Abstract

A major immunological response during neuroinflammation is the activation of microglia, which subsequently release proinflammatory mediators such as prostaglandin E(2) (PGE(2)). Besides its proinflammatory properties, cyclooxygenase-2 (COX-2)-derived PGE(2) has been shown to exhibit anti-inflammatory effects on innate immune responses. Here, we investigated the role of microsomal PGE(2) synthase-1 (mPGES-1), which is functionally coupled to COX-2, in immune responses using a model of lipopolysaccharide (LPS)-induced spinal neuroinflammation. Interestingly, we found that activation of E-prostanoid (EP)2 and EP4 receptors, but not EP1, EP3, PGI(2) receptor (IP), thromboxane A(2) receptor (TP), PGD(2) receptor (DP), and PGF(2) receptor (FP), efficiently blocked LPS-induced tumor necrosis factor α (TNFα) synthesis and COX-2 and mPGES-1 induction as well as prostaglandin synthesis in spinal cultures. In vivo, spinal EP2 receptors were up-regulated in microglia in response to intrathecally injected LPS. Accordingly, LPS priming reduced spinal synthesis of TNFα, interleukin 1β (IL-1β), and prostaglandins in response to a second intrathecal LPS injection. Importantly, this reduction was only seen in wild-type but not in mPGES-1-deficient mice. Furthermore, intrathecal application of EP2 and EP4 agonists as well as genetic deletion of EP2 significantly reduced spinal TNFα and IL-1β synthesis in mPGES-1 knock-out mice after LPS priming. These data suggest that initial inflammation prepares the spinal cord for a negative feedback regulation by mPGES-1-derived PGE(2) followed by EP2 activation, which limits the synthesis of inflammatory mediators during chronic inflammation. Thus, our data suggest a role of mPGES-1-derived PGE(2) in resolution of neuroinflammation.

Highlights

  • Neurodegenerative disorders, including Alzheimer and Parkinson disease, multiple sclerosis, and spinal cord or peripheral nerve injury, are associated with neuroinflammation [1, 2]

  • We used the well described model of chronic neuroinflammation by repetitive spinal LPS injection. In this model, where EP2 expression was up-regulated in microglia, we investigated the pro- and anti-inflammatory properties of PGE2 in vivo as well as the roles of COX-2 and microsomal PGE2 synthase-1 (mPGES-1) during termination of innate immune responses

  • We found that PGE2 attenuates LPS-induced TNF␣ synthesis in the spinal cord via EP2 receptors accompanied by a negative feedback inhibition on COX-2 expression

Read more

Summary

Introduction

Neurodegenerative disorders, including Alzheimer and Parkinson disease, multiple sclerosis, and spinal cord or peripheral nerve injury, are associated with neuroinflammation [1, 2]. We found that PGE2 attenuates LPS-induced TNF␣ synthesis in the spinal cord via EP2 receptors accompanied by a negative feedback inhibition on COX-2 expression. PGE2 Is a Negative Feedback Regulator during Spinal LPSinduced Immune Responses—TNF␣ induces spinal COX-2 and mPGES-1 expression as well as prostanoid synthesis in vivo and in vitro in primary spinal cord cultures [6, 7].

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