Abstract

MitoNEET (mitochondrial protein containing Asn–Glu–Glu–Thr (NEET) sequence) is a 2Fe–2S cluster-containing integral membrane protein that resides in the mitochondrial outer membrane and participates in a redox-sensitive signaling and Fe–S cluster transfer. Thus, mitoNEET is a key regulator of mitochondrial oxidative capacity and iron homeostasis. Moreover, mitochondrial dysfunction and oxidative stress play critical roles in inflammatory diseases such as sepsis. Increased iron levels mediated by mitochondrial dysfunction lead to oxidative damage and generation of reactive oxygen species (ROS). Increasing evidence suggests that targeting mitoNEET to reverse mitochondrial dysfunction deserves further investigation. However, the role of mitoNEET in inflammatory diseases is unknown. Here, we investigated the mechanism of action and function of mitoNEET during lipopolysaccharide (LPS)-induced inflammatory responses in vitro and in vivo. Levels of mitoNEET protein increased during microbial or LPS-induced sepsis. Pharmacological inhibition of mitoNEET using mitoNEET ligand-1 (NL-1) decreased the levels of pro-inflammatory cytokines such as IL-1β, IL-6, and TNF-α in animal models of sepsis, as well as LPS-induced inflammatory responses by macrophages in vitro. Inhibition of mitoNEET using NL-1 or mitoNEET shRNA abrogated LPS-induced ROS formation and mitochondrial dysfunction. Furthermore, mitochondrial iron accumulation led to generation of LPS-induced ROS, a process blocked by NL-1 or shRNA. Taken together, these data suggest that mitoNEET could be a key therapeutic molecule that targets mitochondrial dysfunction during inflammatory diseases and sepsis.

Highlights

  • Inflammation is critical for healing, but uncontrolled and dysregulated inflammation can increase the risk of developing various diseases [1]

  • Prior studies show that mitoNEET, an outer mitochondrial membrane protein, plays an important role in regulating mitochondrial function, especially oxidative capacity [25, 31, 35]

  • We hypothesized that mitoNEET plays a role in inflammation and oxidative stress during sepsis

Read more

Summary

Introduction

Inflammation is critical for healing, but uncontrolled and dysregulated inflammation can increase the risk of developing various diseases [1]. The link between sepsisassociated organ failure and mitochondrial dysfunction is increasing interest to researchers [3]. Sepsis-induced mitochondrial dysfunction mediates hyperinflammation through cellular metabolic disorders, insufficient energy production, and oxidative stress; as such, it plays a key role in the development of sepsisrelated multiorgan failure [4,5,6]. Increased mitochondrial iron accumulation due to pro-inflammatory signaling promotes oxidative damage by catalyzing generation of ROS and causing mitochondrial dysfunction [9, 11]. These processes develop into a vicious inflammatory cycle [12]. Several studies demonstrate that targeting mitochondrial iron accumulation using iron chelators has the potential to improve the prognosis of sepsis [8, 13, 14]

Methods
Results
Conclusion
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