1
|
Villani RM, McKenzie ME, Davidson AL, Spurdle AB. Regional-specific calibration enables application of computational evidence for clinical classification of 5' cis-regulatory variants in Mendelian disease. Am J Hum Genet 2024; 111:1301-1315. [PMID: 38815586 PMCID: PMC11267523 DOI: 10.1016/j.ajhg.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 05/02/2024] [Accepted: 05/03/2024] [Indexed: 06/01/2024] Open
Abstract
To date, clinical genetic testing for Mendelian disease variants has focused heavily on exonic coding and intronic gene regions. This multi-step study was undertaken to provide an evidence base for selecting and applying computational approaches for use in clinical classification of 5' cis-regulatory region variants. Curated datasets of clinically reported disease-causing 5' cis-regulatory region variants and variants from matched genomic regions in population controls were used to calibrate six bioinformatic tools as predictors of variant pathogenicity. Likelihood ratio estimates were aligned to code weights following ClinGen recommendations for application of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology (ACMG/AMP) classification scheme. Considering code assignment across all reference dataset variants, performance was best for CADD (81.2%) and REMM (81.5%). Optimized thresholds provided moderate evidence toward pathogenicity (CADD, REMM) and moderate (CADD) or supporting (REMM) evidence against pathogenicity. Both sensitivity and specificity of prediction were improved when further categorizing variants based on location in an EPDnew-defined promoter region. Combining predictions (CADD, REMM, and location in a promoter region) increased specificity at the expense of sensitivity. Importantly, the optimal CADD thresholds for assigning ACMG/AMP codes PP3 (≥10) and BP4 (≤8) were vastly different from recommendations for protein-coding variants (PP3 ≥25.3; BP4 ≤22.7); CADD <22.7 would incorrectly assign BP4 for >90% of reported disease-causing cis-regulatory region variants. Our results demonstrate the need to consider a tiered approach and tailored score thresholds to optimize bioinformatic impact prediction for clinical classification of 5' cis-regulatory region variants.
Collapse
Affiliation(s)
- Rehan M Villani
- Population Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Maddison E McKenzie
- Population Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Aimee L Davidson
- Population Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Amanda B Spurdle
- Population Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
2
|
Jin R, Zhang H, Lin C, Guo J, Zou W, Chen Z, Liu H. Inhibition of miR338 rescues cleidocranial dysplasia in Runx2 mutant mice partially via the Hif1a-Vegfa axis. Exp Mol Med 2023; 55:69-80. [PMID: 36599929 PMCID: PMC9898552 DOI: 10.1038/s12276-022-00914-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 11/01/2022] [Accepted: 11/08/2022] [Indexed: 01/06/2023] Open
Abstract
Haploinsufficiency of Runt-related transcription factor-2 (RUNX2) is responsible for cleidocranial dysplasia (CCD), a rare hereditary disease with a range of defects, including delayed closure of the cranial sutures and short stature. Symptom-based treatments, such as a combined surgical-orthodontic approach, are commonly used to treat CCD patients. However, there have been few reports of treatments based on Runx2-specific regulation targeting dwarfism symptoms. Previously, we found that the miR338 cluster, a potential diagnostic and therapeutic target for postmenopausal osteoporosis, could directly target Runx2 during osteoblast differentiation in vitro. Here, we generated miR338-/-;Runx2+/- mice to investigate whether inhibition of miR338 could rescue CCD defects caused by Runx2 mutation in vivo. We found that the dwarfism phenotype caused by Runx2 haploinsufficiency was recovered in miR338-/-;Runx2+/- mice, with complete bone density restoration and quicker closure of fontanels. Single-cell RNA-seq analysis revealed that knockout of miR338 specifically rescued the osteoblast lineage priming ability of bone marrow stromal cells in Runx2+/- femurs, which was further confirmed by Osterix-specific conditional knockout of miR338 in Runx2+/- mice (OsxCre; miR338 fl/fl;Runx2+/-). Mechanistically, ablation of the miR338 cluster in Runx2+/- femurs directly rescued the Hif1a-Vegfa pathway in Runx2+/- osteoblasts, as proven by gene expression profiles and ChIP and Re-ChIP assays. Collectively, our data revealed the genetic interaction between Runx2 and the miR338 cluster during osteoblast differentiation and implied that the miR338 cluster could be a potential therapeutic target for CCD.
Collapse
Affiliation(s)
- Runze Jin
- grid.49470.3e0000 0001 2331 6153The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, 430079 China
| | - Hanshu Zhang
- grid.49470.3e0000 0001 2331 6153The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, 430079 China
| | - Chujiao Lin
- grid.49470.3e0000 0001 2331 6153The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, 430079 China ,grid.168645.80000 0001 0742 0364Division of Rheumatology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605 USA
| | - Jinqiang Guo
- grid.49470.3e0000 0001 2331 6153The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, 430079 China
| | - Weiguo Zou
- grid.410726.60000 0004 1797 8419State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Zhi Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, 430079, China.
| | - Huan Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan, 430079, China. .,Department of Periodontology, School of Stomatology, Wuhan University, Wuhan, 430079, China. .,Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
| |
Collapse
|
3
|
Ahmed F, Gi Ho S, Samantasinghar A, Memon FH, Rahim CSA, Soomro AM, Pratibha, Sunildutt N, Kim KH, Choi KH. Drug repurposing in psoriasis, performed by reversal of disease-associated gene expression profiles. Comput Struct Biotechnol J 2022; 20:6097-6107. [DOI: 10.1016/j.csbj.2022.10.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/09/2022] [Accepted: 10/30/2022] [Indexed: 11/10/2022] Open
|
4
|
Legüe M. Relevancia de los mecanismos epigenéticos en el neurodesarrollo normal y consecuencias de sus perturbaciones. REVISTA MÉDICA CLÍNICA LAS CONDES 2022. [DOI: 10.1016/j.rmclc.2022.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
5
|
Liu Y. scDeconv: an R package to deconvolve bulk DNA methylation data with scRNA-seq data and paired bulk RNA-DNA methylation data. Brief Bioinform 2022; 23:6572659. [PMID: 35453146 PMCID: PMC9271220 DOI: 10.1093/bib/bbac150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/26/2022] [Accepted: 04/04/2022] [Indexed: 11/14/2022] Open
Abstract
Many DNA methylation (DNAm) data are from tissues composed of various cell types, and hence cell deconvolution methods are needed to infer their cell compositions accurately. However, a bottleneck for DNAm data is the lack of cell-type-specific DNAm references. On the other hand, scRNA-seq data are being accumulated rapidly with various cell-type transcriptomic signatures characterized, and also, many paired bulk RNA-DNAm data are publicly available currently. Hence, we developed the R package scDeconv to use these resources to solve the reference deficiency problem of DNAm data and deconvolve them from scRNA-seq data in a trans-omics manner. It assumes that paired samples have similar cell compositions. So the cell content information deconvolved from the scRNA-seq and paired RNA data can be transferred to the paired DNAm samples. Then an ensemble model is trained to fit these cell contents with DNAm features and adjust the paired RNA deconvolution in a co-training manner. Finally, the model can be used on other bulk DNAm data to predict their relative cell-type abundances. The effectiveness of this method is proved by its accurate deconvolution on the three testing datasets here, and if given an appropriate paired dataset, scDeconv can also deconvolve other omics, such as ATAC-seq data. Furthermore, the package also contains other functions, such as identifying cell-type-specific inter-group differential features from bulk DNAm data. scDeconv is available at: https://github.com/yuabrahamliu/scDeconv.
Collapse
Affiliation(s)
- Yu Liu
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| |
Collapse
|
6
|
Beck EA, Healey HM, Small CM, Currey MC, Desvignes T, Cresko WA, Postlethwait JH. Advancing human disease research with fish evolutionary mutant models. Trends Genet 2022; 38:22-44. [PMID: 34334238 PMCID: PMC8678158 DOI: 10.1016/j.tig.2021.07.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 01/03/2023]
Abstract
Model organism research is essential to understand disease mechanisms. However, laboratory-induced genetic models can lack genetic variation and often fail to mimic the spectrum of disease severity. Evolutionary mutant models (EMMs) are species with evolved phenotypes that mimic human disease. EMMs complement traditional laboratory models by providing unique avenues to study gene-by-environment interactions, modular mutations in noncoding regions, and their evolved compensations. EMMs have improved our understanding of complex diseases, including cancer, diabetes, and aging, and illuminated mechanisms in many organs. Rapid advancements of sequencing and genome-editing technologies have catapulted the utility of EMMs, particularly in fish. Fish are the most diverse group of vertebrates, exhibiting a kaleidoscope of specialized phenotypes, many that would be pathogenic in humans but are adaptive in the species' specialized habitat. Importantly, evolved compensations can suggest avenues for novel disease therapies. This review summarizes current research using fish EMMs to advance our understanding of human disease.
Collapse
Affiliation(s)
- Emily A Beck
- Data Science, University of Oregon, Eugene, OR 97403, USA; Institute of Ecology and Evolution, University of Oregon, Eugene, OR 97403, USA.
| | - Hope M Healey
- Institute of Ecology and Evolution, University of Oregon, Eugene, OR 97403, USA
| | - Clayton M Small
- Data Science, University of Oregon, Eugene, OR 97403, USA; Institute of Ecology and Evolution, University of Oregon, Eugene, OR 97403, USA
| | - Mark C Currey
- Institute of Ecology and Evolution, University of Oregon, Eugene, OR 97403, USA
| | - Thomas Desvignes
- Institute of Neuroscience, University of Oregon, Eugene, OR 97403, USA
| | - William A Cresko
- Data Science, University of Oregon, Eugene, OR 97403, USA; Institute of Ecology and Evolution, University of Oregon, Eugene, OR 97403, USA
| | | |
Collapse
|
7
|
Saraiva LC, Cappi C, Simpson HB, Stein DJ, Viswanath B, van den Heuvel OA, Reddy YCJ, Miguel EC, Shavitt RG. Cutting-edge genetics in obsessive-compulsive disorder. Fac Rev 2020; 9:30. [PMID: 33659962 PMCID: PMC7886082 DOI: 10.12703/r/9-30] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
This article reviews recent advances in the genetics of obsessive-compulsive disorder (OCD). We cover work on the following: genome-wide association studies, whole-exome sequencing studies, copy number variation studies, gene expression, polygenic risk scores, gene–environment interaction, experimental animal systems, human cell models, imaging genetics, pharmacogenetics, and studies of endophenotypes. Findings from this work underscore the notion that the genetic architecture of OCD is highly complex and shared with other neuropsychiatric disorders. Also, the latest evidence points to the participation of gene networks involved in synaptic transmission, neurodevelopment, and the immune and inflammatory systems in this disorder. We conclude by highlighting that further study of the genetic architecture of OCD, a great part of which remains to be elucidated, could benefit the development of diagnostic and therapeutic approaches based on the biological basis of the disorder. Studies to date revealed that OCD is not a simple homogeneous entity, but rather that the underlying biological pathways are variable and heterogenous. We can expect that translation from bench to bedside, through continuous effort and collaborative work, will ultimately transform our understanding of what causes OCD and thus how best to treat it.
Collapse
Affiliation(s)
- Leonardo Cardoso Saraiva
- Department & Institute of Psychiatry, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Carolina Cappi
- Department & Institute of Psychiatry, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Helen Blair Simpson
- Columbia University Irving Medical Center, Columbia University, New York, NY, 10032, USA
- The New York State Psychiatric Institute, New York, NY, 10032, USA
| | - Dan J Stein
- SA MRC Unit on Risk & Resilience in Mental Disorders, Department of Psychiatry & Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Biju Viswanath
- Molecular Genetics Laboratory, National Institute of Mental Health & Neurosciences (NIMHANS); Accelerator Program for Discovery in Brain disorders using Stem cells (ADBS) Laboratory, NIMHANS, Bangalore, India
| | - Odile A van den Heuvel
- Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Department of Psychiatry, Department of Anatomy & Neuroscience, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - YC Janardhan Reddy
- Obsessive-Compulsive Disorder (OCD) Clinic, Department of Psychiatry, NIMHANS, Bangalore, India
| | - Euripedes C Miguel
- Department & Institute of Psychiatry, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Roseli G Shavitt
- Department & Institute of Psychiatry, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| |
Collapse
|
8
|
Kim WJ, Shin HL, Kim BS, Kim HJ, Ryoo HM. RUNX2-modifying enzymes: therapeutic targets for bone diseases. Exp Mol Med 2020; 52:1178-1184. [PMID: 32788656 PMCID: PMC8080656 DOI: 10.1038/s12276-020-0471-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/21/2020] [Accepted: 05/22/2020] [Indexed: 01/01/2023] Open
Abstract
RUNX2 is a master transcription factor of osteoblast differentiation. RUNX2 expression in the bone and osteogenic front of a suture is crucial for cranial suture closure and membranous bone morphogenesis. In this manner, the regulation of RUNX2 is precisely controlled by multiple posttranslational modifications (PTMs) mediated by the stepwise recruitment of multiple enzymes. Genetic defects in RUNX2 itself or in its PTM regulatory pathways result in craniofacial malformations. Haploinsufficiency in RUNX2 causes cleidocranial dysplasia (CCD), which is characterized by open fontanelle and hypoplastic clavicles. In contrast, gain-of-function mutations in FGFRs, which are known upstream stimulating signals of RUNX2 activity, cause craniosynostosis (CS) characterized by premature suture obliteration. The identification of these PTM cascades could suggest suitable drug targets for RUNX2 regulation. In this review, we will focus on the mechanism of RUNX2 regulation mediated by PTMs, such as phosphorylation, prolyl isomerization, acetylation, and ubiquitination, and we will summarize the therapeutics associated with each PTM enzyme for the treatment of congenital cranial suture anomalies.
Collapse
Affiliation(s)
- Woo-Jin Kim
- Basic Research Lab for "Epigenetic Regeneration of Aged Skeleto-Muscular System (ERASMUS)", Department of Molecular Genetics and Dental Pharmacology, School of Dentistry, Dental Research Institute, Seoul National University, Seoul, South Korea
| | - Hye-Lim Shin
- Basic Research Lab for "Epigenetic Regeneration of Aged Skeleto-Muscular System (ERASMUS)", Department of Molecular Genetics and Dental Pharmacology, School of Dentistry, Dental Research Institute, Seoul National University, Seoul, South Korea
| | - Bong-Soo Kim
- Basic Research Lab for "Epigenetic Regeneration of Aged Skeleto-Muscular System (ERASMUS)", Department of Molecular Genetics and Dental Pharmacology, School of Dentistry, Dental Research Institute, Seoul National University, Seoul, South Korea
| | - Hyun-Jung Kim
- Basic Research Lab for "Epigenetic Regeneration of Aged Skeleto-Muscular System (ERASMUS)", Department of Molecular Genetics and Dental Pharmacology, School of Dentistry, Dental Research Institute, Seoul National University, Seoul, South Korea
| | - Hyun-Mo Ryoo
- Basic Research Lab for "Epigenetic Regeneration of Aged Skeleto-Muscular System (ERASMUS)", Department of Molecular Genetics and Dental Pharmacology, School of Dentistry, Dental Research Institute, Seoul National University, Seoul, South Korea.
| |
Collapse
|
9
|
Gordon JL, Reynolds MM, Brown MA. Nitric Oxide as a Potential Adjuvant Therapeutic for Neuroblastoma: Effects of NO on Murine N2a Cells. Vet Sci 2020; 7:E51. [PMID: 32340209 PMCID: PMC7355501 DOI: 10.3390/vetsci7020051] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 04/16/2020] [Accepted: 04/19/2020] [Indexed: 12/24/2022] Open
Abstract
Neuroblastoma, the most common extracranial solid tumor in children, accounts for 15% of all pediatric cancer deaths. Pharmaceutical applications of S-Nitrosylation, which, under normal conditions is involved with a host of epigenetic and embryological development pathways, have exhibited great potential for use as adjuvant therapeutics in the clinical management of cancer. Herein, an evaluation of the impact of nitric oxide (NO) as a potent anticancer agent on murine neuroblastoma cells is presented. Excitingly cell viability, colony formation, and non-carcinogenic cell analysis illustrate the significance and practicality of NO as a cytotoxic anticancer therapeutic. Resazurin, WST-8 (2-(2-methoxy-4-nitrophenyl)-3-(4-nitrophenyl)-5-(2,4-disulfophenyl)-2H-tetrazolium, monosodium salt), and MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-diphyltetrazolium bromide) assays consistently displayed a moderate, ~20-25% reduction in cell viability after exposure to 1 mM S-Nitrosoglutathione (GSNO). A colony formation assay demonstrated that treated cells no longer exhibited colony formation capacity. Identically GSNO-treated Adult Human Dermal Fibroblasts (HDFa) exhibited no decrease in viability, indicating potential discrimination between neoplastic and normal cells. Collectively, our findings indicate a potential application for NO as an adjuvant therapeutic in the clinical management of neuroblastoma.
Collapse
Affiliation(s)
- Jenna L. Gordon
- Department of Chemistry, Colorado State University, Fort Collins, CO 80521, USA;
| | - Melissa M. Reynolds
- Department of Chemistry, School of Biomedical Engineering, Colorado State University, Campus Delivery 1872, Fort Collins, CO 80523, USA
| | - Mark A. Brown
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523, USA;
| |
Collapse
|
10
|
Perenthaler E, Yousefi S, Niggl E, Barakat TS. Beyond the Exome: The Non-coding Genome and Enhancers in Neurodevelopmental Disorders and Malformations of Cortical Development. Front Cell Neurosci 2019; 13:352. [PMID: 31417368 PMCID: PMC6685065 DOI: 10.3389/fncel.2019.00352] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 07/16/2019] [Indexed: 12/22/2022] Open
Abstract
The development of the human cerebral cortex is a complex and dynamic process, in which neural stem cell proliferation, neuronal migration, and post-migratory neuronal organization need to occur in a well-organized fashion. Alterations at any of these crucial stages can result in malformations of cortical development (MCDs), a group of genetically heterogeneous neurodevelopmental disorders that present with developmental delay, intellectual disability and epilepsy. Recent progress in genetic technologies, such as next generation sequencing, most often focusing on all protein-coding exons (e.g., whole exome sequencing), allowed the discovery of more than a 100 genes associated with various types of MCDs. Although this has considerably increased the diagnostic yield, most MCD cases remain unexplained. As Whole Exome Sequencing investigates only a minor part of the human genome (1-2%), it is likely that patients, in which no disease-causing mutation has been identified, could harbor mutations in genomic regions beyond the exome. Even though functional annotation of non-coding regions is still lagging behind that of protein-coding genes, tremendous progress has been made in the field of gene regulation. One group of non-coding regulatory regions are enhancers, which can be distantly located upstream or downstream of genes and which can mediate temporal and tissue-specific transcriptional control via long-distance interactions with promoter regions. Although some examples exist in literature that link alterations of enhancers to genetic disorders, a widespread appreciation of the putative roles of these sequences in MCDs is still lacking. Here, we summarize the current state of knowledge on cis-regulatory regions and discuss novel technologies such as massively-parallel reporter assay systems, CRISPR-Cas9-based screens and computational approaches that help to further elucidate the emerging role of the non-coding genome in disease. Moreover, we discuss existing literature on mutations or copy number alterations of regulatory regions involved in brain development. We foresee that the future implementation of the knowledge obtained through ongoing gene regulation studies will benefit patients and will provide an explanation to part of the missing heritability of MCDs and other genetic disorders.
Collapse
Affiliation(s)
| | | | | | - Tahsin Stefan Barakat
- Department of Clinical Genetics, Erasmus MC – University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
11
|
Alshiraihi I, Brown MA. Epigenetic Factors of Disease. Diseases 2019; 7:diseases7020042. [PMID: 31197091 PMCID: PMC6630624 DOI: 10.3390/diseases7020042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Accepted: 06/10/2019] [Indexed: 11/21/2022] Open
Abstract
The development of tissues involves the direction of specific programs for gene expression among distinct cell types. These programs are often established in a heritable state by virtue of epigenetic mechanisms and corresponding pathways of cellular memory. Thus, the broad synchronization in patterns of gene expression ultimately dictates cellular consequences. Aberrations in these epigenetic mechanisms are known to be associated with a range of diseases. Herein, we highlight epigenetic factors that, when aberrantly expressed, lead to a broad range of diseases. Further, we call upon the community of biomedical researchers to share their findings related to the epigenetic factors of disease.
Collapse
Affiliation(s)
- Ilham Alshiraihi
- Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO 80523, USA.
| | - Mark A Brown
- Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO 80523, USA.
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523, USA.
- Epidemiology Section, Colorado School of Public Health, Fort Collins, CO 80523, USA.
- Department of Ethnic Studies, Colorado State University, Fort Collins, CO 80523, USA.
| |
Collapse
|