April Newsletter: Rho Family GEFs and Dendritic Spine Structural Plasticity

Dendritic spines are the post-synaptic component of most excitatory glutamatergic synapses and primary site of synaptic structural plasticity1-4 for modulating synaptic function3. Activity-dependent structural plasticity in spines (i.e., spine morphogenesis) depends upon dynamic re-organization of F-actin, the primary structural component of spines1-7. Spine morphogenesis is important for normal learning and memory and the development of neurodegenerative diseases and neurological disorders8-10.

The RhoA, Rac1, and Cdc42 GTPases regulate spine morphogenesis; RhoA inhibits spine growth and stability, whereas Rac1 and Cdc42 exert the opposite effect. In reality, Rho family regulation of spine structural plasticity is much more complex5,7,11-16. Precise spatiotemporal regulation of Rho GTPases is with guanine exchange factors (GEFs) triggering  GTP/GDP exchange and GTPase activating proteins (GAPs) stimulating intrinsic GTPase activity. At least eight Rho family GEFs regulate spine morphogenesis and these GEFs are activated through a variety of receptor signaling pathways, including glutamatergic NMDA receptors (NMDARs) and receptor tyrosine kinases (RTKs)5-7. NMDARs mediate calcium influx and subsequent activation of calcium/calmodulin-dependent kinases (CaMKs), which phosphorylate Rho family GEFs5,7,17,18, essential for GEF activity. In this newsletter, the regulation of spine structural plasticity by the Rho family GEFs Kalirin7 (Kal7; the most abundant isoform in adult brain), Trio-9 (the most abundant isoform in hippocampus), Tiam1, RasGRF2, DOCK10, DOCK180, ephrexin1, and ephrexin5 is discussed (Fig. 1).

 Rac and Cdc42 GEFs  

The Rac GEF Kal7 is highly expressed in the spines of mature hippocampal and cortical neurons. Kal7 is essential for NMDAR-dependent long-term potentiation (LTP), the most studied form of synaptic plasticity and the likely cellular basis of learning and memory19,20. Calcium-activated CaMKII phosphorylates the Thr95 residue, which enables Kal7-mediated activation of Rac1 and re-organization of the actin cytoskeleton necessary for activity-dependent spine morphogenesis17,18,21,22. Elevated Kal7 activity increases spine density and size, whereas Kal7 downregulation decreases spine density in vitro and in vivo17,18,21-24. Similarly, a Kalirin paralog, the GEF Trio-9, is phosphorylated by CaMKII and regulates NMDAR-dependent LTP in hippocampal neurons and presumably the concomitant spine structural plasticity in a manner similar to Kal718. In addition, Kal7 activity is required for spine morphogenesis induced by N-cadherins, a class of trans-synaptic adhesion molecules5,22(Fig. 1).


The Rac GEF Tiam1 is highly expressed in the developing cortex and hippocampus  and also undergoes CaMKII-mediated phosphorylation, resulting in activation of Rac117,25. Selectively blocking Tiam1 function reduces spine density and inhibits NMDAR-dependent formation of new spines, while Tiam1 overexpression increases spine density5,25. Interestingly, Tiam1-induced Rac activation, subsequent spine morphogenesis, and Tiam1's restricted localization to spines is regulated by the polarity protein PAR-3 (partitioning-defective gene 3)26 (Fig. 1).


Other Rac GEFs that regulate actin-based spine plasticity are RasGRF2 and DOCK180. RasGRF2 is required for NMDAR-dependent LTP and the correlated rapid spine enlargement27. Similarly, DOCK180, in a mandatory complex with ELMO1, also positively regulates spine morphogenesis via an activation of Rac GTPases in neurons. Loss of DOCK180 reduces spine density with no effect on spine head size, while over-expression enhances density28 (Fig. 1).


Rac GEFs are also phosphorylated following activation of EphB RTKs5-7,17,22,29 (Fig. 1). Kal7 is necessary for EphB-mediated increases in spine density30 and Kal7 phosphorylation could be mediated by either EphB or associated kinase. A noteworthy candidate is cyclin-dependent kinase 5 (Cdk5), which phosphorylates Kal7 on Thr1590 and affects Kal7's regulation of spine morphogenesis31. Similar to Kal7, Tiam1 is necessary for EphB-mediated increases in spine density and is activated via phosphorylation of Tyr829, also by either EphB or associated kinase32. EphB RTKs also regulate the coordinated activities of Tiam1 and the Rac GAP Bcr, which form a complex essential for spatially-regulated EphB RTK-mediated control of spine morphogenesis33. Athough DOCK10 is a Cdc42/Rac1 GEF, the positive regulation of spine number and head size in cerebellar and hippocampal neurons is mediated by its activation of Cdc4234 (Fig. 1).

418figure_1

Figure 1. Regulation of dendritic spine structural plasticity by Rho family GEFs.

 Rho GEFs  

As opposed to Rac and Cdc42 GEFs, which upregulate spine plasticity, Rho GEFs exert the opposite effect. For instance, GEF-H1 (a.k.a. ARHGEF2 or Lfc), negatively regulates spine density and length35. Following the sequential activation of the RTK EphA4, Cdk5, and the Rho GEF ephexin1 by Src kinase-mediated phosphorylation of Tyr87 and Cdk5-mediated phosphorylation of Thr41 and Ser139 residues on ephexin1, activated RhoA induces spine retraction in hippocampal neurons29 (Fig. 1). Another Rho GEF, ephexin5, also induces spine retraction via RhoA activation. Ephexin5 directly binds and inhibits EphB-mediated spine morphogenesis while activating RhoA. Upon ligand-induced activation of EphB, the RTK inactivates ephexin5 through tyrosine phosphorylation (Tyr361), which induces ubiquitin-mediated degradation. Upon disinhibition, EphB can positively regulate spine morphogenesis36 (Fig. 1).

 Summary   

The dendritic spine is the post-synaptic component of most excitatory neurotransmission and site of synaptic structural plasticity. Spine structural plasticity relies upon re-modeling of the actin cytoskeleton, which is regulated by GEF-mediated activation of Rho family GTPases. Thus, it is imperative to better understand how GEFs function in neurons and how the same GTPase is controlled by different GEFs for precise spatiotemporal regulation. To assist scientists in these studies, Cytoskeleton, Inc. offers SiR-actin live cell imaging probe and multiple purified GEFs and GTPases, along with activation and exchange assays, antibodies, and activators and inhibitors.

 Related Products & Resources  

RasGRF1 GEF Protein (Cdc25 Exchange Domain, aa1038-1270, MBP tag) (Cat. # CS-GE03)

Tiam1 GEF Protein (DHPH Exchange Domain, aa1040-1406, MBP tag) (Cat. # CS-GE04)

Vav1 GEF Protein (DHPHC1 Exchange Domain, Y174D mutant, aa168-522, 6xHis tag) (Cat. # CS-GE05)

Vav2 GEF Protein (DH Exchange Domain, aa189-374, 6xHis tag) (Cat. # CS-GE06)

RhoGEF Exchange Assay (Cat. # BK100)

Spirochrome SiR-Actin Kit (Cat. # CY-SC001)

 References  

  1. Matus A. 2000. Actin-based plasticity in dendritic spines. Science290, 754-758.
  2. Carlisle H.J. and Kennedy M.B. 2005. Spine architecture and synaptic plasticity. Trends Neurosci28, 182-187.
  3. Kasai H. et al. 2003. Structure-stability-function relationships of dendritic spines. Trends Neurosci26, 360-368.
  4. Matsuzaki M. et al. 2004. Structural basis of long-term potentiation in single dendritic spines. Nature429, 761-766.
  5. Tolias K.F. et al. 2011. Control of synapse development and plasticity by Rho GTPase regulatory proteins. Prog. Neurobiol. 94, 133-148.
  6. Hedrick N.G. and Yasuda R. 2017. Regulation of Rho GTPase proteins during spine structural plasticity for the control of local dendritic plasticity. Curr. Opin. Neurobiol. 45, 193-201.
  7. Woolfrey K.M. and Srivastava D.P. 2016. Control of dendritic spine morphological and functional plasticity by small GTPases. Neural Plasticity. 2016, 3025948. DOI: 10.1155/2016/3025948.
  8. Dierssen M. and Ramakers G.J. 2006. Dendritic pathology in mental retardation: from molecular genetics to neurobiology. Genes Brain Behav. 5 (Suppl. 2), 48-60.
  9. Penzes P. and Van Leeuwen J.E. 2011. Impaired regulation of synaptic actin cytoskeleton in Alzheimer's disease. Brain Res. Rev. 67, 184-192.
  10. Penzes P. et al. 2011. Dendritic spine pathology in neuropsychiatric disorders. Nat. Neurosci. 14, 285-292.
  11. Tashiro A. and R. Yuste. 2004. Regulation of dendritic spine motility and stability by Rac1 and Rho kinase: evidence for two forms of spine motility. Mol. Cell. Neurosci. 26, 429–440.
  12. Rex C.S. et al. 2009. Different Rho GTPase–dependent signaling pathways initiate sequential steps in the consolidation of long-term potentiation. J. Cell Biol. 186, 85–97.
  13. Hodges J.L. et al. 2011. Myosin IIb activity and phosphorylation status determines dendritic spine and post-synaptic density morphology. PLoS ONE. 6, e24149.
  14. Murakoshi H. et al. 2011. Local, persistent activation of Rho GTPases during plasticity of single dendritic spines. Nature. 472, 100-104.
  15. Newell-Litwa K. et al. 2015. ROCK1 and 2 differentially regulate actomyosin organization to drive cell and synaptic polarity. J. Cell Biol. 210, 225–242.
  16. Hedrick N.G. et al. 2016. Rho GTPase complementation underlies BDNF-dependent homo- and heterosynaptic plasticity. Nature. 538, 104-108.
  17. Penzes P. et al. 2008. Convergent CaMK and RacGEF signals control dendritic structure and function. Trends Cell Biol. 18, 405-413.
  18. Herring B.E. and Nicoll R.A. 2016. Kalirin and Trio proteins serve critical roles in excitatory synaptic transmission and LTP. Proc. Natl. Acad. Sci. U.S.A. 113, 2264-2269.
  19. Bliss T.V. and Collingridge G.L. 1993. A synaptic model of memory: long-term potentiation in the hippocampus. Nature. 361, 31–39.
  20. Bliss T.V. and Collingridge G.L. 2013. Expression of NMDA-receptor-dependent LTP in the hippocampus: Bridging the divide. Mol. Brain. 6, 5.
  21. Xie Z. et al. 2007. Kalirin-7 controls activity-dependent structural and functional plasticity of dendritic spines. Neuron. 56, 640-656.
  22. Penzes P. and Jones K.A. 2008. Dendritic spine dynamics – a key role for kalirin-7. Trends Neurosci. 31, 419-427.
  23. Ma X.-M. et al. 2008. Kalirin-7 is required for synaptic structure and function. J. Neurosci. 28, 12368–12382.
  24. Ma X. M. et al. 2011. Kalirin-7, an important component of excitatory synapses, is regulated by estradiol in hippocampal neurons. Hippocampus. 21, 661–677.
  25. Tolias K.F. et al. 2005. The Rac1-GEF Tiam1 couples the NMDA receptor to the activity-dependent development of dendritic arbors and spines. Neuron. 45, 525-538.
  26. Zhang H. and Macara I.G. 2006. The polarity protein PAR-3 and TIAM1 cooperate in dendritic spine morphogenesis. Nat. Cell Biol. 8, 227–237.
  27. Schwechter B. et al. 2013. RasGRF2 Rac-GEF activity couples NMDA receptor calcium flux to enhanced synaptic transmission. Proc. Natl. Acad. Sci. U.S.A. 110, 14462-14467.
  28. Kim J.-Y. et al. 2011. The RhoG/ELMO1/Dock180 signaling module is required for spine morphogenesis in hippocampal neurons. J. Biol. Chem. 286, 37615-37624.
  29. Fu W.-Y. et al. 2007. Cdk5 regulates EphA4-mediated dendritic spine retraction through an ephexin1-dependent mechanism. Nat. Neurosci. 10, 67-76.
  30. Penzes P. et al. 2003. Rapid induction of dendritic spine morphogenesis by trans-synaptic ephrinB-ephB receptor activation of the Rho-GEF kalirin. Neuron. 37, 263-274.
  31. Xin X. et al. 2008. Regulation of kalirin by cdk5. J. Cell Sci. 121, 2601–2611.
  32. Tolias K.F. et al. 2007. The Rac1 guanine nucleotide exchange factor Tiam1 mediates EphB receptor-dependent dendritic spine development. Proc. Natl. Acad. Sci. U.S.A. 104, 7265-7270.
  33. Um K. et al. 2014. Dynamic control of excitatory synapse development by a Rac1 GEF/GAP regulatory complex. Dev. Cell. 29, 701-715.
  34. Jaudon F. et al. 2015. The RhoGEF DOCK10 is essential for dendritic spine morphogenesis. Mol. Biol. Cell. 26, 2112-2127.
  35. Kang M.-G. et al. 2009. AMPA receptor and GEF-H1/Lfc complex regulates dendritic spine development through RhoA signaling cascade. Proc. Natl. Acad. Sci. U.S.A. 106, 3549-3554.
  36. Margolis S.S. et al. 2010. EphB-mediated degradation of the RhoA GEF ephexin5 relieves a developmental brake on excitatory synapse formation. Cell. 143, 442-455.