Characterization of the Development of the Mouse Cochlear Epithelium at the Single Cell Level

Total Page:16

File Type:pdf, Size:1020Kb

Characterization of the Development of the Mouse Cochlear Epithelium at the Single Cell Level ARTICLE https://doi.org/10.1038/s41467-020-16113-y OPEN Characterization of the development of the mouse cochlear epithelium at the single cell level Likhitha Kolla1,6, Michael C. Kelly1,6, Zoe F. Mann 2, Alejandro Anaya-Rocha1, Kathryn Ellis1, Abigail Lemons1, Adam T. Palermo 3, Kathy S. So3, Joseph C. Mays 1, Joshua Orvis4, Joseph C. Burns3, Ronna Hertzano4,5, ✉ Elizabeth C. Driver 1 & Matthew W. Kelley 1 Mammalian hearing requires the development of the organ of Corti, a sensory epithelium 1234567890():,; comprising unique cell types. The limited number of each of these cell types, combined with their close proximity, has prevented characterization of individual cell types and/or their developmental progression. To examine cochlear development more closely, we tran- scriptionally profile approximately 30,000 isolated mouse cochlear cells collected at four developmental time points. Here we report on the analysis of those cells including the identification of both known and unknown cell types. Trajectory analysis for OHCs indicates four phases of gene expression while fate mapping of progenitor cells suggests that OHCs and their surrounding supporting cells arise from a distinct (lateral) progenitor pool. Tgfβr1 is identified as being expressed in lateral progenitor cells and a Tgfβr1 antagonist inhibits OHC development. These results provide insights regarding cochlear development and demon- strate the potential value and application of this data set. 1 Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA. 2 Centre for Craniofacial and Regenerative Biology, King’s College London, London, UK. 3 Decibel Therapeutics, 1325 Boylston, Str., Suite 500, Boston, MA 02215, USA. 4 Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA. 5 Department of Otorhinolaryngology Head and Neck Surgery, Anatomy and Neurobiology, and Institute for Genome Sciences, University of Maryland School of Medicine, ✉ Baltimore, MD 21201, USA. 6These authors contributed equally: Likhitha Kolla, Michael C. Kelly. email: [email protected] NATURE COMMUNICATIONS | (2020) 11:2389 | https://doi.org/10.1038/s41467-020-16113-y | www.nature.com/naturecommunications 1 ARTICLE NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-020-16113-y he organ of Corti (OC), located in the floor of the scala manipulations and molecular genetic experiments suggest that Tmedia of the cochlea, acts as the primary sensory trans- otocyst precursor cells proceed through several rounds of lineage ducer of sound in mammals. This structure comprises a restriction that progressively specify subsets of cells as prosensory highly diverse cellular mosaic that includes two different types of cells, and ultimately as either HCs or SCs (ref. 1). mechanosensory hair cells (HCs), and an undefined number of To examine the transcriptional changes that occur during the associated supporting cell (SC) types (Fig. 1a). All of these cells formation of the OC, we dissociate cochlear duct cells at four are believed to arise from a developmental equivalence group developmental time points and then capture individual cells for referred to as the prosensory domain1. The results of embryologic analysis using single-cell RNAseq. Results identify multiple a c IPC Medial Lateral 40 HeC DC3 CC/OSC OPC HC 20 DC1/2 KO4 KO1 ISC KO2 0 tSNE 2 Inner hair cells Deiters cells IdC Outer hair cells Hensen’s cells IPhC –20 Inner phalangeal/ Claudius/ OC90+ border cells outer sulcus KO3 Inner pillar cells cells –25 0 25 Outer pillar cells KO tSNE 1 b d Acbd7 Ccer2 Cib2 Pcp4 Pvalb 4 5 4 5 4 3 4 3 4 3 3 3 2 2 2 2 2 HC 1 1 1 1 1 0 0 0 0 0 Anxa5 Fabp7 Gjb2 Matn4 Prss23 5 5 5 4 4 4 4 4 3 3 3 3 3 2 2 2 2 2 IPhC 1 1 1 1 1 0 0 0 0 0 Cryab Emid1 Igfbpl1 Npy S100b 5 4 6 4 3 3 3 3 4 2 2 2 2 2 1 1 1 1 0 0 0 0 0 Fam159b Ppp1r2 S100b Serpine2 Sm agp 4 4 3 3 3 3 3 2 2 2 2 2 1 1 1 OPC 1 1 0 0 0 0 0 Hes5 Pdzk1ip1 Ppp1r2 S100a1 Serpine2 4 4 4 4 4 3 3 3 3 3 2 2 2 2 2 1 1 1 1 1 DC1/2 0 0 0 0 0 Hes5 Igfbpl1 Lfng Prss23 S100a1 4 4 3 3 4 3 3 3 2 2 2 2 2 DC3 1 1 1 1 1 0 0 0 0 0 Egfl6 Fam159b Fst Nupr1 Pm ch 3 4 3 4 4 2 3 3 3 2 2 2 2 HeC1 1 1 IPC 1 1 0 0 0 0 0 Apoe Bmp4 Fbln2 Fst Npnt 4 4 5 3 4 3 3 4 2 3 3 2 2 2 2 1 1 1 1 1 0 0 0 0 0 CC/OSC Dcn Ddost Pdia6 Rcn3 Sdf2l1 HC IS 4 5 4 DC3 IdC Log-transformed expression level IPhCOPC 4 4 3 KO4 3 3 3 KO1 KO2 KO3 3 2 CC/OSC 2 2 2 2 1 IPC 1 1 1 1 HeC Oc90 0 0 0 0 0 DC1/2 Cpxm2 Ctgf Fkbp9 Kazald1 Tectb 4 4 4 4 5 3 3 4 3 3 3 e 2 2 2 2 2 P1 KO cells only 1 1 1 1 1 KO20 KO1 0 0 0 0 M.KO Cst3 Gjb6 Net1 Tectb Tsen15 L.KO1 Id 5 20 C 4 5 L.KO2 3 3 4 IS 4 3 IS 3 M 3 2 2 L.KO3 .K 2 IdC O L.KO 2 2 1 1 1 2 3 1 1 1 0 KO3 0 0 0 0 0 Calb1 Crabp1 Epyc Itm2a Stmn2 tSNE 2 4 4 5 4 3 3 4 3 –20 3 3 2 2 2 2 2 CALB1 Phal 1 1 1 1 1 KO4 0 0 0 0 0 KO Igf1 Matn1 Meg3 Rgcc Tm4sf1 0 25 -25 4 4 4 3 4 tSNE 1 3 3 3 3 2 2 2 2 2 FABP7 Pou4f3 ISC 1 1 1 1 1 Otoa Calb1 Fabp7 0 0 0 0 0 KO Cndp2 Oc90 Rnase1 Ttr Vmo1 5 6 4 6 5 5 3 4 4 4 4 3 3 3 2 2 2 2 2 FABP7 1 1 1 1 OC90+ 0 0 0 0 0 Cdkn1c Fxyd6 Otoa Ptgds Smoc2 tSNE 2 6 4 4 4 4 4 3 3 3 3 2 2 2 2 IdC 2 1 1 1 1 0 0 0 0 0 tSNE 1 2 NATURE COMMUNICATIONS | (2020) 11:2389 | https://doi.org/10.1038/s41467-020-16113-y | www.nature.com/naturecommunications NATURE COMMUNICATIONS | https://doi.org/10.1038/s41467-020-16113-y ARTICLE Fig. 1 Characterization of cell types in the P1 cochlea. a Line drawing of a cross section of the floor of the cochlear duct at P1. Distinct cell types within the organ of Corti (OC) are color coded. b Heat map for ~14,000 cochlear cells collected from four separate experiments at P1. Top 25 differentially expressed (DE) genes for the 15 identified clusters are shown. Cellular identity for each cluster is indicated by a color bar at the top of the heat map, which corresponds to the legend in a, and by a cell name at the bottom. c tSNE plot for the same cells as in b. Cluster identities are indicated. d Violin plots showing normalized log-transformed expression values for the top five DE genes for each cell type (color coded as in c) by comparison with all other P1 cells (gray on the right in each graph). Bars indicate median expression level. e Upper left panel, tSNE plot of cells determined to be derived from KO (between the OC and medial edge of the cochlear duct). Lower left panel, feature plot for the same cells as in the upper panel indicating high expression of Otoa, Calb1, and Fabp7 (based on color) in different clusters of cells. Lower right panel, cross sections through the cochlear duct at P1, illustrating expression of CALB1 in the medial region of KO and FABP7 directly adjacent to the OC (arrow; scale bars, 20 μm). Lowest panel shows high-magnification view of expression of FABP7 (arrow, gray scale) at the lateral KO border (green line; scale bar, 10 μm). Upper right panel, summary diagram of the spatial distribution of KO cell clusters at P1. HC hair cells, IPhC inner phalangeal cells/border cells, IPC inner pillar cells, OPC outer pillar cells, DC1/2 Deiters’ cells rows 1 and 2, DC3, Deiters’ cells row 3, HeC Hensen’s cells, CC/OSC Claudius cells/outer sulcus cells, IdC interdental cells, ISC inner sulcus cells, KO Kölliker’s organ cells, L.KO lateral Kölliker’s organ cells, M.KO medial Kölliker’s organ cells, OC90 OC90+ cells. unique cell types at each time point, including both known types, during cochlear development. In particular, cells within KO play such as HCs and SCs, and previously unknown cell types, such as a role in the development of the tectorial membrane11, the gen- multiple unique cell types in Kölliker’s organ (KO). Cells col- eration of spontaneous activity required for maturation of spiral lected from E14 and E16 cochleae include prosensory cells; ganglion neurons12 and some cells within this region retain however, unbiased clustering indicates two distinct populations. prosensory potential13–15. However, since KO cells are morpho- Fate mapping of one of these populations demonstrates a strong logically homogenous, the extent of transcriptional heterogeneity bias toward lateral fates (OHCs and surrounding support cells), was unclear.
Recommended publications
  • Downloaded from the National Database for Autism Research (NDAR)
    International Journal of Molecular Sciences Article Phenotypic Subtyping and Re-Analysis of Existing Methylation Data from Autistic Probands in Simplex Families Reveal ASD Subtype-Associated Differentially Methylated Genes and Biological Functions Elizabeth C. Lee y and Valerie W. Hu * Department of Biochemistry and Molecular Medicine, The George Washington University, School of Medicine and Health Sciences, Washington, DC 20037, USA; [email protected] * Correspondence: [email protected]; Tel.: +1-202-994-8431 Current address: W. Harry Feinstone Department of Molecular Microbiology and Immunology, y Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA. Received: 25 August 2020; Accepted: 17 September 2020; Published: 19 September 2020 Abstract: Autism spectrum disorder (ASD) describes a group of neurodevelopmental disorders with core deficits in social communication and manifestation of restricted, repetitive, and stereotyped behaviors. Despite the core symptomatology, ASD is extremely heterogeneous with respect to the severity of symptoms and behaviors. This heterogeneity presents an inherent challenge to all large-scale genome-wide omics analyses. In the present study, we address this heterogeneity by stratifying ASD probands from simplex families according to the severity of behavioral scores on the Autism Diagnostic Interview-Revised diagnostic instrument, followed by re-analysis of existing DNA methylation data from individuals in three ASD subphenotypes in comparison to that of their respective unaffected siblings. We demonstrate that subphenotyping of cases enables the identification of over 1.6 times the number of statistically significant differentially methylated regions (DMR) and DMR-associated genes (DAGs) between cases and controls, compared to that identified when all cases are combined. Our analyses also reveal ASD-related neurological functions and comorbidities that are enriched among DAGs in each phenotypic subgroup but not in the combined case group.
    [Show full text]
  • Expression Gene Network Analyses Reveal Molecular Mechanisms And
    www.nature.com/scientificreports OPEN Diferential expression and co- expression gene network analyses reveal molecular mechanisms and candidate biomarkers involved in breast muscle myopathies in chicken Eva Pampouille1,2, Christelle Hennequet-Antier1, Christophe Praud1, Amélie Juanchich1, Aurélien Brionne1, Estelle Godet1, Thierry Bordeau1, Fréderic Fagnoul2, Elisabeth Le Bihan-Duval1 & Cécile Berri1* The broiler industry is facing an increasing prevalence of breast myopathies, such as white striping (WS) and wooden breast (WB), and the precise aetiology of these occurrences remains poorly understood. To progress our understanding of the structural changes and molecular pathways involved in these myopathies, a transcriptomic analysis was performed using an 8 × 60 K Agilent chicken microarray and histological study. The study used pectoralis major muscles from three groups: slow-growing animals (n = 8), fast-growing animals visually free from defects (n = 8), or severely afected by both WS and WB (n = 8). In addition, a weighted correlation network analysis was performed to investigate the relationship between modules of co-expressed genes and histological traits. Functional analysis suggested that selection for fast growing and breast meat yield has progressively led to conditions favouring metabolic shifts towards alternative catabolic pathways to produce energy, leading to an adaptive response to oxidative stress and the frst signs of infammatory, regeneration and fbrosis processes. All these processes are intensifed in muscles afected by severe myopathies, in which new mechanisms related to cellular defences and remodelling seem also activated. Furthermore, our study opens new perspectives for myopathy diagnosis by highlighting fne histological phenotypes and genes whose expression was strongly correlated with defects. Te poultry industry relies on the production of fast-growing chickens, which are slaughtered at high weights and intended for cutting and processing.
    [Show full text]
  • Seq2pathway Vignette
    seq2pathway Vignette Bin Wang, Xinan Holly Yang, Arjun Kinstlick May 19, 2021 Contents 1 Abstract 1 2 Package Installation 2 3 runseq2pathway 2 4 Two main functions 3 4.1 seq2gene . .3 4.1.1 seq2gene flowchart . .3 4.1.2 runseq2gene inputs/parameters . .5 4.1.3 runseq2gene outputs . .8 4.2 gene2pathway . 10 4.2.1 gene2pathway flowchart . 11 4.2.2 gene2pathway test inputs/parameters . 11 4.2.3 gene2pathway test outputs . 12 5 Examples 13 5.1 ChIP-seq data analysis . 13 5.1.1 Map ChIP-seq enriched peaks to genes using runseq2gene .................... 13 5.1.2 Discover enriched GO terms using gene2pathway_test with gene scores . 15 5.1.3 Discover enriched GO terms using Fisher's Exact test without gene scores . 17 5.1.4 Add description for genes . 20 5.2 RNA-seq data analysis . 20 6 R environment session 23 1 Abstract Seq2pathway is a novel computational tool to analyze functional gene-sets (including signaling pathways) using variable next-generation sequencing data[1]. Integral to this tool are the \seq2gene" and \gene2pathway" components in series that infer a quantitative pathway-level profile for each sample. The seq2gene function assigns phenotype-associated significance of genomic regions to gene-level scores, where the significance could be p-values of SNPs or point mutations, protein-binding affinity, or transcriptional expression level. The seq2gene function has the feasibility to assign non-exon regions to a range of neighboring genes besides the nearest one, thus facilitating the study of functional non-coding elements[2]. Then the gene2pathway summarizes gene-level measurements to pathway-level scores, comparing the quantity of significance for gene members within a pathway with those outside a pathway.
    [Show full text]
  • Product Data Sheet
    Product Data Sheet ExProfileTM Human AMPK Signaling Related Gene qPCR Array For focused group profiling of human AMPK signaling genes expression Cat. No. QG004-A (4 x 96-well plate, Format A) Cat. No. QG004-B (4 x 96-well plate, Format B) Cat. No. QG004-C (4 x 96-well plate, Format C) Cat. No. QG004-D (4 x 96-well plate, Format D) Cat. No. QG004-E (4 x 96-well plate, Format E) Plates available individually or as a set of 6. Each set contains 336 unique gene primer pairs deposited in one 96-well plate. Introduction The ExProfile human AMPK signaling related gene qPCR array profiles the expression of 336 human genes related to AMPK-mediated signal transduction. These genes are carefully chosen for their close pathway correlation based on a thorough literature search of peer-reviewed publications, mainly including genes that encode AMP-activated protein kinase complex,its regulators and targets involved in many important biological processes, such as glucose uptake, β-oxidation of fatty acids and modulation of insulin secretion. This array allows researchers to study the pathway-related genes to gain understanding of their roles in the different biological processes. QG004 plate 01: 84 unique gene PCR primer pairs QG004 plate 02: 84 unique gene PCR primer pairs QG004 plate 03: 84 unique gene PCR primer pairs QG004 plate 04: 84 unique gene PCR primer pairs Shipping and storage condition Shipped at room temperate Stable for at least 6 months when stored at -20°C Array format GeneCopoeia provides five qPCR array formats (A, B, C, D, and E) suitable for use with the following real- time cyclers.
    [Show full text]
  • 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]
  • Supplementary Materials
    1 Supplementary Materials: Supplemental Figure 1. Gene expression profiles of kidneys in the Fcgr2b-/- and Fcgr2b-/-. Stinggt/gt mice. (A) A heat map of microarray data show the genes that significantly changed up to 2 fold compared between Fcgr2b-/- and Fcgr2b-/-. Stinggt/gt mice (N=4 mice per group; p<0.05). Data show in log2 (sample/wild-type). 2 Supplemental Figure 2. Sting signaling is essential for immuno-phenotypes of the Fcgr2b-/-lupus mice. (A-C) Flow cytometry analysis of splenocytes isolated from wild-type, Fcgr2b-/- and Fcgr2b-/-. Stinggt/gt mice at the age of 6-7 months (N= 13-14 per group). Data shown in the percentage of (A) CD4+ ICOS+ cells, (B) B220+ I-Ab+ cells and (C) CD138+ cells. Data show as mean ± SEM (*p < 0.05, **p<0.01 and ***p<0.001). 3 Supplemental Figure 3. Phenotypes of Sting activated dendritic cells. (A) Representative of western blot analysis from immunoprecipitation with Sting of Fcgr2b-/- mice (N= 4). The band was shown in STING protein of activated BMDC with DMXAA at 0, 3 and 6 hr. and phosphorylation of STING at Ser357. (B) Mass spectra of phosphorylation of STING at Ser357 of activated BMDC from Fcgr2b-/- mice after stimulated with DMXAA for 3 hour and followed by immunoprecipitation with STING. (C) Sting-activated BMDC were co-cultured with LYN inhibitor PP2 and analyzed by flow cytometry, which showed the mean fluorescence intensity (MFI) of IAb expressing DC (N = 3 mice per group). 4 Supplemental Table 1. Lists of up and down of regulated proteins Accession No.
    [Show full text]
  • Disrupted Iron Homeostasis Causes Dopaminergic Neurodegeneration in Mice
    Disrupted iron homeostasis causes dopaminergic neurodegeneration in mice Pavle Mataka, Andrija Mataka, Sarah Moustafaa, Dipendra K. Aryalb,c,d,e, Eric J. Bennerf, William Wetselb,c,d,e, and Nancy C. Andrewsa,f,1 aDepartment of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27705; bDepartment of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC 27705; cDepartment of Cell Biology, Duke University School of Medicine, Durham, NC 27705; dDepartment of Neurobiology, Duke University School of Medicine, Durham, NC 27705; eMouse Behavioral and Neuroendocrine Analysis Core Facility, Duke University School of Medicine, Durham, NC 27705; and fDepartment of Pediatrics, Duke University School of Medicine, Durham, NC 27705 This contribution is part of the special series of Inaugural Articles by members of the National Academy of Sciences elected in 2015. Contributed by Nancy C. Andrews, January 27, 2016 (sent for review September 30, 2015; reviewed by Michael K. Georgieff and Barry H. Paw) Disrupted brain iron homeostasis is a common feature of neuro- offers a very sensitive approach to detect cellular iron deficiency degenerative disease. To begin to understand how neuronal iron and surfeit (12). handling might be involved, we focused on dopaminergic neurons Iron homeostasis is altered locally, in the affected part of the and asked how inactivation of transport proteins affected iron brain, in most human neurodegenerative disorders (13). Iron ac- homeostasis in vivo in mice. Loss of the cellular iron exporter, cumulates in the substantia nigra (SN) in Parkinson’s disease (PD) ferroportin, had no apparent consequences. However, loss of (14–16), although it is unsettled whether increased iron is in neu- transferrin receptor 1, involved in iron uptake, caused neuronal ropil (17) or dopaminergic (DA) neurons (18), or both.
    [Show full text]
  • Ubiquitylome Profiling of Parkin-Null Brain Reveals Dysregulation Of
    Neurobiology of Disease 127 (2019) 114–130 Contents lists available at ScienceDirect Neurobiology of Disease journal homepage: www.elsevier.com/locate/ynbdi Ubiquitylome profiling of Parkin-null brain reveals dysregulation of calcium T homeostasis factors ATP1A2, Hippocalcin and GNA11, reflected by altered firing of noradrenergic neurons Key J.a,1, Mueller A.K.b,1, Gispert S.a, Matschke L.b, Wittig I.c, Corti O.d,e,f,g, Münch C.h, ⁎ ⁎ Decher N.b, , Auburger G.a, a Exp. Neurology, Goethe University Medical School, 60590 Frankfurt am Main, Germany b Institute for Physiology and Pathophysiology, Vegetative Physiology and Marburg Center for Mind, Brain and Behavior - MCMBB; Clinic for Neurology, Philipps-University Marburg, 35037 Marburg, Germany c Functional Proteomics, SFB 815 Core Unit, Goethe University Medical School, 60590 Frankfurt am Main, Germany d Institut du Cerveau et de la Moelle épinière, ICM, Paris, F-75013, France e Inserm, U1127, Paris, F-75013, France f CNRS, UMR 7225, Paris, F-75013, France g Sorbonne Universités, Paris, F-75013, France h Institute of Biochemistry II, Goethe University Medical School, 60590 Frankfurt am Main, Germany ARTICLE INFO ABSTRACT Keywords: Parkinson's disease (PD) is the second most frequent neurodegenerative disorder in the old population. Among Parkinson's disease its monogenic variants, a frequent cause is a mutation in the Parkin gene (Prkn). Deficient function of Parkin Mitochondria triggers ubiquitous mitochondrial dysfunction and inflammation in the brain, but it remains unclear howse- Parkin lective neural circuits become vulnerable and finally undergo atrophy. Ubiquitin We attempted to go beyond previous work, mostly done in peripheral tumor cells, which identified protein Calcium targets of Parkin activity, an ubiquitin E3 ligase.
    [Show full text]
  • Proteomics Provides Insights Into the Inhibition of Chinese Hamster V79
    www.nature.com/scientificreports OPEN Proteomics provides insights into the inhibition of Chinese hamster V79 cell proliferation in the deep underground environment Jifeng Liu1,2, Tengfei Ma1,2, Mingzhong Gao3, Yilin Liu4, Jun Liu1, Shichao Wang2, Yike Xie2, Ling Wang2, Juan Cheng2, Shixi Liu1*, Jian Zou1,2*, Jiang Wu2, Weimin Li2 & Heping Xie2,3,5 As resources in the shallow depths of the earth exhausted, people will spend extended periods of time in the deep underground space. However, little is known about the deep underground environment afecting the health of organisms. Hence, we established both deep underground laboratory (DUGL) and above ground laboratory (AGL) to investigate the efect of environmental factors on organisms. Six environmental parameters were monitored in the DUGL and AGL. Growth curves were recorded and tandem mass tag (TMT) proteomics analysis were performed to explore the proliferative ability and diferentially abundant proteins (DAPs) in V79 cells (a cell line widely used in biological study in DUGLs) cultured in the DUGL and AGL. Parallel Reaction Monitoring was conducted to verify the TMT results. γ ray dose rate showed the most detectable diference between the two laboratories, whereby γ ray dose rate was signifcantly lower in the DUGL compared to the AGL. V79 cell proliferation was slower in the DUGL. Quantitative proteomics detected 980 DAPs (absolute fold change ≥ 1.2, p < 0.05) between V79 cells cultured in the DUGL and AGL. Of these, 576 proteins were up-regulated and 404 proteins were down-regulated in V79 cells cultured in the DUGL. KEGG pathway analysis revealed that seven pathways (e.g.
    [Show full text]
  • Warning Sines: Alu Elements, Evolution of the Human Brain, and the Spectrum of Neurological Disease
    Chromosome Res (2018) 26:93–111 https://doi.org/10.1007/s10577-018-9573-4 ORIGINAL ARTICLE Warning SINEs: Alu elements, evolution of the human brain, and the spectrum of neurological disease Peter A. Larsen & Kelsie E. Hunnicutt & Roxanne J. Larsen & Anne D. Yoder & Ann M. Saunders Received: 6 December 2017 /Revised: 14 January 2018 /Accepted: 15 January 2018 /Published online: 19 February 2018 # The Author(s) 2018. This article is an open access publication Abstract Alu elements are a highly successful family of neurological networks are potentially vulnerable to the primate-specific retrotransposons that have fundamen- epigenetic dysregulation of Alu elements operating tally shaped primate evolution, including the evolution across the suite of nuclear-encoded mitochondrial genes of our own species. Alus play critical roles in the forma- that are critical for both mitochondrial and CNS func- tion of neurological networks and the epigenetic regu- tion. Here, we highlight the beneficial neurological as- lation of biochemical processes throughout the central pects of Alu elements as well as their potential to cause nervous system (CNS), and thus are hypothesized to disease by disrupting key cellular processes across the have contributed to the origin of human cognition. De- CNS. We identify at least 37 neurological and neurode- spite the benefits that Alus provide, deleterious Alu generative disorders wherein deleterious Alu activity has activity is associated with a number of neurological been implicated as a contributing factor for the manifes- and neurodegenerative disorders. In particular, tation of disease, and for many of these disorders, this activity is operating on genes that are essential for proper mitochondrial function.
    [Show full text]
  • Supplementary Table S4. FGA Co-Expressed Gene List in LUAD
    Supplementary Table S4. FGA co-expressed gene list in LUAD tumors Symbol R Locus Description FGG 0.919 4q28 fibrinogen gamma chain FGL1 0.635 8p22 fibrinogen-like 1 SLC7A2 0.536 8p22 solute carrier family 7 (cationic amino acid transporter, y+ system), member 2 DUSP4 0.521 8p12-p11 dual specificity phosphatase 4 HAL 0.51 12q22-q24.1histidine ammonia-lyase PDE4D 0.499 5q12 phosphodiesterase 4D, cAMP-specific FURIN 0.497 15q26.1 furin (paired basic amino acid cleaving enzyme) CPS1 0.49 2q35 carbamoyl-phosphate synthase 1, mitochondrial TESC 0.478 12q24.22 tescalcin INHA 0.465 2q35 inhibin, alpha S100P 0.461 4p16 S100 calcium binding protein P VPS37A 0.447 8p22 vacuolar protein sorting 37 homolog A (S. cerevisiae) SLC16A14 0.447 2q36.3 solute carrier family 16, member 14 PPARGC1A 0.443 4p15.1 peroxisome proliferator-activated receptor gamma, coactivator 1 alpha SIK1 0.435 21q22.3 salt-inducible kinase 1 IRS2 0.434 13q34 insulin receptor substrate 2 RND1 0.433 12q12 Rho family GTPase 1 HGD 0.433 3q13.33 homogentisate 1,2-dioxygenase PTP4A1 0.432 6q12 protein tyrosine phosphatase type IVA, member 1 C8orf4 0.428 8p11.2 chromosome 8 open reading frame 4 DDC 0.427 7p12.2 dopa decarboxylase (aromatic L-amino acid decarboxylase) TACC2 0.427 10q26 transforming, acidic coiled-coil containing protein 2 MUC13 0.422 3q21.2 mucin 13, cell surface associated C5 0.412 9q33-q34 complement component 5 NR4A2 0.412 2q22-q23 nuclear receptor subfamily 4, group A, member 2 EYS 0.411 6q12 eyes shut homolog (Drosophila) GPX2 0.406 14q24.1 glutathione peroxidase
    [Show full text]
  • Transcriptomic Profiling of Ca Transport Systems During
    cells Article Transcriptomic Profiling of Ca2+ Transport Systems during the Formation of the Cerebral Cortex in Mice Alexandre Bouron Genetics and Chemogenomics Lab, Université Grenoble Alpes, CNRS, CEA, INSERM, Bâtiment C3, 17 rue des Martyrs, 38054 Grenoble, France; [email protected] Received: 29 June 2020; Accepted: 24 July 2020; Published: 29 July 2020 Abstract: Cytosolic calcium (Ca2+) transients control key neural processes, including neurogenesis, migration, the polarization and growth of neurons, and the establishment and maintenance of synaptic connections. They are thus involved in the development and formation of the neural system. In this study, a publicly available whole transcriptome sequencing (RNA-Seq) dataset was used to examine the expression of genes coding for putative plasma membrane and organellar Ca2+-transporting proteins (channels, pumps, exchangers, and transporters) during the formation of the cerebral cortex in mice. Four ages were considered: embryonic days 11 (E11), 13 (E13), and 17 (E17), and post-natal day 1 (PN1). This transcriptomic profiling was also combined with live-cell Ca2+ imaging recordings to assess the presence of functional Ca2+ transport systems in E13 neurons. The most important Ca2+ routes of the cortical wall at the onset of corticogenesis (E11–E13) were TACAN, GluK5, nAChR β2, Cav3.1, Orai3, transient receptor potential cation channel subfamily M member 7 (TRPM7) non-mitochondrial Na+/Ca2+ exchanger 2 (NCX2), and the connexins CX43/CX45/CX37. Hence, transient receptor potential cation channel mucolipin subfamily member 1 (TRPML1), transmembrane protein 165 (TMEM165), and Ca2+ “leak” channels are prominent intracellular Ca2+ pathways. The Ca2+ pumps sarco/endoplasmic reticulum Ca2+ ATPase 2 (SERCA2) and plasma membrane Ca2+ ATPase 1 (PMCA1) control the resting basal Ca2+ levels.
    [Show full text]