Abstract

Meta-inflammation of hypothalamic areas governing energy homeostasis has recently emerged as a process of potential pathophysiological relevance for the development of obesity and its metabolic sequelae. The current model suggests that diet-induced neuronal injury triggers microgliosis and astrocytosis, conditions which ultimately may induce functional impairment of hypothalamic circuits governing feeding behavior, systemic metabolism, and body weight. Epidemiological data indicate that low circulating HDL levels, besides conveying cardiovascular risk, also correlate strongly with obesity. We simulated that condition by using a genetic loss of function mouse model (apoA-I-/-) with markedly reduced HDL levels to investigate whether HDL may directly modulate hypothalamic inflammation. Astrogliosis was significantly enhanced in the hypothalami of apoA-I-/- compared with apoA-I+/+ mice and was associated with compromised mitochondrial function. apoA-I-/- mice exhibited key components of metabolic disease, like increased fat mass, fasting glucose levels, hepatic triglyceride content, and hepatic glucose output compared with apoA-I+/+ controls. Administration of reconstituted HDL (CSL-111) normalized hypothalamic inflammation and mitochondrial function markers in apoA-I-/- mice. Treatment of primary astrocytes with apoA-I resulted in enhanced mitochondrial activity, implying that circulating HDL levels are likely important for astrocyte function. HDL-based therapies may consequently avert reactive gliosis in hypothalamic astrocytes by improving mitochondrial bioenergetics and thereby offering potential treatment and prevention for obesity and metabolic disease.

Highlights

  • Meta-inflammation of hypothalamic areas governing energy homeostasis has recently emerged as a process of potential pathophysiological relevance for the development of obesity and its metabolic sequelae

  • Using a genetic loss of function mouse model, we report for the first time that low circulating HDL levels promote astrocyte activation and induce mitochondrial dysfunction within the hypothalamic astrocytes that is associated with enhanced adiposity and impaired glucose homeostasis

  • Using immunohistochemical staining of brain sections for glial fibrillary acidic protein (GFAP), we found that astrocytic gliosis is markedly increased in the arcuate nucleus (ARC) of apoA-I / compared with apoA-I+/+ mice (Fig. 1D)

Read more

Summary

Introduction

Meta-inflammation of hypothalamic areas governing energy homeostasis has recently emerged as a process of potential pathophysiological relevance for the development of obesity and its metabolic sequelae. Treatment of primary astrocytes with apoA-I resulted in enhanced mitochondrial activity, implying that circulating HDL levels are likely important for astrocyte function. Emerging evidence indicates that chronic consumption of excess calories induces inflammation in the arcuate nucleus (ARC) of the hypothalamus, a brain area important for the regulation of food intake and energy expenditure [2] This process is considered an early and determining factor for the onset of obesity because it occurs prior to body weight gain and Abbreviations: A , amyloid- peptide; ARC, arcuate nucleus; BBB, blood-brain barrier; 2DG, 2-deoxy-glucose; ECAR, extracellular acidification rate; FCCP, carbonyl cyanide-4-(trifluoromethoxy)phenylhydrazone; GFAP, glial fibrillary acidic protein; Iba-1, ionized calcium-binding adaptor molecule 1; IL, interleukin; OCR, oxygen consumption rate; OXPHOS, oxidative phosphorylation; PGC1- , peroxisome proliferator-activated receptor c coactivator 1 ; PPR, proton production rate; rHDL, reconstituted HDL. Novel therapeutic strategies may be developed to target this impairment

Methods
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call