Skip to main content

Main menu

  • Home
  • Content
    • Published Ahead of Print
    • Current Issue
    • JASN Podcasts
    • Article Collections
    • Archives
    • Kidney Week Abstracts
    • Saved Searches
  • Authors
    • Submit a Manuscript
    • Author Resources
  • Editorial Team
  • Editorial Fellowship
    • Editorial Fellowship Team
    • Editorial Fellowship Application Process
  • More
    • About JASN
    • Advertising
    • Alerts
    • Feedback
    • Impact Factor
    • Reprints
    • Subscriptions
  • ASN Kidney News
  • Other
    • ASN Publications
    • CJASN
    • Kidney360
    • Kidney News Online
    • American Society of Nephrology

User menu

  • Subscribe
  • My alerts
  • Log in
  • Log out
  • My Cart

Search

  • Advanced search
American Society of Nephrology
  • Other
    • ASN Publications
    • CJASN
    • Kidney360
    • Kidney News Online
    • American Society of Nephrology
  • Subscribe
  • My alerts
  • Log in
  • Log out
  • My Cart
Advertisement
American Society of Nephrology

Advanced Search

  • Home
  • Content
    • Published Ahead of Print
    • Current Issue
    • JASN Podcasts
    • Article Collections
    • Archives
    • Kidney Week Abstracts
    • Saved Searches
  • Authors
    • Submit a Manuscript
    • Author Resources
  • Editorial Team
  • Editorial Fellowship
    • Editorial Fellowship Team
    • Editorial Fellowship Application Process
  • More
    • About JASN
    • Advertising
    • Alerts
    • Feedback
    • Impact Factor
    • Reprints
    • Subscriptions
  • ASN Kidney News
  • Follow JASN on Twitter
  • Visit ASN on Facebook
  • Follow JASN on RSS
  • Community Forum
Reviews
You have accessRestricted Access

Mouse Models of Diabetic Nephropathy

Matthew D. Breyer, Erwin Böttinger, Frank C. Brosius, Thomas M. Coffman, Raymond C. Harris, Charles W. Heilig, Kumar Sharma and ; for the AMDCC
JASN January 2005, 16 (1) 27-45; DOI: https://doi.org/10.1681/ASN.2004080648
Matthew D. Breyer
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Erwin Böttinger
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Frank C. Brosius III
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Thomas M. Coffman
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Raymond C. Harris
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Charles W. Heilig
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Kumar Sharma
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data Supps
  • Info & Metrics
  • View PDF
Loading

Abstract

Mice provide an experimental model of unparalleled flexibility for studying mammalian diseases. Inbred strains of mice exhibit substantial differences in their susceptibility to the renal complications of diabetes. Much remains to be established regarding the course of diabetic nephropathy (DN) in mice as well as defining those strains and/or mutants that are most susceptible to renal injury from diabetes. Through the use of the unique genetic reagents available in mice (including knockouts and transgenics), the validation of a mouse model reproducing human DN should significantly facilitate the understanding of the underlying genetic mechanisms that contribute to the development of DN. Establishment of an authentic mouse model of DN will undoubtedly facilitate testing of translational diagnostic and therapeutic interventions in mice before testing in humans.

Diabetic nephropathy (DN) is the major single cause of ESRD in the United States (1), with costs for care of these patients projected to be $12 billion/yr by 2010 (1). Despite the high prevalence of DN, only 20 to 40% of all diabetic patients are prone to developing kidney failure, and family-based studies suggest that a significant genetic component confers risk for DN (2–4). Studies of diabetic mice suggest that, like people, mice exhibit differential susceptibility to diabetes and renal and cardiovascular diseases (5–7). In mice, identification of a strain that is prone to disease provides the relevant discriminator rather than identification of an individual who is at risk for diabetic complications. However, in contrast to people, each inbred mouse strain represents a genetically homogeneous and readily replenished resource that is amenable to repeated experimental study.

Biomedical experimentation in mice affords significant advantages over experimentation in other species. These advantages include the development of diverse and unique genetic resources, including completion of a detailed map of murine genomic sequence that is freely available through the internet, as well as >450 inbred strains of mice that have been generated over the past century, with each strain genetically being homogeneous and offering a unique array of phenotypes (8,9). The availability of murine embryonic stem cells has also provided the ability to disrupt the expression and function of specific preselected genes (10–12). Finally, repositories of mice that bear multiple mutations that alter function in each known gene are gradually being assembled (13–15). These unique resources have the potential to significantly facilitate studies into the pathogenesis of disease in mice. Through the use of the unique genetic reagents that are available in mice (including knockouts and transgenics), the identification of a robust mouse model of DN should significantly facilitate understanding of the underlying genetic mechanisms that contribute to the development of DN in people as well as in mice. Establishment of a valid mouse model of DN should also facilitate testing of translational diagnostic and therapeutic interventions in mice before testing in humans.

Working Definition of DN

In humans, DN manifests as a clinical syndrome that is composed of albuminuria, progressively declining GFR, and increased risk for cardiovascular disease (16,17). The occurrence of cardiovascular disease is an integral component of DN and underscores the systemic nature of this disorder, of which nephropathy is only one aspect. Another key aspect of DN in humans is that it is a late complication of diabetes, occurring progressively in susceptible people only after 15 to 25 yr of diabetes (18–20). Because of issues of cost and convenience, most studies of DN in mice have focused on the earlier harbingers of DN, including the development of albuminuria and histopathologic changes, and have not explicitly used renal insufficiency as an end point (21,22).

Diabetic albuminuria in humans is associated with the development of characteristic histopathologic features, including thickening of the glomerular basement membrane (GBM) and mesangial expansion. As albuminuria progresses and renal insufficiency ensues, glomerulosclerosis, arteriolar hyalinosis, and tubulointerstitial fibrosis develop (23). These pathologic features correlate well with GFR in humans with diabetes and kidney disease (24,25) and would be important features in a robust mouse model of DN.

Characterization of DN in Mice

The major deficiency in animal models of DN is the absence of renal failure. Whereas animal models of diabetic kidney disease exhibit albuminuria and some of the characteristic pathologic changes, reports of renal failure resulting from diabetes in mice or rats are lacking. Furthermore, associated increased risk for cardiovascular disease, neuropathy, and retinopathy have been poorly characterized. Whether the inability to detect renal failure in mice is simply a consequence of studying mice with established diabetes for an insufficient time period or the absence of renal failure reflects an intrinsic resistance to nephropathy of the strains studied thus far remains uncertain.

In contrast to the absence of renal failure, several of the more short-term consequences, including the development of glomerular hyperfiltration, increased albuminuria, and some of the characteristic histopathologic changes, can be detected in animal models. Human DN proceeds through several distinct pathophysiologic stages, including an early stage of glomerular hyperfiltration, followed by the so-called silent phase in which GFR returns to normal (26). This is followed by the sequential development of microalbuminuria, dipstick-positive proteinuria, and then a progressive decline in GFR leading to ESRD (20,27,28). An optimistic view is that the detection of the early functional and histopathologic changes in mouse models of diabetes reflects inadequate duration of hyperglycemia before the study of these models.

Assessment of Hyperglycemia in Mice

In humans, the degree of hyperglycemia is a critical determinant of risk for developing DN (29,30). Similar studies have not been systematically performed in mice, but in small studies, nephropathy seems to correlate with the severity of hyperglycemia. Measurement of glucose control in mice is generally achieved by determination of fasting blood glucose or determination of glycosylated hemoglobin.

Fasting Blood Glucose

Because mice are nocturnal feeders, an overnight fast before measuring blood glucose usually translates into a more prolonged fast of approximately 24 h. This 24-h fast can activate several physiologic counterregulatory mechanisms that obscure the reliability of glucose readings, as a result of this significant distress. Because of this, fasting should be started on the morning of blood sampling. The National Institutes of Health (NIH) has established mouse metabolic phenotyping centers (www.mmpc.org) that have developed a standard protocol that is composed of a fast between 7 a.m. and 1 p.m. with blood drawn at 1 p.m. This protocol has been adapted by the Animal Models of Diabetic Complications Consortium (AMDCC; www.amdcc.org), another NIH consortium whose goal is to identify and establish mouse models of diabetic complications.

Glycosylated Hemoglobin (Including Hemoglobin A1C)

When carbohydrates become nonenzymatically linked to hemoglobin, these species can be separated from normal adult hemoglobin (designated HgbA0) either by altered charge or by formation of a stable complex between the coplanar cis-diol groups of glycated hemoglobin in an affinity column and are collectively designated as HgbA1 (31,32). These hemoglobins can be separated further into HgbA1a, HgbA1b, and HgbA1C, among which HgbA1c is the main glycated form in humans (33). HgbA1c has been used widely to assess glucose control in people, and by extrapolation, it has been widely used to assess glucose control in diabetic mice. Studies in diabetic mice are consistent with the validity of the use of HgbA1C to assess glucose control (34,35), although levels of the various glycosylated hemoglobin assays have not been correlated rigorously to fasting blood glucose values in mice.

Whereas HgA1C correlates well with glucose control in people (36), there seem to be some human populations that exhibit substantially different HgbA1C values despite similar levels of glucose control (37). This observation has been attributed to differences in red cell life span rather than rates of glycation (38). Another intriguing possibility is that people with different endogenous rates of glycation exist despite similar levels of hyperglycemia and that “high glycators” exhibit greater susceptibility to DN and retinopathy (39). Variability in red cell life span among strains of mice seems likely and may have a significant impact on interpretation of HgbA1c levels (35,40,41).

Assessment of Renal Function in Mice

GFR

A serious issue impeding the evaluation of renal function in mice has been the lack of a simple, reproducible method to estimate GFR in conscious mice. As in humans, blood urea nitrogen levels are extremely sensitive to extracellular fluid volume depletion and may be artificially elevated in the individual with diabetes from volume depletion resulting from the osmotic diuresis of hyperglycemia. The use of serum creatinine as a tool to evaluate renal function in mice has also been called into question since Meyer et al. (42) reported that creatinine levels in plasma of mice measured by the Jaffé alkaline picrate method yielded significantly higher levels (three to five times) than those measured by HPLC. These studies concluded that the Jaffé method greatly overestimates the plasma creatinine levels, and attributed this increase to noncreatinine chromagens. Other studies have found similar assay-dependent variability in the accuracy of creatinine as a measure of renal function in mice (43,44) and in humans (45).

The impact of these cross-reacting chromagens on apparent levels of plasma creatinine may be significantly affected by ketoacidosis (e.g., as occurs in diabetes) (46,47). Because absolute creatinine levels as measured by HPLC are substantially lower in mice (0.128 ± 0.026 to 0.207 ± 0.012 mg/dl) than in humans (48,49), the impact of these chromagens on apparent creatinine values may be significantly greater in normal mice than in normal humans. In mice with normal renal function, the picric acid–based method routinely overestimates HPLC-determined serum creatinine by two- to fivefold, yielding apparent serum creatinine values of 0.6 to 1.0 mg/dl (48,49). Conversely, enzymatic creatininase methods coupled to a peroxidase indicator more closely agree with HPLC methods, providing another potentially useful method for determination of serum creatinine in mice (44). Despite the availability of this information regarding the inaccuracy of serum creatinine measured by the Jaffé reaction, the assay remains widely used by the biomedical research community to assess renal function in mice. Thus, artifactual increases in serum creatinine likely contribute to reports of decreased creatinine clearance in many studies that examine DN in mice.

With the increasing use of mice as an experimental model of diabetic kidney disease, awareness of these deficiencies must be heightened. Several approaches have been adopted to re-address this problem. New methods to accurately measure plasma creatinine measurements by HPLC have been established and have confirmed significant overestimation of plasma creatinine using picrate-based methods. When compared with inulin clearance, creatinine clearance determinations using the Jaffé reaction underestimates GFR by >50% (48,49).

Measurement of GFR using inulin clearance, the traditional gold-standard measurement, has also been adapted for use in mice. Clearance of FITC-labeled inulin has been measured in both anesthetized and conscious mice (50,51). Serial measurements of inulin clearance can be determined in conscious mice over a period of months using an intravenous bolus FITC inulin and measuring the decay rate of inulin in plasma (50). Using these techniques, substantial differences in GFR between mice of different strains and sexes have been reported (50,52,53). Consideration of the effects of surgical preparation and/or anesthesia is appropriate when GFR determination is performed in anesthetized mice. It remains to be determined how well HPLC-determined creatinine clearance correlates with FITC inulin clearance in individual mice. Nevertheless, previously published studies in which GFR has been estimated in mice using plasma creatinine values determined by picrate-based methods should be reevaluated carefully in light of the aforementioned problems.

BP

There are several proven methods for monitoring BP in mice that can be applied to the study of diabetic mouse models. Indirect measurements using tail-cuff manometry systems are simple and noninvasive. Their reliability and reproducibility have been well documented across a number of experimental systems (54). These systems measure BP in conscious mice and are inexpensive and adaptable to most laboratory settings. However, they lack the precision and the sensitivity of systems that directly measure intra-arterial pressure. This approach is also complicated by physiologically apparent stress in tested mice that can affect the level of BP, but this can be minimized by training mice adequately beforehand. In our experience, tail-cuff manometry is a useful approach to screen for alterations of BP in mouse models, realizing that very modest differences may be missed because of the relative insensitivity of this method. Using this approach in studies under the auspices of the AMDCC, we have found that BP tend to be reduced in most strains of streptozotocin (STZ)-treated mice when significant hyperglycemia is present (54A). Tail-cuff measurements have also been reported by others in models of diet-induced diabetes and does not seem to be dramatically affected by diabetes (55) and genetically altered mice that are treated with STZ (56,57).

Intra-arterial pressures can also be measured directly in conscious mice. These measurements can be accomplished by cannulating arteries with catheters that are tunneled under the skin, exteriorized, and connected to external transducers and monitors via a tether apparatus. Although this approach requires significant surgical and technical proficiency, it is relatively inexpensive and accessible to most laboratories, providing a reasonable method for verifying BP differences that have been detected by indirect methods. However, it is difficult to maintain line patency for recording BP beyond 1 to 2 wk after catheter placement. The recent development of radiotelemetry units for intra-arterial BP measurements in mice has provided a more suitable approach for chronic measurement of intra-arterial pressures in conscious, unrestrained animals (58). These units provide the capacity to obtain direct and continuous measurements of intra-arterial pressure 24 h/d over a period of 2 to 3 mo. Accordingly, they should prove very useful for correlating levels of BP with the extent of kidney injury in experimental models. The major disadvantage of this system is expense. The initial capital outlay for a small-scale telemetry unit is typically twice that of most tail-cuff monitors. Furthermore, individual transmitters cost approximately $3000 each and must be refurbished periodically for an additional fee.

Albuminuria.

Before developing renal failure, humans with overt DN typically develop nephrotic-range albuminuria (albumin excretion rates exceeding 3 g/24 h). Preceding this, albumin excretion rates gradually and progressively increase over the course of years through a phase of microalbuminuria (between 30 and 300 mg/24 h that is negative by urine dipstick), to albuminuria >300 mg/24 h, a phase that is characterized by an overtly positive urine dipstick (19,59,60). A priori, it is expected that mice that develop DN should progress through similar phases so that the amount of albuminuria should roughly correlate with the severity of DN.

Albuminuria has been quantified in approximately 100 studies of DN in mice and in the absence of accurate measures of GFR probably remains the best functional surrogate for severity of DN. However, comparisons of the severity of nephropathy in the diverse models of diabetes reported are confounded by use of diverse methods (e.g., quantifying total urine protein, anti-rat antibodies or anti-human antibodies to measure mouse albumin on ELISA), differences in units of reporting (e.g., ng/min, μg/24 h, μg Alb/mg Cre; μgAlb/μMol creatinine, total urine protein μg/24 h, μgAlb/ml urine), and nonstandardized methods of urine collection.

Quantification of proteinuria in mice by dipstick is probably inaccurate (unpublished results). The exact reasons for this are not firmly established; however, the inaccuracy of the dipstick proteinuria test in mouse urine may be a consequence of sensitivity of this assay to abundant major urinary proteins, normally present in mouse urine (61,62). Albuminuria determined by an immunoassay generally provides a reliable assessment of proteinuria as a result of glomerular injury. Use of antibodies raised against nonmurine albumin may be particularly problematic as mouse albumin amino acid sequence is only 89% identical to rat albumin and 73% identical to human albumin, allowing for differences in the sensitivity of immunoassays. The AMDCC has chosen to use an anti-mouse albumin ELSIA kit and report albuminuria as μg/24 h urine collection or μg albumin/mg creatinine. A standardized assay used by consortium members is available at the AMDCC web site (www.amdcc.org).

The values of urine albumin excretion rates in mice that signify glomerular disease have not been firmly established. Furthermore, it is likely that significant variation in albumin excretion rates exist between inbred mouse strains. In the absence of well-defined normal values, a first approximation of abnormal albuminuria would be to extrapolate from human disease and test for values 10-fold greater than nondiabetic controls as reflective of incipient disease. Urine albumin excretion rates 100- to 1000-fold more than controls should reflect established diabetic renal disease. We have attempted to compare the severity of diabetic albuminuria in various strains and diabetic models studied by limiting our analysis to those studies performed on a 24-h urine collection and using assays specific for mouse albumin (Figure 1). A single study in KK-Ay mice that were fed a high-cholesterol diet suggested that a 1000-fold increase in albuminuria may be achieved in this model; however, whether renal failure ensues in this case remains to be determined (63).

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Summary of studies in the literature examining 24-h albumin excretion in mouse models of diabetes (#, reference number). For each study, a pair of bars represent mean ± SE. On the left, 24-hr albumin excretion in the diabetic cohort; on the right, the nondiabetic cohort. From left to right are albuminuria in C57BL/6 mice treated with multiple low-dose streptozotocin (STZ); C57BL6 high-dose STZ; C57BL6 db/db mice, a model of type 2 diabetes; four studies measuring albuminuria in the C57BLKS db/db strain show generally higher albuminuria; a single study examining 24-h albuminuria in the KK-Ay strain shows robust albuminuria.

Histopathology.

In human DN, renal histopathologic findings include diffuse thickening of the GBM between the endothelium and the podocyte, together with prominent mesangial expansion. This is accompanied by diffuse and sometimes nodular mesangial sclerosis, as well as arteriolar hyalinosis and tubulointerstitial fibrosis (24,25,64). The best histopathologic correlate of renal function in patients with diabetes is fractional volume of the mesangium, an index of mesangial expansion (24,25). Tubulointerstitial volume also correlates well with renal function.

Given the absence of documented renal failure in mouse models of DN, whether specific pathologic criteria are associated with or predictive of renal insufficiency cannot be readily formulated. GBM thickening has been reported in several but not all mouse models of DN (65–68). In mice, nodular glomerular sclerosis is generally absent as is arteriolar hyalinosis, two features generally seen only in people with more advanced renal disease (24). The reproducible identification of these two lesions in mouse models of DN remains a benchmark yet to be achieved.

Models of Type 1 Diabetes

High-Dose STZ

STZ-induced type 1 diabetes (Table 1) has been used widely as a model for DN; however, interpreting results in this model may be complicated by nonspecific toxicity of STZ. STZ was originally identified as an antibiotic (69) composed of a glucosamine-nitrosourea. It was soon recognized to cause diabetes in animals secondary to pancreatic β cell failure (70,71). STZ is presumed to be especially toxic for pancreatic β cells because its glucose moiety is avidly transported into β cells by GLUT2 (72); however, it may be toxic to a variety of other tissues. Given the relative resistance of mice to single doses of STZ (requiring approximately 150 to 200 mg/kg to obtain chronic hyperglycemia), substantial collateral tissue toxicity may occur (73–75). This model of type 1 diabetes typically develops albuminuria (66,76,77), even on the relatively sclerosis-resistant C57BL/6J background (see below); however, the potential for collateral tissue toxicity complicates the interpretation of the cause of this albuminuria. Evidence for robust oxidative stress in STZ-induced diabetes but not other hyperglycemic models of diabetes (e.g., db/db and KK mice) has been reported (78). Mice that receive high-dose STZ develop more albuminuria than mice that receive the low-dose STZ regimen (see Figure 1 and text below), despite exhibiting similar levels of hyperglycemia, also consistent with an STZ effect independent of hyperglycemia. Nevertheless, it should be noted that the aforementioned potential for nonspecific renal toxicity has not been proven rigorously and that the high-dose STZ model of diabetes is still widely accepted and commonly used. It also should be noted that reversibility of diabetic lesions after treatment of mice with insulin to normalize plasma glucose would provide another means of excluding nonspecific toxicity as a result of high-dose STZ.

View this table:
  • View inline
  • View popup
Table 1.

Some mouse models of diabetes studied for diabetic nephropathya

Low-Dose STZ

To mitigate nonspecific cytotoxicity, multiple low-dose STZ injections have be used to induce diabetes, causing repetitive low-grade β cell damage accompanied by lymphocytic infiltration of the pancreatic islets (79,80). This regimen typically calls for daily intraperitoneally injections of 40 to 50 mg/kg STZ for 5 d (71,81). It is notable that inbred strains of mice exhibit substantial differences in susceptibility to both β cell toxicity (82) and nonspecific toxic effects of low-dose STZ (71), so dose adjustments should be made. In general, when administered in strain-appropriate doses, the low-dose STZ regimen produces comparable levels of hyperglycemia as those obtained using the high-dose regimen. The levels of albuminuria (56,83) are generally lower with low-dose STZ than with high-dose STZ, possibly reflecting reduced direct renal toxicity (Figure 1). In addition, we have confirmed that there is no loss of podocyte markers such as WT-1 or apparent decrement in podocyte number 2 wk after completion of the low-dose STZ regimen in C57BL/6J mice (Siu et al., unpublished data), suggesting that this regimen has no toxic effects of on podocytes.

Our experience using low-dose STZ in mice has shown that hyperglycemia occurs within 2 wk of the low-dose STZ regimen (for the regimen used, see www.AMDCC.org). Albuminuria develops to a variable degree (depending on strain of mice; see below) within 5 wk. At this early time point, the major histologic feature is glomerular hypertrophy, so it is likely that hemodynamic changes contribute to the development of this early albuminuria. As time progresses, mesangial expansion and, in some strains, mesangial sclerosis develop. Only a minority of mouse strains (e.g., KK) develop evidence of arteriolar hyalinosis or nodular glomerulosclerosis. These changes occur later (15 to 30 wk of hyperglycemia). At these later time points, the diabetic mice may exhibit significant weight loss, possibly as a result of catabolic effects of insulin deficiency, as well as volume depletion associated with the osmotic diuresis. This may have a significant impact on subsequent survival and more long-term studies. To circumvent this occurrence, intermittent treatment with insulin, sufficient to reverse weight loss but maintain hyperglycemia, may be desirable.

Insulin-2 Akita

To circumvent the potential nonspecific tissue toxicity that occurs in the STZ model of type 1 diabetes, one may use the recently described insulin-2 Akita (Ins2Akita) mouse mutant model of type 1 diabetes (84–86). These mice develop pancreatic β cell failure as a result of β cell–selective proteotoxicity resulting from misfolding of insulin2 (87). The mice are commercially available through the Jackson Laboratories. Although originally reported to represent a model of maturity-onset diabetes of the young with insulin resistance, subsequent studies clearly show that this model is insulin responsive. Islets from Ins2Akita mice are depleted of β cells, and those remaining β cells release very little mature insulin. This finding together with the finding that mutant mice respond to exogenously administered insulin (84) indicates that Ins2Akita mice should serve as a substitute for mice that are rendered insulin dependent diabetic by treatment with alloxan or STZ.

The Ins2Akita mutation is autosomal dominant. Mice heterozygous for the Akita spontaneous mutation (Ins2Akita) are viable and fertile. Mice homozygous for the Ins2Akita allele exhibit failure to thrive and die within 1 to 2 mo. Symptoms in heterozygous mutant mice include hyperglycemia, hypoinsulinemia, polydipsia, and polyuria beginning at approximately 3 to 4 wk of age.

At present, this mutation exists on the C57BL/6 and C3H/He strains (88). Studies of Ins2Akita mice on the C57BL/6J background show that the hyperglycemia is sexually dimorphic, with the hyperglycemia being substantially worse in male mice than in female mice. As seen with other models of DN in the C57BL/6J strain, albuminuria is not a prominent feature (10.23 ± 2.73 μg/24 h in diabetic mice versus 13.9 ± 8.8 μg/24 h in controls) (89). Renal immunopathologic studies show significant deposition of IgA in the glomeruli of Akita mice (88); however, given the association of IgA deposition in glomeruli of up to 20% of humans with diabetes (90) and rat models of diabetes, the significance of these findings is uncertain (91–93). Overall, the severity of DN in the C57BL/6J Ins2Akita mouse does not seem to be robust.

Nonobese Diabetic Mouse

The genetic mouse model of type 1 diabetes that has been studied most thoroughly is the nonobese diabetic (NOD) mouse (94–96). These animals develop spontaneous autoimmune destruction of β cells at approximately 5 mo of age, although the precise age of onset of diabetes is somewhat variable. However, unlike STZ mice, insulin treatment is required to maintain NOD mice for any extended time after the onset of hyperglycemia, indicating more complete insulin deficiency. Moreover, unlike enhanced susceptibility in male mice for STZ diabetes, there is a female predominance in the NOD line with a female:male ratio of approximately 4:1. The characteristics of autoimmune disease contributing to β cell failure in NOD mice have been studied extensively, and the model has a number of similarities with features of human type 1 diabetes (97–99). These include (1) inheritance of specific MHC class II alleles and many non-MHC loci as polygenic susceptibility loci, (2) transmission of the disease by hematopoietic stem cells, (3) the development of an intraislet inflammatory infiltrate (insulinitis) with anti-islet cell antibodies, and (4) a strict dependence of disease on T cells.

Despite the intensive study of the immunopathogenesis of islet cell destruction in the NOD mouse, very little work has been done to study complications of diabetes in this model. In part, this is because of the late and somewhat variable age of onset of diabetes and the requirement for more intensive management, including daily administration of exogenous insulin. In addition, the genetics of the NOD model is complex. The line was initially developed from an outbred ICR mouse line (94–96). Moreover, there are six or seven background loci that must be retained for development of disease. Accordingly, identifying an appropriate nondiabetic control strain has been a problem. Nonobese nondiabetic (NON) mice were derived from nondiabetic progeny in the original crosses that generated the NOD line. However, NON mice contain a diabetes-resistant MHC haplotype (Kb, I-Anb1, I-Ek, Db) that is distinct from the NOD haplotype (Kd, I-Ag7, I-Enull, Db) associated with diabetes susceptibility (100). Although some studies have used the NON animals as controls for the NOD mouse, NON mice seem to develop spontaneous renal disease of uncertain cause (101). Thus, their utility as controls for studies of DN is questionable.

As discussed above, there have been relatively few studies of kidney disease in the NOD line. Nonetheless, those few studies indicate that albuminuria develops in hyperglycemic NOD animals. The levels of albuminuria, assessed by albumin-to-creatinine ratio, are roughly sevenfold higher than in NOD mice before development of hyperglycemia (102). Furthermore, studies of DN using this model have supported roles for TGF-β and advanced glycosylation end products (AGE) in the pathogenesis of mesangial proliferation and sclerosis (103–106). Because of such results along with the pathogenic similarities between NOD mice and humans with type 1 diabetes, this strain is likely to have utility for developing new models of diabetic complications. To circumvent the problems posed by its complicated genetics, AMDCC is working to generate embryonic stem cell lines from the NOD line to facilitate applications of transgenesis and gene targeting to our efforts at model development.

Mouse Models of Type 2 Diabetes

High-Fat Diet

A high-fat diet provides a commonly used approach to induce obesity and insulin resistance in C57BL6 mice (107–109) and is particularly useful for the study of accelerated atherosclerosis (104,110,111). The effect of a high-fat diet seems to depend on the strain of mouse studied, with A/J mice being relatively resistant to this effect (107). Although one study suggested that this syndrome was associated with the development of hypertension, a subsequent study failed to confirm this and further found no change in GFR (55,112). To our knowledge, the effects of high fat on albuminuria in these models has not been reported, although one study examined its effect on albuminuria in the KK-Ay mouse and found a dramatic effect (63).

LepRdb/LepRdb

The db/db mutation on the C57BLKS background has been investigated intensively and exhibits many features similar to human DN. Details regarding the identification and nomenclature regarding the db/db mouse may be found in a recent review (22). The diabetic gene (db) is transmitted as an autosomal recessive trait. The db gene encodes for a G-to-T point mutation of the leptin receptor, leading to abnormal splicing and defective signaling of the adipocyte-derived hormone leptin (113,114). This obese and diabetic mutant (db) was initially recognized in the C57BLKS/J strain and was subsequently also backcrossed to a pure C57BL/6J background. The C57BLKS/J mouse shares 84% of its alleles with the common C57BL/6 strain and 16% with the DBA/2J strain and was initially maintained by Dr. N. Kaliss (148).

In the C57BLKS/J db/db mouse, hyperinsulinemia is noted by 10 d of age, and blood glucose levels are slightly elevated at 1 mo of age (7.2 ± 2.3 mM) (115). The db/db mouse develops frank hyperglycemia with glucose values of 9.7 ± 1.6 mM by 8 wk of age and 15.7 ± 4.3 mM at 10 wk of age (115). Progressive hyperglycemia is noted with mean levels of glucose of 28.6 ± 13.2 mM at 16 wk of age (115,116). After 5 to 6 mo of age, the body weight and insulin levels begin to fall in association with pancreatic islet cell degeneration (117).

By 2 to 4 mo of age, diabetic mice have a 20 to 30% increase in glomerular size (118). Glomerular hypertrophy at the onset of diabetes may be due to alteration of glomerular hemodynamics as there is evidence of glomerular hyperfiltration in db/db mice during the early stages of diabetes (119). After 16 wk of age, there is a very consistent threefold increase in mesangial matrix expansion based on several independent studies (reviewed in reference 22). The range of albuminuria is between 68 and 600 μg/24 h in the db/db male mouse (22,118,120–122), whereas it is between 4 and 21 μg/24 h in the age-matched heterozygous littermate (118,122). The degree of albuminuria does not consistently increase with the duration of diabetes as there are similar levels of albuminuria between 8 and 25 wk (118,122–124). Arteriolar hyalinosis has been described in this model; however, there is virtually no evidence of advanced tubulointerstitial fibrosis.

The progression of diabetes in db/db mice on the C57BL/6 background in the diabetic phenotype is less severe than that in C57BLKS/J, and as these mice age, plasma glucose seems to normalize (22,125–129). More recently, some investigators have reported observing that a subset of approximately 50% of C57BL/6J db/db mice develop more persistent hyperglycemia (130,131). In these mice, more robust albuminuria and renal histopathologic diabetic changes have been reported (130,131). Unfortunately, the factors that cause this subgroup of C57BL/6J db/db mice to develop persistent hyperglycemia remain to be elucidated. Nevertheless, although the leptin mutation may not produce as robust a model for DN on the C57BL/6 background as it does on the C57BLKS/J background, it does provide a clear advantage for genetic studies because most transgenic and knockout strains are available on this background and can cleanly introgressed onto this strain.

Ob/ob

As opposed to the db/db mutants, the ob/ob recessive obese mouse carries a mutation in leptin, the ligand for the leptin receptor (132,133). The Lepob mutation exists on DBA2/J and C57BL/6J strains (132,134). Renal structure and function in C57BL/6J ob/ob mice is said to be relatively mild (21), but primary studies to support this statement are obscure.

Agouti Mutation

The agouti gene product encodes a 131–amino acid protein that contains a signal sequence. It is produced in the hair follicle, where it acts in a paracrine manner as a high-affinity antagonist of the melanocyte stimulating hormone (MSH) receptor on melanocytes to inhibit α-MSH–induced eumelanin production, causing their yellow coat color (135). Similarly, an agouti-related protein is a potent and selective antagonist of melanocortin receptors 3 and 4 (Mc3r and Mc4r), receptor subtypes, which are expressed in the hypothalamus and implicated in weight regulation (136). Mice that carry the dominant homozygous lethal mutations in agouti (Ay) or viable (Avy) exhibit a complex phenotype of obesity and insulin resistance in addition to yellow fur. The unifying molecular feature of all yellow obese Agouti mutations is that they confer ubiquitous and strong expression of the wild-type agouti protein through use of an alternative transcriptional promoter (137,138). Reports of albuminuria in diabetic KK-Ay mice suggest that this mutation may be useful for the study of nephropathy (see below and Figure 1) (139,140).

New Zealand Obese Mice

New Zealand obese (NZO) mice have a polygenically inherited form of obesity and type 2 diabetes (141–143). They may be prone to autoimmune disease as the kidneys exhibit light microscopic features of both diabetic and lupus nephropathies: Glomerular proliferation, mesangial deposits, mild basement membrane thickening, and glomerulosclerosis (144). Eosinophilic nodules may be seen in some glomeruli, with occasional hyalinization of the glomerular arterioles and healing arteriolar inflammation (144).

From the preceding discussion, it should be apparent that diverse models of both type 1 and type 2 diabetes are available in mice. Although renal changes have been reported in many of these models, renal failure has not as yet been identified as a consequence of diabetes in any of these models. Whether this is a result of inadequate length of diabetes before study or underlying resistance of specific strains to nephropathy remains to be addressed.

Inbred Mice: Strain Dependence of DN

Studies have identified certain strains (e.g., 129, ROP, NON, KK/HIJ) as more prone to glomerulosclerosis than other more commonly studied strains (e.g., C57BL/6, FVB/NJ) that seem relatively resistant to renal disease. It is noteworthy that studies of DN have been performed in fewer than 5% of the available mouse strains. The following section overviews the literature regarding the sensitivity of the kidney to diabetes in the major strains studied so far.

C57BL6

Although C57BL/6 is the most widely used inbred strain, studies of kidney disease in this strain suggest that it is relatively resistant to renal injury (145,146) (other than a possible subgroup, mentioned below, regarding a cohort of this strain that may be more susceptible to DN in the db/db model). The C57BL/6J strain provided the DNA source for the first high-quality draft sequence of the mouse genome. This strain is used in a wide variety of research areas, including cardiovascular biology, developmental biology, diabetes, and obesity. Overall, C57BL/6 mice breed well, are long lived, and have a low susceptibility to tumors. Other characteristics include a high susceptibility to diet-induced obesity, type 2 diabetes, and atherosclerosis. C57BL/6J mice that are fed a high-fat diet develop obesity, hyperglycemia, and hyperinsulinemia (55,112); however, this does not seem to have an impact on renal function (112). In contrast to their resistance to glomerulosclerosis, C57BL/6J mice provide a reasonably robust strain for studying of atherosclerosis when fed an atherogenic diet (111). In the experience of the AMDCC and others, the leptin receptor (db/db) mutation in this background often results in relatively mild and transient hyperglycemia and modest albuminuria (22,147) when compared with the presence of this mutation in the C57BLKS strain (see below).

C57BLKS (Formerly C57BL/KsJ)

The C57BLKS strain was originally derived from C57BL/6J mice maintained by Dr. N. Kaliss (148) and may be somewhat more susceptible to renal disease than the parental C57BL/6. Genomic analysis shows that 84% of the alleles in C57BLKS are shared with C57BL/6 and 16% are shared with DBA/2J, consistent with genetic contamination of the C57BL/6J progenitors early in the strain’s history (148). Studies indicate that on the C57BLKS background, type 2 diabetic db/db mice have lesions consistent with DN (thickened basement membrane, mesangial expansion), whereas they are resistant to DN on a C57Bl/6 background (22,149–151). Although these models have not been completely characterized, this difference suggests two possibilities: Either the worse severity of hyperglycemia that develops in C57BLKS causes this (111) (a combination of peripheral insulin resistance and insulin deficiency develops in C57BLKSLeprdb, whereas C57BL/6Leprdb have only peripheral insulin resistance), or C57BLKS expresses modifier genes that predispose to DN. These mice seem to be relatively prone to atherosclerosis (111) and also develop myocardial disease (152). In support of the former possibility is the general experience including that of the AMDCC that fasting blood glucose is sustained at much higher levels in db/db mice on this background than in db/db mice on a C57BL/6J background.

DBA/2J

In contrast to C57BL/6, the DBA/2J strain seems to be prone to diabetic albuminuria (153,154), but this remains to be rigorously tested. DBA/2J are relatively resistant to the development of atherosclerosis on a semisynthetic high-fat diet (155) and are hyporesponsive to diets that contain high levels of fat and cholesterol (156). Spontaneous calcified heart lesions progress with age, and dystrophic cardiac calcification may be related to disturbed calcium metabolism (157,158).

129/SvJ

Multiple substrains of 129 exist, and care must be taken to determine the precise genetic composition of the specific 129 substrain in use for appropriate interpretation of data. This strain of mice has been found to be more susceptible to elevated BP, nephrosclerosis (both glomerular and tubulointerstitial), and albuminuria in the setting of DOCA-salt hypertension than the C57BL/6 strain (159). In addition, 129/Sv mice develop more significant glomerulosclerosis, proteinuria, increases in BP, and apparent renal failure than do C57BL/6 mice after 5/6 nephrectomy (145). This susceptibility to renal injury and hypertension makes 129/Sv a potentially attractive strain for DN studies. No reports to date have compared renal or BP effects of diabetes in animals on this background with C57BL/6J mice or other strains. Unfortunately, the 129/J strains of mice with the db/db mutation are resistant to hyperglycemia (40).

ROP

The ROP strain of mouse originated from a heterogeneous stock being heterozygous for three mutant alleles: Ra/+ (ragged), Os/+ (osteosyndactylism), and Pt/+ (pintail). Studies of Os/+ ROP mice show that they possess approximately 50% fewer nephrons in the Os/+ mice than in the +/+ mice (160). In general, wild-type mice (Os+/+, Ra+/+, and Pt+/+) of ROP background seem to be more prone to glomerulosclerosis than C57BL/6 mice after a reduction in renal mass (161–163). Mice on the ROP background, in the absence of Os, Ra, or Pt mutant alleles, may also exhibit increased propensity for glomerulosclerosis after combined nephron mass reduction and diabetes (146).

FVB

FVB/NJ was derived from ou bred Swiss mice at NIH inbred for the fv1b allele, which confers sensitivity to the Friend leukemia virus B strain. Because of the prominent pronuclei in their fertilized eggs and the large litter size, FVB/NJ are commonly used for transgenic injection. In this context, FVB/N mice transgenic for a calmodulin minigene regulated by the rat insulin II promoter have been shown to develop type 1 diabetes (164,165). Initial studies characterizing the severity of DN in this line (designated OVE26) suggest that whereas they develop glomerular capillary basement membrane thickening, they do not develop significant albuminuria (68,166). The LepRdb mutation has also been backcrossed onto the FVB/NJ strain, and the kidneys from these mice exhibit mesangial sclerosis, although albuminuria and GFR have not been reported (134).

NON Mice

The NON strain is closely related to NOD mice and was developed as a “control” for NOD mice. The name was derived from “nonobese nondiabetic”; however, NON/LtJ mice are not “normal” as they exhibit tendencies toward autoimmune diseases. NON/LtJ mice harbor genes that predispose to type 2 diabetes, as evidenced by early, impaired glucose tolerance in males and females, and by the development of moderate maturity-onset obesity in the presence of low plasma insulin levels.

The renal phenotype in these mice is not well characterized but is reported to develop spontaneous glomerular lesions that resemble nodular glomerulosclerosis, although they are not overtly diabetic (101). The susceptibility of NON mice to DN using STZ or other models of diabetes has not been reported.

KK Mice

Although exhibiting only mild insulin resistance, the KK mouse seems to be predisposed to development of renal lesions very reminiscent of DN (167,168). These mice were originally established from inbreeding a Japanese mouse by Kondo (169). The severity of hyperglycemia and insulin resistance is exacerbated by introduction of the agouti (Ay) allele into the KK background (170). Of note is that both KK and KK-Ay mice develop nodular glomerulosclerosis and mesangial proliferation (171), which are improved by glycemic control (167,172,173). Albuminuria progressively increases with age; however, reports of renal insufficiency or failure have not been forthcoming.

Monogenic Mutations and Transgenic Mice

Despite the preceding caveats regarding the difficulties in phenotyping renal function in mice, the use of transgenic mouse models has provided a better understanding of the factors that exacerbate DN. The following review is not meant to be comprehensive but only to provide an overview of some of the more recent developments in the field. The reader is referred to other recent articles for additional perspectives on this topic (174).

Apolipoprotein E

Although control of hyperglycemia can decrease the development of DN, there is evidence suggesting that a genetic susceptibility to development of DN is related to abnormalities in lipid metabolism. Hypertriglyceridemia is among the main phenotypic features that distinguish people whoa re prone to nephropathy (175).

Apolipoprotein E (ApoE) is a protein of 34 kD that circulates on plasma lipoproteins at a concentration of 3 to 5 mg/dl (176,177). In the human population, the ApoE gene, located on chromosome 19q13.2, has three common alleles—2, 3, and 4—coding for the three main isoforms of the ApoE protein: E2, E3, and E4 (178). ApoE isoforms differ in their ability to bind to LDL receptors, with E4 having the greatest binding capacity and E2 having defective binding and decreased triglyceride clearance (179). Many but not all studies in patients with either type 1 or type 2 diabetes have implicated the presence of the E2 allele as a risk factor for development and/or progression of DN (180–190). The underlying mechanism(s) that might be involved in the development of DN in patients with the ApoE2 polymorphism have not yet been determined. It is possible that the associated hypertriglyceridemia may predispose to renal injury. It has also been suggested that ApoE may play a role in tissue growth and/or repair after injury separate from its effects on maintenance of lipoprotein homeostasis (191).

In mice with targeted disruption of the ApoE gene locus, no tissues or cells synthesize or secrete ApoE, so ApoE is absent from the plasma. As a consequence, the clearance of remnant lipoproteins is blocked, and serum cholesterol and triglycerides increase to levels of approximately 450 and 250 mg/dl, respectively, similar to levels in human type III hyperlipoproteinemia as a result of ApoE deficiency. ApoE deficiency causes increased susceptibility to atherosclerosis and represents a murine model of spontaneous (non–diet-induced) atherosclerosis (192). Of note, fasting glucose and insulin levels are not elevated in ApoE−/− mice on a normal diet compared with C57BL/6J mice. It is interesting that a recent study in low-dose STZ–treated C57BL6 mice showed that deletion of the ApoE allele was associated with more severe nephropathy as assessed by histopathology and albuminuria (which increased approximately fivefold, albeit still <100 μg/24 h) (193).

Endothelial Nitric Oxide Synthase

Endothelial dysfunction is present in diabetes and is associated with impaired vascular nitric oxide (NO) synthesis. A number of polymorphisms in the endothelial NO synthase gene (eNOS), located on human chromosome 7, have been linked to vasculopathies (194–198). Recent studies have reported an association between eNOS polymorphisms that lead to decreased eNOS expression and development of advanced nephropathy in patients with type 1 (199–201) and type 2 diabetes (202,203). Other studies have found an association of these polymorphisms with ESRD (204,205). However, not all studies have detected any potential association of these eNOS polymorphisms with DN (206–210).

Although direct examination of development of DN has not yet been undertaken in mice with gene deletion of eNOS, it is noteworthy that studies of the phenotype of mice with combined eNOS and ApoE deficiency indicate accelerated atherosclerosis, hypertension, and progressive renal dysfunction characterized by smaller kidney weights and glomerular lipid deposits (211–214)

AGE

Generation of AGE, nonenzymatically glycosylated protein derivatives resulting from prolonged hyperglycemic exposure, is a cardinal feature of the diabetic milieu, and early and advanced steps of the complex process of reactions that lead to glycation of proteins are now well understood (215). Human diabetic kidney (and other tissues) are characterized by glomerular accumulation of AGE, in particular carboxymethyl lysine–modified adducts of proteins (216). AGE may perturb cell function through receptor-independent mechanisms and through receptor-mediated signaling pathways. Several cell surface receptors/binding proteins have been shown to bind AGE, including the macrophage scavenger receptor type II, OST-48, 80K-H, galectin-3, CD36, and receptor for AGE (RAGE) (reviewed in reference 216). Whereas some of these interactions may function to remove/scavenge AGE, AGE binding to RAGE, a signal transduction receptor of the Ig superfamily, mediates diverse cellular responses. For example, Oldfield et al. (217) demonstrated that AGE mediate tubular epithelial to myofibroblast transdifferentiation through RAGE in vitro and that this effect is dependent on the prosclerotic cytokine TGF-β. Thus, an AGE-dependent pathway may play a role in the development of tubulointerstitial fibrosis in the diabetic kidney. RAGE may activate diverse signaling mediators in endothelial cells and macrophages, including p21ras, ERK1/2 MAP kinases, Nf-κB, and NADPH oxidase species (216,218). However, in the diabetic kidney in humans and mice, RAGE is expressed principally by glomerular visceral epithelial cells (podocytes) (219,220). It is interesting that neutralizing soluble RAGE ameliorated glomerular damage in the hyperglycemic db/db mouse (220).

Recently, Yamamoto et al. (221) reported an interesting new transgenic model that provides further evidence of a role of RAGE in acceleration of diabetes-induced glomerular lesions. In this model, the human RAGE gene is overexpressed specifically in endothelial cells under control of an endothelial cell–specific mouse flk-1 promoter (flk1-RAGE) on an inbred CD-1 genetic background. Persistent hyperglycemia was induced in Flk1-RAGE mice by interbreeding with diabetic insulin-eNOS transgenic mice, and flk1-RAGE/ins-eNOS double-transgenic mice exhibited increased HgbA1c and serum AGE levels similar to ins-eNOS diabetic controls. Kidney enlargement and glomerular lesions, including albuminuria, mesangial expansion, glomerular hypertrophy, and glomerulosclerosis, were detectable at younger age (4 mo) in flk1-RAGE/ins-eNOS mice, compared with diabetic ins-eNOS mice (6 mo). Thus, aberrant endothelial expression of RAGE accelerated a glomerular phenotype in diabetic mice that mimics features of diabetic glomerulopathy in humans, including albuminuria, mesangial expansion, and glomerular hypertrophy. However, cardinal histopathologic features of progressive DN in humans, including arteriolar hyalinosis and tubulointerstitial fibrosis, and progressive renal insufficiency (see the Working Definition of Diabetic Nephropathy section), have not been described in this model to date. In collaboration with H. Yamamoto, in-depth phenotype examination and validation using AMDCC standards is under way.

GLUT1

A role for the GLUT1 glucose transporter in the pathogenesis of DN has been suggested over the past decade (222,223). This transporter has been identified as a major glucose transporter of the glomerulus (224), and high glucose exposure or diabetes increases its expression in cultured mesangial cells and whole glomeruli (225,226). Presumably, increased plasma membrane expression of GLUT1 in susceptible cells in the kidney would lead to increased glucose metabolic flux and glucotoxicity perhaps as a result of enhanced generation of reactive oxygen species. In cultured mesangial cells, GLUT1 overexpression leads to enhanced protein kinase C activation and fibronectin accumulation, whereas suppression of GLUT1 levels reduces protein kinase C activation and fibronectin accumulation (225,226). Similar results in transgenic mice have been presented in preliminary form. Normoglycemic GLUT1 transgenic mice develop increased mesangial expansion and albuminuria, whereas mice with an antisense GLUT1 transgene that reduces glomerular GLUT1 expression are protected from these changes despite the presence of hyperglycemia (225,227). Thus, differences in GLUT1 expression could explain part of the variable susceptibility to DN in different strains of mice. Such a possibility is now being explored.

Conclusions

Mice provide an experimental model of unparalleled flexibility for studying mammalian diseases, including DN. Inbred strains of mice exhibit substantial differences in the renal effects of diabetes. Much remains to be established regarding the course of DN in this species and defining those strains and/or mutants that are most susceptible to renal injury from diabetes. It will be especially important to determine whether renal function declines in any of these mouse models of DN. Technical refinements in the measurement of plasma creatinine, GFR, and uniform reporting of albuminuria and histopathology should facilitate progress in this field and allow better comparisons of results obtained by different laboratories. Together with the unique genetic reagents available in mice, the identification of a mouse model of DN that closely mirrors human disease will significantly enhance our understanding of DN and accelerate our progress toward a treatment for this disease.

Acknowledgments

We acknowledge support of the Juvenile Diabetes Research Foundation (JDRF); the National Institute of Diabetes and Digestive and Kidney Diseases; and the National Heart, Lung, and Blood Institute for funding of U01DK61018 (to M.D.B.), NIH UO1-DK60994 (to F.C.B.), U01 DK060995 (to E.P.B.), and U01 HL070523 (to T.M.C.) and bioinformatics and web site creation and maintenance (www.amdcc.org) from Rick McIndoe at Medical College of Georgia (U01 DK060966).

  • © 2005 American Society of Nephrology

References

  1. 1.↵
    USRDS: The United States Renal Data System. Am J Kidney Dis 42[Suppl]: 1–230, 2003
    OpenUrl
  2. 2.↵
    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 320: 1161–1165, 1989
    OpenUrlCrossRefPubMed
  3. 3.
    Krolewski AS: Genetics of diabetic nephropathy: Evidence for major and minor gene effects. Kidney Int 55: 1582–1596, 1999
    OpenUrlCrossRefPubMed
  4. 4.↵
    Iyengar SK, Fox KA, Schachere M, Manzoor F, Slaughter ME, Covic AM, Orloff SM, Hayden PS, Olson JM, Schelling JR, Sedroe JR: Linkage analysis of candidate loci for end-stage renal disease due to diabetic nephropathy. J Am Soc Nephrol 14[Suppl 2]: S195–S201, 2003
    OpenUrl
  5. 5.↵
    Paigen B, Mitchell D, Holmes PA, Albee D: Genetic analysis of strains C57BL/6J and BALB/cJ for Ath-1, a gene determining atherosclerosis susceptibility in mice. Biochem Genet 25: 881–892, 1987
    OpenUrlCrossRefPubMed
  6. 6.
    DiPetrillo K, Tsaih SW, Sheehan S, Johns C, Kelmenson P, Gavras H, Churchill GA, Paigen B: Genetic analysis of blood pressure in C3H/HeJ and SWR/J mice. Physiol Genomics 17: 215–220, 2004
    OpenUrlCrossRefPubMed
  7. 7.↵
    Sugiyama F, Yagami K, Paigen B: Mouse models of blood pressure regulation and hypertension. Curr Hypertens Rep 3: 41–48, 2001
    OpenUrlCrossRefPubMed
  8. 8.↵
    Marshall JD, Mu JL, Cheah YC, Nesbitt MN, Frankel WN, Paigen B: The AXB and BXA set of recombinant inbred mouse strains. Mamm Genome 3: 669–680, 1992
    OpenUrlCrossRefPubMed
  9. 9.↵
    Beck JA, Lloyd S, Hafezparast M, Lennon-Pierce M, Eppig JT, Festing MF, Fisher EM: Genealogies of mouse inbred strains. Nat Genet 24: 23–25, 2000
    OpenUrlCrossRefPubMed
  10. 10.↵
    Bronson SK, Smithies O: Altering mice by homologous recombination using embryonic stem cells. J Biol Chem 269: 27155–27158, 1994
    OpenUrlFREE Full Text
  11. 11.
    Babinet C: Transgenic mice: An irreplaceable tool for the study of mammalian development and biology. J Am Soc Nephrol 11[Suppl 16]: S88–S94, 2000
    OpenUrl
  12. 12.↵
    Mansouri A: Determination of gene function by homologous recombination using embryonic stem cells and knockout mice. Methods Mol Biol 175: 397–413, 2001
    OpenUrlPubMed
  13. 13.↵
    Townley DJ, Avery BJ, Rosen B, Skarnes WC: Rapid sequence analysis of gene trap integrations to generate a resource of insertional mutations in mice. Genome Res 7: 293–298, 1997
    OpenUrlAbstract/FREE Full Text
  14. 14.
    Leighton PA, Mitchell KJ, Goodrich LV, Lu X, Pinson K, Scherz P, Skarnes WC, Tessier-Lavigne M: Defining brain wiring patterns and mechanisms through gene trapping in mice. Nature 410: 174–179, 2001
    OpenUrlCrossRefPubMed
  15. 15.↵
    Hrabe de Angelis MH, Flaswinkel H, Fuchs H, Rathkolb B, Soewarto D, Marschall S, Heffner S, Pargent W, Wuensch K, Jung M, Reis A, Richter T, Alessandrini F, Jakob T, Fuchs E, Kolb H, Kremmer E, Schaeble K, Rollinski B, Roscher A, Peters C, Meitinger T, Strom T, Steckler T, Holsboer F, Klopstock T, Gekeler F, Schindewolf C, Jung T, Avraham K, Behrendt H, Ring J, Zimmer A, Schughart K, Pfeffer K, Wolf E, Balling R: Genome-wide, large-scale production of mutant mice by ENU mutagenesis. Nat Genet 25: 444–447, 2000
    OpenUrlCrossRefPubMed
  16. 16.↵
    Young BA, Maynard C, Boyko EJ: Racial differences in diabetic nephropathy, cardiovascular disease, and mortality in a national population of veterans. Diabetes Care 26: 2392–2399, 2003
    OpenUrlAbstract/FREE Full Text
  17. 17.↵
    Tarnow L, Rossing P, Nielsen FS, Fagerudd JA, Poirier O, Parving HH: Cardiovascular morbidity and early mortality cluster in parents of type 1 diabetic patients with diabetic nephropathy. Diabetes Care 23: 30–33, 2000
    OpenUrlAbstract
  18. 18.↵
    Mogensen CE: The effect of blood pressure intervention on renal function in insulin-dependent diabetes. Diabetes Metab 15: 343–351, 1989
    OpenUrl
  19. 19.↵
    Caramori ML, Fioretto P, Mauer M: The need for early predictors of diabetic nephropathy risk: Is albumin excretion rate sufficient? Diabetes 49: 1399–1408, 2000
    OpenUrlAbstract
  20. 20.↵
    Mogensen CE: Microalbuminuria and hypertension with focus on type 1 and type 2 diabetes. J Intern Med 254: 45–66, 2003
    OpenUrlCrossRefPubMed
  21. 21.↵
    Velasquez MT, Kimmel PL, Michaelis OE: Animal models of spontaneous diabetic kidney disease. FASEB J 4: 2850–2859, 1990
    OpenUrlPubMed
  22. 22.↵
    Sharma K, McCue P, Dunn SR: Diabetic kidney disease in the db/db mouse. Am J Physiol Renal Physiol 284: F1138–F1144, 2003
    OpenUrlCrossRefPubMed
  23. 23.↵
    Mauer SM, Lane P, Zhu D, Fioretto P, Steffes MW: Renal structure and function in insulin-dependent diabetes mellitus in man. J Hypertens Suppl 10: S17–S20, 1992
    OpenUrlCrossRefPubMed
  24. 24.↵
    Chavers BM, Bilous RW, Ellis EN, Steffes MW, Mauer SM: Glomerular lesions and urinary albumin excretion in type I diabetes without overt proteinuria. N Engl J Med 320: 966–970, 1989
    OpenUrlCrossRefPubMed
  25. 25.↵
    Mauer SM, Steffes MW, Ellis EN, Sutherland DE, Brown DM, Goetz FC: Structural-functional relationships in diabetic nephropathy. J Clin Invest 74: 1143–1155, 1984
    OpenUrlCrossRefPubMed
  26. 26.↵
    Rudberg S, Osterby R: Decreasing glomerular filtration rate—An indicator of more advanced diabetic glomerulopathy in the early course of microalbuminuria in IDDM adolescents? Nephrol Dial Transplant 12: 1149–1154, 1997
    OpenUrlCrossRefPubMed
  27. 27.↵
    Hasslacher C, Ritz E, Terpstra J, Gallasch G, Kunowski G, Rall C: Natural history of nephropathy in type I diabetes. Relationship to metabolic control and blood pressure. Hypertension 7: II74–II78, 1985
    OpenUrlPubMed
  28. 28.↵
    Jacobsen P, Rossing K, Tarnow L, Rossing P, Mallet C, Poirier O, Cambien F, Parving HH: Progression of diabetic nephropathy in normotensive type 1 diabetic patients. Kidney Int Suppl 71: S101–S105, 1999
    OpenUrlCrossRefPubMed
  29. 29.↵
    Krolewski AS, Laffel LM, Krolewski M, Quinn M, Warram JH: Glycosylated hemoglobin and the risk of microalbuminuria in patients with insulin-dependent diabetes mellitus. N Engl J Med 332: 1251–1255, 1995
    OpenUrlCrossRefPubMed
  30. 30.↵
    DCCT. 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 329: 977–986, 1993
    OpenUrlCrossRefPubMed
  31. 31.↵
    Peterson KP, Pavlovich JG, Goldstein D, Little R, England J, Peterson CM: What is hemoglobin A1c? An analysis of glycated hemoglobins by electrospray ionization mass spectrometry. Clin Chem 44: 1951–1958, 1998
    OpenUrlAbstract/FREE Full Text
  32. 32.↵
    McDonald MJ, Shapiro R, Bleichman M, Solway J, Bunn HF: Glycosylated minor components of human adult hemoglobin. Purification, identification, and partial structural analysis. J Biol Chem 253: 2327–2332, 1978
    OpenUrlAbstract/FREE Full Text
  33. 33.↵
    Kilpatrick ES: Glycated haemoglobin in the year 2000. J Clin Pathol 53: 335–339, 2000
    OpenUrlFREE Full Text
  34. 34.↵
    Fujii E, Nomoto T: Changes in glycosylated hemoglobin in short- and semi long-term streptozotocin-diabetic mice and rats. Jpn J Pharmacol 34: 113–115, 1984
    OpenUrlPubMed
  35. 35.↵
    Dan K, Fujita H, Seto Y, Kato R: Relation between stable glycated hemoglobin A1C and plasma glucose levels in diabetes-model mice. Exp Anim 46: 135–140, 1997
    OpenUrlCrossRefPubMed
  36. 36.↵
    DCCT. Diabetes Control and Complications Trial (DCCT): Results of feasibility study. The DCCT Research Group. Diabetes Care 10: 1–19, 1987
    OpenUrlAbstract/FREE Full Text
  37. 37.↵
    Kilpatrick ES, Maylor PW, Keevil BG: Biological variation of glycated hemoglobin. Implications for diabetes screening and monitoring. Diabetes Care 21: 261–264, 1998
    OpenUrlAbstract/FREE Full Text
  38. 38.↵
    Jiao Y, Okumiya T, Saibara T, Park K, Sasaki M: Abnormally decreased HbA1c can be assessed with erythrocyte creatine in patients with a shortened erythrocyte age. Diabetes Care 21: 1732–1735, 1998
    OpenUrlAbstract/FREE Full Text
  39. 39.↵
    McCarter RJ, Hempe JM, Gomez R, Chalew SA: Biological variation in HbA1c predicts risk of retinopathy and nephropathy in type 1 diabetes. Diabetes Care 27: 1259–1264, 2004
    OpenUrlAbstract/FREE Full Text
  40. 40.↵
    Kaku K, Fiedorek FT Jr, Province M, Permutt MA: Genetic analysis of glucose tolerance in inbred mouse strains. Evidence for polygenic control. Diabetes 37: 707–713, 1988
    OpenUrlAbstract/FREE Full Text
  41. 41.↵
    Dubuc PU, Scott BK, Peterson CM: Sex differences in glycated hemoglobin in diabetic and non-diabetic C57BL/6 mice. Diabetes Res Clin Pract 21: 95–101, 1993
    OpenUrlCrossRefPubMed
  42. 42.↵
    Meyer MH, Meyer RA Jr, Gray RW, Irwin RL: Picric acid methods greatly overestimate serum creatinine in mice: More accurate results with high-performance liquid chromatography. Anal Biochem 144: 285–290, 1985
    OpenUrlCrossRefPubMed
  43. 43.↵
    O’Donnell MP, Burne M, Daniels F, Rabb H: Utility and limitations of serum creatinine as a measure of renal function in experimental renal ischemia-reperfusion injury. Transplantation 73: 1841–1844, 2002
    OpenUrlCrossRefPubMed
  44. 44.↵
    Jung K, Wesslau C, Priem F, Schreiber G, Zubek A: Specific creatinine determination in laboratory animals using the new enzymatic test kit “Creatinine-PAP.” J Clin Chem Clin Biochem 25: 357–361, 1987
    OpenUrlPubMed
  45. 45.↵
    Kemperman FA, Silberbusch J, Slaats EH, van Zanten AP, Weber JA, Krediet RT, Arisz L: Glomerular filtration rate estimation from plasma creatinine after inhibition of tubular secretion: Relevance of the creatinine assay. Nephrol Dial Transplant 14: 1247–1251, 1999
    OpenUrlCrossRefPubMed
  46. 46.↵
    Kemperman FA, Weber JA, Gorgels J, van Zanten AP, Krediet RT, Arisz L: The influence of ketoacids on plasma creatinine assays in diabetic ketoacidosis. J Intern Med 248: 511–517, 2000
    OpenUrlCrossRefPubMed
  47. 47.↵
    Gerard SK, Khayam-Bashi H: Characterization of creatinine error in ketotic patients. A prospective comparison of alkaline picrate methods with an enzymatic method. Am J Clin Pathol 84: 659–664, 1985
    OpenUrlPubMed
  48. 48.↵
    Yuen PS, Dunn SR, Miyaji T, Yasuda H, Sharma K, Star RA: A simplified method for HPLC determination of creatinine in mouse serum. Am J Physiol Renal Physiol 286: F1116–F1119, 2004
    OpenUrlCrossRefPubMed
  49. 49.↵
    Dunn SR, Qi Z, Bottinger EP, Breyer MD, Sharma K: Utility of endogenous creatinine clearance as a measure of renal function in mice. Kidney Int 65: 1959–1967, 2004
    OpenUrlCrossRefPubMed
  50. 50.↵
    Qi Z, Whitt I, Mehta A, Jin J, Zhao M, Harris RC, Fogo AB, Breyer MD: Serial determination of glomerular filtration rate in conscious mice using FITC-inulin clearance. Am J PhysiolRenal Physiol 286: F590–F596, 2004
    OpenUrlCrossRefPubMed
  51. 51.↵
    Lorenz JN, Gruenstein E: A simple, nonradioactive method for evaluating single-nephron filtration rate using FITC-inulin. Am J Physiol (Lond) 276: F172–F177, 1999
    OpenUrl
  52. 52.↵
    Hackbarth H, Lunebrink J: [Determination of the mouse glomerular filtration rate by total 51Cr- EDTA plasma slope clearance depending on age and sex]. In German. Z Versuchstierkd 21: 38–46, 1979
    OpenUrlPubMed
  53. 53.↵
    Hackbarth H, Hackbarth D: Genetic analysis of renal function in mice. 1. Glomerular filtration rate and its correlation with body and kidney weight. Lab Anim 15: 267–272, 1981
    OpenUrlCrossRefPubMed
  54. 54.↵
    Krege JH, John SW, Langenbach LL, Hodgin JB, Hagaman JR, Bachman ES, Jennette JC, O’Brien DA, Smithies O: Male-female differences in fertility and blood pressure in ACE-deficient mice. Nature 375: 146–148, 1995
    OpenUrlCrossRefPubMed
  55. 54a.↵
    Gurley SB, Snow KP, Meyer TW, Coffmann TM: Influence of genetic background on diabetic kidney disease in mice. J Am Soc Nephrol 15: 727A, 2004
    OpenUrl
  56. 55.↵
    Mills E, Kuhn CM, Feinglos MN, Surwit R: Hypertension in CB57BL/6J mouse model of non-insulin-dependent diabetes mellitus. Am J Physiol 264: R73–R78, 1993
    OpenUrlPubMed
  57. 56.↵
    Huang W, Gallois Y, Bouby N, Bruneval P, Heudes D, Belair MF, Krege JH, Meneton P, Marre M, Smithies O, Alhenc-Gelas F: Genetically increased angiotensin I-converting enzyme level and renal complications in the diabetic mouse. Proc Natl Acad Sci U S A 98: 13330–13334, 2001
    OpenUrlAbstract/FREE Full Text
  58. 57.↵
    Kakoki M, Takahashi N, Jennette JC, Smithies O: Diabetic nephropathy is markedly enhanced in mice lacking the bradykinin B2 receptor. Proc Natl Acad Sci U S A 101: 13302–13305, 2004
    OpenUrlAbstract/FREE Full Text
  59. 58.↵
    Butz GM, Davisson RL: Long-term telemetric measurement of cardiovascular parameters in awake mice: A physiological genomics tool. Physiol Genomics 5: 89–97, 2001
    OpenUrlCrossRefPubMed
  60. 59.↵
    Wajchenberg BL, Sabbaga E, Fonseca JA: The natural history of diabetic nephropathy in type I diabetes and the role of metabolic control in its prevention, reversibility and clinical course. Acta Diabetol Lat 20: 1–18, 1983
    OpenUrlCrossRefPubMed
  61. 60.↵
    Molitch ME, DeFronzo RA, Franz MJ, Keane WF, Mogensen CE, Parving HH, Steffes MW: Nephropathy in diabetes. Diabetes Care 27[Suppl 1]: S79–S83, 2004
    OpenUrl
  62. 61.↵
    Finlayson JS, Asofsky R, Potter M, Runner CC: Major urinary protein complex of normal mice: Origin. Science 149: 981–982, 1965
    OpenUrlAbstract/FREE Full Text
  63. 62.↵
    Shahan K, Denaro M, Gilmartin M, Shi Y, Derman E: Expression of six mouse major urinary protein genes in the mammary, parotid, sublingual, submaxillary, and lachrymal glands and in the liver. Mol Cell Biol 7: 1947–1954, 1987
    OpenUrlAbstract/FREE Full Text
  64. 63.↵
    Yu PH, Wang M, Deng YL, Fan H, Shira-Bock L: Involvement of semicarbazide-sensitive amine oxidase-mediated deamination in atherogenesis in KKAy diabetic mice fed with high cholesterol diet. Diabetologia 45: 1255–1262, 2002
    OpenUrl
  65. 64.↵
    Fioretto P, Steffes MW, Mauer M: Glomerular structure in nonproteinuric IDDM patients with various levels of albuminuria. Diabetes 43: 1358–1364, 1994
    OpenUrlAbstract/FREE Full Text
  66. 65.↵
    Hong SW, Isono M, Chen S, Iglesias-De La Cruz MC, Han DC, Ziyadeh FN: Increased glomerular and tubular expression of transforming growth factor-β1, its type II receptor, and activation of the Smad signaling pathway in the db/db mouse. Am J Pathol 158: 1653–1663, 2001
    OpenUrlCrossRefPubMed
  67. 66.↵
    Fujimoto M, Maezawa Y, Yokote K, Joh K, Kobayashi K, Kawamura H, Nishimura M, Roberts AB, Saito Y, Mori S: Mice lacking Smad3 are protected against streptozotocin-induced diabetic glomerulopathy. Biochem Biophys Res Commun 305: 1002–1007, 2003
    OpenUrlCrossRefPubMed
  68. 67.
    Hammad SM, Hazen-Martin DJ, Sohn M, Eldridge L, Powell-Braxton L, Won W, Lyons TJ: Nephropathy in a hypercholesterolemic mouse model with streptozotocin-induced diabetes. Kidney Blood Press Res 26: 351–361, 2003
    OpenUrlCrossRefPubMed
  69. 68.↵
    Carlson EC, Audette JL, Klevay LM, Nguyen H, Epstein PN: Ultrastructural and functional analyses of nephropathy in calmodulin-induced diabetic transgenic mice. Anat Rec 247: 9–19, 1997
    OpenUrlCrossRefPubMed
  70. 69.↵
    Lewis C, Barbiers AR: Streptozotocin, a new antibiotic. In vitro and in vivo evaluation. Antibiot Annu 7: 247–254, 1959
    OpenUrlPubMed
  71. 70.↵
    Wilson GL, Leiter EH: Streptozotocin interactions with pancreatic beta cells and the induction of insulin-dependent diabetes. Curr Top Microbiol Immunol 156: 27–54, 1990
    OpenUrlPubMed
  72. 71.↵
    Leiter EH: Multiple low-dose streptozotocin-induced hyperglycemia and insulitis in C57BL mice: Influence of inbred background, sex, and thymus. Proc Natl Acad Sci U S A 79: 630–634, 1982
    OpenUrlAbstract/FREE Full Text
  73. 72.↵
    Hosokawa M, Dolci W, Thorens B: Differential sensitivity of GLUT1- and GLUT2-expressing beta cells to streptozotocin. Biochem Biophys Res Commun 289: 1114–1117, 2001
    OpenUrlCrossRefPubMed
  74. 73.↵
    Itagaki S, Nishida E, Lee MJ, Doi K: Histopathology of subacute renal lesions in mice induced by streptozotocin. Exp Toxicol Pathol 47: 485–491, 1995
    OpenUrlPubMed
  75. 74.
    Farr AG, Mannschreck JW, Anderson SK: Expression of Ia antigens by murine kidney epithelium after exposure to streptozotocin. Am J Pathol 126: 561–568, 1987
    OpenUrlPubMed
  76. 75.↵
    Schmezer P, Eckert C, Liegibel UM: Tissue-specific induction of mutations by streptozotocin in vivo. Mutat Res 307: 495–499, 1994
    OpenUrlPubMed
  77. 76.↵
    Katoh M, Ohmachi Y, Kurosawa Y, Yoneda H, Tanaka N, Narita H: Effects of imidapril and captopril on streptozotocin-induced diabetic nephropathy in mice. Eur J Pharmacol 398: 381–387, 2000
    OpenUrlCrossRefPubMed
  78. 77.↵
    Awazu M, Omori S, Ishikura K, Hida M, Fujita H: The lack of cyclin kinase inhibitor p27Kip1 ameliorates progression of diabetic nephropathy. J Am Soc Nephrol 14: 699–708, 2003
    OpenUrlAbstract/FREE Full Text
  79. 78.↵
    Lubec B, Hermon M, Hoeger H, Lubec G: Aromatic hydroxylation in animal models of diabetes mellitus. FASEB J 12: 1581–1587, 1998
    OpenUrlPubMed
  80. 79.↵
    Like AA, Appel MC, Williams RM, Rossini AA: Streptozotocin-induced pancreatic insulitis in mice. Morphologic and physiologic studies. Lab Invest 38: 470–486, 1978
    OpenUrlPubMed
  81. 80.↵
    Like AA, Rossini AA: Streptozotocin-induced pancreatic insulitis: New model of diabetes mellitus. Science 193: 415–417, 1976
    OpenUrlAbstract/FREE Full Text
  82. 81.↵
    Leiter EH: Differential susceptibility of BALB/c sublines to diabetes induction by multi-dose streptozotocin treatment. Curr Top Microbiol Immunol 122: 78–85, 1985
    OpenUrlPubMed
  83. 82.↵
    Rossini AA, Appel MC, Williams RM, Like AA: Genetic influence of the streptozotocin-induced insulitis and hyperglycemia. Diabetes 26: 916–920, 1977
    OpenUrlAbstract/FREE Full Text
  84. 83.↵
    Susztak K, Bottinger E, Novetsky A, Liang D, Zhu Y, Ciccone E, Wu D, Dunn S, McCue P, Sharma K: Molecular profiling of diabetic mouse kidney reveals novel genes linked to glomerular disease. Diabetes 53: 784–794, 2004
    OpenUrlAbstract/FREE Full Text
  85. 84.↵
    Mathews CE, Langley SH, Leiter EH: New mouse model to study islet transplantation in insulin-dependent diabetes mellitus. Transplantation 73: 1333–1336, 2002
    OpenUrlCrossRefPubMed
  86. 85.
    Wang J, Takeuchi T, Tanaka S, Kubo SK, Kayo T, Lu D, Takata K, Koizumi A, Izumi T: A mutation in the insulin 2 gene induces diabetes with severe pancreatic beta-cell dysfunction in the Mody mouse. J Clin Invest 103: 27–37, 1999
    OpenUrlCrossRefPubMed
  87. 86.↵
    Yoshioka M, Kayo T, Ikeda T, Koizumi A: A novel locus, Mody4, distal to D7Mit189 on chromosome 7 determines early-onset NIDDM in nonobese C57BL/6 (Akita) mutant mice. Diabetes 46: 887–894, 1997
    OpenUrlAbstract/FREE Full Text
  88. 87.↵
    Ron D: Proteotoxicity in the endoplasmic reticulum: Lessons from the Akita diabetic mouse. J Clin Invest 109: 443–445, 2002
    OpenUrlCrossRefPubMed
  89. 88.↵
    Haseyama T, Fujita T, Hirasawa F, Tsukada M, Wakui H, Komatsuda A, Ohtani H, Miura AB, Imai H, Koizumi A: Complications of IgA nephropathy in a non-insulin-dependent diabetes model, the Akita mouse. Tohoku J Exp Med 198: 233–244, 2002
    OpenUrlCrossRefPubMed
  90. 89.↵
    Fujita H, Haseyama T, Kayo T, Nozaki J, Wada Y, Ito S, Koizumi A: Increased expression of glutathione S-transferase in renal proximal tubules in the early stages of diabetes: A study of type-2 diabetes in the Akita mouse model. Exp Nephrol 9: 380–386, 2001
    OpenUrlCrossRefPubMed
  91. 90.↵
    Kanauchi M, Kawano T, Dohi K: Serum IgA levels in patients with diabetic nephropathy and IgA nephropathy superimposed on diabetes mellitus. Diabetes Res Clin Pract 48: 113–118, 2000
    OpenUrlCrossRefPubMed
  92. 91.↵
    Miller LL, Izzo MJ, Wemett D, Panner BJ, Schenk EA: Increased plasma IgA, sIgA, and C3- and IgA-containing immune complexes with renal glomerular deposits in diabetic rats. Diabetes 37: 185–193, 1988
    OpenUrlAbstract/FREE Full Text
  93. 92.
    Ainsworth SK, Hirsch HZ, Brackett NC Jr, Brissie RM, Williams AV Jr, Hennigar GR: Diabetic glomerulonephropathy: Histopathologic, immunofluorescent, and ultrastructural studies of 16 cases. Hum Pathol 13: 470–478, 1982
    OpenUrlCrossRefPubMed
  94. 93.↵
    Lee EY, Chung CH, Choi SO: Immunoglobulin A nephropathy in patients with non-insulin dependent diabetes mellitus. J Korean Med Sci 14: 582–585, 1999
    OpenUrlPubMed
  95. 94.↵
    Kolb H: Mouse models of insulin dependent diabetes: Low-dose streptozocin-induced diabetes and nonobese diabetic (NOD) mice. Diabetes Metab Rev 3: 751–778, 1987
    OpenUrlCrossRefPubMed
  96. 95.
    Makino S, Kunimoto K, Muraoka Y, Mizushima Y, Katagiri K, Tochino Y: Breeding of a non-obese, diabetic strain of mice. Jikken Dobutsu 29: 1–13, 1980
    OpenUrlCrossRefPubMed
  97. 96.↵
    Miyagawa J, Hanafusa T, Miyazaki A, Yamada K, Fujino-Kurihara H, Nakajima H, Kono N, Nonaka K, Tochino Y, Tarui S: Ultrastructural and immunocytochemical aspects of lymphocytic submandibulitis in the non-obese diabetic (NOD) mouse. Virchows Arch B Cell Pathol Incl Mol Pathol 51: 215–225, 1986
    OpenUrlPubMed
  98. 97.↵
    Cameron MJ, Meagher C, Delovitch TL: Failure in immune regulation begets IDDM in NOD mice. Diabetes Metab Rev 14: 177–185, 1998
    OpenUrlCrossRefPubMed
  99. 98.
    Leiter EH, Prochazka M, Coleman DL: The non-obese diabetic (NOD) mouse. Am J Pathol 128: 380–383, 1987
    OpenUrlPubMed
  100. 99.↵
    Atkinson MA, Leiter EH: The NOD mouse model of type 1 diabetes: As good as it gets? Nat Med 5: 601–604, 1999
    OpenUrlCrossRefPubMed
  101. 100.↵
    Acha-Orbea H, Scarpellino L: Nonobese diabetic and nonobese nondiabetic mice have unique MHC class II haplotypes. Immunogenetics 34: 57–59, 1991
    OpenUrl
  102. 101.↵
    Watanabe Y, Itoh Y, Yoshida F, Koh N, Tamai H, Fukatsu A, Matsuo S, Hotta N, Sakamoto N: Unique glomerular lesion with spontaneous lipid deposition in glomerular capillary lumina in the NON strain of mice. Nephron 58: 210–218, 1991
    OpenUrlPubMed
  103. 102.↵
    Doi T, Hattori M, Agodoa LY, Sato T, Yoshida H, Striker LJ, Striker GE: Glomerular lesions in nonobese diabetic mouse: Before and after the onset of hyperglycemia. Lab Invest 63: 204–212, 1990
    OpenUrlPubMed
  104. 103.↵
    He CJ, Zheng F, Stitt A, Striker L, Hattori M, Vlassara H: Differential expression of renal AGE-receptor genes in NOD mice: Possible role in nonobese diabetic renal disease. Kidney Int 58: 1931–1940, 2000
    OpenUrlCrossRefPubMed
  105. 104.↵
    Schreyer SA, Wilson DL, LeBoeuf RC: C57BL/6 mice fed high fat diets as models for diabetes-accelerated atherosclerosis. Atherosclerosis 136: 17–24, 1998
    OpenUrlCrossRefPubMed
  106. 105.
    Sharma K, Ziyadeh FN: Renal hypertrophy is associated with upregulation of TGF-beta 1 gene expression in diabetic BB rat and NOD mouse. Am J Physiol 267: F1094–F1101, 1994
    OpenUrlPubMed
  107. 106.↵
    Pankewycz OG, Guan JX, Bolton WK, Gomez A, Benedict JF: Renal TGF-beta regulation in spontaneously diabetic NOD mice with correlations in mesangial cells. Kidney Int 46: 748–758, 1994
    OpenUrlPubMed
  108. 107.↵
    Surwit RS, Feinglos MN, Rodin J, Sutherland A, Petro AE, Opara EC, Kuhn CM, Rebuffe-Scrive M: Differential effects of fat and sucrose on the development of obesity and diabetes in C57BL/6J and A/J mice. Metabolism 44: 645–651, 1995
    OpenUrlCrossRefPubMed
  109. 108.
    Surwit RS, Kuhn CM, Cochrane C, McCubbin JA, Feinglos MN: Diet-induced type II diabetes in C57BL/6J mice. Diabetes 37: 1163–1167, 1988
    OpenUrlAbstract/FREE Full Text
  110. 109.↵
    Petro AE, Cotter J, Cooper DA, Peters JC, Surwit SJ, Surwit RS: Fat, carbohydrate, and calories in the development of diabetes and obesity in the C57BL/6J mouse. Metabolism 53: 454–457, 2004
    OpenUrlCrossRefPubMed
  111. 110.↵
    Towler DA, Bidder M, Latifi T, Coleman T, Semenkovich CF: Diet-induced diabetes activates an osteogenic gene regulatory program in the aortas of low density lipoprotein receptor-deficient mice. J Biol Chem 273: 30427–30434, 1998
    OpenUrlAbstract/FREE Full Text
  112. 111.↵
    Mu JL, Naggert JK, Svenson KL, Collin GB, Kim JH, McFarland C, Nishina PM, Levine DM, Williams KJ, Paigen B: Quantitative trait loci analysis for the differences in susceptibility to atherosclerosis and diabetes between inbred mouse strains C57BL/6J and C57BLKS/J. J Lipid Res 40: 1328–1335, 1999
    OpenUrlAbstract/FREE Full Text
  113. 112.↵
    Noonan WT, Banks RO: Renal function and glucose transport in male and female mice with diet-induced type II diabetes mellitus. Proc Soc Exp Biol Med 225: 221–230, 2000
    OpenUrlCrossRefPubMed
  114. 113.↵
    Chen H, Charlat O, Tartaglia LA, Woolf EA, Weng X, Ellis SJ, Lakey ND, Culpepper J, Moore KJ, Breitbart RE, Duyk GM, Tepper RI, Morgenstern JP. Evidence that the diabetes gene encodes the leptin receptor: Identification of a mutation in the leptin receptor gene in db/db mice. Cell 84: 491–495, 1996
    OpenUrlCrossRefPubMed
  115. 114.↵
    Lee GH, Proenca R, Montez JM, Carroll KM, Darvishzadeh JG, Lee JI, Friedman JM: Abnormal splicing of the leptin receptor in diabetic mice. Nature 379: 632–635, 1996
    OpenUrlCrossRefPubMed
  116. 115.↵
    Lee SM, Bressler R: Prevention of diabetic nephropathy by diet control in the db/db mouse. Diabetes 30: 106–111, 1981
    OpenUrlAbstract/FREE Full Text
  117. 116.↵
    Lee SM, Tutwiler G, Bressler R, Kircher CH: Metabolic control of prevention of nephropathy by 2-tetradecylglycidate in the diabetic mouse (db/db). Diabetes 31: 12–18, 1982
    OpenUrlAbstract/FREE Full Text
  118. 117.↵
    Like AA, Lavine RL, Poffenbarger PL, Chick WL: Studies in the diabetic mutant mouse. VI. Evolution of glomerular lesions and associated proteinuria. Am J Pathol 66: 193–224, 1972
    OpenUrlPubMed
  119. 118.↵
    Cohen MP, Lautenslager GT, Shearman CW: Increased urinary type IV collagen marks the development of glomerular pathology in diabetic d/db mice. Metabolism 50: 1435–1440, 2001
    OpenUrlCrossRefPubMed
  120. 119.↵
    Gartner K: Glomerular hyperfiltration during the onset of diabetes mellitus in two strains of diabetic mice (c57bl/6j db/db and c57bl/ksj db/db). Diabetologia 15: 59–63, 1978
    OpenUrlCrossRefPubMed
  121. 120.↵
    Arakawa K, Ishihara T, Oku A, Nawano M, Ueta K, Kitamura K, Matsumoto M, Saito A: Improved diabetic syndrome in C57BL/KsJ-db/db mice by oral administration of the Na(+)-glucose cotransporter inhibitor T-1095. Br J Pharmacol 132: 578–586, 2001
    OpenUrlCrossRefPubMed
  122. 121.
    Mishra R, Emancipator SN, Miller C, Kern T, Simonson MS: Adipose differentiation-related protein and regulators of lipid homeostasis identified by gene expression profiling in the murine db/db diabetic kidney. Am J Physiol Renal Physiol 286: F913–F921, 2004
    OpenUrlCrossRefPubMed
  123. 122.↵
    Koya D, Haneda M, Nakagawa H, Isshiki K, Sato H, Maeda S, Sugimoto T, Yasuda H, Kashiwagi A, Ways DK, King GL, Kikkawa R: Amelioration of accelerated diabetic mesangial expansion by treatment with a PKC beta inhibitor in diabetic db/db mice, a rodent model for type 2 diabetes. FASEB J 14: 439–447, 2000
    OpenUrlCrossRefPubMed
  124. 123.
    Ha TS, Barnes JL, Stewart JL, Ko CW, Miner JH, Abrahamson DR, Sanes JR, Kasinath BS: Regulation of renal laminin in mice with type II diabetes. J Am Soc Nephrol 10: 1931–1939, 1999
    OpenUrlAbstract/FREE Full Text
  125. 124.↵
    Ziyadeh FN, Hoffman BB, Han DC, Iglesias-De La Cruz MC, Hong SW, Isono M, Chen S, McGowan TA, Sharma K: Long-term prevention of renal insufficiency, excess matrix gene expression, and glomerular mesangial matrix expansion by treatment with monoclonal antitransforming growth factor-beta antibody in db/db diabetic mice. Proc Natl Acad Sci U S A 97: 8015–8020, 2000
    OpenUrlAbstract/FREE Full Text
  126. 125.↵
    Koenig RJ, Araujo DC, Cerami A: Increased hemoglobin AIc in diabetic mice. Diabetes 25: 1–5, 1976
    OpenUrlAbstract/FREE Full Text
  127. 126.
    Leiter EH, Coleman DL, Hummel KP: The influence of genetic background on the expression of mutations at the diabetes locus in the mouse. III. Effect of H-2 haplotype and sex. Diabetes 30: 1029–1034, 1981
    OpenUrlAbstract/FREE Full Text
  128. 127.
    Meade CJ, Brandon DR, Smith W, Simmonds RG, Harris S, Sowter C: The relationship between hyperglycaemia and renal immune complex deposition in mice with inherited diabetes. Clin Exp Immunol 43: 109–120, 1981
    OpenUrlPubMed
  129. 128.
    Leiter EH, Le PH, Coleman DL: Susceptibility to db gene and streptozotocin-induced diabetes in C57BL mice: Control by gender-associated, MHC-unlinked traits. Immunogenetics 26: 6–13, 1987
    OpenUrlCrossRefPubMed
  130. 129.↵
    Leiter EH, Chapman HD, Coleman DL: The influence of genetic background on the expression of mutations at the diabetes locus in the mouse. V. Interaction between the db gene and hepatic sex steroid sulfotransferases correlates with gender-dependent susceptibility to hyperglycemia. Endocrinology 124: 912–922, 1989
    OpenUrlCrossRefPubMed
  131. 130.↵
    Chow F, Ozols E, Nikolic-Paterson DJ, Atkins RC, Tesch GH: Macrophages in mouse type 2 diabetic nephropathy: Correlation with diabetic state and progressive renal injury. Kidney Int 65: 116–128, 2004
    OpenUrlCrossRefPubMed
  132. 131.↵
    Zheng F, Cornacchia F, Schulman I, Banerjee A, Cheng QL, Potier M, Plati AR, Berho M, Elliot SJ, Li J, Fornoni A, Zang YJ, Zisman A, Striker LJ, Striker GE: Development of albuminuria and glomerular lesions in normoglycemic B6 recipients of db/db mice bone marrow: The role of mesangial cell progenitors. Diabetes 53: 2420–2427, 2004
    OpenUrlAbstract/FREE Full Text
  133. 132.↵
    Chua SC Jr, Chung WK, Wu-Peng XS, Zhang Y, Liu SM, Tartaglia L, Leibel RL: Phenotypes of mouse diabetes and rat fatty due to mutations in the OB (leptin) receptor. Science 271: 994–996, 1996
    OpenUrlAbstract/FREE Full Text
  134. 133.↵
    Leibel RL: And finally, genes for human obesity. Nat Genet 16: 218–220, 1997
    OpenUrlCrossRefPubMed
  135. 134.↵
    Chua S Jr, Liu SM, Li Q, Yang L, Thassanapaff VT, Fisher P: Differential beta cell responses to hyperglycaemia and insulin resistance in two novel congenic strains of diabetes (FVB- Lepr (db)) and obese (DBA- Lep (ob)) mice. Diabetologia 45: 976–990, 2002
    OpenUrlCrossRefPubMed
  136. 135.↵
    Lu D, Willard D, Patel IR, Kadwell S, Overton L, Kost T, Luther M, Chen W, Woychik RP, Wilkison WO: Agouti protein is an antagonist of the melanocyte-stimulating-hormone receptor. Nature 371: 799–802, 1994
    OpenUrlCrossRefPubMed
  137. 136.↵
    Ollmann MM, Wilson BD, Yang YK, Kerns JA, Chen Y, Gantz I, Barsh GS: Antagonism of central melanocortin receptors in vitro and in vivo by agouti-related protein. Science 278: 135–138, 1997
    OpenUrlAbstract/FREE Full Text
  138. 137.↵
    Miltenberger RJ, Mynatt RL, Wilkinson JE, Woychik RP: The role of the agouti gene in the yellow obese syndrome. J Nutr 127: 1902S–1907S, 1997
    OpenUrlPubMed
  139. 138.↵
    Michaud EJ, Bultman SJ, Klebig ML, van Vugt MJ, Stubbs LJ, Russell LB, Woychik RP: A molecular model for the genetic and phenotypic characteristics of the mouse lethal yellow (Ay) mutation. Proc Natl Acad Sci U S A 91: 2562–2566, 1994
    OpenUrlAbstract/FREE Full Text
  140. 139.↵
    Ninomiya H, Inomata T, Ogihara K: Obstructive uropathy and hydronephrosis in male KK-Ay mice: A report of cases. J Vet Med Sci 61: 53–57, 1999
    OpenUrlCrossRefPubMed
  141. 140.↵
    Ninomiya H, Inomata T, Ogihara K: Microvasculature of hydronephrotic kidneys in KK-A(Y) mice. J Vet Med Sci 62: 1093–1098, 2000
    OpenUrlCrossRefPubMed
  142. 141.↵
    Leiter EH, Reifsnyder PC: Differential levels of diabetogenic stress in two new mouse models of obesity and type 2 diabetes. Diabetes 53[Suppl 1]: S4–S11, 2004
    OpenUrl
  143. 142.
    Koza RA, Flurkey K, Graunke DM, Braun C, Pan HJ, Reifsnyder PC, Kozak LP, Leiter EH: Contributions of dysregulated energy metabolism to type 2 diabetes development in NZO/H1Lt mice with polygenic obesity. Metabolism 53: 799–808, 2004
    OpenUrlCrossRefPubMed
  144. 143.↵
    Reifsnyder PC, Leiter EH: Deconstructing and reconstructing obesity-induced diabetes (diabesity) in mice. Diabetes 51: 825–832, 2002
    OpenUrlAbstract/FREE Full Text
  145. 144.↵
    Melez KA, Harrison LC, Gilliam JN, Steinberg AD: Diabetes is associated with autoimmunity in the New Zealand obese (NZO) mouse. Diabetes 29: 835–840, 1980
    OpenUrlAbstract/FREE Full Text
  146. 145.↵
    Ma LJ, Fogo AB: Model of robust induction of glomerulosclerosis in mice: Importance of genetic background. Kidney Int 64: 350–355, 2003
    OpenUrlCrossRefPubMed
  147. 146.↵
    Zheng F, Striker GE, Esposito C, Lupia E, Striker LJ: Strain differences rather than hyperglycemia determine the severity of glomerulosclerosis in mice. Kidney Int 54: 1999–2007, 1998
    OpenUrlCrossRefPubMed
  148. 147.↵
    Adhikary L, Chow F, Nikolic-Paterson DJ, Stambe C, Dowling J, Atkins RC, Tesch GH: Abnormal p38 mitogen-activated protein kinase signalling in human and experimental diabetic nephropathy. Diabetologia 47: 1210–1222, 2004
    OpenUrlPubMed
  149. 148.↵
    Naggert JK, Mu JL, Frankel W, Bailey DW, Paigen B: Genomic analysis of the C57BL/Ks mouse strain. Mamm Genome 6: 131–133, 1995
    OpenUrlCrossRefPubMed
  150. 149.↵
    Coleman DL, Hummel KP: Influence of Genetic Background on the Expression of Mutations at the Diabetes Locus in the Mouse. II. Studies on Background Modifiers, New York, Academic Press, 1975, pp 182–187
  151. 150.
    Coleman DL, Hummel KP: Symposium IV: Diabetic syndrome in animals. Influence of genetic background on the expression of mutations at the diabetes locus in the mouse. II. Studies on background modifiers. Isr J Med Sci 11: 708–713, 1975
    OpenUrlPubMed
  152. 151.↵
    Cohen MP, Clements RS, Hud E, Cohen JA, Ziyadeh FN: Evolution of renal function abnormalities in the db/db mouse that parallels the development of human diabetic nephropathy. Exp Nephrol 4: 166–171, 1996
    OpenUrlPubMed
  153. 152.↵
    Giacomelli F, Wiener J: Primary myocardial disease in the diabetic mouse. An ultrastructural study. Lab Invest 40: 460–473, 1979
    OpenUrlPubMed
  154. 153.↵
    Doi K, Matsuzaki H, Tsuda T, Onodera T: Rapid development of renal lesions in diabetic DBA mice infected with the D-variant of encephalomyocarditis virus (EMC-D). Br J Exp Pathol 70: 275–281, 1989
    OpenUrlPubMed
  155. 154.↵
    Shichinohe K, Shimizu M, Ishizaki M, Asakawa M: Kidney complications in EMC virus-induced diabetes in conventional DBA/2 male mice. Jikken Dobutsu 39: 113–116, 1990
    OpenUrlPubMed
  156. 155.↵
    Nishina PM, Wang J, Toyofuku W, Kuypers FA, Ishida BY, Paigen B: Atherosclerosis and plasma and liver lipids in nine inbred strains of mice. Lipids 28: 599–605, 1993
    OpenUrlCrossRefPubMed
  157. 156.↵
    Kirk EA, Moe GL, Caldwell MT, Lernmark JA, Wilson DL, LeBoeuf RC: Hyper- and hypo-responsiveness to dietary fat and cholesterol among inbred mice: Searching for level and variability genes. J Lipid Res 36: 1522–1532, 1995
    OpenUrlAbstract
  158. 157.↵
    Brunnert SR: Morphologic response of myocardium to freeze-thaw injury in mouse strains with dystrophic cardiac calcification. Lab Anim Sci 47: 11–18, 1997
    OpenUrlPubMed
  159. 158.↵
    Brunnert SR, Altman NH: Dystrophic cardiac calcinosis in mice: Abnormal myocardial response to freeze-thaw injury. Lab Anim Sci 40: 616–619, 1990
    OpenUrlPubMed
  160. 159.↵
    Hartner A, Cordasic N, Klanke B, Veelken R, Hilgers KF: Strain differences in the development of hypertension and glomerular lesions induced by deoxycorticosterone acetate salt in mice. Nephrol Dial Transplant 18: 1999–2004, 2003
    OpenUrlCrossRefPubMed
  161. 160.↵
    Zalups RK: The Os/+ mouse: A genetic animal model of reduced renal mass. Am J Physiol 264: F53–F60, 1993
    OpenUrlPubMed
  162. 161.↵
    Fornoni A, Lenz O, Striker LJ, Striker GE: Glucose induces clonal selection and reversible dinucleotide repeat expansion in mesangial cells isolated from glomerulosclerosis-prone mice. Diabetes 52: 2594–2602, 2003
    OpenUrlAbstract/FREE Full Text
  163. 162.
    Cornacchia F, Fornoni A, Plati AR, Thomas A, Wang Y, Inverardi L, Striker LJ, Striker GE: Glomerulosclerosis is transmitted by bone marrow-derived mesangial cell progenitors. J Clin Invest 108: 1649–1656, 2001
    OpenUrlCrossRefPubMed
  164. 163.↵
    Esposito C, He CJ, Striker GE, Zalups RK, Striker LJ: Nature and severity of the glomerular response to nephron reduction is strain-dependent in mice. Am J Pathol 154: 891–897, 1999
    OpenUrlCrossRefPubMed
  165. 164.↵
    Epstein PN, Overbeek PA, Means AR: Calmodulin-induced early-onset diabetes in transgenic mice. Cell 58: 1067–1073, 1989
    OpenUrlCrossRefPubMed
  166. 165.↵
    Epstein PN, Ribar TJ, Decker GL, Yaney G, Means AR: Elevated beta-cell calmodulin produces a unique insulin secretory defect in transgenic mice. Endocrinology 130: 1387–1393, 1992
    OpenUrlCrossRefPubMed
  167. 166.↵
    Carlson EC, Audette JL, Veitenheimer NJ, Risan JA, Laturnus DI, Epstein PN: Ultrastructural morphometry of capillary basement membrane thickness in normal and transgenic diabetic mice. Anat Rec 271A: 332–341, 2003
    OpenUrlCrossRef
  168. 167.↵
    Reddi AS, Oppermann W, Velasco CA, Camerini-Davalos RA: Diabetic microangiopathy in KK mice. II. Suppression of glomerulosclerosis by pyridinolcarbamate. Exp Mol Pathol 26: 325–339, 1977
    OpenUrlCrossRefPubMed
  169. 168.↵
    Reddi AS, Wehner H, Khan MY, Camerini-Davalos RA: Kidney disease in KK mice: Structural, biochemical and functional relationships. Adv Exp Med Biol 246: 135–145, 1988
    OpenUrlPubMed
  170. 169.↵
    Kondo K, Nozawa K, Romida T, Ezaki K: Inbred strains resulting from Japanese mice. Bull Exp Anim 6: 107–112, 1957
    OpenUrl
  171. 170.↵
    Suto J, Matsuura S, Imamura K, Yamanaka H, Sekikawa K: Genetic analysis of non-insulin-dependent diabetes mellitus in KK and KK-Ay mice. Eur J Endocrinol 139: 654–661, 1998
    OpenUrlAbstract
  172. 171.↵
    Liao J, Kobayashi M, Kanamuru Y, Nakamura S, Makita Y, Funabiki K, Horikoshi S, Tomino Y: Effects of candesartan, an angiotensin II type 1 receptor blocker, on diabetic nephropathy in KK/Ta mice. J Nephrol 16: 841–849, 2003
    OpenUrlPubMed
  173. 172.↵
    Camerini-Davalos RA, Oppermann W, Treser G, Ehrenreich T, Lange K, Levine R: Glomerulosclerosis in a strain of genetically diabetic mice. Diabetes 17: 301–849, 1968
    OpenUrlPubMed
  174. 173.↵
    Reddi AS, Velasco CA, Reddy PR, Khan MY, Camerini-Davalos RA: Diabetic microangiopathy in KK mice. VI. Effect of glycemic control on renal glycoprotein metabolism and established glomerulosclerosis. Exp Mol Pathol 53: 140–151, 1990
    OpenUrlCrossRefPubMed
  175. 174.↵
    Allen TJ, Cooper ME, Lan HY: Use of genetic mouse models in the study of diabetic nephropathy. Curr Atheroscler Rep 6: 197–202, 2004
    OpenUrlCrossRefPubMed
  176. 175.↵
    Jenkins AJ, Lyons TJ, Zheng D, Otvos JD, Lackland DT, Mcgee D, Garvey WT, Klein RL: Lipoproteins in the DCCT/EDIC cohort: Associations with diabetic nephropathy. Kidney Int 64: 817–828, 2003
    OpenUrlCrossRefPubMed
  177. 176.↵
    Liberopoulos E, Siamopoulos K, Elisaf M: Apolipoprotein E and renal disease. Am J Kidney Dis 43: 223–233, 2004
    OpenUrlCrossRefPubMed
  178. 177.↵
    Mahley RW: Apolipoprotein E: Cholesterol transport protein with expanding role in cell biology. Science 240: 622–630, 1988
    OpenUrlAbstract/FREE Full Text
  179. 178.↵
    Utermann G: Apolipoprotein E polymorphism in health and disease. Am Heart J 113: 433–440, 1987
    OpenUrlCrossRefPubMed
  180. 179.↵
    Mahley RW, Huang Y: Apolipoprotein E: From atherosclerosis to Alzheimer’s disease and beyond. Curr Opin Lipidol 10: 207–217, 1999
    OpenUrlCrossRefPubMed
  181. 180.↵
    Araki S, Koya D, Makiishi T, Sugimoto T, Isono M, Kikkawa R, Kashiwagi A, Haneda M: APOE polymorphism and the progression of diabetic nephropathy in Japanese subjects with type 2 diabetes: results of a prospective observational follow-up study. Diabetes Care 26: 2416–2420, 2003
    OpenUrlAbstract/FREE Full Text
  182. 181.
    Eto M, Saito M, Okada M, Kume Y, Kawasaki F, Matsuda M, Yoneda M, Matsuki M, Takigami S, Kaku K: Apolipoprotein E genetic polymorphism, remnant lipoproteins, and nephropathy in type 2 diabetic patients. Am J Kidney Dis 40: 243–251, 2002
    OpenUrlCrossRefPubMed
  183. 182.
    Orchard TJ, Chang YF, Ferrell RE, Petro N, Ellis DE: Nephropathy in type 1 diabetes: A manifestation of insulin resistance and multiple genetic susceptibilities? Further evidence from the Pittsburgh Epidemiology of Diabetes Complication Study. Kidney Int 62: 963–970, 2002
    OpenUrlCrossRefPubMed
  184. 183.
    Shcherbak NS: Apolipoprotein E gene polymorphism is not a strong risk factor for diabetic nephropathy and retinopathy in type I diabetes: Case-control study. BMC Med Genet 2: 8–970, 2001
    OpenUrlCrossRefPubMed
  185. 184.
    Hadjadj S, Gallois Y, Simard G, Bouhanick B, Passa P, Grimaldi A, Drouin P, Tichet J, Marre M: Lack of relationship in long-term type 1 diabetic patients between diabetic nephropathy and polymorphisms in apolipoprotein epsilon, lipoprotein lipase and cholesteryl ester transfer protein. Genetique de la Nephropathie Diabetique Study Group. Donnees Epidemiologiques sur le Syndrome D’Insulino-Resistance Study Group. Nephrol Dial Transplant 15: 1971–1976, 2000
    OpenUrlCrossRefPubMed
  186. 185.
    Tarnow L, Stehouwer CD, Emeis JJ, Poirier O, Cambien F, Hansen BV, Parving HH: Plasminogen activator inhibitor-1 and apolipoprotein E gene polymorphisms and diabetic angiopathy. Nephrol Dial Transplant 15: 625–630, 2000
    OpenUrlCrossRefPubMed
  187. 186.
    Chowdhury TA, Dyer PH, Kumar S, Gibson SP, Rowe BR, Davies SJ, Marshall SM, Morris PJ, Gill GV, Feeney S, Maxwell P, Savage D, Boulton AJ, Todd JA, Dunger D, Barnett AH, Bain SC. Association of apolipoprotein epsilon2 allele with diabetic nephropathy in Caucasian subjects with IDDM. Diabetes 47: 278–280, 1998
    OpenUrlFREE Full Text
  188. 187.
    Werle E, Fiehn W, Hasslacher C: Apolipoprotein E polymorphism and renal function in German type 1 and type 2 diabetic patients. Diabetes Care 21: 994–998, 1998
    OpenUrlAbstract/FREE Full Text
  189. 188.
    Boizel R, Benhamou PY, Corticelli P, Valenti K, Bosson JL, Halimi S, Boize R: ApoE polymorphism and albuminuria in diabetes mellitus: A role for LDL in the development of nephropathy in NIDDM? Nephrol Dial Transplant 13: 72–75, 1998
    OpenUrlCrossRefPubMed
  190. 189.
    Onuma T, Laffel LM, Angelico MC, Krolewski AS: Apolipoprotein E genotypes and risk of diabetic nephropathy. J Am Soc Nephrol 7: 1075–1078, 1996
    OpenUrlAbstract
  191. 190.↵
    Eto M, Horita K, Morikawa A, Nakata H, Okada M, Saito M, Nomura M, Abiko A, Iwashima Y, Ikoda A: Increased frequency of apolipoprotein epsilon 2 allele in non-insulin dependent diabetic (NIDDM) patients with nephropathy. Clin Genet 48: 288–292, 1995
    OpenUrlPubMed
  192. 191.↵
    Araki S, Moczulski DK, Hanna L, Scott LJ, Warram JH, Krolewski AS: APOE polymorphisms and the development of diabetic nephropathy in type 1 diabetes: Results of case-control and family-based studies. Diabetes 49: 2190–2195, 2000
    OpenUrlAbstract/FREE Full Text
  193. 192.↵
    Plump AS, Smith JD, Hayek T, Aalto-Setala K, Walsh A, Verstuyft JG, Rubin EM, Breslow JL: Severe hypercholesterolemia and atherosclerosis in apolipoprotein E-deficient mice created by homologous recombination in ES cells. Cell 71: 343–353, 1992
    OpenUrlCrossRefPubMed
  194. 193.↵
    Lassila M, Seah KK, Allen TJ, Thallas V, Thomas MC, Candido R, Burns WC, Forbes JM, Calkin AC, Cooper ME, Jandeleit-Dahm KA: Accelerated nephropathy in diabetic apolipoprotein e-knockout mouse: Role of advanced glycation end products. J Am Soc Nephrol 15: 2125–2138, 2004
    OpenUrlAbstract/FREE Full Text
  195. 194.↵
    Nakayama T, Soma M, Takahashi Y, Izumi Y, Kanmatsuse K, Esumi M: Association analysis of CA repeat polymorphism of the endothelial nitric oxide synthase gene with essential hypertension in Japanese. Clin Genet 51: 26–30, 1997
    OpenUrlPubMed
  196. 195.
    Arngrimsson R, Hayward C, Nadaud S, Baldursdottir A, Walker JJ, Liston WA, Bjarnadottir RI, Brock DJ, Geirsson RT, Connor JM, Soubrier F: Evidence for a familial pregnancy-induced hypertension locus in the eNOS-gene region. Am J Hum Genet 61: 354–362, 1997
    OpenUrlCrossRefPubMed
  197. 196.
    Yokoyama K, Tsukada T, Matsuoka H, Hara S, Yamada A, Kawaguchi Y: High accumulation of endothelial nitric oxide synthase (ecNOS): A gene polymorphism in patients with end-stage renal disease. Nephron 79: 360–361, 1998
    OpenUrlCrossRefPubMed
  198. 197.
    Yoshimura M, Yasue H, Nakayama M, Shimasaki Y, Sumida H, Sugiyama S, Kugiyama K, Ogawa H, Ogawa Y, Saito Y, Miyamoto Y, Nakao K: A missense Glu298Asp variant in the endothelial nitric oxide synthase gene is associated with coronary spasm in the Japanese. Hum Genet 103: 65–69, 1998
    OpenUrlCrossRefPubMed
  199. 198.↵
    Miyamoto Y, Saito Y, Kajiyama N, Yoshimura M, Shimasaki Y, Nakayama M, Kamitani S, Harada M, Ishikawa M, Kuwahara K, Ogawa E, Hamanaka I, Takahashi N, Kaneshige T, Teraoka H, Akamizu T, Azuma N, Yoshimasa Y, Yoshimasa T, Itoh H, Masuda I, Yasue H, Nakao K: Endothelial nitric oxide synthase gene is positively associated with essential hypertension. Hypertension 32: 3–8, 1998
    OpenUrlAbstract/FREE Full Text
  200. 199.↵
    Ksiazek P, Wojewoda P, Muc K, Buraczynska M: Endothelial nitric oxide synthase gene intron 4 polymorphism in type 2 diabetes mellitus. Mol Diagn 7: 119–123, 2003
    OpenUrlCrossRefPubMed
  201. 200.
    Frost D, Chitu J, Meyer M, Beischer W, Pfohl M: Endothelial nitric oxide synthase (ecNOS) 4 a/b gene polymorphism and carotid artery intima-media thickness in type-1 diabetic patients. Exp Clin Endocrinol Diabetes 111: 12–15, 2003
    OpenUrlCrossRefPubMed
  202. 201.↵
    Zanchi A, Moczulski DK, Hanna LS, Wantman M, Warram JH, Krolewski AS: Risk of advanced diabetic nephropathy in type 1 diabetes is associated with endothelial nitric oxide synthase gene polymorphism. Kidney Int 57: 405–413, 2000
    OpenUrlCrossRefPubMed
  203. 202.↵
    Neugebauer S, Baba T, Watanabe T: Association of the nitric oxide synthase gene polymorphism with an increased risk for progression to diabetic nephropathy in type 2 diabetes. Diabetes 49: 500–503, 2000
    OpenUrlAbstract
  204. 203.↵
    Li C, Dong Y, Lu W: [The association between polymorphism of endothelial nitric oxide synthase gene and diabetic nephropathy]. In Chinese. Zhonghua Nei Ke Za Zhi 40: 729–732, 2001
    OpenUrlPubMed
  205. 204.↵
    Asakimori Y, Yorioka N, Yamamoto I, Okumoto S, Doi S, Hirai T, Taniguchi Y: Endothelial nitric oxide synthase intron 4 polymorphism influences the progression of renal disease. Nephron 89: 219–223, 2001
    OpenUrlCrossRefPubMed
  206. 205.↵
    Noiri E, Satoh H, Taguchi J, Brodsky SV, Nakao A, Ogawa Y, Nishijima S, Yokomizo T, Tokunaga K, Fujita T: Association of eNOS Glu298Asp polymorphism with end-stage renal disease. Hypertension 40: 535–540, 2002
    OpenUrlAbstract/FREE Full Text
  207. 206.↵
    Lin S, Qu H, Qiu M: Allele A in intron 4 of ecNOS gene will not increase the risk of diabetic nephropathy in type 2 diabetes of Chinese population. Nephron 91: 768–540, 2002
    OpenUrlCrossRefPubMed
  208. 207.
    Shimizu T, Onuma T, Kawamori R, Makita Y, Tomino Y: Endothelial nitric oxide synthase gene and the development of diabetic nephropathy. Diabetes Res Clin Pract 58: 179–185, 2002
    OpenUrlCrossRefPubMed
  209. 208.
    Rippin JD, Patel A, Belyaev ND, Gill GV, Barnett AH, Bain SC: Nitric oxide synthase gene polymorphisms and diabetic nephropathy. Diabetologia 46: 426–428, 2003
    OpenUrl
  210. 209.
    Degen B, Schmidt S, Ritz E: A polymorphism in the gene for the endothelial nitric oxide synthase and diabetic nephropathy. Nephrol Dial Transplant 16: 185–428, 2001
    OpenUrlCrossRefPubMed
  211. 210.↵
    Freedman BI, Yu H, Anderson PJ, Roh BH, Rich SS, Bowden DW: Genetic analysis of nitric oxide and endothelin in end-stage renal disease. Nephrol Dial Transplant 15: 1794–1800, 2000
    OpenUrlCrossRefPubMed
  212. 211.↵
    Knowles JW, Reddick RL, Jennette JC, Shesely EG, Smithies O, Maeda N: Enhanced atherosclerosis and kidney dysfunction in eNOS(−/−)Apoe(−/−) mice are ameliorated by enalapril treatment. J Clin Invest 105: 451–458, 2000
    OpenUrlCrossRefPubMed
  213. 212.
    Kuhlencordt PJ, Gyurko R, Han F, Scherrer-Crosbie M, Aretz TH, Hajjar R, Picard MH, Huang PL: Accelerated atherosclerosis, aortic aneurysm formation, and ischemic heart disease in apolipoprotein E/endothelial nitric oxide synthase double-knockout mice. Circulation 104: 448–454, 2001
    OpenUrlAbstract/FREE Full Text
  214. 213.
    Chen J, Kuhlencordt PJ, Astern J, Gyurko R, Huang PL: Hypertension does not account for the accelerated atherosclerosis and development of aneurysms in male apolipoprotein e/endothelial nitric oxide synthase double knockout mice. Circulation 104: 2391–2394, 2001
    OpenUrlAbstract/FREE Full Text
  215. 214.↵
    Ozaki M, Kawashima S, Yamashita T, Hirase T, Namiki M, Inoue N, Hirata K, Yasui H, Sakurai H, Yoshida Y, Masada M, Yokoyama M: Overexpression of endothelial nitric oxide synthase accelerates atherosclerotic lesion formation in apoE-deficient mice. J Clin Invest 110: 331–340, 2002
    OpenUrlCrossRefPubMed
  216. 215.↵
    Brownlee M, Cerami A, Vlassara H: Advanced glycosylation end products in tissue and the biochemical basis of diabetic complications. N Engl J Med 318: 1315–1321, 1988
    OpenUrlCrossRefPubMed
  217. 216.↵
    Wendt T, Tanji N, Guo J, Hudson BI, Bierhaus A, Ramasamy R, Arnold B, Nawroth PP, Yan SF, D’Agati V, Schmidt AM: Glucose, glycation, and RAGE: Implications for amplification of cellular dysfunction in diabetic nephropathy. J Am Soc Nephrol 14: 1383–1395, 2003
    OpenUrlAbstract/FREE Full Text
  218. 217.↵
    Oldfield MD, Bach LA, Forbes JM, Nikolic-Paterson D, McRobert A, Thallas V, Atkins RC, Osicka T, Jerums G, Cooper ME: Advanced glycation end products cause epithelial-myofibroblast transdifferentiation via the receptor for advanced glycation end products (RAGE). J Clin Invest 108: 1853–1863, 2001
    OpenUrlCrossRefPubMed
  219. 218.↵
    Li JH, Wang W, Huang XR, Oldfield M, Schmidt AM, Cooper ME, Lan HY: Advanced glycation end products induce tubular epithelial-myofibroblast transition through the RAGE-ERK1/2 MAP kinase signaling pathway. Am J Pathol 164: 1389–1397, 2004
    OpenUrlCrossRefPubMed
  220. 219.↵
    Tanji N, Markowitz GS, Fu C, Kislinger T, Taguchi A, Pischetsrieder M, Stern D, Schmidt AM, D’Agati VD: Expression of advanced glycation end products and their cellular receptor RAGE in diabetic nephropathy and nondiabetic renal disease. J Am Soc Nephrol 11: 1656–1666, 2000
    OpenUrlAbstract/FREE Full Text
  221. 220.↵
    Wendt TM, Tanji N, Guo J, Kislinger TR, Qu W, Lu Y, Bucciarelli LG, Rong LL, Moser B, Markowitz GS, Stein G, Bierhaus A, Liliensiek B, Arnold B, Nawroth PP, Stern DM, D’Agati VD, Schmidt AM: RAGE drives the development of glomerulosclerosis and implicates podocyte activation in the pathogenesis of diabetic nephropathy. Am J Pathol 162: 1123–1137, 2003
    OpenUrlCrossRefPubMed
  222. 221.↵
    Yamamoto Y, Kato I, Doi T, Yonekura H, Ohashi S, Takeuchi M, Watanabe T, Yamagishi S, Sakurai S, Takasawa S, Okamoto H, Yamamoto H: Development and prevention of advanced diabetic nephropathy in RAGE-overexpressing mice. J Clin Invest 108: 261–268, 2001
    OpenUrlCrossRefPubMed
  223. 222.↵
    Weigert C, Brodbeck K, Brosius FC 3rd, Huber M, Lehmann R, Friess U, Facchin S, Aulwurm S, Haring HU, Schleicher ED, Heilig CW: Evidence for a novel TGF-β1-independent mechanism of fibronectin production in mesangial cells overexpressing glucose transporters. Diabetes 52: 527–535, 2003
    OpenUrlAbstract/FREE Full Text
  224. 223.↵
    Heilig CW, Concepcion LA, Riser BL, Freytag SO, Zhu M, Cortes P: Overexpression of glucose transporters in rat mesangial cells cultured in a normal glucose milieu mimics the diabetic phenotype. J Clin Invest 96: 1802–1814, 1995
    OpenUrlCrossRefPubMed
  225. 224.↵
    Heilig C, Zaloga C, Lee M, Zhao X, Riser B, Brosius F, Cortes P: Immunogold localization of high-affinity glucose transporter isoforms in normal rat kidney. Lab Invest 73: 674–684, 1995
    OpenUrlPubMed
  226. 225.↵
    Chen S, Heilig KO, Brosius F III, Heilig C: Diabetes increases glomerular GLUT1 and antisense-GLUT1 protects against diabetic glomerulosclerosis [Abstract]. J Am Soc Nephrol 14: 46A–684, 2003
    OpenUrlCrossRef
  227. 226.↵
    Heilig CW, Liu Y, England RL, Freytag SO, Gilbert JD, Heilig KO, Zhu M, Concepcion LA, Brosius FC 3rd: D-glucose stimulates mesangial cell GLUT1 expression and basal and IGF-I-sensitive glucose uptake in rat mesangial cells: Implications for diabetic nephropathy. Diabetes 46: 1030–1039, 1997
    OpenUrlAbstract/FREE Full Text
  228. 227.↵
    Heilig KO, Chen S, Xiang M, Brosius FC III, Heilig CW: Transgenic overexpression of GLUT1 in glomeruli produces features of diabetic nephropathy in mice. J Am Soc Nephrol 15: 263A–1039, 2004
    OpenUrl
  229. 228.
    Spencer MW, Muhlfeld AS, Segerer S, Hudkins KL, Kirk E, LeBoeuf RC, Alpers CE: Hyperglycemia and hyperlipidemia act synergistically to induce renal disease in LDL receptor-deficient BALB mice. Am J Nephrol 24: 20–31, 2004
    OpenUrlCrossRefPubMed
  230. 229.
    Honjo K, Doi K, Doi C, Mitsuoka T: Histopathology of streptozotocin-induced diabetic DBA/2N and CD-1 mice. Lab Anim 20: 298–303, 1986
    OpenUrlCrossRefPubMed
  231. 230.
    Kimura I, Matsui T, Kimura M: Increase in basal pulse rate and blood pressure by the diabetic state in KK-CAy mice, alloxan-mice and streptozotocin-mice. Jpn J Pharmacol 46: 93–96, 1988
    OpenUrlPubMed
  232. 231.
    Kume E, Doi C, Itagaki S, Nagashima Y, Doi K: Glomerular lesions in unilateral nephrectomized and diabetic (UN-D) mice. J Vet Med Sci 54: 1085–1090, 1992
    OpenUrlPubMed
  233. 232.
    Wolf J, Lilly F, Shin SI: The influence of genetic background on the susceptibility of inbred mice to streptozotocin-induced diabetes. Diabetes 33: 567–571, 1984
    OpenUrlAbstract/FREE Full Text
  234. 233.
    Doi K, Onodera T, Tsuda T, Matsuzaki H, Mitsuoka T: Histopathology of BALB/c mice infected with the D variant of encephalomyocarditis virus. Br J Exp Pathol 69: 395–401, 1988
    OpenUrlPubMed
  235. 234.
    Hirasawa K, Takeda M, Itagaki S, Doi K: Involvement of macrophages in the development of encephalomyocarditis (EMC) virus-induced diabetes in mice. Exp Anim 45: 77–80, 1996
    OpenUrlCrossRefPubMed
  236. 235.
    Maeda M, Yabuki A, Suzuki S, Matsumoto M, Taniguchi K, Nishinakagawa H: Renal lesions in spontaneous insulin-dependent diabetes mellitus in the nonobese diabetic mouse: Acute phase of diabetes. Vet Pathol 40: 187–195, 2003
    OpenUrlCrossRefPubMed
  237. 236.
    Wilson KH, Eckenrode SE, Li QZ, Ruan QG, Yang P, Shi JD, Davoodi-Semiromi A, McIndoe RA, Croker BP, She JX: Microarray analysis of gene expression in the kidneys of new- and post-onset diabetic NOD mice. Diabetes 52: 2151–2159, 2003
    OpenUrlAbstract/FREE Full Text
  238. 237.
    Kim JH, Taylor PN, Young D, Karst SY, Nishina PM, Naggert JK: New leptin receptor mutations in mice: Lepr(db-rtnd), Lepr(db-dmpg) and Lepr(db-rlpy). J Nutr 133: 1265–1271, 2003
    OpenUrlAbstract/FREE Full Text
  239. 238.
    Haluzik M, Colombo C, Gavrilova O, Chua S, Wolf N, Chen M, Stannard B, Dietz KR, Le Roith D, Reitman ML: Genetic background (C57BL/6J versus FVB/N) strongly influences the severity of diabetes and insulin resistance in ob/ob mice. Endocrinology 145: 3258–3264, 2004
    OpenUrlCrossRefPubMed
  240. 239.
    Liu YL, Emilsson V, Cawthorne MA: Leptin inhibits glycogen synthesis in the isolated soleus muscle of obese (ob/ob) mice. FEBS Lett 411: 351–355, 1997
    OpenUrlCrossRefPubMed
  241. 240.
    Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM: Positional cloning of the mouse obese gene and its human homologue. Nature 372: 425–432, 1994
    OpenUrlCrossRefPubMed
  242. 241.
    Suto J, Matsuura S, Imamura K, Yamanaka H, Sekikawa K: Genetics of obesity in KK mouse and effects of A(y) allele on quantitative regulation. Mamm Genome 9: 506–510, 1998
    OpenUrlCrossRefPubMed
  243. 242.
    Suto J, Matsuura S, Yamanaka H, Sekikawa K: Quantitative trait loci that regulate plasma lipid concentration in hereditary obese KK and KK-Ay mice. Biochim Biophys Acta 1453: 385–395, 1999
    OpenUrlPubMed
  244. 243.
    Hustad CM, Perry WL, Siracusa LD, Rasberry C, Cobb L, Cattanach BM, Kovatch R, Copeland NG, Jenkins NA: Molecular genetic characterization of six recessive viable alleles of the mouse agouti locus. Genetics 140: 255–265, 1995
    OpenUrlPubMed
  245. 244.
    Klebig ML, Wilkinson JE, Geisler JG, Woychik RP: Ectopic expression of the agouti gene in transgenic mice causes obesity, features of type II diabetes, and yellow fur. Proc Natl Acad Sci U S A 92: 4728–4732, 1995
    OpenUrlAbstract/FREE Full Text
  246. 245.
    Prpic V, Watson PM, Frampton IC, Sabol MA, Jezek GE, Gettys TW: Differential mechanisms and development of leptin resistance in A/J versus C57BL/6J mice during diet-induced obesity. Endocrinology 144: 1155–1163, 2003
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

Journal of the American Society of Nephrology: 16 (1)
Journal of the American Society of Nephrology
Vol. 16, Issue 1
1 Jan 2005
  • Table of Contents
  • Index by author
View Selected Citations (0)
Print
Download PDF
Sign up for Alerts
Email Article
Thank you for your help in sharing the high-quality science in JASN.
Enter multiple addresses on separate lines or separate them with commas.
Mouse Models of Diabetic Nephropathy
(Your Name) has sent you a message from American Society of Nephrology
(Your Name) thought you would like to see the American Society of Nephrology web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Mouse Models of Diabetic Nephropathy
Matthew D. Breyer, Erwin Böttinger, Frank C. Brosius, Thomas M. Coffman, Raymond C. Harris, Charles W. Heilig, Kumar Sharma
JASN Jan 2005, 16 (1) 27-45; DOI: 10.1681/ASN.2004080648

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Request Permissions
Share
Mouse Models of Diabetic Nephropathy
Matthew D. Breyer, Erwin Böttinger, Frank C. Brosius, Thomas M. Coffman, Raymond C. Harris, Charles W. Heilig, Kumar Sharma
JASN Jan 2005, 16 (1) 27-45; DOI: 10.1681/ASN.2004080648
del.icio.us logo Digg logo Reddit logo Twitter logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like

Jump to section

  • Article
    • Abstract
    • Working Definition of DN
    • Characterization of DN in Mice
    • Assessment of Hyperglycemia in Mice
    • Assessment of Renal Function in Mice
    • Models of Type 1 Diabetes
    • Mouse Models of Type 2 Diabetes
    • Inbred Mice: Strain Dependence of DN
    • Monogenic Mutations and Transgenic Mice
    • Conclusions
    • Acknowledgments
    • References
  • Figures & Data Supps
  • Info & Metrics
  • View PDF

More in this TOC Section

  • Podocyte Aging: Why and How Getting Old Matters
  • Acquired Decline in Ultrafiltration in Peritoneal Dialysis: The Role of Glucose
  • Mendelian Randomization Analysis as a Tool to Gain Insights into Causes of Diseases: A Primer
Show more REVIEWS

Cited By...

  • Vaccination Against Receptor for Advanced Glycation End Products Attenuates the Progression of Diabetic Kidney Disease
  • Drug Testing for Residual Progression of Diabetic Kidney Disease in Mice Beyond Therapy with Metformin, Ramipril, and Empagliflozin
  • Systematic review and meta-analysis of mouse models of diabetes-associated ulcers
  • Paternal multigenerational exposure to an obesogenic diet drives epigenetic predisposition to metabolic disorders
  • Changes in excitability and ion channel expression in neurons of the major pelvic ganglion in female type II diabetic mice
  • Signal integration at the PI3K-p85-XBP1 hub endows coagulation protease activated protein C with insulin-like function
  • Effects of insulin and the glucagon-like peptide 1 receptor agonist liraglutide on the kidney proteome in db/db mice
  • Glomerular Endothelial Mitochondrial Dysfunction Is Essential and Characteristic of Diabetic Kidney Disease Susceptibility
  • Comparison of Glomerular and Podocyte mRNA Profiles in Streptozotocin-Induced Diabetes
  • Low-Dose IL-17 Therapy Prevents and Reverses Diabetic Nephropathy, Metabolic Syndrome, and Associated Organ Fibrosis
  • Four-and-a-Half LIM Domains Protein 2 Is a Coactivator of Wnt Signaling in Diabetic Kidney Disease
  • Diabetes Induces Aberrant DNA Methylation in the Proximal Tubules of the Kidney
  • Albumin stimulates renal tubular inflammation through an HSP70-TLR4 axis in mice with early diabetic nephropathy
  • Vascular Endothelial Growth Factor-A165b Is Protective and Restores Endothelial Glycocalyx in Diabetic Nephropathy
  • Renoprotective effects of combined SGLT2 and ACE inhibitor therapy in diabetic Dahl S rats
  • Progressive Renal Decline: The New Paradigm of Diabetic Nephropathy in Type 1 Diabetes
  • Loss of the Podocyte-Expressed Transcription Factor Tcf21/Pod1 Results in Podocyte Differentiation Defects and FSGS
  • Endogenous Fructose Production and Fructokinase Activation Mediate Renal Injury in Diabetic Nephropathy
  • CD73-Dependent Generation of Adenosine and Endothelial Adora2b Signaling Attenuate Diabetic Nephropathy
  • Knockdown of Glyoxalase 1 Mimics Diabetic Nephropathy in Nondiabetic Mice
  • Phlorizin Pretreatment Reduces Acute Renal Toxicity in a Mouse Model for Diabetic Nephropathy
  • A Role for the Endothelium in Vascular Calcification
  • Celecoxib modifies glomerular basement membrane, mesangium and podocytes in OVE26 mice, but ibuprofen is more detrimental
  • Direct Evidence for Intrarenal Chymase-Dependent Angiotensin II Formation on the Diabetic Renal Microvasculature
  • A Transcriptional Blueprint for Human and Murine Diabetic Kidney Disease
  • eNOS Deficiency Predisposes Podocytes to Injury in Diabetes
  • Intrarenal Dopamine Inhibits Progression of Diabetic Nephropathy
  • Transgenerational Glucose Intolerance With Igf2/H19 Epigenetic Alterations in Mouse Islet Induced by Intrauterine Hyperglycemia
  • Heparanase Is Essential for the Development of Diabetic Nephropathy in Mice
  • Arginase-2 Mediates Diabetic Renal Injury
  • Knockout of Toll-Like Receptor-2 Attenuates Both the Proinflammatory State of Diabetes and Incipient Diabetic Nephropathy
  • Esculetin induced changes in Mmp13 and Bmp6 gene expression and histone H3 modifications attenuate development of glomerulosclerosis in diabetic rats
  • Thyroid hormone ameliorates diabetic nephropathy in a mouse model of type II diabetes
  • Activation of Vascular Bone Morphogenetic Protein Signaling in Diabetes Mellitus
  • The Protective Role of Smad7 in Diabetic Kidney Disease: Mechanism and Therapeutic Potential
  • Antiatherosclerotic and Renoprotective Effects of Ebselen in the Diabetic Apolipoprotein E/GPx1-Double Knockout Mouse
  • BTBR Ob/Ob Mutant Mice Model Progressive Diabetic Nephropathy
  • Deletion of the Receptor for Advanced Glycation End Products Reduces Glomerulosclerosis and Preserves Renal Function in the Diabetic OVE26 Mouse
  • Inhibition or Deletion of Soluble Epoxide Hydrolase Prevents Hyperglycemia, Promotes Insulin Secretion, and Reduces Islet Apoptosis
  • Mouse Models of Diabetic Nephropathy
  • Vasohibin-1, a Negative Feedback Regulator of Angiogenesis, Ameliorates Renal Alterations in a Mouse Model of Diabetic Nephropathy
  • Effect of the Monocyte Chemoattractant Protein-1/CC Chemokine Receptor 2 System on Nephrin Expression in Streptozotocin-Treated Mice and Human Cultured Podocytes
  • Activation of the Succinate Receptor GPR91 in Macula Densa Cells Causes Renin Release
  • The Use of Animal Models in the Study of Diabetes Mellitus
  • Enhanced Expression of Janus Kinase-Signal Transducer and Activator of Transcription Pathway Members in Human Diabetic Nephropathy
  • Combination therapy with AT1 blocker and vitamin D analog markedly ameliorates diabetic nephropathy: Blockade of compensatory renin increase
  • Overexpression of Calmodulin in Pancreatic {beta} Cells Induces Diabetic Nephropathy
  • From Fibrosis to Sclerosis: Mechanisms of Glomerulosclerosis in Diabetic Nephropathy
  • Long-term hyperglucagonaemia induces early metabolic and renal phenotypes of Type 2 diabetes in mice
  • Overexpression of Angiotensinogen Increases Tubular Apoptosis in Diabetes
  • Can rodent models of diabetic kidney disease clarify the significance of early hyperfiltration?: recognizing clinical and experimental uncertainties
  • Thrombospondin-1 Is an Endogenous Activator of TGF-{beta} in Experimental Diabetic Nephropathy In Vivo
  • L-Carnosine, a Substrate of Carnosinase-1, Influences Glucose Metabolism
  • The Vascular Ectonucleotidase ENTPD1 Is a Novel Renoprotective Factor in Diabetic Nephropathy
  • Targeting glucagon receptor signalling in treating metabolic syndrome and renal injury in Type 2 diabetes: theory versus promise
  • Renal Fibrosis and Glomerulosclerosis in a New Mouse Model of Diabetic Nephropathy and Its Regression by Bone Morphogenic Protein-7 and Advanced Glycation End Product Inhibitors
  • Interstitial Vascular Rarefaction and Reduced VEGF-A Expression in Human Diabetic Nephropathy
  • Recipes for Creating Animal Models of Diabetic Cardiovascular Disease
  • Diabetic Endothelial Nitric Oxide Synthase Knockout Mice Develop Advanced Diabetic Nephropathy
  • A Sensitized Screen of N-ethyl-N-nitrosourea-Mutagenized Mice Identifies Dominant Mutants Predisposed to Diabetic Nephropathy
  • Poly(ADP-Ribose) Polymerase Inhibitors Ameliorate Nephropathy of Type 2 Diabetic Leprdb/db Mice
  • Endothelial Nitric Oxide Synthase Deficiency Produces Accelerated Nephropathy in Diabetic Mice
  • Heme Oxygenase-2 Deficiency Contributes to Diabetes-Mediated Increase in Superoxide Anion and Renal Dysfunction
  • Leukocyte Recruitment and Vascular Injury in Diabetic Nephropathy
  • Frontiers in Diabetic Nephropathy: Can We Predict Who Will Get Sick?
  • Glucose-Induced Reactive Oxygen Species Cause Apoptosis of Podocytes and Podocyte Depletion at the Onset of Diabetic Nephropathy
  • Activation of G{alpha}q-Coupled Signaling Pathways in Glomerular Podocytes Promotes Renal Injury
  • Antiangiogenic Endostatin Peptide Ameliorates Renal Alterations in the Early Stage of a Type 1 Diabetic Nephropathy Model
  • Characterization of Susceptibility of Inbred Mouse Strains to Diabetic Nephropathy
  • Accelerated Glucose Intolerance, Nephropathy, and Atherosclerosis in Prostaglandin D2 Synthase Knock-out Mice
  • Google Scholar

Similar Articles

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Articles

  • Current Issue
  • Early Access
  • Subject Collections
  • Article Archive
  • ASN Annual Meeting Abstracts

Information for Authors

  • Submit a Manuscript
  • Author Resources
  • Editorial Fellowship Program
  • ASN Journal Policies
  • Reuse/Reprint Policy

About

  • JASN
  • ASN
  • ASN Journals
  • ASN Kidney News

Journal Information

  • About JASN
  • JASN Email Alerts
  • JASN Key Impact Information
  • JASN Podcasts
  • JASN RSS Feeds
  • Editorial Board

More Information

  • Advertise
  • ASN Podcasts
  • ASN Publications
  • Become an ASN Member
  • Feedback
  • Follow on Twitter
  • Password/Email Address Changes
  • Subscribe to ASN Journals
  • Wolters Kluwer Partnership

© 2022 American Society of Nephrology

Print ISSN - 1046-6673 Online ISSN - 1533-3450

Powered by HighWire