Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2024 Jun 11:13:e85914.
doi: 10.7554/eLife.85914.

The interferon-rich skin environment regulates Langerhans cell ADAM17 to promote photosensitivity in lupus

Affiliations

The interferon-rich skin environment regulates Langerhans cell ADAM17 to promote photosensitivity in lupus

Thomas Morgan Li et al. Elife. .

Abstract

The autoimmune disease lupus erythematosus (lupus) is characterized by photosensitivity, where even ambient ultraviolet radiation (UVR) exposure can lead to development of inflammatory skin lesions. We have previously shown that Langerhans cells (LCs) limit keratinocyte apoptosis and photosensitivity via a disintegrin and metalloprotease 17 (ADAM17)-mediated release of epidermal growth factor receptor (EGFR) ligands and that LC ADAM17 sheddase activity is reduced in lupus. Here, we sought to understand how the lupus skin environment contributes to LC ADAM17 dysfunction and, in the process, differentiate between effects on LC ADAM17 sheddase function, LC ADAM17 expression, and LC numbers. We show through transcriptomic analysis a shared IFN-rich environment in non-lesional skin across human lupus and three murine models: MRL/lpr, B6.Sle1yaa, and imiquimod (IMQ) mice. IFN-I inhibits LC ADAM17 sheddase activity in murine and human LCs, and IFNAR blockade in lupus model mice restores LC ADAM17 sheddase activity, all without consistent effects on LC ADAM17 protein expression or LC numbers. Anti-IFNAR-mediated LC ADAM17 sheddase function restoration is associated with reduced photosensitive responses that are dependent on EGFR signaling and LC ADAM17. Reactive oxygen species (ROS) is a known mediator of ADAM17 activity; we show that UVR-induced LC ROS production is reduced in lupus model mice, restored by anti-IFNAR, and is cytoplasmic in origin. Our findings suggest that IFN-I promotes photosensitivity at least in part by inhibiting UVR-induced LC ADAM17 sheddase function and raise the possibility that anifrolumab ameliorates lupus skin disease in part by restoring this function. This work provides insight into IFN-I-mediated disease mechanisms, LC regulation, and a potential mechanism of action for anifrolumab in lupus.

Keywords: ADAM17; human; immunology; inflammation; interferons; langerhans cells; lupus; mouse; photosensitivity.

PubMed Disclaimer

Conflict of interest statement

TL, VZ, ES, MD, YC, KV, DO, PC, YL, WS, ST, KO, MR, IR No competing interests declared, AD, PL is an employee of AMPEL BioSolutions, but has no financial conflicts of interest to report, NS was awarded the Lupus Therapeutics: The Clinical Trial Network Infrastructure Grant, received by The Albert Einstein College of Medicine. The author received payment for lectures at the Congress of Clinical Rheumatology East and the Congress of Clinical Rheumatology West. The author has no other competing interests to declare, JL has received the grants F31 NIH GM136144 and T32 NIH GM008539. The author has received stock or stock options from NASDAQ/NYSE Ticker: FULC, ABCL, AVXL, VOR, MRNA, BNTX, SAVA, OCGN, CTMX, BCEL, GE. The author has no other competing interests to declare, WA received support for travel and attending Lupus 21st century meeting in 2021. The author has no other competing interests to declare, JZ received a grant from NIH NIAMS, and consulting fees from Hoth Therapeutics and Pfizer. The author received payment for participation on a Data Safety Monitoring Board/ Advisory Board for Hoth Therapeutics and acts as President elect for PASPCR. The author holds stock options from Hoth Therapeutics, FoxWayne Inc and YouV labs. The author has no other competing interests to declare, JK has received grant support from AbbVie, Akros, Allergan, Amgen, Avillion, Biogen, Botanix, Boehringer Ingelheim, Bristol-Myers Squibb, Exicure, Innovaderm, Incyte, Janssen, Kyowa Kirin, Lilly, Nimbus Lackshmi, Novan, Novartis, PAREXEL, Pfizer, Regeneron, UCB, Vitae Pharmaceuticals. The author received consulting fees from AbbVie, Aclaris, Allergan, Almirall, Amgen, Artax Biopharma, Arena, Aristea, Asana, Aurigene, Biogen Idec, Boehringer Ingelheim, Bristol-Myers Squibb, Escalier, Galapagos, Janssen, Kyowa Kirin, Lilly, MoonLake Immunotherapeutics, Nimbus, Novartis, Pfizer, Sanofi, Sienna Biopharmaceuticals, Sun Pharma, Target-Derm, UCB, Valeant, Ventyx. The author has no other competing interests to declare, NA has received the following grants: NIAMS AR080436-01, NIAMS R56AR078686-01 and NIH NIAMS 5R01 GRANT AR070234-05. The author received consulting fees from Immunitas, Shennon Bio and Janssen. The author received payment as a lecturer from 23 and me, Cellino and Bristol Meyer Squibb Genomics. They are also a board member of the Society of Investigative Dermatology. The author has no other competing interests to declare, AJ has received grants from the NIH and the VA and consulting fees from Pfizer. The author has no other competing interests to declare, CB The patent number is US10024844B2 and the title of the patent is "Identification of an inhibitor of iRhom1 or an inhibitor of iRhom2", which is also what the patent relates to. Carl Blobel and the Hospital for Special Surgery have identified iRhom2 inhibitors and have co-founded the start-up company SciRhom in Munich to commercialize these inhibitors, TL has received the following grants: NIH R01AI079178, NIH R21 AR081493, Department of Defense W81XWH-21-LRP-IPA, Lupus Research Alliance Lupus Innovation Award grant, Barbara Volcker Center for Women and Rheumatic Diseases grant. She has also received funding support from the St. Giles Foundation and A Lasting Mark Foundation. She has received consulting fees from Pfizer, and has a received payment from Bristol Meyers Squibb for giving a lecture. The author has received payment for attending Lupus 21st Century meeting. The author has no other competing interests to declare

Figures

Figure 1.
Figure 1.. Analysis of a new cohort shows an IFN-rich environment in non-lesional DLE skin.
(A–H) Microarray analysis of gene expression from non-lesional skin of DLE (n=7, this manuscript), lesional DLE (n=7), psoriasis patients (n=17), and healthy controls (n=13) (Jabbari et al., 2014). In (A, D–F), analyses include lesional DLE or both lesional and psoriasis samples. (A) Principal component analysis (PCA) of patient samples using top 500 genes. (B) Differentially expressed pathways in control and non-lesional DLE skin were determined using QuSAGE pathway analysis against Molecular Signatures Database (MSigDB). (C) Volcano plot of differentially expressed genes. Genes from IFN-α/β (red), IFN-γ (blue) pathways, and IRF transcription factors (green) are marked. (D) Heatmap of z-score transformed gene expression in the IFN-α/β signaling pathway. (E–H) Gene Set Variation Analysis (GSVA) of gene sets relevant to lupus (Martínez et al., 2022), with (G–H) comparing only control and non-lesional skin. (F, H) GSVA using gene sets comprising specific IFN subtypes. (E–H) *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001 by unpaired t-test.
Figure 1—figure supplement 1.
Figure 1—figure supplement 1.. Further analysis of gene expression in human DLE.
Log-transformed expression fold change for genes in the IFN α/β pathway for human DLE skin.
Figure 2.
Figure 2.. Three photosensitive SLE models show upregulated IFN-related gene expression in non-lesional skin by QuSAGE analysis.
(A–I) RNAseq analysis of gene expression and pathway analyses from MRL/lpr (LPR) and control MRL/+ (MRL) (A–D), B6.Sle1yaa and control B6 mice (E), and IMQ and control B6 mice (F–I). For the IMQ model, mice were painted on one ear (ipsilateral) and the unpainted ear (contralateral) was taken as the non-lesional ear. (A, E, F) PCA using top 500 genes. (B, G) Differentially expressed pathways determined by QuSAGE pathway analysis against MSigDB. (C, H) Volcano plot of differentially expressed genes. Genes from IFN-α/β (red), IFN-γ (blue), and IRF transcription factor (green) pathways are marked. (D, I) Heatmap of z-score transformed gene expression in the IFN α/β signaling pathway. (A, E, F) Each symbol represents one mouse.
Figure 2—figure supplement 1.
Figure 2—figure supplement 1.. Further analysis of gene expression in skin of multiple murine lupus models.
(A–B) Log-transformed expression fold change for genes in the IFN α/β pathway for skin of MRL/lpr (A) and IMQ (B) model mice. (C) Genes from yaa locus are expressed at a higher levels in B6.Sle1yaa mice. (D) IRF transcription factors and their targets are expressed at a higher levels and CD207 is expressed at lower level in B6.Sle1yaa mice compared to controls.
Figure 3.
Figure 3.. The SLE models share upregulated IFN signatures in non-lesional skin by gene set variation analysis (GSVA).
(A–F) The RNAseq data from Figure 2 of MRL/lpr (A–B), B6.Sle1yaa (C–D), and IMQ (E–F) models were analyzed by GSVA. (A, C, E) GSVA of gene sets relevant to lupus, adapted for murine models (Kingsmore et al., 2021). (B, D, F) GSVA of gene signatures specific to distinct IFN subtypes. (A–F) Each symbol represents one mouse. *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001 by unpaired t-test.
Figure 3—figure supplement 1.
Figure 3—figure supplement 1.. GSVA of ipsilateral IMQ-painted skin together with contralateral ear skin.
GSVA of skin from ipsilateral IMQ painted ears and contralateral unpainted “non-lesional” ear of IMQ-treated mice.
Figure 4.
Figure 4.. Comparison of human DLE and murine lupus models shows shared upregulation of IFN-associated genes.
(A) Venn diagram of differentially expressed genes among DLE patients, LPR mice, and IMQ mice using FDR <0.05 and logFC >0.5. (B) Heatmap of GSVA scores for shared gene sets between non-lesional skin from DLE patients, LPR mice, B6.Sle1yaa mice, and IMQ mice. Asterisks indicate significant differences in GSVA scores compared to controls for each dataset.
Figure 5.
Figure 5.. IFN-I inhibits LC ADAM17 sheddase activity.
(A) Epidermal cell suspensions from WT and LCAd17 mice lacking ADAM17 in LCs were assayed for LC ADAM17 sheddase activity as indicated by the extent of UVR-induced cell surface TNFR1 loss. Percent change in cell surface TNFR1 mean fluorescence intensity (MFI) after UVR (left); relative LC ADAM17 activity calculated by normalizing TNFR1 loss to that of vehicle controls (right). (B) Representative histograms from cell surface ADAM17 staining of WT and LCAd17 epidermal cell suspensions. assayed. (C–P) Murine and human epidermal cell suspensions were treated ex vivo or mice were treated in vivo with IFN-I prior to assaying for UVR-induced LC ADAM17 sheddase activity (C, F, I, K, N), LC ADAM17 levels (D, G, L, O), and LC numbers (E, H, J, M, P). (C–J) Cells from WT mice were treated with IFN-κ or vehicle (C–E), with or without tofacitinib (F–H), or with IFN-βor vehicle (I–J). (K–M) IFN-κ or vehicle was applied topically to ears of WT mice 16–20 hr prior to examination. (N–P) Suction blister epidermal cell suspensions from healthy human donors were treated with IFN-β or vehicle. In (N), lines connect samples from the same donor. (A,C–P) Each symbol represents cells from a single mouse or donor, bars represent average values, and error bars are SD. n=3–10 per condition over 3–5 independent experiments. *p<0.05, **p<0.01, ***p<0.001, n.s.=not significant by paired (A, C, F, I, K, N (left)) or unpaired (A, C, F, I, K, N (right)), (D–E, G–H, J, L–M, O–P) t-test.
Figure 5—figure supplement 1.
Figure 5—figure supplement 1.. Langerhans cell yield and gating with suction blistering of human skin.
(A) Schematic of suction blistering and sample collection. (B) Gating of Langerhans cells in blister. fluid and in epidermal roof cell suspension. (C) Langerhans cell abundance in blister fluid and. epidermal roof samples. Each symbol represents 1 healthy donor. ****p<0.0001 unpaired t-test.
Figure 6.
Figure 6.. Anti-IFNAR restores LC ADAM17 activity in multiple lupus models.
(A–I) MRL/lpr (A–D), B6.Sle1yaa (E–F), and IMQ (G–I) lupus model mice and their controls were treated twice with anti-IFNAR or isotype control at indicated doses over 6 days prior to collection of non-lesional epidermal cells. (A, C, E, G) UVR-induced LC ADAM17 sheddase activity as in Figure 5. (B, D, F, H) Relative cell surface ADAM17 levels. (I) Relative LC numbers. (A–I) Each symbol represents a mouse, bars represent average values, and error bars are SD. n=3–9 per condition over 3–8 independent experiments. *p<0.05, **p<0.01, ***p<0.001, n.s.=not significant by paired (A, C, E, G (left)) or unpaired (A, C, E, G (right)), (B, D, F, H–I) t-test.
Figure 7.
Figure 7.. Anti-IFNAR reduces photosensitivity in EGFR- and LC ADAM17- dependent manners.
(A–L) MRL/lpr (A–I) and LCAd17 (J–L) mice and controls were treated according to schematics in (A, G, J) and non-lesional ears were harvested. (B, D, H) Ear thickness. (C, E, I) Epidermal permeability as indicated by toluidine blue retention. (F, K) Neutrophils and monocytes, and (L) LCs per ear. (B–F, H–I, K–L) n=4–10 per condition over four to six independent experiments. *p<0.05, **p<0.01, ***p<0.001, n.s.=not significant by paired (B, D, H) and unpaired (C, E, F, I, K–L) t-test.
Figure 8.
Figure 8.. Anti-IFNAR restores UVR-induced LC ROS expression in a lupus model and UVR stimulates cytoplasmic ROS.
(A) Epidermal cell suspensions from control or IMQ mice treated with IgG or anti-IFNAR were exposed to UVR. Cells were loaded with the general ROS indicator CM-H2DCFDA prior to staining for LC markers for flow cytometry analysis. Representative histograms (left), CM-H2DCFDA signal MFI (middle), and fold change with UVR exposure (right). (B–C) Healthy B6 epidermal cells were loaded with the cytoplasmic ROS indicator CellROX (B) or mitochondrial ROS indicator MitoSOX (C) prior to UVR exposure and staining of LC markers. Signal was calculated by dividing the ROS indicator MFI by the MFI of its respective negative control. Each symbol represents one mouse, bars represent average values, and error bars are SD. n=5–6 per condition over four to five experiments. *p<0.05, **p<0.01, n.s.=not significant by paired A (left), (B, C) and unpaired (A (right)) t-test.

Update of

  • doi: 10.1101/2021.08.18.456792

References

    1. Allen ME, Rus V, Szeto GL. Leveraging heterogeneity in systemic lupus erythematosus for new therapies. Trends in Molecular Medicine. 2021;27:152–171. doi: 10.1016/j.molmed.2020.09.009. - DOI - PMC - PubMed
    1. Ambler WG, Howlander M, Chalasani MLS, Seltzer ES, Sim J, Shin J, Schwartz N, Shipman WD, Dasoveanu D, Carballo CB, Sevim E, Siddique S, Rodeo S, Erkan D, Kataru RP, Mehrara B, Lu TT. Lymphatic Dysfunction in Lupus Contributes to Cutaneous Photosensitivity and Lymph Node B Cell Responses. bioRxiv. 2022 doi: 10.1101/2022.06.13.495930. - DOI
    1. Bachen EA, Chesney MA, Criswell LA. Prevalence of mood and anxiety disorders in women with systemic lupus erythematosus. Arthritis and Rheumatism. 2009;61:822–829. doi: 10.1002/art.24519. - DOI - PMC - PubMed
    1. Baechler EC, Batliwalla FM, Karypis G, Gaffney PM, Ortmann WA, Espe KJ, Shark KB, Grande WJ, Hughes KM, Kapur V, Gregersen PK, Behrens TW. Interferon-inducible gene expression signature in peripheral blood cells of patients with severe lupus. PNAS. 2003;100:2610–2615. doi: 10.1073/pnas.0337679100. - DOI - PMC - PubMed
    1. Benci JL, Johnson LR, Choa R, Xu Y, Qiu J, Zhou Z, Xu B, Ye D, Nathanson KL, June CH, Wherry EJ, Zhang NR, Ishwaran H, Hellmann MD, Wolchok JD, Kambayashi T, Minn AJ. Opposing functions of interferon coordinate adaptive and innate immune responses to cancer immune checkpoint blockade. Cell. 2019;178:933–948. doi: 10.1016/j.cell.2019.07.019. - DOI - PMC - PubMed

Associated data