Motor Neuron Disease Occurring in a Mutant Dynactin Mouse Model Is Characterized by Defects in Vesicular Trafficking

Total Page:16

File Type:pdf, Size:1020Kb

Motor Neuron Disease Occurring in a Mutant Dynactin Mouse Model Is Characterized by Defects in Vesicular Trafficking The Journal of Neuroscience, February 27, 2008 • 28(9):1997–2005 • 1997 Neurobiology of Disease Motor Neuron Disease Occurring in a Mutant Dynactin Mouse Model Is Characterized by Defects in Vesicular Trafficking Fiona M. Laird,1,4* Mohamed H. Farah,1,4* Steven Ackerley,1,4*† Ahmet Hoke,2,3 Nicholas Maragakis,3 Jeffrey D. Rothstein,2,3 John Griffin,3 Donald L. Price,1,2,3,4 Lee J. Martin,1,2,4 and Philip C. Wong1,2,4 Departments of 1Pathology, 2Neuroscience, and 3Neurology, and 4Division of Neuropathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205 Amyotrophic lateral sclerosis (ALS), a fatal and progressive neurodegenerative disorder characterized by weakness, muscle atrophy, and spasticity, is the most common adult-onset motor neuron disease. Although the majority of ALS cases are sporadic, ϳ5–10% are familial, including those linked to mutations in SOD1 (Cu/Zn superoxide dismutase). Missense mutations in a dynactin gene (DCTN1) encoding the p150 Glued subunit of dynactin have been linked to both familial and sporadic ALS. To determine the molecular mechanism whereby mutant dynactin p150 Glued causes selective degeneration of motor neurons, we generated and characterized mice expressing either wild-type or mutant human dynactin p150 Glued. Neuronal expression of mutant, but not wild type, dynactin p150 Glued causes motor neuron disease in these animals that are characterized by defects in vesicular transport in cell bodies of motor neurons, axonal swelling and axo-terminal degeneration. Importantly, we provide evidence that autophagic cell death is implicated in the pathogenesis of mutant p150 Glued mice. This novel mouse model will be instrumental for not only clarifying disease mechanisms in ALS, but also for testing therapeutic strategies to ameliorate this devastating disease. Key words: dynactin; p150 Glued; ALS; motor neuron disease model; vesicular trafficking; autophagy Introduction ties of axons in the skin. Since this first report, additional muta- A family with a slowly progressive autosomal dominant lower tions in the dynactin gene (DCTN1) have been associated with motor neuron disease has been linked to a mutation in the motor neuron disease (Munch et al., 2004). p150 Glued subunit (G59S) of the dynactin complex (Puls et al., Dynactin p150 Glued is a critical component of a multiprotein 2003). This disease begins in early adult life with vocal cord pa- complex that differentially copurifies with dynein (Gill et al., ralysis (associated with breathing difficulties), facial weakness, 1991; Schroer and Sheetz, 1991; Schroer, 2004), the molecular and atrophy of muscles in the hands (Puls et al., 2003, 2005). motor required for fast retrograde transport of vesicles, or- Subsequently, weakness and atrophy appear in the distal lower ganelles, RNAs and proteins along microtubules (Schroer, 2004; extremities. There is evidence of degeneration of motor axons, Hirokawa and Takemura, 2005; Price et al., 2005). Although dy- and reduced numbers of motor neurons in the spinal cord and nactin has been postulated to enhance the processivity and effi- hypoglossal nucleus (Puls et al., 2005). Significantly, inclusions ciency of the dynein motor (LaMonte et al., 2002; Schroer, 2004), containing components of dynactin and dynein are evident in previous studies demonstrate in vitro that the dynein-dynactin motor neurons (Puls et al., 2005). Although no sensory deficits complexes exhibit bidirectional processive motility (Ross et al., are reported, examinations of skin biopsies disclose abnormali- 2006) and in vivo that dynactin is required for coordinated an- terograde and retrograde organelle movement (Haghnia et al., 2007). The disruption of the dynactin complex by overexpression Received Sept. 14, 2007; revised Dec. 13, 2007; accepted Jan. 6, 2008. of dynamitin (LaMonte et al., 2002) results in motor neuron This work was supported in part by The Muscular Dystrophy Association, The Robert Packard Center for Amyo- trophic Lateral Sclerosis Research, and National Institute of Neurological Disorder and Stroke Grant RO1 NS40014 disease in mice, but the exact pathogenic mechanism remains (P.C.W.). The Thy1.2 promoter was a gift from P. Caroni. We thank V. Nehus, Y. Pan, D. Prasad, Ashley Moon, and M. unknown. Bidirectional transport of vesicles between the endo- Polley for their technical support, and Drs. H. Cai, A. Ludolph, A. Kolodkin, and T. Lloyd for sharing of unpublished plasmic reticulum (ER) and Golgi apparatus have also been results. Our coauthor and friend, Dr. Steven Ackerley, passed away suddenly on January 14, 2008. With deep sorrow shown to involve dynein/dynactin mediated transport (Lee et al., and mourning, we dedicate this paper to his memory. *F.M.L., M.H.F., and S.A. contributed equally to this work. 2004). Disruption of dynein function also impairs clearance of †Deceased, Jan. 14, 2008. aggregate prone proteins by autophagy (Ravikumar et al., 2005), Correspondence should be addressed to Philip C. Wong, Department of Pathology, The Johns Hopkins University a lysosomal pathway for degrading damaged organelles and ag- School of Medicine, 558 Ross Research Building, 720 Rutland Avenue, Baltimore, MD 21205-2196. E-mail: gregated proteins within the cell. Autophagy has been implicated [email protected]. DOI:10.1523/JNEUROSCI.4231-07.2008 in a number of neurodegenerative disorders, but considerable Copyright © 2008 Society for Neuroscience 0270-6474/08/281997-09$15.00/0 debate exists regarding the role of autophagy in pathogenesis of 1998 • J. Neurosci., February 27, 2008 • 28(9):1997–2005 Laird et al. • A Mutant Dynactin Mouse Model of Motor Neuron Disease specific diseases (Nixon, 2006). The dem- onstration that genetic deletions of auto- phagy genes in neurons are sufficient to cause neurodegeneration supports the view that disruption of basal autophagy participates in pathogenesis of some of these illnesses (Hara et al., 2006; Komatsu et al., 2006). Therefore, given the numerous roles that dynactin plays in intracellular and ax- onal transport, it is conceivable that alter- ation in dynactin function could impact on several critical cellular processes within various compartments of the motor neu- ron. Here, we document that a novel mouse model expressing human mutant p150 Glued dynactin exhibits clinical and pathological hallmarks of motor neuron disease. Importantly, we show that motor neuron degeneration in these mutant dyn- actin mice is associated with abnormalities in intracellular vesicular trafficking, axonal swelling and axo-terminal degeneration. Moreover, autophagic cell death is impli- cated in the pathogenesis of disease in mu- tant p150 Glued mice. Materials and Methods Generation of transgenic mice. We first engi- neered the G59S amyotrophic lateral sclerosis (ALS)-linked mutation (Puls et al., 2003) into the human dynactin p150Glued cDNA by PCR/ oligonucleotide primer-directed mutagenesis strategy. In addition, the full-length human cDNA encoding wild-type and mutant dynactin p150 Glued were modified to include a C-terminal epitope tag (FLAG) to facilitate identification of the transgenic product. Be- cause previous studies show that the Thy1.2 cas- sette works well for generation of mice with neu- ronal expression and was the strategy used for Figure 1. Motor neuron disease in mice expressing mutant but not wild-type p150 Glued. A, Schematic drawing of the con- the overexpression of dynactin p50 (LaMonte et structusedtogeneratetransgenicmice.Humanwild-typeormutantp150 Glued cDNAwithaFLAGtag(indicatedbyastripedbox) al., 2002), the resulting constructs were sub- were cloned into the Thy1.2 expression cassette at the XhoI site. B, Accumulation of mutant G59S dynactin in mice. Total protein cloned into the XhoI site of the mouse Thy1.2 (20 ␮g) extracted from nontransgenic, wild-type, and mutant dynactin mice were electrophoresed on 6% SDS-PAGE gels and expression cassette to generate expression con- immunoblotted using a monoclonal antibody recognizing the p150 Glued (top) or the FLAG (middle) epitope. Identical blot was structs, one encoding wild-type dynactin reprobed with antisera against actin (bottom). C, Kaplan-Meier plot of the onset of tremor for two mutant lines, M1 (gray line) p150 glued (Thy1.2p150WT), and the other G59S Ϯ Ϯ glued and M2 (black line). The average onset of tremor for M1 is 4.8 1.2 months and for M2 is 6.2 1.3months. D, Onset of the dynactin p150 (Thy1.2p150G59S). These reduction in grip strength for line M1 is plotted on a Kaplan-Meier graph. The black and gray dotted lines denote forelimb and vectors were initially tested in cultured cells by hindlimb grip strength, respectively. E, Kaplan-Meier plot for survival of two mutant lines; line M1 (gray) has a shorter lifespan transient transfection to verify their expression than line M2 (black). (data not shown). Subsequently, the purified in- serts from these vectors were submitted to the Indianapolis, IN). Protein extracts were sonicated before protein assay Transgenic Facility at Johns Hopkins University School of Medicine for and protein blot analysis. 20 ␮g of total protein extracts were loaded onto injection into C57BL/6;SJL hybrid mouse embryos. Positive founders a 6 or 16% Tris-glycine gel (Invitrogen, San Diego, CA) and transferred were identified by Southern blot analysis and two lines of mutant and glued to polyvinylidene difluoride membrane (Invitrogen). Immunoblotting three lines of wild-type dynactin p150 were established. F1 offspring, was performed using primary antibodies to p150
Recommended publications
  • Comprehensive Analyses of 723 Transcriptomes Enhance Genetic and Biological Interpretations for Complex Traits in Cattle
    Downloaded from genome.cshlp.org on October 3, 2021 - Published by Cold Spring Harbor Laboratory Press Resource Comprehensive analyses of 723 transcriptomes enhance genetic and biological interpretations for complex traits in cattle Lingzhao Fang,1,2,3,4,8 Wentao Cai,2,5,8 Shuli Liu,1,5,8 Oriol Canela-Xandri,3,4,8 Yahui Gao,1,2 Jicai Jiang,2 Konrad Rawlik,3 Bingjie Li,1 Steven G. Schroeder,1 Benjamin D. Rosen,1 Cong-jun Li,1 Tad S. Sonstegard,6 Leeson J. Alexander,7 Curtis P. Van Tassell,1 Paul M. VanRaden,1 John B. Cole,1 Ying Yu,5 Shengli Zhang,5 Albert Tenesa,3,4 Li Ma,2 and George E. Liu1 1Animal Genomics and Improvement Laboratory, Henry A. Wallace Beltsville Agricultural Research Center, Agricultural Research Service, USDA, Beltsville, Maryland 20705, USA; 2Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland 20742, USA; 3The Roslin Institute, Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian EH25 9RG, United Kingdom; 4Medical Research Council Human Genetics Unit at the Medical Research Council Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh EH4 2XU, United Kingdom; 5College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; 6Acceligen, Eagan, Minnesota 55121, USA; 7Fort Keogh Livestock and Range Research Laboratory, Agricultural Research Service, USDA, Miles City, Montana 59301, USA By uniformly analyzing 723 RNA-seq data from 91 tissues and cell types, we built a comprehensive gene atlas and studied tissue specificity of genes in cattle. We demonstrated that tissue-specific genes significantly reflected the tissue-relevant biol- ogy, showing distinct promoter methylation and evolution patterns (e.g., brain-specific genes evolve slowest, whereas testis- specific genes evolve fastest).
    [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]
  • High-Throughput Discovery of Novel Developmental Phenotypes
    High-throughput discovery of novel developmental phenotypes The Harvard community has made this article openly available. Please share how this access benefits you. Your story matters Citation Dickinson, M. E., A. M. Flenniken, X. Ji, L. Teboul, M. D. Wong, J. K. White, T. F. Meehan, et al. 2016. “High-throughput discovery of novel developmental phenotypes.” Nature 537 (7621): 508-514. doi:10.1038/nature19356. http://dx.doi.org/10.1038/nature19356. Published Version doi:10.1038/nature19356 Citable link http://nrs.harvard.edu/urn-3:HUL.InstRepos:32071918 Terms of Use This article was downloaded from Harvard University’s DASH repository, and is made available under the terms and conditions applicable to Other Posted Material, as set forth at http:// nrs.harvard.edu/urn-3:HUL.InstRepos:dash.current.terms-of- use#LAA HHS Public Access Author manuscript Author ManuscriptAuthor Manuscript Author Nature. Manuscript Author Author manuscript; Manuscript Author available in PMC 2017 March 14. Published in final edited form as: Nature. 2016 September 22; 537(7621): 508–514. doi:10.1038/nature19356. High-throughput discovery of novel developmental phenotypes A full list of authors and affiliations appears at the end of the article. Abstract Approximately one third of all mammalian genes are essential for life. Phenotypes resulting from mouse knockouts of these genes have provided tremendous insight into gene function and congenital disorders. As part of the International Mouse Phenotyping Consortium effort to generate and phenotypically characterize 5000 knockout mouse lines, we have identified 410 Users may view, print, copy, and download text and data-mine the content in such documents, for the purposes of academic research, subject always to the full Conditions of use:http://www.nature.com/authors/editorial_policies/license.html#terms #Corresponding author: [email protected].
    [Show full text]
  • Identification of Conserved Genes Triggering Puberty in European Sea
    Blázquez et al. BMC Genomics (2017) 18:441 DOI 10.1186/s12864-017-3823-2 RESEARCHARTICLE Open Access Identification of conserved genes triggering puberty in European sea bass males (Dicentrarchus labrax) by microarray expression profiling Mercedes Blázquez1,2* , Paula Medina1,2,3, Berta Crespo1,4, Ana Gómez1 and Silvia Zanuy1* Abstract Background: Spermatogenesisisacomplexprocesscharacterized by the activation and/or repression of a number of genes in a spatio-temporal manner. Pubertal development in males starts with the onset of the first spermatogenesis and implies the division of primary spermatogonia and their subsequent entry into meiosis. This study is aimed at the characterization of genes involved in the onset of puberty in European sea bass, and constitutes the first transcriptomic approach focused on meiosis in this species. Results: European sea bass testes collected at the onset of puberty (first successful reproduction) were grouped in stage I (resting stage), and stage II (proliferative stage). Transition from stage I to stage II was marked by an increase of 11ketotestosterone (11KT), the main fish androgen, whereas the transcriptomic study resulted in 315 genes differentially expressed between the two stages. The onset of puberty induced 1) an up-regulation of genes involved in cell proliferation, cell cycle and meiosis progression, 2) changes in genes related with reproduction and growth, and 3) a down-regulation of genes included in the retinoic acid (RA) signalling pathway. The analysis of GO-terms and biological pathways showed that cell cycle, cell division, cellular metabolic processes, and reproduction were affected, consistent with the early events that occur during the onset of puberty.
    [Show full text]
  • Aneuploidy: Using Genetic Instability to Preserve a Haploid Genome?
    Health Science Campus FINAL APPROVAL OF DISSERTATION Doctor of Philosophy in Biomedical Science (Cancer Biology) Aneuploidy: Using genetic instability to preserve a haploid genome? Submitted by: Ramona Ramdath In partial fulfillment of the requirements for the degree of Doctor of Philosophy in Biomedical Science Examination Committee Signature/Date Major Advisor: David Allison, M.D., Ph.D. Academic James Trempe, Ph.D. Advisory Committee: David Giovanucci, Ph.D. Randall Ruch, Ph.D. Ronald Mellgren, Ph.D. Senior Associate Dean College of Graduate Studies Michael S. Bisesi, Ph.D. Date of Defense: April 10, 2009 Aneuploidy: Using genetic instability to preserve a haploid genome? Ramona Ramdath University of Toledo, Health Science Campus 2009 Dedication I dedicate this dissertation to my grandfather who died of lung cancer two years ago, but who always instilled in us the value and importance of education. And to my mom and sister, both of whom have been pillars of support and stimulating conversations. To my sister, Rehanna, especially- I hope this inspires you to achieve all that you want to in life, academically and otherwise. ii Acknowledgements As we go through these academic journeys, there are so many along the way that make an impact not only on our work, but on our lives as well, and I would like to say a heartfelt thank you to all of those people: My Committee members- Dr. James Trempe, Dr. David Giovanucchi, Dr. Ronald Mellgren and Dr. Randall Ruch for their guidance, suggestions, support and confidence in me. My major advisor- Dr. David Allison, for his constructive criticism and positive reinforcement.
    [Show full text]
  • Supplementary Materials
    Supplementary materials Supplementary Table S1: MGNC compound library Ingredien Molecule Caco- Mol ID MW AlogP OB (%) BBB DL FASA- HL t Name Name 2 shengdi MOL012254 campesterol 400.8 7.63 37.58 1.34 0.98 0.7 0.21 20.2 shengdi MOL000519 coniferin 314.4 3.16 31.11 0.42 -0.2 0.3 0.27 74.6 beta- shengdi MOL000359 414.8 8.08 36.91 1.32 0.99 0.8 0.23 20.2 sitosterol pachymic shengdi MOL000289 528.9 6.54 33.63 0.1 -0.6 0.8 0 9.27 acid Poricoic acid shengdi MOL000291 484.7 5.64 30.52 -0.08 -0.9 0.8 0 8.67 B Chrysanthem shengdi MOL004492 585 8.24 38.72 0.51 -1 0.6 0.3 17.5 axanthin 20- shengdi MOL011455 Hexadecano 418.6 1.91 32.7 -0.24 -0.4 0.7 0.29 104 ylingenol huanglian MOL001454 berberine 336.4 3.45 36.86 1.24 0.57 0.8 0.19 6.57 huanglian MOL013352 Obacunone 454.6 2.68 43.29 0.01 -0.4 0.8 0.31 -13 huanglian MOL002894 berberrubine 322.4 3.2 35.74 1.07 0.17 0.7 0.24 6.46 huanglian MOL002897 epiberberine 336.4 3.45 43.09 1.17 0.4 0.8 0.19 6.1 huanglian MOL002903 (R)-Canadine 339.4 3.4 55.37 1.04 0.57 0.8 0.2 6.41 huanglian MOL002904 Berlambine 351.4 2.49 36.68 0.97 0.17 0.8 0.28 7.33 Corchorosid huanglian MOL002907 404.6 1.34 105 -0.91 -1.3 0.8 0.29 6.68 e A_qt Magnogrand huanglian MOL000622 266.4 1.18 63.71 0.02 -0.2 0.2 0.3 3.17 iolide huanglian MOL000762 Palmidin A 510.5 4.52 35.36 -0.38 -1.5 0.7 0.39 33.2 huanglian MOL000785 palmatine 352.4 3.65 64.6 1.33 0.37 0.7 0.13 2.25 huanglian MOL000098 quercetin 302.3 1.5 46.43 0.05 -0.8 0.3 0.38 14.4 huanglian MOL001458 coptisine 320.3 3.25 30.67 1.21 0.32 0.9 0.26 9.33 huanglian MOL002668 Worenine
    [Show full text]
  • The Rab6-Regulated KIF1C Kinesin Motor Domain Contributes to Golgi Organization Peter L Lee, Maikke B Ohlson†, Suzanne R Pfeffer*
    RESEARCH ARTICLE elifesciences.org The Rab6-regulated KIF1C kinesin motor domain contributes to Golgi organization Peter L Lee, Maikke B Ohlson†, Suzanne R Pfeffer* Department of Biochemistry, Stanford University School of Medicine, Stanford, United States Abstract Most kinesins transport cargoes bound to their C-termini and use N-terminal motor domains to move along microtubules. We report here a novel function for KIF1C: it transports Rab6A-vesicles and can influence Golgi complex organization. These activities correlate with KIF1C’s capacity to bind the Golgi protein Rab6A directly, both via its motor domain and C-terminus. Rab6A binding to the motor domain inhibits microtubule interaction in vitro and in cells, decreasing the amount of motile KIF1C. KIF1C depletion slows protein delivery to the cell surface, interferes with vesicle motility, and triggers Golgi fragmentation. KIF1C can protect Golgi membranes from fragmentation in cells lacking an intact microtubule network. Rescue of fragmentation requires sequences that enable KIF1C to bind Rab6A at both ends, but not KIF1C motor function. Rab6A binding to KIF1C’s motor domain represents an entirely new mode of regulation for a kinesin motor, and likely has important consequences for KIF1C’s cellular functions. DOI: 10.7554/eLife.06029.001 *For correspondence: pfeffer@ stanford.edu Introduction Kinesin superfamily proteins (KIFs) are microtubule-based motors that are responsible for the Present address: †Genentech, motility of membrane-bound compartments and transport vesicles (Hirokawa et al., 2009b; South San Francisco, United States Verhey and Hammond, 2009). Of fundamental interest is how these motor proteins link to specific membrane cargoes and how they are regulated.
    [Show full text]
  • Anti-RAB6A Antibody (ARG66404)
    Product datasheet [email protected] ARG66404 Package: 100 μl anti-RAB6A antibody Store at: -20°C Summary Product Description Rabbit Polyclonal antibody recognizes RAB6A Tested Reactivity Hu, Ms Tested Application IHC-P, WB Host Rabbit Clonality Polyclonal Isotype IgG Target Name RAB6A Antigen Species Human Immunogen Synthetic peptide of Human RAB6A. Conjugation Un-conjugated Alternate Names Ras-related protein Rab-6A; Rab-6; RAB6 Application Instructions Application table Application Dilution IHC-P 1:25 - 1:100 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 WB: 293T, Mouse brain and A549. IHC-P: Human skin. Calculated Mw 24 kDa Observed Size ~ 24 kDa Properties Form Liquid Purification Affinity purification with immunogen. Buffer PBS (pH 7.4), 0.05% Sodium azide and 40% Glycerol. Preservative 0.05% Sodium azide Stabilizer 40% Glycerol Concentration 1.3 mg/ml 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. www.arigobio.com 1/2 Note For laboratory research only, not for drug, diagnostic or other use. Bioinformation Gene Symbol RAB6A Gene Full Name RAB6A, member RAS oncogene family Background This gene encodes a member of the RAB family, which belongs to the small GTPase superfamily. GTPases of the RAB family bind to various effectors to regulate the targeting and fusion of transport carriers to acceptor compartments.
    [Show full text]
  • Infection and Transport of Herpes Simplex Virus Type 1 in Neurons: Role of the Cytoskeleton
    viruses Review Infection and Transport of Herpes Simplex Virus Type 1 in Neurons: Role of the Cytoskeleton Monica Miranda-Saksena 1,* ID , Christopher E. Denes 1 ID , Russell J. Diefenbach 2 ID and Anthony L. Cunningham 1,* ID 1 Centre for Virus Research, The Westmead Institute for Medical Research, The University of Sydney, Westmead NSW 2145, Australia; [email protected] 2 Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney NSW 2109, Australia; [email protected] * Correspondence: [email protected] (M.M.S.); [email protected] (A.L.C.); Tel.: +612-8627-3624 (M.M.S.) Received: 24 January 2018; Accepted: 20 February 2018; Published: 23 February 2018 Abstract: Herpes simplex virus type 1 (HSV-1) is a neuroinvasive human pathogen that has the ability to infect and replicate within epithelial cells and neurons and establish a life-long latent infection in sensory neurons. HSV-1 depends on the host cellular cytoskeleton for entry, replication, and exit. Therefore, HSV-1 has adapted mechanisms to promote its survival by exploiting the microtubule and actin cytoskeletons to direct its active transport, infection, and spread between neurons and epithelial cells during primary and recurrent infections. This review will focus on the currently known mechanisms utilized by HSV-1 to harness the neuronal cytoskeleton, molecular motors, and the secretory and exocytic pathways for efficient virus entry, axonal transport, replication, assembly, and exit from the distinct functional compartments (cell body and axon) of the highly polarized sensory neurons. Keywords: herpes simplex virus; neurons; axonal transport; cytoskeleton; microtubules; actin 1.
    [Show full text]
  • RAB6A Cdna Catalog Number: ATGD0203
    RAB6A cDNA Catalog Number: ATGD0203 PRODUCT INPORMATION Catalog number ATGD0203 Product type cDNA Species Human NCBI Accession No. NP_002860.2 Alternative Names RAB6 mRNA Refseq NM_002869.4 OMIM 179513 Chromosome location 11q13.3 PRODUCT SPECIFICATION Formulation Lyophilized Storage Store the plasmid at -20C. cDNA Size 627bp Preparation before usage 1. Centrifuge at 7000rpm for 1 minute. 2. Carefully open the vial and add 100ul of sterile water to dissolve the DNA. Each tube contains approximately 10ug of lyophilized plasmid. Vector description This shuttle vector contains the complete ORF. It is inseted BamH I to Xho I. The gene insert contains multiple cloning sites which can be used to easily cut and transfer the gene and recombination site into your expression vector. Cloning Vector pATGen (puc19-derived cloning vector) General Description 1 RAB6A cDNA Catalog Number: ATGD0203 RAB6A encodes a member of the RAB family, which belongs to the small GTPase superfamily. GTPases of the RAB family bind to various effectors to regulate the targeting and fusion of transport carriers to acceptor compartments. RAB6A is located at the Golgi apparatus, which regulates trafficking in both a retrograde (from early endosomes and Golgi to the endoplasmic reticulum) and an anterograde (from the Golgi to the plasma membrane) directions. Myosin II is an effector of this protein in these processes. RAB6A is also involved in assembly of human cytomegalovirus (HCMV) by interacting with the cellular protein Bicaudal D1, which interacts with the HCMV virion
    [Show full text]
  • Coupling Fission and Exit of RAB6 Vesicles At
    Coupling fission and exit of RAB6 vesicles at Golgi hotspots through kinesin-myosin interactions Stephanie Miserey-Lenkei, Hugo Bousquet, Olena Pylypenko, Sabine Bardin, Ariane Dimitrov, Gaëlle Bressanelli, Raja Bonifay, Vincent Fraisier, Catherine Guillou, Cécile Bougeret, et al. To cite this version: Stephanie Miserey-Lenkei, Hugo Bousquet, Olena Pylypenko, Sabine Bardin, Ariane Dimitrov, et al.. Coupling fission and exit of RAB6 vesicles at Golgi hotspots through kinesin-myosin interactions. Nature Communications, Nature Publishing Group, 2017, 8 (1), pp.1254. 10.1038/s41467-017-01266- 0. hal-02349571 HAL Id: hal-02349571 https://hal.archives-ouvertes.fr/hal-02349571 Submitted on 15 Apr 2020 HAL is a multi-disciplinary open access L’archive ouverte pluridisciplinaire HAL, est archive for the deposit and dissemination of sci- destinée au dépôt et à la diffusion de documents entific research documents, whether they are pub- scientifiques de niveau recherche, publiés ou non, lished or not. The documents may come from émanant des établissements d’enseignement et de teaching and research institutions in France or recherche français ou étrangers, des laboratoires abroad, or from public or private research centers. publics ou privés. Distributed under a Creative Commons Attribution| 4.0 International License ARTICLE DOI: 10.1038/s41467-017-01266-0 OPEN Coupling fission and exit of RAB6 vesicles at Golgi hotspots through kinesin-myosin interactions Stéphanie Miserey-Lenkei1, Hugo Bousquet1, Olena Pylypenko2, Sabine Bardin1, Ariane Dimitrov1, Gaëlle Bressanelli2, Raja Bonifay2, Vincent Fraisier3, Catherine Guillou4, Cécile Bougeret5, Anne Houdusse 2, Arnaud Echard 6 & Bruno Goud1 The actin and microtubule cytoskeletons play important roles in Golgi structure and function, 1234567890 but how they are connected remain poorly known.
    [Show full text]
  • Revealing the Action Mechanisms of Dexamethasone on the Birth Weight
    ® Observational Study Medicine OPEN Revealing the action mechanisms of dexamethasone on the birth weight of infant using RNA-sequencing data of trophoblast cells ∗ Hongkai Shang, MDa, Liping Sun, BMa, Thorsten Braun, MDb, Qi Si, BMa, Jinyi Tong, MMa, Abstract Dexamethasone (DEX) could induce low birth weight of infant, and low birth weight has close associations with glucocorticoid levels, insulin resistance, hypertension, and metabolic syndrome in adulthood. This study was designed to reveal the action mechanisms of DEX on the birth weight of infant. Using quantitative real-time polymerase chain reaction (qRT-PCR), trophoblast cells of human placenta were identified and the optimum treatment time of DEX were determined. Trophoblast cells were treated by DEX (DEX group) or ethanol (control group) (each group had 3 samples), and then were performed with RNA-sequencing. Afterward, the differentially expressed genes (DEGs) were identified by R package, and their potential functions were successively enriched using DAVID database and Enrichr method. Followed by protein–protein interaction (PPI) network was constructed using Cytoscape software. Using Enrichr method and TargetScan software, the transcription factors (TFs) and micorRNAs (miRNAs) targeted the DEGs separately were predicted. Based on MsigDB database, gene set enrichment analysis (GSEA) was performed. There were 391 DEGs screened from the DEX group. Upregulated SRR and potassium voltage-gated channel subfamily J member 4(KCNJ4) and downregulated GALNT1 separately were enriched in PDZ (an acronym of PSD-95, Dlg, and ZO-1) domain binding and Mucin type O-glycan biosynthesis. In the PPI network, CDK2 and CDK4 had higher degrees. TFs ATF2 and E2F4 and miRNA miR-16 were predicted for the DEGs.
    [Show full text]