Established and Newly Proposed Mechanisms of Chronic Cyclosporine Nephropathy

Article information

Korean J Intern Med. 2009;24(2):81-92
Publication date (electronic) : 2009 June 08
doi : https://doi.org/10.3904/kjim.2009.24.2.81
Division of Nephrology, Transplantation research center, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea.
Correspondence to Chul Woo Yang, MD, PhD. Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, 505 Banpo-dong, Seocho-gu, Seoul 137-040, Korea. Tel: 82-2-2258-6037, Fax: 82-2-536-0323, yangch@catholic.ac.kr

Abstract

Cyclosporine (CsA) has improved patient and graft survival rates following solid-organ transplantation and has shown significant clinical benefits in the management of autoimmune diseases. However, the clinical use of CsA is often limited by acute or chronic nephropathy, which remains a major problem. Acute nephropathy depends on the dosage of CsA and appears to be caused by a reduction in renal blood flow related to afferent arteriolar vasoconstriction. However, the mechanisms underlying chronic CsA nephropathy are not completely understood. Activation of the intrarenal renin-angiotensin system (RAS), increased release of endothelin-1, dysregulation of nitric oxide (NO) and NO synthase, up-regulation of transforming growth factor-beta1 (TGF-β1), inappropriate apoptosis, stimulation of inflammatory mediators, enhanced innate immunity, endoplasmic reticulum stress, and autophagy have all been implicated in the pathogenesis of chronic CsA nephropathy. Reducing the CsA dosage or using other renoprotective drugs (angiotensin II receptor antagonist, mycophenolate mofetil, and statins, etc.) may ameliorate chronic CsA-induced renal injury. This review discusses old and new concepts in CsA nephropathy and preventive strategies for this clinical dilemma.

INTRODUCTION

Cyclosporine (CsA) was first approved by the US Food and Drug Administration in the early 1980s for prophylactic anti-rejection therapy in patients receiving allogeneic transplants (kidney, liver, and heart). Its introduction has significantly improved both allograft and patient survival for over two decades [1,2].

The immunosuppressive action of CsA involves the intracellular interaction of CsA and calcineurin phosphatase, which reduces the production of interleukin-2 (IL-2). CsA initially binds to a specific family of receptors known as cyclophilins [3]. This drug-receptor complex inhibits the activation of calcineurin phosphatase, a secondary messenger in the dephosphorylation and activation of the nuclear factor of activation of T-cells (NF-AT). NF-AT is a regulatory protein that promotes the transcription and production of IL-2 and other cytokines that promote the growth and proliferation of T- and B-cells [4]. Inhibition of IL-2 production by CsA halts the proliferation and activation of helper and cytotoxic T-cells [5].

Despite the therapeutic benefits of CsA, several adverse effects have been reported in both transplant and non-transplant settings (i.e., autoimmune disorders), including toxicities (nephrotoxicity, hepatotoxicity, and neurotoxicity), hypertension, dyslipidemia, gingival hyperplasia, hypertrichosis, malignancies, and an increased risk of cardiovascular events [2,6]. The most clinically important complication is chronic CsA nephropathy, which is one of the known non-immunological factors causing chronic allograft nephropathy [7]. Chronic CsA nephropathy is characterized by progressive renal dysfunction, afferent arteriolopathy, inflammatory cell influx, striped tubulointerstitial fibrosis, and increased intrarenal immunogenicity [8-10]. Nankivell et al. [11] showed that almost all recipients presented evidence of chronic CsA nephropathy after 10 years of treatment with calcineurin inhibitors. The exact mechanism of this complication is not fully understood, although many potential mechanisms have been proposed.

Using a well-established animal model, we and other groups recently demonstrated that the mechanism of CsA-induced renal injury includes immunological and non-immunological pathways, as outlined in Figure 1. This review article summarizes our current understanding of the pathogenesis of chronic CsA nephropathy and discusses recent literature on the prevention and delay of this complication.

Figure 1

A schematic diagram of the pathogenesis of chronic CsA-induced renal injury. Chronic treatment with CsA induces renal injury by immunologic and non-immunologic pathways. The non-immunologic pathway includes hemodynamic (low-grade ischemia) and non-hemodynamic (mediators) mechanisms, although both ultimately result in tubulointerstitial inflammation, striped fibrosis, and an increased rate of chronic rejection. The immunologic pathway may be triggered by the endogenous production of HSP 70, a known activator of TLRs. Consequently, activation of the innate immune system by TLRs and MHC class II molecules leads to the activation of NF-κB and AP-1.

PREVIOUSLY ESTABLISHED MECHANISMS OF CsA-INDUCED RENAL INJURY

CsA-induced low-grade ischemia

Long-term administration of CsA rapidly decreases glomerular filtration rate (GFR) and renal blood flow by inducing vasoconstriction or increased intrarenal vascular resistance, which ultimately results in low-grade ischemic injury [2,6]. Chronic ischemia caused by CsA is believed to be associated with reactive oxygen species and lipid peroxidation [12,13]. This hypothesis is supported by the observation that antioxidant therapies significantly attenuate chronic CsA nephropathy [14,15].

The mechanism responsible for vasoconstriction is partially related to the activation of the intrarenal renin-angiotensin system (RAS) and imbalances in prostaglandins and thromboxane [13,16,17]. In addition, hypersecretion of endothelin-1 plays an important role in CsA-induced vasoconstriction [18]. Endothelin-1 is a potent vasoconstrictor, widely released in the kidney and vascular beds, and acts locally to increase vascular tone and regulate blood flow, GFR, and sodium reabsorption. Furthermore, endothelin-1 can also disrupt renal architecture through its effects on extracellular matrix (ECM) accumulation and tubulointerstitial fibrosis. However, after long-term withdrawal of CsA, both vasoconstriction and afferent arteriolopathy are shown to be normalize. CsA may also increase systemic vascular resistance associated with activation of the sympathetic nervous system [19].

CsA-induced activation of the renin-angiotensin system

Activation of the RAS, especially the intrarenal RAS, plays an essential role in the pathogenesis of chronic CsA nephropathy [20,21]. However, the mechanism of RAS activation in this complication remains unknown. One accepted hypothesis is that CsA increases renin release from juxtaglomerular cells. In a reproducible rat model of chronic CsA nephropathy developed by Rosen et al. [22] and Elzinga et al. [23], CsA administration to animals on a low-salt diet induced a significant decrease in GFR and histological changes similar to those described in patients undergoing long-term CsA therapy. Salt depletion activates the RAS, which is implicated in the changes in renal hemodynamics and function that follow CsA administration. Using this model, we and other groups clearly demonstrated that CsA significantly increases renin and angiotensin II (Ang II) immunoreactivity in the kidney [21,24,25], supporting a role for the intrarenal RAS in the pathogenesis of chronic CsA nephropathy.

Activation of the intrarenal RAS induces renal injury via hemodynamic and non-hemodynamic pathways. The RAS mediates vasoconstriction hemodynamically and thus leads to low-grade ischemia, as reviewed above. However, the RAS may also induce renal injury non-hemodynamically via stimulation of tubulointerstitial inflammation, the expression of transforming growth factor-beta 1 (TGF-β1) and vascular endothelial growth factor (VEGF), and increased renal cell apoptosis [26-29]. Blocking this system with either angiotensin-converting enzyme (ACE) inhibitors or angiotensin II (Ang II) receptor type I antagonists mitigates all of the above parameters and confers a renoprotective effect during chronic CsA nephropathy [21,25-28].

The pathogenesis of chronic CsA nephropathy may involve an association between the RAS and expression of the Klotho gene. The Klotho gene is an anti-aging gene involved in the suppression of several aging phenotypes [30]. Klotho gene expression in the kidneys is greatly reduced in patients with chronic renal failure [31] and is also suppressed in acute renal failure in ischemic-reperfusion injury murine models [32]. Recently, we found that the activation of the RAS suppresses Klotho gene expression in an animal model of CsA nephropathy and that blocking the RAS with Ang II receptor type 1 antagonists alleviates suppression of the Klotho gene and tissue injury (unpublished data). These results suggest that RAS activation suppresses Klotho gene expression and leads to chronic CsA-induced renal injury.

Nitric oxide and CsA-induced renal injury

In the kidney, nitric oxide (NO) is a vasodilating factor that plays a key role in maintaining vascular tone. In addition, NO decreases glomerular thrombosis and ischemia, mesangial cell proliferation, ECM protein synthesis, and interstitial inflammatory cell infiltration.

NO is produced from L-arginine by the action of NO synthase (NOS), of which at least three isoforms have been identified: neuronal NOS (nNOS), inducible NOS (iNOS), and endothelial NOS (eNOS). These three NOS isoforms are all found in the kidney: nNOS is specifically expressed in macula densa cells, iNOS has been observed in mesangial and proximal tubular cells, and eNOS is expressed mainly in endothelial cells of the afferent and efferent arterioles and glomerular capillaries [33]. Chronic CsA treatment has been shown to differentially influence NOS isoform expression and interfere with NO production; however, contradictory results have also been reported [34-37].

The role of NO in the pathogenesis of chronic CsA nephropathy has been examined. We demonstrated that exogenous supplementation with L-arginine effectively prevents CsA-induced renal dysfunction, arteriolopathy, and interstitial fibrosis in rats [38]. Shihab et al. [39,40] also reported that L-arginine treatment attenuates CsA-induced TGF-β1 over-expression and ECM deposition, whereas these effects are reversed by treatment with N-nitro-L-arginine, a potent competitive inhibitor of NO biosynthesis.

Osteopontin as a pro-inflammatory cytokine in CsA-induced renal injury

The molecular mechanism underlying chronic CsA nephropathy is multifactorial. Because interstitial inflammatory events precede ongoing fibrosis [9], the up-regulation of chemoattractants and subsequent inflammatory cell infiltration are thought to play important roles in this condition.

Osteopontin (OPN) is a highly acidic phosphoprotein containing an arginine-glycine-aspartic acid (RGD) motif. It is involved in cell adhesion and migration [41] and is expressed by several cell types in a constitutive or inducible fashion. These include osteoclasts, some epithelial cells, macrophages, T-cells, smooth muscle cells, and tumor cells [42-47]. OPN acts as a chemotactic factor for macrophages and monocytes by binding to ligands such as αvβ3 integrin, CD44, collagen type I, and fibronectin [48,49]. The functional role of OPN, with respect to macrophage attraction, has been recently described in vivo and in vitro [41,50]. Moreover, the subcutaneous injection of OPN into rats produces massive macrophage accumulation, which is inhibited by the administration of an anti-OPN antibody [51].

In the kidney, OPN is expressed constitutively in the renal medulla in the loop of Henle and the distal convoluted tubules; it is absent in the normal renal cortex, with the exception of the parietal epithelium of Bowman's capsule. The up-regulation of OPN expression is strongly correlated with macrophage infiltration in several models of kidney diseases [28,52-55]. Young et al. [9] and Pichler et al. [28] reported that CsA treatment up-regulates OPN gene expression and is correlated with interstitial macrophage infiltration and fibrosis. We found that OPN mRNA and protein were constitutively present in the tubular epithelium, collecting ducts, and uroepithelial cells in control rat kidneys, whereas most cortical structures were negative for OPN. In contrast, the levels of OPN mRNA and protein increased dramatically in the tubular epithelium and Bowman's capsule cells in CsA-treated rat kidneys. The most striking change was observed in the renal cortex, which normally expresses very little constitutive OPN. Of note, the sites of strong OPN expression were in areas of macrophage influx and severe tubulointerstitial fibrosis [25,56]. Furthermore, a study in OPN-null mice demonstrated that the lack of OPN expression attenuated chronic CsA nephropathy [57]. These findings imply that OPN plays a pathogenic role in CsA-induced renal injury.

TGF-β1 as a pro-fibrotic cytokine in CsA-induced renal injury

TGF-β1 is a key cytokine implicated in the pathogenesis of a wide range of kidney diseases characterized by glomerulosclerosis and tubulointerstitial fibrosis, including chronic CsA nephropathy. Both in vivo and in vitro studies have shown that CsA administration is associated with dose-dependent increases in TGF-β1 expression. Shihab et al. [58,59] demonstrated that CsA-induced TGF-β1 up-regulation results in tubulointerstitial fibrosis, probably via its actions on ECM synthesis and degradation, and plasminogen activator inhibitor-1 plays a role in this process. Furthermore, administration of a specific TGF-β-neutralizing antibody ameliorated morphological alterations and preserved renal function in a mouse model of chronic CsA nephropathy [60].

TGF-β1 is secreted as a biologically inactive complex requiring in vivo activation. This latent TGF-β1 complex is activated via cleavage of its N-terminal latency-associated peptide to yield mature dimeric TGF-β1 through enzymatic and non-enzymatic mechanisms, or by the presence of the proteoglycan decorin and the scavenging protein α2-macroglobulin [61-65]. Therefore, increased amounts of TGF-β1 mRNA or protein may not actually represent parallel changes in its biologic activity.

Keratoepithelin (βig-h3) is a secreted matrix protein originally identified from a TGF-β1-stimulated human lung adenocarcinoma cell line (A549) [66]. βig-h3 has been proposed as one of the ECM components [67]; although the precise physiologic function of βig-h3 is unclear, it may connect various matrix components and resident cells, thereby serving as a bifunctional linker protein [68,69]. Thus, βig-h3 expression has been used to assess the biological activity of TGF-β1 [70]. Langham et al. [71] reported that βig-h3 production increased significantly in non-renal transplant recipients with chronic CsA nephropathy. More recently, we found that βig-h3 mRNA and protein were normally expressed in the cortex and outer medulla, specifically localized in the terminal portion of afferent arterioles (vascular pole of glomerulus), the S3 segment (parser recta) of the proximal tubules, and the distal convoluted tubules. However, in the CsA-treated rat kidney, βig-h3 gene expression was significantly up-regulated in the interstitium, but not in afferent arterioles or tubules, where interstitial expansion and fibrosis developed [72]. Thus, βig-h3 may be a useful index of TGF-β1 bioactivity and may reflect the degree of tubulointerstitial injury in chronic CsA nephropathy.

CsA-induced cell death

Tubulointerstitial injury is the prominent feature of chronic CsA nephropathy, and the major form of cell death is apoptosis [73]. Excessive loss of cellularity via apoptosis has been observed in fibrotic areas in renal biopsy specimens obtained from patients receiving long-term CsA therapy [74]. Apoptosis is an active mechanism of cell clearance and plays a key role in the regulation of cell number during development, in tissue homeostasis, and following insults. In the kidney, apoptosis may be beneficial [75] but is deleterious if enough resident cells are lost [76].

CsA has been shown to induce apoptotic cell death not only in T-lymphocytes, thus interfering with T-cell function [77], but also in some renal cells, resulting in the deterioration of kidney structure [26,78,79]. Thomas et al. [26] were the first to characterize the close link between apoptosis and interstitial fibrosis in a rat model of chronic CsA nephropathy. Subsequently, we and other groups have reported that CsA-induced renal cell apoptosis is associated with gene families such as the Bcl-2 proteins, Fas and Fas-ligand, p53, and the caspases [73,80], and that Ang II, NO, intrarenal growth factors (TGF-β1), epidermal growth factor (EGF), and macrophages are also involved [21,26,81-83].

CsA-induced activation of nuclear factor-kappa B and activating protein-1

Transcription factors such as nuclear factor-kappa B (NF-κB) and activating protein-1 (AP-1) regulate the gene expression of several cytokines, chemotactic proteins, adhesion molecules, and matrix proteins involved in inflammation, immunologic responses, cell differentiation, and the control of growth [84]. The transcription factors NF-κB and AP-1 are activated by a number of physiological and non-physiological stimuli such as cytokines, mitogens, viruses, mechanical factors, oxidative stress, and a variety of chemical agents [85-87]. Recent studies suggest that the activation of NF-κB and AP-1 is involved in the transcription of monocyte chemoattractant protein-1 and TGF-β1 in the kidney, which is regulated by Ang II or proteinuria [84,88-90]. In rats, Asai et al. [91] reported that the administration of CsA stimulated NF-κB and AP-1 DNA-binding activity, and this effect was blocked by the ACE inhibitor benazepril and by magnesium supplementation. In addition, we reported the involvement of the activation of NF-κB and AP-1 during the activation of innate immunity in chronic CsA nephropathy (described below) [92].

NEWLY DESCRIBED MECHANISMS OF CSA-INDUCED RENAL INJURY

Toll-like receptors and inflammation

The innate immune system primarily evolved as a rapid first line of defense, and is initiated by stimulation of toll-like receptors (TLRs) via their specific ligands [93,94]. Growing evidence indicates that the TLRs play a potential role in the pathophysiology of injury-associated kidney diseases [95]. Previously, we reported that CsA-induced renal injury produces endogenous TLR ligands such as heat shock protein (HSP) 70, up-regulates major histocompatibility complex (MHC) class II protein and TLR expression in renal tubular cells, and promotes the maturation of dendritic cells [92]. HSP 70 is an activator of TLRs and the innate immune system, and TLRs are recognized as sensors of pathogen-associated molecular patterns crucial for the initiation of an innate immune response [96,97]. Recently, we demonstrated that macrophage depletion decreased both TLR2 and MHC class II expression in renal tubular cells [83], which suggests a role for macrophages in immunologic injury. These results suggest the close association between the activation of innate immunity and chronic CsA-induced renal injury.

CsA-mediated impairment of urine concentration

CsA causes tubular dysfunction characterized by polyuria, calcium wasting, distal tubular acidosis, and hyperkalemia. Of these, impaired urine concentration is a predominant feature of chronic CsA nephrotoxicity [9], but the molecular mechanisms underlying this issue remain unknown.

In general, the urine-concentrating process is controlled by the aquaporin (AQP) system and the urea transporters (UTs) [98,99]. AQPs are a family of membrane proteins that play an important role in the reabsorption of water in the kidney. To date, at least 11 different types of AQPs have been cloned [98,100]. AQP1 aids in the rapid reabsorption of large quantities (70-80%) of filtered water and is constitutively present on both the apical and basolateral membranes of epithelial cells in the proximal tubule and the descending thin limbs of Henle's loop, and in endothelial cells of the descending vasa recta [101]. AQPs 2-4 are involved in the movement of water across the apical membrane in collecting duct principal cells and are localized to the basolateral membrane of this cell type. Reabsorbed free water determines the osmolality of the final voided urine, and this AQP-related process is controlled by vasopressin [98]. The UT family includes renal UT (UT-A) and the erythrocyte urea transporter (UT-B) [102]. In the kidney, UT-A1 and UT-A3 are responsible for the accumulation of urea in the inner medulla, and they are present in the inner medullary collecting duct. UT-A2 is present in the descending thin limbs of Henle's loop, and UT-B in the descending vasa recta [103-106]. We have demonstrated that chronic CsA treatment decreases AQP 1-4 and UT (UT-A2, UT-A3, and UT-B) production in rat kidneys, which may account for the impairment of urinary concentrating ability (polyuria, increased fractional excretion of sodium, and decreased free-water reabsorption) [107]. Recently, down-regulation of tonicity-responsive enhancer binding protein (TonEBP) has been proposed as a mediator of decreased AQP and UT transcription [108]. TonEBP is inhibited in the renal medulla secondary to reduced medullary tonicity, which is in turn the result of the down regulation of renal sodium transporters.

CsA-induced urinary calcium wasting

Another manifestation of renal tubular dysfunction caused by CsA is calcium wasting associated with the dysregulation of parathyroid hormone, which ultimately results in high bone turnover (osteopenia). Most of the filtered calcium (approximately 60%) is reabsorbed in the proximal tubule, and the remainder is reabsorbed in the medullary thick ascending limb of Henle's loop, the distal convoluted tubule, and the connecting segment [109]. The calcium binding protein calbindin plays an important role in calcium transport [110]. Two distinct subclasses of this protein with relative molecular masses of 9,000 and 28,000 Da have been recognized. Calbindin D9k is present in high concentrations in the proximal small intestine and facilitates intestinal calcium absorption [111]. Calbindin28k is expressed in the distal nephron segments of the rat kidney and is assumed to take part in calcium reabsorption [112]. Recent experimental studies by our laboratory [113] and by Steiner et al. [114] have demonstrated that chronic CsA treatment reduces calbindin28k immunoreactivity in the distal nephron, and this is accompanied by a significant decrease in serum calcium concentration and an increase in urinary calcium excretion. These results suggest that CsA-mediated suppression of calbindin28k production is a critical factor in renal calcium wasting.

Endoplasmic reticulum stress and autophagy

The endoplasmic reticulum (ER) has evolved a highly specific signaling pathway called the "unfolded protein response," a protective mechanism that reduces protein load and increases nascent protein folding, thus contributing to cell protection [115]. However, when ER stress is excessive and/or prolonged, the initiation of apoptosis is promoted [116,117]. Recently, Pallet et al. [118] demonstrated that CsA induces ER stress in renal tubular cells, which then activates autophagy as a protection against cell death [119]. Autophagy is a protective mechanism against various cellular stresses, including nutrient deprivation, hypoxia, and growth factor deprivation, and promotes cell survival. Recent evidence suggests that ER stress drives autophagy [120-122] and that autophagy alleviates ER stress and reduces cell death. We have demonstrated that prolonged ER stress induces apoptosis in an animal model of chronic CsA nephropathy [123]. Short-term treatment of CsA activated both the ER stress and proapoptotic responses, whereas long-term treatment of CsA decreased the ER stress response and increased the proapoptotic response. These results suggest that an imbalance between the two responses may cause apoptosis by depleting molecular chaperones and activating the proapoptotic pathway in chronic CsA-induced renal injury.

STRATEGIES TO PREVENT OR REDUCE CHRONIC CsA NEPHROPATHY

Reduction or withdrawal of CsA

Whether chronic CsA nephropathy is reversible is controversial. Traditionally, acute CsA nephropathy has been regarded as reversible after CsA dose reduction or complete withdrawal. However, chronic CsA-induced tubulointerstitial injury is considered to be persistent and, in some cases, progressive.

Weir et al. [124] investigated the long-term effects of the reduction or withdrawal of calcineurin inhibitors in a cohort of renal transplant recipients. After reduction or complete cessation of calcineurin inhibitors, a significant improvement in renal function was observed. Although an improvement in renal function was not recorded in all patients, this strategy was beneficial in most patients and reduced the rate of allograft loss in patients with nephrotoxicity. In low-salt diet-fed rats, Elzinga et al. [23] reported a progressive improvement in the GFR at 4 weeks after CsA discontinuation, but cortical fibrosis and tubular atrophy were unchanged. Using the same model, Franceschini et al. [125] carried out a long-term study in which the rats were administered CsA for 5 weeks, followed by the discontinuation of CsA for 2 or 8 weeks. The authors demonstrated that the degree of renal function and arteriolopathy was similar to that in placebo-treated controls after 8 weeks of CsA withdrawal, but tubulointerstitial fibrosis was not reversed. Therefore, there is dissociation between GFR and tissue injury in this animal model, and the low-salt diet is suggested to be an important factor.

To clarify the molecular mechanism of irreversible damage in chronic CsA nephropathy, we carried out two prolonged studies using two dosages of CsA (7.5 and 15 mg/kg) and multiple periods of drug washout [126]. Surprisingly, both renal function and morphological alterations (tubulointerstitial fibrosis and arteriolopathy) were reversed in rats with both dosages of CsA at 5 weeks after withdrawal, and the alleviation of tubulointerstitial fibrosis was observed after 10 weeks. At the molecular level, the reversibility of chronic CsA nephrotoxicity was closely associated with the down-regulation of OPN and TGF-β1 gene expression, the up-regulation of EGF production, and a decrease in renal cell apoptosis [82]. These conflicting results between our studies and previous reports lead us to speculate that the effect of CsA withdrawal in chronic CsA-induced renal injury may depend on the CsA dosage or on the timing of drug elimination.

Given the recently demonstrated association between prolonged ER stress and apoptosis [123], kidney allografts may well adapt to CsA-induced ER stress by inducing the expression of molecular chaperones. The duration and intensity of CsA-induced ER stress may provide a rationale for the reduction or withdrawal of CsA to prevent chronic CsA nephropathy. In addition, because the activation of autophagy indicates that cellular stress has occurred, the early detection of autophagy may facilitate the timely reduction or withdrawal of CsA treatment [119]. Clinical trials are needed to clarify these issues.

Pharmacologic intervention

Chronic CsA nephropathy is a major problem in transplant recipients and patients with autoimmune disorders, as it may lead to end-stage renal disease requiring dialysis. Therefore, renal function should always be carefully monitored, even in patients in whom renal function appears to be stable. In addition, early recognition and pharmacological intervention may be necessary to minimize the rate of allograft loss or chronic renal failure.

It is generally accepted that the modulation of vasoactive factors (Ang II and NO) effectively prevents CsA-induced renal injury. Previous studies clearly demonstrated that the concomitant administration of ACE inhibitors and Ang II type I receptor antagonists significantly reduced arteriolopathy, interstitial fibrosis, and tubular atrophy, independently of hemodynamic effects. However, renal dysfunction was not attenuated, and a potential explanation for this discrepancy has been discussed above. The molecular mechanisms underlying the renoprotective effects of these drugs in chronic CsA nephropathy may be related to their actions on inflammatory mediators, pro-fibrotic cytokines, matrix proteins, apoptotic cell death, and innate immunity [9,16,21,26-28,127). BQ123, an endothelin receptor blocker, partially improved renal function, but did not reduce structural damage [128]. Similarly, exogenous supplementation of L-arginine, a substrate for NO synthase, also conferred a renoprotective effect [38,40]. This effect was associated with the reduction of VEGF expression and the increased availability of NO.

Drugs with anti-fibrotic properties such as the TGF-β antibody [60], statins [129], pirfenidone [59,130], and hepatocyte growth factor [131] have been shown to have renoprotective effects in chronic CsA nephropathy. Statins are competitive inhibitors of 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase, the key enzyme that regulates the synthesis of cholesterol from mevalonic acid by suppressing the conversion of HMG-CoA [132]. The inhibition of HMG-CoA reductase by statins has pleiotropic effects independent of their lipid-lowering effects (e.g., anti-inflammatory and anti-arteriosclerotic effects) [133,134]. These benefits are mirrored in many renal disease models [135-138]. In a rat model of chronic CsA nephropathy, we showed that pravastatin treatment attenuated interstitial inflammation and fibrosis. The suppression of OPN, TGF-β1, and intrarenal C-reactive protein expression along with increased eNOS expression may be responsible for the renoprotective properties of pravastatin [129].

Anti-inflammatory drugs, such as pentosan polysulfate [139] and mycophenolate mofetil (MMF) [25], are also suggested to have protective effects in chronic CsA-induced renal injury. In a study by Schwedler et al. [139], pentosan polysulfate reduced tissue injury but was unable to improve renal function. We have reported that the administration of MMF is partially effective in preventing the development of chronic CsA nephropathy. Moreover, the combination of MMF and losartan (an Ang II type 1 receptor antagonist) or the administration of MMF after CsA withdrawal affords superior protection compared to losartan monotherapy [25] or CsA withdrawal alone [140].

Other strategies include vitamin E [14], recombinant human erythropoietin (rHuEPO) [141], rosiglitazone [142,143], or spironolactone [144,145] therapy. Vitamin E treatment is reported to preserve renal function and reduce free radical production, vasoconstrictive thromboxane levels, and tubulointerstitial fibrosis, probably by suppressing the production of superoxide anion, hydrogen peroxide, malonyldialdehyde, hemeoxygenase I, and some mediators (TGF-β1 and OPN) [14]. The preventive effect of rHuEPO on chronic CsA-induced renal injury is associated with its anti-apoptotic and anti-inflammatory properties [141]. The exogenous administration of rosiglitazone, a peroxisome proliferatoractivated receptor gamma agonist, has been shown to reduce chronic CsA-induced renal injury [142,143], but the mechanism underlying this effect remains unknown. The blockade of aldosterone receptors with spironolactone has been shown to ameliorate chronic CsA nephropathy [144,145]. Interestingly, spironolactone therapy not only attenuated structural damage, but also improved renal function, perhaps via the prevention of TGF-β and extracellular matrix protein over-expression and the reestablishment of renal blood flow.

CONCLUSION

To reduce or avoid chronic CsA-induced renal injury, new immunosuppressive agents and strategies for calcineurin inhibitor minimization, avoidance, or withdrawal have been emerging in the literature. These new strategies may ultimately minimize the incidence of nephrotoxicity and improve allograft and patient survival. Clinical trials are warranted to investigate the appropriate dosage of CsA and the effect of eliminating CsA with the addition of non-nephrotoxic immunosuppressants, such as MMF and sirolimus, which may permit optimal immunosuppression while avoiding the risk of acute rejection. However, CsA is still a major immunosuppressant currently used in both transplant and non-transplant patients. Considering the increasing rate of late allograft loss or chronic renal failure associated with chronic CsA nephropathy, a full understanding of the molecular mechanisms underlying chronic CsA-induced renal injury is essential to improve long-term outcomes and pharmacological interventions to delay the progression of chronic CsA nephropathy.

References

1. Cecka JM, Terasaki PI. The UNOS Scientific Renal Transplant Registry: 1991. Clin Transpl 1991;:1–11. 1820108.
2. de Mattos AM, Olyaei AJ, Bennett WM. Nephrotoxicity of immunosuppressive drugs: long-term consequences and challenges for the future. Am J Kidney Dis 2000;35:333–346. 10676738.
3. Erlanger BF. Do we know the site of action of cyclosporin? Immunol Today 1992;13:487–490. 1334415.
4. Shaw KT, Ho AM, Raghavan A, et al. Immunosuppressive drugs prevent a rapid dephosphorylation of transcription factor NFAT1 in stimulated immune cells. Proc Natl Acad Sci USA 1995;92:11205–11209. 7479966.
5. Suthanthiran M, Morris RE, Strom TB. Immunosuppressants: cellular and molecular mechanisms of action. Am J Kidney Dis 1996;28:159–172. 8768909.
6. Olyaei AJ, de Mattos AM, Bennett WM. Nephrotoxicity of immunosuppressive drugs: new insight and preventive strategies. Curr Opin Crit Care 2001;7:384–389. 11805539.
7. Mourad G, Vela C, Ribstein J, Mimran A. Long-term improvement in renal function after cyclosporine reduction in renal transplant recipients with histologically proven chronic cyclosporine nephropathy. Transplantation 1998;65:661–667. 9521200.
8. Myers BD, Sibley R, Newton L, et al. The long-term course of cyclosporine-associated chronic nephropathy. Kidney Int 1988;33:590–600. 3283402.
9. Young BA, Burdmann EA, Johnson RJ, et al. Cellular proliferation and macrophage influx precede interstitial fibrosis in cyclosporine nephrotoxicity. Kidney Int 1995;48:439–448. 7564111.
10. Young BA, Burdmann EA, Johnson RJ, et al. Cyclosporine A induced arteriolopathy in a rat model of chronic cyclosporine nephropathy. Kidney Int 1995;48:431–438. 7564110.
11. Nankivell BJ, Borrows RJ, Fung CL, O'Connell PJ, Allen RD, Chapman JR. The natural history of chronic allograft nephropathy. N Engl J Med 2003;349:2326–2333. 14668458.
12. Padi SS, Chopra K. Salvage of cyclosporine A-induced oxidative stress and renal dysfunction by carvedilol. Nephron 2002;92:685–692. 12372956.
13. Parra T, de Arriba G, Arribas I, Perez de Lema G, Rodriguez-Puyol D, Rodriguez-Puyol M. Cyclosporine A nephrotoxicity: role of thromboxane and reactive oxygen species. J Lab Clin Med 1998;131:63–70. 9452128.
14. Jenkins JK, Huang H, Ndebele K, Salahudeen AK. Vitamin E inhibits renal mRNA expression of COX II, HO I, TGFbeta, and osteopontin in the rat model of cyclosporine nephrotoxicity. Transplantation 2001;71:331–334. 11213082.
15. Parra T, de Arriba G, Conejo JR, et al. Cyclosporine increases local glomerular synthesis of reactive oxygen species in rats: effect of vitamin E on cyclosporine nephrotoxicity. Transplantation 1998;66:1325–1329. 9846517.
16. Burdmann EA, Andoh TF, Nast CC, et al. Prevention of experimental cyclosporin-induced interstitial fibrosis by losartan and enalapril. Am J Physiol 1995;269:F491–F499. 7485533.
17. Dell K, Bohler T, Gaedeke J, Budde K, Neumayer HH, Waiser J. Impact of PGE1 on cyclosporine A induced up-regulation of TGF-beta1, its receptors, and related matrix production in cultured mesangial cells. Cytokine 2003;22:189–193. 12890451.
18. Ramirez C, Olmo A, O'Valle F, et al. Role of intrarenal endothelin 1, endothelin 3, and angiotensin II expression in chronic cyclosporin A nephrotoxicity in rats. Exp Nephrol 2000;8:161–172. 10810233.
19. Elzinga LW, Rosen S, Burdmann EA, Hatton DC, Lindsley J, Bennett WM. The role of renal sympathetic nerves in experimental chronic cyclosporine nephropathy. Transplantation 2000;69:2149–2153. 10852615.
20. Lassila M. Interaction of cyclosporine A and the renin-angiotensin system: new perspectives. Curr Drug Metab 2002;3:61–71. 11876576.
21. Yang CW, Ahn HJ, Kim WY, et al. Influence of the renin-angiotensin system on epidermal growth factor expression in normal and cyclosporine-treated rat kidney. Kidney Int 2001;60:847–857. 11532080.
22. Rosen S, Greenfeld Z, Brezis M. Chronic cyclosporine-induced nephropathy in the rat: a medullary ray and inner stripe injury. Transplantation 1990;49:445–452. 2305470.
23. Elzinga LW, Rosen S, Bennett WM. Dissociation of glomerular filtration rate from tubulointerstitial fibrosis in experimental chronic cyclosporine nephropathy: role of sodium intake. J Am Soc Nephrol 1993;4:214–221. 8400085.
24. Mazzali M, Kim YG, Suga S, et al. Hyperuricemia exacerbates chronic cyclosporine nephropathy. Transplantation 2001;71:900–905. 11349724.
25. Yang CW, Ahn HJ, Kim WY, et al. Synergistic effects of mycophenolate mofetil and losartan in a model of chronic cyclosporine nephropathy. Transplantation 2003;75:309–315. 12589150.
26. Thomas SE, Andoh TF, Pichler RH, et al. Accelerated apoptosis characterizes cyclosporine-associated interstitial fibrosis. Kidney Int 1998;53:897–908. 9551396.
27. Shihab FS, Bennett WM, Tanner AM, Andoh TF. Angiotensin II blockade decreases TGF-beta1 and matrix proteins in cyclosporine nephropathy. Kidney Int 1997;52:660–673. 9291185.
28. Pichler RH, Franceschini N, Young BA, et al. Pathogenesis of cyclosporine nephropathy: roles of angiotensin II and osteopontin. J Am Soc Nephrol 1995;6:1186–1196. 8589285.
29. Li C, Yang CW, Park CW, et al. Long-term treatment with ramipril attenuates renal osteopontin expression in diabetic rats. Kidney Int 2003;63:454–463. 12631111.
30. Kuro-o M, Matsumura Y, Aizawa H, et al. Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature 1997;390:45–51. 9363890.
31. Koh N, Fujimori T, Nishiguchi S, et al. Severely reduced production of klotho in human chronic renal failure kidney. Biochem Biophys Res Commun 2001;280:1015–1020. 11162628.
32. Sugiura H, Yoshida T, Tsuchiya K, et al. Klotho reduces apoptosis in experimental ischaemic acute renal failure. Nephrol Dial Transplant 2005;20:2636–2645. 16204278.
33. Kone BC, Baylis C. Biosynthesis and homeostatic roles of nitric oxide in the normal kidney. Am J Physiol 1997;272:F561–F578. 9176366.
34. Diederich D, Yang Z, Luscher TF. Chronic cyclosporine therapy impairs endothelium-dependent relaxation in the renal artery of the rat. J Am Soc Nephrol 1992;2:1291–1297. 1627754.
35. Stroes ES, Luscher TF, de Groot FG, Koomans HA, Rabelink TJ. Cyclosporin A increases nitric oxide activity in vivo. Hypertension 1997;29:570–575. 9040440.
36. Takenaka T, Hashimoto Y, Epstein M. Diminished acetylcholine-induced vasodilation in renal microvessels of cyclosporine-treated rats. J Am Soc Nephrol 1992;3:42–50. 1391707.
37. Bobadilla NA, Gamba G, Tapia E, et al. Role of NO in cyclosporin nephrotoxicity: effects of chronic NO inhibition and NO synthases gene expression. Am J Physiol 1998;274:F791–F798. 9575905.
38. Yang CW, Kim YS, Kim J, et al. Oral supplementation of L-arginine prevents chronic cyclosporine nephrotoxicity in rats. Exp Nephrol 1998;6:50–56. 9523173.
39. Shihab FS, Bennett WM, Isaac J, Yi H, Andoh TF. Nitric oxide modulates vascular endothelial growth factor and receptors in chronic cyclosporine nephrotoxicity. Kidney Int 2003;63:522–533. 12631117.
40. Shihab FS, Yi H, Bennett WM, Andoh TF. Effect of nitric oxide modulation on TGF-beta1 and matrix proteins in chronic cyclosporine nephrotoxicity. Kidney Int 2000;58:1174–1185. 10972680.
41. Butler WT. Structural and functional domains of osteopontin. Ann N Y Acad Sci 1995;760:6–11. 7785926.
42. Brown LF, Papadopoulos-Sergiou A, Berse B, et al. Osteopontin expression and distribution in human carcinomas. Am J Pathol 1994;145:610–623. 8080043.
43. Giachelli C, Bae N, Lombardi D, Majesky M, Schwartz S. Molecular cloning and characterization of 2B7, a rat mRNA which distinguishes smooth muscle cell phenotypes in vitro and is identical to osteopontin (secreted phosphoprotein I, 2aR). Biochem Biophys Res Commun 1991;177:867–873. 1828666.
44. Giachelli CM, Liaw L, Murry CE, Schwartz SM, Almeida M. Osteopontin expression in cardiovascular diseases. Ann N Y Acad Sci 1995;760:109–126. 7785890.
45. Hudkins KL, Giachelli CM, Cui Y, Couser WG, Johnson RJ, Alpers CE. Osteopontin expression in fetal and mature human kidney. J Am Soc Nephrol 1999;10:444–457. 10073594.
46. Lopez CA, Hoyer JR, Wilson PD, Waterhouse P, Denhardt DT. Heterogeneity of osteopontin expression among nephrons in mouse kidneys and enhanced expression in sclerotic glomeruli. Lab Invest 1993;69:355–363. 8377476.
47. Patarca R, Freeman GJ, Singh RP, et al. Structural and functional studies of the early T lymphocyte activation 1 (Eta-1) gene: definition of a novel T cell-dependent response associated with genetic resistance to bacterial infection. J Exp Med 1989;170:145–161. 2787378.
48. Hu DD, Lin EC, Kovach NL, Hoyer JR, Smith JW. A biochemical characterization of the binding of osteopontin to integrins alpha v beta 1 and alpha v beta 5. J Biol Chem 1995;270:26232–26238. 7592829.
49. Weber GF, Ashkar S, Glimcher MJ, Cantor H. Receptor-ligand interaction between CD44 and osteopontin (Eta-1). Science 1996;271:509–512. 8560266.
50. Singh RP, Patarca R, Schwartz J, Singh P, Cantor H. Definition of a specific interaction between the early T lymphocyte activation 1 (Eta-1) protein and murine macrophages in vitro and its effect upon macrophages in vivo. J Exp Med 1990;171:1931–1942. 2351930.
51. Ophascharoensuk V, Giachelli CM, Gordon K, et al. Obstructive uropathy in the mouse: role of osteopontin in interstitial fibrosis and apoptosis. Kidney Int 1999;56:571–580. 10432396.
52. Magil AB, Pichler RH, Johnson RJ. Osteopontin in chronic puromycin aminonucleoside nephrosis. J Am Soc Nephrol 1997;8:1383–1390. 9294829.
53. Pichler R, Giachelli CM, Lombardi D, et al. Tubulointerstitial disease in glomerulonephritis: potential role of osteopontin (uropontin). Am J Pathol 1994;144:915–926. 8178943.
54. Wuthrich RP, Fan X, Ritthaler T, et al. Enhanced osteopontin expression and macrophage infiltration in MRL-Fas(lpr) mice with lupus nephritis. Autoimmunity 1998;28:139–150. 9867126.
55. Yu XQ, Wu LL, Huang XR, et al. Osteopontin expression in progressive renal injury in remnant kidney: role of angiotensin II. Kidney Int 2000;58:1469–1480. 11012882.
56. Li C, Yang CW, Ahn HJ, et al. Colchicine suppresses osteopontin expression and inflammatory cell infiltration in chronic cyclosporine nephrotoxicity. Nephron 2002;92:422–430. 12218323.
57. Mazzali M, Hughes J, Dantas M, et al. Effects of cyclosporine in osteopontin null mice. Kidney Int 2002;62:78–85. 12081566.
58. Shihab FS, Andoh TF, Tanner AM, Bennett WM. Sodium depletion enhances fibrosis and the expression of TGF-beta1 and matrix proteins in experimental chronic cyclosporine nephropathy. Am J Kidney Dis 1997;30:71–81. 9214404.
59. Shihab FS, Bennett WM, Yi H, Andoh TF. Pirfenidone treatment decreases transforming growth factor-beta1 and matrix proteins and ameliorates fibrosis in chronic cyclosporine nephrotoxicity. Am J Transplant 2002;2:111–119. 12099512.
60. Ling H, Li X, Jha S, et al. Therapeutic role of TGF-beta-neutralizing antibody in mouse cyclosporin A nephropathy: morphologic improvement associated with functional preservation. J Am Soc Nephrol 2003;14:377–388. 12538738.
61. O'Connor-McCourt MD, Wakefield LM. Latent transforming growth factor-beta in serum: a specific complex with alpha 2-macroglobulin. J Biol Chem 1987;262:14090–14099. 2443501.
62. Oreffo RO, Mundy GR, Seyedin SM, Bonewald LF. Activation of the bone-derived latent TGF beta complex by isolated osteoclasts. Biochem Biophys Res Commun 1989;158:817–823. 2920041.
63. Schultz-Cherry S, Chen H, Mosher DF, et al. Regulation of transforming growth factor-beta activation by discrete sequences of thrombospondin 1. J Biol Chem 1995;270:7304–7310. 7706271.
64. Sharma K, Ziyadeh FN. The transforming growth factor-beta system and the kidney. Semin Nephrol 1993;13:116–128. 8381979.
65. Yamaguchi Y, Mann DM, Ruoslahti E. Negative regulation of transforming growth factor-beta by the proteoglycan decorin. Nature 1990;346:281–284. 2374594.
66. Skonier J, Neubauer M, Madisen L, Bennett K, Plowman GD, Purchio AF. cDNA cloning and sequence analysis of beta ig-h3, a novel gene induced in a human adenocarcinoma cell line after treatment with transforming growth factor-beta. DNA Cell Biol 1992;11:511–522. 1388724.
67. Billings PC, Whitbeck JC, Adams CS, et al. The transforming growth factor-beta-inducible matrix protein (beta)ig-h3 interacts with fibronectin. J Biol Chem 2002;277:28003–28009. 12034705.
68. Billings PC, Herrick DJ, Kucich U, et al. Extracellular matrix and nuclear localization of beta ig-h3 in human bladder smooth muscle and fibroblast cells. J Cell Biochem 2000;79:261–273. 10967553.
69. Gibson MA, Kumaratilake JS, Cleary EG. Immunohistochemical and ultrastructural localization of MP78/70 (betaig-h3) in extracellular matrix of developing and mature bovine tissues. J Histochem Cytochem 1997;45:1683–1696. 9389772.
70. O'Brien ER, Bennett KL, Garvin MR, et al. Beta ig-h3, a transforming growth factor-beta-inducible gene, is overexpressed in atherosclerotic and restenotic human vascular lesions. Arterioscler Thromb Vasc Biol 1996;16:576–584. 8624780.
71. Langham RG, Egan MK, Dowling JP, Gilbert RE, Thomson NM. Transforming growth factor-beta1 and tumor growth factor-beta-inducible gene-H3 in nonrenal transplant cyclosporine nephropathy. Transplantation 2001;72:1826–1829. 11740395.
72. Sun BK, Li C, Lim SW, et al. Expression of transforming growth factor-beta-inducible gene-h3 in normal and cyclosporine-treated rat kidney. J Lab Clin Med 2004;143:175–183. 15007308.
73. Yang CW, Faulkner GR, Wahba IM, et al. Expression of apoptosis-related genes in chronic cyclosporine nephrotoxicity in mice. Am J Transplant 2002;2:391–399. 12123203.
74. Ito H, Kasagi N, Shomori K, Osaki M, Adachi H. Apoptosis in the human allografted kidney: analysis by terminal deoxynucleotidyl transferase-mediated DUTP-botin nick end labeling. Transplantation 1995;60:794–798. 7482737.
75. Ortiz A, Lorz C, Catalan MP, Justo P, Egido J. Role and regulation of apoptotic cell death in the kidney: Y2K update. Front Biosci 2000;5:D735–D749. 10922294.
76. Li C, Yang CW, Ahn HJ, et al. Colchicine decreases apoptotic cell death in chronic cyclosporine nephrotoxicity. J Lab Clin Med 2002;139:364–371. 12066135.
77. Ying S, Khan LN, Meng Q, Barnes NC, Kay AB. Cyclosporin A, apoptosis of BAL T-cells and expression of Bcl-2 in asthmatics. Eur Respir J 2003;22:207–212. 12952249.
78. Healy E, Dempsey M, Lally C, Ryan MP. Apoptosis and necrosis: mechanisms of cell death induced by cyclosporine A in a renal proximal tubular cell line. Kidney Int 1998;54:1955–1966. 9853260.
79. Ortiz A, Lorz C, Catalan M, Coca S, Egido J. Cyclosporine A induces apoptosis in murine tubular epithelial cells: role of caspases. Kidney Int Suppl 1998;68:S25–S29. 9839279.
80. Shihab FS, Andoh TF, Tanner AM, Yi H, Bennett WM. Expression of apoptosis regulatory genes in chronic cyclosporine nephrotoxicity favors apoptosis. Kidney Int 1999;56:2147–2159. 10594790.
81. Amore A, Emancipator SN, Cirina P, et al. Nitric oxide mediates cyclosporine-induced apoptosis in cultured renal cells. Kidney Int 2000;57:1549–1559. 10760090.
82. Li C, Lim SW, Sun BK, et al. Expression of apoptosis-related factors in chronic cyclosporine nephrotoxicity after cyclosporine withdrawal. Acta Pharmacol Sin 2004;25:401–411. 15066204.
83. Ghee JY, Han DH, Song HK, et al. The role of macrophage in the pathogenesis of chronic cyclosporine-induced nephropathy. Nephrol Dial Transplant 2008;23:4061–4069. 18622021.
84. Guijarro C, Egido J. Transcription factor-kappa B (NF-kappa B) and renal disease. Kidney Int 2001;59:415–424. 11168923.
85. Barnes PJ, Karin M. Nuclear factor-kappaB: a pivotal transcription factor in chronic inflammatory diseases. N Engl J Med 1997;336:1066–1071. 9091804.
86. Karin M, Liu Z, Zandi E. AP-1 function and regulation. Curr Opin Cell Biol 1997;9:240–246. 9069263.
87. Tamada S, Nakatani T, Asai T, et al. Inhibition of nuclear factor-kappaB activation by pyrrolidine dithiocarbamate prevents chronic FK506 nephropathy. Kidney Int 2003;63:306–314. 12472797.
88. Klahr S, Morrissey JJ. The role of vasoactive compounds, growth factors and cytokines in the progression of renal disease. Kidney Int Suppl 2000;75:S7–S14. 10828755.
89. Mezzano SA, Barria M, Droguett MA, et al. Tubular NF-kappaB and AP-1 activation in human proteinuric renal disease. Kidney Int 2001;60:1366–1377. 11576350.
90. Ruiz-Ortega M, Bustos C, Hernandez-Presa MA, Lorenzo O, Plaza JJ, Egido J. Angiotensin II participates in mononuclear cell recruitment in experimental immune complex nephritis through nuclear factor-kappa B activation and monocyte chemoattractant protein-1 synthesis. J Immunol 1998;161:430–439. 9647253.
91. Asai T, Nakatani T, Tamada S, et al. Activation of transcription factors AP-1 and NF-kappaB in chronic cyclosporine A nephrotoxicity: role in beneficial effects of magnesium supplementation. Transplantation 2003;75:1040–1044. 12698095.
92. Lim SW, Li C, Ahn KO, et al. Cyclosporine-induced renal injury induces toll-like receptor and maturation of dendritic cells. Transplantation 2005;80:691–699. 16177646.
93. Akira S, Takeda K, Kaisho T. Toll-like receptors: critical proteins linking innate and acquired immunity. Nat Immunol 2001;2:675–680. 11477402.
94. Janeway CA Jr, Medzhitov R. Innate immune recognition. Annu Rev Immunol 2002;20:197–216. 11861602.
95. Anders HJ, Banas B, Schlondorff D. Signaling danger: toll-like receptors and their potential roles in kidney disease. J Am Soc Nephrol 2004;15:854–867. 15034087.
96. Kumaraguru U, Pack CD, Rouse BT. Toll-like receptor ligand links innate and adaptive immune responses by the production of heat-shock proteins. J Leukoc Biol 2003;73:574–583. 12714571.
97. Vabulas RM, Ahmad-Nejad P, Ghose S, Kirschning CJ, Issels RD, Wagner H. HSP70 as endogenous stimulus of the Toll/interleukin-1 receptor signal pathway. J Biol Chem 2002;277:15107–15112. 11842086.
98. Nielsen S, Kwon TH, Christensen BM, Promeneur D, Frokiaer J, Marples D. Physiology and pathophysiology of renal aquaporins. J Am Soc Nephrol 1999;10:647–663. 10073616.
99. Wintour EM. Water channels and urea transporters. Clin Exp Pharmacol Physiol 1997;24:1–9. 9043798.
100. Kishore BK, Krane CM, Di Iulio D, Menon AG, Cacini W. Expression of renal aquaporins 1, 2, and 3 in a rat model of cisplatin-induced polyuria. Kidney Int 2000;58:701–711. 10916093.
101. Nielsen S, Frokiaer J, Marples D, Kwon TH, Agre P, Knepper MA. Aquaporins in the kidney: from molecules to medicine. Physiol Rev 2002;82:205–244. 11773613.
102. Sands JM. Regulation of renal urea transporters. J Am Soc Nephrol 1999;10:635–646. 10073615.
103. Bradford AD, Terris JM, Ecelbarger CA, et al. 97- and 117-kDa forms of collecting duct urea transporter UT-A1 are due to different states of glycosylation. Am J Physiol Renal Physiol 2001;281:F133–F143. 11399654.
104. Terris JM, Knepper MA, Wade JB. UT-A3: localization and characterization of an additional urea transporter isoform in the IMCD. Am J Physiol Renal Physiol 2001;280:F325–F332. 11208608.
105. Timmer RT, Klein JD, Bagnasco SM, et al. Localization of the urea transporter UT-B protein in human and rat erythrocytes and tissues. Am J Physiol Cell Physiol 2001;281:C1318–C1325. 11546670.
106. Wade JB, Lee AJ, Liu J, et al. UT-A2: a 55-kDa urea transporter in thin descending limb whose abundance is regulated by vasopressin. Am J Physiol Renal Physiol 2000;278:F52–F62. 10644655.
107. Lim SW, Li C, Sun BK, et al. Long-term treatment with cyclosporine decreases aquaporins and urea transporters in the rat kidney. Am J Physiol Renal Physiol 2004;287:F139–F151. 14871880.
108. Lim SW, Ahn KO, Sheen MR, et al. Downregulation of renal sodium transporters and tonicity-responsive enhancer binding protein by long-term treatment with cyclosporin A. J Am Soc Nephrol 2007;18:421–429. 17202415.
109. Stier CT Jr, Itskovitz HD. Renal calcium metabolism and diuretics. Annu Rev Pharmacol Toxicol 1986;26:101–116. 3521452.
110. Gross M, Kumar R. Physiology and biochemistry of vitamin D-dependent calcium binding proteins. Am J Physiol 1990;259:F195–F209. 2201202.
111. Staun M, Sjostrom H, Noren O. Calcium-binding protein from human small intestine: purification and characterization of a 10,000 molecular weight protein. Eur J Clin Invest 1986;16:468–472. 3104046.
112. Roth J, Brown D, Norman AW, Orci L. Localization of the vitamin D-dependent calcium-binding protein in mammalian kidney. Am J Physiol 1982;243:F243–F252. 7051856.
113. Yang CW, Kim J, Kim YH, et al. Inhibition of calbindin D28K expression by cyclosporin A in rat kidney: the possible pathogenesis of cyclosporin A-induced hypercalciuria. J Am Soc Nephrol 1998;9:1416–1426. 9697663.
114. Steiner S, Aicher L, Raymackers J, et al. Cyclosporine A decreases the protein level of the calcium-binding protein calbindin-D 28kDa in rat kidney. Biochem Pharmacol 1996;51:253–258. 8573191.
115. Zhang K, Kaufman RJ. The unfolded protein response: a stress signaling pathway critical for health and disease. Neurology 2006;66(2)Suppl 1. :S102–S109. 16432136.
116. Okada K, Minamino T, Tsukamoto Y, et al. Prolonged endoplasmic reticulum stress in hypertrophic and failing heart after aortic constriction: possible contribution of endoplasmic reticulum stress to cardiac myocyte apoptosis. Circulation 2004;110:705–712. 15289376.
117. Oyadomari S, Araki E, Mori M. Endoplasmic reticulum stress-mediated apoptosis in pancreatic beta-cells. Apoptosis 2002;7:335–345. 12101393.
118. Pallet N, Bouvier N, Bendjallabah A, et al. Cyclosporine-induced endoplasmic reticulum stress triggers tubular phenotypic changes and death. Am J Transplant 2008;8:2283–2296. 18785955.
119. Pallet N, Bouvier N, Legendre C, et al. Autophagy protects renal tubular cells against cyclosporine toxicity. Autophagy 2008;4:783–791. 18628650.
120. Ding WX, Ni HM, Gao W, et al. Linking of autophagy to ubiquitin-proteasome system is important for the regulation of endoplasmic reticulum stress and cell viability. Am J Pathol 2007;171:513–524. 17620365.
121. Hoyer-Hansen M, Jaattela M. Connecting endoplasmic reticulum stress to autophagy by unfolded protein response and calcium. Cell Death Differ 2007;14:1576–1582. 17612585.
122. Moretti L, Cha YI, Niermann KJ, Lu B. Switch between apoptosis and autophagy: radiation-induced endoplasmic reticulum stress? Cell Cycle 2007;6:793–798. 17377498.
123. Han SW, Li C, Ahn KO, et al. Prolonged endoplasmic reticulum stress induces apoptotic cell death in an experimental model of chronic cyclosporine nephropathy. Am J Nephrol 2008;28:707–714. 18434710.
124. Weir MR, Ward MT, Blahut SA, et al. Long-term impact of discontinued or reduced calcineurin inhibitor in patients with chronic allograft nephropathy. Kidney Int 2001;59:1567–1573. 11260422.
125. Franceschini N, Alpers CE, Bennett WM, Andoh TF. Cyclosporine arteriolopathy: effects of drug withdrawal. Am J Kidney Dis 1998;32:247–253. 9708608.
126. Li C, Yang CW, Kim WY, et al. Reversibility of chronic cyclosporine nephropathy in rats after withdrawal of cyclosporine. Am J Physiol Renal Physiol 2003;284:F389–F398. 12529276.
127. Ahn KO, Lim SW, Li C, et al. Influence of angiotensin II on expression of toll-like receptor 2 and maturation of dendritic cells in chronic cyclosporine nephropathy. Transplantation 2007;83:938–947. 17460566.
128. Hunley TE, Fogo A, Iwasaki S, Kon V. Endothelin A receptor mediates functional but not structural damage in chronic cyclosporine nephrotoxicity. J Am Soc Nephrol 1995;5:1718–1723. 7780063.
129. Li C, Yang CW, Park JH, et al. Pravastatin treatment attenuates interstitial inflammation and fibrosis in a rat model of chronic cyclosporine-induced nephropathy. Am J Physiol Renal Physiol 2004;286:F46–F57. 14519596.
130. Shihab FS, Bennett WM, Yi H, Andoh TF. Effect of pirfenidone on apoptosis-regulatory genes in chronic cyclosporine nephrotoxicity. Transplantation 2005;79:419–426. 15729167.
131. Mizui M, Isaka Y, Takabatake Y, et al. Electroporation-mediated HGF gene transfer ameliorated cyclosporine nephrotoxicity. Kidney Int 2004;65:2041–2053. 15149317.
132. Goldstein JL, Brown MS. Regulation of the mevalonate pathway. Nature 1990;343:425–430. 1967820.
133. Faggiotto A, Paoletti R. State-of-the-Art lecture: statins and blockers of the renin-angiotensin system: vascular protection beyond their primary mode of action. Hypertension 1999;34:987–996. 10523396.
134. Rosenson RS, Tangney CC. Antiatherothrombotic properties of statins: implications for cardiovascular event reduction. JAMA 1998;279:1643–1650. 9613915.
135. Joyce M, Kelly C, Winter D, Chen G, Leahy A, Bouchier-Hayes D. Pravastatin, a 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor, attenuates renal injury in an experimental model of ischemia-reperfusion. J Surg Res 2001;101:79–84. 11676559.
136. Kim SI, Han DC, Lee HB. Lovastatin inhibits transforming growth factor-beta1 expression in diabetic rat glomeruli and cultured rat mesangial cells. J Am Soc Nephrol 2000;11:80–87. 10616843.
137. Harris KP, Purkerson ML, Yates J, Klahr S. Lovastatin ameliorates the development of glomerulosclerosis and uremia in experimental nephrotic syndrome. Am J Kidney Dis 1990;15:16–23. 2294729.
138. Moriyama T, Kawada N, Nagatoya K, et al. Fluvastatin suppresses oxidative stress and fibrosis in the interstitium of mouse kidneys with unilateral ureteral obstruction. Kidney Int 2001;59:2095–2103. 11380811.
139. Schwedler SB, Bobadilla N, Striker LJ, Vaamonde CA, Herrera-Acosta J, Striker GE. Pentosan polysulfate treatment reduces cyclosporine-induced nephropathy in salt-depleted rats. Transplantation 1999;68:1583–1588. 10589959.
140. Yang CW, Ahn HJ, Kim WY, et al. Cyclosporine withdrawal and mycophenolate mofetil treatment effects on the progression of chronic cyclosporine nephrotoxicity. Kidney Int 2002;62:20–30. 12081560.
141. Lee SH, Li C, Lim SW, et al. Attenuation of interstitial inflammation and fibrosis by recombinant human erythropoietin in chronic cyclosporine nephropathy. Am J Nephrol 2005;25:64–76. 15746540.
142. Ahn KO, Lim SW, Yang HJ, et al. Induction of PPAR gamma mRNA and protein expression by rosiglitazone in chronic cyclosporine nephropathy in the rat. Yonsei Med J 2007;48:308–316. 17461532.
143. Chung BH, Li C, Sun BK, et al. Rosiglitazone protects against cyclosporine-induced pancreatic and renal injury in rats. Am J Transplant 2005;5:1856–1867. 15996232.
144. Feria I, Pichardo I, Juarez P, et al. Therapeutic benefit of spironolactone in experimental chronic cyclosporine A nephrotoxicity. Kidney Int 2003;63:43–52. 12472767.
145. Perez-Rojas JM, Derive S, Blanco JA, et al. Renocortical mRNA expression of vasoactive factors during spironolactone protective effect in chronic cyclosporine nephrotoxicity. Am J Physiol Renal Physiol 2005;289:F1020–F1030. 15998842.

Article information Continued

Figure 1

A schematic diagram of the pathogenesis of chronic CsA-induced renal injury. Chronic treatment with CsA induces renal injury by immunologic and non-immunologic pathways. The non-immunologic pathway includes hemodynamic (low-grade ischemia) and non-hemodynamic (mediators) mechanisms, although both ultimately result in tubulointerstitial inflammation, striped fibrosis, and an increased rate of chronic rejection. The immunologic pathway may be triggered by the endogenous production of HSP 70, a known activator of TLRs. Consequently, activation of the innate immune system by TLRs and MHC class II molecules leads to the activation of NF-κB and AP-1.