1
|
Shayota BJ. Downstream Assays for Variant Resolution: Epigenetics, RNA Sequnncing, and Metabolomics. Pediatr Clin North Am 2023; 70:929-936. [PMID: 37704351 DOI: 10.1016/j.pcl.2023.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
As the availability of advanced molecular testing like whole exome and genome sequencing expands, it comes with the added complication of interpreting inconclusive results, including determining the relevance of variants of uncertain significance or failing to find a variant in an otherwise suspected specific genetic disorder. This complication necessitates the use of alternative testing methods to gather more information in support of, or against, a particular genetic diagnosis. Therefore, new genome-wide approaches, including DNA epigenetic testing, RNA sequencing, and metabolomics, are increasingly being used to increase the diagnostic yield when used in conjunction with more conventional genetic tests.
Collapse
Affiliation(s)
- Brian J Shayota
- University of Utah, 295 Chipeta Way, Salt Lake City, UT 84108, USA; Primary Children's Hospital, Salt Lake City, UT, USA.
| |
Collapse
|
2
|
Chen WX, Liu B, Zhou L, Xiong X, Fu J, Huang ZF, Tan T, Tang M, Wang J, Tang YP. De novo mutations within metabolism networks of amino acid/protein/energy in Chinese autistic children with intellectual disability. Hum Genomics 2022; 16:52. [PMID: 36320054 PMCID: PMC9623983 DOI: 10.1186/s40246-022-00427-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 10/19/2022] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is often accompanied by intellectual disability (ID). Despite extensive studies, however, the genetic basis for this comorbidity is still not clear. In this study, we tried to develop an analyzing pipeline for de novo mutations and possible pathways related to ID phenotype in ASD. Whole-exome sequencing (WES) was performed to screen de novo mutations and candidate genes in 79 ASD children together with their parents (trios). The de novo altering genes and relative pathways which were associated with ID phenotype were analyzed. The connection nodes (genes) of above pathways were selected, and the diagnostic value of these selected genes for ID phenotype in the study population was also evaluated. RESULTS We identified 89 de novo mutant genes, of which 34 genes were previously reported to be associated with ASD, including double hits in the EGF repeats of NOTCH1 gene (p.V999M and p.S1027L). Interestingly, of these 34 genes, 22 may directly affect intelligence quotient (IQ). Further analyses revealed that these IQ-related genes were enriched in protein synthesis, energy metabolism, and amino acid metabolism, and at least 9 genes (CACNA1A, ALG9, PALM2, MGAT4A, PCK2, PLEKHA1, PSME3, ADI1, and TLE3) were involved in all these three pathways. Seven patients who harbored these gene mutations showed a high prevalence of a low IQ score (< 70), a non-verbal language, and an early diagnostic age (< 4 years). Furthermore, our panel of these 9 genes reached a 10.2% diagnostic rate (5/49) in early diagnostic patients with a low IQ score and also reached a 10% diagnostic yield in those with both a low IQ score and non-verbal language (4/40). CONCLUSION We found some new genetic disposition for ASD accompanied with intellectual disability in this study. Our results may be helpful for etiologic research and early diagnoses of intellectual disability in ASD. Larger population studies and further mechanism studies are warranted.
Collapse
Affiliation(s)
- Wen-Xiong Chen
- grid.410737.60000 0000 8653 1072The Assessment and Intervention Center for Autistic Children, Department of Neurology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, 510623 Guangdong China
| | - Bin Liu
- grid.410737.60000 0000 8653 1072Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, 510623 China ,grid.258164.c0000 0004 1790 3548Department of Biobank, Shenzhen Baoan Women’s and Children’s Hospital, Jinan University, Shenzhen, 518102 Guangdong China
| | - Lijie Zhou
- grid.412719.8Department of Pediatric Rehabilitation, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China
| | - Xiaoli Xiong
- grid.410737.60000 0000 8653 1072Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, 510623 China
| | - Jie Fu
- grid.410737.60000 0000 8653 1072Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, 510623 China
| | - Zhi-Fang Huang
- grid.410737.60000 0000 8653 1072The Assessment and Intervention Center for Autistic Children, Department of Neurology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, 510623 Guangdong China
| | - Ting Tan
- grid.410737.60000 0000 8653 1072Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, 510623 China
| | - Mingxi Tang
- grid.488387.8Department of Pathology, Affiliated Hospital of Southwest Medical University, Luzhou, 646000 Sichuan China
| | - Jun Wang
- grid.412719.8Department of Pediatric Rehabilitation, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China
| | - Ya-Ping Tang
- grid.410737.60000 0000 8653 1072Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, 510623 China ,grid.412719.8Department of Pediatric Rehabilitation, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 China ,grid.12981.330000 0001 2360 039XGuangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080 Guangdong China
| |
Collapse
|
3
|
Fetit R, Hillary RF, Price DJ, Lawrie SM. The neuropathology of autism: A systematic review of post-mortem studies of autism and related disorders. Neurosci Biobehav Rev 2021; 129:35-62. [PMID: 34273379 DOI: 10.1016/j.neubiorev.2021.07.014] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/13/2021] [Accepted: 07/10/2021] [Indexed: 02/07/2023]
Abstract
Post-mortem studies allow for the direct investigation of brain tissue in those with autism and related disorders. Several review articles have focused on aspects of post-mortem abnormalities but none has brought together the entire post-mortem literature. Here, we systematically review the evidence from post-mortem studies of autism, and of related disorders that present with autistic features. The literature consists of a small body of studies with small sample sizes, but several remarkably consistent findings are evident. Cortical layering is largely undisturbed, but there are consistent reductions in minicolumn numbers and aberrant myelination. Transcriptomics repeatedly implicate abberant synaptic, metabolic, proliferation, apoptosis and immune pathways. Sufficient replicated evidence is available to implicate non-coding RNA, aberrant epigenetic profiles, GABAergic, glutamatergic and glial dysfunction in autism pathogenesis. Overall, the cerebellum and frontal cortex are most consistently implicated, sometimes revealing distinct region-specific alterations. The literature on related disorders such as Rett syndrome, Fragile X and copy number variations (CNVs) predisposing to autism is particularly small and inconclusive. Larger studies, matched for gender, developmental stage, co-morbidities and drug treatment are required.
Collapse
Affiliation(s)
- Rana Fetit
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK.
| | - Robert F Hillary
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - David J Price
- Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK
| | - Stephen M Lawrie
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH10 5HF, UK; Patrick Wild Centre, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH10 5HF, UK
| |
Collapse
|
4
|
Guan J, Wang Y, Lin Y, Yin Q, Zhuang Y, Ji G. Cell Type-Specific Predictive Models Perform Prioritization of Genes and Gene Sets Associated With Autism. Front Genet 2021; 11:628539. [PMID: 33519924 PMCID: PMC7844401 DOI: 10.3389/fgene.2020.628539] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 12/16/2020] [Indexed: 12/13/2022] Open
Abstract
Bulk transcriptomic analyses of autism spectrum disorder (ASD) have revealed dysregulated pathways, while the brain cell type-specific molecular pathology of ASD still needs to be studied. Machine learning-based studies can be conducted for ASD, prioritizing high-confidence gene candidates and promoting the design of effective interventions. Using human brain nucleus gene expression of ASD and controls, we construct cell type-specific predictive models for ASD based on individual genes and gene sets, respectively, to screen cell type-specific ASD-associated genes and gene sets. These two kinds of predictive models can predict the diagnosis of a nucleus with known cell type. Then, we construct a multi-label predictive model for predicting the cell type and diagnosis of a nucleus at the same time. Our findings suggest that layer 2/3 and layer 4 excitatory neurons, layer 5/6 cortico-cortical projection neurons, parvalbumin interneurons, and protoplasmic astrocytes are preferentially affected in ASD. The functions of genes with predictive power for ASD are different and the top important genes are distinct across different cells, highlighting the cell-type heterogeneity of ASD. The constructed predictive models can promote the diagnosis of ASD, and the prioritized cell type-specific ASD-associated genes and gene sets may be used as potential biomarkers of ASD.
Collapse
Affiliation(s)
- Jinting Guan
- Department of Automation, Xiamen University, Xiamen, China.,National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, China
| | - Yang Wang
- Department of Automation, Xiamen University, Xiamen, China
| | - Yiping Lin
- Department of Automation, Xiamen University, Xiamen, China
| | - Qingyang Yin
- Department of Automation, Xiamen University, Xiamen, China
| | - Yibo Zhuang
- Xiamen YLZ Yihui Technology Co., Ltd., Xiamen, China
| | - Guoli Ji
- Department of Automation, Xiamen University, Xiamen, China.,National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, China.,Innovation Center for Cell Signaling Network, Xiamen University, Xiamen, China
| |
Collapse
|
5
|
Vargason T, Grivas G, Hollowood-Jones KL, Hahn J. Towards a Multivariate Biomarker-Based Diagnosis of Autism Spectrum Disorder: Review and Discussion of Recent Advancements. Semin Pediatr Neurol 2020; 34:100803. [PMID: 32446437 PMCID: PMC7248126 DOI: 10.1016/j.spen.2020.100803] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
An ever-evolving understanding of autism spectrum disorder (ASD) pathophysiology necessitates that diagnostic standards also evolve from being observation-based to include quantifiable clinical measurements. The multisystem nature of ASD motivates the use of multivariate methods of statistical analysis over common univariate approaches for discovering clinical biomarkers relevant to this goal. In addition to characterization of important behavioral patterns for improving current diagnostic instruments, multivariate analyses to date have allowed for thorough investigation of neuroimaging-based, genetic, and metabolic abnormalities in individuals with ASD. This review highlights current research using multivariate statistical analyses to quantify the value of these behavioral and physiological markers for ASD diagnosis. A detailed discussion of a blood-based diagnostic test for ASD using specific metabolite concentrations is also provided. The advancement of ASD biomarker research promises to provide earlier and more accurate diagnoses of the disorder.
Collapse
Affiliation(s)
- Troy Vargason
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY
| | - Genevieve Grivas
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY
| | - Kathryn L Hollowood-Jones
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY
| | - Juergen Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY; Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, NY.
| |
Collapse
|
6
|
Fernandez GJ, Ferreira JH, Vechetti IJ, de Moraes LN, Cury SS, Freire PP, Gutiérrez J, Ferretti R, Dal-Pai-Silva M, Rogatto SR, Carvalho RF. MicroRNA-mRNA Co-sequencing Identifies Transcriptional and Post-transcriptional Regulatory Networks Underlying Muscle Wasting in Cancer Cachexia. Front Genet 2020; 11:541. [PMID: 32547603 PMCID: PMC7272700 DOI: 10.3389/fgene.2020.00541] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 05/05/2020] [Indexed: 12/23/2022] Open
Abstract
Cancer cachexia is a metabolic syndrome with alterations in gene regulatory networks that consequently lead to skeletal muscle wasting. Integrating microRNAs-mRNAs omics profiles offers an opportunity to understand transcriptional and post-transcriptional regulatory networks underlying muscle wasting. Here, we used RNA sequencing to simultaneously integrate and explore microRNAs and mRNAs expression profiles in the tibialis anterior (TA) muscles of the Lewis Lung Carcinoma (LLC) model of cancer cachexia. We found 1,008 mRNAs and 18 microRNAs differentially expressed in cachectic mice compared with controls. Although our transcriptomic analysis demonstrated a high heterogeneity in mRNA profiles of cachectic mice, we identified a reduced number of differentially expressed genes that were uniformly regulated within cachectic muscles. This set of uniformly regulated genes is associated with the extracellular matrix (ECM), proteolysis, and inflammatory response. We also used transcriptomic data to perform enrichment analysis of transcriptional factor binding sites in promoter sequences, which revealed activation of the atrophy-related transcription factors NF-κB, Stat3, AP-1, and FoxO. Furthermore, the integration of mRNA and microRNA expression profiles identified post-transcriptional regulation by microRNAs of genes involved in ECM organization, cell migration, transcription factors binding, ion transport, and the FoxO signaling pathway. Our integrative analysis of microRNA-mRNA co-profiles comprehensively characterized regulatory relationships of molecular pathways and revealed microRNAs targeting ECM-associated genes in cancer cachexia.
Collapse
Affiliation(s)
- Geysson Javier Fernandez
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, Botucatu, Brazil.,Faculty of Medicine, University of Antioquia, Medellín, Colombia
| | - Juarez Henrique Ferreira
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, Botucatu, Brazil
| | - Ivan José Vechetti
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, Botucatu, Brazil
| | - Leonardo Nazario de Moraes
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, Botucatu, Brazil
| | - Sarah Santiloni Cury
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, Botucatu, Brazil
| | - Paula Paccielli Freire
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, Botucatu, Brazil
| | - Jayson Gutiérrez
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
| | - Renato Ferretti
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, Botucatu, Brazil
| | - Maeli Dal-Pai-Silva
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, Botucatu, Brazil
| | - Silvia Regina Rogatto
- Department of Clinical Genetics, University Hospital of Southern Denmark, Vejle, Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Robson Francisco Carvalho
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, Botucatu, Brazil
| |
Collapse
|
7
|
Huang G, Osorio D, Guan J, Ji G, Cai JJ. Overdispersed gene expression in schizophrenia. NPJ SCHIZOPHRENIA 2020; 6:9. [PMID: 32245959 PMCID: PMC7125213 DOI: 10.1038/s41537-020-0097-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 02/13/2020] [Indexed: 02/07/2023]
Abstract
Schizophrenia (SCZ) is a severe, highly heterogeneous psychiatric disorder with varied clinical presentations. The polygenic genetic architecture of SCZ makes identification of causal variants a daunting task. Gene expression analyses hold the promise of revealing connections between dysregulated transcription and underlying variants in SCZ. However, the most commonly used differential expression analysis often assumes grouped samples are from homogeneous populations and thus cannot be used to detect expression variance differences between samples. Here, we applied the test for equality of variances to normalized expression data, generated by the CommonMind Consortium (CMC), from brains of 212 SCZ and 214 unaffected control (CTL) samples. We identified 87 genes, including VEGFA (vascular endothelial growth factor) and BDNF (brain-derived neurotrophic factor), that showed a significantly higher expression variance among SCZ samples than CTL samples. In contrast, only one gene showed the opposite pattern. To extend our analysis to gene sets, we proposed a Mahalanobis distance-based test for multivariate homogeneity of group dispersions, with which we identified 110 gene sets with a significantly higher expression variability in SCZ, including sets of genes encoding phosphatidylinositol 3-kinase (PI3K) complex and several others involved in cerebellar cortex morphogenesis, neuromuscular junction development, and cerebellar Purkinje cell layer development. Taken together, our results suggest that SCZ brains are characterized by overdispersed gene expression-overall gene expression variability among SCZ samples is significantly higher than that among CTL samples. Our study showcases the application of variability-centric analyses in SCZ research.
Collapse
Affiliation(s)
- Guangzao Huang
- Department of Automation, Xiamen University, Xiamen, 361005, China
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, 361005, China
- College of Electrical and Electronic Engineering, Wenzhou University, Wenzhou, 325035, China
| | - Daniel Osorio
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, 77843, USA
| | - Jinting Guan
- Department of Automation, Xiamen University, Xiamen, 361005, China
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, 361005, China
| | - Guoli Ji
- Department of Automation, Xiamen University, Xiamen, 361005, China.
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, 361005, China.
- Innovation Center for Cell Signaling Network, Xiamen University, Xiamen, 361005, China.
| | - James J Cai
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, 77843, USA.
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX, 77843, USA.
- Interdisciplinary Program of Genetics, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
8
|
Chen YJ, Chen CY, Mai TL, Chuang CF, Chen YC, Gupta SK, Yen L, Wang YD, Chuang TJ. Genome-wide, integrative analysis of circular RNA dysregulation and the corresponding circular RNA-microRNA-mRNA regulatory axes in autism. Genome Res 2020; 30:375-391. [PMID: 32127416 PMCID: PMC7111521 DOI: 10.1101/gr.255463.119] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 02/24/2020] [Indexed: 02/07/2023]
Abstract
Circular RNAs (circRNAs), a class of long noncoding RNAs, are known to be enriched in mammalian neural tissues. Although a wide range of dysregulation of gene expression in autism spectrum disorder (ASD) have been reported, the role of circRNAs in ASD remains largely unknown. Here, we performed genome-wide circRNA expression profiling in postmortem brains from individuals with ASD and controls and identified 60 circRNAs and three coregulated modules that were perturbed in ASD. By integrating circRNA, microRNA, and mRNA dysregulation data derived from the same cortex samples, we identified 8170 ASD-associated circRNA-microRNA-mRNA interactions. Putative targets of the axes were enriched for ASD risk genes and genes encoding inhibitory postsynaptic density (PSD) proteins, but not for genes implicated in monogenetic forms of other brain disorders or genes encoding excitatory PSD proteins. This reflects the previous observation that ASD-derived organoids show overproduction of inhibitory neurons. We further confirmed that some ASD risk genes (NLGN1, STAG1, HSD11B1, VIP, and UBA6) were regulated by an up-regulated circRNA (circARID1A) via sponging a down-regulated microRNA (miR-204-3p) in human neuronal cells. Particularly, alteration of NLGN1 expression is known to affect the dynamic processes of memory consolidation and strengthening. To the best of our knowledge, this is the first systems-level view of circRNA regulatory networks in ASD cortex samples. We provided a rich set of ASD-associated circRNA candidates and the corresponding circRNA-microRNA-mRNA axes, particularly those involving ASD risk genes. Our findings thus support a role for circRNA dysregulation and the corresponding circRNA-microRNA-mRNA axes in ASD pathophysiology.
Collapse
Affiliation(s)
- Yen-Ju Chen
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
- Genome and Systems Biology Degree Program, Academia Sinica and National Taiwan University, Taipei 10617, Taiwan
| | - Chia-Ying Chen
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Te-Lun Mai
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Chih-Fan Chuang
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Yu-Chen Chen
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Sachin Kumar Gupta
- Department of Pathology and Immunology
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Laising Yen
- Department of Pathology and Immunology
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Yi-Da Wang
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Trees-Juen Chuang
- Genomics Research Center, Academia Sinica, Taipei 11529, Taiwan
- Genome and Systems Biology Degree Program, Academia Sinica and National Taiwan University, Taipei 10617, Taiwan
| |
Collapse
|
9
|
Single-Cell Expression Variability Implies Cell Function. Cells 2019; 9:cells9010014. [PMID: 31861624 PMCID: PMC7017299 DOI: 10.3390/cells9010014] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 11/26/2019] [Accepted: 11/27/2019] [Indexed: 12/11/2022] Open
Abstract
As single-cell RNA sequencing (scRNA-seq) data becomes widely available, cell-to-cell variability in gene expression, or single-cell expression variability (scEV), has been increasingly appreciated. However, it remains unclear whether this variability is functionally important and, if so, what are its implications for multi-cellular organisms. Here, we analyzed multiple scRNA-seq data sets from lymphoblastoid cell lines (LCLs), lung airway epithelial cells (LAECs), and dermal fibroblasts (DFs) and, for each cell type, selected a group of homogenous cells with highly similar expression profiles. We estimated the scEV levels for genes after correcting the mean-variance dependency in that data and identified 465, 466, and 364 highly variable genes (HVGs) in LCLs, LAECs, and DFs, respectively. Functions of these HVGs were found to be enriched with those biological processes precisely relevant to the corresponding cell type’s function, from which the scRNA-seq data used to identify HVGs were generated—e.g., cytokine signaling pathways were enriched in HVGs identified in LCLs, collagen formation in LAECs, and keratinization in DFs. We repeated the same analysis with scRNA-seq data from induced pluripotent stem cells (iPSCs) and identified only 79 HVGs with no statistically significant enriched functions; the overall scEV in iPSCs was of negligible magnitude. Our results support the “variation is function” hypothesis, arguing that scEV is required for cell type-specific, higher-level system function. Thus, quantifying and characterizing scEV are of importance for our understating of normal and pathological cellular processes.
Collapse
|
10
|
Ansel A, Posen Y, Ellis R, Deutsch L, Zisman PD, Gesundheit B. Biomarkers for Autism Spectrum Disorders (ASD): A Meta-analysis. Rambam Maimonides Med J 2019; 10:RMMJ.10375. [PMID: 31675302 PMCID: PMC6824829 DOI: 10.5041/rmmj.10375] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE To compare the reported accuracy and sensitivity of the various modalities used to diagnose autism spectrum disorders (ASD) in efforts to help focus further biomarker research on the most promising methods for early diagnosis. METHODS The Medline scientific literature database was searched to identify publications assessing potential clinical ASD biomarkers. Reports were categorized by the modality used to assess the putative markers, including protein, genetic, metabolic, or objective imaging methods. The reported sensitivity, specificity, area under the curve, and overall agreement were summarized and analyzed to determine weighted averages for each diagnostic modality. Heterogeneity was measured using the I2 test. RESULTS Of the 71 papers included in this analysis, each belonging to one of five modalities, protein-based followed by metabolite-based markers provided the highest diagnostic accuracy, each with a pooled overall agreement of 83.3% and respective weighted area under the curve (AUC) of 89.5% and 88.3%. Sensitivity provided by protein markers was highest (85.5%), while metabolic (85.9%) and protein markers (84.7%) had the highest specificity. Other modalities showed degrees of sensitivity, specificity, and overall agreements in the range of 73%-80%. CONCLUSIONS Each modality provided for diagnostic accuracy and specificity similar or slightly higher than those reported for the gold-standard Autism Diagnostic Observation Schedule (ADOS) instrument. Further studies are required to identify the most predictive markers within each modality and to evaluate biological pathways or clustering with possible etiological relevance. Analyses will also be necessary to determine the potential of these novel biomarkers in diagnosing pediatric patients, thereby enabling early intervention.
Collapse
Affiliation(s)
| | - Yehudit Posen
- Cell-El Therapeutics Ltd, Jerusalem, Israel
- PSW Ltd, Rehovot, Israel
| | - Ronald Ellis
- Cell-El Therapeutics Ltd, Jerusalem, Israel
- Biotech & Biopharma Consulting, Jerusalem, Israel
| | - Lisa Deutsch
- Biostats Statistical Consulting Ltd, Modiin, Israel
| | | | - Benjamin Gesundheit
- Cell-El Therapeutics Ltd, Jerusalem, Israel
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
11
|
Shnier D, Voineagu MA, Voineagu I. Persistent homology analysis of brain transcriptome data in autism. J R Soc Interface 2019; 16:20190531. [PMID: 31551047 PMCID: PMC6769309 DOI: 10.1098/rsif.2019.0531] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Persistent homology methods have found applications in the analysis of multiple types of biological data, particularly imaging data or data with a spatial and/or temporal component. However, few studies have assessed the use of persistent homology for the analysis of gene expression data. Here we apply persistent homology methods to investigate the global properties of gene expression in post-mortem brain tissue (cerebral cortex) of individuals with autism spectrum disorders (ASD) and matched controls. We observe a significant difference in the geometry of inter-sample relationships between autism and healthy controls as measured by the sum of the death times of zero-dimensional components and the Euler characteristic. This observation is replicated across two distinct datasets, and we interpret it as evidence for an increased heterogeneity of gene expression in autism. We also assessed the topology of gene-level point clouds and did not observe significant differences between ASD and control transcriptomes, suggesting that the overall transcriptome organization is similar in ASD and healthy cerebral cortex. Overall, our study provides a novel framework for persistent homology analyses of gene expression data for genetically complex disorders.
Collapse
Affiliation(s)
- Daniel Shnier
- Department of Mathematics and Statistics, University of New South Wales, Kensington, Sydney, New South Wales 2052, Australia
| | - Mircea A Voineagu
- Department of Mathematics and Statistics, University of New South Wales, Kensington, Sydney, New South Wales 2052, Australia
| | - Irina Voineagu
- Department of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, Sydney, New South Wales 2052, Australia
| |
Collapse
|
12
|
Commonality in dysregulated expression of gene sets in cortical brains of individuals with autism, schizophrenia, and bipolar disorder. Transl Psychiatry 2019; 9:152. [PMID: 31127088 PMCID: PMC6534650 DOI: 10.1038/s41398-019-0488-4] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 04/03/2019] [Accepted: 04/10/2019] [Indexed: 12/14/2022] Open
Abstract
Individuals affected with different neuropsychiatric disorders such as autism (AUT), schizophrenia (SCZ) and bipolar disorder (BPD), may share similar clinical manifestations, suggesting shared genetic influences and common biological mechanisms underlying these disorders. Using brain transcriptome data gathered from postmortem donors affected with AUT, SCZ and BPD, it is now possible to identify shared dysregulated gene sets, i.e., those abnormally expressed in brains of neuropsychiatric patients, compared to non-psychiatric controls. Here, we apply a novel aberrant gene expression analysis method, coupled with consensus co-expression network analysis, to identify gene sets with shared dysregulated expression in cortical brains of individuals affected with AUT, SCZ and BPD. We identify eight gene sets with dysregulated expression shared by AUT, SCZ and BPD, 23 by AUT and SCZ, four by AUT and BPD, and two by SCZ and BPD. The identified genes are enriched with functions relevant to amino acid transport, synapse, neurotransmitter release, oxidative stress, nitric oxide synthase biosynthesis, immune response, protein folding, lysophosphatidic acid-mediated signaling and glycolysis. Our method has been proven to be effective in discovering and revealing multigene sets with dysregulated expression shared by different neuropsychiatric disorders. Our findings provide new insights into the common molecular mechanisms underlying the pathogenesis and progression of AUT, SCZ and BPD, contributing to the study of etiological overlap between these neuropsychiatric disorders.
Collapse
|
13
|
Guan J, Chen M, Ye C, Cai JJ, Ji G. AEGS: identifying aberrantly expressed gene sets for differential variability analysis. Bioinformatics 2018; 34:881-883. [PMID: 29040376 PMCID: PMC6192207 DOI: 10.1093/bioinformatics/btx646] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 10/10/2017] [Indexed: 11/22/2022] Open
Abstract
Motivation In gene expression studies, differential expression (DE) analysis has been widely used
to identify genes with shifted expression mean between groups. Recently, differential
variability (DV) analysis has been increasingly applied as analyzing changed expression
variability (e.g. the changes in expression variance) between groups may reveal
underlying genetic heterogeneity and undetected interactions, which has great
implications in many fields of biology. An easy-to-use tool for DV analysis is
needed. Results We develop AEGS for DV analysis, to identify
aberrantly
expressed gene
sets in diseased cases but not in controls. AEGS
can rank individual genes in an aberrantly expressed gene set by each gene’s relative
contribution to the total degree of aberrant expression, prioritizing top genes. AEGS
can be used for discovering gene sets with disease-specific expression variability
changes. Availability and implementation AEGS web server is accessible at http://bmi.xmu.edu.cn:8003/AEGS, where a stand-alone AEGS application can
also be downloaded.
Collapse
Affiliation(s)
- Jinting Guan
- Department of Automation, Xiamen University.,Xiamen Research Institute of National Center of Healthcare Big Data, Xiamen 361005, China
| | | | - Congting Ye
- College of Environment and Ecology, Xiamen University, Xiamen 361102, China
| | - James J Cai
- Department of Veterinary Integrative Biosciences.,Interdisciplinary Program in Genetics, Texas A&M University, College Station, TX 77843, USA
| | - Guoli Ji
- Department of Automation, Xiamen University.,Xiamen Research Institute of National Center of Healthcare Big Data, Xiamen 361005, China.,Innovation Center for Cell Signaling Network, Xiamen University, Xiamen 361005, China
| |
Collapse
|
14
|
Mukhamedyarov MA, Rizvanov AA, Yakupov EZ, Zefirov AL, Kiyasov AP, Reis HJ, Teixeira AL, Vieira LB, Lima LM, Salafutdinov II, Petukhova EO, Khaiboullina SF, Schlauch KA, Lombardi VC, Palotás A. Transcriptional Analysis of Blood Lymphocytes and Skin Fibroblasts, Keratinocytes, and Endothelial Cells as a Potential Biomarker for Alzheimer's Disease. J Alzheimers Dis 2018; 54:1373-1383. [PMID: 27589530 DOI: 10.3233/jad-160457] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Alzheimer's disease (AD) is a devastating and progressive form of dementia that is typically associated with a build-up of amyloid-β plaques and hyperphosphorylated and misfolded tau protein in the brain. Presently, there is no single test that confirms AD; therefore, a definitive diagnosis is only made after a comprehensive medical evaluation, which includes medical history, cognitive tests, and a neurological examination and/or brain imaging. Additionally, the protracted prodromal phase of the disease makes selection of control subjects for clinical trials challenging. In this study we have utilized a gene-expression array to screen blood and skin punch biopsy (fibroblasts, keratinocytes, and endothelial cells) for transcriptional differences that may lead to a greater understanding of AD as well as identify potential biomarkers. Our analysis identified 129 differentially expressed genes from blood of dementia cases when compared to healthy individuals, and four differentially expressed punch biopsy genes between AD subjects and controls. Additionally, we identified a set of genes in both tissue compartments that showed transcriptional variation in AD but were largely stable in controls. The translational products of these variable genes are involved in the maintenance of the Golgi structure, regulation of lipid metabolism, DNA repair, and chromatin remodeling. Our analysis potentially identifies specific genes in both tissue compartments that may ultimately lead to useful biomarkers and may provide new insight into the pathophysiology of AD.
Collapse
Affiliation(s)
| | | | | | | | | | - Helton J Reis
- Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | | | | | | | | | - Svetlana F Khaiboullina
- Kazan Federal University, Kazan, Russia.,Nevada Center for Biomedical Research, Reno, NV, USA
| | | | - Vincent C Lombardi
- University of Nevada, Reno, NV, USA.,Nevada Center for Biomedical Research, Reno, NV, USA
| | - András Palotás
- Kazan Federal University, Kazan, Russia.,Asklepios-Med (private medical practice and research center), Szeged, Hungary
| |
Collapse
|
15
|
Kremer LS, Bader DM, Mertes C, Kopajtich R, Pichler G, Iuso A, Haack TB, Graf E, Schwarzmayr T, Terrile C, Koňaříková E, Repp B, Kastenmüller G, Adamski J, Lichtner P, Leonhardt C, Funalot B, Donati A, Tiranti V, Lombes A, Jardel C, Gläser D, Taylor RW, Ghezzi D, Mayr JA, Rötig A, Freisinger P, Distelmaier F, Strom TM, Meitinger T, Gagneur J, Prokisch H. Genetic diagnosis of Mendelian disorders via RNA sequencing. Nat Commun 2017; 8:15824. [PMID: 28604674 PMCID: PMC5499207 DOI: 10.1038/ncomms15824] [Citation(s) in RCA: 393] [Impact Index Per Article: 49.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 04/28/2017] [Indexed: 12/23/2022] Open
Abstract
Across a variety of Mendelian disorders, ∼50-75% of patients do not receive a genetic diagnosis by exome sequencing indicating disease-causing variants in non-coding regions. Although genome sequencing in principle reveals all genetic variants, their sizeable number and poorer annotation make prioritization challenging. Here, we demonstrate the power of transcriptome sequencing to molecularly diagnose 10% (5 of 48) of mitochondriopathy patients and identify candidate genes for the remainder. We find a median of one aberrantly expressed gene, five aberrant splicing events and six mono-allelically expressed rare variants in patient-derived fibroblasts and establish disease-causing roles for each kind. Private exons often arise from cryptic splice sites providing an important clue for variant prioritization. One such event is found in the complex I assembly factor TIMMDC1 establishing a novel disease-associated gene. In conclusion, our study expands the diagnostic tools for detecting non-exonic variants and provides examples of intronic loss-of-function variants with pathological relevance.
Collapse
Affiliation(s)
- Laura S. Kremer
- Institute of Human Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Institute of Human Genetics, Klinikum rechts der Isar, Technische Universität München, 81675 München, Germany
| | - Daniel M. Bader
- Department of Informatics, Technische Universität München, 85748 Garching, Germany
- Quantitative Biosciences Munich, Gene Center, Department of Biochemistry, Ludwig Maximilian Universität München, 81377 München, Germany
| | - Christian Mertes
- Department of Informatics, Technische Universität München, 85748 Garching, Germany
| | - Robert Kopajtich
- Institute of Human Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Institute of Human Genetics, Klinikum rechts der Isar, Technische Universität München, 81675 München, Germany
| | - Garwin Pichler
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Arcangela Iuso
- Institute of Human Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Institute of Human Genetics, Klinikum rechts der Isar, Technische Universität München, 81675 München, Germany
| | - Tobias B. Haack
- Institute of Human Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Institute of Human Genetics, Klinikum rechts der Isar, Technische Universität München, 81675 München, Germany
| | - Elisabeth Graf
- Institute of Human Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Institute of Human Genetics, Klinikum rechts der Isar, Technische Universität München, 81675 München, Germany
| | - Thomas Schwarzmayr
- Institute of Human Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Institute of Human Genetics, Klinikum rechts der Isar, Technische Universität München, 81675 München, Germany
| | - Caterina Terrile
- Institute of Human Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Eliška Koňaříková
- Institute of Human Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Institute of Human Genetics, Klinikum rechts der Isar, Technische Universität München, 81675 München, Germany
| | - Birgit Repp
- Institute of Human Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Institute of Human Genetics, Klinikum rechts der Isar, Technische Universität München, 81675 München, Germany
| | - Gabi Kastenmüller
- Institute of Bioinformatics and Systems Biology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Jerzy Adamski
- Institute of Experimental Genetics, Genome Analysis Center, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Peter Lichtner
- Institute of Human Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | | | - Benoit Funalot
- INSERM U1163, Université Paris Descartes—Sorbonne Paris Cité, Institut Imagine, 75015 Paris, France
| | - Alice Donati
- Metabolic Unit, A. Meyer Children’s Hospital, 50139 Florence, Italy
| | - Valeria Tiranti
- Unit of Molecular Neurogenetics, Foundation IRCCS (Istituto di Ricovero e Cura a Carettere Scientifico) Neurological Institute ‘Carlo Besta’, 20126 Milan, Italy
| | - Anne Lombes
- Inserm UMR 1016, Institut Cochin, 75014 Paris, France
- CNRS UMR 8104, Institut Cochin, 75014 Paris, France
- Université Paris V René Descartes, Institut Cochin, 75014 Paris, France
| | - Claude Jardel
- Inserm UMR 1016, Institut Cochin, 75014 Paris, France
- AP/HP, GHU Pitié-Salpêtrière, Service de Biochimie Métabolique, 75013 Paris, France
| | - Dieter Gläser
- Genetikum, Genetic Counseling and Diagnostics, 89231 Neu-Ulm, Germany
| | - Robert W. Taylor
- Wellcome Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Daniele Ghezzi
- Unit of Molecular Neurogenetics, Foundation IRCCS (Istituto di Ricovero e Cura a Carettere Scientifico) Neurological Institute ‘Carlo Besta’, 20126 Milan, Italy
| | - Johannes A. Mayr
- Department of Pediatrics, Paracelsus Medical University, A-5020 Salzburg, Austria
| | - Agnes Rötig
- INSERM U1163, Université Paris Descartes—Sorbonne Paris Cité, Institut Imagine, 75015 Paris, France
| | - Peter Freisinger
- Department of Pediatrics, Klinikum Reutlingen, 72764 Reutlingen, Germany
| | - Felix Distelmaier
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children’s Hospital, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany
| | - Tim M. Strom
- Institute of Human Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Institute of Human Genetics, Klinikum rechts der Isar, Technische Universität München, 81675 München, Germany
| | - Thomas Meitinger
- Institute of Human Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Institute of Human Genetics, Klinikum rechts der Isar, Technische Universität München, 81675 München, Germany
| | - Julien Gagneur
- Department of Informatics, Technische Universität München, 85748 Garching, Germany
- Quantitative Biosciences Munich, Gene Center, Department of Biochemistry, Ludwig Maximilian Universität München, 81377 München, Germany
| | - Holger Prokisch
- Institute of Human Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Institute of Human Genetics, Klinikum rechts der Isar, Technische Universität München, 81675 München, Germany
| |
Collapse
|