N-3 fatty acids modulate Th1 and Th2 dichotomy in diabetic pregnancy and macrosomia

https://doi.org/10.1016/j.jaut.2006.03.003Get rights and content

Abstract

We assessed the implication of Th (helper)-cells and the modulation of the Th1/Th2 dichotomy by n-3 polyunsaturated fatty acids (PUFA) in type I diabetic pregnancy (DP) and macrosomia. Female gestant rats fed a standard diet or n-3 PUFA regimen were rendered diabetic by administration of five low doses of streptozotocin. The macrosomic (MAC) offspring were sacrificed at the age of 90 days. The mRNAs of IL-2 and IFN-γ (Th1 cytokines) and IL-4 (Th2 cytokine) were downregulated in the pancreas and spleen of diabetic pregnant rats. The levels of IL-10 mRNA, another Th2 cytokine, were unchanged in the spleen or upregulated in the pancreas of these animals. Feeding an n-3 PUFA diet to rats with DP upregulated IL-10 mRNA in the pancreas and IL-4 and IL-10 mRNA in the spleen. In MAC offspring, high expression of IL-2 and IFN-γ mRNA, but not of Th2 cytokines, was observed. The n-3 PUFA diet diminished Th1 mRNA quantities and increased the levels of IL-4, but not of IL-10, mRNA in MAC offspring. Our study shows that DP is associated with a decreased Th1 phenotype and IL-4 mRNA expression in the pancreas and spleen, and an n-3 PUFA diet upregulates Th2 profile. In MAC offspring, the Th1 phenotype is upregulated and an n-3 PUFA diet downregulates this phenomenon.

Introduction

Through different experimental models of diabetes, it has been well established that the secretion of cytokines plays an important role in the regulation of tolerance of islet antigens [1]. These cytokines are produced during the islet inflammatory response and their production, in part, may explain the ability of CD4+ Th cells alone to cause β-cell destruction [1]. On the basis of production of cytokines, Th cells can be classified into two principal populations, Th1 and Th2. Th1 cells support cell-mediated immunity and as a consequence promote inflammation, cytotoxicity and delayed type hypersensitivity, whereas Th2 cells support humoral immunity and downregulate the inflammatory actions of Th1 cells [2]. Th1 cells secrete IL-2, IFN-γ and TNF-β, while Th2 cells secrete IL-4, IL-5, IL-6, IL-10 and IL-13 [3].

In the non-obese diabetic (NOD) mouse, the most common animal model of human type I diabetes, insulin-dependent diabetes mellitus (IDDM) results from autoimmune destruction of pancreatic β-cells, mediated by both CD4+ and CD8+T-cells [1]. It has been reported that the pathogenic activity of β-cell autoreactive CD4+T-cells in these mice can be inhibited if the predominant pattern of cytokines is shifted from Th1 (primarily IFN-γ) to Th2 (primarily IL-4) profile [2]. It is interesting to mention that the hyporesponsiveness of Th2 cells may also favor the development of a Th1 cell-mediated environment in the pancreas, leading to loss of immunological tolerance to β-cell autoantigens and onset of diabetes in NOD mice and humans [4]. In human beings suffering from type I diabetes mellitus, a low secretion of IL-4 from mitogen-stimulated T cells has been reported [5]. The general conclusion is drawn that Th1 cytokines play a pathogenic role, while Th2 cytokines assure regulatory function, and thus mediate protection during diabetes [1], [2], [6], [7], [8].

Epidemiological, clinical and experimental studies have suggested that maternal type I diabetes during pregnancy is an important risk factor for fetal overnutrition and macrosomia, and for the development of an increased susceptibility to obesity and diabetes in their offspring [9], [10]. The abnormalities in humoral and cell-mediated immunity in type I diabetic females may persist during pregnancy and, hence may complicate immune-foetal interaction [11]. As far as T-cell activation during pregnancy in type I diabetic mothers and their macrosomic offspring is concerned, only a few studies are available on the subject [11]. In fact, fully activated T-cells are detected in the cord blood of infants and mothers with type I diabetes but not in infants from normal mothers [11]. Moreover, from birth up to 15 years of age, the percentage of total T-cells was higher in children of type I diabetic mothers than in those of healthy mothers [12]. An increase in MHC class II positive lymphocytes has been observed in infants of type I diabetic mothers compared to controls [13]. Moreover, the newborns of type I diabetic mothers showed a significant reduction in natural killer (NK) lymphocytes, indicating a deficit in natural immunity at birth [14]. Newborns of type I diabetic mothers showed an increase in the number of CD4+T-cells [15]. Furthermore, production of IL-1 and IL-2 in the culture supernatants of mitogen-stimulated lymphocytes of these newborns was higher than that in controls [16]. However, the role of pancreatic and splenic Th subsets in the modulation of these pathologies has not yet been explored.

It has been recently well established that n-3 polyunsaturated fatty acids (PUFA) exert immunosuppressive effects [17] and, consequently, these agents have been used in the management of a number of inflammatory and autoimmune diseases, including rheumatoid arthritis and multiple sclerosis [18]. Generally, n-3 PUFA suppress mitogen-stimulated proliferation of lymphocytes isolated from lymph nodes [17]. Hence, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) are the most potent immunomodulators of the n-3 PUFA family [19]. It has been shown that dietary EPA and DHA are equipotent in inhibiting IL-2 production in mice [19] and rats [20]. The production of IFN-γ is also decreased by these fatty acids [21]. The n-3 PUFA have also been used in the management of diabetes mellitus in human beings [22] and experimental models [17].

The above observations demonstrate that type I diabetes mellitus is a Th1-biased disease and n-3 polyunsaturated fatty acids (PUFA) exert immunomodulatory effects. In the present study, we propose a hypothesis that the Th1 and Th2 dichotomy may be involved in diabetes mellitus during pregnancy and macrosomia, and that a diet rich in n-3 PUFA may modulate Th1 and Th2 balance during these pathologies.

Section snippets

Chemicals

EPAX-7010 was generously provided by Polaris, Quimper (France). SYBR® Green supermix was procured from Bio-Rad, Marnes-la-Coquette (France). Vegetable oil (Isio-4) was purchased from Lesieur, Neuilly-sur-Seine (France). All other chemical products were obtained from In-vitrogen Life Technologies, Groningen (The Netherlands).

Animals, diets and experimental design

Adult Wistar rats were obtained from Ifa-Credo (Lyon, France). After mating, the first day of gestation was estimated by the presence of spermatozoids in vaginal smears.

Expression of mRNA of Th1 and Th2 cytokines in the pancreas and spleen of rats with DP

Fig. 1 shows that the expression of mRNA of Th1 cytokines (IL-2 and IFN-γ) and IL-4 was downregulated in the pancreas and spleen of animals with DP in comparison with control rats. However, the expression of IL-10 mRNA was either upregulated in the pancreas or remained unchanged in the spleen of rats with DP as compared to control animals (Fig. 1).

The expression of mRNA of IL-2 and IFN-γ in the pancreas and spleen of rats with DP was not significantly different between the standard diet and the

Discussion

The administration of five low doses of STZ to mice [6], [30] and rats [23] represents a good model of diabetes development, and this is for several reasons: 1) islet lesions in this experimental model resemble to those of human insulitis, with a predominance of CD8+T-cells [30]; 2) the animals used are normal and do not have an underlying immune abnormalities like BB rat, being lymphopenic with few peripheral CD8+T-cells [31], and NOD mice which have systemic immune abnormalities [32]; 3) the

Acknowledgements

We gratefully acknowledge the Office of Scholarship Programme of IDB that granted a scholarship to one of the authors (A. Yessoufou). This work was supported by a financial support from the Burgundy Region, Dijon (France).

References (66)

  • J.P. Mordes et al.

    Animal models of diabetes

    Am J Med

    (1981)
  • L. Aerts et al.

    Intra-uterine transmission of disease

    Placenta

    (2003)
  • G. Reinhard et al.

    Shifts in the TH1/TH2 balance during human pregnancy correlate with apoptotic changes

    Biochem Biophys Res Commun

    (1998)
  • K.J. Pennline et al.

    Recombinant human IL-10 prevents the onset of diabetes in the non-obese diabetic mouse

    Clin Immunol Immunopathol

    (1994)
  • N.A. Khan et al.

    Inhibition of diabetes in NOD mice by human pregnancy factor

    Hum Immunol

    (2001)
  • E. Herrera

    Implications of dietary fatty acids during pregnancy on placental, foetal and postnatal development – a Review

    Placenta

    (2002)
  • K. Ghebremeskel et al.

    Liver triacylglycerols and free fatty acids in streptozotocin-induced diabetic rats have atypical n-6 and n-3 pattern

    Com Biochem Physiol Part C

    (2002)
  • P. Zhang et al.

    Dietary (n-3) polyunsaturated fatty acids modulate murine Th1/Th2 balance toward the Th2 pole by suppression of Th1 development

    J Nutr

    (2005)
  • A. Bonin et al.

    Regulation of calcium signalling by docosahexaenoic acid in human T-cells. Implication of CRAC channels

    J Lipid Res

    (2000)
  • A. Denys et al.

    Eicosapentaenoic acid and docosahexaenoic acid modulate MAP kinase (ERK1/ERK2) signaling in human T cells

    J Lipid Res

    (2001)
  • S. Madani et al.

    Diacylglycerols containing Omega 3 and Omega 6 fatty acids bind to RasGRP and modulate MAP kinase activation

    J Biol Chem

    (2004)
  • M. Lee et al.

    Prevention of autoimmune insulitis by delivery of a chimeric plasmid encoding interleukin-4 and interleukin-10

    J Control Release

    (2003)
  • A. Rabinovitch

    Immunoregulatory and cytokine imbalances in the pathogenesis of IDDM. Therapeutic intervention by immunostimulation?

    Diabetes

    (1994)
  • M.A. Berman et al.

    Decreased IL-4 production in new onset type I insulin-dependent diabetes mellitus

    J Immunol

    (1996)
  • D.V. Serreze et al.

    Th1 to Th2 cytokine shifts in non-obese diabetic mice: sometimes an outcome, rather than the cause, of diabetes resistance elicited by immunostimulation

    J Immunol

    (2001)
  • H. Merzouk et al.

    Implication of lipids in macrosomia of diabetic pregnancy: can n-3 polyunsaturated fatty acids exert beneficial effects?

    Clin Sci

    (2003)
  • I.M. Evers et al.

    Risk of complications of pregnancy in women with type 1 diabetes: nationwide prospective study in the Netherlands

    Br Med J

    (2004)
  • U. Roll et al.

    Alterations of lymphocytes subsets in children of diabetic mothers

    Diabetologia

    (1994)
  • U. Di Mario et al.

    Immunology in diabetic pregnancy: activated T-cells in diabetic mothers and neonates

    Diabetologia

    (1987)
  • Lapolla A, Sanzari MC, Znacanaro F, Masin M, Guerriero A, Piva I, et al. Study on lymphocyte subpopulation in diabetic...
  • C. Giordano et al.

    Analysis of T-lymphocytte subsets after phyhemagglutinin stimulation in normal and type 1 diabetic mothers and their infants

    Am J Reprod Immunol

    (1982)
  • A. El Mohandes et al.

    Lymphocyte populations and responses to mitogens in infants of diabetic mothers

    J Clin Labo Immunol

    (1982)
  • Calder PC. N-3 polyunsaturated fatty acids and inflammation: from molecular biology to the clinic. Lipids...
  • Cited by (45)

    • TFOS DEWS II Management and Therapy Report

      2017, Ocular Surface
      Citation Excerpt :

      As a result, there has been scientific interest in understanding whether increasing systemic ω-3 EFA levels through nutritional intervention, to lower the ω-6:ω-3 ratio, can yield systemic anti-inflammatory effects that are beneficial for conditions with an inflammatory overlay, including DED. Omega-3 EFAs are recognized to have a broad range of systemic anti-inflammatory effects, including inhibiting the production of several key pro-inflammatory cytokines (such as IL-1, IL-2 and TNF-α) [828–830] and preventing T-lymphocyte proliferation [831,832], processes that have been implicated in the pathogenesis of DED. Several laboratory studies have shown that fatty acid enrichment can impart lacrimal gland changes and alter the ocular surface response to pharmacologic-induced models of DED [833–838].

    • 3,10-Dihydroxy-decanoic acid, isolated from royal jelly, stimulates Th1 polarising capability of human monocyte-derived dendritic cells

      2011, Food Chemistry
      Citation Excerpt :

      However, both fatty acids showed the suppressive activity on the KLH-induced immune response in vivo (Vucevic et al., 2007). Many polyunsaturated fatty acids, commonly used as dietary supplements, inhibit inflammatory and immune responses, including T-cell proliferation and IL-2 production (Khan, Yessoufou, Kim, & Hichami, 2006). Cavaglieri et al. (2003) demonstrated that different short-chain fatty acids, either inhibited clonal expansion of T cells, production of IL-2 and IFN-γ, or enhanced IFN-γ and IL-10 production.

    • Intramuscular delivery of a naked DNA plasmid encoding proinsulin and pancreatic regenerating III protein ameliorates type 1 diabetes mellitus

      2011, Pharmacological Research
      Citation Excerpt :

      Cytokines secreted by Th1 cells like IFN-γ and IL-2 were proved to initiate β cell destruction, leading to the development of diabetes [34]; whereas Th2 cytokines such as IL-10 and TGF-β act as protective factors in pathogenesis of T1DM [35]. Previous researches demonstrated that an abnormal Th1 shift was occurred in T1DM patients [36,37]. In this research, intramuscular delivery of pReg/PI or pPI significantly down-regulated production of Th 1 cytokines (IL-2 and IFN-γ), and enhanced concentration of IL-10 and TGF-β in the serum.

    • Docosahexaenoic acid modulates the expression of T-bet and GATA-3 transcription factors, independently of PPARα, through suppression of MAP kinase activation

      2009, Biochimie
      Citation Excerpt :

      Since a variety of strategies which prevent the progression of autoimmune diseases also induce a shift in TH1/TH2 balance, n-3 PUFA have been shown to modulate T-cell differentiation. A diet-enriched with these fatty acids during diabetic pregnancy was found to upregulate the TH2 phenotype in rats [3]. Wallace et al. [4] have observed that, in healthy mice, in vitro production of TH1 cytokines, i.e., IL-2 and IFN-γ, in spleens of animals fed an n-3 PUFA diet for six weeks, was significantly decreased as compared to those fed the standard diet.

    View all citing articles on Scopus
    View full text