Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 May 15.
Article in English | MEDLINE | ID: mdl-38798483

ABSTRACT

Fibrosis is a common outcome of numerous pathologies, including chronic kidney disease (CKD), a progressive renal function deterioration. Current approaches to target activated fibroblasts, key effector contributors to fibrotic tissue remodeling, lack specificity. Here, we report Gucy1α1 as a specific kidney fibroblast marker. Gucy1α1 levels significantly increased over the course of two clinically relevant murine CKD models and directly correlated with established fibrosis markers. Immunofluorescent (IF) imaging showed that Gucy1α1 comprehensively labelled cortical and medullary quiescent and activated fibroblasts in the control kidney and throughout injury progression, respectively. Unlike traditionally used markers platelet derived growth factor receptor beta (Pdgfrß) and vimentin (Vim), Gucy1α1 did not overlap with off-target populations such as podocytes. Notably, Gucy1α1 labelled kidney fibroblasts in both male and female mice. Furthermore, we observed elevated GUCY1α1 expression in the human fibrotic kidney and lung. Studies in the murine models of cardiac and liver fibrosis revealed Gucy1α1 elevation in activated Pdgfrß-, Vim- and alpha smooth muscle actin (αSma)-expressing fibroblasts paralleling injury progression and resolution. Overall, we demonstrate Gucy1α1 as an exclusive fibroblast marker in both sexes. Due to its multiorgan translational potential, GUCY1α1 might provide a novel promising strategy to specifically target and mechanistically examine fibroblasts.

2.
Am J Respir Cell Mol Biol ; 70(1): 63-77, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37734036

ABSTRACT

It is well known that FGFR2 (fibroblast growth factor receptor 2) signaling is critical for proper lung development. Recent studies demonstrate that epithelial FGFR2 signaling during the saccular phase of lung development (sacculation) regulates alveolar type 1 (AT1) and AT2 cell differentiation. During sacculation, PDGFRA (platelet-derived growth factor receptor-α)-positive lung fibroblasts exist as three functional subtypes: contractile myofibroblasts, extracellular matrix-producing matrix fibroblasts, and lipofibroblasts. All three subtypes are required during alveolarization to establish a niche that supports AT2 epithelial cell self-renewal and AT1 epithelial cell differentiation. FGFR2 signaling directs myofibroblast differentiation in PDGFRA+ fibroblasts during alveolar reseptation after pneumonectomy. However, it remains unknown if FGFR2 signaling regulates PDGFRA+ myo-, matrix, or lipofibroblast differentiation during sacculation. In this study, FGFR2 signaling was inhibited by temporal expression of a secreted dominant-negative FGFR2b (dnFGFR2) by AT2 cells from embryonic day (E) 16.5 to E18.5. Fibroblast and epithelial differentiation were analyzed at E18.5 and postnatal days 7 and 21. At all time points, the number of myofibroblasts was reduced and the number of lipo-/matrix fibroblasts was increased. AT2 cells are increased and AT1 cells are reduced postnatally, but not at E18.5. Similarly, in organoids made with PDGFRA+ fibroblasts from dnFGFR2 lungs, increased AT2 cells and reduced AT1 cells were observed. In vitro treatment of primary wild-type E16.5 adherent saccular lung fibroblasts with recombinant dnFGFR2b/c resulted in reduced myofibroblast contraction. Treatment with the PI3K/AKT activator 740 Y-P rescued the lack of myofibroblast differentiation caused by dnFGFR2b/2c. Moreover, treatment with the PI3K/AKT activator 740 Y-P rescued myofibroblast differentiation in E18.5 fibroblasts isolated from dnFGFR2 lungs.


Subject(s)
Myofibroblasts , Receptor, Fibroblast Growth Factor, Type 2 , Myofibroblasts/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Lung/metabolism , Cell Differentiation , Fibroblasts/metabolism , Cells, Cultured
3.
JCI Insight ; 7(5)2022 03 08.
Article in English | MEDLINE | ID: mdl-35113810

ABSTRACT

Infants born prematurely worldwide have up to a 50% chance of developing bronchopulmonary dysplasia (BPD), a clinical morbidity characterized by dysregulated lung alveolarization and microvascular development. It is known that PDGFR alpha-positive (PDGFRA+) fibroblasts are critical for alveolarization and that PDGFRA+ fibroblasts are reduced in BPD. A better understanding of fibroblast heterogeneity and functional activation status during pathogenesis is required to develop mesenchymal population-targeted therapies for BPD. In this study, we utilized a neonatal hyperoxia mouse model (90% O2 postnatal days 0-7, PN0-PN7) and performed studies on sorted PDGFRA+ cells during injury and room air recovery. After hyperoxia injury, PDGFRA+ matrix and myofibroblasts decreased and PDGFRA+ lipofibroblasts increased by transcriptional signature and population size. PDGFRA+ matrix and myofibroblasts recovered during repair (PN10). After 7 days of in vivo hyperoxia, PDGFRA+ sorted fibroblasts had reduced contractility in vitro, reflecting loss of myofibroblast commitment. Organoids made with PN7 PDGFRA+ fibroblasts from hyperoxia in mice exhibited reduced alveolar type 1 cell differentiation, suggesting reduced alveolar niche-supporting PDGFRA+ matrix fibroblast function. Pathway analysis predicted reduced WNT signaling in hyperoxia fibroblasts. In alveolar organoids from hyperoxia-exposed fibroblasts, WNT activation by CHIR increased the size and number of alveolar organoids and enhanced alveolar type 2 cell differentiation.


Subject(s)
Bronchopulmonary Dysplasia , Hyperoxia , Animals , Bronchopulmonary Dysplasia/etiology , Fibroblasts/metabolism , Humans , Hyperoxia/complications , Infant, Newborn , Lung/pathology , Mice , Myofibroblasts/metabolism
4.
Am J Pathol ; 192(3): 410-425, 2022 03.
Article in English | MEDLINE | ID: mdl-34954211

ABSTRACT

Histopathologic evidence of deployment-related constrictive bronchiolitis (DRCB) has been identified in soldiers deployed to Southwest Asia. While inhalational injury to the airway epithelium is suspected, relatively little is known about the pathogenesis underlying this disabling disorder. Club cells are local progenitors critical for repairing the airway epithelium after exposure to various airborne toxins, and a prior study using an inducible transgenic murine model reported that 10 days of sustained targeted club cell injury causes constrictive bronchiolitis. To further understand the mechanisms leading to small airway fibrosis, a murine model was employed to show that sustained club cell injury elicited acute weight loss, caused increased local production of proinflammatory cytokines, and promoted accumulation of numerous myeloid cell subsets in the lung. Transition to a chronic phase was characterized by up-regulated expression of oxidative stress-associated genes, increased activation of transforming growth factor-ß, accumulation of alternatively activated macrophages, and enhanced peribronchiolar collagen deposition. Comparative histopathologic analysis demonstrated that sustained club cell injury was sufficient to induce epithelial metaplasia, airway wall thickening, peribronchiolar infiltrates, and clusters of intraluminal airway macrophages that recapitulated key abnormalities observed in DRCB. Depletion of alveolar macrophages in mice decreased activation of transforming growth factor-ß and ameliorated constrictive bronchiolitis. Collectively, these findings implicate sustained club cell injury in the development of DRCB and delineate pathways that may yield biomarkers and treatment targets for this disorder.


Subject(s)
Bronchiolitis Obliterans , Animals , Bronchioles/pathology , Bronchiolitis Obliterans/pathology , Disease Models, Animal , Lung/pathology , Mice , Transforming Growth Factor beta/metabolism , Transforming Growth Factors/metabolism
5.
JCI Insight ; 6(14)2021 07 22.
Article in English | MEDLINE | ID: mdl-34138759

ABSTRACT

Mutations in the gene SFTPC, encoding surfactant protein C (SP-C), are associated with interstitial lung disease in children and adults. To assess the natural history of disease, we knocked in a familial, disease-associated SFTPC mutation, L188Q (L184Q [LQ] in mice), into the mouse Sftpc locus. Translation of the mutant proprotein, proSP-CLQ, exceeded that of proSP-CWT in neonatal alveolar type 2 epithelial cells (AT2 cells) and was associated with transient activation of oxidative stress and apoptosis, leading to impaired expansion of AT2 cells during postnatal alveolarization. Differentiation of AT2 to AT1 cells was also inhibited in ex vivo organoid culture of AT2 cells isolated from LQ mice; importantly, treatment with antioxidant promoted alveolar differentiation. Upon completion of alveolarization, SftpcLQ expression was downregulated, leading to resolution of chronic stress responses; however, the failure to restore AT2 cell numbers resulted in a permanent loss of AT2 cells that was linked to decreased regenerative capacity in the adult lung. Collectively, these data support the hypothesis that susceptibility to disease in adult LQ mice is established during postnatal lung development, and they provide a potential explanation for the delayed onset of disease in patients with familial pulmonary fibrosis.


Subject(s)
Alveolar Epithelial Cells/pathology , Genetic Predisposition to Disease , Lung Diseases, Interstitial/genetics , Pulmonary Surfactant-Associated Protein C/genetics , Animals , Animals, Newborn , Cell Differentiation/genetics , Female , Gene Knock-In Techniques , Humans , Lung Diseases, Interstitial/pathology , Mice , Mutation
6.
Stem Cells Transl Med ; 10(7): 1021-1032, 2021 07.
Article in English | MEDLINE | ID: mdl-33624948

ABSTRACT

Developing, regenerating, and repairing a lung all require interstitial resident fibroblasts (iReFs) to direct the behavior of the epithelial stem cell niche. During lung development, distal lung fibroblasts, in the form of matrix-, myo-, and lipofibroblasts, form the extra cellular matrix (ECM), create tensile strength, and support distal epithelial differentiation, respectively. During de novo septation in a murine pneumonectomy lung regeneration model, developmental processes are reactivated within the iReFs, indicating progenitor function well into adulthood. In contrast to the regenerative activation of fibroblasts upon acute injury, chronic injury results in fibrotic activation. In murine lung fibrosis models, fibroblasts can pathologically differentiate into lineages beyond their normal commitment during homeostasis. In lung injury, recently defined alveolar niche cells support the expansion of alveolar epithelial progenitors to regenerate the epithelium. In human fibrotic lung diseases like bronchopulmonary dysplasia (BPD), idiopathic pulmonary fibrosis (IPF), and chronic obstructive pulmonary disease (COPD), dynamic changes in matrix-, myo-, lipofibroblasts, and alveolar niche cells suggest differential requirements for injury pathogenesis and repair. In this review, we summarize the role of alveolar fibroblasts and their activation stage in alveolar septation and regeneration and incorporate them into the context of human lung disease, discussing fibroblast activation stages and how they contribute to BPD, IPF, and COPD.


Subject(s)
Fibroblasts , Lung , Stem Cell Niche , Animals , Bronchopulmonary Dysplasia/pathology , Fibroblasts/cytology , Homeostasis , Humans , Idiopathic Pulmonary Fibrosis/pathology , Lung/cytology , Lung/physiopathology , Mice , Pulmonary Disease, Chronic Obstructive/pathology
7.
Front Fungal Biol ; 2: 740845, 2021.
Article in English | MEDLINE | ID: mdl-37744131

ABSTRACT

Pneumocystis species (spp.) are host-obligate fungal parasites that colonize and propagate almost exclusively in the alveolar lumen within the lungs of mammals where they can cause a lethal pneumonia. The emergence of this pneumonia in non-HIV infected persons caused by Pneumocystis jirovecii (PjP), illustrates the continued importance of and the need to understand its associated pathologies and to develop new therapies and preventative strategies. In the proposed life cycle, Pneumocystis spp. attach to alveolar type 1 epithelial cells (AEC1) and prevent gas exchange. This process among other mechanisms of Pneumocystis spp. pathogenesis is challenging to observe in real time due to the absence of a continuous ex vivo or in vitro culture system. The study presented here provides a proof-of-concept for the development of murine lung organoids that mimic the lung alveolar sacs expressing alveolar epithelial type 1 cells (AEC1) and alveolar type 2 epithelial cells (AEC2). Use of these 3-dimensional organoids should facilitate studies of a multitude of unanswered questions and serve as an improved means to screen new anti- PjP agents.

8.
Matrix Biol ; 91-92: 51-74, 2020 09.
Article in English | MEDLINE | ID: mdl-32442602

ABSTRACT

During lung development, the mesenchyme and epithelium are dependent on each other for instructive morphogenic cues that direct proliferation, cellular differentiation and organogenesis. Specification of epithelial and mesenchymal cell lineages occurs in parallel, forming cellular subtypes that guide the formation of both transitional developmental structures and the permanent architecture of the adult lung. While epithelial cell types and lineages have been relatively well-defined in recent years, the definition of mesenchymal cell types and lineage relationships has been more challenging. Transgenic mouse lines with permanent and inducible lineage tracers have been instrumental in identifying lineage relationships among epithelial progenitor cells and their differentiation into distinct airway and alveolar epithelial cells. Lineage tracing experiments with reporter mice used to identify fibroblast progenitors and their lineage trajectories have been limited by the number of cell specific genes and the developmental timepoint when the lineage trace was activated. In this review, we discuss major developmental mesenchymal lineages, focusing on time of origin, major cell type, and other lineage derivatives, as well as the transgenic tools used to find and define them. We describe lung fibroblasts using function, location, and molecular markers in order to compare and contrast cells with similar functions. The temporal and cell-type specific expression of fourteen "fibroblast lineage" genes were identified in single-cell RNA-sequencing data from LungMAP in the LGEA database. Using these lineage signature genes as guides, we clustered murine lung fibroblast populations from embryonic day 16.5 to postnatal day 28 (E16.5-PN28) and generated heatmaps to illustrate expression of transcription factors, signaling receptors and ligands in a temporal and population specific manner.


Subject(s)
Extracellular Matrix Proteins/genetics , Fibroblasts/cytology , Lung/cytology , Mesenchymal Stem Cells/cytology , Mesoderm/cytology , Animals , Cell Differentiation , Cell Lineage/genetics , Cell Tracking/methods , Cytokines/genetics , Cytokines/metabolism , Embryo, Mammalian , Epithelial Cells/cytology , Epithelial Cells/metabolism , Extracellular Matrix Proteins/metabolism , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Lung/growth & development , Lung/metabolism , Mesenchymal Stem Cells/metabolism , Mesoderm/growth & development , Mesoderm/metabolism , Mice , Mice, Transgenic , Organogenesis/genetics , Regeneration/genetics , Signal Transduction
9.
JCI Insight ; 52019 04 16.
Article in English | MEDLINE | ID: mdl-30990794

ABSTRACT

Obliterative bronchiolitis (OB) is a poorly understood airway disease characterized by the generation of fibrotic bronchiolar occlusions. In the lung transplant setting, OB is a pathological manifestation of bronchiolitis obliterans syndrome (BOS), which is a major impediment to long-term recipient survival. Club cells play a key role in bronchiolar epithelial repair, but whether they promote lung transplant tolerance through preventing OB remains unclear. We determined if OB occurs in mouse orthotopic lung transplants following conditional transgene-targeted club cell depletion. In syngeneic lung transplants club cell depletion leads to transient epithelial injury followed by rapid club cell-mediated repair. In contrast, allogeneic lung transplants develop severe OB lesions and poorly regenerate club cells despite immunosuppression treatment. Lung allograft club cell ablation also triggers the recognition of alloantigens, and pulmonary restricted self-antigens reported associated with BOS development. However, CD8+ T cell depletion restores club cell reparative responses and prevents OB. In addition, ex-vivo analysis reveals a specific role for alloantigen-primed effector CD8+ T cells in preventing club cell proliferation and maintenance. Taken together, we demonstrate a vital role for club cells in maintaining lung transplant tolerance and propose a new model to identify the underlying mechanisms of OB.


Subject(s)
Bronchioles/cytology , Bronchiolitis Obliterans/immunology , Epithelial Cells/immunology , Graft Rejection/immunology , Lung Transplantation/adverse effects , Animals , Bronchioles/immunology , Bronchiolitis Obliterans/pathology , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Graft Rejection/pathology , Humans , Mice , Primary Cell Culture , Respiratory Mucosa/cytology , Respiratory Mucosa/immunology , Transplantation, Homologous/adverse effects
10.
JCI Insight ; 3(17)2018 09 06.
Article in English | MEDLINE | ID: mdl-30185671

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a chronic interstitial lung disease causing fibrotic remodeling of the peripheral lung, leading to respiratory failure. Peripheral pulmonary epithelial cells lose normal alveolar epithelial gene expression patterns and variably express genes associated with diverse conducting airway epithelial cells, including basal cells. Single-cell RNA sequencing of pulmonary epithelial cells isolated from IPF lung tissue demonstrated altered expression of LncRNAs, including increased MEG3. MEG3 RNA was highly expressed in subsets of the atypical IPF epithelial cells and correlated with conducting airway epithelial gene expression patterns. Expression of MEG3 in human pulmonary epithelial cell lines increased basal cell-associated RNAs, including TP63, KRT14, STAT3, and YAP1, and enhanced cell migration, consistent with a role for MEG3 in regulating basal cell identity. MEG3 reduced expression of TP73, SOX2, and Notch-associated RNAs HES1 and HEY1, in primary human bronchial epithelial cells, demonstrating a role for MEG3 in the inhibition of genes influencing basal cell differentiation into club, ciliated, or goblet cells. MEG3 induced basal cell genes and suppressed genes associated with terminal differentiation of airway cells, supporting a role for MEG3 in regulation of basal progenitor cell functions, which may contribute to tissue remodeling in IPF.


Subject(s)
Cell Differentiation/physiology , Epithelial Cells/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Binding Sites , Biomarkers , Cell Differentiation/genetics , Cell Line , Cell Movement , Gene Expression Regulation , Humans , Idiopathic Pulmonary Fibrosis/genetics , Keratin-14/genetics , Lung/metabolism , Promoter Regions, Genetic , STAT3 Transcription Factor/genetics , Sequence Analysis, RNA , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics
11.
JCI Insight ; 3(6)2018 03 22.
Article in English | MEDLINE | ID: mdl-29563341

ABSTRACT

Hippo/YAP signaling plays pleiotropic roles in the regulation of cell proliferation and differentiation during organogenesis and tissue repair. Herein we demonstrate increased YAP activity in respiratory epithelial cells in lungs of patients with idiopathic pulmonary fibrosis (IPF), a common, lethal form of interstitial lung disease (ILD). Immunofluorescence staining in IPF epithelial cells demonstrated increased nuclear YAP and loss of MST1/2. Bioinformatic analyses of epithelial cell RNA profiles predicted increased activity of YAP and increased canonical mTOR/PI3K/AKT signaling in IPF. Phospho-S6 (p-S6) and p-PTEN were increased in IPF epithelial cells, consistent with activation of mTOR signaling. Expression of YAP (S127A), a constitutively active form of YAP, in human bronchial epithelial cells (HBEC3s) increased p-S6 and p-PI3K, cell proliferation and migration, processes that were inhibited by the YAP-TEAD inhibitor verteporfin. Activation of p-S6 was required for enhancing and stabilizing YAP, and the p-S6 inhibitor temsirolimus blocked nuclear YAP localization and suppressed expression of YAP target genes CTGF, AXL, and AJUBA (JUB). YAP and mTOR/p-S6 signaling pathways interact to induce cell proliferation and migration, and inhibit epithelial cell differentiation that may contribute to the pathogenesis of IPF.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Epithelial Cells/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Lung Diseases, Interstitial/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Carrier Proteins/metabolism , Cell Culture Techniques , Cell Differentiation , Cell Movement , Cell Proliferation , Epithelial Cells/pathology , Hepatocyte Growth Factor/metabolism , Hippo Signaling Pathway , Humans , Idiopathic Pulmonary Fibrosis/pathology , Lung/metabolism , Lung/pathology , Lung Diseases, Interstitial/pathology , Membrane Proteins/metabolism , Oncogene Protein v-akt/metabolism , Organogenesis , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/antagonists & inhibitors , Phosphorylation , Proto-Oncogene Proteins/metabolism , Ribosomal Protein S6 Kinases , Serine-Threonine Kinase 3 , Signal Transduction/drug effects , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Transcription Factors , Tumor Suppressor Proteins/metabolism , Verteporfin/pharmacology , YAP-Signaling Proteins
12.
Pediatr Pulmonol ; 52(5): 616-624, 2017 05.
Article in English | MEDLINE | ID: mdl-28186703

ABSTRACT

BACKGROUND: Bronchopulmonary dysplasia (BPD) results from alveolar simplification and abnormal development of alveolar and capillary structure. Survivors of BPD display persistent deficits in airflow and membrane and vascular components of alveolar gas diffusion. Despite being the defining feature of BPD, various neonatal hyperoxia models of BPD have not routinely assessed pulmonary gas diffusion. METHODS: To simulate the most commonly-utilized neonatal hyperoxia models, we exposed neonatal mice to room air or ≥90% hyperoxia during key stages of distal lung development: through the first 4 (saccular), 7 (early alveolar), or 14 (bulk alveolar) postnatal days, followed by a period of recovery in room air until 8 weeks of age when alveolar septation is essentially complete. We systematically assessed and correlated the effects of neonatal hyperoxia on the degree of alveolar-capillary structural and functional impairment. We hypothesized that the degree of alveolar-capillary simplification would correlate strongly with worsening diffusion impairment. RESULTS: Neonatal hyperoxia exposure, of any duration, resulted in alveolar simplification and impaired pulmonary gas diffusion. Mean Linear Intercept increased in proportion to the length of hyperoxia exposure while alveolar and total lung volume increased markedly only with prolonged exposure. Surprisingly, despite having a similar effect on alveolar surface area, only prolonged hyperoxia for 14 days resulted in reduced pulmonary microvascular volume. Estimates of alveolar and capillary structure, in general, correlated poorly with assessment of gas diffusion. CONCLUSION: Our results help define the physiological and structural consequences of commonly-employed neonatal hyperoxia models of BPD and inform their clinical utility. Pediatr Pulmonol. 2017;52:616-624. © 2016 Wiley Periodicals, Inc.


Subject(s)
Bronchopulmonary Dysplasia/physiopathology , Hyperoxia/physiopathology , Lung/physiopathology , Pulmonary Alveoli/physiopathology , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/pathology , Capillaries/pathology , Capillaries/physiopathology , Female , Humans , Hyperoxia/pathology , Infant, Newborn , Lung/pathology , Mice , Pulmonary Alveoli/pathology
13.
JCI Insight ; 1(20): e90558, 2016 12 08.
Article in English | MEDLINE | ID: mdl-27942595

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a lethal interstitial lung disease characterized by airway remodeling, inflammation, alveolar destruction, and fibrosis. We utilized single-cell RNA sequencing (scRNA-seq) to identify epithelial cell types and associated biological processes involved in the pathogenesis of IPF. Transcriptomic analysis of normal human lung epithelial cells defined gene expression patterns associated with highly differentiated alveolar type 2 (AT2) cells, indicated by enrichment of RNAs critical for surfactant homeostasis. In contrast, scRNA-seq of IPF cells identified 3 distinct subsets of epithelial cell types with characteristics of conducting airway basal and goblet cells and an additional atypical transitional cell that contributes to pathological processes in IPF. Individual IPF cells frequently coexpressed alveolar type 1 (AT1), AT2, and conducting airway selective markers, demonstrating "indeterminate" states of differentiation not seen in normal lung development. Pathway analysis predicted aberrant activation of canonical signaling via TGF-ß, HIPPO/YAP, P53, WNT, and AKT/PI3K. Immunofluorescence confocal microscopy identified the disruption of alveolar structure and loss of the normal proximal-peripheral differentiation of pulmonary epithelial cells. scRNA-seq analyses identified loss of normal epithelial cell identities and unique contributions of epithelial cells to the pathogenesis of IPF. The present study provides a rich data source to further explore lung health and disease.


Subject(s)
Epithelial Cells/cytology , Idiopathic Pulmonary Fibrosis/pathology , Sequence Analysis, RNA , Gene Expression Profiling , Genetic Markers , Humans , Lung/cytology , Single-Cell Analysis , Transcriptome
14.
Dev Dyn ; 245(5): 590-604, 2016 05.
Article in English | MEDLINE | ID: mdl-26869074

ABSTRACT

BACKGROUND: Lung morphogenesis is regulated by interactions between the canonical Wnt/ß-catenin and Kras/ERK/Foxm1 signaling pathways that establish proximal-peripheral patterning of lung tubules. How these interactions influence the development of respiratory epithelial progenitors to acquire airway as compared to alveolar epithelial cell fate is unknown. During branching morphogenesis, SOX9 transcription factor is normally restricted from conducting airway epithelial cells and is highly expressed in peripheral, acinar progenitor cells that serve as precursors of alveolar type 2 (AT2) and AT1 cells as the lung matures. RESULTS: To identify signaling pathways that determine proximal-peripheral cell fate decisions, we used the SFTPC gene promoter to delete or overexpress key members of Wnt/ß-catenin and Kras/ERK/Foxm1 pathways in fetal respiratory epithelial progenitor cells. Activation of ß-catenin enhanced SOX9 expression in peripheral epithelial progenitors, whereas deletion of ß-catenin inhibited SOX9. Surprisingly, deletion of ß-catenin caused accumulation of atypical SOX9-positive basal cells in conducting airways. Inhibition of Wnt/ß-catenin signaling by Kras(G12D) or its downstream target Foxm1 stimulated SOX9 expression in basal cells. Genetic inactivation of Foxm1 from Kras(G12D) -expressing epithelial cells prevented the accumulation of SOX9-positive basal cells in developing airways. CONCLUSIONS: Interactions between the Wnt/ß-catenin and the Kras/ERK/Foxm1 pathways are essential to restrict SOX9 expression in basal cells. Developmental Dynamics 245:590-604, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Forkhead Box Protein M1/metabolism , Lung/embryology , Morphogenesis , Proto-Oncogene Proteins p21(ras)/metabolism , SOX9 Transcription Factor/metabolism , Signal Transduction/physiology , beta Catenin/metabolism , Animals , Body Patterning , Embryo, Mammalian , Epithelial Cells , Mice , SOX9 Transcription Factor/analysis , Stem Cells
15.
Am J Respir Cell Mol Biol ; 54(4): 532-45, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26414960

ABSTRACT

Epithelial-mesenchymal cell interactions and factors that control normal lung development are key players in lung injury, repair, and fibrosis. A number of studies have investigated the roles and sources of epithelial progenitors during lung regeneration; such information, however, is limited in lung fibroblasts. Thus, understanding the origin, phenotype, and roles of fibroblast progenitors in lung development, repair, and regeneration helps address these limitations. Using a combination of platelet-derived growth factor receptor α-green fluorescent protein (PDGFRα-GFP) reporter mice, microarray, real-time polymerase chain reaction, flow cytometry, and immunofluorescence, we characterized two distinct interstitial resident fibroblasts, myo- and matrix fibroblasts, and identified a role for PDGFRα kinase activity in regulating their activation during lung regeneration. Transcriptional profiling of the two populations revealed a myo- and matrix fibroblast gene signature. Differences in proliferation, smooth muscle actin induction, and lipid content in the two subpopulations of PDGFRα-expressing fibroblasts during alveolar regeneration were observed. Although CD140α(+)CD29(+) cells behaved as myofibroblasts, CD140α(+)CD34(+) appeared as matrix and/or lipofibroblasts. Gain or loss of PDGFRα kinase activity using the inhibitor nilotinib and a dominant-active PDGFRα-D842V mutation revealed that PDGFRα was important for matrix fibroblast differentiation. We demonstrated that PDGFRα signaling promotes alveolar septation by regulating fibroblast activation and matrix fibroblast differentiation, whereas myofibroblast differentiation was largely PDGFRα independent. These studies provide evidence for the phenotypic and functional diversity as well as the extent of specificity of interstitial resident fibroblasts differentiation during regeneration after partial pneumonectomy.


Subject(s)
Lung/cytology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Animals , Antigens, CD/immunology , Green Fluorescent Proteins/genetics , Immunophenotyping , Lung/enzymology , Lung/immunology , Lung/physiology , Mice , Mice, Transgenic , Receptor, Platelet-Derived Growth Factor alpha/genetics , Regeneration
16.
Am J Respir Cell Mol Biol ; 47(4): 517-27, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22652199

ABSTRACT

Although the importance of platelet-derived growth factor receptor (PDGFR)-α signaling during normal alveogenesis is known, it is unclear whether this signaling pathway can regulate realveolarization in the adult lung. During alveolar development, PDGFR-α-expressing cells induce α smooth muscle actin (α-SMA) and differentiate to interstitial myofibroblasts. Fibroblast growth factor (FGF) signaling regulates myofibroblast differentiation during alveolarization, whereas peroxisome proliferator-activated receptor (PPAR)-γ activation antagonizes myofibroblast differentiation in lung fibrosis. Using left lung pneumonectomy, the roles of FGF and PPAR-γ signaling in differentiation of myofibroblasts from PDGFR-α-positive precursors during compensatory lung growth were assessed. FGF receptor (FGFR) signaling was inhibited by conditionally activating a soluble dominant-negative FGFR2 transgene. PPAR-γ signaling was activated by administration of rosiglitazone. Changes in α-SMA and PDGFR-α protein expression were assessed in PDGFR-α-green fluorescent protein (GFP) reporter mice using immunohistochemistry, flow cytometry, and real-time PCR. Immunohistochemistry and flow cytometry demonstrated that the cell ratio and expression levels of PDGFR-α-GFP changed dynamically during alveolar regeneration and that α-SMA expression was induced in a subset of PDGFR-α-GFP cells. Expression of a dominant-negative FGFR2 and administration of rosiglitazone inhibited induction of α-SMA in PDGFR-α-positive fibroblasts and formation of new septae. Changes in gene expression of epithelial and mesenchymal signaling molecules were assessed after left lobe pneumonectomy, and results demonstrated that inhibition of FGFR2 signaling and increase in PPAR-γ signaling altered the expression of Shh, FGF, Wnt, and Bmp4, genes that are also important for epithelial-mesenchymal crosstalk during early lung development. Our data demonstrate for the first time that a comparable epithelial-mesenchymal crosstalk regulates fibroblast phenotypes during alveolar septation.


Subject(s)
Gene Expression Regulation , Myofibroblasts/metabolism , Pulmonary Alveoli/pathology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Regeneration , Actins/metabolism , Animals , Cell Differentiation , Cells, Cultured , Fibroblasts/physiology , Genes, Dominant , Lung/metabolism , Lung/pathology , Lung/physiopathology , Mice , Mice, Transgenic , Myofibroblasts/physiology , PPAR gamma/agonists , Phenotype , Pneumonectomy , Pulmonary Alveoli/physiopathology , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Rosiglitazone , Signal Transduction , Thiazolidinediones/pharmacology , Transcription, Genetic
17.
Am J Respir Crit Care Med ; 183(4): 511-21, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-20870756

ABSTRACT

RATIONALE: The respiratory epithelium has a remarkable capacity to respond to acute injury. In contrast, repeated epithelial injury is often associated with abnormal repair, inflammation, and fibrosis. There is increasing evidence that nonciliated epithelial cells play important roles in the repair of the bronchiolar epithelium after acute injury. Cellular processes underlying the repair and remodeling of the lung after chronic epithelial injury are poorly understood. OBJECTIVES: To identify cell processes mediating epithelial regeneration and remodeling after acute and chronic Clara cell depletion. METHODS: A transgenic mouse model was generated to conditionally express diphtheria toxin A to ablate Clara cells in the adult lung. Epithelial regeneration and peribronchiolar fibrosis were assessed after acute and chronic Clara cell depletion. MEASUREMENTS AND MAIN RESULTS: Acute Clara cell ablation caused squamous metaplasia of ciliated cells and induced proliferation of residual progenitor cells. Ciliated cells in the bronchioles and pro-surfactant protein C-expressing cells in the bronchiolar alveolar duct junctions did not proliferate. Epithelial cell proliferation occurred at multiple sites along the airways and was not selectively associated with regions around neuroepithelial bodies. Chronic Clara cell depletion resulted in ineffective repair and caused peribronchiolar fibrosis. CONCLUSIONS: Colocalization of proliferation and cell type-specific markers demonstrate that Clara cells are critical airway progenitor cells. Continuous depletion of Clara cells resulted in persistent squamous metaplasia, lack of normal reepithelialization, and peribronchiolar fibrosis. Induction of proliferation in subepithelial fibroblasts supports the concept that chronic epithelial depletion caused peribronchiolar fibrosis.


Subject(s)
Bronchi/metabolism , Bronchi/pathology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Stem Cells/metabolism , Stem Cells/pathology , Animals , Bronchi/cytology , Cells, Cultured , Diphtheria Toxin , Disease Models, Animal , Fibrosis , Lung/cytology , Lung/metabolism , Lung/pathology , Mice , Mice, Transgenic , Respiratory Mucosa/cytology , Stem Cells/cytology
18.
Cell Cycle ; 9(14): 2769-76, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20676040

ABSTRACT

Pulmonary fibrosis complicates a number of disease processes and leads to substantial morbidity and mortality. Idiopathic pulmonary fibrosis (IPF) is perhaps the most pernicious and enigmatic form of the greater problem of lung fibrogenesis with a median survival of three years from diagnosis in affected patients. In this review, we will focus on the pathology of IPF as a model of pulmonary fibrotic processes, review possible cellular mechanisms, review current treatment approaches and review two transgenic mouse models of lung fibrosis to provide insight into processes that cause lung fibrosis. We will also summarize the potential utility of signaling pathway inhibitors as a future treatment in pulmonary fibrosis. Finally, we will present data demonstrating a minimal contribution of epithelial-mesenchymal transition in the development of fibrotic lesions in the transforming growth factor-alpha transgenic model of lung fibrosis.


Subject(s)
Epithelial Cells/metabolism , Pulmonary Fibrosis/etiology , Signal Transduction , Animals , Disease Models, Animal , Epithelial Cells/cytology , ErbB Receptors/metabolism , Humans , Mesoderm/cytology , Mesoderm/metabolism , Mice , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/therapy , Pulmonary Surfactant-Associated Protein C/genetics , Pulmonary Surfactant-Associated Protein C/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
19.
Am J Physiol Lung Cell Mol Physiol ; 297(2): L299-308, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19502291

ABSTRACT

Normal alveolarization has been studied in rodents using detailed morphometric techniques and loss of function approaches for growth factors and their receptors. However, it remains unclear how these growth factors direct the formation of secondary septae. We have previously developed a transgenic mouse model in which expression of a soluble dominant-negative FGF receptor (dnFGFR) in the prenatal period results in reduced alveolar septae formation and subsequent alveolar simplification. Retinoic acid (RA), a biologically active derivative of vitamin A, can induce regeneration of alveoli in adult rodents. In this study, we demonstrate that RA induces alveolar reseptation in this transgenic mouse model and that realveolarization in adult mice is FGF dependent. Proliferation in the lung parenchyma, an essential prerequisite for lung regrowth was enhanced after 14 days of RA treatment and was not influenced by dnFGFR expression. During normal lung development, formation of secondary septae is associated with the transient presence of alpha-smooth muscle actin (alphaSMA)-positive interstitial myofibroblasts. One week after completion of RA treatment, alphaSMA expression was detected in interstitial fibroblasts, supporting the concept that RA-initiated realveolarization recapitulates aspects of septation that occur during normal lung development. Expression of dnFGFR blocked realveolarization with increased PDGF receptor-alpha (PDGFRalpha)-positive cells and decreased alphaSMA-positive cells. Taken together, our data demonstrate that FGF signaling is required for the induction of alphaSMA in the PDGFRalpha-positive myofibroblast progenitor and the progression of alveolar regeneration.


Subject(s)
Fibroblasts/physiology , Pulmonary Alveoli/cytology , Pulmonary Alveoli/physiology , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Regeneration/physiology , Signal Transduction/physiology , Actins/metabolism , Age Factors , Animals , Antineoplastic Agents/pharmacology , Cell Division/drug effects , Cell Division/physiology , Elastin/metabolism , Fibroblast Growth Factors/metabolism , Fibroblasts/cytology , Green Fluorescent Proteins/genetics , Humans , Mice , Mice, Transgenic , Pulmonary Alveoli/growth & development , Pulmonary Surfactant-Associated Protein C/genetics , Pulmonary Surfactant-Associated Protein C/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Signal Transduction/drug effects , Tretinoin/pharmacology
20.
Mol Cancer Res ; 6(3): 468-82, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18337453

ABSTRACT

Sprouty (Spry) proteins modulate signal transduction pathways elicited by receptor tyrosine kinases (RTK). Depending on cell type and the particular RTK, Spry proteins exert dual functions: They can either repress RTK-mediated signaling pathways, mainly by interfering with the Ras/Raf/mitogen-activated protein kinase pathway or sustaining RTK signal transduction, for example by sequestering the E3 ubiquitin-ligase c-Cbl and thus preventing ubiquitylation, internalization, and degradation of RTKs. Here, by the inducible expression of murine Spry4 in pancreatic beta cells, we have assessed the functional role of Spry proteins in the development of pancreatic islets of Langerhans in normal mice and in the Rip1Tag2 transgenic mouse model of beta-cell carcinogenesis. beta cell-specific expression of mSpry4 provokes a significant reduction in islet size, an increased number of alpha cells per islet area, and impaired islet cell type segregation. Functional analysis of islet cell differentiation in cultured PANC-1 cells shows that mSpry4 represses adhesion and migration of differentiating pancreatic endocrine cells, most likely by affecting the subcellular localization of the protein tyrosine phosphatase PTP1B. In contrast, transgenic expression of mSpry4 during beta-cell carcinogenesis does not significantly affect tumor outgrowth and progression to tumor malignancy. Rather, tumor cells seem to escape mSpry4 transgene expression.


Subject(s)
Insulin-Secreting Cells/physiology , Islets of Langerhans/physiology , Nerve Tissue Proteins/physiology , Pancreatic Neoplasms/pathology , Animals , Cell Line, Tumor , DNA Primers , Disease Progression , Gene Expression Regulation, Neoplastic , Glucose Tolerance Test , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nuclear Pore Complex Proteins/genetics , Pancreatic Neoplasms/genetics , Polymerase Chain Reaction , RNA-Binding Proteins/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL