tissue Freezing Medium. Forty-micrometer free-floating coronal
cryosections were collected in PBS and processed for immunohisto-
chemistry according to previously published protocol (32). Briefly,
free-floating sections were blocked in 5% normal goat serum in PBS
with 0.1% Triton X-100 for 1hour before incubating in primary
antibody overnight at 4°C. All NE cell bodies were labeled with rabbit
anti-tyrosine hydroxylase (TH) antibody (AB152, Millipore). hM3Dq-
mCherry-expressing NE neurons were labeled with rat anti-mCherry
primary antibody (EST202, Kerafast) and green fluorescent protein
(GFP)–expressing NE neurons were labeled with chicken anti-GFP
primary antibody (AB13970, Abcam). Sections were washed three
times in PBS and incubated for 2 hours in Alexa Fluor 648 anti-rabbit,
Alexa Fluor 568 anti-rat, and Alexa Fluor 488 anti-chicken secondary
antibodies (Invitrogen). Sections were mounted onto glass slides,
Oyarzabal et al., Sci. Adv. 8, eabm9898 (2022) 29 April 2022
SCIENCE ADVANCES | RESEARCH ARTICLE
10 of 12
coverslipped with Vectashield hard-set mounting medium with
4′,6-diamidino-2-phenylindole (DAPI) (H-1500, Vector Labs), and
imaged on a Zeiss LSM 880 inverted confocal microscope.
SUPPLEMENTARY MATERIALS
Supplementary material for this article is available at https://science.org/doi/10.1126/
sciadv.abm9898
REFERENCES AND NOTES
1. M. E. Raichle, The brain’s default mode network. Annu. Rev. Neurosci. 38, 433–447 (2015).
2. R. L. Buckner, J. R. Andrews-Hanna, D. L. Schacter, The brain’s default network: Anatomy,
function, and relevance to disease. Ann. N. Y. Acad. Sci. 1124, 1–38 (2008).
3. J. R. Andrews-Hanna, J. S. Reidler, J. Sepulcre, R. Poulin, R. L. Buckner, Functional-
anatomic fractionation of the brain’s default network. Neuron 65, 550–562 (2010).
4. K. Christoff, Z. C. Irving, K. C. R. Fox, R. N. Spreng, J. R. Andrews-Hanna, Mind-wandering
as spontaneous thought: A dynamic framework. Nat. Rev. Neurosci. 17, 718–731 (2016).
5. M. F. Mason, M. I. Norton, J. D. van Horn, D. M. Wegner, S. T. Grafton, C. N. Macrae,
Wandering minds: The default network and stimulus-independent thought. Science 315,
393–395 (2007).
6. V. Zerbi, A. Floriou-Servou, M. Markicevic, Y. Vermeiren, O. Sturman, M. Privitera,
L. von Ziegler, K. D. Ferrari, B. Weber, P. P. De Deyn, N. Wenderoth, J. Bohacek, Rapid
reconfiguration of the functional connectome after chemogenetic locus coeruleus
activation. Neuron 103, 702–718.e5 (2019).
7. M. Kelberman, S. Keilholz, D. Weinshenker, What’s that (blue) spot on my MRI? Multimodal
neuroimaging of the locus coeruleus in neurodegenerative disease. Front. Neurosci. 14,
583421 (2020).
8. S. J. Sara, S. Bouret, Orienting and reorienting: The locus coeruleus mediates cognition
through arousal. Neuron 76, 130–141 (2012).
9. R. L. van den Brink, T. Pfeffer, T. H. Donner, Brainstem modulation of large-scale intrinsic
cortical activity correlations. Front. Hum. Neurosci. 13, 340 (2019).
10. X. Zhu, Q. Zhu, H. Shen, W. Liao, F. Yuan, Rumination and default mode network
subsystems connectivity in first-episode, drug-naive young patients with major
depressive disorder. Sci. Rep. 7, 43105 (2017).
11. D. J. Sharp, C. F. Beckmann, R. Greenwood, K. M. Kinnunen, V. Bonnelle, X. de Boissezon,
J. H. Powell, S. J. Counsell, M. C. Patel, R. Leech, Default mode network functional
and structural connectivity after traumatic brain injury. Brain 134, 2233–2247 (2011).
12. M. Pievani, N. Filippini, M. P. van den Heuvel, S. F. Cappa, G. B. Frisoni, Brain connectivity
in neurodegenerative diseases—Fom phenotype to proteinopathy. Nat. Rev. Neurol. 10,
620–633 (2014).
13. P. Wang, B. Zhou, H. Yao, Y. Zhan, Z. Zhang, Y. Cui, K. Xu, J. Ma, L. Wang, N. An, X. Zhang,
Y. Liu, T. Jiang, Aberrant intra- and inter-network connectivity architectures in Alzheimer’s
disease and mild cognitive impairment. Sci. Rep. 5, 14824 (2015).
14. B. B. Biswal, M. Mennes, X.-N. Zuo, S. Gohel, C. Kelly, S. M. Smith, C. F. Beckmann,
J. S. Adelstein, R. L. Buckner, S. Colcombe, A.-M. Dogonowski, M. Ernst, D. Fair,
M. Hampson, M. J. Hoptman, J. S. Hyde, V. J. Kiviniemi, R. Kötter, S.-J. Li, C.-P. Lin,
M. J. Lowe, C. Mackay, D. J. Madden, K. H. Madsen, D. S. Margulies, H. S. Mayberg,
K. McMahon, C. S. Monk, S. H. Mostofsky, B. J. Nagel, J. J. Pekar, S. J. Peltier, S. E. Petersen,
V. Riedl, S. A. R. B. Rombouts, B. Rypma, B. L. Schlaggar, S. Schmidt, R. D. Seidler,
G. J. Siegle, C. Sorg, G.-J. Teng, J. Veijola, A. Villringer, M. Walter, L. Wang, X.-C. Weng,
S. Whitfield-Gabrieli, P. Williamson, C. Windischberger, Y.-F. Zang, H.-Y. Zhang,
F. X. Castellanos, M. P. Milham, Toward discovery science of human brain function.
Proc. Natl. Acad. Sci. U.S.A. 107, 4734–4739 (2010).
15. A. Smith, D. Nutt, Noradrenaline and attention lapses. Nature 380, 291 (1996).
16. E. Hoekzema, S. Carmona, J. A. Ramos-Quiroga, V. Richarte Fernández, R. Bosch,
J. C. Soliva, M. Rovira, A. Bulbena, A. Tobeña, M. Casas, O. Vilarroya, An independent
components and functional connectivity analysis of resting state fMRI data points
to neural network dysregulation in adult ADHD. Hum. Brain Mapp. 35, 1261–1272 (2014).
17. F. P. Bymaster, J. S. Katner, D. L. Nelson, S. K. Hemrick-Luecke, P. G. Threlkeld,
J. H. Heiligenstein, S. M. Morin, D. R. Gehlert, K. W. Perry, Atomoxetine increases
extracellular levels of norepinephrine and dopamine in prefrontal cortex of rat:
A potential mechanism for efficacy in attention deficit/hyperactivity disorder.
Neuropsychopharmacology 27, 699–711 (2002).
18. A. Gobert, J. M. Rivet, L. Cistarelli, C. Melon, M. J. Millan, Alpha2-adrenergic receptor
blockade markedly potentiates duloxetine- and fluoxetine-induced increases
in noradrenaline, dopamine, and serotonin levels in the frontal cortex of freely moving
rats. J. Neurochem. 69, 2616–2619 (1997).
19. P. Pais-Roldán, C. Mateo, W.-J. Pan, B. Acland, D. Kleinfeld, L. H. Snyder, X. Yu, S. Keilholz,
Contribution of animal models toward understanding resting state functional
connectivity. Neuroimage 245, 118630 (2021).
20. J. M. Stafford, B. R. Jarrett, O. Miranda-Dominguez, B. D. Mills, N. Cain, S. Mihalas,
G. P. Lahvis, K. M. Lattal, S. H. Mitchell, S. V. David, J. D. Fryer, J. T. Nigg, D. A. Fair,
Large-scale topology and the default mode network in the mouse connectome.
Proc . Natl. Acad. Sci. U.S.A. 111, 18745–18750 (2014).
21. J. Grandjean, V. Zerbi, J. H. Balsters, N. Wenderoth, M. Rudin, Structural basis of large-
scale functional connectivity in the mouse. J. Neurosci. 37, 8092–8101 (2017).
22. J. Grandjean, C. Canella, C. Anckaerts, G. Ayrancı, S. Bougacha, T. Bienert, D. Buehlmann,
L. Coletta, D. Gallino, N. Gass, C. M. Garin, N. A. Nadkarni, N. S. Hübner, M. Karatas,
Y. Komaki, S. Kreitz, F. Mandino, A. E. Mechling, C. Sato, K. Sauer, D. Shah, S. Strobelt,
N. Takata, I. Wank, T. Wu, N. Yahata, L. Y. Yeow, Y. Yee, I. Aoki, M. M. Chakravarty,
W. T. Chang, M. Dhenain, D. von Elverfeldt, L. A. Harsan, A. Hess, T. Jiang, G. A. Keliris,
J. P. Lerch, A. Meyer-Lindenberg, H. Okano, M. Rudin, A. Sartorius, A. van der Linden,
M. Verhoye, W. Weber-Fahr, N. Wenderoth, V. Zerbi, A. Gozzi, Common functional
networks in the mouse brain revealed by multi-centre resting-state fMRI analysis.
Neuroimage 205, 116278 (2020).
23. J. Ferrier, E. Tiran, T. Deffieux, M. Tanter, Z. Lenkei, Functional imaging evidence
for task-induced deactivation and disconnection of a major default mode network hub
in the mouse brain. Proc. Natl. Acad. Sci. U.S.A. 117, 15270–15280 (2020).
24. L. Coletta, M. Pagani, J. D. Whitesell, J. A. Harris, B. Bernhardt, A. Gozzi, Network structure
of the mouse brain connectome with voxel resolution. Sci. Adv. 6, eabb7187 (2020).
25. J. D. Whitesell, A. Liska, L. Coletta, K. E. Hirokawa, P. Bohn, A. Williford, P. A. Groblewski,
N. Graddis, L. Kuan, J. E. Knox, A. Ho, W. Wakeman, P. R. Nicovich, T. N. Nguyen,
C. T. J. van Velthoven, E. Garren, O. Fong, M. Naeemi, A. M. Henry, N. Dee, K. A. Smith,
B. Levi, D. Feng, L. Ng, B. Tasic, H. Zeng, S. Mihalas, A. Gozzi, J. A. Harris, Regional, layer,
and cell-type-specific connectivity of the mouse default mode network. Neuron 109,
545–559.e8 (2021).
26. F. Mandino, R. M. Vrooman, H. E. Foo, L. Y. Yeow, T. A. W. Bolton, P. Salvan, C. L. Teoh,
C. Y. Lee, A. Beauchamp, S. Luo, R. Bi, J. Zhang, G. H. T. Lim, N. Low, J. Sallet, J. Gigg,
J. P. Lerch, R. B. Mars, M. Olivo, Y. Fu, J. Grandjean, A triple-network organization
for the mouse brain. Mol. Psychiatry , (2021).
27. K.-H. Chuang, F. A. Nasrallah, Functional networks and network perturbations in rodents.
Neuroimage 163, 419–436 (2017).
28. F. Sforazzini, A. J. Schwarz, A. Galbusera, A. Bifone, A. Gozzi, Distributed BOLD and CBV-
weighted resting-state networks in the mouse brain. Neuroimage 87, 403–415 (2014).
29. L.-M. Hsu, X. Liang, H. Gu, J. K. Brynildsen, J. A. Stark, J. A. Ash, C. P. Lin, H. Lu, P. R. Rapp,
E. A. Stein, Y. Yang, Constituents and functional implications of the rat default mode
network. Proc. Natl. Acad. Sci. U.S.A. 113, E4541–E4547 (2016).
30. Y.-W. Chen, M. Das, E. A. Oyarzabal, Q. Cheng, N. W. Plummer, K. G. Smith, G. K. Jones,
D. Malawsky, J. L. Yakel, Y.-Y. I. Shih, P. Jensen, Genetic identification of a population of
noradrenergic neurons implicated in attenuation of stress-related responses. Mol. Psychiatry
24, 710–725 (2019).
31. N. R. Sciolino, N. W. Plummer, Y.-W. Chen, G. M. Alexander, S. D. Robertson, S. M. Dudek,
Z. A. McElligott, P. Jensen, Recombinase-dependent mouse lines for chemogenetic
activation of genetically defined cell types. Cell Rep. 15, 2563–2573 (2016).
32. S. D. Robertson, N. W. Plummer, J. de Marchena, P. Jensen, Developmental origins
of central norepinephrine neuron diversity. Nat. Neurosci. 16, 1016–1023 (2013).
33. S.-H. Lee, M. A. Broadwater, W. Ban, T.-W. W. Wang, H.-J. Kim, J. S. Dumas, R. P. Vetreno,
M. A. Herman, A. L. Morrow, J. Besheer, T. L. Kash, C. A. Boettiger, D. L. Robinson,
F. T. Crews, Y.-Y. I. Shih, An isotropic EPI database and analytical pipelines for rat brain
resting-state fMRI. Neuroimage 243, 118541 (2021).
34. J. Paasonen, P. Stenroos, R. A. Salo, V. Kiviniemi, O. Gröhn, Functional connectivity under
six anesthesia protocols and the awake condition in rat brain. Neuroimage 172, 9–20 (2018).
35. C. Nacif-Coelho, C. Correa-Sales, L. L. Chang, M. Maze, Perturbation of ion channel
conductance alters the hypnotic response to the 2-adrenergic agonist
dexmedetomidine in the locus coeruleus of the rat. Anesthesiology 81, 1527–1534 (1994).
36. J. L. Gomez, J. Bonaventura, W. Lesniak, W. B. Mathews, P. Sysa-Shah, L. A. Rodriguez,
R. J. Ellis, C. T. Richie, B. K. Harvey, R. F. Dannals, M. G. Pomper, A. Bonci, M. Michaelides,
Chemogenetics revealed: DREADD occupancy and activation via converted clozapine.
Science 357, 503–507 (2017).
37. H. Lu, Q. Zou, H. Gu, M. E. Raichle, E. A. Stein, Y. Yang, Rat brains also have a default mode
network. Proc. Natl. Acad. Sci. U.S.A. 109, 3979–3984 (2012).
38. W. Tu, Z. Ma, Y. Ma, D. Dopfel, N. Zhang, Suppressing anterior cingulate cortex modulates
default mode network and behavior in awake rats. Cereb. Cortex 31, 312–323 (2021).
39. L. M. Peeters, R. Hinz, J. R. Detrez, S. Missault, W. H. de Vos, M. Verhoye, A. van der Linden,
G. A. Keliris, Chemogenetic silencing of neurons in the mouse anterior cingulate area
modulates neuronal activity and functional connectivity. Neuroimage 220, 117088 (2020).
40. T. J. M. Roelofs, J. P. H. Verharen, G. A. F. van Tilborg, L. Boekhoudt, A. van der Toorn,
J. W. de Jong, M. C. M. Luijendijk, W. M. Otte, R. A. H. Adan, R. M. Dijkhuizen, A novel
approach to map induced activation of neuronal networks using chemogenetics
and functional neuroimaging in rats: A proof-of-concept study on the mesocorticolimbic
system. Neuroimage 156, 109–118 (2017).
Oyarzabal et al., Sci. Adv. 8, eabm9898 (2022) 29 April 2022
SCIENCE ADVANCES | RESEARCH ARTICLE
11 of 12
41. A. Giorgi, S. Migliarini, A. Galbusera, G. Maddaloni, M. Mereu, G. Margiani, M. Gritti,
S. Landi, F. Trovato, S. M. Bertozzi, A. Armirotti, G. M. Ratto, M. A. de Luca, R. Tonini,
A. Gozzi, M. Pasqualetti, Brain-wide mapping of endogenous serotonergic transmission
via chemogenetic fMRI. Cell Rep. 21, 910–918 (2017).
42. L. M. Peeters, M. van den Berg, R. Hinz, G. Majumdar, I. Pintelon, G. A. Keliris, Cholinergic
modulation of the default mode like network in rats. iScience 23, 101455 (2020).
43. S. P. Strebel, C. Kindler, B. Bissonnette, G. Tschalèr, D. Deanovic, The impact of systemic
vasoconstrictors on the cerebral circulation of anesthetized patients. Anesthesiology 89,
67–72 (1998).
44. J. Feng, C. Zhang, J. E. Lischinsky, M. Jing, J. Zhou, H. Wang, Y. Zhang, A. Dong, Z. Wu,
H. Wu, W. Chen, P. Zhang, J. Zou, S. A. Hires, J. J. Zhu, G. Cui, D. Lin, J. Du, Y. Li, A
genetically encoded fluorescent sensor for rapid and specific in vivo detection
of norepinephrine. Neuron 102, 745–761.e8 (2019).
45. H. Dana, B. Mohar, Y. Sun, S. Narayan, A. Gordus, J. P. Hasseman, G. Tsegaye, G. T. Holt,
A. Hu, D. Walpita, R. Patel, J. J. Macklin, C. I. Bargmann, M. B. Ahrens, E. R. Schreiter,
V. Jayaraman, L. L. Looger, K. Svoboda, D. S. Kim, Sensitive red protein calcium indicators
for imaging neural activity. eLife 5, e12727 (2016).
46. J. McBurney-Lin, J. Lu, Y. Zuo, H. Yang, Locus coeruleus-norepinephrine modulation
of sensory processing and perception: A focused review. Neurosci. Biobehav. Rev. 105,
190–199 (2019).
47. M. Michaelides, Y. L. Hurd, DREAMM: A biobehavioral imaging methodology for dynamic
in vivo whole-brain mapping of cell type-specific functional networks.
Neuropsychopharmacology 40, 239–240 (2015).
48. J. D. Bremner, R. B. Innis, C. K. Ng, L. H. Staib, R. M. Salomon, R. A. Bronen, J. Duncan,
S. M. Southwick, J. H. Krystal, D. Rich, G. Zubal, H. Dey, R. Soufer, D. S. Charney, Positron
emission tomography measurement of cerebral metabolic correlates of yohimbine
administration in combat-related posttraumatic stress disorder. Arch. Gen. Psychiatry 54,
246–254 (1997).
49. A. Justice, S. M. Feldman, L. L. Brown, The nucleus locus coeruleus modulates local
cerebral glucose utilization during noise stress in rats. Brain Res. 490, 73–84 (1989).
50. N. French, M. D. Lalies, D. J. Nutt, J. A. Pratt, Idazoxan-induced reductions in cortical
glucose use are accompanied by an increase in noradrenaline release: Complementary
[14C]2-deoxyglucose and microdialysis studies. Neuropharmacology 34, 605–613 (1995).
51. O. Sorg, P. J. Magistretti, Characterization of the glycogenolysis elicited by vasoactive
intestinal peptide, noradrenaline and adenosine in primary cultures of mouse cerebral
cortical astrocytes. Brain Res. 563, 227–233 (1991).
52. T. T. Quach, C. Rose, J. C. Schwartz, [3H]Glycogen hydrolysis in brain slices: Responses
to neurotransmitters and modulation of noradrenaline receptors. J. Neurochem. 30,
1335–1341 (1978).
53. N. Cannella, A. Cosa-Linan, M. Roscher, T. T. Takahashi, N. Vogler, B. Wängler, R. Spanagel,
[18F]-fluorodeoxyglucose-positron emission tomography in rats with prolonged cocaine
self-administration suggests potential brain biomarkers for addictive behavior.
Front. Psych. 8, 218 (2017).
54. V. Menon, L. Q. Uddin, Saliency, switching, attention and control: A network model
of insula function. Brain Struct. Funct. 214, 655–667 (2010).
55. D. Gutierrez-Barragan, M. A. Basson, S. Panzeri, A. Gozzi, Infraslow state fluctuations
govern spontaneous fmri network dynamics. Curr. Biol. 29, 2295–2306.e5 (2019).
56. V. Zerbi, J. Grandjean, M. Rudin, N. Wenderoth, Mapping the mouse brain with rs-fMRI:
An optimized pipeline for functional network identification. Neuroimage 123, 11–21 (2015).
57. C. Canella, F. Rocchi, S. Noei, D. Gutierrez-Barragan, L. Coletta, A. Galbusera, S. Vassanelli,
M. Pasqualetti, G. Iurilli, S. Panzeri, A. Gozzi, Cortical silencing results in paradoxical
fMRI overconnectivity. bioRxiv 2020.08.05.237958 [Preprint]. 15 October 2020.
https:/ /doi.org/10.1101/2020.08.05.237958.
58. C. S. Wood, R. J. Valentino, S. K. Wood, Individual differences in the locus coeruleus-
norepinephrine system: Relevance to stress-induced cardiovascular vulnerability. Physiol.
Behav. 172, 40–48 (2017).
59. N. K. Logothetis, What we can do and what we cannot do with fMRI. Nature 453, 869–878
(2008).
60. L. K. Bekar, H. S. Wei, M. Nedergaard, The locus coeruleus-norepinephrine network
optimizes coupling of cerebral blood volume with oxygen demand. J. Cereb. Blood Flow
Metab. 32, 2135–2145 (2012).
61. P. Pais-Roldán, K. Takahashi, F. Sobczak, Y. Chen, X. Zhao, H. Zeng, Y. Jiang, X. Yu,
Indexing brain state-dependent pupil dynamics with simultaneous fMRI and optical fiber
calcium recording. Proc. Natl. Acad. Sci. U.S.A. 117, 6875–6882 (2020).
62. R. L. van den Brink, T. Pfeffer, C. M. Warren, P. R. Murphy, K.-D. Tona, N. J. A. van der Wee,
E. Giltay, M. S. van Noorden, S. A. R. B. Rombouts, T. H. Donner, S. Nieuwenhuis,
Catecholaminergic neuromodulation shapes intrinsic MRI functional connectivity
in the human brain. J. Neurosci. 36, 7865–7876 (2016).
63. J. M. Shine, R. L. van den Brink, D. Hernaus, S. Nieuwenhuis, R. A. Poldrack,
Catecholaminergic manipulation alters dynamic network topology across cognitive
states. Netw. Neurosci. 2, 381–396 (2018).
64. A. Gozzi, S. Lepore, E. Vicentini, E. Merlo-Pich, A. Bifone, Differential effect of orexin-1
and CRF-1 antagonism on stress circuits: A fMRI study in the rat with the pharmacological
stressor Yohimbine. Neuropsychopharmacology 38, 2120–2130 (2013).
65. X. Toussay, K. Basu, B. Lacoste, E. Hamel, Locus coeruleus stimulation recruits a broad
cortical neuronal network and increases cortical perfusion. J. Neurosci. 33, 3390–3401
(2013).
66. M. E. Raichle, B. K. Hartman, J. O. Eichling, L. G. Sharpe, Central noradrenergic regulation
of cerebral blood flow and vascular permeability. Proc. Natl. Acad. Sci. U.S.A. 72,
3726–3730 (1975).
67. E. Eldar, J. D. Cohen, Y. Niv, The effects of neural gain on attention and learning.
Nat. Neurosci. 16, 1146–1153 (2013).
68. J. M. Shine, Neuromodulatory influences on integration and segregation in the brain.
Trends Cogn. Sci. 23, 572–583 (2019).
69. G. Wainstein, D. Rojas-Líbano, V. Medel, D. Alnæs, K. K. Kolskår, T. Endestad, B. Laeng,
T. Ossandon, N. Crossley, E. Matar, J. M. Shine, The ascending arousal system promotes
optimal performance through mesoscale network integration in a visuospatial
attentional task. Netw. Neurosci. 5, 890–910 (2021).
70. A. T. Baria, M. N. Baliki, T. Parrish, A. V. Apkarian, Anatomical and functional assemblies
of brain BOLD oscillations. J. Neurosci. 31, 7910–7919 (2011).
71. C. Grefkes, L. E. Wang, S. B. Eickhoff, G. R. Fink, Noradrenergic modulation of cortical
networks engaged in visuomotor processing. Cereb. Cortex 20, 783–797 (2010).
72. F. Saviola, E. Pappaianni, A. Monti, A. Grecucci, J. Jovicich, N. de Pisapia, Trait and state
anxiety are mapped differently in the human brain. Sci. Rep. 10, 11112 (2020).
73. M. E. Carter, O. Yizhar, S. Chikahisa, H. Nguyen, A. Adamantidis, S. Nishino, K. Deisseroth,
L. de Lecea, Tuning arousal with optogenetic modulation of locus coeruleus neurons.
Nat. Neurosci. 13, 1526–1533 (2010).
74. E. M. Vazey, G. Aston-Jones, Designer receptor manipulations reveal a role of the locus
coeruleus noradrenergic system in isoflurane general anesthesia. Proc. Natl. Acad. Sci.
U.S.A. 111, 3859–3864 (2014).
75. M. Yang, N. K. Logothetis, O. Eschenko, Phasic activation of the locus coeruleus
attenuates the acoustic startle response by increasing cortical arousal. Sci. Rep. 11, 1409
(2021).
76. J. A. Ross, E. J. Van Bockstaele, The locus coeruleus-norepinephrine system in stress
and arousal: Unraveling historical, current, and future perspectives. Front. Psych. 11,
601519 (2020).
77. G. Aston-Jones, J. D. Cohen, An integrative theory of locus coeruleus-norepinephrine
function: Adaptive gain and optimal performance. Annu. Rev. Neurosci. 28, 403–450
(2005).
78. A. M. C. Kelly, L. Q. Uddin, B. B. Biswal, F. X. Castellanos, M. P. Milham, Competition
between functional brain networks mediates behavioral variability. Neuroimage 39,
527–537 (2008).
79. M. Wang, B. P. Ramos, C. D. Paspalas, Y. Shu, A. Simen, A. Duque, S. Vijayraghavan,
A. Brennan, A. Dudley, E. Nou, J. A. Mazer, D. A. McCormick, A. F. T. Arnsten, 2A-
adrenoceptors strengthen working memory networks by inhibiting cAMP-HCN channel
signaling in prefrontal cortex. Cell 129, 397–410 (2007).
80. D. J. Chandler, Evidence for a specialized role of the locus coeruleus noradrenergic
system in cortical circuitries and behavioral operations. Brain Res. 1641, 197–206
(2016).
81. C. Zarow, S. A. Lyness, J. A. Mortimer, H. C. Chui, Neuronal loss is greater in the locus
coeruleus than nucleus basalis and substantia nigra in Alzheimer and Parkinson diseases.
Arch. Neurol. 60, 337–341 (2003).
82. A. Mohan, A. J. Roberto, A. Mohan, A. Lorenzo, K. Jones, M. J. Carney, L. Liogier-Weyback,
S. Hwang, K. A. Lapidus, The significance of the default mode network (DMN)
in neurological and neuropsychiatric disorders: A review. Yale J. Biol. Med. 89, 49–57
(2016).
83. A. C. Peterson, C.-S. R. Li, Noradrenergic dysfunction in Alzheimer’s and Parkinson’s
diseases—An overview of imaging studies. Front. Aging Neurosci. 10, 127 (2018).
84. A. Bari, S. Xu, M. Pignatelli, D. Takeuchi, J. Feng, Y. Li, S. Tonegawa, Differential attentional
control mechanisms by two distinct noradrenergic coeruleo-frontal cortical pathways.
Proc. Natl. Acad. Sci. U.S.A. 117, 29080–29089 (2020).
85. R. A. Kimmel, D. H. Turnbull, V. Blanquet, W. Wurst, C. A. Loomis, A. L. Joyner, Two lineage
boundaries coordinate vertebrate apical ectodermal ridge formation. Genes Dev. 14,
1377–1389 (2000).
86. B. L. Roth, DREADDs for neuroscientists. Neuron 89, 683–694 (2016).
87. M. Das, E. A. Oyarzabal, L. Chen, S.-H. Lee, N. Shah, G. Gerlach, W. Zhang, T.-H. H. Chao,
N. V. D. Berge, C. Liu, C. Donley, S. A. Montgomery, Y.-Y. I. Shih, One-pot synthesis
of carboxymethyl-dextran coated iron oxide nanoparticles (CION) for preclinical fMRI
and MRA applications. Neuroimage 238, 118213 (2021).
88. J. D. Power, A. Mitra, T. O. Laumann, A. Z. Snyder, B. L. Schlaggar, S. E. Petersen, Methods
to detect, characterize, and remove motion artifact in resting state fMRI. Neuroimage 84,
320–341 (2014).
Oyarzabal et al., Sci. Adv. 8, eabm9898 (2022) 29 April 2022
SCIENCE ADVANCES | RESEARCH ARTICLE
12 of 12
89. L.-M. Hsu, S. Wang, P. Ranadive, W. Ban, T.-H. H. Chao, S. Song, D. H. Cerri, L. R. Walton,
M. A. Broadwater, S.-H. Lee, D. Shen, Y.-Y. I. Shih, Automatic skull stripping of rat
and mouse brain MRI data using U-Net. Front. Neurosci. 14, 568614 (2020).
90. Q. Wang, S.-L. Ding, Y. Li, J. Royall, D. Feng, P. Lesnar, N. Graddis, M. Naeemi, B. Facer,
A. Ho, T. Dolbeare, B. Blanchard, N. Dee, W. Wakeman, K. E. Hirokawa, A. Szafer,
S. M. Sunkin, S. W. Oh, A. Bernard, J. W. Phillips, M. Hawrylycz, C. Koch, H. Zeng,
J. A. Harris, L. Ng, The allen mouse brain common coordinate framework: A 3D reference
atlas. Cell 181, 936–953.e20 (2020).
91. T. T. Liu, A. Nalci, M. Falahpour, The global signal in fMRI: Nuisance or information?
Neuroimage 150, 213–229 (2017).
92. L. Griffanti, G. Douaud, J. Bijsterbosch, S. Evangelisti, F. Alfaro-Almagro, M. F. Glasser,
E. P. Duff, S. Fitzgibbon, R. Westphal, D. Carone, C. F. Beckmann, S. M. Smith, Hand
classification of fMRI ICA noise components. Neuroimage 154, 188–205 (2017).
93. C. Rummel, R. K. Verma, V. Schöpf, E. Abela, M. Hauf, J. F. Z. Berruecos, R. Wiest, Time
course based artifact identification for independent components of resting-state FMRI.
Front. Hum. Neurosci. 7, 214 (2013).
94. K. Schmidt, B. Bari, M. Ralle, C. Washington-Hughes, A. Muchenditsi, E. Maxey,
S. Lutsenko, Localization of the locus coeruleus in the mouse brain. J. Vis. Exp. , (2019).
95. Y.-F. Zang, Y. He, C.-Z. Zhu, Q.-J. Cao, M.-Q. Sui, M. Liang, L.-X. Tian, T.-Z. Jiang, Y.-F. Wang,
Altered baseline brain activity in children with ADHD revealed by resting-state functional
MRI. Brain Dev. 29, 83–91 (2007).
96. D. Cordes, V. M. Haughton, K. Arfanakis, J. D. Carew, P. A. Turski, C. H. Moritz,
M. A. Quigley, M. E. Meyerand, Frequencies contributing to functional connectivity
in the cerebral cortex in “resting-state” data. AJNR Am. J. Neuroradiol. 22, 1326–1333
(2001).
97. E. A. Allen, E. B. Erhardt, E. Damaraju, W. Gruner, J. M. Segall, R. F. Silva, M. Havlicek,
S. Rachakonda, J. Fries, R. Kalyanam, A. M. Michael, A. Caprihan, J. A. Turner, T. Eichele,
S. Adelsheim, A. D. Bryan, J. Bustillo, V. P. Clark, S. W. Feldstein Ewing, F. Filbey, C. C. Ford,
K. Hutchison, R. E. Jung, K. A. Kiehl, P. Kodituwakku, Y. M. Komesu, A. R. Mayer,
G. D. Pearlson, J. P. Phillips, J. R. Sadek, M. Stevens, U. Teuscher, R. J. Thoma, V. D. Calhoun,
A baseline for the multivariate comparison of resting-state networks. Front. Syst. Neurosci.
5, 2 (2011).
98. A. Schaefer, D. S. Margulies, G. Lohmann, K. J. Gorgolewski, J. Smallwood, S. J. Kiebel,
A. Villringer, Dynamic network participation of functional connectivity hubs assessed by
resting-state fMRI. Front. Hum. Neurosci. 8, 195 (2014).
99. A. Abou-Elseoud, T. Starck, J. Remes, J. Nikkinen, O. Tervonen, V. Kiviniemi, The effect
of model order selection in group PICA. Hum. Brain Mapp. 31, 1207–1216 (2010).
100. E. A. Allen, E. B. Erhardt, Y. Wei, T. Eichele, V. D. Calhoun, Capturing inter-subject
variability with group independent component analysis of fMRI data: A simulation study.
Neuroimage 59, 4141–4159 (2012).
101. A. Abou Elseoud, H. Littow, J. Remes, T. Starck, J. Nikkinen, J. Nissilä, M. Timonen,
O. Tervonen, V. Kiviniemi, Group-ICA model order highlights patterns of functional brain
connectivity. Front. Syst. Neurosci. 5, 37 (2011).
102. S. M. Smith, P. T. Fox, K. L. Miller, D. C. Glahn, P. M. Fox, C. E. Mackay, N. Filippini,
K. E. Watkins, R. Toro, A. R. Laird, C. F. Beckmann, Correspondence of the brain’s functional
architecture during activation and rest. Proc. Natl. Acad. Sci. U.S.A. 106, 13040–13045 (2009).
103. V. Kiviniemi, T. Starck, J. Remes, X. Long, J. Nikkinen, M. Haapea, J. Veijola, I. Moilanen,
M. Isohanni, Y.-F. Zang, O. Tervonen, Functional segmentation of the brain cortex using
high model order group PICA. Hum. Brain Mapp. 30, 3865–3886 (2009).
104. X. V. To, V. Vegh, F. A. Nasrallah, Towards data-driven group inferences of resting-state
fMRI data in rodents: Comparison of group ICA, GIG-ICA, and IVA-GL. J. Neurosci. Methods
366, 109411 (2022).
105. B. Jarrahi, Examining the influence of spatial smoothing on spatiotemporal features
of intrinsic connectivity networks at low ICA model order. Annu. Int. Conf. IEEE Eng. Med.
Biol. Soc. 2021, 3221–3224 (2021).
106. X.-N. Zuo, C. Kelly, J. S. Adelstein, D. F. Klein, F. X. Castellanos, M. P. Milham, Reliable
intrinsic connectivity networks: Test-retest evaluation using ICA and dual regression
approach. Neuroimage 49, 2163–2177 (2010).
107. V. D. Blondel, J.-L. Guillaume, R. Lambiotte, E. Lefebvre, Fast unfolding of communities
in large networks. J. Stat. Mech. 2008, P10008 (2008).
108. M. Meilă, Comparing clusterings—An information based distance. J. Multivar. Anal. 98,
873–895 (2007).
109. A. Zalesky, A. Fornito, E. T. Bullmore, Network-based statistic: Identifying differences
in brain networks. Neuroimage 53, 1197–1207 (2010).
110. H.-J. Park, K. J. Friston, C. Pae, B. Park, A. Razi, Dynamic effective connectivity in resting
state fMRI. Neuroimage 180, 594–608 (2018).
111. W. D. Penny, K. E. Stephan, J. Daunizeau, M. J. Rosa, K. J. Friston, T. M. Schofield, A. P. Leff,
Comparing families of dynamic causal models. PLOS Comput. Biol. 6, e1000709 (2010).
112. A. Boucard, A. Marchand, X. Noguès, Reliability and validity of structural equation
modeling applied to neuroimaging data: A simulation study. J. Neurosci. Methods 166,
278–292 (2007).
113. R. M. Baron, D. A. Kenny, The moderator–mediator variable distinction in social
psychological research: Conceptual, strategic, and statistical considerations. J. Pers. Soc.
Psychol. 51, 1173–1182 (1986).
114. K. J. Preacher, A. F. Hayes, Asymptotic and resampling strategies for assessing
and comparing indirect effects in multiple mediator models. Behav. Res. Methods 40,
879–891 (2008).
115. D. Gutierrez-Barragan, N. A. Singh, F. G. Alvino, L. Coletta, F. Rocchi, E. De Guzman,
A. Galbusera, M. Uboldi, S. Panzeri, A. Gozzi, Unique spatiotemporal fMRI dynamics
in the awake mouse brain. Curr. Biol. 32, 631–644.e6 (2022).
116. Y. Liu, P. D. Perez, Z. Ma, Z. Ma, D. Dopfel, S. Cramer, W. Tu, N. Zhang, An open database
of resting-state fMRI in awake rats. Neuroimage 220, 117094 (2020).
117. B. Nieuwenhuis, B. Haenzi, S. Hilton, A. Carnicer-Lombarte, B. Hobo, J. Verhaagen,
J. W. Fawcett, Optimization of adeno-associated viral vector-mediated transduction
of the corticospinal tract: Comparison of four promoters. Gene Ther. 28, 56–74 (2021).
Acknowledgments: We thank the members of the UNC Center for Animal MRI and F. Crews,
Z. McElligott, and B. Roth for inputs. We thank the UNC Small Animal Imaging Core Facility
staff J. Frank and J. Merrill for assistance in PET data acquisition. Funding: This research was
supported by the Extramural Research Programs of U.S. National Institutes of Health, NINDS
(R01NS091236), NIMH (R01MH126518, R01MH111429, and RF1MH117053), NIAAA
(P60AA011605 and U01AA020023), and NICHD (P50HD103573) to Y.-Y.I.S. and the Intramural
Research Program of the U.S. National Institutes of Health, National Institute of Environmental
Health Sciences (ZIA-ES102805 to P.J. and 1ZIAES103310 to G.C.). Author contributions:
E.A.O., M.D., and Y.-Y.I.S. designed the study. E.A.O. and M.D. collected the imaging data. E.A.O.,
L.-M.H., and S.-H.L. analyzed the imaging data. J.Z. and G.C. performed the viral injections and
fiber implantation. E.A.O., T.-H.H.C., and W.Z. collected and analyzed the fiber photometry
data. K.G.S. processed and K.G.S. and P.J. analyzed the histology data. N.R.S., I.Y.E., and P.J.
developed and shared the transgenic mice. E.A.O., L.-M.H., P.J., and Y.-Y.I.S. wrote the
manuscript with input from all authors. Competing interests: The authors declare that they
have no competing interests. Data and materials availability: All data needed to evaluate
the conclusions in the paper are present in the paper and/or the Supplementary Materials. All
MRI, PET, and photometry data from this study are openly available on Mendeley Data
(https://doi.org/10.17632/hxch8htz84.2).
Submitted 22 November 2021
Accepted 15 March 2022
Published 29 April 2022
10.1126/sciadv.abm9898
... Investigators in one study observed that chemogenetic stimulation of tonic LC activity strengthened connectivity in this network. 82 Greater default mode network activity may be a consequence of inadequate modulation of salience and executive control networks by the LC, biasing individuals toward nongoal-directed activity. 34,83 Associations of racial discrimination with altered connectivity of the LC and a default mode network node highlight the potential importance of the LC in pathologic neurophysiologic adaptations to racial discrimination. ...
Article
Full-text available
Importance Racial discrimination increases the risk of adverse brain health outcomes, potentially via neuroplastic changes in emotion processing networks. The involvement of deep brain regions (brainstem and midbrain) in these responses is unknown. Potential associations of racial discrimination with alterations in deep brain functional connectivity and accelerated epigenetic aging, a process that substantially increases vulnerability to health problems, are also unknown. Objective To examine associations of racial discrimination with brainstem and midbrain resting-state functional connectivity (RSFC) and DNA methylation age acceleration (DMAA) among Black women in the US. Design, Setting, and Participants This cohort study was conducted between January 1, 2012, and February 28, 2015, and included a community-based sample of Black women (aged ≥18 years) recruited as part of the Grady Trauma Project. Self-reported racial discrimination was examined in association with seed-to-voxel brain connectivity, including the locus coeruleus (LC), periaqueductal gray (PAG), and superior colliculus (SC); an index of DMAA (Horvath clock) was also evaluated. Posttraumatic stress disorder (PTSD), trauma exposure, and age were used as covariates in statistical models to isolate racial discrimination–related variance. Data analysis was conducted between January 10 and October 30, 2023. Exposure Varying levels of racial discrimination exposure, other trauma exposure, and posttraumatic stress disorder (PTSD). Main Outcomes and Measures Racial discrimination frequency was assessed with the Experiences of Discrimination Scale, other trauma exposure was evaluated with the Traumatic Events Inventory, and current PTSD was evaluated with the PTSD Symptom Scale. Seed-to-voxel functional connectivity analyses were conducted with LC, PAG, and SC seeds. To assess DMAA, the Methylation EPIC BeadChip assay (Illumina) was conducted with whole-blood samples from a subset of 49 participants. Results This study included 90 Black women, with a mean (SD) age of 38.5 (11.3) years. Greater racial discrimination was associated with greater left LC RSFC to the bilateral precuneus (a region within the default mode network implicated in rumination and reliving of past events; cluster size k = 228; t 85 = 4.78; P < .001, false discovery rate-corrected). Significant indirect effects were observed for the left LC-precuneus RSFC on the association between racial discrimination and DMAA (β [SE] = 0.45 [0.16]; 95% CI, 0.12-0.77). Conclusions and Relevance In this study, more frequent racial discrimination was associated with proportionately greater RSFC of the LC to the precuneus, and these connectivity alterations were associated with DMAA. These findings suggest that racial discrimination contributes to accelerated biological aging via altered connectivity between the LC and default mode network, increasing vulnerability for brain health problems.
... Although the LC is a heterogeneous structure with modular organization, it appears that in stressful situations, broad activation of the LC -and subsequent widespread NA release throughout the brain -serves as a broadcast signal to orchestrate re-routing of computational resources to meet situational demands (Likhtik and Johansen, 2019;Poe et al., 2020). On the network level, for example, NA release from the LC is sufficient to trigger a rapid reconfiguration of large-scale networks that shift processing capacity toward salience processing (Zerbi et al., 2019;Oyarzabal et al., 2022). On a circuit level, forebrain regions seem to be particularly important targets of the LC-NA system to influence cognitive processes and ultimately behavior. ...
Article
Full-text available
Exposure to an acute stressor triggers a complex cascade of neurochemical events in the brain. However, deciphering their individual impact on stress-induced molecular changes remains a major challenge. Here, we combine RNA sequencing with selective pharmacological, chemogenetic, and optogenetic manipulations to isolate the contribution of the locus coeruleus-noradrenaline (LC-NA) system to the acute stress response in mice. We reveal that NA release during stress exposure regulates a large and reproducible set of genes in the dorsal and ventral hippocampus via β-adrenergic receptors. For a smaller subset of these genes, we show that NA release triggered by LC stimulation is sufficient to mimic the stress-induced transcriptional response. We observe these effects in both sexes, and independent of the pattern and frequency of LC activation. Using a retrograde optogenetic approach, we demonstrate that hippocampus-projecting LC neurons directly regulate hippocampal gene expression. Overall, a highly selective set of astrocyte-enriched genes emerges as key targets of LC-NA activation, most prominently several subunits of protein phosphatase 1 ( Ppp1r3c , Ppp1r3d , Ppp1r3g ) and type II iodothyronine deiodinase ( Dio2 ). These results highlight the importance of astrocytic energy metabolism and thyroid hormone signaling in LC-mediated hippocampal function and offer new molecular targets for understanding how NA impacts brain function in health and disease.
... No reuse allowed without permission. Pulse repetition rate modulated activity in GP and CPu, but not in SNr, were related to the therapeutic effect of DBS We further determined how STN optogenetic DBS manipulates pathological circling behavior through activity changes in the three identified brain clusters (CPu, GP, and SNr, see Figure 3B) using a mediation model with linear mixed-effect regression 32,33 . First, the data demonstrated that the increased pulse repetition rate of STN optogenetic DBS causally manipulated the reduction of normalized angular velocity shown earlier in Figure 2E (coefficient = -0.735, ...
Preprint
While deep brain stimulation (DBS) is widely employed for managing motor symptoms in Parkinson’s disease (PD), its exact circuit mechanisms remain controversial. To identify the neural targets affected by therapeutic DBS in PD, we analyzed DBS-evoked whole brain activity in female hemi-parkinsonian rats using function magnetic resonance imaging (fMRI). We delivered subthalamic nucleus (STN) DBS at various stimulation pulse repetition rates using optogenetics, allowing unbiased examinations of cell-type specific STN feed-forward neural activity. Unilateral STN optogenetic stimulation elicited pulse repetition rate-dependent alterations of blood-oxygenation-level-dependent (BOLD) signals in SNr (substantia nigra pars reticulata), GP (globus pallidus), and CPu (caudate putamen). Notably, these manipulations effectively ameliorated pathological circling behavior in animals expressing the kinetically faster Chronos opsin, but not in animals expressing ChR2. Furthermore, mediation analysis revealed that the pulse repetition rate-dependent behavioral rescue was significantly mediated by optogenetically induced activity changes in GP and CPu, but not in SNr. This suggests that the activation of GP and CPu are critically involved in the therapeutic mechanisms of STN DBS.
Article
Arousal state is regulated by subcortical neuromodulatory nuclei, such as locus coeruleus, which send wide-reaching projections to cortex. Whether higher-order cortical regions have the capacity to recruit neuromodulatory systems to aid cognition is unclear. Here, we hypothesized that select cortical regions activate the arousal system, which, in turn, modulates large-scale brain activity, creating a functional circuit predicting cognitive ability. We utilized the Human Connectome Project 7T functional magnetic resonance imaging dataset (n = 149), acquired at rest with simultaneous eye tracking, along with extensive cognitive assessment for each subject. First, we discovered select frontoparietal cortical regions that drive large-scale spontaneous brain activity specifically via engaging the arousal system. Second, we show that the functionality of the arousal circuit driven by bilateral posterior cingulate cortex (associated with the default mode network) predicts subjects’ cognitive abilities. This suggests that a cortical region that is typically associated with self-referential processing supports cognition by regulating the arousal system.
Article
The default mode network (DMN) is a large-scale brain network known to be suppressed during a wide range of cognitive tasks. However, our comprehension of its role in naturalistic and unconstrained behaviors has remained elusive because most research on the DMN has been conducted within the restrictive confines of MRI scanners. Here, we use multisite GCaMP (a genetically encoded calcium indicator) fiber photometry with simultaneous videography to probe DMN function in awake, freely exploring rats. We examined neural dynamics in three core DMN nodes—the retrosplenial cortex, cingulate cortex, and prelimbic cortex—as well as the anterior insula node of the salience network, and their association with the rats’ spatial exploration behaviors. We found that DMN nodes displayed a hierarchical functional organization during spatial exploration, characterized by stronger coupling with each other than with the anterior insula. Crucially, these DMN nodes encoded the kinematics of spatial exploration, including linear and angular velocity. Additionally, we identified latent brain states that encoded distinct patterns of time-varying exploration behaviors and found that higher linear velocity was associated with enhanced DMN activity, heightened synchronization among DMN nodes, and increased anticorrelation between the DMN and anterior insula. Our findings highlight the involvement of the DMN in collectively and dynamically encoding spatial exploration in a real-world setting. Our findings challenge the notion that the DMN is primarily a “task-negative” network disengaged from the external world. By illuminating the DMN’s role in naturalistic behaviors, our study underscores the importance of investigating brain network function in ecologically valid contexts.
Article
Full-text available
The striatum, known as the input nucleus of the basal ganglia, is extensively studied for its diverse behavioral roles. However, the relationship between its neuronal and vascular activity, vital for interpreting functional magnetic resonance imaging (fMRI) signals, has not received comprehensive examination within the striatum. Here, we demonstrate that optogenetic stimulation of dorsal striatal neurons or their afferents from various cortical and subcortical regions induces negative striatal fMRI responses in rats, manifesting as vasoconstriction. These responses occur even with heightened striatal neuronal activity, confirmed by electrophysiology and fiber-photometry. In parallel, midbrain dopaminergic neuron optogenetic modulation, coupled with electrochemical measurements, establishes a link between striatal vasodilation and dopamine release. Intriguingly, in vivo intra-striatal pharmacological manipulations during optogenetic stimulation highlight a critical role of opioidergic signaling in generating striatal vasoconstriction. This observation is substantiated by detecting striatal vasoconstriction in brain slices after synthetic opioid application. In humans, manipulations aimed at increasing striatal neuronal activity likewise elicit negative striatal fMRI responses. Our results emphasize the necessity of considering vasoactive neurotransmission alongside neuronal activity when interpreting fMRI signal.
Article
Apathy is a common and disabling complication of Parkinson’s disease characterised by reduced goal-directed behaviour. Several studies have reported dysfunction within prefrontal cortical regions and projections from brainstem nuclei whose neuromodulators include dopamine, serotonin and noradrenaline. Work in animal and human neuroscience have confirmed contributions of these neuromodulators on aspects of motivated decision making. Specifically, these neuromodulators have overlapping contributions to encoding the value of decisions, and influence whether to explore alternative courses of action, or persist in an existing strategy to achieve a rewarding goal. Building upon this work, we hypothesised that apathy in Parkinson’s disease should be associated with an impairment in value-based learning. Using a four-armed restless bandit reinforcement learning task, we studied decision making in 75 volunteers; 53 patients with Parkinson’s Disease, with and without clinical apathy, and 22 age matched healthy controls. Patients with apathy exhibited impaired ability to choose the highest value bandit. Task performance predicted an individual patient’s apathy severity measured using the Lille Apathy Rating scale (R = -0.46, p<0.001). Computational modelling of the patient’s choices confirmed the apathy group made decisions that were indifferent to the learnt value of the options, consistent with previous reports of reward insensitivity. Further analysis demonstrated a shift away from exploiting the highest value option and a reduction in perseveration which also correlated with apathy scores (R = -0.5, p<0.001). We went on to acquire fMRI in 59 volunteers; a group of 19 patients with and 20 without apathy and 20 age matched controls performing the restless bandit task. Analysis of the fMRI signal at the point of reward feedback confirmed diminished signal within ventromedial prefrontal cortex in Parkinson’s disease, which was more marked in apathy, but not predictive of their individual apathy severity. Using a model-based categorisation of choice type, decisions to explore lower value bandits in the apathy group activated prefrontal cortex to a similar degree to the age-matched controls. In contrast, Parkinson’s patients without apathy demonstrated significantly increased activation across a distributed thalamo-cortical network. Enhanced activity in the thalamus predicted individual apathy severity across both patient groups and exhibited functional connectivity with dorsal anterior cingulate cortex and anterior insula. Given that task performance in patients without apathy was no different to the age-matched controls, we interpret the recruitment of this network as a possible compensatory mechanism, which compensates against symptomatic manifestation of apathy in Parkinson’s disease.
Article
While functional brain imaging studies in humans suggest that chronic cocaine use alters functional connectivity (FC) within and between key large-scale brain networks, including the default mode network (DMN), the salience network (SN), and the central executive network (CEN), cross-sectional studies in humans are challenging to obtain brain FC prior to cocaine use. Such information is critical to reveal the relationship between individual's brain FC and the subsequent development of cocaine dependence and brain changes during abstinence. Here, we performed a longitudinal study examining functional magnetic resonance imaging (fMRI) data in male rats ( n = 7), acquired before cocaine self-administration (baseline), on 1 d of abstinence following 10 d of cocaine self-administration, and again after 30 d of experimenter-imposed abstinence. Using repeated-measures analysis of variance (ANOVA) with network-based statistics (NBS), significant connectivity changes were found between anterior insular cortex (AI) of the SN, retrosplenial cortex (RSC) of the DMN, somatosensory cortex, and caudate–putamen (CPu), with AI–RSC FC showing the most robust changes between baseline and 1 d of abstinence. Additionally, the level of escalated cocaine intake is associated with AI–RSC and AI–CPu FC changes between 1 d and 30 d of abstinence; further, the subjects’ AI–RSC FC prior to cocaine intake is a significant moderator for the AI–RSC changes during abstinence. These results provide novel insights into the roles of AI–RSC FC before and after cocaine intake and suggest this circuit to be a potential target to modulate large-scale network and associated behavioral changes in cocaine use disorders.
Preprint
Full-text available
Arousal state is regulated by the brainstem nucleus locus coeruleus, which sends wide-reaching projections to cortex. How this powerful neuromodulatory system is recruited to aid cognition is largely underexplored. Here, we hypothesized that select cortical regions may activate the arousal system, which in turn modulates large-scale brain activity, creating a circuit whose integrity predicts cognitive ability. We utilized the Human Connectome Project 7T functional magnetic resonance imaging dataset (N=149), acquired at rest with simultaneous eye tracking, along with extensive cognitive assessment for each subject. First, we discovered select frontoparietal cortical regions that likely drive large-scale spontaneous brain activity specifically via engaging the arousal system. Second, we show that the integrity of the arousal circuit driven by bilateral posterior cingulate cortex (associated with the default mode network) predicts subjects' cognitive abilities. This suggests that a cortical region that is typically associated with self-referential processing supports cognition by regulating the arousal system.
Article
Fiber photometry is a key technique for characterizing brain-behavior relationships in vivo. Initially, it was primarily used to report calcium dynamics as a proxy for neural activity via genetically encoded indicators. This generated new insights into brain functions including movement, memory, and motivation at the level of defined circuits and cell types. Recently, the opportunity for discovery with fiber photometry has exploded with the development of an extensive range of fluorescent sensors for biomolecules including neuromodulators and peptides that were previously inaccessible in vivo. This critical advance, combined with the new availability of affordable “plug-and-play” recording systems, has made monitoring molecules with high spatiotemporal precision during behavior highly accessible. However, while opening exciting new avenues for research, the rapid expansion in fiber photometry applications has occurred without coordination or consensus on best practices. Here, we provide a comprehensive guide to help end-users execute, analyze, and suitably interpret fiber photometry studies.
Article
Full-text available
Human imaging studies have shown that spontaneous brain activity exhibits stereotypic spatiotemporal reorganization in awake, conscious conditions with respect to minimally conscious states. However, whether and how this phenomenon can be generalized to lower mammalian species remains unclear. Leveraging a robust protocol for resting-state fMRI (rsfMRI) mapping in non-anesthetized, head-fixed mice, we investigated functional network topography and dynamic structure of spontaneous brain activity in wakeful animals. We found that rsfMRI networks in the awake state, while anatomically comparable to those observed under anesthesia, are topologically configured to maximize interregional communication, departing from the underlying community structure of the mouse axonal connectome. We further report that rsfMRI activity in wakeful animals exhibits unique spatiotemporal dynamics characterized by a state-dependent, dominant occurrence of coactivation patterns encompassing a prominent participation of arousal-related forebrain nuclei and functional anti-coordination between visual-auditory and polymodal cortical areas. We finally show that rsfMRI dynamics in awake mice exhibits a stereotypical temporal structure, in which state-dominant coactivation patterns are configured as network attractors. These findings suggest that spontaneous brain activity in awake mice is critically shaped by state-specific involvement of basal forebrain arousal systems and document that its dynamic structure recapitulates distinctive, evolutionarily relevant principles that are predictive of conscious states in higher mammalian species.
Article
Full-text available
s Functional connectivity, which reflects the spatial and temporal organization of intrinsic activity throughout the brain, is one of the most studied measures in human neuroimaging research. The noninvasive acquisition of resting state functional magnetic resonance imaging (rs-fMRI) allows the characterization of features designated as functional networks, functional connectivity gradients, and time-varying activity patterns that provide insight into the intrinsic functional organization of the brain and potential alterations related to brain dysfunction. Functional connectivity, hence, captures dimensions of the brain's activity that have enormous potential for both clinical and preclinical research. However, the mechanisms underlying functional connectivity have yet to be fully characterized, hindering interpretation of rs-fMRI studies. As in other branches of neuroscience, the identification of the neurophysiological processes that contribute to functional connectivity largely depends on research conducted on laboratory animals, which provide a platform where specific, multi-dimensional investigations that involve invasive measurements can be carried out. These highly controlled experiments facilitate the interpretation of the temporal correlations observed across the brain. Indeed, information obtained from animal experimentation to date is the basis for our current understanding of the underlying basis for functional brain connectivity. This review presents a compendium of some of the most critical advances in the field based on the efforts made by the animal neuroimaging community.
Article
Full-text available
The triple-network model of psychopathology is a framework to explain the functional and structural neuroimaging phenotypes of psychiatric and neurological disorders. It describes the interactions within and between three distributed networks: the salience, default-mode, and central executive networks. These have been associated with brain disorder traits in patients. Homologous networks have been proposed in animal models, but their integration into a triple-network organization has not yet been determined. Using resting-state datasets, we demonstrate conserved spatio-temporal properties between triple-network elements in human, macaque, and mouse. The model predictions were also shown to apply in a mouse model for depression. To validate spatial homologies, we developed a data-driven approach to convert mouse brain maps into human standard coordinates. Finally, using high-resolution viral tracers in the mouse, we refined an anatomical model for these networks and validated this using optogenetics in mice and tractography in humans. Unexpectedly, we find serotonin involvement within the salience rather than the default-mode network. Our results support the existence of a triple-network system in the mouse that shares properties with that of humans along several dimensions, including a disease condition. Finally, we demonstrate a method to humanize mouse brain networks that opens doors to fully data-driven trans-species comparisons.
Article
Full-text available
Resting-state functional magnetic resonance imaging (fMRI) has drastically expanded the scope of brain research by advancing our knowledge about the topologies, dynamics, and interspecies translatability of functional brain networks. Several databases have been developed and shared in accordance with recent key initiatives in the rodent fMRI community to enhance the transparency, reproducibility, and interpretability of data acquired at various sites. Despite these pioneering efforts, one notable challenge preventing efficient standardization in the field is the customary choice of anisotropic echo planar imaging (EPI) schemes with limited spatial coverage. Imaging with anisotropic resolution and/or reduced brain coverage has significant shortcomings including reduced registration accuracy and increased deviation in brain feature detection. Here we proposed a high-spatial-resolution (0.4 mm), isotropic, whole-brain EPI protocol for the rat brain using a horizontal slicing scheme that can maintain a functionally relevant repetition time (TR), avoid high gradient duty cycles, and offer unequivocal whole-brain coverage. Using this protocol, we acquired resting-state EPI fMRI data from 87 healthy rats under the widely used dexmedetomidine sedation supplemented with low-dose isoflurane on a 9.4 T MRI system. We developed an EPI template that closely approximates the Paxinos and Watson's rat brain coordinate system and demonstrated its ability to improve the accuracy of group-level approaches and streamline fMRI data pre-processing. Using this database, we employed a multi-scale dictionary-learning approach to identify reliable spatiotemporal features representing rat brain intrinsic activity. Subsequently, we performed k-means clustering on those features to obtain spatially discrete, functional regions of interest (ROIs). Using Euclidean-based hierarchical clustering and modularity-based partitioning, we identified the topological organizations of the rat brain. Additionally, the identified group-level FC network appeared robust across strains and sexes. The “triple-network” commonly adapted in human fMRI were resembled in the rat brain. Through this work, we disseminate raw and pre-processed isotropic EPI data, a rat brain EPI template, as well as identified functional ROIs and networks in standardized rat brain coordinates. We also make our analytical pipelines and scripts publicly available, with the hope of facilitating rat brain resting-state fMRI study standardization.
Article
Full-text available
Previous research has shown that the autonomic nervous system provides essential constraints over ongoing cognitive function. However, there is currently a relative lack of direct empirical evidence for how this interaction manifests in the brain at the macro-scale level. Here, we examine the role of ascending arousal and attentional load on large-scale network dynamics by combining pupillometry, functional MRI and graph theoretical analysis to analyze data from a visual motion-tracking task with a parametric load manipulation. We found that attentional load effects were observable in measures of pupil diameter and in a set of brain regions that parametrically modulated their BOLD activity and meso-scale network-level integration. In addition, the regional patterns of network reconfiguration were correlated with the spatial distribution of the α2a adrenergic receptor. Our results further solidify the relationship between ascending noradrenergic activity, large-scale network integration, and cognitive task performance.
Article
Full-text available
Superparamagnetic iron-oxide nanoparticles are robust contrast agents for magnetic resonance imaging (MRI) and has shown promise for sensitive structural and functional mapping of the cerebral blood volume (CBV) when administered intravenously. To date, many CBV-MRI studies are conducted with Feraheme, manufactured for the treatment of iron-deficiency. Unfortunately, Feraheme is currently not available outside the United States due to commercial and regulatory constraints, making the CBV-MRI method either inaccessible or very costly to achieve. To address this barrier, we developed a simple, one-pot recipe to synthesize Carboxymethyl-dextran coated Iron Oxide Nanoparticles, namely, “CION”, suitable for preclinical CBV-MRI applications. Here we disseminate a step-by-step instruction of our one-pot synthesis protocol, which allows CION to be produced in laboratories with minimal cost. We also characterized CIONs in terms of their size, surface chemistry, and chemical composition, and described the tuning of MR relaxivity and pharmacokinetics by manipulating polymer to metal stoichiometric ratio. We performed several proof-of-concept experiments in vivo, demonstrating the utility of CION for functional and structural MRI applications, including hypercapnic CO2 challenge, visual stimulation, targeted optogenetic stimulation, and microangiography. We also present evidence that CION can serve as a cross-modality research platform by showing concurrent optical and MRI measurement of CBV using fluorescent-labeled CION in vivo. The simplicity and cost-effectiveness of our one-pot synthesis method should allow researchers to reproduce CION and tailor the relaxivity and pharmacokinetics according to their imaging needs. It is our hope that this work would make the CBV-MRI platform openly available and easily affordable for a variety of research applications.
Article
Full-text available
Arousal may be understood on a spectrum, with excessive sleepiness, cognitive dysfunction, and inattention on one side, a wakeful state in the middle, and hypervigilance, panic, and psychosis on the other side. However, historically, the concepts of arousal and stress have been challenging to define as measurable experimental variables. Divergent efforts to study these subjects have given rise to several disciplines, including neurobiology, neuroendocrinology, and cognitive neuroscience. We discuss technological advancements that chronologically led to our current understanding of the arousal system, focusing on the multifaceted nucleus locus coeruleus. We share our contemporary perspective and the hypotheses of others in the context of our current technological capabilities and future developments that will be required to move forward in this area of research.
Conference Paper
Using a relatively high model order of independent component analysis (ICA with 75 ICs) of functional magnetic resonance imaging (fMRI) data, we have reported a clear effect of spatial smoothing Gaussian kernel size on spatiotemporal properties of intrinsic connectivity networks (ICNs). However, many if not the majority of ICA fMRI studies are usually performed at low model order, e.g., 20-IC decomposition, as such low order is generally enough to extract the few networks of interest such as the default-mode network (DMN). The aim of this study is to investigate if we can replicate the spatial smoothing effects on spatiotemporal features of ICNs at low ICA model order. Same resting state fMRI data that we used with 75-IC analysis were used here. Spatial smoothing using an isotropic Gaussian filter kernel with full width at half maximum (FWHM) of 4, 8, and 12 mm was applied during preprocessing. ICNs were identified from 20-IC decomposition and evaluated in terms of three primary features: spatial map intensity, functional network connectivity (FNC), and power spectra. The results identified similar effects of spatial smoothing on spatial map intensities and power spectra at p < 0.01, false discovery rate (FDR) corrected for multiple comparisons. Reduced spatial smoothing kernel size resulted in decreased spatial map intensities as well as a generally decreased low-frequency power (0.01 - 0.10 Hz) but increased high-frequency power (0.15 - 0.25 Hz). FNC, however, did not show a uniform change in correlation values with the size of smoothing kernel. Notably, FNC between DMNs decreased but FNC between central executive and visual networks increased with an increase in smoothing kernel size. These preliminary findings confirm spatial smoothing influences ICN features regardless of model order. The discussion focuses on differences between observed changes at low and high ICA model orders.
Article
Background A trend in the development of resting-state fMRI (rsfMRI) data analysis is the drive towards more data-driven methods. Group Independent Component Analysis (GICA) is a well-proven data-driven method for performing fMRI group analysis, though not without issues, especially the back-reconstruction from group-level independent components to individual-level components. Group information-guided ICA (GIG-ICA) and Independent Vector Analysis (IVA) are recent extensions of GICA that were shown to outperform GICA in the identification of unique rsfMRI biomarkers in psychiatric conditions. New Method In this work, GICA, GIG-ICA, and IVA-GL analysis methods were applied to rsfMRI data acquired from 9 mice under different doses of medetomidine (0.1 – 0.3 mg/kg/h) in the before and after forepaw stimulation, and their performance was compared to determine whether GIG-ICA and IVA-GL outperform GICA in identifying robust and reliable resting-state networks in the rodent brain. Results Our results showed IVA-GL method had certain desirable performance characteristics over the other two methods under minimal data pre-processing and data-driven assumptions in application to analysis of mouse resting-state functional MRI. Comparison with existing methods: IVA-GL provides better stability towards detecting group differences at different model order assumptions and performed better at separating data well-defined and functionally reasonable components in mouse resting-state fMRI. At higher model order and more likely functional component assumptions, GIG-ICA and IVA-GL were found to have greater sensitivity at detecting functional connectivity changes due to physiological challenges compared to GICA. Conclusions This study indicates that IVA-GL yields better detection of resting-state networks in the rodent brain compared to other ICA methods and a promising data-driven analysis method for rodent rsfMRI.