Skip to main content

Advertisement

Log in

Signaling Pathways in Reactive Astrocytes, a Genetic Perspective

  • Published:
Molecular Neurobiology Aims and scope Submit manuscript

Abstract

Reactive astrocytes are associated with a vast array of central nervous system (CNS) pathologies. The activation of astrocytes is characterized by changes in their molecular and morphological features, and depending on the type of damage can also be accompanied by inflammatory responses, neuronal damage, and in severe cases, scar formation. Although reactive astrogliosis is the normal physiological response essential for containing damage, it can also have detrimental effects on neuronal survival and axon regeneration, particularly in neurodegenerative diseases. It is believed that progressive changes in astrocytes as they become reactive are finely regulated by complex intercellular and intracellular signaling mechanisms. However, these have yet to be sorted out. Much has been learned from gain-of-function approaches in vivo and culture paradigms, but in most cases, loss-of-function genetic studies, which are a critical complementary approach, have been lacking. Understanding which signaling pathways are required to control different aspects of astrogliosis will be necessary for designing therapeutic strategies to improve their beneficial effects and limit their detrimental ones in CNS pathologies. In this article, we review recent advances in the mechanisms underlying the regulation of aspects of astrogliosis, with the main focus on the signaling pathways that have been studied using loss-of-function genetic mouse models.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

  1. Norton WT, Aquino DA, Hozumi I, Chiu FC, Brosnan CF (1992) Quantitative aspects of reactive gliosis: a review. Neurochem Res 17:877–885

    Article  PubMed  CAS  Google Scholar 

  2. Ridet JL, Malhotra SK, Privat A, Gage FH (1997) Reactive astrocytes: cellular and molecular cues to biological function. Trends Neurosci 20:570–577

    Article  PubMed  CAS  Google Scholar 

  3. Sofroniew MV (2009) Molecular dissection of reactive astrogliosis and glial scar formation. Trends Neurosci 32:638–647

    Article  PubMed  CAS  Google Scholar 

  4. Pekny M, Nilsson M (2005) Astrocyte activation and reactive gliosis. Glia 50:427–434

    Article  PubMed  Google Scholar 

  5. Sofroniew MV, Vinters HV (2010) Astrocytes: biology and pathology. Acta Neuropathol 119:7–35

    Article  PubMed  Google Scholar 

  6. Babcock AA, Kuziel WA, Rivest S, Owens T (2003) Chemokine expression by glial cells directs leukocytes to sites of axonal injury in the CNS. J Neurosci 23:7922–7930

    PubMed  CAS  Google Scholar 

  7. Buffo A, Rite I, Tripathi P, Lepier A, Colak D, Horn AP, Mori T, Gotz M (2008) Origin and progeny of reactive gliosis: a source of multipotent cells in the injured brain. Proc Natl Acad Sci USA 105:3581–3586

    Article  PubMed  CAS  Google Scholar 

  8. Tatsumi K, Takebayashi H, Manabe T, Tanaka KF, Makinodan M, Yamauchi T, Makinodan E, Matsuyoshi H, Okuda H, Ikenaka K, Wanaka A (2008) Genetic fate mapping of Olig2 progenitors in the injured adult cerebral cortex reveals preferential differentiation into astrocytes. J Neurosci Res 86:3494–3502

    Article  PubMed  CAS  Google Scholar 

  9. Burns KA, Murphy B, Danzer SC, Kuan CY (2009) Developmental and post-injury cortical gliogenesis: a genetic fate-mapping study with Nestin-CreER mice. Glia 57:1115–1129

    Article  PubMed  Google Scholar 

  10. Barnabe-Heider F, Goritz C, Sabelstrom H, Takebayashi H, Pfrieger FW, Meletis K, Frisen J (2010) Origin of new glial cells in intact and injured adult spinal cord. Cell Stem Cell 7:470–482

    Article  PubMed  CAS  Google Scholar 

  11. Dimou L, Simon C, Kirchhoff F, Takebayashi H, Gotz M (2008) Progeny of Olig2-expressing progenitors in the gray and white matter of the adult mouse cerebral cortex. J Neurosci 28:10434–10442

    Article  PubMed  CAS  Google Scholar 

  12. Kang SH, Fukaya M, Yang JK, Rothstein JD, Bergles DE (2010) NG2+ CNS glial progenitors remain committed to the oligodendrocyte lineage in postnatal life and following neurodegeneration. Neuron 68:668–681

    Article  PubMed  CAS  Google Scholar 

  13. Bush TG, Puvanachandra N, Horner CH, Polito A, Ostenfeld T, Svendsen CN, Mucke L, Johnson MH, Sofroniew MV (1999) Leukocyte infiltration, neuronal degeneration, and neurite outgrowth after ablation of scar-forming, reactive astrocytes in adult transgenic mice. Neuron 23:297–308

    Article  PubMed  CAS  Google Scholar 

  14. Faulkner JR, Herrmann JE, Woo MJ, Tansey KE, Doan NB, Sofroniew MV (2004) Reactive astrocytes protect tissue and preserve function after spinal cord injury. J Neurosci 24:2143–2155

    Article  PubMed  CAS  Google Scholar 

  15. Schneider A, Martin-Villalba A, Weih F, Vogel J, Wirth T, Schwaninger M (1999) NF-kappaB is activated and promotes cell death in focal cerebral ischemia. Nat Med 5:554–559

    Article  PubMed  CAS  Google Scholar 

  16. Bethea JR, Castro M, Keane RW, Lee TT, Dietrich WD, Yezierski RP (1998) Traumatic spinal cord injury induces nuclear factor-kappaB activation. J Neurosci 18:3251–3260

    PubMed  CAS  Google Scholar 

  17. Perez-Otano I, McMillian MK, Chen J, Bing G, Hong JS, Pennypacker KR (1996) Induction of NF-kB-like transcription factors in brain areas susceptible to kainate toxicity. Glia 16:306–315

    Article  PubMed  CAS  Google Scholar 

  18. Terai K, Matsuo A, McGeer EG, McGeer PL (1996) Enhancement of immunoreactivity for NF-kappa B in human cerebral infarctions. Brain Res 739:343–349

    Article  PubMed  CAS  Google Scholar 

  19. Terai K, Matsuo A, McGeer PL (1996) Enhancement of immunoreactivity for NF-kappa B in the hippocampal formation and cerebral cortex of Alzheimer's disease. Brain Res 735:159–168

    Article  PubMed  CAS  Google Scholar 

  20. Gabriel C, Justicia C, Camins A, Planas AM (1999) Activation of nuclear factor-kappaB in the rat brain after transient focal ischemia. Brain Res Mol Brain Res 65:61–69

    Article  PubMed  CAS  Google Scholar 

  21. Brambilla R, Bracchi-Ricard V, Hu WH, Frydel B, Bramwell A, Karmally S, Green EJ, Bethea JR (2005) Inhibition of astroglial nuclear factor kappaB reduces inflammation and improves functional recovery after spinal cord injury. J Exp Med 202:145–156

    Article  PubMed  CAS  Google Scholar 

  22. Brambilla R, Hurtado A, Persaud T, Esham K, Pearse DD, Oudega M, Bethea JR (2009) Transgenic inhibition of astroglial NF-kappa B leads to increased axonal sparing and sprouting following spinal cord injury. J Neurochem 110:765–778

    Article  PubMed  CAS  Google Scholar 

  23. Ito M, Natsume A, Takeuchi H, Shimato S, Ohno M, Wakabayashi T, Yoshida J (2009) Type I interferon inhibits astrocytic gliosis and promotes functional recovery after spinal cord injury by deactivation of the MEK/ERK pathway. J Neurotrauma 26:41–53

    Article  PubMed  Google Scholar 

  24. Koistinaho M, Kettunen MI, Goldsteins G, Keinanen R, Salminen A, Ort M, Bures J, Liu D, Kauppinen RA, Higgins LS, Koistinaho J (2002) Beta-amyloid precursor protein transgenic mice that harbor diffuse A beta deposits but do not form plaques show increased ischemic vulnerability: role of inflammation. Proc Natl Acad Sci USA 99:1610–1615

    Article  PubMed  CAS  Google Scholar 

  25. Barone FC, Irving EA, Ray AM, Lee JC, Kassis S, Kumar S, Badger AM, White RF, McVey MJ, Legos JJ, Erhardt JA, Nelson AH, Ohlstein EH, Hunter AJ, Ward K, Smith BR, Adams JL, Parsons AA (2001) SB 239063, a second-generation p38 mitogen-activated protein kinase inhibitor, reduces brain injury and neurological deficits in cerebral focal ischemia. J Pharmacol Exp Ther 296:312–321

    PubMed  CAS  Google Scholar 

  26. Namura S, Iihara K, Takami S, Nagata I, Kikuchi H, Matsushita K, Moskowitz MA, Bonventre JV, Alessandrini A (2001) Intravenous administration of MEK inhibitor U0126 affords brain protection against forebrain ischemia and focal cerebral ischemia. Proc Natl Acad Sci USA 98:11569–11574

    Article  PubMed  CAS  Google Scholar 

  27. Wang X, Wang H, Xu L, Rozanski DJ, Sugawara T, Chan PH, Trzaskos JM, Feuerstein GZ (2003) Significant neuroprotection against ischemic brain injury by inhibition of the MEK1 protein kinase in mice: exploration of potential mechanism associated with apoptosis. J Pharmacol Exp Ther 304:172–178

    Article  PubMed  CAS  Google Scholar 

  28. Mori T, Wang X, Aoki T, Lo EH (2002) Downregulation of matrix metalloproteinase-9 and attenuation of edema via inhibition of ERK mitogen activated protein kinase in traumatic brain injury. J Neurotrauma 19:1411–1419

    Article  PubMed  Google Scholar 

  29. Lennmyr F, Ericsson A, Gerwins P, Ahlstrom H, Terent A (2003) Increased brain injury and vascular leakage after pretreatment with p38-inhibitor SB203580 in transient ischemia. Acta Neurol Scand 108:339–345

    Article  PubMed  CAS  Google Scholar 

  30. Logan A, Frautschy SA, Gonzalez AM, Baird A (1992) A time course for the focal elevation of synthesis of basic fibroblast growth factor and one of its high-affinity receptors (flg) following a localized cortical brain injury. J Neurosci 12:3828–3837

    PubMed  CAS  Google Scholar 

  31. Reilly JF, Kumari VG (1996) Alterations in fibroblast growth factor receptor expression following brain injury. Exp Neurol 140:139–150

    Article  PubMed  CAS  Google Scholar 

  32. Burnstock G (2008) Purinergic signalling and disorders of the central nervous system. Nat Rev Drug Discov 7:575–590

    Article  PubMed  CAS  Google Scholar 

  33. Planas AM, Justicia C, Soriano MA, Ferrer I (1998) Epidermal growth factor receptor in proliferating reactive glia following transient focal ischemia in the rat brain. Glia 23:120–129

    Article  PubMed  CAS  Google Scholar 

  34. Ferrer I, Alcantara S, Ballabriga J, Olive M, Blanco R, Rivera R, Carmona M, Berruezo M, Pitarch S, Planas AM (1996) Transforming growth factor-alpha (TGF-alpha) and epidermal growth factor-receptor (EGF-R) immunoreactivity in normal and pathologic brain. Prog Neurobiol 49:99–123

    Article  PubMed  CAS  Google Scholar 

  35. Birecree E, Whetsell WO Jr, Stoscheck C, King LE Jr, Nanney LB (1988) Immunoreactive epidermal growth factor receptors in neuritic plaques from patients with Alzheimer's disease. J Neuropathol Exp Neurol 47:549–560

    Article  PubMed  CAS  Google Scholar 

  36. Liu B, Chen H, Johns TG, Neufeld AH (2006) Epidermal growth factor receptor activation: an upstream signal for transition of quiescent astrocytes into reactive astrocytes after neural injury. J Neurosci 26:7532–7540

    Article  PubMed  CAS  Google Scholar 

  37. Chen Y, Miles DK, Hoang T, Shi J, Hurlock E, Kernie SG, Lu QR (2008) The basic helix-loop-helix transcription factor olig2 is critical for reactive astrocyte proliferation after cortical injury. J Neurosci 28:10983–10989

    Article  PubMed  CAS  Google Scholar 

  38. Buffo A, Vosko MR, Erturk D, Hamann GF, Jucker M, Rowitch D, Gotz M (2005) Expression pattern of the transcription factor Olig2 in response to brain injuries: implications for neuronal repair. Proc Natl Acad Sci USA 102:18183–18188

    Article  PubMed  CAS  Google Scholar 

  39. Cassiani-Ingoni R, Coksaygan T, Xue H, Reichert-Scrivner SA, Wiendl H, Rao MS, Magnus T (2006) Cytoplasmic translocation of Olig2 in adult glial progenitors marks the generation of reactive astrocytes following autoimmune inflammation. Exp Neurol 201:349–358

    Article  PubMed  CAS  Google Scholar 

  40. Magnus T, Coksaygan T, Korn T, Xue H, Arumugam TV, Mughal MR, Eckley DM, Tang SC, Detolla L, Rao MS, Cassiani-Ingoni R, Mattson MP (2007) Evidence that nucleocytoplasmic Olig2 translocation mediates brain-injury-induced differentiation of glial precursors to astrocytes. J Neurosci Res 85:2126–2137

    Article  PubMed  CAS  Google Scholar 

  41. Amankulor NM, Hambardzumyan D, Pyonteck SM, Becher OJ, Joyce JA, Holland EC (2009) Sonic hedgehog pathway activation is induced by acute brain injury and regulated by injury-related inflammation. J Neurosci 29:10299–10308

    Article  PubMed  CAS  Google Scholar 

  42. Peters CM, Rogers SD, Pomonis JD, Egnaczyk GF, Keyser CP, Schmidt JA, Ghilardi JR, Maggio JE, Mantyh PW (2003) Endothelin receptor expression in the normal and injured spinal cord: potential involvement in injury-induced ischemia and gliosis. Exp Neurol 180:1–13

    Article  PubMed  CAS  Google Scholar 

  43. Li JJ, Wu LH, Cao Q, Yuan Y, Yang L, Guo ZY, Kaur C, Sivakumar V, Ling EA, Wu CY (2010) Endothelins-1/3 and endothelin-A/B receptors expressing glial cells with special reference to activated microglia in experimentally induced cerebral ischemia in the adult rats. Neuroscience 167:665–677

    Article  PubMed  CAS  Google Scholar 

  44. Koyama Y, Takemura M, Fujiki K, Ishikawa N, Shigenaga Y, Baba A (1999) BQ788, an endothelin ET(B) receptor antagonist, attenuates stab wound injury-induced reactive astrocytes in rat brain. Glia 26:268–271

    Article  PubMed  CAS  Google Scholar 

  45. Gadea A, Schinelli S, Gallo V (2008) Endothelin-1 regulates astrocyte proliferation and reactive gliosis via a JNK/c-Jun signaling pathway. J Neurosci 28:2394–2408

    Article  PubMed  CAS  Google Scholar 

  46. Akassoglou K, Adams RA, Bauer J, Mercado P, Tseveleki V, Lassmann H, Probert L, Strickland S (2004) Fibrin depletion decreases inflammation and delays the onset of demyelination in a tumor necrosis factor transgenic mouse model for multiple sclerosis. Proc Natl Acad Sci USA 101:6698–6703

    Article  PubMed  CAS  Google Scholar 

  47. Schachtrup C, Ryu JK, Helmrick MJ, Vagena E, Galanakis DK, Degen JL, Margolis RU, Akassoglou K (2010) Fibrinogen triggers astrocyte scar formation by promoting the availability of active TGF-beta after vascular damage. J Neurosci 30:5843–5854

    Article  PubMed  CAS  Google Scholar 

  48. Chen J, Leong SY, Schachner M (2005) Differential expression of cell fate determinants in neurons and glial cells of adult mouse spinal cord after compression injury. Eur J Neurosci 22:1895–1906

    Article  PubMed  Google Scholar 

  49. Setoguchi T, Nakashima K, Takizawa T, Yanagisawa M, Ochiai W, Okabe M, Yone K, Komiya S, Taga T (2004) Treatment of spinal cord injury by transplantation of fetal neural precursor cells engineered to express BMP inhibitor. Exp Neurol 189:33–44

    Article  PubMed  CAS  Google Scholar 

  50. Setoguchi T, Yone K, Matsuoka E, Takenouchi H, Nakashima K, Sakou T, Komiya S, Izumo S (2001) Traumatic injury-induced BMP7 expression in the adult rat spinal cord. Brain Res 921:219–225

    Article  PubMed  CAS  Google Scholar 

  51. Matsuura I, Taniguchi J, Hata K, Saeki N, Yamashita T (2008) BMP inhibition enhances axonal growth and functional recovery after spinal cord injury. J Neurochem 105:1471–1479

    Article  PubMed  CAS  Google Scholar 

  52. Sahni V, Mukhopadhyay A, Tysseling V, Hebert A, Birch D, McGuire TL, Stupp SI, Kessler JA (2010) BMPR1a and BMPR1b signaling exert opposing effects on gliosis after spinal cord injury. J Neurosci 30:1839–1855

    Article  PubMed  CAS  Google Scholar 

  53. Choi JS, Kim SY, Cha JH, Choi YS, Sung KW, Oh ST, Kim ON, Chung JW, Chun MH, Lee SB, Lee MY (2003) Upregulation of gp130 and STAT3 activation in the rat hippocampus following transient forebrain ischemia. Glia 41:237–246

    Article  PubMed  Google Scholar 

  54. Planas AM, Soriano MA, Berruezo M, Justicia C, Estrada A, Pitarch S, Ferrer I (1996) Induction of Stat3, a signal transducer and transcription factor, in reactive microglia following transient focal cerebral ischaemia. Eur J Neurosci 8:2612–2618

    Article  PubMed  CAS  Google Scholar 

  55. Satriotomo I, Bowen KK, Vemuganti R (2006) JAK2 and STAT3 activation contributes to neuronal damage following transient focal cerebral ischemia. J Neurochem 98:1353–1368

    Article  PubMed  CAS  Google Scholar 

  56. Suzuki S, Tanaka K, Nogawa S, Dembo T, Kosakai A, Fukuuchi Y (2001) Phosphorylation of signal transducer and activator of transcription-3 (Stat3) after focal cerebral ischemia in rats. Exp Neurol 170:63–71

    Article  PubMed  CAS  Google Scholar 

  57. Justicia C, Gabriel C, Planas AM (2000) Activation of the JAK/STAT pathway following transient focal cerebral ischemia: signaling through Jak1 and Stat3 in astrocytes. Glia 30:253–270

    Article  PubMed  CAS  Google Scholar 

  58. Herrmann JE, Imura T, Song B, Qi J, Ao Y, Nguyen TK, Korsak RA, Takeda K, Akira S, Sofroniew MV (2008) STAT3 is a critical regulator of astrogliosis and scar formation after spinal cord injury. J Neurosci 28:7231–7243

    Article  PubMed  CAS  Google Scholar 

  59. Okada S, Nakamura M, Katoh H, Miyao T, Shimazaki T, Ishii K, Yamane J, Yoshimura A, Iwamoto Y, Toyama Y, Okano H (2006) Conditional ablation of Stat3 or Socs3 discloses a dual role for reactive astrocytes after spinal cord injury. Nat Med 12:829–834

    Article  PubMed  CAS  Google Scholar 

  60. Papadopoulos MC, Manley GT, Krishna S, Verkman AS (2004) Aquaporin-4 facilitates reabsorption of excess fluid in vasogenic brain edema. FASEB J 18:1291–1293

    PubMed  CAS  Google Scholar 

  61. Papadopoulos MC, Verkman AS (2005) Aquaporin-4 gene disruption in mice reduces brain swelling and mortality in pneumococcal meningitis. J Biol Chem 280:13906–13912

    Article  PubMed  CAS  Google Scholar 

  62. Verkman AS, Binder DK, Bloch O, Auguste K, Papadopoulos MC (2006) Three distinct roles of aquaporin-4 in brain function revealed by knockout mice. Biochim Biophys Acta 1758:1085–1093

    Article  PubMed  CAS  Google Scholar 

  63. Saadoun S, Papadopoulos MC, Krishna S (2003) Water transport becomes uncoupled from K+ siphoning in brain contusion, bacterial meningitis, to brain tumours: immunohistochemical case review. J Clin Pathol 56:972–975

    Article  PubMed  CAS  Google Scholar 

  64. Saadoun S, Papadopoulos MC, Davies DC, Krishna S, Bell BA (2002) Aquaporin-4 expression is increased in oedematous human brain tumours. J Neurol Neurosurg Psychiatry 72:262–265

    Article  PubMed  CAS  Google Scholar 

  65. Manley GT, Fujimura M, Ma T, Noshita N, Filiz F, Bollen AW, Chan P, Verkman AS (2000) Aquaporin-4 deletion in mice reduces brain edema after acute water intoxication and ischemic stroke. Nat Med 6:159–163

    Article  PubMed  CAS  Google Scholar 

  66. Saadoun S, Papadopoulos MC, Watanabe H, Yan D, Manley GT, Verkman AS (2005) Involvement of aquaporin-4 in astroglial cell migration and glial scar formation. J Cell Sci 118:5691–5698

    Article  PubMed  CAS  Google Scholar 

  67. Brakebusch C, Grose R, Quondamatteo F, Ramirez A, Jorcano JL, Pirro A, Svensson M, Herken R, Sasaki T, Timpl R, Werner S, Fassler R (2000) Skin and hair follicle integrity is crucially dependent on beta 1 integrin expression on keratinocytes. EMBO J 19:3990–4003

    Article  PubMed  CAS  Google Scholar 

  68. Robel S, Mori T, Zoubaa S, Schlegel J, Sirko S, Faissner A, Goebbels S, Dimou L, Gotz M (2009) Conditional deletion of beta1-integrin in astroglia causes partial reactive gliosis. Glia 57:1630–1647

    Article  PubMed  Google Scholar 

  69. Beggs HE, Schahin-Reed D, Zang K, Goebbels S, Nave KA, Gorski J, Jones KR, Sretavan D, Reichardt LF (2003) FAK deficiency in cells contributing to the basal lamina results in cortical abnormalities resembling congenital muscular dystrophies. Neuron 40:501–514

    Article  PubMed  CAS  Google Scholar 

  70. Filbin MT (2003) Myelin-associated inhibitors of axonal regeneration in the adult mammalian CNS. Nat Rev Neurosci 4:703–713

    Article  PubMed  CAS  Google Scholar 

  71. Silver J, Miller JH (2004) Regeneration beyond the glial scar. Nat Rev Neurosci 5:146–156

    Article  PubMed  CAS  Google Scholar 

  72. Schwab ME (2002) Repairing the injured spinal cord. Science 295:1029–1031

    Article  PubMed  CAS  Google Scholar 

  73. Lee JK, Zheng B (2008) Axon regeneration after spinal cord injury: insight from genetically modified mouse models. Restor Neurol Neurosci 26:175–182

    PubMed  Google Scholar 

  74. Zheng B, Ho C, Li S, Keirstead H, Steward O, Tessier-Lavigne M (2003) Lack of enhanced spinal regeneration in Nogo-deficient mice. Neuron 38:213–224

    Article  PubMed  CAS  Google Scholar 

  75. Zheng B, Atwal J, Ho C, Case L, He XL, Garcia KC, Steward O, Tessier-Lavigne M (2005) Genetic deletion of the Nogo receptor does not reduce neurite inhibition in vitro or promote corticospinal tract regeneration in vivo. Proc Natl Acad Sci USA 102:1205–1210

    Article  PubMed  CAS  Google Scholar 

  76. Simonen M, Pedersen V, Weinmann O, Schnell L, Buss A, Ledermann B, Christ F, Sansig G, van der Putten H, Schwab ME (2003) Systemic deletion of the myelin-associated outgrowth inhibitor Nogo-A improves regenerative and plastic responses after spinal cord injury. Neuron 38:201–211

    Article  PubMed  CAS  Google Scholar 

  77. Bartsch U, Bandtlow CE, Schnell L, Bartsch S, Spillmann AA, Rubin BP, Hillenbrand R, Montag D, Schwab ME, Schachner M (1995) Lack of evidence that myelin-associated glycoprotein is a major inhibitor of axonal regeneration in the CNS. Neuron 15:1375–1381

    Article  PubMed  CAS  Google Scholar 

  78. Kim JE, Liu BP, Park JH, Strittmatter SM (2004) Nogo-66 receptor prevents raphespinal and rubrospinal axon regeneration and limits functional recovery from spinal cord injury. Neuron 44:439–451

    Article  PubMed  CAS  Google Scholar 

  79. Cafferty WB, Yang SH, Duffy PJ, Li S, Strittmatter SM (2007) Functional axonal regeneration through astrocytic scar genetically modified to digest chondroitin sulfate proteoglycans. J Neurosci 27:2176–2185

    Article  PubMed  CAS  Google Scholar 

  80. Goldshmit Y, McLenachan S, Turnley A (2006) Roles of Eph receptors and ephrins in the normal and damaged adult CNS. Brain Res Rev 52:327–345

    Article  PubMed  CAS  Google Scholar 

  81. Benson MD, Romero MI, Lush ME, Lu QR, Henkemeyer M, Parada LF (2005) Ephrin-B3 is a myelin-based inhibitor of neurite outgrowth. Proc Natl Acad Sci USA 102:10694–10699

    Article  PubMed  CAS  Google Scholar 

  82. Kullander K, Croll SD, Zimmer M, Pan L, McClain J, Hughes V, Zabski S, DeChiara TM, Klein R, Yancopoulos GD, Gale NW (2001) Ephrin-B3 is the midline barrier that prevents corticospinal tract axons from recrossing, allowing for unilateral motor control. Genes Dev 15:877–888

    Article  PubMed  CAS  Google Scholar 

  83. Wahl S, Barth H, Ciossek T, Aktories K, Mueller BK (2000) Ephrin-A5 induces collapse of growth cones by activating Rho and Rho kinase. J Cell Biol 149:263–270

    Article  PubMed  CAS  Google Scholar 

  84. Yue Y, Su J, Cerretti DP, Fox GM, Jing S, Zhou R (1999) Selective inhibition of spinal cord neurite outgrowth and cell survival by the Eph family ligand ephrin-A5. J Neurosci 19:10026–10035

    PubMed  CAS  Google Scholar 

  85. Goldshmit Y, Galea MP, Wise G, Bartlett PF, Turnley AM (2004) Axonal regeneration and lack of astrocytic gliosis in EphA4-deficient mice. J Neurosci 24:10064–10073

    Article  PubMed  CAS  Google Scholar 

  86. McKeon RJ, Jurynec MJ, Buck CR (1999) The chondroitin sulfate proteoglycans neurocan and phosphacan are expressed by reactive astrocytes in the chronic CNS glial scar. J Neurosci 19:10778–10788

    PubMed  CAS  Google Scholar 

  87. Tang X, Davies JE, Davies SJ (2003) Changes in distribution, cell associations, and protein expression levels of NG2, neurocan, phosphacan, brevican, versican V2, and tenascin-C during acute to chronic maturation of spinal cord scar tissue. J Neurosci Res 71:427–444

    Article  PubMed  CAS  Google Scholar 

  88. Jones LL, Margolis RU, Tuszynski MH (2003) The chondroitin sulfate proteoglycans neurocan, brevican, phosphacan, and versican are differentially regulated following spinal cord injury. Exp Neurol 182:399–411

    Article  PubMed  CAS  Google Scholar 

  89. Schaefer L, Schaefer RM (2010) Proteoglycans: from structural compounds to signaling molecules. Cell Tissue Res 339:237–246

    Article  PubMed  CAS  Google Scholar 

  90. Koprivica V, Cho KS, Park JB, Yiu G, Atwal J, Gore B, Kim JA, Lin E, Tessier-Lavigne M, Chen DF, He Z (2005) EGFR activation mediates inhibition of axon regeneration by myelin and chondroitin sulfate proteoglycans. Science 310:106–110

    Article  PubMed  CAS  Google Scholar 

  91. Erschbamer M, Pernold K, Olson L (2007) Inhibiting epidermal growth factor receptor improves structural, locomotor, sensory, and bladder recovery from experimental spinal cord injury. J Neurosci 27:6428–6435

    Article  PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jean M. Hébert.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Kang, W., Hébert, J.M. Signaling Pathways in Reactive Astrocytes, a Genetic Perspective. Mol Neurobiol 43, 147–154 (2011). https://doi.org/10.1007/s12035-011-8163-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12035-011-8163-7

Keywords

Navigation