Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Oct 1.
Published in final edited form as: Curr Opin Endocrinol Diabetes Obes. 2015 Oct;22(5):325–330. doi: 10.1097/MED.0000000000000182

Hypothalamic inflammation and gliosis in obesity

Mauricio D Dorfman 1, Joshua P Thaler 1
PMCID: PMC4600090  NIHMSID: NIHMS723966  PMID: 26192704

Structured Abstract

Purpose of review

Hypothalamic inflammation and gliosis are recently discovered mechanisms that may contribute to obesity pathogenesis. Current research in this area suggests that investigation of these CNS responses may provide opportunities to develop new weight loss treatments.

Recent findings

In rodents, hypothalamic inflammation and gliosis occur rapidly with high-fat diet consumption prior to significant weight gain. In addition, sensitivity or resistance to diet-induced obesity in rodents generally correlates with the presence or absence of hypothalamic inflammation and reactive gliosis (brain response to injury). Moreover, functional interventions that increase or decrease inflammation in neurons and glia correspondingly alter diet-associated weight gain. However, some conflicting data have recently emerged that question the contribution of hypothalamic inflammation to obesity pathogenesis. However, several studies have detected gliosis and disrupted connectivity in obese humans, highlighting the potential translational importance of this mechanism.

Summary

There is growing evidence that obesity is associated with brain inflammation in humans, particularly in the hypothalamus where its presence may disrupt body weight control and glucose homeostasis. More work is needed to determine whether this response is common in human obesity and to what extent it can be manipulated for therapeutic benefit.

Keywords: obesity, hypothalamus, inflammation, gliosis

INTRODUCTION

Over the past 3 decades, the increased consumption of high calorie “Western” diets along with an ever-more sedentary lifestyle has led to over two-thirds of US adults being currently overweight or obese (body mass index > 25 kg/m2). This trend is particularly concerning due to the strong association of obesity with serious chronic health conditions including diabetes, hypertension, and cardiovascular disease. Few pharmacologic therapies are available that show success in sustaining weight loss over the long term (1), and gastric bypass surgery is an effective but risky procedure that can be offered realistically to only a small number of obese individuals. Thus, new strategies to combat the obesity epidemic are urgently needed, but gaps in our understanding of body weight control continue to limit progress toward this goal.

Body weight is maintained in homeostatic equilibrium by balancing changes in fat mass with compensatory alterations in energy balance (2). This system of energy homeostasis critically depends on the action of leptin, a hormone secreted from adipose tissue in proportion to body fat content. Leptin signals in the brain largely at the level of the hypothalamus, modulating the activity of specific neuronal subsets (including the orexigenic Agouti-related peptide (AgRP) and anorexigenic proopiomelanocortin (POMC) neurons) to reduce appetite and increase energy expenditure (3). This allows lean individuals to maintain stable body weights over many years (4) but fails to protect obese individuals from the deleterious effects of highly palatable, calorie-dense foods and sedentary lifestyles. This differential susceptibility results not from leptin deficiency in obese individuals but rather from an acquired insensitivity to leptin action (5), akin to the central pathogenic role of insulin resistance in type 2 diabetes. In this light, investigations into the potential origins of leptin resistance have drawn inspiration from several decades of research into the mechanisms underlying insulin resistance, focusing particularly on the link between inflammation and impaired hormonal signaling. Beginning with the discovery of hypothalamic inflammation in obese rodents 10 years ago (6••), this area of investigation has led to the hypothesis that obesity progression involves a complex multicellular pathology involving inflammatory signaling, neuronal stress, and reactive gliosis. In this review, we will summarize recent findings on hypothalamic inflammation, the specific CNS cell types involved in this process, the relevance of injury and inflammation to obesity pathogenesis, and the evidence for similar mechanisms in human obesity.

HYPOTHALAMIC INFLAMMATION

An association between obesity and low-grade peripheral inflammation was discovered two decades ago with the identification of increased TNFα expression in the adipose tissue of obese rodents and humans (7). Subsequent work demonstrated that this response includes numerous cytokines and chemokines beyond TNF, is required for obesity-associated insulin resistance, and is propagated by infiltrating macrophages and a variety of other recruited leukocyte cell types (8, 9). Based on these findings, the group of Lício Velloso at the State University of Campinas in Brazil was the first to identify a similar inflammatory response in the hypothalamus (6••). Long-term feeding of a lard-based high fat diet (HFD) to rats increased mediobasal hypothalamic activation of the inflammatory signaling intermediates c-Jun N-terminal kinase (Jnk) and nuclear factor-κB (NF-κB), resulting in the production of proinflammatory cytokines (TNFα, IL-1β, and IL6) and impairment in insulin and leptin signaling. This initial description of hypothalamic inflammation has been reproduced by many other investigators, with extension of the finding to obese mice and non-human primates (1018); models of neonatal overfeeding, type 1 diabetes, and aging (1922); and other hypothalamic nuclei and the hippocampus (19, 23, 24). In addition, HFD-induced inflammation has been associated with hypothalamic resistance to growth hormone and ghrelin signaling (25, 26) and the development of obesity-associated diseases including cognitive dysfunction (27, 28), hypertension (29), hepatic steatosis (30), beta cell dysfunction (31), and, through non-canonical TGF-β signaling, glucose intolerance (32•). Thus, hypothalamic inflammation represents a unifying mechanism of diet-induced obesity (DIO) and metabolic disease.

DIETARY AND CELLULAR MEDIATORS OF HYPOTHALAMIC INFLAMMATION

As mentioned above, the mediobasal hypothalamus is a primary CNS center of metabolic control. Remarkably, the presence of fenestrated capillaries and specialized tanycytes in the median eminence enable this area of the brain to sense and respond rapidly to circulating nutrient and hormonal mediators (33•). This has led to the suggestion that specific macronutrient components—in particular, free fatty acids derived from dietary lipid—are responsible for triggering the inflammatory process. With HFD feeding, mice accumulate a number of fatty acid species in the hypothalamus, including triacylglycerols, diacylglycerols and ceramides (34). Furthermore, acute intracerebroventricular (icv) infusion of lipids such as the saturated fatty acid palmitate and the ω-6 polyunsaturated arachidonate induces inflammatory signaling (11, 3537), alters autophagic protection from cellular stress (38), increases the unfolded protein response in the endoplasmic reticulum (39, 40••), promotes insulin and leptin resistance (11, 41) and possibly triggers apoptosis (42). Likewise, lipid ingestion via milk fat gavage or butter-based HFD consumption increases the hypothalamic levels of saturated fatty acids and proinflammatory gene expression without evidence of systemic inflammation (15••, 17). In addition to fatty acids, several recent studies have suggested that hyperglycemia and fructose exposure can also induce hypothalamic inflammation (21, 43), raising the possibility that nutrient excess itself may be the primary driver of the inflammatory process. Translating these findings to humans is challenging, however, as diet composition, dietary metabolism, and immune function differ greatly between species (44, 45•).

As described above, HFD-induced inflammation in peripheral tissues is tightly associated with infiltration of leukocytes—most notably, classically activated (M1) proinflammatory macrophages (8). In contrast, whether a similar process occurs in the CNS is controversial. In theory, leukocytes cannot enter the brain due to BBB impermeability to diapedesis. Nevertheless, several groups have reported increased numbers of M1-type macrophages in the brains of HFD-fed rodents (46, 47). However, these studies used irradiation-induced myeloablation and bone marrow transplantation to provide tagged donor macrophages, an approach known to compromise the BBB and allow inappropriate ingress of circulating monocytes (48, 49). More recent approaches that included head shielding during irradiation demonstrated the near-complete absence of donor-derived macrophages (15••), indicating that CNS resident cells must be responsible for HFD-induced hypothalamic inflammation. Indeed, the cellular response to HFD in the hypothalamus involves reactive gliosis (15••, 18••, 50), the CNS-specific process of recruitment, proliferation, and morphological transformation of astrocytes and microglia (CNS macrophages) in response to brain injury (51). These changes can be observed as early as 24 hours after the initiation of hypercaloric feeding (18••) and are maintained during prolonged HFD consumption (15••, 18••, 50). Interestingly, hypothalamic gliosis reverses with diet switch, resolving 4 weeks after restarting normal chow feeding (52•); in contrast, caloric restriction of HFD-fed animals does not ameliorate microglial accumulation and activation (53•)(MDD and JPT, unpublished observation), suggesting diet composition is an important determinant of gliosis development. As in other neurodegenerative, infectious, and vascular diseases, HFD-induced reactive gliosis represents an active response to neuron injury (18••), altering neuronal POMC neurovascular coupling through ensheathment of synapses (54), modifying neurotransmitter dynamics through altered astrocyte expression of glutamate and glucose transporters (55), and changing the firing activity of POMC and NPY neurons (56•). Eventually, HFD exposure results in loss of POMC neurons (18••), reduced hypothalamic neurogenesis (57), and impaired synaptic plasticity (54), all potential contributors to excess weight gain. Together, these studies suggest the possibility that glial cells play a primary role in obesity pathogenesis.

Though a new area of investigation, a number of studies have pointed to an active role of glial cells in the control of energy homeostasis. Both astrocyte leptin signaling and overall astrocyte activation can regulate feeding behavior under basal and ghrelin-stimulated conditions (58•, 59•). Tanycytes—specialized glial cells of the median eminence that contribute to blood-brain barrier (BBB) transport and can serve as neural progenitors—promote weight gain during HFD feeding both by limiting leptin access to the mediobasal hypothalamus (33•) and by producing new neurons that increase food intake and decrease energy expenditure (60). Likewise, the oligodendrocyte progenitor cell (OPC) functions through its proliferation/maturation proteoglycan NG2 to increase body weight and fat mass (61). Finally, microglia are critical mediators of HFD-induced inflammatory signaling, modulating hypothalamic neuronal activity and food intake both during basal and HFD feeding (15••, 46, 56•) (MDD and JPT, unpublished data). These data notwithstanding, it should be noted that obesogenic diet consumption is associated with nearly instant glutamate release in the mediobasal hypothalamus, likely stimulated by orosensory perception of food (62), suggesting that glutamatergic neurons themselves may be the primary responders to HFD.

CONTRIBUTION OF HYPOTHALAMIC INFLAMMATION TO OBESITY PATHOGENESIS

Hypothalamic inflammation is a robust and reproducible response to HFD feeding in rodent models, but its relevance as an obesity mechanism is still unclear. In favor of a causative role, hypothalamic inflammation occurs prior to obesity onset within the first few days of HFD feeding (18••). Moreover, blocking the NF-κB pathway in all CNS neurons, only hypothalamic neurons, or AgRP neurons exclusively, reduces levels of HFD-induced leptin resistance and weight gain (40••, 63). Similarly, preventing Toll-like receptor 4 (TLR4) signaling with blocking antibodies or through deletion of MyD88, a TLR and IL-1 signaling intermediate, improves hypothalamic leptin and insulin sensitivity and limits DIO (35, 41). Conversely, deletion of GABA B receptors from POMC neurons results in obesity and increased hypothalamic inflammation (64). Likewise, infusion of low dose TNF-α or viral activation of neuronal NF-κB signaling promotes hyperphagia and weight gain (40••, 65). These data collectively suggest that in some contexts, hypothalamic inflammation is both necessary and sufficient for DIO.

Glial cell inflammation may also contribute to DIO. Use of a virally-expressed dominant negative blocker of NF-κB signaling in astrocytes prevents the acute astrocytosis response induced by HFD (66) while depletion of microglia with a colony stimulating factor-1 receptor (CSF-1R) antagonist reduces food intake in the setting of excess saturated fat ingestion (15••). However, not all forms of obesity require hypothalamic inflammation and gliosis. For example, leptin-deficient ob/ob mice become extremely obese on regular chow without gliosis (53•). Nevertheless, exposing ob/ob mice to HFD feeding increases leptin resistance, weight gain, microglial activity, and hypothalamic inflammation (53•, 67), suggesting that DIO sensitivity correlates with hypothalamic inflammation and gliosis. As another example, HFD-fed male C57BL6 mice are highly DIO sensitive due to increased hypothalamic inflammation from reduced estrogen signaling in neurons and astrocytes (14•). Conversely, female C57Bl6 mice and other DIO resistant models (e.g. DIO-R rats) lack central inflammatory and gliosis responses (14•, 46, 68, 69). Recently, we discovered a sex-specific microglial switch for DIO susceptibility using a mouse model that lacks the fractalkine/CX3CL1 receptor CX3CR1 (MDD and JPT, unpublished data). CX3CL1, a chemokine secreted by neurons to maintain microglial quiescence, increases in the hypothalamus of females exposed to HFD. Remarkably, HFD-fed female Cx3cr1 knockout mice develop hypothalamic microglial activation and obesity compared to their heterozygous littermates. Conversely, HFD-fed males treated with a CNS CX3CL1 infusion have reduced microgliosis and weight gain in a CX3CR1-dependent manner, suggesting that HFD-induced microglial inflammatory activation contributes to obesity pathogenesis.

CONFLICTING DATA

While substantial evidence suggests that hypothalamic inflammation and gliosis are associated with HFD-induced obesity, recent studies cast doubt on their causal role in the pathogenesis of DIO. First, it has long been known that total-body proinflammatory cytokine and cytokine receptor knockout models (e.g., IL-1, IL-6, IL-1 receptor, and TNF receptors) display obesity phenotypes rather than being protected from DIO. Second, several studies have demonstrated dissociation between weight changes and alterations in hypothalamic inflammation. In the first, DIO-sensitive HFD-fed rats switched back to regular chow for 8 weeks lose their excess weight despite sustained elevations in hypothalamic inflammatory gene expression (69). Similarly, chronic ghrelin treatment increases adiposity while reducing hypothalamic inflammation and gliosis (70). Conversely, neuronal peroxisome proliferator–activated receptor-δ and growth hormone receptor knockout mice are resistant to HFD-induced hypothalamic inflammation and gliosis yet more susceptible to DIO than wild-type controls (71). Finally, a stereological analysis of the hypothalamus in HFD-fed mice revealed minimal evidence of neuronal loss and sustained gliosis though the diet composition and duration were different than prior studies (72•). Thus, the evidence supporting a direct role of hypothalamic inflammation and gliosis in promoting weight gain is clearly mixed. These considerations highlight the importance of determining whether hypothalamic inflammation and injury occurs in humans and contributes to obesity pathogenesis.

HYPOTHALAMIC PATHOLOGY IN HUMANS?

Although the extent to which obesity and/or HFD feeding impacts hypothalamic structure and function in humans remains uncertain, translational insights using brain imaging are beginning to support this type of neuropathologic model. A retrospective analysis of MRIs from 34 subjects (BMI range: 17.7–44.1 kg/m2) revealed evidence of gliosis in the mediobasal hypothalamus that correlated with BMI (18••), and recently the findings were replicated in a prospective cohort showing an additional link between gliosis and insulin resistance (Ellen Schur, personal communication). A separate MRI study in 44 overweight/obese subjects reported an inverse correlation between systemic inflammation (measured as serum fibrinogen) and the integrity of brain structures involved in food reward and feeding behavior (73). Most recently, diffusion tensor imaging (DTI) was used to screen for hypothalamic damage in 44 obese patients and controls (74••). In the obese cohort, lower axon diffusivity (i.e. increased damage) was correlated with increased BMI and impaired cognitive performance. Finally, a few studies of structural alterations in the brains of obese subjects have shown reduced connectivity through the corpus callosum (75, 76), reduced hypothalamic volume in obese females with insulin resistance (77), and greater connectivity between the hypothalamus and reward-related brain areas than homeostatic areas (78•). Together, these data support the hypothesis that humans develop hypothalamic pathology akin to that of rodents with gliosis and injury potentially promoting weight gain, insulin resistance, and cognitive impairment.

SUMMARY

The causes and consequences of hypothalamic inflammation and gliosis remain incompletely understood, but the field is progressing rapidly. Hypothalamic inflammation shares several common features with obesity-associated peripheral inflammation, but the involvement of unique CNS cell types identifies it as a fundamentally distinct process. Though there is considerable experimental support for a pathologic role of hypothalamic inflammation and gliosis, recent conflicting data have muddied the waters. Nevertheless, the preponderance of evidence in humans and animal models to date points to structural damage of the hypothalamic energy homeostasis center as a central deleterious consequence of diet-induced inflammation and gliosis and suggests an opportunity to reverse weight gain through manipulation of this complex multicellular process.

Key Points.

  1. High-fat diet feeding is associated with hypothalamic inflammation, reactive gliosis, and neuron injury in rodents.

  2. Interventions that increase or decrease inflammatory pathways in the hypothalamus have corresponding effects to promote or prevent diet-induced obesity.

  3. Glial cells play an important role in transmitting diet-related inflammatory signals and can modulate neuronal regulation of energy balance.

  4. Obese humans show signs of hypothalamic gliosis and impaired connectivity possibly contributing to weight gain, metabolic disease, and cognitive dysfunction.

Acknowledgments

Financial support and sponsorship

This work was supported by a mentor-based postdoctoral fellowship from the American Diabetes Association (ADA) to MDD and by an ADA Pathway to Stop Diabetes Grant 1-14-ACE-51 and NIDDK Career Development Award K08 K08DK088872 to JPT.

Footnotes

Conflicts of interest

None.

Financial Disclosure: This work was supported by a mentor-based postdoctoral fellowship from the American Diabetes Association (ADA) to MDD and by an ADA Pathway to Stop Diabetes Grant 1-14-ACE-51 and NIDDK Career Development Award K08 K08DK088872 to JPT.

References

  • 1.Yanovski SZ, Yanovski JA. Long-term drug treatment for obesity: a systematic and clinical review. Jama. 2014;311(1):74–86. doi: 10.1001/jama.2013.281361. Epub 2013/11/16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Schwartz MW, Woods SC, Porte D, Jr, Seeley RJ, Baskin DG. Central nervous system control of food intake. Nature. 2000;404(6778):661–71. doi: 10.1038/35007534. Epub 2000/04/15. [DOI] [PubMed] [Google Scholar]
  • 3.Morton GJ, Cummings DE, Baskin DG, Barsh GS, Schwartz MW. Central nervous system control of food intake and body weight. Nature. 2006;443(7109):289–95. doi: 10.1038/nature05026. Epub 2006/09/22. [DOI] [PubMed] [Google Scholar]
  • 4.Cohn C, Joseph D. Influence of body weight and body fat on appetite of “normal” lean and obese rats. The Yale journal of biology and medicine. 1962;34:598–607. Epub 1962/06/01. [PMC free article] [PubMed] [Google Scholar]
  • 5.Considine RV, Sinha MK, Heiman ML, Kriauciunas A, Stephens TW, Nyce MR, et al. Serum immunoreactive-leptin concentrations in normal-weight and obese humans. The New England journal of medicine. 1996;334(5):292–5. doi: 10.1056/NEJM199602013340503. Epub 1996/02/01. [DOI] [PubMed] [Google Scholar]
  • ••6.De Souza CT, Araujo EP, Bordin S, Ashimine R, Zollner RL, Boschero AC, et al. Consumption of a fat-rich diet activates a proinflammatory response and induces insulin resistance in the hypothalamus. Endocrinology. 2005;146(10):4192–9. doi: 10.1210/en.2004-1520. This study is the first report showing an association between obesity and hypothalamic inflammation. [DOI] [PubMed] [Google Scholar]
  • 7.Hotamisligil GS, Arner P, Caro JF, Atkinson RL, Spiegelman BM. Increased adipose tissue expression of tumor necrosis factor-alpha in human obesity and insulin resistance. The Journal of clinical investigation. 1995;95(5):2409–15. doi: 10.1172/JCI117936. Epub 1995/05/01. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Lumeng CN, Saltiel AR. Inflammatory links between obesity and metabolic disease. J Clin Invest. 2011;121(6):2111–7. doi: 10.1172/JCI57132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Brestoff JR, Artis D. Immune regulation of metabolic homeostasis in health and disease. Cell. 2015;161(1):146–60. doi: 10.1016/j.cell.2015.02.022. Epub 2015/03/31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Milanski M, Degasperi G, Coope A, Morari J, Denis R, Cintra DE, et al. Saturated fatty acids produce an inflammatory response predominantly through the activation of TLR4 signaling in hypothalamus: implications for the pathogenesis of obesity. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2009;29(2):359–70. doi: 10.1523/JNEUROSCI.2760-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Posey KA, Clegg DJ, Printz RL, Byun J, Morton GJ, Vivekanandan-Giri A, et al. Hypothalamic proinflammatory lipid accumulation, inflammation, and insulin resistance in rats fed a high-fat diet. American journal of physiology Endocrinology and metabolism. 2009;296(5):E1003–12. doi: 10.1152/ajpendo.90377.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Kleinridders A, Schenten D, Könner AC, Belgardt BF, Mauer J, Okamura T, et al. MyD88 signaling in the CNS is required for development of fatty acid-induced leptin resistance and diet-induced obesity. Cell metabolism. 2009;10(4):249–59. doi: 10.1016/j.cmet.2009.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Grayson BE, Levasseur PR, Williams SM, Smith MS, Marks DL, Grove KL. Changes in melanocortin expression and inflammatory pathways in fetal offspring of nonhuman primates fed a high-fat diet. Endocrinology. 2010;151(4):1622–32. doi: 10.1210/en.2009-1019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • •14.Morselli E, Fuente-Martin E, Finan B, Kim M, Frank A, Garcia-Caceres C, et al. Hypothalamic PGC-1alpha protects against high-fat diet exposure by regulating ERalpha. Cell reports. 2014;9(2):633–45. doi: 10.1016/j.celrep.2014.09.025. The hypothalamic response to HFD is sexually dimorphic with male mice being predisposed to diet-induced hypothalamic inflammation from down-regulation of estrogen signaling in neurons and astrocytes. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • ••15.Valdearcos M, Robblee MM, Benjamin DI, Nomura DK, Xu AW, Koliwad SK. Microglia dictate the impact of saturated fat consumption on hypothalamic inflammation and neuronal function. Cell reports. 2014;9(6):2124–38. doi: 10.1016/j.celrep.2014.11.018. This study identified microglia as the primary responders to dietary saturated fat and provided evidence that they are required for lipid-induced neuronal stress, hypothalamic inflammation, leptin resistance, and hyperphagia. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Weissmann L, Quaresma PG, Santos AC, de Matos AH, Pascoal VD, Zanotto TM, et al. IKKepsilon is key to induction of insulin resistance in the hypothalamus, and its inhibition reverses obesity. Diabetes. 2014;63(10):3334–45. doi: 10.2337/db13-1817. Epub 2014/05/09. [DOI] [PubMed] [Google Scholar]
  • 17.Maric T, Woodside B, Luheshi GN. The effects of dietary saturated fat on basal hypothalamic neuroinflammation in rats. Brain, behavior, and immunity. 2014;36:35–45. doi: 10.1016/j.bbi.2013.09.011. Epub 2013/10/01. [DOI] [PubMed] [Google Scholar]
  • ••18.Thaler JP, Yi CX, Schur EA, Guyenet SJ, Hwang BH, Dietrich MO, et al. Obesity is associated with hypothalamic injury in rodents and humans. The Journal of clinical investigation. 2012;122(1):153–62. doi: 10.1172/JCI59660. This study showed an increase in hypothalamic neuronal stress, inflammation and gliosis during the first days of HFD exposure in rodents and found an association between hypothalamic gliosis and BMI in humans. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Ziko I, De Luca S, Dinan T, Barwood JM, Sominsky L, Cai G, et al. Neonatal overfeeding alters hypothalamic microglial profiles and central responses to immune challenge long-term. Brain, behavior, and immunity. 2014;41:32–43. doi: 10.1016/j.bbi.2014.06.014. Epub 2014/07/01. [DOI] [PubMed] [Google Scholar]
  • 20.Wee YS, Weis JJ, Gahring LC, Rogers SW, Weis JH. Age-related onset of obesity corresponds with metabolic dysregulation and altered microglia morphology in mice deficient for Ifitm proteins. PloS one. 2015;10(4):e0123218. doi: 10.1371/journal.pone.0123218. Epub 2015/04/10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Hu P, Thinschmidt JS, Yan Y, Hazra S, Bhatwadekar A, Caballero S, et al. CNS inflammation and bone marrow neuropathy in type 1 diabetes. The American journal of pathology. 2013;183(5):1608–20. doi: 10.1016/j.ajpath.2013.07.009. Epub 2013/10/29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Hu P, Thinschmidt JS, Caballero S, Adamson S, Cole L, Chan-Ling T, et al. Loss of survival factors and activation of inflammatory cascades in brain sympathetic centers in type 1 diabetic mice. American journal of physiology Endocrinology and metabolism. 2015;308(8):E688–98. doi: 10.1152/ajpendo.00504.2014. Epub 2015/02/26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.de Kloet AD, Pioquinto DJ, Nguyen D, Wang L, Smith JA, Hiller H, et al. Obesity induces neuroinflammation mediated by altered expression of the renin-angiotensin system in mouse forebrain nuclei. Physiology & behavior. 2014;136:31–8. doi: 10.1016/j.physbeh.2014.01.016. Epub 2014/02/11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Cano V, Valladolid-Acebes I, Hernandez-Nuno F, Merino B, Del Olmo N, Chowen JA, et al. Morphological changes in glial fibrillary acidic protein immunopositive astrocytes in the hippocampus of dietary-induced obese mice. Neuroreport. 2014 doi: 10.1097/WNR.0000000000000180. Epub 2014/06/10. [DOI] [PubMed] [Google Scholar]
  • 25.Baquedano E, Ruiz-Lopez AM, Sustarsic EG, Herpy J, List EO, Chowen JA, et al. The absence of GH signaling affects the susceptibility to high-fat diet-induced hypothalamic inflammation in male mice. Endocrinology. 2014;155(12):4856–67. doi: 10.1210/en.2014-1367. Epub 2014/09/23. [DOI] [PubMed] [Google Scholar]
  • 26.Naznin F, Toshinai K, Waise TM, Namkoong C, Moin AS, Sakoda H, et al. Diet-induced obesity causes peripheral and central ghrelin resistance by promoting inflammation. The Journal of endocrinology. 2015 doi: 10.1530/JOE-15-0139. Epub 2015/05/29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Pistell PJ, Morrison CD, Gupta S, Knight AG, Keller JN, Ingram DK, et al. Cognitive impairment following high fat diet consumption is associated with brain inflammation. Journal of neuroimmunology. 2010;219(1–2):25–32. doi: 10.1016/j.jneuroim.2009.11.010. Epub 2009/12/17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Jeon BT, Jeong EA, Shin HJ, Lee Y, Lee DH, Kim HJ, et al. Resveratrol attenuates obesity-associated peripheral and central inflammation and improves memory deficit in mice fed a high-fat diet. Diabetes. 2012;61(6):1444–54. doi: 10.2337/db11-1498. Epub 2012/03/01. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Purkayastha S, Zhang G, Cai D. Uncoupling the mechanisms of obesity and hypertension by targeting hypothalamic IKK-β and NF-κB. Nature medicine. 2011;17(7):883–7. doi: 10.1038/nm.2372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Milanski M, Arruda AP, Coope A, Ignacio-Souza LM, Nunez CE, Roman EA, et al. Inhibition of hypothalamic inflammation reverses diet-induced insulin resistance in the liver. Diabetes. 2012;61(6):1455–62. doi: 10.2337/db11-0390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Calegari VC, Torsoni AS, Vanzela EC, Araujo EP, Morari J, Zoppi CC, et al. Inflammation of the hypothalamus leads to defective pancreatic islet function. The Journal of biological chemistry. 2011;286(15):12870–80. doi: 10.1074/jbc.M110.173021. Epub 2011/01/25. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • •32.Yan J, Zhang H, Yin Y, Li J, Tang Y, Purkayastha S, et al. Obesity- and aging-induced excess of central transforming growth factor-beta potentiates diabetic development via an RNA stress response. Nature medicine. 2014;20(9):1001–8. doi: 10.1038/nm.3616. Epub 2014/08/05. This study demonstrated a mechanism of obesity-associated glucose intolerance involving CNS inflammation through atypical TGF-β signaling. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • •33.Balland E, Dam J, Langlet F, Caron E, Steculorum S, Messina A, et al. Hypothalamic tanycytes are an ERK-gated conduit for leptin into the brain. Cell metabolism. 2014;19(2):293–301. doi: 10.1016/j.cmet.2013.12.015. Epub 2014/02/11. Leptin transport across the BBB is disrupted during HFD feeding by excess ERK phosphorylation in tanycytes. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Borg ML, Omran SF, Weir J, Meikle PJ, Watt MJ. Consumption of a high-fat diet, but not regular endurance exercise training, regulates hypothalamic lipid accumulation in mice. The Journal of physiology. 2012;590(Pt 17):4377–89. doi: 10.1113/jphysiol.2012.233288. Epub 2012/06/08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Milanski M, Degasperi G, Coope A, Morari J, Denis R, Cintra DE, 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. 2009;29(2):359–70. doi: 10.1523/JNEUROSCI.2760-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Cheng L, Yu Y, Szabo A, Wu Y, Wang H, Camer D, et al. Palmitic acid induces central leptin resistance and impairs hepatic glucose and lipid metabolism in male mice. The Journal of nutritional biochemistry. 2015;26(5):541–8. doi: 10.1016/j.jnutbio.2014.12.011. Epub 2015/03/01. [DOI] [PubMed] [Google Scholar]
  • 37.Cheng L, Yu Y, Zhang Q, Szabo A, Wang H, Huang XF. Arachidonic acid impairs hypothalamic leptin signaling and hepatic energy homeostasis in mice. Molecular and cellular endocrinology. 2015;412:12–8. doi: 10.1016/j.mce.2015.04.025. Epub 2015/05/20. [DOI] [PubMed] [Google Scholar]
  • 38.Portovedo M, Ignacio-Souza LM, Bombassaro B, Coope A, Reginato A, Razolli DS, et al. Saturated fatty acids modulate autophagy’s proteins in the hypothalamus. PloS one. 2015;10(3):e0119850. doi: 10.1371/journal.pone.0119850. Epub 2015/03/19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Ozcan L, Ergin AS, Lu A, Chung J, Sarkar S, Nie D, et al. Endoplasmic reticulum stress plays a central role in development of leptin resistance. Cell metabolism. 2009;9(1):35–51. doi: 10.1016/j.cmet.2008.12.004. [DOI] [PubMed] [Google Scholar]
  • ••40.Zhang X, Zhang G, Zhang H, Karin M, Bai H, Cai D. Hypothalamic IKKbeta/NF-kappaB and ER stress link overnutrition to energy imbalance and obesity. Cell. 2008;135(1):61–73. doi: 10.1016/j.cell.2008.07.043. Proinflammatory NF-κB signaling in hypothalamic neurons is activated by overnutrition. Disrupting this signaling in the brain or in specific hypothalamic neuron populations (AgRP) reduces inflammation, ER stress, leptin resistance, HFD intake and body weight. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Kleinridders A, Schenten D, Konner AC, Belgardt BF, Mauer J, Okamura T, et al. MyD88 signaling in the CNS is required for development of fatty acid-induced leptin resistance and diet-induced obesity. Cell metabolism. 2009;10(4):249–59. doi: 10.1016/j.cmet.2009.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Moraes JC, Coope A, Morari J, Cintra DE, Roman EA, Pauli JR, et al. High-fat diet induces apoptosis of hypothalamic neurons. PloS one. 2009;4(4):e5045. doi: 10.1371/journal.pone.0005045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Li JM, Ge CX, Xu MX, Wang W, Yu R, Fan CY, et al. Betaine recovers hypothalamic neural injury by inhibiting astrogliosis and inflammation in fructose-fed rats. Molecular nutrition & food research. 2015;59(2):189–202. doi: 10.1002/mnfr.201400307. Epub 2014/10/11. [DOI] [PubMed] [Google Scholar]
  • 44.Mestas J, Hughes CC. Of mice and not men: differences between mouse and human immunology. J Immunol. 2004;172(5):2731–8. doi: 10.4049/jimmunol.172.5.2731. Epub 2004/02/24. [DOI] [PubMed] [Google Scholar]
  • •45.Lai M, Chandrasekera PC, Barnard ND. You are what you eat, or are you? The challenges of translating high-fat-fed rodents to human obesity and diabetes. Nutrition & diabetes. 2014;4:e135. doi: 10.1038/nutd.2014.30. Epub 2014/09/10. A well-written review of the problems with extrapolating mouse metabolic data to human nutrition and obesity. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Morari J, Anhe GF, Nascimento LF, de Moura RF, Razolli D, Solon C, et al. Fractalkine (CX3CL1) is involved in the early activation of hypothalamic inflammation in experimental obesity. Diabetes. 2014;63(11):3770–84. doi: 10.2337/db13-1495. [DOI] [PubMed] [Google Scholar]
  • 47.Buckman LB, Hasty AH, Flaherty DK, Buckman CT, Thompson MM, Matlock BK, et al. Obesity induced by a high-fat diet is associated with increased immune cell entry into the central nervous system. Brain, behavior, and immunity. 2014;35:33–42. doi: 10.1016/j.bbi.2013.06.007. Epub 2013/07/09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Kierdorf K, Katzmarski N, Haas CA, Prinz M. Bone marrow cell recruitment to the brain in the absence of irradiation or parabiosis bias. PloS one. 2013;8(3):e58544. doi: 10.1371/journal.pone.0058544. Epub 2013/03/26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Yang Y, Jorstad NL, Shiao C, Cherne MK, Khademi SB, Montine KS, et al. Perivascular, but not parenchymal, cerebral engraftment of donor cells after non-myeloablative bone marrow transplantation. Experimental and molecular pathology. 2013;95(1):7–17. doi: 10.1016/j.yexmp.2013.03.010. Epub 2013/04/10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Buckman LB, Thompson MM, Moreno HN, Ellacott KL. Regional astrogliosis in the mouse hypothalamus in response to obesity. The Journal of comparative neurology. 2013;521(6):1322–33. doi: 10.1002/cne.23233. Epub 2012/10/11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Burda JE, Sofroniew MV. Reactive gliosis and the multicellular response to CNS damage and disease. Neuron. 2014;81(2):229–48. doi: 10.1016/j.neuron.2013.12.034. Epub 2014/01/28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • •52.Berkseth KE, Guyenet SJ, Melhorn SJ, Lee D, Thaler JP, Schur EA, et al. Hypothalamic gliosis associated with high-fat diet feeding is reversible in mice: a combined immunohistochemical and magnetic resonance imaging study. Endocrinology. 2014;155(8):2858–67. doi: 10.1210/en.2014-1121. Epub 2014/06/11. This study uses MRI scanning to demonstrate that diet-induced gliosis is potentially reversible. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • •53.Gao Y, Ottaway N, Schriever SC, Legutko B, Garcia-Caceres C, de la Fuente E, et al. Hormones and diet, but not body weight, control hypothalamic microglial activity. Glia. 2014;62(1):17–25. doi: 10.1002/glia.22580. Epub 2013/10/30. This paper describes a careful comparative analysis of the differences between genetic and diet-induced obesity on microglial activation. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Horvath TL, Sarman B, García-Cáceres C, Enriori PJ, Sotonyi P, Shanabrough M, et al. Synaptic input organization of the melanocortin system predicts diet-induced hypothalamic reactive gliosis and obesity. Proceedings of the National Academy of Sciences of the United States of America. 2010;107(33):14875–80. doi: 10.1073/pnas.1004282107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Fuente-Martin E, Garcia-Caceres C, Granado M, de Ceballos ML, Sanchez-Garrido MA, Sarman B, et al. Leptin regulates glutamate and glucose transporters in hypothalamic astrocytes. The Journal of clinical investigation. 2012;122(11):3900–13. doi: 10.1172/JCI64102. Epub 2012/10/16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Reis WL, Yi CX, Gao Y, Tschop MH, Stern JE. Brain innate immunity regulates hypothalamic arcuate neuronal activity and feeding behavior. Endocrinology. 2015;156(4):1303–15. doi: 10.1210/en.2014-1849. Microglia can modulate the electrical activity of energy homeostasis-regulating neurons and contribute to food intake regulation. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.McNay DE, Briancon N, Kokoeva MV, Maratos-Flier E, Flier JS. Remodeling of the arcuate nucleus energy-balance circuit is inhibited in obese mice. The Journal of clinical investigation. 2012;122(1):142–52. doi: 10.1172/JCI43134. Epub 2011/12/29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • •58.Kim JG, Suyama S, Koch M, Jin S, Argente-Arizon P, Argente J, et al. Leptin signaling in astrocytes regulates hypothalamic neuronal circuits and feeding. Nature neuroscience. 2014;17(7):908–10. doi: 10.1038/nn.3725. Epub 2014/06/02. The first demonstration of a functional involvement of leptin signaling in glial cells in energy homeostasis regulation. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • •59.Yang L, Qi Y, Yang Y. Astrocytes Control Food Intake by Inhibiting AGRP Neuron Activity via Adenosine A1 Receptors. Cell reports. 2015;11(5):798–807. doi: 10.1016/j.celrep.2015.04.002. Epub 2015/04/30. This study uses DREADD chemogenetic technology to demonstrate a role for astrocytes in the regulation of food intake. [DOI] [PubMed] [Google Scholar]
  • 60.Lee DA, Bedont JL, Pak T, Wang H, Song J, Miranda-Angulo A, et al. Tanycytes of the hypothalamic median eminence form a diet-responsive neurogenic niche. Nature neuroscience. 2012;15(5):700–2. doi: 10.1038/nn.3079. Epub 2012/03/27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Chang Y, She ZG, Sakimura K, Roberts A, Kucharova K, Rowitch DH, et al. Ablation of NG2 proteoglycan leads to deficits in brown fat function and to adult onset obesity. PloS one. 2012;7(1):e30637. doi: 10.1371/journal.pone.0030637. Epub 2012/02/02. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Guyenet SJ, Matsen ME, Morton GJ, Kaiyala KJ, Schwartz MW. Rapid glutamate release in the mediobasal hypothalamus accompanies feeding and is exaggerated by an obesogenic food. Molecular metabolism. 2013;2(2):116–22. doi: 10.1016/j.molmet.2013.02.001. Epub 2013/11/08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Benzler J, Ganjam GK, Pretz D, Oelkrug R, Koch CE, Legler K, et al. Central Inhibition of IKKbeta/NF-kappaB Signaling Attenuates High-Fat Diet-Induced Obesity and Glucose Intolerance. Diabetes. 2015;64(6):2015–27. doi: 10.2337/db14-0093. Epub 2015/01/30. [DOI] [PubMed] [Google Scholar]
  • 64.Ito Y, Banno R, Shibata M, Adachi K, Hagimoto S, Hagiwara D, et al. GABA type B receptor signaling in proopiomelanocortin neurons protects against obesity, insulin resistance, and hypothalamic inflammation in male mice on a high-fat diet. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2013;33(43):17166–73. doi: 10.1523/JNEUROSCI.0897-13.2013. Epub 2013/10/25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Arruda AP, Milanski M, Coope A, Torsoni AS, Ropelle E, Carvalho DP, et al. Low-grade hypothalamic inflammation leads to defective thermogenesis, insulin resistance, and impaired insulin secretion. Endocrinology. 2011;152(4):1314–26. doi: 10.1210/en.2010-0659. [DOI] [PubMed] [Google Scholar]
  • 66.Buckman LB, Thompson MM, Lippert RN, Blackwell TS, Yull FE, Ellacott KL. Evidence for a novel functional role of astrocytes in the acute homeostatic response to high-fat diet intake in mice. Molecular metabolism. 2015;4(1):58–63. doi: 10.1016/j.molmet.2014.10.001. Epub 2015/02/17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Koch CE, Lowe C, Pretz D, Steger J, Williams LM, Tups A. High-fat diet induces leptin resistance in leptin-deficient mice. Journal of neuroendocrinology. 2014;26(2):58–67. doi: 10.1111/jne.12131. Epub 2014/01/03. [DOI] [PubMed] [Google Scholar]
  • 68.Cai G, Dinan T, Barwood JM, De Luca SN, Soch A, Ziko I, et al. Neonatal overfeeding attenuates acute central pro-inflammatory effects of short-term high fat diet. Frontiers in neuroscience. 2014;8:446. doi: 10.3389/fnins.2014.00446. Epub 2015/01/30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Wang X, Ge A, Cheng M, Guo F, Zhao M, Zhou X, et al. Increased hypothalamic inflammation associated with the susceptibility to obesity in rats exposed to high-fat diet. Experimental diabetes research. 2012;2012:847246. doi: 10.1155/2012/847246. Epub 2012/07/31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Garcia-Caceres C, Fuente-Martin E, Diaz F, Granado M, Argente-Arizon P, Frago LM, et al. The opposing effects of ghrelin on hypothalamic and systemic inflammatory processes are modulated by its acylation status and food intake in male rats. Endocrinology. 2014;155(8):2868–80. doi: 10.1210/en.2014-1074. Epub 2014/05/23. [DOI] [PubMed] [Google Scholar]
  • 71.Kocalis HE, Turney MK, Printz RL, Laryea GN, Muglia LJ, Davies SS, et al. Neuron-specific deletion of peroxisome proliferator-activated receptor delta (PPARdelta) in mice leads to increased susceptibility to diet-induced obesity. PloS one. 2012;7(8):e42981. doi: 10.1371/journal.pone.0042981. Epub 2012/08/24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Lemus MB, Bayliss JA, Lockie SH, Santos VV, Reichenbach A, Stark R, et al. A stereological analysis of NPY, POMC, Orexin, GFAP astrocyte, and Iba1 microglia cell number and volume in diet-induced obese male mice. Endocrinology. 2015;156(5):1701–13. doi: 10.1210/en.2014-1961. Epub 2015/03/06. A carefully-conducted immunohistochemical study of neuronal and glial responses to HFD feeding questions previous findings regarding neuron loss and reactive gliosis. [DOI] [PubMed] [Google Scholar]
  • 73.Cazettes F, Cohen JI, Yau PL, Talbot H, Convit A. Obesity-mediated inflammation may damage the brain circuit that regulates food intake. Brain research. 2011;1373:101–9. doi: 10.1016/j.brainres.2010.12.008. Epub 2010/12/15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • ••74.Puig J, Blasco G, Daunis IEJ, Molina X, Xifra G, Ricart W, et al. Hypothalamic damage is associated with inflammatory markers and worse cognitive performance in obese subjects. The Journal of clinical endocrinology and metabolism. 2015;100(2):E276–81. doi: 10.1210/jc.2014-2682. Epub 2014/11/26. The most convincing demonstration to date of hypothalamic structural abnormalities in obese humans. [DOI] [PubMed] [Google Scholar]
  • 75.Mueller K, Anwander A, Moller HE, Horstmann A, Lepsien J, Busse F, et al. Sex-dependent influences of obesity on cerebral white matter investigated by diffusion-tensor imaging. PloS one. 2011;6(4):e18544. doi: 10.1371/journal.pone.0018544. Epub 2011/04/16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Stanek KM, Grieve SM, Brickman AM, Korgaonkar MS, Paul RH, Cohen RA, et al. Obesity is associated with reduced white matter integrity in otherwise healthy adults. Obesity (Silver Spring) 2011;19(3):500–4. doi: 10.1038/oby.2010.312. Epub 2010/12/25. [DOI] [PubMed] [Google Scholar]
  • 77.Ha J, Cohen JI, Tirsi A, Convit A. Association of obesity-mediated insulin resistance and hypothalamic volumes: possible sex differences. Disease markers. 2013;35(4):249–59. doi: 10.1155/2013/531736. Epub 2013/12/18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • •78.Kilpatrick LA, Coveleskie K, Connolly L, Labus JS, Ebrat B, Stains J, et al. Influence of sucrose ingestion on brainstem and hypothalamic intrinsic oscillations in lean and obese women. Gastroenterology. 2014;146(5):1212–21. doi: 10.1053/j.gastro.2014.01.023. Epub 2014/02/01. A well-designed and executed study using a caloric challenge to reveal differences between lean and obese women in connectivity of metabolically important brain areas. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES