Skip to main content
Cellular and Molecular Life Sciences: CMLS logoLink to Cellular and Molecular Life Sciences: CMLS
. 2009 Nov 12;67(2):171–177. doi: 10.1007/s00018-009-0189-x

Rho-kinase inhibitors as therapeutics: from pan inhibition to isoform selectivity

C Hahmann 1, T Schroeter 1,
PMCID: PMC11115778  PMID: 19907920

Abstract

The emerging critical implications of Rho/Rho-kinase (ROCK) signaling in neurodegenerative diseases, glaucoma, renoprotection, diabetes and cancer have sparked growing interest in the pharmacological potential of ROCK inhibitors beyond their current application in cardiovascular disease. This article discusses the therapeutic benefits of novel ROCK inhibitors in development, and highlights the recent advances in the current understanding of disease-dependent and isoform-specific functions of ROCK and their potential impact on future therapeutic strategies.

Keywords: Rho-kinase, ROCK, ROCK1, ROCK2, Fasudil, Y-27632


Rho-associated kinase (ROCK) belongs to the AGC (PKA/PKG/PKC) family of serine-threonine protein kinases and is a major downstream effector of the small GTPase RhoA [13]. To date, two ROCK isoforms have been described; ROCK1 (ROKβ, p160ROCK) and ROCK2 (ROKα). ROCK1 and ROCK2 are highly homologous, sharing 65% homology in amino acid sequence and 92% homology in their kinase domains. Although both isoforms are ubiquitously expressed, ROCK1 expression is enriched in lung, liver, spleen, kidney, and testis, whereas ROCK2 is more prominent in the brain and heart [4]. ROCK activity leads to the phosphorylation of downstream targets including myosin light chain (MLC) [5, 6], MLC phosphatase (MYPT1) [7, 8], ezrin/radixin/moesin (ERM) [9], adducin [10], and LIM kinases (LIMK) [1113], thereby modulating actin cytoskeletal organization, stress fiber formation and cell contraction [14]. ROCK controls vascular smooth muscle contraction and endothelial barrier function [1522]. This key role as a master regulator of the vascular bed explains why cardiovascular diseases were the primary indication in early ROCK drug discovery projects and these remain ongoing efforts.

ROCK1−/− and ROCK2−/− knockout mice develop a similar phenotype resulting in eyes-open at birth and omphalocele [2325], demonstrating that the biological functions of ROCK1 and ROCK2 isoforms are in many cases redundant and cannot be separated. For example, both isoforms phosphorylate the same major downstream substrates such as smooth muscle MLC and MYPT1 in vitro [26]. Given the pathway redundancy and the high degree of homology within the kinase domain of both isoforms, it was believed that isoform selectivity could not be achieved, and early drug discovery efforts concentrated on the development of non-isoform selective ROCK inhibitors. To date, the only clinically approved ROCK inhibitor from these efforts is Fasudil, which was approved in Japan in 1995 for the treatment of vasospasm following subarachnoid haemorrhage. Besides regulating the cardiovascular bed, ROCKs are involved in a myriad of biological functions such that inhibition of ROCK activity could be of potential benefit for the treatment of diseases ranging from glaucoma to neurodegenerative diseases to cancer. For an overview see Table 1.

Table 1.

Effects of pharmacological ROCK inhibitors on disease biology

Therapeutic area Compounds Pharmacological effects Species References
Hypertension Fasudil Decrease in blood pressure and vascular resistance Clinical study [66, 67]
AD Decrease in blood pressure Rat, mouse, dog [59, 60]
AA Decrease in blood pressure Rat [34]
SAR407899 Decrease in blood pressure Rat, mouse [31]
Glaucoma Fasudil Decrease in IOP Rabbit [68]
Y-27632 Decrease in IOP, increase in aqueous outflow Rabbit, mouse [37, 69, 70]
Increase in aqueous outflow Monkey, pig [71, 72]
Y-39983 Decrease in IOP Mouse, rabbit, monkey [36, 37]
Neurodegeneration Fasudil, Y-27632 Axonal sprouting and functional recovery after SCI Rat [7376]
Y-27632 Decreased amount of amyloidogenic Aβ42 Mouse [77]
Fasudil, Y-27632, H-1152 Inhibition of mutant huntingtin aggregation Mouse (in vitro) [78]
Oncology Wf-536 (Y-32885) Inhibition of tumor metastasis Mouse [79]
Inhibition of tumor growth, angiogenesis and metastasis Human lung and prostate cancer xenograft [32, 80]
Y-27632 Inhibition of tumor metastasis (in vivo), angiogenesis (in vitro) Human hepatocellular and prostate cancer xenograft [81, 82]
Fasudil Inhibition of tumor progression and metastasis Human lung and breast cancer xenograft [83]
Renoprotection Y-27632 Macrophage infiltration and interstitial fibrosis Mouse [84]
Fasudil Improved proteinuria, glomerulosclerosis, renal interstitial fibrosis and macrophage infiltration Rat [8589]
Improved progression of diabetic nephropathy Mouse [90]
Diabetes Fasudil Prevention of diabetes development Rat [86]
Corrected glucose and lipid metabolism, improved insulin signaling Rat [91]
Erectile dysfunction Y-27632 Improved erectile function Rat [9294]
H-1152, Y-27632 Improved erectile function Rat [95]

All data are derived from in vivo experiments unless otherwise noted

AA Aminofurazan azabenzimidazoles, AD azaindole derivative, IOP intraocular pressure, SCI spinal cord injury

Most of the studies were performed using chemical inhibitors, and one has to be careful how to interpret these data, especially when using Fasudil and Y-27632, considering their selectivity profile and potency. Y-27632 was non-selective against 4 out of 25 tested kinases [27] and Fasudil exhibited non-selectivity for 8 out of 27 tested kinases [27]. Both inhibitors have biochemical IC50s in the hundreds of nanomolar, but they are typically used at concentrations of 10–50 μM in cell-based studies, opening up the possibility that observed phenotypes could be off-target effects. However, based on the overall promising studies showing efficacy of ROCK inhibitors in a variety of animal disease models, there are significant efforts aimed at developing more potent and selective ROCK inhibitors. Recent activities within the pharmaceutical industry and academia have led to a series of novel ROCK inhibitors with enhanced potency within the low nanomolar range and improved kinase selectivity [2834]. In addition, as of July 2009, two compounds, RKI983 (Novartis) and INS117548 (Inspire), are in phase I/II studies for the treatment of glaucoma (NCT00846989, NCT00767793). Moreover, phase II trials for the treatment of glaucoma investigating DE-104 (Santen) as well as phase I/II trials for the treatment of spinal cord injury investigating BA-210 (Alseres Pharmaceuticals) are completed (NCT00650338, NCT00500812). Recently published data from a phase I clinical trial for the treatment of glaucoma using SNJ-1656 [35] showed no adverse systemic effects and only minor side effects in this 7-day repeated-instillation trial when dosed either once or twice a day, i.e., the occurrence of ocular hyperemia was reported, which is in accordance with previous animal studies using Y-39983 [36]. Consistent with the old paradigm that both ROCK isoforms show similarity in their downstream targets, recent studies of the role of ROCK in glaucoma suggest that neither isoform is predominant in modulating intraocular pressure (IOP), since mice deficient for either ROCK1 or ROCK2 exhibit a significant decrease in IOP compared to wild-type littermates [37]. Off-target effects and systemic toxicity might play a minor role for applications such as for the treatment of glaucoma, where pharmacokinetic and pharmacodynamic parameters of inhibitors can be optimized to reduce or eliminate systemic exposure. However, little is known about side effects and toxicity of ROCK inhibitors for systemic and long-term applications and these need to be further investigated. From the knockout mouse models, it is known that inhibition of both ROCK isoforms results in embryonic death due to placental malfunction, and ROCK inhibitors might be counter-indicated for pregnant women. In addition, given ROCK’s involvement in many biological functions, especially the regulation of blood pressure, isoform-selective ROCK inhibitors may be preferential as systemic drugs. For any disease, blood pressure changes will need to be monitored with respect to the efficacious dose of ROCK inhibitor for the indicated disease, and it has to be decided what level of blood pressure decrease is acceptable or even advantageous and what level would be a health concern.

Recent genetic studies as well as animal disease models, along with short interfering RNA (siRNA)-based gene silencing experiments in vitro, provide significant insights into ROCK isoform biology. Cell type-specific cases, in which either ROCK1 or ROCK2 appear to have distinct non-redundant functions, have been shown in fibroblasts and vascular smooth muscle cells. Whereas knockdown of ROCK1 but not ROCK2 leads to disassembly of stress fibers in fibroblasts [26], in smooth muscle cells this phenotype is mediated by knockdown of ROCK2 [38]. These findings could be explained by the fact that both isoforms are expressed to different levels in individual cell types and have unique interaction partners. ROCK1 but not ROCK2 is regulated by RhoE [3941], which competes with RhoA for interaction with ROCK1. In addition, ROCK1 is cleaved by caspase-3 at a conserved sequence in the C-terminus that does not exist in ROCK2, leading to constitutively active ROCK1, MLC2 phosphorylation, and membrane blebbing during apoptosis [42, 43]. In contrast, during granzyme B-induced cell death, granzyme B specifically cleaves and activates ROCK2 but not ROCK1, resulting in downstream MLC2 phosphorylation and membrane blebbing [44]. ROCK1 and ROCK2 activity and signaling, however, are not only regulated differentially by their upstream modulators but both ROCK isoforms also utilize selective downstream partners to mediate their biological functions. For example, ROCK2 induces degradation of transforming growth factor β type I receptor [45], thereby regulating mesoderm induction. In vascular smooth muscle cells, both ROCK isoforms modulate MYPT1 activity but have different effects on smooth muscle cell morphology. Only ROCK2 binds directly to and phosphorylates MYPT1 [38]. In contrast, upon UV-induced stress, ROCK1 activates c-Jun N-terminal kinase (JNK) and induces apoptosis through binding and phosphorylating JNK-interacting protein 3 (JIP-3) [46]. In addition, cell spreading of breast cancer cells during mesenchymal-mode of 3D-migration depends upon specific ROCK1-LIMK2 interaction [47]. The functional and regulatory significance of each isoform is further highlighted during pathological conditions. Induction of pressure overload cardiac hypertrophy in mice by aortic banding shows that, at the state of stable hypertrophy, after 3 weeks the levels of ROCK1 expression and ERM phosphorylation are increased, while ROCK2 expression levels remain unaltered. Even though the disruption of ROCK1 did not affect the development of cardiac hypertrophy, the development of fibrosis in the myocardium was significantly reduced in the ROCK1−/− [48] and ROCK1+/− mice [49]. ROCK isoforms also have distinct contributions in cancer progression. Increased ROCK2 levels have been reported in hepatocellular [50], colon [51], and bladder [52] cancer. A study of 41 pairs of hepatocellular carcinomas revealed that ROCK2 is frequently overexpressed as compared to non-tumorous livers, while ROCK1 expression is unaltered. Silencing of ROCK2 by short-hairpin RNA reduces stress fiber formation, phosphorylation of MYPT1, migration and invasion in vitro, and lung metastasis in vivo [50]. In contrast, ROCK1 expression levels, but not ROCK2, are significantly higher in human mammary tumors and are associated with poor clinical outcome and overall survival of patients [53], and elevated ROCK1 levels were recently reported to be involved in the transformation of hormone-refractory prostate cancer [54]. Overexpression of ROCK isoforms during disease progression could be the cause for or the effect of the disease. Recent advances in siRNA delivery should allow researchers to answer those questions not only in vitro but also in vivo. It is now possible to not only administer siRNA systemically but also to target specific cells or organs. β1,3-D-glucan-encapsulated siRNA nanoparticles were successfully used to target specifically macrophages [55], and magnetic siRNA nanoparticles were used to target solid tumors [56]. Others successfully applied siRNA coupled to an antibody or cell surface receptor ligand such as transferrin [57] or rabies virus glycoprotein [58] to target a specific cell type. In addition, Alcon Research filed a patent application to treat glaucoma using siRNA-mediated knockdown of ROCK (WO/2007/076367), demonstrating that siRNA treatment in vivo cannot only be used as a tool to investigate pathway biology but also shows the potential for the use of siRNA as therapeutic agent.

Despite the significant progress and the increasing understanding of the functions of ROCK isoforms in disease progression, it is notable that recent patents on ROCK inhibitors [30, 34, 59, 60] do not address isoform selectivity. This might be due to the fact that both isoforms show such a high degree in amino acid homology within the kinase domain that it is not believed to be possible to achieve isoform selectivity. In fact, prior to 2008, all published ROCK inhibitors are equally potent against both isoforms. Only recently has one isoform-selective ROCK inhibitor been described. SLx-2119 is a ROCK2 selective compound that has shown promise in cancer xenograft models [61] and in preclinical models of fibrosis [62]. SLx-2119 has further been reported to attenuate arterial plaque formation in apolipoprotein-E-deficient mice. At the same time, this inhibitor avoids unwanted hemodynamic side effects compared with non-selective ROCK inhibitors [63]. Interestingly, these data are in conflict with the findings that ROCK2 polymorphism [64] and a haplotype block consisting of 4 SNPs within the ROCK2 allele [65] are associated with changes in systemic blood pressure.

In conclusion, ROCK inhibition has shown promise as a therapeutic target for a variety of human diseases, and several small molecule inhibitors are in development. For some diseases, such as glaucoma, ROCK isoform selectivity is not required, and for such topical applications the compound can be easily dosed directly to the target organ. A systemic inhibition of ROCK, however, bears the risk of unwanted side effects such as drop of blood pressure and it has to be carefully evaluated if the benefit is to outweigh the risk in a disease-dependent context. Even though ROCKs are expressed ubiquitously, there is rationale for future efforts towards isoform-selective ROCK inhibitors. A growing body of evidence clearly indicates that both ROCK1 and ROCK2 have distinct expression levels and unique interaction partners in individual tissue types, suggesting that these functional differences could be of therapeutic benefit. Yet, further investigation is required to elucidate the regulatory networks of both isoforms in a disease context, especially in their native in vivo environment. This task may be accomplished by recent advances in siRNA delivery technologies allowing targeting to specific organs or cells. Ultimately, this will not only lead to a deeper understanding of the relative importance of each isoform for the pathophysiology of many human diseases, but will also help the next generation of effective ROCK inhibitors to avoid unwanted systemic side effects by targeting each isoform.

Acknowledgments

We thank Patricia McDonald and Michael Cameron for carefully reading of the manuscript and helpful discussions.

References

  • 1.Ishizaki T, Maekawa M, Fujisawa K, Okawa K, Iwamatsu A, Fujita A, Watanabe N, Saito Y, Kakizuka A, Morii N, Narumiya S. The small GTP-binding protein Rho binds to and activates a 160 kDa Ser/Thr protein kinase homologous to myotonic dystrophy kinase. EMBO J. 1996;15:1885–1893. [PMC free article] [PubMed] [Google Scholar]
  • 2.Leung T, Manser E, Tan L, Lim L. A novel serine/threonine kinase binding the Ras-related RhoA GTPase which translocates the kinase to peripheral membranes. J Biol Chem. 1995;270:29051–29054. doi: 10.1074/jbc.270.49.29051. [DOI] [PubMed] [Google Scholar]
  • 3.Matsui T, Amano M, Yamamoto T, Chihara K, Nakafuku M, Ito M, Nakano T, Okawa K, Iwamatsu A, Kaibuchi K. Rho-associated kinase, a novel serine/threonine kinase, as a putative target for small GTP binding protein Rho. EMBO J. 1996;15:2208–2216. [PMC free article] [PubMed] [Google Scholar]
  • 4.Nakagawa O, Fujisawa K, Ishizaki T, Saito Y, Nakao K, Narumiya S. ROCK-I and ROCK-II, two isoforms of Rho-associated coiled-coil forming protein serine/threonine kinase in mice. FEBS Lett. 1996;392:189–193. doi: 10.1016/0014-5793(96)00811-3. [DOI] [PubMed] [Google Scholar]
  • 5.Amano M, Ito M, Kimura K, Fukata Y, Chihara K, Nakano T, Matsuura Y, Kaibuchi K. Phosphorylation and activation of myosin by Rho-associated kinase (Rho-kinase) J Biol Chem. 1996;271:20246–20249. doi: 10.1074/jbc.271.34.20246. [DOI] [PubMed] [Google Scholar]
  • 6.Kureishi Y, Kobayashi S, Amano M, Kimura K, Kanaide H, Nakano T, Kaibuchi K, Ito M. Rho-associated kinase directly induces smooth muscle contraction through myosin light chain phosphorylation. J Biol Chem. 1997;272:12257–12260. doi: 10.1074/jbc.272.19.12257. [DOI] [PubMed] [Google Scholar]
  • 7.Kawano Y, Fukata Y, Oshiro N, Amano M, Nakamura T, Ito M, Matsumura F, Inagaki M, Kaibuchi K. Phosphorylation of myosin-binding subunit (MBS) of myosin phosphatase by Rho-kinase in vivo. J Cell Biol. 1999;147:1023–1038. doi: 10.1083/jcb.147.5.1023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Kimura K, Ito M, Amano M, Chihara K, Fukata Y, Nakafuku M, Yamamori B, Feng J, Nakano T, Okawa K, Iwamatsu A, Kaibuchi K. Regulation of myosin phosphatase by Rho and Rho-associated kinase (Rho-kinase) Science. 1996;273:245–248. doi: 10.1126/science.273.5272.245. [DOI] [PubMed] [Google Scholar]
  • 9.Matsui T, Maeda M, Doi Y, Yonemura S, Amano M, Kaibuchi K, Tsukita S. Rho-kinase phosphorylates COOH-terminal threonines of ezrin/radixin/moesin (ERM) proteins and regulates their head-to-tail association. J Cell Biol. 1998;140:647–657. doi: 10.1083/jcb.140.3.647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Fukata Y, Oshiro N, Kinoshita N, Kawano Y, Matsuoka Y, Bennett V, Matsuura Y, Kaibuchi K. Phosphorylation of adducin by Rho-kinase plays a crucial role in cell motility. J Cell Biol. 1999;145:347–361. doi: 10.1083/jcb.145.2.347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Maekawa M, Ishizaki T, Boku S, Watanabe N, Fujita A, Iwamatsu A, Obinata T, Ohashi K, Mizuno K, Narumiya S. Signaling from Rho to the actin cytoskeleton through protein kinases ROCK and LIM-kinase. Science. 1999;285:895–898. doi: 10.1126/science.285.5429.895. [DOI] [PubMed] [Google Scholar]
  • 12.Ohashi K, Nagata K, Maekawa M, Ishizaki T, Narumiya S, Mizuno K. Rho-associated kinase ROCK activates LIM-kinase 1 by phosphorylation at threonine 508 within the activation loop. J Biol Chem. 2000;275:3577–3582. doi: 10.1074/jbc.275.5.3577. [DOI] [PubMed] [Google Scholar]
  • 13.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;276:670–676. doi: 10.1074/jbc.M007074200. [DOI] [PubMed] [Google Scholar]
  • 14.Riento K, Ridley AJ. Rocks: multifunctional kinases in cell behaviour. Nat Rev Mol Cell Biol. 2003;4:446–456. doi: 10.1038/nrm1128. [DOI] [PubMed] [Google Scholar]
  • 15.Uehata M, Ishizaki T, Satoh H, Ono T, Kawahara T, Morishita T, Tamakawa H, Yamagami K, Inui J, Maekawa M, Narumiya S. Calcium sensitization of smooth muscle mediated by a Rho-associated protein kinase in hypertension. Nature. 1997;389:990–994. doi: 10.1038/40187. [DOI] [PubMed] [Google Scholar]
  • 16.Carbajal JM, Gratrix ML, Yu CH, Schaeffer RC., Jr ROCK mediates thrombin’s endothelial barrier dysfunction. Am J Physiol Cell Physiol. 2000;279:C195–C204. doi: 10.1152/ajpcell.2000.279.1.C195. [DOI] [PubMed] [Google Scholar]
  • 17.Gavard J, Gutkind JS. Protein kinase C-related kinase and ROCK are required for thrombin-induced endothelial cell permeability downstream from Galpha12/13 and Galpha11/q. J Biol Chem. 2008;283:29888–29896. doi: 10.1074/jbc.M803880200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.McKenzie JA, Ridley AJ. Roles of Rho/ROCK and MLCK in TNF-alpha-induced changes in endothelial morphology and permeability. J Cell Physiol. 2007;213:221–228. doi: 10.1002/jcp.21114. [DOI] [PubMed] [Google Scholar]
  • 19.van Nieuw Amerongen GP, Beckers CM, Achekar ID, Zeeman S, Musters RJ, van Hinsbergh VW. Involvement of Rho kinase in endothelial barrier maintenance. Arterioscler Thromb Vasc Biol. 2007;27:2332–2339. doi: 10.1161/ATVBAHA.107.152322. [DOI] [PubMed] [Google Scholar]
  • 20.van Nieuw Amerongen GP, Musters RJ, Eringa EC, Sipkema P, van Hinsbergh VW. Thrombin-induced endothelial barrier disruption in intact microvessels: role of RhoA/Rho kinase-myosin phosphatase axis. Am J Physiol Cell Physiol. 2008;294:C1234–C1241. doi: 10.1152/ajpcell.00551.2007. [DOI] [PubMed] [Google Scholar]
  • 21.van Nieuw Amerongen GP, van Delft S, Vermeer MA, Collard JG, van Hinsbergh VW. Activation of RhoA by thrombin in endothelial hyperpermeability: role of Rho kinase and protein tyrosine kinases. Circ Res. 2000;87:335–340. doi: 10.1161/01.res.87.4.335. [DOI] [PubMed] [Google Scholar]
  • 22.Vandenbroucke E, Mehta D, Minshall R, Malik AB. Regulation of endothelial junctional permeability. Ann N Y Acad Sci. 2008;1123:134–145. doi: 10.1196/annals.1420.016. [DOI] [PubMed] [Google Scholar]
  • 23.Shimizu Y, Thumkeo D, Keel J, Ishizaki T, Oshima H, Oshima M, Noda Y, Matsumura F, Taketo MM, Narumiya S. ROCK-I regulates closure of the eyelids and ventral body wall by inducing assembly of actomyosin bundles. J Cell Biol. 2005;168:941–953. doi: 10.1083/jcb.200411179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Thumkeo D, Keel J, Ishizaki T, Hirose M, Nonomura K, Oshima H, Oshima M, Taketo MM, Narumiya S. Targeted disruption of the mouse rho-associated kinase 2 gene results in intrauterine growth retardation and fetal death. Mol Cell Biol. 2003;23:5043–5055. doi: 10.1128/MCB.23.14.5043-5055.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Thumkeo D, Shimizu Y, Sakamoto S, Yamada S, Narumiya S. ROCK-I and ROCK-II cooperatively regulate closure of eyelid and ventral body wall in mouse embryo. Genes Cells. 2005;10:825–834. doi: 10.1111/j.1365-2443.2005.00882.x. [DOI] [PubMed] [Google Scholar]
  • 26.Yoneda A, Multhaupt HA, Couchman JR. The Rho kinases I and II regulate different aspects of myosin II activity. J Cell Biol. 2005;170:443–453. doi: 10.1083/jcb.200412043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Davies SP, Reddy H, Caivano M, Cohen P. Specificity and mechanism of action of some commonly used protein kinase inhibitors. Biochem J. 2000;351:95–105. doi: 10.1042/0264-6021:3510095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Chen YT, Bannister TD, Weiser A, Griffin E, Lin L, Ruiz C, Cameron MD, Schurer S, Duckett D, Schroter T, LoGrasso P, Feng Y. Chroman-3-amides as potent Rho kinase inhibitors. Bioorgan Med Chem Lett. 2008;18:6406–6409. doi: 10.1016/j.bmcl.2008.10.080. [DOI] [PubMed] [Google Scholar]
  • 29.Feng Y, Yin Y, Weiser A, Griffin E, Cameron MD, Lin L, Ruiz C, Schurer SC, Inoue T, Rao PV, Schroter T, Lograsso P. Discovery of substituted 4-(pyrazol-4-yl)-phenylbenzodioxane-2-carboxamides as potent and highly selective Rho kinase (ROCK-II) inhibitors. J Med Chem. 2008;51:6642–6645. doi: 10.1021/jm800986w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Goodman KB, Cui H, Dowdell SE, Gaitanopoulos DE, Ivy RL, Sehon CA, Stavenger RA, Wang GZ, Viet AQ, Xu W, Ye G, Semus SF, Evans C, Fries HE, Jolivette LJ, Kirkpatrick RB, Dul E, Khandekar SS, Yi T, Jung DK, Wright LL, Smith GK, Behm DJ, Bentley R, Doe CP, Hu E, Lee D. Development of dihydropyridone indazole amides as selective Rho-kinase inhibitors. J Med Chem. 2007;50:6–9. doi: 10.1021/jm0609014. [DOI] [PubMed] [Google Scholar]
  • 31.Lohn M, Plettenburg O, Ivashchenko Y, Kannt A, Hofmeister A, Kadereit D, Schaefer M, Linz W, Kohlmann M, Herbert JM, Janiak P, O’Connor SE, Ruetten H. Pharmacological characterization of SAR407899, a novel rho-kinase inhibitor. Hypertension. 2009;54:676–683. doi: 10.1161/HYPERTENSIONAHA.109.134353. [DOI] [PubMed] [Google Scholar]
  • 32.Nakajima M, Hayashi K, Katayama K, Amano Y, Egi Y, Uehata M, Goto N, Kondo T. Wf-536 prevents tumor metastasis by inhibiting both tumor motility and angiogenic actions. Eur J Pharmacol. 2003;459:113–120. doi: 10.1016/S0014-2999(02)02869-8. [DOI] [PubMed] [Google Scholar]
  • 33.Sessions EH, Yin Y, Bannister TD, Weiser A, Griffin E, Pocas J, Cameron MD, Ruiz C, Lin L, Schurer SC, Schroter T, LoGrasso P, Feng Y. Benzimidazole- and benzoxazole-based inhibitors of Rho kinase. Bioorgan Med Chem Lett. 2008;18:6390–6393. doi: 10.1016/j.bmcl.2008.10.095. [DOI] [PubMed] [Google Scholar]
  • 34.Stavenger RA, Cui H, Dowdell SE, Franz RG, Gaitanopoulos DE, Goodman KB, Hilfiker MA, Ivy RL, Leber JD, Marino JP, Jr, Oh HJ, Viet AQ, Xu W, Ye G, Zhang D, Zhao Y, Jolivette LJ, Head MS, Semus SF, Elkins PA, Kirkpatrick RB, Dul E, Khandekar SS, Yi T, Jung DK, Wright LL, Smith GK, Behm DJ, Doe CP, Bentley R, Chen ZX, Hu E, Lee D. Discovery of aminofurazan-azabenzimidazoles as inhibitors of Rho-kinase with high kinase selectivity and antihypertensive activity. J Med Chem. 2007;50:2–5. doi: 10.1021/jm060873p. [DOI] [PubMed] [Google Scholar]
  • 35.Tanihara H, Inatani M, Honjo M, Tokushige H, Azuma J, Araie M. Intraocular pressure-lowering effects and safety of topical administration of a selective ROCK inhibitor, SNJ-1656, in healthy volunteers. Arch Ophthalmol. 2008;126:309–315. doi: 10.1001/archophthalmol.2007.76. [DOI] [PubMed] [Google Scholar]
  • 36.Tokushige H, Inatani M, Nemoto S, Sakaki H, Katayama K, Uehata M, Tanihara H. Effects of topical administration of y-39983, a selective rho-associated protein kinase inhibitor, on ocular tissues in rabbits and monkeys. Invest Ophthalmol Vis Sci. 2007;48:3216–3222. doi: 10.1167/iovs.05-1617. [DOI] [PubMed] [Google Scholar]
  • 37.Whitlock NA, Harrison B, Mixon T, Yu XQ, Wilson A, Gerhardt B, Eberhart DE, Abuin A, Rice DS. Decreased intraocular pressure in mice following either pharmacological or genetic inhibition of ROCK. J Ocul Pharmacol Therap. 2009;25:187–194. doi: 10.1089/jop.2008.0142. [DOI] [PubMed] [Google Scholar]
  • 38.Wang Y, Zheng XR, Riddick N, Bryden M, Baur W, Zhang X, Surks HK. ROCK isoform regulation of myosin phosphatase and contractility in vascular smooth muscle cells. Circ Res. 2009;104:531–540. doi: 10.1161/CIRCRESAHA.108.188524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Riento K, Guasch RM, Garg R, Jin B, Ridley AJ. RhoE binds to ROCK I and inhibits downstream signaling. Mol Cell Biol. 2003;23:4219–4229. doi: 10.1128/MCB.23.12.4219-4229.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Garg R, Riento K, Keep N, Morris JD, Ridley AJ. N-terminus-mediated dimerization of ROCK-I is required for RhoE binding and actin reorganization. Biochem J. 2008;411:407–414. doi: 10.1042/BJ20071342. [DOI] [PubMed] [Google Scholar]
  • 41.Komander D, Garg R, Wan PT, Ridley AJ, Barford D. Mechanism of multi-site phosphorylation from a ROCK-I:RhoE complex structure. EMBO J. 2008;27:3175–3185. doi: 10.1038/emboj.2008.226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Coleman ML, Sahai EA, Yeo M, Bosch M, Dewar A, Olson MF. Membrane blebbing during apoptosis results from caspase-mediated activation of ROCK I. Nat Cell Biol. 2001;3:339–345. doi: 10.1038/35070009. [DOI] [PubMed] [Google Scholar]
  • 43.Sebbagh M, Renvoize C, Hamelin J, Riche N, Bertoglio J, Breard J. Caspase-3-mediated cleavage of ROCK I induces MLC phosphorylation and apoptotic membrane blebbing. Nat Cell Biol. 2001;3:346–352. doi: 10.1038/35070019. [DOI] [PubMed] [Google Scholar]
  • 44.Sebbagh M, Hamelin J, Bertoglio J, Solary E, Breard J. Direct cleavage of ROCK II by granzyme B induces target cell membrane blebbing in a caspase-independent manner. J Exp Med. 2005;201:465–471. doi: 10.1084/jem.20031877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Zhang Y, Li X, Qi J, Wang J, Liu X, Zhang H, Lin SC, Meng A. Rock2 controls TGF{beta} signaling and inhibits mesoderm induction in zebrafish embryos. J Cell Sci. 2009;122:2197–2207. doi: 10.1242/jcs.040659. [DOI] [PubMed] [Google Scholar]
  • 46.Ongusaha PP, Qi HH, Raj L, Kim YB, Aaronson SA, Davis RJ, Shi Y, Liao JK, Lee SW (2008) Identification of ROCK1 as an upstream activator of the JIP-3 to JNK signaling axis in response to UVB damage. Sci Signal 1, ra14 [DOI] [PMC free article] [PubMed]
  • 47.Shea KF, Wells CM, Garner AP, Jones GE. ROCK1 and LIMK2 interact in spread but not blebbing cancer cells. PLoS ONE. 2008;3:e3398. doi: 10.1371/journal.pone.0003398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Zhang YM, Bo J, Taffet GE, Chang J, Shi J, Reddy AK, Michael LH, Schneider MD, Entman ML, Schwartz RJ, Wei L. Targeted deletion of ROCK1 protects the heart against pressure overload by inhibiting reactive fibrosis. FASEB J. 2006;20:916–925. doi: 10.1096/fj.05-5129com. [DOI] [PubMed] [Google Scholar]
  • 49.Rikitake Y, Oyama N, Wang CY, Noma K, Satoh M, Kim HH, Liao JK. Decreased perivascular fibrosis but not cardiac hypertrophy in ROCK1 ± haploinsufficient mice. Circulation. 2005;112:2959–2965. doi: 10.1161/CIRCULATIONAHA.105.584623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Wong CC, Wong CM, Tung EK, Man K, Ng IO. Rho-kinase 2 is frequently overexpressed in hepatocellular carcinoma and involved in tumor invasion. Hepatology. 2009;49:1583–1594. doi: 10.1002/hep.22836. [DOI] [PubMed] [Google Scholar]
  • 51.Vishnubhotla R, Sun S, Huq J, Bulic M, Ramesh A, Guzman G, Cho M, Glover SC. ROCK-II mediates colon cancer invasion via regulation of MMP-2 and MMP-13 at the site of invadopodia as revealed by multiphoton imaging. Lab Invest. 2007;87:1149–1158. doi: 10.1038/labinvest.3700674. [DOI] [PubMed] [Google Scholar]
  • 52.Kamai T, Tsujii T, Arai K, Takagi K, Asami H, Ito Y, Oshima H. Significant association of Rho/ROCK pathway with invasion and metastasis of bladder cancer. Clin Cancer Res. 2003;9:2632–2641. [PubMed] [Google Scholar]
  • 53.Lane J, Martin TA, Watkins G, Mansel RE, Jiang WG. The expression and prognostic value of ROCK I and ROCK II and their role in human breast cancer. Int J Oncol. 2008;33:585–593. [PubMed] [Google Scholar]
  • 54.Lin SL, Chang D, Ying SY. Hyaluronan stimulates transformation of androgen-independent prostate cancer. Carcinogenesis. 2007;28:310–320. doi: 10.1093/carcin/bgl134. [DOI] [PubMed] [Google Scholar]
  • 55.Aouadi M, Tesz GJ, Nicoloro SM, Wang M, Chouinard M, Soto E, Ostroff GR, Czech MP. Orally delivered siRNA targeting macrophage Map4k4 suppresses systemic inflammation. Nature. 2009;458:1180–1184. doi: 10.1038/nature07774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Medarova Z, Pham W, Farrar C, Petkova V, Moore A. In vivo imaging of siRNA delivery and silencing in tumors. Nat Med. 2007;13:372–377. doi: 10.1038/nm1486. [DOI] [PubMed] [Google Scholar]
  • 57.Hu-Lieskovan S, Heidel JD, Bartlett DW, Davis ME, Triche TJ. Sequence-specific knockdown of EWS-FLI1 by targeted, nonviral delivery of small interfering RNA inhibits tumor growth in a murine model of metastatic Ewing’s sarcoma. Cancer Res. 2005;65:8984–8992. doi: 10.1158/0008-5472.CAN-05-0565. [DOI] [PubMed] [Google Scholar]
  • 58.Kumar P, Wu H, McBride JL, Jung KE, Kim MH, Davidson BL, Lee SK, Shankar P, Manjunath N. Transvascular delivery of small interfering RNA to the central nervous system. Nature. 2007;448:39–43. doi: 10.1038/nature05901. [DOI] [PubMed] [Google Scholar]
  • 59.Kast R, Schirok H, Figueroa-Perez S, Mittendorf J, Gnoth MJ, Apeler H, Lenz J, Franz JK, Knorr A, Hutter J, Lobell M, Zimmermann K, Munter K, Augstein KH, Ehmke H, Stasch JP. Cardiovascular effects of a novel potent and highly selective azaindole-based inhibitor of Rho-kinase. Br J Pharmacol. 2007;152:1070–1080. doi: 10.1038/sj.bjp.0707484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Schirok H, Kast R, Figueroa-Perez S, Bennabi S, Gnoth MJ, Feurer A, Heckroth H, Thutewohl M, Paulsen H, Knorr A, Hutter J, Lobell M, Munter K, Geiss V, Ehmke H, Lang D, Radtke M, Mittendorf J, Stasch JP. Design and synthesis of potent and selective azaindole-based Rho kinase (ROCK) inhibitors. ChemMedChem. 2008;3:1893–1904. doi: 10.1002/cmdc.200800211. [DOI] [PubMed] [Google Scholar]
  • 61.Shifrin V, Annand RR, Flusberg D, McGonigle S, Wong E, Paradise E, Bartolozzi A, Ram S, Foudoulakis H, Kirk B, Chesworth R, Riesinger S, Grogan M, Tsaioun K, Malchoff A, Ter-Ovanesyan E, Waechter R, Duffy D, Kim E, Schueller O, Campbell S (2005) Effects of SLx-2119, a novel small molecule inhibitor of Rho-associated kinase ROCK (ROK), on growth of human tumor xenografts in nude mice. AACR Meeting Abstracts 2005, 158
  • 62.Boerma M, Fu Q, Wang J, Loose DS, Bartolozzi A, Ellis JL, McGonigle S, Paradise E, Sweetnam P, Fink LM, Vozenin-Brotons MC, Hauer-Jensen M. Comparative gene expression profiling in three primary human cell lines after treatment with a novel inhibitor of Rho kinase or atorvastatin. Blood Coagul Fibrinolysis. 2008;19:709–718. doi: 10.1097/MBC.0b013e32830b2891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Schueller O, Tong W, Ferkany JW, Sweetnam P (2006) Abstract 1216: Selective ROCK 2 inhibition attenuates arterial plaque formation in an ApoE knockout mouse model. Circulation 114, II_228-b-
  • 64.Seasholtz TM, Wessel J, Rao F, Rana BK, Khandrika S, Kennedy BP, Lillie EO, Ziegler MG, Smith DW, Schork NJ, Brown JH, O’Connor DT. Rho kinase polymorphism influences blood pressure and systemic vascular resistance in human twins: role of heredity. Hypertension. 2006;47:937–947. doi: 10.1161/01.HYP.0000217364.45622.f0. [DOI] [PubMed] [Google Scholar]
  • 65.Rankinen T, Church T, Rice T, Markward N, Blair SN, Bouchard C. A major haplotype block at the rho-associated kinase 2 locus is associated with a lower risk of hypertension in a recessive manner: the HYPGENE study. Hypertens Res. 2008;31:1651–1657. doi: 10.1291/hypres.31.1651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Masumoto A, Hirooka Y, Shimokawa H, Hironaga K, Setoguchi S, Takeshita A. Possible involvement of Rho-kinase in the pathogenesis of hypertension in humans. Hypertension. 2001;38:1307–1310. doi: 10.1161/hy1201.096541. [DOI] [PubMed] [Google Scholar]
  • 67.Fukumoto Y, Matoba T, Ito A, Tanaka H, Kishi T, Hayashidani S, Abe K, Takeshita A, Shimokawa H. Acute vasodilator effects of a Rho-kinase inhibitor, fasudil, in patients with severe pulmonary hypertension. Heart. 2005;91:391–392. doi: 10.1136/hrt.2003.029470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Honjo M, Inatani M, Kido N, Sawamura T, Yue BY, Honda Y, Tanihara H. Effects of protein kinase inhibitor, HA1077, on intraocular pressure and outflow facility in rabbit eyes. Arch Ophthalmol. 2001;119:1171–1178. doi: 10.1001/archopht.119.8.1171. [DOI] [PubMed] [Google Scholar]
  • 69.Honjo M, Tanihara H, Inatani M, Kido N, Sawamura T, Yue BY, Narumiya S, Honda Y. Effects of rho-associated protein kinase inhibitor Y-27632 on intraocular pressure and outflow facility. Invest Ophthalmol Vis Sci. 2001;42:137–144. [PubMed] [Google Scholar]
  • 70.Waki M, Yoshida Y, Oka T, Azuma M. Reduction of intraocular pressure by topical administration of an inhibitor of the Rho-associated protein kinase. Curr Eye Res. 2001;22:470–474. doi: 10.1076/ceyr.22.6.470.5489. [DOI] [PubMed] [Google Scholar]
  • 71.Rao PV, Deng PF, Kumar J, Epstein DL. Modulation of aqueous humor outflow facility by the Rho kinase-specific inhibitor Y-27632. Invest Ophthalmol Vis Sci. 2001;42:1029–1037. [PubMed] [Google Scholar]
  • 72.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;80:215–225. doi: 10.1016/j.exer.2004.09.002. [DOI] [PubMed] [Google Scholar]
  • 73.Chan CC, Khodarahmi K, Liu J, Sutherland D, Oschipok LW, Steeves JD, Tetzlaff W. Dose-dependent beneficial and detrimental effects of ROCK inhibitor Y27632 on axonal sprouting and functional recovery after rat spinal cord injury. Exp Neurol. 2005;196:352–364. doi: 10.1016/j.expneurol.2005.08.011. [DOI] [PubMed] [Google Scholar]
  • 74.Fournier AE, Takizawa BT, Strittmatter SM. Rho kinase inhibition enhances axonal regeneration in the injured CNS. J Neurosci. 2003;23:1416–1423. doi: 10.1523/JNEUROSCI.23-04-01416.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Hara M, Takayasu M, Watanabe K, Noda A, Takagi T, Suzuki Y, Yoshida J. Protein kinase inhibition by fasudil hydrochloride promotes neurological recovery after spinal cord injury in rats. J Neurosurg. 2000;93:94–101. doi: 10.3171/spi.2000.93.1.0094. [DOI] [PubMed] [Google Scholar]
  • 76.Tanaka H, Yamashita T, Yachi K, Fujiwara T, Yoshikawa H, Tohyama M. Cytoplasmic p21(Cip1/WAF1) enhances axonal regeneration and functional recovery after spinal cord injury in rats. Neuroscience. 2004;127:155–164. doi: 10.1016/j.neuroscience.2004.05.010. [DOI] [PubMed] [Google Scholar]
  • 77.Zhou Y, Su Y, Li B, Liu F, Ryder JW, Wu X, Gonzalez-DeWhitt PA, Gelfanova V, Hale JE, May PC, Paul SM, Ni B. Nonsteroidal anti-inflammatory drugs can lower amyloidogenic Abeta42 by inhibiting Rho. Science. 2003;302:1215–1217. doi: 10.1126/science.1090154. [DOI] [PubMed] [Google Scholar]
  • 78.Bauer PO, Wong HK, Oyama F, Goswami A, Okuno M, Kino Y, Miyazaki H, Nukina N. Inhibition of Rho kinases enhances the degradation of mutant huntingtin. J Biol Chem. 2009;284:13153–13164. doi: 10.1074/jbc.M809229200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Nakajima M, Hayashi K, Egi Y, Katayama K, Amano Y, Uehata M, Ohtsuki M, Fujii A, Oshita K, Kataoka H, Chiba K, Goto N, Kondo T. Effect of Wf-536, a novel ROCK inhibitor, against metastasis of B16 melanoma. Cancer Chemother Pharmacol. 2003;52:319–324. doi: 10.1007/s00280-003-0641-9. [DOI] [PubMed] [Google Scholar]
  • 80.Somlyo AV, Phelps C, Dipierro C, Eto M, Read P, Barrett M, Gibson JJ, Burnitz MC, Myers C, Somlyo AP. Rho kinase and matrix metalloproteinase inhibitors cooperate to inhibit angiogenesis and growth of human prostate cancer xenotransplants. FASEB J. 2003;17:223–234. doi: 10.1096/fj.02-0655com. [DOI] [PubMed] [Google Scholar]
  • 81.Somlyo AV, Bradshaw D, Ramos S, Murphy C, Myers CE, Somlyo AP. Rho-kinase inhibitor retards migration and in vivo dissemination of human prostate cancer cells. Biochem Biophys Res Commun. 2000;269:652–659. doi: 10.1006/bbrc.2000.2343. [DOI] [PubMed] [Google Scholar]
  • 82.Takamura M, Sakamoto M, Genda T, Ichida T, Asakura H, Hirohashi S. Inhibition of intrahepatic metastasis of human hepatocellular carcinoma by Rho-associated protein kinase inhibitor Y-27632. Hepatology. 2001;33:577–581. doi: 10.1053/jhep.2001.22652. [DOI] [PubMed] [Google Scholar]
  • 83.Ying H, Biroc SL, Li WW, Alicke B, Xuan JA, Pagila R, Ohashi Y, Okada T, Kamata Y, Dinter H. The Rho kinase inhibitor fasudil inhibits tumor progression in human and rat tumor models. Mol Cancer Ther. 2006;5:2158–2164. doi: 10.1158/1535-7163.MCT-05-0440. [DOI] [PubMed] [Google Scholar]
  • 84.Nagatoya K, Moriyama T, Kawada N, Takeji M, Oseto S, Murozono T, Ando A, Imai E, Hori M. Y-27632 prevents tubulointerstitial fibrosis in mouse kidneys with unilateral ureteral obstruction. Kidney Int. 2002;61:1684–1695. doi: 10.1046/j.1523-1755.2002.00328.x. [DOI] [PubMed] [Google Scholar]
  • 85.Kanda T, Wakino S, Hayashi K, Homma K, Ozawa Y, Saruta T. Effect of fasudil on Rho-kinase and nephropathy in subtotally nephrectomized spontaneously hypertensive rats. Kidney Int. 2003;64:2009–2019. doi: 10.1046/j.1523-1755.2003.00300.x. [DOI] [PubMed] [Google Scholar]
  • 86.Kikuchi Y, Yamada M, Imakiire T, Kushiyama T, Higashi K, Hyodo N, Yamamoto K, Oda T, Suzuki S, Miura S. A Rho-kinase inhibitor, fasudil, prevents development of diabetes and nephropathy in insulin-resistant diabetic rats. J Endocrinol. 2007;192:595–603. doi: 10.1677/JOE-06-0045. [DOI] [PubMed] [Google Scholar]
  • 87.Nishikimi T, Akimoto K, Wang X, Mori Y, Tadokoro K, Ishikawa Y, Shimokawa H, Ono H, Matsuoka H. Fasudil, a Rho-kinase inhibitor, attenuates glomerulosclerosis in Dahl salt-sensitive rats. J Hypertens. 2004;22:1787–1796. doi: 10.1097/00004872-200409000-00024. [DOI] [PubMed] [Google Scholar]
  • 88.Nishikimi T, Koshikawa S, Ishikawa Y, Akimoto K, Inaba C, Ishimura K, Ono H, Matsuoka H. Inhibition of Rho-kinase attenuates nephrosclerosis and improves survival in salt-loaded spontaneously hypertensive stroke-prone rats. J Hypertens. 2007;25:1053–1063. doi: 10.1097/HJH.0b013e3280825440. [DOI] [PubMed] [Google Scholar]
  • 89.Satoh S, Yamaguchi T, Hitomi A, Sato N, Shiraiwa K, Ikegaki I, Asano T, Shimokawa H. Fasudil attenuates interstitial fibrosis in rat kidneys with unilateral ureteral obstruction. Eur J Pharmacol. 2002;455:169–174. doi: 10.1016/S0014-2999(02)02619-5. [DOI] [PubMed] [Google Scholar]
  • 90.Kolavennu V, Zeng L, Peng H, Wang Y, Danesh FR. Targeting of RhoA/ROCK signaling ameliorates progression of diabetic nephropathy independent of glucose control. Diabetes. 2008;57:714–723. doi: 10.2337/db07-1241. [DOI] [PubMed] [Google Scholar]
  • 91.Kanda T, Wakino S, Homma K, Yoshioka K, Tatematsu S, Hasegawa K, Takamatsu I, Sugano N, Hayashi K, Saruta T. Rho-kinase as a molecular target for insulin resistance and hypertension. FASEB J. 2006;20:169–171. doi: 10.1096/fj.05-4197fje. [DOI] [PubMed] [Google Scholar]
  • 92.Chitaley K, Wingard CJ, Clinton Webb R, Branam H, Stopper VS, Lewis RW, Mills TM. Antagonism of Rho-kinase stimulates rat penile erection via a nitric oxide-independent pathway. Nat Med. 2001;7:119–122. doi: 10.1038/83258. [DOI] [PubMed] [Google Scholar]
  • 93.Rajasekaran M, White S, Baquir A, Wilkes N. Rho-kinase inhibition improves erectile function in aging male Brown-Norway rats. J Androl. 2005;26:182–188. doi: 10.1002/j.1939-4640.2005.tb01084.x. [DOI] [PubMed] [Google Scholar]
  • 94.Wingard CJ, Johnson JA, Holmes A, Prikosh A. Improved erectile function after Rho-kinase inhibition in a rat castrate model of erectile dysfunction. Am J Physiol Regul Integr Comp Physiol. 2003;284:R1572–R1579. doi: 10.1152/ajpregu.00041.2003. [DOI] [PubMed] [Google Scholar]
  • 95.Teixeira CE, Ying Z, Webb RC. Proerectile effects of the Rho-kinase inhibitor (S)-(+)-2-methyl-1-[(4-methyl-5-isoquinolinyl)sulfonyl]homopiperazine (H-1152) in the rat penis. J Pharmacol Exp Ther. 2005;315:155–162. doi: 10.1124/jpet.105.086041. [DOI] [PubMed] [Google Scholar]

Articles from Cellular and Molecular Life Sciences: CMLS are provided here courtesy of Springer

RESOURCES