Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Development ; 151(8)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38602485

ABSTRACT

Alveologenesis, the final stage in lung development, substantially remodels the distal lung, expanding the alveolar surface area for efficient gas exchange. Secondary crest myofibroblasts (SCMF) exist transiently in the neonatal distal lung and are crucial for alveologenesis. However, the pathways that regulate SCMF function, proliferation and temporal identity remain poorly understood. To address this, we purified SCMFs from reporter mice, performed bulk RNA-seq and found dynamic changes in Hippo-signaling components during alveologenesis. We deleted the Hippo effectors Yap/Taz from Acta2-expressing cells at the onset of alveologenesis, causing a significant arrest in alveolar development. Using single cell RNA-seq, we identified a distinct cluster of cells in mutant lungs with altered expression of marker genes associated with proximal mesenchymal cell types, airway smooth muscle and alveolar duct myofibroblasts. In vitro studies confirmed that Yap/Taz regulates myofibroblast-associated gene signature and contractility. Together, our findings show that Yap/Taz is essential for maintaining functional myofibroblast identity during postnatal alveologenesis.


Subject(s)
Cell Differentiation , Hippo Signaling Pathway , Morphogenesis , Myofibroblasts , Protein Serine-Threonine Kinases , Pulmonary Alveoli , Signal Transduction , YAP-Signaling Proteins , Animals , Mice , Myofibroblasts/metabolism , Myofibroblasts/cytology , YAP-Signaling Proteins/metabolism , YAP-Signaling Proteins/genetics , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/cytology , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Morphogenesis/genetics , Mesoderm/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Lung/metabolism , Organogenesis/genetics , Gene Expression Regulation, Developmental
2.
bioRxiv ; 2024 Mar 09.
Article in English | MEDLINE | ID: mdl-38496421

ABSTRACT

Hermansky-Pudlak syndrome (HPS) is a genetic disorder associated with pulmonary fibrosis in specific subtypes, including HPS-1 and HPS-2. Single mutant HPS1 and HPS2 mice display increased fibrotic sensitivity while double mutant HPS1/2 mice exhibit spontaneous fibrosis with aging, which has been attributed to HPS mutations in alveolar epithelial type II (AT2) cells. Unifying mechanisms of AT2 cell dysfunction in genetic and sporadic fibrotic lung diseases remain unknown. Incorporating AT2 cell lineage tracing in HPS mice, we observed a progressive decline in AT2 cell numbers with aging and aberrant differentiation with increased AT2-derived alveolar epithelial type I cells. HPS AT2 cell proliferation was impaired ex vivo and in vivo , suggesting an intrinsic progenitor defect. Transcriptomic analysis of HPS AT2 cells revealed elevated expression of genes associated with aberrant differentiation and cellular senescence. Through lineage tracing and organoid modeling, we demonstrated that HPS AT2 cells were primed to persist in a Krt8 + reprogrammed transitional state, mediated by p53 activity. These findings suggest that pulmonary fibrosis in HPS may be driven by AT2 cell progenitor dysfunction in the setting of p53-mediated senescence, highlighting a novel potential therapeutic target in HPS and suggesting unifying mechanisms underlying HPS and other forms of pulmonary fibrosis.

5.
JCI Insight ; 8(16)2023 08 22.
Article in English | MEDLINE | ID: mdl-37463053

ABSTRACT

Optimal lung repair and regeneration are essential for recovery from viral infections, including influenza A virus (IAV). We have previously demonstrated that acute inflammation and mortality induced by IAV is under circadian control. However, it is not known whether the influence of the circadian clock persists beyond the acute outcomes. Here, we utilize the UK Biobank to demonstrate an association between poor circadian rhythms and morbidity from lower respiratory tract infections, including the need for hospitalization and mortality after discharge; this persists even after adjusting for common confounding factors. Furthermore, we use a combination of lung organoid assays, single-cell RNA sequencing, and IAV infection in different models of clock disruption to investigate the role of the circadian clock in lung repair and regeneration. We show that lung organoids have a functional circadian clock and the disruption of this clock impairs regenerative capacity. Finally, we find that the circadian clock acts through distinct pathways in mediating lung regeneration - in tracheal cells via the Wnt/ß-catenin pathway and through IL-1ß in alveolar epithelial cells. We speculate that adding a circadian dimension to the critical process of lung repair and regeneration will lead to novel therapies and improve outcomes.


Subject(s)
Circadian Clocks , Influenza A virus , Lung/metabolism , Alveolar Epithelial Cells , Circadian Rhythm , Circadian Clocks/genetics , Influenza A virus/physiology , Regeneration
6.
Am J Respir Crit Care Med ; 208(6): 709-725, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37463497

ABSTRACT

Rationale: Alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV) is a lethal developmental disorder of lung morphogenesis caused by insufficiency of FOXF1 (forkhead box F1) transcription factor function. The cellular and transcriptional mechanisms by which FOXF1 deficiency disrupts human lung formation are unknown. Objectives: To identify cell types, gene networks, and cell-cell interactions underlying the pathogenesis of ACDMPV. Methods: We used single-nucleus RNA and assay for transposase-accessible chromatin sequencing, immunofluorescence confocal microscopy, and RNA in situ hybridization to identify cell types and molecular networks influenced by FOXF1 in ACDMPV lungs. Measurements and Main Results: Pathogenic single-nucleotide variants and copy-number variant deletions involving the FOXF1 gene locus in all subjects with ACDMPV (n = 6) were accompanied by marked changes in lung structure, including deficient alveolar development and a paucity of pulmonary microvasculature. Single-nucleus RNA and assay for transposase-accessible chromatin sequencing identified alterations in cell number and gene expression in endothelial cells (ECs), pericytes, fibroblasts, and epithelial cells in ACDMPV lungs. Distinct cell-autonomous roles for FOXF1 in capillary ECs and pericytes were identified. Pathogenic variants involving the FOXF1 gene locus disrupt gene expression in EC progenitors, inhibiting the differentiation or survival of capillary 2 ECs and cell-cell interactions necessary for both pulmonary vasculogenesis and alveolar type 1 cell differentiation. Loss of the pulmonary microvasculature was associated with increased VEGFA (vascular endothelial growth factor A) signaling and marked expansion of systemic bronchial ECs expressing COL15A1 (collagen type XV α 1 chain). Conclusions: Distinct FOXF1 gene regulatory networks were identified in subsets of pulmonary endothelial and fibroblast progenitors, providing both cellular and molecular targets for the development of therapies for ACDMPV and other diffuse lung diseases of infancy.


Subject(s)
Persistent Fetal Circulation Syndrome , Infant, Newborn , Humans , Persistent Fetal Circulation Syndrome/genetics , Persistent Fetal Circulation Syndrome/pathology , Gene Regulatory Networks/genetics , Vascular Endothelial Growth Factor A/genetics , Endothelial Cells/pathology , Multiomics , Lung/pathology , RNA , Forkhead Transcription Factors/genetics
7.
J Pediatr ; 261: 113564, 2023 10.
Article in English | MEDLINE | ID: mdl-37329980

ABSTRACT

OBJECTIVE: To evaluate associations between cardiac catheterization (cath) hemodynamics, quantitative measures of right ventricular (RV) function by echocardiogram, and survival in patients with congenital diaphragmatic hernia (CDH). STUDY DESIGN: This single-center retrospective cohort study enrolled patients with CDH who underwent index cath from 2003 to 2022. Tricuspid annular plane systolic excursion z score, RV fractional area change, RV free wall and global longitudinal strain, left ventricular (LV) eccentricity index, RV/LV ratio, and pulmonary artery acceleration time were measured from preprocedure echocardiograms. Associations between hemodynamic values, echocardiographic measures, and survival were evaluated by Spearman correlation and Wilcoxon rank sum test, respectively. RESULTS: Fifty-three patients (68% left-sided, 74% liver herniation, 57% extracorporeal membrane oxygenation, 93% survival) underwent cath (39 during index hospitalization, 14 later) including device closure of a patent ductus arteriosus in 5. Most patients (n = 31, 58%) were on pulmonary hypertension treatment at cath, most commonly sildenafil (n = 24, 45%) and/or intravenous treprostinil (n = 16, 30%). Overall, hemodynamics were consistent with precapillary pulmonary hypertension. Pulmonary capillary wedge pressure was >15 mm Hg in 2 patients (4%). Lower fractional area change and worse ventricular strain were associated with higher pulmonary artery pressure while higher LV eccentricity index and higher RV/LV ratio were associated with both higher pulmonary artery pressure and higher pulmonary vascular resistance. Hemodynamics did not differ based on survival status. CONCLUSIONS: Worse RV dilation and dysfunction by echocardiogram correlate with higher pulmonary artery pressure and pulmonary vascular resistance on cath in this CDH cohort. These measures may represent novel, noninvasive clinical trial targets in this population.


Subject(s)
Hernias, Diaphragmatic, Congenital , Hypertension, Pulmonary , Ventricular Dysfunction, Right , Humans , Hernias, Diaphragmatic, Congenital/diagnostic imaging , Hernias, Diaphragmatic, Congenital/complications , Retrospective Studies , Hypertension, Pulmonary/complications , Ventricular Dysfunction, Right/etiology , Ventricular Dysfunction, Right/complications , Echocardiography/methods , Cardiac Catheterization , Hemodynamics , Ventricular Function, Right
9.
Development ; 149(21)2022 11 01.
Article in English | MEDLINE | ID: mdl-36239312

ABSTRACT

There is a growing amount of data uncovering the cellular diversity of the pulmonary circulation and mechanisms governing vascular repair after injury. However, the molecular and cellular mechanisms contributing to the morphogenesis and growth of the pulmonary vasculature during embryonic development are less clear. Importantly, deficits in vascular development lead to significant pediatric lung diseases, indicating a need to uncover fetal programs promoting vascular growth. To address this, we used a transgenic mouse reporter for expression of Cxcl12, an arterial endothelial hallmark gene, and performed single-cell RNA sequencing on isolated Cxcl12-DsRed+ endothelium to assess cellular heterogeneity within pulmonary endothelium. Combining cell annotation with gene ontology and histological analysis allowed us to segregate the developing artery endothelium into functionally and spatially distinct subpopulations. Expression of Cxcl12 is highest in the distal arterial endothelial subpopulation, a compartment enriched in genes for vascular development. Accordingly, disruption of CXCL12 signaling led to, not only abnormal branching, but also distal vascular hypoplasia. These data provide evidence for arterial endothelial functional heterogeneity and reveal conserved signaling mechanisms essential for pulmonary vascular development.


Subject(s)
Endothelium, Vascular , Lung , Mice , Pregnancy , Animals , Female , Endothelium, Vascular/metabolism , Morphogenesis , Mice, Transgenic , Embryonic Development
10.
Mod Pathol ; 35(12): 1870-1881, 2022 12.
Article in English | MEDLINE | ID: mdl-35794233

ABSTRACT

The potential pathogenetic mechanisms underlying the varied morphology of congenital pulmonary airway malformations (CPAMs) have not been molecularly determined, but a subset have been shown to contain clusters of mucinous cells (MCC). These clusters are believed to serve as precursors for potential invasive mucinous adenocarcinoma, and they are associated with KRAS codon 12 mutations. To assess the universality of KRAS mutations in MCCs, we sequenced exon 2 of KRAS in 61 MCCs from 18 patients, and we found a KRAS codon 12 mutation in all 61 MCCs. Furthermore, all MCCs from a single patient always had the same KRAS mutation, and the same KRAS mutation was also found in non-mucinous lesional tissue. Next generation sequencing of seven MCCs showed no other mutations or copy number variations. Sequencing of 46 additional CPAMs with MCCs revealed KRAS mutations in non-mucinous lesional tissue in all cases. RNA in situ hybridization confirmed widespread distribution of cells with mutant KRAS RNA, even extending outside of the bronchiolar type epithelium. We identified 25 additional CPAMs with overall histologic architecture similar to CPAMs with KRAS mutations but without identifiable MCCs, and we found KRAS mutations in 17 (68%). The histologic features of these KRAS mutated CPAMs included type 1 and type 3 morphology, as well as lesions with an intermediate histologic appearance, and analysis revealed a strong correlation between the specific amino acid substitution and histomorphology. These findings, together with previously published model organism data, suggests that the formation of type 1 and 3 CPAMs is driven by mosaic KRAS mutations arising in the lung epithelium early in development and places them within the growing field of mosaic RASopathies. The presence of widespread epithelial mutation explains late metastatic disease in incompletely resected patients and reinforces the recommendation for complete resection of these lesions.


Subject(s)
Adenocarcinoma, Mucinous , Lung Neoplasms , Humans , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , DNA Copy Number Variations , Adenocarcinoma, Mucinous/pathology , Mutation , Lung Neoplasms/genetics , Lung Neoplasms/pathology , RNA , Codon
11.
J Perinatol ; 42(9): 1246-1254, 2022 09.
Article in English | MEDLINE | ID: mdl-35676536

ABSTRACT

OBJECTIVE: To evaluate factors associated with discontinuation of pulmonary vasodilator therapy in bronchopulmonary dysplasia-related pulmonary hypertension (BPD-PH). STUDY DESIGN: Retrospective study of neonatal, echocardiographic, and cardiac catheterization data in 121 infants with BPD-PH discharged on pulmonary vasodilator therapy from 2009-2020 and followed into childhood. RESULT: After median 4.4 years, medications were discontinued in 58%. Those in whom medications were discontinued had fewer days of invasive support, less severe BPD, lower incidence of PDA closure or cardiac catheterization, and higher incidence of fundoplication or tracheostomy decannulation (p < 0.05). On multivariable analysis, likelihood of medication discontinuation was lower with longer period of invasive respiratory support [HR 0.95 (CI:0.91-0.99), p = 0.01] and worse RV dilation on pre-discharge echocardiogram [HR 0.13 (CI:0.03-0.70), p = 0.017]. In those with tracheostomy, likelihood of medication discontinuation was higher with decannulation [HR 10.78 (CI:1.98-58.59), p < 0.001]. CONCLUSION: In BPD-PH, childhood discontinuation of pulmonary vasodilator therapy is associated with markers of disease severity.


Subject(s)
Bronchopulmonary Dysplasia , Hypertension, Pulmonary , Bronchopulmonary Dysplasia/epidemiology , Child , Humans , Hypertension, Pulmonary/complications , Hypertension, Pulmonary/etiology , Infant , Infant, Newborn , Lung , Retrospective Studies , Vasodilator Agents/therapeutic use
12.
Elife ; 112022 05 13.
Article in English | MEDLINE | ID: mdl-35559731

ABSTRACT

Dyskeratosis congenita (DC) is a rare genetic disorder characterized by deficiencies in telomere maintenance leading to very short telomeres and the premature onset of certain age-related diseases, including pulmonary fibrosis (PF). PF is thought to derive from epithelial failure, particularly that of type II alveolar epithelial (AT2) cells, which are highly dependent on Wnt signaling during development and adult regeneration. We use human induced pluripotent stem cell-derived AT2 (iAT2) cells to model how short telomeres affect AT2 cells. Cultured DC mutant iAT2 cells accumulate shortened, uncapped telomeres and manifest defects in the growth of alveolospheres, hallmarks of senescence, and apparent defects in Wnt signaling. The GSK3 inhibitor, CHIR99021, which mimics the output of canonical Wnt signaling, enhances telomerase activity and rescues the defects. These findings support further investigation of Wnt agonists as potential therapies for DC-related pathologies.


Subject(s)
Dyskeratosis Congenita , Induced Pluripotent Stem Cells , Telomerase , Alveolar Epithelial Cells/metabolism , Dyskeratosis Congenita/genetics , Dyskeratosis Congenita/pathology , Glycogen Synthase Kinase 3 , Humans , Induced Pluripotent Stem Cells/metabolism , Mutation , Telomerase/genetics , Telomerase/metabolism , Telomere/metabolism
13.
Nat Commun ; 12(1): 4291, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34257302

ABSTRACT

In utero base editing has the potential to correct disease-causing mutations before the onset of pathology. Mucopolysaccharidosis type I (MPS-IH, Hurler syndrome) is a lysosomal storage disease (LSD) affecting multiple organs, often leading to early postnatal cardiopulmonary demise. We assessed in utero adeno-associated virus serotype 9 (AAV9) delivery of an adenine base editor (ABE) targeting the Idua G→A (W392X) mutation in the MPS-IH mouse, corresponding to the common IDUA G→A (W402X) mutation in MPS-IH patients. Here we show efficient long-term W392X correction in hepatocytes and cardiomyocytes and low-level editing in the brain. In utero editing was associated with improved survival and amelioration of metabolic, musculoskeletal, and cardiac disease. This proof-of-concept study demonstrates the possibility of efficiently performing therapeutic base editing in multiple organs before birth via a clinically relevant delivery mechanism, highlighting the potential of this approach for MPS-IH and other genetic diseases.


Subject(s)
Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/pathology , Animals , Disease Models, Animal , Hepatocytes/metabolism , Humans , Mutation/genetics , Myocytes, Cardiac/metabolism
14.
Matrix Biol ; 102: 1-19, 2021 08.
Article in English | MEDLINE | ID: mdl-34314838

ABSTRACT

This study queried the role of type V collagen in the post-natal growth of temporomandibular joint (TMJ) condylar cartilage, a hybrid tissue with a fibrocartilage layer covering a secondary hyaline cartilage layer. Integrating outcomes from histology, immunofluorescence imaging, electron microscopy and atomic force microscopy-based nanomechanical tests, we elucidated the impact of type V collagen reduction on TMJ condylar cartilage growth in the type V collagen haploinsufficiency and inducible knockout mice. Reduction of type V collagen led to significantly thickened collagen fibrils, decreased tissue modulus, reduced cell density and aberrant cell clustering in both the fibrous and hyaline layers. Post-natal growth of condylar cartilage involves the chondrogenesis of progenitor cells residing in the fibrous layer, which gives rise to the secondary hyaline layer. Loss of type V collagen resulted in reduced proliferation of these cells, suggesting a possible role of type V collagen in mediating the progenitor cell niche. When the knockout of type V collagen was induced in post-weaning mice after the start of physiologic TMJ loading, the hyaline layer exhibited pronounced thinning, supporting an interplay between type V collagen and occlusal loading in condylar cartilage growth. The phenotype in hyaline layer can thus be attributed to the impact of type V collagen on the mechanically regulated progenitor cell activities. In contrast, knee cartilage does not contain the progenitor cell population at post-natal stages, and develops normal structure and biomechanical properties with the loss of type V collagen. Therefore, in the TMJ, in addition to its established role in regulating the assembly of collagen I fibrils, type V collagen also impacts the mechanoregulation of progenitor cell activities in the fibrous layer. We expect such knowledge to establish a foundation for understanding condylar cartilage matrix development and regeneration, and to yield new insights into the TMJ symptoms in patients with classic Ehlers-Danlos syndrome, a genetic disease due to autosomal mutation of type V collagen.


Subject(s)
Cartilage, Articular , Collagen Type V , Animals , Biomechanical Phenomena , Cartilage , Humans , Hyalin , Mandibular Condyle , Mice , Temporomandibular Joint
15.
Cell Stem Cell ; 28(10): 1775-1789.e5, 2021 10 07.
Article in English | MEDLINE | ID: mdl-33974915

ABSTRACT

Regeneration of the architecturally complex alveolar niche of the lung requires precise temporal and spatial control of epithelial cell behavior. Injury can lead to a permanent reduction in gas exchange surface area and respiratory function. Using mouse models, we show that alveolar type 1 (AT1) cell plasticity is a major and unappreciated mechanism that drives regeneration, beginning in the early postnatal period during alveolar maturation. Upon acute neonatal lung injury, AT1 cells reprogram into alveolar type 2 (AT2) cells, promoting alveolar regeneration. In contrast, the ability of AT2 cells to regenerate AT1 cells is restricted to the mature lung. Unbiased genomic assessment reveals that this previously unappreciated level of plasticity is governed by the preferential activity of Hippo signaling in the AT1 cell lineage. Thus, cellular plasticity is a temporally acquired trait of the alveolar epithelium and presents an alternative mode of tissue regeneration in the postnatal lung.


Subject(s)
Alveolar Epithelial Cells , Lung , Animals , Homeostasis , Mice , Respiratory Mucosa , Signal Transduction
16.
J Perinatol ; 41(7): 1528-1539, 2021 07.
Article in English | MEDLINE | ID: mdl-33674714

ABSTRACT

Primary pulmonary vein stenosis (PPVS) is an emerging problem among infants. In contrast to acquired disease, PPVS is the development of stenosis in the absence of preceding intervention. While optimal care approaches remain poorly characterized, over the past decade, understanding of potential pathophysiological mechanisms and development of novel therapeutic strategies are increasing. A multidisciplinary team of health care providers was assembled to review the available evidence and provide a common framework for the diagnosis, management, and treatment of PPVS during infancy. To address knowledge gaps, institutional and multi-institutional approaches must be employed to generate knowledge specific to ex-premature infants with PPVS. Within individual institutions, creation of a team comprised of dedicated health care providers from diverse backgrounds is critical to accelerate clinical learning and provide care for infants with PPVS. Multi-institutional collaborations, such as the PVS Network, provide the infrastructure and statistical power to advance knowledge for this rare disease.


Subject(s)
Infant, Premature, Diseases , Pulmonary Veins , Stenosis, Pulmonary Vein , Constriction, Pathologic , Humans , Infant , Infant, Newborn , Infant, Premature , Stenosis, Pulmonary Vein/diagnostic imaging , Stenosis, Pulmonary Vein/etiology , Stenosis, Pulmonary Vein/therapy
17.
Science ; 371(6534)2021 03 12.
Article in English | MEDLINE | ID: mdl-33707239

ABSTRACT

The lung alveolus is the functional unit of the respiratory system required for gas exchange. During the transition to air breathing at birth, biophysical forces are thought to shape the emerging tissue niche. However, the intercellular signaling that drives these processes remains poorly understood. Applying a multimodal approach, we identified alveolar type 1 (AT1) epithelial cells as a distinct signaling hub. Lineage tracing demonstrates that AT1 progenitors align with receptive, force-exerting myofibroblasts in a spatial and temporal manner. Through single-cell chromatin accessibility and pathway expression (SCAPE) analysis, we demonstrate that AT1-restricted ligands are required for myofibroblasts and alveolar formation. These studies show that the alignment of cell fates, mediated by biophysical and AT1-derived paracrine signals, drives the extensive tissue remodeling required for postnatal respiration.


Subject(s)
Cell Lineage/genetics , Epigenesis, Genetic , Pulmonary Alveoli/embryology , Alveolar Epithelial Cells/cytology , Alveolar Epithelial Cells/metabolism , Animals , Cells, Cultured , Cues , Epigenomics , Humans , Mice , Mice, Transgenic , Myofibroblasts/cytology , Myofibroblasts/metabolism , Pulmonary Alveoli/cytology , Pulmonary Alveoli/metabolism , RNA-Seq/methods , Signal Transduction , Single-Cell Analysis , Transcriptome
18.
Cell Stem Cell ; 28(5): 938-954.e9, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33529599

ABSTRACT

Pathogenic mutations in LAMIN A/C (LMNA) cause abnormal nuclear structure and laminopathies. These diseases have myriad tissue-specific phenotypes, including dilated cardiomyopathy (DCM), but how LMNA mutations result in tissue-restricted disease phenotypes remains unclear. We introduced LMNA mutations from individuals with DCM into human induced pluripotent stem cells (hiPSCs) and found that hiPSC-derived cardiomyocytes, in contrast to hepatocytes or adipocytes, exhibit aberrant nuclear morphology and specific disruptions in peripheral chromatin. Disrupted regions were enriched for transcriptionally active genes and regions with lower LAMIN B1 contact frequency. The lamina-chromatin interactions disrupted in mutant cardiomyocytes were enriched for genes associated with non-myocyte lineages and correlated with higher expression of those genes. Myocardium from individuals with LMNA variants similarly showed aberrant expression of non-myocyte pathways. We propose that the lamina network safeguards cellular identity and that pathogenic LMNA variants disrupt peripheral chromatin with specific epigenetic and molecular characteristics, causing misexpression of genes normally expressed in other cell types.


Subject(s)
Cardiomyopathy, Dilated , Induced Pluripotent Stem Cells , Cardiomyopathy, Dilated/genetics , Chromatin/genetics , Humans , Lamin Type A/genetics , Mutation/genetics , Myocytes, Cardiac
19.
Nat Commun ; 11(1): 5640, 2020 11 06.
Article in English | MEDLINE | ID: mdl-33159078

ABSTRACT

Lymphangioleiomyomatosis (LAM) is a rare fatal cystic lung disease due to bi-allelic inactivating mutations in tuberous sclerosis complex (TSC1/TSC2) genes coding for suppressors of the mechanistic target of rapamycin complex 1 (mTORC1). The origin of LAM cells is still unknown. Here, we profile a LAM lung compared to an age- and sex-matched healthy control lung as a hypothesis-generating approach to identify cell subtypes that are specific to LAM. Our single-cell RNA sequencing (scRNA-seq) analysis reveals novel mesenchymal and transitional alveolar epithelial states unique to LAM lung. This analysis identifies a mesenchymal cell hub coordinating the LAM disease phenotype. Mesenchymal-restricted deletion of Tsc2 in the mouse lung produces a mTORC1-driven pulmonary phenotype, with a progressive disruption of alveolar structure, a decline in pulmonary function, increase of rapamycin-sensitive expression of WNT ligands, and profound female-specific changes in mesenchymal and epithelial lung cell gene expression. Genetic inactivation of WNT signaling reverses age-dependent changes of mTORC1-driven lung phenotype, but WNT activation alone in lung mesenchyme is not sufficient for the development of mouse LAM-like phenotype. The alterations in gene expression are driven by distinctive crosstalk between mesenchymal and epithelial subsets of cells observed in mesenchymal Tsc2-deficient lungs. This study identifies sex- and age-specific gene changes in the mTORC1-activated lung mesenchyme and establishes the importance of the WNT signaling pathway in the mTORC1-driven lung phenotype.


Subject(s)
Lung/metabolism , Lymphangioleiomyomatosis/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mesoderm/metabolism , Age Factors , Aged , Animals , Female , Humans , Lung/drug effects , Lung/physiopathology , Lymphangioleiomyomatosis/drug therapy , Lymphangioleiomyomatosis/genetics , Lymphangioleiomyomatosis/physiopathology , Male , Mechanistic Target of Rapamycin Complex 1/genetics , Mesoderm/drug effects , Mice , Sex Factors , Sirolimus/administration & dosage , Tuberous Sclerosis Complex 2 Protein/genetics , Tuberous Sclerosis Complex 2 Protein/metabolism , Wnt Signaling Pathway
20.
Nat Cell Biol ; 22(10): 1197-1210, 2020 10.
Article in English | MEDLINE | ID: mdl-32989251

ABSTRACT

Alveolar epithelial regeneration is essential for recovery from devastating lung diseases. This process occurs when type II alveolar pneumocytes (AT2 cells) proliferate and transdifferentiate into type I alveolar pneumocytes (AT1 cells). We used genome-wide analysis of chromatin accessibility and gene expression following acute lung injury to elucidate repair mechanisms. AT2 chromatin accessibility changed substantially following injury to reveal STAT3 binding motifs adjacent to genes that regulate essential regenerative pathways. Single-cell transcriptome analysis identified brain-derived neurotrophic factor (Bdnf) as a STAT3 target gene with newly accessible chromatin in a unique population of regenerating AT2 cells. Furthermore, the BDNF receptor tropomyosin receptor kinase B (TrkB) was enriched on mesenchymal alveolar niche cells (MANCs). Loss or blockade of AT2-specific Stat3, Bdnf or mesenchyme-specific TrkB compromised repair and reduced Fgf7 expression by niche cells. A TrkB agonist improved outcomes in vivo following lung injury. These data highlight the biological and therapeutic importance of the STAT3-BDNF-TrkB axis in orchestrating alveolar epithelial regeneration.


Subject(s)
Alveolar Epithelial Cells/cytology , Brain-Derived Neurotrophic Factor/metabolism , Lung Injury/prevention & control , Membrane Glycoproteins/metabolism , Protein-Tyrosine Kinases/metabolism , Receptor, trkB/metabolism , Regeneration , STAT3 Transcription Factor/metabolism , Alveolar Epithelial Cells/metabolism , Animals , Brain-Derived Neurotrophic Factor/genetics , Female , Humans , Lung Injury/etiology , Lung Injury/pathology , Male , Membrane Glycoproteins/genetics , Protein-Tyrosine Kinases/genetics , Receptor, trkB/genetics , STAT3 Transcription Factor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...