Skip to main content
Top
Published in: BioDrugs 3/2007

01-05-2007 | Mechanisms and Targets

Rho GTPase/Rho Kinase Inhibition as a Novel Target for the Treatment of Glaucoma

Authors: Dr Vasantha P. Rao, David L. Epstein

Published in: BioDrugs | Issue 3/2007

Login to get access

Abstract

Rho kinase (ROCK1 and ROCK2) is a serine/threonine kinase that serves as an important downstream effector of Rho GTPase, and plays a critical role in regulating the contractile tone of smooth muscle tissues in a calcium-independent manner. Several lines of experimental evidence indicate that modulating ROCK activity within the aqueous humor outflow pathway using selective inhibitors could achieve very significant benefits for the treatment of increased intraocular pressure in patients with glaucoma. The rationale for such an approach stems from experimental data suggesting that both ROCK and Rho GTPase inhibitors can increase aqueous humor drainage through the trabecular meshwork, leading to a decrease in intraocular pressure. In addition to their ocular hypotensive properties, inhibitors of both ROCK and Rho GTPase have been shown to enhance ocular blood flow, retinal ganglion cell survival and axon regeneration. These properties of the ROCK and Rho GTPase inhibitors indicate that targeting the Rho GTPase/ROCK pathway with selective inhibitors represents a novel therapeutic approach aimed at lowering increased intraocular pressure in glaucoma patients.
Literature
1.
go back to reference Thylefors B, Negrel AD, Pararajasegaram R, et al. Global data on blindness. Bull World Health Organ 1995; 73(1): 115–21PubMed Thylefors B, Negrel AD, Pararajasegaram R, et al. Global data on blindness. Bull World Health Organ 1995; 73(1): 115–21PubMed
2.
go back to reference Quigley HA, Broman AT. The number of people with glaucoma worldwide in 2010 and 2020. Br J Ophthalmol 2006; 90(3): 262–7PubMedCrossRef Quigley HA, Broman AT. The number of people with glaucoma worldwide in 2010 and 2020. Br J Ophthalmol 2006; 90(3): 262–7PubMedCrossRef
4.
5.
go back to reference Gabelt BT, Kaufman PL. Changes in aqueous humor dynamics with age and glaucoma. Prog Retin Eye Res 2005; 24(5): 612–37PubMedCrossRef Gabelt BT, Kaufman PL. Changes in aqueous humor dynamics with age and glaucoma. Prog Retin Eye Res 2005; 24(5): 612–37PubMedCrossRef
6.
go back to reference Lutjen-Drecoll E. Functional morphology of the trabecular meshwork in primate eyes. Prog Retin Eye Res 1999; 18(1): 91–119PubMedCrossRef Lutjen-Drecoll E. Functional morphology of the trabecular meshwork in primate eyes. Prog Retin Eye Res 1999; 18(1): 91–119PubMedCrossRef
7.
go back to reference Woodward DF, Gil DW. The inflow and outflow of anti-glaucoma drugs. Trends Pharmacol Sci 2004; 25(5): 238–41PubMedCrossRef Woodward DF, Gil DW. The inflow and outflow of anti-glaucoma drugs. Trends Pharmacol Sci 2004; 25(5): 238–41PubMedCrossRef
8.
9.
go back to reference Brubaker RF. Targeting outflow facility in glaucoma management. Surv Ophthalmol 2003; 48Suppl. 1: S17–20PubMedCrossRef Brubaker RF. Targeting outflow facility in glaucoma management. Surv Ophthalmol 2003; 48Suppl. 1: S17–20PubMedCrossRef
10.
go back to reference Tan JC, Peters DM, Kaufman PL. Recent developments in understanding the pathophysiology of elevated intraocular pressure. Curr Opin Ophthalmol 2006; 17(2): 168–74PubMed Tan JC, Peters DM, Kaufman PL. Recent developments in understanding the pathophysiology of elevated intraocular pressure. Curr Opin Ophthalmol 2006; 17(2): 168–74PubMed
11.
go back to reference Marquis RE, Whitson JT. Management of glaucoma: focus on pharmacological therapy. Drugs Aging 2005; 22(1): 1–21PubMedCrossRef Marquis RE, Whitson JT. Management of glaucoma: focus on pharmacological therapy. Drugs Aging 2005; 22(1): 1–21PubMedCrossRef
12.
go back to reference Tian B, Geiger B, Epstein DL, et al. Cytoskeletal involvement in the regulation of aqueous humor outflow. Invest Ophthalmol Vis Sci 2000; 41(3): 619–23PubMed Tian B, Geiger B, Epstein DL, et al. Cytoskeletal involvement in the regulation of aqueous humor outflow. Invest Ophthalmol Vis Sci 2000; 41(3): 619–23PubMed
13.
go back to reference Gong H, Tripathi RC, Tripathi BJ. Morphology of the aqueous outflow pathway. Microsc Res Tech 1996; 33(4): 336–67PubMedCrossRef Gong H, Tripathi RC, Tripathi BJ. Morphology of the aqueous outflow pathway. Microsc Res Tech 1996; 33(4): 336–67PubMedCrossRef
14.
go back to reference Johnstone MA. The aqueous outflow system as a mechanical pump: evidence from examination of tissue and aqueous movement in human and non-human primates. J Glaucoma 2004; 13(5): 421–38PubMedCrossRef Johnstone MA. The aqueous outflow system as a mechanical pump: evidence from examination of tissue and aqueous movement in human and non-human primates. J Glaucoma 2004; 13(5): 421–38PubMedCrossRef
15.
go back to reference Francis BA, Alvarado J. The cellular basis of aqueous outflow regulation. Curr Opin Ophthalmol 1997; 8(2): 19–27PubMedCrossRef Francis BA, Alvarado J. The cellular basis of aqueous outflow regulation. Curr Opin Ophthalmol 1997; 8(2): 19–27PubMedCrossRef
16.
go back to reference Brandt JD, O’Donnell ME. How does the trabecular meshwork regulate outflow? Clues from the vascular endothelium. J Glaucoma 1999; 8(5): 328–39PubMedCrossRef Brandt JD, O’Donnell ME. How does the trabecular meshwork regulate outflow? Clues from the vascular endothelium. J Glaucoma 1999; 8(5): 328–39PubMedCrossRef
18.
go back to reference Wiederholt M, Thieme H, Stumpff F. The regulation of trabecular meshwork and ciliary muscle contractility. Prog Retin Eye Res 2000; 19(3): 271–95PubMedCrossRef Wiederholt M, Thieme H, Stumpff F. The regulation of trabecular meshwork and ciliary muscle contractility. Prog Retin Eye Res 2000; 19(3): 271–95PubMedCrossRef
19.
go back to reference Lutjen-Drecoll E. Morphological changes in glaucomatous eyes and the role of TGFbeta2 for the pathogenesis of the disease. Exp Eye Res 2005; 81(1): 1–4PubMedCrossRef Lutjen-Drecoll E. Morphological changes in glaucomatous eyes and the role of TGFbeta2 for the pathogenesis of the disease. Exp Eye Res 2005; 81(1): 1–4PubMedCrossRef
20.
go back to reference Tezel G, Kass MA, Kolker AE, et al. Plasma and aqueous humor endothelin levels in primary open-angle glaucoma. J Glaucoma 1997; 6(2): 83–9PubMedCrossRef Tezel G, Kass MA, Kolker AE, et al. Plasma and aqueous humor endothelin levels in primary open-angle glaucoma. J Glaucoma 1997; 6(2): 83–9PubMedCrossRef
21.
go back to reference Yorio T, Krishnamoorthy R, Prasanna G. Endothelin: is it a contributor to glaucoma pathophysiology? J Glaucoma 2002; 11(3): 259–70PubMedCrossRef Yorio T, Krishnamoorthy R, Prasanna G. Endothelin: is it a contributor to glaucoma pathophysiology? J Glaucoma 2002; 11(3): 259–70PubMedCrossRef
22.
go back to reference Tripathi RC, Li J, Chan WF, et al. Aqueous humor in glaucomatous eyes contains an increased level of TGF-beta 2. Exp Eye Res 1994; 59(6): 723–7PubMedCrossRef Tripathi RC, Li J, Chan WF, et al. Aqueous humor in glaucomatous eyes contains an increased level of TGF-beta 2. Exp Eye Res 1994; 59(6): 723–7PubMedCrossRef
23.
go back to reference Wordinger RJ, Clark AF. Effects of glucocorticoids on the trabecular meshwork: towards a better understanding of glaucoma. Prog Retin Eye Res 1999; 18(5): 629–67PubMedCrossRef Wordinger RJ, Clark AF. Effects of glucocorticoids on the trabecular meshwork: towards a better understanding of glaucoma. Prog Retin Eye Res 1999; 18(5): 629–67PubMedCrossRef
24.
go back to reference Epstein DL, Rowlette LL, Roberts BC. Acto-myosin drug effects and aqueous outflow function. Invest Ophthalmol Vis Sci 1999; 40(1): 74–81PubMed Epstein DL, Rowlette LL, Roberts BC. Acto-myosin drug effects and aqueous outflow function. Invest Ophthalmol Vis Sci 1999; 40(1): 74–81PubMed
25.
go back to reference Rao PV, Deng PF, Kumar J, et al. Modulation of aqueous humor outflow facility by the Rho kinase-specific inhibitor Y-27632. Invest Ophthalmol Vis Sci 2001; 42(5): 1029–37PubMed Rao PV, Deng PF, Kumar J, et al. Modulation of aqueous humor outflow facility by the Rho kinase-specific inhibitor Y-27632. Invest Ophthalmol Vis Sci 2001; 42(5): 1029–37PubMed
26.
go back to reference Yue BY. The extracellular matrix and its modulation in the trabecular meshwork. Surv Ophthalmol 1996; 40(5): 379–90PubMedCrossRef Yue BY. The extracellular matrix and its modulation in the trabecular meshwork. Surv Ophthalmol 1996; 40(5): 379–90PubMedCrossRef
27.
28.
go back to reference deKater AW, Shahsafaei A, Epstein DL. Localization of smooth muscle and nonmuscle actin isoforms in the human aqueous outflow pathway. Invest Ophthalmol Vis Sci 1992; 33(2): 424–9PubMed deKater AW, Shahsafaei A, Epstein DL. Localization of smooth muscle and nonmuscle actin isoforms in the human aqueous outflow pathway. Invest Ophthalmol Vis Sci 1992; 33(2): 424–9PubMed
29.
go back to reference Rao PV, Deng P, Sasaki Y, et al. Regulation of myosin light chain phosphorylation in the trabecular meshwork: role in aqueous humour outflow facility. Exp Eye Res 2005; 80(2): 197–206PubMedCrossRef Rao PV, Deng P, Sasaki Y, et al. Regulation of myosin light chain phosphorylation in the trabecular meshwork: role in aqueous humour outflow facility. Exp Eye Res 2005; 80(2): 197–206PubMedCrossRef
30.
go back to reference Nakamura Y, Hirano S, Suzuki K, et al. Signaling mechanism of TGF-betal-induced collagen contraction mediated by bovine trabecular meshwork cells. Invest Ophthalmol Vis Sci 2002; 43(11): 3465–72PubMed Nakamura Y, Hirano S, Suzuki K, et al. Signaling mechanism of TGF-betal-induced collagen contraction mediated by bovine trabecular meshwork cells. Invest Ophthalmol Vis Sci 2002; 43(11): 3465–72PubMed
31.
go back to reference Wiederholt M. Direct involvement of trabecular meshwork in the regulation of aqueous humor outflow. Curr Opin Ophthalmol 1998; 9(2): 46–9PubMedCrossRef Wiederholt M. Direct involvement of trabecular meshwork in the regulation of aqueous humor outflow. Curr Opin Ophthalmol 1998; 9(2): 46–9PubMedCrossRef
32.
go back to reference Thieme H, Nuskovski M, Nass JU, et al. Mediation of calcium-independent contraction in trabecular meshwork through protein kinase C and rho-A. Invest Ophthalmol Vis Sci 2000; 41(13): 4240–6PubMed Thieme H, Nuskovski M, Nass JU, et al. Mediation of calcium-independent contraction in trabecular meshwork through protein kinase C and rho-A. Invest Ophthalmol Vis Sci 2000; 41(13): 4240–6PubMed
33.
go back to reference Mettu PS, Deng PF, Misra UK, et al. Role of lysophospholipid growth factors in the modulation of aqueous humor outflow facility. Invest Ophthalmol Vis Sci 2004; 45(7): 2263–71PubMedCrossRef Mettu PS, Deng PF, Misra UK, et al. Role of lysophospholipid growth factors in the modulation of aqueous humor outflow facility. Invest Ophthalmol Vis Sci 2004; 45(7): 2263–71PubMedCrossRef
34.
go back to reference Somlyo AP, Somlyo AV. Ca2+ sensitivity of smooth muscle and nonmuscle myosin II: modulated by G proteins, kinases, and myosin phosphatase. Physiol Rev 2003; 83(4): 1325–58PubMed Somlyo AP, Somlyo AV. Ca2+ sensitivity of smooth muscle and nonmuscle myosin II: modulated by G proteins, kinases, and myosin phosphatase. Physiol Rev 2003; 83(4): 1325–58PubMed
35.
go back to reference Kamm KE, Stull JT. Dedicated myosin light chain kinases with diverse cellular functions. J Biol Chem 2001; 276(7): 4527–30PubMedCrossRef Kamm KE, Stull JT. Dedicated myosin light chain kinases with diverse cellular functions. J Biol Chem 2001; 276(7): 4527–30PubMedCrossRef
36.
go back to reference Wettschureck N, Offermanns S. Rho/Rho-kinase mediated signaling in physiology and pathophysiology. J Mol Med 2002; 80(10): 629–38PubMedCrossRef Wettschureck N, Offermanns S. Rho/Rho-kinase mediated signaling in physiology and pathophysiology. J Mol Med 2002; 80(10): 629–38PubMedCrossRef
37.
go back to reference Hartshorne DJ. Myosin phosphatase: subunits and interactions. Acta Physiol Scand 1998; 164(4): 483–93PubMedCrossRef Hartshorne DJ. Myosin phosphatase: subunits and interactions. Acta Physiol Scand 1998; 164(4): 483–93PubMedCrossRef
38.
go back to reference Pfitzer G. Invited review: regulation of myosin phosphorylation in smooth muscle. J Appl Physiol 2001; 91(1): 497–503PubMed Pfitzer G. Invited review: regulation of myosin phosphorylation in smooth muscle. J Appl Physiol 2001; 91(1): 497–503PubMed
39.
go back to reference Fukata Y, Amano M, Kaibuchi K. Rho-Rho-kinase pathway in smooth muscle contraction and cytoskeletal reorganization of non-muscle cells. Trends Pharmacol Sci 2001; 22(1): 32–9PubMedCrossRef Fukata Y, Amano M, Kaibuchi K. Rho-Rho-kinase pathway in smooth muscle contraction and cytoskeletal reorganization of non-muscle cells. Trends Pharmacol Sci 2001; 22(1): 32–9PubMedCrossRef
40.
go back to reference Uehata M, Ishizaki T, Satoh H, et al. Calcium sensitization of smooth muscle mediated by a Rho-associated protein kinase in hypertension. Nature 1997; 389(6654): 990–4PubMedCrossRef Uehata M, Ishizaki T, Satoh H, et al. Calcium sensitization of smooth muscle mediated by a Rho-associated protein kinase in hypertension. Nature 1997; 389(6654): 990–4PubMedCrossRef
41.
go back to reference Zhang M, Rao PV. Blebbistatin, a novel inhibitor of myosin II ATPase activity, increases aqueous humor outflow facility in perfused enucleated porcine eyes. Invest Ophthalmol Vis Sci 2005; 46(11): 4130–8PubMedCrossRef Zhang M, Rao PV. Blebbistatin, a novel inhibitor of myosin II ATPase activity, increases aqueous humor outflow facility in perfused enucleated porcine eyes. Invest Ophthalmol Vis Sci 2005; 46(11): 4130–8PubMedCrossRef
42.
go back to reference Honjo M, Inatani M, Kido N, et al. A myosin light chain kinase inhibitor, ML-9, lowers the intraocular pressure in rabbit eyes. Exp Eye Res 2002; 75(2): 135–42PubMedCrossRef Honjo M, Inatani M, Kido N, et al. A myosin light chain kinase inhibitor, ML-9, lowers the intraocular pressure in rabbit eyes. Exp Eye Res 2002; 75(2): 135–42PubMedCrossRef
43.
go back to reference Tian B, Kaufman PL, Volberg T, et al. H-7 disrupts the actin cytoskeleton and increases outflow facility. Arch Ophthalmol 1998; 116(5): 633–43PubMed Tian B, Kaufman PL, Volberg T, et al. H-7 disrupts the actin cytoskeleton and increases outflow facility. Arch Ophthalmol 1998; 116(5): 633–43PubMed
44.
go back to reference Bill A, Lutjen-Drecoll E, Svedbergh B. Effects of intracameral Na2EDTA and EGTA on aqueous outflow routes in the monkey eye. Invest Ophthalmol Vis Sci 1980; 19(5): 492–504PubMed Bill A, Lutjen-Drecoll E, Svedbergh B. Effects of intracameral Na2EDTA and EGTA on aqueous outflow routes in the monkey eye. Invest Ophthalmol Vis Sci 1980; 19(5): 492–504PubMed
45.
go back to reference Ethier CR, Read AT, Chan DW. Effects of latrunculin-B on outflow facility and trabecular meshwork structure in human eyes. Invest Ophthalmol Vis Sci 2006; 47(5): 1991–8PubMedCrossRef Ethier CR, Read AT, Chan DW. Effects of latrunculin-B on outflow facility and trabecular meshwork structure in human eyes. Invest Ophthalmol Vis Sci 2006; 47(5): 1991–8PubMedCrossRef
46.
go back to reference Johnson DH. The effect of cytochalasin D on outflow facility and the trabecular meshwork of the human eye in perfusion organ culture. Invest Ophthalmol Vis Sci 1997; 38(13): 2790–9PubMed Johnson DH. The effect of cytochalasin D on outflow facility and the trabecular meshwork of the human eye in perfusion organ culture. Invest Ophthalmol Vis Sci 1997; 38(13): 2790–9PubMed
47.
go back to reference Kaufman PL, Erickson KA. Cytochalasin B and D dose-outflow facility response relationships in the cynomolgus monkey. Invest Ophthalmol Vis Sci 1982; 23(5): 646–50PubMed Kaufman PL, Erickson KA. Cytochalasin B and D dose-outflow facility response relationships in the cynomolgus monkey. Invest Ophthalmol Vis Sci 1982; 23(5): 646–50PubMed
48.
go back to reference Epstein DL, Freddo TF, Bassett-Chu S, et al. Influence of ethacrynic acid on outflow facility in the monkey and calf eye. Invest Ophthalmol Vis Sci 1987; 28(12): 2067–75PubMed Epstein DL, Freddo TF, Bassett-Chu S, et al. Influence of ethacrynic acid on outflow facility in the monkey and calf eye. Invest Ophthalmol Vis Sci 1987; 28(12): 2067–75PubMed
49.
go back to reference Ethier CR, Coloma FM. Effects of ethacrynic acid on Schlemm’s canal inner wall and outflow facility in human eyes. Invest Ophthalmol Vis Sci 1999; 40(7): 1599–607PubMed Ethier CR, Coloma FM. Effects of ethacrynic acid on Schlemm’s canal inner wall and outflow facility in human eyes. Invest Ophthalmol Vis Sci 1999; 40(7): 1599–607PubMed
50.
52.
53.
go back to reference Van Aelst L, D’Souza-Schorey C. Rho GTPases and signaling networks. Genes Dev 1997; 11(18): 2295–322PubMedCrossRef Van Aelst L, D’Souza-Schorey C. Rho GTPases and signaling networks. Genes Dev 1997; 11(18): 2295–322PubMedCrossRef
54.
go back to reference Cernuda-Morollon E, Ridley AJ. Rho GTPases and leukocyte adhesion receptor expression and function in endothelial cells. Circ Res 2006; 98(6): 757–67PubMedCrossRef Cernuda-Morollon E, Ridley AJ. Rho GTPases and leukocyte adhesion receptor expression and function in endothelial cells. Circ Res 2006; 98(6): 757–67PubMedCrossRef
55.
go back to reference Chitaley K, Weber D, Webb RC. RhoA/Rho-kinase, vascular changes, and hypertension. Curr Hypertens Rep 2001; 3(2): 139–44PubMedCrossRef Chitaley K, Weber D, Webb RC. RhoA/Rho-kinase, vascular changes, and hypertension. Curr Hypertens Rep 2001; 3(2): 139–44PubMedCrossRef
56.
go back to reference Habas R, Kato Y, He X. Wnt/Frizzled activation of Rho regulates vertebrate gastrulation and requires a novel Formin homology protein Daaml. Cell 2001; 107(7): 843–54PubMedCrossRef Habas R, Kato Y, He X. Wnt/Frizzled activation of Rho regulates vertebrate gastrulation and requires a novel Formin homology protein Daaml. Cell 2001; 107(7): 843–54PubMedCrossRef
57.
go back to reference Wettschureck N, Offermanns S. Mammalian G proteins and their cell type specific functions. Physiol Rev 2005; 85(4): 1159–204PubMedCrossRef Wettschureck N, Offermanns S. Mammalian G proteins and their cell type specific functions. Physiol Rev 2005; 85(4): 1159–204PubMedCrossRef
58.
go back to reference Sah VP, Seasholtz TM, Sagi SA, et al. The role of Rho in G protein-coupled receptor signal transduction. Annu Rev Pharmacol Toxicol 2000; 40: 459–89PubMedCrossRef Sah VP, Seasholtz TM, Sagi SA, et al. The role of Rho in G protein-coupled receptor signal transduction. Annu Rev Pharmacol Toxicol 2000; 40: 459–89PubMedCrossRef
59.
go back to reference Riobo NA, Manning DR. Receptors coupled to heterotrimeric G proteins of the G12 family. Trends Pharmacol Sci 2005; 26(3): 146–54PubMedCrossRef Riobo NA, Manning DR. Receptors coupled to heterotrimeric G proteins of the G12 family. Trends Pharmacol Sci 2005; 26(3): 146–54PubMedCrossRef
60.
go back to reference Coso OA, Teramoto H, Simonds WF, et al. Signaling from G protein-coupled receptors to c-Jun kinase involves beta gamma subunits of heterotrimeric G proteins acting on a Ras and Rac1-dependent pathway. J Biol Chem 1996; 271(8): 3963–6PubMedCrossRef Coso OA, Teramoto H, Simonds WF, et al. Signaling from G protein-coupled receptors to c-Jun kinase involves beta gamma subunits of heterotrimeric G proteins acting on a Ras and Rac1-dependent pathway. J Biol Chem 1996; 271(8): 3963–6PubMedCrossRef
61.
go back to reference Marinissen MJ, Gutkind JS. G-protein-coupled receptors and signaling networks: emerging paradigms. Trends Pharmacol Sci 2001; 22(7): 368–76PubMedCrossRef Marinissen MJ, Gutkind JS. G-protein-coupled receptors and signaling networks: emerging paradigms. Trends Pharmacol Sci 2001; 22(7): 368–76PubMedCrossRef
62.
go back to reference Dutt P, Nguyen N, Toksoz D. Role of Lbc RhoGEF in Galphal2/13-induced signals to Rho GTPase. Cell Signal 2004; 16(2): 201–9PubMedCrossRef Dutt P, Nguyen N, Toksoz D. Role of Lbc RhoGEF in Galphal2/13-induced signals to Rho GTPase. Cell Signal 2004; 16(2): 201–9PubMedCrossRef
63.
go back to reference Tanabe S, Kreutz B, Suzuki N, et al. Regulation of RGS-RhoGEFs by Galphal2 and Galphal3 proteins. Methods Enzymol 2004; 390: 285–94PubMedCrossRef Tanabe S, Kreutz B, Suzuki N, et al. Regulation of RGS-RhoGEFs by Galphal2 and Galphal3 proteins. Methods Enzymol 2004; 390: 285–94PubMedCrossRef
64.
go back to reference Cox AD, Der CJ. Protein prenylation: more than just glue? Curr Opin Cell Biol 1992; 4(6): 1008–16PubMedCrossRef Cox AD, Der CJ. Protein prenylation: more than just glue? Curr Opin Cell Biol 1992; 4(6): 1008–16PubMedCrossRef
66.
67.
go back to reference Budzyn K, Marley PD, Sobey CG. Targeting Rho and Rho-kinase in the treatment of cardiovascular disease. Trends Pharmacol Sci 2006; 27(2): 97–104PubMedCrossRef Budzyn K, Marley PD, Sobey CG. Targeting Rho and Rho-kinase in the treatment of cardiovascular disease. Trends Pharmacol Sci 2006; 27(2): 97–104PubMedCrossRef
68.
go back to reference Fritz G, Kaina B. Rho GTPases: promising cellular targets for novel anticancer drugs. Curr Cancer Drug Targets 2006; 6(1): 1–14PubMed Fritz G, Kaina B. Rho GTPases: promising cellular targets for novel anticancer drugs. Curr Cancer Drug Targets 2006; 6(1): 1–14PubMed
69.
go back to reference Nakagami H, Jensen KS, Liao JK. A novel pleiotropic effect of statins: prevention of cardiac hypertrophy by cholesterol-independent mechanisms. Ann Med 2003; 35(6): 398–403PubMedCrossRef Nakagami H, Jensen KS, Liao JK. A novel pleiotropic effect of statins: prevention of cardiac hypertrophy by cholesterol-independent mechanisms. Ann Med 2003; 35(6): 398–403PubMedCrossRef
70.
go back to reference Kumar J, Rao PV, Rowlette LL, et al. Rho GTPase-mediated actin cytoskeletal reorganization in outflow pathway cells may modulate outflow function. Invest Ophthalmol Vis Sci 1999; 40: 3534 Kumar J, Rao PV, Rowlette LL, et al. Rho GTPase-mediated actin cytoskeletal reorganization in outflow pathway cells may modulate outflow function. Invest Ophthalmol Vis Sci 1999; 40: 3534
71.
go back to reference Liu X, Hu Y, Filla MS, et al. The effect of C3 transgene expression on actin and cellular adhesions in cultured human trabecular meshwork cells and on outflow facility in organ cultured monkey eyes. Mol Vis 2005; 11: 1112–21PubMed Liu X, Hu Y, Filla MS, et al. The effect of C3 transgene expression on actin and cellular adhesions in cultured human trabecular meshwork cells and on outflow facility in organ cultured monkey eyes. Mol Vis 2005; 11: 1112–21PubMed
72.
go back to reference Murthy KS. Signaling for contraction and relaxation in smooth muscle of the gut. Annu Rev Physiol 2006; 68: 345–74PubMedCrossRef Murthy KS. Signaling for contraction and relaxation in smooth muscle of the gut. Annu Rev Physiol 2006; 68: 345–74PubMedCrossRef
73.
go back to reference Lang P, Gesbert F, Delespine-Carmagnat M, et al. Protein kinase A phosphorylation of RhoA mediates the morphological and functional effects of cyclic AMP in cytotoxic lymphocytes. Embo J 1996; 15(3): 510–9PubMed Lang P, Gesbert F, Delespine-Carmagnat M, et al. Protein kinase A phosphorylation of RhoA mediates the morphological and functional effects of cyclic AMP in cytotoxic lymphocytes. Embo J 1996; 15(3): 510–9PubMed
74.
go back to reference Murthy KS, Zhou H, Grider JR, et al. Inhibition of sustained smooth muscle contraction by PKA and PKG preferentially mediated by phosphorylation of RhoA. Am J Physiol Gastrointest Liver Physiol 2003; 284(6): G1006–16PubMed Murthy KS, Zhou H, Grider JR, et al. Inhibition of sustained smooth muscle contraction by PKA and PKG preferentially mediated by phosphorylation of RhoA. Am J Physiol Gastrointest Liver Physiol 2003; 284(6): G1006–16PubMed
75.
go back to reference Qiao J, Huang F, Lum H. PKA inhibits RhoA activation: a protection mechanism against endothelial barrier dysfunction. Am J Physiol Lung Cell Mol Physiol 2003; 284(6): L972–80PubMed Qiao J, Huang F, Lum H. PKA inhibits RhoA activation: a protection mechanism against endothelial barrier dysfunction. Am J Physiol Lung Cell Mol Physiol 2003; 284(6): L972–80PubMed
76.
go back to reference Williams J, Bogwu J, Oyekan A. The role of the RhoA/Rho-kinase signaling pathway in renal vascular reactivity in endothelial nitric oxide synthase null mice. J Hypertens 2006; 24(7): 1429–36PubMedCrossRef Williams J, Bogwu J, Oyekan A. The role of the RhoA/Rho-kinase signaling pathway in renal vascular reactivity in endothelial nitric oxide synthase null mice. J Hypertens 2006; 24(7): 1429–36PubMedCrossRef
77.
go back to reference Musicki B, Burnett AL. eNOS function and dysfunction in the penis. Exp Biol Med (Maywood) 2006; 231(2): 154–65 Musicki B, Burnett AL. eNOS function and dysfunction in the penis. Exp Biol Med (Maywood) 2006; 231(2): 154–65
78.
go back to reference Shiga N, Hirano K, Hirano M, et al. Long-term inhibition of RhoA attenuates vascular contractility by enhancing endothelial NO production in an intact rabbit mesenteric artery. Circ Res 2005; 96(9): 1014–21PubMedCrossRef Shiga N, Hirano K, Hirano M, et al. Long-term inhibition of RhoA attenuates vascular contractility by enhancing endothelial NO production in an intact rabbit mesenteric artery. Circ Res 2005; 96(9): 1014–21PubMedCrossRef
79.
go back to reference Witteck A, Yao Y, Fechir M, et al. Rho protein-mediated changes in the structure of the actin cytoskeleton regulate human inducible NO synthase gene expression. Exp Cell Res 2003; 287(1): 106–15PubMedCrossRef Witteck A, Yao Y, Fechir M, et al. Rho protein-mediated changes in the structure of the actin cytoskeleton regulate human inducible NO synthase gene expression. Exp Cell Res 2003; 287(1): 106–15PubMedCrossRef
80.
go back to reference Ming XF, Viswambharan H, Barandier C, et al. Rho GTPase/Rho kinase negatively regulates endothelial nitric oxide synthase phosphorylation through the inhibition of protein kinase B/Akt in human endothelial cells. Mol Cell Biol 2002; 22(24): 8467–77PubMedCrossRef Ming XF, Viswambharan H, Barandier C, et al. Rho GTPase/Rho kinase negatively regulates endothelial nitric oxide synthase phosphorylation through the inhibition of protein kinase B/Akt in human endothelial cells. Mol Cell Biol 2002; 22(24): 8467–77PubMedCrossRef
81.
go back to reference Kraynack NC, Corey DA, Elmer HL, et al. Mechanisms of NOS2 regulation by Rho GTPase signaling in airway epithelial cells. Am J Physiol Lung Cell Mol Physiol 2002; 283(3): L604–11PubMed Kraynack NC, Corey DA, Elmer HL, et al. Mechanisms of NOS2 regulation by Rho GTPase signaling in airway epithelial cells. Am J Physiol Lung Cell Mol Physiol 2002; 283(3): L604–11PubMed
82.
go back to reference Bivalacqua TJ, Champion HC, Usta MF, et al. RhoA/Rho-kinase suppresses endothelial nitric oxide synthase in the penis: a mechanism for diabetes-associated erectile dysfunction. Proc Natl Acad Sci U S A 2004; 101(24): 9121–6PubMedCrossRef Bivalacqua TJ, Champion HC, Usta MF, et al. RhoA/Rho-kinase suppresses endothelial nitric oxide synthase in the penis: a mechanism for diabetes-associated erectile dysfunction. Proc Natl Acad Sci U S A 2004; 101(24): 9121–6PubMedCrossRef
83.
go back to reference Neufeld AH, Dueker DK, Vegge T, et al. Adenosine 3′,5′-monophosphate increases the outflow of aqueous humor from the rabbit eye. Invest Ophthalmol 1975; 14(1): 40–2PubMed Neufeld AH, Dueker DK, Vegge T, et al. Adenosine 3′,5′-monophosphate increases the outflow of aqueous humor from the rabbit eye. Invest Ophthalmol 1975; 14(1): 40–2PubMed
84.
go back to reference Neufeld AH. Influences of cyclic nucleotides on outflow facility in the vervet monkey. Exp Eye Res 1978; 27(4): 387–97PubMedCrossRef Neufeld AH. Influences of cyclic nucleotides on outflow facility in the vervet monkey. Exp Eye Res 1978; 27(4): 387–97PubMedCrossRef
85.
go back to reference Kee C, Kaufman PL, Gabelt BT. Effect of 8-Br cGMP on aqueous humor dynamics in monkeys. Invest Ophthalmol Vis Sci 1994; 35(6): 2769–73PubMed Kee C, Kaufman PL, Gabelt BT. Effect of 8-Br cGMP on aqueous humor dynamics in monkeys. Invest Ophthalmol Vis Sci 1994; 35(6): 2769–73PubMed
86.
go back to reference Kaufman PL. Adenosine 3′,5′-cyclic-monophosphate and outflow facility in monkey eyes with intact and retrodisplaced ciliary muscle. Exp Eye Res 1987; 44(3): 415–23PubMedCrossRef Kaufman PL. Adenosine 3′,5′-cyclic-monophosphate and outflow facility in monkey eyes with intact and retrodisplaced ciliary muscle. Exp Eye Res 1987; 44(3): 415–23PubMedCrossRef
87.
go back to reference Erickson-Lamy KA, Nathanson JA. Epinephrine increases facility of outflow and cyclic AMP content in the human eye in vitro. Invest Ophthalmol Vis Sci 1992; 33(9): 2672–8PubMed Erickson-Lamy KA, Nathanson JA. Epinephrine increases facility of outflow and cyclic AMP content in the human eye in vitro. Invest Ophthalmol Vis Sci 1992; 33(9): 2672–8PubMed
88.
go back to reference Bartels SP, Lee SR, Neufeld AH. Forskolin stimulates cyclic AMP synthesis, lowers intraocular pressure and increases outflow facility in rabbits. Curr Eye Res 1982; 2(10): 673–81PubMedCrossRef Bartels SP, Lee SR, Neufeld AH. Forskolin stimulates cyclic AMP synthesis, lowers intraocular pressure and increases outflow facility in rabbits. Curr Eye Res 1982; 2(10): 673–81PubMedCrossRef
89.
go back to reference Kotikoski H, Vapaatalo H, Oksala O. Nitric oxide and cyclic GMP enhance aqueous humor outflow facility in rabbits. Curr Eye Res 2003; 26(2): 119–23PubMedCrossRef Kotikoski H, Vapaatalo H, Oksala O. Nitric oxide and cyclic GMP enhance aqueous humor outflow facility in rabbits. Curr Eye Res 2003; 26(2): 119–23PubMedCrossRef
90.
go back to reference Amano M, Chihara K, Kimura K, et al. Formation of actin stress fibers and focal adhesions enhanced by Rho-kinase. Science 1997; 275(5304): 1308–11PubMedCrossRef Amano M, Chihara K, Kimura K, et al. Formation of actin stress fibers and focal adhesions enhanced by Rho-kinase. Science 1997; 275(5304): 1308–11PubMedCrossRef
91.
go back to reference Fujisawa K, Fujita A, Ishizaki T, et al. Identification of the Rho-binding domain of p160ROCK, a Rho-associated coiled-coil containing protein kinase. J Biol Chem 1996; 271(38): 23022–8PubMedCrossRef Fujisawa K, Fujita A, Ishizaki T, et al. Identification of the Rho-binding domain of p160ROCK, a Rho-associated coiled-coil containing protein kinase. J Biol Chem 1996; 271(38): 23022–8PubMedCrossRef
92.
go back to reference Kawano Y, Fukata Y, Oshiro N, et al. Phosphorylation of myosin-binding subunit (MBS) of myosin phosphatase by Rho-kinase in vivo. J Cell Biol 1999; 147(5): 1023–38PubMedCrossRef Kawano Y, Fukata Y, Oshiro N, et al. Phosphorylation of myosin-binding subunit (MBS) of myosin phosphatase by Rho-kinase in vivo. J Cell Biol 1999; 147(5): 1023–38PubMedCrossRef
93.
go back to reference Nakagawa O, Fujisawa K, Ishizaki T, et al. ROCK-I and ROCK-II, two isoforms of Rho-associated coiled-coil forming protein serine/threonine kinase in mice. FEBS Lett 1996; 392(2): 189–93PubMedCrossRef Nakagawa O, Fujisawa K, Ishizaki T, et al. ROCK-I and ROCK-II, two isoforms of Rho-associated coiled-coil forming protein serine/threonine kinase in mice. FEBS Lett 1996; 392(2): 189–93PubMedCrossRef
94.
go back to reference Matsui T, Amano M, Yamamoto T, et al. Rho-associated kinase, a novel serine/threonine kinase, as a putative target for small GTP binding protein Rho. Embo J 1996; 15(9): 2208–16PubMed Matsui T, Amano M, Yamamoto T, et al. Rho-associated kinase, a novel serine/threonine kinase, as a putative target for small GTP binding protein Rho. Embo J 1996; 15(9): 2208–16PubMed
95.
go back to reference Nakajima E, Nakajima T, Minagawa Y, et al. Contribution of ROCK in contraction of trabecular meshwork: proposed mechanism for regulating aqueous outflow in monkey and human eyes. J Pharm Sci 2005; 94(4): 701–8PubMedCrossRef Nakajima E, Nakajima T, Minagawa Y, et al. Contribution of ROCK in contraction of trabecular meshwork: proposed mechanism for regulating aqueous outflow in monkey and human eyes. J Pharm Sci 2005; 94(4): 701–8PubMedCrossRef
96.
go back to reference 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 2001; 276(1): 670–6PubMedCrossRef 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 2001; 276(1): 670–6PubMedCrossRef
97.
go back to reference Maekawa M, Ishizaki T, Boku S, et al. Signaling from Rho to the actin cytoskeleton through protein kinases ROCK and LIM-kinase. Science 1999; 285(5429): 895–8PubMedCrossRef Maekawa M, Ishizaki T, Boku S, et al. Signaling from Rho to the actin cytoskeleton through protein kinases ROCK and LIM-kinase. Science 1999; 285(5429): 895–8PubMedCrossRef
98.
go back to reference Koyama M, Ito M, Feng J, et al. Phosphorylation of CPI-17, an inhibitory phosphoprotein of smooth muscle myosin phosphatase, by Rho-kinase. FEBS Lett 2000; 475(3): 197–200PubMedCrossRef Koyama M, Ito M, Feng J, et al. Phosphorylation of CPI-17, an inhibitory phosphoprotein of smooth muscle myosin phosphatase, by Rho-kinase. FEBS Lett 2000; 475(3): 197–200PubMedCrossRef
99.
go back to reference Li L, Eto M, Lee MR, et al. Possible involvement of the novel CPI-17 protein in protein kinase C signal transduction of rabbit arterial smooth muscle. J Physiol 1998; 508 (Pt 3): 871–81PubMedCrossRef Li L, Eto M, Lee MR, et al. Possible involvement of the novel CPI-17 protein in protein kinase C signal transduction of rabbit arterial smooth muscle. J Physiol 1998; 508 (Pt 3): 871–81PubMedCrossRef
100.
go back to reference Kitazawa T, Eto M, Woodsome TP, et al. Phosphorylation of the myosin phosphatase targeting subunit and CPI-17 during Ca2+ sensitization in rabbit smooth muscle. J Physiol 2003; 546 (Pt 3): 879–89PubMedCrossRef Kitazawa T, Eto M, Woodsome TP, et al. Phosphorylation of the myosin phosphatase targeting subunit and CPI-17 during Ca2+ sensitization in rabbit smooth muscle. J Physiol 2003; 546 (Pt 3): 879–89PubMedCrossRef
101.
go back to reference Woodsome TP, Eto M, Everett A, et al. Expression of CPI-17 and myosin phosphatase correlates with Ca(2+) sensitivity of protein kinase C-induced contraction in rabbit smooth muscle. J Physiol 2001; 535 (Pt 2): 553–64PubMedCrossRef Woodsome TP, Eto M, Everett A, et al. Expression of CPI-17 and myosin phosphatase correlates with Ca(2+) sensitivity of protein kinase C-induced contraction in rabbit smooth muscle. J Physiol 2001; 535 (Pt 2): 553–64PubMedCrossRef
102.
go back to reference Andersson KE. Erectile physiological and pathophysiological pathways involved in erectile dysfunction. J Urol 2003; 170 (2 Pt 2): S6–13; discussion S-4PubMedCrossRef Andersson KE. Erectile physiological and pathophysiological pathways involved in erectile dysfunction. J Urol 2003; 170 (2 Pt 2): S6–13; discussion S-4PubMedCrossRef
103.
go back to reference Chrissobolis S, Sobey CG. Recent evidence for an involvement of rho-kinase in cerebral vascular disease. Stroke 2006; 37(8): 2174–80PubMedCrossRef Chrissobolis S, Sobey CG. Recent evidence for an involvement of rho-kinase in cerebral vascular disease. Stroke 2006; 37(8): 2174–80PubMedCrossRef
104.
go back to reference Hirooka Y, Shimokawa H. Therapeutic potential of rho-kinase inhibitors in cardiovascular diseases. Am J Cardiovasc Drugs 2005; 5(1): 31–9PubMedCrossRef Hirooka Y, Shimokawa H. Therapeutic potential of rho-kinase inhibitors in cardiovascular diseases. Am J Cardiovasc Drugs 2005; 5(1): 31–9PubMedCrossRef
105.
go back to reference Jin L, Burnett AL. RhoA/Rho-kinase in erectile tissue: mechanisms of disease and therapeutic insights. Clin Sci (Lond) 2006; 110(2): 153–65CrossRef Jin L, Burnett AL. RhoA/Rho-kinase in erectile tissue: mechanisms of disease and therapeutic insights. Clin Sci (Lond) 2006; 110(2): 153–65CrossRef
106.
go back to reference Moriyama T, Nagatoya K. The Rho-ROCK system as a new therapeutic target for preventing interstitial fibrosis. Drug News Perspect 2004; 17(1): 29–34PubMedCrossRef Moriyama T, Nagatoya K. The Rho-ROCK system as a new therapeutic target for preventing interstitial fibrosis. Drug News Perspect 2004; 17(1): 29–34PubMedCrossRef
107.
go back to reference Mueller BK, Mack H, Teusch N. Rho kinase, a promising drug target for neurological disorders. Nat Rev Drug Discov 2005; 4(5): 387–98PubMedCrossRef Mueller BK, Mack H, Teusch N. Rho kinase, a promising drug target for neurological disorders. Nat Rev Drug Discov 2005; 4(5): 387–98PubMedCrossRef
108.
go back to reference Mukai Y, Shimokawa H, Matoba T, et al. Involvement of Rho-kinase in hypertensive vascular disease: a novel therapeutic target in hypertension. FASEB J 2001; 15(6): 1062–4PubMed Mukai Y, Shimokawa H, Matoba T, et al. Involvement of Rho-kinase in hypertensive vascular disease: a novel therapeutic target in hypertension. FASEB J 2001; 15(6): 1062–4PubMed
109.
go back to reference Noma K, Oyama N, Liao JK. Physiological role of ROCKs in the cardiovascular system. Am J Physiol Cell Physiol 2006; 290(3): C661–8PubMedCrossRef Noma K, Oyama N, Liao JK. Physiological role of ROCKs in the cardiovascular system. Am J Physiol Cell Physiol 2006; 290(3): C661–8PubMedCrossRef
110.
go back to reference Rikitake Y, Liao JK. ROCKs as therapeutic targets in cardiovascular diseases. Expert Rev Cardiovasc Ther 2005; 3(3): 441–51PubMedCrossRef Rikitake Y, Liao JK. ROCKs as therapeutic targets in cardiovascular diseases. Expert Rev Cardiovasc Ther 2005; 3(3): 441–51PubMedCrossRef
111.
go back to reference Shimokawa H, Takeshita A. Rho-kinase is an important therapeutic target in cardiovascular medicine. Arterioscler Thromb Vasc Biol 2005; 25(9): 1767–75PubMedCrossRef Shimokawa H, Takeshita A. Rho-kinase is an important therapeutic target in cardiovascular medicine. Arterioscler Thromb Vasc Biol 2005; 25(9): 1767–75PubMedCrossRef
112.
go back to reference Wakino S, Kanda T, Hayashi K. Rho/Rho kinase as a potential target for the treatment of renal disease. Drug News Perspect 2005; 18(10): 639–43PubMedCrossRef Wakino S, Kanda T, Hayashi K. Rho/Rho kinase as a potential target for the treatment of renal disease. Drug News Perspect 2005; 18(10): 639–43PubMedCrossRef
113.
go back to reference Hisaoka T, Yano M, Ohkusa T, et al. Enhancement of Rho/Rho-kinase system in regulation of vascular smooth muscle contraction in tachycardia-induced heart failure. Cardiovasc Res 2001; 49(2): 319–29PubMedCrossRef Hisaoka T, Yano M, Ohkusa T, et al. Enhancement of Rho/Rho-kinase system in regulation of vascular smooth muscle contraction in tachycardia-induced heart failure. Cardiovasc Res 2001; 49(2): 319–29PubMedCrossRef
114.
go back to reference Chang S, Hypolite JA, Zderic SA, et al. Increased corpus cavernosum smooth muscle tone associated with partial bladder outlet obstruction is mediated via Rho-kinase. Am J Physiol Regul Integr Comp Physiol 2005; 289(4): R1124–30PubMedCrossRef Chang S, Hypolite JA, Zderic SA, et al. Increased corpus cavernosum smooth muscle tone associated with partial bladder outlet obstruction is mediated via Rho-kinase. Am J Physiol Regul Integr Comp Physiol 2005; 289(4): R1124–30PubMedCrossRef
115.
go back to reference Seko T, Ito M, Kureishi Y, et al. Activation of RhoA and inhibition of myosin phosphatase as important components in hypertension in vascular smooth muscle. Circ Res 2003; 92(4): 411–8PubMedCrossRef Seko T, Ito M, Kureishi Y, et al. Activation of RhoA and inhibition of myosin phosphatase as important components in hypertension in vascular smooth muscle. Circ Res 2003; 92(4): 411–8PubMedCrossRef
116.
go back to reference Oi K, Shimokawa H, Hiroki J, et al. Remnant lipoproteins from patients with sudden cardiac death enhance coronary vasospastic activity through upregulation of Rho-kinase. Arterioscler Thromb Vasc Biol 2004; 24(5): 918–22PubMedCrossRef Oi K, Shimokawa H, Hiroki J, et al. Remnant lipoproteins from patients with sudden cardiac death enhance coronary vasospastic activity through upregulation of Rho-kinase. Arterioscler Thromb Vasc Biol 2004; 24(5): 918–22PubMedCrossRef
117.
go back to reference Jin L, Liu T, Lagoda GA, et al. Elevated RhoA/Rho-kinase activity in the aged rat penis: mechanism for age-associated erectile dysfunction. FASEB J 2006; 20(3): 536–8PubMed Jin L, Liu T, Lagoda GA, et al. Elevated RhoA/Rho-kinase activity in the aged rat penis: mechanism for age-associated erectile dysfunction. FASEB J 2006; 20(3): 536–8PubMed
118.
go back to reference Rao PV, Deng P, Maddala R, et al. Expression of dominant negative Rho-binding domain of Rho-kinase in organ cultured human eye anterior segments increases aqueous humor outflow. Mol Vis 2005; 11: 288–97PubMed Rao PV, Deng P, Maddala R, et al. Expression of dominant negative Rho-binding domain of Rho-kinase in organ cultured human eye anterior segments increases aqueous humor outflow. Mol Vis 2005; 11: 288–97PubMed
119.
go back to reference Honjo M, Inatani M, Kido N, et al. Effects of protein kinase inhibitor, HA1077, on intraocular pressure and outflow facility in rabbit eyes. Arch Ophthalmol 2001; 119(8): 1171–8PubMed Honjo M, Inatani M, Kido N, et al. Effects of protein kinase inhibitor, HA1077, on intraocular pressure and outflow facility in rabbit eyes. Arch Ophthalmol 2001; 119(8): 1171–8PubMed
120.
go back to reference Aktories K, Mohr C, Koch G. Clostridium botulinum C3 ADP-ribosyltransferase. Curr Top Microbiol Immunol 1992; 175: 115–31PubMedCrossRef Aktories K, Mohr C, Koch G. Clostridium botulinum C3 ADP-ribosyltransferase. Curr Top Microbiol Immunol 1992; 175: 115–31PubMedCrossRef
121.
go back to reference Narumiya S, Morii N. Rho gene products, botulinum C3 exoenzyme and cell adhesion. Cell Signal 1993; 5(1): 9–19PubMedCrossRef Narumiya S, Morii N. Rho gene products, botulinum C3 exoenzyme and cell adhesion. Cell Signal 1993; 5(1): 9–19PubMedCrossRef
122.
123.
go back to reference Vittitow JL, Garg R, Rowlette LL, et al. Gene transfer of dominant-negative RhoA increases outflow facility in perfused human anterior segment cultures. Mol Vis 2002; 8: 32–44PubMed Vittitow JL, Garg R, Rowlette LL, et al. Gene transfer of dominant-negative RhoA increases outflow facility in perfused human anterior segment cultures. Mol Vis 2002; 8: 32–44PubMed
124.
go back to reference Song J, Deng PF, Stinnett SS, et al. Effects of cholesterol-lowering statins on the aqueous humor outflow pathway. Invest Ophthalmol Vis Sci 2005; 46(7): 2424–32PubMedCrossRef Song J, Deng PF, Stinnett SS, et al. Effects of cholesterol-lowering statins on the aqueous humor outflow pathway. Invest Ophthalmol Vis Sci 2005; 46(7): 2424–32PubMedCrossRef
125.
go back to reference Nagaoka T, Takahashi A, Sato E, et al. Effect of systemic administration of simvastatin on retinal circulation. Arch Ophthalmol 2006; 124(5): 665–70PubMedCrossRef Nagaoka T, Takahashi A, Sato E, et al. Effect of systemic administration of simvastatin on retinal circulation. Arch Ophthalmol 2006; 124(5): 665–70PubMedCrossRef
126.
go back to reference McGwin G Jr, McNeal S, Owsley C, et al. Statins and other cholesterol-lowering medications and the presence of glaucoma. Arch Ophthalmol 2004; 122(6): 822–6PubMedCrossRef McGwin G Jr, McNeal S, Owsley C, et al. Statins and other cholesterol-lowering medications and the presence of glaucoma. Arch Ophthalmol 2004; 122(6): 822–6PubMedCrossRef
127.
go back to reference Honjo M, Tanihara H, Inatani M, et al. Effects of rho-associated protein kinase inhibitor Y-27632 on intraocular pressure and outflow facility. Invest Ophthalmol Vis Sci 2001; 42(1): 137–44PubMed Honjo M, Tanihara H, Inatani M, et al. Effects of rho-associated protein kinase inhibitor Y-27632 on intraocular pressure and outflow facility. Invest Ophthalmol Vis Sci 2001; 42(1): 137–44PubMed
128.
go back to reference Fukiage C, Mizutani K, Kawamoto Y, et al. Involvement of phosphorylation of myosin phosphatase by ROCK in trabecular meshwork and ciliary muscle contraction. Biochem Biophys Res Commun 2001; 288(2): 296–300PubMedCrossRef Fukiage C, Mizutani K, Kawamoto Y, et al. Involvement of phosphorylation of myosin phosphatase by ROCK in trabecular meshwork and ciliary muscle contraction. Biochem Biophys Res Commun 2001; 288(2): 296–300PubMedCrossRef
129.
go back to reference Rosenthal R, Choritz L, Schlott S, et al. Effects of ML-7 and Y-27632 on carbachol- and endothelin-1-induced contraction of bovine trabecular meshwork. Exp Eye Res 2005; 80(6): 837–45PubMedCrossRef Rosenthal R, Choritz L, Schlott S, et al. Effects of ML-7 and Y-27632 on carbachol- and endothelin-1-induced contraction of bovine trabecular meshwork. Exp Eye Res 2005; 80(6): 837–45PubMedCrossRef
130.
go back to reference Cellini M, Versura P, Trere D, et al. Effects of endothelin-1 on human trabecular meshwork cell contraction: an in vitro cell culture model. Ophthalmic Res 2005; 37(1): 43–9PubMedCrossRef Cellini M, Versura P, Trere D, et al. Effects of endothelin-1 on human trabecular meshwork cell contraction: an in vitro cell culture model. Ophthalmic Res 2005; 37(1): 43–9PubMedCrossRef
131.
go back to reference Waki M, Yoshida Y, Oka T, et al. Reduction of intraocular pressure by topical administration of an inhibitor of the Rho-associated protein kinase. Curr Eye Res 2001; 22(6): 470–4PubMedCrossRef Waki M, Yoshida Y, Oka T, et al. Reduction of intraocular pressure by topical administration of an inhibitor of the Rho-associated protein kinase. Curr Eye Res 2001; 22(6): 470–4PubMedCrossRef
132.
go back to reference Khurana RN, Deng PF, Epstein DL, et al. The role of protein kinase C in modulation of aqueous humor outflow facility. Exp Eye Res 2003; 76(1): 39–47PubMedCrossRef Khurana RN, Deng PF, Epstein DL, et al. The role of protein kinase C in modulation of aqueous humor outflow facility. Exp Eye Res 2003; 76(1): 39–47PubMedCrossRef
133.
go back to reference Koga T, Koga T, Awai M, et al. Rho-associated protein kinase inhibitor, Y-27632, induces alterations in adhesion, contraction and motility in cultured human trabecular meshwork cells. Exp Eye Res 2006; 82(3): 362–70PubMedCrossRef Koga T, Koga T, Awai M, et al. Rho-associated protein kinase inhibitor, Y-27632, induces alterations in adhesion, contraction and motility in cultured human trabecular meshwork cells. Exp Eye Res 2006; 82(3): 362–70PubMedCrossRef
134.
go back to reference Tian B, Kaufman PL. Effects of the Rho kinase inhibitor Y-27632 and the phosphatase inhibitor calyculin A on outflow facility in monkeys. Exp Eye Res 2005; 80(2): 215–25PubMedCrossRef Tian B, Kaufman PL. Effects of the Rho kinase inhibitor Y-27632 and the phosphatase inhibitor calyculin A on outflow facility in monkeys. Exp Eye Res 2005; 80(2): 215–25PubMedCrossRef
135.
go back to reference Tamura M, Nakao H, Yoshizaki H, et al. Development of specific Rho-kinase inhibitors and their clinical application. Biochim Biophys Acta 2005; 1754(1–2): 245–52PubMed Tamura M, Nakao H, Yoshizaki H, et al. Development of specific Rho-kinase inhibitors and their clinical application. Biochim Biophys Acta 2005; 1754(1–2): 245–52PubMed
136.
go back to reference Sasaki 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 2002; 93(2-3): 225–32PubMedCrossRef Sasaki 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 2002; 93(2-3): 225–32PubMedCrossRef
137.
go back to reference Ishizaki T, Uehata M, Tamechika I, et al. Pharmacological properties of Y-27632, a specific inhibitor of rho-associated kinases. Mol Pharmacol 2000; 57(5): 976–83PubMed Ishizaki T, Uehata M, Tamechika I, et al. Pharmacological properties of Y-27632, a specific inhibitor of rho-associated kinases. Mol Pharmacol 2000; 57(5): 976–83PubMed
138.
go back to reference Delaney Y, Walshe TE, O’Brien C. Vasospasm in glaucoma: clinical and laboratory aspects. Optom Vis Sci 2006; 83(7): 406–14PubMedCrossRef Delaney Y, Walshe TE, O’Brien C. Vasospasm in glaucoma: clinical and laboratory aspects. Optom Vis Sci 2006; 83(7): 406–14PubMedCrossRef
139.
140.
go back to reference Stefansson E, Pedersen DB, Jensen PK, et al. Optic nerve oxygenation. Prog Retin Eye Res 2005; 24(3): 307–32PubMedCrossRef Stefansson E, Pedersen DB, Jensen PK, et al. Optic nerve oxygenation. Prog Retin Eye Res 2005; 24(3): 307–32PubMedCrossRef
141.
go back to reference Lama PJ, Fechtner RD. Antifibrotics and wound healing in glaucoma surgery. Surv Ophthalmol 2003; 48(3): 314–46PubMedCrossRef Lama PJ, Fechtner RD. Antifibrotics and wound healing in glaucoma surgery. Surv Ophthalmol 2003; 48(3): 314–46PubMedCrossRef
142.
go back to reference Meyer-ter-Vehn T, Sieprath S, Katzenberger B, et al. Contractility as a prerequisite for TGF-beta-induced myofibroblast transdifferentiation in human tenon fibroblasts. Invest Ophthalmol Vis Sci 2006; 47(11): 4895–904PubMedCrossRef Meyer-ter-Vehn T, Sieprath S, Katzenberger B, et al. Contractility as a prerequisite for TGF-beta-induced myofibroblast transdifferentiation in human tenon fibroblasts. Invest Ophthalmol Vis Sci 2006; 47(11): 4895–904PubMedCrossRef
143.
go back to reference Mimura F, Yamagishi S, Arimura N, et al. Myelin-associated glycoprotein inhibits microtubule assembly by a Rho-kinase-dependent mechanism. J Biol Chem 2006; 281(23): 15970–9PubMedCrossRef Mimura F, Yamagishi S, Arimura N, et al. Myelin-associated glycoprotein inhibits microtubule assembly by a Rho-kinase-dependent mechanism. J Biol Chem 2006; 281(23): 15970–9PubMedCrossRef
144.
go back to reference Alabed YZ, Grados-Munro E, Ferraro GB, et al. Neuronal responses to myelin are mediated by rho kinase. J Neurochem 2006; 96(6): 1616–25PubMedCrossRef Alabed YZ, Grados-Munro E, Ferraro GB, et al. Neuronal responses to myelin are mediated by rho kinase. J Neurochem 2006; 96(6): 1616–25PubMedCrossRef
145.
go back to reference Borisoff JF, Chan CC, Hiebert GW, et al. Suppression of Rho-kinase activity promotes axonal growth on inhibitory CNS substrates. Mol Cell Neurosci 2003; 22(3): 405–16PubMedCrossRef Borisoff JF, Chan CC, Hiebert GW, et al. Suppression of Rho-kinase activity promotes axonal growth on inhibitory CNS substrates. Mol Cell Neurosci 2003; 22(3): 405–16PubMedCrossRef
146.
go back to reference Fournier AE, Takizawa BT, Strittmatter SM. Rho kinase inhibition enhances axonal regeneration in the injured CNS. J Neurosci 2003; 23(4): 1416–23PubMed Fournier AE, Takizawa BT, Strittmatter SM. Rho kinase inhibition enhances axonal regeneration in the injured CNS. J Neurosci 2003; 23(4): 1416–23PubMed
147.
go back to reference Dergham P, Ellezam B, Essagian C, et al. Rho signaling pathway targeted to promote spinal cord repair. J Neurosci 2002; 22(15): 6570–7PubMed Dergham P, Ellezam B, Essagian C, et al. Rho signaling pathway targeted to promote spinal cord repair. J Neurosci 2002; 22(15): 6570–7PubMed
148.
go back to reference Bertrand J, Di Polo A, McKerracher L. Enhanced survival and regeneration of axotomized retinal neurons by repeated delivery of cell-permeable C3-like Rho antagonists. Neurobiol Dis. 2007 Jan; 25(1): 65–72PubMedCrossRef Bertrand J, Di Polo A, McKerracher L. Enhanced survival and regeneration of axotomized retinal neurons by repeated delivery of cell-permeable C3-like Rho antagonists. Neurobiol Dis. 2007 Jan; 25(1): 65–72PubMedCrossRef
149.
go back to reference Bertrand J, Winton MJ, Rodriguez-Hernandez N, et al. Application of Rho antagonist to neuronal cell bodies promotes neurite growth in compartmented cultures and regeneration of retinal ganglion cell axons in the optic nerve of adult rats. J Neurosci 2005; 25(5): 1113–21PubMedCrossRef Bertrand J, Winton MJ, Rodriguez-Hernandez N, et al. Application of Rho antagonist to neuronal cell bodies promotes neurite growth in compartmented cultures and regeneration of retinal ganglion cell axons in the optic nerve of adult rats. J Neurosci 2005; 25(5): 1113–21PubMedCrossRef
150.
go back to reference Kitaoka Y, Kitaoka Y, Kumai T, et al. Involvement of RhoA and possible neuroprotective effect of fasudil, a Rho kinase inhibitor, in NMDA-induced neurotoxicity in the rat retina. Brain Res 2004; 1018(1): 111–8PubMedCrossRef Kitaoka Y, Kitaoka Y, Kumai T, et al. Involvement of RhoA and possible neuroprotective effect of fasudil, a Rho kinase inhibitor, in NMDA-induced neurotoxicity in the rat retina. Brain Res 2004; 1018(1): 111–8PubMedCrossRef
151.
go back to reference Gallo G, Letourneau PC. Regulation of growth cone actin filaments by guidance cues. J Neurobiol 2004; 58(1): 92–102PubMedCrossRef Gallo G, Letourneau PC. Regulation of growth cone actin filaments by guidance cues. J Neurobiol 2004; 58(1): 92–102PubMedCrossRef
152.
153.
go back to reference Ellis JD, Morris AD, MacEwen CJ. Should diabetic patients be screened for glaucoma? DARTS/MEMO Collaboration. Br J Ophthalmol 1999; 83(3): 369–72PubMedCrossRef Ellis JD, Morris AD, MacEwen CJ. Should diabetic patients be screened for glaucoma? DARTS/MEMO Collaboration. Br J Ophthalmol 1999; 83(3): 369–72PubMedCrossRef
154.
go back to reference Sommer A. Glaucoma risk factors observed in the Baltimore Eye Survey. Curr Opin Ophthalmol 1996; 7(2): 93–8PubMedCrossRef Sommer A. Glaucoma risk factors observed in the Baltimore Eye Survey. Curr Opin Ophthalmol 1996; 7(2): 93–8PubMedCrossRef
Metadata
Title
Rho GTPase/Rho Kinase Inhibition as a Novel Target for the Treatment of Glaucoma
Authors
Dr Vasantha P. Rao
David L. Epstein
Publication date
01-05-2007
Publisher
Springer International Publishing
Published in
BioDrugs / Issue 3/2007
Print ISSN: 1173-8804
Electronic ISSN: 1179-190X
DOI
https://doi.org/10.2165/00063030-200721030-00004

Other articles of this Issue 3/2007

BioDrugs 3/2007 Go to the issue

Adis Drug Profile

Certolizumab Pegol