Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 117
Filter
1.
J Clin Invest ; 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38980870

ABSTRACT

Reciprocal interactions between alveolar fibroblasts and epithelial cells are crucial for lung homeostasis, injury repair, and fibrogenesis, but underlying mechanisms remain unclear. To investigate, we administered the fibroblast-selective TGFß1 signaling inhibitor, epigallocatechin gallate (EGCG), to Interstitial Lung Disease (ILD) patients undergoing diagnostic lung biopsy and conducted single-cell RNA sequencing on spare tissue. Biopsies from untreated patients showed higher fibroblast TGFß1 signaling compared to non-disease donor or end-stage ILD tissues. In vivo, EGCG downregulated TGFß1 signaling and several pro-inflammatory and stress pathways in biopsy samples. Notably, EGCG reduced fibroblast secreted frizzle-like receptor protein 2 (sFRP2), an unrecognized TGFß1 fibroblast target gene induced near type II alveolar epithelial cells (AEC2s) in situ. Using AEC2-fibroblast coculture organoids and precision cut lung slices (PCLS) from non-diseased donors, we found TGFß1 signaling promotes a spread AEC2 KRT17+ basaloid state, whereupon sFRP2 then activates a mature Krt5+ basal cell program. Wnt-receptor Frizzled 5 (Fzd5) expression and downstream calcineurin signaling were required for sFRP2-induced nuclear NFATc3 accumulation and KRT5 expression. These findings highlight stage-specific TGFß1 signaling in ILD, the therapeutic potential of EGCG in reducing IPF-related transcriptional changes, and identify TGFß1-non-canonical Wnt pathway crosstalk via sFRP2 as a novel mechanism for dysfunctional epithelial signaling in Idiopathic Pulmonary Fibrosis/ILD.

2.
Am J Respir Cell Mol Biol ; 70(4): 247-258, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38117250

ABSTRACT

BCL-2 family members are known to be implicated in survival in numerous biological settings. Here, we provide evidence that in injury and repair processes in lungs, BCL-2 mainly acts to attenuate endoplasmic reticulum (ER) stress and limit extracellular matrix accumulation. Days after an intratracheal bleomycin challenge, mice lose a fraction of their alveolar type II epithelium from terminal ER stress driven by activation of the critical ER sensor and stress effector IRE1α. This fraction is dramatically increased by BCL-2 inhibition, because IRE1α activation is dependent on its physical association with the BCL-2-proapoptotic family member BAX, and we found BCL-2 to disrupt this association in vitro. In vivo, navitoclax (a BCL-2/BCL-xL inhibitor) given 15-21 days after bleomycin challenge evoked strong activation of IRE-1α in mesenchymal cells and markers of ER stress, but not apoptosis. Remarkably, after BCL-2 inhibition, bleomycin-exposed mice demonstrated persistent collagen accumulation at Day 42, compared with resolution in controls. Enhanced fibrosis proved to be due to the RNAase activity of IRE1α downregulating MRC2 mRNA and protein, a mediator of collagen turnover. The critical role of MRC2 was confirmed in precision-cut lung slice cultures of Day-42 lungs from bleomycin-exposed wild-type and MRC2 null mice. Soluble and tissue collagen accumulated in precision-cut lung slice cultures from navitoclax-treated, bleomycin-challenged mice compared with controls, in a manner nearly identical to that of challenged but untreated MRC2 null mice. Thus, apart from mitochondrial-based antiapoptosis, BCL-2 functions to attenuate ER stress responses, fostering tissue homeostasis and injury repair.


Subject(s)
Aniline Compounds , Pulmonary Fibrosis , Sulfonamides , Mice , Animals , Pulmonary Fibrosis/metabolism , Endoribonucleases , Protein Serine-Threonine Kinases , Endoplasmic Reticulum Stress , Mice, Knockout , Collagen/metabolism , Bleomycin/pharmacology
3.
bioRxiv ; 2023 Aug 04.
Article in English | MEDLINE | ID: mdl-37577522

ABSTRACT

Reciprocal interactions between alveolar fibroblasts and epithelial cells are crucial for lung homeostasis, injury repair, and fibrogenesis, but underlying mechanisms remain unclear. To investigate this, we administered the fibroblast-selective TGFß1 signaling inhibitor, epigallocatechin gallate (EGCG), to Interstitial Lung Disease (ILD) patients undergoing diagnostic lung biopsy and conducted single-cell RNA sequencing on spare tissue. Unexposed biopsy samples showed higher fibroblast TGFß1 signaling compared to non-disease donor or end-stage ILD tissues. In vivo, EGCG significantly downregulated TGFß1 signaling and several pro-inflammatory and stress pathways in biopsy samples. Notably, EGCG reduced fibroblast secreted Frizzle-like Receptor Protein 2 (sFRP2), an unrecognized TGFß1 fibroblast target gene induced near type II alveolar epithelial cells (AEC2s). In human AEC2-fibroblast coculture organoids, sFRP2 was essential for AEC2 trans-differentiation to basal cells. Precision cut lung slices (PCLS) from normal donors demonstrated that TGFß1 promoted KRT17 expression and AEC2 morphological change, while sFRP2 was necessary for KRT5 expression in AEC2-derived basaloid cells. Wnt-receptor Frizzled 5 (Fzd5) expression and downstream calcineurin-related signaling in AEC2s were required for sFRP2-induced KRT5 expression. These findings highlight stage-specific TGFß1 signaling in ILD, the therapeutic potential of EGCG in reducing IPF-related transcriptional changes, and identify the TGFß1-non-canonical Wnt pathway crosstalk via sFRP2 as a novel mechanism for dysfunctional epithelial signaling in Idiopathic Pulmonary Fibrosis/ILD.

4.
J Immunol ; 209(2): 227-237, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35760520

ABSTRACT

Type 1 diabetes (T1D) in both humans and NOD mice is caused by T cell-mediated autoimmune destruction of pancreatic ß cells. Increased frequency or activity of autoreactive T cells and failures of regulatory T cells (Tregs) to control these pathogenic effectors have both been implicated in T1D etiology. Due to the expression of MHC class I molecules on ß cells, CD8 T cells represent the ultimate effector population mediating T1D. Developing autoreactive CD8 T cells normally undergo extensive thymic negative selection, but this process is impaired in NOD mice and also likely T1D patients. Previous studies identified an allelic variant of Nfkbid, a NF-κB signal modulator, as a gene strongly contributing to defective thymic deletion of autoreactive CD8 T cells in NOD mice. These previous studies found ablation of Nfkbid in NOD mice using the clustered regularly interspaced short palindromic repeats system resulted in greater thymic deletion of pathogenic CD8 AI4 and NY8.3 TCR transgenic T cells but an unexpected acceleration of T1D onset. This acceleration was associated with reductions in the frequency of peripheral Tregs. In this article, we report transgenic overexpression of Nfkbid in NOD mice also paradoxically results in enhanced thymic deletion of autoreactive CD8 AI4 T cells. However, transgenic elevation of Nfkbid expression also increased the frequency and functional capacity of peripheral Tregs, in part contributing to the induction of complete T1D resistance. Thus, future identification of a pharmaceutical means to enhance Nfkbid expression might ultimately provide an effective T1D intervention approach.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Animals , CD8-Positive T-Lymphocytes , Diabetes Mellitus, Experimental/pathology , Humans , Mice , Mice, Inbred NOD , Mice, Transgenic , T-Lymphocytes, Regulatory
5.
Nature ; 604(7904): 120-126, 2022 04.
Article in English | MEDLINE | ID: mdl-35355013

ABSTRACT

The human lung differs substantially from its mouse counterpart, resulting in a distinct distal airway architecture affected by disease pathology in chronic obstructive pulmonary disease. In humans, the distal branches of the airway interweave with the alveolar gas-exchange niche, forming an anatomical structure known as the respiratory bronchioles. Owing to the lack of a counterpart in mouse, the cellular and molecular mechanisms that govern respiratory bronchioles in the human lung remain uncharacterized. Here we show that human respiratory bronchioles contain a unique secretory cell population that is distinct from cells in larger proximal airways. Organoid modelling reveals that these respiratory airway secretory (RAS) cells act as unidirectional progenitors for alveolar type 2 cells, which are essential for maintaining and regenerating the alveolar niche. RAS cell lineage differentiation into alveolar type 2 cells is regulated by Notch and Wnt signalling. In chronic obstructive pulmonary disease, RAS cells are altered transcriptionally, corresponding to abnormal alveolar type 2 cell states, which are associated with smoking exposure in both humans and ferrets. These data identify a distinct progenitor in a region of the human lung that is not found in mouse that has a critical role in maintaining the gas-exchange compartment and is altered in chronic lung disease.


Subject(s)
Bronchioles , Ferrets , Multipotent Stem Cells , Pulmonary Alveoli , Animals , Bronchioles/cytology , Cell Lineage , Humans , Lung/pathology , Mice , Multipotent Stem Cells/cytology , Pulmonary Alveoli/cytology , Pulmonary Disease, Chronic Obstructive
6.
Nat Cell Biol ; 24(1): 10-23, 2022 01.
Article in English | MEDLINE | ID: mdl-34969962

ABSTRACT

Loss of alveolar type 2 cells (AEC2s) and the ectopic appearance of basal cells in the alveoli characterize severe lung injuries such as idiopathic pulmonary fibrosis (IPF). Here we demonstrate that human alveolar type 2 cells (hAEC2s), unlike murine AEC2s, transdifferentiate into basal cells in response to fibrotic signalling in the lung mesenchyme, in vitro and in vivo. Single-cell analysis of normal hAEC2s and mesenchymal cells in organoid co-cultures revealed the emergence of pathologic fibroblasts and basaloid cells previously described in IPF. Transforming growth factor-ß1 and anti-bone morphogenic protein signalling in the organoids promoted transdifferentiation. Trajectory and histologic analyses of both hAEC2-derived organoids and IPF epithelium indicated that hAEC2s transdifferentiate into basal cells through alveolar-basal intermediates that accumulate in proximity to pathologic CTHRC1hi/TGFB1hi fibroblasts. Our study indicates that hAEC2 loss and expansion of alveolar metaplastic basal cells in severe human lung injuries are causally connected through an hAEC2-basal cell lineage trajectory driven by aberrant mesenchyme.


Subject(s)
Cell Transdifferentiation/physiology , Epithelial Cells/cytology , Idiopathic Pulmonary Fibrosis/pathology , Keratin-5/metabolism , Pulmonary Alveoli/cytology , Respiratory Mucosa/cytology , Alveolar Epithelial Cells/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Cell Differentiation , Cells, Cultured , Epidermal Cells/cytology , Fibroblasts/cytology , Humans , Mesoderm/cytology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Signal Transduction/physiology , Single-Cell Analysis , Transforming Growth Factor beta1/metabolism
7.
J Clin Invest ; 131(19)2021 10 01.
Article in English | MEDLINE | ID: mdl-34596050

ABSTRACT

IL-33 is a well-studied cytokine that resides normally within nuclei but can be released by cell damage or stress to then signal via a single receptor widely expressed on immune cells to promote host resistance and type 2 allergic immunity. In this issue of the JCI, Wu et al. used a well-established model of mouse Sendai viral infection to show that IL-33 was induced in distal lung airway epithelium, specifically in cell-cycling basal cells. IL-33 induced cell-cycling basal cells to expand and migrate into the alveolar compartment, presumably to restore barrier function. However, restoring barrier function with airway-derived cells may also result in persistent alveolar metaplasia. Surprisingly, nuclear IL-33 in this system acted cell autonomously, independently of release and conventional ST2 (IL1RL1) receptor signaling. The findings uncover a signaling role for nuclear IL-33 in viral activation of mouse basal cells and add to the well-known "alarmin" function of IL-33.


Subject(s)
Hypersensitivity , Interleukin-33 , Alarmins , Animals , Cytokines , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-33/genetics , Lung , Mice
8.
Cell Rep ; 35(3): 109009, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33882319

ABSTRACT

Cancer cells function as primary architects of the tumor microenvironment. However, the molecular features of cancer cells that govern stromal cell phenotypes remain unclear. Here, we show that cancer-associated fibroblast (CAF) heterogeneity is driven by lung adenocarcinoma (LUAD) cells at either end of the epithelial-to-mesenchymal transition (EMT) spectrum. LUAD cells that have high expression of the EMT-activating transcription factor ZEB1 reprogram CAFs through a ZEB1-dependent secretory program and direct CAFs to the tips of invasive projections through a ZEB1-driven CAF repulsion process. The EMT, in turn, sensitizes LUAD cells to pro-metastatic signals from CAFs. Thus, CAFs respond to contextual cues from LUAD cells to promote metastasis.


Subject(s)
Adenocarcinoma of Lung/genetics , Cancer-Associated Fibroblasts/metabolism , Epithelial Cells/metabolism , Kidney Neoplasms/genetics , Lung Neoplasms/genetics , Mesenchymal Stem Cells/metabolism , Zinc Finger E-box-Binding Homeobox 1/genetics , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/secondary , Alpha-Globulins/genetics , Alpha-Globulins/metabolism , Animals , Cancer-Associated Fibroblasts/pathology , Cell Communication , Cell Line, Tumor , Cell Movement , Cell Proliferation , Discoidin Domain Receptor 2/genetics , Discoidin Domain Receptor 2/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/secondary , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mesenchymal Stem Cells/pathology , Mice , Mice, Transgenic , Signal Transduction , Tumor Microenvironment/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism
9.
Thorax ; 76(7): 729-732, 2021 07.
Article in English | MEDLINE | ID: mdl-33472968

ABSTRACT

We recently identified epigallocatechin gallate (EGCG), a trihydroxyphenolic compound, as a dual inhibitor of lysyl oxidase-like2 and transforming growth factor-ß1 (TGFß1) receptor kinase that when given orally to patients with idiopathic pulmonary fibrosis (IPF) reversed profibrotic biomarkers in their diagnostic biopsies. Here, we extend these findings to advanced pulmonary fibrosis using cultured precision-cut lung slices from explants of patients with IPF undergoing transplantation. During these experiments, we were surprised to discover that not only did EGCG attenuate TGFß1 signalling and new collagen accumulation but also activated matrix metalloproteinase-dependent collagen I turnover, raising the possibility of slow fibrosis resolution with continued treatment.


Subject(s)
Amino Acid Oxidoreductases/metabolism , Collagen Type I/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Lung/metabolism , Transforming Growth Factor beta1/metabolism , Cells, Cultured , Humans , Idiopathic Pulmonary Fibrosis/pathology , Immunoblotting , Lung/pathology , Signal Transduction
10.
J Immunol ; 205(8): 2026-2038, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32938729

ABSTRACT

It has become increasingly appreciated that autoimmune responses against neuronal components play an important role in type 1 diabetes (T1D) pathogenesis. In fact, a large proportion of islet-infiltrating B lymphocytes in the NOD mouse model of T1D produce Abs directed against the neuronal type III intermediate filament protein peripherin. NOD-PerIg mice are a previously developed BCR-transgenic model in which virtually all B lymphocytes express the H and L chain Ig molecules from the intra-islet-derived anti-peripherin-reactive hybridoma H280. NOD-PerIg mice have accelerated T1D development, and PerIg B lymphocytes actively proliferate within islets and expand cognitively interactive pathogenic T cells from a pool of naive precursors. We now report adoptively transferred T cells or whole splenocytes from NOD-PerIg mice expectedly induce T1D in NOD.scid recipients but, depending on the kinetics of disease development, can also elicit a peripheral neuritis (with secondary myositis). This neuritis was predominantly composed of CD4+ and CD8+ T cells. Ab depletion studies showed neuritis still developed in the absence of NOD-PerIg CD8+ T cells but required CD4+ T cells. Surprisingly, sciatic nerve-infiltrating CD4+ cells had an expansion of IFN-γ- and TNF-α- double-negative cells compared with those within both islets and spleen. Nerve and islet-infiltrating CD4+ T cells also differed by expression patterns of CD95, PD-1, and Tim-3. Further studies found transitory early B lymphocyte depletion delayed T1D onset in a portion of NOD-PerIg mice, allowing them to survive long enough to develop neuritis outside of the transfer setting. Together, this study presents a new model of peripherin-reactive B lymphocyte-dependent autoimmune neuritis.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Nerve Tissue , Neuritis, Autoimmune, Experimental , Pancreas , Animals , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Nerve Tissue/immunology , Nerve Tissue/pathology , Neuritis, Autoimmune, Experimental/genetics , Neuritis, Autoimmune, Experimental/immunology , Neuritis, Autoimmune, Experimental/pathology , Pancreas/immunology , Pancreas/pathology
11.
Nat Commun ; 11(1): 4520, 2020 09 09.
Article in English | MEDLINE | ID: mdl-32908154

ABSTRACT

Tumor extracellular matrix has been associated with drug resistance and immune suppression. Here, proteomic and RNA profiling reveal increased collagen levels in lung tumors resistant to PD-1/PD-L1 blockade. Additionally, elevated collagen correlates with decreased total CD8+ T cells and increased exhausted CD8+ T cell subpopulations in murine and human lung tumors. Collagen-induced T cell exhaustion occurs through the receptor LAIR1, which is upregulated following CD18 interaction with collagen, and induces T cell exhaustion through SHP-1. Reduction in tumor collagen deposition through LOXL2 suppression increases T cell infiltration, diminishes exhausted T cells, and abrogates resistance to anti-PD-L1. Abrogating LAIR1 immunosuppression through LAIR2 overexpression or SHP-1 inhibition sensitizes resistant lung tumors to anti-PD-1. Clinically, increased collagen, LAIR1, and TIM-3 expression in melanoma patients treated with PD-1 blockade predict poorer survival and response. Our study identifies collagen and LAIR1 as potential markers for immunotherapy resistance and validates multiple promising therapeutic combinations.


Subject(s)
Adenocarcinoma of Lung/drug therapy , Antineoplastic Agents, Immunological/pharmacology , CD8-Positive T-Lymphocytes/immunology , Collagen/metabolism , Drug Resistance, Neoplasm/immunology , Lung Neoplasms/drug therapy , Receptors, Immunologic/metabolism , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/immunology , Adenocarcinoma of Lung/pathology , Amino Acid Oxidoreductases/genetics , Amino Acid Oxidoreductases/metabolism , Animals , Antineoplastic Agents, Immunological/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Biomarkers, Tumor/metabolism , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Datasets as Topic , Disease Models, Animal , Extracellular Matrix/drug effects , Extracellular Matrix/immunology , Extracellular Matrix/pathology , Female , Gene Knockdown Techniques , HEK293 Cells , Hepatitis A Virus Cellular Receptor 2/metabolism , Humans , Lung/immunology , Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Male , Mice , Mice, Transgenic , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , RNA-Seq , Receptors, Immunologic/genetics
12.
Nat Commun ; 11(1): 3559, 2020 07 16.
Article in English | MEDLINE | ID: mdl-32678092

ABSTRACT

The cell type specific sequences of transcriptional programs during lung regeneration have remained elusive. Using time-series single cell RNA-seq of the bleomycin lung injury model, we resolved transcriptional dynamics for 28 cell types. Trajectory modeling together with lineage tracing revealed that airway and alveolar stem cells converge on a unique Krt8 + transitional stem cell state during alveolar regeneration. These cells have squamous morphology, feature p53 and NFkB activation and display transcriptional features of cellular senescence. The Krt8+ state appears in several independent models of lung injury and persists in human lung fibrosis, creating a distinct cell-cell communication network with mesenchyme and macrophages during repair. We generated a model of gene regulatory programs leading to Krt8+ transitional cells and their terminal differentiation to alveolar type-1 cells. We propose that in lung fibrosis, perturbed molecular checkpoints on the way to terminal differentiation can cause aberrant persistence of regenerative intermediate stem cell states.


Subject(s)
Alveolar Epithelial Cells/metabolism , Keratin-8/metabolism , Pulmonary Alveoli/physiology , Pulmonary Fibrosis/pathology , Regeneration , Stem Cells/metabolism , Alveolar Epithelial Cells/cytology , Animals , Cell Communication , Disease Models, Animal , Female , Gene Expression Profiling , Humans , Keratin-8/genetics , Lung Injury/chemically induced , Lung Injury/metabolism , Lung Injury/pathology , Mice , Mice, Inbred C57BL , Pulmonary Alveoli/cytology , Pulmonary Fibrosis/metabolism , Single-Cell Analysis , Stem Cells/cytology
13.
Dev Cell ; 52(5): 546-547, 2020 03 09.
Article in English | MEDLINE | ID: mdl-32155436

ABSTRACT

How lung epithelium and endothelium co-develop to maintain structural integrity of alveoli remains unclear. In this issue of Developmental Cell, Ellis et al. define how epithelial Vegfa directs development of a distinct endothelial cell population that ultimately plays a critical role in ensuring appropriate alveolar septation during alveologenesis.


Subject(s)
Automobiles , Vascular Endothelial Growth Factor A , Animals , Endothelium , Lung , Mice , Pulmonary Alveoli
15.
Cell Stem Cell ; 26(3): 346-358.e4, 2020 03 05.
Article in English | MEDLINE | ID: mdl-31978363

ABSTRACT

Lung injury activates specialized adult epithelial progenitors to regenerate the epithelium. Depending on the extent of injury, both remaining alveolar type II cells (AEC2s) and distal airway stem/progenitors mobilize to cover denuded alveoli and restore normal barriers. The major source of airway stem/progenitors other than basal-like cells remains uncertain. Here, we define a distinct subpopulation (∼5%) of club-like lineage-negative epithelial progenitors (LNEPs) marked by high H2-K1 expression critical for alveolar repair. Quiescent H2-K1high cells account for virtually all in vitro regenerative activity of airway lineages. After bleomycin injury, H2-K1 cells expand and differentiate in vivo to alveolar lineages. However, injured H2-K1 cells eventually develop impaired self-renewal with features of senescence, limiting complete repair. Normal H2-K1high cells transplanted into injured lungs differentiate into alveolar cells and rescue lung function. These findings indicate that small subpopulations of specialized stem/progenitors are required for effective lung regeneration and are a potential therapeutic adjunct after major lung injury.


Subject(s)
Epithelial Cells , Lung Injury , Alveolar Epithelial Cells , Cell Differentiation , Humans , Lung , Stem Cells
16.
JCI Insight ; 4(24)2019 12 19.
Article in English | MEDLINE | ID: mdl-31687975

ABSTRACT

Accumulation of senescent cells is associated with the progression of pulmonary fibrosis, but mechanisms accounting for this linkage are not well understood. To explore this issue, we investigated whether a class of biologically active profibrotic lipids, the leukotrienes (LT), is part of the senescence-associated secretory phenotype. The analysis of conditioned medium (CM), lipid extracts, and gene expression of LT biosynthesis enzymes revealed that senescent cells secreted LT, regardless of the origin of the cells or the modality of senescence induction. The synthesis of LT was biphasic and followed by antifibrotic prostaglandin (PG) secretion. The LT-rich CM of senescent lung fibroblasts (IMR-90) induced profibrotic signaling in naive fibroblasts, which were abrogated by inhibitors of ALOX5, the principal enzyme in LT biosynthesis. The bleomycin-induced expression of genes encoding LT and PG synthases, level of cysteinyl LT in the bronchoalveolar lavage, and overall fibrosis were reduced upon senescent cell removal either in a genetic mouse model or after senolytic treatment. Quantification of ALOX5+ cells in lung explants obtained from idiopathic pulmonary fibrosis (IPF) patients indicated that half of these cells were also senescent (p16Ink4a+). Unlike human fibroblasts from unused donor lungs made senescent by irradiation, senescent IPF fibroblasts secreted LTs but failed to synthesize PGs. This study demonstrates for the first time to our knowledge that senescent cells secrete functional LTs, significantly contributing to the LT pool known to cause or exacerbate IPF.


Subject(s)
Cellular Senescence , Fibroblasts/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Leukotrienes/metabolism , Lung/pathology , Animals , Arachidonate 5-Lipoxygenase/metabolism , Bleomycin/toxicity , Bronchoalveolar Lavage Fluid/chemistry , Cell Line , Culture Media, Conditioned/metabolism , Disease Models, Animal , Disease Progression , Fibroblasts/pathology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Idiopathic Pulmonary Fibrosis/diagnosis , Leukotrienes/analysis , Lipoxygenase Inhibitors/pharmacology , Lung/cytology , Male , Mice , Primary Cell Culture , Prostaglandins/metabolism , Signal Transduction/drug effects
17.
Front Immunol ; 10: 1732, 2019.
Article in English | MEDLINE | ID: mdl-31428087

ABSTRACT

Previous studies indicate that B-lymphocytes play a key role activating diabetogenic T-lymphocytes during the development of autoimmune diabetes. Recently, two transgenic NOD mouse models were generated: the NOD-PerIg and the 116C-NOD mice. In NOD-PerIg mice, B-lymphocytes acquire an activated proliferative phenotype and support accelerated autoimmune diabetes development. In contrast, in 116C-NOD mice, B-lymphocytes display an anergic-like phenotype delaying autoimmune diabetes onset and decreasing disease incidence. The present study further evaluates the T- and B-lymphocyte phenotype in both models. In islet-infiltrating B-lymphocytes (IIBLs) from 116C-NOD mice, the expression of H2-Kd and H2-Ag7 is decreased, whereas that of BAFF, BAFF-R, and TACI is increased. In contrast, IIBLs from NOD-PerIg show an increase in CD86 and FAS expression. In addition, islet-infiltrating T-lymphocytes (IITLs) from NOD-PerIg mice exhibit an increase in PD-1 expression. Moreover, proliferation assays indicate a high capacity of B-lymphocytes from NOD-PerIg mice to secrete high amounts of cytokines and induce T-lymphocyte activation compared to 116C B-lymphocytes. This functional variability between 116C and PerIg B-lymphocytes ultimately results in differences in the ability to shape T-lymphocyte phenotype. These results support the role of B-lymphocytes as key regulators of T-lymphocytes in autoimmune diabetes and provide essential information on the phenotypic characteristics of the T- and B-lymphocytes involved in the autoimmune response in autoimmune diabetes.


Subject(s)
B-Lymphocyte Subsets/immunology , Diabetes Mellitus, Type 1/immunology , T-Lymphocyte Subsets/immunology , Animals , Antigen Presentation , Autoimmunity , Clonal Anergy , Cytokines/blood , Diabetes Mellitus, Type 1/pathology , Disease Models, Animal , Female , Gene Expression Profiling , Genes, Immunoglobulin , Immunophenotyping , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Lymphocyte Activation , Lymphopoiesis , Male , Mice , Mice, Inbred NOD , Mice, Transgenic , Specific Pathogen-Free Organisms , Spleen/immunology , Spleen/pathology
18.
J Clin Invest ; 129(5): 2107-2122, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30985294

ABSTRACT

Alveolar epithelium plays a pivotal role in protecting the lungs from inhaled infectious agents. Therefore, the regenerative capacity of the alveolar epithelium is critical for recovery from these insults in order to rebuild the epithelial barrier and restore pulmonary functions. Here, we show that sublethal infection of mice with Streptococcus pneumoniae, the most common pathogen of community-acquired pneumonia, led to exclusive damage in lung alveoli, followed by alveolar epithelial regeneration and resolution of lung inflammation. We show that surfactant protein C-expressing (SPC-expressing) alveolar epithelial type II cells (AECIIs) underwent proliferation and differentiation after infection, which contributed to the newly formed alveolar epithelium. This increase in AECII activities was correlated with increased nuclear expression of Yap and Taz, the mediators of the Hippo pathway. Mice that lacked Yap/Taz in AECIIs exhibited prolonged inflammatory responses in the lung and were delayed in alveolar epithelial regeneration during bacterial pneumonia. This impaired alveolar epithelial regeneration was paralleled by a failure to upregulate IκBa, the molecule that terminates NF-κB-mediated inflammatory responses. These results demonstrate that signals governing resolution of lung inflammation were altered in Yap/Taz mutant mice, which prevented the development of a proper regenerative niche, delaying repair and regeneration of alveolar epithelium during bacterial pneumonia.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Alveolar Epithelial Cells/cytology , Cell Cycle Proteins/metabolism , Pneumonia, Pneumococcal/pathology , Pulmonary Surfactant-Associated Protein C/metabolism , Trans-Activators/metabolism , Animals , Cell Differentiation , Cell Nucleus/metabolism , Cell Proliferation , Epithelial Cells/metabolism , Epithelium/microbiology , HEK293 Cells , Humans , Inflammation/metabolism , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/metabolism , Regeneration , Signal Transduction , Stem Cells/cytology , Streptococcus pneumoniae , YAP-Signaling Proteins
19.
PLoS One ; 14(1): e0209824, 2019.
Article in English | MEDLINE | ID: mdl-30625178

ABSTRACT

Endoplasmic reticulum stress (ER stress) has been implicated in the pathogenesis of idiopathic pulmonary fibrosis (IPF), a disease of progressive fibrosis and respiratory failure. ER stress activates a signaling pathway called the unfolded protein response (UPR) that either restores homeostasis or promotes apoptosis. The bifunctional kinase/RNase IRE1α is a UPR sensor/effector that promotes apoptosis if ER stress remains high and irremediable (i.e., a "terminal" UPR). Using multiple small molecule inhibitors against IRE1α, we show that ER stress-induced apoptosis of murine alveolar epithelial cells can be mitigated in vitro. In vivo, we show that bleomycin exposure to murine lungs causes early ER stress to activate IRE1α and the terminal UPR prior to development of pulmonary fibrosis. Small-molecule IRE1α kinase-inhibiting RNase attenuators (KIRAs) that we developed were used to evaluate the contribution of IRE1α activation to bleomycin-induced pulmonary fibrosis. One such KIRA-KIRA7-provided systemically to mice at the time of bleomycin exposure decreases terminal UPR signaling and prevents lung fibrosis. Administration of KIRA7 14 days after bleomycin exposure even promoted the reversal of established fibrosis. Finally, we show that KIRA8, a nanomolar-potent, monoselective KIRA compound derived from a completely different scaffold than KIRA7, likewise promoted reversal of established fibrosis. These results demonstrate that IRE1α may be a promising target in pulmonary fibrosis and that kinase inhibitors of IRE1α may eventually be developed into efficacious anti-fibrotic drugs.


Subject(s)
Alveolar Epithelial Cells/drug effects , Endoribonucleases/antagonists & inhibitors , Fibrosis/drug therapy , Lung/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Animals , Apoptosis/drug effects , Cell Line , Endoplasmic Reticulum Stress/drug effects , Fibrosis/metabolism , Fibrosis/pathology , Lung/metabolism , Lung/pathology , Mice , Protein Kinase Inhibitors/therapeutic use , Unfolded Protein Response/drug effects
20.
J Immunol ; 201(7): 1907-1917, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30127089

ABSTRACT

In both NOD mice and humans, the development of type 1 diabetes (T1D) is dependent in part on autoreactive CD8+ T cells recognizing pancreatic ß cell peptides presented by often quite common MHC class I variants. Studies in NOD mice previously revealed that the common H2-Kd and/or H2-Db class I molecules expressed by this strain aberrantly lose the ability to mediate the thymic deletion of pathogenic CD8+ T cell responses through interactions with T1D susceptibility genes outside the MHC. A gene(s) mapping to proximal chromosome 7 was previously shown to be an important contributor to the failure of the common class I molecules expressed by NOD mice to mediate the normal thymic negative selection of diabetogenic CD8+ T cells. Using an inducible model of thymic negative selection and mRNA transcript analyses, we initially identified an elevated Nfkbid expression variant as a likely NOD-proximal chromosome 7 region gene contributing to impaired thymic deletion of diabetogenic CD8+ T cells. CRISPR/Cas9-mediated genetic attenuation of Nfkbid expression in NOD mice resulted in improved negative selection of autoreactive diabetogenic AI4 and NY8.3 CD8+ T cells. These results indicated that allelic variants of Nfkbid contribute to the efficiency of intrathymic deletion of diabetogenic CD8+ T cells. However, although enhancing thymic deletion of pathogenic CD8+ T cells, ablating Nfkbid expression surprisingly accelerated T1D onset that was associated with numeric decreases in both regulatory T and B lymphocytes in NOD mice.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chromosomes, Human, Pair 7/genetics , Diabetes Mellitus, Type 1/immunology , I-kappa B Proteins/genetics , Thymus Gland/immunology , Alleles , Animals , Autoantigens/immunology , Cell Differentiation , Cells, Cultured , Clonal Deletion , Disease Models, Animal , Disease Susceptibility , Humans , I-kappa B Proteins/metabolism , Mice , Mice, Inbred NOD , Polymorphism, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...