Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Immune dysfunction in developmental programming of type 2 diabetes mellitus

Abstract

Intrauterine growth restriction (IUGR) is a common complication of pregnancy and increases the risk of the offspring developing type 2 diabetes mellitus (T2DM) later in life. Alterations in the immune system are implicated in the pathogenesis of IUGR-induced T2DM. The development of the fetal immune system is a delicate balance as it must remain tolerant of maternal antigens whilst also preparing for the post-birth environment. In addition, the fetal immune system is susceptible to an altered intrauterine milieu caused by maternal and placental inflammatory mediators or secondary to nutrient and oxygen deprivation. Pancreatic-resident macrophages populate the pancreas during fetal development, and their phenotype is dynamic through the neonatal period. Furthermore, macrophages in the islets are instrumental in islet development as they influence β-cell proliferation and islet neogenesis. In addition, cytokines, derived from β-cells and macrophages, are important to islet homeostasis in the fetus and adult and, when perturbed, can cause islet dysfunction. Several activated immune pathways have been identified in the islets of people who experienced IUGR, with alternations in the levels of IL-1β and IL-4 as well as changes in TGFβ signalling. Leptin levels are also altered. Immunomodulation has shown therapeutic benefit in T2DM and might be particularly useful in IUGR-induced T2DM.

Key points

  • Fetal immune development is susceptible to an abnormal intrauterine milieu, and alterations have been implicated in the development of type 2 diabetes mellitus (T2DM) following intrauterine growth restriction (IUGR).

  • Pancreatic islet macrophages are instrumental in islet development and homeostasis in adults.

  • Levels of cytokines and immune mediators that are involved in T2DM pathogenesis are also elevated in offspring exposed to IUGR.

  • Leptin stimulates IL-1β production in islets, and levels of leptin are reduced in fetal islets and elevated in adult islets of offspring exposed to IUGR.

  • The results of limited reports evaluating the therapeutic effect of immunomodulation are promising for the treatment of IUGR-induced T2DM.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Fetal immune development.
Fig. 2: Pancreatic immune development.
Fig. 3: Macrophages influence islet development.
Fig. 4: Resident macrophages sense their micro and systemic environments.
Fig. 5: Pancreatic inflammation subsequent to IUGR.

Similar content being viewed by others

References

  1. Boehmer, B. H., Limesand, S. W. & Rozance, P. J. The impact of IUGR on pancreatic islet development and β-cell function. J. Endocrinol. 235, R63–R76 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Rashid, C. S., Bansal, A. & Simmons, R. A. Oxidative stress, intrauterine growth restriction, and developmental programming of type 2 diabetes. Physiology 33, 348–359 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Barker, D. J., Winter, P. D., Osmond, C., Margetts, B. & Simmonds, S. J. Weight in infancy and death from ischaemic heart disease. Lancet 2, 577–580 (1989).

    Article  CAS  PubMed  Google Scholar 

  4. Hales, C. N. & Barker, D. J. Type 2 (non-insulin-dependent) diabetes mellitus: the thrifty phenotype hypothesis. Diabetologia 35, 595–601 (1992).

    Article  CAS  PubMed  Google Scholar 

  5. Kermack, W. O., McKendrick, A. G. & McKinlay, P. L. Death-rates in Great Britain and Sweden: expression of specific mortality rates as products of two factors, and some consequences thereof. J. Hyg. 34, 433–457 (1934).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Ravelli, G. P., Stein, Z. A. & Susser, M. W. Obesity in young men after famine exposure in utero and early infancy. N. Engl. J. Med. 295, 349–353 (1976).

    Article  CAS  PubMed  Google Scholar 

  7. Barker, D. J. et al. Type 2 (non-insulin-dependent) diabetes mellitus, hypertension and hyperlipidaemia (syndrome X): relation to reduced fetal growth. Diabetologia 36, 62–67 (1993).

    Article  CAS  PubMed  Google Scholar 

  8. Fall, C. H. et al. Fetal and infant growth and cardiovascular risk factors in women. BMJ 310, 428–432 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Hales, C. N. et al. Fetal and infant growth and impaired glucose tolerance at age 64. BMJ 303, 1019–1022 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Phipps, K. et al. Fetal growth and impaired glucose tolerance in men and women. Diabetologia 36, 225–228 (1993).

    Article  CAS  PubMed  Google Scholar 

  11. Curhan, G. C. et al. Birth weight and adult hypertension, diabetes mellitus, and obesity in US men. Circulation 94, 3246–3250 (1996).

    Article  CAS  PubMed  Google Scholar 

  12. Egeland, G. M., Skjaerven, R. & Irgens, L. M. Birth characteristics of women who develop gestational diabetes: population based study. BMJ 321, 546–547 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Hales, C. N. & Barker, D. J. The thrifty phenotype hypothesis. Br. Med. Bull. 60, 5–20 (2001).

    Article  CAS  PubMed  Google Scholar 

  14. Jaquet, D., Gaboriau, A., Czernichow, P. & Levy-Marchal, C. Insulin resistance early in adulthood in subjects born with intrauterine growth retardation. J. Clin. Endocrinol. Metab. 85, 1401–1406 (2000).

    CAS  PubMed  Google Scholar 

  15. Leger, J. et al. Reduced final height and indications for insulin resistance in 20 year olds born small for gestational age: regional cohort study. BMJ 315, 341–347 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Lithell, H. O. et al. Relation of size at birth to non-insulin dependent diabetes and insulin concentrations in men aged 50–60 years. BMJ 312, 406–410 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. McKeigue, P. M., Lithell, H. O. & Leon, D. A. Glucose tolerance and resistance to insulin-stimulated glucose uptake in men aged 70 years in relation to size at birth. Diabetologia 41, 1133–1138 (1998).

    Article  CAS  PubMed  Google Scholar 

  18. Rich-Edwards, J. W. et al. Longitudinal study of birth weight and adult body mass index in predicting risk of coronary heart disease and stroke in women. BMJ 330, 1115 (2005).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Valdez, R., Athens, M. A., Thompson, G. H., Bradshaw, B. S. & Stern, M. P. Birthweight and adult health outcomes in a biethnic population in the USA. Diabetologia 37, 624–631 (1994).

    Article  CAS  PubMed  Google Scholar 

  20. Forsen, T. et al. The fetal and childhood growth of persons who develop type 2 diabetes. Ann. Intern. Med. 133, 176–182 (2000).

    Article  CAS  PubMed  Google Scholar 

  21. Davey Smith, G. et al. Education and occupational social class: which is the more important indicator of mortality risk? J. Epidemiol. Community Health 52, 153–160 (1998).

    Article  CAS  PubMed  Google Scholar 

  22. Banderali, G. et al. Short and long term health effects of parental tobacco smoking during pregnancy and lactation: a descriptive review. J. Transl Med. 13, 327 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Lindsay, K. L., Buss, C., Wadhwa, P. D. & Entringer, S. The interplay between maternal nutrition and stress during pregnancy: issues and considerations. Ann. Nutr. Metab. 70, 191–200 (2017).

    Article  CAS  PubMed  Google Scholar 

  24. Parker, J. D., Schoendorf, K. C. & Kiely, J. L. Associations between measures of socioeconomic status and low birth weight, small for gestational age, and premature delivery in the United States. Ann. Epidemiol. 4, 271–278 (1994).

    Article  CAS  PubMed  Google Scholar 

  25. Rich-Edwards, J. W. et al. Birth weight and risk of cardiovascular disease in a cohort of women followed up since 1976. BMJ 315, 396–400 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. de Rooij, S. R., Wouters, H., Yonker, J. E., Painter, R. C. & Roseboom, T. J. Prenatal undernutrition and cognitive function in late adulthood. Proc. Natl Acad. Sci. USA 107, 16881–16886 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Roseboom, T., de Rooij, S. & Painter, R. The Dutch famine and its long-term consequences for adult health. Early Hum. Dev. 82, 485–491 (2006).

    Article  PubMed  Google Scholar 

  28. Ravelli, A. C. et al. Glucose tolerance in adults after prenatal exposure to famine. Lancet 351, 173–177 (1998).

    Article  CAS  PubMed  Google Scholar 

  29. Zur, R. L., Kingdom, J. C., Parks, W. T. & Hobson, S. R. The placental basis of fetal growth restriction. Obstet. Gynecol. Clin. North Am. 47, 81–98 (2020).

    Article  PubMed  Google Scholar 

  30. Geelhoed, J. J. et al. Preeclampsia and gestational hypertension are associated with childhood blood pressure independently of family adiposity measures: the Avon Longitudinal Study of Parents and Children. Circulation 122, 1192–1199 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Palmsten, K., Buka, S. L. & Michels, K. B. Maternal pregnancy-related hypertension and risk for hypertension in offspring later in life. Obstet. Gynecol. 116, 858–864 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Sproul, D., Gilbert, N. & Bickmore, W. A. The role of chromatin structure in regulating the expression of clustered genes. Nat. Rev. Genet. 6, 775–781 (2005).

    Article  CAS  PubMed  Google Scholar 

  33. Pinheiro, T. V., Brunetto, S., Ramos, J. G., Bernardi, J. R. & Goldani, M. Z. Hypertensive disorders during pregnancy and health outcomes in the offspring: a systematic review. J. Dev. Orig. Health Dis. 7, 391–407 (2016).

    Article  CAS  PubMed  Google Scholar 

  34. Prins, J. R. et al. Smoking during pregnancy influences the maternal immune response in mice and humans. Am. J. Obstet. Gynecol. 207, 76.e1–76.e14 (2012).

    Article  CAS  Google Scholar 

  35. Yessoufou, A. & Moutairou, K. Maternal diabetes in pregnancy: early and long-term outcomes on the offspring and the concept of “metabolic memory”. Exp. Diabetes Res. 2011, 218598 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Cornelius, D. C. Preeclampsia: from inflammation to immunoregulation. Clin. Med. Insights Blood Disord. 11, 1179545X17752325 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Harmon, A. C. et al. The role of inflammation in the pathology of preeclampsia. Clin. Sci. 130, 409–419 (2016).

    Article  CAS  Google Scholar 

  38. De Luccia, T. P. B. et al. Unveiling the pathophysiology of gestational diabetes: Studies on local and peripheral immune cells. Scand. J. Immunol. 91, e12860 (2020).

    Article  PubMed  Google Scholar 

  39. Zatterale, F. et al. Chronic adipose tissue inflammation linking obesity to insulin resistance and type 2 diabetes. Front. Physiol. 10, 1607 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Gammill, H. S. & Nelson, J. L. Naturally acquired microchimerism. Int. J. Dev. Biol. 54, 531–543 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Lo, Y. M., Lau, T. K., Chan, L. Y., Leung, T. N. & Chang, A. M. Quantitative analysis of the bidirectional fetomaternal transfer of nucleated cells and plasma DNA. Clin. Chem. 46, 1301–1309 (2000).

    Article  CAS  PubMed  Google Scholar 

  42. Loubiere, L. S. et al. Maternal microchimerism in healthy adults in lymphocytes, monocyte/macrophages and NK cells. Lab. Invest. 86, 1185–1192 (2006).

    Article  CAS  PubMed  Google Scholar 

  43. Maloney, S. et al. Microchimerism of maternal origin persists into adult life. J. Clin. Invest. 104, 41–47 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Mold, J. E. et al. Maternal alloantigens promote the development of tolerogenic fetal regulatory T cells in utero. Science 322, 1562–1565 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Cupedo, T., Nagasawa, M., Weijer, K., Blom, B. & Spits, H. Development and activation of regulatory T cells in the human fetus. Eur. J. Immunol. 35, 383–390 (2005).

    Article  CAS  PubMed  Google Scholar 

  46. Michaelsson, J., Mold, J. E., McCune, J. M. & Nixon, D. F. Regulation of T cell responses in the developing human fetus. J. Immunol. 176, 5741–5748 (2006).

    Article  CAS  PubMed  Google Scholar 

  47. Havran, W. L. & Allison, J. P. Developmentally ordered appearance of thymocytes expressing different T-cell antigen receptors. Nature 335, 443–445 (1988).

    Article  CAS  PubMed  Google Scholar 

  48. Hardy, R. R., Hayakawa, K., Haaijman, J. & Herzenberg, L. A. B-cell subpopulations identifiable by two-color fluorescence analysis using a dual-laser FACS. Ann. NY Acad. Sci. 399, 112–121 (1982).

    Article  CAS  PubMed  Google Scholar 

  49. Mold, J. E. et al. Fetal and adult hematopoietic stem cells give rise to distinct T cell lineages in humans. Science 330, 1695–1699 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Krow-Lucal, E. R., Kim, C. C., Burt, T. D. & McCune, J. M. Distinct functional programming of human fetal and adult monocytes. Blood 123, 1897–1904 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Kumaravelu, P. et al. Quantitative developmental anatomy of definitive haematopoietic stem cells/long-term repopulating units (HSC/RUs): role of the aorta-gonad-mesonephros (AGM) region and the yolk sac in colonisation of the mouse embryonic liver. Development 129, 4891–4899 (2002).

    Article  CAS  PubMed  Google Scholar 

  52. Medvinsky, A. L., Gan, O. I., Semenova, M. L. & Samoylina, N. L. Development of day-8 colony-forming unit-spleen hematopoietic progenitors during early murine embryogenesis: spatial and temporal mapping. Blood 87, 557–566 (1996).

    Article  CAS  PubMed  Google Scholar 

  53. Godin, I. & Cumano, A. Of birds and mice: hematopoietic stem cell development. Int. J. Dev. Biol. 49, 251–257 (2005).

    Article  CAS  PubMed  Google Scholar 

  54. Ginhoux, F. & Guilliams, M. Tissue-resident macrophage ontogeny and homeostasis. Immunity 44, 439–449 (2016).

    Article  CAS  PubMed  Google Scholar 

  55. Park, J. E., Jardine, L., Gottgens, B., Teichmann, S. A. & Haniffa, M. Prenatal development of human immunity. Science 368, 600–603 (2020).

    Article  CAS  PubMed  Google Scholar 

  56. Amdi, C., Lynegaard, J. C., Thymann, T. & Williams, A. R. Intrauterine growth restriction in piglets alters blood cell counts and impairs cytokine responses in peripheral mononuclear cells 24 days post-partum. Sci. Rep. 10, 4683 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Che, L. et al. Flaxseed oil supplementation improves intestinal function and immunity, associated with altered intestinal microbiome and fatty acid profile in pigs with intrauterine growth retardation. Food Funct. 10, 8149–8160 (2019).

    Article  CAS  PubMed  Google Scholar 

  58. Jaeckle Santos, L. J. et al. Neutralizing Th2 inflammation in neonatal islets prevents β-cell failure in adult IUGR rats. Diabetes 63, 1672–1684 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Li, J. et al. Vγ9Vδ2-T lymphocytes have impaired antiviral function in small-for-gestational-age and preterm neonates. Cell Mol. Immunol. 10, 253–260 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Li, J. et al. Impaired NK cell antiviral cytokine response against influenza virus in small-for-gestational-age neonates. Cell Mol. Immunol. 10, 437–443 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Wirbelauer, J., Thomas, W., Rieger, L. & Speer, C. P. Intrauterine growth retardation in preterm infants</=32 weeks of gestation is associated with low white blood cell counts. Am. J. Perinatol. 27, 819–824 (2010).

    Article  PubMed  Google Scholar 

  62. Zhong, X. et al. Impairment of cellular immunity is associated with overexpression of heat shock protein 70 in neonatal pigs with intrauterine growth retardation. Cell Stress. Chaperones 17, 495–505 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Kelly, A. C. et al. RNA sequencing exposes adaptive and immune responses to intrauterine growth restriction in Fetal Sheep Islets. Endocrinology 158, 743–755 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Longo, S., Borghesi, A., Tzialla, C. & Stronati, M. IUGR and infections. Early Hum. Dev. 90 (Suppl. 1), S42–S44 (2014).

    Article  PubMed  Google Scholar 

  65. Hasselbalch, H., Jeppesen, D. L., Ersboll, A. K. & Nielsen, M. B. Thymus size in preterm infants evaluated by ultrasound. A preliminary report. Acta Radiol. 40, 37–40 (1999).

    CAS  PubMed  Google Scholar 

  66. Olearo, E. et al. Thymic volume in healthy, small for gestational age and growth restricted fetuses. Prenat. Diagn. 32, 662–667 (2012).

    Article  PubMed  Google Scholar 

  67. Geutskens, S. B., Otonkoski, T., Pulkkinen, M. A., Drexhage, H. A. & Leenen, P. J. Macrophages in the murine pancreas and their involvement in fetal endocrine development in vitro. J. Leukoc. Biol. 78, 845–852 (2005).

    Article  CAS  PubMed  Google Scholar 

  68. Calderon, B. et al. The pancreas anatomy conditions the origin and properties of resident macrophages. J. Exp. Med. 212, 1497–1512 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Calderon, B., Suri, A., Miller, M. J. & Unanue, E. R. Dendritic cells in islets of Langerhans constitutively present beta cell-derived peptides bound to their class II MHC molecules. Proc. Natl Acad. Sci. USA 105, 6121–6126 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Ferris, S. T. et al. A minor subset of Batf3-dependent antigen-presenting cells in islets of Langerhans is essential for the development of autoimmune diabetes. Immunity 41, 657–669 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Ferris, S. T. et al. The islet-resident macrophage is in an inflammatory state and senses microbial products in blood. J. Exp. Med. 214, 2369–2385 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Banaei-Bouchareb, L. et al. Insulin cell mass is altered in Csf1op/Csf1op macrophage-deficient mice. J. Leukoc. Biol. 76, 359–367 (2004).

    Article  CAS  PubMed  Google Scholar 

  73. Zinselmeyer, B. H. et al. The resident macrophages in murine pancreatic islets are constantly probing their local environment, capturing beta cell granules and blood particles. Diabetologia 61, 1374–1383 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Vomund, A. N. et al. Beta cells transfer vesicles containing insulin to phagocytes for presentation to T cells. Proc. Natl Acad. Sci. USA 112, E5496–E5502 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. German, M. et al. The insulin gene promoter. A simplified nomenclature. Diabetes 44, 1002–1004 (1995).

    Article  CAS  PubMed  Google Scholar 

  76. German, M. S., Moss, L. G., Wang, J. & Rutter, W. J. The insulin and islet amyloid polypeptide genes contain similar cell-specific promoter elements that bind identical beta-cell nuclear complexes. Mol. Cell Biol. 12, 1777–1788 (1992).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. de Koning, E. J. et al. Macrophages and pancreatic islet amyloidosis. Amyloid 5, 247–254 (1998).

    Article  PubMed  Google Scholar 

  78. de Koning, E. J., Bodkin, N. L., Hansen, B. C. & Clark, A. Diabetes mellitus in Macaca mulatta monkeys is characterised by islet amyloidosis and reduction in beta-cell population. Diabetologia 36, 378–384 (1993).

    Article  PubMed  Google Scholar 

  79. Masters, S. L. et al. Activation of the NLRP3 inflammasome by islet amyloid polypeptide provides a mechanism for enhanced IL-1beta in type 2 diabetes. Nat. Immunol. 11, 897–904 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Westwell-Roper, C. Y., Ehses, J. A. & Verchere, C. B. Resident macrophages mediate islet amyloid polypeptide-induced islet IL-1beta production and beta-cell dysfunction. Diabetes 63, 1698–1711 (2014).

    Article  CAS  PubMed  Google Scholar 

  81. Westermark, P., Andersson, A. & Westermark, G. T. Islet amyloid polypeptide, islet amyloid, and diabetes mellitus. Physiol. Rev. 91, 795–826 (2011).

    Article  CAS  PubMed  Google Scholar 

  82. Calderon, B., Carrero, J. A., Miller, M. J. & Unanue, E. R. Cellular and molecular events in the localization of diabetogenic T cells to islets of Langerhans. Proc. Natl Acad. Sci. USA 108, 1561–1566 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Calderon, B., Carrero, J. A., Miller, M. J. & Unanue, E. R. Entry of diabetogenic T cells into islets induces changes that lead to amplification of the cellular response. Proc. Natl Acad. Sci. USA 108, 1567–1572 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Ying, W. et al. Expansion of islet-resident macrophages leads to inflammation affecting beta cell proliferation and function in obesity. Cell Metab. 29, 457–474 (2019).

    Article  CAS  PubMed  Google Scholar 

  85. Ying, W., Fu, W., Lee, Y. S. & Olefsky, J. M. The role of macrophages in obesity-associated islet inflammation and beta-cell abnormalities. Nat. Rev. Endocrinol. 16, 81–90 (2020).

    Article  PubMed  Google Scholar 

  86. Anquetil, F. et al. Alpha cells, the main source of IL-1beta in human pancreas. J. Autoimmun. 81, 68–73 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Arnush, M., Scarim, A. L., Heitmeier, M. R., Kelly, C. B. & Corbett, J. A. Potential role of resident islet macrophage activation in the initiation of autoimmune diabetes. J. Immunol. 160, 2684–2691 (1998).

    Article  CAS  PubMed  Google Scholar 

  88. Boni-Schnetzler, M. et al. Increased interleukin (IL)-1beta messenger ribonucleic acid expression in beta -cells of individuals with type 2 diabetes and regulation of IL-1beta in human islets by glucose and autostimulation. J. Clin. Endocrinol. Metab. 93, 4065–4074 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  89. Maedler, K. et al. Glucose-induced beta cell production of IL-1beta contributes to glucotoxicity in human pancreatic islets. J. Clin. Invest. 110, 851–860 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Haversen, L., Danielsson, K. N., Fogelstrand, L. & Wiklund, O. Induction of proinflammatory cytokines by long-chain saturated fatty acids in human macrophages. Atherosclerosis 202, 382–393 (2009).

    Article  CAS  PubMed  Google Scholar 

  91. Arous, C., Ferreira, P. G., Dermitzakis, E. T. & Halban, P. A. Short term exposure of beta cells to low concentrations of interleukin-1beta improves insulin secretion through focal adhesion and actin remodeling and regulation of gene expression. J. Biol. Chem. 290, 14491 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Ribaux, P. et al. Induction of CXCL1 by extracellular matrix and autocrine enhancement by interleukin-1 in rat pancreatic beta-cells. Endocrinology 148, 5582–5590 (2007).

    Article  CAS  PubMed  Google Scholar 

  93. Ehses, J. A. et al. IL-1 antagonism reduces hyperglycemia and tissue inflammation in the type 2 diabetic GK rat. Proc. Natl Acad. Sci. USA 106, 13998–14003 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Zhao, G., Dharmadhikari, G., Maedler, K. & Meyer-Hermann, M. Possible role of interleukin-1beta in type 2 diabetes onset and implications for anti-inflammatory therapy strategies. PLoS Comput. Biol. 10, e1003798 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  95. Maedler, K. et al. Leptin modulates beta cell expression of IL-1 receptor antagonist and release of IL-1beta in human islets. Proc. Natl Acad. Sci. USA 101, 8138–8143 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Glas, R. et al. Purinergic P2X7 receptors regulate secretion of interleukin-1 receptor antagonist and beta cell function and survival. Diabetologia 52, 1579–1588 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Tabak, A. G. et al. Trajectories of glycaemia, insulin sensitivity, and insulin secretion before diagnosis of type 2 diabetes: an analysis from the Whitehall II study. Lancet 373, 2215–2221 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Herder, C. et al. Elevated levels of the anti-inflammatory interleukin-1 receptor antagonist precede the onset of type 2 diabetes: the Whitehall II study. Diabetes Care 32, 421–423 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  99. Equils, O. et al. Intra-uterine growth restriction downregulates the hepatic toll like receptor-4 expression and function. Clin. Dev. Immunol. 12, 59–66 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Roman, A. et al. Maternal magnesium supplementation reduces intrauterine growth restriction and suppresses inflammation in a rat model. Am. J. Obstet. Gynecol. 208, 383.e1–7 (2013).

    Article  CAS  Google Scholar 

  101. Sanvito, F. et al. TGF-beta 1 influences the relative development of the exocrine and endocrine pancreas in vitro. Development 120, 3451–3462 (1994).

    Article  CAS  PubMed  Google Scholar 

  102. Dichmann, D. S., Miller, C. P., Jensen, J., Scott Heller, R. & Serup, P. Expression and misexpression of members of the FGF and TGFbeta families of growth factors in the developing mouse pancreas. Dev. Dyn. 226, 663–674 (2003).

    Article  CAS  PubMed  Google Scholar 

  103. Miralles, F., Battelino, T., Czernichow, P. & Scharfmann, R. TGF-beta plays a key role in morphogenesis of the pancreatic islets of Langerhans by controlling the activity of the matrix metalloproteinase MMP-2. J. Cell Biol. 143, 827–836 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Smart, N. G. et al. Conditional expression of Smad7 in pancreatic beta cells disrupts TGF-beta signaling and induces reversible diabetes mellitus. PLoS Biol. 4, e39 (2006).

    Article  PubMed  PubMed Central  Google Scholar 

  105. Briana, D. D. et al. Fetal concentrations of the growth factors TGF-alpha and TGF-beta1 in relation to normal and restricted fetal growth at term. Cytokine 60, 157–161 (2012).

    Article  CAS  PubMed  Google Scholar 

  106. Lee, J. H. et al. Protection from beta-cell apoptosis by inhibition of TGF-beta/Smad3 signaling. Cell Death Dis. 11, 184 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Lee, Y. S., Wollam, J. & Olefsky, J. M. An integrated view of immunometabolism. Cell 172, 22–40 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Simmons, R. A., Templeton, L. J. & Gertz, S. J. Intrauterine growth retardation leads to the development of type 2 diabetes in the rat. Diabetes 50, 2279–2286 (2001).

    Article  CAS  PubMed  Google Scholar 

  109. Stoffers, D. A., Desai, B. M., DeLeon, D. D. & Simmons, R. A. Neonatal exendin-4 prevents the development of diabetes in the intrauterine growth retarded rat. Diabetes 52, 734–740 (2003).

    Article  CAS  PubMed  Google Scholar 

  110. Papathanassoglou, E. et al. Leptin receptor expression and signaling in lymphocytes: kinetics during lymphocyte activation, role in lymphocyte survival, and response to high fat diet in mice. J. Immunol. 176, 7745–7752 (2006).

    Article  CAS  PubMed  Google Scholar 

  111. Gabay, C., Dreyer, M., Pellegrinelli, N., Chicheportiche, R. & Meier, C. A. Leptin directly induces the secretion of interleukin 1 receptor antagonist in human monocytes. J. Clin. Endocrinol. Metab. 86, 783–791 (2001).

    CAS  PubMed  Google Scholar 

  112. Francisco, V. et al. Obesity, fat mass and immune system: role for leptin. Front. Physiol. 9, 640 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  113. Fernandez-Riejos, P., Goberna, R. & Sanchez-Margalet, V. Leptin promotes cell survival and activates Jurkat T lymphocytes by stimulation of mitogen-activated protein kinase. Clin. Exp. Immunol. 151, 505–518 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Amarilyo, G. et al. Leptin promotes lupus T-cell autoimmunity. Clin. Immunol. 149, 530–533 (2013).

    Article  CAS  PubMed  Google Scholar 

  115. La Cava, A. Leptin in inflammation and autoimmunity. Cytokine 98, 51–58 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  116. Cohen, S., Danzaki, K. & MacIver, N. J. Nutritional effects on T-cell immunometabolism. Eur. J. Immunol. 47, 225–235 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Pighetti, M. et al. Maternal serum and umbilical cord blood leptin concentrations with fetal growth restriction. Obstet. Gynecol. 102, 535–543 (2003).

    CAS  PubMed  Google Scholar 

  118. Varvarigou, A., Mantzoros, C. S. & Beratis, N. G. Cord blood leptin concentrations in relation to intrauterine growth. Clin. Endocrinol. 50, 177–183 (1999).

    Article  CAS  Google Scholar 

  119. Jaquet, D., Leger, J., Levy-Marchal, C., Oury, J. F. & Czernichow, P. Ontogeny of leptin in human fetuses and newborns: effect of intrauterine growth retardation on serum leptin concentrations. J. Clin. Endocrinol. Metab. 83, 1243–1246 (1998).

    Article  CAS  PubMed  Google Scholar 

  120. Valuniene, M. et al. Leptin levels at birth and in early postnatal life in small- and appropriate-for-gestational-age infants. Medicina 43, 784–791 (2007).

    Article  PubMed  Google Scholar 

  121. Cetin, I. et al. Fetal plasma leptin concentrations: relationship with different intrauterine growth patterns from 19 weeks to term. Pediatr. Res. 48, 646–651 (2000).

    Article  CAS  PubMed  Google Scholar 

  122. Martinez-Cordero, C., Amador-Licona, N., Guizar-Mendoza, J. M., Hernandez-Mendez, J. & Ruelas-Orozco, G. Body fat at birth and cord blood levels of insulin, adiponectin, leptin, and insulin-like growth factor-I in small-for-gestational-age infants. Arch. Med. Res. 37, 490–494 (2006).

    Article  CAS  PubMed  Google Scholar 

  123. Jaquet, D., Leger, J., Tabone, M. D., Czernichow, P. & Levy-Marchal, C. High serum leptin concentrations during catch-up growth of children born with intrauterine growth retardation. J. Clin. Endocrinol. Metab. 84, 1949–1953 (1999).

    CAS  PubMed  Google Scholar 

  124. Yajnik, C. S. et al. Adiposity and hyperinsulinemia in Indians are present at birth. J. Clin. Endocrinol. Metab. 87, 5575–5580 (2002).

    Article  CAS  PubMed  Google Scholar 

  125. Phillips, D. I. et al. Size at birth and plasma leptin concentrations in adult life. Int. J. Obes. Relat. Metab. Disord. 23, 1025–1029 (1999).

    Article  CAS  PubMed  Google Scholar 

  126. Ohashi, K. et al. Adiponectin promotes macrophage polarization toward an anti-inflammatory phenotype. J. Biol. Chem. 285, 6153–6160 (2010).

    Article  CAS  PubMed  Google Scholar 

  127. Luo, Y. & Liu, M. Adiponectin: a versatile player of innate immunity. J. Mol. Cell Biol. 8, 120–128 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Sivan, E. et al. Adiponectin in human cord blood: relation to fetal birth weight and gender. J. Clin. Endocrinol. Metab. 88, 5656–5660 (2003).

    Article  CAS  PubMed  Google Scholar 

  129. Visentin, S. et al. Adiponectin levels are reduced while markers of systemic inflammation and aortic remodelling are increased in intrauterine growth restricted mother-child couple. Biomed. Res. Int. 2014, 401595 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  130. Kyriakakou, M. et al. Leptin and adiponectin concentrations in intrauterine growth restricted and appropriate for gestational age fetuses, neonates, and their mothers. Eur. J. Endocrinol. 158, 343–348 (2008).

    Article  CAS  PubMed  Google Scholar 

  131. Pinney, S. E., Jaeckle Santos, L. J., Han, Y., Stoffers, D. A. & Simmons, R. A. Exendin-4 increases histone acetylase activity and reverses epigenetic modifications that silence Pdx1 in the intrauterine growth retarded rat. Diabetologia 54, 2606–2614 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Tremblay, A. J., Lamarche, B., Deacon, C. F., Weisnagel, S. J. & Couture, P. Effects of sitagliptin therapy on markers of low-grade inflammation and cell adhesion molecules in patients with type 2 diabetes. Metabolism 63, 1141–1148 (2014).

    Article  CAS  PubMed  Google Scholar 

  133. Ahern, T. et al. Glucagon-like peptide-1 analogue therapy for psoriasis patients with obesity and type 2 diabetes: a prospective cohort study. J. Eur. Acad. Dermatol. Venereol. 27, 1440–1443 (2013).

    Article  CAS  PubMed  Google Scholar 

  134. Marcucci, F., Romeo, E., Caserta, C. A., Rumio, C. & Lefoulon, F. Context-dependent pharmacological effects of metformin on the immune system. Trends Pharmacol. Sci. 41, 162–171 (2020).

    Article  CAS  PubMed  Google Scholar 

  135. Vasamsetti, S. B. et al. Metformin inhibits monocyte-to-macrophage differentiation via AMPK-mediated inhibition of STAT3 activation: potential role in atherosclerosis. Diabetes 64, 2028–2041 (2015).

    Article  CAS  PubMed  Google Scholar 

  136. Qing, L. et al. Metformin induces the M2 macrophage polarization to accelerate the wound healing via regulating AMPK/mTOR/NLRP3 inflammasome singling pathway. Am. J. Transl Res. 11, 655–668 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Tsoyi, K. et al. Metformin inhibits HMGB1 release in LPS-treated RAW 264.7 cells and increases survival rate of endotoxaemic mice. Br. J. Pharmacol. 162, 1498–1508 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Jing, Y. et al. Metformin improves obesity-associated inflammation by altering macrophages polarization. Mol. Cell Endocrinol. 461, 256–264 (2018).

    Article  CAS  PubMed  Google Scholar 

  139. Yang, Q. et al. Metformin ameliorates the progression of atherosclerosis via suppressing macrophage infiltration and inflammatory responses in rabbits. Life Sci. 198, 56–64 (2018).

    Article  CAS  PubMed  Google Scholar 

  140. Dandona, P. et al. Increased plasma concentration of macrophage migration inhibitory factor (MIF) and MIF mRNA in mononuclear cells in the obese and the suppressive action of metformin. J. Clin. Endocrinol. Metab. 89, 5043–5047 (2004).

    Article  CAS  PubMed  Google Scholar 

  141. Xiao, H. et al. Metformin is a novel suppressor for transforming growth factor (TGF)-beta1. Sci. Rep. 6, 28597 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. LeBrasseur, N. K. et al. Thiazolidinediones can rapidly activate AMP-activated protein kinase in mammalian tissues. Am. J. Physiol. Endocrinol. Metab. 291, E175–E181 (2006).

    Article  CAS  PubMed  Google Scholar 

  143. Ceriello, A. Thiazolidinediones as anti-inflammatory and anti-atherogenic agents. Diabetes Metab. Res. Rev. 24, 14–26 (2008).

    Article  CAS  PubMed  Google Scholar 

  144. Shimizu, H. et al. Pioglitazone increases circulating adiponectin levels and subsequently reduces TNF-alpha levels in type 2 diabetic patients: a randomized study. Diabet. Med. 23, 253–257 (2006).

    Article  CAS  PubMed  Google Scholar 

  145. Dai, Y., Wang, X., Ding, Z., Dai, D. & Mehta, J. L. DPP-4 inhibitors repress foam cell formation by inhibiting scavenger receptors through protein kinase C pathway. Acta Diabetol. 51, 471–478 (2014).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors acknowledge the support of the National Institutes of Health grant #DK114054 (R.A.S.) and ES01985 (T.N.G.).

Author information

Authors and Affiliations

Authors

Contributions

T.N.G. and R.A.S. researched data for the article, contributed to discussion of its content, wrote the article, and reviewed and edited the manuscript before submission.

Corresponding author

Correspondence to Rebecca A. Simmons.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Endocrinology thanks S. Limesand and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Glossary

Small for gestational age

(SGA). Birth weight that is below the 10th percentile.

Uteroplacental insufficiency

A complication of pregnancy when the placenta is unable to deliver an adequate supply of nutrients and oxygen to the fetus.

Parabiosis studies

A laboratory technique to study physiology whereby two living organisms are joined together surgically to develop a single, shared physiological system.

Glucose disposal

Storage of glucose as glycogen in tissues.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Golden, T.N., Simmons, R.A. Immune dysfunction in developmental programming of type 2 diabetes mellitus. Nat Rev Endocrinol 17, 235–245 (2021). https://doi.org/10.1038/s41574-020-00464-z

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41574-020-00464-z

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing