IF The National Kidney Foundation The Official Journal of

Atnerican Journal of Kidney Diseases

VOL XX, NO 6, DECEMBER 1992

IN-DEPTH REVIEW

Diabetic Nephropathy in Insulin-Dependent Patients Julia A. Breyer, MD • Oiabetic nephropathy is a serious complication of insulin-dependent diabetes mellitus (100M) that affects 30% to 40% of 100M patients with a predictable time of onset. Epidemiologic data suggest that either a genetic susceptibility, perhaps for hypertension (HTN), or an environmental exposure selects out that subset of 100M patients and destines them to develop diabetic nephropathy. Hopefully, assessing glomerular hyperfiltration, urinary albumin excretion rate (AER), glycemic control, mean arterial pressure (MAP), and perhaps early morphologic changes will allow early identification of this high-risk group of 100M patients before overt nephropathy is present. Once nephropathy appears, renal function inexorably declines, although the natural history of this progression may be changing with earlier therapeutic intervention. 100M patients with nephropathy suffer a high mortality rate compared with 100M patients without nephropathy or with nondiabetic end-stage renal disease patients. This is primarily due to malignant atherosclerotic disease manifested as coronary, peripheral, and cerebral arterial disease. Therapeutic interventions of demonstrated benefit in slowing the rate of decline of glomerular filtration rate (GFR) include blood pressure control and low-protein diets. Strict blood sugar control or treatment with aldose reductase inhibitors, converting enzyme inhibitors (eEls), or inhibitors of advanced glycosylation end-product formation are of possible benefit, but are awaiting clinical trial results. © 1992 by the National Kidney Foundation, Inc. INOEX WOROS: Oiabetic nephropathy; insulin-dependent diabetes; kidney failure.

D

IABETIC NEPHROPATHY is the single most common cause of end-stage renal disease (ESRO) in the United States, with diabetic patients accounting for 35% of all the patients enrolled in the Medicare ESRO program. 1 Patients with diabetes will soon account for nearly 50% of all renal failure patients if the current rate of increase continues. 2 In 1988, the cost to Medicare for ESRO patients with diabetes exceeded $1 billion dollars per year, while the amount spent in 1990 is projected to exceed $2 billion dollars per year.3 These Medicare ESRO data includes both patients with insulin-dependent diabetes (100M; type I) and those patients with non-insulin-dependent diabetes (NIOOM; type II). This review will focus on patients with 100M. More striking than the cost in dollars, is the cost in morbidity and mortality for patients with diabetic nephropathy. Patients with 100M and diabetic nephropathy as manifested by proteinuria (>0.5 g protein/24 h) have a 100-fold greater risk of dying relative to a nondiabetic population

(Fig 1).4,5 100M patients without proteinuria have only a twofold increase in their relative mortality. Of 1,475 patients diagnosed with diabetes before 1953, only 10% with nephropathy were alive 40 years after the onset of diabetes, while greater than 70% of the patients without renal disease were alive. 6 Although the overall mortality rate for patients with diabetic nephropathy is improving, death is just as likely to occur from macrovascular disease as from renal failure. 4 ,5,7-9 Oespite the impact of diabetic nephropathy on society and the individual, the epideFrom the Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN. Received May 21, 1992; accepted in revised form July 23, 1992. Address reprint requests to Julia A. Breyer, MD, Assistant Professor of Medicine, Division of Nephrology, Vanderbilt University Medical Center, S-3223 MCN, Nashville, TN 37232-2372. © 1992 by the National Kidney Foundation, Inc. 0272-6386/92/2006-0001$3.00/0

American Journal of Kidney Diseases, Vol XX. No 6 (December), 1992: pp 533-547

533

JULIA A. BREYER

534 125

o~ 11>;:

g-s ~ g.5.c g! g.

> 100 ::::i

II:

o

40

11>",

I-

;:

50

=i: "'0 ~~

75

:E

:::I",

w

u:;;

> 50

~

30 20 10

0-+---===--.----..------,----,

o

30 10 20 Duration of diabetes (years)

....I

~ 25

o

20

40 AGE (VEARS)

60

80

Fig 1. Relative mortality in IDDM patients with proteinuria (-) and without proteinuria (- - -). (Modified with permission.·)

miology, natural history, pathogenesis, and potential therapeutic interventions have only begun to be elucidated, leaving many basic questions unanswered. EPIDEMIOLOGY

Diabetic nephropathy rarely develops before 10 years' duration of diabetes. Approximately 40% ofIDDM patients have proteinuria after 40 years' duration of diabetes. The cumulative incidence of diabetic nephropathy in relation to duration of diabetes is depicted in Fig 2, and similar incidence patterns have been reported. 6,10-13 Interestingly, in two clinical cohorts of patients with IDDM, the cumulative incidence of proteinuria declined for patients whose diabetes was diagnosed in the 1950s compared with those diagnosed in the 1930s.4,10,11 The annual incidence of diabetic nephropathy peaks just before 20 years' duration of diabetes and thereafter declines (Fig 3).6,11 This pattern of increasing and decreasing annual incidence suggests the risk of developing diabetic nephropathy is not constant over the duration of diabetes and that a pool of susceptible patients becomes exhausted. While it is known that approximately 40% of IDDM patients will develop diabetic nephropathy, those patients who survive 35 years of diabetes without developing nephropathy are at extremely low risk of doing so in the future. These data suggest that either a genetic susceptibility or an environmental exposure may influence the risk of developing nephropathy, The reported familial clustering of

40

Fig 2. Cumulative incidence of diabetic nephropathy in relation to duration of diabetes in 907 IDDM patients. (Reprinted with permission. 5 Copyrlght © 1984 by American Diabetes Association, Inc.)

diabetic nephropathy is consistent with a genetically determined susceptibility.14,15 An increased incidence of albuminuria and end-stage renal disease (ESRD) secondary to diabetic nephropathy occurs in diabetic siblings of probands with diabetic nephropathy. Diabetic siblings of duration-matched probands without nephropathy have few renal complications. Although these data support a shared genetic susceptibility, the influence of common environmental factors has not been evaluated. In reviewing data reported to ESRD Network 17, blacks with diabetes (type I and type II) were found to have a 3.6-fold greater risk of developing ESRD than whites with diabetes. 16 This is comparable to the black to white incidence ratio of 2.6 reported in a Michigan study.17 Similarly, Mexican Americans and American Indians with diabetes have higher rates of diabetic nephropathy 4.0

0

;~

3.0

.t:/o

"'~ .>'O.c

am Q)c, oe C.c Q)c, 'OQ)

'u C

.E

2.0

1.0

0 0

30 20 10 Duration of diabetes (years)

40

Fig 3. Annual incidence of diabetic nephropathy in relation to duration of diabetes in 907 IDDM patients. (Reprinted with permission. 5 Copyright © 1984 by American Diabetes Association, Inc.)

535

DIABETIC NEPHROPATHY

than whites?,18 It is not known whether these racial differences are due to genetic, environmental, or other factors.

IDDM patients with GFR less than 125 mLI min and a 53% positive predictive value occurred in patients with GFR greater than 125 mL/min.22 However, the overlap occurring in the GFR of the patients who later develop nephropathy with those who do not, indicates an early measurement ofGFR alone cannot be the sole predictor of the development of renal disease. In contrast, in a small retrospective study and in a study where renal function was estimated only by serum creatinine, hyperfiltration was not found to predict the onset of nephropathy.23,24 Increases in GFR in patients with nephropathy seem to correlate with elevated erythrocyte sodium-lithium countertransport rates and increased glomerular surface areas (renal hypertrophy).25,26 Also, IDDM 'patients appear to have a much greater increase in GFR after amino acid infusions than normal subjects. 27 In animal models of diabetes, glomerular hyperfiltration is associated with increments in renal plasma flow , due to renal vasodilatation, and increases in the glomerular transcapillary hydraulic pressure gradient, which lead to glomerular sderosis?S The pathogenesis of the hyperfiltration is not clearly established, but the presence of the following conditions may contribute: hyperglycemia, insulin deficiency, augmented glucagon and growth hormone levels, increased ketone bodies, dietary protein excess, an altered renin angiotensin axis, altered prostaglandin production, glomerulopressin secretion, elevated kinin production, elevated atrial natriuretic factor levels, and volume expansion. 22 ,29-33

NATURAL HISTORY

At the time of diagnosis of diabetes, functional changes in the kidney are present in virtually all patients (Fig 4). Within a few years, morphologic changes occur in the kidneys of most IDDM patients. During this "silent period" when no overt disease is present, predictors of the development of diabetic renal disease such as hyperfiltration, microalbuminuria, hypertension (HTN), and poor glycemic control frequently occur. Approximately 17 years after the onset of IDDM, frank proteinuria is manifested in those 40% ofIDDM patients destined to develop overt diabetic nephropathy. Once proteinuria is established, renal function inexorably declines, with 50% of patients reaching ESRD within 7 years of the onset of proteinuria. Functional Changes

At the onset of diabetes, virtually all patients experience functional changes such as increased kidney size, albuminuria, which reverses with blood sugar control, and dramatically elevated glomerular filtration rates (GFR), which decrease with the initiation of insulin therapy. 19-21 In a retrospective study and a larger prospective study, IDDM patients had increased GFR long before the onset of incipient or overt nephropathy and the increased GFR predicted the later development of nephropathy.2o,22 A negative predictive value of 95% occurred in

r---INCIPIENT NEPHROPATHY----, Predictors? • Hyperfiltration • MicroaJbuminuria • Rising Blood Pressure

HTN

• Poor Glycemic Control

o I

2

I

5

I

~ Dia~ _________

Onset of

Fig 4_ The natural history of diabetic nephropathy.

Y

Time (years)

11-23

Onset of Proteinuria

FUNCTIONAL CHANGES

STRUCTURAL CHANGES

· i GFR



• Reversible Albuminuria

· i Kidney Size

i Glomerular Basement Memb. Thickening

• Mesangial Expansion

I

13-25

I

Rising Creatinine

15-27

I

End Stage Renal Failure

536

Morphologic Changes

At the onset of diabetes, renal biopsies are normal. However, within 1.5 to 2.5 years, glomerular basement membrane (GBM) thickening begins in virtually all patients with diabetes. 34 Five distinctive lesions can be recognized in the glomeruli of 100M patients in addition to GBM thickening: nodular (Kimmelstiel-Wilson) and diffuse forms of intercapillary glomerulosclerosis, the capsular drop lesion, the fibrin cap lesion, and mesangial expansion. 35 These changes, along with afferent and efferent arteriolar hyalinosis, increased renal extracellular membrane albumin, and IgG localization in the glomeruli, are diagnostic of diabetic nephropathy.36 Although, no correlation exists between the GBM thickening and clinical renal function, mesangial expansion does correlate with creatinine clearance as evidenced by decreased renal function associated with increased mesangial expansion. 34.37 100M patients with nephropathy demonstrate larger mean glomerular volumes and a greater percentage of sclerosed glomeruli than subjects with normal kidney function. 38 Although diabetes is predominantly a glomerular disease, altered structure and function of the renal tubulointerstitium are present. The degree of interstitial fibrosis correlates with the development of reduced GFR. Also, with the tubulointerstitium comprising the bulk of the kidney, the hypertrophy of the kidney in diabetes is contributed to by the expansion of the tubulointerstitium. 39 A variety of important tubular functional defects have been described, including impaired tubular reabsorption of low-molecular weight proteins (N-acetyliJ-D-glucosamidase) and albumin; increased sodium reabsorption leading to a volume expanded state; hypercalciuria; and impaired hydrogen and potassium excretion. 4o Oespite all the tubular and glomerular changes described, no one has yet demonstrated, in humans, that either a single renal biopsy or a series of renal biopsies can predict who will develop clinically significant diabetic nephropathy. The genesis of the morphological changes is not clearly established, yet transplant data do provide some clues. Normal kidneys transplanted into 100M patients develop diabetic morphologic changes, while diabetic kidneys transplanted into nondiabetic ESRO patients resolve their di-

JULIA A. BREYER

abetic lesions. 41 This strongly suggests that an intrinsic defect in the kidney is not the predisposing factor in the development of diabetic nephropathy, but rather something occurring in the diabetic milieu. Glomerular hemodynamic changes also appear to be critical for the development of diabetic nephropathy, since both in diabetic rats who have had their renal artery clipped and in humans with unilateral renal artery stenosis, the kidney distal to the blocked renal artery is protected from developing the morphologic changes of diabetes, while the contralateral unblocked kidney does develop diabetic nephropathy.42,43 Studies in animal models of diabetic nephropathy have demonstrated that maneuvers which alter glomerular hemodynamics affect the progression of the renal disease. 28 Although the diabetic milieu and exposure to the systemic circulation appear necessary for the development of the morphologic changes in the kidney, the pathogenesis of these changes on the molecular level is not clearly established and is beyond the scope of this review. Candidate defects include abnormal regulation of glycosaminoglycans, alteration of extracellular matrix, accumulation of advance glycosylation end-products, disturbed platelet functions, abnormal cytokine or growth factor production, abnormal aldose reductase activity, abnormal glomerular eicosanoid production, and inhibition of glomerular N-deacetylase and, hence, heparan sulfate biosynthesis. 39,44-50 Predictors

Renal hypertrophy, glomerular hyperfiltration, and specific morphologic changes are seen in many, if not most diabetic patients (Fig 4). However, none of these indicators alone identify the subset of 30% to 40% of 100M patients destined to develop clinical diabetic nephropathy. Being able to predict that subset of 100M patients would be an important tool and allow early preventive intervention. In an attempt to develop an early predictor of diabetic nephropathy, a sensitive radioimmunoassay for urinary albumin was developed to determine if subclinical elevations of urinary albumin excretion, in diabetic patients without frank proteinuria, would predict later development of proteinuria and diabetic ESRO. 51 There are currently multiple methods for determining albumin excretion rates (AERs) in a va-

537

DIABETIC NEPHROPATHY

riety of types of urine collections. Normal albumin excretion in the urine is less than 30 mg/24 h. Microalbuminuria is defined as an AER of 20 to 200 f.lg/min or 30 to 300 mg/24 h. Patients with microalbuminuria have a negative urine dipstick for protein and a normal 24-hour urine protein excretion measured in the usual manner. Overt dipstick proteinuria is defined as greater than 300 mg/24 h of albumin excretion. As shown in Table 1, transient causes of microalbuminuria such as hyperglycemia, HTN, congestive heart failure, urinary tract infection, and excessive physical exercise must be considered. If the AER is elevated in a single collection, persistent microalbuminuria needs to be documented by the occurrence of three elevated collections over 3 to 6 months. A single elevated collection is not adequate documentation. The reported prevalence of microalbuminuria in patients with 100M varies widely (3.7% to 23.0%), depending, at least in part, on how the population is selected in terms of the duration of diabetes or degree of microalbuminuria. 52·55 Duration of diabetes, glycemic control, and elevations in the mean arterial pressure (MAP) correlate with the presence of microalbuminuria. The presence of microalbuminuria in 100M patients is highly predictive of progression to clinical diabetic nephropathy during the next 10 to 15 years (Table 2).8,20,56,57 The lower level of microalbuminuria at which risk for diabetic nephropathy begins to increase has varied. GFRs in patients with microalbuminuria are well preserved or increased, but may begin to decrease once the AER exceeds 70 f.lg/min. The excretion of iJ2-microglobulin, a marker of tubular damage, does not increase until the AER exceeds 1,000 f.lg/min.58 Two studies have attempted to correlate micro albuminuria with the morphologic changes seen on biopsy, with conflicting reTable 1. Evaluation of Microalbuminuria

1. Test 100M patients of greater than 5 years' duration every 2 years. 2. Rule out causes of transient microalbuminuria: hyperglycemia, urinary tract infection, physical exercise, essential HTN, congestive heart failure, water loading. 3. If AER is elevated, repeat three times over 3 to 6 months to define persistent microalbuminuria.

Table 2. Predictive Value of Microalbuminuria

No. of Patients

Study

Parving, 198256

25

Viberti, 198630

63

Mogensen, 1984 Mathiesen, 1984

20

57

43 71

AER (!'g/min) Cutoff

% of Patients Developing Nephropathy

>28 30 15 70 0 (,)0 o

10

2

4 Years after onset of proteinuria

6

Fig 5. Cumulative incidence of coronary heart disease in patients with (e - e) and without (0 - 0) nephropathy. (Reprinted with permission.'02)

540

JULIA A. BREYER

Also, not only are there increased rates of coronary artery events, but also excessive cerebral and peripheral arterial complications in IDDM patients on dialysis or posttransplant. 9,104,105 Last, in addition to advanced atherosclerosis, malnutrition may play an important role in the increased mortality of the diabetic dialysis patient. A retrospective logistic regression of analysis of 12,000 hemodialysis patients found that the relative risk of diabetes fell after adjustment for decreased albumin.106 Thus, despite advances in renal replacement therapies, malnutrition, infection, and arthrosclerotic diseases shortens the life of the IDDM patient with ESRD. THERAPEUTIC INTERVENTION

The major therapeutic interventions that have been evaluated include antihypertensive therapy, improved diabetes control, treatment with aldose reductase inhibitors, treatment with inhibitors of the formation of advanced glycosylation endproducts, and the restriction of dietary proteins.

Antihypertensive Therapy Multiple studies have demonstrated that in IDDM patients with proteinuria and declining GFR, lowering MAP dramatically slows the rate of decline in renal function and improves patient survival (Fig 6.)107-111 Slowing the rate of decline in GFR for example from 0.89 mL/min/mo to 0.22 ml/min/mo delays ESRD by approximately 342 months in patients starting with a GFR of 100 mL/min (Fig 6). This dramatic slowing of the rate of decline in GFR occurs despite relatively modest blood pressure control (Fig 6). IDDM patients with microalbuminuria may be overtly hypertensive or may have MAP values that are not overtly hypertensive, but often are significantly higher than the MAP values in IDDM patients without nephropathy."2 Antihypertensive therapy in IDDM patients with microalbuminuria decreases AERs."2-"7 However, studies have not been completed to determine whether a reduction in microalbuminuria will lead to better-preserved GFR. Data from experiments in diabetic rats not only suggest that blood pressure control improves renal survival, but also that specific antihypertensive agents such as angiotensin converting enzyme inhibitors (CEls) have superior effects to other antihypertensive agents. This may be due

1.5

:;:E o

.Ec:

:g

1.0

§. 0:

u..

(!)

c:

.~ 0.5 "0

~

0.0

Mogenson '82

MAP post Rx (mmHg)

111

99

114

Fig 6. The rate of decline in GFR before the initiation of antihypertensive therapy (_) and after the initiation of antihypertensive therapy (~).107-109

to their unique effects on glomerular hemodynamics. 1l8 ,119 In diabetic rat models, increased glomerular capillary pressures contribute to the development of diabetic nephropathy, and lowering these glomerular pressures with CEls preserves renal structure and function. 1 19,120 In animal models, CEls have been effective despite the normal or reduced renin levels typically found in diabetic rats and humans. A number of studies in humans with IDDM have examined the effects of CEls on microalbumunirua, proteinuria, or preservation of GFR with conflicting resultS." 3,14,121-126 Many of these studies were hampered by small patient numbers, the absence of an appropriate control group, or the presence of concurrent illnesses in the patients studied. However, a meta-regression analysis of 77 experimental groups including controlled and uncontrolled studies in IDDM and NIDDM patients concluded that CEls had an ability to decrease proteinuria and to preserve GFR in patients with diabetic nephropathy that was uniquely independent of blood pressure changes. '27 In a study in which normotensive, microalbuminuric IDDM patients were given placebo or CEI, the group given CEI had a 5 mm Hg decrease in MAP and a significant decrease in AER. 113 This suggests the possibility that, even in normotensive IDDM patients, specific antihypertensive agents may be of benefit. The Col-

DIABETIC NEPHROPATHY

541

Table 3. Does Control of Hyperglycemia Alter the Course of Diabetic Nephropathy? No. of Subjects

Treatment Duration

Hyperfiltration

12

12 mo

Hyperfiltration with amino acid infusion Microalbuminuria

12

3wk

Jorgensen, 1992'39

Microalbuminuria

45

Feldt-Rasmussen 1986'40 Kroc, 1984'4' Reichard, 1991'42 Bending , 1984144

Microalbuminuria Microalbuminuria Microalbuminuria

Study

Wiseman, 1985'36 Tuttle, 1991'37 Viberti, 197938

Renal Function

Proteinuria

Glucose Control

Outcome

Remarks

HbA,

.j.GFR

None

Blood glucose

.j.GFR

Questionable blood glucose control

.j.AER

Short-term effects

7 yr

Blood glucose HbA,

.j.AER

36

2 yr

HbA,

.j.AER

Poor BS control, tAER ; Fair and good BS control, no change in AER BP different among groups

70 96

8 mo 5 yr

HbA, HbA,

.j.AER .j.AER

2 yr

HbA,

No effect

7

6

24 h

Only 1 AER at entry Changes relate to mean HbA, not treatment group Small number of subjeCts

Abbreviation: BS, blood sugar.

laborative Study Group of Angiotensin Converting Enzyme Inhibition in Oiabetic Nephropathy is performing an ongoing study designed to determine ifCEls are of unique benefit in preserving renal function in 100M patients with overt nephropathy. In this clinical trial, more than 400 100M patients with overt nephropathy (proteinuria > 0.5 g/24 h, serum creatinine < 221 1Lmol/L [2.5 mg/dL]) were randomly assigned to receive either placebo or captopril. Although both normotensive and hypertensive patients were enrolled, blood pressure goals in both the placebo and captopril groups were identical. 128 So few studies have been conducted examining the effects of calcium channel blockers in 100M patients with incipient or overt nephropathy that no firm conclusions can be drawn about the potential beneficial effects independent of blood pressure control. 1 1 6,129, 130 Antihypertensive therapy slows the rate of decline of renal function in 100M patients with overt nephropathy and is currently the therapeutic intervention with the most established and significant benefit. Important unanswered questions remain. Will the initiation of antihypertensive therapy in the early stage of incipient nephropathy prove to have a long-term beneficial impact? What is normal blood pressure in an 100M patient with nephropathy? Are established normotensive goals (140/90 mm Hg) too high for this group of patients? Are specific antihy-

pertensives or combinations of these of added benefit in IDOM patients with nephropathy? Improved Diabetes Control It is intuitively appealing to postulate that hyperglycemia is the cause of renal damage in diabetic patients, and its control will prevent the disease. Tight control of blood glucose prevents the development of and ameliorates established diabetic nephropathy in animal studies. 131 ,132 Multiple retrospective studies show an association between the development of diabetic nephropathy and poor blood sugar control. 11,79,80, 133·135 In 100M patients who have baseline hyperfiltration and renal hypertrophy or who have an abnormal increase in GFR and kidney size in response to amino acid infusions, short-term strict blood sugar control leads to reductions in kidney size and GFR. 136,137 Short-term strict blood sugar control can also decrease microalbuminuria. 138 Several studies suggest that long-term blood sugar control ameliorates microalbuminuria (Table 3).139.1 42 Criticisms of these trials include lack of stratification based on blood pressure, small sample size, inadequate duration of follow-up, inadequate measurement of entry AER, and varying start points in the disease process. The Oiabetes Complications and Control Trial now ongoing with 1,400 subjects is designed to determine if strict blood sugar control will prevent the development or progression of microalbuminu-

542

ria. 143 This trial, in a preliminary report, notes that the patients in the strict control group have a significantly greater number of severe hypoglycemic episodes. Last, once proteinuria and decreasing renal function are established, strict blood glucose coritrol is both difficult to achieve (due to frequent hypoglycemia) and without benefit for preserving renal function . 144 However, in a single uncontrolled study not designed to address this question, hyperglycemia in patients with established nephropathy contributed to their decline in GFR.145 Aldose Reductase Inhibitors

The tissues most affected by the diabetic disease process, including nerves, kidney, retina, and lens, do not require insulin for their uptake of glucose. Substantial evidence indicates that a significant proportion of the excess glucose in these tissues is metabolized via the polyol pathway. The enzymes that form the polyol pathway are aldose reductase and sorbitol dehydrogenase. Aldose reductase reduces the excess glucose to sorbitol, initiating a cascade of biochemical alterations. This leads to decreases of myo-inositol concentration in many tissues, including the glomerulus. It is postulated that inhibition of the aldose reductase enzyme may reverse or prevent progression of diabetic renal disease. In diabetic rats, aldose reductase inhibitors reverse proteinuria and decrease the rate of mesangial expansion.146.147 In diabetic patients, aldose reductase inhibition in preliminary studies decrease urinary albumin excretion rates. 148 Aldose reductase inhibitors are of potential, but clearly not established, benefit and are currently being studied in a multicenter clinical trial. Prevention of the Formation ofAdvanced Glycosylation End-Products

Hyperglycemia accelerates formation of nonenzymatic advanced glycosylation end-products in tissues and may playa major role in the pathogenesis of diabetic complications. Glucose forms chemically reversible early glycosylation products with proteins at a rate proportional to the glucose concentration. However, some of the early glycosylation products on collagen and other longlived proteins of the vessel walls do not dissociate and instead undergo a slow complex series of chemical rearrangements that form irreversible

JULIA A. BREYER

advanced glycosylation end-products. The levels of these products do not return to normal when hyperglycemia is corrected. 49 Glycation of albumin may contribute to its loss across the GBM, and the accumulation of glycosylated proteins in the glomerulus may stimulate mesangial growth and matrix formation. 49,149.1 52 Macrophages have receptors for these advanced glycosylation endproducts and trigger a series of cytokine events. In 100M patients, advanced glycosylation endproducts accumulate at a faster than normal rate. The increase in circulating advanced glycosylation end-product peptides also parallels the severity of renal functional impairment in diabetic nephropathy.153 Aminoguanidine, a nucleophilic hydrazine compound, prevents both the formation of advanced nonenzymatic glycosylation products and the formation of glucose-derived collagen cross-links in vivo and in vitro. 154 This drug inhibits cross-linking in diabetic rats, as well as inhibits the characteristic increase in GBM thickness seen morphologically in the diabetic rat. 49 Aminoguanidine reduces albuminuria and glomerular sclerosis in diabetic rat models. 155 ,156 Interestingly, aminoguanidine is also an inhibitor of nitric oxide formation and, in diabetic rats, it prevents diabetic vascular dysfunction. 157 Low-Protein Diets

In most experimental animal models of renal disease, high-protein intake accelerates the deterioration in renal fun ction. I 58 In diabetic rats, a high-protein diet increases glomerular blood flow and pressure and accelerates glomerular scaring. 159 Low-protein diets reduce glomerular pressures and preserve kidney function and structure. In 100M patients with no or incipient renal disease, low-protein diets reduce glomerular hyperfiltration. 160 In a study of eight 100M patients with microalbuminuria, alow-protein diet decreased the AER.161 The effects of the low-protein diet in 100M patients with overt nephropathy and declining renal function have also been examined.162-166 In the study with the largest group of patients observed for the longest duration, the rate of decline in GFR was 1.01 mL/ min/mo for patients on the high-protein diet and only 0.26 mL/min/mo for those on the low-protein diet. 166 Dietary restriction of protein can slow the progression of chronic renal failure substantially in patients with diabetic nephropathy.

543

DIABETIC NEPHROPATHY

Other Interventions A variety of other interventions have been attempted in small studies in either diabetic rats or humans with different degrees of renal involvement.

These include octreotide, antiplatelet drugs, pentoxyphyline, thromboxane synthetase inhibitors, camostat mesilate, high-linoleic acid diets, and eicosapentaenoic acid ethyl esters. 167-174

REFERENCES I. Health Care Financing Administration: ESRD Program Management and Medical Information System, March Update, Baltimore, MD, HCFA, 1990 2. Committee for the Study of the Medicare ESRD Program Division of Health Care Services: Kidney Failure and the Federal Government, in Rettig RA, Levinsky NG (eds): Institute of Medicine. Washington, DC, National Academy Press, Summer 1991 3. Health Care Financing Administration: End Stage Renal Disease Program Quarterly Statistical Summary. Baltimore, MD, HCFA, September 1990 4. Borch-Johnsen K, Andersen PK, Deckert T: The effect of proteinuria on relative mortality in type I (insulin-dependent) diabetes mellitus. Diabetologia 28:590-596, 1985 5. Dorman JS, Laporte RE, Kuller LH, et al: The Pittsburgh Insulin-Dependent Diabetes Mellitus (IDDM) Morbidity and Mortality Study. Diabetes 33:271-276, 1984 6. Andersen AR, Christiansen JS, Andersen JK, et al: Diabetic nephropathy in type I (insulin-dependent) diabetes: An epidemiological study. Diabetologia 25:496-50 I, 1983 7. Mathieson ER, Borch-Johnsen K, Jensen DV, et al: Improved survival in patients with diabetic nephropathy. Diabetologia 32:884-886, 1989 8. Messent JWC, Elliott TG, Hill RD, et al: Prognostic significance of microalbuminuria in insulin-dependent diabetes mellitus: A twenty-three year follow-up and study. Kidney Int 41 :836-839, 1992 9. Horl WH, Wanner C, Schollmeyer P, et al: Long-term outcome in diabetic patients on hemodialysis treatment. Contrib Nephrol 73: 163-169, 1989 10. Kofoed-Enevoldsen A, Borch-Johnsen K, Kreiner S, et al: Declining incidence of persistent proteinuria in type I (insulin-dependent) diabetic patients in Denmark. Diabetes 36:205-209, 1987 II. Krolewski AS, Warram JH, Christlieb R, et al: The changing natural history of nephropathy in type I diabetes. Am J Med 78:785-794, 1985 12. White P: Natural course and prognosis of juvenile diabetes. Diabetes 5:445-450, 1956 13 .• Pugh JA: The epidemiology of diabetic nephropathy. Diabetes Metab Rev 5:531-546, 1989 14. Seaquist ER, Goetz FC, Rich S, et al: Familial clustering of diabetic kidney disease-Evidence for genetic susceptibility to diabetic nephropathy. N Eng! J Med 320: 1161-1164, 1989 15. Borch-Johnsen K, Norgaard K, Hommel E, et al: Is diabetic nephropathy an inherited complication? Kidney Int 41:719-722, 1992 16. Stephens GW, Gillaspy JA, Clyne D, et al: Racial differences in the incidence of end-stage renal disease in types I and II diabetes mellitus. Am J Kidney Dis 15:562-567, 1990 17. Cowie CC, Port FK, Wolfe RA, et al: Disparities in incidence of diabetic end-stage renal disease according to type of diabetes. N Engl J Med 321:1074-1079,1989

18. Selby JV, FitzSimmons SC, Newman JM, et al: The natural history and epidemiology of diabetic nephropathyImplications for prevention and control. JAMA 263: 19541960, 1990 19. Mogensen CE, Osterby R, Gundersen HJG: Early functional and morphologic vascular renal consequences of the diabetic state. Diabetologia 17:71-76, 1979 20. Mogensen CE, Christensen CK: Predicting diabetic nephropathy in insulin-dependent patients. N Eng! J Med 311 :89-93, 1984 21. Mogensen CE: Renal function changes in diabetes. Diabetes 25:872-879, 1976 22. Rudberg S, Persson B, Dahlquist G: Increased glomerular filtration rate as a predictor of diabetic nephropathyAn 8-year prospective study. Kidney Int 41 :822-838, 1992 23. Lervang H-H, Jensen S, Brochner-Mortensen J, et al: Early glomerular hyperfiltration and the development oflate nephropathy in type I (insulin-dependent) diabetes mellitus. Diabetologia 31 :723-729, 1988 24. Jensen T, Borch-Johnsen K, Deckert T: Changes in blood pressure and renal function in patients with type I (insulin-dependent) diabetes mellitus prior to clinical diabetic nephropathy. Diabetes Res 4: 159-162, 1987 25. Carr S, Mbanya J-C, Thomas T, et al: Increase in glomerular filtration rate in patients with insulin-dependent diabetes and elevated erythrocyte sodium-lithium countertransport. N Engl J Med 322:500-505, 1990 26. Osterby R, Parving H-H, Nyberg G, et al: A strong correlation between glomerular filtration rate and filtration surface in diabetic nephropathy. Diabetologia 31 :265-270, 1988 27. Tuttle KR, Stein JH, DeFronzo RA: The natural history of diabetic nephropathy. Semin NephroII0:184-193, 1990 28. Hostetter TH, Troy JL, Brenner BM: Glomerular hemodynamics in experimental diabetes. Kidney Int 19:410415,1981 29. Hostetter TH: Pathogenesis of diabetic g!omerulopathy: Hemodynamic considerations. Semin Nephrol 10:219-227, 1990 30. Viberti GC, Wiseman MJ: The kidney in diabetes: Significance of early abnormalities. Clin Endocrinol Metab 15: 753-782, 1986 31. Fioretto P, Muollo B, Faronato PP, et al: Relationships among natriuresis, atrial natiuretic peptide and insulin in insulin-dependent diabetes. Kidney Int 41 :813-821, 1992 32. Correa-Rotter R, Hostetter TH, Rosenberg ME: Renin and angiotensinogen gene expression in experimental diabetes mellitus. Kidney Int 41:796-804, 1992 33. Jaffa A, Chai KX, Chao J, et al: Effects of diabetes and insulin on expression of kallikrein and renin genes in the kidney. Kidney Int 41:789-795, 1992 34. Mauer SM, Steffes MW, Ellis EN, et al: Structuralfunctional relationships in diabetic nephropathy. J Clin Invest 74:1143-1155, 1984

544 35. Reddi AS, Camerini-Davalos RA: Diabetic nephropathy-An update. Arch Intern Med 150:31-43, 1990 36. Mauer SM, Steffes MW, Goetz FC, et al: Diabetic nephropathy-A perspective. Diabetes 32:52-55, 1983 37. Steffes MW, Osterby R, Chavers B, et al: Perspectives in diabetes-Mesangial expansion as a central mechanism for loss of kidney function in diabetic patients. Diabetes 38: 10771081, 1989 38. Bilous RW, Mauer SM, Sutherland DER, et al: Mean glomerular volume and rate of development of diabetic nephropathy. Diabetes 38:1142-1147, 1989 39. Ziyadeh FN, Goldfarb S: The renal tubulointerstitium in diabetes mellitus. Kidney Int 39:464-475, 1991 40. Vaamonde CA, Perez GO: Tubular function in diabetes mellitus. Semin Nephrol 10:203-218, 1990 41. Abouna GM, Kremer GD, Daddah SK, et al: Reversal of diabetic nephropathy in human cadaveris kidneys after transplantation into non-diabetic recipients. Lancet 2:12741276, 1983 42. Berkman J, Rifkin H: Unilateral nodular diabetic glomerulosclerosis (Kimmelstiel-Wilson): Report of a case. Metabolism 22:715-722,1973 43. Mauer SM, Steffes MW, Azar S, et al: The effects of Goldblatt hypertension on development of the glomerular lesions of diabetes mellitus in the rat. Diabetes 27:738-744, 1978 44. Kofoed-Enevoldsen A: Inhibition of glomerular glucosaminyl N-deacetylase in diabetic rats. Kidney Int 41:763767, 1992 45. Noth RH, Krolewski AS, Kaysen GA, et al: Diabetic nephropathy: Hemodynamic basis and implications for disease management. Ann Intern Med 110:795-813, 1989 46. Deckert T, Horowitz 1M, Meyer TW, et al: Possible genetic defects in regulation of glycosaminoglycans in patients with diabetic nephropathy. Diabetes 40:764-770, 1991 47. Raskin P, Rosenstock J: Aldose reductase inhibitors and diabetic complications. Am J Med 83:298-306, 1987 48. Beyer-Mears A, Ku L, Cohen MP: Glomerular polyol accumulation in diabetics and its prevention by oral sorbinil. Diabetes 33:604-607, 1984 49. Brownlee M, Cerami A, Vlassara H: Advanced glycosylation end products in tissue and the biochemical basis of diabetic complication. N Engl J Med 318: 1315-1321, 1988 50. Donadio JV, I1strup DM, Holley KE, et al: Plateletinhibitor treatment of diabetic nephropathy: A IO-year prospective study. Mayo C1in Proc 63:3-15, 1986 51. Keen H, Chlowverakis C: An immunoassay method for urinary albumin at low concentrations. Lancet 2:913-916, 1963 52. Microalbuminuria Collaborative Study Group: Microalbuminuria in type I diabetic patients. Diabetes Care 15: 495-501,1992 53. McKenna MJ, Arias C, Feldkamp CS, et al: Microalbuminuria in clinical practice. Arch Intern Med 151: 17451747,1991 54. Parving H-H, Hommel E, Mathiesen E, et al: Prevalence of microalbuminuria, hypertension, retinopathy and neuropathy in patients with insulin-dependent diabetes. Br Med J 296:156-160, 1988 55. Norgaard K, Feldt-Rasmussen B, Borch-Johnsen K, et al: Prevalence of hypertension in type I (insulin-dependent) diabetes mellitus. Diabetologia 33:407-410, 1990

JULIA A. BREYER

56. Parving H-H, Oxenboll B, Svendsen PA, et al: Early detection of patients at risk of developing diabetic nephropathy. A longitudinal study of urinary albumin excretion. Acta Endocrinol 100:550-555, 1982 57. Mathiesen ER, Oxenboll B, Johansen K, et al: Incipient nephropathy in type I (insulin-dependent) diabetes. Diabetes 26:406-410, 1984 58. Mogensen CE: Microalbuminuria as a predictor of clinical diabetic nephropathy. Kidney Int 31:673-689, 1987 59. Chavers BM, Bilous RW, Ellis EN, et al: Glomerular lesions and urinary albumin excretion in type I diabetes without overt proteinuria. N Engl J Med 320:966-970, 1989 60. Walker JD, Close CF, Jones SL, et al: Glomerular structure in type I (insulin-dependent) diabetic patients with normo- and microalbuminuria. Kidney Int 41:741-748, 1992 61. Mogensen CE: Perspectives in diabetes-Prediction of clinical diabetic nephropathy in IDDM patients-Alternatives to microalbuminuria? Diabetes 39:761-767, 1990 62. Tuck ML, Corry DB: Pathophysiology and management of hypertension in diabetes. Annu Rev Med 42:533548,1991 63. Teuscher A, Egger M, Herman JB: Diabetes and hypertension-blood pressure in clinical diabetic patients and a control population. Arch Intern Med 149:1942-1945, 1989 64. Chase HP, Garg SK, Harris S, et al: High-normal blood pressure and early diabetic nephropathy. Arch Intern Med 150:639-641, 1990 65. Mogensen CE, Christensen CK: Blood pressure changes and renal function in incipient and overt diabetic nephropathy. Hypertension 7:11-64-11-73, 1985 66. Anderson S, Rennke HG, Garcia DL, et al: Short and long term effects of antihypertensive therapy in the diabetic rat. Kidney Int 36:526-536, 1989 67. Holm J, Hemmingsen L, Nielsen NV: Arterial blood pressure related to degree of albuminuria and low-molecularweight proteinuria in IDDM. Diabetes Care 13:443-446, 1990 68. Krolewski AS, Canessa M, Warram JH, et a1: Predisposition to hypertension and susceptibility to renal disease in insulin-dependent diabetes mellitus. N Engl J Med 318: 140145, 1988 69. Mangili R, Bending JJ, Scott G, et al: Increased sodiumlithium countertransport activity in red cells of patients with insulin-dependent diabetes and nephropathy. N Engl J Med 318:146-150,1988 70. Lopes de Faria JB, Friedman R, Tariq T, et al: Prevalence of raised sodium-lithium countertransport activity in type I diabetic patients. Kidney Int 41:877-882, 1992 71. Barzilay J, Warram JH, et al: Predisposition to hypertension: Risk factor for nephropathy and hypertension in IDDM. Kidney Int 41:723-730,1992 72. Jensen JS, Mathiesen ER, Norgaard K, et al: Increased blood pressure and erythrocyte sodium/lithium countertransport activity are not inherited in diabetic nephropathy. Diabetologia 33:619-624, 1990 73. Trevisan R, Nosadini R, Fioretto P, et al: Clustering of risk factors in hypertensive insulin-dependent diabetics with high sodium-lithium countertransport. Kidney Int 41 :855-861, 1992 74. Mauer SM, Sutherland DER, Steffes MW: Relationship of systemic blood pressure to nephropathy in insulin-dependent diabetes mellitus. Kidney Int 41:736-740, 1992

DIABETIC NEPHROPATHY

75. Hansen KW, Christensen CK, et al: Ambulatory blood pressure in microalbuminuric type I diabetic patients. Kidney Int 41:847-854, 1992 76. Ritz E, Hasslacher C, Guo JZ, et al: Role ofhypertension in diabetic nephropathy. Contrib Nephrol 73:91-101, 1989 77. Mathiesen ER, Ronn B, Jensen T, et al: Relationship between blood pressure and urinary albumin excretion in development of microalbuminuria Diabetes 39:245-259, 1990 78. Hasslacher C, Ritz E: Effect of control of diabetes mellitus on progression of renal failure. Kidney Int 32:S53-S56, 1987 (suppl) 79. Kussman MJ, Goldstein H, Gleason RE: The clinical course of diabetic nephropathy. JAMA 236:1861-1863, 1976 80. Orchard TJ, Dorman JS, Maser RE, et al: Factors associated with avoidance of severe complications after 25 years ofIDDM. Diabetes Care 13:741-747, 1990 81. Wilson DM, Luetscher JA: Plasma prorenin activity and complications in children with insulin-dependent diabetes mellitus. N Engl J Med 323:1101-1106, 1990 82. Myers BD: Pathophysiology of proteinuria in diabetic glomerular disease. J Hypertens 8:S41-S46, 1990 (suppl) 83. Deckert T, Feldt-Rasmussen B, et al: Glomerular size and charge selectivity in insulin-dependent diabetes mellitus. Kidney Int 33: 100-106, 1988 84. Kostraba IN, Klein R, et al: The Epidemiology ofDiabetes Complications Study. Am J Epidemiol 133:381-391, 1991 85. Mauer SM, Steffes MW, Ellis EN, et al: Can the insulindependent diabetic patient be managed without kidney biopsy? Symposium on Diabetic Nephropathy: Concepts of Pathogenesis and Treatment, in Robinson RR (ed): Proceedings, 1984 Congress in Nephrology. Springer-Verlag, New York, NY, 1984, pp 1103-1108 86. Parving H-H, Gall M-A, Skott P, et al: Prevalence and causes of albuminuria in non-insulin-dependent diabetic (NIDDM) patients. Kidney Int 37:243, 1990 (abstr) 87. Kussman Mj, Goldstein HH, Gleason RE: The clinical course of diabetic nephropathy. JAMA 236: 1861-1863, 1976 88. Jones RH, Mackay JD, Hayakawa H, et al: Progression of diabetic nephropathy. Lancet I: 1105-1106, 1979 89. Levey AS: Measurement of renal function in chronic renal disease. Kidney Int 38: 167-184, 1990 90. Groop L, Laasonen L, Edgren J: Renal papillary necrosis in patients with IDDM. Diabetes Care 12: 198-202, 1989 9J. Muhlhauser I, Toth G, Sawicki PT, et al: Severe hypoglycemia in type I diabetic patients with impaired kidney function. Diabetes Care 14:344-346, 1991 92. Villoria JG, Nunez JFM, Miralles JM, et al: Hyporeninemic hypoaldosteronism in diabetic patients with chronic renal failure. Am J Nephrol 8:127-137,1988 93. Stone WJ: Saturday conference: Renal disease in the juvenile diabetic. South Med J 72:203-207, 1979 94. Matson M, Kjellstrand CM: Long-term follow-up of 369 diabetic patients undergoing dialysis. Arch Intern Med 148:600-604, 1988 95. Grabensee B, Passlick-Deetjen J: Results of peritoneal dialysis in diabetics. Contrib Nephrol 73:183-198,1989 96. Sutherland DER, Fryd DS, Payne WO, et al: Kidney transplantation in diabetic patients. Transplant Proc 19:9094, 1987

545 97. Robertson RP, Sutherland DER: Pancreas transplantation as therapy for diabetes mellitus. Annu Rev Med 43: 395-415, 1992 98. Brekke IB, Holdaas H, Albrechtsen D, et al: Combined pancreatic and renal transplantation: Improved survival of uremic diabetic patients and renal grafts. Transplant Proc 22: 1580, 1990 99. Gruessner RWG, Dunn DL, Tzardis PJ, et al: Simultaneous pancreas and kidney transplants versus single kidney transplants and previous kidney transplants in uremic patients and single pancreas transplants in non uremic diabetic patients: Comparison of rejection, morbidity, and long-term outcome. Transplant Proc 22:622-623, 1990 100. Bilous RW, Mauer SM, Sutherland DER, et al: The effects of pancreas transplantation on the glomerular structure of renal allografts in patients with insulin-dependent diabetes. N Engl J Med 321 :80-85, 1989 10J. Krolewski AS, Kisinski EJ, Warram JH, et al: Magnitude and determinants of coronary artery disease in juvenileonset, insulin-dependent diabetes mellitus. Am J "Cardiol 59: 750-755, 1987 102. Jensen T, Borch-Johnsen K, Kofoed-Enevoldsen A, et al: Coronary heart disease in young type I (insulin-dependent) diabetic patients with and without diabetic nephropathy: Incidence and risk factors. Diabetologia 30: 144-148, 1987 103. Sampson MJ, Chambers lB, Sprigings DC, et al: Abnormal diastolic function in patients with type I diabetes and early nephropathy. Br Heart J 64:266-271, 1990 104. Lemmers MJ, Barry JM: Major role for arterial disease in morbidity and mortality after kidney transplantation in diabetic recipients. Diabetes Care 14:295-301, 1991 105. Abbott RD, Brand FN, Kannel WB: Epidemiology of some peripheral arterial findings in diabetic men and women: Experiences from the Framingham Study. Am J Med 88:376-381, 1990 106. Lowrie EG, Lew NL: Death risk in hemodialysis patients: The predictive value of commonly measured variables and an evaluation of death rate differences between facilities. Am J Kidney Dis 15:458-482, 1990 107. Mogensen CE: Long-term antihypertensive treatment inhibiting progression of diabetic nephropathy. Br Med J 285: 685-688, 1982 108. Parving H-H, Andersen AR, Smidt UM, et al: Effect of antihypertensive treatment on kidney function in diabetic nephropathy. Br Med J 294:1443-1447, 1987 109. Bjorck S, Nyberg G, Mulec H, et al: Beneficial effects of angiotensin converting enzyme inhibition on renal function in patients with diabetic nephropathy. Br Med J 293:471-474, 1986 110. Mathiesen ER, Borch-Johnsen K, Jensen DV, et al: Improved survival in patients with diabetic nephropathy. Diabetologia 32:884-886, 1989 III. Nyberg G, B10hme G, Norden G: Constant glomerular filtration rate in diabetic nephropathy. Acta Med Scand 219: 67-72, 1986 112. Christiansen CK, Mogensen CE: The course ofincipient diabetic nephropathy. Studies on albumin excretion and blood pressure. Diabetic Med 2:97-102, 1985 113. Marre M, Leblanc H, Suarez L, et al: Converting enzyme inhibition and kidney function in normotensive diabetic

546 patients with persistent microalbuminuria. Br Med J 294: 14481452, 1987 114. Bjorck S, Mulec H, Johnsen SA, et al: Contrasting effects of enalapril and metoprolol on proteinuria in diabetic nephropathy. Br Med J 300:904-907, 1990 115. Slomowitz LA, Bergamo R, Grosvenor M, et al: Enalapril reduces albumin excretion in diabetic patients with low levels of microalbuminuria. Am J Nephrol 10:457-462, 1990 116. Jerums G, Allen TJ, Tsalamandris C, et al: Angiotensin converting enzyme inhibition and calcium channel blockade in incipient diabetic nephropathy. Kidney Int 41: 904-911, 1992 117. Wiseman M, Viterti GC, Mackintosh D, et al: Glycemia, arterial pressure and microalbuminuria in type I (insulin-dependent) diabetes mellitus. Diabetologia 26:401-405, 1984 118. Zatz R, Meyer TW, Rennke HG, et aI: Predominance of hemodynamic rather than metabolic factors in the pathogenesis of diabetic glomerulopathy. Proc Nat! Acad Sci USA 82:5963-5967, 1985 119. Zatz R, Dunn RB, Meyer TW, et al: Prevention of diabetic glomerulopathy by pharmacological amelioration of glomerular capillary hypertension. J Clin Invest 77: 1925-1930, 1986 120. Hostetter TH, Troy JL, Brenner BM: Glomerular hemodynamics in experimental diabetes mellitus. Kidney Int 19:410-415, 1981 121. Pedersen MM, Hansen KW, Schmitz A, et al: Effects of ACE inhibition supplementary to beta blockers and diuretics in early diabetic nephropathy. Kidney Int 41:883-890, 1992 122. Mathiesen ER, Parving H-H, Hommel E: Converting enzyme-inhibition in diabetic nephropathy. Contrib Nephrol 73:160-162,1989 123. Hommel E, Parving H-H, Mathiesen E, et al: Effect of captopril on kidney function in insulin-dependent diabetic patients with nephropathy. Br Med J 293:467-470, 1986 124. Drummond K, Levy-Marchal C, et al: Enalapril does not alter renal function in normotensive, normoalbuminuric, hyperfiltering type I (insulin-dependent) diabetic children. Diabetologia 32:255-260, 1989 125. Parving H-H, Hommel E, Smidt UM: Protection of kidney function and decrease in albuminuria by captopril in insulin-dependent diabetics with nephropathy. Br Med J 297: 1086-1091,1988 126. Taguma Y, Kitamoto Y, Futaki G, et al: Effect of captopril on heavy proteinuria in azotemic diabetics. N Engl J Med 313:1617-1620,1985 127. Kasiske BL, Kalil RSN, Ma JZ, et aI: The effect of blood pressure treatment on the kidney in diabetes: A metaregression analysis. Ann Intern Med (in press) 128. Lemann J, Bidani AK, Bain RP, et al: Use of the serum creatinine to estimate glomerular filtration rate in health and early diabetic nephropathy. Am J Kidney Dis 16:236243,1990 129. Mimran A, Insua A, Ribstein J, et al: Comparative effect of captopril and nifedipine in normotensive patients with incipient diabetic nephropathy. Diabetes Care 11:850853, 1988 130. Melbourne Diabetic Nephropathy Study Group: Comparison between perindopril and nifedipine in hyperten-

JULIA A. BREYER

sive and normotensive diabetic patients with microalbuminuria. Br Med J 302:210-216, 1991 131. Rasch R: Prevention of diabetic g1omerulopathy in streptozotocin diabetic rats by insulin treatment. Diabetologia 18:413-416, 1980 132. Petersen J, Ross J, Rabkin R: Effect of insulin therapy on established diabetic nephropathy in rats. Diabetes 37:13461350, 1988 133. Mogensen CE: Prevention and treatment of renal disease in insulin-dependent diabetes mellitus. Semin Nephrol 10:260-273, 1990 134. Tchobroutsky G: Relation of diabetic control to development of microvascular complications. Diabetologia 15: 143-152, 1978 135. Hasslacher C, Ritz E: Effect of control of diabetes mellitus on progression of renal failure. Kidney Int 32:S53S56, 1987 (suppl) 136. Wiseman MJ, Saunders AJ, Keen H, et al: Effect of blood glucose control on increased glomerular filtration rate and kidney size in insulin-dependent diabetes. N Engl J Med 312:617-621,1985 137. Tuttle KR, Bruton JL, Perusek MC, et al: Effect of strict glycemic control on renal hemodynamic response to amino acids and renal enlargements in insulin-dependent diabetes mellitus. N Engl J Med 324:1626-1632,1991 138. Viberti GC, Pickup JC, Phil D, et al: Effect of control of blood glucose on urinary excretion of albumin and (32 microglobulin in insulin-dependent diabetes. N Engl J Med 300: 638-641, 1979 139. Jorgensen KD, Bjoro T, Kierulf P, et al: Long-term glycemic control and kidney function in insulin-dependent diabetes mellitus. Kidney Int 41 :920-923, 1992 140. Rasmussen BF, Mathiesen ER, Deckert T: Effect of two years of strict metabolic control on progression of incipient nephropathy in insulin-dependent diabetes. Lancet 2:13001304, 1986 141. Kroc Collaborative Study Group: Blood glucose control and the evolution of diabetic retinopathy and albuminuria. A preliminary multicenter trial. N Engl J Med 311:365-372, 1984 142. Reichard P, Berglund B, Britz A, et al: Intensified conventional insulin treatment retards the microvascular complications of insulin dependent diabetes (lDDM): The Stockholm Diabetes Intervention Study (SDIS) after five years. J Intern Med 230:101-108, 1991 143. The DCCT Research Group: Diabetes Control and Complications Trial (DCCT): Results of feasibility study. Diabetes Care 10:1-19, 1987 144. Bending JJ, Pickup JC, Viberti GC, et al: Glycaemic control in diabetic nephropathy. Br Med J 288:1187-1191, 1984 145. Nyberg G, Blohme G, Norden G: Impact of metabolic control on progression of clinical diabetic nephropathy. Diabetologia 30:82-86, 1987 146. Mauer SM, Steffes MW, Azar S, et al: Effects of sorbinil on glomerular structure and function in long-term diabetic rats. Diabetes 38:839-846, 1989 147. Beyer-Mears A, Murray FT, Del Val M, et aI: Reversal of proteinuria by sorbinil, an aldose reductase inhibitor, in spontaneously diabetic (BB) rats. Pharmacology 36:112-120, 1988

DIABETIC NEPHROPATHY

148. Blohme G, Smith V: Aldose reductase inhibition reduces urinary albumin excretion rate in incipient diabetic nephropathy. Diabetologia 32:467A, 1989 (abstr) 149. Narins RG:Diabetic nephropathy: Can the natural history be modified? Am J Med 90:21-70S-2A-75S, 1991 (suppl) 150. Vlassara H, Brownlee M, Cerami A: Advanced nonenzymatic tissue glycosylation: Significance in late diabetic complications. Diabetes Res Clin Pract 7:SI03-SI08, 1989 151. Cerami A, Vlassara H, Brownlee M: Role of advanced glycosylation products in complications of diabetes. Diabetes Care 11:73-79, 1988 152. Craighead JE: Is advanced basement membrane glycosylation reversible? Lab Invest 60:471-472, 1989 153. Makita Z, RadoffS, Rayfield EJ, et al: Advanced glycosylation end products in patients with diabetic nephropathy. N Engl J Med 325:836-842, 1991 154. Brownlee M, Vlassara H, Kooney A, et al: Aminoguanidine prevents diabetes-induced arterial wall protein crosslinking. Science 232:1629-1632, 1986 155. Yamashita K, Yoshikawa H, Bannai C, et al: Glycosylation of glomerular basement membrane (GBM) type IV collagen (lV-C) and proteinuria (PROT). Diabetes 38:25A, 1989 (abstr) 156. Iskandar SS, Patrick JD, Case LD: Protective effect of aminoguanidine against glomerululosclerosis in spontaneous diabetic mice. FASAB J 4:AI227, 1990 (abstr) 157. Corbett JA, Tilton RG, Chang K, et al: Aminoguanidine, a novel inhibitor of nitric oxide formation, prevents diabetic vascular dysfunction. Diabetes 41:552-556, 1992 158. Brenner BM, Meyer TW, Hostetter TH: Dietary protein intake and the progressive nature of kidney disease: The role of hemodynamically mediated glomerular injury in the pathogenesis of progressive glomerular sclerosis in aging, renal ablation, and intrinsic renal disease. N Engl J Med 307:652659, 1982 159. Hostetter TH, Meyer TW, Tennke HG, et al: Chronic effects of dietary protein in the rat with intact and reduced renal mass. Kidney Int 30:509-517, 1986 160. Kupin WL, Cotes P, Dumler F, et al: Effect on renal function of change from high to moderate protein intake in type 1 diabetic patients. Diabetes 36:73-79, 1987 161. Cohen D, Dodds R, Viberti G: Effect of protein restriction in insulin-dependent diabetics at risk of nephropathy. Br Med J 294:795-798, 1987

547 162. Viberti GC, Dodds RA, Bending JJ: Nonglycemic intervention in diabetic nephropathy: The role of dietary protein intake, in Mogensen CE (ed): The Kidney and Hypertension in Diabetes. Boston, MA, Nijhoff, 1988, pp 205-215 163. Barsotti G, Ciardella F, Morelli E, et al: Nutritional treatment of renal failure in type 1 diabetic nephropathy. Clin NephroI29:280-287, 1988 164. Evanoff G, Thompson C, Brown J, et al: Prolonged dietary protein restriction in diabetic nephropathy. Arch Intern Med 149:1129-1133, 1989 165. Walker JD, Bending JJ, Dodds, et al: Restriction of dietary protein and progression of renal failure diabetic nephropathy. Lancet 2:1411-1415,1989 166. Zeller K, Whittaker E, Sullivan L, et al: Effect ofrestricting dietary protein in the progression of renal failure in patients with insulin-dependent diabetes mellitus. N Engl J Med 324:78-84, 1991 167. Flyvbjerg A, Marshall SM, Frystyk J, et al: Octreotide administration in diabetic rats: Effects on renal hypertrophy and urinary albumin excretion. Kidney Int 41 :805-812, 1992 168. Serri 0, Beauregard H, Brazeau P, et al: Somatostatin analogue, octreotide, reduces increased glomerular filtration rate and kidney size in insulin-dependent diabetes. JAMA 265:888-892, 1991 169. Ferrari E, Fioravanti AL, Patti C, et al: Effects oflongterm treatment (4 years) with pentoxifylline on haemorheological changes and vascular complications in diabetic patients. Pharmatherapeutica 5:26-39, 1987 170. Hora K, Oguchi H, Furukawa T, et al: Effects of a selective thromboxane synthetase OKY-046 on experimental diabetic nephropathy. Nephron 56:297-305, 1990 171. Matsubara M, Taguma Y, Kurosawa KM, et al: Effect of camostat mesilate for the treatment of advanced diabetic nephropathy. J Lab Clin Med 116:206-210, 1990 172. Hamazaki T, Takazakura E, Osawa K, et al: Reduction in microalbuminuria in diabetics by eicosapentaenoic acid ethyl ester. Lipids 25:541-545, 1990 173. Aizawa T, Susuki T, Asawa T, et al: Dipyridamole reduces urinary albumin excretion in diabetic patients with normo- or microalbuminuria. Clin NephroI33:130-135, 1990 174. Barcelli VO, Weiss M, Beach D, et al: High linoleic acid diets ameliorate diabetic nephropathy in rats. Am J Kidney Dis 16:244-251,1990

Diabetic nephropathy in insulin-dependent patients.

Diabetic nephropathy is a serious complication of insulin-dependent diabetes mellitus (IDDM) that affects 30% to 40% of IDDM patients with a predictab...
1MB Sizes 0 Downloads 0 Views