You are on page 1of 14

REVIEwS

Neocortical circuits in pain and


pain relief
Linette Liqi Tan ✉ and Rohini Kuner    ✉
Abstract | The sensory, associative and limbic neocortical structures play a critical role in shaping
incoming noxious inputs to generate variable pain perceptions. Technological advances in tracing
circuitry and interrogation of pathways and complex behaviours are now yielding critical knowledge
of neocortical circuits, cellular contributions and causal relationships between pain perception and
its abnormalities in chronic pain. Emerging insights into neocortical pain processing suggest the
existence of neocortical causality and specificity for pain at the level of subdomains, circuits and
cellular entities and the activity patterns they encode. These mechanisms provide opportunities
for therapeutic intervention for improved pain management.

Chronic pain is often poorly managed by existing treat- inputs are only poorly related to the resulting unpleas-
ments and is a major health care problem worldwide ant quality of pain sensation and that the experience
with substantial clinical, social and economic impact. of pain is markedly modulated by cognitive and moti-
A major challenge in understanding pain is due to vational factors as well as emotional states2,3,9, thereby
its multidimensionality: pain is a unique experience highlighting the importance of neocortical contribu-
involving sensory, emotional, motivational and affective tions. This explains major individual-​dependent and
components. context-​dependent variations in pain sensitivity, and also
The overall experience of pain is the result of col- provides a basis for pain control. In support, converging
lective activity arising from numerous areas across the lines of evidence point to the neocortex as a central regu­
brain in conjunction with ascending and descending lator of diverse cortical and subcortical regions that are
pathways1–3. However, given the wide nature of brain involved in pain as well as its control by pharmacological
areas activated during pain, as well as the logic that acti- and cognitive modulation therapies3,8,9. Recent studies
vation does not necessarily indicate a causal contribu- have intensified the quest to understand how specificity
tion, disentangling the identity of regions and pathways and functional integration of nociceptive information
that primarily mediate acute and chronic pain from are encoded and altered at the level of cells and circuits
those that are secondarily recruited as a consequence within neocortical networks during pain chronicity.
of pain or the resulting motor or emotional conse- These studies have also addressed cause–effect relation-
quences is a challenge4. This caveat is beginning to be ships in animal models, which form the main focus of
successfully overcome with the aid of animal models, this Review.
propelled largely by advances in a rapidly expanding At the outset, it is important to acknowledge and
array of technologies, such as optogenetic and chemoge- highlight a difficult conundrum in this field. Neocortical
netic activation or silencing of neurons and their axonal domains are not only recruited during pain perception
projections, dynamic in vivo analysis of cellular activ- but also continuously drive a plethora of brain functions,
ity using single-​photon or multiphoton calcium imag- including cognition, decision-​making, memory, emotion
ing, and electrophysiological analyses of the activity of and homeostatic control. A clear outcome of a large body
hundreds of neurons at the network level as well as the of evidence is that there is no single neocortical domain
single-​cell level5. that is ‘specific’ for pain, unlike its counterparts in the
Several recent reviews have comprehensively dis- peripheral nervous system, spinal superficial dorsal horn
Institute of Pharmacology, cussed progress made in our understanding of spinal and brainstem, where ‘nociception-​specific’ functional
Heidelberg University, and subcortical brain circuits and networks5–8. However, entities have been identified2,3,8,9. However, to the suf-
Heidelberg, Germany. the neocortex and its modulation of key subcortical ferer, pain constitutes a unique percept and experience
✉e-​mail: linette.tan@
domains has remained one of the final frontiers in pain that cannot be confounded with another sensation or
pharma.uni-​heidelberg.de;
research, perhaps owing to its tremendous complexity emotion, and there is unequivocal evidence that the neo-
rohini.kuner@
pharma.uni-​heidelberg.de and plasticity. Although mapping sensory processing cortex plays a critical role in this experience. How spec-
https://doi.org/10.1038/ has traditionally dominated our understanding of pain ificity for pain perception is created, therefore, remains
s41583-021-00468-2 mechanisms, it is now well established that sensory one of the grand challenges in biology and medicine.

458 | August 2021 | volume 22 www.nature.com/nrn

0123456789();:
Reviews

Box 1 | Functional specificity in pain Prefrontal cortex


In humans and non-​human primates, the PFC includes
In terms of regionally specific contributions to pain, recent studies both support and the medial PFC (mPFC), ventrolateral PFC, dorsolat-
challenge the notion of pain specificity in designated cortical domains, such as the eral PFC (dlPFC), ACC and orbitofrontal cortex (OFC),
dorsal anterior cingulate cortex and the insular cortex158–161. New evidence has also encompassing the anatomically defined Brodmann areas
challenged the dogma of broad segregation of cortical areas into purely sensory
8–14, 24, 25, 32 and 44–47 (ref.12). Anatomical and phys-
discriminative versus emotional, and suggests the existence of functional subdivisions
within each of the cortices. At the next level of resolution, functional specificity may iological comparisons suggest that the rodent mPFC can
be imparted by distinct pathways and circuits. In support of this, recent connectivity be subdivided into the ACC, prelimbic (PL) cortex and
analyses and optogenetic studies demonstrate that selective corticofugal pathways, infralimbic (IL) cortex, which are homologues compa-
such as prefrontal projections to the nucleus accumbens or posterior insula projections rable to areas 24, 32 and 25, respectively, of primates.
to the rostroventral medulla enhance pain, while others, such as prefrontal cortical However, thus far, difficulties in reconciling data from
projections to the periaqueductal grey, attenuate pain29,42,44,99. At the next level of human, primate and rodent PFC pain studies have risen
fine-​grained cellular maps, specificity of information coding may be conferred by specific mainly due to the use of ambiguous or hybrid nomen-
neocortical neuronal ensembles (that is, populations of neurons with coordinated clatures (for example, ‘prelimbic mPFC’ and ‘ventral
activity that computationally encode specific functions)162. Specific subsets of neurons mPFC’) and the use of different brain schemes and
that co-​fire in response to noxious stimuli, such as those recently described in the
atlases13, leading to confusion among researchers regard-
amygdala117, may selectively encode pain perception, and their fluctuations upon
intrinsic and extrinsic modulatory influences may account for abnormal variations in ing the actual PFC areas being investigated (see Table 1).
pain perception in chronic pain states. Finally, looping back to the notion that pain is Here we attempt to describe only recent studies where
a network-​driven process, oscillatory brain rhythms, such as alpha, theta and gamma detailed regional information is available so as to get a
frequency rhythms163,164, can provide a mechanistic basis for distinguishing noxious clearer picture of the roles and circuitries of the specific
from innocuous intensity stimulation and explain abnormal fluctuations of neural PFC subdomains in pain.
activities in chronic pain states. Moreover, by serving as a mechanism for short-​range
and long-​range communication between distributed neuronal ensembles and circuits, The PFC in pain. Neuroimaging studies often implicate
they may enable temporal coordination of information across distant, spatially separated PFC activation in response to acute nociceptive stim-
networks and thus help functionally integrate sensory and emotional–motivational uli (lasting milliseconds to seconds) in healthy people9.
aspects into a comprehensive pain percept.
Under longer-​lasting pain conditions, a prominent
role for altered PFC functional connectivity and differ-
In this Review, we propose that functional specificity ential activity in pain modulation have also been well
for pain outcomes in neocortical networks and their cor- documented9,14,15. Here, analysis of neural oscillatory
ticofugal projection targets may come about at least at four responses to tonic experimental noxious stimuli (lasting
distinct, but inter-​related, levels: (1) dynamic activity states minutes) frequently reveals enhanced power of gamma
in neocortical networks, (2) regional, functionally distinct oscillations and suppression of alpha and beta oscilla-
subdomains, (3) specific circuit connections that deline- tions in prefrontal areas that are closely associated with
ate pain from other functions, and (4) cellular ensembles stimulus and pain intensities16–18. Similarly, increases in
of co-​active, functionally homogenous populations of the resting-​state gamma activity and power in the PFC
neurons (see Box 1). In support, we highlight evidence have also been observed during chronic pain conditions,
from recent animal and human work in the context of key reflecting a shift towards an emphasis and recruitment
pain-​related neocortical areas, namely, the prefrontal cor- of emotional–motivational processes in the PFC during
tex (PFC), midcingulate cortex (MCC), anterior cingulate the encoding of chronic ongoing pain17,19–22.
cortex (ACC), anterior insula (AI), posterior insula (PI), In animal models, neuronal oscillations in the
primary somatosensory cortex (S1) and secondary soma- PFC in pain states have not been well studied so far.
tosensory cortex (S2). Several of these regions are also Furthermore, depending on the areas of the PFC under
implicated in anxiodepressive disorders that frequently investigation, conclusions regarding the state of ‘PFC’
accompany chronic pain as co-​morbidities (Box 2). or ‘mPFC’ activity in pathological pain can differ sig-
nificantly, and different maladaptive changes in specific
Functional contributions of distinct cortical PFC domains are reported in different types of chronic
domains in pain pain. In the following sections, we discuss the roles
The PFC, ACC, insula, S1 and S2 neocortices are part of specific PFC subdomains and summarize key PFC
of the ‘neurologic pain signature’, a defined brain activity pathways linked to alterations in pain.
pattern that is derived from a multivariate pattern analysis
of functional MRI data combined with machine learning, Regional and laminar contributions. Although neu-
Multivariate pattern which can be used to predict acute pain associated with rons responsive to both innocuous and noxious stimuli
analysis activation of nociceptors by noxious stimuli2. Moreover, have been recorded in both the PL cortex and the IL
A set of tools that can analyse a similar approach was applied on large imaging data cortex of healthy rodents23,24, structural and functional
and identify neural responses
sets to predict pain beyond nociceptive input to yield data support a more prominent role for the PL cortex
in the brain as spatially
distributed patterns of activity. stimulus intensity-​independent pain signature 1, which in pain processing (Fig. 1). For instance, bilateral lesions
includes the lateral PFC as a neocortical component of the PL cortex, but not the IL cortex, prevented the
Machine learning in addition to the parahippocampal cortices and the development of inflammatory-​induced hyperalgesia and
The use of computer nucleus accumbens (NAc)10. Similarly, rodent imaging pain aversion25,26. Similarly, altered intrinsic excitability
algorithms to learn, analyse,
identify and predict patterns
studies also reveal the S1 and the insular and cingu­ of PL neurons, but not IL neurons, is observed after
from data sets with minimal late cortices to be the neocortical domains that are nerve injury27. Furthermore, using markers of neuronal
human supervision. consistently activated during acute pain11. activation and sensitization (for example, CREB/p38

NAture RevIews | NeurOSCienCe volume 22 | August 2021 | 459

0123456789();:
Reviews

phosphorylation and FOS), others have also reported in neuropathic pain models. Increases in noradren-
enhanced PL cortex activation during acute and persis- ergic innervation38 as well as dendritic complexity
tent pain states28,29. Although the rodent PL cortex is the and the lengths of these layer 2/3 neurons have been
most studied PFC region in pain, its role remains highly described30,37, suggesting a reorganization of intracortical
unclear given that both activation and deactivation have circuits contributing to neuropathic pain. Such morpho-
been reported in diverse models and studies in the con- logical changes are also accompanied by dysregulated
text of pain. While PL cortex activation is suggested to NMDA receptor-​mediated currents, α2-​adrenoceptor
be required for inflammatory pain25,26, the predominant activity and impaired hyperpolarization-​a ctivated
view points to a diminished activity of the mPFC during cyclic nucleotide-​gated (HCN) channels, all of which
chronic neuropathic pain27,30–32. have been correlated with nociceptive hypersensitivity
Differential dysregulation of the various PL cortical in vivo38,39 (Fig. 1).
layers is reported in chronic pain models. For example, Thus far, altered IL cortical activity has been detec­
after a peripheral nerve injury, structural losses in layer 5 ted during inflammatory pain, where it has been linked
PL pyramidal neurons32 and reduced local excitatory to an impaired interaction between presynaptic CB1-​
inputs27 likely contribute to the decreased excitability of type cannabinoid receptors and postsynaptic mGluR5
layer 5 pyramidal neurons that is often seen in chronic receptors 24. Under neuropathic conditions, the IL
pain models27,30–32 (Fig. 1). Such reduced excitability has cortex does not appear to contribute to chronic pain
also been suggested to be partly mediated by enhanced signalling27,30. Rather, chronic pain-​related cognitive
feedforward inhibition from fast-​spiking parvalbu- impairments have recently been linked to morpholog-
min (PV)-​type interneurons31, but not somatostatin ical changes of PV-​type interneurons in layers 5 and 6
(SOM)-​containing GABAergic interneurons27, which are of the IL cortex40.
driven by increased glutamatergic inputs from the baso-
lateral amygdala (BLA) to these PV-​type interneurons PFC–NAc pathway. The PFC projects to the NAc, one of
after nerve injury31,33. Activation of mGluR1-​type glu- the major nodes in the reward circuitry, and in human
tamate receptors (presumably from BLA afferents), but imaging studies, an initial enhanced mPFC–NAc func-
not mGluR5-​type receptors, presynaptic to GABAergic tional connectivity correlated with and predicted the
interneurons is critically involved in driving this mPFC persistence of pain in patients with chronic back pain15,41,
inhibition24. Interestingly, it was recently demonstrated suggesting a facilitatory role for prefrontal–striatal con-
such nerve injury-​induced increases in PV-​type neuronal nectivity in pain. However, in animal models of acute
excitability occur only in male mice34, suggesting that and chronic pain, selective optogenetic activation of the
sex-​specific GABAergic mechanisms underlie chronic PL output to the core region of the NAc significantly
pain. Furthermore, impairments in endocannabinoid, inhibited pain-​related behaviours42,43, while optogenetic
cholinergic, catecholaminergic, opioidergic and dopamin- suppression of the same pathway enhanced nociceptive
ergic signalling also contribute to the altered prefrontal sensitivity in both uninjured and neuropathic animals44
excitability reported during sustained pain5,35,36. (Fig. 1). Thus, additional translational studies are needed
Excitability of layer 2/3 PL neurons is significantly to resolve the role of PFC–NAc connectivity in pain. The
attenuated during inflammatory pain25 but has also IL cortex–NAc projection has not been examined in
been reported to be unaffected37 or even increased30,38 the context of pain.

Box 2 | Neural circuits of pain co-​morbidities PFC–PAG pathway. PFC projections to the periaque-
ductal grey (PAG), a key node within the descending
Chronic pain is a strong trigger of co-​morbid disorders of negative emotions such
pain modulation network, have been extensively char-
as persistent stress, depression, anxiety, addiction vulnerability and memory deficits.
In turn, these affective conditions can aggravate pain duration and sensitivity,
acterized in several mammalian species12. These studies
making it a relentless cycle that is a challenge to treat effectively (see reviews118,165,166). are further supported by functional MRI and diffusion
Unsurprisingly, brain regions such as the prefrontal cortex, anterior cingulate cortex, tensor imaging studies in humans, confirming the exist-
amygdala, insula and nucleus accumbens that are commonly implicated in affective ence of a PFC–PAG circuitry that underlies top-​down
and cognitive disorders are also reported to be involved in regulation of negative affect influences on spinal nociceptive processing9,45. The PAG
in patients with pain. projects to downstream targets that include serotonin-
Specific neural circuits underpinning core co-​morbid traits of chronic pain are only ergic neurons in the raphe nuclei of the rostroventral
just beginning to be explored in animal models. These studies have revealed novel medulla (RVM), which project to the dorsal horn of
functional contributions of various subcortical circuits involving the dorsal raphe the spinal cord, where peripheral noxious inputs are
nucleus, lateral habenula, amygdala and parabrachial nucleus in the regulation of
received and processed.
negative emotions such as anxiety and depression induced by chronic pain134,167,168.
In terms of neocortical contributions, co-​morbid anxiodepressive behaviours in animal
Within the rodent mPFC, a major subpopulation of
models of chronic pain were reported to be driven by persistent hyperactivity of the layer 5 neurons in both the PL region and the IL region
anterior cingulate cortex72, where lesions of the anterior cingulate cortex, but not projects to the PAG27,46. In mouse models of chronic con-
the posterior insula, sufficiently prevented the development of anxiodepressive striction nerve injury, the intrinsic excitability of PL–PAG
behaviours in chronic pain models71. More recently, in a persistent inflammatory pain neurons, but not of IL–PAG neurons, is markedly atten-
model, enhanced activity of the excitatory projections from the primary somatosensory uated bilaterally27, suggesting that reduced activity of
cortex to GABAergic neurons residing in the caudal dorsolateral striatum was also the PL–PAG pathway contributes to the persistence
found to modulate co-​morbid anxiety97, highlighting that the primary somatosensory of neuropathic pain (Fig. 1). Recently, with use of trans-
cortex, in addition to its predominant role in somatosensory processing, can also synaptic tracing with optogenetic, chemogenetic and
contribute to emotional states in pain.
lesioning tools, it was further demonstrated that specific

460 | August 2021 | volume 22 www.nature.com/nrn

0123456789();:
Reviews

Table 1 | Annotations used to describe frontal regions in rodent and human brain been further subdivided into the pregenual (rostral)
versus subgenual (ventral) regions50,51. Mounting evidence
Species Common terms used Frontal subregions Common from numerous human and rodent studies supports a
to describe frontal frequently studied in pain anatomical
subregions involved annotations dual role for the ACC in processing both the sensory and
in pain the emotional aspects of pain29,52–55. Hyperactivation of the
ACC is consistently reported in patients with chronic
Rodent mPFC, dmPFC, vmPFC, ACC, also referred to as mPFC Cg1, Cg2, area
ACC/ACA, rACC, and rACC 24a, area 24b pain and animal models with heightened pain percep-
prelimbic mPFC, tion, and has also been associated with psychological or
Prelimbic, also referred to PL, PrL, Cg3,
prelimbic PFC, PL/PrL,
as mPFC, dmPFC, prelimbic area 32 negative emotional factors in pain, including aversion,
IL/ILA, medial FC, unpleasantness, social rejection and distress1,55. However,
rostral MC, caudal mPFC, dorsal cingulate cortex
MC, medial OC, ORB, and rostral MC it remains unclear whether distinct ACC circuits modu-
MO, AI Infralimbic, also referred to as IL, ILA, area 25 late behavioural responses to noxious stimuli and affec-
mPFC, vmPFC, caudal cingulate tive evaluation and whether they undergo independent
cortex and rostral MC maladaptive changes during pain chronicity.
Human dlPFC, mPFC, dmPFC, dlPFC Brodmann areas
vmPFC, vlPFC, 8, 9 and 46 Dorsal ACC. The human dorsal (dACC), also referred
amPFC, anterior PFC, to as the anterior MCC, is implicated in diverse neuro-
frontopolar PFC, rostral ACC Brodmann areas
PFC, ACC, dACC, 24, 25, 32 and 33 logical processes, including empathy, action evaluation
pregenual (rostral) and negative pain affect, wherein activation of the dACC
ACC, subgenual by noxious stimuli has been consistently reported in the
(ventral) ACC, OFC imaging literature51,56,57. Lesioning of the dACC has also
ACA, anterior cingulate area; ACC, anterior cingulate cortex; AI, agranular insular area; been successful for the treatment of chronic refractory
amPFC, anteromedial prefrontal cortex; Cg, cingulate area; dACC, dorsal anterior cingulate pain and affective disorders58. Interestingly, a recent
cortex; dlPFC, dorsolateral prefrontal cortex; dmPFC, dorsomedial prefrontal cortex;
FC, frontal cortex; IL, infralimbic cortex; ILA, infralimbic area; MC, medial cortex; MO, secondary report demonstrated that positive connectivity between
motor area; mPFC, medial prefrontal cortex; OC, orbital cortex; OFC, orbitofrontal cortex; the dACC and the primary motor cortex (M1) could be a
ORB, orbital area; PFC, prefrontal cortex; PL and PrL, prelimbic cortex; rACC, rostral anterior
cingulate cortex; vlPFC, ventrolateral prefrontal cortex; vmPFC, ventromedial prefrontal cortex.
useful predictor of chronic pain development in survivors
of motor vehicle trauma injuries59.

deactivation of the pathway from the PL cortex to the Rostral and ventral ACC. The perigenual ACC, also
ventrolateral PAG, via enhanced GABAergic tone in referred to as the rostral ACC (rACC), has extensive
the PL cortex, functionally contributes to hypersensitiv- connections to other cingulate areas and the PAG50.
ity in mice with neuropathic pain47. Such an observa- Disrupted rACC connectivity with brain regions of
tion is supported by a large number of human imaging the default mode network has been found in patients
studies which imply that a deactivation of the mPFC in with chronic back pain19, while enhanced connecti­
patients with neuropathic pain is associated with a loss of vity with the salience and sensorimotor networks was
descending inhibition9. The role of the IL–PAG circuit in recently demonstrated in patients with migraine and
chronic pain pathology remains to be clarified. visceral pain60,61, highlighting atypical rACC function
in aberrant pain processing.
PFC–amygdala pathway. The mPFC is reciprocally con- Imaging data suggest a sex-​dependent role of the
nected with the amygdala. Increased intracorticolimbic ventral ACC, also known as the subgenual ACC,
white matter and functional connectivity within the in pain modulation, with female patients with chronic
mPFC–amygdala–hippocampus circuitry has been pain displaying greater subgenual ACC connectivity to
identified as a risk factor for persistence of back pain48. areas of the default mode and sensorimotor networks
In rodents, principal cells in both the PL cortex and the than male patients with chronic pain, suggesting that
IL cortex project to neurons in the BLA and the inter­ subgenual ACC circuitry in women is associated with
calated cell mass of the amygdala; this is critical for corti­ a stronger engagement of descending modulatory sys-
cally driven feedforward inhibition of the amygdala, and tems that likely contribute to better pain adaptation
its impairment leading to hyperactivity of the BLA has and habituation to repeated noxious stimuli62,63.
been linked to nociceptive hypersensitivity in chronic
pain46,49 (Fig. 1). Recently, specific dysfunction in the Rodent ACC. Extensive rodent studies have provided
IL–amygdala pathway has also been reported in chronic detailed insights into ACC mechanisms in pain55,64.
pain models 49, in line with structural tracing data Several types of ACC pyramidal populations have
showing that the BLA is innervated by neurons present been identified on the basis of their firing properties in
in the superficial and deep layers of the IL cortex46. response to noxious and non-noxious inputs 65,66.
In chronic pain models, layer 2/3 neurons undergo
Anterior cingulate cortex structural changes, with increased dendritic complexity
Although the ACC domain of the cingulate is classically and spine densities, and become hyperexcitable37,67,
considered to be part of the PFC, we discuss the roles of similar to observations in layer 2/3 neurons of the PL
the ACC in pain in this section to enable a better compar- cortex. However, unlike in the PL cortex, intrinsic excit-
ison with the other cingulate domain, namely, the MCC, ability of layer 5 ACC pyramidal neurons is potentiated
later. The human ACC is often subdivided into dorsal ver- in chronic pain, while interneurons appear to remain
sus ventral regions, wherein the ventral portion has also unaffected after nerve injury52,66.

NAture RevIews | NeurOSCienCe volume 22 | August 2021 | 461

0123456789();:
Reviews

Seminal work by Zhuo and colleagues has high- in nociception70. However, other studies have demon-
lighted the role of synaptic plasticity in the ACC in both strated that neuropathy-​induced hypersensitivity is
acute and chronic pain, particularly in various forms of not consistently affected by lesions or inhibition of the
long-​term potentiation triggered by activation of NMDA ACC71,72, suggesting that other brain areas may play a
receptors, kainate receptors, adenylyl cyclase and diverse more critical role in the sensory processing during pain
signalling kinases in the ACC, all of which were linked states (such as the MCC and the insula; see later).
to neuronal hyperexcitability55,68. Recently, studies also ACC hyperexcitability has also been linked to
reported altered activity of HCN channels in pyramidal pain-​associated negative affect in various rodent models
neurons, leading to impaired Ih currents and significant of chronic pain, commonly reflected by a reduced
loss of reciprocal synaptic connections between inhib- place escape avoidance or conditioned place aversion
itory and pyramidal layer 5 populations, both of which induced by a noxious stimulus. Lesion or suppression
are suggested to cause disinhibition of the local ACC of neuropathy-​induced or inflammatory pain-​induced
microcircuitry in chronic pain66,69. Optogenetic experi- ACC hyperactivity reduces pain aversion72–74, while elec-
ments indicate a role for the PV-​type, but not SOM-​type, trical or optogenetic stimulation of the ACC in naive
interneurons in the regulation of the ACC excitability animals sufficiently induces aversive behaviours29,75.

a Structural Functional Mechanisms


1
↑ Basal spine density ↑ Excitability (neuropathic) Alterations in HCN channels,
2/3 ↑ Noradrenergic innervation ↑ Excitability (inflammatory) α2-receptor kinetics and
Cortical layers

↓ Excitatory inputs ↓ Inhibition (neuropathic) NMDA-mediated currents

↓ Apical spine complexity ↓ Excitability (neuropathic) Alterations in M1 and CB1


5 ↓ Excitatory inputs ↓ Excitability (inflammatory) receptor expression
↑ Inhibition (neuropathic)
6

CeA LC

Layer 5 Layer 2/3

NAc
ITC

BLA PL vIPAG
cortex

Sensory pathways Excitatory pyramidal neuron


Increased activity in hypersensitivity Excitatory neuron
Decreased activity in hypersensitivity GABAergic neuron
Affective pathways PV-type GABAergic neuron
Decreased activity with more negative pain affect Noradrenergic neuron

Fig. 1 | The PL cortex. a | Structural and functional plasticity reported in layer 2/3 and layer 5 neurons of the prelimbic
(PL) cortex. b | Major input and output pathways of the PL cortex known to modulate sensory states (in green) and negative
affective states (in blue) in acute and chronic pain. Parvalbumin (PV)-​type GABAergic neurons in layer 5 provide feedforward
inhibition leading to PL cortex deactivation and subsequently mechanical hypersensitivity during chronic pain. Modulation
of PL PV-​type GABAergic neurons by basolateral amygdala (BLA) efferents in pain is also shown. Increases (solid line)
and decreases (dotted line) in pathway activity leading to nociceptive hypersensitivity and pain aversion are indicated.
Light-​grey dashed lines indicate the delineation of cortical layers. CeA, central amygdala; HCN channels, hyperpolarization-​
activated cyclic nucleotide-​gated channels; ITC, intercalated cell mass of the amygdala; LC, locus coeruleus; M1, primary
motor cortex; NAc, nucleus accumbens; vlPAG, ventrolateral periaqueductal grey.

462 | August 2021 | volume 22 www.nature.com/nrn

0123456789();:
Reviews

1 ACC MCC PI 1 S1 S2
Cg1
2/3 2/3

5 Cg2 5

6 6

NAc ZI Striatum
Co-morbid
pain

BLA
PAG
Parafascicular
nucleus
MD VTA
RVM

Spinal cord

Sensory pathways Excitatory pyramidal neuron


Increased activity in hypersensitivity Excitatory neuron
Decreased activity in hypersensitivity GABAergic neuron
Affective pathways Dopaminergic neuron
Increased activity with more negative pain affect Serotonergic neuron
Decreased activity with more negative pain affect

Fig. 2 | Overview of major neocortical circuits and their known efferent pathways that modulate nociception
(in black) and negative affective states (in red) in acute and chronic pain. Top-​down circuits with which excitatory
neurons in the deep layers of the anterior cingulate cortex (ACC), posterior insula (PI) and primary somatosensory cortex
(S1) send projections directly and indirectly to the dorsal horn of the spinal cord to modulate descending modulation
of sensory inputs, all of which can contribute to central sensitization under physiological and pathological conditions,
leading to heightened pain responses. Increases (solid line) and decreases (dotted line) in pathway activity leading to
nociceptive hypersensitivity and pain aversion are indicated. BLA, basolateral amygdala; Cg1, cingulate area 1; Cg2,
cingulate area 2; MCC, midcingulate cortex; MD, mediodorsal thalamus; NAc, nucleus accumbens; PAG, periaqueductal
grey; RVM, rostroventral medulla; S2, secondary somatosensory cortex; VTA, ventral tegmental area; ZI, zona incerta.

ACC–NAc and ACC–VTA pathways. Despite strong evi- again that specific prefrontal domains carry out highly
dence implicating its role in pain-​related aversion, the different roles via specific projections.
underlying ACC microcircuitry has not been as exten-
sively studied. Recent data suggest that the ACC regu- ACC–spinal cord pathway. Recently, spinally project-
lates pain-​related aversion via distinct subdomains and ing neurons residing in layer 5 of the ACC in mice were
pathways differentially from behavioural hypersensitivity. linked to pain modulation (Fig. 2). Electrical and selec-
Optogenetic studies29 indicate a dichotomy between the tive optogenetic stimulation of these ACC spinal cord-​
rostral division versus the middle division of the cingulate projecting neurons was found to facilitate spinal sensory
cortex in processing pain affect, with the rACC having a excitation and mechanical hypersensitivity77. Conversely,
key role in generating aversion. In neuropathic models, inhibiting the same pathway alleviated neuropathy-​
viral tracing and chemogenetic tools confirm the role of induced hypersensitivity, providing evidence for a direct
ACC projections to mesolimbic dopaminergic regions, top-​down descending facilitatory role of the ACC in
namely, the NAc and ventral tegmental area (VTA), in chronic pain that is independent of descending pathways
pain aversion but not in mechanical or thermal hyper- that originate in the brainstem77.
sensitivity induced by nerve injury76 (Fig. 2). It remains to
be determined whether this ACC–NAc pathway is related The MCC in pain
to the finding that direct medial thalamus inputs to ACC Since the clarification of the MCC as a cytoarchitec-
subcortically projecting cells modulate pain aversion66. turally and functionally distinct region of the cingulate
These observations are interesting, since an earlier study cortex78, it has been implicated in numerous brain pro-
revealed an opposing role of the PL–NAc pathway (Fig. 1), cesses, including pain anticipation and sensitivity56,79.
that is, inhibition of pain-​related aversion42, highlighting In rodents, as discussed earlier, major efforts have been

NAture RevIews | NeurOSCienCe volume 22 | August 2021 | 463

0123456789();:
Reviews

devoted to investigating the role of the ACC and its Activity rhythms in the S1. Within the S1 region,
associated circuits in pain-​related processes. How­ human electrophysiological studies frequently demon-
ever, although the MCC receives a similarly wide array strate a strong enhancement of gamma-​band power
of inputs from cortical and subcortical regions as the that is driven in response to brief nociceptive stimuli
ACC 80, the identification of specific MCC circuits and associated with the subjective pain intensity18,85–87.
modulating pain has remained sparse. Concurrently, transient decreases in low-​f requency
(below 30 Hz) oscillatory powers in the sensorimotor
MCC–PI pathway. Recently, a study highlighted the regions are often observed, which have been proposed
involvement of the novel pathway from the MCC to the PI to reflect sensory gating mechanisms and to facilitate
that contributes to acute and persistent inflammatory motor reactions to the painful threat17,88,89.
pain, but not to chronic neuropathic pain, and evokes Mechanistic insights into the functional roles of
nociceptive hypersensitivity in the absence of periph- these S1 oscillations in pain were recently explored in
eral injury by PI-​dependent activation of descending animal models. These studies revealed that S1 gamma
serotonergic facilitation, while MCC–NAc projections rhythms were evoked in response to brief noxious
did not modulate nociception29 (Fig. 2). Moreover, opto- somatosensory stimuli but not in response to other
genetic activation experiments indicated that the MCC sensory stimulation (for example, visual), motor
functionally modulates pain independently of affective activity or salience83,90. Such S1 gamma rhythms were
or emotional aspects, and implicated the rACC in these linked to activity of interneurons83,91, whereby opto-
aspects instead29. genetic gamma entrainment of S1 PV-​type interneu-
rons in particular led to nociceptive hypersensitivity
MCC–zona incerta pathway. Recently, Hu and col- via activation of descending serotonergic pathways in
leagues further discovered that selective MCC excitatory the brainstem, providing the first causal evidence
projections (dorsal cingulate area 1 (Cg1) versus ventral in vivo for a cell-​type-​specific and pathway-specific
cingulate area 2 (Cg2)) to the GABAergic population mechanism for S1 gamma rhythms in acute pain83.
residing in the zona incerta (ZI), a subthalamic region, Interestingly, S1 gamma entrainment also elicited
undergo differential activity during chronic neuropathic pain aversion and negative affect, likely via facilitating
pain81, wherein activity of the Cg2–ZI pathway was information flow to the rACC, a cortical area critically
dampened (Fig. 2). Interestingly, restoring activity of this implicated in pain affect29. Under persistent pain con-
pathway in particular, but not of the Cg1–ZI pathway or ditions, enhanced spontaneous gamma activity in the
the Cg2–PAG pathway, significantly reduced nocicep- S1 is also observed, with increases in gamma power
tive hypersensitivity, suggesting that a dysfunction of the positively correlating with the severity of pain83,92, high-
endogenous inhibitory system comprising the MCC Cg2 lighting the significance of S1 gamma activity not only
and ZI is associated with neuropathic pain. Collectively, in physiological pain but also extending to pathological
these studies suggest that distinct MCC subdomains are pain conditions.
operational and perform different roles in the process- Whether and how the S1 contributes to chronic
ing of acute and chronic pain29,81, and suggest that the pain is debated. Several studies have reported a lack of
MCC serves as an origin of pathways mediating inhi- phenotypic alterations in nociception and pain upon
bition (MCC Cg2–ZI) as well as descending facilitation lesions or optogenetic silencing of the S1 in rodents29,93.
(MCC–PI–RVM) of nociception (Fig. 2). Conversely, persistent hyperactivity of S1 neurons
has been reported in models of neuropathic pain94,95,
Primary somatosensory cortex which has been attributed to reduced activity of local
The S1 is one of the most studied cortical areas for pain SOM-​expressing GABAergic interneurons95, unlike the
perception, and has commonly been thought to repre- key role ascribed to PV interneurons in local circuit
sent the sensory-​discriminative component of pain. In dysfunction in the PFC31.
recent years, however, it has been revealed that this is Finally, peripheral nerve injury triggers maladaptive
an overgeneralization and that a number of functions, structural plasticity and cortical reorganization in the
including emotions, can also influence pain processing S1 in chronic pain states of different causes, including
in the S1 (refs82,83). phantom limb pain4,96. Causal relationships between cor-
Recent in vivo calcium imaging experiments in mice tical structural remodelling and pain chronicity have not
revealed complex processing of incoming somatosen- yet been established, and it remains to be determined
sory information in layer 2/3 neurons of the S1, where whether these constitute a cause or a consequence of
distinct neurons have selective tuning properties that chronic pain4. In the following sections, we review effer-
encode multiple features, such as texture, dynamics ent pathways of the S1 that have been implicated in pain
and intensity of cutaneous stimuli, suggesting that tac- modulation.
tile information is well discriminated at the single-​cell
level in the S1 (ref.82). In parallel, imaging and microe- S1‒dorsolateral striatum pathway. Recently, viral tract
lectrode recordings in the primate indicate that there is tracing identified a novel pathway from layer 5 S1 gluta­
a functional and spatial segregation between groups of matergic neurons projecting to GABAergic neurons
neurons processing innocuous tactile versus nociceptive in the caudal dorsolateral striatum (Fig. 2). Selective
heat and cold information within the S1 (ref.84), raising chemogenetic and optogenetic experiments suggest
the possibility of modality-​specific thalamic inputs to that this S1–striatum pathway functionally contributes
S1 subregions. to inflammatory mechanical hypersensitivity97.

464 | August 2021 | volume 22 www.nature.com/nrn

0123456789();:
Reviews

S1‒ACC pathway. A recent study using a combination shown18,29,109, while robust phase-​locked gamma-​band
of tract tracing, optogenetics, in vivo electrophysiology oscillations in response to acute and sustained ther-
and machine learning analysis reported the involvement monociceptive but not non-​nociceptive stimuli can be
of a corticocortical S1–ACC pathway in nociception98. recorded in both the AI and the PI110,111. Intracranial and
The study authors identified an ACC neuronal popu- imaging recordings suggest that information from noci-
lation receiving direct inputs from the S1, which is pre­ ceptive inputs is directed from the PI to the AI, with the
ferentially responsive to noxious stimuli, show enhanced posterior and mid-​regions of the PI strongly connected to
responsivity during inflammatory pain and are function- sensorimotor cortices and associated with encoding for
ally linked to sensory and pain-​related aversive behav- sensory aspects such as intensity, somatotopy and modal-
iours in a chronic pain model98 (Fig. 2). This suggests that ities, while the AI is strongly connected to PFC regions
direct relay of nociceptive information between these and functionally integrates affective processes related to
neocortical centres serves to integrate sensory and affec- the painful sensation112, although a recent study has sug-
tive components of pain. Indeed, previous studies have gested that the AI is also capable of rapidly conveying
shown that electrical responses of the ACC correlated and integrating limbic and sensory inputs in parallel113.
with S1 neuronal activity during peripheral stimulation94 Abnormal intrinsic insular cortical reorganization has
and that optogenetic manipulation of S1 activity can also been associated with prolonged pain in patients, with
lead to enhanced ACC activation83. decreased grey matter volume in various insula sub-
regions being detected as well as enhanced functional
S1–spinal cord pathway. Labelling studies in mice have connectivity between the PI and regions such as the
identified corticospinal projections from the S1 hind- cerebellum and the hippocampus being observed114.
limb area to laminae III–V of the spinal cord, suggesting Animal studies also support a key role for the insula
a role for these projections in tactile sensory processing99. in both nociceptive and affect modulation29,108. Indeed,
These corticospinal projections likely co-​terminate with there is overwhelming evidence for the critical contri-
low-​threshold mechanosensory afferents in the spinal butions of injury-​triggered long-​term potentiation and
cord100, since selective ablation of these neurons reduced plasticity in the insula in pathological pain115,116. Novel
responses to innocuous tactile but not to nociceptive work with viral tracing and pharmacological inhibition
stimuli. Importantly, ablation or functional suppression indicates that acute peripheral hypersensitivity induced
of this pathway attenuated evoked mechanical hyper- by activation of the MCC–PI pathway is mediated by
sensitivity after nerve injury, highlighting that these gluta­matergic projections from the PI to serotonergic
S1 corticospinal projections contribute to the generation neurons in the raphe nuclei and subsequently to the spinal
of pathological nociceptive hypersensitivity99 (Fig. 2). facilitation via descending facilitatory mechanisms29
(Fig. 2). Using fibre photometry and calcium imaging
Secondary somatosensory cortex recordings, an elegant study demonstrated that high
Intracerebral stimulation of the S2 area in patients levels of neural activity in the PI correspond to the
undergoing brain surgery for various reasons elicits animal’s state of anxiety and that a subset of these insu-
somatosensory responses and painful sensations, but lar neurons are also activated upon a painful tail shock
not non-​somatosensory responses such as auditory, ves- stimulus108. Furthermore, optogenetic stimulation of the
tibular or olfactogustatory sensations101. Somatosensory PI–central amygdala (CeA) pathway, but not the PI–NAc
responses in the S2 area correlate with the intensity of projections, strongly modulated aversive and anxiety-​like
stimuli, contributing to the encoding and discrimination behaviours108, suggesting that aversive inputs that induce
of somatosensory stimulus intensities102,103. fear and anxiety arrive at the CeA via the PI. The CeA
Surprisingly, in animal models, the role of the S2 in also receives direct inputs conveying information per-
pain has only recently begun to attract interest. Recent taining to aversive stimuli from the parabrachial nucleus5
imaging data from awake neuropathic mice indicated as well as from the BLA, and recent in vivo calcium
enhanced neural activity in the S2 region of rodents with imaging demonstrated that a distinct neuronal ensem-
nerve injury104. Recently, neurons in the parafascicular ble in the BLA encodes the aversive state during noxious
thalamic nucleus were reported to relay to the S2, and stimulations117. Hence, it would seem that several parallel
the decrease in their excitatory tone was observed to pathways, some involving the neocortex and others of
modulate depression-​associated co-​morbid pain, but not subcortical origin, encode aversion and are related to
inflammatory or neuropathic pain105. It remains to be fear induction following pain perception. It remains to be
further investigated whether and how specific S2 efferent clarified as to how either of these pathways contributes
pathways influence pain processing. to the persisting hyperanxious behaviours that typically
develop in models of chronic pain118.
Insular pathways in pain
The insular cortex is involved in an enormous array Cortical circuits as pharmacological targets
of functions, including homeostatic and interoceptive Opioidergic drugs
processes, motivation, addiction, sensorimotor integra- A well-​described mechanism of opioid analgesia is
tion and processing of emotions such as pain, anxiety attributed to descending inhibition of spinal nocice-
and empathy106,107, and is well connected with numer- ption via actions at diverse stations along descending
ous cortical and subcortical structures108. Acute painful pathways, including the PAG, the RVM and other brain-
responses arising from selective neural stimulation of stem nuclei, and via direct actions on primary nocic-
the insula in both animals and humans are consistently eptors and the spinal cord119. Cortical pathways are a

NAture RevIews | NeurOSCienCe volume 22 | August 2021 | 465

0123456789();:
Reviews

Mindfulness/
OFC self-regulation Opioids

Placebo
Placebo Ketamine CPM rTMS/tDCS
rTMS tACS

vlPFC dlPFC S1–S2 Posterior


rACC sensorimotor insula M1
mPFC

Opioids Cannabinoids Cannabinoids


(pain affect)

Opioids Placebo
Placebo
(sensory)

Opioids
+ (reward) CPM
Amygdala PAG Thalamus
NAc Opioids

CPM
RVM
Opioids

Pharmacological therapy SC Opioids


Non-pharmacological therapy

Fig. 3 | Overview of neocortical structures and their known efferent pathways that serve as targets of
pharmacological analgesics as well as neurostimulation and neuromodulation/cognitive therapies in chronic pain.
Black arrows point to regions and pathways targeted by each therapy. Both neocortical processing and efferent pathways
(represented by grey arrows) to brain centres mediating reward learning (nucleus accumbens (NAc)) and conditioned
learning (amygdala) and descending modulation of nociception (periaqueductal grey (PAG) and rostroventral medulla
(RVM)) are affected. Commonalities as well as differences are found with respect to neocortical mechanisms underlying
diverse therapies, providing a neurobiological basis for an additive or synergistic potential with combination therapies.
Anatomical pathways are depicted by grey arrows. Each mode of therapy is depicted in differently coloured text and
with arrows to the brain regions involved. CPM, conditioned pain modulation; dlPFC, dorsolateral prefrontal cortex;
M1, primary motor cortex; mPFC, medial prefrontal cortex; OFC, orbitofrontal cortex; rACC, rostral anterior cingulate
cortex; rTMS, repetitive transcranial magnetic stimulation; S1, primary secondary somatosensory cortex; S2, secondary
somatosensory cortex; SC, spinal cord; tACS, transcranial alternating current stimulation; tDCS, transcranial direct
current stimulation; vlPFC, ventrolateral prefrontal cortex.

highly promising target, given that opioids particularly to play a role in reward processing, during noxious
modulate the affective dimension of pain, but have not stimulation119.
been studied as widely as the brainstem–spinal cord axis The notion that opioids inhibit the aversive elements
in terms of opioidergic modulation. The expression of of pain more strongly than its sensory component, par-
µ-​opioid receptors (MORs), which account for a major ticularly at lower doses, is consistent with reports that
proportion of opioidergic analgesia, is particularly dense low opioid doses are sufficient to fully block activa-
in cortical areas, including the ACC, the PFC and the tion of the AI and other centres in the limbic system,
insula. For instance, the distribution and modulation of whereas increasing concentrations of opioids inhibit the
opioid receptors in the ACC has been well described in components of the nociceptive matrix, such as the S1
recent studies120,121. and S2 and the PI124. In animal models, seminal work by
Opioids dose dependently decrease pain-​related Navratilova, Porreca and colleagues demonstrated that
blood oxygen level-​dependent signals independently activating opioid receptors specifically in the rACC,
of opioid-​induced vascular changes and suppress using opioid doses lower than those that inhibit nocicep-
pain-​related potentials in humans in the insula, S1, S2 tive hypersensitivity, was sufficient to attenuate ongoing
and ACC119,122 (Fig. 3). Functional near-​infrared spectros- neuropathic pain125. There is a dichotomy of opioider-
copy also revealed that the response of the entire mPFC gic actions on pain affect (via the rACC‒NAc pathway)
region (Brodmann area 10) and the contralateral S1 to and sensory qualities of nociception (via the RVM
noxious stimuli is repressed by morphine in correlation descending axis)126. Opioidergic actions in the rACC
with drug plasma concentration123. Paradoxically, opi- stimulate release of dopamine in the NAc125, indicating
oids have also been linked to increased activity in the activation of prefrontal–NAc connectivity and reward
ACC and the OFC, a region that has been suggested pathways, and are, in turn, modified by the CeA127, thus

466 | August 2021 | volume 22 www.nature.com/nrn

0123456789();:
Reviews

suggesting a role for amygdala‒cingulate–striatal path- of analgesic drugs and thus reduce their side effects136.
ways in opioid analgesia. Opioidergic mechanisms in Addressing the mechanisms and precise circuits under-
the rACC have also been implicated in reduction of lying analgesia by neuromodulation therapies that target
ongoing pain by non-​opioidergic analgesic drugs, cortical domains is now an exciting emerging field136.
including gabapentin128 and α2-​adrenergic agonists, such
as clonidine125. Opioidergic agonists microinjected into Neurostimulation-​induced analgesia
the ventro-​orbital cortex reversed mechanical and cold As per the most recent clinical standpoint, evidence-
hypersensitivity after nerve injury via MORs129. based guidelines indicate definite efficacy of high-
frequency non-invasive repetitive transcranial magnetic
Cannabinoids stimulation (rTMS) of the contralateral M1 for allevi­
While cannabinoidergic actions at the level of periph- ating neuropathic pain and probable efficacy of
eral mechanisms and spinal nociceptive transmission high-frequency rTMS of the left M1 for improving
are well described, insights into modulation of brain quality of life or of the left dlPFC for alleviating pain in
pathways as a basis for cannabinoidergic analgesia are patients with fibromyalgia136 (Fig. 3). Interestingly, neo-
only recently starting to emerge. In a human model of cortical enhancement of supraspinal opioidergic inhibi-
acute pain, Δ9-​tetrahydrocannabinol reduced unpleas- tion may only partly account for the clinical efficacy of
antness and ongoing pain in association with activa- neurostimulation. In rTMS of the M1, release of opioids,
tion of the right amygdala and reduced connectivity but not of dopamine, is reported in diverse neocortical
between the sensorimotor cortex and the amygdala130. areas, including the medial OFC, ACC, left insula and
In patients with chronic radicular neuropathic pain, dlPFC137, but transcranial direct current stimulation
Δ9-​tetrahydrocannabinol reduced functional connectiv- of the M1 failed to lower the required opioid dosage138.
ity between the ACC, the dlPFC and the sensorimotor By contrast, analgesia induced by rTMS of the left dlPFC
cortex in a manner predictive of the analgesic efficacy131 has an opioid-​sparing effect138 and is inhibited by the
(Fig. 3). Thus, the emerging view is that cannabinoids MOR antagonist naloxone139. More recently, manipula­
disrupt higher-​order sensorilimbic connections and tion of somatosensory cortical alpha-band activity
that the resting-​state strength of these connections may using transcranial alternating current stimulation was
be used as a predictive measure for cannabinoid-​based associated with lower levels of reported pain in both
therapies in clinical applications. healthy individuals140 and patients with chronic pain141,
suggesting that targeted oscillatory modulation with
Ketamine and antidepressant drugs transcranial alternating current stimulation could also
Ketamine (or its S enantiomer, esketamine) is gaining be harnessed for pain relief.
clinical prominence for its analgesic actions as well as Rodent models involving direct (invasive) or trans-
rapid-​onset and strong antidepressive qualities132. In ani- dural electrical stimulation of the M1 implicate an
mal models, ketamine has recently been demonstrated inhibition of neuronal activity in thalamic relay nuclei,
to relieve depression-​related behaviours in neuropathic suggesting suppression of nociceptive signals in the
mice, accompanied by restoration of depleted levels of sensory neurons in the somatosensory pathway in
brain-​derived neurotrophic factor and neuroligins in the neuro­pathic rats, as well as disinhibition of neurons
PFC, ACC and hippocampus133. Interestingly, a low dose in the PAG that mediate descending inhibition of
of ketamine attenuates the aversive component of pain spinal nociception142.
for longer than its antinociceptive effect, which was also
tied to suppression of activity in the ACC73 (Fig. 3). Conditioned pain modulation
Antidepressant drugs, which act by blocking reuptake Conditioned pain modulation (CPM) refers to the phe-
of serotonin and noradrenaline and activating brainstem nomenon of pain being inhibited by a conditioning
descending inhibition, show significant clinical benefits pain stimulus applied to another part of the body, and
as analgesic and co-​adjuvants in neuropathic pain8,134. recent data reveal the importance of neocortical regions
Very little is known about how they modulate pain in modulating brainstem nuclei which ultimately elicit
affect, and given the high density of neocortical projec- CPM via suppression of spinal nociception143. CPM
tions of brainstem serotonergic and noradrenergic neu- is compromised in some forms of chronic pain, and a
rons, neocortical contributions deserve to be explored meta-​analysis revealed deficits in CPM in patients with
in detail. Interestingly, a recent study demonstrated chronic low-​back pain in a manner proportional to pain
that chemogenetic activation of prefrontally projecting chronicity144. CPM is associated with increased activity of
noradrenergic neurons located in the locus coeruleus the OFC and the amygdala, followed by suppression
actually induced aversion and spontaneous pain rather of typically pronociceptive brain regions, such as the
than eliciting antinociception135. S1, S2 and insula145 (Fig. 3). Conversely, healthy individ-
uals lacking CPM analgesia in a paradigm of acute pain
Non-​pharmacological therapies demonstrate enhanced activation of the ACC and the
Owing to problems with conventional analgesics, as PFC146.
seen at worst in the current opioid crisis in the United
States with extensive use and harmful reliance on opi- Cognitive control of pain
oid analgesics, there is growing interest in develop- Given that expectancy and attentional factors can
ing non-​pharmacological alternatives, particularly as strongly modulate pain perception, there has been rising
adjunct therapies to help lower the dose and frequency interest in exploring the use of cognitive manipulation

NAture RevIews | NeurOSCienCe volume 22 | August 2021 | 467

0123456789();:
Reviews

for reducing perceived pain and elucidating the neural consisting of the PFC and parietal cortex, and the rACC/
underpinnings thereof. While activation of endogenous mPFC, is suggested to predict and shape expectancy-​
opioidergic pathways has been implicated in several induced modulation of pain156. Woo et al.157 found that
cognitive strategies for pain control, including placebo self-​regulation to enhance or suppress pain in response
analgesia, hypnosis and attentional control147, there are to acute noxious stimuli does not influence the neuro-
substantial reports on opioid-​independent mechanisms, logic pain signature, but rather uses a frontostriatal path­
such as in mindfulness meditation-​induced analgesia148. way between the ventromedial PFC and the NAc, a
There is mounting evidence for a critical role for path- pathway that has been implicated in emotional appraisal.
ways originating in the neocortex in emerging cognitive
strategies for pain relief. Summary and outlook
Taken together, recent studies using modern circuit
Placebo analgesia. Placebo analgesia can be inhibited interrogation tools have unveiled critical insights into
by naloxone, which acts by hindering coupling between how individual neocortical domains process incoming
the rACC and the PAG in placebo responders149 (Fig. 3), noxious and non-​noxious inputs and scale them to create
indicating involvement of descending modulation of pain perceptions of various intensities and aversive qua­
nociception. However, recent human studies essentially lities via interactions with other cortical and subcortical
implicate opioidergic pathways as well as cortical circuits structures. While the emerging picture supports the con-
governing expectation and conditioned learning, orig- cept that pain is essentially the functional product of a
inating mainly from diverse PFC domains, in placebo broad and redundant network of brain structures, there
analgesia. Although suppression of the S2 and other, typ- is also mounting evidence for functional segregation of
ically ‘nociceptive’ cortical regions has been reported in distinct aspects of sensory coding, intensity coding, aver-
placebo analgesia150,151, the neurologic pain signature2 is sion and negative affect across neocortical domains and
modulated by placebo treatment to a minor extent only, specific pathways, with important differences between
suggesting that the mechanisms underlying placebo anal- acute and chronic pain and between inflammatory and
gesia transcend modulation of nociceptive processing152. neuropathic pain.
Moreover, placebo responders show enhanced functional This knowledge, however, is by no means complete,
connectivity of the rACC not only with the PAG but also and the much-​needed fine-​grained analysis of specific
with the amygdala, thus implicating neocortical modu- circuits linking cells to behaviour will need to continue
lation of subcortical structures governing conditioned and be extended to new studies to unravel how attention,
learning153. Direct comparison of brain regions acti- expectation, anxiety, fear and stress modulate and shape
vated during opioid treatment versus placebo analgesia the experience of pain, given their importance in pain
revealed rACC activation in both; however, the lateral chronicity. In the neocortex, encoding of pain modalities
OFC and the ventrolateral PFC show stronger activation in neuronal ensembles and signal processing and modu­
during responsivity to placebo treatment than during lation in precisely organized laminar columns remains
opioid analgesia154, consistent with their role in the pro- to be elucidated. Moreover, there are a number of seem-
cessing of expectation and error signals and contextual ingly paradoxical findings that require clarification, and
evaluation of sensory stimuli. The mPFC and the lateral a further cellular delineation of pathways promoting
PFC are activated during anticipation of placebo analge- pain versus those fostering pain relief is key to achieving
sia, while the right PFC and the dlPFC are most strongly therapeutic success. As knowledge of neural specificities
activated during placebo analgesia150 (Fig. 3). A recent of neocortical efferent pathways controlling pain accrues
study used gabapentin-​induced, naloxone-​sensitive from rodent models, a major challenge ahead is to use
Pavlovian conditioning to model placebo analgesia in these insights to increase the efficacy and improve the
rodents and reported enhanced activity in the PL domain safety of non-​pharmacological neurostimulation-​based
of the PFC, the NAc and the ventrolateral PAG155, thereby and neuromodulation-​based therapies. Finally, the bur-
opening up perspectives for functionally interrogating geoning knowledge of neocortical circuits can also pave
circuits underlying placebo analgesia. the way for designing novel pharmacological therapies
if it is supplemented by detailed analyses of molecular,
Mindfulness and self-​regulation. Mindfulness meditation genetic and epigenetic mechanisms underlying signal-
has been reported to suppress pain intensity and unpleas- ling specificity and plasticity. Here, a critical basis for
antness and use brain mechanisms distinct from those specific cell-​targeted therapies could be achieved via
used in placebo analgesia150, including a stronger activation application of the newly developed single-​cell transcrip-
of the OFC, subgenual ACC and AI, which are primarily tomics and epigenomics technologies to key neocortical
associated with cognitive and affective modulation of circuits in future studies.
pain and interoceptive awareness150 (Fig. 3). The strength
of intrinsic connectivity in the frontoparietal network, Published online 14 June 2021

1. Baliki, M. N. & Apkarian, A. V. Nociception, pain, chronic pain. Nat. Rev. Neurosci. 14, 502–511 6. Braz, J., Solorzano, C., Wang, X. & Basbaum, A. I.
negative moods, and behavior selection. Neuron 87, (2013). Transmitting pain and itch messages: a
474–491 (2015). 4. Kuner, R. & Flor, H. Structural plasticity and contemporary view of the spinal cord circuits
2. Wager, T. D. et al. An fMRI-​based neurologic signature reorganisation in chronic pain. Nat. Rev. Neurosci. 18, that generate gate control. Neuron 82, 522–536
of physical pain. N. Engl. J. Med. 368, 1388–1397 20–30 (2016). (2014).
(2013). 5. Kuner, R. & Kuner, T. Cellular circuits in the brain and 7. Peirs, C. & Seal, R. P. Neural circuits for pain: recent
3. Bushnell, M. C., Ceko, M. & Low, L. A. Cognitive their modulation in acute and chronic pain. Physiol. advances and current views. Science 354, 578–584
and emotional control of pain and its disruption in Rev. 101, 213–258 (2020). (2016).

468 | August 2021 | volume 22 www.nature.com/nrn

0123456789();:
Reviews

8. Finnerup, N. B., Kuner, R. & Jensen, T. S. Neuropathic branching of layer 5 pyramidal cells contribute to an arthritis pain model. ACS Chem. Neurosci. 9,
pain: from mechanisms to treatment. Physiol. Rev. medial prefrontal cortex deactivation in a rat model 2252–2261 (2018).
101, 259–301 (2020). of neuropathic pain. Front. Cell Neurosci. 10, 133 50. Vogt, B. A. Submodalities of emotion in the context of
9. Seminowicz, D. A. & Moayedi, M. The dorsolateral (2016). cingulate subregions. Cortex 59, 197–202 (2014).
prefrontal cortex in acute and chronic pain. J. Pain 18, 33. Huang, J. et al. A neuronal circuit for activating 51. Vogt, B. A. Cingulate Neurobiology and Disease
1027–1035 (2017). descending modulation of neuropathic pain. (Oxford Univ. Press, 2009).
10. Woo, C. W. et al. Quantifying cerebral contributions Nat. Neurosci. 22, 1659–1668 (2019). 52. Blom, S. M., Pfister, J. P., Santello, M., Senn, W.
to pain beyond nociception. Nat. Commun. 8, 14211 This study reports that the synaptic input from & Nevian, T. Nerve injury-​induced neuropathic pain
(2017). the BLA on to GABAergic interneurons in the PL causes disinhibition of the anterior cingulate cortex.
In this study, functional imaging, multivariate pattern cortex is enhanced upon nerve injury via reduced J. Neurosci. 34, 5754–5764 (2014).
analysis and machine learning approaches are used endocannabinoid signalling. This results in 53. Johansen, J. P. & Fields, H. L. Glutamatergic activation
to identify the nature of cerebral regions that are enhanced feedforward inhibition of prefrontal of anterior cingulate cortex produces an aversive
activated in a stimulus intensity-​independent manner neurons that project to the ventrolateral PAG teaching signal. Nat. Neurosci. 7, 398–403 (2004).
to predict pain beyond nociceptive input and help and thereby alters descending noradrenergic and 54. Fuchs, P. N., Peng, Y. B., Boyette-​Davis, J. A. &
explain how psychological manipulations and serotonergic pathways. Uhelski, M. L. The anterior cingulate cortex and pain
expectation modulate pain. 34. Jones, A. F. & Sheets, P. L. Sex-​specific disruption processing. Front. Integr. Neurosci. 8, 35 (2014).
11. Da Silva, J. T. & Seminowicz, D. A. Neuroimaging of distinct mPFC inhibitory neurons in spared-​nerve 55. Bliss, T. V., Collingridge, G. L., Kaang, B. K. & Zhuo, M.
of pain in animal models: a review of recent literature. injury model of neuropathic pain. Cell Rep. 31, Synaptic plasticity in the anterior cingulate cortex
Pain. Rep. 4, e732 (2019). 107729 (2020). in acute and chronic pain. Nat. Rev. Neurosci. 17,
12. Carlen, M. What constitutes the prefrontal cortex? This study is the first to demonstrate that chronic 485–496 (2016).
Science 358, 478–482 (2017). neuropathic pain is modulated in a sex-​specific 56. Kragel, P. A. et al. Generalizable representations
13. Laubach, M., Amarante, L. M., Swanson, K. & manner involving alterations of distinct subclasses of pain, cognitive control, and negative emotion in
White, S. R. What, if anything, is rodent prefrontal of GABAergic neurons that differ between the male medial frontal cortex. Nat. Neurosci. 21, 283–289
cortex? eNeuro 5, ENEURO.0315-18.2018 (2018). mouse PL cortex and the female mouse PL cortex. (2018).
14. Shiers, S. & Price, T. J. Molecular, circuit, and 35. Huang, S., Borgland, S. L. & Zamponi, G. W. This study uses functional imaging and multivariate
anatomical changes in the prefrontal cortex in chronic Dopaminergic modulation of pain signals in the medial pattern analysis to determine that pain
pain. Pain 161, 1726–1729 (2020). prefrontal cortex: challenges and perspectives. representations were localized to the anterior
15. Apkarian, A. V., Baliki, M. N. & Farmer, M. A. Neurosci. Lett. 702, 71–76 (2019). MCC, negative emotion representations were more
Predicting transition to chronic pain. Curr. Opin. 36. Naser, P. V. & Kuner, R. Molecular, cellular and circuit common in the ventromedial PFC and cognitive
Neurol. 26, 360–367 (2013). basis of cholinergic modulation of pain. Neuroscience control representations were localized to parts of
16. Schulz, E. et al. Prefrontal gamma oscillations encode 387, 135–148 (2018). the dorsal MCC, thus suggesting domain-​specific
tonic pain in humans. Cereb. Cortex 25, 4407–4414 37. Metz, A. E., Yau, H. J., Centeno, M. V., Apkarian, A. V. representations for pain, negative emotion and
(2015). & Martina, M. Morphological and functional cognitive control in the mPFC.
17. Ploner, M., Sorg, C. & Gross, J. Brain rhythms of pain. reorganization of rat medial prefrontal cortex in 57. Shackman, A. J. et al. The integration of negative
Trends Cogn. Sci. 21, 100–110 (2017). neuropathic pain. Proc. Natl Acad. Sci. USA 106, affect, pain and cognitive control in the cingulate
18. Kim, J. A. & Davis, K. D. Neural oscillations: 2423–2428 (2009). cortex. Nat. Rev. Neurosci. 12, 154–167 (2011).
understanding a neural code of pain. Neuroscientist 38. Cordeiro Matos, S., Zamfir, M., Longo, G., 58. Strauss, I. et al. Double anterior stereotactic
https://doi.org/10.1177/1073858420958629 Ribeiro-​da-Silva, A. & Seguela, P. Noradrenergic fiber cingulotomy for intractable oncological pain.
(2020). sprouting and altered transduction in neuropathic Stereotact. Funct. Neurosurg. 95, 400–408 (2017).
19. Baliki, M. N. et al. Chronic pain and the emotional prefrontal cortex. Brain Struct. Funct. 223, 59. Fitzgerald, J. M. et al. DACC resting state functional
brain: specific brain activity associated with 1149–1164 (2018). connectivity as a predictor of pain symptoms following
spontaneous fluctuations of intensity of chronic back 39. Cordeiro Matos, S., Zhang, Z. & Seguela, P. Peripheral motor vehicle crash: a preliminary investigation.
pain. J. Neurosci. 26, 12165–12173 (2006). neuropathy induces HCN channel dysfunction in J. Pain 22, 171–179 (2020).
20. Hashmi, J. A. et al. Shape shifting pain: chronification pyramidal neurons of the medial prefrontal cortex. 60. Edes, A. E. et al. Increased activation of the pregenual
of back pain shifts brain representation from J. Neurosci. 35, 13244–13256 (2015). anterior cingulate cortex to citalopram challenge in
nociceptive to emotional circuits. Brain 136, This study reports that nerve injury leads to a migraine: an fMRI study. BMC Neurol. 19, 237
2751–2768 (2013). dysfunction of HCN channels in the dendrites of (2019).
21. May, E. S. et al. Prefrontal gamma oscillations reflect layer 2/3 pyramidal neurons in the mPFC, resulting 61. Icenhour, A. et al. Brain functional connectivity is
ongoing pain intensity in chronic back pain patients. in enhanced excitability and neuronal firing. An associated with visceral sensitivity in women with
Hum. Brain Mapp. 40, 293–305 (2019). altered cAMP–PKA axis was suggested to underlie irritable bowel syndrome. Neuroimage Clin. 15,
22. Ta Dinh, S. et al. Brain dysfunction in alterations to HCN channel function. 449–457 (2017).
chronic pain patients assessed by resting-​state 40. Shiers, S. et al. Neuropathic pain creates an enduring 62. Wang, G., Erpelding, N. & Davis, K. D. Sex differences
electroencephalography. Pain 160, 2751–2765 prefrontal cortex dysfunction corrected by the type II in connectivity of the subgenual anterior cingulate
(2019). diabetic drug metformin but not by gabapentin. cortex. Pain 155, 755–763 (2014).
23. Dale, J. et al. Scaling up cortical control inhibits pain. J. Neurosci. 38, 7337–7350 (2018). 63. Osborne, N. R. et al. Abnormal subgenual anterior
Cell Rep. 23, 1301–1313 (2018). 41. Baliki, M. N. et al. Corticostriatal functional cingulate circuitry is unique to women but not men
24. Thompson, J. M. & Neugebauer, V. Cortico-​limbic pain connectivity predicts transition to chronic back pain. with chronic pain. Pain 162, 97–108 (2021).
mechanisms. Neurosci. Lett. 702, 15–23 (2019). Nat. Neurosci. 15, 1117–1119 (2012). 64. Zhuo, M. Long-​term potentiation in the anterior
25. Wang, G. Q. et al. Deactivation of excitatory neurons 42. Lee, M. et al. Activation of corticostriatal circuitry cingulate cortex and chronic pain. Philos. Trans. R.
in the prelimbic cortex via Cdk5 promotes pain relieves chronic neuropathic pain. J. Neurosci. 35, Soc. Lond. B Biol. Sci. 369, 20130146 (2014).
sensation and anxiety. Nat. Commun. 6, 7660 (2015). 5247–5259 (2015). 65. Koga, K. et al. In vivo whole-​cell patch-​clamp recording
26. Jiang, Z. C. et al. Inactivation of the prelimbic rather 43. Martinez, E. et al. Corticostriatal regulation of acute of sensory synaptic responses of cingulate pyramidal
than infralimbic cortex impairs acquisition and pain. Front. Cell Neurosci. 11, 146 (2017). neurons to noxious mechanical stimuli in adult mice.
expression of formalin-​induced conditioned place 44. Zhou, H. et al. Inhibition of the prefrontal projection to Mol. Pain 6, 62 (2010).
avoidance. Neurosci. Lett. 569, 89–93 (2014). the nucleus accumbens enhances pain sensitivity and 66. Meda, K. S. et al. Microcircuit mechanisms through
27. Cheriyan, J. & Sheets, P. L. Altered excitability affect. Front. Cell Neurosci. 12, 240 (2018). which mediodorsal thalamic input to anterior cingulate
and local connectivity of mPFC-​PAG neurons in a 45. Coulombe, M. A., Erpelding, N., Kucyi, A. & Davis, K. D. cortex exacerbates pain-​related aversion. Neuron
mouse model of neuropathic pain. J. Neurosci. 38, Intrinsic functional connectivity of periaqueductal 102, 944–959.e3 (2019).
4829–4839 (2018). gray subregions in humans. Hum. Brain Mapp. 37, 67. Ning, L., Ma, L. Q., Wang, Z. R. & Wang, Y. W.
28. Fan, X. C. et al. Hypersensitivity of prelimbic cortex 1514–1530 (2016). Chronic constriction injury induced long-​term changes
neurons contributes to aggravated nociceptive 46. Ferreira, A. N., Yousuf, H., Dalton, S. & Sheets, P. L. in spontaneous membrane-​potential oscillations in
responses in rats with experience of chronic Highly differentiated cellular and circuit properties anterior cingulate cortical neurons in vivo. Pain
inflammatory pain. Front. Mol. Neurosci. 11, 85 of infralimbic pyramidal neurons projecting to the Physician 16, E577–E589 (2013).
(2018). periaqueductal gray and amygdala. Front. Cell 68. Li, X. H., Chen, Q. Y. & Zhuo, M. Neuronal adenylyl
29. Tan, L. L. et al. A pathway from midcingulate cortex Neurosci. 9, 161 (2015). cyclase targeting central plasticity for the treatment of
to posterior insula gates nociceptive hypersensitivity. 47. Yin, J. B. et al. dmPFC-​vlPAG projection neurons chronic pain. Neurotherapeutics 17, 861–873 (2020).
Nat. Neurosci. 20, 1591–1601 (2017). contribute to pain threshold maintenance and 69. Santello, M. & Nevian, T. Dysfunction of cortical
In this study, the authors reveal the importance of antianxiety behaviors. J. Clin. Invest. 130, dendritic integration in neuropathic pain reversed
an excitatory pathway from the MCC to the PI and 6555–6570 (2020). by serotoninergic neuromodulation. Neuron 86,
subsequently the raphe nucleus, which recruits This study further highlights the importance of 233–246 (2015).
descending serotonergic facilitatory projections to prefrontal cortical influences on descending pain This study uses elegant electrophysiological
the spinal cord to influence peripheral nociceptive modulatory systems in pain and their dysregulation techniques to demonstrate that HCN channel
responses. in neuropathic pain, reporting an excitatory dysfunction in cortical output neurons in layer 5
30. Mitric, M. et al. Layer- and subregion-​specific pathway from the dorsomedial PFC to the of the PFC in neuropathic pain can be reversed
electrophysiological and morphological changes ventrolateral PAG, which, upon activation, reduces by serotonergic modulation.
of the medial prefrontal cortex in a mouse model of nociception as well as anxiety-​related behaviours 70. Kang, S. J. et al. Bidirectional modulation of
neuropathic pain. Sci. Rep. 9, 9479 (2019). in neuropathic mice. hyperalgesia via the specific control of excitatory and
31. Zhang, Z. et al. Role of prelimbic GABAergic circuits 48. Vachon-​Presseau, E. et al. Corticolimbic anatomical inhibitory neuronal activity in the ACC. Mol. Brain 8,
in sensory and emotional aspects of neuropathic pain. characteristics predetermine risk for chronic pain. 81 (2015).
Cell Rep. 12, 752–759 (2015). Brain 139, 1958–1970 (2016). 71. Barthas, F. et al. The anterior cingulate cortex
32. Kelly, C. J., Huang, M., Meltzer, H. & Martina, M. 49. Kiritoshi, T. & Neugebauer, V. Pathway-​specific is a critical hub for pain-​induced depression.
Reduced glutamatergic currents and dendritic alterations of cortico-​amygdala transmission in Biol. Psychiatry 77, 236–245 (2015).

NAture RevIews | NeurOSCienCe volume 22 | August 2021 | 469

0123456789();:
Reviews

72. Sellmeijer, J. et al. Hyperactivity of anterior cingulate 95. Cichon, J., Blanck, T. J. J., Gan, W. B. & Yang, G. 118. Yalcin, I. & Barrot, M. The anxiodepressive
cortex areas 24a/24b drives chronic pain-​induced Activation of cortical somatostatin interneurons comorbidity in chronic pain. Curr. Opin. Anaesthesiol.
anxiodepressive-​like consequences. J. Neurosci. 38, prevents the development of neuropathic pain. 27, 520–527 (2014).
3102–3115 (2018). Nat. Neurosci. 20, 1122–1132 (2017). 119. Lee, M. C., Wanigasekera, V. & Tracey, I. Imaging
73. Zhou, H. et al. Ketamine reduces aversion in rodent pain 96. Andoh, J. et al. Neural correlates of evoked phantom opioid analgesia in the human brain and its potential
models by suppressing hyperactivity of the anterior limb sensations. Biol. Psychol. 126, 89–97 (2017). relevance for understanding opioid use in chronic pain.
cingulate cortex. Nat. Commun. 9, 3751 (2018). 97. Jin, Y. et al. A somatosensory cortex input to the Neuropharmacology 84, 123–130 (2014).
74. Yao, P. W. et al. Upregulation of tumor necrosis factor-​ caudal dorsolateral striatum controls comorbid 120. Wang, D. et al. Functional divergence of delta and mu
alpha in the anterior cingulate cortex contributes to anxiety in persistent pain. Pain 161, 416–428 opioid receptor organization in CNS pain circuits.
neuropathic pain and pain-​associated aversion. (2020). Neuron 98, 90–108.e5 (2018).
Neurobiol. Dis. 130, 104456 (2019). 98. Singh, A. et al. Mapping cortical integration of 121. Birdsong, W. T. et al. Synapse-​specific opioid
75. Zhang, Q. et al. Chronic pain induces generalized sensory and affective pain pathways. Curr. Biol. 30, modulation of thalamo-​cortico-striatal circuits. eLife 8,
enhancement of aversion. eLife 6, e25302 (2017). 1703–1715.e5 (2020). e45146 (2019).
76. Gao, S. H. et al. The projections from the anterior 99. Liu, Y. et al. Touch and tactile neuropathic pain 122. Wu, W. Y., Liu, C. Y., Tsai, M. L. & Yen, C. T. Nocifensive
cingulate cortex to the nucleus accumbens and ventral sensitivity are set by corticospinal projections. Nature behavior-​related laser heat-​evoked component in
tegmental area contribute to neuropathic pain-​evoked 561, 547–550 (2018). the rostral agranular insular cortex revealed using
aversion in rats. Neurobiol. Dis. 140, 104862 (2020). 100. Abraira, V. E. et al. The cellular and synaptic morphine analgesia. Physiol. Behav. 154, 129–134
77. Chen, T. et al. Top-​down descending facilitation of spinal architecture of the mechanosensory dorsal horn. Cell (2016).
sensory excitatory transmission from the anterior 168, 295–310.e19 (2017). 123. Peng, K. et al. Morphine attenuates fNIRS signal
cingulate cortex. Nat. Commun. 9, 1886 (2018). 101. Mazzola, L., Isnard, J. & Mauguière, F. Somatosensory associated with painful stimuli in the medial
78. Vogt, B. A. Pain and emotion interactions in and pain responses to stimulation of the second frontopolar cortex (medial BA 10). Front. Hum.
subregions of the cingulate gyrus. Nat. Rev. Neurosci. somatosensory area (SII) in humans. A comparison Neurosci. 12, 394 (2018).
6, 533–544 (2005). with SI and insular responses. Cereb. Cortex 16, 124. Oertel, B. G. et al. Differential opioid action on
This review provides an excellent overview of 960–968 (2006). sensory and affective cerebral pain processing.
the importance of cingulate organization and the 102. Liang, M., Mouraux, A. & Iannetti, G. D. Parallel Clin. Pharmacol. Ther. 83, 577–588 (2008).
implications of specific cingulate subregions processing of nociceptive and non-​nociceptive 125. Navratilova, E. et al. Endogenous opioid activity in the
in the processing of emotion and nociceptive somatosensory information in the human primary anterior cingulate cortex is required for relief of pain.
information. and secondary somatosensory cortices: evidence J. Neurosci. 35, 7264–7271 (2015).
79. Almarzouki, A. F., Brown, C. A., Brown, R. J., from dynamic causal modeling of functional magnetic This study reports that endogenous opioidergic
Leung, M. H. K. & Jones, A. K. P. Negative resonance imaging data. J. Neurosci. 31, 8976–8985 signalling in the ACC is required to activate
expectations interfere with the analgesic effect of (2011). dopaminergic signalling in the NAc, which underlies
safety cues on pain perception by priming the cortical 103. Garcia-​Larrea, L. & Peyron, R. Pain matrices and reward associated with pain relief, and that non-​
representation of pain in the midcingulate cortex. neuropathic pain matrices: a review. Pain 154, opioidergic pain-​relieving treatments also engage
PLoS ONE 12, e0180006 (2017). S29–S43 (2013). opioidergic circuits in the ACC to achieve reduction
80. Fillinger, C., Yalcin, I., Barrot, M. & Veinante, P. 104. Inami, C. et al. Visualization of brain activity in a in pain affect.
Afferents to anterior cingulate areas 24a and 24b neuropathic pain model using quantitative activity-​ 126. Gomtsian, L. et al. Morphine effects within the rodent
and midcingulate areas 24a’ and 24b’ in the mouse. dependent manganese magnetic resonance imaging. anterior cingulate cortex and rostral ventromedial
Brain Struct. Funct. 222, 1509–1532 (2017). Front. Neural Circuits 13, 74 (2019). medulla reveal separable modulation of affective and
81. Hu, T. T. et al. Activation of the intrinsic pain inhibitory 105. Zhu, X. et al. A central amygdala input to the sensory qualities of acute or chronic pain. Pain 159,
circuit from the midcingulate Cg2 to zona incerta parafascicular nucleus controls comorbid pain in 2512–2521 (2018).
alleviates neuropathic pain. J. Neurosci. 39, depression. Cell Rep. 29, 3847–3858.e5 (2019). 127. Navratilova, E. et al. Selective modulation of tonic
9130–9144 (2019). 106. Gogolla, N. The insular cortex. Curr. Biol. 27, aversive qualities of neuropathic pain by morphine
82. Kim, J. et al. Somatotopically specific primary R580–R586 (2017). in the central nucleus of the amygdala requires
somatosensory connectivity to salience and default 107. Garcia-​Larrea, L. The posterior insular-​opercular endogenous opioid signaling in the anterior cingulate
mode networks encodes clinical pain. Pain 160, region and the search of a primary cortex for pain. cortex. Pain 161, 609–618 (2020).
1594–1605 (2019). Neurophysiol. Clin. 42, 299–313 (2012). 128. Bannister, K. et al. Multiple sites and actions of
83. Tan, L. L. et al. Gamma oscillations in somatosensory 108. Gehrlach, D. A. et al. Aversive state processing in gabapentin-​induced relief of ongoing experimental
cortex recruit prefrontal and descending serotonergic the posterior insular cortex. Nat. Neurosci. 22, neuropathic pain. Pain 158, 2386–2395 (2017).
pathways in aversion and nociception. Nat. Commun. 1424–1437 (2019). 129. Zhao, M., Wang, J.-Y., Jia, H. & Tang, J.-S. µ- but not
10, 983 (2019). In this study, the authors provide comprehensive δ- and κ-​opioid receptors in the ventrolateral orbital
84. Yang, P. F., Wu, R., Wu, T. L., Shi, Z. & Chen, L. M. data to reveal the importance of the PI and its cortex mediate opioid-​induced antiallodynia in a rat
Discrete modules and mesoscale functional circuits associated circuitries in the detection of aversive neuropathic pain model. Brain Res. 1076, 68–77
for thermal nociception within primate S1 cortex. internal states and triggering of behavioural (2006).
J. Neurosci. 38, 1774–1787 (2018). adaptations critical for survival in mice. 130. Lee, M. C. et al. Amygdala activity contributes to the
85. Zhang, Z. G., Hu, L., Hung, Y. S., Mouraux, A. & 109. Mazzola, L., Mauguiere, F. & Isnard, J. Functional dissociative effect of cannabis on pain perception. Pain
Iannetti, G. D. Gamma-​band oscillations in the primary mapping of the human insula: data from electrical 154, 124–134 (2013).
somatosensory cortex—a direct and obligatory stimulations. Rev. Neurol. 175, 150–156 (2019). 131. Weizman, L. et al. Cannabis analgesia in chronic
correlate of subjective pain intensity. J. Neurosci. 32, 110. Liberati, G. et al. Tonic thermonociceptive stimulation neuropathic pain is associated with altered brain
7429–7438 (2012). selectively modulates ongoing neural oscillations connectivity. Neurology 91, e1285–e1294 (2018).
86. Heid, C. et al. Early gamma-​oscillations as correlate in the human posterior insula: evidence from 132. Kim, J., Farchione, T., Potter, A., Chen, Q. & Temple, R.
of localized nociceptive processing in primary intracerebral EEG. Neuroimage 188, 70–83 (2019). Esketamine for treatment-​resistant depression - first
sensorimotor cortex. J. Neurophysiol. 123, 111. Liberati, G. et al. Gamma-​band oscillations FDA-​approved antidepressant in a new class. N. Engl.
1711–1726 (2020). preferential for nociception can be recorded in the J. Med. 381, 1–4 (2019).
87. Gross, J., Schnitzler, A., Timmermann, L. & Ploner, M. human insula. Cereb. Cortex 28, 3650–3664 (2018). 133. Pan, W. et al. Ketamine differentially restores diverse
Gamma oscillations in human primary somatosensory 112. Frot, M., Faillenot, I. & Mauguiere, F. Processing of alterations of neuroligins in brain regions in a rat
cortex reflect pain perception. PLoS Biol. 5, e133 nociceptive input from posterior to anterior insula in model of neuropathic pain-​induced depression.
(2007). humans. Hum. Brain Mapp. 35, 5486–5499 (2014). Neuroreport 29, 863–869 (2018).
88. Misra, G., Ofori, E., Chung, J. W. & Coombes, S. A. 113. Bastuji, H., Frot, M., Perchet, C., Hagiwara, K. 134. Tappe-​Theodor, A. & Kuner, R. A common ground for
Pain-​related suppression of beta oscillations facilitates & Garcia-​Larrea, L. Convergence of sensory and pain and depression. Nat. Neurosci. 22, 1612–1614
voluntary movement. Cereb. Cortex 27, 2592–2606 limbic noxious input into the anterior insula and the (2019).
(2017). emergence of pain from nociception. Sci. Rep. 8, 135. Hirschberg, S., Li, Y., Randall, A., Kremer, E. J. &
89. Ploner, M., Gross, J., Timmermann, L., Pollok, B. & 13360 (2018). Pickering, A. E. Functional dichotomy in spinal- vs
Schnitzler, A. Pain suppresses spontaneous brain 114. Li, X. et al. The reorganization of insular subregions in prefrontal-​projecting locus coeruleus modules splits
rhythms. Cereb. Cortex 16, 537–540 (2006). individuals with below-​level neuropathic pain following descending noradrenergic analgesia from ascending
90. Hu, L. & Iannetti, G. D. Neural indicators of perceptual incomplete spinal cord injury. Neural Plast. 2020, aversion and anxiety in rats. eLife 6, e29808 (2017).
variability of pain across species. Proc. Natl Acad. 2796571 (2020). 136. Lefaucheur, J. P. et al. Evidence-​based guidelines
Sci. USA 116, 1782–1791 (2019). 115. Zhuo, M. Cortical LTP: a synaptic model for on the therapeutic use of repetitive transcranial
91. Yue, L. P., Iannetti, G. D. & Hu, L. The neural origin chronic pain. Adv. Exp. Med. Biol. 1099, 147–155 magnetic stimulation (rTMS): an update (2014-2018).
of nociceptive-​induced gamma band oscillations. (2018). Clin. Neurophysiol. 131, 474–528 (2020).
J. Neurosci. 40, 2478–3490 (2020). 116. Miao, H. H., Li, X. H., Chen, Q. Y. & Zhuo, M. 137. Lamusuo, S. et al. Neurotransmitters behind pain
92. Wang, J., Wang, J., Xing, G. G., Li, X. & Wan, Y. Calcium-​stimulated adenylyl cyclase subtype 1 is relief with transcranial magnetic stimulation —
Enhanced gamma oscillatory activity in rats with required for presynaptic long-​term potentiation positron emission tomography evidence for release
chronic inflammatory pain. Front. Neurosci. 10, 489 in the insular cortex of adult mice. Mol. Pain 15, of endogenous opioids. Eur. J. Pain 21, 1505–1515
(2016). 1744806919842961 (2019). (2017).
93. Uhelski, M. L., Davis, M. A. & Fuchs, P. N. Pain 117. Corder, G. et al. An amygdalar neural ensemble that 138. Borckardt, J. J. et al. Prefrontal versus motor cortex
affect in the absence of pain sensation: evidence of encodes the unpleasantness of pain. Science 363, transcranial direct current stimulation (tDCS) effects
asomaesthesia after somatosensory cortex lesions 276–281 (2019). on post-​surgical opioid use. Brain Stimul. 10,
in the rat. Pain 153, 885–892 (2012). This study uses time-​lapse in vivo calcium imaging 1096–1101 (2017).
94. Eto, K. et al. Inter-​regional contribution of enhanced in freely behaving mice to identify a distinct neural 139. Taylor, J. J., Borckardt, J. J. & George, M. S.
activity of the primary somatosensory cortex to the ensemble in the BLA that encodes the negative Endogenous opioids mediate left dorsolateral
anterior cingulate cortex accelerates chronic pain affective valence of pain and drives behavioural prefrontal cortex rTMS-​induced analgesia. Pain 153,
behavior. J. Neurosci. 31, 7631–7636 (2011). avoidance responses to noxious stimuli. 1219–1225 (2012).

470 | August 2021 | volume 22 www.nature.com/nrn

0123456789();:
Reviews

140. Arendsen, L. J., Hugh-​Jones, S. & Lloyd, D. M. resonance imaging data. JAMA Neurol. 75, 165. Doan, L., Manders, T. & Wang, J. Neuroplasticity
Transcranial alternating current stimulation at alpha 1321–1330 (2018). underlying the comorbidity of pain and depression.
frequency reduces pain when the intensity of pain is 153. Bingel, U., Lorenz, J., Schoell, E., Weiller, C. & Neural Plast. 2015, 504691 (2015).
uncertain. J. Pain 19, 807–818 (2018). Büchel, C. Mechanisms of placebo analgesia: rACC 166. Nees, F. & Becker, S. Psychological processes in
141. Ahn, S., Prim, J. H., Alexander, M. L., McCulloch, K. L. recruitment of a subcortical antinociceptive network. chronic pain: influences of reward and fear learning
& Fröhlich, F. Identifying and engaging neuronal Pain 120, 8–15 (2006). as key mechanisms — behavioral evidence, neural
oscillations by transcranial alternating current 154. Petrovic, P. et al. A prefrontal non-​opioid mechanism circuits, and maladaptive changes. Neuroscience 387,
stimulation in patients with chronic low back pain: in placebo analgesia. Pain 150, 59–65 (2010). 72–84 (2018).
a randomized, crossover, double-​blind, sham-​controlled 155. Zeng, Y. et al. A voxel-​based analysis of 167. Cai, Y. Q., Wang, W., Paulucci-​Holthauzen, A. &
pilot study. J. Pain 20, 277.e1–277.e11 (2019). neurobiological mechanisms in placebo analgesia in Pan, Z. Z. Brain circuits mediating opposing effects
142. Pagano, R. L. et al. Motor cortex stimulation inhibits rats. Neuroimage 178, 602–612 (2018). on emotion and pain. J. Neurosci. 38, 6340–6349
thalamic sensory neurons and enhances activity of 156. Kong, J. et al. Functional connectivity of the (2018).
PAG neurons: possible pathways for antinociception. frontoparietal network predicts cognitive modulation 168. Zhou, W. et al. A neural circuit for comorbid
Pain 153, 2359–2369 (2012). of pain. Pain 154, 459–467 (2013). depressive symptoms in chronic pain. Nat. Neurosci.
143. Youssef, A. M., Macefield, V. G. & Henderson, L. A. 157. Woo, C. W., Roy, M., Buhle, J. T. & Wager, T. D. Distinct 22, 1649–1658 (2019).
Pain inhibits pain; human brainstem mechanisms. brain systems mediate the effects of nociceptive input
Neuroimage 124, 54–62 (2016). and self-​regulation on pain. PLoS Biol. 13, e1002036 Acknowledgements
144. McPhee, M. E. & Graven-​Nielsen, T. Recurrent (2015). The authors thank all members of their laboratory as well as
low back pain patients demonstrate facilitated 158. Segerdahl, A. R., Mezue, M., Okell, T. W., Farrar, J. T. scientists in the Heidelberg Pain Consortium (CRC1158 of the
pronociceptive mechanisms when in pain, and & Tracey, I. The dorsal posterior insula subserves a Deutsche Forschungsgemeinschaft) for thought-​provoking
impaired antinociceptive mechanisms with fundamental role in human pain. Nat. Neurosci. 18, discussions and expert viewpoints. The authors acknowledge
and without pain. Pain 160, 2866–2876 (2019). 499–500 (2015). funding to R.K. from the Deutsche Forschungsgemeinschaft
145. Moont, R., Crispel, Y., Lev, R., Pud, D. & Yarnitsky, D. This study uses an innovative approach to pinpoint in the form of grants for Collaborative Research Centre 1158
Temporal changes in cortical activation during the dorsal PI as being fundamental to nociception (projects B01 and B06) and from the Baden-​Württemberg
conditioned pain modulation (CPM), a LORETA study. and pain. Foundation (Internationale Spitzenforschung; BWST-​
Pain 152, 1469–1477 (2011). 159. Wiech, K., Jbabdi, S., Lin, C. S., Andersson, J. & ISF2017-069). L.L.T. was partially supported by a postdoc-
146. Youssef, A. M., Macefield, V. G. & Henderson, L. A. Tracey, I. Differential structural and resting state toral fellowship from the European Molecular Biology
Cortical influences on brainstem circuitry responsible connectivity between insular subdivisions and other Organization. R.K. is a member of the Molecular Medicine
for conditioned pain modulation in humans. Hum. pain-​related brain regions. Pain 155, 2047–2055 Partnership Unit in the Medical Faculty in Heidelberg and the
Brain Mapp. 37, 2630–2644 (2016). (2014). European Molecular Biology Laboratory.
147. Zeidan, F. & Vago, D. R. Mindfulness meditation-​based 160. Lieberman, M. D. & Eisenberger, N. I. The dorsal
pain relief: a mechanistic account. Ann. N. Y. Acad. anterior cingulate cortex is selective for pain: results
Author contributions
Sci. 1373, 114–127 (2016). from large-​scale reverse inference. Proc. Natl Acad.
Both authors contributed equally to all aspects of this work.
148. Zeidan, F. et al. Mindfulness-​meditation-based Sci. USA 112, 15250–15255 (2015).
pain relief is not mediated by endogenous opioids. 161. Davis, K. D., Bushnell, M. C., St Iannetti, G. D.,
J. Neurosci. 36, 3391–3397 (2016). Lawrence, K. & Coghill, R. Evidence against pain Competing interests
149. Eippert, F. et al. Activation of the opioidergic specificity in the dorsal posterior insula. F1000Res 4, The authors declare no competing interests.
descending pain control system underlies placebo 362 (2015).
analgesia. Neuron 63, 533–543 (2009). 162. Carrillo-​Reid, L. & Yuste, R. Playing the piano with Publisher’s note
150. Zeidan, F. et al. Mindfulness meditation-​based pain the cortex: role of neuronal ensembles and pattern Springer Nature remains neutral with regard to jurisdictional
relief employs different neural mechanisms than completion in perception and behavior. Curr. Opin. claims in published maps and institutional affiliations.
placebo and sham mindfulness meditation-​induced Neurobiol. 64, 89–95 (2020).
analgesia. J. Neurosci. 35, 15307–15325 (2015). 163. Buzsaki, G. Neural syntax: cell assemblies, Reviewer information
151. Lui, F. et al. Neural bases of conditioned placebo synapsembles, and readers. Neuron 68, 362–385 Nature Reviews Neuroscience thanks Elizabeth May, who
analgesia. Pain 151, 816–824 (2010). (2010). co-​reviewed with Markus Ploner; Patrick Sheets; and Gregory
152. Zunhammer, M., Bingel, U. & Wager, T. D. Placebo 164. Buzsaki, G. & Draguhn, A. Neuronal oscillations Scherrer for their contribution to the peer review of this work.
effects on the neurologic pain signature: a meta-​ in cortical networks. Science 304, 1926–1929
analysis of individual participant functional magnetic (2004). © Springer Nature Limited 2021

NAture RevIews | NeurOSCienCe volume 22 | August 2021 | 471

0123456789();:

You might also like