Frontocoritical Circuits

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 29

Section I Structural and Functional Neuroanatomy

Chapter
Frontal-subcortical circuits

5 David G. Lichter

A series of parallel segregated frontal-subcortical cir- the dorsal caudate nucleus. In contrast, the ventral
cuits (FSCs) connect speciic regions of the frontal cor- architectonic trend, which originates in the ventral
tex (including paralimbic frontal areas) to the stria- orbital region and extends to the ventral portion of
tum, the globus pallidus (GP) and substantia nigra the frontal eye ield, maps onto the ventro-medial
(SN), and the thalamus [1–3]. Together, these con- portion of the caudate and adjacent portion of nucleus
stitute an important efector mechanism that allows accumbens (NAc). Closely connected cortical areas
the individual to interact adaptively with the envi- send converging projections into the striatum [8, 9].
ronment. his chapter reviews the neuroanatomy and Early experimental observations supported a
neurochemistry of FSCs, and outlines the signature role for discrete dorsal and ventral frontostriatal
syndromes of FSC circuit dysfunction. he similar- systems in cognition and behavior. hus, lesions or
ity of cognitive and behavioral changes that accom- electrical stimulation of the dorsolateral prefrontal
pany cortical and subcortical lesions is discussed (DLPF) cortex or of the anterodorsal head of the
within this framework. Select neuropsychiatric disor- caudate nucleus, to which this region projects,
ders that appear linked to FSC dysfunction are then produce deicits in delayed response and delayed
presented. Finally, we explore the manner in which alternation tasks [10, 11]. In contrast, lesions or
these insights may better inform clinical management electrical stimulation of either the orbitofrontal (OF)
of patients with relevant neuropsychological, behav- cortex or of the ventrolateral head of the caudate
ioral, and neuropsychiatric disorders. result in deicits in object alternation or response
inhibition paradigms [12]. Disruption to discrete
cognitive processes following striatal injury can be
Frontostriatal systems interpreted as the “downstream” interruption of
he frontal lobe may be viewed as comprising two anatomically congruent outlow from the frontal
distinct anatomical/functional systems, which relect cortex [12–14].
its dual developmental origin [4]. he sequential
processing of sensory, spatially related, and motiva-
tional information is mediated by a dorsal system, Frontal-subcortical circuits:
which comprises dorsolateral and medial portions of
the frontal lobes, interconnected with the posterior shared anatomy
parietal lobe and cingulate gyrus. Emotional tone is
mediated by a second, ventral system, which involves Basic circuit structure
the orbital surface of the frontal lobes. he architec- he following principal circuits may now be recog-
tonic organization of the prefrontal cortex [5, 6] is nized: a motor circuit that originates in the supple-
relected in the pattern of prefrontostriatal projections mentary motor area (SMA), an oculomotor circuit
[7]. hus, the dorsal architectonic trend, which orig- originating in the frontal eye ields, and three primary
inates in the rostral cingulate gyrus and culminates in circuits/circuit networks mediating cognitive, behav-
the dorsal portion of the frontal eye ield, maps onto ioral, and afective functions [1–3]. Of these, the DLPF

Behavioral Neurology & Neuropsychiatry, eds. David B. Arciniegas, C. Alan Anderson, and Christopher M. Filley.
Published by Cambridge University Press. C Cambridge University Press 2013.

59
Downloaded from https://www.cambridge.org/core. Stockholm University Library, on 25 Nov 2018 at 16:04:24, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781139016919.007
Section I: Structural and Functional Neuroanatomy

Figure 5.1. The general structure shared by


Frontal cortex all frontal-subcortical circuits (direct
connections).

Striatum

Globus pallidus/
substantia nigra

Thalamus

Figure 5.2. The general anatomy of the direct and indirect


frontal-subcortical pathways. 1 – Excitatory cortico-striatal fibers. 2 –
circuit mediates “executive functions,” i.e., the organi- Direct loop’s inhibitory striato-pallidal (GPi) fibers. 3 – Indirect loop’s
zation of information to facilitate a response. he lat- inhibitory striato-pallidal (GPe) fibers. 4 – Indirect loop’s inhibitory
eral OF (LOF) circuit is involved with integration of GPe-subthalamic nucleus fibers. 5 – Indirect loop’s excitatory
subthalamic-GPi/SNr fibers. 6 – Inhibitory outflow from GPi/SNr to
limbic and emotional information into goal-directed specific thalamic regions. 7 – Excitatory fibers returning from
and contextually appropriate behavioral responses. thalamus to cortex (shown for convenience in contralateral
Finally, the rostromedial limbic network consists of the hemisphere). Excitatory fibers are glutaminergic, inhibitory fibers
mediated via GABA. Note: these circuits are unilateral: each set of
medial OF (MOF) circuit, which facilitates integration projections from cortex to subcortical structures to cortex remains
of information pertaining to emotions, and the linked within a single hemisphere. In this figure, projection from cortex to
anterior cingulate (AC) circuitry. he AC network is subcortical structures as well as projections between subcortical
structures are illustrated on the left half of the figure whereas
involved primarily in motivational mechanisms but projections from thalamus to cortex are illustrated on the right side
also, through the activity of its subgenual portion, in of the figure. This does not suggest that these circuits cross from
mood regulation. one hemisphere to the other; instead, they are diagrammed in this
manner here in order to preserve readability of the figure.
All circuits share a basic anatomy, with an ori-
gin in the frontal lobes and sequential projections
to the striatum (caudate, putamen, or ventral stria- caudate area, and the MOF and AC circuitry connects
tum), GP/SN, and then to speciic thalamic nuclei, to the medial striatal/NAc region. Similar anatomic
with a inal link back to the frontal lobe (Figure 5.1). arrangements are maintained in the GP and thalamus
he circuits have two pathways: (1) a direct pathway, (Figure 5.3).
featuring a monosynaptic link between the striatum
and GP interna (GPi)/SN pars reticulata (SNr) com-
plex, and (2) an indirect pathway that projects from Open-loop elements
striatum to GP externa (GPe), linking to GPi/SNr While each circuit comprises a closed loop of anatom-
via the subthalamic nucleus [3] (Figure 5.2). Both ically segregated dedicated neurons, open-loop ele-
direct and indirect circuits project to the thalamus; ments support the functional connectivity of FSCs.
the direct pathway disinhibits the thalamus, whereas Circuit structures receive projections from non-circuit
the indirect pathway inhibits it. he relative inlu- cortical areas, thalamic and amygdaloid nuclei, and
ence of the two pathways therefore determines the also project to regions outside the circuits. Brain
inal output of the circuit. All circuits thus share com- regions linked by these aferent or eferent projec-
mon structures and are parallel and contiguous, but tions are functionally related [15, 16]. Circuits medi-
remain largely segregated anatomically, despite the ating limbic functions, for example, have connections
progressive focusing of succeeding projections onto to other limbic areas whereas those involved with
smaller numbers of neurons. hus, the DLPF cor- executive functions (EFs) interact with brain struc-
tex projects to the dorsolateral region of the cau- tures involved with cognition. In this manner, circuits
date nucleus, the LOF cortex projects to the ventral integrate information from anatomically disparate but

60
Downloaded from https://www.cambridge.org/core. Stockholm University Library, on 25 Nov 2018 at 16:04:24, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781139016919.007
Chapter 5: Frontal-subcortical circuits

cortex [1, 3]. hese areas project principally to the


putamen and thence to ventrolateral GPi, GPe, and
caudolateral SNr. he GP connects to ventral lateral,
ventral anterior, and centromedian nuclei of the tha-
lamus whose major eferents are to the SMA, premo-
tor cortex, and motor cortex, completing the circuit.
hroughout the circuit, the discrete somatotopic orga-
nization of movement-related neurons is maintained,
although information processing in the circuits is not
strictly sequential [3].

The oculomotor circuit


he oculomotor circuit originates in the frontal eye
ield, Brodmann’s area (BA) 8, as well as prefrontal and
posterior parietal cortex and connects sequentially to
the central body of the caudate nucleus, dorsomedial
GPi and ventrolateral SNr, ventral anterior and medial
dorsal thalamic nuclei, and back to the frontal eye ield
[1–3].

The dorsolateral prefrontal circuit


Figure 5.4 illustrates the anatomy of the direct path-
ways of two of the behaviorally relevant FSCs. he
DLPF circuit originates in BA 9 and 10 (areas recip-
rocally connected with adjacent BA 46) on the lat-
eral surface of the anterior frontal lobe. Neurons in
these regions project to the dorsolateral head of the
caudate nucleus [18], thence to the lateral aspect of
the mediodorsal GPi and rostrolateral SNr via the
direct pathway [19]. he indirect pathway projects
sequentially to the dorsal GPe, lateral subthalamic
nucleus [20] and GPi/SNr. Output from the basal gan-
Figure 5.3. The segregated anatomy of the frontal-subcortical glia projects to the ventral anterior and mediodorsal
circuits: dorsolateral prefrontal (horizontal stripes), lateral (parvocellular portion) thalamic nuclei [21–23]. he
orbitofrontal (stippling), and anterior cingulate/medial orbitofrontal
(cross-hatched) circuits in the striatum (top), pallidum (center), and circuit is closed by projections from these thalamic
mediodorsal thalamus (bottom). regions back to the dorsolateral frontal lobe [24, 25].

functionally related brain regions, the cascade of the The lateral orbitofrontal circuit
closed circuit constituting the eventual efector mech- he LOF circuit originates in the lateral orbital gyrus
anism [17]. of BA 11 and the medial inferior frontal gyrus of
areas 10 and 47 [26]. Multiple sensory inputs, includ-
Organization of individual ing olfactory, gustatory, visceral, somatic, and visual
aferents converge on this region. Projections are to
frontal-subcortical circuits the ventrolateral caudate and dorsal edge of the NAc
[27] with subsequent links to the most medial portion
The motor circuit of the mediodorsal GPi and to the dorsomedial SNr
he motor circuit originates from neurons in the SMA, [28]. he ventrolateral caudate also sends an indirect
premotor cortex, motor cortex, and somatosensory loop through the dorsal GPe to the lateral subthalamic

61
Downloaded from https://www.cambridge.org/core. Stockholm University Library, on 25 Nov 2018 at 16:04:24, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781139016919.007
Section I: Structural and Functional Neuroanatomy

The anterior cingulate and subgenual


cingulate circuits
here are two major subdivisions of the AC cortex,
which subserve distinct functions (Figure 5.6). he
dorsal cognitive division (BA 24, 24a’–b’ and 32’),
also referred to as the cognitive efector region [26],
is more developed in its cytoarchitecture than the
rostral-ventral division, and is part of a distributed
attentional network which has reciprocal interconnec-
tions with DLPF cortex (BA 46/9), parietal cortex (BA
7), and premotor and supplementary motor areas [30–
32]. his region is connected via the Muratof bundle
to dorsal and medial portions of the head and body of
the caudate nucleus [7, 18, 33], with succeeding pro-
jections to the GPi/SNr and thence to the parvocellu-
lar mediodorsal thalamus as well as the ventral ante-
rior and midline/intralaminar thalamic nuclei [25, 34].
hese thalamic regions then reconnect with the cogni-
tive division of AC via the inferior thalamic peduncle
and anterior internal capsule (Figure 5.5). Functions
of the cognitive efector region include modulation of
attention or executive functions by inluencing sensory
or response selection; monitoring competition, com-
Figure 5.4. The anatomy of the direct pathways of the plex motor control, motivation, novelty, error detec-
dorsolateral prefrontal and lateral orbitofrontal circuits. NAc, nucleus
accumbens; GPi, globus pallidus, internal segment; SNr, substantia
tion and working memory; and anticipation of cogni-
nigra, pars reticulata; VAmc, ventral anterior nucleus, magnocellular tively demanding tasks [35].
portion; VApc, ventral anterior nucleus, parvocellular portion; MDpc, he rostral-ventral afective division of the AC
mediodorsal nucleus, parvocellular portion; MDmc, mediodorsal
nucleus, magnocellular portion; MDpl, mediodorsal nucleus,
cortex comprises the subgenual cingulate (BA 25),
paralaminar portion. adjacent (caudal) portions of BA 32 (pregenual cingu-
late), area 33, and the rostral portions of AC cortex,
subcallosal areas 24a and 24b (Figure 5.6). hese
nucleus, thence to GPi and SNr [20]. Neurons are sent regions project to the medial part of the ventral stria-
from the GPi and SNr to the medial section of the mag- tum [18, 36], which includes the ventromedial caudate,
nocellular division of the ventral anterior thalamus as ventral putamen [37], and core and shell of the NAc
well as the paralaminar portion and inferomedial sec- (the shell region receiving projections especially from
tor of the magnocellular division of the mediodorsal the subgenual cingulate [38]). Projections from the
thalamus [18, 21, 29]. he circuit then closes with pro- ventral striatum and NAc core innervate the rostrome-
jections from these thalamic regions to the LOF cortex dial GPi and ventral pallidum (VP, the portion of the
(Figure 5.4). globus pallidus inferior to the anterior commissure)
as well as the rostromedial SNr and ventral tegmental
area (VTA) [39–41]. he NAc shell projects to the
The rostromedial limbic circuitry VP, VTA and SNpc, allowing this region to inluence
Emotional, motivational, and afective information dopaminergic inputs to other parts of the striatum
processed by the basal ganglia is represented in the ros- [41]. he VP and GPi then project to the dorsal portion
tromedial limbic circuitry arising from the orbital and of the magnocellular mediodorsal thalamus, midline
medial prefrontal cortex. Closely integrated anatom- and intralaminar (parafascicular) thalamic nuclei
ically and functionally, this comprises AC circuitry [23, 40, 42–44], with closed-loop projections back
(including that related to the subgenual cingulate) and to the anterior and subgenual cingulate (Figure 5.5)
the MOF circuit (Figure 5.5). [25, 34].

62
Downloaded from https://www.cambridge.org/core. Stockholm University Library, on 25 Nov 2018 at 16:04:24, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781139016919.007
Chapter 5: Frontal-subcortical circuits

Figure 5.5. The anatomy of the direct


pathways of the rostromedial limbic
circuits. VP, ventral pallidum; Pf,
parafascicular nucleus. Other
abbreviations follow Figure 5.4. The pale
arrows indicate open-loop connections
between engaged thalamic nuclei and the
lateral orbitofrontal and dorsolateral
prefrontal cortices.

While the OF cortex mediates information con- periaqueductal gray and dorsal brainstem [47]. he
cerning the internal environment, the AC circuitry strong projections to the dorsal raphe suggest a role
facilitates the intentional selection of environmental in regulating the overall function of the serotonergic
stimuli based on their internal relevance [45], thereby system [44]. With its major outlow to autonomic,
mediating motivated behavior. hrough both its dor- visceromotor, and endocrine systems [30, 35, 48], this
sal and rostral-ventral divisions and their connec- circuit has been implicated in salience monitoring of
tions with motor pathways, lateral PFC, and aferents emotional and motivational information, mediation
from the midline thalamus and brainstem (includ- of emotional and autonomic response to socially
ing arousal systems), the AC cortex is ideally posi- signiicant or provocative stimuli, afective processing,
tioned to participate in the willed control of behavior and inhibitory control [35, 49, 50].
[46].
he subgenual cingulate component of AC
circuitry (Figures 5.5 and 5.6) includes substan- The medial orbitofrontal circuit
tial predominantly ipsilateral connections with Arising from the gyrus rectus (areas 14r, 14c) and
the amygdala [44], MOF cortex (see below), other related areas on the medial orbital surface, includ-
portions of both anterior and posterior cingulate, ing BA 11m, 13a and 13b, the MOF cortex is
anterior insula, medial temporal lobe, hypothalamus, integrated anatomically and functionally with the

63
Downloaded from https://www.cambridge.org/core. Stockholm University Library, on 25 Nov 2018 at 16:04:24, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781139016919.007
Section I: Structural and Functional Neuroanatomy

Figure 5.7. The direct and indirect frontal-subcortical pathways.


Filled arrows represent inhibitory (GABAergic) pathways; open
arrows, excitatory (glutamatergic) pathways. Note that
dopaminergic input from the substantia nigra pars compacta and
ventral tegmental area is inhibitory, via D2 receptors, to
GABA/enkephalin neurons projecting to the indirect pathway, and
excitatory, via D1 receptors, to GABA/substance P neurons
projecting to the direct pathway.

specialized connections with the amygdala, the MOF


circuit may serve both as an integrator of visceral
Figure 5.6. The medial surface of the right hemisphere (anterior drives as well as a sensor of information pertaining to
towards the left), showing a schematic representation of
cytoarchitectural areas of the anterior cingulate cortex. The areas emotions [51, 58].
forming the dorsal cognitive division and the skeletomotor effector
region (striped) are outlined by solid lines, the rostro-ventral
affective division areas being outlined by dotted lines. Neurochemical organization
Shared neurochemical organization
infracallosal AC and subgenual cingulate (“visceral Common to all circuits is an origin in the frontal lobes
efector region”) [27, 51], with which it shares sub- with excitatory glutamatergic ibers that terminate in
cortical projections (Figure 5.5). Sequentially, these the striatum (caudate, putamen, and ventral striatum).
links are to the ventromedial caudate and NAc (core); hese striatal cells then project inhibitory gamma-
ventromedial pallidum and dorsomedial SNr; magno- aminobutyric acid (GABA) ibers both to neurons in
cellular portions of the ventral anterior and medial the GPi/SNr (direct loop connection) and to the GPe
mediodorsal thalamic nuclei, with a closed-loop pro- (indirect loop connection). Via the indirect loop, the
jection back to the MOF cortex [18, 23, 38–40, 52, GPe projects inhibitory GABA ibers to the subthala-
53]. In addition to its connections with the infra- mic nucleus which then connects with the GPi/SNr
callosal cingulate, the MOF cortex has strong recip- through excitatory glutamatergic ibers [2, 59]. he
rocal connections with the medial portion of the direct pathway expresses dopamine (DA) D1 recep-
basal and magnocellular division of the accessory tors and utilizes substance P with its GABA projec-
basal amygdala, as well as with the ventromedial tem- tion to the pallidum while the indirect loop receives
poral pole and rostral (agranular) insula [51, 54– its dopaminergic inluence via D2 receptors and com-
57]. he subgenual cingulate provides motivational bines GABA with enkephalin [60]. he GPi/SNr then
input to gustatory, olfactory and alimentary infor- project inhibitory GABA ibers to speciic thalamic
mation from anterior insular processing converging targets, which complete the circuit by sending a inal
on the MOF cortex. Considering its robust connec- excitatory connection to the cortical site of the circuit’s
tions with high-order sensory association cortices and origin in the frontal lobe (Figure 5.7).

64
Downloaded from https://www.cambridge.org/core. Stockholm University Library, on 25 Nov 2018 at 16:04:24, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781139016919.007
Chapter 5: Frontal-subcortical circuits

Circuit-discrete neurochemical organization retrorubral region (cell group A8) and the VTA (A10
cell group) innervate the entire striatum, thereby inlu-
he striatum is organized as two separate systems, the
encing each of the FSCs. his provides an anatomic
striosomes and the matrix. hese elements are dif-
basis for the multifaceted efects of dopaminergic
ferentiated by their distinct ontological, connectional,
agents on motor activity, motivation, thought, and
and chemical characteristics [61]. Relative to matrix
behavior. Relecting DA’s modulatory function, DA-
neurons, striosomal cells mature earlier, have lower
containing axon terminals synapse directly on stri-
concentrations of DA, serotonin and acetylcholine,
atal output neurons, many on the necks of dendritic
and have high concentrations of limbic-associated
spines. he VTA is the primary source of DA for the
membrane protein [62, 63]. Striosomes receive dense
ventral striatum, prefrontal cortex, and limbic targets.
orbitofrontal and insular input and have high levels
he nigra has inhibitory connections, via D1 receptors,
of D1 receptors, with dopaminergic projections from
with the indirect portions of the FSCs and excitatory
the ventral tier of the substantia nigra pars compacta
connections, via D2 receptors, with the direct circuits
(SNpc). In contradistinction, matrix cells receive afer-
(see Figure 5.2). Representing an important conver-
ents predominantly from the sensorimotor cortex and
gence within the otherwise segregated FSCs, the SNpc
express primarily D2 receptors, with dopaminergic
receives difuse input from the limbic circuits, provid-
input from the dorsal tier of the SNpc [64]. he matrix
ing a means for limbic emotional input to inluence
stains selectively for adenylate cyclase whereas the
both motor activity and cognition.
phosphoinositide system is selectively concentrated
in the striosomes of the medial and ventral striatum
[65]. GABAergic output from the striosomes is to the The cholinergic system
medial portion of the SNpc, dedicated to the OF cir-
Cholinergic input to the basal ganglia and most of
cuit, whereas the GABAergic output of the matrix is to
the thalamus is derived from the pedunculopontine
the GPe, GPi, and SNr.
nucleus and laterodorsal tegmentum of the brainstem.
Mirroring the circuit-discrete neurochemical orga-
Second, portions of the mediodorsal, ventroanterior,
nization of the striatum, the NAc region of the ventro-
and reticular nuclei of the thalamus, as well as lim-
medial striatum is also divided into two functionally
bic structures and neocortex, receive cholinergic input
distinct components with discrete histochemistries.
from the basal forebrain, which includes the septum,
hus, the core or dorsolateral portion of the NAc,
diagonal band of Broca, and the nucleus basalis of
which receives projections from the MOF and AC
Meynert [66]. Acetylcholine facilitates thalamic acti-
cortex, is distinguished by a lower concentration of
vation of the cortex and also assists in the septal hip-
mu opiate receptors and a higher concentration of
pocampal pathway supporting mnemonic function.
calcium-binding proteins. his region is virtually iden-
Within the striatum, there are important inter-
tical histologically to the ventromedial caudate. In dis-
actions between the cholinergic and dopaminergic
tinction, the shell or ventromedial portion of NAc,
systems. Activation of D2 DA receptors, located
which receives a segregated pool of projections from
on cholinergic interneurons, inhibits acetylcholine
the subgenual cingulate (BA 25) and pregenual cin-
release, whereas D1 receptor agonists enhance acetyl-
gulate (BA 32), is inversely devoid of calcium-binding
choline release. Conversely, acetylcholine enhances
proteins and replete with mu opiate receptors [38].
DA release via nicotinic and muscarinic receptors
located on presynaptic DA terminals [67].
Neurotransmitter systems inluencing
frontal-subcortical circuits The norepinephrine system
Processing of the detailed information contained in
Noradrenergic neurons from the locus coeruleus
the FSCs is modulated by input from dopaminergic,
project to the entire cortex and hippocampus as well
cholinergic, noradrenergic, and serotonergic systems.
as cerebellum and spinal cord. A ventral pathway orig-
inating below the locus coeruleus innervates the brain-
The dopamine system stem and hypothalamus. Beta 1 receptors are present in
Dopaminergic projections from the SNpc (primarily the cerebral cortex while beta 2 receptors predominate
cell group A9), the caudal extension of SNpc into the in the cerebellum. Electrophysiologic studies suggest

65
Downloaded from https://www.cambridge.org/core. Stockholm University Library, on 25 Nov 2018 at 16:04:24, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781139016919.007
Section I: Structural and Functional Neuroanatomy

that a balance between norepinephrine and DA lev- A recent re-evaluation [41] supports the original
els may set the signal-to-noise ratio of the attentional concept of the NAc part of the ventral striatum as
system [68, 69], which incorporates a distributed net- a functional interface between the limbic and motor
work involving the cingulate, DLPF, and inferior pari- systems [73]. hus, its aferent connections position
etal cortices [70]. this region as a site for integration of signals with
emotional content (amygdala); contextual information
(hippocampus); motivational signiicance (dopamin-
The serotonergic system ergic inputs); information about the state of arousal
he serotonin 5-HT1 receptor is the most abundant (midline thalamus); and executive/cognitive informa-
serotonin receptor in the basal ganglia. 5-HT1C recep- tion (prefrontal cortex) [41]. Outputs of the ventral
tors are very dense in the globus pallidus and mod- striatum include projections to the brainstem dorsal
erately dense in the caudate, putamen, and accum- raphe, the midbrain parabrachial region and to the
bens regions, which also contain intermediate levels of habenula, which may also play an important role in
the 5-HT2 receptor. he highest densities of 5-HT1D integration of the motor and behavioral circuits [74].
receptors are found in the basal ganglia and SN. 5-HT3 he habenula, which receives input from the anterior
is enriched in the striatal matrix and is the most abun- thalamic nuclei and GP, projects to the rostral mid-
dant receptor in the ventral striatum [62, 71] and other brain with connections to the dopaminergic VTA and
areas functionally related to the AC circuit, including to the serotonergic dorsal raphe. Both of these cell
hippocampus, septum, and amygdala. his receptor is groups project difusely to multiple tiers of the FSCs.
linked to a ligand-gated cation channel that also mod- In particular, the limbic ventral striatal and habenular
ulates the release of acetylcholine and DA. input to the dorsal raphe suggests another important
mechanism for cross-talk between circuits and for lim-
Mechanisms for interactions between bic modulation of motor function [75].
Also providing for circuit linkage between
motor and behavioral circuits limbic and motor systems are the cholinergic
Although the FSCs remain largely segregated through- interneurons of the caudate and putamen and somato-
out their course, several mechanisms may allow for statin/neuropeptide Y-containing interneurons that
complex interactions between motor and behavioral ramify between compartments [76]. Diferences
systems [52]. At higher levels, thalamic relay nuclei in peptide cotransmitters and in monoaminergic,
form not only reciprocal but also non-reciprocal corti- GABAergic and glutamatergic synaptic receptor
cal connections, linking multiple frontal cortical areas, expression at subcortical sites (see above) provide
allowing for information low between cortical cir- other mechanisms for diferential response within
cuits [23] (see Figure 5.5 as an example). At the level FSCs and for a complex interplay between these
of the striatum, striosomes interdigitate with the sur- circuits [75].
rounding matrix, so that matrix-striosome borders From a neurophysiologic perspective, integration
may serve as interfaces where the sensorimotor sys- of the temporal coincidence of processing in FSCs,
tems of the matrix interact with the striosomal pro- occurring particularly at the level of the thalamus [74],
cessing of prefrontal and limbic inputs [72]. Linkage may also contribute importantly to linkage and synthe-
between the sensorimotor and limbic related regions sis of information between FSCs.
of the striatum can also occur through corticonigral
inluences on the striatum and through the overlap-
ping eferent striatal projections to the SN. Whereas
the main output from the striatal matrix is to the GPi Prototypical frontal-subcortical
and SNr, striosomes project primarily to the dopamin-
ergic SNpc. In this manner, the ventral striatum is able circuit syndromes
to exert a global regulatory inluence on the dopamin- hree principal frontal lobe symptom complexes are
ergic input to the entire striatum. his provides an recognizable: a medial frontal-AC syndrome with apa-
important mechanism for limbic “motivational” input thy and diminished initiative; an OF syndrome with
to modulate motor behavior and provides for the prominent disinhibition and irritability; and a DLPF
“anchoring” or reinforcement of successful experience. syndrome, with neuropsychological deicits involving

66
Downloaded from https://www.cambridge.org/core. Stockholm University Library, on 25 Nov 2018 at 16:04:24, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781139016919.007
Chapter 5: Frontal-subcortical circuits

EFs. Supporting the concept of circuit-speciic behav- humans have been associated with lesions afecting
ioral syndromes, similar disorders have been observed both rostral and caudal cingulate motor areas [46].
with lesions of subcortical structures of these circuits. Relecting reciprocal connections between the AC
cortex and the MOF cortex [51], orbito-medial pre-
The anterior cingulate syndrome: akinetic frontal cortical lesions may also result in severe forms
of apathy [46, 57]. Data suggest a functional contin-
mutism and abulia uum along the rostral-caudal axis of medial frontal
Akinetic mutism (AM) [77] represents a wakeful state lobe regions from cognitive and emotional functions
of profound apathy, with indiference to pain, thirst, to motor functions devoted to self-initiation of action
or hunger, and absence of motor or psychic initiative, and thought [57]. Relecting this continuum, a sim-
manifested by lack of spontaneous movement, absent ilar disorder afecting the “drive” for willed move-
verbalization, and failure to respond to questions or ment and speech (“motor neglect”) has been seen in
commands. he term abulia, derived from the Greek patients with lesions of the SMA [87–89]. Such patients
boul, or will [78] refers to a similar but less severe psy- exhibit initial global akinesia and neglect, which then
chomotor syndrome, encompassing lack of spontane- lateralizes in unilateral cases. Primarily part of the
ity, apathy, and paucity of speech and movement. motor circuit, the SMA also receives reciprocal pro-
Akinetic mutism has been described with AC jections from the AC (area 24c’). Positron emission
lesions, craniopharyngiomas, obstructive hydro- tomography (PET) studies in humans have shown that
cephalus, tumors in the region of the third ventricle, regional cerebral blood low in the rostral SMA and
and other conditions involving the ventral striatum adjacent mesial frontal cortex is associated with the
(NAc and ventromedial caudate), ventral GP, and self-generation of motor actions but not with exter-
medial thalamus [79]. Although reports of the clinical nally cued ones [90].
consequences of lesions restricted to limbic structures Conceptually distinct, DLPC circuit lesions
of the basal ganglia are scarce [57], larger lesions that may produce a “cognitive” rather than “emotional-
include both ventral striatum and more dorsal areas of afective” form of apathy, cognitive inertia resulting
the basal ganglia are associated with severe apathetic from diiculties in elaborating the plan of actions
states. In a review of patients with focal lesions of necessary for ongoing or forthcoming behavior [57].
the basal ganglia [80], abulia occurred in 18 of 64 his relects links between layer V of the dorsal
(28%) small and large caudate lesions sparing the cingulate cortex and supericial layers of the DLPF
lentiform nucleus, 15 of which were unilateral. Abulia cortex (see Figure 5.5), a powerful avenue of commu-
was also seen in six of 22 (27%) restricted GP lesions, nication between cognitive and motor systems [31].
all bilateral, but did not accompany isolated lesions of hus, while the AC can be considered broadly as the
the putamen (a link in the motor circuit). Lesions of cortical gateway for limbic motivation to inluence
the mediodorsal and anterior nuclei of the thalamus goal-directed behavior [26], subtypes of apathy may
may also result in apathy [81–83]. be deined based on the connections of the cingulate
Unilateral lesions of the AC cortex produce with other regions [30, 91].
transient AM [84] while the most dramatic examples
of AM follow bilateral AC lesions [85], particularly The orbitofrontal syndrome: personality and
lesions that extend from the cognitive efector region
posteriorly into the skeletomotor efector division of emotional changes
the cingulate [51], a region connected with primary he OF cortex is the neocortical representation of the
motor and SMA [29, 86]. here are non-overlapping limbic system [92] and is involved in the determination
representations for hand movements and speech of the appropriate time, place, and strategy for envi-
within the AC cortex, relecting separate AC motor ronmentally elicited behavioral responses. Lesions in
channels [29]. While there are no published reports this area disconnect frontal monitoring systems from
of isolated speech deicits with restricted cingulate limbic input [93]. In particular, visceral sensory input
lesions, bilateral lesions in monkeys to the rostral to OF cortex is normally used to provide informa-
AC cortex, located around the genu of the corpus tion about the internal milieu and guide bodily reac-
callosum, signiicantly impair spontaneous vocaliza- tions to that status (“somatic marker hypothesis”) [94].
tion whereas deicits involving limb movements in Lesions of the LOF may then result in an “interoceptic

67
Downloaded from https://www.cambridge.org/core. Stockholm University Library, on 25 Nov 2018 at 16:04:24, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781139016919.007
Section I: Structural and Functional Neuroanatomy

agnosia,” which may provide the basis for the variety of adjust task performance. Patients with restricted DLPF
emotional and social deicits commonly observed with cortex lesions have diiculty focusing and sustaining
such lesions. attention, generating hypotheses, and maintaining or
Impulsivity and behavioral disinhibition are shiting sets in response to changing task demands,
the hallmark symptoms of OF lesions. Common as required by the WCST [110]. Associated features
manifestations include lack of judgment and social include reduced verbal and design luency, impair-
tact, improper sexual remarks or gestures and other ment of memory search strategies and of organiza-
antisocial acts [95, 96]. Patients may exhibit inappro- tional and constructional strategies on learning and
priate jocularity (witzelsucht) or emotional lability copying tasks, and motor programming disturbances.
and irritability, trivial stimuli oten resulting in abrupt Similar syndromes have been reported in patients
outbursts of anger [97]. Inattention, distractibility, with lesions of subcortical structures of the DLPF
and increased motor activity may be seen, as well circuit [109]. hus, impairment on tests of memory
as hypomania or mania. MOF lesions are associated and EF, including the WCST, have been noted in
with abnormal autonomic responses to socially patients with dorsal caudate lesions [103], bilat-
meaningful behavior and diiculty extinguishing eral GP hemorrhages [111] and bilateral or let
unreinforced behavior, which correlate with antiso- paramedian/medio-dorsal thalamic infarction [112,
cial acts [98]. Large bilateral OF lobe lesions may 113]. Executive functions deicits and other features of
result in enslavement to environmental cues, with “subcortical” dementia [114] in such conditions as HD,
automatic imitation of the gestures of others, or Parkinson’s disease (PD), progressive supranuclear
enforced utilization of environmental objects [99]. palsy (PSP), Wilson’s disease, neuroacanthocytosis
Typically, however, patients with OF dysfunction and other subcortical disorders are believed to relect
exhibit a dissociation between impairment of behav- involvement of the DLPF circuit as it projects through
ior necessary for activities of daily living and normal the basal ganglia [109, 115–117].
performance on psychological tests sensitive to frontal
lobe dysfunction, such as the Wisconsin Card-Sorting Movement disorders and
Test (WCST) [93, 100, 101].
Although the OF syndrome usually follows bilat- frontal-subcortical circuits
eral OF cortex injury [93, 101], unilateral lesions Basal ganglia dysfunction frequently results not only
may produce a similar disorder [102]. Patients with in disorders of movement, but also in alterations in
ventral caudate lesions may also appear disinhibited, intellectual function, mood, personality, and behav-
euphoric, impulsive and inappropriate, reproducing ior. he nature and severity of such changes relect
the corresponding OF lobe syndrome [103]. It is likely the extent of involvement of the behaviorally relevant
that the early appearance of comparable personal- FSC structures, which project through the caudate and
ity alterations in Huntington’s disease (HD) relects ventral striatum, rather than the motor circuit, which
the involvement of medial caudate regions receiving projects to the putamen. Diseases afecting primar-
OF and AC circuit projections [104]. Similarly, mania ily the putamen, such as PD, thus exhibit less strik-
(see below) may result not only from injury to MOF ing intellectual and emotional alterations than dis-
cortex and caudate nuclei but also from lesions to eases that afect primarily the caudate, such as HD. In
the right thalamus [105–108]. Mixed behavioral syn- PD, executive dysfunction and dementia is associated
dromes commonly accompany focal lesions of the GP with involvement of the medial substantia nigra and
and thalamus, relecting the progressive spatial restric- VTA, which project to the caudate nucleus and medial
tion of the parallel circuits at these levels [109]. frontal cortex, and is not present when changes are
conined to the lateral nigral neurons, which project to
The dorsolateral prefrontal syndrome: the putamen [118]. While patients with PSP, a hypoki-
netic disease, exhibit hypoactive behaviors such as
executive function deicits apathy, patients with HD, a hyperkinetic syndrome,
Both experimental and clinical data link the DLPF cor- exhibit predominantly hyperactive behaviors, such as
tex and its subcortical connections with EFs. Exec- agitation, irritation, euphoria, or anxiety. Such behav-
utive functions incorporate anticipation, goal selec- iors may result from an excitatory subcortical output
tion, planning, monitoring, and use of feedback to through the medial and OF cortical circuits [119].

68
Downloaded from https://www.cambridge.org/core. Stockholm University Library, on 25 Nov 2018 at 16:04:24, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781139016919.007
Chapter 5: Frontal-subcortical circuits

Subcortical dementia and In OCD, functional imaging studies have shown


increased glucose metabolism or blood low in the
amnestic syndromes medial and OF cortex and AC gyrus, in the cau-
Subcortical dementia is generally characterized by date nucleus, and, to a lesser extent, in the thalamus.
neuropsychological deicits typical of DLPF circuit his has suggested aberrant OF and limbic circuits as
lesions [114, 120]. Such deicits may be combined with pathophysiologic mechanisms in OCD [130, 140, 141].
amnesia, however, when subcortical lesions involve the he prevailing theory is that the observed cortical, stri-
thalamus [109]. he thalamus is poised at the interface atal, and thalamic overactivity in OCD results from a
of the FSCs and the medial temporal-limbic circuit relative imbalance favoring the direct versus indirect
(incorporating the hippocampus, fornix, hypothala- pathways within this circuitry, leading to failed striato-
mus, and thalamus). While the FSCs mediate memory thalamic inhibition [140, 142, 143].
activation and search functions, the medial temporal- In TS, studies have suggested both anatomi-
thalamic circuit mediates memory storage (both recall cal and functional disturbances in basal ganglia-
and recognition) [113]. thalamocortical circuits. Difusion-tensor (DT)-MRI
A related syndrome involving amnesia, luctuat- has shown smaller let caudate and bilateral thala-
ing inattention, apathy and psychomotor retardation, mic volumes in TS children compared with con-
may occur with capsular genu infarction. It has been trols, tic severity being positively correlated with
inferred that such lesions interrupt the inferior and group diferences in radial water perfusion in the
anterior thalamic peduncles, functionally deactivating right thalamus [144]. Resting state luorodeoxyglucose
the ipsilateral frontal cortex [121]. he anterior thala- (FDG)–PET has suggested reduced activity in a limbic
mic peduncle conveys reciprocal connections between basal ganglia-thalamocortical network, with covariate
the thalamic dorsomedial nucleus and the cingulate decreases in caudate and thalamic metabolism asso-
gyrus, as well as the prefrontal and OF cortex; the infe- ciated with smaller reductions in lentiform and hip-
rior thalamic peduncle carries ibers which connect pocampal activity. he expression of this metabolic
with the OF, insular and temporal cortices, and amyg- pattern correlated closely with ratings on the Tourette
dala. Injury to these tracts thus produces a thalamo- Syndrome Global Scale [133]. Tic severity in TS has
cortical disconnection syndrome combining amnesia also been correlated with hypoperfusion of the let
with FSC circuit deicits. caudate and cingulate gyrus [145] and diferences in
D2 DA receptor binding in the head of the caudate
nucleus has predicted diferences in tic severity within
monozygotic twin pairs [146].
Frontal-subcortical circuit dysfunction he preceding observations link tics to the “asso-
associated with neuropsychiatric ciative” (non-motor) neural circuits in which the cau-
disorders date nucleus (not the putamen) is a key node, and
has suggested that dopaminergic dysfunction in the
Obsessive-compulsive disorder and caudate may underlie the integrated ideational-motor
symptomatology of TS and the “compulsive” quality
Tourette syndrome of tics [146]. Other studies have supported the impor-
Convergent data, including ethological and exper- tance of sensory elements in tic pathophysiology. hus,
imental observations [122, 123], clinicopathologic functional MRI (fMRI) has identiied a brain network
indings [124, 125], behavioral observations [126], of paralimbic and sensory association areas which are
magnetic resonance imaging (MRI) [127–129] and activated before tic onset, analogous to movements
PET studies [130–133] have implicated the basal gan- triggered internally by unpleasant sensations, such as
glia and related cortical and thalamic structures in itch and pain [147].
the pathobiology of both obsessive-compulsive dis- It has been hypothesized that discrete sets of stri-
order (OCD) and Gilles de la Tourette syndrome atal neurons may become overactive in TS, and that
(TS). he neurobiologic substrates for these disor- the production of simple tics (via motor circuit activa-
ders include both corticostriatothalamocortical cir- tion), complex tics (via activation of premotor areas,
cuits and monoaminergic pathways that modulate the SMAs, and cingulate motor areas), and compulsions
activity of these circuits [134–139]. (with OF circuit involvement) might be determined

69
Downloaded from https://www.cambridge.org/core. Stockholm University Library, on 25 Nov 2018 at 16:04:24, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781139016919.007
Section I: Structural and Functional Neuroanatomy

by the speciic FSC that is impacted [148]. Consistent [163]. Comparable PET studies have shown caudate
with this hypothesis, increasing complexity of cogni- hypofunction in response-inhibition paradigms [164].
tive and behavioral symptoms in TS has been associ- he relevance of these indings may relate to the inte-
ated with increasing, apparently dysfunctional synap- gration of the caudate nuclei not only in the DLPF
tic activity within the medial, lateral, and caudal OF (executive function) circuit but also in the OF circuit,
cortices on FDG-PET scans [149]. which subserves delayed responding in primates.
PET data have also suggested an alteration of Impaired signaling of delayed rewards is also inte-
cortical–subcortical interactions in TS, with increased gral to ADHD and has been linked to disturbances
metabolic rates in frontal motor regions and decreases in motivation processes. his implicates the AC cir-
in glucose utilization in paralimbic prefrontal cor- cuit [165], which links the ventral striatum, espe-
tices and in the ventral striatum [131]. Ventral stri- cially NAc [166] to both AC and OF cortex [167].
atal dysfunction has also been shown with PET using Dysfunction in NAc in ADHD is consistent with the
[11C] dihydrotetrabenazine (DTBZ) to label the type observed variability in response to psychostimulant
2 vesicular monoamine transporter, TS patients show- therapy in afected children [168]. A dual pathway
ing increased DTBZ binding in the ventral striatum hypothesis [169, 170] proposes that alterations within
relative to controls [150]. Pertinent to such indings, the DLPF circuit, modulated by mesocortical DA,
and by analogy with animal models of stereotypy, it has and the AC/reward circuit, modulated by mesolim-
been postulated that the pathophysiology of TS may bic DA, constitute discrete neuropsychologic bases for
be related to an imbalance between dorsal and ventral dissociable psychological processes in ADHD, lead-
striato-pallidal systems, perhaps arising from strioso- ing to executive/inhibitory deicits and delay aversion,
mal dysfunction [148, 151, 152]. respectively.
It is likely that both DA and norepinephrine
actions contribute to the therapeutic efects of stim-
Attention-deicit hyperactivity disorder ulants in patients with ADHD. Electrophysiologic
Characterized by inattention, impulsivity and hyper- studies in animals suggest that DA decreases “noise”
activity, attention-deicit hyperactivity disorder through modest levels of stimulation of D1 receptors,
(ADHD) shares clinical features with other neu- abundantly present in prefrontal cortex, while nore-
ropsychiatric conditions, including the OF and pinephrine enhances “signals” through post-synaptic
DLPF syndromes. It has been hypothesized that the alpha2A-adrenoceptors in prefrontal cortex [70, 171].
neural substrates of ADHD involve disturbances in Alpha2-receptor stimulation increases delay-related
frontal-subcortical interactions involving arousal and neuronal iring [172], the cellular measure of working
reward systems [153] which are driven primarily by memory and behavioral inhibition.
dopaminergic activity and modulated by adrener-
gic and serotonergic mechanisms. Dysfunction of
fronto-striatal type in ADHD has been inferred from Depression
neuropsychological studies [154–156], the pattern of Both structural and functional brain-imaging stud-
cognitive deicits resembling those found in “striatal” ies have supported an association between lesions
disorders such as TS [157] and HD [158]. disrupting frontostriatal or paralimbic pathways
Both MRI and quantitative morphologic stud- and depressed mood. OF-inferior prefrontal cortex
ies have shown smaller caudate volumes in ADHD metabolism is lower in depressed when compared
patients compared with controls [159, 160]. Xenon with non-depressed HD patients [173], and PD
inhalation and emission tomography have revealed patients with depression show signiicantly lower
striatal hypoperfusion in childhood ADHD, partially metabolic activity both in the orbital-inferior frontal
reversible by methylphenidate [161], and fMRI has cortex and head of the caudate nucleus compared
also shown diferences between ADHD children and with those without depression [174]. his is consistent
controls in frontal-striatal function and its mod- with pathological evidence in depressed, cognitively
ulation by methylphenidate [162]. Performance on impaired parkinsonian patients of disproportionate
response inhibition tasks have correlated signiicantly degeneration of DA neurons in the VTA, a system that
with fMRI measures of the prefrontal cortex and cau- is linked to motivation and reward (see below) [152]
date nuclei, predominantly in the right hemisphere and which projects to OF and prefrontal cortex.

70
Downloaded from https://www.cambridge.org/core. Stockholm University Library, on 25 Nov 2018 at 16:04:24, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781139016919.007
Chapter 5: Frontal-subcortical circuits

Frontolimbic DA deiciency may underlie clinical An important role for the subgenual cingulate (BA
similarities between the anhedonia and “psychomo- 25) in depression was irst provided by the observa-
tor retardation” (PsR) of major depression and the tion of metabolic change (overactivity) in this region
lethargy of thought, afect, and movement that consti- in treatment-resistant depression, which uniquely pre-
tutes “bradyphrenia” in PD [175]. Anhedonia has been dicted antidepressant response [184, 185]. Deep brain
linked with novelty reward, mediated by dopaminergic stimulation (DBS) of white matter tracts adjacent to
projections to the ventral striatum, and reward scores the subgenual cingulate was subsequently shown to
in response to a dopaminergic challenge correlate with efectively reverse symptoms in otherwise treatment-
activity changes in ventrolateral prefrontal cortex and resistant depression [188, 189] (see below). Distur-
caudate/putamen on fMRI in major depression [176]. bances in the subgenual cingulate circuit have been
Depressed patients with PsR show decreased presy- linked also with the rapid mood shits in bipolar dis-
naptic DA function in the let caudate [177] and ele- order, and may have a role in the pathophysiology of
vated putaminal D2 receptor binding [178]. he DA OCD [190, 191].
metabolite homovanillic acid (HVA) is diminished
in depressed patients with PsR [179] and levodopa
improves the PsR of major depression [180]. Further-
more, clinical improvement in depressed patients with Mania and the lateralization
PsR parallels the dopamimetic speciicity of the antide-
pressants administered [181]. However, DA agonists of emotional behavior
alone have limited efect on depressive symptoms in Afective response to brain injury may relect the
PD [182] and cerebrospinal luid (CSF) HVA levels do hemisphere involved. hus, crying is more common
not correlate with mood in PD [183]. in patients with let hemispheric lesions, while laugh-
Based on convergent indings from patients with ter occurs with right-sided lesions [192]. Let frontal
primary and secondary depression, a model of depres- and let basal ganglia infarctions are most likely to
sion has been proposed which implicates failure of the be associated with depression [103, 193] and in PD
coordinated interactions of a distributed network of depression is more common with right hemiparkin-
cortical-limbic pathways [184]. In this model, a dorsal sonism (let striato-frontal dysfunction) [194]. Con-
(“attention-cognition”) compartment which includes versely, mania frequently results from right-sided tha-
both neocortical (DLPF) and superior limbic (AC) ele- lamic or right medial diencephalic lesions that may
ments is postulated to regulate attentional and cogni- disrupt hypothalamic circuits or disturb modulating
tive aspects of depression, such as apathy, psychomo- transmitters traversing the medial forebrain bundle
tor slowing, and impaired attention and EF. A ventral [106, 107]. Mania has also been observed in patients
(“vegetative-circadian”) compartment, which is com- with MOF cortex lesions and caudate dysfunction in
posed of limbic, paralimbic, and subcortical regions basal ganglia disorders such as HD [193, 195]. he
(anterior insula, hippocampus, subgenual cingulate, increase in appetite drives that oten accompanies
and hypothalamus) is recognized as mediating circa- mania has suggested underlying hyperfunctioning of
dian and vegetative aspects of the illness, including the paleocortical paralimbic belt [51].
sleep, appetite, libidinal, and endocrine disturbances. Whereas depressed patients show bilateral tem-
Both PET and fMRI studies have revealed decreased poral hypometabolism on FDG-PET scans, patients
function of dorsal regions, such as the ventral AC cor- with mania exhibit unilateral, right-sided tempo-
tex, and increased limbic metabolism and activation ral hypometabolism [194]. A diferential biochemi-
in depression [185], while fMRI has shown reciprocal cal response to injury in the two hemispheres may
efects of antidepressant treatment on activity and con- contribute to the polarity of the expressed mood
nectivity in these regions [186]. he rostral anterior disorder. hus, right but not let frontolateral cor-
(pregenual) cingulate is isolated from both the ventral tical lesions in rat models produce hyperactivity,
and dorsal compartments based on its cytoarchitec- widespread depletion of brain norepinephrine, and an
tural characteristics and its reciprocal connections to increased turnover of DA in the NAc. Correspond-
both compartments and may serve an important reg- ingly, right but not let hemispheric stroke in humans
ulatory role in mediating interactions between them leads to an increase in 5-HT-2 serotonin receptor bind-
[187]. ing in both temporal and parietal cortex [194].

71
Downloaded from https://www.cambridge.org/core. Stockholm University Library, on 25 Nov 2018 at 16:04:24, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781139016919.007
Section I: Structural and Functional Neuroanatomy

Bipolar disorder small interneurons in cingulate and prefrontal cor-


tices in schizophrenic patients [212]. It has been sug-
Vulnerability to bipolar disorder (BD) is linked with
gested that if the default network in schizophrenia
the gene coding for diacylglycerol (DAG) kinase
is associated with decreased communication between
eta, an enzyme that metabolizes DAG, inhibiting
the medial prefrontal and posterior cingulate regions,
phosphatidylinositol-protein kinase C intracellular
self-monitoring systems may become split, leading to
signaling [196–198]. he phospho-inositol second
the perception that auditory thoughts are externally
messenger system is concentrated in striosomes in
produced [213].
limbic brain regions. his chemoarchitectural disor-
ganization in BD is consistent with a disease model
that involves dysfunction within both striato-thalamo-
prefrontal networks and limbic modulating regions Substance abuse disorders and impulse
[199–202]. Support for this model includes MRI control disorders
abnormalities in prefrontal cortical areas, striatum and
he principal components of the drug reward circuit
amygdala, and activation diferences in anterior limbic
are the A10 dopaminergic cell group of the VTA, lim-
regions shown by functional imaging studies. Specif-
bic structures of the basal forebrain (OF cortex, AC
ically, reduced ventral and orbital prefrontal activ-
cortex and ventral striatum, particularly NAc) and
ity and increased amygdala activity has been noted,
the dopaminergic connection between the VTA and
both during episodes and in remission. his suggests
basal forebrain limbic system (mesocorticolimbic DA
that dysregulation of mood in BD may result from
system). his network links substance addiction to
diminished prefrontal modulation of subcortical lim-
brain motivational and reward systems. Linked brain
bic structures [199, 203].
regions include the amygdala, which provides afective
salience, and the hippocampus, relaying contextual
Schizophrenia memories. While early phases of drug-seeking behav-
Failures of stimulus iltering and gating in ior and addiction are probably characterized by inter-
schizophrenic patients have been linked with actions between these systems, the dorsal striatum
abnormalities of cortico-striato-pallido-thalamic appears to become involved in later phases when drug-
circuitry [204–206]. Evidence implicating the DLPF taking has become a habit [214–218]. Other compo-
circuit in schizophrenia includes the similarity of nents of the drug reward circuit are the opioid peptide,
observed neuropsychological deicits to symptoms GABA, glutamate, and serotonin systems, and other
associated with DLPF lesions, decreased regional neural inputs that interact with the VTA and basal
metabolism and blood low activation, disruption of forebrain [219–221].
cortical subplate activity (required for connectivity Impulse control disorders (ICDs), including patho-
of thalamocortical neurons), and decreases in major logical gambling, compulsive sexual behavior, and
components of the GABA cortical inhibitory system compulsive buying, are characterized by a failure to
[207–209]. resist an impulse, drive, or temptation to perform a
he DLPF cortex is part of a task-related fronto- typically pleasurable activity. A range of ICDs occur at
parietal neuronal network, the activity of which a greater frequency in PD than in the general popula-
is anticorrelated with a “default” network that is tion and are linked particularly with the use of direct
normally active at rest. he latter network, which DA agonist drugs [222]. Compared with levodopa,
includes medial prefrontal and posterior cortices, DA agonists have signiicantly greater occupation of
has been linked with internally generated “stimulus- D3 DA receptors which are concentrated in limbic
independent” thought as well as self-monitoring and brain regions, including ventral striatum, with high-
salience monitoring [210]. Using a brain-imaging est concentrations in the shell of the NAc [223]. Even
technique based on low-frequency luctuations of within the dorsal striatum, the D3 receptor is primar-
the blood oxygen level-dependent (BOLD) signal, ily localized to the patch/striosome compartment that
schizophrenic patients exhibited striking deicits in is anatomically connected to limbic structures [224,
regions associated with the default network, including 225]. his and other evidence suggests that patholog-
the posterior cingulate and medial prefrontal regions ical gambling and other ICDs in PD may result from
[211]. his supports earlier data showing deicits in dopaminergic overstimulation of a relatively preserved

72
Downloaded from https://www.cambridge.org/core. Stockholm University Library, on 25 Nov 2018 at 16:04:24, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781139016919.007
Chapter 5: Frontal-subcortical circuits

mesocorticolimbic DA pathway in predisposed indi- by clonidine in patients with schizophrenia and Kor-
viduals [226]. sakof ’s syndrome [242–244]. In patients with demen-
Although considered by some as OCD-spectrum tia of the frontal lobe type, EF may be selectively
disorders, there is broader support for categorizing enhanced by the alpha-2 adrenergic antagonist ida-
ICDs as behavioral addictions [227–230]. Supporting zoxan [245].
such a model, a single photon emission computed In ADHD and TS, a variety of agents having impor-
tomography (SPECT) study of PD patients with patho- tant efects on the noradrenergic system, dopaminer-
logical gambling showed resting state overactivity in a gic system, or both, may ameliorate features of both
right hemisphere network that included the OF cortex, DLPF (attentional/executive) and OF (inhibitory) dys-
hippocampus, amygdala, insula, and ventral pallidum function (see below). Such drugs include selegiline,
[226]. he ventral pallidum is part of the limbic cir- stimulant medications, low-dose tricyclic antidepres-
cuitry (see above) and is implicated in the modulation sants, clonidine, and guanfacine [246–251].
of hedonic responses to natural and drug rewards,
and subsequent reward seeking-motivated drive
[231]. The anterior cingulate syndrome: akinetic
mutism and apathy
In animals, a syndrome similar to akinetic mutism
(AM) was irst demonstrated by bilateral or unilateral
Therapeutic interventions for injection of 6-hydroxydopamine into the SN, VTA, or
frontal-subcortical circuit disorders nigrostriatal tract within the medial forebrain bundles
of the lateral hypothalamus [252–254]. hese behav-
Pharmacologic interventions ioral deicits could be reversed by the direct DA agonist
apomorphine [255, 256], and blocked by pretreatment
The dorsolateral prefrontal syndrome: with the DA receptor antagonist spiroperidol [257].
executive dysfunction Similarly, in an early clinical report, AM follow-
In PD, speciic executive functions, including working ing surgical removal of a tumor from the anterior
memory, cognitive sequencing, and attention shiting, hypothalamus responded to the DA receptor agonists
may respond at least partially to dopaminergic thera- lergotrile and bromocriptine but not to the pre-
pies [232, 233]. his relects the combined impact in synaptic dopaminomimetics carbidopa/levodopa or
PD of caudate nuclear DA deiciency, which creates a methylphenidate [258]. his suggested that akinesia
partial “disconnection syndrome” of subcortical origin in this setting resulted from loss of dopaminergic
[234] and a lesser reduction of DA in the DLPF cor- input to AC or other corticolimbic structures rather
tex [235]. However, incomplete reversal of cognitive than to the striatum. Clinico-pathologic correlations
deicits with DA agonists is typically noted in PD [233, have subsequently suggested that isolated damage
236], relecting the importance of non-dopaminergic to any of the projections of brainstem dopaminergic
neuronal dysfunction, especially cholinergic dysfunc- nuclear groups may result in AM [259]. Chronic AM
tion, in PD dementia. secondary to mesencephalic infarction, destroying
Executive dysfunction in PD may also be ame- ventral tegmental DA neurons at their site of origin,
liorated by the selective norepinephrine reuptake may also be reversed with DA agonists [260]. Where
inhibitor atomoxetine [237]. his is consistent with DA receptors have been lost, however, as in patients
psychopharmacologic and anatomical studies, which with lesions involving the AC gyri, response to direct
implicate the noradrenergic as well as the dopamin- DA agonists is typically poor.
ergic system as an important modulator of frontal While requiring conirmation, a recent report sug-
lobe function [172, 238, 239]. Noradrenergic agents gests that AM may respond successfully to intramus-
may also ameliorate executive dysfunction in a vari- cular olanzapine [261], a drug which may also ame-
ety of other clinical states. he alpha-2 adrenergic ago- liorate negative symptoms in schizophrenia. In addi-
nists clonidine and guanfacine both enhance working tion to blockade of DA D2-receptors in the mesolimbic
memory performance in aged monkeys [240, 241] and pathway, olanzapine blocks serotonin 5HT2A recep-
cognitive tasks mediated by prefrontal cortex, such as tors, leading to disinhibition of D2 receptors and
Trails B, Word Fluency and Stroop tasks, are improved enhanced DA release in the mesocortical pathway.

73
Downloaded from https://www.cambridge.org/core. Stockholm University Library, on 25 Nov 2018 at 16:04:24, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781139016919.007
Section I: Structural and Functional Neuroanatomy

A preponderance of the latter action might explain paradigms [279]. In man, both propranolol and pin-
observed increases in levels of DA in the medial pre- dolol have agonist efects at limbic somato-dendritic
frontal cortex in response to olanzapine administra- 5-HT1A receptors at dosages used in the treatment of
tion [261]. aggressive behavior [280–282] and the partial 5-HT1A
Dopaminergic agents may also aford a clinically agonist buspirone may also be efective in the treat-
signiicant and sustained improvement in apathetic ment of aggression in a variety of neuropsychiatric
states encountered in a variety of neuropsychiatric conditions. In addition to their dopaminergic activity,
disorders including Wilson’s disease, PD and human neuroleptics may have a serotonergic mode of action
immunodeiciency virus (HIV)-associated dementia, in the treatment of impulsive aggression by binding
subcortical strokes and anterior communicating artery to and down-regulating the 5-HT2 receptor [274],
aneurysm [209, 262–266]. Efective agents in such con- which is represented in intermediate levels in the NAc
ditions may include direct DA agonists, particularly and striatum. Lithium’s mood-stabilizing action may
pramipexole and ropinirole, which have some selec- be mediated by efects both on the serotonin system
tivity for D3 receptors, amantadine, selegiline, bupro- and on phosphoinositide [283, 284], which is selec-
pion, amphetamine, and methylphenidate. tively concentrated in striosomes of the medial and
Apathy is the most commonly observed behav- ventral striatum [64], regions which receive dense OF
ioral disturbance in Alzheimer’s disease (AD) and is input.
associated with AC hypoperfusion [267]. he doc- Clonidine is an alpha-2 noradrenergic agonist,
umented improvement in AD-related apathy with which reduces central noradrenergic transmission by
cholinesterase inhibitor therapy [268] may relect par- stimulating presynaptic autoreceptors [285, 286]. Its
tial correction of cholinergic disconnection of AC eicacy for OF syndrome is exempliied by the report
structures. he latter include the basal nucleus of of a patient with OF dysfunction including mania sec-
the amygdala [51], innervated by cholinergic projec- ondary to bilateral OF contusions [105]. he rapid
tions from basal forebrain structures, and the mid- response to clonidine in this case was attributed to
line thalamic nuclei which receive input both from the reduction of noradrenergic overactivity induced by
basal forebrain and from cholinergic pedunculopon- lesions of prefrontal areas projecting to noradrenergic
tine projections that form part of the ascending retic- systems [287] which, in turn, innervate and modulate
ular activating system. Cholinesterase inhibitors may prefrontal cortex [238, 239, 288]. Clonidine may also
also ameliorate apathy in traumatic brain injury [269, successfully ameliorate symptoms characteristic of OF
270]. circuit dysfunction, including distractibility, impulsiv-
ity and emotional lability, in children with ADHD and
The orbitofrontal syndrome: personality change TS [246, 248, 289].
A variety of pharmacologic agents may ameliorate, at Several classes of drugs thus have the potential to
least partially, the disinhibited behavior of the patient favorably inluence symptoms of OF circuit dysfunc-
with OF circuit dysfunction [271]. Such drugs include tion, relecting serotonergic, dopaminergic, and nora-
the major and minor tranquilizers, propranolol, bus- drenergic modulation of functions of the OF cortex
pirone, carbamazepine, sodium valproate, lithium, and connected brain regions.
clonidine, and selective serotonin reuptake inhibitors
(SSRIs). Obsessive-compulsive disorder
Robust data, including studies using 5HT1B Serotonin is robustly implicated in the pathophysiol-
receptor gene knockout mice [272], link behavioral ogy of OCD and SSRIs are efective treatments for
disinhibition with central serotonergic deiciency this condition [290, 291]. he serotonergic innerva-
[273–276]. he eicacy of serotonergic agonists, tion of the striatum is dense and is localized to those
including luoxetine and clomipramine, for impulsive, basal ganglia regions which receive input from the
aggressive, or sexually disinhibited behaviors [275, OF and AC cortices, via the ventromedial caudate
277, 278] may relate to the density of serotonin nucleus head and ventral striatum, respectively [62].
receptors in the ventral striatum and other limbic Glucose metabolic rates in the head of the caudate
brain regions. Certain 5-HT1A agonists (“serenics”) nucleus diminish when OCD is treated successfully
exert a dose-dependent decrease in aggression with with SSRIs, a result that may be attributable to the
a concomitant increase in social interest in animal action of serotonin aferents from the dorsal raphe on

74
Downloaded from https://www.cambridge.org/core. Stockholm University Library, on 25 Nov 2018 at 16:04:24, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781139016919.007
Chapter 5: Frontal-subcortical circuits

caudate interneurons [130, 292]. he thalamofrontal Depression


pathways, lesioned at diferent sites in anterior capsu- Five potential targets have been identiied in the litera-
lotomy and subcaudate tractotomy (see below), con- ture as potential surgical targets for DBS in treatment-
tain both serotonergic and dopaminergic tracts [293]. resistant depression: (1) ventral striatum/NAc; (2) sub-
he improvement in OCD that may be observed with genual cingulate cortex (area 25); (3) inferior thalamic
adjunctive DA receptor blockers, particularly in TS peduncle; (4) rostral cingulate cortex (area 24a); and
[294], relects the functionally coupled interactions (5) lateral habenula [319]. Although it has been sug-
between brain 5-HT and DA systems [295]. gested that the subgenual cingulate region may prove
to be most efective, based on its anatomic connectiv-
ity [189], further studies are required in larger patient
Neurosurgical interventions groups to assess both the safety and eicacy of these
targets.
Obsessive-compulsive disorder and Tourette syndrome
Anterior cingulotomy or limbic leucotomy have been
used successfully in the past to treat disabling ritualis- Addiction
tic behaviors in selected patients with OCD [296] and In the past, procedures such as cingulotomy, hypotha-
TS [297–299]. he rationale for lesioning the anterior lamotomy, and resection of the substantia innominata
cingulate in these disorders derives from its role as the and NAc have been recommended as treatments for
conduit for frontal cortex input to the Papez circuit and severe addictive disorders. With expansion of knowl-
limbic system [300, 301], while limbic leucotomy selec- edge concerning its neurobiology, refractory addictive
tively targets both anterior cingulate cortex and fron- states might also prove amenable to DBS of relevant
tothalamic projections. he beneicial efect of anterior brain targets [320].
capsulotomy in OCD [302] would also be predicted by
the proposed models, as this procedure severs a path-
way for reciprocal tracts interconnecting the OF cor- Conclusion
tex with the dorsomedial and related thalamic nuclei Frontal-subcortical circuits (FSCs) are efector mech-
[143, 303]. More recently this has been replaced efec- anisms that allow the organism to act on the envi-
tively by DBS of the ventral anterior internal capsule ronment. he DLPF circuit allows the organization
[304], a procedure which has been shown to modu- of information to facilitate a response; AC circuitry
late activity in the dorsal and ventral striatum, subgen- is required for motivated behavior; the LOF cir-
ual cingulate cortex, MOF cortex, and thalamus [191, cuit allows the integration of limbic and emotional
305, 306]. his demonstrates how the stimulation of a information into contextually appropriate behavioral
single region can generate complex changes through- responses; and mood regulation and integration of
out the interconnected network [303]. Electrode place- information pertaining to emotions are functions,
ments in ventral caudate, STN, zona incerta (near the respectively, of the subgenual cingulate and MOF cir-
STN), and ventral striatum have also been efective for cuits. Correspondingly, impaired EFs, apathy, impul-
refractory OCD [98]. sivity, and depression are hallmarks of FSC dysfunc-
Deep brain stimulation of several targets has tion. Movement disorders typically involve not only
been used efectively to treat severe pharmacologi- the motor circuit but also other FSCs as they project
cally refractory TS. hese include the nucleus ven- through the basal ganglia. A variety of other neuropsy-
tralis oralis, the motor and limbic portions of GPi chiatric disorders may result from disturbances that
[307–311], NAc/anterior limb of the internal capsule impact directly or indirectly on the integrity or func-
[312], and the centromedian parafascicular complex tioning of these circuits. Examples of such conditions
of the median and intralaminar thalamic nuclei [311, include OCD and TS, ADHD, substance abuse and
313–317], a unit which has signiicant projections into impulse control disorders, BD and schizophrenia. he
motor striatum as well as limbic and associative areas circuits involve a number of transmitters, receptor sub-
of the subthalamic nucleus [318]. Targeting of these types, and second messengers that can be manipu-
sites oten results in at least 70% long-term reductions lated pharmacologically. In addition, for an increas-
in vocal or motor tics, with accompanying disappear- ing number of conditions, such as disabling OCD, TS
ance of the preceding sensory urge. and depression, discrete neurosurgical approaches to

75
Downloaded from https://www.cambridge.org/core. Stockholm University Library, on 25 Nov 2018 at 16:04:24, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781139016919.007
Section I: Structural and Functional Neuroanatomy

speciic FSC dysfunctions are being explored more 11. Divac I, Rosvold HE, Szwarcbart MK. Behavioral
actively, with advances in DBS showing particular efects of selective ablation of the caudate nucleus.
promise. J Comp Physiol Psychol. 1967;63(2):184–90.
12. Rosvold HE. he prefrontal cortex and caudate
nucleus: a system for efecting correction in response
mechanisms. In Rupp C, editor. Mind as a Tissue;
Acknowledgments Proceedings. New York, NY: Hoeber Medical Division;
Special thanks are ofered to John Nyquist for prepara- 1968, pp. 21–38.
tion of the igures. 13. Divac I. Neostriatum and functions of prefrontal
cortex. Acta Neurobiol Exp (Wars.) 1972;32(2):
461–77.
References
14. Johnson TN, Rosvold HE, Mishkin M. Projections
1. Alexander GE, DeLong MR, Strick PL. Parallel
from behaviorally-deined sectors of the prefrontal
organization of functionally segregated circuits linking
cortex to the basal ganglia, septum, and diencephalon
basal ganglia and cortex. Annu Rev Neurosci.
of the monkey. Exp Neurol. 1968;21(1):20–34.
1986;9:357–81.
15. Groenewegen HJ, Berendse HW, Wolters JG, Lohman
2. Alexander GE, Crutcher MD. Functional architecture
AH. he anatomical relationship of the prefrontal
of basal ganglia circuits: neural substrates of parallel
cortex with the striatopallidal system, the thalamus
processing. Trends Neurosci. 1990;13(7):266–71.
and the amygdala: evidence for a parallel organization.
3. Alexander GE, Crutcher MD, DeLong MR. Basal Prog Brain Res. 1990;85:95–116;discussion 116–18.
ganglia-thalamocortical circuits: parallel substrates for
16. Parent A. Extrinsic connections of the basal ganglia.
motor, oculomotor, “prefrontal” and “limbic”
Trends Neurosci. 1990;13(7):254–8.
functions. Prog Brain Res. 1990;85:119–46.
17. Lichter DG, Cummings J. Introduction and overview.
4. Pandya D, Barnes CL. Architecture and connections of
In Lichter DG, Cummings JL, editors. Frontal-
the frontal lobe. In Perecman E, editor. he Frontal
subcortical Circuits in Psychiatric and Neurological
Lobes Revisited. New York, NY: IRBN Press; 1987,
Disorders. New York, NY: Guilford Press; 2001,
pp. 41–72.
pp. 1–43.
5. Sanides F. Comparative architectonics of the neocortex
18. Selemon LD, Goldman-Rakic PS. Longitudinal
of mammals and their evolutionary interpretation.
topography and interdigitation of corticostriatal
Annals N Y Acad Sci. 1969;167(1):404–23.
projections in the rhesus monkey. J Neurosci.
6. Sanides F. Functional architecture of motor and 1985;5(3):776–94.
sensory cortices in primates in the light of a new
19. Parent A, Bouchard C, Smith Y. he striatopallidal and
concept of neocortex evolution. In Noback CR,
striatonigral projections: two distinct iber systems in
Montagna W, editors. he Primate Brain. New York,
primate. Brain Res. 1984;303(2):385–90.
NY: Meredith Corp.; 1970, pp. 137–209.
20. Smith Y, Hazrati LN, Parent A. Eferent projections
7. Yeterian EH, Pandya DN. Prefrontostriatal
of the subthalamic nucleus in the squirrel monkey as
connections in relation to cortical architectonic
studied by the PHA-L anterograde tracing method.
organization in rhesus monkeys. J Comp Neurol.
J Comp Neurol. 1990;294(2):306–23.
1991;312(1):43–67.
21. Ilinsky IA, Jouandet ML, Goldman-Rakic PS.
8. Yeterian EH, Van Hoesen GW. Cortico-striate
Organization of the nigrothalamocortical system in the
projections in the rhesus monkey: the organization of
rhesus monkey. J Comp Neurol. 1985;236(3):315–30.
certain cortico-caudate connections. Brain Res.
1978;139(1):43–63. 22. Kim R, Nakano K, Jayaraman A, Carpenter MB.
Projections of the globus pallidus and adjacent
9. Selemon LD, Goldman-Rakic PS. Common cortical
structures: an autoradiographic study in the monkey.
and subcortical targets of the dorsolateral prefrontal
J Comp Neurol. 1976;169(3):263–90.
and posterior parietal cortices in the rhesus monkey:
evidence for a distributed neural network subserving 23. McFarland NR, Haber SN. halamic relay nuclei of the
spatially guided behavior. J Neurosci. 1988;8(11): basal ganglia form both reciprocal and nonreciprocal
4049–68. cortical connections, linking multiple frontal cortical
areas. J Neurosci. 2002;22(18):8117–32.
10. Rosvold HE, Szwarcbart MK. Neural structures
involved in delayed response performance. In Warren 24. Kievit J, Kuypers HG. Organization of the
JM, Akert K, , editors. he Frontal Granular Cortex and thalamo-cortical connexions to the frontal lobe in the
Behavior. New York, NY: McGraw-Hill; 1964, pp. 1–15. rhesus monkey. Exp Brain Res. 1977;29(3–4):299–322.

76
Downloaded from https://www.cambridge.org/core. Stockholm University Library, on 25 Nov 2018 at 16:04:24, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781139016919.007
Chapter 5: Frontal-subcortical circuits

25. Giguere M, Goldman-Rakic PS. Mediodorsal nucleus: 38. Kopell BH, Greenberg BD. Anatomy and physiology of
areal, laminar, and tangential distribution of aferents the basal ganglia: implications for DBS in psychiatry.
and eferents in the frontal lobe of rhesus monkeys. Neurosci Biobehav Rev. 2008;32(3):408–22.
J Comp Neurol. 1988;277(2):195–213. 39. Haber SN, Lynd E, Klein C, Groenewegen HJ.
26. Mega M, Cummings J. he cingulate and cingulate Topographic organization of the ventral striatal
syndromes. In Trimble MR, Cummings JL, editors. eferent projections in the rhesus monkey: an
Contemporary Behavioral Neurology. Boston, MA: anterograde tracing study. J Comp Neurol. 1990;
Butterworth-Heinemann; 1997, pp. 189–214. 293(2):282–98.
27. Ongur D, Price JL. he organization of networks 40. Haber SN, Kunishio K, Mizobuchi M, Lynd-Balta E.
within the orbital and medial prefrontal cortex of rats, he orbital and medial prefrontal circuit through the
monkeys and humans. Cereb Cortex. 2000;10(3): primate basal ganglia. J Neurosci. 1995;15(7 Pt 1):
206–19. 4851–67.
28. Johnson TN, Rosvold HE. Topographic projections on 41. Groenewegen HJ, Trimble M. he ventral striatum as
the globus pallidus and the substantia nigra of an interface between the limbic and motor systems.
selectively placed lesions in the precommissural CNS Spectr. 2007;12(12):887–92.
caudate nucleus and putamen in the monkey. Exp 42. Haber SN, Lynd-Balta E, Mitchell SJ. he organization
Neurol. 1971;33(3):584–96. of the descending ventral pallidal projections in the
29. Middleton F, Strick PL. Revised neuroanatomy of monkey. J Comp Neurol. 1993;329(1):111–28.
frontal-subcortical circuits. In Lichter DG, Cummings 43. Ongur D, Ferry AT, Price JL. Architectonic
JL, editors. Frontal-subcortical Circuits in Psychiatric subdivision of the human orbital and medial
and Neurological Disorders. New York, NY: Guilford prefrontal cortex. J Comp Neurol. 2003;460(3):425–49.
Press; 2001, pp. 44–58.
44. Freedman LJ, Insel TR, Smith Y. Subcortical
30. Devinsky O, Morrell MJ, Vogt BA. Contributions projections of area 25 (subgenual cortex) of the
of anterior cingulate cortex to behaviour. Brain macaque monkey. J Comp Neurol. 2000;421(2):172–88.
1995;118(Pt 1):279–306.
45. Tekin S, Cummings JL. Frontal-subcortical neuronal
31. Barbas H, Pandya DN. Architecture and intrinsic circuits and clinical neuropsychiatry: an update.
connections of the prefrontal cortex in the rhesus J Psychosom Res. 2002;53(2):647–54.
monkey. J Comp Neurol. 1989;286(3):353–75.
46. Paus T. Primate anterior cingulate cortex: where
32. Morecrat RJ, Geula C, Mesulam MM. Architecture of motor control, drive and cognition interface. Nat Rev
connectivity within a cingulo-fronto-parietal Neurosci. 2001;2(6):417–24.
neurocognitive network for directed attention. Arch
Neurol. 1993;50(3):279–84. 47. Gutman DA, Holtzheimer PE, Behrens TE,
Johansen-Berg H, Mayberg HS. A tractography
33. Schmahmann JD, Pandya DN. Fiber Pathways of the analysis of two deep brain stimulation white matter
Brain. Oxford: Oxford University Press; 2006. targets for depression. Biol Psychiatry 2009;
34. Goldman-Rakic PS, Porrino LJ. he primate 65(4):276–82.
mediodorsal (MD) nucleus and its projection 48. Vogt BA, Finch DM, Olson CR. Functional
to the frontal lobe. J Comp Neurol. 1985;242(4): heterogeneity in cingulate cortex: the anterior
535–60. executive and posterior evaluative regions. Cereb
35. Bush G, Luu P, Posner MI. Cognitive and emotional Cortex 1992;2(6):435–43.
inluences in anterior cingulate cortex. Trends Cogn 49. Drevets WC, Price JL, Simpson JR, Jr. et al. Subgenual
Sci. 2000;4(6):215–22. prefrontal cortex abnormalities in mood disorders.
36. Kunishio K, Haber SN. Primate cingulostriatal Nature 1997;386(6627):824–7.
projection: limbic striatal versus sensorimotor striatal 50. Yang TT, Simmons AN, Matthews SC et al. Adolescent
input. J Comp Neurol. 1994;350(3):337–56. subgenual anterior cingulate activity is related to harm
37. Heimer L. he olfactory cortex and the ventral avoidance. Neuroreport 2009;20(1):19–23.
triatum. In Livingston KE, Hornykiewicz O, editors. 51. Mega MS, Cummings JL, Salloway S, Malloy P.
Limbic Mechanisms: he Continuing Evolution of the he limbic system: an anatomic, phylogenetic, and
Limbic System Concept. Proceedings of the Limbic clinical perspective. J Neuropsychiatry Clin Neurosci.
System Symposium held at the University of Toronto, 1997;9(3):315–30.
ON, Canada, November 5–6, 1976, as a satellite to the
sixth annual meeting of the Society of Neuroscience. 52. Haber SN. he primate basal ganglia: parallel and
New York, NY: Plenum Press; 1978. integrative networks. J Chem Neuroanat. 2003;
26(4):317–30.

77
Downloaded from https://www.cambridge.org/core. Stockholm University Library, on 25 Nov 2018 at 16:04:24, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781139016919.007
Section I: Structural and Functional Neuroanatomy

53. An X, Ongur D, Price J. Prefrontostriatal projections thalamus and brainstem in cats and monkeys. J Comp
in relation to cortico-cortical networks in the macaque Neurol. 1988;277(2):281–301.
monkey. Soc Neurosci Abstr. 1997;23:901. 67. McGeer PL, McGeer EG. Neurotransmitters and
54. Vogt BA, Pandya DN. Cingulate cortex of the rhesus their receptors in the basal ganglia. Adv Neurol.
monkey: II. Cortical aferents. J Comp Neurol. 1993;60:93–101.
1987;262(2):271–89. 68. Daniel DG, Weinberger DR, Jones DW et al. he efect
55. Morecrat RJ, Geula C, Mesulam MM. of amphetamine on regional cerebral blood low
Cytoarchitecture and neural aferents of orbitofrontal during cognitive activation in schizophrenia.
cortex in the brain of the monkey. J Comp Neurol. J Neurosci. 1991;11(7):1907–17.
1992;323(3):341–58. 69. Sawaguchi T. Catecholamine sensitivities of neurons
56. Pandya DN, Van Hoesen GW, Mesulam MM. Eferent related to a visual reaction time task in the monkey
connections of the cingulate gyrus in the rhesus prefrontal cortex. J Neurophysiol. 1987;58(5):1100–22.
monkey. Exp Brain Res. 1981;42(3–4):319–30. 70. Arnsten AF. Fundamentals of attention-deicit/
57. Levy R, Dubois B. Apathy and the functional anatomy hyperactivity disorder: circuits and pathways. J Clin
of the prefrontal cortex-basal ganglia circuits. Cereb Psychiatry 2006;67 (Suppl. 8):7–12.
Cortex. 2006;16(7):916–28. 71. Hoyer D, Palacios JM, Mengod G. 5-HT receptor
58. Barbas H. Specialized elements of orbitofrontal distribution in the human brain: autoradiographic
cortex in primates. Ann N Y Acad Sci. 2007;1121: studies. In Marsden CA, Heal DJ, editors. Central
10–32. Serotonin Receptors and Psychotropic Drugs. London:
59. Albin RL, Young AB, Penney JB. he functional Blackwell Scientiic Publications; 1992, pp. 100–25.
anatomy of basal ganglia disorders. Trends Neurosci. 72. Flaherty AW, Graybiel AM. Anatomy of the basal
1989;12(10):366–75. ganglia. In Marsden CD, Fahn S, editors. Movement
60. Groenewegen HJ, Roeling TAP, Voorn P, Berendse Disorders. Boston: Butterworth-Heinemann Ltd.;
HW. he parallel arrangement of basal 1994, pp. 3–27.
ganglia-thalamocortical circuits: a neuronal substrate 73. Mogenson GJ, Jones DL, Yim CY. From motivation to
for the role of dopamine in motor and cognitive action: functional interface between the limbic system
functions. In Wolters EC, Scheltens P, editors. Mental and the motor system. Prog Neurobiol. 1980;14(2–3):
Dysfunction in Parkinson’s Disease. Amsterdam: Vrije 69–97.
Universiteit; 1993, pp. 3–18. 74. Scheibel AB. he thalamus and neuropsychiatric
61. Graybiel AM. Neurotransmitters and illness. J Neuropsychiatry Clin Neurosci. 1997;
neuromodulators in the basal ganglia. Trends Neurosci. 9(3):342–53.
1990;13(7):244–54. 75. Salloway S, Cummings J. Subcortical structures and
62. Lavoie B, Parent A. Immunohistochemical study of the neuropsychiatric illness. Neuroscientist 1996;2(1):
serotoninergic innervation of the basal ganglia in the 66–75.
squirrel monkey. J Comp Neurol. 1990;299(1):1–16. 76. Gerfen CR. he neostriatal mosaic:
63. Chesselet MF, Gonzales C, Levitt P. Heterogeneous compartmentalization of corticostriatal input and
distribution of the limbic system-associated membrane striatonigral output systems. Nature 1984;
protein in the caudate nucleus and substantia nigra of 311(5985):461–4.
the cat. Neuroscience 1991;40(3):725–33. 77. Cairns H, Oldield R, Pennybacker JB, Whitteridge
64. Gibb WR. Melanin, tyrosine hydroxylase, calbindin DC. Akinetic mutism with an epidermoid cyst at the
and substance P in the human midbrain and third ventricle. Brain. 1941;64:275–90.
substantia nigra in relation to nigrostriatal projections 78. Auerbach S. Beitrag zur diagnostik der geschwülste
and diferential neuronal susceptibility in Parkinson’s des stirnhirns. J Neurol. 1902;22(3):312–32.
disease. Brain Res. 1992;581(2):283–91.
79. van Domburg PH, ten Donkelaar HJ, Notermans SL.
65. Fotuhi M, Dawson TM, Sharp AH et al. Akinetic mutism with bithalamic infarction.
Phosphoinositide second messenger system is Neurophysiological correlates. J Neurol Sci.
enriched in striosomes: immunohistochemical 1996;139(1):58–65.
demonstration of inositol 1,4,5-trisphosphate
receptors and phospholipase C beta and gamma in 80. Bhatia KP, Marsden CD. he behavioural and motor
primate basal ganglia. J Neurosci. 1993;13(8):3300–8. consequences of focal lesions of the basal ganglia in
man. Brain 1994;117 (Pt 4):859–76.
66. Parent A, Pare D, Smith Y, Steriade M. Basal forebrain
cholinergic and noncholinergic projections to the 81. Engelborghs S, Marien P, Pickut BA, Verstraeten S, De
Deyn PP. Loss of psychic self-activation ater

78
Downloaded from https://www.cambridge.org/core. Stockholm University Library, on 25 Nov 2018 at 16:04:24, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781139016919.007
Chapter 5: Frontal-subcortical circuits

paramedian bithalamic infarction. Stroke 2000; 97. Stuss DT, Gow CA, Hetherington CR. “No longer
31(7):1762–5. Gage”: frontal lobe dysfunction and emotional
82. Bogousslavsky J, Regli F, Delaloye B et al. Loss of changes. J Consult Clin Psychol. 1992;60(3):349–59.
psychic self-activation with bithalamic infarction. 98. Chow TW, Cummings J. Frontal-subcortical circuits.
Neurobehavioural, CT, MRI and SPECT correlates. In Miller BL, Cummings JL, editors. he Human
Acta Neurol Scand. 1991;83(5):309–16. Frontal Lobes: Functions and Disorders. 2nd edition.
83. Ghika-Schmid F, Bogousslavsky J. he acute New York, NY: Guilford Press; 2007, pp. 25–73.
behavioral syndrome of anterior thalamic infarction: 99. Lhermitte F, Pillon B, Serdaru M. Human autonomy
a prospective study of 12 cases. Ann Neurol. 2000; and the frontal lobes. Part I: Imitation and utilization
48(2):220–7. behavior: a neuropsychological study of 75 patients.
84. Damasio H, Damasio AR. Lesion Analysis in Ann Neurol. 1986;19(4):326–34.
Neuropsychology. New York, NY: Oxford University 100. Laiacona M, De Santis A, Barbarotto R et al.
Press; 1989. Neuropsychological follow-up of patients operated for
85. Barris RW, Schuman HR. [Bilateral anterior cingulate aneurysms of anterior communicating artery. Cortex
gyrus lesions; syndrome of the anterior cingulate gyri.] 1989;25(2):261–73.
Neurology 1953;3(1):44–52. 101. Vanderploeg RD, Haley JA. Pseudosociopathy with
86. Koski L, Paus T. Functional connectivity of the intact higher-order cognitive abilities in patients with
anterior cingulate cortex within the human frontal orbitofrontal cortical damage. J Clin Exp Neuropsychol.
lobe: a brain-mapping meta-analysis. Exp Brain Res. 1990;12:54–5.
2000;133(1):55–65. 102. Meyers CA, Berman SA, Scheibel RS, Hayman A. Case
87. Damasio AR, Vanhoesen GW. Structure and function report: acquired antisocial personality disorder
of the supplementary motor area. Neurology associated with unilateral let orbital frontal lobe
1980;30(4):359. damage. J Psychiatry Neurosci. 1992;17(3):121–5.
88. Laplane D, Degos JD. Motor neglect. J Neurol 103. Mendez MF, Adams NL, Lewandowski KS.
Neurosurg Psychiatry 1983;46(2):152–8. Neurobehavioral changes associated with caudate
lesions. Neurology 1989;39(3):349–54.
89. von Giesen HJ, Schlaug G, Steinmetz H et al. Cerebral
network underlying unilateral motor neglect: evidence 104. Vonsattel JP, Myers RH, Stevens TJ et al.
from positron emission tomography. J Neurol Sci. Neuropathological classiication of Huntington’s
1994;125(1):29–38. disease. J Neuropathol Exp Neurol. 1985;44(6):559–77.
90. Jenkins IH, Jahanshahi M, Jueptner M, Passingham 105. Bakchine S, Lacomblez L, Benoit N et al. Manic-like
RE, Brooks DJ. Self-initiated versus externally state ater bilateral orbitofrontal and right
triggered movements. II. he efect of movement temporoparietal injury: eicacy of clonidine.
predictability on regional cerebral blood low. Brain Neurology 1989;39(6):777–81.
2000;123 (Pt 6):1216–28. 106. Bogousslavsky J, Ferrazzini M, Regli F et al. Manic
91. Stuss DT. Diferentiation of states and causes of apathy. delirium and frontal-like syndrome with paramedian
In Borod JC, editor. he Neuropsychology of Emotion. infarction of the right thalamus. J Neurol Neurosurg
New York, NY: Oxford University Press; 2000, Psychiatry 1988;51(1):116–19.
pp. 340–63. 107. Cummings JL, Mendez MF. Secondary mania with
92. Nauta WJ. he problem of the frontal lobe: a focal cerebrovascular lesions. Am J Psychiatry
reinterpretation. J Psychiatr Res. 1971;8(3):167–87. 1984;141(9):1084–7.
93. Eslinger PJ, Damasio AR. Severe disturbance of higher 108. Starkstein SE, Boston JD, Robinson RG. Mechanisms
cognition ater bilateral frontal lobe ablation: patient of mania ater brain injury. 12 case reports and review
EVR. Neurology 1985;35(12):1731–41. of the literature. J Nerv Ment Dis. 1988;176(2):87–100.
94. Damasio AR. Descartes’ Error: Emotion, Reason, and 109. Cummings JL. Frontal-subcortical circuits and human
the Human Brain. New York, NY: Putnam; 1994. behavior. Arch Neurol. 1993;50(8):873–80.
95. Blumer D, Benson DF. Personality changes with 110. Milner B. Efects of diferent brain lesions on card
frontal and temporal lobe lesions. In Benson DF, sorting: the role of the frontal lobes. Arch Neurol.
Blumer D, editors. Psychiatric Aspects of Neurological 1963;9(1):90–100.
Disease. New York, NY: Grune & Stratton; 1975. 111. Strub RL. Frontal lobe syndrome in a patient with
96. Reitman F. Orbital cortex syndrome following bilateral globus pallidus lesions. Arch Neurol.
leucotomy. Am J Psychiatry 1946;103(2):238–41. 1989;46(9):1024–7.

79
Downloaded from https://www.cambridge.org/core. Stockholm University Library, on 25 Nov 2018 at 16:04:24, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781139016919.007
Section I: Structural and Functional Neuroanatomy

112. Sandson TA, Dafner KR, Carvalho PA, Mesulam MM. evidence for cortico-striatal network dysfunction?
Frontal lobe dysfunction following infarction of the J Psychopharmacol. 2000;14(1):37–9.
let-sided medial thalamus. Arch Neurol. 1991;48(12): 127. Robinson D, Wu H, Munne RA et al. Reduced caudate
1300–3. nucleus volume in obsessive-compulsive disorder.
113. Stuss DT, Guberman A, Nelson R, Larochelle S. he Arch Gen Psychiatry 1995;52(5):393–8.
neuropsychology of paramedian thalamic infarction. 128. Peterson B, Riddle MA, Cohen DJ et al. Reduced basal
Brain Cogn. 1988;8(3):348–78. ganglia volumes in Tourette’s syndrome using
114. Albert ML, Feldman RG, Willis AL. he ‘subcortical three-dimensional reconstruction techniques from
dementia’ of progressive supranuclear palsy. J Neurol magnetic resonance images. Neurology 1993;
Neurosurg Psychiatry 1974;37(2):121–30. 43(5):941–9.
115. Peinemann A, Schuller S, Pohl C et al. Executive 129. Singer HS, Reiss AL, Brown JE et al. Volumetric MRI
dysfunction in early stages of Huntington’s disease is changes in basal ganglia of children with Tourette’s
associated with striatal and insular atrophy: a syndrome. Neurology 1993;43(5):950–6.
neuropsychological and voxel-based morphometric 130. Baxter LR, Jr., Schwartz JM, Bergman KS et al.
study. J Neurol Sci. 2005;239(1):11–19. Caudate glucose metabolic rate changes with both
116. Sawamoto N, Piccini P, Hotton G et al. Cognitive drug and behavior therapy for obsessive-compulsive
deicits and striato-frontal dopamine release in disorder. Arch Gen Psychiatry 1992;49(9):681–9.
Parkinson’s disease. Brain 2008;131(Pt 5):1294–302. 131. Braun AR, Stoetter B, Randolph C et al. he functional
117. Owen AM. Cognitive dysfunction in Parkinson’s neuroanatomy of Tourette’s syndrome: an FDG-PET
disease: the role of frontostriatal circuitry. study. I. Regional changes in cerebral glucose
Neuroscientist 2004;10(6):525–37. metabolism diferentiating patients and controls.
118. Rinne JO, Rummukainen J, Paljarvi L, Rinne UK. Neuropsychopharmacology 1993;9(4):277–91.
Dementia in Parkinson’s disease is related to neuronal 132. Jefries KJ, Schooler C, Schoenbach C et al. he
loss in the medial substantia nigra. Ann Neurol. functional neuroanatomy of Tourette’s syndrome: an
1989;26(1):47–50. FDG PET study III: functional coupling of regional
119. Litvan I, Paulsen JS, Mega MS, Cummings JL. cerebral metabolic rates. Neuropsychopharmacology
Neuropsychiatric assessment of patients with 2002;27(1):92–104.
hyperkinetic and hypokinetic movement disorders. 133. Eidelberg D, Moeller JR, Antonini A et al. he
Arch Neurol. 1998;55(10):1313–19. metabolic anatomy of Tourette’s syndrome. Neurology
120. Cummings J (editor). Introduction. In Subcortical 1997;48(4):927–34.
Dementia. New York, NY: Oxford University Press; 134. Gerard E, Peterson BS. Developmental processes
1990, pp. 3–16. and brain imaging studies in Tourette syndrome.
121. Tatemichi TK, Desmond DW, Prohovnik I et al. J Psychosom Res. 2003;55(1):13–22.
Confusion and memory loss from capsular genu 135. Singer HS, Minzer K. Neurobiology of Tourette’s
infarction: a thalamocortical disconnection syndrome? syndrome: concepts of neuroanatomic localization and
Neurology 1992;42(10):1966–79. neurochemical abnormalities. Brain Dev. 2003;25
122. Kolb B. Studies on the caudate-putamen and the (S1):S70–84.
dorsomedial thalamic nucleus of the rat: implications 136. Peterson B, Leckman JF, Arnsten AF. Neuroanatomical
for mammalian frontal-lobe functions. Physiol Behav. circuitry. In Leckman JF, Cohen DJ, editors. Tourette’s
1977;18(2):237–44. Syndrome – Tics, Obsessions, Compulsions:
123. Rapoport JL. Recent advances in obsessive-compulsive Developmental Psychopathology and Clinical Care.
disorder. Neuropsychopharmacology 1991;5(1):1–10. New York, NY: John Wiley & Sons; 1998, pp. 230–60.
124. Cummings JL, Cunningham K. Obsessive-compulsive 137. Leckman JF, Knorr AM, Rasmusson AM, Cohen DJ.
disorder in Huntington’s disease. Biol Psychiatry Basal ganglia research and Tourette’s syndromes.
1992;31(3):263–70. Trends Neurosci. 1991;14(3):94.
125. Swedo SE, Rapoport JL, Cheslow DL et al. High 138. Leckman JF, Pauls DL, Peterson BS et al. Pathogenesis
prevalence of obsessive-compulsive symptoms in of Tourette syndrome. Clues from the clinical
patients with Sydenham’s chorea. Am J Psychiatry phenotype and natural history. Adv Neurol.
1989;146(2):246–9. 1992;58:15–24.
126. Dursun SM, Burke JG, Reveley MA. Antisaccade eye 139. Leckman JF, Walker DE, Goodman WK, Pauls DL,
movement abnormalities in Tourette syndrome: Cohen DJ. “Just right” perceptions associated with

80
Downloaded from https://www.cambridge.org/core. Stockholm University Library, on 25 Nov 2018 at 16:04:24, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781139016919.007
Chapter 5: Frontal-subcortical circuits

compulsive behavior in Tourette’s syndrome. Am J 152. Canales JJ, Graybiel AM. A measure of striatal
Psychiatry 1994;151(5):675–80. function predicts motor stereotypy. Nat Neurosci.
140. Modell JG, Mountz JM, Curtis GC, Greden JF. 2000;3(4):377–83.
Neurophysiologic dysfunction in basal ganglia/ 153. Wise RA. Catecholamine theories of reward: a critical
limbic striatal and thalamocortical circuits as a review. Brain Res. 1978;152(2):215–47.
pathogenetic mechanism of obsessive-compulsive 154. Barkley RA, Grodzinsky G, DuPaul GJ. Frontal lobe
disorder. J Neuropsychiatry Clin Neurosci. 1989;1(1): functions in attention deicit disorder with and
27–36. without hyperactivity: a review and research report.
141. Insel TR. Toward a neuroanatomy of J Abnorm Child Psychol. 1992;20(2):163–88.
obsessive-compulsive disorder. Arch Gen Psychiatry 155. Shue KL, Douglas VI. Attention deicit hyperactivity
1992;49(9):739–44. disorder and the frontal lobe syndrome. Brain Cogn.
142. Rauch SL. Neuroimaging and neurocircuitry models 1992;20(1):104–24.
pertaining to the neurosurgical treatment of 156. Kemp SL, Kirk U. An investigation of frontal executive
psychiatric disorders. Neurosurg Clin N Am. dysfunction in attention deicit disorder subgroups.
2003;14(2):213–23, vii–viii. Ann N Y Acad Sci. 1993;682:363–5.
143. Masterman DL, Cummings JL. Frontal-subcortical 157. Towbin KE, Riddle MA. Attention deicit hyperactivity
circuits: the anatomic basis of executive, social and disorder. In Kurlan R, editor. Handbook of Tourette’s
motivated behaviors. J Psychopharmacol. 1997; Syndrome and Related Tic and Behavioral Disorders.
11(2):107–14. New York, NY: M. Dekker; 1993, pp. 89–109.
144. Makki MI, Behen M, Bhatt A, Wilson B, Chugani HT. 158. Bamford KA, Caine ED, Kido DK, Plassche WM,
Microstructural abnormalities of striatum and Shoulson I. Clinical-pathologic correlation in
thalamus in children with Tourette syndrome. Mov Huntington’s disease: a neuropsychological and
Disord. 2008;23(16):2349–56. computed tomography study. Neurology 1989;
145. Moriarty J, Costa DC, Schmitz B et al. Brain perfusion 39(6):796–801.
abnormalities in Gilles de la Tourette’s syndrome. Br J 159. Hynd GW, Hern KL, Novey ES et al. Attention
Psychiatry 1995;167(2):249–54. deicit-hyperactivity disorder and asymmetry of the
146. Wolf SS, Jones DW, Knable MB et al. Tourette caudate nucleus. J Child Neurol. 1993;8(4):339–47.
syndrome: prediction of phenotypic variation in 160. Castellanos FX, Giedd JN, Eckburg P et al.
monozygotic twins by caudate nucleus D2 Quantitative morphology of the caudate nucleus in
receptor binding. Science 1996;273(5279): attention deicit hyperactivity disorder. Am J
1225–7. Psychiatry 1994;151(12):1791–6.
147. Bohlhalter S, Goldine A, Matteson S et al. Neural 161. Lou HC, Henriksen L, Bruhn P, Borner H, Nielsen JB.
correlates of tic generation in Tourette syndrome: Striatal dysfunction in attention deicit and
an event-related functional MRI study. Brain hyperkinetic disorder. Arch Neurol. 1989;46(1):48–52.
2006;129(8):2029–37.
162. Vaidya CJ, Austin G, Kirkorian G et al. Selective efects
148. Mink JW. Neurobiology of basal ganglia and Tourette of methylphenidate in attention deicit hyperactivity
syndrome: basal ganglia circuits and thalamocortical disorder: a functional magnetic resonance study. Proc
outputs. Adv Neurol. 2006;99:89–98. Natl Acad Sci USA 1998;95(24):14 494–9.
149. Braun AR, Randolph C, Stoetter B et al. he functional 163. Casey BJ, Castellanos FX, Giedd JN et al. Implication
neuroanatomy of Tourette’s syndrome: an FDG-PET of right frontostriatal circuitry in response inhibition
Study. II: Relationships between regional cerebral and attention-deicit/hyperactivity disorder. J Am
metabolism and associated behavioral and cognitive Acad Child Adolesc Psychiatry 1997;36(3):374–83.
features of the illness. Neuropsychopharmacology
1995;13(2):151–68. 164. Dickstein SG, Bannon K, Castellanos FX, Milham MP.
he neural correlates of attention deicit hyperactivity
150. Albin RL, Koeppe RA, Bohnen NI et al. Increased disorder: an ALE meta-analysis. J Child Psychol
ventral striatal monoaminergic innervation in Psychiatry 2006;47(10):1051–62.
Tourette syndrome. Neurology 2003;1(3):310–15.
165. McClure SM, York MK, Montague PR. he neural
151. Groenewegen HJ, van den Heuvel OA, Cath DC, substrates of reward processing in humans: the
Voorn P, Veltman DJ. Does an imbalance between the modern role of FMRI. Neuroscientist 2004;10(3):
dorsal and ventral striatopallidal systems play a role in 260–8.
Tourette’s syndrome? A neuronal circuit approach.
Brain Dev. 2003;25(S1):S3–S14. 166. Zink CF, Pagnoni G, Martin-Skurski ME, Chappelow
JC, Berns GS. Human striatal responses to monetary

81
Downloaded from https://www.cambridge.org/core. Stockholm University Library, on 25 Nov 2018 at 16:04:24, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781139016919.007
Section I: Structural and Functional Neuroanatomy

reward depend on saliency. Neuron 2004;42(3): 179. Praag HM, Korf J, Lakke J, Schut T. Dopamine
509–17. metabolism in depressions, psychoses, and Parkinson’s
167. Kringelbach ML, Rolls ET. he functional disease: the problem of the speciicity of biological
neuroanatomy of the human orbitofrontal cortex: variables in behaviour disorders. Psychol Med.
evidence from neuroimaging and neuropsychology. 1975;5(2):138–46.
Prog Neurobiol. 2004;72(5):341–72. 180. Goodwin FK, Murphy DL, Brodie HK, Bunney WE,
168. Voeller KK. What can neurological models of Jr. L-DOPA, catecholamines, and behavior: a clinical
attention, intention, and arousal tell us about and biochemical study in depressed patients. Biol
attention-deicit hyperactivity disorder? Psychiatry 1970;2(4):341–66.
J Neuropsychiatry Clin Neurosci. 1991;3(2): 181. Rampello L, Nicoletti G, Rafaele R. Dopaminergic
209–16. hypothesis for retarded depression: a symptom proile
169. Sonuga-Barke EJ. Psychological heterogeneity in for predicting therapeutical responses. Acta Psychiatr
AD/HD – a dual pathway model of behaviour and Scand. 1991;84(6):552–4.
cognition. Behav Brain Res. 2002;130(1–2): 182. Marsh GG, Markham CH. Does levodopa alter
29–36. depression and psychopathology in Parkinsonism
170. Sonuga-Barke EJ. he dual pathway model of AD/HD: patients? J Neurol Neurosurg Psychiatry 1973;
an elaboration of neuro-developmental characteristics. 36(6):925–35.
Neurosci Biobehav Rev. 2003;27(7):593–604. 183. Mayeux R, Stern Y, Williams JB et al. Clinical and
171. Brennan AR, Arnsten AF. Neuronal mechanisms biochemical features of depression in Parkinson’s
underlying attention deicit hyperactivity disorder: the disease. Am J Psychiatry 1986;143(6):756–9.
inluence of arousal on prefrontal cortical function. 184. Mayberg HS. Limbic-cortical dysregulation: a
Ann N Y Acad Sci. 2008;1129:236–45. proposed model of depression. J Neuropsychiatry Clin
172. Li BM, Mao ZM, Wang M, Mei ZT. Alpha-2 adrenergic Neurosci. 1997;9(3):471–81.
modulation of prefrontal cortical neuronal activity 185. Mayberg HS, Liotti M, Brannan SK et al. Reciprocal
related to spatial working memory in monkeys. limbic-cortical function and negative mood:
Neuropsychopharmacology 1999;21(5):601–10. converging PET indings in depression and normal
173. Mayberg HS, Starkstein SE, Peyser CE et al. Paralimbic sadness. Am J Psychiatry 1999;156(5):675–82.
frontal lobe hypometabolism in depression associated 186. Anand A, Li Y, Wang Y, Gardner K, Lowe MJ.
with Huntington’s disease. Neurology 1992;42(9): Reciprocal efects of antidepressant treatment on
1791–7. activity and connectivity of the mood regulating
174. Mayberg HS, Starkstein SE, Sadzot B et al. Selective circuit: an FMRI study. J Neuropsychiatry Clin
hypometabolism in the inferior frontal lobe in Neurosci. 2007;19(3):274–82.
depressed patients with Parkinson’s disease. Ann 187. Mayberg HS. Depression and frontal-subcortical
Neurol. 1990;28(1):57–64. circuits: focus on prefrontal-limbic interactions. In
175. Rogers D. Bradyphrenia in Parkinson’s disease. In Lichter DG, Cummings JL, editors. Frontal-subcortical
Huber SJ, Cummings JL, editors. Parkinson’s Disease: Circuits in Psychiatric and Neurological Disorders. New
Neurobehavioral Aspects. New York, NY: Oxford York, NY: Guilford Press; 2001, pp. 177–206.
University Press; 1992, pp. 86–96. 188. Mayberg HS, Lozano AM, Voon V et al. Deep brain
176. Tremblay LK, Naranjo CA, Graham SJ et al. Functional stimulation for treatment-resistant depression. Neuron
neuroanatomical substrates of altered reward 2005;45(5):651–60.
processing in major depressive disorder revealed by a 189. Johansen-Berg H, Gutman DA, Behrens TE et al.
dopaminergic probe. Arch Gen Psychiatry 2005; Anatomical connectivity of the subgenual cingulate
62(11):1228–36. region targeted with deep brain stimulation for
177. Martinot M, Bragulat V, Artiges E et al. Decreased treatment-resistant depression. Cereb Cortex
presynaptic dopamine function in the let caudate of 2008;18(6):1374–83.
depressed patients with afective lattening and 190. Drevets WC, Savitz J, Trimble M. he subgenual
psychomotor retardation. Am J Psychiatry 2001; anterior cingulate cortex in mood disorders. CNS
158(2):314–16. Spectr. 2008;13(8):663–81.
178. Meyer JH, McNeely HE, Sagrati S et al. Elevated 191. Van Laere K, Nuttin B, Gabriels L et al. Metabolic
putamen D(2) receptor binding potential in major imaging of anterior capsular stimulation in refractory
depression with motor retardation: an [11C]raclopride obsessive-compulsive disorder: a key role for the
positron emission tomography study. Am J Psychiatry subgenual anterior cingulate and ventral striatum.
2006;163(9):1594–602. J Nucl Med. 2006;47(5):740–7.

82
Downloaded from https://www.cambridge.org/core. Stockholm University Library, on 25 Nov 2018 at 16:04:24, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781139016919.007
Chapter 5: Frontal-subcortical circuits

192. Sackeim HA, Greenberg MS, Weiman AL et al. and cerebral cortex in schizophrenia. Arch Gen
Hemispheric asymmetry in the expression of positive Psychiatry 1991;48(10):881–90.
and negative emotions. Neurologic evidence. Arch 207. Grebb JA, Weinberger DR, Wyatt RJ. Schizophrenia. In
Neurol. 1982;39(4):210–18. Asbury AK, McKahann GM, McDonald WI, editors.
193. Starkstein SE, Robinson RG, Price TR. Comparison Diseases of the Nervous System: Clinical Neurobiology.
of cortical and subcortical lesions in the production 2nd edition. Philadelphia, PA: W.B. Saunders; 1992,
of poststroke mood disorders. Brain 1987;110 pp. 839–48.
(Pt 4):1045–59. 208. Goldman-Rakic PS, Selemon LD. Functional and
194. Starkstein SE, Preziosi TJ, Bolduc PL, Robinson RG. anatomical aspects of prefrontal pathology in
Depression in Parkinson’s disease. J Nerv Ment Dis. schizophrenia. Schizophr Bull. 1997;23(3):437–58.
1990;178(1):27–31. 209. Bunney WE, Bunney BG. Evidence for a compromised
195. Jorge RE, Robinson RG, Starkstein SE et al. Secondary dorsolateral prefrontal cortical parallel circuit in
mania following traumatic brain injury. Am J schizophrenia. Brain Res Brain Res Rev. 2000;
Psychiatry 1993;150(6):916–21. 31(2–3):138–46.
196. Newberg AR, Catapano LA, Zarate CA, Manji HK. 210. Gusnard DA, Raichle ME. Searching for a baseline:
Neurobiology of bipolar disorder. Expert Rev functional imaging and the resting human brain. Nat
Neurother. 2008;8(1):93–110. Rev Neurosci. 2001;2(10):685–94.
197. Arnsten AF. Ameliorating prefrontal cortical 211. Garrity AG, Pearlson GD, McKiernan K et al. Aberrant
dysfunction in mental illness: inhibition of “default mode” functional connectivity in
phosphotidyl inositol-protein kinase C signaling. schizophrenia. Am J Psychiatry 2007;164(3):
Psychopharmacology (Berl.) 2009;202(1–3): 450–7.
445–55. 212. Benes FM, McSparren J, Bird ED, SanGiovanni JP,
198. Manji HK, Lenox RH. Protein kinase C signaling in Vincent SL. Deicits in small interneurons in
the brain: molecular transduction of mood prefrontal and cingulate cortices of schizophrenic and
stabilization in the treatment of manic-depressive schizoafective patients. Arch Gen Psychiatry
illness. Biol Psychiatry 1999;46(10):1328–51. 1991;48(11):996–1001.
199. Strakowski SM, Delbello MP, Adler CM. he 213. Williamson P. Are anticorrelated networks in the brain
functional neuroanatomy of bipolar disorder: a review relevant to schizophrenia? Schizophr Bull. 2007;
of neuroimaging indings. Mol Psychiatry 33(4):994–1003.
2005;10(1):105–16. 214. Everitt BJ, Wolf ME. Psychomotor stimulant addiction:
200. DelBello MP, Adler CM, Strakowski SM. he a neural systems perspective. J Neurosci. 2002;
neurophysiology of childhood and adolescent bipolar 22(9):3312–20.
disorder. CNS Spectr. 2006;11(4):298–311. 215. Everitt BJ, Robbins TW. Neural systems of
201. Adler CM, DelBello MP, Strakowski SM. Brain reinforcement for drug addiction: from actions to
network dysfunction in bipolar disorder. CNS Spectr. habits to compulsion. Nat Neurosci. 2005;
2006;11(4):312–20. 8(11):1481–9.
202. Caetano SC, Olvera RL, Glahn D et al. Fronto-limbic 216. Volkow ND, Wang GJ, Telang F et al. Cocaine cues and
brain abnormalities in juvenile onset bipolar disorder. dopamine in dorsal striatum: mechanism of craving in
Biol Psychiatry 2005;58(7):525–31. cocaine addiction. J Neurosci. 2006;26(24):6583–8.
203. Haldane M, Frangou S. New insights help deine the 217. Yin HH, Knowlton BJ. he role of the basal ganglia in
pathophysiology of bipolar afective disorder: habit formation. Nat Rev Neurosci. 2006;7(6):464–76.
neuroimaging and neuropathology indings. Prog 218. Belin D, Everitt BJ. Cocaine seeking habits depend
Neuropsychopharmacol Biol Psychiatry 2004; upon dopamine-dependent serial connectivity linking
28(6):943–60. the ventral with the dorsal striatum. Neuron 2008;
204. Swerdlow NR, Koob GF. Toward a uniied hypothesis 57(3):432–41.
of cortico-striato-pallido-thalamus function. Behav 219. Koob GF, Nestler EJ. he neurobiology of drug
Brain Sci. 1990;13(1):172–177. addiction. J Neuropsychiatry Clin Neurosci.
205. Braf DL, Geyer MA. Sensorimotor gating and 1997;9(3):482–97.
schizophrenia. Human and animal model studies. Arch 220. Gardner E. Brain reward mechanisms. In Lowinson
Gen Psychiatry 1990;47(2):181–8. JH, editor. Substance Abuse: A Comprehensive
206. Jernigan TL, Zisook S, Heaton RK et al. Magnetic Textbook. 3rd edition. Baltimore, MD: Williams &
resonance imaging abnormalities in lenticular nuclei Wilkins; 1997, pp. 51–85.

83
Downloaded from https://www.cambridge.org/core. Stockholm University Library, on 25 Nov 2018 at 16:04:24, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781139016919.007
Section I: Structural and Functional Neuroanatomy

221. Ivanov IS, Schulz KP, Palmero RC, Newcorn JH. 234. Taylor AE, Saint-Cyr JA, Lang AE. Frontal lobe
Neurobiology and evidence-based biological dysfunction in Parkinson’s disease. he cortical focus
treatments for substance abuse disorders. CNS Spectr. of neostriatal outlow. Brain 1986;109(Pt 5):845–83.
2006;11(11):864–77. 235. Scatton B, Javoy-Agid F, Rouquier L, Dubois B, Agid Y.
222. Weintraub D. Dopamine and impulse control Reduction of cortical dopamine, noradrenaline,
disorders in Parkinson’s disease. Ann Neurol. 2008; serotonin and their metabolites in Parkinson’s disease.
64(Suppl. 2):S93–100. Brain Res. 1983;275(2):321–8.
223. Joyce JN, Gurevich EV. D3 receptors and the actions of 236. Portin R, Rinne UK. Predictive factors for cognitive
neuroleptics in the ventral striatopallidal system of deterioration and dementia in Parkinson’s disease. Adv
schizophrenics. Ann N Y Acad Sci. 1999;877:595–613. Neurol. 1987;45:413–16.
224. Murray AM, Ryoo HL, Gurevich E, Joyce JN. 237. Marsh L, Biglan K, Gerstenhaber M, Williams JR.
Localization of dopamine D3 receptors to mesolimbic Atomoxetine for the treatment of executive
and D2 receptors to mesostriatal regions of human dysfunction in Parkinson’s disease: a pilot open-label
forebrain. Proc Natl Acad Sci USA 1994;91(23): study. Mov Disord. 2009;24(2):277–82.
11 271–5. 238. Arnsten AF, Contant TA. Alpha-2 adrenergic agonists
225. Chen J, Marmur R, Paredes W, Pulles A, Gardner EL. decrease distractibility in aged monkeys performing
Systemic cocaine challenge ater chronic cocaine the delayed response task. Psychopharmacology (Berl.)
treatment reveals sensitization of extracellular 1992;108(1–2):159–69.
dopamine levels in nucleus accumbens but direct 239. Goldman-Rakic PS, Lidow MS, Gallager DW. Overlap
cocaine perfusion into nucleus accumbens does not: of dopaminergic, adrenergic, and serotoninergic
implications for the neural locus of cocaine receptors and complementarity of their subtypes in
sensitization. Life Sci. 1996;58(8):PL139–46. primate prefrontal cortex. J. Neurosci. 1990;10(7):
226. Cilia R, Siri C, Marotta G et al. Functional 2125–38.
abnormalities underlying pathological gambling in 240. Arnsten AF, Cai JX, Goldman-Rakic PS. he alpha-2
Parkinson disease. Arch Neurol. 2008;65(12):1604–11. adrenergic agonist guanfacine improves memory in
227. Blanco C, Moreyra P, Nunes EV, Saiz-Ruiz J, Ibanez A. aged monkeys without sedative or hypotensive side
Pathological gambling: addiction or compulsion? efects: evidence for alpha-2 receptor subtypes.
Semin Clin Neuropsychiatry 2001;6(3):167–76. J Neurosci. 1988;8(11):4287–98.
228. Potenza MN. Should addictive disorders include 241. Arnsten AF, Goldman-Rakic PS. Alpha 2-adrenergic
non-substance-related conditions? Addiction mechanisms in prefrontal cortex associated with
2006;101(Suppl. 1):142–51. cognitive decline in aged nonhuman primates. Science
229. Hollander E. Behavioral and substance addictions: A 1985;230(4731):1273–6.
new proposed DSM-V category characterized by 242. Fields RB, Van Kammen DP, Peters JL et al. Clonidine
impulsive choice, reward sensitivity, and fronto-striatal improves memory function in schizophrenia
circuit impairment. CNS Spectr. 2006;11(11):814. independently from change in psychosis. Preliminary
230. Potenza MN. he neurobiology of pathological indings. Schizophr Res. 1988;1(6):417–23.
gambling and drug addiction: an overview and new 243. Mair RG, McEntee WJ. Cognitive enhancement in
indings. Philos R Soc B. 2008;363(1507):3181–9. Korsakof’s psychosis by clonidine: a comparison with
231. Smith KS, Berridge KC. Opioid limbic circuit for L-dopa and ephedrine. Psychopharmacology (Berl.)
reward: interaction between hedonic hotspots of 1986;88(3):374–80.
nucleus accumbens and ventral pallidum. J Neurosci. 244. Mofoot A, O’Carroll RE, Murray C et al. Clonidine
2007;27(7):1594–605. infusion increases uptake of 99mTc-Exametazime in
232. Lange KW, Robbins TW, Marsden CD et al. L-dopa anterior cingulate cortex in Korsakof’s psychosis.
withdrawal in Parkinson’s disease selectively impairs Psychol Med. 1994;24(1):53–61.
cognitive performance in tests sensitive to frontal lobe 245. Sahakian BJ, Coull JJ, Hodges JR. Selective
dysfunction. Psychopharmacology (Berl.) 1992; enhancement of executive function by idazoxan in a
107(2–3):394–404. patient with dementia of the frontal lobe type. J Neurol
233. Cooper JA, Sagar HJ, Doherty SM et al. Diferent Neurosurg Psychiatry 1994;57(1):120–1.
efects of dopaminergic and anticholinergic therapies 246. Denckla MB, Reader MJ. Education and psychosocial
on cognitive and motor function in Parkinson’s interdentions: executive dysfunction and its
disease. A follow-up study of untreated patients. Brain consequences. In Kurlan R, editor. Handbook of
1992;115(Pt 6):1701–25. Tourette’s Syndrome and Related Tic and Behavioral

84
Downloaded from https://www.cambridge.org/core. Stockholm University Library, on 25 Nov 2018 at 16:04:24, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781139016919.007
Chapter 5: Frontal-subcortical circuits

Disorders. New York, NY: M. Dekker; 1993, 260. Alexander MP. Reversal of chronic akinetic mutism
pp. 431–51. ater mesencephalic infarction with dopaminergic
247. Golden GS. Treatment of attention deicit hyperactivity agents. Neurology 1995;45(4):A330.
disorder. In Kurlan R, editor. Handbook of Tourette’s 261. Spiegel DR, Casella DP, Callender DM, Dhadwal N.
Syndrome and Related Tic and Behavioral Disorders. Treatment of akinetic mutism with intramuscular
New York, NY: M. Dekker; 1993, pp. 423–30. olanzapine: a case series. J Neuropsychiatry Clin
248. Leckman JF, Hardin MT, Riddle MA et al. Clonidine Neurosci. 2008;20(1):93–5.
treatment of Gilles de la Tourette’s syndrome. Arch 262. Parks RW, Crockett DJ, Manji HK, Ammann W.
Gen Psychiatry 1991;48(4):324–8. Assessment of bromocriptine intervention for the
249. Tannock R, Schachar RJ, Carr RP, Chajczyk D, Logan treatment of frontal lobe syndrome: a case study.
GD. Efects of methylphenidate on inhibitory control J Neuropsychiatry Clin Neurosci. 1992;4(1):109–11.
in hyperactive children. J Abnorm Child Psychol. 263. Barrett K. Treating organic abulia with bromocriptine
1989;17(5):473–91. and lisuride: four case studies. J Neurol Neurosurg
250. Hunt RD, Arnsten AF, Asbell MD. An open trial of Psychiatry 1991;54(8):718–21.
guanfacine in the treatment of attention-deicit 264. Holmes VF, Fernandez F, Levy JK. Psychostimulant
hyperactivity disorder. J Am Acad Child Adolesc response in AIDS-related complex patients. J Clin
Psychiatry 1995;34(1):50–4. Psychiatry 1989;50(1):5–8.
251. Arnsten AF, Steere JC, Hunt RD. he contribution of 265. Czernecki V, Schupbach M, Yaici S et al. Apathy
alpha 2-noradrenergic mechanisms of prefrontal following subthalamic stimulation in Parkinson
cortical cognitive function. Potential signiicance for disease: a dopamine responsive symptom. Mov Disord.
attention-deicit hyperactivity disorder. Arch Gen 2008;23(7):964–9.
Psychiatry 1996;53(5):448–55. 266. Marin RS, Fogel BS, Hawkins J, Dufy J, Krupp B.
252. Ungerstedt U. Is interruption of the nigro-striatal Apathy: a treatable syndrome. J Neuropsychiatry Clin
dopamine system producing the “lateral hypothalamus Neurosci. 1995;7(1):23–30.
syndrome”? Acta Physiol Scand. 1970;80(4): 267. Craig AH, Cummings JL, Fairbanks L et al. Cerebral
35A–6A. blood low correlates of apathy in Alzheimer disease.
253. Ungerstedt U. Adipsia and aphagia ater Arch Neurol. 1996;53(11):1116–20.
6-hydroxydopamine induced degeneration of the 268. Kaufer DI, Cummings JL, Christine D. Efect of tacrine
nigro-striatal dopamine system. Acta Physiol Scand on behavioral symptoms in Alzheimer’s disease: an
Suppl. 1971;367:95–122. open-label study. J Geriatr Psychiatry Neurol.
254. Marshall JF, Richardson JS, Teitelbaum P. Nigrostriatal 1996;9(1):1–6.
bundle damage and the lateral hypothalamic 269. Masanic CA, Bayley MT, VanReekum R, Simard M.
syndrome. J Comp Physiol Psychol. 1974;87(5):808–30. Open-label study of donepezil in traumatic brain
255. Ljungberg T, Ungerstedt U. Reinstatement of eating by injury. Arch Phys Med Rehabil. 2001;82(7):896–901.
dopamine agonists in aphagic dopamine denervated 270. Morey CE, Cilo M, Berry J, Cusick C. he efect of
rats. Physiol Behav. 1976;16(3):277–83. Aricept in persons with persistent memory disorder
256. Marshall JF, Ungerstedt U. Apomorphine-induced following traumatic brain injury: a pilot study. Brain
restoration of drinking to thirst challenges in Inj. 2003;17(9):809–15.
6-hydroxydopamine-treated rats. Physiol Behav. 271. Cummings J. Behavioral disorders associated with
1976;17(5):817–22. frontal lobe injury. In Cummings JL, editor. Clinical
257. Marshall JF, Gotthelf T. Sensory inattention in rats Neuropsychiatry. Orlando, FL: Grune & Stratton; 1985,
with 6-hydroxydopamine-induced degeneration of pp. 57–67.
ascending dopaminergic neurons: apomorphine- 272. Lesch KP, Merschdorf U. Impulsivity, aggression, and
induced reversal of deicits. Exp Neurol. serotonin: a molecular psychobiological perspective.
1979;65(2):398–411. Behav Sci Law 2000;18(5):581–604.
258. Ross ED, Stewart RM. Akinetic mutism from 273. Brown GL, Linnoila MI. CSF serotonin metabolite
hypothalamic damage: successful treatment with (5-HIAA) studies in depression, impulsivity, and
dopamine agonists. Neurology 1981;31(11):1435–9. violence. J Clin. Psychiatry 1990;51(Suppl.):31–41;
259. Nemeth G, Hegedus K, Molnar L. Akinetic mutism discussion 2–3.
and locked-in syndrome: the functional-anatomical 274. Coccaro EF. Central serotonin and impulsive
basis for their diferentiation. Funct Neurol. aggression. Br J Psychiatry Suppl. 1989;8:52–62.
1986;1(2):128–39.

85
Downloaded from https://www.cambridge.org/core. Stockholm University Library, on 25 Nov 2018 at 16:04:24, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781139016919.007
Section I: Structural and Functional Neuroanatomy

275. Stein DJ, Hollander E, Liebowitz MR. Neurobiology of and pathophysiology. Biol Psychiatry
impulsivity and the impulse control disorders. 1987;22(6):667–87.
J Neuropsychiatry Clin Neurosci. 1993;5(1):9–17. 291. Goodman WK, Price LH, Delgado PL et al. Speciicity
276. Dougherty DM, Moeller FG, Bjork JM, Marsh DM. of serotonin reuptake inhibitors in the treatment of
Plasma L-tryptophan depletion and aggression. Adv obsessive-compulsive disorder. Comparison of
Exp Med Biol. 1999;467:57–65. luvoxamine and desipramine. Arch Gen Psychiatry
277. Leo RJ, Kim KY. Clomipramine treatment of 1990;47(6):577–85.
paraphilias in elderly demented patients. J Geriatr 292. Greybiel AM, Ragsdale CW. Biochemical anatomy of
Psychiatry Neurol. 1995;8(2):123–4. the striatum. In Emson PC, editor. Chemical
278. Hollander E, Wong CM. Body dysmorphic disorder, Neuroanatomy. New York, NY: Raven Press; 1983,
pathological gambling, and sexual compulsions. J Clin pp. 427–507.
Psychiatry 1995;56(Suppl. 4):7–12; discussion 3. 293. Nieuwenhuys R. Chemoarchitecture of the Brain.
279. Olivier B, Mos J. Serenics and aggression. Stress Berlin: Springer-Verlag; 1985.
Medicine 1986;2(3):197–209. 294. McDougle CJ, Goodman WK, Price LH. Dopamine
280. Pazos A, Palacios JM. Quantitative autoradiographic antagonists in tic-related and psychotic spectrum
mapping of serotonin receptors in the rat brain. I. obsessive compulsive disorder. J Clin Psychiatry
Serotonin-1 receptors. Brain Res. 1985;346(2):205–30. 1994;55(Suppl.):24–31.
281. Hjorth S, Carlsson A. Is pindolol a mixed 295. Korsgaard S, Gerlach J, Christensson E. Behavioral
agonist-antagonist at central serotonin (5-HT) aspects of serotonin-dopamine interaction in the
receptors? Eur J Pharmacol. 1986;129(1–2):131–8. monkey. Eur J Pharmacol. 1985;118(3):245–52.
282. Bel N, Romero L, Celada P et al. editors. 296. Ballantine HT, Jr., Bouckoms AJ, homas EK, Giriunas
Neurobiological basis for the potentiation of the IE. Treatment of psychiatric illness by stereotactic
antidepressant efect of 5-HT reuptake inhibitors by cingulotomy. Biol Psychiatry 1987;22(7):807–19.
the 5-HT1A antagonist pindolol. In Monitoring 297. Sawle GV, Lees AJ, Hymas NF, Brooks DJ, Frackowiak
Molecules in Neuroscience: Proceeding of the 6th RS. he metabolic efects of limbic leucotomy in Gilles
International Conference of in vivo Methods; 1994. de la Tourette syndrome. J Neurol Neurosurg Psychiatry
Bordeaux: University of Bordeaux. 1993;56(9):1016–19.
283. Bunney WE, Garland-Bunney BL. Mechanisms of 298. Robertson M, Doran M, Trimble M, Lees AJ. he
action of lithium in afective illness: basic and clinical treatment of Gilles de la Tourette syndrome by limbic
implications. In: Meltzer HY, editor. leucotomy. J Neurol Neurosurg Psychiatry 1990;
Psychopharmacology: he hird Generation of Progress. 53(8):691–4.
New York, NY: Raven Press; 1987, pp. 553–65. 299. Kurlan R, Kersun J, Ballantine HT, Jr.,
284. Snyder SH. Second messengers and afective illness. Caine ED. Neurosurgical treatment of severe
Focus on the phosphoinositide cycle. obsessive-compulsive disorder associated with
Pharmacopsychiatry 1992;25(1):25–8. Tourette’s syndrome. Mov Disord. 1990;5(2):
285. Jouvent R, Lecrubier Y, Puech AJ, Simon P, Widlocher 152–5.
D. Antimanic efect of clonidine. Am J Psychiatry 300. Papez JW. A proposed mechanism of emotion. Arch
1980;137(10):1275–6. Neurol Psychiatry 1937;38(4):725–43.
286. Shen WW. Mania, clonidine, and dopamine. Am J 301. Mesulam M. Patterns in behavioral neuoranatomy:
Psychiatry 1986;143(1):127. association areas, the limbic system, and hemispheric
287. Arnsten AF, Goldman-Rakic PS. Selective prefrontal specialization. In Mesulam MM, editor. Principles of
cortical projections to the region of the locus coeruleus Behavioral Neurology. Philadelphia. PA: F.A. Davis;
and raphe nuclei in the rhesus monkey. Brain Res. 1985, pp. 1–70.
1984;306(1–2):9–18. 302. Bingley T, Leksell L, Meyerson BA, Rylander G.
288. Cummings JL. Organic psychoses. Delusional Long-term results of stereotactic anterior capsulotomy
disorders and secondary mania. Psychiatr Clin North in chronic obsessive-compulsive neurosis. In Sweet
Am. 1986;9(2):293–311. WH, Obrador S, Martı́n-Rodrı́guez JG, editors.
Neurosurgical Treatment in Psychiatry, Pain, and
289. Jankovic J. Deprenyl in attention deicit associated with Epilepsy. Proceedings of the Fourth World Congress of
Tourette’s syndrome. Arch Neurol. 1993;50(3):286–8. Psychiatric Surgery, September 7–10, 1975, Madrid,
290. Zohar J, Insel TR. Obsessive-compulsive disorder: Spain. Baltimore, MD: University Park Press; 1977,
psychobiological approaches to diagnosis, treatment, pp. 287–99.

86
Downloaded from https://www.cambridge.org/core. Stockholm University Library, on 25 Nov 2018 at 16:04:24, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781139016919.007
Chapter 5: Frontal-subcortical circuits

303. Lujan JL, Chaturvedi A, McIntyre CC. Tracking the 312. Flaherty AW, Williams ZM, Amirnovin R et al. Deep
mechanisms of deep brain stimulation for brain stimulation of the anterior internal capsule for
neuropsychiatric disorders. Front Biosci. 2008; the treatment of Tourette syndrome: technical case
13:5892–904. report. Neurosurgery 2005;57(4 Suppl.):E403;
304. Greenberg BD, Malone DA, Friehs GM et al. discussion E.
hree-year outcomes in deep brain stimulation for 313. Servello D, Porta M, Sassi M, Brambilla A, Robertson
highly resistant obsessive-compulsive disorder. MM. Deep brain stimulation in 18 patients with severe
Neuropsychopharmacology 2006;31(11):2384–93. Gilles de la Tourette syndrome refractory to treatment:
305. Rauch SL, Dougherty DD, Malone D et al. A the surgery and stimulation. J Neurol Neurosurg
functional neuroimaging investigation of deep brain Psychiatry 2008;79(2):136–42.
stimulation in patients with obsessive-compulsive 314. Bajwa RJ, de Lotbiniere AJ, King RA et al. Deep brain
disorder. J Neurosurg. 2006;104(4):558–65. stimulation in Tourette’s syndrome. Mov Disord.
306. Nuttin BJ, Gabriels LA, Cosyns PR et al. Long-term 2007;22(9):1346–50.
electrical capsular stimulation in patients with 315. Maciunas RJ, Maddux BN, Riley DE et al. Prospective
obsessive-compulsive disorder. Neurosurgery randomized double-blind trial of bilateral thalamic
2003;52(6):1263–72; discussion 72–4. deep brain stimulation in adults with Tourette
307. Ackermans L, Temel Y, Cath D et al. Deep brain syndrome. J Neurosurg. 2007;107(5):1004–14.
stimulation in Tourette’s syndrome: two targets? Mov 316. Visser-Vandewalle V. DBS in Tourette syndrome:
Disord. 2006;21(5):709–13. rationale, current status and future prospects. Acta
308. Shahed J, Poysky J, Kenney C, Simpson R, Jankovic J. Neurochir Suppl. 2007;97(Pt 2):215–22.
GPi deep brain stimulation for Tourette syndrome 317. Temel Y, Visser-Vandewalle V. Surgery in Tourette
improves tics and psychiatric comorbidities. Neurology syndrome. Mov Disord. 2004;19(1):3–14.
2007;68(2):159–60. 318. Hamani C, Saint-Cyr JA, Fraser J, Kaplitt M,
309. Welter ML, Mallet L, Houeto JL et al. Internal pallidal Lozano AM. he subthalamic nucleus in the
and thalamic stimulation in patients with Tourette context of movement disorders. Brain 2004;127(Pt 1):
syndrome. Arch Neurol. 2008;65(7):952–7. 4–20.
310. Dehning S, Mehrkens JH, Muller N, Botzel K. 319. Hauptman JS, DeSalles AA, Espinoza R, Sedrak M,
herapy-refractory Tourette syndrome: beneicial Ishida W. Potential surgical targets for deep brain
outcome with globus pallidus internus deep brain stimulation in treatment-resistant depression.
stimulation. Mov Disord. 2008;23(9):1300–2. Neurosurg Focus 2008;25(1):E3.
311. Houeto JL, Karachi C, Mallet L et al. Tourette’s 320. Stelten BM, Noblesse LH, Ackermans L, Temel Y,
syndrome and deep brain stimulation. J Neurol Visser-Vandewalle V. he neurosurgical treatment of
Neurosurg Psychiatry 2005;76(7):992–5. addiction. Neurosurg Focus 2008;25(1):E5.

87
Downloaded from https://www.cambridge.org/core. Stockholm University Library, on 25 Nov 2018 at 16:04:24, subject to the Cambridge Core terms of use, available at
https://www.cambridge.org/core/terms. https://doi.org/10.1017/CBO9781139016919.007

You might also like