Prediction of Novel Bioactive Micropeptides in the Immune System

Total Page:16

File Type:pdf, Size:1020Kb

Prediction of Novel Bioactive Micropeptides in the Immune System Prediction of novel bioactive micropeptides in the immune system Fengyuan Hu The Babraham Institute St Edmund’s College, University of Cambridge June 2020 Submitted for the degree of Doctor of Philosophy at the University of Cambridge Declaration of Originality This thesis is the result of my own work and includes nothing which is the outcome of work done in collaboration except as declared in the Preface and specified in the text. It is not substantially the same as any that I have submitted, or, is being concurrently submitted for a degree or diploma or other qualification at the University of Cambridge or any other University or similar institution except as declared in the Preface and specified in the text. I further state that no substantial part of my dissertation has already been submitted, or, is being concurrently submitted for any such degree, diploma or other qualification at the University of Cambridge or any other University or similar institution except as declared in the Preface and specified in the text. It does not exceed the prescribed word limit for the relevant Degree Committee. Fengyuan Hu 1 Table of Contents Acknowledgements ……...……………………………………………………………………...06 Abstract……...…………………………………………………………………………………..08 Abbreviation……………………………………………………………………………………..09 1 Introduction……………………...…………..10 1.1 Roles of bioactive peptides and small proteins in the immune system ....................................... 111 1.2 Small open reading frames (smORFs) and micropeptides ............................................................. 122 1.3 Identification of smORFs and micropeptides ..................................................................................... 188 Approaches to identify protein-coding smORFs .......................................................................................... 21. Bioinformatics ................................................................................................................... 22 Transcriptomics .............................................................................................................. 244 Proteomics ...................................................................................................................... 277 1.4 Functional characterization of micropeptides..................................................................................... 288 1.5 smORF and mircopeptide categories ...................................................................................................... 30 Canonical smORFs ................................................................................................................................................. 33 Upstream ORFs ........................................................................................................................................................ 33 Downstream smORFs ............................................................................................................................................ 34 smORFs in non-coding RNAs ............................................................................................................................. 34 1.6 Importance of smORFs to health and disease ...................................................................................... 35 1.7 Research objectives ...................................................................................................................................... 35 2 Materials and Methods…………….………..37 2.1 Datasets ............................................................................................................................................................ 38 2.2 Reference genome, transcriptome and annotation .............................................................................. 40 2.3 Identifying putative smORFs .................................................................................................................... 42 2 2.4 Sequencing data processing ....................................................................................................................... 43 2.5 ORF discovery ............................................................................................................................................... 44 2.6 Analysis of predicted smORFs ................................................................................................................. 45 2.7 Plasmid design and smORF cloning ....................................................................................................... 46 3 Computational Pipeline to Predict Actively Translated smORFs…….………………….……49 3.1 Summary ......................................................................................................................................................... 50 3.2 Overview of the pipeline ............................................................................................................................ 51 3.3 Prediction of putative smORFs ................................................................................................................. 55 3.4 Sequencing data QC and processing ....................................................................................................... 56 Adapter trimming .................................................................................................................................................... 56 Contaminant removal ............................................................................................................................................. 57 FastQ screen .............................................................................................................................................................. 57 Sequence alignment to the reference genome................................................................................................. 58 Metagene Analysis .................................................................................................................................................. 58 RPF mapping rules and P-site offsets calculation ......................................................................................... 59 Read phasing in Ribo-Seq ..................................................................................................................................... 61 Transcript expression estimation ........................................................................................................................ 61 3.5 ORF discovery ............................................................................................................................................... 61 ORF discovery steps ............................................................................................................................................... 61 3.6 Pipeline Output .............................................................................................................................................. 64 3.7 Results .............................................................................................................................................................. 65 Ribosome profiling data quality .......................................................................................................................... 65 Missing rRNA reference sequences ................................................................................................................... 67 Genome alignment .................................................................................................................................................. 70 Determine P-site offset and sub-codon phasing ............................................................................................. 73 Sufficient read coverage to predict smORFs .................................................................................................. 75 Reproduce the published smORFs ..................................................................................................................... 76 Comparison between ORFLine and RiboCode .............................................................................................. 77 3.8 Pipeline availability ...................................................................................................................................... 80 3.9 Discussion ............................................................................................................................... 80 3 4 Properties of smORFs and Functional Validation of Micropeptide…………...………...82 4.1 Summary ......................................................................................................................................................... 83 4.2 Predicted smORFs ........................................................................................................................................ 83 4.3 smORF classification ................................................................................................................................... 86 4.4 Start codon usage in smORFs ................................................................................................................. 877 4.5 smORF conservation ................................................................................................................................. 899 4.6 Canonical smORFs ......................................................................................................................................
Recommended publications
  • Daniel Bernstein, MD Associate Dean for Curriculum and Scholarship Stanford University School of Medicine Alfred Woodley Salter and Mabel G
    RESEARCHER PROFILES Daniel Bernstein, MD Associate Dean for Curriculum and Scholarship Stanford University School of Medicine Alfred Woodley Salter and Mabel G. Salter Endowed Professor of Pediatrics (Cardiology) Stanford University Former Division Chief, Pediatric Cardiology Former Director, Children’s Heart Center, Lucile Packard Children’s Hospital at Stanford EMAIL [email protected] PROFILE med.stanford.edu/profiles/Daniel-Bernstein LAB murinecvcore.stanford.edu CURRENT RESEARCH EDUCATION/TRAINING Our recent work has focused on the mechanism by which mutations in sarcomeric proteins such MD New York University as myosin lead to the clinical phenotypes of hypertrophic cardiomyopathy (HCM). Utilizing human induced pluripotent stem cell-derived cardiomyocytes, with mutations induced by CRISPR/Cas9 PEDIATRICS RESIDENCY Montefiore Medical Center gene editing, we are undertaking a multi-scale approach ranging from structural and function studies on the single myosin molecule, to the individual myofibril, to whole cells and to microengineered MEDICAL EDUCATION FELLOWSHIP Albert Einstein College of Medicine tissues. To better understand cardiomyocyte mechano-transduction, we are applying FRET sensors in critical sarcomeric and junctional proteins. We are also studying a large biobank of myocardial PEDIATRIC CARDIOLOGY FELLOWSHIP UCSF samples from patients with HCM, combining transcriptomics and metabolomics with measurements of mitochondrial function to determine the degree to which HCM is a disease of altered cardiac BOARD CERTIFICATION Pediatrics, ABP energetics. These studies will allow us to correlate findings from hiPSC-CMs with actual patient samples. Another focus of our lab has been on the molecular mechanisms of RV hypertrophy Pediatric Cardiology, ABP and its transition to RV failure, and how this differs from LV failure.
    [Show full text]
  • Requirement of the Fusogenic Micropeptide Myomixer for Muscle Formation in Zebrafish
    Requirement of the fusogenic micropeptide myomixer for muscle formation in zebrafish Jun Shia,b,1, Pengpeng Bia,b,c,1, Jimin Peid, Hui Lia,b,c, Nick V. Grishind,e, Rhonda Bassel-Dubya,b,c, Elizabeth H. Chena,b,2, and Eric N. Olsona,b,c,2 aDepartment of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390; bHamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390; cSenator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390; dHoward Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390; and eDepartment of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX 75390 Contributed by Eric N. Olson, September 28, 2017 (sent for review August 29, 2017; reviewed by Chen-Ming Fan and Thomas A. Rando) Skeletal muscle formation requires fusion of mononucleated myo- CRISPR/Cas9, we show that myomixer is required for zebrafish blasts to form multinucleated myofibers. The muscle-specific mem- myoblast fusion. We also identify additional distantly related brane proteins myomaker and myomixer cooperate to drive myomixer orthologs from genomes of turtle and elephant shark, mammalian myoblast fusion. Whereas myomaker is highly conserved which like zebrafish myomixer can substitute for mouse myomixer across diverse vertebrate species, myomixer is a micropeptide that to promote cell fusion together with myomaker in cultured cells. shows relatively weak cross-species conservation. To explore the By comparison of amino acid sequences across these diverse functional conservation of myomixer, we investigated the expres- species, we identify a unique protein motif required for the sion and function of the zebrafish myomixer ortholog.
    [Show full text]
  • ELABELA, a Peptide Hormone for Heart Development
    RESEARCH HIGHLIGHTS parasites and confirmed its role in OXA resistance by showing that RNA The cancer epigenome of mice and men interference knockdown of Smp_089320 in drug-sensitive parasites Genetically engineered mouse models (GEMMs) of human resulted in an increase in resistance. They found that the Smp_089320 disease rarely take into account epigenetic alterations, which protein in sensitive but not resistant strains showed sulfonation activity, contribute to many human cancers. Now, Stephen Tapscott and identifying a mechanism for the drug in which it acts as a sulfotrans- colleagues compare genome-wide patterns of cancer-specific ferase that activates OXA. Finally, the authors determined the crystal DNA methylation in human patients and three GEMMs of structure of Smp_089320 protein from sensitive parasites with OXA medulloblastoma (Epigenetics 8, 1254–1260, 2013). Using two bound and suggest that the mechanism of resistance involves disruption independent methods to measure CpG methylation, they find, of the drug-protein interaction in resistant strains. Comparative and in contrast to the hypermethylation patterns previously observed phylogenetic analysis with other schistosomes also suggests the basis in patients with medulloblastoma, that GEMMs had only modest for the species specificity in OXA drug action. OB increase in CpG methylation of gene promoters relative to wild- type controls. Whereas a human medulloblastoma tumor sample showed >60% increase in methylation at 121 loci, consistent ELABELA, a peptide hormone for heart with the authors’ previous work, there were only 0–16 such loci development in the GEMMs. They further extend these findings to mouse models of Burkitt lymphoma and breast cancer, which showed Bruno Reversade and colleagues identify a highly conserved gene encod- similar results.
    [Show full text]
  • DNA Damage Response
    biomolecules Editorial DNA Damage Response Valentyn Oksenych 1,2,3,4,* and Denis E. Kainov 1,5,* 1 Department for Cancer Research and Molecular Medicine (IKOM), Norwegian University of Science and Technology, 7491 Trondheim, Norway 2 Department of Biosciences and Nutrition (BioNuT), Karolinska Institutet, 14183 Huddinge, Sweden 3 KG Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, 0316 Oslo, Norway 4 Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway 5 Institute of Technology, University of Tartu, 50090 Tartu, Estonia * Correspondence: [email protected] (V.O.); [email protected] (D.E.K.) DNA in our cells is constantly modified by internal and external factors. For exam- ple, metabolic byproducts, ionizing radiation (IR), ultraviolet (UV) light, and medicines can induce spontaneous DNA lesions [1–3]. However, DNA modifications can also be programmed. In particular, the recombination activating gene (RAG) can induce breaks generated during the V(D)J recombination in developing B and T lymphocytes [1,2]. In addition, activation-induced cytidine deaminase (AID) makes DNA break during the class-switch recombination (CSR) and somatic hypermutation (SHM) in B cells [1,2]. One focus of this Special Issue is on the non-homologous end-joining (NHEJ) DNA repair pathway and DNA repair and DNA damage response (DDR) factors. Oksenych et al. and others found the functional redundancy of these factors in mammalian cells. In particu- lar, a genetic interaction was found between the X-ray repair cross-complementing protein 4 (XRCC4)-like factor (XLF, also known as Cernunnos) and the DNA-dependent protein kinase catalytic subunit (DNA-PKcs) [4,5], the paralog of XRCC4 and XLF (PAXX) [6–9], and the modulator of retrovirus infection (MRI, also known as Cyren) [10].
    [Show full text]
  • DPP9 Is an Endogenous and Direct Inhibitor of the NLRP1 Inflammasome That Guards Against Human Auto-Inflammatory Diseases
    bioRxiv preprint doi: https://doi.org/10.1101/260919; this version posted February 7, 2018. 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. DPP9 is an endogenous and direct inhibitor of the NLRP1 inflammasome that guards against human auto-inflammatory diseases Franklin L. Zhong1,2,3,*, Kim Robinson2,3, Chrissie Lim1, Cassandra R. Harapas4, Chien- Hsiung Yu4, William Xie2, Radoslaw M. Sobota1, Veonice Bijin Au1, Richard Hopkins1, John E. Connolly1,6,7, Seth Masters4,5 , Bruno Reversade1,2,8,9,10 *, # 1. Institute of Molecular and Cell Biology, A*STAR, 61 Biopolis Drive, Proteos, Singapore 138673 2. Institute of Medical Biology, A*STAR, 8A Biomedical Grove, Immunos, Singapore 138648 3. Skin Research Institute of Singapore (SRIS), 8A Biomedical Grove, Immunos, Singapore 138648 4. Inflammation division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia. 5. Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010 Australia 6. Institute of Biomedical Studies, Baylor University, Waco, Texas 76712, USA 7. Department of Microbiology and Immunology, National University of Singapore, 5 Science Drive 2, Singapore 117545 8. Reproductive Biology Laboratory, Obstetrics and Gynaecology, Academic Medical Center (AMC), Meibergdreef 9, 1105 AZ Amsterdam-Zuidoost, Netherlands 9. Department of Paediatrics, National University of Singapore, 1E Kent Ridge Road, Singapore 119228 10. Medical Genetics Department, Koç University School of Medicine, 34010 Istanbul, Turkey * Corresponding authors. F.L.Z., [email protected]; B.R., [email protected] # Lead contact 1 bioRxiv preprint doi: https://doi.org/10.1101/260919; this version posted February 7, 2018.
    [Show full text]
  • Generation of a Mouse Model Lacking the Non-Homologous End-Joining Factor Mri/Cyren
    biomolecules Article Generation of a Mouse Model Lacking the Non-Homologous End-Joining Factor Mri/Cyren 1,2, 1,2, 1,2, 1,2 Sergio Castañeda-Zegarra y , Camilla Huse y, Øystein Røsand y, Antonio Sarno , Mengtan Xing 1,2, Raquel Gago-Fuentes 1,2, Qindong Zhang 1,2, Amin Alirezaylavasani 1,2, Julia Werner 1,2,3, Ping Ji 1, Nina-Beate Liabakk 1, Wei Wang 1, Magnar Bjørås 1,2 and Valentyn Oksenych 1,2,4,* 1 Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, 7491 Trondheim, Norway; [email protected] (S.C.-Z.); [email protected] (C.H.); [email protected] (Ø.R.); [email protected] (A.S.); [email protected] (M.X.); [email protected] (R.G.-F.); [email protected] (Q.Z.); [email protected] (A.A.); [email protected] (J.W.); [email protected] (P.J.); [email protected] (N.-B.L.); [email protected] (W.W.); [email protected] (M.B.) 2 St. Olavs Hospital, Trondheim University Hospital, Clinic of Medicine, Postboks 3250, Sluppen, 7006 Trondheim, Norway 3 Molecular Biotechnology MS programme, Heidelberg University, 69120 Heidelberg, Germany 4 Department of Biosciences and Nutrition (BioNut), Karolinska Institutet, 14183 Huddinge, Sweden * Correspondence: [email protected]; Tel.: +47-913-43-084 These authors contributed equally to this work. y Received: 7 November 2019; Accepted: 26 November 2019; Published: 28 November 2019 Abstract: Classical non-homologous end joining (NHEJ) is a molecular pathway that detects, processes, and ligates DNA double-strand breaks (DSBs) throughout the cell cycle.
    [Show full text]
  • Computational Analysis Predicts Hundreds of Coding Lncrnas in Zebrafish
    biology Communication Computational Analysis Predicts Hundreds of Coding lncRNAs in Zebrafish Shital Kumar Mishra 1,2 and Han Wang 1,2,* 1 Center for Circadian Clocks, Soochow University, Suzhou 215123, China; [email protected] 2 School of Biology & Basic Medical Sciences, Medical College, Soochow University, Suzhou 215123, China * Correspondence: [email protected] or [email protected]; Tel.: +86-512-6588-2115 Simple Summary: Noncoding RNAs (ncRNAs) regulate a variety of fundamental life processes such as development, physiology, metabolism and circadian rhythmicity. RNA-sequencing (RNA- seq) technology has facilitated the sequencing of the whole transcriptome, thereby capturing and quantifying the dynamism of transcriptome-wide RNA expression profiles. However, much remains unrevealed in the huge noncoding RNA datasets that require further bioinformatic analysis. In this study, we applied six bioinformatic tools to investigate coding potentials of approximately 21,000 lncRNAs. A total of 313 lncRNAs are predicted to be coded by all the six tools. Our findings provide insights into the regulatory roles of lncRNAs and set the stage for the functional investigation of these lncRNAs and their encoded micropeptides. Abstract: Recent studies have demonstrated that numerous long noncoding RNAs (ncRNAs having more than 200 nucleotide base pairs (lncRNAs)) actually encode functional micropeptides, which likely represents the next regulatory biology frontier. Thus, identification of coding lncRNAs from ever-increasing lncRNA databases would be a bioinformatic challenge. Here we employed the Coding Potential Alignment Tool (CPAT), Coding Potential Calculator 2 (CPC2), LGC web server, Citation: Mishra, S.K.; Wang, H. Coding-Non-Coding Identifying Tool (CNIT), RNAsamba, and MicroPeptide identification tool Computational Analysis Predicts (MiPepid) to analyze approximately 21,000 zebrafish lncRNAs and computationally to identify Hundreds of Coding lncRNAs in 2730–6676 zebrafish lncRNAs with high coding potentials, including 313 coding lncRNAs predicted Zebrafish.
    [Show full text]
  • Stem Cell Metabolic and Epigenetic Reprogramming
    RESEARCH HIGHLIGHTS Cell 16, 39–50, 2015). They discovered that inactivating Rb facilitates Enhancer evolution in mammals reprogramming of fibroblasts to a pluripotent state. Surprisingly, their Widespread changes in regulatory genomic regions have data indicate that this does not involve interference with the cell cycle but underpinned mammalian evolution, but our knowledge about instead that Rb directly binds to and represses pluripotency-associated these regions is still incomplete. Paul Flicek, Duncan Odom and loci such as Oct4 (Pou5f1) and Sox2. Loss of Rb seems to compensate colleagues have now contributed to a better understanding of for the omission of Sox2 from the cocktail of reprogramming factors. how the noncoding portions of mammalian genomes have been Furthermore, genetic disruption of Sox2 precludes tumor formation in reshaped over the last 180 million years (Cell 160, 554–566, mice lacking functional Rb protein. This study positions Rb as a repres- 2015). They characterized active promoters and enhancers in sor of the pluripotency gene regulatory network and suggests that loss of liver samples from 20 mammalian species—from Tasmanian Rb might clear the path for Sox2, or other master regulators of stem cell devil to human—by examining the genome-wide enrichment identity, to induce cancer. It will be interesting to analyze the potential profiles of H3K4me3 and H3K27ac, two histone modifications role of Rb in other types of in vitro reprogramming. TF associated with transcriptional activity. Their analyses suggest that rapid enhancer evolution and high promoter conservation are fundamental traits of mammalian genomes. Intriguingly, they Micropeptide regulates muscle performance find that the majority of newly evolved enhancers originated via It is becoming increasingly recognized that some transcripts anno- functional exaptation of ancestral DNA and not through clade- tated as long noncoding RNAs (lncRNAs) contain short ORFs that specific expansions of repeat elements.
    [Show full text]
  • Novel Mutations in the Ciliopathy-Associated Gene CPLANE1 (C5orf42) Cause OFD Syndrome Type VI Rather Than Joubert Syndrome
    Accepted Manuscript Novel mutations in the ciliopathy-associated gene CPLANE1 (C5orf42) cause OFD syndrome type VI rather than Joubert syndrome Carine Bonnard, Mohammad Shboul, Seyed Hassan Tonekaboni, Alvin Yu Jin Ng, Sumanty Tohari, Kakaly Ghosh, Angeline Lai, Jiin Ying Lim, Ene Choo Tan, Louise Devisme, Morgane Stichelbout, Adila Alkindi, Nazreen Banu, Zafer Yüksel, Jamal Ghoumid, Nadia Elkhartoufi, Lucile Boutaud, Alessia Micalizzi, Maggie Siewyan Brett, Byrappa Venkatesh, Enza Maria Valente, Tania Attié-Bitach, Bruno Reversade, Ariana Kariminejad PII: S1769-7212(17)30410-X DOI: 10.1016/j.ejmg.2018.03.012 Reference: EJMG 3440 To appear in: European Journal of Medical Genetics Received Date: 30 June 2017 Revised Date: 28 March 2018 Accepted Date: 28 March 2018 Please cite this article as: C. Bonnard, M. Shboul, S.H. Tonekaboni, A.Y.J. Ng, S. Tohari, K. Ghosh, A. Lai, J.Y. Lim, E.C. Tan, L. Devisme, M. Stichelbout, A. Alkindi, N. Banu, Z. Yüksel, J. Ghoumid, N. Elkhartoufi, L. Boutaud, A. Micalizzi, M.S. Brett, B. Venkatesh, E.M. Valente, T. Attié-Bitach, B. Reversade, A. Kariminejad, Novel mutations in the ciliopathy-associated gene CPLANE1 (C5orf42) cause OFD syndrome type VI rather than Joubert syndrome, European Journal of Medical Genetics (2018), doi: 10.1016/j.ejmg.2018.03.012. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
    [Show full text]
  • The Lncrna Toolkit: Databases and in Silico Tools for Lncrna Analysis
    non-coding RNA Review The lncRNA Toolkit: Databases and In Silico Tools for lncRNA Analysis Holly R. Pinkney † , Brandon M. Wright † and Sarah D. Diermeier * Department of Biochemistry, University of Otago, Dunedin 9016, New Zealand; [email protected] (H.R.P.); [email protected] (B.M.W.) * Correspondence: [email protected] † These authors contributed equally to this work. Received: 29 November 2020; Accepted: 15 December 2020; Published: 16 December 2020 Abstract: Long non-coding RNAs (lncRNAs) are a rapidly expanding field of research, with many new transcripts identified each year. However, only a small subset of lncRNAs has been characterized functionally thus far. To aid investigating the mechanisms of action by which new lncRNAs act, bioinformatic tools and databases are invaluable. Here, we review a selection of computational tools and databases for the in silico analysis of lncRNAs, including tissue-specific expression, protein coding potential, subcellular localization, structural conformation, and interaction partners. The assembled lncRNA toolkit is aimed primarily at experimental researchers as a useful starting point to guide wet-lab experiments, mainly containing multi-functional, user-friendly interfaces. With more and more new lncRNA analysis tools available, it will be essential to provide continuous updates and maintain the availability of key software in the future. Keywords: non-coding RNAs; long non-coding RNAs; databases; computational analysis; bioinformatic prediction software; RNA interactions; coding potential; RNA structure; RNA function 1. Introduction For decades, the human genome was thought to be a desert of ‘junk DNA’ with sporadic oases of transcriptionally active genes, most of them coding for proteins.
    [Show full text]
  • The Response to DNA Damage at Telomeric Repeats and Its Consequences for Telomere Function
    Review The Response to DNA Damage at Telomeric Repeats and Its Consequences for Telomere Function Ylli Doksani IFOM, The FIRC Institute of Molecular Oncology, via Adamello 16, 20139 Milan, Italy; [email protected]; Tel.: +39-02-574303258 Received: 26 March 2019; Accepted: 18 April 2019; Published: 24 April 2019 Abstract: Telomeric repeats, coated by the shelterin complex, prevent inappropriate activation of the DNA damage response at the ends of linear chromosomes. Shelterin has evolved distinct solutions to protect telomeres from different aspects of the DNA damage response. These solutions include formation of t-loops, which can sequester the chromosome terminus from DNA-end sensors and inhibition of key steps in the DNA damage response. While blocking the DNA damage response at chromosome ends, telomeres make wide use of many of its players to deal with exogenous damage and replication stress. This review focuses on the interplay between the end-protection functions and the response to DNA damage occurring inside the telomeric repeats, as well as on the consequences that telomere damage has on telomere structure and function. Keywords: telomere maintenance; shelterin complex; end-protection problem; telomere damage; telomeric double strand breaks; telomere replication; alternative lengthening of telomeres 1. Telomere Structure and End-Protection Functions Mammalian telomeres are made of tandem TTAGGG repeats that extend several kilobases and terminate with a 3′ single-stranded overhang about 50–400 nucleotides long. Telomeric repeats are coated in a sequence-specific fashion by a 6-protein complex named shelterin that prevents the activation of the Double Strand Break (DSB) response at chromosome ends [1-3].
    [Show full text]
  • Endothelial Cell Dynamics in Vascular Development: Insights from Live-Imaging in Zebrafish
    fphys-11-00842 July 20, 2020 Time: 12:10 # 1 REVIEW published: 22 July 2020 doi: 10.3389/fphys.2020.00842 Endothelial Cell Dynamics in Vascular Development: Insights From Live-Imaging in Zebrafish Kazuhide S. Okuda1,2 and Benjamin M. Hogan1,2,3* 1 Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia, 2 Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia, 3 Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, VIC, Australia The formation of the vertebrate vasculature involves the acquisition of endothelial cell identities, sprouting, migration, remodeling and maturation of functional vessel networks. To understand the cellular and molecular processes that drive vascular development, live-imaging of dynamic cellular events in the zebrafish embryo have proven highly informative. This review focusses on recent advances, new tools and new insights from imaging studies in vascular cell biology using zebrafish as a model system. Keywords: vasculogenesis, angiogenesis, lymphangiogenesis, anastomosis, endothelial cell, zebrafish Edited by: INTRODUCTION Elizabeth Anne Vincent Jones, KU Leuven, Belgium Vascular development is an early and essential process in the formation of a viable vertebrate Reviewed by: embryo. Blood and lymphatic vascular networks are composed of a complex mix of cell types: for Anna Rita Cantelmo, example smooth muscle cells, fibroblasts, pericytes and immune cells are intimately associated and Université Lille Nord de France, even integrated within mature vessel walls (Rouget, 1873; Horstmann, 1952; Nicosia and Madri, France 1987; Fantin et al., 2010; Gordon et al., 2010). The inner lining of the vasculature, made up of Jingjing Zhang, endothelial cells (ECs), is the earliest part of the vasculature to develop in the embryo and is Affiliated Hospital of Guangdong Medical University, China instructive in recruiting other lineages and cell types as the vascular system matures.
    [Show full text]