A Chromosome Level Genome of Astyanax Mexicanus Surface Fish for Comparing Population

Total Page:16

File Type:pdf, Size:1020Kb

A Chromosome Level Genome of Astyanax Mexicanus Surface Fish for Comparing Population bioRxiv preprint doi: https://doi.org/10.1101/2020.07.06.189654; this version posted July 6, 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 The copyright holder for this preprint (which was not certified by peer review) is the author/funder.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. 1 Title 2 A chromosome level genome of Astyanax mexicanus surface fish for comparing population- 3 specific genetic differences contributing to trait evolution. 4 5 Authors 6 Wesley C. Warren1, Tyler E. Boggs2, Richard Borowsky3, Brian M. Carlson4, Estephany 7 Ferrufino5, Joshua B. Gross2, LaDeana Hillier6, Zhilian Hu7, Alex C. Keene8, Alexander Kenzior9, 8 Johanna E. Kowalko5, Chad Tomlinson10, Milinn Kremitzki10, Madeleine E. Lemieux11, Tina 9 Graves-Lindsay10, Suzanne E. McGaugh12, Jeff T. Miller12, Mathilda Mommersteeg7, Rachel L. 10 Moran12, Robert Peuß9, Edward Rice1, Misty R. Riddle13, Itzel Sifuentes-Romero5, Bethany A. 11 Stanhope5,8, Clifford J. Tabin13, Sunishka Thakur5, Yamamoto Yoshiyuki14, Nicolas Rohner9,15 12 13 Authors for correspondence: Wesley C. Warren ([email protected]), Nicolas Rohner 14 ([email protected]) 15 16 Affiliation 17 1Department of Animal Sciences, Department of Surgery, Institute for Data Science and 18 Informatics, University of Missouri, Bond Life Sciences Center, Columbia, MO 19 2 Department of Biological Sciences, University of Cincinnati, Cincinnati, OH 20 3 Department of Biology, New York University, New York, NY 21 4 Department of Biology, The College of Wooster, Wooster, OH 22 5 Harriet L. Wilkes Honors College, Florida Atlantic University, Jupiter FL 23 6 Department of Genome Sciences, University of Washington, Seattle, WA 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.07.06.189654; this version posted July 6, 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 The copyright holder for this preprint (which was not certified by peer review) is the author/funder.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. 24 25 7 Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United 26 Kingdom 27 8 Department of Biological Sciences, Florida Atlantic University, Jupiter FL 28 9 Stowers Institute for Medical Research, Kansas City, MO 29 10 McDonnell Genome Institute, Washington University, St Louis, MO 30 11 Bioinfo, Plantagenet, ON K0B 1L0, Canada 31 12 Department of Ecology, Evolution, and Behavior, University of Minnesota, Saint Paul, MN 32 13 Genetics Department, Blavatnik Institute, Harvard Medical School, Boston, MA 33 14 Department of Cell and Developmental Biology, University College London, London, United 34 Kingdom 35 15 Department of Molecular & Integrative Physiology, KU Medical Center, Kansas City, KS 36 37 Abstract 38 Identifying the genetic factors that underlie complex traits is central to understanding the 39 mechanistic underpinnings of evolution. In nature, adaptation to severe environmental change, 40 such as encountered following colonization of caves, has dramatically altered genomes of species 41 over varied time spans. Genomic sequencing approaches have identified mutations associated with 42 troglomorphic trait evolution, but the functional impacts of these mutations remain poorly 43 understood. The Mexican Tetra, Astyanax mexicanus, is abundant in the surface waters of 44 northeastern Mexico, and also inhabits at least 30 different caves in the region. Cave-dwelling A. 45 mexicanus morphs are well adapted to subterranean life and many populations appear to have 46 evolved troglomorphic traits independently, while the surface-dwelling populations can be used as 2 bioRxiv preprint doi: https://doi.org/10.1101/2020.07.06.189654; this version posted July 6, 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. 47 a proxy for the ancestral form. Here we present a high-resolution, chromosome-level surface fish 48 genome, enabling the first genome-wide comparison between surface fish and cavefish 49 populations. Using this resource, we performed quantitative trait locus (QTL) mapping analyses 50 for pigmentation and eye size and found new candidate genes for eye loss such as dusp26. We 51 used CRISPR gene editing in A. mexicanus to confirm the essential role of a gene within an eye 52 size QTL, rx3, in eye formation. We also generated the first genome-wide evaluation of deletion 53 variability that includes an analysis of the impact on protein-coding genes across cavefish 54 populations to gain insight into this potential source of cave adaptation. The new surface fish 55 genome reference now provides a more complete resource for comparative, functional, 56 developmental and genetic studies of drastic trait differences within a species. 57 58 Main Text 59 Establishing the relationship between natural environmental factors and the genetic basis of trait 60 evolution has been challenging. The ecological shift from surface to cave environments provides 61 a tractable system to address this, as in this instance the polarity of evolutionary change is known. 62 Across the globe, subterranean animals, including fish, salamanders, rodents, and myriads of 63 invertebrate species have converged on reductions in metabolic rate, eye size, and pigmentation1- 64 3. The robust phenotypic differences from surface relatives provide the opportunity to investigate 65 the mechanistic underpinnings of evolution and to determine whether genotype-phenotype 66 interactions are deeply conserved. 67 The Mexican cavefish, Astyanax mexicanus, has emerged as a powerful model to 68 investigate complex trait evolution4. A. mexicanus comprises at least 30 cave-dwelling populations 69 in the Sierra de El Abra, Sierra de Colmena and Sierra de Guatemala regions of northeastern 3 bioRxiv preprint doi: https://doi.org/10.1101/2020.07.06.189654; this version posted July 6, 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. 70 Mexico, and surface-dwelling fish of the same species inhabit rivers and lakes throughout Mexico 71 and southern Texas5. The ecology of surface and cave environments differ dramatically, allowing 72 for functional and genomic comparisons between populations that have evolved in distinct 73 environments. Dozens of evolved trait differences have been identified in cavefish including 74 changes in morphology, physiology, and behavior4,6 . It is also worth noting that comparing 75 cavefish to surface fish reveals substantial differences in many traits of possible relevance to 76 human disease, including sleep duration, circadian rhythmicity, anxiety, aggression, heart 77 regeneration, eye and retina development, craniofacial structure, insulin resistance, appetite, and 78 obesity7-17. Further, generation of fertile surface-cave hybrids in a laboratory setting has allowed 79 for genetic mapping in these fish10,18-25. Clear phenotypic differences, combined with availability 80 of genetic tools, positions A. mexicanus as a natural model system for identifying the genetic basis 81 of ecologically and evolutionary relevant phenotypes17,26,27. 82 Recently, the genome of an individual from the Pachón cave population was sequenced28. 83 While this work uncovered genomic intervals and candidate genes linked to cave traits, an 84 important computational limitation was sequence fragmentation due to the use of short-read 85 sequencing. In addition, lack of a surface fish genome prevents direct comparisons between surface 86 and cavefish populations at the genomic level. Here, we address these two obstacles by presenting 87 the first de novo genome assembly of the surface fish morph using long-read sequencing 88 technology. This approach yielded a much more comprehensive genome that allows for direct 89 genome-wide comparisons between surface fish and Pachón cavefish. As a proof-of-principle, we 90 first confirmed known genetic mutations associated with pigmentation and eye loss, then 91 discovered novel quantitative trait loci (QTL), and identified coding and deletion mutations that 92 highlight putative contributions to cave trait biology. 4 bioRxiv preprint doi: https://doi.org/10.1101/2020.07.06.189654; this version posted July 6, 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. 93 94 Results 95 De novo assembly and curation of the surface fish genome. We set out to generate a robust 96 reference genome for the surface morph of A. mexicanus from a single lab-reared female, 97 descended from wild caught individuals from known Mexico localities (Fig. 1a, Supplementary 98 Figure 1). We sequenced and assembled the genome using Pacific Biosciences single molecule 99 real time (SMRT) sequencing (~73x genome coverage) and the wtdbg2 assembler29 to an 100 ungapped size of 1.29 Gb. Initial scaffolding of assembled contigs was accomplished with the aid 101 of an A. mexicanus surface fish physical map (BioNano) followed by manual assignment of 70% 102 of the assembly scaffolds to 25 total chromosomes using the existing A. mexicanus genetic linkage 103 map markers30. The final genome assembly, Astyanax mexicanus 2.0, comprises a total of 2,415 104 scaffolds (including single contig scaffolds) with N50 contig and scaffold lengths of 1.7 Mb and 105 35 Mb, respectively, which is comparable to other similarly sequenced and assembled teleost 106 fishes (Supplementary Table 1). The assembled regions (394 Mb) that we were
Recommended publications
  • Genetic Variations in the PSMA6 and PSMC6 Proteasome Genes Are Associated with Multiple Sclerosis and Response to Interferon‑Β Therapy in Latvians
    EXPERIMENTAL AND THERAPEUTIC MEDICINE 21: 478, 2021 Genetic variations in the PSMA6 and PSMC6 proteasome genes are associated with multiple sclerosis and response to interferon‑β therapy in Latvians NATALIA PARAMONOVA1, JOLANTA KALNINA1, KRISTINE DOKANE1, KRISTINE DISLERE1, ILVA TRAPINA1, TATJANA SJAKSTE1 and NIKOLAJS SJAKSTE1,2 1Genomics and Bioinformatics, Institute of Biology of The University of Latvia; 2Department of Medical Biochemistry of The University of Latvia, LV‑1004 Riga, Latvia Received July 8, 2020; Accepted December 8, 2020 DOI: 10.3892/etm.2021.9909 Abstract. Several polymorphisms in genes related to the Introduction ubiquitin‑proteasome system exhibit an association with pathogenesis and prognosis of various human autoimmune Multiple sclerosis (MS) is a lifelong demyelinating disease of diseases. Our previous study reported the association the central nervous system. The clinical onset of MS tends to between multiple sclerosis (MS) and the PSMA3‑rs2348071 be between the second and fourth decade of life. Similarly to polymorphism in the Latvian population. The current study other autoimmune diseases, women are affected 3‑4 times more aimed to evaluate the PSMA6 and PSMC6 genetic variations, frequently than men (1). About 10% of MS patients experience their interaction between each other and with the rs2348071, a primary progressive MS form characterized by the progres‑ on the susceptibility to MS risk and response to therapy in sion of neurological disability from the onset. In about 90% the Latvian population. PSMA6‑rs2277460, ‑rs1048990 and of MS patients, the disease undergoes the relapse‑remitting PSMC6‑rs2295826, ‑rs2295827 were genotyped in the MS MS course (RRMS); in most of these patients, the condition case/control study and analysed in terms of genotype‑protein acquires secondary progressive course (SPMS) (2).
    [Show full text]
  • Transcriptome Analyses of Rhesus Monkey Pre-Implantation Embryos Reveal A
    Downloaded from genome.cshlp.org on September 23, 2021 - Published by Cold Spring Harbor Laboratory Press Transcriptome analyses of rhesus monkey pre-implantation embryos reveal a reduced capacity for DNA double strand break (DSB) repair in primate oocytes and early embryos Xinyi Wang 1,3,4,5*, Denghui Liu 2,4*, Dajian He 1,3,4,5, Shengbao Suo 2,4, Xian Xia 2,4, Xiechao He1,3,6, Jing-Dong J. Han2#, Ping Zheng1,3,6# Running title: reduced DNA DSB repair in monkey early embryos Affiliations: 1 State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China 2 Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China 3 Yunnan Key Laboratory of Animal Reproduction, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China 4 University of Chinese Academy of Sciences, Beijing, China 5 Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China 6 Primate Research Center, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China * Xinyi Wang and Denghui Liu contributed equally to this work 1 Downloaded from genome.cshlp.org on September 23, 2021 - Published by Cold Spring Harbor Laboratory Press # Correspondence: Jing-Dong J. Han, Email: [email protected]; Ping Zheng, Email: [email protected] Key words: rhesus monkey, pre-implantation embryo, DNA damage 2 Downloaded from genome.cshlp.org on September 23, 2021 - Published by Cold Spring Harbor Laboratory Press ABSTRACT Pre-implantation embryogenesis encompasses several critical events including genome reprogramming, zygotic genome activation (ZGA) and cell fate commitment.
    [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]
  • Mouse Eaf2 Conditional Knockout Project (CRISPR/Cas9)
    https://www.alphaknockout.com Mouse Eaf2 Conditional Knockout Project (CRISPR/Cas9) Objective: To create a Eaf2 conditional knockout Mouse model (C57BL/6J) by CRISPR/Cas-mediated genome engineering. Strategy summary: The Eaf2 gene (NCBI Reference Sequence: NM_001113401 ; Ensembl: ENSMUSG00000022838 ) is located on Mouse chromosome 16. 6 exons are identified, with the ATG start codon in exon 1 and the TGA stop codon in exon 6 (Transcript: ENSMUST00000114829). Exon 4 will be selected as conditional knockout region (cKO region). Deletion of this region should result in the loss of function of the Mouse Eaf2 gene. To engineer the targeting vector, homologous arms and cKO region will be generated by PCR using BAC clone RP24-400K10 as template. Cas9, gRNA and targeting vector will be co-injected into fertilized eggs for cKO Mouse production. The pups will be genotyped by PCR followed by sequencing analysis. Note: Mice homozygous for a null allele exhibit premature death, enlarged heart and prostate associate with hypertrophy, and increased incidence of tumors. Exon 4 starts from about 43.13% of the coding region. The knockout of Exon 4 will result in frameshift of the gene. The size of intron 3 for 5'-loxP site insertion: 2311 bp, and the size of intron 4 for 3'-loxP site insertion: 7170 bp. The size of effective cKO region: ~646 bp. The cKO region does not have any other known gene. Page 1 of 8 https://www.alphaknockout.com Overview of the Targeting Strategy Wildtype allele gRNA region 5' gRNA region 3' 1 4 6 Targeting vector Targeted allele Constitutive KO allele (After Cre recombination) Legends Exon of mouse Eaf2 Homology arm cKO region loxP site Page 2 of 8 https://www.alphaknockout.com Overview of the Dot Plot Window size: 10 bp Forward Reverse Complement Sequence 12 Note: The sequence of homologous arms and cKO region is aligned with itself to determine if there are tandem repeats.
    [Show full text]
  • A Species-Specific Retrotransposon Drives a Conserved Cdk2ap1 Isoform Essential for Preimplantation Development
    bioRxiv preprint doi: https://doi.org/10.1101/2021.03.24.436683; this version posted March 25, 2021. 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. Title: A species-specific retrotransposon drives a conserved Cdk2ap1 isoform essential for preimplantation development Authors: Andrew Modzelewski1†, Wanqing Shao2†, Jingqi Chen1, Angus Lee1, Xin Qi1, Mackenzie Noon1, Kristy Tjokro1, Gabriele Sales3, Anne Biton4, Terence Speed5, Zhenyu Xuan6, Ting Wang2#, Davide Risso3# and Lin He1# Affiliations: 1 Division of Cellular and Developmental Biology, MCB department, University of California at Berkeley, Berkeley, CA, USA. 2 Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA 3 Department of Statistical Sciences, University of Padova, Italy. 4 Division of Biostatistics, University of California at Berkeley, Berkeley, CA 94720, USA. 5 Bioinformatics Division, WEHI, Parkville, VIC 3052, Australia. 6 Department of Biological Sciences, The University of Texas at Dallas, Richardson Texas, USA # Correspondence to: [email protected], [email protected] and [email protected] †These authors contributed equally. Abstract: Retrotransposons mediate gene regulation in multiple developmental and pathological processes. Here, we characterized the transient retrotransposon induction in preimplantation development of eight mammalian species. While species-specific in sequences, induced retrotransposons exhibit a similar preimplantation profile, conferring gene regulatory activities particularly through LTR retrotransposon promoters. We investigated a mouse-specific MT2B2 retrotransposon promoter, which generates an N-terminally truncated, preimplantation-specific Cdk2ap1ΔN isoform to promote cell proliferation. Cdk2ap1ΔN functionally contrasts to the canonical Cdk2ap1, which represses cell proliferation and peaks in mid-gestation stage.
    [Show full text]
  • Supplemental Table 1. Complete Gene Lists and GO Terms from Figure 3C
    Supplemental Table 1. Complete gene lists and GO terms from Figure 3C. Path 1 Genes: RP11-34P13.15, RP4-758J18.10, VWA1, CHD5, AZIN2, FOXO6, RP11-403I13.8, ARHGAP30, RGS4, LRRN2, RASSF5, SERTAD4, GJC2, RHOU, REEP1, FOXI3, SH3RF3, COL4A4, ZDHHC23, FGFR3, PPP2R2C, CTD-2031P19.4, RNF182, GRM4, PRR15, DGKI, CHMP4C, CALB1, SPAG1, KLF4, ENG, RET, GDF10, ADAMTS14, SPOCK2, MBL1P, ADAM8, LRP4-AS1, CARNS1, DGAT2, CRYAB, AP000783.1, OPCML, PLEKHG6, GDF3, EMP1, RASSF9, FAM101A, STON2, GREM1, ACTC1, CORO2B, FURIN, WFIKKN1, BAIAP3, TMC5, HS3ST4, ZFHX3, NLRP1, RASD1, CACNG4, EMILIN2, L3MBTL4, KLHL14, HMSD, RP11-849I19.1, SALL3, GADD45B, KANK3, CTC- 526N19.1, ZNF888, MMP9, BMP7, PIK3IP1, MCHR1, SYTL5, CAMK2N1, PINK1, ID3, PTPRU, MANEAL, MCOLN3, LRRC8C, NTNG1, KCNC4, RP11, 430C7.5, C1orf95, ID2-AS1, ID2, GDF7, KCNG3, RGPD8, PSD4, CCDC74B, BMPR2, KAT2B, LINC00693, ZNF654, FILIP1L, SH3TC1, CPEB2, NPFFR2, TRPC3, RP11-752L20.3, FAM198B, TLL1, CDH9, PDZD2, CHSY3, GALNT10, FOXQ1, ATXN1, ID4, COL11A2, CNR1, GTF2IP4, FZD1, PAX5, RP11-35N6.1, UNC5B, NKX1-2, FAM196A, EBF3, PRRG4, LRP4, SYT7, PLBD1, GRASP, ALX1, HIP1R, LPAR6, SLITRK6, C16orf89, RP11-491F9.1, MMP2, B3GNT9, NXPH3, TNRC6C-AS1, LDLRAD4, NOL4, SMAD7, HCN2, PDE4A, KANK2, SAMD1, EXOC3L2, IL11, EMILIN3, KCNB1, DOK5, EEF1A2, A4GALT, ADGRG2, ELF4, ABCD1 Term Count % PValue Genes regulation of pathway-restricted GDF3, SMAD7, GDF7, BMPR2, GDF10, GREM1, BMP7, LDLRAD4, SMAD protein phosphorylation 9 6.34 1.31E-08 ENG pathway-restricted SMAD protein GDF3, SMAD7, GDF7, BMPR2, GDF10, GREM1, BMP7, LDLRAD4, phosphorylation
    [Show full text]
  • A Single-Cell Transcriptomic Landscape of Primate Arterial Aging
    ARTICLE https://doi.org/10.1038/s41467-020-15997-0 OPEN A single-cell transcriptomic landscape of primate arterial aging Weiqi Zhang 1,2,3,4,5,13, Shu Zhang6,7,13, Pengze Yan3,8,13, Jie Ren7,9,13, Moshi Song3,5,8, Jingyi Li2,3,8, Jinghui Lei4, Huize Pan2,3, Si Wang3,5,8, Xibo Ma3,10, Shuai Ma2,3,8, Hongyu Li2,3, Fei Sun2,3, Haifeng Wan3,5,11, ✉ ✉ ✉ Wei Li 3,5,11, Piu Chan4, Qi Zhou3,5,11, Guang-Hui Liu 2,3,4,5,8 , Fuchou Tang 6,7,9,12 & Jing Qu 3,5,11 Our understanding of how aging affects the cellular and molecular components of the vas- 1234567890():,; culature and contributes to cardiovascular diseases is still limited. Here we report a single-cell transcriptomic survey of aortas and coronary arteries in young and old cynomolgus monkeys. Our data define the molecular signatures of specialized arteries and identify eight markers discriminating aortic and coronary vasculatures. Gene network analyses characterize tran- scriptional landmarks that regulate vascular senility and position FOXO3A, a longevity- associated transcription factor, as a master regulator gene that is downregulated in six subtypes of monkey vascular cells during aging. Targeted inactivation of FOXO3A in human vascular endothelial cells recapitulates the major phenotypic defects observed in aged monkey arteries, verifying FOXO3A loss as a key driver for arterial endothelial aging. Our study provides a critical resource for understanding the principles underlying primate arterial aging and contributes important clues to future treatment of age-associated vascular disorders. 1 CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China.
    [Show full text]
  • Reconstructing Cell Cycle Pseudo Time-Series Via Single-Cell Transcriptome Data—Supplement
    School of Natural Sciences and Mathematics Reconstructing Cell Cycle Pseudo Time-Series Via Single-Cell Transcriptome Data—Supplement UT Dallas Author(s): Michael Q. Zhang Rights: CC BY 4.0 (Attribution) ©2017 The Authors Citation: Liu, Zehua, Huazhe Lou, Kaikun Xie, Hao Wang, et al. 2017. "Reconstructing cell cycle pseudo time-series via single-cell transcriptome data." Nature Communications 8, doi:10.1038/s41467-017-00039-z This document is being made freely available by the Eugene McDermott Library of the University of Texas at Dallas with permission of the copyright owner. All rights are reserved under United States copyright law unless specified otherwise. File name: Supplementary Information Description: Supplementary figures, supplementary tables, supplementary notes, supplementary methods and supplementary references. CCNE1 CCNE1 CCNE1 CCNE1 36 40 32 34 32 35 30 32 28 30 30 28 28 26 24 25 Normalized Expression Normalized Expression Normalized Expression Normalized Expression 26 G1 S G2/M G1 S G2/M G1 S G2/M G1 S G2/M Cell Cycle Stage Cell Cycle Stage Cell Cycle Stage Cell Cycle Stage CCNE1 CCNE1 CCNE1 CCNE1 40 32 40 40 35 30 38 30 30 28 36 25 26 20 20 34 Normalized Expression Normalized Expression Normalized Expression 24 Normalized Expression G1 S G2/M G1 S G2/M G1 S G2/M G1 S G2/M Cell Cycle Stage Cell Cycle Stage Cell Cycle Stage Cell Cycle Stage Supplementary Figure 1 | High stochasticity of single-cell gene expression means, as demonstrated by relative expression levels of gene Ccne1 using the mESC-SMARTer data. For every panel, 20 sample cells were randomly selected for each of the three stages, followed by plotting the mean expression levels at each stage.
    [Show full text]
  • Screening and Analysis of Pathogenic Genes During DMBA- Induced Buccal Mucosa Carcinogenesis in Golden Hamsters
    1619-1624.qxd 23/4/2010 09:56 Ì ™ÂÏ›‰·1619 ONCOLOGY REPORTS 23: 1619-1624, 2010 Screening and analysis of pathogenic genes during DMBA- induced buccal mucosa carcinogenesis in golden hamsters KAI YANG1, GUODONG ZHANG1, JIE MEI1, DAN CHEN1 and MINGJUN WU2 1Department of Oral and Maxillofacial Surgery, the First Affiliated Hospital, Chongqing Medical University, Chongqing; 2Institute of Life Sciences of Chongqing Medical University, Chongqing 400016, P.R. China Received December 18, 2009; Accepted February 9, 2010 DOI: 10.3892/or_00000803 Abstract. We designed to screen pathogenic genes related to Introduction the occurrence and development of oral buccal mucosa cancer by whole genome microarray and analyze the mechanisms There are 274,000 new cases of oral cancer in the world every of carcinogenesis. The golden hamster model of buccal year (1). Squamous cell carcinoma accounts for 90% of mucosa cancer was established by induction with DMBA. oral cancer, and buccal carcinoma is one of the most common cRNAs labeled with Cy3 were synthesized and hybridized oral cancers (2). Although the treatment of oral cancers has with Agilent Whole Rat Genome Arrays containing 41,000 markedly improved in recent decades, the 5-year survival genes/ESTs. A Venn diagram analysis was performed to rate for buccal carcinoma and other oral cancers is only 55- screen the continuously abnormally expressed genes. Our 60% (2-4). Therefore, it is of great significance to explore results show 5,255 significantly differentially expressed the molecular mechanisms of the occurrence and develop- genes in golden hamster pouch mucosa during the ment of oral mucosa carcinomas and search for effective progression of normal buccal mucosa to squamous cell therapeutic targets.
    [Show full text]
  • Genome-Wide DNA Methylation and Long-Term Ambient Air Pollution
    Lee et al. Clinical Epigenetics (2019) 11:37 https://doi.org/10.1186/s13148-019-0635-z RESEARCH Open Access Genome-wide DNA methylation and long- term ambient air pollution exposure in Korean adults Mi Kyeong Lee1 , Cheng-Jian Xu2,3,4, Megan U. Carnes5, Cody E. Nichols1, James M. Ward1, The BIOS consortium, Sung Ok Kwon6, Sun-Young Kim7*†, Woo Jin Kim6*† and Stephanie J. London1*† Abstract Background: Ambient air pollution is associated with numerous adverse health outcomes, but the underlying mechanisms are not well understood; epigenetic effects including altered DNA methylation could play a role. To evaluate associations of long-term air pollution exposure with DNA methylation in blood, we conducted an epigenome- wide association study in a Korean chronic obstructive pulmonary disease cohort (N = 100 including 60 cases) using Illumina’s Infinium HumanMethylation450K Beadchip. Annual average concentrations of particulate matter ≤ 10 μmin diameter (PM10) and nitrogen dioxide (NO2) were estimated at participants’ residential addresses using exposure prediction models. We used robust linear regression to identify differentially methylated probes (DMPs) and two different approaches, DMRcate and comb-p, to identify differentially methylated regions (DMRs). Results: After multiple testing correction (false discovery rate < 0.05), there were 12 DMPs and 27 DMRs associated with PM10 and 45 DMPs and 57 DMRs related to NO2. DMP cg06992688 (OTUB2) and several DMRs were associated with both exposures. Eleven DMPs in relation to NO2 confirmed previous findings in Europeans; the remainder were novel. Methylation levels of 39 DMPs were associated with expression levels of nearby genes in a separate dataset of 3075 individuals.
    [Show full text]
  • Large Meta-Analysis of Genome-Wide Association Studies
    medRxiv preprint doi: https://doi.org/10.1101/2020.10.01.20200659; this version posted October 4, 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. It is made available under a CC-BY-NC-ND 4.0 International license . Large meta-analysis of genome-wide association studies expands knowledge of the genetic etiology of Alzheimer’s disease and highlights potential translational opportunities Céline Bellenguez1,*,#, Fahri Küçükali2,3,4*, Iris Jansen5,6*, Victor Andrade7,8*, Sonia Morenau- Grau9,10,*, Najaf Amin11,12, Benjamin Grenier-Boley1, Anne Boland13, Luca Kleineidam7,8, Peter Holmans14, Pablo Garcia9,10, Rafael Campos Martin7, Adam Naj15,16, Yang Qiong17, Joshua C. Bis18, Vincent Damotte1, Sven Van der Lee5,6,19, Marcos Costa1, Julien Chapuis1, Vilmentas Giedraitis20, María Jesús Bullido10,21, Adolfo López de Munáin10,22, Jordi Pérez- Tur10,23, Pascual Sánchez-Juan10,24, Raquel Sánchez-Valle25, Victoria Álvarez26, Pau Pastor27, Miguel Medina10,28, Jasper Van Dongen2,3,4, Christine Van Broeckhoven2,3,4, Rik Vandenberghe29,30, Sebastiaan Engelborghs31,32, Gael Nicolas33, Florence Pasquier34, Olivier Hanon35, Carole Dufouil36, Claudine Berr37, Stéphanie Debette36, Jean-François Dartigues36, Gianfranco Spalletta38, Benedetta Nacmias39,40, Vincenzo Solfrezzi41, Barbara Borroni42, Lucio Tremolizzo43, Davide Seripa44, Paolo Caffarra45, Antonio Daniele46,47, Daniela Galimberti48,49, Innocenzo Rainero50, Luisa Benussi51, Alesio Squassina52, Patrizia Mecoci53, Lucilla Parnetti54, Carlo Masullo55, Beatrice Arosio56, John Hardy57, Simon Mead58, Kevin Morgan59, Clive Holmes60, Patrick Kehoe61, Bob Woods62, EADB, Charge, ADGC, Jin Sha15,16, Yi Zhao15,63, Chien-Yueh Lee15,63, Pavel P.
    [Show full text]
  • WO 2019/079361 Al 25 April 2019 (25.04.2019) W 1P O PCT
    (12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT) (19) World Intellectual Property Organization I International Bureau (10) International Publication Number (43) International Publication Date WO 2019/079361 Al 25 April 2019 (25.04.2019) W 1P O PCT (51) International Patent Classification: CA, CH, CL, CN, CO, CR, CU, CZ, DE, DJ, DK, DM, DO, C12Q 1/68 (2018.01) A61P 31/18 (2006.01) DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, C12Q 1/70 (2006.01) HR, HU, ID, IL, IN, IR, IS, JO, JP, KE, KG, KH, KN, KP, KR, KW, KZ, LA, LC, LK, LR, LS, LU, LY, MA, MD, ME, (21) International Application Number: MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, PCT/US2018/056167 OM, PA, PE, PG, PH, PL, PT, QA, RO, RS, RU, RW, SA, (22) International Filing Date: SC, SD, SE, SG, SK, SL, SM, ST, SV, SY, TH, TJ, TM, TN, 16 October 2018 (16. 10.2018) TR, TT, TZ, UA, UG, US, UZ, VC, VN, ZA, ZM, ZW. (25) Filing Language: English (84) Designated States (unless otherwise indicated, for every kind of regional protection available): ARIPO (BW, GH, (26) Publication Language: English GM, KE, LR, LS, MW, MZ, NA, RW, SD, SL, ST, SZ, TZ, (30) Priority Data: UG, ZM, ZW), Eurasian (AM, AZ, BY, KG, KZ, RU, TJ, 62/573,025 16 October 2017 (16. 10.2017) US TM), European (AL, AT, BE, BG, CH, CY, CZ, DE, DK, EE, ES, FI, FR, GB, GR, HR, HU, ΓΕ , IS, IT, LT, LU, LV, (71) Applicant: MASSACHUSETTS INSTITUTE OF MC, MK, MT, NL, NO, PL, PT, RO, RS, SE, SI, SK, SM, TECHNOLOGY [US/US]; 77 Massachusetts Avenue, TR), OAPI (BF, BJ, CF, CG, CI, CM, GA, GN, GQ, GW, Cambridge, Massachusetts 02139 (US).
    [Show full text]