U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Feingold KR, Anawalt B, Blackman MR, et al., editors. Endotext [Internet]. South Dartmouth (MA): MDText.com, Inc.; 2000-.

Cover of Endotext

Endotext [Internet].

Show details

Diabetic Kidney Disease

, MD, PhD, MSc and , MD, DMSc.

Author Information and Affiliations

Last Update: August 3, 2022.

ABSTRACT

Diabetes is the most common cause of end-stage kidney disease (ESKD) in the US and other developed countries. Diabetic nephropathy is a chronic condition characterized by a gradual increase in urinary albumin excretion, blood pressure levels and cardiovascular risk, and declining glomerular filtration rate (GFR), which can progress to ESKD. Chronic kidney disease (CKD) is common among patients with diabetes, and it develops in approximately 50% of the patients with type 1 diabetes (T1D) and 30% of those with type 2 diabetes (T2D). Patients with diabetes should be screened for CKD annually. Screening should include both albuminuria measurements and estimates of GFR. The kidney structural changes of diabetic nephropathy are unique to this disease, and closely correlate with kidney function. Multiple factors are associated with CKD in diabetes, and patients with diabetes often require multiple therapies aimed at prevention of progressive CKD and its associated co-morbidities and mortality. Management of cardiorenal risk factors, including lifestyle modifications (diet, exercise, and stop smoking), glucose, blood pressure and lipid control, use of agents blocking the renin angiotensin aldosterone system and use of SGLT2 inhibitors in patients with T2D and other agents with proven renal or cardiovascular benefit are the cornerstones of therapy. For complete coverage of all related areas of Endocrinology, please visit our on-line FREE web-text, WWW.ENDOTEXT.ORG.

INTRODUCTION AND EPIDEMIOLOGY

Diabetes and its complications are a substantial public health problem. In 2021, 10% of the global population (about 537 million adults) were living with diabetes (1). It is estimated that by 2045 this will rise to 784 million (1). Moreover, in a large proportion of patients, diabetes is undiagnosed. The estimates for the increased number of adults with diabetes vary largely according to the geographic region, going from a predicted 13% increase in Europe to a predicted 129% increase in Africa in the next 25 years (1), including a 24% increase in North America and Caribbean. It is estimated that over one in ten (37.3 million) Americans have diabetes, and one in three adult Americans (96 million Americans) have prediabetes (https://www.cdc.gov/diabetes/library/features/diabetes-stat-report.html).

While in populations of European origin, nearly all children and adolescents have type 1 diabetes (T1D), in certain populations (e.g., Japan), type 2 diabetes (T2D) is more common than T1D in this age group. Although the incidence of T1D is also increasing around the globe (2, 3), the rapid increase in the incidence of T2D among children and adolescents is alarming, and it has been linked to increased obesity rates and physical inactivity in this group.

Diabetes is associated with increased mortality and morbidity, and it is the main cause of incident end-stage kidney disease (ESKD) in the US and other developed countries (4). In the US alone, diabetes is responsible for more than 47% of the new ESKD cases. This is in large part due to T2D as most patients with diabetes have T2D rather than T1D. However, the proportion of individuals starting kidney replacement therapy due to diabetes varies significantly, ranging from 13% in China to 66% in Singapore (4). The likelihood of a patient with diabetes developing chronic kidney disease (CKD) is about 40% for patients with T1D and 30% for those with T2D, while the likelihood of a patient with diabetes developing ESKD is lower than that, as a large proportion of these die prematurely, especially from cardiovascular causes, before progressing to ESKD. ESKD is devastating to the individual and of enormous financial and social consequences.

PATHOPHYSIOLOGY

Diabetic nephropathy is a chronic condition that develops over many years. It is characterized by a gradual increase in urinary albumin excretion, blood pressure levels, and cardiovascular risk, declining glomerular filtration rate (GFR) and eventual ESKD. Diabetic nephropathy is associated with characteristic histopathological features (5, 6). About 25 to 50% of individuals with T1D (7, 8) and 45-57% of those with T2D (9-12) have progressively declining GFR with no or minimal albuminuria. Non-albuminuric renal impairment was the predominant phenotype among youth with T1D (13) and also among patients with T2D (14) in Italy, and a strong predictor of mortality (15). T1D patients with non-albuminuric CKD were older (8, 16) at evaluation and at T1D onset (16), were more often female (8, 16), had lower HbA1c (8, 16), total cholesterol, LDL-cholesterol, triglyceride levels (8), and serum uric acid levels (8, 16), had higher estimated GFR (eGFR) (8), were less often hypertensive (8, 16) and less likely to have retinopathy (8, 16) or to smoke (8, 16) than patients with albuminuric CKD (14, 15, 17, 18). HbA1c and blood pressure levels were higher and HDL-cholesterol was lower among non-albuminuric youth with type 1 diabetes and CKD as compared to patients with normal renal function (13). T2D patients with non-albuminuric CKD were also older (19), more often female (10, 11, 19), non-smokers (10, 11), Caucasian or Asian (10), had shorter diabetes duration (11), lower HbA1c (11), total cholesterol (12), LDL-cholesterol (12), triglyceride (11, 12), and systolic blood pressure levels (11, 12), higher eGFR (12, 19), and less often had retinopathy (11, 12) or a history of cardiovascular disease (11) than T2D patients with albuminuric CKD.

CKD in people with diabetes can be the result of diabetic nephropathy, other associated conditions such as hypertensive renal disease and obesity-related glomerulopathy, or other renal diseases, such as IgA nephropathy, focal segmental glomerulosclerosis, acute tubular necrosis, membranous nephropathy, among others (13-15). The frequency of other renal diseases depends, among others, on the prevalence of these conditions in the background population (see Excluding Other Causes of Kidney Disease below).

SCREENING, DIAGNOSIS, STAGES, AND MONITORING

Diabetic kidney disease, or CKD in diabetes, is diagnosed by measurements of kidney function. CKD diagnosis and staging in diabetes follows the same criteria as for patients without diabetes. In the clinical setting, CKD is classically diagnosed by estimates of GFR and measurements of urinary albumin. A decreased GFR indicates loss of filtration capacity, while an elevated albuminuria indicates that an abnormal (elevated) proportion of the albumin filtered by the kidneys is being eliminated in the urine, indicating changes in barrier selectivity.

Screening

Multiple guidelines recommend annual CKD screening of patients with diabetes, starting about 5 years after diagnosis in patients with T1D and at diagnosis in patients with T2D (20-22). Screening tests should include both albuminuria measurements and estimates of GFR.

ALBUMINURIA

Albuminuria screening should be undertaken when the person is free from acute illness and in reasonably stable glucose control, as acute illnesses and acute hyperglycemia can transiently increase albuminuria. Albuminuria may also increase in the upright posture and with exercise, thus measurements are best made in an early-morning urine sample; however, a spot urine sample is acceptable if there is no alternative. Because of the high day-to-day variation in urinary albumin excretion, if the first sample is abnormal, further samples should be obtained, ideally within 1–3 months. At least two out of three measurements should be abnormal before a diagnosis of albuminuria is made. First-morning void urinary albumin-to-creatinine ratio (ACR) measurement is the test of choice, as it is less cumbersome than timed urine collections and has lower day-to-day variability as compared to other methods (23).

GFR

In the clinical setting, GFR is estimated using equations that include patients’ age, sex, and serum creatinine. Serum creatinine should be measured annually, using an accredited assay standardized to the recommended isotope dilution mass spectrometry reference method (IDMS-traceable). Most laboratories currently calculate the eGFR using the serum creatinine CKD-EPI equation (https://www.mdcalc.com/ckd-epi-equations-glomerular-filtration-rate-gfr). Race is now optional on this equation, as its inclusion may or may not provide more precise estimates of GFR. The CKD-EPI equation estimates measured GFR more accurately than previous equations, particularly when GFR levels are greater than 60 mL/min/1.73 m2 (24). The CKD-EPI equation also categorizes risk of mortality and ESKD more accurately than the previous MDRD equation in a wide range of populations, including those with diabetes (25, 26). In elderly patients and in those with obesity, it has been suggested that equations based on creatinine lack precision, particularly in situations where weight loss is significant, as muscle mass usually changes without changes in eGFR (27).

Although there are data suggesting that GFR estimations based on cystatin C measurements may be slightly more precise than those based on serum creatinine (28), there is no agreement that cystatin C-based estimates are superior to creatinine-based GFR estimates (29, 30). Moreover, cystatin-C measurements are not interchangeable among laboratories, and not routinely available in the majority of the centers. The Kidney Disease: Improving Global Outcomes (KDIGO) guidelines recommend calculating cystatin-based eGFR in adults whose creatinine-based eGFR is 45–59 mL/min/1.73 m2 without other markers of kidney disease (31). Although this may help identify individuals with falsely reduced GFR, it is unclear if this approach improves the identification of individuals with progressive CKD compared with sequential measurements of creatinine-based eGFR. Recently, it was proposed the use of a CKD-Epi equation including both creatinine and cystatin C, and without race, for optimal precision when needed (32).

Diagnosis

CKD is diagnosed when two eGFR, at least 3 months apart, are <60 mL/min/1.73 m2 and/or 2 out of 3 albuminuria measurements are abnormal (ACR ≥ 30 mg/g creatinine). Diagnosis should be made in the absence of an acute serious illness (31).

CKD Stages

The 2020 KDIGO Clinical Practice Guideline for the Evaluation and Management of CKD advocates that final screening status should indicate both the GFR and albuminuria status (Tables 1 and 2) (22). The information can then be used as a measure of risk of progression to ESKD, and this classifier is also a good indicator of cardiovascular morbidity and mortality (Figure 1).

Table 1.

Glomerular Filtration Rate (GFR) Categories in Chronic Kidney Disease.

GFR categoryGFR (mL/min/1.73 m2)Description
G1≥90Normal or high
G260–89Mildly decreaseda
G3a45–59Mildly to moderately decreased
G3b30–44Moderately to severely decreased
G415–29Severely decreased
G5<15Kidney failure
a

Relative to young adult level.

Table 2.

Albuminuria Categories in Chronic Kidney Disease.

CategoryAER (mg/24 h)ACR (approximate equivalent)DescriptionPrevious terminology
mg/mmolmg/g
A1<30<3<30Normal to mildly increasedNormal
A230–3003–3030–300Moderately increasedaMicroalbuminuria
A3>300>30>300Severely increasedbProteinuria
a

Relative to young adult level.

b

Including nephrotic syndrome.

ACR, urine albumin:creatinine ratio; AER, albumin excretion rate.

Figure 1. . Classification and prognosis of chronic kidney disease by estimated glomerular filtration rate and albuminuria.

Figure 1.

Classification and prognosis of chronic kidney disease by estimated glomerular filtration rate and albuminuria. Source: Reprinted by permission from Macmillan Publishers Ltd: Kidney International, Levin A, Stevens PE (21), copyright 2014.

Monitoring Kidney Disease

Once urinary albumin excretion is abnormal, the ACR should be measured every 3 months and eGFR every 3–6 months, depending on the CKD stage (Figure 2).

Figure 2. . Risk of progression by intensity of coloring (green, yellow, orange, red, deep red).

Figure 2.

Risk of progression by intensity of coloring (green, yellow, orange, red, deep red). The numbers in the boxes are a guide to the frequency of monitoring (number of times per year). These are general parameters only based on expert opinion and must take into account underlying comorbid conditions and disease state, as well as the likelihood of impacting a change in management for any individual patient. CKD, chronic kidney disease; GFR, glomerular filtration rate. Source: Reprinted by permission from American Diabetes (252).

EXCLUDING OTHER TREATABLE CAUSES OF KIDNEY DISEASE

Excluding other causes of kidney disease is especially important among patients who do not follow the classical course of diabetic nephropathy disease progression. Diabetic nephropathy is a chronic disease, thus if acute decline in GFR is present, other causes should be sought. Other causes of kidney dysfunction should also be considered if proteinuria is present before 5 years of T1D duration, in the presence of active urinary sediment (acanthocytes, cellular casts, etc.), and if there are signs or symptoms of other systemic diseases. Retinopathy may or may not be present in patients with T2D and diabetic nephropathy. The frequency of other kidney diseases will also depend on the frequency of specific diseases (IgA nephropathy, for example) in the background (non-diabetic) population. Urinalysis, ultrasound of the kidney tract, measurement of autoantibodies and immunoglobulins, and kidney biopsy may help clarify the diagnosis. Studies evaluating the frequency of other kidney diseases in patients with diabetes indicate that the frequency of other diseases varies depending on the policy and on the reasons for a kidney biopsy (33-35). When kidney biopsies are done for research purposes, the frequency of other kidney disease is extremely low among patients with T1D without CKD (36, 37) and in Pima Indians with T2D (38).

STRUCTURAL KIDNEY LESIONS IN DIABETES

In patients with T1D, glomerular lesions can be demonstrated after diabetes has been present for a few years, while in T2D they can be present at diagnosis, probably reflecting delayed diagnosis. The changes in kidney structure caused by diabetes are specific, creating a pattern not seen in any other kidney disease. The severity of these diabetic lesions correlates with functional abnormalities (decreased GFR and albuminuria) (5, 6, 36) and it is also related to diabetes duration, glycemic control, and genetic factors. These later relationships are not precise and are in line with the marked variability in diabetic nephropathy susceptibility among patients with diabetes (see Relationships between Kidney Structure and Function below).

Light Microscopy

Renal hypertrophy, the earliest renal structural change in T1D, is not reflected in any specific light microscopy findings. In some patients, glomerular structure may remain normal or near normal for many decades, while others develop progressive disease. Early changes often include arteriolar hyalinosis, thickening of the glomerular basement membrane (GBM), and diffuse mesangial expansion (5, 6, 36). In about 40-50% of patients developing proteinuria, areas of extreme mesangial expansion called Kimmelstiel-Wilson nodules, or nodular mesangial expansion can be observed. Although Kimmelstiel-Wilson nodules are diagnostic of diabetic nephropathy, they are not necessary for severe renal dysfunction to develop. Global glomerulosclerosis can also be observed, especially with progressive disease (Figure 3). Atubular glomeruli and glomerulotubular junction abnormalities can also be present in proteinuric patients with T1D (39, 40). Tubular atrophy and interstitial fibrosis, common to most chronic renal disorders, can be present at later stages.

Figure 3. . Light microscopy photographs of glomeruli in sequential kidney biopsies performed at baseline and after 5 and 10 years of follow-up in a long-standing normoalbuminuric type 1 diabetic patient with progressive mesangial expansion and renal function deterioration.

Figure 3.

Light microscopy photographs of glomeruli in sequential kidney biopsies performed at baseline and after 5 and 10 years of follow-up in a long-standing normoalbuminuric type 1 diabetic patient with progressive mesangial expansion and renal function deterioration. A. Note the diffuse and nodular mesangial expansion and arteriolar hyalinosis in this glomerulus from a patient who was normotensive and normoalbuminuric at the time of this baseline biopsy, 21 years after diabetes onset [Periodic Acid Schiff (PAS) X 400]. B. 5-year follow-up biopsy showing worsening of the diffuse and nodular mesangial expansion and arteriolar hyalinosis in this now microalbuminuric patient with declining GFR (PAS X 400). C. 10-year follow-up biopsy showing more advanced diabetic glomerulopathy in this now proteinuric patient with further reduced GFR. Note also the multiple small glomerular probably efferent arterioles in the hilar region of this glomerulus (PAS X 400), and in the glomerulus in Fig. 3A above. Source: Reprinted with permission from National Kidney Foundation. Pathogenesis and Pathophysiology of Diabetic Nephropathy. Caramori ML, Mauer M. Primer on Kidney Diseases, 5th Edition, Greenberg A, et al., Copyright 2009 (253).

Immunofluorescence

Immunofluorescence findings include linear GBM and tubular basement membrane, as well as Bowman’s capsule, increased staining IgG (mainly IgG4), and albumin staining. The intensity of staining is not related to the severity of the underlying lesions.

Electron Microscopy

Using morphometric techniques, the first measurable diabetic nephropathy change is thickening of the GBM, which can be detected as early as 1 and 1/2 to 2 and 1/2 years after onset of type 1 diabetes (6, 41-44) (Figure 4). Tubular basement membrane thickening can also be detected, and it parallels GBM thickening (45). Increase in the relative area of the mesangium becomes measurable by 4-5 years (6, 36, 42). Immunohistochemical studies indicate that these changes in mesangium, GBM, and tubular basement membrane represent expansion of the intrinsic extracellular matrix components at these sites, likely including types IV and VI collagen, laminin, and fibronectin. Foot processes (podocyte) changes can be observed by electron microscopy, and the severity of these abnormalities has been associated with kidney function (46, 47). Changes in fenestrated endothelium have also been described in diabetes (47). Interstitial expansion is common to many kidney diseases. Early on in diabetes, interstitial expansion is associated with cellular alterations, while later in the disease process, when GFR is already reduced, there is increase in fibrillar collagen in the interstitium (48).

Figure 4. . Electron microscopy photographs of mesangial area in normal control (A) and in type 1 diabetic patient (B) [X 3,900].

Figure 4.

Electron microscopy photographs of mesangial area in normal control (A) and in type 1 diabetic patient (B) [X 3,900]. Note the increase in mesangial matrix and cell content, the glomerular basement membrane thickening and the decrease in the capillary luminal space in the diabetic patient (B). Source: Reprinted with permission from National Kidney Foundation. Pathogenesis and Pathophysiology of Diabetic Nephropathy. Caramori ML, Mauer M. Primer on Kidney Diseases, 5th Edition, Greenberg A, et al., Copyright 2009 (253).

While about 30% of patients with T2D and microalbuminuria who have had a kidney biopsy performed for research rather than clinical reasons had the classical diabetic nephropathy lesions described above, 41% have disproportionally severe interstitial fibrosis and tubular atrophy while the remaining 29% had minimal lesions with normal or near normal glomerular structure (49) (Figure 5).

Figure 5. . Light microscopy photographs of glomeruli of patients with type 1 (A) and type 2 diabetes (B-D).

Figure 5.

Light microscopy photographs of glomeruli of patients with type 1 (A) and type 2 diabetes (B-D). A. Diffuse and nodular mesangial expansion and arteriolar hyalinosis in this glomerulus from a microalbuminuric type 1 diabetic patient [Periodic Acid Schiff (PAS) X 400]. B. Normal or near normal renal structure in this glomerulus from a microalbuminuric type 2 diabetic patient (PAS X 400). This photograph was kindly provided by Dr. Paola Fioretto. C. Changes "typical" of diabetic nephropathology (glomerular, tubulo-interstitial and arteriolar changes occurring in parallel) in this renal biopsy from a microalbuminuric type 2 diabetic patient (PAS X 400). D. “Atypical" patterns of injury, with absent or only mild diabetic glomerular changes associated with disproportionately severe tubulo-interstitial changes. Note also a glomerulus undergoing glomerular sclerosis (PAS X 400). Source: Reprinted with permission from National Kidney Foundation. Pathogenesis and Pathophysiology of Diabetic Nephropathy. Caramori ML, Mauer M. Primer on Kidney Diseases, 5th Edition, Greenberg A, et al., Copyright 2009 (253).

RELATIONSHIPS BETWEEN KIDNEY STRUCTURE AND FUNCTION

In type 1 diabetes, the relationships between kidney structure and function are strong (5, 50, 51). Mesangial fractional volume and GBM width are inversely correlated with GFR, and directly correlated with albuminuria (5, 51) and blood pressure (51, 52). Importantly, GBM width is a strong independent predictor of progression to clinically advanced kidney disease among normoalbuminuric patients with T1D (53). Among these patients, global glomerular sclerosis (53, 54) and interstitial expansion (53, 55) are present and are additional independent predictors of GFR loss (53). Although increases in podocyte foot process width also correlates with albuminuria increases in T1D (56-58), our studies in patients with T1D who had no clinical manifestations of CKD at time of their research kidney biopsies indicate that podocyte parameters did not predict long-term progression to clinical CKD (59).

RISK FACTORS

Many factors are associated with CKD in diabetes. Associations may be with both albuminuria and GFR or with one measurement only. Factors that influence the initial development of kidney disease may not be the same as factors influencing progression. Duration of diabetes is one of the strongest risk factors for diabetic nephropathy, particularly in T1D.

Glucose Control

Glucose control is an important risk factor for the development and progression of diabetic nephropathy. Data from multiple observational and intervention studies in both T1D and T2D support this view (60). There is a strong positive association between HbA1c and incident CKD (eGFR <60 mL/min/1.73 m2), independent of other risk factors, and present even in the absence of albuminuria (61). Greater variability in HbA1c is independently associated with albuminuria and diabetic nephropathy (62-64), and variability in blood glucose levels as detected by continues glucose monitoring (CGM) has also been associated with complications (65, 66).

Blood Pressure

Blood pressure is critical in the development and progression of diabetic kidney disease. The excess prevalence of hypertension in T1D is confined to those with nephropathy (67). In young people with moderately elevated albuminuria, changes in blood pressure are subtle, perhaps manifesting only as reduced nocturnal diastolic blood pressure dipping (68). Once severely increased albuminuria is present, frank hypertension is present in 80% of patients, and is almost universal in ESKD. Variability in systolic and diastolic blood pressure independently predicts the development of albuminuria in T1D (62).

In T2D, the link between hypertension and kidney disease is less striking, perhaps due to the fact that hypertension is very common among these patients, present in 70-80% of the patients with T2D at the time of diagnosis. Almost all patients with moderately elevated albuminuria or worse have hypertension. In people with diabetic nephropathy, variability in systolic blood pressure is independently associated with the development of ESKD in patients with T1D (62) and T2D (69).

Other Metabolic Factors

Blood lipids, including triglycerides (70, 71), are associated with the development and progression of nephropathy, although the lipid phenotype alters as nephropathy progresses (72-74). Current smoking predicts the development of albuminuria (75). Insulin resistance increases the risk of albuminuria and rapid eGFR decline in patients with T1D (76) and of albuminuria in those with T2D (77). Individuals with T1D or T2D and nephropathy are more likely to have the metabolic syndrome (78, 79). Uric acid predicts the development of severely increased albuminuria (80) and decline in GFR as well as cardiovascular events (81). Probably this association is not causal as a reduction in uric acid by treatment with allopurinol could not slow GFR decline in patients with T1D (17).

Hyperfiltration

Hyperfiltration is common at onset of T1D and it is also present in some individuals at T2D diagnosis. GFR often returns to normal as glucose is controlled, but it may remain elevated in certain individuals. Whether individuals with persistent hyperfiltration are at increased diabetic nephropathy risk remains controversial (82-85). Sodium glucose cotransporter 2 inhibitors (SGLT2i) were introduced to lower glucose in T2D and have been demonstrated to slow progression of kidney disease (see below). A marked effect on hyperfiltration in T1D with SGLT2i was suggested to reflect lowering of intraglomerular hypertension and to support lowering of hyperfiltration as an important kidney protective measure (86). On the other hand, the results in T2D were less clear (87).

Genetic Factors

Genetic factors influence susceptibility to diabetic nephropathy (85, 86). If one sibling with T1D has nephropathy, the risk for the second sibling is increased 4–8 fold compared with siblings where neither have nephropathy (88). The clustering of conventional cardiovascular risk factors and cardiovascular disease (CVD) in people with diabetic nephropathy also occurs in their parents (89, 90). This suggests that the genetic susceptibility to nephropathy also influences the associated CVD. Research kidney biopsies in siblings with T1D also demonstrated heritability in the severity and patterns of renal lesions (91). Sodium-hydrogen antiport activity (92) and mRNA expression of catalase, an antioxidant enzyme associated with diabetic nephropathy risk, (93) were also found to be, at least in part, genetically regulated in siblings concordant for T1D. It is likely multiple genes are associated with DKD, and they can be either protective or deleterious. Moreover, different loci may influence albuminuria and GFR (94). Epigenetic modifications may also be important (95).

Ethnicity

In the Unites States, the prevalence of early CKD (defined as moderately elevated albuminuria or greater and eGFR<60 mL/min/1.73 m2) is higher in Latino and African American individuals than white people (96). A similar pattern is seen in Europe, where United Kingdom Afro-Caribbean and South Asian individuals more often have albuminuria and advanced CKD (stages 4-5) than white European individuals (97, 98). Albuminuria and CKD are also more common in Pima Indians (99) and in Māoris and Pacific Islanders (100, 101) than white Europeans. Reasons for this varying prevalence may include differing genetic influences and altered response to, or poorer access to, treatments.

Development of T2D in Youth

Individuals who develop T2D in youth have a high prevalence of hypertension and moderately elevated albuminuria (102). ESKD and death are particularly common in young people from ethnic minorities (103-105). However, in some of these populations, there is a high prevalence of non-diabetic kidney disease (106).

Albuminuria and GFR

Baseline albuminuria and eGFR independently influence the development and rate of progression of CKD (75, 107). Baseline albuminuria strongly predicts ESKD (108). Higher levels of albuminuria in the normoalbuminuric range (109, 110) and lower eGFR (111) predict a faster decline in eGFR. Conversely a short-term reduction in albuminuria with intervention suggest reduced progression of kidney and cardiovascular complications (112, 113).

Other Risk Factors

Other risk factors for nephropathy include pre-eclampsia (114), inflammatory markers (115, 116), cytokines and growth factors (117), periodontitis (118), and serum bilirubin levels (119, 120). Obstructive sleep apnea (121) and non-alcoholic fatty liver disease are both independently associated with diabetic nephropathy (122, 123). Circulating levels of tumor necrosis factor-α receptor 1 are independently associated with the cumulative risk of ESKD in T1D and T2D (124-126).

CO-MORBIDITIES AND ASSOCIATED COMPLICATIONS

The prognosis for people with diabetes and CKD is much poorer than for those without CKD. Both albuminuria and eGFR <60 mL/min/1.73 m2 (Figure 6 and 7) contribute independently and synergistically to the increased all-cause and cardiovascular risk (127-131).

Figure 6. . Declining glomerular filtration rate is associated with all-cause and cardiovascular mortality in individuals with and without diabetes.

Figure 6.

Declining glomerular filtration rate is associated with all-cause and cardiovascular mortality in individuals with and without diabetes. (A, B) All-cause mortality. (C, D) Cardiovascular mortality. Panels A and C use one reference point (diamond, eGFR of 95 mL/min per 1.73 m2 in the no diabetes group) for both individuals with and without diabetes to show the main effect of diabetes on risk. Panels B and D use separate references (diamonds) in the diabetes and no diabetes groups to assess interaction with diabetes specifically. Hazard ratios were adjusted for age, sex, race, smoking, history of cardiovascular disease, serum total cholesterol concentration, body-mass index, and albuminuria (log albumin-to-creatinine ratio, log protein-to-creatinine, or categorical dipstick proteinuria [negative, trace, 1+, ≥2+]). Blue and red circles denote p<0.05 as compared with the reference (diamond). Significant interaction between diabetes and eGFR is shown by x signs. eGFR=estimated glomerular filtration rate. Reproduced from Fox et al. 2012 (254), Copyright 2012, with permission from Elsevier.

Figure 7. . Increasing albuminuria is associated with all-cause and cardiovascular mortality in individuals with and without diabetes.

Figure 7.

Increasing albuminuria is associated with all-cause and cardiovascular mortality in individuals with and without diabetes. (A, B) All-cause mortality. (C, D) Cardiovascular mortality. Panels A and C use one reference point (diamond, ACR of 5 mg/g in the no diabetes group), for both individuals with and without hypertension to show the main effect of diabetes on risk. Panels B and D use separate references (diamonds) in the diabetes and no diabetes groups to assess interaction with diabetes specifically. Hazard ratios were adjusted for age, sex, race, smoking, history of cardiovascular disease, serum total cholesterol concentration, body-mass index, and estimated glomerular filtration rate. Blue and red circles denote p<0.05 as compared with the reference (diamond). Significant interaction between diabetes and ACR is shown by x signs. ACR=albumin-to-creatinine ratio. Reproduced from Fox et al. 2012 (254), Copyright 2012, with permission from Elsevier.

Association of Diabetic Kidney Disease with Cardiovascular Disease

TYPE 1 DIABETES

In T1D, the relative risk of premature mortality is 2–3-fold higher in moderately elevated albuminuria, 9-fold in severely increased albuminuria, and 18-fold in ESKD compared with the non-diabetic population (132). Individuals with T1D and normoalbuminuria do not have a higher risk of premature death (132, 133). CVD is 1.2-fold more common in people with moderately increased albuminuria (134) and 10-fold higher in those with severely increased albuminuria compared with those with normoalbuminuria (135). The cumulative incidence of CVD by the age of 40 years is 43% in people with T1D and severely increased albuminuria, compared with 7% in individuals with normoalbuminuria, with a 10-fold risk of coronary heart disease and stroke. In ESKD, the risk of CVD is even higher. Median survival on kidney replacement therapy is 3.84 years (136).

TYPE 2 DIABETES

In T2D, CVD risk is increased 2–4-fold with moderately increased albuminuria (137) and 9-fold in severely increased albuminuria (138). Once serum creatinine is outside the normal range, cardiovascular risk increases exponentially (139). Median survival from initiation of kidney replacement therapy is 2.16 years (136).

Microvascular Complications

Patients with diabetic nephropathy often have other microvascular complications. Significant retinopathy is almost always present in people with T1D and moderately elevated albuminuria or more. Progression of retinopathy and development of nephropathy each increases the risk for the other, supporting the notion of a common etiology (140). In people with T2D, the relationship is less strong (141). Those with classical nephropathy and progressively increasing albuminuria usually have significant retinopathy, and indeed moderately elevated albuminuria predicts the development and progression of retinopathy in T2D (142-144). In those with non-classical disease, retinopathy may be absent.

Peripheral neuropathy is also more common in diabetic nephropathy and associated with both albuminuria and declining GFR (144). Autonomic neuropathy, diagnosed by loss of nocturnal blood pressure dipping, occurs frequently (145, 146) and predicts kidney function decline (147).

PREVENTION AND TREATMENT

Although multiple strategies are now available to slow diabetic nephropathy progression, prevention of kidney disease remains crucial. The risk of developing diabetic nephropathy is particularly reduced by achievement and maintenance of good blood glucose and blood pressure control (22).

A guideline on management of diabetes in CKD from Kidney Disease Improving Global Outcomes (KDIGO) emphasize management of cardiorenal risk factors lifestyle factors (diet, exercise, and stop smoking), glucose, blood pressure, and lipids including blockade of the renin angiotensin aldosterone system and in T2D SGLT2 inhibition (Figure 8) (148).

Figure 8. . Putative promoters of progression of diabetic nephropathy.

Figure 8.

Putative promoters of progression of diabetic nephropathy. Source: Reproduced from Fox et al. 2012 (254), Copyright 2012, with permission from Elsevier.

Glucose Control

GLUCOSE CONTROL IN T1D

Among the participants in the DCCT who initially had normoalbuminuria, the relative risk reduction for development of moderately elevated albuminuria was 39% and for grade A3 (macroalbuminuria or proteinuria) 54% in those allocated to the intensively treated group compared with those in the conventionally managed group over the 6.5-year study (149). Mean achieved HbA1c was 7.0% and 9.1%, respectively. There is no HbA1c threshold below which risk is not reduced (150).

In the open follow-up of the DCCT cohort, the EDIC study, HbA1c in the previously intensive and conventional treatment groups became similar, ~8.0%. Despite this, the incidence of moderately and severely increased albuminuria grades (151), eGFR <60 mL/min/1.73 m2, and ESKD (151) were significantly reduced in those who had previously received intensive management, as summarized in Table 3. These results are supported by an observational study of individuals with T1D and CKD stages 1–3 with severely increased albuminuria at baseline (152). The cumulative risk of ESKD after 15 years was significantly lower in those whose HbA1c improved compared with those whose HbA1c remained stable or deteriorated. Hence improving glucose control significantly reduces the risk of development and progression of all stages of diabetic nephropathy in T1D. The beneficial effects extend far beyond the actual period of good glucose control, a phenomenon termed “metabolic memory.” In highly selected patients undergoing serial kidney biopsies after successful pancreas transplantation, kidney structural changes regressed after 10 but not 5 years (153). Thus, prolonged periods of “normoglycemia” are necessary to reverse kidney structural changes. It has been suggested that not only mean glycemic level as reflected by HbA1c, but also time in target glycemic range is important for the development of renal complications (154). In a small, study insulin pump therapy was associated with less variability compared to multiple daily insulin injections, and the reduced variability and improved time in range contributed to decline in albuminuria in T1D with increased albuminuria, beyond change in HbA1c (65).

Table 3.

Kidney Benefits of Intensive Insulin Therapy Demonstrated by the Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications Cohort.

ParameterDuration of observation (years)Conventional insulin therapyIntensive insulin therapy
Moderately elevated albuminuria815.8%6.8%
Severely increased albuminuria89.4%1.4%
eGFR <60 mL/min/1.73 m22246 (n)24 (n)
ESKD2216 (n)8 (n)

n, Number.

eGFR estimated glomerular filtration rate; ESKD, end-stage kidney disease.

Source: Data from (142, 145).

GLUCOSE CONTROL IN T2D

In the UKPDS, although the mean achieved HbA1c in the intensively managed group was 7.0% compared with 7.9% in the less strictly managed group, there was a 30% reduction in the relative risk of developing moderately or severely increased albuminuria after 9–12 years (155). No threshold of HbA1c and risk was observed, suggesting that the lower the HbA1c, the lower is the risk of nephropathy (156). In the open follow-up of the UKPDS cohort, HbA1c was similar in the previously intensively and conventionally managed groups after 1 year (157). Despite this, microvascular risk remained lower, confirming the “metabolic memory” seen in the DCCT/EDIC study. In the ADVANCE study, the HbA1c achieved in the intensively managed group was 6.5%, compared with 7.3% in the standard care group (158). In the intensive group there was a 9% relative risk reduction in new-onset moderately elevated albuminuria, a 30% reduction in the development of severely increased albuminuria, and a 65% reduction in ESKD over 5 years (159). The ACCORD study also demonstrated significant reductions in new onset moderately and severely increased albuminuria and of ESKD with intensive glucose management (160). Progression of albuminuria was reduced and regression increased. However, in those with CKD at baseline, the risk of all-cause and cardiovascular mortality was significantly increased in the intensive glucose management group (161). Hence the kidney benefits of extremely tight glucose control were outweighed by the excess mortality. A less intensive HbA1c target in individuals with T2D and duration >10 years seems sensible.

GLUCOSE CONTROL IN ESKD

Most (162-164) but not all (165) observational studies have demonstrated increasing all-cause and cardiovascular mortality with increasing HbA1c in people with diabetes on kidney replacement therapy. Some also showed a U-shaped relationship, with mortality increasing at low HbA1c levels (162, 164, 166). However, there have been no studies that demonstrated improved survival in patients with ESKD with improving glucose control. Among patients undergoing kidney transplant, improved allograft survival was demonstrated in patients with more strict blood glucose control (167).

Glucose Lowering Medications and Organ Protection

SGLT2 INHIBITORS

For over twenty years renin angiotensin system (RAS) blockade was the only recommended treatment for diabetic nephropathy. After many unsuccessful attempts in developing new therapies the first success has been with SGLT2 inhibitors. When initially tested for safety in cardiovascular outcome trials, empagliflozin showed not only a benefit on the primary endpoint major adverse cardiovascular events (168) but also a significant benefit on hospitalization for heart failure was also observed. In addition, a reduction in incident or worsening nephropathy occurred (HR 0.61; 95% CI, 0.53 to 0.70) (169). These findings were confirmed in cardiovascular outcome trials with canagliflozin, dapagliflozin and ertugliflozin (170). Importantly the benefits on kidney outcomes were independent of baseline eGFR from <45 ml/min/1.73m2 to >90 ml/min/1.73m2 and also independent of urinary albumin creatinine ratio <30mg/g, 30-300 or >300 mg/g (171). The first study with hard renal endpoints (end stage kidney disease, significant loss of renal function) as primary endpoint using a SGLT2 inhibitor was CREDENCE showing a major benefit on renal outcome, but also on heart failure and major adverse cardiovascular events in people with type 2 diabetes, urine albumin creatinine ratio >300 mg/g and eGFR 30-90 ml/min/1.73m2 (172). The primary outcome was a composite of end stage kidney disease, a doubling of the serum creatinine level, or death from renal or cardiovascular causes. The study was stopped early showing a benefit of canagliflozin with a HR 0.70; (95% CI, 0.59 to 0.82). These data were confirmed and extended by the DAPA-CKD study including subjects with chronic kidney disease with or without diabetes (173). EMPA-KIDNEY included participants with CKD with and without T2D as DAPA-CKD, but in addition to participants with albuminuria, EMPA-KIDNEY also included a group of study participants with impaired eGFR (20-45 mL/min/1.73m2) and normal albumin excretion (174). This study was recently stopped for positive findings which remain to be disclosed. Whereas SGLT2i’s were introduced to treat hyperglycemia, they also provide organ protection in diabetes with eGFR <45 mL/min/1.73m2 where there is no effect on blood glucose. Dapagliflozin and empagliflozin were also able to reduce heart failure hospitalization in people with heart failure with reduced ejection fraction (175), and empagliflozin was the first agent reported to reduce hospitalization for heart failure in people with heart failure with preserved ejection fraction, with similar benefit in those with and without diabetes (86, 176). In the DAPA-CKD study it was also demonstrated that dapagliflozin was able to reduce progression of CKD, hospitalization for heart failure and mortality in people with CKD with type 2 diabetes, but just as well in people with non-diabetic CKD (173) (Table 4).

Table 4.

Summary of SGLT2 Inhibitors on Renal Disease

NumberMean Follow-up (years)Hazard Ratio* (95% CI)P value
EMPA-REG
Empagliflozin
7,0203.10.54
(0.40-0.75
<0.001
CANVAS
Canagliflozin
10,1423.60.60
(0.47-0.77)
--
DECLARE-TIMI 58
Dapagliflozin
17,1604.20.53
(0.43-0.66)
<0.001
VERTIS-CV
Ertugliflozin
8,2463.00.81
(0.63-1.04)
0.08
CREDENCE
Canagliflozin
4,4012.60.66
(0.53-0.81)
<0.01
DAPA-HF
Dapagliflozin
4,7741.50.71
(0.44-1.16)
0.17
EMPEROR
Empagliflozin
3,7301.30.52
(0.32-0.77)
0.026
DAPA-CKD
Dapagliflozin
43042.40.56
(0.45-0.68)
<0.001
*

Renal composite outcomes Adapted from (177)

The explanation for the renal and cardiac benefits is not clear but multiple mechanisms have been suggested and probably glucose reduction is not very important. The inhibition of SGLT2 in the proximal tubule leads to blockade of glucose and sodium reabsorption, thus increasing distal tubular sodium delivery, which via macula densa and tubulo-glomerular feedback reduces intraglomerular pressure through constriction of the afferent glomerular arterioles. This is reflected clinically in the small dip in GFR when starting SGLT2i treatment and this mechanism has been suggested as the key mechanism behind the kidney protective effects. Reduction in blood pressure, body weight, increased uric acid excretion, and change in fuel metabolites have also been suggested to contribute (169). Blocking uptake of sodium in the proximal tubule has also been suggested to reduce oxygen consumption, thereby reducing hypoxia, leading to less inflammation and fibrosis in experimental studies and acute studies in humans were able to demonstrate improved renal oxygen availability (178).

In T2D with CKD metformin is recommended as first glucose lowering agent after lifestyle intervention, as in others with T2D, and then SGLT2 inhibitors are recommended independent of HbA1c for their organ protective effect, particularly in patients with albuminuria or heart failure (179, 180)(181) (Figure 9). In Europe the SGLT inhibitors sotagliflozin and dapagliflozin were initially approved for treatment of T1D, however the risk for normoglycemic diabetic ketoacidosis is increased compared to T2D and there are no studies of the kidney benefit in diabetic nephropathy in T1D. Currently, sotagliflozin is not marketed and the indication for dapagliflozin for treatment of T1D was stopped, and additional studies are needed to determine whether these agents can be safely used in patients with T1D to prevent CKD and cardiovascular progression.

Figure 9. . Patients with diabetes and CKD should be treated with a comprehensive strategy to reduce risks of kidney disease progression and cardiovascular disease Source: Reproduced with permission from Kidney Disease: Improving Global Outcomes (KDIGO) (172).

Figure 9.

Patients with diabetes and CKD should be treated with a comprehensive strategy to reduce risks of kidney disease progression and cardiovascular disease Source: Reproduced with permission from Kidney Disease: Improving Global Outcomes (KDIGO) (172).

GLUCAGON LIKE PEPTIDE 1 RECEPTOR AGONISTS

For some long-acting glucagon-like peptide-1 receptor agonists (GLP1-RA) (liraglutide, semaglutide, and dulaglutide) the cardiovascular outcome trials in type 2 diabetes demonstrated cardiovascular benefits, in subjects with already existing atherosclerotic CVD (180). The benefit on CVD outcomes was also demonstrated in CKD populations and thus GLP1-RA are recommended in the treatment of T2D with diabetic nephropathy when metformin and SGLT2 inhibition cannot control glucose (Figure 10). Studies also demonstrated positive kidney effects as secondary endpoints, mostly driven by reductions in albuminuria, but also some potential effects on eGFR. A kidney benefit was supported by the AWARD 7 study with dulaglutide in T2D with CKD although the primary endpoint was glycemic control (182). Semaglutide is being tested in the FLOW study (ClinicalTrials.gov NCT03819153) to determine whether it will confer benefits on hard renal and cardiovascular outcomes among participants with T2D when compared to placebo.

Figure 10. . Antihyperglycemic Therapies in Patients with Diabetes and CKD Source KDIGO guideline on management of diabetes in CKD Source: Reproduced with permission from Kidney Disease: Improving Global Outcomes (KDIGO) (172).

Figure 10.

Antihyperglycemic Therapies in Patients with Diabetes and CKD Source KDIGO guideline on management of diabetes in CKD Source: Reproduced with permission from Kidney Disease: Improving Global Outcomes (KDIGO) (172).

Blood Pressure Control

Rigorous blood pressure control improves the prognosis in diabetic nephropathy dramatically. Conservative estimates suggest that good blood pressure management doubles the time taken from first appearance of severely increased albuminuria to need for kidney replacement therapy, from a mean of 9 to 18 years. Improved management in moderately elevated albuminuria may prevent progression and promote regression normoalbuminuria. Blood pressure and blood glucose lowering effects are independent of one another but have synergistic effects (183, 184). In contrast to glucose “metabolic memory,” the benefits of blood pressure reduction are lost rapidly when control deteriorates (157).

TYPE 1 DIABETES

RAS inhibitors do not prevent moderately elevated albuminuria in normotensive people with T1D (37, 185, 186). There is also no evidence that control of hypertension in T1D and normoalbuminuria prevents progression of albuminuria and decline in kidney function. However, it seems highly likely.

Once moderately or severely increased albuminuria is present, inhibition of the RAS is the backbone of therapy, because it reduces intraglomerular pressure. A meta-analysis summarized the effects of ACE inhibitors in people with T1D and moderately elevated albuminuria (187). The odds ratio for progression to severely increased albuminuria was reduced by ACE inhibition to 0.35, and for regression to normoalbuminuria it increased to 3.07, compared with placebo treatment. After 2 years of treatment, the mean reduction in albumin excretion was 50.5% with ACE inhibition and it was greatest in those with highest baseline levels. However, the response to treatment plateaued with time, suggesting that treatment delays, rather than prevents, progression.

Addition of an ACE inhibitor to non-ACE inhibitor antihypertensive therapy reduced the risk of a doubling of the serum creatinine by 48% and the composite end-point of death, need for dialysis or kidney transplantation, by 50%, in people with T1D and with severely increased albuminuria and hypertension (188). Both benefits were independent of blood pressure. In short-term studies, the effects of angiotensin receptor blockers (ARBs) on blood pressure and urinary albumin excretion were similar to those of ACE inhibitors in T1D and severely increased albuminuria (189).

For a similar reduction in blood pressure, there is a greater reduction in protein excretion using ACE inhibitors compared with other classes of antihypertensive agents (190). This may be beneficial, as the passage of protein across the glomerular filtration barrier may accelerate the progression of nephropathy (191). Animal data show that this is due to preferential reduction in intraglomerular pressure with ACE inhibitors due to a dilatation of the efferent vessels (192). An effect on the filtration barrier has also been suggested (193).

RAS inhibitors should be offered to all individuals with T1D and albuminuria, regardless of blood pressure. The dose should be titrated up to the maximum recommended or tolerated, to obtain maximal antiproteinuric effect. If blood pressure remains >125/75 mmHg on maximum dose of RAS inhibitor, antihypertensive therapy should be intensified. Lower blood pressure reduces the rate of decline of GFR from 10–12 mL/min/year untreated to <5 mL/min/year (194). Regression from severely to moderately increased albuminuria can be achieved, with the fall in GFR reduced to <1 mL/min/year (71). The choice of agent should be made on an individual basis, as there is no evidence in T1D that any one add-on agent is better than any other. Often multiple agents are needed in CKD stage 3 and beyond.

TYPE 2 DIABETES

Control of hypertension reduces the risk of developing moderately or severely increased albuminuria (195-198). There may be a particular benefit of RAS inhibition in prevention of nephropathy (199-201) but lowering blood pressure sufficiently is the key. Achieved blood pressure in these studies was generally ~140/80 mmHg, but most guidelines now suggest a blood pressure target of 130/80 mmHg in T2D (20, 21).

As with T1D, there is good evidence in T2D that inhibition of the RAS should be the backbone of therapy if albuminuria is elevated. RAS blockade reduces progression of moderately elevated albuminuria to severely increased albuminuria (196, 202) and increases regression to normoalbuminuria (202). The benefits are at least partly independent of blood pressure lowering. In more advanced diabetic nephropathy, RAS inhibition with ARB reduces progression, defined as doubling of serum creatinine, ESKD, or death (203, 204). Hence people with T2D and moderately or severely increased albuminuria should be prescribed a RAS inhibitor, titrated to the maximum tolerated dose (205). Hyperkalemia is common in individuals with T2D and nephropathy taking an ARB and is associated with increased risk of kidney failure (206). General steps to lower potassium such as dietary advice, diuretics, discontinuation of other medications or dietary supplements which might be increasing potassium levels, or potassium binders should be considered before stopping RAS blockade (179). Introduction of a RAS inhibitor often leads to an acute decline in GFR, which then stabilizes. Individuals with the greatest initial fall in GFR have the slowest subsequent decline in kidney function (207).

Most people with T2D and albuminuria will require additional antihypertensive therapy. The choice of additional agents should be made on an individual basis, with diuretics and calcium channel blockers often being appropriate. In resistant hypertension with preserved renal function mineralocorticoid receptor antagonists may be useful (208).

In the UKPDS, there was no blood pressure level below which risk of developing moderately elevated albuminuria or beyond increased, i.e., no “J” shape (209). The ADVANCE study explored the effects of reduction of blood pressure below the currently recommended targets of 130/80 mmHg in individuals with normal or moderately increased albuminuria and 125/75 mmHg in those with severely increased albuminuria (210). Over 4 years, the risk of kidney events was reduced by 21%, mainly because of reduced risk of developing moderately or severely elevated albuminuria. However, an achieved systolic blood pressure below 120–130 mmHg was associated with increased mortality and ESKD (211). Therefore, extremely tight blood pressure control should be avoided.

DUAL BLOCKADE OF THE RAS

Addition of an ARB to an ACE inhibitor (212, 213) or of the direct renin inhibitor aliskiren to an ARB reduces blood pressure and albuminuria more than each agent individually. However, in the longer term, dual blockade increases the risk of hyperkalemia, hypotension, and acute, irreversible kidney failure (214-217). Hence dual blockade is not recommended.

MINERALOCORTICOID RECEPTOR ANTAGONISM

Prevention of diabetic nephropathy was attempted in the PRIORITY trial including T2D with normoalbuminuria. High risk for progression to CKD/moderately elevated albuminuria was identified with a urinary proteomic based risk score (CKD-273). High risk individuals were randomized to spironolactone or placebo, and although the biomarker predicted progression of kidney disease, spironolactone was not able to reduce progression compared to placebo over three years (218).

Short term studies in established diabetic nephropathy revealed ~30% reduction in albuminuria with the steroidal mineralocorticoid receptor antagonists (MRAs) spironolactone or eplerenone (219). Preventing over activation of the mineralocorticoid receptor reduces inflammation and fibrosis, but due to potassium problems, diabetes with kidney disease became a contraindication for these agents. Non-steroidal MRAs have been developed and may cause less potassium issues. The non-steroidal MRAs esaxerenone and finerenone reduced moderately elevated albuminuria in T2D in short term studies with a good safety profile with very little hyperkalemia (220, 221). This led to two large studies testing finerenone in T2D with CKD.

FIDELIO-DKD tested finerenone on a background or RAS blockade with an angiotensin converting enzyme inhibitor (ACEi) or ARB and included 5734 subjects with relatively advanced CKD and T2D (UACR ≥30–≤5000 mg/g, eGFR ≥25–<75 mL/min/1.73 m2 and the primary endpoint (kidney failure, sustained decrease of eGFR ≥40% or kidney death) was reduced with a hazard rate (HR) 0.82 (95%CI 0.73-0.93, p=0.001). The key secondary outcome (cardiovascular death, myocardial infarction, stroke, or hospitalization for heart failure) was also reduced (HR: 0.86; 95% CI 0.75–0.99; p=0.03). The incidence of hyperkalemia-related treatment discontinuation was rare, but higher with finerenone than placebo (2.3% and 0.9%, respectively) (222).

FIGARO-DKD also tested finerenone, but included patients with T2D with less advanced CKD, including a greater number of patients with albuminuria in the range 30-300 and impaired eGFR or albuminuria >300 with normal eGFR. FIGARO-DKD was a randomized double-blind phase III study of CV morbidity and mortality, and the primary endpoint was time to first occurrence of CV death, nonfatal myocardial infarction (MI), nonfatal stroke, or hospitalization for HF. The key secondary composite outcome was time to kidney failure, sustained ≥40% decrease in eGFR from baseline, or renal death (223). The study randomized 7437 patients, and the results demonstrated a significant reduction in the primary CV composite endpoint with finerenone compared with placebo (HR: 0.87; 95% CI, 0.76–0.98; P = 0.03). The effect on the ≥40% kidney composite endpoint was not significant with finerenone versus placebo (HR: 0.87; 95% CI, 0.76–1.01 P = 0.07) (223). However, the standard kidney composite endpoint with a ≥57% decline in eGFR (equivalent to doubling of serum creatinine) instead of the ≥40% decline in eGFR was significantly reduced with finerenone compared with placebo (HR: 0.77; 95% CI, 0.60–0.99; P = 0.04) (223).

Finerenone has now been approved for treatment of CKD in T2D by FDA, and will thus be a new opportunity for treatment of diabetic nephropathy. It is not clear where finerenone will be placed in guidelines compared to SGLT2i, but a subgroup analysis from FIDELIOIO-DKD suggest that finerenone is just as efficient when added to SGLT2i and thus it will be interesting to study if the combination provides added benefit (224).

SODIUM INTAKE

Short-term dietary sodium restriction (target sodium intake 50 mmol or 1150 mg Na+ per day), added to RAS blockade, reduces albuminuria (225). The treatment effects of ARB are greater in patients with lower rather than higher dietary sodium intake (226). Hence dietary counselling to reduce sodium intake is essential and an intake of <2 g of sodium per day (or <90 mmol or 2070 mg of sodium per day, or <5 g of sodium chloride per day) is recommended (179).

NON-CLASSICAL DIABETIC KIDNEY DISEASE

There is no specific evidence for the use of RAS inhibition in individuals without albuminuria. However, control of blood pressure remains crucial to slow progression. Ongoing studies are investigating the effect of the SGLT2 inhibitor empagliflozin on CKD including low eGFR (20-45 ml/min/1.73m2) but normal urinary albumin excretion (227).

Endothelin Receptor Antagonists

Atrasentan is an endothelin receptor A antagonist which demonstrated ability to lower proteinuria without significant edema (228). Previously edema had been a concern with this class of agents (229). The SONAR study tested atrasentan in T2D with severely increased albuminuria with progression of kidney disease, ESKD and mortality as the primary outcome (230). Although stopped early for concern of futility, the study eventually showed a kidney benefit of the same magnitude as with the SGLT inhibitors, but without effect on major adverse cardiovascular events and with a tendency to increased risk of heart failure. The primary endpoint was a composite of doubling of serum creatinine (sustained for ≥30 days) or end-stage kidney disease (eGFR <15 mL/min per 1.73 m2 sustained for ≥90 days, chronic dialysis for ≥90 days, kidney transplantation, or death from kidney failure). The hazard ratio for atrasentan compared to placebo was 0.65 (95% CI 0.49 to 0.88) p=0·0047). The mode of action may relate to an effect on inflammation, but also an effect on podocytes and endothelium and glycocalyx has been proposed from experimental data (231).

Low-Protein Diet

A meta-analysis concluded that a low protein diet significantly improves GFR but not albuminuria, across all subtypes of diabetes and stages of nephropathy (232). A randomized trial of 82 patients with T1D,, severely increased proteinuria and progressive loss of kidney function demonstrated reduced mortality and ESKD (relative risk 0.23; 95% CI 0.07 to 0.72) for patients assigned to a low-protein diet targeting 0.8 g protein/kg body weight/day compared to usual diet (233). Protein intake should not be restricted to less than 0.7 g protein/kg body weight/day because of concerns about malnutrition in ESKD. In line with recommendations for the general population a protein intake of 0.8 g protein/kg body weight/day is recommended for diabetes and CKD, except for people on peritoneal dialysis where a higher intake (1.0-1.2 g protein/kg body weight/day is recommended (179).

Lipids

In diabetic nephropathy lipid lowering medications are recommended to reduce the risk for CVD. There is some evidence that lipid-lowering agents are beneficial to the kidney. In a post hoc analysis of the Collaborative Atorvastatin Diabetes Study, the rate of decline of eGFR was significantly less in those individuals taking atorvastatin 10 mg daily compared with placebo. Fibrates also reduce albuminuria, although they reversibly increase serum creatinine (234).

Cardiovascular Risk—Other Factors

Smoking increases the likelihood for development of diabetic nephropathy as discussed above. There have been no good trials of smoking cessation. However, smoking cessation should clearly be encouraged. There are no studies in diabetic kidney disease with aspirin evaluating long term benefits although short term studies suggest no effect on urinary albumin excretion or GFR (235). In many individuals with established CVD or high risk for CVD aspirin should be considered for prevention of cardiovascular events. There is an increased risk for atrial fibrillation in diabetes and in CKD, and higher morbidity and mortality associated with thromboembolic events including stroke in diabetes with atrial fibrillation (236). In diabetes with atrial fibrillation anticoagulation is often recommended, and direct oral anticoagulants are usually preferred compared to vitamin K antagonists. In addition to a reduced risk for bleeding and similar or better effects on reducing risk for thrombosis, observational studies suggest reduction in progression of CKD. Thus, a recent study using a health claim database included patients with nonvalvular atrial fibrillation and diabetes that newly initiated rivaroxaban (N=10,017) or warfarin (N=11,665) (237). Patients were matched using propensity scores. In comparison to warfarin, rivaroxaban was associated with lower risks of acute kidney injury events (HR: 0.83; 95% CI, 0.74 to 0.92) and development of stage 5 CKD or need for hemodialysis (HR: 0.82; 95% CI, 0.70 to 0.96) (237). The mechanism could be reduced vascular calcification but needs to be confirmed in randomized controlled trials.

Weight Loss

In a trial comparing intensive lifestyle intervention with diabetes support and education in T2D, individuals randomized to intensive lifestyle modification were less likely to develop CKD over 8 years (238). The effect was partly attributable to reductions in body weight, HbA1c, and systolic blood pressure. Low carbohydrate, Mediterranean, and low-fat diets have similar beneficial effects on change in eGFR and albuminuria over 2 years (239). In individuals with T2D who have undergone bariatric surgery, moderately and severely increased albuminuria regresses to normoalbuminuria (240). Similar benefits were described in a 5-year study in severely obese adolescents with and without T2D (241).

FURTHER MANAGEMENT OF CHRONIC KIDNEY DISEASE STAGE 3 OR POORER

Monitoring Anemia and Bone Chemistry

In progressive CKD from stage 3 onwards, bone chemistry, full blood count, and iron stores should be assessed every 3–6 months.

Monitoring Glucose Control

Red blood cell and protein turnover are abnormal in CKD, making the interpretation of HbA1c, glycated albumin, and fructosamine results difficult, particularly in subjects with CKD 4+. Thus, more reliance should be placed on self-monitoring of blood glucose and continuous glucose monitoring, particularly if treatment can cause hypoglycemia (179).

With declining kidney function, it is important to be aware of the increased risk for hypoglycemia. The glycemic target may have to be increased to avoid hypoglycemic episodes (179) and glucose lowering agents may have to be changed or have their dose adjusted (Table 5). There are several explanations for this: a) the kidney is important for the metabolism of many glucose lowering medications and this function is impaired in advanced CKD; b) the kidney contributes to total endogenous glucose production by approximately 30% which declines with loss of kidney function; c) in advanced CKD acidosis affects the liver’s ability to produce glucose and compensate for failing kidney gluconeogenesis, and malnutrition and muscle wasting contributes to the risk for hypoglycemia; d) people with diabetic nephropathy are often older, have longer diabetes duration, and more frequently suffer from comorbidities, especially cardiovascular disease, and are thus more likely to be on multiple medications with can have potential interactions with glucose lowering medications (242).

Table 5.

Glucose-Lowering Agents in Chronic Kidney Disease

DrugComment
MetforminRisk of accumulation and possibly lactic acidosis
Caution when eGFR <45 mL/min/1.73 m2
Stop when eGFR <30 mL/min/1.73 m2
SulfonylureasGlibenclamide, gliclazide, and tolbutamide predominantly renally excreted; may need to reduce dose
Meglitinides~10% excreted via kidney; usually safe
ThiazolidinedionesPredominantly hepatic metabolism; use may be limited by fluid retention
Dipeptidyl peptidase IV inhibitorsDose may need to be reduced in some agents
Glucagon-like peptide-1 receptor agonistsFew data when eGFR <15 mL/min/1.73 m2
Sodium–glucose co-transporter 2 inhibitorsProtect kidney and heart down to eGFR>25, but ineffective at reducing glucose at eGFR <45 mL/min/1.73 m2
InsulinExcreted by kidney; may need to reduce dose and/or switch to shorter-acting preparations

Metformin and its metabolites are excreted mainly by the kidney. In kidney failure, they accumulate and inhibit lactate oxidation. Metformin should therefore be used cautiously in those with eGFR <45 mL/min/1.73 m2, and stopped completely when eGFR <30 mL/min/1.73 m2 (243).

The sulfonylureas glibenclamide, gliclazide, and tolbutamide are excreted predominantly by the kidneys and accumulate in CKD. Their dose, and indeed the dose of any sulfonylurea, may need to be reduced as CKD progresses. Only ~10% of the meglitinides, repaglinide and nateglinide, are excreted by the kidneys, making them suitable alternative agents. The thiazolidinediones, rosiglitazone and pioglitazone, are predominantly metabolized in the liver. However, their use in ESKD may be limited by fluid retention.

Insulin is also excreted by the kidney so that reduced dosage, and perhaps a switch to shorter acting preparations, may be required.

The dose of some but not all DPP-4 inhibitors and GLP-1 receptor agonists may need to be reduced as kidney function deteriorates. The SGLT-2 inhibitors become less effective at decreasing glucose levels as GFR falls.

Anemia

Anemia is common in people with diabetes and CKD stage 3 or poorer (244). Full investigation of iron deficiency anemia may be needed to exclude a non-kidney cause. Those with anemia have a higher mortality, higher rates of hospital admission with heart failure, and poorer quality of life. Iron stores should be repleted with oral or parenteral iron as necessary, and erythropoietin replacement commenced if indicated. In the TREAT trial it was investigated if treatment of anemia in T2D with CKD would improve renal or cardiovascular outcome, but the trial showed no benefit (245).

When to Refer to Nephrology

Patients who begin dialysis as an emergency do less well than those in whom treatment is planned (246). Referral to nephrology should be made when eGFR is declining rapidly (>5 mL/min/1.73m2/year or when eGFR is <30-45 mL/min/1.73 m2. This allows structured physical and psychological preparation for kidney replacement therapy. Earlier referral may be necessary in particular circumstances (Table 6). The need for kidney replacement therapy should be discussed with all patients and those who wish it should have access. People without significant comorbidities will usually be offered transplantation. Full cardiovascular assessment and treatment are essential before transplantation.

Table 6.

Indications for Referral to Nephrology

Diagnosis uncertain
Hypertension difficult to control
Fluid overload
Anemia unresponsive to oral iron
Abnormal bone chemistry (calcium, phosphorus, PTH)
eGFR 30–45 mL/min/1.73 m2
Nephrotic syndrome
eGFR fall >5 mL/min/1.73 m2 per year

Organization of Care

Structured care, delivered by trained specialists working with clear protocols with specific, multiple treatment goals for all the variables described above, reduces the incidence of moderately elevated albuminuria (247, 248) and provides greater kidney and cardiovascular benefits than routine care for individuals with T2D and CKD (179, 249, 250). Progression to ESKD or death, need for laser therapy for management of retinopathy, and cardiovascular endpoints including stroke and heart failure are all reduced by such multifactorial interventions (251-254). When structured intensive multifactorial intervention targeting lifestyle factors (diet, exercise, smoking) and heart and kidney risk factors (blood glucose, blood pressure, lipid management) compared to usual care was started already in T2D with moderately elevated albuminuria, long-term follow-up of the Steno-2 study demonstrated that eight years of intervention translated into almost 8 years of extended median survival (Figure 11) (251).

Figure 11. . Steno-2 post-trial: Twenty-one years sustained effect of intensive multifactorial intervention compared to standard of care for 8 years targeting lifestyle and heart and kidney risk factors.

Figure 11.

Steno-2 post-trial: Twenty-one years sustained effect of intensive multifactorial intervention compared to standard of care for 8 years targeting lifestyle and heart and kidney risk factors.

Pregnancy in Women with Diabetes and Chronic Kidney Disease

Women with diabetic nephropathy have poor pregnancy outcomes (255). They remain at increased risk of hypertension, preeclampsia, abnormal fetal growth, and preterm delivery (256). In a recent series, the prevalence of diabetic nephropathy and moderately elevated albuminuria in early pregnancy was similar in women with T1D or T2D, and pregnancy outcomes were comparable regardless of the type of diabetes (257). Women with any evidence of CKD therefore should be counselled pre-pregnancy. RAS inhibitors should be stopped and therapies safe in pregnancy, such as methyldopa, labetolol, and nifedipine, used as substitutes. In women with T1D, maintenance of BP <135/85 mmHg and proteinuria <300 mg/24 h with methyldopa improves outcomes (208, 258).

REFERENCES

1.
Federation ID. IDF Diabetes Atlas Brussels, Belgium2019 [9th Edition:[Available from: https://www​.diabetesatlas.org.
2.
Group DP. Incidence and trends of childhood Type 1 diabetes worldwide 1990-1999. Diabet Med. 2006;23(8):857–66. [PubMed: 16911623]
3.
Patterson CC, Harjutsalo V, Rosenbauer J, Neu A, Cinek O, Skrivarhaug T, et al. Trends and cyclical variation in the incidence of childhood type 1 diabetes in 26 European centres in the 25 year period 1989-2013: a multicentre prospective registration study. Diabetologia. 2019;62(3):408–17. [PubMed: 30483858]
4.
United States Renal Data System. 2020 USRDS Annual Data Report: Epidemiology of Kidney Disease in the United States. In: National Institutes of Health NIoDaDaKD, editor. Bethesda, MD2020.
5.
Caramori ML, Kim Y, Huang C, Fish AJ, Rich SS, Miller ME, et al. Cellular basis of diabetic nephropathy: 1. Study design and renal structural-functional relationships in patients with long-standing type 1 diabetes. Diabetes. 2002;51(2):506–13. [PubMed: 11812762]
6.
Mauer SM, Steffes MW, Brown DM. The kidney in diabetes. The American journal of medicine. 1981;70(3):603–12. [PubMed: 7011015]
7.
Molitch ME, Steffes M, Sun W, Rutledge B, Cleary P, de Boer IH, et al. Development and progression of renal insufficiency with and without albuminuria in adults with type 1 diabetes in the diabetes control and complications trial and the epidemiology of diabetes interventions and complications study. Diabetes care. 2010;33(7):1536–43. [PMC free article: PMC2890355] [PubMed: 20413518]
8.
Lamacchia O, Viazzi F, Fioretto P, Mirijello A, Giorda C, Ceriello A, et al. Normoalbuminuric kidney impairment in patients with T1DM: insights from annals initiative. Diabetol Metab Syndr. 2018;10:60. [PMC free article: PMC6069993] [PubMed: 30083251]
9.
Retnakaran R, Cull CA, Thorne KI, Adler AI, Holman RR, Group US. Risk factors for renal dysfunction in type 2 diabetes: U.K. Prospective Diabetes Study 74. Diabetes. 2006;55(6):1832–9. [PubMed: 16731850]
10.
Thomas MC, Macisaac RJ, Jerums G, Weekes A, Moran J, Shaw JE, et al. Nonalbuminuric renal impairment in type 2 diabetic patients and in the general population (national evaluation of the frequency of renal impairment cO-existing with NIDDM [NEFRON] 11). Diabetes care. 2009;32(8):1497–502. [PMC free article: PMC2713618] [PubMed: 19470839]
11.
Penno G, Solini A, Bonora E, Fondelli C, Orsi E, Zerbini G, et al. Clinical significance of nonalbuminuric renal impairment in type 2 diabetes. J Hypertens. 2011;29(9):1802–9. [PubMed: 21738053]
12.
Pichaiwong W, Homsuwan W, Leelahavanichkul A. The prevalence of normoalbuminuria and renal impairment in type 2 diabetes mellitus. Clin Nephrol. 2019;92(2):73–80. [PubMed: 31198168]
13.
Di Bonito P, Mozzillo E, Rosanio FM, Maltoni G, Piona CA, Franceschi R, et al. Albuminuric and non-albuminuric reduced eGFR phenotypes in youth with type 1 diabetes: Factors associated with cardiometabolic risk. Nutr Metab Cardiovasc Dis. 2021;31(7):2033–41. [PubMed: 34083127]
14.
Pugliese G, Solini A, Bonora E, Fondelli C, Orsi E, Nicolucci A, et al. Chronic kidney disease in type 2 diabetes: lessons from the Renal Insufficiency And Cardiovascular Events (RIACE) Italian Multicentre Study. Nutr Metab Cardiovasc Dis. 2014;24(8):815–22. [PubMed: 24780515]
15.
Penno G, Solini A, Orsi E, Bonora E, Fondelli C, Trevisan R, et al. Non-albuminuric renal impairment is a strong predictor of mortality in individuals with type 2 diabetes: the Renal Insufficiency And Cardiovascular Events (RIACE) Italian multicentre study. Diabetologia. 2018;61(11):2277–89. [PubMed: 30032426]
16.
Afkarian M, Polsky S, Parsa A, Aronson R, Caramori ML, Cherney DZ, et al. Preventing Early Renal Loss in Diabetes (PERL) Study: A Randomized Double-Blinded Trial of Allopurinol-Rationale, Design, and Baseline Data. Diabetes care. 2019;42(8):1454–63. [PMC free article: PMC6647051] [PubMed: 31186299]
17.
Doria A, Galecki AT, Spino C, Pop-Busui R, Cherney DZ, Lingvay I, et al. Serum Urate Lowering with Allopurinol and Kidney Function in Type 1 Diabetes. N Engl J Med. 2020;382(26):2493–503. [PMC free article: PMC7375708] [PubMed: 32579810]
18.
Garofolo M, Russo E, Miccoli R, Lucchesi D, Giusti L, Sancho-Bornez V, et al. Albuminuric and non-albuminuric chronic kidney disease in type 1 diabetes: Association with major vascular outcomes risk and all-cause mortality. Journal of diabetes and its complications. 2018;32(6):550–7. [PubMed: 29705091]
19.
MacIsaac RJ, Tsalamandris C, Panagiotopoulos S, Smith TJ, McNeil KJ, Jerums G. Nonalbuminuric renal insufficiency in type 2 diabetes. Diabetes care. 2004;27(1):195–200. [PubMed: 14693989]
20.
American Diabetes Association Professional Practice C. 11. Chronic Kidney Disease and Risk Management: Standards of Medical Care in Diabetes-2022. Diabetes care. 2022;45 Suppl 1:S175–S84. Draznin B, Aroda VR, Bakris G, Benson G, Brown FM, et al.
21.
Diabetes Canada Clinical Practice Guidelines Expert C. McFarlane P, Cherney D, Gilbert RE, Senior P. Chronic Kidney Disease in Diabetes. Can J Diabetes. 2018;42 Suppl 1:S201–S9. [PubMed: 29650098]
22.
Kidney Disease: Improving Global Outcomes Diabetes Work G. KDIGO 2020 Clinical Practice Guideline for Diabetes Management in Chronic Kidney Disease. Kidney international. 2020;98(4S):S1–S115. [PubMed: 32998798]
23.
Gansevoort RT, Brinkman J, Bakker SJ, De Jong PE, de Zeeuw D. Evaluation of measures of urinary albumin excretion. Am J Epidemiol. 2006;164(8):725–7. [PubMed: 16936069]
24.
Levin A, Stevens PE. Summary of KDIGO 2012 CKD Guideline: behind the scenes, need for guidance, and a framework for moving forward. Kidney international. 2014;85(1):49–61. [PubMed: 24284513]
25.
Targher G, Zoppini G, Mantovani W, Chonchol M, Negri C, Stoico V, et al. Comparison of two creatinine-based estimating equations in predicting all-cause and cardiovascular mortality in patients with type 2 diabetes. Diabetes care. 2012;35(11):2347–53. [PMC free article: PMC3476924] [PubMed: 22837367]
26.
Matsushita K, Mahmoodi BK, Woodward M, Emberson JR, Jafar TH, Jee SH, et al. Comparison of risk prediction using the CKD-EPI equation and the MDRD study equation for estimated glomerular filtration rate. JAMA. 2012;307(18):1941–51. [PMC free article: PMC3837430] [PubMed: 22570462]
27.
von Scholten BJ, Persson F, Svane MS, Hansen TW, Madsbad S, Rossing P. Effect of large weight reductions on measured and estimated kidney function. BMC nephrology. 2017;18(1):52. [PMC free article: PMC5294831] [PubMed: 28166744]
28.
Inker LA, Schmid CH, Tighiouart H, Eckfeldt JH, Feldman HI, Greene T, et al. Estimating glomerular filtration rate from serum creatinine and cystatin C. N Engl J Med. 2012;367(1):20–9. [PMC free article: PMC4398023] [PubMed: 22762315]
29.
Iliadis F, Didangelos T, Ntemka A, Makedou A, Moralidis E, Gotzamani-Psarakou A, et al. Glomerular filtration rate estimation in patients with type 2 diabetes: creatinine- or cystatin C-based equations? Diabetologia. 2011;54(12):2987–94. [PubMed: 21947381]
30.
Tsai CW, Grams ME, Inker LA, Coresh J, Selvin E. Cystatin C- and creatinine-based estimated glomerular filtration rate, vascular disease, and mortality in persons with diabetes in the U.S. Diabetes care. 2014;37(4):1002–8. [PMC free article: PMC3964484] [PubMed: 24271191]
31.
Chapter 2: Definition, identification, and prediction of CKD progression. Kidney Int Suppl (2011). 2013;3(1):63-72. [PMC free article: PMC4089637] [PubMed: 25018976]
32.
Inker LA, Eneanya ND, Coresh J, Tighiouart H, Wang D, Sang Y, et al. New Creatinine- and Cystatin C-Based Equations to Estimate GFR without Race. N Engl J Med. 2021;385(19):1737–49. [PMC free article: PMC8822996] [PubMed: 34554658]
33.
Mazzucco G, Bertani T, Fortunato M, Bernardi M, Leutner M, Boldorini R, et al. Different patterns of renal damage in type 2 diabetes mellitus: a multicentric study on 393 biopsies. American journal of kidney diseases : the official journal of the National Kidney Foundation. 2002;39(4):713–20. [PubMed: 11920336]
34.
Chong YB, Keng TC, Tan LP, Ng KP, Kong WY, Wong CM, et al. Clinical predictors of non-diabetic renal disease and role of renal biopsy in diabetic patients with renal involvement: a single centre review. Ren Fail. 2012;34(3):323–8. [PubMed: 22250665]
35.
Sharma SG, Bomback AS, Radhakrishnan J, Herlitz LC, Stokes MB, Markowitz GS, et al. The modern spectrum of renal biopsy findings in patients with diabetes. Clinical journal of the American Society of Nephrology : CJASN. 2013;8(10):1718–24. [PMC free article: PMC3789339] [PubMed: 23886566]
36.
Mauer M, Drummond K. The early natural history of nephropathy in type 1 diabetes: I. Study design and baseline characteristics of the study participants. Diabetes. 2002;51(5):1572–9. [PubMed: 11978658]
37.
Mauer M, Zinman B, Gardiner R, Suissa S, Sinaiko A, Strand T, et al. Renal and retinal effects of enalapril and losartan in type 1 diabetes. N Engl J Med. 2009;361(1):40–51. [PMC free article: PMC2978030] [PubMed: 19571282]
38.
Nelson RG, Meyer TW, Myers BD, Bennett PH. Clinical and pathological course of renal disease in non-insulin-dependent diabetes mellitus: the Pima Indian experience. Semin Nephrol. 1997;17(2):124–31. [PubMed: 9148378]
39.
Najafian B, Crosson JT, Kim Y, Mauer M. Glomerulotubular junction abnormalities are associated with proteinuria in type 1 diabetes. Journal of the American Society of Nephrology : JASN. 2006;17(4) Suppl 2:S53–60. [PubMed: 16565248]
40.
Najafian B, Kim Y, Crosson JT, Mauer M. Atubular glomeruli and glomerulotubular junction abnormalities in diabetic nephropathy. Journal of the American Society of Nephrology : JASN. 2003;14(4):908–17. [PubMed: 12660325]
41.
Osterby R. Morphometric studies of the peripheral glomerular basement membrane in early juvenile diabetes. I. Development of initial basement membrane thickening. Diabetologia. 1972;8(2):84–92. [PubMed: 5031267]
42.
Østerby R. Early phases in the development of diabetic glomerulopathy. Acta Med Scand Suppl. 1974;574:3–82. [PubMed: 4533587]
43.
Osterby R, Hartmann A, Bangstad HJ. Structural changes in renal arterioles in Type I diabetic patients. Diabetologia. 2002;45(4):542–9. [PubMed: 12032631]
44.
Fioretto P, Stehouwer CD, Mauer M, Chiesura-Corona M, Brocco E, Carraro A, et al. Heterogeneous nature of microalbuminuria in NIDDM: studies of endothelial function and renal structure. Diabetologia. 1998;41(2):233–6. [PubMed: 9498659]
45.
Brito PL, Fioretto P, Drummond K, Kim Y, Steffes MW, Basgen JM, et al. Proximal tubular basement membrane width in insulin-dependent diabetes mellitus. Kidney international. 1998;53(3):754–61. [PubMed: 9507223]
46.
Toyoda M, Najafian B, Kim Y, Caramori ML, Mauer M. Podocyte detachment and reduced glomerular capillary endothelial fenestration in human type 1 diabetic nephropathy. Diabetes. 2007;56(8):2155–60. [PubMed: 17536064]
47.
Weil EJ, Lemley KV, Mason CC, Yee B, Jones LI, Blouch K, et al. Podocyte detachment and reduced glomerular capillary endothelial fenestration promote kidney disease in type 2 diabetic nephropathy. Kidney international. 2012;82(9):1010–7. [PMC free article: PMC3472108] [PubMed: 22718189]
48.
Katz A, Caramori ML, Sisson-Ross S, Groppoli T, Basgen JM, Mauer M. An increase in the cell component of the cortical interstitium antedates interstitial fibrosis in type 1 diabetic patients. Kidney international. 2002;61(6):2058–66. [PubMed: 12028446]
49.
Fioretto P, Mauer M, Brocco E, Velussi M, Frigato F, Muollo B, et al. Patterns of renal injury in NIDDM patients with microalbuminuria. Diabetologia. 1996;39(12):1569–76. [PubMed: 8960844]
50.
Ellis EN, Steffes MW, Goetz FC, Sutherland DE, Mauer SM. Glomerular filtration surface in type I diabetes mellitus. Kidney international. 1986;29(4):889–94. [PubMed: 3712971]
51.
Mauer SM, Steffes MW, Ellis EN, Sutherland DE, Brown DM, Goetz FC. Structural-functional relationships in diabetic nephropathy. The Journal of clinical investigation. 1984;74(4):1143–55. [PMC free article: PMC425280] [PubMed: 6480821]
52.
Mauer SM, Sutherland DE, Steffes MW. Relationship of systemic blood pressure to nephropathology in insulin-dependent diabetes mellitus. Kidney Int. 1992;41(4):736–40. [PubMed: 1513095]
53.
Caramori ML, Parks A, Mauer M. Renal lesions predict progression of diabetic nephropathy in type 1 diabetes. Journal of the American Society of Nephrology : JASN. 2013;24(7):1175–81. [PMC free article: PMC3699823] [PubMed: 23687360]
54.
Harris RD, Steffes MW, Bilous RW, Sutherland DE, Mauer SM. Global glomerular sclerosis and glomerular arteriolar hyalinosis in insulin dependent diabetes. Kidney international. 1991;40(1):107–14. [PubMed: 1921145]
55.
Lane PH, Steffes MW, Fioretto P, Mauer SM. Renal interstitial expansion in insulin-dependent diabetes mellitus. Kidney international. 1993;43(3):661–7. [PubMed: 8455365]
56.
Ellis EN, Steffes MW, Chavers B, Mauer SM. Observations of glomerular epithelial cell structure in patients with type I diabetes mellitus. Kidney international. 1987;32(5):736–41. [PubMed: 3430959]
57.
Bjorn SF, Bangstad HJ, Hanssen KF, Nyberg G, Walker JD, Viberti GC, et al. Glomerular epithelial foot processes and filtration slits in IDDM patients. Diabetologia. 1995;38(10):1197–204. [PubMed: 8690172]
58.
Pagtalunan ME, Miller PL, Jumping-Eagle S, Nelson RG, Myers BD, Rennke HG, et al. Podocyte loss and progressive glomerular injury in type II diabetes. The Journal of clinical investigation. 1997;99(2):342–8. [PMC free article: PMC507802] [PubMed: 9006003]
59.
Harindhanavudhi T, Parks A, Mauer M, Caramori ML. Podocyte structural parameters do not predict progression to diabetic nephropathy in normoalbuminuric type 1 diabetic patients. Am J Nephrol. 2015;41(4-5):277–83. [PMC free article: PMC4514533] [PubMed: 26021523]
60.
Nordwall M, Abrahamsson M, Dhir M, Fredrikson M, Ludvigsson J, Arnqvist HJ. Impact of HbA1c, followed from onset of type 1 diabetes, on the development of severe retinopathy and nephropathy: the VISS Study (Vascular Diabetic Complications in Southeast Sweden). Diabetes care. 2015;38(2):308–15. [PubMed: 25510400]
61.
Bash LD, Selvin E, Steffes M, Coresh J, Astor BC. Poor glycemic control in diabetes and the risk of incident chronic kidney disease even in the absence of albuminuria and retinopathy: Atherosclerosis Risk in Communities (ARIC) Study. Arch Intern Med. 2008;168(22):2440–7. [PMC free article: PMC2766035] [PubMed: 19064828]
62.
Rotbain Curovic V, Theilade S, Winther SA, Tofte N, Tarnow L, Jorsal A, et al. Visit-to-visit variability of clinical risk markers in relation to long-term complications in type 1 diabetes. Diabet Med. 2021;38(5):e14459. [PubMed: 33179275]
63.
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):2198–202. [PMC free article: PMC2571045] [PubMed: 18650371]
64.
Hsu CC, Chang HY, Huang MC, Hwang SJ, Yang YC, Lee YS, et al. HbA1c variability is associated with microalbuminuria development in type 2 diabetes: a 7-year prospective cohort study. Diabetologia. 2012;55(12):3163–72. [PubMed: 22923064]
65.
Ranjan AG, Rosenlund SV, Hansen TW, Rossing P, Andersen S, Norgaard K. Improved Time in Range Over 1 Year Is Associated With Reduced Albuminuria in Individuals With Sensor-Augmented Insulin Pump-Treated Type 1 Diabetes. Diabetes care. 2020;43(11):2882–5. [PubMed: 32887707]
66.
Ceriello A. Glucose Variability and Diabetic Complications: Is It Time to Treat? Diabetes care. 2020;43(6):1169–71. [PubMed: 32434893]
67.
Norgaard K, Feldt-Rasmussen B, Borch-Johnsen K, Saelan H, Deckert T. Prevalence of hypertension in type 1 (insulin-dependent) diabetes mellitus. Diabetologia. 1990;33(7):407–10. [PubMed: 2401396]
68.
Dost A, Klinkert C, Kapellen T, Lemmer A, Naeke A, Grabert M, et al. Arterial hypertension determined by ambulatory blood pressure profiles: contribution to microalbuminuria risk in a multicenter investigation in 2,105 children and adolescents with type 1 diabetes. Diabetes care. 2008;31(4):720–5. [PubMed: 18174497]
69.
McMullan CJ, Lambers Heerspink HJ, Parving HH, Dwyer JP, Forman JP, de Zeeuw D. Visit-to-visit variability in blood pressure and kidney and cardiovascular outcomes in patients with type 2 diabetes and nephropathy: a post hoc analysis from the RENAAL study and the Irbesartan Diabetic Nephropathy Trial. American journal of kidney diseases : the official journal of the National Kidney Foundation. 2014;64(5):714–22. [PubMed: 25064674]
70.
Daousi C, Bain SC, Barnett AH, Gill GV. Hypertriglyceridaemia is associated with an increased likelihood of albuminuria in extreme duration (> 50 years) Type 1 diabetes. Diabet Med. 2008;25(10):1234–6. [PubMed: 19046204]
71.
Hovind P, Rossing P, Tarnow L, Smidt UM, Parving HH. Progression of diabetic nephropathy. Kidney international. 2001;59(2):702–9. [PubMed: 11168952]
72.
Thomas MC, Rosengard-Barlund M, Mills V, Ronnback M, Thomas S, Forsblom C, et al. Serum lipids and the progression of nephropathy in type 1 diabetes. Diabetes care. 2006;29(2):317–22. [PubMed: 16443880]
73.
Tolonen N, Forsblom C, Thorn L, Waden J, Rosengard-Barlund M, Saraheimo M, et al. Relationship between lipid profiles and kidney function in patients with type 1 diabetes. Diabetologia. 2008;51(1):12–20. [PubMed: 17994214]
74.
Tofte N, Suvitaival T, Ahonen L, Winther SA, Theilade S, Frimodt-Moller M, et al. Lipidomic analysis reveals sphingomyelin and phosphatidylcholine species associated with renal impairment and all-cause mortality in type 1 diabetes. Sci Rep. 2019;9(1):16398. [PMC free article: PMC6841673] [PubMed: 31705008]
75.
Rossing P, Hougaard P, Parving HH. Risk factors for development of incipient and overt diabetic nephropathy in type 1 diabetic patients: a 10-year prospective observational study. Diabetes care. 2002;25(5):859–64. [PubMed: 11978681]
76.
Bjornstad P, Snell-Bergeon JK, Rewers M, Jalal D, Chonchol MB, Johnson RJ, et al. Early diabetic nephropathy: a complication of reduced insulin sensitivity in type 1 diabetes. Diabetes care. 2013;36(11):3678–83. [PMC free article: PMC3816872] [PubMed: 24026551]
77.
Hsu CC, Chang HY, Huang MC, Hwang SJ, Yang YC, Tai TY, et al. Association between insulin resistance and development of microalbuminuria in type 2 diabetes: a prospective cohort study. Diabetes care. 2011;34(4):982–7. [PMC free article: PMC3064061] [PubMed: 21335369]
78.
Thorn LM, Forsblom C, Waden J, Saraheimo M, Tolonen N, Hietala K, et al. Metabolic syndrome as a risk factor for cardiovascular disease, mortality, and progression of diabetic nephropathy in type 1 diabetes. Diabetes care. 2009;32(5):950–2. [PMC free article: PMC2671127] [PubMed: 19196885]
79.
Kilpatrick ES, Rigby AS, Atkin SL. Insulin resistance, the metabolic syndrome, and complication risk in type 1 diabetes: "double diabetes" in the Diabetes Control and Complications Trial. Diabetes care. 2007;30(3):707–12. [PubMed: 17327345]
80.
Hovind P, Rossing P, Tarnow L, Johnson RJ, Parving HH. Serum uric acid as a predictor for development of diabetic nephropathy in type 1 diabetes: an inception cohort study. Diabetes. 2009;58(7):1668–71. [PMC free article: PMC2699868] [PubMed: 19411615]
81.
Piehlmeier W, Renner R, Schramm W, Kimmerling T, Garbe S, Proetzsch R, et al. Screening of diabetic patients for microalbuminuria in primary care--The PROSIT-Project. Proteinuria Screening and Intervention. Exp Clin Endocrinol Diabetes. 1999;107(4):244–51. [PubMed: 10433063]
82.
Caramori ML, Gross JL, Pecis M, de Azevedo MJ. Glomerular filtration rate, urinary albumin excretion rate, and blood pressure changes in normoalbuminuric normotensive type 1 diabetic patients: an 8-year follow-up study. Diabetes care. 1999;22(9):1512–6. [PubMed: 10480518]
83.
Magee GM, Bilous RW, Cardwell CR, Hunter SJ, Kee F, Fogarty DG. Is hyperfiltration associated with the future risk of developing diabetic nephropathy? A meta-analysis. Diabetologia. 2009;52(4):691–7. [PubMed: 19198800]
84.
Jerums G, Premaratne E, Panagiotopoulos S, MacIsaac RJ. The clinical significance of hyperfiltration in diabetes. Diabetologia. 2010;53(10):2093–104. [PubMed: 20496053]
85.
Thomas MC, Moran JL, Harjutsalo V, Thorn L, Waden J, Saraheimo M, et al. Hyperfiltration in type 1 diabetes: does it exist and does it matter for nephropathy? Diabetologia. 2012;55(5):1505–13. [PubMed: 22322918]
86.
Cherney DZ, Perkins BA, Soleymanlou N, Maione M, Lai V, Lee A, et al. Renal hemodynamic effect of sodium-glucose cotransporter 2 inhibition in patients with type 1 diabetes mellitus. Circulation. 2014;129(5):587–97. [PubMed: 24334175]
87.
van Bommel EJM, Muskiet MHA, van Baar MJB, Tonneijck L, Smits MM, Emanuel AL, et al. The renal hemodynamic effects of the SGLT2 inhibitor dapagliflozin are caused by post-glomerular vasodilatation rather than pre-glomerular vasoconstriction in metformin-treated patients with type 2 diabetes in the randomized, double-blind RED trial. Kidney international. 2020;97(1):202–12. [PubMed: 31791665]
88.
Seaquist ER, Goetz FC, Rich S, Barbosa J. Familial clustering of diabetic kidney disease. Evidence for genetic susceptibility to diabetic nephropathy. N Engl J Med. 1989;320(18):1161–5. [see comments] [PubMed: 2710189]
89.
Fagerudd JA, Pettersson-Fernholm KJ, Gronhagen-Riska C, Groop PH. The impact of a family history of Type II (non-insulin-dependent) diabetes mellitus on the risk of diabetic nephropathy in patients with Type I (insulin-dependent) diabetes mellitus. Diabetologia. 1999;42(5):519–26. [PubMed: 10333042]
90.
Thorn LM, Forsblom C, Fagerudd J, Pettersson-Fernholm K, Kilpikari R, Groop PH, et al. Clustering of risk factors in parents of patients with type 1 diabetes and nephropathy. Diabetes care. 2007;30(5):1162–7. [PubMed: 17337502]
91.
Fioretto P, Steffes MW, Barbosa J, Rich SS, Miller ME, Mauer M. Is diabetic nephropathy inherited? Studies of glomerular structure in type 1 diabetic sibling pairs. Diabetes. 1999;48(4):865–9. [PubMed: 10102705]
92.
Trevisan R, Fioretto P, Barbosa J, Mauer M. Insulin-dependent diabetic sibling pairs are concordant for sodium-hydrogen antiport activity. Kidney international. 1999;55(6):2383–9. [PubMed: 10354286]
93.
Caramori ML, Kim Y, Fioretto P, Huang C, Rich SS, Miller ME, et al. Cellular basis of diabetic nephropathy: IV Antioxidant enzyme mRNA expression levels in skin fibroblasts of type 1 diabetic sibling pairs. Nephrol Dial Transplant. 2006 [PubMed: 16877492]
94.
Wuttke M, Li Y, Li M, Sieber KB, Feitosa MF, Gorski M, et al. A catalog of genetic loci associated with kidney function from analyses of a million individuals. Nat Genet. 2019;51(6):957–72. [PMC free article: PMC6698888] [PubMed: 31152163]
95.
Keating ST, van Diepen JA, Riksen NP, El-Osta A. Epigenetics in diabetic nephropathy, immunity and metabolism. Diabetologia. 2018;61(1):6–20. [PMC free article: PMC6448927] [PubMed: 29128937]
96.
Sinha SK, Shaheen M, Rajavashisth TB, Pan D, Norris KC, Nicholas SB. Association of race/ethnicity, inflammation, and albuminuria in patients with diabetes and early chronic kidney disease. Diabetes care. 2014;37(4):1060–8. [PMC free article: PMC4069363] [PubMed: 24550221]
97.
Allawi J, Rao PV, Gilbert R, Scott G, Jarrett RJ, Keen H, et al. Microalbuminuria in non-insulin-dependent diabetes: its prevalence in Indian compared with Europid patients. Br Med J (Clin Res Ed). 1988;296(6620):462–4. [PMC free article: PMC2545043] [PubMed: 3126860]
98.
Dreyer G, Hull S, Aitken Z, Chesser A, Yaqoob MM. The effect of ethnicity on the prevalence of diabetes and associated chronic kidney disease. QJM. 2009;102(4):261–9. [PubMed: 19147658]
99.
Nelson RG, Knowler WC, Pettitt DJ, Hanson RL, Bennett PH. Incidence and determinants of elevated urinary albumin excretion in Pima Indians with NIDDM. Diabetes care. 1995;18(2):182–7. [PubMed: 7729295]
100.
Joshy G, Dunn P, Fisher M, Lawrenson R. Ethnic differences in the natural progression of nephropathy among diabetes patients in New Zealand: hospital admission rate for renal complications, and incidence of end-stage renal disease and renal death. Diabetologia. 2009;52(8):1474–8. [PubMed: 19436991]
101.
Collins VR, Dowse GK, Finch CF, Zimmet PZ, Linnane AW. Prevalence and risk factors for micro- and macroalbuminuria in diabetic subjects and entire population of Nauru. Diabetes. 1989;38(12):1602–10. [PubMed: 2583379]
102.
Group TS. Rapid rise in hypertension and nephropathy in youth with type 2 diabetes: the TODAY clinical trial. Diabetes care. 2013;36(6):1735–41. [PMC free article: PMC3661847] [PubMed: 23704672]
103.
Dart AB, Sellers EA, Martens PJ, Rigatto C, Brownell MD, Dean HJ. High burden of kidney disease in youth-onset type 2 diabetes. Diabetes care. 2012;35(6):1265–71. [PMC free article: PMC3357249] [PubMed: 22432116]
104.
Dyck RF, Jiang Y, Osgood ND. The long-term risks of end stage renal disease and mortality among First Nations and non-First Nations people with youth-onset diabetes. Can J Diabetes. 2014;38(4):237–43. [PubMed: 24986804]
105.
Chan JC, Lau ES, Luk AO, Cheung KK, Kong AP, Yu LW, et al. Premature mortality and comorbidities in young-onset diabetes: a 7-year prospective analysis. The American journal of medicine. 2014;127(7):616–24. [PubMed: 24680795]
106.
Sellers EA, Blydt-Hansen TD, Dean HJ, Gibson IW, Birk PE, Ogborn M. Macroalbuminuria and renal pathology in First Nation youth with type 2 diabetes. Diabetes care. 2009;32(5):786–90. [PMC free article: PMC2671101] [PubMed: 19228860]
107.
Murussi M, Campagnolo N, Beck MO, Gross JL, Silveiro SP. High-normal levels of albuminuria predict the development of micro- and macroalbuminuria and increased mortality in Brazilian Type 2 diabetic patients: an 8-year follow-up study. Diabet Med. 2007;24(10):1136–42. [PubMed: 17561963]
108.
de Zeeuw D, Ramjit D, Zhang Z, Ribeiro AB, Kurokawa K, Lash JP, et al. Renal risk and renoprotection among ethnic groups with type 2 diabetic nephropathy: a post hoc analysis of RENAAL. Kidney international. 2006;69(9):1675–82. [PubMed: 16572114]
109.
Caramori ML, Fioretto P, Mauer M. Long-term follow-up of normoalbuminuric longstanding type 1 diabetic patients: Progression is associated with worse baseline glomerular lesions and lower glomerular filtration rate. Journal of the American Society of Nephrology : JASN. 1999;10:126A. [Abstract]
110.
Babazono T, Nyumura I, Toya K, Hayashi T, Ohta M, Suzuki K, et al. Higher levels of urinary albumin excretion within the normal range predict faster decline in glomerular filtration rate in diabetic patients. Diabetes care. 2009;32(8):1518–20. [PMC free article: PMC2713629] [PubMed: 19435960]
111.
Zoppini G, Targher G, Chonchol M, Ortalda V, Negri C, Stoico V, et al. Predictors of estimated GFR decline in patients with type 2 diabetes and preserved kidney function. Clinical journal of the American Society of Nephrology : CJASN. 2012;7(3):401–8. [PubMed: 22282481]
112.
Rossing P, Hommel E, Smidt UM, Parving HH. Reduction in albuminuria predicts a beneficial effect on diminishing the progression of human diabetic nephropathy during antihypertensive treatment. Diabetologia. 1994;37(5):511–6. [PubMed: 8056190]
113.
Heerspink HJL, Greene T, Tighiouart H, Gansevoort RT, Coresh J, Simon AL, et al. Change in albuminuria as a surrogate endpoint for progression of kidney disease: a meta-analysis of treatment effects in randomised clinical trials. Lancet Diabetes Endocrinol. 2019;7(2):128–39. [PubMed: 30635226]
114.
Gordin D, Hiilesmaa V, Fagerudd J, Ronnback M, Forsblom C, Kaaja R, et al. Pre-eclampsia but not pregnancy-induced hypertension is a risk factor for diabetic nephropathy in type 1 diabetic women. Diabetologia. 2007;50(3):516–22. [PubMed: 17216281]
115.
Niewczas MA, Pavkov ME, Skupien J, Smiles A, Md Dom ZI, Wilson JM, et al. A signature of circulating inflammatory proteins and development of end-stage renal disease in diabetes. Nat Med. 2019;25(5):805–13. [PMC free article: PMC6508971] [PubMed: 31011203]
116.
Rotbain Curovic V, Theilade S, Winther SA, Tofte N, Eugen-Olsen J, Persson F, et al. Soluble Urokinase Plasminogen Activator Receptor Predicts Cardiovascular Events, Kidney Function Decline, and Mortality in Patients With Type 1 Diabetes. Diabetes care. 2019;42(6):1112–9. [PubMed: 30885954]
117.
Schrijvers BF, De Vriese AS, Flyvbjerg A. From hyperglycemia to diabetic kidney disease: the role of metabolic, hemodynamic, intracellular factors and growth factors/cytokines. Endocr Rev. 2004;25(6):971–1010. [PubMed: 15583025]
118.
Shultis WA, Weil EJ, Looker HC, Curtis JM, Shlossman M, Genco RJ, et al. Effect of periodontitis on overt nephropathy and end-stage renal disease in type 2 diabetes. Diabetes care. 2007;30(2):306–11. [PubMed: 17259499]
119.
Riphagen IJ, Deetman PE, Bakker SJ, Navis G, Cooper ME, Lewis JB, et al. Bilirubin and progression of nephropathy in type 2 diabetes: a post hoc analysis of RENAAL with independent replication in IDNT. Diabetes. 2014;63(8):2845–53. [PubMed: 24677717]
120.
Mashitani T, Hayashino Y, Okamura S, Tsujii S, Ishii H. Correlations between serum bilirubin levels and diabetic nephropathy progression among Japanese type 2 diabetic patients: a prospective cohort study (Diabetes Distress and Care Registry at Tenri [DDCRT 5]). Diabetes care. 2014;37(1):252–8. [PubMed: 24009299]
121.
Tahrani AA, Ali A, Raymond NT, Begum S, Dubb K, Altaf QA, et al. Obstructive sleep apnea and diabetic nephropathy: a cohort study. Diabetes care. 2013;36(11):3718–25. [PMC free article: PMC3816897] [PubMed: 24062320]
122.
Targher G, Bertolini L, Rodella S, Zoppini G, Lippi G, Day C, et al. Non-alcoholic fatty liver disease is independently associated with an increased prevalence of chronic kidney disease and proliferative/laser-treated retinopathy in type 2 diabetic patients. Diabetologia. 2008;51(3):444–50. [PubMed: 18058083]
123.
Gohda T, Niewczas MA, Ficociello LH, Walker WH, Skupien J, Rosetti F, et al. Circulating TNF receptors 1 and 2 predict stage 3 CKD in type 1 diabetes. Journal of the American Society of Nephrology : JASN. 2012;23(3):516–24. [PMC free article: PMC3294299] [PubMed: 22266664]
124.
Niewczas MA, Gohda T, Skupien J, Smiles AM, Walker WH, Rosetti F, et al. Circulating TNF receptors 1 and 2 predict ESRD in type 2 diabetes. Journal of the American Society of Nephrology : JASN. 2012;23(3):507–15. [PMC free article: PMC3294310] [PubMed: 22266663]
125.
Forsblom C, Moran J, Harjutsalo V, Loughman T, Waden J, Tolonen N, et al. Added value of soluble tumor necrosis factor-alpha receptor 1 as a biomarker of ESRD risk in patients with type 1 diabetes. Diabetes care. 2014;37(8):2334–42. [PubMed: 24879837]
126.
Pavkov ME, Nelson RG, Knowler WC, Cheng Y, Krolewski AS, Niewczas MA. Elevation of circulating TNF receptors 1 and 2 increases the risk of end-stage renal disease in American Indians with type 2 diabetes. Kidney international. 2015;87(4):812–9. [PMC free article: PMC4382420] [PubMed: 25272234]
127.
Amin AP, Whaley-Connell AT, Li S, Chen SC, McCullough PA, Kosiborod MN, et al. The synergistic relationship between estimated GFR and microalbuminuria in predicting long-term progression to ESRD or death in patients with diabetes: results from the Kidney Early Evaluation Program (KEEP). American journal of kidney diseases : the official journal of the National Kidney Foundation. 2013;61(4) Suppl 2:S12–23. [PMC free article: PMC4492431] [PubMed: 23507266]
128.
Afkarian M, Sachs MC, Kestenbaum B, Hirsch IB, Tuttle KR, Himmelfarb J, et al. Kidney disease and increased mortality risk in type 2 diabetes. Journal of the American Society of Nephrology : JASN. 2013;24(2):302–8. [PMC free article: PMC3559486] [PubMed: 23362314]
129.
McCullough PA, Jurkovitz CT, Pergola PE, McGill JB, Brown WW, Collins AJ, et al. Independent components of chronic kidney disease as a cardiovascular risk state: results from the Kidney Early Evaluation Program (KEEP). Arch Intern Med. 2007;167(11):1122–9. [PubMed: 17563019]
130.
So WY, Kong AP, Ma RC, Ozaki R, Szeto CC, Chan NN, et al. Glomerular filtration rate, cardiorenal end points, and all-cause mortality in type 2 diabetic patients. Diabetes care. 2006;29(9):2046–52. [PubMed: 16936151]
131.
Bruno G, Merletti F, Bargero G, Novelli G, Melis D, Soddu A, et al. Estimated glomerular filtration rate, albuminuria and mortality in type 2 diabetes: the Casale Monferrato study. Diabetologia. 2007;50(5):941–8. [PubMed: 17333106]
132.
Groop PH, Thomas MC, Moran JL, Waden J, Thorn LM, Makinen VP, et al. The presence and severity of chronic kidney disease predicts all-cause mortality in type 1 diabetes. Diabetes. 2009;58(7):1651–8. [PMC free article: PMC2699848] [PubMed: 19401416]
133.
Orchard TJ, Secrest AM, Miller RG, Costacou T. In the absence of renal disease, 20 year mortality risk in type 1 diabetes is comparable to that of the general population: a report from the Pittsburgh Epidemiology of Diabetes Complications Study. Diabetologia. 2010;53(11):2312–9. [PMC free article: PMC3057031] [PubMed: 20665208]
134.
Deckert T, Yokoyama H, Mathiesen E, Ronn B, Jensen T, Feldt-Rasmussen B, et al. Cohort study of predictive value of urinary albumin excretion for atherosclerotic vascular disease in patients with insulin dependent diabetes. BMJ. 1996;312(7035):871–4. [PMC free article: PMC2350594] [PubMed: 8611873]
135.
Tuomilehto J, Borch-Johnsen K, Molarius A, Forsen T, Rastenyte D, Sarti C, et al. Incidence of cardiovascular disease in Type 1 (insulin-dependent) diabetic subjects with and without diabetic nephropathy in Finland. Diabetologia. 1998;41(7):784–90. [PubMed: 9686919]
136.
Bell S, Fletcher EH, Brady I, Looker HC, Levin D, Joss N, et al. End-stage renal disease and survival in people with diabetes: a national database linkage study. QJM. 2015;108(2):127–34. [PMC free article: PMC4309927] [PubMed: 25140030]
137.
Dinneen SF, Gerstein HC. The association of microalbuminuria and mortality in non-insulin-dependent diabetes mellitus. A systematic overview of the literature. Arch Intern Med. 1997;157(13):1413–8. [PubMed: 9224218]
138.
Fuller JH, Stevens LK, Wang SL. Risk factors for cardiovascular mortality and morbidity: the WHO Mutinational Study of Vascular Disease in Diabetes. Diabetologia. 2001;44 Suppl 2:S54–64. [PubMed: 11587051]
139.
Adler AI, Stevens RJ, Manley SE, Bilous RW, Cull CA, Holman RR. Development and progression of nephropathy in type 2 diabetes: the United Kingdom Prospective Diabetes Study (UKPDS 64). Kidney international. 2003;63(1):225–32. [PubMed: 12472787]
140.
Kramer CK, Retnakaran R. Concordance of retinopathy and nephropathy over time in Type 1 diabetes: an analysis of data from the Diabetes Control and Complications Trial. Diabet Med. 2013;30(11):1333–41. [PubMed: 23909911]
141.
Penno G, Solini A, Zoppini G, Orsi E, Zerbini G, Trevisan R, et al. Rate and determinants of association between advanced retinopathy and chronic kidney disease in patients with type 2 diabetes: the Renal Insufficiency And Cardiovascular Events (RIACE) Italian multicenter study. Diabetes care. 2012;35(11):2317–23. [PMC free article: PMC3476898] [PubMed: 23093684]
142.
Chen YH, Chen HS, Tarng DC. More impact of microalbuminuria on retinopathy than moderately reduced GFR among type 2 diabetic patients. Diabetes care. 2012;35(4):803–8. [PMC free article: PMC3308275] [PubMed: 22338100]
143.
Moriya T, Tanaka S, Kawasaki R, Ohashi Y, Akanuma Y, Yamada N, et al. Diabetic retinopathy and microalbuminuria can predict macroalbuminuria and renal function decline in Japanese type 2 diabetic patients: Japan Diabetes Complications Study. Diabetes care. 2013;36(9):2803–9. [PMC free article: PMC3747927] [PubMed: 23620478]
144.
Margolis DJ, Hofstad O, Feldman HI. Association between renal failure and foot ulcer or lower-extremity amputation in patients with diabetes. Diabetes care. 2008;31(7):1331–6. [PMC free article: PMC2453658] [PubMed: 18390800]
145.
Ko SH, Park SA, Cho JH, Song KH, Yoon KH, Cha BY, et al. Progression of cardiovascular autonomic dysfunction in patients with type 2 diabetes: a 7-year follow-up study. Diabetes care. 2008;31(9):1832–6. [PMC free article: PMC2518354] [PubMed: 18509202]
146.
Nielsen S, Schmitz A, Bacher T, Rehling M, Ingerslev J, Mogensen CE. Transcapillary escape rate and albuminuria in Type II diabetes. Effects of short-term treatment with low-molecular weight heparin. Diabetologia. 1999;42(1):60–7. [PubMed: 10027580]
147.
Tahrani AA, Dubb K, Raymond NT, Begum S, Altaf QA, Sadiqi H, et al. Cardiac autonomic neuropathy predicts renal function decline in patients with type 2 diabetes: a cohort study. Diabetologia. 2014;57(6):1249–56. [PubMed: 24623102]
148.
Group DER, de Boer IH, Sun W, Cleary PA, Lachin JM, Molitch ME, et al. Intensive diabetes therapy and glomerular filtration rate in type 1 diabetes. N Engl J Med. 2011;365(25):2366–76. [PMC free article: PMC3270008] [PubMed: 22077236]
149.
The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. The Diabetes Control and Complications Trial Research Group. N Engl J Med. 1993;329(14):977–86. [PubMed: 8366922]
150.
The absence of a glycemic threshold for the development of long-term complications: the perspective of the Diabetes Control and Complications Trial. Diabetes. 1996;45(10):1289–98. [PubMed: 8826962]
151.
Writing Team for the Diabetes C. Complications Trial/Epidemiology of Diabetes I, Complications Research G. Sustained effect of intensive treatment of type 1 diabetes mellitus on development and progression of diabetic nephropathy: the Epidemiology of Diabetes Interventions and Complications (EDIC) study. JAMA. 2003;290(16):2159–67. [PMC free article: PMC2622725] [PubMed: 14570951]
152.
Skupien J, Warram JH, Smiles A, Galecki A, Stanton RC, Krolewski AS. Improved glycemic control and risk of ESRD in patients with type 1 diabetes and proteinuria. Journal of the American Society of Nephrology : JASN. 2014;25(12):2916–25. [PMC free article: PMC4243344] [PubMed: 24904086]
153.
Fioretto P, Steffes MW, Sutherland DE, Goetz FC, Mauer M. Reversal of lesions of diabetic nephropathy after pancreas transplantation. N Engl J Med. 1998;339(2):69–75. [PubMed: 9654536]
154.
Beck RW, Bergenstal RM, Riddlesworth TD, Kollman C, Li Z, Brown AS, et al. Validation of Time in Range as an Outcome Measure for Diabetes Clinical Trials. Diabetes care. 2019;42(3):400–5. [PMC free article: PMC6905478] [PubMed: 30352896]
155.
Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). UK Prospective Diabetes Study (UKPDS) Group. Lancet. 1998;352(9131):837–53. [published erratum appears in Lancet 1999 Aug 14;354(9178):602] [see comments] [PubMed: 9742976]
156.
Stratton IM, Adler AI, Neil HA, Matthews DR, Manley SE, Cull CA, et al. Association of glycaemia with macrovascular and microvascular complications of type 2 diabetes (UKPDS 35): prospective observational study. BMJ. 2000;321(7258):405–12. [PMC free article: PMC27454] [PubMed: 10938048]
157.
Holman RR, Paul SK, Bethel MA, Neil HA, Matthews DR. Long-term follow-up after tight control of blood pressure in type 2 diabetes. N Engl J Med. 2008;359(15):1565–76. [PubMed: 18784091]
158.
Group AC, Patel A, MacMahon S, Chalmers J, Neal B, Billot L, et al. Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. N Engl J Med. 2008;358(24):2560–72. [PubMed: 18539916]
159.
Perkovic V, Heerspink HL, Chalmers J, Woodward M, Jun M, Li Q, et al. Intensive glucose control improves kidney outcomes in patients with type 2 diabetes. Kidney international. 2013;83(3):517–23. [PubMed: 23302714]
160.
Ismail-Beigi F, Craven T, Banerji MA, Basile J, Calles J, Cohen RM, et al. Effect of intensive treatment of hyperglycaemia on microvascular outcomes in type 2 diabetes: an analysis of the ACCORD randomised trial. Lancet. 2010;376(9739):419–30. [PMC free article: PMC4123233] [PubMed: 20594588]
161.
Papademetriou V, Lovato L, Doumas M, Nylen E, Mottl A, Cohen RM, et al. Chronic kidney disease and intensive glycemic control increase cardiovascular risk in patients with type 2 diabetes. Kidney international. 2015;87(3):649–59. [PubMed: 25229335]
162.
Shurraw S, Hemmelgarn B, Lin M, Majumdar SR, Klarenbach S, Manns B, et al. Association between glycemic control and adverse outcomes in people with diabetes mellitus and chronic kidney disease: a population-based cohort study. Arch Intern Med. 2011;171(21):1920–7. [PubMed: 22123800]
163.
Duong U, Mehrotra R, Molnar MZ, Noori N, Kovesdy CP, Nissenson AR, et al. Glycemic control and survival in peritoneal dialysis patients with diabetes mellitus. Clinical journal of the American Society of Nephrology : CJASN. 2011;6(5):1041–8. [PMC free article: PMC3087769] [PubMed: 21511838]
164.
Ricks J, Molnar MZ, Kovesdy CP, Shah A, Nissenson AR, Williams M, et al. Glycemic control and cardiovascular mortality in hemodialysis patients with diabetes: a 6-year cohort study. Diabetes. 2012;61(3):708–15. [PMC free article: PMC3282812] [PubMed: 22315308]
165.
Williams ME, Lacson E Jr, Wang W, Lazarus JM, Hakim R. Glycemic control and extended hemodialysis survival in patients with diabetes mellitus: comparative results of traditional and time-dependent Cox model analyses. Clinical journal of the American Society of Nephrology : CJASN. 2010;5(9):1595–601. [PMC free article: PMC2974399] [PubMed: 20671217]
166.
Hoshino J, Hamano T, Abe M, Hasegawa T, Wada A, Ubara Y, et al. Glycated albumin versus hemoglobin A1c and mortality in diabetic hemodialysis patients: a cohort study. Nephrol Dial Transplant. 2018;33(7):1150–8. [PubMed: 29528439]
167.
Morath C, Zeier M, Dohler B, Schmidt J, Nawroth PP, Opelz G. Metabolic control improves long-term renal allograft and patient survival in type 1 diabetes. Journal of the American Society of Nephrology : JASN. 2008;19(8):1557–63. [PMC free article: PMC2488262] [PubMed: 18495965]
168.
Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, et al. Empagliflozin, Cardiovascular Outcomes, and Mortality in Type 2 Diabetes. N Engl J Med. 2015;373(22):2117–28. [PubMed: 26378978]
169.
Wanner C, Inzucchi SE, Lachin JM, Fitchett D, von Eynatten M, Mattheus M, et al. Empagliflozin and Progression of Kidney Disease in Type 2 Diabetes. N Engl J Med. 2016;375(4):323–34. [PubMed: 27299675]
170.
McGuire DK, Shih WJ, Cosentino F, Charbonnel B, Cherney DZI, Dagogo-Jack S, et al. Association of SGLT2 Inhibitors With Cardiovascular and Kidney Outcomes in Patients With Type 2 Diabetes: A Meta-analysis. JAMA Cardiol. 2021;6(2):148–58. [PMC free article: PMC7542529] [PubMed: 33031522]
171.
Neuen BL, Young T, Heerspink HJL, Neal B, Perkovic V, Billot L, et al. SGLT2 inhibitors for the prevention of kidney failure in patients with type 2 diabetes: a systematic review and meta-analysis. Lancet Diabetes Endocrinol. 2019;7(11):845–54. [PubMed: 31495651]
172.
Perkovic V, Jardine MJ, Neal B, Bompoint S, Heerspink HJL, Charytan DM, et al. Canagliflozin and Renal Outcomes in Type 2 Diabetes and Nephropathy. N Engl J Med. 2019;380(24):2295–306. [PubMed: 30990260]
173.
Heerspink HJL, Stefansson BV, Correa-Rotter R, Chertow GM, Greene T, Hou FF, et al. Dapagliflozin in Patients with Chronic Kidney Disease. N Engl J Med. 2020;383(15):1436–46. [PubMed: 32970396]
174.
Group E-KC. Design, recruitment, and baseline characteristics of the EMPA-KIDNEY trial. Nephrol Dial Transplant. 2022;37(7):1317–29. [PMC free article: PMC9217655] [PubMed: 35238940]
175.
Packer M, Anker SD, Butler J, Filippatos G, Pocock SJ, Carson P, et al. Cardiovascular and Renal Outcomes with Empagliflozin in Heart Failure. N Engl J Med. 2020;383(15):1413–24. [PubMed: 32865377]
176.
Anker SD, Butler J, Filippatos G, Ferreira JP, Bocchi E, Bohm M, et al. Empagliflozin in Heart Failure with a Preserved Ejection Fraction. N Engl J Med. 2021;385(16):1451–61. [PubMed: 34449189]
177.
Kang A, Jardine MJ. SGLT2 inhibitors may offer benefit beyond diabetes. Nat Rev Nephrol. 2021;17(2):83–4. [PubMed: 33361782]
178.
Laursen JC, Sondergaard-Heinrich N, de Melo JML, Haddock B, Rasmussen IKB, Safavimanesh F, et al. Acute effects of dapagliflozin on renal oxygenation and perfusion in type 1 diabetes with albuminuria: A randomised, double-blind, placebo-controlled crossover trial. EClinicalMedicine. 2021;37:100895. [PMC free article: PMC8343250] [PubMed: 34386735]
179.
de Boer IH, Caramori ML, Chan JCN, Heerspink HJL, Hurst C, Khunti K, et al. Executive summary of the 2020 KDIGO Diabetes Management in CKD Guideline: evidence-based advances in monitoring and treatment. Kidney international. 2020;98(4):839–48. [PubMed: 32653403]
180.
Buse JB, Wexler DJ, Tsapas A, Rossing P, Mingrone G, Mathieu C, et al. 2019 Update to: Management of Hyperglycemia in Type 2 Diabetes, 2018. A Consensus Report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes care. 2020;43(2):487–93. [PMC free article: PMC6971782] [PubMed: 31857443]
181.
(NICE) NIoHaCE. Nice 2022 - Type 2 diabetes in adults: Management [web content].: National Health Service in England; 2022 [updated 31 March 2022, amended June 2022. https://www​.breathewellbeing​.in/blog/type-2-diabetes-management-in-2022/ .
182.
Tuttle KR, Lakshmanan MC, Rayner B, Busch RS, Zimmermann AG, Woodward DB, et al. Dulaglutide versus insulin glargine in patients with type 2 diabetes and moderate-to-severe chronic kidney disease (AWARD-7): a multicentre, open-label, randomised trial. Lancet Diabetes Endocrinol. 2018;6(8):605–17. [PubMed: 29910024]
183.
Stratton IM, Cull CA, Adler AI, Matthews DR, Neil HA, Holman RR. Additive effects of glycaemia and blood pressure exposure on risk of complications in type 2 diabetes: a prospective observational study (UKPDS 75). Diabetologia. 2006;49(8):1761–9. [PubMed: 16736131]
184.
Zoungas S, de Galan BE, Ninomiya T, Grobbee D, Hamet P, Heller S, et al. Combined effects of routine blood pressure lowering and intensive glucose control on macrovascular and microvascular outcomes in patients with type 2 diabetes: New results from the ADVANCE trial. Diabetes care. 2009;32(11):2068–74. [PMC free article: PMC2768202] [PubMed: 19651921]
185.
Randomised placebo-controlled trial of lisinopril in normotensive patients with insulin-dependent diabetes and normoalbuminuria or microalbuminuria. The EUCLID Study Group. Lancet. 1997;349(9068):1787–92. [PubMed: 9269212]
186.
Bilous R, Chaturvedi N, Sjolie AK, Fuller J, Klein R, Orchard T, et al. Effect of candesartan on microalbuminuria and albumin excretion rate in diabetes: three randomized trials. Ann Intern Med. 2009;151(1):11-20, W3-4. [PubMed: 19451554]
187.
Group ACEIiDNT. Should all patients with type 1 diabetes mellitus and microalbuminuria receive angiotensin-converting enzyme inhibitors? A meta-analysis of individual patient data. Ann Intern Med. 2001;134(5):370–9. [PubMed: 11242497]
188.
Lewis EJ, Hunsicker LG, Bain RP, Rohde RD. The effect of angiotensin-converting-enzyme inhibition on diabetic nephropathy. The Collaborative Study Group. N Engl J Med. 1993;329(20):1456–62. [see comments] [published erratum appears in N Engl J Med 1993 Jan 13;330(2):152] [PubMed: 8413456]
189.
Andersen S, Tarnow L, Rossing P, Hansen BV, Parving HH. Renoprotective effects of angiotensin II receptor blockade in type 1 diabetic patients with diabetic nephropathy. Kidney international. 2000;57(2):601–6. [PubMed: 10652037]
190.
Kasiske BL, Kalil RS, Ma JZ, Liao M, Keane WF. Effect of antihypertensive therapy on the kidney in patients with diabetes: a meta-regression analysis. Ann Intern Med. 1993;118(2):129–38. [PubMed: 8416309]
191.
Remuzzi G, Benigni A. Progression of proteinuric diabetic and nondiabetic renal diseases: a possible role for renal endothelin. Kidney Int Suppl. 1997;58:S66–8. [PubMed: 9067947]
192.
Zatz R, Dunn BR, Meyer TW, Anderson S, Rennke HG, Brenner BM. Prevention of diabetic glomerulopathy by pharmacological amelioration of glomerular capillary hypertension. The Journal of clinical investigation. 1986;77(6):1925–30. [PMC free article: PMC370553] [PubMed: 3011862]
193.
Andersen S, Blouch K, Bialek J, Deckert M, Parving HH, Myers BD. Glomerular permselectivity in early stages of overt diabetic nephropathy. Kidney international. 2000;58(5):2129–37. [PubMed: 11044234]
194.
Parving HH, Hommel E, Jensen BR, Hansen HP. Long-term beneficial effect of ACE inhibition on diabetic nephropathy in normotensive type 1 diabetic patients. Kidney international. 2001;60(1):228–34. [PubMed: 11422755]
195.
Tight blood pressure control and risk of macrovascular and microvascular complications in type 2 diabetes: UKPDS 38. UK Prospective Diabetes Study Group. Bmj. 1998;317(7160):703–13. [see comments] [published erratum appears in BMJ 1999 Jan 2;318(7175):29] [PMC free article: PMC28659] [PubMed: 9732337]
196.
Effects of ramipril on cardiovascular and microvascular outcomes in people with diabetes mellitus: results of the HOPE study and MICRO-HOPE substudy. Heart Outcomes Prevention Evaluation Study Investigators. Lancet. 2000;355(9200):253–9. [PubMed: 10675071]
197.
Ruggenenti P, Fassi A, Ilieva AP, Bruno S, Iliev IP, Brusegan V, et al. Preventing microalbuminuria in type 2 diabetes. N Engl J Med. 2004;351(19):1941–51. [PubMed: 15516697]
198.
Patel A, Group AC, MacMahon S, Chalmers J, Neal B, Woodward M, et al. Effects of a fixed combination of perindopril and indapamide on macrovascular and microvascular outcomes in patients with type 2 diabetes mellitus (the ADVANCE trial): a randomised controlled trial. Lancet. 2007;370(9590):829–40. [PubMed: 17765963]
199.
Persson F, Lindhardt M, Rossing P, Parving HH. Prevention of microalbuminuria using early intervention with renin-angiotensin system inhibitors in patients with type 2 diabetes: A systematic review. J Renin Angiotensin Aldosterone Syst. 2016;17(3) [PMC free article: PMC5843870] [PubMed: 27488274]
200.
Haller H, Ito S, Izzo JL Jr, Januszewicz A, Katayama S, Menne J, et al. Olmesartan for the delay or prevention of microalbuminuria in type 2 diabetes. N Engl J Med. 2011;364(10):907–17. [PubMed: 21388309]
201.
Strippoli GF, Craig M, Schena FP, Craig JC. Antihypertensive agents for primary prevention of diabetic nephropathy. Journal of the American Society of Nephrology : JASN. 2005;16(10):3081–91. [PubMed: 16135776]
202.
Parving HH, Lehnert H, Brochner-Mortensen J, Gomis R, Andersen S, Arner P. The effect of irbesartan on the development of diabetic nephropathy in patients with type 2 diabetes. N Engl J Med. 2001;345(12):870–8. [PubMed: 11565519]
203.
Brenner BM, Cooper ME, de Zeeuw D, Keane WF, Mitch WE, Parving HH, et al. Effects of losartan on renal and cardiovascular outcomes in patients with type 2 diabetes and nephropathy. N Engl J Med. 2001;345(12):861–9. [PubMed: 11565518]
204.
Lewis EJ, Hunsicker LG, Clarke WR, Berl T, Pohl MA, Lewis JB, et al. Renoprotective effect of the angiotensin-receptor antagonist irbesartan in patients with nephropathy due to type 2 diabetes. N Engl J Med. 2001;345(12):851–60. [PubMed: 11565517]
205.
de Boer IH, Bangalore S, Benetos A, Davis AM, Michos ED, Muntner P, et al. Diabetes and Hypertension: A Position Statement by the American Diabetes Association. Diabetes care. 2017;40(9):1273–84. [PubMed: 28830958]
206.
Miao Y, Dobre D, Heerspink HJ, Brenner BM, Cooper ME, Parving HH, et al. Increased serum potassium affects renal outcomes: a post hoc analysis of the Reduction of Endpoints in NIDDM with the Angiotensin II Antagonist Losartan (RENAAL) trial. Diabetologia. 2011;54(1):44–50. [PMC free article: PMC2995871] [PubMed: 20882268]
207.
Holtkamp FA, de Zeeuw D, Thomas MC, Cooper ME, de Graeff PA, Hillege HJ, et al. An acute fall in estimated glomerular filtration rate during treatment with losartan predicts a slower decrease in long-term renal function. Kidney international. 2011;80(3):282–7. [PubMed: 21451458]
208.
Oxlund CS, Henriksen JE, Tarnow L, Schousboe K, Gram J, Jacobsen IA. Low dose spironolactone reduces blood pressure in patients with resistant hypertension and type 2 diabetes mellitus: a double blind randomized clinical trial. J Hypertens. 2013;31(10):2094–102. [PubMed: 24107738]
209.
Adler AI, Stratton IM, Neil HA, Yudkin JS, Matthews DR, Cull CA, et al. Association of systolic blood pressure with macrovascular and microvascular complications of type 2 diabetes (UKPDS 36): prospective observational study. BMJ. 2000;321(7258):412–9. [PMC free article: PMC27455] [PubMed: 10938049]
210.
de Galan BE, Perkovic V, Ninomiya T, Pillai A, Patel A, Cass A, et al. Lowering blood pressure reduces renal events in type 2 diabetes. Journal of the American Society of Nephrology : JASN. 2009;20(4):883–92. [PMC free article: PMC2663832] [PubMed: 19225038]
211.
Sim JJ, Shi J, Kovesdy CP, Kalantar-Zadeh K, Jacobsen SJ. Impact of achieved blood pressures on mortality risk and end-stage renal disease among a large, diverse hypertension population. J Am Coll Cardiol. 2014;64(6):588–97. [PMC free article: PMC4136516] [PubMed: 25104529]
212.
Jacobsen P, Andersen S, Rossing K, Jensen BR, Parving HH. Dual blockade of the renin-angiotensin system versus maximal recommended dose of ACE inhibition in diabetic nephropathy. Kidney international. 2003;63(5):1874–80. [PubMed: 12675866]
213.
Mogensen CE, Neldam S, Tikkanen I, Oren S, Viskoper R, Watts RW, et al. Randomised controlled trial of dual blockade of renin-angiotensin system in patients with hypertension, microalbuminuria, and non-insulin dependent diabetes: the candesartan and lisinopril microalbuminuria (CALM) study. BMJ. 2000;321(7274):1440–4. [PMC free article: PMC27545] [PubMed: 11110735]
214.
Parving HH, Brenner BM, McMurray JJ, de Zeeuw D, Haffner SM, Solomon SD, et al. Cardiorenal end points in a trial of aliskiren for type 2 diabetes. N Engl J Med. 2012;367(23):2204–13. [PubMed: 23121378]
215.
Fried LF, Emanuele N, Zhang JH, Brophy M, Conner TA, Duckworth W, et al. Combined angiotensin inhibition for the treatment of diabetic nephropathy. N Engl J Med. 2013;369(20):1892–903. [PubMed: 24206457]
216.
Mann JF, Schmieder RE, McQueen M, Dyal L, Schumacher H, Pogue J, et al. Renal outcomes with telmisartan, ramipril, or both, in people at high vascular risk (the ONTARGET study): a multicentre, randomised, double-blind, controlled trial. Lancet. 2008;372(9638):547–53. [PubMed: 18707986]
217.
Ren F, Tang L, Cai Y, Yuan X, Huang W, Luo L, et al. Meta-analysis: the efficacy and safety of combined treatment with ARB and ACEI on diabetic nephropathy. Ren Fail. 2015;37(4):548–61. [PubMed: 25707526]
218.
Tofte N, Lindhardt M, Adamova K, Bakker SJL, Beige J, Beulens JWJ, et al. Early detection of diabetic kidney disease by urinary proteomics and subsequent intervention with spironolactone to delay progression (PRIORITY): a prospective observational study and embedded randomised placebo-controlled trial. Lancet Diabetes Endocrinol. 2020;8(4):301–12. [PubMed: 32135136]
219.
Currie G, Taylor AH, Fujita T, Ohtsu H, Lindhardt M, Rossing P, et al. Effect of mineralocorticoid receptor antagonists on proteinuria and progression of chronic kidney disease: a systematic review and meta-analysis. BMC nephrology. 2016;17(1):127. [PMC free article: PMC5015203] [PubMed: 27609359]
220.
Ito S, Shikata K, Nangaku M, Okuda Y, Sawanobori T. Efficacy and Safety of Esaxerenone (CS-3150) for the Treatment of Type 2 Diabetes with Microalbuminuria: A Randomized, Double-Blind, Placebo-Controlled, Phase II Trial. Clinical journal of the American Society of Nephrology : CJASN. 2019;14(8):1161–72. [PMC free article: PMC6682830] [PubMed: 31248950]
221.
Bakris GL, Agarwal R, Chan JC, Cooper ME, Gansevoort RT, Haller H, et al. Effect of Finerenone on Albuminuria in Patients With Diabetic Nephropathy: A Randomized Clinical Trial. JAMA. 2015;314(9):884–94. [PubMed: 26325557]
222.
Bakris GL, Agarwal R, Anker SD, Pitt B, Ruilope LM, Rossing P, et al. Effect of Finerenone on Chronic Kidney Disease Outcomes in Type 2 Diabetes. N Engl J Med. 2020;383(23):2219–29. [PubMed: 33264825]
223.
Pitt B, Filippatos G, Agarwal R, Anker SD, Bakris GL, Rossing P, et al. Cardiovascular Events with Finerenone in Kidney Disease and Type 2 Diabetes. N Engl J Med. 2021;385(24):2252–63. [PubMed: 34449181]
224.
Rossing P, Filippatos G, Agarwal R, Anker SD, Pitt B, Ruilope LM, et al. Finerenone in Predominantly Advanced CKD and Type 2 Diabetes With or Without Sodium-Glucose Cotransporter-2 Inhibitor Therapy. Kidney Int Rep. 2022;7(1):36–45. [PMC free article: PMC8720648] [PubMed: 35005312]
225.
Kwakernaak AJ, Krikken JA, Binnenmars SH, Visser FW, Hemmelder MH, Woittiez AJ, et al. Effects of sodium restriction and hydrochlorothiazide on RAAS blockade efficacy in diabetic nephropathy: a randomised clinical trial. Lancet Diabetes Endocrinol. 2014;2(5):385–95. [PubMed: 24795252]
226.
Lambers Heerspink HJ, Holtkamp FA, Parving HH, Navis GJ, Lewis JB, Ritz E, et al. Moderation of dietary sodium potentiates the renal and cardiovascular protective effects of angiotensin receptor blockers. Kidney international. 2012;82(3):330–7. [PubMed: 22437412]
227.
Herrington WG, Preiss D, Haynes R, von Eynatten M, Staplin N, Hauske SJ, et al. The potential for improving cardio-renal outcomes by sodium-glucose co-transporter-2 inhibition in people with chronic kidney disease: a rationale for the EMPA-KIDNEY study. Clin Kidney J. 2018;11(6):749–61. [PMC free article: PMC6275453] [PubMed: 30524708]
228.
de Zeeuw D, Coll B, Andress D, Brennan JJ, Tang H, Houser M, et al. The endothelin antagonist atrasentan lowers residual albuminuria in patients with type 2 diabetic nephropathy. Journal of the American Society of Nephrology : JASN. 2014;25(5):1083–93. [PMC free article: PMC4005314] [PubMed: 24722445]
229.
Mann JF, Green D, Jamerson K, Ruilope LM, Kuranoff SJ, Littke T, et al. Avosentan for overt diabetic nephropathy. Journal of the American Society of Nephrology : JASN. 2010;21(3):527–35. [PMC free article: PMC2831858] [PubMed: 20167702]
230.
Heerspink HJL, Parving HH, Andress DL, Bakris G, Correa-Rotter R, Hou FF, et al. Atrasentan and renal events in patients with type 2 diabetes and chronic kidney disease (SONAR): a double-blind, randomised, placebo-controlled trial. Lancet. 2019;393(10184):1937–47. [PubMed: 30995972]
231.
Garsen M, Lenoir O, Rops AL, Dijkman HB, Willemsen B, van Kuppevelt TH, et al. Endothelin-1 Induces Proteinuria by Heparanase-Mediated Disruption of the Glomerular Glycocalyx. Journal of the American Society of Nephrology : JASN. 2016;27(12):3545–51. [PMC free article: PMC5118481] [PubMed: 27026367]
232.
Nezu U, Kamiyama H, Kondo Y, Sakuma M, Morimoto T, Ueda S. Effect of low-protein diet on kidney function in diabetic nephropathy: meta-analysis of randomised controlled trials. BMJ Open. 2013;3(5) [PMC free article: PMC3664345] [PubMed: 23793703]
233.
Hansen HP, Tauber-Lassen E, Jensen BR, Parving HH. Effect of dietary protein restriction on prognosis in patients with diabetic nephropathy. Kidney international. 2002;62(1):220–8. [PubMed: 12081581]
234.
Jun M, Zhu B, Tonelli M, Jardine MJ, Patel A, Neal B, et al. Effects of fibrates in kidney disease: a systematic review and meta-analysis. J Am Coll Cardiol. 2012;60(20):2061–71. [PubMed: 23083786]
235.
Gaede P, Hansen HP, Parving HH, Pedersen O. Impact of low-dose acetylsalicylic acid on kidney function in type 2 diabetic patients with elevated urinary albumin excretion rate. Nephrol Dial Transplant. 2003;18(3):539–42. [PubMed: 12584276]
236.
Kreutz R, Camm AJ, Rossing P. Concomitant diabetes with atrial fibrillation and anticoagulation management considerations. Eur Heart J Suppl. 2020;22 Suppl O:O78–O86. [PMC free article: PMC7753879] [PubMed: 33380946]
237.
Hernandez AV, Bradley G, Khan M, Fratoni A, Gasparini A, Roman YM, et al. Rivaroxaban vs. warfarin and renal outcomes in non-valvular atrial fibrillation patients with diabetes. Eur Heart J Qual Care Clin Outcomes. 2020;6(4):301–7. [PubMed: 31432074]
238.
Look ARG. Effect of a long-term behavioural weight loss intervention on nephropathy in overweight or obese adults with type 2 diabetes: a secondary analysis of the Look AHEAD randomised clinical trial. Lancet Diabetes Endocrinol. 2014;2(10):801–9. [PMC free article: PMC4443484] [PubMed: 25127483]
239.
Tirosh A, Golan R, Harman-Boehm I, Henkin Y, Schwarzfuchs D, Rudich A, et al. Renal function following three distinct weight loss dietary strategies during 2 years of a randomized controlled trial. Diabetes care. 2013;36(8):2225–32. [PMC free article: PMC3714527] [PubMed: 23690533]
240.
Jackson S, le Roux CW, Docherty NG. Bariatric surgery and microvascular complications of type 2 diabetes mellitus. Curr Atheroscler Rep. 2014;16(11):453. [PubMed: 25239035]
241.
Bjornstad P, Nehus E, Jenkins T, Mitsnefes M, Moxey-Mims M, Dixon JB, et al. Five-year kidney outcomes of bariatric surgery differ in severely obese adolescents and adults with and without type 2 diabetes. Kidney international. 2020;97(5):995–1005. [PMC free article: PMC7183894] [PubMed: 32229096]
242.
Pugliese G, Penno G, Natali A, Barutta F, Di Paolo S, Reboldi G, et al. Diabetic kidney disease: new clinical and therapeutic issues. Joint position statement of the Italian Diabetes Society and the Italian Society of Nephrology on "The natural history of diabetic kidney disease and treatment of hyperglycemia in patients with type 2 diabetes and impaired renal function". J Nephrol. 2020;33(1):9–35. [PMC free article: PMC7007429] [PubMed: 31576500]
243.
Petrie JR, Rossing PR, Campbell IW. Metformin and cardiorenal outcomes in diabetes: A reappraisal. Diabetes Obes Metab. 2020;22(6):904–15. [PMC free article: PMC7317924] [PubMed: 32009286]
244.
Thomas MC, Cooper ME, Rossing K, Parving HH. Anaemia in diabetes: Is there a rationale to TREAT? Diabetologia. 2006;49(6):1151–7. [PubMed: 16586069]
245.
Pfeffer MA, Burdmann EA, Chen CY, Cooper ME, de Zeeuw D, Eckardt KU, et al. A trial of darbepoetin alfa in type 2 diabetes and chronic kidney disease. N Engl J Med. 2009;361(21):2019–32. [PubMed: 19880844]
246.
Smart NA, Dieberg G, Ladhani M, Titus T. Early referral to specialist nephrology services for preventing the progression to end-stage kidney disease. Cochrane Database Syst Rev. 2014;(6):CD007333. [PubMed: 24938824]
247.
Lim LL, Lau ESH, Ozaki R, Chung H, Fu AWC, Chan W, et al. Association of technologically assisted integrated care with clinical outcomes in type 2 diabetes in Hong Kong using the prospective JADE Program: A retrospective cohort analysis. PLoS Med. 2020;17(10):e1003367. [PMC free article: PMC7531841] [PubMed: 33007052]
248.
Tu ST, Chang SJ, Chen JF, Tien KJ, Hsiao JY, Chen HC, et al. Prevention of diabetic nephropathy by tight target control in an asian population with type 2 diabetes mellitus: a 4-year prospective analysis. Arch Intern Med. 2010;170(2):155–61. [PubMed: 20101010]
249.
Gaede P, Lund-Andersen H, Parving HH, Pedersen O. Effect of a multifactorial intervention on mortality in type 2 diabetes. N Engl J Med. 2008;358(6):580–91. [PubMed: 18256393]
250.
Chan JC, So WY, Yeung CY, Ko GT, Lau IT, Tsang MW, et al. Effects of structured versus usual care on renal endpoint in type 2 diabetes: the SURE study: a randomized multicenter translational study. Diabetes care. 2009;32(6):977–82. [PMC free article: PMC2681013] [PubMed: 19460913]
251.
Gaede P, Oellgaard J, Carstensen B, Rossing P, Lund-Andersen H, Parving HH, et al. Years of life gained by multifactorial intervention in patients with type 2 diabetes mellitus and microalbuminuria: 21 years follow-up on the Steno-2 randomised trial. Diabetologia. 2016;59(11):2298–307. [PMC free article: PMC5506099] [PubMed: 27531506]
252.
Gaede P, Oellgaard J, Kruuse C, Rossing P, Parving HH, Pedersen O. Beneficial impact of intensified multifactorial intervention on risk of stroke: outcome of 21 years of follow-up in the randomised Steno-2 Study. Diabetologia. 2019;62(9):1575–80. [PMC free article: PMC6677690] [PubMed: 31154479]
253.
Oellgaard J, Gaede P, Rossing P, Persson F, Parving HH, Pedersen O. Intensified multifactorial intervention in type 2 diabetics with microalbuminuria leads to long-term renal benefits. Kidney international. 2017;91(4):982–8. [PubMed: 28187983]
254.
Oellgaard J, Gaede P, Rossing P, Rorth R, Kober L, Parving HH, et al. Reduced risk of heart failure with intensified multifactorial intervention in individuals with type 2 diabetes and microalbuminuria: 21 years of follow-up in the randomised Steno-2 study. Diabetologia. 2018;61(8):1724–33. [PMC free article: PMC6061176] [PubMed: 29850922]
255.
Mathiesen ER. Diabetic nephropathy in pregnancy: new insights from a retrospective cohort study. Diabetologia. 2015;58(4):649–50. [PubMed: 25690842]
256.
Klemetti MM, Laivuori H, Tikkanen M, Nuutila M, Hiilesmaa V, Teramo K. Obstetric and perinatal outcome in type 1 diabetes patients with diabetic nephropathy during 1988-2011. Diabetologia. 2015;58(4):678–86. [PubMed: 25575985]
257.
Damm JA, Asbjornsdottir B, Callesen NF, Mathiesen JM, Ringholm L, Pedersen BW, et al. Diabetic nephropathy and microalbuminuria in pregnant women with type 1 and type 2 diabetes: prevalence, antihypertensive strategy, and pregnancy outcome. Diabetes care. 2013;36(11):3489–94. [PMC free article: PMC3816914] [PubMed: 24009298]
258.
Nielsen LR, Damm P, Mathiesen ER. Improved pregnancy outcome in type 1 diabetic women with microalbuminuria or diabetic nephropathy: effect of intensified antihypertensive therapy? Diabetes care. 2009;32(1):38–44. [PMC free article: PMC2606826] [PubMed: 18945922]
Copyright © 2000-2024, MDText.com, Inc.

This electronic version has been made freely available under a Creative Commons (CC-BY-NC-ND) license. A copy of the license can be viewed at http://creativecommons.org/licenses/by-nc-nd/2.0/.

Bookshelf ID: NBK279103PMID: 25905328

Views

  • PubReader
  • Print View
  • Cite this Page

LINKS TO WWW.ENDOTEXT.ORG

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Similar articles in PubMed

See reviews...See all...

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...