Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.242
Filtrer
1.
AAPS PharmSciTech ; 25(6): 150, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38954161

RÉSUMÉ

Nintedanib, a primary treatment for lung fibrosis, has gathered substantial attention due to its multifaceted potential. A tyrosine kinase inhibitor, nintedanib, inhibits multiple signalling receptors, including endothelial growth factor receptor (VEGFR), platelet-derived growth factor receptor (PDGFR), and fibroblast growth factor receptor (FGFR) and ultimately inhibits fibroblast proliferation and differentiation. Therefore, nintedanib has been studied widely for other ailments like cancers and hepatic fibrosis, apart from lung disorders. Commercially, nintedanib is available as soft gelatin capsules for treatment against idiopathic pulmonary fibrosis. Since it has very low oral bioavailability (4.7%), high doses of a drug, such as 100-150 mg, are administered, which can cause problems of gastrointestinal irritation and hepatotoxicity. The article begins with exploring the mechanism of action of nintedanib, elucidating its complex interactions within cellular pathways that govern fibrotic processes. It also emphasizes the pharmacokinetics of nintedanib, clinical trial insights, and the limitations of conventional formulations. The article mainly focuses on the emerging landscape of nanoparticle-based carriers such as hybrid liposome-exosome, nano liquid crystals, discoidal polymeric, and magnetic systems, offering promising avenues to optimize drug targeting, address its efficacy issues and minimise adverse effects. However, none of these delivery systems are commercialised, and further research is required to ensure safety and effectiveness in clinical settings. Yet, as research progresses, these advanced delivery systems promise to revolutionise the treatment landscape for various fibrotic disorders and cancers, potentially improving patient outcomes and quality of life.


Sujet(s)
Systèmes de délivrance de médicaments , Indoles , Humains , Indoles/administration et posologie , Indoles/pharmacocinétique , Systèmes de délivrance de médicaments/méthodes , Animaux , Maladies pulmonaires/traitement médicamenteux , Inhibiteurs de protéines kinases/administration et posologie , Inhibiteurs de protéines kinases/pharmacocinétique , Fibrose pulmonaire idiopathique/traitement médicamenteux , Fibrose pulmonaire idiopathique/métabolisme
2.
Cells ; 13(13)2024 Jul 06.
Article de Anglais | MEDLINE | ID: mdl-38995003

RÉSUMÉ

Lung parenchymal hypoxia has emerged as a cardinal feature of idiopathic pulmonary fibrosis (IPF). Hypoxia promotes cancer cell invasion and metastasis through signaling that is dependent upon the lysophosphatidic acid (LPA) receptor, LPA1 (LPAR1). Abundant data indicate that LPA1-dependent signaling also enhances lung fibrogenesis in IPF. We recently reported that fibroblasts isolated from the lungs of individuals with IPF have an increased capacity to form subcellular matrix-degradative structures known as invadosomes, an event that correlates with the degree of lung fibrosis. We therefore hypothesized that hypoxia promotes invadosome formation in lung fibroblasts through LPA1-dependent signaling. Here, it is demonstrated that invadosome formation by fibroblasts from the lungs of individuals with advanced IPF is inhibited by both the tyrosine receptor kinase inhibitor nintedanib and inhibition of LPA1. In addition, exposure of normal human lung fibroblasts to either hypoxia or LPA increased their ability to form invadosomes. Mechanistically, the hypoxia-induced invadosome formation by lung fibroblasts was found to involve LPA1 and PDGFR-Akt signaling. We concluded that hypoxia increases the formation of invadosomes in lung fibroblasts through the LPA1 and PDGFR-Akt signaling axis, which represents a potential target for suppressing lung fibrosis.


Sujet(s)
Fibroblastes , Poumon , Podosomes , Récepteurs à l'acide phosphatidique , Transduction du signal , Humains , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Poumon/anatomopathologie , Poumon/métabolisme , Podosomes/métabolisme , Récepteurs à l'acide phosphatidique/métabolisme , Fibrose pulmonaire idiopathique/anatomopathologie , Fibrose pulmonaire idiopathique/métabolisme , Hypoxie cellulaire , Lysophospholipides/métabolisme , Indoles/pharmacologie , Protéines proto-oncogènes c-akt/métabolisme , Récepteurs aux facteurs de croissance dérivés des plaquettes/métabolisme
3.
Sci Rep ; 14(1): 15778, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38982264

RÉSUMÉ

Idiopathic pulmonary fibrosis (IPF) is the most predominant type of idiopathic interstitial pneumonia and has an increasing incidence, poor prognosis, and unclear pathogenesis. In order to investigate the molecular mechanisms underlying IPF further, we performed single-cell RNA sequencing analysis on three healthy controls and five IPF lung tissue samples. The results revealed a significant shift in epithelial cells (ECs) phenotypes in IPF, which may be attributed to the differentiation of alveolar type 2 cells to basal cells. In addition, several previously unrecognized basal cell subtypes were preliminarily identified, including extracellular matrix basal cells, which were increased in the IPF group. We identified a special population of fibroblasts that highly expressed extracellular matrix-related genes, POSTN, CTHRC1, COL3A1, COL5A2, and COL12A1. We propose that the close interaction between ECs and fibroblasts through ligand-receptor pairs may have a critical function in IPF development. Collectively, these outcomes provide innovative perspectives on the complexity and diversity of basal cells and fibroblasts in IPF and contribute to the understanding of possible mechanisms in pathological lung fibrosis.


Sujet(s)
Fibroblastes , Fibrose pulmonaire idiopathique , Analyse de séquence d'ARN , Analyse sur cellule unique , Fibrose pulmonaire idiopathique/génétique , Fibrose pulmonaire idiopathique/anatomopathologie , Fibrose pulmonaire idiopathique/métabolisme , Humains , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Analyse sur cellule unique/méthodes , Cellules épithéliales/métabolisme , Cellules épithéliales/anatomopathologie , Femelle , Mâle , Poumon/anatomopathologie , Poumon/métabolisme , Matrice extracellulaire/métabolisme , Adulte d'âge moyen
4.
Respir Res ; 25(1): 284, 2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39026235

RÉSUMÉ

Idiopathic pulmonary fibrosis is a lethal, progressive, and irreversible condition that has become a significant focus of medical research due to its increasing incidence. This rising trend presents substantial challenges for patients, healthcare providers, and researchers. Despite the escalating burden of pulmonary fibrosis, the available therapeutic options remain limited. Currently, the United States Food and Drug Administration has approved two drugs for the treatment of pulmonary fibrosis-nintedanib and pirfenidone. However, their therapeutic effectiveness is limited, and they cannot reverse the fibrosis process. Additionally, these drugs are associated with significant side effects. Myofibroblasts play a central role in the pathophysiology of pulmonary fibrosis, significantly contributing to its progression. Consequently, strategies aimed at inhibiting myofibroblast differentiation or promoting their dedifferentiation hold promise as effective treatments. This review examines the regulation of myofibroblast dedifferentiation, exploring various signaling pathways, regulatory targets, and potential pharmaceutical interventions that could provide new directions for therapeutic development.


Sujet(s)
Dédifférenciation cellulaire , Myofibroblastes , Humains , Myofibroblastes/anatomopathologie , Myofibroblastes/métabolisme , Myofibroblastes/effets des médicaments et des substances chimiques , Dédifférenciation cellulaire/effets des médicaments et des substances chimiques , Dédifférenciation cellulaire/physiologie , Animaux , Fibrose pulmonaire idiopathique/anatomopathologie , Fibrose pulmonaire idiopathique/métabolisme , Fibrose pulmonaire idiopathique/traitement médicamenteux , Transduction du signal/physiologie , Antifibrotiques/usage thérapeutique , Antifibrotiques/pharmacologie , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/métabolisme
5.
Mol Biol Cell ; 35(8): ar114, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38985514

RÉSUMÉ

Idiopathic pulmonary fibrosis (IPF), one of the most common forms of interstitial lung disease, is a poorly understood, chronic, and often fatal fibroproliferative condition with only two FDA-approved medications. Understanding the pathobiology of the fibroblast in IPF is critical to evaluating and discovering novel therapeutics. Using a decellularized lung matrix derived from patients with IPF, we generate three-dimensional hydrogels as in vitro models of lung physiology and characterize the phenotype of fibroblasts seeded into the hydrogels. When cultured in IPF extracellular matrix hydrogels, IPF fibroblasts display differential contractility compared with their normal counterparts, lose the classical myofibroblast marker α-smooth muscle actin, and increase expression of proinflammatory cytokines compared with fibroblasts seeded two-dimensionally on tissue culture dishes. We validate this proinflammatory state in fibroblast-conditioned media studies with monocytes and monocyte-derived macrophages. These findings add to a growing understanding of the lung microenvironment effect on fibroblast phenotypes, shed light on the potential role of fibroblasts as immune signaling hubs during lung fibrosis, and suggest intervention in fibroblast-immune cell cross-talk as a possible novel therapeutic avenue.


Sujet(s)
Matrice extracellulaire , Fibroblastes , Hydrogels , Fibrose pulmonaire idiopathique , Poumon , Humains , Fibrose pulmonaire idiopathique/métabolisme , Fibrose pulmonaire idiopathique/anatomopathologie , Fibroblastes/métabolisme , Poumon/anatomopathologie , Poumon/métabolisme , Matrice extracellulaire/métabolisme , Cytokines/métabolisme , Macrophages/métabolisme , Myofibroblastes/métabolisme , Inflammation/métabolisme , Inflammation/anatomopathologie , Cellules cultivées , Milieux de culture conditionnés/pharmacologie , Actines/métabolisme , Monocytes/métabolisme
6.
Eur Respir Rev ; 33(172)2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-39009409

RÉSUMÉ

Lysophosphatidic acid (LPA)-mediated activation of LPA receptor 1 (LPAR1) contributes to the pathophysiology of fibrotic diseases such as idiopathic pulmonary fibrosis (IPF) and systemic sclerosis (SSc). These diseases are associated with high morbidity and mortality despite current treatment options. The LPA-producing enzyme autotaxin (ATX) and LPAR1 activation contribute to inflammation and mechanisms underlying fibrosis in preclinical fibrotic models. Additionally, elevated levels of LPA have been detected in bronchoalveolar lavage fluid from patients with IPF and in serum from patients with SSc. Thus, ATX and LPAR1 have gained considerable interest as pharmaceutical targets to combat fibrotic disease and inhibitors of these targets have been investigated in clinical trials for IPF and SSc. The goals of this review are to summarise the current literature on ATX and LPAR1 signalling in pulmonary fibrosis and to help differentiate the novel inhibitors in development. The mechanisms of action of ATX and LPAR1 inhibitors are described and preclinical studies and clinical trials of these agents are outlined. Because of their contribution to numerous physiologic events underlying fibrotic disease, ATX and LPAR1 inhibition presents a promising therapeutic strategy for IPF, SSc and other fibrotic diseases that may fulfil unmet needs of the current standard of care.


Sujet(s)
Fibrose pulmonaire idiopathique , Phosphodiesterases , Récepteurs à l'acide phosphatidique , Transduction du signal , Humains , Récepteurs à l'acide phosphatidique/antagonistes et inhibiteurs , Récepteurs à l'acide phosphatidique/métabolisme , Animaux , Transduction du signal/effets des médicaments et des substances chimiques , Phosphodiesterases/métabolisme , Fibrose pulmonaire idiopathique/traitement médicamenteux , Fibrose pulmonaire idiopathique/métabolisme , Thérapie moléculaire ciblée , Poumon/effets des médicaments et des substances chimiques , Poumon/physiopathologie , Poumon/métabolisme , Antifibrotiques/usage thérapeutique , Lysophospholipides/métabolisme , Résultat thérapeutique , Fibrose pulmonaire/traitement médicamenteux , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/physiopathologie , Inhibiteurs de la phosphodiestérase/usage thérapeutique
7.
Mol Biol Rep ; 51(1): 795, 2024 Jul 13.
Article de Anglais | MEDLINE | ID: mdl-39001907

RÉSUMÉ

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a disease of unknown etiology characterized by a constant incidence rate. Unfortunately, effective pharmacological treatments for this condition are lacking and the identification of novel therapeutic approaches and underlying pathological mechanisms are required. This study investigated the potential of quercetin in alleviating pulmonary fibrosis by promoting autophagy and activation of the SIRT1/AMPK pathway. METHODS: Mouse models of IPF were divided into four treatment groups: control, bleomycin (BLM), quercetin (Q), and quercetin + EX-527 (Q + E) treatment. Pulmonary fibrosis was induced in the mouse models through intratracheal instillation of BLM. Various indexes were identified through histological staining, Western blotting analysis, enzyme-linked immunosorbent assay, immunohistochemistry, and transmission electron microscopy. RESULTS: Quercetin treatment ameliorated the pathology of BLM-induced pulmonary fibrosis of mice by reducing α-smooth muscle actin (α-SMA), collagen I (Col I), and collagen III (Col III) levels, and also improved the level of E-cadherin in lung tissue. Furthermore, Quercetin significantly enhanced LC3II/LC3I levels, decreased P62 expression, and increased the number of autophagosomes in lung tissue. These effects were accompanied by the activation of the SIRT1/AMPK pathway. Treatment with EX-527, an inhibitor for SIRT1, reversed all effects induced by quercetin. CONCLUSION: This study showed that quercetin could alleviate pulmonary fibrosis and improve epithelial-mesenchymal transition by acting on the SIRT1/AMPK signaling pathway, which may be achieved by regulating the level of autophagy.


Sujet(s)
AMP-Activated Protein Kinases , Autophagie , Bléomycine , Fibrose pulmonaire , Quercétine , Transduction du signal , Sirtuine-1 , Animaux , Bléomycine/effets indésirables , Quercétine/pharmacologie , Sirtuine-1/métabolisme , Autophagie/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Souris , AMP-Activated Protein Kinases/métabolisme , Fibrose pulmonaire/traitement médicamenteux , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/induit chimiquement , Fibrose pulmonaire/anatomopathologie , Modèles animaux de maladie humaine , Mâle , Poumon/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Poumon/métabolisme , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Fibrose pulmonaire idiopathique/traitement médicamenteux , Fibrose pulmonaire idiopathique/métabolisme , Fibrose pulmonaire idiopathique/induit chimiquement , Fibrose pulmonaire idiopathique/anatomopathologie , Souris de lignée C57BL
8.
Theranostics ; 14(9): 3603-3622, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948058

RÉSUMÉ

Background: Myofibroblasts (MYFs) are generally considered the principal culprits in excessive extracellular matrix deposition and scar formation in the pathogenesis of lung fibrosis. Lipofibroblasts (LIFs), on the other hand, are defined by their lipid-storing capacity and are predominantly found in the alveolar regions of the lung. They have been proposed to play a protective role in lung fibrosis. We previously reported that a LIF to MYF reversible differentiation switch occurred during fibrosis formation and resolution. In this study, we tested whether WI-38 cells, a human embryonic lung fibroblast cell line, could be used to study fibroblast differentiation towards the LIF or MYF phenotype and whether this could be relevant for idiopathic pulmonary fibrosis (IPF). Methods: Using WI-38 cells, Fibroblast (FIB) to MYF differentiation was triggered using TGF-ß1 treatment and FIB to LIF differentiation using Metformin treatment. We also analyzed the MYF to LIF and LIF to MYF differentiation by pre-treating the WI-38 cells with TGF-ß1 or Metformin respectively. We used IF, qPCR and bulk RNA-Seq to analyze the phenotypic and transcriptomic changes in the cells. We correlated our in vitro transcriptome data from WI-38 cells (obtained via bulk RNA sequencing) with the transcriptomic signature of LIFs and MYFs derived from the IPF cell atlas as well as with our own single-cell transcriptomic data from IPF patients-derived lung fibroblasts (LF-IPF) cultured in vitro. We also carried out alveolosphere assays to evaluate the ability of the proposed LIF and MYF cells to support the growth of alveolar epithelial type 2 cells. Results: WI-38 cells and LF-IPF display similar phenotypical and gene expression responses to TGF-ß1 and Metformin treatment. Bulk RNA-Seq analysis of WI-38 cells and LF-IPF treated with TGF-ß1, or Metformin indicate similar transcriptomic changes. We also show the partial conservation of the LIF and MYF signature extracted from the Habermann et al. scRNA-seq dataset in WI-38 cells treated with Metformin or TGF-ß1, respectively. Alveolosphere assays indicate that LIFs enhance organoid growth, while MYFs inhibit organoid growth. Finally, we provide evidence supporting the MYF to LIF and LIF to MYF reversible switch using WI-38 cells. Conclusions: WI-38 cells represent a versatile and reliable model to study the intricate dynamics of fibroblast differentiation towards the MYF or LIF phenotype associated with lung fibrosis formation and resolution, providing valuable insights to drive future research.


Sujet(s)
Différenciation cellulaire , Fibroblastes , Fibrose pulmonaire idiopathique , Myofibroblastes , Facteur de croissance transformant bêta-1 , Humains , Myofibroblastes/métabolisme , Fibroblastes/métabolisme , Lignée cellulaire , Fibrose pulmonaire idiopathique/anatomopathologie , Fibrose pulmonaire idiopathique/métabolisme , Facteur de croissance transformant bêta-1/métabolisme , Facteur de croissance transformant bêta-1/génétique , Poumon/anatomopathologie , Poumon/cytologie , Transcriptome , Metformine/pharmacologie , Plasticité cellulaire/effets des médicaments et des substances chimiques , Phénotype
9.
Cells ; 13(13)2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-38994981

RÉSUMÉ

Aging is the main risk factor for chronic lung diseases (CLDs) including idiopathic pulmonary fibrosis (IPF) and chronic obstructive pulmonary disease (COPD). Accordingly, hallmarks of aging like cellular senescence are increased in these patients in different lung cell types including fibroblasts. However, little is known about the different triggers that induce a senescence phenotype in different disease backgrounds and its role in CLD pathogenesis. Therefore, we characterized senescence in primary human lung fibroblasts (phLF) from control, IPF, or COPD patients at baseline and after exposure to disease-relevant insults (H2O2, bleomycin, TGF-ß1) and studied their capacity to support progenitor cell potential in a lung organoid model. Bulk-RNA sequencing revealed that phLF from IPF and COPD activate different transcriptional programs but share a similar senescence phenotype at baseline. Moreover, H2O2 and bleomycin but not TGF-ß1 induced senescence in phLF from different disease origins. Exposure to different triggers resulted in distinct senescence programs in phLF characterized by different SASP profiles. Finally, co-culture with bleomycin- and H2O2-treated phLF reduced the progenitor cell potential of alveolar epithelial progenitor cells. In conclusion, phLF from COPD and IPF share a conserved senescence response that varies depending on the insult and impairs alveolar epithelial progenitor capacity ex vivo.


Sujet(s)
Bléomycine , Vieillissement de la cellule , Fibroblastes , Peroxyde d'hydrogène , Fibrose pulmonaire idiopathique , Poumon , Cellules souches , Humains , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Fibrose pulmonaire idiopathique/anatomopathologie , Fibrose pulmonaire idiopathique/métabolisme , Poumon/cytologie , Poumon/anatomopathologie , Bléomycine/pharmacologie , Cellules souches/métabolisme , Cellules souches/effets des médicaments et des substances chimiques , Cellules souches/cytologie , Peroxyde d'hydrogène/pharmacologie , Broncho-pneumopathie chronique obstructive/anatomopathologie , Broncho-pneumopathie chronique obstructive/métabolisme , Facteur de croissance transformant bêta-1/pharmacologie , Facteur de croissance transformant bêta-1/métabolisme , Pneumocytes/métabolisme , Pneumocytes/effets des médicaments et des substances chimiques , Cellules cultivées
10.
Cells ; 13(11)2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38891078

RÉSUMÉ

Pulmonary fibrosis is a chronic, progressive, irreversible lung disease characterized by fibrotic scarring in the lung parenchyma. This condition involves the excessive accumulation of extracellular matrix (ECM) due to the aberrant activation of myofibroblasts in the alveolar environment. Transforming growth factor beta (TGF-ß) signaling is a crucial driver of fibrogenesis because it promotes excessive ECM deposition, thereby leading to scar formation and lung damage. A primary target of TGF-ß signaling in fibrosis is Collagen Triple Helix Repeat Containing 1 (CTHRC1), a secreted glycoprotein that plays a pivotal role in ECM deposition and wound repair. TGF-ß transcriptionally regulates CTHRC1 in response to tissue injury and controls the wound healing response through functional activity. CTHRC1 may also play an essential role in re-establishing and maintaining tissue homeostasis after wound closure by modulating both the TGF-ß and canonical Wnt signaling pathways. This dual function suggests that CTHRC1 regulates tissue remodeling and homeostasis. However, deregulated CTHRC1 expression in pathogenic fibroblasts has recently emerged as a hallmark of fibrosis in multiple organs and tissues. This review highlights recent studies suggesting that CTHRC1 can serve as a diagnostic and prognostic biomarker for fibrosis in idiopathic pulmonary fibrosis, systemic sclerosis, and post-COVID-19 lung fibrosis. Notably, CTHRC1 expression is responsive to antifibrotic drugs that target the TGF-ß pathway, such as pirfenidone and bexotegrast, indicating its potential as a biomarker of treatment success. These findings suggest that CTHRC1 may present new opportunities for diagnosing and treating patients with lung fibrosis.


Sujet(s)
Protéines de la matrice extracellulaire , Fibroblastes , Fibrose pulmonaire , Humains , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Protéines de la matrice extracellulaire/métabolisme , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/métabolisme , Animaux , Facteur de croissance transformant bêta/métabolisme , Matrice extracellulaire/métabolisme , Fibrose pulmonaire idiopathique/anatomopathologie , Fibrose pulmonaire idiopathique/métabolisme
11.
Int J Nanomedicine ; 19: 6161-6176, 2024.
Article de Anglais | MEDLINE | ID: mdl-38911503

RÉSUMÉ

Idiopathic pulmonary fibrosis (IPF) poses a formidable clinical challenge, characterized by the thickening of alveolar septa and the onset of pulmonary fibrosis. The pronounced activation of oxidative stress emerges as a pivotal hallmark of inflammation. Traditional application of exogenous antioxidants proves inadequate in addressing oxidative stress, necessitating exploration into strategies to augment their antioxidant efficacy. Exosomes, nano-sized extracellular vesicles harboring a diverse array of bioactive factors, present as promising carriers with the potential to meet this challenge. Recent attention has been directed towards the clinical applications of exosomes in IPF, fueling the impetus for this comprehensive review. We have compiled fresh insights into the role of exosomes in modulating oxidative stress in IPF and delved into their potential as carriers for regulating endogenous reactive oxygen species generation. This review endeavors to bridge the divide between exosome research and IPF, traversing from bedside to bench. Through the synthesis of recent findings, we propose exosomes as a novel and promising strategy for improving the outcomes of IPF therapy.


Sujet(s)
Antioxydants , Exosomes , Fibrose pulmonaire idiopathique , Stress oxydatif , Espèces réactives de l'oxygène , Humains , Exosomes/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Fibrose pulmonaire idiopathique/traitement médicamenteux , Fibrose pulmonaire idiopathique/métabolisme , Espèces réactives de l'oxygène/métabolisme , Antioxydants/pharmacologie , Animaux
12.
Respir Res ; 25(1): 245, 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38886743

RÉSUMÉ

Idiopathic Pulmonary Fibrosis (IPF) represents a chronic and progressive pulmonary disorder distinguished by a notable mortality rate. Despite the elusive nature of the pathogenic mechanisms, several signaling pathways have been elucidated for their pivotal roles in the progression of this ailment. This manuscript aims to comprehensively review the existing literature on the signaling pathways linked to the pathogenesis of IPF, both within national and international contexts. The objective is to enhance the comprehension of the pathogenic mechanisms underlying IPF and offer a scholarly foundation for the advancement of more efficacious therapeutic strategies, thereby fostering research and clinical practices within this domain.


Sujet(s)
Fibrose pulmonaire idiopathique , Transduction du signal , Fibrose pulmonaire idiopathique/métabolisme , Humains , Transduction du signal/physiologie , Animaux
13.
Respir Res ; 25(1): 241, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38872139

RÉSUMÉ

Idiopathic pulmonary fibrosis (IPF) is a chronic disease of unknown etiology that lacks a specific treatment. In IPF, macrophages play a key regulatory role as a major component of the lung immune system, especially during inflammation and fibrosis. However, our understanding of the cellular heterogeneity and molecular characterization of macrophages in IPF, as well as their relevance in the clinical setting, is relatively limited. In this study, we analyzed in-depth single-cell transcriptome sequencing (scRNA-seq) data from lung tissues of IPF patients, identified macrophage subpopulations in IPF, and probed their molecular characteristics and biological functions. hdWGCNA identified co-expressed gene modules of a subpopulation of IPF-associated macrophages (IPF-MΦ), and probed the IPF-MΦ by a machine-learning approach. hdWGCNA identified a subpopulation of IPF-associated macrophage subpopulations and probed the IPF-MΦ signature gene (IRMG) for its prognostic value, and a prediction model was developed on this basis. In addition, IPF-MΦ was obtained after recluster analysis of macrophages in IPF lung tissues. Coexpressed gene modules of IPF-MΦ were identified by hdWGCNA. Then, a machine learning approach was utilized to reveal the characteristic genes of IPF-MΦ, and a prediction model was built on this basis. In addition, we discovered a type of macrophage unique to IPF lung tissue named ATP5-MΦ. Its characteristic gene encodes a subunit of the mitochondrial ATP synthase complex, which is closely related to oxidative phosphorylation and proton transmembrane transport, suggesting that ATP5-MΦ may have higher ATP synthesis capacity in IPF lung tissue. This study provides new insights into the pathogenesis of IPF and provides a basis for evaluating disease prognosis and predictive medicine in IPF patients.


Sujet(s)
Marqueurs biologiques , Fibrose pulmonaire idiopathique , Apprentissage machine , Macrophages , Analyse sur cellule unique , Fibrose pulmonaire idiopathique/métabolisme , Fibrose pulmonaire idiopathique/diagnostic , Fibrose pulmonaire idiopathique/génétique , Fibrose pulmonaire idiopathique/anatomopathologie , Humains , Analyse sur cellule unique/méthodes , Macrophages/métabolisme , Marqueurs biologiques/métabolisme , Mâle , Femelle , Poumon/métabolisme , Poumon/anatomopathologie
14.
J Cell Mol Med ; 28(11): e18414, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38872435

RÉSUMÉ

Idiopathic pulmonary fibrosis (IPF) is a debilitating interstitial lung disease characterized by progressive fibrosis and poor prognosis. Despite advancements in treatment, the pathophysiological mechanisms of IPF remain elusive. Herein, we conducted an integrated bioinformatics analysis combining clinical data and carried out experimental validations to unveil the intricate molecular mechanism of IPF. Leveraging three IPF datasets, we identified 817 upregulated and 560 downregulated differentially expressed genes (DEGs). Of these, 14 DEGs associated with copper metabolism were identified, shedding light on the potential involvement of disrupted copper metabolism in IPF progression. Immune infiltration analysis revealed dysregulated immune cell infiltration in IPF, with a notable correlation between copper metabolism-related genes and immune cells. Weighted gene co-expression network analysis (WGCNA) identified a central module correlated with IPF-associated genes, among which STEAP2 emerged as a key hub gene. Subsequent in vivo and in vitro studies confirmed the upregulation of STEAP2 in IPF model. Knockdown of STEAP2 using siRNA alleviated fibrosis in vitro, suggesting potential pathway related to copper metabolism in the pathophysiological progression of IPF. Our study established a novel link between immune cell infiltration and dysregulated copper metabolism. The revelation of intracellular copper overload and upregulated STEAP2 unravelled a potential therapeutic option. These findings offer valuable insights for future research and therapeutic interventions targeting STEAP2 and associated pathways in IPF.


Sujet(s)
Cuivre , Fibrose pulmonaire idiopathique , Fibrose pulmonaire idiopathique/métabolisme , Fibrose pulmonaire idiopathique/génétique , Fibrose pulmonaire idiopathique/anatomopathologie , Cuivre/métabolisme , Humains , Animaux , Biologie informatique/méthodes , Réseaux de régulation génique , Souris , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes
15.
Mol Pharm ; 21(7): 3684-3692, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38899595

RÉSUMÉ

Early detection of pulmonary fibrosis is a critical yet insufficiently met clinical necessity. This study evaluated the effectiveness of FAPI-LM3, a 68Ga-radiolabeled heterobivalent molecular probe that targets fibroblast activating protein (FAP) and somatostatin receptor 2 (SSTR2), in the early detection of pulmonary fibrosis, leveraging its potential for early disease identification. A bleomycin-induced early pulmonary fibrosis model was established in C57BL/6 mice for 7 days. FAP and SSTR2 expression levels were quantitatively assessed in human idiopathic pulmonary fibrosis lung tissue samples and bleomycin-treated mouse lung tissues by using western blotting, real-time quantitative PCR (RT-qPCR), and immunofluorescence techniques. The diagnostic performance of FAPI-LM3 was investigated by synthesizing monomeric radiotracers 68Ga-FAPI-46 and 68Ga-DOTA-LM3 alongside the heterobivalent probe 68Ga-FAPI-LM3. These imaging radiopharmaceuticals were used in small-animal PET to compare their uptake in fibrotic and normal lung tissues. Results indicated significant upregulation of FAP and SSTR2 at both RNA and protein levels in fibrotic lung tissues compared with that in normal controls. PET imaging demonstrated significantly enhanced uptake of the 68Ga-FAPI-LM3 probe in fibrotic lung tissues, with superior visual effects compared to monomeric tracers. At 60 min postinjection, early stage fibrotic tissues (day 7) demonstrated low-to-medium uptake of monomeric probes, including 68Ga-DOTA-LM3 (0.45 ± 0.04% ID/g) and 68Ga-FAPI-46 (0.78 ± 0.09% ID/g), whereas the uptake of the heterobivalent probe 68Ga-FAPI-LM3 (1.90 ± 0.10% ID/g) was significantly higher in fibrotic lesions than in normal lung tissue. Blockade experiments confirmed the specificity of 68Ga-FAPI-LM3 uptake, which was attributed to synergistic targeting of FAP and SSTR2. This study demonstrates the potential of 68Ga-FAPI-LM3 for early pulmonary fibrosis detection via molecular imaging, offering significant benefits over monomeric tracers 68Ga-FAPI-46 and 68Ga-DOTA-LM3. This strategy offers new possibilities for noninvasive and precise early detection of pulmonary fibrosis.


Sujet(s)
Radio-isotopes du gallium , Souris de lignée C57BL , Tomographie par émission de positons , Radiopharmaceutiques , Récepteur somatostatine , Animaux , Souris , Récepteur somatostatine/métabolisme , Humains , Tomographie par émission de positons/méthodes , Radiopharmaceutiques/pharmacocinétique , Radiopharmaceutiques/composition chimique , Fibrose pulmonaire/imagerie diagnostique , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/induit chimiquement , Poumon/imagerie diagnostique , Poumon/anatomopathologie , Poumon/métabolisme , Mâle , Bléomycine , Endopeptidases/métabolisme , Modèles animaux de maladie humaine , Femelle , Fibrose pulmonaire idiopathique/imagerie diagnostique , Fibrose pulmonaire idiopathique/métabolisme , Protéines membranaires/métabolisme , Serine endopeptidases/métabolisme , Quinoléines
16.
Int J Biol Macromol ; 273(Pt 1): 132896, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38851619

RÉSUMÉ

Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive pulmonary disease with an unclear pathogenesis and no available specific drug treatment. The principal etiological factors are lung inflammation caused by environmental factors, damage to alveolar epithelial cells, leading to epithelial-mesenchymal transition (EMT), and the abnormal proliferation of fibroblasts. Here, we have demonstrated that fibroblast growth factor 21 (FGF21) ameliorates IPF via the autophagy pathway. We administered FGF21 to bleomycin (BLM)-treated mice, which ameliorated their defects in lung function, reduced the accumulation of collagen, restored tissue structure, reduced the deposition of hydroxyproline, reduced the expression of collagen I and α-SMA and increased the expression of E-cadherin. The expression of LC3BII and the number of autophagosomes were significantly higher in the lungs. The expression of AKT and mTOR was significantly reduced by FGF21 treatment. We also determined the effects of FGF21 in A549 cells treated with TGF-ß, and found that FGF21 significantly inhibits activation of the AKT signaling pathway, thereby reducing TGF-ß-induced EMT and preventing the uncontrolled proliferation of fibroblasts. We conclude that FGF21 ameliorates IPF by inhibiting the PI3K-AKT-mTOR signaling pathway and activating autophagy, which provides a theoretical basis for FGF21 to be used for the treatment of IPF.


Sujet(s)
Autophagie , Bléomycine , Transition épithélio-mésenchymateuse , Facteurs de croissance fibroblastique , Fibrose pulmonaire idiopathique , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Transduction du signal , Sérine-thréonine kinases TOR , Sérine-thréonine kinases TOR/métabolisme , Autophagie/effets des médicaments et des substances chimiques , Animaux , Fibrose pulmonaire idiopathique/traitement médicamenteux , Fibrose pulmonaire idiopathique/métabolisme , Fibrose pulmonaire idiopathique/anatomopathologie , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Humains , Souris , Facteurs de croissance fibroblastique/métabolisme , Facteurs de croissance fibroblastique/pharmacologie , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Bléomycine/effets indésirables , Cellules A549 , Mâle , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Poumon/effets des médicaments et des substances chimiques , Poumon/métabolisme , Souris de lignée C57BL , Facteur de croissance transformant bêta/métabolisme
17.
Sci Adv ; 10(25): eadm9817, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38896611

RÉSUMÉ

Precision management of fibrotic lung diseases is challenging due to their diverse clinical trajectories and lack of reliable biomarkers for risk stratification and therapeutic monitoring. Here, we validated the accuracy of CMKLR1 as an imaging biomarker of the lung inflammation-fibrosis axis. By analyzing single-cell RNA sequencing datasets, we demonstrated CMKLR1 expression as a transient signature of monocyte-derived macrophages (MDMφ) enriched in patients with idiopathic pulmonary fibrosis (IPF). Consistently, we identified MDMφ as the major driver of the uptake of CMKLR1-targeting peptides in a murine model of bleomycin-induced lung fibrosis. Furthermore, CMKLR1-targeted positron emission tomography in the murine model enabled quantification and spatial mapping of inflamed lung regions infiltrated by CMKLR1-expressing macrophages and emerged as a robust predictor of subsequent lung fibrosis. Last, high CMKLR1 expression by bronchoalveolar lavage cells identified an inflammatory endotype of IPF with poor survival. Our investigation supports the potential of CMKLR1 as an imaging biomarker for endotyping and risk stratification of fibrotic lung diseases.


Sujet(s)
Fibrose pulmonaire idiopathique , Pneumopathie infectieuse , Animaux , Humains , Souris , Fibrose pulmonaire idiopathique/imagerie diagnostique , Fibrose pulmonaire idiopathique/anatomopathologie , Fibrose pulmonaire idiopathique/métabolisme , Fibrose pulmonaire idiopathique/induit chimiquement , Pneumopathie infectieuse/métabolisme , Pneumopathie infectieuse/imagerie diagnostique , Pneumopathie infectieuse/anatomopathologie , Macrophages/métabolisme , Macrophages/anatomopathologie , Marqueurs biologiques , Modèles animaux de maladie humaine , Tomographie par émission de positons/méthodes , Fibrose pulmonaire/imagerie diagnostique , Fibrose pulmonaire/anatomopathologie , Fibrose pulmonaire/métabolisme , Fibrose pulmonaire/induit chimiquement , Bléomycine , Poumon/anatomopathologie , Poumon/imagerie diagnostique , Poumon/métabolisme , Mâle , Femelle , Souris de lignée C57BL
18.
Sci Rep ; 14(1): 14376, 2024 06 22.
Article de Anglais | MEDLINE | ID: mdl-38909094

RÉSUMÉ

Idiopathic pulmonary fibrosis (IPF) is believed to be associated with a notable disruption of cellular energy metabolism. By detecting the changes of energy metabolites in the serum of patients with pulmonary fibrosis, we aimed to investigate the diagnostic and prognostic value of energy metabolites in IPF, and further elucidated the mechanism of their involvement in pulmonary fibrosis. Through metabolomics research, it was discovered that the TCA cycle intermediates changed dramatically in IPF patients. In another validation cohort of 55 patients with IPF compared to 19 healthy controls, it was found that succinate, an intermediate product of TCA cycle, has diagnostic and prognostic value in IPF. The cut-off levels of serum succinate were 98.36 µM for distinguishing IPF from healthy controls (sensitivity, 83.64%; specificity, 63.16%; likelihood ratio, 2.27, respectively). Moreover, a high serum succinate level was independently associated with higher rates of disease progression (OR 13.087, 95%CI (2.819-60.761)) and mortality (HR 3.418, 95% CI (1.308-8.927)). In addition, accumulation of succinate and increased expression of the succinate receptor GPR91 were found in both IPF patients and BLM mouse models of pulmonary fibrosis. Reducing succinate accumulation in BLM mice alleviated pulmonary fibrosis and 21d mortality, while exogenous administration of succinate can aggravate pulmonary fibrosis in BLM mice. Furthermore, GPR91 deficiency protected against lung fibrosis caused by BLM. In vitro, succinate promoted the activation of lung fibroblasts by activating ERK pathway through GPR91. In summary, succinate is a promising biomarker for diagnosis and prognosis of IPF. The accumulation of succinate may promote fibroblast activation through GPR91 and pulmonary fibrosis.


Sujet(s)
Fibrose pulmonaire idiopathique , Récepteurs couplés aux protéines G , Acide succinique , Acide succinique/métabolisme , Acide succinique/sang , Récepteurs couplés aux protéines G/métabolisme , Récepteurs couplés aux protéines G/génétique , Humains , Fibrose pulmonaire idiopathique/métabolisme , Fibrose pulmonaire idiopathique/anatomopathologie , Fibrose pulmonaire idiopathique/mortalité , Animaux , Mâle , Souris , Femelle , Adulte d'âge moyen , Pronostic , Sujet âgé , Modèles animaux de maladie humaine , Marqueurs biologiques/sang , Fibroblastes/métabolisme , Cycle citrique
19.
J Cell Mol Med ; 28(12): e18499, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38887981

RÉSUMÉ

Idiopathic pulmonary fibrosis (IPF) is a common, chronic, and progressive lung disease that severely impacts human health and survival. However, the intricate molecular underpinnings of IPF remains elusive. This study aims to delve into the nuanced molecular interplay of cellular interactions in IPF, thereby laying the groundwork for innovative therapeutic approaches in the clinical field of IPF. Sophisticated bioinformatics methods were employed to identify crucial biomarkers essential for the progression of IPF. The GSE122960 single-cell dataset was obtained from the Gene Expression Omnibus (GEO) compendium, and intercellular communication potentialities were scrutinized via CellChat. The random survival forest paradigm was established using the GSE70866 dataset. Quintessential genes were selected through Kaplan-Meier (KM) curves, while immune infiltration examinations, functional enrichment critiques and nomogram paradigms were inaugurated. Analysis of intercellular communication revealed an intimate potential connections between macrophages and various cell types, pinpointing five cardinal genes influencing the trajectory and prognosis of IPF. The nomogram paradigm, sculpted from these seminal genes, exhibits superior predictive prowess. Our research meticulously identified five critical genes, confirming their intimate association with the prognosis, immune infiltration and transcriptional governance of IPF. Interestingly, we discerned these genes' engagement with the EPITHELIAL_MESENCHYMAL_TRANSITION signalling pathway, which may enhance our understanding of the molecular complexity of IPF.


Sujet(s)
Communication cellulaire , Fibrose pulmonaire idiopathique , Analyse sur cellule unique , Transcriptome , Fibrose pulmonaire idiopathique/génétique , Fibrose pulmonaire idiopathique/anatomopathologie , Fibrose pulmonaire idiopathique/métabolisme , Humains , Communication cellulaire/génétique , Analyse sur cellule unique/méthodes , Transcriptome/génétique , Analyse de profil d'expression de gènes , Biologie informatique/méthodes , Pronostic , Marqueurs biologiques/métabolisme , Régulation de l'expression des gènes , Réseaux de régulation génique , Nomogrammes
20.
Biomed Pharmacother ; 176: 116896, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38876049

RÉSUMÉ

Idiopathic pulmonary fibrosis (IPF) is a severe disability due to progressive lung dysfunction. IPF has long been viewed as a non-immune form of pulmonary fibrosis, but nowadays it is accepted that a chronic inflammatory response can exacerbate fibrotic patterns. IL-1-like cytokines and ATP are highly detected in the lung and broncho-alveolar lavage fluid of IPF patients. Because ATP binds the purinergic receptor P2RX7 involved in the release of IL-1-like cytokines, we aimed to understand the role of P2RX7 in IPF. PBMCs from IPF patients were treated with nintedanib or pirfenidone in the presence of ATP. Under these conditions, PBMCs still released IL-1-like cytokines and the pro-fibrotic TGFß. Bulk and scRNAseq demonstrated that lung tissues of IPF patients had higher levels of P2RX7, especially on macrophages, which were correlated to T cell activity and inflammatory response with a TGFBI and IL-10 signature. A subcluster of macrophages in IPF lung tissues had 2055 genes that were not in common with the other subclusters, and that were involved in metabolic and PDGF, FGF and VEGF associated pathways. These data confirmed what observed on circulating cells that, although treated with anti-fibrotic agents, nintedanib or pirfenidone, they were still able to release IL-1 cytokines and the fibrogenic TGFß. In conclusion, these data imply that because nintedanib and pirfenidone do not block ATP-induced IL-1-like cytokines and TGFß induced during P2RX7 activation, it is plausible to consider P2RX7 on circulating cells and/or tissue biopsies as potential pharmacological tool for IPF patients.


Sujet(s)
Adénosine triphosphate , Fibrose pulmonaire idiopathique , Indoles , Pyridones , Récepteurs purinergiques P2X7 , Humains , Fibrose pulmonaire idiopathique/traitement médicamenteux , Fibrose pulmonaire idiopathique/métabolisme , Fibrose pulmonaire idiopathique/anatomopathologie , Pyridones/pharmacologie , Pyridones/usage thérapeutique , Indoles/pharmacologie , Indoles/usage thérapeutique , Adénosine triphosphate/métabolisme , Récepteurs purinergiques P2X7/métabolisme , Mâle , Poumon/effets des médicaments et des substances chimiques , Poumon/anatomopathologie , Poumon/métabolisme , Femelle , Cytokines/métabolisme , Sujet âgé , Agranulocytes/effets des médicaments et des substances chimiques , Agranulocytes/métabolisme , Adulte d'âge moyen , Facteur de croissance transformant bêta/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE