Next Article in Journal
Effect of Single High Dose Vitamin D Substitution in Hospitalized COVID-19 Patients with Vitamin D Deficiency on Length of Hospital Stay
Previous Article in Journal
RNA Therapeutics: A Healthcare Paradigm Shift
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Dopaminergic Modulation of Prefrontal Cortex Inhibition

Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(5), 1276; https://doi.org/10.3390/biomedicines11051276
Submission received: 31 March 2023 / Revised: 21 April 2023 / Accepted: 23 April 2023 / Published: 25 April 2023
(This article belongs to the Special Issue Synaptic Transmission: From Molecular to Neural Network Levels 2.0)

Abstract

:
The prefrontal cortex is the highest stage of integration in the mammalian brain. Its functions vary greatly, from working memory to decision-making, and are primarily related to higher cognitive functions. This explains the considerable effort devoted to investigating this area, revealing the complex molecular, cellular, and network organization, and the essential role of various regulatory controls. In particular, the dopaminergic modulation and the impact of local interneurons activity are critical for prefrontal cortex functioning, controlling the excitatory/inhibitory balance and the overall network processing. Though often studied separately, the dopaminergic and GABAergic systems are deeply intertwined in influencing prefrontal network processing. This mini review will focus on the dopaminergic modulation of GABAergic inhibition, which plays a significant role in shaping prefrontal cortex activity.

1. The Prefrontal Cortex

The prefrontal cortex (PFC) is thought to be the highest association area in the mammalian cortex and is required for proper executive control. Task flexibility and planning [1], selective attention, attentional set-shifting, rule learning, strategy switching, and goal-directed behavior [2,3,4] are just some of the many PFC functions. This considered, it is not surprising that PFC alterations have been associated with a variety of psychiatric conditions. For example, several investigations reported PFC-related impaired working memory [5,6,7,8] and altered network oscillations [9,10] in schizophrenia. Though rodent PFC is less complex than that of primates, it exerts similar functions in the executive domain [11]. For this reason, the rodent represents a valuable model to investigate how PFC functions are determined at the molecular, cellular, and network levels. However, investigations in rodents are complicated by the lack of a univocal and unambiguous nomenclature of PFC subdivisions. Due to its recent evolution and inter-species variability, it is challenging to identify proper structural and functional criteria to define PFC regions [12,13]. This has been the subject of many studies aiming at characterizing differences and similarities of mammalian PFC [14]. Ref [15] introduced a hodological criterium based on the assumption that the mediodorsal thalamic nucleus (MD) is the primary site of projections toward the PFC. Therefore, according to this definition, the mammalian PFC could be identified based on the connectivity with the MD. Following this perspective, the effective existence in rats of two prefrontal cortex areas receiving projections from the MD, indicated as medial and orbitofrontal, was demonstrated [16]. Clearly, this definition bears some limitations. Indeed, other criteria were then adopted. For example, other researchers proposed a cytoarchitectural criterion, though this method was deemed valid only for closely related species [12]. To date, the best way to define PFC parcellation is proposed to be a combination of four criteria: function, architecture, connectivity, and topography [17,18]. In particular, the relevance of the connectivity aspect grew over time. Recent works have described the organization of cortical interconnectivity into modules along the whole brain [18,19] and identified a prefrontal cortical module. The areas within the prefrontal module show dense interconnections [20,21] and are believed to be devoted to similar functions [22]. The regions recognized as a component of the prefrontal module are the prelimbic area, the infralimbic area, the anterior cingulate area, the frontal pole cerebral cortex, and the orbital areas. Another widely used distinction, mainly based on connectivity mapping including thalamocortical, corticothalamic, corticostriatal, and corticocortical projections, recognizes three broad PFC subdivisions: the dorsomedial PFC (dmPFC), ventromedial PFC (vmPFC), and ventrolateral PFC (vlPFC). Considering the complex scenario of rodent PFC nomenclature and the absence of a standard reference for the different studies available in the literature, it is not surprising that many studies focusing on the PFC report vague indications of the subregion actually subjected to analysis. In particular, most investigations on the highest-level cognitive functioning in rodents target the so-called medial PFC (mPFC), comprising the infralimbic, prelimbic, and anterior cingulate areas [2,13]. It is worth specifying that there is no direct anatomical equivalence between human and rodent PFC. However, the rodent mPFC is anatomically located in correspondence with the anterior cingulate cortex (ACC) in humans (see [13] for a detailed review of the comparison between rodent and human PFC). Here, we will mainly refer to rodent reports on the mPFC, which is the most commonly addressed PFC area. The cytoarchitecture and the connectivity patterns are similar in rodents and humans, with the significant difference represented by the lack of the granular layer (layer IV) in rodent PFC. In both cases, the PFC is mainly composed of pyramidal neurons (PN, 80–90%) and inhibitory interneurons (IN, 10–20%) [23]. The main excitatory output is provided by the PNs, which are strongly interconnected to form a local network that projects to other cortical and subcortical areas. PN activity is modulated by a strong network of GABAergic INs [24,25], which proved to be essential for controlling PN firing and generating neuronal network oscillations [26,27,28]. The interplay between PNs and INs modulates PFC activity and is crucial to maintain proper cognitive functions.

2. Dopamine Receptors in the PFC

Dopamine (DA) is released in the mPFC by projections originating from the midbrain nuclei of the ventral tegmental area (VTA) and substantia nigra pars compacta [29,30]. Once released, DA interacts with five different receptors subtypes (D1, D2, D3, D4, D5) subdivided into two families: D1-like receptors comprising D1 and D5, and D2-like receptors comprising D2, D3, and D4 [29,31,32]. Receptors belonging to the D1-like family are more abundant than those of the D2-like family and are expressed in all PFC layers. On the other hand, receptors of the D2-like family are primarily expressed in deeper layers (mainly layer V) [33], and their affinity is 10–100 times higher than that of D1-like receptors [34]. Both DA receptor families are expressed on pyramidal and non-pyramidal neurons, thus modulating excitation and inhibition [29,33]. Finally, these two receptor classes differ in the intracellular signaling pathway mediating their effects. Since DA receptors are G-protein coupled receptors (GPCRs), they all activate heteromeric G-proteins, but the second messenger and the effector proteins activated are usually different for different receptors and, in most cases, mediate opposite responses.
In particular, D1-like receptors activation is coupled with the G-proteins Gαs and Gαolf which, in turn, are associated with adenylyl cyclase (AC) that, once activated, increases the level of cyclic adenosine monophosphate (cAMP) leading to the activation of protein kinase A (PKA). PKA modulates most D1-like functions by phosphorylating many substrates including voltage-gated K+, Na+, and Ca2+ channels, GABA receptors, and NMDA receptors [32,35]. One of the main PKA targets is the DA and cAMP-regulated phosphoprotein DARPP-32, which is crucial in regulating downstream signaling pathways. When phosphorylated, DARPP-32 inhibits the protein phosphatase 1 (PP1) that opposes PKA action, eventually amplifying PKA signaling. On the other hand, the activation of D2-like receptors leads to the opposite effect. When activated, these receptors couple with Gαi and Gαo that inhibit the activation of AC, thus limiting PKA signaling. Moreover, the activation of D2-like receptors determines the activation of the calmodulin-dependent protein phosphatase (PP2B), which turns DARPP-32 into a strong inhibitor of PKA signaling [32]. Thus, DARPP-32 can bidirectionally modulate PKA activity. Besides their regulation through PKA pathways, ion channels can also be modulated directly via binding the Gβγ subunit or indirectly via activation of the phospholipase C (PLC) by both D1-like and D2-like receptors (Figure 1). The latter is most common for modulating Ca2+ conductance, determining a decrease in CaV2.2 (N-type) and CaV1 (L-type) currents. PLC can also be activated through coupling with Gαq, though limited to cells expressing D5 and D1/D2 heterodimers [36,37]. Lastly, D1-like and D2-like receptors can modulate NMDA and GABA receptors through direct protein–protein interactions or PKA/IP3 signaling [35]. The mechanism by which D2-like receptors, particularly D4, regulate GABA receptors involves a pathway comprising the dephosphorylation of cofilin (an actin depolymerizing factor) via PP1 activation. This leads to the loss of actin stability, with a consequent interruption of myosin motor-mediated transport of GABA receptor-containing vesicles in the membrane, resulting in a reduced GABA receptor-mediated current [38].

3. Dopamine Modulation of GABAergic Inhibition

3.1. On Pyramidal Neurons (PN)

As nicely reviewed by [29], the net effect of DA release onto the PFC also depends on cell type, synaptic properties, and interactions with other neurotransmitters. One of the critical DA roles in the PFC is the modulation of the GABAergic system. This modulation contributes to setting the proper excitation/inhibition (E/I) balance in the PFC, which requires fine-tuning to ensure correct network activity. Indeed, the E/I ratio is disrupted in a broad range of psychiatric disorders [39,40,41]. Many studies focused on the role of D4 receptors in preserving the correct E/I balance. D4 receptors are enriched in the PFC and are usually expressed in dendritic processes [42,43,44], while D1 receptors are most prominent at PN dendritic spines [45]. In particular, D4 receptors are mainly expressed nearby GABAA receptors in PFC PNs [46]. Experimental evidence showed that D2/D4 receptor agonists decrease the inhibitory post-synaptic currents (IPSCs) of layer V PN in rodent PFC, while a D1 receptor agonist increases IPSCs amplitude in the same neurons [46,47,48]. When D1- and D2-like receptors activation combines, an initial downregulation of the IPSCs mediated by D2-like receptors is followed by a D1-like receptors-dependent IPSCs increase. This suggests the biphasic nature of DA modulation of GABAergic responses in PFC PNs [29,47].
DA is reported to regulate inhibition through different intracellular mechanisms. In particular, high DA concentrations increase spontaneous inhibitory postsynaptic potentials (sIPSP) in PFC layer II/III [49] and layer V-VI PNs [50], revealing DA-mediated enhancement of GABA release. On the other hand, DA can depress evoked IPSP (eIPSP) in layer V-VI PNs [47,51,52]. This evidence shows that DA can modulate spontaneous and evoked IPSPs affecting GABA release mechanisms, hence regulating the presynaptic machinery [29]. This effect was also described in IN-PN pair recordings [53]. A possible explanation of the different DA impact on spontaneous and evoked IPSCs is proposed by [29]. The authors highlighted that the eIPSCs derive from activating a specific fiber through electrical stimulation, while sIPSCs derive from multiple diverse inputs. Therefore, the effect of DA on IPSCs may depend on the neuronal type generating the IPSC and the different neurons originating the GABAergic terminals impinging on that same neuron [29]. The heterogeneity of DA modulation reported in different studies might also depend on the recording sites. Indeed, D1- and D2-like receptors have different expression patterns: while D1-like receptors mRNA are also expressed in superficial layers, D2-like receptors are restricted to deeper layers such as layer V [33] (Figure 2).

3.2. On Inhibitory Interneurons (IN)

DA receptors are expressed in a wide array of GABAergic interneurons and, therefore, DA release onto the PFC affects IN activity, too [33,54,55]. DA is known to induce an increase in intrinsic excitability favoring depolarization in fast-spiking interneurons (FS) via a D1-like receptor-dependent mechanism [56,57]. Moreover, the effect of D1-like and D2-like receptors on PFC GABAergic INs may differ on a temporal scale. The activation of D1-like receptors induces both a depolarization and an increase in the neuronal excitability of FS. Different mechanisms mediate these two effects. The DA-induced depolarization lasts less than the increased excitability, meaning that DA can act through the same receptors to modulate different ionic currents at different time scales [56]. Interestingly, the activation of D2-like receptors at the peak of D1-like mediated IPSC determines a decrease in the IPSC amplitude [47,56]. Consistent with the biphasic hypothesis of DA modulation of the GABAergic system, D2-like receptors mediate a reduction in inhibition, and D1-like receptors mediate an increase in inhibition on PFC PNs, influencing IN activity (Figure 2). Lastly, D1-like receptors in superficial layers are often associated with vasoactive intestinal peptide (VIP) GABAergic INs and inhibit deeper INs via internal loops and interactions [58]. This supports the D1-like receptor role in determining circuit disinhibition, which is fundamental to appropriately modulating the PFC range of activity.

3.3. Evidence In Vivo

Several studies showed that DA exerts a predominantly inhibitory effect on PFC PN in vivo, primarily suppressing spontaneous firing [59,60,61]. Importantly, microdialysis data in vivo revealed a tonic level of DA in the PFC [62,63]. Most studies reported here were performed on anesthetized animals, where little VTA activity is presumably present at rest. Nevertheless, the stimulation of fiber bundles at the medial forebrain, or direct VTA stimulation, effectively increased DA levels in the PFC. It should also be considered that the absence of not experimentally evoked DA release is an advantage in characterizing transient DA effects on PFC neurons. For these reasons, these studies are considered suitable to address the consequences of DA release on the PFC in vivo. Indeed, VTA stimulation induces a fast EPSP-IPSP sequence in PFC PNs, with the IPSP consistent with GABAA receptors activation [60]. Interestingly, the inhibitory component is eliminated not only by GABAA receptor antagonists [64] but also by D2-like receptor antagonists, which tonically inhibit neuronal excitability [65,66,67]. When the D2-like receptor tone is abolished, the entire network physiology changes: neurons increase their firing, and the inhibition produced by VTA stimulation is occluded [29]. Overall, these studies show that DA released from dopaminergic terminals in the PFC, as well as exogenous DA, modulates spontaneous firing in vivo through complex mechanisms depending on the endogenous DA tone, the amount of DA released, and the activated receptor subtype. This effect was also confirmed by a computational model in which increasing DA concentrations elicited the facilitation of FS activity, with consequent suppression of pyramidal neurons firing. Moreover, enhancing basal DA levels rescues the initial condition, through the downregulation of the GABAergic tone, with consequent hyperactivity of PN firing [68]. Interestingly, computational models primarily based on in vivo studies have proposed a dual mechanism by which D1-like receptors can modulate working memory. First, the spontaneous activity of PN is decreased by upregulating inhibitory GABA currents; then, high-activity states are induced by upregulating excitatory NMDA currents [69,70]. This effect is believed to be mediated by D1-like receptors, which might induce inhibition by amplifying IPSCs in PNs [71], or an excitatory effect by enhancing NMDA receptor-mediated responses [72,73]. The same computational model was also used to implement D2-like receptors modulation of PFC activity. It was proposed that D2-like receptors activation decreases inhibitory currents in PNs while increasing IN excitability to maintain E/I balance [74].
Taken together, these findings provide evidence for a delicate homeostatic interplay between dopaminergic and GABAergic systems necessary to maintain PFC network stability and output selectivity.

3.4. Comments on PFC Regional Specificity

As pointed out in the first section, the PFC can be subdivided into several regions. It might then be of interest to consider whether a regional specificity has been observed in dopaminergic and GABAergic interplay. However, the intricate PFC subregions identification and nomenclature complicate the picture. Considering the literature reviewed here and mentioning to whatever extent the interaction between dopaminergic and GABAergic systems, it is not possible to infer a region specificity. Indeed, out of 24 studies, 14 reported to be generally on the mPFC (10) or PFC (4), 8 addressed the prelimbic or prelimbic/infralimbic region (without discrimination), and 2 specified the anterior cingulate cortex and the shoulder region or Fr2 region of the frontal cortex (without discrimination). Therefore, it is not possible to extrapolate differences in dopaminergic–GABAergic interaction among the mPFC subdivisions. Indeed, the prelimbic region seems to be the preferred target of most studies.
However, searching for a regional distinction might be pointless. Accumulating evidence suggests that current subdivisions might not reflect actual PFC functioning segregation. Recently, besides the cytoarchitecture and connectivity distinction criteria, the PFC gene expression profile has also been reported [75]. Interestingly, this study did not identify distinct subregions, but the genetic profile was, in fact, common to the multiple regions composing the PFC. Therefore, the PFC subdivisions based on connectivity or cytoarchitecture criteria, already not matching one another, are not confirmed by gene expression. This is of particular interest since it highlights a crucial aspect when considering PFC functions. The scenario that is emerging suggests that assigning different functions to the different PFC subdivisions is indeed deeply misleading. Based on connectivity alone, some distinctions seem to emerge, at least among the three main subdivisions (dmPFC, vmPFC, and vlPFC), which show different densities of specific connections. Nevertheless, their connectivity is not entirely differentiated, and the connections are shared but differ quantitatively [18]. Further based on this evidence, the dmPFC is often studied for sensorimotor behavior, the vmPFC is often associated with emotions and memory, while the vlPFC, though much less studied than the other two subdivisions, is often correlated to reward-related information and addictive behavior. However, this might reflect the common practice of the researchers rather than actual functional segregation. Several behavioral studies suggested that the perturbation of any PFC subdivision is sufficient to disrupt behavior and the whole cortical activity, independent of the type of task at hand (see [18] for an extensive discussion on this topic). Therefore, despite the different supposed roles of each subdivision, it is most likely that the PFC processes higher cognitive functions as a whole and cannot be assigned to a specific subregion [76].

4. Clinical Relevance

Given the evidence summarized so far, it is not surprising that several PFC-related pathologies involve alterations in both the dopaminergic and GABAergic systems. In the following paragraphs, we will briefly summarize the involvement of the dopaminergic and GABAergic systems in the main pathologies with a prominent PFC component, in particular schizophrenia and autism spectrum disorders.
Schizophrenia is one of the most studied cognitive pathologies, with a renowned involvement of the dopaminergic system, which is responsible for maintaining the proper E/I balance [77]. The “revised dopamine hypothesis” proposes that schizophrenic patients have hyperactive dopamine transmission in mesolimbic areas and hypoactive dopamine transmission in PFC [78]. The positive symptoms of schizophrenia include hallucinations and delusion due to an augmented dopamine release in subcortical areas, leading to an increase in D2-like receptors activation [79], and are thought to be caused by disrupted cortical pathways through the nucleus accumbens [80]. On the other hand, negative symptoms, such as anhedonia, lack of motivation, and speech impairments, result from reduced D1-like receptors activation in the PFC [79]. As computational models highlighted, the imbalance between D1-like and D2-like receptor activity might explain the positive and negative symptoms and the cognitive alterations in schizophrenia [81]. Interestingly, besides other players recently found involved (as the glutamatergic system and the NMDA receptors, [82,83]), the GABAergic system has been reported to be altered. In particular, a reduction in GABAergic inhibition is often reported (e.g., a reduced expression of GAD67, GAT1, and GABAA receptors; a decreased number of inhibitory interneurons; reduced inhibitory currents; [84] for details). The investigations on GABAergic disruption in schizophrenia are complicated since the alterations differ depending on the specific targeted PFC region [85]. In any case, GABAergic signaling alterations will contribute to the E/I balance disruption associated with this disease, both in humans and animal models. Alterations in GABA release have been correlated with impaired gamma oscillations and, as such, to the cognitive symptoms of the disease [86]. Interestingly, the GABAergic system deficit in the PFC has been proposed to result from the altered dopaminergic tone in the striatum in a mouse model with striatal D2 receptors overexpression [87]. Though the idea that the GABAergic and dopaminergic systems influence each other and collaborate in determining the pathological alterations in schizophrenia is not new [88,89], further research on this interaction might reveal critical to disentangle the complex pathophysiology of the disease. This would have a relevant impact from the clinical perspective. Independent of where the primary alteration occurred, a clinical intervention might need to impact both systems to regain a proper balance in PFC network activity. Moreover, a complete view of such a complex pathology will need to integrate the alterations seen in other neurotransmitter systems (such as the glutamatergic one) and the impact on the E/I balance of the glutamate/GABA interplay [77].
Autism spectrum disorders (ASD) are neurodevelopmental disorders characterized by deficits in social cognition, repetitive and stereotyped behavior, and restricted interests. The investigation of the pathophysiology of ASD is complicated by the incredibly heterogeneous genetic and phenotypic profiles that can be found in humans and the several animal models of the disease [90,91]. Nevertheless, the diverse molecular, cellular, and network alterations reported in literature seem to converge on a common outcome characterized by altered E/I balance (in favor of excitation), network hyperexcitability, and hyperresponsivity, often accompanied by altered long-range connectivity [92,93,94]. The GABAergic system is considered central for ASD research, and its interplay with the glutamatergic one to determine the E/I balance is one of the most studied topics in this field [95]. The most common alteration reported is a decrease inhibition efficiency, ultimately leading to the complex cognitive dysfunctions reported and the comorbidity with anxiety and other disorders [84]. The involvement of the dopaminergic system in ASD is supported by significant evidence in humans and animal models [96] and confirmed by the contribution of alterations in genes related to DA neurotransmission and its modulation [97]. The prefrontal cortex and striatum are considered the most affected brain regions. Given the role of the dopaminergic system in fine-tuning network transmission and signal-to-noise ratio during behavior, alterations in this system are considered causal for the reduced sociability and increased repetitive behavior that characterize ASD phenotype in mice and, most likely, in humans [98,99]. Therefore, ASD physiopathology could be the ideal ground to study the correlation between DA and GABAergic system alterations.
Affective disorders, such as major depression and bipolar disorder, and anxiety disorders are commonly associated with altered serotoninergic tone and glutamate/GABA systems imbalance. Nevertheless, many symptoms are considered to rely on dopaminergic miscontrol leading, for example, to a lack of motivation and anhedonia in depression [100,101]. In particular, many forms of depression have been correlated with PFC hyperactivity, and acting on the systems controlling the E/I balance in this region is the primary treatment approach to date [100]. The circuits responsible for the stress response, including the hippocampus and amygdala, are also involved in the altered PFC-related communication found in these disorders [100,102]. Altered DA signaling is also reported in post-traumatic stress disorder [103]. Moreover, the involvement of the dopaminergic system in pain modulation and chronic pain can be considered related to the previous disorders [104,105]. Interestingly, the increased mPFC output observed in neuropathic pain conditions has been correlated with altered VTA-mediated DA control over the prelimbic region in rats, associated with impaired integration of GABAergic inhibition [106].

5. Conclusions

The dopaminergic system modulates the PFC network activity state, finely tuning the signal-to-noise ratio and the E/I balance. These effects are partially exerted influencing the GABAergic system through complex intracellular pathways that modify GABA receptors expression and activity, and modulate GABA release by INs. DA control of the PFC activity state and responsiveness modulates the gain of signal transmission modifying the tonic DA level and regulates the timing of neuronal responses through its complex phasic component. The PFC is one of the most integrative areas in the brain, and the interplay between the dopaminergic and GABAergic systems is one of the critical features that influence input integration by this network and therefore deserves special attention. Further effort should also be devoted to exploring the reciprocal influence of these two systems in PFC-related neuropathologies. More often than not, the alterations in DA and GABAergic systems and their impact on the clinical perspective are studied separately. This is undoubtedly due to the intrinsic difficulty in disentangling the relative contribution of the two systems to the alterations observed and to the limitations of using animal models for addressing cognitive phenotype. Nevertheless, the data summarized in this mini review strongly support the idea that the interplay between these two systems significantly contributes to originate the unbalance seen in pathological models, possibly with a primarily affected system causing the impairment of the other. The recent technological advancements and the application of computational models could boost the research in this field and allow us to address this issue with a renewed effort.

Author Contributions

Conceptualization, L.M.; writing—original draft preparation, D.D.D. and L.M.; writing—review and editing, L.M.; visualization, D.D.D.; supervision, L.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ACadenylyl cyclase
ACCanterior cingulate cortex
ASDautism spectrum disorders
cAMPcyclic adenosine monophosphate
DAdopamine
DARPP-32DA and cAMP-regulated phosphoprotein
dmPFCdorsomedial prefrontal cortex
eIPSPevoked inhibitory postsynaptic potential
EPSPexcitatory postsynaptic potential
E/Iexcitation/inhibition
FSfast-spiking interneurons
GAD67glutamate decarboxylase 67
GAT1GABA transporter type 1
INinhibitory interneuron
IP3inositol triphosphate
IPSPinhibitory postsynaptic potential
mPFCmedial prefrontal cortex
PFCprefrontal cortex
PNpyramidal neuron
VTAventral tegmental area
GPCRG-protein coupled receptor
IPSCinhibitory post-synaptic current
PKAprotein kinase A
PLCphospholipase C
PP1protein phosphatase 1
PP2Bcalmodulin-dependent protein phosphatase
sIPSPspontaneous inhibitory postsynaptic potential
VIPvasoactive intestinal peptide
vlPFCventrolateral prefrontal cortex
vmPFCventromedial prefrontal cortex

References

  1. Elliott, R. Executive Functions and Their Disorders. Br. Med. Bull. 2003, 65, 49–59. [Google Scholar] [CrossRef] [PubMed]
  2. Huang, Y.Y.; Simpson, E.; Kellendonk, C.; Kandel, E.R. Genetic Evidence for the Bidirectional Modulation of Synaptic Plasticity in the Prefrontal Cortex by D1 Receptors. Proc. Natl. Acad. Sci. USA 2004, 101, 3236–3241. [Google Scholar] [CrossRef] [PubMed]
  3. Aron, A.R.; Robbins, T.W.; Poldrack, R.A. Inhibition and the Right Inferior Frontal Cortex. Trends Cogn. Sci. 2004, 8, 170–177. [Google Scholar] [CrossRef] [PubMed]
  4. Buschman, T.J.; Miller, E.K. Top-down versus bottom-up control of attention in the prefrontal and posterior parietal cortices. Science 2007, 315, 1860–1862. [Google Scholar] [CrossRef]
  5. Goldman-Rakic, P.S. The Cortical Dopamine System: Role in Memory and Cognition. Adv. Pharmacol. 1997, 42, 707–711. [Google Scholar] [CrossRef]
  6. Goldman-Rakic, P.S. Cellular Basis of Working Memory. Neuron 1995, 14, 477–485. [Google Scholar] [CrossRef]
  7. Goldman-Rakic, P.S. The Physiological Approach: Functional Architecture of Working Memory and Disordered Cognition in Schizophrenia. Biol. Psychiatry 1999, 46, 650–661. [Google Scholar] [CrossRef]
  8. Kesner, R.P.; Churchwell, J.C. An Analysis of Rat Prefrontal Cortex in Mediating Executive Function. Neurobiol. Learn. Mem. 2011, 96, 417–431. [Google Scholar] [CrossRef]
  9. Arnsten, A.F.T.; Wang, M.J.; Paspalas, C.D. Neuromodulation of Thought: Flexibilities and Vulnerabilities in Prefrontal Cortical Network Synapses. Neuron 2012, 76, 223–239. [Google Scholar] [CrossRef]
  10. Volk, D.; Lewis, D. GABA Targets for the Treatment of Cognitive Dysfunction in Schizophrenia. Curr. Neuropharmacol. 2005, 3, 45–62. [Google Scholar] [CrossRef]
  11. Seamans, J.K.; Lapish, C.C.; Durstewitz, D. Comparing the Prefrontal Cortex of Rats and Primates: Insights from Electrophysiology. Neurotox. Res. 2008, 14, 249–262. [Google Scholar] [CrossRef] [PubMed]
  12. Carlén, M. What Constitutes the Prefrontal Cortex? Science 2017, 358, 478–482. [Google Scholar] [CrossRef]
  13. Laubach, M.; Amarante, L.M.; Swanson, K.; White, S.R. What, If Anything, Is Rodent Prefrontal Cortex? eNeuro 2018, 5, ENEURO.0315-18.2018. [Google Scholar] [CrossRef] [PubMed]
  14. Fuster, J.M. The Prefrontal Cortex—An Update: Time Is of the Essence. Neuron 2001, 30, 319–333. [Google Scholar] [CrossRef]
  15. Rose, J.E.; Woolsey, C.N. The Orbitofrontal Cortex and Its Connections with the Mediodorsal Nucleus in Rabbit, Sheep and Cat. Res. Publ. Assoc. Res. Nerv. Ment. Dis. 1948, 27, 210–232. [Google Scholar]
  16. Otani, S. (Ed.) Prefrontal Cortex: From Synaptic Plasticity to Cognition; Springer Science & Business Media: Berlin/Heidelberg, Germany, 2004. [Google Scholar]
  17. Van Essen, D.C.; Glasser, M.F. Parcellating Cerebral Cortex: How Invasive Animal Studies Inform Noninvasive Mapmaking in Humans. Neuron 2018, 99, 640–663. [Google Scholar] [CrossRef] [PubMed]
  18. Le Merre, P.; Ährlund-Richter, S.; Carlén, M. The Mouse Prefrontal Cortex: Unity in Diversity. Neuron 2021, 109, 1925–1944. [Google Scholar] [CrossRef]
  19. Harris, J.A.; Mihalas, S.; Hirokawa, K.E.; Whitesell, J.D.; Choi, H.; Bernard, A.; Bohn, P.; Caldejon, S.; Casal, L.; Cho, A.; et al. Hierarchical Organization of Cortical and Thalamic Connectivity. Nature 2019, 575, 195–202. [Google Scholar] [CrossRef]
  20. Ercsey-Ravasz, M.; Markov, N.T.; Lamy, C.; VanEssen, D.C.; Knoblauch, K.; Toroczkai, Z.; Kennedy, H. A Predictive Network Model of Cerebral Cortical Connectivity Based on a Distance Rule. Neuron 2013, 80, 184–197. [Google Scholar] [CrossRef]
  21. Gămănuţ, R.; Kennedy, H.; Toroczkai, Z.; Ercsey-Ravasz, M.; Van Essen, D.C.; Knoblauch, K.; Burkhalter, A. The Mouse Cortical Connectome, Characterized by an Ultra-Dense Cortical Graph, Maintains Specificity by Distinct Connectivity Profiles. Neuron 2018, 97, 698–715.e10. [Google Scholar] [CrossRef]
  22. Bullmore, E.; Sporns, O. Complex Brain Networks: Graph Theoretical Analysis of Structural and Functional Systems. Nat. Rev. Neurosci. 2009, 10, 186–198. [Google Scholar] [CrossRef] [PubMed]
  23. Riga, D.; Matos, M.R.; Glas, A.; Smit, A.B.; Spijker, S.; Van den Oever, M.C. Optogenetic Dissection of Medial Prefrontal Cortex Circuitry. Front. Syst. Neurosci. 2014, 8, 230. [Google Scholar] [CrossRef] [PubMed]
  24. Palmer, L.; Murayama, M.; Larkum, M. Inhibitory Regulation of Dendritic Activity in Vivo. Front. Neural Circuits 2012, 6, 26. [Google Scholar] [CrossRef]
  25. Anastasiades, P.G.; Carter, A.G. Circuit Organization of the Rodent Medial Prefrontal Cortex. Trends Neurosci. 2021, 44, 550–563. [Google Scholar] [CrossRef] [PubMed]
  26. Whittington, M.A.; Traub, R.D. Interneuron Diversity Series: Inhibitory Interneurons and Network Oscillations in Vitro. Trends Neurosci. 2003, 26, 676–682. [Google Scholar] [CrossRef] [PubMed]
  27. Kvitsiani, D.; Ranade, S.; Hangya, B.; Taniguchi, H.; Huang, J.Z.; Kepecs, A. Distinct Behavioural and Network Correlates of Two Interneuron Types in Prefrontal Cortex. Nature 2013, 498, 363–366. [Google Scholar] [CrossRef]
  28. Cobb, S.R.; Buhl, E.H.; Halasy, K.; Paulsen, O.; Somogyi, P. Synchronization of Neuronal Activity in Hippocampus by Individual GABAergic Interneurons. Nature 1995, 378, 75–78. [Google Scholar] [CrossRef]
  29. Seamans, J.K.; Yang, C.R. The Principal Features and Mechanisms of Dopamine Modulation in the Prefrontal Cortex. Prog. Neurobiol. 2004, 74, 1–58. [Google Scholar] [CrossRef]
  30. Puig, M.V.; Rose, J.; Schmidt, R.; Freund, N. Dopamine Modulation of Learning and Memory in the Prefrontal Cortex: Insights from Studies in Primates, Rodents, and Birds. Front. Neural Circuits 2014, 8, 93. [Google Scholar] [CrossRef]
  31. Ott, T.; Nieder, A. Dopamine and Cognitive Control in Prefrontal Cortex. Trends Cogn. Sci. 2019, 23, 213–234. [Google Scholar] [CrossRef]
  32. Tritsch, N.X.; Sabatini, B.L. Dopaminergic Modulation of Synaptic Transmission in Cortex and Striatum. Neuron 2012, 76, 33–50. [Google Scholar] [CrossRef] [PubMed]
  33. Santana, N.; Mengod, G.; Artigas, F. Quantitative Analysis of the Expression of Dopamine D1 and D2 Receptors in Pyramidal and GABAergic Neurons of the Rat Prefrontal Cortex. Cereb. Cortex 2009, 19, 849–860. [Google Scholar] [CrossRef] [PubMed]
  34. Beaulieu, J.M.; Gainetdinov, R.R. The Physiology, Signaling, and Pharmacology of Dopamine Receptors. Pharmacol. Rev. 2011, 63, 182–217. [Google Scholar] [CrossRef]
  35. Huang, S.; Borgland, S.L.; Zamponi, G.W. Dopaminergic Modulation of Pain Signals in the Medial Prefrontal Cortex: Challenges and Perspectives. Neurosci. Lett. 2019, 702, 71–76. [Google Scholar] [CrossRef] [PubMed]
  36. Lee, S.P.; So, C.H.; Rashid, A.J.; Varghese, G.; Cheng, R.; Lança, A.J.; O’Dowd, B.F.; George, S.R. Dopamine D1 and D2 Receptor Co-Activation Generates a Novel Phospholipase C-Mediated Calcium Signal. J. Biol. Chem. 2004, 279, 35671–35678. [Google Scholar] [CrossRef]
  37. Sahu, A.; Tyeryar, K.R.; Vongtau, H.O.; Sibley, D.R.; Undieh, A.S. D 5 Dopamine Receptors Are Required for Dopaminergic Activation of Phospholipase C. Mol. Pharmacol. 2009, 75, 447–453. [Google Scholar] [CrossRef]
  38. Graziane, N.M.; Yuen, E.Y.; Yan, Z. Dopamine D4 Receptors Regulate GABAA Receptor Trafficking via an Actin/Cofilin/Myosin-Dependent Mechanism. J. Biol. Chem. 2009, 284, 8329–8336. [Google Scholar] [CrossRef]
  39. Lewis, D.A.; Gonzalez-Burgos, G. Pathophysiologically Based Treatment Interventions in Schizophrenia. Nat. Med. 2006, 12, 1016–1022. [Google Scholar] [CrossRef]
  40. O’Donnell, P. Adolescent Onset of Cortical Disinhibition in Schizophrenia: Insights from Animal Models. Schizophr. Bull. 2011, 37, 484–492. [Google Scholar] [CrossRef]
  41. Tseng, K.Y.; Chambers, R.A.; Lipska, B.K. The Neonatal Ventral Hippocampal Lesion as a Heuristic Neurodevelopmental Model of Schizophrenia. Behav. Brain Res. 2009, 204, 295–305. [Google Scholar] [CrossRef]
  42. Mrzljak, L.; Bergson, C.; Pappy, M.; Huff, R.; Levenson, R.; Goldman-Rakic, P.S. Localization of Dopamine D4 Receptors in GABAergic Neurons of the Primate Brain. Nature 1996, 381, 245–248. [Google Scholar] [CrossRef] [PubMed]
  43. Ariano, M.A.; Wang, J.; Noblett, K.L.; Larson, E.R.; Sibley, D.R. Cellular Distribution of the Rat D4 Dopamine Receptor Protein in the CNS Using Anti-Receptor Antisera. Brain Res. 1997, 752, 26–34. [Google Scholar] [CrossRef] [PubMed]
  44. Wȩdzony, K.; Chocyk, A.; Maćkowiak, M.; Fijał, K.; Czyrak, A. Cortical Localization of Dopamine D4 Receptors in the Rat Brain—Immunocytochemical Study. J. Physiol. Pharmacol. 2000, 51, 205–221. [Google Scholar] [PubMed]
  45. Smiley, J.F.; Levey, A.I.; Ciliax, B.J.; Goldman-Rakic, P.S. D1 Dopamine Receptor Immunoreactivity in Human and Monkey Cerebral Cortex: Predominant and Extrasynaptic Localization in Dendritic Spines. Proc. Natl. Acad. Sci. USA 1994, 91, 5720–5724. [Google Scholar] [CrossRef] [PubMed]
  46. Wang, X.; Zhong, P.; Yan, Z. Dopamine D4 Modulate GABAergic Signaling in Pyramidal Neurons. J. Neurosci. 2002, 22, 9185–9193. [Google Scholar] [CrossRef]
  47. Seamans, J.K.; Gorelova, N.; Durstewitz, D.; Yang, C.R. Bidirectional Dopamine Modulation of GABAergic Inhibition in Prefrontal Cortical Pyramidal Neurons. J. Neurosci. 2001, 21, 3628–3638. [Google Scholar] [CrossRef]
  48. Chiu, C.Q.; Puente, N.; Grandes, P.; Castillo, P.E. Dopaminergic Modulation of Endocannabinoid-Mediated Plasticity at GABAergic Synapses in the Prefrontal Cortex. J. Neurosci. 2010, 30, 7236–7248. [Google Scholar] [CrossRef]
  49. Zhou, F.M.; Hablitz, J.J. Dopamine Modulation of Membrane and Synaptic Properties of Interneurons in Rat Cerebral Cortex. J. Neurophysiol. 1999, 81, 967–976. [Google Scholar] [CrossRef]
  50. Penit-Soria, J.; Audinat, E.; Crepel, F. Excitation of Rat Prefrontal Cortical Neurons by Dopamine: An in Vitro Electrophysiological Study. Brain Res. 1987, 425, 263–274. [Google Scholar] [CrossRef]
  51. Law-Tho, D.; Desce, J.M.; Crepel, F. Dopamine Favours the Emergence of Long-Term Depression versus Long-Term Potentiation in Slices of Rat Prefrontal Cortex. Neurosci. Lett. 1995, 188, 125–128. [Google Scholar] [CrossRef]
  52. Gonzalez-Islas, C.; Hablitz, J.J. Dopamine Inhibition of Evoked IPSCs in Rat Prefrontal Cortex. J. Neurophysiol. 2001, 86, 2911–2918. [Google Scholar] [CrossRef] [PubMed]
  53. Gao, W.J.; Goldman-Rakic, P.S. Selective Modulation of Excitatory and Inhibitory Microcircuits by Dopamine. Proc. Natl. Acad. Sci. USA 2003, 100, 2836–2841. [Google Scholar] [CrossRef] [PubMed]
  54. Chris Muly, E.; Szigeti, K.; Goldman-Rakic, P.S. D1 Receptor in Interneurons of Macaque Prefrontal Cortex: Distribution and Subcellular Localization. J. Neurosci. 1998, 18, 10553–10565. [Google Scholar] [CrossRef] [PubMed]
  55. Glausier, J.R.; Khan, Z.U.; Muly, E.C. Dopamine D1 and D5 Receptors Are Localized to Discrete Populations of Interneurons in Primate Prefrontal Cortex. Cereb. Cortex 2009, 19, 1820–1834. [Google Scholar] [CrossRef] [PubMed]
  56. Gorelova, N.; Seamans, J.K.; Yang, C.R. Mechanisms of Dopamine Activation of Fast-Spiking Interneurons That Exert Inhibition in Rat Prefrontal Cortex. J. Neurophysiol. 2002, 88, 3150–3166. [Google Scholar] [CrossRef] [PubMed]
  57. Tseng, K.Y.; O’Donnell, P. Dopamine Modulation of Prefrontal Cortical Interneurons Changes during Adolescence. Cereb. Cortex 2007, 17, 1235–1240. [Google Scholar] [CrossRef] [PubMed]
  58. Anastasiades, P.G.; Boada, C.; Carter, A.G. Cell-Type-Specific D1 Dopamine Receptor Modulation of Projection Neurons and Interneurons in the Prefrontal Cortex. Cereb. Cortex 2019, 29, 3224–3242. [Google Scholar] [CrossRef] [PubMed]
  59. Ferron, A.; Thierry, A.M.; Le Douarin, C.; Glowinski, J. Inhibitory Influence of the Mesocortical Dopaminergic System on Spontaneous Activity or Excitatory Response Induced from the Thalamic Mediodorsal Nucleus in the Rat Medial Prefrontal Cortex. Brain Res. 1984, 302, 257–265. [Google Scholar] [CrossRef]
  60. Lewis, B.L.; O’Donnell, P. Ventral Tegmental Area Afferents to the Prefrontal Cortex Maintain Membrane Potential “up” States in Pyramidal Neurons via D1 Dopamine Receptors. Cereb. Cortex 2000, 10, 1168–1175. [Google Scholar] [CrossRef]
  61. Tseng, K.Y.; Mallet, N.; Toreson, K.L.; Le Moine, C.; Gonon, F.; O’Donnell, P. Excitatory Response of Prefrontal Cortical Fast-Spiking Interneurons to Ventral Tegmental Area Stimulation in Vivo. Synapse 2006, 59, 412–417. [Google Scholar] [CrossRef]
  62. Garris, P.A.; Collins, L.B.; Jones, S.R.; Wightman, R.M. Evoked Extracellular Dopamine In Vivo in the Medial Prefrontal Cortex. J. Neurochem. 1993, 61, 637–647. [Google Scholar] [CrossRef] [PubMed]
  63. Garris, P.A.; Wightman, R.M. Different Kinetics Govern Dopaminergic Transmission in the Amygdala, Prefrontal Cortex, and Striatum: An in Vivo Voltammetric Study. J. Neurosci. 1994, 14, 442–450. [Google Scholar] [CrossRef] [PubMed]
  64. Pirot, S.; Godbout, R.; Mantz, J.; Tassin, J.P.; Glowinski, J.; Thierry, A.M. Inhibitory Effects of Ventral Tegmental Area Stimulation on the Activity of Prefrontal Cortical Neurons: Evidence for the Involvement of Both Dopaminergic and GABAergic Components. Neuroscience 1992, 49, 857–865. [Google Scholar] [CrossRef] [PubMed]
  65. West, A.R.; Grace, A.A. Opposite Influences of Endogenous Dopamine D1 and D2 Receptor Activation on Activity States and Electrophysiological Properties of Striatal Neurons: Studies Combining in Vivo Intracellular Recordings and Reverse Microdialysis. J. Neurosci. 2002, 22, 294–304. [Google Scholar] [CrossRef] [PubMed]
  66. Rubinstein, M.; Cepeda, C.; Hurst, R.S.; Flores-Hernandez, J.; Ariano, M.A.; Falzone, T.L.; Kozell, L.B.; Meshul, C.K.; Bunzow, J.R.; Low, M.J.; et al. Dopamine D4 Receptor-Deficient Mice Display Cortical Hyperexcitability. J. Neurosci. 2001, 21, 3756–3763. [Google Scholar] [CrossRef]
  67. Wang, X.; Zhong, P.; Gu, Z.; Yan, Z. Regulation of NMDA Receptors by Dopamine D4 Signaling in Prefrontal Cortex. J. Neurosci. 2003, 23, 9852–9861. [Google Scholar] [CrossRef]
  68. Lew, S.E.; Tseng, K.Y. Dopamine Modulation of GABAergic Function Enables Network Stability and Input Selectivity for Sustaining Working Memory in a Computational Model of the Prefrontal Cortex. Neuropsychopharmacology 2014, 39, 3067–3076. [Google Scholar] [CrossRef] [PubMed]
  69. Durstewitz, D.; Seamans, J.K. The Computational Role of Dopamine D1 Receptors in Working Memory. Neural Networks 2002, 15, 561–572. [Google Scholar] [CrossRef]
  70. Brunel, N.; Sup, E.N.; Wang, X. Effects of Neuromodulation in a Cortical Network Model of Object Working. J. Comput. Neurosci. 2001, 11, 63–85. [Google Scholar] [CrossRef]
  71. Trantham-Davidson, H.; Neely, L.C.; Lavin, A.; Seamans, J.K. Mechanisms Underlying Differential D1 versus D2 Dopamine Receptor Regulation of Inhibition in Prefrontal Cortex. J. Neurosci. 2004, 24, 10652–10659. [Google Scholar] [CrossRef]
  72. Seamans, J.K.; Durstewitz, D.; Christie, B.R.; Stevens, C.F.; Sejnowski, T.J. Dopamine D1/D5 Receptor Modulation of Excitatory Synaptic Inputs to Layer V Prefrontal Cortex Neurons. Proc. Natl. Acad. Sci. USA 2001, 98, 301–306. [Google Scholar] [CrossRef]
  73. Tseng, K.Y.; O’Donnell, P. Dopamine-Glutamate Interactions Controlling Prefrontal Cortical Pyramidal Cell Excitability Involve Multiple Signaling Mechanisms. J. Neurosci. 2004, 24, 5131–5139. [Google Scholar] [CrossRef]
  74. Ott, T.; Nieder, A. Dopamine D2 Receptors Enhance Population Dynamics in Primate Prefrontal Working Memory Circuits. Cereb. Cortex 2017, 27, 4423–4435. [Google Scholar] [CrossRef] [PubMed]
  75. Ortiz, C.; Navarro, J.F.; Jurek, A.; Märtin, A.; Lundeberg, J.; Meletis, K. Molecular Atlas of the Adult Mouse Brain. Sci. Adv. 2020, 6, eabb3446. [Google Scholar] [CrossRef]
  76. Wilson, C.R.E.; Gaffan, D.; Browning, P.G.F.; Baxter, M.G. Functional Localization within the Prefrontal Cortex: Missing the Forest for the Trees? Trends Neurosci. 2010, 33, 533–540. [Google Scholar] [CrossRef] [PubMed]
  77. Liu, Y.; Ouyang, P.; Zheng, Y.; Mi, L.; Zhao, J.; Ning, Y.; Guo, W. A Selective Review of the Excitatory-Inhibitory Imbalance in Schizophrenia: Underlying Biology, Genetics, Microcircuits, and Symptoms. Front. Cell Dev. Biol. 2021, 9, 2917. [Google Scholar] [CrossRef] [PubMed]
  78. da Silva Alves, F.; Figee, M.; van Avamelsvoort, T.; Veltman, D.; de Haan, L. The Revised Dopamine Hypothesis of Schizophrenia: Evidence from Pharmacological MRI Studies with Atypical Antipsychotic Medication. Psychopharmacol. Bull. 2008, 41, 121–132. [Google Scholar]
  79. Shen, L.H.; Liao, M.H.; Tseng, Y.C. Recent Advances in Imaging of Dopaminergic Neurons for Evaluation of Neuropsychiatric Disorders. J. Biomed. Biotechnol. 2012, 2012, 259349. [Google Scholar] [CrossRef]
  80. O’Donnell, P.; Grace, A.A. Dysfunctions in Multiple Interrelated Systems as the Neurobiological Bases of Schizophrenic Symptom Clusters. Schizophr. Bull. 1998, 24, 267–283. [Google Scholar] [CrossRef]
  81. Durstewitz, D.; Seamans, J.K. The Dual-State Theory of Prefrontal Cortex Dopamine Function with Relevance to Catechol-O-Methyltransferase Genotypes and Schizophrenia. Biol. Psychiatry 2008, 64, 739–749. [Google Scholar] [CrossRef]
  82. Nakazawa, K.; Sapkota, K. The Origin of NMDA Receptor Hypofunction in Schizophrenia. Pharmacol. Ther. 2020, 205, 107426. [Google Scholar] [CrossRef]
  83. Kruse, A.O.; Bustillo, J.R. Glutamatergic Dysfunction in Schizophrenia. Transl. Psychiatry 2022, 12, 500. [Google Scholar] [CrossRef]
  84. Zhang, W.; Xiong, B.R.; Zhang, L.Q.; Huang, X.; Yuan, X.; Tian, Y.K.; Tian, X.B. The Role of the GABAergic System in Diseases of the Central Nervous System. Neuroscience 2021, 470, 88–99. [Google Scholar] [CrossRef] [PubMed]
  85. Benes, F.M. The GABA System in Schizophrenia: Cells, Molecules and Microcircuitry. Schizophr. Res. 2015, 167, 1–3. [Google Scholar] [CrossRef]
  86. Crabtree, G.W.; Park, A.J.; Gordon, J.A.; Gogos, J.A. Cytosolic Accumulation of L-Proline Disrupts GABA-Ergic Transmission through GAD Blockade. Cell Rep. 2016, 17, 570–582. [Google Scholar] [CrossRef] [PubMed]
  87. Li, Y.C.; Kellendonk, C.; Simpson, E.H.; Kandel, E.R.; Gao, W.J. D2 Receptor Overexpression in the Striatum Leads to a Deficit in Inhibitory Transmission and Dopamine Sensitivity in Mouse Prefrontal Cortex. Proc. Natl. Acad. Sci. USA 2011, 108, 12107–12112. [Google Scholar] [CrossRef] [PubMed]
  88. Garbutt, J.C.; Van Kammen, D.P. The Interaction between GABA and Dopamine: Implications for Schizophrenia. Schizophr. Bull. 1983, 9, 336–353. [Google Scholar] [CrossRef]
  89. Benes, F.M. The Role of Stress and Dopamine-GABA Interactions in the Vulnerability for Schizophrenia. J. Psychiatr. Res. 1997, 31, 257–275. [Google Scholar] [CrossRef]
  90. Bey, A.; Jiang, Y.H. Overview of mouse models of autism spectrum disorders. Curr Protoc Pharmacol. 2014, 66, 1–26. [Google Scholar] [CrossRef]
  91. Mapelli, L.; Soda, T.; D’Angelo, E.; Prestori, F. The Cerebellar Involvement in Autism Spectrum Disorders: From the Social Brain to Mouse Models. Int. J. Mol. Sci. 2022, 23, 3894. [Google Scholar] [CrossRef]
  92. Uzunova, G.; Pallanti, S.; Hollander, E. Excitatory/Inhibitory Imbalance in Autism Spectrum Disorders: Implications for Interventions and Therapeutics. World J. Biol. Psychiatry 2016, 17, 174–186. [Google Scholar] [CrossRef] [PubMed]
  93. Rinaldi, T.; Perrodin, C.; Markram, H. Hyper-Connectivity and Hyper-Plasticity in the Medial Prefrontal Cortex in the Valproic Acid Animal Model of Autism. Front. Neural Circuits 2008, 2, 4. [Google Scholar] [CrossRef]
  94. Soda, T.; Mapelli, L.; Locatelli, F.; Botta, L.; Goldfarb, M.; Prestori, F.; D’Angelo, E.U. Hyperexcitability and Hyperplasticity Disrupt Cerebellar Signal Transfer in the Ib2 Ko Mouse Model of Autism. J. Neurosci. 2019, 39, 2383–2397, Erratum in J. Neurosci. 2019, 39, 7029. [Google Scholar] [CrossRef] [PubMed]
  95. Antoine, M.W.; Langberg, T.; Schnepel, P.; Feldman, D.E. Increased Excitation-Inhibition Ratio Stabilizes Synapse and Circuit Excitability in Four Autism Mouse Models. Neuron 2019, 101, 648–661.e4. [Google Scholar] [CrossRef] [PubMed]
  96. Kosillo, P.; Bateup, H.S. Dopaminergic Dysregulation in Syndromic Autism Spectrum Disorders: Insights From Genetic Mouse Models. Front. Neural Circuits 2021, 15, 700968. [Google Scholar] [CrossRef]
  97. Nguyen, M.; Roth, A.; Kyzar, E.J.; Poudel, M.K.; Wong, K.; Stewart, A.M.; Kalueff, A.V. Decoding the Contribution of Dopaminergic Genes and Pathways to Autism Spectrum Disorder (ASD). Neurochem. Int. 2014, 66, 15–26. [Google Scholar] [CrossRef] [PubMed]
  98. Gunaydin, L.A.; Grosenick, L.; Finkelstein, J.C.; Kauvar, I.V.; Fenno, L.E.; Adhikari, A.; Lammel, S.; Mirzabekov, J.J.; Airan, R.D.; Zalocusky, K.A.; et al. Natural Neural Projection Dynamics Underlying Social Behavior. Cell 2014, 157, 1535–1551. [Google Scholar] [CrossRef]
  99. Lee, Y.; Kim, H.; Kim, J.E.; Park, J.Y.; Choi, J.; Lee, J.E.; Lee, E.H.; Han, P.L. Excessive D1 Dopamine Receptor Activation in the Dorsal Striatum Promotes Autistic-Like Behaviors. Mol. Neurobiol. 2018, 55, 5658–5671. [Google Scholar] [CrossRef]
  100. Grace, A.A. Dysregulation of the Dopamine System in the Pathophysiology of Schizophrenia and Depression. Nat. Rev. Neurosci. 2016, 17, 524–532. [Google Scholar] [CrossRef]
  101. Wise, R.A. Dopamine and Reward: The Anhedonia Hypothesis 30 Years On. Neurotox. Res. 2008, 14, 169–183. [Google Scholar] [CrossRef]
  102. McKlveen, J.M.; Moloney, R.D.; Scheimann, J.R.; Myers, B.; Herman, J.P. “Braking” the Prefrontal Cortex: The Role of Glucocorticoids and Interneurons in Stress Adaptation and Pathology. Biol. Psychiatry 2019, 86, 669–681. [Google Scholar] [CrossRef]
  103. Yan, R.; Wang, T.; Zhou, Q. Elevated Dopamine Signaling from Ventral Tegmental Area to Prefrontal Cortical Parvalbumin Neurons Drives Conditioned Inhibition. Proc. Natl. Acad. Sci. USA 2019, 116, 13077–13086. [Google Scholar] [CrossRef] [PubMed]
  104. Kummer, K.K.; Mitrić, M.; Kalpachidou, T.; Kress, M. The Medial Prefrontal Cortex as a Central Hub for Mental Comorbidities Associated with Chronic Pain. Int. J. Mol. Sci. 2020, 21, 3440. [Google Scholar] [CrossRef] [PubMed]
  105. Bushnell, M.C.; Čeko, M.; Low, L.A. Cognitive and Emotional Control of Pain and Its Disruption in Chronic Pain. Nat. Rev. Neurosci. 2013, 14, 502–511. [Google Scholar] [CrossRef] [PubMed]
  106. Huang, S.; Zhang, Z.; Gambeta, E.; Xu, S.C.; Thomas, C.; Godfrey, N.; Chen, L.; M’Dahoma, S.; Borgland, S.L.; Zamponi, G.W. Dopamine Inputs from the Ventral Tegmental Area into the Medial Prefrontal Cortex Modulate Neuropathic Pain-Associated Behaviors in Mice. Cell Rep. 2020, 31, 107812. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Main intracellular pathways activated by dopamine receptors. The scheme shows different pathways in which dopamine (DA) affects the modulation of intracellular signaling. DA can regulate the activation state of (a) adenylyl cyclase (AC) or (b) phospholipase C (PLC) binding either D1-like or D2-like receptors. (c) Both pathways lead to a modulation (either positive or negative) of DARPP-32 which regulates the expression of GABA receptors. DA also affects neuronal excitability by modulating voltage-dependent ion channels via activation of (d) β/γ subunit or (e) AC pathway. The forward and stop arrows indicate activation or inhibition of the next element in the chain, respectively. This figure was created with BioRender.com.
Figure 1. Main intracellular pathways activated by dopamine receptors. The scheme shows different pathways in which dopamine (DA) affects the modulation of intracellular signaling. DA can regulate the activation state of (a) adenylyl cyclase (AC) or (b) phospholipase C (PLC) binding either D1-like or D2-like receptors. (c) Both pathways lead to a modulation (either positive or negative) of DARPP-32 which regulates the expression of GABA receptors. DA also affects neuronal excitability by modulating voltage-dependent ion channels via activation of (d) β/γ subunit or (e) AC pathway. The forward and stop arrows indicate activation or inhibition of the next element in the chain, respectively. This figure was created with BioRender.com.
Biomedicines 11 01276 g001
Figure 2. Dopaminergic receptors distribution in the PFC and main effects on inhibition. The distribution of dopamine (DA) receptors among PFC layers and their expression on different neuronal types can variably affect inhibition. In layer II/III, DA (green dots) binding D1-like receptors (orange) on pyramidal neurons (PNs, (a)) increases spontaneous IPSC (sIPSC); (b) on vasoactive intestinal peptide (VIP) neurons, it starts internal loops inhibiting deeper layers’ inhibitory interneurons (INs); and (c) on fast-spiking interneurons (FS) increases intrinsic excitability. DA binding D1-like receptor expressed in layer V PNs (d) increases the IPSC. DA binding D2-like receptors (blue) expressed in layer V PNs (e) decreases the IPSC. Expression of both D1-like and D2-like receptors in layer V PNs (f) increases the IPSC mediated by D2-like receptor activation (1) followed by a IPSC decrease mediated by D1-like receptor activation (2). On INs (g), the decrease in the IPSC mediated by D1-like receptors (1) is followed by an increased IPSC mediated by D2-like receptors (2). (h) In layer VI PNs, the activation of DA receptors by high DA concentration leads to an increase in sIPSC and a decrease in evoked IPSC (eIPSC).
Figure 2. Dopaminergic receptors distribution in the PFC and main effects on inhibition. The distribution of dopamine (DA) receptors among PFC layers and their expression on different neuronal types can variably affect inhibition. In layer II/III, DA (green dots) binding D1-like receptors (orange) on pyramidal neurons (PNs, (a)) increases spontaneous IPSC (sIPSC); (b) on vasoactive intestinal peptide (VIP) neurons, it starts internal loops inhibiting deeper layers’ inhibitory interneurons (INs); and (c) on fast-spiking interneurons (FS) increases intrinsic excitability. DA binding D1-like receptor expressed in layer V PNs (d) increases the IPSC. DA binding D2-like receptors (blue) expressed in layer V PNs (e) decreases the IPSC. Expression of both D1-like and D2-like receptors in layer V PNs (f) increases the IPSC mediated by D2-like receptor activation (1) followed by a IPSC decrease mediated by D1-like receptor activation (2). On INs (g), the decrease in the IPSC mediated by D1-like receptors (1) is followed by an increased IPSC mediated by D2-like receptors (2). (h) In layer VI PNs, the activation of DA receptors by high DA concentration leads to an increase in sIPSC and a decrease in evoked IPSC (eIPSC).
Biomedicines 11 01276 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Di Domenico, D.; Mapelli, L. Dopaminergic Modulation of Prefrontal Cortex Inhibition. Biomedicines 2023, 11, 1276. https://doi.org/10.3390/biomedicines11051276

AMA Style

Di Domenico D, Mapelli L. Dopaminergic Modulation of Prefrontal Cortex Inhibition. Biomedicines. 2023; 11(5):1276. https://doi.org/10.3390/biomedicines11051276

Chicago/Turabian Style

Di Domenico, Danila, and Lisa Mapelli. 2023. "Dopaminergic Modulation of Prefrontal Cortex Inhibition" Biomedicines 11, no. 5: 1276. https://doi.org/10.3390/biomedicines11051276

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop