Cellular and humoral immune responses in the early stages of diabetic nephropathy in NOD mice

https://doi.org/10.1016/j.jaut.2008.12.003Get rights and content

Abstract

This study was designed to examine immunopathology of diabetic nephropathy in non-obese diabetic (NOD) mice and to investigate the involvement of cellular and humoral immunity at various time points after diabetes onset. We found that the glomeruli of diabetic NOD mice were infiltrated with T and B cells, as well as CD11c+ dendritic cells, which had close contact with CD4+ and CD8+ T cells in the infiltrates. We also found that IgG deposits in the glomeruli of diabetic NOD mice were accompanied by the presence of complement C3. Moreover, the serum from diabetic mice contained autoantibodies directed towards components of the glomeruli and these antibodies were not present in non-diabetic NOD mice. The immune changes in the kidney occurred together with increasing kidney weight and urinary albumin excretion along with duration of diabetes. We provide evidence that infiltrating lymphocytes and anti-kidney autoantibodies may be involved in diabetic nephropathy in autoimmune diabetes in the NOD mouse. Understanding the role that the immune system plays in the pathogenesis of diabetic nephropathy could lead to identification of new strategies and/or additional therapeutic targets for prevention and treatment of diabetic nephropathy.

Introduction

Diabetic nephropathy is the leading cause of end-stage renal failure in young people and affects about 40% of individuals with long-standing type 1 diabetes [1]. Current therapy, which includes treatment for hyperglycemia, hypertension and dyslipidemia can slow the rate of progression of diabetic nephropathy [2], but eventually end-stage renal failure will still occur in a proportion of patients [3]. Therefore, the effects of currently used treatments must be maximized and identification of new strategies and additional therapeutic targets for treating diabetic nephropathy will be important.

Many factors are involved in the pathogenesis of diabetic nephropathy, which in humans is defined by a progressive rise in albuminuria, together with increasing blood pressure, leading to a fall in glomerular filtration and ultimately end-stage renal failure [3]. Microalbuminuria may occur as early as 5 years after diagnosis of diabetes [3]. Structural abnormalities include glomerular basement membrane thickening, increased number of mesangial cells and mesangial expansion [4]. Metabolic factors, such as the formation of advanced glycation end products and increased flux through the polyol pathways, have been implicated [5]. In addition, growth factors (transforming growth factor TGF-β, insulin-like growth factor IGF-1 and vascular endothelial growth factor VEGF) may also play a role [5], [6]. Although the pathogenesis of diabetic nephropathy is not considered to be primarily immune mediated, nevertheless studies examining renal biopsies in patients with type 1 diabetes have noted T cell infiltration of the juxtaglomerular apparatus [7], [8]. Macrophages have also been detected in kidney sections from patients with diabetic nephropathy although no distinction was made between patients with type 1 or type 2 diabetes [9].

To facilitate dissection of pathogenesis of diabetic nephropathy, a variety of experimental rat and mouse models have been used, many of which relate to type 2 diabetes. In the present study, we focused on the non-obese diabetic (NOD) mouse model that develops spontaneous autoimmune diabetes similar to human type 1 diabetes [10]. Many studies of diabetic nephropathy were performed in chemically-induced diabetic animal models and NOD mice have not been studied much in terms of diabetic nephropathy, perhaps because of the potentially long and variable time to onset of diabetes [11]. However, a few studies have shown that NOD mice could develop renal pathology after the onset of diabetes [12], [13]. To test the hypothesis that immune responses are involved in the development of diabetic nephropathy, we investigated cellular and humoral immune parameters in spontaneous diabetic NOD mice.

Section snippets

Animals

Female NOD/Caj mice were used in this study. All the mice were housed in individually-ventilated filter cages with autoclaved food under specific pathogen-free (SPF) conditions with a 12-h dark/light cycle. The use of the animals and the procedures applied in this study were approved by the Institutional Animal Care and Use Committee of Yale University.

Determination of diabetes and diabetic nephropathy

NOD mice were observed for diabetes development by weekly screening for glycosuria. Diabetes was confirmed following 2 consecutive readings of

Cellular immune responses in diabetic kidneys

To investigate whether cellular immune responses are involved in the pathogenesis of diabetic nephropathy, we examined mononuclear cell infiltration in glomeruli with confocal microscopy at monthly intervals after the development of diabetes. All the kidneys were perfused before preparation of tissue sections. We found that CD4+ (Fig. 1B–D, F–H) and CD8+ (Fig. 1J–L) T cells were present in the glomeruli in diabetic mice. Dendritic cells (CD11c+) were also present in the infiltrates, in close

Discussion

Our study has shown that, firstly, in the early stages of diabetes in the NOD mouse, both T and B cells infiltrate the glomeruli. Secondly, we have demonstrated that serum components from diabetic NOD mice, but not non-diabetic NOD mice, bind to normal kidney sections, indicating the presence of autoantibodies to the kidney. These autoantibodies only develop after the mice become diabetic and appear to be directed towards renal endothelial cells and the glomerular basement membrane. Thirdly, we

Acknowledgements

Xiaoyan Xiao is a scholarship recipient from the China Scholarship Council (2007-102113). This study was supported by an innovative partnership grant to Li Wen and F. Susan Wong from the JDRF (19-2006-1075). We are grateful to Arthur Hand (University of Connecticut) for the electron microscopy work, Octavian Hengariu for measurement of insulin autoantibodies, Jan Czyzyk for help with independent evaluation of nephropathy and Bin Lu for help with animal care.

References (29)

  • A. Lewis et al.

    Diabetic nephropathy, inflammation, hyaluronan and interstitial fibrosis

    Histol Histopathol

    (2008)
  • N. Giarratana et al.

    Animal models of spontaneous autoimmune disease: type 1 diabetes in the nonobese diabetic mouse

    Methods Mol Biol

    (2007)
  • M.D. Breyer et al.

    Mouse models of diabetic nephropathy

    J Am Soc Nephrol

    (2005)
  • M. Maeda et al.

    Renal lesions in spontaneous insulin-dependent diabetes mellitus in the nonobese diabetic mouse: acute phase of diabetes

    Vet Pathol

    (2003)
  • Cited by (76)

    • IDO1 as a new immune biomarker for diabetic nephropathy and its correlation with immune cell infiltration

      2021, International Immunopharmacology
      Citation Excerpt :

      Among the infiltrating immune cells, monocytes/macrophages comprised a high proportion and caused podocyte damage through multiple pathways in DN patients, which concurred with the results of previous studies [15]. Xiao et al. [29] found the glomeruli of non-obese diabetic mice were infiltrated with D11c + dendritic cells, which suggested their role in DN pathogenesis. Increased levels of CD4 + CXCR5 + PD-1 + T follicular helper cells were also discovered in the peripheral blood of DN patients [30].

    • Renal diseases and the role of complement: Linking complement to immune effector pathways and therapeutics

      2021, Advances in Immunology
      Citation Excerpt :

      Unfortunately, these findings could not be replicated by others (Østergaard et al., 2016). In contrast, the nonobese diabetic and the OVE26 mouse models both feature C3 deposition (Xiao et al., 2009; Yang et al., 2011), and in other models C5a inhibition ameliorates tubulointerstitial damage (Yiu et al., 2018). In rat models, C5 inhibition also reduces C3 staining and mesangial expansion (Fujita et al., 1999) and treatment with C3aR and C5ar antagonists led to downregulation of alpha smooth muscle actin, TGF-b, fibronectin and beta catenin, which are major components of kidney scarring, and preserved renal function (Li et al., 2015, 2014).

    View all citing articles on Scopus
    1

    F.S. Wong and L. Wen contributed equally to this study.

    View full text