Abstract
The results of recent outcome trials challenge hypotheses that tight control of both glycohemoglobin and blood pressure diminishes macrovascular events and survival among type 2 diabetic patients. Relevant questions exist regarding the adequacy of glycohemoglobin alone as a measure of diabetes control. Are we ignoring mechanisms of vasculotoxicity (profibrosis, altered angiogenesis, hypertrophy, hyperplasia, and endothelial injury) inherent in current antihyperglycemic medications? Is the polypharmacy for lowering cholesterol, triglyceride, glucose, and systolic blood pressure producing drug interactions that are too complex to be clinically identified? We review angiotensin–aldosterone mechanisms of tissue injury that magnify microvascular damage caused by hyperglycemia and hypertension. Many studies describe interruption of these mechanisms, without hemodynamic consequence, in the preservation of function in type 1 diabetes. Possible interactions between the renin–angiotensin–aldosterone system and physiologic glycemic control (through pulsatile insulin release) suggest opportunities for further clinical investigation.
Keywords: angiotensin-converting enzyme inhibitor, pulsatile insulin, diabetic nephropathy, cardiac autonomic neuropathy, podocytes, beta cells
Introduction
The Diabetes Control and Complications Trial (DCCT)1 established that multiple injections of insulin reduce microvascular complications in type 1 diabetes (Table 1). In the Captopril Study, angiotensin-converting enzyme (ACE) inhibition was demonstrated to preserve renal function in type 1 diabetes.2 Microvascular benefits from intensive glycemic management and ACE inhibition in type 2 diabetic patients have been reported in the UK Prospective Diabetes Study.3,4 Until recently, aggressive control of glycemia and blood pressure in type 2 diabetic patients was felt to be effective in the reduction of cardiovascular endpoints. Now that macrovascular endpoints have been found to be unresponsive to the highest doses of medications to lower glucose5 and blood pressure,6 we must consider alternative research involving the preservation of renal function as an indirect way of preserving myocardial function.
Table 1.
A. Diabetes control and complications trial |
Multiple insulin injections were found to be associated with a lower incidence of diabetic nephropathy, retinopathy, and neuropathy |
B. Captopril trial |
Angiotensin-converting enzyme inhibitor was found to decrease the rate of loss of renal function, using the time to doubling of serum creatinine |
C. Pulsatile insulin study14 |
Control of high and low blood glucose in type 1 diabetic patients was found to be improved beyond use of the DCCT protocol 7 days/week by use of the DCCT protocol 6 days/week + intravenous infusions of insulin in pulses 1 day/week. Each patient was his/her own control. This study did not address long-term complications |
D. Orthostatic hypotension15 |
Improved lifestyle with stabilization of locomotion through diminution of this neurological complication when pulsatile insulin was added 1 day/week to the DCCT protocol 6 days/week. Each patient was his/her own control |
E. Hypertension16 |
Lower doses of antihypertensives required during 3-month rotations of the pulsatile infusion 1 day/week were added to the DCCT protocol 6 days/week |
Abbreviation: DCCT, Diabetes Control and Complications Trial.
Mechanisms by which kidney glomerular, interstitial, and vascular anatomy are injured include hypertension, inflammation, enhanced hemostasis, oxidative stress, diminished endothelial function, pathological angiogenesis, and accelerated fibrosis. High glucose and increased angiotensin have been shown to have additive pathological effects on renal tubules with accelerated fibrosis of the interstitium through enhanced expression of transforming growth factor (TGF)-β.7,8 Positive endpoint responses that cannot be explained by hemodynamic variations alone have been demonstrated.
The interruption of angiotensin–aldosterone mechanisms and optimal insulinization need further study. We call for prospective investigation of nonhemodynamic angiotensin effects that may alter intracellular insulin signaling with effects on renal and myocardial physiology.
Clinical trials
Losartan or enalapril prevented the onset of retinopathy by photography, but did not alter the onset of nephropathy by histology in type 1 diabetic patients.9 In type 2 diabetic patients, an improvement in albuminuria with angiotensin receptor blockade was associated with a reduction in markers for acceleration of inflammation and thrombosis, suggesting effects beyond blood pressure reduction.10 In type 1 diabetic patients, pulsatile use of insulin magnified the benefits of ACE inhibition.
Improved cardiac autonomic function and reversal of left ventricular hypertrophy were associated with improved glycemia in type 1 diabetic nephropathy patients. Renal and pancreatic transplantation has also been associated with reversal of left ventricular hypertrophy.
Although most studies demonstrate the prevention of a new appearance of retinopathy in type 1 diabetic patients treated with angiotensin-active medications, there is major inconsistency in the findings of protection from retinopathy progression for both type 1 and type 2 diabetic patients treated with angiotensin-active medications.
Kidney studies
In the Renin–Angiotensin–Aldosterone System (RAAS) Study, normotensive, normoalbuminuric type 1 diabetic patients with minimal evidence of retinopathy were observed for 5 years. A renal biopsy was done at baseline and repeated at 5 years. When compared with placebo, neither enalapril nor losartan had an effect on renal biopsy histology despite the fact that blood pressures were significantly lower than with placebo.9
The Irbesartan in Patients with Type 2 Diabetes and Microalbuminuria (IRMA2) Study (Table 2) found improvement in albuminuria associated with reduction in markers for inflammation, including C-reactive protein, interleukin-6 (IL-6), and fibrinogen.10 Patients were maintained on their usual diabetes treatment, which was associated with no significant decrease in advanced glycation end products (AGE) concentration. Improvement of albuminuria without change in blood pressure over 52 weeks was found in diabetic11 and nondiabetic12 kidney disease patients when spironolactone was added to a regimen of ACE inhibitor or ACE inhibitor plus angiotensin receptor blocker (ARB)12 as shown in Table 2. The latter study was associated with diminution of type 4 collagen, a marker of fibrosis. Improvement in AGE and glycohemoglobin A1c was associated with a reduction in elevated levels of fibrinogen and factor VII13 acute-phase reactants with hemostasis effects.
Table 2.
References | Drug/patient | Primary endpoint | Comment |
---|---|---|---|
Persson et al10 | Irbesartan Type 2 diabetes |
Inflammatory markers decreased a. IL-6 b. C-reactive protein c. Fibrinogen |
Albuminuria decreased |
Flammer et al44 | Losartan versus atenolol Type 2 diabetes and hypertension |
a. Flow-mediated vasodilation decreased b. 8-Isoprostane decreased with losartan, but not atenolol |
8-Isoprostane is generated from membrane phospholipid by free radicals |
Kramer et al45 | Losartan Hypertension, no diabetes |
a. Platelet aggregation decreased after 8 h b. Endothelial cells (human)AT2-induced PDGF2-α + thromboxane blocked by incubation with EXP3179 |
Losartan metabolites: EXP3174 uses AT2 receptor EXP3179 no receptor Metabolism of losartan requires 8 h |
Fortuno et al46 | EXP3179 or losartan (but not EXP3174), irbesartan or quinapril, no diabetes | Human phagocytic mononuclear cells: a. NADPH oxidase b. Protein kinase C expression inhibited |
Expression of matrix metalloproteinase inhibited |
Furumatsu et al11 | Enalapril, losartan, and spironolactone, no diabetes | Albuminuria decreased over 1 year | Urine type 4 collagen decreased |
Mehdi et al12 | Lisinopril, spironolactone diabetes a. Type 2: 80% b. Type 1: 20% |
Albuminuria decreased over 1 year |
Table 1 summarizes the added benefit found with pulsatile insulin infusions for the stabilization of high and low blood glucose,14 postural hypotension,15 and supine hypertension16 in type 1 diabetic patients with emerging complications. Crossover studies demonstrated lower blood pressure medication requirements during pulsatile intravenous insulin infusions.16
A prospective randomized study of weekly pulsatile intravenous insulin infusions in type 1 diabetic patients with proteinuria and hypertension, including a control group on the multiple insulin injection (DCCT) protocol, was completed in several centers (Table 3). After 18 months, a statistically significant difference in progression of renal dysfunction was noted between the groups.17 For the pulsatile insulin group, it would take 15 years for the creatinine clearance (Ccreat) to fall from 50–60 mL/min to 10–15 mL/min compared with 5 years utilizing the DCCT protocol without weekly pulsatile infusions. This outcome could be expected to result in quality of life benefits in the trade-off of 1 day/week spent with insulin infusions versus multiple days per week with peritoneal or hemodialysis. The mechanism for added benefit of pulsatile infusion could not be shown to involve any of the hemostatic, echocardiographic, ambulatory blood pressure, or ambulatory electrocardiographic measures that were incorporated into the protocol.18
Table 3.
A. Randomized trial of DCCT protocol (control) versus DCCT protocol 6 days/week + pulsatile insulin (infusion group) 1 day/week in type 1 diabetic patients with proteinuria, effect on progression of loss of renal function as measured by Ccreat
B. ACE inhibitors preferred for blood pressure control
C. Blood pressures (mmHg) by 24-h ambulatory method not significantly different at baseline, 52 weeks, and 78 weeks | |||
Baseline | Infusion group (n = 37) | Control group (n = 34) | P value |
Systolic | 133.6 ± 3.2 | 132.5 ± 2.6 | 0.79 |
Diastolic | 77.8 ± 1.5 | 79.6 ± 1.7 | 0.44 |
52 weeks | Infusion group (n = 37) | Control group (n = 34) | P value |
Systolic | 136.0 ± 2.7 | 133.2 ± 2.6 | 0.46 |
Diastolic | 76.9 ± 1.8 | 78.7 ± 1.9 | 0.50 |
Baseline | Infusion group (n = 23) | Control group (n = 26) | P value |
Systolic | 134.8 ± 4.7 | 134.5 ± 3.1 | 0.96 |
Diastolic | 78.3 ± 1.8 | 80.4 ± 2.1 | 0.46 |
78 weeks | Infusion group (n = 23) | Control group (n = 26) | P value |
Systolic | 131.6 ± 3.8 | 135.1 ± 3.4 | 0.49 |
Diastolic | 74.7 ± 1.8 | 78.8 ± 2.2 | 0.17 |
Notes: Slopes of loss of Ccreat not significantly different at 52 weeks (n = 71); significantly different at 78 weeks (n = 49); did not change when the graph was drawn from 52 to 78 weeks.
In light of newer angiotensin mechanisms in the research literature, our total data set,17 which has now become more relevant,19 can be summarized as follows (Table 4).
Table 4.
A. Normal population
B. Type 1 diabetic nephropathy patients
C. Mean arterial pressure for patients with highest slope of loss of Ccreat baseline, endpoint
D. Mean arterial pressure for patients with lowest slope of loss of Ccreat baseline, endpoint |
Note:
P < 0.05.
The level of blood pressure was not lower with better preservation of renal function and was not higher with the faster loss of Ccreat in both study groups.
Pulsatile infusion added no benefit to multiple injections in the absence of ACE inhibitors.
The DCCT protocol group required significantly larger doses of ACE inhibitors (captopril, enalapril, fosinopril, lisinopril, and quinapril) compared with the pulsatile infusion group as predicted.16
The combination of ACE inhibition with pulsatile infusion was significantly better in preservation of Ccreat than ACE inhibition with multiple injections of insulin. When calculations were limited to individuals treated with ACE inhibitors, the estimated time for Ccreat to fall from 50–60 to 10–15 mL/min was ∼5 years for the DCCT protocol group versus ∼40 years for the DCCT + pulsatile infusion group.
Heart studies
The positive rationale for use of insulin therapy in type 2 diabetic patients with normal renal function has recently been reviewed.20 Results from the United Kingdom Prospective Diabetes Study (UKPDS)21 found both metformin and insulin to be superior to sulfonylureas in the first 10 years with insulin superior to metformin in the second 10 years, using separate endpoints for death due to myocardial infarction, complications of diabetes mellitus, or a composite of all causes. Microvascular complications were also lowest in the insulin-treated group in the 20-year follow-up.
With left ventricular mass (LVM) increase, a long-term increased risk of heart failure and other morbid cardiovascular events is observed. The prevalence of morbid events also increases as renal function decreases. LVM increases proportionately with pressure/volume increase, particularly if associated with calcification/stenosis of the cardiac valves, or the fluid overload of progressive renal dysfunction. Adverse ventricular remodeling therapy has focused on hemodynamic alteration. In our limited experience, however, lowering of glycated hemoglobin A1c concentration was associated with beneficial ventricular remodeling in diabetic nephropathy patients at equivalent blood pressures, suggesting a metabolic nonhemodynamic mechanism22 (Table 5). Increased LVM in these patients would be seen as interstitial collagen/fibrosis deposition superimposed on myocyte hypertrophy.23 Reversal of diabetic cardiomyopathy (a definition restricted to individuals without coronary obstructive disease) was observed after successful renal transplantation.24 Elimination of fiber stretch may be a signal for downregulation of local angiotensin elaboration in the left ventricle, a subject of much research interest in both heart and kidney protocols.
Table 5.
A. Objective measures of autonomic nervous system function
B. Subjective response to questionnaire27
|
A study of glycemia control in type 1 diabetic patients with nephropathy involved 23 patients with cardiac autonomic neuropathy (CAN). Of these, 10 were classified as early autonomic neuropathy and 13 as advanced CAN. None of the patients were receiving β-blocker medications. Glycohemoglobin fell significantly at 3, 6, and 12 months in the early CAN group, but only at 6 months for advanced CAN. There were no changes in heart rate variability (HRV) for the advanced CAN group on 24-h ambulatory EKG over the course of the study. There were statistically significant increases in several tests within the time and frequency domains for HRV, suggesting an improvement in parasympathetic function over the course of 1 year in the early CAN group.25 Most of the patients were receiving ACE inhibitors. In this same study, an improvement in glycohemoglobin A1c was associated with a significant reduction in LVM.22 Patients who did not achieve a significant improvement in A1c% also did not have a significant loss of LVM despite similar blood pressure control. Variation of HRV by the coefficient of variation of the RR interval (CVNN) test was related to loss of excess LVM.26 A1c was related to both autonomic nerve function and LVM, so finding an association between autonomic nerve function and LVM change was expected (Table 5).
A questionnaire was used to determine subjective neurologic endpoints in the study groups. Patients who experienced relative stability of Ccreat reported significantly fewer problems with visual blurring, postural imbalance, intestinal disturbance (diarrhea), sexual dysfunction, and peripheral neuropathic sensations (numbness, tingling, burning, and other pain).27 Although AGE may interfere with recovery from objective sensory neuropathy,28 improvement in plasma concentration did not discriminate those patients who had subjective neurological benefits from those who did not.27 Intermittent visual blurring may be due to dysfunction of corneal nerves that can now be studied with confocal microscopy.29 Use of this technology has documented early regeneration of corneal nerves following pancreas transplantation.30
Eye studies
Diabetic Retinopathy Control Trial-Prevention (DIRECT-Prevent 1) and Diabetic Retinopathy Control Trial-Protection (DIRECT-Protect 1) studies are two randomized, double-shielded, parallel-design, placebo-controlled trials that studied the effect of the ARB candesartan in normotensive, normoalbuminuric type 1 diabetic patients without or with retinopathy, respectively. In DIRECT-Prevent 1, during 4.7 years, the incidence of new retinopathy was significantly lower in the candesartan group.31 However, among patients with existing retinopathy (DIRECT-Protect 1), candesartan did not have a beneficial effect on progression,31 unlike results in the EURODIAB-controlled trial of lisinopril in insulin-dependent diabetes (EUCLID) in which lisinopril decreased retinopathy progression in nonhypertensive patients who had type 1 diabetes with little or no nephropathy.32 With regard to type 2 diabetic patients with mild to moderate retinopathy studied in the DIRECT-Protect 2 trial, candesartan significantly promoted the regression of retinopathy,33 as opposed to findings in the ADVANCE study in which perindopril–indapamide-based blood pressure lowering or intensive glucose control did not significantly reduce the incidence and progression of retinopathy.34 Although most studies demonstrate the prevention of the new appearance of retinopathy in type 1 diabetic patients treated with angiotensin-active medications, there is major inconsistency in findings of protection from retinopathy progression for both type 1 and type 2 diabetic patients treated with angiotensin-active medications.
ACE inhibition and blockade of angiotensin receptor have been studied in multiple centers with an emphasis on small vessel complications of retinal and renal circulations. The effect of blood pressure lowering was demonstrated in the UKPDS study in which there was not much difference in retinopathy outcomes between an ACE inhibitor (captopril) and a β-blocker (atenolol), as long as similar blood pressure control was obtained (average attained BP 144/82, mean pressure 108 mmHg), suggesting that the origin of retinal benefits in newly diagnosed type 2 diabetic patients was hemodynamic.35 When compared with placebo, progression of retinopathy in the RAAS study was reduced by 65% with enalapril and by 70% with losartan. Blood pressures were significantly lower in the enalapril and losartan groups when compared with placebo.9 Confirmation of the relationship between microvascular complications, markers of inflammation, and glycemia control can now be achieved through the use of computer-assisted intravitreal microscopy.36 Future studies with technology that is capable of quantifying macular edema37 by optical coherence tomography and retinal neurodegeneration38 may be useful in the evaluation of treatments.
Use of the standard grading system established by the Early Treatment Diabetic Retinopathy Study (ETDRS)39 at the central reading center (University of Wisconsin, Madison, WI, USA) provided an adequate measure of objectivity in evaluating a subset (n = 57) of the Pulsatile Intravenous Insulin Treatment Study.17 There were no statistically significant differences in the two insulin study groups with respect to changes in retinal grading, despite significant differences in objective renal and subjective neurologic function over the course of the study.40 Baseline blood pressure, glycohemoglobin A1c, and use of ACE inhibitors were not statistically significantly different. This study was not powered to define an effect of pulsatile intravenous insulin on the progression of retinopathy. In this small study, when aggressive lowering of glycemia in the total group was analyzed, it appeared that progression of retinopathy was associated with a higher degree of variability of A1c despite lower levels of A1c, although not meeting statistical significance.41 An effect of high degrees of A1c variability has been reported to adversely affect incidence and progression of nephropathy/retinopathy in type 1 diabetes;42,43 however, in both instances, variability was highest when A1c was highest.
Research studies
Research studies in humans indicate that the protective effects on microvasculature from the ARB losartan are related to its minor metabolite, which does not block the angiotensin receptor. Instead, EXP3179 has anti-inflammatory, antiplatelet aggregation and antioxidant properties not found in other ACE inhibitors or ARBs. In animal models, angiotensin receptor blockade and ACE inhibition have been found to have antifibrotic effects, related in part to their effect on profibrotic local RAAS in the kidney, heart, and eyes.
Human studies
A pathogenetic role of angiotensin 2 (AT2) in microvascular complications in which blood pressure was not the central issue came from a study of 13 type 2 diabetic patients treated in a randomized fashion with either atenolol or losartan.44 Flow-mediated dilation (endothelial-dependent) via hyperemia increased significantly following losartan compared with atenolol (P = 0.01) despite similar blood pressure control. Levels of 8-isoprostane, a marker for oxidative stress, did not change with atenolol but decreased significantly with losartan. The rise in flow-mediated dilation versus the fall in concentration of 8-isoprostane was statistically significant (Table 6).
Table 6.
Inflammation | Hemostasis | Oxidative stress | Vasodilation | Angiogenesis | Fibrosis | |
---|---|---|---|---|---|---|
A. Endothelial function | ||||||
1. Human study | ||||||
a. Losartan: Flammer et al44 | + | + | + | |||
Type 2 diabetes | ||||||
b. Losartan: Kramer et al45 | + | |||||
No diabetes | ||||||
c. EXP3179: Fortuno et al46 | + | |||||
No diabetes | ||||||
2. Animal study | ||||||
a. EXP3179: Watanabe et al63 | + | + | ||||
No diabetes | ||||||
b. Valsartan: Michel et al64 | + | |||||
Spironolactone | + | |||||
No diabetes | ||||||
B. Retinal function | ||||||
1. Animal study | ||||||
a. Candesartan: Kim et al60 | + | + | ||||
b. Fosinopril: Zheng et al58 | + | |||||
c. Enalapril: Kim et al57 | + | |||||
d. Candesartan: Fukumoto et al59 | + | + | + | |||
e. Valsartan: Wilkinson-Berka et al61 | + | |||||
Spironolactone | + | + | ||||
C. Renal function | ||||||
1. Human study | ||||||
a. Irbesartan: Persson et al10 | + | |||||
b. Protein kinase C inhibitors: Gruden et al56 | + | |||||
Tyrosine kinase inhibitors | ||||||
Human mesangial cells: no diabetes | ||||||
c. Enalapril, losartan, and spironolactone: Furumatsu et al11 | + | |||||
Nondiabetic | ||||||
2. Animal study | ||||||
a. ACE2: Qudit et al47 | + | + | + | |||
Akita diabetic mouse | ||||||
b. Quinapril: Blanco et al83 | + | + | ||||
Zucker obese rat | ||||||
D. Cardiac function | ||||||
1. Animal study | ||||||
a. Enalapril: Ma et al49 | + | + | ||||
Losartan | + | + | ||||
Sprague–Dawley rat | ||||||
b. Trandolapril: Onozato et al50 | + | + | ||||
Eplerenone | + | + | ||||
Dahl salt-sensitive rat | ||||||
c. Quinapril: Nemeth et al51 | + | |||||
Spironolactone | + |
Note: + indicates a favorable response in returning marker toward control level.
Losartan, an angiotensin receptor 1 (AT1) blocker, functions through its major metabolite, known as EXP3174. Its minor metabolite, EXP3179, has important physiologic effects not involving blockade of the AT1 receptor. In a study involving 28 subjects with normal renal function, losartan treatment was associated with a significant decrease in platelet aggregation and concentrations of prostaglandin F2-α (PGF2α). The same researchers treated human endothelial cells ex vivo with EXP3179, which has molecular homology with indomethacin, prior to treatment with the proinflammatory agents (AT2, lipopolysaccharide), demonstrating inhibition of cyclooxygenase and arachidonic acid stimulation of platelet aggregation.45 Mononuclear cells from hypertensive subjects (n = 153) that were evaluated ex vivo through the use of EXP317946 demonstrated inhibition of superoxide generation by nicotine adenine dinucleotide phosphate (NADPH) oxidase. Levels of plasma matrix metalloproteinase were also suppressed. This antioxidant response could not be demonstrated with an ACE inhibitor (quinapril), AT1 blockers (irbesartan and losartan), or EXP3174 (Table 6).
Reported functions of EXP3179 include cyclooxygenase blockade (anti-inflammation), nitric oxide synthase stimulation (vasodilation), tumor necrosis factor inhibition (antiapoptosis), peroxisome proliferator-activated agonist response (protects proximal tubule from toxicity of fatty acids in nephrotic syndrome), and decreased platelet aggregation by collagen inhibition (diminishes thrombosis).
Animal models
Fibrosis of the kidney and heart has been studied in experimental animals with or without diabetes. Anti-angiotensin–aldosterone treatments have been found to interrupt fibrosis. In some instances, the impact occurred without change in blood pressure. The diabetic Akita mouse model was used to test ACE2, a homolog for ACE, and was found to be associated with a decrease in AT2 levels in the plasma and renal cortex.47 Urinary albumin and 24-h albumin excretion were significantly lower compared with untreated controls. Glomerular hypertrophy, basement membrane thickening, and mesangial matrix expansion with collagen and smooth muscle actin were significantly less prominent than observed in controls. Renal cortical expression of nicotine adenine dinucleotide (NAD) phosphate oxidase, nitric oxide synthase oxidase, and protein kinase C was significantly lower in treated animals. These findings provide evidence that blockade of signals of inflammation and oxidative stress diminished renal interstitial and mesangial fibrosis in the Akita diabetic mouse (Tables 6 and 7).
Table 7.
A. Alloxan diabetic dog
B. Akita diabetic mouse
C. Zucker obese rat
|
Demonstration of the effect of AT2 on the cardiac fibroblast was undertaken in a newborn Wistar rat model (nondiabetic). Genes for fibronectin and collagen increased within 45 min, but TGF as a marker for extracellular matrix remodeling did not rise until after 60 min of AT2 superimposed upon hypoxia.48 The investigators concluded that the combined influence of AT2 and hypoxia may promote remodeling of myocardial interstitial matrix even in the absence of diabetes.
Three additional animal models for fibrosis (glomerulosclerosis) have concentrated on relevant mechanisms although are unable to eliminate hypertension as a factor (Table 6). Two of the studies used 5/6 nephrectomy in the rat, and one employed the Dahl salt-sensitive hypertensive rat. Mechanisms for fibrosis included increased plasminogen activator inhibitor,49 increased TGF-β (TGFB) + NADPH oxidase,50 and TGFB + collagen type 4.51 In these three animal models, treatment included enalapril, losartan, or both;49 trandolapril, eplerenone, or both;48 and quinapril, spironolactone, or both.51 Glomerulosclerosis was reduced toward baseline with all of these therapies along with reversal of increased profibrosis mechanisms. Such studies are encouraging in that use of anti-angiotensin–aldosterone medications may prevent irreversible remodeling of the left ventricle. Chronic kidney disease models associated with deficiency of vitamin D52 or vitamin D receptor53 demonstrate increased interstitial inflammation/fibrosis of the kidney52,53 or heart.54 Either losartan53 or 1,25-dihydroxyvitamin D54 may impede interstitial fibrosis of the kidney53 or heart54 in chronic kidney disease models.
When alloxan diabetic dogs were observed for 6 months, with no significant differences in aortic pressure, heart rate, or ejection fraction compared with normal controls, an increase in myocardial collagen was found (Tables 7). This collagen was linked to AGE and associated with decreased end-diastolic volume and increased end-diastolic pressure (and could be prevented by either aminoguanidine or enalapril), again suggesting a biochemical nonhemodynamic target.55
A local RAAS exists in both the kidney and heart. Tension to the glomerular mesangium increases expression of protein kinase C and vascular endothelial growth factor (VEGF), resulting in fibrosis of the kidney,57 eye, and heart.23
Mechanisms of microangiopathic remodeling as they relate to AT2 signaling in the retina are summarized in Table 6. A combination of reverse transcriptase polymerase chain reaction, immunohistochemistry, and Western blotting demonstrated increased expression of VEGF in the retina of streptozotocin diabetic (STZ) rats. Eight weeks of treatment with enalapril (10 mg/kg) prevented an increase in retinal VEGF expression.56 Increased VEGF expression both in the retina of STZ rats and in bovine retinal endothelial cells (BREC) exposed to hyperglycemia has also been demonstrated. This increased VEGF expression was attenuated by an ACE inhibitor that is associated with a diminution in histologic markers for retinal vascular damage.58 This study showed the protective effect of perindopril on mitochondrial dysfunction-induced reactive oxygen species (ROS).57 The Tori rat, a model of spontaneous type 2 diabetes, was used to demonstrate that retinal expression of genes for both VEGF and NADPH oxidase were promoted by AT2, which was reduced by candesartan.59 This RAAS-ROS connection via AT2 infusion was associated with increased expression of VEGF and a subunit of the ROS-generating NADPH oxidase;59 these increases in expression (Table 6). were attenuated by treatment with the ARB candesartan. It was found that retinopathy in the STZ diabetic animal model was associated with vascular leakage and increased VEGF expression (Table 6). Treatment with perindopril was able to reverse all of these abnormalities and restore the integrity of tight junction proteins that occlude the spaces between cells.60
An RAAS process was associated with inflammation, angiogenesis, and enhanced expression of NADPH oxidase in oxygen-induced retinopathy. Spironolactone and valsartan both prevented inflammation and angiogenesis.61 Because aldosterone inhibits nitric oxide synthase, spironolactone may have been helpful in promoting the generation of nitric oxide.62
The post-insulin receptor signal is conveyed to mitochondria via phosphoinositol 3-phosphate (PI3p), Akt, and adenosine monophosphate (AMP) with inhibition by AT2. This PI3p/Akt signal system operates in bovine aortic endothelial cells. Studies using losartan have shown that metabolite EXP3179 inhibits the angiotensin response, protects the receptor for VEGF, and therefore diminishes cellular death through apoptosis.63 In the mouse limb ischemic model, induction of neovascularization and VEGF protein are demonstrated in aldosterone-treated animals and inhibited equally by spironolactone, valsartan, and antibody to VEGF64 (Table 6). Because aldosterone is instrumental in neovascularization of both the ischemic limb and the retina exposed to excess oxygen, the interactions of angiotensin/aldosterone, VEGF, and the insulin signal require further elucidation. Further research could be done on whether the new class of renin inhibitors might also exhibit nonhemodynamic synergy with pulsatile insulin release with microvascular benefits.65
Type 2 diabetes: glomerular podocyte and pancreatic β-cell undergo similar dysfunction
Ultrastructural studies of the podocyte and pancreatic β-cell reveal similarities in cytoskeletal proteins, such as nephrin, that are related to cell trafficking as well as to leveraging proteins in cardiac myocytes. Inflammatory effects of obesity interfere with efficient functioning of those proteins and thus interfere with the podocyte slit diaphragm and insulin release. Type 2 diabetes is characterized by increased size and disordering of the amplitude of pulsatile insulin release from β-cells and eventually by decreased size of the pulse, and may be related to a local angiotensin–aldosterone effect on somatostatin. The interruption of adequate delivery of glucose to the medullary thick ascending limb leads to injury from excess angiotensin–aldosterone. Future research may show that correction of excess angiotensin–aldosterone may help to reverse these effects.
Developments in researching the ultrastructure of the kidney epithelial cell podocyte, the pancreatic islet β-cell, and certain muscular structures have identified similar proteins through which the cytoskeleton provides leverage for timely functions. Resisting the excretion of albumin, enhancing the secretion of insulin, or relaxing a muscle appropriately after contraction requires proteins, some of which are structurally similar, that can function through attachment to the cytoskeletal protein actin. Electron microscopy of the β-cell has identified an actin latticework that is available for attachment.66
The cytoskeleton is no longer viewed as a capsule to prevent entry of bacteria and viruses. Actin filaments form polymers that have been described as a polygon on electron microscopy.67 Much is known about the β-cell, which mainly employs nephrin and syntaxin interaction with actin. The podocyte utilizes large numbers of cooperating proteins, among which are nephrin, NCK protein, podocin, and synaptopodin. When the extracellular portion of nephrin engages neighboring foot processes,68 the intracellular portion engages actin to promote leverage (Table 8).
Table 8.
Podocyte of kidney | Pancreas β-cells |
---|---|
Anchors adjacent foot processes by extracellular domain through attachment to actin cytoskeleton by its intracellular domain68 | Anchors intracellular filaments of actin cytoskeleton66,67 |
Requires assistance of other proteins (NCK, podocin, and syn protein family). Receptor for advanced glycated end products colocated with synaptopodin72 | Requires assistance of syntaxin71 intracellularly in a way more closely cooperative than the functions of podocin and synaptopodin, where the leverage effect occurs extracellularly |
Cooperates in translocation of glucose transporter protein vesicles across cytoplasm to dock at plasma membrane.69 Accompanied by VAMP, a protein that moves with other proteins that have taken the form of vesicles (granules). Process is critical for intake of fuel; insulin and adiponectin have a signaling role | Promotes translocation of insulin vesicles across cytoplasmic ‘cell web’ to dock at plasma membrane. Accompanied by VAMP. Process critical for timely insulin secretion70 |
Inhibits nuclear factor-κβ (NF-κβ).82 Animal model of mutant nephrin79,80 adiponectin knockout81 have elevated NF-κβ, proteinuria, glomerular pathology corrected with replacement of nephrin | AGEs cause apoptosis. AGEs also can cross-link actin filament to alter the timing and amplitude of insulin secretion73 |
Coexpressed with myocardin, a protein of both cardiac and smooth muscle origin. Other podocyte proteins (smoothelin and calponin) relate strictly to a smooth muscle origin. Through smoothelin, podocyte responds to angiotensin 2 by contraction in actin-dependent manner.74 Podocytes have similarities to pericytes of the central nervous system and retina | Angiotensin 2 provokes apoptosis through NADPH oxidase; prevented by telmisartan75 |
In both the podocyte and the β-cell, coordinated function is most critical after the intake of food. Efficient function of the nephron and the islets of Langerhans requires opening and closing of transit pathways. The β-cell has a cytoplasmic latticework66 ‘cell web’ that conducts vesicles of insulin to the outer cell membrane. The podocyte has efficient glucose absorption once the glucose transport (GLUT) apparatus has been conducted to the outer cell membrane. Both podocytes and β-cells have a vesicular-binding protein, VAMP (vesicle-associated membrane protein), that associates with moving components and then separates as movement ends with docking at the plasma membrane. In the podocyte, nephrin and VAMP associate with GLUT protein vesicles that are then able to move to the plasma membrane with a signal from insulin.69 In the β-cell, nephrin and VAMP associate with insulin vesicles70 that are then able to move to the plasma membrane with the help of syntaxin71 and a signal from glucose (Table 8).
The effects of diabetes or obesity may inhibit the impact of nephrin and adiponectin. AGEs impede podocyte function by cross-linking of filaments of actin and associated leverage proteins, making it difficult to keep the foot processes of adjacent podocytes aligned. The receptor for AGE is colocalized with synaptopodin, one of the leverage proteins responsible for the retention of albumin.72 AGEs also accelerate apoptosis of β-cells and podocytes by elaboration of an apoptosis effector protein.73
Podocyte chemistry has remote relationships to cardiac physiology. Nephrin is coexpressed with myocardin, a protein of both cardiac and smooth muscle origin. Other podocyte proteins (smoothelin and calponin) relate strictly to a smooth muscle origin. Through smoothelin, the podocyte responds to AT2 by contraction in an actin-dependent manner74 (Table 8). Insulin resistance is associated with inhibition of AMP kinase through increased protein concentration of NOX4, an oxidase of NAD. Angiotensin-induced oxidative stress in heart failure may respond to the use of anti-angiotensin medications, such as receptor blockers. Telmisartan inhibits the oxidase of NADPH and promotes AMP kinase, which leads to more efficient generation of adenosine triphosphate (ATP). This same sequence occurs in the β-cell, which would otherwise experience a reduction in glucose-stimulated insulin secretion,75 and might also be at risk for apoptosis.76 Metformin enhances AMP kinase, which is protective in experimental models of β-cell apoptosis77 and heart failure.78
Nephrin suppresses the inflammation cascade by inhibiting nuclear factor-κβ (NF-κβ), as does adiponectin, the adipokine most active in promoting the postreceptor insulin signal. Animal models of mutant nephrin79,80 or adiponectin knockout81 demonstrate that proteinuria is found to be reversible with restoration of functional nephrin or adiponectin (Table 8). Adiponectin inhibits NF-κβ through a cyclic AMP-dependent pathway,82 the last major point in the insulin signal before activation of mitochondrial glucose oxidation80 (Table 8). The obese diabetic Zucker rat develops glomerulosclerosis and interstitial infiltrate. Glomerular nephrin is depleted. Quinapril, but not diltiazem, reversed all of these abnormalities without a change in blood pressure83 (Table 6).
Insulinization utilizing pumps has been an immense step forward in glycemia control. Nevertheless, there is no method of physiological infusion of insulin in routine use. Insulin is secreted in pulses every 10 ± 2 min. It has been considered that the first phase of insulin secretion is the release of older granules that are already docked at the plasma membrane. The second phase of insulin secretion involves younger insulin vesicles bound to their intracytoplasmic protein carriers (nephrin and syntaxin) co-operating in propulsion through the transit pathway toward the plasma membrane (Table 8). Recent cellular research has suggested that under certain experimental conditions, the younger insulin granules from healthy human β-cells may be the first to be released in response to glucose.84 Both oral intake and intravenous infusion of 30 g of glucose increases the rate of pulsation by 40%, but the incretin effect with oral intake results in a 70% greater mass of insulin secreted in these same pulsations.85
The onset of type 2 diabetes is characterized by increased amplitude and disordering of the rhythm of insulin pulses. In later stages, as the size of the pulse decreases, the patient becomes insulin dependent. Several drugs increase the amplitude but do not change the rhythm of pulses: sulfonylurea,86 glucagon-like peptide,87 and sodium salicylate.88 Decreases in pulse amplitude of insulin secretion may occur as a result of catechol stimulation. A link between type 2 diabetes and pheochromocytoma has been described in patients with a genetic mutation, resulting in an increased number of α-2 adrenoreceptors with a decreased number of insulin granules docked at the β-cell plasma membrane.89 This could explain the observation of hyperglycemia during increased catecholamine expression as being related to increased glycogenolysis and decreased secretion of insulin pulses.90
Metabolic efficiency is improved by pulsatile hormonal delivery when compared with steady infusion91,92 (Table 9). The islets of Langerhans secrete glucagon,91 insulin,92 and somatostatin91 in a pulsatile fashion from the α-, β-, and γ-cells. There is general agreement that somatostatin acts as a governor of these cells through receptors on α- and β-cells,93 preventing overwork to the point of exhaustion. β-cells also have local AT2-generating systems94 through angiotensin-converting enzyme with receptors (AT1 and AT2) in place. Action on the β-cell angiotensin axis can be expected to affect metabolism of somatostatin, insulin, and glucagon. Studies of insulin secretion following infusion of AT2 in healthy volunteers have shown that pressor doses diminished levels of basal and glucose-stimulated oscillations. The regularity of insulin oscillation was not changed by AT2 infusion.95 We suspect that future studies will demonstrate that inhibition of excess angiotensin–aldosterone has widespread benefits in hormonal balance that may not be corrected simply with physiological insulinization.
Table 9.
A. Insulin secretion
B. Islets of Langerhans
C. Type 2 diabetes
D. Drugs that increase amplitude, but do not change rate of oscillations E. Drugs that decrease amplitude, but do not change secretion rate |
Preservation of function in chronic glomerular disorders depends on remodeling of the interstitium. Disorders of blood supply or oxygen transfer, such as diabetes or sickle cell anemia, are associated with medullary papillary necrosis while the cortex is spared. The cortex can utilize fuels generated under stress,96,97 including ketones (β-hydroxybutyrate), fatty acids (palmitate), glycerol, triglycerides, glutamine, lactate, mannose, and fructose.97
Medullary energy production depends on anaerobic glycolysis in the thick ascending limb of the distal tubule where mitochondria exists. Energy for sodium/chloride transport against an interstitial gradient for water resorption in the collecting ducts depletes medullary reserves.98 As the renal medulla operates at lower levels of oxygen concentration/consumption,99,100 hematocrit,101 and blood flow101 than the cortex, pulsatile insulin delivery may be critical. These observations pinpoint the renal medullary interstitium as a susceptible focus of injury from excess angiotensin–aldosterone when imperfect insulin supply occurs.
Individuals with type 1 diabetes are not exempt from the growing epidemic of obesity, which would be expected to add insulin resistance to insulin deficiency. In type 1 diabetic nephropathy patients treated with pulsatile insulin infusions, we have observed an increase in respiratory quotient (RQ) in every instance, indicating a direction of fuel oxidation away from β-oxidation of fatty acids to aerobic oxidation of glucose in mitochondria.102 In several patients, transitory elevations of RQ to >1.0 may have been the result of excess fuel with synthesis of fatty acid, leading to deposition of triglyceride, because diacylglycerol is also an endpoint of insulin action. Deposition of triglyceride may occur in the liver, adipose, muscle (skeletal and cardiac), and pancreas (β-cell). We have no experience with type 2 diabetic nephropathy patients undergoing experimental use of pulsatile insulin infusion.
From these biological observations it can be speculated that glomerular and β-cells are injured in similar ways by mechanisms seen in uncontrolled hyperglycemia. Therefore, treatments that eliminate these injury patterns may be expected to preserve the function of both the nephron and the islets of Langerhans. The function of the myocardium tends to improve in situations that are favorable to either the pancreas or the kidney. When the function of the myocardium has been improved, there is improvement in renal function.
Future potential interventions to control cellular metabolic derangement
Studies of the use of TNF-α inhibitors in rheumatologic disease suggest clinical (reduction of cardiac events) and laboratory evidence (improved HDL antioxidant capacity and endothelial responsiveness) for cardiovascular benefit.103 A role of both TNF-α and IL-1 has been demonstrated in postinfarction human heart failure and isolated rat cardiac fibroblast. In both instances the process is augmented by angiotensin.104–106
Demonstration that RAAS activation, mechanical stretch, and myocardial injury stimulate production of TNF-α,107 hydroxyl radicals, IL-1β, and NF-κβ has led to studies that demonstrate the possibility that the benefit of medications such as carvedilol108,109 and statins110 may be related not only to their effect on heart rate, blood pressure, and lipids but also to regulation of oxidative stress. Animal studies support TNF-α inhibition as a therapy for the reduction of oxidative stress, myocardial mitochondrial dysfunction, and apoptosis.111,112 Laboratory and clinical evidence additionally links TNF-α and oxidative stress to postmyocardial infarction progression to heart failure.113 Thiazolidinediones may enhance TNF-α induction of IL-1, and this may explain problems within this class of drugs.104
There are no clinical outcome studies involving inhibition of TNF-α in patients with diabetic nephropathy. The effect of both AT2114 and TNF-α115 on sodium potassium ATPase of the medullary thick ascending limb of the loop of Henle is to inhibit resorption of sodium and chloride. We have referred to this site in the renal medulla, which is critical for water preservation, as a potential site for injury in uncontrolled glycemia with insulin deficiency/resistance.98 The oxidative stress that attacks the most vulnerable position in the renal medulla, where energy is required for transport of sodium chloride, has been shown to be overcome by candesartan/valsartan in type 2 diabetes.116 Because this therapeutic process was associated with inhibition of expression of IL-6, there is a suggestion that future studies should involve a combination of therapy of ARBs with inhibitors of IL or TNF-α. A note of caution is required, however, due to reports of the development of immune glomerulonephritis in rheumatoid arthritis patients treated with etanercept, adalimumab, or infliximab.117
The primary focus for the preservation of cardiovascular, renal, and retinal integrity in diabetes has been related to RAAS activation, blood pressure, and glycemic control. Recent studies have demonstrated relationships between these factors and cellular signaling that may be additional targets for intervention. The goal is to alter rates of programmed cell death (apoptosis), oxidative stress, thrombosis, inflammation, and fibrosis. Given the impact of successes in the rheumatologic therapies, future research using these additional cellular targets may be worthwhile.
Conclusion
We have reviewed angiotensin–aldosterone mechanisms of tissue injury that magnify microvascular damage caused by hyperglycemia and hypertension. Many studies describe interruption of these mechanisms, without hemodynamic consequence, in the preservation of function in type 1 diabetes. Possible interactions between the RAAS and physiologic glycemic control (through pulsatile insulin release) suggest opportunities for clinical research. Until we have better markers of risk for microvascular complications, therapy must be directed at minimizing variability of hemoglobin A1c.118
The results of recent outcome trials challenge hypotheses that tight control of both glycohemoglobin and blood pressure diminishes macrovascular events and survival among type 2 diabetic patients. These results raise relevant questions. Is glycohemoglobin an adequate measure of diabetes control? Are we ignoring mechanisms of vasculotoxicity (profibrosis, altered angiogenesis, hypertrophy, hyperplasia, and endothelial injury) inherent in current antihyperglycemic medications? Is the polypharmacy for lowering cholesterol, triglyceride, glucose, and systolic blood pressure producing drug interactions that are too complex to be clinically identified? Answers to these questions will most certainly improve our understanding of disease mechanisms and further refine therapies.
On the basis of our review of the literature, we suggest that these nonhemodynamic effects on renal, cardiac, and ocular microvasculature are as important, if not more so, as the hemodynamic effects of ACE inhibition and AT2 receptor blockade. Although the macrovascular effects of uncontrolled hypertension and diabetes are clear, we are only now beginning to understand the microvascular complications. Answers to these questions will most certainly improve our understanding of disease mechanisms and allows us to further refine therapies, such as the interruption of local RAAS and use of pulsatile insulin to reduce proinflammatory and profibrotic forces. These therapies, in turn, will help to improve the lives of our patients.
Acknowledgments
We wish to thank Bruce Bistrian, Nathan Norris, Aldo Rossini, and Diane Young for their painstaking and helpful research in the preparation of this manuscript.
Footnotes
Disclosure
The authors report no conflicts of interest in this work.
References
- 1.The Diabetes Control and Complications Trial Research Group The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. N Engl J Med. 1993;329(14):977–986. doi: 10.1056/NEJM199309303291401. [DOI] [PubMed] [Google Scholar]
- 2.Lewis E, Hunsicker LG, Bain RP, Rohde RD. The effect of angiotensin-converting-enzyme inhibition on diabetic nephropathy. N Engl J Med. 1993;329(20):1456–1462. doi: 10.1056/NEJM199311113292004. [DOI] [PubMed] [Google Scholar]
- 3.UK Prospective Diabetes Study (UKPDS) Group Intensive blood glucose control with sulfonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33) Lancet. 1998;352(9131):837–853. [PubMed] [Google Scholar]
- 4.UK Prospective Diabetes Study (UKPDS) Group Tight blood pressure control and risk of macrovascular and microvascular complications in type 2 diabetes: UKPDS 38. BMJ. 1998;317(7160):703–713. [PMC free article] [PubMed] [Google Scholar]
- 5.Gerstein HC, Miller ME, et al. ACCORD Study Group Effects of intensive glucose lowering in type 2 diabetes. N Engl J Med. 2008;358(24):2545–2559. doi: 10.1056/NEJMoa0802743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Cushman WC, Evans GW, et al. ACCORD Study Group Effects of intensive blood-pressure control in type 2 diabetes mellitus. N Engl J Med. 2010;362:1575–1585. doi: 10.1056/NEJMoa1001286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Wolf G, Ziyadeh FN, Helmchen U, Zahner G, Schroeder R, Stahl RA. ANG II is a mitogen for a murine cell line isolated from medullary thick ascending limb of Henle’s loop. Am J Physiol. 1995;268(5 Pt 2):F940–F947. doi: 10.1152/ajprenal.1995.268.5.F940. [DOI] [PubMed] [Google Scholar]
- 8.Ziyadeh FN, Sharma K, Ericksen M, Wolf G. Stimulation of collagen gene expression and protein synthesis in murine mesangial cells by high glucose is mediated by autocrine activation of transforming growth factor-beta. J Clin Invest. 1994;93(2):536–542. doi: 10.1172/JCI117004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Mauer M, Zinman B, Gardiner R, et al. Renal and retinal effects of enalapril and losartan in type 1 diabetes. N Engl J Med. 2009;361(1):40–51. doi: 10.1056/NEJMoa0808400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Persson F, Rossing P, Hovind P, et al. Irbesartan treatment reduces biomarkers of inflammatory activity in patients with type 2 diabetes and microalbuminuria: an IRMA 2 substudy. Diabetes. 2006;55(12):3550–3555. doi: 10.2337/db06-0827. [DOI] [PubMed] [Google Scholar]
- 11.Furumatsu Y, Nagasawa Y, Tomida K, et al. Effect of renin-angiotensin-aldosterone system triple blockade on non-diabetic renal disease: addition of an aldosterone blocker, spironolactone, to combination treatment with an angiotensin-converting enzyme inhibitor and angiotensin II receptor blocker. Hypertens Res. 2008;31(1):59–67. doi: 10.1291/hypres.31.59. [DOI] [PubMed] [Google Scholar]
- 12.Mehdi UF, Adams-Huet B, Raskin P, Vega GL, Toto RD. Addition of angiotensin receptor blockade or mineralocorticoid antagonism to maximal angiotensin-converting enzyme inhibition in diabetic nephropathy. J Am Soc Nephrol. 2009;20(12):2641–2650. doi: 10.1681/ASN.2009070737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.D’Elia JA, Weinrauch LA, Gleason RE, Lipinska I, Pendse S, Roshan B, Lee AT, Tofler G. Fibrinogen and factor VII levels improve with glycemic control in patients with type 1 diabetes mellitus who have microvascular complications. Arch Intern Med. 2001;161(1):98–101. doi: 10.1001/archinte.161.1.98. [DOI] [PubMed] [Google Scholar]
- 14.Aoki TT, Benbarka MM, Okimura MC, et al. Long-term intermittent intravenous insulin therapy and type 1 diabetes mellitus. Lancet. 1993;342(8870):515–518. doi: 10.1016/0140-6736(93)91645-3. [DOI] [PubMed] [Google Scholar]
- 15.Aoki TT, Grecu EO, Arcangeli MA. Chronic intermittent intravenous insulin therapy corrects orthostatic hypotension of diabetes. Am J Med. 1995;99(6):683–684. doi: 10.1016/s0002-9343(99)80257-5. [DOI] [PubMed] [Google Scholar]
- 16.Aoki TT, Grecu EO, Prendergast JJ, Arcangeli MA, Meisenheimer R. Effect of chronic intermittent intravenous insulin therapy on antihypertensive medication requirements in IDDM subjects with hypertension and nephropathy. Diabetes Care. 1995;18(9):1260–1265. doi: 10.2337/diacare.18.9.1260. [DOI] [PubMed] [Google Scholar]
- 17.Dailey GE, Boden GH, Creech RH, Johnson D, Gleason RE, Kennedy FP, Weinrauch LA, Weir M, D’Elia J. Effects of pulsatile intravenous insulin therapy on the progression of diabetic nephropathy. Metabolism. 2000;49(11):1491–1495. doi: 10.1053/meta.2000.17700. [DOI] [PubMed] [Google Scholar]
- 18.Weinrauch LA, Burger AJ, Aepfelbacher F, Lee AT, Gleason RE, D’Elia JA. A pilot study to test the effect of pulsatile insulin infusion on cardiovascular mechanisms that might contribute to attenuation of renal compromise in type 1 diabetes mellitus patients with proteinuria. Metabolism. 2007;56(11):1453–1457. doi: 10.1016/j.metabol.2007.05.016. [DOI] [PubMed] [Google Scholar]
- 19.Weinrauch LA, Gleason RE, D’Elia JA. What have trials of pulsatile intravenous insulin taught us? Metabolism. 2010;59(5):764–765. [Google Scholar]
- 20.Niswender K. Early and aggressive initiation of insulin therapy for type 2 diabetes: what is the evidence? Clin Diabetes. 2009;27(2):60–68. [Google Scholar]
- 21.Holman RR, Paul SK, Bethel MA, Matthews DR, Neil HA. 10-year follow-up of intensive glucose control in type 2 diabetes. N Engl J Med. 2008;359(15):1577–1589. doi: 10.1056/NEJMoa0806470. [DOI] [PubMed] [Google Scholar]
- 22.Aepfelbacher FC, Yeon SB, Weinrauch LA, D’Elia J, Burger AJ. Improved glycemic control induces regression of left ventricular mass in patients with type 1 diabetes mellitus. Int J Cardiol. 2004;94(1):47–51. doi: 10.1016/j.ijcard.2003.04.012. [DOI] [PubMed] [Google Scholar]
- 23.Spiro MJ, Kumar BR, Crowley TJ. Myocardial glycoproteins in diabetes: type VI collagen is a major PAS-reactive extracellular matrix protein. J Moll Cell Cardiol. 1992;24(4):397–410. doi: 10.1016/0022-2828(92)93194-o. [DOI] [PubMed] [Google Scholar]
- 24.D’Elia JA, Weinrauch LA, Healy RW, Libertino JA, Bradley RF, Leland OS., Jr Myocardial dysfunction without coronary artery disease in diabetic renal failure. Am J Cardiol. 1979;43(2):193–199. doi: 10.1016/s0002-9149(79)80004-1. [DOI] [PubMed] [Google Scholar]
- 25.Burger AJ, Weinrauch LA, D’Elia JA, Aronson D. Effect of glycemic control on heart rate variability in type I diabetic patients with cardiac autonomic neuropathy. Am J Cardiol. 1999;84(6):687–691. doi: 10.1016/s0002-9149(99)00417-8. [DOI] [PubMed] [Google Scholar]
- 26.Weinrauch LA, Burger A, Gleason RE, Lee AT, D’Elia JA. Left ventricular mass reduction in type 1 diabetic patients with nephropathy. J Clin Hypertens (Greenwich) 2005;7(3):159–164. doi: 10.1111/j.1524-6175.2005.04307.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Weinrauch LA, Bayliss G, Gleason RE, Lee AT, D’Elia JA. Utilization of an abbreviated diabetes impact management scale to assess change in subjective disability during a trial of pulsatile insulin delivery demonstrates benefit. Metabolism. 2009;58(4):488–491. doi: 10.1016/j.metabol.2008.11.006. [DOI] [PubMed] [Google Scholar]
- 28.Duran-Jimenez B, Dobler D, Moffatt S, et al. Advanced glycation end products in extracellular matrix proteins contribute to the failure of sensory nerve regeneration in diabetes. Diabetes. 2009;58(12):2893–2903. doi: 10.2337/db09-0320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Quattrini C, Tavakoli M, Jeziorska M, et al. Surrogate markers of small fiber damage in human diabetic neuropathy. Diabetes. 2007;56(8):2148–2154. doi: 10.2337/db07-0285. [DOI] [PubMed] [Google Scholar]
- 30.Mehra S, Tavakoli M, Kallinikos PA, et al. Corneal confocal microscopy detects early nerve regeneration after pancreas transplantation in patients with type 1 diabetes. Diabetes Care. 2007;30(10):2608–2612. doi: 10.2337/dc07-0870. [DOI] [PubMed] [Google Scholar]
- 31.Chaturvedi N, Porta M, Klein R, et al. Effect of candesartan on prevention (DIRECT-Prevent 1) and progression (Direct-Protect 1) of retinopathy in type 1 diabetes: randomized, placebo-controlled trials. Lancet. 2008;372(9647):1394–1402. doi: 10.1016/S0140-6736(08)61412-9. [DOI] [PubMed] [Google Scholar]
- 32.Chaturvedi N, Sjolie AK, Stephenson JM, et al. Effect of lisinopril on progression of retinopathy in normotensive people with type 1 diabetes. The EUCLID study group. EURODIAB controlled trial of lisinopril in insulin dependant diabetes mellitus. Lancet. 1998;351(9095):28–31. doi: 10.1016/s0140-6736(97)06209-0. [DOI] [PubMed] [Google Scholar]
- 33.Sjolie A, Klein R, Porta M, et al. Effect of candesartan on progression and regression of retinopathy in type 2 diabetes (DIRECT-Protect 2): a randomized placebo-controlled trial. Lancet. 2008;372(9647):1385–1393. doi: 10.1016/S0140-6736(08)61411-7. [DOI] [PubMed] [Google Scholar]
- 34.Beulens JW, Patel A, Vingerling JR, et al. Effects of blood pressure lowering and intensive glucose control on the incidence and progression of retinopathy in patients with type 2 diabetes mellitus: a randomised controlled trial. Diabetologia. 2009;52(10):2027–2036. doi: 10.1007/s00125-009-1457-x. [DOI] [PubMed] [Google Scholar]
- 35.Hansson L, Zanchetti A, Carruthers SG, et al. Effects of intensive blood-pressure lowering and low-dose aspirin in patients with hypertension: principal results of the Hypertension Optimal Treatment (HOT) randomized trial. Lancet. 1998;351(9118):1755–1762. doi: 10.1016/s0140-6736(98)04311-6. [DOI] [PubMed] [Google Scholar]
- 36.Devaraj S, Cheung AT, Jialal I, et al. Evidence of increased inflammation and microcirculatory abnormalities in patients with type 1 diabetes and their role in microvascular complications. Diabetes. 2007;56(11):2790–2796. doi: 10.2337/db07-0784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Gardner TW, Sander B, Larsen ML, et al. An extension of the Early Treatment Diabetic Retinopathy Study (ETDRS) system for grading of diabetic macular edema in the astemizole retinopathy trial. Curr Eye Res. 2006;31(6):535–547. doi: 10.1080/02713680600746112. [DOI] [PubMed] [Google Scholar]
- 38.Silva KC, Rosales MAB, Biswas SK, Lopes de Faria JB, Lopes de Faria JM. Diabetic retinal neurodegeneration is associated with mitochondrial oxidative stress and is improved by an angiotensin receptor blocker in a model combining hypertension and diabetes. Diabetes. 2009;58(6):1382–1390. doi: 10.2337/db09-0166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Early Treatment Diabetic Retinopathy Study Group Fundus photographic risk factors for progression of diabetic retinopathy. ETDRS report number 12. Opthalmology. 1991;98(Suppl 5):823–833. [PubMed] [Google Scholar]
- 40.Weinrauch LA, Sun J, Gleason RE, Boden GH, Creech RH, Dailey G, Kennedy FP, Weir MR, D’Elia JA. Pulsatile intermittent intravenous insulin therapy for attenuation of retinopathy and nephropathy in type 1 diabetes mellitus. Metabolism. 2010;59(10):1429–1434. doi: 10.1016/j.metabol.2010.01.004. [DOI] [PubMed] [Google Scholar]
- 41.Weinrauch LA, Sun J, Gleason RE, Boden GH, Creech RH, Dailey G, Kennedy FP, Weir MR, D’Elia JA. Progression of retinopathy in type 1 diabetic patients with nephropathy: impact of glycemia control as measured by variation in glycohemoglobin A1C. J Am Soc Nephrol. 2009;20 Abstract. [Google Scholar]
- 42.Waden J, Forsblom C, Thorn LM, Gordin D, Saraheimo M, Groop P-H. A1C variability predicts incident cardiovascular events, microalbuminuria, and overt diabetic nephropathy in patients with type 1 diabetes. Diabetes. 2009;58(11):2649–2655. doi: 10.2337/db09-0693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Kilpatrick ES, Rigby AS, Atkin SL. A1C variability and the risk of microvascular complications in type 1 diabetes: data from the Diabetes Control and Complications Trial. Diabetes Care. 2008;31(11):2198–2202. doi: 10.2337/dc08-0864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Flammer AJ, Hermann F, Wiesli P, et al. Effect of losartan, compared with atenolol, on endothelial function and oxidative stress in patients with type 2 diabetes and hypertension. J Hypertens. 2007;25(4):785–791. doi: 10.1097/HJH.0b013e3280287a72. [DOI] [PubMed] [Google Scholar]
- 45.Kramer C, Sunkomat J, Witte J, et al. Angiotensin II receptor-independent anti-inflammatory and antiaggregatory properties of losartan: role of active metabolite EXP3179. Circ Res. 2002;90(7):770–776. doi: 10.1161/01.res.0000014434.48463.35. [DOI] [PubMed] [Google Scholar]
- 46.Fortuno A, Bidegain J, Robador P, et al. Losartan metabolite EXP3179 blocks NADPH oxidase-mediated superoxide production by inhibiting protein kinase C: potential clinical implications in hypertension. Hypertension. 2009;54(4):744–750. doi: 10.1161/HYPERTENSIONAHA.109.129353. [DOI] [PubMed] [Google Scholar]
- 47.Qudit GY, Liu GC, Zhong J, et al. Human recombinant ACE2 reduces the progression of diabetic nephropathy. Diabetes. 2010;59(2):529–538. doi: 10.2337/db09-1218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Sharma HS, van Heugten HA, Goedbloed MA, Verdouw PD, Lamers JM. Angiotensin II induced expression of transcription factors precedes increase in transforming growth factor-beta 1 mRNA in neonatal cardiac fibroblasts. Biochem Biophys Res Commun. 1994;205(1):105–112. doi: 10.1006/bbrc.1994.2636. [DOI] [PubMed] [Google Scholar]
- 49.Ma LJ, Nakamura S, Aldigier JC, et al. Regression of glomerulosclerosis with high-dose angiotensin inhibition is linked to decreased plasminogen activator inhibitor-1. J Am Soc Nephrol. 2005;16(4):966–976. doi: 10.1681/ASN.2004060492. [DOI] [PubMed] [Google Scholar]
- 50.Onozato ML, Tojo A, Kobayashi N, Goto A, Matsuoka H, Fujita T. Dual blockade of aldosterone and angiotensin II additively suppresses TGF-beta and NADPH oxidase in the hypertensive kidney. Nephrol Dial Transplant. 2007;22(5):1314–1322. doi: 10.1093/ndt/gfl780. [DOI] [PubMed] [Google Scholar]
- 51.Nemeth Z, Kokeny G, Godo M, et al. Increased renoprotection with ACE inhibitor plus aldosterone antagonist as compared to montherapies – the effect on podocytes. Nephrol Dial Transplant. 2009;24(12):3640–3651. doi: 10.1093/ndt/gfp371. [DOI] [PubMed] [Google Scholar]
- 52.Agarwal R. Vitamin D, proteinuria, diabetic nephropathy, and progression of CKD. Clin J Am Soc Nephrol. 2009;4(9):1523–1528. doi: 10.2215/CJN.02010309. [DOI] [PubMed] [Google Scholar]
- 53.Zhang Y, Kong J, Deb DK, Chang A, Li YC. Vitamin D receptor attenuates renal fibrosis by suppressing the renin-angiotensin system. J Am Soc Nephrol. 2010;21(6):966–973. doi: 10.1681/ASN.2009080872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Mizobuchi M, Nakamura H, Tokumoto M, et al. Myocardial effects of VDR activators in renal failure. J Steroid Biochem Mol Biol. 2010;121(1–2):188–192. doi: 10.1016/j.jsbmb.2010.03.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Avendano GF, Agarwal RK, Bashey RI, et al. Effects of glucose intolerance on myocardial function and collagen-linked glycation. Diabetes. 1999;48(7):1443–1447. doi: 10.2337/diabetes.48.7.1443. [DOI] [PubMed] [Google Scholar]
- 56.Gruden G, Thomas S, Burt D, et al. Mechanical stretch induces vascular permeability factor in human mesangial cells: mechanisms of signal transduction. Proc Natl Acad Sci U S A. 1997;94(22):12112–12116. doi: 10.1073/pnas.94.22.12112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Kim HW, Kim JL, Lee HK, Hur DY, Yun IH, Kim SD. Enalapril alters expression of key growth factors in experimental diabetic retinopathy. Curr Eye Res. 2009;34(11):976–987. doi: 10.3109/02713680903249913. [DOI] [PubMed] [Google Scholar]
- 58.Zheng Z, Chen H, Xu X, Li C, Gu Q. Effects of angiotensin-converting enzyme inhibitors and beta adrenergic blockers on retinal vascular endothelial growth factor expression in rat diabetic retinopathy. Exp Eye Res. 2007;84(4):745–752. doi: 10.1016/j.exer.2006.12.013. [DOI] [PubMed] [Google Scholar]
- 59.Fukumoto M, Takai S, Ishizaki E, et al. Involvement of angiotensin II-dependent vascular endothelial growth factor gene expression via NADPH oxidase in the retina in a type 2 diabetic rat model. Curr Eye Res. 2008;33(10):885–891. doi: 10.1080/02713680802389851. [DOI] [PubMed] [Google Scholar]
- 60.Kim JH, Kim JH, Yu YS, Cho CS, Kim KW. Blockade of angiotensin II attenuates VEGF-medicated blood-retinal barrier breakdown in diabetic retinopathy. J Cereb Blood Flow Metab. 2009;29(3):621–628. doi: 10.1038/jcbfm.2008.154. [DOI] [PubMed] [Google Scholar]
- 61.Wilkinson-Berka JL, Tan G, Jaworski K, Miller AG. Identification of a retinal aldosterone system and the protective effects of mineralocorticoid receptor antagonism on retinal vascular pathology. Circ Res. 2009;104(1):124–133. doi: 10.1161/CIRCRESAHA.108.176008. [DOI] [PubMed] [Google Scholar]
- 62.Leopold JA, Dam A, Maron BA, et al. Aldosterone impairs vascular reactivity by decreasing glucose-6-phosphate dehydrogenase activity. Nat Med. 2007;13(2):189–197. doi: 10.1038/nm1545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Watanabe T, Suzuki J, Yamawaki H, Sharma VK, Sheu SS, Berk BC. Losartan metabolite EXP3179 activates Akt and endothelial nitric oxide synthase via vascular endothelial growth factor receptor-2 in endothelial cells: angiotensin II type 1 receptor-independent effects of EXP3179. Circulation. 2005;112(12):1798–1805. doi: 10.1161/CIRCULATIONAHA.104.509760. [DOI] [PubMed] [Google Scholar]
- 64.Michel F, Ambroisine ML, Duriez M, Delcayre C, Levy B, Silvestre JS. Aldosterone enhances ischemia-induced neovascularization through angiotensin II-dependent pathway. Circulation. 2004;109(16):1933–1937. doi: 10.1161/01.CIR.0000127112.36796.9B. [DOI] [PubMed] [Google Scholar]
- 65.Parving HH, Brenner BM, McMurray JJ, et al. Aliskiren trial in type 2 diabetes using cardio-renal endpoints (ALTITUDE): rationale and study design. Nephrol Dial Transplant. 2009;24(5):1663–1671. doi: 10.1093/ndt/gfn721. [DOI] [PubMed] [Google Scholar]
- 66.Lacy PE, Howell SL, Young DA, Fink CJ. New hypothesis of insulin secretion. Nature. 1968;219(5159):1177–1179. doi: 10.1038/2191177a0. [DOI] [PubMed] [Google Scholar]
- 67.Orci L, Gabbay K, Malaisse WJ. Pancreatic beta-cell web: it’s possible role in insulin secretion. Science. 1972;175(26):1128–1130. doi: 10.1126/science.175.4026.1128. [DOI] [PubMed] [Google Scholar]
- 68.Verma R, Kovari I, Soofi A, Nihalani D, Patrie K, Holzman LB. Nephrin ectodomain engagement results in SRC kinase activation, nephrin phosphorylation, NCK recruitment, and actin polymerization. J Clin Invest. 2006;116(5):1346–1359. doi: 10.1172/JCI27414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Coward RJ, Welsh GI, Koziell A, et al. Nephrin is critical for the action of insulin on human glomerular podocytes. Diabetes. 2007;56(4):1127–1135. doi: 10.2337/db06-0693. [DOI] [PubMed] [Google Scholar]
- 70.Fornoni A, Jeon J, Varona Santos J, et al. Nephrin is expressed on the surface of insulin vesicles and facilitates glucose-stimulated insulin release. Diabetes. 2010;59(1):190–199. doi: 10.2337/db09-0655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Jewell JL, Luo W, Oh E, Wang Z, Thurmond DC. Filamentous actin regulates insulin exocytosis through direct interaction with syntaxin 4. J Biol Chem. 2008;283(16):10716–10726. doi: 10.1074/jbc.M709876200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Wendt T, Tanji N, Guo J, et al. Glucose, glycation, and RAGE: implications for amplification of cellular dysfunction in diabetic nephropathy. J Am Soc Nephrol. 2003;14(5):1383–1395. doi: 10.1097/01.asn.0000065100.17349.ca. [DOI] [PubMed] [Google Scholar]
- 73.Chuang PY, Yu Q, Fang W, Uribarri J, He JC. Advanced glycation endproducts induce podocyte apoptosis by activation of the FOXO4 transcription factor. Kidney Int. 2007;72(8):965–976. doi: 10.1038/sj.ki.5002456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Saleem S, Zavadil J, Bailly M, et al. The molecular and functional phenotype of glomerular podocytes reveals key features of contractile smooth muscle cells. Am J Physiol Renal Physiol. 2008;295(4):F959–F970. doi: 10.1152/ajprenal.00559.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Saitoh Y, Hongwei W, Ueno H, Mizuta M, Nakazato M. Telmisartan attenuates fatty-acid-induced oxidative stress and NAD(P)H oxidase activity in pancreatic beta-cells. Diabetes Metab. 2009;35(5):392–397. doi: 10.1016/j.diabet.2009.04.005. [DOI] [PubMed] [Google Scholar]
- 76.Chu KY, Cheng Q, Chen C, et al. Angiotensin exerts glucose-dependent effects on Kv current in mouse pancreatic beta-cells via angiotensin II type 2 receptors. Am J Physiol. 2010;298(2):C313–C323. doi: 10.1152/ajpcell.00575.2008. [DOI] [PubMed] [Google Scholar]
- 77.Marchetti P, Del Guerra S, Marselli L, et al. Pancreatic islets from type 2 diabetic patients have functional defects and increased apoptosis that are ameliorated by metformin. J Clin Endocrinol Metab. 2004;89(11):5535–5541. doi: 10.1210/jc.2004-0150. [DOI] [PubMed] [Google Scholar]
- 78.Sasaki H, Asanuma H, Fujita M, et al. Metformin prevents progression of heart failure in dogs: role of AMP-activated protein kinase. Circulation. 2009;119(19):2568–2577. doi: 10.1161/CIRCULATIONAHA.108.798561. [DOI] [PubMed] [Google Scholar]
- 79.Hussain S, Romio L, Saleem M, et al. Nephrin deficiency activates NF-kappaB and promotes glomerular injury. J Am Soc Nephrol. 2009;20(8):1733–1743. doi: 10.1681/ASN.2008111219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Sheerin NS. A novel role for nephrin in the maintenance of glomerular structure. J Am Soc Nephrol. 2009;20(8):1661–1663. doi: 10.1681/ASN.2009060596. [DOI] [PubMed] [Google Scholar]
- 81.Sharma K, Ramachandra Rao S, Qui G, et al. Adiponectin regulates albuminuria and podocyte function in mice. J Clin Invest. 2008;118(5):1645–1656. doi: 10.1172/JCI32691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Ouchi N, Kihara S, Arita Y, et al. Adiponectin, an adipocyte-derived plasma protein, inhibits endothelial NF-kappaB signaling through a cAMP-dependent pathway. Circulation. 2000;102(11):1296–1301. doi: 10.1161/01.cir.102.11.1296. [DOI] [PubMed] [Google Scholar]
- 83.Blanco S, Bonet J, Lopez D, Casas I, Romero R. ACE inhibitors improve nephrin expression in Zucker rats with glomerulosclerosis. Kidney Int Suppl. 2005;(93):S10–S14. doi: 10.1111/j.1523-1755.2005.09303.x. [DOI] [PubMed] [Google Scholar]
- 84.Michael D, Xiong W, Geng X, Drain P, Chow RH. Human insulin vesicle dynamics during pulsatile secretion. Diabetes. 2007;56(5):1277–1288. doi: 10.2337/db06-0367. [DOI] [PubMed] [Google Scholar]
- 85.Porksen N, Munn S, Steers J, Veldhuis J, Butler PC. Effects of glucose ingestion versus infusion on pulsatile insulin secretion: the incretin effect is achieved by amplification of insulin secretory burst mass. Diabetes. 1996;45(10):1317–1323. doi: 10.2337/diab.45.10.1317. [DOI] [PubMed] [Google Scholar]
- 86.Juhl CB, Porksen N, Pincus SM, Hansen AP, Veldhuis JD, Schmitz O. Acute and short-term administration of a sulfonylurea (gliclazide) increases pulsatile insulin secretion in type 2 diabetes. Diabetes. 2001;50(8):1778–1784. doi: 10.2337/diabetes.50.8.1778. [DOI] [PubMed] [Google Scholar]
- 87.Ritzel R, Schulte M, Porksen N, et al. Glucagon-like peptide 1 increases secretory burst mass of pulsatile insulin secretion in patients with type 2 diabetes and impaired glucose tolerance. Diabetes. 2001;50(4):776–784. doi: 10.2337/diabetes.50.4.776. [DOI] [PubMed] [Google Scholar]
- 88.Matthews DR, Lang DA, Burnett MA, Turner RC. Control of pulsatile insulin secretion in man. Diabetologia. 1983;24(4):231–237. doi: 10.1007/BF00282705. [DOI] [PubMed] [Google Scholar]
- 89.Rosengren AH, Jokubka R, Tojjar D, et al. Overexpression of alpha 2 A-adrenergic receptors contributes to type 2 diabetes. Science. 2010;327(5962):217–220. doi: 10.1126/science.1176827. [DOI] [PubMed] [Google Scholar]
- 90.Gribble FM. Alpha 2 A-adrenergic receptors and type 2 diabetes. N Engl J Med. 2010;362(4):361–362. doi: 10.1056/NEJMcibr0911499. [DOI] [PubMed] [Google Scholar]
- 91.Weigle DS, Koerker DJ, Goodner CJ. Pulsatile glucagon delivery enhances glucose production by perifused rat hepatocytes. Am J Physiol. 1984;247(4 Pt 1):E564–E568. doi: 10.1152/ajpendo.1984.247.4.E564. [DOI] [PubMed] [Google Scholar]
- 92.Lang DA, Matthews DR, Petro J, Turner RC. Cyclic oscillations of basal plasma glucose and insulin concentrations in human beings. N Engl J Med. 1979;301(19):1023–1027. doi: 10.1056/NEJM197911083011903. [DOI] [PubMed] [Google Scholar]
- 93.Jaspan JB, Lever E, Polonsky KS, van Cauter E. In vivo pulsatility of pancreatic islet peptides. Am J Physiol. 1986;251(2 Pt 1):E215–E226. doi: 10.1152/ajpendo.1986.251.2.E215. [DOI] [PubMed] [Google Scholar]
- 94.Lau T, Carlsson PO, Leung PS. Evidence for a local angiotensingenerating system and dose-dependent inhibition of glucose-stimulated insulin release by angiotensin II in isolated pancreatic islets. Diabetologia. 2004;47(2):240–248. doi: 10.1007/s00125-003-1295-1. [DOI] [PubMed] [Google Scholar]
- 95.Fliser D, Schaefer F, Schmid D, Veldhuis JD, Ritz E. Angiotensin II affects basal, pulsatile and glucose-stimulated insulin secretion in humans. Hypertension. 1997;30(5):1156–1161. doi: 10.1161/01.hyp.30.5.1156. [DOI] [PubMed] [Google Scholar]
- 96.Bernanke D, Epstein FH. Metabolism of the renal medulla. Am J Physiol. 1965;208:541–545. doi: 10.1152/ajplegacy.1965.208.3.541. [DOI] [PubMed] [Google Scholar]
- 97.Lee JB, Vance VK, Cahill GF., Jr Metabolism of C14-labeled substrates by rabbit kidney cortex and medulla. Am J Physiol. 1962;203:27–36. doi: 10.1152/ajplegacy.1962.203.1.27. [DOI] [PubMed] [Google Scholar]
- 98.Martinez-Maldonado M, Allen JC, Eknoyan G, Suki W, Schwartz A. Renal concentrating mechanism: possible role for sodium-potassium activated adenosine triphosphatase. Science. 1969;165(895):807–808. doi: 10.1126/science.165.3895.807. [DOI] [PubMed] [Google Scholar]
- 99.Leichtweiss H, Lubbers D, Weiss C, Baumgartl H, Resche W. The oxygen supply of the rat kidney: measurements of intrarenal pO2. Pfluegers Arch. 1969;309:328–349. doi: 10.1007/BF00587756. [DOI] [PubMed] [Google Scholar]
- 100.Aukland K, Krog J. Renal oxygen tension. Nature. 1960;188:671. doi: 10.1038/188671a0. [DOI] [PubMed] [Google Scholar]
- 101.Lilienfield LS, Rose JC, Lassen NA. Diverse distribution of red cells and albumin in the dog kidney. Circ Res. 1958;6(6):810–815. doi: 10.1161/01.res.6.6.810. [DOI] [PubMed] [Google Scholar]
- 102.Aoki TT, Vlachokosta FV, Foss MC, Meistas MT. Evidence for restoration of hepatic glucose processing in type I diabetes mellitus. J Clin Invest. 1983;71(4):837–839. doi: 10.1172/JCI110837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Jacobsson LT, Turesson C, Gulfe A, et al. Treatment with tumor necrosis factor blockers is associated with a lower incidence of first cardiovascular events in patients with rheumatoid arthritis. J Rheumatol. 2005;32(7):1213–1218. [PubMed] [Google Scholar]
- 104.Porter KE, Turner NA. Cardiac fibroblasts: at the heart of myocardial remodeling. Pharmacol Ther. 2009;123(2):255–278. doi: 10.1016/j.pharmthera.2009.05.002. [DOI] [PubMed] [Google Scholar]
- 105.Sekiguchi K, Li X, Coker M, et al. Cross-regulation between the renin-angiotensin system and inflammatory mediators in cardiac hypertrophy and failure. Cadiovasc Res. 2004;63(3):433–442. doi: 10.1016/j.cardiores.2004.02.005. [DOI] [PubMed] [Google Scholar]
- 106.Hwang MW, Matsumori A, Furukawa Y, et al. Neutralization of interleukin-1beta in the acute phase of myocardial infarction promotes the progression of left ventricular remodeling. J Am Coll Cardiol. 2001;38(5):1546–1553. doi: 10.1016/s0735-1097(01)01591-1. [DOI] [PubMed] [Google Scholar]
- 107.Flesch M, Höper A, Dell’Italia L, et al. Activation and functional significance of the reninangiotensin system in mice with cardiac restricted over-expression of tumor necrosis factor. Circulation. 2003;108(5):598–604. doi: 10.1161/01.CIR.0000081768.13378.BF. [DOI] [PubMed] [Google Scholar]
- 108.Nakamura K, Kusano K, Nakamura Y, et al. Carvedilol decreases elevated oxidative stress in human failing myocardium. Circulation. 2002;105(24):2867–2871. doi: 10.1161/01.cir.0000018605.14470.dd. [DOI] [PubMed] [Google Scholar]
- 109.Mochizuki M, Yano M, Oda T, et al. Scavenging free radicals by low-dose carvedilol prevents redox-dependent Ca2+ leak via stabilization of ryanodine receptor in heart failure. J Am Coll Cardiol. 2007;49(16):1722–1732. doi: 10.1016/j.jacc.2007.01.064. [DOI] [PubMed] [Google Scholar]
- 110.Delbosc S, Cristol JP, Descomps B, Mimran A, Jover B. Simvastatin prevents angiotensin II-induced cardiac alteration and oxidative stress. Hypertension. 2002;40(2):142–147. doi: 10.1161/01.hyp.0000024348.87637.6f. [DOI] [PubMed] [Google Scholar]
- 111.Moe GW, Marin-Garcia J, Konig A, Goldenthal M, Lu X, Feng Q. In vivo TNF-alpha inhibition ameliorates cardiac mitochondrial dysfunction, oxidative stress, and apoptosis in experimental heart failure. Am J Physiol Heart Circ Physiol. 2004;287(4):H1813–H1820. doi: 10.1152/ajpheart.00036.2004. [DOI] [PubMed] [Google Scholar]
- 112.Li S, Jiao X, Tao L, et al. Tumor necrosis factor-alpha in mechanic trauma plasma mediates cardiomyocyte apoptosis. Am J Physiol Heart Circ Physiol. 2007;293:H1847–H1852. doi: 10.1152/ajpheart.00578.2007. [DOI] [PubMed] [Google Scholar]
- 113.Valgimigli M, Merli E, Malagutti P, et al. Hydroxyl radical generation, levels of tumor necrosis factor-alpha, and progression to heart failure after acute myocardial infarction. J Am Coll Cardiol. 2004;43(11):2000–2008. doi: 10.1016/j.jacc.2004.01.036. [DOI] [PubMed] [Google Scholar]
- 114.Lerolle N, Bourgeois S, Leviel F, Lebrun G, Paillard M, Houillier P. Angiotensin II inhibits NaCl absorption in the rat medullary thick ascending limb. Am J Physiol Renal Physiol. 2004;287(3):F404–F410. doi: 10.1152/ajprenal.00265.2003. [DOI] [PubMed] [Google Scholar]
- 115.Shahid M, Francis J, Majid DS. Tumor necrosis factor-alpha induces renal vasoconstriction as well as natriuresis in mice. Am J Physiol Renal Physiol. 2008;295(6):F1836–F1844. doi: 10.1152/ajprenal.90297.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Ogawa S, Mori T, Nako K, Kato T, Takeuchi K, Ito S. Angiotensin II type 1 receptor blockers reduce oxidative stress markers in hypertensive diabetic nephropathy. Hypertension. 2006;47(4):699–705. doi: 10.1161/01.HYP.0000203826.15076.4b. [DOI] [PubMed] [Google Scholar]
- 117.Stokes MB, Foster K, Markowitz GS, et al. Development of glomerulonephritis during anti-TNF-alpha therapy for rheumatoid arthritis. Nephrol Dial Transplant. 2005;20(7):1400–1406. doi: 10.1093/ndt/gfh832. [DOI] [PubMed] [Google Scholar]
- 118.Hirsch IB, Brownlee M. Beyond hemoglobin A1c–need for additional markers of risk for diabetic microvascular complications. JAMA. 2010;303(22):2291–2292. doi: 10.1001/jama.2010.785. [DOI] [PubMed] [Google Scholar]