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:

Hypothalamic innate immune reaction in obesity

Key Points

  • Hypothalamic metabolic sensing requires an interactive network of neurons, glial cells and vasculature to enable appropriate integration of complex metabolic feedback signals

  • Exposure to hypercaloric environments induces inflammatory-like responses not only in peripheral tissues but also in the central nervous system, especially in the hypothalamus

  • The term 'hypothalamic inflammation' is being used more and more frequently to describe a complex of hypothalamic processes that occurs in response to hypercaloric diets

  • The phenomenon of hypothalamic inflammation in obesity is more similar to an innate immune reaction than to conventionally defined inflammation

  • To address the hypothalamic innate immune reaction therapeutically, cell-specific strategies should be developed that enable prevention of the adverse effects of systemic treatment approaches

Abstract

Findings from rodent and human studies show that the presence of inflammatory factors is positively correlated with obesity and the metabolic syndrome. Obesity-associated inflammatory responses take place not only in the periphery but also in the brain. The hypothalamus contains a range of resident glial cells including microglia, macrophages and astrocytes, which are embedded in highly heterogenic groups of neurons that control metabolic homeostasis. This complex neural–glia network can receive information directly from blood-borne factors, positioning it as a metabolic sensor. Following hypercaloric challenge, mediobasal hypothalamic microglia and astrocytes enter a reactive state, which persists during diet-induced obesity. In established mouse models of diet-induced obesity, the hypothalamic vasculature displays angiogenic alterations. Moreover, proopiomelanocortin neurons, which regulate food intake and energy expenditure, are impaired in the arcuate nucleus, where there is an increase in local inflammatory signals. The sum total of these events is a hypothalamic innate immune reactivity, which includes temporal and spatial changes to each cell population. Although the exact role of each participant of the neural–glial–vascular network is still under exploration, therapeutic targets for treating obesity should probably be linked to individual cell types and their specific signalling pathways to address each dysfunction with cell-selective compounds.

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

Figure 1: The cytoarchitecture of hypothalamic NPY and POMC neurons, microglia, astrocytes and vasculature.
Figure 2: The diversity of hypothalamic microglia and macrophages visualized in CX3Cr1-GFP mice.
Figure 3: Morphological comparison of microglial reactivity in diet-induced obesity as visualized by AIF-1-ir.
Figure 4: The distribution pattern of the GFP+ astrocytes (green) and GFAP-ir astrocytes (red) in hGFAP-GFP transgenic mouse hypothalamus.
Figure 5: The diverse cell populations in the local microenvironment of the mediobasal hypothalamus and their respective transitions from basal conditions to reactive stages in the metabolic syndrome.

Similar content being viewed by others

References

  1. Zhang, Y. et al. Positional cloning of the mouse obese gene and its human homologue. Nature 372, 425–432 (1994).

    Article  CAS  PubMed  Google Scholar 

  2. Williams, K. W. & Elmquist, J. K. From neuroanatomy to behavior: central integration of peripheral signals regulating feeding behavior. Nat. Neurosci. 15, 1350–1355 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Begg, D. P. & Woods, S. C. The endocrinology of food intake. Nat. Rev. Endocrinol. 9, 584–597 (2013).

    Article  CAS  PubMed  Google Scholar 

  4. Furuhashi, M. et al. Adipocyte/macrophage fatty acid-binding proteins contribute to metabolic deterioration through actions in both macrophages and adipocytes in mice. J. Clin. Invest. 118, 2640–2650 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Mathis, D. Immunological goings-on in visceral adipose tissue. Cell Metab. 17, 851–859 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Zhang, X. et al. Hypothalamic IKKβ/NF-κB and ER stress link overnutrition to energy imbalance and obesity. Cell 135, 61–73 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. De Souza, C. T. et al. Consumption of a fat-rich diet activates a proinflammatory response and induces insulin resistance in the hypothalamus. Endocrinology 146, 4192–4199 (2005).

    Article  CAS  PubMed  Google Scholar 

  8. Yi, C. X., Zeltser, L. & Tschöp, M. H. Metabolic Syndrome ePoster—Brain & Neuron. Nat. Med. [online], (2011).

  9. Yi, C. X. & Tschöp, M. H. Brain-gut-adipose-tissue communication pathways at a glance. Dis. Model. Mech. 5, 583–587 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Yi, C. X., Scherer, T. & Tschöp, M. H. Cajal revisited: does the VMH make us fat? Nat. Neurosci. 14, 806–808 (2011).

    Article  CAS  PubMed  Google Scholar 

  11. Kim, K. W. et al. CNS-specific ablation of steroidogenic factor 1 results in impaired female reproductive function. Mol. Endocrinol. 24, 1240–1250 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Xu, B. et al. Brain-derived neurotrophic factor regulates energy balance downstream of melanocortin-4 receptor. Nat. Neurosci. 6, 736–742 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Obici, S., Zhang, B. B., Karkanias, G. & Rossetti, L. Hypothalamic insulin signaling is required for inhibition of glucose production. Nat. Med. 8, 1376–1382 (2002).

    Article  CAS  PubMed  Google Scholar 

  14. Bruinstroop, E. et al. The autonomic nervous system regulates postprandial hepatic lipid metabolism. Am. J. Physiol. Endocrinol. Metab. 304, E1089–E1096 (2013).

    Article  CAS  PubMed  Google Scholar 

  15. Lam, T. K. et al. Hypothalamic sensing of circulating fatty acids is required for glucose homeostasis. Nat. Med. 11, 320–327 (2005).

    Article  CAS  PubMed  Google Scholar 

  16. Cottrell, G. T. & Ferguson, A. V. Sensory circumventricular organs: central roles in integrated autonomic regulation. Regul. Pept. 117, 11–23 (2004).

    Article  CAS  PubMed  Google Scholar 

  17. Milanski, M. et al. Saturated fatty acids produce an inflammatory response predominantly through the activation of TLR4 signaling in hypothalamus: implications for the pathogenesis of obesity. J. Neurosci. 29, 359–370 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Thaler, J. P. et al. Obesity is associated with hypothalamic injury in rodents and humans. J. Clin. Invest. 122, 153–162 (2012).

    Article  PubMed  Google Scholar 

  19. Yi, C. X. et al. High calorie diet triggers hypothalamic angiopathy. Mol. Metab. 1, 95–100 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Moraes, J. C. et al. High-fat diet induces apoptosis of hypothalamic neurons. PLoS ONE 4, e5045 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Li, J., Tang, Y. & Cai, D. IKKβ/NF-κB disrupts adult hypothalamic neural stem cells to mediate a neurodegenerative mechanism of dietary obesity and pre-diabetes. Nat. Cell Biol. 14, 999–1012 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Heneka, M. T., Kummer, M. P. & Latz, E. Innate immune activation in neurodegenerative disease. Nat. Rev. Immunol. 14, 463–477 (2014).

    Article  CAS  PubMed  Google Scholar 

  23. Heppner, F., Ransohoff, R. & Becher, B. Immune attack! The role of inflammation in Alzheimer's disease. Nat. Rev. Neurosci. (in press).

  24. Ginhoux, F. et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 330, 841–845 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Kierdorf, K. et al. Microglia emerge from erythromyeloid precursors via Pu.1- and Irf8-dependent pathways. Nat. Neurosci. 16, 273–280 (2013).

    Article  CAS  PubMed  Google Scholar 

  26. Alliot, F., Godin, I. & Pessac, B. Microglia derive from progenitors, originating from the yolk sac, and which proliferate in the brain. Brain Res. Dev. Brain Res. 117, 145–152 (1999).

    Article  CAS  PubMed  Google Scholar 

  27. Perry, V. H., Hume, D. A. & Gordon, S. Immunohistochemical localization of macrophages and microglia in the adult and developing mouse brain. Neuroscience 15, 313–326 (1985).

    Article  CAS  PubMed  Google Scholar 

  28. Massberg, S. et al. Immunosurveillance by hematopoietic progenitor cells trafficking through blood, lymph, and peripheral tissues. Cell 131, 994–1008 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Mildner, A. et al. Microglia in the adult brain arise from Ly-6ChiCCR2+ monocytes only under defined host conditions. Nat. Neurosci. 10, 1544–1553 (2007).

    Article  CAS  PubMed  Google Scholar 

  30. Varvel, N. H. et al. Microglial repopulation model reveals a robust homeostatic process for replacing CNS myeloid cells. Proc. Natl Acad. Sci. USA 109, 18150–18155 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Li, Y., Du, X. F., Liu, C. S., Wen, Z. L. & Du, J. L. Reciprocal regulation between resting microglial dynamics and neuronal activity in vivo. Dev. Cell 23, 1189–1202 (2012).

    Article  CAS  PubMed  Google Scholar 

  32. Prinz, M. & Priller, J. Microglia and brain macrophages in the molecular age: from origin to neuropsychiatric disease. Nat. Rev. Neurosci. 15, 300–312 (2014).

    Article  CAS  PubMed  Google Scholar 

  33. Kettenmann, H., Hanisch, U. K., Noda, M. & Verkhratsky, A. Physiology of microglia. Physiol. Rev. 91, 461–553 (2011).

    Article  CAS  PubMed  Google Scholar 

  34. Daikoku, S., Kotsu, T. & Hashimoto, M. Electron microscopic observations on the development of the median eminence in perinatal rats. Z. Anat. Entwicklungsgesch. 134, 311–327 (1971).

    Article  CAS  PubMed  Google Scholar 

  35. Bitsch, P. & Schiebler, T. H. Postnatal development of the median eminence in the rat [German]. Z. Mikrosk. Anat. Forsch. 93, 1–20 (1979).

    CAS  PubMed  Google Scholar 

  36. Pow, D. V., Perry, V. H., Morris, J. F. & Gordon, S. Microglia in the neurohypophysis associate with and endocytose terminal portions of neurosecretory neurons. Neuroscience 33, 567–578 (1989).

    Article  CAS  PubMed  Google Scholar 

  37. Jung, S. et al. Analysis of fractalkine receptor CX3CR1 function by targeted deletion and green fluorescent protein reporter gene insertion. Mol. Cell. Biol. 20, 4106–4114 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Hawkes, C. A. & McLaurin, J. Selective targeting of perivascular macrophages for clearance of β-amyloid in cerebral amyloid angiopathy. Proc. Natl Acad. Sci. USA 106, 1261–1266 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Grossmann, R. et al. Juxtavascular microglia migrate along brain microvessels following activation during early postnatal development. Glia 37, 229–240 (2002).

    Article  PubMed  Google Scholar 

  40. Davalos, D. et al. Fibrinogen-induced perivascular microglial clustering is required for the development of axonal damage in neuroinflammation. Nat. Commun. 3, 1227 (2012).

    Article  CAS  PubMed  Google Scholar 

  41. Neumann, H., Kotter, M. R. & Franklin, R. J. Debris clearance by microglia: an essential link between degeneration and regeneration. Brain 132, 288–295 (2009).

    Article  CAS  PubMed  Google Scholar 

  42. Douglas, S. D. & Musson, R. A. Phagocytic defects--monocytes/macrophages. Clin. Immunol. Immunopathol. 40, 62–68 (1986).

    Article  CAS  PubMed  Google Scholar 

  43. Vedeler, C. et al. Fc receptor for IgG (FcR) on rat microglia. J. Neuroimmunol. 49, 19–24 (1994).

    Article  CAS  PubMed  Google Scholar 

  44. Quan, Y., Moller, T. & Weinstein, J. R. Regulation of Fcγ receptors and immunoglobulin G-mediated phagocytosis in mouse microglia. Neurosci. Lett. 464, 29–33 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Webster, S. D., Park, M., Fonseca, M. I. & Tenner, A. J. Structural and functional evidence for microglial expression of C1qRP, the C1q receptor that enhances phagocytosis. J. Leukoc. Biol. 67, 109–116 (2000).

    Article  CAS  PubMed  Google Scholar 

  46. Bard, F. et al. Peripherally administered antibodies against amyloid β-peptide enter the central nervous system and reduce pathology in a mouse model of Alzheimer disease. Nat. Med. 6, 916–919 (2000).

    Article  CAS  PubMed  Google Scholar 

  47. Citron, M. Alzheimer's disease: strategies for disease modification. Nat. Rev. Drug Discov. 9, 387–398 (2010).

    Article  CAS  PubMed  Google Scholar 

  48. Fishman, P. S. & Savitt, J. M. Selective localization by neuroglia of immunoglobulin G in normal mice. J. Neuropathol. Exp. Neurol. 48, 212–220 (1989).

    Article  CAS  PubMed  Google Scholar 

  49. Yi, C. X., Tschöp, M. H., Woods, S. C. & Hofmann, S. M. High-fat-diet exposure induces IgG accumulation in hypothalamic microglia. Dis. Model. Mech. 5, 686–690 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Ferreira, R. et al. Neuropeptide Y inhibits interleukin-1β-induced phagocytosis by microglial cells. J. Neuroinflammation 8, 169 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Krabbe, G. et al. Functional impairment of microglia coincides with β-amyloid deposition in mice with Alzheimer-like pathology. PLoS ONE 8, e60921 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Wake, H., Moorhouse, A. J., Jinno, S., Kohsaka, S. & Nabekura, J. Resting microglia directly monitor the functional state of synapses in vivo and determine the fate of ischemic terminals. J. Neurosci. 29, 3974–3980 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Paolicelli, R. C. et al. Synaptic pruning by microglia is necessary for normal brain development. Science 333, 1456–1458 (2011).

    Article  CAS  PubMed  Google Scholar 

  54. Zhan, Y. et al. Deficient neuron-microglia signaling results in impaired functional brain connectivity and social behavior. Nat. Neurosci. 17, 400–406 (2014).

    Article  CAS  PubMed  Google Scholar 

  55. Bouret, S. G., Draper, S. J. & Simerly, R. B. Formation of projection pathways from the arcuate nucleus of the hypothalamus to hypothalamic regions implicated in the neural control of feeding behavior in mice. J. Neurosci. 24, 2797–2805 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Loffreda, S. et al. Leptin regulates proinflammatory immune responses. FASEB J. 12, 57–65 (1998).

    Article  CAS  PubMed  Google Scholar 

  57. Schafer, D. P. et al. Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner. Neuron 74, 691–705 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Nimmerjahn, A., Kirchhoff, F. & Helmchen, F. Resting microglial cells are highly dynamic surveillants of brain parenchyma in vivo. Science 308, 1314–1318 (2005).

    Article  CAS  PubMed  Google Scholar 

  59. Schaeffer, M. et al. Rapid sensing of circulating ghrelin by hypothalamic appetite-modifying neurons. Proc. Natl Acad. Sci. USA 110, 1512–1517 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Gao, Y. et al. Hormones and diet, but not body weight, control hypothalamic microglial activity. Glia 62, 17–25 (2014).

    Article  PubMed  Google Scholar 

  61. Lafrance, V., Inoue, W., Kan, B. & Luheshi, G. N. Leptin modulates cell morphology and cytokine release in microglia. Brain Behav. Immun. 24, 358–365 (2010).

    Article  CAS  PubMed  Google Scholar 

  62. Kleinridders, A. et al. MyD88 signaling in the CNS is required for development of fatty acid-induced leptin resistance and diet-induced obesity. Cell Metab. 10, 249–259 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Choi, S. J., Kim, F., Schwartz, M. W. & Wisse, B. E. Cultured hypothalamic neurons are resistant to inflammation and insulin resistance induced by saturated fatty acids. Am. J. Physiol. Endocrinol. Metab. 298, E1122–E1130 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Douard, V. & Ferraris, R. P. Regulation of the fructose transporter GLUT5 in health and disease. Am. J. Physiol. Endocrinol. Metab. 295, E227–E237 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Maher, F., Vannucci, S. J. & Simpson, I. A. Glucose transporter proteins in brain. FASEB J. 8, 1003–1011 (1994).

    Article  CAS  PubMed  Google Scholar 

  66. Vannucci, S. J., Maher, F. & Simpson, I. A. Glucose transporter proteins in brain: delivery of glucose to neurons and glia. Glia 21, 2–21 (1997).

    Article  CAS  PubMed  Google Scholar 

  67. Vom Berg, J. et al. Inhibition of IL-12/IL-23 signaling reduces Alzheimer's disease-like pathology and cognitive decline. Nat. Med. 18, 1812–1819 (2012).

    Article  CAS  PubMed  Google Scholar 

  68. Zhang, B. et al. Integrated systems approach identifies genetic nodes and networks in late-onset Alzheimer's disease. Cell 153, 707–720 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Streit, W. J., Braak, H., Xue, Q. S. & Bechmann, I. Dystrophic (senescent) rather than activated microglial cells are associated with tau pathology and likely precede neurodegeneration in Alzheimer's disease. Acta Neuropathol. 118, 475–485 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Parkhurst, C. N. et al. Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor. Cell 155, 1596–1609 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. De Filippo, C. et al. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc. Natl Acad. Sci. USA 107, 14691–14696 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  72. Zhao, L. The gut microbiota and obesity: from correlation to causality. Nat. Rev. Microbiol. 11, 639–647 (2013).

    Article  CAS  PubMed  Google Scholar 

  73. Inoue, K. Microglial activation by purines and pyrimidines. Glia 40, 156–163 (2002).

    Article  PubMed  Google Scholar 

  74. James, G. & Butt, A. M. P2Y and P2X purinoceptor mediated Ca2+ signalling in glial cell pathology in the central nervous system. Eur. J. Pharmacol. 447, 247–260 (2002).

    Article  CAS  PubMed  Google Scholar 

  75. Samuels, S. E., Lipitz, J. B., Wang, J., Dahl, G. & Muller, K. J. Arachidonic acid closes innexin/pannexin channels and thereby inhibits microglia cell movement to a nerve injury. Dev. Neurobiol. 73, 621–631 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Bermudez-Silva, F. J., Cardinal, P. & Cota, D. The role of the endocannabinoid system in the neuroendocrine regulation of energy balance. J. Psychopharmacol. 26, 114–124 (2012).

    Article  CAS  PubMed  Google Scholar 

  77. Cardona, A. E. et al. Control of microglial neurotoxicity by the fractalkine receptor. Nat. Neurosci. 9, 917–924 (2006).

    Article  CAS  PubMed  Google Scholar 

  78. Morari, J. et al. Fractalkine (CX3CL1) is involved in the early activation of hypothalamic inflammation in experimental obesity. Diabetes 63, 3770–3784 (2014).

    Article  CAS  PubMed  Google Scholar 

  79. Ruchaya, P. J., Antunes, V. R., Paton, J. F., Murphy, D. & Yao, S. T. The cardiovascular actions of fractalkine/CX3CL1 in the hypothalamic paraventricular nucleus are attenuated in rats with heart failure. Exp. Physiol. 99, 111–122 (2014).

    Article  CAS  PubMed  Google Scholar 

  80. Buckman, L. B. et al. Obesity induced by a high-fat diet is associated with increased immune cell entry into the central nervous system. Brain Behav. Immun. 35, 33–42 (2014).

    Article  CAS  PubMed  Google Scholar 

  81. Mildner, A. et al. Distinct and non-redundant roles of microglia and myeloid subsets in mouse models of Alzheimer's disease. J. Neurosci. 31, 11159–11171 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Kierdorf, K., Katzmarski, N., Haas, C. A. & Prinz, M. Bone marrow cell recruitment to the brain in the absence of irradiation or parabiosis bias. PLoS ONE 8, e58544 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Gordon, S. Alternative activation of macrophages. Nat. Rev. Immunol. 3, 23–35 (2003).

    Article  CAS  PubMed  Google Scholar 

  84. Murray, P. J. & Wynn, T. A. Protective and pathogenic functions of macrophage subsets. Nat. Rev. Immunol. 11, 723–737 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Michelucci, A., Heurtaux, T., Grandbarbe, L., Morga, E. & Heuschling, P. Characterization of the microglial phenotype under specific pro-inflammatory and anti-inflammatory conditions: effects of oligomeric and fibrillar amyloid-β. J. Neuroimmunol. 210, 3–12 (2009).

    Article  CAS  PubMed  Google Scholar 

  86. Ponomarev, E. D., Maresz, K., Tan, Y. & Dittel, B. N. CNS-derived interleukin-4 is essential for the regulation of autoimmune inflammation and induces a state of alternative activation in microglial cells. J. Neurosci. 27, 10714–10721 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Prinz, M., Priller, J., Sisodia, S. S. & Ransohoff, R. M. Heterogeneity of CNS myeloid cells and their roles in neurodegeneration. Nat. Neurosci. 14, 1227–1235 (2011).

    Article  CAS  PubMed  Google Scholar 

  88. Town, T., Nikolic, V. & Tan, J. The microglial “activation” continuum: from innate to adaptive responses. J. Neuroinflammation 2, 24 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Murray, P. J. et al. Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity 41, 14–20 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Wieghofer, P., Knobeloch, K. P. & Prinz, M. Genetic targeting of microglia. Glia 63, 1–22 (2014).

    Article  PubMed  Google Scholar 

  91. Goldmann, T. et al. A new type of microglia gene targeting shows TAK1 to be pivotal in CNS autoimmune inflammation. Nat. Neurosci. 16, 1618–1626 (2013).

    Article  CAS  PubMed  Google Scholar 

  92. Heppner, F. L. et al. Experimental autoimmune encephalomyelitis repressed by microglial paralysis. Nat. Med. 11, 146–152 (2005).

    Article  CAS  PubMed  Google Scholar 

  93. Grathwohl, S. A. et al. Formation and maintenance of Alzheimer's disease beta-amyloid plaques in the absence of microglia. Nat. Neurosci. 12, 1361–1363 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Yi, C. X., Habegger, K. M., Chowen, J. A., Stern, J. & Tschöp, M. H. A role for astrocytes in the central control of metabolism. Neuroendocrinology 93, 143–149 (2011).

    Article  CAS  PubMed  Google Scholar 

  95. Broer, S. et al. Comparison of lactate transport in astroglial cells and monocarboxylate transporter 1 (MCT 1) expressing Xenopus laevis oocytes. Expression of two different monocarboxylate transporters in astroglial cells and neurons. J. Biol. Chem. 272, 30096–30102 (1997).

    Article  CAS  PubMed  Google Scholar 

  96. Le Foll, C., Dunn-Meynell, A. A., Miziorko, H. M. & Levin, B. E. Regulation of hypothalamic neuronal sensing and food intake by ketone bodies and fatty acids. Diabetes 63, 1259–1269 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Dixit, V. D. et al. Ghrelin and the growth hormone secretagogue receptor constitute a novel autocrine pathway in astrocytoma motility. J. Biol. Chem. 281, 16681–16690 (2006).

    Article  CAS  PubMed  Google Scholar 

  98. Hsuchou, H. et al. Obesity induces functional astrocytic leptin receptors in hypothalamus. Brain 132, 889–902 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  99. Heni, M. et al. Insulin promotes glycogen storage and cell proliferation in primary human astrocytes. PLoS ONE 6, e21594 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Kim, J. G. et al. Leptin signaling in astrocytes regulates hypothalamic neuronal circuits and feeding. Nat. Neurosci. 17, 908–910 (2014).

    Article  CAS  PubMed  Google Scholar 

  101. Diano, S., Leonard, J. L., Meli, R., Esposito, E. & Schiavo, L. Hypothalamic type II iodothyronine deiodinase: a light and electron microscopic study. Brain Res. 976, 130–134 (2003).

    Article  CAS  PubMed  Google Scholar 

  102. Herwig, A., Ross, A. W., Nilaweera, K. N., Morgan, P. J. & Barrett, P. Hypothalamic thyroid hormone in energy balance regulation. Obes. Facts 1, 71–79 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Courtin, F. et al. Thyroid hormone deiodinases in the central and peripheral nervous system. Thyroid 15, 931–942 (2005).

    Article  CAS  PubMed  Google Scholar 

  104. Kamphuis, W. et al. GFAP isoforms in adult mouse brain with a focus on neurogenic astrocytes and reactive astrogliosis in mouse models of Alzheimer disease. PLoS ONE 7, e42823 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Horvath, T. L. et al. Synaptic input organization of the melanocortin system predicts diet-induced hypothalamic reactive gliosis and obesity. Proc. Natl Acad. Sci. USA 107, 14875–14880 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  106. Frederich, R. C. et al. Leptin levels reflect body lipid content in mice: evidence for diet-induced resistance to leptin action. Nat. Med. 1, 1311–1314 (1995).

    Article  CAS  PubMed  Google Scholar 

  107. Campbell, I. L. et al. Neurologic disease induced in transgenic mice by cerebral overexpression of interleukin 6. Proc. Natl Acad. Sci. USA 90, 10061–10065 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Haugh, R. M. & Markesbery, W. R. Hypothalamic astrocytoma. Syndrome of hyperphagia, obesity, and disturbances of behavior and endocrine and autonomic function. Arch. Neurol. 40, 560–563 (1983).

    Article  CAS  PubMed  Google Scholar 

  109. Emsley, J. G. & Macklis, J. D. Astroglial heterogeneity closely reflects the neuronal-defined anatomy of the adult murine CNS. Neuron Glia Biol. 2, 175–186 (2006).

    Article  PubMed  PubMed Central  Google Scholar 

  110. Langlet, F. et al. Tanycytic VEGF-A boosts blood-hypothalamus barrier plasticity and access of metabolic signals to the arcuate nucleus in response to fasting. Cell Metab. 17, 607–617 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Milanski, M. et al. Inhibition of hypothalamic inflammation reverses diet-induced insulin resistance in the liver. Diabetes 61, 1455–1462 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Hotamisligil, G. S., Shargill, N. S. & Spiegelman, B. M. Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science 259, 87–91 (1993).

    Article  CAS  PubMed  Google Scholar 

  113. Ventre, J. et al. Targeted disruption of the tumor necrosis factor-alpha gene: metabolic consequences in obese and nonobese mice. Diabetes 46, 1526–1531 (1997).

    Article  CAS  PubMed  Google Scholar 

  114. Pamir, N., McMillen, T. S., Kaiyala, K. J., Schwartz, M. W. & LeBoeuf, R. C. Receptors for tumor necrosis factor-α play a protective role against obesity and alter adipose tissue macrophage status. Endocrinology 150, 4124–4134 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Aggarwal, B. B. Signalling pathways of the TNF superfamily: a double-edged sword. Nat. Rev. Immunol. 3, 745–756 (2003).

    Article  CAS  PubMed  Google Scholar 

  116. Baker, R. G., Hayden, M. S. & Ghosh, S. NF-κB, inflammation, and metabolic disease. Cell Metab. 13, 11–22 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Perkins, N. D. Integrating cell-signalling pathways with NF-κB and IKK function. Nat. Rev. Mol. Cell Biol. 8, 49–62 (2007).

    Article  CAS  PubMed  Google Scholar 

  118. Meng, Q. & Cai, D. Defective hypothalamic autophagy directs the central pathogenesis of obesity via the IκB kinase β (IKKβ)/NF-κB pathway. J. Biol. Chem. 286, 32324–32332 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Purkayastha, S., Zhang, G. & Cai, D. Uncoupling the mechanisms of obesity and hypertension by targeting hypothalamic IKK-β and NF-κB. Nat. Med. 17, 883–887 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Koulich, E., Nguyen, T., Johnson, K., Giardina, C. & D'Mello, S. NF-κB is involved in the survival of cerebellar granule neurons: association of Iκβ phosphorylation with cell survival. J. Neurochem. 76, 1188–1198 (2001).

    Article  CAS  PubMed  Google Scholar 

  121. Culmsee, C. et al. Reciprocal inhibition of p53 and nuclear factor-κB transcriptional activities determines cell survival or death in neurons. J. Neurosci. 23, 8586–8595 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Kuno, R. et al. Autocrine activation of microglia by tumor necrosis factor-α. J. Neuroimmunol. 162, 89–96 (2005).

    Article  CAS  PubMed  Google Scholar 

  123. Listwak, S. J., Rathore, P. & Herkenham, M. Minimal NF-κB activity in neurons. Neuroscience 250, 282–299 (2013).

    Article  CAS  PubMed  Google Scholar 

  124. Tantiwong, P. et al. NF-κB activity in muscle from obese and type 2 diabetic subjects under basal and exercise-stimulated conditions. Am. J. Physiol. Endocrinol. Metab. 299, E794–E801 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Dietrich, M. O. & Horvath, T. L. Hypothalamic control of energy balance: insights into the role of synaptic plasticity. Trends Neurosci. 36, 65–73 (2013).

    Article  CAS  PubMed  Google Scholar 

  126. Zeltser, L. M., Seeley, R. J. & Tschöp, M. H. Synaptic plasticity in neuronal circuits regulating energy balance. Nat. Neurosci. 15, 1336–1342 (2012).

    Article  CAS  PubMed  Google Scholar 

  127. Mattson, M. P., Gleichmann, M. & Cheng, A. Mitochondria in neuroplasticity and neurological disorders. Neuron 60, 748–766 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Chang, D. T., Honick, A. S. & Reynolds, I. J. Mitochondrial trafficking to synapses in cultured primary cortical neurons. J. Neurosci. 26, 7035–7045 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Sheng, Z. H. & Cai, Q. Mitochondrial transport in neurons: impact on synaptic homeostasis and neurodegeneration. Nat. Rev. Neurosci. 13, 77–93 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. de Brito, O. M. & Scorrano, L. Mitofusin 2 tethers endoplasmic reticulum to mitochondria. Nature 456, 605–610 (2008).

    Article  CAS  PubMed  Google Scholar 

  131. Vannuvel, K., Renard, P., Raes, M. & Arnould, T. Functional and morphological impact of ER stress on mitochondria. J. Cell. Physiol. 228, 1802–1818 (2013).

    Article  CAS  PubMed  Google Scholar 

  132. Schneeberger, M. et al. Mitofusin 2 in POMC neurons connects ER stress with leptin resistance and energy imbalance. Cell 155, 172–187 (2013).

    Article  CAS  PubMed  Google Scholar 

  133. Diano, S. et al. Peroxisome proliferation-associated control of reactive oxygen species sets melanocortin tone and feeding in diet-induced obesity. Nat. Med. 17, 1121–1127 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Dietrich, M. O., Liu, Z. W. & Horvath, T. L. Mitochondrial dynamics controlled by mitofusins regulate Agrp neuronal activity and diet-induced obesity. Cell 155, 188–199 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Cnop, M. et al. Mechanisms of pancreatic β-cell death in type 1 and type 2 diabetes: many differences, few similarities. Diabetes 54 (Suppl.2), S97–S107 (2005).

    Article  CAS  PubMed  Google Scholar 

  136. Schapira, A. H., Olanow, C. W., Greenamyre, J. T. & Bezard, E. Slowing of neurodegeneration in Parkinson's disease and Huntington's disease: future therapeutic perspectives. Lancet 384, 545–555 (2014).

    Article  CAS  PubMed  Google Scholar 

  137. Alkemade, A. et al. AgRP and NPY expression in the human hypothalamic infundibular nucleus correlate with body mass index, whereas changes in αMSH are related to type 2 diabetes. J. Clin. Endocrinol. Metab. 97, E925–E933 (2012).

    Article  CAS  PubMed  Google Scholar 

  138. Watkins, L. R. & Hutchinson, M. R. A concern on comparing 'apples' and 'oranges' when differences between microglia used in human and rodent studies go far, far beyond simply species: comment on Smith and Dragunow. Trends Neurosci. 37, 189–190 (2014).

    Article  CAS  PubMed  Google Scholar 

  139. Smith, A. M. & Dragunow, M. The human side of microglia. Trends Neurosci. 37, 125–135 (2014).

    Article  CAS  PubMed  Google Scholar 

  140. Finan, B. et al. Targeted estrogen delivery reverses the metabolic syndrome. Nat. Med. 18, 1847–1856 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Komm, B. S. & Mirkin, S. An overview of current and emerging SERMs. J. Steroid Biochem. Mol. Biol. 143, 207–222 (2014).

    Article  CAS  PubMed  Google Scholar 

  142. van der Goes, A., Hoekstra, K., van den Berg, T. K. & Dijkstra, C. D. Dexamethasone promotes phagocytosis and bacterial killing by human monocytes/macrophages in vitro. J. Leukoc. Biol. 67, 801–807 (2000).

    Article  CAS  PubMed  Google Scholar 

  143. Caro, J. F. & Amatruda, J. M. Glucocorticoid-induced insulin resistance: the importance of postbinding events in the regulation of insulin binding, action, and degradation in freshly isolated and primary cultures of rat hepatocytes. J. Clin. Invest. 69, 866–875 (1982).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Kennedy, B., Elayan, H. & Ziegler, M. G. Glucocorticoid induction of epinephrine synthesizing enzyme in rat skeletal muscle and insulin resistance. J. Clin. Invest. 92, 303–307 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Yi, C. X. et al. Glucocorticoid signaling in the arcuate nucleus modulates hepatic insulin sensitivity. Diabetes 61, 339–345 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

F.L.H. is supported by grants from the Deutsche Forschungsgemeinschaft (SFB TRR 43, NeuroCure Exc 257 and HE 3130/6-1), the Federal Ministry of Education and Research (DLR/BMBF; Kompetenznetz Degenerative Demenzen) and by a collaborative research grant of the Berlin Institute of Health (BIH). I.B. is supported by the Deutsche Forschungsgemeinsschaft (FOR 1336 and SFB 1052), ICEMED and DZD. M.H.T. is supported by the Alexander von Humboldt Foundation, the Deutsches Zentrum für Diabetesforschung (DZD), and the Helmholtz Alliance ICEMED–Imaging and Curing Environmental Metabolic Diseases, through the Initiative and Networking Fund of the Helmholtz Association.

Author information

Authors and Affiliations

Authors

Contributions

M.H.T. and C.-X.Y. provided substantial contribution to discussion of the content. S.K. and C.-X.Y. wrote the article. F.L.H., I.B., M.P., M.H.T. and C.-X.Y. reviewed and edited the manuscript before submission.

Corresponding author

Correspondence to Matthias H. Tschöp.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kälin, S., Heppner, F., Bechmann, I. et al. Hypothalamic innate immune reaction in obesity. Nat Rev Endocrinol 11, 339–351 (2015). https://doi.org/10.1038/nrendo.2015.48

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrendo.2015.48

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