Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Circ Res ; 134(11): e133-e149, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38639105

ABSTRACT

BACKGROUND: The precise origin of newly formed ACTA2+ (alpha smooth muscle actin-positive) cells appearing in nonmuscularized vessels in the context of pulmonary hypertension is still debatable although it is believed that they predominantly derive from preexisting vascular smooth muscle cells (VSMCs). METHODS: Gli1Cre-ERT2; tdTomatoflox mice were used to lineage trace GLI1+ (glioma-associated oncogene homolog 1-positive) cells in the context of pulmonary hypertension using 2 independent models of vascular remodeling and reverse remodeling: hypoxia and cigarette smoke exposure. Hemodynamic measurements, right ventricular hypertrophy assessment, flow cytometry, and histological analysis of thick lung sections followed by state-of-the-art 3-dimensional reconstruction and quantification using Imaris software were used to investigate the contribution of GLI1+ cells to neomuscularization of the pulmonary vasculature. RESULTS: The data show that GLI1+ cells are abundant around distal, nonmuscularized vessels during steady state, and this lineage contributes to around 50% of newly formed ACTA2+ cells around these normally nonmuscularized vessels. During reverse remodeling, cells derived from the GLI1+ lineage are largely cleared in parallel to the reversal of muscularization. Partial ablation of GLI1+ cells greatly prevented vascular remodeling in response to hypoxia and attenuated the increase in right ventricular systolic pressure and right heart hypertrophy. Single-cell RNA sequencing on sorted lineage-labeled GLI1+ cells revealed an Acta2high fraction of cells with pathways in cancer and MAPK (mitogen-activated protein kinase) signaling as potential players in reprogramming these cells during vascular remodeling. Analysis of human lung-derived material suggests that GLI1 signaling is overactivated in both group 1 and group 3 pulmonary hypertension and can promote proliferation and myogenic differentiation. CONCLUSIONS: Our data highlight GLI1+ cells as an alternative cellular source of VSMCs in pulmonary hypertension and suggest that these cells and the associated signaling pathways represent an important therapeutic target for further studies.


Subject(s)
Hypertension, Pulmonary , Vascular Remodeling , Zinc Finger Protein GLI1 , Animals , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein GLI1/genetics , Mice , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/pathology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Mice, Inbred C57BL , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Mice, Transgenic , Male , Humans , Hypoxia/metabolism , Hypoxia/physiopathology
2.
Stem Cells ; 40(6): 605-617, 2022 06 22.
Article in English | MEDLINE | ID: mdl-35437594

ABSTRACT

Bronchopulmonary dysplasia (BPD) is a neonatal lung disease developing in premature babies characterized by arrested alveologenesis and associated with decreased Fibroblast growth factor 10 (FGF10) expression. One-week hyperoxia (HYX) exposure of newborn mice leads to a permanent arrest in alveologenesis. To test the role of Fgf10 signaling to promote de novo alveologenesis following hyperoxia, we used transgenic mice allowing inducible expression of Fgf10 and recombinant FGF10 (rFGF10) protein delivered intraperitoneally. We carried out morphometry analysis, and IF on day 45. Alveolospheres assays were performed co-culturing AT2s from normoxia (NOX) with FACS-isolated Sca1Pos resident mesenchymal cells (rMC) from animals exposed to NOX, HYX-PBS, or HYX-FGF10. scRNAseq between rMC-Sca1Pos isolated from NOX and HYX-PBS was also carried out. Transgenic overexpression of Fgf10 and rFGF10 administration rescued the alveologenesis defects following HYX. Alveolosphere assays indicate that the activity of rMC-Sca1Pos is negatively impacted by HYX and partially rescued by rFGF10 treatment. Analysis by IF demonstrates a significant impact of rFGF10 on the activity of resident mesenchymal cells. scRNAseq results identified clusters expressing Fgf10, Fgf7, Pdgfra, and Axin2, which could represent the rMC niche cells for the AT2 stem cells. In conclusion, we demonstrate that rFGF10 administration is able to induce de novo alveologenesis in a BPD mouse model and identified subpopulations of rMC-Sca1Pos niche cells potentially representing its cellular target.


Subject(s)
Bronchopulmonary Dysplasia , Hyperoxia , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/genetics , Bronchopulmonary Dysplasia/metabolism , Fibroblast Growth Factor 10/genetics , Fibroblast Growth Factor 10/metabolism , Humans , Hyperoxia/metabolism , Infant, Newborn , Lung/metabolism , Mice , Mice, Transgenic
3.
Infection ; 51(5): 1357-1367, 2023 Oct.
Article in English | MEDLINE | ID: mdl-36787015

ABSTRACT

PURPOSE: SARS-CoV-2 infections cause COVID-19 and have a wide spectrum of morbidity. Severe disease courses among children are rare. To date, data on the variability of morbidity in relation to variant of concern (VOC) in children has been sparse and inconclusive. We compare the clinical severity of SARS-CoV-2 infection among children and adolescents in Germany during the Wildtype and Alpha combined, Delta and Omicron phases of the COVID-19 pandemic. METHODS: Comparing risk of COVID-19-related hospitalization, intensive care unit (ICU) admission and death due to COVID-19 in children and adolescents, we used: (1) a multi-center seroprevalence study (SARS-CoV-2-KIDS study); (2) a nationwide registry of pediatric patients hospitalized with SARS-CoV-2 infections; and (3) compulsory national reporting for RT-PCR-confirmed SARS-CoV-2 infections in Germany. RESULTS: During the Delta predominant phase, risk of COVID-19-related hospitalization among all SARS-CoV-2 seropositive children was 3.35, ICU admission 1.19 and fatality 0.09 per 10,000; hence about halved for hospitalization and ICU admission and unchanged for deaths as compared to the Wildtype- and Alpha-dominant period. The relative risk for COVID-19-related hospitalization and ICU admission compared to the alpha period decreased during Delta [0.60 (95% CI 0.54; 0.67) and 0.51 (95% CI 0.42; 0.61)] and Omicron [0.27 (95% CI 0.24; 0.30) and 0.06 (95% CI 0.05; 0.08)] period except for the < 5-year-olds. The rate of case fatalities decreased slightly during Delta, and substantially during Omicron phase. CONCLUSION: Morbidity caused by SARS-CoV-2 infections among children and adolescents in Germany decreased over the course of the COVID-19 pandemic, as different VOCs) emerged.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Adolescent , Child , Child, Preschool , COVID-19/epidemiology , Risk , Pandemics , Seroepidemiologic Studies , Hospitalization , Germany/epidemiology , Intensive Care Units
4.
Cell Mol Life Sci ; 79(12): 609, 2022 Nov 29.
Article in English | MEDLINE | ID: mdl-36445537

ABSTRACT

The specification, characterization, and fate of alveolar type 1 and type 2 (AT1 and AT2) progenitors during embryonic lung development are poorly defined. Current models of distal epithelial lineage formation fail to capture the heterogeneity and dynamic contribution of progenitor pools present during early development. Furthermore, few studies explore the pathways involved in alveolar progenitor specification and fate. In this paper, we build upon our previously published work on the regulation of airway epithelial progenitors by fibroblast growth factor receptor 2b (FGFR2b) signalling during early (E12.5) and mid (E14.5) pseudoglandular stage lung development. Our results suggest that a significant proportion of AT2 and AT1 progenitors are lineage-flexible during late pseudoglandular stage development, and that lineage commitment is regulated in part by FGFR2b signalling. We have characterized a set of direct FGFR2b targets at E16.5 which are likely involved in alveolar lineage formation. These signature genes converge on a subpopulation of AT2 cells later in development and are downregulated in AT2 cells transitioning to the AT1 lineage during repair after injury in adults. Our findings highlight the extensive heterogeneity of pneumocytes by elucidating the role of FGFR2b signalling in these cells during early airway epithelial lineage formation, as well as during repair after injury.


Subject(s)
Alveolar Epithelial Cells , Lung , Receptor, Fibroblast Growth Factor, Type 2 , Stem Cells , Animals , Mice , Embryonic Development , Receptor, Fibroblast Growth Factor, Type 2/genetics , Signal Transduction , Lung/embryology , Cell Lineage
5.
Int J Mol Sci ; 24(13)2023 Jul 07.
Article in English | MEDLINE | ID: mdl-37446407

ABSTRACT

Bronchopulmonary dysplasia (BPD) is a chronic lung disease commonly seen in preterm infants, and is triggered by infection, mechanical ventilation, and oxygen toxicity. Among other problems, lifelong limitations in lung function and impaired psychomotor development may result. Despite major advances in understanding the disease pathologies, successful interventions are still limited to only a few drug therapies with a restricted therapeutic benefit, and which sometimes have significant side effects. As a more promising therapeutic option, mesenchymal stem cells (MSCs) have been in focus for several years due to their anti-inflammatory effects and their secretion of growth and development promoting factors. Preclinical studies provide evidence in that MSCs have the potential to contribute to the repair of lung injuries. This review provides an overview of MSCs, and other stem/progenitor cells present in the lung, their identifying characteristics, and their differentiation potential, including cytokine/growth factor involvement. Furthermore, animal studies and clinical trials using stem cells or their secretome are reviewed. To bring MSC-based therapeutic options further to clinical use, standardized protocols are needed, and upcoming side effects must be critically evaluated. To fill these gaps of knowledge, the MSCs' behavior and the effects of their secretome have to be examined in more (pre-) clinical studies, from which only few have been designed to date.


Subject(s)
Bronchopulmonary Dysplasia , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Infant, Newborn , Animals , Humans , Bronchopulmonary Dysplasia/therapy , Bronchopulmonary Dysplasia/pathology , Infant, Premature , Lung/pathology , Stem Cells/pathology , Mesenchymal Stem Cells/pathology , Mesenchymal Stem Cell Transplantation/methods
6.
Int J Mol Sci ; 23(13)2022 Jun 30.
Article in English | MEDLINE | ID: mdl-35806292

ABSTRACT

Pulmonary manifestation (PM) of inflammatory bowel disease (IBD) in children is a rare condition. The exact pathogenesis is still unclear, but several explanatory concepts were postulated and several case reports in children were published. We performed a systematic Medline search between April 1976 and April 2022. Different pathophysiological concepts were identified, including the shared embryological origin, "miss-homing" of intestinal based neutrophils and T lymphocytes, inflammatory triggering via certain molecules (tripeptide proline-glycine-proline, interleukin 25), genetic factors and alterations in the microbiome. Most pediatric IBD patients with PM are asymptomatic, but can show alterations in pulmonary function tests and breathing tests. In children, the pulmonary parenchyma is more affected than the airways, leading histologically mainly to organizing pneumonia. Medication-associated lung injury has to be considered in pulmonary symptomatic pediatric IBD patients treated with certain agents (i.e., mesalamine, sulfasalazine or infliximab). Furthermore, the risk of pulmonary embolism is generally increased in pediatric IBD patients. The initial treatment of PM is based on corticosteroids, either inhaled for the larger airways or systemic for smaller airways and parenchymal disease. In summary, this review article summarizes the current knowledge about PM in pediatric IBD patients, focusing on pathophysiological and clinical aspects.


Subject(s)
Inflammatory Bowel Diseases , Child , Humans , Inflammatory Bowel Diseases/complications , Inflammatory Bowel Diseases/diagnosis , Infliximab/therapeutic use , Lung , Mesalamine , Proline
7.
Int J Mol Sci ; 23(17)2022 Sep 04.
Article in English | MEDLINE | ID: mdl-36077511

ABSTRACT

Insulin receptor substrates (IRSs) are proteins that are involved in signaling through the insulin receptor (IR) and insulin-like growth factor (IGFR). They can also interact with other receptors including growth factor receptors. Thus, they represent a critical node for the transduction and regulation of multiple signaling pathways in response to extracellular stimuli. In addition, IRSs play a central role in processes such as inflammation, growth, metabolism, and proliferation. Previous studies have highlighted the role of IRS proteins in lung diseases, in particular asthma. Further, the members of the IRS family are the common proteins of the insulin growth factor signaling cascade involved in lung development and disrupted in bronchopulmonary dysplasia (BPD). However, there is no study focusing on the relationship between IRS proteins and BPD yet. Unfortunately, there is still a significant gap in knowledge in this field. Thus, in this review, we aimed to summarize the current knowledge with the major goal of exploring the possible roles of IRS in BPD and asthma to foster new perspectives for further investigations.


Subject(s)
Asthma , Bronchopulmonary Dysplasia , Humans , Infant, Newborn , Insulin/metabolism , Insulin Receptor Substrate Proteins/metabolism , Phosphoproteins/metabolism , Receptor, Insulin/metabolism
8.
Int J Mol Sci ; 23(12)2022 Jun 18.
Article in English | MEDLINE | ID: mdl-35743234

ABSTRACT

The intestinal microbiota is known to influence local immune homeostasis in the gut and to shape the developing immune system towards elimination of pathogens and tolerance towards self-antigens. Even though the lung was considered sterile for a long time, recent evidence using next-generation sequencing techniques confirmed that the lower airways possess their own local microbiota. Since then, there has been growing evidence that the local respiratory and intestinal microbiota play a role in acute and chronic pediatric lung diseases. The concept of the so-called gut-lung axis describing the mutual influence of local microbiota on distal immune mechanisms was established. The mechanisms by which the intestinal microbiota modulates the systemic immune response include the production of short-chain fatty acids (SCFA) and signaling through pattern recognition receptors (PRR) and segmented filamentous bacteria. Those factors influence the secretion of pro- and anti-inflammatory cytokines by immune cells and further modulate differentiation and recruitment of T cells to the lung. This article does not only aim at reviewing recent mechanistic evidence from animal studies regarding the gut-lung axis, but also summarizes current knowledge from observational studies and human trials investigating the role of the respiratory and intestinal microbiota and their modulation by pre-, pro-, and synbiotics in pediatric lung diseases.


Subject(s)
Gastrointestinal Microbiome , Lung Diseases , Microbiota , Animals , Child , Fatty Acids, Volatile , Gastrointestinal Microbiome/physiology , Humans , Lung
9.
Hum Mol Genet ; 28(9): 1429-1444, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30566624

ABSTRACT

Bronchopulmonary dysplasia (BPD), characterized by alveoli simplification and dysmorphic pulmonary microvasculature, is a chronic lung disease affecting prematurely born infants. Pulmonary hypertension (PH) is an important BPD feature associated with morbidity and mortality. In human BPD, inflammation leads to decreased fibroblast growth factor 10 (FGF10) expression but the impact on the vasculature is so far unknown. We used lungs from Fgf10+/- versus Fgf10+/+ pups to investigate the effect of Fgf10 deficiency on vascular development in normoxia (NOX) and hyperoxia (HOX, BPD mouse model). To assess the role of fibroblast growth factor receptor 2b (Fgfr2b) ligands independently of early developmentaldefects, we used an inducible double transgenic system in mice allowing inhibition of Fgfr2b ligands activity. Using vascular morphometry, we quantified the pathological changes. Finally, we evaluated changes in FGF10, surfactant protein C (SFTPC), platelet endothelial cell adhesion molecule (PECAM) and alpha-smooth muscle actin 2 (α-SMA) expression in human lung samples from patients suffering from BPD. In NOX, no major difference in the lung vasculature between Fgf10+/- and control pups was detected. In HOX, a greater loss of blood vessels in Fgf10+/- lungs is associated with an increase of poorly muscularized vessels. Fgfr2b ligands inhibition postnatally in HOX is sufficient to decrease the number of blood vessels while increasing the level of muscularization, suggesting a PH phenotype. BPD lungs exhibited decreased FGF10, SFTPC and PECAM but increased α-SMA. Fgf10 deficiency-associated vascular defects are enhanced in HOX and could represent an additional cause of morbidity in human patients with BPD.


Subject(s)
Bronchopulmonary Dysplasia/etiology , Bronchopulmonary Dysplasia/pathology , Disease Susceptibility , Fibroblast Growth Factor 10/deficiency , Lung/blood supply , Lung/metabolism , Animals , Biomarkers , Bronchopulmonary Dysplasia/metabolism , Computational Biology/methods , Disease Models, Animal , Gene Expression , Gene Expression Profiling , Genotype , Hypoxia , Lung/pathology , Mice , Mutation , Neovascularization, Physiologic/genetics , Oxygen Consumption , Phosphorylation , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Signal Transduction
10.
Harm Reduct J ; 18(1): 122, 2021 12 07.
Article in English | MEDLINE | ID: mdl-34872586

ABSTRACT

BACKGROUND: Worldwide, alcohol-related road traffic accidents represent a major avoidable health risk. The aim of this study was to evaluate the accuracy of self-estimating the degree of acute alcohol intoxication regarding the legal driving limit, and to identify risk factors for misjudgement. METHODS: In this prospective randomised controlled crossover trial, 90 social drinkers (mean age 23.9 ± 3.5 years, 50% female) consumed either beer or wine. Study group subjects were made aware when exceeding the legal driving limit (BrAC = 0.05%). Controls received no information about their BrAC. For crossover, beer or wine were consumed in the opposite order. RESULTS: 39-53% of all participants exceeded the legal driving limit whilst under the impression to be still permitted to drive. Self-estimation was significantly more accurate on study day 2 (p = 0.009). Increasing BrAC positively correlated with self-estimation inaccuracy, which was reproducible during crossover. Multiple regression analysis revealed fast drinking and higher alcohol levels as independent risk factors for inaccurate self-estimation. CONCLUSIONS: Social drinkers are commonly unaware of exceeding the legal driving limit when consuming alcohol. Self-estimating alcohol intoxication can be improved through awareness. Dedicated awareness programs, social media campaigns and government advice communications should be utilised to address this avoidable hazard. Trial registration The trial was registered prospectively at the Witten/Herdecke University Ethics Committee (trial registration number 140/2016 on 04/11/2016) and at the DRKS-German Clinical Trials Register (trial registration number DRKS00015285 on 08/22/2018-Retrospectively registered). Trial protocol can be accessed online.


Subject(s)
Alcoholic Intoxication , Automobile Driving , Adult , Alcohol Drinking , Female , Humans , Male , Perception , Prospective Studies , Young Adult
11.
Int J Mol Sci ; 22(3)2021 Jan 24.
Article in English | MEDLINE | ID: mdl-33498887

ABSTRACT

Bronchopulmonary dysplasia (BPD) remains one of the most devastating consequences of preterm birth resulting in life-long restrictions in lung function. Distorted lung development is caused by its inflammatory response which is mainly provoked by mechanical ventilation, oxygen toxicity and bacterial infections. Dysfunction of resident lung mesenchymal stem cells (MSC) represents one key hallmark that drives BPD pathology. Despite all progress in the understanding of pathomechanisms, therapeutics to prevent or treat BPD are to date restricted to a few drugs. The limited therapeutic efficacy of established drugs can be explained by the fact that they fail to concurrently tackle the broad spectrum of disease driving mechanisms and by the huge overlap between distorted signal pathways of lung development and inflammation. The great enthusiasm about MSC based therapies as novel therapeutic for BPD arises from the capacity to inhibit inflammation while simultaneously promoting lung development and repair. Preclinical studies, mainly performed in rodents, raise hopes that there will be finally a broadly acting, efficient therapy at hand to prevent or treat BPD. Our narrative review gives a comprehensive overview on preclinical achievements, results from first early phase clinical studies and challenges to a successful translation into the clinical setting.


Subject(s)
Bronchopulmonary Dysplasia/therapy , Mesenchymal Stem Cell Transplantation , Bronchopulmonary Dysplasia/prevention & control , Humans , Lung
13.
Genet Med ; 22(3): 511-523, 2020 03.
Article in English | MEDLINE | ID: mdl-31680123

ABSTRACT

PURPOSE: Fetal akinesia has multiple clinical subtypes with over 160 gene associations, but the genetic etiology is not yet completely understood. METHODS: In this study, 51 patients from 47 unrelated families were analyzed using next-generation sequencing (NGS) techniques aiming to decipher the genomic landscape of fetal akinesia (FA). RESULTS: We have identified likely pathogenic gene variants in 37 cases and report 41 novel variants. Additionally, we report putative pathogenic variants in eight cases including nine novel variants. Our work identified 14 novel disease-gene associations for fetal akinesia: ADSSL1, ASAH1, ASPM, ATP2B3, EARS2, FBLN1, PRG4, PRICKLE1, ROR2, SETBP1, SCN5A, SCN8A, and ZEB2. Furthermore, a sibling pair harbored a homozygous copy-number variant in TNNT1, an ultrarare congenital myopathy gene that has been linked to arthrogryposis via Gene Ontology analysis. CONCLUSION: Our analysis indicates that genetic defects leading to primary skeletal muscle diseases might have been underdiagnosed, especially pathogenic variants in RYR1. We discuss three novel putative fetal akinesia genes: GCN1, IQSEC3 and RYR3. Of those, IQSEC3, and RYR3 had been proposed as neuromuscular disease-associated genes recently, and our findings endorse them as FA candidate genes. By combining NGS with deep clinical phenotyping, we achieved a 73% success rate of solved cases.


Subject(s)
Fetal Diseases/genetics , Guanine Nucleotide Exchange Factors/genetics , RNA-Binding Proteins/genetics , Ryanodine Receptor Calcium Release Channel/genetics , Trans-Activators/genetics , Adolescent , Adult , Arthrogryposis/genetics , Arthrogryposis/pathology , Child , Child, Preschool , DNA Copy Number Variations/genetics , Female , Fetal Diseases/pathology , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Humans , Infant , Infant, Newborn , Male , Muscular Diseases/genetics , Muscular Diseases/pathology , Young Adult
14.
Cell Mol Life Sci ; 76(14): 2817-2832, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30887098

ABSTRACT

The respiratory epithelium arises from alveolar epithelial progenitors which differentiate into alveolar epithelial type 1 (AT1) and type 2 (AT2) cells. AT2 cells are stem cells in the lung critical for the repair process after injury. Mechanisms regulating AT1 and AT2 cell maturation are poorly defined. We report that the activation of the glucocorticoid pathway in an in vitro alveolar epithelial lineage differentiation assay led to increased AT2 marker Sftpc and decreased miR-142 expression. Using miR-142 KO mice, we demonstrate an increase in the AT2/AT1 cell number ratio. Overexpression of miR-142 in alveolar progenitor cells in vivo led to the opposite effect. Examination of the KO lungs at E18.5 revealed enhanced expression of miR-142 targets Apc, Ep300 and Kras associated with increased ß-catenin and p-Erk signaling. Silencing of miR-142 expression in lung explants grown in vitro triggers enhanced Sftpc expression as well as increased AT2/AT1 cell number ratio. Pharmacological inhibition of Ep300-ß-catenin but not Erk in vitro prevented the increase in Sftpc expression triggered by loss of miR-142. These results suggest that the glucocorticoid-miR-142-Ep300-ß-catenin signaling axis controls pneumocyte maturation.


Subject(s)
Alveolar Epithelial Cells/cytology , Cell Lineage , Lung/growth & development , MicroRNAs/genetics , Organogenesis , Respiratory Mucosa/cytology , Alveolar Epithelial Cells/metabolism , Animals , Cells, Cultured , Mice , Mice, Knockout , MicroRNAs/metabolism , Respiratory Mucosa/physiology
15.
J Pathol ; 245(2): 153-159, 2018 06.
Article in English | MEDLINE | ID: mdl-29574785

ABSTRACT

Trophic functions for macrophages are emerging as key mediators of developmental processes, including bone, vessel, and mammary gland development. Yolk sac-derived macrophages mature in the distal lung shortly after birth. Myeloid-lineage macrophages are recruited to the lung and are activated under pathological conditions. These pathological conditions include bronchopulmonary dysplasia (BPD), a common complication of preterm birth characterized by stunted lung development, where the formation of alveoli is blocked. No study has addressed causal roles for immune cells in lung alveolarization. We employed antibody-based and transgenic death receptor-based depletion approaches to deplete or prevent lung recruitment of immune cell populations in a hyperoxia-based mouse model of BPD. Neither neutrophils nor exudate macrophages (which might include lung interstitial macrophages) contributed to structural perturbations to the lung that were provoked by hyperoxia; however, cells of the Csf1r-expressing monocyte/macrophage lineage were implicated as causal mediators of stunted lung development. We propose that resident alveolar macrophages differentiate into a population of CD45+ CD11c+ SiglecF+ CD11b+ CD68+ MHCII+ cells, which are activated by hyperoxia, and contribute to disturbances to the structural development of the immature lung. This is the first report that causally implicates immune cells in pathological disturbances to postnatal lung organogenesis. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Bronchopulmonary Dysplasia/pathology , Macrophage Activation , Macrophages, Alveolar/pathology , Pulmonary Alveoli/pathology , Animals , Animals, Newborn , Biomarkers/metabolism , Bronchopulmonary Dysplasia/etiology , Bronchopulmonary Dysplasia/immunology , Bronchopulmonary Dysplasia/metabolism , Cell Proliferation , Disease Models, Animal , Hyperoxia/complications , Hyperoxia/metabolism , Hyperoxia/pathology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Mice, Inbred C57BL , Organogenesis , Phenotype , Pulmonary Alveoli/immunology , Pulmonary Alveoli/metabolism , Signal Transduction
16.
Development ; 142(23): 4139-50, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26511927

ABSTRACT

Lipid-containing alveolar interstitial fibroblasts (lipofibroblasts) are increasingly recognized as an important component of the epithelial stem cell niche in the rodent lung. Although lipofibroblasts were initially believed merely to assist type 2 alveolar epithelial cells in surfactant production during neonatal life, recent evidence suggests that these cells are indispensable for survival and growth of epithelial stem cells during adulthood. Despite increasing interest in lipofibroblast biology, little is known about their cellular origin or the molecular pathways controlling their formation during embryonic development. Here, we show that a population of lipid-droplet-containing stromal cells emerges in the developing mouse lung between E15.5 and E16.5. This is accompanied by significant upregulation, in the lung mesenchyme, of peroxisome proliferator-activated receptor gamma (master switch of lipogenesis), adipose differentiation-related protein (marker of mature lipofibroblasts) and fibroblast growth factor 10 (previously shown to identify a subpopulation of lipofibroblast progenitors). We also demonstrate that although only a subpopulation of total embryonic lipofibroblasts derives from Fgf10(+) progenitor cells, in vivo knockdown of Fgfr2b ligand activity and reduction in Fgf10 expression lead to global reduction in the expression levels of lipofibroblast markers at E18.5. Constitutive Fgfr1b knockouts and mutants with conditional partial inactivation of Fgfr2b in the lung mesenchyme reveal the involvement of both receptors in lipofibroblast formation and suggest a possible compensation between the two receptors. We also provide data from human fetal lungs to demonstrate the relevance of our discoveries to humans. Our results reveal an essential role for Fgf10 signaling in the formation of lipofibroblasts during late lung development.


Subject(s)
Fibroblast Growth Factor 10/metabolism , Fibroblasts/cytology , Gene Expression Regulation, Developmental , Lung/embryology , Pulmonary Alveoli/metabolism , Adipose Tissue/metabolism , Animals , Cell Differentiation , Cell Line , Cell Separation , Cells, Cultured , Epithelial Cells/cytology , Female , Flow Cytometry , Gene Deletion , Humans , Lipids/chemistry , Lung/metabolism , Mice , Mice, Transgenic , PPAR gamma/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Time Factors , Up-Regulation
17.
Stem Cells ; 35(6): 1566-1578, 2017 06.
Article in English | MEDLINE | ID: mdl-28370670

ABSTRACT

ACTA2 expression identifies pulmonary airway and vascular smooth muscle cells (SMCs) as well as alveolar myofibroblasts (MYF). Mesenchymal progenitors expressing fibroblast growth factor 10 (Fgf10), Wilms tumor 1 (Wt1), or glioma-associated oncogene 1 (Gli1) contribute to SMC formation from early stages of lung development. However, their respective contribution and specificity to the SMC and/or alveolar MYF lineages remain controversial. In addition, the contribution of mesenchymal cells undergoing active WNT signaling remains unknown. Using Fgf10CreERT2 , Wt1CreERT2 , Gli1CreERT2 , and Axin2CreERT2 inducible driver lines in combination with a tdTomatoflox reporter line, the respective differentiation of each pool of labeled progenitor cells along the SMC and alveolar MYF lineages was quantified. The results revealed that while FGF10+ and WT1+ cells show a minor contribution to the SMC lineage, GLI1+ and AXIN2+ cells significantly contribute to both the SMC and alveolar MYF lineages, but with limited specificity. Lineage tracing using the Acta2-CreERT2 transgenic line showed that ACTA2+ cells labeled at embryonic day (E)11.5 do not expand significantly to give rise to new SMCs at E18.5. However, ACTA2+ cells labeled at E15.5 give rise to the majority (85%-97%) of the SMCs in the lung at E18.5 as well as alveolar MYF progenitors in the lung parenchyma. Fluorescence-activated cell sorting-based isolation of different subpopulations of ACTA2+ lineage-traced cells followed by gene arrays, identified transcriptomic signatures for alveolar MYF progenitors versus airway and vascular SMCs at E18.5. Our results establish a new transcriptional landscape for further experiments addressing the function of signaling pathways in the formation of different subpopulations of ACTA2+ cells. Stem Cells 2017;35:1566-1578.


Subject(s)
Actins/metabolism , Lung/cytology , Myocytes, Smooth Muscle/metabolism , Animals , Animals, Newborn , Cell Differentiation , Cell Lineage , Cell Separation , Fibroblast Growth Factor 10/metabolism , Lung/embryology , Mice , Models, Biological , Myofibroblasts/cytology , Myofibroblasts/metabolism , Pulmonary Alveoli/cytology , Signal Transduction/genetics , Zinc Finger Protein GLI1/metabolism
18.
J Pathol ; 241(1): 91-103, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27770432

ABSTRACT

Inflammation-induced FGF10 protein deficiency is associated with bronchopulmonary dysplasia (BPD), a chronic lung disease of prematurely born infants characterized by arrested alveolar development. So far, experimental evidence for a direct role of FGF10 in lung disease is lacking. Using the hyperoxia-induced neonatal lung injury as a mouse model of BPD, the impact of Fgf10 deficiency in Fgf10+/- versus Fgf10+/+ pups was investigated. In normoxia, no lethality of Fgf10+/+ or Fgf10+/- pups was observed. By contrast, all Fgf10+/- pups died within 8 days of hyperoxic injury, with lethality starting at day 5, whereas Fgf10+/+ pups were all alive. Lungs of pups from the two genotypes were collected on postnatal day 3 following normoxia or hyperoxia exposure for further analysis. In hyperoxia, Fgf10+/- lungs exhibited increased hypoalveolarization. Analysis by FACS of the Fgf10+/- versus control lungs in normoxia revealed a decreased ratio of alveolar epithelial type II (AECII) cells over total Epcam-positive cells. In addition, gene array analysis indicated reduced AECII and increased AECI transcriptome signatures in isolated AECII cells from Fgf10+/- lungs. Such an imbalance in differentiation is also seen in hyperoxia and is associated with reduced mature surfactant protein B and C expression. Attenuation of the activity of Fgfr2b ligands postnatally in the context of hyperoxia also led to increased lethality with decreased surfactant expression. In summary, decreased Fgf10 mRNA levels lead to congenital lung defects, which are compatible with postnatal survival, but which compromise the ability of the lungs to cope with sub-lethal hyperoxic injury. Fgf10 deficiency affects quantitatively and qualitatively the formation of AECII cells. In addition, Fgfr2b ligands are also important for repair after hyperoxia exposure in neonates. Deficient AECII cells could be an additional complication for patients with BPD. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Bronchopulmonary Dysplasia/metabolism , Fibroblast Growth Factor 10/deficiency , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/etiology , Bronchopulmonary Dysplasia/genetics , Bronchopulmonary Dysplasia/pathology , Cells, Cultured , Disease Models, Animal , Female , Fibroblast Growth Factor 10/genetics , Fibroblast Growth Factor 10/metabolism , Gene Expression Regulation/physiology , Hyperoxia/complications , Hyperoxia/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Pulmonary Surfactants/metabolism , RNA, Messenger/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism
19.
Development ; 141(2): 296-306, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24353064

ABSTRACT

The lung mesenchyme consists of a widely heterogeneous population of cells that play crucial roles during development and homeostasis after birth. These cells belong to myogenic, adipogenic, chondrogenic, neuronal and other lineages. Yet, no clear hierarchy for these lineages has been established. We have previously generated a novel Fgf10(iCre) knock-in mouse line that allows lineage tracing of Fgf10-positive cells during development and postnatally. Using these mice, we hereby demonstrate the presence of two waves of Fgf10 expression during embryonic lung development: the first wave, comprising Fgf10-positive cells residing in the submesothelial mesenchyme at early pseudoglandular stage (as well as their descendants); and the second wave, comprising Fgf10-positive cells from late pseudoglandular stage (as well as their descendants). Our lineage-tracing data reveal that the first wave contributes to the formation of parabronchial and vascular smooth muscle cells as well as lipofibroblasts at later developmental stages, whereas the second wave does not give rise to smooth muscle cells but to lipofibroblasts as well as an Nkx2.1(-) E-Cad(-) Epcam(+) Pro-Spc(+) lineage that requires further in-depth analysis. During alveologenesis, Fgf10-positive cells give rise to lipofibroblasts rather than alveolar myofibroblasts, and during adult life, a subpopulation of Fgf10-expressing cells represents a pool of resident mesenchymal stromal (stem) cells (MSCs) (Cd45(-) Cd31(-) Sca-1(+)). Taken together, we show for the first time that Fgf10-expressing cells represent a pool of mesenchymal progenitors in the embryonic and postnatal lung. Our findings suggest that Fgf10-positive cells could be useful for developing stem cell-based therapies for treating interstitial lung diseases.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Fibroblast Growth Factor 10/metabolism , Lung/embryology , Lung/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Animals , Cell Lineage , Cell Movement , Female , Fibroblast Growth Factor 10/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Gestational Age , Lung/growth & development , Mice , Mice, Transgenic , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Pregnancy , Pulmonary Alveoli/embryology , Pulmonary Alveoli/growth & development , Pulmonary Alveoli/metabolism
20.
Development ; 141(6): 1272-81, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24553287

ABSTRACT

The regulation of the balance between proliferation and differentiation in the mesenchymal compartment of the lung is largely uncharacterized, unlike its epithelial counterpart. In this study, we determined that miR-142-3p contributes to the proper proliferation of mesenchymal progenitors by controlling the level of WNT signaling. miR-142-3p can physically bind to adenomatous polyposis coli mRNA, functioning to regulate its expression level. In miR-142-3p loss-of-function experiments, proliferation of parabronchial smooth muscle cell progenitors is significantly impaired, leading to premature differentiation. Activation of WNT signaling in the mesenchyme, or Apc loss of function, can both rescue miR-142-3p knockdown. These findings show that in the embryonic lung mesenchyme, the microRNA machinery modulates the level of WNT signaling, adding an extra layer of control in the feedback loop between FGFR2C and ß-catenin-mediated WNT signaling.


Subject(s)
Lung/embryology , Lung/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Adenomatous Polyposis Coli Protein/metabolism , Animals , Cell Differentiation , Cell Proliferation , Female , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Genes, APC , Lung/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/antagonists & inhibitors , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Pregnancy , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL