Increased Microvascular Permeability in Mice Lacking Epac1 (Rapgef3)

Total Page:16

File Type:pdf, Size:1020Kb

Increased Microvascular Permeability in Mice Lacking Epac1 (Rapgef3) Increased microvascular permeability in mice lacking Epac1 (Rapgef3) Kopperud, R. K.; Rygh, C. Brekke; Karlsen, T. V.; Krakstad, C.; Kleppe, R.; Hoivik, E. A.; Bakke, M.; Tenstad, O.; Selheim, F.; Lidén, Å.; Madsen, Lise; Pavlin, T.; Taxt, T.; Kristiansen, Karsten; Curry, F. -R. E.; Reed, R. K.; Døskeland, S. O. Published in: Acta Physiologica DOI: 10.1111/apha.12697 Publication date: 2017 Document version Publisher's PDF, also known as Version of record Document license: CC BY-NC-ND Citation for published version (APA): Kopperud, R. K., Rygh, C. B., Karlsen, T. V., Krakstad, C., Kleppe, R., Hoivik, E. A., Bakke, M., Tenstad, O., Selheim, F., Lidén, Å., Madsen, L., Pavlin, T., Taxt, T., Kristiansen, K., Curry, F. -R. E., Reed, R. K., & Døskeland, S. O. (2017). Increased microvascular permeability in mice lacking Epac1 (Rapgef3). Acta Physiologica, 219(2), 441-452. https://doi.org/10.1111/apha.12697 Download date: 30. Sep. 2021 Acta Physiol 2017, 219, 441–452 Increased microvascular permeability in mice lacking Epac1 (Rapgef3) † § R. K. Kopperud,1,2,*, C. Brekke Rygh,1,*, T. V. Karlsen,1 C. Krakstad,1 R. Kleppe,1 † ¶ ‡ E. A. Hoivik,1, M. Bakke,1 O. Tenstad,1 F. Selheim,1 A. Liden, 1, L. Madsen,1,3, T. Pavlin,1 T. Taxt,1 K. Kristiansen,3 F.-R. E. Curry,4 R. K. Reed1,2 and S. O. Døskeland1 1 Department of Biomedicine, University of Bergen, Bergen, Norway 2 Centre for Cancer Biomarkers (CCBIO), University of Bergen, Bergen, Norway 3 Department of Biology, University of Copenhagen, Copenhagen, Denmark 4 Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, CA, USA Received 18 February 2016, Abstract revision requested 15 March Aim: Maintenance of the blood and extracellular volume requires tight con- 2016, trol of endothelial macromolecule permeability, which is regulated by cAMP revision received 12 April 2016, signalling. This study probes the role of the cAMP mediators rap guanine accepted 14 April 2016 Correspondence: S. O. nucleotide exchange factor 3 and 4 (Epac1 and Epac2) for in vivo control of Døskeland, MD, PhD, Jonas Lies microvascular macromolecule permeability under basal conditions. À À À À vei 91, N-5009 Bergen, Norway. Methods: Epac1 / and Epac2 / C57BL/6J mice were produced and E-mail: [email protected] compared with wild-type mice for transvascular flux of radio-labelled albumin in skin, adipose tissue, intestine, heart and skeletal muscle. The See Editorial Commentary: transvascular leakage was also studied by dynamic contrast-enhanced Waschke, J. Epac1 – a tonic stabilizer of the endothelial magnetic resonance imaging (DCE-MRI) using the MRI contrast agent barrier. Acta Physiol (Oxf) 219, Gadomer-17 as probe. À/À 339–340. Results: Epac1 mice had constitutively increased transvascular macro- molecule transport, indicating Epac1-dependent restriction of baseline per- À À *These authors contributed meability. In addition, Epac1 / mice showed little or no enhancement of equally to this work. † vascular permeability in response to atrial natriuretic peptide (ANP), Present address: Department of À/À Clinical Sciences, University of whether probed with labelled albumin or Gadomer-17. Epac2 and Bergen, Bergen, Norway wild-type mice had similar basal and ANP-stimulated clearances. ‡ À/À Present address: National Ultrastructure analysis revealed that Epac1 microvascular interendothe- Institute of Nutrition Research lial junctions had constitutively less junctional complex. (NIFES), Bergen, Norway § Conclusion: Epac1 exerts a tonic inhibition of in vivo basal microvascular Present address: Bergen Univer- permeability. The loss of this tonic action increases baseline permeability, sity College, Bergen, Norway ¶ Present address: Galderma presumably by reducing the interendothelial permeability resistance. Part Nordic AB, Uppsala, Sweden of the action of ANP to increase permeability in wild-type microvessels may involve inhibition of the basal Epac1-dependent activity. The copyright line for this article Keywords atrial natriuretic peptide, cAMP, endothelial junction, Epac was changed on 11th August, 2016 deletion (mouse), microvascular permeability (in vivo), Rapgef. after original online publication. The primary aim of the study was to investigate cAMP-dependent protein kinase (PKA) only, but the whether the cAMP-stimulated Rap activator Epac likely involvement of Epac was demonstrated using contributes to the tonic control of microvessel base- Epac-activating cAMP analogues (Christensen et al., line permeability in major organs of the mouse. The 2003) that lowered the permeability of monolayers of well-recognized endothelial permeability lowering human embryonic vascular endothelial cells and atten- effect of cAMP was first thought to be mediated by uated the response to inflammatory conditions such © 2016 The Authors. Acta Physiologica published by John Wiley & Sons Ltd on behalf of Scandinavian Physiological Society, doi: 10.1111/apha.12697 441 This is an open access article under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made. Microvessel permeability in Epac1KO mice · R K Kopperud et al. Acta Physiol 2017, 219, 441–452 as exposure to thrombin (Cullere et al. 2005). A role et al. 2010, Lin et al. 2011), and also shown that it is of Epac1 and its target, the small GTPase Rap, to attenuated when cAMP is elevated, and proposed that modulate inflammation-associated endothelial hyper- ANP might increase permeability via decreased Epac permeability has been confirmed in numerous in vitro activity (Lin et al. 2011). We show here that Epac1- studies and noted in some in situ perfusion studies of deficient mice, but not Epac2-deficient mice have an accessible microvessels, although the relative roles of attenuated response to ANP. PKA, cGMP-dependent protein kinase (PKG) and Epac in different organ vascular beds and under dif- Materials and methods ferent experimental conditions still remain uncertain (for recent reviews, see Spindler et al. 2010, Surapi- Ethical approval sitchat & Beavo 2011, Kuhn 2012, Curry & Adam- son 2013). The animal experiments were approved by the Norwe- The role of Epac to maintain stable baseline perme- gian Animal Research Authority and conducted accord- ability in intact microvessels under non-inflammatory ing to the European Convention for the Protection of conditions is less well studied. A major reason is the Vertebrates Used for Scientific Purposes, Norway. lack of ways to reliably inhibit Epac and other cAMP- dependent pathways pharmacologically in vivo (Spind- Materials ler & Waschke 2011), in part due to rapid metabolism and renal clearance of cAMP analogues used to modu- The anti-Epac1 antibody was kindly provided by late Epac and PKA activity (Kleppe et al. 2015). J. Bos, University of Utrecht, the Netherlands. The À À In this study, Epac-deficient mice (Epac1 / or anti-Epac2 antibody (5B1) was from Cell Signaling À À Epac2 / ) were generated to elucidate the potential technology (www.cellsignal.com). The anti-CD31 anti- in vivo role of Epac in regulation of baseline macro- body (DIA-310) was from Dianova, Hamburg, Ger- molecule flux across microvascular endothelium. many. ANP and rolipram were from Sigma-Aldrich À À À À Others have reported that Epac1 / and Epac2 / mice (St. Louis, MO, USA). Gadomer-17 was from in vivo À À have only subtle phenotypes. Epac2 / mice have a Contrast GmbH, Schering Pharma (Berlin, Germany). slightly perturbed pathway of pancreatic beta cell insu- Human serum albumin (HSA) was labelled with 131I À À lin exocytosis (Shibasaki et al. 2007), while Epac1 / or 125I (from Perkin Elmer, Waltham, MA, USA) by mice have been reported to gain slightly more (Kai et al. the iodogen method, as previously described (Curry 2013) or slightly less (Yan et al. 2013) body weight on et al. 2010). Any free iodide was removed by spinning both low- and high-fat diets, and be less susceptible to twice on a 3-kDa cut-off column (Microcon filter; rickettsial invasion (Gong et al. 2013). Millipore, Bedford, MA, USA) prior to experiments. We report here an increased basal microvascular The labelled HSA eluted like native HSA upon anionic transvascular albumin flux in skin, skeletal muscle and exchange and high-resolution size exclusion chro- À À À À intestines of Epac1 / , but not Epac2 / mice. matography with online gamma radiation detection in Dynamic contrast-enhanced magnetic resonance imag- series with UV detector. Low molecular weight ing (DCE-MRI) studies of the masseter muscle using radioactivity accounted for <0.3% of the total activity. Gadomer-17 demonstrated transvascular flux of the À À À À high molecular weight MR contrast agent, which con- Generation of Epac1 / and Epac2 / knock-out À À À À À À firmed that Epac1 / mice had constitutively increased models. Epac1 / and Epac2 / mice were generated microvascular permeability to macromolecules. Trans- by genomic deletion of exons 7–10 in Rapgef3 (encod- mission electron microscopy was used to compare the ing Epac1) and exons 12–13 of Rapgef4 (encoding ultrastructure of the endothelial junctions in masseter Epac2) encoding the cAMP-binding domains of both À À muscle biopsies from basal state in Wt and Epac1 / proteins (Fig. 1a,d) by use of the Cre–LoxP system. An mice. They revealed decrease of junctional complex in additional frame shift codon was introduced in exon À À the interendothelial
Recommended publications
  • Protein Interaction Network of Alternatively Spliced Isoforms from Brain Links Genetic Risk Factors for Autism
    ARTICLE Received 24 Aug 2013 | Accepted 14 Mar 2014 | Published 11 Apr 2014 DOI: 10.1038/ncomms4650 OPEN Protein interaction network of alternatively spliced isoforms from brain links genetic risk factors for autism Roser Corominas1,*, Xinping Yang2,3,*, Guan Ning Lin1,*, Shuli Kang1,*, Yun Shen2,3, Lila Ghamsari2,3,w, Martin Broly2,3, Maria Rodriguez2,3, Stanley Tam2,3, Shelly A. Trigg2,3,w, Changyu Fan2,3, Song Yi2,3, Murat Tasan4, Irma Lemmens5, Xingyan Kuang6, Nan Zhao6, Dheeraj Malhotra7, Jacob J. Michaelson7,w, Vladimir Vacic8, Michael A. Calderwood2,3, Frederick P. Roth2,3,4, Jan Tavernier5, Steve Horvath9, Kourosh Salehi-Ashtiani2,3,w, Dmitry Korkin6, Jonathan Sebat7, David E. Hill2,3, Tong Hao2,3, Marc Vidal2,3 & Lilia M. Iakoucheva1 Increased risk for autism spectrum disorders (ASD) is attributed to hundreds of genetic loci. The convergence of ASD variants have been investigated using various approaches, including protein interactions extracted from the published literature. However, these datasets are frequently incomplete, carry biases and are limited to interactions of a single splicing isoform, which may not be expressed in the disease-relevant tissue. Here we introduce a new interactome mapping approach by experimentally identifying interactions between brain-expressed alternatively spliced variants of ASD risk factors. The Autism Spliceform Interaction Network reveals that almost half of the detected interactions and about 30% of the newly identified interacting partners represent contribution from splicing variants, emphasizing the importance of isoform networks. Isoform interactions greatly contribute to establishing direct physical connections between proteins from the de novo autism CNVs. Our findings demonstrate the critical role of spliceform networks for translating genetic knowledge into a better understanding of human diseases.
    [Show full text]
  • Protein-Coding Variants Implicate Novel Genes Related to Lipid Homeostasis
    bioRxiv preprint doi: https://doi.org/10.1101/352674; this version posted June 30, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. 1 PROTEIN-CODING VARIANTS IMPLICATE NOVEL GENES RELATED TO LIPID HOMEOSTASIS 2 CONTRIBUTING TO BODY FAT DISTRIBUTION 3 Anne E Justice¥,1,2, Tugce Karaderi¥,3,4, Heather M Highland¥,1,5, Kristin L Young¥,1, Mariaelisa Graff¥,1, 4 Yingchang Lu¥,6,7,8, Valérie Turcot9, Paul L Auer10, Rebecca S Fine11,12,13, Xiuqing Guo14, Claudia 5 Schurmann7,8, Adelheid Lempradl15, Eirini Marouli16, Anubha Mahajan3, Thomas W Winkler17, Adam E 6 Locke18,19, Carolina Medina-Gomez20,21, Tõnu Esko11,13,22, Sailaja Vedantam11,12,13, Ayush Giri23, Ken Sin 7 Lo9, Tamuno Alfred7, Poorva Mudgal24, Maggie CY Ng24,25, Nancy L Heard-Costa26,27, Mary F Feitosa28, 8 Alisa K Manning11,29,30 , Sara M Willems31, Suthesh Sivapalaratnam30,32,33, Goncalo Abecasis18, Dewan S 9 Alam34, Matthew Allison35, Philippe Amouyel36,37,38, Zorayr Arzumanyan14, Beverley Balkau39, Lisa 10 Bastarache40, Sven Bergmann41,42, Lawrence F Bielak43, Matthias Blüher44,45, Michael Boehnke18, Heiner 11 Boeing46, Eric Boerwinkle47,48, Carsten A Böger49, Jette Bork-Jensen50, Erwin P Bottinger7, Donald W 12 Bowden24,25,51, Ivan Brandslund52,53, Linda Broer21, Amber A Burt54, Adam S Butterworth55,56, Mark J 13 Caulfield16,57, Giancarlo Cesana58, John C Chambers59,60,61,62,63, Daniel I Chasman11,64,65,66, Yii-Der Ida 14 Chen14, Rajiv Chowdhury55, Cramer Christensen67,
    [Show full text]
  • Epac2a-Null Mice Exhibit Obesity-Prone Nature More Susceptible to Leptin Resistance
    OPEN International Journal of Obesity (2017) 41, 279–288 www.nature.com/ijo ORIGINAL ARTICLE Epac2a-null mice exhibit obesity-prone nature more susceptible to leptin resistance M Hwang1,5,YGo2,5,6, J-H Park1, S-K Shin1, SE Song1, B-C Oh3, S-S Im1, I Hwang1, YH Jeon2, I-K Lee2, S Seino4 and D-K Song1 BACKGROUND: The exchange protein directly activated by cAMP (Epac), which is primarily involved in cAMP signaling, has been known to be essential for controlling body energy metabolism. Epac has two isoforms: Epac1 and Epac2. The function of Epac1 on obesity was unveiled using Epac1 knockout (KO) mice. However, the role of Epac2 in obesity remains unclear. METHODS: To evaluate the role of Epac2 in obesity, we used Epac2a KO mice, which is dominantly expressed in neurons and endocrine tissues. Physiological factors related to obesity were analyzed: body weight, fat mass, food intake, plasma leptin and adiponectin levels, energy expenditure, glucose tolerance, and insulin and leptin resistance. To determine the mechanism of Epac2a, mice received exogenous leptin and then hypothalamic leptin signaling was analyzed. RESULTS: Epac2a KO mice appeared to have normal glucose tolerance and insulin sensitivity until 12 weeks of age, but an early onset increase of plasma leptin levels and decrease of plasma adiponectin levels compared with wild-type mice. Acute leptin injection revealed impaired hypothalamic leptin signaling in KO mice. Consistently, KO mice fed a high-fat diet (HFD) were significantly obese, presenting greater food intake and lower energy expenditure. HFD-fed KO mice were also characterized by greater impairment of hypothalamic leptin signaling and by weaker leptin-induced decrease in food consumption compared with HFD-fed wild-type mice.
    [Show full text]
  • DLL1- and DLL4-Mediated Notch Signaling Is Essential for Adult Pancreatic Islet
    Page 1 of 41 Diabetes DLL1- and DLL4-mediated Notch signaling is essential for adult pancreatic islet homeostasis (running title –Role of Delta ligands in adult pancreas) Marina Rubey1,2,6*, Nirav Florian Chhabra1,2*, Daniel Gradinger1,2,7, Adrián Sanz-Moreno1, Heiko Lickert2,4,5, Gerhard K. H. Przemeck1,2, Martin Hrabě de Angelis1,2,3** 1 Helmholtz Zentrum München, Institute of Experimental Genetics and German Mouse Clinic, Neuherberg, Germany 2 German Center for Diabetes Research (DZD), Neuherberg, Germany 3 Chair of Experimental Genetics, Centre of Life and Food Sciences, Weihenstephan, Technische Universität München, Freising, Germany 4 Helmholtz Zentrum München, Institute of Diabetes and Regeneration Research and Institute of Stem Cell Research, Neuherberg, Germany 5 Technische Universität München, Medical Faculty, Munich, Germany 6 Present address Marina Rubey: WMC Healthcare GmbH, Munich, Germany 7 Present address Daniel Gradinger: PSI CRO AG, Munich, Germany *These authors contributed equally **Corresponding author: Prof. Dr. Martin Hrabě de Angelis, Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Experimental Genetics, Ingolstädter Landstr.1, 85764 Neuherberg, Germany. Phone: +49-89-3187-3502. Fax: +49- 89-3187-3500. E-mail address: [email protected] Word count – 4088 / Figures – 7 Diabetes Publish Ahead of Print, published online February 6, 2020 Diabetes Page 2 of 41 Abstract Genes of the Notch signaling pathway are expressed in different cell types and organs at different time points during embryonic development and adulthood. The Notch ligand Delta- like 1 (DLL1) controls the decision between endocrine and exocrine fates of multipotent progenitors in the developing pancreas, and loss of Dll1 leads to premature endocrine differentiation.
    [Show full text]
  • Human Induced Pluripotent Stem Cell–Derived Podocytes Mature Into Vascularized Glomeruli Upon Experimental Transplantation
    BASIC RESEARCH www.jasn.org Human Induced Pluripotent Stem Cell–Derived Podocytes Mature into Vascularized Glomeruli upon Experimental Transplantation † Sazia Sharmin,* Atsuhiro Taguchi,* Yusuke Kaku,* Yasuhiro Yoshimura,* Tomoko Ohmori,* ‡ † ‡ Tetsushi Sakuma, Masashi Mukoyama, Takashi Yamamoto, Hidetake Kurihara,§ and | Ryuichi Nishinakamura* *Department of Kidney Development, Institute of Molecular Embryology and Genetics, and †Department of Nephrology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan; ‡Department of Mathematical and Life Sciences, Graduate School of Science, Hiroshima University, Hiroshima, Japan; §Division of Anatomy, Juntendo University School of Medicine, Tokyo, Japan; and |Japan Science and Technology Agency, CREST, Kumamoto, Japan ABSTRACT Glomerular podocytes express proteins, such as nephrin, that constitute the slit diaphragm, thereby contributing to the filtration process in the kidney. Glomerular development has been analyzed mainly in mice, whereas analysis of human kidney development has been minimal because of limited access to embryonic kidneys. We previously reported the induction of three-dimensional primordial glomeruli from human induced pluripotent stem (iPS) cells. Here, using transcription activator–like effector nuclease-mediated homologous recombination, we generated human iPS cell lines that express green fluorescent protein (GFP) in the NPHS1 locus, which encodes nephrin, and we show that GFP expression facilitated accurate visualization of nephrin-positive podocyte formation in
    [Show full text]
  • Selective Small-Molecule EPAC Activators
    Heriot-Watt University Research Gateway Selective small-molecule EPAC activators Citation for published version: Luchowska-Staska, U, Morgan, D, Yarwood, SJ & Barker, G 2019, 'Selective small-molecule EPAC activators', Biochemical Society Transactions, vol. 47, no. 5, pp. 1415-1427. https://doi.org/10.1042/BST20190254 Digital Object Identifier (DOI): 10.1042/BST20190254 Link: Link to publication record in Heriot-Watt Research Portal Document Version: Publisher's PDF, also known as Version of record Published In: Biochemical Society Transactions Publisher Rights Statement: © 2019 The Author(s). This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY). General rights Copyright for the publications made accessible via Heriot-Watt Research Portal is retained by the author(s) and / or other copyright owners and it is a condition of accessing these publications that users recognise and abide by the legal requirements associated with these rights. Take down policy Heriot-Watt University has made every reasonable effort to ensure that the content in Heriot-Watt Research Portal complies with UK legislation. If you believe that the public display of this file breaches copyright please contact [email protected] providing details, and we will remove access to the work immediately and investigate your claim. Download date: 30. Sep. 2021 Biochemical Society Transactions (2019) https://doi.org/10.1042/BST20190254 Review Article Selective small-molecule EPAC activators Urszula Luchowska-Stanská 1, David Morgan2, Stephen J. Yarwood1 and Graeme Barker2 1 2 Institute of Biological Chemistry, Biophysics, and Bioengineering, Heriot-Watt University, Edinburgh EH14 4AS, U.K.; Institute of Chemical Sciences, Heriot-Watt University, Downloaded from https://portlandpress.com/biochemsoctrans/article-pdf/doi/10.1042/BST20190254/857286/bst-2019-0254c.pdf by Heriot-Watt University user on 21 October 2019 Edinburgh EH14 4AS, U.K.
    [Show full text]
  • Loss of EPAC2 Results in Altered Synaptic Composition of Dendritic
    bioRxiv preprint doi: https://doi.org/10.1101/526772; this version posted January 21, 2019. 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 4.0 International license. Loss of EPAC2 results in altered synaptic composition of dendritic spines and excitatory and inhibitory balance Kelly A. Jonesa, Michiko Sumyiab,c, Kevin M. Woolfreya, Deepak P. Srivastavaa,b,c§, and Peter Penzesa,d§ aDepartment of Physiology, bDepartment of Basic and Clinical Neuroscience, Institute of Psychiatry Psychology and Neuroscience, King's College London, 125 Coldharbour Lane, London, SE5 8NU, UK; cMRC Centre for Neurodevelopmental Disorders, King’s College London, London, UK; dDepartment of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 E. Chicago Avenue, Chicago, IL 60611 USA § = corresponding authors: Peter Penzes, Department of Physiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA, e-mail: p- [email protected]; or Deepak Srivastava, Department of Basic and Clinical Neuroscience, King's College London, London, SE5 9RT, UK, e-mail: [email protected] Keywords: EPAC2, dendritic spines, dendritic arborization, synaptic plasticity, autism spectrum disorders, excitatory and inhibitory balance bioRxiv preprint doi: https://doi.org/10.1101/526772; this version posted January 21, 2019. 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 4.0 International license.
    [Show full text]
  • Epac2 (A-7): Sc-28326
    SAN TA C RUZ BI OTEC HNOL OG Y, INC . Epac2 (A-7): sc-28326 BACKGROUND STORAGE 3',5' cyclic adenosine monophosphate (cAMP)-regulated guanine nucleotide Store at 4° C, **DO NOT FREEZE**. Stable for one year from the date of exchange factors Epac1 (cAMP-GEFI) and Epac2 (cAMP-GEFII) activate the shipment. Non-hazardous. No MSDS required. Ras family GTPases Rap 1 and Rap 2 by promoting GTP binding in a cAMP- dependent manner. Eukaryotic cAMP is a second messenger that induces DATA physiological responses such as gene expression, growth, differentiation, secretion and neurotransmission. Human EPAC2 contains at least 31 exons ABC and maps to chromosome 2q31.1. The 4.4-kb Epac2 transcript is prominent in brain and adrenal gland. Within the brain, expression is strong in cortex, 132K– occipital pole, frontal lobe, temporal lobe, amygdala, putamen, hippocampus < Epac2 and cerebellum. 90K– CHROMOSOMAL LOCATION Genetic locus: RAPGEF4 (human) mapping to 2q31.1; Rapgef4 (mouse) Epac2 (A-7): sc-28326. Western blot analysis of Epac2 Epac2 (A-7): sc-28326. Immunoperoxidase staining of mapping to 2 C3. expression in rat cerebellum (A), mouse brain (B) and formalin fixed, paraffin-embedded human skeletal rat brain (C) tissue extracts. muscle tissue showing cytoplasmic staining of myo- cytes. Kindly provided by The Swedish Human Protein SOURCE Atlas (HPA) program. Epac2 (A-7) is a mouse monoclonal antibody raised against amino acids 1-220 of Epac2 of human origin. SELECT PRODUCT CITATIONS 1. Wang, Z., et al. 2006. Rap1-mediated activation of extracellular signal- PRODUCT regulated kinases by cyclic AMP is dependent on the mode of Rap1 acti vation.
    [Show full text]
  • Small Gtpases of the Ras and Rho Families Switch On/Off Signaling
    International Journal of Molecular Sciences Review Small GTPases of the Ras and Rho Families Switch on/off Signaling Pathways in Neurodegenerative Diseases Alazne Arrazola Sastre 1,2, Miriam Luque Montoro 1, Patricia Gálvez-Martín 3,4 , Hadriano M Lacerda 5, Alejandro Lucia 6,7, Francisco Llavero 1,6,* and José Luis Zugaza 1,2,8,* 1 Achucarro Basque Center for Neuroscience, Science Park of the Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), 48940 Leioa, Spain; [email protected] (A.A.S.); [email protected] (M.L.M.) 2 Department of Genetics, Physical Anthropology, and Animal Physiology, Faculty of Science and Technology, UPV/EHU, 48940 Leioa, Spain 3 Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Granada, 180041 Granada, Spain; [email protected] 4 R&D Human Health, Bioibérica S.A.U., 08950 Barcelona, Spain 5 Three R Labs, Science Park of the UPV/EHU, 48940 Leioa, Spain; [email protected] 6 Faculty of Sport Science, European University of Madrid, 28670 Madrid, Spain; [email protected] 7 Research Institute of the Hospital 12 de Octubre (i+12), 28041 Madrid, Spain 8 IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain * Correspondence: [email protected] (F.L.); [email protected] (J.L.Z.) Received: 25 July 2020; Accepted: 29 August 2020; Published: 31 August 2020 Abstract: Small guanosine triphosphatases (GTPases) of the Ras superfamily are key regulators of many key cellular events such as proliferation, differentiation, cell cycle regulation, migration, or apoptosis. To control these biological responses, GTPases activity is regulated by guanine nucleotide exchange factors (GEFs), GTPase activating proteins (GAPs), and in some small GTPases also guanine nucleotide dissociation inhibitors (GDIs).
    [Show full text]
  • Anxiety and Depression with Neurogenesis Defects in Exchange
    OPEN Citation: Transl Psychiatry (2016) 6, e881; doi:10.1038/tp.2016.129 www.nature.com/tp ORIGINAL ARTICLE Anxiety and depression with neurogenesis defects in exchange protein directly activated by cAMP 2-deficient mice are ameliorated by a selective serotonin reuptake inhibitor, Prozac L Zhou1,10,SLMa2,10, PKK Yeung1,3, YH Wong4,5, KWK Tsim5,6,KFSo1,7,8,9, LCW Lam2 and SK Chung1,3,7 Intracellular cAMP and serotonin are important modulators of anxiety and depression. Fluoxetine, a selective serotonin reuptake inhibitor (SSRI) also known as Prozac, is widely used against depression, potentially by activating cAMP response element-binding protein (CREB) and increasing brain-derived neurotrophic factor (BDNF) through protein kinase A (PKA). However, the role of Epac1 and Epac2 (Rap guanine nucleotide exchange factors, RAPGEF3 and RAPGEF4, respectively) as potential downstream targets of SSRI/cAMP in mood regulations is not yet clear. Here, we investigated the phenotypes of Epac1 (Epac1− / −) or Epac2 (Epac2− / −) knockout mice by comparing them with their wild-type counterparts. Surprisingly, Epac2− / − mice exhibited a wide range of mood disorders, including anxiety and depression with learning and memory deficits in contextual and cued fear-conditioning tests without affecting Epac1 expression or PKA activity. Interestingly, rs17746510, one of the three single-nucleotide polymorphisms (SNPs) in RAPGEF4 associated with cognitive decline in Chinese Alzheimer’s disease (AD) patients, was significantly correlated with apathy and mood disturbance, whereas no significant association was observed between RAPGEF3 SNPs and the risk of AD or neuropsychiatric inventory scores. To further determine the detailed role of Epac2 in SSRI/serotonin/cAMP-involved mood disorders, we treated Epac2− / − mice with a SSRI, Prozac.
    [Show full text]
  • Itga4 Cldn16 Cldn9 Cldn15 Cldn22 Ocln Esam
    Supplementary material J Med Genet Table S1. List of 263 genes included in the AGS-LEUK panel. Axonal Guidance Signaling genes as AGS and Leukocyte transvasation genes as LEUK. List of genes (AGS) List of genes (LEUK) ABLIM1 CLDN11 ACTR3 MMP14 ADAM11 MMP15 ADAM23 CTNNA1 ADAMTS1 ENSG00000130396 ADAMTS4 CLDN6 ADAMTS9 MMP24 ARHGEF15 ARHGAP12 ARHGEF6 DLC1 ARPC1B TIMP2 BDNF RAPGEF3 BMP1 F11R BMP4 CLDN23 BMP6 CLDN8 BMP7 JAM3 CXCL12 CLDN3 CXCR4 ARHGAP8 DPYSL5 ICAM1 EFNA1 MMP16 EFNA5 JAM2 ENPEP CLDN7 EPHA1 TIMP3 EPHA3 VCAM1 EPHA5 CLDN5 EPHA7 MSN EPHB1 NOX3 EPHB2 ACTC1 EPHB4 VAV2 FGFR2 CLDN10 FZD1 RAP1GAP FZD10 VAV3 FZD5 MAPK10 FZD6 CTNNA2 GAB1 CDH5 GLI1 PECAM1 GLI3 CTNND1 GNA14 ITGA4 GNAI1 CLDN16 GNAO1 CLDN9 GNAS CLDN15 GNB4 CLDN22 GNG11 OCLN GNG2 ESAM Gallego-Martinez A, et al. J Med Genet 2019; 0:1–7. doi: 10.1136/jmedgenet-2019-106159 Supplementary material J Med Genet GNG7 ACTB IGF1 CYBA IRS1 CTNNB1 IRS2 MMP9 ITGA3 MAPK14 ITGB1 MAPK11 LIMK1 MAPK12 LIMK2 MAPK13 LINGO1 PRKCB LRRC4C PXN MME BCAR1 MMP11 THY1 MMP2 ARHGAP5 MRAS MYL2 MYL9 MYLPF NFATC4 RAP1A NGFR RAP1B NOTUM VASP NRP1 ACTN4 NTN3 ACTN1 NTRK2 VCL NTRK3 RAPGEF4 PAK3 ITK PAK4 VAV1 PAPPA2 PDGFA PDGFC PIK3CB PIK3R1 PLCE1 PLCH1 PLCH2 PLXNA2 PLXNB1 PLXND1 PPP3CA PRKACB PRKAR2A PRKAR2B PRKCA PRKCZ PRKD3 ROBO2 SDC2 SDCBP Gallego-Martinez A, et al. J Med Genet 2019; 0:1–7. doi: 10.1136/jmedgenet-2019-106159 Supplementary material J Med Genet SEMA3B SEMA3C SEMA3E SEMA3F SEMA4F SEMA4G SEMA5A SEMA6B SEMA6D SEMA7A SHC1 SLIT2 SLIT3 STK36 TUBA4A TUBB2B TUBB4A TUBB4B TUBB6 UNC5C UNC5D ENSG00000165197 WIPF1 WNT3 WNT5A WNT7A WNT7B NTNG1 NTNG2 LRRC4 NTN4 TRPC1 TRPC3 TRPC6 PPP3CB PPP3CC PPP3R1 NFATC2 NFATC3 PTK2 FYN RAC1 CDC42 ABLIM2 NCK1 PAK1 PAK2 Gallego-Martinez A, et al.
    [Show full text]
  • Reprogrammed Mrna Translation Drives Metabolic Response to Therapeutic Targeting of Ribosome Biogenesis
    bioRxiv preprint doi: https://doi.org/10.1101/847723; this version posted December 12, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Reprogrammed mRNA translation drives metabolic response to therapeutic targeting of ribosome biogenesis E. P. Kusnadi1,2, A. S. Trigos1,2, C. Cullinane1, D. L. Goode1,2, O. Larsson3, J. R. Devlin1,2, K. T. Chan1,2, D. P. De Souza4, M. J. McConville4,5, G. A. McArthur1,2, G. Thomas6, E. Sanij1,7, G. Poortinga1,2, R. D. Hannan1,2,5,8,9,10, K. M. Hannan5,8,†, J. Kang1,†, and R. B. Pearson1,2,5,9,†,* Affiliations 1 Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, Victoria, Australia, 3000. 2 Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia, 3010. 3 Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, 171 65 Solna, Sweden. 4 Metabolomics Australia, Bio21 Molecular Science and Biotechnology Institute, Parkville, Victoria, Australia, 3010. 5 Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, Victoria, Australia, 3010. 6 Metabolism and Cancer Group, Molecular Mechanisms and Experimental Therapy In Oncology Program, Bellvitge Biomedical Research Institute, IDIBELL, 08908 Barcelona, Spain 7 Department of Clinical Pathology, The University of Melbourne, Parkville, Victoria, Australia, 3010. 8 ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, ACT, Australia, 2601. 9 Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia, 3168. 10 School of Biomedical Sciences, University of Queensland, Brisbane, Queensland, Australia, 4072.
    [Show full text]