Abstract

The mitogen-activated protein kinase (MAPK) cascades are activated in innate immune cells such as macrophages upon the detection of microbial infection, critically regulating the expression of proinflammatory cytokines and chemokines such as TNF-α, IL-6, and MCP-1. As a result, activation of MAPKs is tightly regulated to ensure appropriate and adequate immune responses. Dual-specificity phosphatases (DUSPs) are a family of proteins which specifically dephosphorylates threonine and tyrosine residues essential for MAPK activation to negatively regulate their activation. DUSP12 is a member of atypical DUSPs that lack MAPK-binding domain. Its substrate and function in immune cells are unknown. In this study, we demonstrated that DUSP12 is able to interact with all the three groups of MAPKs, including extracellular signal-regulated protein kinase, JNK, and p38. To investigate the function of DUSP12 in macrophages in response to TLR activation and microbial infection, we established RAW264.7 cell lines stably overexpressing DUSP12 and found that overexpression of DUSP12 inhibited proinflammatory cytokine and chemokine production in response to TLR4 activation, heat-inactivated Mycobacterium tuberculosis stimulation as well as infections by intracellular bacteria including Listeria moncytogenesis and Mycobacterium bovis BCG by specifically inhibiting p38 and JNK. In addition, a scaffold protein known as signal transducing adaptor protein 2 (STAP2), was found to mediate the interaction between DUSP12 and p38. Thus, DUSP12 is a bona fide MAPK phosphatase, playing an important role in MAPK-regulated responses to bacterial infection. Our study provides a model where atypical DUSPs regulate MAPKs via scaffold, thereby regulating immune responses to microbial infection.

Highlights

  • The innate immune system is the first line of defense against microbe infection

  • We demonstrated that dual-specificity phosphatase 12 (DUSP12) interacts with all three groups of mitogen-activated protein kinase (MAPK) in HEK293T cells despite the lack of MAPK binding domain (MKB) and the interaction between DUSP12 and p38 was mediated by signal transducing adaptor protein 2 (STAP2)

  • We found that LPS stimulation resulted in the upregulation of DUSP12 expression in macrophages (Figure 1B), the upregulation appeared to be weaker as compared to that of MKP5

Read more

Summary

Introduction

The innate immune system is the first line of defense against microbe infection. It responses rapidly upon pathogen recognition and triggering inflammation. The pathogen-associated molecular patterns (PAMPs) derived from invading pathogens can be recognized by a family of transmembrane pattern recognition receptors (PRRs) called Toll-like receptors (TLRs) located on the cellular. Pathogens may present various ligands which can be recognized by multiple TLRs. For instance, Listeria monocytogenes, a gram-positive intracellular bacterium, expresses a myriad of TLR ligands, including peptidoglycan, flagellin, and bacterial DNA. Recognition of peptidoglycan by a heterodimer of TLR2 and TLR6, lipoteichoic acid by TLR4, flagellin by TLR5, and bacterial DNA by TLR9 lead to the detection of invading L. monocytogenes by innate immune cells and activate the overall immune response [3,4,5]

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