Abstract

BackgroundMurine monocytes (MC) are classified into Ly6Chigh and Ly6Clow MC. Ly6Chigh MC is the pro-inflammatory subset and the counterpart of human CD14++CD16+ intermediate MC which contributes to systemic and tissue inflammation in various metabolic disorders, including hyperhomocysteinemia (HHcy). This study aims to explore molecule signaling mediating MC subset differentiation in HHcy and control mice.MethodsRNA-seq was performed in blood Ly6Chigh and Ly6Clow MC sorted by flow cytometry from control and HHcy cystathionine β-synthase gene-deficient (Cbs -/-) mice. Transcriptome data were analyzed by comparing Ly6Chigh vs. Ly6Clow in control mice, Ly6Chigh vs. Ly6Clow in Cbs-/- mice, Cbs-/- Ly6Chigh vs. control Ly6Chigh MC and Cbs-/- Ly6Clow vs. control Ly6Clow MC by using intensive bioinformatic strategies. Significantly differentially expressed (SDE) immunological genes and transcription factor (TF) were selected for functional pathways and transcriptional signaling identification.ResultsA total of 7,928 SDE genes and 46 canonical pathways derived from it were identified. Ly6Chigh MC exhibited activated neutrophil degranulation, lysosome, cytokine production/receptor interaction and myeloid cell activation pathways, and Ly6Clow MC presented features of lymphocyte immunity pathways in both mice. Twenty-four potential transcriptional regulatory pathways were identified based on SDE TFs matched with their corresponding SDE immunological genes. Ly6Chigh MC presented downregulated co-stimulatory receptors (CD2, GITR, and TIM1) which direct immune cell proliferation, and upregulated co-stimulatory ligands (LIGHT and SEMA4A) which trigger antigen priming and differentiation. Ly6Chigh MC expressed higher levels of macrophage (MΦ) markers, whereas, Ly6Clow MC highly expressed lymphocyte markers in both mice. HHcy in Cbs -/- mice reinforced inflammatory features in Ly6Chigh MC by upregulating inflammatory TFs (Ets1 and Tbx21) and strengthened lymphocytes functional adaptation in Ly6Clow MC by increased expression of CD3, DR3, ICOS, and Fos. Finally, we established 3 groups of transcriptional models to describe Ly6Chigh to Ly6Clow MC subset differentiation, immune checkpoint regulation, Ly6Chigh MC to MΦ subset differentiation and Ly6Clow MC to lymphocyte functional adaptation.ConclusionsLy6Chigh MC displayed enriched inflammatory pathways and favored to be differentiated into MΦ. Ly6Clow MC manifested activated T-cell signaling pathways and potentially can adapt the function of lymphocytes. HHcy reinforced inflammatory feature in Ly6Chigh MC and strengthened lymphocytes functional adaptation in Ly6Clow MC.

Highlights

  • Monocytes (MC) are bone marrow (BM) derived mononuclear phagocytes that play an important role in innate immune response and are the major immune cell population in chronic tissue inflammatory [1, 2]

  • We identified 2641 secretome, 1176 cytokines and 377 surface markers collected in Protein Atlas [45] and 49 immune checkpoint gene based on the current literature [25], and newly suggested leukocyte signature genes from recent scRNA-seq study [46, 47]

  • Principle components analysis (PCA) presented a clear separation between Ly6Chigh and Ly6Clow in both control and Cbs-/- samples (Figure 2D)

Read more

Summary

Introduction

Monocytes (MC) are bone marrow (BM) derived mononuclear phagocytes that play an important role in innate immune response and are the major immune cell population in chronic tissue inflammatory [1, 2]. Murine Ly6Chigh and Ly6Cmiddle MC subsets perform pro-inflammatory functions, which are considered the counterpart of human CD14++ CD16+ intermediate MC or CD14+CD40+ inflammatory MC [4, 5]. The Abbreviations: MC, Monocyte; HHcy, Hyperhomocysteinemia; Cbs-/-, Cystathionine b-synthase gene-deficient; SDE, Significantly differentially expressed; TF, Transcription factor; Ly6C, Lymphocyte antigen 6 complex, locus C; BM, Bone marrow; DC, Dendritic cells; Mf, Macrophages; NK, Natural killer; IL, Interleukin; MCP, Monocyte chemoattractant protein; CD, Cluster of differentiation; RNA-seq, RNA sequencing; scRNA-seq, Single-cell RNA sequencing; Tg-hCBS, Human CBS transgenic; FACS, Fluorescenceactivated cell sorting; PBS, Phosphate‐buffered saline; EDTA, Ethylenediaminetetraacetic acid; FC, Fold change; PCA, Principle components analysis; PC, Principal components; IPA, Ingenuity Pathway Analysis; IFN-I, Type I interferon; PAMP, Pathogen-associated molecular pattern; DAMP, Damageassociated molecular pattern; MADS, Metabolic-associated danger signals; Th, T helper; Tfh, T follicular helper; T-reg, Regulatory T; ME, Myelo-erythroid. This study aims to explore molecule signaling mediating MC subset differentiation in HHcy and control mice

Objectives
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