scholarly journals Single Cell RNA-seq reveals ectopic and aberrant lung resident cell populations in Idiopathic Pulmonary Fibrosis

2019 ◽  
Author(s):  
Taylor S. Adams ◽  
Jonas C. Schupp ◽  
Sergio Poli ◽  
Ehab A. Ayaub ◽  
Nir Neumark ◽  
...  

AbstractWe provide a single cell atlas of Idiopathic Pulmonary Fibrosis (IPF), a fatal interstitial lung disease, focusing on resident lung cell populations. By profiling 312,928 cells from 32 IPF, 29 healthy control and 18 chronic obstructive pulmonary disease (COPD) lungs, we demonstrate that IPF is characterized by changes in discrete subpopulations of cells in the three major parenchymal compartments: the epithelium, endothelium and stroma. Among epithelial cells, we identify a novel population of IPF enriched aberrant basaloid cells that co-express basal epithelial markers, mesenchymal markers, senescence markers, developmental transcription factors and are located at the edge of myofibroblast foci in the IPF lung. Among vascular endothelial cells in the in IPF lung parenchyma we identify an expanded cell population transcriptomically identical to vascular endothelial cells normally restricted to the bronchial circulation. We confirm the presence of both populations by immunohistochemistry and independent datasets. Among stromal cells we identify fibroblasts and myofibroblasts in both control and IPF lungs and leverage manifold-based algorithms diffusion maps and diffusion pseudotime to infer the origins of the activated IPF myofibroblast. Our work provides a comprehensive catalogue of the aberrant cellular transcriptional programs in IPF, demonstrates a new framework for analyzing complex disease with scRNAseq, and provides the largest lung disease single-cell atlas to date.

2020 ◽  
Vol 6 (28) ◽  
pp. eaba1983 ◽  
Author(s):  
Taylor S. Adams ◽  
Jonas C. Schupp ◽  
Sergio Poli ◽  
Ehab A. Ayaub ◽  
Nir Neumark ◽  
...  

We provide a single-cell atlas of idiopathic pulmonary fibrosis (IPF), a fatal interstitial lung disease, by profiling 312,928 cells from 32 IPF, 28 smoker and nonsmoker controls, and 18 chronic obstructive pulmonary disease (COPD) lungs. Among epithelial cells enriched in IPF, we identify a previously unidentified population of aberrant basaloid cells that coexpress basal epithelial, mesenchymal, senescence, and developmental markers and are located at the edge of myofibroblast foci in the IPF lung. Among vascular endothelial cells, we identify an ectopically expanded cell population transcriptomically identical to bronchial restricted vascular endothelial cells in IPF. We confirm the presence of both populations by immunohistochemistry and independent datasets. Among stromal cells, we identify IPF myofibroblasts and invasive fibroblasts with partially overlapping cells in control and COPD lungs. Last, we confirm previous findings of profibrotic macrophage populations in the IPF lung. Our comprehensive catalog reveals the complexity and diversity of aberrant cellular populations in IPF.


2021 ◽  
pp. bmjspcare-2020-002831
Author(s):  
Meena Kalluri ◽  
Sarah Younus ◽  
Nathan Archibald ◽  
Janice Richman-Eisenstat ◽  
Charlotte Pooler

BackgroundIdiopathic pulmonary fibrosis (IPF) is a progressive, incurable fibrotic lung disease in which patients and caregivers report a high symptom burden. Symptoms are often poorly managed and patients and caregivers struggle to alleviate their distress in the absence of self-management support.AimTo explore perceptions of symptoms, symptom management strategies and self-efficacy for patients with IPF and caregivers who received self-management education and action plans created and provided in a Multidisciplinary Collaborative Interstitial Lung Disease (MDC-ILD) Clinic.DesignA qualitative study was conducted with participants recruited from the MDC-ILD Clinic. Participants received an early integrated palliative approach; most attended ILD pulmonary rehabilitation and some received home care support. Semistructured interviews were conducted. Patient participants completed Measure Yourself Medical Outcome Profile (MYMOP) for symptom assessment and Chronic Obstructive Pulmonary Disease Self-Efficacy Scale to assess self-management efficacy.ResultsThirteen patients and eight self-declared caregiver participants were interviewed. IPF severity ranged from mild to advanced disease. Participants integrated and personalised self-management strategies. They were intentional and confident, focused on living well and engaged in anticipatory planning. Twelve participants completed the MYMOP. Five reported dyspnoea. Four reported fatigue as an additional or only symptom. One reported cough. Five declared no dyspnoea, cough or fatigue. Participants reported 80% self-efficacy in symptom management.ConclusionsThe approach to symptom self-management and education was beneficial to patients with IPF and caregiver participants. Participants personalised the strategies, focusing on living, and planned both in the moment and for the future. They were confident and expressed dignity and meaning in their lives.


2016 ◽  
Vol 310 (3) ◽  
pp. L249-L262 ◽  
Author(s):  
Andrew J. Bryant ◽  
Ryan P. Carrick ◽  
Melinda E. McConaha ◽  
Brittany R. Jones ◽  
Sheila D. Shay ◽  
...  

Pulmonary hypertension (PH) complicating chronic parenchymal lung disease, such as idiopathic pulmonary fibrosis, results in significant morbidity and mortality. Since the hypoxia-inducible factor (HIF) signaling pathway is important for development of pulmonary hypertension in chronic hypoxia, we investigated whether HIF signaling in vascular endothelium regulates development of PH related to pulmonary fibrosis. We generated a transgenic model in which HIF is deleted within vascular endothelial cells and then exposed these mice to chronic intraperitoneal bleomycin to induce PH associated with lung fibrosis. Although no differences in the degree of fibrotic remodeling were observed, we found that endothelial HIF-deficient mice were protected against development of PH, including right ventricle and pulmonary vessel remodeling. Similarly, endothelial HIF-deficient mice were protected from PH after a 4-wk exposure to normobaric hypoxia. In vitro studies of pulmonary vascular endothelial cells isolated from the HIF-targeted mice and controls revealed that endothelial HIF signaling increases endothelial cell expression of connective tissue growth factor, enhances vascular permeability, and promotes pulmonary artery smooth muscle cell proliferation and wound healing ability, all of which have the potential to impact the development of PH in vivo. Taken together, these studies demonstrate that vascular endothelial cell HIF signaling is necessary for development of hypoxia and pulmonary fibrosis associated PH. As such, HIF and HIF-regulated targets represent a therapeutic target in these conditions.


Author(s):  
Shengnan Yang ◽  
Peipei Liu ◽  
Yale Jiang ◽  
Zai Wang ◽  
Huaping Dai ◽  
...  

Idiopathic pulmonary fibrosis (IPF) is an interstitial disease of unknown etiology characterized by progressive pulmonary fibrosis. Pirfenidone and nintedanib are the only drugs that can prolong the time to disease progression, slow down the decline in lung function, and prolong survival. However, they do not offer a cure and are associated with tolerability issues. The pluripotency of mesenchymal stem cells (MSCs) and their ability to regulate immunity, inhibit inflammation, and promote epithelial tissue repair highlight the promise of MSC therapy for treating interstitial lung disease. However, optimal protocols are lacking for multi-parameter selection in MSC therapy. This review summarizes preclinical studies on MSC transplantation for the treatment of interstitial lung disease and clinical studies with known results. An analysis of relevant factors for the optimization of treatment plans is presented, including MSCs with different sources, administration routes and timing, dosages, frequencies, and pretreatments with MSCs. This review proposes an optimized plan for guiding the design of future clinical research to identify therapeutic options for this complex disease.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3869-3869
Author(s):  
Mathew Angelos ◽  
Juan Abrahante ◽  
Dan Kaufman

Abstract Hemogenic endothelium is a rare and highly specialized subset of vascular endothelial cell that functions as a precursor cell population to definitive blood development. One of the mechanistic hallmarks of definitive hematopoiesis is the endothelial-to-hematopoietic transition (EHT), a process where hemogenic endothelial cells phenotypically switches to produce a detached and free-moving hematopoietic cell. While EHT has been visualized both in vitro and in vivo via lineage tracing studies, the regulation of this fate change at both a cellular and molecular level remains unclear. Human pluripotent stem cells, such as human embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs) can serve as a useful platform to characterize human hemogenic endothelium and to understand basic mechanisms underlying human EHT. We hypothesized that human hemogenic endothelium derived from hESCs is phenotypically and transcriptionally distinct from other vascular endothelial cells and hematopoietic progenitor cells. To identify human hemogenic endothelium, we used combined expression of endothelial cell surface antigens and RUNX1c expression via a fluorescent reporter previously validated in our lab. Specifically, we employed defined culture methods to differentiate hemogenic endothelial cells (HE) and vascular endothelial cells without hematopoietic potential (non-HE) from hESC harboring a RUNX1c-tdTomato reporter (hESC-RUNX1c-tdTomato). At Day 11 of differentiation, CD31+CD144+ endothelial cells were present, with approximately 40% of these cells tdTomato+. We next sorted HE (CD31+CD144+CD41-CD43-CD45-CD73-tdTomato+) and non-HE (CD31+CD144+CD41-CD43-CD45-CD73-tdTomato-) and cultured both populations in endothelial growth media (EGM) or hematopoietic growth media. HE retained characteristic cobblestone morphology and CD31 expression over the course of 5 days in EGM similar to control human umbilical vein endothelial cells. HE, but not non-HE, was able to generate non-adherent, tdTomato+ hematopoietic progenitor cells in hematopoietic growth media. Using these defined cell populations, we next performed single-cell RNASeq on HE, non-HE, and early hematopoietic progenitor cells (HP; CD34+CD43+tdTomato+). We captured a total of 55 HE, 47 non-HE, and 35 HP using the Fluidgm C1 single cell system and performed next generation sequencing of validated libraries. We analyzed single cell gene expression using Seurat, an R-based bioinformatics software package developed for the analysis of single cell NGS experiments. Populations were first validated based on expression of known genetic identifiers for vascular endothelium, hemogenic endothelium, and hematopoietic progenitor cells. HE and HP were both highly enriched for CDH5, ERG, ESAM, and FLI as compared to non-HE; these genes have been previously implicated in HE functionality. All single cell transcriptional profiles were similarly enriched for KDR, PECAM1, and LMO2, which are characteristic genes expressed in vascular endothelium. We next performed t-distributed stochastic neighbor embedding (t-SNE) using statistically significant principal components to distinguish groups of cells with similar transcriptional expression. Interestingly, we found overlap between individual HE and HP cells (Cluster 1); however, these cells were distinct from two, separate clusters of non-HE (Cluster 2 & 3; Figure 1). Further analysis of these clusters revealed novel biomarkers for HE/HP, such as TIMP3 (DGE: 2.06, Power: 0.91), ERG (DGE: 2.08, Power: 0.83), NOTCH4 (DGE: 2.35, Power: 0.78), and HEY1 (DGE: 2.47, Power: 0.74), Non-HE were found to cluster into two, distinct groups (Clusters 2 and 3), with Cluster 3 enriched for extracellular matrix genes (COL1A1 (DGE: 6.58, Power: 1.00), DCN (DGE: 6.09, Power: 1.00), VCAN (DGE: 3.55, Power: 1.00), FN1 (DGE: 2.19, Power: 0.96)). This profile suggests that some hESC-derived non-HE further differentiate into mesenchymal cells, in a process known as the endothelial-to mesenchymal transition (EndMT). Taken together, we demonstrate that hESC-derived HE and HP share a common developmental pathway, while non-HE is heterogeneous, but transcriptionally distinct. Our novel findings will be instrumental for testing new genetic targets to optimize the production of definitive hematopoietic cells. Figure 1 t-SNE analysis of individual HE, non-HE, and HP cells. Figure 1. t-SNE analysis of individual HE, non-HE, and HP cells. Disclosures Kaufman: Fate Therapeutics: Consultancy.


2021 ◽  
Vol 12 ◽  
Author(s):  
Eleanor Valenzi ◽  
Tracy Tabib ◽  
Anna Papazoglou ◽  
John Sembrat ◽  
Humberto E. Trejo Bittar ◽  
...  

Idiopathic pulmonary fibrosis (IPF) and systemic sclerosis-associated interstitial lung disease (SSc-ILD) differ in the predominant demographics and identified genetic risk alleles of effected patients, however both diseases frequently progress to respiratory failure and death. Contrasting advanced SSc-ILD to IPF provides insight to the role dysregulated immunity may play in pulmonary fibrosis. To analyze cell-type specific transcriptome commonalities and differences between IPF and SSc-ILD, we compared single-cell RNA-sequencing (scRNA-seq) of 21 explanted lung tissue specimens from patients with advanced IPF, SSc-ILD, and organ donor controls. Comparison of IPF and SSc-ILD tissue identified divergent patterns of interferon signaling, with interferon-gamma signaling upregulated in the SPP1hi and FABP4hi macrophages, cytotoxic T cells, and natural kill cells of IPF, while type I interferon signaling and production was upregulated in the corresponding SSc-ILD populations. Plasmacytoid dendritic cells were found in diseased lungs only, and exhibited upregulated cellular stress pathways in SSc-ILD compared to IPF. Alveolar type I cells were dramatically decreased in both IPF and SSc-ILD, with a distinct transcriptome signature separating these cells by disease. KRT5-/KRT17+ aberrant basaloid cells exhibiting markers of cellular senescence and epithelial-mesenchymal transition were identified in SSc-ILD for the first time. In summary, our study utilizes the enriched capabilities of scRNA-seq to identify key divergent cell types and pathways between IPF and SSc-ILD, providing new insights into the shared and distinct mechanisms between idiopathic and autoimmune interstitial lung diseases.


2021 ◽  
Author(s):  
EA Ayaub ◽  
S Poli ◽  
J Ng ◽  
T Adams ◽  
J Schupp ◽  
...  

AbstractIn this study, we leveraged a combination of single cell RNAseq, cytometry by time of flight (CyTOF), and flow cytometry to study the biology of a unique macrophage population in pulmonary fibrosis. Using the profiling data from 312,928 cells derived from 32 idiopathic pulmonary fibrosis (IPF), 29 healthy control and 18 chronic obstructive pulmonary disease (COPD) lungs, we identified an expanded population of macrophages in IPF that have a unique transcriptional profile associated with pro-fibrotic signature. These macrophages attain a hybrid transitional state between alveolar and interstitial macrophages, are enriched with biological processes of pro-fibrotic immune cells, and express novel surface markers and genes that have not been previously reported. We then applied single cell CyTOF to simultaneously measure 37 markers to precisely phenotype the uniquely expanded macrophage subset in IPF lungs. The SPADE algorithm independently identified an expanded macrophage cluster, and validated CD84 and CD36 as novel surface markers that highly label this cluster. Using a separate validation cohort, we confirmed an increase in CD84++CD36++ macrophage population in IPF compared to control and COPD lungs by flow cytometry. Further, using the signature from the IPF-specific macrophages and the LINCS drug database, we predicted small molecules that could reverse the signature of IPF-specific macrophages, and validated two molecules, CRT and Cucur, using THP-1 derived human macrophages and precision-cut lung slices (PCLS) from IPF patients. Utilizing a multi-dimensional translational approach, our work identified a novel and targetable population of macrophages found in end-stage pulmonary fibrosis.One Sentence SummarySingle cell RNAseq, CyTOF, and flow cytometry reveal the presence of an aberrant macrophage population in pulmonary fibrosis


Sign in / Sign up

Export Citation Format

Share Document