You are on page 1of 15

NeuroImage 62 (2012) 2281–2295

Contents lists available at SciVerse ScienceDirect

NeuroImage
journal homepage: www.elsevier.com/locate/ynimg

Review

Resting state functional connectivity in addiction: Lessons learned and a road ahead
Matthew T. Sutherland, Meredith J. McHugh, Vani Pariyadath, Elliot A. Stein ⁎
Neuroimaging Research Branch, National Institute on Drug Abuse, Intramural Research Program, NIH/DHHS, Baltimore, MD, USA

a r t i c l e i n f o a b s t r a c t

Article history: Despite intensive scientific investigation and public health imperatives, drug addiction treatment out-
Received 10 August 2011 comes have not significantly improved in more than 50 years. Non-invasive brain imaging has, over the
Revised 22 December 2011 past several decades, contributed important new insights into the neuroplastic adaptations that result
Accepted 24 January 2012
from chronic drug intake, but additional experimental approaches and neurobiological hypotheses are
Available online 1 February 2012
needed to better capture the totality of the motivational, affective, cognitive, genetic and pharmacological
Keywords:
complexities of the disease. Recent advances in assessing network dynamics through resting-state func-
fMRI tional connectivity (rsFC) may allow for such systems-level assessments. In this review, we first summa-
Connectivity rize the nascent addiction-related rsFC literature and suggest that in using this tool, circuit connectivity
Drug abuse may inform specific neurobiological substrates underlying psychological dysfunctions associated with re-
Nicotine ward, affective and cognitive processing often observed in drug addicts. Using nicotine addiction as an ex-
Insula emplar, we subsequently provide a heuristic framework to guide future research by linking recent findings
Default mode from intrinsic network connectivity studies with those interrogating nicotine's neuropharmacological ac-
Attention
tions. Emerging evidence supports a critical role for the insula in nicotine addiction. Likewise, the anterior
insula, potentially together with the anterior cingulate cortex, appears to pivotally influence the dynamics
between large-scale brain networks subserving internal (default-mode network) and external (executive
control network) information processing. We suggest that a better understanding of how the insula mod-
ulates the interaction between these networks is critical for elucidating both the cognitive impairments
often associated with withdrawal and the performance-enhancing effects of nicotine administration.
Such an understanding may be usefully applied in the design and development of novel smoking cessation
treatments.
Published by Elsevier Inc.

Contents

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2281
Addiction-related rsFC studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2282
Reward dysregulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2282
Emotional dysregulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2284
Cognitive dysregulation. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2284
Disease severity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2285
Nicotine and large-scale networks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2286
Insula, network switching and interoception . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2289
A network model of nicotine addiction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2290
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2293
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2293

Introduction

Drug addiction is a multifaceted neuropsychiatric disorder char-


acterized by the compulsive seeking and taking of a drug, despite
⁎ Corresponding author at: Neuroimaging Research Branch, National Institute on
Drug Abuse, 251 Bayview Blvd, Suite 200, Baltimore, MD 21224, USA. Fax: + 1 443
the high likelihood of negative consequences. Addiction is notable
740 2734. for the complex, and still only partially understood, interactions be-
E-mail address: estein@mail.nih.gov (E.A. Stein). tween neurobiological, environmental, pharmacological, and genetic

1053-8119/$ – see front matter. Published by Elsevier Inc.


doi:10.1016/j.neuroimage.2012.01.117
2282 M.T. Sutherland et al. / NeuroImage 62 (2012) 2281–2295

components. Multiple theories regarding the underlying mecha- in the last few years that rsFC has begun to be considered as a character-
nisms of addiction have been proposed (e.g., Everitt and Robbins, ization and potentially diagnostic tool.
2005; Goldstein and Volkow, 2002; Koob and Le Moal, 2008; Koob To date, rsFC has been applied in only a handful of drug addiction-
and Volkow, 2010; Redish, 2004; Wise, 2008), and although original- related studies and the current review begins with an overview of
ly generated mostly from rodent studies, many, but not all, of these those findings. As expected in an emerging field, many of these stud-
neurobiological mechanisms appear well conserved in non-human ies have been somewhat exploratory in nature, leading to conflicting
primates and, where studied, in humans. However, despite tremen- findings. Applications of this tool in addiction research may therefore
dous advances over the past several decades, translating preclinical benefit from experimental approaches grounded by stronger a priori
findings of the molecular and cellular neuroadaptations following theoretical models. To this end, throughout the review we relate ex-
chronic drug intake has not as yet significantly improved clinical tant addiction-related rsFC findings to a larger corpus of neuroimag-
outcomes. As drug abuse is a uniquely human disease, it would ing research from healthy controls and other neuropsychiatric
seem that a better understanding of the profound disruption of mo- conditions. In the latter half of the review we continue this theme
tivational, affective and cognitive processes following, and/or predis- and in greater detail link recent findings from network connectivity
posing individuals to drug use is critical to expedite treatment and nicotine-related neuroimaging studies to develop a heuristic
development. framework yielding empirically testable hypotheses regarding the ef-
While neuroimaging cognitive-subtraction task paradigms have fects of both nicotine abstinence and drug administration on brain
been essential for delineating the acute effects of addictive drugs and behavior.
and the long-term consequences of use within circumscribed brain
areas (e.g., Bickel et al., 2007; Breiter et al., 1997; Diekhof et al., Addiction-related rsFC studies
2008; Kumari et al., 2003; Stein et al., 1998), additional, complemen-
tary insight will most likely be obtained by considering alterations in The earliest addiction-related rsFC study reported marked reduc-
circuit-level interactions between brain regions (Koob and Volkow, tions in connectivity within the primary visual and motor cortices
2010). The last decade has witnessed an explosion in the study of after cocaine administration to addicted individuals, presumably
functional connectivity using fMRI, largely because it allows for the reflecting changes in coherent neuronal firing patterns (Li et al.,
exploration of large-scale networks and their interactions, thus mov- 2000). Subsequently, using a functional connectivity analysis on
ing towards a systems-level understanding of brain functioning [ 15O] PET data, Daglish et al. (2003) identified two networks during
(Bressler and Menon, 2010; van den Heuvel and Hulshoff Pol, opiate craving, one including anterior cingulate cortex (ACC) and
2010). By extension, resting-state functional connectivity (rsFC) temporal cortex and a second involving orbitofrontal cortex (OFC),
may allow for the identification of neural circuitry dysfunction under- parietal cortex, and insula, suggesting engagement of motivational
lying various neuropsychiatric disorders (Fox and Greicius, 2010). and attentional circuits. These networks are similar to those seen fol-
Among its advantages as a potential characterization and diagnostic lowing cocaine cue provocation (Garavan et al., 2000), again suggest-
tool are that: 1) identified rsFC circuit-alterations are less likely to ing that circuits identified at rest reflect those engaged during active
be confounded by subtle differences in specific task-based experi- task performance. More recent rsFC endeavors in drug addiction
mental paradigms; 2) networks so identified appear to be consistent have interrogated altered connectivity between specific regions of in-
across time within and between individuals (Chen et al., 2008); 3) terest (i.e., “seed” regions) and their interconnected brain regions. As
such networks appear to reflect the entirety of the cognitive elements a broad organization principle, we have grouped findings from these
necessary for task processing (Smith et al., 2009); and 4) relative to initial studies according to general psychological constructs often as-
task-based fMRI methods, data collection is relatively quick and sociated with impairment in drug addiction: 1) reward dysregulation,
straight-forward — a useful quality when assessing patient 2) emotional dysregulation, and 3) cognitive dysregulation.
populations with variable constraints on attentional, executive, and
motor control. There are, however, certain methodological issues Reward dysregulation
associated with rsFC-based techniques, a few of which will be briefly
discussed at points throughout the review. Nonetheless, this emerg- Large and rapid dopamine increases in the mesocorticolimbic
ing neuroimaging tool has provided researchers with additional in- (MCL) system are thought to underlie the initial reinforcing effects
sights and spurred novel theories about the underlying neural of abused drugs (Nestler, 2005). As such, preclinical and clinical stud-
substrates of various neuropsychiatric disorders (e.g., Menon, 2011). ies have generally focused on neuroadaptations in midbrain dopami-
Circuits synchronized “at rest” are constrained to known, direct or nergic areas (e.g., ventral tegmental area, substantia nigra pars
polysynaptic anatomically interconnected regions (Damoiseaux and compacta) and the structures to which they project (e.g., the nucleus
Greicius, 2009; Greicius et al., 2009; Honey et al., 2009), thus constitut- accumbens [NAc] of the ventral striatum), following an extended his-
ing plausible functional networks. Critically, the strength of these net- tory of drug administration (Everitt and Robbins, 2005; Koob and
works “at rest” predicts both behavioral performance and subsequent Volkow, 2010). While understanding of the molecular and cellular
activation of the same brain areas during task performance (e.g., drug-induced changes within constituent components of the MCL
Hampson et al., 2006; Kelly et al., 2008; Kim et al., 2009; Seeley et al., system has advanced (Morón and Green, 2010), much less is known
2007; Tambini et al., 2010). Further, there is growing evidence that al- about the circuit-level manifestations of such regional alterations
terations in rsFC strength can potentially be used to assess various neu- when considering interactions among and between MCL regions
ropsychiatric disease trajectories, and where available treatment and other subcortical and cortical structures. Insofar as drug addiction
outcomes, including: Alzheimer's disease (Greicius et al., 2004; Lustig has historically been viewed as a dopaminergic dysregulation disor-
et al., 2003; Rombouts et al., 2005; Wang et al., 2006), autism der (Di Chiara et al., 2004; Wise, 2008), rsFC investigations have
(Kennedy et al., 2006), depression (Anand et al., 2005), multiple sclero- begun to interrogate the MCL system in the service of elucidating
sis (Lowe et al., 2002), and attention deficit/hyperactive disorder circuit-level alterations associated with reward deficits in the
(ADHD; Tian et al., 2006). Emerging evidence further suggests a genetic human addict.
linkage between rsFC networks and various behavioral phenotypes Alterations in rsFC strength between ventral striatum and vari-
(Glahn et al., 2010; Meyer-Lindenberg, 2009), raising promise that ous subcortical and cortical regions have been observed when com-
functional connectivity may serve as a systems-level biomarker to iden- paring cocaine (Gu et al., 2010; Tomasi et al., 2010; Wilcox et al.,
tify individual differences in and provide differential disease diagnoses 2011), prescription opioid (Upadhyay et al., 2010), and heroin de-
for various neuropsychiatric disorders. Despite such promise, it is only pendent individuals (Ma et al., 2010) with matched, non-drug
M.T. Sutherland et al. / NeuroImage 62 (2012) 2281–2295 2283

using controls. Although altered MCL circuitry has been consistently observed a general decrease in rsFC between most regions within
observed in drug addicts, synthesis of a cogent narrative surround- the MCL reward pathway and interconnected brain areas (with the
ing the precise circuits and direction of change is difficult from notable exception of the NAc, whose connectivity remained
these initial studies. While, on the one hand, some studies have unchanged between groups) (Fig. 1A). Such widespread reductions
reported increased rsFC between MCL regions and subcortical and in the connectivity of multiple MCL system components may reflect
cortical areas (Ma et al., 2010; Wilcox et al., 2011), others have putative difficulty in appropriately engaging reward, motivational,
reported decreased connectivity (Gu et al., 2010; Tomasi et al., and emotional circuitry, which is consistent with perspectives sug-
2010; Upadhyay et al., 2010; Wang et al., 2010). For example, test- gesting that the transition from drug-use to addiction is driven by re-
ing the hypothesis that MCL circuits are altered in heroin addicts, duced functioning of reward systems, with concurrently increased
Ma et al. (2010) noted stronger rsFC between NAc and the ventral activation of ‘anti-reward’ systems (Koob and Le Moal, 1997, 2005).
aspects of medial prefrontal cortex (including rostral ACC and medi- Tomasi et al. (2010) arrived at similar conclusions when observing
al OFC). Although using a small (n = 14) and a heterogeneous sub- lower connectivity between midbrain dopaminergic regions and me-
ject group (i.e., both methadone maintained and abstinent users), dial PFC regions in cocaine abusers (n = 20) relative to healthy con-
these results suggest enhanced connectivity within reward and mo- trols performing a sustained attention task.
tivation circuits that may be interpreted in the perspective of al- While clearly more work is needed to untangle the nature of rsFC
tered incentive salience for drugs and drug-associated stimuli changes in reward-related circuitry, a general pattern of perturbed
(Berridge and Robinson, 1998). Similarly, Wilcox et al. (2011) ob- connectivity across heterogeneous drug-using cohorts is consistent
served increased rsFC between ventral striatum and ventromedial with a reward dysregulation hypothesis of drug addiction. That said,
PFC (vmPFC) regions in abstinent cocaine-users. any effort to draw conclusions at this early stage must be tempered
In apparent contrast to the above findings suggesting increased by consideration of the various methodological issues inherent to
striatal-PFC rsFC strength, a widespread reduction in connectivity be- this literature. For example, relatively small samples sizes (Ma et al.,
tween NAc and various subcortical (hippocampus and amygdala) and 2010; Upadhyay et al., 2010; Wilcox et al., 2011) necessitate caution
cortical (parietal, cingulate, prefrontal) regions in prescription opioid when interpreting findings. Additionally, the duration since last
addicts (n = 10) has been described (Upadhyay et al., 2010). Individ- drug use and collection of imaging data has often not been adequately
uals in that study were current users, thus results may in part reflect considered. Factors such as acute withdrawal (Gu et al., 2010; Ma et
effects of recent opiate use. Nonetheless, a similar pattern of MCL cir- al., 2010; Tomasi et al., 2010; Wilcox et al., 2011) or acute drug effects
cuit reductions was reported by Gu et al. (2010) who conducted rsFC (Ma et al., 2010; Upadhyay et al., 2010) likely contribute significantly
assessments in a relatively large population (n = 39) of active to variance both within and between rsFC studies, confounding inter-
cocaine-users and a matched, non-using control group. They pretation of results. Finally, generalizing across different abused

Fig. 1. (A) Summary of regions showing reduced rsFC strength with MCL seed regions in cocaine-users relative to matched non-using controls. (B) Difference map illustrating re-
duced rsFC between an amygdala seed and medial PFC/rostral ACC regions in cocaine-users relative to matched non-using controls. (C) Top displays a difference map showing
stronger interhemispheric connectivity between lateral PFC regions among non-using controls relative to cocaine-users. Bottom shows self-reported cognitive failures as a function
of reduced lateral PFC interhemispheric connectivity.
Panel A is reproduced from Gu et al. (2010). Panel B is reproduced from Gu et al. (2010). Panel C is reproduced from Kelly et al. (2011).
2284 M.T. Sutherland et al. / NeuroImage 62 (2012) 2281–2295

drugs is complicated by the potential for drug-specific effects on un- tentatively concluded that more efficient amygdala-vmPFC neurobio-
derlying neural circuitry. In sum, careful consideration of the above logical coupling likely yields beneficial behavioral outcomes in terms
methodological factors will be necessary for future work to elucidate of elevated emotional regulation and reduced anxiety. Initial
the precise nature of rsFC alterations in the reward-related neurocir- addiction-related rsFC studies, combined with evidence from healthy
cuitry of drug addiction. samples and other neuropsychiatric disorders, suggest the intriguing
possibility that abnormalities in functional and structural connectivity
Emotional dysregulation between amygdala and medial PFC may, at least partly, mediate as-
pects of emotional dysregulation often observed in drug dependent
Accompanying alterations in reward-related neurocircuitry, drug- individuals.
induced changes in amygdala-centered ‘anti-reward’ circuits have
been associated with increased anxiety, irritability or aversive Cognitive dysregulation
stress-like states that may mediate negative reinforcement mecha-
nisms perpetuating drug use (Koob and Le Moal, 2005). Amygdala's Drug-addicted individuals are known to exhibit deficits in neural
interactions with medial prefrontal, cingulate, hippocampal, and systems associated with cognitive control (Goldstein et al., 2004;
insula regions are further implicated in processing emotional stimuli, Hester and Garavan, 2004; Hester et al., 2009). Neurobiological
generating affective states, and/or regulating emotion (Pezawas et al., models of cognitive control emphasize a network of regions centered
2005; Phillips et al., 2003; Stein et al., 2007). Working under the hy- on the ACC, lateral PFC and parietal areas. Substantial evidence and
pothesis that amygdala and its interconnected circuitry are critical recent theories suggest that ACC subserves a monitoring role for the
neural substrates mediating continued drug use, preliminary rsFC detection of salient events, particularly erroneous or error-prone ac-
studies suggest that altered functional and structural amygdala-PFC tions (Carter and van Veen, 2007; Ridderinkhof et al., 2004). Upon de-
connectivity may underlie aspects of emotional dysregulation often tection of such salient/erroneous events, the ACC is thought to signal
noted in addicted individuals. the need for the top-down reorientation of attention, implemented
Altered amygdala-centered connectivity has been noted in individ- by lateral PFC and parietal regions (Kerns et al., 2004; King et al.,
uals addicted to multiple pharmacological classes of drugs (Gu et al., 2010; Miller and Cohen, 2001). The top-down influence of lateral
2010; Liu et al., 2009; Upadhyay et al., 2010; Wang et al., 2010; Xie et PFC appears to bias information processing in lower-level sensorimo-
al., 2011). More specifically, Gu et al. (2010) reported decreased rsFC tor cortices towards relevant input (Egner and Hirsch, 2005; King et
strength between amygdala and a region of medial PFC (encompassing al., 2010) for the optimization of goal-directed behavior (Frank et
aspects of vmPFC and rostral ACC) in cocaine addicts (Fig. 1B). Similar al., 2005; Hester et al., 2008; Magno et al., 2006). Performance decre-
decreases in amygdala-vmPFC rsFC strength have also been observed ments on measures of cognitive control in drug addicted individuals
in a sample (n = 15) of active heroin abusers (Wang et al., 2010). (Franken et al., 2010; Hester and Garavan, 2004; Hester et al., 2007,
Widespread reductions in amygdala's connectivity with multiple re- 2009) are associated with reduced functional engagement (Hester
gions, including medial, ventrolateral, and dorsolateral PFC regions, and Garavan, 2004; Hester et al., 2009; Li and Sinha, 2008) and struc-
have also been documented in prescription-opioid addicts (Upadhyay tural integrity across these regions (Barrós-Loscertales et al., 2011;
et al., 2010). Moreover, amygdala-vmPFC (i.e., subgenual ACC) connec- Nakama et al., 2011; Yuan et al., 2009, 2010).
tivity was inversely related to duration of opioid dependence, such that Recent rsFC studies have also suggested patterns of abnormal con-
longer periods of use were associated with greater rsFC reductions nectivity between these primary nodes of a “cognitive control net-
(Upadhyay et al., 2010). Additionally, using diffusion tensor imaging, work” in both heroin (Ma et al., 2010; Yuan et al., 2010) and
these authors also showed that opioid use was associated with reduced cocaine addicted samples (Kelly et al., 2011). For example, reduced
structural integrity of the uncinate fasiculus, the primary white matter rsFC strength between the ACC and dlPFC has been noted in heroin
tract connecting amygdala and medial PFC. Thus, while the number of addicts (Ma et al., 2010). Additionally, in a sample of chronic
studies is still limited, initial observations in drug addicted samples ap- cocaine-using individuals (n = 25), Kelly et al. (2011) observed sig-
pear to be converging on functional and structural connectivity abnor- nificant reductions in rsFC within and between lateral PFC and parie-
malities in an amygdala-medial PFC circuit. tal areas, where reduced interhemispheric connectivity between
Addiction-related alterations between amygdala and medial PFC lateral PFC regions predicted a higher incidence of self-reported cog-
are particularly intriguing when considered in light of a larger corpus nitive failures (Fig. 1C). Yuan et al. (2010) observed a similar pattern
of research relating such circuit-level interactions to emotional regula- in abstinent heroin users such that reduced rsFC between lateral PFC
tion, subjective anxiety, and other neuropsychiatric disorders (Foland- and parietal regions was accompanied by reductions in gray matter
Ross et al., 2010; Hahn et al., 2011; Hariri et al., 2003; Kim et al., 2011a, density in those same regions, with years of use predicting greater re-
2011b; Motzkin et al., 2011; Pezawas et al., 2005). The vmPFC has been ductions across both measures. Reduced rsFC between nodes within
posited to actively suppress amygdala functioning (Foland-Ross et al., this putative cognitive control network is consistent with the behav-
2010; Hariri et al., 2003; Kim et al., 2011b), which in turn, is thought ioral and task-based imaging findings referenced above as well as
to alleviate emotional distress (Berkman and Leiberman, 2009; self-reported cognitive deficits in drug-addicted populations (Ersche
Ochsner et al., 2004). Such a regulation-circuit perspective is bolstered et al., 2006; Gruber et al., 2007; Hester and Garavan, 2004; Kelly et
by rsFC studies demonstrating that amygdala-vmPFC rsFC strength is al., 2011).
inversely related to self-reported anxiety levels in non-clinical samples, Camchong et al. (2011) recently observed increased positive con-
where increased anxiety is associated with reduced connectivity (Kim nectivity between the ACC and dlPFC in a sample of active cocaine
et al., 2011a; Pezawas et al., 2005). Taken further, reduced amygdala- users (n = 27). While the direction of this connectivity change initial-
vmPFC rsFC strength, as well as compromised uncinate fasiculus struc- ly seems counter-intuitive and contradictory to those results dis-
tural integrity, has been observed in neuropsychiatric conditions char- cussed above, greater rsFC in this ACC-dlPFC circuit was in fact
acterized by pathological levels of anxiety (Hahn et al., 2011; Phan et associated with poorer task performance during reversal learning.
al., 2009). Diminished amygdala-vmPFC functional and structural con- Of note, the ACC seed chosen was located rostral to the dorsal ACC re-
nectivity has similarly been noted in a sample of psychopathic versus gion typically associated with situations necessitating elevated cogni-
non-psychopathic criminals (Motzkin et al., 2011), which may offer a tive control (Carter and van Veen, 2007; Ridderinkhof et al., 2004).
neurobiological account of the aberrant emotional and social behaviors Rostral ACC and adjacent vmPFC regions are considered part of a
associated with psychopathy (Blair, 2008). Kim et al. (2011a, 2011b) “task-negative” or default-mode network (DMN) that typically deac-
have reviewed extensive evidence regarding this circuitry and tivates during task performance (Buckner et al., 2008; Gusnard and
M.T. Sutherland et al. / NeuroImage 62 (2012) 2281–2295 2285

Raichle, 2001) and shows negative connectivity with cognitive con- addiction-related circuit alterations, other rsFC studies have just
trol regions during tasks and “at rest” (Fox et al., 2005; Kelly et al., begun to consider the possibility that this approach could be used
2008; Prado and Weissman, 2011). Moreover, altered connectivity as a diagnostic tool to assess individual differences in addiction sever-
between default-mode and cognitive control regions has been ity and, by extension, provide a biomarker for treatment efficacy. To
reported in smokers following nicotine abstinence and linked with this end, Hong et al. (2009) sought to identify neural circuits in ciga-
withdrawal-related cognitive deficits (Cole et al., 2010). Importantly, rette smokers that were modulated as a function of: 1) acute nicotine
Camchong et al. (2011) examined rsFC in current cocaine users, indi- administration, and 2) severity of nicotine addiction. Based on the hy-
viduals likely to be experiencing at least some degree of acute with- pothesis that the cingulate is an integral component of many
drawal, and the enhanced positive connectivity they observed addiction-related deficits, seven bilateral cingulate sub-regions were
between default-mode (rostral ACC) and cognitive control (dlPFC) re- defined and used as seeds in separate rsFC analyses. Two distinct
gions may reflect state changes associated with withdrawal. In the groups of networks were identified. The first consisted of seven
latter sections of this review, we describe in greater detail how mal- cingulate-neocortical pathways that demonstrated enhanced connec-
adaptive interactions within and between these two large-scale net- tivity strength in the presence (versus absence) of an acute nicotine
works following acute abstinence may provide a neurobiologically patch (Fig. 2A), including ACC, parietal and medial superior frontal re-
plausible account of the cognitive deficits often observed in addicted gions. These and other identified circuits are consistent with those
individuals. implicated in nicotine's performance-enhancing properties
(Heishman et al., 2010). However, in a double dissociation fashion,
Disease severity two bilateral dorsal ACC to ventral striatal circuits were identified
whose connectivity strengths were inversely proportional to an indi-
While the preceding sections have highlighted the use of function- vidual's level of nicotine addiction as measured by Fagerström scores,
al connectivity assessments as a tool for the characterization of but were unaltered following nicotine patch administration (Fig. 2B),

Fig. 2. State and trait components of nicotine addiction. (A) Two of the seven resting state networks that showed enhanced connectivity under nicotine relative to placebo admin-
istration in minimally deprived (~ 4.5 h) smokers. Left image shows enhanced rsFC between a dorsal ACC (dACC) seed (blue oval) and a region encompassing the superior parietal
lobe and post-central gyrus. Right image shows enhanced rsFC between a mid cingulate cortex (MCC) seed and a region encompassing the post-central gyrus and inferior parietal
lobe. (B) Negative correlation between scores on the Fagerström Test for Nicotine Dependence (FTND) and rsFC strength in smokers between the right striatum (blue) and a dACC
seed in minimally deprived (gray triangles) and nicotine sated (red diamonds) conditions.
Reproduced from Hong et al. (2009).
2286 M.T. Sutherland et al. / NeuroImage 62 (2012) 2281–2295

suggesting specific circuits related to addiction severity and which, Dissociable large-scale brain networks are thought to subserve
the authors speculate, may serve as a biomarker for studies of treat- both task-relevant and -irrelevant cognitive operations during
ment outcome. attention-demanding tasks. One ensemble, termed the “task-posi-
Subsequently, Hong et al. (2010) went on to demonstrate that a tive” network (TPN; Fox et al., 2005), consists of regions routinely
gene variant of the α5 subunit of nicotinic acetylcholine receptors is showing activity increases during demanding tasks (e.g., lateral pre-
associated with a very similar “addiction related circuit”. Specifically, frontal, lateral parietal, posterior medial PFC, and insula) presumably
this α5 gene variant, the most replicated genetic marker of smoking supporting exogenous attentional orientation (Corbetta and
(Bierut et al., 2008), now identified a dorsal ACC-ventral striatum/ Shulman, 2002). A second ensemble, termed the “task-negative” (or
extended amygdala circuit, such that the risk allele was associated “default-mode”) network consists of regions routinely showing activ-
with decreased rsFC between these structures. This circuit, represent- ity increases during passive states and reciprocal activity decreases
ing a “trait-like” biomarker, was impaired in smokers, not altered by during task performance (e.g., dorsomedial prefrontal cortex
acute nicotine administration, and was anatomically consistent with [dmPFC], vmPFC, posterior cingulate cortex [PCC] and parahippocam-
(although not identical to) that previously shown to predict addiction pal regions) presumably subserving task-independent endogenous
severity using the phenotypic Fagerström index (Hong et al., 2009). information processing (Buckner et al., 2008; Gusnard and Raichle,
Another independent smoking-related variant in the same gene clus- 2001; Raichle et al., 2001). Most efforts to elucidate the psychological
ter (α3) (Bierut et al., 2008) was associated with a separate circuit functions supported by the DMN have converged on the view that it is
between dorsal ACC and anterior thalamus that was related to recen- associated with internally directed cognitive operations 1 (e.g., self-
cy of smoking but not addiction severity, resembling a “state-like” reflection on past and future events, autobiographical, social or emo-
marker for smoking, perhaps related to craving or withdrawal in tional functions; Amodio and Frith, 2006; Buckner et al., 2008;
these mildly deprived smokers. The results of these initial studies Gusnard et al., 2001; Schacter et al., 2007). In the absence of explicit
suggest the intriguing possibility that alterations in specific neural task demands (i.e., “at rest”), intrinsic activity in the TPN and DMN
circuits may provide systems-level biomarkers of addiction severity fluctuate in a temporally anti-correlated fashion (Fox et al., 2005),
that could be leveraged to track cessation treatment trajectories. such that decreased activity in one coincides with increased activity
In sum, rsFC studies are beginning to shed light on circuit-level al- in the other, 2 suggesting they subserve opposing cognitive processes
terations associated with drug addiction. While initial findings are competing for limited processing resources (Fransson, 2006).
limited and have not been totally consistent, potentially due to Potentially one of the more heuristically useful perspectives to
between-study methodological and participant-characteristic issues, emerge from the nascent functional connectivity literature relates
supplementing traditional task-based neuroimaging data with rsFC this antagonistic TPN and DMN dynamic to consequences during
analyses may provide a deeper level of understanding regarding psy- goal-directed behavior (e.g., Figs. 3A,B). Specifically, intermittent fail-
chological deficits associated with reward, emotional and cognitive ures to adequately suppress DMN activity during goal-directed be-
processing often related to an extended drug use history. By relating havior have been identified as one source of interference limiting
extant addiction-related rsFC findings with a larger body of literature optimal performance (Sonuga-Barke and Castellanos, 2007). Task-
related to emotional regulation and cognitive control, we have induced DMN suppression is parametrically altered as a function of
attempted to provide a heuristic framework allowing for a transition cognitive load (Fransson, 2006; McKiernan et al., 2003), suggesting
from these early exploratory studies to more model-driven hypothe- reallocation of processing resources as dictated by task demands
sis testing. The section below extends this theme and provides in along a continuum rather than an “all-or-none” phenomenon. Fluctu-
greater detail an exemplar model-based framework derived from ations along this continuum manifest during monotonous task per-
existing connectivity and task-based activation studies, relating formance, where decreases in DMN suppression and concomitant
system-level neural circuit interactions with the cognitive deficits reductions in regional TPN activity increase the probability of error
often observed during smoking abstinence and the performance- commission (Fig. 3C; Eichele et al., 2008) and protracted response
enhancing effects of nicotine administration. times (Weissman et al., 2006). Maladaptive interactions between
DMN and TPN regions partly underlie suboptimal performance
Nicotine and large-scale networks (Prado and Weissman, 2011), such that decreased negative coupling
between these networks predicts increased variability in trial-to-
Nicotine's performance-enhancing properties manifest in multiple trial response times across individuals (Kelly et al., 2008). As such, al-
cognitive domains, particularly when considering abstinent smokers tered network dynamics and/or a compromised ability to suppress
(Heishman et al., 1994, 2010; Newhouse et al., 2004). Previous neuro- DMN activity have been proposed as a neurobiological explanation
imaging studies exploring such nicotinic or cholinergic effects have for attentional-control maladjustments in conditions such as ADHD
often done so in the context of cognitive task paradigms, providing (Fassbender et al., 2009; Sonuga-Barke and Castellanos, 2007), autism
information regarding pharmacological actions generally within spe- spectrum disorders (Broyd et al., 2009; Uddin and Menon, 2009),
cific brain regions (for review, see Bentley et al., 2011). rsFC studies
may provide additional, complementary insight by considering circuit 1
In fact, during tasks designed to interrogate such introspective functions, regions of
interactions between regions (Bressler and Menon, 2010), thus the DMN show increased activity (e.g. Sestieri et al., 2011). Additionally, while in the
allowing for a systems-level mechanistic account of nicotine's current review we generally refer to the DMN as if it were a single unitary entity, it
is important to note that this canonical network appears to be comprised of multiple
performance-enhancing properties, which has remained elusive. dissociable components subserving specific aspects of internally oriented cognitive
Below, we synthesize a heuristic framework that may serve to guide processes (e.g., Andrews-Hanna et al., 2010; Uddin et al., 2008).
future research by integrating recent findings from intrinsic network 2
Although not the intent of the current review, a methodological issue requires
connectivity neuroimaging studies with those from investigations in- mention here. It has been argued that anti-correlations between TPN and DMN simply
reflect an artifact resulting from a commonly employed preprocessing step in rsFC an-
terrogating nicotine's neuropharmacological actions. Given that a
alyses involving the removal of non-neuronal, physiological noise (e.g., cardiac and
hallmark feature of the tobacco abstinence syndrome is difficulty con- respiration cycles) common across the entire brain (i.e., mean global signal regression)
centrating (Hughes, 2007; Parrott et al., 1996), a systems-level theo- (Anderson et al., 2011; Murphy et al., 2009). Arguing against an artifactual explanation,
retical account of nicotine's effects on cognition could inform the such anti-correlated networks have been observed in the absence of mean global signal
development of improved smoking cessation pharmacotherapies regression (e.g., Fox et al., 2009), appear modulated by pharmacological manipulations
(e.g., Cole et al., 2010), and correlate with aspects of behavioral performance (e.g., Kel-
(Lerman et al., 2007) and, additionally, may hold therapeutic utility ly et al., 2008; Prado and Weissman, 2011). Nonetheless, the extent to which anti-
for other disease states involving attentional dysfunction (Levin et correlations between large-scale brain networks reflect an intrinsic property of brain
al., 2006). organization or merely a signal processing artifact is an ongoing debate.
M.T. Sutherland et al. / NeuroImage 62 (2012) 2281–2295 2287

Fig. 3. Two components derived from a spatial Independent Components Analysis of BOLD activity during performance of a speeded flanker task. (A) A “task positive” component
that includes the dorsal ACC and supplementary motor area (SMA: pictured in red/yellow) as well as the anterior insula and dorsal premotor area (not pictured), showing a pattern
of activation consistent with engagement of these regions in performance monitoring. (B) A “task negative” component that includes the posterior cingulate cortex (PCC: pictured
in blue/green), precuneus and retrosplenial cortex (not pictured) showing task-related deactivation consistent with the identification of these regions as part of the default-mode
network. Graphs illustrate component time courses in response to error commission. (C) Activity in the “task negative” component shows a linear increase in activity preceding
response errors.
Reproduced from Eichele et al. (2008).

chronic pain (Baliki et al., 2008; Tagliazucchi et al., 2010), depression forward states (high acetylcholine levels) described at the cellular-
(Sheline et al., 2010), schizophrenia (Williamson, 2007), anxiety, and level (Bentley et al., 2011; Hasselmo and McGaughy, 2004).
dementia (Menon, 2011). Thus, the waning of concentration during During acute nicotine withdrawal, smokers often report subjec-
monotonous task performance may arise from persistent, re- tively experienced “difficulty concentrating” and display objectively
emergent, and/or spontaneously occurring DMN activity supporting assessed impairments in task-based performance (Heishman, 1998;
task-irrelevant, internally oriented information processing. Hendricks et al., 2006; Kozink et al., 2010). In keeping with the dy-
On the other hand, nicotine-induced performance enhancement is namic network view discussed above, increased DMN functioning,
consistently observed during monotonous tasks requiring sustained at- decreased TPN operations, and/or maladaptive interactions between
tention, vigilance, and visuospatial orientation (Hahn et al., 2007; components of these networks may account for abstinence-induced
Lawrence et al., 2002; Newhouse et al., 2004). Emerging evidence sug- cognitive impairments. Supporting such a proposal, self-reported im-
gests nicotine augments performance by suppressing DMN processes proved concentration following nicotine administration to abstinent
while also enhancing those associated with the TPN. For example, in smokers has been associated with increased negative coupling be-
minimally-abstinent smokers, nicotine enhances deactivations in re- tween DMN and TPN regions (Fig. 4D; Cole et al., 2010). Additionally,
gions overlapping those of the DMN during task cue presentation nicotine decreases intra-individual response time variability (Hahn
which is also associated with faster responding to subsequently pre- et al., 2007); variability thought to arise from maladaptive dynamic
sented targets (Figs. 4A,B; Hahn et al., 2007). Enhanced suppression interactions between DMN and TPN (Kelly et al., 2008). Relative to
of DMN regions may be a general mechanism by which nicotine ele- the drug-sated state, 24-h abstinence leads to reduced activation in
vates global task-based focus, as similar enhanced deactivations, occur- TPN regions (e.g. lateral PFC) during performance of a sustained at-
ring concurrently with augmented performance, have been observed tention task (Ettinger et al., 2009). Nicotinic stimulation with vareni-
when probing different cognitive constructs such as stimulus detec- cline, a modestly efficacious pharmacotherapy for smoking cessation,
tion, selective/divided attention (Hahn et al., 2009), sustained atten- increases activity in core TPN nodes (i.e., lateral and posterior-medial
tion (Beaver et al., 2011), and overt attentional shifting (Ettinger et PFC) during demanding working memory performance following
al., 2009). Nicotine administered to non-smokers decreases DMN activ- smoking abstinence (Loughead et al., 2010). Thus, we suggest acute
ity “at rest” (Fig. 4C; Tanabe et al., 2011), suggesting such effects are not nicotine withdrawal may be a particularly relevant endogenous stim-
constrained to task-specific contexts nor limited to the amelioration of ulus necessitating further processing resources in the service of
abstinence-induced effects in smokers. In contrast to nicotine-induced returning the individual to a euthymic, homeostatic set point, but at
decreases in DMN and consistent with enhancement of sensory-based the expense of reduced processing efficiency for exogenous stimuli.
information processing, nicotine potentiates rsFC in cingulate- In addition to negatively impacting attention to exogenous stimu-
neocortical circuits of minimally-deprived smokers (Fig. 2A; Hong et li, increased DMN activity may give rise to the percept of drug urges,
al., 2009) and in extrastriate visual circuits of non-smokers (Tanabe cravings, and/or ruminative thoughts about use. For example, in-
et al., 2011). Such nicotine-induced suppression of DMN and reciprocal creased cerebral blood flow to multiple regions, including some over-
enhancement of TPN activity at the systems-level, may parallel acetyl- lapping the DMN (e.g., vmPFC, hippocampus) predicts the severity of
choline's role in toggling circuit dynamics between cortico-cortical abstinence-induced smoking urges (Wang et al., 2007). Moreover, in-
feedback states (low acetylcholine levels) and thalamo-cortical feed- dependent of withdrawal, increased activity in DMN regions such as
2288 M.T. Sutherland et al. / NeuroImage 62 (2012) 2281–2295

Fig. 4. Nicotine's impact on default-mode functioning. (A) Nicotine enhanced deactivation in DMN regions (PCC, dmPFC) in minimally deprived (~3 h) smokers under nicotine (rel-
ative to placebo) administration during a spatial attention task. (B) Nicotine-induced deactivation in the PCC correlates with reduced reaction time (difference values reflect Nic-
otine–Placebo). (C) Nicotine (relative to pre-nicotine baseline) reduced activity in DMN regions (vmPFC, PCC, precuneus) of non-smokers. (D) Example time-courses of DMN (blue)
and ECN (red) activity during the resting state under nicotine and placebo conditions in two abstinent (~ 12 h) smokers. Top graphs illustrate enhanced negative coupling between
the DMN and ECN following nicotine in an individual reporting decreased withdrawal symptoms following nicotine replacement. Bottom graphs show little change in DMN-ECN
coupling following nicotine administration in an individual reporting no change in withdrawal symptoms.
Panels A and B are reproduced from Hahn et al. (2007). Panel C is reproduced from Tanabe et al. (2011). Panel D is reproduced from Cole et al. (2010).

vmPFC, PCC and (para)hippocampus is observed as a function of reac- clinically anxious and depressed populations characterized by a pro-
tivity to drug cues and/or use-urges (Fig. 5; Brenhouse et al., 2008; pensity to perseverate on negative self-reflections (Gentili et al.,
Franklin et al., 2011; Garavan et al., 2000; Goudriaan et al., 2010; 2009; Hamilton et al., 2011; Lanius et al., 2010; Sheline et al., 2010;
Langleben et al., 2008; Li and Sinha, 2008; Naqvi and Bechara, 2009; Zhou et al., 2010). Increased DMN activity and/or maladaptive inter-
Weinstein et al., 2010; Wilcox et al., 2011; Zhang et al., 2011b). Cog- actions with TPN regions may reflect an intermediate endophenotype
nitive down-regulation of cue-induced cravings is accompanied by associated with acute withdrawal, resulting in cognitive impairments
increased activity in TPN regions (e.g., lateral and posterior-medial as well as increased use-urges.
PFC) and concomitant decreases in reward-related and DMN regions Based on the above, we hypothesize that nicotine withdrawal can
(e.g., ventral striatum, ACC and vmPFC) (Kober et al., 2010). Finally, enhance and nicotine administration suppress, DMN functioning. Addi-
given the overlap between symptoms of major depressive disorder tionally, maladaptive interactions between DMN and TPN may provide
and acute nicotine withdrawal, for example, depressed mood, prob- a systems-level mechanistic account regarding deficits in sustained at-
lems concentrating, restlessness and sleeping difficulties (American tention, performance monitoring and inhibitory control following
Psychiatric Association, 1994), it is noteworthy that enhanced rsFC acute abstinence from addictive drugs (Garavan and Hester, 2007;
and reduced deactivation in DMN regions has been observed among Garavan and Stout, 2005; Heishman et al., 1994; Verdejo-García et al.,
M.T. Sutherland et al. / NeuroImage 62 (2012) 2281–2295 2289

Fig. 5. Increased activity in DMN regions in response to cues. (A) BOLD activation in DMN regions (PCC, precuneus) to heroin-related versus neutral cues in opioid-dependent in-
dividuals. (B,C). Heroin-related cues increase BOLD activation in DMN regions (vmPFC, hippocampus) and the insula immediately prior to and after a methadone dose in opioid-
dependent individuals.
Panel A reproduced from Wang et al. (2010). Panels B and C are reproduced from Langleben et al. (2008).

2008). Specifically, nicotine may enhance performance via a shift in ac- mediating such dynamic activity switching between large-scale brain
tivity from a network subserving internally oriented, to one or more networks, and is there a role for nicotine in such a process?
networks mediating externally oriented information processing. Such
enhancing effects likely are more evident in populations experiencing Insula, network switching and interoception
state- (e.g., abstinent drug users, sleep deprived or fatigued partici-
pants) or trait-related (e.g., ADHD, chronic pain) dysfunctions in exter- The brain is inundated with a continuous flow of information aris-
nally oriented information processing (Newhouse et al., 2004). ing from exogenous and endogenous sources, necessitating control
However, the question remains: what are the neural substrates mechanisms to identify, and in turn act upon, the currently most
2290 M.T. Sutherland et al. / NeuroImage 62 (2012) 2281–2295

salient stimuli. A distinct network, composed of insula and ACC nodes, these insular functional subdivisions using rsFC analyses has generally
has been suggested to play a critical and causal role in the initiation, bolstered such a conceptualization (Cauda et al., 2011; Deen et al.,
maintenance, and adjustment of attentional control (Bressler and 2011; Taylor et al., 2009). Accordingly, multiple perspectives have
Menon, 2010; Dosenbach et al., 2006, 2007, 2008; Menon and emerged relating insula with subjective drug urges (Naqvi and
Uddin, 2010). Such findings suggest the fractionation of the TPN dis- Bechara, 2009; Garavan, 2010), impaired behavioral monitoring
cussed above into at least two dissociable sub-networks. Indeed, in (Goldstein et al., 2009), and maladaptive decision-making (Paulus,
the absence of explicit task demands, two distinct networks appear 2007) in drug addicts. Naqvi and Bechara (2010) have elucidated a
to exist, each subserving different psychological processes: one an- rather thorough model of insula's interoceptive role at multiple stages
chored in anterior insula and ACC termed the “salience network” of the addiction cycle (e.g., drug-taking, -withdrawal, -urges). Of par-
(SN), and a second composed of lateral prefrontal and parietal regions ticular note, during acute abstinence, the insula is theorized to track
termed the “executive control network” (ECN; Seeley et al., 2007). homeostatically relevant withdrawal-induced bodily changes and in
The SN has been proposed to influence moment-to-moment infor- turn interact with other regions (e.g., vmPFC, amygdala and striatum),
mation processing by identifying the most subjectively relevant stim- thereby altering affective (e.g., anxiety, irritability) and motivational
uli (Seeley et al., 2007), whether arising from internal or external loci, (e.g., cue reactivity, smoking urges) states (Naqvi and Bechara, 2010).
and “toggling” activity between the DMN and ECN accordingly To this list we suggest adding attentional states, hypothesizing that in-
(Hamilton et al., 2011; Sridharan et al., 2008; Uddin et al., 2011). In creased withdrawal-related insula engagement during abstinence in-
cognitive task paradigms, insula and dorsal ACC activity routinely terferes with normal network switching processes and perhaps
co-occur (Lawrence et al., 2003; Medford and Critchley, 2010), likely underlies the breakdown in negative coupling between the DMN and
in the service of monitoring ongoing goal-directed behavior for salient ECN. That is to say, during periods of abstinence the most homeostati-
stimuli (e.g., errors, response conflict) which precipitates a cascade of cally relevant stimuli are often internal bodily sensations subjectively
neural events resulting in attentional and behavioral adjustments to experienced as withdrawal symptoms, which result in objectively
optimize behavior (Botvinick et al., 1999; Egner and Hirsch, 2005; manifest decreased behavioral performance due to a shift in activity to-
Kerns et al., 2004). By assessing the temporal dynamics and direction- wards DMN and away from ECN.
al interactions between specific nodes of the SN, DMN and ECN, Given the discussion above, it is not surprising that the insula ap-
Sridharan et al. (2008) identified the insula as a causal outflow hub pears to play a pivotal role in nicotine addiction. Critically, damage to
mediating dynamic switching between DMN and ECN activity as dic- the insula can result in a sudden and profound disruption of smoking
tated by task-events and also occurring intrinsically “at rest”. 3 They behavior (Naqvi et al., 2007). Further supporting insula's involve-
further postulated that such network switching is facilitated by a set ment, a recent study observed greater gray matter density in the an-
of specialized spindle neurons (Allman et al., 2011), found exclusively, terior insula of smokers in comparison to non-smokers (Fig. 6A;
and in high quantities, within the human anterior insula and dorsal Zhang et al., 2011a). Additionally, when deprived of nicotine, elevated
ACC underlying fast, adaptive switching of the prevailing dominant activity in the insula, along with other brain regions, covaries with in-
network state. More recently, the same research group has shown creased abstinence-induced smoking urges (Wang et al., 2007). Mul-
that insula's causal influence on DMN and ECN nodes matures over tiple studies of cigarette-cue reactivity have noted positive
the course of normal development, as does the underlying structural associations between insula responses and subjective use-urges
connectivity between such regions (Uddin et al., 2011). Further sug- (Naqvi and Bechara, 2009). Connecting insula activity with attention-
gesting insula's role in “toggling” between DMN and TPN, Hamilton al processes, Janes et al. (2010) reported that attentional bias for
et al. (2011) have reported insular engagement at points of transition smoking-related stimuli assessed in a Stroop task variant was posi-
between prevailing DMN or TPN activity in patients suffering from tively correlated with greater insula cue-reactivity (Fig. 6B). Con-
major depressive disorder and healthy controls. From a drug abuse versely, during performance of a sustained attention task not
perspective, the proposal that the SN orients attention to the most involving drug-related stimuli, nicotine administered to minimally-
homeostatically-relevant source of information arising from endoge- deprived smokers decreased insula activity while also improving per-
nous or exogenous stimuli (Seeley et al., 2007) may provide one par- formance (Fig. 6C; Lawrence et al., 2002). Thus, we propose that
simonious neurobiological account of some of the cognitive deficits insula hyperactivity following nicotine-abstinence or in response to
often noted, not only in abstinent-smokers, but also in other drug smoking-related cues may precipitate decreased cognitive perfor-
using cohorts (Garavan and Stout, 2005; Verdejo-García et al., 2008). mance. Such a view is consistent with previous proposals relating
Complementing a network-switching function, insula's role in insula dysfunction with other neuropsychiatric disorders such as
interoception (Craig, 2002; Craig, 2009), and, by extension, drug addic- anxiety (hyperactivity: Paulus and Stein, 2006) and autism spectrum
tion (Naqvi and Bechara, 2009, 2010), has been of substantial recent in- disorders (hypoactivity: Uddin and Menon, 2009).
terest. Interoception refers to the monitoring of internal bodily states to
maintain or procure homeostasis, possibly via rousing the organism A network model of nicotine addiction
through affective, motivational, and attentional alterations. The insula
appears to be parsed into multiple subregions along a posterior-to-an- Drawing upon the above literature, we propose a model that inte-
terior gradient (Craig, 2009), such that more posterior regions have grates contemporary understanding of intrinsic network dynamics
been related to primary interoceptive operations and more anterior re- with evidence of alterations within and between these networks as
gions associated with affective and cognitive processes. Exploration of a function of nicotine abstinence and administration, cue reactivity
and/or drug urges. We hypothesize that during acute abstinence,
the insula monitors salient interoceptive states, thus marking the
3
A second methodological point warrants discussion. In additional to assessing presence of endogenous, withdrawal-related somatic, affective, and/
functional connectivity between brain regions, the directional influence of one region
or motivational events. The insula in turn interacts with DMN regions
onto another is often of particular interest. One approach for inferring directionality,
termed Granger causality analysis has been increasingly applied in task-based and rsFC in the service of orienting attention to resolve this inner tumult and
studies (e.g., Sridharan et al., 2008; Uddin et al., 2011). It is important to note that such return the system to homeostasis, thereby shifting network dynamics
“lag-based” directionality/causality methods may not be appropriate for inferring cau- and biasing processing towards the DMN and away from the ECN.
sality within fMRI datasets given variability in the hemodynamic response function Such a shift may underlie the cognitive impairments observed across
across brain regions (e.g., Smith et al., 2011). Thus, additional studies using alterative
analysis strategies are needed to verify the veracity of seminal studies suggesting insu-
various task domains during abstinence. Additionally, preoccupation
la's association with dynamic switching between large-scale networks (e.g., Hamilton with endogenous, withdrawal-related stimuli may hinder the capaci-
et al., 2011). ty of the SN to engage in extrinsic performance monitoring, further
M.T. Sutherland et al. / NeuroImage 62 (2012) 2281–2295 2291

Fig. 6. Insula involvement in nicotine addiction and attentional processes. (A) Greater gray matter density in smokers (n = 48) relative to matched controls in the left insula.
(B) Increased insula activity to smoking-related versus neutral cues is positively correlated with attention to smoking-cues in an affective Stroop task. (C). Difference map and
bar graph illustrating enhanced BOLD deactivations in the insula under nicotine relative to placebo conditions during a sustained attention task (RVIP) but not a sensorimotor con-
trol task in minimally deprived (~ 3 h) smokers.
Panel A is reproduced from Zhang et al. (2011a). Panel B is reproduced from Janes et al. (2010). Panel C is reproduced from Lawrence et al. (2002).

contributing to cognitive deficits. Given the functional heterogeneity interference effects associated with this pattern of co-activation
within insula, it remains for future research to identify those subre- (Janes et al., 2010). We would argue that drug cues elicit a salient in-
gions most critically involved in such processes. teroceptive state increasing insula activity, and in turn the DMN,
This hypothesized shift in network dynamics during abstinence drawing resources away from extrinsic task-positive regions, produc-
(Fig. 7A) would result in one or more of the following observable ing slower response times and increased error rates.
changes in rsFC: 1) enhanced rsFC between insula and DMN, which While the above focus lies with nicotine abstinence, this
would correlate with the severity of self-reported withdrawal symp- network-based perspective could apply equally well to acute, and
toms and impaired task performance; 2) reduced rsFC between insula perhaps protracted, withdrawal from other drugs of abuse. Such a
and the ECN; 3) enhanced rsFC within the DMN; 4) reduced rsFC with- model would account, at least in part, for the coincidence of cogni-
in the ECN; (e.g., Cole et al., 2010; Kelly et al., 2011); and 5) a break- tive deficits observed across drug addicted populations, typically
down in negative coupling between the DMN and ECN (e.g., Cole et assessed during acute abstinent states (Forman et al., 2004;
al., 2010). Conversely, acute nicotine administration (Fig. 7B) may Goldstein et al., 2004; Hester and Garavan, 2004; Hester et al.,
bias processing away from the DMN, resulting in enhanced rsFC with- 2007, 2009; Sofuoglu, 2010). Reduced recruitment of dorsal ACC
in the ECN and between the ECN and insula. Such network dynamics and ECN regions accompanying these deficits (Forman et al., 2004;
may not only underlie the amelioration of withdrawal symptoms, Goldstein et al., 2004; Hester and Garavan, 2004; Hester et al.,
but may also reflect an inherent property of nicotine to suppress 2009) is also in accordance with such an abstinence model. Of
DMN activity and enhance processing of extrinsic task-based stimuli. course, any differences arising between abstinent drug-using popu-
This latter view derives from the cognitive enhancing effects observed lations and non-using controls may also reflect pre-existing vulner-
in nicotine-naïve populations (Heishman et al., 2010) as well as evi- abilities or drug-induced functional and/or structural changes
dence that nicotine suppresses the DMN activity in non-smokers independent of acute withdrawal processes (e.g. Zhang et al.,
(Tanabe et al., 2011). Accordingly, a shift away from an insula-DMN 2011b). That said, while there exist obvious limitations to studying
biased pattern of rsFC and towards an insula-ECN pattern may be ob- acute withdrawal processes in drug-addicted individuals, we sug-
served in both smokers and non-smokers, accompanied by enhanced gest that following such individuals across the course of treatment
attentional performance. may present a means of testing this heuristic framework within dif-
In addition to providing a network-based account of abstinence ferent drug addicted cohorts and in turn, screening for relapse risk.
induced cognitive impairment (and conversely, enhancement follow- Specifically, as acute and protracted withdrawal symptoms subside,
ing acute nicotine administration), this model may provide a frame- altered network dynamics may ‘normalize’. Individuals showing
work within which to interpret the co-activation of insula and DMN the least change in rsFC dynamics are hypothesized to present the
regions often observed during reactivity to drug cues and/or drug greatest risk for recidivism to drug use. Finally, while we have em-
urges (Franklin et al., 2011; Goudriaan et al., 2010; Janes et al., phasized a role for the insula in mediating some of the psychological
2010; Wang et al., 2007), and potentially also drug Stroop deficits associated with drug use, given the multifaceted nature of
2292 M.T. Sutherland et al. / NeuroImage 62 (2012) 2281–2295

Fig. 7. A proposed model of activity within and between the default mode network (DMN), executive control network (ECN) and salience network (SN) under nicotine absti-
nence (A) and following acute nicotine administration (B). Arrow thickness between and within networks reflects the hypothesized strength of interactions between networks.
The thick arrow between the insula and endogenously relevant interoceptive events in (A) reflects an influx of such events during nicotine abstinence. Similarly, thick arrows
between the dACC and ECN and their conceptual outputs in (B) reflects an enhanced capacity to engage in task execution and performance monitoring following nicotine
administration.

this neuropsychiatric disease, alternative circuit disruptions (such as dynamic activity between two large-scale brain networks, the default-
those described earlier) also represent potential targets for future mode network (DMN) and the executive control network (ECN).
treatment development. These networks associated with endogenously oriented processes and
In sum, rsFC provides a useful tool for studying multifaceted neuro- exogenously oriented attention, respectively, competitively interact
psychiatric diseases like addiction at a systems-level of assessment. To during task performance, with suppression of DMN activity often asso-
efficiently leverage the capabilities of this tool, stronger, model-driven ciated with optimal behavioral performance. By modulating dynamic
approaches need to be utilized. To this end, we attempt to formulate a network activity, the anterior insula is hypothesized to expedite proces-
framework of dynamic large-scale network interactions derived from sing of the most homeostatically relevant stimuli arising from either in-
recent advances in intrinsic functional connectivity to explain the con- ternal or external events. During nicotine abstinence, the insula may
sequences of acute nicotine abstinence and attention enhancing effects track withdrawal-induced bodily sensations and in turn direct attention
of nicotine administration. Complementing an interoceptive monitor- towards this homeostatically salient internal state via increased interac-
ing role, emerging evidence implicates insula involvement in directing tions with the DMN at the expense of decreased exogenously directed
attention towards either internal or external stimuli by mediating attention mediated by ECN.
M.T. Sutherland et al. / NeuroImage 62 (2012) 2281–2295 2293

Acknowledgments Craig, A.D., 2002. How do you feel? Interoception: the sense of the physiological condi-
tion of the body. Nat. Rev. Neurosci. 10 (1), 59–70.
Craig, A.D., 2009. How do you feel—now? The anterior insula and human awareness.
This work was supported by the National Institute on Drug Abuse, Nat. Rev. Neurosci. 10, 59–70.
Intramural Research Program, NIH/DHHS. Daglish, M.R.C., Weinstein, A., Malizia, A.L., Wilson, S., Melichar, J.K., Lingford-Hughes,
A., Myles, J.S., Grasby, P., Nutt, D.J., 2003. Functional connectivity analysis of the
neural circuits of opiate craving: “more” rather than “different”? Neuroimage 20
References (4), 1964–1970.
Damoiseaux, J.S., Greicius, M.D., 2009. Greater than the sum of its parts: a review of
Allman, J.M., Tetreault, N.A., Hakeem, A.Y., Park, S., 2011. The von economo neurons in studies combining structural connectivity and resting-state functional connectivi-
apes and humans. Am. J. Hum. Biol. 23, 5–21. ty. Brain Struct. Funct. 213, 525–533.
American Psychiatric Association, 1994. Diagnostic and Statistical Manual of Mental Deen, B., Pitskel, N.B., Pelphrey, K.A., 2011. Three systems of insular functional connec-
Disorders 4th edn. American Psychiatric Publishing, Washington, DC. tivity identified with cluster analysis. Cereb. Cortex 21 (7), 1498–1506.
Amodio, D.M., Frith, C.D., 2006. Meeting of minds: the medial frontal cortex and social Di Chiara, G., Bassareo, V., Fenu, S., De Luca, M.A., Spina, L., Cadoni, C., Acquas, E.,
cognition. Nat. Rev. Neurosci. 7, 268–277. Carboni, E., Valentini, V., Lecca, D., 2004. Dopamine and drug addiction: the nucleus
Anand, A., Li, Y., Wang, Y., Wu, J., Gao, S., Bukhari, L., Mathews, V.P., Kalnin, A., Lowe, accumbens shell connection. Neuropharmacology 47 (Suppl. 1), 227–241.
M.J., 2005. Activity and connectivity of brain mood regulating circuit in depression: Diekhof, E.K., Falkai, P., Gruber, O., 2008. Functional neuroimaging of reward processing
a functional magnetic resonance study. Biol. Psychiatry 57, 1079–1088. and decision-making: a review of aberrant motivational and affective processing in
Anderson, J.S., Druzgal, T.J., Lopez-Larson, M., Jeong, E.-K., Desai, K., Yurgelun-Todd, D., addiction and mood disorders. Brain Res. Rev. 59, 164–184.
2011. Network anticorrelations, global regression, and phase-shifted soft tissue Dosenbach, N.U.F., Visscher, K.M., Palmer, E.D., Miezin, F.M., Wenger, K.K., Kang, H.C.,
correction. Hum. Brain Mapp. 32, 919–934. Burgund, E.D., Grimes, A.L., Schlaggar, B.L., Petersen, S.E., 2006. A core system for
Andrews-Hanna, J.R., Reidler, J.S., Sepulcre, J., Poulin, R., Buckner, R.L., 2010. Functional- the implementation of task sets. Neuron 50, 799–812.
anatomic fractionation of the brain's default network. Neuron 65, 550–562. Dosenbach, N.U.F., Fair, D.A., Miezin, F.M., Cohen, A.L., Wenger, K.K., Dosenbach, R.A.T.,
Baliki, M.N., Geha, P.Y., Apkarian, A.V., Chialvo, D.R., 2008. Beyond feeling: chronic pain Fox, M.D., Snyder, A.Z., Vincent, J.L., Raichle, M.E., Schlaggar, B.L., Petersen, S.E.,
hurts the brain, disrupting the default-mode network dynamics. J. Neurosci. 28, 2007. Distinct brain networks for adaptive and stable task control in humans.
1398–1403. Proc. Natl. Acad. Sci. U. S. A. 104, 11073–11078.
Barrós-Loscertales, A., Garavan, H., Bustamante, J.C., Ventura-Campos, N., Llopis, J.J., Dosenbach, N.U.F., Fair, D.A., Cohen, A.L., Schlaggar, B.L., Petersen, S.E., 2008. A dual-
Belloch, V., Parcet, M.A., et al., 2011. Reduced striatal volume in cocaine- networks architecture of top-down control. Trends Cogn. Sci. 12, 99–105.
dependent patients. Neuroimage 56 (3), 1021–1026. Egner, T., Hirsch, J., 2005. Cognitive control mechanisms resolve conflict through corti-
Beaver, J.D., Long, C.J., Cole, D.M., Durcan, M.J., Bannon, L.C., Mishra, R.G., Matthews, cal amplification of task-relevant information. Nat. Neurosci. 8, 1784–1790.
P.M., 2011. The effects of nicotine replacement on cognitive brain activity during Eichele, T., Debener, S., Calhoun, V.D., Specht, K., Engel, A.K., Hugdahl, K., von Cramon,
smoking withdrawal studied with simultaneous fMRI/EEG. Neuropsychopharma- D.Y., Ullsperger, M., 2008. Prediction of human errors by maladaptive changes in
cology 36 (9), 1792–1800. event-related brain networks. Proc. Natl. Acad. Sci. U. S. A. 105, 6173–6178.
Bentley, P., Driver, J., Dolan, R.J., 2011. Cholinergic modulation of cognition: insights Ersche, K.D., Clark, L., London, M., Robbins, T.W., Sahakian, B.J., 2006. Profile of execu-
from human pharmacological functional neuroimaging. Prog. Neurobiol. 94, tive and memory function associated with amphetamine and opiate dependence.
360–388. Neuropsychopharmacology 31 (5), 1036–1047.
Berkman, E.T., Lieberman, M.D., 2009. Using neuroscience to broaden emotion regula- Ettinger, U., Williams, S.C.R., Patel, D., Michel, T.M., Nwaigwe, A., Caceres, A., Mehta,
tion: theoretical and methodological considerations. Soc. Pers. Psychol. Compass 3, M.A., Anilkumar, A.P., Kumari, V., 2009. Effects of acute nicotine on brain function
1–19. in healthy smokers and non-smokers: estimation of inter-individual response het-
Berridge, K.C., Robinson, T.E., 1998. What is the role of dopamine in reward: hedonic erogeneity. Neuroimage 45, 549–561.
impact, reward learning, or incentive salience? Brain Res. Rev. 28, 309–369. Everitt, B.J., Robbins, T.W., 2005. Neural systems of reinforcement for drug addiction:
Bickel, W.K., Miller, M.L., Yi, R., Kowal, B.P., Lindquist, D.M., Pitcock, J.A., 2007. Behavior- from actions to habits to compulsion. Nat. Neurosci. 8, 1481–1489.
al and neuroeconomics of drug addiction: competing neural systems and temporal Fassbender, C., Zhang, H., Buzy, W.M., Cortes, C.R., Mizuiri, D., Beckett, L., Schweitzer,
discounting processes. Drug Alcohol Depend. 90 (Suppl. 1), S85–S91. J.B., 2009. A lack of default network suppression is linked to increased distractibil-
Bierut, L.J., Stitzel, J.A., Wang, J.C., Hinrichs, A.L., Grucza, R.A., Xuei, X., Saccone, N.L., ity in ADHD. Brain Res. 1273, 114–128.
Saccone, S.F., Bertelsen, S., Fox, L., Horton, W.J., Breslau, N., Budde, J., Cloninger, Foland-Ross, L.C., Altshuler, L.L., Bookheimer, S.Y., Lieberman, M.D., Townsend, J.,
C.R., Dick, D.M., Foroud, T., Hatsukami, D., Hesselbrock, V., Johnson, E.O., Kramer, Penfold, C., Moody, T., Ahlf, K., Shen, J.K., Madsen, S.K., Rasser, P.E., Toga, A.W.,
J., Kuperman, S., Madden, P.A., Mayo, K., Nurnberger Jr., J., Pomerleau, O., Porjesz, Thompson, P.M., 2010. Amygdala reactivity in healthy adults is correlated with
B., Reyes, O., Schuckit, M., Swan, G., Tischfield, J.A., Edenberg, H.J., Rice, J.P., Goate, prefrontal cortical thickness. J. Neurosci. 30, 16673–16678.
A.M., 2008. Variants in nicotinic receptors and risk for nicotine dependence. Am. Forman, S.D., Dougherty, G.G., Casey, B.J., Siegle, G.J., Braver, T.S., Barch, D.M., Stenger, V.A.,
J. Psychiatry 165, 1163–1171. Wick-Hull, C., Pisarov, L.A., Lorensen, E., 2004. Opiate addicts lack error-dependent ac-
Blair, R.J., 2008. The amygdala and ventromedial prefrontal cortex: functional contribu- tivation of rostral anterior cingulate. Biological Psychiatry 55 (5), 531–537.
tions and dysfunction in psychopathy. Philos. Trans. R. Soc. Lond. B Biol. Sci. 363, Fox, M.D., Greicius, M., 2010. Clinical applications of resting state functional connectiv-
2557–2565. ity. Frontiers in systems neuroscience 4, 19. doi:10.3389/fnsys.2010.00019.
Botvinick, M., Nystrom, L.E., Fissell, K., Carter, C.S., Cohen, J.D., 1999. Conflict monitor- Fox, M.D., Snyder, A.Z., Vincent, J.L., Corbetta, M., Van Essen, D.C., Raichle, M.E., 2005.
ing versus selection-for-action in anterior cingulate cortex. Nature 402, 179–181. The human brain is intrinsically organized into dynamic, anticorrelated functional
Breiter, H.C., Gollub, R.L., Weisskoff, R.M., Kennedy, D.N., Makris, N., Berke, J.D., networks. Proc. Natl. Acad. Sci. U. S. A. 102, 9673–9678.
Goodman, J.M., Kantor, H.L., Gastfriend, D.R., Riorden, J.P., Mathew, R.T., Rosen, Fox, M.D., Zhang, D.Y., Snyder, A.Z., Raichle, M.E., 2009. The global signal and ob-
B.R., Hyman, S.E., 1997. Acute effects of cocaine on human brain activity and emo- served anticorrelated resting state brain networks. J. Neurophysiol. 101,
tion. Neuron 19, 591–611. 3270–3283.
Brenhouse, H.C., Sonntag, K.C., Andersen, 2008. Transient D1 dopamine receptor ex- Frank, M.J., Woroch, B.S., Curran, T., 2005. Error-related negativity predicts reinforce-
pression on prefrontal cortex projection neurons: relationship to enhanced moti- ment learning and conflict biases. Neuron 47 (4), 495–501.
vational salience of drug cues in adolescence. J. Neurosci. 28, 2375–2382. Franken, I.H., van Strien, J.W., Kuijpers, I., 2010. Evidence for a deficit in the salience at-
Bressler, S.L., Menon, V., 2010. Large-scale brain networks in cognition: emerging tribution to errors in smokers. Drug and Alcohol Dependence 106 (2–3), 181–185.
methods and principles. Trends Cogn. Sci. 14, 277–290. Franklin, T., Wang, Z., Suh, J.J., Hazan, R., Cruz, J., Li, Y., Goldman, M., Detre, J.A., O'Brien,
Broyd, S.J., Demanuele, C., Debener, S., Helps, S.K., James, C.J., Sonuga-Barke, E.J.S., 2009. C.P., Childress, A.-R., 2011. Effects of varenicline on smoking cue-triggered neural
Default-mode brain dysfunction in mental disorders: a systematic review. Neu- and craving responses. Arch. Gen. Psychiatry 68, 516–526.
rosci. Biobehav. Rev. 33, 279–296. Fransson, P., 2006. How default is the default mode of brain function? Further evidence
Buckner, R.L., Andrews-Hanna, J.R., Schacter, D.L., 2008. The brain's default network: from intrinsic BOLD signal fluctuations. Neuropsychologia 44, 2836–2845.
anatomy, function, and relevance to disease. Ann. N. Y. Acad. Sci. 1124, 1–38. Garavan, H., 2010. Insula and drug cravings. Brain Struct. Funct. 214, 593–601.
Camchong, J., MacDonald, A.W., Nelson, B., Bell, C., Mueller, B.A., Specker, S., Lim, K.O., Garavan, H., Hester, R., 2007. The role of cognitive control in cocaine dependence. Neu-
2011. Frontal hyperconnectivity related to discounting and reversal learning in co- ropsychol. Rev. 17, 337–345.
caine subjects. Biol. Psychiatry 69, 1117–1123. Garavan, H., Stout, J.C., 2005. Neurocognitive insights into substance abuse. Trends
Carter, C.S., van Veen, V., 2007. Anterior cingulate cortex and conflict detection: an up- Cogn. Sci. 9, 195–201.
date of theory and data. Cognitive, affective & behavioral neuroscience 7 (4), Garavan, H., Pankiewicz, J., Bloom, A., Cho, J.K., Sperry, L., Ross, T.J., Salmeron, B.J., Risinger,
367–379. R., Kelley, D., Stein, E.A., 2000. Cue-induced cocaine craving: neuroanatomical speci-
Cauda, F., D'Agata, F., Sacco, K., Duca, S., Geminiani, G., Vercelli, A., 2011. Functional ficity for drug users and drug stimuli. Am. J. Psychiatry 157, 1789–1798.
connectivity of the insula in the resting brain. Neuroimage 55, 8–23. Gentili, C., Ricciardi, E., Gobbini, M.I., Santarelli, M.F., Haxby, J.V., Pietrini, P., Guazzelli,
Chen, S., Ross, T.J., Zhan, W., Myers, C.S., Chuang, K.-S., Heishman, S.J., Stein, E.A., Yang, M., 2009. Beyond amygdala: default mode network activity differs between pa-
Y., 2008. Group independent component analysis reveals consistent resting-state tients with social phobia and healthy controls. Brain Res. Bull. 79, 409–413.
networks across multiple sessions. Brain Res. 1239, 141–151. Glahn, D.C., Winkler, A.M., Kochunov, P., Almasy, L., Duggirala, R., Carless, M.A., Curran,
Cole, D.M., Beckmann, C.F., Long, C.J., Matthews, P.M., Durcan, M.J., Beaver, J.D., 2010. Nic- J.C., Olvera, R.L., Laird, A.R., Smith, S.M., Beckmann, C.F., Fox, P.T., Blangero, J., 2010.
otine replacement in abstinent smokers improves cognitive withdrawal symptoms Genetic control over the resting brain. Proc. Natl. Acad. Sci. U. S. A. 107, 1223–1228.
with modulation of resting brain network dynamics. Neuroimage 52, 590–599. Goldstein, R.Z., Volkow, N.D., 2002. Drug addiction and its underlying neurobiological
Corbetta, M., Shulman, G.L., 2002. Control of goal-directed and stimulus-driven atten- basis: neuroimaging evidence for the involvement of the frontal cortex. Am. J. Psy-
tion in the brain. Nat. Rev. Neurosci. 3, 201–215. chiatry 159, 1642–1652.
2294 M.T. Sutherland et al. / NeuroImage 62 (2012) 2281–2295

Goldstein, R.Z., Leskovjan, A.C., Hoff, A.L., Hitzemann, R., Bashan, F., Khalsa, S.S., Wang, Kelly, C., Zuo, X.-N., Gotimer, K., Cox, C.L., Lynch, L., Brock, D., Imperati, D., Garavan, H.,
G.-J., et al., 2004. Severity of neuropsychological impairment in cocaine and alcohol Rotrosen, J., Castellanos, F.X., Milham, M.P., 2011. Reduced interhemispheric rest-
addiction: association with metabolism in the prefrontal cortex. Neuropsychologia ing state functional connectivity in cocaine addiction. Biol. Psychiatry 69, 684–692.
42 (11), 1447–1458. Kennedy, D.P., Redcay, E., Courchesne, E., 2006. Failing to deactivate: resting functional
Goldstein, R.Z., Alia-Klein, N., Tomasi, D., Carrillo, J.H., Maloney, T., Woicik, P.A., Wang, R., abnormalities in autism. Proc. Natl. Acad. Sci. U. S. A. 103, 8275–8280.
Telang, F., Volkow, N.D., 2009. Anterior cingulate cortex hypoactivations to an emo- Kerns, J.G., Cohen, J.D., MacDonald, A.W., Cho, R.Y., Stenger, V.A., Carter, C.S., 2004. An-
tionally salient task in cocaine addiction. Proc. Natl. Acad. Sci. U. S. A. 106, 9453–9458. terior cingulate conflict monitoring and adjustments in control. Science 303,
Goudriaan, A.E., de Ruiter, M.B., van den Brink, W., Oosterlaan, J., Veltman, D.J., 2010. 1023–1026.
Brain activation patterns associated with cue reactivity and craving in abstinent Kim, E., Ku, J., Namkoong, K., Lee, W., Lee, K.S., Park, J.-Y., Lee, S.Y., Kim, J.-J., Kim, S.I.,
problem gamblers, heavy smokers and healthy controls: an fMRI study. Addict. Jung, Y.-C., 2009. Mammillothalamic functional connectivity and memory function
Biol. 15, 491–503. in Wernicke's encephalopathy. Brain 132, 369–376.
Greicius, M.D., Srivastava, G., Reiss, A.L., Menon, V., 2004. Default-mode network activ- Kim, M.J., Gee, D.G., Loucks, R.A., Davis, F.C., Whalen, P.J., 2011a. Anxiety dissociates
ity distinguishes Alzheimer's disease from healthy aging: evidence from functional dorsal and ventral medial prefrontal cortex functional connectivity with the amyg-
MRI. Proc. Natl. Acad. Sci. U. S. A. 101, 4637–4642. dala at rest. Cereb. Cortex 21, 1667–1673.
Greicius, M.D., Supekar, K., Menon, V., Dougherty, R.F., 2009. Resting-state functional Kim, M.J., Loucks, R.A., Palmer, A.L., Brown, A.C., Solomon, K.M., Marchante, A.N.,
connectivity reflects structural connectivity in the default mode network. Cereb. Whalen, P.J., 2011b. The structural and functional connectivity of the amygdala:
Cortex 19, 72–78. from normal emotion to pathological anxiety. Behav. Brain Res. 223, 403–410.
Gruber, S.A., Silveri, M.M., Yurgelun-Todd, D.A., 2007. Neuropsychological conse- King, J.A., Korb, F.M., von Cramon, D.Y., Ullsperger, M., 2010. Post-error behavioral ad-
quences of opiate use. Neuropsychol. Rev. 17 (3), 299–315. justments are facilitated by activation and suppression of task-relevant and task-
Gu, H., Salmeron, B.J., Ross, T.J., Geng, X., Zhan, W., Stein, E.A., Yang, Y., 2010. Mesocor- irrelevant information processing. The Journal of neuroscience: the official journal
ticolimbic circuits are impaired in chronic cocaine users as demonstrated by of the Society for Neuroscience 30 (38), 12759–12769.
resting-state functional connectivity. Neuroimage 53, 593–601. Kober, H., Mende-Siedlecki, P., Kross, E.F., Weber, J., Mischel, W., Hart, C.L., Ochsner,
Gusnard, D.A., Raichle, M.E., 2001. Searching for a baseline: functional imaging and the K.N., 2010. Prefrontal–striatal pathway underlies cognitive regulation of craving.
resting human brain. Nat. Rev. Neurosci. 2, 685–694. Proc. Natl. Acad. Sci. U. S. A. 107, 14811–14816.
Gusnard, D.A., Akbudak, E., Shulman, G.L., Raichle, M.E., 2001. Medial prefrontal cortex Koob, G.F., Le Moal, M., 1997. Drug abuse: hedonic homeostatic dysregulation. Science
and self-referential mental activity: relation to a default mode of brain function. 278, 52–58.
Proc. Natl. Acad. Sci. U. S. A. 98, 4259–4264. Koob, G.F., Le Moal, M., 2005. Plasticity of reward neurocircuitry and the “dark side” of
Hahn, B., Ross, T.J., Yang, Y., Kim, I., Huestis, M.A., Stein, E.A., 2007. Nicotine enhances drug addiction. Nat. Neurosci. 8, 1442–1444.
visuospatial attention by deactivating areas of the resting brain default network. Koob, G.F., Le Moal, M., 2008. Review. Neurobiological mechanisms for opponent moti-
J. Neurosci. 27, 3477–3489. vational processes in addiction. Philos. Trans. R. Soc. Lond. B Biol. Sci. 363,
Hahn, B., Ross, T.J., Wolkenberg, F.A., Shakleya, D.M., Huestis, M.A., Stein, E.A., 2009. 3113–3123.
Performance effects of nicotine during selective attention, divided attention, and Koob, G.F., Volkow, N.D., 2010. Neurocircuitry of addiction. Neuropsychopharmacology
simple stimulus detection: an fMRI study. Cereb. Cortex 19, 1990–2000. 35, 217–238.
Hahn, A., Stein, P., Windischberger, C., Weissenbacher, A., Spindeleggera, C., Moser, E., Kozink, R.V., Lutz, A.M., Rose, J.E., Froeliger, B., McClernon, F.J., 2010. Smoking with-
Kaspera, S., Lanzenberger, R., 2011. Reduced resting-state functional connectivity drawal shifts the spatiotemporal dynamics of neurocognition. Addict. Biol. 15,
between amygdala and orbitofrontal cortex in social anxiety disorder. Neuroimage 480–490.
56 (3), 881–889. Kumari, V., Gray, J.A., Ffytche, D.H., Mitterschiffthaler, M.T., Das, M., Zachariah, E.,
Hamilton, J.P., Furman, D.J., Chang, C., Thomason, M.E., Dennis, E., Gotlib, I.H., 2011. De- Vythelingum, G.N., Williams, S.C.R., Simmons, A., Sharma, T., 2003. Cognitive ef-
fault-mode and task-positive network activity in major depressive disorder: impli- fects of nicotine in humans: an fMRI study. Neuroimage 19, 1002–1013.
cations for adaptive and maladaptive rumination. Biol. Psychiatry 70 (4), 327–333. Langleben, D.D., Ruparel, K., Elman, I., Busch-Winokur, S., Pratiwadi, R., Loughead, J.,
Hampson, M., Driesen, N.R., Skudlarski, P., Gore, J.C., Constable, R.T., 2006. Brain con- O'Brien, C.P., Childress, A.R., 2008. Acute effect of methadone maintenance dose
nectivity related to working memory performance. J. Neurosci. 26, 13338–13343. on brain FMRI response to heroin-related cues. Am. J. Psychiatry 165 (3), 390–394.
Hariri, A.R., Mattay, V.S., Tessitore, A., Fera, F., Weinberger, D.R., 2003. Neocortical mod- Lanius, R.A., Bluhm, R.I., Coupland, N.J., Hegadoren, K.M., Rowe, B., 2010. Default mode
ulation of the amygdala response to fearful stimuli. Biol. Psychiatry 53, 494–501. network connectivity as a predictor of post-traumatic stress disorder symptom se-
Hasselmo, M.E., McGaughy, J., 2004. High acetylcholine levels set circuit dynamics for verity in acutely traumatized subjects. J. Psychiatry Neurosci. 33–40.
attention and encoding and low acetylcholine levels set dynamics for consolida- Lawrence, N.S., Ross, T.J., Stein, E.A., 2002. Cognitive mechanisms of nicotine on visual
tion. Prog. Brain Res. 145, 207–231. attention. Neuron 36, 539–548.
Heishman, S.J., 1998. What aspects of human performance are truly enhanced by nico- Lawrence, N.S., Ross, T.J., Hoffmann, R., Garavan, H., Stein, E.A., 2003. Multiple neuronal
tine? Addiction 93, 317–320. networks mediate sustained attention. J. Cogn. Neurosci. 15, 1028–1038.
Heishman, S.J., Taylor, R.C., Henningfield, J.E., 1994. Nicotine and smoking: a review of Lerman, C., LeSage, M.G., Perkins, K.A., O'Malley, S.S., Siegel, S.J., Benowitz, N.L.,
effects on human performance. Exp. Clin. Psychopharmacol. 2 (4), 345–395. Corrigall, W.A., 2007. Translational research in medication development for nico-
Heishman, S.J., Kleykamp, B.A., Singleton, E.G., 2010. Meta-analysis of the acute effects tine dependence. Nat. Rev. Drug Discov. 6, 746–762.
of nicotine and smoking on human performance. Psychopharmacology 210, Levin, E.D., McClernon, F.J., Rezvani, A.H., 2006. Nicotinic effects on cognitive function:
453–469. behavioral characterization, pharmacological specification, and anatomic localiza-
Hendricks, P.S., Ditre, J.W., Drobes, D.J., Brandon, T.H., 2006. The early time course of tion. Psychopharmacology 184, 523–539.
smoking withdrawal effects. Psychopharmacology 187, 385–396. Li, C-shan R., Sinha, R., 2008. Inhibitory control and emotional stress regulation: neuro-
Hester, R., Garavan, H., 2004. Executive dysfunction in cocaine addiction: evidence for imaging evidence for frontal-limbic dysfunction in psycho-stimulant addiction.
discordant frontal, cingulate, and cerebellar activity. J. Neurosci. 24 (49), Neurosci. Biobehav. Rev. 32, 581–597.
11017–11022. Li, S.J., Biswal, B., Li, Z., Risinger, R., Rainey, C., Cho, J.K., Salmeron, B.J., Stein, E.A., 2000.
Hester, R., Simões-Franklin, C., Garavan, H., 2007. Post-error behavior in active cocaine Cocaine administration decreases functional connectivity in human primary visual
users: poor awareness of errors in the presence of intact performance adjustments. and motor cortex as detected by functional MRI. Magn. Reson. Med. 43, 45–51.
Neuropsychopharmacology 32 (9), 1974–1984. Liu, J., Liang, J., Qin, W., Tian, J., Yuan, K., Bai, L., Zhang, Y., Wang, W., Wang, Y., Li, Q.,
Hester, R., Barre, N., Murphy, K., Silk, T.J., Mattingley, J.B., 2008. Human medial frontal Zhao, L., Lu, L., von Deneen, K.M., Liu, Y., Gold, M.S., 2009. Dysfunctional connectiv-
cortex activity predicts learning from errors. Cerebral cortex (New York, N.Y.: ity patterns in chronic heroin users: an fMRI study. Neurosci. Lett. 460, 72–77.
1991) 18 (8), 1933–1940. Loughead, J., Ray, R., Wileyto, E.P., Ruparel, K., Sanborn, P., Siegel, S., Gur, R.C., Lerman,
Hester, R., Nestor, L., Garavan, H., 2009. Impaired error awareness and anterior cingu- C., 2010. Effects of the alpha4beta2 partial agonist varenicline on brain activity and
late cortex hypoactivity in chronic cannabis users. Neuropsychopharmacology 34 working memory in abstinent smokers. Biological Psychiatry 67 (8), 715–721.
(11), 2450–2458. Lowe, M.J., Phillips, M.D., Lurito, J.T., Mattson, D., Dzemidzic, M., Mathews, V.P., 2002.
Honey, C.J., Sporns, O., Cammoun, L., Gigandet, X., Thiran, J.P., Meuli, R., Hagmann, P., Multiple sclerosis: low-frequency temporal blood oxygen level-dependent fluctu-
2009. Predicting human resting-state functional connectivity from structural con- ations indicate reduced functional connectivity initial results. Radiology 224,
nectivity. Proc. Natl. Acad. Sci. U. S. A. 106, 2035–2040. 184–192.
Hong, L.E., Gu, H., Yang, Y., Ross, T.J., Salmeron, B.J., Buchholz, B., Thaker, G.K., Stein, E.A., Lustig, C., Snyder, A.Z., Bhakta, M., O'Brien, K.C., McAvoy, M., Raichle, M.E., Morris, J.C.,
2009. Association of nicotine addiction and nicotine's actions with separate cingu- Buckner, R.L., 2003. Functional deactivations: change with age and dementia of
late cortex functional circuits. Arch. Gen. Psychiatry 66, 431–441. the Alzheimer type. Proc. Natl. Acad. Sci. U. S. A. 100, 14504–14509.
Hong, L.E., Hodgkinson, C.A., Yang, Y., Sampath, H., Ross, T.J., Buchholz, B., Salmeron, Ma, N., Liu, Y., Li, N., Wang, C.-X., Zhang, H., Jiang, X.-F., Xu, H.-S., Fu, X.-M., Hu, X.,
B.J., Srivastava, V., Thaker, G.K., Goldman, D., Stein, E.A., 2010. A genetically modu- Zhang, D.-R., 2010. Addiction related alteration in resting-state brain connectivity.
lated, intrinsic cingulate circuit supports human nicotine addiction. Proc. Natl. Neuroimage 49, 738–744.
Acad. Sci. U. S. A. 107, 13509–13514. Magno, E., Foxe, J.J., Molholm, S., Robertson, I.H., Garavan, H., 2006. The anterior cingu-
Hughes, J.R., 2007. Effects of abstinence from tobacco: valid symptoms and time course. late and error avoidance. J. Neurosci 26, 4769–4773.
Nicotine Tob. Res. 9, 315–327. McKiernan, K.A., Kaufman, J.N., Kucera-Thompson, J., Binder, J.R., 2003. A parametric
Janes, A.C., Pizzagalli, D.A., Richardt, S., Frederick Bde, B., Holmes, A.J., Sousa, J., Fava, M., manipulation of factors affecting task-induced deactivation in functional neuroim-
Evins, A.E., Kaufman, M.J., 2010. Neural substrates of attentional bias for smoking- aging. J. Cogn. Neurosci. 15, 394–408.
related cues: an FMRI study. Neuropsychopharmacology 35 (12), 2339–2345. Medford, N., Critchley, H.D., 2010. Conjoint activity of anterior insular and anterior cin-
Kelly, A.M.C., Uddin, L.Q., Biswal, B.B., Castellanos, F.X., Milham, M.P., 2008. Competi- gulate cortex: awareness and response. Brain Struct. Funct. 214, 535–549.
tion between functional brain networks mediates behavioral variability. Neuro- Menon, V., 2011. Large-scale brain networks and psychopathology: a unifying triple
image 39, 527–537. network model. Trends Cogn. Sci. 15 (10), 483–506.
M.T. Sutherland et al. / NeuroImage 62 (2012) 2281–2295 2295

Menon, V., Uddin, L.Q., 2010. Saliency, switching, attention and control: a network Stein, E.A., Pankiewicz, J., Harsch, H.H., Cho, J.K., Fuller, S.A., Hoffmann, R.G., Hawkins,
model of insula function. Brain Struct. Funct. 214, 655–667. M., Rao, S.M., Bandettini, P.A., Bloom, A.S., 1998. Nicotine-induced limbic cortical
Meyer-Lindenberg, A., 2009. Neural connectivity as an intermediate phenotype: brain activation in the human brain: a functional MRI study. Am. J. Psychiatry 155,
networks under genetic control. Hum. Brain Mapp. 30, 1938–1946. 1009–1015.
Miller, E.K., Cohen, J.D., 2001. An integrative theory of prefrontal cortex function. Annu- Stein, J.L., Wiedholz, L.M., Bassett, D.S., Weinberger, D.R., Zink, C.F., Mattay, V.S.,
al Review of Neuroscience (1), 167. Meyer-Lindenberg, A., 2007. A validated network of effective amygdala connec-
Morón, J.A., Green, T.A., 2010. Exploring the molecular basis of addiction: drug-induced tivity. Neuroimage 36, 736–745.
neuroadaptations. Neuropsychopharmacology 35, 337–338. Tagliazucchi, E., Balenzuela, P., Fraiman, D., Chialvo, D.R., 2010. Brain resting state is
Motzkin, J.C., Newman, J.P., Kiehl, K.A., Koenigs, M., 2011. Reduced prefrontal connec- disrupted in chronic back pain patients. Neurosci. Lett. 485, 26–31.
tivity in psychopathy. J. Neurosci. 31 (48), 17348–17357. Tambini, A., Ketz, N., Davachi, L., 2010. Enhanced brain correlations during rest are re-
Murphy, K., Birn, R.M., Handwerker, D.A., Jones, T.B., Bandettini, P.A., 2009. The impact lated to memory for recent experiences. Neuron 65, 280–290.
of global signal regression on resting state correlations: are anti-correlated net- Tanabe, J., Nyberg, E., Martin, L.F., Martin, J., Cordes, D., Kronberg, E., Tregellas, J.R.,
works introduced? Neuroimage 44 (3), 893–905. 2011. Nicotine effects on default mode network during resting state. Psychophar-
Nakama, H., Chang, L., Fein, G., Shimotsu, R., Jiang, C.S., Ernst, T., 2011. Methamphet- macology 216, 287–295.
amine users show greater than normal age-related cortical gray matter loss. Addic- Taylor, K.S., Seminowicz, D.A., Davis, K.D., 2009. Two systems of resting state con-
tion 106 (8), 1474–1483. nectivity between the insula and cingulate cortex. Hum. Brain Mapp. 30,
Naqvi, N.H., Bechara, A., 2009. The hidden island of addiction: the insula. Trends Neu- 2731–2745.
rosci. 32, 56–67. Tian, L., Jiang, T., Wang, Y., Zang, Y., He, Y., Liang, M., Sui, M., Cao, Q., Hu, S., Peng, M.,
Naqvi, N.H., Bechara, A., 2010. The insula and drug addiction: an interoceptive view of Zhuo, Y., 2006. Altered resting-state functional connectivity patterns of anterior
pleasure, urges, and decision-making. Brain Struct. Funct. 214, 435–450. cingulate cortex in adolescents with attention deficit hyperactivity disorder. Neu-
Naqvi, N.H., Rudrauf, D., Damasio, H., Bechara, A., 2007. Damage to the insula disrupts rosci. Lett. 400, 39–43.
addiction to cigarette smoking. Science 315, 531–534. Tomasi, D., Volkow, N.D., Wang, R., Carrillo, J.H., Maloney, T., Alia-Klein, N., Woicik, P.A.,
Nestler, E.J., 2005. Is there a common molecular pathway for addiction? Nat. Neurosci. Telang, F., Goldstein, R.Z., 2010. Disrupted functional connectivity with dopaminer-
8, 1445–1449. gic midbrain in cocaine abusers. PLoS One 5, e11509.
Newhouse, P.A., Potter, A., Singh, A., 2004. Effects of nicotinic stimulation on cognitive Uddin, L.Q., Menon, V., 2009. The anterior insula in autism: under-connected and
performance. Curr. Opin. Pharmacol. 4, 36–46. under-examined. Neurosci. Biobehav. Rev. 33, 1198–1203.
Ochsner, K.N., Ray, R.D., Cooper, J.C., Robertson, E.R., Chopra, S., Gabrieli, J.D.E., Gross, Uddin, L.Q., Kelly, A.M.C., Biswal, B.B., Castellanos, F.X., Milham, M.P., 2008. Functional
J.J., 2004. For better or for worse: neural systems supporting the cognitive down- connectivity of default mode network components: correlation, anticorrelation
and up-regulation of negative emotion. Neuroimage 23 (2), 483–499. and causality. Hum. Brain Mapp. 30, 625–637.
Parrott, A.C., Garnham, N.J., Wesnes, K., Pincock, C., 1996. Cigarette smoking and absti- Uddin, L.Q., Supekar, K.S., Ryali, S., Menon, V., 2011. Dynamic reconfiguration of struc-
nence: comparative effects upon cognitive task performance and mood state over tural and functional connectivity across core neurocognitive brain networks with
24 hours. Human Psychopharmacol. Clin. Exp. 11, 391–400. development. J. Neurosci. 31 (50), 18578–18589.
Paulus, M.P., 2007. Decision-making dysfunctions in psychiatry-altered homeostatic Upadhyay, J., Maleki, N., Potter, J., Elman, I., Rudrauf, D., Knudsen, J., Wallin, D., Pendse,
processing? Science 318, 602–606. G., McDonald, L., Griffin, M., Anderson, J., Nutile, L., Renshaw, P., Weiss, R., Becerra,
Paulus, M.P., Stein, M.B., 2006. An insular view of anxiety. Biol. Psychiatry 60, 383–387. L., Borsook, D., 2010. Alterations in brain structure and functional connectivity in
Pezawas, L., Meyer-Lindenberg, A., Drabant, E.M., Verchinski, B.A., Munoz, K.E., prescription opioid-dependent patients. Brain 133, 2098–2114.
Kolachana, B.S., Egan, M.F., Mattay, V.S., Hariri, A.R., Weinberger, D.R., 2005. 5- van den Heuvel, M.P., Hulshoff Pol, H.E., 2010. Exploring the brain network: a review
HTTLPR polymorphism impacts human cingulate-amygdala interactions: a genetic on resting-state fMRI functional connectivity. Eur. Neuropsychopharmacol. 20,
susceptibility mechanism for depression. Nat. Neurosci. 8, 828–834. 519–534.
Phan, K.L., Orlichenko, A., Boyd, E., Angstadta, M., Coccarod, E.F., Liberzona, I., Arfanakis, Verdejo-García, A., Lawrence, A.J., Clark, L., 2008. Impulsivity as a vulnerability mark-
K., 2009. Preliminary evidence of white matter abnormality in the uncinate fascic- er for substance-use disorders: review of findings from high-risk research, prob-
ulus in generalized social anxiety disorder. Biol. Psychiatry 66, 691–694. lem gamblers and genetic association studies. Neurosci. Biobehav. Rev. 32,
Phillips, M.L., Drevets, W.C., Rauch, S.L., Lane, R., 2003. Neurobiology of emotion per- 777–810.
ception I: the neural basis of normal emotion perception. Biol. Psychiatry 54, Wang, K., Jiang, T., Liang, M., Wang, L., Tian, L., Zhang, X., Li, K., Liu, Z., 2006. Discrimi-
504–514. native analysis of early Alzheimer's disease based on two intrinsically anti-
Prado, J., Weissman, D.H., 2011. Heightened interactions between a key default-mode correlated networks with resting-state fMRI. Med. Image Comput. Comput. Assist.
region and a key task-positive region are linked to suboptimal current perfor- Interv. 9, 340–347.
mance but to enhanced future performance. Neuroimage 56, 2276–2282. Wang, Z., Faith, M., Patterson, F., Tang, K., Kerrin, K., Wileyto, E.P., Detre, J.A., Lerman, C.,
Raichle, M.E., MacLeod, A.M., Snyder, A.Z., Powers, W.J., Gusnard, D.A., Shulman, G.L., 2007. Neural substrates of abstinence-induced cigarette cravings in chronic
2001. A default mode of brain function. Proc. Natl. Acad. Sci. U. S. A. 98, 676–682. smokers. J. Neurosci. 27, 14035–14040.
Redish, 2004. Addiction as a computational process gone awry. Science 306, Wang, W., Wang, Y.R., Qin, W., Yuan, K., Tian, J., Li, Q., Yang, L.Y., Lu, L., Guo, Y.M., 2010.
1944–1947. Changes in functional connectivity of ventral anterior cingulate cortex in heroin
Ridderinkhof, K.R., Ullsperger, M., Crone, E.A., Nieuwenhuis, S., 2004. The role of the abusers. Chin. Med. J. (Engl) 123 (12), 1582–1588.
medial frontal cortex in cognitive control. Science (New York, N.Y.) 306 (5695), Weinstein, A., Greif, J., Yemini, Z., Lerman, H., Weizman, A., Even-Sapir, E., 2010. Atten-
443–447. uation of cue-induced smoking urges and brain reward activity in smokers treated
Rombouts, S.A.R.B., Barkhof, F., Goekoop, R., Stam, C.J., Scheltens, P., 2005. Altered rest- successfully with bupropion. J. Psychopharmacol. 24, 829–838.
ing state networks in mild cognitive impairment and mild Alzheimer's disease: an Weissman, D.H., Roberts, K.C., Visscher, K.M., Woldorff, M.G., 2006. The neural bases of
fMRI study. Hum. Brain Mapp. 26, 231–239. momentary lapses in attention. Nat. Neurosci. 9, 971–978.
Schacter, D.L., Addis, D.R., Buckner, R.L., 2007. Remembering the past to imagine the fu- Wilcox, C.E., Teshiba, T.M., Merideth, F., Ling, J., Mayer, A.R., 2011. Enhanced cue reac-
ture: the prospective brain. Nat. Rev. Neurosci. 8, 657–661. tivity and fronto-striatal functional connectivity in cocaine use disorders. Drug and
Seeley, W.W., Menon, V., Schatzberg, A.F., Keller, J., Glover, G.H., Kenna, H., Reiss, A.L., alcohol dependence 115 (1-2), 137–144. doi:10.1016/j. drugalcdep. 2011.01.009.
Greicius, M.D., 2007. Dissociable intrinsic connectivity networks for salience pro- Williamson, P., 2007. Are anticorrelated networks in the brain relevant to schizophre-
cessing and executive control. J. Neurosci. 27, 2349–2356. nia? Schizophr. Bull. 33, 994–1003.
Sestieri, C., Corbetta, M., Romani, G.L., Shulman, G.L., 2011. Episodic memory retrieval, Wise, R.A., 2008. Dopamine and reward: the anhedonia hypothesis 30 years on. Neuro-
parietal cortex, and the default mode network: functional and topographic ana- tox. Res. 14, 169–183.
lyses. J. Neurosci. 31, 4407–4420. Xie, C., Li, S., Shao, Y., Fu, L., Goveas, J., Ye, E., Li, W., Cohen, A.D., Chen, G., Zhang, Z.,
Sheline, Y.I., Price, J.L., Yan, Z., Mintun, M.A., 2010. Resting-state functional MRI in de- Yang, Z., 2011. Identification of hyperactive intrinsic amygdala network connec-
pression unmasks increased connectivity between networks via the dorsal nexus. tivity associated with impulsivity in abstinent heroin addicts. Behav. Brain Res.
Proc. Natl. Acad. Sci. U. S. A. 107, 11020–11025. 216, 639–646.
Smith, S.M., Fox, P.T., Miller, K.L., Glahn, D.C., Fox, P.M., Mackay, C.E., Filippini, N., Yuan, Y., Zhu, Z., Shi, J., Zou, Z., Yuan, F., Liu, Y., Lee, T.M.C., et al., 2009. Gray matter den-
Watkins, K.E., Toro, R., Laird, A.R., Beckmann, C.F., 2009. Correspondence of the sity negatively correlates with duration of heroin use in young lifetime heroin-
brain's functional architecture during activation and rest. Proc. Natl. Acad. Sci. U. dependent individuals. Brain Cogn. 71 (3), 223–228.
S. A. 106, 13040–13045. Yuan, K., Qin, W., Dong, M., Liu, J., Sun, J., Liu, P., Zhang, Y., et al., 2010. Gray matter def-
Smith, S.M., Miller, K.L., Salimi-Khorshidi, G., Webster, M., Beckmann, C.F., Nichols, T.E., icits and resting-state abnormalities in abstinent heroin-dependent individuals.
Ramsey, J.D., Woolrich, M.W., 2011. Network modelling methods for FMRI. Neuro- Neurosci. Lett. 482 (2), 101–105.
image 54 (2), 875–891. Zhang, X., Salmeron, B.J., Ross, T.J., Geng, X., Yang, Y., Stein, E.A., 2011a. Factors under-
Sofuoglu, M., 2010. Cognitive enhancement as a pharmacotherapy target for stimulant lying prefrontal and insula structural alterations in smokers. Neuroimage 54 (1),
addiction. Addiction 105 (1), 38–48. 42–48.
Sonuga-Barke, E.J.S., Castellanos, F.X., 2007. Spontaneous attentional fluctuations in im- Zhang, X., Salmeron, B.J., Ross, T.J., Gu, H., Geng, X., Yang, Y., Stein, E.A., 2011b. Anatom-
paired states and pathological conditions: a neurobiological hypothesis. Neurosci. ical differences and network characteristics underlying smoking cue reactivity.
Biobehav. Rev. 31, 977–986. Neuroimage 54 (1), 131–141.
Sridharan, D., Levitin, D.J., Menon, V., 2008. A critical role for the right fronto-insular Zhou, Y., Yu, C., Zheng, H., Liu, Y., Song, M., Qin, W., Li, K., Jiang, T., 2010. Increased neu-
cortex in switching between central-executive and default-mode networks. Proc. ral resources recruitment in the intrinsic organization in major depression. J. Af-
Natl. Acad. Sci. U. S. A. 105, 12569–12574. fect. Disord. 121, 220–230.

You might also like