1,318
Views
0
CrossRef citations to date
0
Altmetric
Research Article

Overlap in synaptic neurological condition susceptibility pathways and the neural pannexin 1 interactome revealed by bioinformatics analyses

ORCID Icon, ORCID Icon & ORCID Icon
Article: 2253102 | Received 12 May 2023, Accepted 22 Aug 2023, Published online: 08 Oct 2023

ABSTRACT

Many neurological conditions exhibit synaptic impairments, suggesting mechanistic convergence. Additionally, the pannexin 1 (PANX1) channel and signaling scaffold is linked to several of these neurological conditions and is an emerging regulator of synaptic development and plasticity; however, its synaptic pathogenic contributions are relatively unexplored. To this end, we explored connections between synaptic neurodevelopmental disorder and neurodegenerative disease susceptibility genes discovered by genome-wide association studies (GWASs), and the neural PANX1 interactome (483 proteins) identified from mouse Neuro2a (N2a) cells. To identify shared susceptibility genes, we compared synaptic suggestive GWAS candidate genes amongst autism spectrum disorders, schizophrenia, Parkinson’s disease, and Alzheimer’s disease. To further probe PANX1 signaling pathways at the synapse, we used bioinformatics tools to identify PANX1 interactome signaling pathways and protein–protein interaction clusters. To shed light on synaptic disease mechanisms potentially linking PANX1 and these four neurological conditions, we performed additional cross-analyses between gene ontologies enriched for the PANX1 synaptic and disease-susceptibility gene sets. Finally, to explore the regional specificity of synaptic PANX1-neurological condition connections, we identified brain region-specific elevations of synaptic PANX1 interactome and GWAS candidate gene set transcripts. Our results confirm considerable overlap in risk genes for autism spectrum disorders and schizophrenia and identify potential commonalities in genetic susceptibility for neurodevelopmental disorders and neurodegenerative diseases. Our findings also pinpointed novel putative PANX1 links to synaptic disease-associated pathways, such as regulation of vesicular trafficking and proteostasis, warranting further validation.

Introduction

Dendritic spines are the site of post-synaptic communication between neurons. Several neurodevelopmental and neurodegenerative conditions, such as autism spectrum disorders (ASD), schizophrenia, Parkinson’s disease, and Alzheimer’s disease, exhibit divergent dendritic spine size, stability, and/or function [Citation1–8]. These alterations often precede obvious clinical symptoms, suggesting they could be involved in disease susceptibility and progression. Despite this, current understanding of the mechanisms affecting dendritic spine dynamics in these conditions is limited.

The pannexin 1 (PANX1) channel and cytoskeleton-regulating protein is emerging as a key regulator of dendritic spines. PANX1, highly expressed at post-synaptic densities [Citation9], oligomerizes to form ion and metabolite channels, and also acts as a channel-independent signaling hub [Citation10]. For example, we discovered protein–protein interactions (PPIs) between PANX1 and key cytoskeleton-regulating proteins involved in dendritic spine formation and stability [Citation11–13], including collapsin response mediator protein 2 (CRMP2) and actin-related protein 3 (ARP3 of the ARP2/3 complex) [Citation14–18]. As follows, PANX1 knock out mice exhibit several synaptic and behavioral abnormalities, such as altered hippocampal long-term potentiation and long-term depression, impaired object recognition, spatial memory and reversal learning, and increased anxiety [Citation19–21]. Consistent with these findings, we discovered that PANX1 limits cortical neuron network size and complexity through inhibition of dendritic spine density and stability [Citation22,Citation23], and similarly, others have shown that PANX1 hinders hippocampal neuron spine maturation [Citation24]. Not surprisingly, PANX1 and/or its protein interaction partners CRMP2 and ARP2/3 are implicated in several neurological conditions associated with dendritic spine abnormalities [Citation25–31], raising the possibility that PANX1 and its interaction partners may contribute to disease risk and/or progression via dendritic spine modulation.

Therefore, here we aimed to identify potential links between synaptic neurological condition susceptibility genes and PANX1 by exploring the extent of overlap in (1) synaptic genes involved in four common neurological conditions associated with broad genetic susceptibility [Citation32–36] and atypical dendritic spines, and (2) these same genes with the neural PANX1 interactome. The neurological conditions we selected, ASD, schizophrenia, Parkinson’s disease, and Alzheimer’s disease, are commonly known and contribute to substantial socioeconomic disease burden [Citation37–40]. Moreover, we performed an in-depth bioinformatics analysis of the neural PANX1 interactome to identify PANX1 interaction partners potentially relevant to dendritic spine dynamics using in silico tools (). We first cross-referenced findings from Genome-Wide Association Studies (GWASs) for ASD, schizophrenia, Parkinson’s disease, and Alzheimer’s disease, to identify common suggestive risk susceptibility genes. We then performed enrichment analyses of the total neural PANX1 interactome to identify overrepresented biological pathways (e.g., relating to neurological diseases), including implicated PANX1-interacting proteins. We next identified links between existing PPI networks and synaptic PANX1-interacting proteins, the latter obtained by identifying protein hits from our mouse N2a cell PANX1-EGFP interactome annotated to the Gene Ontology (GO) term “synapse”. This was done to gain further insight into the molecular mechanisms that might underlie PANX1 regulation of dendritic spines. To investigate potential links between PANX1 and neurological conditions exhibiting dendritic spine pathology, we compared cellular localizations and biological functions of the synaptic PANX1 interactome with synaptic-enriched susceptibility genes for ASD, schizophrenia, Parkinson’s disease, and Alzheimer’s disease identified by GWASs.

Figure 1. Workflow for the current study from materials and methods to results. In this study, a number of subanalyses were conducted, which explored the neurodevelopmental disorder and neurodegenerative disease susceptibility genes, and the PANX1 interactome in mouse N2a cells overlapping with the gene ontology (GO) synapse. The input data was obtained from three sources: The genome-wide association study (GWAS) catalog (Homo sapiens) [Citation41], Swayne lab [our lab] (Mus musculus), and the GO knowledge base (Homo sapiens and Mus musculus) via PANTHER [protein ANalysis THrough Evolutionary Relationships] [Citation42,Citation43], which is a classification system. To enable cross analyses between these datasets, the resources UniProt Retrieve/ID mapping tool, biological DataBase network (bioDBnet) [Citation44], HUGO gene Nomenclature Committee (HGNC) database [Citation45] and mouse genome informatics (MGI) international database [Citation46,Citation47] were used (e.g., to identify human orthologs for mouse UniProtKB IDs). The data were then analyzed using the statistical computing environment R and the following bioinformatics tools/databases: PANTHER [Citation42,Citation48,Citation49], the Human Protein Atlas [Citation50,Citation51] and STRING [Search tool for the Retrieval of interacting genes/proteins] [Citation52–54]. In addition to these subanalyses, the findings were compared to provide a comprehensive overview for each neurodevelopment disorder and neurodegenerative disease in relation to the PANX1 interactome, and overrepresented PANTHER pathways were identified for the PANX1 interactome.

Figure 1. Workflow for the current study from materials and methods to results. In this study, a number of subanalyses were conducted, which explored the neurodevelopmental disorder and neurodegenerative disease susceptibility genes, and the PANX1 interactome in mouse N2a cells overlapping with the gene ontology (GO) synapse. The input data was obtained from three sources: The genome-wide association study (GWAS) catalog (Homo sapiens) [Citation41], Swayne lab [our lab] (Mus musculus), and the GO knowledge base (Homo sapiens and Mus musculus) via PANTHER [protein ANalysis THrough Evolutionary Relationships] [Citation42,Citation43], which is a classification system. To enable cross analyses between these datasets, the resources UniProt Retrieve/ID mapping tool, biological DataBase network (bioDBnet) [Citation44], HUGO gene Nomenclature Committee (HGNC) database [Citation45] and mouse genome informatics (MGI) international database [Citation46,Citation47] were used (e.g., to identify human orthologs for mouse UniProtKB IDs). The data were then analyzed using the statistical computing environment R and the following bioinformatics tools/databases: PANTHER [Citation42,Citation48,Citation49], the Human Protein Atlas [Citation50,Citation51] and STRING [Search tool for the Retrieval of interacting genes/proteins] [Citation52–54]. In addition to these subanalyses, the findings were compared to provide a comprehensive overview for each neurodevelopment disorder and neurodegenerative disease in relation to the PANX1 interactome, and overrepresented PANTHER pathways were identified for the PANX1 interactome.

The outcomes of this work provide potential new insights into the role of PANX1 in the central nervous system (CNS) and suggest links between PANX1 and neurological conditions.

Materials and methods

An overview of the study workflow can be found in . For the comparisons and analysis, the R statistical computing environment v4.2.2 was applied. On 2 June 2020, the University of Victoria’s Human Research Ethics Board exempted the study from ethical review as the study: 1) is limited to accessing publicly available data sets and 2) does not involve human participants. Biosafety approval was obtained from the University of Victoria Biosafety committee to undertake the experiments identifying PANX1 interacting proteins. For clarity, it is important to note that according to international conventions (https://www.informatics.jax.org/mgihome/nomen/gene.shtml), all mouse protein abbreviations are fully capitalized. Accordingly, the mouse and human pannexin 1 (also referred to pannexin-1 and pannexin1 in the literature) protein abbreviation is “PANX1.” The gene abbreviation for the mouse pannexin 1 gene is Panx1, and the gene abbreviation for the human pannexin 1 gene is PANX1. These are the conventions we follow in this manuscript.

Data inputs

Extraction of suggestive GWAS candidate genes involved in neurodevelopmental disorders and neurodegenerative diseases in humans

On 13 November 2022, we searched for the Experimental Factor Ontology (EFO) [Citation55] trait labels “autism spectrum disorder”, “schizophrenia”, “Parkinson disease”, and “Alzheimer disease” separately in the NHGRI-EBI GWAS catalog API [Citation41,Citation56] by means of the gwasrapidd R package [Citation57] and extracted information about the studies (some publications contain multiple GWASs), variants (or single nucleotide polymorphisms; SNPs) and associations (SNP-trait associations). For Parkinson’s disease, 61 studies and 494 associations were available; for Alzheimer’s disease, 117 studies and 1988 associations were available; for schizophrenia, 131 studies, 4961 associations were available; and for ASD, 35 studies and 1275 associations were available. Information about the original publications, including accession ID of the GWAS Catalog study, can be found in Supplemental Table S1.

Using the extracted data, associations without gene information were excluded (gene symbols or entrez IDs; symbols beginning with LOC were kept) and only suggestive GWAS candidate genes reported more than once were included for further analysis. Based on these filtering criteria, a total of 461, 881, 74, and 86 suggestive GWAS candidate genes were identified for ASD, schizophrenia, Parkinson’s disease, and Alzheimer’s disease, respectively. The complete lists of suggestive GWAS candidate genes obtained after filtering were used as inputs for the cross-analyses.

Identification of the PANX1 interactome in mouse N2a cells

We previously identified the putative PANX1 interactome from mouse N2a neuroblastoma-derived cells, using methods that were comprehensively described in that work [Citation11,Citation13]. Briefly, proteins co-precipitating with PANX1EGFP [enhanced green fluorescent protein] or EGFP from N2a cells were identified by the UVIC-Genome BC Proteomics Centre using high-performance liquid chromatography-tandem mass spectrometry (LC-MS/MS) followed by analysis with Proteome Discoverer v1.3.0.339 (Thermo Scientific) and Mascot v2.2 [Citation58] [percolator settings: Max delta Cn 0.05, Target false discovery rate (FDR) strict 0.01, Target FDR relaxed 0.05 with validation based on q-value]. The q-value refers to the minimal FDR at which a peptide spectrum match was accepted [Citation59]. The full list of PANX1-interacting proteins can be found in Supplemental Table S2. The first column contains the UniProt mouse accession ID number and abbreviation, and the second column contains the full protein name. This accession number can be queried at uniprot.org for a full entry on the protein, including information such as alternative names, gene name, function, GO annotations, subcellular location, and post-translational modifications. To identify proteins selectively interacting with PANX1, all proteins co-precipitating with anti-GFP antibody from EGFP-expressing cells were removed from the list of PANX1 interactors. This paradigm was repeated three times, and the results were pooled. The complete list of PANX1-interacting proteins identified from those experiments (not previously published in its entirety) was used as input for the cross-analyses.

Cross-analysis

Cross-analysis of the suggestive disorder/disease susceptibility human GWAS candidate genes and mouse neural PANX1 interactome with the GO term “synapse”

To identify known synaptic genes, genes annotated to the “synapse” GO term were extracted for Homo sapiens and Mus musculus using the PANTHER database v17.0 and the GO Ontology database released on 1 July 2022 [Citation42,Citation48,Citation49,Citation60,Citation61]. Second, the entrez IDs were obtained by converting UniProtKB IDs from the Homo sapiens synapse GO term gene list using the UniProt Retrieve/ID mapping tool (https://www.uniprot.org/id-mapping), and finding the human orthologs for the MGI IDs from the Mus musculus GO term gene list using the biological DataBase network (bioDBnet; http://biodbnet.abcc.ncifcrf.gov) [Citation44]. When unable to obtain the entrez IDs, we manually looked up the entrez IDs in the HUGO Gene Nomenclature Committee (HGNC) database [Citation45], or the human orthologs in the Mouse Genome Informatics (MGI) international database [Citation46,Citation47]. The gene lists were combined and used as input for the cross-analyses.

To allow for comparison with the GWAS findings, human orthologs in the form of entrez IDs were identified for the mouse UniProtKB IDs forming the PANX1 interactome using bioDBnet [Citation44]. As described above, for the genes we were unable to obtain entrez IDs, we manually looked up the human orthologs in the MGI international database [Citation46].

Next, we conducted cross-analyses between the GO term “synapse” (GO:0045202; identified using the PANTHER database [Citation42,Citation43,Citation49]) and the (i) suggestive GWAS candidate genes involved in the selected human neurodegenerative diseases and neurodevelopmental disorders, and (ii) proteins comprising the mouse PANX1 interactome. This resulted in the identification of synaptic genes/proteins, and subsequently overlaps between the lists of synaptic suggestive GWAS candidate genes for the neurological conditions and synaptic PANX1-interacting proteins were examined.

Downstream analyses

Functional annotation, categorization, and overrepresentation analyses of the synaptic suggestive GWAS candidate genes and PANX1 interactome using the PANTHER database

Bioinformatics analysis of the synaptic suggestive GWAS candidate genes for each neurodevelopmental disorder and neurodegenerative disease (using Homo sapiens Entrez gene identifiers), proteins from our PANX1-interacting protein list (using Mus musculus UniProtKB unique identifiers), and synaptic proteins from our PANX1-interacting protein list (using Mus musculus UniProtKB unique identifiers or Homo sapiens Entrez gene identifiers) was carried out using the curated database PANTHER v17.0 [Citation42,Citation43,Citation49]. The genes/proteins were annotated to (i) PANTHER pathways [Citation62] created using the CellDesigner tool, a modeling tool for biochemical networks [Citation63], and/or (ii) GO terms within the Biological Process and Cellular Component domains from the GO knowledge base [Citation60,Citation61]. Right-tailed Fisher’s exact tests were used to identify overrepresented PANTHER pathways and GO terms (present in greater abundance than would be expected). FDR-corrected p-values <0.05 (to account for multiple testing) were considered statistically significant. In addition, only PANTHER pathways and GO terms with at least 10 annotated proteins/genes were presented (to reduce the likelihood of false positives; to allow for comparison, this was not done for and corresponding analysis).

Figure 2. Overlap between genes associated with neurodevelopmental disorders and neurodegenerative diseases in humans based on genome-wide association study (GWAS) findings. Venn diagram displaying the number of suggestive candidate genes the selected neurodevelopmental disorders, autism spectrum disorder (ASD) and schizophrenia, and the selected neurodegenerative diseases, Parkinson’s disease and Alzheimer’s disease, have in common.

Figure 2. Overlap between genes associated with neurodevelopmental disorders and neurodegenerative diseases in humans based on genome-wide association study (GWAS) findings. Venn diagram displaying the number of suggestive candidate genes the selected neurodevelopmental disorders, autism spectrum disorder (ASD) and schizophrenia, and the selected neurodegenerative diseases, Parkinson’s disease and Alzheimer’s disease, have in common.

PPI network for the PANX1 synaptic interactome

The synaptic PPI network, based on interaction evidence [from STRING-defined categories: known interactions (curated databases, and experimentally determined)], was created for Mus musculus using the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) v11.5 [Citation52–54], and the identified PANX1-interacting synaptic proteins were used as input (those proteins overlapping with the Mus musculus synapse GO term using the PANTHER v17.0 database and the GO Ontology database released on 1 July 2022 [Citation42,Citation48,Citation49,Citation60,Citation61]). Edges (also termed PPIs) were formed if the interaction score was at least 0.4 (medium confidence, “estimated likelihood that a given interaction is biologically meaningful, specific and reproducible, given the supporting evidence” [Citation64]), and the thickness of the edges in the PPI network indicates the strength of data support and dashed line edges reveal PPIs between clusters. First, clusters were identified using the unsupervised Markov Cluster (MCL) algorithm with inflation factor 1.3 (higher inflation factor leads to more clusters but noting that “MCL is remarkably robust to graph alterations”) [Citation65–67]. The outcome of this analysis was used to select the final number of clusters in the PPI network, and k-means clustering was carried out. A two-step process was used for quality control purposes (i.e., combining bioinformatics approaches and expert knowledge for enhanced confidence in the findings). The potential function of the clusters (formed by at least four proteins) was investigated using the PANTHER database v17.0 [Citation42,Citation48,Citation49] focusing specifically on PANTHER protein classes and GO biological process terms.

Synaptic PANX1-interacting proteins and suggestive GWAS candidate genes expressed in specific brain regions in humans

RNA expression in human brain regions was explored using the Human Protein Atlas v22.0 (proteinatlas.org) [Citation50,Citation51], and focusing specifically on genes classified as “regionally elevated.” The findings were compared with the synaptic PANX1 interactome and suggestive GWAS candidate genes for ASD, schizophrenia, Parkinson’s disease, and Alzheimer’s disease. Genes elevated in the following nine brain regions were included in the cross-analysis (UniProtKB IDs were converted to entrez IDs as described above): Cerebral cortex, hippocampal formation, amygdala, thalamus, hypothalamus, midbrain, pons, cerebellum, and medulla oblongata (https://www.proteinatlas.org/humanproteome/brain).

Results

Comparison amongst neurological conditions exhibiting impaired synapse structure and/or stability revealed shared synaptic suggestive GWAS candidate genes

We focused our study on four major neurological conditions exhibiting synapse instability, namely ASD, schizophrenia, Parkinson’s disease, and Alzheimer’s disease. ASD and schizophrenia had more synaptic suggestive GWAS candidate genes in common than any other disease–disorder combination we studied (; Supplemental Table S3), consistent with other recent findings [Citation68]. In fact, all the synaptic suggestive GWAS candidate genes identified for ASD overlapped with those identified for schizophrenia. These included well-known synaptic genes, such as those encoding for the scaffold-protein ANKG (ANK3), the ionotropic glutamate receptor GluN2a (GRIN2A), and Ras GTPase activating protein 1 (SYNGAP1).

Figure 3. Overlap between genes associated with neurodevelopmental disorders and neurodegenerative diseases in humans based on genome-wide association study (GWAS) findings focusing on those overlapping with the GO “synapse”. Venn diagram visualizing overlap between synaptic suggestive candidate genes associated with the selected neurodevelopmental disorders and neurodegenerative diseases. The synaptic suggestive candidate genes, APOE and MAPT were associated with both Alzheimer’s and Parkinson’s disease, ADAM10 and PTK2B with both Alzheimer’s disease and schizophrenia, DLG2 and FYN with both Parkinson’s disease and schizophrenia, CLU and MEF2C with Alzheimer’s disease, schizophrenia and ASD, and IGSF9B with Parkinson’s disease, schizophrenia and ASD. See table S3 for full lists of genes.

Figure 3. Overlap between genes associated with neurodevelopmental disorders and neurodegenerative diseases in humans based on genome-wide association study (GWAS) findings focusing on those overlapping with the GO “synapse”. Venn diagram visualizing overlap between synaptic suggestive candidate genes associated with the selected neurodevelopmental disorders and neurodegenerative diseases. The synaptic suggestive candidate genes, APOE and MAPT were associated with both Alzheimer’s and Parkinson’s disease, ADAM10 and PTK2B with both Alzheimer’s disease and schizophrenia, DLG2 and FYN with both Parkinson’s disease and schizophrenia, CLU and MEF2C with Alzheimer’s disease, schizophrenia and ASD, and IGSF9B with Parkinson’s disease, schizophrenia and ASD. See table S3 for full lists of genes.

Comparison of the synaptic suggestive GWAS candidate genes across other combinations of diseases and disorders (Table S2) revealed the following overlapping findings: (i) apolipoprotein E (APOE) and microtubule associated protein tau (MAPT) for Parkinson’s disease and Alzheimer’s disease, (ii) ADAM metallopeptidase domain 10 (ADAM10) and protein tyrosine kinase 2 beta (PTK2B) for Alzheimer’s disease and schizophrenia, (iii) clusterin (CLU) and myocyte enhancer factor 2C (MEF2C) for Alzheimer’s disease, schizophrenia, and ASD, (iv) discs large MAGUK scaffold protein 2 (DLG2) and FYN proto-oncogene, Src family tyrosine kinase (FYN) for Parkinson’s disease and schizophrenia, and (v) immunoglobulin superfamily member 9B (IGSF9B) for Parkinson’s disease, schizophrenia & ASD (; Supplemental Table S3). Based on the STRING database (v11.5), the majority of these genes are directly or indirectly connected either through PPIs (identified from experimental/biochemical data or reported in curated databases) or co-mentioning in PubMed abstracts, indicating involvement in similar biological mechanisms. PANX1 is connected to this STRING network via the proto-oncogene tyrosine-protein kinase (Src), which regulates GLUN2A and GLUN2B receptors and synaptic metaplasticity [Citation69] and mediates PANX1-NMDA receptor crosstalk in pathophysiological synaptic plasticity [Citation70,Citation71] through physical interaction [Citation70]. Given that PANX1 is connected to several of these neurological conditions and has been reported to be enriched at post-synaptic membranes, we characterized the neural PANX1 interactome (originally identified by our lab [Citation11,Citation13]) by performing PANTHER pathway and PPI analyses to expand our understanding of its biological roles.

Bioinformatics analyses of the PANX1 interactome revealed involvement of genes associated with cell structure regulation, proteostasis, neurodegeneration, and synaptic enrichment

In addition to revealing possible biological roles of the PANX1 interactome, we also conducted pathway and PPI analyses to better understand (i) potential implications in disease and (ii) interactions between the PANX1-interacting proteins, based on existing knowledge. PANTHER enrichment analysis of the 483 Mus musculus PANX1-interacting proteins (unique UniProt accession numbers) revealed enrichment for six PANTHER pathways, namely the ubiquitin proteasome pathway (P00060, p-value = 3.65E–04), Parkinson’s disease (P00049, p-value = 1.05E–03), integrin signaling pathway (P00034, p-value = 1.61E–03), nicotinic acetylcholine receptor signaling pathway (P00044, p-value = 3.93E–03), inflammation mediated by chemokine and cytokine signaling pathway (P00031, p-value = 1.93E–02) and Huntington disease (P00029, p-value = 1.99E–02). An overview of the PANX1-interacting proteins annotated to the Parkinson’s disease PANTHER pathway can be found in .

Table 1. Mus musculus PANX1-interacting proteins annotated to the Parkinson disease PANTHER pathway (P00049), other PANTHER pathways, and PANTHER protein classes.

As PANX1 is involved in a wide range of synaptic functions, we decided to identify synaptic PANX1-interacting proteins (~18% of the neural PANX1 interactome) via cross-analysis with the Mus musculus cellular component GO term “synapse.” Using the 89 identified synaptic PANX1-interacting proteins as input, we created a STRING PPI network formed by 66 synaptic PANX1-interacting proteins () and identified 4 clusters involved in gene expression and translation, cytoskeleton organization, vesicle-mediated transport, and cell communication and its regulation, respectively (). When conducting the cross-analysis with both the Mus musculus and Homo sapiens synapse GO term (combined), 92 synaptic PANX1-interacting proteins were identified (see for an overview of these proteins).

Figure 4. STRING protein–protein interaction (PPI) network to explore the synaptic Mus musculus PANX1 interactome and to identify clusters (potential functions presented in ). A total of 66 synaptic PANX1-interacting proteins formed the PPI network (~74% of the synaptic Mus musculus PANX1 interactome), based on our chosen methodology. The thickness of the edges in the PPI network indicates the strength of data support and dashed line edges reveal PPIs between clusters.

Figure 4. STRING protein–protein interaction (PPI) network to explore the synaptic Mus musculus PANX1 interactome and to identify clusters (potential functions presented in Table 2). A total of 66 synaptic PANX1-interacting proteins formed the PPI network (~74% of the synaptic Mus musculus PANX1 interactome), based on our chosen methodology. The thickness of the edges in the PPI network indicates the strength of data support and dashed line edges reveal PPIs between clusters.

Table 2. Synaptic Mus musculus PANX1-interacting proteins forming the STRING protein–protein interaction network clusters annotated to selected PANTHER protein classes and gene ontology (GO) biological process (BP) terms.

Table 3. Overview of Mus musculus synaptic PANX1-interacting proteins.

Cross-analyses revealed overlap between the full neural and synapse-specific PANX1 interactome and neurological disorder/disease susceptibility genes

To explore connections between PANX1 and the four disorders/diseases of interest, we compared the entire PANX1 interactome with the suggestive candidate genes for each neurological condition (). Overall, the two neurodevelopmental disorder susceptibility gene sets exhibited a relatively larger degree of overlap with the PANX1 interactome than did the neurodegenerative disease susceptibility gene sets. Twenty schizophrenia susceptibility genes overlapped with the PANX1 interactome, which was the largest number in common, while ASD had 12 genes in common with the PANX1 interactome (see for an overview). Several of these genes are involved in protein folding (GO:0006457; Hspa1b, Hspa1l, Hspd1, Hspe1, and St13) or regulation of translation (GO:0006417; Cnot1, Ddx39b, Etf1, Lsm1, and Mapk3).

Table 4. Genes encoding for PANX1-interacting proteins that have also been identified as suggestive GWAS candidate genes ().

Since synaptic dysfunction has been associated with neurological conditions such as Parkinson’s disease [Citation74], to identify synaptic-specific associations, we compared the (Mus musculus and Homo sapiens synapse GO term-defined) synaptic PANX1 interactome with (Mus musculus and Homo sapiens synapse GO term-defined) synaptic suggestive GWAS candidate genes ( and Table S2). Based on our methodological workflow (e.g., restricted to genes reported more than once in the GWAS catalog and genes annotated to the Mus musculus and Homo sapiens synapse GO term), we identified the gene coding for N-ethylmaleimide sensitive factor, vesicle-fusing ATPase (NSF) as both a suggestive candidate gene for Parkinson’s disease and a PANX1-interacting protein. It is important to note that the GO definition of “synapse” does not include several important synaptic proteins, as further elaborated in the discussion.

Therefore, to circumvent the limitations of the restricted GO definition of “synapse” genes in our exploration of PANX1-related disease connections of a synaptic nature, we made a broader comparison between GO terms enriched within the synaptic PANX1 interactome and GO terms enriched for the four neurological conditions (based on synaptic disorder/disease susceptibility genes; ). We included terms associated with the GO domains “cellular component” and “biological process” (choosing domains fitting the aim of this study), as these two relatively broader GO domains could help provide insight into common pathophysiological mechanisms and potential PANX1 involvement. The “cellular component” domain was selected to explore localization to structures associated with synapses, and/or structures influencing synapses and their development or stability (e.g., “somatodendritic tree” should have some bearing on dendritic spines, “cytoplasmic vesicle” would be associated with transport processes required to bring cargo to nascent spines and so on). The following GO terms were enriched for the synaptic PANX1 interactome and each of the neurological conditions: Asymmetric synapse (GO:0032279), glutamatergic synapse (GO:0098978), neuron to neuron synapse (GO:0098984), postsynapse (GO:0098794), postsynaptic density (GO:0014069), and postsynaptic specialization (GO:0099572; ).

Table 5. Enrichment analysis (GO terms within the “cellular component” and “biological process” GO domains) for the synaptic PANX1 interactome showing GO terms also found for the investigated neurological conditions (as indicated with X).

Focusing on the GO “biological process”, two GO terms overlapped (i.e., cell junction organization (GO:0034330) and regulation of vesicle-mediated transport (GO:0060627)) when comparing the synaptic PANX1 interactome and the neurological conditions ().

Specific human brain regions associated with ASD, schizophrenia, Parkinson’s disease, and Alzheimer’s disease

Since the neurological conditions we investigated are well known to exhibit spatiotemporal specificity in terms of progression in affected brain regions (for example, Parkinson’s disease initially presents with striatal dysfunction), we investigated regional specificity in elevated transcript expression for the synaptic PANX1 interactome and synaptic GWAS candidate genes by means of cross-analysis. Note that a gene was considered as “expressed” in a given brain region when the normalized transcript per million (nTPM; i.e. expression value) was above 1. Using the Human Protein Atlas v22.0 (proteinatlas.org) [Citation50,Citation51], we found three of the synaptic PANX1-interactors exhibited regionally elevated transcript expression: RPL9 in cerebral cortex, SLC18A2 in pons, midbrain, and hypothalamus, and TANC2 in cerebral cortex, hippocampal formation, and amygdala (; based on dataset on summarized expression in main brain regions not the specific expression in the more than 200 regions, areas, and subfields, separately). Regionally elevated expression of synaptic neurological disorder/disease susceptibility genes was also found, with the highest number of genes noted for schizophrenia (16) and ASD (11; ). Most of the disorder/disease-associated genes were elevated in the cerebral cortex, cerebellum, hippocampal formation, and/or amygdala ().

Table 6. Overview of the overlap between the elevated genes in specific brain regions (Human Protein Atlas) and the synaptic PANX1 interactome/suggestive neurological disorder/disease susceptibility genes.

Discussion

The goal of this study was to identify common synaptic genes and molecular pathways amongst neurological conditions and PANX1 using bioinformatics approaches. Using large-scale bioinformatics approaches has many advantages including but not limited to the: (i) ability to efficiently analyze a large amount of data, (ii) approaching analysis beyond tradition (e.g., data integration to get new insights), and (iii) possibility of making novel discoveries. Our study revealed multiple potential links between the PANX1 interactome and the investigated neurological conditions that now warrant validation. Overall, the two investigated neurodevelopmental disorders exhibited the largest overlap in synaptic susceptibility genes and were more abundantly represented in the PANX1 interactome. Our results suggest that the molecular mechanisms underlying synaptic dysfunction in neurodevelopmental disorders may be more closely linked to one another and to PANX1 than is the case for neurodegenerative diseases, although it should be noted that this larger overlap also may be influenced by the size of these gene sets compared with those extracted for the investigated neurogenerative diseases. While the research on PANX1 involvement in neurological conditions has primarily focused on neurodegenerative diseases, it is perhaps not surprising that the PANX1 interactome is linked to neurodevelopmental disorder molecular players, given expanding evidence for overlap between molecular mechanisms underlying neurodevelopmental disorders and neurodegenerative diseases, particularly at the synapse and the role for PANX1 in regulation of synapse development and plasticity [Citation19–24,Citation75]. There is mounting evidence that synaptic dysfunction associated with neurodegenerative diseases can be detected at early pre-symptomatic stages, and there are growing links in neurodevelopmental and neurodegenerative gene expression changes and genetic risk susceptibilities [Citation76–86].

Further evidence for common genetic elements of ASD and schizophrenia

Notably, several of the susceptibility genes common to ASD and schizophrenia in our analysis have previously been linked to both disorders [Citation87,Citation88] and play key roles at the synapse (reviewed in [Citation1,Citation3,Citation4]). For example, ANK3 (ANKG protein), GRIN2A (GluN2A), and SYNGAP1 have established synaptic roles [Citation89–92] and connections to ASD, schizophrenia, and other co-morbid neurodevelopmental disorders like epilepsy [Citation93–98]. Although GluN2A was not identified in our PANX1 interactome, we identified the closely related protein GluN2D (GRIN2D). PANX1 has been previously reported to form a complex with GluN1 (obligatory NMDA receptor subunit) and Src [Citation70] in juvenile rat brain hippocampal slices. For our interaction screen, we used the N2a mouse cell line derived from a neuroblastoma (neural crest in origin) that is widely used to study cell biological aspects of neuronal differentiation [Citation99]. The relatively immature state of the undifferentiated N2a cell model and the high stringency of our immunoprecipitation conditions (RIPA buffer which contains harsh detergents and preserves only robust interactions) could partially account for the lack of identification of GluN1 as a PANX1 interactor. It is surprising that we would detect GluN2D and not GluN1, but perhaps GluN2D plays an early non-synaptic, non-receptor role that has yet to be identified; this may be consistent with its decrease in levels with increasing age/development [Citation100]. Conversely, the lipid raft scaffolding protein, flotillin, was common to ASD and schizophrenia but has not been extensively studied in either neurodevelopmental disorder. Given lipid raft/cholesterol dysfunction has been implicated in the monogenic common ASD Fragile X syndrome and has been linked to glutamatergic synapse formation [Citation101]; further exploration of the role of flotillin in synaptic dysfunction in ASD and schizophrenia could be informative. Further analysis is now warranted to explore the shared synaptic genetic susceptibility between ASD and schizophrenia, including individual variability in risk, onset, severity, and progression. Some studies have looked into this shared genetic basis. For instance, significant genetic correlations have been identified between ASD and schizophrenia (but not between Alzheimer’s disease and Parkinson’s disease) [Citation68], similar to what we observed in this study. Even though ASD and schizophrenia share genetic risks, they seem to have distinct developmental profiles [Citation102]; multi-omics studies could help tease out the underpinnings.

The established synaptic roles of these neurological disorder/disease susceptibility genes and our recent discovery of PANX1 regulation of dendritic spine stability prompted our synaptic PANX1 interactome STRING analysis, which identified clusters in gene expression and translation, cytoskeleton organization, vesicle-mediated transport, and cell communication and its regulation (). Dysregulation in these same cellular processes are observed in ASD [Citation103,Citation104], schizophrenia [Citation105,Citation106], Parkinson’s disease [Citation107,Citation108], and Alzheimer’s disease [Citation109,Citation110], and PANX1 has been linked to these conditions (reviewed in [Citation75,Citation111]). Of particular note, there are >40 non-coding PANX1 variants in the VariCarta ASD database (https://varicarta.msl.ubc.ca/index [Citation112]), and there are several ASD-linked PANX1 SNPs associated with brain-specific PANX1 gene expression changes [Citation113]. Our cross-analysis of PANX1 interactors, synaptic-expressed genes, and genetic risk susceptibility to neurological conditions is a key step in bridging our gaps in understanding how PANX1 is linked to these various conditions.

Given the key synaptic findings from our broader analyses, we thought it prudent to refine our approach with the GO synaptic gene set; however, the results were somewhat surprising. Despite the strong representation of well-established and important synaptic genes associated with neurological conditions in the PANX1 set (refer to , e.g. Dpysl2, Actr3, Hip1, several Rabs etc.), when we restricted our disease-associated analysis to “GO synaptic” genes, we only identified the vesicular trafficking regulator NSF as the sole common gene shared by PANX1 and a neurological disease susceptibility gene set (specifically for Parkinson’s disease). Given that studies in Drosophila have shown that expression of a dominant-negative form of Nsf2 leads to disrupted neuromuscular junction and synaptic structural development [Citation114,Citation115] and that this is linked to actin-cytoskeleton regulation [Citation116], this finding suggests that investigating the NSF-PANX1 interaction could be of key interest in terms of exploring a putative role for PANX1 in synaptic dysfunction in Parkinson’s disease [Citation117]. The connection to Parkinson’s disease is further bolstered by our PANTHER pathway analysis, which identified Parkinson’s disease as one of six pathways exhibiting enrichment within the PANX1 interactome. Importantly, the limited output of the GO synaptic analysis (one gene identified) underscores a key caveat of bioinformatics and GO, in that many key connections may be missed as the outputs are limited by the inputs (a gene may not have met overly restrictive inclusion criteria or certain literature has been missed). Given this limitation, we broadened our approach to compare enriched synapse-specific GO terms (). In accordance with casting a wide net, we focused on cellular component locations (“cellular component” domain) and biological processes (“biological process” domain), which are relatively broader than the third GO domain, “molecular function”. We identified similar neurological condition-associated cellular components/processes as those identified in our STRING analysis of PANX1 PPIs, such as “vesicle-mediated transport”. These similarities provide potential avenues of insight into the connection between PANX1 and various neurological conditions, as discussed above, such as the link between vesicle-mediated transport genes and Parkinson’s disease risk. These potential links between PANX1, vesicle transport, and Parkinson’s disease may be worth pursuing given the cell-to-cell propagation of alpha-synuclein fibrils, implicated in Parkinson’s disease, involves macropinocytosis [Citation118], a process in which PANX1 has been implicated [Citation119–121]. Further dissection of brain region-specific expression of PANX1 interactors in relevant animal models could also provide important clues.

Limitations of GWAS and GO analysis can obscure key synaptic PPIs

GWAS exhibits several key caveats. Importantly, GWAS was historically somewhat restricted to European populations, thereby limiting their broader use and application [Citation122]; importantly, some recent progress has been made in expanding GWAS to more groups, especially Asian populations (see for details of GWASs used in the current study). Additionally, although GWAS identify genetic differences associated with disease (and beyond), these are not necessarily causal, and where mechanistic links might be present, these can be challenging and complicated to unravel [Citation123]. In terms of the analysis of synaptic representation within suggestive candidate genes for neurological conditions, there are several types of connections that can be missed, as briefly discussed above. For example, given the limitations of gene ontologies [Citation124], not all proteins that could be present at synapses during their lifespan may have their genes annotated to GO “synapse” – for example, PANX1 itself is not found in GO “synapse”, despite its described localization and functional characterization at the post-synapse. Similarly, a recently created synaptic gene ontology database (https://www.syngoportal.org/) does not contain PANX1. Furthermore, identification of a connection does not imply causality; this requires careful and in-depth follow-up of cell biology studies amongst other analyses. Additionally, the analysis is limited to gene level associations with disease and does not account for disorder/disease-associated differences in expression levels, post-translational modifications or protein-targeting pathophysiological mechanisms, like autoantibody production, all of which have been described for CRMP2 (e.g., changes in protein expression levels in several neurological conditions [Citation125], hyperphosphorylation in Alzheimer’s disease [Citation126–128], and auto-antibodies in autism spectrum disorders [Citation129]). Finally, these genetic approaches cannot account for the influence of environmental factors such as inflammation due to injury or infection, which would be expected to have a major impact on PANX1 function and regulation [Citation111,Citation130–132], including neuronal PANX1. For example, the inflammatory mediator TNF-α increases PANX1 expression and surface localization in human umbilical vein endothelial cells, resulting in increased intracellular Ca2+ and release of the pro-inflammatory cytokine IL-β1 [Citation133]. If neuronal PANX1 levels are similarly upregulated by inflammatory mediators, this could inhibit morphological plasticity mechanisms like neurite outgrowth [Citation11,Citation12] and contribute to dendritic spine instability [Citation22–24]. Supporting this, recent work suggests that spine and synaptic plasticity deficits in a mouse model of Alzheimer’s disease can be mitigated by blocking PANX1 [Citation26]. In a similar fashion, PANX1 could be involved in impaired cognitive function associated with a wide variety of diseases and conditions in which heightened peripheral inflammation leads to neuroinflammation (reviewed in [Citation134]), often associated with increases in TNF-α, and as such, could be a potential therapeutic target.

Table 7. Overview of the specified study populations in the genome-wide association studies (GWASs) extracted from the GWAS catalog for each neurological condition (prior to applying filtering criteria).

Where do we go from here?

To fully appreciate both the complexity and validity of putative pathophysiological mechanisms hinted at by the analyses presented here, additional work in rodent and human iPSC-derived models of neuropsychiatric conditions would be a logical next step. For example, while we have previously validated interactions of PANX1 with ARP3 and CRMP2 in cell lines, investigating their interplay within living neurons and brain will help to shed light on how this and related PPI networks intersect in synapse development in health and disease states. Finally, we currently have limited insight into how PANX1 protein interactions could be involved in or regulate channel function, or how changes in ion and/or metabolite fluxes may impact the interactions or the function of the interacting proteins. Disentangling the impact of interacting proteins on PANX1 properties like channel function (trafficking, post-translational modifications), and vice versa, as well as PANX1 channel-independent signaling (i.e., scaffold or other functions) will be critical for fulsome understanding of the role of PANX1 in brain health and disease.

Supplemental material

Supplemental Material

Download MS Excel (56.5 KB)

Acknowledgments

Additionally, the authors are grateful to Juan C. Sanchez-Arias for his initial input on the study design and discussion for the first bioRxiv preprint version of this work (posted October 11, 2019). LAS and SDF designed the study with inputs from LEWS. LEWS identified the PANX1 interactome in mouse N2a cells, SDF performed the bioinformatics analyses, and SDF, LEWS, and LAS wrote the manuscript. All authors approved the final manuscript. A preprint of this manuscript (prior to final peer review edits) can be found on bioRxiv: https://www.biorxiv.org/content/10.1101/801563v3.

Disclosure statement

No potential conflict of interest was reported by the authors.

Data availability statement

Information about the publicly available data used in this study is outlined throughout the manuscript and supplemental tables. For the proteomics data, RAW files are deposited in the Center for Computational Mass Spectrometry (MassIVE MSV000093036). Please contact Dr. Leigh Anne Swayne ([email protected]) for additional information, if required. The R code is available and can be obtained from Dr. Simona Denise Frederiksen, upon reasonable request.

Supplementary material

Supplemental data for this article can be accessed online at https://doi.org/10.1080/19336950.2023.2253102

Additional information

Funding

This project was supported by operating grants from the Canadian Institutes of Health Research [MOP142215], the Natural Sciences and Engineering Research Council of Canada [NSERC; 402270-2011], and the University of Victoria Division of Medical Sciences to LAS. LAS was also supported by a Michael Smith Foundation for Health Research and British Columbia Schizophrenia Society Foundation Scholar Award [5900]. LEWS was supported by a Vanier Canada Graduate Scholarship (NSERC).

References

  • Forrest MP, Parnell E, Penzes P. Dendritic structural plasticity and neuropsychiatric disease. Nat Rev Neurosci. 2018;19(4):215–21. doi: 10.1038/nrn.2018.16
  • Maiti P, Manna J, Ilavazhagan G, et al. Molecular regulation of dendritic spine dynamics and their potential impact on synaptic plasticity and neurological diseases. Neurosci Biobehav Rev. 2015;59:208–237. doi: 10.1016/j.neubiorev.2015.09.020
  • Penzes P, Cahill ME, Jones KA, et al. Dendritic spine pathology in neuropsychiatric disorders. Nat Neurosci. 2011;14(3):285–293. doi: 10.1038/nn.2741
  • Nishiyama J. Plasticity of dendritic spines: molecular function and dysfunction in neurodevelopmental disorders. Psychiatry Clin Neurosci. 2019;73(9):541–550. doi: 10.1111/pcn.12899
  • Phillips M, Pozzo-Miller L. Dendritic spine dysgenesis in autism related disorders. Neurosci Lett. 2015;601:30–40. doi: 10.1016/j.neulet.2015.01.011
  • Zhang K, Liao P, Wen J, et al. Synaptic plasticity in schizophrenia pathophysiology. IBRO Neurosci Reports. 2022;13:478–487.
  • Herms J, Dorostkar MM. Dendritic spine pathology in neurodegenerative diseases. Annu Rev Pathol. 2016;11(1):221–250. doi: 10.1146/annurev-pathol-012615-044216
  • Villalba RM, Smith Y. Loss and remodeling of striatal dendritic spines in Parkinson’s disease: from homeostasis to maladaptive plasticity? J Neural Transm. 2018;125(3):431–447. doi: 10.1007/s00702-017-1735-6
  • Zoidl G, Petrasch-Parwez E, Ray A, et al. Localization of the pannexin1 protein at postsynaptic sites in the cerebral cortex and hippocampus. Neuroscience. 2007;146(1):9–16. doi: 10.1016/j.neuroscience.2007.01.061
  • Boyce AKJ, Epp AL, Nagarajan A, et al. Transcriptional and post-translational regulation of pannexins. Biochim Biophys Acta Biomembr. 2018;1860(1):72–82. doi: 10.1016/j.bbamem.2017.03.004
  • Wicki-Stordeur LE, Swayne LA. Panx1 regulates neural stem and progenitor cell behaviours associated with cytoskeletal dynamics and interacts with multiple cytoskeletal elements. Cell Commun Signal. 2013;11(1):62. doi: 10.1186/1478-811X-11-62
  • Xu X, Wicki-Stordeur LE, Sanchez-Arias JC, et al. Probenecid disrupts a novel pannexin 1-Collapsin response mediator protein 2 interaction and increases microtubule stability. Front Cell Neurosci. 2018;12:124. doi: 10.3389/fncel.2018.00124
  • Wicki-Stordeur LE. Pannexin 1 regulates ventricular zone neuronal development. [ PhD Thesis], Division Of Medical Sciences (University Of Victoria); 2015.
  • Boycott KM, Rath A, Chong JX, et al. International cooperation to enable the diagnosis of all Rare genetic diseases. Am J Hum Genet. 2017;100(5):695–705. doi: 10.1016/j.ajhg.2017.04.003
  • Zhang H, Kang E, Wang Y, et al. Brain-specific Crmp2 deletion leads to neuronal development deficits and behavioural impairments in mice. Nat Commun. 2016;7(1). doi: 10.1038/ncomms11773
  • Ziak J, Weissova R, Jeřábková K, et al. CRMP2 mediates Sema3F-dependent axon pruning and dendritic spine remodeling. EMBO Rep. 2020;21(3):e48512. doi: 10.15252/embr.201948512
  • Spence EF, Kanak DJ, Carlson BR, et al. The Arp2/3 complex is essential for distinct stages of spine synapse maturation, including synapse unsilencing. J Neurosci Off J Soc Neurosci. 2016;36(37):9696–9709. doi: 10.1523/JNEUROSCI.0876-16.2016
  • Kim IH, Racz B, Wang H, et al. Disruption of Arp2/3 results in asymmetric structural plasticity of dendritic spines and progressive synaptic and behavioral abnormalities. J Neurosci Off J Soc Neurosci. 2013;33(14):6081–6092. doi: 10.1523/JNEUROSCI.0035-13.2013
  • Ardiles AO, Flores-Muñoz C, Toro-Ayala G, et al. Pannexin 1 regulates bidirectional hippocampal synaptic plasticity in adult mice. Front Cell Neurosci. 2014;8:326. doi: 10.3389/fncel.2014.00326
  • Gajardo I, Salazar CS, Lopez-Espíndola D, et al. Lack of pannexin 1 alters synaptic GluN2 subunit composition and spatial reversal learning in mice. Front Mol Neurosci. 2018;11:114. doi: 10.3389/fnmol.2018.00114
  • Prochnow N, Abdulazim A, Kurtenbach S, et al. Pannexin1 stabilizes synaptic plasticity and is needed for learning. PLoS One. 2012;7(12):e51767. doi: 10.1371/journal.pone.0051767
  • Sanchez-Arias JC, Liu M, Choi CSW, et al. Pannexin 1 regulates network ensembles and dendritic spine development in cortical neurons. eNeuro. 2019;6(3):6. doi: 10.1523/ENEURO.0503-18.2019
  • Sanchez-Arias JC, Candlish RC, van der Slagt E, et al. Pannexin 1 regulates dendritic protrusion dynamics in immature cortical neurons. eNeuro. 2020;7(4):ENEURO.0079–20.2020. doi: 10.1523/ENEURO.0079-20.2020
  • Flores-Muñoz C, García-Rojas F, Pérez MA, et al. The long-term pannexin 1 ablation produces structural and functional modifications in hippocampal neurons. Cells. 2022;11(22):11. doi: 10.3390/cells11223646
  • Liu Y, Qu H-Q, Chang X, et al. Mutation burden analysis of six common mental disorders in African Americans by whole genome sequencing. Hum Mol Genet. 2022;31(22):3769–3776. doi: 10.1093/hmg/ddac129
  • Flores-Muñoz C, Gómez B, Mery E, et al. Acute pannexin 1 blockade mitigates early synaptic plasticity defects in a mouse model of Alzheimer’s disease. Front Cell Neurosci. 2020;14:46. doi: 10.3389/fncel.2020.00046
  • Ni M, He J-G, Zhou H-Y, et al. Pannexin-1 channel dysfunction in the medial prefrontal cortex mediates depressive-like behaviors induced by chronic social defeat stress and administration of mefloquine in mice. Neuropharmacology. 2018;137:256–267. doi: 10.1016/j.neuropharm.2017.12.004
  • Lee H, Joo J, Nah S-S, et al. Changes in Dpysl2 expression are associated with prenatally stressed rat offspring and susceptibility to schizophrenia in humans. Int J Mol Med. 2015;35(6):1574–1586. doi: 10.3892/ijmm.2015.2161
  • Liu Y, Pham X, Zhang L, et al. Functional variants in DPYSL2 sequence increase risk of schizophrenia and suggest a link to mTOR signaling. G3 (Bethesda). 2014;5(1):61–72. doi: 10.1534/g3.114.015636
  • Datta D, Arion D, Roman KM, et al. Altered expression of ARP2/3 complex signaling pathway genes in prefrontal layer 3 pyramidal cells in schizophrenia. Am J Psychiatry. 2017;174(2):163–171. doi: 10.1176/appi.ajp.2016.16020204
  • Haan N, Westacott LJ, Carter J, et al. Haploinsufficiency of the schizophrenia and autism risk gene Cyfip1 causes abnormal postnatal hippocampal neurogenesis through microglial and Arp2/3 mediated actin dependent mechanisms. Transl Psychiatry. 2021;11(1):313. doi: 10.1038/s41398-021-01415-6
  • Rylaarsdam L, Guemez-Gamboa A. Genetic causes and modifiers of autism spectrum disorder. Front Cell Neurosci. 2019;13:385. doi: 10.3389/fncel.2019.00385
  • Ikeda M, Takahashi A, Kamatani Y, et al. Genome-wide association study detected novel susceptibility genes for schizophrenia and shared trans-populations/diseases genetic effect. Schizophr Bull. 2019;45(4):824–834. doi: 10.1093/schbul/sby140
  • Redenšek S, Trošt M, Dolžan V. Genetic determinants of parkinson’s disease: can they help to stratify the patients based on the underlying molecular defect? Front Aging Neurosci. 2017;9:20. doi: 10.3389/fnagi.2017.00020
  • Neuner SM, Tcw J, Goate AM. Genetic architecture of Alzheimer’s disease. Neurobiol Dis. 2020;143:104976. doi: 10.1016/j.nbd.2020.104976
  • Bettens K, Sleegers K, Van Broeckhoven C. Genetic insights in Alzheimer’s disease. Lancet Neurol. 2013;12(1):92–104. doi: 10.1016/S1474-4422(12)70259-4
  • Ding C, Wu Y, Chen X, et al. Global, regional, and national burden and attributable risk factors of neurological disorders: the Global burden of disease study 1990-2019. Front Public Health. 2022;10:952161. doi: 10.3389/fpubh.2022.952161
  • GBD. Global, regional, and national burden of 12 mental disorders in 204 countries and territories, 1990–2019: a systematic analysis for the Global burden of disease study 2019. Lancet Psychiatry. 2022;9(2):137–150. doi: 10.1016/S2215-0366(21)00395-3
  • Lavelle TA, Weinstein MC, Newhouse JP, et al. Economic burden of childhood autism spectrum disorders. Pediatrics. 2014;133(3):e520–529. doi: 10.1542/peds.2013-0763
  • Chong HY, Teoh SL, DB-C W, et al. Global economic burden of schizophrenia: a systematic review. Neuropsychiatr Dis Treat. 2016;12:357–373. doi: 10.2147/NDT.S96649
  • Buniello A, MacArthur JAL, Cerezo M, et al. The NHGRI-EBI GWAS catalog of published genome-wide association studies, targeted arrays and summary statistics 2019. Nucleic Acids Res. 2019;47(D1):D1005–D1012. doi: 10.1093/nar/gky1120
  • Mi H, Muruganujan A, Huang X, et al. Protocol update for large-scale genome and gene function analysis with the PANTHER classification system (v.14.0). Nat Protoc. 2019;14(3):703–721. doi: 10.1038/s41596-019-0128-8
  • Mi H, Ebert D, Muruganujan A, et al. PANTHER version 16: a revised family classification, tree-based classification tool, enhancer regions and extensive API. Nucleic Acids Res [Internet]. 2020;49:D394–D403. doi: 10.1093/nar/gkaa1106
  • Mudunuri U, Che A, Yi M, et al. bioDbnet: the biological database network. Bioinformatics [Internet]. 2009;25(4):555–556. doi: 10.1093/bioinformatics/btn654
  • Seal RL, Braschi B, Gray K, et al. Genenames.Org: the HGNC resources in 2023. Nucleic Acids Res [Internet. 2023;51:D1003–1009. doi: 10.1093/nar/gkac888
  • Bult CJ, Blake JA, Smith CL, et al. Mouse genome database (MGD) 2019. Nucleic Acids Res. 2019;47:D801–806. doi: 10.1093/nar/gky1056
  • Blake JA, Baldarelli R, Kadin JA, et al. Mouse Genome Database (MGD): knowledgebase for mouse-human comparative biology. Nucleic Acids Res. 2021;49:D981–987. doi: 10.1093/nar/gkaa1083
  • Mi H, Ebert D, Muruganujan A, et al. PANTHER version 16: a revised family classification, tree-based classification tool, enhancer regions and extensive API. Nucleic Acids Res [Internet]. 2021;49:D394–403. doi: 10.1093/nar/gkaa1106.
  • Thomas PD, Ebert D, Muruganujan A, et al. PANTHER: making genome-scale phylogenetics accessible to all. Protein Sci. 2022;31(1):8–22. doi: 10.1002/pro.4218
  • Sjöstedt E, Zhong W, Fagerberg L, et al. An atlas of the protein-coding genes in the human, pig, and mouse brain. Science. 2020;367(6482). doi: 10.1126/science.aay5947
  • Uhlen M, Oksvold P, Fagerberg L, et al. Towards a knowledge-based human protein Atlas. Nat Biotechnol. 2010;28(12):1248–1250. doi: 10.1038/nbt1210-1248
  • Szklarczyk D, Gable AL, Lyon D, et al. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2019;47:D607–613. doi: 10.1093/nar/gky1131
  • Szklarczyk D, Gable AL, Nastou KC, et al. The STRING database in 2021: customizable protein-protein networks, and functional characterization of user-uploaded gene/measurement sets. Nucleic Acids Res. 2021;49:D605–612. doi: 10.1093/nar/gkaa1074
  • Szklarczyk D, Kirsch R, Koutrouli M, et al. The STRING database in 2023: protein-protein association networks and functional enrichment analyses for any sequenced genome of interest. Nucleic Acids Res. 2023;51:D638–646. doi: 10.1093/nar/gkac1000
  • Malone J, Holloway E, Adamusiak T, et al. Modeling sample variables with an experimental factor ontology. Bioinformatics. 2010;26(8):1112–1118. doi: 10.1093/bioinformatics/btq099
  • Sollis E, Mosaku A, Abid A, et al. The NHGRI-EBI GWAS catalog: knowledgebase and deposition resource. Nucleic Acids Res. 2023;51(D1):D977–D985. doi: 10.1093/nar/gkac1010
  • Magno R, Maia A-T, Wren J. Gwasrapidd: an R package to query, download and wrangle GWAS catalog data. Bioinformatics [Internet]. 2020;36(2):649–650. doi: 10.1093/bioinformatics/btz605.
  • Perkins DN, Pappin DJ, Creasy DM, et al. Probability-based protein identification by searching sequence databases using mass spectrometry data. Electrophoresis. 1999;20(18):3551–3567. doi: 10.1002/(SICI)1522-2683(19991201)20:18<3551:AID-ELPS3551>3.0.CO;2-2
  • Brosch M, Yu L, Hubbard T, et al. Accurate and sensitive peptide identification with Mascot Percolator. J Proteome Res. 2009;8(6):3176–3181. doi: 10.1021/pr800982s
  • Ashburner M, Ball CA, Blake JA, et al. Gene ontology: tool for the unification of biology. The gene ontology consortium. Nat Genet. 2000;25(1):25–29. doi: 10.1038/75556
  • Carbon S, Douglass E, Good BM, et al. The gene ontology resource: enriching a GOld mine. Nucleic Acids Res. 2021;49:D325–D334. doi: 10.1093/nar/gkaa1113
  • Mi H, Thomas P. PANTHER pathway: an ontology-based pathway database coupled with data analysis tools. Methods Mol Biol. 2009;563:123–140.
  • Funahashi A, Matsuoka Y, Jouraku A, et al. CellDesigner 3.5: a versatile modeling tool for biochemical networks. Proc IEEE. 2008;96(8):1254–1265. doi: 10.1109/JPROC.2008.925458
  • Szklarczyk D, Morris JH, Cook H, et al. The STRING database in 2017: quality-controlled protein–protein association networks, made broadly accessible. Nucleic Acids Res. 2017;45(D1):D362–D368. doi: 10.1093/nar/gkw937
  • Brohée S, van Helden J, van Helden J. Evaluation of clustering algorithms for protein-protein interaction networks. BMC Bioinf. 2006;7(1):488. doi: 10.1186/1471-2105-7-488
  • Enright AJ, Van Dongen S, Ouzounis CA. An efficient algorithm for large-scale detection of protein families. Nucleic Acids Res. 2002;30(7):1575–1584. doi: 10.1093/nar/30.7.1575
  • Van Dongen S. Graph clustering by flow simulation. [ PhD thesis], Centers for mathematics and computer science (CWI), University of Utr; 2000.
  • Anttila V, Bulik-Sullivan B, Finucane HK, et al. Analysis of shared heritability in common disorders of the brain. Science. 2018;360(6395). doi: 10.1126/science.aap8757
  • Yang K, Trepanier C, Sidhu B, et al. Metaplasticity gated through differential regulation of GluN2A versus GluN2B receptors by src family kinases. EMBO J. 2012;31(4):805–816. doi: 10.1038/emboj.2011.453
  • Weilinger NL, Lohman AW, Rakai BD, et al. Metabotropic NMDA receptor signaling couples src family kinases to pannexin-1 during excitotoxicity. Nat Neurosci. 2016;19(3):432–442. doi: 10.1038/nn.4236
  • Lohman AW, Weilinger NL, Santos SM, et al. Regulation of pannexin channels in the central nervous system by src family kinases. Neurosci Lett. 2019;695:65–70. doi: 10.1016/j.neulet.2017.09.019
  • Liu Y, Deng J, Liu Y, et al. FGF, mechanism of action, role in parkinson’s disease, and therapeutics. Front Pharmacol. 2021;12:675725. doi: 10.3389/fphar.2021.675725
  • Goldman JG, Goetz CG, Berry-Kravis E, et al. Genetic polymorphisms in Parkinson disease subjects with and without hallucinations: an analysis of the cholecystokinin system. Arch Neurol [Internet]. 2004;61(8):1280–1284. doi: 10.1001/archneur.61.8.1280
  • Picconi B, Piccoli G, Calabresi P. Synaptic dysfunction in Parkinson’s disease. Adv Exp Med Biol. 2012;970:553–572.
  • Yeung AK, Patil CS, Jackson MF. Pannexin-1 in the CNS: emerging concepts in health and disease. J Neurochem. 2020;154(5):468–485. doi: 10.1111/jnc.15004
  • Li C, Yang T, Ou R, et al. Overlapping genetic architecture between schizophrenia and neurodegenerative disorders. Front Cell Dev Biol. 2021;9:797072. doi: 10.3389/fcell.2021.797072
  • Reynolds RH, Wagen AZ, Lona-Durazo F, et al. Local genetic correlations exist among neurodegenerative and neuropsychiatric diseases. NPJ Parkinsons Dis. 2023;9(1):70. doi: 10.1038/s41531-023-00504-1
  • Taoufik E, Kouroupi G, Zygogianni O, et al. Synaptic dysfunction in neurodegenerative and neurodevelopmental diseases: an overview of induced pluripotent stem-cell-based disease models. Open Biol. 2018;8(9). doi: 10.1098/rsob.180138
  • Sadeghi I, Gispert JD, Palumbo E, et al. Brain transcriptomic profiling reveals common alterations across neurodegenerative and psychiatric disorders. Comput Struct Biotechnol J. 2022;20:4549–4561. doi: 10.1016/j.csbj.2022.08.037
  • Milnerwood AJ, Gladding CM, Pouladi MA, et al. Early increase in extrasynaptic NMDA receptor signaling and expression contributes to phenotype onset in Huntington’s disease mice. Neuron. 2010;65:178–190. doi: 10.1016/j.neuron.2010.01.008
  • Chambon J, Komal P, Lewitus GM, et al. Early TNF-Dependent regulation of excitatory and inhibitory synapses on striatal direct pathway medium spiny neurons in the YAC128 mouse model of huntington’s disease. J Neurosci Off J Soc Neurosci. 2023;43(4):672–680. doi: 10.1523/JNEUROSCI.1655-22.2022
  • Sha S, Chaigneau T, Krantic S. Pre-symptomatic synaptic dysfunction and longitudinal decay of hippocampal synaptic function in APPPS1 mouse model of Alzheimer’s disease is sex-independent. Brain Res Bull. 2023;198:36–49. doi: 10.1016/j.brainresbull.2023.04.005
  • Litwa K. Shared mechanisms of neural circuit disruption in tuberous sclerosis across lifespan: bridging neurodevelopmental and neurodegenerative pathology. Front Genet. 2022;13:997461. doi: 10.3389/fgene.2022.997461
  • Parnell E, Shapiro LP, Voorn RA, et al. KALRN: acentral regulator of synaptic function and synaptopathies. Gene. 2021;768:145306. doi: 10.1016/j.gene.2020.145306
  • Batool S, Raza H, Zaidi J, et al. Synapse formation: from cellular and molecular mechanisms to neurodevelopmental and neurodegenerative disorders. J Neurophysiol. 2019;121(4):1381–1397. doi: 10.1152/jn.00833.2018
  • Milnerwood AJ, Raymond LA. Early synaptic pathophysiology in neurodegeneration: insights from huntington’s disease. Trends Neurosci. 2010;33(11):513–523. doi: 10.1016/j.tins.2010.08.002
  • Liu X, Li Z, Fan C, et al. Genetics implicate common mechanisms in autism and schizophrenia: synaptic activity and immunity. J Med Genet. 2017;54(8):511.2–520. doi: 10.1136/jmedgenet-2016-104487
  • Ray A, Zoidl G, Weickert S, et al. Site-specific and developmental expression of pannexin1 in the mouse nervous system. Eur J Neurosci. 2005;21(12):3277–3290. doi: 10.1111/j.1460-9568.2005.04139.x
  • Smith KR, Kopeikina KJ, Fawcett-Patel JM, et al. Psychiatric risk factor ANK3/ankyrin-G nanodomains regulate the structure and function of glutamatergic synapses. Neuron. 2014;84(2):399–415. doi: 10.1016/j.neuron.2014.10.010
  • Tseng WC, Jenkins PM, Tanaka M, et al. Giant ankyrin-G stabilizes somatodendritic GABAergic synapses through opposing endocytosis of GABAA receptors. Proc Natl Acad Sci U S A. 2015;112(4):1214–1219. doi: 10.1073/pnas.1417989112
  • Gardoni F, Di Luca M. Protein-protein interactions at the NMDA receptor complex: from synaptic retention to synaptonuclear protein messengers. Neuropharmacology. 2021;190:108551. doi: 10.1016/j.neuropharm.2021.108551
  • Gamache TR, Araki Y, Huganir RL. Twenty years of SynGAP Research: from synapses to cognition. J Neurosci Off J Soc Neurosci. 2020;40(8):1596–1605. doi: 10.1523/JNEUROSCI.0420-19.2020
  • Yoon S, Piguel NH, Penzes P. Roles and mechanisms of ankyrin-G in neuropsychiatric disorders. Exp Mol Med. 2022;54(7):867–877. doi: 10.1038/s12276-022-00798-w
  • Yu Y, Lin Y, Takasaki Y, et al. Rare loss of function mutations in N-methyl-D-aspartate glutamate receptors and their contributions to schizophrenia susceptibility. Transl Psychiatry. 2018;8(1):12. doi: 10.1038/s41398-017-0061-y
  • Mangano GD, Riva A, Fontana A, et al. De Novo GRIN2A variants associated with epilepsy and autism and literature review. Epilepsy Behav. 2022;129:108604. doi: 10.1016/j.yebeh.2022.108604
  • Trifiletti R, Lachman HM, Manusama O, et al. Identification of ultra-rare genetic variants in pediatric acute onset neuropsychiatric syndrome (PANS) by exome and whole genome sequencing. Sci Rep. 2022;12(1):11106. doi: 10.1038/s41598-022-15279-3
  • Singh T, Poterba T, Curtis D, et al. Rare coding variants in ten genes confer substantial risk for schizophrenia. Nature. 2022;604(7906):509–516. doi: 10.1038/s41586-022-04556-w
  • Kilinc M, Creson T, Rojas C, et al. Species-conserved SYNGAP1 phenotypes associated with neurodevelopmental disorders. Mol Cell Neurosci. 2018;91:140–150. doi: 10.1016/j.mcn.2018.03.008
  • Tremblay RG, Sikorska M, Sandhu JK, et al. Differentiation of mouse neuro 2A cells into dopamine neurons. J Neurosci Methods. 2010;186(1):60–67. doi: 10.1016/j.jneumeth.2009.11.004
  • Camp CR, Yuan H. GRIN2D/GluN2D NMDA receptor: unique features and its contribution to pediatric developmental and epileptic encephalopathy. Eur J Paediatr Neurol EJPN Off J Eur Paediatr Neurol Soc. 2020;24:89–99. doi: 10.1016/j.ejpn.2019.12.007
  • Swanwick CC, Shapiro ME, Vicini S, et al. Flotillin-1 promotes formation of glutamatergic synapses in hippocampal neurons. Dev Neurobiol. 2010;70:875–883. doi: 10.1002/dneu.20828
  • St Pourcain B, Robinson EB, Anttila V, et al. ASD and schizophrenia show distinct developmental profiles in common genetic overlap with population-based social communication difficulties. Mol Psychiatry. 2018;23(2):263–270. doi: 10.1038/mp.2016.198
  • Lin Y-C, Frei JA, Kilander MBC, et al. A subset of autism-associated genes regulate the structural stability of neurons. Front Cell Neurosci [Internet]. 2016;10:263. doi: 10.3389/fncel.2016.00263.
  • Ebert DH, Greenberg ME. Activity-dependent neuronal signalling and autism spectrum disorder. Nature. 2013;493(7432):327–337. doi: 10.1038/nature11860
  • McGuire JL, Depasquale EA, Funk AJ, et al. Abnormalities of signal transduction networks in chronic schizophrenia. NPJ Schizophr. 2017;3(1):30. doi: 10.1038/s41537-017-0032-6
  • Cannon TD. How schizophrenia develops: cognitive and brain mechanisms underlying onset of psychosis. Trends Cogn Sci. 2015;19(12):744–756. doi: 10.1016/j.tics.2015.09.009
  • Singh PK, Muqit MMK. Parkinson’s: a disease of aberrant vesicle trafficking. Annu Rev Cell Dev Biol [Internet]. 2020;36(1):237–264. doi: 10.1146/annurev-cellbio-100818-125512.
  • Pellegrini L, Wetzel A, Grannó S, et al. Back to the tubule: microtubule dynamics in Parkinson’s disease. Cell Mol Life Sci. 2017;74(3):409–434. doi: 10.1007/s00018-016-2351-6
  • Bamburg JR, Bloom GS. Cytoskeletal pathologies of Alzheimer disease. Cell Motil Cytoskeleton. 2009;66(8):635–649. doi: 10.1002/cm.20388
  • Martínez-Cué C, Rueda N. Signalling pathways implicated in Alzheimer’s disease neurodegeneration in Individuals with and without down syndrome. Int J Mol Sci. 2020;21(18):21. doi: 10.3390/ijms21186906
  • Sanchez-Arias JC, van der Slagt E, Vecchiarelli HA, et al. Purinergic signaling in nervous system health and disease: focus on pannexin 1. Pharmacol Ther. 2021;225:107840. doi: 10.1016/j.pharmthera.2021.107840
  • Belmadani M, Jacobson M, Holmes N, et al. VariCarta: a comprehensive database of harmonized genomic variants found in autism spectrum disorder sequencing studies. Autism Res Off J Int Soc Autism Res. 2019;12(12):1728–1736. doi: 10.1002/aur.2236
  • Davis LK, Gamazon ER, Kistner-Griffin E, et al. Loci nominally associated with autism from genome-wide analysis show enrichment of brain expression quantitative trait loci but not lymphoblastoid cell line expression quantitative trait loci. Mol Autism. 2012;3(1):3. doi: 10.1186/2040-2392-3-3
  • Stewart BA, Pearce J, Bajec M, et al. Disruption of synaptic development and ultrastructure by drosophila NSF2 alleles. J Comp Neurol. 2005;488(1):101–111. doi: 10.1002/cne.20603
  • Stewart BA, Mohtashami M, Rivlin P, et al. Dominant-negative NSF2 disrupts the structure and function of drosophila neuromuscular synapses. J Neurobiol. 2002;51(4):261–271. doi: 10.1002/neu.10059
  • Peyre J-B, Seabrooke S, Randlett O, et al. Interaction of cytoskeleton genes with NSF2-induced neuromuscular junction overgrowth. Genesis. 2006;44(12):595–600. doi: 10.1002/dvg.20254
  • Chen S, Liu F. Interaction of dopamine D1 receptor with N-ethylmaleimide-sensitive factor is important for the membrane localization of the receptor. J Neurosci Res. 2010;88:2504–2512. doi: 10.1002/jnr.22401
  • Bayati A, Banks E, Han C, et al. Rapid macropinocytic transfer of α-synuclein to lysosomes. Cell Rep. 2022;40(3):111102. doi: 10.1016/j.celrep.2022.111102
  • Boyce AKJ, Kim MS, Wicki-Stordeur LE, et al. ATP stimulates pannexin 1 internalization to endosomal compartments. Biochem J. 2015;470(3):319–330. doi: 10.1042/BJ20141551
  • Boyce AKJ, Swayne LA. P2X7 receptor cross-talk regulates ATP-induced pannexin 1 internalization. Biochem J. 2017;474(13):2133–2144. doi: 10.1042/BCJ20170257
  • Boyce AKJ, van der Slagt E, Sanchez-Arias JC, et al. ATP triggers macropinocytosis that internalizes and is regulated by PANX1. bioRxiv [Internet]. 2020. https://www.biorxiv.org/content/early/2020/12/08/2020.11.19.389072
  • Yang G, Mishra M, Perera MA. Multi-omics studies in historically excluded populations: the road to equity. Clin Pharmacol Ther. 2023;113(3):541–556. doi: 10.1002/cpt.2818
  • Frederiksen SD. Prioritizing suggestive candidate genes in migraine: an opinion. Front Neurol. 2022;13:910366. doi: 10.3389/fneur.2022.910366
  • Gaudet P, Dessimoz C. Gene ontology: pitfalls, biases, and remedies. Methods Mol Biol. 2017;1446:189–205.
  • Beasley CL, Pennington K, Behan A, et al. Proteomic analysis of the anterior cingulate cortex in the major psychiatric disorders: evidence for disease-associated changes. Proteomics. 2006;6(11):3414–3425. doi: 10.1002/pmic.200500069
  • Hensley K, Kursula P, Butterfield DA. Collapsin response mediator protein-2 (CRMP2) is a plausible etiological factor and potential therapeutic target in alzheimer’s disease: comparison and contrast with microtubule-associated protein Tau. J Alzheimers Dis. 2016;53(1):1–14. doi: 10.3233/JAD-160076
  • Williamson R, van Aalten L, Mann DMA, et al. CRMP2 hyperphosphorylation is characteristic of Alzheimer’s disease and not a feature common to other neurodegenerative diseases. J Alzheimers Dis. 2011;27(3):615–625. doi: 10.3233/JAD-2011-110617
  • Cole AR, Noble W, van Aalten L, et al. Collapsin response mediator protein-2 hyperphosphorylation is an early event in Alzheimer’s disease progression. J Neurochem. 2007;103(3):1132–1144. doi: 10.1111/j.1471-4159.2007.04829.x
  • Braunschweig D, Krakowiak P, Duncanson P, et al. Autism-specific maternal autoantibodies recognize critical proteins in developing brain. Transl Psychiatry. 2013;3(7):e277. doi: 10.1038/tp.2013.50
  • Swayne LA, Johnstone SR, Ng CS, et al. Consideration of pannexin 1 channels in COVID-19 pathology and treatment. Am J Physiol Lung Cell Mol Physiol. 2020;319(1):L121–L125. doi: 10.1152/ajplung.00146.2020
  • Sanchez Arias JC, Wicki-Stordeur LE, Candlish RC, et al. PANX1 in inflammation heats up: new mechanistic insights with implications for injury and infection. Cell Calcium. 2020;90:102253. doi: 10.1016/j.ceca.2020.102253
  • Seo JH, Dalal MS, Contreras JE. Pannexin-1 channels as mediators of neuroinflammation. Int J Mol Sci. 2021;22(10):22. doi: 10.3390/ijms22105189
  • Yang Y, Delalio LJ, Best AK, et al. Endothelial pannexin 1 channels control inflammation by regulating intracellular Calcium. J Immunol [Internet]. 2020;204(11):2995–3007. doi: 10.4049/jimmunol.1901089
  • Süβ P, Lana AJ, Schlachetzki JCM. Chronic peripheral inflammation: a possible contributor to neurodegenerative diseases. Neural Regen Res [Internet]. 2021;16:1711. doi: 10.4103/1673-5374.306060.