Skip to main content

The regulatory role of AP-2β in monoaminergic neurotransmitter systems: insights on its signalling pathway, linked disorders and theragnostic potential

Abstract

Monoaminergic neurotransmitter systems play a central role in neuronal function and behaviour. Dysregulation of these systems gives rise to neuropsychiatric and neurodegenerative disorders with high prevalence and societal burden, collectively termed monoamine neurotransmitter disorders (MNDs). Despite extensive research, the transcriptional regulation of monoaminergic neurotransmitter systems is not fully explored. Interestingly, certain drugs that act on these systems have been shown to modulate central levels of the transcription factor AP-2 beta (AP-2β, gene: TFAP2Β). AP-2β regulates multiple key genes within these systems and thereby its levels correlate with monoamine neurotransmitters measures; yet, its signalling pathways are not well understood. Moreover, although dysregulation of TFAP2Β has been associated with MNDs, the underlying mechanisms for these associations remain elusive. In this context, this review addresses AP-2β, considering its basic structural aspects, regulation and signalling pathways in the controlling of monoaminergic neurotransmitter systems, and possible mechanisms underpinning associated MNDS. It also underscores the significance of AP-2β as a potential diagnostic biomarker and its potential and limitations as a therapeutic target for specific MNDs as well as possible pharmaceutical interventions for targeting it. In essence, this review emphasizes the role of AP-2β as a key regulator of the monoaminergic neurotransmitter systems and its importance for understanding the pathogenesis and improving the management of MNDs.

Background

The monoaminergic neurotransmitter systems, including dopaminergic (DA), adrenergic, noradrenergic (NA) and serotonergic (5-HT) circuitries, regulate a wide range of neurological functions. Dysregulation of these systems is associated with a variety of neuropsychiatric, neurodevelopmental and neurodegenerative disorders, which forms the basis for the so-called monoamine neurotransmitter disorders (MNDs) [1, 2], including obesity, type 2 diabetes (T2D), anxiety, depression, alcoholism, Alzheimer's disease (AD), and neuroblastoma, several of which are among the leading causes of death and disability worldwide [3].

The intricate interplay between monoamine neurotransmitters in the pathophysiology of MNDs poses a substantial challenge for treatment strategies. For instance, dopamine and serotonin, together with norepinephrine, regulate neurological functions ranging from eating behaviour and memory to psychiatric disorders, including aggression, anxiety and depression [4,5,6,7,8,9,10]. On the other hand, current treatment options targeting different proteins in monoaminergic systems are far from optimal. In particular, tricyclic antidepressants (TCA), selective serotonin reuptake inhibitors (SSRIs), serotonin and norepinephrine reuptake inhibitors (SNRIs), monoamine oxidase inhibitors (MAOIs) and catechol-O-methyltransferase inhibitors (COMTIs) are associated with serious adverse and off-target effects as well as interactions with foods and other drugs, resulting in poor patient compliance and treatment outcomes [11,12,13,14,15,16,17,18].

The transcription factor activating protein 2 beta (AP-2β, gene: TFAP2Β) AP-2β has emerged as a vital transcription factor, regulating multiple key genes in monoaminergic neurotransmitter systems such as the serotonin transporter (5-HTT) [19, 20], COMT [21], dopamine-beta-hydroxylase (DBH) [22], vesicular monoamine transporter 2 (VMAT2) [23] and others. Subsequently, AP-2β levels correlate positively with monoamine neurotransmitter indices in the brain [24, 25]. In terms of MNDs, not only is TFAP2Β associated with reduced anxiety [26, 27], alcoholism [28], obesity [29], binge-eating disorder (BED) [30]and T2D [31], but it also plays a neuroprotective role in AD [32] and neuroblastoma [33]. Together with its role in MNDs, the ability of AP-2β to selectively regulate key genes in the monoamine neurotransmitter pathways underline its potential in the early diagnosis and management of associated MNDs [33,34,35,36,37]. This is supported by the fact that certain drugs acting on monoaminergic neurotransmitter systems, phenelzine (MAOI) and citalopram (SNRI), have been shown to modulate the brainstem levels of AP-2β [38, 39]. Therefore, in order to improve current treatment options for MNDs, a better understanding of AP-2β role in the regulation of monoaminergic neurotransmitter systems is essentially required.

Despite extensive studies on AP-2β crucial role in monoaminergic neurotransmitter systems, little is known about its signalling pathways in controlling these systems, making the underlying mechanisms for associated MNDs rather ambiguous. Thus, we review studies on the AP-2β basic structure, its overarching signalling pathways in the control of monoaminergic neurotransmitters and its polymorphisms associated with MNDs. Also, we discuss possible underlying mechanisms for these associations. Most importantly, we explore its usefulness as a diagnostic biomarker for specific MNDs and underline key opportunities and challenges for targeting it.

The AP-2 transcription factor family, structure and transduction mechanisms

The transcription factor activating protein 2 (AP-2) was first cloned in 1987 [40]. At present, five members of the transcription factor AP-2 have been identified: AP-2α, AP-2β, AP-2γ, AP-2δ and AP-2ε, which are encoded by TFAP2A/α, TFAP2Β/β, TFAP2C/γ, TFAP2D/δ, TFAP2E/ε respectively [41]. The AP-2 proteins are highly conserved across species [42] and have differential expression and functions, thereby their mutations give rise to diverse disorders, see Table 1. Structurally, they can form either hetero- or homodimers with a molecular weight of around 50 kDa. They share three regions: a highly conserved C-terminal helix–span–helix homodimerization motif, which starts with a glutamine amino acid; followed by a central basic region, both regions constitute the DNA binding domain; and a less conserved N-terminal proline- and glutamine-rich transactivation region responsible for protein binding domain [42,43,44,45] (Fig. 1).

Table 1 Expression, function, and linked diseases and disorders for transcription factors AP-2
Fig. 1
figure 1

Schematic representation of the possible structure of AP-2β. DNA binding domain is comprised of helix-span-helix and a central basic region, whereas the protein binding domain, also known as the transactivation region, has a special PY motif that is highly conserved. X denotes any amino acid. The picture was influenced by [42, 44, 45, 54], and SwissProt ID: Q92481 & Q61313 and created with Biorender.com

There are two DNA binding sites for AP-2: 2 cis-acting DNA sequences 5′-(G/C)CCCA(G/C)(G/C)(G/C)-3′ and the palindromic sequence 5′-GCCNNNGGC-3′ [27, 46, 47]. Except for AP-2δ, the transactivation domain of AP-2 proteins is characterized by a PY motif (XPPXY) and other highly conserved residues in the protein binding domain [48]. Despite structure similarities (76% homology) between AP-2α & AP-2β [49], their gene mutations give rise to different phenotypes (Table 1). Moreover, AP-2α-null mice exhibit severe craniofacial defects while AP-2β KO mice display massive apoptosis of renal cells and terminal renal failure [49,50,51,52]. This might indicate that they are involved in differential signalling pathways. Although the crystal structure of each AP-2 protein has not yet been established, a highly accurate prediction of their protein structures has been produced by AlphaFold [53] and is available at Uniprot (Q92481).

AP-2 transcription factors are prominent regulators of multiple genes involved in embryonic neural crest development, cell differentiation and haemostasis in a range of tissues but mainly in central and peripheral nervous and urogenital systems [42, 43, 55]. They mediate induction of the target genes in response to two signal-transduction mechanisms: phorbol-ester- and diacylglycerol-activated protein kinase C (PKC) and cAMP-dependent protein kinase A (PKA) [30, 51, 56, 57]. AP-2 transcription factors can act both as transcriptional activators or repressors of the target genes; therefore, we suggest that the name 'transcription factor activating protein 2 (AP-2)' used in the literature is actually a misnomer and should be considered as 'transcription factor trans-regulator protein 2 (TP-2)'.

AP-2β transcription factor

AP-2β was first characterized in 1995 [41]. Two splice variants for AP-2βhave been identified, both can exert transcriptional regulatory activity but have a little different in tissue expressions: one short at 198 aa (22.1 kDa) (UniProt ID X6R4Y8), which is not well studied, and one long at 460 aa (50.5 kDa) (UniProt Q92481) that has been thoroughly investigated [58, 59]. AP-2β is highly expressed in the brain and peripheral neurons, eyes, smooth muscles of ductus arteriosus, skin, bone marrow, adipose and lymphoid tissues, proximal digestive tract, the adrenal medulla of the kidney and across the urogenital system [36, 58, 60]. In addition to its function in monoaminergic neurons, TFAP2Β is also involved in specification of glycinergic and GABAergic interneurons [61,62,63]. TFAP2Β mutations lead to the development of severe disorders such as craniosynostosis, Char Syndrome, dental anomalies, defects in patent ductus arteriosus, terminal renal failure and others [64,65,66,67] (Table 1).

Regulation of AP-2β activity

To better understand its associated MNDs, the mechanisms regulating AP-2β activity should be first articulated. Two key mechanisms have been shown to modulate the DNA-binding and/or transcriptional activities of AP-2β: protein–protein interactions through the protein-binding domain and post-translational modifications employing the DNA-binding domain (Figs. 1 and 2).

Fig. 2
figure 2

Protein interaction network of AP-2β factor. The image was taken from the STRING database [76] where the active interaction source is only experiments and the interaction score is of medium confidence (0.4). The blue line indicates binding, the purple catalysis, and the black reaction. YEATS4: YEATS domain-containing protein 4; KCTD1: potassium channel tetramerization domain 1; MYC: MYC proto-oncogene; TP53, cellular tumour antigen p53; UBC: ubiquitin carrier protein 9; UBE2I, ubiquitin-conjugating enzyme E2 I; SYT4: Synaptotagmin-4; CITED2 & CITED4: Cpb/p300-interacting transactivator 2 & 4

Protein–protein interactions

AP-2β has a specific protein-binding domain, also known as a transactivation domain [44, 45, 67]. Binding to this domain modulates the DNA-binding ability of AP-2β at the adjacent DNA-binding domain [67]. A study by Ding et al. has shown, for example, that the potassium channel tetramerization domain 1 (KCTD1) binds to the N-terminal protein-binding domain of AP-2β and inhibits its transcriptional activity in human cell lines [68]. Another study by Zarelli et al. has reported that in zebrafish, potassium channel tetramerization domain 15 (KCTD15) inhibits the expression of AP-2 [69]. Interestingly, KCTD1 and KCTD15 have remarkably similar amino acid sequences [70], which may explain the similarity of their effects on AP-2β. Additional inhibitors of AP-2β transcriptional activity are the protein kinase D (PKD) [71], which, by phosphorylating AP-2β, inhibits its DNA binding activity, as well as the hypoxia-inducible factor-2alpha (HIF-2α), which negatively regulates the expression AP-2β [72].

The dynamics of protein–protein interaction can also lead to enhanced AP-2β activities. Cpb/p300-interacting transactivator 2 & 4 (CITED2 & CITED4), are coactivators and enhancers of AP-2β-induced transcriptional activation [73, 74]. Additionally, YEATS domain-containing protein 4 (YEATS4, also known as GAS41) augments both, the DNA-binding and the transcriptional activity of AP-2β [75]. For more information about other protein interactions with AP-2β, see Fig. 2.

Post-translational modifications

Post-translational modification is another mechanism that can modulate the transcriptional or DNA-binding activity of AP-2β. For example, the sumoylation of AP-2β by ubiquitin carrier protein 9 (UBC9) has been shown to decrease its transcriptional activity [77]. KCTD15   mentioned earlier can also enhance the activity of AP-2β through sumoylation [78, 79]. Accordingly, KCTD15 can modulate the activity of AP-2β directly (blocking) and post-translationally (activating) (Table 2).

Table 2 The coactivators and suppressors of AP-2β

In summary, different modulators have been shown to regulate the activity of the AP-2β by two mechanisms: the protein–protein interaction through the transactivation domain and post-transcriptional modification through the DNA-binding domain. These two mechanisms could be utilized to pharmacologically modulate AP-2β activity, possibily by targeting these modulators.

AP-2β signalling regulation of monoaminergic neurotransmitter systems

AP-2β is a vital transcription factor for the proper development and function of the monoaminergic neurotransmitter systems [24, 62, 80]. It plays an essential role in the maturation of chromaffin cells, sympathetic neuronal differentiation, and regulation of monoaminergic transmission, including adrenergic, noradrenergic and dopaminergic, and serotonergic transmission, during both development and adulthood [24, 46, 62, 80,81,82]. Several key genes regulating the monoaminergic neurotransmitters' biosynthesis, degradation and synaptic activity have a recognition site for AP-2β in their promoter regions [21,22,23, 27, 83, 84]. Since AP-2β enhances both monoaminergic transmissions, AP-2β levels correlate with monoamine neurotransmitter transmission in the brainstem [24, 25, 62]. This might explain why the central level of AP-2β has been changed after treatment with certain drugs that act on monoaminergic neurotransmitters, such as phenelzine (MAOI) and citalopram (SNRI) [24, 38, 85].

AP-2β regulation of catecholaminergic transmission

AP-2β plays a crucial role in the regulation of catecholamine levels in the brain by altering the expression of several key genes in the catecholaminergic pathway (Fig. 3). AP-2β enhances adrenergic transmission, boosting norepinephrine and epinephrine availability [24, 46, 86,87,88]. It activates the transcription of the genes of catecholamine-synthesizing enzymes, such as tyrosine hydroxylase (TH) [33, 82, 89], dopamine-beta-hydroxylase (DBH) [33, 46, 82, 89, 90] and phenylethanolamine N-methyltransferase (PNMT) [46, 86,87,88] Furthermore, it lowers the genes of catecholamine-degrading enzymes, such as monoamine oxidase-A & B (MAO-A), (MAO-B) [28, 91,92,93] and COMT [21, 92].

Fig. 3
figure 3

The signalling pathway of AP-2β on catecholaminergic transmission where '–' symbolizes repressing and '+ ' activating effect of AP-2β on the transcription of its target genes. AP-2β stimulate the expression of key genes involved in catecholamine synthesis such as TH [33, 82, 89], DBH [33, 46, 82, 89, 90], PNMT [46, 86,87,88] while it lowers genes coding catecholamine degrading enzymes such as MAO [28, 91,92,93] and COMT [21, 92]. AP-2β also repress the transcription of DRD1A through binding to its D1AS1 [94]. AP-2β: transcription activating protein 2 beta; TH: tyrosine hydroxylase; DBH: dopamine-beta-hydroxylase; PNMT: phenylethanolamine N-methyltransferase; MAO: monoamine oxidase; COMT: catechol-O-methyltransferase; HVA: homovanillic acid; VMA: 3-methoxy-4-hydroxymandelic acid; MHPG: 3- methoxy-4-hydroxy-phenylglycol; D1AS1: dopamine receptor 1A silencer 1; DRD1A: dopamine receptor 1A; AC: adenylyl cyclase; and cAMP: cyclic adenosine monophosphate

Furthermore, AP-2β represses the Dopamine receptor D1A (DRD1A) receptor through binding to the D1A silencer 1 (D1AS1) [94]. Blocking this receptor leads to decreased adenylyl cyclase (AC) activity and lowered cyclic adenosine monophosphate (cAMP), resulting in dopamine-related behavioural changes such as reward, cognition and learning [94,95,96] (Fig. 3). DRD1A plays a significant role in the modulation of memory and cognition [97,98,99]; hence, it would be interesting to explore the association of TFAP2B polymorphisms with cognitive functions.

The effect of AP-2β on dopamine turnover is under debate. Damberg et al. have reported that AP-2β is positively correlated with the indices of dopamine metabolites, namely homovanillic acid (HAV) and 3-methoxy-4-hydroxy-phenylgly (MGPG) in rat forebrain [24] but not in CSF of humans [25]. Moreover, DBH enzyme is responsible for converting dopamine into norepinephrine in catecholaminergic neurons [100]; thus, given its transcriptional activation effect on DBH [33, 46, 82, 89, 90], AP-2β might enhance the turnover of dopamine by boosting its conversion into norepinephrine. On the other hand, Schabram et al. have suggested that AP-2β enhances dopamine availability and decreases its turnover through its repressing effect on MAO-A and COMT [92]. In total, it is difficult to conclude the influence of AP-2β on dopamine levels and hence, more investigations in this area are recommended, especially in the AP-2β-associated MNDs where dopamine is a key player, e.g. obesity and anxiety.

AP-2β regulation of serotonergic transmission

AP-2β controls multiple key genes in the serotonergic (5-HT) pathway (Fig. 4) and has been shown to enhance serotonergic turnover and transmission in the brain [24], perhaps by increasing serotonin availability in the synaptic cleft. Two key genes in the serotonergic system have been shown to be negatively regulated by AP-2β: MAO-A [28, 91,92,93] and 5-HTT [19, 20, 101]. 5-HTT transports 5-HT into presynaptic where it is degraded by MAO. Thus, inhibiting 5-HTT and MAO genes by AP-2β may result in increased serotonin levels in the synaptic cleft and serotonergic transmission [24, 25]. In addition, AP-2β has a binding site on tryptophan hydroxylase (TPH), aromatic l‐Amino acid decarboxylase (AADC) as well as rat 5-Htr2 and human 5HT3R [62, 102,103,104,105,106,107]; yet, its regulatory effect on these genes is still unknown. Given the positive correlation between AP-2β and serotonergic activity in the brainstem [24, 25], it is reasonable to suggest that AP-2β may enhance the expression of TPH and AADC as well, although studies are needed to confirm this suggestion.

Fig. 4
figure 4

The effect of AP-2β on serotonergic transmission in the brain. AP-2β enhances the 5-HT levels in the synaptic cleft by increasing the transcription Vmat [81] and repressing 5-HTT [19, 20, 101] and MAO-A [28, 91,92,93]. AP-2β has binding sites on TPH & AADC genes and 5-HT receptor [62, 102,103,104,105,106,107], but its effect on them is unknown. 5-HT: serotonin; Vmat: Drosophila vesicular monoamine transporter; 5-HTT: 5-HT transporters; MAO-A: monoamine oxidase-A; TPH: tryptophan hydroxylase; AADC: aromatic L‐Amino acid decarboxylase; 5-HT receptor: serotonin receptor

The vesicular monoamine transporter 2 gene (VMAT2), a key regulator of the monoamine neurotransmitters' availability in the synaptic cleft, has a putative AP-2β binding site in its promotor region [23]. However, the regulatory effect of AP-2β on this gene has not been fully explored. In an attempt to explore this, using Drosophila melanogaster, our group has shown that the Drosophila analogue of TFAP2Β, TfAP-2, positively regulates the expression levels of Drosophila VMAT2, Vmat, and activates octopaminergic neurons (analogous to human noradrenergic neurons) [81]. Considering that TFAP2Β is conserved in Drosophila (TfAP-2) and there is a high homology between Drosophila Vmat and human VMAT2 (weighted score 14/15) [108, 109], AP-2β might also activate VMAT2 in humans. Consistent with this suggestion is the observations that, similar to AP-2β [24, 25], activation of VMAT2 enhances monoaminergic transmission in the synaptic cleft, not to mention that VMAT2 dysregulation results in disorders that are also associated with TFAP2Β, namely depression [110], AD [111], alcoholism [112] and obesity [113, 114]. Altogether, more research on AP-2β effects on human VMAT2, for instance in human neuronal cell culture, are warranted.

Taken together, AP-2β can modulate both arms of monoaminergic neurotransmitter systems, by controlling the key genes in catecholaminergic and serotonergic transmission, indicating its role as a key regulator of monoaminergic neurotransmitter systems and its importance for better understanding and management of MNDS.

TFAP2Β polymorphisms

The TFAP2Β gene is located on chromosome 6p12-p21, and it has eleven axons with a size of 29,910 bases [115, 116]. TFAP2B has many polymorphic regions that affect mostly its transcriptional activity rather than its protein structure [29, 34, 117]. This in turn influences the expression of its target genes in the monoaminergic neurotransmitter systems. Consequently, TFAP2Β functional polymorphisms have been associated with different MNDs. To date, three functional polymorphisms of TFAP2Β are known to be associated with MNDs. One is located in intron 1, in which polymorphic regions (SNP at intron 1 + 774T/G and a nearby variable number tandem repeat, VNTR, allele), have been shown to postively regulate TFAP2B transcriptional activity, enhancing  its expression. Subjects who carry the disease-related alleles (S-allele: T-nucleotide for SNP, nine repeats for VNTR) show higher expression of TFAP2Β, which is associated with a higher risk for T2D and a lower risk for depression [34, 36, 102, 118].

The second polymorphism is located in intron 2 in a polymorphic region [CAAA] close to the 3`-splice site of exon 2 between nucleotides 12593 and 12612 [27, 51]. Repeats of this sequence in variable regions suggest its important role in functional polymorphisms [25, 27, 60]. Interestingly, the 5-repeat alleles of indel intron 2 of TFAP2B have been shown to increase the expression of TFAP2Β [19]. The intron 2 polymorphism is associated with adiposity, neonatal temperament and anxiety-related personality traits [19, 26, 27, 29]. Strikingly, there is significant linkage disequilibrium between the polymorphisms of intron 1 and intron 2 [27, 29, 119], indicating that their association is not by chance. This suggests that the associations observed with intron 2 polymorphisms are most likely a result of the functional polymorphism in intron 1 [28].

The third TFAP2Β polymorphism, rs987237, is located in intron 3 and has three different genotypes, AA, AG and GG that are associated with obesity, BMI, waist circumference and differential effects on weight loss [120,121,122,123]. Stocks et al. have suggested that the TFAP2Β rs987237 variant may be linked to enhanced activity of this transcription factor since it is also in complete linkage disequilibrium with intron 1 [120].

Overall, three TFAP2Β polymorphisms have been shown to mostly enhance its transcription activity. This in turn affects the expression of the key genes in the monoaminergic neurotransmitter systems that have putative binding sites for AP-2β.

Monoamine neurotransmitter disorders associated with TFAP2Β dysregulation and possible underlying mechanisms

Obesity and type 2 diabetes (T2D)

Studies have shown that TFAP2Β polymorphisms of intron 1,2 & 3 that lead to higher expression of TFAP2Β, are associated with reward fixation, insulin resistance, T2D and adiposity, lipid droplet biogenesis as well as eating disorders [29, 34, 36, 124,125,126,127,128]. GWAS studies have indicated an association of TFAP2Β rs987237 within intron 3 with obesity, BMI and waist circumference in women [120, 129, 130]. There is, however, a debate whether rs987237 is associated with improved weight loss, depending on the genotype (AA, GG or AG) [120,121,122] and the type of diet (high/low-fat diet or high/low protein diet) [120, 131].

The mechanisms underlying AP-2β-induced insulin resistance and adiposity are rather unclear. Several studies have attributed the role of AP-2β in insulin resistance and adiposity to its regulatory effect on genes that encode adipocytokines, which have AP-2β binding sites in their promoters. For instance, TFAP2Β overexpression has been shown to inhibit the expression of adiponectin and leptin but also enhance the expression of interleukin 6 (IL-6), monocyte chemoattractant protein-1 (MCP) and tumour necrosis factor-alpha (TNF-alpha) [34, 124, 132,133,134,135]. However, Tsukada et al. have demonstrated that overexpressed TFAP2Β has no effect on most adipocytokines like adiponectin, leptin and IL-6 [34]. Additionally, a study by Nordquist et al. have shown no association of adiponectin levels to insulin resistance or to the TFAP2Β polymorphism [29]. Furthermore, AP-2β has been reported to enhance glucose uptake (enhancing the translocation of the glucose transport 4, GLUT4) but also reduce insulin sensitivity through both, repressing the expression of insulin receptor substrate 1 (IRS-I) and lipid accumulation [29, 31, 36, 133, 134]. The enhanced glucose uptake effect, however, is not in line with the inhibitory effect of AP-2β on adiponectin and leptin mentioned above since higher adiponectin and leptin have been shown to enhance glucose uptake [136, 137]. Given such discrepancies, it is reasonable to suggest another pathway by which AP-2β contributes to obesity and T2D, possibly through monoaminergic neurotransmitter systems.

Many research studies have revealed that polymorphisms or overexpression of TFAP2Β contribute to eating disorder-related behaviour through catecholamine-induced orexigenic signals [78, 79]. In terms of glucose uptake and insulin resistance, AP-2β augments norepinephrine availability [24, 25], and elevated norepinephrine has been reported to increase glucose uptake and transport in adipose tissue [138, 139]. Moreover, AP-2β increases both norepinephrine and epinephrine levels [24, 25], both of which can contribute to insulin resistance and obesity [140,141,142,143,144,145]. Furthermore, AP-2β upregulates PNMT [46, 86,87,88], and upregulated PNMT has been associated with elevated epinephrine and reduced circulating leptin levels [146], both of which can give rise to obesity [141, 142, 146].

The association of TFAP2Β with obesity-related disorders might also be linked to its repression effect on 5-HTT. First, 5-HTT re-uptakes 5-HT from the synaptic cleft into the presynaptic neurons. As we mentioned previously, AP-2β represses the expression of 5-HTT [19, 20, 101] and 5-HTT gene promotor methylation is associated with reward and eating behaviour as well as human obesity [147] and suppression of 5-HTT, by hypermethylation, in humans is associated with a high prevalence of obesity [148].

The genetic interaction between TFAP2Β and KCTD15, an obesity-linked gene, is also likely to positively contribute to AP-2β-induced obesity and metabolic dysfunction. In this regard, our group has recently shown that in Drosophila, the homologue of KCTD15, Twz, genetically interacts with Drosophila TfAP-2 to regulate octopaminergic (human noradrenergic) signalling. We have further demonstrated that mouse TFAP2β and Kctd15 directly interact with Ube2i, a mouse sumoylation enzyme, to induce consummatory behaviour [79]. Strikingly, consistent with these findings is a recent study by Gamero-Villarroel et al. that has identified the genetic interaction between KCTD15 and TFAP2Β in individuals with eating disorders [78]. The same study has suggested that after sumoylation of TFAP2Β by KCTD15, a post-translational effect is initiated by E1‐3 enzymes and ATP, resulting in elevated catecholamine levels and induction of feeding behaviour [78].

Alcoholism

That dysregulation of monoamine neurotransmitters contributes to the development of alcoholism is well known. Reports have indicated that TFAP2Β polymorphism in the intron 1, which can enhance AP-2β transcription, is associated with severe alcoholism in women [28]. This association has been linked to the effect of AP-2β on several targets in the monoaminergic system. AP-2β lowers the levels of MAO and lowered levels of MAO are linked to alcoholism [28, 149,150,151]. AP-2β activation of dopamine-beta-hydroxylase (DBH) [82] may also contribute to alcoholism, since in alcohol-dependent persons, DBH is hypomethylated and its enzyme is more active, resulting in a reduction of dopamine levels [152, 153]. However, inhibition of DRD1 or DRD1 KO mice has been shown to reduce alcohol-seeking behaviour [154,155,156,157], suggesting that AP-2β-induced alcohol intake might be independent of its repressing effect on DRD1 [94].

Moreover, AP-2β-enhanced serotonergic transmission may also contribute to alcohol abuse, perhaps through inhibition of 5-HTT [19, 20, 101] and lowering MAO [28, 91,92,93]. In agreement with this notion, increased serotonin levels or 5-HTT KO mice have been found to trigger alcoholism [158,159,160], and MAO-A methylation is associated with alcoholism in women [161]. Furthermore, alcoholics have higher levels of both, the transcript and protein of tryptophan hydroxylase, TPH [162], the rate-liming enzyme in serotonin synthesis, which has a binding site for AP-2β in its promotor [107]. Increased levels of TPH have been reported to enhance serotonin levels [163].

Anxiety and depression

TFAP2Β intron 2 polymorphisms are associated with low anxiety [26, 27, 93], and its intron 1 polymorphism, which enhances its expression, protects against the risk of depression in patients with attention deficit hyperactivity disorder [102]. However, when psychosocial adversity is considered, individuals with TFAP2Β intron 2 polymorphisms who are homozygous for the short TFAP2Β allele exhibited higher depression scores [117].

This association might be ascribed to the AP-2β regulatory effect on the monoaminergic neurotransmitters in the brain. Mechanistically, AP-2β inhibits 5-HTT expression [19, 20, 101] and inhibition of 5-HTT, by SSRIs for instance, leads to increased serotonin, which alleviates anxiety and depression [164]. Moreover, AP-2β reduces MAO levels [28, 91,92,93], which may also contribute to reduced anxiety and depression. In agreement with this possibility, deficiency of MAO-A or MAO-B leads to reduced anxiety-like behaviour in mice, as well as inhibition of MAO-A reduces depression in mice, and likewise, drugs that inhibit MAO-B reduce depression as well [165, 166]. Interestingly, MAO inhibitors enhance the levels of noradrenaline and serotonin levels in the brain to alleviate anxiety [167] and depression [168]. In consonance with these effects, AP-2β enhances noradrenaline and serotonin transmission [24], both of which have been reported to exert anxiolytic and antidepressant effects [9, 169,170,171].

Since certain antidepressant/anxiolytic drugs, e.g. citalopram (SSRI) and imipramine (TCA), have been reported to modulate the levels of AP-2β in the brainstem of the rat [24, 38, 84], probably due to its transcriptional regulation of the key targets in monoaminergic neurotransmitter systems, future clinical  anxiolytic/antidepressant drug development should monitor the level of AP-2β for better management and/or prevention of depression and anxiety.

Antisocial behaviour

Antisocial behaviour in children and adolescents refers to a heterogeneous set of actions outside the norms of society, including aggression, impulsive behaviour and criminal acts, which are linked to monoaminergic neurotransmitter systems [172,173,174,175]. TFAP2Β has been associated with aggression in fruit flies and humans, as well as with general antisocial behaviour in humans [27, 28, 81, 176]. However, the TFAP2Β signalling mechanisms that underlie such association have not been fully elucidated. The repressing effect of AP-2β on monoamine-degrading enzymes, such as COMT and MAO might explain such an association since several reports have revealed that lowered expression of MAO-A, or MAO-A KO mice, as well as lower activity of COMT or COMT-deficiency in mice are associated with higher aggression [177,178,179,180,181,182]. Furthermore, activation of DBH expression by AP-2β may also play a part in provoking high aggression. Activation of DBH enhances the conversion of dopamine to adrenaline and it has been reported that higher adrenergic signalling provokes aggression [183], and DBH KO mice have lower levels of aggression [184], all linking AP-2β-mediated higher aggression to enhanced adrenergic signalling.

Nevertheless, the AP-2β association with higher aggression might be independent of its enhancement effect on serotonergic signalling, specifically, its repressing effect on 5-HTT. This is because several preclinical and clinical studies have indicated that high levels of serotonin and lower expression or blocking of 5-HTT or knocking out 5-HTT in mice lead to reduced aggression outbursts and violent behaviour [185,186,187,188], an effect opposite to that of AP-2β. It would be interesting to investigate the association of TFAP2Β polymorphisms and their interaction with key monoamine neurotransmitter genes to aggression and antisocial behaviour.

Alzheimer's disease (AD)

While genetic variants of TFAP2Β have been suggested to play a role in resilience to AD [189], increased AP-2β has been shown to bestow a neuroprotective effect in AD due to the AP-2β-enhancing effect on the expression of apolipoprotein E (apoE), an important protective protein in AD pathogenesis [32, 190]. The effects of AP-2β on catecholamines might also contribute to its protective effect in AD. Firstly, lowered catecholamine levels contribute to the development and pathogenesis of AD [191,192,193] and, by the same token, AP-2β has been shown to increase catecholamine signalling in the brain [24, 46, 86,87,88]. AD is also associated with lower levels of DBH and PNMT [194,195,196,197,198], which both are activated by AP-2β [46, 86,87,88]. In addition, COMT and MAO-B levels are higher in AD [192, 199,200,201], and both are lowered by AP-2β [21, 92, 93]. Supporting this notion, COMT and MAO inhibitors have been repurposed for the treatment of AD [202,203,204]. Secondly, AP-2β enhances serotonin activity in the brain which might add further protective effects in AD. Consistent with this extrapolation is the observation that reduced serotonin levels in the brain can enhance the risk for AD [205], providing the rationale for SSRI use to delay the onset of AD [206]. Taken together, it might be suggested that, through its enhancing catecholaminergic and serotonergic activities, AP-2β may exert a protective role in AD. Thus, elucidating the association of TFAP2Β polymorphisms and their interactions with COMT, MAO, to AD could unveil a potential biomarker for early diagnosis and management of AD.

Neuroblastoma

Transcription factors serve as essential regulators of cell development, proliferation and differentiation; consequently, dysregulation of them brings about oncogenic transformation and cancers [207, 208]. Neuroblastoma is an embryonal pediatric malignant tumour originating from the sympathetic nervous system and characterized by extremely low noradrenergic neuronal differentiation [33]. However, the molecular mechanisms underlying lowered neuronal differentiation in neuroblastoma are still under investigation.

Recent studies have revealed an important role of AP-2β in the pathogenesis and progression of neuroblastoma [33, 37, 209, 210]. Thorell et al. have identified TFAP2Β as a potential tumour suppressor gene in neuroblastoma [210]. Ikram et al. have also indicated that low AP-2β expression results in lower noradrenergic neuronal differentiation and is thereby significantly associated with poor prognostic markers and unfavourable patient outcomes [33]. In contrast, induction of AP-2β expression has been found to impair tumour cell proliferation and slow tumour progression by enhancing both differentiation of noradrenergic neurons as well as noradrenergic signalling through increased expression of TH and DBH [33]. In addition to its effects on sympathetic neurons, AP-2β has been demonstrated to enhance retinoic acid (RA) responsiveness, which potentiates neuronal differentiation and therefore is used in neuroblastoma therapy [33].

Altogether, AP-2β is associated with certain MNDS, although the underlying mechanisms are not fully elucidated. As we mentioned above, the underlying mechanisms for this association might be linked to AP-2β regulatory effects on key genes of monoaminergic neurotransmitters. However, further preclinical studies investigating the molecular mechanisms underlying AP-2β associated MNDs, e.g. manipulating the expression of TFAP2B in neuronal cell culture or rats’ models of MNDs and examining the levels of the key enzymes of monoaminergic neurotransmission, are warranted.

AP-2β as a biomarker and a potential therapeutic target

AP-2β as a biomarker

Transcription factors are currently widely used as diagnostic biomarkers for the early detection of several diseases [211,212,213]. A wide range of brain-related disorders and cancers are accompanied by alterations in the levels and activity of TFAP2Β/AP-2β, which underscores its diagnostic importance for such diseases. TFAP2Β overexpression, for instance, has been implicated not only as a protective or favourable prognostic factor in several cancers like breast, renal cell, cervical and endometrial cancers but also as a poor prognostic factor in thyroid cancer and lung adenocarcinoma [214,215,216,217,218,219,220,221,222,223,224,225] (See Table 1).

Along the same line, AP-2β might also constitute a diagnostic biomarker for other MNDs. For example, while TFAP2Β/AP-2β overexpression has already been suggested as a favourable prognostic marker in neuroblastoma[33, 210], it might further be considered as a biomarker for other MNDs where TFAP2Β/AP-2β overexpression is implicated, such as alcoholism, obesity, T2D and aggression. Nevertheless, clinical studies are needed to address its specificity and characterize its validity as a biomarker for the early diagnosis of these disorders.

AP-2β as a potential therapeutic target

In the last decade, targeting transcription factors with selective therapeutic agents has gained growing interest because transcription factors act as on/off switches of gene expression, a process that leads to neurological disorders and cancers when disrupted. In this context, clinical reports have highlighted the potential of AP-2β as a therapeutic target for specific cancers such as lung adenocarcinoma [216], renal cell tumorigenesis [220], and breast [221] and endometrial cancers [217].

TFAP2Β/AP-2β might also be considered an interesting therapeutic target for the management of specific MNDs, where its synergistic effects on multiple targets may be beneficial for the treatment or prevention of complex diseases. This concerns specifically the neurometabolic disorders, such as obesity and type 2 diabetes, that are characterised by widespread systemic alterations through diverse factors including, behavioural, neural, hormonal, adipose and intestinal along with the involvement of different mediators such as monoamine neurotransmitters, insulin and adipocytokines. In this regard, researchers have emphasized the importance of AP-2β as a promising drug target for the prevention and treatment of obesity and T2D [34,35,36]. AP-2β can target multiple features of obesity and T2D centrally and peripherally by modulating key-obesity linked genes such as IRS-1, GLUT4, adipocytokines related genes, as well as catecholaminergic and serotonergic genes involved in reward, consummatory behaviour and insulin resistance (Table 3). Some of these genes are, intriguingly, recognized as therapeutic targets of several drugs used for obesity-linked diseases.

Table 3 Summary of the mechanisms underlying AP-2β effects, and potential therapeutic interventions for specific monoamine neurotransmitter disorders (MNDs)

A second possible application of AP-2β as a therapeutic target is in the treatment of neuroblastoma [37], where the benefits of treatment success could outweigh the possible side effects. As mentioned previously, lower expression of TFAP2Β is associated with a poor prognosis of neuroblastoma since patients whose tumour cells have lowered TFAP2Β showed poor treatment outcomes [33]. In contrast, elevated TFAP2Β has been reported to improve patient outcomes [33] by enhancing noradrenergic neuronal differentiation through different target genes in monoaminergic transmission [33, 210] and subsequently repress the progression of neuroblastoma. Most importantly, induction of TFAP2Β expression by tetracycline has already been successfully employed to impair tumour cell proliferation and slow neuroblastoma progression [33]. Collectively, boosting AP-2β levels might represent a potential therapeutic approach, perhaps in combination with other anti-neuroblastoma therapies, to treat neuroblastoma, possibly through a localized drug delivery, which could minimize peripheral undesired effects. This is supported by the fact that Trichostatin A, a potential drug used for neuroblastoma, has been shown to augment monoamine pools by inhibiting COMT and MAO-A genes [226], a mechanism similar to that of AP-2β.

Like other drug targets at the transcription level, off-target effects could pose a substantial challenge. AP-2β has been reported to modulate multiple key genes within and outside the monoaminergic neurotransmitter systems, suggesting off-target effects. Consequently, such ubiquitous off-target effects might limit its potential as a therapeutic target due to the lack of specificity and the risk of adverse effects.

Targeting AP-2β (druggability), challenges and future insights

Until recently, transcription factors had been considered very challenging targets (undruggable) since transcription factors lack obvious druggable pockets and the transcription process is carried out in the nucleus; therefore, the therapeutic agent should have the appropriate physicochemical properties to cross many biological barriers and reach the nucleus with sufficient concentration. Moreover, many crucial components involved in the transcription process do not have the enzymatic activity adequate for pharmaco-chemical interventions [227, 228].

So far, manipulation of transcription factor activity at protein–protein interaction levels has been successfully implemented and a few drugs have been approved for various disorders [227, 229]. As mentioned previously, several proteins have been reported to modulate the levels and activity of AP-2β (see Table 2). Such modulator proteins can be targeted to regulate TFAP2Β/AP-2β at different levels: at transcription, post-translation or the DNA-binding site (Fig. 5). Yet, potency and selectivity may pose a substantial issue because targeting these proteins can elicit a chain of inadvertent adverse effects since they are also involved in diverse biological processes and regulate other AP-2 transcription factors [42].

Fig. 5
figure 5

Possible sites and targets that can be potentially exploited to modulate AP-2β activities and/or levels. One proposal for modulating AP-2β is through protein–protein interactions whereby co-activators/suppressors bind to the transactivation domain and modify AP-2β transcription activity and DNA-binding activities, as indicated within the yellow square. Inducing degradation of AP-2β could be achieved through designing peptide inhibitors binding selectively to its transactivation domain to form a non-functional complex or by enhancing PKD phosphorylation of AP-2β [71] or by developing specific monoclonal antibodies that can bind and inactivate AP-2β. By contrast, enhancing AP-2β activity could be feasible by designing artificial transcription factor analogues (TFA) that can act as AP-2β agonists. Some monoaminergic drugs, such as phenelzine and citalopram, also have been shown to alter the brain levels of AP-2β [38, 39]] while tetracycline induces its gene expression[33]. KCTD1 & KCTD15: potassium channel tetramerization domain 1 & 15; UBC9: ubiquitin carrier protein 9; HIF-2α: hypoxia-inducible factor-2alpha; YEATS4: YEATS domain-containing protein 4; CITED2 & 4: Cpb/p300-interacting transactivator 2 & 4; PKD: the protein kinase D; DAG: diacylglycerol. The Figure was created with BioRender.com

Despite these challenges, in the last decade, advances in pharmacological interventions have facilitated the druggability of transcription factors by specifically modulating their DNA-binding and transcription regulation activities at specific pockets. Interestingly, such pharmacological strategies have been successfully applied preclinically [228, 230,231,232,233,234] and clinically [235, 236], and at least 10% of drugs obtained FDA approval [208, 228, 237, 238]. Moreover, a recent article in Nature Reviews Drug Discovery by Henley et al. has discussed the renewed interest in the advent of pharmacological interventions for targeting transcription factors [208]. Currently, about ten drugs acting on transcription factors are in clinical trials [208], indicating a driving trend toward targeting transcription factors.

For example, inducing protein degradation of the transcription factor can be driven by exploiting its transactivation domain. Binding to this part with a specific peptide can form a non-functional complex for degradation. Such technology has facilitated abolishing of transcription factor activity by designing peptide inhibitors, also known as peptide therapeutics [233] that binds to the transactivation domain of the transcription factor and thereby hinder its interactions with other proteins and ultimately induce its degradation (Fig. 5). An interesting example in this context is YK-4-279, which peptide inhibitor that binds to the oncogenic transcription factor EWS-FLI1 to inhibit its activities. YK-4-279 is used to treat Ewing sarcomas [231, 239].

In parallel, providing the basic knowledge of its domain structure and binding sites, direct inhibition of AP-2β, by peptide inhibitors capable of selectively binding to its transactivation domain and inducing its degradation could be attractively applicable. Such specific peptide inhibitors could interfere with the dynamic of AP-2β protein–protein interactions and subsequently block its transcriptional activity on the target genes. This represents an interesting therapeutic approach for the management of obesity and related neurometabolic disorders where higher AP-2β levels are implicated.

On the other hand, mimicking transcription factor activity has been made feasible, especially in cancer and neurometabolic disorders. Many artificial transcription factors, also called transcription factor analogues (TFAs), have been successfully developed for several cancers and loss-of-function mutations to restore the overall functions of the transcription factors [231]. Dimethyl fumarate (DMF), for example, is an NRF2 activator, which is FDA-approved for multiple sclerosis (MS) [237]. In a similar vein, induction of AP-2β-related transcriptional effects on its target genes could be achieved by designing an AP-2β analogue with sufficient selectivity and potency (Fig. 5). Such AP-2β agonists could mediate effects similar to that of AP-2β and thereby be used in disorders where higher AP-2β is therapeutically advantageous such as in neuroblastoma. Nevertheless, full characterization of the protein crystal structure of AP-2β is a prerequisite for developing such therapeutic agents.

Concluding remarks

AP-2β is a central regulator of monoamine neurotransmitters and its dysregulation is associated with MNDs. The underlying mechanisms for its associated MNDs could be linked to its regulatory role in monoaminergic transmission, yet more studies are recommended. For example, knockout/overexpression of TFAP2B in rat models of MNDs and examining the expression of the key monoaminergic genes can help clarify the molecular pathogenesis of the associated MNDs. Moreover, the molecular functions of AP-2β within the monoaminergic systems underline its importance as a promising biomarker for the early diagnosis of relevant MNDs. Given its effect on multiple targets within and outside monoaminergic systems, AP-2β could be considered a dirty drug target due to the odds of having adverse effects. Nevertheless, several dirty drugs that aim for multiple targets at once are clinically effective in the treatment of complex diseases [240, 241], which might also underscore the therapeutic potential of the AP-2β for complex MNDs like obesity and neuroblastoma. Future drug development targeting monoaminergic systems could take advantage of monitoring the AP-2β levels during clinical studies for better management and treatment of MNDs.

Availability of data and materials

Not applicable.

Abbreviations

CNS:

Central nerve system

PNS:

Prepheral nerve system

MNDs:

Monoamine neurotransmitter disorders

DA:

Dopamine

NA:

Noradrenaline

5-HT:

Serotonin

T2D:

Type 2 diabetes

AD:

Alzheimer's disease

TCA:

Tricyclic antidepressants

SSRIs:

Selective serotonin reuptake inhibitors

SNRIs:

Serotonin and norepinephrine reuptake inhibitors

MAOIs:

Monoamine oxidase inhibitors

COMTIs:

Catechol-O-methyltransferase inhibitors

AP-2β:

Transcription factor activating protein 2 beta

5-HTT :

Serotonin transporter

DBH :

Dopamine-beta-hydroxylase

VMAT2 :

Vesicular monoamine transporter 2

WWOX :

WW Domain Containing Oxidoreductase

GREB1 :

Growth Regulating Estrogen Receptor Binding 1

 CDH2 :

Cadherin-2

HPSE :

Heparanase

IGSF11:

Immunoglobulin Superfamily Member 11

BOFS:

Branchio-oculo-facial syndrome

COL2A1:

Core promoter of type II collagen

HMOX1 :

Heme Oxygenase 1

IRS-1:

Insulin resistance substrate 1

PKC:

Protein kinase C

PKA:

CAMP-dependent protein kinase A

KCTD1:

The potassium channel tetramerization domain 1

KCTD15:

Potassium channel tetramerization domain 15

PKD:

Protein kinase D

HIF-2α:

Hypoxia-inducible factor-2alpha

CITED2:

Cpb/p300-interacting transactivator 2

CITED4:

Cpb/p300-interacting transactivator 4

YEATS4:

YEATS Domain-containing protein 4

MYC:

MYC Proto-Oncogene, bHLH transcription factor

TP53:

Cellular tumor antigen p53

UBC:

Ubiquitin carrier protein 9

UBE2I:

Ubiquitin conjugating enzyme E2 I

SYT4:

Synaptotagmin-4

TH :

Tyrosine hydroxylase

PNMT :

Phenylethanolamine N-methyltransferase

MAO-A :

Monoamine oxidase-A

DRD1A :

Dopamine receptor D1A

AC:

Adenylyl cyclase

cAMP:

Cyclic adenosine monophosphate

HAV:

Homovanillic acid

MGPG:

3- Methoxy-4-hydroxy-phenylgly

TPH :

Tryptophan hydroxylase

AADC :

Aromatic l‐Amino acid decarboxylase

VMAT2 :

Vesicular monoamine transporter 2 gene

IL-6:

Interleukin 6

MCP:

Monocyte chemoattractant protein-1

TNF-alpha:

Tumour necrosis factor-alpha

IRS-I:

Insulin receptor substrate 1

GLUT4:

Glucose transport 4

apoE:

Apolipoprotein E

TFAs:

Transcription factor analogues

DMF:

Dimethyl fumarate

References

  1. Kurian MA, Gissen P, Smith M, Heales SJR, Clayton PT. The monoamine neurotransmitter disorders: an expanding range of neurological syndromes. Lancet Neurol. 2011;10:721–33.

    Article  CAS  PubMed  Google Scholar 

  2. Ng J, Papandreou A, Heales SJ, Kurian MA. Monoamine neurotransmitter disorders—clinical advances and future perspectives. Nat Rev Neurol. 2015;11:567–84.

    Article  CAS  PubMed  Google Scholar 

  3. World Health Organization (WHO). Leading causes of death and disability worldwide: 2000–2019. https://www.who.int/news/item/09-12-2020-who-reveals-leading-causes-of-death-and-disability-worldwide-2000-2019. 2020.

  4. Hauser TU, Eldar E, Purg N, Moutoussis M, Dolan RJ. Distinct roles of dopamine and noradrenaline in incidental memory. J Neurosci. 2019;39:7715–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Narvaes R, de Almeida RMM. Aggressive behavior and three neurotransmitters: dopamine, GABA, and serotonin—a review of the last 10 years. Psychol Neurosci. 2014;7:601–7.

    Article  Google Scholar 

  6. Seo D, Patrick CJ, Kennealy PJ. Role of serotonin and dopamine system interactions in the neurobiology of impulsive aggression and its comorbidity with other clinical disorders. Aggress Violent Beh. 2008;13:383–95.

    Article  Google Scholar 

  7. Hull EM, Muschamp JW, Sato S. Dopamine and serotonin: Influences on male sexual behavior. Physiol Behav. 2004;83:291–307.

    Article  CAS  PubMed  Google Scholar 

  8. Moret C, Briley M. The importance of norepinephrine in depression. Neuropsychiatric Dis Treat. 2011;7:9–13.

    CAS  Google Scholar 

  9. Montoya A, Bruins R, Katzman MA, Blier P. The noradrenergic paradox: implications in the management of depression and anxiety. Neuropsychiatr Dis Treat. 2016;12:541–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. González-Burgos I, Feria-Velasco A. Serotonin/dopamine interaction in memory formation. Prog Brain Res. 2008;172:603–23.

    Article  PubMed  CAS  Google Scholar 

  11. Walker AJ, Card T, Bates TE, Muir K. Tricyclic antidepressants and the incidence of certain cancers: a study using the GPRD. Br J Cancer. 2011. https://doi.org/10.1038/sj.bjc.6605996.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Yamada M, Yasuhara H. Clinical pharmacology of MAO inhibitors: safety and future. Neurotoxicology. 2004. https://doi.org/10.1016/S0161-813X(03)00097-4.

    Article  PubMed  Google Scholar 

  13. Von Kleist L, Michaelis S, Bartho K, Graebner O, Schlief M, Dreger M, et al. Identification of Potential off-target toxicity liabilities of catechol-O-methyltransferase inhibitors by differential competition capture compound mass spectrometry. J Med Chem. 2016. https://doi.org/10.1021/acs.jmedchem.5b01970.

    Article  Google Scholar 

  14. Scotton WJ, Hill LJ, Williams AC, Barnes NM. Serotonin syndrome: pathophysiology, clinical features, management, and potential future directions. Int J Tryptophan Res. 2019;12:117864691987392–117864691987392.

    Article  Google Scholar 

  15. Low Y, Setia S, Lima G. Drug–drug interactions involving antidepressants: focus on desvenlafaxine. Neuropsychiatr Dis Treat. 2018;14:567–80.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Sansone RA, Sansone LA. Antidepressant adherence: are patients taking their medications? Innov Clin Neurosci. 2012;9:41–6.

    PubMed  PubMed Central  Google Scholar 

  17. Ramsay RR, De Deurwaerdère P, Di Giovanni G. Updating neuropathology and neuropharmacology of monoaminergic systems. Br J Pharmacol. 2016;173:2065–2065.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Cohen LJ, Sclar DA. MAOIs: issues in treatment adherence and rates of treatment failure. J Clin Psychiatry. 2013;74:26367.

    Article  Google Scholar 

  19. Ivorra JL, D’Souza UM, Jover M, Arranz MJ, Williams BP, Henry SE, et al. Association between neonatal temperament, SLC6A4, DRD4and a functional polymorphism located in TFAP2B. Genes Brain Behav. 2011;10:570–8.

    Article  CAS  PubMed  Google Scholar 

  20. Hu XZ, Lipsky RH, Zhu G, Akhtar LA, Taubman J, Greenberg BD, et al. Serotonin transporter promoter gain-of-function genotypes are linked to obsessive-compulsive disorder. Am J Hum Genet. 2006;78:815–26.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Tenhunen J. Characterization of the rat catechol-O-methyltransferase gene proximal promoter: identification of a nuclear protein-DNA interaction that contributes to the tissue-specific regulation. Larchmont: Mary Ann Liebert, Inc.; 1996.

    Google Scholar 

  22. Greco D, Zellmer E, Zhang Z, Lewis E. Transcription factor AP-2 regulates expression of the dopamine beta-hydroxylase gene. J Neurochem. 1995;65:510–6.

    Article  CAS  PubMed  Google Scholar 

  23. Uhl GR, Li S, Takahashi N, Itokawa K, Lin Z, Hazama M, et al. The VMAT2 gene in mice and humans: amphetamine responses, locomotion, cardiac arrhythmias, aging, and vulnerability to dopaminergic toxins. FASEB J. 2000;14:2459–65.

    Article  CAS  PubMed  Google Scholar 

  24. Damberg M, Eller M, Tõnissaar M, Oreland L, Harro J. Levels of transcription factors AP-2α and AP-2β in the brainstem are correlated to monoamine turnover in the rat forebrain. Neurosci Lett. 2001;313:102–4.

    Article  CAS  PubMed  Google Scholar 

  25. Damberg M, Berggård C, Farde L, Sedvall GC, Jönsson EG. Transcription factor AP-2β genotype, striatal dopamine D2 receptor density and cerebrospinal fluid monoamine metabolite concentrations in humans. J Neural Transm. 2004. https://doi.org/10.1007/s00702-003-0097-4.

    Article  PubMed  Google Scholar 

  26. Damberg M, Berggård C, Mattila-Evenden M, Rylander G, Forslund K, Garpenstrand H, et al. Transcription factor AP-2β genotype associated with anxiety-related personality traits in women: a replication study. Neuropsychobiology. 2003;48:169–75.

    Article  CAS  PubMed  Google Scholar 

  27. Damberg M, Garpenstrand H, Alfredsson J, Ekblom J, Forslund K, Rylander G, et al. A polymorphic region in the human transcription factor AP-2β gene is associated with specific personality traits. Mol Psychiatry. 2000;5:220–4.

    Article  CAS  PubMed  Google Scholar 

  28. Nordquist N, Göktürk C, Comasco E, Nilsson KW, Oreland L, Hallman J. Transcription factor AP2 beta involved in severe female alcoholism. Brain Res. 2009;1305:S20–6.

    Article  CAS  PubMed  Google Scholar 

  29. Nordquist N, Göktürk C, Comasco E, Eensoo D, Merenäkk L, Veidebaum T, et al. The transcription factor TFAP2B is associated with insulin resistance and adiposity in healthy adolescents. Obesity. 2009;17:1762–7.

    Article  CAS  PubMed  Google Scholar 

  30. Damberg M, Garpenstrand H, Hallman J, Oreland L. Genetic mechanisms of behavior—don’t forget about the transcription factors. Mol Psychiatry. 2001;6:503–10.

    Article  CAS  PubMed  Google Scholar 

  31. Tao Y, Maegawa H, Ugi S, Ikeda K, Nagai Y, Egawa K, et al. The Transcription factor AP-2β causes cell enlargement and insulin resistance in 3T3-L1 adipocytes. Endocrinology. 2006;147:1685–96.

    Article  CAS  PubMed  Google Scholar 

  32. Rossello XS, Igbavboa U, Weisman GA, Sun GY, Wood WG. AP-2β regulates amyloid beta-protein stimulation of apolipoprotein e transcription in astrocytes. Brain Res. 2012;1444:87–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Ikram F, Ackermann S, Kahlert Y, Volland R, Roels F, Engesser A, et al. Transcription factor activating protein 2 beta (TFAP2B) mediates noradrenergic neuronal differentiation in neuroblastoma. Mol Oncol. 2016;10:344–59.

    Article  CAS  PubMed  Google Scholar 

  34. Tsukada S, Tanaka Y, Maegawa H, Kashiwagi A, Kawamori R, Maeda S. Intronic polymorphisms within TFAP2B regulate transcriptional activity and affect adipocytokine gene expression in differentiated adipocytes. Mol Endocrinol. 2006;20:1104–11.

    Article  CAS  PubMed  Google Scholar 

  35. Albuquerque D, Nóbrega C, Rodríguez-López R, Manco L. Association study of common polymorphisms in MSRA, TFAP2B, MC4R, NRXN3, PPARGC1A, TMEM18, SEC16B, HOXB5 and OLFM4 genes with obesity-related traits among Portuguese children. J Hum Genet. 2014;59:307–13.

    Article  CAS  PubMed  Google Scholar 

  36. Maeda S, Tsukada S, Kanazawa A, Sekine A, Tsunoda T, Koya D, et al. Genetic variations in the gene encoding TFAP2B are associated with type 2 diabetes mellitus. J Hum Genet. 2005;50:283–92.

    Article  CAS  PubMed  Google Scholar 

  37. Jansky S, Sharma AK, Körber V, Quintero A, Toprak UH, Wecht EM, et al. Single-cell transcriptomic analyses provide insights into the developmental origins of neuroblastoma. Nat Genet. 2021;53:683–93.

    Article  CAS  PubMed  Google Scholar 

  38. Berggard C, Damberg M, Oreland L. Brainstem levels of transcription factor AP-2 in rat are changed after treatment with phenelzine, but not with citalopram. BMC Pharmacol. 2005. https://doi.org/10.1186/1471-2210-5-1.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Berggård C, Damberg M, Oreland L. Chronic citalopram treatment induces time-dependent changes in the expression and DNA-binding activity of transcription factor AP-2 in rat brain. Eur Neuropsychopharmacol. 2003;13:11–7.

    Article  PubMed  Google Scholar 

  40. Mitchell PJ, Wang C, Tjian R. Positive and negative regulation of transcription in vitro: enhancer-binding protein AP-2 is inhibited by SV40 T antigen. Cell. 1987;50:847–61.

    Article  CAS  PubMed  Google Scholar 

  41. Wang HV, Vaupel K, Buettner R, Bosserhoff AK, Moser M. Identification and embryonic expression of a new AP-2 transcription factor, AP-2ε. Dev Dyn. 2004;231:128–35.

    Article  CAS  PubMed  Google Scholar 

  42. Eckert D, Buhl S, Weber S, Jäger R, Schorle H. The AP-2 family of transcription factors. Genome Biol. 2005;6:246–246.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Hilger-Eversheim K, Moser M, Schorle H, Buettner R. Regulatory roles of AP-2 transcription factors in vertebrate development, apoptosis and cell-cycle control. Gene. 2000;260:1–12.

    Article  CAS  PubMed  Google Scholar 

  44. Williams T, Tijan R. Characterization of a dimerization motif in AP-2 and its function in heterologous DNA-binding proteins. Science. 1991;251:1067–71.

    Article  CAS  PubMed  Google Scholar 

  45. Williams T, Tjian R. Analysis of the DNA-binding and activation properties of the human transcription factor AP-2. Genes Dev. 1991;5:670–82.

    Article  CAS  PubMed  Google Scholar 

  46. Hong SJ, Huh YH, Leung A, Choi HJ, Ding Y, Kang UJ, et al. Transcription factor AP-2β regulates the neurotransmitter phenotype and maturation of chromaffin cells. Mol Cell Neurosci. 2011;46:245–51.

    Article  CAS  PubMed  Google Scholar 

  47. Bosher JM, Totty NF, Hsuan JJ, Williams T, Hurst HC. A family of AP-2 proteins regulates c-erbB-2 expression in mammary carcinoma. Oncogene. 1996;13:1701–7.

    CAS  PubMed  Google Scholar 

  48. Zhao F, Satoda M, Licht JD, Hayashizaki Y, Gelb BD. Cloning and characterization of a novel mouse AP-2 transcription factor, Ap-2δ, with unique DNA binding and transactivation properties *. J Biol Chem. 2001;276:40755–60.

    Article  CAS  PubMed  Google Scholar 

  49. Moser M, Rüschoff J, Rüschoff R, Buettner R. Comparative analysis of AP-2a and AP-2b gene expression during murine embryogenesis. Dev Dyn. 1997;208:115–24.

    Article  CAS  PubMed  Google Scholar 

  50. Moser M, Dahmen S, Kluge R, Gröne H, Dahmen J, Kunz D, et al. Terminal renal failure in mice lacking transcription factor AP-2β. Lab Investig. 2003;83:571–8.

    Article  CAS  PubMed  Google Scholar 

  51. Moser M, Pscherer A, Roth C, Becker J, Mücher G, Zerres K, et al. Enhanced apoptotic cell death of renal epithelial cells in mice lacking transcription factor AP-2β. Genes Dev. 1997;11:1938–1938.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Schorle H, Meier P, Buchert M, Jaenisch R, Mitchell PJ. Transcription factor AP-2 essential for cranial closure and craniofacial development. Nature. 1996;381:235–8.

    Article  CAS  PubMed  Google Scholar 

  53. Jumper J, Evans R, Pritzel A, Green T, Figurnov M, Ronneberger O, et al. Highly accurate protein structure prediction with AlphaFold. Nature. 2021;596:583–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Duvaud S, Gabella C, Lisacek F, Stockinger H, Ioannidis V, Durinx C. Expasy, the Swiss Bioinformatics Resource Portal, as designed by its users. Nucleic Acids Res. 2021;49:W216–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Nelms BL, Labosky PA. AP genes, transcriptional control of neural crest development. Dev Biol. 2010;1:1–227.

    Google Scholar 

  56. Imagawa M, Chiu R, Karin M. Transcription factor AP-2 mediates induction by two different signal-transduction pathways: protein kinase C and cAMP. Cell. 1987;51:251–60.

    Article  CAS  PubMed  Google Scholar 

  57. Roeslers WJ, Vandenbark GR, Hanson RW. Cyclic AMP and the induction of eukaryotic gene transcription*. J biol Chem. 1988;263:9063–6.

    Article  Google Scholar 

  58. Uhlén M, Fagerberg L, Hallström BM, Lindskog C, Oksvold P, Mardinoglu A, et al. Tissue-based map of the human proteome. Science. 2015;347:1260419.

    Article  PubMed  CAS  Google Scholar 

  59. Pichler K, Warner K, Magrane M. SPIN: submitting sequences determined at protein level to UniProt. Curr Protoc Bioinform. 2018. https://doi.org/10.1002/cpbi.52.

    Article  Google Scholar 

  60. Moser M, Rüschoff J, Buettner R. Comparative Analysis of AP-2 Alpha and AP-2 Beta Gene Expression During Murine Embryogenesis. Dev Dyn. 1997;208:115–24.

    Article  CAS  PubMed  Google Scholar 

  61. Lamontagne JO, Zhang H, Zeid AM, Strittmatter K, Rocha AD, Williams T, et al. Transcription factors AP-2α and AP-2β regulate distinct segments of the distal nephron in the mammalian kidney. Nat Commun. 2022;13:1–18.

    Article  CAS  Google Scholar 

  62. Damberg M. Transcription factor AP-2 and monoaminergic functions in the central nervous system review. J Neural Transm. 2005;112:1281–96.

    Article  CAS  PubMed  Google Scholar 

  63. Zainolabidin N, Kamath SP, Thanawalla AR, Chen AI. Distinct activities of Tfap2A and Tfap2B in the specification of GABAergic interneurons in the developing cerebellum. Front Mol Neurosci. 2017;10:281–281.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Timberlake AT, Jin SC, Nelson-Williams C, Wu R, Furey CG, Islam B, et al. Mutations in TFAP2B and previously unimplicated genes of the BMP, Wnt, and Hedgehog pathways in syndromic craniosynostosis. Proc Natl Acad Sci USA. 2019;116:15116–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Zhao F, Weismann CG, Satoda M, Pierpont MEM, Sweeney E, Thompson EM, et al. Novel TFAP2B mutations that cause char syndrome provide a genotype-phenotype correlation. Am J Hum Genet. 2001;69:695–695.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Kusuma L, Dinesh SM, Savitha MR, Krishnamurthy B, Narayanappa D, Ramachandra NB. Mutations of TFAP2B in congenital heart disease patients in Mysore, south India. Indian J Med Res. 2011;134:621–6.

    Article  CAS  Google Scholar 

  67. Ji W, Benson MA, Bhattacharya S, Chen Y, Hu J, Li F. Characterization of transcription factor AP-2 beta mutations involved in familial isolated patent ductus arteriosus suggests haploinsufficiency. J Surg Res. 2014;188:466–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Ding X, Luo C, Zhou J, Zhong Y, Hu X, Zhou F, et al. The interaction of KCTD1 with transcription factor AP-2α inhibits its transactivation. J Cell Biochem. 2009;106:285–95.

    Article  CAS  PubMed  Google Scholar 

  69. Zarelli VE, Dawid IB. Inhibition of neural crest formation by Kctd15 involves regulation of transcription factor AP-2. Proc Natl Acad Sci USA. 2013. https://doi.org/10.1073/pnas.1300203110.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Liu Z, Xiang Y, Sun G. The KCTD family of proteins: Structure, function, disease relevance. Cell Biosci. 2013;3:1–5.

    Article  CAS  Google Scholar 

  71. Iwamoto N, Yokoyama S. Protein kinase D regulates the adiponectin gene expression through phosphorylation of AP-2: a common pathway to the ABCA1 gene regulation. Atherosclerosis. 2011;216:90–6.

    Article  CAS  PubMed  Google Scholar 

  72. Ivey KN, Sutcliffe D, Richardson J, Clyman RI, Garcia JA, Srivastava D. Transcriptional regulation during development of the ductus arteriosus. Circ Res. 2008. https://doi.org/10.1161/CIRCRESAHA.108.180661.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Bragança J, Eloranta JJ, Bamforth SD, Ibbitt JC, Hurst HC, Bhattacharya S. Physical and functional interactions among AP-2 transcription factors, p300/CREB-binding protein, and CITED2. J Biol Chem. 2003. https://doi.org/10.1074/jbc.M208144200.

    Article  PubMed  Google Scholar 

  74. Bragança J, Swingler T, Marques FIR, Jones T, Eloranta JJ, Hurst HC, et al. Human CREB-binding protein/p300-interacting transactivator with ED-rich tail (CITED) 4, a new member of the CITED family, functions as a co-activator for transcription factor AP-2. J Biol Chem. 2002;277:8559–65.

    Article  PubMed  CAS  Google Scholar 

  75. Ding X, Fan C, Zhou J, Zhong Y, Liu R, Ren K, et al. GAS41 interacts with transcription factor AP-2β and stimulates AP-2β-mediated transactivation. Nucleic Acids Res. 2006;34:2570–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Szklarczyk D, Franceschini A, Wyder S, Forslund K, Heller D, Huerta-Cepas J, et al. STRING v10: protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res. 2015;43:D447–52.

    Article  CAS  PubMed  Google Scholar 

  77. Eloranta JJ, Hurst HC. Transcription factor AP-2 interacts with the SUMO-conjugating enzyme UBC9 and is sumolated in vivo. J Biol Chem. 2002;277:30798–804.

    Article  CAS  PubMed  Google Scholar 

  78. Gamero-Villarroel C, González LM, Rodríguez-López R, Albuquerque D, Carrillo JA, García-Herráiz A, et al. Influence of TFAP2B and KCTD15 genetic variability on personality dimensions in anorexia and bulimia nervosa. Brain Behav. 2017;7:e00784–e00784.

    Article  PubMed  PubMed Central  Google Scholar 

  79. Williams MJ, Goergen P, Rajendran J, Zheleznyakova G, Hägglund MG, Perland E, et al. Obesity-linked homologues TfAP-2 and Twz establish meal frequency in drosophila melanogaster. PLoS Genet. 2014;10:e1004499–e1004499.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  80. Schmidt M, Huber L, Majdazari A, Schütz G, Williams T, Rohrer H. The transcription factors AP-2β and AP-2α are required for survival of sympathetic progenitors and differentiated sympathetic neurons. Dev Biol. 2011;355:89–100.

    Article  CAS  PubMed  Google Scholar 

  81. Williams MJ, Goergen P, Rajendran J, Klockars A, Kasagiannis A, Fredriksson R, et al. Regulation of aggression by obesity-linked genes TfAP-2 and Twz through octopamine signaling in Drosophila. Genetics. 2014;196:349–62.

    Article  CAS  PubMed  Google Scholar 

  82. Jong Hong S, Lardaro T, Sook OhM, Huh Y, Ding Y, Jung Kang U, et al. Regulation of the noradrenaline neurotransmitter phenotype by the transcription factor AP-2β. J Biol Chem. 2008;283:16860–7.

    Article  CAS  Google Scholar 

  83. Heils A, Mößner R, Lesch KP. The human serotonin transporter gene polymorphism-basic research and clinical implications. J Neural Transm. 1997;104:1005.

    Article  CAS  PubMed  Google Scholar 

  84. Lesch KP, Heils A. Serotonergic gene transcriptional control regions: targets for antidepressant drug development ? Int J Neuropsychopharmacol. 2000;3:67–79.

    Article  CAS  PubMed  Google Scholar 

  85. Damberg M, Berggård C, Oreland L. Phenelzine treatment increases transcription factor AP-2 levels in rat brain. BMC Pharmacol. 2003;3:10–10.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Her S, Bell RA, Bloom AK, Siddall BJ, Wong DL. Phenylethanolamine N-methyltransferase gene expression SP1 AND MAZ potential for tissue-specific expression*. J Biol Chem. 1999;274:8698–707.

    Article  CAS  PubMed  Google Scholar 

  87. Ebert SN, Ficklin MB, Her S, Siddall BJ, Bell RA, Ganguly K, et al. Glucocorticoid-dependent action of neural crest factor AP-2: stimulation of phenylethanolamine N-Methyltransferase gene expression. J Neurochem. 2002;70:2286–95.

    Article  Google Scholar 

  88. Wong DL, Siddall BJ, Ebert SN, Bell RA, Her S. Phenylethanolamine N-methyltransferase gene expression: synergistic activation by Egr-1, AP-2 and the glucocorticoid receptor. Mol Brain Res. 1998;61:154–61.

    Article  CAS  PubMed  Google Scholar 

  89. Kim H-S, Hong SJ, LeDoux MS, Kim K-S. Regulation of the tyrosine hydroxylase and dopamine β-hydroxylase genes by the transcription factor AP-2. J Neurochem. 2009;76:280–94.

    Article  Google Scholar 

  90. Kim HS, Seo H, Yang C, Brunet JF, Kim KS. Noradrenergic-specific transcription of the dopamine β-hydroxylase gene requires synergy of multiple cis-acting elements including at least two Phox2a-binding sites. J Neurosci. 1998. https://doi.org/10.1523/jneurosci.18-20-08247.1998.

    Article  PubMed  PubMed Central  Google Scholar 

  91. Sari Y, Johnson VR, Weedman JM. Role of the serotonergic system in alcohol dependence: From animal models to clinics. Progr Mol Biol Transl Sci. 2011;98:401–43.

    Article  CAS  Google Scholar 

  92. Schabram I, Eggermann T, Siegel SJ, Gründer G, Zerres K, Vernaleken I. Neuropsychological correlates of transcription factor AP-2Beta, and its interaction with COMT and MAOA in healthy females. Neuropsychobiology. 2013;68:79–90.

    Article  CAS  PubMed  Google Scholar 

  93. Damberg M, Garpenstrand H, Berggård C, Åsberg M, Hallman J, Oreland L. The genotype of human transcription factor AP-2β is associated with platelet monoamine oxidase B activity. Neurosci Lett. 2000;291:204–6.

    Article  CAS  PubMed  Google Scholar 

  94. Takeuchi S, Imafuku I, Waragai M, Roth C, Kanazawa I, Buettner R, et al. AP-2β represses D(1A) dopamine receptor gene transcription in Neuro2a cells. Mol Brain Res. 1999;74:208–16.

    Article  CAS  PubMed  Google Scholar 

  95. Nishi A, Kuroiwa M, Shuto T. Mechanisms for the modulation of dopamine D1 receptor signaling in striatal neurons. Front Neuroanatomy. 2011;5:43.

    Article  CAS  Google Scholar 

  96. Bhatia A, Lenchner JR, Saadabadi A. Biochemistry, dopamine receptors. Tampa: StatPearls Publishing; 2022.

    Google Scholar 

  97. Ortiz O, Delgado-García JM, Espadas I, Bahí A, Trullas R, Dreyer JL, et al. Associative learning and CA3–CA1 synaptic plasticity are impaired in D1R Null, Drd1a−/− mice and in hippocampal siRNA silenced Drd1a Mice. J Neurosci. 2010;30:12288–12288.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Fazio L, Pergola G, Papalino M, Carlo PD, Monda A, Gelao B, et al. Transcriptomic context of DRD1 is associated with prefrontal activity and behavior during working memory. Proc Natl Acad Sci USA. 2018;115:5582–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Tsang J, Fullard JF, Giakoumaki SG, Katsel P, Karagiorga VE, Greenwood TA, et al. The relationship between dopamine receptor D1 and cognitive performance. NPJ Schizophr. 2015;1:14002–14002.

    Article  PubMed  PubMed Central  Google Scholar 

  100. Weinshenker D. Dopamine beta-hydroxylase. In: Enna SJ, Bylund DB, editors. xPharm: the comprehensive pharmacology reference. New York: Elsevier; 2007. p. 1–15.

    Google Scholar 

  101. Ikegame T, Hidaka Y, Nakachi Y, Murata Y, Watanabe R, Sugawara H, et al. Identification and functional characterization of the extremely long allele of the serotonin transporter-linked polymorphic region. Transl Psychiatry. 2021;11:1–7.

    Article  CAS  Google Scholar 

  102. Nilsson KW, Sonnby K, Nordquist N, Comasco E, Leppert J, Oreland L, et al. Transcription factor activating protein-2β (TFAP-2β) genotype and symptoms of attention deficit hyperactivity disorder in relation to symptoms of depression in two independent samples. Eur Child Adolesc Psychiatry. 2014;23:207–17.

    Article  PubMed  Google Scholar 

  103. Hahn SL, Hahn M, Kang UJ, Joh TH. Structure of the rat aromatic l-amino acid decarboxylase gene: evidence for an alternative promoter usage. J Neurochem. 1993;60:1058–64.

    Article  CAS  PubMed  Google Scholar 

  104. Garlow SJ, Ciaranello RD. Transcriptional control of the rat serotonin-2 receptor gene. Mol Brain Res. 1995;31:201–9.

    Article  CAS  PubMed  Google Scholar 

  105. Du YL, Wilcox BD, Teitler M, Jeffrey JJ. Isolation and characterization of the rat 5-hydroxytryptamine type 2 receptor promoter: constitutive and inducible activity in myometrial smooth muscle cells. Mol Pharmacol. 1994;45:1125–31.

    CAS  PubMed  Google Scholar 

  106. Bedford FK, Julius D, Ingraham HA. Neuronal expression of the 5HT3 serotonin receptor gene requires nuclear factor 1 complexes. J Neurosci. 1998. https://doi.org/10.1523/jneurosci.18-16-06186.1998.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Boularand S, Darmon MC, Ravassard P, Mallet J. Characterization of the human tryptophan hydroxylase gene promoter: transcriptional regulation bY cAMP requires a new motif distinct from the cAMP-responsive element (). J Biol Chem. 1995;270:3757–64.

    Article  CAS  PubMed  Google Scholar 

  108. Zirin J, Hu Y, Liu L, Yang-Zhou D, Colbeth R, Yan D, et al. Large-scale transgenic drosophila resource collections for loss- and gain-of-function studies. Genetics. 2020. https://doi.org/10.1534/genetics.119.302964.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Thurmond J, Goodman JL, Strelets VB, Attrill H, Gramates LS, Marygold SJ, et al. FlyBase 2.0: the next generation. Nucleic Acids Res. 2019;47:D759–65.

    Article  CAS  PubMed  Google Scholar 

  110. Schwartz K, Yadid G, Weizman A, Rehavi M. Decreased limbic vesicular monoamine transporter 2 in a genetic rat model of depression. Brain Res. 2003;965:174–9.

    Article  CAS  PubMed  Google Scholar 

  111. Villemagne VL, Okamura N, Pejoska S, Drago J, Mulligan RS, Chételat G, et al. Differential diagnosis in Alzheimer’s disease and dementia with Lewy bodies via VMAT2 and amyloid imaging. Neurodegener Dis. 2012;10:161–5.

    Article  CAS  PubMed  Google Scholar 

  112. Fehr C, Sommerlad D, Sander T, Anghelescu I, Dahmen N, Szegedi A, et al. Association of VMAT2 gene polymorphisms with alcohol dependence. J Neural Transm. 2013;120:1161–9.

    Article  CAS  PubMed  Google Scholar 

  113. Avsar O, Kuskucu A, Sancak S, Genc E. Do vesicular monoamine transporter 2 genotypes relate to obesity and eating behavior? Neuropsychiatry. 2017. https://doi.org/10.4172/neuropsychiatry.1000310.

    Article  Google Scholar 

  114. Xu Y, Lu Y, Xu P, Mangieri LR, Isingrini E, Xu Y, et al. VMAT2-mediated neurotransmission from midbrain leptin receptor neurons in feeding regulation. eNeuro. 2017. https://doi.org/10.1523/ENEURO.0083-17.2017.

    Article  PubMed  PubMed Central  Google Scholar 

  115. Kolat D, Kaluzinska Z, Bednarek AK, Pluciennik E. The biological characteristics of transcription factors AP-2α and AP-2γ and their importance in various types of cancers. 2019. Biosci Rep. https://doi.org/10.1042/BSR20181928.

  116. Safran M, Rosen N, Twik M, BarShir R, Stein TI, Dahary D, et al. The GeneCards Suite. Practical Guide to Life Science Databases. 2021;27–56.

  117. Nilsson KW, Sjöberg RL, Leppert J, Oreland L, Damberg M. Transcription factor AP-2β genotype and psychosocial adversity in relation to adolescent depressive symptomatology. J Neural Transm. 2009;116:363–70.

    Article  CAS  PubMed  Google Scholar 

  118. Maeda S, Osawa N, Hayashi T, Tsukada S, Kobayashi M, Kikkawa R. Genetic variations associated with diabetic nephropathy and type II diabetes in a Japanese population. Kidney Int. 2007;72(SUPPL. 106):S43–8.

    Article  CAS  Google Scholar 

  119. Hensch T, Wargelius HL, Herold U, Strobel A, Oreland L, Brocke B. Electrophysiological and behavioral correlates of polymorphisms in the transcription factor AP-2β coding gene. Neurosci Lett. 2008;436:67–71.

    Article  CAS  PubMed  Google Scholar 

  120. Stocks T, Ängquist L, Banasik K, Harder MN, Taylor MA, Hager J, et al. TFAP2B influences the effect of dietary fat on weight loss under energy restriction. PLoS ONE. 2012;7:e43212–e43212.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Tomei S, Mamtani R, Al Ali R, Elkum N, Abdulmalik M, Ismail A, et al. Obesity susceptibility loci in Qataris, a highly consanguineous Arabian population. J Transl Med. 2015;13:1.

    Article  CAS  Google Scholar 

  122. Iłowiecka K, Glibowski P, Skrzypek M, Styk W. The long-term dietitian and psychological support of obese patients who have reduced their weight allows them to maintain the effects. Nutrients. 2021;13:2020–2020.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  123. Sandholt CH, Hansen T, Pedersen O. Beyond the fourth wave of genome-wide obesity association studies. Nutr Diabetes. 2012;2:37–37.

    Article  CAS  Google Scholar 

  124. Kondo M, Maegawa H, Obata T, Ugi S, Ikeda K, Morino K, et al. Transcription factor activating protein-2β: a positive regulator of monocyte chemoattractant protein-1 gene expression. Endocrinology. 2009. https://doi.org/10.1210/en.2008-1361.

    Article  PubMed  PubMed Central  Google Scholar 

  125. Joost U, Villa I, Comasco E, Oreland L, Veidebaum T, Harro J. Association between Transcription Factor AP-2B genotype, obesity, insulin resistance and dietary intake in a longitudinal birth cohort study. Int J Obes. 2019;43:2095–106.

    Article  CAS  Google Scholar 

  126. Hebbar P, Abubaker JA, Abu-Farha M, Tuomilehto J, Al-Mulla F, Thanaraj TA. A perception on genome-wide genetic analysis of metabolic traits in Arab populations. Front Endocrinol. 2019;10:8.

    Article  Google Scholar 

  127. Katus U, Villa I, Ringmets I, Pulver A, Veidebaum T, Harro J. The role of reward sensitivity in obesity and its association with Transcription Factor AP-2B: a longitudinal birth cohort study. Neurosci Lett. 2020;735:135158.

    Article  CAS  PubMed  Google Scholar 

  128. Scott CC, Vossio S, Rougemont J, Gruenberg J. TFAP2 transcription factors are regulators of lipid droplet biogenesis. Elife. 2018;7:e36330.

    Article  PubMed  PubMed Central  Google Scholar 

  129. Bille DS, Banasik K, Justesen JM, Sandholt CH, Sandbæk A, Lauritzen T, et al. Implications of central obesity-related variants in LYPLAL1, NRXN3, MSRA, and TFAP2B on quantitative metabolic traits in adult danes. PLoS ONE. 2011;6: e20640.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Lindgren CM, Heid IM, Randall JC, Lamina C, Steinthorsdottir V, Qi L, et al. Genome-wide association scan meta-analysis identifies three loci influencing adiposity and fat distribution. PLoS Genet. 2009;5: e1000508.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  131. Stocks T, Ängquist L, Hager J, Charon C, Holst C, Martinez JA, et al. TFAP2B-dietary protein and glycemic index interactions and weight maintenance after weight loss in the DiOGenes trial. Hum Hered. 2013;75:213–9.

    Article  CAS  PubMed  Google Scholar 

  132. Isse N, Ogawa Y, Tamura N, Masuzaki H, Mori K, Okazaki T, et al. Structural organization and chromosomal assignment of the human obese gene. J Biol Chem. 1995;270:27728–33.

    Article  CAS  PubMed  Google Scholar 

  133. Fuke T, Yoshizaki T, Kondo M, Morino K, Obata T, Ugi S, et al. Transcription factor AP-2Β inhibits expression and secretion of leptin, an insulin-sensitizing hormone, in 3T3-L1 adipocytes. Int J Obes. 2010;34:670–8.

    Article  CAS  Google Scholar 

  134. Meng X, Kondo M, Morino K, Fuke T, Obata T, Yoshizaki T, et al. Transcription factor AP-2β: A negative regulator of IRS-1 gene expression. Biochem Biophys Res Commun. 2010;392:526–32.

    Article  CAS  PubMed  Google Scholar 

  135. Ikeda K, Maegawa H, Ugi S, Tao Y, Nishio Y, Tsukada S, et al. Transcription factor activating enhancer-binding protein-2β: a negative regulator of adiponectin gene expression. J Biol Chem. 2006;281:31245–53.

    CAS  PubMed  Google Scholar 

  136. Mojiminiyi OA, Abdella NA, Al Arouj M, Ben NA. Adiponectin, insulin resistance and clinical expression of the metabolic syndrome in patients with Type 2 diabetes. Int J Obes. 2005;2007(31):213–20.

    Google Scholar 

  137. Lihn AS, Pedersen SB, Richelsen B. Adiponectin: action, regulation and association to insulin sensitivity. Obes Rev. 2005;6:13–21.

    Article  CAS  PubMed  Google Scholar 

  138. Marette A, Bukowiecki LJ. Noradrenaline stimulates glucose transport in rat brown adipocytes by activating thermogenesis. Evidence that fatty acid activation of mitochondrial respiration enhances glucose transport. Biochemical J. 1991;277:119–24.

    Article  CAS  Google Scholar 

  139. Chernogubova E, Cannon B, Bengtsson T. Norepinephrine increases glucose transport in brown adipocytes via β 3 -Adrenoceptors through a cAMP, PKA, and PI3-Kinase-dependent pathway stimulating conventional and novel PKCs. Endocrinology. 2004;145:269–80.

    Article  CAS  PubMed  Google Scholar 

  140. Esler M, Straznicky N, Eikelis N, Masuo K, Lambert G, Lambert E. Mechanisms of sympathetic activation in obesity-related hypertension. Hypertension. 2006. https://doi.org/10.1161/01.HYP.0000242642.42177.49.

    Article  PubMed  Google Scholar 

  141. Deibert DC, DeFronzo RA. Epinephrine-induced insulin resistance in man. J Clin Investig. 1980;65:717–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. LeeAbraham D, Hansen PA, Schluter J, Gulve EA, Gao J, Holloszy JO. Effects of epinephrine on insulin-stimulated glucose uptake and GLUT-4 phosphorylation in muscle. Am J Physiol. 1997;273:3.

    Google Scholar 

  143. Khoury N, Mcgill JB. Reduction in insulin sensitivity following administration of the clinically used low-dose pressor, norepinephrine. Diabetes Metab Res Rev. 2011;27:604–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Penesova A, Radikova Z, Cizmarova E, Kvetňanský R, Blazicek P, Vlcek M, et al. The role of norepinephrine and insulin resistance in an early stage of hypertension. Ann NY Acad Sci. 2008;1148:490–4.

    Article  CAS  PubMed  Google Scholar 

  145. Mannelli M, Parenti G, Zampetti B, Canu L, Mannucci E. Diabetes from Catecholamine Excess. In: Frontiers in Diabetes. S. Karger AG; 2014. p. 44–51.

  146. Bottner A, Haidan A, Eisenhofer G, Kristensen K, Castle AL, Scherbaum WA, et al. Increased body fat mass and suppression of circulating leptin levels in response to hypersecretion of epinephrine in phenylethanolamine-N-methyltransferase (PNMT)-overexpressing mice. Endocrinology. 2000;141:4239–46.

    Article  CAS  PubMed  Google Scholar 

  147. Drabe M, Rullmann M, Luthardt J, Boettcher Y, Regenthal R, Ploetz T, et al. Serotonin transporter gene promoter methylation status correlates with in vivo prefrontal 5-HTT availability and reward function in human obesity. Transl Psychiatry. 2017;7:e1167–e1167.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Zhao J, Goldberg J, Vaccarino V. Promoter methylation of serotonin transporter gene is associated with obesity measures: a monozygotic twin study. Int J Obes. 2013;37:140–5.

    Article  CAS  Google Scholar 

  149. Oreland L, Hallman J, Damberg M. Platelet MAO and personality—function and dysfunction. Curr Med Chem. 2012;11:2007–16.

    Article  Google Scholar 

  150. Oreland L, Nilsson K, Damberg M, Hallman J. Monoamine oxidases—activities, genotypes and the shaping of behaviour. J Neural Transm. 2007;114(6):817–22.

    Article  CAS  PubMed  Google Scholar 

  151. Tikkanen R, Ducci F, Goldman D, Holi M, Lindberg N, Tiihonen J, et al. MAOA alters the effects of heavy drinking and childhood physical abuse on risk for severe impulsive acts of violence among alcoholic violent offenders. Alcohol Clin Exp Res. 2010. https://doi.org/10.1111/j.1530-0277.2010.01157.x.

    Article  PubMed  PubMed Central  Google Scholar 

  152. Preuss UW, Wurst FM, Ridinger M, Rujescu D, Fehr C, Koller G, et al. Association of functional DBH genetic variants with alcohol dependence risk and related depression and suicide attempt phenotypes: Results from a large multicenter association study. Drug Alcohol Depend. 2013;133:459–67.

    Article  CAS  PubMed  Google Scholar 

  153. Zhao R, Zhang R, Li W, Liao Y, Tang J, Miao Q, et al. Genome-wide DNA methylation patterns in discordant sib pairs with alcohol dependence. Asia Pac Psychiatry. 2013. https://doi.org/10.1111/appy.12010.

    Article  PubMed  Google Scholar 

  154. Hodge CW, Samson HH, Chappelle AM. Alcohol self-administration: further examination of the role of dopamine receptors in the nucleus accumbens. Alcohol Clin Exp Res. 1997;21:1083–91.

    CAS  PubMed  Google Scholar 

  155. Bahi A, Dreyer JL. Involvement of nucleus accumbens dopamine D1 receptors in ethanol drinking, ethanol-induced conditioned place preference, and ethanol-induced psychomotor sensitization in mice. Psychopharmacology. 2012;222:141–53.

    Article  CAS  PubMed  Google Scholar 

  156. Abrahao KP, Quadros IMH, Souza-Formigoni MLO. Nucleus accumbens dopamine D1 receptors regulate the expression of ethanol-induced behavioural sensitization. Int J Neuropsychopharmacol. 2011;14:175–85.

    Article  CAS  PubMed  Google Scholar 

  157. El-Ghundi M, George SR, Drago J, Fletcher PJ, Fan T, Nguyen T, et al. Disruption of dopamine D1 receptor gene expression attenuates alcohol-seeking behavior. Eur J Pharmacol. 1998;353:149–58.

    Article  CAS  PubMed  Google Scholar 

  158. Boyce-Rustay JM, Wiedholz LM, Millstein RA, Carroll J, Murphy DL, Daws LC, et al. Ethanol-related behaviors in serotonin transporter knockout mice. Alcohol Clin Exp Res. 2006. https://doi.org/10.1111/j.1530-0277.2006.00241.x.

    Article  PubMed  Google Scholar 

  159. Atigari OV, Kelly AM, Jabeen Q, Healy D. New onset alcohol dependence linked to treatment with selective serotonin reuptake inhibitors. Int J Risk Saf Med. 2013;25:105–9.

    Article  PubMed  Google Scholar 

  160. Brookwell L, Hogan C, Healy D, Mangin D. Ninety-three cases of alcohol dependence following SSRI treatment. Int J Risk Saf Med. 2014;26:99–107.

    Article  PubMed  Google Scholar 

  161. Philibert RA, Gunter TD, Beach SRH, Brody GH, Madan A. Rapid publication: MAOA methylation is associated with nicotine and alcohol dependence in women. Am J Med Genet Part B Neuropsychiatric Genet. 2008. https://doi.org/10.1002/ajmg.b.30778.

    Article  Google Scholar 

  162. Bach H, Arango V, Kassir SA, Tsaava T, Dwork AJ, Mann JJ, et al. Alcoholics have more tryptophan hydroxylase 2 mRNA and protein in the dorsal and median raphe nuclei. Alcohol Clin Exp Res. 2014. https://doi.org/10.1111/acer.12414.

    Article  PubMed  PubMed Central  Google Scholar 

  163. Kuhn DM, Hasegawa H. Tryptophan hydroxylase and serotonin synthesis regulation. Handb Behav Neurosci. 2020;31:239–56.

    Article  Google Scholar 

  164. SSRIs in Depression and Anxiety. SSRIs in Depression and Anxiety. 2001https://doi.org/10.1002/0470846518

  165. Garcia-Miralles M, Ooi J, Ferrari Bardile C, Tan LJJ, George M, Drum CLL, et al. Treatment with the MAO-A inhibitor clorgyline elevates monoamine neurotransmitter levels and improves affective phenotypes in a mouse model of Huntington disease. Exp Neurol. 2016. https://doi.org/10.1016/j.expneurol.2016.01.019.

    Article  PubMed  Google Scholar 

  166. Bortolato M, Shih JC, et al. Behavioral outcomes of monoamine oxidase deficiency: preclinical and clinical evidence. Int Rev Neurobiol. 2011;100:13–42.

    Article  PubMed  PubMed Central  Google Scholar 

  167. Tyrer P, Srrawcuosst C. Monoamine oxidase inhibitors in anxiety disorders. J Psychiatric Res. 1988;22:87–98.

    Article  Google Scholar 

  168. Shulman KII, Herrmann N, Walker SEE. Current place of monoamine oxidase inhibitors in the treatment of depression. CNS Drugs. 2013. https://doi.org/10.1007/s40263-013-0097-3.

    Article  PubMed  Google Scholar 

  169. Thase ME. The role of monoamine oxidase inhibitors in depression treatment guidelines. J Clin Psychiatry. 2012. https://doi.org/10.4088/JCP.11096su1c.02.

    Article  PubMed  Google Scholar 

  170. Jakubovski E, Johnson JA, Nasir M, Müller-Vahl K, Bloch MH. Systematic review and meta-analysis: dose–response curve of SSRIs and SNRIs in anxiety disorders. Depress Anxiety. 2019. https://doi.org/10.1002/da.22854.

    Article  PubMed  Google Scholar 

  171. Jans LAW, Riedel WJ, Markus CR, Blokland A. Serotonergic vulnerability and depression: assumptions, experimental evidence and implications. Mol Psychiatry. 2007. https://doi.org/10.1038/sj.mp.4001920.

    Article  PubMed  Google Scholar 

  172. Mentis A-FA, Dardiotis E, Katsouni E, Chrousos GP. From warrior genes to translational solutions: novel insights into monoamine oxidases (MAOs) and aggression. Transl Psychiatry. 2021;11:130–130.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Kolla NJ, Vinette SA. Monoamine oxidase A in antisocial personality disorder and borderline personality disorder. Curr Behav Neurosci Rep. 2017;4:41–41.

    Article  PubMed  PubMed Central  Google Scholar 

  174. Huizinga D, Haberstick BC, Smolen A, Menard S, Young SE, Corley RP, et al. Childhood maltreatment, subsequent antisocial behavior, and the role of monoamine oxidase A genotype. Biol Psychiat. 2006;60:677–83.

    Article  CAS  PubMed  Google Scholar 

  175. Pattij T, Vanderschuren LJMJ. The neuropharmacology of impulsive behaviour. Trends Pharmacol Sci. 2008;29:192–9.

    Article  CAS  PubMed  Google Scholar 

  176. Prichard ZM, Jorm AF, Mackinnon A, Easteal S. Association analysis of 15 polymorphisms within 10 candidate genes for antisocial behavioural traits. Psychiatr Genet. 2007. https://doi.org/10.1097/YPG.0b013e32816ebc9e.

    Article  PubMed  Google Scholar 

  177. Scott AL, Bortolato M, Chen K, Shih JC. Novel monoamine oxidase A knock out mice with human-like spontaneous mutation. NeuroReport. 2008. https://doi.org/10.1097/WNR.0b013e3282fd6e88.

    Article  PubMed  PubMed Central  Google Scholar 

  178. Ramakrishnan V. MAOA gene associated with aggressive behavior in humans. J Down Synd Chromosome Abnorm. 2017. https://doi.org/10.4172/2472-1115.1000120.

    Article  Google Scholar 

  179. Cases O, Seif I, Grimsby J, Gaspar P, Chen K, Pournin S, et al. Aggressive behavior and altered amounts of brain serotonin and norepinephrine in mice lacking MAOA. Science. 1995. https://doi.org/10.1126/science.7792602.

    Article  PubMed  PubMed Central  Google Scholar 

  180. Gogos JA, Morgan M, Luine V, Santha M, Ogawa S, Pfaff D, et al. Catechol-O-methyltransferase-deficient mice exhibit sexually dimorphic changes in catecholamine levels and behavior. Proc Natl Acad Sci USA. 1998. https://doi.org/10.1073/pnas.95.17.9991.

    Article  PubMed  PubMed Central  Google Scholar 

  181. Volavka J, Kennedy JL, Ni X, Czobor P, Nolan K, Sheitman B, et al. COMT158 polymorphism and hostility. Am J Med Genet Neuropsychiatric Genet. 2004. https://doi.org/10.1002/ajmg.b.20149.

    Article  Google Scholar 

  182. Strous RD, Bark N, Parsia SS, Volavka J, Lachman HM. Analysis of a functional catechol-O-methyltransferase gene polymorphism in schizophrenia: evidence for association with aggressive and antisocial behavior. Psychiatry Res. 1997. https://doi.org/10.1016/S0165-1781(96)03111-3.

    Article  PubMed  Google Scholar 

  183. Lee RJ, Coccaro EF. Neurotransmitters and Intermittent Explosive Disorder. In: Intermittent Explosive Disorder: Etiology, Assessment, and Treatment. Elsevier; 2019. p. 87–110.

  184. Marino MD, Bourdélat-Parks BN, Cameron Liles L, Weinshenker D. Genetic reduction of noradrenergic function alters social memory and reduces aggression in mice. Behav Brain Res. 2005. https://doi.org/10.1016/j.bbr.2005.02.005.

    Article  PubMed  Google Scholar 

  185. Holmes A, Murphy DL, Crawley JN. Abnormal behavioral phenotypes of serotonin transporter knockout mice: Parallels with human anxiety and depression. Biol Psychiat. 2003. https://doi.org/10.1016/j.biopsych.2003.09.003.

    Article  PubMed  Google Scholar 

  186. Quadros IM, Takahashi A, Miczek KA. Serotonin and aggression—an update. In: Handbook of Behavioral Neuroscience. Elsevier B.V.; 2020. p. 635–63.

  187. Quadros IM, Takahashi A, Miczek KA. Serotonin and Aggression. In: Handbook of Behavioral Neuroscience. Elsevier; 2010. p. 687–713.

  188. Veroude K, Zhang-James Y, Fernàndez-Castillo N, Bakker MJ, Cormand B, Faraone SV. Genetics of aggressive behavior: an overview. Am J Med Genet Part B Neuropsychiatric Genet. 2016. https://doi.org/10.1002/ajmg.b.32364.

    Article  Google Scholar 

  189. Dumitrescu L, Mahoney ER, Mukherjee S, Lee ML, Bush WS, Engelman CD, et al. Genetic variants and functional pathways associated with resilience to Alzheimer’s disease. Brain. 2020;143:2561–75.

    Article  PubMed  PubMed Central  Google Scholar 

  190. García MA, Vázquez J, Giménez C, Valdivieso F, Zafra F. Transcription factor AP-2 regulates human apolipoprotein E gene expression in astrocytoma cells. J Neurosci. 1996;16:7550–6.

    Article  PubMed  PubMed Central  Google Scholar 

  191. Šimić G, Babić Leko M, Wray S, Harrington CR, Delalle I, Jovanov-Milošević N, et al. Monoaminergic neuropathology in Alzheimer’s disease. Prog Neurobiol. 2017;151:101–38.

    Article  PubMed  CAS  Google Scholar 

  192. Babić Leko M, Nikolac Perković M, Klepac N, Švob Štrac D, Borovečki F, Pivac N, et al. Relationships of cerebrospinal fluid alzheimer’s disease biomarkers and COMT, DBH, and MAOB single nucleotide polymorphisms. J Alzheimer’s Dis. 2020;73:135–45.

    Article  CAS  Google Scholar 

  193. Pan X, Kaminga AC, Jia P, Wen SW, Acheampong K, Liu A. Catecholamines in Alzheimer’s disease: a systematic review and meta-analysis. Front Aging Neurosci. 2020;12:184.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Burke WJ, Chung HD, Marshall GL, Gillespie KN, Joh TH. Evidence for decreased transport of PNMT protein in advanced Alzheimer’s Disease. J Am Geriatr Soc. 1990. https://doi.org/10.1111/j.1532-5415.1990.tb03448.x.

    Article  PubMed  Google Scholar 

  195. Mustapić M, Presečki P, Mimica N, Pivac N, Folnegović Šmalc V, Mück-Šeler D. Dopamine beta-hydroxylase and inflammatory cytokines in Alzheimer’s disease. Periodicum biologorum 112, Suppl 1 - Final Programme and Abstract Book of the 6th Croatian Congress of Pharmacology with International Participation. 2010.

  196. Trillo L, Das D, Hsieh W, Medina B, Moghadam S, Lin B, et al. Ascending monoaminergic systems alterations in Alzheimer’s disease Translating basic science into clinical care. Neurosci Biobehav Rev. 2013;37:1363–79.

    Article  CAS  PubMed  Google Scholar 

  197. Mann MB, Wu S, Rostamkhani M, Tourtellotte W, MacMurray J, Comings DE. Phenylethanolamine N-methyltransferase (PNMT) gene and early-onset Alzheimer disease. Am J Med Genet Neuropsychiatric Genet. 2001;105:312–6.

    Article  CAS  Google Scholar 

  198. Mustapic M, Presecki P, Pivac N, Mimica N, Hof PR, Simic G, et al. Genotype-independent decrease in plasma dopamine beta-hydroxylase activity in Alzheimer’s disease. Prog Neuropsychopharmacol Biol Psychiatry. 2013. https://doi.org/10.1016/j.pnpbp.2013.02.002.

    Article  PubMed  PubMed Central  Google Scholar 

  199. Perkovic MN, Strac DS, Tudor L, Konjevod M, Erjavec GN, Pivac N. Catechol-O-methyltransferase, cognition and Alzheimer’s Disease. Curr Alzheimer Res. 2018;15:408–19.

    Article  CAS  PubMed  Google Scholar 

  200. Lukiw WJ, Rogaev EI. Genetics of aggression in Alzheimer’s Disease (AD). Front Aging Neurosci. 2017;9:87–87.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  201. Adolfsson R, Gottfries CG, Oreland L, Wiberg A, Winblad B. Increased activity of brain and platelet monoamine oxidase in dementia of Alzheimer type. Life Sci. 1980;27:1029–34.

    Article  CAS  PubMed  Google Scholar 

  202. Patel CN, Georrge JJ, Modi KM, Narechania MB, Patel DP, Gonzalez FJ, et al. Pharmacophore-based virtual screening of catechol-o-methyltransferase (COMT) inhibitors to combat Alzheimer’s disease. J Biomol Struct Dyn. 2018. https://doi.org/10.1080/07391102.2017.1404931.

    Article  PubMed  Google Scholar 

  203. Cai Z. Monoamine oxidase inhibitors: promising therapeutic agents for Alzheimer’s disease (Review). Mol Med Rep. 2014. https://doi.org/10.3892/mmr.2014.2040.

    Article  PubMed  PubMed Central  Google Scholar 

  204. Serretti A, Olgiati P. Catechol-O-Methyltransferase and Alzheimer’s Disease: A Review of Biological and Genetic Findings. CNS & Neurol Disord Drug Targets. 2012;11:299–305.

    Article  CAS  Google Scholar 

  205. Smith GS, Barrett FS, Joo JH, Nassery N, Savonenko A, Sodums DJ, et al. Molecular imaging of serotonin degeneration in mild cognitive impairment. Neurobiol Dis. 2017. https://doi.org/10.1016/j.nbd.2017.05.007.

    Article  PubMed  PubMed Central  Google Scholar 

  206. Mdawar B, Ghossoub E, Khoury R. Selective serotonin reuptake inhibitors and Alzheimer’s disease. Neural Regen Res. 2020;15:41–6.

    Article  PubMed  Google Scholar 

  207. Bradner JE, Young DHRA. Transcriptional addiction in cancer. Cell. 2017;168:629–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Henley MJ, Koehler AN. Advances in targeting ‘undruggable’ transcription factors with small molecules. Nat Rev Drug Discov. 2021;20:669–88.

    Article  CAS  PubMed  Google Scholar 

  209. Durbin AD, Zimmerman MW, Dharia NV, Abraham BJ, Iniguez AB, Weichert-Leahey N, et al. Selective gene dependencies in MYCN-amplified neuroblastoma include the core transcriptional regulatory circuitry. Nat Genet. 2018;50:1240–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Thorell K, Bergman A, Carén H, Nilsson S, Kogner P, Martinsson T, et al. Verification of genes differentially expressed in neuroblastoma tumours: a study of potential tumour suppressor genes. BMC Med Genom. 2009;2:53.

    Article  CAS  Google Scholar 

  211. Li B, Xie S, Xia A, Suo T, Huang H, Zhang X, et al. Recent advance in the sensing of biomarker transcription factors. TrAC Trends Anal Chem. 2020;132:116039–116039.

    Article  CAS  Google Scholar 

  212. Ahsen ME, Chun Y, Grishin A, Grishina G, Stolovitzky G, Pandey G, et al. NeTFactor, a framework for identifying transcriptional regulators of gene expression-based biomarkers. Sci Rep. 2019. https://doi.org/10.1038/s41598-019-49498-y.

    Article  PubMed  PubMed Central  Google Scholar 

  213. Kaur M, MacPherson CR, Schmeier S, Narasimhan K, Choolani M, Bajic VB. In Silico discovery of transcription factors as potential diagnostic biomarkers of ovarian cancer. BMC Syst Biol. 2011. https://doi.org/10.1186/1752-0509-5-144.

    Article  PubMed  PubMed Central  Google Scholar 

  214. Raap M, Gierendt L, Kreipe HH, Christgen M. Transcription factor AP-2beta in development, differentiation and tumorigenesis. Int J Cancer. 2021;149:1221–7.

    Article  CAS  PubMed  Google Scholar 

  215. Fu X, Zhang H, Chen Z, Yang Z, Shi D, Liu T, et al. TFAP2B overexpression contributes to tumor growth and progression of thyroid cancer through the COX-2 signaling pathway. Cell Death Dis. 2019;10:1–13.

    Article  CAS  Google Scholar 

  216. Fu L, Shi K, Wang J, Chen W, Shi D, Tian Y, et al. TFAP2B overexpression contributes to tumor growth and a poor prognosis of human lung adenocarcinoma through modulation of ERK and VEGF/PEDF signaling. Mol Cancer. 2014;13:89–89.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  217. Wu H, Zhang J. Decreased expression of TFAP2B in endometrial cancer predicts poor prognosis: A study based on TCGA data. Gynecol Oncol. 2018;149:592–7.

    Article  CAS  PubMed  Google Scholar 

  218. Oya M, Mikami S, Mizuno R, Miyajima A, Horiguchi Y, Nakashima J, et al. Differential expression of activator protein-2 isoforms in renal cell carcinoma. Urology. 2004;64:162–7.

    Article  PubMed  Google Scholar 

  219. Tun HW, Marlow LA, von Roemeling CA, Cooper SJ, Kreinest P, Wu K, et al. Pathway signature and cellular differentiation in clear cell renal cell carcinoma. PLoS ONE. 2010;5:e10696.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  220. Zhu M, Zou L, Lu F, Ye L, Su B, Yang K, et al. miR-142-5p promotes renal cell tumorigenesis by targeting TFAP2B. Oncol Lett. 2020;20:1–1.

    CAS  Google Scholar 

  221. Li Z, Xu X, Luo M, Hao J, Zhao S, Yu W, et al. Activator protein-2β promotes tumor growth and predicts poor prognosis in breast cancer. Cell Physiol Biochem. 2018;47:1925–35.

    Article  CAS  PubMed  Google Scholar 

  222. Pellikainen JM, Kosma V-M. Activator protein-2 in carcinogenesis with a special reference to breast cancer—a mini review. Int J Cancer. 2007;120:2061–7.

    Article  CAS  PubMed  Google Scholar 

  223. Raap M, Gronewold M, Christgen H, Glage S, Bentires-Alj M, Koren S, et al. Lobular carcinoma in situ and invasive lobular breast cancer are characterized by enhanced expression of transcription factor AP-2β. Lab Investg. 2018. https://doi.org/10.1038/labinvest.2017.106.

    Article  Google Scholar 

  224. Wang F, Huang W, Hu X, Chen C, Li X, Qiu J, et al. Transcription factor AP-2β suppresses cervical cancer cell proliferation by promoting the degradation of its interaction partner β-catenin. Mol Carcinog. 2017;56:1909–23.

    Article  CAS  PubMed  Google Scholar 

  225. Beger M, Butz K, Denk C, Williams T, Hurst HC, Hoppe-Seyler F. Expression pattern of AP-2 transcription factors in cervical cancer cells and analysis of their influence on human papillomavirus oncogene transcription. J Mol Med. 2001;79:314–20.

    Article  CAS  PubMed  Google Scholar 

  226. Bence M, Koller J, Sasvari-Szekely M, Keszler G. Transcriptional modulation of monoaminergic neurotransmission genes by the histone deacetylase inhibitor trichostatin A in neuroblastoma cells. J Neural Transm. 2012;119:17–24.

    Article  CAS  PubMed  Google Scholar 

  227. Fontaine F, Overman J, François M. Pharmacological manipulation of transcription factor protein-protein interactions: opportunities and obstacles. Cell Regen. 2015. https://doi.org/10.1186/s13619-015-0015-x.

    Article  PubMed  PubMed Central  Google Scholar 

  228. Yan C, Higgins PJ. Drugging the undruggable: Transcription therapy for cancer. Biochim Biophys Acta Rev Cancer. 2013;1835:76–85.

    Article  CAS  Google Scholar 

  229. Mohamad Anuar NN, Nor Hisam NS, Liew SL, Ugusman A. Clinical review: navitoclax as a pro-apoptotic and anti-fibrotic agent. Front Pharmacol. 2020;11:1817–1817.

    Article  CAS  Google Scholar 

  230. Lamhamedi-Cherradi SE, Menegaz BA, Ramamoorthy V, Aiyer RA, Maywald RL, Buford AS, et al. An oral formulation of YK-4-279: Preclinical efficacy and acquired resistance patterns in Ewing sarcoma. Mol Cancer Ther. 2015. https://doi.org/10.1158/1535-7163.MCT-14-0334.

    Article  PubMed  Google Scholar 

  231. Lambert M, Jambon S, Depauw S, David-Cordonnier MH. Targeting transcription factors for cancer treatment. Molecules. 2018;23:1479.

    Article  PubMed Central  CAS  Google Scholar 

  232. Bushweller JH. Targeting transcription factors in cancer — from undruggable to reality. Nat Rev Cancer. 2019. https://doi.org/10.1038/s41568-019-0196-7.

    Article  PubMed  PubMed Central  Google Scholar 

  233. Inamoto I, Shin JA. Peptide therapeutics that directly target transcription factors. Pept Sci. 2019;111:e24048–e24048.

    Article  CAS  Google Scholar 

  234. Ozers MS, Warren CL, Ansari AZ. Determining DNA sequence specificity of natural and artificial transcription factors by cognate site identifier analysis. Methods Mol Biol. 2009;544:637–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  235. Allen-Petersen BL, Sears RC. Mission possible: advances in MYC therapeutic targeting in cancer. BioDrugs. 2019. https://doi.org/10.1007/s40259-019-00370-5.

    Article  PubMed  PubMed Central  Google Scholar 

  236. Ludwig JA, Federman NC, Anderson PM, Macy ME, Riedel RF, Davis LE, et al. TK216 for relapsed/refractory Ewing sarcoma: Interim phase 1/2 results. JCO. 2021;39:11500–11500.

    Article  Google Scholar 

  237. Robledinos-Antón N, Fernández-Ginés R, Manda G, Cuadrado A. Activators and Inhibitors of NRF2: a review of their potential for clinical development. Oxid Med Cell Longev. 2019. https://doi.org/10.1155/2019/9372182.

    Article  PubMed  PubMed Central  Google Scholar 

  238. Ghosh D, Papavassiliou AG. Transcription factor therapeutics: long-shot or lodestone. Curr Med Chem. 2005;12:691–701.

    Article  CAS  PubMed  Google Scholar 

  239. Erkizan HV, Kong Y, Merchant M, Schlottmann S, Barber-Rotenberg JS, Abaan OD, et al. Small molecule selected to disrupt oncogenic protein EWS-FLI1 interaction with RNA Helicase A inhibits Ewing’s Sarcoma. Nat Med. 2009;15:750–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  240. Branca MA. Multi-kinase inhibitors create buzz at ASCO. Nat Biotechnol. 2005;23:639–639.

    Article  CAS  PubMed  Google Scholar 

  241. Frantz S. Playing dirty. Nature. 2005;437:942–3.

    Article  CAS  PubMed  Google Scholar 

  242. Rothstein M, Simoes-Costa M. Heterodimerization of TFAP2 pioneer factors drives epigenomic remodeling during neural crest specification. Genome Res. 2020;30:35–48.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  243. Huang Z, Xu H, Sandell L. Negative regulation of chondrocyte differentiation by transcription factor AP-2alpha. J Bone Miner Res. 2004;19:245–55.

    Article  CAS  PubMed  Google Scholar 

  244. Huckle WR. Molecular biology of placental development and disease. Progr Mol Biol Transl Sci. 2017;145:29–37.

    Article  CAS  Google Scholar 

  245. Xie WF, Kondo S, Sandell LJ. Regulation of the mouse cartilage-derived retinoic acid-sensitive protein gene by the transcription factor AP-2. J Biol Chem. 1998;273:5026–32.

    Article  CAS  PubMed  Google Scholar 

  246. Davies SR, Sakano S, Zhu Y, Sandell LJ. Distribution of the transcription factors Sox9, AP-2, and [delta]EF1 in adult murine articular and meniscal cartilage and growth plate. J Histochem Cytochem. 2002;50:1059–65.

    Article  CAS  PubMed  Google Scholar 

  247. Seberg HE, Van Otterloo E, Loftus SK, Liu H, Bonde G, Sompallae R, et al. TFAP2 paralogs regulate melanocyte differentiation in parallel with MITF. PLoS Genet. 2017;13:e1006636–e1006636.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  248. Chambers BE, Gerlach GF, Clark EG, Chen KH, Levesque AE, Leshchiner I, et al. Tfap2a is a novel gatekeeper of nephron differentiation during kidney development. Development. 2019;146:dev172387.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  249. Kuckenberg P, Kubaczka C, Schorle H. The role of transcription factor Tcfap2c/TFAP2C in trophectoderm development. Reprod Biomed Online. 2012;25:12–20.

    Article  CAS  PubMed  Google Scholar 

  250. Milunsky JM, Maher TA, Zhao G, Roberts AE, Stalker HJ, Zori RT, et al. TFAP2A mutations result in Branchio-Oculo-facial syndrome. The Am J Hum Genet. 2008;82:1171–7.

    Article  CAS  PubMed  Google Scholar 

  251. Huang HX, Yang G, Yang Y, Yan J, Tang XY, Pan Q. TFAP2A is a novel regulator that modulates ferroptosis in gallbladder carcinoma cells via the Nrf2 signalling axis. Eur Rev Med Pharmacol Sci. 2020;24:4745–55.

    PubMed  Google Scholar 

  252. Li Q, Dashwood RH. Activator Protein 2α associates with adenomatous polyposis Coli/β-catenin and inhibits β-Catenin/T-cell factor transcriptional activity in colorectal cancer cells. J Biol Chem. 2004;279:45669–45669.

    Article  CAS  PubMed  Google Scholar 

  253. Makhov PB, Golovine KV, Kutikov A, Canter DJ, Rybko VA, Roshchin DA, et al. Reversal of epigenetic silencing of AP-2alpha results in increased zinc uptake in DU-145 and LNCaP prostate cancer cells. Carcinogenesis. 2011;32:1773–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  254. Hallberg AR, Vorrink SU, Hudachek DR, Cramer-Morales K, Milhem MM, Cornell RA, et al. Aberrant CpG methylation of the TFAP2A gene constitutes a mechanism for loss of TFAP2A expression in human metastatic melanoma. Epigenetics. 2014;9:1641–7.

    Article  PubMed  Google Scholar 

  255. Lian W, Zhang L, Yang L, Chen W. AP-2α reverses vincristine-induced multidrug resistance of SGC7901 gastric cancer cells by inhibiting the Notch pathway. Apoptosis. 2017;22:933–41.

    Article  CAS  PubMed  Google Scholar 

  256. Su W, Xia J, Chen X, Xu M, Nie L, Chen N, et al. Ectopic expression of AP-2α transcription factor suppresses glioma progression. Int J Clin Exp Pathol. 2014;7:8666–8666.

    PubMed  PubMed Central  Google Scholar 

  257. Sotiriou C, Wirapati P, Loi S, Harris A, Fox S, Smeds J, et al. Gene expression profiling in breast cancer: understanding the molecular basis of histologic grade to improve prognosis. J Natl Cancer Inst. 2006;98:262–72.

    Article  CAS  PubMed  Google Scholar 

  258. Shi D, Xie F, Zhang Y, Tian Y, Chen W, Fu L, et al. TFAP2A regulates nasopharyngeal carcinoma growth and survival by targeting HIF-1α signaling pathway. Cancer Prev Res (Phila). 2014;7:266–77.

    Article  CAS  Google Scholar 

  259. Bennett KL, Romigh T, Eng C. AP-2alpha induces epigenetic silencing of tumor suppressive genes and microsatellite instability in head and neck squamous cell carcinoma. PLoS ONE. 2009;4:e6931.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  260. Ding X, Yang Z, Zhou F, Wang F, Li X, Chen C, et al. Transcription factor AP-2α regulates acute myeloid leukemia cell proliferation by influencing Hoxa gene expression. Int J Biochem Cell Biol. 2013;45:1647–56.

    Article  CAS  PubMed  Google Scholar 

  261. Carrière C, Mirocha S, Deharvengt S, Gunn JR, Korc M. Aberrant expressions of AP-2α splice variants in pancreatic cancer. Pancreas. 2011;40:695–700.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  262. Schulte JH, Kirfel J, Lim S, Schramm A, Friedrichs N, Deubzer HE, et al. Transcription factor AP2alpha (TFAP2a) regulates differentiation and proliferation of neuroblastoma cells. Cancer Lett. 2008;271:56–63.

    Article  CAS  PubMed  Google Scholar 

  263. Yang Y-L, Zhao L-Y. AP-2 family of transcription factors: critical regulators of human development and cancer. J Cancer Treat Diagn. 2021;5:1–4.

    Article  CAS  Google Scholar 

  264. Kaiser S, Koch Y, Kühnel E, Sharma N, Gellhaus A, Kuckenberg P, et al. Reduced gene dosage of Tfap2c impairs trophoblast lineage differentiation and alters maternal blood spaces in the mouse placenta. Biol Reprod. 2015;93:31–2.

    Article  PubMed  CAS  Google Scholar 

  265. Satoda M, Zhao F, Diaz GA, Burn J, Goodship J, Davidson HR, et al. Mutations in TFAP2B cause Char syndrome, a familial form of patent ductus arteriosus. Nat Genet. 2000;25:42–6.

    Article  CAS  PubMed  Google Scholar 

  266. Gao SL, Wang LZ, Liu HY, Liu DL, Xie LM, Zhang ZW. miR-200a inhibits tumor proliferation by targeting AP-2γ in neuroblastoma cells. Asian Pac J Cancer Prev. 2014;15:4671–6.

    Article  PubMed  Google Scholar 

  267. Hoei-Hansen CE, Nielsen JE, Almstrup K, Sonne SB, Graem N, Skakkebaek NE, et al. Transcription factor AP-2gamma is a developmentally regulated marker of testicular carcinoma in situ and germ cell tumors. Clin Cancer Res. 2004;10:8521–30.

    Article  CAS  PubMed  Google Scholar 

  268. Lal G, Contreras PG, Kulak M, Woodfield G, Bair T, Domann FE, et al. Human Melanoma cells over-express extracellular matrix 1 (ECM1) which is regulated by TFAP2C. PLoS ONE. 2013;8:e73953.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  269. Williams CMJ, Scibetta AG, Friedrich JK, Canosa M, Berlato C, Moss CH, et al. AP-2gamma promotes proliferation in breast tumour cells by direct repression of the CDKN1A gene. EMBO J. 2009;28:3591–601.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  270. Cheng C, Ying K, Xu M, Zhao W, Zhou Z, Huang Y, et al. Cloning and characterization of a novel human transcription factor AP-2 beta like gene (TFAP2BL1). Int J Biochem Cell Biol. 2002;34:78–86.

    Article  CAS  PubMed  Google Scholar 

  271. Zhao F, Lufkin T, Gelb BD. Expression of Tfap2d, the gene encoding the transcription factor Ap-2 delta, during mouse embryogenesis. Gene Expr Patterns. 2003;3:213–7.

    Article  CAS  PubMed  Google Scholar 

  272. Jain S, Glubrecht DD, Germain DR, Moser M, Godbout R. AP-2ε expression in developing retina: contributing to the molecular diversity of amacrine cells. Sci Rep. 2018;8:1–13.

    Article  Google Scholar 

  273. Sun L, Zhao Y, Gu S, Mao Y, Ji C, Xin X. Regulation of the HMOX1 gene by the transcription factor AP-2δ with unique DNA binding site. Mol Med Rep. 2014;10:423–8.

    Article  CAS  PubMed  Google Scholar 

  274. Fraune C, Harms L, Büscheck F, Höflmayer D, Tsourlakis MC, Clauditz TS, et al. Upregulation of the transcription factor TFAP2D is associated with aggressive tumor phenotype in prostate cancer lacking the TMPRSS2:ERG fusion. Mol Med. 2020;26:1–13.

    Article  CAS  Google Scholar 

  275. Wenke AK, Bosserhoff AK. Roles of AP-2 transcription factors in the regulation of cartilage and skeletal development. FEBS J. 2010;277:894–902.

    Article  CAS  PubMed  Google Scholar 

  276. Lin JM, Taroc EZM, Frias JA, Prasad A, Catizone AN, Sammons MA, et al. The transcription factor Tfap2e/AP-2ε plays a pivotal role in maintaining the identity of basal vomeronasal sensory neurons. Dev Biol. 2018;441:67–82.

    Article  CAS  PubMed  Google Scholar 

  277. Enomoto T, Ohmoto M, Iwata T, Uno A, Saitou M, Yamaguchi T, et al. Development/Plasticity/Repair Bcl11b/Ctip2 controls the differentiation of vomeronasal sensory neurons in mice. J Neurosci. 2011. https://doi.org/10.1523/JNEUROSCI.1245-11.2011.

    Article  PubMed  PubMed Central  Google Scholar 

  278. Wenke AK, Grässel S, Moser M, Bosserhoff AK. The cartilage-specific transcription factor Sox9 regulates AP-2ε expression in chondrocytes. FEBS J. 2009;276:2494–504.

    Article  CAS  PubMed  Google Scholar 

  279. Feng W, Simoes-de-Souza F, Finger TE, Restrepo D, Williams T. Disorganized olfactory bulb lamination in mice deficient for transcription factor AP-2e{open}. Mol Cell Neurosci. 2009;42:161–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  280. Niebler S, Bosserhoff AK. The transcription factor activating enhancer-binding protein epsilon (AP-2ε) regulates the core promoter of type II collagen (COL2A1). FEBS J. 2013;280:1397–408.

    Article  CAS  PubMed  Google Scholar 

  281. Wenke AK, Rothhammer T, Moser M, Bosserhoff AK. Regulation of integrin alpha10 expression in chondrocytes by the transcription factors AP-2epsilon and Ets-1. Biochem Biophys Res Commun. 2006;345:495–501.

    Article  CAS  PubMed  Google Scholar 

  282. Ebert MPA, Tänzer M, Balluff B, Burgermeister E, Kretzschmar AK, Hughes DJ, et al. TFAP2E-DKK4 and chemoresistance in colorectal cancer. N Engl J Med. 2012;366:44–53.

    Article  CAS  PubMed  Google Scholar 

  283. Hoshi R, Watanabe Y, Ishizuka Y, Hirano T, Nagasaki-Maeoka E, Yoshizawa S, et al. Depletion of TFAP2E attenuates adriamycin-mediated apoptosis in human neuroblastoma cells. Oncol Rep. 2017;37:2459–64.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Nour Aldin Kahlous, a Ph.D. student at the department of Cell and Molecular Biology, Computational Biology and Bioinformatics, Uppsala University, for his valuable suggestions on the druggability part of this review.

Funding

Open access funding provided by Uppsala University. HBS is supported by the Swedish Research Council, The Swedish Brain Research Foundation and the Novo Nordisk Foundation.

Author information

Authors and Affiliations

Authors

Contributions

MHA-S, MN, MW contributed to the conception of the manuscript. The first draft of the manuscript was written by MHA-S and MN. LEC, MMA, MJW, MR-A, JM and HBS commented on previous versions of the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Helgi B. Schiöth.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Al-Sabri, M.H., Nikpour, M., Clemensson, L.E. et al. The regulatory role of AP-2β in monoaminergic neurotransmitter systems: insights on its signalling pathway, linked disorders and theragnostic potential. Cell Biosci 12, 151 (2022). https://doi.org/10.1186/s13578-022-00891-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13578-022-00891-7

Keywords