Mouse Ndufb6 Knockout Project (CRISPR/Cas9)

Total Page:16

File Type:pdf, Size:1020Kb

Mouse Ndufb6 Knockout Project (CRISPR/Cas9) https://www.alphaknockout.com Mouse Ndufb6 Knockout Project (CRISPR/Cas9) Objective: To create a Ndufb6 knockout Mouse model (C57BL/6J) by CRISPR/Cas-mediated genome engineering. Strategy summary: The Ndufb6 gene (NCBI Reference Sequence: NM_001033305 ; Ensembl: ENSMUSG00000071014 ) is located on Mouse chromosome 4. 4 exons are identified, with the ATG start codon in exon 1 and the TGA stop codon in exon 4 (Transcript: ENSMUST00000095128). Exon 1~3 will be selected as target site. Cas9 and gRNA will be co-injected into fertilized eggs for KO Mouse production. The pups will be genotyped by PCR followed by sequencing analysis. Note: Exon 1 starts from about 0.26% of the coding region. Exon 1~3 covers 82.81% of the coding region. The size of effective KO region: ~6526 bp. The KO region does not have any other known gene. Page 1 of 8 https://www.alphaknockout.com Overview of the Targeting Strategy Wildtype allele 5' gRNA region gRNA region 3' 1 2 3 4 Legends Exon of mouse Ndufb6 Knockout region Page 2 of 8 https://www.alphaknockout.com Overview of the Dot Plot (up) Window size: 15 bp Forward Reverse Complement Sequence 12 Note: The 2000 bp section upstream of start codon is aligned with itself to determine if there are tandem repeats. No significant tandem repeat is found in the dot plot matrix. So this region is suitable for PCR screening or sequencing analysis. Overview of the Dot Plot (down) Window size: 15 bp Forward Reverse Complement Sequence 12 Note: The 2000 bp section downstream of Exon 3 is aligned with itself to determine if there are tandem repeats. No significant tandem repeat is found in the dot plot matrix. So this region is suitable for PCR screening or sequencing analysis. Page 3 of 8 https://www.alphaknockout.com Overview of the GC Content Distribution (up) Window size: 300 bp Sequence 12 Summary: Full Length(2000bp) | A(28.65% 573) | C(21.3% 426) | T(28.9% 578) | G(21.15% 423) Note: The 2000 bp section upstream of start codon is analyzed to determine the GC content. No significant high GC-content region is found. So this region is suitable for PCR screening or sequencing analysis. Overview of the GC Content Distribution (down) Window size: 300 bp Sequence 12 Summary: Full Length(2000bp) | A(28.0% 560) | C(18.55% 371) | T(33.65% 673) | G(19.8% 396) Note: The 2000 bp section downstream of Exon 3 is analyzed to determine the GC content. No significant high GC-content region is found. So this region is suitable for PCR screening or sequencing analysis. Page 4 of 8 https://www.alphaknockout.com BLAT Search Results (up) QUERY SCORE START END QSIZE IDENTITY CHROM STRAND START END SPAN ----------------------------------------------------------------------------------------------- browser details YourSeq 2000 1 2000 2000 100.0% chr4 - 40279338 40281337 2000 browser details YourSeq 228 1 327 2000 84.2% chr9 + 53088553 53088864 312 browser details YourSeq 162 265 567 2000 84.7% chr7 + 27621956 27622209 254 browser details YourSeq 153 384 646 2000 85.7% chrX + 142382416 142382623 208 browser details YourSeq 153 384 586 2000 87.0% chr2 + 170202929 170203127 199 browser details YourSeq 150 384 568 2000 91.4% chr5 - 92175594 92175776 183 browser details YourSeq 150 331 568 2000 90.7% chr12 - 69369654 69370208 555 browser details YourSeq 149 384 568 2000 91.8% chr16 - 11265926 11266113 188 browser details YourSeq 148 12 555 2000 84.8% chr19 - 7465133 7465533 401 browser details YourSeq 147 386 568 2000 91.2% chr9 + 118367160 118367345 186 browser details YourSeq 147 384 568 2000 90.6% chr8 + 123286696 123286881 186 browser details YourSeq 147 379 585 2000 88.5% chr8 + 10981389 10981593 205 browser details YourSeq 145 381 567 2000 90.5% chrX - 140425083 140425271 189 browser details YourSeq 145 384 568 2000 91.1% chr4 + 133658536 133658726 191 browser details YourSeq 145 384 586 2000 87.5% chr17 + 31120794 31120990 197 browser details YourSeq 145 384 568 2000 89.9% chr15 + 99482876 99483059 184 browser details YourSeq 145 386 568 2000 89.1% chr14 + 105954395 105954574 180 browser details YourSeq 145 384 565 2000 88.9% chr11 + 86446580 86446755 176 browser details YourSeq 144 384 566 2000 87.8% chr5 - 140537375 140537547 173 browser details YourSeq 144 384 567 2000 88.6% chr2 - 117445291 117445471 181 Note: The 2000 bp section upstream of start codon is BLAT searched against the genome. No significant similarity is found. BLAT Search Results (down) QUERY SCORE START END QSIZE IDENTITY CHROM STRAND START END SPAN ----------------------------------------------------------------------------------------------- browser details YourSeq 2000 1 2000 2000 100.0% chr4 - 40270810 40272809 2000 browser details YourSeq 201 928 1136 2000 99.6% chr9 + 108037651 108037864 214 browser details YourSeq 200 895 1122 2000 94.8% chr8 - 57734341 57734557 217 browser details YourSeq 197 929 1136 2000 99.5% chr18 + 34575681 34575892 212 browser details YourSeq 195 918 1124 2000 97.6% chr13 - 100525606 100525832 227 browser details YourSeq 195 927 1136 2000 96.7% chr12 - 84492588 84492803 216 browser details YourSeq 194 930 1136 2000 98.5% chr11 + 98876174 98876381 208 browser details YourSeq 192 932 1140 2000 98.5% chr2 - 75690375 75690591 217 browser details YourSeq 192 932 1124 2000 100.0% chr9 + 59376160 59376354 195 browser details YourSeq 192 932 1131 2000 98.5% chr5 + 100283909 100284108 200 browser details YourSeq 191 932 1127 2000 99.0% chr11 - 102389647 102389844 198 browser details YourSeq 191 930 1123 2000 99.5% chr4 + 45863866 45864060 195 browser details YourSeq 191 932 1127 2000 99.0% chr18 + 12745273 12745470 198 browser details YourSeq 190 932 1123 2000 99.5% chr8 - 71501382 71501573 192 browser details YourSeq 190 930 1122 2000 99.5% chr2 - 172968395 172968602 208 browser details YourSeq 190 923 1121 2000 98.0% chr14 - 44836771 44837018 248 browser details YourSeq 190 928 1121 2000 99.5% chr10 - 78478675 78478876 202 browser details YourSeq 190 930 1122 2000 99.5% chrX + 152557797 152558006 210 browser details YourSeq 190 930 1122 2000 99.5% chr5 + 31745266 31745463 198 browser details YourSeq 190 930 1122 2000 99.5% chr10 + 120936070 120936271 202 Note: The 2000 bp section downstream of Exon 3 is BLAT searched against the genome. No significant similarity is found. Page 5 of 8 https://www.alphaknockout.com Gene and protein information: Ndufb6 NADH:ubiquinone oxidoreductase subunit B6 [ Mus musculus (house mouse) ] Gene ID: 230075, updated on 12-Aug-2019 Gene summary Official Symbol Ndufb6 provided by MGI Official Full Name NADH:ubiquinone oxidoreductase subunit B6 provided by MGI Primary source MGI:MGI:2684983 See related Ensembl:ENSMUSG00000071014 Gene type protein coding RefSeq status REVIEWED Organism Mus musculus Lineage Eukaryota; Metazoa; Chordata; Craniata; Vertebrata; Euteleostomi; Mammalia; Eutheria; Euarchontoglires; Glires; Rodentia; Myomorpha; Muroidea; Muridae; Murinae; Mus; Mus Also known as Gm137; CI-B17 Summary This gene encodes a subunit of complex I (NADH:ubiquinone oxidoreductase) of the mitochondrial respiratory chain. This Expression complex functions in electron transport and generation of a proton gradient across the inner mitochondrial membrane to drive ATP synthesis. Data from human cell lines suggests that the encoded subunit may be required for electron transfer activity. [provided by RefSeq, Aug 2015] Orthologs Ubiquitous expression in heart adult (RPKM 89.9), kidney adult (RPKM 75.1) and 28 other tissues See more human all Genomic context Location: 4; 4 A5 See Ndufb6 in Genome Data Viewer Exon count: 4 Annotation release Status Assembly Chr Location 108 current GRCm38.p6 (GCF_000001635.26) 4 NC_000070.6 (40270591..40279421, complement) Build 37.2 previous assembly MGSCv37 (GCF_000001635.18) 4 NC_000070.5 (40217696..40226401, complement) Chromosome 4 - NC_000070.6 Page 6 of 8 https://www.alphaknockout.com Transcript information: This gene has 2 transcripts Gene: Ndufb6 ENSMUSG00000071014 Description NADH:ubiquinone oxidoreductase subunit B6 [Source:MGI Symbol;Acc:MGI:2684983] Gene Synonyms LOC230075 Location Chromosome 4: 40,270,591-40,279,421 reverse strand. GRCm38:CM000997.2 About this gene This gene has 2 transcripts (splice variants), 194 orthologues and is a member of 1 Ensembl protein family. Transcripts Name Transcript ID bp Protein Translation ID Biotype CCDS UniProt Flags Ndufb6-201 ENSMUST00000095128.9 659 128aa ENSMUSP00000092746.3 Protein coding CCDS18045 A2AP31 Q3UIU2 TSL:1 GENCODE basic APPRIS P1 Ndufb6-202 ENSMUST00000108108.2 480 97aa ENSMUSP00000103743.2 Protein coding - A2AP32 TSL:3 GENCODE basic 28.83 kb Forward strand 40.265Mb 40.270Mb 40.275Mb 40.280Mb 40.285Mb Genes Smim27-201 >protein coding (Comprehensive set... Smim27-202 >lncRNA Contigs AL831793.4 > Genes (Comprehensive set... < Topors-201protein coding < Ndufb6-201protein coding < Ndufb6-202protein coding Regulatory Build 40.265Mb 40.270Mb 40.275Mb 40.280Mb 40.285Mb Reverse strand 28.83 kb Regulation Legend CTCF Promoter Promoter Flank Transcription Factor Binding Site Gene Legend Protein Coding merged Ensembl/Havana Ensembl protein coding Non-Protein Coding RNA gene Page 7 of 8 https://www.alphaknockout.com Transcript: ENSMUST00000095128 < Ndufb6-201protein coding Reverse strand 8.83 kb ENSMUSP00000092... Transmembrane heli... PDB-ENSP mappings Low complexity (Seg) Pfam NADH dehydrogenase 1, beta subcomplex, subunit 6 PANTHER NADH dehydrogenase 1, beta subcomplex, subunit 6 All sequence SNPs/i... Sequence variants (dbSNP and all other sources) Variant Legend missense variant synonymous variant Scale bar 0 20 40 60 80 100 128 We wish to acknowledge the following valuable scientific information resources: Ensembl, MGI, NCBI, UCSC. Page 8 of 8.
Recommended publications
  • The Pathogenetic Role of Β-Cell Mitochondria in Type 2 Diabetes
    236 3 Journal of M Fex et al. Mitochondria in β-cells 236:3 R145–R159 Endocrinology REVIEW The pathogenetic role of β-cell mitochondria in type 2 diabetes Malin Fex1, Lisa M Nicholas1, Neelanjan Vishnu1, Anya Medina1, Vladimir V Sharoyko1, David G Nicholls1, Peter Spégel1,2 and Hindrik Mulder1 1Department of Clinical Sciences in Malmö, Unit of Molecular Metabolism, Lund University Diabetes Centre, Clinical Research Center, Malmö University Hospital, Lund University, Malmö, Sweden 2Department of Chemistry, Center for Analysis and Synthesis, Lund University, Sweden Correspondence should be addressed to H Mulder: [email protected] Abstract Mitochondrial metabolism is a major determinant of insulin secretion from pancreatic Key Words β-cells. Type 2 diabetes evolves when β-cells fail to release appropriate amounts f TCA cycle of insulin in response to glucose. This results in hyperglycemia and metabolic f coupling signal dysregulation. Evidence has recently been mounting that mitochondrial dysfunction f oxidative phosphorylation plays an important role in these processes. Monogenic dysfunction of mitochondria is a f mitochondrial transcription rare condition but causes a type 2 diabetes-like syndrome owing to β-cell failure. Here, f genetic variation we describe novel advances in research on mitochondrial dysfunction in the β-cell in type 2 diabetes, with a focus on human studies. Relevant studies in animal and cell models of the disease are described. Transcriptional and translational regulation in mitochondria are particularly emphasized. The role of metabolic enzymes and pathways and their impact on β-cell function in type 2 diabetes pathophysiology are discussed. The role of genetic variation in mitochondrial function leading to type 2 diabetes is highlighted.
    [Show full text]
  • SARS-Cov-2 Restructures the Host Chromatin Architecture 2
    bioRxiv preprint doi: https://doi.org/10.1101/2021.07.20.453146; this version posted July 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 1 SARS-CoV-2 Restructures the Host Chromatin Architecture 2 3 Authors: Ruoyu Wang 1,2,†, Joo-Hyung Lee 1,†, Feng Xiong 1, Jieun Kim 3,4, Lana Al Hasani 1,2, Xiaoyi 4 Yuan 3,4, Pooja Shivshankar 1,3,4, Joanna Krakowiak 1, Chuangye Qi 1, Yanyu Wang 3,4, Holger K. 5 Eltzschig 2,3,4, Wenbo Li 1,2,* 6 7 Affiliations: 8 1 Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas 9 Health Science Center, Houston, 77030, TX, USA 10 2 Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center and 11 UTHealth, Houston, 77030, TX, USA 12 3 Department of Anesthesiology, McGovern Medical School, University of Texas Health Science Center, 13 Houston, 77030, TX, USA 14 4 Center for Perioperative Medicine, McGovern Medical School, University of Texas Health Science 15 Center, Houston, 77030, TX, USA 16 17 † These authors contributed equally 18 * Correspondence: Wenbo Li, Ph.D. ([email protected]). MSB 6.161, Fannin Street, Houston, 19 Texas, 77030, USA; Tel: 713-500-6103. 1 bioRxiv preprint doi: https://doi.org/10.1101/2021.07.20.453146; this version posted July 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity.
    [Show full text]
  • NDUFB6 (NM 182739) Human Tagged ORF Clone Product Data
    OriGene Technologies, Inc. 9620 Medical Center Drive, Ste 200 Rockville, MD 20850, US Phone: +1-888-267-4436 [email protected] EU: [email protected] CN: [email protected] Product datasheet for RC219791 NDUFB6 (NM_182739) Human Tagged ORF Clone Product data: Product Type: Expression Plasmids Product Name: NDUFB6 (NM_182739) Human Tagged ORF Clone Tag: Myc-DDK Symbol: NDUFB6 Synonyms: B17; CI Vector: pCMV6-Entry (PS100001) E. coli Selection: Kanamycin (25 ug/mL) Cell Selection: Neomycin ORF Nucleotide >RC219791 representing NM_182739 Sequence: Red=Cloning site Blue=ORF Green=Tags(s) TTTTGTAATACGACTCACTATAGGGCGGCCGGGAATTCGTCGACTGGATCCGGTACCGAGGAGATCTGCC GCCGCGATCGCC ATGACGGGGTACACTCCGGATGAGAAACTGCGGCTGCAGCAGCTGCGAGAGCTGAGAAGGCGATGGCTGA AGGACCAGGAGCTGAGCCCTCGGGAGCCGGTGCTGCCCCCACAGAAGATGGGGCCTATGGAGAAATTCTG GAATAAATTTTTGGAGAATAAATCCCCTTGGAGGAAAATGGTCCATGGGGTATACAAAAAGAGTATCTTT GTTTTCACTCATGTACTTGTACCTGTCTGGATTATTCATTATTACATGAAGTATCATGTTTCTGGTGATA CAATTCTGGAGACTGGAGAAGTAATTCCACCAATGAAAGAATTTCCTGATCAACATCAT ACGCGTACGCGGCCGCTCGAGCAGAAACTCATCTCAGAAGAGGATCTGGCAGCAAATGATATCCTGGATT ACAAGGATGACGACGATAAGGTTTAA Protein Sequence: >RC219791 representing NM_182739 Red=Cloning site Green=Tags(s) MTGYTPDEKLRLQQLRELRRRWLKDQELSPREPVLPPQKMGPMEKFWNKFLENKSPWRKMVHGVYKKSIF VFTHVLVPVWIIHYYMKYHVSGDTILETGEVIPPMKEFPDQHH TRTRPLEQKLISEEDLAANDILDYKDDDDKV Restriction Sites: SgfI-MluI This product is to be used for laboratory only. Not for diagnostic or therapeutic use. View online » ©2021 OriGene Technologies, Inc., 9620 Medical Center Drive, Ste 200, Rockville,
    [Show full text]
  • THE FUNCTIONAL SIGNIFICANCE of MITOCHONDRIAL SUPERCOMPLEXES in C. ELEGANS by WICHIT SUTHAMMARAK Submitted in Partial Fulfillment
    THE FUNCTIONAL SIGNIFICANCE OF MITOCHONDRIAL SUPERCOMPLEXES in C. ELEGANS by WICHIT SUTHAMMARAK Submitted in partial fulfillment of the requirements For the degree of Doctor of Philosophy Dissertation Advisor: Drs. Margaret M. Sedensky & Philip G. Morgan Department of Genetics CASE WESTERN RESERVE UNIVERSITY January, 2011 CASE WESTERN RESERVE UNIVERSITY SCHOOL OF GRADUATE STUDIES We hereby approve the thesis/dissertation of _____________________________________________________ candidate for the ______________________degree *. (signed)_______________________________________________ (chair of the committee) ________________________________________________ ________________________________________________ ________________________________________________ ________________________________________________ ________________________________________________ (date) _______________________ *We also certify that written approval has been obtained for any proprietary material contained therein. Dedicated to my family, my teachers and all of my beloved ones for their love and support ii ACKNOWLEDGEMENTS My advanced academic journey began 5 years ago on the opposite side of the world. I traveled to the United States from Thailand in search of a better understanding of science so that one day I can return to my homeland and apply the knowledge and experience I have gained to improve the lives of those affected by sickness and disease yet unanswered by science. Ultimately, I hoped to make the academic transition into the scholarly community by proving myself through scientific research and understanding so that I can make a meaningful contribution to both the scientific and medical communities. The following dissertation would not have been possible without the help, support, and guidance of a lot of people both near and far. I wish to thank all who have aided me in one way or another on this long yet rewarding journey. My sincerest thanks and appreciation goes to my advisors Philip Morgan and Margaret Sedensky.
    [Show full text]
  • Short-Term Exercise Training Does Not Stimulate Skeletal Muscle ATP Synthesis in Relatives of Humans with Type 2 Diabetes
    Diabetes Publish Ahead of Print, published online March 5, 2009 Short-term exercise training does not stimulate skeletal muscle ATP synthesis in relatives of humans with type 2 diabetes Gertrud Kacerovsky-Bielesz1,2, Marek Chmelik2,3, Charlotte Ling4, Rochus Pokan5, Julia Szendroedi1,2, Michaela Farukuoye2, Michaela Kacerovsky2, Albrecht I. Schmid2,3, Stephan Gruber3, Michael Wolzt6, Ewald Moser3, Giovanni Pacini7, Gerhard Smekal5, Leif Groop4, Michael Roden1,2,8 1 1. Medical Department, Hanusch Hospital, Vienna, Austria, 2 Karl-Landsteiner Institute for Endocrinology and Metabolism, Vienna, Austria, 3 MR Center of Excellence, Medical University of Vienna, Austria, 4 Department of Clinical Sciences, Lund University, Malmö, Sweden, 5 Department of Sports and Exercise Physiology, University of Vienna, Austria, 6 Department of Clinical Pharmacology, Medical University of Vienna, Austria, 7 Metabolic Unit, Institute of Biomedical Engineering, CNR, Padova, Italy 8 Institute for Clinical Diabetology, German Diabetes Center - Leibniz Center for Diabetes Research, Department of Medicine/Metabolic Diseases, Heinrich Heine University Düsseldorf, Düsseldorf, Germany Corresponding Author: Michael Roden, MD Email: [email protected] Clinical trial reg. No. NCT00710008, clinicaltrials.gov. Additional information for this article can be found in an online appendix at http://diabetes.diabetesjournals.org Submitted 6 September 2008 and accepted 27 February 2009. This is an uncopyedited electronic version of an article accepted for publication in Diabetes. The American Diabetes Association, publisher of Diabetes, is not responsible for any errors or omissions in this version of the manuscript or any version derived from it by third parties. The definitive publisher-authenticated version will be available in a future issue of Diabetes in print and online at http://diabetes.diabetesjournals.org.
    [Show full text]
  • Biogenesis of NDUFS3-Less Complex I Indicates TMEM126A/OPA7 As an Assembly Factor of the ND4-Module
    bioRxiv preprint doi: https://doi.org/10.1101/2020.10.22.350587; this version posted October 23, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC 4.0 International license. Biogenesis of NDUFS3-less complex I indicates TMEM126A/OPA7 as an assembly factor of the ND4-module Running title: NDUFS3-dependent CI disassembly pathway Luigi D’Angelo,1* Elisa Astro,1* Monica De Luise,2 Ivana Kurelac,2 Nikkitha Umesh-Ganesh,2 Shujing Ding,3 Ian M. Fearnley,3 Massimo Zeviani,3,4 Giuseppe Gasparre,2,5 Anna Maria Porcelli,1,6# Erika Fernandez-Vizarra,3,7# and Luisa Iommarini1# 1Department of Pharmacy and Biotechnology (FABIT), University of Bologna, Bologna, Italy 2Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy 3Medical Research Council-Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK 4Venetian Institute of Molecular Medicine, Via Orus 2, 35128 Padova, Italy; Department of Neurosciences, University of Padova, via Giustiniani 2, 35128 Padova, Italy 5Center for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy 6Interdepartmental Center of Industrial Research (CIRI) Life Science and Health Technologies, University of Bologna, Ozzano dell'Emilia, Italy 7Institute of Molecular, Cell and Systems Biology, University of Glasgow, Glasgow, UK. *Co-first authors #Co-last authors To whom correspondence should be addressed: Luisa Iommarini Department of Pharmacy and Biotechnology (FABIT) University of Bologna Via Francesco Selmi 3, 40126 Bologna, Italy Tel. +39 051 2091282 e-mail [email protected] Erika Fernandez-Vizarra Institute of Molecular, Cell and Systems Biology University of Glasgow University Avenue Glasgow G12 8QQ, UK Tel.
    [Show full text]
  • Metabolic Programming of MEST DNA Methylation by Intrauterine Exposure to Gestational Diabetes Mellitus
    ORIGINAL ARTICLE Metabolic Programming of MEST DNA Methylation by Intrauterine Exposure to Gestational Diabetes Mellitus Nady El Hajj,1 Galyna Pliushch,1 Eberhard Schneider,1 Marcus Dittrich,2 Tobias Müller,2 Michael Korenkov,3 Melanie Aretz,4 Ulrich Zechner,5 Harald Lehnen,4 and Thomas Haaf1 Epigenetic processes are primary candidates when searching for The prevalence of type 2 diabetes mellitus and obesity is mechanisms that can stably modulate gene expression and met- increasing in most populations worldwide. In addition to abolic pathways according to early life conditions. To test the overnutrition and physical inactivity, prenatal factors ap- effects of gestational diabetes mellitus (GDM) on the epigenome pear to make a significant contribution to the metabolic of the next generation, cord blood and placenta tissue were disease epidemic. It is well-known that the offspring of obtained from 88 newborns of mothers with dietetically treated diabetic and/or obese mothers who are exposed to high GDM, 98 with insulin-dependent GDM, and 65 without GDM. fi concentrations of glucose, free fatty acids, and amino Bisul te pyrosequencing was used to compare the methylation acids in utero are at increased life-long risk for de- levels of seven imprinted genes involved in prenatal and post- natal growth, four genes involved in energy metabolism, one anti- velopment of metabolic disorders (6,7). Gestational di- inflammatory gene, one tumor suppressor gene, one pluripotency abetes mellitus (GDM) presents during pregnancy in gene, and two repetitive DNA families. The maternally imprinted women without pregestational diabetes and typically dis- MEST gene, the nonimprinted glucocorticoid receptor NR3C1 appears again after delivery.
    [Show full text]
  • Mitochondrial Oxidative Phosphorylation Defect in the Heart
    www.nature.com/scientificreports OPEN Mitochondrial Oxidative Phosphorylation defect in the Heart of Subjects with Coronary Artery Received: 23 August 2018 Accepted: 6 December 2018 Disease Published: xx xx xxxx Karima Ait-Aissa 1, Scott C. Blaszak1, Gisela Beutner2, Shirng-Wern Tsaih3, Garrett Morgan4, Janine H. Santos5, Michael J. Flister3, David L. Joyce6, Amadou K. S. Camara3,7, David D. Gutterman1, Anthony J. Donato4,8, George A. Porter Jr. 2,9,10 & Andreas M. Beyer1,3 Coronary artery disease (CAD) is a leading cause of death worldwide and frequently associated with mitochondrial dysfunction. Detailed understanding of abnormalities in mitochondrial function that occur in patients with CAD is lacking. We evaluated mitochondrial damage, energy production, and mitochondrial complex activity in human non-CAD and CAD hearts. Fresh and frozen human heart tissue was used. Cell lysate or mitochondria were isolated using standard techniques. Mitochondrial DNA (mtDNA), NAD + and ATP levels, and mitochondrial oxidative phosphorylation capacity were evaluated. Proteins critical to the regulation of mitochondrial metabolism and function were also evaluated in tissue lysates. PCR analysis revealed an increase in mtDNA lesions and the frequency of mitochondrial common deletion, both established markers for impaired mitochondrial integrity in CAD compared to non-CAD patient samples. NAD+ and ATP levels were signifcantly decreased in CAD subjects compared to Non-CAD (NAD+ fold change: non-CAD 1.00 ± 0.17 vs. CAD 0.32 ± 0.12* and ATP fold change: non-CAD 1.00 ± 0.294 vs. CAD 0.01 ± 0.001*; N = 15, P < 0.005). We observed decreased respiration control index in CAD tissue and decreased activity of complexes I, II, and III.
    [Show full text]
  • Download Download
    ISSN: 2469-6706 Vol. 6 2019 Role of epigenetics in the pathogenesis and management of type 2 diabetes mellitus Tajudeen Yahaya a , 1 Esther Oladele b, Ufuoma Shemishere c and Mohammad Abdulrau’f c aDepartment of Biology, Federal University Birnin Kebbi, Nigeria,bBiology Unit, Distance Learning Institute, University of Lagos, Nigeria, and cDepartment of Biochem- istry and Molecular Biology, Federal University Birnin Kebbi, Nigeria The need to reverse the growing incidence and burden of dia- ence the risk of T2DM, most of the candidate genes impair insulin betes mellitus (DM) worldwide has led to more studies on the synthesis rather than insulin metabolism (12, 13). This suggests that causes of the disease. Scientists have long suspected genetic pancreatic islet developmental error might be the main mechanism and environmental factors in the pathogenesis of type 2 diabetes of T2DM pathogenesis (12, 14). The suspect genes do not account mellitus (T2DM). However, recent studies suggest that epigenetic for the full transmission of T2DM, meaning that more genetic as- changes may cause some cases of the disease. This review high- lights the role of epigenetic modifications in the pathogenesis pect exists (12, 15). The search for this additional genetic factor led and management of T2DM. Peer-reviewed studies on the sub- to the discovery that modifications of the chemical tags above the ject were retrieved from electronic databases such as PubMed, genome, often known as epigenetic change, may modulate T2DM Google Scholar, SpringerLink, and Scopus. Most of the studies (1). implicated epigenetic modifications in the pathogenesis of some cases of T2DM. DNA methylation, histone modification, and mi- Epigenetic changes are heritable modifications in gene expres- croRNAs mediated pathways are the main mechanisms of epi- sion and function without affecting the nucleotide sequence (16, genetic changes.
    [Show full text]
  • Instability in NAD Metabolism Leads to Impaired Cardiac Mitochondrial
    RESEARCH ARTICLE Instability in NAD+ metabolism leads to impaired cardiac mitochondrial function and communication Knut H Lauritzen1*, Maria Belland Olsen1, Mohammed Shakil Ahmed2, Kuan Yang1, Johanne Egge Rinholm3, Linda H Bergersen4,5, Qin Ying Esbensen6, Lars Jansen Sverkeli7, Mathias Ziegler7, Ha˚ vard Attramadal2, Bente Halvorsen1,8, Pa˚ l Aukrust1,8,9, Arne Yndestad1,8 1Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet and University of Oslo, Oslo, Norway; 2Institute for Surgical Research, Oslo University Hospital and University of Oslo, Oslo, Norway; 3Department of Microbiology, Oslo University Hospital, Oslo, Norway; 4Department of Oral Biology, University of Oslo, Oslo, Norway; 5Department of Neuroscience and Pharmacology, Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark; 6Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, Nordbyhagen, Norway; 7Department of Biomedicine, University of Bergen, Bergen, Norway; 8Institute of Clinical Medicine, University of Oslo, Faculty of Medicine, Oslo, Norway; 9Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway Abstract Poly(ADP-ribose) polymerase (PARP) enzymes initiate (mt)DNA repair mechanisms and use nicotinamide adenine dinucleotide (NAD+) as energy source. Prolonged PARP activity can drain cellular NAD+ reserves, leading to de-regulation of important molecular processes. Here, we provide evidence of a pathophysiological mechanism that connects mtDNA damage to cardiac *For correspondence: dysfunction via reduced NAD+ levels and loss of mitochondrial function and communication. Using Knut.Huso.Lauritzen@rr-research. a transgenic model, we demonstrate that high levels of mice cardiomyocyte mtDNA damage cause no a reduction in NAD+ levels due to extreme DNA repair activity, causing impaired activation of Competing interests: The NAD+-dependent SIRT3.
    [Show full text]
  • Bayesian Hidden Markov Tree Models for Clustering Genes with Shared Evolutionary History
    Submitted to the Annals of Applied Statistics arXiv: arXiv:0000.0000 BAYESIAN HIDDEN MARKOV TREE MODELS FOR CLUSTERING GENES WITH SHARED EVOLUTIONARY HISTORY By Yang Liy,{,∗, Shaoyang Ningy,∗, Sarah E. Calvoz,x,{, Vamsi K. Moothak,z,x,{ and Jun S. Liuy Harvard Universityy, Broad Institutez, Harvard Medical Schoolx, Massachusetts General Hospital{, and Howard Hughes Medical Institutek Determination of functions for poorly characterized genes is cru- cial for understanding biological processes and studying human dis- eases. Functionally associated genes are often gained and lost together through evolution. Therefore identifying co-evolution of genes can predict functional gene-gene associations. We describe here the full statistical model and computational strategies underlying the orig- inal algorithm CLustering by Inferred Models of Evolution (CLIME 1.0) recently reported by us [Li et al., 2014]. CLIME 1.0 employs a mixture of tree-structured hidden Markov models for gene evolution process, and a Bayesian model-based clustering algorithm to detect gene modules with shared evolutionary histories (termed evolutionary conserved modules, or ECMs). A Dirichlet process prior was adopted for estimating the number of gene clusters and a Gibbs sampler was developed for posterior sampling. We further developed an extended version, CLIME 1.1, to incorporate the uncertainty on the evolution- ary tree structure. By simulation studies and benchmarks on real data sets, we show that CLIME 1.0 and CLIME 1.1 outperform traditional methods that use simple metrics (e.g., the Hamming distance or Pear- son correlation) to measure co-evolution between pairs of genes. 1. Introduction. The human genome encodes more than 20,000 protein- coding genes, of which a large fraction do not have annotated function to date [Galperin and Koonin, 2010].
    [Show full text]
  • Mitochondrial H2O2 Generated from Electron Transport Chain Complex I Stimulates Muscle Differentiation
    Cell Research (2011) 21:817-834. © 2011 IBCB, SIBS, CAS All rights reserved 1001-0602/11 $ 32.00 npg ORIGINAL ARTICLE www.nature.com/cr Mitochondrial H2O2 generated from electron transport chain complex I stimulates muscle differentiation Seonmin Lee1, Eunyoung Tak1, Jisun Lee1, MA Rashid1, Michael P Murphy2, Joohun Ha1, Sung Soo Kim1 1Department of Biochemistry and Molecular Biology, Medical Science and Engineering Research Center for Bioreaction to Reac- tive Oxygen Species and Biomedical Science Institute (BK-21), School of Medicine, Kyung Hee University, #1, Hoegi-dong, Dong- daemoon-gu, Seoul 130-701, Korea; 2MRC Mitochondrial Biology Unit, Hills Road, Cambridge CB2 0XY, UK Mitochondrial reactive oxygen species (mROS) have been considered detrimental to cells. However, their physi- ological roles as signaling mediators have not been thoroughly explored. Here, we investigated whether mROS gener- ated from mitochondrial electron transport chain (mETC) complex I stimulated muscle differentiation. Our results showed that the quantity of mROS was increased and that manganese superoxide dismutase (MnSOD) was induced via NF-κB activation during muscle differentiation. Mitochondria-targeted antioxidants (MitoQ and MitoTEMPOL) and mitochondria-targeted catalase decreased mROS quantity and suppressed muscle differentiation without af- fecting the amount of ATP. Mitochondrial alterations, including the induction of mitochondrial transcription factor A and an increase in the number and size of mitochondria, and functional activations were observed during muscle differentiation. In particular, increased expression levels of mETC complex I subunits and a higher activity of com- plex I than other complexes were observed. Rotenone, an inhibitor of mETC complex I, decreased the mitochondrial NADH/NAD+ ratio and mROS levels during muscle differentiation.
    [Show full text]