Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
2.
Front Cell Dev Biol ; 9: 639162, 2021.
Article in English | MEDLINE | ID: mdl-34124033

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) represents the most aggressive form of pulmonary fibrosis (PF) and is a highly debilitating disorder with a poorly understood etiology. The lung epithelium seems to play a critical role in the initiation and progression of the disease. A repeated injury of lung epithelial cells prompts type II alveolar cells to secrete pro-fibrotic cytokines, which induces differentiation of resident mesenchymal stem cells into myofibroblasts, thus promoting aberrant deposition of extracellular matrix (ECM) and formation of fibrotic lesions. Reactivation of developmental pathways such as the Wnt-ß-catenin signaling cascade in lung epithelial cells plays a critical role in this process, but the underlying mechanisms are still enigmatic. Here, we demonstrate that the membrane-associated protein NUMB is required for pathological activation of ß-catenin signaling in lung epithelial cells following bleomycin-induced injury. Importantly, depletion of Numb and Numblike reduces accumulation of fibrotic lesions, preserves lung functions, and increases survival rates after bleomycin treatment of mice. Mechanistically, we demonstrate that NUMB interacts with casein kinase 2 (CK2) and relies on CK2 to activate ß-catenin signaling. We propose that pharmacological inhibition of NUMB signaling may represent an effective strategy for the development of novel therapeutic approaches against PF.

3.
EMBO Mol Med ; 10(2): 276-293, 2018 02.
Article in English | MEDLINE | ID: mdl-29217661

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal parenchymal lung disease with limited therapeutic options, with fibroblast-to-myofibroblast transdifferentiation and hyperproliferation playing a major role. Investigating ex vivo-cultured (myo)fibroblasts from human IPF lungs as well as fibroblasts isolated from bleomycin-challenged mice, Forkhead box O3 (FoxO3) transcription factor was found to be less expressed, hyperphosphorylated, and nuclear-excluded relative to non-diseased controls. Downregulation and/or hyperphosphorylation of FoxO3 was reproduced by exposure of normal human lung fibroblasts to various pro-fibrotic growth factors and cytokines (FCS, PDGF, IGF1, TGF-ß1). Moreover, selective knockdown of FoxO3 in the normal human lung fibroblasts reproduced the transdifferentiation and hyperproliferation phenotype. Importantly, mice with global- (Foxo3-/-) or fibroblast-specific (Foxo3f.b-/-) FoxO3 knockout displayed enhanced susceptibility to bleomycin challenge, with augmented fibrosis, loss of lung function, and increased mortality. Activation of FoxO3 with UCN-01, a staurosporine derivative currently investigated in clinical cancer trials, reverted the IPF myofibroblast phenotype in vitro and blocked the bleomycin-induced lung fibrosis in vivo These studies implicate FoxO3 as a critical integrator of pro-fibrotic signaling in lung fibrosis and pharmacological reconstitution of FoxO3 as a novel treatment strategy.


Subject(s)
Fibroblasts , Forkhead Box Protein O3/genetics , Idiopathic Pulmonary Fibrosis/metabolism , Myofibroblasts , Animals , Cell Proliferation , Cell Transdifferentiation , Cells, Cultured , Cytokines/pharmacology , Down-Regulation , Fibroblasts/metabolism , Fibroblasts/pathology , Forkhead Box Protein O3/metabolism , Gene Knockout Techniques , Humans , Idiopathic Pulmonary Fibrosis/pathology , Idiopathic Pulmonary Fibrosis/therapy , Models, Animal , Myofibroblasts/metabolism , Myofibroblasts/pathology , Phosphorylation , Staurosporine/chemistry , Staurosporine/pharmacology
4.
Am J Respir Cell Mol Biol ; 57(5): 589-602, 2017 11.
Article in English | MEDLINE | ID: mdl-28678521

ABSTRACT

Acute respiratory distress syndrome constitutes a significant disease burden with regard to both morbidity and mortality. Current therapies are mostly supportive and do not address the underlying pathophysiologic mechanisms. Removal of protein-rich alveolar edema-a clinical hallmark of acute respiratory distress syndrome-is critical for survival. Here, we describe a transforming growth factor (TGF)-ß-triggered mechanism, in which megalin, the primary mediator of alveolar protein transport, is negatively regulated by glycogen synthase kinase (GSK) 3ß, with protein phosphatase 1 and nuclear inhibitor of protein phosphatase 1 being involved in the signaling cascade. Inhibition of GSK3ß rescued transepithelial protein clearance in primary alveolar epithelial cells after TGF-ß treatment. Moreover, in a bleomycin-based model of acute lung injury, megalin+/- animals (the megalin-/- variant is lethal due to postnatal respiratory failure) showed a marked increase in intra-alveolar protein and more severe lung injury compared with wild-type littermates. In contrast, wild-type mice treated with the clinically relevant GSK3ß inhibitors, tideglusib and valproate, exhibited significantly decreased alveolar protein concentrations, which was associated with improved lung function and histopathology. Together, we discovered that the TGF-ß-GSK3ß-megalin axis is centrally involved in disturbances of alveolar protein clearance in acute lung injury and provide preclinical evidence for therapeutic efficacy of GSK3ß inhibition.


Subject(s)
Acute Lung Injury/metabolism , Acute Lung Injury/therapy , Low Density Lipoprotein Receptor-Related Protein-2/metabolism , Pulmonary Alveoli/metabolism , Acute Lung Injury/genetics , Animals , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta/metabolism , Low Density Lipoprotein Receptor-Related Protein-2/deficiency , Lung/metabolism , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Edema/metabolism , Pulmonary Edema/therapy , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/therapy , Transforming Growth Factor beta/metabolism
5.
Am J Respir Crit Care Med ; 191(4): 437-47, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25536148

ABSTRACT

RATIONALE: Recent studies indicate that tumor-associated macrophages (MΦ) with an M2 phenotype can influence cancer progression and metastasis, but the regulatory pathways remain poorly characterized. OBJECTIVES: This study investigated the role of tumor-associated MΦ in lung cancer. METHODS: Coculturing of MΦ with mouse Lewis lung carcinoma (LLC1) and 10 different human lung cancer cell lines (adenocarcinoma, squamous cell carcinoma, and large cell carcinoma) caused up-regulation of CCR2/CCL2 and CX3CR1/CX3CL1 in both the cancer cells and the MΦ. MEASUREMENTS AND MAIN RESULTS: In the MΦ-tumor cell system, IL-10 drove CCR2 and CX3CR1 up-regulation, whereas CCL1, granulocyte colony-stimulating factor, and MIP1α were required for the up-regulation of CCL2 and CX3CL1. Downstream phenotypic effects included enhanced LLC1 proliferation and migration and MΦ M2 polarization. In vivo, MΦ depletion (clodronate, MΦ Fas-induced apoptosis mice) and genetic ablation of CCR2 and CX3CR1 all inhibited LLC1 tumor growth and metastasis, shifted tumor-associated MΦ toward M1 polarization, suppressed tumor vessel growth, and enhanced survival (metastasis model). Furthermore, mice treated with CCR2 antagonist mimicked genetic ablation of CCR2, showing reduced tumor growth and metastasis. In human lung cancer samples, tumor MΦ infiltration and CCR2 expression correlated with tumor stage and metastasis. CONCLUSIONS: Tumor-associated MΦ play a central role in lung cancer growth and metastasis, with bidirectional cross-talk between MΦ and cancer cells via CCR2 and CX3CR1 signaling as a central underlying mechanism. These findings suggest that the therapeutic strategy of blocking CCR2 and CX3CR1 may prove beneficial for halting lung cancer progression.


Subject(s)
Adenocarcinoma/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Large Cell/metabolism , Carcinoma, Squamous Cell/metabolism , Lung Neoplasms/metabolism , Macrophages/metabolism , Adenocarcinoma/pathology , Animals , CX3C Chemokine Receptor 1 , Carcinoma, Large Cell/pathology , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Chemokine CCL2/metabolism , Chemokine CX3CL1/metabolism , Humans , Lung Neoplasms/pathology , Mice , Neoplasm Metastasis , Neoplasm Staging , Receptor Cross-Talk , Receptors, CCR2/metabolism , Receptors, Chemokine/metabolism , Up-Regulation
6.
Nat Med ; 20(11): 1289-300, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25344740

ABSTRACT

Pulmonary hypertension (PH) is characterized by increased proliferation and apoptosis resistance of pulmonary artery smooth muscle cells (PASMCs). Forkhead box O (FoxO) transcription factors are key regulators of cellular proliferation. Here we show that in pulmonary vessels and PASMCs of human and experimental PH lungs, FoxO1 expression is downregulated and FoxO1 is inactivated via phosphorylation and nuclear exclusion. These findings could be reproduced using ex vivo exposure of PASMCs to growth factors and inflammatory cytokines. Pharmacological inhibition and genetic ablation of FoxO1 in smooth muscle cells reproduced PH features in vitro and in vivo. Either pharmacological reconstitution of FoxO1 activity using intravenous or inhaled paclitaxel, or reconstitution of the transcriptional activity of FoxO1 by gene therapy, restored the physiologically quiescent PASMC phenotype in vitro, linked to changes in cell cycle control and bone morphogenic protein receptor type 2 (BMPR2) signaling, and reversed vascular remodeling and right-heart hypertrophy in vivo. Thus, PASMC FoxO1 is a critical integrator of multiple signaling pathways driving PH, and reconstitution of FoxO1 activity offers a potential therapeutic option for PH.


Subject(s)
Forkhead Transcription Factors/metabolism , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Signal Transduction , Adult , Animals , Apoptosis/drug effects , Apoptosis/genetics , Bone Morphogenetic Proteins/metabolism , Bromodeoxyuridine/metabolism , Cell Proliferation/drug effects , Cytokines/metabolism , Female , Forkhead Transcription Factors/genetics , Gene Deletion , Gene Expression Regulation/drug effects , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/physiopathology , Intercellular Signaling Peptides and Proteins/metabolism , Lung/blood supply , Lung/pathology , Lung/physiopathology , Male , Mice , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Paclitaxel/pharmacology , Pulmonary Artery/pathology , Quinolones/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Signal Transduction/drug effects , Signal Transduction/genetics , Small Molecule Libraries/pharmacology , Vascular Remodeling/drug effects , Vascular Remodeling/genetics
7.
BMC Pulm Med ; 10: 26, 2010 May 05.
Article in English | MEDLINE | ID: mdl-20444277

ABSTRACT

BACKGROUND: Pulmonary fibrosis (PF) is a group of devastating and largely irreversible diseases. Phosphodiesterase (PDE) 4 is involved in the processes of remodeling and inflammation, which play key role in tissue fibrosis. The aim of the study was, therefore, to investigate the effect of PDE4 inhibition in experimental model of PF. METHODS: PF was induced in C57BL/6N mice by instillation of bleomycin. Pharmacological inhibition of PDE4 was achieved by using cilomilast, a selective PDE4 inhibitor. Changes in either lung inflammation or remodeling were evaluated at different stages of experimental PF. Lung inflammation was assessed by bronchoalveolar lavage fluid (BALF) differential cell count and reverse transcription quantitative polymerase chain reaction (RT-qPCR) for inflammatory cytokines. Changes in tissue remodeling were evaluated by pulmonary compliance measurement, quantified pathological examination, measurement of collagen deposition and RT-qPCR for late remodeling markers. Survival in all groups was analyzed as well. RESULTS: PDE4 inhibition significantly reduced the total number of alveolar inflammatory cells in BALF of mice with bleomycin-induced PF at early fibrosis stage (days 4 and 7). Number of macrophages and lymphocytes, but not neutrophils, was significantly reduced as well. Treatment decreased lung tumor necrosis factor (TNF)-alpha mRNA level and increased mRNA level of interleukin (IL)-6 but did not influence IL-1 beta. At later stage (days 14 and 24) cilomilast improved lung function, which was shown by increase in lung compliance. It also lowered fibrosis degree, as was shown by quantified pathological examination of Hematoxilin-Eosin stained lung sections. Cilomilast had no significant effect on the expression of late remodeling markers such as transforming growth factor (TGF)-beta1 and collagen type Ia1 (COL(I)alpha1). However, it tended to restore the level of lung collagen, assessed by SIRCOL assay and Masson's trichrome staining, and to improve the overall survival. CONCLUSIONS: Selective PDE4 inhibition suppresses early inflammatory stage and attenuates the late stage of experimental pulmonary fibrosis.


Subject(s)
Bleomycin/adverse effects , Phosphodiesterase 4 Inhibitors , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/prevention & control , Animals , Carboxylic Acids/pharmacology , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Cyclohexanecarboxylic Acids , Disease Models, Animal , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Lung/drug effects , Lung/pathology , Lung/physiopathology , Male , Mice , Mice, Inbred C57BL , Nitriles/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Pneumonia/chemically induced , Pneumonia/metabolism , Pneumonia/prevention & control , Pulmonary Fibrosis/metabolism , Transforming Growth Factor beta1/metabolism , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...