Plasticity Related Gene 3 (PRG3) overcomes myelin-associated growth inhibition and promotes functional recovery after spinal cord injury

The Plasticity Related Gene family covers five, brain-specific, transmembrane proteins (PRG1-5, also termed LPPR1-5) that operate in neuronal plasticity during development, aging and brain trauma. Here we investigated the role of the PRG family on axonal and filopodia outgrowth. Comparative analysis revealed the strongest outgrowth induced by PRG3 (LPPR1). During development, PRG3 is ubiquitously located at the tip of neuronal processes and at the plasma membrane and declines with age. In utero electroporation of PRG3 induced dendritic protrusions and accelerated spine formations in cortical pyramidal neurons. The neurite growth promoting activity of PRG3 requires RasGRF1 (RasGEF1/Cdc25) mediated downstream signaling. Moreover, in axon collapse assays, PRG3-induced neurites resisted growth inhibitors such as myelin, Nogo-A (Reticulon/RTN-4), thrombin and LPA and impeded the RhoA-Rock-PIP5K induced neurite repulsion. Transgenic adult mice with constitutive PRG3 expression displayed strong axonal sprouting distal to a spinal cord lesion. Moreover, fostered PRG3 expression promoted complex motor-behavioral recovery compared to wild type controls as revealed in the Schnell swim test (SST). Thus, PRG3 emerges as a developmental RasGRF1-dependent conductor of filopodia formation and axonal growth enhancer. PRG3-induced neurites resist brain injury-associated outgrowth inhibitors and contribute to functional recovery after spinal cord lesions. Here, we provide evidence that PRG3 operates as an essential neuronal growth promoter in the nervous system. Maintaining PRG3 expression in aging brain may turn back the developmental clock for neuronal regeneration and plasticity.

[1]  S. Tenzer,et al.  PRG-1 Regulates Synaptic Plasticity via Intracellular PP2A/β1-Integrin Signaling. , 2016, Developmental cell.

[2]  A. Doerfler,et al.  PRG3 induces Ras-dependent oncogenic cooperation in gliomas , 2016, Oncotarget.

[3]  H. M. Geller,et al.  Cooperative interactions of LPPR family members in membrane localization and alteration of cellular morphology , 2015, Journal of Cell Science.

[4]  D. Muller,et al.  Structural plasticity: mechanisms and contribution to developmental psychiatric disorders , 2014, Front. Neuroanat..

[5]  M. Schwab,et al.  The Sphingolipid Receptor S1PR2 Is a Receptor for Nogo-A Repressing Synaptic Plasticity , 2014, PLoS biology.

[6]  D. Ingber,et al.  Nogo-A is a negative regulator of CNS angiogenesis , 2013, Proceedings of the National Academy of Sciences.

[7]  A. Bräuer,et al.  Plasticity-related gene 3 promotes neurite shaft protrusion , 2013, BMC Neuroscience.

[8]  A. Bräuer,et al.  Plasticity-related gene 3 promotes neurite shaft protrusion , 2013, BMC Neuroscience.

[9]  François Guillemot,et al.  Visualization and Genetic Manipulation of Dendrites and Spines in the Mouse Cerebral Cortex and Hippocampus using In utero Electroporation , 2012, Journal of visualized experiments : JoVE.

[10]  L. Mendell,et al.  Combined delivery of Nogo-A antibody, neurotrophin-3 and the NMDA-NR2d subunit establishes a functional ‘detour’ in the hemisected spinal cord , 2011, The European journal of neuroscience.

[11]  P. Hordijk,et al.  Rac controls PIP5K localisation and PtdIns(4,5)P2 synthesis, which modulates vinculin localisation and neurite dynamics , 2010, Journal of Cell Science.

[12]  M. Frotscher,et al.  L1 syndrome mutations impair neuronal L1 function at different levels by divergent mechanisms , 2010, Neurobiology of Disease.

[13]  R. Nitsch,et al.  Plasticity-related Gene 5 (PRG5) Induces Filopodia and Neurite Growth and Impedes Lysophosphatidic Acid– and Nogo-A–mediated Axonal Retraction , 2010, Molecular biology of the cell.

[14]  H. Mellor The role of formins in filopodia formation. , 2010, Biochimica et biophysica acta.

[15]  S. Strittmatter,et al.  Rho-Associated Kinase II (ROCKII) Limits Axonal Growth after Trauma within the Adult Mouse Spinal Cord , 2009, The Journal of Neuroscience.

[16]  Carmen Birchmeier,et al.  Synaptic PRG-1 Modulates Excitatory Transmission via Lipid Phosphate-Mediated Signaling , 2009, Cell.

[17]  J. Fawcett,et al.  Chronic spinal hemisection in rats induces a progressive decline in transmission in uninjured fibers to motoneurons , 2009, Experimental Neurology.

[18]  A. Prokop,et al.  Formin Proteins of the DAAM Subfamily Play a Role during Axon Growth , 2008, The Journal of Neuroscience.

[19]  C. Shatz,et al.  PirB is a Functional Receptor for Myelin Inhibitors of Axonal Regeneration , 2008, Science.

[20]  P. Mattila,et al.  Filopodia: molecular architecture and cellular functions , 2008, Nature Reviews Molecular Cell Biology.

[21]  C. Nimsky,et al.  Small interfering RNA–mediated xCT silencing in gliomas inhibits neurodegeneration and alleviates brain edema , 2008, Nature Medicine.

[22]  F. Zipp,et al.  Sirt1 contributes critically to the redox-dependent fate of neural progenitors , 2008, Nature Cell Biology.

[23]  C. Lopez,et al.  Progressive Motor Neuronopathy: A Critical Role of the Tubulin Chaperone TBCE in Axonal Tubulin Routing from the Golgi Apparatus , 2007, The Journal of Neuroscience.

[24]  H. Kuhn,et al.  Role for glutathione peroxidase-4 in brain development and neuronal apoptosis: specific induction of enzyme expression in reactive astrocytes following brain injury. , 2007, Free radical biology & medicine.

[25]  A. Morris,et al.  Cdc42 and ARP2/3-independent regulation of filopodia by an integral membrane lipid-phosphatase-related protein , 2007, Journal of Cell Science.

[26]  A. Ridley Rho GTPases and actin dynamics in membrane protrusions and vesicle trafficking. , 2006, Trends in cell biology.

[27]  Noam Y. Harel,et al.  Can regenerating axons recapitulate developmental guidance during recovery from spinal cord injury? , 2006, Nature Reviews Neuroscience.

[28]  K. Fouad,et al.  Nogo‐A antibody improves regeneration and locomotion of spinal cord–injured rats , 2005, Annals of neurology.

[29]  Tomoyuki Yamada,et al.  dsCheck: highly sensitive off-target search software for double-stranded RNA-mediated RNA interference , 2005, Nucleic Acids Res..

[30]  J. Feramisco,et al.  Rho‐mediated cytoskeletal rearrangement in response to LPA is functionally antagonized by Rac1 and PIP2 , 2004, Journal of neurochemistry.

[31]  Jerry Silver,et al.  Regeneration beyond the glial scar , 2004, Nature Reviews Neuroscience.

[32]  R. Nitsch,et al.  Molecular cloning and expression regulation of PRG‐3, a new member of the plasticity‐related gene family , 2004, The European journal of neuroscience.

[33]  A. B. Huber,et al.  Nogo-A Inhibits Neurite Outgrowth and Cell Spreading with Three Discrete Regions , 2003, The Journal of Neuroscience.

[34]  Charles Boone,et al.  Formins: signaling effectors for assembly and polarization of actin filaments , 2003, Journal of Cell Science.

[35]  R. Nitsch,et al.  A new phospholipid phosphatase, PRG-1, is involved in axon growth and regenerative sprouting , 2003, Nature Neuroscience.

[36]  Vincenzo De Paola,et al.  AMPA receptors regulate dynamic equilibrium of presynaptic terminals in mature hippocampal networks , 2003, Nature Neuroscience.

[37]  Gary G. Borisy,et al.  Mechanism of filopodia initiation by reorganization of a dendritic network , 2003, The Journal of cell biology.

[38]  T. Takenawa,et al.  Small GTPase Tc10 and its homologue RhoT induce N-WASP-mediated long process formation and neurite outgrowth , 2003, Journal of Cell Science.

[39]  A. Hall,et al.  Rho GTPases in cell biology , 2002, Nature.

[40]  B. Eickholt,et al.  Essential Role of Type Iα Phosphatidylinositol 4-Phosphate 5-Kinase in Neurite Remodeling , 2002, Current Biology.

[41]  Sheila M. Thomas,et al.  N-WASP deficiency reveals distinct pathways for cell surface projections and microbial actin-based motility , 2001, Nature Cell Biology.

[42]  K. Rottner,et al.  Actin pedestal formation by enteropathogenic Escherichia coli and intracellular motility of Shigella flexneri are abolished in N‐WASP‐defective cells , 2001, EMBO reports.

[43]  W. Lim,et al.  Integration of multiple signals through cooperative regulation of the N-WASP-Arp2/3 complex. , 2000, Science.

[44]  V. Berezin,et al.  Neural Cell Adhesion Molecule-Stimulated Neurite Outgrowth Depends on Activation of Protein Kinase C and the Ras–Mitogen-Activated Protein Kinase Pathway , 2000, The Journal of Neuroscience.

[45]  Martin E. Schwab,et al.  Nogo-A is a myelin-associated neurite outgrowth inhibitor and an antigen for monoclonal antibody IN-1 , 2000, Nature.

[46]  R. Prinjha,et al.  Neurobiology: Inhibitor of neurite outgrowth in humans , 2000, Nature.

[47]  A. Hall,et al.  Activation of RhoA by lysophosphatidic acid and Galpha12/13 subunits in neuronal cells: induction of neurite retraction. , 1999, Molecular biology of the cell.

[48]  M. Kirschner,et al.  The Interaction between N-WASP and the Arp2/3 Complex Links Cdc42-Dependent Signals to Actin Assembly , 1999, Cell.

[49]  Laura M. Machesky,et al.  Scar1 and the related Wiskott–Aldrich syndrome protein, WASP, regulate the actin cytoskeleton through the Arp2/3 complex , 1998, Current Biology.

[50]  L. Lim,et al.  Rho family GTPases and neuronal growth cone remodelling: relationship between increased complexity induced by Cdc42Hs, Rac1, and acetylcholine and collapse induced by RhoA and lysophosphatidic acid , 1997, Molecular and cellular biology.

[51]  C. Nobes,et al.  Rho, rac and cdc42 GTPases: regulators of actin structures, cell adhesion and motility. , 1995, Biochemical Society transactions.

[52]  C. Nobes,et al.  Rho, Rac, and Cdc42 GTPases regulate the assembly of multimolecular focal complexes associated with actin stress fibers, lamellipodia, and filopodia , 1995, Cell.

[53]  M. Schwab,et al.  Axonal regeneration in the rat spinal cord produced by an antibody against myelin-associated neurite growth inhibitors , 1990, Nature.

[54]  R. Nitsch,et al.  Autotaxin (NPP-2) in the brain: cell type-specific expression and regulation during development and after neurotrauma , 2006, Cellular and Molecular Life Sciences.

[55]  B. Eickholt,et al.  Essential role of type I(alpha) phosphatidylinositol 4-phosphate 5-kinase in neurite remodeling. , 2002, Current biology : CB.

[56]  R. Nitsch,et al.  Molecules Involved in Reactive Sprouting in the Hippocampus , 2001, Reviews in the neurosciences.

[57]  R. Evans,et al.  Initiation of neuropathic pain requires lysophosphatidic acid receptor signaling , 2022 .