Defining cell type-specific immune responses in a mouse model of allergic contact dermatitis by single-cell transcriptomics

Curation statements for this article:
  • Curated by eLife

    eLife logo

    eLife assessment

    This study on scRNA-seq of allergic contact dermatitis (ACD) is important in that it presents new data on fibroblasts in ACD and links to recent studies on other cell types and their signatures. The evidence presented is solid in that the data support claims of unique roles for subtypes of fibroblasts in ACD. Overall, this paper will be used as a resource by many in the skin inflammation field.

This article has been Reviewed by the following groups

Read the full article See related articles

Abstract

Allergic contact dermatitis (ACD), a prevalent inflammatory skin disease, is elicited upon repeated skin contact with protein-reactive chemicals through a complex and poorly characterized cellular network between immune cells and skin resident cells. Here, single-cell transcriptomic analysis of the murine hapten-elicited model of ACD reveals that upon elicitation of ACD, infiltrated CD4 + or CD8 + lymphocytes were primarily the IFNγ-producing type 1 effector phenotype. In contrast, type 2 cytokines (IL4 and IL13) were dominantly expressed by basophils, IL17A was primarily expressed by δγ T cells, and IL1β was identified as the primary cytokine expressed by activated neutrophils and macrophages. Furthermore, analysis of skin resident cells identified a sub-cluster of dermal fibroblasts with preadipocyte signature as a prominent target for IFNγ + lymphocytes and dermal source for key T cell chemokines CXCL9/10. IFNγ treatment shifted dermal fibroblasts from collagen-producing to CXCL9/10-producing, which promoted T cell polarization toward the type-1 phenotype through a CXCR3-dependent mechanism. Furthermore, targeted deletion of Ifngr1 in dermal fibroblasts in mice reduced Cxcl9/10 expression, dermal infiltration of CD8 + T cell, and alleviated ACD inflammation in mice. Finally, we showed that IFNγ + CD8 + T cells and CXCL10-producing dermal fibroblasts co-enriched in the dermis of human ACD skin. Together, our results define the cell type-specific immune responses in ACD, and recognize an indispensable role of dermal fibroblasts in shaping the development of type-1 skin inflammation through the IFNGR-CXCR3 signaling circuit during ACD pathogenesis.

Article activity feed

  1. eLife assessment

    This study on scRNA-seq of allergic contact dermatitis (ACD) is important in that it presents new data on fibroblasts in ACD and links to recent studies on other cell types and their signatures. The evidence presented is solid in that the data support claims of unique roles for subtypes of fibroblasts in ACD. Overall, this paper will be used as a resource by many in the skin inflammation field.

  2. Reviewer #1 (Public Review):

    Summary:

    In this manuscript, Liu et al. used scRNA-seq to characterize cell type-specific responses during allergic contact dermatitis (ACD) in a mouse model, specifically the hapten-induced DNFB model. Using the scRNA-seq data, they deconvolved the cell types responsible for the expression of major inflammatory cytokines such as IFNG (from CD4 and CD8 T cells), IL4/13 (from basophils), IL17A (from gd T cells), and IL1B from neutrophils and macrophages. They found the highest upregulation of a type 1 inflammatory response, centering around IFNG produced by CD4 and CD8 T cells. They further identified a subpopulation of dermal fibroblasts that upregulate CXCL9/10 during ACD and provided functional genetic evidence in their mouse model that disrupting IFNG signaling to fibroblasts decreases CD8 T cell infiltration and overall inflammation. They identify an increase in IFNG-expressing CD8 T cells in human patient samples of ACD vs. healthy control skin and co-localization of CD8 T cells with PDGFRA+ fibroblasts, which suggests this mechanism is relevant to human ACD. This mechanism is reminiscent of recent work (Xu et al., Nature 2022) showing that IFNG signaling in dermal fibroblasts upregulates CXCL9/10 to recruit CD8 T cells in a mouse model of vitiligo. Overall, this is a very well-presented, clear, and comprehensive manuscript. The conclusions of the study are mostly well supported by data, but some aspects of the work could be improved by additional clarification of the identity of the cell types shown to be involved, including the exact subpopulation discovered by scRNA-seq and the subtype of CD8 T cell involved. The study was limited by its use of one ACD model (DNFB), which prevents an assessment of how broadly relevant this axis is. The human sample validation is slightly circumstantial and limited by the multiplexing capacity of immunofluorescence markers.

    Strengths:

    Through deep characterization of the in vivo ACD model, the authors were able to determine which cell types were expressing the major cytokines involved in ACD inflammation, such as IFNG, IL4/13, IL17A, and IL1B. These analyses are well-presented and thoughtful, showing first that the response is IFNG-dominant, then focusing on deeper characterization of lymphocytes, myeloid cells, and fibroblasts, which are also validated and complemented by FACS experiments using canonical markers of these cell types as well as IF staining. Crosstalk analyses from the scRNA-seq data led the authors to focus on IFNG signaling fibroblasts, and in vitro experiments demonstrate that CXCL9 and CXCL10 are expressed by fibroblasts stimulated by IFNG. In vivo functional genetic evidence demonstrates an important role for IFNG signaling in fibroblasts, as KO of Ifngr1 using Pdgfra-Cre Ifngr1 fl/fl mice, showed a reduction in inflammation and CD8 T cell recruitment.

    Weaknesses:

    The use of one model limits an understanding of how broad this fibroblast-T cell axis is during ACD. However, the authors chose the most commonly employed model and cited additional work in a vitiligo model (another type 1 immune response). The identity of the involved fibroblasts and T cells in the mouse model is difficult to assess as scRNA-seq identified subpopulations of these cell types, but most work in the Pdgfra-Cre Ifngr1 fl/fl mice used broad markers for these cell types as opposed to matched subpopulation markers from their scRNA-seq data. Human patient samples of ACD were co-stained with two markers at a time, demonstrating the presence of CD8+IFNG+ T cells, PDGFRA+CXCL10+ fibroblasts, and co-localization of PDGFRA+ fibroblasts and CD8+ T cells. However, no IF staining demonstrates co-expression of all 4 markers at once; thus, the human validation of co-localization of CD8+IFNG+ T cells and PDGFRA+CXCL10+ fibroblasts is ultimately indirect, although not a huge leap of faith. Although n=3 samples of healthy control and ACD samples are used, there is no quantification of any results to demonstrate the robustness of differences.

  3. Reviewer #2 (Public Review):

    Summary:

    The investigators apply scRNA seq and bioinformatics to identify biomarkers associated with DNFB-induced contact dermatitis in mice. The bioinformatics component of the study appears reasonable and may provide new insights regarding TH1-driven immune reactions in ACD in mice. However, the IF data and images of tissue sections are not clear and should be improved to validate the model.

    Strengths:

    The bioinformatics analysis.

    Weaknesses:

    The IF data presented in 4H, 6H, 7E and 7F are not convincing and need to be correlated with routine staining on histology and different IF markers for PDGFR. Some of the IF staining data demonstrates a pattern inconsistent with its target.