Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 252
1.
Int J Med Sci ; 21(6): 1079-1090, 2024.
Article En | MEDLINE | ID: mdl-38774751

Idiopathic pulmonary fibrosis (IPF) is a rare, chronic and progressively worsening lung disease that poses a significant threat to patient prognosis, with a mortality rate exceeding that of some common malignancies. Effective methods for early diagnosis and treatment remain for this condition are elusive. In our study, we used the GEO database to access second-generation sequencing data and associated clinical information from IPF patients. By utilizing bioinformatics techniques, we identified crucial disease-related genes and their biological functions, and characterized their expression patterns. Furthermore, we mapped out the immune landscape of IPF, which revealed potential roles for novel kinase 1 and CD8+T cells in disease progression and outcome. These findings can aid the development of new strategies for the clinical diagnosis and treatment of IPF.


CD8-Positive T-Lymphocytes , Idiopathic Pulmonary Fibrosis , Humans , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/immunology , Idiopathic Pulmonary Fibrosis/pathology , Idiopathic Pulmonary Fibrosis/drug therapy , CD8-Positive T-Lymphocytes/immunology , Computational Biology , Disease Progression , Prognosis
2.
Int Immunopharmacol ; 135: 112269, 2024 Jun 30.
Article En | MEDLINE | ID: mdl-38781610

Idiopathic pulmonary fibrosis (IPF) presents a challenging progression characterized by lung tissue scarring and abnormal extracellular matrix deposition. This review examines the influence of immune responses, emphasizing their complex role in initiating and perpetuating fibrosis. It highlights how metabolic pathways modulate immune cell function during IPF. Immune cell modulation holds promise in managing pulmonary fibrosis (PF). Inhibiting neutrophil recruitment and monitoring mast cell levels offer insights into PF progression. Low-dose IL-2 therapy and regulation of fibroblast recruitment present potential therapeutic avenues, while the role of innate lymphoid cells (ILC2s) in allergic lung inflammation sheds light on disease mechanisms. The review focuses on metabolic reprogramming's role in shaping immune cell function during IPF progression. While some immune cells use glycolysis for pro-inflammatory responses, others favor fatty acid oxidation for regulatory functions. Targeting specialized pro-resolving lipid mediators (SPMs) presents significant potential for managing fibrotic disorders. Additionally, it highlights the pivotal role of amino acid metabolism in synthesizing serine and glycine as crucial regulators of collagen production and exploring the interconnectedness of lipid metabolism, mitochondrial dysfunction, and adipokines in driving fibrotic processes. Moreover, the review discusses the impact of metabolic disorders such as obesity and diabetes on lung fibrosis. Advocating for a holistic approach, it emphasizes the importance of considering this interplay between immune cell function and metabolic pathways in developing effective and personalized treatments for IPF.


Idiopathic Pulmonary Fibrosis , Humans , Idiopathic Pulmonary Fibrosis/immunology , Idiopathic Pulmonary Fibrosis/metabolism , Animals , Lung/immunology , Lung/pathology , Lung/metabolism , Immunity, Innate , Lipid Metabolism
3.
Int Immunopharmacol ; 132: 111999, 2024 May 10.
Article En | MEDLINE | ID: mdl-38581994

Idiopathic pulmonary fibrosis (IPF) is a heterogeneous group of lung diseases with different etiologies and characterized by progressive fibrosis. This disease usually causes pulmonary structural remodeling and decreased pulmonary function. The median survival of IPF patients is 2-5 years. Predominantly accumulation of type II innate immune cells accelerates fibrosis progression by secreting multiple pro-fibrotic cytokines. Group 2 innate lymphoid cells (ILC2) and monocytes/macrophages play key roles in innate immunity and aggravate the formation of pro-fibrotic environment. As a potent immunosuppressant, tacrolimus has shown efficacy in alleviating the progression of pulmonary fibrosis. In this study, we found that tacrolimus is capable of suppressing ILC2 activation, monocyte differentiation and the interaction of these two cells. This effect further reduced activation of monocyte-derived macrophages (Mo-M), thus resulting in a decline of myofibroblast activation and collagen deposition. The combination of tacrolimus and nintedanib was more effective than either drug alone. This study will reveal the specific process of tacrolimus alleviating pulmonary fibrosis by regulating type II immunity, and explore the potential feasibility of tacrolimus combined with nintedanib in the treatment of pulmonary fibrosis. This project will provide new ideas for clinical optimization of anti-pulmonary fibrosis drug strategies.


Idiopathic Pulmonary Fibrosis , Immunosuppressive Agents , Mice, Inbred C57BL , Monocytes , Tacrolimus , Tacrolimus/therapeutic use , Tacrolimus/pharmacology , Animals , Monocytes/drug effects , Monocytes/immunology , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/immunology , Idiopathic Pulmonary Fibrosis/pathology , Mice , Immunosuppressive Agents/therapeutic use , Immunosuppressive Agents/pharmacology , Humans , Lymphocytes/drug effects , Lymphocytes/immunology , Immunity, Innate/drug effects , Indoles/therapeutic use , Indoles/pharmacology , Macrophages/drug effects , Macrophages/immunology , Disease Progression , Lung/pathology , Lung/drug effects , Lung/immunology , Cells, Cultured , Male , Cytokines/metabolism , Myofibroblasts/drug effects , Cell Differentiation/drug effects , Disease Models, Animal
4.
Pharmacol Res ; 203: 107178, 2024 May.
Article En | MEDLINE | ID: mdl-38583686

Idiopathic pulmonary fibrosis (IPF) is one of the most fatal chronic interstitial lung diseases with unknown pathogenesis, current treatments cannot truly reverse the progression of the disease. Pulmonary macrophages, especially bone marrow derived pro-fibrotic macrophages, secrete multiple kinds of profibrotic mediators (SPP1, CD206, CD163, IL-10, CCL18…), thus further promote myofibroblast activation and fibrosis procession. IL20Rb is a cell-surface receptor that belongs to IL-20 family. The role of IL20Rb in macrophage activation and pulmonary fibrosis remains unclear. In this study, we established a bleomycin-induced pulmonary fibrosis model, used IL4/13-inducing THP1 cells to induce profibrotic macrophage (M2-like phenotype) polarization models. We found that IL20Rb is upregulated in the progression of pulmonary fibrosis, and its absence can alleviate the progression of pulmonary fibrosis. In addition, we demonstrated that IL20Rb promote the activation of bone marrow derived profibrotic macrophages by regulating the Jak2/Stat3 and Pi3k/Akt signaling pathways. In terms of therapeutic strategy, we used IL20Rb neutralizing antibodies for animal administration, which was found to alleviate the progression of IPF. Our results suggest that IL20Rb plays a profibrotic role by promoting profibrotic macrophage polarization, and IL20Rb may become a potential therapeutic target for IPF. Neutralizing antibodies against IL20Rb may become a potential drug for the clinical treatment of IPF.


Bleomycin , Macrophage Activation , Macrophages , Animals , Humans , Male , Mice , Bleomycin/toxicity , Idiopathic Pulmonary Fibrosis/pathology , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/immunology , Janus Kinase 2/metabolism , Lung/pathology , Lung/metabolism , Lung/immunology , Lung/drug effects , Macrophages/metabolism , Macrophages/immunology , Mice, Inbred C57BL , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/chemically induced , Receptors, Interleukin/metabolism , Signal Transduction , STAT3 Transcription Factor/metabolism , THP-1 Cells
5.
Sheng Li Xue Bao ; 76(2): 346-352, 2024 Apr 25.
Article Zh | MEDLINE | ID: mdl-38658383

Programmed death-ligand 1 (PD-L1) is important in maintaining central and peripheral immune tolerance in normal tissues, mediating tumor immune escape and keeping the balance between anti- and pro-inflammatory responses. Inflammation plays an important role in inflammatory lung diseases. This article reviews the research progress and potential clinical value of PD-L1 in inflammatory lung diseases, including acute lung injury, chronic obstructive pulmonary disease, asthma and idiopathic pulmonary fibrosis.


Asthma , B7-H1 Antigen , Pulmonary Disease, Chronic Obstructive , Humans , B7-H1 Antigen/metabolism , B7-H1 Antigen/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Asthma/immunology , Acute Lung Injury/immunology , Inflammation/immunology , Idiopathic Pulmonary Fibrosis/immunology , Idiopathic Pulmonary Fibrosis/metabolism , Lung Diseases/immunology , Lung Diseases/metabolism , Animals
7.
Int Immunopharmacol ; 113(Pt A): 109424, 2022 Dec.
Article En | MEDLINE | ID: mdl-36461589

Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic lung disease of unknown cause and characterized by excessive proliferation of fibroblasts and the irregular remodeling of extracellular matrix (ECM), which ultimately cause the severe distortion of the alveolar architecture. The median survival of IPF patients is 2-5 years. IPF patients are predominantly infiltrated by M2 macrophages during the course of disease development and progression. Predominantly accumulation of M2 macrophages accelerates fibrosis progression by secreting multiple cytokines that promote fibroblast to myofibroblast transition. In the process of M2 macrophage polarization, JAK2/STAT3 signaling plays a key role, thus, targeting activated macrophages to inhibit the pro-fibrotic phenotype is considered as an approach to the potential treatment of IPF. Tacrolimus is a macrolide antibiotic that as a specific inhibitor of T-lymphocyte function and has been used widely as an immunosuppressant in human organ transplantation. In this study we explored the potential effect and mechanism of tacrolimus on pulmonary fibrosis in vivo and vitro. Here, we found that tacrolimus is capable of suppressing M2 macrophages polarization by inhibiting pro-fibrotic factors secreted by M2 macrophages. This effect further alleviates M2-induced myofibroblast activation, thus resulting in a decline of collagen deposition, pro-fibrotic cytokines secretion, recovering of lung function, ultimately relieving the progression of fibrosis in vivo. Mechanistically, we found that tacrolimus can inhibit the activation of JAK2/STAT3 signaling by targeting JAK2. Our findings indicate a potential anti-fibrotic effect of tacrolimus by regulating macrophage polarization and might be meaningful in clinical settings.


Bleomycin , Idiopathic Pulmonary Fibrosis , Macrophages , Tacrolimus , Humans , Bleomycin/adverse effects , Cytokines , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/immunology , Janus Kinase 2/immunology , Macrophages/drug effects , Macrophages/immunology , STAT3 Transcription Factor/immunology , Tacrolimus/pharmacology , Tacrolimus/therapeutic use
8.
Respir Med ; 203: 106992, 2022 11.
Article En | MEDLINE | ID: mdl-36252462

BACKGROUND: Acute exacerbation (AE) is a life-threatening clinical event that occurs during the clinical course of idiopathic pulmonary fibrosis (IPF). Several studies have reported that AE also occurs in interstitial lung disease (ILD) other than IPF. However, the incidence, clinical features, risk factors for AE, and major causes of death in antineutrophil cytoplasmic antibody (ANCA)-associated ILD (ANCA-ILD) patients have not been well established. METHODS: We retrospectively reviewed the data of 54 ANCA-ILD patients and 304 IPF patients. We investigated the frequency of AE, post-AE prognoses, risk factors for AE, and major causes of death in ANCA-ILD patients. We also compared the data of ANCA-ILD with that of IPF. RESULTS: Fourteen (25.9%) ANCA-ILD patients and 84 (27.6%) IPF patients developed AE. The median survival times (MSTs) after AE in ANCA-ILD and IPF patients were 35.5 and 60 days, respectively (p = 0.588, log-rank test). In a multivariate analysis, the percentage of predicted forced vital capacity (%FVC) [O.R. 0.750 (95% CI 0.570, 0.986), p < 0.01] and serum C-reactive protein (CRP) [O.R. 2.202 (95% CI 1.037, 4.674), p < 0.01] were independent risk factors for AE. AE was the most frequent cause of death in ANCA-ILD and IPF patients. CONCLUSION: ANCA-ILD patients could develop AE, and the frequency of AE in ANCA-ILD is similar to that in IPF. AE is the most frequent cause of death in ANCA-ILD patients. A low %FVC and a high serum CRP level were independent predictive factors for AE in ANCA-ILD. The prognosis after AE in ANCA-ILD was poor, as it was in IPF.


Antibodies, Antineutrophil Cytoplasmic , Idiopathic Pulmonary Fibrosis , Lung Diseases, Interstitial , Humans , C-Reactive Protein , Disease Progression , Idiopathic Pulmonary Fibrosis/immunology , Idiopathic Pulmonary Fibrosis/metabolism , Lung Diseases, Interstitial/immunology , Lung Diseases, Interstitial/metabolism , Prognosis , Retrospective Studies , Risk Factors
9.
J Immunol ; 208(5): 1259-1271, 2022 03 01.
Article En | MEDLINE | ID: mdl-35149532

Idiopathic pulmonary fibrosis (IPF) is an irreversible, age-related diffuse parenchymal lung disease of poorly defined etiology. Many patients with IPF demonstrate distinctive lymphocytic interstitial infiltrations within remodeled lung tissue with uncertain pathogenetic relevance. Histopathological examination of explant lung tissue of patients with IPF revealed accentuated lymphoplasmacellular accumulations in close vicinity to, or even infiltrating, remodeled lung tissue. Similarly, we found significant accumulations of B cells interfused with T cells within remodeled lung tissue in two murine models of adenoviral TGF-ß1 or bleomycin (BLM)-induced lung fibrosis. Such B cell accumulations coincided with significantly increased lung collagen deposition, lung histopathology, and worsened lung function in wild-type (WT) mice. Surprisingly, B cell-deficient µMT knockout mice exhibited similar lung tissue remodeling and worsened lung function upon either AdTGF-ß1 or BLM as for WT mice. Comparative transcriptomic profiling of sorted B cells collected from lungs of AdTGF-ß1- and BLM-exposed WT mice identified a large set of commonly regulated genes, but with significant enrichment observed for Gene Ontology terms apparently not related to lung fibrogenesis. Collectively, although we observed B cell accumulations in lungs of IPF patients as well as two experimental models of lung fibrosis, comparative profiling of characteristic features of lung fibrosis between WT and B cell-deficient mice did not support a major involvement of B cells in lung fibrogenesis in mice.


B-Lymphocytes/immunology , Idiopathic Pulmonary Fibrosis/immunology , Idiopathic Pulmonary Fibrosis/pathology , Transforming Growth Factor beta1/metabolism , Animals , Bleomycin/toxicity , Collagen/metabolism , Female , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Parenchymal Tissue/pathology , T-Lymphocytes/immunology
10.
JCI Insight ; 7(4)2022 02 22.
Article En | MEDLINE | ID: mdl-34990413

Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal disease characterized by collagen deposition within the lung interstitium. Bacterial infection is associated with increased morbidity and more rapid mortality in IPF patient populations, and pathogens such as methicillin-resistant Staphylococcus aureus (MRSA) are commonly isolated from the lungs of hospitalized patients with IPF. Despite this, the effects of fibrotic lung injury on critical immune responses to infection remain unknown. In the present study, we show that, like humans with IPF, fibrotic mice infected with MRSA exhibit increased morbidity and mortality compared with uninfected fibrotic mice. We determine that fibrosis conferred a defect in MRSA clearance compared with nonfibrotic mice, resulting from blunted innate immune responses. We show that fibrosis inhibited neutrophil intracellular killing of MRSA through impaired neutrophil elastase release and oxidative radical production. Additionally, we demonstrate that lung macrophages from fibrotic mice have impaired phagocytosis of MRSA. Our study describes potentially novel impairments of antimicrobial responses upon pulmonary fibrosis development, and our findings suggest a possible mechanism for why patients with IPF are at greater risk of morbidity and mortality related to infection.


Idiopathic Pulmonary Fibrosis/immunology , Immunity, Innate/immunology , Macrophages, Alveolar/metabolism , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Neutrophils/pathology , Pneumonia, Staphylococcal/pathology , Animals , Disease Models, Animal , Idiopathic Pulmonary Fibrosis/complications , Macrophages, Alveolar/pathology , Mice , Mice, Inbred C57BL , Neutrophils/metabolism , Phagocytosis , Pneumonia, Staphylococcal/etiology , Pneumonia, Staphylococcal/immunology
11.
J Pathol ; 256(3): 310-320, 2022 03.
Article En | MEDLINE | ID: mdl-34825713

The clinical significance of B7H3 (CD276) and its cleavage product soluble B7H3 (sB7H3) in idiopathic pulmonary fibrosis (IPF) is unknown. Mounting evidence suggests the potential utility of peripheral blood myeloid cell enumeration to predict disease outcome and indicate active lung disease. Here we hypothesized that sB7H3 is involved in regulation of circulating myeloid cells in pulmonary fibrosis. In support of this possibility, both plasma sB7H3 and B7H3+ cells were elevated in IPF patient blood samples, which correlated negatively with lung function. To analyze its function, the effects of sB7H3 on naïve or bleomycin-treated mice were examined. The results revealed that sB7H3 injection induced an influx of myeloid-derived suppressor cells (MDSCs) and Ccl2 expression in lung tissue of naïve mice, accompanied by enhanced overall inflammation. Additionally, sB7H3 caused accumulation of MDSCs in bone marrow with increased expression of inflammatory cytokines. Notably, in vitro assays revealed chemotaxis of MDSCs to sB7H3, which was dependent on TLT-2 (TREML2), a putative receptor for sB7H3. Thus, increased circulating sB7H3 and/or B7H3+ cells in IPF patient blood samples correlated with lung function decline and potential immunosuppressive status. The correlation of sB7H3 with deterioration of lung function might be due to its ability to enhance inflammation and recruitment of MDSCs into the lung and their expansion in the bone marrow, and thus potentially contribute to IPF exacerbation. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


B7 Antigens/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Lung/metabolism , Aged , Animals , B7 Antigens/genetics , B7 Antigens/toxicity , Bleomycin , Case-Control Studies , Cells, Cultured , Chemokine CCL2/metabolism , Chemotaxis , Disease Models, Animal , Disease Progression , Female , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/immunology , Idiopathic Pulmonary Fibrosis/pathology , Lung/drug effects , Lung/immunology , Lung/pathology , Male , Mice, Inbred C57BL , Middle Aged , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Receptors, Immunologic/metabolism , Signal Transduction
12.
Front Immunol ; 12: 762594, 2021.
Article En | MEDLINE | ID: mdl-34880861

Background: Idiopathic pulmonary fibrosis (IPF) has high mortality worldwide. The CD247 molecule (CD247, as known as T-cell surface glycoprotein CD3 zeta chain) has been reported as a susceptibility locus in systemic sclerosis, but its correlation with IPF remains unclear. Methods: Datasets were acquired by researching the Gene Expression Omnibus (GEO). CD247 was identified as the hub gene associated with percent predicted diffusion capacity of the lung for carbon monoxide (Dlco% predicted) and prognosis according to Pearson correlation, logistic regression, and survival analysis. Results: CD247 is significantly downregulated in patients with IPF compared with controls in both blood and lung tissue samples. Moreover, CD247 is significantly positively associated with Dlco% predicted in blood and lung tissue samples. Patients with low-expression CD247 had shorter transplant-free survival (TFS) time and more composite end-point events (CEP, death, or decline in FVC >10% over a 6-month period) compared with patients with high-expression CD247 (blood). Moreover, in the follow-up 1st, 3rd, 6th, and 12th months, low expression of CD247 was still the risk factor of CEP in the GSE93606 dataset (blood). Thirteen genes were found to interact with CD247 according to the protein-protein interaction network, and the 14 genes including CD247 were associated with the functions of T cells and natural killer (NK) cells such as PD-L1 expression and PD-1 checkpoint pathway and NK cell-mediated cytotoxicity. Furthermore, we also found that a low expression of CD247 might be associated with a lower activity of TIL (tumor-infiltrating lymphocytes), checkpoint, and cytolytic activity and a higher activity of macrophages and neutrophils. Conclusion: These results imply that CD247 may be a potential T cell-derived disease severity and prognostic biomarker for IPF.


CD3 Complex/immunology , Idiopathic Pulmonary Fibrosis/immunology , T-Lymphocytes/immunology , Aged , CD3 Complex/blood , CD3 Complex/genetics , Down-Regulation , Female , Gene Expression , Humans , Idiopathic Pulmonary Fibrosis/blood , Idiopathic Pulmonary Fibrosis/genetics , Lung/immunology , Male , Middle Aged , Myosin Light Chains/blood , Myosin Light Chains/genetics , Prognosis , Protein Interaction Maps , Severity of Illness Index
13.
Cells ; 10(11)2021 11 17.
Article En | MEDLINE | ID: mdl-34831433

Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal lung disease of unknown etiology. Immune disorders play an important role in IPF pathogenesis. Here, we show that Th9 cells differentiate and activate in the lung tissue of patients with IPF and bleomycin (BLM)-induced lung fibrosis mice. Moreover, we found that Th9 cells promote pulmonary fibrosis in two ways. On the one hand, Th9 cells promote fibroblast differentiation, activation, and collagen secretion by secreting IL-9. On the other hand, they promote differentiation of Th0 cells into Th2 cells by secreting IL-4. Th9 cells and Th2 cells can promote each other, accelerating the Th1/Th2 imbalance and eventually forming a positive feedback of pulmonary fibrosis. In addition, we found that neutralizing IL-9 in both preventive and therapeutic settings ameliorates bleomycin-induced pulmonary fibrosis. Furthermore, we identified several critical signaling pathways involved in the effect of neutralizing IL-9 on pulmonary fibrosis by proteomics study. From an immunological perspective, we elucidated the novel role and underlying mechanism of Th9 cells in pulmonary fibrosis. Our study suggested that Th9-based immunotherapy may be employed as a treatment strategy for IPF.


Idiopathic Pulmonary Fibrosis/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Antibodies, Neutralizing/metabolism , Bleomycin , Case-Control Studies , Cell Differentiation , Cell Proliferation , Collagen/metabolism , Female , Fibroblasts/pathology , Humans , Idiopathic Pulmonary Fibrosis/pathology , Interleukin-9/metabolism , Leukocytes, Mononuclear/metabolism , Lung/immunology , Lung/pathology , Lymphocyte Activation/immunology , Male , Middle Aged , Proteomics , Signal Transduction
14.
Front Immunol ; 12: 747335, 2021.
Article En | MEDLINE | ID: mdl-34804029

Interstitial lung diseases (ILDs) are a heterogeneous group of diseases characterized by varying degrees of inflammation and fibrosis of the pulmonary interstitium. The interrelations between multiple immune cells and stromal cells participate in the pathogenesis of ILDs. While fibroblasts contribute to the development of ILDs through secreting extracellular matrix and proinflammatory cytokines upon activation, T cells are major mediators of adaptive immunity, as well as inflammation and autoimmune tissue destruction in the lung of ILDs patients. Fibroblasts play important roles in modulating T cell recruitment, differentiation and function and conversely, T cells can balance fibrotic sequelae with protective immunity in the lung. A more precise understanding of the interrelation between fibroblasts and T cells will enable a better future therapeutic design by targeting this interrelationship. Here we highlight recent work on the interactions between fibroblasts and T cells in ILDs, and consider the implications of these interactions in the future development of therapies for ILDs.


Fibroblasts/pathology , Idiopathic Pulmonary Fibrosis/pathology , Lung Diseases, Interstitial/pathology , T-Lymphocytes/pathology , Animals , Humans , Idiopathic Pulmonary Fibrosis/immunology , Lung Diseases, Interstitial/immunology , T-Lymphocytes/immunology
15.
Int J Mol Sci ; 22(19)2021 Sep 28.
Article En | MEDLINE | ID: mdl-34638790

Idiopathic pulmonary fibrosis (IPF), one of the most common fibrosing interstitial lung diseases (ILD), is a chronic-age-related respiratory disease that rises from repeated micro-injury of the alveolar epithelium. Environmental influences, intrinsic factors, genetic and epigenetic risk factors that lead to chronic inflammation might be implicated in the development of IPF. The exact triggers that initiate the fibrotic response in IPF remain enigmatic, but there is now increasing evidence supporting the role of chronic exposure of viral infection. During viral infection, activation of the NLRP3 inflammasome by integrating multiple cellular and molecular signaling implicates robust inflammation, fibroblast proliferation, activation of myofibroblast, matrix deposition, and aberrant epithelial-mesenchymal function. Overall, the crosstalk of the NLRP3 inflammasome and viruses can activate immune responses and inflammasome-associated molecules in the development, progression, and exacerbation of IPF.


Idiopathic Pulmonary Fibrosis/immunology , Inflammasomes/immunology , Lung Diseases, Interstitial/immunology , Myofibroblasts/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Virus Diseases/immunology , Animals , Humans , Idiopathic Pulmonary Fibrosis/virology , Lung Diseases, Interstitial/virology
16.
Cells ; 10(10)2021 10 15.
Article En | MEDLINE | ID: mdl-34685744

Pulmonary epithelial cells are widely considered to be the first line of defence in the lung and are responsible for coordinating the innate immune response to injury and subsequent repair. Consequently, epithelial cells communicate with multiple cell types including immune cells and fibroblasts to promote acute inflammation and normal wound healing in response to damage. However, aberrant epithelial cell death and damage are hallmarks of pulmonary disease, with necrotic cell death and cellular senescence contributing to disease pathogenesis in numerous respiratory diseases such as idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD) and coronavirus disease (COVID)-19. In this review, we summarise the literature that demonstrates that epithelial damage plays a pivotal role in the dysregulation of the immune response leading to tissue destruction and abnormal remodelling in several chronic diseases. Specifically, we highlight the role of epithelial-derived damage-associated molecular patterns (DAMPs) and senescence in shaping the immune response and assess their contribution to inflammatory and fibrotic signalling pathways in the lung.


COVID-19/immunology , Epithelium/immunology , Idiopathic Pulmonary Fibrosis/immunology , Lung/immunology , Alarmins , Animals , Cellular Senescence , Coculture Techniques , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Fibrosis/metabolism , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Immunity , Inflammation/metabolism , Ligands , Necroptosis , Necrosis/pathology , Pulmonary Disease, Chronic Obstructive , SARS-CoV-2 , Signal Transduction
17.
Am J Physiol Lung Cell Mol Physiol ; 321(5): L925-L940, 2021 11 01.
Article En | MEDLINE | ID: mdl-34524907

Acute exacerbation of idiopathic pulmonary fibrosis has a poor prognosis associated with neutrophilic inflammation. Interleukin-23 is a proinflammatory cytokine involved in neutrophilic inflammation. However, little is known about its role in acute exacerbation of pulmonary fibrosis. This study was performed to determine the role of interleukin-23 in acute exacerbation of pulmonary fibrosis. For assessment of acute exacerbation of pulmonary fibrosis, mice were intratracheally administered bleomycin followed by lipopolysaccharide. Inflammatory cells, cytokine levels, and morphological morphometry of the lungs were analyzed. Cytokine levels were measured in the bronchoalveolar lavage fluid of idiopathic pulmonary fibrosis patients with or without acute exacerbation. Interleukin-23, -17A, and -22 levels were increased in the airway of mice with acute exacerbation of pulmonary fibrosis. Interleukin-23p19-deficient mice with acute exacerbation of pulmonary fibrosis had markedly reduced airway inflammation and fibrosis associated with decreased levels of interleukin-17A and -22 compared with wild-type mice. Treatment with an anti-interleukin-23 antibody attenuated airway inflammation and fibrosis and reduced interleukin-17A and -22 levels in mice with acute exacerbation of pulmonary fibrosis. T-helper type 17 cells were the predominant source of interleukin-17A in mice with acute exacerbation of pulmonary fibrosis. Interleukin-23 levels in bronchoalveolar lavage fluid tended to be higher in idiopathic pulmonary fibrosis patients with than without acute exacerbation. The data presented here suggest that interleukin-23 is essential for the development of acute exacerbation of pulmonary fibrosis and that blockade of interleukin-23 may be a new therapeutic strategy for acute exacerbation of pulmonary fibrosis.


Idiopathic Pulmonary Fibrosis/etiology , Idiopathic Pulmonary Fibrosis/immunology , Inflammation/metabolism , Interleukin-23/metabolism , Acute Disease , Animals , Idiopathic Pulmonary Fibrosis/metabolism , Inflammation/pathology , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukin-23/immunology , Interleukins/immunology , Interleukins/metabolism , Lung/immunology , Lung/pathology , Mice , Th17 Cells/immunology , Th17 Cells/metabolism
18.
J Ethnopharmacol ; 281: 114522, 2021 Dec 05.
Article En | MEDLINE | ID: mdl-34391863

ETHNOPHARMACOLOGICAL RELEVANCE: Firstly prescribed in the ancient Chinese book Jingui Yaolue, Gancao Ganjiang decoction (GGD) is a traditional Chinese herbal formula that has been widely used to treat "atrophic lung disease". GGD is a popular and widely used traditional Chinese medicine. The decoction is extracted from the dried rhizomes and roots of Glycyrrhiza uralensis Fisch. and Zingiber officinale Roscoe (2:1). AIM OF STUDY: To investigate the therapeutic effect of idiopathic pulmonary fibrosis (IPF) of GGD, a bleomycin-induced IPF murine model was used in this study. MATERIALS AND METHODS: Mice were induced by bleomycin instillation and GGD was orally administered. Changes on mice weight were recorded during the experiment. Lung weight was recorded on days 14 and 28, and pulmonary index was calculated accordingly. Pathological evaluation, including fibrosis analysis of lung tissue, was assessed by H&E and Masson staining. The expression of PD-1, p-STAT3 and IL-17A were detected by immunohistochemistry (IHC). The expression of p-STAT3 in lung tissues of mice were detected by Western blot. The level of IL-17A in lung tissue were detected by ELISA. The expression of PD-1 in CD4+ T cells in peripheral blood of mice was detected by flow cytometry. The levels of hydroxyproline and TGF-ß1 in lung tissue were detected by ELISA. The expression of E-cadherin, vimentin and α-SMA in lung tissues of mice were detected by qRT-PCR and Western blot. RESULTS: GGD can increase body weight and reduce pulmonary index in mice with pulmonary fibrosis. As such, GGD can significantly improve the inflammatory and alleviate IPF in the lung tissue of mice. GGD treatment was capable of reducing the content of PD-1 in lung tissue as well as the expression of PD-1 in CD4+ T cells in peripheral blood. Likewise, GGD was able to reduce the content of p-STAT3, IL-17A and TGF-ß1. In addition, GGD stimulation could inhibit epithelial-mesenchymal transformation (EMT) by increasing the expression of E-cadherin and reducing vimentin and α-SMA, thus reducing extracellular matrix (ECM) deposition. CONCLUSION: Our results indicate that GGD positively affects IPF by regulating PD-1/TGF-ß1/IL-17A pathway.


Anti-Inflammatory Agents/therapeutic use , Drugs, Chinese Herbal/therapeutic use , Idiopathic Pulmonary Fibrosis/drug therapy , Immunologic Factors/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacology , Bleomycin , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cadherins/genetics , Drugs, Chinese Herbal/pharmacology , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/immunology , Idiopathic Pulmonary Fibrosis/pathology , Immunologic Factors/pharmacology , Interleukin-17 , Lung/drug effects , Lung/immunology , Lung/pathology , Male , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor , Signal Transduction , Transforming Growth Factor beta1 , Vimentin/genetics
19.
Clin Immunol ; 230: 108827, 2021 09.
Article En | MEDLINE | ID: mdl-34428741

BAL cellularity and lymphocyte immunophenotyping offer insights into lung inflammatory status. Natural killer (NK) cells are efficient effector cells, producing pro-inflammatory cytokines. A better understanding of the biology of NK cells in BAL in the lungs is necessary to improve the pathogenesis of fibrotic ILD and develop prospective targeted treatments. Our aim was to analyse NK and NKT-like cell percentages in BAL from 159 patients with different ILD: f-HP, f-NSIP, IPF and CTD-ILD, to evaluate their potential diagnostic/prognostic role. BAL NK cell percentages showed significantly higher values in IPF than in f-HP and f-NSIP, while BAL NKT-like cells showed significantly lower values in the f-NSIP than the f-HP and IPF. A cut-off of 4%NK cells in BAL of IPF showed a significant difference in survival rate. It suggests a possible new marker of survival and raises the possibility of new targeted approach in treatment and management of IPF.


Killer Cells, Natural/immunology , Lung Diseases, Interstitial/immunology , Aged , Aged, 80 and over , Alveolitis, Extrinsic Allergic/immunology , Alveolitis, Extrinsic Allergic/pathology , Alveolitis, Extrinsic Allergic/physiopathology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Connective Tissue Diseases/immunology , Connective Tissue Diseases/pathology , Connective Tissue Diseases/physiopathology , Female , Fibrosis , Humans , Idiopathic Pulmonary Fibrosis/immunology , Idiopathic Pulmonary Fibrosis/pathology , Idiopathic Pulmonary Fibrosis/physiopathology , Immunophenotyping , Kaplan-Meier Estimate , Killer Cells, Natural/classification , Killer Cells, Natural/pathology , Lung/immunology , Lung/pathology , Lung/physiopathology , Lung Diseases, Interstitial/pathology , Lung Diseases, Interstitial/physiopathology , Male , Middle Aged , Natural Killer T-Cells/classification , Natural Killer T-Cells/immunology , Natural Killer T-Cells/pathology , Prognosis , Respiratory Function Tests
20.
Front Immunol ; 12: 661811, 2021.
Article En | MEDLINE | ID: mdl-34220810

Fibrotic Interstitial lung diseases (ILDs) are complex disorders of variable clinical behaviour. The majority of them cause significant morbidity, whilst Idiopathic Pulmonary Fibrosis (IPF) is recognised as the most relentless. NLRP3, AIM2, and NLRC4 inflammasomes are multiprotein complexes driving IL-1ß release; a proinflammatory and profibrotic cytokine. Several pathogenetic factors associated with IPF are identified as inflammasome activators, including increases in mtROS and bacterial burden. Mitochondrial oxidation and alterations in bacterial burden in IPF and other ILDs may lead to augmented inflammasome activity in airway macrophages (AMs). IPF (n=14), non-IPF-ILDs (n=12) patients and healthy subjects (n=12) were prospectively recruited and AMs were isolated from bronchoalveolar lavage. IL-1ß release resulting from NLRP3, AIM2 and NLRC4 inflammasomes stimulation in AMs were determined and baseline levels of mitochondrial ROS and microbial burden were also measured. Our results showed that NLRP3 was more inducible in IPF and other ILDs compared to controls. Additionally, following AIM2 activation IL-1ß release was significantly higher in IPF compared to controls, whereas similar trends were observed in Non-IPF-ILDs. NLRC4 activation was similar across groups. mtROS was significantly associated with heightened NLRP3 and AIM2 activation, and mitochondrial antioxidant treatment limited inflammasome activation. Importantly, microbial burden was linked to baseline IL-1ß release and AIM2 and IL-18 relative expression independently of mtROS. In conclusion, the above findings suggested a link between the overactivation of NLRP3 and AIM2 inflammasomes, driven by mitochondrial oxidation, in the pathogenesis of lung fibrosis while changes in the microbiota may prime the inflammasome in the lungs.


DNA-Binding Proteins/immunology , Idiopathic Pulmonary Fibrosis/immunology , Inflammasomes/drug effects , Inflammasomes/immunology , Interleukin-1beta/analysis , Macrophages/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Adult , Aged , Aged, 80 and over , Bronchoalveolar Lavage Fluid/cytology , Female , Humans , Idiopathic Pulmonary Fibrosis/physiopathology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Lipopolysaccharides/pharmacology , Lung/cytology , Lung/immunology , Macrophages/drug effects , Male , Middle Aged , Prospective Studies
...