Hedgehog interacting protein-expressing lung fibroblasts suppress lymphocytic inflammation in mice

Jeong H Yun, ChangHee Lee, Tao Liu, Siqi Liu, Edy Y Kim, Shuang Xu, Jeffrey L Curtis, Luca Pinello, Russell P Bowler, Edwin K Silverman, Craig P Hersh, Xiaobo Zhou, Jeong H Yun, ChangHee Lee, Tao Liu, Siqi Liu, Edy Y Kim, Shuang Xu, Jeffrey L Curtis, Luca Pinello, Russell P Bowler, Edwin K Silverman, Craig P Hersh, Xiaobo Zhou

Abstract

Chronic obstructive pulmonary disease (COPD) is mainly caused by cigarette smoking and characterized by chronic inflammation in vulnerable individuals. However, it is unknown how genetic factors may shape chronic inflammation in COPD. To understand how hedgehog interacting protein, encoded by HHIP gene identified in the genome-wide association study in COPD, plays a role in inflammation, we utilized Hhip+/- mice that present persistent inflammation and emphysema upon aging similar to that observed in human COPD. By performing single-cell RNA sequencing of the whole lung from mice at different ages, we found that Hhip+/- mice developed a cytotoxic immune response with a specific increase in killer cell lectin-like receptor G1-positive CD8+ T cells with upregulated Ifnγ expression recapitulating human COPD. Hhip expression was restricted to a lung fibroblast subpopulation that had increased interaction with CD8+ T lymphocytes in Hhip+/- compared with Hhip+/+ during aging. Hhip-expressing lung fibroblasts had upregulated IL-18 pathway genes in Hhip+/- lung fibroblasts, which was sufficient to drive increased levels of IFN-γ in CD8+ T cells ex vivo. Our finding provides insight into how a common genetic variation contributes to the amplified lymphocytic inflammation in COPD.

Trial registration: ClinicalTrials.gov NCT00608764.

Keywords: COPD; Inflammation; Mouse models; Pulmonology.

Figures

Figure 1. Single-cell RNA sequencing of Hhip…
Figure 1. Single-cell RNA sequencing of Hhip+/– and Hhip+/+ murine lungs.
(A) Experimental design of lung single-cell RNA sequencing from age-matched Hhip+/– and WT littermate Hhip+/+ mice. (B) Unbiased clustering of 38,875 cells from 26 clusters by uniform manifold approximation and projection (UMAP) plot. (C) Expression of representative marker genes across cell type clusters. (D) UMAP plot indicating fibroblast cluster expressing Hhip (red dots). (E) Nine subclusters of lung fibroblasts showing heterogeneity. (F) Expression of representative fibroblast subcluster marker genes shown in dot plot. Hhip is a marker gene for myofibroblast subcluster. (G) Hhip expression is decreased in Hhip+/– lungs. Expression values are normalized unique molecular identifier (UMI) count. Lymph.agg, lymphoid aggregates; AE: alveolar epithelial cell; AT1, type 1 alveolar epithelial cell; Cap Endothelial, capillary endothelial cell; Lymph Endothelial, lymphatic endothelial cell; AM, alveolar macrophage; IM, interstitial macrophage; Mesprogenitor: mesenchymal progenitor; MonoC, classical monocyte; MonoInt, intermediate monocyte; MonoNC, nonclassical monocyte; DC, dendritic cell; CD8TE, CD8+ effector memory T cell; CD8TTE, CD8+ terminal effector T cell; gdT, γδ T cell; NK, natural killer cell; Treg, regulatory T cell; ILC, innate lymphoid cell.
Figure 2. Number of CDTTEs increased in…
Figure 2. Number of CDTTEs increased in Hhip+/– mice with age.
(A) Naive CD8+ T cells (Ccr7+CD62L+) differentiate into terminal effector cells (Klrg1+CD127–). Proportion of CD8+ T cells including naive CD8+ T (CD8T) cells, effector memory CD8+ T cells (CD8TE), and terminal effector CD8+ T cells (CD8TTE) increases with age in Hhip+/– (Het) mice. (B) FACS gating strategy for CD8TTE cells. CD8+ T cells were gated for KLRG1hiCD127– populations. (C) Proportion of CD8TTEs in total CD8+ T cells. Each point represents an individual biological replicate. Error bars indicate standard error of the mean (SEM). *P < 0.05. Wilcoxon matched pairs (by age- and sex-matched littermates) signed rank test.
Figure 3. Increased activation of CDTTEs in…
Figure 3. Increased activation of CDTTEs in Hhip+/– mice.
(A) Average expression (normalized UMI count) of Ifnγ in various lymphocyte subtypes showing terminal effector CD8+ T (CD8TTE) cells with increased Ifnγ expression in Hhip+/– mice. (B) Ifnγ expression is increased in Hhip+/– lung CD8+ T cells as measured by qPCR (n = 4 mice/group, 4 months of age). Error bars indicate SEM. (C) Average expression of Tbx21 in various lymphocytes showing CD8TTE cells with increased Tbx21 expression in Hhip+/– mice. (D) Average expression of Rorc in various lymphocyte subtypes. (E) Average expression of Gata3 in various lymphocytes with enrichment in Tregs, NKT cells, and ILCs. *P < 0.05, unpaired Student’s t test. CD8TTE, CD8+ terminal effector T cell; CD8TE, CD8+ effector memory T cell; CD8T, CD8+ naive T cell; CD4T, CD4+ T cell; NKT, natural killer T cell; gdT, γδ T cell; Treg, regulatory T cell; NK, natural killer cell; ILC, innate lymphoid cell.
Figure 4. Inflammatory response–related pathways are enriched…
Figure 4. Inflammatory response–related pathways are enriched in Hhip+/– CD8+ T cells and fibroblasts.
Selected pathways that are significantly enriched with upregulated genes in Hhip+/– compared with Hhip+/+ mice by Gene Ontology (GO) analysis in (A) all CD8+ T cells and (B) fibroblasts. Pathway analysis was performed with gprofiler2. P value is FDR adjusted.
Figure 5. Receptor-ligand analysis suggests dysregulated fibroblast-T…
Figure 5. Receptor-ligand analysis suggests dysregulated fibroblast-T cell interaction in lungs from Hhip-haploinsufficient mice.
(A) Genotype-dependent differential receptor-ligand interactions across various cell types from mice at 4 months (4m) and 8 months (8m) of age. Color scale bar indicates number of interaction pairs between cell types. (B) Selected ligand (blue)-receptor (pink) pairs between fibroblasts and immune cells from mice at different age points. Select receptor-ligand interaction pairs that are different by genotype are marked by black rectangles. Dot size indicates P values and the means of expression levels (normalized UMI counts) of interacting molecules are indicated by color of each dot. Wt, Hhip+/+; Het, Hhip+/–; AE, alveolar epithelial cell; Endo, endothelial cell; AM, alveolar macrophage; IM, interstitial macrophage; MonoC, classical monocyte; MonoNC, nonclasscial monocyte; DC, dendritic cell; B, B cell; CD8T, CD8+ naive T cell; CD8TE, CD8+ effector memory T cell; CD8TTE, CD8+ terminal effector T cell; gdT, γδ T cell; NK, natural killer cell; Treg, regulatory T cell; ILC, innate lymphoid cell.
Figure 6. Hhip +/– lung fibroblasts induce…
Figure 6. Hhip+/– lung fibroblasts induce increased IFN-γ production from CD8+ T cells.
(A) Experimental design. Splenic CD8+ T cells from Hhip+/+ mice were treated with conditioned medium (CM) from lung fibroblasts from 11-month-old Hhip+/+ and Hhip+/– mice for 24 hours. (B) WT CD8+ T cells incubated with Hhip+/–-derived CM produced increased levels of IFN-γ. (C) Lung fibroblasts from Hhip+/– mice have higher expression of Il18. (D) IL-18 level is elevated in the Hhip+/–-derived CM measured by ELISA. (E) Knockdown of HHIP by siRNA in adult primary human lung fibroblasts (n = 3) and (F) human fetal lung fibroblast cell line MRC5 led to increased expression of Il18. (G) Expression of Ifnγ in WT splenic CD8+ T cells treated with conditioned medium derived from Hhip+/+ or Hhip+/– fibroblasts for 24 hours, with or without IL-18 neutralizing Ab (30 μg/mL) as measured by qPCR with Cd8a as the reference gene. Each point represents an individual biological replicate. Data are represented as mean ± SD, with n ≥ 3 per group. *P < 0.05, **P < 0.01, ***P < 0.001, unpaired Student’s t test (BF), paired Student’s t test (G). Representative results are shown (from ≥2 replicated experiments with 2–4 mice in each repeat for BD and F). KD, knockdown.

References

    1. Lutz SM, et al. A genome-wide association study identifies risk loci for spirometric measures among smokers of European and African ancestry. BMC Genet. 2015;16(1):138. doi: 10.1186/s12863-015-0299-4.
    1. Cho MH, et al. Risk loci for chronic obstructive pulmonary disease: a genome-wide association study and meta-analysis. Lancet Respir Med. 2014;2(3):214–225. doi: 10.1016/S2213-2600(14)70002-5.
    1. Hobbs BD, et al. Genetic loci associated with chronic obstructive pulmonary disease overlap with loci for lung function and pulmonary fibrosis. Nat Genet. 2017;49(3):426–432. doi: 10.1038/ng.3752.
    1. Pillai SG, et al. A genome-wide association study in chronic obstructive pulmonary disease (COPD): identification of two major susceptibility loci. PLoS Genet. 2009;5(3):e1000421. doi: 10.1371/journal.pgen.1000421.
    1. Cho MH, et al. A genome-wide association study of emphysema and airway quantitative imaging phenotypes. Am J Respir Crit Care Med. 2015;192(5):559–569. doi: 10.1164/rccm.201501-0148OC.
    1. Boueiz A, et al. Genome-wide association study of the genetic determinants of emphysema distribution. Am J Respir Crit Care Med. 2017;195(6):757–771. doi: 10.1164/rccm.201605-0997OC.
    1. van der Plaat DA, et al. Genome-wide association study on the FEV1/FVC ratio in never-smokers identifies HHIP and FAM13A. J Allergy Clin Immunol. 2017;139(2):533–540. doi: 10.1016/j.jaci.2016.06.062.
    1. Repapi E, et al. Genome-wide association study identifies five loci associated with lung function. Nat Genet. 2010;42(1):36–44. doi: 10.1038/ng.501.
    1. Zhou X, et al. Identification of a chronic obstructive pulmonary disease genetic determinant that regulates HHIP. Hum Mol Genet. 2012;21(6):1325–1335. doi: 10.1093/hmg/ddr569.
    1. Chuang P-T, et al. Feedback control of mammalian Hedgehog signaling by the Hedgehog-binding protein, Hip1, modulates Fgf signaling during branching morphogenesis of the lung. Genes Dev. 2003;17(3):342–347. doi: 10.1101/gad.1026303.
    1. Lao T, et al. Haploinsufficiency of Hedgehog interacting protein causes increased emphysema induced by cigarette smoke through network rewiring. Genome Med. 2015;7(1):12. doi: 10.1186/s13073-015-0137-3.
    1. Hogg JC, et al. The nature of small-airway obstruction in chronic obstructive pulmonary disease. N Engl J Med. 2004;350(26):2645–2653. doi: 10.1056/NEJMoa032158.
    1. Lao T et al. Hhip haploinsufficiency sensitizes mice to age-related emphysema. Proc Natl Acad Sci U S A. 2016;113(32):E4681–E4687. doi: 10.1073/pnas.1602342113.
    1. Wain LV, et al. Novel insights into the genetics of smoking behaviour, lung function, and chronic obstructive pulmonary disease (UK BiLEVE): a genetic association study in UK Biobank. Lancet Respir Med. 2015;3(10):769–781. doi: 10.1016/S2213-2600(15)00283-0.
    1. Perera WR, et al. Inflammatory changes, recovery and recurrence at COPD exacerbation. Eur Respir J. 2007;29(3):527–534. doi: 10.1183/09031936.00092506.
    1. Butler A, et al. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat Biotechnol. 2018;36(5):411–420. doi: 10.1038/nbt.4096.
    1. Satija R, et al. Spatial reconstruction of single-cell gene expression data. Nat Biotechnol. 2015;33(5):495–502. doi: 10.1038/nbt.3192.
    1. Han X, et al. Mapping the mouse cell atlas by microwell-seq. Cell. 2018;172(5):1091–1107. doi: 10.1016/j.cell.2018.02.001.
    1. Xie T, et al. Single-cell deconvolution of fibroblast heterogeneity in mouse pulmonary fibrosis. Cell Rep. 2018;22(13):3625–3640. doi: 10.1016/j.celrep.2018.03.010.
    1. Aran D, et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat Immunol. 2019;20(2):163–172. doi: 10.1038/s41590-018-0276-y.
    1. Buchholz VR, et al. T cell fate at the single-cell level. Annu Rev Immunol. 2016;34:65–92. doi: 10.1146/annurev-immunol-032414-112014.
    1. Herndler-Brandstetter D, et al. KLRG1+ effector CD8+ T cells lose KLRG1, differentiate into all memory T cell lineages, and convey enhanced protective immunity. Immunity. 2018;48(4):716–729. doi: 10.1016/j.immuni.2018.03.015.
    1. Efremova M, et al. CellPhoneDB: inferring cell-cell communication from combined expression of multi-subunit ligand-receptor complexes. Nat Protoc. 2020;15(4):1484–1506. doi: 10.1038/s41596-020-0292-x.
    1. Abel S, et al. The transmembrane CXC-chemokine ligand 16 is induced by IFN-gamma and TNF-alpha and shed by the activity of the disintegrin-like metalloproteinase ADAM10. J Immunol. 2004;172(10):6362–6372. doi: 10.4049/jimmunol.172.10.6362.
    1. Yang J, et al. Induction of interferon-gamma production in Th1 CD4+ T cells: evidence for two distinct pathways for promoter activation. Eur J Immunol. 1999;29(2):548–555. doi: 10.1002/(SICI)1521-4141(199902)29:02<548::AID-IMMU548>;2-Z.
    1. Dinarello CA, et al. Interleukin-18 and IL-18 binding protein. Front Immunol. 2013;4:289.
    1. Regan EA, et al. Genetic epidemiology of COPD (COPDGene) study design. COPD. 2011;7(1):32–43. doi: 10.3109/15412550903499522.
    1. Grumelli S, et al. An immune basis for lung parenchymal destruction in chronic obstructive pulmonary disease and emphysema. PLoS Med. 2004;1(1):075–083.
    1. Takabatake N, et al. Impaired systemic cell-mediated immunity and increased susceptibility to acute respiratory tract infections in patients with COPD. Respir Med. 2005;99(4):485–492. doi: 10.1016/j.rmed.2004.09.013.
    1. Barnes PJ. Inflammatory endotypes in COPD. Allergy. 2019;74(7):1249–1256.
    1. Henson S, et al. KLRG-1 signalling induced defective Akt (ser473) phosphorylation and proliferative dysfunction of highly differentiated CD8+ T cells. Blood. 2009;113(26):6619–6628. doi: 10.1182/blood-2009-01-199588.
    1. Cui W, Kaech SM. Generation of effector CD8+ T cells and their conversion to memory T cells. Immunol Rev. 2010;236(1):151–166. doi: 10.1111/j.1600-065X.2010.00926.x.
    1. Li L, et al. KLRG1 restricts memory T cell antitumor immunity. Oncotarget. 2016;7(38):61670–61678. doi: 10.18632/oncotarget.11430.
    1. Saetta M, et al. CD8+ T-lymphocytes in peripheral airways of smokers with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 1998;157(3 pt 1):822–826.
    1. Freeman CM, et al. CC chemokine receptor 5 and CXC chemokine receptor 6 expression by lung CD8+ cells correlates with chronic obstructive pulmonary disease severity. Am J Pathol. 2007;171(3):767–776. doi: 10.2353/ajpath.2007.061177.
    1. Wang Z, et al. Interferon gamma induction of pulmonary emphysema in the adult murine lung. J Exp Med. 2000;192(11):1587–1600. doi: 10.1084/jem.192.11.1587.
    1. Kang MJ, et al. IL-18 induces emphysema and airway and vascular remodeling via IFN-γ, IL-17A, and IL-13. Am J Respir Crit Care Med. 2012;185(11):1205–1217. doi: 10.1164/rccm.201108-1545OC.
    1. Hardaker EL, et al. Regulation of TNF-alpha- and IFN-gamma-induced CXCL10 expression: participation of the airway smooth muscle in the pulmonary inflammatory response in chronic obstructive pulmonary disease. FASEB J. 2004;18(1):191–193. doi: 10.1096/fj.03-0170fje.
    1. Barnes PJ, Cosio MG. Characterization of T lymphocytes in chronic obstructive pulmonary disease. PLoS Med. 2004;1(1):e20. doi: 10.1371/journal.pmed.0010020.
    1. Hoshino T, et al. Pulmonary inflammation and emphysema: role of the cytokines IL-18 and IL-13. Am J Respir Crit Care Med. 2007;176(1):49–62. doi: 10.1164/rccm.200603-316OC.
    1. Dima E, et al. Implication of Interleukin (IL)-18 in the pathogenesis of chronic obstructive pulmonary disease (COPD) Cytokine. 2015;74(2):313–317. doi: 10.1016/j.cyto.2015.04.008.
    1. Briend E, et al. IL-18 associated with lung lymphoid aggregates drives IFNγ production in severe COPD. Respir Res. 2017;18(1):159. doi: 10.1186/s12931-017-0641-7.
    1. Freeman CM, et al. Cytotoxic potential of lung CD8+ T cells increases with chronic obstructive pulmonary disease severity and with in vitro stimulation by IL-18 or IL-15. J Immunol. 2010;184(11):6504–6513. doi: 10.4049/jimmunol.1000006.
    1. Wang C, et al. Expansion of hedgehog disrupts mesenchymal identity and induces emphysema phenotype. J Clin Invest. 2018;128(10):4343–4358. doi: 10.1172/JCI99435.
    1. Schaum N, et al. Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris. Nature. 2018;562(7727):367–372. doi: 10.1038/s41586-018-0590-4.
    1. Tsukui T, et al. Collagen-producing lung cell atlas identifies multiple subsets with distinct localization and relevance to fibrosis. Nat Commun. 2020;11(1):1920. doi: 10.1038/s41467-020-15647-5.
    1. Raredon MSB, et al. Single-cell connectomic analysis of adult mammalian lungs. Sci Adv. 2019;5(12):eaaw3851. doi: 10.1126/sciadv.aaw3851.
    1. Travaglini KJ, et al. A molecular cell atlas of the human lung from single-cell RNA sequencing. Nature. 2020;587(7835):619–625. doi: 10.1038/s41586-020-2922-4.
    1. Ardini-Poleske ME, et al. LungMAP: the molecular atlas of lung development program. Am J Physiol Lung Cell Mol Physiol. 2017;313(5):L733–L740. doi: 10.1152/ajplung.00139.2017.
    1. Barnes PJ, et al. Chronic obstructive pulmonary disease. Nat Rev Dis Primers. 2015;1:15076. doi: 10.1038/nrdp.2015.76.
    1. Sun W, et al. Common genetic polymorphisms influence blood biomarker measurements in COPD. PLoS Genet. 2016;12(8):e1006011. doi: 10.1371/journal.pgen.1006011.
    1. Chuang PT, McMahon AP. Vertebrate Hedgehog signalling modulated by induction of a Hedgehog-binding protein. Nature. 1999;397(6720):617–621. doi: 10.1038/17611.
    1. Watanabe M, et al. A novel flow cytometry-based cell capture platform for the detection, capture and molecular characterization of rare tumor cells in blood. J Transl Med. 2014;12(1):143. doi: 10.1186/1479-5876-12-143.
    1. Klein AM, et al. Droplet barcoding for single-cell transcriptomics applied to embryonic stem cells. Cell. 2015;161(5):1187–1201. doi: 10.1016/j.cell.2015.04.044.
    1. DeMaio L, et al. Characterization of mouse alveolar epithelial cell monolayers. Am J Physiol Lung Cell Mol Physiol. 2009;296(6):L1051–L1058. doi: 10.1152/ajplung.00021.2009.
    1. Stuart T, et al. Comprehensive integration of single-cell data. Cell. 2019;177(7):1888–1902. doi: 10.1016/j.cell.2019.05.031.
    1. doi: 10.1101/653253. Korsunsky I, et al. Presto scales Wilcoxon and auROC analyses to millions of observations [preprint]. Posted on bioRxiv May 29, 2019.
    1. Heng TSP, et al. The Immunological Genome Project: networks of gene expression in immune cells. Nat Immunol. 2008;9(10):1091–1094. doi: 10.1038/ni1008-1091.
    1. Hafemeister C, Satija R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 2019;20(1):296. doi: 10.1186/s13059-019-1874-1.
    1. Durinck S, et al. BioMart and Bioconductor: a powerful link between biological databases and microarray data analysis. Bioinformatics. 2005;21(16):3439–3440. doi: 10.1093/bioinformatics/bti525.
    1. Raudvere U, et al. g:Profiler: a web server for functional enrichment analysis and conversions of gene lists (2019 update) Nucleic Acids Res. 2019;47(w1):W191–W198. doi: 10.1093/nar/gkz369.

Source: PubMed

Подписаться