Open access

ROCK Inhibition – A New Therapeutic Avenue in Kidney Protection

Written By

Stefan Reuter, Dominik Kentrup and Eckhart Büssemaker

Submitted: 02 November 2010 Published: 23 August 2011

DOI: 10.5772/19200

From the Edited Volume

Kidney Transplantation - New Perspectives

Edited by Dr.(Md) Magdalena Trzcinska

Chapter metrics overview

2,544 Chapter Downloads

View Full Metrics

1. Introduction

Chronic allograft nephropathy (CAN) remains the main cause of renal transplant loss besides the death of patients with a functioning graft. Its prevention and treatment still lacks any significant breakthrough since many years (Meier-Kriesche et al. 2004; Pascual et al. 2002). Pathological manifestations of CAN include interstitial fibrosis, tubular atrophy, vascular occlusive changes, glomerulosclerosis and a progressive renal dysfunction accompanied by hypertension and proteinuria (Joosten et al. 2005; Racusen et al. 1999). This histopathologic constellation is now more formally and descriptively referred to as interstitial fibrosis and tubular atrophy (IF/TA) without evidence of any specific aetiology (Solez et al. 2007). IF/TA or CAN describe the common final path of different injuries causing renal damage, whereas their precise pathogenesis is complex and only incompletely understood (Joosten et al. 2005). The process encompasses multifactorial aetiologies including alloantigen-dependent as well as alloantigen-independent factors (Gottmann et al. 2003; Joosten et al. 2005; Kerjaschki et al. 2006; Nankivell et al. 2003; Reuter et al. 2010; Tullius & Tilney, 1995). Hence, an effective treatment is not available so far.

Rho effectors Rho-associated, coiled-coil containing protein kinases (ROCK) and their associated signaling pathways have emerged as important players in cardiovascular and renal pathophysiology. Recently, it has been shown that ROCK inhibition is protective in diabetic and obstructive nephropathy, hypertensive nephrosclerosis, ischemia reperfusion injury, and chronic allograft nephropathy (Kanda et al. 2003a; Kentrup D. et al. 10 A.D.; Kentrup D. et al. 2010; Komers et al. 2011a; Komers et al. 2011b; Liu et al. 2009; Nagatoya et al. 2002; Nishikimi et al. 2004a; Satoh et al. 2002; Song et al. 2008; Versteilen et al. 2011).

ROCKs have been initially identified as downstream targets of the small GTP binding protein Rho. Members of the Rho family include Rho (isoforms A–E, and G), Rac (isoforms 1 and 2), Cdc42 and TC10 (Nobes & Hall, 1994). After translocation to the plasma membrane, GTP-RhoA activates its effectors, including the two isoforms of ROCK, ROCK1 (ROKβ, p160ROCK) and ROCK2 (ROKα, Rho kinase) (Nakagawa et al. 1996). Although, ROCK1 and 2 can be differentially regulated under distinct circumstances there is no evidence that ROCK1 and ROCK2 have different functions at present (Nobes & Hall, 1994). ROCKs are protein serine/threonine kinases belonging to the AGC (PKA/PKG/PKC) family (Ishizaki et al. 1996; Leung et al. 1995; Matsui et al. 1996). ROCK activity is involved in actin cytoskeletal organization, stress fiber formation, and cell contraction thereby controlling vascular smooth muscle contraction, endothelial barrier and leukocyte functions (e.g., cellular motility, migration, adhesion and transmigration) (Loirand, Guerin & Pacaud, 2006; Riento & Ridley, 2003). Both isoforms of ROCK can be the target of effective inhibitors such as TAT-C3, HMG-CoA reductase inhibitors, mTOR inhibitors, angiotensin II antagonists, Rad GTPase, dominant-negative ROCK, and the specific ROCK inhibitors fasudil, hydroxyfasudil, and Y-27632 (Liao, Seto & Noma, 2007; Oka et al. 2008). We herein discuss favourable effects of ROCK inhibitors in kidney transplantation-related diseases, and highlight their potential impact on novel therapeutic strategies to improve long-term renal graft survival.

1.1. Rock Inhibition in Diabetic nephropathy

Diabetic nephropathy (DN) is the most common cause of chronic kidney disease (CKD) with a considerable risk of progression to end stage renal disease (ESRD) (Zimmet, Alberti & Shaw, 2001). Thus, it is not surprising that DN is the main cause of patients entering permanent renal replacement programs (dialysis/transplantation) worldwide (Rugg, 2003). When dialysis is initiated, the survival of patients with DN is inferior when compared with other renal diseases. Despite an inferior survival of diabetics, which is primarily due to cardiovascular disease generally present prior to transplantation, kidney transplantation has been established as the renal replacement therapy of choice for these patients (Cosio et al. 2008). Because longer waiting times on dialysis negatively impact post-transplant graft survival (Meier-Kriesche et al. 2000) and successful transplantation was shown to confess a substantial survival benefit to patients with diabetic-ESRD, transplantation should be performed early (Becker et al. 2006; Hirschl, 1996; Son et al. 2010; Wolfe et al. 1999). As diabetes persists (or occurs, e.g. post-transplant diabetes (Rodrigo et al. 2006)) after renal transplantation it contributes to delayed graft function and long-term (re-)graft loss (Arnol et al. 2008; Fellstrom et al. 2005; Khalkhali et al. 2010; Parekh, Bostrom & Feng, 2010; Wilkinson et al. 2005). Interestingly, in a study by Wiesbauer et al. maximal glucose, HbA1c, or diabetes treatment did not influence death-censored functional graft survival but mortality (Wiesbauer et al. 2010). However, there is evidence that DN can re-occur after transplantation and that reversal or stabilization of the course of DN (still difficult to achieve) may slower the progress to ESRD again (Bhalla et al. 2003; Osterby et al. 1991; Salifu et al. 2004; Wojciechowski, Onozato & Gonin, 2009).

What happens in DN? The mechanistic driving force of DN still remains undetermined. Only 30-50% of people with diabetes develop overt nephropathy over a lifetime. This suggests that other factors besides diabetes are required to share in for the progression of DN (Nakagawa et al. 2011; Rugg, 2003). One important parameter for the development of DN identified is glomerular hypertension due to (intra renal) vascular alterations (Johnston et al. 1998). Vascular damage of the glomerular capillaries causes leakage of albumin and other proteins across the filter into the urine. It was postulated that the degree of DN correlates to the amount of urinary albumin excretion. However, others have found that DN is an independent risk factor for the development of microalbuminuria (Chang et al. 2011a). In contrast, progressive renal failure in diabetics can also occur in the absence of proteinuria even though histology present with typical signs of DN (Caramori, Fioretto & Mauer, 2003). Thus, it was questioned whether the decline of renal function is linked to proteinuria or whether both simply appear in parallel (Perkins et al. 2007) (reviewed in (Jefferson, Shankland & Pichler, 2008)). Because classical cardiovascular risk factors like hyperglycemia, hypertension, and hyperlipidemia are well-described to promote DN it would stand to reason that mechanisms causing vascular damage, such as endothelial dysfunction, oxidative stress, advanced glycation end products (AGEs), and angiogenesis, are involved in the pathogenesis of DN (Brownlee, 2001; Chang et al. 2011b; Jansson, 2007). It was suggested that hyperglycemia accelerates the polyol and the hexosamine pathway, activates protein kinase C (PKC), and induces nonenzymatic glycosylation AGEs (Brownlee, 2001; Schena & Gesualdo, 2005). Especially AGEs lead to fibrosis via accumulation of interstitial collagens. AGEs also induce oxidative stress which activates NF-kappa b. NF-kappa b and PKC-activation are associated with the release of (proinflammatory) cytokines and growth factors such as vascular endothelial growth factor (VEGF), tumor necrosis factor a (TNFa), fibroblast growth factor (FGF), tissue factor, transforming growth factor-beta (TGF-ß), Interleukin 1 (IL-1), IL-6 and IL-18 (Brownlee, 2001; Johnston et al. 1998). PKC activation is also related to hemodynamic changes (predominantly through the activation of the renin-angiotensin aldosterone system (RAAS) which in term promotes hypertension, oxidative stress, and fibrosis.

Therefore, current renoprotective treatment strategies for DN are rather classical and include the control of blood glucose, blood pressure, lipids (notably, many cholesterol-independent or "pleiotropic" effects of statins are mediated by ROCK inhibition (Liao, 2007)) and body weight as well as RAAS blockade and physical training (Alicic & Tuttle, 2010; Van Buren & Toto, 2011). The interest emerged on ROCK inhibitors when it was observed that Rho/ROCK play important roles in hypertension/cardiovascular system and in the kidney in models of diabetes (Arita et al. 2009; Gojo et al. 2007; Kawamura et al. 2004; Kolavennu et al. 2008; Miao et al. 2002; Peng et al. 2008; Rikitake & Liao, 2005). Besides hyperglycemia, further factors of the diabetic milieu, such as reactive oxygen species (ROS), oxidized LDL, acceleration of the hexosamine pathway, and AGEs, can activate the Rho/ROCK pathway in vascular and renal cells (Komers, 2011). Interestingly, among other actions AGEs can increase endothelial permeability/hyperpermeability of vessels through the RAGE/Rho signaling pathway which can be inhibited by application of Y-27632 (Hirose et al. 2010). Moreover, Rho/ROCK have been identified as key mediators of VEGF-induced endothelial cell hyperpermeability (Zeng et al. 2005). As mentioned above, VEGF expression is increased in response to hyperglycemia. Because albuminuria, due to glomerular leakage, is a common feature of DN, ROCK activity might be a mechanism involved in renal proteinuria in these patients. In addition, the Rho/ROCK pathway can be activated by hormones or cytokines involved in DN pathophysiology, like AngII, aldosterone, TGF-ß, and VEGF (Komers, 2011). It was stated that this activation of ROCK-dependent pathways, e.g. due to production of osteopontin, plasminogen activator inhibitor 1, or extracellular matrix, is involved in the pathogenesis of DN. Cell culture studies by Peng et al. and Kolavennu et al. supported these findings. They observed that the activity of Rho/ROCK in mesangial cells increased by glucose treatment. This caused reorganization of cytoskeleton and increased production of fibronectin, collagen IV, VEGF and AP-1, a transcription factor promoting the expression of e.g. TGF-ß. Inhibition of the Rho/ROCK pathway saved the cells from these changes (Kolavennu et al. 2008; Peng et al. 2008). Moreover, glomerular hypertension is a well described factor participating in the progression of DN. Hypertension causes mechanical stress which activates Rho (see below). In congruence, Komers et al. described improved renal hemodynamics after ROCK inhibition in diabetic rats (Komers et al. 2011a). In addition, there is some evidence that Rho is involved in the actions of endothelin 1 (ET-1), a potent vasoconstrictor, which is upregulated in diabetics (Yousif, 2006).

Because of this and comparable evidence from other studies it was assumed that ROCK inhibition is advantageous in diabetes. Komers, Peng and Gojo et al. treated diabetic rats (diabetes type 1 model) with the orally administered ROCK-inhibitor fasudil (Gojo et al. 2007; Komers et al. 2011b; Peng et al. 2008). Their diabetic rats rapidly developed albuminuria, glomerulosclerosis, and renal interstitial fibrosis, as well as decreased glomerular filtration rates (GFR), and increased expression of molecular markers of DN. Sustained ROCK inhibition reduced diabetes-related kidney damage (as confirmed by histology and urinalysis) in the kidney and expression of the molecular markers (including markers of epithelial–mesenchymal transition (EMT)) in association with a slight anti-proteinuric effect (including beneficial effects of fasudil on podocyte foot process effacement) which was independent of blood pressure or glucose control. So far, all long-term studies evidenced nephroprotective effects of ROCK inhibitors being independent from systemic blood pressure. In contrast, Kikuchi et al. failed to suppress the progression of nephropathy by fasudil application (but ameliorated some features of DN) in a rat model of type 2 diabetes (Kikuchi et al. 2007) but Kolavennu et al. found ROCK inhibition being kidney protective in type 2 diabetes mice (Kolavennu et al. 2008). Interestingly, Peng observed that fasudil and ACE-inhibitors had a comparable effectiveness preventing DN while Komers et al. noted that the combination of fasudil and losartan was not more effective than losartan alone (Komers et al. 2011b; Peng et al. 2008). ACE-inhibitors and AT1-blockers are well known agents used for kidney protection in (proteinuric) DN for many years. Interestingly, RAAS blockade was effective to inhibit Rho/ROCK activity in several studies suggesting that ROCK activation might follow AT1 activation under certain conditions (Higuchi et al. 2007; Komers et al. 2011b; Ohtsu et al. 2006). Thus, the effects of RAAS blockade partly rely on ROCK inhibition: Angiotensin II (AngII) mediated vasoconstriction (via myosin light chain phosphorylation), proinflammatory effects (via PAI-1 and monocyte chemoattractant protein-1 (MCP-1) induction) and JNK-dependent hypertrophy/cell migration. They can be attributed to Rho activity and are therefore within the therapeutic target range of ROCK inhibitors (Higuchi et al. 2007; Ohtsu et al. 2006). More evidence comes from Rikitake et al. who showed in ROCK1+/- haploinsufficient mice that perivascular fibrosis induced by AngII was significantly lower than in wild type individuals (Rikitake et al. 2005b). In the context of diabetes it is of note that AngII-dependent activation of the Rho/ROCK pathway is partly mediated by NADPH oxidase-dependent ROS (Jin et al. 2006). As mentioned above, ROS are present in the hyperglycemic milieu in excess probably promoting AngII effects in diabetics.

1.2. Rock Inhibition in Urethral obstruction

Urethral obstruction (UO) is a typical cause of ESRD in children. In this patient group, UO is responsible for approximately 15 to 25% of kidney failures (Koo et al. 1999). This is even a problem after renal transplantation, because obstruction-related side effects influence the success of transplantation. Allograft function and survival are often limited due to urinary tract infection or surgical complications, implying deleterious actions of chronic elevated intravesical pressure (Cairns et al. 1991; Churchill et al. 1988; Sheldon et al. 1994). However, more recent studies claim that patients with severe lower urinary tract abnormalities and ESRD may receive a kidney transplant with comparable safeness and success to patients without abnormalities (Broniszczak et al. 2010; Nahas & David-Neto, 2009; Rigamonti et al. 2005).

In experimental settings, UO is commonly employed as a normotensive, non-proteinuric and non-hyperlipidemic model of tubulointerstitial fibrosis without any toxic renal insult (Nagatoya et al. 2002). Fibrosis is of major interest, not only because it is a common final path of different injuries causing renal damage, but, as a substantial part of IF/TA, also related to the renal (graft) prognosis (Couser & Johnson, 1994; Meier-Kriesche et al. 2004; Morgan et al. 2011; Nath, 1992; Pascual et al. 2002). Depending on the side where it mainly occurs, fibrosis is classified as glomerulosclerosis or tubulointerstitial fibrosis. Nevertheless, in most situations fibrosis is detected in both compartments, hinting towards a not yet fully understood cross-talk in between (Klein et al. 2011). Briefly, examples of fibrosis initiating factors are: increased intravascular pressure (hypertension), increased pressure in the tubular lumen (due to obstruction of the urinary tract), hyperglycemia (diabetes), increased urinary albumin concentrations (proteinuria), and toxic substances (puromycin or adriamycin). After ignition of the process due to different stimuli the pathways converge independently of the activator. Signaling includes induction of cytokines and chemokines, like TGF-β, platelet-derived growth factor (PDGF), FGF, ET-1, and osteopontin, leading to renal inflammation (predominant cell types are monocytes and macrophage (Mphi)) and cell activation.

It was suggested that Mphi are central players in the fibroproliferative response (Vernon, Mylonas & Hughes, 2010). They release profibrogenic growth factors i.e., TGF-β, PDGF and FGF, which activate/recruit myofibroblasts and stimulate resident interstitial fibroblasts. These fibroblasts are the main source of interstitial ECM (e.g., collagens and fibronectin, and expression of factors interfering with ECM crosslinking (transglutaminases, high glucose-induced AGEs) and/or turnover (matrix metalloproteinases, plasminogen activator, and their inhibitors) (well summarized in (Klein et al. 2011)) that cause fibrosis. In congruence, we and others observed e.g., that the level of L-Plastin, a cytoskeletal protein mainly expressed in interstitial fibroblasts, is increased in IF/TA (Reuter et al. 2010). Recently, it was reported that Rho/ROCK are central to differentiation, migration, and contractility of myofibroblasts (Mack et al. 2001; Parizi, Howard & Tomasek, 2000). Thus, ROCK inhibition might suppress the destructive activity of myofibroblasts. If not applied, the fibrotic process continues. Blocking of Mphi recruitment and activation ameliorates renal inflammation and fibrosis (Vielhauer et al. 2010). As ROCK is essential for Mphi migration its blockade can protect from fibrosis in UO (Nagatoya et al. 2002; Satoh et al. 2002). In renal graft recipients TGF-ß expression was analyzed in protocol biopsies (performed 3, 6, and 12 months after kidney transplantation). An increased level of TGF-ß was associated with a larger extent of interstitial fibrosis (Baboolal et al. 2002). Interestingly, when ROCK is inhibited in vivo the tissue level of the fibrogenic TGF-ß is lower than in control kidneys (Nagatoya et al. 2002).

Besides, TGF-ß promotes EMT, a process related to the development of renal fibrosis (Zavadil & Bottinger, 2005). As mentioned before, Rho/ROCK are important players in the (re-)organisation of the cytoskeleton. This implies that they are key mediators/effectors of epithelial plasticity and EMT induced by TGF-ß (Patel et al. 2005; Rodrigues-Diez et al. 2008; Zavadil & Bottinger, 2005). In summary, kidney protection due to ROCK inhibition can be achieved in UO because Rho/ROCK is involved in different key mechanisms in renal fibrosis.

1.3. Rock Inhibition in Hypertensive Nephrosclerosis

Increased blood pressure is an important health problem and a major risk factor for cardiovascular morbidity and mortality throughout the world (Staessen et al. 2003). To date, both the pathogenesis of arterial hypertension and the molecular mechanisms involved in blood pressure control remain poorly understood. Hypertension is characterized by high arterial pressure resulting from increased peripheral vascular resistance that can be attributed to both enhanced contractility of vascular smooth muscle cells and arterial wall remodeling. Increased activity of the Rho/ROCK pathway has been proposed to play an important role in the development and maintenance of hypertension. In various animal models of experimental hypertension a role of RhoA and ROCK has been demonstrated. Blocking ROCK activity with Y-27632 has blood pressure lowering effects in spontaneously hypertensive rats (SHR), deoxycorticosterone-acetate (DOCA)/salt-treated and renal hypertensive rats (Uehata et al. 1997). Similarly, oral administration of fasudil to SHR rats significantly lowered blood pressure (Mukai et al. 2001). In addition, several studies have addressed changes in RhoA activity in isolated vascular segments from hypertensive animals suggesting that increased RhoA activity is responsible for enhanced ROCK function in the pathology of hypertension. In mesenteric and cerebral arteries from SHR and normotensive rats the relaxation induced by treatment with Y-27632 was markedly higher in arteries from SHR rats (Asano & Nomura, 2003; Chrissobolis & Sobey, 2001). This has also been shown for mesenteric arteries from DOCA/salt-treated rats (Weber & Webb, 2001). In animals treated with L-NAME, an inhibitor of NO-synthase, oral administration of Y-27632 lowered blood pressure and the level of active, GTP-bound RhoA was markedly increased in vessels from L-NAME treated rats (Weber & Webb, 2001). Similarly, direct evidence for increased amounts of active RhoA have been found in stroke-prone SHR, DOCA/salt- and renal hypertensive rats (Moriki et al. 2004; Seasholtz et al. 2001; Seko et al. 2003). Analysis of the expression levels of RhoA has led to controversial results with some studies showing an increased expression of RhoA under hypertensive conditions (Seasholtz et al. 2001) whereas in other reports no differences in the expression profile of proteins from the RhoA/ROCK pathway have been detected (Seko et al. 2003).

Impaired endothelial function and decreased NO-production have been implicated in the etiology of hypertension. Decreased expression of endothelial nitric oxide synthase (eNOS) is found in aortae from SHR rats (Chou et al. 1998) and eNOS-deficient mice have an elevated blood pressure (Huang et al. 1995). Interestingly, there seems to be extensive crosstalk between NO and RhoA/ROCK-signaling. There is compelling evidence that the NO/cGK pathway leads to an inhibition of RhoA/ROCK signaling (Carter, Begaye & Kanagy, 2002; Chitaley & Webb, 2002; Sauzeau et al. 2000). On the other hand, the RhoA/ROCK cascade seems to reciprocally influence NO-signaling. The mechanism by which ROCK influences NO production seems to be the regulation of eNOS mRNA stability (Eto et al. 2001; Rikitake et al. 2005a; Takemoto et al. 2002).

Chronic hypertension leads to end organ damage in a substantial number of patients. Hypertensive nephrosclerosis is a disorder that is usually associated with chronic hypertension. Histologically it is characterized by vascular, glomerular, and tubulointerstitial changes. The vascular pathology consists of intimal thickening and luminal narrowing of the large and small renal arteries and the glomerular arterioles. Two different processes appear to contribute to the development of the vascular lesions: A hypertrophic response to chronic hypertension evident by medial hypertrophy and fibroblastic intimal thickening, resulting in narrowing of the vascular lumen (Zucchelli & Zuccala, 1994). In the beginning this process is adaptive by minimizing the degree to which the rise in systemic pressure is transmitted to the downstream arterioles and capillaries (Zucchelli & Zuccala, 1994). The other process contributing to the vascular pathology is the deposition of hyaline-like material (plasma protein constituents, such as inactive C3b, part of the third component of complement) into the damaged, more permeable arteriolar wall (Zucchelli & Zuccala, 1994). Arterial hypertension may lead to focal global (involving the whole glomerulus) or focal segemental glomerulosclerosis (Marcantoni et al. 2002; Zucchelli & Zuccala, 1994). Global sclerosis is thought to reflect ischemic injury, leading to nephron loss (Marcantoni et al. 2002). Focal segmental sclerosis is typically associated with glomerular enlargement, which is probably a compensatory response to nephron loss (Harvey et al. 1992). The vascular and glomerular alterations are associated with an often severe interstitial nephritis. Its etiology is incompletely understood. At least in part immunologic processes may be involved. They are probably started by ischemia-induced alterations in antigen expression on the surface of the tubular epithelial cells (Truong et al. 1992). Nephrosclerosis is seen with aging but is clearly exacerbated in arterial hypertension (Lindeman, Tobin & Shock, 1984; Rule et al. 2010). The incidence of progressive renal disease in hypertensive nephrosclerosis is low, however, three groups of patients are at increased risk to develop progressive kidney function deterioration: patients with more marked elevations in blood pressure, afro-american patients (they have an approximate eight-fold elevation in the risk of hypertension-induced ESRD(Toto, 2003); this increase in risk may persist even with "adequate" blood pressure control) and patients with underlying chronic kidney disease, especially diabetics. Patients with nephrosclerosis typically present with a long history of hypertension. If present, decline in kidney function is slow in progression as indicated by serum-creatinine and blood-urea-nitrogen. Urinalysis is typically benign without appearance of cast or dsymorph erythrocytes. Urinary protein excretion is typically mildly elevated (less than 1 gram per day). Concerning the incidence of renal failure in hypertensive nephrosclerosis there seems to be a clinical paradox as among patients (especially considering afro-americans) entering the chronic hemodialysis program. Hypertensive nephrosclerosis is one of the most common diagnoses, whereas the risk for a hypertensive patient to develop ESRD is rather small. However, at least three large trials might explain this paradox: the number of hypertensive patients is so large that even a small percentage of patients at risk gives a large number; the rate of progression might be so slow that trials that mostly run over 5-7 years might not detect patients at risk (Freedman, Iskandar & Appel, 1995; Madhavan et al. 1995). Recent work has shed some light on the importance of the Rho/ROCK pathway in kidney disease. ROCK is constitutionally active in the renal circulation. Studies on glomerular hemodynamics demonstrated that ROCK inhibition by Y-27632 and fasudil dilates the basal tone of afferent and efferent arterioles (Cavarape et al. 2003; Nakamura et al. 2003). Importantly, both inihibitors reverse angiotensin II-induced vasoconstriction of efferent and afferent arterioles (Nakamura et al. 2003). Thus, ROCK inhibition might protect against deleterious hemodynamic effects in kidney disease. In addition, to its critical role for the renal microvasculature the Rho/ROCK pathway is an important regulator of several cell function including proliferation, migration and apoptosis as already stated above (diabetic nephropathy). Of importance for the development of glomerulosclerosis, as a key feature of hypertensive nephrosclerosis, it has been demonstrated that Rho regulates the formation of stress fibers, focal adhesions and peripheral bundles through reorganization of the actin cytoskeleton in a renal epithelial cell line (Nakano et al. 1999). Renal epithelial cells are able to transform to mesenchymal-like cells via EMT. These changes have been observed in renal tubulointerstitial fibrosis, another hallmark of hypertensive nephrosclerosis. Mesangial cells reside in the renal glomeruli and produce ECM. Its increased accumulation causes glomerulosclerosis, where TGF-ß has been shown as a causative factor (Nakano et al. 1999). As mentioned above Rho/ROCK are key mediators/effectors of epithelial plasticity and EMT induced by TGF-ß (Patel et al. 2005; Rodrigues-Diez et al. 2008; Zavadil & Bottinger, 2005). Finally, in mesangial cells, mechanical stress, which is considered to cause glomerular hypertension and glomerulosclerosis, enhances mitogen-activated protein kinase (MAP kinase) activity, stress fiber formation, and cellular proliferation (Bruijn et al. 1994). In this process of disease the Rho/ROCK pathway plays a pivotal role, acting as a modulator of MAP kinase and the downstream cellular impact. Taken together, these data strengthen an important role of the Rho/ROCK pathway in the development of glomerulosclerotic disease.

Podocytes are highly differentiated cells that are located in the renal glomerulus. Cytoskeleton rearrangement is closely associated with podocyte shape changes and dysfunction in various renal diseases (Zucchelli & Zuccala, 1994). Using the ROCK-inhibitor Y-27632 Endlich et al. could inhibit the reorganization of the cytoskeleton induced by mechanical stress in podocytes. Moreover, inhibition of ROCK prevents TGF-ß-induced increase in CTGF accumulation in fibroblast cells (Heusinger-Ribeiro et al. 2001). All these observation strongly suggest a pivotal role of the Rho/ROCK pathway in the progression of renal injury. To date, the body of evidence given by in vivo studies is growing. As the Rho/ROCK pathway regulates glomerular hemodynamics and has profound effects on mesangial cell proliferation and matrix production, ROCK-inhibitors are candidates to serve as therapeutic tools to treat glomerulosclerotic disease. Thus, it has been demonstrated that Y-27632 and fasudil prevent tubulointerstitial fibrosis in a model of unilateral ureteral obstruction [for detail see section on ureteral obstruction]. In 5/6 nephrectomized spontaneously hypertensive rats (SHR), a model of hypertensive glomerulosclerosis, the Rho/ROCK pathway was activated. Treatment with fasudil reduced urinary protein excretion, improved glomerular and tubulointerstitial injury score, and reduced the infiltration of ED-1 positive cells and proliferating cell nuclear antigen positive cells in the kidney of SHR treated by 5/6 nephrectomy. Interestingly, these effects were obtained without lowering blood pressure, indicating blood pressure-independent effects of ROCK (Kanda et al. 2003b). Fasudil up-regulated the expression of p27kip1, a cyclin-dependent kinase inhibitor, and increased the p27kip1 immunopositive cells in both glomeruli and tubulointerstitium, indicating inhibition of cell proliferation and macrophage recruitment under fasudil treatment. Another group reported similar beneficial effects in Dahl salt-sensitive rats. In these animals, fasudil improved renal function, proteinuria, and histological findings without changes in blood pressure (Nishikimi et al. 2004b). These beneficial effects were most likely accompanied by decreased expression of TGF-ß, collagen-I, and collagen-III mRNA in the renal cortex. In salt-loaded spontaneously hypertensive stroke-prone rats serving as a model of severe hypertension fasudil improved kidney function, proteinuria, histological findings and decreased expression of genes encoding for extracellular matrix, oxidative stress, adhesion molecules and antifibrinolysis. Of note, these effects were independent of the blood pressure-lowering activity of fasudil (Nishikimi et al. 2004b). As stated above there is compelling evidence for an extensive crosstalk between NO and RhoA/ROCK-signaling. Indeed, in SHR fasudil partly reversed the progressive nephrosclerosis initiated by administration of the nitric oxide-synthase inhibitor nitro-L-arginine methyl ester (Koshikawa et al. 2008).

Although multiple factors contribute to the development and progression of chronic renal disease, the renin-angiotensin-aldosterone system seems to be of major importance. Angiotensin II is considered to be the main mediator of this system. It is a potent vasoconstrictor acting directly on vascular smooth muscle cells, thereby regulating the vascular tone. Besides it alters renal sodium and water absorption by stimulating synthesis and secretion of aldosterone. Further, it is involved in the generation of thirst and the excretion of vasopressin. Hence, it has a pivotal role in acute and chronic regulation of blood pressure. Studies in chronic renal disease have shown that angII contributes to deterioration of renal function even if blood pressure is unaltered (Anderson, Rennke & Brenner, 1986). In this view it is important to keep in mind that angII activates ROCK in vascular smooth muscle cells (Yamakawa et al. 2000). In rats infused with angII, treatment with Y-27632 reduced renal inflammatory cell infiltration and tubular damage. AngII activated nuclear factor-kappaB and initiated overexpression of proinflammatory factors, including TNF-alpha and monocyte chemotactic protein-1 (MCP-1), and of CTGF. Treatment of angII-infused rats with Y-27632 reduced the upregulation of these proinflammatory and profibrotic mediators (Ruperez et al. 2005).

Taken together these studies imply that blockade of the Rho/ROCK pathway might prove beneficial in hypertensive nephrosclerosis.

1.4. Rock Inhibition in Ischemia Reperfusion Injury

Renal ischemia-reperfusion (IR) injury (IRI) is a common and important trigger of acute renal injury (AKI). It occurs in a broad spectrum of clinical settings including (transplantation) surgery, trauma, dehydration or sepsis leading to renal hypoperfusion, acute tubular necrosis (ATN), and functional disturbances - namely AKI. Inevitably linked to renal transplantation it is a well known risk factor for delayed graft function associated with prolonged hospitalization, elevated costs, and increased complexity of immunosuppressive drug management. Moreover, by reducing the overall number of nephrons and increasing the risk of acute rejection episodes, IRI might cause a significantly reduced graft survival. Involving both, the innate and the adaptive immune response, causing subsequent sterile inflammation, IRI is composed of a complex cascade of events including the generation of reactive oxygen and nitrogen species, chemotaxis, and phagocytosis. All of which are functional properties of the key effectors of the inflammatory cascade, neutrophiles, the most abundant leukocyte population in circulation, which accumulate as early as 30 minutes after IR particularly in the peritubular capillary network of the outer medulla (Li et al. 2008). Attracted leukocytes subsequently transmigrate into the interstitium. This is associated with increased vascular permeability and loss of endothelial and tubular epithelial cell integrity (Awad et al. 2009) due to degranulation of neutrophils. Upon activation these neutrophils release proteases, myeloperoxidase, cytokines, and generate reactive species leading to aggravation of injury and damage of endothelial and epithelial cells especially in the outer medulla (Bolisetty & Agarwal, 2009; Jang & Rabb, 2009; Li & Okusa, 2006; Okusa et al. 2000). In regard to this, it has recently been shown that ROCK) and their associated signaling pathways play pivotal roles in the development of (experimental) IRI. ROCK-inhibitors such as fasudil or Y-27632 have been shown to provide beneficial effects concerning different ischemic events such as renal (Kentrup D. et al. 10 A.D.; Kentrup D. et al. 2010; Prakash et al. 2008; Teraishi et al. 2004; Versteilen et al. 2006; Versteilen et al. 2011) myocardial (Bao et al. 2004; Hamid, Bower & Baxter, 2007; Wolfrum et al. 2004), cerebral (Satoh et al. 2008; Toshima et al. 2000), hepatic (Du & Hannon, 2004; Ikeda et al. 2003; Takeda et al. 2003), and gastrointestinal (Santen et al. 2010) ischemia. However, the exact mechanisms involved remain to be fully elucidated.

In the context of IRI it seems to be of interest that ROCKs are involved in the regulation of leukocyte cellular motility, migration, adhesion, and transmigration (Alblas et al. 2001; Honing et al. 2004; Lee et al. 2004; Samaniego et al. 2007; Takesono et al. 2010; Vemula et al. 2010; Worthylake et al. 2001; Worthylake & Burridge, 2003) Regarding this, Teraishi et al. were the first group to test the effectiveness of ROCK-inhibitors in an animal model of renal IRI. For this, male Sprague-Dawley rats underwent unilateral nephrectomy of the right kidney two weeks before inducing 45 min of warm ischemia in the remaining kidney by clamping the left renal artery and vein. Y-27632 was hereby applied 5 min pre ischemia or 5 min post ischemia. They observed a protective effect for both treatments (i.e. improved renal function, less histological damage) which was based according to them on reduced infiltration by neutrophils as shown by myeloperoxidase assays. However, even though the latter is a non specific detection method and the data regarding the cell types typically involved varies, (e.g. due to the models used (Rabb et al. 2003; Thornton et al. 1989) or due to non specific detection methods, e.g. myeloperoxidase, naphthol chloroacetate esterase, or HIS-48 staining (Ysebaert et al. 2000)), it is well known that the increased influx of neutrophiles, T- and B-lymphocytes as well as macrophages/monocytes significantly contributes to the pathogenesis of AKI (Kinsey, Li & Okusa, 2008). This first study is also in congruence with later work by Versteilen et al.. By pre-treating male Wistar rats with Y-27632 they could show that leukocyte accumulation (60-70% neutrophils) was markedly reduced by ROCK-inhibitor treatment in the microvasculature of the corticomedullary junction and medulla (Versteilen et al. 2011). They hypothesize that this may be partly due to NO-mediated effects via activated endothelial cells (i.e. limited expression of adhesion molecules and cytokines leading to attenuation of leukocyte accumulation), eNOS mediated alterations of the renal blood flow (Versteilen et al. 2006) and direct effects on the leukocytes.

Further, Prakash et al. also applied the ROCK-inhibitor Y-27632 in a rat model of renal ischemia, but used a Y-27632-lysozyme conjugate (Prakash et al. 2008). Thus, they tried to guarantee a renal-specific uptake into proximal tubular cells via megalin receptors. In unison with the aforementioned data they describe substantially attenuated tubular damage as indicated by reduced expression of dedifferentiation markers kidney injury molecule 1 (KIM-1) and vimentin. Additionally, they observed reduced fibrosis and inflammation as determined by reduced gene expressions of MCP-1, procollagen Iα1, TGF-β1, tissue inhibitor of metalloproteinase 1 and α-smooth muscle actin, as well as reduced immunohistochemical staining of macrophage infiltration, α-smooth muscle actin, collagen I, collagen III and fibronection compared to untreated animals. However, in contrast to the previous data, they describe adverse systemic effects (e.g. leucopenia) when animals were treated with the unconjugated Y-27632, as performed by others groups. An effect, which cannot easily be explained due to the fact that the beneficial effects of ROCK-inhibitors have not only been shown for the kidney but for other organs as well.

Although and apart from the above described effects, it has recently been shown by Kroening et al. that, in some cases, ROCK inhibition may not adversely influence the migratory capabilities of specific cell types as it has been repeatedly shown. Instead, at least in the case of tubular epithelial cells the migratory capacity may actually be promoted and thus ROCK inhibition may favor repair processes in renal tubules (Kroening et al. 2010). The beneficial effects of ROCK inhibition are further supported by work published by Unbekandt et al. who could show that transient ROCK inhibition allows cells from disaggregated embryonic kidneys to form ureteric bud and nephron epithelia (Unbekandt & Davies, 2010). Moreover, one last positive effect of ROCK inhibition related to the prevention of IRI mentioned might be that ROCK inhibition promotes cell survival in human stem cells (Watanabe et al. 2007) and is able to reduce apoptosis in conventional embryonic kidney culture (Meyer et al. 2006).

1.5. Rock Inhibition in Chronic Allograft Nephropathy

Compared to hemodialysis kidney transplantation significantly reduces mortality when it is applied to the appropriate patient (Wolfe et al. 1999). However, even after renal transplantation mortality of transplant recipients is still increased when compared to the general population. This is mainly due to death from cardiovascular disease with a functioning graft as well as immunological and non-immunological factors resulting in graft loss. Despite introduction of new immunosuppressive drugs within the last two decades a substantial increase in graft survival was not achieved. Consequently prevention of graft loss and treatment still lack any significant breakthrough since many years (Gjertson, 1991; Lamb, Lodhi & Meier-Kriesche, 2011; Meier-Kriesche et al. 2004; Meier-Kriesche, Schold & Kaplan, 2004). The pathological manifestations seen in chronic graft loss were summarized as chronic allograft nephropathy. In the recent Banff-classification (“Banff `05”) which gives a classification of the different pathological features seen in chronic graft loss, the term chronic allograft nephropathy does no longer occur (Solez et al. 2007). The rationale for this update of the Banff schema was the misusage of CAN as a generic term for all causes of chronic renal allograft dysfunction with fibrosis that inhibits the accurate diagnosis and appropriate therapy. Thus, the authors of the Banff-classification aimed to present a pathological classification that specifies the underlying disease to facilitate causal treatment. Pathological manifestations of chronic allograft injury include interstitial fibrosis, tubular atrophy, vascular occlusive changes, glomerulosclerosis and a progressive renal dysfunction accompanied by hypertension and proteinuria (Cornell & Colvin, 2005). This histopathologic constellation is now more formally and descriptively referred to as interstitial fibrosis and tubular atrophy (IF/TA) without evidence of any specific aetiology (Solez et al. 2007). IF/TA or CAN describe the common final path of different injuries causing renal damage, whereas their precise pathogenesis is complex and only incompletely understood. The process encompasses multifactorial aetiologies including alloantigen-dependent as well as alloantigen-independent factors (Cornell & Colvin, 2005). The latter mainly comprise arterial hypertension, chronic obstruction and calcineurin-inhibitor toxicity (Busauschina, Schnuelle & van der Woude, 2004; Klahr & Morrissey, 2003; Mihatsch, Ryffel & Gudat, 1995; Morozumi et al. 2004). Besides, chronic polyoma virus nephrotixicity can lead to IF/TA (Drachenberg et al. 2005). Recurrent and de novo glomerular or vascular diseases can also lead to glomerulosclerosis and IF/TA, both early and late post-transplant. It is important to mention that de novo diabetic changes are becoming more common in allografts. As mentioned chronic alloimmune injury is an important cause of IF/TA in kidney grafts. Data on alloantibodies and C4d, a product of the complement cascade in chronically failing grafts, hint at a pathogenic role for humoral immunity in chronic allograft injury. In a prospective study de novo appearance of donor-specific HLA-antibodies was associated with increased graft failure at one year (Terasaki & Ozawa, 2004). These data are consistent with the importance of immunological factors in chronic graft injury.

The Rho/ROCK pathway is potentially crucial in mediating immunological as well as non-immunological injury in chronic graft loss as it is involved in adhesion, migration, proliferation, and cytokine release (Amano et al. 1997; Chihara et al. 1997; Tharaux et al. 2003). The activation of T-cells is involved in alloimmune responses. Here the Rho/ROCK pathway plays a pivotal role in T-cell activation during cellular immune responses by promoting structural rearrangements that are critical for T-cell signaling (Tharaux et al. 2003). Besides, a role for Rho/ROCK in HLA class I signaling pathways that have been implicated in the process of chronic rejection has been shown. Ligation of HLA class I molecules by anti-HLA-antibodies resulted in activation of Rho/ROCK and increased stress fiber formation. Inhibitors of Rho-GTPase and ROCK blocked HLA class I-mediated posphorylation of paxillin and FAK, both central elements of the focal adhesion signaling complex (Lepin et al. 2004). As stated above not only immunological factors but also non-immunological factors, such as arterial hypertension, are responsible for chronic graft injury. Recent data also show relevance of the Rho/ROCK pathway for the regulation of hemodynamics and arterial hypertension. In 1997 it was shown that the Rho/ROCK pathway is involved in the generation of arterial hypertension in different animal models of hypertension (Uehata et al. 1997). Interestingly, these data were confirmed in hypertensive patients showing involvement of Rho/ROCK in the generation of the increased vascular tone in these patients (Masumoto et al. 2001). As already stated above there is a close interaction between the RAAS and the Rho/ROCK pathway. Activation of the RAAS in arterial hypertension and vascular disease is associated with increased activation of Rho/ROCK (Higashi et al. 2003; Yamakawa et al. 2000). Taken together accumulating data suggest a role of the Rho/ROCK pathway in chronic allograft dysfunction. Recent studies in animal models of allograft dysfunction strongly support this view. In a well established model of kidney transplantation Lewis rats acted as kidney graft recipients and Fisher rats as donors. Cyclosporine A was given for immunosuppression. One group of animals additionally received the specific ROCK-inhibitor Y-27632. Renal function deteriorated progressively in the group that received cyclosporine A alone and histology revealed the typical features of IF/TA. ROCK inhibition by Y-27632 significantly prevented the deterioration of kidney function, reduced proteinuria and preserved renal function. These effects were accompanied by down-regulation of the expression of tubular MCP-1, RANTES, and phosphorylated NF-kB. The profibrotic TGF-ß1 and α-SMA, a marker of EMT, were downregulated by the treatment with Y-27632. These data indicated that the Rho/ROCK pathway is critically involved in renal interstitial inflammation and fibrosis, thus efficiently retarding the development of chronic allograft failure (Liu et al. 2009). The same group reported that atorvastatin, an inhibitor of the 3-hydroxy-3-methylglutaryl-coenzyme A reductase, exerted protective effects in CAN (Zhang et al. 2007). Because 3-hydroxy-3-methylglutaryl-coenzyme A reductase interferes with isoprenylation and activation of Rho, these data support the view that inhibition of the Rho/ROCK pathway could be attractive in prevention of allograft nephropathy. Of note, these data confirmed what was seen earlier in a model of cardiac allograft vasculopathy in mice. Here, coronary remodeling in the allografts characterized by intimal thickening and perivascular fibrosis was dose-dependently suppressed by the ROCK-inhibitor fasudil. These data were strengthened as gene transfer of dominant-negative ROCK mimicked the effects of fasudil. Vascular inflammation and expression of profibrotic mediators were significantly reduced in this model (Hattori et al. 2004). Song et al. observed expression of RhoA and ROCK1 mRNA and protein in measangial and tubular cells in the Fisher-to-Lewis model of CAN. Interestingly, they found a negative correlation between RhoA/ROCK1 mRNA and the Banff score. MMF, a potent immunosuppressive drug used in solid organ transplantation attenuated CAN by downregulating the expression of RhoA/ROCK1 (Song et al. 2008).

The data available suggest an important role of the Rho/ROCK pathway in chronic allograft dysfunction. Further studies also in humans are needed to support this view and possibly add a new therapeutic strategy to the nephrologist´s therapeutic options in preserving graft function.

Advertisement

2. Conclusion

Recent experimental data provide promising data that kidney protection in diabetic nephropathy, urethral obstruction, hypertensive nephrosclerosis and chronic allograft nephropathy can be brought about by inhibition of the Rho/ROCK pathway. As reviewed above ROCK activity is involved in actin cytoskeletal organization, stress fiber formation, and cell contraction thereby controlling vascular smooth muscle contraction, endothelial barrier and leukocyte functions (e.g., cellular motility, migration, adhesion and transmigration). All of these aspects are important in the pathogenesis of the renal diseases discussed here. However, clinical trials are needed to develop future strategies and transfer new treatment options by ROCK-inhibition from bench to bedside.

References

  1. 1. Alblas, J., Ulfman, L., Hordijk, P. & Koenderman, L. (2001). Activation of Rhoa and ROCK are essential for detachment of migrating leukocytes. Mol.Biol.Cell 7 12 2137 2145
  2. 2. Alicic R. Z. Tuttle K. R. 2010Management of the diabetic patient with advanced chronic kidney disease. Semin.Dial. 23 2 140 147
  3. 3. Amano M. Chihara K. Kimura K. Fukata Y. Nakamura N. Matsuura Y. Kaibuchi K. 1997Formation of actin stress fibers and focal adhesions enhanced by Rho-kinase. Science 275 5304 1308 1311
  4. 4. Anderson, S., Rennke, H.G. & Brenner, B.M. (1986). Therapeutic advantage of converting enzyme inhibitors in arresting progressive renal disease associated with systemic hypertension in the rat. J.Clin.Invest 6 77 1993 2000
  5. 5. Arita R. Hata Y. Nakao S. Kita T. Miura M. Kawahara S. Zandi S. Almulki L. Tayyari F. Shimokawa H. Hafezi-Moghadam A. Ishibashi T. 2009Rho kinase inhibition by fasudil ameliorates diabetes-induced microvascular damage. Diabetes 58 1 215 226
  6. 6. Arnol M. Prather J. C. Mittalhenkle A. Barry J. M. Norman D. J. 2008Long-term kidney regraft survival from deceased donors: risk factors and outcomes in a single center. Transplantation 86 8 1084 1089
  7. 7. Asano M. Nomura Y. 2003Comparison of inhibitory effects of Y-27632, a Rho kinase inhibitor, in strips of small and large mesenteric arteries from spontaneously hypertensive and normotensive Wistar-Kyoto rats. Hypertens.Res. 26 1 97 106
  8. 8. Awad A. S. Rouse M. Huang L. Vergis A. L. Reutershan J. Cathro H. P. Linden J. Okusa M. D. 2009Compartmentalization of neutrophils in the kidney and lung following acute ischemic kidney injury. Kidney Int. 75 7 689 698
  9. 9. Baboolal K. Jones G. A. Janezic A. Griffiths D. R. Jurewicz W. A. 2002Molecular and structural consequences of early renal allograft injury. Kidney Int. 61 2 686 696
  10. 10. Bao W. Hu E. Tao L. Boyce R. Mirabile R. Thudium D. T. Ma X. L. Willette R. N. Yue T. L. 2004Inhibition of Rho-kinase protects the heart against ischemia/reperfusion injury. Cardiovasc.Res. 61 3 548 558
  11. 11. Becker B. N. Rush S. H. Dykstra D. M. Becker Y. T. Port F. K. 2006Preemptive transplantation for patients with diabetes-related kidney disease. Arch.Intern.Med. 166 1 44 48
  12. 12. Bhalla V. Nast C. C. Stollenwerk N. Tran S. Barba L. Kamil E. S. Danovitch G. Adler S. G. 2003Recurrent and de novo diabetic nephropathy in renal allografts. Transplantation 75 1 66 71
  13. 13. Bolisetty S. Agarwal A. 2009Neutrophils in acute kidney injury: not neutral any more. Kidney Int. 75 7 674 676
  14. 14. Broniszczak D. Ismail H. Nachulewicz P. Szymczak M. Drewniak T. Markiewicz-Kijewska M. Kowalski A. Jobs K. Smirska E. Rubik J. Skobejko-Wlodarska L. Gastol P. Mikolajczyk A. Kalicinski P. 2010Kidney transplantation in children with bladder augmentation or ileal conduit diversion. Eur.J.Pediatr.Surg. 20 1 5 10
  15. 15. Brownlee M. 2001Biochemistry and molecular cell biology of diabetic complications. Nature 414 6865 813 820
  16. 16. Bruijn J. A. Roos A. de G. B. de H. E. 1994Transforming growth factor-beta and the glomerular extracellular matrix in renal pathology. J.Lab Clin.Med. 123 1 34 47
  17. 17. Busauschina A. Schnuelle P. van der Woude F. J. 2004Cyclosporine nephrotoxicity. Transplant Proc. 36(2 Suppl):229S-233S.
  18. 18. Cairns H. S. Leaker B. Woodhouse C. R. Rudge C. J. Neild G. H. 1991Renal transplantation into abnormal lower urinary tract. Lancet 338 8779 1376 1379
  19. 19. Caramori M. L. Fioretto P. Mauer M. 2003Low glomerular filtration rate in normoalbuminuric type 1 diabetic patients: an indicator of more advanced glomerular lesions. Diabetes 52 4 1036 1040
  20. 20. Carter R. W. Begaye M. Kanagy N. L. 2002Acute and chronic NOS inhibition enhances alpha(2)- adrenoreceptor-stimulated RhoA and Rho kinase in rat aorta. Am.J.Physiol Heart Circ.Physiol 283(4):H1361H1369.
  21. 21. Cavarape A. Endlich N. Assaloni R. Bartoli E. Steinhausen M. Parekh N. Endlich K. 2003Rho-kinase inhibition blunts renal vasoconstriction induced by distinct signaling pathways in vivo. J.Am.Soc.Nephrol. 14 1 37 45
  22. 22. Chang T. I. Park J. T. Kim J. K. Kim S. J. Oh H. J. Yoo D. E. Han S. H. Yoo T. H. Kang S. W. 2011aRenal outcomes in patients with type 2 diabetes with or without coexisting non-diabetic renal disease. Diabetes Res.Clin.Pract.
  23. 23. Chang T. I. Park J. T. Kim J. K. Kim S. J. Oh H. J. Yoo D. E. Han S. H. Yoo T. H. Kang S. W. 2011bRenal outcomes in patients with type 2 diabetes with or without coexisting non-diabetic renal disease. Diabetes Res.Clin.Pract.
  24. 24. Chihara K. Amano M. Nakamura N. Yano T. Shibata M. Tokui T. Ichikawa H. Ikebe R. Ikebe M. Kaibuchi K. 1997Cytoskeletal rearrangements and transcriptional activation of c-fos serum response element by Rho-kinase. J.Biol.Chem. 272 40 25121 25127
  25. 25. Chitaley K. Webb R. C. 2002Nitric oxide induces dilation of rat aorta via inhibition of rho-kinase signaling. Hypertension 39(2 Pt 2):438 EOF 42 EOF
  26. 26. Chou T. C. Yen M. H. Li C. Y. Ding Y. A. 1998Alterations of nitric oxide synthase expression with aging and hypertension in rats. Hypertension 31 2 643 648
  27. 27. Chrissobolis S. Sobey C. G. 2001Evidence that Rho-kinase activity contributes to cerebral vascular tone in vivo and is enhanced during chronic hypertension: comparison with protein kinase C. Circ.Res. 88 8 774 779
  28. 28. Churchill B. M. Sheldon C. A. Mc Lorie G. A. Arbus G. S. 1988Factors influencing patient and graft survival in 300 cadaveric pediatric renal transplants. J.Urol. 140(5 Pt 2):1129 EOF 33 EOF
  29. 29. Cornell L. D. Colvin R. B. 2005Chronic allograft nephropathy. Curr.Opin.Nephrol.Hypertens. 14 3 229 234
  30. 30. Cosio F. G. Hickson L. J. Griffin M. D. Stegall M. D. Kudva Y. 2008Patient survival and cardiovascular risk after kidney transplantation: the challenge of diabetes. Am.J.Transplant. 8 3 593 599
  31. 31. Couser W. G. Johnson R. J. 1994Mechanisms of progressive renal disease in glomerulonephritis. Am.J.Kidney Dis. 23 2 193 198
  32. 32. Drachenberg C. B. Hirsch H. H. Ramos E. Papadimitriou J. C. 2005Polyomavirus disease in renal transplantation: review of pathological findings and diagnostic methods. Hum.Pathol. 36 12 1245 1255
  33. 33. Du J. Hannon G. J. 2004Suppression of 160ROCKbypasses cell cycle arrest after Aurora-A/STK15 depletion. Proc.Natl.Acad.Sci.U.S.A 101(24):8975-8980.
  34. 34. Eto M. Barandier C. Rathgeb L. Kozai T. Joch H. Yang Z. Luscher T. F. 2001Thrombin 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. 89 7 583 590
  35. 35. Fellstrom B. Holdaas H. Jardine A. G. Nyberg G. Gronhagen-Riska C. Madsen S. Neumayer H. H. Cole E. Maes B. Ambuhl P. Olsson A. G. Staffler B. Pedersen T. R. 2005Risk factors for reaching renal endpoints in the assessment of Lescol in renal transplantation (ALERT) trial. Transplantation 79 2 205 212
  36. 36. Freedman B. I. Iskandar S. S. Appel R. G. 1995The link between hypertension and nephrosclerosis. Am.J.Kidney Dis. 25 2 207 221
  37. 37. Gjertson D. W. 1991Survival trends in long-term first cadaver-donor kidney transplants. Clin.Transpl. 225 235
  38. 38. Gojo A. Utsunomiya K. Taniguchi K. Yokota T. Ishizawa S. Kanazawa Y. Kurata H. Tajima N. 2007The Rho-kinase inhibitor, fasudil, attenuates diabetic nephropathy in streptozotocin-induced diabetic rats. Eur.J.Pharmacol. 568(1-3):242 EOF 247 EOF
  39. 39. Gottmann U. Oltersdorf J. Schaub M. Knoll T. Back W. E. van der Woude F. J. Braun C. 2003Oxidative stress in chronic renal allograft nephropathy in rats: effects of long-term treatment with carvedilol, BM 91.0228, or alpha-tocopherol. J.Cardiovasc.Pharmacol. 42 3 442 450
  40. 40. Hamid S. A. Bower H. S. Baxter G. F. 2007Rho kinase activation plays a major role as a mediator of irreversible injury in reperfused myocardium. Am.J.Physiol Heart Circ.Physiol 292(6):H2598H2606.
  41. 41. Harvey J. M. Howie A. J. Lee S. J. Newbold K. M. Adu D. Michael J. Beevers D. G. 1992Renal biopsy findings in hypertensive patients with proteinuria. Lancet 340 8833 1435 1436
  42. 42. Hattori T. Shimokawa H. Higashi M. Hiroki J. Mukai Y. Kaibuchi K. Takeshita A. 2004Long-term treatment with a specific Rho-kinase inhibitor suppresses cardiac allograft vasculopathy in mice. Circ.Res. 94 1 46 52
  43. 43. Heusinger-Ribeiro, J., Eberlein, M., Wahab, N.A. & Goppelt-Struebe, M. (2001). Expression of connective tissue growth factor in human renal fibroblasts: regulatory roles of RhoA and cAMP. J.Am.Soc.Nephrol. 9 12 1853 1861
  44. 44. Higashi M. Shimokawa H. Hattori T. Hiroki J. Mukai Y. Morikawa K. Ichiki T. Takahashi S. Takeshita A. 2003Long-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. 93 8 767 775
  45. 45. Higuchi S. Ohtsu H. Suzuki H. Shirai H. Frank G. D. Eguchi S. 2007Angiotensin II signal transduction through the AT1 receptor: novel insights into mechanisms and pathophysiology. Clin.Sci. (Lond) 112 8 417 428
  46. 46. Hirose A. Tanikawa T. Mori H. Okada Y. Tanaka Y. 2010Advanced glycation end products increase endothelial permeability through the RAGE/Rho signaling pathway. FEBS Lett. 584 1 61 66
  47. 47. Hirschl M. M. 1996Type II diabetes mellitus and chronic renal insufficiency: renal transplantation or haemodialysis treatment? Nephrol.Dial.Transplant. 11 Suppl 9 98 99
  48. 48. Honing H. van den Berg. T. K. van der Pol S. M. Dijkstra C. D. van der Kammen R. A. Collard J. G. de Vries H. E. 2004RhoA activation promotes transendothelial migration of monocytes via ROCK. J.Leukoc.Biol. 75 3 523 528
  49. 49. Huang P. L. Huang Z. Mashimo H. Bloch K. D. Moskowitz M. A. Bevan J. A. Fishman M. C. 1995Hypertension in mice lacking the gene for endothelial nitric oxide synthase. Nature 377 6546 239 242
  50. 50. Ikeda F. Terajima H. Shimahara Y. Kondo T. Yamaoka Y. 2003Reduction of hepatic ischemia/reperfusion-induced injury by a specific ROCK/Rho kinase inhibitor Y-27632. J.Surg.Res. 109 2 155 160
  51. 51. Ishizaki, T., Maekawa, M., Fujisawa, K., Okawa, K., Iwamatsu, A., Fujita, A., Watanabe, N., Saito, Y., Kakizuka, A., Morii, N. & Narumiya, S. (1996). The small GTP-binding protein Rho binds to and activates a 160 kDa Ser/Thr protein kinase homologous to myotonic dystrophy kinase. EMBO J. 8 15 1885 1893
  52. 52. Jang H. R. Rabb H. 2009The innate immune response in ischemic acute kidney injury. Clin.Immunol. 130 1 41 50
  53. 53. Jansson P. A. 2007Endothelial dysfunction in insulin resistance and type 2 diabetes. J.Intern.Med. 262 2 173 183
  54. 54. Jefferson J. A. Shankland S. J. Pichler R. H. 2008Proteinuria in diabetic kidney disease: a mechanistic viewpoint. Kidney Int. 74 1 22 36
  55. 55. Jin L. Ying Z. Hilgers R. H. Yin J. Zhao X. Imig J. D. Webb R. C. 2006Increased RhoA/Rho-kinase signaling mediates spontaneous tone in aorta from angiotensin II-induced hypertensive rats. J.Pharmacol.Exp.Ther. 318 1 288 295
  56. 56. Johnston C. I. Risvanis J. Naitoh M. Tikkanen I. 1998Mechanism of progression of renal disease: current hemodynamic concepts. J.Hypertens.Suppl 16(4):S3S7.
  57. 57. Joosten S. A. Sijpkens Y. W. van K. C. Paul L. C. 2005Chronic renal allograft rejection: pathophysiologic considerations. Kidney Int. 68 1 1 13
  58. 58. Kanda, T., Wakino, S., Hayashi, K., Homma, K., Ozawa, Y. & Saruta, T. (2003a). Effect of fasudil on Rho-kinase and nephropathy in subtotally nephrectomized spontaneously hypertensive rats. Kidney Int. 6 64 2009 2019
  59. 59. Kanda, T., Wakino, S., Hayashi, K., Homma, K., Ozawa, Y. & Saruta, T. (2003b). Effect of fasudil on Rho-kinase and nephropathy in subtotally nephrectomized spontaneously hypertensive rats. Kidney Int. 6 64 2009 2019
  60. 60. Kawamura H. Yokote K. Asaumi S. Kobayashi K. Fujimoto M. Maezawa Y. Saito Y. Mori S. 2004High glucose-induced upregulation of osteopontin is mediated via Rho/Rho kinase pathway in cultured rat aortic smooth muscle cells. Arterioscler.Thromb.Vasc.Biol. 24 2 276 281
  61. 61. Kentrup D. Reuter S. Schnöckel U. Edemir B. Grabner A. Pavenstädt H. Schlatter E. Büssemaker E. 2010Alterations in gene expression profile associated with renal ischemia-reperfusion injury are attenuated by hydroxyfasudil mediated Rho-kinase inhibition in a rat model of acute renal failure. Transpl Int 23, 50(Abstract)
  62. 62. Kentrup D. Reuter S. Schnöckel U. Klokkers J. Edemir B. Pavenstädt H. Schäfers M. Schlatter E. Büssemaker E. (. A. D. -Inhibition R. O. C. K. by Hydroxyfasudil. Significantly Improves. Kidney Function. Attenuates-Inflammatory Pro. Infiltration in. a. Rat Model. of Acute. Renal Ischemia. Am J. Am.J.Transplant. 10, 429(Abstract)
  63. 63. Kerjaschki D. Huttary N. Raab I. Regele H. Bojarski-Nagy K. Bartel G. Krober S. M. Greinix H. Rosenmaier A. Karlhofer F. Wick N. Mazal P. R. 2006Lymphatic endothelial progenitor cells contribute to de novo lymphangiogenesis in human renal transplants. Nat.Med. 12 2 230 234
  64. 64. Khalkhali H. R. Ghafari A. Hajizadeh E. Kazemnejad A. 2010Risk factors of long-term graft loss in renal transplant recipients with chronic allograft dysfunction. Exp.Clin.Transplant. 8 4 277 282
  65. 65. Kikuchi Y. Yamada M. Imakiire T. Kushiyama T. Higashi K. Hyodo N. Yamamoto K. Oda T. Suzuki S. Miura S. 2007A Rho-kinase inhibitor, fasudil, prevents development of diabetes and nephropathy in insulin-resistant diabetic rats. J.Endocrinol. 192 3 595 603
  66. 66. Kinsey G. R. Li L. Okusa M. D. 2008Inflammation in acute kidney injury. Nephron Exp.Nephrol. 109(4):e102e107.
  67. 67. Klahr S. Morrissey J. 2003Obstructive nephropathy and renal fibrosis: The role of bone morphogenic protein-7 and hepatocyte growth factor. Kidney Int.Suppl (87):S105S112.
  68. 68. Klein J. Kavvadas P. Prakoura N. Karagianni F. Schanstra J. P. Bascands J. L. Charonis A. 2011Renal fibrosis: Insight from proteomics in animal models and human disease. Proteomics. 11 4 805 815
  69. 69. Kolavennu V. Zeng L. Peng H. Wang Y. Danesh F. R. 2008Targeting of RhoA/ROCK signaling ameliorates progression of diabetic nephropathy independent of glucose control. Diabetes 57 3 714 723
  70. 70. Komers R. 2011Rho kinase inhibition in diabetic nephropathy. Curr. Opin. Nephrol. Hypertens. 20 1 77 83
  71. 71. Komers R. Oyama T. T. Beard D. R. Anderson S. 2011aEffects of systemic inhibition of Rho kinase on blood pressure and renal haemodynamics in diabetic rats. Br.J.Pharmacol. 162 1 163 174
  72. 72. Komers R. Oyama T. T. Beard D. R. Tikellis C. Xu B. Lotspeich D. F. Anderson S. 2011bRho kinase inhibition protects kidneys from diabetic nephropathy without reducing blood pressure. Kidney Int. 79 4 432 442
  73. 73. Koo H. P. Bunchman T. E. Flynn J. T. Punch J. D. Schwartz A. C. Bloom D. A. 1999Renal transplantation in children with severe lower urinary tract dysfunction. J.Urol. 161 1 240 245
  74. 74. Koshikawa, S., Nishikimi, T., Inaba, C., Akimoto, K. & Matsuoka, H. (2008). Fasudil, a Rho-kinase inhibitor, reverses L-NAME exacerbated severe nephrosclerosis in spontaneously hypertensive rats. J.Hypertens. 9 26 1837 1848
  75. 75. Kroening S. Stix J. Keller C. Streiff C. Goppelt-Struebe M. 2010Matrix-independent stimulation of human tubular epithelial cell migration by Rho kinase inhibitors. J.Cell Physiol 223 3 703 712
  76. 76. Lamb K. E. Lodhi S. Meier-Kriesche H. U. 2011Long-term renal allograft survival in the United States: a critical reappraisal. Am.J.Transplant 11 3 450 462
  77. 77. Lee J. H. Katakai T. Hara T. Gonda H. Sugai M. Shimizu A. 2004Roles of p-ERM and Rho-ROCK signaling in lymphocyte polarity and uropod formation. J.Cell Biol. 167 2 327 337
  78. 78. Lepin E. J. Jin Y. P. Barwe S. P. Rozengurt E. Reed E. F. 2004HLA class I signal transduction is dependent on Rho GTPase and ROK. Biochem.Biophys.Res.Commun. 323 1 213 217
  79. 79. Leung T. Manser E. Tan L. Lim L. 1995A novel serine/threonine kinase binding the Ras-related RhoA GTPase which translocates the kinase to peripheral membranes. J.Biol.Chem. 270 49 29051 29054
  80. 80. Li L. Huang L. Sung S. S. Vergis A. L. Rosin D. L. Rose C. E. Jr Lobo P. I. Okusa M. D. 2008The chemokine receptors CCR2 and CX3CR1 mediate monocyte/macrophage trafficking in kidney ischemia-reperfusion injury. Kidney Int. 74 12 1526 1537
  81. 81. Li L. Okusa M. D. 2006Blocking the immune response in ischemic acute kidney injury: the role of adenosine 2A agonists. Nat.Clin.Pract.Nephrol. 2 8 432 444
  82. 82. Liao J. K. 2007Does it matter whether or not a lipid-lowering agent inhibits Rho kinase? Curr.Atheroscler.Rep. 9 5 384 388
  83. 83. Liao J. K. Seto M. Noma K. 2007Rho kinase (ROCK) inhibitors. J.Cardiovasc.Pharmacol. 50 1 17 24
  84. 84. Lindeman R. D. Tobin J. D. Shock N. W. 1984Association between blood pressure and the rate of decline in renal function with age. Kidney Int. 26 6 861 868
  85. 85. Liu M. Gu M. Wu Y. Zhu P. Zhang W. Yin C. Zhang W. J. 2009Therapeutic effect of Y-27632 on chronic allograft nephropathy in rats. J.Surg.Res. 157(1):e117e127.
  86. 86. Loirand G. Guerin P. Pacaud P. 2006Rho kinases in cardiovascular physiology and pathophysiology. Circ.Res. 98 3 322 334
  87. 87. Mack C. P. Somlyo A. V. Hautmann M. Somlyo A. P. Owens G. K. 2001Smooth muscle differentiation marker gene expression is regulated by RhoA-mediated actin polymerization. J.Biol.Chem. 276 1 341 347
  88. 88. Madhavan S. Stockwell D. Cohen H. Alderman M. H. 1995Renal function during antihypertensive treatment. Lancet 345 8952 749 751
  89. 89. Marcantoni C. Ma L. J. Federspiel C. Fogo A. B. 2002Hypertensive nephrosclerosis in African Americans versus Caucasians. Kidney Int. 62 1 172 180
  90. 90. Masumoto A. Hirooka Y. Shimokawa H. Hironaga K. Setoguchi S. Takeshita A. 2001Possible involvement of Rho-kinase in the pathogenesis of hypertension in humans. Hypertension 38 6 1307 1310
  91. 91. Matsui, T., Amano, M., Yamamoto, T., Chihara, K., Nakafuku, M., Ito, M., Nakano, T., Okawa, K., Iwamatsu, A. & Kaibuchi, K. (1996). Rho-associated kinase, a novel serine/threonine kinase, as a putative target for small GTP binding protein Rho. EMBO J. 9 15 2208 2216
  92. 92. Meier-Kriesche H. U. Port F. K. Ojo A. O. Rudich S. M. Hanson J. A. Cibrik D. M. Leichtman A. B. Kaplan B. 2000Effect of waiting time on renal transplant outcome. Kidney Int. 58 3 1311 1317
  93. 93. Meier-Kriesche H. U. Schold J. D. Kaplan B. 2004Long-term renal allograft survival: have we made significant progress or is it time to rethink our analytic and therapeutic strategies? Am.J.Transplant 4 8 1289 1295
  94. 94. Meier-Kriesche H. U. Schold J. D. Srinivas T. R. Kaplan B. 2004Lack of improvement in renal allograft survival despite a marked decrease in acute rejection rates over the most recent era. Am.J.Transplant. 4 3 378 383
  95. 95. Meyer T. N. Schwesinger C. Sampogna R. V. Vaughn D. A. Stuart R. O. Steer D. L. Bush K. T. Nigam S. K. 2006Rho kinase acts at separate steps in ureteric bud and metanephric mesenchyme morphogenesis during kidney development. Differentiation 74(9-10):638 EOF 647 EOF
  96. 96. Miao L. Calvert J. W. Tang J. Zhang J. H. 2002Upregulation of small GTPase RhoA in the basilar artery from diabetic (mellitus) rats. Life Sci. 71 10 1175 1185
  97. 97. Mihatsch M. J. Ryffel B. Gudat F. 1995The differential diagnosis between rejection and cyclosporine toxicity. Kidney Int.Suppl 52:S63S69.
  98. 98. Morgan C. Sis B. Pinsk M. Yiu V. 2011Renal interstitial fibrosis in children treated with FK506 for nephrotic syndrome. Nephrol.Dial.Transplant.
  99. 99. Moriki N. Ito M. Seko T. Kureishi Y. Okamoto R. Nakakuki T. Kongo M. Isaka N. Kaibuchi K. Nakano T. 2004RhoA activation in vascular smooth muscle cells from stroke-prone spontaneously hypertensive rats. Hypertens.Res. 27 4 263 270
  100. 100. Morozumi K. Takeda A. Uchida K. Mihatsch M. J. 2004Cyclosporine nephrotoxicity: how does it affect renal allograft function and transplant morphology? Transplant Proc. 36(2 Suppl):251S-256S.
  101. 101. Mukai Y. Shimokawa H. Matoba T. Kandabashi T. Satoh S. Hiroki J. Kaibuchi K. Takeshita A. 2001Involvement of Rho-kinase in hypertensive vascular disease: a novel therapeutic target in hypertension. FASEB J. 15 6 1062 1064
  102. 102. Nagatoya K. Moriyama T. Kawada N. Takeji M. Oseto S. Murozono T. Ando A. Imai E. Hori M. 2002Y-27632 prevents tubulointerstitial fibrosis in mouse kidneys with unilateral ureteral obstruction. Kidney Int. 61 5 1684 1695
  103. 103. Nahas W. C. David-Neto E. 2009Strategies to treat children with end-stage renal dysfunction and severe lower urinary tract anomalies for receiving a kidney transplant. Pediatr.Transplant. 13 5 524 535
  104. 104. Nakagawa O. Fujisawa K. Ishizaki T. Saito Y. Nakao K. Narumiya S. 1996ROCK-I and ROCK-II, two isoforms of Rho-associated coiled-coil forming protein serine/threonine kinase in mice. FEBS Lett. 392 2 189 193
  105. 105. Nakagawa T. Tanabe K. Croker B. P. Johnson R. J. Grant M. B. Kosugi T. Li Q. 2011Endothelial dysfunction as a potential contributor in diabetic nephropathy. Nat.Rev.Nephrol. 7 1 36 44
  106. 106. Nakamura A. Hayashi K. Ozawa Y. Fujiwara K. Okubo K. Kanda T. Wakino S. Saruta T. 2003Vessel- and vasoconstrictor-dependent role of rho/rho-kinase in renal microvascular tone. J.Vasc.Res. 40 3 244 251
  107. 107. Nakano, K., Takaishi, K., Kodama, A., Mammoto, A., Shiozaki, H., Monden, M. & Takai, Y. (1999). Distinct actions and cooperative roles of ROCK and mDia in Rho small G protein-induced reorganization of the actin cytoskeleton in Madin-Darby canine kidney cells. Mol.Biol.Cell 8 10 2481 2491
  108. 108. Nankivell, B.J., Borrows, R.J., Fung, C.L., O’Connell, P.J., Allen, R.D. & Chapman, J.R. (2003). The natural history of chronic allograft nephropathy. N.Engl.J.Med. 24 349 2326 2333
  109. 109. Nath K. A. 1992Tubulointerstitial changes as a major determinant in the progression of renal damage. Am.J.Kidney Dis. 20 1 1 17
  110. 110. Nishikimi T. Akimoto K. Wang X. Mori Y. Tadokoro K. Ishikawa Y. Shimokawa H. Ono H. Matsuoka H. 2004aFasudil, a Rho-kinase inhibitor, attenuates glomerulosclerosis in Dahl salt-sensitive rats. J.Hypertens. 22 9 1787 1796
  111. 111. Nishikimi T. Akimoto K. Wang X. Mori Y. Tadokoro K. Ishikawa Y. Shimokawa H. Ono H. Matsuoka H. 2004bFasudil, a Rho-kinase inhibitor, attenuates glomerulosclerosis in Dahl salt-sensitive rats. J.Hypertens. 22 9 1787 1796
  112. 112. Nobes C. Hall A. 1994Regulation and function of the Rho subfamily of small GTPases. Curr.Opin.Genet.Dev. 4 1 77 81
  113. 113. Ohtsu H. Suzuki H. Nakashima H. Dhobale S. Frank G. D. Motley E. D. Eguchi S. 2006Angiotensin II signal transduction through small GTP-binding proteins: mechanism and significance in vascular smooth muscle cells. Hypertension 48 4 534 540
  114. 114. Oka M. Fagan K. A. Jones P. L. Mc Murtry I. F. 2008Therapeutic potential of RhoA/Rho kinase inhibitors in pulmonary hypertension. Br.J.Pharmacol. 155 4 444 454
  115. 115. Okusa M. D. Linden J. Huang L. Rieger J. M. Macdonald T. L. Huynh L. P. 2000A(2A) adenosine receptor-mediated inhibition of renal injury and neutrophil adhesion. Am.J.Physiol Renal Physiol 279(5):F809F818.
  116. 116. Osterby R. Nyberg G. Hedman L. Karlberg I. Persson H. Svalander C. 1991Kidney transplantation in type 1 (insulin-dependent) diabetic patients. Early glomerulopathy. Diabetologia 34 9 668 674
  117. 117. Parekh J. Bostrom A. Feng S. 2010Diabetes mellitus: a risk factor for delayed graft function after deceased donor kidney transplantation. Am.J.Transplant. 10 2 298 303
  118. 118. Parizi M. Howard E. W. Tomasek J. J. 2000Regulation of LPA-promoted myofibroblast contraction: role of Rho, myosin light chain kinase, and myosin light chain phosphatase. Exp.Cell Res. 254 2 210 220
  119. 119. Pascual M. Theruvath T. Kawai T. Tolkoff-Rubin N. Cosimi A. B. 2002Strategies to improve long-term outcomes after renal transplantation. N.Engl.J.Med. 346 8 580 590
  120. 120. Patel, S., Takagi, K.I., Suzuki, J., Imaizumi, A., Kimura, T., Mason, R.M., Kamimura, T. & Zhang, Z. (2005). RhoGTPase activation is a key step in renal epithelial mesenchymal transdifferentiation. J.Am.Soc.Nephrol. 7 16 1977 1984
  121. 121. Peng F. Wu D. Gao B. Ingram A. J. Zhang B. Chorneyko K. Mc Kenzie R. Krepinsky J. C. 2008RhoA/Rho-kinase contribute to the pathogenesis of diabetic renal disease. Diabetes 57 6 1683 1692
  122. 122. Perkins B. A. Ficociello L. H. Ostrander B. E. Silva K. H. Weinberg J. Warram J. H. Krolewski A. S. 2007Microalbuminuria and the risk for early progressive renal function decline in type 1 diabetes. J.Am.Soc.Nephrol. 18 4 1353 1361
  123. 123. Prakash J. de Borst M. H. Lacombe M. Opdam F. Klok P. A. van G. H. Meijer D. K. Moolenaar F. Poelstra K. Kok R. J. 2008Inhibition of Renal Rho Kinase Attenuates Ischemia/Reperfusion-Induced Injury. J.Am.Soc.Nephrol.
  124. 124. Rabb H. Wang Z. Nemoto T. Hotchkiss J. Yokota N. Soleimani M. 2003Acute renal failure leads to dysregulation of lung salt and water channels. Kidney Int. 63 2 600 606
  125. 125. Racusen L. C. Solez K. Colvin R. B. Bonsib S. M. Castro M. C. Cavallo T. Croker B. P. Demetris A. J. Drachenberg C. B. Fogo A. B. Furness P. Gaber L. W. Gibson I. W. Glotz D. Goldberg J. C. Grande J. Halloran P. F. Hansen H. E. Hartley B. Hayry P. J. Hill C. M. Hoffman E. O. Hunsicker L. G. Lindblad A. S. Yamaguchi Y. . 1999The Banff 97 working classification of renal allograft pathology. Kidney Int. 55 2 713 723
  126. 126. Reuter, S., Reiermann, S., Worner, R., Schroter, R., Edemir, B., Buck, F., Henning, S., Peter-Katalinic, J., Vollenbroker, B., Amann, K., Pavenstadt, H., Schlatter, E. & Gabriels, G. (2010). IF/TA-related metabolic changes--proteome analysis of rat renal allografts. Nephrol.Dial.Transplant. 8 25 2492 2501
  127. 127. Riento K. Ridley A. J. 2003Rocks: multifunctional kinases in cell behaviour. Nat.Rev.Mol.Cell Biol. 4 6 446 456
  128. 128. Rigamonti W. Capizzi A. Zacchello G. Capizzi V. Zanon G. F. Montini G. Murer L. Glazel G. P. 2005Kidney transplantation into bladder augmentation or urinary diversion: long-term results. Transplantation 80 10 1435 1440
  129. 129. Rikitake, Y., Kim, H.H., Huang, Z., Seto, M., Yano, K., Asano, T., Moskowitz, M.A. & Liao, J.K. (2005a). Inhibition of Rho kinase (ROCK) leads to increased cerebral blood flow and stroke protection. Stroke 10 36 2251 2257
  130. 130. Rikitake Y. Liao J. K. 2005Rho-kinase mediates hyperglycemia-induced plasminogen activator inhibitor-1 expression in vascular endothelial cells. Circulation 111 24 3261 3268
  131. 131. Rikitake Y. Oyama N. Wang C. Y. Noma K. Satoh M. Kim H. H. Liao J. K. 2005bDecreased perivascular fibrosis but not cardiac hypertrophy in ROCK1+/- haploinsufficient mice. Circulation 112 19 2959 2965
  132. 132. Rodrigo E. Fernandez-Fresnedo G. Valero R. Ruiz J. C. Pinera C. Palomar R. Gonzalez-Cotorruelo J. Gomez-Alamillo C. Arias M. 2006New-onset diabetes after kidney transplantation: risk factors. J.Am.Soc.Nephrol. 17(12 Suppl 3):S291S295.
  133. 133. Rodrigues-Diez, R., Carvajal-Gonzalez, G., Sanchez-Lopez, E., Rodriguez-Vita, J., Rodrigues, D.R., Selgas, R., Ortiz, A., Egido, J., Mezzano, S. & Ruiz-Ortega, M. (2008). Pharmacological modulation of epithelial mesenchymal transition caused by angiotensin II. Role of ROCK and MAPK pathways. Pharm.Res. 10 25 2447 2461
  134. 134. Rugg, V. 2003 Diabetes and kidney disease: time to Act. International Diabetes Federation and the International Society of Nephrology.. 20-2-2011. (GENERIC
  135. 135. Ref Type:Electronic Citation
  136. 136. Rule A. D. Amer H. Cornell L. D. Taler S. J. Cosio F. G. Kremers W. K. Textor S. C. Stegall M. D. 2010The association between age and nephrosclerosis on renal biopsy among healthy adults. Ann.Intern.Med. 152 9 561 567
  137. 137. Ruperez M. Sanchez-Lopez E. Blanco-Colio L. M. Esteban V. Rodriguez-Vita J. Plaza J. J. Egido J. Ruiz-Ortega M. 2005The Rho-kinase pathway regulates angiotensin II-induced renal damage. Kidney Int.Suppl (99):S39S45.
  138. 138. Salifu M. O. Nicastri A. D. Markell M. S. Ghali H. Sommer B. G. Friedman E. A. 2004Allograft diabetic nephropathy may progress to end-stage renal disease. Pediatr.Transplant. 8 4 351 356
  139. 139. Samaniego R. Sanchez-Martin L. Estecha A. Sanchez-Mateos P. 2007Rho/ROCK and myosin II control the polarized distribution of endocytic clathrin structures at the uropod of moving T lymphocytes. J.Cell Sci. 120(Pt 20):3534 EOF 43 EOF
  140. 140. Santen S. Wang Y. Laschke M. W. Menger M. D. Jeppsson B. Thorlacius H. 2010Rho-kinase signalling regulates CXC chemokine formation and leukocyte recruitment in colonic ischemia-reperfusion. Int.J.Colorectal Dis. 25 9 1063 1070
  141. 141. Satoh S. Toshima Y. Hitomi A. Ikegaki I. Seto M. Asano T. 2008Wide therapeutic time window for Rho-kinase inhibition therapy in ischemic brain damage in a rat cerebral thrombosis model. Brain Res. 1193 102 108
  142. 142. Satoh S. Yamaguchi T. Hitomi A. Sato N. Shiraiwa K. Ikegaki I. Asano T. Shimokawa H. 2002Fasudil attenuates interstitial fibrosis in rat kidneys with unilateral ureteral obstruction. Eur.J.Pharmacol. 455(2-3):169 EOF 74 EOF
  143. 143. Sauzeau V. Le J. H. Cario-Toumaniantz C. Smolenski A. Lohmann S. M. Bertoglio J. Chardin P. Pacaud P. Loirand G. 2000Cyclic GMP-dependent protein kinase signaling pathway inhibits RhoA-induced Ca2+ sensitization of contraction in vascular smooth muscle. J.Biol.Chem. 275 28 21722 21729
  144. 144. Schena F. P. Gesualdo L. 2005Pathogenetic mechanisms of diabetic nephropathy. J.Am.Soc.Nephrol. 16 Suppl 1:S30S33.
  145. 145. Seasholtz T. M. Zhang T. Morissette M. R. Howes A. L. Yang A. H. Brown J. H. 2001Increased expression and activity of RhoA are associated with increased DNA synthesis and reduced 27Kip1) expression in the vasculature of hypertensive rats. Circ.Res. 89(6):488-495.
  146. 146. Seko T. Ito M. Kureishi Y. Okamoto R. Moriki N. Onishi K. Isaka N. Hartshorne D. J. Nakano T. 2003Activation of RhoA and inhibition of myosin phosphatase as important components in hypertension in vascular smooth muscle. Circ.Res. 92 4 411 418
  147. 147. Sheldon C. A. Gonzalez R. Burns M. W. Gilbert A. Buson H. Mitchell M. E. 1994Renal transplantation into the dysfunctional bladder: the role of adjunctive bladder reconstruction. J.Urol. 152 3 972 975
  148. 148. Solez K. Colvin R. B. Racusen L. C. Sis B. Halloran P. F. Birk P. E. Campbell P. M. Cascalho M. Collins A. B. Demetris A. J. Drachenberg C. B. Gibson I. W. Grimm P. C. Haas M. Lerut E. Liapis H. Mannon R. B. Marcus P. B. Mengel M. Mihatsch M. J. Nankivell B. J. Nickeleit V. Papadimitriou J. C. Platt J. L. Randhawa P. Roberts I. Salinas-Madriga L. Salomon D. R. Seron D. Sheaff M. Weening J. J. 2007Banff ‘05 Meeting Report: differential diagnosis of chronic allograft injury and elimination of chronic allograft nephropathy (‘CAN’). Am.J.Transplant. 7 3 518 526
  149. 149. Son Y. K. Oh J. S. Kim S. M. Jeon J. M. Shin Y. H. Kim J. K. 2010Clinical outcome of preemptive kidney transplantation in patients with diabetes mellitus. Transplant.Proc. 42 9 3497 3502
  150. 150. Song J. Lu Y. P. Luo G. H. Yang L. Ma X. Xia Q. J. Shi Y. J. Li Y. P. 2008Effects of mycophenolate mofetil on chronic allograft nephropathy by affecting RHO/ROCK signal pathways. Transplant.Proc. 40 8 2790 2794
  151. 151. Staessen J. A. Wang J. Bianchi G. Birkenhager W. H. 2003Essential hypertension. Lancet 361 9369 1629 1641
  152. 152. Takeda K. Jin M. B. Fujita M. Fukai M. Sakurai T. Nakayama M. Taniguchi M. Suzuki T. Shimamura T. Furukawa H. Todo S. 2003A novel inhibitor of Rho-associated protein kinase, Y-27632, ameliorates hepatic ischemia and reperfusion injury in rats. Surgery 133 2 197 206
  153. 153. Takemoto M. Sun J. Hiroki J. Shimokawa H. Liao J. K. 2002Rho-kinase mediates hypoxia-induced downregulation of endothelial nitric oxide synthase. Circulation 106 1 57 62
  154. 154. Takesono A. Heasman S. J. Wojciak-Stothard B. Garg R. Ridley A. J. 2010Microtubules regulate migratory polarity through Rho/ROCK signaling in T cells. PLoS.ONE. 5(1):e8774 EOF
  155. 155. Teraishi K. Kurata H. Nakajima A. Takaoka M. Matsumura Y. 2004Preventive effect of Y-27632, a selective Rho-kinase inhibitor, on ischemia/reperfusion-induced acute renal failure in rats. Eur.J.Pharmacol. 505(1-3):205 EOF 11 EOF
  156. 156. Terasaki P. I. Ozawa M. 2004Predicting kidney graft failure by HLA antibodies: a prospective trial. Am.J.Transplant 4 3 438 443
  157. 157. Tharaux P. L. Bukoski R. C. Rocha P. N. Crowley S. D. Ruiz P. Nataraj C. Howell D. N. Kaibuchi K. Spurney R. F. Coffman T. M. 2003Rho kinase promotes alloimmune responses by regulating the proliferation and structure of T cells. J.Immunol. 171 1 96 105
  158. 158. Thornton M. A. Winn R. Alpers C. E. Zager R. A. 1989An evaluation of the neutrophil as a mediator of in vivo renal ischemic-reperfusion injury. Am.J.Pathol. 135 3 509 515
  159. 159. Toshima, Y., Satoh, S., Ikegaki, I. & Asano, T. (2000). A new model of cerebral microthrombosis in rats and the neuroprotective effect of a Rho-kinase inhibitor. Stroke 9 31 2245 2250
  160. 160. Toto, R.B. (2003). Hypertensive nephrosclerosis in African Americans. Kidney Int. 6 64 2331 2341
  161. 161. Truong L. D. Farhood A. Tasby J. Gillum D. 1992Experimental chronic renal ischemia: morphologic and immunologic studies. Kidney Int. 41 6 1676 1689
  162. 162. Tullius S. G. Tilney N. L. 1995Both alloantigen-dependent and-independent factors influence chronic allograft rejection. Transplantation 59 3 313 318
  163. 163. Uehata M. Ishizaki T. Satoh H. Ono T. Kawahara T. Morishita T. Tamakawa H. Yamagami K. Inui J. Maekawa M. Narumiya S. 1997Calcium sensitization of smooth muscle mediated by a Rho-associated protein kinase in hypertension. Nature 389 6654 990 994
  164. 164. Unbekandt M. Davies J. A. 2010Dissociation of embryonic kidneys followed by reaggregation allows the formation of renal tissues. Kidney Int. 77 5 407 416
  165. 165. Van Buren P. N. Toto R. 2011Hypertension in diabetic nephropathy: epidemiology, mechanisms, and management. Adv.Chronic.Kidney Dis. 18 1 28 41
  166. 166. Vemula S. Shi J. Hanneman P. Wei L. Kapur R. 2010ROCK1 functions as a suppressor of inflammatory cell migration by regulating PTEN phosphorylation and stability. Blood 115 9 1785 1796
  167. 167. Vernon M. A. Mylonas K. J. Hughes J. 2010Macrophages and renal fibrosis. Semin.Nephrol. 30 3 302 317
  168. 168. Versteilen A. M. Blaauw N. Di M. F. Groeneveld A. B. Sipkema P. Musters R. J. Tangelder G. J. 2011Rho-Kinase Inhibition Reduces Early Microvascular Leukocyte Accumulation in the Rat Kidney following Ischemia-Reperfusion Injury: Roles of Nitric Oxide and Blood Flow. Nephron Exp.Nephrol. 118(4):e79e86.
  169. 169. Versteilen A. M. Korstjens I. J. Musters R. J. Groeneveld A. B. Sipkema P. 2006Rho kinase regulates renal blood flow by modulating eNOS activity in ischemia-reperfusion of the rat kidney. Am.J.Physiol Renal Physiol 291(3):F606F611.
  170. 170. Vielhauer V. Kulkarni O. Reichel C. A. Anders H. J. 2010Targeting the recruitment of monocytes and macrophages in renal disease. Semin.Nephrol. 30 3 318 333
  171. 171. Watanabe K. Ueno M. Kamiya D. Nishiyama A. Matsumura M. Wataya T. Takahashi J. B. Nishikawa S. Nishikawa S. Muguruma K. Sasai Y. 2007A ROCK inhibitor permits survival of dissociated human embryonic stem cells. Nat.Biotechnol. 25 6 681 686
  172. 172. Weber D. S. Webb R. C. 2001Enhanced relaxation to the rho-kinase inhibitor Y-27632 in mesenteric arteries from mineralocorticoid hypertensive rats. Pharmacology 63 3 129 133
  173. 173. Wiesbauer F. Heinze G. Regele H. Horl W. H. Schernthaner G. H. Schwarz C. Kainz A. Kramar R. Oberbauer R. 2010Glucose control is associated with patient survival in diabetic patients after renal transplantation. Transplantation 89 5 612 619
  174. 174. Wilkinson A. Davidson J. Dotta F. Home P. D. Keown P. Kiberd B. Jardine A. Levitt N. Marchetti P. Markell M. Naicker S. O’Connell P. Schnitzler M. Standl E. Torregosa J. V. Uchida K. Valantine H. Villamil F. Vincenti F. Wissing M. 2005Guidelines for the treatment and management of new-onset diabetes after transplantation. Clin.Transplant. 19 3 291 298
  175. 175. Wojciechowski D. Onozato M. L. Gonin J. 2009Rapid onset of diabetic nephropathy in three renal allografts despite normoglycemia. Clin.Nephrol. 71 6 719 724
  176. 176. Wolfe R. A. Ashby V. B. Milford E. L. Ojo A. O. Ettenger R. E. Agodoa L. Y. Held P. J. Port F. K. 1999Comparison of mortality in all patients on dialysis, patients on dialysis awaiting transplantation, and recipients of a first cadaveric transplant. N.Engl.J.Med. 341 23 1725 1730
  177. 177. Wolfrum, S., Dendorfer, A., Rikitake, Y., Stalker, T.J., Gong, Y., Scalia, R., Dominiak, P. & Liao, J.K. (2004). Inhibition of Rho-kinase leads to rapid activation of phosphatidylinositol 3-kinase/protein kinase Akt and cardiovascular protection. Arterioscler.Thromb.Vasc.Biol. 10 24 1842 1847
  178. 178. Worthylake R. A. Burridge K. 2003RhoA and ROCK promote migration by limiting membrane protrusions. J.Biol.Chem. 278 15 13578 13584
  179. 179. Worthylake R. A. Lemoine S. Watson J. M. Burridge K. 2001RhoA is required for monocyte tail retraction during transendothelial migration. J.Cell Biol. 154 1 147 160
  180. 180. Yamakawa T. Tanaka S. Numaguchi K. Yamakawa Y. Motley E. D. Ichihara S. Inagami T. 2000Involvement of Rho-kinase in angiotensin II-induced hypertrophy of rat vascular smooth muscle cells. Hypertension 35(1 Pt 2):313 EOF 8 EOF
  181. 181. Yousif M. H. 2006Role of protein kinases in mediating diabetes-induced augmented vasoconstriction to endothelin-1 in the renal arteries of STZ-diabetic rats. Cell Biochem.Funct. 24 5 397 405
  182. 182. Ysebaert D. K. De Greef K. E. Vercauteren S. R. Ghielli M. Verpooten G. A. Eyskens E. J. De Broe M. E. 2000Identification and kinetics of leukocytes after severe ischaemia/reperfusion renal injury. Nephrol.Dial.Transplant. 15 10 1562 1574
  183. 183. Zavadil J. Bottinger E. P. 2005TGF-beta and epithelial-to-mesenchymal transitions. Oncogene 24 37 5764 5774
  184. 184. Zeng, L., Xu, H., Chew, T.L., Eng, E., Sadeghi, M.M., Adler, S., Kanwar, Y.S. & Danesh, F.R. (2005). HMG CoA reductase inhibition modulates VEGF-induced endothelial cell hyperpermeability by preventing RhoA activation and myosin regulatory light chain phosphorylation. FASEB J. 13 19 1845 1847
  185. 185. Zhang W. Liu M. Wu Y. Zhu P. Yin C. Zhang W. Gu M. 2007Protective effects of atorvastatin on chronic allograft nephropathy in rats. J.Surg.Res. 143 2 428 436
  186. 186. Zimmet P. Alberti K. G. Shaw J. 2001Global and societal implications of the diabetes epidemic. Nature 414 6865 782 787
  187. 187. Zucchelli P. Zuccala A. 1994Primary hypertension--how does it cause renal failure? Nephrol.Dial.Transplant. 9 3 223 225

Written By

Stefan Reuter, Dominik Kentrup and Eckhart Büssemaker

Submitted: 02 November 2010 Published: 23 August 2011