Вы находитесь на странице: 1из 10

Protein kinase C and the vascular complications of diabetes mellitus

Pgina 1 de 10

Official reprint from UpToDate


www.uptodate.com 2015 UpToDate

Protein kinase C and the vascular complications of diabetes mellitus


Author
Eli A Friedman, MD

Section Editors
Richard J Glassock, MD, MACP
David M Nathan, MD

Deputy Editor
John P Forman, MD, MSc

All topics are updated as new evidence becomes available and our peer review process is complete.
Literature review current through: May 2015. | This topic last updated: May 06, 2015.
INTRODUCTION Among patients who have had diabetes mellitus for five or more years, vascular
dysfunction results in nephropathy, retinopathy and at least some forms of neuropathy. The most likely cause of
vascular dysfunction is chronic hyperglycemia, although hyperglycemia alone may not be sufficient to cause
microvascular disease [1,2]. (See "Glycemic control and vascular complications in type 1 diabetes mellitus" and
"Glycemic control and vascular complications in type 2 diabetes mellitus".)
Numerous mechanisms have been proposed to explain the association between hyperglycemia and vascular
complications (algorithm 1) [3,4]. These include:
Increased activation of the polyol (or aldose reductase) pathway. Increased aldose reductase activity
results in the depletion of NADPH, a decrease in cellular reduced glutathione levels, and increased
oxidative stress [4].
Increased activity of the hexosamine pathway, which may alter gene expression and protein function [4].
Increased formation of advanced glycated end products (AGEs) [5], which may activate protein kinase C
(PKC) isoforms [6]. The mechanisms of oxidant-induced interaction between PKC isoforms and AGEs
resulting in vascular disease are incompletely defined [7].
Hemodynamic alterations (increased capillary pressures and flows).
Tissue hypoxia and oxidative stress.
Increased activity of vascular PKC isoform(s) [8].
This topic reviews the role of PKC in the pathogenesis of diabetic complications. The pathogenesis of diabetes
mellitus and the roles of specific proteins in insulin regulation and resistance are discussed elsewhere. (See
"Pathogenesis of type 1 diabetes mellitus" and "Pathogenesis of type 2 diabetes mellitus" and "Classification of
diabetes mellitus and genetic diabetic syndromes", section on 'Genetic defects'.)
STRUCTURE AND FUNCTION The reversible phosphorylation of proteins is a common means of governing
protein activity within cells. Phosphates are transferred from ATP molecules onto target molecules by protein
kinases and are removed by protein phosphatases (figure 1). In most, but not all, circumstances, the activated
kinase "turns on" the next target by the addition of the phosphate and is "turned off" with the removal of the
phosphate by a protein phosphatase. Within each cell, a large variety of kinases and phosphatases play a
central role in intracellular signaling.
Protein kinases belong to a large family of enzymes that contain a similar 250 amino acid catalytic (kinase)
domain that is responsible for the phosphate transfer to the target protein. They differ according to the amino
acids on either side of the kinase domain. These variable amino acids provide specificity and permit the
recognition and phosphorylation of unique sets of proteins by each protein kinase. Other amino acid sequences
within the kinase domain permit enzyme activity to be regulated (on or off) in response to specific signals.
Kinases recognize target phosphorylation sites by the surrounding amino acid sequences, which are common
to many proteins.

http://www.uptodate.com/contents/protein-kinase-c-and-the-vascular-complications-of-di... 16/06/2015

Protein kinase C and the vascular complications of diabetes mellitus

Pgina 2 de 10

ROLE OF PKC IN VASCULAR COMPLICATIONS Protein kinase C (PKC) activity is increased in both
experimental animals [9,10] and humans [11-13] with diabetes. The hyperglycemia-induced increase in PKC
activity is most likely mediated by enhanced de novo synthesis of diacylglycerol (DAG), which is a major
endogenous activator of PKC [14]. Diacylglycerol (DAG) levels are chronically elevated in the setting of
hyperglycemia because of the increased availability of the DAG precursor, glycerol 3-phosphate [3]. Reactive
oxygen species, free fatty acids, and growth factors may also contribute to PKC activation in diabetic patients
[15]. Multiple PKC isoforms appear to be activated by hyperglycemia [3].
The activation of PKC initiates a complex network of intracellular signaling that may alter transcription factor
binding to the promoter regions of responsive genes, which alters gene expression [16]. Increases in vascular
permeability, angiogenesis, cell growth and apoptosis, vessel dilation, cytokine activation, basement membrane
thickening and extracellular matrix expansion have all been observed with increased PKC activity [3,17].
Sustained alterations in PKC-regulated gene expression in vascular cells may contribute to some of the
characteristic vascular abnormalities in diabetes mellitus.
Diabetic nephropathy Experiments in diabetic mice and rats support a role for PKC in the pathogenesis of
diabetic nephropathy:
PKC activity is increased in glomeruli isolated from diabetic rats [18] and in mesangial cells cultured in
high glucose [10].
PKC inhibition prevents the glycated albumin-induced or glucose-induced increase in basement
membrane type IV collagen production by glomerular endothelial [19] and mesangial [20] cells.
PKC increases levels of mRNA that encode matrix components in glomeruli isolated from streptozotocin
(STZ)-induced diabetic rats [21].
PKCdelta promotes tubular cell injury and death in kidneys of STZ-induced diabetic mice [22].
PKC activation has been associated with alterations in renal and glomerular blood flow, basement
membrane thickening, extracellular matrix expansion, increased vascular permeability, enhanced
angiogenesis, excessive apoptosis, and alterations in enzyme activation of Na(+)-K(+)-ATPase, cPLA(2),
PI3Kinase, and MAP kinase in diabetic rats [23].
PKC inhibitors decrease hyperfiltration and proteinuria in animal models. (See 'Inhibitors of PKC' below.)
Multiple PKC isoform(s) contribute to the pathogenesis of diabetic nephropathy. Experiments with isoformspecific knockout mice suggest that activation of PKCalpha underlies the development of albuminuria and that
of PKCbeta contributes to glomerular hypertrophy and mesangial expansion [15].
Both isoforms appear to contribute to reactive oxygen species generation, which plays a substantive pathogenic
role in the progression of diabetic nephropathy [15,24-26].
Role of PKC in extrarenal complications Hyperglycemia-induced increases in PKC activity have been
observed in nonrenal tissue including retina, endothelial, smooth muscle cells, monocytes and macrophages
and cardiac tissue [3,27-30].
Animal studies have suggested that increased PKC activity contributes to atherosclerosis, cardiomyopathy,
diabetic retinopathy and diabetic neuropathy [3].
Hyperglycemia-induced PKC activation appears to contribute to endothelial dysfunction by increasing
expression of vascular endothelium growth factor (VEGF), thromboxane and endothelin, and decreasing levels
of nitric oxide (NO) and prostacyclin resulting in increases in vasoconstriction and permeability [3,31,32].
Extracellular matrix synthesis, fibrosis, dysregulation of cytokines and monocyte activation and impaired insulinstimulated glucose transport in muscle may all result from PKC activation [3,33].

http://www.uptodate.com/contents/protein-kinase-c-and-the-vascular-complications-of-di... 16/06/2015

Protein kinase C and the vascular complications of diabetes mellitus

Pgina 3 de 10

The identification of compounds that inhibit the PKC signaling pathway and block expression of PKC-regulated
genes may lead to novel therapeutic approaches to prevent and/or treat diabetic complications [34].
GENETICS Mutations that alter the expression or activity of protein kinases may underlie both abnormalities
in glucose metabolism and the predisposition to vascular complications of diabetes [35-39]. (See "Pathogenesis
of type 2 diabetes mellitus".)
Mutations that increase the activity of protein kinase C (PKC) isoforms may enhance susceptibility to diabetic
nephropathy in humans. This was suggested by the following two studies that showed an association of
PKCbeta1 single-nucleotide polymorphisms (SNPs) and diabetic nephropathy:
Nine PKCbeta1 SNPs were identified among patients who had type 1 diabetes and diabetic nephropathy
[40]. The frequency of each SNP was compared among case patients (defined as patients with type 1
diabetes and persistent proteinuria or end-stage renal disease (ESRD) as a result of diabetic
nephropathy) and control subjects (defined as patients with type diabetes for >15 years and who have an
albumin-to-creatinine ratio <17 mg/g for men, and <25 mg/g for women on spot urine specimens). The
frequency of two SNPs in the promoter region was greater among case patients compared with control
patients. The association was stronger among those who had a duration of diabetes less than 24 years.
Eighteen common SNPs were genotyped in 1172 Chinese patients without renal disease who were
recruited from 1995-1998 and compared with 1049 patients with early onset type 2 diabetes without renal
disease who were recruited after 1998 [41]. After a mean of 7.9 years, 90 patients (7.7 percent)
progressed to ESRD. ESRD was associated with four common SNPs.
INHIBITORS OF PKC Therapies aimed at lowering protein kinase C (PKC) may be beneficial in slowing the
progression of diabetic nephropathy, as shown in animal studies; however, studies in humans to date are rather
inconclusive in this regard [42,43]. The most widely studied isoform-specific PKC inhibitor is ruboxistaurin
mesylate, which is highly specific for PKCbeta isoforms [44]. Ruboxistaurin has been examined in animal
models of diabetes mellitus and a few pilot studies in humans [45].
Animal studies of ruboxistaurin The effects of ruboxistaurin on diabetic neuropathy, vascular permeability
and renal function have been studied in multiple animal models:
Two weeks of treatment with ruboxistaurin completely reversed the diabetes-induced reduction in sciatic
motor and saphenous sensory nerve conduction velocity (NCV) in streptozotocin (STZ)-induced diabetic
rats [46]. When combined with the antioxidants vitamin E or alpha-lipoic acid, motor and sensory NCV and
sciatic nerve perfusion improved to the nondiabetic range, demonstrating synergism between PKCbeta
and blockers of oxidative stress.
The topical application of ruboxistaurin to newly grown vessels in granulation tissue blocked glucoseinduced increases in both blood flow and permeability (figure 2) [47]. Oral administration of ruboxistaurin
for eight weeks prevented diabetes-induced increases in albumin permeation in the retina, nerve, and
aorta, but not in muscle or brain of diabetic rats, an effect possibly mediated by inhibition of transcription
factor binding to specific PKC-regulated genes that are involved in vascular function.
The oral administration of ruboxistaurin was associated with reversal of glomerular hyperfiltration (from 4.6
mL/min to the normal range of 3.0 0.2 mL/min) and reduction of urinary albumin excretion (11.7 to 4.9
mg/day [normal 1.6 mg/day]) in diabetic rats [48]. This occurred without a change in glycated hemoglobin
or DAG content in the retina or glomeruli.
Ruboxistaurin normalized glomerular hyperfiltration, decreased urinary albumin excretion, and reduced
glomerular transforming growth factor (TGF)-beta1 and extracellular matrix protein production in the STZ
rat, Lepr(db)/Lepr(db) mouse, and STZ-Ren-2 rat models of diabetes [49]. Improvements were also noted
in mesangial expansion, glomerulosclerosis, tubulointerstitial fibrosis, and renal function.

http://www.uptodate.com/contents/protein-kinase-c-and-the-vascular-complications-of-di... 16/06/2015

Protein kinase C and the vascular complications of diabetes mellitus

Pgina 4 de 10

Ruboxistaurin significantly attenuated increases in PKC-induced osteopontin expression in tubular


epithelial cells of the renal cortex, macrophage recruitment and tubulointerstitial injury associated with
activity of TGF-beta in the Ren-2 diabetic rats [50].
Human studies of ruboxistaurin
Orally administered ruboxistaurin did not prevent the progression of retinopathy in initial clinical trials of
moderately severe to very severe nonproliferative diabetic retinopathy [51]. (See "Diabetic retinopathy:
Prevention and treatment", section on 'Nonproliferative diabetic retinopathy'.)
Ruboxistaurin stabilized the progression of nephropathy in 123 patients with type 2 diabetes and early
diabetic nephropathy (albumin-to-creatinine ratios of 200 to 2000 mg/gm and serum creatinine 1.7 mg/dL
[150 micromol/L] in women, and 2.0 mg/dL [177 micromol/L] in men) who were on stable doses of
angiotensin-converting enzyme (ACE) inhibitors or angiotensin II receptor blockers (ARBs) [52]. Patients
were randomly assigned to receive ruboxistaurin, 32 mg/day, or placebo. After one year, treatment with
ruboxistaurin was associated with a reduction in albuminuria and stabilization of estimated glomerular
filtration rate (eGFR), with no change in albuminuria and worsening of eGFR in the placebo group. There
was no difference between groups in mean blood pressure at baseline and at the end of the study.
The effect of ruboxistaurin on long-term (greater than one year) renal outcomes was evaluated in a
secondary analysis of three diabetic retinopathy trials [53]. During median follow-up at 33 to 39 months, no
differences in kidney outcomes were observed between ruboxistaurin and placebo. The result of a
prospective study of the effect of ruboxistaurin versus placebo in 9 diabetic and 11 control patients was
also disappointing; no difference was found between cohorts in peripheral nerve conduction velocity or
symptoms of sensory neuropathy [54]. Adding to disappointment in the use of ruboxistaurin in diabetes is
a randomized, 14-day double-blind, crossover, placebo-controlled trial of ruboxistaurin mesylate in 13
subjects with type 2 diabetes without evidence of cardiovascular disease and 15 healthy control subjects.
No significant change in endothelium-dependent or endothelium-independent vasodilation or blood-based
markers of inflammation, fibrinolysis, endothelial damage, and oxidative stress was noted in either diabetic
or healthy subjects [55].
Much larger trials are required before determining the potential clinical role of this agent [56]. Ruboxistaurin is in
phase 2 clinical trials in diabetic retinopathy and cardiac ventricular hypertrophy cardiomyopathy.
Other PKC inhibitors The treatment of diabetic rats with d-alpha-tocopherol, which decreases DAG and
inhibits PKC activation, prevents glomerular hyperfiltration and minimizes the development of proteinuria (2.4
versus 9.1 mg/day in control diabetic rats [versus 1.2 mg/day in nondiabetic rats]) [42].
Elevated cytosolic glucose promotes the accumulation of glyceraldehyde 3-phosphate, which activates PKC [4].
Large doses of thiamine and the thiamine monophosphate derivative, benfotiamine decreased the formation of
glyceraldehyde 3-phosphate and mitigated PKC activation in an experimental model of diabetes [57]. This was
associated with a decrease in the development of moderately increased albuminuria (the new term for what
was previously called "microalbuminuria" [58]) and prevention of hyperfiltration, without a change in elevated
plasma glucose concentration and glycated hemoglobin.
SUMMARY
Increased activity of protein kinase C (PKC) appears to contribute to the micro- and macrovascular
complications of diabetes mellitus. Mechanisms by which this occurs likely include PKC-induced changes
in vascular permeability, angiogenesis, cell growth and apoptosis, vessel dilation, cytokine activation,
basement membrane thickening and extracellular matrix expansion. (See 'Introduction' above and 'Role of
PKC in vascular complications' above.)
Multiple PKC isoforms appear to be involved in the development of diabetic nephropathy. Studies of
isoform-specific knockout mice suggest that PKCalpha contributes to albuminuria and PKCbeta to

http://www.uptodate.com/contents/protein-kinase-c-and-the-vascular-complications-of-di... 16/06/2015

Protein kinase C and the vascular complications of diabetes mellitus

Pgina 5 de 10

glomerular hypertrophy and mesangial expansion. Both isoforms may play a role in the generation of
reactive oxygen species. (See 'Diabetic nephropathy' above.)
Increased PKC activity may also underlie the development of atherosclerosis, cardiomyopathy, diabetic
retinopathy and diabetic neuropathy. (See 'Role of PKC in extrarenal complications' above.)
Mutations that alter expression or activity of PKC may enhance susceptibility to diabetic nephropathy in
humans. Single-nucleotide polymorphisms (SNPs) of the PKCbeta1 gene have been associated with
proteinuria and with end-stage renal disease (ESRD). (See 'Genetics' above.)
The clinical benefit of therapies aimed at lowering PKC activity in preventing or delaying the complications
of diabetes mellitus is not clear at this time. (See 'Inhibitors of PKC' above.)
Use of UpToDate is subject to the Subscription and License Agreement.
REFERENCES
1. 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.
2. 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.
3. Geraldes P, King GL. Activation of protein kinase C isoforms and its impact on diabetic complications.
Circ Res 2010; 106:1319.
4. Brownlee M. Biochemistry and molecular cell biology of diabetic complications. Nature 2001; 414:813.
5. Garay-Sevilla ME, Regalado JC, Malacara JM, et al. Advanced glycosylation end products in skin, serum,
saliva and urine and its association with complications of patients with type 2 diabetes mellitus. J
Endocrinol Invest 2005; 28:223.
6. He Z, King GL. Protein kinase Cbeta isoform inhibitors: a new treatment for diabetic cardiovascular
diseases. Circulation 2004; 110:7.
7. Cai W, Torreggiani M, Zhu L, et al. AGER1 regulates endothelial cell NADPH oxidase-dependent oxidant
stress via PKC-delta: implications for vascular disease. Am J Physiol Cell Physiol 2010; 298:C624.
8. Craven PA, Studer RK, Negrete H, DeRubertis FR. Protein kinase C in diabetic nephropathy. J Diabetes
Complications 1995; 9:241.
9. Koya D, Jirousek MR, Lin YW, et al. 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.
10. Craven PA, DeRubertis FR. Protein kinase C is activated in glomeruli from streptozotocin diabetic rats.
Possible mediation by glucose. J Clin Invest 1989; 83:1667.
11. Koya D, King GL. Protein kinase C activation and the development of diabetic complications. Diabetes
1998; 47:859.
12. Ways DK, Sheetz MJ. The role of protein kinase C in the development of the complications of diabetes.
Vitam Horm 2000; 60:149.
13. Schena FP, Gesualdo L. Pathogenetic mechanisms of diabetic nephropathy. J Am Soc Nephrol 2005; 16
Suppl 1:S30.
14. Inoguchi T, Battan R, Handler E, et al. 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.
15. Menne J, Meier M, Park JK, Haller H. Inhibition of protein kinase C in diabetic nephropathy--where do we
stand? Nephrol Dial Transplant 2009; 24:2021.

http://www.uptodate.com/contents/protein-kinase-c-and-the-vascular-complications-of-di... 16/06/2015

Protein kinase C and the vascular complications of diabetes mellitus

Pgina 6 de 10

16. Karin M. The AP-1 complex and its role in transcriptional control by protein kinase C. In: Molecular
Aspects of Cellular Regulation, Cohen P, Foulkes JG (Eds) (Eds), Elsevier, New York 1991. p.235.
17. Noh H, King GL. The role of protein kinase C activation in diabetic nephropathy. Kidney Int Suppl 2007;
:S49.
18. Haneda M, Kikkawa R, Sugimoto T, et al. Abnormalities in protein kinase C and MAP kinase cascade in
mesangial cells cultured under high glucose conditions. J Diabetes Complications 1995; 9:246.
19. Chen S, Cohen MP, Lautenslager GT, et al. Glycated albumin stimulates TGF-beta 1 production and
protein kinase C activity in glomerular endothelial cells. Kidney Int 2001; 59:673.
20. 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.
21. Ziyadeh FN, Fumo P, Rodenberger CH, et al. Role of protein kinase C and cyclic AMP/protein kinase A in
high glucose-stimulated transcriptional activation of collagen alpha 1 (IV) in glomerular mesangial cells. J
Diabetes Complications 1995; 9:255.
22. Li X, Pabla N, Wei Q, et al. PKC-delta promotes renal tubular cell apoptosis associated with proteinuria. J
Am Soc Nephrol 2010; 21:1115.
23. Das Evcimen N, King GL. The role of protein kinase C activation and the vascular complications of
diabetes. Pharmacol Res 2007; 55:498.
24. Swaminathan S, Shah SV. Novel approaches targeted toward oxidative stress for the treatment of chronic
kidney disease. Curr Opin Nephrol Hypertens 2008; 17:143.
25. Kanwar YS, Wada J, Sun L, et al. Diabetic nephropathy: mechanisms of renal disease progression. Exp
Biol Med (Maywood) 2008; 233:4.
26. Wolf G. New insights into the pathophysiology of diabetic nephropathy: from haemodynamics to
molecular pathology. Eur J Clin Invest 2004; 34:785.
27. Kunisaki M, Bursell SE, Clermont AC, et al. Vitamin E prevents diabetes-induced abnormal retinal blood
flow via the diacylglycerol-protein kinase C pathway. Am J Physiol 1995; 269:E239.
28. Hattori Y, Kawasaki H, Fukao M, Kanno M. Phorbol esters elicit Ca(2+)-dependent delayed contractions
in diabetic rat aorta. Eur J Pharmacol 1995; 279:51.
29. Haneda M, Koya D, Kikkawa R. Cellular mechanisms in the development and progression of diabetic
nephropathy: activation of the DAG-PKC-ERK pathway. Am J Kidney Dis 2001; 38:S178.
30. Srivastava AK. High glucose-induced activation of protein kinase signaling pathways in vascular smooth
muscle cells: a potential role in the pathogenesis of vascular dysfunction in diabetes (review). Int J Mol
Med 2002; 9:85.
31. Shiba T, Inoguchi T, Sportsman JR, et al. Correlation of diacylglycerol level and protein kinase C activity
in rat retina to retinal circulation. Am J Physiol 1993; 265:E783.
32. Wolf BA, Williamson JR, Easom RA, et al. Diacylglycerol accumulation and microvascular abnormalities
induced by elevated glucose levels. J Clin Invest 1991; 87:31.
33. Temofonte N, Sajan MP, Nimal S, et al. Combined thiazolidinedione-metformin treatment synergistically
improves insulin signalling to insulin receptor substrate-1-dependent phosphatidylinositol 3-kinase,
atypical protein kinase C and protein kinase B/Akt in human diabetic muscle. Diabetologia 2009; 52:60.
34. Kuo DY, Yang SF, Chu SC, et al. Amphetamine-evoked changes of oxidative stress and neuropeptide Y
gene expression in hypothalamus: regulation by the protein kinase C-delta signaling. Chem Biol Interact
2009; 180:193.
35. O'Rahilly S, Barroso I, Wareham NJ. Genetic factors in type 2 diabetes: the end of the beginning?
Science 2005; 307:370.
36. Rangwala SM, Lazar MA. Peroxisome proliferator-activated receptor gamma in diabetes and metabolism.
Trends Pharmacol Sci 2004; 25:331.
37. Wilkin TJ. Diabetes: 1 and 2, or one and the same? Progress with the accelerator hypothesis. Pediatr
Diabetes 2008; 9:23.
38. Perry JR, Frayling TM. New gene variants alter type 2 diabetes risk predominantly through reduced betacell function. Curr Opin Clin Nutr Metab Care 2008; 11:371.

http://www.uptodate.com/contents/protein-kinase-c-and-the-vascular-complications-of-di... 16/06/2015

Protein kinase C and the vascular complications of diabetes mellitus

Pgina 7 de 10

39. Pleyer L, Egle A, Hartmann TN, Greil R. Molecular and cellular mechanisms of CLL: novel therapeutic
approaches. Nat Rev Clin Oncol 2009; 6:405.
40. Araki S, Ng DP, Krolewski B, et al. Identification of a common risk haplotype for diabetic nephropathy at
the protein kinase C-beta1 (PRKCB1) gene locus. J Am Soc Nephrol 2003; 14:2015.
41. Ma RC, Tam CH, Wang Y, et al. Genetic variants of the protein kinase C-beta 1 gene and development of
end-stage renal disease in patients with type 2 diabetes. JAMA 2010; 304:881.
42. Koya D, Lee IK, Ishii H, et al. Prevention of glomerular dysfunction in diabetic rats by treatment with
d-alpha-tocopherol. J Am Soc Nephrol 1997; 8:426.
43. Williams ME, Tuttle KR. The next generation of diabetic nephropathy therapies: an update. Adv Chronic
Kidney Dis 2005; 12:212.
44. Gassel M, Breitenlechner CB, Knig N, et al. The protein kinase C inhibitor bisindolyl maleimide 2 binds
with reversed orientations to different conformations of protein kinase A. J Biol Chem 2004; 279:23679.
45. Glvez MI. Rubosixtaurin and other PKC inhibitors in diabetic retinopathy and macular edema. Review.
Curr Diabetes Rev 2009; 5:14.
46. 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.
47. Birch KA, Heath WF, Hermeling RN, et al. LY290181, an inhibitor of diabetes-induced vascular
dysfunction, blocks protein kinase C-stimulated transcriptional activation through inhibition of transcription
factor binding to a phorbol response element. Diabetes 1996; 45:642.
48. Ishii H, Jirousek MR, Koya D, et al. Amelioration of vascular dysfunctions in diabetic rats by an oral PKC
beta inhibitor. Science 1996; 272:728.
49. Tuttle KR, Anderson PW. A novel potential therapy for diabetic nephropathy and vascular complications:
protein kinase C beta inhibition. Am J Kidney Dis 2003; 42:456.
50. Kelly DJ, Chanty A, Gow RM, et al. Protein kinase Cbeta inhibition attenuates osteopontin expression,
macrophage recruitment, and tubulointerstitial injury in advanced experimental diabetic nephropathy. J
Am Soc Nephrol 2005; 16:1654.
51. PKC-DRS Study Group. The effect of ruboxistaurin on visual loss in patients with moderately severe to
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.
52. Tuttle KR, Bakris GL, Toto RD, et al. The effect of ruboxistaurin on nephropathy in type 2 diabetes.
Diabetes Care 2005; 28:2686.
53. Tuttle KR, McGill JB, Haney DJ, et al. Kidney outcomes in long-term studies of ruboxistaurin for diabetic
eye disease. Clin J Am Soc Nephrol 2007; 2:631.
54. Brooks B, Delaney-Robinson C, Molyneaux L, Yue DK. Endothelial and neural regulation of skin
microvascular blood flow in patients with diabetic peripheral neuropathy: effect of treatment with the
isoform-specific protein kinase C beta inhibitor, ruboxistaurin. J Diabetes Complications 2008; 22:88.
55. Beckman JA, Goldfine AB, Goldin A, et al. Inhibition of protein kinase Cbeta does not improve endothelial
function in type 2 diabetes. J Clin Endocrinol Metab 2010; 95:3783.
56. Mehta NN, Sheetz M, Price K, et al. Selective PKC beta inhibition with ruboxistaurin and endothelial
function in type-2 diabetes mellitus. Cardiovasc Drugs Ther 2009; 23:17.
57. Babaei-Jadidi R, Karachalias N, Ahmed N, et al. Prevention of incipient diabetic nephropathy by highdose thiamine and benfotiamine. Diabetes 2003; 52:2110.
58. KDIGO. Chapter 1: Definition and classification of CKD. Kidney Int Suppl 2013; 3:19.
http://www.kdigo.org/clinical_practice_guidelines/pdf/CKD/KDIGO_2012_CKD_GL.pdf (Accessed on
March 04, 2013).
Topic 3079 Version 9.0

http://www.uptodate.com/contents/protein-kinase-c-and-the-vascular-complications-of-di... 16/06/2015

Protein kinase C and the vascular complications of diabetes mellitus

Pgina 8 de 10

GRAPHICS
Mechanisms of hyperglycemia-induced vascular
complications

Flow diagram showing how too much glucose may lead to the long-term
complications of diabetes.
AGE: advanced glycosylation endproducts; DAG: diacylglycerol; PKC: protein
kinase C.
Graphic 61356 Version 2.0

http://www.uptodate.com/contents/protein-kinase-c-and-the-vascular-complications-of-di... 16/06/2015

Protein kinase C and the vascular complications of diabetes mellitus

Pgina 9 de 10

Action of protein kinases

Schematic representation of the action of protein kinases. Phosphates


are transferred from ATP molecules onto target molecules by protein
kinases; these phosphates are then removed by protein phosphatases.
Graphic 78694 Version 1.0

http://www.uptodate.com/contents/protein-kinase-c-and-the-vascular-complications-of-di... 16/06/2015

Protein kinase C and the vascular complications of diabetes mellitus

Pgina 10 de 10

Protein kinase C inhibition protects against


hyperglycemia-induced vascular dysfunction

Effect of the PKC inhibitor LY290181 on vascular dysfunction in skin


chamber granulation tissue induced by elevated glucose levels in
nondiabetic rats. Skin chamber granulation tissue was treated with
either 5 or 30 mmol/L of glucose alone or 30 mmol/L of glucose plus 20
to 50 mol/L of LY290181 twice daily for seven days. Exposure to the
high glucose solution increased both blood flow and vascular
permeability; these effects were prevented by LY290181.
Graphic 73671 Version 1.0

Disclosures
Disclosures: Eli A Friedman, MD Nothing to disclose. Richard J Glassock, MD, MACP Consultant/Advisory Boards: Bristol-MyersSquibb [lupus nephritis (Abatacept)]; Abbvie [lupus nephritis (no product)]; Sanofi-Genzyme [FSGS (Fresolimumab)]; Chemocentryx
[diabetes (CCR2 antagonist)]; QuestCor [nephrotic syndrome (ACTH Gel)]; Eli Lily [kidney disease (no product)]; Astellas [kidney
disease (Tacrolimus)]. Speaker's Bureau: Genentech [vasculitis (Rituximab)]. Other Financial Interest: Karger [editor]; AAKP [patients
with kidney disease]; American Renal Associates [dialysis]. Equity Ownership/Stock Options: Reata Stock [diabetes]. David M Nathan,
MD Nothing to disclose. John P Forman, MD, MSc Nothing to disclose.
Contributor disclosures are reviewed for conflicts of interest by the editorial group. When found, these are addressed by vetting through a
multi-level review process, and through requirements for references to be provided to support the content. Appropriately referenced
content is required of all authors and must conform to UpToDate standards of evidence.
Conflict of interest policy

http://www.uptodate.com/contents/protein-kinase-c-and-the-vascular-complications-of-di... 16/06/2015

Вам также может понравиться