Retinoic acid signaling after nerve injury [Elektronische Ressource] / vorgelegt von Kirsten Schrage
84 pages
Deutsch

Découvre YouScribe en t'inscrivant gratuitement

Je m'inscris

Retinoic acid signaling after nerve injury [Elektronische Ressource] / vorgelegt von Kirsten Schrage

Découvre YouScribe en t'inscrivant gratuitement

Je m'inscris
Obtenez un accès à la bibliothèque pour le consulter en ligne
En savoir plus
84 pages
Deutsch
Obtenez un accès à la bibliothèque pour le consulter en ligne
En savoir plus

Description

“Retinoic acid signaling after nerve injury” Von der Fakultät für Mathematik, Informatik und Naturwissenschaften der Rheinisch-Westfälischen Technischen Hochschule Aachen zur Erlangung des akademischen Grades einer Doktorin der Naturwissenschaften genehmigte Dissertation vorgelegt von Diplom-Biologin Kirsten Schrage aus Hagen (NRW) Berichter: PD DR. Jörg Mey Prof. Dr. Hermann Wagner Tag der mündlichen Prüfung: 01.12.2005 Diese Dissertation ist auf den Internetseiten der Hochschulbibliothek online verfügbar. Table of contents 1 Introduction 1.1 Peripheral nerve lesions – Axon - Schwann cell interactions 1 1.2 Retinoic acid and axonal regeneration 3 1.3 CNS lesions – Spinal cord injuries 5 1.4 Retinoic acid signaling after spinal cord injury 8 1.5 The retinoic acid signaling system 8 1.6 Goals 11 2 Schwann cells are targets of retinoic acid signaling 2.1 Abstract 12 2.2 Materials and methods 13 2.2.1 Preparation of Schwann cell primary cultures from newborn rat sciatic nerves 2.2.2 Western blotting 14 2.2.3 Determination of protein distribution 16 2.2.4 RNase Protection Assay 17 2.2.5 Cytokine treatment 19 2.3 Results 2.3.1 Molecules of the retinoic acid signaling cascade are present in Schwann cell primary cultures of newborn rats 21 2.3.

Sujets

Informations

Publié par
Publié le 01 janvier 2005
Nombre de lectures 59
Langue Deutsch
Poids de l'ouvrage 1 Mo

Extrait

“Retinoic acid signaling after nerve injury”
Von der Fakultät für Mathematik, Informatik und Naturwissenschaften der Rheinisch-Westfälischen Technischen Hochschule Aachen zur Erlangung des akademischen Grades einer Doktorin der Naturwissenschaften genehmigte Dissertation vorgelegt von Diplom-Biologin Kirsten Schrage aus Hagen (NRW) Berichter: PD DR. Jörg Mey Prof. Dr. Hermann Wagner Tag der mündlichen Prüfung: 01.12.2005 Diese Dissertation ist auf den Internetseiten der Hochschulbibliothek online verfügbar.
Table of contents 1 Introduction 1.1 Peripheral nerve lesions – Axon - Schwann cell interactions 1.2 Retinoic acid and axonal regeneration 1.3 CNS lesions – Spinal cord injuries 1.4 Retinoic acid signaling after spinal cord injury 1.5 The retinoic acid signaling system 1.6 Goals 2 Schwann cells are targets of retinoic acid signaling 2.1 Abstract 2.2 Materials and methods 2.2.1 Preparation of Schwann cell primary cultures from newborn rat sciatic nerves 2.2.2 Western blotting 2.2.3 Determination of protein distribution 2.2.4 RNase Protection Assay 2.2.5 Cytokine treatment 2.3 Results2.3.1 Molecules of the retinoic acid signaling cascade are present in Schwann cell primary cultures of newborn rats 2.3.2 RA treatment induces translocation of retinoid X receptors and CRABP-II into the nucleus 2.3.3 Autoregulatory functions of retinoid signal transduction in Schwann cells 2.3.4 Cytokine expression in Schwann cells 2.3.5 Molecular mechanisms of RA-/cytokine interactions in Schwann cells 2.4 Discussion 2.4.1 Intracellular (trans-)location of RA signaling components 2.4.2 Autoregulatory functions of RA 2.4.3 RA – cytokine interactions
1 3 5 8 8 11
12 13
13 14 16 17 19
21
22
25 27
29
33 35 36
3
3.1 3.2
3.3
3.4
4 5 6
Reactive microglia/macrophages and neurons are targets of retinoic acid signaling after spinal cord injury Abstract Materials and methods 3.2.1 Animal experimentation 3.2.2 Western blotting 3.2.3 Immunohistochemistry 3.2.4 Quantification of the intracellular localization of retinoid receptors Results 3.3.1 Behavioral effects of spinal cord contusion 3.3.2 Morphological changes after spinal cord injury 3.3.3 RARαand all RXRs are present in the rat spinal cord 3.3.4 SCI causes a small reduction in the amount of retinoid receptor proteins 3.3.5 Cellular distribution of retinoid receptors in the adult spinal cord 3.3.6 Nuclear localization of retinoid receptors in reactive microglia/macrophages 3.3.7 Retinoid receptor expression in neurons 3.3.8 Reactive astrocytes in the glial scar are immunoreactive for retinoid receptors 3.3.9 Retinoid receptor distribution in oligodendrocytes Discussion 3.4.1 Cellular targets of retinoic acid signaling after spinal cord contusion 3.4.2 Intracellular translocation of retinoid receptors 3.4.3 Possible physiological functions of RA after spinal cord injury
General discussion
Summary
Bibliography
40
41 42 42 43
44 44 46
46 48
50 52
52 55
56 5758
60
62
63
Abbreviations
ANOVA APS Ara CBBBBCABDNFBMPBSAcAMPCNPase CNSCNTFCOXCRABP CRBPCyp26DAPIDEPC DMEM DMSODNA dpo DRG ECL ELISAerbBErkFCSGACUGAPGAPDH GDNFGFAPGFPGM-CSF gpHeLa cells HEPES HRP IFNIg ILIL-6Rα JakLIFLtMAGMAP2MAPK
analysis of variance ammoniumpersulfate cytosine arabinosid Basso, Beattie, Bresnahan bicinchoninic acid brain-derived neurotrophic factor bone morphogenetic protein bovine serum albumin cyclic adenosine mono-phosphate 2´,3´-cyclic nucleotide 3´-phosphodiesterase central nervous system ciliary neurotrophic factor cyclooxygenase cellular retinoic acid binding protein cellular retinol binding protein cytochrome P450-oxidase, number 26 4’,6-diamidino-2-phenylindole dihydrochloride diethyl-pyrocarbonat Dulbecco´s Modified Eagle´s Medium dimethyl sulfoxide desoxyribonucleic acid days post operation dorsal root ganglia enhanced chemiluminiscence Enzyme-Linked Immunosorbent Assay erythroblastosis gene B extracellular-signal-regulated kinases fetal calf serum guanine, adenine, cytosine, uracil growth associated protein glycerinaldehyde-3-phosphate dehydrogenase glial cell line-derived neurotrophic factor glial fibrillary acidic protein green fluorescent protein granulocyte macrophage colony stimulating factor glycoprotein cells from the cervix of Henrietta Lacks N-2-Hydroxyethylpiperazine-N´-2ethanesulfonic acid horseradish peroxidase interferon immune globuline interleukin interleukin-6 receptorαJanus kinase leukemia inhibitory factor lymphotoxin myelin-associated glycoprotein microtubule associated protein 2 mitogen-activated protein kinase
MIF NC NCAM N-CoRNF-κBNGFNGFINGS NLSNTNYUOD P0 p75PBSPC12PFAPKAPMSFPNS PPAR RARALDH RARRARERasRNA ROLDH RPA RT RXRSCISDS-PAGE SEM SMADSMRTSOCSSSC cells StatTEMEDΤGFβTNFTRTrkVDR
macrophage inhibitory factor nitrocellulose neural cell adhesion molecule nuclear receptor corepressor nuclear factor-kappa B nerve growth factor nerve growth factor-inducible normal goat serum nuclear localization sequence neurotrophin New York University optic density myelin protein zero protein 75 phosphate buffered saline rat phaeochromocytomapara-formaldehyde cAMP-dependent protein kinase phenylmethylsulfonylfluorid peripheral nervous system peroxisomal proliferator-activated receptor retinoic acid retinaldehyde dehydrogenase retinoic acid receptors retinoic acid responsive elements rat sarcoma viral oncogene homolog ribonucleic acid retinol dehydrogenase ribonuclease protection assay room temperature retinoid X receptors spinal cord injury sodium dodecyl sulfate – polyacrylamide gel electrophoresis standard error of mean vertebrate homologues ofSmaandMadofXenopussilencing mediator for retinoid and thyroid hormone receptors suppressors of cytokine signaling stromal stem cells signal transducer and activator of transcriptionN,N,N´,N´-tetramethylethylenediaminetransforming growth factorβtumor necrosis factor thyroid hormone receptor tyrosin kinase vitamin D receptor
1
1 Introduction 1.1 Peripheral nerve lesions - Axon-Schwann cell interactions Neurons of the peripheral nervous system (PNS) are able to regenerate after nerve injury. This is in contrast to neurons of the central nervous system (CNS; Ramón y Cajal, 1914). Crushing or cutting a peripheral nerve triggers a well characterized cascade of cellular and molecular events, collectively described as Wallerian degeneration, throughout the distal extent of that nerve. This process involves a number of phases in which the distal portions of all affected axons, irrespective of size and modality, degenerate and disappear. Within two days associated myelin sheaths are degraded. Recruited macrophages invade the distal segment and
remove axonal and myelin debris. Schwann cells proliferate showing a maximum after 3 – 4 days leading to steadily increasing numbers up to day 14 after lesion (Oaklander, 1988). The de-differentiated daughter cells line up within each basal lamina tube to form the bands of Büngner and become permissive for regeneration. Proximal to the site of injury, axons give rise to one or more sprouts, each of which is tipped by a growth cone (Ramón y Cajal, 1928). For axonal regeneration to be successful, growth cones must first reach the distal nerve stump, which is even possible when they have to cross a gap between the proximal and distal nerve stumps. Growth cones then contact the basal lamina, and Schwann cells that re-differentiate after a second burst of proliferation induced by ingrowing axons enwrap the ingrowing axon forming a new myelin sheath (Fawcett and Keynes, 1990; Fig. 1.1). Schwann cells have long been known to be central players in Wallerian degeneration and subsequent regeneration. A differentiated axon-associated Schwann cell is a polarized cell with a basal, abaxonal surface apposed to a basal lamina (which it secretes), an apical surface apposed to an axon or group of axons, and lateral surfaces tipped with microvilli, which interdigitate with adjacent Schwann cells. Functionally, they have been defined almost entirely in terms of their reciprocal relationships with the axons they ensheathe. Axonal signals, whether acting by direct contact or diffusible molecules, regulate the expression of
many Schwann cell genes and control both proliferation and differentiation of these cells (Bolin and Shooter, 1993; Jessen and Richardson, 2001; Lemke and Chao, 1988; Maurel and Salzer, 2000; Thomson et al., 1993). The relative contributions of the Schwann cells and their basal laminae have been evaluated in vivo by comparing regeneration in cellular versus a-cellular nerves: PNS axons will regenerate also in a-cellular nerve grafts, but regeneration is better in nerves that contain living Schwann cells (Sketelj et al., 1989).
2
Fig. 1.1: Changes in peripheral nerve during Wallerian degeneration. a: Schwann cells surrounded by a basal lamina enwrap an uninjured axon.b:A few days after injury, the axon and its myelin sheath degenerate distal to the lesion site, Schwann cells proliferate and macrophages invade the distal nerve stump to phagocytose myelin debris.c: A few weeks later, the regenerating axon has been re-myelinated by Schwann cells. Jessen, 2001. After axotomy this tightly regulated relationship between axons and Schwann cells is disrupted. Acutely denervated Schwann cells downregulate transcripts of myelin-specific proteins, like myelin-associated glycoprotein (MAG), myelin basic protein, protein P0, periaxin, peripheral myelin protein 22kD, plasmolipin, and the fatty acid-binding protein P2(Gillen et al., 1997; Mitchell et al., 1990; Willison et al., 1988). They also reduce the expression of connexin 32, a gap junction protein which forms reflexive contacts within individual myelinating Schwann cells at paranodes and incisures (Arroyo and Scherer, 2000; Chandross, 1998), and caveolin-1, a putative regulator of signal transduction and/or cholesterol transport in myelinating Schwann cells (Nishio et al., 2002). De-differentiated proliferating Schwann cells upregulate gene expression of the low affinity neurotrophin receptor p75 (Heumann et al., 1987a; Heumann et al., 1987b; Taniuchi et al., 1986), GAP-43 (Curtis et al., 1992), nerve growth factor (NGF; Matsuoka et al., 1991), brain-derived neurotrophic factor (BDNF; Korsching, 1993; Meyer et al., 1992), the neuregulin receptors erbB2, erbB3 and erbB4 (Cohen et al., 1992; Li et al., 1997a; Li et al., 1998), N-CAM and L1 (Jessen et al., 1987; Martini and Schachner, 1988). They increase the expression of laminin (Kuecherer-Ehret et al., 1990), tenascin (Martini et al., 1990), fibronectin (Lefcort et al., 1992), the neuropoietic cytokines leukemia inhibitory factor (LIF) and interleukin-6 (IL-6; Kurek et al., 1996), and netrin-1, a secreted protein which influences growth cone and axon
3
guidance (Madison et al., 2000). Gene expression of the gap junction protein connexin 43, which may mediate junctional coupling in de-differentiated Schwann cells and so facilitate the rapid diffusion of signaling molecules between cells downstream of the lesion site (Chandross, 1998) and TGFβ-1 (Scherer et al., 1993) is also upregulated by proliferating Schwann cells. These changes in gene expression mean that acutely denervated Schwann cells are in a reactive, axon-responsive state in the first weeks after injury. Re-innervated Schwann cells cease dividing, downregulate expression of the molecules listed above and revert to an axon-associated phenotype. 1.2 Retinoic acid and axonal regeneration Schwann cells are the most important modulators of axonal regeneration. They are the main source of trophic factors that promote regeneration after peripheral nerve injury. It is still unknown how these trophic factors themselves are modulated. My hypothesis is that retinoic acid (RA), a transcriptional regulator of gene expression, whose importance in embryonic development is well established (Ross et al., 2000), is one of the factors that control Schwann cell reactions after nerve injury. All necessary components of the RA signal transduction pathway are detectable in mammalian peripheral nerves. Among the molecular targets that are known to be expressed by Schwann cells and regulated by RA in various cell culture systems unrelated to the nervous system (Mey, 2001) are neuropoietic cytokines, neurotrophins and
members of the transforming growth factor-β (TGFβ) family. They all belong to a diverse group of proteins with small molecular weight (6-26 kDa), secreted by all cell types that participate in traumatic interactions after nerve injury (Zhao and Schwartz, 1998), and they are known to be regulated early after sciatic nerve crush or transection (Gillen et al., 1997). The neurotrophin family consists of four members in mammals: NGF, BDNF, neurotrophin (NT)-3 and NT-4 (Ip and Yancopoulos, 1996; Korsching, 1993; Lewin and Barde, 1996; Snider, 1994). Each neurotrophin binds selectively to high-affinity neurotrophin receptors, TrkA (tyrosin kinase), TrkB and TrkC and non-selectively to the low-affinity neurotrophin receptor p75 (Ip and Yancopoulos, 1996). Upon binding their cognate neurotrophin(s), Trk receptors dimerize and are autophosphorylated via their intracellular kinase domain; this, in turn, activates various intracellular signaling pathways including Ras, phosphatidylinositol 3-kinase and phospholipase Cγ(Segal and Greenberg, 1996; Skaper and Walsh, 1998).
In mammals, the TGFβ family is comprised of three isoforms termed TGFβ1, -β2 and -β3. They initiate their cellular action by binding to heterodimeric receptors type I and type II, with intrinsic serine/threonine kinase activity (Wrana et al., 1994). The TGF receptors
  • Univers Univers
  • Ebooks Ebooks
  • Livres audio Livres audio
  • Presse Presse
  • Podcasts Podcasts
  • BD BD
  • Documents Documents