Human Pluripotent Reprogramming with CRISPR Activators

Total Page:16

File Type:pdf, Size:1020Kb

Human Pluripotent Reprogramming with CRISPR Activators ARTICLE DOI: 10.1038/s41467-018-05067-x OPEN Human pluripotent reprogramming with CRISPR activators Jere Weltner1, Diego Balboa 1, Shintaro Katayama2, Maxim Bespalov1, Kaarel Krjutškov2,3, Eeva-Mari Jouhilahti1, Ras Trokovic1, Juha Kere 1,2,4,5 & Timo Otonkoski 1,6 CRISPR-Cas9-based gene activation (CRISPRa) is an attractive tool for cellular reprogram- ming applications due to its high multiplexing capacity and direct targeting of endogenous 1234567890():,; loci. Here we present the reprogramming of primary human skin fibroblasts into induced pluripotent stem cells (iPSCs) using CRISPRa, targeting endogenous OCT4, SOX2, KLF4, MYC, and LIN28A promoters. The low basal reprogramming efficiency can be improved by an order of magnitude by additionally targeting a conserved Alu-motif enriched near genes involved in embryo genome activation (EEA-motif). This effect is mediated in part by more efficient activation of NANOG and REX1. These data demonstrate that human somatic cells can be reprogrammed into iPSCs using only CRISPRa. Furthermore, the results unravel the invol- vement of EEA-motif-associated mechanisms in cellular reprogramming. 1 Research Programs Unit, Molecular Neurology and Biomedicum Stem Cell Centre, Faculty of Medicine, University of Helsinki, Helsinki 00014, Finland. 2 Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge 141 83, Sweden. 3 Competence Centre on Health Technologies, Tartu 50410, Estonia. 4 School of Basic and Medical Biosciences, Guy’s Hospital, King’s College London, London SE1 9RT, UK. 5 Folkhälsan Institute of Genetics, Helsinki 00290, Finland. 6 Children’s Hospital, Helsinki University Central Hospital, University of Helsinki, Helsinki 00290, Finland. These authors contributed equally: Juha Kere, Timo Otonkoski. Correspondence and requests for materials should be addressed to J.W. (email: jere.weltner@helsinki.fi) or to J.K. (email: [email protected]) or to T.O. (email: timo.otonkoski@helsinki.fi) NATURE COMMUNICATIONS | (2018) 9:2643 | DOI: 10.1038/s41467-018-05067-x | www.nature.com/naturecommunications 1 ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/s41467-018-05067-x RISPRa system relies on sequence specific recruitment of a reprogramming human cells, including primary adult human catalytically inactivated version of Cas9 protein (dCas9) to skin fibroblasts, into induced pluripotent stem cells by CRISPRa. C fi genomic sequences de ned by short guide RNA (gRNA) This opens up important possibilities for the development of molecules1–3. The fact that dCas9 effectors can be used to control more extensive CRISPRa reprogramming approaches for human transcription of targeted endogenous loci makes it useful for cells. Efficiency of the method depends on the targeting of the mediating cellular reprogramming, which requires silencing and EEA-motif, which results in improved activation of a subset of activation of endogenous gene sets for proper cell type conver- endogenous genes that work as reprogramming factors, including sion. CRISPRa may therefore be beneficial in overcoming NANOG and ZFP42 (REX1). These results also exemplify the reprogramming barriers that limit reprogramming efficiency and potential in targeting cell type enriched regulatory elements for contribute to the emergence of partially reprogrammed stable cell controlling cell fate. populations, often associated with inadequate endogenous gene activation or silencing4–6. Previously, dCas9 effectors have been Results used to mediate differentiation, transdifferentiation, and repro- gramming of various mouse and human cell types, but complete CRISPRa-mediated reprogramming of NSCs and EEA target- ing. We began human cell reprogramming with CRISPRa using a pluripotent reprogramming of human cells using only CRISPRa fi has not yet been reported7–15. simpli ed reprogramming scheme. CRISPRa-mediated POU5F1 In addition to gene activation, dCas9 effector mediated DNA (OCT4) activation has been used to replace transgenic OCT4 in human fibroblast reprogramming, while the transgenic expres- targeting can be used to decipher the functions of genomic reg- fi ulatory elements16–18. Combining reprogramming factor pro- sion of only OCT4 has been shown to be suf cient for the reprogramming of neuroepithelial stem cells (NSCs) into moter targeting gRNAs with targeting of other regulatory 12,22 elements has high potential in mediating comprehensive resetting iPSCs . We therefore combined CRISPRa-mediated OCT4 of gene regulatory networks. A conserved Alu-motif was recently activation and NSC reprogramming as an initial model using fi trimethoprim (TMP) stabilized SpdCas9VP192 fused with P65- reported to be enriched in the promoter areas of the rst genes 23 expressed during human embryo genome activation (EGA)19. HSF1 activator domain (DDdCas9VPH) under doxycycline This sequence is thus likely to be involved in the control of early (DOX) inducible promoter (Fig. 1a, b). Expression of embryonic transcriptional networks. As human embryos can DDdCas9VPH and OCT4 targeting guides in iPSC-derived NSCs 20 resulted in the emergence of pluripotent cells in a DOX and TMP reprogram somatic cell nuclei , we hypothesized that targeting – this EGA-enriched Alu-motif (EEA-motif) could enhance dependent manner (Fig. 1c e). These cells could be expanded into reprogramming of somatic cells to pluripotency. stable dCas9 independent cell lines (Fig. 1c and Supplementary Fig. 1). This demonstrated that CRISPRa mediated activation of Development of reprogramming approaches for faithful reca- fi pitulation of cellular phenotypes is an important task, considering endogenous OCT4 alone was suf cient to reprogram NSCs to the increasing pace with which reprogrammed cells are moving iPSCs. toward clinical trials21. Here we describe a method for To determine if EEA-motif targeting could improve CRISPRa reprogramming of NSCs, we designed a set of five 14 nt gRNAs a c OCT4 SOX2 TRA-1-60 NANOG TRA-1-81 OCT4 dCas9VPH dCas9 VP192 P65 HSF1 b dCas9VPH + VIM TUBB3 α-SMA AFP SOX17 iPSC NSC OCT4 activation iPSC d EGA-enriched Alu-motif targeting gRNAs e CRISPRa NSC reprogramming with dCas9VPH 4000 3500 EEA-g10 3000 EEA-g7 2 2500 1 Bits 2000 0 1 3 5 6 7 8 9 2 4 1500 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 EEA-g3 1000 EEA-g1 500 EEA-g2 Colonies per 100,000 cells 0 no DOX no OCT4 gRNA OCT4 gRNA + EEA gRNA TMP EEA gRNA + DOX + TMP Fig. 1 CRISPRa-mediated reprogramming of NSCs and EEA-motif targeting. a Schematic representation of dCas9VPH structure. b Schematic representation of NSC reprogramming into iPSCs with dCas9VPH mediated OCT4 activation. c Immunocytochemical detection of pluripotency markers in NCS-derived iPSCs (top row) and tri-lineage differentiation in plated embryoid bodies (bottom row). Nuclei stained blue. Scale bar = 200 µm. d Targeting of EGA enriched Alu-motif with SpdCas9 gRNAs. e Quantification of iPSC-like alkaline phosphatase positive colonies induced from NSCs. n = 6 independent inductions (P = 0.053, OCT4 targeting with EEA-gRNAs vs. without EEA-gRNAs). Data presented as mean ± s.e.m., two-tailed Student’s t-test 2 NATURE COMMUNICATIONS | (2018) 9:2643 | DOI: 10.1038/s41467-018-05067-x | www.nature.com/naturecommunications NATURE COMMUNICATIONS | DOI: 10.1038/s41467-018-05067-x ARTICLE a OCT4 4 53 2 1 SOX2 1 23 45 KLF4 5 4 321 MYC 54 3 21 LIN28A 5 4 32 1 NANOG 1 23 45 GENE –600 bp –400 bp –200 bp TSS b d dCas9VPH HEK293 gene activation with dCas9VPH + OMKSL gRNA plasmid 100 *** * 10 OCT4 4 53 2 1 Pool 1–5 *** 1 *** 0.1 ** SOX2 1 23 4 5 Pool 1–5 0.01 0.001 KLF4 5 43 2 1 Pool 1–5 mRNA expression relative to hESC 0.0001 TdT ctrl TdT ctrl TdT ctrl TdT ctrl TdT ctrl MYC 5 43 2 1 Pool 1–5 OMKSL OMKSL OMKSL OMKSL OMKSL OCT4 SOX2 KLF4 LIN28A MYC Fibroblast gene activation with dCas9VPH + OMKSL gRNA plasmid LIN28A 5 43 2 1 Pool 1–5 100 10 1 NANOG 1 23 4 5 Pool 1–5 ** ** 0.1 c MYC.1 OCT4.4 SOX2.3 LIN28.4 0.01 OCT4.1 KLF4.3 OCT4.5 LIN28.1 LIN28.3 SOX2.4 0.001 mRNA expression relative to hESC 0.0001 TdT ctrl TdT ctrl TdT ctrl TdT ctrl TdT ctrl OMKSL gRNA plasmid OMKSL OMKSL OMKSL OMKSL OMKSL OCT4 SOX2 KLF4 LIN28A MYC Fig. 2 Optimization of dCas9 activator and gRNA targeting in HEK293 for reprogramming factor activation. a Locations of promoter targeting gRNAs for reprogramming factors (OCT4, SOX2, KLF4, C-MYC, LIN28A, and NANOG) in relation to transcription start site. b Immunocytochemical staining of reprogramming factors after single gRNA activation and pooled mixture of five guides in HEK293 with dCas9VPH. Pictures are in similar order to guides in Fig. 2a. Best performing guides used for plasmid cloning are marked with dotted lines. Scale bar = 400 µm. c Schematic representation of concatenated reprogramming factor gRNA plasmid construction. d Reprogramming factor activation by qRT-PCR, in HEK293 cells 3 days after transfection and HFFs 4 days after electroporation, using transiently expressed dCas9VPH effector. n = 3, data are from three independently treated samples. Data presented as mean ± s.e.m., two tailed Student’s t-test. *P < 0.05, **P < 0.01, ***P < 0.001 targeting the 36 bp EEA consensus sequence (Fig. 1d). Addition rest of the factors. This suggested that additional guides for KLF4, of the EEA-motif gRNAs in the reprogramming mixture LIN28A, and MYC targeting would be required for efficient demonstrated a trend in increasing the number of alkaline activation of these genes. phosphatase (AP) positive colonies (P = 0.053, Student’s t-test) (Fig. 1e). This suggested that EEA-motif targeting could be useful Fibroblast reprogramming with CRISPRa. Electroporation of for improving CRISPRa reprogramming efficiency. primary skin fibroblasts with episomally replicating dCas9VPH plasmid, containing TP53 targeting shRNA, EEA-motif targeting Pluripotency factor activation with CRISPRa. To devise a gRNA plasmid, reprogramming factor targeting gRNA plasmid reprogramming system for fibroblasts based solely on CRISPRa, (OMKSL), and an additional KLF4 and MYC targeting gRNA we optimized the promoter targeting of single gRNAs to the plasmid (KM) resulted in the emergence of iPSC-like colonies canonical reprogramming factors OCT4, MYC, KLF4, SOX2, (Fig.
Recommended publications
  • An Alu Element-Based Model of Human Genome Instability George Wyndham Cook, Jr
    Louisiana State University LSU Digital Commons LSU Doctoral Dissertations Graduate School 2013 An Alu element-based model of human genome instability George Wyndham Cook, Jr. Louisiana State University and Agricultural and Mechanical College, [email protected] Follow this and additional works at: https://digitalcommons.lsu.edu/gradschool_dissertations Recommended Citation Cook, Jr., George Wyndham, "An Alu element-based model of human genome instability" (2013). LSU Doctoral Dissertations. 2090. https://digitalcommons.lsu.edu/gradschool_dissertations/2090 This Dissertation is brought to you for free and open access by the Graduate School at LSU Digital Commons. It has been accepted for inclusion in LSU Doctoral Dissertations by an authorized graduate school editor of LSU Digital Commons. For more information, please [email protected]. AN ALU ELEMENT-BASED MODEL OF HUMAN GENOME INSTABILITY A Dissertation Submitted to the Graduate Faculty of the Louisiana State University and Agricultural and Mechanical College in partial fulfillment of the requirements for the degree of Doctor of Philosophy in The Department of Biological Sciences by George Wyndham Cook, Jr. B.S., University of Arkansas, 1975 May 2013 TABLE OF CONTENTS LIST OF TABLES ...................................................................................................... iii LIST OF FIGURES .................................................................................................... iv LIST OF ABBREVIATIONS ......................................................................................
    [Show full text]
  • Table of Contents (PDF)
    July 26, 2011 u vol. 108 u no. 30 u 12187–12560 Cover image: Pictured is a Tasmanian devil (Sarcophilus harrisii), a carnivorous marsupial whose numbers are dwindling due to an infectious facial cancer called Devil Facial Tumor Disease. Webb Miller et al. sequenced the genome of devils from northwest and south- east Tasmania, spanning the range of this threatened species on the Australian island. The authors report that the sequences reveal a worrisome dearth of genetic diversity among devils, suggesting the need for genetically characterized stocks to help breed hardier devils that might be better equipped to fight diseases. See the article by Miller et al. on pages 12348–12353. Image courtesy of Stephan C. Schuster. From the Cover 12348 Decoding the Tasmanian devil genome 12283 Illuminating chromosomal architecture 12295 Symmetry of cultured cells 12319 Caloric restriction and infertility 12366 Genetic diversity among ants Contents COMMENTARIES 12189 Methyl fingerprinting of the nucleosome reveals the molecular mechanism of high-mobility group THIS WEEK IN PNAS nucleosomal-2 (HMGN2) association Catherine A. Musselman and Tatiana G. Kutateladze See companion article on page 12283 12187 In This Issue 12191 Examining the establishment of cellular axes using intrinsic chirality LETTERS (ONLINE ONLY) Jason C. McSheene and Rebecca D. Burdine See companion article on page 12295 E341 Difference between restoring and predicting 3D 12193 Secrets of palm oil biosynthesis revealed structures of the loops in G-protein–coupled Toni Voelker receptors by molecular modeling See companion article on page 12527 Gregory V. Nikiforovich, Christina M. Taylor, Garland R. Marshall, and Thomas J. Baranski E342 Reply to Nikiforovich et al.: Restoration of the loop regions of G-protein–coupled receptors Dahlia A.
    [Show full text]
  • Metallothionein Monoclonal Antibody, Clone N11-G
    Metallothionein monoclonal antibody, clone N11-G Catalog # : MAB9787 規格 : [ 50 uL ] List All Specification Application Image Product Rabbit monoclonal antibody raised against synthetic peptide of MT1A, Western Blot (Recombinant protein) Description: MT1B, MT1E, MT1F, MT1G, MT1H, MT1IP, MT1L, MT1M, MT2A. Immunogen: A synthetic peptide corresponding to N-terminus of human MT1A, MT1B, MT1E, MT1F, MT1G, MT1H, MT1IP, MT1L, MT1M, MT2A. Host: Rabbit enlarge Reactivity: Human, Mouse Immunoprecipitation Form: Liquid Enzyme-linked Immunoabsorbent Assay Recommend Western Blot (1:1000) Usage: ELISA (1:5000-1:10000) The optimal working dilution should be determined by the end user. Storage Buffer: In 20 mM Tris-HCl, pH 8.0 (10 mg/mL BSA, 0.05% sodium azide) Storage Store at -20°C. Instruction: Note: This product contains sodium azide: a POISONOUS AND HAZARDOUS SUBSTANCE which should be handled by trained staff only. Datasheet: Download Applications Western Blot (Recombinant protein) Western blot analysis of recombinant Metallothionein protein with Metallothionein monoclonal antibody, clone N11-G (Cat # MAB9787). Lane 1: 1 ug. Lane 2: 3 ug. Lane 3: 5 ug. Immunoprecipitation Enzyme-linked Immunoabsorbent Assay ASSP5 MT1A MT1B MT1E MT1F MT1G MT1H MT1M MT1L MT1IP Page 1 of 5 2021/6/2 Gene Information Entrez GeneID: 4489 Protein P04731 (Gene ID : 4489);P07438 (Gene ID : 4490);P04732 (Gene ID : Accession#: 4493);P04733 (Gene ID : 4494);P13640 (Gene ID : 4495);P80294 (Gene ID : 4496);P80295 (Gene ID : 4496);Q8N339 (Gene ID : 4499);Q86YX0 (Gene ID : 4490);Q86YX5
    [Show full text]
  • DNA Sequence Insertion and Evolutionary Variation in Gene Regulation (Mobile Elements/Long Terminal Repeats/Alu Sequences/Factor-Binding Sites) Roy J
    Proc. Natl. Acad. Sci. USA Vol. 93, pp. 9374-9377, September 1996 Colloquium Paper This paper was presented at a colloquium entitled "Biology of Developmental Transcription Control, " organized by Eric H. Davidson, Roy J. Britten, and Gary Felsenfeld, held October 26-28, 1995, at the National Academy of Sciences in Irvine, CA. DNA sequence insertion and evolutionary variation in gene regulation (mobile elements/long terminal repeats/Alu sequences/factor-binding sites) RoY J. BRITrEN Division of Biology, California Institute of Technology, 101 Dahlia Avenue, Corona del Mar, CA 92625 ABSTRACT Current evidence on the long-term evolution- 3 and 4). Sequence change, obscuring the original structure, ary effect of insertion of sequence elements into gene regions has occurred in the long history, and the underlying rate of is reviewed, restricted to cases where a sequence derived from base substitution that is responsible is known (5). a past insertion participates in the regulation of expression of The requirements for a convincing example are: (i) that a useful gene. Ten such examples in eukaryotes demonstrate there be a trace of a known class of elements present in gene that segments of repetitive DNA or mobile elements have been region; (ii) that there is evidence that it has been there long inserted in the past in gene regions, have been preserved, enough to not just be a transient mutation; (iii) that some sometimes modified by selection, and now affect control of sequence residue of the mobile element or repeat participates transcription ofthe adjacent gene. Included are only examples in regulation of expression of the gene; (iv) that the gene have in which transcription control was modified by the insert.
    [Show full text]
  • Epigenome-Wide Association Study of Wellbeing
    Twin Research and Human Genetics Volume 18 Number 6 pp. 710–719 C The Author(s) 2015 doi:10.1017/thg.2015.85 Epigenome-Wide Association Study of Wellbeing Bart M. L. Baselmans,1,2 Jenny van Dongen,1,2 Michel G. Nivard,1 Bochao D. Lin,1 BIOS Consortium,3 Nuno R. Zilhao,˜ 1 Dorret I. Boomsma,1,2,4 and Meike Bartels1,2,4 1Department of Biological Psychology, VU University, Amsterdam, the Netherlands 2EMGO+ Institute for Health and Care Research, VU University Medical Center, Amsterdam, the Netherlands 3The Biobank-Based Integrative Omics Study (BIOS) Consortium 4Neuroscience Campus Amsterdam, Amsterdam, the Netherlands Wellbeing (WB) is a major topic of research across several scientific disciplines, partly driven by its strong association with psychological and mental health. Twin-family studies have found that both genotype and environment play an important role in explaining the variance in WB. Epigenetic mechanisms, such as DNA methylation, regulate gene expression, and may mediate genetic and environmental effects on WB. Here, for the first time, we apply an epigenome-wide association study (EWAS) approach to identify differentially methylated sites associated with individual differences in WB. Subjects were part of the longitudinal survey studies of the Netherlands Twin Register (NTR) and participated in the NTR biobank project between 2002 and 2011. WB was assessed by a short inventory that measures satisfaction with life (SAT). DNA methylation was measured in whole blood by the Illumina Infinium HumanMethylation450 BeadChip (HM450k array) and the association between WB and DNA methylation level was tested at 411,169 autosomal sites. Two sites (cg10845147, p = 1.51 ∗ 10-8 and cg01940273, p = 2.34 ∗ 10-8) reached genome-wide significance following Bonferonni correction.
    [Show full text]
  • Table S1 the Four Gene Sets Derived from Gene Expression Profiles of Escs and Differentiated Cells
    Table S1 The four gene sets derived from gene expression profiles of ESCs and differentiated cells Uniform High Uniform Low ES Up ES Down EntrezID GeneSymbol EntrezID GeneSymbol EntrezID GeneSymbol EntrezID GeneSymbol 269261 Rpl12 11354 Abpa 68239 Krt42 15132 Hbb-bh1 67891 Rpl4 11537 Cfd 26380 Esrrb 15126 Hba-x 55949 Eef1b2 11698 Ambn 73703 Dppa2 15111 Hand2 18148 Npm1 11730 Ang3 67374 Jam2 65255 Asb4 67427 Rps20 11731 Ang2 22702 Zfp42 17292 Mesp1 15481 Hspa8 11807 Apoa2 58865 Tdh 19737 Rgs5 100041686 LOC100041686 11814 Apoc3 26388 Ifi202b 225518 Prdm6 11983 Atpif1 11945 Atp4b 11614 Nr0b1 20378 Frzb 19241 Tmsb4x 12007 Azgp1 76815 Calcoco2 12767 Cxcr4 20116 Rps8 12044 Bcl2a1a 219132 D14Ertd668e 103889 Hoxb2 20103 Rps5 12047 Bcl2a1d 381411 Gm1967 17701 Msx1 14694 Gnb2l1 12049 Bcl2l10 20899 Stra8 23796 Aplnr 19941 Rpl26 12096 Bglap1 78625 1700061G19Rik 12627 Cfc1 12070 Ngfrap1 12097 Bglap2 21816 Tgm1 12622 Cer1 19989 Rpl7 12267 C3ar1 67405 Nts 21385 Tbx2 19896 Rpl10a 12279 C9 435337 EG435337 56720 Tdo2 20044 Rps14 12391 Cav3 545913 Zscan4d 16869 Lhx1 19175 Psmb6 12409 Cbr2 244448 Triml1 22253 Unc5c 22627 Ywhae 12477 Ctla4 69134 2200001I15Rik 14174 Fgf3 19951 Rpl32 12523 Cd84 66065 Hsd17b14 16542 Kdr 66152 1110020P15Rik 12524 Cd86 81879 Tcfcp2l1 15122 Hba-a1 66489 Rpl35 12640 Cga 17907 Mylpf 15414 Hoxb6 15519 Hsp90aa1 12642 Ch25h 26424 Nr5a2 210530 Leprel1 66483 Rpl36al 12655 Chi3l3 83560 Tex14 12338 Capn6 27370 Rps26 12796 Camp 17450 Morc1 20671 Sox17 66576 Uqcrh 12869 Cox8b 79455 Pdcl2 20613 Snai1 22154 Tubb5 12959 Cryba4 231821 Centa1 17897
    [Show full text]
  • Transcriptome-Wide Effects of Inverted Sines on Gene Expression And
    Tajaddod et al. Genome Biology (2016) 17:220 DOI 10.1186/s13059-016-1083-0 RESEARCH Open Access Transcriptome-wide effects of inverted SINEs on gene expression and their impact on RNA polymerase II activity Mansoureh Tajaddod1†, Andrea Tanzer3†, Konstantin Licht2†, Michael T. Wolfinger2,3, Stefan Badelt3, Florian Huber1,4, Oliver Pusch2, Sandy Schopoff1, Michael Janisiw2, Ivo Hofacker3 and Michael F. Jantsch2,5* Abstract Background: Short interspersed elements (SINEs) represent the most abundant group of non-long-terminal repeat transposable elements in mammalian genomes. In primates, Alu elements are the most prominent and homogenous representatives of SINEs. Due to their frequent insertion within or close to coding regions, SINEs have been suggested to play a crucial role during genome evolution. Moreover, Alu elements within mRNAs have also been reported to control gene expression at different levels. Results: Here, we undertake a genome-wide analysis of insertion patterns of human Alus within transcribed portions of the genome. Multiple, nearby insertions of SINEs within one transcript are more abundant in tandem orientation than in inverted orientation. Indeed, analysis of transcriptome-wide expression levels of 15 ENCODE cell lines suggests a cis-repressive effect of inverted Alu elements on gene expression. Using reporter assays, we show that the negative effect of inverted SINEs on gene expression is independent of known sensors of double-stranded RNAs. Instead, transcriptional elongation seems impaired, leading to reduced mRNA levels. Conclusions: Our study suggests that there is a bias against multiple SINE insertions that can promote intramolecular base pairing within a transcript. Moreover, at a genome-wide level, mRNAs harboring inverted SINEs are less expressed than mRNAs harboring single or tandemly arranged SINEs.
    [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]
  • Anti-ALPPL2 Antibody (ARG58232)
    Product datasheet [email protected] ARG58232 Package: 100 μl anti-ALPPL2 antibody Store at: -20°C Summary Product Description Rabbit Polyclonal antibody recognizes ALPPL2 Tested Reactivity Hu, Ms Tested Application WB Host Rabbit Clonality Polyclonal Isotype IgG Target Name ALPPL2 Antigen Species Human Immunogen Recombinant fusion protein corresponding to aa. 20-280 of Human ALPPL2 (NP_112603.2). Conjugation Un-conjugated Alternate Names Alkaline phosphatase, placental-like; Placental alkaline phosphatase-like; PLAP-like; EC 3.1.3.1; Germ cell alkaline phosphatase; ALPG; Alkaline phosphatase Nagao isozyme; ALP-1; ALPPL; GCAP Application Instructions Application table Application Dilution WB 1:500 - 1:2000 Application Note * The dilutions indicate recommended starting dilutions and the optimal dilutions or concentrations should be determined by the scientist. Positive Control HeLa Calculated Mw 57 kDa Observed Size 70 kDa Properties Form Liquid Purification Affinity purified. Buffer PBS (pH 7.3), 0.02% Sodium azide and 50% Glycerol. Preservative 0.02% Sodium azide Stabilizer 50% Glycerol Storage instruction For continuous use, store undiluted antibody at 2-8°C for up to a week. For long-term storage, aliquot and store at -20°C. Storage in frost free freezers is not recommended. Avoid repeated freeze/thaw cycles. Suggest spin the vial prior to opening. The antibody solution should be gently mixed before use. Note For laboratory research only, not for drug, diagnostic or other use. www.arigobio.com 1/2 Bioinformation Gene Symbol ALPPL2 Gene Full Name alkaline phosphatase, placental-like 2 Background There are at least four distinct but related alkaline phosphatases: intestinal, placental, placental-like, and liver/bone/kidney (tissue non-specific).
    [Show full text]
  • Mapping of Quantitative Trait Loci for Milk Yield Traits on Bovine Chromosome 5 in the Fleckvieh Cattle
    From the Department of Veterinary Sciences Faculty of Veterinary Medicine Ludwig-Maximilians-Universität München Arbeit angefertigt unter der Leitung von Univ. Prof. Dr. Dr. habil. Martin Förster Mapping of Quantitative Trait Loci for Milk Yield Traits on Bovine Chromosome 5 in the Fleckvieh Cattle Inaugural–Dissertation For the attainment of Doctor Degree in Veterinary Medicine From the Faculty of Veterinary Medicine of the Ludwig-Maximilians-Universität München by Ashraf Fathy Said Awad from Sharkia- Egypt München 2011 Gedruckt mit Genehmigung der Tierärztlichen Fakultät der Ludwig–Maximilians–Universität München Dekan: Univ. Prof. Dr. Braun Berichterstatter: Univ. Prof. Dr. Dr. habil Förster Korreferent: Univ. Prof. Dr. Mansfeld Tag der Promotion: 12. February 2011 This work is dedicated to My Parents, my wife and my lovely daughters; Sama, Shaza, Hana CONTENTS CONTENTS ABBREVIATION……………………………………………………………… IV CHAPTER 1: GENERAL INTRODUCTION……………………………….. 1 CHAPTER 2: REVIEW OF LITERATURE………………………………… 3 2.1. DNA Markers……………………………………………………….. 3 2.1.1. Microsatellites………………………………………………………….. 3 2.1.2. Single Nucleotide Polymorphism (SNPs)…………………………… 4 2.2. Mapping of Quantitative Trait Loci (QTL)…………………….. 5 2.2.1. QTL Mapping Designs………………………………………………... 6 2.2.1.1. Daughter Design………………………………………………... 6 2.2.1.2. Granddaughter Design………………………………………… 7 2.2.1.3. Complex Pedigree Design…………………………………….. 9 2.2.2. QTL Mapping Strategies……………………………………………… 10 2.2.2.1. Candidate Gene Approach……………………………………. 10 2.2.2.2. Genome Scan Approach……………………………………… 11 2.3. Principles of Linkage Mapping…………………………………. 12 2.4. QTL Fine Mapping………………………………………………… 14 2.4.1. Linkage Disequilibrium……………………………………………… 15 2.4.2. Combined Linkage Disequilibrium and Linkage (LDL) Mapping… 17 2.5. Identification of Candidate Genes……………………………… 18 2.6.
    [Show full text]
  • DNA Methylation Analysis Identifies Patterns in Progressive Glioma Grades to Predict Patient Survival
    International Journal of Molecular Sciences Article DNA Methylation Analysis Identifies Patterns in Progressive Glioma Grades to Predict Patient Survival Jingyin Weng and Nicole Salazar * Department of Biology, San Francisco State University, San Francisco, CA 94132, USA; [email protected] * Correspondence: [email protected] Abstract: DNA methylation is an epigenetic change to the genome that impacts gene activities with- out modification to the DNA sequence. Alteration in the methylation pattern is a naturally occurring event throughout the human life cycle which may result in the development of diseases such as cancer. In this study, we analyzed methylation data from The Cancer Genome Atlas, under the Lower-Grade Glioma (LGG) and Glioblastoma Multiforme (GBM) projects, to identify methylation markers that exhibit unique changes in DNA methylation pattern along with tumor grade progression, to predict patient survival. We found ten glioma grade-associated Cytosine-phosphate-Guanine (CpG) sites that targeted four genes (SMOC1, KCNA4, SLC25A21, and UPP1) and the methylation pattern is strongly associated with glioma specific molecular alterations, primarily isocitrate dehydrogenase (IDH) mutation and chromosome 1p/19q codeletion. The ten CpG sites collectively distinguished a cohort of diffuse glioma patients with remarkably poor survival probability. Our study highlights genes (KCNA4 and SLC25A21) that were not previously associated with gliomas to have contributed to the poorer patient outcome. These CpG sites can aid glioma tumor progression monitoring and serve as prognostic markers to identify patients diagnosed with less aggressive and malignant gliomas that exhibit similar survival probability to GBM patients. Keywords: glioma; glioblastoma; DNA methylation; progression; TCGA; WGCNA; prognosis Citation: Weng, J.; Salazar, N.
    [Show full text]
  • CD29 Identifies IFN-Γ–Producing Human CD8+ T Cells With
    + CD29 identifies IFN-γ–producing human CD8 T cells with an increased cytotoxic potential Benoît P. Nicoleta,b, Aurélie Guislaina,b, Floris P. J. van Alphenc, Raquel Gomez-Eerlandd, Ton N. M. Schumacherd, Maartje van den Biggelaarc,e, and Monika C. Wolkersa,b,1 aDepartment of Hematopoiesis, Sanquin Research, 1066 CX Amsterdam, The Netherlands; bLandsteiner Laboratory, Oncode Institute, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands; cDepartment of Research Facilities, Sanquin Research, 1066 CX Amsterdam, The Netherlands; dDivision of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands; and eDepartment of Molecular and Cellular Haemostasis, Sanquin Research, 1066 CX Amsterdam, The Netherlands Edited by Anjana Rao, La Jolla Institute for Allergy and Immunology, La Jolla, CA, and approved February 12, 2020 (received for review August 12, 2019) Cytotoxic CD8+ T cells can effectively kill target cells by producing therefore developed a protocol that allowed for efficient iso- cytokines, chemokines, and granzymes. Expression of these effector lation of RNA and protein from fluorescence-activated cell molecules is however highly divergent, and tools that identify and sorting (FACS)-sorted fixed T cells after intracellular cytokine + preselect CD8 T cells with a cytotoxic expression profile are lacking. staining. With this top-down approach, we performed an un- + Human CD8 T cells can be divided into IFN-γ– and IL-2–producing biased RNA-sequencing (RNA-seq) and mass spectrometry cells. Unbiased transcriptomics and proteomics analysis on cytokine- γ– – + + (MS) analyses on IFN- and IL-2 producing primary human producing fixed CD8 T cells revealed that IL-2 cells produce helper + + + CD8 Tcells.
    [Show full text]