SlideShare a Scribd company logo
Corticolimbic anatomical characteristics
predetermine risk for chronic pain
Etienne Vachon-Presseau,1
Pascal Te´treault,1
Bogdan Petre,1
Lejian Huang,1
Sara E. Berger,1
Souraya Torbey,2
Alexis T. Baria,1
Ali R. Mansour,1
Javeria A. Hashmi,3
James W. Griffith,4
Erika Comasco,5
Thomas J. Schnitzer,6
Marwan N. Baliki1,7
and A. Vania Apkarian1
Mechanisms of chronic pain remain poorly understood. We tracked brain properties in subacute back pain patients longitudinally
for 3 years as they either recovered from or transitioned to chronic pain. Whole-brain comparisons indicated corticolimbic, but not
pain-related circuitry, white matter connections predisposed patients to chronic pain. Intra-corticolimbic white matter connectivity
analysis identified three segregated communities: dorsal medial prefrontal cortex–amygdala–accumbens, ventral medial prefrontal
cortex–amygdala, and orbitofrontal cortex–amygdala–hippocampus. Higher incidence of white matter and functional connections
within the dorsal medial prefrontal cortex–amygdala–accumbens circuit, as well as smaller amygdala volume, represented inde-
pendent risk factors, together accounting for 60% of the variance for pain persistence. Opioid gene polymorphisms and negative
mood contributed indirectly through corticolimbic anatomical factors, to risk for chronic pain. Our results imply that persistence of
chronic pain is predetermined by corticolimbic neuroanatomical factors.
1 Department of Physiology, Feinberg School of Medicine, Northwestern University 303 E. Chicago Ave., Chicago, IL 60611, USA
2 Department of Psychiatry and Neurobehavioral Sciences, University of Virginia , 2955 Ivy Rd, Suite 210, Charlottesville, VA
22903, USA
3 Department of Anesthesia, Pain Management and Perioperative Medicine Dalhousie University, Halifax, NS, Canada B3H 4R2
4 Department of Medical Social Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
5 Department of Neuroscience, Science for Life Laboratory, Uppsala University, BMC, Pob 593, 75124, Uppsala, Sweden
6 Northwestern University Feinberg School of Medicine, Departments of Physical Medicine and Rehabilitation and Internal
Medicine/Rheumatology, 710 N. Lake Shore Drive, Room 1020, Chicago, IL 60611, USA
7 Rehabilitation Istitute of Chicago, 345 E Superior St, Chicago, IL 60611, USA
Correspondence to: A. V. Apkarian,
Department of Physiology,
Feinberg School of Medicine,
Northwestern University 303 E. Chicago Ave.,
Chicago, IL 60611, USA
E-mail: a-apkarian@northwestern.edu
Correspondence may also be addressed to: Marwan N. Baliki,
E-mail: marwanbaliki2008@u.northwestern.edu
Keywords: chronic pain; brain network; limbic system; magnetic resonance imaging (MRI); diffusion tensor imaging (DTI)
Abbreviations: DTI = diffusion tensor imaging; PFC = prefrontal cortex; SBP = subacute back pain; SNP = single nucleotide
polymorphism
doi:10.1093/brain/aww100 BRAIN 2016: Page 1 of 13 | 1
Received December 18, 2015. Revised February 11, 2016. Accepted March 16, 2016.
ß The Author (2016). Published by Oxford University Press on behalf of the Guarantors of Brain. All rights reserved.
For Permissions, please email: journals.permissions@oup.com
Brain Advance Access published May 5, 2016
byguestonMay9,2016http://brain.oxfordjournals.org/Downloadedfrom
Introduction
Chronic pain is a state of continued negative emotional suf-
fering representing a leading source of worldwide disability
(Murray and Lopez, 2013) with a staggeringly high healthcare
cost (Medicine, 2011). Risk factors for developing chronic
pain remain poorly understood, yet its negative emotional
valence implicates the involvement of limbic circuitry. In
fact, both Maclean’s (1955) original formulation of the
limbic brain and Melzack and Casey’s (1968) concept of
the gate control theory of pain (Melzack and Wall, 1965)
hypothesized that the limbic brain plays an integral role in
pain states. Consistently, long-standing clinical evidence has
demonstrated that injury to limbic structures diminishes the
emotional overtones of pain (Raz, 2009). However, only
recent human neuroimaging (Apkarian et al., 2004; Baliki
et al., 2006, 2010, 2012; Geha et al., 2008; Hashmi et al.,
2013; Mansour et al., 2013; Mutso et al., 2014) and comple-
mentary animal model (Neugebauer et al., 2003; Metz et al.,
2009; Ji and Neugebauer, 2011; Ren et al., 2011; Mutso
et al., 2012; Baliki et al., 2014; Chang et al., 2014;
Schwartz et al., 2014) studies have begun to unravel the
role of limbic brain properties in chronic pain.
In the present study, we focus on intrinsic properties of the
corticolimbic portion of the mesocorticolimbic system. A net-
work comprised of the prefrontal cortex (PFC) (medial and
orbital cortices), nucleus accumbens, hippocampus/parahippo-
campus, and amygdala. Collectively, these structures support
emotion, behaviour, motivation and memory functions.
Converging evidence reinforces the idea that components of
this circuitry are important for chronic pain. In humans, the
nucleus accumbens encodes salience of impending pain and
the reward value of analgesia, which is distorted in chronic
back pain (Baliki et al., 2010), and functional connectivity
between nucleus accumbens and medial PFC prospectively
predicts chronification of pain (Baliki et al., 2012).
Prefrontal cortical activity reflects subjective reports of back
pain intensity in patients with chronic back pain (Baliki et al.,
2006; Hashmi et al., 2013) and is thought to regulate the
meaning of affective responses and influence emotional deci-
sion-making. Furthermore, the size of the amygdala and
hippocampus show strong genetic dependences (Hibar et al.,
2015) and both structures are implicated in emotional learn-
ing (Phelps and LeDoux, 2005), anxiety (Russo et al., 2012),
and stress regulation (Vachon-Presseau et al., 2013), and ex-
hibit changes in activity and functional connectivity during the
transition to chronic pain (Mutso et al., 2012; Hashmi et al.,
2013). Comparative evidence further bolsters corticolimbic in-
volvement in chronic pain by demonstrating that single
neuron morphology, excitability, and short- or long-term po-
tentiation are distorted within and across the hippocampus
(Ren et al., 2011; Mutso et al., 2012), amygdala
(Neugebauer et al., 2003; Ji and Neugebauer, 2011), nucleus
accumbens (Chang et al., 2014; Schwartz et al., 2014), and
PFC (Metz et al., 2009; Ji and Neugebauer, 2011).
Consistently, the persistence of pain-like behaviours in rodents
is accompanied by a reorganization of resting state connect-
ivity primarily across limbic brain structures (Baliki et al.,
2014). Despite this compelling evidence, a comprehensive
examination of corticolimbic brain contributions to chronic
pain remains to be undertaken. Here we interrogate the func-
tional and anatomical properties of the corticolimbic neural
network, and its subcircuits, to unravel the interrelationships
between change in pain intensity, negative emotion, and their
potential genetic mediators. We report the novel finding that
anatomical characteristics of specific networks and structures
of the corticolimbic system predetermine risk for developing
chronic pain.
Materials and methods
Additional information regarding whole brain region of inter-
est parcellation schemes, pain and corticolimbic nodes, corti-
colimbic parcellation, modularity analyses of limbic structural
network, and genetic analyses can be found in the
Supplementary material.
Participants
The data presented are from a longitudinal observational study
where we followed patients with subacute back pain (SBP) over 3
years. This report is based on the final total sample of patients.
Portions of the data were used in previous publications (Baliki
et al., 2012; Hashmi et al., 2013; Mansour et al., 2013; Petre
et al., 2015). The study recruited 159 patients with SBP and 29
healthy control subjects. All patients with SBP were diagnosed by
a clinician for back pain and reported pain intensity greater than
40/100 on a visual analogue scale (VAS, 0–100, where
100 = maximum imaginable pain and 0 = no pain) prior to en-
rolling in the study. Patients with SBP were recruited only if the
duration of their pain was between 4–16 weeks with no back
pain reported during the previous 12 months. Subjects were
excluded if they reported other chronic painful conditions, sys-
temic disease, psychiatric diseases, or history of head injuries.
Subjects were also excluded if they reported high depression,
which was defined as scores 419 using the Beck’s Depression
Inventory (BDI) (Beck et al., 1961). From these participants, 69
SBP (34 females, 43.1 Æ 10.4 years of age) and 20 healthy con-
trols (nine females, 37.4 Æ 7.5 years of age) completed the study
to 1-year follow-up. These patients with SBP were dichotomized
into groups with persisting pain (SBPp; n = 39) or recovery from
pain at the end of the year (with recovery operationalized as 20%
reduction in pain from Week 0 to Week 56; SBPr n = 30). A
subsample of 39 patients with SBP (18 females, 44.2 Æ 11.3
years of age) was followed for an additional visit at 3 years
from pain onset. Dates of visits are shown for all subjects in
Supplementary Fig. 1. The Institutional Review Board of
Northwestern University approved the study and all participants
signed a consent form.
Additional groups of patients and healthy controls were used
to cross-sectionally examine subcortical limbic volumes: 23
with chronic back pain (nine females, 46.6 Æ 6.0 years of
age), 40 osteoarthritis patients (22 females, 58.6 Æ 7.6 years
of age), and 20 healthy age-matched controls (10 females,
57.9 Æ 6.7 years of age).
2 | BRAIN 2016: Page 2 of 13 E. Vachon-Presseau et al.
byguestonMay9,2016http://brain.oxfordjournals.org/Downloadedfrom
Pain characteristics, depressive
mood, and negative affect
Pain intensity was measured using the visual analogue scale
(VAS) from the short version of the McGill Pain
Questionnaire (Melzack, 1975) and disability was assessed
using the Pain Disability Index (PDI) measuring the degree at
which chronic pain disrupts aspects of the patients’ lives (Tait
et al., 1990). Negative affect was calculated with the negative
scale of the Positive and Negative Affectivity Scale (PANAS).
This scale consists of a series of items describing different emo-
tions experienced over the previous week (Watson and Clark,
1999). Depressive mood was assessed using BDI-1b. All ques-
tionnaires were given within an hour before brain scanning.
Summary results for pain characteristics, depressive mood and
negative affect are presented in Supplementary Table 1.
Scanning parameters
Anatomical (T1), diffusion tensor imaging (DTI), and functional
MRI were collected in the same scanning session. High-reso-
lution T1-anatomical brain images were acquired with a 3 T
Siemens Trio whole-body scanner with echo-planar imaging
capability using the standard radio-frequency head coil using
the following parameters: voxel size = 1 Â 1 Â 1 mm, repetition
time = 2.50 ms, echo time = 3.36 ms, flip angle = 9
, in-plane
matrix resolution = 256 Â 256; 160 slices, field of
view = 256mm.
Functional MRI images were acquired using the following
parameters: multi-slice T2*-weighted echo-planar images with
repetition time = 2.5 s, echo time = 30 ms, flip angle = 90
,
number of volumes = 244, slice thickness = 3 mm, in plane
resolution = 64 Â 64. The 36 slices covered the whole brain
from the cerebellum to the vertex.
DTI images were acquired using echo planar imaging
(72 Â 2-mm thick axial slices; matrix size = 128 Â 128;
field of view = 256 Â 256 mm2
, resulting in a voxel size of
2 Â 2 Â 2 mm). Images had an isotropic distribution along 60
directions using a b-value of 1000 s/mm2
. For each set of diffu-
sion-weighted data, eight volumes with no diffusion weighting
were acquired at equidistant points throughout the acquisition.
Morphometric analyses of subcortical
structures
Structural data were analysed with the standard automated
processing stream of FSL 4.1 that shows very high reliability
across labs (Nugent et al., 2013). The analysis entailed skull
extraction, a two-stage linear subcortical registration, and seg-
mentation using FMRIB’s Integrated Registration and
Segmentation Tool (FIRST). The volumes of right and left nu-
cleus accumbens, amygdala, hippocampus, and thalamus were
calculated for each subject. Group comparisons were per-
formed using repeated analyses of covariance with volume as
the dependent factor, time of the scan and hemisphere as
within-subject factors, the group as a between-subject factor
(SBPp, SBPr, and healthy), and age, sex and total grey matter
volume [estimated using FSL SIENAX (Smith et al., 2002)] as
covariates of no interest. Post hoc comparisons were
Bonferroni corrected for multiple comparisons.
From the initial 89 participants, nine were excluded because
T1 was missing or because of quality control failure of the T1
image at one of the four visits. Quality control included: (i)
visual inspection of the subcortical segmentation, where gross
mismatches between underlying anatomy and FIRST outcome
were excluded; and (ii) large deviations from the mean volume
of a given structure were also excluded [greater than Æ2
standard deviations (SD)]. Outliers were identified independ-
ently for hippocampus and amygdala volumes. From a total of
80 subjects, 11 were rejected from the amygdala analyses and
four were rejected from the hippocampus analyses.
Importantly, removing these individuals did not change the
overall statistical outcomes of our results. Our subsample of
patients scanned a fifth time at Week 156 included 33 patients
with accurate segmentation of subcortical structures.
Differences between SBPp and SBPr in amygdala and hippo-
campus volumes, initially discovered using FSL, were repli-
cated using the standard automated cortical and subcortical
segmentations by Freesurfer 5.0 (http://surfer.nmr.mgh.har
vard.edu/). The procedure includes motion correction, removal
of the skull using watershed/surface deformation procedure,
normalization in Talairach space, and segmentation of the sub-
cortical structures based on the existing atlas containing prob-
abilistic information on the location of structures (Fischl et al.,
2002).
As hippocampus and amygdala exhibited significant group
differences, we investigated whether these changes can be
attributed to localized structural deformations. Vertex repre-
sentation of both structures were therefore generated using
FIRST (Patenaude et al., 2011). The model uses a Bayesian
approach representing the structure in 3D from meshes fitting
the form of the subcortical structure to a training dataset that
was manually segmented. The shapes of the subcortical struc-
tures were represented from meshes parameterized by the co-
ordinates of their vertices. A multivariate permutation test on
the 3D coordinates of corresponding vertices provide the
F-values representing group differences (SBPp and SBPr).
Group differences in vertex localized at the boundary provided
a geometric change shape of the amygdala and the hippocam-
pus. No corrections for multiple comparisons were performed
on these analyses.
Diffusion tensor imaging
preprocessing
FAST (FMRIB Automated Segmentation Tool) was used to
segment the 3D T1 image of the brain into different tissue
types (grey matter, white matter, and CSF) while also correct-
ing for spatial intensity variations (Supplementary material).
For each subject, the grey matter and white matter masks
were transformed into standard space using FSL linear regis-
tration tool (FLIRT). Those masks were then thresholded
(probability 4 0.25) and multiplied with each other, yielding
a subject specific grey–white matter interface composed of re-
gions having relatively equal probabilities of being in either
category, thus constituting the grey–white matter boundary.
Probabilistic tractography of the whole brain networks were
constructed from voxels composing this grey–white matter
boundary.
Analysis was performed using tools from the FMRIB
Diffusion Toolbox (FDT; Behrens et al., 2003) and in-house
Anatomical risk factors for chronic pain BRAIN 2016: Page 3 of 13 | 3
byguestonMay9,2016http://brain.oxfordjournals.org/Downloadedfrom
software. DTI data were manually checked for obvious arte-
facts. Eddy current correction was used and simple head
motion correction using affine registration to a no-diffusion
reference volume for each participant. Data were then skull
extracted (BET) and a diffusion tensor model was fit at each
voxel determining voxel-wise fractional anisotropy. Bayesian
Estimation of Diffusion Parameters Obtained using Sampling
Techniques (BEDPOSTX) and probabilistic tractography
(PROBTRACX) modelling two fibre tracks per voxel were
used to determine the probability of connection between re-
gions (Behrens et al., 2007). Voxel-wise principal diffusion
directions were used to repetitively compute a probabilistic
streamline from 5000 samples to generate posterior distribu-
tion on the ‘connectivity distribution’. Probabilistic tractogra-
phy was performed in the diffusion space of each participant
and then transformed to MNI space using FSL linear registra-
tion (FLIRT; Jenkinson et al., 2002). The connectivity distri-
bution was averaged across voxels of each of the 264 and 480
regions of interest (Supplementary Fig. 3) for whole brain net-
work analyses or across voxels of the subsample in
4 Â 4 Â 4 mm for the corticolimbic network analyses (1068
regions of interest; Supplementary Fig. 4). From the 89 par-
ticipants, 15 were excluded because DTI images were missing
at one of the four visits or because of quality control failure.
From the remaining 74 patients, two patients were further
excluded from the limbic network analyses because they
showed connectivity counts in the dorsal medial PFC–amyg-
dala–nucleus accumbens 43 SD from the group mean.
Diffusion scans were collected in 36 patients out of the sub-
sample of patients scanned a fifth time at Week 156.
Functional MRI preprocessing
Ten minutes of functional MRI were acquired at each visit of
the study, where subjects rated spontaneous fluctuations of
their pain. These data were used to examine intrinsic brain
connectivity differences between groups (SBPp and SBPr).
The preprocessing of each subject’s time series of functional
MRI volumes was performed using the FMRIB Expert
Analysis Tool (FEAT). The sequence included skull extraction
using BET from FSL, slice time correction, and motion correc-
tion (Jenkinson et al., 2002). Temporal filtering between 0.01
and 0.1 Hz was applied on the blood oxygenation level-de-
pendent (BOLD) time series. The six parameters of head
motion, global mean signal measured over all voxels of the
brain, the signal from a ventricular region of interest, and
the signal from a region centred in the white matter were re-
gressed as covariates of no interest from the BOLD time series.
The first four volumes were removed for signal stabilization.
After preprocessing, functional scans were registered into a
standard MNI space using FSL FLIRT. From the 69 patients
that rated their spontaneous pain fluctuations, seven were
excluded because of missing functional scans at one of the
four visits. Our subsample of patients scanned a fifth time at
Week 156 included 34 patients with functional scans.
Construction of white matter
networks
Whole brain DTI-based connectivity networks for the 480 re-
gions of interest for SBPp and SBPr at Week 0 were
constructed using methods described in (Rubinov and
Sporns, 2010). For all regions of interest, the connection prob-
ability between a seed region of interest and any target region
of interest in the brain is defined as the sum of sample fibre
lengths connecting these two regions of interest. For each sub-
ject, a 480 Â 480 and a 264 Â 264-weighted directed matrices
were generated from connection probabilities for all seed re-
gions of interest with the number of target regions of interest.
The resultant matrices were then symmetrized and adjusted for
distance constraints by multiplication with Euclidean distance
matrix. Finally, the matrix was thresholded at a value that
yielded a binary undirected network of different link densities
ranging from 0.02 to 0.20 (i.e. the top 2–20% of connections
were assigned a value of 1, whereas the rest of the connections
were assigned a value of 0; Supplementary Fig. 6). Group dif-
ferences (SBPp and SBPr) in edges connecting nodes of the
network were initially determined using permutation test con-
trolling for family-wise error rate using Network Based
Statistics (NBS; Zalesky et al., 2010). Corticolimbic white
matter networks formed between 1068 Â 1068 nodes were
generated for all subjects (SBPp, SBPr and healthy controls),
at each of the five visits using the same methods.
Construction of functional brain
networks
To construct the limbic functional connectivity networks for
each subject, we first computed the Pearson correlation coeffi-
cient (r) for the all possible pairs of the n = 1068 of cortical
and subcortical limbic regions of interest time series from the
preprocessed resting state functional MRI data. Similar to
white matter networks, threshold was calculated to produce
a fixed number of connections across link densities ranging
from 0.02 to 0.20 (top 2–20% of r-values binarized), for
each subject (Achard et al., 2012). The values of the threshold
are therefore subject-dependent but the number of connections
in the network is the same for every subject. The density of
structural connections was examined with respect to the com-
munity-based organization of the white matter network.
Therefore, group differences (SBPp and SBPr) in functional
connectivity within and across modules determined from
white matter tractography were tested using repeated measure
ANOVAs entering the visits as repeated variable.
Genetic association with mesolimbic
brain outcome measures
To test the idea that some of the mesolimbic brain properties
associated with risk of chronic pain may underlie a genetic pre-
disposition, we looked for associations between single nucleotide
polymorphisms (SNPs) and limbic brain properties. Saliva sam-
ples were collected from all subjects who completed the study.
Given the limited number of subjects in the study we decided to
only examine 30 SNPs. The choice of SNPs was determined prior
to having knowledge regarding the primary functional or ana-
tomical determinants of risk for chronic pain. The 30 candidate
SNPs located in 12 different genes were selected for their known
involvement in neurotransmitters, receptors and metabolites asso-
ciated with corticolimbic structures, and also for being related to
pain or nociception. Only one gene was selected as a marker for
peripheral encoding of nociception, SCN9A. A description of
4 | BRAIN 2016: Page 4 of 13 E. Vachon-Presseau et al.
byguestonMay9,2016http://brain.oxfordjournals.org/Downloadedfrom
each gene is presented in Supplementary Table 8. Genetic analysis
was done independently and with no knowledge of the results of
brain analyses. Four brain outcome parameters were tested for
association with the 30 selected SNPs. SNPs that passed statistical
threshold (or were borderline significant) after Bonferroni mul-
tiple comparison correction were also tested for discrimination
between SBPp and SBPr.
Statistical analyses
Path analyses and multiple regression models
Path analysis was used to examine the direct and mediating
links between multiple variables to test a given theoretical
model empirically, specifically testing the prediction for SBP
grouping at Week 56 using a logistic regression model. Path
analyses were performed with Mplus 7.3 using the robust
weighted least squares estimator method accounting for the
non-normality of the data. We calculated 95% confidence
intervals for the indirect effect using a recommended bias-
corrected bootstrapped method (bootstrap of 1000 iterations;
Mackinnon et al., 2004); we rejected the null hypothesis when
the 95% confidence interval did not contain zero.
Results
This report presents the complete results of a longitudinal
study. Patients recruited while in subacute back pain were
followed over the course of 1 year (n = 69 patients with
SBP), with a smaller subset followed for 3 years (n = 39).
Thirty-nine patients with SBP were dichotomized into per-
sisting pain (SBPp) and 30 SBP were dichotomized into
recovery from pain as they showed 20% reduction in
pain intensity between Week 0 and Week 56 (SBPr).
Classification of patients with SBP at 3 years was also
based on the Week 56 criterion (SBPp n = 23, SBPr
n = 16). Group differences in pain intensity and disability
were observed as early as 8 weeks after entry into the study
and maintained temporal stability up to 3 years after back
pain onset (Fig. 1B). Over 3 years, no changes were
observed in negative psychological traits for SBPp or SBPr
(Supplementary Table 1), and medication use was not dif-
ferent between SBP groups (Supplementary Fig. 2).
Whole-brain white matter
connectivity
We used DTI probabilistic tractography-based structural
connectivity at study entry to identify brain areas for
which differences in white matter connectivity conferred
greater risk for chronic pain. The DTI scans at study
entry (10.8 Æ 5.5 weeks from onset of back pain) were
used to construct whole-brain structural networks
(Supplementary Fig. 3) from different parcellation schemes
of 480 and 264 regions (Fig. 1C and Supplementary Fig.
6). Permutation tests were used to identify group differ-
ences in the incidence of connections, for each parcellation
scheme, across link densities ranging from 0.02 to 0.2.
Example group contrast results (SBPp 4 SBPr, and
SBPr 4 SBPp; Supplementary Tables 3 and 4) for both par-
cellations, at 0.1 link density, are illustrated in Fig. 1D and
E. Connections were denser in SBPp and mostly localized
across corticolimbic nodes (Supplementary Tables 5 and 6).
In addition to corticolimbic nodes, we also identified nodes
related to pain perception using a term-based meta-analysis
tool (based on a map derived for ‘pain’ from 420 PubMed
publications; www.neurosynth.org). Group differences in
structural connections were localized outside pain-related
nodes. Instead, the likelihood for denser connections in
SBPp, compared to SBPr, was consistently higher within
the corticolimbic brain compared to random networks
(Fig. 1F and G). Collectively, these analyses indicate that
structural connections between regions involved in pain
processing were not involved in the development of chronic
pain. In contrast, denser white matter connections were
observed between nodes concentrated within the cortico-
limbic system, implying that these connections were more
likely to predispose individuals to transition to chronic
pain.
Corticolimbic modules contribute to
risk for chronic pain
The identification of dense corticolimbic white matter con-
nections does not provide information about underlying
anatomical and functional properties of this system that
may impart risk for chronic pain. Therefore, we next inter-
rogated the intrinsic structural and functional network
organization of the corticolimbic brain throughout the
3 years of observation, using higher spatial resolution
data. Anatomical networks were first constructed from con-
nections between 1068 nodes (4 Â 4 Â 4 mm voxels) be-
longing to the corticolimbic system (nucleus accumbens,
amygdala, hippocampus-parahippocampus, and the pre-
frontal cortex) (Fig. 2A and Supplementary Fig. 4). Edges
between the nodes were drawn from DTI-based probabil-
istic white matter tractography and from Pearson correl-
ations between functional MRI time series (i.e. functional
connections derived from functional MRI activity as pa-
tients rated back pain intensity). The structural (white
matter network) and functional (functional connectivity
network) graphs were constructed by binarizing the net-
works across different densities ranging from 0.02 to
0.20 (Supplementary Fig. 6). Derived networks were
tested and confirmed to be insensitive to standard con-
founds (Supplementary Fig. 5).
We first studied the architecture of the limbic white
matter network (Fig. 2A). This fine-grained, voxel-based,
intra-limbic probabilistic tractography-based white matter
network revealed dense, bilateral connections of subcortical
structures with each other and with the prefrontal cortex.
Modularity analysis revealed that this network was com-
posed of three distinct communities, including modules
composed of (i) dorsal and medial portion of PFC,
Anatomical risk factors for chronic pain BRAIN 2016: Page 5 of 13 | 5
byguestonMay9,2016http://brain.oxfordjournals.org/Downloadedfrom
medial-inferior part of the amygdala, and most of the nu-
cleus accumbens (dorsal medial PFC–amygdala–nucleus
accumbens); (ii) most of the ventral medial frontal cortex
(ventral medial PFC) and medial-superior part of the amyg-
dala (ventral medial PFC–amygdala); and (iii) a thin layer
of orbital cortex linked to lateral amygdala and most of
hippocampus (orbitofrontal cortex–amygdala–hippocam-
pus) (Fig. 2B). The stability of the community structure
was preserved across different clustering thresholds
(Supplementary Fig. 7). Note that our results are consistent
Figure 1 Whole-brain structural connectivity identifies corticolimbic regions conferring risk for chronic pain. (A) Timeline of
brain scans and type and number of subjects studied. (B) Recovering (SBPr) and persisting (SBPp) patients were dichotomized based on 20%
reduction in back pain intensity at Week 56. The two groups showed time-dependent divergence (Weeks 0–56) in pain intensity [F(2.52, 140.92);
P 5 0.001 repeated measure ANOVA] and pain disability [F(2.58, 110.74) = 3.44; P = 0.025 repeated measure ANOVA], sustained in the 3-year
subsamples [Week 156 pain intensity t(1,33) = 3.67; P = 0.001; pain disability t(1,36) = 3.28; P = 0.002 two-sided unpaired t-tests]. *
P 5 0.05
between group comparison; +
P 5 0.05 within group comparison relative to Week 0. (C) Structural white matter-based networks were con-
structed from probabilistic tractography between nodes covering the whole brain. (D and F) Group comparison at link density of 0.1 at Week 0
was studied from network constructed from 480 and 264 parcellation schemes. In both networks SBPp show more white matter connections
between corticolimbic nodes (P 5 0.05, FDR corrected). (E) In 480-parcellation, location and number of nodes within pain and corticolimbic
systems are illustrated. The SBPp 4 SBPr contrast indicates denser structural connections in SBPp across corticolimbic nodes, but not for pain-
related nodes, for all link densities. Observed incidence of connections compared to expected incidence in a random network was significantly
higher for corticolimbic nodes (chi-square test, P 5 0.0001 for all link densities) but not for pain-related nodes (P 4 0.28 for all link densities).
Histogram is the cumulative probability (total connectivity = 1.0) for corticolimbic nodes and pain-related nodes compared to expected con-
nectivity in a random network, averaged for all densities. (G) Concordant results were observed in 264-parcellation. Observed incidence of
connections in SBPp was much higher than expected across corticolimbic nodes (chi-square test, P 5 0.0001 for all link densities) but not for pain-
related nodes (P 4 0.28 for all link densities). Number of subjects is shown in parentheses. Error bars indicate SEM.
6 | BRAIN 2016: Page 6 of 13 E. Vachon-Presseau et al.
byguestonMay9,2016http://brain.oxfordjournals.org/Downloadedfrom
with the anatomical subdivisions of the amygdala, based on
meta-analytic functional co-activation maps of the amyg-
dala with the rest of the brain, which also show a close
correspondence to the region’s cytoarchitectonic subdiv-
isions (Bzdok et al., 2013) (Supplementary Fig. 8).
We next studied group differences and temporal variabil-
ity in density of intra-limbic white matter and functional
connectivity networks, both within and between the three
corticolimbic circuits. Although white matter network
modular organization was common across SBPr, SBPp
and healthy individuals (Fig. 2C), SBPp consistently dis-
played a greater frequency of white matter connections
within the dorsal medial PFC–amygdala–nucleus accum-
bens module across all link densities (Fig. 2D), across the
four visits (Fig. 2E) and 3 years after pain onset (Fig. 2E).
The white matter networks of ventral medial PFC–amyg-
dala and orbitofrontal cortex–amygdala–hippocampus
modules did not differ between SBP groups
(Supplementary Fig. 9A). The dorsal medial PFC–amyg-
dala–nucleus accumbens white matter network was stable
over 3 years because individual SBP values were strongly
correlated between Week 0 and Week 156 for all SBP (Fig.
2E). This finding is consistent with and generalizes from a
previous report that white matter regional fractional anisot-
ropy distinguishes between SBP groups (based on a sub-
sample of the current cohort; Mansour et al., 2013).
Examining the functional connectivity network connec-
tions in each one of the modules, defined in Fig. 2B,
showed that SBPp displayed higher incidence of connec-
tions within the dorsal medial PFC–amygdala–nucleus
accumbens module at all densities (Fig. 2F), at all four
visits (Fig. 2G), a finding that expands on our previous
report of stronger (seed-to-target) medial PFC–nucleus
accumbens functional connectivity in a subgroup of SBPp
(Baliki et al., 2012). However, the SBP group difference in
dorsal medial PFC–amygdala–nucleus accumbens func-
tional network connectivity was not maintained 3 years
later (Fig. 2G and Supplementary Fig. 10A and B), with
non-significant correlations between study entry and Week
156 (Fig. 2H). The functional connectivity networks of ven-
tral medial PFC–amygdala and orbitofrontal cortex–amyg-
dala–hippocampus modules did not differ between SBP
groups (Supplementary Fig. 9B). Taken together, these re-
sults reinforce that the functional connectivity within the
dorsal medial PFC–amygdala–nucleus accumbens is predict-
ive of the transition to chronic pain 1 year later; in con-
trast, its impact dissipates by Year 3, suggesting that it is
not required for the longer term maintenance of chronic
pain.
Amygdala, hippocampus and risk for
chronic pain
Morphometric measures of the amygdala and hippocampus
(Fig. 3A and B) were examined because they both show
strong genetic dependence (Hibar et al., 2015), represent
risk factors for developing pathological emotional states
such as anxiety (Russo et al., 2012) and depression
(Drevets et al., 2008), and previous studies suggest that
both may be involved in pain chronification (Metz et al.,
2009; Ji and Neugebauer, 2011; Baliki et al., 2012; Hashmi
et al., 2013). Given our white matter and functional con-
nectivity network findings, we also examined the volumes
of nucleus accumbens (a main component of the dorsal
medial PFC–amygdala–nucleus accumbens module) and
thalamus, as the latter is a primary relay for nociceptive
transmission to the cortex through the spinothalamocorti-
cal projections. SBPp exhibited $10% smaller amygdala
(Fig. 3C) and hippocampus (Fig. 3D) total volumes (left + -
right), as compared to SBPr and healthy controls
(Supplementary Table 7). This effect was sustained over 3
years with strong correlations for volumes between study
entry to Week 156 for all SBP, for the amygdala and
hippocampus. These results were replicated using
Freesurfer as an alternative segmentation tool for extracting
subcortical volumes (Supplementary Fig. 11). However, no
significant group differences were observed for volumes of
the nucleus accumbens or the thalamus. Exploratory ana-
lyses further revealed that smaller amygdala and hippocam-
pus volumes were accompanied by thinning of mainly the
right amygdala shape in SBPp at all visits (Fig. 3G). We
previously have shown that the hippocampus volume is
smaller in patients with chronic pain (Mutso et al.,
2012). Here we replicate the finding in two new patient
groups suffering from longstanding chronic back pain and
osteoarthritis and show reduced volumes for both amyg-
dala and hippocampus. More importantly comparison be-
tween these groups and SBPp shows that extent of volume
decrease is similar across chronic pain patients and SBPp at
Week 0 (Fig. 3E and F). Given that both amygdala and
hippocampus volumes distinguished between SBP groups,
their morphologies did not change over 3 years, and smal-
ler amygdala and hippocampus volumes were comparable
between SBPp and multiple chronic pain conditions, these
volumetric measures (derived and validated using two un-
biased measurements, which are replicated in many labs in
more than 30 000 subjects; Hibar et al., 2015) most likely
reflect pre-existing circuit abnormalities that induce risk for
chronic pain, across multiple chronic pain conditions.
Genetic associations with limbic risk
factors
Data from twin studies show that chronic back pain is 20–
67% heritable (Ferreira et al., 2013). Given the recent
notion that genetic variants of some pathologies are more
penetrant at the level of brain measures (Rose and
Donohoe, 2013), we explored the hypothesis that gene vari-
ations can be linked to corticolimbic risk factors for chronic
pain. We used an imaging–genetics approach to examine
associations between SNPs and corticolimbic brain risk fac-
tors. In total, 30 candidate SNPs located in 12 different
Anatomical risk factors for chronic pain BRAIN 2016: Page 7 of 13 | 7
byguestonMay9,2016http://brain.oxfordjournals.org/Downloadedfrom
genes were selected for their known involvement in limbic
functions and also to pain or nociception (Supplementary
Tables 9–11). Genotypes were compared with regard to
amygdala and hippocampus volumes, and dorsal medial
PFC–amygdala–nucleus accumbens white matter and func-
tional connectivity network connectivity for Week 0 values.
Then, we secondarily tested if these genes could also
differentiate between SBP groups as identified at Week 56.
After correcting for multiple comparisons, the opioid recep-
tor delta 1 (OPRD1) rs678849 SNP was associated with
amygdala volume, and opioid receptor mu 1 (OPRM1)
rs1799971 SNP was associated with incidence of connec-
tions within the dorsal medial PFC–amygdala–nucleus
accumbens white matter network. Moreover, OPRD1
Figure 2 Intra-corticolimbic white matter connectivity identifies three modules, only the dorsal medial PFC–amygdala–nu-
cleus accumbens module imparts risk for chronic pain. (A) Display of the intra-corticolimbic white matter-based structural networks
generated from 1068 nodes (white matter networks) at 0.1 link density. (B) Modularity analysis performed on this network separated the system
into three communities: (i) dorsal medial PFC–amygdala–nucleus accumbens (mPFCdorsal-amy-NAc); (ii) ventral medial PFC–amygdala
(mPFCventral-Amy); and (iii) orbitofrontal cortex–amygdala–hippocampus (OFC-amy-hipp). (C) Normalized mutual information was used to
quantify similarity between individual subject community structure and the mean group community structure presented in (B) SBPp, SBPr and
healthy controls displayed similar modular organization across all time points, due to absence of group differences [F(2,69) = 2.34; P = 0.10
repeated measure ANOVA] or a time  group interaction [F(5.73, 200.65) = 0.72; P = 0.63 repeated measure ANOVA]. (D) Structural con-
nection probability for SBPp, SBPr and healthy within the dorsal medial PFC–amygdala–nucleus accumbens module calculated across link densities
ranging between 0.02 and 0.2. Incidence of white matter connections was higher in SBPp at all densities [F(2,78) = 6.10; P = 0.003 repeated
measure ANOVA]. The histogram displays group differences at 0.1 link density. (E) Per cent structural connections for SBPp and SBPr and healthy
controls over 3 years at 0.1 link density. Incidence of connections in the dorsal medial PFC–amygdala–nucleus accumbens module was higher in
SBPp [F(2,69) = 4.74; P = 0.012 repeated measure ANOVA] for visits 0–56 weeks. Concordant results were seen at Week 156 [t(1,34) = 1.79;
P = 0.08 two-sided unpaired t-tests]. Scatterplot shows 3-year stability of dorsal medial PFC–amygdala–nucleus accumbens module connectivity
for all SBP. (F) Incidence of functional connections in the dorsal medial PFC–amygdala–nucleus accumbens module was higher in SBPp across 0.02
to 0.2 link densities [F(1,60) = 4.00; P = 0.05]. The histogram displays group differences at 0.1 link density. (G) SBPp showed higher incidence of
functional connections in the dorsal medial PFC–amygdala–nucleus accumbens module [F(1,60) = 8.00; P = 0.006 repeated measure ANOVA], for
visits 0–56 weeks at 0.1 link density. This functional connectivity, however, was not different between SBPp and SBPr subsamples at Week 156, and
the scatter plot showed no within subject relationship between Weeks 0 and 156 functional connections. *
P 5 0.05 post hoc comparisons between
SBPp and SBPp. 
P = 0.08. Number of subjects is shown in parentheses. Error bars indicate standard error of the mean (SEM).
8 | BRAIN 2016: Page 8 of 13 E. Vachon-Presseau et al.
byguestonMay9,2016http://brain.oxfordjournals.org/Downloadedfrom
rs678849 SNP was also related to SBP groups at Week 56
(Supplementary Fig. 12 and Supplementary Table 12).
These findings provide suggestive evidence that the
OPRD1 polymorphism is a genetic candidate for mediating
corticolimbic-based risk of chronic pain.
Modelling risk and symptom severity
for chronic pain
We performed path analysis to unravel the relationships
between the identified gene, pain at time of entry into the
study, and corticolimbic risk factors for predicting chronic
pain (Fig. 4). The three primary identified risk factors, each
of which predicted SBPp/r groupings at 1 year based on
measures collected at study entry, explained 60% of the
variance for pain chronification (the explained variance
would be closer to 90% if only right amygdala volume
was used in the model, yet the model using bilateral amyg-
dala volume should be more robust as this measure is
replicated using different segmentation tools), and the
gene–chronic pain relationship was fully mediated through
the indirect effect of the amygdala volume, accounting for
4% of the total variance (Fig. 4A).
Although back pain intensity measured at entry in the
study did not influence risk factor for chronic pain, it
was the only parameter of the risk factors that explained
30% and 38% of the variance in pain intensity at 1 year
and at 3 years, respectively (Fig. 4B). Restricting the regres-
sion model only to SBPp subjects, back pain at the time of
entry into the study was the only parameter that explained
76% and 51% of the variance of back pain at 1 year and 3
years, respectively. These results imply that although the
injury or inciting event-related back pain at time of entry
into the study does not influence SBP groupings, initial
Figure 3 Amygdala and hippocampus volumes are risk factors for chronic pain. (A and B) Subcortical structures were segmented,
corresponding volumes summed across hemispheres, and compared between groups as a function of time (Supplementary Tables 4 and 5). Heat maps
display overlap of automated segmentation across SBPp, SBPr, and healthy controls at week 0. (C and D) Repeated measure ANCOVAs (group and
time effects; covariate for age, gender, and grey matter volume) revealed that SBPp showed smaller amygdala [F(2,63) = 7.94; P 5 0.001] and
hippocampus [F(2,70) = 6.35; P = 0.003], but no main effect of time (P-values 4 0.69) or group by time interaction (P-values4 0.23). Bar graphs
illustrate group differences at 3 years. Scatterplots show volume reproducibility over 3 years across all SBP. *
P 5 0.05 SBPp versus SBPr, and SBPp
versus healthy controls. (E and F) Age-matched chronic back pain patients showed significant smaller amygdala [t(1,42) = 2.71; P = 0.009 two-sided
unpaired t-tests] and hippocampus [t(1,43) = 1.99; P = 0.05 two-sided unpaired t-tests] than SBPr, matching the volumes observed in SBPp. Similarly,
osteoarthritis patients displayed at least marginally smaller amygdala [t(1,52) = 2.21; P = 0.03 two-sided unpaired t-tests] and hippocampus
[t(1,52) = 1.95; P = 0.057 two-sided unpaired t-tests] when compared to age matched healthy controls. *
P 5 0.05, 
P 5 0.06. (G) Vertex-wise shape
analyses of the amygdala and the hippocampus indicated that SBPp had thinner right amygdala across all visits. Number of subjects is shown in
parentheses. Error bars indicate SEM.
Anatomical risk factors for chronic pain BRAIN 2016: Page 9 of 13 | 9
byguestonMay9,2016http://brain.oxfordjournals.org/Downloadedfrom
back pain intensity has a large influence on symptom se-
verity, especially in subjects where the pain persists.
Influence of negative affect on risk
for chronic pain
Epidemiological studies indicate high prevalence of depres-
sion and negative affect in chronic pain patients, and pro-
pose these conditions reciprocally aggravate each other
(Gerrits et al., 2015). Our data showed that neither nega-
tive affect nor depression changed over 3 years in SBP
(Supplementary Table 1), which at least in this cohort dis-
counts the influence of the development of chronic pain on
negative moods. The converse influence of mood on the
development of chronic pain remains a possibility. We ad-
dressed this issue by testing whether depression, and nega-
tive affect, influence future pain outcome through the
mediating effects of uncovered corticolimbic risk factors,
using path analysis. We observed that the density of con-
nections within dorsal medial PFC–amygdala–nucleus
accumbens white matter network mediated an indirect
effect of depression on transition to chronic pain (account-
ing for 5% of total variance), with additional independent
contributions of the volume of bilateral total amygdala
volume and connections within dorsal medial PFC–
amygdala–nucleus accumbens functional connectivity net-
work (Supplementary Fig. 13). A similar mediation path
was observed for negative affect (Supplementary Fig. 13).
It is noteworthy that SBP were only mildly depressed, yet
their negative mood influenced pain chronification through
white matter connectivity of the medial dorsal PFC–amyg-
dala–nucleus accumbens module. Maladaptive genotype-
driven neural responsiveness may pave the way to develop-
ment of chronic pain. As the dorsal medial PFC–amygdala–
nucleus accumbens white matter network path is also influ-
enced by the OPRM1 rs1799971, we here uncover a
brain–gene relationship for the influence of mood on devel-
opment of chronic pain.
Discussion
The present study tested the role of the corticolimbic system
in development of chronic back pain. Earlier analyses of
subsamples of these data (Baliki et al., 2012; Mansour
et al., 2013) show that functional and white matter proper-
ties of some limbic regions impart risk for chronic pain. Yet
identification of these factors was based on whole-brain
contrasts. Here we used a completely unbiased approach,
with around double the number of subjects, and tracked
properties of identified risk factors for 3 years. Whole-brain
multi-resolution white matter network comparisons at
study entry indicated that structural connections predispos-
ing the development of chronic pain at 1 year were located
outside pain-related regions but within the corticolimbic
brain. Higher-resolution examination of the corticolimbic
white matter network identified three separate modules,
where only the dorsal medial PFC–amygdala–nucleus
accumbens module differentiated between SBP groups
based on both white matter and functional connectivity.
The white matter network connectivity of dorsal medial
PFC–amygdala–nucleus accumbens remained constant
over 3 years, but its functional network connectivity dissi-
pated by 3 years relative to values at time 0. Both amyg-
dala and hippocampus volumes differentiated between SBP
groups, remained invariant over 3 years, and robustly dis-
tinguished between SBPp and SBPr.
The present results identify three segregated white matter
network connectivity-based corticolimbic circuits, with only
the dorsal medial PFC–amygdala–nucleus accumbens net-
work contributing to risk for chronic pain. What are the
functional properties of this module? The amygdala subdiv-
isions of each mesolimbic module show correspondence
with its function-based and cytoarchitecture-based parcella-
tion (Bzdok et al., 2013). The portion of the amygdala
incorporated within dorsal medial PFC–amygdala–nucleus
accumbens network matched best to the basolateral nu-
cleus, which functionally shows preferential involvement
in sensory input processing (Bzdok et al., 2013). On the
other hand, the corticolimbic circuits also subdivided the
prefrontal cortex into dorsal, ventral and orbital subdiv-
isions. These divisions may be viewed equivalent to the
Figure 4 Corticolimbic characteristics determine risk for
chronic pain but symptom severity depends only on initial
pain intensity. (A) Path analysis was used to identify the contri-
bution of rs678849 SNP, pain at Week 0, and brain parameters on
risk for developing chronic pain (SBPp/r at Week 56). The white
matter network (White matter) and functional connectivity net-
work (Functional) connections of dorsal medial PFC–amygdala–nu-
cleus accumbens (mPFCdorsal-amy-NAc) module and bilateral total
amygdala volume, at Week 0, were independent predictors of pain
persistence or recovery (SBPp/r) at Week 56 (all P-values 4 0.01).
The relationship between OPRD1 rs678849 and pain persistence/
recovery was fully mediated through the bilateral amygdala volume
(indirect pathway 95% confidence interval: À0.021 and À0.173;
R2
= 0.04 of unique variance). Covariance coverage within and be-
tween variables exceeded 88%. Numbers of subjects are shown in
parentheses. (B) Scatterplots show that pain intensity at Week 0
significantly correlated with pain intensity and pain disability at
Week 156 (P-values 4 0.001).
10 | BRAIN 2016: Page 10 of 13 E. Vachon-Presseau et al.
byguestonMay9,2016http://brain.oxfordjournals.org/Downloadedfrom
rodent medial frontal cortex with respective correspond-
ences with pre-limbic, infra-limbic, and orbital cortices.
Within this perspective, the cortical component of the
dorsal medial PFC–amygdala–nucleus accumbens network
becomes analogous to the rodent pre-limbic cortex. In the
rodent, pre-limbic is necessary for expression of acquired
fear but not for innate fear (Corcoran and Quirk, 2007),
which parallels the evidence that human medial PFC repre-
sents intensity of chronic but not acute back pain (Hashmi
et al., 2013). In rodent models of persistent pain, pre-limbic
pyramidal neurons undergo rapid morphological changes
(Metz et al., 2009), and amygdala inputs shift the balance
between inhibitory and excitatory synaptic transmission in
pre-limbic neurons (Ji et al., 2010). Three recent independ-
ent studies now also show that activation of pre-limbic
neurons diminishes or blocks persistent, but do not influ-
ence acute pain behaviour (Lee et al., 2015; Wang et al.,
2015; Zhang et al., 2015). One of these reports also dem-
onstrates that projections from pre-limbic to nucleus
accumbens mediate the relief from persistent pain (Lee
et al., 2015). Therefore, the mesolimbic pre-limbic circuitry
and the control of persistent pain by pre-limbic activity
suggest that the dorsal medial PFC–amygdala–nucleus
accumbens module should be considered its human
homologue.
We recently proposed that, from a behaviour selection
viewpoint, pain and negative emotions must be conceptua-
lized as a continuum of processes necessary for survival
(Baliki and Apkarian, 2015). Current results support the
idea by demonstrating that smaller amygdala and hippo-
campal volumes, parameters commonly associated with
negative affective disorders (Gilbertson et al., 2002), are
also predictive of pain chronification. In SBP, amygdala
and hippocampus volumes remained invariant over 3
years, and the reduced volumes seen in SBPp matched
those observed in patients with chronic pain. Thus, we
can generalize these results to chronic negative mood dis-
orders where we presume the observed decreased volumes
most likely also reflect predispositions. Recent evidence in-
dicates that amygdala and hippocampus volumes impact
emotional learning and sociability abilities. Healthy sub-
jects with larger amygdala volume perform better in fear
acquisition (Winkelmann et al., 2015), while subjects with
larger hippocampus show higher fear contingency aware-
ness (Cacciaglia et al., 2015) and more accurate discrimin-
ation of contextual fear (Pohlack et al., 2012). These limbic
volume-related differential response characteristics may,
therefore, represent personality traits predisposing subjects
to both chronic pain and negative mood disorders.
We observed that the OPRD1 rs678849 SNP mediated
through amygdala volume risk for chronic pain. Earlier
studies associate this gene with cocaine, opiate, and
heroin addiction, as well as with smaller regional brain
volume (http://www.snpedia.com/index.php/Rs678849).
This genetic evidence is consistent and complimentary to
the role of dorsal medial PFC–amygdala–nucleus accum-
bens module in determining risk for chronic pain, as the
latter module is also commonly implicated in addictive
pathologies. Collectively these observations argue for
chronic pain being a maladaptive response, contingent on
corticolimbic properties that render the brain addicted to
pain. We, however, emphasize the important caveat that
the gene–brain–behaviour associations illustrated here are
based on sampling few SNPs in a small number of subjects.
They must be viewed as proof-of-concept exemplars, and
larger and multi-centred collaborative gene–brain studies
are necessary to unravel genes critical in the transition to
chronic pain.
In conclusion, these results challenge long-standing views
of the science of pain. Traditionally, the field has empha-
sized the inciting injury and related nociceptive spinal pro-
cesses as primary culprits of pain chronification. Instead,
here we establish that the gross anatomical properties of
the corticolimbic brain, not the initial back pain, determine
most of the risk for developing chronic pain. As the ana-
tomical risk factors were stable across 3 years, they were
presumably hardwired and present prior to the event initi-
ating back pain. These results pave the way for the devel-
opment of novel and distinct approaches to prevention and
treatment of chronic pain.
Acknowledgements
We thank all patients and volunteers that participated in
the study, and members of Apkarian lab for technical sup-
port and helpful suggestions.
Funding
This work was supported by funds from the National
Institute of Neurological Disorders and Stroke
(NS035115, A.V.A.). E.V.P. and P.T. were funded from
Canadian Institutes of Health Research (CIHR), E.C. was
funded from Swedish Council for Working Life and Social
Research (FAS: 2011-0627) and Uppsala University.
M.N.B. was partially funded by an anonymous foundation.
Supplementary material
Supplementary material is available at Brain online.
References
Achard S, Delon-Martin C, Vertes PE, Renard F, Schenck M,
Schneider F, et al. Hubs of brain functional networks are radically
reorganized in comatose patients. Proc Natl Acad Sci USA 2012;
109: 20608–13.
Apkarian AV, Sosa Y, Sonty S, Levy RE, Harden RN, Parrish TB,
et al. Chronic back pain is associated with decreased prefrontal and
thalamic gray matter density. J Neurosci 2004; 24: 10410–5.
Baliki MN, Apkarian AV. Nociception, pain, negative moods, and
behavior selection. Neuron 2015; 87: 474–91.
Anatomical risk factors for chronic pain BRAIN 2016: Page 11 of 13 | 11
byguestonMay9,2016http://brain.oxfordjournals.org/Downloadedfrom
Baliki MN, Chang PC, Baria AT, Centeno MV, Apkarian AV.
Resting-sate functional reorganization of the rat limbic system fol-
lowing neuropathic injury. Sci Rep 2014; 4: 6186.
Baliki MN, Chialvo DR, Geha PY, Levy RM, Harden RN, Parrish TB,
et al. Chronic pain and the emotional brain: specific brain activity
associated with spontaneous fluctuations of intensity of chronic back
pain. J Neurosci 2006; 26: 12165–73.
Baliki MN, Geha PY, Fields HL, Apkarian AV. Predicting value of
pain and analgesia: nucleus accumbens response to noxious stimuli
changes in the presence of chronic pain. Neuron 2010; 66: 149–60.
Baliki MN, Petre B, Torbey S, Herrmann KM, Huang L, Schnitzer TJ,
et al. Corticostriatal functional connectivity predicts transition to
chronic back pain. Nat Neurosci 2012; 15: 117–19.
Beck AT, Ward CH, Mendelson M, Mock J, Erbaugh J. An inventory
for measuring depression. Arch Gen Psychiatry 1961; 4: 561–71.
Behrens TE, Berg HJ, Jbabdi S, Rushworth MF, Woolrich MW.
Probabilistic diffusion tractography with multiple fibre orientations:
what can we gain? Neuroimage 2007; 34: 144–55.
Behrens TE, Woolrich MW, Jenkinson M, Johansen-Berg H, Nunes
RG, Clare S, et al. Characterization and propagation of uncertainty
in diffusion-weighted MR imaging. Magn Reson Med 2003; 50:
1077–88.
Bzdok D, Laird AR, Zilles K, Fox PT, Eickhoff SB. An investigation of
the structural, connectional, and functional subspecialization in the
human amygdala. Hum Brain Mapp 2013; 34: 3247–66.
Cacciaglia R, Pohlack ST, Flor H, Nees F. Dissociable roles for hip-
pocampal and amygdalar volume in human fear conditioning. Brain
Struct Funct 2015; 220: 2575–86.
Chang PC, Pollema-Mays SL, Centeno MV, Procissi D, Contini M,
Baria AT, et al. Role of nucleus accumbens in neuropathic pain:
linked multi-scale evidence in the rat transitioning to neuropathic
pain. Pain 2014; 155: 1128–39.
Corcoran KA, Quirk GJ. Activity in prelimbic cortex is necessary for
the expression of learned, but not innate, fears. J Neurosci 2007; 27:
840–4.
Drevets WC, Price JL, Furey ML. Brain structural and functional
abnormalities in mood disorders: implications for neurocircuitry
models of depression. Brain Struct Funct 2008; 213: 93–118.
Ferreira PH, Beckenkamp P, Maher CG, Hopper JL, Ferreira ML.
Nature or nurture in low back pain? Results of a systematic
review of studies based on twin samples. Eur J Pain 2013; 17:
957–71.
Fischl B, Salat DH, Busa E, Albert M, Dieterich M, Haselgrove C,
et al. Whole brain segmentation: automated labeling of neuroana-
tomical structures in the human brain. Neuron 2002; 33: 341–55.
Geha PY, Baliki MN, Harden RN, Bauer WR, Parrish TB, Apkarian
AV. The brain in chronic CRPS pain: abnormal gray-white matter
interactions in emotional and autonomic regions. Neuron 2008; 60:
570–81.
Gerrits MM, van Marwijk HW, van Oppen P, van der Horst H,
Penninx BW. Longitudinal association between pain, and depression
and anxiety over four years. J Psychosom Res 2015; 78: 64–70.
Gilbertson MW, Shenton ME, Ciszewski A, Kasai K, Lasko NB, Orr
SP, et al. Smaller hippocampal volume predicts pathologic vulner-
ability to psychological trauma. Nat Neurosci 2002; 5: 1242–7.
Hashmi JA, Baliki MN, Huang L, Baria AT, Torbey S, Hermann KM,
et al. Shape shifting pain: chronification of back pain shifts brain
representation from nociceptive to emotional circuits. Brain 2013;
136(Pt 9): 2751–68.
Hibar DP, Stein JL, Renteria ME, Arias-Vasquez A, Desrivieres S,
Jahanshad N, et al. Common genetic variants influence human sub-
cortical brain structures. Nature 2015; 520: 224–9.
Jenkinson M, Bannister P, Brady M, Smith S. Improved optimization
for the robust and accurate linear registration and motion correction
of brain images. Neuroimage 2002; 17: 825–41.
Ji G, Neugebauer V. Pain-related deactivation of medial prefrontal
cortical neurons involves mGluR1 and GABA(A) receptors. J
Neurophysiol 2011; 106: 2642–52.
Ji G, Sun H, Fu Y, Li Z, Pais-Vieira M, Galhardo V, et al. Cognitive
impairment in pain through amygdala-driven prefrontal cortical de-
activation. J Neurosci 2010; 30: 5451–64.
Lee M, Manders TR, Eberle SE, Su C, D’Amour J, Yang R, et al.
Activation of corticostriatal circuitry relieves chronic neuropathic
pain. J Neurosci 2015; 35: 5247–59.
Mackinnon DP, Lockwood CM, Williams J. Confidence limits for the
indirect effect: distribution of the product and resampling methods.
Multivariate Behav Res 2004; 39: 99.
MacLean PD. The limbic system (“visceral brain”) in relation to the
central gray and reticulum of the brain stem.Psychosom Med 1955;
17: 355–66.
Mansour AR, Baliki MN, Huang L, Torbey S, Herrmann KM,
Schnitzer TJ, et al. Brain white matter structural properties predict
transition to chronic pain. Pain 2013; 154: 2160–8.
Medicine Io. Relieving pain in America: a blueprint for transforming
prevention, care, education, and research. 2011. Available from:
http://www.iom.edu.
Melzack R. The McGill Pain Questionnaire: major properties and
scoring methods. Pain 1975; 1: 277–99.
Melzack R, Casey K. Sensory, motivational, and central contol deter-
minants of pain. The skin senses. Springfield: Charles C. Thomas;
1968. p. 423–43.
Melzack R, Wall PD. Pain mechanisms: a new theory. Science 1965;
150: 971–9.
Metz AE, Yau HJ, Centeno MV, Apkarian AV, Martina M.
Morphological and functional reorganization of rat medial pre-
frontal cortex in neuropathic pain. Proc Natl Acad Sci USA 2009;
106: 2423–8.
Murray CJ, Lopez AD. Measuring the global burden of disease. N
Engl J Med 2013; 369: 448–57.
Mutso AA, Petre B, Huang L, Baliki MN, Torbey S, Herrmann K,
et al. Reorganization of hippocampal functional connectivity with
transition to chronic back pain. J Neurophysiol 2014; 111:
1065–76.
Mutso AA, Radzicki D, Baliki MN, Huang L, Banisadr G, Centeno
MV, et al. Abnormalities in hippocampal functioning with persistent
pain. J Neurosci 2012; 32: 5747–56.
Neugebauer V, Li W, Bird GC, Bhave G, Gereau RW. Synaptic plas-
ticity in the amygdala in a model of arthritic pain: differential roles
of metabotropic glutamate receptors 1 and 5. J Neurosci 2003; 23:
52–63.
Nugent AC, Luckenbaugh DA, Wood SE, Bogers W, Zarate CA, Jr.,
Drevets WC. Automated subcortical segmentation using FIRST: test-
retest reliability, interscanner reliability, and comparison to manual
segmentation. Hum Brain Mapp 2013; 34: 2313–29.
Patenaude B, Smith SM, Kennedy DN, Jenkinson M. A Bayesian
model of shape and appearance for subcortical brain segmentation.
Neuroimage 2011; 56: 907–22.
Petre B, Torbey S, Griffith JW, De Oliveira G, Herrmann K, Mansour
A, et al. Smoking increases risk of pain chronification through
shared corticostriatal circuitry. Hum Brain Mapp 2015; 36: 683–94.
Phelps EA, LeDoux JE. Contributions of the amygdala to emotion
processing: from animal models to human behavior. Neuron 2005;
48: 175–87.
Pohlack ST, Nees F, Liebscher C, Cacciaglia R, Diener SJ, Ridder S,
et al. Hippocampal but not amygdalar volume affects contextual
fear conditioning in humans. Hum Brain Mapp 2012; 33: 478–88.
Raz M. The painless brain: lobotomy, psychiatry, and the treatment of
chronic pain and terminal illness. Perspect Biol Med 2009; 52:
555–65.
Ren WJ, Liu Y, Zhou LJ, Li W, Zhong Y, Pang RP, et al. Peripheral
nerve injury leads to working memory deficits and dysfunction of
the hippocampus by upregulation of TNF-alpha in rodents.
Neuropsychopharmacology 2011; 36: 979–92.
Rose EJ, Donohoe G. Brain vs behavior: an effect size comparison of
neuroimaging and cognitive studies of genetic risk for schizophrenia.
Schizophr Bull 2013; 39: 518–26.
12 | BRAIN 2016: Page 12 of 13 E. Vachon-Presseau et al.
byguestonMay9,2016http://brain.oxfordjournals.org/Downloadedfrom
Rubinov M, Sporns O. Complex network measures of brain connect-
ivity: uses and interpretations. Neuroimage 2010; 52: 1059–69.
Russo SJ, Murrough JW, Han MH, Charney DS, Nestler EJ.
Neurobiology of resilience. Nat Neurosci 2012; 15: 1475–84.
Schwartz N, Temkin P, Jurado S, Lim BK, Heifets BD, Polepalli JS,
et al. Chronic pain. Decreased motivation during chronic pain re-
quires long-term depression in the nucleus accumbens. Science 2014;
345: 535–42.
Smith SM, Zhang Y, Jenkinson M, Chen J, Matthews PM, Federico A,
et al. Accurate, robust, and automated longitudinal and cross-sec-
tional brain change analysis. Neuroimage 2002; 17: 479–89.
Tait RC, Chibnall JT, Krause S. The pain disability index: psychomet-
ric properties. Pain 1990; 40: 171–82.
Vachon-Presseau E, Roy M, Martel MO, Caron E, Marin MF, Chen J,
et al. The stress model of chronic pain: evidence from basal cortisol
and hippocampal structure and function in humans. Brain 2013;
136(Pt 3): 815–27.
Wang GQ, Cen C, Li C, Cao S, Wang N, Zhou Z, et al.
Deactivation of excitatory neurons in the prelimbic cortex via
Cdk5 promotes pain sensation and anxiety. Nat Commun
2015; 6: 7660.
Watson D, Clark LA. The PANAS-X manual for the positive and
negative affect schedule–Expanded form. University of Iowa.
1999.
Winkelmann T, Grimm O, Pohlack ST, Nees F, Cacciaglia R, Dinu-
Biringer R, et al. Brain morphology correlates of interindividual
differences in conditioned fear acquisition and extinction learning.
Brain Struct Funct 2015, in press.
Zalesky A, Fornito A, Bullmore ET. Network-based statistic: iden-
tifying differences in brain networks. Neuroimage 2010; 53:
1197–207.
Zhang Z, Gadotti VM, Chen L, Souza IA, Stemkowski PL, Zamponi
GW. Role of prelimbic GABAergic circuits in sensory and emotional
aspects of neuropathic pain. Cell Rep 2015; 12: 752–9.
Anatomical risk factors for chronic pain BRAIN 2016: Page 13 of 13 | 13
byguestonMay9,2016http://brain.oxfordjournals.org/Downloadedfrom

More Related Content

What's hot

Predicting transition to chronic pain.
Predicting transition to chronic pain.Predicting transition to chronic pain.
Predicting transition to chronic pain.
Paul Coelho, MD
 
Opioid Induced Hyperalgesia
Opioid Induced HyperalgesiaOpioid Induced Hyperalgesia
Opioid Induced Hyperalgesia
Ade Wijaya
 
Dolore pelvico cronico: epidemiologia ed eziopatogenesi
Dolore pelvico cronico: epidemiologia ed eziopatogenesiDolore pelvico cronico: epidemiologia ed eziopatogenesi
Dolore pelvico cronico: epidemiologia ed eziopatogenesi
GLUP2010
 
Pain, Heat, & Emotion. NEJM
Pain, Heat, & Emotion. NEJMPain, Heat, & Emotion. NEJM
Pain, Heat, & Emotion. NEJM
Paul Coelho, MD
 
E-book Dissertatie Coen Itz
E-book Dissertatie Coen ItzE-book Dissertatie Coen Itz
E-book Dissertatie Coen Itz
Coen Itz
 
Chronic Pain After Surgery
Chronic Pain After SurgeryChronic Pain After Surgery
Chronic Pain After Surgery
Ade Wijaya
 
2016 Brandeis Student Conference POSTER_Kilcoyne_FINAL (3)
2016 Brandeis Student Conference POSTER_Kilcoyne_FINAL (3)2016 Brandeis Student Conference POSTER_Kilcoyne_FINAL (3)
2016 Brandeis Student Conference POSTER_Kilcoyne_FINAL (3)
Hannah Kilcoyne
 
Widerstrom noga talk 7-12_11
Widerstrom noga talk 7-12_11Widerstrom noga talk 7-12_11
Widerstrom noga talk 7-12_11
National Neurotrauma Symposium
 
Tapering Long Term Opioid Therapy in Chronic Noncancer Pain
Tapering Long Term Opioid Therapy in Chronic Noncancer PainTapering Long Term Opioid Therapy in Chronic Noncancer Pain
Tapering Long Term Opioid Therapy in Chronic Noncancer Pain
Ade Wijaya
 
Medical shockwaves for chronic low back pain - a case series
Medical shockwaves for chronic low back pain - a case seriesMedical shockwaves for chronic low back pain - a case series
Medical shockwaves for chronic low back pain - a case series
Kenneth Craig
 
The use of pulsed radiofrequency for the treatment of pudendal neuralgia a c...
The use of pulsed radiofrequency for the treatment of pudendal neuralgia  a c...The use of pulsed radiofrequency for the treatment of pudendal neuralgia  a c...
The use of pulsed radiofrequency for the treatment of pudendal neuralgia a c...
Jason Attaman
 
Treatment Chronic Pain [Autosaved].pptx
Treatment Chronic Pain [Autosaved].pptxTreatment Chronic Pain [Autosaved].pptx
Treatment Chronic Pain [Autosaved].pptx
putusukedana1
 
Postherpetic Neuralgia.pptx
Postherpetic Neuralgia.pptxPostherpetic Neuralgia.pptx
Postherpetic Neuralgia.pptx
putusukedana1
 
The neuroanatomy, neurophysiology, and neurochemistry of pain, stress, and an...
The neuroanatomy, neurophysiology, and neurochemistry of pain, stress, and an...The neuroanatomy, neurophysiology, and neurochemistry of pain, stress, and an...
The neuroanatomy, neurophysiology, and neurochemistry of pain, stress, and an...
South Dakota Pain Capable Unborn Child Protection Act
 
Frozen Shoulder.pdf
Frozen Shoulder.pdfFrozen Shoulder.pdf
Frozen Shoulder.pdf
putusukedana1
 
Haptic medicine - sustainable, accessible, low-cost
Haptic medicine - sustainable, accessible, low-costHaptic medicine - sustainable, accessible, low-cost
Haptic medicine - sustainable, accessible, low-cost
0neW0rldT0gether
 
Emerging concepts in pain management
Emerging concepts in pain managementEmerging concepts in pain management
Emerging concepts in pain management
Vikram Kumar
 
Still’s Disease and Recurrent Complex Regional Pain Syndrome Type-I: The Firs...
Still’s Disease and Recurrent Complex Regional Pain Syndrome Type-I: The Firs...Still’s Disease and Recurrent Complex Regional Pain Syndrome Type-I: The Firs...
Still’s Disease and Recurrent Complex Regional Pain Syndrome Type-I: The Firs...
Samantha Adcock
 
ANTIDEPRESSANTS IN CHRONIC PAIN RELIEF- A REVIEW
ANTIDEPRESSANTS IN CHRONIC PAIN RELIEF- A REVIEWANTIDEPRESSANTS IN CHRONIC PAIN RELIEF- A REVIEW
ANTIDEPRESSANTS IN CHRONIC PAIN RELIEF- A REVIEW
Dr. Raghavendra Kumar Gunda
 
CIPNDrRusso
CIPNDrRussoCIPNDrRusso
CIPNDrRusso
Dr Fred Russo
 

What's hot (20)

Predicting transition to chronic pain.
Predicting transition to chronic pain.Predicting transition to chronic pain.
Predicting transition to chronic pain.
 
Opioid Induced Hyperalgesia
Opioid Induced HyperalgesiaOpioid Induced Hyperalgesia
Opioid Induced Hyperalgesia
 
Dolore pelvico cronico: epidemiologia ed eziopatogenesi
Dolore pelvico cronico: epidemiologia ed eziopatogenesiDolore pelvico cronico: epidemiologia ed eziopatogenesi
Dolore pelvico cronico: epidemiologia ed eziopatogenesi
 
Pain, Heat, & Emotion. NEJM
Pain, Heat, & Emotion. NEJMPain, Heat, & Emotion. NEJM
Pain, Heat, & Emotion. NEJM
 
E-book Dissertatie Coen Itz
E-book Dissertatie Coen ItzE-book Dissertatie Coen Itz
E-book Dissertatie Coen Itz
 
Chronic Pain After Surgery
Chronic Pain After SurgeryChronic Pain After Surgery
Chronic Pain After Surgery
 
2016 Brandeis Student Conference POSTER_Kilcoyne_FINAL (3)
2016 Brandeis Student Conference POSTER_Kilcoyne_FINAL (3)2016 Brandeis Student Conference POSTER_Kilcoyne_FINAL (3)
2016 Brandeis Student Conference POSTER_Kilcoyne_FINAL (3)
 
Widerstrom noga talk 7-12_11
Widerstrom noga talk 7-12_11Widerstrom noga talk 7-12_11
Widerstrom noga talk 7-12_11
 
Tapering Long Term Opioid Therapy in Chronic Noncancer Pain
Tapering Long Term Opioid Therapy in Chronic Noncancer PainTapering Long Term Opioid Therapy in Chronic Noncancer Pain
Tapering Long Term Opioid Therapy in Chronic Noncancer Pain
 
Medical shockwaves for chronic low back pain - a case series
Medical shockwaves for chronic low back pain - a case seriesMedical shockwaves for chronic low back pain - a case series
Medical shockwaves for chronic low back pain - a case series
 
The use of pulsed radiofrequency for the treatment of pudendal neuralgia a c...
The use of pulsed radiofrequency for the treatment of pudendal neuralgia  a c...The use of pulsed radiofrequency for the treatment of pudendal neuralgia  a c...
The use of pulsed radiofrequency for the treatment of pudendal neuralgia a c...
 
Treatment Chronic Pain [Autosaved].pptx
Treatment Chronic Pain [Autosaved].pptxTreatment Chronic Pain [Autosaved].pptx
Treatment Chronic Pain [Autosaved].pptx
 
Postherpetic Neuralgia.pptx
Postherpetic Neuralgia.pptxPostherpetic Neuralgia.pptx
Postherpetic Neuralgia.pptx
 
The neuroanatomy, neurophysiology, and neurochemistry of pain, stress, and an...
The neuroanatomy, neurophysiology, and neurochemistry of pain, stress, and an...The neuroanatomy, neurophysiology, and neurochemistry of pain, stress, and an...
The neuroanatomy, neurophysiology, and neurochemistry of pain, stress, and an...
 
Frozen Shoulder.pdf
Frozen Shoulder.pdfFrozen Shoulder.pdf
Frozen Shoulder.pdf
 
Haptic medicine - sustainable, accessible, low-cost
Haptic medicine - sustainable, accessible, low-costHaptic medicine - sustainable, accessible, low-cost
Haptic medicine - sustainable, accessible, low-cost
 
Emerging concepts in pain management
Emerging concepts in pain managementEmerging concepts in pain management
Emerging concepts in pain management
 
Still’s Disease and Recurrent Complex Regional Pain Syndrome Type-I: The Firs...
Still’s Disease and Recurrent Complex Regional Pain Syndrome Type-I: The Firs...Still’s Disease and Recurrent Complex Regional Pain Syndrome Type-I: The Firs...
Still’s Disease and Recurrent Complex Regional Pain Syndrome Type-I: The Firs...
 
ANTIDEPRESSANTS IN CHRONIC PAIN RELIEF- A REVIEW
ANTIDEPRESSANTS IN CHRONIC PAIN RELIEF- A REVIEWANTIDEPRESSANTS IN CHRONIC PAIN RELIEF- A REVIEW
ANTIDEPRESSANTS IN CHRONIC PAIN RELIEF- A REVIEW
 
CIPNDrRusso
CIPNDrRussoCIPNDrRusso
CIPNDrRusso
 

Similar to Corticolimbic anatomic characteristics predetermine risk for chronic pain.

Abordaje de Factores Psicosociales en Educación Para la Salud en Fisioterapia
Abordaje de Factores Psicosociales en Educación Para la Salud en FisioterapiaAbordaje de Factores Psicosociales en Educación Para la Salud en Fisioterapia
Abordaje de Factores Psicosociales en Educación Para la Salud en Fisioterapia
Raúl Ferrer Peña PT MSc PhD Candidate
 
fnsys-09-00015
fnsys-09-00015fnsys-09-00015
fnsys-09-00015
John Caccaviello
 
fnsys-09-00015
fnsys-09-00015fnsys-09-00015
fnsys-09-00015
John Caccaviello
 
Sensory caracteristics
Sensory caracteristicsSensory caracteristics
Sensory caracteristics
Meziat
 
2015.01.26 ben kligler integrative pain2015
2015.01.26 ben kligler integrative pain20152015.01.26 ben kligler integrative pain2015
2015.01.26 ben kligler integrative pain2015
Cara Feldman-Hunt
 
Endogenous Opioid Dysregulation
Endogenous Opioid DysregulationEndogenous Opioid Dysregulation
Endogenous Opioid Dysregulation
Paul Coelho, MD
 
Central Sensitization In LBP
Central Sensitization In LBPCentral Sensitization In LBP
Central Sensitization In LBP
Paul Coelho, MD
 
Common Brain Mechanisms Between Pain & Addiction
Common Brain Mechanisms Between Pain & AddictionCommon Brain Mechanisms Between Pain & Addiction
Common Brain Mechanisms Between Pain & Addiction
Paul Coelho, MD
 
Psychological correlates of acute post surgical pain.
Psychological correlates of acute post surgical pain.Psychological correlates of acute post surgical pain.
Psychological correlates of acute post surgical pain.
Paul Coelho, MD
 
Presentatie Drs. Ronald Kan - Even wat rechtzetten
Presentatie Drs. Ronald Kan - Even wat rechtzetten Presentatie Drs. Ronald Kan - Even wat rechtzetten
Presentatie Drs. Ronald Kan - Even wat rechtzetten
NVMT-symposium
 
Austin Journal of Sleep Disorders
Austin Journal of Sleep Disorders Austin Journal of Sleep Disorders
Austin Journal of Sleep Disorders
Austin Publishing Group
 
Ian Persaud Conte Symposium 2016
Ian Persaud Conte Symposium 2016Ian Persaud Conte Symposium 2016
Ian Persaud Conte Symposium 2016
Ian Persaud
 
Recovery from chronic pain is possible
Recovery from chronic pain is possibleRecovery from chronic pain is possible
Recovery from chronic pain is possible
SIRPA Ltd
 
Modern neurosciapproachcs ppaper13
Modern neurosciapproachcs ppaper13Modern neurosciapproachcs ppaper13
Modern neurosciapproachcs ppaper13
Nathanael Amparo
 
How the brain heals emotional wounds the functional neuroanatomy of forgivene...
How the brain heals emotional wounds the functional neuroanatomy of forgivene...How the brain heals emotional wounds the functional neuroanatomy of forgivene...
How the brain heals emotional wounds the functional neuroanatomy of forgivene...
Elsa von Licy
 
BBB480FinalAcademiaPub
BBB480FinalAcademiaPubBBB480FinalAcademiaPub
BBB480FinalAcademiaPub
Timothy Judge
 
العقل البشري
العقل البشريالعقل البشري
العقل البشري
Dr. Ahmed El Watidy
 
No association between prepulse inhibition of the startle reflex and neuropsyc...
No association between prepulse inhibition of the startle reflex and neuropsyc...No association between prepulse inhibition of the startle reflex and neuropsyc...
No association between prepulse inhibition of the startle reflex and neuropsyc...
Benjamin Cortes
 
Cyriax research articles mi lb 11 4-14
Cyriax research articles mi lb 11 4-14Cyriax research articles mi lb 11 4-14
Cyriax research articles mi lb 11 4-14
Satoshi Kajiyama
 
PPT Presentation
PPT PresentationPPT Presentation
PPT Presentation
Dr K K Pandey
 

Similar to Corticolimbic anatomic characteristics predetermine risk for chronic pain. (20)

Abordaje de Factores Psicosociales en Educación Para la Salud en Fisioterapia
Abordaje de Factores Psicosociales en Educación Para la Salud en FisioterapiaAbordaje de Factores Psicosociales en Educación Para la Salud en Fisioterapia
Abordaje de Factores Psicosociales en Educación Para la Salud en Fisioterapia
 
fnsys-09-00015
fnsys-09-00015fnsys-09-00015
fnsys-09-00015
 
fnsys-09-00015
fnsys-09-00015fnsys-09-00015
fnsys-09-00015
 
Sensory caracteristics
Sensory caracteristicsSensory caracteristics
Sensory caracteristics
 
2015.01.26 ben kligler integrative pain2015
2015.01.26 ben kligler integrative pain20152015.01.26 ben kligler integrative pain2015
2015.01.26 ben kligler integrative pain2015
 
Endogenous Opioid Dysregulation
Endogenous Opioid DysregulationEndogenous Opioid Dysregulation
Endogenous Opioid Dysregulation
 
Central Sensitization In LBP
Central Sensitization In LBPCentral Sensitization In LBP
Central Sensitization In LBP
 
Common Brain Mechanisms Between Pain & Addiction
Common Brain Mechanisms Between Pain & AddictionCommon Brain Mechanisms Between Pain & Addiction
Common Brain Mechanisms Between Pain & Addiction
 
Psychological correlates of acute post surgical pain.
Psychological correlates of acute post surgical pain.Psychological correlates of acute post surgical pain.
Psychological correlates of acute post surgical pain.
 
Presentatie Drs. Ronald Kan - Even wat rechtzetten
Presentatie Drs. Ronald Kan - Even wat rechtzetten Presentatie Drs. Ronald Kan - Even wat rechtzetten
Presentatie Drs. Ronald Kan - Even wat rechtzetten
 
Austin Journal of Sleep Disorders
Austin Journal of Sleep Disorders Austin Journal of Sleep Disorders
Austin Journal of Sleep Disorders
 
Ian Persaud Conte Symposium 2016
Ian Persaud Conte Symposium 2016Ian Persaud Conte Symposium 2016
Ian Persaud Conte Symposium 2016
 
Recovery from chronic pain is possible
Recovery from chronic pain is possibleRecovery from chronic pain is possible
Recovery from chronic pain is possible
 
Modern neurosciapproachcs ppaper13
Modern neurosciapproachcs ppaper13Modern neurosciapproachcs ppaper13
Modern neurosciapproachcs ppaper13
 
How the brain heals emotional wounds the functional neuroanatomy of forgivene...
How the brain heals emotional wounds the functional neuroanatomy of forgivene...How the brain heals emotional wounds the functional neuroanatomy of forgivene...
How the brain heals emotional wounds the functional neuroanatomy of forgivene...
 
BBB480FinalAcademiaPub
BBB480FinalAcademiaPubBBB480FinalAcademiaPub
BBB480FinalAcademiaPub
 
العقل البشري
العقل البشريالعقل البشري
العقل البشري
 
No association between prepulse inhibition of the startle reflex and neuropsyc...
No association between prepulse inhibition of the startle reflex and neuropsyc...No association between prepulse inhibition of the startle reflex and neuropsyc...
No association between prepulse inhibition of the startle reflex and neuropsyc...
 
Cyriax research articles mi lb 11 4-14
Cyriax research articles mi lb 11 4-14Cyriax research articles mi lb 11 4-14
Cyriax research articles mi lb 11 4-14
 
PPT Presentation
PPT PresentationPPT Presentation
PPT Presentation
 

More from Paul Coelho, MD

Suicidality Poster References.docx
Suicidality Poster References.docxSuicidality Poster References.docx
Suicidality Poster References.docx
Paul Coelho, MD
 
Opioid Milli-equivalence Conversion Factors CDC
Opioid Milli-equivalence Conversion Factors CDCOpioid Milli-equivalence Conversion Factors CDC
Opioid Milli-equivalence Conversion Factors CDC
Paul Coelho, MD
 
Labeling Woefulness: The Social Construction of Fibromyalgia
Labeling Woefulness: The Social Construction of FibromyalgiaLabeling Woefulness: The Social Construction of Fibromyalgia
Labeling Woefulness: The Social Construction of Fibromyalgia
Paul Coelho, MD
 
Outcomes in Long-term Opioid Tapering and Buprenorphine Transition: A Retrosp...
Outcomes in Long-term Opioid Tapering and Buprenorphine Transition: A Retrosp...Outcomes in Long-term Opioid Tapering and Buprenorphine Transition: A Retrosp...
Outcomes in Long-term Opioid Tapering and Buprenorphine Transition: A Retrosp...
Paul Coelho, MD
 
Pain Phenotyping Tool For Primary Care
Pain Phenotyping Tool For Primary CarePain Phenotyping Tool For Primary Care
Pain Phenotyping Tool For Primary Care
Paul Coelho, MD
 
Fibromyalgia & Somatic Symptom Disorder Patient Handout
Fibromyalgia & Somatic Symptom Disorder Patient HandoutFibromyalgia & Somatic Symptom Disorder Patient Handout
Fibromyalgia & Somatic Symptom Disorder Patient Handout
Paul Coelho, MD
 
Community Catastrophizing
Community CatastrophizingCommunity Catastrophizing
Community Catastrophizing
Paul Coelho, MD
 
Risk-Benefit High-Dose LTOT
Risk-Benefit High-Dose LTOTRisk-Benefit High-Dose LTOT
Risk-Benefit High-Dose LTOT
Paul Coelho, MD
 
Space Trial
Space TrialSpace Trial
Space Trial
Paul Coelho, MD
 
Mortality quadrupled among opioid-driven hospitalizations notably within lowe...
Mortality quadrupled among opioid-driven hospitalizations notably within lowe...Mortality quadrupled among opioid-driven hospitalizations notably within lowe...
Mortality quadrupled among opioid-driven hospitalizations notably within lowe...
Paul Coelho, MD
 
Prescriptions filled following an opioid-related hospitalization.
Prescriptions filled following an opioid-related hospitalization.Prescriptions filled following an opioid-related hospitalization.
Prescriptions filled following an opioid-related hospitalization.
Paul Coelho, MD
 
Jamapsychiatry santavirta 2017_oi_170084
Jamapsychiatry santavirta 2017_oi_170084Jamapsychiatry santavirta 2017_oi_170084
Jamapsychiatry santavirta 2017_oi_170084
Paul Coelho, MD
 
Correlation of opioid mortality with prescriptions and social determinants -a...
Correlation of opioid mortality with prescriptions and social determinants -a...Correlation of opioid mortality with prescriptions and social determinants -a...
Correlation of opioid mortality with prescriptions and social determinants -a...
Paul Coelho, MD
 
Gao recommendations to CMS
Gao recommendations to CMSGao recommendations to CMS
Gao recommendations to CMS
Paul Coelho, MD
 
Prescribing by specialty
Prescribing by specialtyPrescribing by specialty
Prescribing by specialty
Paul Coelho, MD
 
The place-of-antipsychotics-in-the-therapy-of-anxiety-disorders-and-obsessive...
The place-of-antipsychotics-in-the-therapy-of-anxiety-disorders-and-obsessive...The place-of-antipsychotics-in-the-therapy-of-anxiety-disorders-and-obsessive...
The place-of-antipsychotics-in-the-therapy-of-anxiety-disorders-and-obsessive...
Paul Coelho, MD
 
Structured opioid refill clinic epic smartphrases
Structured opioid refill clinic epic smartphrases Structured opioid refill clinic epic smartphrases
Structured opioid refill clinic epic smartphrases
Paul Coelho, MD
 
Opioids for the Treatment of Chronic Pain: Mistakes Made, Lessons Learned, an...
Opioids for the Treatment of Chronic Pain: Mistakes Made, Lessons Learned, an...Opioids for the Treatment of Chronic Pain: Mistakes Made, Lessons Learned, an...
Opioids for the Treatment of Chronic Pain: Mistakes Made, Lessons Learned, an...
Paul Coelho, MD
 
The potential adverse influence of physicians’ words.
The potential adverse influence of physicians’ words.The potential adverse influence of physicians’ words.
The potential adverse influence of physicians’ words.
Paul Coelho, MD
 
Ctaf chronic pain__evidence_report_100417
Ctaf chronic pain__evidence_report_100417Ctaf chronic pain__evidence_report_100417
Ctaf chronic pain__evidence_report_100417
Paul Coelho, MD
 

More from Paul Coelho, MD (20)

Suicidality Poster References.docx
Suicidality Poster References.docxSuicidality Poster References.docx
Suicidality Poster References.docx
 
Opioid Milli-equivalence Conversion Factors CDC
Opioid Milli-equivalence Conversion Factors CDCOpioid Milli-equivalence Conversion Factors CDC
Opioid Milli-equivalence Conversion Factors CDC
 
Labeling Woefulness: The Social Construction of Fibromyalgia
Labeling Woefulness: The Social Construction of FibromyalgiaLabeling Woefulness: The Social Construction of Fibromyalgia
Labeling Woefulness: The Social Construction of Fibromyalgia
 
Outcomes in Long-term Opioid Tapering and Buprenorphine Transition: A Retrosp...
Outcomes in Long-term Opioid Tapering and Buprenorphine Transition: A Retrosp...Outcomes in Long-term Opioid Tapering and Buprenorphine Transition: A Retrosp...
Outcomes in Long-term Opioid Tapering and Buprenorphine Transition: A Retrosp...
 
Pain Phenotyping Tool For Primary Care
Pain Phenotyping Tool For Primary CarePain Phenotyping Tool For Primary Care
Pain Phenotyping Tool For Primary Care
 
Fibromyalgia & Somatic Symptom Disorder Patient Handout
Fibromyalgia & Somatic Symptom Disorder Patient HandoutFibromyalgia & Somatic Symptom Disorder Patient Handout
Fibromyalgia & Somatic Symptom Disorder Patient Handout
 
Community Catastrophizing
Community CatastrophizingCommunity Catastrophizing
Community Catastrophizing
 
Risk-Benefit High-Dose LTOT
Risk-Benefit High-Dose LTOTRisk-Benefit High-Dose LTOT
Risk-Benefit High-Dose LTOT
 
Space Trial
Space TrialSpace Trial
Space Trial
 
Mortality quadrupled among opioid-driven hospitalizations notably within lowe...
Mortality quadrupled among opioid-driven hospitalizations notably within lowe...Mortality quadrupled among opioid-driven hospitalizations notably within lowe...
Mortality quadrupled among opioid-driven hospitalizations notably within lowe...
 
Prescriptions filled following an opioid-related hospitalization.
Prescriptions filled following an opioid-related hospitalization.Prescriptions filled following an opioid-related hospitalization.
Prescriptions filled following an opioid-related hospitalization.
 
Jamapsychiatry santavirta 2017_oi_170084
Jamapsychiatry santavirta 2017_oi_170084Jamapsychiatry santavirta 2017_oi_170084
Jamapsychiatry santavirta 2017_oi_170084
 
Correlation of opioid mortality with prescriptions and social determinants -a...
Correlation of opioid mortality with prescriptions and social determinants -a...Correlation of opioid mortality with prescriptions and social determinants -a...
Correlation of opioid mortality with prescriptions and social determinants -a...
 
Gao recommendations to CMS
Gao recommendations to CMSGao recommendations to CMS
Gao recommendations to CMS
 
Prescribing by specialty
Prescribing by specialtyPrescribing by specialty
Prescribing by specialty
 
The place-of-antipsychotics-in-the-therapy-of-anxiety-disorders-and-obsessive...
The place-of-antipsychotics-in-the-therapy-of-anxiety-disorders-and-obsessive...The place-of-antipsychotics-in-the-therapy-of-anxiety-disorders-and-obsessive...
The place-of-antipsychotics-in-the-therapy-of-anxiety-disorders-and-obsessive...
 
Structured opioid refill clinic epic smartphrases
Structured opioid refill clinic epic smartphrases Structured opioid refill clinic epic smartphrases
Structured opioid refill clinic epic smartphrases
 
Opioids for the Treatment of Chronic Pain: Mistakes Made, Lessons Learned, an...
Opioids for the Treatment of Chronic Pain: Mistakes Made, Lessons Learned, an...Opioids for the Treatment of Chronic Pain: Mistakes Made, Lessons Learned, an...
Opioids for the Treatment of Chronic Pain: Mistakes Made, Lessons Learned, an...
 
The potential adverse influence of physicians’ words.
The potential adverse influence of physicians’ words.The potential adverse influence of physicians’ words.
The potential adverse influence of physicians’ words.
 
Ctaf chronic pain__evidence_report_100417
Ctaf chronic pain__evidence_report_100417Ctaf chronic pain__evidence_report_100417
Ctaf chronic pain__evidence_report_100417
 

Recently uploaded

THE SPECIAL SENCES- Unlocking the Wonders of the Special Senses: Sight, Sound...
THE SPECIAL SENCES- Unlocking the Wonders of the Special Senses: Sight, Sound...THE SPECIAL SENCES- Unlocking the Wonders of the Special Senses: Sight, Sound...
THE SPECIAL SENCES- Unlocking the Wonders of the Special Senses: Sight, Sound...
Nursing Mastery
 
HEALTH ASSESSMENT IN NURSING USING THE NURSING PROCESSpptx
HEALTH ASSESSMENT IN NURSING USING THE NURSING PROCESSpptxHEALTH ASSESSMENT IN NURSING USING THE NURSING PROCESSpptx
HEALTH ASSESSMENT IN NURSING USING THE NURSING PROCESSpptx
Rommel Luis III Israel
 
English Drug and Alcohol Commissioners June 2024.pptx
English Drug and Alcohol Commissioners June 2024.pptxEnglish Drug and Alcohol Commissioners June 2024.pptx
English Drug and Alcohol Commissioners June 2024.pptx
MatSouthwell1
 
3. User Guide Activity Budget Tracking App Steps to apply.pptx
3. User Guide Activity Budget Tracking App Steps to apply.pptx3. User Guide Activity Budget Tracking App Steps to apply.pptx
3. User Guide Activity Budget Tracking App Steps to apply.pptx
habtegirma
 
National Rural Health Mission(NRHM).pptx
National Rural Health Mission(NRHM).pptxNational Rural Health Mission(NRHM).pptx
National Rural Health Mission(NRHM).pptx
Jyoti Chand
 
Health Tech Market Intelligence Prelim Questions -
Health Tech Market Intelligence Prelim Questions -Health Tech Market Intelligence Prelim Questions -
Health Tech Market Intelligence Prelim Questions -
Gokul Rangarajan
 
Simple Steps to Make Her Choose You Every Day
Simple Steps to Make Her Choose You Every DaySimple Steps to Make Her Choose You Every Day
Simple Steps to Make Her Choose You Every Day
Lucas Smith
 
Daughter's of Dr Ranjit Jagtap (Poulami & Aditi)
Daughter's of Dr Ranjit Jagtap (Poulami & Aditi)Daughter's of Dr Ranjit Jagtap (Poulami & Aditi)
Daughter's of Dr Ranjit Jagtap (Poulami & Aditi)
Aditi Jagtap Pune
 
Data-Driven Dispensing- Rise of AI in Pharmacies.pdf
Data-Driven Dispensing- Rise of AI in Pharmacies.pdfData-Driven Dispensing- Rise of AI in Pharmacies.pdf
Data-Driven Dispensing- Rise of AI in Pharmacies.pdf
Jasper Colin
 
Top 5 Benefits of Cancer Registry Services
Top 5 Benefits of Cancer Registry ServicesTop 5 Benefits of Cancer Registry Services
Top 5 Benefits of Cancer Registry Services
Cardiac Registry Support
 
Faridkot ℂ𝕒𝕝𝕝 𝔾𝕚𝕣𝕝𝕤 7742996321 ℂ𝕒𝕝𝕝 𝔾𝕚𝕣𝕝𝕤 Faridkot
Faridkot ℂ𝕒𝕝𝕝 𝔾𝕚𝕣𝕝𝕤 7742996321 ℂ𝕒𝕝𝕝 𝔾𝕚𝕣𝕝𝕤 FaridkotFaridkot ℂ𝕒𝕝𝕝 𝔾𝕚𝕣𝕝𝕤 7742996321 ℂ𝕒𝕝𝕝 𝔾𝕚𝕣𝕝𝕤 Faridkot
Faridkot ℂ𝕒𝕝𝕝 𝔾𝕚𝕣𝕝𝕤 7742996321 ℂ𝕒𝕝𝕝 𝔾𝕚𝕣𝕝𝕤 Faridkot
varun0kumar00
 
Mohali Call Girls 7742996321 Call Girls Mohali
Mohali Call Girls  7742996321 Call Girls  MohaliMohali Call Girls  7742996321 Call Girls  Mohali
Mohali Call Girls 7742996321 Call Girls Mohali
Digital Marketing
 
Monopoly PCD Pharma Franchise in Tripura
Monopoly PCD Pharma Franchise in TripuraMonopoly PCD Pharma Franchise in Tripura
Monopoly PCD Pharma Franchise in Tripura
SKG Internationals
 
Friendly Massage in Ajman - Malayali Kerala Spa Ajman
Friendly Massage in Ajman - Malayali Kerala Spa AjmanFriendly Massage in Ajman - Malayali Kerala Spa Ajman
Friendly Massage in Ajman - Malayali Kerala Spa Ajman
Malayali Kerala Spa Ajman
 
NURSING MANAGEMENT OF PATIENT WITH EMPHYSEMA .PPT
NURSING MANAGEMENT OF PATIENT WITH EMPHYSEMA .PPTNURSING MANAGEMENT OF PATIENT WITH EMPHYSEMA .PPT
NURSING MANAGEMENT OF PATIENT WITH EMPHYSEMA .PPT
blessyjannu21
 
Test bank clinical nursing skills a concept based approach 4e pearson educati...
Test bank clinical nursing skills a concept based approach 4e pearson educati...Test bank clinical nursing skills a concept based approach 4e pearson educati...
Test bank clinical nursing skills a concept based approach 4e pearson educati...
rightmanforbloodline
 
The Importance of Black Women Understanding the Chemicals in Their Personal C...
The Importance of Black Women Understanding the Chemicals in Their Personal C...The Importance of Black Women Understanding the Chemicals in Their Personal C...
The Importance of Black Women Understanding the Chemicals in Their Personal C...
bkling
 
nurs fpx 4050 assessment 4 final care coordination plan.pdf
nurs fpx 4050 assessment 4 final care coordination plan.pdfnurs fpx 4050 assessment 4 final care coordination plan.pdf
nurs fpx 4050 assessment 4 final care coordination plan.pdf
Carolyn Harker
 
nursing management of patient with Empyema ppt
nursing management of patient with Empyema pptnursing management of patient with Empyema ppt
nursing management of patient with Empyema ppt
blessyjannu21
 
一比一原版(UoA毕业证)昆士兰科技大学毕业证如何办理
一比一原版(UoA毕业证)昆士兰科技大学毕业证如何办理一比一原版(UoA毕业证)昆士兰科技大学毕业证如何办理
一比一原版(UoA毕业证)昆士兰科技大学毕业证如何办理
xkute
 

Recently uploaded (20)

THE SPECIAL SENCES- Unlocking the Wonders of the Special Senses: Sight, Sound...
THE SPECIAL SENCES- Unlocking the Wonders of the Special Senses: Sight, Sound...THE SPECIAL SENCES- Unlocking the Wonders of the Special Senses: Sight, Sound...
THE SPECIAL SENCES- Unlocking the Wonders of the Special Senses: Sight, Sound...
 
HEALTH ASSESSMENT IN NURSING USING THE NURSING PROCESSpptx
HEALTH ASSESSMENT IN NURSING USING THE NURSING PROCESSpptxHEALTH ASSESSMENT IN NURSING USING THE NURSING PROCESSpptx
HEALTH ASSESSMENT IN NURSING USING THE NURSING PROCESSpptx
 
English Drug and Alcohol Commissioners June 2024.pptx
English Drug and Alcohol Commissioners June 2024.pptxEnglish Drug and Alcohol Commissioners June 2024.pptx
English Drug and Alcohol Commissioners June 2024.pptx
 
3. User Guide Activity Budget Tracking App Steps to apply.pptx
3. User Guide Activity Budget Tracking App Steps to apply.pptx3. User Guide Activity Budget Tracking App Steps to apply.pptx
3. User Guide Activity Budget Tracking App Steps to apply.pptx
 
National Rural Health Mission(NRHM).pptx
National Rural Health Mission(NRHM).pptxNational Rural Health Mission(NRHM).pptx
National Rural Health Mission(NRHM).pptx
 
Health Tech Market Intelligence Prelim Questions -
Health Tech Market Intelligence Prelim Questions -Health Tech Market Intelligence Prelim Questions -
Health Tech Market Intelligence Prelim Questions -
 
Simple Steps to Make Her Choose You Every Day
Simple Steps to Make Her Choose You Every DaySimple Steps to Make Her Choose You Every Day
Simple Steps to Make Her Choose You Every Day
 
Daughter's of Dr Ranjit Jagtap (Poulami & Aditi)
Daughter's of Dr Ranjit Jagtap (Poulami & Aditi)Daughter's of Dr Ranjit Jagtap (Poulami & Aditi)
Daughter's of Dr Ranjit Jagtap (Poulami & Aditi)
 
Data-Driven Dispensing- Rise of AI in Pharmacies.pdf
Data-Driven Dispensing- Rise of AI in Pharmacies.pdfData-Driven Dispensing- Rise of AI in Pharmacies.pdf
Data-Driven Dispensing- Rise of AI in Pharmacies.pdf
 
Top 5 Benefits of Cancer Registry Services
Top 5 Benefits of Cancer Registry ServicesTop 5 Benefits of Cancer Registry Services
Top 5 Benefits of Cancer Registry Services
 
Faridkot ℂ𝕒𝕝𝕝 𝔾𝕚𝕣𝕝𝕤 7742996321 ℂ𝕒𝕝𝕝 𝔾𝕚𝕣𝕝𝕤 Faridkot
Faridkot ℂ𝕒𝕝𝕝 𝔾𝕚𝕣𝕝𝕤 7742996321 ℂ𝕒𝕝𝕝 𝔾𝕚𝕣𝕝𝕤 FaridkotFaridkot ℂ𝕒𝕝𝕝 𝔾𝕚𝕣𝕝𝕤 7742996321 ℂ𝕒𝕝𝕝 𝔾𝕚𝕣𝕝𝕤 Faridkot
Faridkot ℂ𝕒𝕝𝕝 𝔾𝕚𝕣𝕝𝕤 7742996321 ℂ𝕒𝕝𝕝 𝔾𝕚𝕣𝕝𝕤 Faridkot
 
Mohali Call Girls 7742996321 Call Girls Mohali
Mohali Call Girls  7742996321 Call Girls  MohaliMohali Call Girls  7742996321 Call Girls  Mohali
Mohali Call Girls 7742996321 Call Girls Mohali
 
Monopoly PCD Pharma Franchise in Tripura
Monopoly PCD Pharma Franchise in TripuraMonopoly PCD Pharma Franchise in Tripura
Monopoly PCD Pharma Franchise in Tripura
 
Friendly Massage in Ajman - Malayali Kerala Spa Ajman
Friendly Massage in Ajman - Malayali Kerala Spa AjmanFriendly Massage in Ajman - Malayali Kerala Spa Ajman
Friendly Massage in Ajman - Malayali Kerala Spa Ajman
 
NURSING MANAGEMENT OF PATIENT WITH EMPHYSEMA .PPT
NURSING MANAGEMENT OF PATIENT WITH EMPHYSEMA .PPTNURSING MANAGEMENT OF PATIENT WITH EMPHYSEMA .PPT
NURSING MANAGEMENT OF PATIENT WITH EMPHYSEMA .PPT
 
Test bank clinical nursing skills a concept based approach 4e pearson educati...
Test bank clinical nursing skills a concept based approach 4e pearson educati...Test bank clinical nursing skills a concept based approach 4e pearson educati...
Test bank clinical nursing skills a concept based approach 4e pearson educati...
 
The Importance of Black Women Understanding the Chemicals in Their Personal C...
The Importance of Black Women Understanding the Chemicals in Their Personal C...The Importance of Black Women Understanding the Chemicals in Their Personal C...
The Importance of Black Women Understanding the Chemicals in Their Personal C...
 
nurs fpx 4050 assessment 4 final care coordination plan.pdf
nurs fpx 4050 assessment 4 final care coordination plan.pdfnurs fpx 4050 assessment 4 final care coordination plan.pdf
nurs fpx 4050 assessment 4 final care coordination plan.pdf
 
nursing management of patient with Empyema ppt
nursing management of patient with Empyema pptnursing management of patient with Empyema ppt
nursing management of patient with Empyema ppt
 
一比一原版(UoA毕业证)昆士兰科技大学毕业证如何办理
一比一原版(UoA毕业证)昆士兰科技大学毕业证如何办理一比一原版(UoA毕业证)昆士兰科技大学毕业证如何办理
一比一原版(UoA毕业证)昆士兰科技大学毕业证如何办理
 

Corticolimbic anatomic characteristics predetermine risk for chronic pain.

  • 1. Corticolimbic anatomical characteristics predetermine risk for chronic pain Etienne Vachon-Presseau,1 Pascal Te´treault,1 Bogdan Petre,1 Lejian Huang,1 Sara E. Berger,1 Souraya Torbey,2 Alexis T. Baria,1 Ali R. Mansour,1 Javeria A. Hashmi,3 James W. Griffith,4 Erika Comasco,5 Thomas J. Schnitzer,6 Marwan N. Baliki1,7 and A. Vania Apkarian1 Mechanisms of chronic pain remain poorly understood. We tracked brain properties in subacute back pain patients longitudinally for 3 years as they either recovered from or transitioned to chronic pain. Whole-brain comparisons indicated corticolimbic, but not pain-related circuitry, white matter connections predisposed patients to chronic pain. Intra-corticolimbic white matter connectivity analysis identified three segregated communities: dorsal medial prefrontal cortex–amygdala–accumbens, ventral medial prefrontal cortex–amygdala, and orbitofrontal cortex–amygdala–hippocampus. Higher incidence of white matter and functional connections within the dorsal medial prefrontal cortex–amygdala–accumbens circuit, as well as smaller amygdala volume, represented inde- pendent risk factors, together accounting for 60% of the variance for pain persistence. Opioid gene polymorphisms and negative mood contributed indirectly through corticolimbic anatomical factors, to risk for chronic pain. Our results imply that persistence of chronic pain is predetermined by corticolimbic neuroanatomical factors. 1 Department of Physiology, Feinberg School of Medicine, Northwestern University 303 E. Chicago Ave., Chicago, IL 60611, USA 2 Department of Psychiatry and Neurobehavioral Sciences, University of Virginia , 2955 Ivy Rd, Suite 210, Charlottesville, VA 22903, USA 3 Department of Anesthesia, Pain Management and Perioperative Medicine Dalhousie University, Halifax, NS, Canada B3H 4R2 4 Department of Medical Social Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA 5 Department of Neuroscience, Science for Life Laboratory, Uppsala University, BMC, Pob 593, 75124, Uppsala, Sweden 6 Northwestern University Feinberg School of Medicine, Departments of Physical Medicine and Rehabilitation and Internal Medicine/Rheumatology, 710 N. Lake Shore Drive, Room 1020, Chicago, IL 60611, USA 7 Rehabilitation Istitute of Chicago, 345 E Superior St, Chicago, IL 60611, USA Correspondence to: A. V. Apkarian, Department of Physiology, Feinberg School of Medicine, Northwestern University 303 E. Chicago Ave., Chicago, IL 60611, USA E-mail: a-apkarian@northwestern.edu Correspondence may also be addressed to: Marwan N. Baliki, E-mail: marwanbaliki2008@u.northwestern.edu Keywords: chronic pain; brain network; limbic system; magnetic resonance imaging (MRI); diffusion tensor imaging (DTI) Abbreviations: DTI = diffusion tensor imaging; PFC = prefrontal cortex; SBP = subacute back pain; SNP = single nucleotide polymorphism doi:10.1093/brain/aww100 BRAIN 2016: Page 1 of 13 | 1 Received December 18, 2015. Revised February 11, 2016. Accepted March 16, 2016. ß The Author (2016). Published by Oxford University Press on behalf of the Guarantors of Brain. All rights reserved. For Permissions, please email: journals.permissions@oup.com Brain Advance Access published May 5, 2016 byguestonMay9,2016http://brain.oxfordjournals.org/Downloadedfrom
  • 2. Introduction Chronic pain is a state of continued negative emotional suf- fering representing a leading source of worldwide disability (Murray and Lopez, 2013) with a staggeringly high healthcare cost (Medicine, 2011). Risk factors for developing chronic pain remain poorly understood, yet its negative emotional valence implicates the involvement of limbic circuitry. In fact, both Maclean’s (1955) original formulation of the limbic brain and Melzack and Casey’s (1968) concept of the gate control theory of pain (Melzack and Wall, 1965) hypothesized that the limbic brain plays an integral role in pain states. Consistently, long-standing clinical evidence has demonstrated that injury to limbic structures diminishes the emotional overtones of pain (Raz, 2009). However, only recent human neuroimaging (Apkarian et al., 2004; Baliki et al., 2006, 2010, 2012; Geha et al., 2008; Hashmi et al., 2013; Mansour et al., 2013; Mutso et al., 2014) and comple- mentary animal model (Neugebauer et al., 2003; Metz et al., 2009; Ji and Neugebauer, 2011; Ren et al., 2011; Mutso et al., 2012; Baliki et al., 2014; Chang et al., 2014; Schwartz et al., 2014) studies have begun to unravel the role of limbic brain properties in chronic pain. In the present study, we focus on intrinsic properties of the corticolimbic portion of the mesocorticolimbic system. A net- work comprised of the prefrontal cortex (PFC) (medial and orbital cortices), nucleus accumbens, hippocampus/parahippo- campus, and amygdala. Collectively, these structures support emotion, behaviour, motivation and memory functions. Converging evidence reinforces the idea that components of this circuitry are important for chronic pain. In humans, the nucleus accumbens encodes salience of impending pain and the reward value of analgesia, which is distorted in chronic back pain (Baliki et al., 2010), and functional connectivity between nucleus accumbens and medial PFC prospectively predicts chronification of pain (Baliki et al., 2012). Prefrontal cortical activity reflects subjective reports of back pain intensity in patients with chronic back pain (Baliki et al., 2006; Hashmi et al., 2013) and is thought to regulate the meaning of affective responses and influence emotional deci- sion-making. Furthermore, the size of the amygdala and hippocampus show strong genetic dependences (Hibar et al., 2015) and both structures are implicated in emotional learn- ing (Phelps and LeDoux, 2005), anxiety (Russo et al., 2012), and stress regulation (Vachon-Presseau et al., 2013), and ex- hibit changes in activity and functional connectivity during the transition to chronic pain (Mutso et al., 2012; Hashmi et al., 2013). Comparative evidence further bolsters corticolimbic in- volvement in chronic pain by demonstrating that single neuron morphology, excitability, and short- or long-term po- tentiation are distorted within and across the hippocampus (Ren et al., 2011; Mutso et al., 2012), amygdala (Neugebauer et al., 2003; Ji and Neugebauer, 2011), nucleus accumbens (Chang et al., 2014; Schwartz et al., 2014), and PFC (Metz et al., 2009; Ji and Neugebauer, 2011). Consistently, the persistence of pain-like behaviours in rodents is accompanied by a reorganization of resting state connect- ivity primarily across limbic brain structures (Baliki et al., 2014). Despite this compelling evidence, a comprehensive examination of corticolimbic brain contributions to chronic pain remains to be undertaken. Here we interrogate the func- tional and anatomical properties of the corticolimbic neural network, and its subcircuits, to unravel the interrelationships between change in pain intensity, negative emotion, and their potential genetic mediators. We report the novel finding that anatomical characteristics of specific networks and structures of the corticolimbic system predetermine risk for developing chronic pain. Materials and methods Additional information regarding whole brain region of inter- est parcellation schemes, pain and corticolimbic nodes, corti- colimbic parcellation, modularity analyses of limbic structural network, and genetic analyses can be found in the Supplementary material. Participants The data presented are from a longitudinal observational study where we followed patients with subacute back pain (SBP) over 3 years. This report is based on the final total sample of patients. Portions of the data were used in previous publications (Baliki et al., 2012; Hashmi et al., 2013; Mansour et al., 2013; Petre et al., 2015). The study recruited 159 patients with SBP and 29 healthy control subjects. All patients with SBP were diagnosed by a clinician for back pain and reported pain intensity greater than 40/100 on a visual analogue scale (VAS, 0–100, where 100 = maximum imaginable pain and 0 = no pain) prior to en- rolling in the study. Patients with SBP were recruited only if the duration of their pain was between 4–16 weeks with no back pain reported during the previous 12 months. Subjects were excluded if they reported other chronic painful conditions, sys- temic disease, psychiatric diseases, or history of head injuries. Subjects were also excluded if they reported high depression, which was defined as scores 419 using the Beck’s Depression Inventory (BDI) (Beck et al., 1961). From these participants, 69 SBP (34 females, 43.1 Æ 10.4 years of age) and 20 healthy con- trols (nine females, 37.4 Æ 7.5 years of age) completed the study to 1-year follow-up. These patients with SBP were dichotomized into groups with persisting pain (SBPp; n = 39) or recovery from pain at the end of the year (with recovery operationalized as 20% reduction in pain from Week 0 to Week 56; SBPr n = 30). A subsample of 39 patients with SBP (18 females, 44.2 Æ 11.3 years of age) was followed for an additional visit at 3 years from pain onset. Dates of visits are shown for all subjects in Supplementary Fig. 1. The Institutional Review Board of Northwestern University approved the study and all participants signed a consent form. Additional groups of patients and healthy controls were used to cross-sectionally examine subcortical limbic volumes: 23 with chronic back pain (nine females, 46.6 Æ 6.0 years of age), 40 osteoarthritis patients (22 females, 58.6 Æ 7.6 years of age), and 20 healthy age-matched controls (10 females, 57.9 Æ 6.7 years of age). 2 | BRAIN 2016: Page 2 of 13 E. Vachon-Presseau et al. byguestonMay9,2016http://brain.oxfordjournals.org/Downloadedfrom
  • 3. Pain characteristics, depressive mood, and negative affect Pain intensity was measured using the visual analogue scale (VAS) from the short version of the McGill Pain Questionnaire (Melzack, 1975) and disability was assessed using the Pain Disability Index (PDI) measuring the degree at which chronic pain disrupts aspects of the patients’ lives (Tait et al., 1990). Negative affect was calculated with the negative scale of the Positive and Negative Affectivity Scale (PANAS). This scale consists of a series of items describing different emo- tions experienced over the previous week (Watson and Clark, 1999). Depressive mood was assessed using BDI-1b. All ques- tionnaires were given within an hour before brain scanning. Summary results for pain characteristics, depressive mood and negative affect are presented in Supplementary Table 1. Scanning parameters Anatomical (T1), diffusion tensor imaging (DTI), and functional MRI were collected in the same scanning session. High-reso- lution T1-anatomical brain images were acquired with a 3 T Siemens Trio whole-body scanner with echo-planar imaging capability using the standard radio-frequency head coil using the following parameters: voxel size = 1 Â 1 Â 1 mm, repetition time = 2.50 ms, echo time = 3.36 ms, flip angle = 9 , in-plane matrix resolution = 256 Â 256; 160 slices, field of view = 256mm. Functional MRI images were acquired using the following parameters: multi-slice T2*-weighted echo-planar images with repetition time = 2.5 s, echo time = 30 ms, flip angle = 90 , number of volumes = 244, slice thickness = 3 mm, in plane resolution = 64 Â 64. The 36 slices covered the whole brain from the cerebellum to the vertex. DTI images were acquired using echo planar imaging (72 Â 2-mm thick axial slices; matrix size = 128 Â 128; field of view = 256 Â 256 mm2 , resulting in a voxel size of 2 Â 2 Â 2 mm). Images had an isotropic distribution along 60 directions using a b-value of 1000 s/mm2 . For each set of diffu- sion-weighted data, eight volumes with no diffusion weighting were acquired at equidistant points throughout the acquisition. Morphometric analyses of subcortical structures Structural data were analysed with the standard automated processing stream of FSL 4.1 that shows very high reliability across labs (Nugent et al., 2013). The analysis entailed skull extraction, a two-stage linear subcortical registration, and seg- mentation using FMRIB’s Integrated Registration and Segmentation Tool (FIRST). The volumes of right and left nu- cleus accumbens, amygdala, hippocampus, and thalamus were calculated for each subject. Group comparisons were per- formed using repeated analyses of covariance with volume as the dependent factor, time of the scan and hemisphere as within-subject factors, the group as a between-subject factor (SBPp, SBPr, and healthy), and age, sex and total grey matter volume [estimated using FSL SIENAX (Smith et al., 2002)] as covariates of no interest. Post hoc comparisons were Bonferroni corrected for multiple comparisons. From the initial 89 participants, nine were excluded because T1 was missing or because of quality control failure of the T1 image at one of the four visits. Quality control included: (i) visual inspection of the subcortical segmentation, where gross mismatches between underlying anatomy and FIRST outcome were excluded; and (ii) large deviations from the mean volume of a given structure were also excluded [greater than Æ2 standard deviations (SD)]. Outliers were identified independ- ently for hippocampus and amygdala volumes. From a total of 80 subjects, 11 were rejected from the amygdala analyses and four were rejected from the hippocampus analyses. Importantly, removing these individuals did not change the overall statistical outcomes of our results. Our subsample of patients scanned a fifth time at Week 156 included 33 patients with accurate segmentation of subcortical structures. Differences between SBPp and SBPr in amygdala and hippo- campus volumes, initially discovered using FSL, were repli- cated using the standard automated cortical and subcortical segmentations by Freesurfer 5.0 (http://surfer.nmr.mgh.har vard.edu/). The procedure includes motion correction, removal of the skull using watershed/surface deformation procedure, normalization in Talairach space, and segmentation of the sub- cortical structures based on the existing atlas containing prob- abilistic information on the location of structures (Fischl et al., 2002). As hippocampus and amygdala exhibited significant group differences, we investigated whether these changes can be attributed to localized structural deformations. Vertex repre- sentation of both structures were therefore generated using FIRST (Patenaude et al., 2011). The model uses a Bayesian approach representing the structure in 3D from meshes fitting the form of the subcortical structure to a training dataset that was manually segmented. The shapes of the subcortical struc- tures were represented from meshes parameterized by the co- ordinates of their vertices. A multivariate permutation test on the 3D coordinates of corresponding vertices provide the F-values representing group differences (SBPp and SBPr). Group differences in vertex localized at the boundary provided a geometric change shape of the amygdala and the hippocam- pus. No corrections for multiple comparisons were performed on these analyses. Diffusion tensor imaging preprocessing FAST (FMRIB Automated Segmentation Tool) was used to segment the 3D T1 image of the brain into different tissue types (grey matter, white matter, and CSF) while also correct- ing for spatial intensity variations (Supplementary material). For each subject, the grey matter and white matter masks were transformed into standard space using FSL linear regis- tration tool (FLIRT). Those masks were then thresholded (probability 4 0.25) and multiplied with each other, yielding a subject specific grey–white matter interface composed of re- gions having relatively equal probabilities of being in either category, thus constituting the grey–white matter boundary. Probabilistic tractography of the whole brain networks were constructed from voxels composing this grey–white matter boundary. Analysis was performed using tools from the FMRIB Diffusion Toolbox (FDT; Behrens et al., 2003) and in-house Anatomical risk factors for chronic pain BRAIN 2016: Page 3 of 13 | 3 byguestonMay9,2016http://brain.oxfordjournals.org/Downloadedfrom
  • 4. software. DTI data were manually checked for obvious arte- facts. Eddy current correction was used and simple head motion correction using affine registration to a no-diffusion reference volume for each participant. Data were then skull extracted (BET) and a diffusion tensor model was fit at each voxel determining voxel-wise fractional anisotropy. Bayesian Estimation of Diffusion Parameters Obtained using Sampling Techniques (BEDPOSTX) and probabilistic tractography (PROBTRACX) modelling two fibre tracks per voxel were used to determine the probability of connection between re- gions (Behrens et al., 2007). Voxel-wise principal diffusion directions were used to repetitively compute a probabilistic streamline from 5000 samples to generate posterior distribu- tion on the ‘connectivity distribution’. Probabilistic tractogra- phy was performed in the diffusion space of each participant and then transformed to MNI space using FSL linear registra- tion (FLIRT; Jenkinson et al., 2002). The connectivity distri- bution was averaged across voxels of each of the 264 and 480 regions of interest (Supplementary Fig. 3) for whole brain net- work analyses or across voxels of the subsample in 4 Â 4 Â 4 mm for the corticolimbic network analyses (1068 regions of interest; Supplementary Fig. 4). From the 89 par- ticipants, 15 were excluded because DTI images were missing at one of the four visits or because of quality control failure. From the remaining 74 patients, two patients were further excluded from the limbic network analyses because they showed connectivity counts in the dorsal medial PFC–amyg- dala–nucleus accumbens 43 SD from the group mean. Diffusion scans were collected in 36 patients out of the sub- sample of patients scanned a fifth time at Week 156. Functional MRI preprocessing Ten minutes of functional MRI were acquired at each visit of the study, where subjects rated spontaneous fluctuations of their pain. These data were used to examine intrinsic brain connectivity differences between groups (SBPp and SBPr). The preprocessing of each subject’s time series of functional MRI volumes was performed using the FMRIB Expert Analysis Tool (FEAT). The sequence included skull extraction using BET from FSL, slice time correction, and motion correc- tion (Jenkinson et al., 2002). Temporal filtering between 0.01 and 0.1 Hz was applied on the blood oxygenation level-de- pendent (BOLD) time series. The six parameters of head motion, global mean signal measured over all voxels of the brain, the signal from a ventricular region of interest, and the signal from a region centred in the white matter were re- gressed as covariates of no interest from the BOLD time series. The first four volumes were removed for signal stabilization. After preprocessing, functional scans were registered into a standard MNI space using FSL FLIRT. From the 69 patients that rated their spontaneous pain fluctuations, seven were excluded because of missing functional scans at one of the four visits. Our subsample of patients scanned a fifth time at Week 156 included 34 patients with functional scans. Construction of white matter networks Whole brain DTI-based connectivity networks for the 480 re- gions of interest for SBPp and SBPr at Week 0 were constructed using methods described in (Rubinov and Sporns, 2010). For all regions of interest, the connection prob- ability between a seed region of interest and any target region of interest in the brain is defined as the sum of sample fibre lengths connecting these two regions of interest. For each sub- ject, a 480 Â 480 and a 264 Â 264-weighted directed matrices were generated from connection probabilities for all seed re- gions of interest with the number of target regions of interest. The resultant matrices were then symmetrized and adjusted for distance constraints by multiplication with Euclidean distance matrix. Finally, the matrix was thresholded at a value that yielded a binary undirected network of different link densities ranging from 0.02 to 0.20 (i.e. the top 2–20% of connections were assigned a value of 1, whereas the rest of the connections were assigned a value of 0; Supplementary Fig. 6). Group dif- ferences (SBPp and SBPr) in edges connecting nodes of the network were initially determined using permutation test con- trolling for family-wise error rate using Network Based Statistics (NBS; Zalesky et al., 2010). Corticolimbic white matter networks formed between 1068 Â 1068 nodes were generated for all subjects (SBPp, SBPr and healthy controls), at each of the five visits using the same methods. Construction of functional brain networks To construct the limbic functional connectivity networks for each subject, we first computed the Pearson correlation coeffi- cient (r) for the all possible pairs of the n = 1068 of cortical and subcortical limbic regions of interest time series from the preprocessed resting state functional MRI data. Similar to white matter networks, threshold was calculated to produce a fixed number of connections across link densities ranging from 0.02 to 0.20 (top 2–20% of r-values binarized), for each subject (Achard et al., 2012). The values of the threshold are therefore subject-dependent but the number of connections in the network is the same for every subject. The density of structural connections was examined with respect to the com- munity-based organization of the white matter network. Therefore, group differences (SBPp and SBPr) in functional connectivity within and across modules determined from white matter tractography were tested using repeated measure ANOVAs entering the visits as repeated variable. Genetic association with mesolimbic brain outcome measures To test the idea that some of the mesolimbic brain properties associated with risk of chronic pain may underlie a genetic pre- disposition, we looked for associations between single nucleotide polymorphisms (SNPs) and limbic brain properties. Saliva sam- ples were collected from all subjects who completed the study. Given the limited number of subjects in the study we decided to only examine 30 SNPs. The choice of SNPs was determined prior to having knowledge regarding the primary functional or ana- tomical determinants of risk for chronic pain. The 30 candidate SNPs located in 12 different genes were selected for their known involvement in neurotransmitters, receptors and metabolites asso- ciated with corticolimbic structures, and also for being related to pain or nociception. Only one gene was selected as a marker for peripheral encoding of nociception, SCN9A. A description of 4 | BRAIN 2016: Page 4 of 13 E. Vachon-Presseau et al. byguestonMay9,2016http://brain.oxfordjournals.org/Downloadedfrom
  • 5. each gene is presented in Supplementary Table 8. Genetic analysis was done independently and with no knowledge of the results of brain analyses. Four brain outcome parameters were tested for association with the 30 selected SNPs. SNPs that passed statistical threshold (or were borderline significant) after Bonferroni mul- tiple comparison correction were also tested for discrimination between SBPp and SBPr. Statistical analyses Path analyses and multiple regression models Path analysis was used to examine the direct and mediating links between multiple variables to test a given theoretical model empirically, specifically testing the prediction for SBP grouping at Week 56 using a logistic regression model. Path analyses were performed with Mplus 7.3 using the robust weighted least squares estimator method accounting for the non-normality of the data. We calculated 95% confidence intervals for the indirect effect using a recommended bias- corrected bootstrapped method (bootstrap of 1000 iterations; Mackinnon et al., 2004); we rejected the null hypothesis when the 95% confidence interval did not contain zero. Results This report presents the complete results of a longitudinal study. Patients recruited while in subacute back pain were followed over the course of 1 year (n = 69 patients with SBP), with a smaller subset followed for 3 years (n = 39). Thirty-nine patients with SBP were dichotomized into per- sisting pain (SBPp) and 30 SBP were dichotomized into recovery from pain as they showed 20% reduction in pain intensity between Week 0 and Week 56 (SBPr). Classification of patients with SBP at 3 years was also based on the Week 56 criterion (SBPp n = 23, SBPr n = 16). Group differences in pain intensity and disability were observed as early as 8 weeks after entry into the study and maintained temporal stability up to 3 years after back pain onset (Fig. 1B). Over 3 years, no changes were observed in negative psychological traits for SBPp or SBPr (Supplementary Table 1), and medication use was not dif- ferent between SBP groups (Supplementary Fig. 2). Whole-brain white matter connectivity We used DTI probabilistic tractography-based structural connectivity at study entry to identify brain areas for which differences in white matter connectivity conferred greater risk for chronic pain. The DTI scans at study entry (10.8 Æ 5.5 weeks from onset of back pain) were used to construct whole-brain structural networks (Supplementary Fig. 3) from different parcellation schemes of 480 and 264 regions (Fig. 1C and Supplementary Fig. 6). Permutation tests were used to identify group differ- ences in the incidence of connections, for each parcellation scheme, across link densities ranging from 0.02 to 0.2. Example group contrast results (SBPp 4 SBPr, and SBPr 4 SBPp; Supplementary Tables 3 and 4) for both par- cellations, at 0.1 link density, are illustrated in Fig. 1D and E. Connections were denser in SBPp and mostly localized across corticolimbic nodes (Supplementary Tables 5 and 6). In addition to corticolimbic nodes, we also identified nodes related to pain perception using a term-based meta-analysis tool (based on a map derived for ‘pain’ from 420 PubMed publications; www.neurosynth.org). Group differences in structural connections were localized outside pain-related nodes. Instead, the likelihood for denser connections in SBPp, compared to SBPr, was consistently higher within the corticolimbic brain compared to random networks (Fig. 1F and G). Collectively, these analyses indicate that structural connections between regions involved in pain processing were not involved in the development of chronic pain. In contrast, denser white matter connections were observed between nodes concentrated within the cortico- limbic system, implying that these connections were more likely to predispose individuals to transition to chronic pain. Corticolimbic modules contribute to risk for chronic pain The identification of dense corticolimbic white matter con- nections does not provide information about underlying anatomical and functional properties of this system that may impart risk for chronic pain. Therefore, we next inter- rogated the intrinsic structural and functional network organization of the corticolimbic brain throughout the 3 years of observation, using higher spatial resolution data. Anatomical networks were first constructed from con- nections between 1068 nodes (4 Â 4 Â 4 mm voxels) be- longing to the corticolimbic system (nucleus accumbens, amygdala, hippocampus-parahippocampus, and the pre- frontal cortex) (Fig. 2A and Supplementary Fig. 4). Edges between the nodes were drawn from DTI-based probabil- istic white matter tractography and from Pearson correl- ations between functional MRI time series (i.e. functional connections derived from functional MRI activity as pa- tients rated back pain intensity). The structural (white matter network) and functional (functional connectivity network) graphs were constructed by binarizing the net- works across different densities ranging from 0.02 to 0.20 (Supplementary Fig. 6). Derived networks were tested and confirmed to be insensitive to standard con- founds (Supplementary Fig. 5). We first studied the architecture of the limbic white matter network (Fig. 2A). This fine-grained, voxel-based, intra-limbic probabilistic tractography-based white matter network revealed dense, bilateral connections of subcortical structures with each other and with the prefrontal cortex. Modularity analysis revealed that this network was com- posed of three distinct communities, including modules composed of (i) dorsal and medial portion of PFC, Anatomical risk factors for chronic pain BRAIN 2016: Page 5 of 13 | 5 byguestonMay9,2016http://brain.oxfordjournals.org/Downloadedfrom
  • 6. medial-inferior part of the amygdala, and most of the nu- cleus accumbens (dorsal medial PFC–amygdala–nucleus accumbens); (ii) most of the ventral medial frontal cortex (ventral medial PFC) and medial-superior part of the amyg- dala (ventral medial PFC–amygdala); and (iii) a thin layer of orbital cortex linked to lateral amygdala and most of hippocampus (orbitofrontal cortex–amygdala–hippocam- pus) (Fig. 2B). The stability of the community structure was preserved across different clustering thresholds (Supplementary Fig. 7). Note that our results are consistent Figure 1 Whole-brain structural connectivity identifies corticolimbic regions conferring risk for chronic pain. (A) Timeline of brain scans and type and number of subjects studied. (B) Recovering (SBPr) and persisting (SBPp) patients were dichotomized based on 20% reduction in back pain intensity at Week 56. The two groups showed time-dependent divergence (Weeks 0–56) in pain intensity [F(2.52, 140.92); P 5 0.001 repeated measure ANOVA] and pain disability [F(2.58, 110.74) = 3.44; P = 0.025 repeated measure ANOVA], sustained in the 3-year subsamples [Week 156 pain intensity t(1,33) = 3.67; P = 0.001; pain disability t(1,36) = 3.28; P = 0.002 two-sided unpaired t-tests]. * P 5 0.05 between group comparison; + P 5 0.05 within group comparison relative to Week 0. (C) Structural white matter-based networks were con- structed from probabilistic tractography between nodes covering the whole brain. (D and F) Group comparison at link density of 0.1 at Week 0 was studied from network constructed from 480 and 264 parcellation schemes. In both networks SBPp show more white matter connections between corticolimbic nodes (P 5 0.05, FDR corrected). (E) In 480-parcellation, location and number of nodes within pain and corticolimbic systems are illustrated. The SBPp 4 SBPr contrast indicates denser structural connections in SBPp across corticolimbic nodes, but not for pain- related nodes, for all link densities. Observed incidence of connections compared to expected incidence in a random network was significantly higher for corticolimbic nodes (chi-square test, P 5 0.0001 for all link densities) but not for pain-related nodes (P 4 0.28 for all link densities). Histogram is the cumulative probability (total connectivity = 1.0) for corticolimbic nodes and pain-related nodes compared to expected con- nectivity in a random network, averaged for all densities. (G) Concordant results were observed in 264-parcellation. Observed incidence of connections in SBPp was much higher than expected across corticolimbic nodes (chi-square test, P 5 0.0001 for all link densities) but not for pain- related nodes (P 4 0.28 for all link densities). Number of subjects is shown in parentheses. Error bars indicate SEM. 6 | BRAIN 2016: Page 6 of 13 E. Vachon-Presseau et al. byguestonMay9,2016http://brain.oxfordjournals.org/Downloadedfrom
  • 7. with the anatomical subdivisions of the amygdala, based on meta-analytic functional co-activation maps of the amyg- dala with the rest of the brain, which also show a close correspondence to the region’s cytoarchitectonic subdiv- isions (Bzdok et al., 2013) (Supplementary Fig. 8). We next studied group differences and temporal variabil- ity in density of intra-limbic white matter and functional connectivity networks, both within and between the three corticolimbic circuits. Although white matter network modular organization was common across SBPr, SBPp and healthy individuals (Fig. 2C), SBPp consistently dis- played a greater frequency of white matter connections within the dorsal medial PFC–amygdala–nucleus accum- bens module across all link densities (Fig. 2D), across the four visits (Fig. 2E) and 3 years after pain onset (Fig. 2E). The white matter networks of ventral medial PFC–amyg- dala and orbitofrontal cortex–amygdala–hippocampus modules did not differ between SBP groups (Supplementary Fig. 9A). The dorsal medial PFC–amyg- dala–nucleus accumbens white matter network was stable over 3 years because individual SBP values were strongly correlated between Week 0 and Week 156 for all SBP (Fig. 2E). This finding is consistent with and generalizes from a previous report that white matter regional fractional anisot- ropy distinguishes between SBP groups (based on a sub- sample of the current cohort; Mansour et al., 2013). Examining the functional connectivity network connec- tions in each one of the modules, defined in Fig. 2B, showed that SBPp displayed higher incidence of connec- tions within the dorsal medial PFC–amygdala–nucleus accumbens module at all densities (Fig. 2F), at all four visits (Fig. 2G), a finding that expands on our previous report of stronger (seed-to-target) medial PFC–nucleus accumbens functional connectivity in a subgroup of SBPp (Baliki et al., 2012). However, the SBP group difference in dorsal medial PFC–amygdala–nucleus accumbens func- tional network connectivity was not maintained 3 years later (Fig. 2G and Supplementary Fig. 10A and B), with non-significant correlations between study entry and Week 156 (Fig. 2H). The functional connectivity networks of ven- tral medial PFC–amygdala and orbitofrontal cortex–amyg- dala–hippocampus modules did not differ between SBP groups (Supplementary Fig. 9B). Taken together, these re- sults reinforce that the functional connectivity within the dorsal medial PFC–amygdala–nucleus accumbens is predict- ive of the transition to chronic pain 1 year later; in con- trast, its impact dissipates by Year 3, suggesting that it is not required for the longer term maintenance of chronic pain. Amygdala, hippocampus and risk for chronic pain Morphometric measures of the amygdala and hippocampus (Fig. 3A and B) were examined because they both show strong genetic dependence (Hibar et al., 2015), represent risk factors for developing pathological emotional states such as anxiety (Russo et al., 2012) and depression (Drevets et al., 2008), and previous studies suggest that both may be involved in pain chronification (Metz et al., 2009; Ji and Neugebauer, 2011; Baliki et al., 2012; Hashmi et al., 2013). Given our white matter and functional con- nectivity network findings, we also examined the volumes of nucleus accumbens (a main component of the dorsal medial PFC–amygdala–nucleus accumbens module) and thalamus, as the latter is a primary relay for nociceptive transmission to the cortex through the spinothalamocorti- cal projections. SBPp exhibited $10% smaller amygdala (Fig. 3C) and hippocampus (Fig. 3D) total volumes (left + - right), as compared to SBPr and healthy controls (Supplementary Table 7). This effect was sustained over 3 years with strong correlations for volumes between study entry to Week 156 for all SBP, for the amygdala and hippocampus. These results were replicated using Freesurfer as an alternative segmentation tool for extracting subcortical volumes (Supplementary Fig. 11). However, no significant group differences were observed for volumes of the nucleus accumbens or the thalamus. Exploratory ana- lyses further revealed that smaller amygdala and hippocam- pus volumes were accompanied by thinning of mainly the right amygdala shape in SBPp at all visits (Fig. 3G). We previously have shown that the hippocampus volume is smaller in patients with chronic pain (Mutso et al., 2012). Here we replicate the finding in two new patient groups suffering from longstanding chronic back pain and osteoarthritis and show reduced volumes for both amyg- dala and hippocampus. More importantly comparison be- tween these groups and SBPp shows that extent of volume decrease is similar across chronic pain patients and SBPp at Week 0 (Fig. 3E and F). Given that both amygdala and hippocampus volumes distinguished between SBP groups, their morphologies did not change over 3 years, and smal- ler amygdala and hippocampus volumes were comparable between SBPp and multiple chronic pain conditions, these volumetric measures (derived and validated using two un- biased measurements, which are replicated in many labs in more than 30 000 subjects; Hibar et al., 2015) most likely reflect pre-existing circuit abnormalities that induce risk for chronic pain, across multiple chronic pain conditions. Genetic associations with limbic risk factors Data from twin studies show that chronic back pain is 20– 67% heritable (Ferreira et al., 2013). Given the recent notion that genetic variants of some pathologies are more penetrant at the level of brain measures (Rose and Donohoe, 2013), we explored the hypothesis that gene vari- ations can be linked to corticolimbic risk factors for chronic pain. We used an imaging–genetics approach to examine associations between SNPs and corticolimbic brain risk fac- tors. In total, 30 candidate SNPs located in 12 different Anatomical risk factors for chronic pain BRAIN 2016: Page 7 of 13 | 7 byguestonMay9,2016http://brain.oxfordjournals.org/Downloadedfrom
  • 8. genes were selected for their known involvement in limbic functions and also to pain or nociception (Supplementary Tables 9–11). Genotypes were compared with regard to amygdala and hippocampus volumes, and dorsal medial PFC–amygdala–nucleus accumbens white matter and func- tional connectivity network connectivity for Week 0 values. Then, we secondarily tested if these genes could also differentiate between SBP groups as identified at Week 56. After correcting for multiple comparisons, the opioid recep- tor delta 1 (OPRD1) rs678849 SNP was associated with amygdala volume, and opioid receptor mu 1 (OPRM1) rs1799971 SNP was associated with incidence of connec- tions within the dorsal medial PFC–amygdala–nucleus accumbens white matter network. Moreover, OPRD1 Figure 2 Intra-corticolimbic white matter connectivity identifies three modules, only the dorsal medial PFC–amygdala–nu- cleus accumbens module imparts risk for chronic pain. (A) Display of the intra-corticolimbic white matter-based structural networks generated from 1068 nodes (white matter networks) at 0.1 link density. (B) Modularity analysis performed on this network separated the system into three communities: (i) dorsal medial PFC–amygdala–nucleus accumbens (mPFCdorsal-amy-NAc); (ii) ventral medial PFC–amygdala (mPFCventral-Amy); and (iii) orbitofrontal cortex–amygdala–hippocampus (OFC-amy-hipp). (C) Normalized mutual information was used to quantify similarity between individual subject community structure and the mean group community structure presented in (B) SBPp, SBPr and healthy controls displayed similar modular organization across all time points, due to absence of group differences [F(2,69) = 2.34; P = 0.10 repeated measure ANOVA] or a time  group interaction [F(5.73, 200.65) = 0.72; P = 0.63 repeated measure ANOVA]. (D) Structural con- nection probability for SBPp, SBPr and healthy within the dorsal medial PFC–amygdala–nucleus accumbens module calculated across link densities ranging between 0.02 and 0.2. Incidence of white matter connections was higher in SBPp at all densities [F(2,78) = 6.10; P = 0.003 repeated measure ANOVA]. The histogram displays group differences at 0.1 link density. (E) Per cent structural connections for SBPp and SBPr and healthy controls over 3 years at 0.1 link density. Incidence of connections in the dorsal medial PFC–amygdala–nucleus accumbens module was higher in SBPp [F(2,69) = 4.74; P = 0.012 repeated measure ANOVA] for visits 0–56 weeks. Concordant results were seen at Week 156 [t(1,34) = 1.79; P = 0.08 two-sided unpaired t-tests]. Scatterplot shows 3-year stability of dorsal medial PFC–amygdala–nucleus accumbens module connectivity for all SBP. (F) Incidence of functional connections in the dorsal medial PFC–amygdala–nucleus accumbens module was higher in SBPp across 0.02 to 0.2 link densities [F(1,60) = 4.00; P = 0.05]. The histogram displays group differences at 0.1 link density. (G) SBPp showed higher incidence of functional connections in the dorsal medial PFC–amygdala–nucleus accumbens module [F(1,60) = 8.00; P = 0.006 repeated measure ANOVA], for visits 0–56 weeks at 0.1 link density. This functional connectivity, however, was not different between SBPp and SBPr subsamples at Week 156, and the scatter plot showed no within subject relationship between Weeks 0 and 156 functional connections. * P 5 0.05 post hoc comparisons between SBPp and SBPp. P = 0.08. Number of subjects is shown in parentheses. Error bars indicate standard error of the mean (SEM). 8 | BRAIN 2016: Page 8 of 13 E. Vachon-Presseau et al. byguestonMay9,2016http://brain.oxfordjournals.org/Downloadedfrom
  • 9. rs678849 SNP was also related to SBP groups at Week 56 (Supplementary Fig. 12 and Supplementary Table 12). These findings provide suggestive evidence that the OPRD1 polymorphism is a genetic candidate for mediating corticolimbic-based risk of chronic pain. Modelling risk and symptom severity for chronic pain We performed path analysis to unravel the relationships between the identified gene, pain at time of entry into the study, and corticolimbic risk factors for predicting chronic pain (Fig. 4). The three primary identified risk factors, each of which predicted SBPp/r groupings at 1 year based on measures collected at study entry, explained 60% of the variance for pain chronification (the explained variance would be closer to 90% if only right amygdala volume was used in the model, yet the model using bilateral amyg- dala volume should be more robust as this measure is replicated using different segmentation tools), and the gene–chronic pain relationship was fully mediated through the indirect effect of the amygdala volume, accounting for 4% of the total variance (Fig. 4A). Although back pain intensity measured at entry in the study did not influence risk factor for chronic pain, it was the only parameter of the risk factors that explained 30% and 38% of the variance in pain intensity at 1 year and at 3 years, respectively (Fig. 4B). Restricting the regres- sion model only to SBPp subjects, back pain at the time of entry into the study was the only parameter that explained 76% and 51% of the variance of back pain at 1 year and 3 years, respectively. These results imply that although the injury or inciting event-related back pain at time of entry into the study does not influence SBP groupings, initial Figure 3 Amygdala and hippocampus volumes are risk factors for chronic pain. (A and B) Subcortical structures were segmented, corresponding volumes summed across hemispheres, and compared between groups as a function of time (Supplementary Tables 4 and 5). Heat maps display overlap of automated segmentation across SBPp, SBPr, and healthy controls at week 0. (C and D) Repeated measure ANCOVAs (group and time effects; covariate for age, gender, and grey matter volume) revealed that SBPp showed smaller amygdala [F(2,63) = 7.94; P 5 0.001] and hippocampus [F(2,70) = 6.35; P = 0.003], but no main effect of time (P-values 4 0.69) or group by time interaction (P-values4 0.23). Bar graphs illustrate group differences at 3 years. Scatterplots show volume reproducibility over 3 years across all SBP. * P 5 0.05 SBPp versus SBPr, and SBPp versus healthy controls. (E and F) Age-matched chronic back pain patients showed significant smaller amygdala [t(1,42) = 2.71; P = 0.009 two-sided unpaired t-tests] and hippocampus [t(1,43) = 1.99; P = 0.05 two-sided unpaired t-tests] than SBPr, matching the volumes observed in SBPp. Similarly, osteoarthritis patients displayed at least marginally smaller amygdala [t(1,52) = 2.21; P = 0.03 two-sided unpaired t-tests] and hippocampus [t(1,52) = 1.95; P = 0.057 two-sided unpaired t-tests] when compared to age matched healthy controls. * P 5 0.05, P 5 0.06. (G) Vertex-wise shape analyses of the amygdala and the hippocampus indicated that SBPp had thinner right amygdala across all visits. Number of subjects is shown in parentheses. Error bars indicate SEM. Anatomical risk factors for chronic pain BRAIN 2016: Page 9 of 13 | 9 byguestonMay9,2016http://brain.oxfordjournals.org/Downloadedfrom
  • 10. back pain intensity has a large influence on symptom se- verity, especially in subjects where the pain persists. Influence of negative affect on risk for chronic pain Epidemiological studies indicate high prevalence of depres- sion and negative affect in chronic pain patients, and pro- pose these conditions reciprocally aggravate each other (Gerrits et al., 2015). Our data showed that neither nega- tive affect nor depression changed over 3 years in SBP (Supplementary Table 1), which at least in this cohort dis- counts the influence of the development of chronic pain on negative moods. The converse influence of mood on the development of chronic pain remains a possibility. We ad- dressed this issue by testing whether depression, and nega- tive affect, influence future pain outcome through the mediating effects of uncovered corticolimbic risk factors, using path analysis. We observed that the density of con- nections within dorsal medial PFC–amygdala–nucleus accumbens white matter network mediated an indirect effect of depression on transition to chronic pain (account- ing for 5% of total variance), with additional independent contributions of the volume of bilateral total amygdala volume and connections within dorsal medial PFC– amygdala–nucleus accumbens functional connectivity net- work (Supplementary Fig. 13). A similar mediation path was observed for negative affect (Supplementary Fig. 13). It is noteworthy that SBP were only mildly depressed, yet their negative mood influenced pain chronification through white matter connectivity of the medial dorsal PFC–amyg- dala–nucleus accumbens module. Maladaptive genotype- driven neural responsiveness may pave the way to develop- ment of chronic pain. As the dorsal medial PFC–amygdala– nucleus accumbens white matter network path is also influ- enced by the OPRM1 rs1799971, we here uncover a brain–gene relationship for the influence of mood on devel- opment of chronic pain. Discussion The present study tested the role of the corticolimbic system in development of chronic back pain. Earlier analyses of subsamples of these data (Baliki et al., 2012; Mansour et al., 2013) show that functional and white matter proper- ties of some limbic regions impart risk for chronic pain. Yet identification of these factors was based on whole-brain contrasts. Here we used a completely unbiased approach, with around double the number of subjects, and tracked properties of identified risk factors for 3 years. Whole-brain multi-resolution white matter network comparisons at study entry indicated that structural connections predispos- ing the development of chronic pain at 1 year were located outside pain-related regions but within the corticolimbic brain. Higher-resolution examination of the corticolimbic white matter network identified three separate modules, where only the dorsal medial PFC–amygdala–nucleus accumbens module differentiated between SBP groups based on both white matter and functional connectivity. The white matter network connectivity of dorsal medial PFC–amygdala–nucleus accumbens remained constant over 3 years, but its functional network connectivity dissi- pated by 3 years relative to values at time 0. Both amyg- dala and hippocampus volumes differentiated between SBP groups, remained invariant over 3 years, and robustly dis- tinguished between SBPp and SBPr. The present results identify three segregated white matter network connectivity-based corticolimbic circuits, with only the dorsal medial PFC–amygdala–nucleus accumbens net- work contributing to risk for chronic pain. What are the functional properties of this module? The amygdala subdiv- isions of each mesolimbic module show correspondence with its function-based and cytoarchitecture-based parcella- tion (Bzdok et al., 2013). The portion of the amygdala incorporated within dorsal medial PFC–amygdala–nucleus accumbens network matched best to the basolateral nu- cleus, which functionally shows preferential involvement in sensory input processing (Bzdok et al., 2013). On the other hand, the corticolimbic circuits also subdivided the prefrontal cortex into dorsal, ventral and orbital subdiv- isions. These divisions may be viewed equivalent to the Figure 4 Corticolimbic characteristics determine risk for chronic pain but symptom severity depends only on initial pain intensity. (A) Path analysis was used to identify the contri- bution of rs678849 SNP, pain at Week 0, and brain parameters on risk for developing chronic pain (SBPp/r at Week 56). The white matter network (White matter) and functional connectivity net- work (Functional) connections of dorsal medial PFC–amygdala–nu- cleus accumbens (mPFCdorsal-amy-NAc) module and bilateral total amygdala volume, at Week 0, were independent predictors of pain persistence or recovery (SBPp/r) at Week 56 (all P-values 4 0.01). The relationship between OPRD1 rs678849 and pain persistence/ recovery was fully mediated through the bilateral amygdala volume (indirect pathway 95% confidence interval: À0.021 and À0.173; R2 = 0.04 of unique variance). Covariance coverage within and be- tween variables exceeded 88%. Numbers of subjects are shown in parentheses. (B) Scatterplots show that pain intensity at Week 0 significantly correlated with pain intensity and pain disability at Week 156 (P-values 4 0.001). 10 | BRAIN 2016: Page 10 of 13 E. Vachon-Presseau et al. byguestonMay9,2016http://brain.oxfordjournals.org/Downloadedfrom
  • 11. rodent medial frontal cortex with respective correspond- ences with pre-limbic, infra-limbic, and orbital cortices. Within this perspective, the cortical component of the dorsal medial PFC–amygdala–nucleus accumbens network becomes analogous to the rodent pre-limbic cortex. In the rodent, pre-limbic is necessary for expression of acquired fear but not for innate fear (Corcoran and Quirk, 2007), which parallels the evidence that human medial PFC repre- sents intensity of chronic but not acute back pain (Hashmi et al., 2013). In rodent models of persistent pain, pre-limbic pyramidal neurons undergo rapid morphological changes (Metz et al., 2009), and amygdala inputs shift the balance between inhibitory and excitatory synaptic transmission in pre-limbic neurons (Ji et al., 2010). Three recent independ- ent studies now also show that activation of pre-limbic neurons diminishes or blocks persistent, but do not influ- ence acute pain behaviour (Lee et al., 2015; Wang et al., 2015; Zhang et al., 2015). One of these reports also dem- onstrates that projections from pre-limbic to nucleus accumbens mediate the relief from persistent pain (Lee et al., 2015). Therefore, the mesolimbic pre-limbic circuitry and the control of persistent pain by pre-limbic activity suggest that the dorsal medial PFC–amygdala–nucleus accumbens module should be considered its human homologue. We recently proposed that, from a behaviour selection viewpoint, pain and negative emotions must be conceptua- lized as a continuum of processes necessary for survival (Baliki and Apkarian, 2015). Current results support the idea by demonstrating that smaller amygdala and hippo- campal volumes, parameters commonly associated with negative affective disorders (Gilbertson et al., 2002), are also predictive of pain chronification. In SBP, amygdala and hippocampus volumes remained invariant over 3 years, and the reduced volumes seen in SBPp matched those observed in patients with chronic pain. Thus, we can generalize these results to chronic negative mood dis- orders where we presume the observed decreased volumes most likely also reflect predispositions. Recent evidence in- dicates that amygdala and hippocampus volumes impact emotional learning and sociability abilities. Healthy sub- jects with larger amygdala volume perform better in fear acquisition (Winkelmann et al., 2015), while subjects with larger hippocampus show higher fear contingency aware- ness (Cacciaglia et al., 2015) and more accurate discrimin- ation of contextual fear (Pohlack et al., 2012). These limbic volume-related differential response characteristics may, therefore, represent personality traits predisposing subjects to both chronic pain and negative mood disorders. We observed that the OPRD1 rs678849 SNP mediated through amygdala volume risk for chronic pain. Earlier studies associate this gene with cocaine, opiate, and heroin addiction, as well as with smaller regional brain volume (http://www.snpedia.com/index.php/Rs678849). This genetic evidence is consistent and complimentary to the role of dorsal medial PFC–amygdala–nucleus accum- bens module in determining risk for chronic pain, as the latter module is also commonly implicated in addictive pathologies. Collectively these observations argue for chronic pain being a maladaptive response, contingent on corticolimbic properties that render the brain addicted to pain. We, however, emphasize the important caveat that the gene–brain–behaviour associations illustrated here are based on sampling few SNPs in a small number of subjects. They must be viewed as proof-of-concept exemplars, and larger and multi-centred collaborative gene–brain studies are necessary to unravel genes critical in the transition to chronic pain. In conclusion, these results challenge long-standing views of the science of pain. Traditionally, the field has empha- sized the inciting injury and related nociceptive spinal pro- cesses as primary culprits of pain chronification. Instead, here we establish that the gross anatomical properties of the corticolimbic brain, not the initial back pain, determine most of the risk for developing chronic pain. As the ana- tomical risk factors were stable across 3 years, they were presumably hardwired and present prior to the event initi- ating back pain. These results pave the way for the devel- opment of novel and distinct approaches to prevention and treatment of chronic pain. Acknowledgements We thank all patients and volunteers that participated in the study, and members of Apkarian lab for technical sup- port and helpful suggestions. Funding This work was supported by funds from the National Institute of Neurological Disorders and Stroke (NS035115, A.V.A.). E.V.P. and P.T. were funded from Canadian Institutes of Health Research (CIHR), E.C. was funded from Swedish Council for Working Life and Social Research (FAS: 2011-0627) and Uppsala University. M.N.B. was partially funded by an anonymous foundation. Supplementary material Supplementary material is available at Brain online. References Achard S, Delon-Martin C, Vertes PE, Renard F, Schenck M, Schneider F, et al. Hubs of brain functional networks are radically reorganized in comatose patients. Proc Natl Acad Sci USA 2012; 109: 20608–13. Apkarian AV, Sosa Y, Sonty S, Levy RE, Harden RN, Parrish TB, et al. Chronic back pain is associated with decreased prefrontal and thalamic gray matter density. J Neurosci 2004; 24: 10410–5. Baliki MN, Apkarian AV. Nociception, pain, negative moods, and behavior selection. Neuron 2015; 87: 474–91. Anatomical risk factors for chronic pain BRAIN 2016: Page 11 of 13 | 11 byguestonMay9,2016http://brain.oxfordjournals.org/Downloadedfrom
  • 12. Baliki MN, Chang PC, Baria AT, Centeno MV, Apkarian AV. Resting-sate functional reorganization of the rat limbic system fol- lowing neuropathic injury. Sci Rep 2014; 4: 6186. Baliki MN, Chialvo DR, Geha PY, Levy RM, Harden RN, Parrish TB, et al. Chronic pain and the emotional brain: specific brain activity associated with spontaneous fluctuations of intensity of chronic back pain. J Neurosci 2006; 26: 12165–73. Baliki MN, Geha PY, Fields HL, Apkarian AV. Predicting value of pain and analgesia: nucleus accumbens response to noxious stimuli changes in the presence of chronic pain. Neuron 2010; 66: 149–60. Baliki MN, Petre B, Torbey S, Herrmann KM, Huang L, Schnitzer TJ, et al. Corticostriatal functional connectivity predicts transition to chronic back pain. Nat Neurosci 2012; 15: 117–19. Beck AT, Ward CH, Mendelson M, Mock J, Erbaugh J. An inventory for measuring depression. Arch Gen Psychiatry 1961; 4: 561–71. Behrens TE, Berg HJ, Jbabdi S, Rushworth MF, Woolrich MW. Probabilistic diffusion tractography with multiple fibre orientations: what can we gain? Neuroimage 2007; 34: 144–55. Behrens TE, Woolrich MW, Jenkinson M, Johansen-Berg H, Nunes RG, Clare S, et al. Characterization and propagation of uncertainty in diffusion-weighted MR imaging. Magn Reson Med 2003; 50: 1077–88. Bzdok D, Laird AR, Zilles K, Fox PT, Eickhoff SB. An investigation of the structural, connectional, and functional subspecialization in the human amygdala. Hum Brain Mapp 2013; 34: 3247–66. Cacciaglia R, Pohlack ST, Flor H, Nees F. Dissociable roles for hip- pocampal and amygdalar volume in human fear conditioning. Brain Struct Funct 2015; 220: 2575–86. Chang PC, Pollema-Mays SL, Centeno MV, Procissi D, Contini M, Baria AT, et al. Role of nucleus accumbens in neuropathic pain: linked multi-scale evidence in the rat transitioning to neuropathic pain. Pain 2014; 155: 1128–39. Corcoran KA, Quirk GJ. Activity in prelimbic cortex is necessary for the expression of learned, but not innate, fears. J Neurosci 2007; 27: 840–4. Drevets WC, Price JL, Furey ML. Brain structural and functional abnormalities in mood disorders: implications for neurocircuitry models of depression. Brain Struct Funct 2008; 213: 93–118. Ferreira PH, Beckenkamp P, Maher CG, Hopper JL, Ferreira ML. Nature or nurture in low back pain? Results of a systematic review of studies based on twin samples. Eur J Pain 2013; 17: 957–71. Fischl B, Salat DH, Busa E, Albert M, Dieterich M, Haselgrove C, et al. Whole brain segmentation: automated labeling of neuroana- tomical structures in the human brain. Neuron 2002; 33: 341–55. Geha PY, Baliki MN, Harden RN, Bauer WR, Parrish TB, Apkarian AV. The brain in chronic CRPS pain: abnormal gray-white matter interactions in emotional and autonomic regions. Neuron 2008; 60: 570–81. Gerrits MM, van Marwijk HW, van Oppen P, van der Horst H, Penninx BW. Longitudinal association between pain, and depression and anxiety over four years. J Psychosom Res 2015; 78: 64–70. Gilbertson MW, Shenton ME, Ciszewski A, Kasai K, Lasko NB, Orr SP, et al. Smaller hippocampal volume predicts pathologic vulner- ability to psychological trauma. Nat Neurosci 2002; 5: 1242–7. Hashmi JA, Baliki MN, Huang L, Baria AT, Torbey S, Hermann KM, et al. Shape shifting pain: chronification of back pain shifts brain representation from nociceptive to emotional circuits. Brain 2013; 136(Pt 9): 2751–68. Hibar DP, Stein JL, Renteria ME, Arias-Vasquez A, Desrivieres S, Jahanshad N, et al. Common genetic variants influence human sub- cortical brain structures. Nature 2015; 520: 224–9. Jenkinson M, Bannister P, Brady M, Smith S. Improved optimization for the robust and accurate linear registration and motion correction of brain images. Neuroimage 2002; 17: 825–41. Ji G, Neugebauer V. Pain-related deactivation of medial prefrontal cortical neurons involves mGluR1 and GABA(A) receptors. J Neurophysiol 2011; 106: 2642–52. Ji G, Sun H, Fu Y, Li Z, Pais-Vieira M, Galhardo V, et al. Cognitive impairment in pain through amygdala-driven prefrontal cortical de- activation. J Neurosci 2010; 30: 5451–64. Lee M, Manders TR, Eberle SE, Su C, D’Amour J, Yang R, et al. Activation of corticostriatal circuitry relieves chronic neuropathic pain. J Neurosci 2015; 35: 5247–59. Mackinnon DP, Lockwood CM, Williams J. Confidence limits for the indirect effect: distribution of the product and resampling methods. Multivariate Behav Res 2004; 39: 99. MacLean PD. The limbic system (“visceral brain”) in relation to the central gray and reticulum of the brain stem.Psychosom Med 1955; 17: 355–66. Mansour AR, Baliki MN, Huang L, Torbey S, Herrmann KM, Schnitzer TJ, et al. Brain white matter structural properties predict transition to chronic pain. Pain 2013; 154: 2160–8. Medicine Io. Relieving pain in America: a blueprint for transforming prevention, care, education, and research. 2011. Available from: http://www.iom.edu. Melzack R. The McGill Pain Questionnaire: major properties and scoring methods. Pain 1975; 1: 277–99. Melzack R, Casey K. Sensory, motivational, and central contol deter- minants of pain. The skin senses. Springfield: Charles C. Thomas; 1968. p. 423–43. Melzack R, Wall PD. Pain mechanisms: a new theory. Science 1965; 150: 971–9. Metz AE, Yau HJ, Centeno MV, Apkarian AV, Martina M. Morphological and functional reorganization of rat medial pre- frontal cortex in neuropathic pain. Proc Natl Acad Sci USA 2009; 106: 2423–8. Murray CJ, Lopez AD. Measuring the global burden of disease. N Engl J Med 2013; 369: 448–57. Mutso AA, Petre B, Huang L, Baliki MN, Torbey S, Herrmann K, et al. Reorganization of hippocampal functional connectivity with transition to chronic back pain. J Neurophysiol 2014; 111: 1065–76. Mutso AA, Radzicki D, Baliki MN, Huang L, Banisadr G, Centeno MV, et al. Abnormalities in hippocampal functioning with persistent pain. J Neurosci 2012; 32: 5747–56. Neugebauer V, Li W, Bird GC, Bhave G, Gereau RW. Synaptic plas- ticity in the amygdala in a model of arthritic pain: differential roles of metabotropic glutamate receptors 1 and 5. J Neurosci 2003; 23: 52–63. Nugent AC, Luckenbaugh DA, Wood SE, Bogers W, Zarate CA, Jr., Drevets WC. Automated subcortical segmentation using FIRST: test- retest reliability, interscanner reliability, and comparison to manual segmentation. Hum Brain Mapp 2013; 34: 2313–29. Patenaude B, Smith SM, Kennedy DN, Jenkinson M. A Bayesian model of shape and appearance for subcortical brain segmentation. Neuroimage 2011; 56: 907–22. Petre B, Torbey S, Griffith JW, De Oliveira G, Herrmann K, Mansour A, et al. Smoking increases risk of pain chronification through shared corticostriatal circuitry. Hum Brain Mapp 2015; 36: 683–94. Phelps EA, LeDoux JE. Contributions of the amygdala to emotion processing: from animal models to human behavior. Neuron 2005; 48: 175–87. Pohlack ST, Nees F, Liebscher C, Cacciaglia R, Diener SJ, Ridder S, et al. Hippocampal but not amygdalar volume affects contextual fear conditioning in humans. Hum Brain Mapp 2012; 33: 478–88. Raz M. The painless brain: lobotomy, psychiatry, and the treatment of chronic pain and terminal illness. Perspect Biol Med 2009; 52: 555–65. Ren WJ, Liu Y, Zhou LJ, Li W, Zhong Y, Pang RP, et al. Peripheral nerve injury leads to working memory deficits and dysfunction of the hippocampus by upregulation of TNF-alpha in rodents. Neuropsychopharmacology 2011; 36: 979–92. Rose EJ, Donohoe G. Brain vs behavior: an effect size comparison of neuroimaging and cognitive studies of genetic risk for schizophrenia. Schizophr Bull 2013; 39: 518–26. 12 | BRAIN 2016: Page 12 of 13 E. Vachon-Presseau et al. byguestonMay9,2016http://brain.oxfordjournals.org/Downloadedfrom
  • 13. Rubinov M, Sporns O. Complex network measures of brain connect- ivity: uses and interpretations. Neuroimage 2010; 52: 1059–69. Russo SJ, Murrough JW, Han MH, Charney DS, Nestler EJ. Neurobiology of resilience. Nat Neurosci 2012; 15: 1475–84. Schwartz N, Temkin P, Jurado S, Lim BK, Heifets BD, Polepalli JS, et al. Chronic pain. Decreased motivation during chronic pain re- quires long-term depression in the nucleus accumbens. Science 2014; 345: 535–42. Smith SM, Zhang Y, Jenkinson M, Chen J, Matthews PM, Federico A, et al. Accurate, robust, and automated longitudinal and cross-sec- tional brain change analysis. Neuroimage 2002; 17: 479–89. Tait RC, Chibnall JT, Krause S. The pain disability index: psychomet- ric properties. Pain 1990; 40: 171–82. Vachon-Presseau E, Roy M, Martel MO, Caron E, Marin MF, Chen J, et al. The stress model of chronic pain: evidence from basal cortisol and hippocampal structure and function in humans. Brain 2013; 136(Pt 3): 815–27. Wang GQ, Cen C, Li C, Cao S, Wang N, Zhou Z, et al. Deactivation of excitatory neurons in the prelimbic cortex via Cdk5 promotes pain sensation and anxiety. Nat Commun 2015; 6: 7660. Watson D, Clark LA. The PANAS-X manual for the positive and negative affect schedule–Expanded form. University of Iowa. 1999. Winkelmann T, Grimm O, Pohlack ST, Nees F, Cacciaglia R, Dinu- Biringer R, et al. Brain morphology correlates of interindividual differences in conditioned fear acquisition and extinction learning. Brain Struct Funct 2015, in press. Zalesky A, Fornito A, Bullmore ET. Network-based statistic: iden- tifying differences in brain networks. Neuroimage 2010; 53: 1197–207. Zhang Z, Gadotti VM, Chen L, Souza IA, Stemkowski PL, Zamponi GW. Role of prelimbic GABAergic circuits in sensory and emotional aspects of neuropathic pain. Cell Rep 2015; 12: 752–9. Anatomical risk factors for chronic pain BRAIN 2016: Page 13 of 13 | 13 byguestonMay9,2016http://brain.oxfordjournals.org/Downloadedfrom