Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

A systematic review of non-coding RNA genes with differential expression profiles associated with autism spectrum disorders

  • Jon Stott,

    Roles Conceptualization, Data curation, Investigation, Methodology, Project administration, Writing – original draft, Writing – review & editing

    Affiliations Child Oriented Mental Health Intervention Collaborative (COMIC), University of York in Collaboration with Leeds and York Partnership NHS Foundation Trust, York, United Kingdom, Tees, Esk & Wear Valleys NHS Foundation Trust, Foss Park Hospital, York, United Kingdom

  • Thomas Wright,

    Roles Conceptualization, Investigation, Writing – review & editing

    Affiliations Manchester Centre for Genomic Medicine, Clinical Genetics Service, Saint Mary’s Hospital, Manchester University NHS Foundation Trust, Manchester, United Kingdom, Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom

  • Jannah Holmes ,

    Roles Funding acquisition, Investigation, Writing – review & editing

    jannah.holmes@hotmail.com

    Affiliations Child Oriented Mental Health Intervention Collaborative (COMIC), University of York in Collaboration with Leeds and York Partnership NHS Foundation Trust, York, United Kingdom, Hull York Medical School, University of York, Heslington, York, United Kingdom

  • Julie Wilson,

    Roles Formal analysis, Writing – review & editing

    Affiliation Department of Mathematics, University of York, Heslington, York, United Kingdom

  • Sam Griffiths-Jones,

    Roles Writing – review & editing

    Affiliation Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom

  • Deborah Foster,

    Roles Investigation

    Affiliation Tees, Esk & Wear Valleys NHS Foundation Trust, Foss Park Hospital, York, United Kingdom

  • Barry Wright

    Roles Conceptualization, Investigation, Methodology, Supervision, Writing – review & editing

    Affiliations Child Oriented Mental Health Intervention Collaborative (COMIC), University of York in Collaboration with Leeds and York Partnership NHS Foundation Trust, York, United Kingdom, Hull York Medical School, University of York, Heslington, York, United Kingdom

Abstract

Aims

To identify differential expression of shorter non-coding RNA (ncRNA) genes associated with autism spectrum disorders (ASD).

Background

ncRNA are functional molecules that derive from non-translated DNA sequence. The HUGO Gene Nomenclature Committee (HGNC) have approved ncRNA gene classes with alignment to the reference human genome. One subset is microRNA (miRNA), which are highly conserved, short RNA molecules that regulate gene expression by direct post-transcriptional repression of messenger RNA. Several miRNA genes are implicated in the development and regulation of the nervous system. Expression of miRNA genes in ASD cohorts have been examined by multiple research groups. Other shorter classes of ncRNA have been examined less. A comprehensive systematic review examining expression of shorter ncRNA gene classes in ASD is timely to inform the direction of research.

Methods

We extracted data from studies examining ncRNA gene expression in ASD compared with non-ASD controls. We included studies on miRNA, piwi-interacting RNA (piRNA), small NF90 (ILF3) associated RNA (snaR), small nuclear RNA (snRNA), small nucleolar RNA (snoRNA), transfer RNA (tRNA), vault RNA (vtRNA) and Y RNA. The following electronic databases were searched: Cochrane Library, EMBASE, PubMed, Web of Science, PsycINFO, ERIC, AMED and CINAHL for papers published from January 2000 to May 2022. Studies were screened by two independent investigators with a third resolving discrepancies. Data was extracted from eligible papers.

Results

Forty-eight eligible studies were included in our systematic review with the majority examining miRNA gene expression alone. Sixty-four miRNA genes had differential expression in ASD compared to controls as reported in two or more studies, but often in opposing directions. Four miRNA genes had differential expression in the same direction in the same tissue type in at least 3 separate studies. Increased expression was reported in miR-106b-5p, miR-155-5p and miR-146a-5p in blood, post-mortem brain, and across several tissue types, respectively. Decreased expression was reported in miR-328-3p in bloods samples. Seven studies examined differential expression from other classes of ncRNA, including piRNA, snRNA, snoRNA and Y RNA. No individual ncRNA genes were reported in more than one study. Six studies reported differentially expressed snoRNA genes in ASD. A meta-analysis was not possible because of inconsistent methodologies, disparate tissue types examined, and varying forms of data presented.

Conclusion

There is limited but promising evidence associating the expression of certain miRNA genes and ASD, although the studies are of variable methodological quality and the results are largely inconsistent. There is emerging evidence associating differential expression of snoRNA genes in ASD. It is not currently possible to say whether the reports of differential expression in ncRNA may relate to ASD aetiology, a response to shared environmental factors linked to ASD such as sleep and nutrition, other molecular functions, human diversity, or chance findings. To improve our understanding of any potential association, we recommend improved and standardised methodologies and reporting of raw data. Further high-quality research is required to shine a light on possible associations, which may yet yield important information.

Introduction

Autistic people are thought to account for at least 1% of the global population [1]. Individuals with a diagnosis of autism have differences in social communication and are more likely to have intense interests [24]. People with autism belong within a spectrum of neurodiversity that is important for society and evolution [5]. For the purpose of this systematic review we have followed the established international diagnostic criteria and the corresponding nomenclature [6]. From herein we will use the associated terminology, autism spectrum disorder (ASD), although we acknowledge that different perspectives exist regarding language and terminology preferences [79]. The genomic landscape of ASD is complex [10], however a strong genetic aetiology is recognised [11, 12] with twin studies estimating heritability between 70–90% [13, 14]. Access to broad genomic testing is reshaping our understanding of ASD, which appears to encompass a collection of broad, heterogenous [15] and variable conditions with overlapping neurobehavioral phenotypes [16]. These may be considered on one hand as complex or syndromic when ASD symptomatology features alongside intellectual disability, facial dysmorphism or congenital malformations [17]. On the other hand, non-syndromic ASD symptomatology may comprise a broader understanding of neurodiversity [5]. High impact genetic variants are reported to occur in around 15% of individuals with ASD, which are predominantly caused by nuclear sequence-level and structural variants, or less commonly mitochondrial variants [18]. It is important to recognise the variable contribution genetic variants have made towards ASD symptomatology, which frequently demonstrate incomplete penetrance and variable expressivity [19].

Proposed explanations for the high heritability, but low monogenic diagnostic findings in ASD include oligogenic and polygenic models of aetiology [20]. Other proposed genetic aetiologies include the imprinted brain theory where there is a paternal bias in the expression of imprinted genes [21] and epigenetic contribution [22]. Given that most nucleotides in the human genome are outside of open reading frames of protein coding genes [23], yet around 75% of the genome are transcribed [24], this draws our attention inexorably to non-coding RNA transcripts that comprise functional molecules that may play an important role in gene expression and gene-environment interactions in ASD. A good starting point is a synthesis of the ncRNA gene expression literature to delineate further promising avenues of enquiry for ASD research [25].

Classification of non-coding RNA

ncRNA are described in detail elsewhere [26, 27]. They have historically been categorised by size, where long non-coding RNA (lncRNA) are 200 or more nucleotides and short ncRNA are less than 200 nucleotides in length [28]. The terms “short” or “small” however, are being used less to describe ncRNA, and do not feature in the current approved nomenclature [26]. Many ncRNA molecules regulate gene expression via RNA interference, epigenetic modification and inhibition of translation related mechanisms [29]. Secreted extracellular circulating ncRNA are, in many cases, highly stable and detectable in multiple biological fluids such as blood, saliva and urine [30, 31]. There is great interest in developing ncRNA expression assays translatable into a clinical setting that may be capable of supporting ASD diagnostics and providing phenotypic or prognostic information to enhance ASD care [32, 33]. The HUGO Gene Nomenclature Committee (HGNC) have worked with specialist advisors to define the accepted nomenclature for ncRNA [26]. HGNC define nine major classes of ncRNA annotated in the human genome. In this systematic review, we are interested in the shorter classes ncRNA and their relative gene expression in ASD. We are not considering genomic variation within ncRNA genes [34, 35], or the expression of larger ncRNA such as ribosomal RNA [36, 37] or long non-coding RNA (lncRNA) [38, 39]. The shorter ncRNA HGNC approved gene classes included in this systematic review are: microRNA (miRNA), piwi-interacting-RNA (piRNA), small NF90 (ILF3) associated RNA (snaR), small nuclear RNA (snRNA), small nucleolar RNA (snoRNA), transfer RNA (tRNA), vault RNA (vtRNA), and Y RNA. They have been summarised in Table 1. Whilst we acknowledge that HGNC approval is only in place for piRNA gene clusters, given the likely expansion to include individual piRNA genes in the future and given that annotation exists elsewhere [40], they have also been included. For simplicity, we will collectively refer to the shorter ncRNA classes included in this systematic review as ncRNA from herein.

thumbnail
Table 1. Shorter HGNC approved ncRNA gene classes included in this systematic review.

https://doi.org/10.1371/journal.pone.0287131.t001

Rationale for systematic review

A systematic review is warranted for a few key reasons. Firstly, much of the early research examining ncRNA expression profiles in association with ASD examines miRNA alone [41]. We may be missing other important classes of ncRNA. To our knowledge there has been no systematic review exploring gene expression of other ncRNA. Secondly, a large proportion of early research in this field is from post-mortem samples from brain tissue [42, 43]. These are important for discovery but may lack clinical translatability. To realise the potential of ASD ncRNA gene expression assays for biomarker use, we require an appreciation of the combined expression data from living patients with ASD from clinically available samples. To date there have been some narrative, discursive, selective or scoping reviews [25, 42, 4448] and just one recent systematic review that only examines miRNA expression associated with ASD that is missing some studies [41]. Finally, in view of the recent international nomenclature describing ncRNA with HGNC approved human genome annotation [26], we are keen to collate and present up to date and standardised ncRNA gene expression data associated with ASD. We acknowledge that there may be a paucity of evidence for classes of ncRNA other than miRNA, but demonstrating and delineating this clearly by systematic review is important to help shape future research directions.

Methods

PROSPERO registration number: CRD42020208233.

Study eligibility criteria

The inclusion criteria were as follows:

Population: Human subjects with a diagnosis of ASD compared with controls without ASD.

Exposure: ncRNA gene expression profiles from biosamples measuring HGNC approved ncRNA genes or piRNA genes listed in piRBase v3.0.

Outcome(s): Expression profile of any of the following ncRNA genes: miRNA, piRNA, snaR, snRNA, snoRNA, tRNA, vtRNA, and Y RNA; using validated scientific methodologies.

Studies: Peer reviewed publications, conference abstracts or dissertations.

The exclusion criteria were as follows: studies not published in English, duplicated data, non-human studies, review articles, hypothesis papers, narrative reviews, fact sheets and letters to the editor that did not present unique or new data, unpublished materials and studies published before 2000.

Search strategy

The following electronic databases were searched: Cochrane, EMBASE, Science Direct, Medline, PubMed, Scopus, Web of Science, PsychInfo, ERIC, AMED, and CINAHL. We searched databases from January 2000 to May 2022. Medical Subjective Heading (MeSH) search terms were used for autism spectrum conditions including ‘autism’, ‘autistic’, autism spectrum disorder, ‘ASD’, autism spectrum condition (ASC), ‘Asperger’, ‘pervasive developmental disorder’ and ‘PDD’ in all combinations with the terms ‘short non coding RNA’, ‘non-coding RNA’, ‘RNA’, ‘miRNA’, ‘miRNA’, ‘piwi interacting RNA’, ‘piRNA’, ‘ribosomal RNA’, ‘rRNA’, ‘small NF90 associated RNA’, ‘small NF90 (ILF3) associated RNA’, ‘snaRs’, ‘small nuclear RNA’, ‘snRNA’, ‘small nucleolar RNA’, ‘snoRNA’, ‘transfer RNA’, ‘tRNA’, ‘vault RNA’, and ‘Y RNA’. The references cited in identified publications were also searched to locate additional studies. Data related to ncRNA expression profiles was extracted where available, including information related to normalisation strategies, ncRNA gene expression fold change, P values and confidence intervals. Given the varied nomenclature used for ncRNA, gene names will be recorded together with HGNC codes, accession IDs from miRBase database v22.1 (mirbase.org) or piRBase database v3.0 (bigdata.ibp.ac.cn/piRBase).

Procedure

Two reviewers independently screened the titles and abstracts to identify all eligible studies identified by the searches. Any discrepancies were adjudicated by a third reviewer. The reference lists of selected articles were used to identify additional papers for screening. The included studies were assessed using the Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA) guidelines [62]. Data extraction took place and was recorded in a dedicated data extraction form generated using Microsoft Excel for further evaluation of study quality and data synthesis including functional enrichment analysis of the significant differentially expression miRNA genes. Raw data was retrieved from published papers, supplementary materials or by contacting the corresponding authors.

Data synthesis and quality assessment

We planned to perform meta-analysis of ncRNA gene expression using the statistical techniques employed by Zhu and Leung [63], including a random effects model [64] to examine differentially expressed ncRNA genes in ASD compared with controls. We expected between study heterogeneity and subgroup analysis were to be used to explore possible sources, including source of patients, source of control (such as healthy control or disease control), participant ethnicity, ncRNA profile (single ncRNA and multiple ncRNA) and sample specimen (blood, saliva, urine, cultured lymphoblastoid cells, fibroblast cells, neural tissues, and others); living or post-mortem. We planned to analyse the statistical heterogeneity of the meta-analysis by x-squared (x2)-based Q statistic test when I2 (I-squared or I2) exceeded 50% or P < 0.1. Receiver-operating characteristics (ROC) curves were planned to be generated with sensitivity, specificity and positive predictive values based on known assessments of participants with ASD or without ASD. The area under the curve (AUC) was planned to be calculated both overall and for any subgroup analysis. Statistical tests were intended to be two-sided, with P < 0.05 considered statistically significant. Functional enrichment analysis of statistically significant differentially expressed miRNA genes as determined by meta-analysis would be performed using DIANA-miRPath v3.0 [65] and executed using the online DIANA-microT-CDS web-server algorithm to examine Gene Ontology (GO) with ‘categories union’. P-value and microT thresholds would be set at < 0.05 and 0.8, respectively with False Discovery Rate (FDR) correction applied. Targeted pathways and significance clusters will be generated and a related heatmap constructed.

We planned an assessment of publication bias [66] using Egger’s graphical test to construct a funnel plot of all studies included in the meta-analysis and explore the symmetry of the study distribution on the plot [64]. Begg and Mazumdar’s Rank Correlation test would be used to correlate the ranks of effect sizes and the ranks of their variances [67] and Orwin’s Fail-Safe N test would determine the presence of missing studies that may skew the regression line in the funnel plot, with Duval and Tweedie’s Trim and Fill method being used for imputation of the missing studies [68, 69]. The methodological quality of all included studies was assessed by two reviewers independently using a quality assessment template based on Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2) [70].

Results

Studies identified for selection

The systematic review search strategy yielded 5250 publications, with 1221 being duplications. The titles and abstracts of 4029 papers were screened and 168 papers were assessed in full for eligibility. 48 studies were identified for inclusion in the systematic review for data extraction. This process is outlined along with reasons for exclusion in the PRISMA flow chart (Fig 1).

thumbnail
Fig 1. PRISMA flow chart illustrating the process of study selection.

https://doi.org/10.1371/journal.pone.0287131.g001

Summary of eligible studies

This systematic review has brought together the findings of 48 studies involving over 1800 individuals with ASD compared with over 1400 controls. The year of publication ranged from 2008 to 2021. ASD ncRNA gene expression studies have been conducted in numerous countries across the world, including Brazil, Bulgaria, China, Egypt, Iran, Italy, Japan, United Kingdom, and United States of America (USA). The most prolific country for publication was the USA with 12 studies. Considering all included studies, the diagnosis of ASD of study participants in 16 studies reported the use of both a validated assessment tool and Diagnostic and Statistical Manual of Mental Disorders (DSM-5) criteria. There were 14 studies that only reported the use of a validated assessment tool, the most common being the Autism Diagnostic Interview-Revised (ADI-R) and the Autism Diagnostic Observation Schedule (ADOS). Eight studies solely used The World Health Organisation (WHO) or DSM diagnostic criteria without a validated assessment tool and 10 studies did not state the method of ASD diagnosis. The vast majority of studies (N = 46) examined miRNA gene expression; 41 studies did so exclusively and 7 studies examined other classes of ncRNA, of which 5 studies also measured miRNA gene expression (including a genome wide study ncRNA expression study encompassing miRNA genomic loci). Fourteen studies used pre-selected candidate-driven ncRNA expression approaches, for example where specific miRNAs had been investigated, in contrast to 34 studies that investigated unselected or larger populations of ncRNA genes including those examined using genome wide approaches. Many of these studies went on to examine (‘validate’) a selected population of miRNA genes identified by an initial unselected approach such as microarray or from RNA-seq. Thirty-three studies reported ncRNA expression findings using tissue samples and laboratory methodologies that could feasibly be implemented into clinical practice (i.e., those from living individuals, with routine sampling methodology of easily obtainable tissue such as blood or saliva and routine laboratory work). These studies had a male to female ratio of participants of 3.5 to 1. There were 15 studies that exclusively reported findings from studies with less or unfeasible clinical implementation possibilities (i.e., when samples derived from post-mortem brain tissue or studies from living individuals requiring specialist sampling procedures such as biopsies, or those with complex or time-consuming laboratory work such as cell culturing). These studies had a male to female ratio of participants of 4.8 to 1. There were two studies that examined ncRNA expression from both clinically feasible and unfeasible samples.

Characteristics of eligible studies

Table 2 provides a summary of 33 studies describing methods feasible for clinical implementation. Of these, 29 reported ncRNA gene expression from peripheral blood and 4 reported from saliva samples. We found no studies exploring ncRNA gene expression from other bodily fluids such urine or sweat. Table 3 summarises the studies with less or unfeasible clinical implementation. From these 17 studies, 10 were from post-mortem brain tissue samples, five were from cultured lymphoblastoid cell lines, one was from reprogrammed induced pluripotent stem cell-derived neurons, and a further study reporting both olfactory mucosal stem cells and primary skin fibroblasts [71]. Two of these studies examined ncRNA gene expression from both clinically feasible and unfeasible samples [71, 72], and therefore feature in both Tables 2 and 3. Table 4 provides an overview of the individual ncRNA genes (all of which are miRNA genes) that have been reported to have increased or decreased expression in ASD cohorts in more than one study. The individual miRNA genes are listed with the direction of expression change presented by broad tissue sample types: blood, saliva, cultured lymphoblastoid cells (unless otherwise specified) and post-mortem brain samples. The seven studies examining differential expression of ncRNA genes other than miRNA have been presented in a separate table (Table 5).

thumbnail
Table 2. Studies examining ncRNA gene expression in ASD using tissue samples and laboratory methodologies that could be feasibly implemented into clinical practice (N = 33).

https://doi.org/10.1371/journal.pone.0287131.t002

thumbnail
Table 3. Studies examining ncRNA gene expression in ASD using tissue samples or laboratory methodologies that require complex additional processing or are from deceased persons (N = 17).

https://doi.org/10.1371/journal.pone.0287131.t003

thumbnail
Table 4. Overview of individual miRNA genes with differential expression in ASD reported in two or more studies presented by tissue.

https://doi.org/10.1371/journal.pone.0287131.t004

thumbnail
Table 5. Studies examining differential expression of ncRNA classes other than miRNA in ASD.

https://doi.org/10.1371/journal.pone.0287131.t005

Non-coding RNA with differential expression in ASD

The systematic review revealed 64 miRNA genes with differential expression in more than one study (Table 4). Twenty-nine of these miRNA genes had differential expression in opposing directions. Four miRNA genes had differential expression in the same direction in the same tissue type in at least 3 separate studies. These were in bloods samples for miR-106b-5p [7375] and miR-328-3p [73, 76, 77], which had increased and decreased expression, respectively. The other miRNA gene was miR-155-5p which had increased expression in post-mortem brain samples [7880]. Finally, miR-146a-5p had consistent, increased differential expression across several different tissue types as reported in four studies [71, 8183]. These were from saliva, primary skin fibroblasts, lymphoblastoid cell lines, olfactory mucosal cells and post-mortem brain samples from the pre-fontal cortex and temporal lobe, respectively. Seven research studies examined ncRNA gene expression, other than miRNA, in association with ASD (Table 5). From these studies, differential expression was reported in individual genes from ncRNA classes including: snoRNA [43, 73, 8487], snRNA [84, 88], piRNA [73] and Y RNA genes [73]. Differential expression of one or more snoRNA genes was reported by six studies, but no individual snoRNA gene or other individual ncRNA genes (excluding miRNA genes) had differential expression reported in more than one study.

Data synthesis and meta-analysis

Functional enrichment analysis using DIANA-miRPath v3.0 online interface [65] was performed with interrogation of the four key miRNA genes identified in this review (miR-106b-5p, miR-328-3p, miR-146a-5p and miR-155-5p) versus Gene Ontology (GO) categories. Clustering with the highest enrichment significance levels were seen in ‘ion binding’ and ‘organelle function’ GO categories, which can be visualised within the heatmap generated (S1 Fig). For the planned meta-analysis, we extracted all available ncRNA expression data from each study. Data for most studies was incomplete for meta-analysis, therefore we contacted corresponding authors, but were only able to obtain raw datasets from a small number of studies. Considering all included studies, a range of data elements were used to capture ncRNA gene differential expression. Only 17 papers included both fold change and associated statistical findings [43, 73, 75, 77, 78, 8082, 85, 90, 101103, 110, 113, 114, 117] and often the latter was not corrected for multiple testing. The different data types, levels of data processing and in many instances inaccessible data, made meta-analysis unsuitable [118]. Many papers only reported p-values for miRNAs that were found to be differentially expressed (i.e. did not report those with non-significant expression). Although methods to combine p-values have been proposed [119], we found that the use of different statistical tests, different hypotheses (one-sided or two-sided) and any adjustment for multiple comparisons often being unknown, made this inappropriate. Originally plans were made for a series of statistical and publication bias analytical assessments as part of a meta-analysis, but these were not possible [64, 68, 69]. We added a field into the quality assessment related to statistical analysis given the complexities of analysing complex data sets and multiple testing in the included studies [120]. The quality assessment using adapted QUADAS-2 [70] is shown in S1 Table.

Discussion

We consider here our findings from 46 studies that examined miRNA gene expression and 7 studies examining other classes of ncRNA.

Differential expression of miRNA genes in ASD

Several miRNA genes have been reported to have differential expression in two or more studies (Table 4). Whilst this initially appears promising, many of these are in opposing directions. This may relate to tissue specificity of miRNA gene expression [71], type I errors related to high numbers of miRNA genes tested and/or statistical tests being performed [121] or reflect the heterogenous nature of ASD aetiology or the study populations examined [15, 122]. Further issues around study quality and bias are considered later in the discussion. Only four miRNAs had differential expression in the same direction and tissue type in at least three studies: miR-106b-5p, miR-146a-5p, miR-155-5p and miR-328-3p. Intriguingly, in addition to the studies in our systematic review, a further single case study examining genome-wide differential miRNA gene expression from the post-mortem prefrontal cortex of a single deceased individual with ASD compared with a non-ASD sibling control without ASD (i.e. ASD of N = 1) also found miR-106b-5p and miR-146a-5p were in their top six differentially expressed miRNA genes [123]. It is instructive to consider the 4 notable miRNA genes identified in our systematic review in more detail, although caution should be exercised given the possibility of selective research and/or reporting and high levels of potential bias found from our quality assessments.

Four notable miRNA genes with differential expression in ASD

miR-106b-5p.

It has previously been reported that miR-106b-5p has altered expression in schizophrenia [124]. The finding that ASD and childhood onset schizophrenia both share altered expression is under research scrutiny [125], although both these groups also have high associated rates of pathogenic copy number variants and brain trauma [126] and there is a long history of some diagnostic overlap [127]. miR-106b-5p has a wide influence on various biological processes including cancer [128, 129] and in isolation is unlikely to demonstrate disease specificity.

miR-146a-5.

miR-146a-5p was found to have uniformly increased expression in our systematic review across a wide range of tissue types including saliva, primary skin fibroblasts, lymphoblastoid cell line, olfactory mucosal cells and post-mortem brain samples from the prefrontal cortex and temporal lobe, respectively [71, 78, 8183]. One of these studies examined tissue and disease specificity of miR-146a-5p (with three other miRNA genes) and found no differential expression in peripheral blood mononuclear cells (PBMC) from a group of ASD patients compared to controls [71]. miR-146a-5p has also been implicated in a number of biological processes including regulation of the development of viral infections [130] and cancer tumour suppression [131], for example in the inhibition of both EGFR and NF-kB signalling and reduction of the metastatic potential of cancers [132].

miR-155-5p.

miR-155-5p showed a degree of uniformity in our systematic review, with increased expression in the amygdala, prefrontal cortex and temporal cortex regions in three post-mortem studies [7880] but with no significant differential expression found in dorsolateral prefrontal cortex [79]. miR-155-5p has been implicated in inflammatory processes [133, 134] and the modulation of cancer [135]. miR-155-5p expression appears to be involved in impaired development of dendritic cells, B cells and T cells and is important for immune response [136, 137]. Moreover, it was one of several differentially expressed miRNA genes associated with a basket of neurodegenerative diseases, including idiopathic Parkinson’s disease, where miR-155-5p has been reported to have increased expression [138].

miR-328-3p.

In our systematic review, miR-328-3p was found to have decreased expression in peripheral blood samples in three studies examining serum [76, 77] and plasma [73], respectively [73, 76, 77] but a further study reported increased expression in peripheral blood [102]. miR-328-3p has been thought to have a role in cancer, whereby suppression is believed to impair stem cell function, a mechanism hypothesised to prevent ovarian cancer metastasis [139].

Functional enrichment analysis.

The output of functional enrichment analysis by DIANA-miRPath v3.0 [65] with the four key miRNA genes identified in this systematic review (miR-106b-5p, miR-146a-5p, miR-155-5p and miR-328-3p) versus gene ontology categories identified the most significant levels of enrichment in ‘ion binding’ and ‘organelle function’ GO categories (S1 Fig). Ion binding is an interesting finding, given the theories of channelopathy dysregulation in the pathogenesis of ASD [140143]. However, there are well articulated concerns related to the cautious interpretation of functional enrichment and pathway analysis of miRNA that have been raised within the miRNA research community [144147]. For example, there have been suggestions that the results from standard analyses are biased by over-represented terms and may suffer from ascertainment bias for the most studied molecular pathways and be limited by selective coverage of annotated genes within a gene set [144]. Some solutions to these challenges have been proposed [144, 145, 147] but are beyond the scope of this review.

Other ncRNA with differential expression in ASD

Whilst the majority of papers identified in this systematic review examined miRNA gene expression, other ncRNA genes with differential expression were reported in seven papers including differential expression of snoRNA [43, 73, 8486], snRNA [84, 88], piRNA [73] and Y RNA genes [73] (Table 5). One of these studies Salloum-Asfar and colleagues (2021) [73] was the first to report stable expression of piRNA, snoRNA, Y RNA and tRNA genes in plasma, a helpful attribute for further research. Two of the seven papers were published by the same research group [84, 88], and described overlapping ASD ncRNA ‘diagnostic signatures’ that derived from re-annotation and analysis of expression data from an external dataset with validation using recruited participants. Together these two studies described nine snRNA genes [84], one snoRNA gene [88] and one Y RNA pseudogene in overlapping ncRNA expression diagnostic models measured in blood (Table 5). Unfortunately the corresponding raw data, strength and direction of expression change, and how each ncRNA gene contributed to their ‘signature formula’ models were not clearly reported [84, 88]. The small number of ncRNA gene expression studies in cohorts of individuals with a diagnosis of ASD is in itself an important finding to report, to help shape future research directions, given their cellular mechanisms and theoretical links with ASD. Each ncRNA class with reports of differential expression in ASD found in our review, have been discussed further, in turn.

Small nucleolar RNA.

Six studies examined differential expression in snoRNA genes in ASD. snoRNA can be divided into three major classes: C/D box snoRNAs (SNORDs), H/ACA box snoRNAs (SNORAs) and small Cajal body‐specific RNAs (scaRNAs) (Table 1). snoRNAs accumulate in the nucleoli of the cell and have roles in post-transcriptional modification and maturation of ribosomal RNA and snRNA [56, 85, 148, 149] and roles in mRNA processing and splicing [150]. There is interest in snoRNA splicing disruption affecting neuronal development and function [151153]. snoRNA have been associated with a range of human diseases [154] including ASD, and are gathering interest [43, 73, 8486]. Differentially methylated genomic regions of paternal sperm samples have been associated with ASD-related phenotype at 12 months of age [22]. The paternal sperm genomic loci region exhibiting differential methylation in this study contains fifteen snoRNA genes within the SNORD-115 cluster, which lies within the Prader-Willi syndrome critical region on chromosome 15. Prader-Willi syndrome is an imprinting condition that can manifest with a neurobehavioral phenotype with aspects of ASD symptomatology [155].

Small nuclear RNA.

Most snRNA are involved in the major and minor spliceosome complex to splice the introns from pre-messenger RNA [53]. snRNA and the related core spliceosomal U-snRNP complexes are associated with numerous diseases including those with neurological manifestations such as spinal muscular atrophy (SMA), amyotrophic lateral sclerosis and Burn‐McKeown syndrome [156159]. Some authors have proposed an association of with snRNA with ASD [84, 88, 160], including Zhou and colleagues (2019), identified in this review, who report an ASD-ncRNA ‘diagnostic signature’ in blood comprising entirely of snRNA genes [88].

Piwi-interacting RNA.

piRNA are frequently considered with miRNA, given their comparable size, and overlapping molecular functions [51]. In contrast to miRNA, piRNA are predominantly expressed in germline cells and function to silence transposable elements and regulate gene expression through RNA cleavage and methylation mechanisms. The role of piRNA is increasingly being described in somatic cells, such as in the nervous system and they have been implicated in neurodevelopmental and neurodegenerative disorders [161]. Rett syndrome is an X-linked dominant neurodevelopmental condition affecting females caused by pathogenic variants in the MECP2 gene [162]. Rett Syndrome is characterised by developmental regression following a period of apparently normal development, an ASD neurobehavioural phenotype and repetitive hand movements. The MECP2 gene is responsible for binding to methylated genomic DNA and has epigenetic functions required for neuronal development [163]. Interestingly, MECP2 knockout mice have increased piRNA expression profiles in the cerebellum [164]. MECP2 also has roles related to miRNA biogenesis, miRNA binding and lncRNA interactions [163].

Y RNA.

One study identified in this systematic review reported five Y RNA genes (RNY4P36, RNY4P6, RNY4, RNY4P25 and RNY4P18) with decreased plasma expression in ASD compared with controls [73]. The same study reported four other Y RNA genes with differential expression associated with ‘more symptoms’ of ASD, with increased expression of RNY4P29 and decreased expression of RNY3P1, RNY3 and RNY4P28, respectively. Whilst there were no other studies reporting Y RNAs, Cheng and colleagues (2020) included a single Y RNA pseudogene known as RNY1P11 within their ASD ncRNA diagnostic signature in blood [88], but had no HGNC approved Y RNA genes within their model. Y RNAs were first discovered in the serum of people with systemic lupus erythematosus (SLE), a multisystemic autoimmune condition that can involve the brain [165]. Y RNA have cellular roles related to DNA replication, RNA stability and cellular stress responses [59, 60].

Other classes of ncRNA lacking ASD differential expression evidence.

Whilst no differential expression findings were forthcoming from this systematic review in relation to vtRNA, tRNA and snaR, we have highlighted some interesting literature relevant to ASD, worthy of further discussion.

Vault RNA.

vtRNA plays a role in neuronal synapse formation and so are of interest in ASD given postulated aetiologies such as altered neurone development including synapse formation [166]. vtRNA bind to and activate a mitogen-activated protein kinase (MEK) to amplify the RAS-MAPK signalling pathway [167]. There is emerging evidence associating RASopathies (a group of inherited disorders caused by pathogenic variants of genes encoding regulatory proteins within the RAS-MAPK signalling pathway) with an increased prevalence of ASD [168]. One such RASopathy is Legius syndrome, which interestingly has a murine model where the ASD-like neurobehavioral phenotype is ameliorated by MEK inhibitors [169]. Further work related to vtRNA expression in ASD could complement this research to support the possible clinical translation of ASD-related MEK inhibitor drug therapy [169].

Transfer RNA.

tRNA genes are encoded for by both nuclear and mitochondrial genomes. The mitochondrial genome has been proposed as a genetic modifier for ASD [170] and theories related to mitochondrial dysfunction in ASD have been hypothesised [171]. The mitochondrial genome encodes 22 transfer RNA genes and harbours the majority of pathogenic variants that result in broad and disparate disorders [172]. One report demonstrated a mitochondrial tRNA variant within a single family that was attributed as causative for a heterogeneous group of neurological disorders where ASD was a feature [173].

Small NF90 (ILF3) associated RNA.

snaR gene expression may also be worthy of further examination in ASD, given their abundant expression within the testis and discrete regions of the brain [52]. Evidence from meta-analysis reports that advanced paternal age as a risk factor for ASD [174], which may be related to increased rates of genomic and epigenomic abnormalities within the germline cells [175]. It is also interesting that polymorphisms of SNAR-I (one of twenty snaR genes), is associated with increased lateral ventricle volume [176], which is one of two neuroimaging distinguishing features (alongside increased Pallidum volume) found in a large ASD cohort that underwent high-resolution structural brain scans [177].

Limitations and quality assessments of studies

Quality of data and reporting.

There are several limitations that need to be taken seriously both in interpreting the results from this systematic review and in planning for future research. The exact number of ASD participants from all included studies was difficult to ascertain as certain studies were not explicit in descriptions of study populations, and there were occasions where it was difficult to exclude some study population overlap [77, 104, 178]. The use of external datasets and biobank sample resources also made this challenging, with some instances where the same Gene Expression Omnibus (GEO) dataset was used (for example GSE18123 in three studies) [84, 88, 94]. Two of these studies were from one research group that also appeared to use the same internal datasets in both of their studies, but this was not readily apparent in their described methodologies [84, 88]. Most studies use small sample sizes and several studies do not report how the diagnosis of ASD was established (e.g., whether they used validated measures). We identified studies that included participants with ASD present alongside confounding phenotypes for example, individuals with ‘high-functioning’ ASD [102], those who recruited both ASD and control participants from an allergy/immunology clinic [99], and individuals with high levels of consanguinity, epilepsy and dysmorphism [112], that may influence miRNA expression [179181]. Participants were frequently recruited from convenience samples or clinic populations and many studies had a limited description of control groups with few or no assessments to characterise phenotype variations. These factors are further challenged by the heterogeneity of ASD and the use of small sample sizes [15, 182]. We also recognised a large variation in the methods used to determine ncRNA gene expression and many studies omit important methodological details related to these.

Meta-analysis and data synthesis.

Statistical methodological quality in the studies are highly variable with many instances of small sample sizes and studies using inappropriate statistical tests. It is unclear in some studies whether correction for multiple testing has been applied and, where stated, different methods have been used such as Bonferroni or Benjamini-Hochberg correction. For meta-analysis, we considered methods to combine p-values [119], such as Stouffer’s method [183] that is generally preferred when different weights are attributed to the p-values being combined. However, it is not clear how the direction of differential expression (often presented as fold change) should be incorporated. Some authors recommend the removal of genes with conflicting differential expression, so that only the genes with the same fold change are combined [184] and others suggest that one-sided p-values can be used to take the direction of fold-change into account. When not specified, the p-values given are presumably two-sided but one-sided p-values are sometimes reported. We also observed different statistical tests, including t-tests, Mann-Whitney U-tests and Tukey’s multiple comparison tests to provide the p-values. These were often reported as simple inequalities rather than precise values, making it unlikely that useful information could be extracted from their combination. High degrees of heterogeneity were apparent across studies with respect to participants, sample types and expression assays. It is well recognised that different cell types have tissue specific ‘miRNomes’ and comparing this ncRNA expression data therefore might not be appropriate [185]. Despite contacting several authors, we were not able to obtain full data sets in several cases. In summary, our planned strategy for meta-analysis and integration of the findings from different studies was not possible [118] because of the large variation in data presentation, availability, statistical analysis used and many instances of poor reporting.

Factors affecting ncRNA gene expression.

The field of ncRNA gene expression studies is littered with challenges in the interpretation of findings. Disease or developmental states may not be the only factors altering ncRNA expression. Exercise [186], sleep [187], nutritional intake [188, 189] and infection [190] are just some factors that may impact ncRNA expression. Interestingly, sleep [191], nutrition [192], bowel habit [193] and exercise [194] may be markedly different in people with ASD compared to neurotypical people, raising the prospect that ncRNA differential expression findings may be as a result of ASD and its patterns, lifestyles and associations rather than (or as well as) aetiological. This is currently unclear and so research methodologies should attempt to examine and control for this where possible. There are numerous ways that ncRNAs are deployed in biological processes. As in multifactorial models of ASD aetiology [195], the role of ncRNAs may also be multifaceted and interactive.

We also know that sample collection, RNA extraction, purification, storage, handling, and testing conditions can greatly impact ncRNA expression [196198]. For example, the use of an EDTA anticoagulant appears to influence specific miRNA expression, particularly after longer EDTA exposure times [196]. In our systematic review, EDTA blood tubes were used in several studies [73, 75, 76, 89, 90, 93, 101, 105] with only a few studies using PAXgene blood RNA tubes [94, 102, 104, 110] and many studies omitted details about blood sample collection, including anticoagulant exposure timings. Quantity and quality of centrifuging in blood has also been shown to alter the proportion of intra and extracellular components that may demonstrate different miRNA expression properties [197]. Challenges related to ncRNA data normalisation approaches also support the need for standardisation [199]. Caution is also required for the interpretation of post-mortem samples. In life, hypoxia is known to change miRNA function and expression [200] and so it is not surprising that post-mortem miRNAs are altered through the process of death with degradation happening in different ways at different rates [201, 202]. Post-mortem ncRNA gene expression studies therefore need to include supplementary tests to explore degradation to aid interpretation. In summary, the process of measuring ncRNA gene expression requires quality control and clear detailed reporting to allow comparison between studies for meaningful interpretation.

Differential gene expression in opposing directions.

Our review findings of studies reporting miRNA genes with differential expression in opposite directions needs further consideration. Another systematic review in type two diabetes mellitus reported that two thirds of differentially expressed miRNA genes were found in opposite directions [63]. Whilst this may suggest poor methodologies or reporting bias we should be cautious about how we interpret this. Some miRNA genes appear to have greater tissue specificity than others [203]. In the context of cancer, opposing directions of miRNA differential gene expression in miR-125b is thought to represent oncogenic characteristics when expression is increased and loss of tumour suppressive functions when expression is decreased [204]. Differential expression in opposing directions of individual miRNA genes was observed in this systematic review on a population level, but also on an individual level [112]. There is evidence that direction of miRNA (and other ncRNA) differential expression may change with age [86] or over time and may respond to environmental exposures such as smoking [205] and alcohol [206]. Whilst numerous miRNA genes have been associated with neurodevelopmental or neurodegenerative diseases [138] there is still much work to be done to understand whether miRNA differential expression may play a role in aetiology or to the numerous other factors described above including a response to the condition itself.

Expression assays for ncRNA.

Various technologies for measuring ncRNA expression levels have been used in the studies, each with different strengths and limitations [207]. Quantitative polymerase chain reaction assays (qPCR) are based on the amplification of target ncRNA genes of known sequence. Although qPCR assays are known for their high sensitivity and specificity, the sensitivity does depend on the target abundance and the efficiency of the amplification [208]. If there are closely related sequences to the target sequence, there is a risk of false amplification. The many different protocols, reagents, and analysis methods and lack of technical information led to recommendations for qPCR assay design and data reporting, or “minimum information for the publication of qPCR experiments” (MIQE) [209]. qPCR assays can be expensive as each target requires specific primers and probes and they are commonly used to validate gene expression changes identified by other methods, such as microarrays or Next-Generation Sequencing (NGS). Microarrays are cost-effective and have been widely used in ncRNA gene expression research. However, they may not be sensitive enough to detect expression of low-abundance ncRNA genes and can suffer from dynamic range issues which affect the quantification of highly abundant transcripts [210]. Microarray results can also be influenced by probe design bias, as the performance of the probes may vary depending on their sequence. Differences in hybridisation as well as normalisation issues mean that RNA sequencing is sometimes preferred [211]. NGS has revolutionised ncRNA research by allowing comprehensive profiling of ncRNAs. However, biases in library preparation methods, including at ligation, reverse transcription, and amplification steps, and sequencing errors, can all affect the accuracy of ncRNA identification and quantification [212]. Furthermore, NGS generates huge amounts of data, requiring advanced bioinformatics tools and computational resources for data analysis.

Implications for clinical practice.

At the current time there are no implications for clinical practice that we could reliably draw from these results, with limited evidence to support ncRNA gene expression as biomarkers for ASD. The ncRNA genes with differential expression identified in this systematic review have all been implicated in several other diseases and biological processes and there is limited or no reporting of any high sensitivity and/or specificity scores or validation studies. There are also limited descriptions of phenotypes in the ASD groups. There is, however, enough promise to suggest that continuing to research in this field has potential to improve our understanding of mechanisms associated with neurodevelopmental differences such as ASD.

Implications for research.

By contrast there are many implications for research to consider. The finding that there is limited research examining gene expression in classes of ncRNA other than miRNA is important to report. This shines a light on the omission in the research literature. Given that miRNA gene silencing occurs in many tissue types including in the developing brain [213], it is intriguing that four proteins critical for miRNA biogenesis [214] are encoded by genes associated with Mendelian disorders where ASD and overlapping neurobehavioral phenotypes are highly prevalent: DRCG8 (included within the deleted region in chromosome 22q11.2) [215], MECP2 (Rett syndrome) [216], FOXG1 [217] and FMR1 (Fragile X) [218, 219]. As key regulators of gene expression, miRNA may have a role in modifying genetic variants demonstrating incomplete penetrance and variable expressivity [220]. This theory is interesting, considering the multiple examples of recurrent pathogenic CNVs associated with variable ASD risk [19].

Some standardisation is required to overcome the large variability in quality and reporting of ncRNA gene expression in ASD. Improved methodologies and reporting would greatly benefit the research endeavour. Alongside MIQE mentioned above, we recommend researchers work to the FAIR Guiding Principles for scientific data management and stewardship (2016) [221] to improve the findability, accessibility, interoperability, and reusability of ncRNA expression data in ASD and other ncRNA expression studies. This would provide the standardisation and authentication necessary for data to be reusable. Feature level extraction output (FLEO) files have been recommended as published gene lists (PGL data) and gene expression data matrices (GEDMs) have been deemed unsuitable for meta-analysis due to their dependence on the pre-processing used [118]. Sharing research data between research groups comes with challenges [222] and public sharing of raw data in biomedical microarray studies appears to be more likely for studies published in high impact journals and when lead authors are more experienced researchers [223, 224]. The majority of journals and funders now have data sharing policies. National and international data protection laws restrict data sharing by genomic researchers but a number of initiatives have been developed to promote successful data sharing including those hosted by the European Molecular Biology Laboratory’s European Bioinformatics Institute [225], the International Cancer Genome Consortium’s project [226], the Pan-Cancer Analysis of Whole Genomes (PCAWG) [227] and the Human Cell Atlas [228]. The researchers involved in setting up PCAWG have called for an international code of conduct to overcome issues with data protection and provide guidelines for researchers [229].

Conclusion

The search for discrete genetic, immunological, metabolic, neurological/neurophysiological and behavioural associations with ASD continues [32]. Differential expression of ncRNA genes have shown much promise in various conditions and may be playing a role in the multifactorial aetiology of ASD. At present, no clear conclusions can be drawn from this systematic review for implementation into clinical practice. The key recommendations from our study are to improve research methodologies, reporting and data sharing in this field and to fund and deliver larger studies with more power that will increase the likelihood of being able to answer important questions.

Supporting information

S1 Fig. Gene Ontology analysis heatmap using four most notable differentially expressed miRNA genes in ASD identified by this systematic review.

DIANA-miRPath v3.0 online interface DIANA-microT-CDS was used to perform analysis of Gene Ontology Categories (x axis) versus the four key miRNA genes identified in this systematic review (miR-106b-5p, miR-328-3p, miR-146a-5p and miR-155-5p) (y axis). P-value and microT threshold were set at < 0.05 and 0.8, respectively and False Discovery Rate (FDR) applied. The heatmap shows the levels of enrichment as determined by Log(p values).

https://doi.org/10.1371/journal.pone.0287131.s001

(TIF)

S1 Table. Quality assessment using adapted QUADAS-2.

https://doi.org/10.1371/journal.pone.0287131.s002

(DOCX)

S2 Table. Overview of ncRNA gene expression profiles in ASD from all included studies.

https://doi.org/10.1371/journal.pone.0287131.s003

(XLSX)

Acknowledgments

We thank David Brown from the Academic Liaison Information Services Team at the University of York for helpful guidance related to the library databases and the search strategy.

References

  1. 1. Elsabbagh M, Divan G, Koh YJ, Kim YS, Kauchali S, Marcín C, et al. Global Prevalence of Autism and Other Pervasive Developmental Disorders. Autism Res. 2012;5: 160–179. pmid:22495912
  2. 2. American Psychiatric Association. Diagnostic and statistical manual of mental disorders. Washington, DC: American Psychiatric Association; 2013.
  3. 3. Lord C, Risi S, Lambrecht L, Cook EH, Leventhal BL, DiLavore PC, et al. The Autism Diagnostic Observation Schedule—Generic: A standard measure of social and communication deficits associated with the spectrum of autism. J Autism Dev Disord. 2000;30: 205–223. pmid:11055457
  4. 4. Scott FJ, Baron-Cohen S. Imagining Real and Unreal Things: Evidence of a Dissociation in Autism. J Cogn Neurosci. 1996;8: 371–382. pmid:23971507
  5. 5. Wright B, Spikins P, Pearson H. Should Autism Spectrum Conditions Be Characterised in a More Positive Way in Our Modern World? Medicina; 2020. pmid:32413984
  6. 6. Volkmar FR. Categorical approaches to the diagnosis of autism: An overview of DSM-IV and ICD-10. Autism. 1998;2: 45–59.
  7. 7. Vivanti G. Ask the Editor: What is the Most Appropriate Way to Talk About Individuals with a Diagnosis of Autism? J Autism Dev Disord. 2020;50: 691–693. pmid:31676917
  8. 8. Bury SM, Jellett R, Spoor JR, Hedley D. “It Defines Who I Am” or “It’s Something I Have”: What Language Do [Autistic] Australian Adults [on the Autism Spectrum] Prefer? J Autism Dev Disord. 2023;53: 677–687. pmid:32112234
  9. 9. Botha M, Hanlon J, Williams GL. Does Language Matter? Identity-First Versus Person-First Language Use in Autism Research: A Response to Vivanti. J Autism Dev Disord. 2023;53: 870–878. pmid:33474662
  10. 10. Qiu S, Qiu Y, Li Y. Genetics of autism spectrum disorder: an umbrella review of systematic reviews and meta-analyses. Transl Psychiatry. 2022;12. pmid:35705542
  11. 11. Huguet G, Benabou M, Bourgeron T. The Genetics of Autism Spectrum Disorders. Sassone-Corsi P, Christen Y, editors. Cham: Springer; 2016. https://doi.org/10.1007/978-3-319-27069-2_11
  12. 12. Tick B, Bolton P, Happé F, Rutter M, Rijsdijk F. Heritability of autism spectrum disorders: a meta‐analysis of twin studies. J Child Psychol Psychiatry. 2016;57: 585–595. pmid:26709141
  13. 13. Basu SN, Kollu R, Banerjee-Basu S. AutDB: a gene reference resource for autism research. Nucleic Acids Res. 2009;37: 832–6. pmid:19015121
  14. 14. Genovese A, Butler MG. Clinical assessment, genetics, and treatment approaches in autism spectrum disorder (ASD. Int J Mol Sci. 2020;21: 4726. pmid:32630718
  15. 15. Mottron L, Bzdok D. Autism spectrum heterogeneity: fact or artifact? Mol Psychiatry. 2020;25: 3178–85. pmid:32355335
  16. 16. Ziats CA, Patterson WG, Friez M. Syndromic Autism Revisited: Review of the Literature and Lessons Learned. Pediatric Neurology. 2021. pp. 21–25. pmid:33189026
  17. 17. Warrier V, Zhang X, Reed P, Havdahl A, Moore TM, Cliquet F, et al. Genetic correlates of phenotypic heterogeneity in autism. Nat Genet. 2022;54: 1293–1304. pmid:35654973
  18. 18. Trost B, Thiruvahindrapuram B, Chan AJS, Engchuan W, Higginbotham EJ, Howe JL, et al. Genomic architecture of autism from comprehensive whole-genome sequence annotation. Cell. 2022;185: 4409–4427.e18. pmid:36368308
  19. 19. Vicari S, Napoli E, Cordeddu V, Menghini D, Alesi V, Loddo S, et al. Copy number variants in autism spectrum disorders. Progress in Neuro-Psychopharmacology and Biological Psychiatry. 2019. pp. 421–427. pmid:30797015
  20. 20. Wang T, Zhao PA, Eichler EE. Rare variants and the oligogenic architecture of autism. Trends Genet. 2022;38: 895–903. pmid:35410794
  21. 21. Badcock C. The imprinted brain: how genes set the balance between autism and psychosis. Epigenomics. 2011;3: 345–359. pmid:22122342
  22. 22. Feinberg JI, Bakulski KM, Jaffe AE, Tryggvadottir R, Brown SC, Goldman LR, et al. Paternal sperm DNA methylation associated with early signs of autism risk in an autism-enriched cohort. Int J Epidemiol. 2015;44: 1199–1210. pmid:25878217
  23. 23. Aspden JL, Wallace EWJ, Whiffin N. Not all exons are protein coding: Addressing a common misconception. Cell Genomics. 2023;3: 100296. pmid:37082142
  24. 24. Djebali S, Davis CA, Merkel A, Dobin A, Lassmann T, Mortazavi A, et al. Landscape of transcription in human cells. Nature. 2012;489: 101–108. pmid:22955620
  25. 25. Tonacci A, Bagnato G, Pandolfo G, Billeci L, Sansone F, Conte R, et al. miRNA Cross-Involvement in Autism Spectrum Disorders and Atopic Dermatitis: A Literature Review. J Clin Med. 2019;8: 88. pmid:30646527
  26. 26. Seal RL, Chen L, Griffiths‐Jones S, Lowe TM, Mathews MB, O’Reilly D, et al. A guide to naming human non‐coding RNA genes. EMBO J. 2020;39: 103777. pmid:32090359
  27. 27. Watson CN, Belli A, Di Pietro V. Small Non-coding RNAs: New Class of Biomarkers and Potential Therapeutic Targets in Neurodegenerative Disease. Front Genet. 2019;10: 364. pmid:31080456
  28. 28. Nagano T, Fraser P. No-Nonsense Functions for Long Noncoding RNA. Cell. 2011;145: 178–181. pmid:21496640
  29. 29. Sperling R. Small non-coding RNA within the endogenous spliceosome and alternative splicing regulation. Biochim Biophys Acta BBA—Gene Regul Mech. 2019;1862: 194406. pmid:31323432
  30. 30. Kim S, Jeon OH, Jeon Y-J. Extracellular RNA: Emerging roles in cancer cell communication and biomarkers. Cancer Lett. 2020;95: 33–40. pmid:32916182
  31. 31. O’Brien J, Hayder H, Zayed Y, Peng C. Overview of miRNA Biogenesis, Mechanisms of Actions, and Circulation. Front Endocrinol. 2018;9: 402. pmid:30123182
  32. 32. Frye RE, Vassall S, Kaur G, Lewis C, Karim M, Rossignol D. Emerging biomarkers in autism spectrum disorder: a systematic review. Ann Transl Med. 2019;7: 792–792. pmid:32042808
  33. 33. Chen Q, Yan M, Cao Z, Li X, Zhang Y, Shi J, et al. Sperm tsRNAs contribute to intergenerational inheritance of an acquired metabolic disorder. Science. 2016;351: 397–400. pmid:26721680
  34. 34. Borges-Monroy R, Chu C, Dias C, Choi J, Lee S, Gao Y, et al. Whole-genome analysis reveals the contribution of non-coding de novo transposon insertions to autism spectrum disorder. Mob DNA. 2021;12. pmid:34838103
  35. 35. Brandler WM, Antaki D, Gujral M, Kleiber ML, Whitney J, Maile MS, et al. Paternally inherited cis-regulatory structural variants are associated with autism. Science. 2018;360: 327–331. pmid:29674594
  36. 36. Lombardo MV. Ribosomal protein genes in post-mortem cortical tissue and iPSC-derived neural progenitor cells are commonly upregulated in expression in autism. Mol Psychiatry. 2020;26: 1432–1435. pmid:32404943
  37. 37. Porokhovnik L. Individual Copy Number of Ribosomal Genes as a Factor of Mental Retardation and Autism Risk and Severity. Cells. 2019;8: 1151. pmid:31561466
  38. 38. Francoeur N, Gandal M, Xu X, Sarpong K, Johnson J, Sklar P, et al. ASSESSING THE ROLE OF LONG NONCODING RNA (LNCRNA) IN AUTISM SPECTRUM DISORDERS. Eur Neuropsychopharmacol. 2019;29: 960.
  39. 39. Tang J, Yu Y, Yang W. Long noncoding RNA and its contribution to autism spectrum disorders. CNS Neurosci Ther. 2017;23: 645–656. pmid:28635106
  40. 40. Wang J, Shi Y, Zhou H, Zhang P, Song T, Ying Z, et al. piRBase: integrating piRNA annotation in all aspects. Nucleic Acids Res. 2022;50: D265–D272. pmid:34871445
  41. 41. Huang ZX, Chen Y, Guo HR, Chen GF. Systematic Review and Bioinformatic Analysis of microRNA Expression in Autism Spectrum Disorder Identifies Pathways Associated With Cancer, Metabolism. Cell Signal Cell Adhes Front Psychiatry. 2021;12: 630876. pmid:34744804
  42. 42. Hicks SD, Middleton FA. A Comparative Review of miRNA Expression Patterns in Autism Spectrum Disorder. Front Psychiatry. 2016;7: 176. pmid:27867363
  43. 43. Wright C, Shin JH, Rajpurohit A, Deep-Soboslay A, Collado-Torres L, Brandon NJ, et al. Altered expression of histamine signaling genes in autism spectrum disorder. Transl Psychiatry. 2017;7: 1126–1126. pmid:28485729
  44. 44. Fregeac J, Colleaux L, Nguyen LS. The emerging roles of miRNA in autism spectrum disorders. Neurosci Biobehav Rev. 2016;71: 729–738. pmid:27793596
  45. 45. Geaghan M, Cairns MJ. miRNA and posttranscriptional dysregulation in psychiatry. Biol Psychiatry. 2015;78: 231–239. pmid:25636176
  46. 46. Konečná B, Radošinská J, Keményová P, Repiská G. Detection of disease-associated microRNAs—application for autism spectrum disorders. Rev Neurosci. 2020;31: 757–769. pmid:32813679
  47. 47. Salloum-Asfar S, Satheesh NJ, Abdulla SA. Circulating miRNAs, Small but Promising Biomarkers for Autism Spectrum Disorder. Front Mol Neurosci. 2019;12: 253. pmid:31680857
  48. 48. Vasu MM, Sumitha PS, Rahna P, Thanseem I, Anitha A. microRNAs in Autism Spectrum Disorders. Curr Pharm Des. 2020;25: 4368–4378. pmid:31692427
  49. 49. Kozomara A, Birgaoanu M, Griffiths-Jones S. miRBase: from microRNA sequences to function. Nucleic Acids Res. 2018;47: 155–162. pmid:30423142
  50. 50. Bartel DP. Metazoan miRNA. Cell. 2018;173: 20–51. pmid:29570994
  51. 51. Ozata DM, Gainetdinov I, Zoch A, O’Carroll D, Zamore PD. PIWI-interacting RNAs: small RNAs with big functions. Nat Rev Genet. 2019;20: 89–108. pmid:30446728
  52. 52. Parrott AM, Tsai M, Batchu P, Ryan K, Ozer HL, Tian B, et al. The evolution and expression of the snaR family of small non-coding RNA. Nucleic Acids Res. 2011;39: 1485–1500. pmid:20935053
  53. 53. Karijolich J, Yu Y-T. Spliceosomal snRNA modifications and their function. RNA Biol. 2010;7: 192–204. pmid:20215871
  54. 54. Ma J, Zhang L, Chen S, Liu H. A brief review of RNA modification related database resources. Methods. 2022;203: 342–353. pmid:33705860
  55. 55. Lestrade L. snoRNA-LBME-db, a comprehensive database of human H/ACA and C/D box snoRNAs. Nucleic Acids Res. 2006;34: D158–D162. pmid:16381836
  56. 56. Bratkovič T, Božič J, Rogelj B. Functional diversity of small nucleolar RNA. Nucleic Acids Res. 2019;48: 1627–1651. pmid:31828325
  57. 57. Chan PP, Lowe TM. GtRNAdb 2.0: an expanded database of transfer RNA genes identified in complete and draft genomes. Nucleic Acids Res. 2015;44: 184–189. pmid:26673694
  58. 58. Büscher M, Horos R, Hentze MW. ‘High vault-age’: non-coding RNA control of autophagy. Open Biol. 2020;10: 190307. pmid:32070232
  59. 59. Kowalski MP, Krude T. Functional roles of non-coding Y RNA. Int J Biochem Cell Biol. 2015;66: 20–29. pmid:26159929
  60. 60. Valkov N, Das S. YRNA Biogenesis, Function and Implications for the Cardiovascular System. Adv Exp Med Biol. 2020;1229: 327–342. pmid:32285422
  61. 61. HUGO Gene Nomenclature Commitee. HUGO Gene Nomenclature Committee Statistics and Download files. HGNC; 2023. Available: https://www.genenames.org/download/statistics-and-files/
  62. 62. Moher D, Liberati A, Tetzlaff J, Altman DG. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. BMJ. 2009;339: 2535. pmid:19622551
  63. 63. Zhu H, Leung S. Identification of Potential miRNA Biomarkers by Meta-analysis. In: Gore M, Jagtap U, editors. Methods in Molecular Biology. New York: Springer; 2018. pp. 473–484. https://doi.org/10.1007/978-1-4939-7756-7_24 pmid:29594787
  64. 64. Hedges LV, Vevea JL. Fixed- and random-effects models in meta-analysis. Psychol Methods. 1998;3: 486–504.
  65. 65. Vlachos IS, Zagganas K, Paraskevopoulou MD, Georgakilas G, Karagkouni D, Vergoulis T, et al. DIANA-miRPath v3. 0: deciphering microRNA function with experimental support. Nucleic Acids Res. 2015; 43 1 460–466. pmid:25977294
  66. 66. Jayaraj R, Kumarasamy C. Systematic review and meta-analysis of cancer studies evaluating diagnostic test accuracy and prognostic values: approaches to improve clinical interpretation of results. Cancer Manag Res. 2018;10: 4669–4670. pmid:30410400
  67. 67. Begg CB, Mazumdar M. Operating Characteristics of a Rank Correlation Test for Publication Bias. Biometrics. 1994;50: 1088. pmid:7786990
  68. 68. Duval S, Tweedie R. Trim and Fill: A Simple Funnel-Plot-Based Method of Testing and Adjusting for Publication Bias in Meta-Analysis. Biometrics. 2020;56: 455–463. pmid:10877304
  69. 69. Orwin RG. A Fail-Safe N for Effect Size in Meta-Analysis. J Educ Stat. 1983;8: 157.
  70. 70. Whiting PF. QUADAS-2: A Revised Tool for the Quality Assessment of Diagnostic Accuracy Studies. Ann Intern Med. 2011;155: 529. pmid:22007046
  71. 71. Nguyen LS, Lepleux M, Makhlouf M, Martin C, Fregeac J, Siquier-Pernet K, et al. Profiling olfactory stem cells from living patients identifies miRNAs relevant for autism pathophysiology. Mol Autism. 2016;7. pmid:26753090
  72. 72. Pagan C, Goubran-Botros H, Delorme R, Benabou M, Lemière N, Murray K, et al. Disruption of melatonin synthesis is associated with impaired 14-3-3 and miR-451 levels in patients with autism spectrum disorders. Sci Rep. 2017;7. pmid:28522826
  73. 73. Salloum-Asfar S, Elsayed AK, Elhag SF, Abdulla SA. Circulating Non-Coding RNAs as a Signature of Autism Spectrum Disorder Symptomatology. Int J Mol Sci. 2021;22. pmid:34207213
  74. 74. Vasu MM, Anitha A, Thanseem I, Suzuki K, Yamada K, Takahashi T, et al. Serum microRNA profiles in children with autism. Mol Autism. 2014;5. pmid:25126405
  75. 75. Yu D, Jiao X, Cao T, Huang F. Serum miRNA expression profiling reveals miR-486-3p may play a significant role in the development of autism by targeting ARID1B. Neuroreport. 2018;29: 1431–1436. pmid:30260819
  76. 76. Kichukova TM, Popov NT, Ivanov IS, Vachev TI. Profiling of circulating serum microRNAs in children with autism spectrum disorder using stem-loop qRT-PCR assay. Folia Med. 2017;59: 43–52. pmid:28384108
  77. 77. Popov NT, Minchev DS, Naydenov MM, Minkov IN, Vachev TI. Investigation of Circulating Serum MicroRNA-328-3p and MicroRNA-3135a Expression as Promising Novel Biomarkers for Autism Spectrum Disorder. Balk J Med Genet. 2018;21: 5–12. pmid:30984518
  78. 78. Mor M, Nardone S, Sams DS, Elliott E. Hypomethylation of miR-142 promoter and upregulation of microRNAs that target the oxytocin receptor gene in the autism prefrontal cortex. Mol Autism. 2015;6. pmid:26273428
  79. 79. Almehmadi KA, Tsilioni I, Theoharides TC. Increased Expression of miR-155p5 in Amygdala of Children With Autism Spectrum Disorder. Autism Res. 2020;13: 18–23. pmid:31502418
  80. 80. Wu YE, Parikshak NN, Belgard TG, Geschwind DH. Genome-wide, integrative analysis implicates microRNA dysregulation in autism spectrum disorder. Nat Neurosci. 2016;19: 1463–1476. pmid:27571009
  81. 81. Ragusa M, Santagati M, Mirabella F, Lauretta G, Cirnigliaro M, Brex D, et al. Potential Associations Among Alteration of Salivary miRNAs. Saliva Microbiome Struct Cogn Impair Autistic Child Int J Mol Sci. 2020;21. pmid:32867322
  82. 82. Nguyen LS, Fregeac J, Bole-Feysot C, Cagnard N, Iyer A, Anink J, et al. Role of miR-146a in neural stem cell differentiation and neural lineage determination: relevance for neurodevelopmental disorders. Mol Autism. 2018;9. pmid:29951184
  83. 83. Talebizadeh Z, Butler MG, Theodoro MF. Feasibility and relevance of examining lymphoblastoid cell lines to study role of microRNAs in autism. Autism Res. 2008;Aug;1(4):240–50. pmid:19360674
  84. 84. Cheng W, Zhou S, Zhou J, Wang X. Identification of a robust non-coding RNA signature in diagnosing autism spectrum disorder by cross-validation of microarray data from peripheral blood samples. Med Baltim. 2020;99. pmid:32176083
  85. 85. Ander BP, Barger N, Stamova B, Sharp FR, Schumann CM. Atypical miRNA expression in temporal cortex associated with dysregulation of immune, cell cycle, and other pathways in autism spectrum disorders. Mol Autism. 2015;6. pmid:26146533
  86. 86. Stamova B, Ander BP, Barger N, Sharp FR, Schumann CM. Specific Regional and Age-Related Small Noncoding RNA Expression Patterns Within Superior Temporal Gyrus of Typical Human Brains Are Less Distinct in Autism Brains. J Child Neurol. 2015;30: 1930–46. pmid:26350727
  87. 87. Gandal MJ, Zhang P, Hadjimichael E, Walker RL, Chen C, Liu S, et al. Transcriptome-wide isoform-level dysregulation in ASD, schizophrenia, and bipolar disorder. Science. 2018;362. pmid:30545856
  88. 88. Zhou J, Hu Q, Wang X, Cheng W, Pan C, Xing X. Development and validation of a novel and robust blood small nuclear RNA signature in diagnosing autism spectrum disorder. Med Baltim. 2019;98. pmid:31702648
  89. 89. Abdelrahman AH. Evaluation of circulating miRNAs and mRNAs expression patterns in autism spectrum disorder. Egypt J Med Hum Genet. 2021;22.
  90. 90. Atwan H, Assarehzadegan MA, Shekarabi M, Jazayeri SM, Barfi S, Shokouhi Shoormasti R, et al. Assessment of miR-181b-5p, miR-23a-3p, BCL-2, and IL-6 in Peripheral Blood Mononuclear Cells of Autistic Patients; Likelihood of Reliable Biomarkers. Iran J Allergy Asthma Immunol. 2020;19: 74–83. pmid:32245323
  91. 91. Cirnigliaro M, Barbagallo C, Gulisano M, Domini CN, Barone R, Barbagallo D, et al. Expression and Regulatory Network Analysis of miR-140-3p, a New Potential Serum Biomarker for Autism Spectrum Disorder. Front Mol Neurosci. 2017;10. pmid:28848387
  92. 92. Cui L, Du W, Xu N, Dong J, Xia B, Ma J, et al. Impact of MicroRNAs in Interaction With Environmental Factors on Autism Spectrum Disorder: An Exploratory Pilot Study. Front Psychiatry. 2021;12. pmid:34675825
  93. 93. Eftekharian MM, Komaki A, Oskooie VK, Namvar A, Taheri M, Ghafouri-Fard S. Assessment of Apoptosis Pathway in Peripheral Blood of Autistic Patients. J Mol Neurosci. 2019;69: 588–596. pmid:31363911
  94. 94. Gao H, Zhong J, Huang Q, Wu X, Mo X, Lu L, et al. Integrated Systems Analysis Explores Dysfunctional Molecular Modules and Regulatory Factors in Children with Autism Spectrum Disorder. J Mol Neurosci. 2021;Feb;71(2):358–368. pmid:32653993
  95. 95. Hicks SD, Ignacio C, Gentile K, Middleton FA. Salivary miRNA profiles identify children with autism spectrum disorder, correlate with adaptive behavior, and implicate ASD candidate genes involved in neurodevelopment. BMC Pediatr. 2016;16. pmid:27105825
  96. 96. Hicks SD, Carpenter RL, Wagner KE, Pauley R, Barros M, Tierney-Aves C, et al. Saliva MicroRNA Differentiates Children With Autism From Peers With Typical and Atypical Development. J Am Acad Child Adolesc Psychiatry. 2020;59: 296–308. pmid:30926572
  97. 97. Hirsch MM, Deckmann I, Fontes-Dutra M, Bauer-Negrini G, Della-Flora Nunes G, Nunes W, et al. Behavioral alterations in autism model induced by valproic acid and translational analysis of circulating microRNA. Food Chem Toxicol. 2018;115: 336–343. pmid:29510222
  98. 98. Huang F, Long Z, Chen Z, Li J, Hu Z, Qiu R. Investigation of Gene Regulatory Networks Associated with Autism Spectrum Disorder Based on MiRNA Expression in China. PLoS ONE. 2015;10: 0129052. pmid:26061495
  99. 99. Jyonouchi H, Geng L, Streck DL, Dermody JJ, Toruner GA. MicroRNA expression changes in association with changes in interleukin-1ß/interleukin10 ratios produced by monocytes in autism spectrum disorders: their association with neuropsychiatric symptoms and comorbid conditions (observational study. J Neuroinflammation. 2017;14. pmid:29178897
  100. 100. Jyonouchi H, Geng L, Toruner GA, Rose S, Bennuri SC, Frye RE. Serum microRNAs in ASD: Association With Monocyte Cytokine Profiles and Mitochondrial Respiration. Front Psychiatry. 2019;10. pmid:31551826
  101. 101. Kichukova T, Petrov V, Popov N, Minchev D, Naimov S, Minkov I, et al. Identification of serum microRNA signatures associated with autism spectrum disorder as promising candidate biomarkers. Heliyon. 2021;7. pmid:34286132
  102. 102. Nakata M, Kimura R, Funabiki Y, Awaya T, Murai T, Hagiwara M. MicroRNA profiling in adults with high-functioning autism spectrum disorder. Mol Brain. 2019;12. pmid:31639010
  103. 103. Ozkul Y, Taheri S, Bayram KK, Sener EF, Mehmetbeyoglu E, Öztop DB, et al. A heritable profile of six miRNAs in autistic patients and mouse models. Sci Rep. 2020;10. pmid:32514154
  104. 104. Popov NT, Minkov IN, Petrova ND, Andreenko EA, Stoyanova VK, Ivanov HY, et al. Evaluation of microRNA‑486‑3p Molecular Signature in Patients with Autism Spectrum Disorder. COMPTES RENDUS Acad Bulg Sci. 2021;74: 852–860.
  105. 105. Popov NT, Petrov VD. Comparative expression analysis of miR-619-5p in serum and PBMCs as a promising candidate biomarker for autism spectrum disorder. Mol Biol. 2021;74: 537–543.
  106. 106. Sehovic E, Spahic L, Smajlovic-Skenderagic L, Pistoljevic N, Dzanko E, Hajdarpasic A. Identification of developmental disorders including autism spectrum disorder using salivary miRNAs in children from Bosnia and Herzegovina. PloS One. 2020;15: e0232351. pmid:32353026
  107. 107. Sell SL, Widen SG, Prough DS, Hellmich HL. Principal component analysis of blood microRNA datasets facilitates diagnosis of diverse diseases. PloS One. 2020;15: e0234185. pmid:32502186
  108. 108. Shen L, Lin Y, Sun Z, Yuan X, Chen L, Shen B. Knowledge-Guided Bioinformatics Model for Identifying Autism Spectrum Disorder Diagnostic MicroRNA Biomarkers. Sci Rep. 2016;6. pmid:28000768
  109. 109. Vaccaro TDS, Sorrentino JM, Salvador S, Veit T, Souza DO, Almeida RF. Alterations in the MicroRNA of the Blood of Autism Spectrum Disorder Patients: Effects on Epigenetic Regulation and Potential Biomarkers. Behav Sci Basel. 2018;8. pmid:30111726
  110. 110. Vachev TI, Minkov IN. Down regulation of miRNA let-7b-3p and let-7d-3p in the peripheral blood of children with autism spectrum disorder. Int J Curr Microbiol Appl Sci. 2013;2: 384–388.
  111. 111. Zamil BM, Ali-Labib R. Evaluation of miR-106a and ADARB1 in autistic children. Gene Rep. 2019;18.
  112. 112. Abu-Elneel K, Liu T, Gazzaniga FS, Nishimura Y, Wall DP, Geschwind DH, et al. Heterogeneous dysregulation of microRNAs across the autism spectrum. Neurogenetics. 2008; 3 153–61. pmid:18563458
  113. 113. Bleazard T. Investigating the role of microRNAs in autism. PhD thesis, The University of Manchester. 2017. Available: https://www.research.manchester.ac.uk/portal/files/86865547/FULL_TEXT.PDF
  114. 114. Frye RE, Rose S, McCullough S, Bennuri SC, Porter-Gill PA, Dweep H, et al. MicroRNA Expression Profiles in Autism Spectrum Disorder: Role for miR-181 in Immunomodulation. J Med. 2021;11. pmid:34575699
  115. 115. Moore D, Meays BM, Madduri LSV, Shahjin F, Chand S, Niu M, et al. Downregulation of an Evolutionary Young miR-1290 in an iPSC-Derived Neural Stem Cell Model of Autism Spectrum Disorder. Stem Cells Int. 2019. pmid:31191687
  116. 116. Sarachana T, Zhou R, Chen G, Manji HK, Hu VW. Investigation of post-transcriptional gene regulatory networks associated with autism spectrum disorders by microRNA expression profiling of lymphoblastoid cell lines. Genome Med. 2010;2. pmid:20374639
  117. 117. Seno MMG, Hu P, Gwadry FG, Pinto D, Marshall CR, Casallo G, et al. Gene and miRNA expression profiles in autism spectrum disorders. Brain Res. 2011;1380: 85–97. pmid:20868653
  118. 118. Ramasamy A, Mondry A, Holmes CC, Altman DG. Key issues in conducting a meta-analysis of gene expression microarray datasets. PLoS Med. 2008;5: e184. pmid:18767902
  119. 119. Heard NA, Rubin-Delanchy P. Choosing between methods of combining-values. Biometrika. 2018;105: 239–246.
  120. 120. Barkhordarian A, Pellionisz P, Dousti M, Lam V, Gleason L, Dousti M, et al. Assessment of risk of bias in translational science. J Transl Med. 2013;11: 1–6. pmid:23927081
  121. 121. Li Y, Ge X, Peng F, Li W, Li JJ. Exaggerated false positives by popular differential expression methods when analyzing human population samples. Genome Biol. 2022;23: 79. pmid:35292087
  122. 122. Jeste SS, Geschwind DH. Disentangling the heterogeneity of autism spectrum disorder through genetic findings. Nat Rev Neurol. 2014;10: 74–81. pmid:24468882
  123. 123. Lin CY, Chang KW, Lin CY, Wu JY, Coon H, Huang PH, et al. Allele-specific expression in a family quartet with autism reveals mono-to-biallelic switch and novel transcriptional processes of autism susceptibility genes. Sci Rep. 2018;8.
  124. 124. Moreau MP, Bruse SE, David-Rus R, Buyske S, Brzustowicz LM. Altered micro-RNA expression profiles in postmortem brain samples from individuals with schizophrenia and bipolar disorder. Biol Psychiatry. 2011;69: 188–193. pmid:21183010
  125. 125. Rapoport J, Chavez A, Greenstein D, Addington A, Gogtay N. Autism spectrum disorders and childhood-onset schizophrenia: clinical and biological contributions to a relation revisited. J Am Acad Child Adolesc Psychiatry. 2009;48: 10–18. pmid:19218893
  126. 126. Fernandez A, Drozd MM, Thümmler S, Dor E, Capovilla M, Askenazy F, et al. Childhood-onset schizophrenia: a systematic overview of its genetic heterogeneity from classical studies to the genomic era. Front Genet. 2019;10: 1137. pmid:31921276
  127. 127. Pina-Camacho L, Parellada M, Kyriakopoulos M. Autism spectrum disorder and schizophrenia: boundaries and uncertainties. BJPsych Adv. 2016;22: 316–324.
  128. 128. Farré PL, Duca RB, Massillo C, Dalton GN, Graña KD, Gardner K, et al. MiR-106b-5p: A master regulator of potential biomarkers for breast cancer aggressiveness and prognosis. Int J Mol Sci. 2021;22. pmid:34681793
  129. 129. Sagar SK. miR-106b as an emerging therapeutic target in cancer. Genes Dis. 2022;9. pmid:35685464
  130. 130. Nahand JS, Karimzadeh MR, Nezamnia M, Fatemipour M, Khatami A, Jamshidi S, et al. The role of miR‐146a in viral infection. IUBMB Life. 2020;72: 343–360. pmid:31889417
  131. 131. Iacona JR, Lutz CS. miR‐146a‐5p: expression, regulation, and functions in cancer. Wiley Interdiscip Rev RNA. 2019;10: e1533. pmid:30895717
  132. 132. Labbaye C, Testa U. The emerging role of MIR-146A in the control of hematopoiesis, immune function and cancer. J Hematol Oncol. 2012;5: 13. pmid:22453030
  133. 133. Wang X, Chen Y, Yuan W, Yao L, Wang S, Jia Z, et al. miRNA-155-5p is a key regulator of allergic inflammation, modulating the epithelial barrier by targeting PKIα. Cell Death Dis. 2019;10: 1–14. pmid:31767859
  134. 134. Pasca S, Jurj A, Petrushev B, Tomuleasa C, Matei D. miRNA-155 implication in M1 polarization and the impact in inflammatory diseases. Front Immunol. 2020;11: 625. pmid:32351507
  135. 135. Chen G, Wang D, Zhao X. miR-155-5p modulates malignant behaviors of hepatocellular carcinoma by directly targeting CTHRC1 and indirectly regulating GSK-3β-involved Wnt/β-catenin signaling. Cancer Cell Int. 2017;17. pmid:29234238
  136. 136. Mahesh G, Biswas R. MicroRNA-155: a master regulator of inflammation. J Interferon Cytokine Res. 2019;39: 321–330. pmid:30998423
  137. 137. Testa U, Pelosi E, Castelli G, Labbaye C. miR-146 and miR-155: two key modulators of immune response and tumor development. Non-Coding RNA MDPI. 2017;3. pmid:29657293
  138. 138. Juźwik CA, Drake SS, Zhang Y, Paradis-Isler N, Sylvester A, Amar-Zifkin A, et al. miRNA dysregulation in neurodegenerative diseases: A systematic review. Prog Neurobiol. 2019;182: 101664. pmid:31356849
  139. 139. Srivastava AK, Banerjee A, Cui T, Han C, Cai S, Liu L, et al. Inhibition of miR-328–3p Impairs Cancer Stem Cell Function and Prevents Metastasis in Ovarian Cancer. Cancer Res. 2019;79: 2314–2326. pmid:30894370
  140. 140. Pinggera A, Mackenroth L, Rump A, Schallner J, Beleggia F, Wollnik B, et al. New gain-of-function mutation shows CACNA1D as recurrently mutated gene in autism spectrum disorders and epilepsy. Hum Mol Genet. 2017;26: 2923–2932. pmid:28472301
  141. 141. Ben-Shalom R, Keeshen CM, Berrios KN, An JY, Sanders SJ, Bender KJ. Opposing Effects on Na V 1.2 Function Underlie Differences Between SCN2A Variants Observed in Individuals With Autism Spectrum Disorder or Infantile Seizures. Biol Psychiatry. 2017;2017;82(3):224–232. pmid:28256214
  142. 142. Guglielmi L. Update on the implication of potassium channels in autism: K+ channel autism spectrum disorder. Front Cell Neurosci. 2015;2015. pmid:25784856
  143. 143. Schmunk G, Gargus JJ. Channelopathy pathogenesis in autism spectrum disorders. Front Genet. 2013;4. pmid:24204377
  144. 144. Ayllon-Benitez A, Bourqui R, Thébault P, Mougin F. GSAn: an alternative to enrichment analysis for annotating gene sets. NAR Genomics Bioinforma. 2020;2: 017. pmid:33575577
  145. 145. Bleazard T, Lamb JA, Griffiths-Jones S. Bias in miRNA functional enrichment analysis. Bioinformatics. 2015;31: 1592–1598. pmid:25609791
  146. 146. Fridrich A, Hazan Y, Moran Y. Too Many False Targets for MicroRNAs: Challenges and Pitfalls in Prediction of miRNA Targets and Their Gene Ontology in Model and Non‐model Organisms. BioEssays. 2019;41: 1800169. pmid:30919506
  147. 147. Godard P, Eyll J. Pathway analysis from lists of miRNA: common pitfalls and alternative strategy. Nucleic Acids Res. 2015;43: 3490–3497. pmid:25800743
  148. 148. Liang J, Wen J, Huang Z, Chen XP, Zhang BX, Chu L. Small Nucleolar RNA: insight into their function in Cancer. Front Oncol. 2019;9: 587. pmid:31338327
  149. 149. Reichow SL, Hamma T, Ferre-D’Amare AR, Varani G. The structure and function of small nucleolar ribonucleoproteins. Nucleic Acids Res. 2007;35: 1452–64. pmid:17284456
  150. 150. Wajahat M, Bracken CP, Orang A. Emerging Functions for snoRNAs and snoRNA-Derived Fragments. Int J Mol Sci. 2021;22: 10193. pmid:34638533
  151. 151. Al-Dewik N, Alsharshani M. New Horizons for Molecular Genetics Diagnostic and Research in Autism Spectrum Disorder. In: Essa MM, Qoronfleh MW, editors. Personalized Food Intervention and Therapy for Autism Spectrum Disorder Management. Cham: Springer International Publishing; 2020. pp. 43–81. https://doi.org/10.1007/978-3-030-30402-7_2 pmid:32006356
  152. 152. Quesnel-Vallières M, Weatheritt RJ, Cordes SP, Blencowe BJ. Autism spectrum disorder: insights into convergent mechanisms from transcriptomics. Nat Rev Genet. 2019;20: 51–63. pmid:30390048
  153. 153. Zhang P, Omanska A, Ander BP, Gandal MJ, Stamova B, Schumann CM. Neuron-specific transcriptomic signatures indicate neuroinflammation and altered neuronal activity in ASD temporal cortex. Proc Natl Acad Sci. 2023;120: e2206758120. pmid:36862688
  154. 154. Girardot M, Cavaille J, Feil R. Small regulatory RNA controlled by genomic imprinting and their contribution to human disease. Epigenetics Off J DNA Methylation Soc. 2012;7: 1341–1348. pmid:23154539
  155. 155. Bennett JA, Germani T, Haqq AM, Zwaigenbaum L. Autism spectrum disorder in Prader-Willi syndrome: A systematic review. Am J Med Genet A. 2015;167: 2936–2944. pmid:26331980
  156. 156. Wood KA, Ellingford JM, Thomas HB, Douzgou S, Beaman GM, Hobson E, et al. Expanding the genotypic spectrum of TXNL4A variants in Burn‐McKeown syndrome. Clin Genet. 2021;101: 255–259. pmid:34713892
  157. 157. Vazquez-Arango P, O’Reilly D. Variant snRNPs: New players within the spliceosome system. RNA Biol. 2018;15: 17–25. pmid:28876172
  158. 158. Vazquez-Arango P, Vowles J, Browne C, Hartfield E, Fernandes HJR, Mandefro B, et al. Variant U1 snRNAs are implicated in human pluripotent stem cell maintenance and neuromuscular disease. Nucleic Acids Res. 2016;44: 10960–10973. pmid:27536002
  159. 159. Gabanella F, Butchbach MER, Saieva L, Carissimi C, Burghes AHM, Pellizzoni L. Ribonucleoprotein Assembly Defects Correlate with Spinal Muscular Atrophy Severity and Preferentially Affect a Subset of Spliceosomal snRNPs. Valcarcel J, editor. PLoS ONE. 2007;2: e921. pmid:17895963
  160. 160. Singh RK, Cooper TA. Pre-mRNA splicing in disease and therapeutics. Trends Mol Med. 2012;18: 472–482. pmid:22819011
  161. 161. Kim KW. PIWI Proteins and piRNA in the Nervous System. Mol Cells. 2019;42: 828–835. pmid:31838836
  162. 162. Neul JL, Zoghbi HY. Rett syndrome: a prototypical neurodevelopmental disorder. The Neuroscientist. 2004;10: 118–128. pmid:15070486
  163. 163. Good KV, Vincent JB, Ausió J. MeCP2: The Genetic Driver of Rett Syndrome Epigenetics. Front Genet. 2021;12. pmid:33552148
  164. 164. Saxena A, Tang D, Carninci P. piRNAs Warrant Investigation in Rett Syndrome: An Omics Perspective. Dis Markers. 2012;33: 261–275. pmid:22976001
  165. 165. Lerner MR, Boyle JA, Hardin JA, Steitz JA. Two Novel Classes of Small Ribonucleoproteins Detected by Antibodies Associated with Lupus Erythematosus. Science. 1981;211: 400–402. pmid:6164096
  166. 166. Nisar S, Haris M. Neuroimaging genetics approaches to identify new biomarkers for the early diagnosis of autism spectrum disorder. Mol Psychiatry. 2023 [cited 30 Apr 2023]. pmid:37069342
  167. 167. Wakatsuki S, Takahashi Y, Shibata M, Adachi N, Numakawa T, Kunugi H, et al. Small noncoding vault RNA modulates synapse formation by amplifying MAPK signaling. J Cell Biol. 2021;220: 201911078. pmid:33439240
  168. 168. Geoffray M-M, Falissard B, Green J, Kerr B, Evans DG, Huson S, et al. Autism Spectrum Disorder Symptom Profile Across the RASopathies. Front Psychiatry. 2021;11. pmid:33519543
  169. 169. Borrie SC, Plasschaert E, Callaerts-Vegh Z, Yoshimura A, D’Hooge R, Elgersma Y, et al. MEK inhibition ameliorates social behavior phenotypes in a Spred1 knockout mouse model for RASopathy disorders. Mol Autism. 2021;12: 1–19. pmid:34311771
  170. 170. Caporali L, Fiorini C, Palombo F, Romagnoli M, Baccari F, Zenesini C, et al. Dissecting the multifaceted contribution of the mitochondrial genome to autism spectrum disorder. Front Genet. 2022; 13. pmid:36419830
  171. 171. Citrigno L, Muglia M, Qualtieri A, Spadafora P, Cavalcanti F, Pioggia G, et al. The Mitochondrial Dysfunction Hypothesis in Autism Spectrum Disorders: Current Status and Future Perspectives. Int J Mol Sci. 2020;21: 5785. pmid:32806635
  172. 172. Slade A, Kattini R, Campbell C, Holcik M. Diseases Associated with Defects in tRNA CCA Addition. Int J Mol Sci. 2020;21: 3780. pmid:32471101
  173. 173. Graf WD, Marin-Garcia J, Gao HG, Pizzo S, Naviaux RK, Markusic D, et al. Autism Associated With the Mitochondrial DNA G8363A Transfer RNALys Mutation. J Child Neurol. 2000;15: 357–361. pmid:10868777
  174. 174. Hultman CM, Sandin S, Levine SZ, Lichtenstein P, Reichenberg A. Advancing paternal age and risk of autism: new evidence from a population-based study and a meta-analysis of epidemiological studies. Mol Psychiatry. 2010;16: 1203–1212. pmid:21116277
  175. 175. Foldi CJ, Eyles DW, Flatscher-Bader T, McGrath JJ, Burne THJ. New Perspectives on Rodent Models of Advanced Paternal Age: Relevance to Autism. Front Behav Neurosci. 2011;5. pmid:21734873
  176. 176. Vojinovic D, Adams HH, Jian X, Yang Q, Smith AV, Bis JC, et al. Genome-wide association study of 23,500 individuals identifies 7 loci associated with brain ventricular volume. Nat Commun. 2018;9. pmid:30258056
  177. 177. Turner AH, Greenspan KS, Erp TGM. Pallidum and lateral ventricle volume enlargement in autism spectrum disorder. Psychiatry Res Neuroimaging. 2016;252: 40–45. pmid:27179315
  178. 178. Popov NT, Madjirova NP, Minkov IN, Vachev TI. Micro RNA HSA-486-3P Gene Expression Profiling in the Whole Blood of Patients with Autism. Biotechnol Biotechnol Equip. 2012;26: 3385–3388.
  179. 179. Monies D, Abouelhoda M, Assoum M, Moghrabi N, Rafiullah R, Almontashiri N, et al. Lessons Learned from Large-Scale, First-Tier Clinical Exome Sequencing in a Highly Consanguineous Population. Am J Hum Genet. 2019;104: 1182–1201. pmid:31130284
  180. 180. Hemmat M, Rumple MJ, Mahon LW, Strom CM, Anguiano A, Talai M, et al. Short stature, digit anomalies and dysmorphic facial features are associated with the duplication of miR-17 ~ 92 cluster. Mol Cytogenet. 2014;7: 27. pmid:24739087
  181. 181. Henshall DC. MicroRNA and epilepsy: profiling, functions and potential clinical applications. Curr Opin Neurol. 2014;27: 199–205. pmid:24553459
  182. 182. Lombardo MV, Lai M-C, Baron-Cohen S. Big data approaches to decomposing heterogeneity across the autism spectrum. Mol Psychiatry. 2019;24: 1435–1450. pmid:30617272
  183. 183. Stouffer SA, Suchman EA, DeVinney LC, Star SA, Williams RM Jr. The american soldier: Adjustment during army life.(studies in social psychology in world war ii), vol. 1. 1949.
  184. 184. Toro-Domínguez D, Villatoro-García JA, Martorell-Marugán J, Román-Montoya Y, Alarcón-Riquelme ME, Carmona-Sáez P. A survey of gene expression meta-analysis: methods and applications. Brief Bioinform. 2021;22: 1694–1705. pmid:32095826
  185. 185. Leidinger P, Backes C, Meder B, Meese E, Keller A. The human miRNA repertoire of different blood compounds. BMC Genomics. 2014;15. pmid:24928098
  186. 186. da Silva FC, da Rosa Iop R, Andrade A, Costa VP, Gutierres Filho PJB, da Silva R. Effects of physical exercise on the expression of MicroRNAs: a systematic review. J Strength Cond Res. 2020;34: 270–280. pmid:31877120
  187. 187. Hijmans JG, Levy MA, Garcia V, Lincenberg GM, Diehl KJ, Greiner JJ, et al. Insufficient sleep is associated with a pro‐atherogenic circulating miRNA signature. Exp Physiol. 2019;104: 975–982. pmid:31016755
  188. 188. McNaughton SA, Danaher J, Russell AP. Diet and microRNA expression: a systematic review. FASEB J. 2017;31: 644–3.
  189. 189. Palmer JD, Soule BP, Simone BA, Zaorsky NG, Jin L, Simone NL. MicroRNA expression altered by diet: can food be medicinal? Ageing Res Rev. 2014;17: 16–24. pmid:24833329
  190. 190. Benz F, Roy S, Trautwein C, Roderburg C, Luedde T. Circulating miRNA as biomarkers for sepsis. Int J Mol Sci. 2016;17: 78. pmid:26761003
  191. 191. Díaz-Román A, Zhang J, Delorme R, Beggiato A, Cortese S. Sleep in youth with autism spectrum disorders: systematic review and meta-analysis of subjective and objective studies. Evid Based Ment Health. 2018;21: 146–54. pmid:30361331
  192. 192. Marí-Bauset S, Zazpe I, Mari-Sanchis A, Llopis-González A, Morales-Suárez-Varela M. Food selectivity in autism spectrum disorders: a systematic review. J Child Neurol. 2014;29: 1554–61. pmid:24097852
  193. 193. Smith R, Farnworth H, Wright B, Allgar V. Are there more bowel symptoms in children with autism compared to normal children and children with other developmental and neurological disorders? A case control study. Autism. 2009;13: 343–355. pmid:19535465
  194. 194. Hillier A, Buckingham A, Schena D. Physical activity among adults with autism: Participation, attitudes, and barriers. Percept Mot Skills. 2020;127: 874–890. pmid:32443953
  195. 195. Lopes LT, Rodrigues JM, Baccarin C, Oliveira K, Abreu M, Ribeiro V, et al. Autism Spectrum as an Etiologic Systemic Disorder: A Protocol for an Umbrella Review. Healthcare. 2022;10: 2200. pmid:36360541
  196. 196. Leidinger P, Backes C, Rheinheimer S, Keller A, Meese E. Towards clinical applications of blood-borne miRNA signatures: the influence of the anticoagulant EDTA on miRNA abundance. PloS One. 2015;23;10(11):e0143321. pmid:26599228
  197. 197. Mitchell AJ, Gray WD, Hayek SS, Ko YA, Thomas S, Rooney K, et al. Platelets confound the measurement of extracellular miRNA in archived plasma. Sci Rep. 2016;13;6(1):1–1. pmid:27623086
  198. 198. Wright K, Silva K, Purdie AC, Plain KM. Comparison of methods for miRNA isolation and quantification from ovine plasma. Sci Rep. 2020;10. pmid:31964966
  199. 199. Schlosser K, McIntyre LA, White RJ, Stewart DJ. Customized Internal Reference Controls for Improved Assessment of Circulating MicroRNAs in Disease. Jeyaseelan K, editor. PLOS ONE. 2015;10: 0127443. pmid:26010841
  200. 200. Kulshreshtha R, Ferracin M, Negrini M, Calin GA, Davuluri RV, Ivan M. Regulation of microRNA expression: the hypoxic component. Cell Cycle. 2007;6: 1425–30. pmid:17582223
  201. 201. Ferrer I, Martinez A, Boluda S, Parchi P, Barrachina M. Brain banks: benefits, limitations and cautions concerning the use of post-mortem brain tissue for molecular studies. Cell Tissue Bank. 2008;9: 181–94. pmid:18543077
  202. 202. Wang H, Mao J, Li Y, Luo H, Wu J, Liao M, et al. 5 miRNA expression analyze in post-mortem interval (PMI) within 48 h. Forensic Sci Int Genet Suppl Ser. 2013;4: 190–1.
  203. 203. Lu M, Zhang Q, Deng M, Miao J, Guo Y, Gao W, et al. An analysis of human miRNA and disease associations. PloS One. 2008;3: 3420. pmid:18923704
  204. 204. Banzhaf-Strathmann J, Edbauer D. Good guy or bad guy: the opposing roles of miRNA 125b in cancer. Cell Commun Signal. 2014;12: 1–13. pmid:24774301
  205. 205. Vrijens K, Bollati V, Nawrot TS. miRNA as potential signatures of environmental exposure or effect: a systematic review. Environ Health Perspect. 2015;123: 399–411. pmid:25616258
  206. 206. Lewohl JM, Nunez YO, Dodd PR, Tiwari GR, Harris RA, Mayfield RD. Up‐regulation of miRNA in brain of human alcoholics. Alcohol Clin Exp Res. 2011;35: 1928–1937. pmid:21651580
  207. 207. Git A, Dvinge H, Salmon-Divon M, Osborne M, Kutter C, Hadfield J, et al. Systematic comparison of microarray profiling, real-time PCR, and next-generation sequencing technologies for measuring differential microRNA expression. RNA. 2010;16: 991–1006. pmid:20360395
  208. 208. Forero DA, González-Giraldo Y, Castro-Vega LJ, Barreto GE. qPCR-based methods for expression analysis of miRNAs. BioTechniques. 2019;67: 192–199. pmid:31560239
  209. 209. Bustin SA, Benes V, Garson JA, Hellemans J, Huggett J, Kubista M, et al. The MIQE Guidelines: Minimum Information for Publication of Quantitative Real-Time PCR Experiments. Clin Chem. 2009;55: 611–622. pmid:19246619
  210. 210. Nersisyan S, Shkurnikov M, Poloznikov A, Turchinovich A, Burwinkel B, Anisimov N, et al. A Post-Processing Algorithm for miRNA Microarray Data. Int J Mol Sci. 2020;21: 1228. pmid:32059403
  211. 211. Goodwin S, McPherson JD, McCombie WR. Coming of age: ten years of next-generation sequencing technologies. Nat Rev Genet. 2016;17: 333–351. pmid:27184599
  212. 212. Wright C, Rajpurohit A, Burke EE, Williams C, Collado-Torres L, Kimos M, et al. Comprehensive assessment of multiple biases in small RNA sequencing reveals significant differences in the performance of widely used methods. BMC Genomics. 2019;20: 513. pmid:31226924
  213. 213. Wang W, Kwon EJ, Tsai LH. miRNA in learning, memory, and neurological diseases. Learn Mem. 2012;19: 359–368. pmid:22904366
  214. 214. Treiber T, Treiber N, Meister G. Regulation of microRNA biogenesis and its crosstalk with other cellular pathways. Nat Rev Mol Cell Biol. 2019;20: 5–20. pmid:30228348
  215. 215. Fine SE, Weissman A, Gerdes M, Pinto-Martin J, Zackai EH, McDonald-McGinn DM, et al. Autism Spectrum Disorders and Symptoms in Children with Molecularly Confirmed 22q11.2 Deletion Syndrome. J Autism Dev Disord. 2005;35: 461–470. pmid:16134031
  216. 216. Percy AK. Rett Syndrome. Arch Neurol. 2011;68: 985. pmid:21825235
  217. 217. Mariani J, Coppola G, Zhang P, Abyzov A, Provini L, Tomasini L, et al. FOXG1-dependent dysregulation of GABA/glutamate neuron differentiation in autism spectrum disorders. Cell. 2015;16;162(2):375–90. pmid:26186191
  218. 218. Jin P, Zarnescu DC, Ceman S, Nakamoto M, Mowrey J, Jongens TA, et al. Biochemical and genetic interaction between the fragile X mental retardation protein and the microRNA pathway. Nat Neurosci. 2004;7: 113–117. pmid:14703574
  219. 219. Plante I, Davidovic L, Ouellet DL, Gobeil L-A, Tremblay S, Khandjian EW, et al. Dicer-derived microRNAs are utilized by the fragile X mental retardation protein for assembly on target RNAs. J Biomed Biotechnol. 2006;2006. pmid:17057366
  220. 220. Ahluwalia JK, Hariharan M, Bargaje R, Pillai B, Brahmachari V. Incomplete penetrance and variable expressivity: is there a microRNA connection? Bioessays. 2009;31: 981–92. pmid:19642110
  221. 221. Wilkinson MD, Dumontier M, Aalbersberg IJ, Appleton G, Axton M, Baak A, et al. The FAIR Guiding Principles for scientific data management and stewardship. Sci Data. 2016;3: 1–9. pmid:26978244
  222. 222. Byrd JB, Greene AC, Prasad DV, Jiang X, Greene CS. Responsible, practical genomic data sharing that accelerates research. Nat Rev Genet. 2020;21: 615–629. pmid:32694666
  223. 223. Piwowar HA, Chapman WW. Recall and bias of retrieving gene expression microarray datasets through PubMed identifiers. J Biomed Discov Collab. 2010;5: 7. pmid:20349403
  224. 224. Piwowar HA, Chapman WW. Public sharing of research datasets: a pilot study of associations. J Informetr. 2010;4: 148–156. pmid:21339841
  225. 225. Cook CE, Stroe O, Cochrane G, Birney E, Apweiler R. The European Bioinformatics Institute in 2020: building a global infrastructure of interconnected data resources for the life sciences. Nucleic Acids Res. 2020;48: D17–D23. pmid:31701143
  226. 226. Joly Y, Dove ES, Knoppers BM, Bobrow M, Chalmers D. Data Sharing in the Post-Genomic World: The Experience of the International Cancer Genome Consortium (ICGC) Data Access Compliance Office (DACO). Bourne PE, editor. PLoS Comput Biol. 2012;8: e1002549. pmid:22807659
  227. 227. The ICGC/TCGA Pan-Cancer Analysis of Whole Genomes Consortium, Aaltonen LA, Abascal F, Abeshouse A, Aburatani H, Adams DJ, et al. Pan-cancer analysis of whole genomes. Nature. 2020;578: 82–93. pmid:32025007
  228. 228. Knoppers BM, Bernier A, Bowers S, Kirby E. Open Data in the Era of the GDPR: Lessons from the Human Cell Atlas. Annu Rev Genomics Hum Genet. 2023;24: annurev-genom-101322-113255. pmid:36791787
  229. 229. Phillips M, Molnár-Gábor F, Korbel JO, Thorogood A, Joly Y, Chalmers D, et al. Genomics: data sharing needs an international code of conduct. Nature. 2020;578: 31–311. pmid:32025008