Abstract

The Nod-like receptor 3 (NLRP3) inflammasome is an intracellular sensor that sets off the innate immune system in response to microbial-derived and endogenous metabolic danger signals. We previously reported that γ-tocotrienol (γT3) attenuated adipose tissue inflammation and insulin resistance in diet-induced obesity, but the underlying mechanism remained elusive. Here, we investigated the effects of γT3 on NLRP3 inflammasome activation and attendant consequences on type 2 diabetes. γT3 repressed inflammasome activation, caspase-1 cleavage, and interleukin (IL) 1β secretion in murine macrophages, implicating the inhibition of NLRP3 inflammasome in the anti-inflammatory and antipyroptotic properties of γT3. Furthermore, supplementation of leptin-receptor KO mice with γT3 attenuated immune cell infiltration into adipose tissue, decreased circulating IL-18 levels, preserved pancreatic β-cells, and improved insulin sensitivity. Mechanistically, γT3 regulated the NLRP3 inflammasome via a two-pronged mechanism: 1) the induction of A20/TNF-α interacting protein 3 leading to the inhibition of the TNF receptor-associated factor 6/nuclear factor κB pathway and 2) the activation of AMP-activated protein kinase/autophagy axis leading to the attenuation of caspase-1 cleavage. Collectively, we demonstrated, for the first time, that γT3 inhibits the NLRP3 inflammasome thereby delaying the progression of type 2 diabetes. This study also provides an insight into the novel therapeutic values of γT3 for treating NLRP3 inflammasome-associated chronic diseases.

Highlights

  • The Nod-like receptor 3 (NLRP3) inflammasome is an intracellular sensor that sets off the innate immune system in response to microbial-derived and endogenous metabolic danger signals

  • We investigated the role of ␥T3 in autophagy in peritoneal macrophages of db/db mice. p-AMPactivated protein kinase (AMPK)␣ (Thr 172) as well as markers of autophagic activation, including Beclin-1, autophagy protein 5 (ATG5), and LC3II, were markedly increased by ␥T3, whereas p62 was decreased by ␥T3 (Fig. 4E)

  • The activation of the NOD-like receptor protein 3 (NLRP3) inflammasome is positively associated with the prevalence of type 2 diabetes

Read more

Summary

RESEARCH DESIGN AND METHODS

Chemicals and reagents ␥T3 (with 90% purity) was kindly provided by Carotech. ␥T3 was prepared as previously described [18]. Primary bone marrow cells were isolated from the femurs of 6- to 10-week-old C57BL/6 mice and stimulated to differentiate for 7–10 days in L-cell conditioned medium (CM) as we described previously [5]. The resulting differentiated bone marrow-derived macrophages (BMDMs) were pretreated with ␥T3 or vehicle (DMSO) for 24 h, primed with lipopolysaccharide (LPS) (100 ng/ml) for 1 h, and stimulated either with nigericin (Ng; 6.5 ␮M, a K+/H+ ionophore) for 1 h or palmitate (PA; 400 ␮M complexed with BSA) for 12 h. Pro-IL-1␤-Gaussia luciferase reporter assay The J774 macrophages stably expressing pro-IL-1␤-Gaussia luciferase (iGLuc) fusion construct (Fig. 1A) were a generous gift. ␥T3 suppressed NLRP3 inflammasome procaspase reporter activity in iJ774 macrophages. A: Structure of the iGLuc (NLRP3 inflammasome and caspase activity reporter constructs). The cleavage product of the pro-IL1␤-iGLuc fusion protein was detected in the media by Western blot analysis using anti-GLuc antibody (NEB Inc.)

Glucose and insulin tolerance tests
Isolation of stromal vascular cells and flow cytometric analysis
RESULTS
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