Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Nov 1.
Published in final edited form as: Am J Kidney Dis. 2015 Aug 10;66(5):846–856. doi: 10.1053/j.ajkd.2015.06.026

Remote Ischemic Preconditioning for Kidney Protection: GSK3β-Centric Insights Into the Mechanism of Action

Zhangsuo Liu 1, Rujun Gong 2
PMCID: PMC4623948  NIHMSID: NIHMS711803  PMID: 26271146

Abstract

Preventing acute kidney injury (AKI) in high-risk patients following medical interventions is a paramount challenge for clinical practice. Recent data from animal experiments and clinical trials indicate that remote ischemic preconditioning (IPC), represented by limb IPC, confers a protective action on the kidney. IPC is effective in reducing the risk of AKI following cardiovascular interventions and the use of iodinated radiocontrast media. Nevertheless, the underlying mechanisms for this protective effect remain elusive. A protective signal is conveyed from the remote site undergoing IPC, like the limb, to target organs, like the kidney, via multiple potential communication pathways, which may involve a humoral, neuronal, and systemic mechanisms. Diverse transmitting pathways trigger a variety of signaling cascades, including the reperfusion injury salvage kinase and survivor activating factor enhancement pathways, all of which converge on glycogen synthase kinase (GSK)3β. Inhibition of GSK3β subsequent to IPC reinforces the Nrf2-mediated antioxidant defense, diminishes the NFκB-dependent pro-inflammatory response, and exerts prosurvival effects ensuing from the desensitized mitochondria permeability transition. Thus, therapeutic targeting of GSK3β by IPC or by pharmacologic preconditioning with existing FDA-approved drugs having GSK3β inhibitory activities might represent a pragmatic and cost-effective adjuvant strategy for kidney protection and prophylaxis against AKI.

Keywords: acute kidney injury (AKI), remote ischemic preconditioning (IPC), limb IPC, ischemia/reperfusion, glycogen synthase kinase 3β (GSK3β), NFκB, Nrf2, inflammation, mitochondria permeability transition (MPT), lithium, renal protection, review

BACKGROUND

Acute kidney injury (AKI) is a common and potentially life-threatening complication annually affecting about 2,000 to 3,000 per million population, with two-thirds of cases occurring in intensive care units1,2. Common causes of AKI include sepsis, ischemia/reperfusion injury, trauma, and exposure to nephrotoxic agents1,2. In examining the epidemiologic risk factors, it is apparent that patients with older age, hypovolemia, heart dysfunction, preexisting chronic kidney disease (CKD), and exposure to nephrotoxic medications are more susceptible to developing AKI following diagnostic or therapeutic medical interventions (i.e. iatrogenic AKI)1. Specific medical interventions linked to AKI include intravascular use of iodinated radiocontrast media, administration of nephrotoxic medications, and major surgeries involving cardiopulmonary bypass (CPB) or aortic cross clamping1,2.

In the past 30 years, clinical management of AKI has been principally confined to treatment of symptoms and general supportive care including fluid resuscitation and kidney replacement therapy. These treatments are, however, of limited utility with unsatisfying therapeutic efficacy, given the poor prognosis. Although only a minority of patients with AKI requires hemodialysis after their initial hospital discharge, long-term follow-up studies show that the number of patients whose kidney recovery is incomplete has been underestimated and that AKI per se is an independent risk factor for subsequent transition to CKD24. Therefore, it is imperative to develop a novel, pragmatic, and effective therapy for prophylaxis against AKI in these susceptible patients. Recently, a burgeoning body of evidence from both experimental and clinical studies points to ischemic preconditioning (IPC) as a promising and feasible approach to kidney protection and prophylaxis against AKI5.

CASE VIGNETTE

A 65-year-old man with a history of diabetes and hypertension for over 30 years presented to the emergency room with unstable angina pectoris. Laboratory testing revealed an elevated level of cardiac enzymes and serum creatinine level of 2.1 mg/dL (186 μmol/L; corresponding to an estimated glomerular filtration rate [eGFR] of 32 mL/min/1.73 m2 as calculated using the CKD-EPI creatinine equation6), consistent with stage 3 CKD. Urinalysis demonstrated an albumin-creatinine ratio of 2.6 mg/mg. The patient underwent urgent coronary angiogram, which revealed 90% stenosis of right coronary artery (RCA) and 75% stenosis of left anterior descending branch. An attempt at percutaneous coronary angioplasty of the RCA failed. The patient was referred for surgical coronary artery bypass grafting (CABG) with CPB but was considered to be a poor candidate for surgery because of high risk of AKI (risk score of 8 using the Thakar model of dialysis risk after cardiac surgery7). The patient was subsequently maintained on nonsurgical treatments, including insulin, furosemide, valsartan, metopralol, amlodipine, acetyl salicylic acid, and lovastatin.

Although not currently standard of care, remote IPC may prove very helpful for patients like the one presented above. In future clinical practice, the approach to this patient might change. After induction of anesthesia for CABG surgery, this patient might undergo 4 cycles of a 5-minute period of upper arm ischemia, brought about placing a 9-cm blood pressure cuff around the upper arm and inflating it to a pressure 503mm3Hg greater than his systolic blood pressure. Each period of ischemia would be followed by a 5-minute period of reperfusion induced by deflation of the blood pressure cuff. Remote IPC would occur in the anesthetic room during patient monitoring and placement of intravascular and bladder catheters. Immediately after the remote IPC protocol is completed, the patient would undergo CABG surgery with a significant reduction in the risk of AKI.

PATHOGENESIS

Ischemic preconditioning (IPC) is an innate tissue adaptation, whereby brief episodes of ischemic insult to a tissue or solid organ make both local and remote organs more resistant to a later prolonged exposure to the same or other injuries8. The concept of IPC was first advanced in 1986 by Murry et al9, who described a protective effect of repeated brief episodes of coronary artery ischemia/reperfusion on a subsequent myocardial infarction induced by sustained occlusion of the coronary artery in dogs. Before the introduction of IPC, Zager et al10 had already made a similar observation in the kidney, namely that in rats, prior exposure of the kidney to ischemia confers protection against additional ischemic kidney insults. In fact, it has been known for almost a century that a previous sublethal heavy metal exposure renders the kidney resistant to the additional injury11.

More recently, studies have demonstrated that brief ischemia imposed even on non-target organs, most commonly in the limbs, exerts a protective effect on remote solid organs (eg, heart, lung, kidney or intestine) against injuries associated with ischemia and with other insults, including toxicants, hemorrhagic shock/resuscitation, and iodinated radiocontrast media12. Limb IPC achieved by cycling between inflating and deflating blood pressure cuffs on either the arm or leg is a cost-free, straightforward, and attractive protocol. This technique has been reproducibly shown to attenuate acute injuries in the heart and in other organs, including the kidney13. A meta-analysis by Wever et al14 examined a total of 58 experimental studies on the effect of IPC (including limb IPC) in animal models of AKI and concluded that IPC has a protective effect on AKI induced by ischemia reperfusion injury. IPC successfully prevents reductions in kidney function as assessed by serum creatinine and serum urea nitrogen and it also minimizes kidney histological damage. Moreover, the kidney-protective properties of IPC seem to be most effective in animals when the IPC stimulus is applied 24 hours before the ischemic injury (late window of protection)14.

In an effort to evaluate the safety and effectiveness of IPC in humans, 13 randomized controlled trials (1,334 participants) have been completed so far in patients at risk of AKI following cardiac or vascular interventions. In 11 of the 13 trials (1,216 participants), limb IPC was found to reduce the risk of AKI compared with the control group (risk ratio, 0.70; 95% CI, 0.48–1.02)15. A substantial trend toward statistical significance suggests that limb IPC might be beneficial for prevention of AKI, although more adequately powered trials are still needed to validate its effectiveness15. The kidney-protective effect of limb IPC seem more pronounced in patients who are at high risk for AKI. This is supported by the recent Renal Protection (RenPro) Trial16, in which 100 adult patients with stable angina pectoris were recruited for elective coronary angiography using the contrast agent iohexol, a nephrotoxic iodinated radiocontrast media. This group of patients also had preexisting CKD and thus was at an increased risk of developing superimposed AKI.16 A total of 26 patients developed contrast-induced AKI, 6 (12%) in the limb IPC group and 20 (40%) in the control group. Multivariable analysis suggested limb IPC as a strong independent contributor to prevention of contrast -induced AKI16.

Notwithstanding the positive findings made by most of the clinical and animal studies, a few clinical trials1721 have demonstrated no benefit of IPC in preventing AKI. The conflicting results may be attributable to confounding variables like IPC protocols and patient characteristics. To date, the optimal protocol for remote IPC to trigger organ protection in humans remains unknown, but at least in rats, 3 or 4 (not 1 or 2) cycles of 5-minute remote IPC are necessary to confer heart protection22. Most of the clinical trials showing beneficial effects of IPC employed more than 3 cycles of 5 to 10-minute IPC, whereas in a study of 2 cycles of 10-minute iliac clamping, there was no evidence of any favorable changes in kidney outcome indices in patients undergoing open abdominal aortic aneurysm repair19. Also to be determined is whether use of upper versus lower limbs makes a difference in remote IPC preconditioning. Loukogeorgakis et al23 have shown that when using the leg rather than the arm for conditioning stimulus, more cycles are required to reach a preconditioning threshold and protect against ischemia/reperfusion-induced endothelial dysfunction in healthy volunteers. Anesthesia might be another confounding variable in examining the kidney effect of remote IPC. In some studies, such as those involving cardiac surgery, limb IPC was performed after anesthetic induction and, thus, patients were pain free21. However, as suggested by some experimental studies, pain may strongly stimulate pre-conditioning24, and remote IPC is dependent, in part, on intact local neural pathways25. In addition, some inhalational anesthetics can themselves cause preconditioning and protect from kidney injury26. This may mask the effect of remote IPC if IPC is performed during anesthesia. Thus, future studies are required to verify if variations in the mode of remote IPC, like the number of IPC cycles, upper versus lower limb IPC, or anesthesia, make a difference in the kidney-protective effects of IPC.

Different patient characteristics might be another important contributor to the contradictory findings from different trials. For instance, remote IPC is seemingly beneficial in patients at intermediate or high risk for AKI, whereas little kidney protective effect can be seen in patients with low risk or those who do not have reduced kidney function27. This variable should be validated in future studies by stratifying according to patients’ propensity for AKI. Furthermore, recent evidence suggests that, at least for cardioprotection, remote IPC is likely more effective in male than female patients28. We don’t as yet know if gender disparity also exists with IPC-associated kidney protection; however, it seems that clinical trials favoring a kidney protective effect for IPC often recruited more male patients29,30 and most of the negative studies1720 enrolled more female patients.

In view of the limitations of previous clinical trials as well as some of the previously described confounding factors, more studies aiming to provide definitive evidence in defining the role of remote IPC are in progress. Among these trials, the Remote Ischaemic Preconditioning for Heart Surgery (RIPHeart) study31 is a prospective, randomized, double-blind, multicentre, controlled trial including 2,070 adult cardiac surgical patients. It has been meticulously designed and adequately powered to determine whether remote IPC may improve clinical outcomes, including AKI, following all types of cardiac surgeries in which CPB is used.

Despite a number of reproducible observations suggesting the benefit of IPC in AKI, the underlying molecular mechanisms accounting for the kidney protective activity remain largely unknown. The most intriguing aspect of remote IPC is how the protective signal generated by remote IPC is transmitted to the target organs like the kidney. To date, there have been many studies investigating the mechanisms of remote IPC on heart protection32. From available evidence, the substance generating the remote IPC-induced protective activity is dialyzable, transferable, and receptor-mediated, suggesting the involvement of one or more blood-borne humoral mediators33. These mediators purportedly include an as-yet unidentified small (~3.5–15 kDa) hydrophobic molecule released during brief IPC, which is cardioprotective when purified and enriched by reverse phase chromatography and administered to naïve hearts12. In addition, autonomic reflex and other neurogenic pathways that are stimulated by the release of opioid, adenosine, and bradykinin may be involved in IPC-induced organ protection. In support of this view, blocking the autonomic ganglion abolishes the cardioprotective effects of remote IPC33. Moreover, systemic immune modulation and an anti-inflammatory effect on immune-competent cells following IPC might also contribute to the protection of target organs against subsequent acute injury33. The mediating pathways by which the protective signal is carried from the site subjected to IPC (e.g. limbs) to target organs may differ among experimental models and may involve humoral, neuronal, and systemic mechanisms (Figure 1).

Figure 1. GSK3β is a convergence point of multiple protective signaling pathways triggered by limb ischemia preconditioning (IPC).

Figure 1

The protective signals of limb IPC may be transmitted to remote target organs like the kidney via different communication mechanisms, including humoral mediators, neurogenic pathways, and systemic regulation of immune-competent cells. These mediating pathways function on the kidney and activate the reperfusion injury salvage kinase (RISK) pathway (involving PI3K/Akt signaling) and the survivor activating factor enhancement (SAFE) pathway (involving JAK/STAT3 signaling), both of which have been shown to converge on GSK3β. GSK3β is situated at the nexus of a number of pivotal signaling cascades and controls NFκB phosphorylation, Nrf2 nuclear exclusion and degradation, as well as the opening of the mitochondria permeability transition (MPT) pore. Inhibition of GSK3β in the kidney following limb IPC desensitizes MPT, reduces proinflammatory NFκB activation, and stabilizes Nrf2 in the nucleus in kidney cells, resulting in pro-survival/anti-apoptotic, anti-inflammatory/immunomodulatory and antioxidant effects, which contribute to limb IPC–conferred kidney protection against various injuries elicited by contrast media or other insults. GSK3β, glycogen synthase kinase 3β; JAK, Janus kinase; PI3K, phosphoinositide 3-kinase; RISK, reperfusion injury salvage kinase; SAFE, survivor activating factor enhancement; STAT3, signal transducer and activator of transcription 3.

RECENT ADVANCES

Regardless of the exact communication pathways transmitting the protective signal of IPC to the kidney, it is nevertheless important to decipher the molecular signaling mechanisms behind IPC so that pharmaceuticals can be developed as an alternative means of preconditioning that is able to precisely and effectively target the responsible signal transducers. This is particularly relevant for patients with preexisting CKD and comorbidities, like peripheral vascular disease, hypercoagulability, and diabetes mellitus. If subjected to limb IPC, in particular lower-limb IPC, these patients would be at significant risk for thrombosis, rhabdomyolysis, limb ischemia, nerve injury and other complications3436,37,.

IPC triggers a number of cell signaling transducers that contribute to the organ protective effects. Within the complex signaling network, the RISK (reperfusion injury salvage kinase) and SAFE (survivor activating factor enhancement) pathways have emerged as two major prosurvival signaling cascades mediating the beneficial action of IPC38. Activation of receptor tyrosine kinase or G-protein–coupled receptor by IPC in target organs triggers phosphoinositide 3-kinase (PI3K)/Akt and mitogen-activated protein kinases (MAPK)/extracellular regulated kinase (ERK) signal transduction as part of the RISK pathway (although the role of ERK is debatable). In parallel, IPC increases the levels of growth factors and cytokines like tumor necrosis factor (TNF)-α, which bind cell surface receptors in target organs and activate JAK (Janus kinase) and STAT3 (signal transducer and activator of transcription 3), as part of the SAFE pathway. Inhibition of either the RISK or the SAFE pathway totally abolishes the protective activity of IPC39,40, consistent with a cross-talk between the two pathways. Whether these two pathways converge on the same signaling transducer needs to be clarified, but glycogen synthase kinase (GSK) 3β has been shown to be a convergence point for the RISK and SAFE pathways41, justifying the choice of this signaling transducer as a target to both potentiate prosurvival cascades and maximize organ protection.

To understand the mechanism of action responsible for kidney protection elicited by remote IPC, Liu et al42 dissected the RISK signaling cascades43,44. Rats with preexisting CKD caused by partial nephrectomy were subjected to limb IPC prior to iohexol-induced AKI. The limb IPC–induced kidney protection could be abrogated by pretreatment with wortmannin (a specific inhibitor of PI3K) but not by U0126 (a ERK1/2 inhibitor)42. This observation was confirmed using another selective inhibitor of PI3K, LY294002, and another ERK1/2 inhibitor, PD98059. Moreover, the limb IPC–induced kidney protection was found to be associated with inhibitory phosphorylation of the signaling transducer GSK3β, which is downstream of the PI3K/Akt signaling pathway. Inhibition of GSK3β by SB216763, a selective small molecule inhibitor, mimicked the kidney protective effect of limb IPC and likewise attenuated loss of kidney function and prevented the acute kidney histological injury elicited by iohexol42.

GSK3β is a highly conserved serine/threonine protein kinase that is ubiquitously expressed and was found in the early 1980s to phosphorylate glycogen synthase and regulate glucose metabolism45,46. GSK3β is constitutively active in quiescent cells, while both the Wnt signaling pathway and PI3K/Akt downregulate its activity via inhibitory phosphorylation of the serine at amino acid 9 (ie, near the amino terminus). Its activity can be amplified by reactive oxygen species following oxidative injury47,48. Interest in GSK3β has heightened considerably following the finding that it is an important regulator of not just glycogen metabolism but also several other key cellular events such as signal transduction, insulin action, gene transcription, protein translation, cytoskeletal organization, cell cycle progression, and cell death and survival45. In addition, GSK3β has been implicated in a multitude of pathophysiologic processes, including embryo development, tissue injury, repair, and regeneration.

As a redox-sensitive serine/threonine protein kinase, GSK3β is interconnected with multiple cellular signaling cascades, including the Wnt, Nrf2 antioxidant response, and NF (nuclear factor) κB pathways, and more46. A number of transcription factors, such as Nrf2 (NRF2 in humans, ie, the product of the NFE2L2 gene) and the NFκB subunit RelA/p65, have been found to be cognate substrates for GSK3β and are subjected to GSK3β-directed phosphorylation and regulation of transcriptional activity49(Figure 1). Studies from our and other groups have indicated that GSK3β determines RelA/p65 phosphorylation at serine 468, thereby specifying the transcription of an array of NFκB target molecules involved in immune reaction and inflammatory response5052.

In studies we have performed in animal models, inhibition of GSK3β mitigates pro-inflammatory NFκB activation in kidney tubules50 and the glomerulus53, exerts an anti-inflammatory and immunoregulatory activity (Figure 2), but largely preserves other NFκB-dependent biological activities, including prosurvival/anti-apoptosis and anti-inflammation, resulting in attenuated kidney histologic evidence of kidney injury50. In addition, GSK3β is responsible for phosphorylation, nuclear exclusion, and degradation of Nrf254, which drives the primary cellular defense signaling against the cytotoxic effects of oxidative stress in mammalian cells55,56. Inhibition of GSK3β has been shown to promote the Nrf2-dependent antioxidant response and confer protection in several organ systems (Figure 3)56,57. Our studies have found that GSK3β inhibition by lithium reinforces the NRF2 antioxidant response and ameliorates hepatic injury in patients with hepatitis C virus infection57.

Figure 2. Ischemic preconditioning prevents inflammation in an acutely injured kidney by regulating the NFκκ κB-dependent inflammatory response.

Figure 2

NFκB RelA/p65 is a cognate substrate of GSK3β and can be phosphorylated by GSK3β at serine 468, which has been shown to specify the expression of an array of NFκB target genes involved in kidney inflammation. Upon injury, the activity of the redox-sensitive GSK3β is elevated. This promotes the phosphorylation of NFκB RelA/p65, and amplifies the transcription of pro-inflammatory genes, including chemokines, cytokines and inflammatory mediators. Ischemic preconditioning inhibits GSK3β by inducing phosphorylation at serine 9 via a number of communication pathways and possibly through reperfusion injury salvage kinase (RISK) and survivor activating factor enhancement (SAFE) signaling. GSK3β inhibition subsequent to ischemic preconditioning will attenuate NFκB RelA/p65 phosphorylation, repress the expression of proinflammatory molecules, and prevent inflammatory injury in the kidney, resulting in a kidney-protective effect. AKI, acute kidney injury; GSK3β, glycogen synthase kinase 3β.

Figure 3. Ischemic preconditioning exerts kidney protection and prevents acute kidney injury by regulating the GSK3β-dictated Nrf2 detoxification/antioxidant cellular defense pathway.

Figure 3

Nrf2, as a cognate substrate of GSK3β, can be phosphorylated directly by GSK3β or indirectly by Fyn kinase, which requires phosphorylation and activation by GSK3β. Phosphorylation of Nrf2 facilitates its nuclear export, which is followed by its ubiquitination and degradation. Upon injury, the activity of the redox-sensitive GSK3β is upregulated. This will promote Nrf2 phosphorylation, nuclear exit, and degradation, resulting in a reduced antioxidant gene expression and response. Ischemic preconditioning inhibits GSK3β by inducing phosphorylation at serine 9. This effect may be attained via a number of communication pathways and possibly through reperfusion injury salvage kinase (RISK) and survivor activating factor enhancement (SAFE) signaling. GSK3β inhibition diminishes Nrf2 phosphorylation, mitigate the nuclear export of Nrf2, enhance Nrf2 nuclear accumulation, and the antioxidant response, culminating in a kidney protective effect. AKI, acute kidney injury; GSK3β, glycogen synthase kinase 3β.

Kidney cells have a discrete mitochondrial pool of GSK3β48,58,59. Components of the mitochondria permeability transition (MPT) pore, including cyclophilin F and the voltage-dependent anion channel (VDAC), possess GSK3β phosphorylation consensus motifs and serve as putative substrates for GSK3β 48,5861. By controlling the phosphorylation and activity of cyclophilin F and VDAC, GSK3β may thereby determine how readily the MPT pore opens. Since induction of the MPT pore opening can lead to cell death, GSK3β can dictate the sensitivity of cells to pro-death stimuli. In our work, inhibition of GSK3β by selective inhibitors desensitizes the MPT pore to stimuli that would ordinarily cause it open, ameliorates mitochondria dysfunction in kidney tubular and glomerular cells, attenuates kidney dysfunction and proteinuria, and improves the histology of kidney tubules and the glomerulus following injury by the pro-oxidant paraquat48 and also high-dose diclofenac58 and adriamycin59 (Figure 4)48,58,59. Consistent with this observation, inhibition of GSK3β by limb IPC or by SB216763 (the small molecule inhibitor) ameliorates mitochondrial injury in kidney tubules after iohexol exposure in rats, enhances the tubular expression of Nrf2, reduces oxidative stress, blunts NFκB activation, and lessens kidney inflammation.42

Figure 4. Ischemic preconditioning protects against acute kidney injury by preventing mitochondria dysfunction and damage.

Figure 4

There is a discrete mitochondrial pool of GSK3β. Structural components of the mitochondrial permeability transition pore, including cyclophilin F and voltage-dependent anion channel (VDAC), possess multiple GSK3β phosphorylation consensus motifs and serve as cognate substrates for GSK3β. Phosphorylation of cyclophilin F and VDAC by GSK3β has been shown to reduce the threshold MPT pore (sensitizing it to open more readily). Upon oxidative injury, the activity of the redox sensitive GSK3β is elevated. This promotes the phosphorylation of cyclophilin F and VDAC, subsequently augments MPT and potentiates cell death. Ischemic preconditioning inhibits GSK3β by inducing phosphorylation at serine 9 via a number of communication pathways and possibly through reperfusion injury salvage kinase (RISK) and survivor activating factor enhancement (SAFE) signaling. GSK3β inhibition reduces the phosphorylation of cyclophilin F and VDAC, desensitizes MPT, and gives kidney cells resistance to prodeath insults, contributing to a kidney-protective effect. AKI, acute kidney injury; Cyp-F, cyclophilin F; GSK3β, glycogen synthase kinase 3β; MPT, mitochondria permeability transition; VDAC, voltage-dependent anion channel.

Besides IPC, a multitude of treatments and drugs are known to attenuate acute organ injury via inhibition of GSK3β. These include insulin, estrogen, valproic acid, lithium, and more62,63. Moreover, most of the communication pathways utilized by IPC for remote organ protection (Figure 1), such as adenosine, opioid, nitric oxide, reactive oxygen species, and autonomic reflex, are capable of inducing inhibitory phosphorylation of GSK3β64. Thus, GSK3β is likely the point of convergence for multiple protective pathways. Of note, GSK3 exists in two isoforms, GSK3a and GSK3β, both of which are widely expressed in almost all organ systems45,65, including the kidney. Although GSK3α and GSK3β display 84% structural homology and are similarly regulated and functionally redundant in many aspects of cellular signaling65, it seems that only GSK3β is involved in mediating the protective effect of IPC and other organ protective treatments66. Indeed, in cardiac myocytes, knockdown of GSK3α fails to provide any protective effects, even though the protective effects of insulin and hypoxic pre-conditioning remain intact. Knockdown of GSK3β by itself achieves levels of protection comparable to the effects of insulin66. Moreover, insulin does not increase the level of protection above that already achieved in the GSK3β-silenced cells. Conversely, in murine cardiac myocytes expressing a mutant form of GSK3β that cannot be inhibited (the serine 9 residue is replaced by an alanine), the cellular protective effect of hypoxic preconditioning is markedly blunted66. Therefore, inhibition of the β, but not the α, isoform of GSK3 is responsible for the protective signaling in IPC. Accordingly, therapeutic targeting of GSK3β by using pharmacological inhibitors is an attractive substitute for IPC as a prophylactic measure against AKI, in particular for patients unable to tolerate IPC.

Recent structure-activity relationship studies have advanced the development of a number of highly selective small-molecule chemical inhibitors of GSK3β. However, most of these novel inhibitors are currently in preclinical evaluation and may take years to enter clinical trials. Fortunately, several US Food and Drug Administration (FDA)-approved agents, like lithium, are able to effectively block the activity of GSK3β67,68. The lightest alkali metal on earth, lithium is the best-known GSK3β inhibitor67. It is an essential trace element required for physical and mental health in humans69 and has been in clinical use for a half century as an FDA-approved first-line treatment for bipolar affective disorders70. The beneficial effect of lithium in diverse organ systems has been noted for decades. For example, lithium exhibits a neuroprotective and neurotrophic activity71 and reinforces neural repair in acute brain injury (e.g. stroke or ischemia)72,73 as well as in chronic neurodegenerative diseases, such as Alzheimer’s, Parkinson’s, and Huntington’s diseases74,75. In addition, lithium can ameliorate neutropenia associated with cancer chemotherapy76 and has been one of the treatment choices for neutropenia77. Recent research has deciphered the mechanism of this general protective activity: lithium targets GSK3β, which as discussed previously is a key cell signaling molecule central to the pathogenesis of multiple human diseases, including AKI.

Studies from our and other laboratories have demonstrated that lithium-induced blockade of GSK3β can prevent AKI arising from endotoxin78, nephrotoxic drugs79 and ischemia/reperfusion injury79. As an alkali ion, lithium requires high blood levels to pass through the blood brain barrier80, thus the FDA-approved lithium dose for psychiatric disorders is quite high, with a relatively narrow therapeutic index. Long-term (usually >10 years) high-dose lithium therapy primarily for psychiatric disorders is occasionally associated with some kidney adverse effects, such as nephrogenic diabetes insipidus, interstitial nephritis, and even glomerular disease81. However, according to a large-scale epidemiology study of two regions in Sweden with a population of 2.7 million, end-stage kidney disease was uncommon in lithium-treated patients, although the incidence (0.53%) was noted to be higher than in the general population82. In our experience with animal models, the kidney-protective dose of lithium is much lower than its neurobiologic dose83. Short-term treatment with low-dose lithium (approximately 1/3 to 1/2 of the neurobiologic dose) results in no detectable changes in kidney histology or function in healthy mice, but clearly prevents kidney histologic injury in mouse models of AKI79. Therefore, it seems that short-term use of low-dose lithium might be a promising approach for preventing AKI. Taken together, if the pivotal role of GSK3β in mediating the kidney protection by IPC is validated, preconditioning with lithium might be a feasible and cost-effective alternative to IPC, especially for patients at risk of complications if subjected to IPC.

SUMMARY

There is increasing scientific evidence supporting a role for IPC in protecting against AKI. The possible underlying mechanisms are less clear because of the limitations in many of these studies. First, all of the clinical trials published so far are small-scale low-powered studies, which unavoidably undermine the reliability of the observations15. We believe that large-scale multicenter randomized controlled trials with high power would be worthwhile to conclusively validate the safety, effectiveness, and efficacy of IPC in preventing AKI. Second, published mechanistic studies have relied exclusively on the use of pharmacological inhibitors42, thus should be interpreted with the caveat that these inhibitors may act in a concentration-dependent way and may have other, as-yet unknown, functions. Researchers should consider using transgenic mice with kidney tubule-specific conditional knockout of the kinases being studied in order to validate the results in in-depth confirmatory studies. Mice with kidney tubule-specific conditional knockout of GSK3β have been well established84 and demonstrated resistance to mercury-induced AKI85. Whether ischemia/reperfusion injury–, cardiac surgery–, or contrast media–induced AKI is preventable in this line of knockout mice merits further investigation.

To date, available evidence demonstrates only that GSK3β inhibition is sufficient for IPC-associated kidney protection42. It remains unknown whether inhibitory phosphorylation of GSK3β is required and essential for the kidney protective activity of IPC. To address this question, gene-targeted knock-in mice with mutant (impervious to inhibition) GSK3β86 may be considered.

Furthermore, recent studies have indicated that there is cross-talk between the RISK and SAFE pathways and that the JAK-STAT3 pathway is regulated by the activity of PI3K-Akt-GSK3β cascade in the IPC-induced cardioprotection87,88. Future studies are needed to explore if the SAFE pathway is involved and is under the regulation by GSK3β in IPC-conferred kidney protection.

Despite these pitfalls, the key findings of the latest studies exploring kidney protection by IPC are very consistent with the mechanism of action of IPC revealed so far for heart protection, where GSK3β inactivation has been found to mediate a variety of divergent protection signaling pathways triggered by IPC to prevent myocardial injury66,89,90.

In conjunction with recent evidence indicating a general benefit of targeting GSK3β in various organ systems46,9193, it seems that GSK3β is situated at the convergence point of multiple protective pathways of IPC. Therapeutic targeting of GSK3β by IPC (Figure 1) or by using existing FDA-approved drugs with GSK3β inhibitory activities, such as lithium79, may represent a promising and pragmatic strategy for kidney protection and prevention of AKI. In future clinical practice, patients with high risk of AKI like the one presented in the case vignette may routinely undergo remote IPC as a standard preprocedural prophylactic measure to prevent AKI prior to medical interventions such as major surgery involving CPB or the use of contrast media.

Acknowledgments

Support: This work was supported in part by the U.S. National Institutes of Health grant R01DK092485 and the Natural Science Foundation of China 81070574 and 81270807.

Footnotes

Financial Disclosure: The authors declare that they have no relevant financial interests.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Chertow GM, Burdick E, Honour M, Bonventre JV, Bates DW. Acute kidney injury, mortality, length of stay, and costs in hospitalized patients. J Am Soc Nephrol. 2005 Nov;16(11):3365–3370. doi: 10.1681/ASN.2004090740. [DOI] [PubMed] [Google Scholar]
  • 2.Murugan R, Kellum JA. Acute kidney injury: what's the prognosis? Nat Rev Nephrol. 2011 Apr;7(4):209–217. doi: 10.1038/nrneph.2011.13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Chawla LS, Kimmel PL. Acute kidney injury and chronic kidney disease: an integrated clinical syndrome. Kidney Int. 2012 Sep;82(5):516–524. doi: 10.1038/ki.2012.208. [DOI] [PubMed] [Google Scholar]
  • 4.Coca SG, Singanamala S, Parikh CR. Chronic kidney disease after acute kidney injury: a systematic review and meta-analysis. Kidney Int. 2012 Mar;81(5):442–448. doi: 10.1038/ki.2011.379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Endre ZH. Renal ischemic preconditioning: finally some good news for prevention of acute kidney injury. Kidney Int. 2011 Oct;80(8):796–798. doi: 10.1038/ki.2011.193. [DOI] [PubMed] [Google Scholar]
  • 6.Levey AS, Stevens LA, Schmid CH, et al. A new equation to estimate glomerular filtration rate. Ann Intern Med. 2009 May 5;150(9):604–612. doi: 10.7326/0003-4819-150-9-200905050-00006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Thakar CV, Arrigain S, Worley S, Yared JP, Paganini EP. A clinical score to predict acute renal failure after cardiac surgery. J Am Soc Nephrol. 2005 Jan;16(1):162–168. doi: 10.1681/ASN.2004040331. [DOI] [PubMed] [Google Scholar]
  • 8.Tomai F, Crea F, Chiariello L, Gioffre PA. Ischemic preconditioning in humans: models, mediators, and clinical relevance. Circulation. 1999 Aug 3;100(5):559–563. doi: 10.1161/01.cir.100.5.559. [DOI] [PubMed] [Google Scholar]
  • 9.Murry CE, Jennings RB, Reimer KA. Preconditioning with ischemia: a delay of lethal cell injury in ischemic myocardium. Circulation. 1986 Nov;74(5):1124–1136. doi: 10.1161/01.cir.74.5.1124. [DOI] [PubMed] [Google Scholar]
  • 10.Zager RA, Baltes LA, Sharma HM, Jurkowitz MS. Responses of the ischemic acute renal failure kidney to additional ischemic events. Kidney Int. 1984 Nov;26(5):689–700. doi: 10.1038/ki.1984.204. [DOI] [PubMed] [Google Scholar]
  • 11.Bonventre JV. Limb ischemia protects against contrast-induced nephropathy. Circulation. 2012 Jul 24;126(4):384–387. doi: 10.1161/CIRCULATIONAHA.112.119701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Przyklenk K, Whittaker P. Remote ischemic preconditioning: current knowledge, unresolved questions, and future priorities. J Cardiovasc Pharmacol Ther. 2011 Sep-Dec;16(3–4):255–259. doi: 10.1177/1074248411409040. [DOI] [PubMed] [Google Scholar]
  • 13.Lim SY, Hausenloy DJ. Remote ischemic conditioning: from bench to bedside. Front Physiol. 2012;3:27. doi: 10.3389/fphys.2012.00027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Wever KE, Menting TP, Rovers M, et al. Ischemic preconditioning in the animal kidney, a systematic review and meta-analysis. PLoS One. 2012;7(2):e32296. doi: 10.1371/journal.pone.0032296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Yang Y, Lang XB, Zhang P, Lv R, Wang YF, Chen JH. Remote ischemic preconditioning for prevention of acute kidney injury: a meta-analysis of randomized controlled trials. Am J Kidney Dis. 2014 Oct;64(4):574–583. doi: 10.1053/j.ajkd.2014.04.029. [DOI] [PubMed] [Google Scholar]
  • 16.Er F, Nia AM, Dopp H, et al. Ischemic preconditioning for prevention of contrast medium-induced nephropathy: randomized pilot RenPro Trial (Renal Protection Trial) Circulation. 2012 Jul 17;126(3):296–303. doi: 10.1161/CIRCULATIONAHA.112.096370. [DOI] [PubMed] [Google Scholar]
  • 17.Choi YS, Shim JK, Kim JC, et al. Effect of remote ischemic preconditioning on renal dysfunction after complex valvular heart surgery: a randomized controlled trial. J Thorac Cardiovasc Surg. 2011 Jul;142(1):148–154. doi: 10.1016/j.jtcvs.2010.11.018. [DOI] [PubMed] [Google Scholar]
  • 18.Rahman IA, Mascaro JG, Steeds RP, et al. Remote ischemic preconditioning in human coronary artery bypass surgery: from promise to disappointment? Circulation. 2010 Sep 14;122(11 Suppl):S53–59. doi: 10.1161/CIRCULATIONAHA.109.926667. [DOI] [PubMed] [Google Scholar]
  • 19.Walsh SR, Sadat U, Boyle JR, et al. Remote ischemic preconditioning for renal protection during elective open infrarenal abdominal aortic aneurysm repair: randomized controlled trial. Vasc Endovascular Surg. 2010 Jul;44(5):334–340. doi: 10.1177/1538574410370788. [DOI] [PubMed] [Google Scholar]
  • 20.Pedersen KR, Ravn HB, Povlsen JV, Schmidt MR, Erlandsen EJ, Hjortdal VE. Failure of remote ischemic preconditioning to reduce the risk of postoperative acute kidney injury in children undergoing operation for complex congenital heart disease: a randomized single-center study. J Thorac Cardiovasc Surg. 2012 Mar;143(3):576–583. doi: 10.1016/j.jtcvs.2011.08.044. [DOI] [PubMed] [Google Scholar]
  • 21.Gallagher SM, Jones DA, Kapur A, et al. Remote ischemic preconditioning has a neutral effect on the incidence of kidney injury after coronary artery bypass graft surgery. Kidney Int. 2015 Feb;87(2):473–481. doi: 10.1038/ki.2014.259. [DOI] [PubMed] [Google Scholar]
  • 22.Xin P, Zhu W, Li J, et al. Combined local ischemic postconditioning and remote perconditioning recapitulate cardioprotective effects of local ischemic preconditioning. Am J Physiol Heart Circ Physiol. 2010 Jun;298(6):H1819–1831. doi: 10.1152/ajpheart.01102.2009. [DOI] [PubMed] [Google Scholar]
  • 23.Loukogeorgakis SP, Williams R, Panagiotidou AT, et al. Transient limb ischemia induces remote preconditioning and remote postconditioning in humans by a K(ATP)-channel dependent mechanism. Circulation. 2007 Sep 18;116(12):1386–1395. doi: 10.1161/CIRCULATIONAHA.106.653782. [DOI] [PubMed] [Google Scholar]
  • 24.Jones WK, Fan GC, Liao S, et al. Peripheral nociception associated with surgical incision elicits remote nonischemic cardioprotection via neurogenic activation of protein kinase C signaling. Circulation. 2009 Sep 15;120(11 Suppl):S1–9. doi: 10.1161/CIRCULATIONAHA.108.843938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Loukogeorgakis SP, Panagiotidou AT, Broadhead MW, Donald A, Deanfield JE, MacAllister RJ. Remote ischemic preconditioning provides early and late protection against endothelial ischemia-reperfusion injury in humans: role of the autonomic nervous system. J Am Coll Cardiol. 2005 Aug 2;46(3):450–456. doi: 10.1016/j.jacc.2005.04.044. [DOI] [PubMed] [Google Scholar]
  • 26.Ma D, Lim T, Xu J, et al. Xenon preconditioning protects against renal ischemic-reperfusion injury via HIF-1alpha activation. J Am Soc Nephrol. 2009 Apr;20(4):713–720. doi: 10.1681/ASN.2008070712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Deftereos S, Giannopoulos G, Tzalamouras V, et al. Renoprotective effect of remote ischemic post-conditioning by intermittent balloon inflations in patients undergoing percutaneous coronary intervention. J Am Coll Cardiol. 2013 May 14;61(19):1949–1955. doi: 10.1016/j.jacc.2013.02.023. [DOI] [PubMed] [Google Scholar]
  • 28.Pei H, Wu Y, Wei Y, Yang Y, Teng S, Zhang H. Remote ischemic preconditioning reduces perioperative cardiac and renal events in patients undergoing elective coronary intervention: a meta-analysis of 11 randomized trials. PLoS One. 2014;9(12):e115500. doi: 10.1371/journal.pone.0115500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Venugopal V, Laing CM, Ludman A, Yellon DM, Hausenloy D. Effect of remote ischemic preconditioning on acute kidney injury in nondiabetic patients undergoing coronary artery bypass graft surgery: a secondary analysis of 2 small randomized trials. Am J Kidney Dis. 2010 Dec;56(6):1043–1049. doi: 10.1053/j.ajkd.2010.07.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Zimmerman RF, Ezeanuna PU, Kane JC, et al. Ischemic preconditioning at a remote site prevents acute kidney injury in patients following cardiac surgery. Kidney Int. 2011 Oct;80(8):861–867. doi: 10.1038/ki.2011.156. [DOI] [PubMed] [Google Scholar]
  • 31.Meybohm P, Zacharowski K, Cremer J, et al. Remote ischaemic preconditioning for heart surgery. The study design for a multi-center randomized double-blinded controlled clinical trial--the RIPHeart-Study. Eur Heart J. 2012 Jun;33(12):1423–1426. [PubMed] [Google Scholar]
  • 32.Hausenloy DJ, Yellon DM. Remote ischaemic preconditioning: underlying mechanisms and clinical application. Cardiovasc Res. 2008 Aug 1;79(3):377–386. doi: 10.1093/cvr/cvn114. [DOI] [PubMed] [Google Scholar]
  • 33.Gassanov N, Nia AM, Caglayan E, Er F. Remote ischemic preconditioning and renoprotection: from myth to a novel therapeutic option? J Am Soc Nephrol. 2014 Feb;25(2):216–224. doi: 10.1681/ASN.2013070708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Srinivasan C, Kuppuswamy B. Rhabdomyolysis complicating non-invasive blood pressure measurement. Indian J Anaesth. 2012 Jul;56(4):428–430. doi: 10.4103/0019-5049.100847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Celoria G, Dawson JA, Teres D. Compartment syndrome in a patient monitored with an automated blood pressure cuff. J Clin Monit. 1987 Apr;3(2):139–141. doi: 10.1007/BF00858363. [DOI] [PubMed] [Google Scholar]
  • 36.Lin CC, Jawan B, de Villa MV, Chen FC, Liu PP. Blood pressure cuff compression injury of the radial nerve. J Clin Anesth. 2001 Jun;13(4):306–308. doi: 10.1016/s0952-8180(01)00262-8. [DOI] [PubMed] [Google Scholar]
  • 37.Devbhandari M, Shariff Z, Duncan AJ. Skin necrosis in a critically ill patient due to a blood pressure cuff. J Postgrad Med. 2006 Apr-Jun;52(2):136–138. [PubMed] [Google Scholar]
  • 38.Lecour S. Activation of the protective Survivor Activating Factor Enhancement (SAFE) pathway against reperfusion injury: Does it go beyond the RISK pathway? J Mol Cell Cardiol. 2009 Jul;47(1):32–40. doi: 10.1016/j.yjmcc.2009.03.019. [DOI] [PubMed] [Google Scholar]
  • 39.Smith RM, Suleman N, Lacerda L, et al. Genetic depletion of cardiac myocyte STAT-3 abolishes classical preconditioning. Cardiovasc Res. 2004 Sep 1;63(4):611–616. doi: 10.1016/j.cardiores.2004.06.019. [DOI] [PubMed] [Google Scholar]
  • 40.Suleman N, Somers S, Smith R, Opie LH, Lecour SC. Dual activation of STAT-3 and Akt is required during the trigger phase of ischaemic preconditioning. Cardiovasc Res. 2008 Jul 1;79(1):127–133. doi: 10.1093/cvr/cvn067. [DOI] [PubMed] [Google Scholar]
  • 41.Gross ER, Hsu AK, Gross GJ. The JAK/STAT pathway is essential for opioid-induced cardioprotection: JAK2 as a mediator of STAT3, Akt, and GSK-3 beta. Am J Physiol Heart Circ Physiol. 2006 Aug;291(2):H827–834. doi: 10.1152/ajpheart.00003.2006. [DOI] [PubMed] [Google Scholar]
  • 42.Liu T, Fang Y, Liu S, et al. Limb ischemic preconditioning protects against contrast-induced acute kidney injury in rats via phosphorylation of GSK-3beta. Free Radic Biol Med. 2015 Oct 31;81:170–182. doi: 10.1016/j.freeradbiomed.2014.10.509. [DOI] [PubMed] [Google Scholar]
  • 43.Hausenloy DJ, Yellon DM. New directions for protecting the heart against ischaemia-reperfusion injury: targeting the Reperfusion Injury Salvage Kinase (RISK)-pathway. Cardiovasc Res. 2004 Feb 15;61(3):448–460. doi: 10.1016/j.cardiores.2003.09.024. [DOI] [PubMed] [Google Scholar]
  • 44.Hausenloy DJ, Tsang A, Yellon DM. The reperfusion injury salvage kinase pathway: a common target for both ischemic preconditioning and postconditioning. Trends Cardiovasc Med. 2005 Feb;15(2):69–75. doi: 10.1016/j.tcm.2005.03.001. [DOI] [PubMed] [Google Scholar]
  • 45.Cohen P, Frame S. The renaissance of GSK3. Nat Rev Mol Cell Biol. 2001 Oct;2(10):769–776. doi: 10.1038/35096075. [DOI] [PubMed] [Google Scholar]
  • 46.Rayasam GV, Tulasi VK, Sodhi R, Davis JA, Ray A. Glycogen synthase kinase 3: more than a namesake. Br J Pharmacol. 2009 Mar;156(6):885–898. doi: 10.1111/j.1476-5381.2008.00085.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Chiara F, Gambalunga A, Sciacovelli M, et al. Chemotherapeutic induction of mitochondrial oxidative stress activates GSK-3alpha/beta and Bax, leading to permeability transition pore opening and tumor cell death. Cell Death Dis. 2012;3:e444. doi: 10.1038/cddis.2012.184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Wang Z, Ge Y, Bao H, Dworkin L, Peng A, Gong R. Redox-sensitive glycogen synthase kinase 3beta-directed control of mitochondrial permeability transition: rheostatic regulation of acute kidney injury. Free Radic Biol Med. 2013 Dec;65:849–858. doi: 10.1016/j.freeradbiomed.2013.08.169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Sutherland C. What Are the bona fide GSK3 Substrates? Int J Alzheimers Dis. 2011;2011:505607. doi: 10.4061/2011/505607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Gong R, Rifai A, Ge Y, Chen S, Dworkin LD. Hepatocyte growth factor suppresses proinflammatory NFkappaB activation through GSK3beta inactivation in renal tubular epithelial cells. J Biol Chem. 2008 Mar 21;283(12):7401–7410. doi: 10.1074/jbc.M710396200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Buss H, Dorrie A, Schmitz ML, et al. Phosphorylation of serine 468 by GSK-3beta negatively regulates basal p65 NF-kappaB activity. J Biol Chem. 2004 Nov 26;279(48):49571–49574. doi: 10.1074/jbc.C400442200. [DOI] [PubMed] [Google Scholar]
  • 52.Steinbrecher KA, Wilson W, 3rd, Cogswell PC, Baldwin AS. Glycogen synthase kinase 3beta functions to specify gene-specific, NF-kappaB-dependent transcription. Mol Cell Biol. 2005 Oct;25(19):8444–8455. doi: 10.1128/MCB.25.19.8444-8455.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Bao H, Ge Y, Peng A, Gong R. Fine-tuning of NFkappaB by glycogen synthase kinase 3beta directs the fate of glomerular podocytes upon injury. Kidney Int. 2015 Jan 28;87(6):1176–1190. doi: 10.1038/ki.2014.428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Salazar M, Rojo AI, Velasco D, de Sagarra RM, Cuadrado A. Glycogen synthase kinase-3beta inhibits the xenobiotic and antioxidant cell response by direct phosphorylation and nuclear exclusion of the transcription factor Nrf2. J Biol Chem. 2006 May 26;281(21):14841–14851. doi: 10.1074/jbc.M513737200. [DOI] [PubMed] [Google Scholar]
  • 55.Motohashi H, Yamamoto M. Nrf2-Keap1 defines a physiologically important stress response mechanism. Trends Mol Med. 2004 Nov;10(11):549–557. doi: 10.1016/j.molmed.2004.09.003. [DOI] [PubMed] [Google Scholar]
  • 56.Rojo AI, Sagarra MR, Cuadrado A. GSK-3beta down-regulates the transcription factor Nrf2 after oxidant damage: relevance to exposure of neuronal cells to oxidative stress. J Neurochem. 2008 Apr;105(1):192–202. doi: 10.1111/j.1471-4159.2007.05124.x. [DOI] [PubMed] [Google Scholar]
  • 57.Jiang Y, Bao H, Ge Y, et al. Therapeutic targeting of GSK3beta enhances the Nrf2 antioxidant response and confers hepatic cytoprotection in hepatitis C. Gut. 2015 Jan;64(1):168–179. doi: 10.1136/gutjnl-2013-306043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Bao H, Ge Y, Zhuang S, Dworkin LD, Liu Z, Gong R. Inhibition of glycogen synthase kinase-3beta prevents NSAID-induced acute kidney injury. Kidney Int. 2012 Apr;81(7):662–673. doi: 10.1038/ki.2011.443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Wang Z, Bao H, Ge Y, Zhuang S, Peng A, Gong R. Pharmacological targeting of GSK3beta confers protection against podocytopathy and proteinuria by desensitizing mitochondrial permeability transition. Br J Pharmacol. 2014 Sep 29;172(3):895–909. doi: 10.1111/bph.12952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Tanno M, Kuno A, Ishikawa S, et al. Translocation of glycogen synthase kinase-3beta (GSK-3beta), a trigger of permeability transition, is kinase activity-dependent and mediated by interaction with voltage-dependent anion channel 2 (VDAC2) J Biol Chem. 2014 Oct 17;289(42):29285–29296. doi: 10.1074/jbc.M114.563924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Zhu J, Rebecchi MJ, Glass PS, Brink PR, Liu L. Interactions of GSK-3beta with mitochondrial permeability transition pore modulators during preconditioning: age-associated differences. J Gerontol A Biol Sci Med Sci. 2013 Apr;68(4):395–403. doi: 10.1093/gerona/gls205. [DOI] [PubMed] [Google Scholar]
  • 62.Fink MP. What do insulin, estrogen, valproic acid, and TDZD-8 have in common? Crit Care Med. 2005 Sep;33(9):2115–2117. doi: 10.1097/01.ccm.0000178348.82829.ad. [DOI] [PubMed] [Google Scholar]
  • 63.Dugo L, Collin M, Allen DA, et al. GSK-3beta inhibitors attenuate the organ injury/dysfunction caused by endotoxemia in the rat. Crit Care Med. 2005 Sep;33(9):1903–1912. doi: 10.1097/01.ccm.0000178350.21839.44. [DOI] [PubMed] [Google Scholar]
  • 64.Murphy E, Steenbergen C. Inhibition of GSK-3beta as a target for cardioprotection: the importance of timing, location, duration and degree of inhibition. Expert Opin Ther Targets. 2005 Jun;9(3):447–456. doi: 10.1517/14728222.9.3.447. [DOI] [PubMed] [Google Scholar]
  • 65.Doble BW, Woodgett JR. GSK-3: tricks of the trade for a multi-tasking kinase. J Cell Sci. 2003 Apr 1;116(Pt 7):1175–1186. doi: 10.1242/jcs.00384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Juhaszova M, Zorov DB, Kim SH, et al. Glycogen synthase kinase-3beta mediates convergence of protection signaling to inhibit the mitochondrial permeability transition pore. J Clin Invest. 2004 Jun;113(11):1535–1549. doi: 10.1172/JCI19906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Jope RS. Lithium and GSK-3: one inhibitor, two inhibitory actions, multiple outcomes. Trends Pharmacol Sci. 2003 Sep;24(9):441–443. doi: 10.1016/S0165-6147(03)00206-2. [DOI] [PubMed] [Google Scholar]
  • 68.Freland L, Beaulieu JM. Inhibition of GSK3 by lithium, from single molecules to signaling networks. Front Mol Neurosci. 2012 Jan 27;5:14. doi: 10.3389/fnmol.2012.00014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Zarse K, Terao T, Tian J, Iwata N, Ishii N, Ristow M. Low-dose lithium uptake promotes longevity in humans and metazoans. Eur J Nutr. 2011 Aug;50(5):387–389. doi: 10.1007/s00394-011-0171-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Cade JF. Lithium salts in the treatment of psychotic excitement. Med J Aust. 1949 Sep 3;2(10):349–352. doi: 10.1080/j.1440-1614.1999.06241.x. [DOI] [PubMed] [Google Scholar]
  • 71.Moore GJ, Bebchuk JM, Wilds IB, Chen G, Manji HK. Lithium-induced increase in human brain grey matter. Lancet. 2000 Oct 7;356(9237):1241–1242. doi: 10.1016/s0140-6736(00)02793-8. [DOI] [PubMed] [Google Scholar]
  • 72.Wada A, Yokoo H, Yanagita T, Kobayashi H. Lithium: potential therapeutics against acute brain injuries and chronic neurodegenerative diseases. J Pharmacol Sci. 2005 Dec;99(4):307–321. doi: 10.1254/jphs.crj05009x. [DOI] [PubMed] [Google Scholar]
  • 73.Kang K, Kim YJ, Kim YH, et al. Lithium pretreatment reduces brain injury after intracerebral hemorrhage in rats. Neurol Res. 2012 Jun;34(5):447–454. doi: 10.1179/1743132812Y.0000000015. [DOI] [PubMed] [Google Scholar]
  • 74.Forlenza OV, De-Paula VJ, Diniz BS. Neuroprotective Effects of Lithium: Implications for the Treatment of Alzheimer's Disease and Related Neurodegenerative Disorders. ACS Chem Neurosci. 2014 May 6;5(6):443–450. doi: 10.1021/cn5000309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Bauer M, Alda M, Priller J, Young LT. Implications of the neuroprotective effects of lithium for the treatment of bipolar and neurodegenerative disorders. Pharmacopsychiatry. 2003 Nov;36 (Suppl 3):S250–254. doi: 10.1055/s-2003-45138. [DOI] [PubMed] [Google Scholar]
  • 76.Focosi D, Azzara A, Kast RE, Carulli G, Petrini M. Lithium and hematology: established and proposed uses. J Leukoc Biol. 2009 Jan;85(1):20–28. doi: 10.1189/jlb.0608388. [DOI] [PubMed] [Google Scholar]
  • 77.Lyman GH, Williams CC, Preston D. The use of lithium carbonate to reduce infection and leukopenia during systemic chemotherapy. N Engl J Med. 1980 Jan 31;302(5):257–260. doi: 10.1056/NEJM198001313020503. [DOI] [PubMed] [Google Scholar]
  • 78.Wang Y, Huang WC, Wang CY, et al. Inhibiting glycogen synthase kinase-3 reduces endotoxaemic acute renal failure by down-regulating inflammation and renal cell apoptosis. Br J Pharmacol. 2009 Jul;157(6):1004–1013. doi: 10.1111/j.1476-5381.2009.00284.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Bao H, Ge Y, Wang Z, et al. Delayed administration of a single dose of lithium promotes recovery from AKI. J Am Soc Nephrol. 2014 Mar;25(3):488–500. doi: 10.1681/ASN.2013040350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Platman SR, Fieve RR. Biochemical aspects of lithium in affective disorders. Arch Gen Psychiatry. 1968 Dec;19(6):659–663. doi: 10.1001/archpsyc.1968.01740120019003. [DOI] [PubMed] [Google Scholar]
  • 81.Markowitz GS, Radhakrishnan J, Kambham N, Valeri AM, Hines WH, D'Agati VD. Lithium nephrotoxicity: a progressive combined glomerular and tubulointerstitial nephropathy. J Am Soc Nephrol. 2000 Aug;11(8):1439–1448. doi: 10.1681/ASN.V1181439. [DOI] [PubMed] [Google Scholar]
  • 82.Bendz H, Schon S, Attman PO, Aurell M. Renal failure occurs in chronic lithium treatment but is uncommon. Kidney Int. 2010 Feb;77(3):219–224. doi: 10.1038/ki.2009.433. [DOI] [PubMed] [Google Scholar]
  • 83.Xu W, Ge Y, Liu Z, Gong R. Glycogen synthase kinase 3beta orchestrates microtubule remodeling in compensatory glomerular adaptation to podocyte depletion. J Biol Chem. 2015 Jan 16;290(3):1348–1363. doi: 10.1074/jbc.M114.593830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Ge Y, Si J, Tian L, Zhuang S, Dworkin LD, Gong R. Conditional ablation of glycogen synthase kinase 3beta in postnatal mouse kidney. Lab Invest. 2011 Jan;91(1):85–96. doi: 10.1038/labinvest.2010.142. [DOI] [PubMed] [Google Scholar]
  • 85.Howard C, Tao S, Yang HC, et al. Specific deletion of glycogen synthase kinase-3beta in the renal proximal tubule protects against acute nephrotoxic injury in mice. Kidney Int. 2012 Nov;82(9):1000–1009. doi: 10.1038/ki.2012.239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.McManus EJ, Sakamoto K, Armit LJ, et al. Role that phosphorylation of GSK3 plays in insulin and Wnt signalling defined by knockin analysis. EMBO J. 2005 Apr 20;24(8):1571–1583. doi: 10.1038/sj.emboj.7600633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Tamareille S, Mateus V, Ghaboura N, et al. RISK and SAFE signaling pathway interactions in remote limb ischemic perconditioning in combination with local ischemic postconditioning. Basic Res Cardiol. 2011 Nov;106(6):1329–1339. doi: 10.1007/s00395-011-0210-z. [DOI] [PubMed] [Google Scholar]
  • 88.Heusch G. No risk, no ... cardioprotection? A critical perspective. Cardiovasc Res. 2009 Nov 1;84(2):173–175. doi: 10.1093/cvr/cvp298. [DOI] [PubMed] [Google Scholar]
  • 89.Murphy E. Inhibit GSK-3beta or there's heartbreak dead ahead. J Clin Invest. 2004 Jun;113(11):1526–1528. doi: 10.1172/JCI21986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Juhaszova M, Zorov DB, Yaniv Y, Nuss HB, Wang S, Sollott SJ. Role of glycogen synthase kinase-3beta in cardioprotection. Circ Res. 2009 Jun 5;104(11):1240–1252. doi: 10.1161/CIRCRESAHA.109.197996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Miura T, Miki T. GSK–3beta, a therapeutic target for cardiomyocyte protection. Circ J. 2009 Jul;73(7):1184–1192. doi: 10.1253/circj.cj-09-0284. [DOI] [PubMed] [Google Scholar]
  • 92.Fu H, Xu H, Chen H, et al. Inhibition of glycogen synthase kinase 3 ameliorates liver ischemia/reperfusion injury via an energy-dependent mitochondrial mechanism. J Hepatol. 2014 Oct;61(4):816–824. doi: 10.1016/j.jhep.2014.05.017. [DOI] [PubMed] [Google Scholar]
  • 93.Nelson PJ, Cantley L. GSK3beta plays dirty in acute kidney injury. J Am Soc Nephrol. 2010 Feb;21(2):199–200. doi: 10.1681/ASN.2009121214. [DOI] [PubMed] [Google Scholar]

RESOURCES