Next Article in Journal
Effects of Heat Stress on Motion Characteristics and Metabolomic Profiles of Boar Spermatozoa
Next Article in Special Issue
SNP-by-CpG Site Interactions in ABCA7 Are Associated with Cognition in Older African Americans
Previous Article in Journal
Differences in Adipose Gene Expression Profiles between Male and Female Even Reindeer (Rangifer tarandus) in Sakha (Yakutia)
Previous Article in Special Issue
Exploration of the Moderating Effects of Physical Activity and Early Life Stress on the Relation between Brain-Derived Neurotrophic Factor (BDNF) rs6265 Variants and Depressive Symptoms among Adolescents
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recent Developments in Autism Genetic Research: A Scientometric Review from 2018 to 2022

1
Psychology Program, School of Social Sciences, Nanyang Technological University, Singapore 639818, Singapore
2
Department of Psychology and Cognitive Science, University of Trento, 38068 Rovereto, Italy
3
Sleep Education and Research Laboratory, UCL Institute of Education, London WC1H 0AA, UK
*
Author to whom correspondence should be addressed.
Genes 2022, 13(9), 1646; https://doi.org/10.3390/genes13091646
Submission received: 9 August 2022 / Revised: 12 September 2022 / Accepted: 12 September 2022 / Published: 14 September 2022

Abstract

:
Genetic research in Autism Spectrum Disorder (ASD) has progressed tremendously in recent decades. Dozens of genetic loci and hundreds of alterations in the genetic sequence, expression, epigenetic transformation, and interactions with other physiological and environmental systems have been found to increase the likelihood of developing ASD. There is therefore a need to represent this wide-ranging yet voluminous body of literature in a systematic manner so that this information can be synthesised and understood at a macro level. Therefore, this study made use of scientometric methods, particularly document co-citation analysis (DCA), to systematically review literature on ASD genetic research from 2018 to 2022. A total of 14,818 articles were extracted from Scopus and analyzed with CiteSpace. An optimized DCA analysis revealed that recent literature on ASD genetic research can be broadly organised into 12 major clusters representing various sub-topics. These clusters are briefly described in the manuscript and potential applications of this study are discussed.

1. Introduction

With its beginnings as a category of abnormal personality types ranging from antisocial, cyclothymic, and autistic to epileptic [1], autism became formally recognised as a disorder after Leo Kanner’s clinical description [2] and eventual inclusion in the third edition of the Diagnostic and Statistical Manual (DSM-III; [3]). Presently, Autism Spectrum Disorder (ASD) in the DSM-5 is characterised by deficits in social situations and restricted and/or repetitive behaviours, interests, or activities [4]. Recent studies estimate that ASD affects approximately 1 in 54 children in the United States (i.e., 18.52/1000), with varying estimates between 4.76 and 31.3/1000 in the European Union [5]. However, due to discrepancies in definitions, testing methods, and the age range of children tested in the national surveys, it is currently not possible to arrive at a worldwide prevalence, although most studies have concluded that prevalence rates have generally increased over time [6,7,8].
Among the scientific community, there has always been a suspicion that there was a hereditary basis to the development of autism. The first biologically grounded investigation into the etiology was conducted by MAY and Dunn [9], and made use of blood typing and antigen analysis. Less than a century later, beginning with Mendel’s discoveries in genetics [10], and in tandem with progress in biotechnological tools, genetic research in ASD is now made possible and more sophisticated than ever. Since the advent of genetic sequencing and manipulation, thousands of articles have been published on the genetic underpinnings of ASD, and many reviews synthesizing these findings have also been written (for example, see [11,12]). The latest review in this area is a meta-analysis written by Qiu et al. [13], which discussed the significant effect of polymorphisms on 12 candidate gene loci. However, rather than a singular disorder with a monogenic cause, ASD is an umbrella of neurodevelopmental abnormalities. ASD has been linked to dozens of genetic loci and hundreds of alterations from the genetic sequence to its expression, epigenetic transformation, and interactions with other physiological and environmental systems, all of which can be studied under the umbrella of ASD genetic research. Admirable attempts to summarise and report findings across these sub-categories have surfaced (e.g., Ref. [14]), but a majority of reviews in this area are restricted to only a particular aspect of ASD genetics. For example, while the narrative review by Cataldo et al. [15] focused on oxytocin and arginine-vasopressin receptors, the systematic review by Azhari et al. [16] examined the mechanisms related to the gut–brain hypothesis. Rather than narrative reviews which may organise the existing literature informally without declaring a methodology and may inevitably miss out on relevant publications [17], or meta-analyses that require a much more homogeneous set of studies and data, an alternative approach may be needed to systematically chart out the latest developments in ASD genetic research where the available literature is wide-ranging and varied.
The present paper therefore aims to make use of scientometric methods to perform a systematic review of the literature related to ASD genetic research in the last 5 years (i.e., 2018 to 2022). Further, based on the generated research clusters that arise from scientometric analysis, a brief survey of prominent citing and cited papers in each cluster will be described. Scientometry is a quantitative method of systematic review that measures the production of knowledge in a particular field by mapping out the quantitative relationships between publications [18,19]. Scientometric reviews proved useful to detect the developments of sub-specializations in topical research (e.g., [20]), where thematic research clusters are formed organically through the detection of quantitative relationships among documents. Compared to manual narrative reviews, scientometric reviews are data-driven, systematic, objective, and they are able to present findings representative of the entire scope of research with less bias [21].

2. Materials and Methods

2.1. Data Collection and Conversion

Prior to conducting scientometric analysis, relevant literature has to be systematically collated and their metadata exported. To achieve this, most scientometric reviews rely on searches on established citation databases such as Web of Science, Scopus, and other similar options. For the present review, a literature search was conducted on 24 July 2022 on Scopus with the search string: “TITLE-ABS-KEY((“ASD” OR “autism” OR autist* OR “Autism Spectrum Disorder”) AND (gene* OR geno*)) AND LIMIT-TO(LANGUAGE, “English”) AND LIMIT-TO(PUBYEAR, 2022) OR LIMIT-TO(PUBYEAR, 2021) OR LIMIT-TO(PUBYEAR, 2020) OR LIMIT-TO(PUBYEAR, 2019) OR LIMIT-TO(PUBYEAR, 2018))”. While the use of multiple databases in this step is possible, it would result in numerous duplicate entries and irregularities in citation formatting that would be difficult to resolve. Therefore, only one citation platform was chosen. Scopus was chosen over other platforms for its broader coverage in terms of number of indexed journals [22]. This method of literature search was previously adopted in [23] and is recommended by Chen [24]. Based on these criteria, a total of 14,818 articles was found to (1) be indexed with a combination of autism- and genetics-related terms in their title, abstract, or keywords, (2) be published in the English language, and (3) be published between 2018 and 2022. The research was limited to documents written in English to ensure that only the international scientific literature on the field was included in the data sample.
Unlike other systematic reviews, which rely heavily on a screening process to determine documents’ relevance, this dataset did not go through a screening process after extracting articles. This is because it is recommended that scientometric analysis on CiteSpace defers this screening process during qualitative analysis of each cluster in order to avoid systematic bias of excluding potential ambiguous terms [24]. The downloaded sample of documents was then imported into CiteSpace (version 6.1.R2) for scientometric analysis. Developed by Chen [25], CiteSpace is a freely available application built on Java that can be used to systematically map scientific literature. CiteSpace supports citation data exported from major databases such as Web of Science, PubMed and Scopus, and can be used to map temporal and structural patterns in a field of study. These patterns can be detected based on co-citations (such as in the present review), or author collaborations and directed citations. In generating these maps, clusters are produced where there are dense but common networks of co-citations (such as in the present review), representing sub-themes or specializations in a given field of research. The software also has applications in tracking the development of research over time, and is able to identify specific documents which are pivotal and have wide-ranging influence in a given field of research, as well as identify prolific authors. When importing data into CiteSpace, out of the total 948,381 references identified in the downloaded documents, 937,915 references were successfully converted, indicating a success rate of 98.90% (i.e., data loss of 1.10%). The data loss of the current work is within the acceptable bounds of 1.00% to 5.00%, and it may be considered negligible [26]. After the conversion step, the “Remove Alias” function was turned on in CiteSpace in order to eliminate repeated or identical entries.

2.2. Document Co-Citation Analysis

Using the converted references, a document co-citation analysis (DCA) was conducted on CiteSpace. DCA is a form of scientometric analysis that makes use of the frequency with which two or more papers are cited together (co-cited) in source articles [22,27]. The foundation of DCA rests on the assumption that higher co-citation frequencies between two or more documents are a marker of common lines of research between the cited documents [28]. Based on these principles, the final DCA network consists of two types of nodes: documents that are frequently cited together and main citing documents. To construct a DCA network, there are a variety of criteria that may be used to select the network’s nodes. Mainly, the three node selection criteria are: G-index, Top N, and Top N%. The G-index is a representation of an author’s citation score (i.e., higher G generally indicates higher author citations) and was designed as an improvement of the previously established H-index as it weighs more heavily the author’s most cited g publications [26,29,30,31]. To calculate G-index, the “largest number that equals the average number of citations of the most highly cited g publications” [29] is taken. The scaling factor k for the G-index criterion is introduced by CiteSpace to regulate the size of the network and can be any positive number [22]. The larger the k value, the more nodes included in the network. On the other hand, Top N and Top N% select the most cited N or N% references in a given period of time (i.e., time slice) [32]. In this review, the time slice is always kept consistent at 1 year per slice. For example, a Top N = 25 will select only the top 25 most cited references in each year (i.e., 2018 to 2022) for inclusion as nodes, whereas a Top N% = 25 will select the top quartile of most cited references in each year for inclusion as nodes.
Additionally, to allow CiteSpace to explore all possible links between older and more recent references and documents, the Look Back Years function was set to −1 (i.e., unlimited). Following the procedure of previously published papers [22], in this review, the following criteria were tested to optimize the results from the final DCA: G-index with scaling factor k set at 15, 25, and 50; Top N with N set at 25 and 50; Top N% with N set at 5, 10, and 15. The generated DCAs were evaluated to arrive at a final network that consists of coherent and distinct research clusters, and that is visually balanced and representative of the dataset. Particularly, the overall effects on the network’s structural metrics, the number of included nodes, and the amount of identified clusters drove the selection of the optimal node selection criteria to generate the final DCA. Based on this approach, the optimal DCA network was obtained using G-index with scaling factor k set at 50. Finally, clusters were obtained on CiteSpace using the “Clustering” function, which would systematically group the common co-citation pathways into clusters representing sub-themes and specializations within the research. The study flow diagram beginning with identification and retrieval from Scopus to the final number of nodes included in DCA can be found in Figure 1.

2.3. Metrics

Two types of metric are used to evaluate results in CiteSpace—namely, structural and temporal metrics. Structural metrics include modularity Q, silhouette score, and betweenness centrality. Modularity Q represents the degree of divisibility of a network into groups of nodes (i.e., modules or clusters) [34]. Modularity Q values range from 0 to 1, where values closer to 1 mean high network divisibility (i.e., distinct clusters and good structure) [28]. Conversely, silhouette refers to the consistency (i.e., cohesion and separation) within each cluster and ranges from −1 to 1, where values closer to 1 mean greater homogeneity within the cluster [35,36]. Finally, betweenness centrality measures the extent to which a node connects an arbitrary pair of nodes in the network [32,37]. While the values of betweenness centrality can range from 0 to 1, a value closer to 1 indicates that a publication is wide-reaching, due to its tendency to be referenced between other (less related) references and papers in the network [36]. The group of temporal metrics include citation burstness and sigma. Citation burstness represents a sudden increase in the number of citations received by a particular document over a period of time, and it allows detecting prominent publications that have received widespread attention from the scientific community [24]. Citation burstness is computed using Kleinberg’s algorithm [38]. While the lowest possible value for citation burstness is 0 (i.e., the document does not report a citation burst in its citation history), there is no theoretical upper limit for citation burstness. Finally, betweenness centrality and citation burstness values are combined with the equation (centrality + 1) b u r s t n e s s to derive sigma. The sigma metric is an index of a document’s novelty and impact on the network.

3. Results

3.1. Structural Properties of DCA Network

The final DCA network consists of 1424 nodes and 7660 links (averaging approximately 5.38 links/node). Its modularity Q value is 0.6631, indicating a moderately divisible network with distinct clusters, while the average silhouette score is 0.875, indicating high homogeneity within the clusters. The network contains 12 major clusters (see Figure 2). Cluster #0 (size = 218; silhouette = 0.823; mean publication year = 2018), cluster #1 (size = 158; silhouette = 0.920; mean publication year = 2019), and cluster #2 (size = 132; silhouette = 0.850; mean publication year = 2018) are the largest clusters in the network. In terms of silhouette score, the clusters wth higher internal homogeneity were, in order, cluster #14 (silhouette = 1.000; size = 4; mean publication year = 2020), cluster #10 (silhouette = 0.985; size = 49; mean publication year = 2019), and cluster #1. The size, individual silhouette scores, average year of publication and recommended cluster labels of the major clusters are found in Table 1. Recommended labels of the clusters are generated using the log-likelihood ratio (LLR) algorithm available on CiteSpace, which identifies the most unique and yet representative terms present in each cluster [32]. While LLR provides the most accurate labels compared to other automated labelling methods on CiteSpace [39], it may nonetheless lack precision as compared to manual labelling [40]. References and papers contributing to each cluster are subsequently summarised in the Discussion section. Where renaming the clusters is deemed more appropriate, alternative cluster labels that are more representative of the references comprising the clusters are proposed.

3.2. Documents with a Citation Burst

A total of 163 documents are recorded with a citation burst in their history. All 163 documents have a citation burst higher than 1.9. In the network, the document with the highest citation burstness is the review of Autism Spectrum Disorder authored by Lord et al. [41] (strength of burst = 14.357; burst duration = 2). The following documents in order of citation burst strength are authored by Grove et al. [42] (strength of burst = 9.462; burst duration = 2) and Iakoucheva et al. [43] (strength of burst = 8.080; burst duration = 2). A partial list of the 20 documents with the highest citation burstness and their metrics are summarised in Table 2, in accordance with Carollo et al. [33], Gaggero et al. [39], Lim et al. [44]. It should also be noted that documents with their “Burst End” year marked as 2022 could be considered as still experiencing their citation bursts, implying that several of these references may continue to be highly cited even beyond 2022.

4. Discussion

To reiterate the aims of the review, the present paper will make use of scientometric methods to perform a systematic review of the literature related to ASD genetic research in the last 5 years (i.e., 2018 to 2022). Subsequently, based on the generated research clusters from scientometric analysis, a brief qualitative survey of prominent citing and cited papers in each cluster will be described.
As can be seen in Figure 2, there are some hints of overlapping contributing papers between clusters, which corroborate the quantitative finding that the generated DCA network had only moderately divisible clusters (modularity Q = 0.6631). On one hand, while it attests to the wide relevance of these papers towards sub-themes in ASD genetic research, it also shows that the different clusters are intrinsically related and rely on findings from beyond their various specializations.
In the subsequent discussion by cluster, the main citing papers that contributed to the cluster in terms of their coverage (i.e., number of references in the cluster that are cited by that paper) and the relevant cited references are reported and described, in accordance with [33]. Additionally, citing papers’ global citing score (GCS) is also reported. GCS indicates the total number of papers’ citations in Scopus. Cited references, on the other hand, are described below where applicable in terms of their frequency of being cited by the citing papers in each cluster. Where the renaming of clusters was deemed necessary, the renamed label was decided upon based on content covered by both citing documents and their corresponding references.

4.1. Cluster #0: Networks and Pathways

The largest cluster—namely, Cluster #0—is the least homogeneous of the major clusters, based on the silhouette metric. The major citing document in Cluster #0 is authored by Joensuu et al. [62], with a coverage of 57 documents and a GCS of 37. Based on a survey of the contributing papers, the thematic focus of the cluster appears to be on “Networks and Pathways”—the mechanisms by which genetic alterations may lead to differences in gene expression, physiological function and eventually ASD symptoms. A majority of citing documents focus on neural mechanisms and circuitry of ASD [15,63,64,65,66,67], particularly making use of evidence from chromatin remodelling and transcriptome analyses as cited papers [68,69,70]. The top 10 citing papers of Cluster #0 are seen in Table 3.

4.2. Cluster #1: Gut Microbiota

Cluster #1 was labelled as “Gut Microbiota”. In this cluster, the main citing document is authored by Guang et al. [71] with a coverage of 38 and a GCS of 98. As for the cluster’s label, many citing and cited papers in the cluster focus on the impact of microbes present in gut microbiomes of individuals with ASD on ASD symptoms (e.g., Refs. [45,75,76,77,78]; please see [79] for a critical analysis). A recent expansion of research in this area links gut microbiota to ASD via the gut–immune–brain axis [54,80,81], where maternal and fetal inflammatory responses to gut dysbacteriosis may have affected fetal neurodevelopment [16]. Some of the most frequently cited references in this cluster also support the gut–brain link in ASD, where gene expression, particularly those related to synaptic function and potentiation, may be altered due to gut microbiota, and that ASD severity is correlated with severity of gastrointestinal tract dysfunction [82]. Conversely, it has also been found that genetic variation may lead to differential composition of gut microbiota [83]. In this area of research, a recently published article (published a mere two days before the present review) has also reviewed the characteristics of gut microbiota that have been found to lead to ASD, as well as potential therapeutic applications to alleviate ASD symptoms by regulating the microbiome [84]. The top 10 citing papers of Cluster #1 are seen in Table 4.

4.3. Clusters #2 and #3: Mouse Models

Unlike the preceding clusters which had topical focuses, Clusters #2 (manually renamed “Fragile X Syndrome”) and #3 (manually renamed “SHANK1,2,3 Genes”) are discussed together primarily because of the use of animal models in their research methodology (e.g., Refs. [65,67,90,91,92,93]). The documents authored by Wang et al. [94] and by Verma et al. [91] are the major citing documents in cluster #2 with a coverage of 64 (GCS = 8) and 46 (GCS = 23), respectively. The major citing documents in Cluster #3 are authored by Wang et al. [94] and Soler et al. [95] with a coverage of 54 (GCS = 8) and 40 (GCS = 26), respectively. Mouse models in ASD research can be traced back to the work by Consorthium et al. [96], who used FMR1 knockout mice to investigate fragile X syndrome, one of the monogenic causes of ASD [97,98,99]. In fact, mouse model research in Cluster #2 focuses heavily on fragile X, where citing documents relied most frequently on Fragile X studies (e.g., Ref. [100] with a citation frequency of 52, Ref. [101] with a citation frequency of 47 and [98] with a citation frequency of 44). On the other hand, Cluster #3 uses mouse models to investigate the SHANK1, SHANK2, and SHANK3 genes that code for synaptic formation and transmissions [93,102,103,104], where mutations in these loci may also be a monogenic cause of ASD [71,91,94,105]. Therefore, proposed labels may be “Fragile X Syndrome” and “SHANK1,2,3 Genes” for Clusters #2 and #3, respectively. The top 10 citing papers of Clusters #2 and #3 are seen in Table 5 and Table 6 respectively.

4.4. Clusters #4 and #6: Stem Cell Technology

The major citing documents for Clusters #4 and #6 are authored by St. Clair and Johnstone [108] (coverage = 22; GCS = 13) and Lord et al. [109] (coverage = 23; GCS = 211), respectively. While Clusters #4 and #6 were originally labelled “Valproic Acid” and “Brain Organoid”, respectively, a more representative label for both clusters may be “Stem Cell Technology”, as papers in the two clusters focus on the exposure of stem cells to various molecules, including valproic acid (a molecule that is associated with higher likelihood of developing ASD [110]), to investigate the development of ASD-like phenotypes [108,111,112,113,114]. Papers in Cluster #4 use stem cells to mimic the prenatal development processes in order to trace the pathogeneses of ASD. In Cluster #6, research methodologies take it one step further through the use of brain organoids—a self-organising tissue made of these stem cells to simulate the structure and function of the human brain [115,116,117,118,119] (please see [120] for a review of organoid technology). Generally, in contrast to animal models which may be difficult to validate in human ASD [121,122], stem cell technology offers a valuable alternative in modelling genetic variants implicated in autism by using induced pluripotent stem cells (iPSC or its human-only variant, hiPSC) from the reprogramming of somatic cells. The combination of stem cell with CRISPR gene editing, three-dimensional organoid development, and in vitro to in vivo engraftment technologies opens up avenues for stem cell therapies in the future [123]. The top 10 citing papers of Cluster #4 and #6 are seen in Table 7 and Table 8.

4.5. Cluster #5: Genomic Architecture

In Cluster #5, the major citing document is written by Al-Dewik et al. [132] and has a coverage of 20 documents and a GCS of 5. As the term “Genomic Architectured” implies, many studies in this cluster make use of big data, such as genome-wide associations or transcriptome analyses, to uncover the genetic bases of ASD [133,134,135,136,137,138,139,140]. The top 10 citing papers of Cluster #5 are seen in Table 9.

4.6. Cluster #7: Psychiatric Disorder

The document by Lord et al. [41] with a coverage of 18 and a GCS of 211 is the main citing document of Cluster #7 “Psychiatric Disorder”. This cluster represents an expansion of genetic studies beyond ASD to other neurological and psychiatric disorders such as major depressive disorder [148], schizophrenia [95,108,149,150,151], obsessive-compulsive disorders [152], Parkinson’s disease [153], Tourette’s syndrome [154], as well as behavioural indicators of psychiatric disorders such as self-harm and suicidality [155], and sociability [156]. Studies in this cluster either are comparative in nature (i.e., analyzing and comparing genetic bases of the various disorders), or apply the success of genetic research in ASD to other disorders. Generally, the cluster is testament to the progress and accomplishments in the field of ASD that researchers of other psychiatric disorders may take inspiration from. The top 10 citing papers of Cluster #7 are seen in Table 10.

4.7. Cluster #8: Sex Difference

In Cluster #8, Rylaarsdam and Guemez-Gamboa [14] authored the major citing document with a coverage of 11 references and a GCS of 118. The current label “Sex Difference” is derived from some studies in the cluster investigating the significant discrepancy in male-to-female individuals diagnosed with ASD and the effects of sex on ASD [125,165,166,167,168], with a small extension of research into ASD and gender dysphoria [169]. Notably, one of the most frequently cited references, with a citation frequency of 112, in the cluster is a systematic review and meta-analysis by [170] on the sex ratio in ASD. The top 10 citing papers of Cluster #8 are seen in Table 11.

4.8. Cluster #9: Copy Number Variations (CNVs)

In Cluster #9, the major citing document is Jønch et al. [176], with a coverage of 11 and a GCS of 19. A more appropriate label for this cluster is “Copy Number Variations" (CNVs) due to the focus of contributing papers on CNV profiles in ASD. CNVs, which are the most common form of structural variations in the human genome [177] and may take the form of duplications or deletions, have been identified to be one of the risk factors for developing ASD [178]. According to the contributing papers in this cluster, the most-researched gene loci is 16p11.2 [179,180,181,182,183,184,185], which codes for proteins involved in cortical development [186]. Of the cited documents, some of the most widely cited documents indeed focused on 16p11.2 microdeletion and microduplication [187] (citation frequency of 69) and others examined CNV similarities between ASD and schizophrenia (e.g., Marshall et al. [188] with a citation frequency of 56 and Stefansson et al. [189] with a citation frequency of 50). The top 10 citing papers of Cluster #9 are seen in Table 12.

4.9. Cluster #10: Developmental Perspectives

Cluster #10 has Zwaigenbaum et al. [193] as major citing document with a coverage of 7 and a GCS of 20. Cluster #10 is also the most internally homogeneous cluster out of all major clusters. Contributing papers in this cluster discuss the developmental trajectory of ASD from diagnosis to adulthood [193,194,195], as well as the unique issues faced in early childhood [196,197,198] to the intersection between ASD and other health and social conditions in adulthood [199,200,201]. A major cited reference in this cluster, with a citation frequency of 100, dealt with the psychiatric co-morbidities of ASD in childhood [202]. Therefore, rather than “Autistic Adult”, which does not capture the development of ASD across a lifespan, a more appropriate label for this cluster may be “Developmental Perspectives”. The top 10 citing papers of Cluster #10 are seen in Table 13.

4.10. Cluster #14: Antiseizure Drug

The smallest cluster—namely, Cluster #14 “Antiseizure Drug”—has the paper authored by Hakami [208] as major citing document, with a coverage of 4 and a GCS of 2. The naming of this cluster is based on a series of two papers written by Hakami [208,209] on antiseizure drugs, and may be related to ASD due to the higher prevalence of seizure and epilepsy diagnoses among these individuals [210]. Similarly, other contributing papers focused on understanding the genetic bases of epilepsy [211,212,213] and epilepsy treatment [214,215]. In this cluster, contributing papers frequently cited references from the International League Against Epilepsy (ILAE) in their clinical descriptions and classifications of different seizures and epilepsies (e.g., Scheffer et al. [216], Fisher et al. [217,218] with citation frequencies of 43, 12, and 8, respectively). The top 10 citing papers of Cluster #14 are seen in Table 14.

4.11. Limitations and Future Recommendations

Despite the emergence of meaningful research clusters from scientometric analysis, this review nonetheless still has some limitations. Firstly, due to the choice of keywords used, relevant papers that had not indexed autism- and gene-related terms in their titles, abstracts, or keywords would inevitably be excluded from the dataset [25]. Papers that instead discussed ASD under the larger umbrella of neurodevelopmental disorders may also be missed as a result. Of course, unpublished but relevant papers would also be excluded from the analysis [222].
Secondly, DCA relies on the theory that higher co-citation frequencies between two or more documents are a marker of common lines of research or of emerging research trends within the field of study [28]. However, there are some caveats to a simplistic interpretation of DCA analyses. For example, a purely quantitative measure of co-citation does not reveal the relationship between two papers. A citing paper may be in agreement or disagreement with the cited reference [33]. Therefore, it is recommended by scientometricians that analysis be accompanied with a qualitative discussion [223] in order to fully understand the context of the clusters. Furthermore, as can be seen in the mean years of publication in the above clusters, DCA tends to under-represent newer publications as compared to older publications, as later publications tend to have fewer citations. Thus, it is important to consider here that citation numbers are not wholly indicative of the paper’s importance or impact.
Finally, it must be emphasised that findings regarding the citation burstness and duration are not conclusive, due to the short time period we used to conduct the scientometric review. While the search results yielded a significant amount of literature based on the time period under consideration (approximately 14 thousand entries), an alternative search without publication year limitations actually yielded 37 thousand entries, and would be more appropriate in assessing citation bursts of documents over time. Nonetheless, it was the intention of the present paper to conduct a review of only the most recent studies in the field of ASD and genetics, particularly because of the fast-paced nature of technological advancements. Future studies that are more interested in the development of this research over time may instead opt for a full coverage of the literature from its inception to the present day.

5. Conclusions

It is hoped that findings from this scientometric review will encourage researchers to make full use of the available literature in this field and integrate findings from various clusters and subspecialties into their future work. Additionally, it is noted that none of the clusters predominantly focused on the translation of laboratory findings to clinical applications, as well as the development of interdisciplinary treatments with medical, psychological, and occupational perspectives. The future of genetic research in ASD may see more interdisciplinary collaborations to facilitate the process from ‘bench to bedside’. Nonetheless, genetic findings on their own have exciting potential in personalized medicine [14], as well as genetic counselling and early intervention strategies [224].

Author Contributions

Conceptualization: A.C., G.E.; Data curation: M.L., A.C.; Formal analysis: A.C.; Supervision: G.E.; Writing—original draft: M.L., A.C.; Writing—review & editing: M.L., A.C., D.D., G.E. All authors read and agreed to the published version of the manuscript.

Funding

D.D. was funded by John and Lorna Wing Foundation, UK.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

Abbreviations

The following abbreviations are used in this manuscript:
ASDAutism Spectrum Disorder
DCADocument Co-Citation Analysis
LLRLog-Likelihood Ratio
GCSGlobal Citing Score
ILAEInternational League Against Epilepsy

References

  1. Rosanoff, A.J. A Theory of Personality Based Mainly on Psychiatric Experience. Psychol. Bull. 1920, 17, 281. [Google Scholar] [CrossRef]
  2. Kanner, L. Early infantile autism. J. Pediatr. 1944, 25, 211–217. [Google Scholar] [CrossRef]
  3. Volkmar, F.R.; Bregman, J.; Cohen, D.J.; Cicchetti, D.V. DSM-III and DSM-III-R diagnoses of autism. Am. J. Psychiatry 1988, 145, 1404–1408. [Google Scholar] [PubMed]
  4. Grzadzinski, R.; Huerta, M.; Lord, C. DSM-5 and Autism Spectrum Disorders (ASDs): An opportunity for identifying ASD subtypes. Mol. Autism 2013, 4, 12. [Google Scholar] [CrossRef]
  5. Chiarotti, F.; Venerosi, A. Epidemiology of Autism Spectrum Disorders: A review of worldwide prevalence estimates since 2014. Brain Sci. 2020, 10, 274. [Google Scholar] [CrossRef] [PubMed]
  6. Fombonne, E. Is there an epidemic of autism? Pediatrics 2001, 107, 411–412. [Google Scholar] [CrossRef]
  7. Randall, M.; Sciberras, E.; Brignell, A.; Ihsen, E.; Efron, D.; Dissanayake, C.; Williams, K. Autism Spectrum Disorder: Presentation and prevalence in a nationally representative Australian sample. Aust. N. Z. J. Psychiatry 2016, 50, 243–253. [Google Scholar] [CrossRef]
  8. Ouellette-Kuntz, H.; Coo, H.; Lam, M.; Breitenbach, M.M.; Hennessey, P.E.; Jackman, P.D.; Lewis, M.; Dewey, D.; Bernier, F.P.; Chung, A.M. The changing prevalence of autism in three regions of Canada. J. Autism Dev. Disord. 2014, 44, 120–136. [Google Scholar] [CrossRef]
  9. MAY, J.; Dunn, L. A preliminary study of inherited blood group antigens in families having an index case of early infantile autism. J. Hered. 1961, 52, 239–240. [Google Scholar] [CrossRef]
  10. Gayon, J. From Mendel to epigenetics: History of genetics. Comptes Rendus Biol. 2016, 339, 225–230. [Google Scholar] [CrossRef] [Green Version]
  11. Muhle, R.; Trentacoste, S.V.; Rapin, I. The genetics of autism. Pediatrics 2004, 113, e472–e486. [Google Scholar] [CrossRef] [PubMed]
  12. Geschwind, D.H. Genetics of Autism Spectrum Disorders. Trends Cogn. Sci. 2011, 15, 409–416. [Google Scholar] [CrossRef] [PubMed]
  13. Qiu, S.; Qiu, Y.; Li, Y.; Cong, X. Genetics of Autism Spectrum Disorder: An umbrella review of systematic reviews and meta-analyses. Transl. Psychiatry 2022, 12, 249. [Google Scholar] [CrossRef]
  14. Rylaarsdam, L.; Guemez-Gamboa, A. Genetic causes and modifiers of Autism Spectrum Disorder. Front. Cell. Neurosci. 2019, 13, 385. [Google Scholar] [CrossRef] [PubMed]
  15. Cataldo, I.; Azhari, A.; Esposito, G. A review of oxytocin and arginine-vasopressin receptors and their modulation of Autism Spectrum Disorder. Front. Mol. Neurosci. 2018, 11, 27. [Google Scholar] [CrossRef] [PubMed]
  16. Azhari, A.; Azizan, F.; Esposito, G. A systematic review of gut-immune-brain mechanisms in Autism Spectrum Disorder. Dev. Psychobiol. 2019, 61, 752–771. [Google Scholar] [CrossRef]
  17. Jahan, N.; Naveed, S.; Zeshan, M.; Tahir, M.A. How to conduct a systematic review: A narrative literature review. Cureus 2016, 8, e864. [Google Scholar] [CrossRef]
  18. Mulchenko, Z. Measurement of science. study of the development of science as an information process. In USAF Foreign Technology Division Translation AD735634; National Technical Information Service: Springfield, VA, USA, 1969. [Google Scholar]
  19. Börner, K.; Chen, C.; Boyack, K.W. Visualizing knowledge domains. Annu. Rev. Inf. Sci. Technol. 2003, 37, 179–255. [Google Scholar] [CrossRef]
  20. Junior, O.F. The Legacy of David Bohm in Physics—An Essay in Scientometry. In David Bohm; Springer: Cham, Switzerland, 2019; pp. 223–240. [Google Scholar]
  21. Su, H.N.; Lee, P.C. Mapping knowledge structure by keyword co-occurrence: A first look at journal papers in Technology Foresight. Scientometrics 2010, 85, 65–79. [Google Scholar] [CrossRef]
  22. Cataldo, I.; Lieu, A.A.; Carollo, A.; Bornstein, M.H.; Gabrieli, G.; Lee, A.; Esposito, G. From the cradle to the web: The growth of “sharenting”—A scientometric perspective. Hum. Behav. Emerg. Technol. 2022, 2022, 5607422. [Google Scholar] [CrossRef]
  23. Aryadoust, V.; Zakaria, A.; Lim, M.H.; Chen, C. An extensive knowledge mapping review of measurement and validity in language assessment and SLA research. Front. Psychol. 2020, 11, 1941. [Google Scholar] [CrossRef] [PubMed]
  24. Chen, C. Science mapping: A systematic review of the literature. J. Data Inf. Sci. 2017, 2, 1–40. [Google Scholar] [CrossRef]
  25. Chen, C. CiteSpace II: Detecting and visualizing emerging trends and transient patterns in scientific literature. J. Am. Soc. Inf. Sci. Technol. 2006, 57, 359–377. [Google Scholar] [CrossRef]
  26. Chen, C. CiteSpace: A Practical Guide for Mapping Scientific Literature; Nova Science Publishers: Hauppauge, NY, USA, 2016. [Google Scholar]
  27. Small, H. Co-citation context analysis and the structure of paradigms. J. Doc. 1980, 36, 183–196. [Google Scholar] [CrossRef]
  28. Chen, C.; Ibekwe-SanJuan, F.; Hou, J. The structure and dynamics of cocitation clusters: A multiple-perspective cocitation analysis. J. Am. Soc. Inf. Sci. Technol. 2010, 61, 1386–1409. [Google Scholar] [CrossRef]
  29. Egghe, L. An improvement of the h-index: The G-index. ISSI Newsl. 2006, 2, 8–9. [Google Scholar]
  30. Bornmann, L.; Daniel, H.D. What do we know about the h index? J. Am. Soc. Inf. Sci. Technol. 2007, 58, 1381–1385. [Google Scholar] [CrossRef]
  31. Alonso, S.; Cabrerizo, F.J.; Herrera-Viedma, E.; Herrera, F. h-Index: A review focused in its variants, computation and standardization for different scientific fields. J. Inf. 2009, 3, 273–289. [Google Scholar] [CrossRef]
  32. Chen, C. The citespace manual. Coll. Comput. Inform. 2014, 1, 1–84. [Google Scholar]
  33. Carollo, A.; Balagtas, J.P.M.; Neoh, M.J.Y.; Esposito, G. A scientometric approach to review the role of the medial preoptic area (MPOA) in parental behavior. Brain Sci. 2021, 11, 393. [Google Scholar] [CrossRef]
  34. Newman, M.E. Modularity and community structure in networks. Proc. Natl. Acad. Sci. USA 2006, 103, 8577–8582. [Google Scholar] [CrossRef] [PubMed]
  35. Rousseeuw, P.J. Silhouettes: A graphical aid to the interpretation and validation of cluster analysis. J. Comput. Appl. Math. 1987, 20, 53–65. [Google Scholar] [CrossRef] [Green Version]
  36. Aryadoust, V.; Ang, B.H. Exploring the frontiers of eye tracking research in language studies: A novel co-citation scientometric review. Comput. Assist. Lang. Learn. 2019, 34, 898–933. [Google Scholar] [CrossRef]
  37. Freeman, L.C. A set of measures of centrality based on betweenness. Sociometry 1977, 40, 35–41. [Google Scholar] [CrossRef]
  38. Kleinberg, J. Bursty and hierarchical structure in streams. Data Min. Knowl. Discov. 2003, 7, 373–397. [Google Scholar] [CrossRef]
  39. Gaggero, G.; Bonassi, A.; Dellantonio, S.; Pastore, L.; Aryadoust, V.; Esposito, G. A scientometric review of alexithymia: Mapping thematic and disciplinary shifts in half a century of research. Front. Psychiatry 2020, 11, 1405. [Google Scholar] [CrossRef]
  40. Carollo, A.; Lim, M.; Aryadoust, V.; Esposito, G. Interpersonal Synchrony in the Context of Caregiver-Child Interactions: A Document Co-citation Analysis. Front. Psychol. 2021, 12, 2977. [Google Scholar] [CrossRef]
  41. Lord, C.; Elsabbagh, M.; Baird, G.; Veenstra-Vanderweele, J. Autism Spectrum Disorder. Lancet 2018, 392, 508–520. [Google Scholar] [CrossRef]
  42. Grove, J.; Ripke, S.; Als, T.D.; Mattheisen, M.; Walters, R.K.; Won, H.; Pallesen, J.; Agerbo, E.; Andreassen, O.A.; Anney, R.; et al. Identification of common genetic risk variants for autism spectrum disorder. Nat. Genet. 2019, 51, 431–444. [Google Scholar] [CrossRef]
  43. Iakoucheva, L.M.; Muotri, A.R.; Sebat, J. Getting to the cores of autism. Cell 2019, 178, 1287–1298. [Google Scholar] [CrossRef]
  44. Lim, M.; Carollo, A.; Chen, S.A.; Esposito, G. Surveying 80 years of psychodrama research: A scientometric review. Front. Psychiatry 2021, 12, 780542. [Google Scholar] [CrossRef] [PubMed]
  45. Sharon, G.; Cruz, N.J.; Kang, D.W.; Gandal, M.J.; Wang, B.; Kim, Y.M.; Zink, E.M.; Casey, C.P.; Taylor, B.C.; Lane, C.J.; et al. Human gut microbiota from Autism Spectrum Disorder promote behavioral symptoms in mice. Cell 2019, 177, 1600–1618. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Ruzzo, E.K.; Pérez-Cano, L.; Jung, J.Y.; Wang, L.k.; Kashef-Haghighi, D.; Hartl, C.; Singh, C.; Xu, J.; Hoekstra, J.N.; Leventhal, O.; et al. Inherited and de novo genetic risk for autism impacts shared networks. Cell 2019, 178, 850–866. [Google Scholar] [CrossRef] [PubMed]
  47. Kim, Y.S.; Leventhal, B.L.; Koh, Y.J.; Fombonne, E.; Laska, E.; Lim, E.C.; Cheon, K.A.; Kim, S.J.; Kim, Y.K.; Lee, H.; et al. Prevalence of Autism Spectrum Disorders in a total population sample. Am. J. Psychiatry 2011, 168, 904–912. [Google Scholar] [CrossRef]
  48. Abraham, A.; Milham, M.P.; Di Martino, A.; Craddock, R.C.; Samaras, D.; Thirion, B.; Varoquaux, G. Deriving reproducible biomarkers from multi-site resting-state data: An Autism-based example. NeuroImage 2017, 147, 736–745. [Google Scholar] [CrossRef]
  49. Lim, E.T.; Uddin, M.; De Rubeis, S.; Chan, Y.; Kamumbu, A.S.; Zhang, X.; D’Gama, A.M.; Kim, S.N.; Hill, R.S.; Goldberg, A.P.; et al. Rates, distribution and implications of postzygotic mosaic mutations in Autism Spectrum Disorder. Nat. Neurosci. 2017, 20, 1217–1224. [Google Scholar] [CrossRef]
  50. Yang, M.; Bozdagi, O.; Scattoni, M.L.; Wöhr, M.; Roullet, F.I.; Katz, A.M.; Abrams, D.N.; Kalikhman, D.; Simon, H.; Woldeyohannes, L.; et al. Reduced excitatory neurotransmission and mild autism-relevant phenotypes in adolescent Shank3 null mutant mice. J. Neurosci. 2012, 32, 6525–6541. [Google Scholar] [CrossRef]
  51. Lee, P.H.; Anttila, V.; Won, H.; Feng, Y.C.A.; Rosenthal, J.; Zhu, Z.; Tucker-Drob, E.M.; Nivard, M.G.; Grotzinger, A.D.; Posthuma, D.; et al. Genomic relationships, novel loci, and pleiotropic mechanisms across eight psychiatric disorders. Cell 2019, 179, 1469–1482. [Google Scholar] [CrossRef]
  52. Nowakowski, T.J.; Bhaduri, A.; Pollen, A.A.; Alvarado, B.; Mostajo-Radji, M.A.; Di Lullo, E.; Haeussler, M.; Sandoval-Espinosa, C.; Liu, S.J.; Velmeshev, D.; et al. Spatiotemporal gene expression trajectories reveal developmental hierarchies of the human cortex. Science 2017, 358, 1318–1323. [Google Scholar] [CrossRef]
  53. Velmeshev, D.; Schirmer, L.; Jung, D.; Haeussler, M.; Perez, Y.; Mayer, S.; Bhaduri, A.; Goyal, N.; Rowitch, D.H.; Kriegstein, A.R. Single-cell genomics identifies cell type–specific molecular changes in autism. Science 2019, 364, 685–689. [Google Scholar] [CrossRef]
  54. Matta, S.M.; Hill-Yardin, E.L.; Crack, P.J. The influence of neuroinflammation in Autism Spectrum Disorder. Brain Behav. Immun. 2019, 79, 75–90. [Google Scholar] [CrossRef] [PubMed]
  55. Estes, M.L.; McAllister, A.K. Immune mediators in the brain and peripheral tissues in autism spectrum disorder. Nat. Rev. Neurosci. 2015, 16, 469–486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Goines, P.E.; Ashwood, P. Cytokine dysregulation in Autism Spectrum Disorders (ASD): Possible role of the environment. Neurotoxicol. Teratol. 2013, 36, 67–81. [Google Scholar] [CrossRef] [PubMed]
  57. Pantelis, C.; Papadimitriou, G.N.; Papiol, S.; Parkhomenko, E.; Pato, M.T.; Paunio, T.; Pejovic-Milovancevic, M.; Perkins, D.O.; Pietiläinen, O.; Pimm, J. Biological insights from 108 schizophrenia-associated genetic loci. Nature 2014, 511, 421–427. [Google Scholar]
  58. Yuen, R.K.; Thiruvahindrapuram, B.; Merico, D.; Walker, S.; Tammimies, K.; Hoang, N.; Chrysler, C.; Nalpathamkalam, T.; Pellecchia, G.; Liu, Y.; et al. Whole-genome sequencing of quartet families with autism spectrum disorder. Nat. Med. 2015, 21, 185–191. [Google Scholar] [CrossRef]
  59. Antoine, M.W.; Langberg, T.; Schnepel, P.; Feldman, D.E. Increased excitation-inhibition ratio stabilizes synapse and circuit excitability in four autism mouse models. Neuron 2019, 101, 648–661. [Google Scholar] [CrossRef]
  60. Schafer, S.T.; Paquola, A.; Stern, S.; Gosselin, D.; Ku, M.; Pena, M.; Kuret, T.J.; Liyanage, M.; Mansour, A.A.; Jaeger, B.N.; et al. Pathological priming causes developmental gene network heterochronicity in autistic subject-derived neurons. Nat. Neurosci. 2019, 22, 243–255. [Google Scholar] [CrossRef]
  61. Stahl, E.A.; Breen, G.; Forstner, A.J.; McQuillin, A.; Ripke, S.; Trubetskoy, V.; Mattheisen, M.; Wang, Y.; Coleman, J.R.; Gaspar, H.A.; et al. Genome-wide association study identifies 30 loci associated with bipolar disorder. Nat. Genet. 2019, 51, 793–803. [Google Scholar] [CrossRef]
  62. Joensuu, M.; Lanoue, V.; Hotulainen, P. Dendritic spine actin cytoskeleton in Autism Spectrum Disorder. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2018, 84, 362–381. [Google Scholar] [CrossRef]
  63. Alonso-Gonzalez, A.; Rodriguez-Fontenla, C.; Carracedo, A. De novo mutations (DNMs) in Autism Spectrum Disorder (ASD): Pathway and network analysis. Front. Genet. 2018, 9, 406. [Google Scholar] [CrossRef]
  64. Ayhan, F.; Konopka, G. Regulatory genes and pathways disrupted in Autism Spectrum Disorders. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2019, 89, 57–64. [Google Scholar] [CrossRef] [PubMed]
  65. Hui, K.; Katayama, Y.; Nakayama, K.I.; Nomura, J.; Sakurai, T. Characterizing vulnerable brain areas and circuits in mouse models of autism: Towards understanding pathogenesis and new therapeutic approaches. Neurosci. Biobehav. Rev. 2020, 110, 77–91. [Google Scholar] [CrossRef] [PubMed]
  66. Eyring, K.W.; Geschwind, D.H. Three decades of ASD genetics: Building a foundation for neurobiological understanding and treatment. Hum. Mol. Genet. 2021, 30, R236–R244. [Google Scholar] [CrossRef]
  67. Gandhi, T.; Lee, C.C. Neural mechanisms underlying repetitive behaviors in rodent models of Autism Spectrum Disorders. Front. Cell. Neurosci. 2021, 14, 592710. [Google Scholar] [CrossRef] [PubMed]
  68. Voineagu, I.; Wang, X.; Johnston, P.; Lowe, J.K.; Tian, Y.; Horvath, S.; Mill, J.; Cantor, R.M.; Blencowe, B.J.; Geschwind, D.H. Transcriptomic analysis of autistic brain reveals convergent molecular pathology. Nature 2011, 474, 380–384. [Google Scholar] [CrossRef]
  69. Parikshak, N.N.; Luo, R.; Zhang, A.; Won, H.; Lowe, J.K.; Chandran, V.; Horvath, S.; Geschwind, D.H. Integrative functional genomic analyses implicate specific molecular pathways and circuits in autism. Cell 2013, 155, 1008–1021. [Google Scholar] [CrossRef]
  70. De Rubeis, S.; He, X.; Goldberg, A.P.; Poultney, C.S.; Samocha, K.; Ercument Cicek, A.; Kou, Y.; Liu, L.; Fromer, M.; Walker, S.; et al. Synaptic, transcriptional and chromatin genes disrupted in autism. Nature 2014, 515, 209–215. [Google Scholar] [CrossRef]
  71. Guang, S.; Pang, N.; Deng, X.; Yang, L.; He, F.; Wu, L.; Chen, C.; Yin, F.; Peng, J. Synaptopathology involved in Autism Spectrum Disorder. Front. Cell. Neurosci. 2018, 12, 470. [Google Scholar] [CrossRef]
  72. Garcia-Forn, M.; Boitnott, A.; Akpinar, Z.; De Rubeis, S. Linking autism risk genes to disruption of cortical development. Cells 2020, 9, 2500. [Google Scholar] [CrossRef]
  73. Diaz-Caneja, C.M.; State, M.; Hagerman, R.; Jacquemont, S.; Marín, O.; Bagni, C.; Umbricht, D.; Simonoff, E.; de Andrés-Trelles, F.; Kaale, A.; et al. A white paper on a neurodevelopmental framework for drug discovery in autism and other neurodevelopmental disorders. Eur. Neuropsychopharmacol. 2021, 48, 49–88. [Google Scholar] [CrossRef]
  74. DiCarlo, G.E.; Wallace, M.T. Modeling dopamine dysfunction in Autism Spectrum Disorder: From invertebrates to vertebrates. Neurosci. Biobehav. Rev. 2021, 133, 104494. [Google Scholar] [CrossRef] [PubMed]
  75. Zheng, Y.; Verhoeff, T.A.; Perez Pardo, P.; Garssen, J.; Kraneveld, A.D. The gut–brain axis in Autism Spectrum Disorder: A focus on the metalloproteases ADAM10 and ADAM17. Int. J. Mol. Sci. 2020, 22, 118. [Google Scholar] [CrossRef] [PubMed]
  76. Cryan, J.F.; O’Riordan, K.J.; Cowan, C.S.; Sandhu, K.V.; Bastiaanssen, T.F.; Boehme, M.; Codagnone, M.G.; Cussotto, S.; Fulling, C.; Golubeva, A.V.; et al. The microbiota-gut–brain axis. Physiol. Rev. 2019, 99, 1877–2013. [Google Scholar] [CrossRef] [PubMed]
  77. Fattorusso, A.; Di Genova, L.; Dell’Isola, G.B.; Mencaroni, E.; Esposito, S. Autism Spectrum Disorders and the gut microbiota. Nutrients 2019, 11, 521. [Google Scholar] [CrossRef]
  78. Srikantha, P.; Mohajeri, M.H. The possible role of the microbiota-gut–brain-axis in autism spectrum disorder. Int. J. Mol. Sci. 2019, 20, 2115. [Google Scholar] [CrossRef] [PubMed]
  79. Hooks, K.B.; Konsman, J.P.; O’Malley, M.A. Microbiota-gut–brain research: A critical analysis. Behav. Brain Sci. 2019, 42, e60. [Google Scholar] [CrossRef] [PubMed]
  80. Azhari, A.; Azizan, F.; Esposito, G. Beyond a gut feeling: How the immune system impacts the effect of gut microbiota in neurodevelopment. Behav. Brain Sci. 2019, 42. [Google Scholar] [CrossRef]
  81. van Sadelhoff, J.H.; Perez Pardo, P.; Wu, J.; Garssen, J.; Van Bergenhenegouwen, J.; Hogenkamp, A.; Hartog, A.; Kraneveld, A.D. The gut-immune-brain axis in Autism Spectrum Disorders; a focus on amino acids. Front. Endocrinol. 2019, 10, 247. [Google Scholar] [CrossRef]
  82. Tomova, A.; Husarova, V.; Lakatosova, S.; Bakos, J.; Vlkova, B.; Babinska, K.; Ostatnikova, D. Gastrointestinal microbiota in children with autism in Slovakia. Physiol. Behav. 2015, 138, 179–187. [Google Scholar] [CrossRef]
  83. Liu, Z.; Mao, X.; Dan, Z.; Pei, Y.; Xu, R.; Guo, M.; Liu, K.; Zhang, F.; Chen, J.; Su, C.; et al. Gene variations in Autism Spectrum Disorder are associated with alternation of gut microbiota, metabolites and cytokines. Gut Microbes 2021, 13, 1854967. [Google Scholar] [CrossRef]
  84. Taniya, M.A.; Chung, H.J.; Al Mamun, A.; Alam, S.; Aziz, M.; Emon, N.U.; Islam, M.; Podder, B.R.; Ara Mimi, A.; Aktar Suchi, S.; et al. Role of Gut Microbiome in Autism Spectrum Disorder and Its Therapeutic Regulation. Front. Cell. Infect. Microbiol. 2022, 12, 915701. [Google Scholar] [CrossRef] [PubMed]
  85. Patel, J.; Lukkes, J.L.; Shekhar, A. Overview of genetic models of Autism Spectrum Disorders. Prog. Brain Res. 2018, 241, 1–36. [Google Scholar] [PubMed]
  86. Yang, G.; Shcheglovitov, A. Probing disrupted neurodevelopment in autism using human stem cell-derived neurons and organoids: An outlook into future diagnostics and drug development. Dev. Dyn. 2020, 249, 6–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Panisi, C.; Guerini, F.R.; Abruzzo, P.M.; Balzola, F.; Biava, P.M.; Bolotta, A.; Brunero, M.; Burgio, E.; Chiara, A.; Clerici, M.; et al. Autism Spectrum Disorder from the womb to adulthood: Suggestions for a paradigm shift. J. Pers. Med. 2021, 11, 70. [Google Scholar] [CrossRef]
  88. Liu, Q.; Chen, M.X.; Sun, L.; Wallis, C.U.; Zhou, J.S.; Ao, L.J.; Li, Q.; Sham, P.C. Rational use of mesenchymal stem cells in the treatment of autism spectrum disorders. World J. Stem Cells 2019, 11, 55. [Google Scholar] [CrossRef]
  89. Lombardo, M.V.; Moon, H.M.; Su, J.; Palmer, T.D.; Courchesne, E.; Pramparo, T. Maternal immune activation dysregulation of the fetal brain transcriptome and relevance to the pathophysiology of autism spectrum disorder. Mol. Psychiatry 2018, 23, 1001–1013. [Google Scholar] [CrossRef]
  90. Möhrle, D.; Fernández, M.; Peñagarikano, O.; Frick, A.; Allman, B.; Schmid, S. What we can learn from a genetic rodent model about autism. Neurosci. Biobehav. Rev. 2020, 109, 29–53. [Google Scholar] [CrossRef]
  91. Verma, V.; Paul, A.; Amrapali Vishwanath, A.; Vaidya, B.; Clement, J.P. Understanding intellectual disability and Autism Spectrum Disorders from common mouse models: Synapses to behaviour. Open Biol. 2019, 9, 180265. [Google Scholar] [CrossRef]
  92. Nakai, N.; Takumi, T.; Nakai, J.; Sato, M. Common defects of spine dynamics and circuit function in neurodevelopmental disorders: A systematic review of findings from in vivo optical imaging of mouse models. Front. Neurosci. 2018, 12, 412. [Google Scholar] [CrossRef]
  93. Sungur, A.Ö.; Schwarting, R.K.; Woehr, M. Behavioral phenotypes and neurobiological mechanisms in the Shank1 mouse model for Autism Spectrum Disorder: A translational perspective. Behav. Brain Res. 2018, 352, 46–61. [Google Scholar] [CrossRef]
  94. Wang, X.; Kery, R.; Xiong, Q. Synaptopathology in Autism Spectrum Disorders: Complex effects of synaptic genes on neural circuits. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2018, 84, 398–415. [Google Scholar] [CrossRef] [PubMed]
  95. Soler, J.; Fañanás, L.; Parellada, M.; Krebs, M.O.; Rouleau, G.A.; Fatjó-Vilas, M. Genetic variability in scaffolding proteins and risk for schizophrenia and autism-spectrum disorders: A systematic review. J. Psychiatry Neurosci. 2018, 43, 223–244. [Google Scholar] [CrossRef] [PubMed]
  96. Consorthium, T.D.B.F.X.; Bakker, C.E.; Verheij, C.; Willemsen, R.; van der Helm, R.; Oerlemans, F.; Vermey, M.; Bygrave, A.; Hoogeveen, A.; Oostra, B.A.; et al. Fmr1 knockout mice: A model to study fragile X mental retardation. Cell 1994, 78, 23–33. [Google Scholar]
  97. Bagni, C.; Zukin, R.S. A synaptic perspective of fragile X syndrome and autism spectrum disorders. Neuron 2019, 101, 1070–1088. [Google Scholar] [CrossRef] [Green Version]
  98. Kaufmann, W.E.; Kidd, S.A.; Andrews, H.F.; Budimirovic, D.B.; Esler, A.; Haas-Givler, B.; Stackhouse, T.; Riley, C.; Peacock, G.; Sherman, S.L.; et al. Autism Spectrum Disorder in fragile X syndrome: Cooccurring conditions and current treatment. Pediatrics 2017, 139, S194–S206. [Google Scholar] [CrossRef]
  99. Clifford, S.; Dissanayake, C.; Bui, Q.M.; Huggins, R.; Taylor, A.K.; Loesch, D.Z. Autism spectrum phenotype in males and females with fragile X full mutation and premutation. J. Autism Dev. Disord. 2007, 37, 738–747. [Google Scholar] [CrossRef]
  100. Verkerk, A.J.; Pieretti, M.; Sutcliffe, J.S.; Fu, Y.H.; Kuhl, D.P.; Pizzuti, A.; Reiner, O.; Richards, S.; Victoria, M.F.; Zhang, F.; et al. Identification of a gene (FMR-1) containing a CGG repeat coincident with a breakpoint cluster region exhibiting length variation in fragile X syndrome. Cell 1991, 65, 905–914. [Google Scholar] [CrossRef]
  101. Gibson, J.R.; Bartley, A.F.; Hays, S.A.; Huber, K.M. Imbalance of neocortical excitation and inhibition and altered UP states reflect network hyperexcitability in the mouse model of fragile X syndrome. J. Neurophysiol. 2008, 100, 2615–2626. [Google Scholar] [CrossRef]
  102. Yoo, T.; Cho, H.; Park, H.; Lee, J.; Kim, E. Shank3 exons 14–16 deletion in glutamatergic neurons leads to social and repetitive behavioral deficits associated with increased cortical layer 2/3 neuronal excitability. Front. Cell. Neurosci. 2019, 13, 458. [Google Scholar] [CrossRef]
  103. Mossa, A.; Giona, F.; Pagano, J.; Sala, C.; Verpelli, C. SHANK genes in autism: Defining therapeutic targets. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2018, 84, 416–423. [Google Scholar] [CrossRef]
  104. Yoo, T.; Cho, H.; Lee, J.; Park, H.; Yoo, Y.E.; Yang, E.; Kim, J.Y.; Kim, H.; Kim, E. GABA neuronal deletion of Shank3 exons 14–16 in mice suppresses striatal excitatory synaptic input and induces social and locomotor abnormalities. Front. Cell. Neurosci. 2018, 12, 341. [Google Scholar] [CrossRef] [PubMed]
  105. Monteiro, P.; Feng, G. SHANK proteins: Roles at the synapse and in Autism Spectrum Disorder. Nat. Rev. Neurosci. 2017, 18, 147–157. [Google Scholar] [CrossRef] [PubMed]
  106. Chaudry, S.; Vasudevan, N. mTOR-dependent spine dynamics in autism. Front. Mol. Neurosci. 2022, 15, 877609. [Google Scholar] [CrossRef]
  107. Ali Rodriguez, R.; Joya, C.; Hines, R.M. Common ribs of inhibitory synaptic dysfunction in the umbrella of neurodevelopmental disorders. Front. Mol. Neurosci. 2018, 11, 132. [Google Scholar] [CrossRef] [Green Version]
  108. St. Clair, D.; Johnstone, M. Using mouse transgenic and human stem cell technologies to model genetic mutations associated with schizophrenia and autism. Philos. Trans. R. Soc. Biol. Sci. 2018, 373, 20170037. [Google Scholar] [CrossRef] [PubMed]
  109. Lord, C.; Brugha, T.S.; Charman, T.; Cusack, J.; Dumas, G.; Frazier, T.; Jones, E.J.; Jones, R.M.; Pickles, A.; State, M.W.; et al. Autism Spectrum Disorder. Nat. Rev. Dis. Prim. 2020, 6, 1–23. [Google Scholar] [CrossRef]
  110. Vallortigara, G. Born Knowing: Imprinting and the Origins of Knowledge; MIT Press: Cambridge, MA, USA, 2021. [Google Scholar]
  111. Tartaglione, A.M.; Schiavi, S.; Calamandrei, G.; Trezza, V. Prenatal valproate in rodents as a tool to understand the neural underpinnings of social dysfunctions in Autism Spectrum Disorder. Neuropharmacology 2019, 159, 107477. [Google Scholar] [CrossRef]
  112. Fink, J.J.; Levine, E.S. Uncovering true cellular phenotypes: Using induced pluripotent stem cell-derived neurons to study early insults in neurodevelopmental disorders. Front. Neurol. 2018, 9, 237. [Google Scholar] [CrossRef]
  113. Maussion, G.; Rocha, C.; Bernard, G.; Beitel, L.K.; Durcan, T.M. Patient-derived stem cells, another in vitro model, or the missing link toward novel therapies for Autism Spectrum Disorders? Front. Pediatr. 2019, 7, 225. [Google Scholar] [CrossRef]
  114. Sato, A.; Kotajima-Murakami, H.; Tanaka, M.; Katoh, Y.; Ikeda, K. Influence of prenatal drug exposure, maternal inflammation, and parental aging on the development of Autism Spectrum Disorder. Front. Psychiatry 2022, 13, 821455. [Google Scholar] [CrossRef]
  115. Ilieva, M.; Svenningsen, Å.F.; Thorsen, M.; Michel, T.M. Psychiatry in a dish: Stem cells and brain organoids modeling autism spectrum disorders. Biol. Psychiatry 2018, 83, 558–568. [Google Scholar] [CrossRef] [PubMed]
  116. Chan, W.K.; Griffiths, R.; Price, D.J.; Mason, J.O. Cerebral organoids as tools to identify the developmental roots of autism. Mol. Autism 2020, 11, 1–14. [Google Scholar] [CrossRef] [PubMed]
  117. Baldassari, S.; Musante, I.; Iacomino, M.; Zara, F.; Salpietro, V.; Scudieri, P. Brain organoids as model systems for genetic neurodevelopmental disorders. Front. Cell Dev. Biol. 2020, 8, 590119. [Google Scholar] [CrossRef] [PubMed]
  118. Shou, Y.; Liang, F.; Xu, S.; Li, X. The application of brain organoids: From neuronal development to neurological diseases. Front. Cell Dev. Biol. 2020, 8, 579659. [Google Scholar] [CrossRef]
  119. de Jong, J.O.; Llapashtica, C.; Genestine, M.; Strauss, K.; Provenzano, F.; Sun, Y.; Zhu, H.; Cortese, G.P.; Brundu, F.; Brigatti, K.W.; et al. Cortical overgrowth in a preclinical forebrain organoid model of CNTNAP2-associated Autism Spectrum Disorder. Nat. Commun. 2021, 12, 1–14. [Google Scholar] [CrossRef]
  120. Sun, N.; Meng, X.; Liu, Y.; Song, D.; Jiang, C.; Cai, J. Applications of brain organoids in neurodevelopment and neurological diseases. J. Biomed. Sci. 2021, 28, 1–16. [Google Scholar] [CrossRef]
  121. Silverman, J.L.; Thurm, A.; Ethridge, S.B.; Soller, M.M.; Petkova, S.P.; Abel, T.; Bauman, M.D.; Brodkin, E.S.; Harony-Nicolas, H.; Wöhr, M.; et al. Reconsidering animal models used to study Autism Spectrum Disorder: Current state and optimizing future. Genes Brain Behav. 2022, 21, e12803. [Google Scholar] [CrossRef]
  122. Tordjman, S.; Drapier, D.; Bonnot, O.; Graignic, R.; Fortes, S.; Cohen, D.; Millet, B.; Laurent, C.; Roubertoux, P.L. Animal models relevant to schizophrenia and autism: Validity and limitations. Behav. Genet. 2007, 37, 61–78. [Google Scholar] [CrossRef]
  123. Siniscalco, D.; Sapone, A.; Cirillo, A.; Giordano, C.; Maione, S.; Antonucci, N. Autism Spectrum Disorders: Is mesenchymal stem cell personalized therapy the future? J. Biomed. Biotechnol. 2012, 2012, 480289. [Google Scholar] [CrossRef]
  124. Filice, F.; Janickova, L.; Henzi, T.; Bilella, A.; Schwaller, B. The parvalbumin hypothesis of Autism Spectrum Disorder. Front. Cell. Neurosci. 2020, 14, 577525. [Google Scholar] [CrossRef]
  125. Napolitano, A.; Schiavi, S.; La Rosa, P.; Rossi-Espagnet, M.C.; Petrillo, S.; Bottino, F.; Tagliente, E.; Longo, D.; Lupi, E.; Casula, L.; et al. Sex Differences in Autism Spectrum Disorder: Diagnostic, Neurobiological, and Behavioral Features. Front. Psychiatry 2022, 13, 889636. [Google Scholar] [CrossRef] [PubMed]
  126. Courchesne, E.; Gazestani, V.H.; Lewis, N.E. Prenatal origins of ASD: The when, what, and how of ASD development. Trends Neurosci. 2020, 43, 326–342. [Google Scholar] [CrossRef] [PubMed]
  127. Hoffmann, A.; Ziller, M.; Spengler, D. Childhood-onset schizophrenia: Insights from induced pluripotent stem cells. Int. J. Mol. Sci. 2018, 19, 3829. [Google Scholar] [CrossRef] [PubMed]
  128. Hong, S.J.; Vogelstein, J.T.; Gozzi, A.; Bernhardt, B.C.; Yeo, B.T.; Milham, M.P.; Di Martino, A. Toward neurosubtypes in autism. Biol. Psychiatry 2020, 88, 111–128. [Google Scholar] [CrossRef] [Green Version]
  129. Niu, W.; Parent, J.M. Modeling genetic epilepsies in a dish. Dev. Dyn. 2020, 249, 56–75. [Google Scholar] [CrossRef]
  130. Fetit, R.; Hillary, R.F.; Price, D.J.; Lawrie, S.M. The neuropathology of autism: A systematic review of post-mortem studies of autism and related disorders. Neurosci. Biobehav. Rev. 2021, 129, 35–62. [Google Scholar] [CrossRef]
  131. Griesi-Oliveira, K.; Fogo, M.; Pinto, B.; Alves, A.; Suzuki, A.; Morales, A.; Ezquina, S.; Sosa, O.; Sutton, G.; Sunaga-Franze, D.; et al. Transcriptome of iPSC-derived neuronal cells reveals a module of co-expressed genes consistently associated with Autism Spectrum Disorder. Mol. Psychiatry 2021, 26, 1589–1605. [Google Scholar] [CrossRef]
  132. Al-Dewik, N.; Al-Jurf, R.; Styles, M.; Tahtamouni, S.; Alsharshani, D.; Alsharshani, M.; Ahmad, A.I.; Khattab, A.; Al Rifai, H.; Walid Qoronfleh, M. Overview and introduction to Autism Spectrum Disorder (ASD). In Personalized Food Intervention and Therapy for Autism Spectrum Disorder Management; Springer: Cham, Switzerland, 2020; pp. 3–42. [Google Scholar]
  133. Breen, M.S.; Browne, A.; Hoffman, G.E.; Stathopoulos, S.; Brennand, K.; Buxbaum, J.D.; Drapeau, E. Transcriptional signatures of participant-derived neural progenitor cells and neurons implicate altered Wnt signaling in Phelan-McDermid syndrome and autism. Mol. Autism 2020, 11, 53. [Google Scholar] [CrossRef]
  134. Seabra, C.M.; Aneichyk, T.; Erdin, S.; Tai, D.J.; De Esch, C.E.; Razaz, P.; An, Y.; Manavalan, P.; Ragavendran, A.; Stortchevoi, A.; et al. Transcriptional consequences of MBD5 disruption in mouse brain and CRISPR-derived neurons. Mol. Autism 2020, 11, 45. [Google Scholar] [CrossRef]
  135. Brusco, A.; Ferrero, G.B. Genomic Architecture of ASD. In Psychopathology in Adolescents and Adults with Autism Spectrum Disorders; Springer: Cham, Switzerland, 2019; pp. 23–34. [Google Scholar]
  136. Rogozin, I.B.; Gertz, E.M.; Baranov, P.V.; Poliakov, E.; Schaffer, A.A. Genome-wide changes in protein translation efficiency are associated with autism. Genome Biol. Evol. 2018, 10, 1902–1919. [Google Scholar] [CrossRef]
  137. Chen, T.; Froehlich, T.; Li, T.; Lu, L. Big data approaches to develop a comprehensive and accurate tool aimed at improving Autism Spectrum Disorder diagnosis and subtype stratification. Libr. Tech. 2020, 38, 819–833. [Google Scholar] [CrossRef]
  138. Nehme, R.; Barrett, L.E. Using human pluripotent stem cell models to study autism in the era of big data. Mol. Autism 2020, 11, 21. [Google Scholar] [CrossRef]
  139. Liu, H.; Barnes, J.; Pedrosa, E.; Herman, N.S.; Salas, F.; Wang, P.; Zheng, D.; Lachman, H.M. Transcriptome analysis of neural progenitor cells derived from Lowe syndrome induced pluripotent stem cells: Identification of candidate genes for the neurodevelopmental and eye manifestations. J. Neurodev. Disord. 2020, 12, 14. [Google Scholar] [CrossRef] [PubMed]
  140. Cantor, R.M.; Lowe, J.K. Genome-wide association studies and neurodevelopment: Autism spectrum disorders and related traits. In Factors Affecting Neurodevelopment; Elsevier: Cham, Switzerland, 2021; pp. 27–37. [Google Scholar]
  141. Culotta, L.; Penzes, P. Exploring the mechanisms underlying excitation/inhibition imbalance in human iPSC-derived models of ASD. Mol. Autism 2020, 11, 32. [Google Scholar] [CrossRef] [PubMed]
  142. Gordon, A.; Geschwind, D.H. Human in vitro models for understanding mechanisms of autism spectrum disorder. Mol. Autism 2020, 11, 26. [Google Scholar] [CrossRef]
  143. Prem, S.; Millonig, J.H.; DiCicco-Bloom, E. Dysregulation of neurite outgrowth and cell migration in autism and other neurodevelopmental disorders. In Neurodevelopmental Disorders; Springer: Cham, Switzerland, 2020; pp. 109–153. [Google Scholar]
  144. Muhle, R.A.; Reed, H.E.; Stratigos, K.A.; Veenstra-VanderWeele, J. The emerging clinical neuroscience of Autism Spectrum Disorder: A review. JAMA Psychiatry 2018, 75, 514–523. [Google Scholar] [CrossRef]
  145. Grabrucker, A. Biometals in Autism Spectrum Disorders; Academic Press: Cambridge, MA, USA, 2020. [Google Scholar]
  146. Saxena, R.; Babadi, M.; Namvarhaghighi, H.; Roullet, F.I. Role of environmental factors and epigenetics in autism spectrum disorders. Prog. Mol. Biol. Transl. Sci. 2020, 173, 35–60. [Google Scholar]
  147. Scuderi, C.; Verkhratsky, A. The role of neuroglia in Autism Spectrum Disorders. Prog. Mol. Biol. Transl. Sci. 2020, 173, 301–330. [Google Scholar]
  148. Zhang, X.; Abdellaoui, A.; Rucker, J.; de Jong, S.; Potash, J.B.; Weissman, M.M.; Shi, J.; Knowles, J.A.; Pato, C.; Pato, M.; et al. Genome-wide burden of rare short deletions is enriched in major depressive disorder in four cohorts. Biol. Psychiatry 2019, 85, 1065–1073. [Google Scholar] [CrossRef]
  149. Campbell, P.D.; Granato, M. Zebrafish as a tool to study schizophrenia-associated copy number variants. Dis. Model. Mech. 2020, 13, dmm043877. [Google Scholar] [CrossRef]
  150. Nehme, R.; Pietiläinen, O.; Artomov, M.; Tegtmeyer, M.; Valakh, V.; Lehtonen, L.; Bell, C.; Singh, T.; Trehan, A.; Sherwood, J.; et al. The 22q11. 2 region regulates presynaptic gene-products linked to schizophrenia. Nat. Commun. 2022, 13, 3690. [Google Scholar] [CrossRef] [PubMed]
  151. Degenhardt, F. Update on the genetic architecture of schizophrenia. Med. Genet. 2020, 32, 19–24. [Google Scholar] [CrossRef]
  152. Strom, N.I.; Grove, J.; Meier, S.M.; Bækvad-Hansen, M.; Nissen, J.B.; Als, T.D.; Halvorsen, M.; Nordentoft, M.; Mortensen, P.B.; Hougaard, D.M.; et al. Polygenic heterogeneity across obsessive-compulsive disorder subgroups defined by a comorbid diagnosis. Front. Genet. 2021, 12, 711624. [Google Scholar] [CrossRef] [PubMed]
  153. Bryois, J.; Skene, N.G.; Hansen, T.F.; Kogelman, L.J.; Watson, H.J.; Liu, Z.; Brueggeman, L.; Breen, G.; Bulik, C.M.; Arenas, E.; et al. Genetic identification of cell types underlying brain complex traits yields insights into the etiology of Parkinson’s disease. Nat. Genet. 2020, 52, 482–493. [Google Scholar] [CrossRef]
  154. Yang, Z.; Wu, H.; Lee, P.H.; Tsetsos, F.; Davis, L.K.; Yu, D.; Lee, S.H.; Dalsgaard, S.; Haavik, J.; Barta, C.; et al. Investigating shared genetic basis across tourette syndrome and comorbid neurodevelopmental disorders along the impulsivity-compulsivity spectrum. Biol. Psychiatry 2021, 90, 317–327. [Google Scholar] [CrossRef]
  155. Warrier, V.; Baron-Cohen, S. Childhood trauma, life-time self-harm, and suicidal behaviour and ideation are associated with polygenic scores for autism. Mol. Psychiatry 2021, 26, 1670–1684. [Google Scholar] [CrossRef] [Green Version]
  156. Bralten, J.; Mota, N.R.; Klemann, C.J.; De Witte, W.; Laing, E.; Collier, D.A.; de Kluiver, H.; Bauduin, S.E.; Arango, C.; Ayuso-Mateos, J.L.; et al. Genetic underpinnings of sociability in the general population. Neuropsychopharmacology 2021, 46, 1627–1634. [Google Scholar] [CrossRef]
  157. Park, C.Y.; Zhou, J.; Wong, A.K.; Chen, K.M.; Theesfeld, C.L.; Darnell, R.B.; Troyanskaya, O.G. Genome-wide landscape of RNA-binding protein target site dysregulation reveals a major impact on psychiatric disorder risk. Nat. Genet. 2021, 53, 166–173. [Google Scholar] [CrossRef]
  158. Jiang, C.C.; Lin, L.S.; Long, S.; Ke, X.Y.; Fukunaga, K.; Lu, Y.M.; Han, F. Signalling pathways in Autism Spectrum Disorder: Mechanisms and therapeutic implications. Signal Transduct. Target. Ther. 2022, 7, 1–36. [Google Scholar] [CrossRef]
  159. Urresti, J.; Zhang, P.; Moran-Losada, P.; Yu, N.K.; Negraes, P.D.; Trujillo, C.A.; Antaki, D.; Amar, M.; Chau, K.; Pramod, A.B.; et al. Cortical organoids model early brain development disrupted by 16p11. 2 copy number variants in autism. Mol. Psychiatry 2021, 26, 7560–7580. [Google Scholar] [CrossRef]
  160. Walker, R.L.; Ramaswami, G.; Hartl, C.; Mancuso, N.; Gandal, M.J.; De La Torre-Ubieta, L.; Pasaniuc, B.; Stein, J.L.; Geschwind, D.H. Genetic control of expression and splicing in developing human brain informs disease mechanisms. Cell 2019, 179, 750–771. [Google Scholar] [CrossRef] [PubMed]
  161. Willsey, H.R.; Willsey, A.J.; Wang, B.; State, M.W. Genomics, convergent neuroscience and progress in understanding Autism Spectrum Disorder. Nat. Rev. Neurosci. 2022, 23, 323–341. [Google Scholar] [CrossRef] [PubMed]
  162. Sullivan, P.F.; Geschwind, D.H. Defining the genetic, genomic, cellular, and diagnostic architectures of psychiatric disorders. Cell 2019, 177, 162–183. [Google Scholar] [CrossRef] [PubMed]
  163. Rees, E.; Owen, M.J. Translating insights from neuropsychiatric genetics and genomics for precision psychiatry. Genome Med. 2020, 12, 1–16. [Google Scholar] [CrossRef]
  164. Mullins, N.; Forstner, A.J.; O’Connell, K.S.; Coombes, B.; Coleman, J.R.; Qiao, Z.; Als, T.D.; Bigdeli, T.B.; Børte, S.; Bryois, J.; et al. Genome-wide association study of more than 40,000 bipolar disorder cases provides new insights into the underlying biology. Nat. Genet. 2021, 53, 817–829. [Google Scholar] [CrossRef]
  165. Rujeedawa, T.; Zaman, S.H. The Diagnosis and Management of Autism Spectrum Disorder (ASD) in Adult Females in the Presence or Absence of an Intellectual Disability. Int. J. Environ. Res. Public Health 2022, 19, 1315. [Google Scholar] [CrossRef]
  166. Ruigrok, A.N.; Lai, M.C. Sex/gender differences in neurology and psychiatry: Autism. Handb. Clin. Neurol. 2020, 175, 283–297. [Google Scholar]
  167. Lombardo, M.V.; Auyeung, B.; Pramparo, T.; Quartier, A.; Courraud, J.; Holt, R.J.; Waldman, J.; Ruigrok, A.N.; Mooney, N.; Bethlehem, R.A.; et al. Sex-specific impact of prenatal androgens on social brain default mode subsystems. Mol. Psychiatry 2020, 25, 2175–2188. [Google Scholar] [CrossRef]
  168. Nowak, S.; Jacquemont, S. The effects of sex on prevalence and mechanisms underlying neurodevelopmental disorders. In Handbook of Clinical Neurology; Elsevier: Cham, Switzerland, 2020; Volume 173, pp. 327–339. [Google Scholar]
  169. Kallitsounaki, A.; Williams, D.M. Autism Spectrum Disorder and Gender Dysphoria/Incongruence. A systematic Literature Review and Meta-Analysis. J. Autism Dev. Disord. 2022, 1–15. [Google Scholar] [CrossRef]
  170. Loomes, R.; Hull, L.; Mandy, W.P.L. What is the male-to-female ratio in Autism Spectrum Disorder? A systematic review and meta-analysis. J. Am. Acad. Child Adolesc. Psychiatry 2017, 56, 466–474. [Google Scholar] [CrossRef]
  171. Lai, M.C.; Anagnostou, E.; Wiznitzer, M.; Allison, C.; Baron-Cohen, S. Evidence-based support for autistic people across the lifespan: Maximising potential, minimising barriers, and optimising the person–environment fit. Lancet Neurol. 2020, 19, 434–451. [Google Scholar] [CrossRef]
  172. Müller, R.A.; Fishman, I. Brain connectivity and neuroimaging of social networks in autism. Trends Cogn. Sci. 2018, 22, 1103–1116. [Google Scholar] [CrossRef] [PubMed]
  173. Wilson, H.A.; Creighton, C.; Scharfman, H.; Choleris, E.; MacLusky, N.J. Endocrine insights into the pathophysiology of autism spectrum disorder. Neuroscientist 2021, 27, 650–667. [Google Scholar] [CrossRef] [PubMed]
  174. Howes, O.D.; Rogdaki, M.; Findon, J.L.; Wichers, R.H.; Charman, T.; King, B.H.; Loth, E.; McAlonan, G.M.; McCracken, J.T.; Parr, J.R.; et al. Autism Spectrum Disorder: Consensus guidelines on assessment, treatment and research from the British Association for Psychopharmacology. J. Psychopharmacol. 2018, 32, 3–29. [Google Scholar] [CrossRef]
  175. Yuen, T.; Carter, M.T.; Szatmari, P.; Ungar, W.J. Cost-effectiveness of genome and exome sequencing in children diagnosed with Autism Spectrum Disorder. Appl. Health Econ. Health Policy 2018, 16, 481–493. [Google Scholar] [CrossRef]
  176. Jønch, A.E.; Douard, E.; Moreau, C.; Van Dijck, A.; Passeggeri, M.; Kooy, F.; Puechberty, J.; Campbell, C.; Sanlaville, D.; Lefroy, H.; et al. Estimating the effect size of the 15Q11. 2 BP1–BP2 deletion and its contribution to neurodevelopmental symptoms: Recommendations for practice. J. Med. Genet. 2019, 56, 701–710. [Google Scholar] [CrossRef]
  177. Rosenberg, L.E.; Rosenberg, D.D. Human Genes and Genomes: Science, Health, Society; Academic Press: Cambridge, MA, USA, 2012; Volume 2. [Google Scholar]
  178. Shishido, E.; Aleksic, B.; Ozaki, N. Copy-number variation in the pathogenesis of autism spectrum disorder. Psychiatry Clin. Neurosci. 2014, 68, 85–95. [Google Scholar] [CrossRef]
  179. Egolf, L.E.; Vaksman, Z.; Lopez, G.; Rokita, J.L.; Modi, A.; Basta, P.V.; Hakonarson, H.; Olshan, A.F.; Diskin, S.J. Germline 16p11. 2 microdeletion predisposes to neuroblastoma. Am. J. Hum. Genet. 2019, 105, 658–668. [Google Scholar] [CrossRef]
  180. Lengyel, A.; Pinti, É.; Pikó, H.; Jávorszky, E.; David, D.; Tihanyi, M.; Gönczi, É.; Kiss, E.; Tóth, Z.; Tory, K.; et al. Clinical and genetic findings in Hungarian pediatric patients carrying chromosome 16p copy number variants and a review of the literature. Eur. J. Med. Genet. 2020, 63, 104027. [Google Scholar] [CrossRef]
  181. Bristow, G.C.; Thomson, D.M.; Openshaw, R.L.; Mitchell, E.J.; Pratt, J.A.; Dawson, N.; Morris, B.J. 16p11 Duplication disrupts hippocampal-orbitofrontal-amygdala connectivity, revealing a neural circuit endophenotype for schizophrenia. Cell Rep. 2020, 31, 107536. [Google Scholar] [CrossRef]
  182. Pucilowska, J.; Vithayathil, J.; Pagani, M.; Kelly, C.; Karlo, J.C.; Robol, C.; Morella, I.; Gozzi, A.; Brambilla, R.; Landreth, G.E. Pharmacological inhibition of ERK signaling rescues pathophysiology and behavioral phenotype associated with 16p11. 2 chromosomal deletion in mice. J. Neurosci. 2018, 38, 6640–6652. [Google Scholar] [CrossRef] [PubMed]
  183. Niarchou, M.; Chawner, S.J.; Doherty, J.L.; Maillard, A.M.; Jacquemont, S.; Chung, W.K.; Green-Snyder, L.; Bernier, R.A.; Goin-Kochel, R.P.; Hanson, E.; et al. Psychiatric disorders in children with 16p11. 2 deletion and duplication. Transl. Psychiatry 2019, 9, 1–8. [Google Scholar]
  184. Ip, J.P.; Nagakura, I.; Petravicz, J.; Li, K.; Wiemer, E.A.; Sur, M. Major vault protein, a candidate gene in 16p11. 2 microdeletion syndrome, is required for the homeostatic regulation of visual cortical plasticity. J. Neurosci. 2018, 38, 3890–3900. [Google Scholar] [CrossRef] [PubMed]
  185. Poot, M. Syndromes hidden within the 16p11. 2 deletion region. Mol. Syndromol. 2018, 9, 171–174. [Google Scholar] [CrossRef]
  186. Rein, B.; Yan, Z. 16p11. 2 copy number variations and neurodevelopmental disorders. Trends Neurosci. 2020, 43, 886–901. [Google Scholar] [CrossRef]
  187. Weiss, L.A.; Shen, Y.; Korn, J.M.; Arking, D.E.; Miller, D.T.; Fossdal, R.; Saemundsen, E.; Stefansson, H.; Ferreira, M.A.; Green, T.; et al. Association between microdeletion and microduplication at 16p11. 2 and autism. N. Engl. J. Med. 2008, 358, 667–675. [Google Scholar] [CrossRef] [PubMed]
  188. Marshall, C.R.; Howrigan, D.P.; Merico, D.; Thiruvahindrapuram, B.; Wu, W.; Greer, D.S.; Antaki, D.; Shetty, A.; Holmans, P.A.; Pinto, D.; et al. Contribution of copy number variants to schizophrenia from a genome-wide study of 41,321 subjects. Nat. Genet. 2017, 49, 27–35. [Google Scholar] [CrossRef]
  189. Stefansson, H.; Meyer-Lindenberg, A.; Steinberg, S.; Magnusdottir, B.; Morgen, K.; Arnarsdottir, S.; Bjornsdottir, G.; Walters, G.B.; Jonsdottir, G.A.; Doyle, O.M.; et al. CNVs conferring risk of autism or schizophrenia affect cognition in controls. Nature 2014, 505, 361–366. [Google Scholar] [CrossRef] [Green Version]
  190. Deshpande, A.; Weiss, L.A. Recurrent reciprocal copy number variants: Roles and rules in neurodevelopmental disorders. Dev. Neurobiol. 2018, 78, 519–530. [Google Scholar] [CrossRef]
  191. Takumi, T.; Tamada, K. CNV biology in neurodevelopmental disorders. Curr. Opin. Neurobiol. 2018, 48, 183–192. [Google Scholar] [CrossRef]
  192. Kushima, I.; Aleksic, B.; Nakatochi, M.; Shimamura, T.; Okada, T.; Uno, Y.; Morikawa, M.; Ishizuka, K.; Shiino, T.; Kimura, H.; et al. Comparative analyses of copy-number variation in autism spectrum disorder and schizophrenia reveal etiological overlap and biological insights. Cell Rep. 2018, 24, 2838–2856. [Google Scholar] [CrossRef] [PubMed]
  193. Zwaigenbaum, L.; Duku, E.; Fombonne, E.; Szatmari, P.; Smith, I.M.; Bryson, S.E.; Mirenda, P.; Vaillancourt, T.; Volden, J.; Georgiades, S.; et al. Developmental functioning and symptom severity influence age of diagnosis in Canadian preschool children with autism. Paediatr. Child Health 2019, 24, e57–e65. [Google Scholar] [CrossRef] [PubMed]
  194. Atherton, G.; Edisbury, E.; Piovesan, A.; Cross, L. Autism through the ages: A mixed methods approach to understanding how age and age of diagnosis affect quality of life. J. Autism Dev. Disord. 2022, 52, 3639–3654. [Google Scholar] [CrossRef]
  195. Pender, R.; Fearon, P.; Heron, J.; Mandy, W. The longitudinal heterogeneity of autistic traits: A systematic review. Res. Autism Spectr. Disord. 2020, 79, 101671. [Google Scholar] [CrossRef]
  196. Kirst, S.; Diehm, R.; Bögl, K.; Wilde-Etzold, S.; Bach, C.; Noterdaeme, M.; Poustka, L.; Ziegler, M.; Dziobek, I. Fostering socio-emotional competencies in children on the autism spectrum using a parent-assisted serious game: A multicenter randomized controlled trial. Behav. Res. Ther. 2022, 152, 104068. [Google Scholar] [CrossRef] [PubMed]
  197. Rozenblatt-Perkal, Y.; Zaidman-Zait, A. Mother–child interaction in families of children with autism: Interpersonal dyadic processes. Res. Autism Spectr. Disord. 2020, 79, 101689. [Google Scholar] [CrossRef]
  198. Reinhartsen, D.; Tapia, A.; Watson, L.; Crais, E.; Bradley, C.; Fairchild, J.; Herring, A.; Daniels, J. Expressive dominant versus receptive dominant language patterns in young children: Findings from the study to explore early development. J. Autism Dev. Disord. 2019, 49, 2447–2460. [Google Scholar] [CrossRef]
  199. Belcher, H.L.; Morein-Zamir, S.; Stagg, S.D.; Ford, R.M. Shining a Light on a Hidden Population: Social Functioning and Mental Health in Women Reporting Autistic Traits But Lacking Diagnosis. J. Autism Dev. Disord. 2022, 1–15. [Google Scholar] [CrossRef]
  200. White, R.; Livingston, L.A.; Taylor, E.C.; Close, S.A.; Shah, P.; Callan, M.J. Understanding the contributions of trait autism and anxiety to extreme demand avoidance in the adult general population. J. Autism Dev. Disord. 2022, 1–9. [Google Scholar] [CrossRef]
  201. Brown, C.M.; Stokes, M.A. Intersection of eating disorders and the female profile of autism. Psychiatr. Clin. 2020, 43, 735–743. [Google Scholar]
  202. Simonoff, E.; Pickles, A.; Charman, T.; Chandler, S.; Loucas, T.; Baird, G. Psychiatric disorders in children with Autism Spectrum Disorders: Prevalence, comorbidity, and associated factors in a population-derived sample. J. Am. Acad. Child Adolesc. Psychiatry 2008, 47, 921–929. [Google Scholar] [CrossRef] [PubMed]
  203. Barros, F.; Figueiredo, C.; Costa, A.; Soares, S.C. Sensory processing in the Autism Spectrum: The role of attention to detail and somatic trait anxiety in the olfactory perception of the general population. J. Autism Dev. Disord. 2021, 51, 2338–2353. [Google Scholar] [CrossRef] [PubMed]
  204. Lacroix, A.; Nalborczyk, L.; Dutheil, F.; Kovarski, K.; Chokron, S.; Garrido, M.; Gomot, M.; Mermillod, M. High spatial frequency filtered primes hastens happy faces categorization in autistic adults. Brain Cogn. 2021, 155, 105811. [Google Scholar] [CrossRef] [PubMed]
  205. Hollin, G. From the profound to the mundane: Questionnaires as emerging technologies in autism genetics. Sci. Technol. Hum. Values 2019, 44, 634–659. [Google Scholar] [CrossRef]
  206. Nebel, M.B.; Lidstone, D.E.; Wang, L.; Benkeser, D.; Mostofsky, S.H.; Risk, B.B. Accounting for motion in resting-state fMRI: What part of the spectrum are we characterizing in Autism Spectrum Disorder? NeuroImage 2022, 257, 119296. [Google Scholar] [CrossRef]
  207. McCracken, J.T.; Anagnostou, E.; Arango, C.; Dawson, G.; Farchione, T.; Mantua, V.; McPartland, J.; Murphy, D.; Pandina, G.; Veenstra-VanderWeele, J.; et al. Drug development for Autism Spectrum Disorder (ASD): Progress, challenges, and future directions. Eur. Neuropsychopharmacol. 2021, 48, 3–31. [Google Scholar] [CrossRef]
  208. Hakami, T. Efficacy and tolerability of antiseizure drugs. Ther. Adv. Neurol. Disord. 2021, 14, 17562864211037430. [Google Scholar] [CrossRef]
  209. Hakami, T. Neuropharmacology of antiseizure drugs. Neuropsychopharmacol. Rep. 2021, 41, 336–351. [Google Scholar] [CrossRef]
  210. Frye, R.E.; Casanova, M.F.; Fatemi, S.H.; Folsom, T.D.; Reutiman, T.J.; Brown, G.L.; Edelson, S.M.; Slattery, J.C.; Adams, J.B. Neuropathological mechanisms of seizures in Autism Spectrum Disorder. Front. Neurosci. 2016, 10, 192. [Google Scholar] [CrossRef] [Green Version]
  211. Hawkins, N.A.; Misra, S.N.; Jurado, M.; Kang, S.K.; Vierra, N.C.; Nguyen, K.; Wren, L.; George, A.L., Jr.; Trimmer, J.S.; Kearney, J.A. Epilepsy and neurobehavioral abnormalities in mice with a dominant-negative KCNB1 pathogenic variant. Neurobiol. Dis. 2021, 147, 105141. [Google Scholar] [CrossRef]
  212. De Maria, B.; Balestrini, S.; Mei, D.; Melani, F.; Pellacani, S.; Pisano, T.; Rosati, A.; Scaturro, G.M.; Giordano, L.; Cantalupo, G.; et al. Expanding the genetic and phenotypic spectrum of CHD2-related disease: From early neurodevelopmental disorders to adult-onset epilepsy. Am. J. Med. Genet. Part A 2022, 188, 522–533. [Google Scholar] [CrossRef]
  213. Cali, E.; Rocca, C.; Salpietro, V.; Houlden, H. Epileptic Phenotypes Associated with SNAREs and Related Synaptic Vesicle Exocytosis Machinery. Front. Neurol. 2021, 12, 806506. [Google Scholar] [CrossRef] [PubMed]
  214. Galanopoulou, A.S.; Löscher, W.; Lubbers, L.; O’Brien, T.J.; Staley, K.; Vezzani, A.; D’Ambrosio, R.; White, H.S.; Sontheimer, H.; Wolf, J.A.; et al. Antiepileptogenesis and disease modification: Progress, challenges, and the path forward—Report of the Preclinical Working Group of the 2018 NINDS-sponsored antiepileptogenesis and disease modification workshop. Epilepsia Open 2021, 6, 276–296. [Google Scholar] [CrossRef] [PubMed]
  215. Raga, S.; Specchio, N.; Rheims, S.; Wilmshurst, J.M. Developmental and epileptic encephalopathies: Recognition and approaches to care. Epileptic Disord. 2021, 23, 40–52. [Google Scholar] [CrossRef] [PubMed]
  216. Scheffer, I.E.; Berkovic, S.; Capovilla, G.; Connolly, M.B.; French, J.; Guilhoto, L.; Hirsch, E.; Jain, S.; Mathern, G.W.; Moshé, S.L.; et al. ILAE classification of the epilepsies: Position paper of the ILAE Commission for Classification and Terminology. Epilepsia 2017, 58, 512–521. [Google Scholar] [CrossRef]
  217. Fisher, R.S.; Cross, J.H.; French, J.A.; Higurashi, N.; Hirsch, E.; Jansen, F.E.; Lagae, L.; Moshé, S.L.; Peltola, J.; Roulet Perez, E.; et al. Operational classification of seizure types by the International League Against Epilepsy: Position Paper of the ILAE Commission for Classification and Terminology. Epilepsia 2017, 58, 522–530. [Google Scholar] [CrossRef]
  218. Fisher, R.S.; Acevedo, C.; Arzimanoglou, A.; Bogacz, A.; Cross, J.H.; Elger, C.E.; Engel, J., Jr.; Forsgren, L.; French, J.A.; Glynn, M.; et al. ILAE official report: A practical clinical definition of epilepsy. Epilepsia 2014, 55, 475–482. [Google Scholar] [CrossRef]
  219. Stamberger, H.; Hammer, T.B.; Gardella, E.; Vlaskamp, D.R.; Bertelsen, B.; Mandelstam, S.; de Lange, I.; Zhang, J.; Myers, C.T.; Fenger, C.; et al. NEXMIF encephalopathy: An X-linked disorder with male and female phenotypic patterns. Genet. Med. 2021, 23, 363–373. [Google Scholar] [CrossRef]
  220. Fan, H.C.; Lee, H.F.; Chi, C.S. SCN8A encephalopathy: Case report and literature review. Neurol. Int. 2021, 13, 143–150. [Google Scholar] [CrossRef]
  221. Crawford, K.; Xian, J.; Helbig, K.L.; Galer, P.D.; Parthasarathy, S.; Lewis-Smith, D.; Kaufman, M.C.; Fitch, E.; Ganesan, S.; O’Brien, M.; et al. Computational analysis of 10,860 phenotypic annotations in individuals with SCN2A-related disorders. Genet. Med. 2021, 23, 1263–1272. [Google Scholar] [CrossRef]
  222. Haddaway, N.R.; Collins, A.M.; Coughlin, D.; Kirk, S. The role of Google Scholar in evidence reviews and its applicability to grey literature searching. PLoS ONE 2015, 10, e0138237. [Google Scholar] [CrossRef] [PubMed]
  223. Hicks, D.; Wouters, P.; Waltman, L.; De Rijcke, S.; Rafols, I. Bibliometrics: The Leiden Manifesto for research metrics. Nat. News 2015, 520, 429. [Google Scholar] [CrossRef] [PubMed]
  224. Hens, K.; Peeters, H.; Dierickx, K. Genetic testing and counseling in the case of an autism diagnosis: A caregivers perspective. Eur. J. Med. Genet. 2016, 59, 452–458. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Study flow diagram (adapted from [33]).
Figure 1. Study flow diagram (adapted from [33]).
Genes 13 01646 g001
Figure 2. Network of publications generated through the Document Co-Citation Analysis (DCA) on the literature about the genetics of autism from 2018 to 2022. The major clusters are highlighted and divided by color.
Figure 2. Network of publications generated through the Document Co-Citation Analysis (DCA) on the literature about the genetics of autism from 2018 to 2022. The major clusters are highlighted and divided by color.
Genes 13 01646 g002
Table 1. Details of the 12 major clusters identified with the document co-citation analysis (DCA). Log-Likelihood Ratio (LLR) label is automatically generated by the software.
Table 1. Details of the 12 major clusters identified with the document co-citation analysis (DCA). Log-Likelihood Ratio (LLR) label is automatically generated by the software.
Cluster IDSizeSilhouetteMean Publication YearLLR LabelSuggested Label
02180.8232018Intellectual DisabilityNetworks and Pathways
11580.9202019Gut MicrobiotaGut Microbiota
21320.8502018Mouse ModelFragile X Syndrome
31200.8852018Mutant MiceSHANK1,2,3 Genes
41190.8312018Valproic AcidValproic Acid
51100.9192019Genomic ArchitectureGenomic Architecture
61060.8252019Brain OrganoidBrain Organoid
71020.8932020Psychiatric DisorderPsychiatric Disorder
8720.9052019Sex DifferenceSex Difference
9590.9112018Autism Spectrum DisorderCopy Number Variations (CNVs)
10490.9852019Autistic AdultDevelopmental Perspectives
1441.0002020Antiseizure DrugAntiseizure Drug
Table 2. Identifying characteristics of 20 documents with higher citation burtness metric generated in the document co-citation analysis (DCA).
Table 2. Identifying characteristics of 20 documents with higher citation burtness metric generated in the document co-citation analysis (DCA).
ReferenceCitation BurstnessPublication YearBurst BeginBurst EndDurationBetweenness CentralitySigma
Lord et al. [41]14.35720182020202220.00101.01
Grove et al. [42]9.46220192020202220.01281.13
Iakoucheva et al. [43]8.08020192020202220.00011.00
Sharon et al. [45]7.82720192020202220.00661.05
Ruzzo et al. [46]7.38920192020202220.01001.08
Kim et al. [47]7.17220112018201910.00001.00
Abraham et al. [48]6.81620172020202220.00311.02
Lim et al. [49]6.70220172019202010.00031.00
Yang et al. [50]6.69320122018201910.00131.01
Lee et al. [51]6.31120192020202220.00061.00
Nowakowski et al. [52]6.31120172020202220.00681.04
Velmeshev et al. [53]6.31120192020202220.00131.01
Matta et al. [54]6.31120192020202220.00111.01
Estes and McAllister [55]6.21420152018201910.00201.01
Goines and Ashwood [56]6.05820132020202220.00111.01
Pantelis et al. [57]6.05820142020202220.00041.00
Yuen et al. [58]5.97520152018201910.00081.01
Antoine et al. [59]5.90920192020202220.00741.04
Schafer et al. [60]5.80520192020202220.00311.02
Stahl et al. [61]5.80520192020202220.00191.01
Table 3. Top 10 citing papers in Cluster #0 identified using DCA.
Table 3. Top 10 citing papers in Cluster #0 identified using DCA.
TitleCoverageGlobal Citing Score
Joensuu et al. [62]5737
Gandhi and Lee [67]529
Guang et al. [71]4798
Garcia-Forn et al. [72]458
Hui et al. [65]448
Diaz-Caneja et al. [73]4411
Alonso-Gonzalez et al. [63]4025
DiCarlo and Wallace [74]402
Eyring and Geschwind [66]395
Iakoucheva et al. [43]3994
Table 4. Top 10 citing papers in Cluster #1 identified using DCA.
Table 4. Top 10 citing papers in Cluster #1 identified using DCA.
TitleCoverageGlobal Citing Score
Guang et al. [71]3898
Patel et al. [85]379
Yang and Shcheglovitov [86]3310
Panisi et al. [87]3021
Matta et al. [54]2778
Zheng et al. [75]268
DiCarlo and Wallace [74]252
Liu et al. [88]2511
Lombardo et al. [89]2573
Fattorusso et al. [77]25140
Table 5. Top 10 citing papers in Cluster #2 identified using DCA.
Table 5. Top 10 citing papers in Cluster #2 identified using DCA.
TitleCoverageGlobal Citing Score
Wang et al. [94]648
Verma et al. [91]4623
Gandhi and Lee [67]439
Joensuu et al. [62]4037
Guang et al. [71]3498
Sungur et al. [93]3412
Bagni and Zukin [97]33109
Chaudry and Vasudevan [106]310
Patel et al. [85]319
Möhrle et al. [90]2821
Table 6. Top 10 citing papers in Cluster #3 identified using DCA.
Table 6. Top 10 citing papers in Cluster #3 identified using DCA.
TitleCoverageGlobal Citing Score
Wang et al. [94]548
Soler et al. [95]4026
Mossa et al. [103]3519
Yoo et al. [104]3522
Ali Rodriguez et al. [107]3410
Joensuu et al. [62]3337
Sungur et al. [93]3112
Yoo et al. [102]2915
Yang and Shcheglovitov [86]2910
Verma et al. [91]2923
Table 7. Top 10 citing papers in Cluster #4 identified using DCA.
Table 7. Top 10 citing papers in Cluster #4 identified using DCA.
TitleCoverageGlobal Citing Score
St. Clair and Johnstone [108]2213
Tartaglione et al. [111]1928
Hui et al. [65]188
Filice et al. [124]1718
Rylaarsdam and Guemez-Gamboa [14]16118
Napolitano et al. [125]160
DiCarlo and Wallace [74]142
Fink and Levine [112]1414
Patel et al. [85]149
Nakai et al. [92]1424
Table 8. Top 10 citing papers in Cluster #6 identified using DCA.
Table 8. Top 10 citing papers in Cluster #6 identified using DCA.
TitleCoverageGlobal Citing Score
Lord et al. [109]23211
Courchesne et al. [126]1540
Hoffmann et al. [127]1514
Ilieva et al. [115]1439
Hong et al. [128]1231
Chan et al. [116]1212
Niu and Parent [129]1218
Fetit et al. [130]116
Griesi-Oliveira et al. [131]1022
Hui et al. [65]108
Table 9. Top 10 citing papers in Cluster #5 identified using DCA.
Table 9. Top 10 citing papers in Cluster #5 identified using DCA.
TitleCoverageGlobal Citing Score
Al-Dewik et al. [132]205
Culotta and Penzes [141]1812
Breen et al. [133]1413
Gordon and Geschwind [142]137
Prem et al. [143]137
Muhle et al. [144]1276
Grabrucker [145]142
Saxena et al. [146]125
Scuderi and Verkhratsky [147]118
Fink and Levine [112]1114
Table 10. Top 10 citing papers in Cluster #7 identified using DCA.
Table 10. Top 10 citing papers in Cluster #7 identified using DCA.
TitleCoverageGlobal Citing Score
Lord et al. [41]18211
Park et al. [157]1623
Jiang et al. [158]150
Urresti et al. [159]1211
Walker et al. [160]1262
Willsey et al. [161]121
Hoffmann et al. [127]1214
Sullivan and Geschwind [162]12156
Rees and Owen [163]1228
Mullins et al. [164]1194
Table 11. Top 10 citing papers in Cluster #8 identified using DCA.
Table 11. Top 10 citing papers in Cluster #8 identified using DCA.
TitleCoverageGlobal Citing Score
Rylaarsdam and Guemez-Gamboa [14]11118
Lord et al. [41]10211
Napolitano et al. [125]90
Rujeedawa and Zaman [165]90
Lai et al. [171]853
Kallitsounaki and Williams [169]60
Müller and Fishman [172]647
Wilson et al. [173]67
Howes et al. [174]6105
Yuen et al. [175]612
Table 12. Top 10 citing papers in Cluster #9 identified using DCA.
Table 12. Top 10 citing papers in Cluster #9 identified using DCA.
TitleCoverageGlobal Citing Score
Jønch et al. [176]1119
Egolf et al. [179]1114
Deshpande and Weiss [190]1122
Lengyel et al. [180]101
Takumi and Tamada [191]955
Rylaarsdam and Guemez-Gamboa [14]9118
Kushima et al. [192]8114
Bristow et al. [181]711
Pucilowska et al. [182]739
Campbell and Granato [149]73
Table 13. Top 10 citing papers in Cluster #10 identified using DCA.
Table 13. Top 10 citing papers in Cluster #10 identified using DCA.
TitleCoverageGlobal Citing Score
Zwaigenbaum et al. [193]720
Barros et al. [203]62
Al-Dewik et al. [132]65
Lacroix et al. [204]61
Kirst et al. [196]50
Hollin [205]50
Nebel et al. [206]50
Belcher et al. [199]50
Rozenblatt-Perkal and Zaidman-Zait [197]51
McCracken et al. [207]55
Table 14. Top 10 citing papers in Cluster #14 identified using DCA.
Table 14. Top 10 citing papers in Cluster #14 identified using DCA.
TitleCoverageGlobal Citing Score
Hakami [208]42
Hakami [209]43
Stamberger et al. [219]310
Hawkins et al. [211]35
Fan et al. [220]24
De Maria et al. [212]24
Crawford et al. [221]25
Galanopoulou et al. [214]26
Cali et al. [213]20
Raga et al. [215]26
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Lim, M.; Carollo, A.; Dimitriou, D.; Esposito, G. Recent Developments in Autism Genetic Research: A Scientometric Review from 2018 to 2022. Genes 2022, 13, 1646. https://doi.org/10.3390/genes13091646

AMA Style

Lim M, Carollo A, Dimitriou D, Esposito G. Recent Developments in Autism Genetic Research: A Scientometric Review from 2018 to 2022. Genes. 2022; 13(9):1646. https://doi.org/10.3390/genes13091646

Chicago/Turabian Style

Lim, Mengyu, Alessandro Carollo, Dagmara Dimitriou, and Gianluca Esposito. 2022. "Recent Developments in Autism Genetic Research: A Scientometric Review from 2018 to 2022" Genes 13, no. 9: 1646. https://doi.org/10.3390/genes13091646

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

Article Metrics

Back to TopTop