Abstract

The pulmonary collectins, surfactant proteins A and D (SP-A and SP-D) have been implicated in the regulation of the innate immune system within the lung. In particular, SP-D appears to have both pro- and anti-inflammatory signaling functions. At present, the molecular mechanisms involved in switching between these functions remain unclear. SP-D differs in its quaternary structure from SP-A and the other members of the collectin family, such as C1q, in that it forms large multimers held together by the N-terminal domain, rather than aligning the triple helix domains in the traditional “bunch of flowers” arrangement. There are two cysteine residues within the hydrophobic N terminus of SP-D that are critical for multimer assembly and have been proposed to be involved in stabilizing disulfide bonds. Here we show that these cysteines exist within the reduced state in dodecameric SP-D and form a specific target for S-nitrosylation both in vitro and by endogenous, pulmonary derived nitric oxide (NO) within a rodent acute lung injury model. S-nitrosylation is becoming increasingly recognized as an important post-translational modification with signaling consequences. The formation of S-nitrosothiol (SNO)-SP-D both in vivo and in vitro results in a disruption of SP-D multimers such that trimers become evident. SNO-SP-D but not SP-D, either dodecameric or trimeric, is chemoattractive for macrophages and induces p38 MAPK phosphorylation. The signaling capacity of SNO-SP-D appears to be mediated by binding to calreticulin/CD91. We propose that NO controls the dichotomous nature of this pulmonary collectin and that posttranslational modification by S-nitrosylation causes quaternary structural alterations in SP-D, causing it to switch its inflammatory signaling role. This represents new insight into both the regulation of protein function by S-nitrosylation and NO's role in innate immunity.

Highlights

  • Nitric oxide (NO) has long presented a curious dichotomy within biologic systems, namely that it is both an important physiological regulator and the mediator of many pathologies [1,2,3]

  • Cells of the lung lining secrete a microbe-binding molecule called surfactant protein D (SP-D) that helps activate the inflammatory system against invading pathogens

  • SP-D is held together in its multimeric state by interacting cysteine residues, which are susceptible to modification by the gaseous second messenger, nitric oxide, to form S-nitrosothiols

Read more

Summary

Introduction

Nitric oxide (NO) has long presented a curious dichotomy within biologic systems, namely that it is both an important physiological regulator and the mediator of many pathologies [1,2,3]. Nowhere is this more clearly demonstrated than within the pulmonary system. Of particular interest is the capability of NO to induce signaling functions via the post-translational modification of proteins [10,11,12]. The potential for SNO as a post-translational regulator of protein function has been highlighted by recent proteomic and targeted studies [16,17,18]

Methods
Results
Discussion
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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.