Abstract

Indomethacin (Indo), a nonsteroidal antiinflammatory drug, has been shown to promote murine brown adipogenesis both in vitro and in vivo, possibly due to its peroxisome proliferator‐activated receptor gamma (PPARγ)‐agonist activities. However, it is unclear whether Indo induces browning of white adipocytes from both murine and human origins or induces human brown adipogenesis. To bridge the gap, this study investigated the effects of increasing concentrations of Indo on murine 3T3‐L1, human primary subcutaneous white adipocytes (HPsubQ), and human brown (HBr) adipocytes. The results show that Indo dose‐dependently enhanced 3T3‐L1 adipocyte differentiation and upregulated both mRNA and protein expression of brown and beige adipocyte markers, while simultaneously suppressing white adipocyte‐specific marker mRNA expression. mRNA and protein expression of mitochondrial biogenesis and structural genes were dose‐dependently enhanced in Indo treated 3T3‐L1 adipocytes. This was accompanied by augmented mitochondrial DNA, enhanced oxygen consumption, proton leak, and maximal and spare respiratory capacity. Dose‐dependent transactivation of PPARγ confirmed Indo's PPARγ‐agonist activity in 3T3‐L1 cells. Knockdown of PPARγ significantly attenuated Indo's activities in selective browning genes, demonstrating PPARγ dependence of these effects. Moreover, Indo enhanced mRNA and protein expression of brown markers in HPsubQ adipocytes. Interestingly, Indo‐induced differential effects on individual PPARγ isoforms with significant dose‐dependent induction of PPARγ‐2 and suppression of PPARγ‐1 protein expression. Finally, Indo significantly promoted brown adipogenesis in HBr cells. Taken together, these results demonstrate Indo to be a potent thermogenic compound in both murine and human fat cells and may be explored as a therapeutic agent for obesity treatment and prevention.

Highlights

  • Energy-dense diets and sedentary lifestyles are primary contributing factors to the still-growing obesity epidemic facing the United States and worldwide.[1,2] Obesity-related conditions such as diabetes, heart disease, and cancer are among the leading causes of preventable deaths in America.[1]

  • Our findings that Indo activates PPARγ and promotes 3T3-L1 adipocyte differentiation are consistent with previous reports demonstrating that TZDs and Indo promote white adipocyte differentiation in vitro and in vivo.[15,21,28,29]

  • We show that Indo significantly induced browning in 3T3-L1 adipocytes as shown by enhanced uncoupling protein 1 (UCP1) mRNA and protein expression as well as beige fat-specific marker, Cd137, with simultaneous suppression of white fat-specific marker, Igfbp[3]

Read more

Summary

| INTRODUCTION

Energy-dense diets and sedentary lifestyles are primary contributing factors to the still-growing obesity epidemic facing the United States and worldwide.[1,2] Obesity-related conditions such as diabetes, heart disease, and cancer are among the leading causes of preventable deaths in America.[1] Advocating for improved diet and lifestyle factors while increasing physical activity is always the first line of treatment. These pursuits are difficult to maintain and may not be feasible for ill or disabled individuals. To bridge the gap, the current study investigated the effects of Indo on differentiating murine and human white fat cells and human brown fat cells

| MATERIALS AND METHODS
Findings
| DISCUSSION
Full Text
Published version (Free)

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