Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2009 Jun 8.
Published in final edited form as: Am J Physiol Cell Physiol. 2006 Mar;290(3):C661–C668. doi: 10.1152/ajpcell.00459.2005

Physiological role of ROCKs in the cardiovascular system

Kensuke Noma 1, Naotsugu Oyama 1, James K Liao 1
PMCID: PMC2692274  NIHMSID: NIHMS89184  PMID: 16469861

Abstract

Rho-associated kinases (ROCKs), the immediate downstream targets of RhoA, are ubiquitously expressed serine-threonine protein kinases that are involved in diverse cellular functions, including smooth muscle contraction, actin cytoskeleton organization, cell adhesion and motility, and gene expression. Recent studies have shown that ROCKs may play a pivotal role in cardiovascular diseases such as vasospastic angina, ischemic stroke, and heart failure. Indeed, inhibition of ROCKs by statins or other selective inhibitors leads to the upregulation and activation of endothelial nitric oxide synthase (eNOS) and reduction of vascular inflammation and atherosclerosis. Thus inhibition of ROCKs may contribute to some of the cholesterol-independent beneficial effects of statin therapy. Currently, two ROCK isoforms have been identified, ROCK1 and ROCK2. Because ROCK inhibitors are nonselective with respect to ROCK1 and ROCK2 and also, in some cases, may be nonspecific with respect to other ROCK-related kinases such as myristolated alanine-rich C kinase substrate (MARCKS), protein kinase A, and protein kinase C, the precise role of ROCKs in cardiovascular disease remains unknown. However, with the recent development of ROCK1- and ROCK2-knockout mice, further dissection of ROCK signaling pathways is now possible. Herein we review what is known about the physiological role of ROCKs in the cardiovascular system and speculate about how inhibition of ROCKs could provide cardiovascular benefits.

Keywords: Rho GTPase, Rho-kinase, vascular endothelium, contraction, actin cytoskeleton, nitric oxide, statins


The small GTP-binding proteins belonging to the Ras, Rho, Rab, and Ran subfamilies act as molecular “on-off” switches that control multiple signaling pathways (24, 36, 116). Among them, the Rho GTPases are implicated in a variety of physiological functions associated with changes in the actin cytoskeleton, such as cell adhesion, motility, migration, and contraction (77, 109). Downstream targets of Rho GTPases that are involved in actin cytoskeletal reorganization include citron kinase, p140mDia, protein kinase N (PKN), p21-activated protein kinase (PAK), rhophillin, and rhotekin. The Rho-associated kinases (ROCKs) were found to be one of the first downstream targets of RhoA (41, 60, 68). ROCKs are important regulators of cell growth, migration, and apoptosis via control of actin cytoskeletal assembly. They regulate cell contraction through serine-threonine phosphorylation of adducin, ezrin-radixin-moesin (ERM) proteins, LIM kinase, myosin light chain phosphatase (MLCP), and Na/H exchanger (NHE)1 (21, 87). For example, ROCKs increase myosin light chain (MLC) phosphorylation through phosphorylating the myosin binding subunit (MBS) on MLCP and inhibiting the activity of MLCP, leading to vascular smooth muscle cell (VSMC) contraction (49, 53, 102). ROCKs can also phosphorylate and inhibit LIM kinase, which phosphorylates the cofilin/actindepolymerizing factor complex involved in the depolymerization and severing of actin filaments (64). Thus the overall physiological effects of ROCKs are to enhance actin-myosin association through increasing MLC phosphorylation and preventing actin depolymerization.

In the mammalian system, ROCKs consist of two isoforms, ROCK1 and ROCK2. ROCK1, which is also known as ROKβ and p160ROCK, is located on chromosome 18 and encodes a 1,354-amino acid protein (41, 68). ROCK2, which is also known as ROKα and often referred to as Rho-kinase, is located on chromosome 12 and contains 1,388 amino acids (87). Y-27632 and fasudil are selective ROCK inhibitors that target their ATP-dependent kinase domains and therefore are equipotent in terms of inhibiting both ROCK1 and ROCK2. However, at higher concentrations, these ROCK inhibitors could also inhibit other kinases such as PKA and PKC (88). Thus current commercially available ROCK inhibitors are not entirely specific for ROCKs compared with other serine-threonine kinases in vivo and cannot distinguish between ROCK1 and ROCK2. Nevertheless, with Y-27632 and fasudil, some evidence is emerging regarding the physiological role of ROCKs, particularly in cardiovascular disease (38, 94, 100).

The emergence of ROCKs in cardiovascular disease coincides with the growing acceptance of the so-called “pleiotropic” or cholesterol-independent effects of 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors or statins. In particular, accumulating evidence indicates that endothelial nitric oxide synthase (eNOS), which is an important mediator of endothelial function, is regulated by the RhoA/ROCK pathway (55, 58, 71, 110, 124). For example, inhibition of RhoA geranylgeranylation by statins decreases membrane GTP-bound active RhoA and subsequent ROCK activity, leading to the upregulation and activation of eNOS (58). Similarly, direct inhibition of the RhoA/ROCK pathway by ROCK inhibitors or dominant-negative mutant of RhoA has been shown to increase eNOS expression (88, 124). Because eNOS is protective of the vasculature, these studies suggest that the RhoA/ROCK pathway may play a significant pathophysiological role in various aspects of cardiovascular disease. Indeed, ROCKs are activated in patients with cardiovascular disease or its associated risk factors, suggesting that ROCK inhibition may be a useful therapeutic target in the cardiovascular system (50, 66, 79, 99).

REGULATION OF RhoA

The Rho GTPases, Rho, Rac, and Cdc42, are involved in the regulation of vital cellular functions such as cytoskeleton organization, motility, proliferation, adhesion, apoptosis, and gene expression (10, 76, 77, 109). In particular, RhoA regulates the assembly of actin stress fibers (85), whereas Rac and Cdc42 regulate actin polymerization and cellular protrusions (86). As with other Rho GTPases, RhoA functions as a molecular switch, cycling between an active GTP-bound form and an inactive GDP-bound form. The cycle between active and inactive forms is controlled by several regulatory proteins, including guanine dissociation inhibitors (GDIs), guanine nucleotide exchange factors (GEFs), and GTPase-activating proteins (GAPs) (24, 73, 95, 116). Rho GEFs activate RhoA by catalyzing the exchange of GDP to GTP, whereas Rho GAPs inactivate RhoA by dephosphorylating GTP to GDP. The GDIs prevent the membrane translocation of RhoA from the cytosol. To date, numerous proteins have been identified as potential Rho GEFs and GAPs. In addition, numerous proteins function as downstream effectors of Rho GTPase, including ROCKs, suggesting the central importance of these proteins in cellular homeostasis (24).

ISOFORM AND STRUCTURE OF ROCKs

ROCK1 and ROCK2 are protein serine-threonine kinases that share 45–50% homology with other actin cytoskeletal kinases such as citron kinase, myotonic dystrophy kinase (DMPK), and myotonic dystrophy-related cdc42-binding kinase (MRCK) (87). ROCKs consist of an amino-terminal kinase domain, followed by a mid-coiled-coil-forming lesion containing a Rho-binding domain (RBD), and a carboxyterminal cysteine-rich domain (CRD) located within the pleckstrin homology (PH) motif. ROCK1 and ROCK2 share overall 65% homology in amino acid sequence and 92% homology in their kinase domains. ROCK1 and ROCK2 are ubiquitously expressed in mouse tissues from early embryonic development to adulthood. ROCK1 mRNA is preferentially expressed in lung, liver, spleen, kidney, and testis, whereas ROCK2 mRNA is highly expressed in the heart and brain (22, 75, 121). In addition, immunolocalization and cell fractionation studies have shown that ROCK2 is distributed mainly in the cytoplasm (60, 68). When activated by GTP-bound RhoA, ROCK2 translocates from cytoplasm to membranes. Indeed, a small amount of ROCK2 has been found in the membrane fraction, and some immunostaining is detectable at the cell periphery or membranes of growing cells (49). In contrast, little is known regarding the intracellular localization of ROCK1. However, a recent study suggests that ROCK1 may be colocalized to centrosomes (15).

Recently, ROCK1-deficient (ROCK1−/−) and ROCK2-deficient (ROCK2−/−) mice have been generated (98, 111). Both mutant ROCK mice were generated by knocking in a lacZ reporter gene. In the ROCK2−/− mouse, LacZ staining was observed in many locations throughout the embryo, including the dorsal root ganglions, heart, liver, and umbilical blood vessels. In the placenta, strong LacZ staining was also observed in the labyrinth layer. In the ROCK1−/− mouse, LacZ staining was also detected throughout the embryo, including the aorta, dorsal root ganglia, heart, skin, and umbilical blood vessels. ROCK2−/− embryos are embryonically lethal because of placental dysfunction and intrauterine growth retardation caused by thrombus formation in the labyrinth layer of the placenta. ROCK1−/− mice, however, exhibit a completely different phenotype compared with ROCK2−/− mice. They die postnatally because of an abnormality of filamentous actin accumulation that leads to impairment in umbilical ring closure. The ROCK1−/− mice also exhibit eyes open at birth (EOB) due to disorganization of actin filaments in the epithelial cells of the eyelid. Interestingly, a substantial proportion of ROCK2−/− mouse embryos exhibit EOB and omphalocele, indicating that both ROCK1 and ROCK2 may play important roles in mediating eye and umbilical ring closure. Thus further studies regarding the specific roles of ROCK1 and ROCK2 with haploinsufficient or conditional ROCK1- and ROCK2-knockout mice are needed. Recently, it was reported that haploinsufficient ROCK1-knockout mice are viable and healthy at birth and exhibit decreased cardiac fibrosis, but not hypertrophy, in response to angiotensin II infusion (90).

REGULATION OF ROCKs

The carboxy-terminal regions of ROCKs, which contain the PH domain and the RBD, serve as an autoregulatory inhibitor of the amino-terminal kinase domain (4). The interaction of GTP-bound RhoA to the RBD of ROCKs increases ROCK activity through derepression of the carboxy-terminal RBD-PH domains on the amino-terminal kinase domain, leading to an active “open” kinase conformation. This open conformation could also be caused by the binding of arachidonic acid to the PH domain (28) or by cleavage of the carboxy terminus by caspase-3 (18, 96). Such a regulatory mechanism could also be found with DMPK and MRCK activation (4, 13) and is consistent with studies showing that overexpression of various carboxy-terminal constructs of ROCKs or kinase-defective forms of full-length ROCKs function as dominant-negative ROCK mutants (3, 41, 59). Interestingly, ROCKs could also be activated independently of RhoA through amino-terminal transphosphorylation caused by protein oligomerization (13, 113). Other small GTP-binding proteins such as Gem and Rad specifically regulate either ROCK1- or ROCK2-mediated cell rounding and neurite retraction (120). Although further studies are needed to uncover the precise mechanism, these results indicate that ROCK1 and ROCK2 may have different physiological roles in cellular function.

DOWNSTREAM TARGETS OF ROCKs

ROCKs phosphorylate various targets and mediate a broad range of cellular responses that involve the actin cytoskeleton in response to GTP-bound RhoA by activators of RhoA such as lysophosphatidic acid (LPA) or sphingosine-1 phosphate (S1P), which stimulate Rho GEFs. ROCKs control assembly of the actin cytoskeleton and cell contractility via phosphorylation of various downstream target proteins, including MBS on MLCP, MLC, ERM proteins, protein LIM kinase, and adducin (Fig. 1). The consensus amino acid sequences for phosphorylation are R/KXS/T or R/KXXS/T (R, arginine; K, lysine; X, any amino acid; S, serine; T, threonine) (46, 106). Of interest, ROCKs can be autophosphorylated, suggesting that the function of ROCKs may be dependent in part on autoregulation (41, 60). MBS on MLCP is an important downstream target protein of ROCKs. Phosphorylation of MBS on MLCP by ROCKs leads to the phosphorylation of MLC and subsequent contraction of VSMCs (103). MLCP holoenzyme is composed of three subunits: a catalytic subunit (PP1δ), a MBS composed of a 58-kDa head and a 32-kDa tail region, and a small noncatalytic subunit, M21. ROCK2 phosphorylates MBS at Thr697, Ser854, and Thr855 (46). The functional significance of MBS phosphorylation at Ser854, however, is not known. Phosphorylation of Thr697 or Thr855 attenuates MLCP activity and, in some instances, the dissociation of MLCP from myosin (27, 119). In addition, MLC is one of the major downstream target proteins of ROCKs. ROCK2 phosphorylates Ser19 of MLC, the same residue that is phosphorylated by MLC kinase. Thus ROCK2 may increase cellular contractility via dual effects on MLC kinase and MLCP. Indeed, ROCK2 can alter the sensitivity of VSMC contraction in response to changes in Ca2+ concentration (5). ROCKs also phosphorylate ERM proteins, which serve as cross-linkers between actin filaments and membrane proteins at the cell surface. ROCK-mediated phosphorylation of ERM proteins, namely, Thr567 of ezrin, Thr564 of radixin, and Thr558 of moesin, leads to the disruption of the head-to-tail association of ERM proteins and actin cytoskeletal reorganization (69). However, it is not known whether phosphorylation of MBS on MLCP or ERM proteins is specific to ROCK isoforms. Nevertheless, ROCK1 phosphorylates LIM kinase-1 at Thr508 and LIM kinase-2 at Thr505, which inhibits cofilin-mediated actin filament disassembly by phosphorylating cofilin (64, 81, 106). Adducin, which is a membrane skeletal protein that associates with and promotes the association of spectrin with F-actin, is also a downstream target of ROCK2 (32, 48). Adducin is localized at cell-cell contact sites and is thought to participate in the assembly of the spectrin-actin network by capping the fast-growing ends of actin filaments and recruiting spectrin to the filament ends. The phosphorylation of α-adducin by ROCK2 enhances the binding activity of α-adducin to F-actin, thereby increasing the contractile response.

Fig. 1.

Fig. 1

Role of Rho-associated kinases (ROCKs) in cardiovascular disease. Activated ROCKs, mediated through GTP-binding active RhoA, phosphorylate various downstream targets of ROCKs. Most of these processes lead to cardiovascular injury, including hypertension, pulmonary hypertension, erectile dysfunction, atherosclerosis, vasospasm, cerebral ischemia, neointima formation, myocardial hypertrophy, ischemia-reperfusion injury, and so forth. MLC, myosin light chain; MLCP, MLC phosphatase; ERM, ezrin-radixin-moesin proteins.

Although the homology of the kinase domain between ROCK1 and ROCK2 is 92%, ROCK1 and ROCK2 may serve different functions and may have different downstream target proteins. Most of the downstream targets of ROCKs are cellular proteins associated with the regulation of the actin cytoskeleton. The difficulty with studying the roles of ROCKs is due to the lack of specific inhibitors that can distinguish not only the roles of ROCK isoforms but also the roles of ROCKs from other serine-threonine kinases, such as PKA, PKC, and citron kinase. Therefore, a genetic approach using conditional or haploinsufficient ROCK1- and ROCK2-knockout mice offers the best opportunity to dissect the functions of ROCK1 and ROCK2.

FUNCTIONS OF ROCKs

ROCKs are important regulators of cellular apoptosis, growth, metabolism, and migration via control of the actin cytoskeletal assembly and cell contraction (87). ROCK1 and ROCK2 are differentially expressed and regulated in specific tissues. For instance, only ROCK1 is cleaved by caspase-3 during apoptosis (18, 96). On the other hand, smooth muscle-specific basic calponin is phosphorylated only by ROCK2 (44). Furthermore, ROCK1 is highly expressed ubiquitously, whereas ROCK2 is preferentially expressed in cardiac and brain tissues (22, 75, 121). With the deletion of ROCK1 and ROCK2 alleles in mice, distinguishing the physiological roles of ROCK isoforms is now feasible.

Stimulation of tyrosine kinase and G protein-coupled receptors recruits and activates Rho GEFs, leading to activation of RhoA, the direct upstream activator of ROCKs (37, 52). ROCKs are pivotal downstream effectors of RhoA in regulating the actin cytoskeleton. Inhibitors of ROCKs such as Y-27632 and fasudil, or overexpression of dominant-negative mutants of ROCK, cause loss of actin stress fibers and focal adhesion complexes (41, 114). This is predominantly due to the phosphorylation and inhibition of MLCP by ROCKs, which increase MLC phosphorylation and cellular contraction by facilitating interaction of myosin with F-actin. Thus ROCKs regulate cell polarity and migration through cellular contractions, protrusions, and focal adhesions. By affecting tight and adherens junctions through actin cytoskeletal contractions, ROCKs can also regulate macrophage phagocytic activity and endothelial cell permeability (122, 123).

ROCKs could regulate other cellular functions independent of their effects on the actin cytoskeleton. For example, ROCKs inhibit insulin signaling via direct phosphorylation of insulin receptor substrate-1 (IRS-1). Phosphorylation of IRS-1 uncouples the insulin receptor from phosphatidylinositol 3-kinase (PI3K) and potentially Glut4 activation (26). ROCKs could also regulate cell size by enhancing IGF-induced cAMP response element binding protein (CREB) phosphorylation (104), which may be the underlying mechanism by which ROCK inhibitors reduce cardiac hypertrophy (80, 83). However, a recent study in haploinsufficient ROCK1+/− mice indicates that ROCK1 is required for the development of cardiac fibrosis, not hypertrophy (90). Interestingly, ROCKs may be involved in tissue differentiation from adipocytes to myocyte. In p190-B Rho GAP-deficient mice in which the RhoA/ROCK pathway is chronically activated, there is a defect in adipogenesis, with a predilection toward myogenesis (104, 105). Treatment of p190-B Rho GAP-deficient mice with Y-27632 restores normal adipogenesis (105), suggesting that ROCKs are involved in the regulation of the myocyte differentiation.

REGULATION OF eNOS BY ROCKs

The vascular endothelium secretes vasoactive substances such as NO, prostacyclin, endothelium-derived hyperpolarizing factor, and endothelins (63, 84, 117). In particular, endothelium-derived NO plays an important role in the regulation of vascular tone, inhibition of platelet aggregation, suppression of smooth muscle cell proliferation, and prevention of leukocyte recruitment to the vessel wall (115, 118). Increased bioavailability of NO is, in part, dependent on increased expression and activity of eNOS as well as on decreased inactivation of NO by reactive oxygen species (ROS), specifically the superoxide anion (Fig. 2). Although various conditions and factors such as laminar shear stress (19), oxygen tension (62), and TGF-β1 (93) can regulate eNOS expression at the transcriptional level, eNOS expression can be also regulated at the posttranscriptional level. For example, chronic hypoxia (55), TNF-α (125), thrombin (25), oxidized LDL (61), and cellular proliferation (6, 31) are known to decrease eNOS mRNA stability. Chronic hypoxia and cellular proliferation are known to activate RhoA and ROCKs (110). In contrast, statins, which have been shown to increase eNOS mRNA stability, inhibit RhoA geranylgeranylation (58) and ROCK activity (110). Thus RhoA/ROCK inversely regulates eNOS expression through alteration in eNOS mRNA stability.

Fig. 2.

Fig. 2

Regulation of endothelial nitric oxide synthase (eNOS) by ROCKs. Statins decrease the synthesis of isoprenoids, resulting in the indirect inhibition of the RhoA/ROCK pathway. The indirect inhibition of the RhoA/ROCK pathway by statins as well as the direct inhibition of ROCKs by ROCK inhibitors leads to increased eNOS expression and eNOS activity by stabilization of eNOS mRNA or phosphorylation of eNOS, respectively. AII, angiotensin II; ET-1, endothelin 1; GEF, guanine nucleotide exchange factor; GAP, GTPase activating protein; HMG-CoA, 3-hydroxy-3-methylglutaryl-CoA; HSP, heat shock protein; ox-LDL, oxidized LDL; PI3K, phosphatidylinositol 3-kinase; ROS, reactive oxygen species; PP, pyrophosphate; GG, isoprenoid geranylgeranyl.

A recent study has shown that 3′-untranslated region complexes (3′-UTR) of eNOS mRNA, which consist of a UC-rich region and a AU-rich region, regulate the stability of eNOS mRNA by multiple cytosolic proteins binding to the 3′-UTR of eNOS mRNA (42). For example, cytosolic proteins, which bind to the eNOS mRNA 3′-UTR after TNF-α stimulation, lead to the reduction of eNOS mRNA half-life in human endothelial cells (2, 125). Because the RhoA/ROCK pathway and the actin cytoskeleton also regulate eNOS mRNA half-life (88, 110), it is likely that cytosolic proteins, which bind to the 3′-UTR eNOS mRNA, are actin cytoskeleton-associated proteins.

The RhoA/ROCK pathway may also be important in regulating eNOS activity. The activity of eNOS is dependent on intracellular Ca2+ concentration (11). There is increasing evidence that eNOS activity is also partly dependent on eNOS-interacting proteins and posttranslational modification of eNOS (11, 20). For example, eNOS activity could be regulated in part through association with caveolin-1 (35), porin (107), dynamin (12), G protein-coupled receptors (43), heat shock protein 90 (91), and various protein kinases (14, 23, 30, 34). In addition, the regulation of eNOS activity can occur via eNOS phosphorylation. The phosphorylation of Ser1177 of eNOS leads to the rapid activation of eNOS by fluid shear stress, insulin, estrogen, bradykinin, and VEGF (9, 23, 30, 47, 54). Interestingly, inhibition of RhoA or ROCKs leads to the rapid activation of PI3K/Akt and phosphorylation of eNOS (71, 124), suggesting the potential role of ROCKs in regulating eNOS activation in addition to eNOS expression. Although the phosphorylation of Ser1177 leads to increased eNOS activity, the phosphorylation of Thr495 is associated with a decrease in eNOS activity (30). In addition, although other phosphorylation sites have been identified on eNOS proteins, such as Ser114, Ser633, and other tyrosine residues, the precise functional roles of these phosphorylation sites remain to be determined (29, 108).

ROCKs IN CARDIOVASCULAR DISEASE

Selective ROCK inhibitors such as fasudil have been shown to prevent cerebral vasospasm after subarachnoid hemorrhage. Furthermore, another ROCK inhibitor, Y-27632, has been shown to inhibit the development of atherosclerosis and arterial remodeling after vascular injury (65, 94). We recently showed (89) that ROCK activity is involved in the expression of plasminogen activator inhibitor-1 (PAI-1) mediated by hyperglycemia, indicating that the RhoA/ROCK pathway may function as a key regulator of cardiovascular injury in patients with diabetes mellitus. Thus there is increasing evidence that inhibition of the RhoA/ROCK pathway may be beneficial in cardiovascular diseases and could be a potential therapeutic target (Fig. 1). Specifically, the RhoA/ROCK pathway has been shown to be involved in angiogenesis (40, 51), atherosclerosis (65), cerebral and coronary vasospasm (45, 92), cerebral ischemia (112), erectile dysfunction (8, 17), glomerulosclerosis (78), hypertension (114), myocardial hypertrophy (38), myocardial ischemia-reperfusion injury (7, 124), neointima formation (70, 94), pulmonary hypertension (1), and vascular remodeling (72). In many of these studies, the underlying protective mechanism of ROCK inhibition was mediated by the upregulation of eNOS. However, in other studies, the protective effects of ROCK inhibition were mediated by the acute activation of eNOS via the PI3K/Akt pathway (124). Moreover, ROCKs may also play a role in Alzheimer disease (74), bronchial asthma (16), cancers (101), demyelinating diseases (74), glaucoma (39), and osteoporosis (82). However, it remains to be determined how ROCKs are involved in these disease states.

CLINICAL IMPLICATIONS OF ROCKs

Inhibition of ROCKs by fasudil leads to beneficial effects in patients with systemic hypertension (66), pulmonary hypertension (33), vasospastic angina (67), stable effort angina (99), stroke (97), and chronic heart failure (50). Indeed, perhaps many of the so-called pleiotropic effects of statins are mediated by ROCK inhibition (5558, 110, 124). Thus assessing ROCK activity in patients undergoing statin therapy may provide greater insights into the mechanism underlying statin nonresponders in statin clinical trials. Furthermore, the development of ROCK inhibitors could be useful therapeutically in patients without elevated cholesterol levels or in cardiovascular conditions such as ischemic stroke and heart failure in which elevated serum cholesterol level is not a risk factor.

In conclusion, there is growing evidence that the RhoA/ROCK pathway plays an important pathophysiological role in cardiovascular diseases and that inhibition of ROCKs by ROCK inhibitors or statins may be beneficial. To date, a great number of cellular and physiological functions are mediated by ROCKs, and ROCK activity is often elevated in disorders of the cardiovascular system. Thus inhibition of ROCKs may be a potential therapeutic target in reducing cardiovascular disease. However, a greater understanding of the physiological role of each ROCK isoform in the cardiovascular system and the development of isoform-specific inhibitors are needed to resolve the specificity and safety of ROCK inhibitors.

Acknowledgments

GRANTS

This work was supported by National Institutes of Health Grants HL-52233 and NS-10828 and the American Heart Association Bugher Foundation Award. K. Noma and N. Oyama are recipients of the Japan Heart Foundation/Bayer-Yakuhin Research Grant Abroad.

REFERENCES

  • 1.Abe K, Shimokawa H, Morikawa K, Uwatoku T, Oi K, Matsumoto Y, Hattori T, Nakashima Y, Kaibuchi K, Sueishi K, Takeshita A. Long-term treatment with a Rho-kinase inhibitor improves monocrotaline-induced fatal pulmonary hypertension in rats. Circ Res. 2004;94:385–393. doi: 10.1161/01.RES.0000111804.34509.94. [DOI] [PubMed] [Google Scholar]
  • 2.Alonso J, Sanchez de Miguel L, Monton M, Casado S, Lopez-Farre A. Endothelial cytosolic proteins bind to the 3′ untranslated region of endothelial nitric oxide synthase mRNA: regulation by tumor necrosis factor alpha. Mol Cell Biol. 1997;17:5719–5726. doi: 10.1128/mcb.17.10.5719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Amano M, Chihara K, Kimura K, Fukata Y, Nakamura N, Matsuura Y, Kaibuchi K. Formation of actin stress fibers and focal adhesions enhanced by Rho-kinase. Science. 1997;275:1308–1311. doi: 10.1126/science.275.5304.1308. [DOI] [PubMed] [Google Scholar]
  • 4.Amano M, Chihara K, Nakamura N, Kaneko T, Matsuura Y, Kaibuchi K. The COOH terminus of Rho-kinase negatively regulates rho-kinase activity. J Biol Chem. 1999;274:32418–32424. doi: 10.1074/jbc.274.45.32418. [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.Arnal JF, Yamin J, Dockery S, Harrison DG. Regulation of endothelial nitric oxide synthase mRNA, protein, and activity during cell growth. Am J Physiol Cell Physiol. 1994;267:C1381–C1388. doi: 10.1152/ajpcell.1994.267.5.C1381. [DOI] [PubMed] [Google Scholar]
  • 7.Bao W, Hu E, Tao L, Boyce R, Mirabile R, Thudium DT, Ma XL, Willette RN, Yue TL. Inhibition of Rho-kinase protects the heart against ischemia/reperfusion injury. Cardiovasc Res. 2004;61:548–558. doi: 10.1016/j.cardiores.2003.12.004. [DOI] [PubMed] [Google Scholar]
  • 8.Bivalacqua TJ, Champion HC, Usta MF, Cellek S, Chitaley K, Webb RC, Lewis RL, Mills TM, Hellstrom WJ, Kadowitz PJ. RhoA/Rho-kinase suppresses endothelial nitric oxide synthase in the penis: a mechanism for diabetes-associated erectile dysfunction. Proc Natl Acad Sci USA. 2004;101:9121–9126. doi: 10.1073/pnas.0400520101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Boo YC, Sorescu G, Boyd N, Shiojima I, Walsh K, Du J, Jo H. Shear stress stimulates phosphorylation of endothelial nitric-oxide synthase at Ser1179 by Akt-independent mechanisms: role of protein kinase A. J Biol Chem. 2002;277:3388–3396. doi: 10.1074/jbc.M108789200. [DOI] [PubMed] [Google Scholar]
  • 10.Burridge K, Wennerberg K. Rho and Rac take center stage. Cell. 2004;116:167–179. doi: 10.1016/s0092-8674(04)00003-0. [DOI] [PubMed] [Google Scholar]
  • 11.Busse R, Mulsch A. Calcium-dependent nitric oxide synthesis in endothelial cytosol is mediated by calmodulin. FEBS Lett. 1990;265:133–136. doi: 10.1016/0014-5793(90)80902-u. [DOI] [PubMed] [Google Scholar]
  • 12.Cao S, Yao J, McCabe TJ, Yao Q, Katusic ZS, Sessa WC, Shah V. Direct interaction between endothelial nitric-oxide synthase and dynamin-2. Implications for nitric-oxide synthase function. J Biol Chem. 2001;276:14249–14256. doi: 10.1074/jbc.M006258200. [DOI] [PubMed] [Google Scholar]
  • 13.Chen XQ, Tan I, Ng CH, Hall C, Lim L, Leung T. Characterization of RhoA-binding kinase ROKα implication of the pleckstrin homology domain in ROKα function using region-specific antibodies. J Biol Chem. 2002;277:12680–12688. doi: 10.1074/jbc.M109839200. [DOI] [PubMed] [Google Scholar]
  • 14.Chen ZP, Mitchelhill KI, Michell BJ, Stapleton D, Rodriguez-Crespo I, Witters LA, Power DA, Ortiz de Montellano PR, Kemp BE. AMP-activated protein kinase phosphorylation of endothelial NO synthase. FEBS Lett. 1999;443:285–289. doi: 10.1016/s0014-5793(98)01705-0. [DOI] [PubMed] [Google Scholar]
  • 15.Chevrier V, Piel M, Collomb N, Saoudi Y, Frank R, Paintrand M, Narumiya S, Bornens M, Job D. The Rho-associated protein kinase p160ROCK is required for centrosome positioning. J Cell Biol. 2002;157:807–817. doi: 10.1083/jcb.200203034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Chiba Y, Misawa M. The role of RhoA-mediated Ca2+ sensitization of bronchial smooth muscle contraction in airway hyperresponsiveness. J Smooth Muscle Res. 2004;40:155–167. doi: 10.1540/jsmr.40.155. [DOI] [PubMed] [Google Scholar]
  • 17.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]
  • 18.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]
  • 19.Davis ME, Grumbach IM, Fukai T, Cutchins A, Harrison DG. Shear stress regulates endothelial nitric-oxide synthase promoter activity through nuclear factor κB binding. J Biol Chem. 2004;279:163–168. doi: 10.1074/jbc.M307528200. [DOI] [PubMed] [Google Scholar]
  • 20.Dedio J, Konig P, Wohlfart P, Schroeder C, Kummer W, Muller-Esterl W. NOSIP, a novel modulator of endothelial nitric oxide synthase activity. FASEB J. 2001;15:79–89. doi: 10.1096/fj.00-0078com. [DOI] [PubMed] [Google Scholar]
  • 21.Denker SP, Huang DC, Orlowski J, Furthmayr H, Barber DL. Direct binding of the Na-H exchanger NHE1 to ERM proteins regulates the cortical cytoskeleton and cell shape independently of H+ translocation. Mol Cell. 2000;6:1425–1436. doi: 10.1016/s1097-2765(00)00139-8. [DOI] [PubMed] [Google Scholar]
  • 22.Di Cunto F, Imarisio S, Hirsch E, Broccoli V, Bulfone A, Migheli A, Atzori C, Turco E, Triolo R, Dotto GP, Silengo L, Altruda F. Defective neurogenesis in citron kinase knockout mice by altered cytokinesis and massive apoptosis. Neuron. 2000;28:115–127. doi: 10.1016/s0896-6273(00)00090-8. [DOI] [PubMed] [Google Scholar]
  • 23.Dimmeler S, Fleming I, Fisslthaler B, Hermann C, Busse R, Zeiher AM. Activation of nitric oxide synthase in endothelial cells by Akt-dependent phosphorylation. Nature. 1999;399:601–605. doi: 10.1038/21224. [DOI] [PubMed] [Google Scholar]
  • 24.Etienne-Manneville S, Hall A. Rho GTPases in cell biology. Nature. 2002;420:629–635. doi: 10.1038/nature01148. [DOI] [PubMed] [Google Scholar]
  • 25.Eto M, Barandier C, Rathgeb L, Kozai T, Joch H, Yang Z, Luscher TF. Thrombin suppresses endothelial nitric oxide synthase and upregulates endothelin-converting enzyme-1 expression by distinct pathways: role of Rho/ROCK and mitogen-activated protein kinase. Circ Res. 2001;89:583–590. doi: 10.1161/hh1901.097084. [DOI] [PubMed] [Google Scholar]
  • 26.Farah S, Agazie Y, Ohan N, Ngsee JK, Liu XJ. A rho-associated protein kinase, ROKα binds insulin receptor substrate-1 and modulates insulin signaling. J Biol Chem. 1998;273:4740–4746. doi: 10.1074/jbc.273.8.4740. [DOI] [PubMed] [Google Scholar]
  • 27.Feng J, Ito M, Ichikawa K, Isaka N, Nishikawa M, Hartshorne DJ, Nakano T. Inhibitory phosphorylation site for Rho-associated kinase on smooth muscle myosin phosphatase. J Biol Chem. 1999;274:37385–37390. doi: 10.1074/jbc.274.52.37385. [DOI] [PubMed] [Google Scholar]
  • 28.Feng J, Ito M, Kureishi Y, Ichikawa K, Amano M, Isaka N, Okawa K, Iwamatsu A, Kaibuchi K, Hartshorne DJ, Nakano T. Rho-associated kinase of chicken gizzard smooth muscle. J Biol Chem. 1999;274:3744–3752. doi: 10.1074/jbc.274.6.3744. [DOI] [PubMed] [Google Scholar]
  • 29.Fleming I, Busse R. Molecular mechanisms involed in the regulation of the endothelial nitric oxide synthase. Am J Physiol Regul Integr Comp Physiol. 2003;284:R1–R12. doi: 10.1152/ajpregu.00323.2002. [DOI] [PubMed] [Google Scholar]
  • 30.Fleming I, Fisslthaler B, Dimmeler S, Kemp BE, Busse R. Phosphorylation of Thr495 regulates Ca2+/calmodulin-dependent endothelial nitric oxide synthase activity. Circ Res. 2001;88:E68–E75. doi: 10.1161/hh1101.092677. [DOI] [PubMed] [Google Scholar]
  • 31.Flowers MA, Wang Y, Stewart RJ, Patel B, Marsden PA. Reciprocal regulation of endothelin-1 and endothelial constitutive NOS in proliferating endothelial cells. Am J Physiol Heart Circ Physiol. 1995;269:H1988–H1997. doi: 10.1152/ajpheart.1995.269.6.H1988. [DOI] [PubMed] [Google Scholar]
  • 32.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]
  • 33.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]
  • 34.Fulton D, Gratton JP, McCabe TJ, Fontana J, Fujio Y, Walsh K, Franke TF, Papapetropoulos A, Sessa WC. Regulation of endothelium-derived nitric oxide production by the protein kinase Akt. Nature. 1999;399:597–601. doi: 10.1038/21218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Garcia-Cardena G, Martasek P, Masters BS, Skidd PM, Couet J, Li S, Lisanti MP, Sessa WC. Dissecting the interaction between nitric oxide synthase (NOS) and caveolin. Functional significance of the NOS caveolin binding domain in vivo. J Biol Chem. 1997;272:25437–25440. doi: 10.1074/jbc.272.41.25437. [DOI] [PubMed] [Google Scholar]
  • 36.Hall A. Rho GTPases and the actin cytoskeleton. Science. 1998;279:509–514. doi: 10.1126/science.279.5350.509. [DOI] [PubMed] [Google Scholar]
  • 37.Hart MJ, Jiang X, Kozasa T, Roscoe W, Singer WD, Gilman AG, Sternweis PC, Bollag G. Direct stimulation of the guanine nucleotide exchange activity of p115 RhoGEF by Gα13. Science. 1998;280:2112–2114. doi: 10.1126/science.280.5372.2112. [DOI] [PubMed] [Google Scholar]
  • 38.Higashi M, Shimokawa H, Hattori T, Hiroki J, Mukai Y, Morikawa K, Ichiki T, Takahashi S, Takeshita A. Long-term inhibition of Rho-kinase suppresses angiotensin II-induced cardiovascular hypertrophy in rats in vivo: effect on endothelial NAD(P)H oxidase system. Circ Res. 2003;93:767–775. doi: 10.1161/01.RES.0000096650.91688.28. [DOI] [PubMed] [Google Scholar]
  • 39.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]
  • 40.Hyvelin JM, Howell K, Nichol A, Costello CM, Preston RJ, McLoughlin P. Inhibition of Rho-kinase attenuates hypoxia-induced angiogenesis in the pulmonary circulation. Circ Res. 2005;97:185–191. doi: 10.1161/01.RES.0000174287.17953.83. [DOI] [PubMed] [Google Scholar]
  • 41.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]
  • 42.Jimenez A, Arriero MM, Lopez-Blaya A, Gonzalez-Fernandez F, Garcia R, Fortes J, Millas I, Velasco S, Sanchez De Miguel L, Rico L, Farre J, Casado S, Lopez-Farre A. Regulation of endothelial nitric oxide synthase expression in the vascular wall and in mononuclear cells from hypercholesterolemic rabbits. Circulation. 2001;104:1822–1830. doi: 10.1161/hc3901.095769. [DOI] [PubMed] [Google Scholar]
  • 43.Ju H, Venema VJ, Marrero MB, Venema RC. Inhibitory interactions of the bradykinin B2 receptor with endothelial nitric-oxide synthase. J Biol Chem. 1998;273:24025–24029. doi: 10.1074/jbc.273.37.24025. [DOI] [PubMed] [Google Scholar]
  • 44.Kaneko T, Amano M, Maeda A, Goto H, Takahashi K, Ito M, Kaibuchi K. Identification of calponin as a novel substrate of Rhokinase. Biochem Biophys Res Commun. 2000;273:110–116. doi: 10.1006/bbrc.2000.2901. [DOI] [PubMed] [Google Scholar]
  • 45.Katsumata N, Shimokawa H, Seto M, Kozai T, Yamawaki T, Kuwata K, Egashira K, Ikegaki I, Asano T, Sasaki Y, Takeshita A. Enhanced myosin light chain phosphorylations as a central mechanism for coronary artery spasm in a swine model with interleukin-1β. Circulation. 1997;96:4357–4363. doi: 10.1161/01.cir.96.12.4357. [DOI] [PubMed] [Google Scholar]
  • 46.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]
  • 47.Kim F, Gallis B, Corson MA. TNF-α inhibits flow and insulin signaling leading to NO production in aortic endothelial cells. Am J Physiol Cell Physiol. 2001;280:C1057–C1065. doi: 10.1152/ajpcell.2001.280.5.C1057. [DOI] [PubMed] [Google Scholar]
  • 48.Kimura K, Fukata Y, Matsuoka Y, Bennett V, Matsuura Y, Okawa K, Iwamatsu A, Kaibuchi K. Regulation of the association of adducin with actin filaments by Rho-associated kinase (Rho-kinase) and myosin phosphatase. J Biol Chem. 1998;273:5542–5548. doi: 10.1074/jbc.273.10.5542. [DOI] [PubMed] [Google Scholar]
  • 49.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]
  • 50.Kishi T, Hirooka Y, Masumoto A, Ito K, Kimura Y, Inokuchi K, Tagawa T, Shimokawa H, Takeshita A, Sunagawa K. Rho-kinase inhibitor improves increased vascular resistance and impaired vasodilation of the forearm in patients with heart failure. Circulation. 2005;111:2741–2747. doi: 10.1161/CIRCULATIONAHA.104.510248. [DOI] [PubMed] [Google Scholar]
  • 51.Kishore R, Qin G, Luedemann C, Bord E, Hanley A, Silver M, Gavin M, Goukassain D, Losordo DW. The cytoskeletal protein ezrin regulates EC proliferation and angiogenesis via TNF-α-induced transcriptional repression of cyclin A. J Clin Invest. 2005;115:1785–1796. doi: 10.1172/JCI22849. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Kozasa T, Jiang X, Hart MJ, Sternweis PM, Singer WD, Gilman AG, Bollag G, Sternweis PC. p115 RhoGEF, a GTPase activating protein for Gα12 and Gα13. Science. 1998;280:2109–2111. doi: 10.1126/science.280.5372.2109. [DOI] [PubMed] [Google Scholar]
  • 53.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]
  • 54.Lantin-Hermoso RL, Rosenfeld CR, Yuhanna IS, German Z, Chen Z, Shaul PW. Estrogen acutely stimulates nitric oxide synthase activity in fetal pulmonary artery endothelium. Am J Physiol Lung Cell Mol Physiol. 1997;273:L119–L126. doi: 10.1152/ajplung.1997.273.1.L119. [DOI] [PubMed] [Google Scholar]
  • 55.Laufs U, Endres M, Stagliano N, Amin-Hanjani S, Chui DS, Yang SX, Simoncini T, Yamada M, Rabkin E, Allen PG, Huang PL, Bohm M, Schoen FJ, Moskowitz MA, Liao JK. Neuroprotection mediated by changes in the endothelial actin cytoskeleton. J Clin Invest. 2000;106:15–24. doi: 10.1172/JCI9639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Laufs U, Fata VL, Liao JK. Inhibition of 3-hydroxy-3-methylglutaryl (HMG)-CoA reductase blocks hypoxia-mediated down-regulation of endothelial nitric oxide synthase. J Biol Chem. 1997;272:31725–31729. doi: 10.1074/jbc.272.50.31725. [DOI] [PubMed] [Google Scholar]
  • 57.Laufs U, La Fata V, Plutzky J, Liao JK. Upregulation of endothelial nitric oxide synthase by HMG CoA reductase inhibitors. Circulation. 1998;97:1129–1135. doi: 10.1161/01.cir.97.12.1129. [DOI] [PubMed] [Google Scholar]
  • 58.Laufs U, Liao JK. Post-transcriptional regulation of endothelial nitric oxide synthase mRNA stability by Rho GTPase. J Biol Chem. 1998;273:24266–24271. doi: 10.1074/jbc.273.37.24266. [DOI] [PubMed] [Google Scholar]
  • 59.Leung T, Chen XQ, Manser E, Lim L. The p160 RhoA-binding kinase ROK alpha is a member of a kinase family and is involved in the reorganization of the cytoskeleton. Mol Cell Biol. 1996;16:5313–5327. doi: 10.1128/mcb.16.10.5313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.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]
  • 61.Liao JK, Shin WS, Lee WY, Clark SL. Oxidized low-density lipoprotein decreases the expression of endothelial nitric oxide synthase. J Biol Chem. 1995;270:319–324. doi: 10.1074/jbc.270.1.319. [DOI] [PubMed] [Google Scholar]
  • 62.Liao JK, Zulueta JJ, Yu FS, Peng HB, Cote CG, Hassoun PM. Regulation of bovine endothelial constitutive nitric oxide synthase by oxygen. J Clin Invest. 1995;96:2661–2666. doi: 10.1172/JCI118332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Luscher TF. Imbalance of endothelium-derived relaxing and contracting factors. A new concept in hypertension? Am J Hypertens. 1990;3:317–330. doi: 10.1093/ajh/3.4.317. [DOI] [PubMed] [Google Scholar]
  • 64.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]
  • 65.Mallat Z, Gojova A, Sauzeau V, Brun V, Silvestre JS, Esposito B, Merval R, Groux H, Loirand G, Tedgui A. Rho-associated protein kinase contributes to early atherosclerotic lesion formation in mice. Circ Res. 2003;93:884–888. doi: 10.1161/01.RES.0000099062.55042.9A. [DOI] [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.Masumoto A, Mohri M, Shimokawa H, Urakami L, Usui M, Takeshita A. Suppression of coronary artery spasm by the Rho-kinase inhibitor fasudil in patients with vasospastic angina. Circulation. 2002;105:1545–1547. doi: 10.1161/hc1002.105938. [DOI] [PubMed] [Google Scholar]
  • 68.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]
  • 69.Matsui T, Maeda M, Doi Y, Yonemura S, Amano M, Kaibuchi K, Tsukita S, 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]
  • 70.Matsumoto Y, Uwatoku T, Oi K, Abe K, Hattori T, Morishige K, Eto Y, Fukumoto Y, Nakamura K, Shibata Y, Matsuda T, Takeshita A, Shimokawa H. Long-term inhibition of Rho-kinase suppresses neointimal formation after stent implantation in porcine coronary arteries: involvement of multiple mechanisms. Arterioscler Thromb Vasc Biol. 2004;24:181–186. doi: 10.1161/01.ATV.0000105053.46994.5B. [DOI] [PubMed] [Google Scholar]
  • 71.Ming XF, Viswambharan H, Barandier C, Ruffieux J, Kaibuchi K, Rusconi S, Yang Z. 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;22:8467–8477. doi: 10.1128/MCB.22.24.8467-8477.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Miyata K, Shimokawa H, Kandabashi T, Higo T, Morishige K, Eto Y, Egashira K, Kaibuchi K, Takeshita A. Rho-kinase is involved in macrophage-mediated formation of coronary vascular lesions in pigs in vivo. Arterioscler Thromb Vasc Biol. 2000;20:2351–2358. doi: 10.1161/01.atv.20.11.2351. [DOI] [PubMed] [Google Scholar]
  • 73.Moon SY, Zheng Y. Rho GTPase-activating proteins in cell regulation. Trends Cell Biol. 2003;13:13–22. doi: 10.1016/s0962-8924(02)00004-1. [DOI] [PubMed] [Google Scholar]
  • 74.Mueller BK, Mack H, Teusch N. Rho kinase, a promising drug target for neurological disorders. Nat Rev Drug Discov. 2005;4:387–398. doi: 10.1038/nrd1719. [DOI] [PubMed] [Google Scholar]
  • 75.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]
  • 76.Narumiya S. The small GTPase Rho: cellular functions and signal transduction. J Biochem (Tokyo) 1996;120:215–228. doi: 10.1093/oxfordjournals.jbchem.a021401. [DOI] [PubMed] [Google Scholar]
  • 77.Narumiya S, Ishizaki T, Watanabe N. Rho effectors and reorganization of actin cytoskeleton. FEBS Lett. 1997;410:68–72. doi: 10.1016/s0014-5793(97)00317-7. [DOI] [PubMed] [Google Scholar]
  • 78.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]
  • 79.Noma K, Higashi Y, Jitsuiki D, Hara K, Kimura M, Nakagawa K, Goto C, Oshima T, Yoshizumi M, Chayama K. Smoking activates rho-kinase in smooth muscle cells of forearm vasculature in humans. Hypertension. 2003;41:1102–1105. doi: 10.1161/01.HYP.0000067062.92836.9E. [DOI] [PubMed] [Google Scholar]
  • 80.O’Cochlain DF, Perez-Terzic C, Reyes S, Kane GC, Behfar A, Hodgson DM, Strommen JA, Liu XK, van den Broek W, Wansink DG, Wieringa B, Terzic A. Transgenic overexpression of human DMPK accumulates into hypertrophic cardiomyopathy, myotonic myopathy and hypotension traits of myotonic dystrophy. Hum Mol Genet. 2004;13:2505–2518. doi: 10.1093/hmg/ddh266. [DOI] [PubMed] [Google Scholar]
  • 81.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]
  • 82.Ohnaka K, Shimoda S, Nawata H, Shimokawa H, Kaibuchi K, Iwamoto Y, Takayanagi R. Pitavastatin enhanced BMP-2 and osteocalcin expression by inhibition of Rho-associated kinase in human osteoblasts. Biochem Biophys Res Commun. 2001;287:337–342. doi: 10.1006/bbrc.2001.5597. [DOI] [PubMed] [Google Scholar]
  • 83.Pan J, Singh US, Takahashi T, Oka Y, Palm-Leis A, Herbelin BS, Baker KM. PKC mediates cyclic stretch-induced cardiac hypertrophy through Rho family GTPases and mitogen-activated protein kinases in cardiomyocytes. J Cell Physiol. 2005;202:536–553. doi: 10.1002/jcp.20151. [DOI] [PubMed] [Google Scholar]
  • 84.Rees DD, Palmer RM, Moncada S. Role of endothelium-derived nitric oxide in the regulation of blood pressure. Proc Natl Acad Sci USA. 1989;86:3375–3378. doi: 10.1073/pnas.86.9.3375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Ridley AJ, Hall A. The small GTP-binding protein rho regulates the assembly of focal adhesions and actin stress fibers in response to growth factors. Cell. 1992;70:389–399. doi: 10.1016/0092-8674(92)90163-7. [DOI] [PubMed] [Google Scholar]
  • 86.Ridley AJ, Paterson HF, Johnston CL, Diekmann D, Hall A. The small GTP-binding protein rac regulates growth factor-induced membrane ruffling. Cell. 1992;70:401–410. doi: 10.1016/0092-8674(92)90164-8. [DOI] [PubMed] [Google Scholar]
  • 87.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]
  • 88.Rikitake Y, Kim HH, Huang Z, Seto M, Yano K, Asano T, Moskowitz MA, Liao JK. Inhibition of Rho kinase (ROCK) leads to increased cerebral blood flow and stroke protection. Stroke. 2005;36:2251–2257. doi: 10.1161/01.STR.0000181077.84981.11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Rikitake Y, Liao JK. Rho-kinase mediates hyperglycemia-induced plasminogen activator inhibitor-1 expression in vascular endothelial cells. Circulation. 2005;111:3261–3268. doi: 10.1161/CIRCULATIONAHA.105.534024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.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]
  • 91.Russell KS, Haynes MP, Caulin-Glaser T, Rosneck J, Sessa WC, Bender JR. Estrogen stimulates heat shock protein 90 binding to endothelial nitric oxide synthase in human vascular endothelial cells. Effects on calcium sensitivity and NO release. J Biol Chem. 2000;275:5026–5030. doi: 10.1074/jbc.275.7.5026. [DOI] [PubMed] [Google Scholar]
  • 92.Sato M, Tani E, Fujikawa H, Kaibuchi K. Involvement of Rho-kinase-mediated phosphorylation of myosin light chain in enhancement of cerebral vasospasm. Circ Res. 2000;87:195–200. doi: 10.1161/01.res.87.3.195. [DOI] [PubMed] [Google Scholar]
  • 93.Saura M, Zaragoza C, Cao W, Bao C, Rodriguez-Puyol M, Rodriguez-Puyol D, Lowenstein CJ. Smad2 mediates transforming growth factor-β induction of endothelial nitric oxide synthase expression. Circ Res. 2002;91:806–813. doi: 10.1161/01.res.0000040397.23817.e5. [DOI] [PubMed] [Google Scholar]
  • 94.Sawada N, Itoh H, Ueyama K, Yamashita J, Doi K, Chun TH, Inoue M, Masatsugu K, Saito T, Fukunaga Y, Sakaguchi S, Arai H, Ohno N, Komeda M, Nakao K. Inhibition of rho-associated kinase results in suppression of neointimal formation of balloon-injured arteries. Circulation. 2000;101:2030–2033. doi: 10.1161/01.cir.101.17.2030. [DOI] [PubMed] [Google Scholar]
  • 95.Schmidt A, Hall A. Guanine nucleotide exchange factors for Rho GTPases: turning on the switch. Genes Dev. 2002;16:1587–1609. doi: 10.1101/gad.1003302. [DOI] [PubMed] [Google Scholar]
  • 96.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]
  • 97.Shibuya M, Hirai S, Seto M, Satoh S, Ohtomo E Fasudil Ischemic Stroke Study Group. Effects of fasudil in acute ischemic stroke: results of a prospective placebo-controlled double-blind trial. J Neurol Sci. 2005;238:31–39. doi: 10.1016/j.jns.2005.06.003. [DOI] [PubMed] [Google Scholar]
  • 98.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]
  • 99.Shimokawa H, Hiramori K, Iinuma H, Hosoda S, Kishida H, Osada H, Katagiri T, Yamauchi K, Yui Y, Minamino T, Nakashima M, Kato K. Anti-anginal effect of fasudil, a Rho-kinase inhibitor, in patients with stable effort angina: a multicenter study. J Cardiovasc Pharmacol. 2002;40:751–761. doi: 10.1097/00005344-200211000-00013. [DOI] [PubMed] [Google Scholar]
  • 100.Shimokawa H, Takeshita A. Rho-kinase is an important therapeutic target in cardiovascular medicine. Arterioscler Thromb Vasc Biol. 2005;25:1767–1775. doi: 10.1161/01.ATV.0000176193.83629.c8. [DOI] [PubMed] [Google Scholar]
  • 101.Somlyo AP, Somlyo AV. Ca2+ sensitivity of smooth muscle and nonmuscle myosin II: modulated by G proteins, kinases, and myosin phosphatase. Physiol Rev. 2003;83:1325–1358. doi: 10.1152/physrev.00023.2003. [DOI] [PubMed] [Google Scholar]
  • 102.Somlyo AP, Somlyo AV. Signal transduction and regulation in smooth muscle. Nature. 1994;372:231–236. doi: 10.1038/372231a0. [DOI] [PubMed] [Google Scholar]
  • 103.Somlyo AP, Somlyo AV. Signal transduction by G-proteins, rho-kinase and protein phosphatase to smooth muscle and non-muscle myosin II. J Physiol. 2000;522:177–185. doi: 10.1111/j.1469-7793.2000.t01-2-00177.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Sordella R, Classon M, Hu KQ, Matheson SF, Brouns MR, Fine B, Zhang L, Takami H, Yamada Y, Settleman J. Modulation of CREB activity by the Rho GTPase regulates cell and organism size during mouse embryonic development. Dev Cell. 2002;2:553–565. doi: 10.1016/s1534-5807(02)00162-4. [DOI] [PubMed] [Google Scholar]
  • 105.Sordella R, Jiang W, Chen GC, Curto M, Settleman J. Modulation of Rho GTPase signaling regulates a switch between adipogenesis and myogenesis. Cell. 2003;113:147–158. doi: 10.1016/s0092-8674(03)00271-x. [DOI] [PubMed] [Google Scholar]
  • 106.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]
  • 107.Sun J, Liao JK. Functional interaction of endothelial nitric oxide synthase with a voltage-dependent anion channel. Proc Natl Acad Sci USA. 2002;99:13108–13113. doi: 10.1073/pnas.202260999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Tai SC, Robb GB, Marsden PA. Endothelial nitric oxide synthase: a new paradigm for gene regulation in the injured blood vessel. Arterioscler Thromb Vasc Biol. 2004;24:405–412. doi: 10.1161/01.ATV.0000109171.50229.33. [DOI] [PubMed] [Google Scholar]
  • 109.Takai Y, Sasaki T, Tanaka K, Nakanishi H. Rho as a regulator of the cytoskeleton. Trends Biochem Sci. 1995;20:227–231. doi: 10.1016/s0968-0004(00)89022-2. [DOI] [PubMed] [Google Scholar]
  • 110.Takemoto M, Sun J, Hiroki J, Shimokawa H, Liao JK. Rho-kinase mediates hypoxia-induced downregulation of endothelial nitric oxide synthase. Circulation. 2002;106:57–62. doi: 10.1161/01.cir.0000020682.73694.ab. [DOI] [PubMed] [Google Scholar]
  • 111.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]
  • 112.Toshima Y, Satoh S, Ikegaki I, Asano T. A new model of cerebral microthrombosis in rats and the neuroprotective effect of a Rho-kinase inhibitor. Stroke. 2000;31:2245–2250. doi: 10.1161/01.str.31.9.2245. [DOI] [PubMed] [Google Scholar]
  • 113.Turner MS, Fen Fen L, Trauger JW, Stephens J, LoGrasso P. Characterization and purification of truncated human Rho-kinase II expressed in Sf-21 cells. Arch Biochem Biophys. 2002;405:13–20. doi: 10.1016/s0003-9861(02)00249-7. [DOI] [PubMed] [Google Scholar]
  • 114.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]
  • 115.Vallance P, Collier J, Moncada S. Effects of endothelium-derived nitric oxide on peripheral arteriolar tone in man. Lancet. 1989;2:997–1000. doi: 10.1016/s0140-6736(89)91013-1. [DOI] [PubMed] [Google Scholar]
  • 116.Van Aelst L, D’Souza-Schorey C. Rho GTPases and signaling networks. Genes Dev. 1997;11:2295–2322. doi: 10.1101/gad.11.18.2295. [DOI] [PubMed] [Google Scholar]
  • 117.Vane JR, Anggard EE, Botting RM. Regulatory functions of the vascular endothelium. N Engl J Med. 1990;323:27–36. doi: 10.1056/NEJM199007053230106. [DOI] [PubMed] [Google Scholar]
  • 118.Vanhoutte PM. Endothelium and control of vascular function. State of the Art lecture. Hypertension. 1989;13:658–667. doi: 10.1161/01.hyp.13.6.658. [DOI] [PubMed] [Google Scholar]
  • 119.Velasco G, Armstrong C, Morrice N, Frame S, Cohen P. Phosphorylation of the regulatory subunit of smooth muscle protein phosphatase 1M at Thr850 induces its dissociation from myosin. FEBS Lett. 2002;527:101–104. doi: 10.1016/s0014-5793(02)03175-7. [DOI] [PubMed] [Google Scholar]
  • 120.Ward Y, Yap SF, Ravichandran V, Matsumura F, Ito M, Spinelli B, Kelly K. The GTP binding proteins Gem and Rad are negative regulators of the Rho-Rho kinase pathway. J Cell Biol. 2002;157:291–302. doi: 10.1083/jcb.200111026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Wei L, Roberts W, Wang L, Yamada M, Zhang S, Zhao Z, Rivkees SA, Schwartz RJ, Imanaka-Yoshida K. Rho kinases play an obligatory role in vertebrate embryonic organogenesis. Development. 2001;128:2953–2962. doi: 10.1242/dev.128.15.2953. [DOI] [PubMed] [Google Scholar]
  • 122.Wojciak-Stothard B, Potempa S, Eichholtz T, Ridley AJ. Rho and Rac but not Cdc42 regulate endothelial cell permeability. J Cell Sci. 2001;114:1343–1355. doi: 10.1242/jcs.114.7.1343. [DOI] [PubMed] [Google Scholar]
  • 123.Wojciak-Stothard B, Ridley AJ. Rho GTPases and the regulation of endothelial permeability. Vascul Pharmacol. 2002;39:187–199. doi: 10.1016/s1537-1891(03)00008-9. [DOI] [PubMed] [Google Scholar]
  • 124.Wolfrum S, Dendorfer A, Rikitake Y, Stalker TJ, Gong Y, Scalia R, Dominiak P, Liao JK. Inhibition of Rho-kinase leads to rapid activation of phosphatidylinositol 3-kinase/protein kinase Akt and cardiovascular protection. Arterioscler Thromb Vasc Biol. 2004;24:1842–1847. doi: 10.1161/01.ATV.0000142813.33538.82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Yoshizumi M, Perrella MA, Burnett JC, Jr, Lee ME. Tumor necrosis factor downregulates an endothelial nitric oxide synthase mRNA by shortening its half-life. Circ Res. 1993;73:205–209. doi: 10.1161/01.res.73.1.205. [DOI] [PubMed] [Google Scholar]

RESOURCES