Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
JCI Insight ; 8(14)2023 07 24.
Article in English | MEDLINE | ID: mdl-37279065

ABSTRACT

During alveolar repair, alveolar type 2 (AT2) epithelial cell progenitors rapidly proliferate and differentiate into flat AT1 epithelial cells. Failure of normal alveolar repair mechanisms can lead to loss of alveolar structure (emphysema) or development of fibrosis, depending on the type and severity of injury. To test if ß1-containing integrins are required during repair following acute injury, we administered E. coli lipopolysaccharide (LPS) by intratracheal injection to mice with a postdevelopmental deletion of ß1 integrin in AT2 cells. While control mice recovered from LPS injury without structural abnormalities, ß1-deficient mice had more severe inflammation and developed emphysema. In addition, recovering alveoli were repopulated with an abundance of rounded epithelial cells coexpressing AT2 epithelial, AT1 epithelial, and mixed intermediate cell state markers, with few mature type 1 cells. AT2 cells deficient in ß1 showed persistently increased proliferation after injury, which was blocked by inhibiting NF-κB activation in these cells. Lineage tracing experiments revealed that ß1-deficient AT2 cells failed to differentiate into mature AT1 epithelial cells. Together, these findings demonstrate that functional alveolar repair after injury with terminal alveolar epithelial differentiation requires ß1-containing integrins.


Subject(s)
Emphysema , Lipopolysaccharides , Mice , Animals , Lipopolysaccharides/toxicity , Escherichia coli , Lung , Integrins
2.
Am J Respir Crit Care Med ; 206(5): 596-607, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35728047

ABSTRACT

Rationale: Although persistent fibroblast activation is a hallmark of idiopathic pulmonary fibrosis (IPF), mechanisms regulating persistent fibroblast activation in the lungs have not been fully elucidated. Objectives: On the basis of our observation that lung fibroblasts express TBXA2R (thromboxane-prostanoid receptor) during fibrosis, we investigated the role of TBXA2R signaling in fibrotic remodeling. Methods: We identified TBXA2R expression in lungs of patients with IPF and mice and studied primary mouse and human lung fibroblasts to determine the impact of TBXA2R signaling on fibroblast activation. We used TBXA2R-deficient mice and small-molecule inhibitors to investigate TBXA2R signaling in preclinical lung fibrosis models. Measurements and Main Results: TBXA2R expression was upregulated in fibroblasts in the lungs of patients with IPF and in mouse lungs during experimental lung fibrosis. Genetic deletion of TBXA2R, but not inhibition of thromboxane synthase, protected mice from bleomycin-induced lung fibrosis, thereby suggesting that an alternative ligand activates profibrotic TBXA2R signaling. In contrast to thromboxane, F2-isoprostanes, which are nonenzymatic products of arachidonic acid induced by reactive oxygen species, were persistently elevated during fibrosis. F2-isoprostanes induced TBXA2R signaling in fibroblasts and mediated a myofibroblast activation profile due, at least in part, to potentiation of TGF-ß (transforming growth factor-ß) signaling. In vivo treatment with the TBXA2R antagonist ifetroban reduced profibrotic signaling in the lungs, protected mice from lung fibrosis in three preclinical models (bleomycin, Hermansky-Pudlak mice, and radiation-induced fibrosis), and markedly enhanced fibrotic resolution after bleomycin treatment. Conclusions: TBXA2R links oxidative stress to fibroblast activation during lung fibrosis. TBXA2R antagonists could have utility in treating pulmonary fibrosis.


Subject(s)
Idiopathic Pulmonary Fibrosis , Receptors, Thromboxane , Animals , Bleomycin/pharmacology , F2-Isoprostanes/metabolism , Fibroblasts/metabolism , Humans , Idiopathic Pulmonary Fibrosis/genetics , Lung/metabolism , Mice , Mice, Inbred C57BL , Prostaglandins/metabolism , Receptors, Thromboxane/metabolism , Thromboxanes/metabolism , Transforming Growth Factor beta/metabolism
3.
Dev Cell ; 57(7): 839-853.e6, 2022 04 11.
Article in English | MEDLINE | ID: mdl-35303432

ABSTRACT

Although increased neuropeptides are often detected in lungs that exhibit respiratory distress, whether they contribute to the condition is unknown. Here, we show in a mouse model of neuroendocrine cell hyperplasia of infancy, a pediatric disease with increased pulmonary neuroendocrine cells (PNECs), excess PNEC-derived neuropeptides are responsible for pulmonary manifestations including hypoxemia. In mouse postnatal lung, prolonged signaling from elevated neuropeptides such as calcitonin gene-related peptide (CGRP) activate receptors enriched on endothelial cells, leading to reduced cellular junction gene expression, increased endothelium permeability, excess lung fluid, and hypoxemia. Excess fluid and hypoxemia were effectively attenuated by either prevention of PNEC formation, inactivation of CGRP gene, endothelium-specific inactivation of CGRP receptor gene, or treatment with CGRP receptor antagonist. Neuropeptides were increased in human lung diseases with excess fluid such as acute respiratory distress syndrome. Our findings suggest that restricting neuropeptide function may limit fluid and improve gas exchange in these conditions.


Subject(s)
Calcitonin Gene-Related Peptide , Neuropeptides , Animals , Calcitonin Gene-Related Peptide/metabolism , Endothelial Cells/metabolism , Humans , Hypoxia/metabolism , Lung/metabolism , Mice , Neuropeptides/metabolism
4.
Development ; 148(24)2021 12 15.
Article in English | MEDLINE | ID: mdl-34927678

ABSTRACT

Lung organogenesis requires precise timing and coordination to effect spatial organization and function of the parenchymal cells. To provide a systematic broad-based view of the mechanisms governing the dynamic alterations in parenchymal cells over crucial periods of development, we performed a single-cell RNA-sequencing time-series yielding 102,571 epithelial, endothelial and mesenchymal cells across nine time points from embryonic day 12 to postnatal day 14 in mice. Combining computational fate-likelihood prediction with RNA in situ hybridization and immunofluorescence, we explore lineage relationships during the saccular to alveolar stage transition. The utility of this publicly searchable atlas resource (www.sucrelab.org/lungcells) is exemplified by discoveries of the complexity of type 1 pneumocyte function and characterization of mesenchymal Wnt expression patterns during the saccular and alveolar stages - wherein major expansion of the gas-exchange surface occurs. We provide an integrated view of cellular dynamics in epithelial, endothelial and mesenchymal cell populations during lung organogenesis.


Subject(s)
Embryonic Development/genetics , Lung/growth & development , Mesenchymal Stem Cells/cytology , Organogenesis/genetics , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Embryo, Mammalian/ultrastructure , Epithelial Cells/cytology , Epithelial Cells/ultrastructure , Gene Expression Regulation, Developmental/genetics , Lung/ultrastructure , Mesenchymal Stem Cells/ultrastructure , Mice , RNA-Seq , Single-Cell Analysis , Transcriptome/genetics
5.
J Clin Invest ; 131(1)2021 01 04.
Article in English | MEDLINE | ID: mdl-33180746

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) novel coronavirus 2019 (COVID-19) global pandemic has led to millions of cases and hundreds of thousands of deaths. While older adults appear at high risk for severe disease, hospitalizations and deaths due to SARS-CoV-2 among children have been relatively rare. Integrating single-cell RNA sequencing (scRNA-seq) of developing mouse lung with temporally resolved immunofluorescence in mouse and human lung tissue, we found that expression of SARS-CoV-2 Spike protein primer TMPRSS2 was highest in ciliated cells and type I alveolar epithelial cells (AT1), and TMPRSS2 expression increased with aging in mice and humans. Analysis of autopsy tissue from fatal COVID-19 cases detected SARS-CoV-2 RNA most frequently in ciliated and secretory cells in airway epithelium and AT1 cells in peripheral lung. SARS-CoV-2 RNA was highly colocalized in cells expressing TMPRSS2. Together, these data demonstrate the cellular spectrum infected by SARS-CoV-2 in lung epithelium and suggest that developmental regulation of TMPRSS2 may underlie the relative protection of infants and children from severe respiratory illness.


Subject(s)
Alveolar Epithelial Cells/enzymology , COVID-19/enzymology , COVID-19/metabolism , Gene Expression Regulation, Enzymologic , SARS-CoV-2/metabolism , Serine Endopeptidases/biosynthesis , Adult , Aging , Alveolar Epithelial Cells/pathology , Alveolar Epithelial Cells/virology , Animals , COVID-19/pathology , Child, Preschool , Disease Models, Animal , Female , Humans , Infant , Male , Mice
6.
J Clin Invest ; 131(1)2021 01 04.
Article in English | MEDLINE | ID: mdl-33108351

ABSTRACT

Emerging evidence indicates that early life events can increase the risk for developing chronic obstructive pulmonary disease (COPD). Using an inducible transgenic mouse model for NF-κB activation in the airway epithelium, we found that a brief period of inflammation during the saccular stage (P3-P5) but not alveolar stage (P10-P12) of lung development disrupted elastic fiber assembly, resulting in permanent reduction in lung function and development of a COPD-like lung phenotype that progressed through 24 months of age. Neutrophil depletion prevented disruption of elastic fiber assembly and restored normal lung development. Mechanistic studies uncovered a role for neutrophil elastase (NE) in downregulating expression of critical elastic fiber assembly components, particularly fibulin-5 and elastin. Further, purified human NE and NE-containing exosomes from tracheal aspirates of premature infants with lung inflammation downregulated elastin and fibulin-5 expression by saccular-stage mouse lung fibroblasts. Together, our studies define a critical developmental window for assembling the elastin scaffold in the distal lung, which is required to support lung structure and function throughout the lifespan. Although neutrophils play a well-recognized role in COPD development in adults, neutrophilic inflammation may also contribute to early-life predisposition to COPD.


Subject(s)
Elastin/metabolism , Neutrophils/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Animals , Elastin/genetics , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Leukocyte Elastase/genetics , Leukocyte Elastase/metabolism , Mice , Mice, Transgenic , Neutrophils/pathology , Pulmonary Alveoli/pathology , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/pathology
7.
bioRxiv ; 2020 Aug 03.
Article in English | MEDLINE | ID: mdl-32511364

ABSTRACT

The SARS-CoV-2 novel coronavirus global pandemic (COVID-19) has led to millions of cases and hundreds of thousands of deaths around the globe. While the elderly appear at high risk for severe disease, hospitalizations and deaths due to SARS-CoV-2 among children have been relatively rare. Integrating single-cell RNA sequencing (scRNA-seq) of the developing mouse lung with temporally-resolved RNA-in-situ hybridization (ISH) in mouse and human lung tissue, we found that expression of SARS-CoV-2 Spike protein primer TMPRSS2 was highest in ciliated cells and type I alveolar epithelial cells (AT1), and TMPRSS2 expression was increased with aging in mice and humans. Analysis of autopsy tissue from fatal COVID-19 cases revealed SARS-CoV-2 RNA was detected most frequently in ciliated and secretory cells in the airway epithelium and AT1 cells in the peripheral lung. SARS-CoV-2 RNA was highly colocalized in cells expressing TMPRSS2. Together, these data demonstrate the cellular spectrum infected by SARS-CoV-2 in the lung epithelium, and suggest that developmental regulation of TMPRSS2 may underlie the relative protection of infants and children from severe respiratory illness.

8.
JCI Insight ; 5(2)2020 01 30.
Article in English | MEDLINE | ID: mdl-31873073

ABSTRACT

Integrins, the extracellular matrix receptors that facilitate cell adhesion and migration, are necessary for organ morphogenesis; however, their role in maintaining adult tissue homeostasis is poorly understood. To define the functional importance of ß1 integrin in adult mouse lung, we deleted it after completion of development in type 2 alveolar epithelial cells (AECs). Aged ß1 integrin-deficient mice exhibited chronic obstructive pulmonary disease-like (COPD-like) pathology characterized by emphysema, lymphoid aggregates, and increased macrophage infiltration. These histopathological abnormalities were preceded by ß1 integrin-deficient AEC dysfunction such as excessive ROS production and upregulation of NF-κB-dependent chemokines, including CCL2. Genetic deletion of the CCL2 receptor, Ccr2, in mice with ß1 integrin-deficient type 2 AECs impaired recruitment of monocyte-derived macrophages and resulted in accelerated inflammation and severe premature emphysematous destruction. The lungs exhibited reduced AEC efferocytosis and excessive numbers of inflamed type 2 AECs, demonstrating the requirement for recruited monocytes/macrophages in limiting lung injury and remodeling in the setting of a chronically inflamed epithelium. These studies support a critical role for ß1 integrin in alveolar homeostasis in the adult lung.


Subject(s)
Alveolar Epithelial Cells/metabolism , Integrin beta1/genetics , Integrin beta1/metabolism , Pneumonia/metabolism , Aging/metabolism , Alveolar Epithelial Cells/pathology , Animals , Cell Adhesion , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokines/genetics , Chemokines/metabolism , Disease Models, Animal , Epithelium , Lung/pathology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia/pathology , Pulmonary Disease, Chronic Obstructive/metabolism , Receptors, CCR2/genetics
10.
Am J Respir Cell Mol Biol ; 58(5): 566-574, 2018 05.
Article in English | MEDLINE | ID: mdl-29190429

ABSTRACT

Defining the mechanisms of cellular pathogenesis in rare lung diseases such as Hermansky-Pudlak syndrome (HPS) is often complicated by loss of the differentiated phenotype of cultured primary alveolar type 2 (AT2) cells, as well as by a lack of durable cell lines that are faithful to both AT2-cell and rare disease phenotypes. We used CRISPR/Cas9 gene editing to generate a series of HPS-specific mutations in the MLE-15 cell line. The resulting MLE-15/HPS cell lines exhibit preservation of AT2 cellular functions, including formation of lamellar body-like organelles, complete processing of surfactant protein B, and known features of HPS specific to each trafficking complex, including loss of protein targeting to lamellar bodies. MLE-15/HPS1 and MLE-15/HPS2 (with a mutation in Ap3ß1) express increased macrophage chemotactic protein-1, a well-described mediator of alveolitis in patients with HPS and in mouse models. We show that MLE-15/HPS9 and pallid AT2 cells (with a mutation in Bloc1s6) also express increased macrophage chemotactic protein-1, suggesting that mice and humans with BLOC-1 mutations may also be susceptible to alveolitis. In addition to providing a flexible platform to examine the role of HPS-specific mutations in trafficking AT2 cells, MLE-15/HPS cell lines provide a durable resource for high-throughput screening and studies of cellular pathophysiology that are likely to accelerate progress toward developing novel therapies for this rare lung disease.


Subject(s)
Alveolar Epithelial Cells/metabolism , CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Editing/methods , Hermanski-Pudlak Syndrome/genetics , Mutation , Alveolar Epithelial Cells/pathology , Animals , CRISPR-Associated Protein 9/metabolism , Cell Line , Disease Models, Animal , Genetic Markers , Genetic Predisposition to Disease , Hermanski-Pudlak Syndrome/metabolism , Hermanski-Pudlak Syndrome/pathology , Humans , Mice, Inbred C57BL , Mice, Transgenic , Phenotype
11.
JCI Insight ; 1(17): e88947, 2016 Oct 20.
Article in English | MEDLINE | ID: mdl-27777976

ABSTRACT

Alveolar epithelial cell (AEC) dysfunction underlies the pathogenesis of pulmonary fibrosis in Hermansky-Pudlak syndrome (HPS) and other genetic syndromes associated with interstitial lung disease; however, mechanisms linking AEC dysfunction and fibrotic remodeling are incompletely understood. Since increased macrophage recruitment precedes pulmonary fibrosis in HPS, we investigated whether crosstalk between AECs and macrophages determines fibrotic susceptibility. We found that AECs from HPS mice produce excessive MCP-1, which was associated with increased macrophages in the lungs of unchallenged HPS mice. Blocking MCP-1/CCR2 signaling in HPS mice with genetic deficiency of CCR2 or targeted deletion of MCP-1 in AECs normalized macrophage recruitment, decreased AEC apoptosis, and reduced lung fibrosis in these mice following treatment with low-dose bleomycin. We observed increased TGF-ß production by HPS macrophages, which was eliminated by CCR2 deletion. Selective deletion of TGF-ß in myeloid cells or of TGF-ß signaling in AECs through deletion of TGFBR2 protected HPS mice from AEC apoptosis and bleomycin-induced fibrosis. Together, these data reveal a feedback loop in which increased MCP-1 production by dysfunctional AECs results in recruitment and activation of lung macrophages that produce TGF-ß, thus amplifying the fibrotic cascade through AEC apoptosis and stimulation of fibrotic remodeling.


Subject(s)
Epithelial Cells/cytology , Hermanski-Pudlak Syndrome/immunology , Macrophages/cytology , Pulmonary Fibrosis/immunology , Animals , Bleomycin , Chemokine CCL2/metabolism , Disease Susceptibility , Female , Male , Mice , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/metabolism , Pulmonary Alveoli/cytology , Receptor, Transforming Growth Factor-beta Type II , Receptors, CCR2/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/metabolism
12.
Oncoimmunology ; 5(6): e1168549, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27471643

ABSTRACT

Several studies have demonstrated that NF-κB activation is common in lung cancer; however, the mechanistic links between NF-κB signaling and tumorigenesis remain to be fully elucidated. We investigated the function of NF-κB signaling in epidermal growth factor receptor (EGFR)-mutant lung tumors using a transgenic mouse model with doxycycline (dox)-inducible expression of oncogenic EGFR in the lung epithelium with or without a dominant inhibitor of NF-κB signaling. NF-κB inhibition resulted in a significant reduction in tumor burden in both EGFR tyrosine kinase inhibitor (TKI)-sensitive and resistant tumors. However, NF-κB inhibition did not alter epithelial cell survival in vitro or in vivo, and no changes were detected in activation of EGFR downstream signaling pathways. Instead, we observed an influx of inflammatory cells (macrophages and neutrophils) in the lungs of mice with oncogenic EGFR expression that was blocked in the setting of NF-κB inhibition. To investigate whether inflammatory cells play a role in promoting EGFR-mutant lung tumors, we depleted macrophages and neutrophils during tumorigenesis and found that neutrophil depletion had no effect on tumor formation, but macrophage depletion caused a significant reduction in tumor burden. Together, these data suggest that epithelial NF-κB signaling supports carcinogenesis in a non-cell autonomous manner in EGFR-mutant tumors through recruitment of pro-tumorigenic macrophages.

13.
Oncotarget ; 7(5): 5470-82, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26756215

ABSTRACT

Nuclear Factor (NF)-κB is positioned to provide the interface between COPD and carcinogenesis through regulation of chronic inflammation in the lungs. Using a tetracycline-inducible transgenic mouse model that conditionally expresses activated IκB kinase ß (IKKß) in airway epithelium (IKTA), we found that sustained NF-κB signaling results in chronic inflammation and emphysema by 4 months. By 11 months of transgene activation, IKTA mice develop lung adenomas. Investigation of lung inflammation in IKTA mice revealed a substantial increase in M2-polarized macrophages and CD4+/CD25+/FoxP3+ regulatory T lymphocytes (Tregs). Depletion of alveolar macrophages in IKTA mice reduced Tregs, increased lung CD8+ lymphocytes, and reduced tumor numbers following treatment with the carcinogen urethane. Alveolar macrophages from IKTA mice supported increased generation of inducible Foxp3+ Tregs ex vivo through expression of TGFß and IL-10. Targeting of TGFß and IL-10 reduced the ability of alveolar macrophages from IKTA mice to induce Foxp3 expression on T cells. These studies indicate that sustained activation of NF-κB pathway links COPD and lung cancer through generation and maintenance of a pro-tumorigenic inflammatory environment consisting of alternatively activated macrophages and regulatory T cells.


Subject(s)
Epithelium/immunology , Inflammation/immunology , Lung Neoplasms/immunology , Lung/immunology , Macrophages, Alveolar/immunology , NF-kappa B/metabolism , Pulmonary Disease, Chronic Obstructive/immunology , Animals , Blotting, Western , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Female , Flow Cytometry , Humans , I-kappa B Kinase/physiology , Immunosuppressive Agents/immunology , Interleukin-10/genetics , Interleukin-10/metabolism , Lung/metabolism , Lung/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice , Mice, Transgenic , NF-kappa B/genetics , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
14.
Development ; 141(24): 4751-62, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25395457

ABSTRACT

Integrin-dependent interactions between cells and extracellular matrix regulate lung development; however, specific roles for ß1-containing integrins in individual cell types, including epithelial cells, remain incompletely understood. In this study, the functional importance of ß1 integrin in lung epithelium during mouse lung development was investigated by deleting the integrin from E10.5 onwards using surfactant protein C promoter-driven Cre. These mutant mice appeared normal at birth but failed to gain weight appropriately and died by 4 months of age with severe hypoxemia. Defects in airway branching morphogenesis in association with impaired epithelial cell adhesion and migration, as well as alveolarization defects and persistent macrophage-mediated inflammation were identified. Using an inducible system to delete ß1 integrin after completion of airway branching, we showed that alveolarization defects, characterized by disrupted secondary septation, abnormal alveolar epithelial cell differentiation, excessive collagen I and elastin deposition, and hypercellularity of the mesenchyme occurred independently of airway branching defects. By depleting macrophages using liposomal clodronate, we found that alveolarization defects were secondary to persistent alveolar inflammation. ß1 integrin-deficient alveolar epithelial cells produced excessive monocyte chemoattractant protein 1 and reactive oxygen species, suggesting a direct role for ß1 integrin in regulating alveolar homeostasis. Taken together, these studies define distinct functions of epithelial ß1 integrin during both early and late lung development that affect airway branching morphogenesis, epithelial cell differentiation, alveolar septation and regulation of alveolar homeostasis.


Subject(s)
Epithelial Cells/metabolism , Integrin beta1/metabolism , Lung/embryology , Organogenesis/physiology , Pulmonary Alveoli/embryology , Animals , Bronchoalveolar Lavage , Cell Adhesion/physiology , Cell Movement/physiology , Chemokine CCL2/metabolism , Enzyme-Linked Immunosorbent Assay , Extracellular Matrix/metabolism , Integrases/metabolism , Mice , Microscopy, Confocal , Pulmonary Surfactant-Associated Protein C/metabolism , Reactive Oxygen Species/metabolism , Thiobarbituric Acid Reactive Substances
15.
Am J Respir Crit Care Med ; 186(10): 1014-24, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23043085

ABSTRACT

RATIONALE: Hermansky-Pudlak syndrome (HPS) is a family of recessive disorders of intracellular trafficking defects that are associated with highly penetrant pulmonary fibrosis. Naturally occurring HPS mice reliably model important features of the human disease, including constitutive alveolar macrophage activation and susceptibility to profibrotic stimuli. OBJECTIVES: To decipher which cell lineage(s) in the alveolar compartment is the predominant driver of fibrotic susceptibility in HPS. METHODS: We used five different HPS and Chediak-Higashi mouse models to evaluate genotype-specific fibrotic susceptibility. To determine whether intrinsic defects in HPS alveolar macrophages cause fibrotic susceptibility, we generated bone marrow chimeras in HPS and wild-type mice. To directly test the contribution of the pulmonary epithelium, we developed a transgenic model with epithelial-specific correction of the HPS2 defect in an HPS mouse model. MEASUREMENTS AND MAIN RESULTS: Bone marrow transplantation experiments demonstrated that both constitutive alveolar macrophage activation and increased susceptibility to bleomycin-induced fibrosis were conferred by the genotype of the lung epithelium, rather than that of the bone marrow-derived, cellular compartment. Furthermore, transgenic epithelial-specific correction of the HPS defect significantly attenuated bleomycin-induced alveolar epithelial apoptosis, fibrotic susceptibility, and macrophage activation. Type II cell apoptosis was genotype specific, caspase dependent, and correlated with the degree of fibrotic susceptibility. CONCLUSIONS: We conclude that pulmonary fibrosis in naturally occurring HPS mice is driven by intracellular trafficking defects that lower the threshold for pulmonary epithelial apoptosis. Our findings demonstrate a pivotal role for the alveolar epithelium in the maintenance of alveolar homeostasis and regulation of alveolar macrophage activation.


Subject(s)
Genetic Predisposition to Disease , Hermanski-Pudlak Syndrome/genetics , Pulmonary Alveoli/physiopathology , Pulmonary Fibrosis/physiopathology , Respiratory Mucosa/physiopathology , Adaptor Protein Complex 3/genetics , Animals , Bleomycin/pharmacology , Hermanski-Pudlak Syndrome/complications , Hermanski-Pudlak Syndrome/physiopathology , Humans , Lung/pathology , Macrophage Activation , Macrophages, Alveolar/physiology , Mice , Mice, Knockout , Mutation , Protein Transport , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/complications , Pulmonary Fibrosis/pathology
16.
Chest ; 138(3): 674-81, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20382711

ABSTRACT

OBJECTIVES: The majority of women with lymphangioleiomyomatosis (LAM) present with cystic lung disease, and most require lung biopsy for definitive diagnosis. The purpose of this study was to determine the prospective diagnostic usefulness of a serologic test for vascular endothelial growth factor-D (VEGF-D), a lymphangiogenic growth factor. METHODS: We prospectively measured serum VEGF-D levels by enzyme-linked immunoassay in 48 women presenting with cystic lung disease. Diagnostic test performance was determined from a cohort of 195 women, with tuberous sclerosis complex (TSC), TSC-LAM, sporadic LAM (S-LAM), and other cystic lung diseases in the differential diagnosis, including biopsy-proven or genetically proven pulmonary Langerhans cell histiocytosis, emphysema, Sjögren syndrome, or Birt-Hogg-Dubé syndrome. RESULTS: Serum VEGF-D levels were significantly greater in S-LAM (median 1,175 [interquartile range (IQR): 780-2,013] pg/mL; n = 56) than in other cystic lung diseases (median 281 [IQR 203-351] pg/mL; n = 44, P < .001). In the cohort evaluated prospectively, 12 of the 15 individuals ultimately diagnosed with LAM by biopsy had VEGF-D levels of > 800 pg/mL, whereas levels were < 600 pg/mL in all 18 subjects later diagnosed with other causes of cystic lung disease. Receiver operating characteristic curves demonstrated that VEGF-D effectively identified LAM, with an area under the curve of 0.961(95% CI, 0.923-0.992). A VEGF-D level of > 600 pg/mL was highly associated with a diagnosis of LAM (specificity 97.6%, likelihood ratio 35.2) and values > 800 pg/mL were diagnostically specific. Serum VEGF-D levels were significantly elevated in women with TSC-LAM (median 3,465 [IQR 1,970-7,195] pg/mL) compared with women with TSC only (median 370 [IQR 291-520] pg/mL), P < .001). CONCLUSIONS: A serum VEGF-D level of > 800 pg/mL in a woman with typical cystic changes on high-resolution CT (HRCT) scan is diagnostically specific for S-LAM and identifies LAM in women with TSC. A negative VEGF-D result does not exclude the diagnosis of LAM. The usefulness of serum VEGF-D testing in men or in women who do not have cystic lung disease on HRCT scan is unknown.


Subject(s)
Lung Diseases/blood , Lung Diseases/pathology , Lymphangioleiomyomatosis/blood , Lymphangioleiomyomatosis/diagnosis , Vascular Endothelial Growth Factor D/blood , Adolescent , Adult , Biomarkers, Tumor/blood , Diagnosis, Differential , Female , Humans , Middle Aged , Predictive Value of Tests , Prospective Studies , ROC Curve , Reproducibility of Results , Young Adult
17.
Am J Respir Cell Mol Biol ; 37(1): 67-74, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17363777

ABSTRACT

Pulmonary inflammation, abnormalities in type II cell and macrophage morphology, and pulmonary fibrosis are features of Hermansky-Pudlak Syndrome (HPS), a recessive disorder associated with intracellular trafficking defects. We have previously reported that "Pearl" (HPS2) and "Pale Ear" (HPS1) mouse models have pulmonary inflammatory dysregulation and constitutive alveolar macrophage (AM) activation (Young LR et al., J Immunol 2006;176:4361-4368). In the current study, we used these HPS models to investigate mechanisms of lung fibrosis. Unchallenged HPS1 and HPS2 mice have subtle airspace enlargement and foamy AMs, but little or no histologic evidence of lung fibrosis. Seven days after intratracheal bleomycin (0.025 units), HPS1 and HPS2 mice exhibited increased mortality and diffuse pulmonary fibrosis compared to strain-matched C57BL/6J wild-type (WT) mice. HPS mice had significantly increased collagen deposition, and reduced quasi-static and static compliance consistent with a restrictive defect. The early airway and parenchymal cellular inflammatory responses to bleomycin were similar in HPS2 and WT mice. Greater elevations in levels of TGF-beta and IL-12p40 were produced in the lungs and AMs from bleomycin-challenged HPS mice than in WT mice. TUNEL staining revealed apoptosis of type II cells as early as 5 h after low-dose bleomycin challenge in HPS mice, suggesting that type II cell susceptibility to apoptosis may play a role in the fibrotic response. We conclude that the trafficking abnormalities in HPS promote alveolar apoptosis and pulmonary fibrosis in response to bleomycin challenge.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Apoptosis , Bleomycin/pharmacology , Fibrosis , Hermanski-Pudlak Syndrome/drug therapy , Hermanski-Pudlak Syndrome/genetics , Adaptor Protein Complex 3/metabolism , Animals , Collagen/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Inflammation , Lung/pathology , Macrophages, Alveolar/metabolism , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...