Transcriptome Alterations of Vascular Smooth Muscle Cells in Aortic Wall of Myocardial Infarction Patients

Total Page:16

File Type:pdf, Size:1020Kb

Transcriptome Alterations of Vascular Smooth Muscle Cells in Aortic Wall of Myocardial Infarction Patients This document is downloaded from DR‑NTU (https://dr.ntu.edu.sg) Nanyang Technological University, Singapore. Transcriptome alterations of vascular smooth muscle cells in aortic wall of myocardial infarction patients Wongsurawat, Thidathip; Woo, Chin Cheng; Giannakakis, Antonis; Lin, Xiao Yun; Cheow, Esther Sok Hwee; Lee, Chuen Neng; Richards, Mark; Sze, Siu Kwan; Nookaew, Intawat; Sorokin, Vitaly; Kuznetsov, Vladimir Andreevich 2018 Wongsurawat, T., Woo, C. C., Giannakakis, A., Lin, X. Y., Cheow, E. S. H., Lee, C. N., et al. (2018). Transcriptome alterations of vascular smooth muscle cells in aortic wall of myocardial infarction patients. Data in Brief, 17, 1112‑1135. https://hdl.handle.net/10356/85590 https://doi.org/10.1016/j.dib.2018.01.108 © 2018 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/). Downloaded on 09 Oct 2021 06:21:01 SGT Data in Brief 17 (2018) 1112–1135 Contents lists available at ScienceDirect Data in Brief journal homepage: www.elsevier.com/locate/dib Data Article Transcriptome alterations of vascular smooth muscle cells in aortic wall of myocardial infarction patients Thidathip Wongsurawat a,b, Chin Cheng Woo c, Antonis Giannakakis a, Xiao Yun Lin d, Esther Sok Hwee Cheow e, Chuen Neng Lee c,d, Mark Richards f,g, Siu Kwan Sze e, Intawat Nookaew b, Vladimir A. Kuznetsov a,h, Vitaly Sorokin c,d,⁎ a Department of Genome and Gene Expression Data Analysis, Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), Singapore 138671, Singapore b Department of Biomedical Informatics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA c Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore d Department of Cardiac, Thoracic and Vascular Surgery, National University Heart Centre, Singapore, National University Health System, Singapore 119228, Singapore e School of Biological Sciences, Nanyang Technological University, Singapore 639798, Singapore f Cardiovascular Research Institute, National University Heart Centre, Singapore, 119228, Singapore g Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore h School of Computer Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore article info abstract Article history: This article contains further data and information from our pub- Received 23 January 2018 lished manuscript [1]. We aim to identify significant transcriptome Accepted 30 January 2018 alterations of vascular smooth muscle cells (VSMCs) in the aortic Available online 6 February 2018 wall of myocardial infarction (MI) patients. Microarray gene ana- lysis was applied to evaluate VSMCs of MI and non-MI patients. Prediction Analysis of Microarray (PAM) identified genes that sig- nificantly discriminated the two groups of samples. Incorporation of gene ontology (GO) identified a VSMCs-associated classifier that DOI of original article: https://doi.org/10.1016/j.atherosclerosis.2018.01.024 ⁎ Corresponding author at: Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Sin- gapore 119228, Singapore. E-mail address: [email protected] (V. Sorokin). https://doi.org/10.1016/j.dib.2018.01.108 2352-3409/& 2018 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/). T. Wongsurawat et al. / Data in Brief 17 (2018) 1112–1135 1113 discriminated between the two groups of samples. Mass spectro- metry-based iTRAQ analysis revealed proteins significantly differ- entiating these two groups of samples. Ingenuity Pathway Analysis (IPA) revealed top pathways associated with hypoxia signaling in cardiovascular system. Enrichment analysis of these proteins sug- gested an activated pathway, and an integrated transcriptome- proteome pathway analysis revealed that it is the most implicated pathway. The intersection of the top candidate molecules from the transcriptome and proteome highlighted overexpression. & 2018 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/). Specifications Table Subject area Biology More specific sub- Genomics, Proteomics, Bioinformatics, Cardiovascular ject area Type of data Tables, figures How data was Microarray (Gene Titan Instrument, Affymetrix), mass spectrometry (LC-MS/ acquired MS system comprised of a Dionex Ultimate 3000 RSLC nano-HPLC system, coupled to an online Q-Exactive hybrid quadrupole-Orbitrap mass spectro- meter (Thermo Scientific, Hudson, NH, USA)), RT-qPCR (QuantStudio™ 12K Flex system (Life Technologies; Thermo Fisher Scientific Inc, USA)) Data format Raw, analyzed Experimental Laser capture microdissection, total RNA extraction and protein extraction factors from aortic tissues from surgical patients Experimental Data analysis with Principal Component Analysis (PCA), Prediction Analysis features of Microarray (PAM), Gene Ontology (GO), Ingenuity Pathway Analysis (IPA) Data source Singapore location Data accessibility Data is with this article. Value of the data Combination of multiple technologies and bioinformatics analysis performed in this study reveals the molecular changes induced by myocardial infarction on aortic smooth cells in humans. The alterations of the VSMCs transcriptome are congruent with alterations at the protein levels. Both levels show notably the up-regulation of the superoxide dismutase (SOD) with the activation of superoxide radical degradation pathway. Differentially expressed genes and pathways identified in these comparisons may be used in future experiments investigating response in myocardial infarction. 1. Data 1.1. Clinical analysis The characteristics of the myocardial infarction (MI) and non-MI samples undergoing tran- scriptomics and proteomics studies are presented in Tables 1(A) and 1(B) respectively. The baseline demographic and clinical characteristics of samples undergoing transcriptomics study were 1114 T. Wongsurawat et al. / Data in Brief 17 (2018) 1112–1135 Table 1A Demographic characteristics of MI and non-MI groups undergoing transcriptomics analysis. Characteristics Transcriptomics Transcriptomics p-value MI Non-MI (n¼17) (n ¼19) Ethnic Chinese 12 10 0.557 Malay 2 6 Indian 2 2 Others 1 1 Gender Male 14 16 0.881 Female 3 3 Age (Mean 7 SD) 59.53 7 8.28 59.68 7 8.85 0.957 Ejection Fraction Good (445%) 11 13 0.292 Fair (30–45%) 4 6 Poor (o30%) 2 0 Smoking No 8 9 0.985 Yes 9 10 Renal Impairment No 15 19 0.124 Yes 2 0 Diabetes Mellitus No 9 7 0.332 Yes 8 12 Hypertension No 1 3 0.345 Yes 16 16 Hyperlipidaemia No 0 0 – Yes 17 19 Antihyperlipidemic Medication No 0 0 – Yes 17 19 Troponin I (µg/L) 12.20 7 20.86 0.01 7 0.004 o0.05 (Mean 7 SD) (n¼15) (n ¼4) Table 1B Demographic characteristics of MI and non-MI proteomics groups. Characteristics Proteomics Proteomics p-value MI Non-MI n¼25 n¼25 Ethnic Chinese 11 13 0.745 Malay 8 7 Indian 5 5 Others 1 0 Gender Male 20 18 0.508 Female 5 7 Age (Mean 7 SD) 60.88712.34 61.6878.26 0.789 Ejection Fraction Good (445%) 14 16 0.344 Fair (30–45%) 9 9 Poor (o30%) 2 0 Smoking No 12 12 1 Yes 13 13 Renal Impairment No 25 25 NA Yes 0 0 Diabetes Mellitus No 10 9 0.771 Yes 15 16 Hypertension No 3 1 0.297 Yes 22 24 Hyperlipidaemia No 1 0 0.312 Yes 24 25 Antihyperlipidemic Medication No 4 1 0.157 Yes 21 24 Troponin I (µg/L) 19.54 7 19.24 0.015 7 0.006 o0.05 (Mean 7 SD) (n¼22) (n¼9) T. Wongsurawat et al. / Data in Brief 17 (2018) 1112–1135 1115 Table 2 Comparative demographic characteristics of transcriptomics and proteomics groups. Characteristics Transcriptomics Proteomics p-value n¼36 n¼50 Ethnic Chinese 22 24 0.404 Malay 8 15 Indian 4 10 Others 2 1 Gender Male 30 38 0.41 Female 6 12 Age (Mean 7 SD) 59.61 7 8.47 61.28 7 10.40 0.431 Ejection Fraction Good (445%) 24 30 0.708 Fair (30–45%) 10 18 Poor (o30%) 2 2 Smoking No 17 24 0.943 Yes 19 26 Renal Impairment No 34 50 0.092 Yes 2 0 Diabetes Mellitus No 16 19 0.548 Yes 20 31 Hypertension No 4 4 0.624 Yes 32 46 Hyperlipidaemia No 0 1 0.393 Yes 36 49 Antihyperlipidemic Medication No 0 5 0.051 Yes 36 45 Troponin I (µg/L) 9.63 719.09 13.87 7 18.45 0.445 (Mean 7 SD) (n¼19) (n ¼31) compared with those of the samples from the proteomics study (Table 2). In addition, the char- acteristics of the transcriptomic MI and non-MI samples with those of the independent cohorts comprising additional MI and non-MI patients undergoing RT-qPCR were compared (Tables 3 and 4). 1.2. Gene expression data analysis and class prediction by Prediction Analysis of Microarray (PAM) The samples were preprocessed through several steps, including quality assessment and outlier identification, normalization, batch effect correction and evaluation (Fig. 1). To interrogate differen- tially expressed genes between MI and non-MI we conducted gene-expression profiling using the Affymetrix U219 microarray platform. The R ‘limma’ package (https://www.bioconductor.org/help/ workflows/arrays/) identified 4,357 probe sets, selected at a ‘limma’-defined p-value o 0.05. Based on this set of differentially expressed genes (DEGs), we performed principal component analysis (PCA) (Fig. 1). To determine subgroup of genes distinguishing MI from non-MI subjects, we performed super- vised PAM [2] and identified a set of differentially expressed genes (DEGs) that discriminated between the two subtypes at Wilcox FDR o 0.1 (Table 5). Gene Ontology (GO) analysis of the DEGs was performed using DAVID Bioinformatics tools [3] (http://david.abcc.ncifcrf.gov/).
Recommended publications
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • Circular RNA Hsa Circ 0005114‑Mir‑142‑3P/Mir‑590‑5P‑ Adenomatous
    ONCOLOGY LETTERS 21: 58, 2021 Circular RNA hsa_circ_0005114‑miR‑142‑3p/miR‑590‑5p‑ adenomatous polyposis coli protein axis as a potential target for treatment of glioma BO WEI1*, LE WANG2* and JINGWEI ZHAO1 1Department of Neurosurgery, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033; 2Department of Ophthalmology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin 130021, P.R. China Received September 12, 2019; Accepted October 22, 2020 DOI: 10.3892/ol.2020.12320 Abstract. Glioma is the most common type of brain tumor APC expression with a good overall survival rate. UALCAN and is associated with a high mortality rate. Despite recent analysis using TCGA data of glioblastoma multiforme and the advances in treatment options, the overall prognosis in patients GSE25632 and GSE103229 microarray datasets showed that with glioma remains poor. Studies have suggested that circular hsa‑miR‑142‑3p/hsa‑miR‑590‑5p was upregulated and APC (circ)RNAs serve important roles in the development and was downregulated. Thus, hsa‑miR‑142‑3p/hsa‑miR‑590‑5p‑ progression of glioma and may have potential as therapeutic APC‑related circ/ceRNA axes may be important in glioma, targets. However, the expression profiles of circRNAs and their and hsa_circ_0005114 interacted with both of these miRNAs. functions in glioma have rarely been studied. The present study Functional analysis showed that hsa_circ_0005114 was aimed to screen differentially expressed circRNAs (DECs) involved in insulin secretion, while APC was associated with between glioma and normal brain tissues using sequencing the Wnt signaling pathway. In conclusion, hsa_circ_0005114‑ data collected from the Gene Expression Omnibus database miR‑142‑3p/miR‑590‑5p‑APC ceRNA axes may be potential (GSE86202 and GSE92322 datasets) and explain their mecha‑ targets for the treatment of glioma.
    [Show full text]
  • Discovery of Progenitor Cell Signatures by Time-Series Synexpression Analysis During Drosophila Embryonic Cell Immortalization
    Correction DEVELOPMENTAL BIOLOGY Correction for “Discovery of progenitor cell signatures by time- series synexpression analysis during Drosophila embryonic cell immortalization,” by Mary-Lee Dequéant, Delphine Fagegaltier, Yanhui Hu, Kerstin Spirohn, Amanda Simcox, Gregory J. Hannon, and Norbert Perrimon, which appeared in issue 42, October 20, 2015, of Proc Natl Acad Sci USA (112:12974–12979; first published October 5, 2015; 10.1073/pnas.1517729112). The authors note that Delphine Fagegaltier should be credited for designing research and performing research. The authors also note that Delphine Fagegaltier, Amanda Simcox, and Gregory J. Hannon should be credited for contributing to the writing of the paper. The corrected author contributions footnote appears below. Author contributions: M.-L.D., D.F., A.S., G.J.H., and N.P. designed research; M.-L.D., D.F., K.S., and A.S. performed research; M.-L.D., D.F., and Y.H. analyzed data; and M.-L.D. and N.P. wrote the paper with contributions from D.F., A.S., and G.J.H. www.pnas.org/cgi/doi/10.1073/pnas.1520482112 E6408 | PNAS | November 17, 2015 | vol. 112 | no. 46 www.pnas.org Downloaded by guest on September 25, 2021 Discovery of progenitor cell signatures by time-series synexpression analysis during Drosophila embryonic cell immortalization Mary-Lee Dequéanta,1, Delphine Fagegaltierb, Yanhui Hua, Kerstin Spirohna, Amanda Simcoxc, Gregory J. Hannond, and Norbert Perrimona,e,1 aDepartment of Genetics, Harvard Medical School, Boston, MA 02115, bCold Spring Harbor Laboratories, Cold Spring Harbor, NY 11724; cDepartment of Molecular Genetics, The Ohio State University, Columbus, OH 43210; dHoward Hughes Medical Institute, Cold Spring Harbor Laboratories, Cold Spring Harbor, NY 11724; and eHoward Hughes Medical Institute, Harvard Medical School, Boston, MA 02115 Contributed by Norbert Perrimon, September 10, 2015 (sent for review May 18, 2015; reviewed by Peter Cherbas, Gary Karpen, and Renato Paro) The use of time series profiling to identify groups of functionally population contributing to adult muscles (4–7).
    [Show full text]
  • Investigation of the Underlying Hub Genes and Molexular Pathogensis in Gastric Cancer by Integrated Bioinformatic Analyses
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.20.423656; this version posted December 22, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Investigation of the underlying hub genes and molexular pathogensis in gastric cancer by integrated bioinformatic analyses Basavaraj Vastrad1, Chanabasayya Vastrad*2 1. Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, Karnataka 582103, India. 2. Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karanataka, India. * Chanabasayya Vastrad [email protected] Ph: +919480073398 Chanabasava Nilaya, Bharthinagar, Dharwad 580001 , Karanataka, India bioRxiv preprint doi: https://doi.org/10.1101/2020.12.20.423656; this version posted December 22, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Abstract The high mortality rate of gastric cancer (GC) is in part due to the absence of initial disclosure of its biomarkers. The recognition of important genes associated in GC is therefore recommended to advance clinical prognosis, diagnosis and and treatment outcomes. The current investigation used the microarray dataset GSE113255 RNA seq data from the Gene Expression Omnibus database to diagnose differentially expressed genes (DEGs). Pathway and gene ontology enrichment analyses were performed, and a proteinprotein interaction network, modules, target genes - miRNA regulatory network and target genes - TF regulatory network were constructed and analyzed. Finally, validation of hub genes was performed. The 1008 DEGs identified consisted of 505 up regulated genes and 503 down regulated genes.
    [Show full text]
  • Supporting Information
    Supporting Information Pouryahya et al. SI Text Table S1 presents genes with the highest absolute value of Ricci curvature. We expect these genes to have significant contribution to the network’s robustness. Notably, the top two genes are TP53 (tumor protein 53) and YWHAG gene. TP53, also known as p53, it is a well known tumor suppressor gene known as the "guardian of the genome“ given the essential role it plays in genetic stability and prevention of cancer formation (1, 2). Mutations in this gene play a role in all stages of malignant transformation including tumor initiation, promotion, aggressiveness, and metastasis (3). Mutations of this gene are present in more than 50% of human cancers, making it the most common genetic event in human cancer (4, 5). Namely, p53 mutations play roles in leukemia, breast cancer, CNS cancers, and lung cancers, among many others (6–9). The YWHAG gene encodes the 14-3-3 protein gamma, a member of the 14-3-3 family proteins which are involved in many biological processes including signal transduction regulation, cell cycle pro- gression, apoptosis, cell adhesion and migration (10, 11). Notably, increased expression of 14-3-3 family proteins, including protein gamma, have been observed in a number of human cancers including lung and colorectal cancers, among others, suggesting a potential role as tumor oncogenes (12, 13). Furthermore, there is evidence that loss Fig. S1. The histogram of scalar Ricci curvature of 8240 genes. Most of the genes have negative scalar Ricci curvature (75%). TP53 and YWHAG have notably low of p53 function may result in upregulation of 14-3-3γ in lung cancer Ricci curvatures.
    [Show full text]
  • Inhibition of Mitochondrial Complex II in Neuronal Cells Triggers Unique
    www.nature.com/scientificreports OPEN Inhibition of mitochondrial complex II in neuronal cells triggers unique pathways culminating in autophagy with implications for neurodegeneration Sathyanarayanan Ranganayaki1, Neema Jamshidi2, Mohamad Aiyaz3, Santhosh‑Kumar Rashmi4, Narayanappa Gayathri4, Pulleri Kandi Harsha5, Balasundaram Padmanabhan6 & Muchukunte Mukunda Srinivas Bharath7* Mitochondrial dysfunction and neurodegeneration underlie movement disorders such as Parkinson’s disease, Huntington’s disease and Manganism among others. As a corollary, inhibition of mitochondrial complex I (CI) and complex II (CII) by toxins 1‑methyl‑4‑phenylpyridinium (MPP+) and 3‑nitropropionic acid (3‑NPA) respectively, induced degenerative changes noted in such neurodegenerative diseases. We aimed to unravel the down‑stream pathways associated with CII inhibition and compared with CI inhibition and the Manganese (Mn) neurotoxicity. Genome‑wide transcriptomics of N27 neuronal cells exposed to 3‑NPA, compared with MPP+ and Mn revealed varied transcriptomic profle. Along with mitochondrial and synaptic pathways, Autophagy was the predominant pathway diferentially regulated in the 3‑NPA model with implications for neuronal survival. This pathway was unique to 3‑NPA, as substantiated by in silico modelling of the three toxins. Morphological and biochemical validation of autophagy markers in the cell model of 3‑NPA revealed incomplete autophagy mediated by mechanistic Target of Rapamycin Complex 2 (mTORC2) pathway. Interestingly, Brain Derived Neurotrophic Factor
    [Show full text]
  • Appendix 2. Significantly Differentially Regulated Genes in Term Compared with Second Trimester Amniotic Fluid Supernatant
    Appendix 2. Significantly Differentially Regulated Genes in Term Compared With Second Trimester Amniotic Fluid Supernatant Fold Change in term vs second trimester Amniotic Affymetrix Duplicate Fluid Probe ID probes Symbol Entrez Gene Name 1019.9 217059_at D MUC7 mucin 7, secreted 424.5 211735_x_at D SFTPC surfactant protein C 416.2 206835_at STATH statherin 363.4 214387_x_at D SFTPC surfactant protein C 295.5 205982_x_at D SFTPC surfactant protein C 288.7 1553454_at RPTN repetin solute carrier family 34 (sodium 251.3 204124_at SLC34A2 phosphate), member 2 238.9 206786_at HTN3 histatin 3 161.5 220191_at GKN1 gastrokine 1 152.7 223678_s_at D SFTPA2 surfactant protein A2 130.9 207430_s_at D MSMB microseminoprotein, beta- 99.0 214199_at SFTPD surfactant protein D major histocompatibility complex, class II, 96.5 210982_s_at D HLA-DRA DR alpha 96.5 221133_s_at D CLDN18 claudin 18 94.4 238222_at GKN2 gastrokine 2 93.7 1557961_s_at D LOC100127983 uncharacterized LOC100127983 93.1 229584_at LRRK2 leucine-rich repeat kinase 2 HOXD cluster antisense RNA 1 (non- 88.6 242042_s_at D HOXD-AS1 protein coding) 86.0 205569_at LAMP3 lysosomal-associated membrane protein 3 85.4 232698_at BPIFB2 BPI fold containing family B, member 2 84.4 205979_at SCGB2A1 secretoglobin, family 2A, member 1 84.3 230469_at RTKN2 rhotekin 2 82.2 204130_at HSD11B2 hydroxysteroid (11-beta) dehydrogenase 2 81.9 222242_s_at KLK5 kallikrein-related peptidase 5 77.0 237281_at AKAP14 A kinase (PRKA) anchor protein 14 76.7 1553602_at MUCL1 mucin-like 1 76.3 216359_at D MUC7 mucin 7,
    [Show full text]
  • Functional Characterization of TBR1 Variants in Neurodevelopmental Disorder Received: 14 May 2018 Joery Den Hoed1, Elliot Sollis1, Hanka Venselaar2, Sara B
    www.nature.com/scientificreports OPEN Functional characterization of TBR1 variants in neurodevelopmental disorder Received: 14 May 2018 Joery den Hoed1, Elliot Sollis1, Hanka Venselaar2, Sara B. Estruch1, Pelagia Deriziotis1 & Accepted: 31 August 2018 Simon E. Fisher 1,3 Published: xx xx xxxx Recurrent de novo variants in the TBR1 transcription factor are implicated in the etiology of sporadic autism spectrum disorders (ASD). Disruptions include missense variants located in the T-box DNA- binding domain and previous work has demonstrated that they disrupt TBR1 protein function. Recent screens of thousands of simplex families with sporadic ASD cases uncovered additional T-box variants in TBR1 but their etiological relevance is unclear. We performed detailed functional analyses of de novo missense TBR1 variants found in the T-box of ASD cases, assessing many aspects of protein function, including subcellular localization, transcriptional activity and protein-interactions. Only two of the three tested variants severely disrupted TBR1 protein function, despite in silico predictions that all would be deleterious. Furthermore, we characterized a putative interaction with BCL11A, a transcription factor that was recently implicated in a neurodevelopmental syndrome involving developmental delay and language defcits. Our fndings enhance understanding of molecular functions of TBR1, as well as highlighting the importance of functional testing of variants that emerge from next-generation sequencing, to decipher their contributions to neurodevelopmental disorders like ASD. TBR1 (T-box brain, 1; OMIM *604616) encodes a neuron-specifc transcription factor of the T-box family1. Te TBR1 protein is highly expressed in the deep layers of the cortex, where it is involved in diferentiation of subsets of projection neurons2–4.
    [Show full text]
  • (12) Patent Application Publication (10) Pub. No.: US 2014/0304845 A1 Loboda Et Al
    US 201403.04845A1 (19) United States (12) Patent Application Publication (10) Pub. No.: US 2014/0304845 A1 Loboda et al. (43) Pub. Date: Oct. 9, 2014 54) ALZHEMIERS DISEASE SIGNATURE Publicationublication ClassificatiClassification MARKERS AND METHODS OF USE (51) Int. Cl. (71) Applicant: MERCKSHARP & DOHME CORP, CI2O I/68 (2006.01) Rahway, NJ (US) AOIK 67/027 (2006.01) (52) U.S. Cl. (72) Inventors: SIES yet.sS); CPC .......... CI2O I/6883 (2013.01); A0IK 67/0275 Icnaei NepoZnyn, Yeadon, (2013.01) le.East,Italia; David J. Stone, Wyncote, USPC .............. 800/12:536/23.5:536/23.2:506/17 (US); Keith Tanis, Quakertown, PA (US); William J. Ray, Juniper, FL (US) (57) ABSTRACT (21) Appl. No.: 14/354,622 Methods, biomarkers, and expression signatures are dis 1-1. closed for assessing the disease progression of Alzheimer's (22) PCT Filed: Oct. 26, 2012 disease (AD). In one embodiment, BioAge (biological age), NdStress (neurodegenerative stress), Alz (Alzheimer), and (86). PCT No.: PCT/US12A62218 Inflame (inflammation) are used as biomarkers of AD pro S371 (c)(1), gression. In another aspect, the invention comprises a gene (2), (4) Date: Apr. 28, 2014 signature for evaluating disease progression. In still another Related U.S. Application Data abiliticises in (60) Provisional application No. 61/553,400, filed on Oct. used to identify animal models for use in the development and 31, 2011. evaluation of therapeutics for the treatment of AD. Patent Application Publication Oct. 9, 2014 Sheet 1 of 16 US 2014/0304845 A1 Y : : O O O O O O O O D O v v CN On cy Patent Application Publication Oct.
    [Show full text]
  • Circular RNA Expression Profiles in Pediatric Ependymomas Ulvi Ahmadov1, Meile M
    medRxiv preprint doi: https://doi.org/10.1101/2020.08.04.20167312; this version posted August 5, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. All rights reserved. No reuse allowed without permission. Circular RNA expression profiles in pediatric ependymomas Ulvi Ahmadov1, Meile M. Bendikas2, Karoline K. Ebbesen2,3, Astrid M. Sehested4, Jørgen Kjems2,3, Helle Broholm5 and Lasse S. Kristensen1# 1. Department of Biomedicine, Aarhus University, Aarhus, Denmark 2. Molecular Biology and Genetics (MBG), Aarhus University, Aarhus, Denmark 3. Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus, Denmark 4. Department of Pediatrics and Adolescent Medicine, Copenhagen University Hospital, Copenhagen, Denmark 5. Department of Pathology, Center of Diagnostic Investigation, Rigshospitalet, Copenhagen, Denmark # corresponding author Running title: CircRNAs expression profiles in pediatric ependymomas Correspondence should be addressed to: Lasse Sommer Kristensen, PhD, Department of Biomedicine, Høegh- Guldbergs Gade 10, building 1116, room 268, Aarhus University, 8000 Aarhus, Denmark. Phone: +45 28880562, E-mail: [email protected] Key words: Pediatric ependymoma, pilocytic astrocytoma, medulloblastoma, circular RNA, RNA-sequencing, NanoString nCounter 1 NOTE: This preprint reports new research that has not been certified by peer review and should not be used to guide clinical practice. medRxiv preprint doi: https://doi.org/10.1101/2020.08.04.20167312; this version posted August 5, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity.
    [Show full text]
  • Reduction of BCL11A in Hematopoietic Stem Cells Through
    Science Bulletin 64 (2019) 1562–1564 Contents lists available at ScienceDirect Science Bulletin journal homepage: www.elsevier.com/locate/scib Research Highlight Reduction of BCL11A in hematopoietic stem cells through gene editing: new strategy to ameliorate the severe b-globin disorders sickle cell disease ⇑ Weiqi Hong, Mengyuan Huang, Yuquan Wei, Xiawei Wei Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China Site-specific gene editing is of great importance in precise Sickle-cell anemia is a prototypical monogenic disorder caused medicine. Two conventional genome editing methods, Zine finger by mutation of b-globin subunit. It is a promising therapy strategy nucleases (ZFNs) and transcription activator-like effector nucleases to induct fetal hemoglobin (HbF, a2c2) by re-expressing the paral- (TALENs), are based on protein-DNA recognition, with tedious ogous c-globin genes (HBG1/2) for severe b-globin disorders sickle work in constructing target protein [1,2]. Developed from immune cell disease (SCD) and b-thalassemia [9]. Researches in the past response of bacteria, CRISPR/Cas9 has been widely investigated as have shown that the core of the +58 erythroid enhancer of BCL11A a promising tool for therapeutic genome editing in clinical settings was crucial for repression of HBF in adult stage erythroid. Wu et al nowadays [3,4]. This system succeeds in gene deletion, insertion found that chemically modified synthetic sgRNAs (MS-sgRNAs) and frameshift mutations with higher efficiency, less cost, was more efficient than in vitro transcribed sgRNAs. Targeting improved flexibility and simplified designing process [5].
    [Show full text]
  • Characterization of Transcription Factor Networks Involved in Umbilical Cord Blood CD34+ Stem Cells-Derived Erythropoiesis
    Characterization of Transcription Factor Networks Involved in Umbilical Cord Blood CD34+ Stem Cells-Derived Erythropoiesis Biaoru Li1, Lianghao Ding2, Chinrang Yang2, Baolin Kang1, Li Liu3, Michael D. Story2, Betty S. Pace1* 1 Department of Pediatrics, Hematology/Oncology Division, Georgia Regents University, Augusta, Georgia, United States of America, 2 Department of Radiation Oncology and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America, 3 Department of Molecular and Cell Biology, University of Texas at Dallas, Richardson, Texas, United States of America Abstract Fetal stem cells isolated from umbilical cord blood (UCB) possess a great capacity for proliferation and differentiation and serve as a valuable model system to study gene regulation. Expanded knowledge of the molecular control of hemoglobin synthesis will provide a basis for rational design of therapies for b-hemoglobinopathies. Transcriptome data are available for erythroid progenitors derived from adult stem cells, however studies to define molecular mechanisms controlling globin gene regulation during fetal erythropoiesis are limited. Here, we utilize UCB-CD34+ stem cells induced to undergo erythroid differentiation to characterize the transcriptome and transcription factor networks (TFNs) associated with the c/b-globin switch during fetal erythropoiesis. UCB-CD34+ stem cells grown in the one-phase liquid culture system displayed a higher proliferative capacity than adult CD34+ stem cells. The c/b-globin switch was observed after day 42 during fetal erythropoiesis in contrast to adult progenitors where the switch occurred around day 21. To gain insights into transcription factors involved in globin gene regulation, microarray analysis was performed on RNA isolated from UCB-CD34+ cell-derived erythroid progenitors harvested on day 21, 42, 49 and 56 using the HumanHT-12 Expression BeadChip.
    [Show full text]