Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Rho kinase, a promising drug target for neurological disorders

Key Points

  • A large number of Rho kinase (ROCK) inhibitors have been developed during the past decade, and to our knowledge none of these distinguish between the two ROCK isoforms, ROCKI and ROCKII. In addition, the commercially available inhibitors (fasudil, Y-27632 and H-1152) target other closely related kinases. Despite this, the reasonably selective ROCK inhibitor fasudil has been marketed in Japan since 1995 for the treatment of cerebral vasospasm after subarachnoid haemorrhage and is currently undergoing clinical evaluation in angina pectoris.

  • Most interestingly, knockouts of either ROCK genes yielded the surprising result that despite their embryonic phenotypes, homozygous mice are able to develop to maturity and are fertile and healthy.

  • In different animal models of spinal-cord injury, ROCK inhibitors induced neurite outgrowth and sprouting and enhanced tissue preservation. Both mechanisms account for the improved regeneration and functional recovery of the inhibitor-treated animals in comparison to control animals. These results turn ROCK inhibitors into the currently most potent small-molecule stimulators of neuroregeneration.

  • In a transgenic mouse model of Alzheimer's disease, the ROCK inhibitor Y-27632 decreased the amount of toxic Aβ42, thereby interfering with the pathological degenerative process. In animal models of neuropathic pain and in multiple sclerosis models, data have shown that inhibiting the Rho–ROCK pathway has positive effects, which indicate an excellent therapeutic opportunity.

  • No crystallographic structural data of the kinase domain of ROCKs are currently available but a structural surrogate kinase approach with protein kinase A yielded valuable information that explains the binding mode of three different ROCK inhibitors (fasudil, Y-27632 and H-1152). Crystallographic data on the ROCK kinase structure will help to design more selective and more potent ROCK inhibitors.

Abstract

Rho kinases (ROCKs), the first Rho effectors to be described, are serine/threonine kinases that are important in fundamental processes of cell migration, cell proliferation and cell survival. Abnormal activation of the Rho/ROCK pathway has been observed in various disorders of the central nervous system. Injury to the adult vertebrate brain and spinal cord activates ROCKs, thereby inhibiting neurite growth and sprouting. Inhibition of ROCKs results in accelerated regeneration and enhanced functional recovery after spinal-cord injury in mammals, and inhibition of the Rho/ROCK pathway has also proved to be efficacious in animal models of stroke, inflammatory and demyelinating diseases, Alzheimer's disease and neuropathic pain. ROCK inhibitors therefore have potential for preventing neurodegeneration and stimulating neuroregeneration in various neurological disorders.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Molecular structure of Rho kinase (ROCK) I and II.
Figure 2: Activation mechanism of Rho kinase (ROCK).
Figure 3: Major ROCK substrates.
Figure 4: Spinal-cord injury.
Figure 5: Growth-cone collapse.
Figure 6: Structures of Rho kinase (ROCK) inhibitors.

Similar content being viewed by others

References

  1. Van Aelst, L. & D'Souza-Schorey, C. Rho GTPases and signaling networks. Genes Dev. 11, 2295–2322 (1997).

    CAS  PubMed  Google Scholar 

  2. Burridge, K. & Wennerberg, K. Rho and Rac take center stage. Cell 116, 167–179 (2004).

    CAS  PubMed  Google Scholar 

  3. Bishop, A. & Hall, A. Rho GTPases and their effector proteins. Biochem. J. 348, 241–255 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Rossman, K. L., Der, C. J. & Sondek, J. GEF means go: turning on Rho GTPases with guanine nucleotide-exchange factors. Nature Rev. Mol. Cell Biol. 6, 167–180 (2005).

    CAS  Google Scholar 

  5. Matsui et al. Rho-associated kinase, a novel serine/threonine kinase, as a putative target for the small GTP binding protein Rho. EMBO J. 15, 2208–2216 (1996). An important paper describing identification of ROCK for the first time.

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Ishizaki et al. The small GTP-binding proteinRho binds to and activates a 160 kDa Ser/Thr protein kinase homologous to myotonic dystrophy kinase. EMBO J. 15, 1885–1893 (1996). Seminal paper, which along with reference 7 provides the first description of ROCK.

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Leung, T., Manser, E., Tan, L. & Lim, L. A novel serine/threonine kinase binding the Ras-related RhoA GTPase which translocates the kinase to the peripheral membranes. J. Biol. Chem. 270, 29051–29054 (1996).

    Google Scholar 

  8. Nagawa, O. et al. ROCK-I and ROCK-II, two isoforms of Rho-associated coiled-coil forming protein serine/threonine kinase in mice. FEBS Lett. 392, 189–193 (1996).

    Google Scholar 

  9. Amano, M., Fukata, Y. & Kaibuchi, K. Regulation and function of Rho-associated kinase. Exp. Cell Res. 261, 44–51 (2000).

    CAS  PubMed  Google Scholar 

  10. Riento, K. & Ridley, A. ROCKs: multifunctional kinases in cell behaviour. Nature Rev. Mol. Cell. Bio. 4, 446–456 (2003). Outstanding review that summarizes the known cellular functions of ROCKs.

    CAS  Google Scholar 

  11. Hashimoto, R. et al. Distribution of Rho–kinase in the bovine brain. Biochem. Biophys. Res. Commun. 263, 575–579 (1999).

    CAS  PubMed  Google Scholar 

  12. Komagome, R., Kimura, K. & Saito, M. Postnatal changes in Rho and Rho–related proteins in the mouse brain. Jpn J. Vet. Res. 47, 127–133 (2000).

    CAS  PubMed  Google Scholar 

  13. Amano, M. et al. The COOH terminus of Rho-kinase negatively regulates Rho-kinase activity. J. Biol. Chem. 274, 32418–32424 (1999).

    CAS  PubMed  Google Scholar 

  14. Tan, I., Seow, K. T., Lim, L. & Leung, T. Intermolecular and intramolecular interactions regulate the catalytic activity of myotonic dystrophy kinase-related Cdc42-binding kinase. Mol. Cell. Biol. 21, 2767–2778 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Bush, E. W., Helmke, S. M., Birnbaum, R. A. & Perryman, M. B. Myotonic dystrophy protein kinase domains mediate localization, oligomerization, novel catalytic activity and autoinhibition. Biochemistry 39, 8480–8490 (2000).

    CAS  PubMed  Google Scholar 

  16. Shimizu, T. Ihara, K., Maesaki, R., Amano, M., Kaibuchi, K. & Hakoshima, T. Parallel coiled-coil association of the RhoA binding domain in Rho-kinase. J. Biol. Chem. 278, 46046–46051 (2003).

    CAS  PubMed  Google Scholar 

  17. Dvorsky, R., Blumenstein, L., Vetter, I. & Ahmadian, M. R. Structural insights into the interaction of ROCKI with the switch regions of RhoA. J. Biol. Chem. 279, 7098–7104 (2004).

    CAS  PubMed  Google Scholar 

  18. Doran, J., Lui, X., Taslimi, P. Saadat, A. & Fox, T. New insights into the structure-function relationships of Rho-associated kinase: A thermodynamic and hydrodynamic study of the dimer-to-monomer transition and its kinetic implications. Biochem. J. 384, 255–262 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Turner, M. S., Fen Fen, L., Trauger, J. W., Stephens, J. & LoGrasso, P. Characterization and purification of truncated human Rho-kinase II expressed in Sf-21 cells. Arch. Biochem. Biophys. 405, 13–20 (2002).

    CAS  PubMed  Google Scholar 

  20. Fu, X., Gong, M. C., Jia, T., Somlyo A. V. & Somlyo, A. P. The effects of the Rho-kinase inhibitor Y-27632 on arachidonic acid-, GTPγS-, and phorbol ester-induced Ca-sensitization of smooth muscle. FEBS Lett. 440, 183–187 (1998).

    CAS  PubMed  Google Scholar 

  21. Shirao, S. et al. Sphingosylphosphorylcholine is a novel messenger for Rho-kinase-mediated Ca2+ sensitization in the bovine cerebral artery: unimportant role for protein kinase C. Circ. Res. 91, 112–119 (2002).

    CAS  PubMed  Google Scholar 

  22. Sebbagh, M. et al. Caspase-3-mediated cleavage of ROCKI induces MLC phosphorylation and apoptotic membrane blebbing. Nature Cell Biol. 3, 346–352 (2001).

    CAS  PubMed  Google Scholar 

  23. Coleman, M. L. et al. Membrane blebbing during apoptosis results from Caspase-mediated activation of ROCKI. Nature Cell Biol. 3, 339–345 (2001).

    CAS  PubMed  Google Scholar 

  24. Leung, T., Chen, X. Q., Manser, E. & Lim, L. The p160 RhoA-binding kinase ROKα is a member of a kinase family and is involved in the reorganization of the cytoskeleton. Mol. Cell. Biol. 16, 5313–5327 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Somlyo, A. P. & Somlyo, A. V. Signal transduction by G-proteins, rho-kinase and protein phosphatase to smooth muscle and non-muscle myosin II. J. Physiol. 522, 177–185 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Kimura, K. et al. Regulation of myosin phosphatase by Rho and Rho-associated kinase (Rho-kinase). Science 273, 245–248 (1996).

    CAS  PubMed  Google Scholar 

  27. Sumi, T., Matsumoto, K. & Nakamura, T. Specific activation of LIM kinase 2 via phosphorylation of threonine 505 by ROCK, a Rho-dependent protein kinase. J. Biol. Chem. 276, 670–676 (2001).

    CAS  PubMed  Google Scholar 

  28. Ohashi, K. et al. Rho-associated kinase ROCK activates LIM-kinase 1 by phosphorylation at threonine 508 within the activation loop. J. Biol. Chem. 275, 3577–3582 (2000).

    CAS  PubMed  Google Scholar 

  29. Goto, H. et al. Phosphorylation of vimentin by Rho-associated kinase at a unique amino-terminal site that is specificially phosphorylated during cytokinesis. J. Biol. Chem. 273, 11728–11736 (1998).

    CAS  PubMed  Google Scholar 

  30. Kosako, H. et al. Phosphorylation of glial fibrillary acidic protein at the same sites by cleavage furrow kinase and Rho-associated kinase. J. Biol. Chem. 272, 10333–10336 (1997).

    CAS  PubMed  Google Scholar 

  31. Hashimoto, R. et al. Domain and site-specific phosphorylation of bovine NF-L by Rho-associated kinase. Biochem. Biophys. Res. Commun. 245, 407–411 (1998).

    CAS  PubMed  Google Scholar 

  32. Amano, M. et al. Identification of Tau and MAP2 as novel substrates of Rho-kinase and myosin phosphatase. J. Neurochem. 87, 780–790 (2003).

    CAS  PubMed  Google Scholar 

  33. Arimura, N. et al. Phosphorylation of collapsin response mediator protein-2 by Rho-kinase. Evidence for two seperate signaling pathway for growth cone collapse. J. Biol. Chem. 275, 23973–23980 (2000).

    CAS  PubMed  Google Scholar 

  34. Shimizu, Y. et al. ROCK-I regulates closure of the eyelids and ventral body wall by inducing assembly of actomyosin bundles. J. Cell Biol. 168, 941–963 (2005). First description of ROCKI-knockout mice with a milder phenotype, as expected based on the many developmental functions the kinase has.

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Thumkeo, D. et al. Targeted disruption of the mouse Rho-associated kinase 2 gene results in intrauterine growth retardation and fetal death. Mol. Cell. Biol. 23, 5043–5055 (2003). This paper describes the surprising result that ROCKII-knockout mice develop normally after surviving the embryonic stage.

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Shibuya, M. et al. Effect of AT877 on cerebral vasospasm after aneurysmal subarachnoid hemorrhage. Results of a prospective placebo-controlled double-blind trial. J. Neurosurg. 76, 571–577 (1992).

    CAS  PubMed  Google Scholar 

  37. Shimokawa, M. et al. Anti-anginal effect of fasudil, a Rho-kinase inhibitor, in patients with stable effort angina: a multicenter study. J Cardiovasc Pharmacol. 40, 751–761 (2002).

    CAS  PubMed  Google Scholar 

  38. Hu, E. & Lee, D. ROCK inhibitors as potential therapeutic agents for cardiovascular diseases. Curr. Opin. Investig. Drugs. 4, 1065–1075 (2003).

    CAS  PubMed  Google Scholar 

  39. Wettschureck, N. & Offermanns, S. Rho/Rho-kinase mediated signaling in physiology and pathophysiology. J. Mol. Med. 80, 629–638 (2002). An excellent review that extensively describes the many therapeutic indication areas for Rho and ROCK inhibitors.

    CAS  PubMed  Google Scholar 

  40. Somlyo, A. P. & Somlyo, A. V. Ca2+ sensitivity of smooth muscle and nonmuscle myosin II: modulated by G proteins, kinases, and myosin phosphatase. Physiol. Rev. 83, 1325–1358 (2003).

    CAS  PubMed  Google Scholar 

  41. Sylvester, J. T. The tone of pulmonary smooth muscle: ROK and Rho music? Am. J. Physiol. Lung Cell. Mol. Physiol. 287, L624–L630 (2004).

    CAS  PubMed  Google Scholar 

  42. Aznar, S., Fernandez-Valeron, P., Espina, C. & Lacal, J. C. Rho GTPases: potential candidates for anticancer therapy. Cancer Lett. 206, 181–191 (2004).

    CAS  PubMed  Google Scholar 

  43. Sharpe, C. C. & Hendry, B. M. Signaling: focus on Rho in renal disease. J. Am. Soc. Nephrol. 14, 261–264 (2003).

    PubMed  Google Scholar 

  44. Andersson, K. E. & Hedlund, P. New directions for erectile dysfunction therapies. Int. J. Impot. Res. 14 (Suppl. 1) S82–S92 (2002).

    PubMed  Google Scholar 

  45. Dobkin, B. H. & Havton, L. A. Basic advances and new avenues in therapy of spinal cord injury. Annu. Rev. Med. 55, 255–282 (2004). Excellent summary of the field of spinal-cord injury with an overview of current therapeutic approaches.

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Harkey, H. L., White, E. A., Tibbs, R. E. Jr & Haines, D. E. A clinician's view of spinal cord injury. Anat. Rec. B New Anat. 271, 41–48 (2003).

    PubMed  Google Scholar 

  47. McDonald, J. W. & Sadowsky, C. Spinal-cord injury. Lancet 359, 417–425 (2002).

    PubMed  Google Scholar 

  48. Dumont, R. J. et al. Acute spinal cord injury, part I: pathophysiologic mechanisms. Clin. Neuropharmacol. 24, 254–264 (2001).

    CAS  PubMed  Google Scholar 

  49. Schwab, M. E. Nogo and axon regeneration. Curr. Opin. Neurobiol. 14, 118–124 (2004).

    CAS  PubMed  Google Scholar 

  50. Fournier, A. E., GrandPre, T. & Strittmatter, S. M. Identification of a receptor mediating Nogo-66 inhibition of axonal regeneration. Nature 409, 341–346 (2001). An outstanding paper that describes the discovery of the first receptor for NOGO-A.

    CAS  PubMed  Google Scholar 

  51. Domeniconi, M. et al. Myelin-associated glycoprotein interacts with the Nogo66 receptor to inhibit neurite outgrowth. Neuron 35, 283–290 (2002).

    CAS  PubMed  Google Scholar 

  52. Liu, B. P., Fournier, A., GrandPre, T. & Strittmatter, S. M. Myelin-associated glycoprotein as a functional ligand for the Nogo-66 receptor. Science 297, 1190–1193 (2002).

    CAS  PubMed  Google Scholar 

  53. Wang, K. C. et al. Oligodendrocyte-myelin glycoprotein is a Nogo receptor ligand that inhibits neurite outgrowth. Nature 417, 941–944 (2002).

    CAS  PubMed  Google Scholar 

  54. Venkatesh, K. et al. The Nogo-66 receptor homolog NgR2 is a sialic acid-dependent receptor selective for myelin-associated glycoprotein. J. Neurosci. 25, 808–822 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Wang, K. C., Kim, J. A., Sivasankaran, R., Segal, R. & He, Z. P75 interacts with the Nogo receptor as a co-receptor for Nogo, MAG and OMgp. Nature 420, 74–78 (2002).

    CAS  PubMed  Google Scholar 

  56. Wong, S. T. et al. A p75(NTR) and Nogo receptor complex mediates repulsive signaling by myelin-associated glycoprotein. Nature Neurosci. 5, 1302–1308 (2002).

    CAS  PubMed  Google Scholar 

  57. Mi, S. et al. LINGO-1 is a component of the Nogo-66 receptor/p75 signaling complex. Nature Neurosci. 7, 221–228 (2004).

    CAS  PubMed  Google Scholar 

  58. Park, J. B. et al. A TNF receptor family member, TROY, is a coreceptor with Nogo receptor in mediating the inhibitory activity of myelin inhibitors. Neuron 45, 345–351 (2005).

    CAS  PubMed  Google Scholar 

  59. Shao, Z. et al. TAJ/TROY, an orphan TNF receptor family member, binds Nogo-66 receptor 1 and regulates axonal regeneration. Neuron 45, 353–359 (2005).

    CAS  PubMed  Google Scholar 

  60. Schweigreiter, R. et al. Versican V2 and the central inhibitory domain of Nogo-A inhibit neurite growth via p75NTR/NgR-independent pathways that converge at RhoA. Mol. Cell. Neurosi. 27, 163–174 (2004).

    CAS  Google Scholar 

  61. Niederost, B., Oertle, T., Fritsche, J., McKinney, R. A. & Bandtlow, C. E. Nogo-A and myelin-associated glycoprotein mediate neurite growth inhibition by antagonistic regulation of RhoA and Rac1. J. Neurosci. 22, 10368–10376 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Schwab, J. M. et al. Spinal cord injury induces lesional expression pattern of the Repulsive Guidance Molecule (RGM). Eur. J. Neurosci. (in the press).

  63. He, Z. & Koprivica, V. The Nogo signaling pathway for regeneration block. Annu. Rev. Neurosci. 27, 341–368 (2004).

    CAS  PubMed  Google Scholar 

  64. Kullander, K. et al. Role of EphA4 and EphrinB3 in local neuronal circuits that control walking. Science 299, 1889–1892 (2003).

    CAS  PubMed  Google Scholar 

  65. Moreau-Fauvarque, C. et al. The transmembrane semaphorin Sema4D/CD100, an inhibitor of axonal growth, is expressed on oligodendrocytes and upregulated after CNS lesion. Neuroscience 23, 9229–9239 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Willson, C. A. et al. Upregulation of EphA receptor expression in the injured adult rat spinal cord. Cell Transplant. 11, 229–239 (2002).

    PubMed  Google Scholar 

  67. Miranda, J. D. et al. Induction of Eph B3 after spinal cord injury. Exp. Neurol. 156, 218–222 (1999).

    CAS  PubMed  Google Scholar 

  68. Monnier, P. P., Sierra, A., Schwab, J. M., Henke-Fahle, S. & Mueller, B. K. The Rho/ROCK pathway mediates neurite growth-inhibitory activity associated with the chondroitin sulfate proteoglycans of the CNS glial scar. Mol. Cell. Neurosci. 22, 319–330 (2003).

    CAS  PubMed  Google Scholar 

  69. Borisoff, J. F. et al. Suppression of Rho-kinase activity promotes axonal growth on inhibitory CNS substrates. Mol Cell Neurosci. 22, 405–416 (2003).

    CAS  PubMed  Google Scholar 

  70. Swiercz, J. M., Kuner, R., Behrens, J. & Offermanns, S. Plexin-B1 directly interacts with PDZ-RhoGEF/LARG to regulate RhoA and growth cone morphology. Neuron 35, 51–63 (2002).

    CAS  PubMed  Google Scholar 

  71. Shamah, S. M. et al. EphA receptors regulate growth cone dynamics through the novel guanine nucleotide exchange factor ephexin. Cell 105, 233–244 (2001).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Brabeck, C. et al. Lesional expression of RhoA and RhoB following traumatic brain injury in humans. J. Neurotrauma 21, 697–706 (2004).

    PubMed  Google Scholar 

  74. Aimone, J., Leisure L., Perreau, V. & Tallmair, M. Spatial an temporal gene epression profiling of the contused rat spinal cord. Exp. Neurol. 189, 204–221 (2004).

    CAS  PubMed  Google Scholar 

  75. Dubreuil, C. I., Winton, M. J. & McKerracher, L. Rho activation patterns after spinal cord injury and the role of activated Rho in apoptosis in the central nervous system. J. Cell Biol. 162, 233–243 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Madura, T. et al. Activation of Rho in the injured axons following spinal cord injury. EMBO Rep. 5, 412–417 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Dergham, P. et al. Rho signaling pathway targeted to promote spinal cord repair. J. Neurosci. 22, 6570–6577 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Hara, M. et al. Protein kinase inhibition by fasudil hydrochloride promotes neurological recovery after spinal cord injury in rats. J. Neurosurg. Spine 93, 94–101 (2000).

    CAS  Google Scholar 

  79. Fournier, A. E., Takizawa, B. T. & Strittmatter, S. M. ROCK inhibition enhances axonal regeneration in the injured CNS. J. Neurosci. 23, 1416–1423 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Sung, J. K. et al. A possible role of RhoA/Rho-kinase in experimental spinal cord injury in rat. Brain Res. 959, 29–38 (2003).

    CAS  PubMed  Google Scholar 

  81. Tanaka, H. et al. Cytoplasmic p21(Cip1/WAF1) enhances axonal regeneration and functional recovery after spinal cord injury in rats. Neuroscience 127, 155–164 (2004). Important paper showing that three different ROCK inhibitors result in enhanced functional recovery in a moderate–severe rat spinal-cord injury model.

    CAS  PubMed  Google Scholar 

  82. Citron, M. Strategies for disease modification in Alzheimer's disease. Nature Rev. Neurosci. 5, 677–685 (2004).

    CAS  Google Scholar 

  83. Selkoe, D. J. Cell biology of protein misfolding: the examples of Alzheimer's and Parkinson's diseases. Nature Cell Biol. 6, 1054–1061 (2004).

    CAS  PubMed  Google Scholar 

  84. McGeer, P. L., Schulzer, M. & McGeer, E. G. Arthritis and anti-inflammatory agents as possible protective factors for Alzheimer's disease: a review of 17 epidemiologic studies. Neurology 47, 425–432 (1996).

    CAS  PubMed  Google Scholar 

  85. Anthony, J. C. et al. Reduced prevalence of AD in users of NSAIDs and H2 receptor antagonists: the Cache County study. Neurology 54, 2066–2071 (2000).

    CAS  PubMed  Google Scholar 

  86. Weggen, S. et al. A subset of NSAIDs lower amyloidogenic Aβ42 independently of cyclooxygenase activity. Nature 414, 212–216 (2001).

    CAS  PubMed  Google Scholar 

  87. Eriksen, J. L. et al. NSAIDs and enantiomers of flurbiprofen target γ-secretase and lower Aβ42 in vivo. J. Clin. Invest. 112, 440–449 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Zhou, Y. et al. Nonsteroidal anti-inflammatory drugs can lower amyloidogenic Aβ42 by inhibiting Rho. Science 302, 1215–1217 (2003). A key paper that describes the effect of Rho or ROCK inhibitors on Aβ42 secretion both in vitro and in vivo.

    CAS  PubMed  Google Scholar 

  89. Pedrini, S. et al. Modulation of statin-activated shedding of Alzheimer APP ectodomain by ROCK. PLoS Med. 2, 69–78 (2005).

    CAS  Google Scholar 

  90. Neuhaus, O., Stuve, O., Zamvil, S. S. & Hartung, H. P. Are statins a treatment option for multiple sclerosis? Lancet Neurol. 3, 369–371 (2004).

    CAS  PubMed  Google Scholar 

  91. Hendriks, J. J. et al. Flavonoids influence monocytic GTPase activity and are protective in experimental allergic encephalitis. J. Exp. Med. 200, 1667–1672 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Greenwood, J. et al. Lovastatin inhibits brain endothelial cell Rho-mediated lymphocyte migration and attenuates experimental autoimmune encephalomyelitis. FASEB J. 17, 905–907 (2003).

    CAS  PubMed  Google Scholar 

  93. Walters, C. E. et al. Inhibition of Rho GTPases with protein prenyltransferase inhibitors prevents leukocyte recruitment to the central nervous system and attenuates clinical signs of disease in an animal model of multiple sclerosis. J. Immunol. 168, 4087–4094 (2002).

    CAS  PubMed  Google Scholar 

  94. Honing, H. et al. RhoA activation promotes transendothelial migration of monocytes via ROCK. J. Leukoc. Biol. 75, 523–528 (2004).

    CAS  PubMed  Google Scholar 

  95. Kakulas, B. A. A review of the neuropathology of human spinal cord injury with emphasis on special features. J. Spinal Cord Med. 22, 119–124 (1999). An excellent report on spinal-cord injury with autopsy data and a description of the most relevant injury-related mechanisms. Most importantly, this paper provides an overview of the amount of motor and sensory fibre necessary for residual motor and sensory function.

    CAS  PubMed  Google Scholar 

  96. Kohama, I. et al. Transplantation of cryopreserved adult human Schwann cells enhances axonal conduction in demyelinated spinal cord. J. Neurosci. 21, 944–950 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Melendez-Vasquez, C. V., Einheber, S. & Salzer, J. L. ROCK regulates schwann cell myelination and formation of associated axonal domains. J. Neurosci. 24, 3953–3963 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Minagar, A., Toledo, E. G., Alexander, J. S. & Kelley, R. E. Pathogenesis of brain and spinal cord atrophy in multiple sclerosis. J. Neuroimaging 14, 5s–10s (2004).

    PubMed  Google Scholar 

  99. Trapp, B. D. et al. Axonal transection in the lesions of multiple sclerosis. N. Engl. J. Med. 338, 278–285 (1998).

    CAS  PubMed  Google Scholar 

  100. Toshima Y, Satoh S, Ikegaki I, Asano T. A new model of cerebral microthrombosis in rats and the neuroprotective effect of a Rho-kinase inhibitor. Stroke 31, 2245–2250 (2000).

    CAS  PubMed  Google Scholar 

  101. Satoh, S. et al. Pharmacological profile of hydroxy fasudil as a selective ROCK inhibitor on ischemic brain damage. Life Sci. 69, 1441–1453 (2001).

    CAS  PubMed  Google Scholar 

  102. Kitaoka, Y. et al. Involvement of RhoA and possible neuroprotective effect of fasudil, a ROCK inhibitor, in NMDA-induced neurotoxicity in the rat retina. Brain Res. 1018, 111–118 (2004).

    CAS  PubMed  Google Scholar 

  103. Lee, J. K., Kim, J. E., Sivula, M. & Strittmatter, S. M. Nogo receptor antagonism promotes stroke recovery by enhancing axonal plasticity. J. Neurosci. 24, 6209–6217 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Wiessner, C. et al. Anti-Nogo-A antibody infusion 24 hours after experimental stroke improved behavioral outcome and corticospinal plasticity in normotensive and spontaneously hypertensive rats. J. Cereb. Blood Flow Metab. 23, 154–165 (2003).

    CAS  PubMed  Google Scholar 

  105. Schwab, J. M. et al. CNS injury-induced RGM (Repulsive Guidance Molecule) expression in the adult human brain. Arch. Neurol. (in the press).

  106. Ji, R. R. & Strichartz, G. Cell signaling and the genesis of neuropathic pain. Science STKE 252, re 14, 1–19 (2004).

    Google Scholar 

  107. Inoue, M. et al. Initiation of neuropathic pain requires lysophosphatidic acid receptor signaling. Nature Med. 10, 712–718 (2004).

    CAS  PubMed  Google Scholar 

  108. Ramer, L. M., Borisoff, J. F. & Ramer, M. S. Rho-kinase inhibition enhances axonal plasticity and attenuates cold hyperalgesia after dorsal rhizotomy. J Neurosci. 24, 10796–10805 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Tatsumi, S. et al. Involvement of Rho-kinase in inflammatory and neuropathic pain through phosphorylation of myristoylated alanine-rich C-kinase substrate (MARCKS). Neuroscience 131, 491–498 (2005).

    CAS  PubMed  Google Scholar 

  110. Uehata, M. et al. Calcium sensitization of smooth muscle mediated by a Rho-associated protein kinase in hypertension. Nature 389, 990–994 (1997). A landmark paper describing the first low-molecular-mass Rho-kinase inhibitor.

    CAS  PubMed  Google Scholar 

  111. Uehata M., Ono, T., Satoh, H., Yamagami, K. & Kawahara, T. Medicines comprising ROCK inhibitor. European Patent EP0956865A1 (1998).

  112. Shimokawa, H. Rho-kinase as a novel therapeutic target in treatment of cardiovascular diseases. J. Cardiovasc. Pharmacol. 39, 319–327 (2002).

    CAS  PubMed  Google Scholar 

  113. Hidaka, H., Matsuura, A. & Matsuzaki, T. Isoquinoline derivatives and drugs. European Patent EP0885888(A1) (1997).

  114. Iwakubo, M. & Okada, Y. Isoquinoline derivatives having kinase inhibitory activity and drugs containing the same. World Patent WO 2004024717A1 (2004).

  115. Takanashi, S., Naito, Y., Tanaka, H., Uehata, M. & Katayama, K. Amide compounds and use thereof. European Patent EP1270570A1 (2001).

  116. Takanashi, S. & Hamaguchi, S. ROCK inhibitor comprising amide compound. Japanese Patent JP2003073357 A2 (2003).

  117. Gingras, K. et al. Bioaxone. Synthesis and evaluation of 4-(1-aminoalkyl)-N-(4-pyridyl)–cyclohexanecarboxamides as ROCK inhibitors and neurite outgrowth promoters. Bioorg. Med. Chem. Lett. 14, 4931–4934 (2004).

    CAS  PubMed  Google Scholar 

  118. McKerracher, L., Thouin, E. & Lubell, W. D. 1,4-substituted cyclohexane derivatives. World Patent WO 2004022541A1 (2004).

  119. Takami, A., Iijima, H., Iwakubo, M. & Okada, Y. Nitrogen-containing compounds having kinase inhibitory activity and drugs containing the same. European Patent EP1256574A1 (2001).

  120. Nagarathnam, D. Wang, C. H. ROCK inhibitors. World Patent WO2002076977A2 (2002).

  121. Feurer, A. et al. Heteroaryloxy-substituted phenylaminopyrimidines as ROCK inhibitors. World Patent WO2004039796A1 (2004).

  122. Ishizaki, T. et al. Pharmacological properties of Y-27632, a specific inhibitor of Rho-associated kinases. Molecular Pharmacology 57, 976–983 (2000).

    CAS  PubMed  Google Scholar 

  123. Davies, S. P., Reddy, H., Caivano, M. & Cohen, P. Specificity and mechanism of action of some commonly used protein kinase inhibitors. Biochem. J. 351, 95–105 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Sazaki, Y., Suzuki, M. & Hidaka, H. The novel and specific Rho-kinase inhibitor (S)-(+)-2-methyl-1-[(4-methyl-5-isoquinoline)sulfonyl]-homopiperazine as a probing molecule for Rho-kinase-involved pathway. Pharmacol. Ther. 93, 225–232 (2002).

    Google Scholar 

  125. Ikenoya, M. et al. Inhibition of Rho-kinase-induced myristoylated alanine-rich C kinase substrate (MARCKS) phosphorylation in human neuronal cells by H-1152, a novel and specific Rho-kinase inhibitor. J. Neurochem. 81, 9–16 (2002).

    CAS  PubMed  Google Scholar 

  126. Breitenlechner, C. et al. Protein kinase A in complex with Rho-kinase inhibitors Y-27632, Fasudil, and H–1152P: structural basis of selectivity. Structure 11, 1595–1607 (2003).

    CAS  PubMed  Google Scholar 

  127. Hirooka, Y. & Shimokawa, H. Therapeutic potential of rho-kinase inhibitors in cardiovascular diseases. Am. J. Cardiovasc. Drugs 5, 31–39 (2005).

    CAS  PubMed  Google Scholar 

  128. Lu, M. et al. Assessment of absortion of ROCK inhibitor at different sites of the human gastrointestinal tract. Am. Soc. Clin. Pharmacol. Ther. P6, PI–13 (2004).

    Google Scholar 

  129. Takanashi, Y. et al. Efficacy of intrathecal liposomal fasudil for experimental cerebral vasospasm after subarachnoid hemorrhage. Neurosurgery 48, 894–900 (2001).

    CAS  PubMed  Google Scholar 

  130. Ishida, T., Takanashi, Y., Doi, H., Yamamoto, I. & Kiwada, H. Encapsulation of an antivasospastic drug, fasudil, into liposomes, and in vitro stability of the fasudil-loaded liposomes. Int. J. Pharm. 232, 59–67 (2002).

    CAS  PubMed  Google Scholar 

  131. Nagatoya, K. et al. Y-27632 prevents tubulointerstitial fibrosis in mouse kidneys with unilateral ureteral obstruction. Kidney Int. 61, 1684–1695 (2002).

    CAS  PubMed  Google Scholar 

  132. Narumiya, S., Ishizaki, T. & Uehata, M. Use and properties of ROCK-specific inhibitor Y-27632. Meth Enzymol. 325, 273–284 (2000).

    CAS  Google Scholar 

  133. Wei, L. et al. Rho kinases play an obligatory role in vertebrate embryonic organogenesis. Development 128, 2953–2962 (2001).

    CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Bernhard K. Mueller.

Related links

Related links

DATABASES

Entrez Gene

APP

CRMP2

DMPK

GFAP

LIMK1

LIMK2

MAG

MAP2

MAPKAPK1b

MLCP

MRCK

MSK1

NgR1

NgR2

NOGO-A

OMgp

p21CIP1/WAF1

PKN2

ROCKI

ROCKII

TROY

FURTHER INFORMATION

Alzheimer's Disease International

Alzheimer Research Forum

CareCure Community

Christopher and Dana Reeve Paralysis Resource Center

MedicinePlus - Alzheimer's Disease

National Institute of Neurological Disorders and Stroke

National Multiple Sclerosis Society

NeuroTrauma Registry Spinal Cord Map

Glossary

NEUROPATHIC PAIN

A pain state initiated or caused by a primary lesion or dysfunction in the nervous system.

AGC KINASES

A group of kinases with a high degree of amino-acid sequence conservation in their kinase domains.

OMPHALOCOELE

Inability to close the ventral body wall during ontogenetic development, which causes the protrusion of intestinal organs.

HAEMORRHAGE

Excessive bleeding, with increased blood release from blood vessels into surrounding tissues or organs.

SUBARACHNOID SPACE

The brain and spinal cord are enclosed by three layers of connective tissue called the meninges.The space below (or under) the outermost meningeal layer and the middle arachnoid meningeal layer is the subarachnoid space.

ISCHAEMIA

Insufficient blood supply of organs or tissues, very often caused by a thrombus or a clot blocking a blood vessel.

REPERFUSION

Restoration of blood flow after obstruction of blood vessels.

EXCITOTOXICITY

The fundamental concept that neurons can be damaged by overstimulation or overexcitation with excitatory amino acids such as glutamate.

NEURONAL GROWTH CONES

Hand-like structures at the tip of growing neurites, axons and dendrites.

RETRACTION BULB

Large bulbs at the ends of injured nerve fibres in humans suffering from brain injury or spinal-cord injury.

CHONDROITIN SULPHATE PROTEOGLYCAN

(CSPG). The most abundant proteoglycan found in the mammalian CNS, CSPG consists of a protein core and long, unbranched polysaccharides comprising chondroitin sulphate (CS) disaccharide unit repeats.

C3 TRANSFERASE

Bacterial exoenzyme produced by a diverse group of bacteria that inactivates Rho proteins by covalently attaching an ADP-ribose to certain amino acids.

PDAPP MICE

Transgenic mice that express mutated human amyloid precursor protein under control of the platelet-derived growth factor promoter. These mice develop extracellular fibrils and plaques.

LYSOPHOSPHATIDIC ACID (LPA)

A natural phospholipid and a major active constituent of serum that induces growth-factor-like activities in many cell types by binding to specific G-protein-coupled cell-surface receptors.

ALLODYNIA

Abnormal pain state, in which normally non-painful stimuli evoke painful responses.

HYPERALGESIA

An increased or excessive pain response to noxious stimuli.

RHIZOTOMY

Surgical transection of a spinal nerve root.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Mueller, B., Mack, H. & Teusch, N. Rho kinase, a promising drug target for neurological disorders. Nat Rev Drug Discov 4, 387–398 (2005). https://doi.org/10.1038/nrd1719

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrd1719

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing