Extensive Additivity of Gene Expression Differentiates

Total Page:16

File Type:pdf, Size:1020Kb

Extensive Additivity of Gene Expression Differentiates Genetics: Published Articles Ahead of Print, published on October 18, 2007 as 10.1534/genetics.107.076190 Extensive additivity of gene expression differentiates subspecies of the house mouse Ruth Rottscheidt & Bettina Harr Institute for Genetics, University of Cologne, Zülpicher Strasse 47, 50674 Cologne, Germany Address for correspondence: Dr. Bettina Harr, Institute for Genetics, University of Cologne, Zülpicher Strasse 47, 50674 Cologne, Germany Email: [email protected] Tel: ++49 221 470 6617 Fax: ++49 221 470 5975 Keywords: house mouse, gene expression, F1 hybrid, speciation Running title: Additivity of gene expression Abstract We have studied different subspecies of the house mouse and their reciprocal F1 hybrids to estimate the within-locus mode of inheritance for subspecies differences in gene expression in three tissues (brain, liver, testis) of male mice. This study investigates mode of inheritance in crosses at a larger taxonomic distance than have been previously systematically investigated. We found the vast majority of transcripts to be additively expressed with only a few transcripts showing dominance or overdominance in expression, except for one direction of one cross, which showed large mis-expression in the testis. We suggest that as time passes, more genes come to influence expression, and if there is no directional dominance, additivity becomes increasingly more likely, up to a threshold beyond which there is F1 hybrid breakdown. Some previous studies on different organisms have found a large degree of dominance, commonly at shorter taxonomic differences. We surveyed these findings and show that the most consistent association exists between the amount of additivity detected in a study and expression analysis method (in particular microarray platform), suggesting that at least some of the differences among studies might be methodological. Most studies agree with ours in that within-locus additivity seems to be general mode of inheritance for transcript expression. Differentially expressed transcripts identified in our screen among subspecies of house mice are candidate genes that could be involved in reproductive isolation between these subspecies. Introduction The methods of quantitative genetics are now being applied to transcript abundance, as measured using microarrays. “Expression Quantitative Trait Locus” mapping (eQTL) identifies the genetic basis of expression differences between organisms (ROCKMAN and KRUGLYAK 2006). Such mapping studies are being used to ask how many loci contribute to a quantitative trait, what is the size of the effect of the loci, and how individual loci interact to generate a quantitative trait (MACKAY 2001). Treating gene expression levels as measured by microarrays as quantitative traits enables the simultaneous assessment of thousands of traits in parallel. eQTL studies can potentially also be used to ask whether regulatory variants, especially those that are important in evolution, are located in cis (i.e. directly linked to the gene that shows the differential expression) or in trans (i.e. somewhere else in the genome) (HOEKSTRA and COYNE 2007; WRAY 2007). Two approaches have been used to study the genetics of expression traits. In the first, DNA sequence polymorphisms are correlated with expression differences (BREM et al. 2002; DOSS et al. 2005; SCHADT et al. 2003; STOREY et al. 2005). A key finding from these studies is that expression traits are often affected by multiple underlying loci and interactions among them. The second approach makes use of F1 hybrids generated from a cross between divergent taxa to ask if a transcript’s expression is intermediate (“additive”) to that of the two parents. These studies have produced varying results. GIBSON et al. (2004) crossed two lines of Drosophila melanogaster and found most transcripts to be “nonadditivly” (i.e. the hybrids showed expression levels that were more closely to either on of the parents) expressed in F1 hybrids. Widespread nonadditivity was also identified in the Pacific Oyster (HEDGECOCK et al. 2007) and to a lesser extend in Arabidopsis (VUYLSTEKE et al. 2005). However, later studies on Drosophila (HUGHES et al. 2006), maize (STUPAR and SPRINGER 2006; SWANSON-WAGNER et al. 2006), and laboratory strains of the house mouse (CUI et al. 2006) were consistent with mostly additivity of expression. HUGHES et al. (2006) suggested the discrepancy between the different Drosophila studies might be explained in two ways. First, inbreeding might affect the results. Much within-locus additivity was observed using natural populations of D. melanogaster (HUGHES et al. 2006) while nonadditivity was observed with strongly inbred lines (GIBSON et al. 2004). It is well known that crosses between inbred strains produce offspring that exhibit greater biomass, speed of development, and fertility than both parents (heterosis) (COMINGS and MACMURRAY 2000). Heterosis is attributed to overdominance (superiority of heterozygotes at genes affecting fitness) or dominance (masking of recessive deleterious mutations) and plausibly could result in overdominant or dominant expression at the transcript level. However the inbreeding explanation seems to be an unlikely general explanation because additional recent studies have found strong evidence for additivity despite the use of inbred parental lines (CUI et al. 2006; STUPAR and SPRINGER 2006; SWANSON-WAGNER et al. 2006). A second explanation for the Drosophila differences is that genetic architecture of an expression trait might depend critically on the taxonomic level at which the variation is investigated (i.e., between populations as in the GIBSON et al. (2004) study as opposed to within populations as in the HUGHES et al. (2006) study). These authors assume that the more highly diverged the parents are the greater the nonadditivity in gene expression that will be observed. However, we might expect additivity of expression to become more likely as taxa diverge and the trait becomes influenced by more and more genes, provided that they do not show directional dominance. This would apply up to some certain threshold, beyond which incompatible genes lead to massive mis-expression in F1 hybrids. Here we investigate patterns of expression in the house mouse subspecies complex of Mus musculus. We used parental mouse strains that were collected in the wild and inbred in the laboratory for several generations. The strains belong to the three different subspecies of house mice, M. m. musculus, M. m. domesticus and M. m. castaneus, with estimated divergence times of between 300,000 to 1 Million years (BOURSOT et al. 1993). Previous systematic studies focused on comparisons within a single species or used house mouse laboratory strains. In contrast, our study employs natural-derived lines that are rather highly diverged. We found predominantly additive gene expression differences between these subspecies. Materials and Methods Mouse strains Wild-derived strains of domesticus (JPC 2705, from Germany), musculus (JPC 2821, from the Czech Republic) and castaneus (CIM, from India) were used for the microarray experiment. The domesticus and musculus strains were provided by J. Pialek. These strains were collected in the wild and inbred by brother-sister matings for ~13 generations in the laboratory of J. Pialek in the Department of Population Biology in Studenec, Czech Republic. The castaneus strain was provided by A. Orth and F. Bonhomme. This strain has been kept in the Laboratory Génome, Populations, Interactions, Adaptation, Montpellier, France for more than 30 generations in a closed colony. We set up reciprocal crosses between domesticus and musculus and between musculus and castaneus to obtain F1 hybrids. For each parental strain and each reciprocal cross two male individuals were analyzed. Altogether, we performed 42 microarray experiments, 14 in each of three tissues (brain, liver and testis). An overview of the samples used in this study is given in Table 1. All mice were raised under identical standard laboratory conditions and were sacrificed at the age of 8-10 weeks. RNA extraction We extracted RNA from three different tissues (brain, liver and testis) using Trizol® (Invitrogen, Carlsbad, CA) following the manufacturer’s protocol. Quality and integrity of the total RNA was controlled by using the Agilent Technologies 2100 Bioanalyzer and the RNA 6000 Nano LabChip® Kit (Agilent Technologies; Waldbronn, Germany). Microarray Expression profiles were determined for over 39,000 mouse transcripts using the Mouse Genome 430 2.0 Affymetrix® GeneChip ®. For biotin-labeled target synthesis starting from 3 µg of total RNA we used standard protocols supplied by the manufacturer (Affymetrix; Santa Clara, CA). After hybridization the GeneChips ® were washed, stained with SA-PE and read using an Affymetrix ® GeneChip ® fluidic station and scanner. Data analysis All data processing and statistical analyses were performed using the statistical language R. Raw signal intensities were normalized and summarized according to the standard Affymetrix MA Suite 5.0 algorithm using the program Bioconductor (http://www.bioconductor.org/). Signal intensities were ln-transformed prior to statistical analyses. MA Suite 5.0 expression values have been submitted to the Gene Expression Omnibus (accession number XX). We define a “group” as one of domesticus, musculus, castaneus, F1 hybrids from one direction of the cross, or F1 hybrids from the other direction of the cross. We called a transcript “expressed” in this
Recommended publications
  • Circular RNA Hsa Circ 0005114‑Mir‑142‑3P/Mir‑590‑5P‑ Adenomatous
    ONCOLOGY LETTERS 21: 58, 2021 Circular RNA hsa_circ_0005114‑miR‑142‑3p/miR‑590‑5p‑ adenomatous polyposis coli protein axis as a potential target for treatment of glioma BO WEI1*, LE WANG2* and JINGWEI ZHAO1 1Department of Neurosurgery, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033; 2Department of Ophthalmology, The First Hospital of Jilin University, Jilin University, Changchun, Jilin 130021, P.R. China Received September 12, 2019; Accepted October 22, 2020 DOI: 10.3892/ol.2020.12320 Abstract. Glioma is the most common type of brain tumor APC expression with a good overall survival rate. UALCAN and is associated with a high mortality rate. Despite recent analysis using TCGA data of glioblastoma multiforme and the advances in treatment options, the overall prognosis in patients GSE25632 and GSE103229 microarray datasets showed that with glioma remains poor. Studies have suggested that circular hsa‑miR‑142‑3p/hsa‑miR‑590‑5p was upregulated and APC (circ)RNAs serve important roles in the development and was downregulated. Thus, hsa‑miR‑142‑3p/hsa‑miR‑590‑5p‑ progression of glioma and may have potential as therapeutic APC‑related circ/ceRNA axes may be important in glioma, targets. However, the expression profiles of circRNAs and their and hsa_circ_0005114 interacted with both of these miRNAs. functions in glioma have rarely been studied. The present study Functional analysis showed that hsa_circ_0005114 was aimed to screen differentially expressed circRNAs (DECs) involved in insulin secretion, while APC was associated with between glioma and normal brain tissues using sequencing the Wnt signaling pathway. In conclusion, hsa_circ_0005114‑ data collected from the Gene Expression Omnibus database miR‑142‑3p/miR‑590‑5p‑APC ceRNA axes may be potential (GSE86202 and GSE92322 datasets) and explain their mecha‑ targets for the treatment of glioma.
    [Show full text]
  • Genetic and Genomic Analysis of Hyperlipidemia, Obesity and Diabetes Using (C57BL/6J × TALLYHO/Jngj) F2 Mice
    University of Tennessee, Knoxville TRACE: Tennessee Research and Creative Exchange Nutrition Publications and Other Works Nutrition 12-19-2010 Genetic and genomic analysis of hyperlipidemia, obesity and diabetes using (C57BL/6J × TALLYHO/JngJ) F2 mice Taryn P. Stewart Marshall University Hyoung Y. Kim University of Tennessee - Knoxville, [email protected] Arnold M. Saxton University of Tennessee - Knoxville, [email protected] Jung H. Kim Marshall University Follow this and additional works at: https://trace.tennessee.edu/utk_nutrpubs Part of the Animal Sciences Commons, and the Nutrition Commons Recommended Citation BMC Genomics 2010, 11:713 doi:10.1186/1471-2164-11-713 This Article is brought to you for free and open access by the Nutrition at TRACE: Tennessee Research and Creative Exchange. It has been accepted for inclusion in Nutrition Publications and Other Works by an authorized administrator of TRACE: Tennessee Research and Creative Exchange. For more information, please contact [email protected]. Stewart et al. BMC Genomics 2010, 11:713 http://www.biomedcentral.com/1471-2164/11/713 RESEARCH ARTICLE Open Access Genetic and genomic analysis of hyperlipidemia, obesity and diabetes using (C57BL/6J × TALLYHO/JngJ) F2 mice Taryn P Stewart1, Hyoung Yon Kim2, Arnold M Saxton3, Jung Han Kim1* Abstract Background: Type 2 diabetes (T2D) is the most common form of diabetes in humans and is closely associated with dyslipidemia and obesity that magnifies the mortality and morbidity related to T2D. The genetic contribution to human T2D and related metabolic disorders is evident, and mostly follows polygenic inheritance. The TALLYHO/ JngJ (TH) mice are a polygenic model for T2D characterized by obesity, hyperinsulinemia, impaired glucose uptake and tolerance, hyperlipidemia, and hyperglycemia.
    [Show full text]
  • Gwas Meta-Analysis of Intelligence – Supplementary Note 1
    GWAS META-ANALYSIS OF INTELLIGENCE – SUPPLEMENTARY NOTE 1 Supplementary Note for GWAS meta-analysis (N=279,930) identifies new genes and functional links to intelligence Jeanne E Savage1#, Philip R Jansen1,2#, Sven Stringer1, Kyoko Watanabe1, Julien Bryois3, Christiaan A de Leeuw1, Mats Nagel1, Swapnil Awasthi4, Peter B Barr5, Jonathan R I Coleman6,7, Katrina L Grasby8, Anke R Hammerschlag1, Jakob Kaminski4,9, Robert Karlsson3, Eva Krapohl6, Max Lam10, Marianne Nygaard11,12, Chandra A. Reynolds13, Joey W. Trampush14, Hannah Young15, Delilah Zabaneh6, Sara Hägg3, Narelle K Hansell16, Ida K Karlsson3, Sten Linnarsson17, Grant W Montgomery8,18, Ana B. Muñoz-Manchado17, Erin B Quinlan6, Gunter Schumann6, Nathan Skene17, Bradley T Webb19,20,21, Tonya White2, Dan E Arking22, Deborah K Attix23,24, Dimitrios Avramopoulos22,25, Robert M Bilder26, Panos Bitsios27, Katherine E Burdick28,29,30, Tyrone D Cannon31, Ornit Chiba-Falek32, Andrea Christoforou23, Elizabeth T Cirulli33, Eliza Congdon26, Aiden Corvin34, Gail Davies35, 36, Ian J Deary35,36, Pamela DeRosse37, Dwight Dickinson38, Srdjan Djurovic39,40, Gary Donohoe41, Emily Drabant Conley42, Johan G Eriksson43,44, Thomas Espeseth45,46, Nelson A Freimer26,Stella Giakoumaki47, Ina Giegling48, Michael Gill34, David C Glahn49, Ahmad R Hariri50, Alex Hatzimanolis51,52,53, Matthew C Keller54, Emma Knowles49, Bettina Konte48, Jari Lahti55,56, Stephanie Le Hellard23,40, Todd Lencz37,57,58, David C Liewald36, Edythe London26, Astri J Lundervold59, Anil K Malhotra37,57,58, Ingrid Melle40,46, , Derek Morris41,
    [Show full text]
  • Whole Exome Sequencing in Families at High Risk for Hodgkin Lymphoma: Identification of a Predisposing Mutation in the KDR Gene
    Hodgkin Lymphoma SUPPLEMENTARY APPENDIX Whole exome sequencing in families at high risk for Hodgkin lymphoma: identification of a predisposing mutation in the KDR gene Melissa Rotunno, 1 Mary L. McMaster, 1 Joseph Boland, 2 Sara Bass, 2 Xijun Zhang, 2 Laurie Burdett, 2 Belynda Hicks, 2 Sarangan Ravichandran, 3 Brian T. Luke, 3 Meredith Yeager, 2 Laura Fontaine, 4 Paula L. Hyland, 1 Alisa M. Goldstein, 1 NCI DCEG Cancer Sequencing Working Group, NCI DCEG Cancer Genomics Research Laboratory, Stephen J. Chanock, 5 Neil E. Caporaso, 1 Margaret A. Tucker, 6 and Lynn R. Goldin 1 1Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD; 2Cancer Genomics Research Laboratory, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD; 3Ad - vanced Biomedical Computing Center, Leidos Biomedical Research Inc.; Frederick National Laboratory for Cancer Research, Frederick, MD; 4Westat, Inc., Rockville MD; 5Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD; and 6Human Genetics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD, USA ©2016 Ferrata Storti Foundation. This is an open-access paper. doi:10.3324/haematol.2015.135475 Received: August 19, 2015. Accepted: January 7, 2016. Pre-published: June 13, 2016. Correspondence: [email protected] Supplemental Author Information: NCI DCEG Cancer Sequencing Working Group: Mark H. Greene, Allan Hildesheim, Nan Hu, Maria Theresa Landi, Jennifer Loud, Phuong Mai, Lisa Mirabello, Lindsay Morton, Dilys Parry, Anand Pathak, Douglas R. Stewart, Philip R. Taylor, Geoffrey S. Tobias, Xiaohong R. Yang, Guoqin Yu NCI DCEG Cancer Genomics Research Laboratory: Salma Chowdhury, Michael Cullen, Casey Dagnall, Herbert Higson, Amy A.
    [Show full text]
  • A Meta-Analysis of Gene Expression Data Highlights Synaptic Dysfunction
    www.nature.com/scientificreports OPEN A meta-analysis of gene expression data highlights synaptic dysfunction in the hippocampus of brains with Alzheimer’s disease Saeedeh Hosseinian1, Ehsan Arefan2,3 ✉ , Hassan Rakhsh-Khorshid4, Mehdi Eivani 5, Ameneh Rezayof6, Hamid Pezeshk 7,8 & Sayed-Amir Marashi 1 Since the world population is ageing, dementia is going to be a growing concern. Alzheimer’s disease is the most common form of dementia. The pathogenesis of Alzheimer’s disease is extensively studied, yet unknown remains. Therefore, we aimed to extract new knowledge from existing data. We analysed about 2700 upregulated genes and 2200 downregulated genes from three studies on the CA1 of the hippocampus of brains with Alzheimer’s disease. We found that only the calcium signalling pathway enriched by 48 downregulated genes was consistent between all three studies. We predicted miR-129 to target nine out of 48 genes. Then, we validated miR-129 to regulate six out of nine genes in HEK cells. We noticed that four out of six genes play a role in synaptic plasticity. Finally, we confrmed the upregulation of miR-129 in the hippocampus of brains of rats with scopolamine-induced amnesia as a model of Alzheimer’s disease. We suggest that future research should investigate the possible role of miR-129 in synaptic plasticity and Alzheimer’s disease. This paper presents a novel framework to gain insight into potential biomarkers and targets for diagnosis and treatment of diseases. Alzheimer’s disease (AD) is the most common form of dementia. It mostly afects people aged 65 and older, pro- gresses slowly and leads to death in an average of nine years afer diagnosis.
    [Show full text]
  • Data Sheet 140
    Rudolf-Wissell-Str. 28a Background 37079 Göttingen, Germany Phone: +49 551-50556-0 RIMs are presynaptic active zone proteins that regulate Ca2+ triggered release of neurotransmitters. Fax: +49 551-50556-384 RIM 1α and RIM 2α are composed of an N-terminal zinc-finger domain, a central PDZ domain and two E-mail: [email protected] C-terminal C2 domains that are seperated by long alternatively spliced sequences. Web: www.sysy.com RIM 2β consists of a specific N-terminus, the central PDZ domain and the C-terminal C2 domains. The RIM 2 mRNA for RIM 2β is transcribed from an internal promoter of the RIM 2α gene. Shorter variants of RIM 2 which comprise only the C-terminal C2B domain and some flanking regions Cat.No. 140 303; Polyclonal rabbit antibody, 50 µg specific antibody (lyophilized) are referred to as NIM 2 / RIM 2γ and NIM 3 / RIM 3γ. Data Sheet Selected References for 140 303 RIM1/2-Mediated Facilitation of Cav1.4 Channel Opening Is Required for Ca2+-Stimulated Release in Mouse Rod Reconstitution/ 50 µg specific antibody, lyophilized. Affinity purified with the immunogen. Photoreceptors. Grabner CP, Gandini MA, Rehak R, Le Y, Zamponi GW, Schmitz F Storage Albumin was added for stabilization. For reconstitution add 50 µl H2O to get a 1mg/ml solution in PBS. Then aliquot and store at -20°C to -80°C until use. The Journal of neuroscience : the official journal of the Society for Neuroscience (2015) 3538: 13133-47. IHC, WB For detailed information, see back of the data sheet. Analysis of RIM Expression and Function at Mouse Photoreceptor Ribbon Synapses.
    [Show full text]
  • Loss of Twist1 in the Mesenchymal Compartment Promotes Increased
    Loss of Twist1 in the Mesenchymal Compartment Promotes Increased Fibrosis in Experimental Lung Injury by Enhanced Expression of CXCL12 This information is current as of October 2, 2021. Jiangning Tan, John R. Tedrow, Mehdi Nouraie, Justin A. Dutta, David T. Miller, Xiaoyun Li, Shibing Yu, Yanxia Chu, Brenda Juan-Guardela, Naftali Kaminski, Kritika Ramani, Partha S. Biswas, Yingze Zhang and Daniel J. Kass J Immunol 2017; 198:2269-2285; Prepublished online 8 Downloaded from February 2017; doi: 10.4049/jimmunol.1600610 http://www.jimmunol.org/content/198/6/2269 http://www.jimmunol.org/ Supplementary http://www.jimmunol.org/content/suppl/2017/02/07/jimmunol.160061 Material 0.DCSupplemental References This article cites 65 articles, 15 of which you can access for free at: http://www.jimmunol.org/content/198/6/2269.full#ref-list-1 Why The JI? Submit online. by guest on October 2, 2021 • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication *average Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Author Choice Freely available online through The Journal of Immunology Author Choice option Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2017 by The American Association of Immunologists, Inc.
    [Show full text]
  • Role of PDZ-Binding Motif from West Nile Virus NS5 Protein on Viral
    www.nature.com/scientificreports OPEN Role of PDZ‑binding motif from West Nile virus NS5 protein on viral replication Emilie Giraud1*, Chloé Otero del Val2, Célia Caillet‑Saguy2, Nada Zehrouni2, Cécile Khou5, Joël Caillet4, Yves Jacob3, Nathalie Pardigon5 & Nicolas Wolf2 West Nile virus (WNV) is a Flavivirus, which can cause febrile illness in humans that may progress to encephalitis. Like any other obligate intracellular pathogens, Flaviviruses hijack cellular protein functions as a strategy for sustaining their life cycle. Many cellular proteins display globular domain known as PDZ domain that interacts with PDZ‑Binding Motifs (PBM) identifed in many viral proteins. Thus, cellular PDZ‑containing proteins are common targets during viral infection. The non‑structural protein 5 (NS5) from WNV provides both RNA cap methyltransferase and RNA polymerase activities and is involved in viral replication but its interactions with host proteins remain poorly known. In this study, we demonstrate that the C‑terminal PBM of WNV NS5 recognizes several human PDZ‑ containing proteins using both in vitro and in cellulo high‑throughput methods. Furthermore, we constructed and assayed in cell culture WNV replicons where the PBM within NS5 was mutated. Our results demonstrate that the PBM of WNV NS5 is important in WNV replication. Moreover, we show that knockdown of the PDZ‑containing proteins TJP1, PARD3, ARHGAP21 or SHANK2 results in the decrease of WNV replication in cells. Altogether, our data reveal that interactions between the PBM of NS5 and PDZ‑containing proteins afect West Nile virus replication. Arboviruses include numerous human and animal pathogens that are important global health threats responsible for arboviroses.
    [Show full text]
  • The Molecular Machinery of Neurotransmitter Release Nobel Lecture, 7 December 2013
    The Molecular Machinery of Neurotransmitter Release Nobel Lecture, 7 December 2013 by Thomas C. Südhof Dept. of Molecular and Cellular Physiology, and Howard Hughes Medical Institute, Stanford University, USA. 1. THE NEUROTRANSMITTER RELEASE ENIGMA Synapses have a long history in science. Synapses were frst functionally demon- strated by Emil duBois-Reymond (1818–1896), were morphologically identifed by classical neuroanatomists such as Rudolf von Kölliker (1817–1905) and San- tiago Ramon y Cajal (1852–1934), and named in 1897 by Michael Foster (1836– 1907). Although the chemical nature of synaptic transmission was already sug- gested by duBois-Reymond, it was long disputed because of its incredible speed. Over time, however, overwhelming evidence established that most synapses use chemical messengers called neurotransmitters, most notably with the pioneer- ing contributions by Otto Loewi (1873–1961), Henry Dale (1875–1968), Ulf von Euler (1905–1983), and Julius Axelrod (1912–2004). In parallel, arguably the most important advance to understanding how synapses work was provided by Bernard Katz (1911–2003), who elucidated the principal mechanism of syn- aptic transmission (Katz, 1969). Most initial studies on synapses were carried out on the neuromuscular junction, and central synapses have only come to the fore in recent decades. Here, major contributions by many scientists, including George Palade, Rodolfo Llinas, Chuck Stevens, Bert Sakmann, Eric Kandel, and Victor Whittaker, to name just a few, not only confrmed the principal results obtained in the neuromuscular junction by Katz, but also revealed that synapses 259 6490_Book.indb 259 11/4/14 2:29 PM 260 The Nobel Prizes exhibit an enormous diversity of properties as well as an unexpected capacity for plasticity.
    [Show full text]
  • Rabbit Anti-Epac2/FITC Conjugated Antibody
    SunLong Biotech Co.,LTD Tel: 0086-571- 56623320 Fax:0086-571- 56623318 E-mail:[email protected] www.sunlongbiotech.com Rabbit Anti-Epac2/FITC Conjugated antibody SL10955R-FITC Product Name: Anti-Epac2/FITC Chinese Name: FITC标记的鸟嘌呤核苷酸交换因子4 CAMP GEFII; cAMP regulated guanine nucleotide exchange factor II; cAMP-GEFII; cAMP-regulated guanine nucleotide exchange factor II; CAMPGEFII; CGEF 2; CGEF2; Epac 2; EPAC2; Exchange factor directly activated by cAMP 2; Exchange Alias: protein directly activated by cAMP 2; Nbla00496; Putative protein product of Nbla00496; Rap guanine nucleotide exchange factor (GEF) 4; RAP guanine nucleotide exchange factor 4; RAPGEF 4; RAPGEF4; RPGF4_HUMAN. Organism Species: Rabbit Clonality: Polyclonal React Species: ICC=1:50-200IF=1:50-200 Applications: not yet tested in other applications. optimal dilutions/concentrations should be determined by the end user. Molecular weight: 116kDa Form: Lyophilized or Liquid Concentration: 1mg/ml immunogen: KLHwww.sunlongbiotech.com conjugated synthetic peptide derived from human Epac2 Lsotype: IgG Purification: affinity purified by Protein A Storage Buffer: 0.01M TBS(pH7.4) with 1% BSA, 0.03% Proclin300 and 50% Glycerol. Store at -20 °C for one year. Avoid repeated freeze/thaw cycles. The lyophilized antibody is stable at room temperature for at least one month and for greater than a year Storage: when kept at -20°C. When reconstituted in sterile pH 7.4 0.01M PBS or diluent of antibody the antibody is stable for at least two weeks at 2-4 °C. background: 3',5' cyclic adenosine monophosphate (cAMP)-regulated guanine nucleotide exchange Product Detail: factors Epac1 (cAMP-GEFI) and Epac2 (cAMP-GEFII) activate the ras family GTPases Rap1 and Rap2 by promoting GTP binding in a cAMP-dependent manner (1,2).
    [Show full text]
  • GSE50161, (C) GSE66354, (D) GSE74195 and (E) GSE86574
    Figure S1. Boxplots of normalized samples in five datasets. (A) GSE25604, (B) GSE50161, (C) GSE66354, (D) GSE74195 and (E) GSE86574. The x‑axes indicate samples, and the y‑axes represent the expression of genes. Figure S2. Volanco plots of DEGs in five datasets. (A) GSE25604, (B) GSE50161, (C) GSE66354, (D) GSE74195 and (E) GSE86574. Red nodes represent upregulated DEGs and green nodes indicate downregulated DEGs. Cut‑off criteria were P<0.05 and |log2 FC|>1. DEGs, differentially expressed genes; FC, fold change; adj.P.Val, adjusted P‑value. Figure S3. Transcription factor‑gene regulatory network constructed using the Cytoscape iRegulion plug‑in. Table SI. Primer sequences for reverse transcription‑quantitative polymerase chain reaction. Genes Sequences hsa‑miR‑124 F: 5'‑ACACTCCAGCTGGGCAGCAGCAATTCATGTTT‑3' R: 5'‑CTCAACTGGTGTCGTGGA‑3' hsa‑miR‑330‑3p F: 5'‑CATGAATTCACTCTCCCCGTTTCTCCCTCTGC‑3' R: 5'‑CCTGCGGCCGCGAGCCGCCCTGTTTGTCTGAG‑3' hsa‑miR‑34a‑5p F: 5'‑TGGCAGTGTCTTAGCTGGTTGT‑3' R: 5'‑GCGAGCACAGAATTAATACGAC‑3' hsa‑miR‑449a F: 5'‑TGCGGTGGCAGTGTATTGTTAGC‑3' R: 5'‑CCAGTGCAGGGTCCGAGGT‑3' CD44 F: 5'‑CGGACACCATGGACAAGTTT‑3' R: 5'‑TGTCAATCCAGTTTCAGCATCA‑3' PCNA F: 5'‑GAACTGGTTCATTCATCTCTATGG‑3' F: 5'‑TGTCACAGACAAGTAATGTCGATAAA‑3' SYT1 F: 5'‑CAATAGCCATAGTCGCAGTCCT‑3' R: 5'‑TGTCAATCCAGTTTCAGCATCA‑3' U6 F: 5'‑GCTTCGGCAGCACATATACTAAAAT‑3' R: 5'‑CGCTTCACGAATTTGCGTGTCAT‑3' GAPDH F: 5'‑GGAAAGCTGTGGCGTGAT‑3' R: 5'‑AAGGTGGAAGAATGGGAGTT‑3' hsa, homo sapiens; miR, microRNA; CD44, CD44 molecule (Indian blood group); PCNA, proliferating cell nuclear antigen;
    [Show full text]
  • Analysis and Functional Evaluation of the Hair-Cell Transcriptome
    Analysis and functional evaluation of the hair-cell transcriptome Brian M. McDermott, Jr.*, Jessica M. Baucom, and A. J. Hudspeth† Howard Hughes Medical Institute and Laboratory of Sensory Neuroscience, The Rockefeller University, 1230 York Avenue, New York, NY 10021-6399 Contributed by A. J. Hudspeth, May 17, 2007 (sent for review March 31, 2007) An understanding of the molecular bases of the morphogenesis, ciated from the lagena, a receptor organ of the zebrafish’s ear. organization, and functioning of hair cells requires that the genes Linear amplification of the RNA from 200 hair cells yielded Ϸ40 expressed in these cells be identified and their functions ascer- ␮g of aRNA, an enhancement of Ϸ1 millionfold. The resultant tained. After purifying zebrafish hair cells and detecting mRNAs labeled aRNA was hybridized to an Affymetrix microarray with oligonucleotide microarrays, we developed a subtractive (Affymetrix, Santa Clara, CA) containing Ϸ15,000 oligonucle- strategy that identified 1,037 hair cell-expressed genes whose otide probe sets. Averaging the outcomes of three experiments cognate proteins subserve functions including membrane trans- (SI Data Set 1) resulted in the identification of 6,472 transcripts port, synaptic transmission, transcriptional control, cellular adhe- scored as ‘‘present’’ (SI Data Set 2). sion and signal transduction, and cytoskeletal organization. To In the second step, we defined the transcriptome from cells of assess the validity of the subtracted hair-cell data set, we verified a nonsensory organ, the liver (SI Data Set 1). Hepatocytes were the presence of 11 transcripts in inner-ear tissue. Functional eval- selected for the subtraction process for three reasons: they are uation of two genes from the subtracted data set revealed their nonneuronal and thus unlikely to express synaptic factors; they importance in hair bundles: zebrafish larvae bearing the seahorse lack cilia (http://members.global2000.net/bowser/cilialist.html) and ift 172 mutations display specific kinociliary defects.
    [Show full text]