Abstract

Myeloid cells play a central role in the context of viral eradication, yet precisely how these cells differentiate throughout the course of acute infections is poorly understood. In this study, we have developed a novel quantitative temporal in vivo proteomics (QTiPs) platform to capture proteomic signatures of temporally transitioning virus-driven myeloid cells directly in situ, thus taking into consideration host–virus interactions throughout the course of an infection. QTiPs, in combination with phenotypic, functional, and metabolic analyses, elucidated a pivotal role for inflammatory CD11b+, Ly6G–, Ly6Chigh-low cells in antiviral immune response and viral clearance. Most importantly, the time-resolved QTiPs data set showed the transition of CD11b+, Ly6G–, Ly6Chigh-low cells into M2-like macrophages, which displayed increased antigen-presentation capacities and bioenergetic demands late in infection. We elucidated the pivotal role of myeloid cells in virus clearance and show how these cells phenotypically, functionally, and metabolically undergo a timely transition from inflammatory to M2-like macrophages in vivo. With respect to the growing appreciation for in vivo examination of viral–host interactions and for the role of myeloid cells, this study elucidates the use of quantitative proteomics to reveal the role and response of distinct immune cell populations throughout the course of virus infection.

Highlights

  • Myeloid immune cell populations are phenotypically dynamic and arise from a common pluripotent hematopoietic stem cell lineage

  • Bone marrow (BM)-emigrating immature monocytic myeloid cells identified in mice as CD11b+, Ly6G−, Ly6C+ cells are recruited to the site of infection and mediate antimicrobial, as well as inflammatory, functions

  • Reovirus induces the accumulation of otherwise absent CD11b+, Ly6G−, Ly6Chigh cells at the site of infection as early as 1 d.p.i., which subsequently exhibited a gradual loss of Ly6C expression over time

Read more

Summary

■ INTRODUCTION

Myeloid immune cell populations are phenotypically dynamic and arise from a common pluripotent hematopoietic stem cell lineage. Through a temporal transition, these inflammatory immature myeloid cells differentiate into monocytes and macrophages that acquire specific phenotypes, functionalities, and metabolic profiles throughout infection. Such plasticity permits these myeloid cells to be associated with a plethora of pathological conditions including pathogenic infections,[1−3] inflammatory diseases/responses,[4,5] cancer progression,[6−9] and antitumor immune responses.[10]. DAF-FM diacetate (4-amino-5-methylamino2′,7′-difluorofluorescein diacetate) (D23844), and CM-H2DCFDA (C6827) were purchased from Molecular Probes (ThermoFisher Scientific, Rochford, IL, USA)

Ethics Statement
■ RESULTS
■ DISCUSSION
■ ACKNOWLEDGMENTS
■ REFERENCES
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