Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Apr 30.
Published in final edited form as: Circ Res. 2010 Apr 30;106(8):1319–1331. doi: 10.1161/CIRCRESAHA.110.217117

Activation of Protein Kinase C Isoforms & Its Impact on Diabetic Complications

Pedro Geraldes 1, George L King 1,2
PMCID: PMC2877591  NIHMSID: NIHMS198866  PMID: 20431074

Abstract

Both cardio- and microvascular complications adversely affect the life quality of patients with diabetes and have been the leading cause of mortality and morbidity in this population. Cardiovascular pathologies of diabetes have an effect on microvenules, arteries, and myocardium. It is believed that hyperglycemia is one of the most important metabolic factors in the development of both micro- and macrovascular complications in diabetic patients. Several prominent hypotheses exist to explain the adverse effect of hyperglycemia. One of them is the chronic activation by hyperglycemia of protein kinase C (PKC), a family of enzymes that are involved in controlling the function of other proteins. PKC has been associated with vascular alterations such as increases in permeability, contractility, extracellular matrix synthesis, cell growth and apoptosis, angiogenesis, leukocyte adhesion, and cytokine activation and inhibition. These perturbations in vascular cell homeostasis caused by different PKC isoforms (PKC-α, -β1/2, and PKC-δ) are linked to the development of pathologies affecting large vessel (atherosclerosis, cardiomyopathy) and small vessel (retinopathy, nephropathy and neuropathy) complications. Clinical trials using a PKC-β isoform inhibitor have been conducted, with some positive results for diabetic nonproliferative retinopathy, nephropathy and endothelial dysfunction. This paper reviews current understanding of how PKC isoforms cause vascular dysfunctions and pathologies in diabetes.

Keywords: Protein Kinase C, Vascular cell biology, Diabetes, Cardiovascular diseases

Introduction

Diabetes currently affects nearly 170 million people worldwide and this number will double by 2030, affecting both developed and developing countries.1 The major cause of suffering in diabetic patients is complications, which are the result of interactions among systemic metabolic abnormalities such as hyperglycemia, dyslipidemia, genetic and epi-genetic modulators and local tissue responses to toxic metabolites. Complications involve large-vessel obstructions such as coronary artery diseases, atherosclerosis and peripheral vascular diseases, and microvascular pathologies including retinopathy, nephropathy and neuropathy. Hyperglycemia is one of the major systemic risk factors for diabetic complications. The Diabetes Control and Complications Trial (DCCT) in type 1 diabetes2 and the United Kingdom Prospective Diabetes Study in type 2 diabetes3 demonstrated that intensive blood glucose control delays the onset and retards the progression of diabetic microvascular complications. In contrast, reducing cardiovascular pathologies in type 2 diabetes requires the control of multiple metabolic factors, including hyperglycemia, free fatty acids, lipids, insulin resistance and others. Hyperglycemia alone may not be sufficient to trigger diabetic alterations since only 30–40% of type 1 diabetic patients will ultimately develop chronic renal failure, suggesting that genetic and protective tissue factors may also play an important role4. For example, the 50-Year Medalist Study at Joslin Diabetes Center, which recognizes patients who survive more than 50 years with type 1 diabetes, showed that significant numbers of diabetic patients live without severe complications regardless of their HBA1c levels and other classical markers thought to be predictive for diabetic vascular complications.4 These data suggest that some patients possess genetic factors that can neutralize the adverse effects of hyperglycemia.

Multiple biochemical pathways have been proposed to link the adverse effects of hyperglycemia with vascular complications. Cellular mechanisms include increased activation of the polyol pathway,5 nonenzymatic glycation and advanced glycation end products pathway,6 enhanced reactive oxygen production and actions,7 and activation of the diacylglycerol (DAG)-protein kinase C (PKC) pathway.8 It is likely that hyperglycemia-induced intra- and extracellular changes lead to alterations of signal transduction pathways, affecting gene expression and protein function to cause cellular dysfunction and damage. The DAG-PKC pathway is one of the most studied pathways in cellular signaling induced by diabetes.9 In this review, we will focus on the role of PKC actions and related signaling changes in the development of diabetic vascular complications.

Activation of DAG-PKC pathway in diabetes

DAG levels are elevated chronically in the hyperglycemic or diabetic environment due to an increase in the glycolytic intermediate dihydroxyacetone phosphate. This intermediate is reduced to glycerol-3-phosphate, which subsequently increases de novo synthesis of DAG.10 In diabetes, total DAG levels are elevated in vascular tissues such as the retina,11 aorta, heart12 and renal glomeruli,13,14 and in nonvascular tissues such as liver and skeletal muscles.15 However, there is no consistent change in DAG levels in the central nervous system and peripheral nerves.16 Various cell culture studies have shown that DAG levels increase in a time-dependent manner as glucose levels elevate from 5.5 to 22 mM in aortic endothelial cells,12 retinal pericytes,17 smooth muscle cells10 and renal mesangial cells.18

PKC, a group of enzyme members of the AGC (cAMP-dependent protein kinase/protein kinase G/protein kinase C) family, is a serine/threonine-related protein kinase that plays a key role in many cellular functions and affects many signal transduction pathways.19 There are multiple isoforms of PKC that function in a wide variety of biological systems.20 The conventional PKC (cPKC) isoforms (PKC-α, -β1, -β2, and -γ) are activated by phosphatidylserine (PS), calcium, and DAG or phorbol esters such as phorbol 12-myristate 13-acetate (PMA), whereas novel PKCs (nPKC; PKC-δ, -ε, -θ and -η) are activated by PS, DAG or PMA, but not by calcium. The atypical PKCs (aPKC; PKC-ζ and -ι/λ) are not activated by calcium, DAG or PMA (Figure 1). Extensive and excellent reviews concerning PKC structural basic activation have been published.1922 Given the current breadth of knowledge in this area, we will focus our attention on how hyperglycemia modulates PKC activation. PKCs can also be activated by oxidants such as H2O2 in a manner unrelated to lipid second messengers23 and by mitochondrial superoxide induced by elevated glucose levels.24 Many abnormal vascular and cellular processes and deregulations, including endothelial dysfunction, vascular permeability, angiogenesis, cell growth and apoptosis, changes in vessel dilation, basement membrane thickening and extracellular matrix expansion, enzymatic activity alterations such as mitogen-activated protein kinase (MAPK), cytosolic phospholipase A2 (PLA2), Na+–K+–ATPase and alterations in several transcription factors (Figure 2), are attributed to multiple PKC isoforms that are changed by diabetes (Table 1). These PKC-induced vascular and tissue pathologies will be discussed in detail in the following paragraphs.

Figure 1.

Figure 1

Schematic representation of the domain structure of PKC isoforms (adapted from Newton19,20 and Steinberg19,20).

Figure 2.

Figure 2

Schematic representation of biological targets of PKC isoform activation and synthesis.

Table 1.

PKC isoforms detected in vascular tissues and cell types by immunoblot under normal conditions or following exposure to hyperglycemia or diabetic state.

Tissue and cultured cell type PKC isoforms detected PKC isoforms activated (translocation and/or abundance) by hyperglycemia or diabetes
Mouse retina α, β1, β2, δ, ε, ζ β2, δ
Rat retina α, β1, β2, δ, ε α, β1, β2, δ, ε
Bovine pericytes α, β1, β2, δ, ε, θ, ζ δ
Bovine endothelial cells α, β1, β2, δ, ε, γ, ζ α, β2, δ
Rat kidney α, β1, β2, δ, ε, ζ α, ε
Rat glomeruli α, β1, β2, δ, ε α, β1, β2, δ, ε
Rat mesangial cell α, δ, ε, ζ α, ζ
Rat corpus cavernosum α, β1, β2, δ, ε β2
Rat sciatic nerve α, β1, β2, δ, ε α or no change
Rat aorta α, β2, δ, ε β2
Rat aortic smooth muscle cells α, β2, δ β2, δ
Rat heart α, β2, δ, ε, γ, ζ α, β2, δ, ζ
Rat cardiac myocytes β2, δ, ε β2, ε

PKC Activation: Cell Culture Studies

Endothelial cells (EC)

EC regulate both vasodilator and vasoconstrictor substances mediating coagulation, platelet adhesion and immune function and control volume and electrolyte content of the intravascular and extravascular spaces. Tight junctions between EC form a vascular barrier, which in the diabetic state becomes more vulnerable and permeable as a result of EC abnormalities. PKC activation directly increases the permeability of albumin and other macromolecules through barriers formed by EC.25, 26 Moreover, Inoguchi and collaborators reported that hyperglycemia or PMA inhibits gap junction intercellular communication in bovine aortic EC. Staurosporine, a serine/threonine kinase inhibitor, prevented these effects of high glucose.27 High glucose exposure induced translocation of PKC-α, -β1, -β2, and PKC-δ but not PKC-ε or PKC-ζ in retinal EC.28 A previous study showed that overexpression of PKC-β1 in human dermal microvascular EC enhances phorbol ester-induced increased permeability to albumin.29 Hempel and colleagues also showed that PMA increases permeability via translocation of PKC-α similar to that caused by high glucose levels. This permeability is prevented by staurosporine.30 PKC activation also results in endothelium-dependent vasodilator dysfunction by altering the bioavailability of nitric oxide (NO), affecting vascular endothelial growth factor (VEGF) expression and actions and decreasing production of prostacyclin, as well as increasing the production of thromboxane, other cyclogenase-dependent vasoconstrictors and endothelin-1 (ET-1)3133 (Figure 3). A previous study demonstrated that endothelial nitric oxide synthase (eNOS) expression is decreased in aortic EC cultured in high glucose concentrations, resulting in a reduction of NO.34 Treatment with calphostin C, an inhibitor of c- and nPKC, prevented high glucose levels from reducing NO production.33 Another potential pathway by which high concentrations of glucose reduce NO bioavailability is by increasing superoxide production from nicotinamide adenine dinucleotide phosphate (NADPH) oxidase in aortic EC.35 The induction of several subunits of NADPH oxidase by constant or intermittent high glucose concentrations is decreased with treatment of a PKC-β inhibitor (LY379196).36 PKC activation also modulates vascular endothelial permeability and neovascularization via the expression of growth factors, such as VEGF/vascular permeability factor (VPF). One study showed that both VEGF/VPF mitogenic and permeability actions are, in part, the result of the membrane expression of PKC-α and -β through tyrosine phosphorylation of phospholipase-Cγ, which is reduced by a PKC-β inhibitor, LY333531.37 Moreover, PKC-β is required for VEGF-induced retinal EC permeability by altering occluding phosphorylation.38 One possible mechanism for vasoconstriction in retinal EC caused by PKC action is probably increased expression of ET-1, a potent vasoconstrictor. Indeed, studies reported that ET-1 mRNA expression elevates in cultured retinal EC exposed to high glucose levels.28

Figure 3.

Figure 3

Schematic representation of hyperglycemia-induced PKC activation affecting multiple cellular functions.

Smooth muscle cells (SMC)

Vascular SMC are involved in regulation of vascular tone and could be an important target for injurious effects of hyperglycemia. An emerging body of evidence suggests that hyperglycemia-induced SMC pathogenesis, similar to EC pathogenesis, results from an imbalance between cell growth and cell death. In the presence of high glucose levels, SMC have elevated DNA synthesis and contraction by PMA stimulation.3941 Another group reported that high glucose levels reduce SMC apoptosis which is blocked by PKC inhibitor (calphostin C).42 Moreover, rat SMC exposed to high glucose concentrations exhibited increases membrane fraction expression of cPKC-β and nPKC-δ p38 MAPK phosphorylation and arachidonic acid release (Figure 3).41

Monocytes – macrophages

Monocyte activation and transformation into macrophages are key steps in the atherosclerotic and inflammatory process. One of the earliest events in the pathogenesis of atherosclerosis is lipid accumulation in the arterial wall and formation of foam cells through uptake of modified or oxidized low density lipoprotein (oxLDL) by monocyte-derived macrophages.43 Little is known about how hyperglycemia or diabetes affects PKC activity in circulating monocytes. However, activation of PKC in monocytes may participate in their initial adhesion to the vasculature and differentiation into macrophages. Hyperglycemia regulates the leptin-like oxidized LDL receptor-1 (LOX-1) and enhanced membrane PKC-β2 expression in human monocyte-derived macrophages.44 These effects of high glucose levels in LOX-1 expression are prevented by treatment with calphostin C or PKC-β inhibitor (LY379196). Ceolotto and collaborators reported that monocytes isolated from diabetic patients showed higher membrane PKC activity compared to those from control subjects.45 This study also demonstrated that membrane PKC-β activity, but not -α, isoform, is increased in diabetic monocytes.45 Further, Osto and collaborators reported that inhibition of PKC-β reduced LDL uptake by macrophages.46 This study also showed that treatment with LY333531 significantly attenuates macrophage activation by reducing intercellular adhesion molecules-1 (ICAM-1) and monocyte chemotactic protein-1 (MCP-1) protein expression.47 A recent study from Dasu and colleagues reported that elevation of glucose levels in the media increases the expression of toll-like receptors 2/4 in the human acute monocytic leukemia cell line, while inhibition of PKC-α and PKC-δ prevents this effect.48 Overall, these data suggest that PKC signaling could be in part responsible for macrophage activation/attachment and foam cell formation induced in the hyperglycemic state (Figure 3).

Mesangial cells

The hallmark of diabetic kidney disease is the thickening of glomerular basement membrane and accumulation of extracellular matrix (ECM) in glomerular mesangium and tubulointerstitium.49 Histological analysis showed increases in type IV and VI collagen, fibronectin and laminin and decreases in proteoglycans in the mesangium of diabetic patients with nephropathy, and probably in the vascular endothelium in general.50 In vitro experiments reported that hyperglycemia or PMA increases type IV collagen and fibronectin expression in mesangial cells, which is prevented by calphostin C.51, 52 Kapor-Drezgic and collaborators reported that total PKC activity measured by in situ 32P-phosphorylation of epidermal growth factor substrate is increased by hyperglycemia in rat mesangial cells.53 Length of exposure to high glucose also influences PKC isoform translocation in these cells.54 Previous studies reported that exposure to high glucose levels or PMA stimulation increases PKC-α, -β1, -β2, PKC-δ and -ε membrane phosphorylated fraction in mesangial cells.5357 Advanced glycation end products (AGEs) also play a significant role in the pathogenesis of vascular and renal complications associated with diabetes. Two groups indicated that AGEs modulate, directly or indirectly via oxidative stress, particulate and membrane localization of PKC-β in mesangial cells.58, 59 In addition, the development of mesangial expansion and basement membrane thickening in diabetes correlates with increased expression of transforming growth factor-beta (TGF-β). The implication of PKC causing elevated production of ECM and TGF-β is further supported by several reports showing that LY333531 prevents hyperglycemia-increased ECM production and TGF-β expression in mesangial cells.60 Another group demonstrated that high glucose concentrations stimulate PKC-ζ activity, as measured by immune complex kinase assay and immunofluorescence confocal microscopy. This activity is suppressed by a TGF-β receptor inhibitor (LY364947).61

PKC Activation: Animal Model Studies

Atherosclerosis

Diabetes macrovasculopathy is associated with structural and functional changes in large vessels that lead to blood flow obstruction, hypertension, myocardial infarction (MI) and possibly death. Atherosclerosis, characterized by endothelial dysfunction, cytokine expression, monocyte infiltration, SMC proliferation, impaired fibrinolysis combined with increased thrombosis, and chronic inflammation causes ischemic heart diseases, MI, stroke, and peripheral arterial diseases.6264 Hyperglycemia is believed to participate in atherosclerotic plaque formation. Intensive therapy treatment (DCCT study) showed a significant decrease in carotid artery intima/media thickness.65 However, the disease state associated with insulin resistance can also accelerate atherosclerosis which suggests that metabolic abnormalities associated with diabetes, and not hyperglycemia per se, can increase the risk for macrovascular complications. Nevertheless, activation of PKC isoforms, especially the β isoform, has been the subject of recent investigation. Schmidt and Yan’s group recently published supportive data on the potential role of PKC-β in atherosclerotic plaque formation. Depletion of PKC-β gene or treatment with LY333531 in apolipoprotein e–deficient mice decreased atherosclerosis by inhibiting the early growth response (Egr)-1 protein, which regulates vascular cell adhesion molecule (VCAM) expression and matrix metalloproteinase-2 (MMP-2) activity preferentially in EC.66 However, additional isoforms may be involved in other steps of atherosclerosis. Novel PKC-δ participates in SMC apoptosis and deletion of this isoform, leading to formation of arteriosclerosis.67 Therefore, whether it will be beneficial to inhibit multiple PKC isoforms to halt or prevent complex mechanisms related to atherosclerosis remains to be resolved.

Cardiomyopathy

Myocardial pathologies in diabetic patients include diastolic dysfunction, microvascular diseases, and interstitial fibrosis, the last of which could be the result of active tissue remodeling contributing to reduction of cardiac contractility.68 Independent of the severity of coronary artery disease, hypertension and other risk factors, diabetic patients have elevated risk of congestive heart failure, inadequate collateral vascular formation in response to ischemia69 and greater likelihood of developing heart failure and re-infarction after MI.70 Quantitative immunoblotting revealed a significant increase in membrane fraction expression of PKC-β1 and -β2 in human failed hearts compared to nonfailed.71 This study also showed that treatment with LY333531 decreases PKC activation in human heart.71 Previously, animal studies reported that PKC-α, -β1 and -β2 isoforms, mRNA and protein expression in the membrane fraction are increased in the heart and aorta in the hyperglycemic condition.12, 72 Genetically modified mice have been developed to better examine the role of PKC in heart diseases. Transgenic mice overexpressing PKC-β2 develop cardiomyopathy, supporting the role of PKC-β2 in myocardial hypertrophy and fibrosis.73 Both angiotensin-converting enzyme (ACE) and PKC-β inhibitors ameliorate the metabolic gene profile and affect PKC activity in diabetic hearts without altering circulating metabolites.74 In cardiomyocytes, free fatty acids increased metabolic genes such as PDK4 and UCP3 and mediated the inhibition of basal and insulin-stimulated glucose oxidation, all of which are prevented by treatment with LY333531. Further, treatment with islet cell transplantation, LY333531 and ACE inhibitor (captopril) improved diastolic function, increased glucose utilization by 36%74 and attenuated myocyte hypertrophy and collagen deposition75 in diabetic rat hearts. Together, these results suggest that both ACE and PKC-β inhibitors regulate fuel metabolic gene expression directly in the myocardium and consequently improve cardiac function and metabolism in diabetes. Other PKC isoforms may also play a role in cardiomyopathy. A recent study reported that PKC-α–deficient mice exhibit increased cardiac contractility and are less susceptible to heart failure following long-term pressure-overload stimulation.76

Connective tissue growth factor (CTGF) expression may contribute to cardiac fibrosis. Interestingly, transgenic PKC-β2 mice showed increased CTGF expression in the myocardium, suggesting that CTGF may act directly or indirectly with other cytokines to induce cardiac fibrosis.73 By opposition, inhibition of PKC-δ exacerbated CTGF expression and potentially cardiac fibrosis in the presence of angiotensin II.77 However, careful attention should be given to PKC-δ activation since this isoform regulates pro-apoptotic signalling in cardiomyocytes during myocardial ischemia/reperfusion injury.78 Further, cardiac-specific activation of PKC-ε prevented diabetes-induced pathogenetic changes in the heart, including ventricular function79 (Figure 4).

Figure 4.

Figure 4

Schematic representation of various biological targets of PKC activation leading to selective insulin resistance and diabetic cardiomyopathy.

Among the processes induced by hyperglycemia, activation of PKC may contribute to cardiomyopathy by inhibiting insulin’s metabolic actions, perhaps by phosphorylation of serine/threonine residues on the insulin receptors or their substrates.80 Cardiomyocytes isolated from nondiabetic animals and cultured in high glucose concentrations exhibited impaired insulin-stimulated glucose uptake compared to myocytes in normal glucose levels.81 Loss of insulin action in myocardium is associated with lower basal expression of hypoxia-inducible factor-1 alpha, which affects VEGF expression in the myocardium.82 In contrast to nondiabetic patients, insulin-resistant and diabetic patients showed a downregulation of VEGF and its receptors.83 Moreover, several studies of diabetic animals confirmed this finding, showing a reduction of mRNA and protein expression of VEGF and its receptors, cardiac VEGF signaling and coronary capillary density in the myocardium.84, 85 Interestingly, another group reported that diabetic patients with chronic coronary heart diseases show an elevation of myocardial VEGF expression but a reduction of its receptor expression and subsequently its downstream signal transduction.86 Nevertheless, VEGF signaling impairment results in decreased formation of collateral circulation, which explains bigger infarct size and congestive heart failure post MI in patients with diabetes.

Retinal tissues

The pathogenesis of diabetic retinopathy is a complex process involving multiple factors.87 The early stage of diabetic retinopathy is characterized by loss of pericytes around capillaries in the retina. Indeed, pericyte loss occurs prior to clinically discernible retinopathy. This is followed by development of weakness in the capillary wall, leading to capillary aneurysm formation (microaneurysms) and fluid leakage from capillaries as their walls become more permeable, with increased adhesion of leucocytes and monocytes to the endothelium.87 Hyperglycemia activates several PKC isoforms in retinal tissues, including PKC-α, -β-δ, and -ε.12, 88 PKC activation causes retinal vascular dysfunction by altering enzyme activities in EC (NO, ET-1, VEGF) and pericytes (platelet-derived growth factor; PDGF, reactive oxygen species; ROS, nuclear factor-kappa B; NF-κB and MAPK).89 In diabetic animals, blood flow is decreased in the retina but elevated in glomeruli.9092 In streptozotocin-induced diabetic rats, treatment with LY333531 (0.1 to 10 mg/kg) improved retinal hemodynamic abnormalities.13 Interestingly, systemic oral administration for two weeks of LY333531 to diabetic rats from the onset of the disease normalized the RBF13 and prevented the induction of ET-1 mRNA expression.93 Furthermore, local intravitreous injection of LY333531 (5 nM) reduced retinal PKC activation and retinal circulation time and restored retinal blood flow (RBF) in diabetic rats.94 Alterations in NO production and eNOS expression directly influence vascular hemodynamics such as contraction and relaxation, which may affect RBF. In vessels isolated from diabetic patients and animals, acetylcholine stimulation, which induces vessel relaxation, appears to be delayed.95, 96 PKC agonist PMA provokes vascular relaxation impairment in normal arteries.97

How diabetes regulates and increases pro-apoptotic factors has been researched for many years. However, the impact of hyperglycemia on reducing survival factors has been much less studied. We recently reported that both PKC-β and PKC-δ translocate to the membrane fraction in total retinal lysates of diabetic mice, but the consequences of these two PKC isoform activations were very different: PKC-δ induced cellular apoptosis,17 whereas PKC-β enhanced cellular growth.98 Accordingly, an increase in membrane PKC-δ levels for several months of diabetes correlated with the appearance of retinal pericyte apoptosis in vitro and acellular capillaries in vivo. In vivo studies showed that induction of retinal PKC-δ of diabetic mice leads to PDGF resistance, which is not observed in Prkcd−/− mice. Further, specific inhibition of PKC-δ in pericytes in vitro or disruption of Prkcd−/− mice prevented NF-κB activation and restored signal transduction of PDGF. Our study found that hyperglycemia through PKC-δ actions promotes two distinct and equally important pathways by 1) increasing ROS production and NF-κB activity and 2) decreasing the important survival signaling pathway of PDGF by upregulating the expression of a protein tyrosine phosphatase, SHP-1. These findings identify a pivotal role for PKC-δ in causing retinal cell apoptosis and the formation of acellular capillaries (Figure 5).17

Figure 5.

Figure 5

Schematic representation of potential biological targets of PKC activation causing diabetic retinopathy.

Renal tissues

Many studies have correlated changes in hyperglycemia with severity and progression of nephropathy affecting both the glomeruli and renal proximal tubules.2, 3, 99, 100 The specific effects of PKC isoforms on diabetic complications in the kidneys are just beginning to be determined using pharmacological approaches and genetically altered mice. Menne and colleagues reported that diabetic Prkca−/− mice are resistant to the development of albuminuria and glomerular hyperfiltration stimulated by VEGF.101 The translocation of PKC-α to renal cortical membranes by diabetes is associated with NADPH-dependent superoxide production and elevated renal serum and urinary VEGF.102 Furthermore, previous animal studies reported that deletion of the PKC-β gene or treatment with LY333531 protects against diabetes by causing increases in renal hypertrophy, glomerular hyperfiltration, ECM production, expression of CTGF and production of TGF-β1 and ROS (Figure 6).13, 60, 103105 Even in nondiabetic kidney disease, LY333531 treatment attenuated the impairment in glomerular filtration rate and reduced the extent of both glomerulosclerosis and tubulointerstitial fibrosis in subtotally nephrectomized rats.106 Interestingly, analysis of the renal phenotype in Prkce−/− mice showed elevated albuminuria at 6 and 16 weeks of age, with increased tubulointerstitial fibrosis and mesangial cell expansion.107 These data suggest that not all PKC isoforms have adverse effects on kidney pathologies.

Figure 6.

Figure 6

Schematic representation of various biological targets of PKC activation leading to diabetic nephropathy.

Neuronal tissues

The contribution of PKC activation to diabetic neuropathy still requires clarification. PKC contributes to diabetic neuropathy by a neurovascular mechanism such as blood flow and conduction velocity. Immunochemical analysis demonstrated the presence of PKC-α, -β1, -β2, -γ, PKC-δ and -ε isoforms in nerve.108, 109 A previous report demonstrated a reduction of PKC activity by direct measurement of sciatic nerve tissues in streptozotocin diabetic rats.110 These results contrast with more recent studies showing that treatment with nonselective PKC isoform inhibitor as well as selective PKC-β inhibitor improve neural function in diabetic animals.111, 112 Initial evidence suggested that PKC is involved in the mechanism leading to reduced Na+–K+–ATPase activity, resulting in decreased nerve conduction and nerve regeneration.113 Membrane-associated PKC activity was reduced in diabetic mice treated with nonselective PKC inhibitors that restored or maintained Na+–K+–ATPase activity.111, 114 Further, previous studies examined the effect of diabetes in rat sciatic nerve and demonstrated that LY333531 treatment prevents the development of diabetic nerve dysfunction.115 Indeed, Cameron and colleagues showed that treatment with LY333531 at low dose improves motor nerve conduction velocity, normalizes nerve blood flow and restores Na+–K+–ATPase activity in diabetic rats.116 However, in this study, diabetes did not affect nerve PKC activity or DAG levels. Overall, these studies suggest that PKC inhibition on nerve conduction improves nerve blood flow rather than nerve Na+–K+–ATPase activity.

Insulin resistance

Although the principal function of insulin is to regulate and maintain glycemic control, this hormone has many vasotropic actions. Insulin normally stimulates vasorelaxation through a direct effect on blood vessels mediated by endothelium-derived NO.117 Insulin also promotes endothelial NO production by rapid post-translational mechanisms118 as well as eNOS gene expression.119 Therefore, insulin resistance, diabetes or reduction of insulin-stimulated regulation of endothelium-derived NO may be an important factor for vascular homeostasis.120 Insulin signaling in vasculature of obese Zucker rats showed inhibition of the phosphoinositide 3-kinases (PI3K) pathway but no change in the extracellular signal-regulated kinases (ERK)1/2 cascade, suggesting a selective insulin resistance phenomenon.121 This observation was subsequently reported in skeletal muscle from obese people and patients with type 2 diabetes122 and in the myocardium of obese Zucker rats.123 Vasculature of obese Zucker rats also exhibited elevated PKC activity124 and reduced insulin-stimulated insulin receptor substrate tyrosine phosphorylation.121 The question of which PKC isoform’s elevation is involved in this process has generated much interest. Treatment with LY333531 normalized the reduction of insulin-stimulated NO production in the aorta of obese Zucker rats.124 Transgenic mice overexpressing PKC-β2 exhibited decrease Akt activation in vascular cells after insulin stimulation.124 Although strong evidence corroborates PKC-induced insulin signaling inhibition, the complex mechanisms involved are not fully understood. PKC activation prevents PI3K pathway at IRS level, which affects several signaling molecules related to this pathway (see elegant review by Sampson A.R. et al.).125 Insulin actions mediating the ERK1/2 pathway are accentuated by PKC actions.126 Thus, selective insulin resistance, caused by PKC activation, enhances insulin’s pro-atherosclerotic mechanisms via ERK1/2 signaling or inhibits its anti-atherosclerotic mechanisms by inhibiting the PI3K/Akt pathway.127

PKC Inhibitors and Human Clinical Trials

General PKC isoform inhibitors exist. However, these non–isoform-specific inhibitors interact with other ATP binding kinases and therefore display toxic and severe side effects in vivo. Suitable specific PKC isoform inhibitors for therapeutic or clinical studies should target the phospholipid or phorbol ester binding site of the PKC structure, called regulatory domain, or bind to the substrate or ATP binding site of the catalytic domain. For example, indolcarbazole or bisindoylmaleimide are selective inhibitors that target the ATP-binding site of catalytic domain. The PKC-β inhibitor ruboxistaurin (LY333531, RBX or Arxxant™; Eli Lilly and Company, Indianapolis, IN), is a class of bisindoylmaleimide.13, 104 Rotterlin (mallotoxin), a natural product derived from Mallotus philippensis, has higher affinity for PKC-δ (IC50 3–6 μM) but also inhibits other isoforms of PKC (IC50 >30μM)128 and other non-PKC kinases such as MAPK, protein kinase A and GSK-3.129 In contrast, RBX shows selective inhibition for PKC-β1 and PKC-β2 with IC50 values at 4.5 and 5.9 nM, respectively.130

RBX is the most studied PKC inhibitor in cellular, animal and especially human studies (Table 2). Phase I studies using RBX included patients who had diabetes for less than 10 years and no evidence of clinical retinopathy. This study determined the dose needed to normalize RBF, an early marker of diabetic retinopathy.131 The dose-response curve showed that a minimum dosage of 32 mg/day orally is required to prevent decreases in RBF in diabetic patients.131 Low doses did not completely normalize RBF. Few side effects were found during clinical trials of up to four years’ duration.132 Phase II and phase III clinical trials were conducted in late stages of nonproliferative diabetic retinopathy, with the loss of visual acuity as the primary end point. The first two clinical trials, PKC-Diabetic Retinopathy Study (PKC-DRS) and PKC-Diabetic Macular Edema Study (PKC-DMES),133, 134 failed to reach primary outcomes due to multiple factors (unpowered, three treatment arms of differing dosages, high drop-out rate of patients). However, there was a significant reduction in the secondary endpoint of the progression of diabetic macular edema to a certain stage. A much larger clinical trial, PKC-DRS2, was undertaken using a single oral dose (32 mg versus placebo), with the primary endpoint of visual acuity rather that diabetic retinopathy progression.135 The results showed that RBX significantly prevents reduction of visual acuity in diabetic patients with moderate visual loss and decreases the onset of diabetic macular edema.133 These clinical results suggest that PKC activation, especially of the -β isoform, participates in the development of diabetic retinopathy. However, because treatment with RBX preserves visual acuity by decreasing capillary permeability or targeting the neural retina but cannot significantly delay the progression of diabetic retinopathy, these data suggest that inhibition of the PKC-β isoform alone is not enough to stop the early metabolic changes that are likely driving the progression of pre-proliferative DR.

Table 2.

Clinical trials using a PKC inhibitor.

Clinical trials Patients Treatment Clinical outcomes
Aiello et al. (2006)131 N = 29 RBX 4, 16, 32 mg/day (1 month, dose-dependent) Normalization of RBF at 32 mg/day
PKC-DRS (2005)133 N = 252 RBX 8, 16, 32 mg/day (36–46 months) vs. placebo Significantly delayed occurrence of moderate visual impairment; no reduction in progression of proliferative DR
PKC-DMES (2007)134 N = 686 RBX 4, 16, 32 mg/day (min 30 months) vs. placebo No significant effect of RBX to reduce progression of diabetic macular edema
PKC-DRS2 (2006)135 N = 685 RBX 32 mg/day (36 months) vs. placebo 40% risk reduction in vision loss
Tuttle et al. (2005)137 N = 123 RBX 32 mg/day (12 months) vs. placebo Decrease in urinary ACR
Vinik et al. (2005)138 N = 205 RBX 32, 64 mg/day (1, 3, 6, 12 months) vs. placebo Improvement with RBX 64 mg/day in patients with NTSS-6 > 6 at baseline
Beckman et al. (2002)143 N = 15 RBX 32 mg/day (7 days) vs. placebo Significant inhibition of endothelium-dependent vasodilatation after hyperglycemic clamp
Mehta et al. (2009)144 N = 108 RBX 32 mg/day (6–8 weeks) vs. placebo Trend but not significant improvement in flow-mediated dilatation (FMD)

The role of PKC-β was also evaluated in patients with diabetic nephropathy. In kidney biopsies of diabetic patients, quantitative real-time PCR analysis showed a 9.9-fold increase in PKC-β mRNA expression as compared to control subjects.136 A phase II clinical trial was conducted using RBX (32 mg/day) to determine whether PKC-β inhibition can be effective in type 1 and 2 diabetic patients with high proteinuria (>300 mg/day) treated with ACE inhibitors or angiotensin-receptor blockers. Results suggested that one year of treatment with RBX in addition to angiotensin II inhibitors or receptor blockers decreases the loss of glomerular filtration rate and proteinuria in diabetic patients.137 However, more data and phase III clinical trials are needed to confirm the role of PKC and its subsequent signaling pathways in DN, especially in very early histopathological manifestations.

Vinik and collaborators conducted a one-year trial of RBX treatment, using a standardized clinical neurologic examination to measure changes in neuropathy sensory symptoms by the Neuropathy Total Symptoms Score–6 (NTSS-6) and quantitative sensory testing by the vibration detection threshold.138 Patients with diabetic peripheral neuropathy taking placebo versus either 32 or 64 mg of RBX were evaluated at baseline and at 1, 3, 6, and 12 months in each group.138 RBX had no significant effect on the overall patient population. However, results showed a significant change in the NTSS-6 at 6 months and 12 months in patients who had clinically significant neuropathy sensory symptoms (NTSS-6 >6) at baseline when treated with RBX 64 mg compared with placebo. There were no treatment-related differences in change from baseline to endpoint for vibration detection threshold among all symptomatic patients. However, in a subset of patients with less severe symptomatic diabetic peripheral neuropathy, as defined by a measurable sural nerve action potential, RBX (32 mg and 64 mg) statistically improved vibration detection threshold compared with placebo.138

Abnormal endothelial function has been demonstrated in type 1 and type 2 diabetes, as well as in obese, insulin-resistant patients whose insulin sensitivity correlated with the magnitude of endothelium-dependent vasodilation.139141 Vascular permeability is increased as early as 4–6 weeks’ duration of diabetes in patients, supporting the concept of EC dysfunction.142 A previous study also reported that infusion of glucose for six hours decreases endothelium-dependent vasodilation in nondiabetic and healthy people.143 In the natural history of type 2 diabetes, the onset of hyperglycemia precedes endothelial dysfunction,141 whereas onset of endothelial dysfunction coincides with the presence of hyperglycemia in type 1 diabetes. Treatment with RBX (32 mg/day for 7 days) prevented endothelium-dependent vasodilatation abnormalities induced by hyperglycemia.143 Moreover, a recent small, double-masked, placebo-controlled study in type 2 diabetes showed that RBX (32 mg/day) after 6 weeks improves femoral-mediated dilatation at 1 and 5 minutes after cuff deflation as compared to placebo.144 These data suggest that PKC activation, especially the -β isoform induced by hyperglycemia, may be responsible for the endothelial dysfunction observed in diabetic patients. Clearly, further studies are needed to determine whether RBX can effectively improve endothelial function in type 1 and 2 diabetic patients.

Summary

In summary, many years of human and animal data confirm the long-held belief that hyperglycemia perturbs arteries and vascular cell function. The source of such complications in diabetes is certainly multifactorial, with key roles identified for oxidants and glycation metabolites. However, a large body of literature supports hypothesis that hyperglycemia or diabetes leads to vascular DAG accumulation and ensuing PKC activation, causing a variety of cardiovascular defects. Indeed, modulation of PKC signaling transduction pathways affects the pathogenesis of various diabetic vascular complications. Activation of specific PKC isoforms by hyperglycemia and lipid metabolites is likely to be responsible for specific vascular pathologies such as EC and SMC dysfunction, ECM synthesis and fibrosis, monocyte activation, deregulation of cytokines, and vascular insulin dysfunction. These intermediate cellular alterations contribute significantly to the development of macrovascular complications such as cardiomyopathy and acceleration of atherosclerosis, and microvascular complications such as retinopathy and nephropathy. Clinical trials have shown potential for PKC-β inhibitor as a therapy for diabetic vascular complications. However, not all clinical trials demonstrated positive effects of PKC-β inhibition. For example, RBX trials did not exhibit robust effects on the improvement of painful neuropathy or sexual dysfunction in diabetic patients.145 Therefore, more clinical trials targeting multiple PKC isoforms are urgently needed to test the effectiveness in delaying, stopping and even reversing diabetic vascular complications.

Acknowledgments

This review used data supported by grants from the ADA (1-08-RA-93), NIH (R01 EY016150, R01 DK071359), DERC (5 P30 DK36836-23), and JDRF (1-2008-330, 8-2008-363, 25-2008-383) awarded to George King.

List of abbreviations

DCCT

Diabetes Control and Complications Trial

DAG

diacylglycerol

PKC

protein kinase C

AGC

cAMP-dependent protein kinase/protein kinase G/protein kinase C

EC

endothelial cells

SMC

smooth muscle cells

PS

phosphatidylserine

PMA

phorbol 12-myristate 13-acetate

MAPK

mitogen-activated protein kinases

PLA2

phospholipase A2

NO

nitric oxide

VEGF

vascular endothelial growth factor

ET-1

endothelin-1

eNOS

endothelial nitric oxide synthase

NADPH

nicotinamide adenine dinucleotide phosphate

VPF

vascular permeability factor

oxLDL

oxidized low density lipoprotein

LOX-1

leptin-like oxidized LDL receptor-1

ICAM-1

intercellular adhesion molecules-1

MCP-1

monocyte chemotactic protein-1

ECM

extracellular matrix

AGEs

advanced glycation end products

TGF-β

transforming growth factor-beta

MI

myocardial infarction

Erg-1

early growth response-1

VCAM

vascular cell adhesion molecule

MMP-2

matrix metalloproteinase-2

ACE

angiotensin-converting enzyme

CTGF

connective tissue growth factor

PDGF

platelet-derived growth factor

ROS

reactive oxygen species

NF-κB

nuclear factor-kappa B

RBF

retinal blood flow

PI3K

phosphoinositide 3-kinases

ERK

extracellular signal-regulated kinases

RBX

ruboxistaurin

PKC-DRS

PKC-Diabetic Retinopathy Study

PKC-DMES

PKC-Diabetic Macular Edema Study

NTSS-6

Neuropathy Total Symptoms Score–6

Footnotes

Disclosure

The authors declare no financial, personal or professional relationship with other people or organizations.

References

  • 1.King H, Aubert RE, Herman WH. Global burden of diabetes, 1995–2025: Prevalence, numerical estimates, and projections. Diabetes Care. 1998;21:1414–1431. doi: 10.2337/diacare.21.9.1414. [DOI] [PubMed] [Google Scholar]
  • 2.The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. The diabetes control and complications trial research group. N Engl J Med. 1993;329:977–986. doi: 10.1056/NEJM199309303291401. [DOI] [PubMed] [Google Scholar]
  • 3.Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (ukpds 33) Uk prospective diabetes study (UKPDS) group. Lancet. 1998;352:837–853. [PubMed] [Google Scholar]
  • 4.Keenan HA, Costacou T, Sun JK, Doria A, Cavellerano J, Coney J, Orchard TJ, Aiello LP, King GL. Clinical factors associated with resistance to microvascular complications in diabetic patients of extreme disease duration: The 50-year medalist study. Diabetes Care. 2007;30:1995–1997. doi: 10.2337/dc06-2222. [DOI] [PubMed] [Google Scholar]
  • 5.Obrosova IG, Minchenko AG, Vasupuram R, White L, Abatan OI, Kumagai AK, Frank RN, Stevens MJ. Aldose reductase inhibitor fidarestat prevents retinal oxidative stress and vascular endothelial growth factor overexpression in streptozotocin-diabetic rats. Diabetes. 2003;52:864–871. doi: 10.2337/diabetes.52.3.864. [DOI] [PubMed] [Google Scholar]
  • 6.Wendt T, Harja E, Bucciarelli L, Qu W, Lu Y, Rong LL, Jenkins DG, Stein G, Schmidt AM, Yan SF. Rage modulates vascular inflammation and atherosclerosis in a murine model of type 2 diabetes. Atherosclerosis. 2006;185:70–77. doi: 10.1016/j.atherosclerosis.2005.06.013. [DOI] [PubMed] [Google Scholar]
  • 7.Brownlee M. Biochemistry and molecular cell biology of diabetic complications. Nature. 2001;414:813–820. doi: 10.1038/414813a. [DOI] [PubMed] [Google Scholar]
  • 8.Koya D, King GL. Protein kinase C activation and the development of diabetic complications. Diabetes. 1998;47:859–866. doi: 10.2337/diabetes.47.6.859. [DOI] [PubMed] [Google Scholar]
  • 9.Nishizuka Y. Intracellular signaling by hydrolysis of phospholipids and activation of protein kinase C. Science. 1992;258:607–614. doi: 10.1126/science.1411571. [DOI] [PubMed] [Google Scholar]
  • 10.Xia P, Inoguchi T, Kern TS, Engerman RL, Oates PJ, King GL. Characterization of the mechanism for the chronic activation of diacylglycerol-protein kinase C pathway in diabetes and hypergalactosemia. Diabetes. 1994;43:1122–1129. doi: 10.2337/diab.43.9.1122. [DOI] [PubMed] [Google Scholar]
  • 11.Shiba T, Inoguchi T, Sportsman JR, Heath WF, Bursell S, King GL. Correlation of diacylglycerol level and protein kinase C activity in rat retina to retinal circulation. Am J Physiol. 1993;265:E783–793. doi: 10.1152/ajpendo.1993.265.5.E783. [DOI] [PubMed] [Google Scholar]
  • 12.Inoguchi T, Battan R, Handler E, Sportsman JR, Heath W, King GL. Preferential elevation of protein kinase C isoform beta II and diacylglycerol levels in the aorta and heart of diabetic rats: Differential reversibility to glycemic control by islet cell transplantation. Proc Natl Acad Sci U S A. 1992;89:11059–11063. doi: 10.1073/pnas.89.22.11059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Ishii H, Jirousek MR, Koya D, Takagi C, Xia P, Clermont A, Bursell SE, Kern TS, Ballas LM, Heath WF, Stramm LE, Feener EP, King GL. Amelioration of vascular dysfunctions in diabetic rats by an oral PKCbeta inhibitor. Science (New York, N Y. 1996;272:728–731. doi: 10.1126/science.272.5262.728. [DOI] [PubMed] [Google Scholar]
  • 14.Craven PA, Davidson CM, DeRubertis FR. Increase in diacylglycerol mass in isolated glomeruli by glucose from de novo synthesis of glycerolipids. Diabetes. 1990;39:667–674. doi: 10.2337/diab.39.6.667. [DOI] [PubMed] [Google Scholar]
  • 15.van Herpen NA, Schrauwen-Hinderling VB. Lipid accumulation in non-adipose tissue and lipotoxicity. Physiol Behav. 2008;94:231–241. doi: 10.1016/j.physbeh.2007.11.049. [DOI] [PubMed] [Google Scholar]
  • 16.Ido Y, McHowat J, Chang KC, Arrigoni-Martelli E, Orfalian Z, Kilo C, Corr PB, Williamson JR. Neural dysfunction and metabolic imbalances in diabetic rats. Prevention by acetyl-L-carnitine. Diabetes. 1994;43:1469–1477. doi: 10.2337/diab.43.12.1469. [DOI] [PubMed] [Google Scholar]
  • 17.Geraldes P, Hiraoka-Yamamoto J, Matsumoto M, Clermont A, Leitges M, Marette A, Aiello LP, Kern TS, King GL. Activation of PKC-delta and SHP-1 by hyperglycemia causes vascular cell apoptosis and diabetic retinopathy. Nat Med. 2009;15:1298–1306. doi: 10.1038/nm.2052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Ayo SH, Radnik R, Garoni JA, Troyer DA, Kreisberg JI. High glucose increases diacylglycerol mass and activates protein kinase Cin mesangial cell cultures. Am J Physiol. 1991;261:F571–577. doi: 10.1152/ajprenal.1991.261.4.F571. [DOI] [PubMed] [Google Scholar]
  • 19.Newton AC. Regulation of the abc kinases by phosphorylation: Protein kinase C as a paradigm. Biochem J. 2003;370:361–371. doi: 10.1042/BJ20021626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Steinberg SF. Structural basis of protein kinase C isoform function. Physiol Rev. 2008;88:1341–1378. doi: 10.1152/physrev.00034.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Corbalan-Garcia S, Gomez-Fernandez JC. Protein kinase C regulatory domains: The art of decoding many different signals in membranes. Biochim Biophys Acta. 2006;1761:633–654. doi: 10.1016/j.bbalip.2006.04.015. [DOI] [PubMed] [Google Scholar]
  • 22.Churchill EN, Qvit N, Mochly-Rosen D. Rationally designed peptide regulators of protein kinase C. Trends Endocrinol Metab. 2009;20:25–33. doi: 10.1016/j.tem.2008.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Konishi H, Tanaka M, Takemura Y, Matsuzaki H, Ono Y, Kikkawa U, Nishizuka Y. Activation of protein kinase C by tyrosine phosphorylation in response to H2O2. Proc Natl Acad Sci U S A. 1997;94:11233–11237. doi: 10.1073/pnas.94.21.11233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Nishikawa T, Edelstein D, Du XL, Yamagishi S, Matsumura T, Kaneda Y, Yorek MA, Beebe D, Oates PJ, Hammes HP, Giardino I, Brownlee M. Normalizing mitochondrial superoxide production blocks three pathways of hyperglycaemic damage. Nature. 2000;404:787–790. doi: 10.1038/35008121. [DOI] [PubMed] [Google Scholar]
  • 25.Lynch JJ, Ferro TJ, Blumenstock FA, Brockenauer AM, Malik AB. Increased endothelial albumin permeability mediated by protein kinase C activation. J Clin Invest. 1990;85:1991–1998. doi: 10.1172/JCI114663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Wolf BA, Williamson JR, Easom RA, Chang K, Sherman WR, Turk J. Diacylglycerol accumulation and microvascular abnormalities induced by elevated glucose levels. J Clin Invest. 1991;87:31–38. doi: 10.1172/JCI114988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Inoguchi T, Ueda F, Umeda F, Yamashita T, Nawata H. Inhibition of intercellular communication via gap junction in cultured aortic endothelial cells by elevated glucose and phorbol ester. Biochem Biophys Res Commun. 1995;208:492–497. doi: 10.1006/bbrc.1995.1365. [DOI] [PubMed] [Google Scholar]
  • 28.Park JY, Takahara N, Gabriele A, Chou E, Naruse K, Suzuma K, Yamauchi T, Ha SW, Meier M, Rhodes CJ, King GL. Induction of endothelin-1 expression by glucose: An effect of protein kinase Cactivation. Diabetes. 2000;49:1239–1248. doi: 10.2337/diabetes.49.7.1239. [DOI] [PubMed] [Google Scholar]
  • 29.Nagpala PG, Malik AB, Vuong PT, Lum H. Protein kinase C beta 1 overexpression augments phorbol ester-induced increase in endothelial permeability. J Cell Physiol. 1996;166:249–255. doi: 10.1002/(SICI)1097-4652(199602)166:2<249::AID-JCP2>3.0.CO;2-P. [DOI] [PubMed] [Google Scholar]
  • 30.Hempel A, Maasch C, Heintze U, Lindschau C, Dietz R, Luft FC, Haller H. High glucose concentrations increase endothelial cell permeability via activation of protein kinase C alpha. Circ Res. 1997;81:363–371. doi: 10.1161/01.res.81.3.363. [DOI] [PubMed] [Google Scholar]
  • 31.Hink U, Li H, Mollnau H, Oelze M, Matheis E, Hartmann M, Skatchkov M, Thaiss F, Stahl RA, Warnholtz A, Meinertz T, Griendling K, Harrison DG, Forstermann U, Munzel T. Mechanisms underlying endothelial dysfunction in diabetes mellitus. Circ Res. 2001;88:E14–22. doi: 10.1161/01.res.88.2.e14. [DOI] [PubMed] [Google Scholar]
  • 32.Cardillo C, Campia U, Bryant MB, Panza JA. Increased activity of endogenous endothelin in patients with type 2diabetes mellitus. Circulation. 2002;106:1783–1787. doi: 10.1161/01.cir.0000032260.01569.64. [DOI] [PubMed] [Google Scholar]
  • 33.Cosentino F, Eto M, De Paolis P, van der Loo B, Bachschmid M, Ullrich V, Kouroedov A, Delli Gatti C, Joch H, Volpe M, Luscher TF. High glucose causesupregulation of cyclooxygenase-2 and alters prostanoid profile in human endothelial cells: Role of protein kinase C and reactive oxygen species. Circulation. 2003;107:1017–1023. doi: 10.1161/01.cir.0000051367.92927.07. [DOI] [PubMed] [Google Scholar]
  • 34.Ding Y, Vaziri ND, Coulson R, Kamanna VS, Roh DD. Effects of simulated hyperglycemia, insulin, and glucagon on endothelial nitric oxide synthase expression. Am J Physiol Endocrinol Metab. 2000;279:E11–17. doi: 10.1152/ajpendo.2000.279.1.E11. [DOI] [PubMed] [Google Scholar]
  • 35.Inoguchi T, Li P, Umeda F, Yu HY, Kakimoto M, Imamura M, Aoki T, Etoh T, Hashimoto T, Naruse M, Sano H, Utsumi H, Nawata H. High glucose level and free fatty acid stimulate reactive oxygen species production through protein kinase C-dependent activation of NAD(P)H oxidase in cultured vascular cells. Diabetes. 2000;49:1939–1945. doi: 10.2337/diabetes.49.11.1939. [DOI] [PubMed] [Google Scholar]
  • 36.Quagliaro L, Piconi L, Assaloni R, Martinelli L, Motz E, Ceriello A. Intermittent high glucose enhances apoptosis related to oxidative stress in human umbilical vein endothelial cells: The role of protein kinase C and NAD(P)H-oxidase activation. Diabetes. 2003;52:2795–2804. doi: 10.2337/diabetes.52.11.2795. [DOI] [PubMed] [Google Scholar]
  • 37.Xia P, Aiello LP, Ishii H, Jiang ZY, Park DJ, Robinson GS, Takagi H, Newsome WP, Jirousek MR, King GL. Characterization of vascular endothelial growth factor’s effect on the activation of protein kinase c, its isoforms, and endothelial cell growth. J Clin Invest. 1996;98:2018–2026. doi: 10.1172/JCI119006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Harhaj NS, Felinski EA, Wolpert EB, Sundstrom JM, Gardner TW, Antonetti DA. VEGF activation of protein kinase C stimulates occludin phosphorylation and contributes to endothelial permeability. Invest Ophthalmol Vis Sci. 2006;47:5106–5115. doi: 10.1167/iovs.06-0322. [DOI] [PubMed] [Google Scholar]
  • 39.Natarajan R, Gonzales N, Xu L, Nadler JL. Vascular smooth muscle cells exhibit increased growth in response to elevated glucose. Biochem Biophys Res Com. 1992;187:552–560. doi: 10.1016/s0006-291x(05)81529-3. [DOI] [PubMed] [Google Scholar]
  • 40.Sakai Y, Inazu M, Shamoto A, Zhu B, Homma I. Contractile hyperreactivity and alteration of PKC activity in gastric fundus smooth muscle of diabetic rats. Pharmacol Biochem Behav. 1994;49:669–674. doi: 10.1016/0091-3057(94)90086-8. [DOI] [PubMed] [Google Scholar]
  • 41.Igarashi M, Wakasaki H, Takahara N, Ishii H, Jiang ZY, Yamauchi T, Kuboki K, Meier M, Rhodes CJ, King GL. Glucose or diabetes activates p38 mitogen-activated protein kinase via different pathways. J Clin Invest. 1999;103:185–195. doi: 10.1172/JCI3326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Hall JL, Matter CM, Wang X, Gibbons GH. Hyperglycemia inhibits vascular smooth muscle cell apoptosis through a protein kinase C-dependent pathway. Circ Res. 2000;87:574–580. doi: 10.1161/01.res.87.7.574. [DOI] [PubMed] [Google Scholar]
  • 43.Osterud B, Bjorklid E. Role of monocytes in atherogenesis. Physiol Rev. 2003;83:1069–1112. doi: 10.1152/physrev.00005.2003. [DOI] [PubMed] [Google Scholar]
  • 44.Li L, Sawamura T, Renier G. Glucose enhances human macrophage LOX-1 expression: Role for LOX-1 in glucose-induced macrophage foam cell formation. CircRes. 2004;94:892–901. doi: 10.1161/01.RES.0000124920.09738.26. [DOI] [PubMed] [Google Scholar]
  • 45.Ceolotto G, Gallo A, Miola M, Sartori M, Trevisan R, Del Prato S, Semplicini A, Avogaro A. Protein kinase C activity is acutely regulated by plasma glucose concentration in human monocytes in vivo. Diabetes. 1999;48:1316–1322. doi: 10.2337/diabetes.48.6.1316. [DOI] [PubMed] [Google Scholar]
  • 46.Osto E, Kouroedov A, Mocharla P, Akhmedov A, Besler C, Rohrer L, von Eckardstein A, Iliceto S, Volpe M, Luscher TF, Cosentino F. Inhibition of protein kinase Cbeta prevents foam cell formation by reducing scavenger receptor a expression in human macrophages. Circulation. 2008;118:2174–2182. doi: 10.1161/CIRCULATIONAHA.108.789537. [DOI] [PubMed] [Google Scholar]
  • 47.Wu Y, Wu G, Qi X, Lin H, Qian H, Shen J, Lin S. Protein kinase C beta inhibitor LY333531 attenuates intercellular adhesion molecule-1 and monocyte chemotactic protein-1 expression in the kidney in diabetic rats. J Pharmacol Sci. 2006;101:335–343. doi: 10.1254/jphs.fp0050896. [DOI] [PubMed] [Google Scholar]
  • 48.Dasu MR, Devaraj S, Zhao L, Hwang DH, Jialal I. High glucose induces toll-like receptor expression in human monocytes: Mechanism of activation. Diabetes. 2008;57:3090–3098. doi: 10.2337/db08-0564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Scheinman JI, Fish AJ, Matas AJ, Michael AF. The immunohistopathology of glomerular antigens. II. The glomerular basement membrane, actomyosin, and fibroblast surface antigens in normal, diseased, and transplanted human kidneys. Am J Pathol. 1978;90:71–88. [PMC free article] [PubMed] [Google Scholar]
  • 50.Bruneval P, Foidart JM, Nochy D, Camilleri JP, Bariety J. Glomerular matrix proteins in nodular glomerulosclerosis in association with light chain deposition disease and diabetes mellitus. Hum Patholy. 1985;16:477–484. doi: 10.1016/s0046-8177(85)80086-1. [DOI] [PubMed] [Google Scholar]
  • 51.Studer RK, Craven PA, DeRubertis FR. Role for protein kinase C in the mediation of increased fibronectin accumulation by mesangial cells grown in high-glucose medium. Diabetes. 1993;42:118–126. doi: 10.2337/diab.42.1.118. [DOI] [PubMed] [Google Scholar]
  • 52.Fumo P, Kuncio GS, Ziyadeh FN. Pkc and high glucose stimulate collagen alpha 1 (IV) transcriptional activity in a reporter mesangial cell line. Am J Physiol. 1994;267:F632–638. doi: 10.1152/ajprenal.1994.267.4.F632. [DOI] [PubMed] [Google Scholar]
  • 53.Kapor-Drezgic J, Zhou X, Babazono T, Dlugosz JA, Hohman T, Whiteside C. Effect of high glucose on mesangial cell protein kinase C-delta and -epsilon is polyol pathway-dependent. J Am Soc Nephrol. 1999;10:1193–1203. doi: 10.1681/ASN.V1061193. [DOI] [PubMed] [Google Scholar]
  • 54.Baccora MH, Cortes P, Hassett C, Taube DW, Yee J. Effects of long-term elevated glucose on collagen formation by mesangial cells. Kidney Int. 2007;72:1216–1225. doi: 10.1038/sj.ki.5002517. [DOI] [PubMed] [Google Scholar]
  • 55.Padival AK, Hawkins KS, Huang C. High glucose-induced membrane translocation of PKC beta1 is associated with Arf6 in glomerular mesangial cells. Mol Cell Biochem. 2004;258:129–135. doi: 10.1023/b:mcbi.0000012847.86529.07. [DOI] [PubMed] [Google Scholar]
  • 56.Wu D, Peng F, Zhang B, Ingram AJ, Kelly DJ, Gilbert RE, Gao B, Krepinsky JC. PKC-beta1 mediates glucose-induced Akt activation and TGF-beta1 upregulation in mesangial cells. J Am Soc Nephrol. 2009;20:554–566. doi: 10.1681/ASN.2008040445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Whiteside CI, Dlugosz JA. Mesangial cell protein kinase C isozyme activation in the diabetic milieu. Am J Physiol. 2002;282:F975–980. doi: 10.1152/ajprenal.00014.2002. [DOI] [PubMed] [Google Scholar]
  • 58.Lal MA, Brismar H, Eklof AC, Aperia A. Role of oxidative stress in advanced glycation end product-induced mesangial cell activation. Kidney Int. 2002;61:2006–2014. doi: 10.1046/j.1523-1755.2002.00367.x. [DOI] [PubMed] [Google Scholar]
  • 59.Scivittaro V, Ganz MB, Weiss MF. AGEs induce oxidative stress and activate protein kinase C-beta(II) in neonatal mesangial cells. Am J Physiol. 2000;278:F676–683. doi: 10.1152/ajprenal.2000.278.4.F676. [DOI] [PubMed] [Google Scholar]
  • 60.Koya D, Jirousek MR, Lin YW, Ishii H, Kuboki K, King GL. Characterization of protein kinase c beta isoform activation on the gene expression of transforming growth factor-beta, extracellular matrix components, and prostanoids in the glomeruli of diabetic rats. J Clin Invest. 1997;100:115–126. doi: 10.1172/JCI119503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Xia L, Wang H, Munk S, Kwan J, Goldberg HJ, Fantus IG, Whiteside CI. High glucose activates PKC-zeta and NADPH oxidase through autocrine TGF-beta1 signaling in mesangial cells. Am J Physiol. 2008;295:F1705–1714. doi: 10.1152/ajprenal.00043.2008. [DOI] [PubMed] [Google Scholar]
  • 62.Ross R. Atherosclerosis--an inflammatory disease. N Engl J Med. 1999;340:115–126. doi: 10.1056/NEJM199901143400207. [DOI] [PubMed] [Google Scholar]
  • 63.Glass CK, Witztum JL. Atherosclerosis. The road ahead. Cell. 2001;104:503–516. doi: 10.1016/s0092-8674(01)00238-0. [DOI] [PubMed] [Google Scholar]
  • 64.Libby P. Inflammation in atherosclerosis. Nature. 2002;420:868–874. doi: 10.1038/nature01323. [DOI] [PubMed] [Google Scholar]
  • 65.Nathan DM, Lachin J, Cleary P, Orchard T, Brillon DJ, Backlund JY, O’Leary DH, Genuth S. Intensive diabetes therapy and carotid intima-media thickness in type 1 diabetes mellitus. N Engl J Med. 2003;348:2294–2303. doi: 10.1056/NEJMoa022314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Harja E, Chang JS, Lu Y, Leitges M, Zou YS, Schmidt AM, Yan SF. Mice deficient in PKCbeta and apolipoprotein Edisplay decreased atherosclerosis. FASEBJ. 2009;23:1081–1091. doi: 10.1096/fj.08-120345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Leitges M, Mayr M, Braun U, Mayr U, Li C, Pfister G, Ghaffari-Tabrizi N, Baier G, Hu Y, Xu Q. Exacerbated vein graft arteriosclerosis in protein kinase cdelta-null mice. J Clin Invest. 2001;108:1505–1512. doi: 10.1172/JCI12902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Bell DS. Diabetic cardiomyopathy. A unique entity or a complication of coronary artery disease? Diabetes care. 1995;18:708–714. doi: 10.2337/diacare.18.5.708. [DOI] [PubMed] [Google Scholar]
  • 69.Abaci A, Oguzhan A, Kahraman S, Eryol NK, Unal S, Arinc H, Ergin A. Effect of diabetes mellitus on formation of coronary collateral vessels. Circulation. 1999;99:2239–2242. doi: 10.1161/01.cir.99.17.2239. [DOI] [PubMed] [Google Scholar]
  • 70.Stone PH, Muller JE, Hartwell T, York BJ, Rutherford JD, Parker CB, Turi ZG, Strauss HW, Willerson JT, Robertson T, et al. The effect of diabetes mellitus on prognosis and serial left ventricular function after acute myocardial infarction: Contribution of both coronary disease and diastolic left ventricular dysfunction to the adverse prognosis. The milis study group. J Am Coll Cardiol. 1989;14:49–57. doi: 10.1016/0735-1097(89)90053-3. [DOI] [PubMed] [Google Scholar]
  • 71.Bowling N, Walsh RA, Song G, Estridge T, Sandusky GE, Fouts RL, Mintze K, Pickard T, Roden R, Bristow MR, Sabbah HN, Mizrahi JL, Gromo G, King GL, Vlahos CJ. Increased protein kinase C activity and expression of Ca2+-sensitive isoforms in the failing human heart. Circulation. 1999;99:384–391. doi: 10.1161/01.cir.99.3.384. [DOI] [PubMed] [Google Scholar]
  • 72.Guo M, Wu MH, Korompai F, Yuan SY. Upregulation of PKCgenes and isozymes in cardiovascular tissues during early stages of experimental diabetes. Physiol Genomics. 2003;12:139–146. doi: 10.1152/physiolgenomics.00125.2002. [DOI] [PubMed] [Google Scholar]
  • 73.Way KJ, Isshiki K, Suzuma K, Yokota T, Zvagelsky D, Schoen FJ, Sandusky GE, Pechous PA, Vlahos CJ, Wakasaki H, King GL. Expression of connective tissue growth factor is increased in injured myocardium associated with protein kinase C beta2 activation and diabetes. Diabetes. 2002;51:2709–2718. doi: 10.2337/diabetes.51.9.2709. [DOI] [PubMed] [Google Scholar]
  • 74.Arikawa E, Ma RC, Isshiki K, Luptak I, He Z, Yasuda Y, Maeno Y, Patti ME, Weir GC, Harris RA, Zammit VA, Tian R, King GL. Effects of insulin replacements, inhibitors of angiotensin, and PKCbeta’s actions to normalize cardiac gene expression and fuel metabolism in diabetic rats. Diabetes. 2007;56:1410–1420. doi: 10.2337/db06-0655. [DOI] [PubMed] [Google Scholar]
  • 75.Connelly KA, Kelly DJ, Zhang Y, Prior DL, Advani A, Cox AJ, Thai K, Krum H, Gilbert RE. Inhibition of protein kinase Cin diabetic cardiomyopathy. Circ Heart Fail. 2009;2:129–137. doi: 10.1161/CIRCHEARTFAILURE.108.765750. [DOI] [PubMed] [Google Scholar]
  • 76.Liu Q, Chen X, Macdonnell SM, Kranias EG, Lorenz JN, Leitges M, Houser SR, Molkentin JD. Protein kinase C{alpha}, but not PKC{beta} or PKC{gamma}, regulates contractility and heart failure susceptibility: Implications for ruboxistaurin as a novel therapeutic approach. Circ Res. 2009;105:194–200. doi: 10.1161/CIRCRESAHA.109.195313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.He Z, Way KJ, Arikawa E, Chou E, Opland DM, Clermont A, Isshiki K, Ma RC, Scott JA, Schoen FJ, Feener EP, King GL. Differential regulation of angiotensin II-induced expression of connective tissue growth factor by protein kinase C isoforms in the myocardium. J Biol Chem. 2005;280:15719–15726. doi: 10.1074/jbc.M413493200. [DOI] [PubMed] [Google Scholar]
  • 78.Churchill EN, Mochly-Rosen D. The roles of PKCdelta and epsilon isoenzymes in the regulation of myocardial ischaemia/reperfusion injury. Biochem Soc Trans. 2007;35:1040–1042. doi: 10.1042/BST0351040. [DOI] [PubMed] [Google Scholar]
  • 79.Malhotra A, Kang BP, Hashmi S, Meggs LG. PKCepsilon inhibits the hyperglycemia-induced apoptosis signal in adult rat ventricular myocytes. Mol Cell Biochem. 2005;268:169–173. doi: 10.1007/s11010-005-3858-6. [DOI] [PubMed] [Google Scholar]
  • 80.Kolter T, Uphues I, Eckel J. Molecular analysis of insulin resistance in isolated ventricular cardiomyocytes of obese zucker rats. Am J Physiol. 1997;273:E59–67. doi: 10.1152/ajpendo.1997.273.1.E59. [DOI] [PubMed] [Google Scholar]
  • 81.Davidoff AJ, Davidson MB, Carmody MW, Davis ME, Ren J. Diabetic cardiomyocyte dysfunction and myocyte insulin resistance: Role of glucose-induced PKC activity. Mol Cell Biochem. 2004;262:155–163. doi: 10.1023/b:mcbi.0000038231.68078.4b. [DOI] [PubMed] [Google Scholar]
  • 82.McQueen AP, Zhang D, Hu P, Swenson L, Yang Y, Zaha VG, Hoffman JL, Yun UJ, Chakrabarti G, Wang Z, Albertine KH, Abel ED, Litwin SE. Contractile dysfunction in hypertrophied hearts with deficient insulin receptor signaling: Possible role of reduced capillary density. J Mol Cell Cardiol. 2005;39:882–892. doi: 10.1016/j.yjmcc.2005.07.017. [DOI] [PubMed] [Google Scholar]
  • 83.Yoon YS, Uchida S, Masuo O, Cejna M, Park JS, Gwon HC, Kirchmair R, Bahlman F, Walter D, Curry C, Hanley A, Isner JM, Losordo DW. Progressive attenuation of myocardial vascular endothelial growth factor expression is a seminal event in diabetic cardiomyopathy: Restoration of microvascular homeostasis and recovery of cardiac function in diabetic cardiomyopathy after replenishment of local vascular endothelial growth factor. Circulation. 2005;111:2073–2085. doi: 10.1161/01.CIR.0000162472.52990.36. [DOI] [PubMed] [Google Scholar]
  • 84.Chou E, Suzuma I, Way KJ, Opland D, Clermont AC, Naruse K, Suzuma K, Bowling NL, Vlahos CJ, Aiello LP, King GL. Decreased cardiac expression of vascular endothelial growth factor and its receptors in insulin-resistant and diabetic states: A possible explanation for impaired collateral formation in cardiac tissue. Circulation. 2002;105:373–379. doi: 10.1161/hc0302.102143. [DOI] [PubMed] [Google Scholar]
  • 85.Jesmin S, Zaedi S, Shimojo N, Iemitsu M, Masuzawa K, Yamaguchi N, Mowa CN, Maeda S, Hattori Y, Miyauchi T. Endothelin antagonism normalizes vegf signaling and cardiac function in stz-induced diabetic rat hearts. Am J Physiol. 2007;292:E1030–1040. doi: 10.1152/ajpendo.00517.2006. [DOI] [PubMed] [Google Scholar]
  • 86.Sasso FC, Torella D, Carbonara O, Ellison GM, Torella M, Scardone M, Marra C, Nasti R, Marfella R, Cozzolino D, Indolfi C, Cotrufo M, Torella R, Salvatore T. Increased vascular endothelial growth factor expression but impaired vascular endothelial growth factor receptor signaling in the myocardium of type 2 diabetic patients with chronic coronary heart disease. J Am Coll Cardiol. 2005;46:827–834. doi: 10.1016/j.jacc.2005.06.007. [DOI] [PubMed] [Google Scholar]
  • 87.Frank RN. Diabetic retinopathy. N Engl J Med. 2004;350:48–58. doi: 10.1056/NEJMra021678. [DOI] [PubMed] [Google Scholar]
  • 88.Idris I, Gray S, Donnelly R. Protein kinase c activation: Isozyme-specific effects on metabolism and cardiovascular complications in diabetes. Diabetologia. 2001;44:659–673. doi: 10.1007/s001250051675. [DOI] [PubMed] [Google Scholar]
  • 89.Way KJ, Katai N, King GL. Protein kinase C and the development of diabetic vascular complications. Diabet Med. 2001;18:945–959. doi: 10.1046/j.0742-3071.2001.00638.x. [DOI] [PubMed] [Google Scholar]
  • 90.Small KW, Stefansson E, Hatchell DL. Retinal blood flow in normal and diabetic dogs. Invest Ophthalmol Vis Sci. 1987;28:672–675. [PubMed] [Google Scholar]
  • 91.Bursell SE, Clermont AC, Kinsley BT, Simonson DC, Aiello LM, Wolpert HA. Retinal blood flow changes in patients with insulin-dependent diabetes mellitus and no diabeticretinopathy. Invest Ophthalmol Vis Sci. 1996;37:886–897. [PubMed] [Google Scholar]
  • 92.Hostetter TH, Troy JL, Brenner BM. Glomerular hemodynamics in experimental diabetes mellitus. Kidney Int. 1981;19:410–415. doi: 10.1038/ki.1981.33. [DOI] [PubMed] [Google Scholar]
  • 93.Yokota T, Ma RC, Park JY, Isshiki K, Sotiropoulos KB, Rauniyar RK, Bornfeldt KE, King GL. Role of protein kinase C on the expression of platelet-derived growth factor and endothelin-1 in the retina of diabetic rats and cultured retinal capillary pericytes. Diabetes. 2003;52:838–845. doi: 10.2337/diabetes.52.3.838. [DOI] [PubMed] [Google Scholar]
  • 94.Bursell SE, Takagi C, Clermont AC, Takagi H, Mori F, Ishii H, King GL. Specific retinal diacylglycerol and protein kinase C beta isoform modulation mimics abnormal retinal hemodynamics in diabetic rats. Invest Ophthalmol Vis Sci. 1997;38:2711–2720. [PubMed] [Google Scholar]
  • 95.Kamata K, Miyata N, Kasuya Y. Involvement of endothelial cells in relaxation and contraction responses of the aorta to isoproterenol in naive and streptozotocin-induced diabetic rats. The J Pharmacol Exp Ther. 1989;249:890–894. [PubMed] [Google Scholar]
  • 96.Mayhan WG. Impairment of endothelium-dependent dilatation of cerebral arterioles during diabetes mellitus. Am J Physiol. 1989;256:H621–62. doi: 10.1152/ajpheart.1989.256.3.H621. [DOI] [PubMed] [Google Scholar]
  • 97.Kamata K, Chikada S, Umeda F, Kasuya Y. Effects of phorbol ester on vasodilation induced by endothelium-dependent or endothelium-independent vasodilators in the mesenteric arterial bed. J Cardiovasc Pharmacol. 1995;26:645–652. doi: 10.1097/00005344-199510000-00021. [DOI] [PubMed] [Google Scholar]
  • 98.Suzuma K, Takahara N, Suzuma I, Isshiki K, Ueki K, Leitges M, Aiello LP, King GL. Characterization of protein kinase C beta isoform’s action on retinoblastoma protein phosphorylation, vascular endothelial growth factor-induced endothelial cell proliferation, and retinal neovascularization. Proc Natl Acad Sci U S A. 2002;99:721–726. doi: 10.1073/pnas.022644499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Seaquist ER, Goetz FC, Rich S, Barbosa J. Familial clustering of diabetic kidney disease. Evidence for genetic susceptibility to diabetic nephropathy. N Engl J Med. 1989;320:1161–1165. doi: 10.1056/NEJM198905043201801. [DOI] [PubMed] [Google Scholar]
  • 100.Krolewski AS, Warram JH, Christlieb AR, Busick EJ, Kahn CR. The changing natural history of nephropathy in type Idiabetes. Am J Med. 1985;78:785–794. doi: 10.1016/0002-9343(85)90284-0. [DOI] [PubMed] [Google Scholar]
  • 101.Menne J, Park JK, Boehne M, Elger M, Lindschau C, Kirsch T, Meier M, Gueler F, Fiebeler A, Bahlmann FH, Leitges M, Haller H. Diminished loss of proteoglycans and lack of albuminuria in protein kinase C-alpha-deficient diabetic mice. Diabetes. 2004;53:2101–2109. doi: 10.2337/diabetes.53.8.2101. [DOI] [PubMed] [Google Scholar]
  • 102.Thallas-Bonke V, Thorpe SR, Coughlan MT, Fukami K, Yap FY, Sourris KC, Penfold SA, Bach LA, Cooper ME, Forbes JM. Inhibition of NADPH oxidase prevents advanced glycation end product-mediated damage in diabetic nephropathy through a protein kinase C-alpha-dependent pathway. Diabetes. 2008;57:460–469. doi: 10.2337/db07-1119. [DOI] [PubMed] [Google Scholar]
  • 103.Ohshiro Y, Ma RC, Yasuda Y, Hiraoka-Yamamoto J, Clermont AC, Isshiki K, Yagi K, Arikawa E, Kern TS, King GL. Reduction of diabetes-induced oxidative stress, fibrotic cytokine expression, and renal dysfunction in protein kinase Cbeta-null mice. Diabetes. 2006;55:3112–3120. doi: 10.2337/db06-0895. [DOI] [PubMed] [Google Scholar]
  • 104.Koya D, Haneda M, Nakagawa H, Isshiki K, Sato H, Maeda S, Sugimoto T, Yasuda H, Kashiwagi A, Ways DK, King GL, Kikkawa R. Amelioration of accelerated diabetic mesangial expansion by treatment with a PKC beta inhibitor in diabetic db/db mice, a rodent model for type 2 diabetes. FASEBJ. 2000;14:439–447. doi: 10.1096/fasebj.14.3.439. [DOI] [PubMed] [Google Scholar]
  • 105.Kelly DJ, Cox AJ, Tolcos M, Cooper ME, Wilkinson-Berka JL, Gilbert RE. Attenuation of tubular apoptosis by blockade of the renin-angiotensin system in diabetic ren-2 rats. Kidney Int. 2002;61:31–39. doi: 10.1046/j.1523-1755.2002.00088.x. [DOI] [PubMed] [Google Scholar]
  • 106.Kelly DJ, Edgley AJ, Zhang Y, Thai K, Tan SM, Cox AJ, Advani A, Connelly KA, Whiteside CI, Gilbert RE. Protein kinase C-beta inhibition attenuates the progression of nephropathy in non-diabetic kidney disease. Nephrol Dial Transplant. 2009;24:1782–1790. doi: 10.1093/ndt/gfn729. [DOI] [PubMed] [Google Scholar]
  • 107.Meier M, Menne J, Park JK, Holtz M, Gueler F, Kirsch T, Schiffer M, Mengel M, Lindschau C, Leitges M, Haller H. Deletion of protein kinase C-epsilon signaling pathway induces glomerulosclerosis and tubulointerstitial fibrosis in vivo. J Am Soc Nephrol. 2007;18:1190–1198. doi: 10.1681/ASN.2005070694. [DOI] [PubMed] [Google Scholar]
  • 108.Borghini I, Ania-Lahuerta A, Regazzi R, Ferrari G, Gjinovci A, Wollheim CB, Pralong WF. Alpha, beta I, beta II, delta, and epsilon protein kinase C isoforms and compound activity in the sciatic nerve of normal and diabetic rats. J Neurochem. 1994;62:686–696. [PubMed] [Google Scholar]
  • 109.Roberts RE, McLean WG. Protein kinase C isozyme expression in sciatic nerves and spinal cords of experimentally diabetic rats. Brain Res. 1997;754:147–156. doi: 10.1016/s0006-8993(97)00062-0. [DOI] [PubMed] [Google Scholar]
  • 110.Kim J, Rushovich EH, Thomas TP, Ueda T, Agranoff BW, Greene DA. Diminished specific activity of cytosolic protein kinase C in sciatic nerve of streptozocin-induced diabetic rats and its correction by dietary myo-inositol. Diabetes. 1991;40:1545–1554. doi: 10.2337/diab.40.11.1545. [DOI] [PubMed] [Google Scholar]
  • 111.Cameron NE, Cotter MA, Jack AM, Basso MD, Hohman TC. Protein kinase C effects on nerve function, perfusion, Na(+), K(+)-atpase activity and glutathione content in diabetic rats. Diabetologia. 1999;42:1120–1130. doi: 10.1007/s001250051280. [DOI] [PubMed] [Google Scholar]
  • 112.Yamagishi S, Uehara K, Otsuki S, Yagihashi S. Differential influence of increased polyol pathway on protein kinase C expressions between endoneurial and epineurial tissues in diabetic mice. J Neurochem. 2003;87:497–507. doi: 10.1046/j.1471-4159.2003.02011.x. [DOI] [PubMed] [Google Scholar]
  • 113.Lehning EJ, LoPachin RM, Mathew J, Eichberg J. Changes in Na-K Atpase and protein kinase c activities in peripheral nerve of acrylamide-treated rats. J Toxicol Environ Health. 1994;42:331–342. doi: 10.1080/15287399409531883. [DOI] [PubMed] [Google Scholar]
  • 114.Hermenegildo C, Felipo V, Minana MD, Grisolia S. Inhibition of protein kinase C restores Na+, K(+)-Atpase activity in sciatic nerve of diabetic mice. J Neurochem. 1992;58:1246–1249. doi: 10.1111/j.1471-4159.1992.tb11335.x. [DOI] [PubMed] [Google Scholar]
  • 115.Nakamura J, Kato K, Hamada Y, Nakayama M, Chaya S, Nakashima E, Naruse K, Kasuya Y, Mizubayashi R, Miwa K, Yasuda Y, Kamiya H, Ienaga K, Sakakibara F, Koh N, Hotta N. A protein kinase C-beta-selective inhibitor ameliorates neural dysfunction in streptozotocin-induced diabetic rats. Diabetes. 1999;48:2090–2095. doi: 10.2337/diabetes.48.10.2090. [DOI] [PubMed] [Google Scholar]
  • 116.Cameron NE, Cotter MA. Effects of protein kinase Cbeta inhibition on neurovascular dysfunction in diabetic rats: Interaction with oxidative stress and essential fatty acid dysmetabolism. Diabetes Metab Res Rev. 2002;18:315–323. doi: 10.1002/dmrr.307. [DOI] [PubMed] [Google Scholar]
  • 117.Steinberg HO, Brechtel G, Johnson A, Fineberg N, Baron AD. Insulin-mediated skeletal muscle vasodilation is nitric oxide dependent. A novel action of insulin to increase nitric oxide release. J Clin Invest. 1994;94:1172–1179. doi: 10.1172/JCI117433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Montagnani M, Chen H, Barr VA, Quon MJ. Insulin-stimulated activation of enos is independent of Ca2+ but requires phosphorylation by Akt at ser(1179) J Biol Chem. 2001;276:30392–30398. doi: 10.1074/jbc.M103702200. [DOI] [PubMed] [Google Scholar]
  • 119.Kuboki K, Jiang ZY, Takahara N, Ha SW, Igarashi M, Yamauchi T, Feener EP, Herbert TP, Rhodes CJ, King GL. Regulation of endothelial constitutive nitric oxide synthase gene expression in endothelial cells and in vivo: A specific vascular action of insulin. Circulation. 2000;101:676–681. doi: 10.1161/01.cir.101.6.676. [DOI] [PubMed] [Google Scholar]
  • 120.Rask-Madsen C, King GL. Proatherosclerotic mechanisms involving protein kinase C in diabetes and insulin resistance. Arterioscler Thromb VascBiol. 2005;25:487–496. doi: 10.1161/01.ATV.0000155325.41507.e0. [DOI] [PubMed] [Google Scholar]
  • 121.Jiang ZY, Lin YW, Clemont A, Feener EP, Hein KD, Igarashi M, Yamauchi T, White MF, King GL. Characterization of selective resistance to insulin signaling in the vasculature of obese zucker (fa/fa) rats. J Clin Invest. 1999;104:447–457. doi: 10.1172/JCI5971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Cusi K, Maezono K, Osman A, Pendergrass M, Patti ME, Pratipanawatr T, DeFronzo RA, Kahn CR, Mandarino LJ. Insulin resistance differentially affects the PI3-kinase- and MAP kinase-mediated signaling in human muscle. J Clin Invest. 2000;105:311–320. doi: 10.1172/JCI7535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.He Z, Opland DM, Way KJ, Ueki K, Bodyak N, Kang PM, Izumo S, Kulkarni RN, Wang B, Liao R, Kahn CR, King GL. Regulation of vascular endothelial growth factor expression and vascularization in the myocardium by insulin receptor and PI3K/Akt pathways in insulin resistance and ischemia. Arterioscler Thromb Vasc Biol. 2006;26:787–793. doi: 10.1161/01.ATV.0000209500.15801.4e. [DOI] [PubMed] [Google Scholar]
  • 124.Naruse K, Rask-Madsen C, Takahara N, Ha SW, Suzuma K, Way KJ, Jacobs JR, Clermont AC, Ueki K, Ohshiro Y, Zhang J, Goldfine AB, King GL. Activation of vascular protein kinase C-beta inhibits Akt-dependent endothelial nitric oxide synthase function in obesity-associated insulin resistance. Diabetes. 2006;55:691–698. doi: 10.2337/diabetes.55.03.06.db05-0771. [DOI] [PubMed] [Google Scholar]
  • 125.Sampson SR, Cooper DR. Specific protein kinase c isoforms as transducers and modulators of insulin signaling. Mol Genet Metab. 2006;89:32–47. doi: 10.1016/j.ymgme.2006.04.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Bakker W, Sipkema P, Stehouwer CD, Serne EH, Smulders YM, van Hinsbergh VW, Eringa EC. Protein kinase C theta activation induces insulin-mediated constriction of muscle resistance arteries. Diabetes. 2008;57:706–713. doi: 10.2337/db07-0792. [DOI] [PubMed] [Google Scholar]
  • 127.Rask-Madsen C, King GL. Mechanisms of disease: Endothelial dysfunction in insulin resistance and diabetes. Nat Clin Pract Endocrinol Metab. 2007;3:46–56. doi: 10.1038/ncpendmet0366. [DOI] [PubMed] [Google Scholar]
  • 128.Parmer TG, Ward MD, Hait WN. Effects of rottlerin, an inhibitor of calmodulin-dependent protein kinase III, on cellular proliferation, viability, and cell cycle distribution in malignant glioma cells. Cell Growth Differ. 1997;8:327–334. [PubMed] [Google Scholar]
  • 129.Soltoff SP. Rottlerin: An inappropriate and ineffective inhibitor of PKCdelta. Trends Pharmacol Sci. 2007;28:453–458. doi: 10.1016/j.tips.2007.07.003. [DOI] [PubMed] [Google Scholar]
  • 130.Jirousek MR, Gillig JR, Gonzalez CM, Heath WF, McDonald JH, 3rd, Neel DA, Rito CJ, Singh U, Stramm LE, Melikian-Badalian A, Baevsky M, Ballas LM, Hall SE, Winneroski LL, Faul MM. (S)-13-[(dimethylamino)methyl]-10,11,14,15-tetrahydro-4,9:16, 21-dimetheno-1H, 13H-dibenzo[e,k]pyrrolo[3,4-h][1,4,13]oxadiazacyclohexadecene-1,3(2H)-d ione ( LY333531) and related analogues: isozyme selective inhibitors of protein kinase C beta. J Med Chem. 1996;39:2664–2671. doi: 10.1021/jm950588y. [DOI] [PubMed] [Google Scholar]
  • 131.Aiello LP, Clermont A, Arora V, Davis MD, Sheetz MJ, Bursell SE. Inhibition of PKC beta by oral administration of ruboxistaurin is well tolerated and ameliorates diabetes-induced retinal hemodynamic abnormalities in patients. Invest Ophthalmol Visl Sci. 2006;47:86–92. doi: 10.1167/iovs.05-0757. [DOI] [PubMed] [Google Scholar]
  • 132.McGill JB, King GL, Berg PH, Price KL, Kles KA, Bastyr EJ, Hyslop DL. Clinical safety of the selective PKC-beta inhibitor, ruboxistaurin. Expert Opin Drug Saf. 2006;5:835–845. doi: 10.1517/14740338.5.6.835. [DOI] [PubMed] [Google Scholar]
  • 133.PKC-DRS Study Group. The effect of ruboxistaurin on visual loss in patients with moderately severeto very severe nonproliferative diabetic retinopathy: Initial results of the protein kinase C beta inhibitor diabetic retinopathy study (PKC-DRS) multicenter randomized clinical trial. Diabetes. 2005;54:2188–2197. doi: 10.2337/diabetes.54.7.2188. [DOI] [PubMed] [Google Scholar]
  • 134.PKC-DMES Study Group. Effect of ruboxistaurin in patients with diabetic macular edema: Thirty-month results of the randomized PKC-DMESclinical trial. Arch Ophthalmol. 2007;125:318–324. doi: 10.1001/archopht.125.3.318. [DOI] [PubMed] [Google Scholar]
  • 135.Aiello LP, Davis MD, Girach A, Kles KA, Milton RC, Sheetz MJ, Vignati L, Zhi XE. Effect of ruboxistaurin on visual loss in patients with diabetic retinopathy. Ophthalmology. 2006;113:2221–2230. doi: 10.1016/j.ophtha.2006.07.032. [DOI] [PubMed] [Google Scholar]
  • 136.Langham RG, Kelly DJ, Gow RM, Zhang Y, Cox AJ, Qi W, Thai K, Pollock CA, Christensen PK, Parving HH, Gilbert RE. Increased renal gene transcription of protein kinase C-beta in human diabetic nephropathy: Relationship to long-term glycaemic control. Diabetologia. 2008;51:668–674. doi: 10.1007/s00125-008-0927-x. [DOI] [PubMed] [Google Scholar]
  • 137.Tuttle KR, Bakris GL, Toto RD, McGill JB, Hu K, Anderson PW. The effect of ruboxistaurin on nephropathy in type 2 diabetes. Diabetes Care. 2005;28:2686–2690. doi: 10.2337/diacare.28.11.2686. [DOI] [PubMed] [Google Scholar]
  • 138.Vinik AI, Bril V, Kempler P, Litchy WJ, Tesfaye S, Price KL, Bastyr EJ., 3rd Treatment of symptomatic diabetic peripheral neuropathy with the protein kinase C beta-inhibitor ruboxistaurin mesylate during a 1-year, randomized, placebo-controlled, double-blind clinical trial. Clinical therapeutics. 2005;27:1164–1180. doi: 10.1016/j.clinthera.2005.08.001. [DOI] [PubMed] [Google Scholar]
  • 139.Calver A, Collier J, Vallance P. Inhibition and stimulation of nitric oxide synthesis in the human forearm arterial bed of patients with insulin-dependent diabetes. J Clin Invest. 1992;90:2548–2554. doi: 10.1172/JCI116149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.McVeigh GE, Brennan GM, Johnston GD, McDermott BJ, McGrath LT, Henry WR, Andrews JW, Hayes JR. Impaired endothelium-dependent and independent vasodilation in patients with type 2 (non-insulin-dependent) diabetes mellitus. Diabetologia. 1992;35:771–776. doi: 10.1007/BF00429099. [DOI] [PubMed] [Google Scholar]
  • 141.Steinberg HO, Chaker H, Leaming R, Johnson A, Brechtel G, Baron AD. Obesity/insulin resistance is associated with endothelial dysfunction. Implications for the syndrome of insulin resistance. J Clin Invest. 1996;97:2601–2610. doi: 10.1172/JCI118709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Williamson JR, Chang K, Tilton RG, Prater C, Jeffrey JR, Weigel C, Sherman WR, Eades DM, Kilo C. Increased vascular permeability in spontaneously diabetic bb/w rats and in rats with mild versus severe streptozocin-induced diabetes. Prevention by aldose reductase inhibitors and castration. Diabetes. 1987;36:813–821. doi: 10.2337/diab.36.7.813. [DOI] [PubMed] [Google Scholar]
  • 143.Beckman JA, Goldfine AB, Gordon MB, Garrett LA, Creager MA. Inhibition of protein kinase Cbeta prevents impaired endothelium-dependent vasodilation caused by hyperglycemia in humans. Circ Res. 2002;90:107–111. doi: 10.1161/hh0102.102359. [DOI] [PubMed] [Google Scholar]
  • 144.Mehta NN, Sheetz M, Price K, Comiskey L, Amrutia S, Iqbal N, Mohler ER, Reilly MP. Selective PKC beta inhibition with ruboxistaurin and endothelial function in type-2 diabetes mellitus. Cardiovasc Drugs and Ther. 2009;23:17–24. doi: 10.1007/s10557-008-6144-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Casellini CM, Barlow PM, Rice AL, Casey M, Simmons K, Pittenger G, Bastyr EJ, 3rd, Wolka AM, Vinik AI. A 6-month, randomized, double-masked, placebo-controlled study evaluating the effects of the protein kinase c-beta inhibitor ruboxistaurin on skin microvascular blood flow and other measures of diabetic peripheral neuropathy. Diabetes Care. 2007;30:896–902. doi: 10.2337/dc06-1699. [DOI] [PubMed] [Google Scholar]

RESOURCES