Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 519
1.
Int J Rheum Dis ; 27(5): e15174, 2024 May.
Article En | MEDLINE | ID: mdl-38720423

OBJECTIVES: This study investigates the role of TNF-induced protein 3 (TNFAIP3) and CCAAT/enhancer-binding protein ß (C/EBPß) in alveolar macrophages (AMs) of patients with systemic sclerosis-associated interstitial lung disease (SSc-ILD) and their influence on pulmonary fibrosis. METHODS: Transfection of HEK293T cells and AMs with plasmids carrying TNFAIP3 and C/EBPß was performed, followed by co-culturing AMs with pulmonary fibroblasts. Immunoblotting analysis was then utilized to assess the expression of TNFAIP3, C/EBPß, and collagen type 1 (Col1). Quantitative PCR analysis was conducted to quantify the mRNA levels of C/EBPß, IL-10, and TGF-ß1. STRING database analysis, and immunoprecipitation assays were employed to investigate the interactions between TNFAIP3 and C/EBPß. RESULTS: TNFAIP3 expression was significantly reduced in SSc-ILD AMs, correlating with increased Col1 production in fibroblasts. Overexpression of TNFAIP3 inhibited this pro-fibrotic activity. Conversely, C/EBPß expression was elevated in SSc-ILD AMs, and its reduction through TNFAIP3 restoration decreased pro-fibrotic cytokines IL-10 and TGFß1 levels. Protein-protein interaction studies confirmed the regulatory relationship between TNFAIP3 and C/EBPß. CONCLUSIONS: This study highlights the important role of TNFAIP3 in regulating pulmonary fibrosis in SSc-ILD by modulating C/EBPß expression in AMs. These findings suggest that targeting TNFAIP3 could be a potential therapeutic strategy for managing SSc-ILD patients.


CCAAT-Enhancer-Binding Protein-beta , Coculture Techniques , Fibroblasts , Lung Diseases, Interstitial , Macrophages, Alveolar , Scleroderma, Systemic , Tumor Necrosis Factor alpha-Induced Protein 3 , Female , Humans , Male , Middle Aged , CCAAT-Enhancer-Binding Protein-beta/metabolism , CCAAT-Enhancer-Binding Protein-beta/genetics , Collagen Type I/metabolism , Collagen Type I/genetics , Fibroblasts/metabolism , HEK293 Cells , Interleukin-10/metabolism , Interleukin-10/genetics , Lung/metabolism , Lung/pathology , Lung Diseases, Interstitial/metabolism , Lung Diseases, Interstitial/etiology , Macrophages, Alveolar/metabolism , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/etiology , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/complications , Signal Transduction , Transforming Growth Factor beta1/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Adult , Aged
2.
J Transl Med ; 22(1): 457, 2024 May 14.
Article En | MEDLINE | ID: mdl-38745204

BACKGROUND AND PURPOSE: Interstitial lung disease (ILD) represents a significant complication of rheumatoid arthritis (RA) that lacks effective treatment options. This study aimed to investigate the intrinsic mechanism by which resveratrol attenuates rheumatoid arthritis complicated with interstitial lung disease through the AKT/TMEM175 pathway. METHODS: We established an arthritis model by combining chicken type II collagen and complete Freund's adjuvant. Resveratrol treatment was administered via tube feeding for 10 days. Pathological changes in both the joints and lungs were evaluated using HE and Masson staining techniques. Protein expression of TGF-ß1, AKT, and TMEM175 was examined in lung tissue. MRC-5 cells were stimulated using IL-1ß in combination with TGF-ß1 as an in vitro model of RA-ILD, and agonists of AKT, metabolic inhibitors, and SiRNA of TMEM175 were used to explore the regulation and mechanism of action of resveratrol RA-ILD. RESULTS: Resveratrol mitigates fibrosis in rheumatoid arthritis-associated interstitial lung disease and reduces oxidative stress and inflammation in RA-ILD. Furthermore, resveratrol restored cellular autophagy. When combined with the in vitro model, it was further demonstrated that resveratrol could suppress TGF-ß1 expression, and reduce AKT metamorphic activation, consequently inhibiting the opening of AKT/MEM175 ion channels. This, in turn, lowers lysosomal pH and enhances the fusion of autophagosomes with lysosomes, ultimately ameliorating the progression of RA-ILD. CONCLUSION: In this study, we demonstrated that resveratrol restores autophagic flux through the AKT/MEM175 pathway to attenuate inflammation as well as fibrosis in RA-ILD by combining in vivo and in vitro experiments. It further provides a theoretical basis for the selection of therapeutic targets for RA-ILD.


Arthritis, Rheumatoid , Fibrosis , Inflammation , Lung Diseases, Interstitial , Proto-Oncogene Proteins c-akt , Resveratrol , Signal Transduction , Resveratrol/pharmacology , Resveratrol/therapeutic use , Arthritis, Rheumatoid/complications , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/pathology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Lung Diseases, Interstitial/drug therapy , Lung Diseases, Interstitial/complications , Lung Diseases, Interstitial/pathology , Lung Diseases, Interstitial/metabolism , Humans , Inflammation/pathology , Inflammation/drug therapy , Signal Transduction/drug effects , Transforming Growth Factor beta1/metabolism , Membrane Proteins/metabolism , Autophagy/drug effects , Oxidative Stress/drug effects , Cell Line , Lung/pathology , Lung/drug effects , Male
3.
Cell Rep ; 43(4): 114114, 2024 Apr 23.
Article En | MEDLINE | ID: mdl-38625791

Patients afflicted with Stimulator of interferon gene (STING) gain-of-function mutations frequently present with debilitating interstitial lung disease (ILD) that is recapitulated in mice expressing the STINGV154M mutation (VM). Prior radiation chimera studies revealed an unexpected and critical role for non-hematopoietic cells in initiating ILD. To identify STING-expressing non-hematopoietic cell types required for the development of ILD, we use a conditional knockin (CKI) model and direct expression of the VM allele to hematopoietic cells, fibroblasts, epithelial cells, or endothelial cells. Only endothelial cell-targeted VM expression results in enhanced recruitment of immune cells to the lung associated with elevated chemokine expression and the formation of bronchus-associated lymphoid tissue, as seen in the parental VM strain. These findings reveal the importance of endothelial cells as instigators of STING-driven lung disease and suggest that therapeutic targeting of STING inhibitors to endothelial cells could potentially mitigate inflammation in the lungs of STING-associated vasculopathy with onset in infancy (SAVI) patients or patients afflicted with other ILD-related disorders.


Endothelial Cells , Gain of Function Mutation , Lung , Membrane Proteins , Animals , Membrane Proteins/metabolism , Membrane Proteins/genetics , Endothelial Cells/metabolism , Endothelial Cells/pathology , Mice , Lung/pathology , Lung/metabolism , Lymphocytes/metabolism , Lung Diseases, Interstitial/pathology , Lung Diseases, Interstitial/genetics , Lung Diseases, Interstitial/metabolism , Mice, Inbred C57BL , Humans
4.
Int J Mol Sci ; 25(8)2024 Apr 12.
Article En | MEDLINE | ID: mdl-38673881

Interstitial lung diseases (ILDs) are characterized by inflammation or fibrosis of the pulmonary parenchyma. Despite the involvement of immune cells and soluble mediators in pulmonary fibrosis, the influence of antimicrobial peptides (AMPs) remains underexplored. These effector molecules display a range of activities, which include immunomodulation and wound repair. Here, we investigate the role of AMPs in the development of fibrosis in ILD. We compare the concentration of different AMPs and different cytokines in 46 fibrotic (F-ILD) and 17 non-fibrotic (NF-ILD) patients by ELISA and using peripheral blood mononuclear cells from in vitro stimulation in the presence of lysozyme or secretory leukocyte protease inhibitor (SLPI) from 10 healthy donors. We observed that bronchoalveolar lavage (BAL) levels of AMPs were decreased in F-ILD patients (lysozyme: p < 0.001; SLPI: p < 0.001; LL-37: p < 0.001; lactoferrin: p = 0.47) and were negatively correlated with levels of TGF-ß (lysozyme: p = 0.02; SLPI: p < 0.001) and IL-17 (lysozyme: p < 0.001; SLPI: p < 0.001). We observed that lysozyme increased the percentage of CD86+ macrophages (p < 0.001) and the production of TNF-α (p < 0.001). We showed that lysozyme and SLPI were associated with clinical parameters (lysozyme: p < 0.001; SLPI: p < 0.001) and disease progression (lysozyme: p < 0.001; SLPI: p = 0.01). These results suggest that AMPs may play an important role in the anti-fibrotic response, regulating the effect of pro-fibrotic cytokines. In addition, levels of lysozyme in BAL may be a potential biomarker to predict the progression in F-ILD patients.


Bronchoalveolar Lavage Fluid , Lung Diseases, Interstitial , Muramidase , Secretory Leukocyte Peptidase Inhibitor , Humans , Muramidase/metabolism , Male , Female , Middle Aged , Secretory Leukocyte Peptidase Inhibitor/metabolism , Bronchoalveolar Lavage Fluid/chemistry , Lung Diseases, Interstitial/metabolism , Lung Diseases, Interstitial/pathology , Aged , Cytokines/metabolism , Adult , Biomarkers , Bronchoalveolar Lavage , Leukocytes, Mononuclear/metabolism
5.
Front Immunol ; 15: 1328781, 2024.
Article En | MEDLINE | ID: mdl-38550597

Metabolic changes are coupled with alteration in protein glycosylation. In this review, we will focus on macrophages that are pivotal in the pathogenesis of pulmonary fibrosis and sarcoidosis and thanks to their adaptable metabolism are an attractive therapeutic target. Examples presented in this review demonstrate that protein glycosylation regulates metabolism-driven immune responses in macrophages, with implications for fibrotic processes and granuloma formation. Targeting proteins that regulate glycosylation, such as fucosyltransferases, neuraminidase 1 and chitinase 1 could effectively block immunometabolic changes driving inflammation and fibrosis, providing novel avenues for therapeutic interventions.


Lung Diseases, Interstitial , Pulmonary Fibrosis , Sarcoidosis , Humans , Glycosylation , Lung Diseases, Interstitial/metabolism , Pulmonary Fibrosis/etiology , Sarcoidosis/metabolism , Fibrosis
6.
J Clin Invest ; 134(2)2024 Jan 16.
Article En | MEDLINE | ID: mdl-38226623

Mutations in ATP-binding cassette A3 (ABCA3), a phospholipid transporter critical for surfactant homeostasis in pulmonary alveolar type II epithelial cells (AEC2s), are the most common genetic causes of childhood interstitial lung disease (chILD). Treatments for patients with pathological variants of ABCA3 mutations are limited, in part due to a lack of understanding of disease pathogenesis resulting from an inability to access primary AEC2s from affected children. Here, we report the generation of AEC2s from affected patient induced pluripotent stem cells (iPSCs) carrying homozygous versions of multiple ABCA3 mutations. We generated syngeneic CRISPR/Cas9 gene-corrected and uncorrected iPSCs and ABCA3-mutant knockin ABCA3:GFP fusion reporter lines for in vitro disease modeling. We observed an expected decreased capacity for surfactant secretion in ABCA3-mutant iPSC-derived AEC2s (iAEC2s), but we also found an unexpected epithelial-intrinsic aberrant phenotype in mutant iAEC2s, presenting as diminished progenitor potential, increased NFκB signaling, and the production of pro-inflammatory cytokines. The ABCA3:GFP fusion reporter permitted mutant-specific, quantifiable characterization of lamellar body size and ABCA3 protein trafficking, functional features that are perturbed depending on ABCA3 mutation type. Our disease model provides a platform for understanding ABCA3 mutation-mediated mechanisms of alveolar epithelial cell dysfunction that may trigger chILD pathogenesis.


ATP-Binding Cassette Transporters , Lung Diseases, Interstitial , Pluripotent Stem Cells , Humans , Alveolar Epithelial Cells/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Lung/pathology , Lung Diseases, Interstitial/genetics , Lung Diseases, Interstitial/metabolism , Lung Diseases, Interstitial/pathology , Mutation , Pluripotent Stem Cells/metabolism , Surface-Active Agents/metabolism
7.
Respir Res ; 24(1): 320, 2023 Dec 18.
Article En | MEDLINE | ID: mdl-38111019

BACKGROUND: Pulmonary Langerhans cell histiocytosis (PLCH) is a rare interstitial lung disease (ILD) associated with smoking, whose definitive diagnosis requires the exclusion of other forms of ILD and a compatible surgical lung biopsy. Bronchoalveolar lavage (BAL) is commonly proposed for the diagnosis of ILD, including PLCH, but the diagnostic value of this technique is limited. Here, we have analyzed the levels of a panel of cytokines and chemokines in BAL from PLCH patients, in order to identify a distinct immune profile to discriminate PLCH from other smoking related-ILD (SR-ILD), and comparing the results with idiopathic pulmonary fibrosis (IPF) as another disease in which smoking is considered a risk factor. METHODS: BAL samples were collected from thirty-six patients with different ILD, including seven patients with PLCH, sixteen with SR-ILD and thirteen with IPF. Inflammatory profiles were analyzed using the Human Cytokine Membrane Antibody Array. Principal component analysis (PCA) was performed to reduce dimensionality and protein-protein interaction (PPI) network analysis using STRING 11.5 database were conducted. Finally, Random forest (RF) method was used to build a prediction model. RESULTS: We have found significant differences (p < 0.05) on thirty-two cytokines/chemokines when comparing BAL from PLCH patients with at least one of the other ILD. Four main groups of similarly regulated cytokines were established, identifying distinct sets of markers for each cluster. Exploratory analysis using PCA (principal component analysis) showed clustering and separation of patients, with the two first components capturing 69.69% of the total variance. Levels of TARC/CCL17, leptin, oncostatin M (OSM) and IP-10/CXCL10 were associated with lung function parameters, showing positive correlation with FVC. Finally, random forest (RF) algorithm demonstrates that PLCH patients can be differentiated from the other ILDs based solely on inflammatory profile (accuracy 96.25%). CONCLUSIONS: Our results show that patients with PLCH exhibit a distinct BAL immune profile to SR-ILD and IPF. PCA analysis and RF model identify a specific immune profile useful for discriminating PLCH.


Histiocytosis, Langerhans-Cell , Idiopathic Pulmonary Fibrosis , Lung Diseases, Interstitial , Humans , Bronchoalveolar Lavage Fluid , Lung Diseases, Interstitial/diagnosis , Lung Diseases, Interstitial/etiology , Lung Diseases, Interstitial/metabolism , Histiocytosis, Langerhans-Cell/diagnosis , Histiocytosis, Langerhans-Cell/pathology , Smoking/adverse effects , Cytokines , Immunoglobulins , Chemokines
8.
Respir Res ; 24(1): 318, 2023 Dec 17.
Article En | MEDLINE | ID: mdl-38105232

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a progressive interstitial lung disease (ILD) with unknown etiology, characterized by sustained damage repair of epithelial cells and abnormal activation of fibroblasts, the underlying mechanism of the disease remains elusive. METHODS: To evaluate the role of Tuftelin1 (TUFT1) in IPF and elucidate its molecular mechanism. We investigated the level of TUFT1 in the IPF and bleomycin-induced mouse models and explored the influence of TUFT1 deficiency on pulmonary fibrosis. Additionally, we explored the effect of TUFT1 on the cytoskeleton and illustrated the relationship between stress fiber and pulmonary fibrosis. RESULTS: Our results demonstrated a significant upregulation of TUFT1 in IPF and the bleomycin (BLM)-induced fibrosis model. Disruption of TUFT1 exerted inhibitory effects on pulmonary fibrosis in both in vivo and in vitro. TUFT1 facilitated the assembly of microfilaments in A549 and MRC-5 cells, with a pronounced association between TUFT1 and Neuronal Wiskott-Aldrich syndrome protein (N-WASP) observed during microfilament formation. TUFT1 can promote the phosphorylation of tyrosine residue 256 (Y256) of the N-WASP (pY256N-WASP). Furthermore, TUFT1 promoted transforming growth factor-ß1 (TGF-ß1) induced fibroblast activation by increasing nuclear translocation of pY256N-WASP in fibroblasts, while wiskostatin (Wis), an N-WASP inhibitor, suppressed these processes. CONCLUSIONS: Our findings suggested that TUFT1 plays a critical role in pulmonary fibrosis via its influence on stress fiber, and blockade of TUFT1 effectively reduces pro-fibrotic phenotypes. Pharmacological targeting of the TUFT1-N-WASP axis may represent a promising therapeutic approach for pulmonary fibrosis.


Idiopathic Pulmonary Fibrosis , Lung Diseases, Interstitial , Animals , Mice , Bleomycin/toxicity , Fibroblasts/metabolism , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/metabolism , Lung/metabolism , Lung Diseases, Interstitial/metabolism , Mice, Inbred C57BL , Stress Fibers/metabolism , Transforming Growth Factor beta1/pharmacology
9.
J Transl Med ; 21(1): 857, 2023 11 27.
Article En | MEDLINE | ID: mdl-38012636

BACKGROUND: The prognosis of patients with lung cancer accompanied by interstitial pneumonia is poorer than that of patients with lung cancer but without interstitial pneumonia. Moreover, the available therapeutic interventions for lung cancer patients with interstitial pneumonia are limited. Therefore, a new treatment strategy for these patients is required. The aim of the present study was to investigate the pathophysiological relationship between interstitial pneumonia and lung cancer and explore potential therapeutic agents. METHODS: A novel hybrid murine model of lung cancer with interstitial pneumonia was established via bleomycin-induced pulmonary fibrosis followed by orthotopic lung cancer cell transplantation into the lungs. Changes in tumor progression, lung fibrosis, RNA expression, cytokine levels, and tumor microenvironment in the lung cancer with interstitial pneumonia model were investigated, and therapeutic agents were examined. Additionally, clinical data and samples from patients with lung cancer accompanied by interstitial pneumonia were analyzed to explore the potential clinical significance of the findings. RESULTS: In the lung cancer with interstitial pneumonia model, accelerated tumor growth was observed based on an altered tumor microenvironment. RNA sequencing analysis revealed upregulation of the hypoxia-inducible factor 1 signaling pathway. These findings were consistent with those obtained for human samples. Moreover, we explored whether ascorbic acid could be an alternative treatment for lung cancer with interstitial pneumonia to avoid the disadvantages of hypoxia-inducible factor 1 inhibitors. Ascorbic acid successfully downregulated the hypoxia-inducible factor 1 signaling pathway and inhibited tumor progression and lung fibrosis. CONCLUSIONS: The hypoxia-inducible factor 1 pathway is critical in lung cancer with interstitial pneumonia and could be a therapeutic target for mitigating interstitial pneumonia-mediated lung cancer progression.


Hypoxia-Inducible Factor 1, alpha Subunit , Lung Diseases, Interstitial , Lung Neoplasms , Pneumonia , Pulmonary Fibrosis , Animals , Humans , Mice , Ascorbic Acid , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung/pathology , Lung Diseases, Interstitial/complications , Lung Diseases, Interstitial/metabolism , Lung Diseases, Interstitial/pathology , Lung Neoplasms/genetics , Pulmonary Fibrosis/pathology , Tumor Microenvironment
10.
Matrix Biol ; 124: 1-7, 2023 Dec.
Article En | MEDLINE | ID: mdl-37922998

Elastin is a long-lived fibrous protein that is abundant in the extracellular matrix of the lung. Elastic fibers provide the lung the characteristic elasticity during inhalation with recoil during exhalation thereby ensuring efficient gas exchange. Excessive deposition of elastin and other extracellular matrix proteins reduces lung compliance by impairing ventilation and compromising gas exchange. Notably, the degree of elastosis is associated with the progressive decline in lung function and survival in patients with interstitial lung diseases. Currently there are no proven therapies which effectively reduce the elastin burden in the lung nor prevent dysregulated elastosis. This review describes elastin's role in the healthy lung, summarizes elastosis in pulmonary diseases, and evaluates the current understanding of elastin regulation and dysregulation with the goal of guiding future research efforts to develop novel and effective therapies.


Lung Diseases, Interstitial , Lung , Humans , Lung/metabolism , Lung Diseases, Interstitial/metabolism , Fibrosis , Elastin , Elastic Tissue/metabolism
11.
Am J Physiol Cell Physiol ; 325(5): C1190-C1200, 2023 Nov 01.
Article En | MEDLINE | ID: mdl-37661917

Interstitial lung diseases can result in poor patient outcomes, especially in idiopathic pulmonary fibrosis (IPF), a severe interstitial lung disease with unknown causes. The lack of treatment options requires further understanding of the pathological process/mediators. Membrane-associated RING-CH 8 (MARCH8) has been implicated in immune function regulation and inflammation, however, its role in the development of pulmonary fibrosis and particularly the fibroblast to myofibroblast transition (FMT) remains a gap in existing knowledge. In this study, we demonstrated decreased MARCH8 expression in patients with IPF compared with non-PF controls and in bleomycin-induced PF. TGF-ß dose- and time-dependently decreased MARCH8 expression in normal and IPF human lung fibroblast (HLFs), along with induction of FMT markers α-SMA, collagen type I (Col-1), and fibronectin (FN). Interestingly, overexpression of MARCH8 significantly suppressed TGF-ß-induced expression of α-SMA, Col-1, and FN. By contrast, the knockdown of MARCH8 using siRNA upregulated basal expression of α-SMA/Col-1/FN. Moreover, MARCH8 knockdown enhanced TGF-ß-induced FMT marker expression. These data clearly show that MARCH8 is a critical "brake" for FMT and potentially affects PF. We further found that TGF-ß suppressed MARCH8 mRNA expression and the proteasome inhibitor MG132 failed to block MARCH8 decrease induced by TGF-ß. Conversely, TGF-ß decreases mRNA levels of MARCH8 in a dose- and time-dependent manner, suggesting the transcriptional regulation of MARCH8 by TGF-ß. Mechanistically, MARCH8 overexpression suppressed TGF-ß-induced Smad2/3 phosphorylation, which may account for the observed effects. Taken together, this study demonstrated an unrecognized role of MARCH8 in negatively regulating FMT and profibrogenic responses relevant to interstitial lung diseases.NEW & NOTEWORTHY MARCH8 is an important modulator of inflammation, immunity, and other cellular processes. We found that MARCH8 expression is downregulated in the lungs of patients with idiopathic pulmonary fibrosis (IPF) and experimental models of pulmonary fibrosis. Furthermore, TGF-ß1 decreases MARCH8 transcriptionally in human lung fibroblasts (HLFs). MARCH8 overexpression blunts TGF-ß1-induced fibroblast to myofibroblast transition while knockdown of MARCH8 drives this profibrotic change in HLFs. The findings support further exploration of MARCH8 as a novel target in IPF.


Idiopathic Pulmonary Fibrosis , Lung Diseases, Interstitial , Humans , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/pharmacology , Myofibroblasts , Down-Regulation , Lung/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Fibroblasts/metabolism , Lung Diseases, Interstitial/metabolism , Transforming Growth Factor beta/metabolism , Bleomycin/pharmacology , Inflammation/metabolism , RNA, Messenger/metabolism
12.
Semin Perinatol ; 47(6): 151811, 2023 10.
Article En | MEDLINE | ID: mdl-37775368

Bronchopulmonary dysplasia (BPD) is a multi-factorial disease that results from multiple clinical factors, including lung immaturity, mechanical ventilation, oxidative stress, pulmonary congestion due to increasing cardiac blood shunting, nutritional and immunological factors. Twin studies have indicated that susceptibility to BPD can be strongly inherited in some settings. Studies have reported associations between common genetic variants and BPD in preterm infants. Recent genomic studies have highlighted a potential role for molecular pathways involved in inflammation and lung development in affected infants. Rare mutations in genes encoding the lipid transporter ATP-binding cassette, sub-family A, member 3 (ABCA3 gene) which is involved in surfactant synthesis in alveolar type II cells, as well as surfactant protein B (SFTPB) and C (SFTPC) can also result in severe form of neonatal-onset interstitial lung diseases and may also potentially affect the course of BPD. This chapter summarizes the current state of knowledge on the genetics of BPD.


Bronchopulmonary Dysplasia , Lung Diseases, Interstitial , Infant , Infant, Newborn , Humans , Bronchopulmonary Dysplasia/genetics , Infant, Premature , Lung , Lung Diseases, Interstitial/genetics , Lung Diseases, Interstitial/metabolism , Surface-Active Agents
13.
Sci Rep ; 13(1): 13076, 2023 08 11.
Article En | MEDLINE | ID: mdl-37567908

Chronic obstructive pulmonary disease (COPD) and interstitial lung disease (ILD) are clinically and molecularly heterogeneous diseases. We utilized clustering and integrative network analyses to elucidate roles for microRNAs (miRNAs) and miRNA isoforms (isomiRs) in COPD and ILD pathogenesis. Short RNA sequencing was performed on 351 lung tissue samples of COPD (n = 145), ILD (n = 144) and controls (n = 64). Five distinct subclusters of samples were identified including 1 COPD-predominant cluster and 2 ILD-predominant clusters which associated with different clinical measurements of disease severity. Utilizing 262 samples with gene expression and SNP microarrays, we built disease-specific genetic and expression networks to predict key miRNA regulators of gene expression. Members of miR-449/34 family, known to promote airway differentiation by repressing the Notch pathway, were among the top connected miRNAs in both COPD and ILD networks. Genes associated with miR-449/34 members in the disease networks were enriched among genes that increase in expression with airway differentiation at an air-liquid interface. A highly expressed isomiR containing a novel seed sequence was identified at the miR-34c-5p locus. 47% of the anticorrelated predicted targets for this isomiR were distinct from the canonical seed sequence for miR-34c-5p. Overexpression of the canonical miR-34c-5p and the miR-34c-5p isomiR with an alternative seed sequence down-regulated NOTCH1 and NOTCH4. However, only overexpression of the isomiR down-regulated genes involved in Ras signaling such as CRKL and GRB2. Overall, these findings elucidate molecular heterogeneity inherent across COPD and ILD patients and further suggest roles for miR-34c in regulating disease-associated gene-expression.


Lung Diseases, Interstitial , MicroRNAs , Pulmonary Disease, Chronic Obstructive , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Lung/pathology , Lung Diseases, Interstitial/metabolism , Genomics
14.
Inflammation ; 46(6): 2120-2131, 2023 Dec.
Article En | MEDLINE | ID: mdl-37561311

The effectiveness of corticosteroids (GCs) varies greatly in interstitial lung diseases (ILDs). In this study, we aimed to compare the gene expression profiles of patients with cryptogenic organizing pneumonia (COP), idiopathic pulmonary fibrosis (IPF), and non-specific interstitial pneumonia (NSIP) and identify the molecules and pathways responsible for GCs sensitivity in ILDs. Three datasets (GSE21411, GSE47460, and GSE32537) were selected. Differentially expressed genes (DEGs) among COP, IPF, NSIP, and healthy control (CTRL) groups were identified. Functional enrichment analysis and protein-protein interaction network analysis were performed to examine the potential functions of DEGs. There were 128 DEGs when COP versus CTRL, 257 DEGs when IPF versus CTRL, 205 DEGs when NSIP versus CTRL, and 270 DEGs when COP versus IPF. The DEGs in different ILDs groups were mainly enriched in the inflammatory response. Further pathway analysis showed that "interleukin (IL)-17 signaling pathway" (hsa04657) and "tumor necrosis factor (TNF) signaling pathway" were associated with different types of ILDs. A total of 10 genes associated with inflammatory response were identified as hub genes and their expression levels in the IPF group were higher than those in the COP group. Finally, we identified two GCs' response-related differently expressed genes (FOSL1 and DDIT4). Our bioinformatics analysis demonstrated that the inflammatory response played a pathogenic role in the progression of ILDs. We also illustrated that the inflammatory reaction was more severe in the IPF group compared to the COP group and identified two GCs' response-related differently expressed genes (FOSL1 and DDIT4) in ILDs.


Idiopathic Interstitial Pneumonias , Idiopathic Pulmonary Fibrosis , Lung Diseases, Interstitial , Humans , Lung Diseases, Interstitial/drug therapy , Lung Diseases, Interstitial/genetics , Lung Diseases, Interstitial/metabolism , Idiopathic Interstitial Pneumonias/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Steroids/metabolism , Inflammation/metabolism , Lung/metabolism
15.
Front Immunol ; 14: 1098602, 2023.
Article En | MEDLINE | ID: mdl-37409133

Background: Krebs von den Lungen 6 (KL-6) is a potential biomarker for determining the severity of interstitial lung disease (ILD) in patients with connective tissue disease (CTD). Whether KL-6 levels can be affected by potential confounders such as underlying CTD patterns, patient-associated demographics, and comorbidities needs further investigation. Methods: From the database created by Xiangya Hospital, 524 patients with CTD, with or without ILD, were recruited for this retrospective analysis. Recorded data included demographic information, comorbidities, inflammatory biomarkers, autoimmune antibodies, and the KL-6 level at admission. Results of CT and pulmonary function tests were collected one week before or after KL-6 measurements. The percent of predicted diffusing capacity of the lung for carbon monoxide (DLCO%) and computed tomography (CT) scans were used to determine the severity of ILD. Results: Univariate linear regression analysis showed that BMI, lung cancer, TB, lung infections, underlying CTD type, white blood cell (WBC) counts, neutrophil (Neu) counts, and hemoglobin (Hb) were related to KL-6 levels. Multiple linear regression confirmed that Hb and lung infections could affect KL-6 levels independently; the ß were 9.64 and 315.93, and the P values were 0.015 and 0.039, respectively. CTD-ILD patients had higher levels of KL-6 (864.9 vs 463.9, P < 0.001) than those without ILD. KL-6 levels were closely correlated to the severity of ILD assessed both by CT and DLCO%. Additionally, we found that KL-6 level was an independent predictive factor for the presence of ILD and further constructed a decision tree model to rapidly determine the risk of developing ILD among CTD patients. Conclusion: KL-6 is a potential biomarker for gauging the incidence and severity of ILD in CTD patients. To use this typical value of KL-6, however, doctors should take Hb and the presence of lung infections into account.


Connective Tissue Diseases , Lung Diseases, Interstitial , Humans , Retrospective Studies , Connective Tissue Diseases/complications , Connective Tissue Diseases/epidemiology , Lung Diseases, Interstitial/metabolism , Lung/metabolism , Biomarkers
16.
Metabolism ; 144: 155587, 2023 07.
Article En | MEDLINE | ID: mdl-37156409

BACKGROUND: Systemic sclerosis (SSc) is a chronic and systemic autoimmune disease marked by the skin and visceral fibrosis. Metabolic alterations have been found in SSc patients; however, serum metabolomic profiling has not been thoroughly conducted. Our study aimed to identify alterations in the metabolic profile in both SSc patients before and during treatment, as well as in mouse models of fibrosis. Furthermore, the associations between metabolites and clinical parameters and disease progression were explored. METHODS: High-performance liquid chromatography quadrupole time-of-flight mass spectrometry (HPLC-Q-TOF-MS)/MS was performed in the serum of 326 human samples and 33 mouse samples. Human samples were collected from 142 healthy controls (HC), 127 newly diagnosed SSc patients without treatment (SSc baseline), and 57 treated SSc patients (SSc treatment). Mouse serum samples were collected from 11 control mice (NaCl), 11 mice with bleomycin (BLM)-induced fibrosis and 11 mice with hypochlorous acid (HOCl)-induced fibrosis. Both univariate analysis and multivariate analysis (orthogonal partial least-squares discriminate analysis (OPLS-DA)) were conducted to unravel differently expressed metabolites. KEGG pathway enrichment analysis was performed to characterize the dysregulated metabolic pathways in SSc. Associations between metabolites and clinical parameters of SSc patients were identified by Pearson's or Spearman's correlation analysis. Machine learning (ML) algorithms were applied to identify the important metabolites that have the potential to predict the progression of skin fibrosis. RESULTS: The newly diagnosed SSc patients without treatment showed a unique serum metabolic profile compared to HC. Treatment partially corrected the metabolic changes in SSc. Some metabolites (phloretin 2'-O-glucuronide, retinoyl b-glucuronide, all-trans-retinoic acid, and betaine) and metabolic pathways (starch and sucrose metabolism, proline metabolism, androgen and estrogen metabolism, and tryptophan metabolism) were dysregulated in new-onset SSc, but restored upon treatment. Some metabolic changes were associated with treatment response in SSc patients. Metabolic changes observed in SSc patients were mimicked in murine models of SSc, indicating that they may reflect general metabolic changes associated with fibrotic tissue remodeling. Several metabolic changes were associated with SSc clinical parameters. The levels of allysine and all-trans-retinoic acid were negatively correlated, while D-glucuronic acid and hexanoyl carnitine were positively correlated with modified Rodnan skin score (mRSS). In addition, a panel of metabolites including proline betaine, phloretin 2'-O-glucuronide, gamma-linolenic acid and L-cystathionine were associated with the presence of interstitial lung disease (ILD) in SSc. Specific metabolites identified by ML algorithms, such as medicagenic acid 3-O-b-D-glucuronide, 4'-O-methyl-(-)-epicatechin-3'-O-beta-glucuronide, valproic acid glucuronide, have the potential to predict the progression of skin fibrosis. CONCLUSIONS: Serum of SSc patients demonstrates profound metabolic changes. Treatment partially restored the metabolic changes in SSc. Moreover, certain metabolic changes were associated with clinical manifestations such as skin fibrosis and ILD, and could predict the progression of skin fibrosis.


Lung Diseases, Interstitial , Scleroderma, Systemic , Humans , Mice , Animals , Glucuronides/adverse effects , Lung Diseases, Interstitial/complications , Lung Diseases, Interstitial/metabolism , Scleroderma, Systemic/metabolism , Fibrosis , Biomarkers , Tretinoin/adverse effects
17.
Int J Mol Sci ; 24(8)2023 Apr 20.
Article En | MEDLINE | ID: mdl-37108718

ATP-binding cassette subfamily A member 3 (ABCA3) is a lipid transporter within alveolar type II cells. Patients with bi-allelic variants in ABCA3 may suffer from a variable severity of interstitial lung disease. We characterized and quantified ABCA3 variants' overall lipid transport function by assessing the in vitro impairment of its intracellular trafficking and pumping activity. We expressed the results relative to the wild type, integrated the quantitative readouts from eight different assays and used newly generated data combined with previous results to correlate the variants' function and clinical phenotype. We differentiated normal (within 1 normalized standard deviation (nSD) of the wild-type mean), impaired (within 1 to 3 nSD) and defective (beyond 3 nSD) variants. The transport of phosphatidylcholine from the recycling pathway into ABCA3+ vesicles proved sensitive to the variants' dysfunction. The sum of the quantitated trafficking and pumping predicted a clinical outcome. More than an approximately 50% loss of function was associated with considerable morbidity and mortality. The in vitro quantification of ABCA3 function enables detailed variant characterization, substantially improves the phenotype prediction of genetic variants and possibly supports future treatment decisions.


ATP-Binding Cassette Transporters , Lung Diseases, Interstitial , Humans , ATP-Binding Cassette Transporters/metabolism , A549 Cells , Lung Diseases, Interstitial/metabolism , Alveolar Epithelial Cells/metabolism , Phosphatidylcholines/metabolism , Mutation , Lung/metabolism
18.
Acta Histochem ; 125(3): 152024, 2023 Apr.
Article En | MEDLINE | ID: mdl-36958084

Pulmonary fibrosis is a severe condition in interstitial lung diseases (ILD) such as idiopathic pulmonary fibrosis (IPF) and systemic sclerosis-ILD, where the underlying mechanism is not well defined and with no curative treatments available. Serotonin (5-HT) signaling via the 5-HT2B receptor has been recognized as a promising preclinical target for fibrosis. Despite this, the involvement of the 5-HT2B receptor in fibrotic ILD is widely unexplored. This work highlights the spatial pulmonary distribution of the 5-HT2B receptor in patients with IPF and systemic sclerosis-ILD. We show that the 5-HT2B receptor is located in typical pathological structures e.g. honeycomb cysts and weakly in fibroblast foci. Together with immunohistochemistry and immunofluorescence stainings of patient derived distal lung tissues, we identified cell targets for 5-HT2B receptor interference in type II alveolar epithelial cells, endothelial cells and M2 macrophages. Our results emphasize the role of 5-HT2B receptor as a target in lung fibrosis, warranting further consideration in targeting fibrotic ILDs.


Idiopathic Pulmonary Fibrosis , Lung Diseases, Interstitial , Scleroderma, Systemic , Humans , Serotonin , Endothelial Cells/metabolism , Lung Diseases, Interstitial/metabolism , Lung Diseases, Interstitial/pathology , Lung/metabolism , Fibrosis , Idiopathic Pulmonary Fibrosis/pathology , Scleroderma, Systemic/pathology
19.
Sci Rep ; 13(1): 1839, 2023 02 01.
Article En | MEDLINE | ID: mdl-36725894

Oxygenated hemoglobin (OxyHem) in arterial blood may reflect disease severity in patients with systemic sclerosis (SSc). The aim of this study was to analyze the predictive value of OxyHem in SSc patients screened for pulmonary hypertension (PH). OxyHem (g/dl) was measured by multiplying the concentration of hemoglobin with fractional oxygen saturation in arterialized capillary blood. Prognostic power was compared with known prognostic parameters in SSc using uni- and multivariable analysis. A total of 280 SSc patients were screened, 267 were included in the analysis. No signs of pulmonary vascular disease were found in 126 patients, while 141 patients presented with mean pulmonary arterial pressure ≥ 21 mmHg. Interstitial lung disease (ILD) was identified in 70 patients. Low OxyHem ≤ 12.5 g/dl at baseline was significantly associated with worse survival (P = 0.046). In the multivariable analysis presence of ILD, age ≥ 60 years and diffusion capacity for carbon monoxide (DLCO) ≤ 65% were negatively associated with survival. The combination of low DLCO and low OxyHem at baseline could predict PH at baseline (sensitivity 76.1%). This study detected for the first time OxyHem ≤ 12.5 g/dl as a prognostic predictor in SSc patients. Further studies are needed to confirm these results.


Oxyhemoglobins , Scleroderma, Systemic , Humans , Middle Aged , Hypertension, Pulmonary/metabolism , Lung/metabolism , Lung Diseases, Interstitial/metabolism , Prognosis , Scleroderma, Systemic/metabolism , Oxyhemoglobins/metabolism
20.
JCI Insight ; 8(3)2023 02 08.
Article En | MEDLINE | ID: mdl-36752201

Patients with progressive fibrosing interstitial lung diseases (PF-ILDs) carry a poor prognosis and have limited therapeutic options. A hallmark feature is fibroblast resistance to apoptosis, leading to their persistence, accumulation, and excessive deposition of extracellular matrix. A complex balance of the B cell lymphoma 2 (BCL-2) protein family controlling the intrinsic pathway of apoptosis and fibroblast reliance on antiapoptotic proteins has been hypothesized to contribute to this resistant phenotype. Examination of lung tissue from patients with PF-ILD (idiopathic pulmonary fibrosis and silicosis) and mice with PF-ILD (repetitive bleomycin and silicosis) showed increased expression of antiapoptotic BCL-2 family members in α-smooth muscle actin-positive fibroblasts, suggesting that fibroblasts from fibrotic lungs may exhibit increased susceptibility to inhibition of antiapoptotic BCL-2 family members BCL-2, BCL-XL, and BCL-W with the BH3 mimetic ABT-263. We used 2 murine models of PF-ILD to test the efficacy of ABT-263 in reversing established persistent pulmonary fibrosis. Treatment with ABT-263 induced fibroblast apoptosis, decreased fibroblast numbers, and reduced lung collagen levels, radiographic disease, and histologically evident fibrosis. Our studies provide insight into how fibroblasts gain resistance to apoptosis and become sensitive to the therapeutic inhibition of antiapoptotic proteins. By targeting profibrotic fibroblasts, ABT-263 offers a promising therapeutic option for PF-ILDs.


Idiopathic Pulmonary Fibrosis , Lung Diseases, Interstitial , Silicosis , Mice , Animals , Apoptosis Regulatory Proteins/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Apoptosis/genetics , Lung Diseases, Interstitial/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Fibroblasts/metabolism , Silicosis/metabolism
...