4.8 Article

Macrophage-specific PPARγ controls alternative activation and improves insulin resistance

Journal

NATURE
Volume 447, Issue 7148, Pages 1116-U12

Publisher

NATURE PUBLISHING GROUP
DOI: 10.1038/nature05894

Keywords

-

Funding

  1. NHLBI NIH HHS [R01 HL076746-01, R01 HL076746, R01 HL076746-05, R01 HL076746-03, R01 HL076746-02, R01 HL076746-04] Funding Source: Medline
  2. NIAID NIH HHS [T32 AI007290, AI07290, F31 AI066402] Funding Source: Medline
  3. NIDDK NIH HHS [R01 DK076760, K08 DK062386, K08 DK062386-06, R01 DK066525, K08 DK062386-05, R01 DK066525-05] Funding Source: Medline

Ask authors/readers for more resources

Obesity and insulin resistance, the cardinal features of metabolic syndrome, are closely associated with a state of low-grade inflammation(1,2). In adipose tissue chronic overnutrition leads to macrophage infiltration, resulting in local inflammation that potentiates insulin resistance(3,4). For instance, transgenic expression of Mcp1 (also known as chemokine ligand 2, Ccl2) in adipose tissue increases macrophage infiltration, inflammation and insulin resistance(5,6). Conversely, disruption of Mcp1 or its receptor Ccr2 impairs migration of macrophages into adipose tissue, thereby lowering adipose tissue inflammation and improving insulin sensitivity(5,7). These findings together suggest a correlation between macrophage content in adipose tissue and insulin resistance. However, resident macrophages in tissues display tremendous heterogeneity in their activities and functions, primarily reflecting their local metabolic and immune microenvironment(8). While Mcp1 directs recruitment of pro-inflammatory classically activated macrophages to sites of tissue damage(5,8), resident macrophages, such as those present in the adipose tissue of lean mice, display the alternatively activated phenotype(9). Despite their higher capacity to repair tissue(10), the precise role of alternatively activated macrophages in obesity-induced insulin resistance remains unknown. Using mice with macrophage-specific deletion of the peroxisome proliferator activated receptor-gamma (PPAR gamma), we showhere that PPAR gamma is required for maturation of alternatively activated macrophages. Disruption of PPAR gamma in myeloid cells impairs alternative macrophage activation, and predisposes these animals to development of diet-induced obesity, insulin resistance, and glucose intolerance. Furthermore, gene expression profiling revealed that downregulation of oxidative phosphorylation gene expression in skeletal muscle and liver leads to decreased insulin sensitivity in these tissues. Together, our findings suggest that resident alternatively activated macrophages have a beneficial role in regulating nutrient homeostasis and suggest that macrophage polarization towards the alternative state might be a useful strategy for treating type 2 diabetes.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available