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

Country/Region as subject
Affiliation country
Publication year range
1.
Am J Physiol Cell Physiol ; 326(6): C1659-C1668, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38646784

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is marked by the activation of fibroblasts, leading to excessive production and deposition of extracellular matrix (ECM) within the lung parenchyma. Despite the pivotal role of ECM overexpression in IPF, potential negative regulators of ECM production in fibroblasts have yet to be identified. Semaphorin class 3B (SEMA3B), a secreted protein highly expressed in lung tissues, has established roles in axonal guidance and tumor suppression. However, the role of SEMA3B in ECM production by fibroblasts in the pathogenesis of IPF remains unexplored. Here, we show the downregulation of SEMA3B and its cognate binding receptor, neuropilin 1 (NRP1), in IPF lungs compared with healthy controls. Notably, the reduced expression of SEMA3B and NRP1 is associated with a decline in lung function in IPF. The downregulation of SEMA3B and NRP1 transcripts was validated in the lung tissues of patients with IPF, and two alternative mouse models of pulmonary fibrosis. In addition, we show that transforming growth factor-ß (TGFß) functions as a negative regulator of SEMA3B and NRP1 expression in lung fibroblasts. Furthermore, we demonstrate the antifibrotic effects of SEMA3B against TGFß-induced ECM production in IPF lung fibroblasts. Overall, our findings uncovered a novel role of SEMA3B in the pathogenesis of pulmonary fibrosis and provided novel insights into modulating the SEMA3B-NRP1 axis to attenuate pulmonary fibrosis.NEW & NOTEWORTHY The excessive production and secretion of collagens and other extracellular matrix proteins by fibroblasts lead to the scarring of the lung in severe fibrotic lung diseases. This study unveils an antifibrotic role for semaphorin class 3B (SEMA3B) in the pathogenesis of idiopathic pulmonary fibrosis. SEMA3B functions as an inhibitor of transforming growth factor-ß-driven fibroblast activation and reduced levels of SEMA3B and its receptor, neuropilin 1, are associated with decreased lung function in idiopathic pulmonary fibrosis.


Subject(s)
Extracellular Matrix Proteins , Fibroblasts , Idiopathic Pulmonary Fibrosis , Lung , Neuropilin-1 , Semaphorins , Transforming Growth Factor beta , Animals , Female , Humans , Male , Mice , Middle Aged , Cells, Cultured , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/genetics , Fibroblasts/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Idiopathic Pulmonary Fibrosis/genetics , Lung/metabolism , Lung/pathology , Membrane Glycoproteins , Mice, Inbred C57BL , Neuropilin-1/metabolism , Neuropilin-1/genetics , Semaphorins/metabolism , Semaphorins/genetics , Transforming Growth Factor beta/metabolism
2.
Pediatr Surg Int ; 40(1): 102, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38589706

ABSTRACT

PURPOSE: The utility of pulmonary function testing (PFT) in pectus excavatum (PE) has been subject to debate. Although some evidence shows improvement from preoperative to postoperative values, the clinical significance is uncertain. A high failure-to-completion rate for operative PFT (48%) was identified in our large institutional cohort. With such a high non-completion rate, we questioned the overall utility of PFT in the preoperative assessment of PE and sought to evaluate if other measures of PE severity or cardiopulmonary function could explain this finding. METHODS: Demographics, clinical findings, and results from cardiac MRI, PFT (spirometry and plethysmography), and cardiopulmonary exercise tests (CPET) were reviewed in 270 patients with PE evaluated preoperatively between 2015 and 2018. Regression modeling was used to measure associations between PFT completion and cardiopulmonary function. RESULTS: There were no differences in demographics, symptoms, connective tissue disorders, or multiple indices of pectus severity and cardiac deformation in PFT completers versus non-completers. While regression analysis revealed higher RVEF, LVEF, and LVEF-Z scores, lower RV-ESV/BSA, LV-ESV/BSA, and LV-ESV/BSA-Z scores, and abnormal breathing reserve in PFT completers vs. non-completers, these findings were not consistent across continuous and binary analyses. CONCLUSIONS: We found that PFT completers were not significantly different from non-completers in most structural and functional measures of pectus deformity and cardiopulmonary function. Inability to complete PFT is not an indicator of pectus severity.


Subject(s)
Funnel Chest , Humans , Funnel Chest/surgery , Spirometry
3.
Pediatr Surg Int ; 39(1): 52, 2022 Dec 16.
Article in English | MEDLINE | ID: mdl-36525122

ABSTRACT

PURPOSE: We sought to analyze differences in presentation and cardiopulmonary function between those referred for surgical consultation as adolescents (11-17 years) versus adults (18 + years). METHODS: Presenting symptoms, past medical history, and results from cardiac MRI (CMR), pulmonary function testing (PFT), and cardiopulmonary exercise testing (CPET) were reviewed in 329 patients evaluated preoperatively between 2015 and 2018. Adjusted regression modeling was used to measure associations between pectus indices and clinical endpoints of cardiopulmonary function. RESULTS: Our sample included 276 adolescents and 53 adults. Adults presented more frequently with chest pain (57% vs. 38%, p = 0.01), shortness of breath (76% vs. 59%, p = 0.02), palpitations (21% vs. 11%, p = 0.04), and exercise intolerance (76% vs. 59%, p = 0.02). Their Haller indices (5.2 [4.2, 7.0] vs. 4.7 [4.0, 5.7], p = 0.05) and cardiac asymmetry (1.8 [0.5] vs. 1.6 [0.5], p = 0.02) were also higher. In continuous outcome analysis, adolescents had higher FEV1/FVC on PFT and higher work on CPET (p < 0.01). CONCLUSIONS: Adults with pectus excavatum were more symptomatic than adolescents with deeper, more asymmetric deformities, decreased FEV1/FVC and exercise capacity. These findings may support earlier versus later repair to prevent age-related decline. Further studies are warranted.


Subject(s)
Funnel Chest , Humans , Adolescent , Adult , Funnel Chest/surgery , Respiratory Function Tests/methods , Magnetic Resonance Imaging
4.
Pediatr Surg Int ; 36(11): 1281-1286, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32940825

ABSTRACT

PURPOSE: Pectus excavatum (PE) is a chest wall deformity of variable severity and symptomatology. Existing female-specific literature highlights breast asymmetry and cosmetic reconstruction. We sought to evaluate gender differences in cardiopulmonary function. METHODS: Cardiac MRIs, pulmonary function tests (PFTs), and cardiopulmonary exercise tests (CPETs) were reviewed in 345 patients undergoing preoperative evaluation for PE. Regression modeling was used to evaluate associations between gender and clinical endpoints of cardiopulmonary function. RESULTS: Mean age was 15.2 years, 19% were female, 98% were white. Pectus indices included median Haller Index (HI) of 4.8, mean depression index (DI) of 0.63, correction index (CI) of 33.6%, and Cardiac Compression Index (CCI) of 2.79. Cardiac assessment revealed decreased right and left ventricular ejection fraction (RVEF, LVEF) in 16% and 22% of patients, respectively. PFTs and CPETs were abnormal in ~ 30% of patients. While females had deeper PE deformities-represented by higher pectus indices-they had superior function with higher RVEF, LVEF Z-scores, FEV1, VO2 max, O2 pulse, work, and breathing reserve (p < 0.05). CONCLUSION: Despite worse PE deformity and symptomatology, females had a better cardiopulmonary function and exercise tolerance than males. Further research is needed to assess the precise mechanisms of this phenomenon and postoperative outcomes in this population.


Subject(s)
Exercise Tolerance/physiology , Funnel Chest/physiopathology , Heart Rate/physiology , Stroke Volume/physiology , Thoracic Wall/physiopathology , Ventricular Function, Left/physiology , Adolescent , Adult , Child , Child, Preschool , Female , Funnel Chest/epidemiology , Humans , Incidence , Magnetic Resonance Imaging , Male , Sex Factors , United States/epidemiology , Young Adult
5.
Am J Physiol Lung Cell Mol Physiol ; 315(3): L456-L465, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29877096

ABSTRACT

Cystic fibrosis (CF) produces variable lung disease phenotypes that are, in part, independent of the CF transmembrane conductance regulator ( CFTR) genotype. Transforming growth factor-ß (TGFß) is the best described genetic modifier of the CF phenotype, but its mechanism of action is unknown. We hypothesized that TGFß is sufficient to drive pathognomonic features of CF lung disease in vivo and that CFTR deficiency enhances susceptibility to pathological TGFß effects. A CF mouse model and littermate controls were exposed intratracheally to an adenoviral vector containing the TGFß1 cDNA (Ad-TGFß), empty vector, or PBS only. Studies were performed 1 wk after treatment, including lung mechanics, collection of bronchoalveolar lavage fluid, and analysis of lung histology, RNA, and protein. CF and non-CF mice showed similar weight loss, inflammation, goblet cell hyperplasia, and Smad pathway activation after Ad-TGFß treatment. Ad-TGFß produced greater abnormalities in lung mechanics in CF versus control mice, which was uniquely associated with induction of phosphoinositide 3-kinase and mitogen-activated protein kinase signaling. CFTR transcripts were reduced, and epithelial sodium channel transcripts were increased in CF and non-CF mice, whereas the goblet cell transcription factors, forkhead ortholog A3 and SAM-pointed domain-containing ETS-like factor, were increased in non-CF but not CF mice following Ad-TGFß treatment. Pulmonary TGFß1 expression was sufficient to produce pulmonary remodeling and abnormalities in lung mechanics that were associated with both shared and unique cell signaling pathway activation in CF and non-CF mice. These results highlight the multifunctional impact of TGFß on pulmonary pathology in vivo and identify cellular-response differences that may impact CF lung pathology.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/metabolism , Gene Expression Regulation , Goblet Cells/metabolism , Lung/metabolism , Transforming Growth Factor beta1/biosynthesis , Adenoviridae , Animals , Cystic Fibrosis/genetics , Cystic Fibrosis/pathology , Cystic Fibrosis/physiopathology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Goblet Cells/pathology , Hyperplasia , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Lung/pathology , Lung/physiopathology , Mice , Mice, Transgenic , Transduction, Genetic , Transforming Growth Factor beta1/genetics
7.
Am J Physiol Lung Cell Mol Physiol ; 312(4): L488-L499, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28130263

ABSTRACT

Pulmonary fibrosis contributes to morbidity and mortality in a range of diseases, and there are no approved therapies for reversing its progression. To understand the mechanisms underlying pulmonary fibrosis and assess potential therapies, mouse models are central to basic and translational research. Unfortunately, metrics commonly used to assess murine pulmonary fibrosis require animals to be grouped and euthanized, increasing experimental difficulty and cost. We examined the ability of magnetic resonance imaging (MRI) to noninvasively assess lung fibrosis progression and resolution in a doxycycline (Dox) regulatable, transgenic mouse model that overexpresses transforming growth factor-α (TGF-α) under control of a lung-epithelial-specific promoter. During 7 wk of Dox treatment, fibrotic lesions were readily observed as high-signal tissue. Mean weighted signal and percent signal volume were found to be the most robust MRI-derived measures of fibrosis, and these metrics correlated significantly with pleural thickness, histology scores, and hydroxyproline content (R = 0.75-0.89). When applied longitudinally, percent high signal volume increased by 1.5% wk-1 (P < 0.001) and mean weighted signal increased at a rate of 0.0065 wk-1 (P = 0.0062). Following Dox treatment, lesions partially resolved, with percent high signal volume decreasing by -3.2% wk-1 (P = 0.0034) and weighted mean signal decreasing at -0.015 wk-1 (P = 0.0028). Additionally, longitudinal MRI revealed dynamic remodeling in a subset of lesions, a previously unobserved behavior in this model. These results demonstrate MRI can noninvasively assess experimental lung fibrosis progression and resolution and provide unique insights into its pathobiology.


Subject(s)
Disease Progression , Magnetic Resonance Imaging/methods , Pulmonary Fibrosis/pathology , Animals , Disease Models, Animal , Hydroxyproline/metabolism , Imaging, Three-Dimensional , Mice , Mice, Transgenic , Transforming Growth Factor alpha/pharmacology
8.
Am J Pathol ; 186(5): 1066-77, 2016 05.
Article in English | MEDLINE | ID: mdl-27021937

ABSTRACT

Fibrogenesis involves a dynamic interplay between factors that promote the biosynthesis and deposition of extracellular matrix along with pathways that degrade the extracellular matrix and eliminate the primary effector cells. Opposing the often held perception that fibrotic tissue is permanent, animal studies and clinical data now demonstrate the highly plastic nature of organ fibrosis that can, under certain circumstances, regress. This review describes the current understanding of the mechanisms whereby the lung is known to resolve fibrosis focusing on degradation of the extracellular matrix, removal of myofibroblasts, and the role of inflammatory cells. Although there are significant gaps in understanding lung fibrosis resolution, accelerated improvements in biotechnology and bioinformatics are expected to improve the understanding of these mechanisms and have high potential to lead to novel and effective restorative therapies in the treatment not only of pulmonary fibrosis, but also of a wide-ranging spectrum of chronic disorders.


Subject(s)
Extracellular Matrix/metabolism , Pulmonary Fibrosis/physiopathology , Animals , Collagen/physiology , Enzymes/physiology , Extracellular Matrix/physiology , Humans , Lysosomes/metabolism , Mice , Models, Animal , Proteolysis , Pulmonary Fibrosis/metabolism
9.
J Immunol ; 195(8): 3978-91, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26371248

ABSTRACT

Collagen-producing myofibroblast transdifferentiation is considered a crucial determinant in the formation of scar tissue in the lungs of patients with idiopathic pulmonary fibrosis. Multiple resident pulmonary cell types and bone marrow-derived fibrocytes have been implicated as contributors to fibrotic lesions because of the transdifferentiation potential of these cells into myofibroblasts. In this study, we assessed the expression of Wilms tumor 1 (WT1), a known marker of mesothelial cells, in various cell types in normal and fibrotic lungs. We demonstrate that WT1 is expressed by both mesothelial and mesenchymal cells in idiopathic pulmonary fibrosis lungs but has limited or no expression in normal human lungs. We also demonstrate that WT1(+) cells accumulate in fibrotic lung lesions, using two different mouse models of pulmonary fibrosis and WT1 promoter-driven fluorescent reporter mice. Reconstitution of bone marrow cells into a TGF-α transgenic mouse model demonstrated that fibrocytes do not transform into WT1(+) mesenchymal cells, but they do augment accumulation of WT1(+) cells in severe fibrotic lung disease. Importantly, the number of WT1(+) cells in fibrotic lesions was correlated with severity of lung disease as assessed by changes in lung function, histology, and hydroxyproline levels in mice. Finally, inhibition of WT1 expression was sufficient to attenuate collagen and other extracellular matrix gene production by mesenchymal cells from both murine and human fibrotic lungs. Thus, the results of this study demonstrate a novel association between fibrocyte-driven WT1(+) cell accumulation and severe fibrotic lung disease.


Subject(s)
Gene Expression Regulation/immunology , Idiopathic Pulmonary Fibrosis/immunology , Lung/immunology , Repressor Proteins/immunology , WT1 Proteins/immunology , Animals , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/immunology , Female , Humans , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Lung/pathology , Male , Mice , Mice, Transgenic , Repressor Proteins/genetics , Transforming Growth Factor alpha/genetics , Transforming Growth Factor alpha/immunology , WT1 Proteins/genetics
10.
Am J Respir Cell Mol Biol ; 55(6): 792-803, 2016 12.
Article in English | MEDLINE | ID: mdl-27438654

ABSTRACT

The p70 ribosomal S6 kinase (p70S6K) is a downstream substrate that is phosphorylated and activated by the mammalian target of rapamycin complex and regulates multiple cellular processes associated with pulmonary fibrogenesis. Two isoforms of the p70S6K have been identified (S6K1 and S6K2), but their relative contributions in mediating pulmonary fibrosis are unknown. To interrogate the roles of the p70S6K isoforms, we overexpressed transforming growth factor (TGF)-α in mice deficient for the S6K1 or S6K2 genes and measured changes in lung histology, morphometry, total lung collagen, lung function, and proliferation between wild-type and isoform-deficient mice. Deficiency of S6K1, but not S6K2, had a significant effect on reducing proliferation in subpleural fibrotic lesions during TGF-α-induced fibrosis. Migration was significantly decreased in mesenchymal cells isolated from the lungs of S6K1 knockout mice compared with wild-type or S6K2 knockout mice. Conversely, increases in subpleural thickening were significantly decreased in S6K2-deficient mice compared with wild type. Deficiency of S6K2 significantly reduced phosphorylation of the downstream S6 ribosomal protein in lung homogenates and isolated mesenchymal cells after TGF-α expression. However, deficiency of neither isoform alone significantly altered TGF-α-induced collagen accumulation or lung function decline in vivo. Furthermore, deficiency in neither isoform prevented changes in collagen accumulation or lung compliance decline after administration of intradermal bleomycin. Together, these findings demonstrate that the p70S6K isoforms have unique and redundant functions in mediating fibrogenic processes, including proliferation, migration, and S6 phosphorylation, signifying that both isoforms must be targeted to modulate p70S6K-mediated pulmonary fibrosis.


Subject(s)
Cell Movement , Mesoderm/pathology , Pulmonary Fibrosis/enzymology , Pulmonary Fibrosis/pathology , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Animals , Bleomycin , Cell Proliferation , Collagen/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Isoenzymes/metabolism , Ki-67 Antigen/metabolism , Lung/metabolism , Lung/pathology , Lung/physiopathology , Mice, Transgenic , Phosphorylation , Pulmonary Fibrosis/physiopathology , Ribosomal Protein S6 Kinases, 70-kDa/deficiency , Signal Transduction , Transforming Growth Factor alpha/metabolism
11.
Am J Physiol Lung Cell Mol Physiol ; 310(2): L175-86, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26566903

ABSTRACT

The p70 ribosomal S6 kinase (S6K) is a downstream substrate that is phosphorylated and activated by the mammalian target of rapamycin complex and regulates multiple cellular processes associated with fibrogenesis. Recent studies demonstrate that aberrant mTORC1-S6K signaling contributes to various pathological conditions, but a direct role in pulmonary fibroproliferation has not been established. Increased phosphorylation of the S6K pathway is detected immediately following transforming growth factor-α (TGF-α) expression in a transgenic model of progressive lung fibrosis. To test the hypothesis that the S6K directly regulates pulmonary fibroproliferative disease we determined the cellular sites of S6K phosphorylation during the induction of fibrosis in the TGF-α model and tested the efficacy of specific pharmacological inhibition of the S6K pathway to prevent and reverse fibrotic disease. Following TGF-α expression increased phosphorylation of the S6K was detected in the airway and alveolar epithelium and the mesenchyme of advanced subpleural fibrotic regions. Specific inhibition of the S6K with the small molecule inhibitor LY-2584702 decreased TGF-α and platelet-derived growth factor-ß-induced proliferation of lung fibroblasts in vitro. Administration of S6K inhibitors to TGF-α mice prevented the development of extensive subpleural fibrosis and alterations in lung mechanics, and attenuated the increase in total lung hydroxyproline. S6K inhibition after fibrosis was established attenuated the progression of subpleural fibrosis. Together these studies demonstrate targeting the S6K pathway selectively modifies the progression of pulmonary fibrosis in the subpleural compartment of the lung.


Subject(s)
Lung/metabolism , Lung/pathology , Pulmonary Fibrosis/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Transforming Growth Factor alpha/metabolism , Animals , Mice, Transgenic , Phosphorylation , Platelet-Derived Growth Factor/metabolism , Pulmonary Fibrosis/pathology , Signal Transduction/drug effects , Sirolimus/pharmacology
12.
Am J Respir Cell Mol Biol ; 50(4): 777-86, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24199692

ABSTRACT

Pulmonary fibrosis is caused by excessive proliferation and accumulation of stromal cells. Fibrocytes are bone marrow (BM)-derived cells that contribute to pathologic stromal cell accumulation in human lung disease. However, the cellular source for these stromal cells and the degree of fibrocyte contribution to pulmonary fibrosis remain unclear. To determine the etiology of stromal cell excess during pulmonary fibrosis, we measured fibrocytes during the progression of fibrosis in the transforming growth factor (TGF)-α transgenic mouse model. Lung epithelial-specific overexpression of TGF-α led to progressive pulmonary fibrosis associated with increased accumulation of fibrocytes in the fibrotic lesions. Although reconstitution of BM cells into TGF-α mice demonstrated accumulation of these cells in fibrotic lesions, the majority of the cells did not express α-smooth muscle actin, suggesting that fibrocytes did not transform into myofibroblasts. To explore the mechanisms of fibrocytes in pulmonary fibrogenesis, adoptive cell-transfer experiments were performed. Purified fibrocytes were transferred intravenously into TGF-α transgenic mice, and fibrosis endpoints were compared with controls. Analysis of lung histology and hydroxyproline levels demonstrated that fibrocyte transfers augment TGF-α-induced lung fibrosis. A major subset of TGF-α-induced fibrocytes expressed CD44 and displayed excessive invasiveness, which is attenuated in the presence of anti-CD44 antibodies. Coculture experiments of resident fibroblasts with fibrocytes demonstrated that fibrocytes stimulate proliferation of resident fibroblasts. In summary, fibrocytes are increased in the progressive, fibrotic lesions of TGF-α-transgenic mice and activate resident fibroblasts to cause severe lung disease.


Subject(s)
Bone Marrow Cells/metabolism , Cell Movement , Cell Proliferation , Fibroblasts/metabolism , Lung/metabolism , Pulmonary Fibrosis/metabolism , Stromal Cells/metabolism , Transforming Growth Factor alpha/metabolism , Adoptive Transfer , Animals , Bone Marrow Cells/pathology , Bone Marrow Transplantation , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Disease Progression , Fibroblasts/pathology , Fibroblasts/transplantation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hyaluronan Receptors/metabolism , Hydroxyproline/metabolism , Lung/pathology , Mice , Mice, Transgenic , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Stromal Cells/pathology , Stromal Cells/transplantation , Time Factors , Transforming Growth Factor alpha/genetics , Up-Regulation
13.
Am J Physiol Lung Cell Mol Physiol ; 306(8): L726-35, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24508732

ABSTRACT

A number of growth factors and signaling pathways regulate matrix deposition and fibroblast proliferation in the lung. The epidermal growth factor receptor (EGFR) family of receptors and the transforming growth factor-ß (TGF-ß) family are active in diverse biological processes and are central mediators in the initiation and maintenance of fibrosis in many diseases. Transforming growth factor-α (TGF-α) is a ligand for the EGFR, and doxycycline (Dox)-inducible transgenic mice conditionally expressing TGF-α specifically in the lung epithelium develop progressive fibrosis accompanied with cachexia, changes in lung mechanics, and marked pleural thickening. Although recent studies demonstrate that EGFR activation modulates the fibroproliferative effects involved in the pathogenesis of TGF-ß induced pulmonary fibrosis, in converse, the direct role of EGFR induction of the TGF-ß pathway in the lung is unknown. The αvß6 integrin is an important in vivo activator of TGF-ß activation in the lung. Immunohistochemical analysis of αvß6 protein expression and bronchoalveolar analysis of TGF-ß pathway signaling indicates activation of the αvß6/TGF-ß pathway only at later time points after lung fibrosis was already established in the TGF-α model. To determine the contribution of the αvß6/TGF-ß pathway on the progression of established fibrotic disease, TGF-α transgenic mice were administered Dox for 4 wk, which leads to extensive fibrosis; these mice were then treated with a function-blocking anti-αvß6 antibody with continued administration of Dox for an additional 4 wk. Compared with TGF-α transgenic mice treated with control antibody, αvß6 inhibition significantly attenuated pleural thickening and altered the decline in lung mechanics. To test the effects of genetic loss of the ß6 integrin, TGF-α transgenic mice were mated with ß6-null mice and the degree of fibrosis was compared in adult mice following 8 wk of Dox administration. Genetic ablation of the ß6 integrin attenuated histological and physiological changes in the lungs of TGF-α transgenic mice although a significant degree of fibrosis still developed. In summary, inhibition of the ß6 integrin led to a modest, albeit significant, effect on pleural thickening and lung function decline observed with TGF-α-induced pulmonary fibrosis. These data support activation of the αvß6/TGF-ß pathway as a secondary effect contributing to TGF-α-induced pleural fibrosis and suggest a complex contribution of multiple mediators to the maintenance of progressive fibrosis in the lung.


Subject(s)
Integrins/antagonists & inhibitors , Pulmonary Fibrosis/pathology , Transforming Growth Factor alpha/pharmacology , Animals , Anti-Bacterial Agents/toxicity , Antibodies, Neutralizing , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Bronchoalveolar Lavage , Collagen , Doxycycline/toxicity , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoenzyme Techniques , Integrins/genetics , Integrins/metabolism , Male , Mice , Mice, Transgenic , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/pharmacology , Uteroglobin/physiology
14.
Front Surg ; 11: 1356501, 2024.
Article in English | MEDLINE | ID: mdl-38831786

ABSTRACT

Introduction: Patients with congenital heart disease (CHD) often have pulmonary abnormalities and exercise intolerance following cardiac surgery. Cardiac rehabilitation (CR) improves exercise capacity in patients with CHD, but minimal study has been performed to see if resting and dynamic pulmonary performance improves following CR in those with prior cardiac surgery. Methods: This was a retrospective cohort study of all patients who completed ≥12 weeks of CR from 2018 through 2022. Demographic, cardiopulmonary exercise test (CPET), spirometry, 6-minute walk, functional strength measures, and outcomes data were collected. Data are presented as median[IQR]. A Student's t-test was used for comparisons between groups and serial measurements were measured with a paired t-test. A p < 0.05 was considered significant. Results: There were a total of 37 patients [age 16.7 (14.2-20.1) years; 46% male] included. Patients with prior surgery (n = 26) were more likely to have abnormal spirometry data than those without heart disease (n = 11) (forced vital capacity [FVC] 76.7 [69.1-84.3]% vs. 96.4 [88.1-104.7]%, p = 0.002), but neither group experienced a significant change in spirometry. On CPET, peak oxygen consumption increased but there was no change in other pulmonary measures during exercise. Percent predicted FVC correlated with hand grip strength (r = 0.57, p = 0.0003) and percent predicted oxygen consumption (r = 0.43, p = 0.009). The number of prior sternotomies showed negative associations with both percent predicted FVC (r = -0.43, p = 0.04) and FEV1 (r = -0.47, p = 0.02). Discussion: Youth and young adults with a prior history of cardiac surgery have resting and dynamic pulmonary abnormalities that do not improve following CR. Multiple sternotomies are associated with worse pulmonary function.

15.
Matrix Biol ; 124: 1-7, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37922998

ABSTRACT

Elastin is a long-lived fibrous protein that is abundant in the extracellular matrix of the lung. Elastic fibers provide the lung the characteristic elasticity during inhalation with recoil during exhalation thereby ensuring efficient gas exchange. Excessive deposition of elastin and other extracellular matrix proteins reduces lung compliance by impairing ventilation and compromising gas exchange. Notably, the degree of elastosis is associated with the progressive decline in lung function and survival in patients with interstitial lung diseases. Currently there are no proven therapies which effectively reduce the elastin burden in the lung nor prevent dysregulated elastosis. This review describes elastin's role in the healthy lung, summarizes elastosis in pulmonary diseases, and evaluates the current understanding of elastin regulation and dysregulation with the goal of guiding future research efforts to develop novel and effective therapies.


Subject(s)
Lung Diseases, Interstitial , Lung , Humans , Lung/metabolism , Lung Diseases, Interstitial/metabolism , Fibrosis , Elastin , Elastic Tissue/metabolism
16.
J Clin Transl Sci ; 7(1): e155, 2023.
Article in English | MEDLINE | ID: mdl-37528939

ABSTRACT

Introduction: Research is an important aspect of many students' training. However, formal research training is rarely included in curricula. Thus, we developed an online, asynchronous series of modules to introduce trainees to multiple topics that are relevant to the conduct of research. Methods: Research 101 was utilized by first-year medical students and undergraduate students conducting mentored research projects. Students' knowledge, confidence, and satisfaction were assessed using pre- and post-module surveys with five-point Likert scaled questions, open-ended text responses, and a final quiz. Results: Pre-module survey results showed that learners felt most confident with the Conducting a literature search and Race and racism in medicine modules and least confident with the Submitting an Institutional Review Board protocol at UC module. Post-module survey responses were significantly increased compared to pre-module results for all modules and questions (p < 0.0001). The response to "The content of this module met my needs" was endorsed across all modules (84.9% "yes" responses). A final quiz of 25 multiple-choice questions was completed by 92 participants who received a median score of 21. Content analysis of open-ended post-module survey responses identified several strengths and opportunities for improvement in course content and instructional methods. Conclusions: These data demonstrate that significant learning resulted from completion of Research 101, as post-module survey scores were significantly higher than pre-module survey scores for all modules and questions. Final quiz scores were positive but also highlighted opportunity for additional trainee learning and will guide evolution of future modules.

17.
Am J Respir Cell Mol Biol ; 46(3): 380-8, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22021337

ABSTRACT

Pulmonary fibrosis remains a significant public health burden with no proven therapies. The mitogen-activated protein kinase (MAPK)/MAPK kinase (MEK)/extracellular signal-regulated kinase (ERK) signaling cascade is a major pathway controlling cellular processes associated with fibrogenesis, including growth, proliferation, and survival. Activation of the MAPK/ERK pathway is detected in the lungs of human fibrosis samples; however, the effect of modulating the pathway in vivo is unknown. Overexpression of transforming growth factor (TGF)-α in the lung epithelium of transgenic mice causes a progressive pulmonary fibrosis associated with increased MEK/ERK activation localized primarily in mesenchymal cells. To determine the role of the MEK pathway in the induction of TGF-α-induced lung fibrosis, TGF-α was overexpressed for 4 weeks while mice were simultaneously treated with the specific MEK inhibitor, ARRY-142886 (ARRY). Treatment with ARRY prevented increases in lung cell proliferation and total lung collagen, attenuated production of extracellular matrix genes, and protected mice from changes in lung function. ARRY administered as a rescue treatment after fibrosis was already established inhibited fibrosis progression, as assessed by lung histology, changes in body weights, extracellular matrix gene expression, and lung mechanics. These findings demonstrate that MEK inhibition prevents progression of established fibrosis in the TGF-α model, and provides proof of concept of targeting the MEK pathway in fibrotic lung disease.


Subject(s)
Benzimidazoles/pharmacology , ErbB Receptors/metabolism , Lung/drug effects , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pulmonary Fibrosis/prevention & control , Animals , Cell Proliferation/drug effects , Cells, Cultured , Collagen/genetics , Collagen/metabolism , Disease Models, Animal , Enzyme Activation , Fibroblasts/drug effects , Fibroblasts/enzymology , Gene Expression Regulation , Humans , Lung/enzymology , Lung/pathology , Lung/physiopathology , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphorylation , Pulmonary Fibrosis/enzymology , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/physiopathology , Time Factors , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
18.
Respir Res ; 13: 51, 2012 Jun 22.
Article in English | MEDLINE | ID: mdl-22726462

ABSTRACT

BACKGROUND: Resistin-like molecule alpha or found in inflammatory zone protein (Fizz1) is increased in pulmonary epithelial cells and also in limited amounts by other lung cells during various lung injuries and fibrosis. However, the direct role of Fizz1 produced in the pulmonary epithelium has not been determined. METHODS: Fizz1 Transgenic mice (CCSP/Fizz1) were generated that overexpress Fizz1 in the lung epithelium under the control of a doxycycline (Dox) inducible lung epithelial cell specific promoter Scgb1a1 (Clara cell secretory protein, CCSP). Histology and FACS analysis of lung cells were used to identify the direct effects of Fizz1 in the transgenic mice (Dox treated) when compared with control (CCSP/-) mice. Intratracheal bleomycin sulfate or silica in saline and saline alone were used to study the role of Fizz1 during bleomycin- and silica-induced pulmonary fibrosis in CCSP/Fizz1 and CCSP/- mice. Weight change, pulmonary inflammation, and fibrosis were assessed 10 days post bleomycin or 28 days post silica challenge. RESULTS: When CCSP/Fizz1 mice were fed Dox food, elevated Fizz1 protein was detected in lung homogenates by western blot. Lungs of mice in which Fizz1 was induced in the epithelium contained increased lung cells staining for CD11c and F4/80 by FACS analysis consistent with increased dendritic cells however, no changes were observed in the percentage of interstitial macrophages compared to CCSP/- controls. No significant changes were found in the lung histology of CCSP/Fizz1 mice after up to 8 weeks of overexpression compared to CCSP/- controls. Overexpression of Fizz1 prior to challenge or following challenge with bleomycin or silica did not significantly alter airway inflammation or fibrosis compared to control mice. CONCLUSIONS: The current study demonstrates that epithelial cell derived Fizz1 is sufficient to increase the bone-marrow derived dendritic cells in the lungs, but it is not sufficient to cause lung fibrosis or alter chemical or particle-induced fibrosis.


Subject(s)
Cell Movement/physiology , Dendritic Cells/metabolism , Intercellular Signaling Peptides and Proteins/physiology , Lung/metabolism , Lung/pathology , Pulmonary Fibrosis , Animals , Dendritic Cells/pathology , Female , Mice , Mice, Transgenic , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/pathology
19.
J Clin Transl Sci ; 6(1): e102, 2022.
Article in English | MEDLINE | ID: mdl-36106129

ABSTRACT

Introduction: Research is an important aspect of many medical students' training. However, many medical students are not required to complete a scholarly project, and formal research training is often fragmented across the medical school curriculum. Thus, we developed an online, structured, asynchronous set of modules to introduce trainees to multiple topics relevant to the conduct of research. Methods: Research 101 was piloted by 27 first-year medical students at the University of Cincinnati College of Medicine. Students' knowledge, confidence, and satisfaction were assessed using a final quiz and pre- and post-module surveys with five-point Likert-scaled questions and open-ended text responses. Results: Pre-module survey results showed that learners felt most confident in Conducting a literature search and least confident in Submitting an Institutional Review Board (IRB) protocol at UC. Post-module mean scores were significantly increased compared to pre-module results for all modules and questions (P < 0.05). The response to "The content of this module met my needs" was high across all modules with 236 (84.0%) "yes" responses. Thematic analysis of open-ended text responses from post-module surveys identified several improvements to individual modules and to the overall structure of Research 101. A final quiz of 25 multiple choice questions covering content from all required modules was required. The median score was 21. Conclusions: Comparison of post-module to pre-module survey scores provided clear evidence of improved learning across all topics. The modules developed were responsive to the students' needs, and students provided additional improvements for subsequent iterations of Research 101.

20.
Am J Physiol Lung Cell Mol Physiol ; 300(3): L414-21, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21224214

ABSTRACT

Increases in the epidermal growth factor receptor (EGFR) have been associated with the severity of airway thickening in chronic asthmatic subjects, and EGFR signaling is induced by asthma-related cytokines and inflammation. The goal of this study was to determine the role of EGFR signaling in a chronic allergic model of asthma and specifically in epithelial cells, which are increasingly recognized as playing an important role in asthma. EGFR activation was assessed in mice treated with intranasal house dust mite (HDM) for 3 wk. EGFR signaling was inhibited in mice treated with HDM for 6 wk, by using either the drug erlotinib or a genetic approach that utilizes transgenic mice expressing a mutant dominant negative epidermal growth factor receptor in the lung epithelium (EGFR-M mice). Airway hyperreactivity (AHR) was assessed by use of a flexiVent system after increasing doses of nebulized methacholine. Airway smooth muscle (ASM) thickening was measured by morphometric analysis. Sensitization to HDM (IgG and IgE), inflammatory cells, and goblet cell changes were also assessed. Increased EGFR activation was detected in HDM-treated mice, including in bronchiolar epithelial cells. In mice exposed to HDM for 6 wk, AHR and ASM thickening were reduced after erlotinib treatment and in EGFR-M mice. Sensitization to HDM and inflammatory cell counts were similar in all groups, except neutrophil counts, which were lower in the EGFR-M mice. Goblet cell metaplasia with HDM treatment was reduced by erlotinib, but not in EGFR-M transgenic mice. This study demonstrates that EGFR signaling, especially in the airway epithelium, plays an important role in mediating AHR and remodeling in a chronic allergic asthma model.


Subject(s)
Airway Remodeling/physiology , Asthma/physiopathology , Bronchial Hyperreactivity/complications , Epithelial Cells/enzymology , ErbB Receptors/metabolism , Signal Transduction , Animals , Asthma/complications , Asthma/parasitology , Asthma/pathology , Bronchial Hyperreactivity/parasitology , Bronchial Hyperreactivity/pathology , Bronchial Hyperreactivity/physiopathology , Chronic Disease , Disease Models, Animal , Enzyme Activation , Epithelial Cells/pathology , ErbB Receptors/antagonists & inhibitors , Goblet Cells/pathology , Inflammation/complications , Inflammation/pathology , Lung/parasitology , Lung/pathology , Lung/physiopathology , Metaplasia , Mice , Muscle, Smooth/pathology , Pyroglyphidae/physiology
SELECTION OF CITATIONS
SEARCH DETAIL