Elsevier

Journal of Autoimmunity

Volume 35, Issue 4, December 2010, Pages 414-423
Journal of Autoimmunity

Successful modulation of type 2 diabetes in db/db mice with intra-bone marrow–bone marrow transplantation plus concurrent thymic transplantation

https://doi.org/10.1016/j.jaut.2010.09.001Get rights and content

Abstract

There is increasing evidence that both autoimmune and autoinflammatory mechanisms are involved in the development of not only type 1 diabetes mellitus (T1 DM), but also type 2 diabetes mellitus (T2 DM). Our laboratory has focused on this concept, and in earlier efforts replaced the bone marrow cells (BMCs) of leptin receptor-deficient (db/db) mice, an animal model of T2DM, with those of normal C57BL/6 (B6) mice by IBM–BMT. However, the outcome was poor due to incomplete recovery of T cell function. Therefore, we hypothesized that intra-bone marrow–bone marrow transplantation plus thymus transplantation (IBM–BMT + TT) could be used to treat T2 DM by normalizing the T cell imbalance. Hence we addressed this issue by using such dual transplantation and demonstrate herein that seven weeks later, recipient db/db mice manifested improved body weight, reduced levels of blood glucose, and a reduction of plasma IL-6 and IL-1β. More importantly, this treatment regimen showed normal CD4/CD8 ratios, and increased plasma adiponectin levels, insulin sensitivity, and the number of insulin-producing cells. Furthermore, the expression of pancreatic pAKT, pLKB1, pAMPK and HO-1 was increased in the mice treated with IBM–BMT + TT. Our data show that IBM–BMT + TT treatment normalizes T cell subsets, cytokine imbalance and insulin sensitivity in the db/db mouse, suggesting that IBM–BMT + TT is a viable therapeutic option in the treatment of T2 DM.

Introduction

There is a virtual epidemic of type 2 diabetes, and although the mechanisms for this increase are not entirely clear, it has become the focus of both genetic and environmental research [1]. Clearly, inflammation has a critical role in the development of metabolic diseases, including obesity and T2 DM [1]. Recently, it has been shown that obese adipose tissue activates CD8T cells, resulting in promoting the recruitment and activation of macrophages in the adipose tissue [2]; macrophages have been shown to infiltrate the adipose tissue in mice and humans [3]. Adipocytes regulate and mediate inflammatory cytokines such as tumor necrosis factor-α (TNFα), IL-6, matrixmetalloproteinases (MMPs), peroxisome proliferation activated receptor-r (PPAR-r) and fatty acid-binding protein –4. These cytokines inhibit or enhance each other, and their activities contribute to insulin resistance [4]. As such, both an autoinflammatory as well as an autoimmune response are involved in the pathogenesis of T2 DM.

Bone marrow transplantation (BMT) has been demonstrated to treat hematopoietic disorders, metabolic disorders and autoimmune diseases [5], [6], [7], [8], [9], [10], [11], [12], [13], [14], [15], [16], [17], [18], [19]. We have recently found that intra-bone marrow–BMT (IBM–BMT) treatment is an advantageous strategy for allogeneic BMT, compared with conventional intravenous BMT [23], since IBM–BMT can replace not only hemopoietic cells (including hemopoietic stem cells:HSCs) but also stromal cells (including mesenchymal stem cells:MSCs). In addition, we have very recently found that thymus transplantation combined with BMT (BMT + TT) is a powerful strategy to ameliorate thymic involution in recipient mice due to aging or irradiation [20], [21], [22].

Based on these findings, we carried out IBM–BMT in combination with newborn thymus transplantation (TT) in db/db mice. We here demonstrate that, after IBM–BMT + TT treatment in db/db mice, insulin sensitivity increases and blood glucose levels decrease, resulting from the normalization of balance of lymphocyte subsets and cytokines, followed by enhanced expression of pAKT, pLKB1, pAMPK, insulin receptor phosphorylation and HO-1. This suggests that the maintenance of the balance of lymphocyte subsets and cytokine production by IBM–BMT + TT treatment is essential for the amelioration of T2 DM in db/db mice.

Section snippets

Animals

Five-week-old BKS.Cg-m+Leprdb/+Leprdb/J (H-2kd) (db/db) mice, BKS. Cg-m+/+Leprdb/J(H-2kd) (lean) mice and C57BL/6 (B6) (H-2kb) mice were purchased from Charles River Laboratories (Yokohama, Japan) and SLC (Shizuoka, Japan) and maintained in animal facilities under specific pathogen-free conditions. All procedures were performed under protocols approved by the Institutional Animal Care and Use Committee at Kansai Medical University. Body weight and blood glucose levels were measured each week.

Body weight, blood glucose levels, insulin sensitivity, and plasma adiponectin, insulin, IL-6 and IL-1β levels

In our preliminary experiments, we carried out IBM–BMT alone (without TT). The IBM–BMT-treated db/db mice showed decreased blood glucose levels (<150 mg/ml) one week after the treatment but rapid increases in blood glucose levels 2 weeks after the treatment; the mice became susceptible to severe infection due to a rebound phenomenon, and died. Therefore, in the present study, we carried out IBM–BMT + TT, and non-treated db/db mice were used as the control.

As seen in Fig. 1A, a gain in body

Discussion

Leptin is an adipocyte-derived hormone that links nutritional status with neuroendocrine and immune functions. Leptin has been shown to modulate T cell proliferation, to promote Th1 responses, and to protect thymocytes from corticosteroid-induced apoptosis in vitro [26], [27], [28], [29]. Leptin-deficient ob/ob mice and leptin receptor-deficient db/db mice exhibit severe hereditary obesity [30], [31] and display hormonal imbalances and hematolymphoid defects [32], [33]. Db/db mice exhibit a

Acknowledgments

We would like to thank Mr. Hilary Eastwick-Field and Ms. K. Ando for their help in the preparation of the manuscript. This study was mainly supported by the 21st Century Center of Excellence (COE) program of the Ministry of Education, Culture, Sports, Science and Technology. This study was also supported by grants from Haiteku Research Center of the Ministry of Education, Health and Labour Sciences Research Grants, the Science Frontier program of the Ministry of Education, Culture, Sports,

References (58)

  • S. Deane et al.

    On reversing the persistence of memory: hematopoietic stem cell transplant for autoimmune disease in the first ten years

    J Autoimmun

    (2008)
  • T. Kushida et al.

    Intra-bone marrow injection of allogeneic bone marrow cells: a powerful new strategy for treatment of intractable autoimmune diseases in MRL/lpr mice

    Blood

    (2001)
  • S.J. Peterson et al.

    The L-4F mimetic peptide prevents insulin resistance through increased levels of HO-1, pAMPK, and pAKT in obese mice

    J Lipid Res

    (2009)
  • C. Martin-Romero et al.

    Human leptin enhances activation and proliferation of human circulating T lymphocytes

    Cell Immunol

    (2000)
  • M. Kimura et al.

    T lymphopenia in obese diabetic (db/db) mice is non-selective and thymus independent

    Life Sci

    (1998)
  • L.K. Gilliam et al.

    Autoimmunity and clinical course in children with type 1, type 2, and type 1.5 diabetes

    J Autoimmun

    (2005)
  • M. Flaquer et al.

    Bone marrow transplantation induces normoglycemia in a type 2 diabetes mellitus murine model

    Transplant Proc

    (2009)
  • G. Fantuzzi

    Adipose tissue, adipokines, and inflammation

    J Allergy Clin Immunol

    (2005)
  • I. Boumaza et al.

    Autologous bone marrow-derived rat mesenchymal stem cells promote PDX-1 and insulin expression in the islets, alter T cell cytokine pattern and preserve regulatory T cells in the periphery and induce sustained normoglycemia

    J Autoimmun

    (2009)
  • N.D. Theise et al.

    Liver from bone marrow in humans

    Hepatology

    (2000)
  • F. Chow et al.

    Macrophages in mouse type 2 diabetic nephropathy: correlation with diabetic state and progressive renal injury

    Kidney Int

    (2004)
  • J.P. Despres et al.

    Abdominal obesity and metabolic syndrome

    Nature

    (2006)
  • S. Nishimura et al.

    CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity

    Nat Med

    (2009)
  • G.S. Hotamisligil

    Inflammation and metabolic disorders

    Nature

    (2006)
  • T. Suganami et al.

    A paracrine loop between adipocytes and macrophages aggravates inflammatory changes: role of free fatty acids and tumor necrosis factor alpha

    Arterioscler Thromb Vasc Biol

    (2005)
  • R.A. Clift et al.

    Follow-up 26 years after treatment for acute myelogenous leukemia

    N Engl J Med

    (2004)
  • S. Ikehara et al.

    Prevention of type I diabetes in nonobese diabetic mice by allogenic bone marrow transplantation

    Proc Natl Acad Sci U S A

    (1985)
  • S. Ikehara et al.

    Rationale for bone marrow transplantation in the treatment of autoimmune diseases

    Proc Natl Acad Sci U S A

    (1985)
  • S. Than et al.

    Bone marrow transplantation as a strategy for treatment of non-insulin-dependent diabetes mellitus in KK-Ay mice

    J Exp Med

    (1992)
  • Cited by (0)

    View full text