Skip to main content
American Journal of Physiology - Renal Physiology logoLink to American Journal of Physiology - Renal Physiology
. 2015 Sep 2;309(10):F807–F820. doi: 10.1152/ajprenal.00266.2015

Twenty years after ACEIs and ARBs: emerging treatment strategies for diabetic nephropathy

Stacy A Johnson 1, Robert F Spurney 1,
PMCID: PMC4652076  PMID: 26336162

Abstract

Diabetic nephropathy (DN) is a serious complication of both type 1 and type 2 diabetes mellitus. The disease is now the most common cause of end-stage kidney disease (ESKD) in developed countries, and both the incidence and prevalence of diabetes mellitus is increasing worldwide. Current treatments are directed at controlling hyperglycemia and hypertension, as well as blockade of the renin angiotensin system with angiotensin-converting enzyme inhibitors (ACEIs), and angiotensin receptor blockers. Despite these therapies, DN progresses to ESKD in many patients. As a result, much interest is focused on developing new therapies. It has been over two decades since ACEIs were shown to have beneficial effects in DN independent of their blood pressure-lowering actions. Since that time, our understanding of disease mechanisms in DN has evolved. In this review, we summarize major cell signaling pathways implicated in the pathogenesis of diabetic kidney disease, as well as emerging treatment strategies. The goal is to identify promising targets that might be translated into therapies for the treatment of patients with diabetic kidney disease.

Keywords: diabetic nephropathy, diabetes mellitus, cell signaling, oxidative stress


diabetic nephropathy (dn) is the leading cause of end-stage kidney disease in developed countries (123). Current clinical management is directed at strict control of blood glucose and blood pressure (BP) as well as inhibition of the renin angiotensin system (RAS) using angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin receptor blockers (ARBs). While several smaller human studies had suggested beneficial effects of ACEIs in DN, the beneficial actions of these agents were convincingly demonstrated in a large, randomized, controlled trial in 1993 (83). In this study, captopril attenuated the decline in renal function in Type 1 diabetic patients with heavy proteinuria more effectively than BP control alone (83). Since this landmark study, data from several large human studies have consistently demonstrated renoprotective benefits of ACEIs and ARBs in diabetic patients with kidney disease (13, 84, 107). As a result, ACEIs and ARBs are now a mainstay of management for DN. Despite their clinical effectiveness, diabetic kidney disease continues to progress ultimately causing end-stage kidney disease (ESKD) in ∼20% of patients (92, 97). Thus, new treatment strategies are needed. In this review, we examine major cell-signaling pathways implicated in the pathogenesis of DN beginning with G protein-coupled receptors (GPCRs) and proceeding in rough chronological order to more recently identified signaling pathways that might be exploited for the treatment of diabetic kidney disease. Where applicable, we will discuss recently completed or ongoing clinical trials; however, the primary focus will be data arising from in vitro and in vivo experiments that may inform future clinical studies.

Targeting GPCR Signaling Pathways in DN

GPCRs are the target of ∼30–40% of all modern medicinal drugs (140). In this section, we discuss promising strategies to maximize RAS blockade, as well as other GPCR systems that might be targeted for the treatment of diabetic kidney disease.

Maximizing RAS blockade.

While targeting the RAS was a major advance in the treatment of DN (13, 83, 84, 107), initial attempts to maximize RAS blockade produced disappointing results (42, 170). For example, combined ACEI and ARB therapy was associated with an increased risk of the composite primary end-point of death from cardiovascular causes in the ONTARGET clinical trial (170). In a secondary analysis of renal outcomes, dialysis or doubling of the serum creatinine was similar in the monotherapy ACEI or ARB group but increased with combination treatment (94). Other investigators have targeted additional components of the RAS. For example, the direct renin inhibitor aliskiren showed promise as an adjunct to ACEI and ARBs in the treatment of DN (108, 112). Unfortunately, when the effect of aliskiren in combination with the RAS blockade was tested in a large cohort of patients with Type 2 diabetes and chronic kidney disease (CKD), investigators observed no difference in the composite end-point of death from cardiovascular or renal events (106). In this trial, aliskiren reduced albuminuria but increased the rate of decline in renal function and was associated with an increased risk of hypotension and hyperkalemia (106). An alternative approach to enhance RAS blockade is inhibition of the mineralocorticoid receptor (MR), a nuclear receptor that is not a member of the GPCR family. In vitro, aldosterone promotes apoptosis in podocytes (81) and induces expression of the inflammatory chemokine CCL2 (C-C motif ligand 2, formerly termed monocyte chemoattractant protein 1) in an NF-κB-dependent manner in both mesangial and proximal tubule cells (54). Furthermore, administration of MR antagonists to rodents with either type 1 or type 2 diabetes decreases albuminuria and inhibits fibrosis (51, 54). In small clinical trials, short-term treatment with spironolactone decreased 24-h urinary albumin excretion in Type 1 and Type 2 diabetic patients with persistent albuminuria despite RAS blockade (68, 127, 128). Similarly, combined treatment with an ACEI and the selective aldosterone inhibitor eplerenone reduced albuminuria to a greater extent than the ACEI alone without causing a significant increase in hyperkalemia (37). Additional MR antagonists have been developed (Table 1) and phase 2 clinical trials are currently evaluating these agents in patients with Type 2 diabetes and albuminuria (Table 2). Ultimately, MR blockade may prove to be an effective treatment strategy for DN; however, larger trials with longer followup are required to assess its effect on progression to ESRD and/or mortality.

Table 1.

Selected approved and investigational drug products

Pathway Approved Drug(s)* Investigational Drug(s)* Mechanism(s) of action Major adverse side-effect(s)
Adenosine GPCRs Dipyridamole Adenosine re-uptake inhibitor Bleeding, Myocardial ischemia at high doses
ATL146e, CGS212680 α2A receptor agonists
AMPK Metformin Adiponectin, AICAR** Activates AMPK (indirectly inhibits mTOR) Lactic acidosis
Calcineurin Cyclosporine, Tacrolimus VIVIT Inhibitors Immunosuppression Nephrotoxicity
Chemokine GPCRs CCX140-B, INCB3344, RS504393, RO5234444 BMS-813160 CCR2 antagonists GI symptoms, Infection
PF-04634817
CCR2/5 antagonist
MLN1202 CCR2 antibody
Endothelin GPCRs Atrasentan, Avosentan Endothelin antagonists Fluid retention, Heart failure
JAK/STAT Tofacitinib, Ruxolitinib AG490 JAK inhibitors Anemia, Thrombocytopenia Immunosuppression
Barcitinib
MR Spironolactone, Eplerenone Finerenone (BAY94-8862) MR antagonists Hyperkalemia
MT-3995
mTOR Sirolimus, Everolimus Inhibitors Immunosuppression, Proteinuria
Oxidative stress
    Antioxidants Vitamin E, Probucol, Resveratrol α-lipoic acid Anitoxidants
    Nrf2 Bardoxolone, RTA405 Nrf2 activators Heart failure, Proteinuria
    NOX
Apocynin NOX inhibitor
GKT137831 NOX1/4 inhibitor
TGF-β Pirfenidone, Inhibits the TGF-β promoter activity GI symptoms, Photosensitivity rash
Anti-TGF-β antibodies TGF-β antibodies
LY2382770 Small molecule TGF-β inhibitors
Trabedersen (AP12009) TGF-β antisense
VDR Calcitriol, Paricalcitol, Doxercalceferiol VDR agonists Hypercalcemia
VEGF Bevacizumab VEGF antibody Proteinuria, Hypertension
Ramucirumab VEGFR2 antibody Thrombotic microangiopathy Edema
Sorafenib Sunitinib Small molecule VEGFR2 inhibitors Ascites
Wnt/β-catenin ICG-001 Inhibits β-catenin induced transcription
*

Available drug products are dietary supplements or drugs approved for an indication other than DN. Investigational drug products are drugs used in animal studies or in early phase clinical trials.

**

5-aminoimidazole-4-carboxamide-1-β-d-ribonucleoside,

Valine-isoleucine-valine-isoleucine-threonine.

Table 2.

Selected ongoing and recently completed clinical trials in diabetic nephropathy

Trial/phase Drug Mechanism(s) of action Patient characteristics Primary outcome
NCT01712061 (Phase 2) PF-04634817 CCR2/5 antagonist Type 2 diabetes, CKD stages 2–4, urine ACR*>300 mg/gram Change in urine ACR* from baseline
NCT01752985 (Phase 2) BMS-813160 CCR2/5 antagonist Type 2 diabetes, urine ACR* 200–3500 mg/gram Change in urine ACR* from baseline
NCT01447147 (Phase 2) CCX140-B CCR2 antagonist Type 2 diabetes, urine ACR* 100–3000 mg/gram eGFR ≥25 ml/min/1.73m2 Safety and tolerability
NCT01440257 (Phase 2) CCX140-B CCR2 antagonist Type 2 diabetes, urine ACR* ≥200–3000 mg/gram, eGFR ≥25 ml/min/1.73m2 Change in 24 h urine ACR*
NCT02410499 (Phase 2) MLN1202 CCR2 monoclonal antibody Type 2 diabetes, albuminuria, eGFR 25–59 ml/min/1.73m2 Change in urine ACR*
NCT01858532 (Phase 3) SONAR Atrasentan Selective ETA antagonist Type 2 diabetes, urine ACR* ≥300 and <5000 mg/gram, eGFR 25–75 ml/min/1.73m2 and BNP ≤200 ng/l Doubling of serum creatinine or progression to ESRD
NCT01683409 (Phase 2) Barcitinib JAK1/2 inhibitor Type 2 diabetes, albuminuria, eGFR 25–70 ml/min/1.73m2 Change in urine ACR* after 24 wk
NCT02345057 (Phase 2) CS-3150 MR antagonist Type 2 diabetes, urine ACR* ≥45 and <300 mg/gram, eGFR >30 ml/min/1.73m2 Change in urine ACR*
NCT01968668 (Phase 1/2) ARTS-DN Japan Finerenone (BAY94-8862) MR antagonist Type 2 diabetes, urine ACR* ≥300 mg/gram, eGFR 30–90 ml/min/1.73m2 Change in urine ACR* at 90 days
NCT01874431 (Phase 2) ARTS-DN Finerenone (BAY94-8862) MR antagonist Type 2 diabetes, urine ACR* ≥300 mg/gram, eGFR 30–90 ml/min/1.73m2 Change in urine ACR* at 90 days
NCT01889277 (Phase 1/2) MT-3995 MR antagonist Type 2 diabetes, albuminuria Safety and tolerability
NCT02517320 (Phase 2) MT-3995 MR antagonist Type 2 diabetes, urine ACR* ≥50–300 mg/gram, eGFR ≥30 ml/min/1.73m2 Change in urine ACR* after 24 wk
NCT01726816 (Phase 2) Probucol Antioxidant Type 2 diabetes, urine ACR* ≥300 mg/gram, eGFR 15–90 ml/min/1.73m2 Change in urine ACR from baseline after 16 wk
NCT02316821 (Phase 2) RTA 402 Nrf2 activator Type 2 diabetes and chronic kidney disease Change in GFR as measured by inulin clearance
NCT02010242 (Phase 2) GKT137831 NOX1/4 inhibitor Type 2 diabetes, urine ACR* 300–3500 mg/gram, eGFR ≥30 ml/min/1.73m2 Change in urine ACR* at 8,10 and 12 wk
NCT01113801 (Phase 2) LY2382770 TGF-β antibody Type 1 or type 2 diabetes, urine ACR ≥800 mg/gram GFR 20–60 ml/min/1.73m2, Change in serum creatinine from baseline after 12 mo
NCT00552409 (Phase 2/3) Cholecalciferol Precusor of the VDR agonist calcitriol Type 2 diabetes, urine ACR* 30–1000 mg/gram, eGFR ≥60 ml/min/1.73m2 Change in 24 h urine ACR*
NCT01393808, (Phase 2) PROCEED Paricalcitol VDR agonist Type 2 diabetes, low or high salt diet, urine albumin >300 mg/24 h, creatinine <2 mg/dL Change in urine ACR* after 4 mo
NCT02410005 (Phase 2/3) IDEAL Calcitriol Losartan VDR agonist ARB§ Type 2 diabetes, urine ACR* >300 mg/gram, eGFR 30–90 ml/min/1.73m2 Change in 24 h albuminuria
*

Albumin creatinine ratio (ACR),

Estimated glomerular filtration rate (eGFR),

§

Angiotensin receptor blocker. Additional information on these studies is available at https://clinicaltrials.gov/.

A second promising approach to enhance RAS blockade is activating the vitamin D receptor (VDR), another nuclear receptor system. In addition to its well-known role in calcium homeostasis, the VDR ligand, 1,25-dihydroxyvitamin D3 has also been shown to negatively regulate renin gene expression in vitro (169) and in vivo (86). VDR knockout mice treated with the β-cell toxin streptozotocin (STZ) develop more severe kidney damage than wild-type mice (174), suggesting a protective role for VDR in diabetic nephropathy. Furthermore, treatment of diabetic mice with either paricalcitol or doxercalciferol abrogates renal damage by preserving glomerular basement membrane width, as well as blunting inflammation and fibrosis (30, 173, 175). In both cases, a synergistic effect with ARBs was observed. The mechanisms mediating the beneficial effects of VDR activation in diabetes are not known with certainty; however, it may be due to anti-inflammatory effects (29, 121) and/or by inhibiting the compensatory increase in renin expression that occurs in the setting of ARB therapy (175). VDR activation has also shown promise in clinical trials. In the VITAL (selective VITamin D receptor activator for Albuminuria Lowering) study, paricalcitol (2 μg per day) decreased urine albumin excretion by 18% compared with placebo in patients with Type 2 diabetes (26). As a result of these promising results, VDR agonists are currently being tested in additional clinical trials (Table 2).

Endothelin receptors.

The endothelin receptors, ETAR (ETA receptor) and ETBR, are G protein-coupled receptors that mediate the biological effects of endothelin peptides. The ETAR and ETBR are expressed throughout the kidney (12), and endothelin peptides are key regulators of blood pressure and sodium homeostasis (72). Both the ETAR and ETBR cause vasoconstriction; but the ETBR is also a vasodilator due to ETBR-induced generation of the vasodilator prostaglandins and nitric oxide (12). In the kidney, endothelins exert a net natriuretic effect that is largely mediated by the ETBR, although the hemodynamic and natriuretic actions of endothelins and their receptors in the kidney are complex (72). In addition to effects on vascular tone and natriuresis, endothelin receptors promote kidney injury. Serum levels of endothelin a (ET-1) are increased in both Type 1 and Type 2 diabetes (52), and overexpression of the human ET-1 in mice results in glomerulosclerosis and tubulointerstitial fibrosis (59). Infusion of ET-1 into rats produces a dose-dependent increase in glomerular permeability to albumin and increases glomerular expression of the inflammatory chemokine CCL2. These effects are blocked by a selective ETAR antagonist (120). Furthermore, deletion of both ETAR and ETBR from podocytes protects against glomerulosclerosis (82), while diabetic ETB receptor-deficient rats exhibit higher levels of albuminuria and enhanced glomerular injury compared with wild-type controls (115). Taken together, these data suggest that ETAR receptor activation promotes podocyte damage and albuminuria.

This differential role of the ETAR and ETBR in the kidney is reflected in the effects of receptor antagonists. For example, treatment with the nonspecific ETAR/ETBR antagonist bosentan did not improve renal pathology or albumin excretion in hypertensive diabetic rats (69). In contrast, selective ETAR blockade with avosentan, alone or in combination with lisinopril, improved albuminuria and glomerulosclerosis in uninephrectomized, diabetic rats (43). ETAR antagonists have also shown promise in clinical trials. Avosentan significantly reduced albuminuria in patients with either Type 1 or Type 2 diabetes and macroalbuminuria on standard RAS blockade (162). Similarly, treatment with the highly selective ETA antagonist atrasentan significantly reduced the urinary albumin to creatinine ratio in patients with diabetic nephropathy already receiving stable doses of RAS inhibitors (28, 71). The main impediment to using ETA antagonists for the treatment of DN is fluid retention, which results in edema and may aggravate heart failure (28, 71, 93, 162). While promising, it remains to be seen whether these off target effects can be overcome by more selective ETAR antagonists.

Chemokine receptors.

Another promising target for the treatment of diabetes includes the family of chemokine receptors, which are predominantly expressed on T cells and monocytes (163). These GPCRs play a critical role in chronic inflammatory diseases (163). Accumulating evidence suggests that chronic inflammation also plays an important role in metabolic diseases linked to obesity, including insulin resistance and diabetes mellitus (163). Adipose tissue produces both CCL2 and CCL5 (46, 143, 165), which bind to the chemokine receptors CCR2 and CCR5, respectively (163). Both ligands are upregulated in obese rodents (46, 143, 165). In obese mice, enhanced chemokine levels are associated with an increased number of inflammatory cells in adipose tissue (161, 165), which ostensibly causes insulin resistance by indirect mechanisms. In addition, the CCL2-CCR axis may have a direct role in insulin resistance because treatment with CCL2 reduces insulin-stimulated glucose uptake in cultured adipocytes (125). In support of an important role for CCR2 signaling in diabetes, knockout of CCR2 improves insulin resistance and fasting blood glucose levels in obese mice (161). On the basis of these results, small-molecule CCR2 antagonists have been developed and are being tested in preclinical animal studies (67, 126, 135). Moreover, several pharmacological agents have entered phase 2 clinical trials, including studies of patients with overt diabetic nephropathy (see Tables 1 and 2) (55, 163). Preliminary results of these clinical studies suggest that CCR2 antagonism reduces both fasting plasma glucose and hemoglobin A1c levels (55). It remains to be determined, however, whether or not these pharmacological agents will have beneficial effects on diabetic kidney disease, independent of effects on blood glucose levels.

Adenosine receptors.

Activation of adenosine receptor subtypes has both hemodynamic and anti-inflammatory actions in the kidney (79, 144). For example, glomerular hyperfiltration in diabetic kidney disease can be inhibited by the adenosine reuptake inhibitor dipyridamole (145). More recent studies using small-molecule agonists and antagonists found that the adenosine A2A receptor-mediated arteriolar vasodilation in diabetes-induced glomerular hyperfiltration (114). Other studies have investigated the anti-inflammatory effects of A2A receptor signaling in animal models of diabetic kidney disease. Awad et al. (4) found that a selective A2A agonist reduced albuminuria, mesangial expansion, and glomerular basement membrane width in diabetic rats. Moreover, knockout of the A2A receptor aggravated diabetic kidney disease in this same rodent model (4). Similarly, A2A receptor activation reduced proteinuria and promoted an anti-inflammatory phenotype characterized by reduced proinflammatory tumor necrosis factor-α (TNF-α) levels and enhanced anti-inflammatory IL-10 levels, as well as decreased macrophage infiltration and glomerular injury (113). Taken together, these data suggest that the A2A receptor may be a novel future therapeutic target for the treatment of DN.

Regulation of Inflammation and Fibrosis by Transforming growth Factor-β

The secreted protein-transforming growth factor-β (TGF-β) is a master regulator of inflammation and fibrosis. Activation of TGF-β in the kidney initiates a profibrotic program characterized by extracellular matrix production, cellular hypertrophy, basement membrane thickening, and apoptosis. These changes eventually lead to glomerulosclerosis and tubulointerstitial fibrosis (89). In CKD, the link between TGF-β and fibrosis has been well established, and a full discussion of the topic is beyond the scope of this review. Therefore, we will focus on the therapeutic potential of TGF-β inhibition. To assess the role of TGF-β in early diabetes, Sharma et al. (131, 133) treated mice with STZ and then measured the expression of TGF-β. Within 3 days of STZ administration, the expression of TGF-β increased in the kidney cortex. Treating the mice with an anti-TGF-β antibody blocked the increase and also inhibited glomerular hypertrophy and upregulation of collagen α1, suggesting that TGF-β induction by hyperglycemia in vivo is an early event and highlighting the potential of a neutralizing anti-TGF-β antibody. In a long-term experiment, this same anti-TGF-β antibody was administered to db/db mice (a genetic model of Type 2 diabetes) starting at the time when the mice become hyperglycemic. Again, anti-TGB-β antibody blocked the increase in collagen-α1 and fibronectin in the kidney cortex (179). However, it had no effect on albuminuria (179). Given that DN is often well established before renoprotective therapies are initiated, strategies to ameliorate established DN are required. In this regard, treatment with an anti-TGF-β antibody reduced glomerular basement membrane thickness and mesangial expansion in db/db mice with established diabetic kidney disease (17). In another study, diabetic rats were treated with either a murine or human anti-TGF-β antibody in combination with lisinopril (8). In the absence of additional treatment, both murine and human antibodies decreased blood pressure, albuminuria, and fibrosis. The human anti-TGF-β antibody was also able to abrogate glomerulosclerosis and had a synergistic effect with lisinopril (8). As a result, a phase 2 clinical trial is studying an anti-TGF-β antibody in patients with DN (Table 2) (1). The TGF-β pathway has also been targeted with pirfenidone, a synthetic compound that inhibits TGF-β promoter activity in vitro (117). Treatment of mouse mesangial cells with pirfenidone inhibited both TGF-β signaling and reactive oxygen species (ROS) production in a dose-dependent manner. Additionally, pirfenidone decreased mesangial expansion and the expression of collagen IV and fibronectin in db/db mice (117). A small randomized control trial to determine the effect of pirfenidone in diabetic nephropathy patients already on RAS blockade demonstrated a significant improvement in estimated glomerular filtration rate compared with placebo, but there was a high dropout rate in the pirfenidone group, which complicated interpretation of the study results (130).

Despite these promising findings, the use of TGF-β inhibitors for the treatment of fibrotic disease processes is tempered by the dual biological roles of TGF-β as both a profibrotic and anti-inflammatory mediator (167). For example, the use of a TGF-β-neutralizing antibody in patients with systemic sclerosis was complicated by serious adverse effects, including several deaths, with no evidence of a beneficial treatment effect (31). As a result, investigators have explored other approaches. One promising strategy is amplifying bone morphogenic protein (BMP) 7 signaling (167). BMP7 activates members of the TGF-β receptor superfamily and inhibits both inflammation and fibrosis (167). BMP7 is highly expressed in the kidney and overexpression of BMP7 in podocytes and proximal renal tubular cells (RTCs) reduces albuminuria, prevents podocyte loss, and decreases tubulointerstitial fibrosis (154). Similarly, pharmacological doses of BMP7 reduce albuminuria and glomerulosclerosis in diabetic rats (153). Unfortunately, the bioavailability of BMP7 in the kidney is quite low (167). A potential strategy to circumvent this problem is targeting the predominant BMP7 receptor in proximal RTCs, activin-like kinase 3 or Alk3 (141). Consistent with an important role for Alk3 in regulating fibrosis, knockout of Alk3 in proximal RTCs enhanced TGF-β signaling and increased kidney fibrosis (141). Other treatment strategies include targeting endogenous inhibitors of TGF-β signaling, such as gremlin and uterine sensitization gene-1 (167). Thus, while much work remains to be accomplished, significant advancement has been made in exploiting the TGF-β superfamily to treat the fibrotic disease process, including diabetic kidney disease.

Reducing Oxidative Stress in Diabetes

A large body of evidence links oxidative stress to the development of diabetic kidney disease (14, 139). The increase in reactive oxygen species is the result of both increased production, as well as reduced and/or inadequate antioxidant function (139). Sources of ROS include enzymatic activity, including xanthine oxidase, cytochrome P-450 enzymes, cyclooxygenases, and uncoupled nitric oxide synthase (NOS), as well as NADPH oxidases and mitochondrial superoxide production (139, 166). A large component of the increase in ROS generation in diabetes appears to be caused by hyperglycemia (139). Although not universally accepted (34), a prevailing theory suggests that hyperglycemia causes enhanced mitochondrial ROS generation (15). In this model, metabolism of glucose generates the electron donors NADH and FADH2, which donate their electrons to the mitochondrial electron transport chain leading to the production of ATP and water (15). Under hyperglycemic conditions, metabolism of glucose leads to excessive electron transport, eventually overloading the pathway and resulting in donation of electrons to molecular oxygen, generating superoxide (15). Another major source of ROS is NADPH oxidase (NOX) protein complexes (14, 139). Diabetes promotes increased expression of NOX proteins, resulting in enhanced superoxide generation. In diabetic animals, this pathway has been shown to be a major source of ROS (7, 15, 48). In support of an important role for NOX proteins in the pathogenesis of DN, NOX inhibitors reduce renal injury in animal models of diabetic kidney disease (142).

Counterbalancing excessive ROS generation is the antioxidant system, which includes superoxide dismutase, catalase, and the glutathione system (glucose-6 phosphate dehydrogenase, glutathione reductase, and glutathione peroxidase) (139). Numerous abnormalities of the antioxidant system have been described in animal models, including increased, decreased, and unchanged antioxidant levels (139). Overall, however, there does appear to be an inadequate antioxidant response, either as a result of altered levels and/or abnormal function (139). One consequence of enhanced ROS generation is excessive conversion of NAD to NADH (see above), resulting in depletion of cellular NAD. This cofactor is required to convert glyceraldehyde 3-phosphate to pyruvate during glycolysis, resulting in blockade of the glycolytic pathway and accumulation of pyruvate precursor compounds (15, 139). The metabolic intermediates can then be metabolized to advanced glycation end products (AGE), sorbitol, glucose-6-phosphate, and diacyl glyercol, an activator of PKC (15, 139). These compounds have numerous adverse effects, including enhancing expression of TGF-β, vascular endothelial growth factor (VEGF) and NADPH oxidases, activating NF-κB and decreasing expression of endothelial NOS (eNOS) (15). Exacerbating the decrease in eNOS expression is depletion of cofactors required for eNOS enzymatic activity as a result of oxidation by ROS (101). This leads to uncoupled eNOS activity, which results in the generation of superoxide instead of nitric oxide (NO) and, in turn, enhanced ROS generation (101).

The consequence of enhanced ROS generation is to oxidize adjacent molecules, including protein, lipids, nucleic acids, and carbohydrates (139). As a result, multiple cellular processes are disrupted, including organelle function, gene regulation, and cellular signaling, which ultimately may affect cell survival (139). A variety of genetic and pharmacological strategies have been used to “boost” the antioxidant system in diabetes. These strategies have shown promise in animal models of diabetic kidney disease. For example, treatment with the antioxidants vitamin E or probucol reduced ROS generation in STZ-treated diabetic rats (74). Another antioxidant α-lipoic acid inhibits podocyte loss, decreases albuminuria, and attenuates glomerulopathy in animal models of diabetes (137, 164). Alternatively, overexpression of an ubiquitously expressed superoxide dismutase transgene ameliorated kidney injury in diabetic mice (22, 32). Other groups have tried to target specific sources of ROS generation. This strategy has been used to reduce uncoupled eNOS activity in diabetes by supplementing cofactors oxidized in the diabetic environment (19). These supplements reduced albuminuria and glomerular basement membrane thickness in a mouse model of Type 2 diabetes (19). Unfortunately, the beneficial effects of antioxidant supplementation in animal models have not translated to beneficial effects in diabetic humans. For example, diabetic patients were treated with 400 IU vitamin E per day in the MICRO-HOPE clinical trial (microalbuminuria cardiovascular renal outcomes arm of HOPE) (90). In this study, vitamin E had no effect on either the development or progression of diabetic kidney disease (90). In contrast, treatment with the lipid-lowering and antioxidant probucol demonstrated a trend toward improved renal function, reduced proteinuria and less ESKD in diabetic patients over a three-year time period (36). This trial was, however, a small, open-label study, and it is difficult to discern whether the effects were directly related to probucol's antioxidant properties. Results of additional clinical trials (Table 2) may provide more definitive information on the efficacy of probucol in DN.

Another promising strategy to enhance the antioxidant system is targeting the transcription factor Nrf2 [nuclear factor (erythroid-derived 2)-like 2, also known as NFE2L2 or Nrf2], a key regulator of antioxidant protein expression (44). Nrf2 interacts with Keap 1 (Kelch-like-ECH-associated protein 1), which promotes ubiquitination and proteosomal degradation of Nrf2 (44). In diabetic rats, proteosomal inhibition upregulates expression of both Nrf2 and antioxidant genes (91). Thus, Nrf2 and the negative regulator Keap1 are attractive targets for modulating antioxidant expression in diabetes. In support of an important role for Nrf2 in DN, Nrf2 deletion exacerbates diabetic kidney disease in mice (66). Enthusiasm for this pathway in treating human disease stimulated a number of clinical trials examining Nrf2 agonists and Nrf2 inducers in diverse disease processes (reviewed in Ref. 44). In this regard, bardoxolone (Table 1) had shown great promise in an early-phase 2 clinical trial in patients with moderate to severe diabetic kidney disease (110, 111). Unfortunately, the phase 3 study was terminated early due to a higher rate of cardiovascular events (27). In the phase 3 study, treatment with bardoxolone improved glomerular filtration rate but increased albuminuria (27). Similarly, bardoxolone-treated monkeys also demonstrated enhanced proteinuria, likely as a result of decreased megalin-mediated protein uptake by the proximal tubule (119). It has been suggested that compounds such as bardoxolone may have a narrow therapeutic window and that higher dosages of the drug may stimulate expression of proinflammatory genes (53). Therefore, it is possible that titrating the dosage to maintain renal function, as opposed to improving renal function, may have a more favorable side effect profile (53). There is, however, a general concern that activation of the Nrf2 pathway may cause cancer cells to acquire a growth advantage (157). Indeed, Keap1 expression is downregulated in some cancers (157, 171). Thus, targeting the Nrf2/Keap1 system holds great promise, but further studies in animal models are needed to optimize this therapeutic approach.

Lastly, while there is strong evidence that ROS contribute to diabetic kidney disease, it is important to understand that ROS also play a key role in normal physiology. The system is designed to maintain ROS generation at some optimal level. For example, ROS play a role in cellular proliferation, and overexpression of antioxidant proteins inhibits cell growth and increases apoptosis (139). Another caveat is that blood glucose levels are often markedly and chronically elevated in the animal models used to study diabetic kidney disease compared with diabetic patients, who are usually receiving treatment for hyperglycemia. If ROS generation is largely driven by the severity of hyperglycemia, then the beneficial effects of antioxidant therapy in animal models may overestimate the beneficial effects of antioxidants in humans with diabetes, at least over the short term. Although we acknowledge that the time-averaged effect over decades of the disease could be similar, if may be difficult to design clinical trials to study the effects of antioxidant therapies in diabetic humans if detectable effects of the treatment may take decades to discern.

Dysregulated VEGF Expression in Diabetes

VEGF was discovered in 1983 and was initially named vasopermeability factor due to its ability to increase permeability of tumor-associated blood vessels (96). VEGF is now recognized as the critical angiogenic factor regulating endothelial cell migration, proliferation, differentiation, and cell survival by inhibiting apoptosis (96, 104). The VEGF family is composed of secreted, dimeric glycoproteins, including VEGF-A, VEGF-B, VEGF-C, VEGF-D, and placental growth factor (21, 96, 129). The VEGF-A (hereafter referred to as VEGF) is recognized as the prototypical family member and is the key regulator of physiological and pathological angiogenesis. Within the kidney, VEGF is expressed predominantly by podocytes but can also be detected in distal tubules, collecting ducts, and, to a lesser extent, the proximal tubules (21, 96, 129). Alternative splicing of VEGF results in multiple isoforms (96), which act by binding to receptor tyrosine kinases (RTKs) on target cells. The VEGF receptor 2 (VEGFR2, also termed Flk1 or KDR for kinase-insert domain receptor) is the RTK responsible for the known biological actions of VEGFR2 on endothelial cells (96, 104). A second receptor (VEGFR1 or Flt1) is also found on endothelium and seems to play a role in regulating endothelial cell VEGFR2 activity (104). VEGFR1 is, however, functional on other cell types. For example, VEGFR1 promotes hematopoietic stem cell recruitment and monocyte/macrophage migration (96, 104). In the kidney, VEGFR2 expression has been reported on glomerular endothelial cells, peritubular endothelial cells, medullary interstitial cells, mesangial cells, and glomerular podocytes (10, 21, 96, 147). Expression of VEGFR2 on podocytes is, however, controversial with several groups unable to detect VEGFR2 mRNA or protein in cultured podocytes (18, 41) or in vivo (136), although all of these studies were able to detect podocyte expression of VEGFR1 (18, 41, 136). Indeed, the biological effects of VEGF on podocytes may be mediated by activating VEGFR1, including antiapoptotic actions (41) and stimulating synthesis of glomerular basement membrane components (18). Given the proximity of podocytes to glomerular endothelial cells and mesangial cells, both autocrine and paracrine signaling has been proposed to contribute to VEGF actions within the glomerulus (96). Although recent studies have questioned the presence of autocrine VEGF signaling under normal physiological conditions (136), the presence of both paracrine and autocrine signaling is supported by both cell culture experiments (18, 41) and in vivo studies (147) and might be mediated by activating VEGFR1.

In diabetes, renal VEGF expression is markedly upregulated with prominent VEGF expression in glomerular podocytes (21, 96, 129). This enhanced expression is, in part, related to activation of mammalian target of rapamycin (mTOR) target genes, as well as blockade of glycolytic pathways [see oxidative stress above and (15)]. Within the diabetic kidney, the effects of VEGF are complex. In endothelial cells, VEGF induces eNOS expression and NO release, which under normal conditions, promotes vasorelaxation, inhibits VEGF-induced capillary growth, blocks endothelial cell activation by inflammatory cytokines, such as TNF and inhibits release of prothrombotic von Willebrand factor from Weibel-Palade bodies (101). In the diabetic milieu, however, VEGF stimulates “uncoupled” NO generation and, in turn, NO production is decreased, and generation of ROS is enhanced (101). In this model, enhanced VEGF generation in diabetes might have adverse effects on endothelial cells by stimulating apoptosis-promoting ROS generation, endothelial cell activation, and intravascular thrombosis. In podocytes, the proapoptotic effect of ROS may be mitigated by prosurvival, autocrine VEGF signaling. In this scenario, inhibition of paracrine VEGF signaling in endothelial cells would be predicted to have beneficial effects in diabetic kidney disease. In contrast, inhibition of autocrine VEGF signaling in podocytes would be predicted to enhance podocyte loss. Thus, global inhibition of VEGF in diabetes might have both adverse and beneficial effects on the disease process.

The role of VEGF inhibition in diabetic kidney disease has been studied using rodent models of diabetes. In this regard, VEGF inhibition attenuates albuminuria and some histological features of diabetic kidney disease in Type 1 and Type 2 diabetes (25, 39, 76). For example, a VEGF antibody ameliorates diabetic kidney disease in rodent models of Type 2 diabetes (25, 39). Moreover, treatment with a soluble splice variant of VEGFR1 (sFlt1) reduced albuminuria, mesangial expansion, and glomerular basement membrane (GBM), thickening in diabetic mice (76). In support of a pathogenetic role for VEGF in diabetic nephropathy, overexpression of VEGF in podocytes causes proteinuria, GBM thickening, and mesangial expansion (146, 147). Enthusiasm for these promising findings is, however, tempered by the observation that the absence of podocyte VEGF enhances glomerular injury in diabetic mice (138), suggesting some basal level of VEGF expression is required for glomerular maintenance.

The effects of VEGF are also regulated by additional angiogenic mediators. Angiopoietin-1 (Angpt1) is produced by specialized pericytes, such as podocytes and mesangial cells (65). It binds to the Tie-2 receptor on endothelial cells and maintains endothelial cell quiescence and constrains angiogenesis and fibrosis following vessel injury (65). A second Angpt family member, Angpt2, is produced by endothelial cells and is the natural antagonist of Angpt1 (24). In diabetes, expression of Angpt1 and Angpt2 is altered, resulting in an elevation in the Angpt2/Angpt1 ratio (65). In support of an important role for the Angpts in DN, deletion of glomerular Angpt1 enhances albuminuria and the severity of glomerular injury in diabetic kidney disease. Moreover, enhancing glomerular expression of Angpt1 ameliorates glomerular damage in diabetic mice (33, 80). Thus, angiogenic signaling is a complex process with multiple, mutually antagonistic mediators.

While the Tie2 receptor system is also a potential target for developing therapeutic agents, a large number of VEGF inhibitors have been developed and are used extensively for the treatment of solid tumors, including renal cell carcinoma, hepatocellular carcinoma, gastrointestinal stromal tumors, and soft tissue sarcomas (40). These drugs include blocking antibodies and small-molecule inhibitors (Table 1) (40). While the clinical use of these agents has been associated with a low risk of adverse events (40), 1–2% of patients develop proteinuria (178), and a small percentage develop thrombotic microangiopathy (38). A pathogenic role for VEGF in the latter is suggested by the observation that animals lacking glomerular VEGF develop a thrombotic glomerular disease (38). Given adverse effects of current VEGF inhibitors, as well as complexity of VEGF signaling within the glomerulus, a more complete understanding of VEGF biology in diabetes will be essential for designing effective treatment strategies that target angiogenic signaling in DN.

Activation of Janus Kinases and Signal Transducers and Activators of Transcription in DN

JAKs are a group of tyrosine kinases that mediate cellular response to cytokines and chemokines through interactions with STAT transcription regulators (47). The JAK/STAT pathway is involved in several developmental processes, including hematopoiesis, immune system development, and cell growth (47). In canonical JAK/STAT signaling, the pathway is activated by extracellular ligand binding to its receptor. This receptor-ligand interaction results in JAK phosphorylation. Phosphorylated JAKs then phosphorylate and activate cytoplasmic STAT proteins. Activated STATs translocate to the nucleus, where they bind to target genes (57). One group of STAT target genes, the suppressors of cytokine signaling (SOCS) family of proteins, inhibit JAK/STAT phosphorylation, thus creating a negative feedback loop (57). Altered JAK/STAT signaling has been implicated in several human diseases. For example, activating mutations in JAK2 were identified in myeloproliferative disorders (6, 75). In DN, a role for the JAK/STAT pathway in disease pathogenesis was described over a decade ago in a series of studies by Marrero et al. (95). These findings have more recently been supported by microarray analysis of RNA isolated from human kidney biopsies, which demonstrated significant upregulation of multiple JAK/STAT gene family members in both the glomerular and tubulointerstitial compartments (9). In this study, there was a significant inverse correlation between expression of JAK/STAT genes and glomerular filtration rate (9). The mechanisms promoting JAK/STAT activation in DN have not been fully elucidated; however, work by several groups found that ANG II and high glucose are upstream activators of JAK2, STATs 1, 3, and 5, as well as SOCS 1 and 3 in vitro (2, 95, 156). In vivo, ANG II infusion increases phosphorylation of JAK2 and STAT1 (58). Furthermore, treating diabetic rats with candesartan or the JAK2 inhibitor, AG490, decreases albuminuria (5). Taken together, these data implicate JAK/STAT family members in the pathogenesis of DN and suggest that inhibition of JAK/STAT might be beneficial. Currently, there are two JAK inhibitors on the market, tofacitinib, which preferentially inhibits JAKs 1 and 3 over JAK2, and ruxolitinib, which blocks JAK2 (Table 1) (122, 148). A major hurdle to using JAK inhibition in patients with DN is the risk of myelosuppression (148). JAK2, in particular, mediates signaling by several important hematopoetic cytokines, including erythropoietin and GM-CS (105). If these agents are proven effective for the treatment of human diabetic kidney disease (Table 2), there is likely to be a significant risk-benefit ratio to consider and, in turn, this class of medications may only be considered for the patients with the greatest risk for disease progression.

Enhanced Calcineurin/Nuclear Factor of Activated T-Cell Signaling in DN

CN is a calcium-activated phosphatase that plays a key role in diverse disease processes. An important CN substrate is the family of NFAT transcription factors (60). NFAT family members were originally discovered in cells of the lymphoid lineage, but abundant evidence indicates that NFAT isoforms are expressed in nonimmune cells with some family members expressed ubiquitously (61). In quiescent cells, NFAT isoforms are phosphorylated and located in the cytoplasm (61). CN dephosphorylates NFAT, which permits translocation to the nucleus and stimulation of gene transcription.

A growing literature suggests that CN is activated in kidneys of diabetic rodents (50, 150). This increase in CN activation may contribute to the apoptosis of cell types that play an important role in the pathogenesis of DN, including renal tubular cells (20) and podocytes (85, 150). CN directly promotes apoptosis through dephosphorylation of the proapoptotic protein BAD (155) and Drp1 (134). CN may also indirectly cause apoptosis by stimulating gene transcription. In this regard, a cell-permeable peptide inhibitor termed VIVIT that specifically blocks CN-dependent NFAT activation attenuates apoptosis of cultured podocytes (85, 150). Indeed, expression of a constitutively active NFAT isoform specifically in podocytes promotes proteinuria, glomerulosclerosis, and a decrease in podocyte numbers (159). Important gene targets of CN include TRPC6 (transient receptor potential cation channel C6), COX2 (cyclooxygenase 2), and RCAN1 (regulator of CN 1) (77, 152). These CN gene targets are upregulated in animal models of diabetic kidney disease and in patients with diabetes (73, 109). In support of a pathogenic role for CN in DN, inhibition CN ameliorates animal models of diabetic kidney disease (50, 116, 172). For example, CN is required for glomerular hypertrophy and extracellular matrix accumulation in diabetic rats (50). In diabetic rodents, albuminuria, extracellular matrix accumulation, and interstitial injury are attenuated by either pharmacologic CN inhibition (116) or the NFAT inhibitor VIVIT (172). The beneficial effects of VIVIT suggest that NFAT-mediated gene transcription is, at least in part, responsible for the beneficial effects of CN inhibition in diabetic kidney disease.

Currently available pharmacological CN inhibitors include cyclosporine A and tacrolimus/FK506 (Table 1) (45). Both of these drugs are immunosuppressive agents (45). As with JAK-STAT and mTOR inhibitors, the risk-benefit ratio will have to be carefully considered if CN inhibition is undertaken for treatment of DN. Moreover, both cyclosporine A and FK506 have adverse effects, such as nephrotoxicity and promoting the development of posttransplant diabetes. The development, however, of pharmacological agents that selectively inhibit CN-NFAT signaling (3) without affecting dephosphorylation of other CN substrates may provide a therapeutic strategy for inhibiting the CN-NFAT signaling pathway, while avoiding the adverse side effects of pharmacologic CN inhibitors (168). In this regard, the cell-permeable peptide NFAT inhibitor VIVIT prevented rejection of pancreatic islets in vivo without the adverse effects on glucose metabolism (102). Further research is needed but, if these initial findings are confirmed, then the results might provide the impetus for the development of highly specific pharmacological agents that block CN-dependent NFAT signaling without some of the adverse side effects of current pharmacological CN inhibitors. Indeed, a recent study identified dipyridamole as a small-molecule inhibitor of CN-dependent NFAT activation that does not affect CN phosphatase activity (99).

Regulation of Cell Metabolism by mTOR/AMPK

The serine/threonine kinase mTOR regulates cell growth and metabolism. mTOR forms two distinct signaling complexes called mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2) (78). mTORC1 integrates input from both intracellular and extracellular signals to regulate several processes, including lipid metabolism, protein synthesis, and autophagy (35). Upstream regulators of mTORC1 include oxygen, growth factors, and amino acids (35, 78). Emerging evidence supports the role of mTORC1 in renal development and kidney disease. Embryonic deletion of the mTORC1 component Raptor in mouse podocytes causes robust albuminuria and glomerulosclerosis (49). This phenotype is present, although less severe when Raptor is deleted in adult mice (49). Moreover, genetic activation of mTORC1 by deletion of an upstream inhibitor in podocytes recapitulates histologic features of DN, including mesangial expansion and glomerulosclerosis (62), and genetic inhibition of mTORC1 by deletion of one copy of Raptor confers resistance to albuminuria and mesangial expansion in a mouse model of Type 2 diabetes (62). In support of an important role for enhanced mTORC1 activity in diabetic kidney disease, phosphorylation of a downstream target of mTORC1 (p70S6 kinase) is enhanced in glomeruli from diabetic mice and in biopsy specimens from patients with DN (49, 98). Taken together, these findings support an important role for mTORC1 in diabetic kidney disease and suggest that a balance of mTORC1 signaling is required for maintenance of glomerular integrity. Targeting this pathway using pharmacological inhibitors may, therefore, have a narrow therapeutic window. Preclinical studies in rodents, however, suggest that this strategy is feasible because mTORC1 inhibition ameliorates mesangial expansion and interstitial fibrosis in STZ-treated Sprague-Dawley rats (88). There are currently two mTOR inhibitors in clinical use, sirolimus (also known as rapamycin) and everolimus (Table 1). Unfortunately, these drugs are immunosuppressive agents (149), which may limit their use in all but high-risk patients. In addition, both drugs have been associated with development of proteinuria (100), consistent with the narrow therapeutic window required to obtain a beneficial therapeutic effect. While the mechanisms of this proteinuric effect are not known with certainty, mTORC1 target genes include VEGF isoforms (49). As discussed above, VEGF plays a key role in maintenance of glomerular filtration barrier integrity. Alternatively, mTORC2 is required for activation of prosurvival Akt signaling (16, 118). While mTORC2 is classically thought to be insensitive to current mTOR inhibitors, recent studies suggest that both mTOR complexes can be inhibited by these agents (78, 124). Thus, one possible mechanism to reduce toxicity may be the development of more selective mTOR inhibitors, which preferentially target mTORC1 (149). mTORC2, however, also activates Rac 1 (64), and aberrant activation of Rho GTPases family members cause proteinuria (11, 151, 177). Thus, inhibition of mTORC2 signaling may also contribute to the beneficial effects of mTOR blockade in diabetic kidney disease. Additional studies will, therefore, be necessary to optimize this therapeutic approach.

Another therapeutic approach is to target upstream regulators of mTORC1 activity, such as AMPK. In this regard, AMPK is a potent negative regulator of mTORC1 (56, 132), and strategies to augment AMPK activity could theoretically have therapeutic benefits in DN, perhaps without immunosuppressive effects. In this regard, the adipocyte-secreted hormone adiponectin is a potent activator of AMPK activity (132). Adiponectin is a circulating plasma protein, and adiponectin serum levels negatively correlate with Type 2 diabetes, coronary artery disease, and obesity (132). In support of a role for adiponectin in DN, Sharma et al. (132) found that plasma adiponectin levels were inversely correlated with albuminuria in obese patients. Moreover, knockout of adiponectin caused albuminuria and foot process effacement in mice, which was improved with adiponectin treatment in association with AMPK activation. These beneficial effects were not the result of improved glucose control because this knockout model has normal glucose tolerance and insulin sensitivity. These data suggest that AMPK activation may be a useful therapeutic strategy for ameliorating the development of diabetes and DN. In support of this notion, treatment with the AMPK activator AICAR (5-aminoimidazole-4-carboxamide-1-β-d-ribonucleoside) (Table 1) improved albuminuria and kidney histology in two mouse models of diabetic kidney disease without altering blood glucose levels (34). Similarly, treatment with another AMPK activator metformin (Table 1) (56) reduced albuminuria and mesangial expansion in a rat model of Type 2 diabetes (70). Unfortunately, this study did not control for the beneficial effects of reduced hyperglycemia observed in the metformin-treated group. While these data are promising, additional basic research is needed. Moreover, given that metformin is already used clinically, examining this agent in humans with diabetic kidney disease may also be of benefit. Unfortunately, current guidelines limit the use of metformin in patients with CKD (63, 87), but carefully monitored clinical trials that control for effects of hyperglycemia in patients with DN and relatively well-preserved kidney function might be feasible.

Induction of Wnt/β-Catenin Signaling in DN

Wnt/β-catenin signaling was initially identified as a proto-oncogene in a mouse model of breast cancer (103). Since then, activated Wnt signaling has been identified in a variety of cancers and diverse biological processes, including embryonic patterning, bone biology, and stem cell maintenance (103). Recent studies have implicated Wnt/β-catenin signaling in the pathogenesis of DN. In this regard, components of the Wnt/β-catenin signaling cascade are upregulated in proteinuric renal diseases, including animal models of diabetic kidney disease and humans with DN (23). The mechanisms of Wnt/β-catenin gene induction are not known with certainty, but CN is activated in diabetic kidneys (50, 150), and expression of a constitutively active NFAT isoform in podocytes in vivo upregulates Wnt genes (159). Moreover, Wnt/β-catenin upregulates multiple genes of the RAS, suggesting a potentially important role for this pathway as a major mediator of kidney damage in DN (175). Small molecule inhibitors of Wnt/β-catenin signaling have been developed (Table 1), and treatment with these inhibitors has beneficial effects in animal models of glomerular disease (175). Thus, while the studies are still in their early stages, the Wnt/β-catenin signaling pathway looks promising as a potential target for the treatment of DN.

Summary and Conclusions

We have attempted to summarize a large body of literature on emerging treatment strategies for DN. For this review, we focused predominantly on signaling pathways that have been implicated relatively recently and selected pathways in which therapeutic agents and/or small molecule inhibitors were readily available (Table 1). Unfortunately, many of these inhibitors have significant risks associated with their use, including currently available inhibitors of JAK/STAT, mTOR, CN, and VEGF signaling. As a result, using inhibitors of these pathways to treat diabetic patients will likely require identifying patients at high risk for disease progression. In contrast, other treatment strategies look more promising as a general approach to patients with DN. Combined therapies using aldosterone antagonists, endothelin inhibitors, and chemokine receptor blockers with RAS blockade are an obvious approach that is currently being studied in clinical trials (Table 2). Another approach is the use of AMPK activators. Metformin activates AMPK and could be studied in clinical trials, but it may be difficult to control for effects of treatment on hyperglycemia and insulin sensitivity. Moreover, the use of metformin is restricted to patients with relatively normal renal function (63, 87), although a number of investigators have suggested that the guidelines for using this agent in patients with CKD could be liberalized (63, 87). Additional AMPK activators are available that have beneficial effects in animal models of diabetic kidney disease without affecting blood glucose levels (34), but these agents would need to be approved for use in humans if clinical studies are considered. Vitamin D is another agent that has demonstrated beneficial effects in animal models of diabetic kidney disease (30, 158, 160, 173) and has been shown to lower albuminuria in macroalbuminuric patients with Type 2 diabetes treated with RAS blockade (26). Vitamin D supplementation might, therefore, be a useful adjunction to current therapeutic approaches in DN that could also be rigorously tested in clinical trials (Table 2). Lastly, there may be strategies to reduce toxicities of other pharmacologic agents with a significant risk of adverse effects. For example, the development of mTOR inhibitors, which are more selective for mTORC1 or CN inhibitors that specifically block CN-NFAT signaling without affecting other signaling cascades (see Regulation of Cell Metabolism by mTOR/AMPK and Enhanced CN/NFAT Signaling in DN). Modification of the dosing regimens used for bardoxolone-like drugs may also be of benefit (see Reducing Oxidative Stress in Diabetes). In summary, promising new therapeutic approaches for the treatment of diabetic kidney disease are rapidly expanding. While more research is needed, a major challenge to the development of new therapies will be deciding which promising ideas to pursue and then translating these ideas into innovative health care strategies for treating patients with diabetic kidney disease.

GRANTS

R. F. Spurney received salary support through grants from the National Institutes of Health, National Institute of Diabetes, Digestive and Kidney Diseases (RO1 DK-087707, RO1 DK-094987, and RO1 DK-103694), as well as the Duke O'Brien Center for Kidney Research (P30 DK-096493). S. A. Johnson received salary support through a postdoctoral fellowship from the Edna and Fred L. Mandel, Jr. Center for Hypertension and Atherosclerosis research and from a research fellowship from the American Society of Nephrology Foundation for Kidney Research (Ben J. Lipps Research Fellowship).

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the authors.

AUTHOR CONTRIBUTIONS

Author contributions: S.A.J. and R.F.S. drafted manuscript; S.A.J. and R.F.S. edited and revised manuscript; R.F.S. approved final version of manuscript.

REFERENCES

  • 1.Akhurst RJ, Hata A. Targeting the TGF-β signalling pathway in disease. Nat Rev Drug Discov 11: 790–811, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Amiri F, Shaw S, Wang X, Tang J, Waller JL, Eaton DC, Marrero MB. Angiotensin II activation of the JAK/STAT pathway in mesangial cells is altered by high glucose. Kidney Int 61: 1605–1616, 2002. [DOI] [PubMed] [Google Scholar]
  • 3.Aramburu J, Yaffe MB, Lopez-Rodriguez C, Cantley LC, Hogan PG, Rao A. Affinity-driven peptide selection of an NFAT inhibitor more selective than cyclosporin A. Science 285: 2129–2133, 1999. [DOI] [PubMed] [Google Scholar]
  • 4.Awad AS, Huang L, Ye H, Duong ET, Bolton WK, Linden J, Okusa MD. Adenosine A2A receptor activation attenuates inflammation and injury in diabetic nephropathy. Am J Physiol Renal Physiol 290: F828–F837, 2006. [DOI] [PubMed] [Google Scholar]
  • 5.Banes AK, Shaw S, Jenkins J, Redd H, Amiri F, Pollock DM, Marrero MB. Angiotensin II blockade prevents hyperglycemia-induced activation of JAK and STAT proteins in diabetic rat kidney glomeruli. Am J Physiol Renal Physiol 286: F653–F659, 2004. [DOI] [PubMed] [Google Scholar]
  • 6.Baxter EJ, Scott LM, Campbell PJ, East C, Fourouclas N, Swanton S, Vassiliou GS, Bench AJ, Boyd EM, Curtin N, Scott MA, Erber WN, Green AR. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet 365: 1054–1061, 2005. [DOI] [PubMed] [Google Scholar]
  • 7.Bedard K, Krause KH. The NOX family of ROS-generating NADPH oxidases: physiology and pathophysiology. Physiol Rev 87: 245–313, 2007. [DOI] [PubMed] [Google Scholar]
  • 8.Benigni A, Zoja C, Corna D, Zatelli C, Conti S, Campana M, Gagliardini E, Rottoli D, Zanchi C, Abbate M, Ledbetter S, Remuzzi G. Add-on anti-TGF-beta antibody to ACE inhibitor arrests progressive diabetic nephropathy in the rat. J Am Soc Nephrol 14: 1816–1824, 2003. [DOI] [PubMed] [Google Scholar]
  • 9.Berthier CC, Zhang H, Schin M, Henger A, Nelson RG, Yee B, Boucherot A, Neusser MA, Cohen CD, Carter-Su C, Argetsinger LS, Rastaldi MP, Brosius FC, Kretzler M. Enhanced expression of Janus kinase-signal transducer and activator of transcription pathway members in human diabetic nephropathy. Diabetes 58: 469–477, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Bertuccio C, Veron D, Aggarwal PK, Holzman L, Tufro A. Vascular endothelial growth factor receptor 2 direct interaction with nephrin links VEGF-A signals to actin in kidney podocytes. J Biol Chem 286: 39,933–39,944, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Blattner SM, Hodgin JB, Nishio M, Wylie SA, Saha J, Soofi AA, Vining C, Randolph A, Herbach N, Wanke R, Atkins KB, Gyung Kang H, Henger A, Brakebusch C, Holzman LB, Kretzler M. Divergent functions of the Rho GTPases Rac1 and Cdc42 in podocyte injury. Kidney Int 84: 920–930, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Boesen EI. Endothelin receptors, renal effects and blood pressure. Curr Opin Pharmacol 21: 25–34, 2015. [DOI] [PubMed] [Google Scholar]
  • 13.Brenner BM, Cooper ME, de Zeeuw D, Keane WF, Mitch WE, Parving HH, Remuzzi G, Snapinn SM, Zhang Z, Shahinfar S. Effects of losartan on renal and cardiovascular outcomes in patients with type 2 diabetes and nephropathy. N Engl J Med 345: 861–869, 2001. [DOI] [PubMed] [Google Scholar]
  • 14.Brosius FC 3rd, Alpers CE. New targets for treatment of diabetic nephropathy: what we have learned from animal models. Curr Opin Nephrol Hypertens 22: 17–25, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Brownlee M. The pathobiology of diabetic complications: a unifying mechanism. Diabetes 54: 1615–1625, 2005. [DOI] [PubMed] [Google Scholar]
  • 16.Canaud G, Bienaime F, Viau A, Treins C, Baron W, Nguyen C, Burtin M, Berissi S, Giannakakis K, Muda AO, Zschiedrich S, Huber TB, Friedlander G, Legendre C, Pontoglio M, Pende M, Terzi F. AKT2 is essential to maintain podocyte viability and function during chronic kidney disease. Nat Med 19: 1288–1296, 2013. [DOI] [PubMed] [Google Scholar]
  • 17.Chen S, Iglesias-de la Cruz MC, Jim B, Hong SW, Isono M, Ziyadeh FN. Reversibility of established diabetic glomerulopathy by anti-TGF-β antibodies in db/db mice. Biochem Biophys Res Commun 300: 16–22, 2003. [DOI] [PubMed] [Google Scholar]
  • 18.Chen S, Kasama Y, Lee JS, Jim B, Marin M, Ziyadeh FN. Podocyte-derived vascular endothelial growth factor mediates the stimulation of α3(IV) collagen production by transforming growth factor-β1 in mouse podocytes. Diabetes 53: 2939–2949, 2004. [DOI] [PubMed] [Google Scholar]
  • 19.Cheng H, Wang H, Fan X, Paueksakon P, Harris RC. Improvement of endothelial nitric oxide synthase activity retards the progression of diabetic nephropathy in db/db mice. Kidney Int 82: 1176–1183, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Cho SG, Du Q, Huang S, Dong Z. Drp1 dephosphorylation in ATP depletion-induced mitochondrial injury and tubular cell apoptosis. Am J Physiol Renal Physiol 299: F199–F206, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Cooper ME, Vranes D, Youssef S, Stacker SA, Cox AJ, Rizkalla B, Casley DJ, Bach LA, Kelly DJ, Gilbert RE. Increased renal expression of vascular endothelial growth factor (VEGF) and its receptor VEGFR-2 in experimental diabetes. Diabetes 48: 2229–2239, 1999. [DOI] [PubMed] [Google Scholar]
  • 22.Craven PA, Melhem MF, Phillips SL, DeRubertis FR. Overexpression of Cu2+/Zn2+ superoxide dismutase protects against early diabetic glomerular injury in transgenic mice. Diabetes 50: 2114–2125, 2001. [DOI] [PubMed] [Google Scholar]
  • 23.Dai C, Stolz DB, Kiss LP, Monga SP, Holzman LB, Liu Y. Wnt/β-catenin signaling promotes podocyte dysfunction and albuminuria. J Am Soc Nephrol 20: 1997–2008, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Davis B, Dei Cas A, Long DA, White KE, Hayward A, Ku CH, Woolf AS, Bilous R, Viberti G, Gnudi L. Podocyte-specific expression of angiopoietin-2 causes proteinuria and apoptosis of glomerular endothelia. J Am Soc Nephrol 18: 2320–2329, 2007. [DOI] [PubMed] [Google Scholar]
  • 25.de Vriese AS, Tilton RG, Elger M, Stephan CC, Kriz W, Lameire NH. Antibodies against vascular endothelial growth factor improve early renal dysfunction in experimental diabetes. J Am Soc Nephrol 12: 993–1000, 2001. [DOI] [PubMed] [Google Scholar]
  • 26.de Zeeuw D, Agarwal R, Amdahl M, Audhya P, Coyne D, Garimella T, Parving HH, Pritchett Y, Remuzzi G, Ritz E, Andress D. Selective vitamin D receptor activation with paricalcitol for reduction of albuminuria in patients with type 2 diabetes (VITAL study): a randomised controlled trial. Lancet 376: 1543–1551, 2010. [DOI] [PubMed] [Google Scholar]
  • 27.de Zeeuw D, Akizawa T, Audhya P, Bakris GL, Chin M, Christ-Schmidt H, Goldsberry A, Houser M, Krauth M, Lambers Heerspink HJ, McMurray JJ, Meyer CJ, Parving HH, Remuzzi G, Toto RD, Vaziri ND, Wanner C, Wittes J, Wrolstad D, Chertow GM. Bardoxolone methyl in type 2 diabetes and stage 4 chronic kidney disease. N Engl J Med 369: 2492–2503, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.de Zeeuw D, Coll B, Andress D, Brennan JJ, Tang H, Houser M, Correa-Rotter R, Kohan D, Lambers Heerspink HJ, Makino H, Perkovic V, Pritchett Y, Remuzzi G, Tobe SW, Toto R, Viberti G, Parving HH. The endothelin antagonist atrasentan lowers residual albuminuria in patients with type 2 diabetic nephropathy. J Am Soc Nephrol 25: 1083–1093, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Deb DK, Chen Y, Zhang Z, Zhang Y, Szeto FL, Wong KE, Kong J, Li YC. 1,25-Dihydroxyvitamin D3 suppresses high glucose-induced angiotensinogen expression in kidney cells by blocking the NF-κB pathway. Am J Physiol Renal Physiol 296: F1212–F1218, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Deb DK, Sun T, Wong KE, Zhang Z, Ning G, Zhang Y, Kong J, Shi H, Chang A, Li YC. Combined vitamin D analog and AT1 receptor antagonist synergistically block the development of kidney disease in a model of type 2 diabetes. Kidney Int 77: 1000–1009, 2010. [DOI] [PubMed] [Google Scholar]
  • 31.Denton CP, Merkel PA, Furst DE, Khanna D, Emery P, Hsu VM, Silliman N, Streisand J, Powell J, Akesson A, Coppock J, Hoogen F, Herrick A, Mayes MD, Veale D, Haas J, Ledbetter S, Korn JH, Black CM, Seibold JR. Recombinant human anti-transforming growth factor β1 antibody therapy in systemic sclerosis: a multicenter, randomized, placebo-controlled phase I/II trial of CAT-192. Arthritis Rheum 56: 323–333, 2007. [DOI] [PubMed] [Google Scholar]
  • 32.DeRubertis FR, Craven PA, Melhem MF, Salah EM. Attenuation of renal injury in db/db mice overexpressing superoxide dismutase: evidence for reduced superoxide-nitric oxide interaction. Diabetes 53: 762–768, 2004. [DOI] [PubMed] [Google Scholar]
  • 33.Dessapt-Baradez C, Woolf AS, White KE, Pan J, Huang JL, Hayward AA, Price KL, Kolatsi-Joannou M, Locatelli M, Diennet M, Webster Z, Smillie SJ, Nair V, Kretzler M, Cohen CD, Long DA, Gnudi L. Targeted glomerular angiopoietin-1 therapy for early diabetic kidney disease. J Am Soc Nephrol 25: 33–42, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Dugan LL, You YH, Ali SS, Diamond-Stanic M, Miyamoto S, DeCleves AE, Andreyev A, Quach T, Ly S, Shekhtman G, Nguyen W, Chepetan A, Le TP, Wang L, Xu M, Paik KP, Fogo A, Viollet B, Murphy A, Brosius F, Naviaux RK, Sharma K. AMPK dysregulation promotes diabetes-related reduction of superoxide and mitochondrial function. J Clin Invest 123: 4888–4899, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Efeyan A, Zoncu R, Sabatini DM. Amino acids and mTORC1: from lysosomes to disease. Trends Mol Med 18: 524–533, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Endo K, Miyashita Y, Sasaki H, Ohira M, Saiki A, Koide N, Otsuka M, Oyama T, Takeyoshi M, Ito Y, Shirai K. Probucol delays progression of diabetic nephropathy. Diabetes Res Clin Pract 71: 156–163, 2006. [DOI] [PubMed] [Google Scholar]
  • 37.Epstein M, Williams GH, Weinberger M, Lewin A, Krause S, Mukherjee R, Patni R, Beckerman B. Selective aldosterone blockade with eplerenone reduces albuminuria in patients with type 2 diabetes. Clin J Am Soc Nephrol 1: 940–951, 2006. [DOI] [PubMed] [Google Scholar]
  • 38.Eremina V, Jefferson JA, Kowalewska J, Hochster H, Haas M, Weisstuch J, Richardson C, Kopp JB, Kabir MG, Backx PH, Gerber HP, Ferrara N, Barisoni L, Alpers CE, Quaggin SE. VEGF inhibition and renal thrombotic microangiopathy. N Engl J Med 358: 1129–1136, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Flyvbjerg A, Dagnaes-Hansen F, De Vriese AS, Schrijvers BF, Tilton RG, Rasch R. Amelioration of long-term renal changes in obese type 2 diabetic mice by a neutralizing vascular endothelial growth factor antibody. Diabetes 51: 3090–3094, 2002. [DOI] [PubMed] [Google Scholar]
  • 40.Fontanella C, Ongaro E, Bolzonello S, Guardascione M, Fasola G, Aprile G. Clinical advances in the development of novel VEGFR2 inhibitors. Ann Transl Med 2: 123, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Foster RR, Hole R, Anderson K, Satchell SC, Coward RJ, Mathieson PW, Gillatt DA, Saleem MA, Bates DO, Harper SJ. Functional evidence that vascular endothelial growth factor may act as an autocrine factor on human podocytes. Am J Physiol Renal Physiol 284: F1263–F1273, 2003. [DOI] [PubMed] [Google Scholar]
  • 42.Fried LF, Emanuele N, Zhang JH, Brophy M, Conner TA, Duckworth W, Leehey DJ, McCullough PA, O'Connor T, Palevsky PM, Reilly RF, Seliger SL, Warren SR, Watnick S, Peduzzi P, Guarino P. Combined angiotensin inhibition for the treatment of diabetic nephropathy. N Engl J Med 369: 1892–1903, 2013. [DOI] [PubMed] [Google Scholar]
  • 43.Gagliardini E, Corna D, Zoja C, Sangalli F, Carrara F, Rossi M, Conti S, Rottoli D, Longaretti L, Remuzzi A, Remuzzi G, Benigni A. Unlike each drug alone, lisinopril if combined with avosentan promotes regression of renal lesions in experimental diabetes. Am J Physiol Renal Physiol 297: F1448–F1456, 2009. [DOI] [PubMed] [Google Scholar]
  • 44.Gao B, Doan A, Hybertson BM. The clinical potential of influencing Nrf2 signaling in degenerative and immunological disorders. Clin Pharmacol Adv Applic 6: 19–34, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Gaston RS. Chronic calcineurin inhibitor nephrotoxicity: reflections on an evolving paradigm. Clin J Am Soc Nephrol 4: 2029–2034, 2009. [DOI] [PubMed] [Google Scholar]
  • 46.Gerhardt CC, Romero IA, Cancello R, Camoin L, Strosberg AD. Chemokines control fat accumulation and leptin secretion by cultured human adipocytes. Mol Cell Endocrinol 175: 81–92, 2001. [DOI] [PubMed] [Google Scholar]
  • 47.Ghoreschi K, Laurence A, O'Shea JJ. Janus kinases in immune cell signaling. Immunol Rev 228: 273–287, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Gill PS, Wilcox CS. NADPH oxidases in the kidney. Antiox Redox Signal 8: 1597–1607, 2006. [DOI] [PubMed] [Google Scholar]
  • 49.Godel M, Hartleben B, Herbach N, Liu S, Zschiedrich S, Lu S, Debreczeni-Mor A, Lindenmeyer MT, Rastaldi MP, Hartleben G, Wiech T, Fornoni A, Nelson RG, Kretzler M, Wanke R, Pavenstadt H, Kerjaschki D, Cohen CD, Hall MN, Ruegg MA, Inoki K, Walz G, Huber TB. Role of mTOR in podocyte function and diabetic nephropathy in humans and mice. J Clin Invest 121: 2197–2209, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Gooch JL, Barnes JL, Garcia S, Abboud HE. Calcineurin is activated in diabetes and is required for glomerular hypertrophy and ECM accumulation. Am J Physiol Renal Physiol 284: F144–F154, 2003. [DOI] [PubMed] [Google Scholar]
  • 51.Guo C, Martinez-Vasquez D, Mendez GP, Toniolo MF, Yao TM, Oestreicher EM, Kikuchi T, Lapointe N, Pojoga L, Williams GH, Ricchiuti V, Adler GK. Mineralocorticoid receptor antagonist reduces renal injury in rodent models of types 1 and 2 diabetes mellitus. Endocrinology 147: 5363–5373, 2006. [DOI] [PubMed] [Google Scholar]
  • 52.Haak T, Jungmann E, Felber A, Hillmann U, Usadel KH. Increased plasma levels of endothelin in diabetic patients with hypertension. Am J Hypertens 5: 161–166, 1992. [DOI] [PubMed] [Google Scholar]
  • 53.Hall ET, Bhalla V. Is there a sweet spot for Nrf2 activation in the treatment of diabetic kidney disease? Diabetes 63: 2904–2905, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Han SY, Kim CH, Kim HS, Jee YH, Song HK, Lee MH, Han KH, Kim HK, Kang YS, Han JY, Kim YS, Cha DR. Spironolactone prevents diabetic nephropathy through an anti-inflammatory mechanism in type 2 diabetic rats. J Am Soc Nephrol 17: 1362–1372, 2006. [DOI] [PubMed] [Google Scholar]
  • 55.Hanefeld M. Orally administered chemokine receptor CCR2 Antagonist CCX140-B in Type 2 diabetes: a pilot double-blind, randomized clinical trial. J Diabetes Metab 3: 225, 2012. [Google Scholar]
  • 56.Hardie DG. AMPK: a target for drugs and natural products with effects on both diabetes and cancer. Diabetes 62: 2164–2172, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Harrison DA. The Jak/STAT pathway. Cold Spring Harbor Perspect Biol 4: 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Hernandez-Vargas P, Lopez-Franco O, Sanjuan G, Ruperez M, Ortiz-Munoz G, Suzuki Y, Aguado-Roncero P, Perez-Tejerizo G, Blanco J, Egido J, Ruiz-Ortega M, Gomez-Guerrero C. Suppressors of cytokine signaling regulate angiotensin II-activated Janus kinase-signal transducers and activators of transcription pathway in renal cells. J Am Soc Nephrol 16: 1673–1683, 2005. [DOI] [PubMed] [Google Scholar]
  • 59.Hocher B, Thone-Reineke C, Rohmeiss P, Schmager F, Slowinski T, Burst V, Siegmund F, Quertermous T, Bauer C, Neumayer HH, Schleuning WD, Theuring F. Endothelin-1 transgenic mice develop glomerulosclerosis, interstitial fibrosis, and renal cysts but not hypertension. J Clin Invest 99: 1380–1389, 1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Hogan PG, Chen L, Nardone J, Rao A. Transcriptional regulation by calcium, calcineurin, and NFAT. Genes Dev 17: 2205–2232, 2003. [DOI] [PubMed] [Google Scholar]
  • 61.Horsley V, Pavlath GK. NFAT: ubiquitous regulator of cell differentiation and adaptation. J Cell Biol 156: 771–774, 2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Inoki K, Mori H, Wang J, Suzuki T, Hong S, Yoshida S, Blattner SM, Ikenoue T, Ruegg MA, Hall MN, Kwiatkowski DJ, Rastaldi MP, Huber TB, Kretzler M, Holzman LB, Wiggins RC, Guan KL. mTORC1 activation in podocytes is a critical step in the development of diabetic nephropathy in mice. J Clin Invest 121: 2181–2196, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Inzucchi SE, Lipska KJ, Mayo H, Bailey CJ, McGuire DK. Metformin in patients with type 2 diabetes and kidney disease: a systematic review. J Am Med Assoc 312: 2668–2675, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Jacinto E, Loewith R, Schmidt A, Lin S, Ruegg MA, Hall A, Hall MN. Mammalian TOR complex 2 controls the actin cytoskeleton and is rapamycin insensitive. Nat Cell Biol 6: 1122–1128, 2004. [DOI] [PubMed] [Google Scholar]
  • 65.Jeansson M, Gawlik A, Anderson G, Li C, Kerjaschki D, Henkelman M, Quaggin SE. Angiopoietin-1 is essential in mouse vasculature during development and in response to injury. J Clin Invest 121: 2278–2289, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Jiang T, Huang Z, Lin Y, Zhang Z, Fang D, Zhang DD. The protective role of Nrf2 in streptozotocin-induced diabetic nephropathy. Diabetes 59: 850–860, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Kang YS, Lee MH, Song HK, Ko GJ, Kwon OS, Lim TK, Kim SH, Han SY, Han KH, Lee JE, Han JY, Kim HK, Cha DR. CCR2 antagonism improves insulin resistance, lipid metabolism, and diabetic nephropathy in type 2 diabetic mice. Kidney Int 78: 883–894, 2010. [DOI] [PubMed] [Google Scholar]
  • 68.Kato S, Maruyama S, Makino H, Wada J, Ogawa D, Uzu T, Araki H, Koya D, Kanasaki K, Oiso Y, Goto M, Nishiyama A, Kobori H, Imai E, Ando M, Matsuo S. Anti-albuminuric effects of spironolactone in patients with type 2 diabetic nephropathy: a multicenter, randomized clinical trial. Clin Exp Nephrol 1–9, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Kelly DJ, Skinner SL, Gilbert RE, Cox AJ, Cooper ME, Wilkinson-Berka JL. Effects of endothelin or angiotensin II receptor blockade on diabetes in the transgenic (mRen-2)27 rat. Kidney Int 57: 1882–1894, 2000. [DOI] [PubMed] [Google Scholar]
  • 70.Kim J, Shon E, Kim CS, Kim JS. Renal podocyte injury in a rat model of type 2 diabetes is prevented by metformin. Exp Diabetes Res 2012: 210821, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Kohan DE, Pritchett Y, Molitch M, Wen S, Garimella T, Audhya P, Andress DL. Addition of atrasentan to renin-angiotensin system blockade reduces albuminuria in diabetic nephropathy. J Am Soc Nephrol 22: 763–772, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Kohan DE, Rossi NF, Inscho EW, Pollock DM. Regulation of blood pressure and salt homeostasis by endothelin. Physiol Rev 91: 1–77, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Komers R, Lindsley JN, Oyama TT, Schutzer WE, Reed JF, Mader SL, Anderson S. Immunohistochemical and functional correlations of renal cyclooxygenase-2 in experimental diabetes. J Clin Invest 107: 889–898, 2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Koya D, Hayashi K, Kitada M, Kashiwagi A, Kikkawa R, Haneda M. Effects of antioxidants in diabetes-induced oxidative stress in the glomeruli of diabetic rats. J Am Soc Nephrol 14: S250–S253, 2003. [DOI] [PubMed] [Google Scholar]
  • 75.Kralovics R, Passamonti F, Buser AS, Teo SS, Tiedt R, Passweg JR, Tichelli A, Cazzola M, Skoda RC. A gain-of-function mutation of JAK2 in myeloproliferative disorders. N Engl J Med 352: 1789–1790, 2005. [DOI] [PubMed] [Google Scholar]
  • 76.Ku CH, White KE, Dei Cas A, Hayward A, Webster Z, Bilous R, Marshall S, Viberti G, Gnudi L. Inducible overexpression of sFlt-1 in podocytes ameliorates glomerulopathy in diabetic mice. Diabetes 57: 2824–2833, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Kuwahara K, Wang Y, McAnally J, Richardson JA, Bassel-Duby R, Hill JA, Olson EN. TRPC6 fulfills a calcineurin signaling circuit during pathologic cardiac remodeling. J Clin Invest 116: 3114–3126, 2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Laplante M, Sabatini DM. mTOR signaling in growth control and disease. Cell 149: 274–293, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Lee HT, Gallos G, Nasr SH, Emala CW. A1 adenosine receptor activation inhibits inflammation, necrosis, and apoptosis after renal ischemia-reperfusion injury in mice. J Am Soc Nephrol 15: 102–111, 2004. [DOI] [PubMed] [Google Scholar]
  • 80.Lee S, Kim W, Moon SO, Sung MJ, Kim DH, Kang KP, Jang KY, Lee SY, Park BH, Koh GY, Park SK. Renoprotective effect of COMP-angiopoietin-1 in db/db mice with type 2 diabetes. Nephrol Dial Transpl 22: 396–408, 2007. [DOI] [PubMed] [Google Scholar]
  • 81.Lee SH, Yoo TH, Nam BY, Kim DK, Li JJ, Jung DS, Kwak SJ, Ryu DR, Han SH, Lee JE, Moon SJ, Han DS, Kang SW. Activation of local aldosterone system within podocytes is involved in apoptosis under diabetic conditions. Am J Physiol Renal Physiol 297: F1381–F1390, 2009. [DOI] [PubMed] [Google Scholar]
  • 82.Lenoir O, Milon M, Virsolvy A, Henique C, Schmitt A, Masse JM, Kotelevtsev Y, Yanagisawa M, Webb DJ, Richard S, Tharaux PL. Direct action of endothelin-1 on podocytes promotes diabetic glomerulosclerosis. J Am Soc Nephrol 25: 1050–1062, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Lewis EJ, Hunsicker LG, Bain RP, Rohde RD. The effect of angiotensin-converting-enzyme inhibition on diabetic nephropathy. The Collaborative Study Group. N Engl J Med 329: 1456–1462, 1993. [DOI] [PubMed] [Google Scholar]
  • 84.Lewis EJ, Hunsicker LG, Clarke WR, Berl T, Pohl MA, Lewis JB, Ritz E, Atkins RC, Rohde R, Raz I. Renoprotective effect of the angiotensin-receptor antagonist irbesartan in patients with nephropathy due to type 2 diabetes. N Engl J Med 345: 851–860, 2001. [DOI] [PubMed] [Google Scholar]
  • 85.Li R, Zhang L, Shi W, Zhang B, Liang X, Liu S, Wang W. NFAT2 mediates high glucose-induced glomerular podocyte apoptosis through increased Bax expression. Exp Cell Res 319: 992–1000, 2013. [DOI] [PubMed] [Google Scholar]
  • 86.Li YC, Kong J, Wei M, Chen ZF, Liu SQ, Cao LP. 1,25-Dihydroxyvitamin D3 is a negative endocrine regulator of the renin-angiotensin system. J Clin Invest 110: 229–238, 2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Lipska KJ, Bailey CJ, Inzucchi SE. Use of metformin in the setting of mild-to-moderate renal insufficiency. Diabetes Care 34: 1431–1437, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Lloberas N, Cruzado JM, Franquesa M, Herrero-Fresneda I, Torras J, Alperovich G, Rama I, Vidal A, Grinyo JM. Mammalian target of rapamycin pathway blockade slows progression of diabetic kidney disease in rats. J Am Soc Nephrol 17: 1395–1404, 2006. [DOI] [PubMed] [Google Scholar]
  • 89.Loeffler I, Wolf G. Transforming growth factor-beta and the progression of renal disease. Nephrol Dial Transpl 29 Suppl 1: i37–i45, 2014. [DOI] [PubMed] [Google Scholar]
  • 90.Lonn E, Yusuf S, Hoogwerf B, Pogue J, Yi Q, Zinman B, Bosch J, Dagenais G, Mann JF, Gerstein HC. Effects of vitamin E on cardiovascular and microvascular outcomes in high-risk patients with diabetes: results of the HOPE study and MICRO-HOPE substudy. Diabetes Care 25: 1919–1927, 2002. [DOI] [PubMed] [Google Scholar]
  • 91.Luo ZF, Qi W, Feng B, Mu J, Zeng W, Guo YH, Pang Q, Ye ZL, Liu L, Yuan FH. Prevention of diabetic nephropathy in rats through enhanced renal antioxidative capacity by inhibition of the proteasome. Life Sci 88: 512–520, 2011. [DOI] [PubMed] [Google Scholar]
  • 92.Maahs DM, West NA, Lawrence JM, Mayer-Davis EJ. Epidemiology of type 1 diabetes. Endocrinol Metab Clin North Am 39: 481–497, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Mann JF, Green D, Jamerson K, Ruilope LM, Kuranoff SJ, Littke T, Viberti G. Avosentan for overt diabetic nephropathy. J Am Soc Nephrol 21: 527–535, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Mann JF, Schmieder RE, McQueen M, Dyal L, Schumacher H, Pogue J, Wang X, Maggioni A, Budaj A, Chaithiraphan S, Dickstein K, Keltai M, Metsarinne K, Oto A, Parkhomenko A, Piegas LS, Svendsen TL, Teo KK, Yusuf S. Renal outcomes with telmisartan, ramipril, or both, in people at high vascular risk (the ONTARGET study): a multicentre, randomised, double-blind, controlled trial. Lancet 372: 547–553, 2008. [DOI] [PubMed] [Google Scholar]
  • 95.Marrero MB, Banes-Berceli AK, Stern DM, Eaton DC. Role of the JAK/STAT signaling pathway in diabetic nephropathy. Am J Physiol Renal Physiol 290: F762–F768, 2006. [DOI] [PubMed] [Google Scholar]
  • 96.Mironidou-Tzouveleki M, Tsartsalis S, Tomos C. Vascular endothelial growth factor (VEGF) in the pathogenesis of diabetic nephropathy of type 1 diabetes mellitus. Curr Drug Targets 12: 107–114, 2011. [DOI] [PubMed] [Google Scholar]
  • 97.Molitch ME, DeFronzo RA, Franz MJ, Keane WF, Mogensen CE, Parving HH, Steffes MW. Nephropathy in diabetes. Diabetes Care 27 Suppl 1: S79–S83, 2004. [DOI] [PubMed] [Google Scholar]
  • 98.Mori H, Inoki K, Masutani K, Wakabayashi Y, Komai K, Nakagawa R, Guan KL, Yoshimura A. The mTOR pathway is highly activated in diabetic nephropathy and rapamycin has a strong therapeutic potential. Biochem Biophys Res Commun 384: 471–475, 2009. [DOI] [PubMed] [Google Scholar]
  • 99.Mulero MC, Aubareda A, Orzaez M, Messeguer J, Serrano-Candelas E, Martinez-Hoyer S, Messeguer A, Perez-Paya E, Perez-Riba M. Inhibiting the calcineurin-NFAT (nuclear factor of activated T cells) signaling pathway with a regulator of calcineurin-derived peptide without affecting general calcineurin phosphatase activity. J Biol Chem 284: 9394–9401, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Murakami N, Riella LV, Funakoshi T. Risk of metabolic complications in kidney transplantation after conversion to mTOR inhibitor: a systematic review and meta-analysis. Am J Transplant 14: 2317–2327, 2014. [DOI] [PubMed] [Google Scholar]
  • 101.Nakagawa T, Tanabe K, Croker BP, Johnson RJ, Grant MB, Kosugi T, Li Q. Endothelial dysfunction as a potential contributor in diabetic nephropathy. Nat Rev Nephrol 7: 36–44, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Noguchi H, Matsushita M, Okitsu T, Moriwaki A, Tomizawa K, Kang S, Li ST, Kobayashi N, Matsumoto S, Tanaka K, Tanaka N, Matsui H. A new cell-permeable peptide allows successful allogeneic islet transplantation in mice. Nat Med 10: 305–309, 2004. [DOI] [PubMed] [Google Scholar]
  • 103.Nusse R, Varmus H. Three decades of Wnts: a personal perspective on how a scientific field developed. EMBO J 31: 2670–2684, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Olsson AK, Dimberg A, Kreuger J, Claesson-Welsh L. VEGF receptor signalling—in control of vascular function. Nat Rev Mol Cell Biol 7: 359–371, 2006. [DOI] [PubMed] [Google Scholar]
  • 105.Parganas E, Wang D, Stravopodis D, Topham DJ, Marine JC, Teglund S, Vanin EF, Bodner S, Colamonici OR, van Deursen JM, Grosveld G, Ihle JN. Jak2 is essential for signaling through a variety of cytokine receptors. Cell 93: 385–395, 1998. [DOI] [PubMed] [Google Scholar]
  • 106.Parving HH, Brenner BM, McMurray JJ, de Zeeuw D, Haffner SM, Solomon SD, Chaturvedi N, Persson F, Desai AS, Nicolaides M, Richard A, Xiang Z, Brunel P, Pfeffer MA. Cardiorenal end points in a trial of aliskiren for type 2 diabetes. N Engl J Med 367: 2204–2213, 2012. [DOI] [PubMed] [Google Scholar]
  • 107.Parving HH, Lehnert H, Brochner-Mortensen J, Gomis R, Andersen S, Arner P. The effect of irbesartan on the development of diabetic nephropathy in patients with type 2 diabetes. N Engl J Med 345: 870–878, 2001. [DOI] [PubMed] [Google Scholar]
  • 108.Parving HH, Persson F, Lewis JB, Lewis EJ, Hollenberg NK. Aliskiren combined with losartan in type 2 diabetes and nephropathy. N Engl J Med 358: 2433–2446, 2008. [DOI] [PubMed] [Google Scholar]
  • 109.Peiris H, Raghupathi R, Jessup CF, Zanin MP, Mohanasundaram D, Mackenzie KD, Chataway T, Clarke JN, Brealey J, Coates PT, Pritchard MA, Keating DJ. Increased expression of the glucose-responsive gene, RCAN1, causes hypoinsulinemia, beta-cell dysfunction, and diabetes. Endocrinology 153: 5212–5221, 2012. [DOI] [PubMed] [Google Scholar]
  • 110.Pergola PE, Krauth M, Huff JW, Ferguson DA, Ruiz S, Meyer CJ, Warnock DG. Effect of bardoxolone methyl on kidney function in patients with T2D and Stage 3b-4 CKD. Am J Nephrol 33: 469–476, 2011. [DOI] [PubMed] [Google Scholar]
  • 111.Pergola PE, Raskin P, Toto RD, Meyer CJ, Huff JW, Grossman EB, Krauth M, Ruiz S, Audhya P, Christ-Schmidt H, Wittes J, Warnock DG. Bardoxolone methyl and kidney function in CKD with type 2 diabetes. N Engl J Med 365: 327–336, 2011. [DOI] [PubMed] [Google Scholar]
  • 112.Persson F, Lewis JB, Lewis EJ, Rossing P, Hollenberg NK, Parving HH. Aliskiren in combination with losartan reduces albuminuria independent of baseline blood pressure in patients with type 2 diabetes and nephropathy. Clin J Am Soc Nephrol 6: 1025–1031, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Persson P, Friederich-Persson M, Fasching A, Hansell P, Inagi R, Palm F. Adenosine A a receptor stimulation prevents proteinuria in diabetic rats by promoting an anti-inflammatory phenotype without affecting oxidative stress. Acta Physiol (Oxf) 214: 311–318, 2015. [DOI] [PubMed] [Google Scholar]
  • 114.Persson P, Hansell P, Palm F. Reduced adenosine A2A receptor-mediated efferent arteriolar vasodilation contributes to diabetes-induced glomerular hyperfiltration. Kidney Int 87: 109–115, 2015. [DOI] [PubMed] [Google Scholar]
  • 115.Pfab T, Thone-Reineke C, Theilig F, Lange I, Witt H, Maser-Gluth C, Bader M, Stasch JP, Ruiz P, Bachmann S, Yanagisawa M, Hocher B. Diabetic endothelin B receptor-deficient rats develop severe hypertension and progressive renal failure. J Am Soc Nephrol 17: 1082–1089, 2006. [DOI] [PubMed] [Google Scholar]
  • 116.Qi XM, Wu YG, Liang C, Zhang P, Dong J, Ren KJ, Zhang W, Fang F, Shen JJ. FK506 ameliorates renal injury in early experimental diabetic rats induced by streptozotocin. Int Immunopharmacol 11: 1613–1619, 2011. [DOI] [PubMed] [Google Scholar]
  • 117.RamachandraRao SP, Zhu Y, Ravasi T, McGowan TA, Toh I, Dunn SR, Okada S, Shaw MA, Sharma K. Pirfenidone is renoprotective in diabetic kidney disease. J Am Soc Nephrol 20: 1765–1785, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Reiser J. Akt2 relaxes podocytes in chronic kidney disease. Nat Med 19: 1212–1213, 2013. [DOI] [PubMed] [Google Scholar]
  • 119.Reisman SA, Chertow GM, Hebbar S, Vaziri ND, Ward KW, Meyer CJ. Bardoxolone methyl decreases megalin and activates nrf2 in the kidney. J Am Soc Nephrol 23: 1663–1673, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Saleh MA, Boesen EI, Pollock JS, Savin VJ, Pollock DM. Endothelin-1 increases glomerular permeability and inflammation independent of blood pressure in the rat. Hypertension 56: 942–949, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Sanchez-Nino MD, Bozic M, Cordoba-Lanus E, Valcheva P, Gracia O, Ibarz M, Fernandez E, Navarro-Gonzalez JF, Ortiz A, Valdivielso JM. Beyond proteinuria: VDR activation reduces renal inflammation in experimental diabetic nephropathy. Am J Physiol Renal Physiol 302: F647–F657, 2012. [DOI] [PubMed] [Google Scholar]
  • 122.Sandborn WJ, Ghosh S, Panes J, Vranic I, Su C, Rousell S, Niezychowski W. Tofacitinib, an oral Janus kinase inhibitor, in active ulcerative colitis. N Engl J Med 367: 616–624, 2012. [DOI] [PubMed] [Google Scholar]
  • 123.Saran R, Li Y, Robinson B, Ayanian J, Balkrishnan R, Bragg-Gresham J, Chen JT, Cope E, Gipson D, He K, Herman W, Heung M, Hirth RA, Jacobsen SS, Kalantar-Zadeh K, Kovesdy CP, Leichtman AB, Lu Y, Molnar MZ, Morgenstern H, Nallamothu B, O'Hare AM, Pisoni R, Plattner B, Port FK, Rao P, Rhee CM, Schaubel DE, Selewski DT, Shahinian V, Sim JJ, Song P, Streja E, Kurella Tamura M, Tentori F, Eggers PW, Agodoa LY, Abbott KC. U.S. Renal Data System 2014 Annual Data Report. Epidemiology of kidney disease in the United States. Am J Kidney Dis 65: A7, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Sarbassov DD, Ali SM, Sengupta S, Sheen JH, Hsu PP, Bagley AF, Markhard AL, Sabatini DM. Prolonged rapamycin treatment inhibits mTORC2 assembly and Akt/PKB. Mol Cell 22: 159–168, 2006. [DOI] [PubMed] [Google Scholar]
  • 125.Sartipy P, Loskutoff DJ. Monocyte chemoattractant protein 1 in obesity and insulin resistance. Proc Natl Acad Sci USA 100: 7265–7270, 2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Sayyed SG, Ryu M, Kulkarni OP, Schmid H, Lichtnekert J, Gruner S, Green L, Mattei P, Hartmann G, Anders HJ. An orally active chemokine receptor CCR2 antagonist prevents glomerulosclerosis and renal failure in type 2 diabetes. Kidney Int 80: 68–78, 2011. [DOI] [PubMed] [Google Scholar]
  • 127.Schjoedt KJ, Rossing K, Juhl TR, Boomsma F, Rossing P, Tarnow L, Parving HH. Beneficial impact of spironolactone in diabetic nephropathy. Kidney Int 68: 2829–2836, 2005. [DOI] [PubMed] [Google Scholar]
  • 128.Schjoedt KJ, Rossing K, Juhl TR, Boomsma F, Tarnow L, Rossing P, Parving HH. Beneficial impact of spironolactone on nephrotic range albuminuria in diabetic nephropathy. Kidney Int 70: 536–542, 2006. [DOI] [PubMed] [Google Scholar]
  • 129.Schrijvers BF, Flyvbjerg A, De Vriese AS. The role of vascular endothelial growth factor (VEGF) in renal pathophysiology. Kidney Int 65: 2003–2017, 2004. [DOI] [PubMed] [Google Scholar]
  • 130.Sharma K, Ix JH, Mathew AV, Cho M, Pflueger A, Dunn SR, Francos B, Sharma S, Falkner B, McGowan TA, Donohue M, Ramachandrarao S, Xu R, Fervenza FC, Kopp JB. Pirfenidone for diabetic nephropathy. J Am Soc Nephrol 22: 1144–1151, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Sharma K, Jin Y, Guo J, Ziyadeh FN. Neutralization of TGF-β by anti-TGF-β antibody attenuates kidney hypertrophy and the enhanced extracellular matrix gene expression in STZ-induced diabetic mice. Diabetes 45: 522–530, 1996. [DOI] [PubMed] [Google Scholar]
  • 132.Sharma K, Ramachandrarao S, Qiu G, Usui HK, Zhu Y, Dunn SR, Ouedraogo R, Hough K, McCue P, Chan L, Falkner B, Goldstein BJ. Adiponectin regulates albuminuria and podocyte function in mice. J Clin Invest 118: 1645–1656, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Sharma K, Ziyadeh FN. Hyperglycemia and diabetic kidney disease. The case for transforming growth factor-β as a key mediator. Diabetes 44: 1139–1146, 1995. [DOI] [PubMed] [Google Scholar]
  • 134.Shibasaki F, Hallin U, Uchino H. Calcineurin as a multifunctional regulator. J Biochem 131: 1–15, 2002. [DOI] [PubMed] [Google Scholar]
  • 135.Shin N, Baribaud F, Wang K, Yang G, Wynn R, Covington MB, Feldman P, Gallagher KB, Leffet LM, Lo YY, Wang A, Xue CB, Newton RC, Scherle PA. Pharmacological characterization of INCB3344, a small molecule antagonist of human CCR2. Biochem Biophys Res Commun 387: 251–255, 2009. [DOI] [PubMed] [Google Scholar]
  • 136.Sison K, Eremina V, Baelde H, Min W, Hirashima M, Fantus IG, Quaggin SE. Glomerular structure and function require paracrine, not autocrine, VEGF-VEGFR-2 signaling. J Am Soc Nephrol 21: 1691–1701, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Siu B, Saha J, Smoyer WE, Sullivan KA, Brosius FC 3rd. Reduction in podocyte density as a pathologic feature in early diabetic nephropathy in rodents: prevention by lipoic acid treatment. BMC Nephrol 7: 6, 2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Sivaskandarajah GA, Jeansson M, Maezawa Y, Eremina V, Baelde HJ, Quaggin SE. Vegfa protects the glomerular microvasculature in diabetes. Diabetes 61: 2958–2966, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Stanton RC. Oxidative stress and diabetic kidney disease. Curr Diabetes Rep 11: 330–336, 2011. [DOI] [PubMed] [Google Scholar]
  • 140.Stevens RC, Cherezov V, Katritch V, Abagyan R, Kuhn P, Rosen H, Wuthrich K. The GPCR Network: a large-scale collaboration to determine human GPCR structure and function. Nat Rev Drug Discov 12: 25–34, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Sugimoto H, LeBleu VS, Bosukonda D, Keck P, Taduri G, Bechtel W, Okada H, Carlson W Jr, Bey P, Rusckowski M, Tampe B, Tampe D, Kanasaki K, Zeisberg M, Kalluri R. Activin-like kinase 3 is important for kidney regeneration and reversal of fibrosis. Nat Med 18: 396–404, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Susztak K, Raff AC, Schiffer M, Bottinger EP. Glucose-induced reactive oxygen species cause apoptosis of podocytes and podocyte depletion at the onset of diabetic nephropathy. Diabetes 55: 225–233, 2006. [PubMed] [Google Scholar]
  • 143.Takahashi K, Mizuarai S, Araki H, Mashiko S, Ishihara A, Kanatani A, Itadani H, Kotani H. Adiposity elevates plasma MCP-1 levels leading to the increased CD11b-positive monocytes in mice. J Biol Chem 278: 46,654–46,660, 2003. [DOI] [PubMed] [Google Scholar]
  • 144.Vallon V, Muhlbauer B, Osswald H. Adenosine and kidney function. Physiol Rev 86: 901–940, 2006. [DOI] [PubMed] [Google Scholar]
  • 145.Vallon V, Osswald H. Dipyridamole prevents diabetes-induced alterations of kidney function in rats. Naunyn-Schmiedeberg's Arch Pharmacol 349: 217–222, 1994. [DOI] [PubMed] [Google Scholar]
  • 146.Veron D, Bertuccio CA, Marlier A, Reidy K, Garcia AM, Jimenez J, Velazquez H, Kashgarian M, Moeckel GW, Tufro A. Podocyte vascular endothelial growth factor (Vegf(1)(6)(4)) overexpression causes severe nodular glomerulosclerosis in a mouse model of type 1 diabetes. Diabetologia 54: 1227–1241, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Veron D, Reidy KJ, Bertuccio C, Teichman J, Villegas G, Jimenez J, Shen W, Kopp JB, Thomas DB, Tufro A. Overexpression of VEGF-A in podocytes of adult mice causes glomerular disease. Kidney Int 77: 989–999, 2010. [DOI] [PubMed] [Google Scholar]
  • 148.Verstovsek S, Mesa RA, Gotlib J, Levy RS, Gupta V, DiPersio JF, Catalano JV, Deininger M, Miller C, Silver RT, Talpaz M, Winton EF, Harvey JH Jr, Arcasoy MO, Hexner E, Lyons RM, Paquette R, Raza A, Vaddi K, Erickson-Viitanen S, Koumenis IL, Sun W, Sandor V, Kantarjian HM. A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis. N Engl J Med 366: 799–807, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Wander SA, Hennessy BT, Slingerland JM. Next-generation mTOR inhibitors in clinical oncology: how pathway complexity informs therapeutic strategy. J Clin Invest 121: 1231–1241, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Wang L, Chang JH, Paik SY, Tang Y, Eisner W, Spurney RF. Calcineurin (CN) activation promotes apoptosis of glomerular podocytes both in vitro and in vivo. Mol Endocrinol 25: 1376–1386, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Wang L, Ellis MJ, Gomez JA, Eisner W, Fennell W, Howell DN, Ruiz P, Fields TA, Spurney RF. Mechanisms of the proteinuria induced by Rho GTPases. Kidney Int 81: 1075–1085, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Wang L, Flannery PJ, Rosenberg PB, Fields TA, Spurney RF. Gq-dependent signaling upregulates COX2 in glomerular podocytes. J Am Soc Nephrol 19: 2108–2118, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Wang S, Chen Q, Simon TC, Strebeck F, Chaudhary L, Morrissey J, Liapis H, Klahr S, Hruska KA. Bone morphogenic protein-7 (BMP-7), a novel therapy for diabetic nephropathy. Kidney Int 63: 2037–2049, 2003. [DOI] [PubMed] [Google Scholar]
  • 154.Wang S, de Caestecker M, Kopp J, Mitu G, Lapage J, Hirschberg R. Renal bone morphogenetic protein-7 protects against diabetic nephropathy. J Am Soc Nephrol 17: 2504–2512, 2006. [DOI] [PubMed] [Google Scholar]
  • 155.Wang W, Wang Y, Long J, Wang J, Haudek SB, Overbeek P, Chang BH, Schumacker PT, Danesh FR. Mitochondrial fission triggered by hyperglycemia is mediated by ROCK1 activation in podocytes and endothelial cells. Cell Metab 15: 186–200, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Wang X, Shaw S, Amiri F, Eaton DC, Marrero MB. Inhibition of the Jak/STAT signaling pathway prevents the high glucose-induced increase in TGF-β and fibronectin synthesis in mesangial cells. Diabetes 51: 3505–3509, 2002. [DOI] [PubMed] [Google Scholar]
  • 157.Wang XJ, Sun Z, Villeneuve NF, Zhang S, Zhao F, Li Y, Chen W, Yi X, Zheng W, Wondrak GT, Wong PK, Zhang DD. Nrf2 enhances resistance of cancer cells to chemotherapeutic drugs, the dark side of Nrf2. Carcinogenesis 29: 1235–1243, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Wang Y, Deb DK, Zhang Z, Sun T, Liu W, Yoon D, Kong J, Chen Y, Chang A, Li YC. Vitamin D receptor signaling in podocytes protects against diabetic nephropathy. J Am Soc Nephrol 23: 1977–1986, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Wang Y, Jarad G, Tripathi P, Pan M, Cunningham J, Martin DR, Liapis H, Miner JH, Chen F. Activation of NFAT signaling in podocytes causes glomerulosclerosis. J Am Soc Nephrol 21: 1657–1666, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Wang Y, Zhou J, Minto AW, Hack BK, Alexander JJ, Haas M, Li YC, Heilig CW, Quigg RJ. Altered vitamin D metabolism in type II diabetic mouse glomeruli may provide protection from diabetic nephropathy. Kidney Int 70: 882–891, 2006. [DOI] [PubMed] [Google Scholar]
  • 161.Weisberg SP, Hunter D, Huber R, Lemieux J, Slaymaker S, Vaddi K, Charo I, Leibel RL, Ferrante AW Jr. CCR2 modulates inflammatory and metabolic effects of high-fat feeding. J Clin Invest 116: 115–124, 2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Wenzel RR, Littke T, Kuranoff S, Jurgens C, Bruck H, Ritz E, Philipp T, Mitchell A. Avosentan reduces albumin excretion in diabetics with macroalbuminuria. J Am Soc Nephrol 20: 655–664, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.White GE, Iqbal AJ, Greaves DR. CC chemokine receptors and chronic inflammation—therapeutic opportunities and pharmacological challenges. Pharmacol Rev 65: 47–89, 2013. [DOI] [PubMed] [Google Scholar]
  • 164.Winiarska K, Malinska D, Szymanski K, Dudziak M, Bryla J. Lipoic acid ameliorates oxidative stress and renal injury in alloxan diabetic rabbits. Biochimie 90: 450–459, 2008. [DOI] [PubMed] [Google Scholar]
  • 165.Wu H, Ghosh S, Perrard XD, Feng L, Garcia GE, Perrard JL, Sweeney JF, Peterson LE, Chan L, Smith CW, Ballantyne CM. T-cell accumulation and regulated on activation, normal T cell expressed and secreted upregulation in adipose tissue in obesity. Circulation 115: 1029–1038, 2007. [DOI] [PubMed] [Google Scholar]
  • 166.Yan Y, Wei CL, Zhang WR, Cheng HP, Liu J. Cross-talk between calcium and reactive oxygen species signaling. Acta Pharmacol Sin 27: 821–826, 2006. [DOI] [PubMed] [Google Scholar]
  • 167.Yanagita M. Inhibitors/antagonists of TGF-β system in kidney fibrosis. Nephrol Dial Transpl 27: 3686–3691, 2012. [DOI] [PubMed] [Google Scholar]
  • 168.Yu H, van Berkel TJ, Biessen EA. Therapeutic potential of VIVIT, a selective peptide inhibitor of nuclear factor of activated T cells, in cardiovascular disorders. Cardiovasc Drug Rev 25: 175–187, 2007. [DOI] [PubMed] [Google Scholar]
  • 169.Yuan W, Pan W, Kong J, Zheng W, Szeto FL, Wong KE, Cohen R, Klopot A, Zhang Z, Li YC. 1,25-dihydroxyvitamin D3 suppresses renin gene transcription by blocking the activity of the cyclic AMP response element in the renin gene promoter. J Biol Chem 282: 29,821–29,830, 2007. [DOI] [PubMed] [Google Scholar]
  • 170.Yusuf S, Teo KK, Pogue J, Dyal L, Copland I, Schumacher H, Dagenais G, Sleight P, Anderson C. Telmisartan, ramipril, or both in patients at high risk for vascular events. N Engl J Med 358: 1547–1559, 2008. [DOI] [PubMed] [Google Scholar]
  • 171.Zhang DD. The Nrf2-Keap1-ARE signaling pathway: The regulation and dual function of Nrf2 in cancer. Antiox Redox Signal 13: 1623–1626, 2010. [DOI] [PubMed] [Google Scholar]
  • 172.Zhang L, Li R, Shi W, Liang X, Liu S, Ye Z, Yu C, Chen Y, Zhang B, Wang W, Lai Y, Ma J, Li Z, Tan X. NFAT2 inhibitor ameliorates diabetic nephropathy and podocyte injury in db/db mice. Br J Pharmacol 170: 426–439, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Zhang Y, Deb DK, Kong J, Ning G, Wang Y, Li G, Chen Y, Zhang Z, Strugnell S, Sabbagh Y, Arbeeny C, Li YC. Long-term therapeutic effect of vitamin D analog doxercalciferol on diabetic nephropathy: strong synergism with AT1 receptor antagonist. Am J Physiol Renal Physiol 297: F791–F801, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Zhang Z, Sun L, Wang Y, Ning G, Minto AW, Kong J, Quigg RJ, Li YC. Renoprotective role of the vitamin D receptor in diabetic nephropathy. Kidney Int 73: 163–171, 2008. [DOI] [PubMed] [Google Scholar]
  • 175.Zhang Z, Zhang Y, Ning G, Deb DK, Kong J, Li YC. Combination therapy with AT1 blocker and vitamin D analog markedly ameliorates diabetic nephropathy: blockade of compensatory renin increase. Proc Nat Acad Sci USA 105: 15,896–15,901, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Zhou L, Li Y, Hao S, Zhou D, Tan RJ, Nie J, Hou FF, Kahn M, Liu Y. Multiple genes of the renin-angiotensin system are novel targets of Wnt/β-catenin signaling. J Am Soc Nephrol 26: 107–120, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Zhu L, Jiang R, Aoudjit L, Jones N, Takano T. Activation of RhoA in podocytes induces focal segmental glomerulosclerosis. J Am Soc Nephrol 22: 1621–1630, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Zhu X, Wu S, Dahut WL, Parikh CR. Risks of proteinuria and hypertension with bevacizumab, an antibody against vascular endothelial growth factor: systematic review and meta-analysis. Am J Kidney Dis 49: 186–193, 2007. [DOI] [PubMed] [Google Scholar]
  • 179.Ziyadeh FN, Hoffman BB, Han DC, Iglesias-De La Cruz MC, Hong SW, Isono M, Chen S, McGowan TA, Sharma K. Long-term prevention of renal insufficiency, excess matrix gene expression, and glomerular mesangial matrix expansion by treatment with monoclonal antitransforming growth factor-beta antibody in db/db diabetic mice. Proc Nat Acad Sci USA 97: 8015–8020, 2000. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Physiology - Renal Physiology are provided here courtesy of American Physiological Society

RESOURCES