Wnt pathways are critical developmental signaling cascades that are conserved across multicellular life. A clear role for Wnt signaling in proper neural development has been well-established, yet less is known about its sustained expression and signaling in the mature nervous system. The precise role for Wnt pathways, canonical or otherwise, and individual Wnt components (ligands, receptors, transducers, effectors, and regulators) in the mature brain are poorly understood. However, genetic evidence implicating Wnt-related components in both neurodevelopmental and neurodegenerative disorders suggests that fine-tuned regulation of Wnt signaling is required for proper nervous system development and long-term homeostasis. Much has been documented about down-regulated Wnt signaling and its association with neurological conditions. Hence, the focus of this review is to consolidate and highlight the evidence for up-regulated Wnt transcription and/or signaling in neurodevelopmental and neurodegenerative disorders with a brief discussion on the role of deregulated Wnt in cancer. Finally, we touch upon the therapeutic prospect of Wnt inhibition in the nervous system.

The wingless/integrated (Wnt) family of genes encode highly conserved secreted ligands that are critical for embryogenesis as well as tissue development and homeostasis [1]. These morphogens play an important role in developmental patterning and have many diverse functions in controlling proliferation, migration, differentiation, polarity, and death. The ligand was first discovered in Drosophila melanogaster as the wingless (wg) gene [2] and established a role in developmental patterning [3]. The name ‘Wnt’ comes from a fusion of the name of the Drosophila gene wg and the name of the vertebrate homolog, integrated, or int-1 [4]. In mammals, 19 Wnt ligands have been identified [5]. These ligands are highly conserved across multicellular animals in both vertebrates and invertebrates. Although there is a recent appreciation for an increasing number of Wnt pathways [6], there are three main cascades: the Wnt/β-catenin-dependent, also known as the canonical pathway, the Wnt/planar cell polarity (PCP) pathway, and the Wnt/Ca2+ pathway (Figure 1).

Wnt signaling pathways.

Figure 1:
Wnt signaling pathways.

In the β-catenin-dependent pathway, Wnt is considered ‘off’ when Wnt is not bound to FZD receptors and LRP5/6 co-receptors. β-catenin associates with the destruction complex (APC, Axin, CK1, GSK33), where it is phosphorylated by GSK-3 and CK1, followed by subsequent degradation via the proteasome. Wnt is considered ‘on’ when a Wnt ligand binds to LRP5/6 and FZD receptors, and the scaffold protein Dvl recruits Axin along with the kinases CK1 and GSK-3 to the membrane. This disrupts the formation of the destruction complex, preventing the phosphorylation and degradation of β-catenin. As a result, β-catenin accumulates in the cytoplasm and then translocates to the nucleus, where it activates transcription of Wnt target genes. In the Wnt/PCP pathway, small G proteins RhoA and RAC are activated via Dvl, resulting in signaling via the ROCK and Jnk signaling pathways. In the Wnt/Ca2+ signaling pathway, signaling is via heterotrimeric G proteins that activate PLC, which leads to a rise in intracellular Ca2+. This leads to the activation of calcineurin (Ca), calcium/calmodulin-dependent kinase II (CamKII), and PKC. Both CamKII and PKC activate various regulatory proteins such as NF-κB and CREB, and Cn can activate the cytoplasmic protein NFAT. CamKII, calcium/calmodulin-dependent kinase II; Wnt, wingless/integrated.

Figure 1:
Wnt signaling pathways.

In the β-catenin-dependent pathway, Wnt is considered ‘off’ when Wnt is not bound to FZD receptors and LRP5/6 co-receptors. β-catenin associates with the destruction complex (APC, Axin, CK1, GSK33), where it is phosphorylated by GSK-3 and CK1, followed by subsequent degradation via the proteasome. Wnt is considered ‘on’ when a Wnt ligand binds to LRP5/6 and FZD receptors, and the scaffold protein Dvl recruits Axin along with the kinases CK1 and GSK-3 to the membrane. This disrupts the formation of the destruction complex, preventing the phosphorylation and degradation of β-catenin. As a result, β-catenin accumulates in the cytoplasm and then translocates to the nucleus, where it activates transcription of Wnt target genes. In the Wnt/PCP pathway, small G proteins RhoA and RAC are activated via Dvl, resulting in signaling via the ROCK and Jnk signaling pathways. In the Wnt/Ca2+ signaling pathway, signaling is via heterotrimeric G proteins that activate PLC, which leads to a rise in intracellular Ca2+. This leads to the activation of calcineurin (Ca), calcium/calmodulin-dependent kinase II (CamKII), and PKC. Both CamKII and PKC activate various regulatory proteins such as NF-κB and CREB, and Cn can activate the cytoplasmic protein NFAT. CamKII, calcium/calmodulin-dependent kinase II; Wnt, wingless/integrated.

Close modal

Wnt pathways

The canonical Wnt/β-catenin pathway initiates diverse signaling pathways through Wnt molecules acting as ligands that primarily interact with Frizzled (Fzd) receptors, which are seven-transmembrane G protein-coupled receptors [7]. Without active Wnt signaling, the destruction complex, composed of Axin, APC, and GSK-3, constitutively phosphorylates β-catenin, leading to its ubiquitination and proteasomal degradation. Upon secretion of Wnt from a signal-sending cell, Wnt binds Fzd and the LRP5/6 co-receptors on the signal-receiving cell. This leads to the cytosolic protein dishevelled (Dvl) becoming activated and inhibiting the destruction complex. β-catenin subsequently stabilizes, accumulates, and translocates to the nucleus, forming a transcriptional complex with Tcf./Lef family transcription factors to activate Wnt-responsive genes [8]. A growing list of regulators and interactors modulate the Wnt/β-catenin pathway. Dickkopf (DKK) inhibits Wnt signaling by binding to LRP5/6. Secreted Fzd-related proteins (SFRPs) and Wnt inhibitory factor (WIF) sequester Wnt ligands extracellularly, adding layers of regulation that define Wnt activity in a context-dependent manner pre- and post-development [9].

The Wnt/PCP pathway involves three main steps [10]. First, upstream PCP components such as Four-jointed, Dachsous, Fat, and Atrophin provide a long-range signal for global polarity direction [11]. Core PCP components such as Fzd, Dvl, Celsr, Vangl, and Prickle establish planar polarity within various cells of the body. Specifically, this is done by directing asymmetric localization at cell membranes. This co-ordination involves Fzd (the receptor for the PCP signal) and Dvl (the transducer of the signal) accumulating on one side of the cell with Vangl (membrane component) and Prickle (cytoplasmic component) on the opposite side, as Celsr facilitates intercellular communication to ensure the propagation of polarity across tissues [12,13]. PCP signaling plays a vital role in neural tube closure, co-ordinated ciliogenesis, and the organization of epithelial tissues, highlighting its importance in normal development and the maintenance of tissue structure. Some of these processes are orchestrated by downstream PCP effectors such as Daam1, Rho, Rac, Rho, JNK, and Profilin [14-17].

Finally, the Wnt/Ca2+ pathway was identified when injections of Wnt5a or Wnt11 mRNA into zebrafish embryos increased calcium transients in the blastodisc [18,19]. Wnt-Fzd binding activates phospholipase C (PLC) via G proteins, increasing intracellular Ca2+ levels and activating downstream effectors like protein kinase C (PKC). This pathway is crucial for development and is implicated in cancer [20].

To highlight the key differences between the Wnt pathways, the canonical Wnt/β-catenin pathway primarily regulates long-term processes such as gene expression, neurogenesis, cell proliferation, and synaptic stability. In contrast, the non-canonical pathways control immediate cellular responses, such as cell polarity, calcium maintenance, cell migration, cytoskeletal dynamics, and inflammation. In neurodevelopmental and neurodegenerative disorders, these pathways can interact synergistically or antagonistically. This highlights the need for fine-tuned regulation to maintain neural health [21,22].

Wnt function

Proper Wnt signaling is essential for embryogenesis, development, cell proliferation, migration, and other processes [1]. It is also important for cell maintenance and survival [23]. Wnt is crucial for key neurodevelopmental processes such as cell proliferation, dendrite development, differentiation, migration, synaptogenesis, and axon guidance, while also maintaining synaptic plasticity and neural integrity in the mature brain [24,25]. Due to its broad role in proper brain development, when altered Wnt signaling occurs, there is a potential for disorders of the brain such as neurodevelopmental and neurodegenerative diseases.

Wnt activity varies based on temporal and cell-specific factors, such as neurons or glia, and in development or the mature nervous system [26,27]. An example of this in the context of neurodegeneration would be the hyperactivation of Wnt signaling in glial cells, which may exacerbate chronic inflammation and contribute to neuronal dysfunction in Alzheimer’s disease (AD) and frontotemporal dementia [28,29]. In comparison, in neurodevelopmental disorders (NDDs) such as autism spectrum disorder (ASD), excessive Wnt activity may disrupt synaptic pruning and lead to abnormal connectivity [30]. This variability shows the importance of tailoring therapeutic strategies to the specific context of Wnt dysregulation in each condition, as interventions should address not only the underlying molecular mechanisms but also the timing and cellular environment where Wnt signaling operates. Advances in inducible and conditional animal models, such as adult-specific knockout/knock-in systems, are critical tools for dissecting these complex dynamics.

Building on the diverse functions of Wnt signaling, its role in brain development highlights how tightly regulated Wnt activity is essential for processes such as neural specification, cell migration, and synaptic formation, which are critical for establishing functional neural circuits [25,31]. Canonical Wnt plays an important role in numerous homeostatic functions and is critical for early proper central nervous system (CNS) development. Haploinsufficiency of the gene CTNNB1 that encodes β-catenin causes intellectual disability and NDDs, where β-catenin is critical for neural specification and synaptic development [32,33]. Another example of this would be the loss of function mutations in the gene FZD3 (a gene that regulates axon guidance and neuronal migration) associated with schizophrenia [34]. Lack of Wnt1 causes severe midbrain and cerebellar atrophy in mice [35]. Mutations in the Wnt secretion mediator wntless (WLS) in humans cause multiorgan defects, including microcephaly [36]. Hence, core Wnt components are critical for development, and their loss is associated with nervous system disorders.

However, what about overactivation of Wnt? The sustained expression of Wnt signaling in the mature nervous system is important for maintaining homeostasis as it regulates processes such as synaptic plasticity, neurogenesis, neuronal survival, glial cell function, and inflammation, all of which are vital for the stability and proper functioning of the nervous system [22,25]. However, proper negative regulation of Wnt is also crucial. Dkk1 knockout mice, lacking anterior head structures, demonstrate the need for Wnt inhibition during axis formation and limb morphogenesis [37]. Loss of the destruction complex member, APC2, leads to lissencephaly, characterized by intellectual disability and neuromotor impairments [38]. LRP6 gene variants in infants with spina bifida showed an increase in noncanonical Wnt/PCP signaling [39]. An excess of Wnt signaling, particularly through the canonical Wnt/β-catenin pathway, can disrupt the precise cellular processes required for neural tube closure, which can cause congenital neurodevelopmental abnormalities such as neural tube defects [40,41]. The following review highlights specific examples of when the Wnt pathway is deregulated in CNS disorders, including neurodevelopmental, neurodegenerative, and neurooncological conditions. First, we will discuss excess Wnt in rare Mendelian NDDs.

Excess Wnt in rare Mendelian NDDs

NDDs encompass a wide range of conditions that affect the growth and development of the CNS [42,43]. Within this spectrum, the literature often distinguishes between rare Mendelian subtypes and more common NDDs. Rare Mendelian conditions, such as Fragile X and Angelman syndromes, result from specific single mutations. These contrast with more prevalent clinical groups, including ASD, epilepsy, and schizophrenia, which are generally thought to have complex, heterogeneous, and polygenic inheritance patterns, although they can also be caused by single gene mutations [44]. Next-generation sequencing, such as whole-genome, exome, or long-read sequencing, has helped increase the identification of gene variants causing NDDs [45]. In both rare NDDs and common disorders of neurodevelopment such as ASD, there is an increased burden of de novo variants in affected individuals [46,47]. Recent advances in understanding the molecular basis of these disorders have provided renewed hope for achieving more precise and timely diagnoses and identified the genes and pathways implicated in NDDs.

Table 1 provides a list of rare NDDs associated with specific genetic variants that disrupt the Wnt signaling pathway. As mentioned above, loss of APC2 leads to a congenital lissencephaly. In Apc2 null mice, disrupted neuronal migration results in defects in the lamination of the cerebral cortex and cerebellum, which manifest as lissencephaly, characterized by a smooth brain surface, severe intellectual disability, seizures, and motor dysfunction [38,64]. Hence, the loss of APC2 implicates increased Wnt/β-catenin in lissencephaly. Upon Wnt stimulation, tuberous sclerosis complex 2 (TSC2) represses β-catenin-dependent transcriptional activity. This suggests that certain phenotypic features of the TSC, such as benign tumors (hamartomas), seizures, skin abnormalities, and cognitive impairment, may arise from abnormal β-catenin signaling caused by mutations in the TSC2 [61,62,65]. De novo truncations in the intronless gene interferon regulatory factor 2 binding protein-like (IRF2BPL) cause neurodevelopmental disorder with regression, abnormal movements, loss of speech, and seizures (NEDAMSS) [66]. Recent studies in the Drosophila and zebrafish CNS and patient chemically reprogrammed astrocytes show increased transcription of WNT1 and subsequent activation of downstream Wnt signaling pathways. Moreover, pharmacological suppression of Wnt/β-catenin signaling mitigated neurobehavioral phenotypes in fruit flies and zebrafish [52]. This identified IRF2BPL as a negative regulator of Wnt/β-catenin signaling and its dysregulation in NEDAMSS. Additionally, mutations in UBE3A, which is a HECT domain E3 ubiquitin ligase, are linked to ASD, Angelman syndrome, and cancer. Through quantitative proteomics and reporter assays, it has been shown that a de novo autism-linked UBE3A variant p.T485A can activate Wnt/β-catenin signaling to a greater extent in comparison with the wild type [67]. Moreover, CHD8 haploinsufficiency causes intellectual disability, ASD, and macrocephaly. CHD8 negatively regulates β-catenin-targeted gene expression by promoting the association of β-catenin and histone H1 [49,68,69]. Moreover, pathogenic variants in CHD2, also known as N-cadherin, lead to a rare disease known as agenesis of the corpus callosum, cardiac, ocular, and genital syndrome [48]. Relatedly, CHD2 negatively regulates canonical Wnt signaling by forming a molecular complex with Axin and LRP5, thus leading to increased β-catenin degradation [70]. Finally, damaging variants in PRICKLE1 are associated with progressive myoclonic epilepsy, and it has been shown to negatively regulate Wnt/β-catenin signaling through binding with Dvl and facilitating Dvl ubiquitination and subsequent degradation [53]. In sum, these examples show how deregulation of Wnt/β-catenin signaling is a likely relevant pathogenic signature of a subset of Mendelian NDDs.

Table 1:
Mendelian neurological disorders associated with deregulated Wnt signaling.
GeneNervous system-related disorderInheritanceMIM#Role in Wnt regulationReferences
APC2 Cortical dysplasia, complex, with other brain malformations 10 AR 618,677 Degrades β-catenin [38
CDH2 Agenesis of corpus callosum, cardiac, ocular, and genital syndrome AD 618,929 Interacts with axin and LRP5 increasing β-catenin degradation [48
CHD8 Autism spectrum disorder 18 (risk) AD 615,032 Negatively regulates β-catenin gene expression [49
CUL3 Neurodevelopmental disorder with or without autism or seizures AD 619,239 Ubiquitination of dishevelled [50
FUZ Neural tube defects (risk) AD 182,940 Increased Wnt/β-catenin signaling in Fuz-/- mouse embryos [51
IRF2BPL Neurodevelopmental disorder with regression, abnormal movements, loss of speech, and seizure AD 618,088 Ubiquitinate β-catenin, also interacts with Ck1a [52
PRICKLE1 Epilepsy, progressive myoclonic 1B AR 612,437 Prickle-1 bound with Dvl3 and facilitated Dvl3 ubiquitination/degradation [53
PRKN Parkinson disease, juvenile, type 2 AR 600,116 PRKN regulates beta-catenin protein levels [54
PSMD12 Stankiewicz-Isidor syndrome AD 617,516 Up-regulation of some proteasome proteins [55
RNF213 Moyamoya disease risk AD / AR 607,151 Ubiquitination and degradation of FLNA and NFATC2 in nooncannolcal Wnt Ca [56
SCYL2 Arthrogryposis multiplex congenita 4, neurogenic, with agenesis of the corpus callosum AR 618,766 SCYL2 selectively induces the lysosomal degradation of Frizzled5 [57
SOX10 Peripheral demyelinating neuropathy, central dysmyelination, Waardenburg syndrome, and Hirschsprung disease AD 609,136 SOX10 competes with Tcf.4 to bind β-catenin and transrepresses its downstream target genes via its own DNA-binding [58
SOX2 Optic nerve hypoplasia and abnormalities of the central nervous system AD 206,900 SOX2 disrupts β-catenin-Tcf./LEF mediated transcriptional activation [59
TLE1 Microcephaly and a severe neurodevelopmental disorder? AR  Tcf. repressor [60
TSC2 Tuberous sclerosis-2 AD 613,254 Associates with GSK3 and axin and promotes β-catenin degradation [61,62
WWOX Developmental and epileptic encephalopathy 28 AR 616,211 Preventing the nuclear import of the Dvl proteins [63
GeneNervous system-related disorderInheritanceMIM#Role in Wnt regulationReferences
APC2 Cortical dysplasia, complex, with other brain malformations 10 AR 618,677 Degrades β-catenin [38
CDH2 Agenesis of corpus callosum, cardiac, ocular, and genital syndrome AD 618,929 Interacts with axin and LRP5 increasing β-catenin degradation [48
CHD8 Autism spectrum disorder 18 (risk) AD 615,032 Negatively regulates β-catenin gene expression [49
CUL3 Neurodevelopmental disorder with or without autism or seizures AD 619,239 Ubiquitination of dishevelled [50
FUZ Neural tube defects (risk) AD 182,940 Increased Wnt/β-catenin signaling in Fuz-/- mouse embryos [51
IRF2BPL Neurodevelopmental disorder with regression, abnormal movements, loss of speech, and seizure AD 618,088 Ubiquitinate β-catenin, also interacts with Ck1a [52
PRICKLE1 Epilepsy, progressive myoclonic 1B AR 612,437 Prickle-1 bound with Dvl3 and facilitated Dvl3 ubiquitination/degradation [53
PRKN Parkinson disease, juvenile, type 2 AR 600,116 PRKN regulates beta-catenin protein levels [54
PSMD12 Stankiewicz-Isidor syndrome AD 617,516 Up-regulation of some proteasome proteins [55
RNF213 Moyamoya disease risk AD / AR 607,151 Ubiquitination and degradation of FLNA and NFATC2 in nooncannolcal Wnt Ca [56
SCYL2 Arthrogryposis multiplex congenita 4, neurogenic, with agenesis of the corpus callosum AR 618,766 SCYL2 selectively induces the lysosomal degradation of Frizzled5 [57
SOX10 Peripheral demyelinating neuropathy, central dysmyelination, Waardenburg syndrome, and Hirschsprung disease AD 609,136 SOX10 competes with Tcf.4 to bind β-catenin and transrepresses its downstream target genes via its own DNA-binding [58
SOX2 Optic nerve hypoplasia and abnormalities of the central nervous system AD 206,900 SOX2 disrupts β-catenin-Tcf./LEF mediated transcriptional activation [59
TLE1 Microcephaly and a severe neurodevelopmental disorder? AR  Tcf. repressor [60
TSC2 Tuberous sclerosis-2 AD 613,254 Associates with GSK3 and axin and promotes β-catenin degradation [61,62
WWOX Developmental and epileptic encephalopathy 28 AR 616,211 Preventing the nuclear import of the Dvl proteins [63

MIM# from Mendelian inheritance in man.

Excess Wnt in ASD

ASD is a common condition characterized by a range of social communication deficits and restricted behaviors [71]. A notable 30–40% of individuals with ASD are comorbid with NDDs, such as seizures and motor anomalies. The etiology of ASD is complex, but twin studies and cohort studies have consistently demonstrated that genetic factors play a significant role in the etiology of ASD [72-74].

One area of significant interest in ASD research is the expression of genes associated with Wnt signaling regulation in ASD brains [75]. The convergence of gain and loss of function in Wnt/β-catenin signaling pathways in the context of ASD is particularly noteworthy. The essential function of β-catenin in brain development, growth, and the differentiation and proliferation pathways of neuronal progenitors underscores the potential role of Wnt signaling in ASD development [76]. This suggests that a wide spectrum of Wnt signaling activation may underlie the diverse manifestations of ASD. Over half of the top 102 ASD-related genes [77] (59 genes) are associated with the Wnt signaling pathway, with 18/59 acting as known negative regulators (Figure 2).

Wnt in ASD.

Figure 2:
Wnt in ASD.

Manually curated association of 59/102 top ASD genes and 16 negative regulators with known roles in Wnt/β-catenin signaling. ASD, Autism spectrum disorders; Wnt, wingless/integrated.

Figure 2:
Wnt in ASD.

Manually curated association of 59/102 top ASD genes and 16 negative regulators with known roles in Wnt/β-catenin signaling. ASD, Autism spectrum disorders; Wnt, wingless/integrated.

Close modal

Increasing evidence suggests that dysregulated Wnt signaling plays a crucial role in the neurodevelopmental alterations observed in ASD. Several genes, including TBL1XR1, MED13L, and PTK7 (Figure 2), provide insight into how increased Wnt/β-catenin activity may contribute to ASD pathology. TBL1XR1 is part of the NCoR/SMRT complex, which regulates Wnt/β-catenin signaling by modulating transcriptional repression, and mutations in TBL1XR1 have been implicated in ASD and intellectual disabilities [78]. Similarly, MED13L, a component of the Mediator complex that regulates Wnt/β-catenin signaling [79], has been linked to ASD and intellectual disability [80]. Finally, PTK7 is a key regulator of Wnt signaling, particularly in the PCP pathway and proper embryogenesis [81] and has been associated with neural tube defects and ASD [82]. Together, these genes highlight the role of deregulated Wnt pathways in ASD and provide potential targets for further research and therapeutic intervention.

Excess Wnt in other neuropsychiatric disorders

Major psychiatric disorders such as schizophrenia, bipolar affective disorder (BPAD), and attention-deficit hyperactivity disorder (ADHD) are common and particularly debilitating conditions. These disorders are increasingly recognized as caused by subtle dysregulations of CNS development, particularly concerning synapse formation and maintenance. However, our understanding of the biological basis for these psychiatric disorders remains limited [83]. Since Wnt signaling plays a crucial role in organizing the developing brain, it is not surprising that molecular links have tied these conditions to Wnt [84].

Schizophrenia has an estimated heritability of 65–80% and is characterized by ‘positive’ symptoms such as delusions, hallucinations, and disorganized speech, as well as ‘negative’ symptoms, including a lack of emotional affect and motivation [85]. Neuroanatomical abnormalities in schizophrenia, though subtle, include enlarged lateral ventricles, disorganized forebrain structures, loss of neuropil, and fewer synapses in pyramidal neurons [86,87]. The neurodevelopmental hypothesis suggests that these abnormalities arise from dysregulated brain development, with the Wnt signaling pathway being a key candidate for such dysregulation [88,89]. Given Wnt signaling’s critical role in brain regionalization, hippocampal development, neuronal proliferation, migration, and synapse formation, its dysregulation could lead to hippocampal volume reduction, forebrain disorganization, and synaptic defects such as those observed in schizophrenia [84]. The WNT1 ligand has been reported to be up-regulated in individuals with schizophrenia [90], and several schizophrenia-associated genes are core components of the Wnt/β-catenin signaling pathway. Notably, a single nucleotide polymorphism (SNP) in the Wnt/β-catenin-activated transcription factor Tcf.4 shows a significant genome-wide correlation with schizophrenia [91]. Tcf.4 overexpression has been linked to memory impairments and deficits in prepulse inhibition, a neurophysiological correlate of schizophrenia and other psychiatric disorders [92]. Moreover, down-regulation of DKK proteins and GSK3β, both negative regulators of Wnt signaling, are genetically and pharmacologically connected to schizophrenia [93-97]. Together, a gain of Wnt/β-catenin signaling is associated with schizophrenia. However, it is known that the loss of disrupted in schizophrenia 1 (DISC1), a scaffold protein involved in synaptic development and neural migration, potentiates Wnt/β-catenin by inhibiting the destruction complex member GSK3β [98-100]. Thus, Wnt signaling loss has also been implicated in schizophrenia.

BPAD has an estimated heritability of 60–85% and is characterized by alternating phases of mania and depression [101]. Similar to schizophrenia, genomic studies have revealed disruptions in the Wnt/β-catenin signaling pathway in BPAD, implicating genes such as DISC1, WNT7A, WNT2B, and Tcf.7L2 [102-107]. Additionally, SNP-based family studies have identified the Wnt/β-catenin target gene PPARD and the high-mobility group (HMG) box family member HMG2L1, the latter functioning as a negative regulator of Wnt/β-catenin signaling, as significantly associated with BPAD [107-109]. A recent genome-wide association study (GWAS) identified a noncoding SNP in the SEC14 and spectrin domain-containing 1 (SESTD1) locus, strongly linked to lithium-responsive BPAD. SESTD1, involved in noncanonical Wnt/PCP signaling, interacts with key proteins during neurodevelopment and in the mature CNS [107,110].

ADHD is a prevalent neurodevelopmental condition marked by symptoms of inattention, hyperactivity, and impulsivity with an estimated heritability of 77 to 88% [111,112]. ADHD affects over 5% of the global population and often coexists with other conditions, including but not limited to ASD [113]. The etiology and pathophysiology of ADHD remain largely unknown, though emerging evidence points to the interplay of genetic and environmental factors, particularly those involving the Wnt signaling pathway [114]. A recent study investigated the timing and extent of Wnt pathway alterations in ADHD across different developmental stages. Researchers performed proteomic and functional analyses on induced pluripotent stem cells (iPSCs), neural stem cells (NSCs), and differentiated neurons. Although no differences were observed at the iPSC stage, ADHD NSCs displayed altered protein expression of GSK3β and β-catenin, indicating increased Wnt activity. This heightened Wnt activity in NSCs was confirmed by reporter assays and correlated with genetic and clinical ADHD traits [114]. In addition, in the thyroid-hormone responsive-overexpressing ADHD mouse model, hippocampal proteomic analysis revealed up-regulation of Ctnnb1 (encoding β-catenin) and other changes in Wnt-related genes, including reduced Wnt7a and increased Wnt inhibitors Dkk4 and Igfbp5 [115]. On the other hand, research has shown loss of Wnt/β-catenin signaling in immortalized patient lymphoblasts with treatment of the Wnt agonist methylphenidate [116]. Human studies have linked sex-specific genetic variations in LRP5/6 co-receptors with ADHD, further implicating Wnt signaling [117]. Additionally, in a large meta-GWAS, several genes (e.g., DUSP6, SEMA6D, ST3GAL3, FOXP1 and FOXP2, and SORCS3) linked to the Wnt pathways (canonical and non-canonical) were found to be associated with ADHD, further supporting the role of this pathway [118,119]. It is not necessarily clear from the studies above whether Wnt is lost or gained as the consequence of these genetic variations is not always known [115]. Regardless, these findings suggest that Wnt signaling dysregulation may occur at the NSC stage in ADHD, potentially contributing to the developmental deficits observed in patients. Further research into specific Wnt signaling changes in defined cell types will be critical for understanding and treating ADHD. Relatedly, CTNNB1 is involved in multiple NDDs such as intellectual disability, schizophrenia, and ASD [32], providing further genetic evidence linking Wnt-related components across NDDs.

It has been well-established that Wnt signaling has an important role in brain development. However, in the mature CNS, it is known for its role in regeneration and plasticity, but its involvement in long-term neuronal health and neurodegenerative diseases is less clear. Importantly, all core Wnt signaling components remain expressed in the adult CNS of most animals [120]. As the brain matures and ages, the highly controlled regulation of Wnt signaling continues to exert its influence, and recent research has shed light on the pathogenesis of the following neurodegenerative disorders. Understanding how Wnt signaling contributes to the progression of these conditions can provide valuable insights into potential therapeutic interventions.

Excess Wnt in Alzheimer’s disease

AD is the most common neurodegenerative disorder, characterized by cognitive decline, memory loss, amyloid beta (Aβ) deposition, tau aggregates, and widespread neuronal death [121]. Synapse loss is an early feature strongly associated with cognitive decline. GWAS has revealed that Wnt effectors were associated with both presenilin1 and granulin (GRN), which cause dominantly inherited forms of AD and frontotemporal dementia (FTD), respectively. Interestingly, they found that Wnt decreases GRN expression in cultured human neural progenitor cells, and knockdown of GRN resulted in an increase in Wnt expression, demonstrating the reciprocal relationship between Wnt and GRN [29]. Hence, taken together, although the down-regulation of Wnt is associated with aging and AD, there could be contexts in the CNS where deregulated Wnt contributes to neurodegeneration seen in AD and/or FTD. However, it should be noted that most evidence suggests that Wnt signaling is down-regulated in AD. Aβ may reduce Wnt signaling, contributing to synapse dysfunction and degeneration [107,108]. Studies show down-regulation of Wnt components in aging brains, with SFRP1 and Dkk1 up-regulation inhibiting Wnt pathways [122,123]. This inhibition increases Gsk3β activity and reduces β-catenin, promoting Aβ production and Tau hyperphosphorylation, both key AD features [124,125]. Thus, diminished Wnt signaling probably exacerbates AD pathology. While reduced Wnt signaling is widely implicated in aging and AD pathology, emerging evidence indicates that increased or dysregulated Wnt activity may also play a role in neurodegeneration, suggesting a more complex relationship within the CNS.

Excess Wnt in Parkinson’s disease

Parkinson’s disease (PD) is the most common movement disorder and the second most diagnosed neurodegenerative disorder after AD, as it affects almost 1% of the population over age 60 [126]. PD motor symptoms occur when the progressive and selective degeneration of midbrain dopaminergic neurons of the substantia nigra pars compacta and their projections into the caudate putamen (striatum) result in a substantial decrease in dopamine levels. The second hallmark of PD is the accumulation of pathological α-synuclein, which is a major component of Lewy bodies and Lewy neurites present in surviving dopaminergic neurons [127].

Relating to canonical Wnt/β-catenin signaling, a main feature of PD is the regression of dopaminergic neurogenesis from neurodevelopment through aging and neurodegeneration [128,129]. It has been shown that autosomal recessive mutations in the E3 ubiquitin ligase Parkin, encoded by PARK2, cause early-onset PD. Moreover, Parkin represses β-catenin by inducing both β-catenin ubiquitination and degradation [54]. The loss of Parkin can lead to the accumulation of β-catenin, resulting in up-regulation in Wnt/β-catenin signaling. Moreover, autosomal-dominant mutations in LRRK2 (leucine-rich repeat kinase 2), which is encoded by PARK8, cause PD. Relatedly, LRRK2 is an important scaffolding protein involved in the three branches of Wnt signaling [130,131]. These studies used Lrrk2 p.G2019S-mutant knock-in mice, thought of as hyperkinase gain-of-function, and the Lrrk2 knockout mice to show that both canonical and non-canonical Wnt signaling were elevated. Hence, by examining models of familial PD, excess Wnt is implicated in the pathogenesis of this debilitating movement disorder.

Excess Wnt in Huntington’s disease

Huntington’s disease (HD) is an autosomal dominantly inherited neurological condition that leads to involuntary choreatic movements, behavioral and psychiatric issues, and dementia [132]. It is a rare and progressive neurodegenerative disorder caused by an elongated CAG repeat HTT gene [133]. Multiple studies have shown that the Wnt/β-catenin pathway is altered in HD models, especially the stability and levels of β-catenin. In most cases, levels of β-catenin are increased in HD. This has been shown in multiple models, including iPSC-derived neuronal cultures [134,135], human transfected cells [136,137], mouse cell lines [138], Drosophila HD models [137,139], and post-mortem striatal samples from HD patients [137]. Additionally, HD neuronal cells derived from patient iPSCs have a significant dysregulation of several members of the Wnt/β-catenin signaling pathway, including the down-regulation of the components of the destruction complex and the increased expression of Tcf.3 and FZD transcripts, as well as the increased expression of Wnt transcriptional targets such as cyclin D1 (CCND1) [140]. This up-regulation of the gene target of the Wnt/β-catenin pathway in HD was also reproduced by other groups [135], solidifying deregulated Wnt/β-catenin signaling as an important pathogenic process in HD.

Excess Wnt in spinocerebellar ataxia type 1

Spinocerebellar ataxia type 1 (SCA1) is a dominantly inherited neurodegenerative disease that is characterized by progressive ataxia and the degeneration of specific neuronal populations such as Purkinje cells in the cerebellum, brainstem cranial nerve nuclei, and inferior olive neurons [141]. Genetically, this disease is caused by a trinucleotide repeat expansion of a glutamine-encoding CAG tract in ATXN1 [141]. In relation to Wnt, there are numerous genes that encode key components in the Wnt/β-catenin signaling pathway, including Apc, Gsk3β, Ctnnb1 (encoding β-catenin), and Lef-1, as well as its target genes, including Ccnd1 and Myc, that are expressed in adult Purkinje cells [142,143]. It has been found that ataxin-1 in a SCA1 mouse model enhances Wnt/β-catenin signaling activity at numerous levels of the pathway, prior to pathology, and that this activity increases with the size of the CAG expansion [144]. This discovery reveals an inherent pathological mechanism that enhances the activation of Wnt/β-catenin signaling in SCA1 Bergman glia (adjacent to the Purkinje cells) and likely in other cell types containing ataxin-1. Additionally, this Wnt/β-catenin enhancement potentially occurs by direct interaction with Tcf./LEF family members. Enhanced Wnt ligand secretion may also explain the dysregulation of genes associated with PCP signaling, which has been previously observed in SCA1 in previous studies [145]. Hence, the excess Wnt/β-catenin pathway is likely an important contributor to SCA1.

The above genetic evidence indicates that in sum, the complex orchestration of the availability of Wnt ligands (e.g. Wnt3a, Wnt5a) is balanced by antagonists such as DKK1 and SFRPs, which inhibit Wnt signaling. This balance is important in the prevention of both overactivation or suppression of the pathway; hence, the role of Wnt in neurodegeneration is of increasing interest [146]. The consequences of dysregulation of this organized process include cancer, neurodevelopmental, and neurodegenerative disorders. Therefore, the Wnt pathway and its players can be targeted in therapeutic strategies for various diseases, including cancer, that is discussed in the following section.

Elevated Wnt is implicated in various cancers because it plays critical roles in regulating proliferation, death, migration, and cell fate decisions. Wnt dysregulation can affect any of these processes and lead to tumorigenesis [147]. For instance, continuous activation of β-catenin increases the proliferation of mouse neural progenitor cells [147]. Also, a proviral insertion into the Wnt1 mouse gene causes excess transcription and can lead to mammary hyperplasia and tumors [148-150]. Both β-catenin-dependent and independent Wnt pathways play a role in embryonic neurogenesis by regulating NSC behavior during embryonic development [151]. Deficiencies or overexpression of Wnt pathways can have severe consequences, resulting in regional CNS defects or altered progenitor cell proliferation in the spinal cord and cerebral cortex [152,153].

Apart from having a role in neurogenesis, Wnt pathways are also involved in gliomagenesis and are also associated with the most malignant, common, and lethal brain cancer in adults called glioblastoma (GBM). According to the Cancer Genome Atlas classification, GBM has four classes: proneural, neural, classical, and mesenchymal [154]. Several components of multiple Wnt pathways are involved in the mesenchymal GBM subtype. These include two frizzled receptors, β-catenin, Tcf.7L1/2 and LEF1, transcriptional factors, E-cadherin, PLC gamma, calmodulins, calcineurin, and NFAT (nuclear factor of activated T cells) [155]. GBM is not associated with mutations in CTNNB1 or APC but instead originates from loss/deletion (copy number loss) of FAT Atypical Cadherin 1 (FAT1) [156]. FAT1 is a negative effector of β-catenin, and its loss causes an overactivation of Wnt/β-catenin signaling, resulting in GBM. This is observed in 20% of GBM cases [157].

Large-scale genomic approaches have shown that GBM cells display decreased epigenetic-dependent expression of Wnt pathway inhibitors and tumor suppressors such as WIF1s, DKKs, and SFRPs [158]. These changes occur through DNA methylation and histone modification in the promoter regions [158]. These findings were significant in explaining the role of epigenetic alterations and overexpression of Wnt components, eventually leading to tumor generation. Restoring the function of these Wnt inhibitors can be a promising strategy in controlling the proliferation of GBM.

Mutations in WNT signaling genes such as CTNNB1, APC, and AXIN1 are associated with another form of tumor found in the cerebellum called medulloblastoma (MB) [159]. β-catenin mutations in exon 3 relate to the phosphorylation site targeted by the destruction complex and result in hyperactivation of Wnt/β-catenin [158,160]. The important contrast between MB and GBM can be that most cases of MB are due to genomic mutations and not epigenetic alterations.

Numerous studies have found that several Wnt-related genes associated with ASD susceptibility also have associations with cancer [161]. For example, data obtained from the Brainbase database confirm that ARID1B, ILF2, and KMT2C are associated with glioma. What is particularly surprising about the intriguing overlap between ASD and cancer is the shared functions of these genes in chromatin remodeling and genome maintenance, as well as transcription factors [162,163]. For instance, ADNP, PAX5, FOXP1, Tcf.7L2, and TBLXR1 are transcription factors genetically implicated in both ASD and cancer [161]. Hence, a common inference made from this finding is that since people diagnosed with ASD carry alterations in similar genes to those associated with cancer, they might have an increased risk of developing cancer. However, upon examining 702 individuals with ASD, there was no increase in childhood cancer in those with ASD [164]. This finding suggests that ASD begins to develop as a consequence of errors during fetal life at critical time periods for the proliferation of neuronal precursors, while cancer incidence is significantly correlated with age, which means errors more commonly occur during adult life in cell types susceptible to tumors [161].

Although this additional dimension adds complexity to our attempts to understand ASD, looking at mechanistic similarities can be leveraged into therapeutic strategies [165]. It may be possible to repurpose available cancer drugs with reasonable safety profiles as targeted treatments for ASD. For example, the evaluation of a rapamycin analog in patients with tuberous sclerosis included outcome measures for ASD features, along with seizures, sleep disturbances, and academic skills [161]. Therefore, ongoing research into these areas is crucial for developing a deeper understanding and more effective interventions for individuals affected by ASD. Regardless, the enrichment of ASD genes implicated in Wnt signaling points to a Wnt-related molecular signature of some forms of ASD that could inform unified diagnostic and therapeutic strategies.

Since Wnt pathways are altered in many cell pathways and regulation should be tightly controlled, it may be a potential therapeutic target for many disease states. However, it is also a challenge because Wnt is so critical for multiple processes, and appropriate levels, target cells, and timing must be considered. Although both decreased and increased Wnt signaling appear to share overlapping symptoms and clinical presentations, the underlying molecular mechanisms and potential treatments will likely differ due to the distinct imbalances in Wnt activity. As a result of this complex dynamic, treatment strategies will need to be tailored specifically for different conditions involving Wnt signaling to address the specific underlying mechanisms. Some Wnt-targeting compounds are currently under evaluation in clinical trials for many neurological disorders, including PD, such as SB216763, a GSK3 inhibitor [166]. There are currently few methods for halting or slowing the progression of PD, AD, and amyotrophic lateral sclerosis (ALS) [129]. Table 2 highlights some promising compounds.

Table 2:
Selected list of Wnt inhibitors.
CompoundTargetInhibitor/activator of the targetReference
LGK974 Porcupine Inhibitor [167
XAV939 Tankyrase 1/Axin Activates axin [168
Pyrvinium CK1 Stabilizes CK1 [169
NSC668036 Dsh Inhibitor [170
WAY-316606 SFRP Inhibitor [171
Sulindac Dsh Inhibitor [172
Curcumin β-catenin Inhibitor [173
CompoundTargetInhibitor/activator of the targetReference
LGK974 Porcupine Inhibitor [167
XAV939 Tankyrase 1/Axin Activates axin [168
Pyrvinium CK1 Stabilizes CK1 [169
NSC668036 Dsh Inhibitor [170
WAY-316606 SFRP Inhibitor [171
Sulindac Dsh Inhibitor [172
Curcumin β-catenin Inhibitor [173

The reason why Wnt1 specifically is targeted in therapeutics is because Wnt1 is the mediator for the communication between glial cells and Wnt1 agonists. It is responsible for providing a critical neuroprotective mechanism against oxidative stress, growth factor deprivation, and inflammation. Additionally, microglia and astrocytes are both responsible for the primary secretion of Wnt1 and Wnt1 agonists [174]. This is why in neurodevelopmental and/or neurodegenerative diseases where there is excess Wnt production, inhibitors of the Wnt pathway may have strong potential to delay and/or lessen the symptomologies of numerous neurological diseases. Many of these drugs need to be tested in respective neurodegenerative preclinical models and assessed for long-term safety and efficacy. For example, the Wnt antagonist LGK974 is currently in Phase I trials to treat solid malignancies and has also shown promising results in both the cancer field [167] and has been recently used in cultured neurons of ALS mouse models [175]. This drug is a small-molecule inhibitor of porcupine, which is a membrane-bound O-acyltransferase responsible for the secretion and palmitoylation of all Wnt ligands [176]. LGK974 is a potent down regulator of the Wnt/β-catenin signaling pathway, and further research may lead to possible therapeutics in the future for other neurological diseases that show aberrant Wnt signaling. Another Wnt inhibitor being used in the cancer field that has the potential to be used in neurological disease is the Wnt/β-catenin signaling inhibitor XAV939. This drug inhibits β-catenin translocation to the nucleus without affecting their proliferation and viability in vitro, as shown in LNCaP cells and CD4+ BRPCa lymphocytes [177]. XAV939 has also been shown to ameliorate climbing defects in the NEDAMSS disease model in Drosophila when the fly ortholog of IRF2BPL is reduced in neurons [52].

Unfortunately, these drugs are not approved and are still in clinical trials. Hence, another avenue to consider would be to use drugs and/or supplements that are readily available over-the-counter that can inhibit Wnt signaling to some degree. An example of this would be curcumin, a polyphenol that has been shown to inhibit cell growth via the Wnt/β-catenin pathway in non-small-cell lung cancer cells, resulting in decreased β-catenin [173], and is a candidate for treating neurodegenerative diseases such as PD [178]. Another example of a drug that is both readily available and has the potential to be used as a therapeutic in neurological disease is sulindac, which is primarily used as a nonsteroidal anti-inflammatory drug to treat mild-to-moderate pain. Sulindac has been shown to bind to the PDZ domain of Dvl and thereby suppress Wnt3A-induced β-catenin signaling at the level of Dvl [172]. Numerous studies demonstrate the efficacy of sulindac in models of neurodegenerative disease [179-181]. Importantly, sulindac can cross the blood–brain barrier, potentially allowing it to interact with therapeutic targets in the CNS [182].

As discussed, Wnt signaling pathways have context-dependent regulation and are involved in numerous cellular processes. It may be an ideal druggable pathway as understanding increases, since the regulation of Wnt must be tightly controlled for homeostasis. However, disrupting such a complex pathway poses a risk of harmful side effects warranting caution.

Although the gain of Wnt is well-established in cancers in and outside the CNS, this review highlighted the genetic and molecular evidence for increased Wnt signaling in neurodevelopmental and neurodegenerative disorders. The role of up-regulated Wnt signaling varies significantly across neurodevelopmental, degenerative, and oncological disorders, largely depending on the timing and cell-specific context. Wnt signaling is highly context-dependent as its effects are influenced by the temporal and spatial expression patterns in specific cell types in the brain, such as neurons or glia. Although not yet fully understood, this variability emphasizes the complexity of Wnt signaling pathways and highlights the need for further research to unravel its diverse roles and implications in these conditions. Relatedly, excess Wnt in development can contribute to NDDs such as ASD and other neuropsychiatric disorders such as schizophrenia. Yet Wnt in the CNS is not only important for development but also for maintaining neuronal health and function over time. This was evidenced by deregulated Wnt in various neurodegenerative disorders, including AD, PD, HD, and SCA1. A burgeoning question is how enhanced Wnt signaling could lead to neurodegeneration or cancer, as these broad disease states have traditionally been thought of as polar in pathogenesis. Cell context is likely critical, along with the effects of deregulated Wnt in terminally differentiated cells like neurons compared with glia. Nevertheless, understanding the interplay between Wnt pathways and these conditions may lead to possible therapeutic interventions for multiple disorders.

It is important to note that Wnt signaling is a highly context-dependent pathway that plays critical roles in neural development, degeneration, and disease. Its interactions with other pathways, such as Notch [183] and Sonic Hedgehog [184], add layers of complexity that underscore its broad influence on neural processes. These interactions highlight the risk of targeting Wnt signaling therapeutically, as interfering with this cross-talk could result in unpredictable outcomes. Additionally, the context-dependent nature of Wnt signaling further complicates therapeutic targeting. Multiple variables such as ligand concentration, receptor expression, and environmental factors such as stress, inflammation, and injury can significantly influence Wnt activity [185]. These factors emphasize the need for patient-specific approaches in developing Wnt-targeted therapies, as a one-size-fits-all strategy is unlikely to be effective. In the mature brain, the role of Wnt signaling is especially challenging to study compared with its role in development. Unlike the relatively well-defined processes of development, Wnt signaling in the adult brain is more subtle and complex, with cell-specific interactions between Wnt ligands, receptors, and downstream signaling pathways that are harder to pinpoint [186]. This complexity necessitates advanced tools and research, such as conditional animal models, including adult-specific knockout systems and inducible mouse models. These tools are critical for dissecting the precise roles of Wnt signaling in the adult brain and for understanding how its dysregulation contributes to neurodegeneration and other disorders.

All authors have no competing interests to report.

D.M.P.: Writing—Original draft, Writing—review and editing. D.J.R.: Writing—Original draft, Writing—review and editing. H.K.: Writing—Original draft, Writing—review and editing. P.C.M.: Conceptualization, Writing— review and editing.

AD

Alzheimer’s disease

ADHD

attention deficit hyperactivity disorder

ALS

amyotrophic lateral sclerosis

ASD

autism spectrum disorders

amyloid beta

BPAD

bipolar affective disorder

CCND1

cyclin D1

CNS

central nervous system

CamKII

calcium/calmodulin-dependent kinase II

DISC1

disrupted in schizophrenia 1

DKK

dickkopf

DNVs

de novo coding variants

DVL

dishevelled

FAT1

FAT atypical cadherin 1

FTD

frontotemporal dementia

Fzd

frizzled

GBM

glioblastoma

GRN

granulin

HD

Huntington's disease

HMG

high mobility group

IRF2BPL

interferon regulatory factor 2 binding protein-like

LRRK2

leucine-rich repeat kinase 2

MB

medulloblastoma

NDDs

neurodevelopmental disorders

NEDAMSS

neurodevelopmental disorder with regression, abnormal movements, loss of speech, and seizures

NFAT

nuclear factor of activated T cells

NSCs

neural stem cells

PCP

planar cell polarity

PD

Parkinson’s disease

PKC

protein kinase C

SCA1

spinocerebellar ataxia type 1

SESTD1

SEC14 and spectrin domain-containing 1

SFRPs

secreted Fzd-related proteins

SNP

single nucleotide polymorphism

TSC2

tuberous sclerosis complex 2

WIF, WLS

wntless,Wnt inhibitory factor

Wnt

wingless/integrated

iPSCs

induced pluripotent stem cells

1
Liu
,
J.
,
Xiao
,
Q.
,
Xiao
,
J.
,
Niu
,
C.
,
Li
,
Y.
,
Zhang
,
X.
et al.
(
2022
)
Wnt/β-catenin signalling: function, biological mechanisms, and therapeutic opportunities
.
Signal Transduct. Target. Ther.
7
, 3 https://doi.org/10.1038/s41392-021-00762-6
2
Sharma
,
R.P.
and
Chopra
,
V.L
. (
1976
)
Effect of the wingless (wg1) mutation on wing and haltere development in Drosophila melanogaster
.
Dev. Biol. (NY)
48
,
461
465
https://doi.org/10.1016/0012-1606(76)90108-1
3
Nüsslein-Volhard
,
C.
and
Wieschaus
,
E
. (
1980
)
Mutations affecting segment number and polarity in Drosophila
.
Nature
287
,
795
801
https://doi.org/10.1038/287795a0
4
Wodarz
,
A.
and
Nusse
,
R
. (
1998
)
Mechanisms of Wnt signaling in development
.
Annu. Rev. Cell Dev. Biol.
14
,
59
88
https://doi.org/10.1146/annurev.cellbio.14.1.59
5
Miller
,
J.R
. (
2002
)
The Wnts
.
Genome Biol.
3
, reviews3001.1 https://doi.org/10.1186/gb-2001-3-1-reviews3001
6
Semenov
,
M.V.
,
Habas
,
R.
,
Macdonald
,
B.T.
and
He
,
X
. (
2007
)
SnapShot: noncanonical Wnt signaling pathways
.
Cell
131
, 1378 https://doi.org/10.1016/j.cell.2007.12.011
7
Zhan
,
T.
,
Rindtorff
,
N.
and
Boutros
,
M
. (
2017
)
Wnt signaling in cancer
.
Oncogene
36
,
1461
1473
https://doi.org/10.1038/onc.2016.304
8
Huang
,
H.
and
He
,
X
. (
2008
)
Wnt/beta-catenin signaling: new (and old) players and new insights
.
Curr. Opin. Cell Biol.
20
,
119
125
https://doi.org/10.1016/j.ceb.2008.01.009
9
Freese
,
J.L.
,
Pino
,
D.
and
Pleasure
,
S.J
. (
2010
)
Wnt signaling in development and disease
.
Neurobiol. Dis.
38
,
148
153
https://doi.org/10.1016/j.nbd.2009.09.003
10
Tree
,
D.R.P.
,
Ma
,
D.
and
Axelrod
,
J.D
. (
2002
)
A three-tiered mechanism for regulation of planar cell polarity
.
Semin. Cell Dev. Biol.
13
,
217
224
https://doi.org/10.1016/s1084-9521(02)00042-3
11
Strutt
,
H.
and
Strutt
,
D
. (
2005
)
Long-range coordination of planar polarity in Drosophila
.
Bioessays
27
,
1218
1227
https://doi.org/10.1002/bies.20318
12
Axelrod
,
J.D
. (
2001
)
Unipolar membrane association of dishevelled mediates frizzled planar cell polarity signaling
.
Genes Dev.
15
,
1182
1187
https://doi.org/10.1101/gad.890501
13
Song
,
Y.
,
Jian
,
S.
,
Teng
,
J.
,
Zheng
,
P.
and
Zhang
,
Z
. (
2025
)
Structural basis of human VANGL-PRICKLE interaction
.
Nat. Commun.
16
, 132 https://doi.org/10.1038/s41467-024-55396-3
14
Schlessinger
,
K.
,
Hall
,
A.
and
Tolwinski
,
N
. (
2009
)
Wnt signaling pathways meet Rho GTPases
.
Genes Dev.
23
,
265
277
https://doi.org/10.1101/gad.1760809
15
Corkins
,
M.E.
,
Krneta-Stankic
,
V.
,
Kloc
,
M.
,
McCrea
,
P.D.
,
Gladden
,
A.B.
and
Miller
,
R.K
. (
2019
)
Divergent roles of the Wnt/PCP Formin Daam1 in renal ciliogenesis
.
Plos One
14
, e0221698 https://doi.org/10.1371/journal.pone.0221698
16
Zallen
,
J.A
. (
2007
)
Planar polarity and tissue morphogenesis
.
Cell
129
,
1051
1063
https://doi.org/10.1016/j.cell.2007.05.050
17
Shi
,
D.L
. (
2022
)
Wnt/planar cell polarity signaling controls morphogenetic movements of gastrulation and neural tube closure
.
Cell. Mol. Life Sci.
79
, 586 https://doi.org/10.1007/s00018-022-04620-8
18
Slusarski
,
D.C.
,
Yang-Snyder
,
J.
,
Busa
,
W.B.
and
Moon
,
R.T
. (
1997
)
Modulation of embryonic intracellular Ca2+signaling byWnt-5A
.
Dev. Biol. (NY)
182
,
114
120
https://doi.org/10.1006/dbio.1996.8463
19
Westfall
,
T.A.
,
Brimeyer
,
R.
,
Twedt
,
J.
,
Gladon
,
J.
,
Olberding
,
A.
,
Furutani-Seiki
,
M.
et al.
(
2003
)
Wnt-5/pipetail functions in vertebrate axis formation as a negative regulator of Wnt/beta-catenin activity
.
J. Cell Biol.
162
,
889
898
https://doi.org/10.1083/jcb.200303107
20
Kohn
,
A.D.
and
Moon
,
R.T
. (
2005
)
Wnt and calcium signaling: beta-catenin-independent pathways
.
Cell Calcium
38
,
439
446
https://doi.org/10.1016/j.ceca.2005.06.022
21
Qin
,
K.
,
Yu
,
M.
,
Fan
,
J.
,
Wang
,
H.
,
Zhao
,
P.
,
Zhao
,
G.
et al.
(
2024
)
Canonical and noncanonical Wnt signaling: multilayered mediators, signaling mechanisms and major signaling crosstalk
.
Genes Dis.
11
,
103
134
https://doi.org/10.1016/j.gendis.2023.01.030
22
Ramakrishna
,
K.
,
Nalla
,
L.V.
,
Naresh
,
D.
,
Venkateswarlu
,
K.
,
Viswanadh
,
M.K.
,
Nalluri
,
B.N.
et al.
(
2023
)
WNT-β catenin signaling as a potential therapeutic target for neurodegenerative diseases: current status and future perspective
.
Diseases
11
, 89 https://doi.org/10.3390/diseases11030089
23
Yu
,
M.
,
Qin
,
K.
,
Fan
,
J.
,
Zhao
,
G.
,
Zhao
,
P.
,
Zeng
,
W.
et al.
(
2024
)
The evolving roles of Wnt signaling in stem cell proliferation and differentiation, the development of human diseases, and therapeutic opportunities
.
Genes Dis.
11
, 101026 https://doi.org/10.1016/j.gendis.2023.04.042
24
Mulligan
,
K.A.
and
Cheyette
,
B.N.R
. (
2017
)
Neurodevelopmental perspectives on Wnt signaling in psychiatry
.
Mol. Neuropsychiatry
2
,
219
246
https://doi.org/10.1159/000453266
25
Rosso
,
S.B.
and
Inestrosa
,
N.C
. (
2013
)
WNT signaling in neuronal maturation and synaptogenesis
.
Front. Cell. Neurosci.
7
, 103 https://doi.org/10.3389/fncel.2013.00103
26
Patapoutian
,
A.
and
Reichardt
,
L.F
. (
2000
)
Roles of Wnt proteins in neural development and maintenance
.
Curr. Opin. Neurobiol.
10
,
392
399
https://doi.org/10.1016/s0959-4388(00)00100-8
27
Arredondo
,
S.B.
,
Valenzuela-Bezanilla
,
D.
,
Mardones
,
M.D.
and
Varela-Nallar
,
L
. (
2020
)
Role of Wnt signaling in adult hippocampal neurogenesis in health and disease
.
Front. Cell Dev. Biol.
8
, 860 https://doi.org/10.3389/fcell.2020.00860
28
Kostes
,
W.W.
and
Brafman
,
D.A
. (
2023
)
The multifaceted role of WNT signaling in alzheimer’s disease onset and age-related progression
.
Cells
12
, 1204 https://doi.org/10.3390/cells12081204
29
Wexler
,
E.M.
,
Rosen
,
E.
,
Lu
,
D.
,
Osborn
,
G.E.
,
Martin
,
E.
,
Raybould
,
H.
et al.
(
2011
)
Genome-wide analysis of a Wnt1-regulated transcriptional network implicates neurodegenerative pathways
.
Sci. Signal.
4
,
ra65
ra65
https://doi.org/10.1126/scisignal.2002282
30
Kwan
,
V.
,
Unda
,
B.K.
and
Singh
,
K.K
. (
2016
)
Wnt signaling networks in autism spectrum disorder and intellectual disability
.
J. Neurodev. Disord.
8
, 45 https://doi.org/10.1186/s11689-016-9176-3
31
Bocchi
,
R.
,
Egervari
,
K.
,
Carol-Perdiguer
,
L.
,
Viale
,
B.
,
Quairiaux
,
C.
,
De Roo
,
M.
et al.
(
2017
)
Perturbed Wnt signaling leads to neuronal migration delay, altered interhemispheric connections and impaired social behavior
.
Nat. Commun.
8
, 1158 https://doi.org/10.1038/s41467-017-01046-w
32
Zhuang
,
W.
,
Ye
,
T.
,
Wang
,
W.
,
Song
,
W.
and
Tan
,
T
. (
2023
)
CTNNB1 in neurodevelopmental disorders
.
Front. Psychiatry
14
, 1143328 https://doi.org/10.3389/fpsyt.2023.1143328
33
Sudnawa
,
K.K.
,
Garber
,
A.
,
Cohen
,
R.
,
Calamia
,
S.
,
Kanner
,
C.H.
,
Montes
,
J.
et al.
(
2024
)
Clinical phenotypic spectrum of CTNNB1 neurodevelopmental disorder
.
Clin. Genet.
105
,
523
532
https://doi.org/10.1111/cge.14487
34
Katsu
,
T.
,
Ujike
,
H.
,
Nakano
,
T.
,
Tanaka
,
Y.
,
Nomura
,
A.
,
Nakata
,
K.
et al.
(
2003
)
The human frizzled-3 (FZD3) gene on chromosome 8p21, a receptor gene for Wnt ligands, is associated with the susceptibility to schizophrenia
.
Neurosci. Lett.
353
,
53
56
https://doi.org/10.1016/j.neulet.2003.09.017
35
McMahon
,
A.P.
and
Bradley
,
A
. (
1990
)
The Wnt-1 (int-1) proto-oncogene is required for development of a large region of the mouse brain
.
Cell
62
,
1073
1085
https://doi.org/10.1016/0092-8674(90)90385-r
36
Chai
,
G.
,
Szenker-Ravi
,
E.
,
Chung
,
C.
,
Li
,
Z.
,
Wang
,
L.
,
Khatoo
,
M.
et al.
(
2021
)
A human pleiotropic multiorgan condition caused by deficient Wnt secretion
.
N. Engl. J. Med.
385
,
1292
1301
https://doi.org/10.1056/NEJMoa2033911
37
Mukhopadhyay
,
M.
,
Shtrom
,
S.
,
Rodriguez-Esteban
,
C.
,
Chen
,
L.
,
Tsukui
,
T.
,
Gomer
,
L.
et al.
(
2001
)
Dickkopf1 is required for embryonic head induction and limb morphogenesis in the mouse
.
Dev. Cell
1
,
423
434
https://doi.org/10.1016/s1534-5807(01)00041-7
38
Lee
,
S.
,
Chen
,
D.Y.
,
Zaki
,
M.S.
,
Maroofian
,
R.
,
Houlden
,
H.
,
Di Donato
,
N.
et al.
(
2019
)
Bi-allelic loss of human APC2, encoding adenomatous polyposis coli protein 2, leads to lissencephaly, subcortical heterotopia, and global developmental delay
.
Am. J. Hum. Genet.
105
,
844
853
https://doi.org/10.1016/j.ajhg.2019.08.013
39
Lei
,
Y.
,
Fathe
,
K.
,
McCartney
,
D.
,
Zhu
,
H.
,
Yang
,
W.
,
Ross
,
M.E.
et al.
(
2015
)
Rare LRP6 variants identified in spina bifida patients
.
Hum. Mutat.
36
,
342
349
https://doi.org/10.1002/humu.22750
40
Greco
,
T.L.
,
Takada
,
S.
,
Newhouse
,
M.M.
,
McMahon
,
J.A.
,
McMahon
,
A.P.
and
Camper
,
S.A
. (
1996
)
Analysis of the vestigial tail mutation demonstrates that Wnt-3a gene dosage regulates mouse axial development
.
Genes Dev.
10
,
313
324
https://doi.org/10.1101/gad.10.3.313
41
Van Amerongen
,
R.
and
Berns
,
A
. (
2006
)
Knockout mouse models to study Wnt signal transduction
.
Trends Genet.
22
,
678
689
https://doi.org/10.1016/j.tig.2006.10.001
42
Haghshenas
,
S.
,
Bhai
,
P.
,
Aref-Eshghi
,
E.
and
Sadikovic
,
B
. (
2020
)
Diagnostic utility of genome-wide DNA methylation analysis in mendelian neurodevelopmental disorders
.
Int. J. Mol. Sci.
21
, 9303 https://doi.org/10.3390/ijms21239303
43
Parenti
,
I.
,
Rabaneda
,
L.G.
,
Schoen
,
H.
and
Novarino
,
G
. (
2020
)
Neurodevelopmental disorders: from genetics to functional pathways
.
Trends Neurosci.
43
,
608
621
https://doi.org/10.1016/j.tins.2020.05.004
44
Kim
,
Y.S.
and
State
,
M.W
. (
2014
)
Recent challenges to the psychiatric diagnostic nosology: a focus on the genetics and genomics of neurodevelopmental disorders
.
Int. J. Epidemiol.
43
,
465
475
https://doi.org/10.1093/ije/dyu037
45
Splinter
,
K.
,
Adams
,
D.R.
,
Bacino
,
C.A.
,
Bellen
,
H.J.
,
Bernstein
,
J.A.
,
Cheatle-Jarvela
,
A.M.
et al.
(
2018
)
Effect of genetic diagnosis on patients with previously undiagnosed disease
.
N. Engl. J. Med.
379
,
2131
2139
https://doi.org/10.1056/NEJMoa1714458
46
Iossifov
,
I.
,
O’Roak
,
B.J.
,
Sanders
,
S.J.
,
Ronemus
,
M.
,
Krumm
,
N.
,
Levy
,
D.
et al.
(
2014
)
The contribution of de novo coding mutations to autism spectrum disorder
.
Nature
515
,
216
221
https://doi.org/10.1038/nature13908
47
Study
,
D.D.D
. (
2017
)
Prevalence and architecture of de novo mutations in developmental disorders
.
Nature
542
,
433
438
https://doi.org/10.1038/nature21062
48
Accogli
,
A.
,
Calabretta
,
S.
,
St-Onge
,
J.
,
Boudrahem-Addour
,
N.
,
Dionne-Laporte
,
A.
,
Joset
,
P.
et al.
(
2019
)
De novo pathogenic variants in N-cadherin cause a syndromic neurodevelopmental disorder with corpus collosum, axon, cardiac, ocular, and genital defects
.
Am. J. Hum. Genet.
105
,
854
868
https://doi.org/10.1016/j.ajhg.2019.09.005
49
Bernier
,
R.
,
Golzio
,
C.
,
Xiong
,
B.
,
Stessman
,
H.A.
,
Coe
,
B.P.
,
Penn
,
O.
et al.
(
2014
)
Disruptive CHD8 mutations define a subtype of autism early in development
.
Cell
158
,
263
276
https://doi.org/10.1016/j.cell.2014.06.017
50
Angers
,
S.
,
Thorpe
,
C.J.
,
Biechele
,
T.L.
,
Goldenberg
,
S.J.
,
Zheng
,
N.
,
MacCoss
,
M.J.
et al.
(
2006
)
The KLHL12-Cullin-3 ubiquitin ligase negatively regulates the Wnt-beta-catenin pathway by targeting dishevelled for degradation
.
Nat. Cell Biol.
8
,
348
357
https://doi.org/10.1038/ncb1381
51
Zhang
,
Z.
,
Wlodarczyk
,
B.J.
,
Niederreither
,
K.
,
Venugopalan
,
S.
,
Florez
,
S.
,
Finnell
,
R.H.
et al.
(
2011
)
Fuz regulates craniofacial development through tissue specific responses to signaling factors
.
Plos One
6
, e24608 https://doi.org/10.1371/journal.pone.0024608
52
Marcogliese
,
P.C.
,
Dutta
,
D.
,
Ray
,
S.S.
,
Dang
,
N.D.P.
,
Zuo
,
Z.
,
Wang
,
Y.
et al.
(
2022
)
Loss of IRF2BPL impairs neuronal maintenance through excess Wnt signaling
.
Sci. Adv.
8
, eabl5613 https://doi.org/10.1126/sciadv.abl5613
53
Chan
,
D.W.
,
Chan
,
C.Y.
,
Yam
,
J.W.P.
,
Ching
,
Y.P.
and
Ng
,
I.O.L
. (
2006
)
Prickle-1 negatively regulates Wnt/beta-catenin pathway by promoting Dishevelled ubiquitination/degradation in liver cancer
.
Gastroenterology
131
,
1218
1227
https://doi.org/10.1053/j.gastro.2006.07.020
54
Rawal
,
N.
,
Corti
,
O.
,
Sacchetti
,
P.
,
Ardilla-Osorio
,
H.
,
Sehat
,
B.
,
Brice
,
A.
et al.
(
2009
)
Parkin protects dopaminergic neurons from excessive Wnt/beta-catenin signaling
.
Biochem. Biophys. Res. Commun.
388
,
473
478
https://doi.org/10.1016/j.bbrc.2009.07.014
55
Maurizi
,
E.
,
Schiroli
,
D.
,
Zini
,
R.
,
Limongelli
,
A.
,
Mistò
,
R.
,
Macaluso
,
C.
et al.
(
2020
)
A fine-tuned β-catenin regulation during proliferation of corneal endothelial cells revealed using proteomics analysis
.
Sci. Rep.
10
, 13841 https://doi.org/10.1038/s41598-020-70800-w
56
Scholz
,
B.
,
Korn
,
C.
,
Wojtarowicz
,
J.
,
Mogler
,
C.
,
Augustin
,
I.
,
Boutros
,
M.
et al.
(
2016
)
Endothelial RSPO3 controls vascular stability and pruning through non-canonical WNT/Ca(2+)/NFAT signaling
.
Dev. Cell
36
,
79
93
https://doi.org/10.1016/j.devcel.2015.12.015
57
Terabayashi
,
T.
,
Funato
,
Y.
,
Fukuda
,
M.
and
Miki
,
H
. (
2009
)
A coated vesicle-associated kinase of 104 kDa (CVAK104) induces lysosomal degradation of frizzled 5 (Fzd5)
.
J. Biol. Chem.
284
,
26716
26724
https://doi.org/10.1074/jbc.M109.039313
58
Tong
,
X.
,
Li
,
L.
,
Li
,
X.
,
Heng
,
L.
,
Zhong
,
L.
,
Su
,
X.
et al.
(
2014
)
SOX10, a novel HMG-box-containing tumor suppressor, inhibits growth and metastasis of digestive cancers by suppressing the Wnt/β-catenin pathway
.
Oncotarget
5
,
10571
10583
https://doi.org/10.18632/oncotarget.2512
59
Kelberman
,
D.
,
de Castro
,
S.C.P.
,
Huang
,
S.
,
Crolla
,
J.A.
,
Palmer
,
R.
,
Gregory
,
J.W.
et al.
(
2008
)
SOX2 plays a critical role in the pituitary, forebrain, and eye during human embryonic development
.
J. Clin. Endocrinol. Metab.
93
,
1865
1873
https://doi.org/10.1210/jc.2007-2337
60
Gaudet
,
P.
,
Livstone
,
M.S.
,
Lewis
,
S.E.
and
Thomas
,
P.D
. (
2011
)
Phylogenetic-based propagation of functional annotations within the gene ontology consortium
.
Brief. Bioinformatics
12
,
449
462
https://doi.org/10.1093/bib/bbr042
61
Mak
,
B.C.
,
Kenerson
,
H.L.
,
Aicher
,
L.D.
,
Barnes
,
E.A.
and
Yeung
,
R.S
. (
2005
)
Aberrant beta-catenin signaling in tuberous sclerosis
.
Am. J. Pathol.
167
,
107
116
https://doi.org/10.1016/s0002-9440(10)62958-6
62
Mak
,
B.C.
,
Takemaru
,
K.I.
,
Kenerson
,
H.L.
,
Moon
,
R.T.
and
Yeung
,
R.S
. (
2003
)
The tuberin-hamartin complex negatively regulates beta-catenin signaling activity
.
J. Biol. Chem.
278
,
5947
5951
https://doi.org/10.1074/jbc.C200473200
63
Bouteille
,
N.
,
Driouch
,
K.
,
Hage
,
P.E.
,
Sin
,
S.
,
Formstecher
,
E.
,
Camonis
,
J.
et al.
(
2009
)
Inhibition of the Wnt/beta-catenin pathway by the WWOX tumor suppressor protein
.
Oncogene
28
,
2569
2580
https://doi.org/10.1038/onc.2009.120
64
Shintani
,
T.
,
Takeuchi
,
Y.
,
Fujikawa
,
A.
and
Noda
,
M
. (
2012
)
Directional neuronal migration is impaired in mice lacking adenomatous polyposis coli 2
.
J. Neurosci.
32
,
6468
6484
https://doi.org/10.1523/JNEUROSCI.0590-12.2012
65
Mizuguchi
,
M.
,
Ohsawa
,
M.
,
Kashii
,
H.
and
Sato
,
A
. (
2021
)
Brain symptoms of tuberous sclerosis complex: pathogenesis and treatment
.
Int. J. Mol. Sci.
22
, 6677 https://doi.org/10.3390/ijms22136677
66
Marcogliese
,
P.C.
,
Shashi
,
V.
,
Spillmann
,
R.C.
,
Stong
,
N.
,
Rosenfeld
,
J.A.
,
Koenig
,
M.K.
et al.
(
2018
)
IRF2BPL is associated with neurological phenotypes
.
Am. J. Hum. Genet.
103
,
245
260
https://doi.org/10.1016/j.ajhg.2018.07.006
67
Yi
,
J.J.
,
Paranjape
,
S.R.
,
Walker
,
M.P.
,
Choudhury
,
R.
,
Wolter
,
J.M.
,
Fragola
,
G
et al.
(
2017
)
The autism-linked UBE3A T485A mutant E3 ubiquitin ligase activates the Wnt/β-catenin pathway by inhibiting the proteasome
.
J. Biol. Chem.
292
,
12503
12515
https://doi.org/10.1074/jbc.M117.788448
68
Thompson
,
B.A.
,
Tremblay
,
V.
,
Lin
,
G.
and
Bochar
,
D.A
. (
2008
)
CHD8 is an ATP-dependent chromatin remodeling factor that regulates beta-catenin target genes
.
Mol. Cell. Biol.
28
,
3894
3904
https://doi.org/10.1128/MCB.00322-08
69
Nishiyama
,
M.
,
Skoultchi
,
A.I.
and
Nakayama
,
K.I
. (
2012
)
Histone H1 recruitment by CHD8 is essential for suppression of the Wnt-β-catenin signaling pathway
.
Mol. Cell. Biol.
32
,
501
512
https://doi.org/10.1128/MCB.06409-11
70
Haÿ
,
E.
,
Laplantine
,
E.
,
Geoffroy
,
V.
,
Frain
,
M.
,
Kohler
,
T.
,
Müller
,
R.
et al.
(
2009
)
N-cadherin interacts with axin and LRP5 to negatively regulate Wnt/beta-catenin signaling, osteoblast function, and bone formation
.
Mol. Cell. Biol.
29
,
953
964
https://doi.org/10.1128/MCB.00349-08
71
Hodges
,
H.
,
Fealko
,
C.
and
Soares
,
N
. (
2020
)
Autism spectrum disorder: definition, epidemiology, causes, and clinical evaluation
.
Transl. Pediatr.
9
,
S55
S65
https://doi.org/10.21037/tp.2019.09.09
72
Gaugler
,
T.
,
Klei
,
L.
,
Sanders
,
S.J.
,
Bodea
,
C.A.
,
Goldberg
,
A.P.
,
Lee
,
A.B.
et al.
(
2014
)
Most genetic risk for autism resides with common variation
.
Nat. Genet.
46
,
881
885
https://doi.org/10.1038/ng.3039
73
Hallmayer
,
J.
,
Cleveland
,
S.
,
Torres
,
A.
,
Phillips
,
J.
,
Cohen
,
B.
,
Torigoe
,
T.
et al.
(
2011
)
Genetic heritability and shared environmental factors among twin pairs with autism
.
Arch. Gen. Psychiatry
68
,
1095
1102
https://doi.org/10.1001/archgenpsychiatry.2011.76
74
Klei
,
L.
,
Sanders
,
S.J.
,
Murtha
,
M.T.
,
Hus
,
V.
,
Lowe
,
J.K.
,
Willsey
,
A.J.
et al.
(
2012
)
Common genetic variants, acting additively, are a major source of risk for autism
.
Mol. Autism
3
, 9 https://doi.org/10.1186/2040-2392-3-9
75
Zeidán-Chuliá
,
F.
,
de Oliveira
,
B.H.N.
,
Salmina
,
A.B.
,
Casanova
,
M.F.
,
Gelain
,
D.P.
,
Noda
,
M.
et al.
(
2014
)
Altered expression of alzheimer’s disease-related genes in the cerebellum of autistic patients: a model for disrupted brain connectome and therapy
.
Cell Death Dis.
5
, e1250 https://doi.org/10.1038/cddis.2014.227
76
Caracci
,
M.O.
,
Avila
,
M.E.
,
Espinoza-Cavieres
,
F.A.
,
López
,
H.R.
,
Ugarte
,
G.D.
and
De Ferrari
,
G.V
. (
2021
)
Wnt/β-catenin-dependent transcription in autism spectrum disorders
.
Front. Mol. Neurosci.
14
, 764756 https://doi.org/10.3389/fnmol.2021.764756
77
Satterstrom
,
F.K.
,
Kosmicki
,
J.A.
,
Wang
,
J.
,
Breen
,
M.S.
,
De Rubeis
,
S.
,
An
,
J.Y.
et al.
(
2020
)
Large-scale exome sequencing study implicates both developmental and functional changes in the neurobiology of autism
.
Cell
180
,
568
584
https://doi.org/10.1016/j.cell.2019.12.036
78
Mastrototaro
,
G.
,
Zaghi
,
M.
,
Massimino
,
L.
,
Moneta
,
M.
,
Mohammadi
,
N.
,
Banfi
,
F.
et al.
(
2021
)
TBL1XR1 ensures balanced neural development through NCOR complex-mediated regulation of the MAPK pathway
.
Front. Cell Dev. Biol.
9
, 641410 https://doi.org/10.3389/fcell.2021.641410
79
Rosenbluh
,
J.
,
Wang
,
X.
and
Hahn
,
W.C
. (
2014
)
Genomic insights into WNT/β-catenin signaling
.
Trends Pharmacol. Sci.
35
,
103
109
https://doi.org/10.1016/j.tips.2013.11.007
80
Heilmann
,
R.
,
Pfalzer
,
A.
,
Bichell
,
T.J.
,
Terala
,
A.
,
Campbell
,
A.
,
Taatjes
,
D.
et al.
(
2024
)
The MED13L foundation strategic research plan: a roadmap to the future
.
Ther. Adv. Rare Dis.
5
, 26330040241290252 https://doi.org/10.1177/26330040241290252
81
Hayes
,
M.
,
Naito
,
M.
,
Daulat
,
A.
,
Angers
,
S.
and
Ciruna
,
B
. (
2013
)
Ptk7 promotes non-canonical Wnt/PCP-mediated morphogenesis and inhibits Wnt/β-catenin-dependent cell fate decisions during vertebrate development
.
Development (Rome)
140
,
1807
1818
https://doi.org/10.1242/dev.090183
82
Li
,
N.
,
Zhou
,
P.
,
Tang
,
H.
,
He
,
L.
,
Fang
,
X.
,
Zhao
,
J.
et al.
(
2022
)
In-depth analysis reveals complex molecular aetiology in a cohort of idiopathic cerebral palsy
.
Brain (Bacau)
145
,
119
141
https://doi.org/10.1093/brain/awab209
83
Bray
,
N.J.
and
O’Donovan
,
M.C
. (
2019
)
The genetics of neuropsychiatric disorders
.
Brain Neurosci. Adv.
2
, 2398212818799271 https://doi.org/10.1177/2398212818799271
84
Okerlund
,
N.D.
and
Cheyette
,
B.N.R
. (
2011
)
Synaptic Wnt signaling-a contributor to major psychiatric disorders?
J. Neurodev. Disord.
3
,
162
174
https://doi.org/10.1007/s11689-011-9083-6
85
Lichtenstein
,
P.
,
Yip
,
B.H.
,
Björk
,
C.
,
Pawitan
,
Y.
,
Cannon
,
T.D.
,
Sullivan
,
P.F.
et al.
(
2009
)
Common genetic determinants of schizophrenia and bipolar disorder in Swedish families: a population-based study
.
Lancet
373
,
234
239
https://doi.org/10.1016/S0140-6736(09)60072-6
86
Ross
,
C.A.
,
Margolis
,
R.L.
,
Reading
,
S.A.J.
,
Pletnikov
,
M.
and
Coyle
,
J.T
. (
2006
)
Neurobiology of schizophrenia
.
Neuron
52
,
139
153
https://doi.org/10.1016/j.neuron.2006.09.015
87
Hill
,
J.J.
,
Hashimoto
,
T.
and
Lewis
,
D.A
. (
2006
)
Molecular mechanisms contributing to dendritic spine alterations in the prefrontal cortex of subjects with schizophrenia
.
Mol. Psychiatry
11
,
557
566
https://doi.org/10.1038/sj.mp.4001792
88
Harrison
,
P.J.
and
Weinberger
,
D.R
. (
2005
)
Schizophrenia genes, gene expression, and neuropathology: on the matter of their convergence
.
Mol. Psychiatry
10
,
40
68
https://doi.org/10.1038/sj.mp.4001558
89
Guilmatre
,
A.
,
Dubourg
,
C.
,
Mosca
,
A.L.
,
Legallic
,
S.
,
Goldenberg
,
A.
,
Drouin-Garraud
,
V.
et al.
(
2009
)
Recurrent rearrangements in synaptic and neurodevelopmental genes and shared biologic pathways in schizophrenia, autism, and mental retardation
.
Arch. Gen. Psychiatry
66
,
947
956
https://doi.org/10.1001/archgenpsychiatry.2009.80
90
Miyaoka
,
T.
,
Seno
,
H.
and
Ishino
,
H
. (
1999
)
Increased expression of Wnt-1 in schizophrenic brains
.
Schizophr. Res.
38
,
1
6
https://doi.org/10.1016/s0920-9964(98)00179-0
91
Stefansson
,
H.
,
Ophoff
,
R.A.
,
Steinberg
,
S.
,
Andreassen
,
O.A.
,
Cichon
,
S.
,
Rujescu
,
D.
et al.
(
2009
)
Common variants conferring risk of schizophrenia
.
Nature.
460
,
744
747
https://doi.org/10.1038/nature08186
92
Brzózka
,
M.M.
,
Radyushkin
,
K.
,
Wichert
,
S.P.
,
Ehrenreich
,
H.
and
Rossner
,
M.J
. (
2010
)
Cognitive and sensorimotor gating impairments in transgenic mice overexpressing the Schizophrenia susceptibility gene Tcf4 in the brain
.
Biol. Psychiatry Cogn. Neurosci. Neuroimaging
68
,
33
40
https://doi.org/10.1016/j.biopsych.2010.03.015
93
Aleksic
,
B.
,
Kushima
,
I.
,
Ito
,
Y.
,
Nakamura
,
Y.
,
Ujike
,
H.
,
Suzuki
,
M.
et al.
(
2010
)
Genetic association study of KREMEN1 and DKK1 and schizophrenia in a Japanese population
.
Schizophr. Res.
118
,
113
117
https://doi.org/10.1016/j.schres.2010.01.014
94
Ftouh
,
S.
,
Akbar
,
M.T.
,
Hirsch
,
S.R.
,
de Belleroche,
,
J.S.D
(
2005
)
Down-regulation of dickkopf 3, a regulator of the Wnt signalling pathway, in elderly schizophrenic subjects
.
J. Neurochem.
94
,
520
530
https://doi.org/10.1111/j.1471-4159.2005.03239.x
95
Proitsi
,
P.
,
Li
,
T.
,
Hamilton
,
G.
,
Di Forti
,
M.
,
Collier
,
D.
,
Killick
,
R.
et al.
(
2008
)
Positional pathway screen of wnt signaling genes in schizophrenia: association with DKK4
.
Biol. Psychiatry Cogn. Neurosci. Neuroimaging
63
,
13
16
https://doi.org/10.1016/j.biopsych.2007.03.014
96
Scassellati
,
C.
,
Bonvicini
,
C.
,
Perez
,
J.
,
Bocchio-Chiavetto
,
L.
,
Tura
,
G.B.
,
Rossi
,
G.
et al.
(
2004
)
Association study of -1727 A/T, -50 C/T and (CAA)n repeat GSK-3beta gene polymorphisms with schizophrenia
.
Neuropsychobiology
50
,
16
20
https://doi.org/10.1159/000077936
97
Souza
,
R.P.
,
Romano-Silva
,
M.A.
,
Lieberman
,
J.A.
,
Meltzer
,
H.Y.
,
Wong
,
A.H.C.
and
Kennedy
,
J.L
. (
2008
)
Association study of GSK3 gene polymorphisms with schizophrenia and clozapine response
.
Psychopharmacology (Berl.)
200
,
177
186
, 177 https://doi.org/10.1007/s00213-008-1193-9
98
Hayashi-Takagi
,
A.
,
Takaki
,
M.
,
Graziane
,
N.
,
Seshadri
,
S.
,
Murdoch
,
H.
,
Dunlop
,
A.J.
et al.
(
2010
)
Disrupted-in-schizophrenia 1 (DISC1) regulates spines of the glutamate synapse via Rac1
.
Nat. Neurosci.
13
,
327
332
https://doi.org/10.1038/nn.2487
99
Mao
,
Y.
,
Ge
,
X.
,
Frank
,
C.L.
,
Madison
,
J.M.
,
Koehler
,
A.N.
,
Doud
,
M.K.
et al.
(
2009
)
Disrupted in schizophrenia 1 regulates neuronal progenitor proliferation via modulation of GSK3beta/beta-catenin signaling
.
Cell
136
,
1017
1031
https://doi.org/10.1016/j.cell.2008.12.044
100
Singh
,
K.K.
,
Ge
,
X.
,
Mao
,
Y.
,
Drane
,
L.
,
Meletis
,
K.
,
Samuels
,
B.A.
et al.
(
2010
)
Dixdc1 is a critical regulator of DISC1 and embryonic cortical development
.
Neuron
67
,
33
48
https://doi.org/10.1016/j.neuron.2010.06.002
101
McGuffin
,
P.
,
Rijsdijk
,
F.
,
Andrew
,
M.
,
Sham
,
P.
,
Katz
,
R.
and
Cardno
,
A
. (
2003
)
The heritability of bipolar affective disorder and the genetic relationship to unipolar depression
.
Arch. Gen. Psychiatry
60
,
497
502
https://doi.org/10.1001/archpsyc.60.5.497
102
Hennah
,
W.
,
Thomson
,
P.
,
McQuillin
,
A.
,
Bass
,
N.
,
Loukola
,
A.
,
Anjorin
,
A.
et al.
(
2009
)
DISC1 association, heterogeneity and interplay in schizophrenia and bipolar disorder
.
Mol. Psychiatry
14
,
865
873
https://doi.org/10.1038/mp.2008.22
103
Hodgkinson
,
C.A.
,
Goldman
,
D.
,
Jaeger
,
J.
,
Persaud
,
S.
,
Kane
,
J.M.
,
Lipsky
,
R.H.
et al.
(
2004
)
Disrupted in schizophrenia 1 (DISC1): association with schizophrenia, schizoaffective disorder, and bipolar disorder
.
Am. J. Hum. Genet.
75
,
862
872
https://doi.org/10.1086/425586
104
Millar
,
J.K.
,
Christie
,
S.
,
Anderson
,
S.
,
Lawson
,
D.
,
Hsiao-Wei Loh
,
D.
,
Devon
,
R.S.
et al.
(
2001
)
Genomic structure and localisation within a linkage hotspot of Disrupted In Schizophrenia 1, a gene disrupted by a translocation segregating with schizophrenia
.
Mol. Psychiatry
6
,
173
178
https://doi.org/10.1038/sj.mp.4000784
105
Palo
,
O.M.
,
Antila
,
M.
,
Silander
,
K.
,
Hennah
,
W.
,
Kilpinen
,
H.
,
Soronen
,
P.
et al.
(
2007
)
Association of distinct allelic haplotypes of DISC1 with psychotic and bipolar spectrum disorders and with underlying cognitive impairments
.
Hum. Mol. Genet.
16
,
2517
2528
https://doi.org/10.1093/hmg/ddm207
106
Winham
,
S.J.
,
Cuellar-Barboza
,
A.B.
,
Oliveros
,
A.
,
McElroy
,
S.L.
,
Crow
,
S.
,
Colby
,
C.
et al.
(
2014
)
Genome-wide association study of bipolar disorder accounting for effect of body mass index identifies a new risk allele in TCF7L2
.
Mol. Psychiatry
19
,
1010
1016
https://doi.org/10.1038/mp.2013.159
107
Zandi
,
P.P.
,
Belmonte
,
P.L.
,
Willour
,
V.L.
,
Goes
,
F.S.
,
Badner
,
J.A.
,
Simpson
,
S.G.
et al.
(
2008
)
Association study of Wnt signaling pathway genes in bipolar disorder
.
Arch. Gen. Psychiatry
65
,
785
793
https://doi.org/10.1001/archpsyc.65.7.785
108
Potash
,
J.B.
,
Buervenich
,
S.
,
Cox
,
N.J.
,
Zandi
,
P.P.
,
Akula
,
N.
,
Steele
,
J.
et al.
(
2008
)
Gene‐based SNP mapping of a psychotic bipolar affective disorder linkage region on 22q12.3: association with HMG2L1 and TOM1
.
American J. of Med. Genetics Pt. B
147B
,
59
67
https://doi.org/10.1002/ajmg.b.30574
109
Yamada
,
M.
,
Ohkawara
,
B.
,
Ichimura
,
N.
,
Hyodo-Miura
,
J.
,
Urushiyama
,
S.
,
Shirakabe
,
K.
et al.
(
2003
)
Negative regulation of Wnt signalling by HMG2L1, a novel NLK-binding protein
.
Genes Cells
8
,
677
684
https://doi.org/10.1046/j.1365-2443.2003.00666.x
110
Yang
,
X.
and
Cheyette
,
B.N.R
. (
2013
)
SEC14 and spectrin domains 1 (Sestd1) and Dapper antagonist of catenin 1 (Dact1) scaffold proteins cooperatively regulate the Van Gogh-like 2 (Vangl2) four-pass transmembrane protein and planar cell polarity (PCP) pathway during embryonic development in mice
.
J. Biol. Chem.
288
,
20111
20120
https://doi.org/10.1074/jbc.M113.465427
111
Association
,
A.P
. (
2013
)
Diagnostic and Statistical Manual of Mental Disorders,DSM-5
. https://doi.org/10.1176/appi.books.9780890425596
112
Faraone
,
S.V.
and
Larsson
,
H
. (
2019
)
Genetics of attention deficit hyperactivity disorder
.
Mol. Psychiatry
24
,
562
575
https://doi.org/10.1038/s41380-018-0070-0
113
Antshel
,
K.M.
,
Zhang-James
,
Y.
,
Wagner
,
K.E.
,
Ledesma
,
A.
and
Faraone
,
S.V
. (
2016
)
An update on the comorbidity of ADHD and ASD: a focus on clinical management
.
Expert Rev. Neurother.
16
,
279
293
https://doi.org/10.1586/14737175.2016.1146591
114
Walter
,
N.M.
,
Yde Ohki
,
C.M.
,
Rickli
,
M.
,
Smigielski
,
L.
,
Walitza
,
S.
and
Grünblatt
,
E
. (
2024
)
An investigation on the alterations in Wnt signaling in ADHD across developmental stages
.
Neuroscience Applied
3
,
104070
https://doi.org/10.1016/j.nsa.2024.104070
115
Custodio
,
R.J.P.
,
Kim
,
H.J.
,
Kim
,
J.
,
Ortiz
,
D.M.
,
Kim
,
M.
,
Buctot
,
D.
et al.
(
2023
)
Hippocampal dentate gyri proteomics reveals Wnt signaling involvement in the behavioral impairment in the THRSP-overexpressing ADHD mouse model
.
Commun. Biol.
6
, 55 https://doi.org/10.1038/s42003-022-04387-5
116
Yde Ohki
,
C.M.
,
Grossmann
,
L.
,
Alber
,
E.
,
Dwivedi
,
T.
,
Berger
,
G.
,
Werling
,
A.M.
et al.
(
2020
)
The stress-Wnt-signaling axis: a hypothesis for attention-deficit hyperactivity disorder and therapy approaches
.
Transl. Psychiatry
10
, 315 https://doi.org/10.1038/s41398-020-00999-9
117
Grünblatt
,
E.
,
Nemoda
,
Z.
,
Werling
,
A.M.
,
Roth
,
A.
,
Angyal
,
N.
,
Tarnok
,
Z.
et al.
(
2019
)
The involvement of the canonical Wnt-signaling receptor LRP5 and LRP6 gene variants with ADHD and sexual dimorphism: association study and meta-analysis
.
Am. J. Med. Genet. B Neuropsychiatr. Genet.
180
,
365
376
https://doi.org/10.1002/ajmg.b.32695
118
Demontis
,
D.
,
Walters
,
R.K.
,
Martin
,
J.
,
Mattheisen
,
M.
,
Als
,
T.D.
,
Agerbo
,
E.
et al.
(
2019
)
Discovery of the first genome-wide significant risk loci for attention deficit/hyperactivity disorder
.
Nat. Genet.
51
,
63
75
https://doi.org/10.1038/s41588-018-0269-7
119
Demontis
,
D.
,
Walters
,
G.B.
,
Athanasiadis
,
G.
,
Walters
,
R.
,
Therrien
,
K.
,
Nielsen
,
T.T.
et al.
(
2023
)
Genome-wide analyses of ADHD identify 27 risk loci, refine the genetic architecture and implicate several cognitive domains
.
Nat. Genet.
55
,
198
208
https://doi.org/10.1038/s41588-022-01285-8
120
MacDonald
,
B.T.
,
Tamai
,
K.
and
He
,
X
. (
2009
)
Wnt/beta-catenin signaling: components, mechanisms, and diseases
.
Dev. Cell
17
,
9
26
https://doi.org/10.1016/j.devcel.2009.06.016
121
Apostolova
,
L.G
. (
2016
)
Alzheimer disease. contin.: lifelong learn
.
Neurol
22
,
419
434
https://doi.org/10.1212/CON.0000000000000307
122
Caricasole
,
A.
,
Copani
,
A.
,
Caraci
,
F.
,
Aronica
,
E.
,
Rozemuller
,
A.J.
,
Caruso
,
A.
et al.
(
2004
)
Induction of dickkopf-1, a negative modulator of the Wnt pathway, is associated with neuronal degeneration in Alzheimer’s brain
.
J. Neurosci.
24
,
6021
6027
https://doi.org/10.1523/JNEUROSCI.1381-04.2004
123
Rosi
,
M.C.
,
Luccarini
,
I.
,
Grossi
,
C.
,
Fiorentini
,
A.
,
Spillantini
,
M.G.
,
Prisco
,
A.
et al.
(
2010
)
Increased dickkopf-1 expression in transgenic mouse models of neurodegenerative disease
.
J. Neurochem.
112
,
1539
1551
https://doi.org/10.1111/j.1471-4159.2009.06566.x
124
Zhang
,
Z.
,
Hartmann
,
H.
,
Minh Do
,
V.
,
Abramowski
,
D.
,
Sturchler-Pierrat
,
C.
,
Staufenbiel
,
M.
et al.
(
1998
)
Destabilization of β-catenin by mutations in presenilin-1 potentiates neuronal apoptosis
.
Nature.
395
,
698
702
https://doi.org/10.1038/27208
125
Pei
,
J.J.
,
Braak
,
E.
,
Braak
,
H.
,
Grundke-Iqbal
,
I.
,
Iqbal
,
K.
,
Winblad
,
B.
et al.
(
1999
)
Distribution of active glycogen synthase kinase 3beta (GSK-3beta) in brains staged for Alzheimer disease neurofibrillary changes
.
J. Neuropathol. Exp. Neurol.
58
,
1010
1019
https://doi.org/10.1097/00005072-199909000-00011
126
Blauwendraat
,
C.
,
Heilbron
,
K.
,
Vallerga
,
C.L.
,
Bandres-Ciga
,
S.
,
von Coelln,
,
R.,
,
Pihlstrøm
,
L.
et al.
(
2019
)
Parkinson’s disease age at onset genome-wide association study: defining heritability, genetic loci, and α-synuclein mechanisms
.
Mov. Disord.
34
,
866
875
https://doi.org/10.1002/mds.27659
127
Del Tredici
,
K.
and
Braak
,
H
. (
2012
)
Lewy pathology and neurodegeneration in premotor Parkinson’s disease
.
Mov. Disord.
27
,
597
607
https://doi.org/10.1002/mds.24921
128
Inestrosa
,
N.C.
and
Arenas
,
E
. (
2010
)
Emerging roles of Wnts in the adult nervous system
.
Nat. Rev. Neurosci.
11
,
77
86
https://doi.org/10.1038/nrn2755
129
Nusse
,
R.
and
Clevers
,
H
. (
2017
)
Wnt/β-catenin signaling, disease, and emerging therapeutic modalities
.
Cell
169
,
985
999
https://doi.org/10.1016/j.cell.2017.05.016
130
Salašová
,
A.
,
Yokota
,
C.
,
Potěšil
,
D.
,
Zdráhal
,
Z.
,
Bryja
,
V.
and
Arenas
,
E
. (
2017
)
A proteomic analysis of LRRK2 binding partners reveals interactions with multiple signaling components of the WNT/PCP pathway
.
Mol. Neurodegener.
12
, 54 https://doi.org/10.1186/s13024-017-0193-9
131
Berwick
,
D.C.
and
Harvey
,
K
. (
2012
)
LRRK2 functions as a Wnt signaling scaffold, bridging cytosolic proteins and membrane-localized LRP6
.
Hum. Mol. Genet.
21
,
4966
4979
https://doi.org/10.1093/hmg/dds342
132
Roos
,
R.A.C
. (
2010
)
Huntington’s disease: a clinical review
.
Orphanet J. Rare Dis.
5
, 40 https://doi.org/10.1186/1750-1172-5-40
133
Macdonald
,
M
. (
1993
)
A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington’s disease chromosomes
.
Cell
72
,
971
983
https://doi.org/10.1016/0092-8674(93)90585-E
134
Ghatak
,
S.
and
Raha
,
S
. (
2015
)
Micro RNA-214 contributes to proteasome independent downregulation of beta catenin in Huntington’s disease knock-in striatal cell model STHdhQ111/Q111
.
Biochem. Biophys. Res. Commun.
459
,
509
514
https://doi.org/10.1016/j.bbrc.2015.02.137
135
Lim
,
R.G.
,
Quan
,
C.
,
Reyes-Ortiz
,
A.M.
,
Lutz
,
S.E.
,
Kedaigle
,
A.J.
,
Gipson
,
T.A.
et al.
(
2017
)
Huntington’s disease iPSC-derived brain microvascular endothelial cells reveal WNT-mediated angiogenic and blood-brain barrier deficits
.
Cell Rep.
19
,
1365
1377
https://doi.org/10.1016/j.celrep.2017.04.021
136
Carmichael
,
J.
,
Sugars
,
K.L.
,
Bao
,
Y.P.
and
Rubinsztein
,
D.C
. (
2002
)
Glycogen synthase kinase-3beta inhibitors prevent cellular polyglutamine toxicity caused by the Huntington’s disease mutation
.
J. Biol. Chem.
277
,
33791
33798
https://doi.org/10.1074/jbc.M204861200
137
Godin
,
J.D.
,
Poizat
,
G.
,
Hickey
,
M.A.
,
Maschat
,
F.
and
Humbert
,
S
. (
2010
)
Mutant huntingtin-impaired degradation of beta-catenin causes neurotoxicity in huntington’s disease
.
EMBO J.
29
,
2433
2445
https://doi.org/10.1038/emboj.2010.117
138
Gines
,
S.
,
Ivanova
,
E.
,
Seong
,
I.S.
,
Saura
,
C.A.
and
MacDonald
,
M.E
. (
2003
)
Enhanced Akt signaling is an early pro-survival response that reflects N-methyl-D-aspartate receptor activation in huntington’s disease knock-in striatal cells
.
J. Biol. Chem.
278
,
50514
50522
https://doi.org/10.1074/jbc.M309348200
139
Dupont
,
P.
,
Besson
,
M.T.
,
Devaux
,
J.
and
Liévens
,
J.C
. (
2012
)
Reducing canonical Wingless/Wnt signaling pathway confers protection against mutant huntingtin toxicity in drosophila
.
Neurobiol. Dis.
47
,
237
247
https://doi.org/10.1016/j.nbd.2012.04.007
140
Smith-Geater
,
C.
,
Hernandez
,
S.J.
,
Lim
,
R.G.
,
Adam
,
M.
,
Wu
,
J.
,
Stocksdale
,
J.T
. et al.
(
2020
)
Aberrant development corrected in adult-onset huntington’s disease iPSC-derived neuronal cultures via WNT signaling modulation
.
Stem Cell Reports
14
,
406
419
https://doi.org/10.1016/j.stemcr.2020.01.015
141
Orr
,
H.T.
,
Chung
,
M.Y.
,
Banfi
,
S.
,
Kwiatkowski
,
T.J.
Jr
,
Servadio
,
A.
,
Beaudet
,
A.L.
et al.
(
1993
)
Expansion of an unstable trinucleotide CAG repeat in spinocerebellar ataxia type 1
.
Nat. Genet.
4
,
221
226
https://doi.org/10.1038/ng0793-221
142
Lein
,
E.S.
,
Hawrylycz
,
M.J.
,
Ao
,
N.
,
Ayres
,
M.
,
Bensinger
,
A.
,
Bernard
,
A.
et al.
(
2007
)
Genome-wide atlas of gene expression in the adult mouse brain
.
Nature.
445
,
168
176
https://doi.org/10.1038/nature05453
143
Coyle-Rink
,
J.
,
Del Valle
,
L.
,
Sweet
,
T.
,
Khalili
,
K.
and
Amini
,
S
. (
2002
)
Developmental expression of Wnt signaling factors in mouse brain
.
Cancer Biol. Ther.
1
,
640
645
https://doi.org/10.4161/cbt.313
144
Luttik
,
K.
,
Tejwani
,
L.
,
Ju
,
H.
,
Driessen
,
T.
,
Smeets
,
C.J.L.M.
,
Edamakanti
,
C.R.
et al.
(
2022
)
Differential effects of Wnt-β-catenin signaling in Purkinje cells and Bergmann glia in spinocerebellar ataxia type 1
.
Proc. Natl. Acad. Sci. U.S.A.
119
, e2208513119 https://doi.org/10.1073/pnas.2208513119
145
Ingram
,
M.
,
Wozniak
,
E.A.L.
,
Duvick
,
L.
,
Yang
,
R.
,
Bergmann
,
P.
,
Carson
,
R.
et al.
(
2016
)
Cerebellar transcriptome profiles of ATXN1 transgenic mice reveal SCA1 disease progression and protection pathways
.
Neuron
89
,
1194
1207
https://doi.org/10.1016/j.neuron.2016.02.011
146
Libro
,
R.
,
Bramanti
,
P.
and
Mazzon
,
E
. (
2016
)
The role of the Wnt canonical signaling in neurodegenerative diseases
.
Life Sci. (1962).
158
,
78
88
https://doi.org/10.1016/j.lfs.2016.06.024
147
Lee
,
Y.
,
Lee
,
J.K.
,
Ahn
,
S.H.
,
Lee
,
J.
and
Nam
,
D.H
. (
2016
)
WNT signaling in glioblastoma and therapeutic opportunities
.
Lab. Invest.
96
,
137
150
https://doi.org/10.1038/labinvest.2015.140
148
Tsukamoto
,
A.S.
,
Grosschedl
,
R.
,
Guzman
,
R.C.
,
Parslow
,
T.
and
Varmus
,
H.E
. (
1988
)
Expression of the int-1 gene in transgenic mice is associated with mammary gland hyperplasia and adenocarcinomas in male and female mice
.
Cell
55
,
619
625
https://doi.org/10.1016/0092-8674(88)90220-6
149
Nusse
,
R.
,
van Ooyen
,
A.
,
Cox
,
D.
,
Fung
,
Y.K.T.
and
Varmus
,
H
. (
1984
)
Mode of proviral activation of a putative mammary oncogene (int-1) on mouse chromosome 15
.
Nature.
307
,
131
136
https://doi.org/10.1038/307131a0
150
Nusse
,
R.
and
Varmus
,
H.E
. (
1982
)
Many tumors induced by the mouse mammary tumor virus contain a provirus integrated in the same region of the host genome
.
Cell
31
,
99
109
https://doi.org/10.1016/0092-8674(82)90409-3
151
Lie
,
D.C
,
Colamarino
,
S.A.
,
Song
,
H.J.
,
Désiré
,
L.
,
Mira
,
H.
,
Consiglio
,
A.
et al.
(
2005
)
Wnt signalling regulates adult hippocampal neurogenesis
.
Nature.
437
,
1370
1375
https://doi.org/10.1038/nature04108
152
Chenn
,
A.
and
Walsh
,
C.A
. (
2002
)
Regulation of cerebral cortical size by control of cell cycle exit in neural precursors
.
Science
297
,
365
369
https://doi.org/10.1126/science.1074192
153
Megason
,
S.G.
and
McMahon
,
A.P
. (
2002
)
A mitogen gradient of dorsal midline Wnts organizes growth in the CNS
.
Development (Rome)
129
,
2087
2098
https://doi.org/10.1242/dev.129.9.2087
154
Verhaak
,
R.G.W.
,
Hoadley
,
K.A.
,
Purdom
,
E.
,
Wang
,
V.
,
Qi
,
Y.
,
Wilkerson
,
M.D.
et al.
(
2010
)
Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1
.
Cancer Cell
17
,
98
110
https://doi.org/10.1016/j.ccr.2009.12.020
155
Park
,
A.K.
,
Kim
,
P.
,
Ballester
,
L.Y.
,
Esquenazi
,
Y.
and
Zhao
,
Z
. (
2019
)
Subtype-specific signaling pathways and genomic aberrations associated with prognosis of glioblastoma
.
Neuro-oncology
21
,
59
70
https://doi.org/10.1093/neuonc/noy120
156
Logan
,
C.Y.
and
Nusse
,
R
. (
2004
)
The Wnt signaling pathway in development and disease
.
Annu. Rev. Cell Dev. Biol.
20
,
781
810
https://doi.org/10.1146/annurev.cellbio.20.010403.113126
157
Morris
,
L.G.T.
,
Kaufman
,
A.M.
,
Gong
,
Y.
,
Ramaswami
,
D.
,
Walsh
,
L.A.
,
Turcan
,
Ş.
et al.
(
2013
)
Recurrent somatic mutation of FAT1 in multiple human cancers leads to aberrant Wnt activation
.
Nat. Genet.
45
,
253
261
https://doi.org/10.1038/ng.2538
158
Manfreda
,
L.
,
Rampazzo
,
E.
and
Persano
,
L
. (
2023
)
Wnt signaling in brain tumors: a challenging therapeutic target
.
Biology (Basel)
12
, 729 https://doi.org/10.3390/biology12050729
159
Silva
,
R. da
,
Marie
,
S.K.N.
,
Uno
,
M.
,
Matushita
,
H.
,
Wakamatsu
,
A.
,
Rosemberg
,
S.
et al.
(
2013
)
CTNNB1, AXIN1 and APC expression analysis of different medulloblastoma variants
.
Clinics (Sao Paulo)
68
,
167
172
https://doi.org/10.6061/clinics/2013(02)oa08
160
Northcott
,
P.A.
,
Buchhalter
,
I.
,
Morrissy
,
A.S.
,
Hovestadt
,
V.
,
Weischenfeldt
,
J.
,
Ehrenberger
,
T.
et al.
(
2017
)
The whole-genome landscape of medulloblastoma subtypes
.
Nature.
547
,
311
317
https://doi.org/10.1038/nature22973
161
Crawley
,
J.N.
,
Heyer
,
W.D.
and
LaSalle
,
J.M
. (
2016
)
Autism and cancer share risk genes, pathways, and drug targets
.
Trends Genet.
32
,
139
146
https://doi.org/10.1016/j.tig.2016.01.001
162
Pinto
,
D.
,
Delaby
,
E.
,
Merico
,
D.
,
Barbosa
,
M.
,
Merikangas
,
A.
,
Klei
,
L.
et al.
(
2014
)
Convergence of genes and cellular pathways dysregulated in autism spectrum disorders
.
Am. J. Hum. Genet.
94
,
677
694
https://doi.org/10.1016/j.ajhg.2014.03.018
163
Zhou
,
J.
and
Parada
,
L.F
. (
2012
)
PTEN signaling in autism spectrum disorders
.
Curr. Opin. Neurobiol.
22
,
873
879
https://doi.org/10.1016/j.conb.2012.05.004
164
Blatt
,
J.
,
Deal
,
A.M.
and
Mesibov
,
G
. (
2010
)
Autism in children and adolescents with cancer
.
Pediatr. Blood Cancer
54
,
144
147
https://doi.org/10.1002/pbc.22303
165
Wen
,
Y.
and
Herbert
,
M.R
. (
2017
)
Connecting the dots: overlaps between autism and cancer suggest possible common mechanisms regarding signaling pathways related to metabolic alterations
.
Med. Hypotheses
103
,
118
123
https://doi.org/10.1016/j.mehy.2017.05.004
166
Serafino
,
A.
,
Giovannini
,
D.
,
Rossi
,
S.
and
Cozzolino
,
M
. (
2020
)
Targeting the Wnt/β-catenin pathway in neurodegenerative diseases: recent approaches and current challenges
.
Expert Opin. Drug Discov.
15
,
803
822
https://doi.org/10.1080/17460441.2020.1746266
167
Rodon
,
J.
,
Argilés
,
G.
,
Connolly
,
R.M.
,
Vaishampayan
,
U.
,
de Jonge
,
M.
,
Garralda
,
E.
et al.
(
2021
)
Phase 1 study of single-agent WNT974, a first-in-class porcupine inhibitor, in patients with advanced solid tumours
.
Br. J. Cancer
125
,
28
37
https://doi.org/10.1038/s41416-021-01389-8
168
Huang
,
S.M.A.
,
Mishina
,
Y.M.
,
Liu
,
S.
,
Cheung
,
A.
,
Stegmeier
,
F.
,
Michaud
,
G.A.
et al.
(
2009
)
Tankyrase inhibition stabilizes axin and antagonizes Wnt signalling
.
Nature.
461
,
614
620
https://doi.org/10.1038/nature08356
169
Thorne
,
C.A.
,
Hanson
,
A.J.
,
Schneider
,
J.
,
Tahinci
,
E.
,
Orton
,
D.
,
Cselenyi
,
C.S.
et al.
(
2010
)
Small-molecule inhibition of Wnt signaling through activation of casein kinase 1α
.
Nat. Chem. Biol.
6
,
829
836
https://doi.org/10.1038/nchembio.453
170
Shan
,
J.
,
Shi
,
D.L.
,
Wang
,
J.
and
Zheng
,
J
. (
2005
)
Identification of a specific inhibitor of the dishevelled PDZ domain
.
Biochemistry
44
,
15495
15503
https://doi.org/10.1021/bi0512602
171
Bodine
,
P.V.N.
,
Stauffer
,
B.
,
Ponce-de-Leon
,
H.
,
Bhat
,
R.A.
,
Mangine
,
A.
,
Seestaller-Wehr
,
L.M.
et al.
(
2009
)
A small molecule inhibitor of the Wnt antagonist secreted frizzled-related protein-1 stimulates bone formation
.
Bone
44
,
1063
1068
https://doi.org/10.1016/j.bone.2009.02.013
172
Lee
,
H.J.
,
Wang
,
N.X.
,
Shi
,
D.L.
and
Zheng
,
J.J
. (
2009
)
Sulindac inhibits canonical Wnt signaling by blocking the PDZ domain of the protein dishevelled
.
Angew. Chem. Int. Ed. Engl.
48
,
6448
6452
https://doi.org/10.1002/anie.200902981
173
Wang
,
J.Y.
,
Wang
,
X.
,
Wang
,
X.J.
,
Zheng
,
B.Z.
,
Wang
,
Y.
,
Wang
,
X.
et al.
(
2018
)
Curcumin inhibits the growth via Wnt/β-catenin pathway in non-small-cell lung cancer cells
.
Eur. Rev. Med. Pharmacol. Sci.
22
,
7492
7499
https://doi.org/10.26355/eurrev_201811_16290
174
L’episcopo
,
F.
,
Serapide
,
M.F.
,
Tirolo
,
C.
,
Testa
,
N.
,
Caniglia
,
S.
,
Morale
,
M.C.
et al.
(
2011
)
A Wnt1 regulated Frizzled-1/β-Catenin signaling pathway as a candidate regulatory circuit controlling mesencephalic dopaminergic neuron-astrocyte crosstalk: therapeutical relevance for neuron survival and neuroprotection
.
Mol. Neurodegener.
6
, 49 https://doi.org/10.1186/1750-1326-6-49
175
Zhang
,
N.
,
Westerhaus
,
A.
,
Wilson
,
M.
,
Wang
,
E.
,
Goff
,
L.
and
Sockanathan
,
S
. (
2024
)
Physiological regulation of neuronal Wnt activity is essential for TDP-43 localization and function
.
EMBO J.
43
,
3388
3413
https://doi.org/10.1038/s44318-024-00156-8
176
Liu
,
J.
,
Pan
,
S.
,
Hsieh
,
M.H.
,
Ng
,
N.
,
Sun
,
F.
,
Wang
,
T.
et al.
(
2013
)
Targeting Wnt-driven cancer through the inhibition of Porcupine by LGK974
.
Proc. Natl. Acad. Sci. U.S.A.
110
,
20224
20229
https://doi.org/10.1073/pnas.1314239110
177
Stakheev
,
D.
,
Taborska
,
P.
,
Strizova
,
Z.
,
Podrazil
,
M.
,
Bartunkova
,
J.
and
Smrz
,
D
. (
2019
)
The WNT/β-catenin signaling inhibitor XAV939 enhances the elimination of LNCaP and PC-3 prostate cancer cells by prostate cancer patient lymphocytes in vitro
.
Sci. Rep.
9
, 4761 https://doi.org/10.1038/s41598-019-41182-5
178
Singh
,
S.S.
,
Rai
,
S.N.
,
Birla
,
H.
,
Zahra
,
W.
,
Rathore
,
A.S.
and
Singh
,
S.P
. (
2020
)
NF-κB-mediated neuroinflammation in parkinson’s disease and potential therapeutic effect of polyphenols
.
Neurotox. Res.
37
,
491
507
https://doi.org/10.1007/s12640-019-00147-2
179
Vlad
,
S.C.
,
Miller
,
D.R.
,
Kowall
,
N.W.
and
Felson
,
D.T
. (
2008
)
Protective effects of NSAIDs on the development of Alzheimer disease
.
Neurology (ECronicon).
70
,
1672
1677
https://doi.org/10.1212/01.wnl.0000311269.57716.63
180
Côté
,
S.
,
Carmichael
,
P.H.
,
Verreault
,
R.
,
Lindsay
,
J.
,
Lefebvre
,
J.
and
Laurin
,
D
. (
2012
)
Nonsteroidal anti-inflammatory drug use and the risk of cognitive impairment and Alzheimer’s disease
.
Alzheimers. Dement.
8
,
219
226
https://doi.org/10.1016/j.jalz.2011.03.012
181
Landi
,
F.
,
Cesari
,
M.
,
Onder
,
G.
,
Russo
,
A.
,
Torre
,
S.
and
Bernabei
,
R
. (
2003
)
Non-steroidal anti-inflammatory drug (NSAID) use and Alzheimer disease in community-dwelling elderly patients
.
Am. J. Geriatr. Psychiatry
11
,
179
185
https://doi.org/10.1097/00019442-200303000-00008
182
Modi
,
J.P.
,
Prentice
,
H.
and
Wu
,
J.Y
. (
2014
)
Sulindac for stroke treatment: neuroprotective mechanism and therapy
.
Neural Regen. Res.
9
,
2023
2025
https://doi.org/10.4103/1673-5374.147919
183
Collu
,
G.M.
,
Hidalgo-Sastre
,
A.
and
Brennan
,
K
. (
2014
)
Wnt-Notch signalling crosstalk in development and disease
.
Cell. Mol. Life Sci.
71
,
3553
3567
https://doi.org/10.1007/s00018-014-1644-x
184
Wilson
,
N.H.
and
Stoeckli
,
E.T
. (
2012
)
Sonic hedgehog regulates Wnt activity during neural circuit formation
.
Vitam. Horm.
88
,
173
209
https://doi.org/10.1016/B978-0-12-394622-5.00008-0
185
Matoba
,
N.
,
Le
,
B.D.
,
Valone
,
J.M.
,
Wolter
,
J.M.
,
Mory
,
J.T.
,
Liang
,
D.
et al.
(
2024
)
Stimulating Wnt signaling reveals context-dependent genetic effects on gene regulation in primary human neural progenitors
.
Nat. Neurosci.
27
,
2430
2442
https://doi.org/10.1038/s41593-024-01773-6
186
Noelanders
,
R.
and
Vleminckx
,
K
. (
2017
)
How Wnt signaling builds the brain: bridging development and disease
.
Neuroscientist
23
,
314
329
https://doi.org/10.1177/1073858416667270
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY).