Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 186(7): 1478-1492.e15, 2023 03 30.
Article in English | MEDLINE | ID: mdl-36870331

ABSTRACT

Lungs undergo mechanical strain during breathing, but how these biophysical forces affect cell fate and tissue homeostasis are unclear. We show that biophysical forces through normal respiratory motion actively maintain alveolar type 1 (AT1) cell identity and restrict these cells from reprogramming into AT2 cells in the adult lung. AT1 cell fate is maintained at homeostasis by Cdc42- and Ptk2-mediated actin remodeling and cytoskeletal strain, and inactivation of these pathways causes a rapid reprogramming into the AT2 cell fate. This plasticity induces chromatin reorganization and changes in nuclear lamina-chromatin interactions, which can discriminate AT1 and AT2 cell identity. Unloading the biophysical forces of breathing movements leads to AT1-AT2 cell reprogramming, revealing that normal respiration is essential to maintain alveolar epithelial cell fate. These data demonstrate the integral function of mechanotransduction in maintaining lung cell fate and identifies the AT1 cell as an important mechanosensor in the alveolar niche.


Subject(s)
Alveolar Epithelial Cells , Mechanotransduction, Cellular , Alveolar Epithelial Cells/metabolism , Cells, Cultured , Lung , Cell Differentiation/physiology , Respiration
2.
Cell ; 170(6): 1134-1148.e10, 2017 Sep 07.
Article in English | MEDLINE | ID: mdl-28886382

ABSTRACT

The lung is an architecturally complex organ comprising a heterogeneous mixture of various epithelial and mesenchymal lineages. We use single-cell RNA sequencing and signaling lineage reporters to generate a spatial and transcriptional map of the lung mesenchyme. We find that each mesenchymal lineage has a distinct spatial address and transcriptional profile leading to unique niche regulatory functions. The mesenchymal alveolar niche cell is Wnt responsive, expresses Pdgfrα, and is critical for alveolar epithelial cell growth and self-renewal. In contrast, the Axin2+ myofibrogenic progenitor cell preferentially generates pathologically deleterious myofibroblasts after injury. Analysis of the secretome and receptome of the alveolar niche reveals functional pathways that mediate growth and self-renewal of alveolar type 2 progenitor cells, including IL-6/Stat3, Bmp, and Fgf signaling. These studies define the cellular and molecular framework of lung mesenchymal niches and reveal the functional importance of developmental pathways in promoting self-renewal versus a pathological response to tissue injury.


Subject(s)
Lung/cytology , Mesoderm/cytology , Algorithms , Animals , Epithelial Cells/metabolism , Fibrosis/metabolism , Gene Expression Profiling , Lung/pathology , Lung/physiology , Lung Injury/pathology , Mice , Organoids/cytology , Paracrine Communication , Regeneration , Signal Transduction , Single-Cell Analysis , Stem Cells/metabolism
3.
Cell ; 171(3): 573-587.e14, 2017 Oct 19.
Article in English | MEDLINE | ID: mdl-29033129

ABSTRACT

Progenitor cells differentiate into specialized cell types through coordinated expression of lineage-specific genes and modification of complex chromatin configurations. We demonstrate that a histone deacetylase (Hdac3) organizes heterochromatin at the nuclear lamina during cardiac progenitor lineage restriction. Specification of cardiomyocytes is associated with reorganization of peripheral heterochromatin, and independent of deacetylase activity, Hdac3 tethers peripheral heterochromatin containing lineage-relevant genes to the nuclear lamina. Deletion of Hdac3 in cardiac progenitor cells releases genomic regions from the nuclear periphery, leading to precocious cardiac gene expression and differentiation into cardiomyocytes; in contrast, restricting Hdac3 to the nuclear periphery rescues myogenesis in progenitors otherwise lacking Hdac3. Our results suggest that availability of genomic regions for activation by lineage-specific factors is regulated in part through dynamic chromatin-nuclear lamina interactions and that competence of a progenitor cell to respond to differentiation signals may depend upon coordinated movement of responding gene loci away from the nuclear periphery.


Subject(s)
Chromatin/metabolism , Gene Expression Regulation, Developmental , Histone Deacetylases/metabolism , Nuclear Lamina/metabolism , Stem Cells/cytology , Animals , Genome , Mice , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Stem Cells/metabolism
4.
Nature ; 604(7904): 120-126, 2022 04.
Article in English | MEDLINE | ID: mdl-35355013

ABSTRACT

The human lung differs substantially from its mouse counterpart, resulting in a distinct distal airway architecture affected by disease pathology in chronic obstructive pulmonary disease. In humans, the distal branches of the airway interweave with the alveolar gas-exchange niche, forming an anatomical structure known as the respiratory bronchioles. Owing to the lack of a counterpart in mouse, the cellular and molecular mechanisms that govern respiratory bronchioles in the human lung remain uncharacterized. Here we show that human respiratory bronchioles contain a unique secretory cell population that is distinct from cells in larger proximal airways. Organoid modelling reveals that these respiratory airway secretory (RAS) cells act as unidirectional progenitors for alveolar type 2 cells, which are essential for maintaining and regenerating the alveolar niche. RAS cell lineage differentiation into alveolar type 2 cells is regulated by Notch and Wnt signalling. In chronic obstructive pulmonary disease, RAS cells are altered transcriptionally, corresponding to abnormal alveolar type 2 cell states, which are associated with smoking exposure in both humans and ferrets. These data identify a distinct progenitor in a region of the human lung that is not found in mouse that has a critical role in maintaining the gas-exchange compartment and is altered in chronic lung disease.


Subject(s)
Bronchioles , Ferrets , Multipotent Stem Cells , Pulmonary Alveoli , Animals , Bronchioles/cytology , Cell Lineage , Humans , Lung/pathology , Mice , Multipotent Stem Cells/cytology , Pulmonary Alveoli/cytology , Pulmonary Disease, Chronic Obstructive
5.
Nature ; 575(7782): 375-379, 2019 11.
Article in English | MEDLINE | ID: mdl-31618756

ABSTRACT

Mitochondrial homeostasis depends on mitophagy, the programmed degradation of mitochondria. Only a few proteins are known to participate in mitophagy. Here we develop a multidimensional CRISPR-Cas9 genetic screen, using multiple mitophagy reporter systems and pro-mitophagy triggers, and identify numerous components of parkin-dependent mitophagy1. Unexpectedly, we find that the adenine nucleotide translocator (ANT) complex is required for mitophagy in several cell types. Whereas pharmacological inhibition of ANT-mediated ADP/ATP exchange promotes mitophagy, genetic ablation of ANT paradoxically suppresses mitophagy. Notably, ANT promotes mitophagy independently of its nucleotide translocase catalytic activity. Instead, the ANT complex is required for inhibition of the presequence translocase TIM23, which leads to stabilization of PINK1, in response to bioenergetic collapse. ANT modulates TIM23 indirectly via interaction with TIM44, which regulates peptide import through TIM232. Mice that lack ANT1 show blunted mitophagy and consequent profound accumulation of aberrant mitochondria. Disease-causing human mutations in ANT1 abrogate binding to TIM44 and TIM23 and inhibit mitophagy. Together, our findings show that ANT is an essential and fundamental mediator of mitophagy in health and disease.


Subject(s)
Mitophagy , Animals , Cell Line , Mice , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Precursor Protein Import Complex Proteins , Nucleotides/metabolism , Protein Binding , Protein Kinases/genetics , Protein Kinases/metabolism
6.
Nature ; 573(7774): 430-433, 2019 09.
Article in English | MEDLINE | ID: mdl-31511695

ABSTRACT

Fibrosis is observed in nearly every form of myocardial disease1. Upon injury, cardiac fibroblasts in the heart begin to remodel the myocardium by depositing excess extracellular matrix, resulting in increased stiffness and reduced compliance of the tissue. Excessive cardiac fibrosis is an important factor in the progression of various forms of cardiac disease and heart failure2. However, clinical interventions and therapies that target fibrosis remain limited3. Here we demonstrate the efficacy of redirected T cell immunotherapy to specifically target pathological cardiac fibrosis in mice. We find that cardiac fibroblasts that express a xenogeneic antigen can be effectively targeted and ablated by adoptive transfer of antigen-specific CD8+ T cells. Through expression analysis of the gene signatures of cardiac fibroblasts obtained from healthy and diseased human hearts, we identify an endogenous target of cardiac fibroblasts-fibroblast activation protein. Adoptive transfer of T cells that express a chimeric antigen receptor against fibroblast activation protein results in a significant reduction in cardiac fibrosis and restoration of function after injury in mice. These results provide proof-of-principle for the development of immunotherapeutic drugs for the treatment of cardiac disease.


Subject(s)
CD8-Positive T-Lymphocytes , Endomyocardial Fibrosis/therapy , Immunotherapy, Adoptive , Animals , Antigens, Surface/immunology , CD8-Positive T-Lymphocytes/immunology , Endomyocardial Fibrosis/immunology , Fibroblasts/immunology , Humans , Male , Mice , Ovalbumin/immunology , Wound Healing
8.
Genes Dev ; 31(9): 889-903, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28546511

ABSTRACT

A subset of long noncoding RNAs (lncRNAs) is spatially correlated with transcription factors (TFs) across the genome, but how these lncRNA-TF gene duplexes regulate tissue development and homeostasis is unclear. We identified a feedback loop within the NANCI (Nkx2.1-associated noncoding intergenic RNA)-Nkx2.1 gene duplex that is essential for buffering Nkx2.1 expression, lung epithelial cell identity, and tissue homeostasis. Within this locus, Nkx2.1 directly inhibits NANCI, while NANCI acts in cis to promote Nkx2.1 transcription. Although loss of NANCI alone does not adversely affect lung development, concurrent heterozygous mutations in both NANCI and Nkx2.1 leads to persistent Nkx2.1 deficiency and reprogramming of lung epithelial cells to a posterior endoderm fate. This disruption in the NANCI-Nkx2.1 gene duplex results in a defective perinatal innate immune response, tissue damage, and progressive degeneration of the adult lung. These data point to a mechanism in which lncRNAs act as rheostats within lncRNA-TF gene duplex loci that buffer TF expression, thereby maintaining tissue-specific cellular identity during development and postnatal homeostasis.


Subject(s)
Gene Expression Regulation, Developmental , Homeostasis , Lung/growth & development , Lung/physiology , Nuclear Proteins/metabolism , RNA, Long Noncoding/metabolism , Transcription Factors/genetics , Animals , Epithelial Cells/immunology , Epithelial Cells/metabolism , Humans , Immunity, Cellular , Lung/immunology , Mice , Nuclear Proteins/genetics , RNA, Long Noncoding/genetics , Thyroid Nuclear Factor 1 , Transcription Factors/metabolism
9.
Nature ; 555(7695): 251-255, 2018 03 08.
Article in English | MEDLINE | ID: mdl-29489752

ABSTRACT

Functional tissue regeneration is required for the restoration of normal organ homeostasis after severe injury. Some organs, such as the intestine, harbour active stem cells throughout homeostasis and regeneration; more quiescent organs, such as the lung, often contain facultative progenitor cells that are recruited after injury to participate in regeneration. Here we show that a Wnt-responsive alveolar epithelial progenitor (AEP) lineage within the alveolar type 2 cell population acts as a major facultative progenitor cell in the distal lung. AEPs are a stable lineage during alveolar homeostasis but expand rapidly to regenerate a large proportion of the alveolar epithelium after acute lung injury. AEPs exhibit a distinct transcriptome, epigenome and functional phenotype and respond specifically to Wnt and Fgf signalling. In contrast to other proposed lung progenitor cells, human AEPs can be directly isolated by expression of the conserved cell surface marker TM4SF1, and act as functional human alveolar epithelial progenitor cells in 3D organoids. Our results identify the AEP lineage as an evolutionarily conserved alveolar progenitor that represents a new target for human lung regeneration strategies.


Subject(s)
Epithelial Cells/cytology , Evolution, Molecular , Pulmonary Alveoli/cytology , Regeneration , Stem Cells/cytology , Acute Lung Injury/pathology , Acute Lung Injury/surgery , Animals , Antigens, Surface/metabolism , Axin Protein/metabolism , Biomarkers/metabolism , Cell Cycle , Cell Lineage , Chromatin/genetics , Chromatin/metabolism , Epigenomics , Epithelial Cells/metabolism , Female , Fibroblast Growth Factors/metabolism , Humans , Male , Mice , Neoplasm Proteins/metabolism , Organoids/cytology , Organoids/metabolism , Stem Cells/metabolism , Transcriptome , Wnt Signaling Pathway
10.
Circ Res ; 127(6): 761-777, 2020 08 28.
Article in English | MEDLINE | ID: mdl-32529949

ABSTRACT

RATIONALE: Identifying genetic markers for heterogeneous complex diseases such as heart failure is challenging and requires prohibitively large cohort sizes in genome-wide association studies to meet the stringent threshold of genome-wide statistical significance. On the other hand, chromatin quantitative trait loci, elucidated by direct epigenetic profiling of specific human tissues, may contribute toward prioritizing subthreshold variants for disease association. OBJECTIVE: Here, we captured noncoding genetic variants by performing epigenetic profiling for enhancer H3K27ac chromatin immunoprecipitation followed by sequencing in 70 human control and end-stage failing hearts. METHODS AND RESULTS: We have mapped a comprehensive catalog of 47 321 putative human heart enhancers and promoters. Three thousand eight hundred ninety-seven differential acetylation peaks (FDR [false discovery rate], 5%) pointed to pathways altered in heart failure. To identify cardiac histone acetylation quantitative trait loci (haQTLs), we regressed out confounding factors including heart failure disease status and used the G-SCI (Genotype-independent Signal Correlation and Imbalance) test1 to call out 1680 haQTLs (FDR, 10%). RNA sequencing performed on the same heart samples proved a subset of haQTLs to have significant association also to gene expression (expression quantitative trait loci), either in cis (180) or through long-range interactions (81), identified by Hi-C (high-throughput chromatin conformation assay) and HiChIP (high-throughput protein centric chromatin) performed on a subset of hearts. Furthermore, a concordant relationship between the gain or disruption of TF (transcription factor)-binding motifs, inferred from alternative alleles at the haQTLs, implied a surprising direct association between these specific TF and local histone acetylation in human hearts. Finally, 62 unique loci were identified by colocalization of haQTLs with the subthreshold loci of heart-related genome-wide association studies datasets. CONCLUSIONS: Disease and phenotype association for 62 unique loci are now implicated. These loci may indeed mediate their effect through modification of enhancer H3K27 acetylation enrichment and their corresponding gene expression differences (bioRxiv: https://doi.org/10.1101/536763). Graphical Abstract: A graphical abstract is available for this article.


Subject(s)
Epigenome , Genetic Variation , Heart Failure/genetics , Histones/genetics , Acetylation , Adult , Aged , Aged, 80 and over , Case-Control Studies , Chromatin Immunoprecipitation , Databases, Genetic , Epigenesis, Genetic , Epigenomics , Female , Genetic Predisposition to Disease , Heart Failure/diagnosis , Heart Failure/physiopathology , High-Throughput Nucleotide Sequencing , Histones/metabolism , Humans , Male , Middle Aged , Phenotype , Quantitative Trait Loci
11.
Proc Natl Acad Sci U S A ; 116(10): 4362-4371, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30782824

ABSTRACT

During the stepwise specification and differentiation of tissue-specific multipotent progenitors, lineage-specific transcriptional networks are activated or repressed to orchestrate cell specification. The gas-exchange niche in the lung contains two major epithelial cell types, alveolar type 1 (AT1) and AT2 cells, and the timing of lineage specification of these cells is critical for the correct formation of this niche and postnatal survival. Integrating cell-specific lineage tracing studies, spatially specific mRNA transcript and protein expression, and single-cell RNA-sequencing analysis, we demonstrate that specification of alveolar epithelial cell fate begins concomitantly with the proximal-distal specification of epithelial progenitors and branching morphogenesis earlier than previously appreciated. By using a newly developed dual-lineage tracing system, we show that bipotent alveolar cells that give rise to AT1 and AT2 cells are a minor contributor to the alveolar epithelial population. Furthermore, single-cell assessment of the transcriptome identifies specified AT1 and AT2 progenitors rather than bipotent cells during sacculation. These data reveal a paradigm of organ formation whereby lineage specification occurs during the nascent stages of development coincident with broad tissue-patterning processes, including axial patterning of the endoderm and branching morphogenesis.


Subject(s)
Cell Lineage , Lung/cytology , Pulmonary Alveoli/cytology , Animals , Cell Differentiation , Female , In Situ Hybridization, Fluorescence , Mice , Pregnancy , Transcriptome
12.
Eur Heart J ; 42(20): 2000-2011, 2021 05 21.
Article in English | MEDLINE | ID: mdl-33677556

ABSTRACT

AIMS: Our objective was to better understand the genetic bases of dilated cardiomyopathy (DCM), a leading cause of systolic heart failure. METHODS AND RESULTS: We conducted the largest genome-wide association study performed so far in DCM, with 2719 cases and 4440 controls in the discovery population. We identified and replicated two new DCM-associated loci on chromosome 3p25.1 [lead single-nucleotide polymorphism (SNP) rs62232870, P = 8.7 × 10-11 and 7.7 × 10-4 in the discovery and replication steps, respectively] and chromosome 22q11.23 (lead SNP rs7284877, P = 3.3 × 10-8 and 1.4 × 10-3 in the discovery and replication steps, respectively), while confirming two previously identified DCM loci on chromosomes 10 and 1, BAG3 and HSPB7. A genetic risk score constructed from the number of risk alleles at these four DCM loci revealed a 3-fold increased risk of DCM for individuals with 8 risk alleles compared to individuals with 5 risk alleles (median of the referral population). In silico annotation and functional 4C-sequencing analyses on iPSC-derived cardiomyocytes identify SLC6A6 as the most likely DCM gene at the 3p25.1 locus. This gene encodes a taurine transporter whose involvement in myocardial dysfunction and DCM is supported by numerous observations in humans and animals. At the 22q11.23 locus, in silico and data mining annotations, and to a lesser extent functional analysis, strongly suggest SMARCB1 as the candidate culprit gene. CONCLUSION: This study provides a better understanding of the genetic architecture of DCM and sheds light on novel biological pathways underlying heart failure.


Subject(s)
Cardiomyopathy, Dilated , Heart Failure, Systolic , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis Regulatory Proteins , Cardiomyopathy, Dilated/genetics , Chromosomes , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Heart Failure, Systolic/genetics , Humans , Polymorphism, Single Nucleotide/genetics
13.
Genes Dev ; 28(12): 1363-79, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24939938

ABSTRACT

Long noncoding RNAs (lncRNAs) are thought to play important roles in regulating gene transcription, but few have well-defined expression patterns or known biological functions during mammalian development. Using a conservative pipeline to identify lncRNAs that have important biological functions, we identified 363 lncRNAs in the lung and foregut endoderm. Importantly, we show that these lncRNAs are spatially correlated with transcription factors across the genome. In-depth expression analyses of lncRNAs with genomic loci adjacent to the critical transcription factors Nkx2.1, Gata6, Foxa2 (forkhead box a2), and Foxf1 mimic the expression patterns of their protein-coding neighbor. Loss-of-function analysis demonstrates that two lncRNAs, LL18/NANCI (Nkx2.1-associated noncoding intergenic RNA) and LL34, play distinct roles in endoderm development by controlling expression of critical developmental transcription factors and pathways, including retinoic acid signaling. In particular, we show that LL18/NANCI acts upstream of Nkx2.1 and downstream from Wnt signaling to regulate lung endoderm gene expression. These studies reveal that lncRNAs play an important role in foregut and lung endoderm development by regulating multiple aspects of gene transcription, often through regulation of transcription factor expression.


Subject(s)
Gene Expression Regulation, Developmental , Lung/growth & development , RNA, Long Noncoding/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation/genetics , Cell Line , Endoderm/cytology , Endoderm/metabolism , Gene Expression Profiling , Gene Knockdown Techniques , Genome , HEK293 Cells , Humans , Lung/embryology , Mice , Nuclear Proteins/genetics , RNA, Long Noncoding/genetics , Thyroid Nuclear Factor 1 , Transcription Factors/genetics , Wnt Signaling Pathway
14.
Nature ; 526(7574): 578-82, 2015 Oct 22.
Article in English | MEDLINE | ID: mdl-26436454

ABSTRACT

Postnatal tissue quiescence is thought to be a default state in the absence of a proliferative stimulus such as injury. Although previous studies have demonstrated that certain embryonic developmental programs are reactivated aberrantly in adult organs to drive repair and regeneration, it is not well understood how quiescence is maintained in organs such as the lung, which displays a remarkably low level of cellular turnover. Here we demonstrate that quiescence in the adult lung is an actively maintained state and is regulated by hedgehog signalling. Epithelial-specific deletion of sonic hedgehog (Shh) during postnatal homeostasis in the murine lung results in a proliferative expansion of the adjacent lung mesenchyme. Hedgehog signalling is initially downregulated during the acute phase of epithelial injury as the mesenchyme proliferates in response, but returns to baseline during injury resolution as quiescence is restored. Activation of hedgehog during acute epithelial injury attenuates the proliferative expansion of the lung mesenchyme, whereas inactivation of hedgehog signalling prevents the restoration of quiescence during injury resolution. Finally, we show that hedgehog also regulates epithelial quiescence and regeneration in response to injury via a mesenchymal feedback mechanism. These results demonstrate that epithelial-mesenchymal interactions coordinated by hedgehog actively maintain postnatal tissue homeostasis, and deregulation of hedgehog during injury leads to aberrant repair and regeneration in the lung.


Subject(s)
Hedgehog Proteins/metabolism , Lung Injury/metabolism , Lung Injury/pathology , Lung/cytology , Lung/metabolism , Regeneration , Wound Healing , Animals , Cell Proliferation , Down-Regulation , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Feedback, Physiological , Hedgehog Proteins/deficiency , Hedgehog Proteins/genetics , Homeostasis , Lung/pathology , Male , Mesoderm/cytology , Mesoderm/metabolism , Mice , Paracrine Communication
15.
Dev Biol ; 454(2): 108-117, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31242446

ABSTRACT

Lung endoderm development occurs through a series of finely coordinated transcriptional processes that are regulated by epigenetic mechanisms. However, the role of DNA methylation in regulating lung endoderm development remains poorly understood. We demonstrate that DNA methyltransferase 1 (Dnmt1) is required for early branching morphogenesis of the lungs and for restraining epithelial fate specification. Loss of Dnmt1 leads to an early branching defect, a loss of epithelial polarity and proximal endodermal cell differentiation, and an expansion of the distal endoderm compartment. Dnmt1 deficiency also disrupts epithelial-mesenchymal crosstalk and leads to precocious distal endodermal cell differentiation with premature expression of alveolar type 2 cell restricted genes. These data reveal an important requirement for Dnmt1 mediated DNA methylation in early lung development to promote proper branching morphogenesis, maintain proximal endodermal cell fate, and suppress premature activation of the distal epithelial fate.


Subject(s)
Alveolar Epithelial Cells/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Animals , Cell Differentiation/genetics , Cell Lineage/physiology , Cell Polarity , Cell Proliferation/genetics , DNA (Cytosine-5-)-Methyltransferase 1/physiology , DNA Methylation/genetics , Endoderm/metabolism , Epigenesis, Genetic/genetics , Epithelial Cells/cytology , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Developmental , Lung/cytology , Lung/metabolism , Lung/pathology , Male , Mice , Morphogenesis , Organogenesis/physiology , Signal Transduction/physiology , Transcription Factors/metabolism
16.
Development ; 142(1): 108-17, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25516972

ABSTRACT

The development of the lung epithelium is regulated in a stepwise fashion to generate numerous differentiated and stem cell lineages in the adult lung. How these different lineages are generated in a spatially and temporally restricted fashion remains poorly understood, although epigenetic regulation probably plays an important role. We show that the Polycomb repressive complex 2 component Ezh2 is highly expressed in early lung development but is gradually downregulated by late gestation. Deletion of Ezh2 in early lung endoderm progenitors leads to the ectopic and premature appearance of Trp63+ basal cells that extend the entire length of the airway. Loss of Ezh2 also leads to reduced secretory cell differentiation. In their place, morphologically similar cells develop that express a subset of basal cell genes, including keratin 5, but no longer express high levels of either Trp63 or of standard secretory cell markers. This suggests that Ezh2 regulates the phenotypic switch between basal cells and secretory cells. Together, these findings show that Ezh2 restricts the basal cell lineage during normal lung endoderm development to allow the proper patterning of epithelial lineages during lung formation.


Subject(s)
Cell Lineage , Endoderm/cytology , Endoderm/embryology , Lung/cytology , Lung/embryology , Polycomb Repressive Complex 2/metabolism , Animals , Biomarkers/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Proliferation , Enhancer of Zeste Homolog 2 Protein , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelium/embryology , Epithelium/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Ontology , Goblet Cells/cytology , Goblet Cells/metabolism , Hedgehog Proteins/metabolism , Keratin-5/metabolism , Lung/metabolism , Mice , Mutation/genetics , Neuroendocrine Cells/cytology , Neuroendocrine Cells/metabolism , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Phosphoproteins/metabolism , Software , Thyroid Nuclear Factor 1 , Trans-Activators/metabolism , Transcription Factors/metabolism
17.
Proc Natl Acad Sci U S A ; 111(34): 12444-9, 2014 Aug 26.
Article in English | MEDLINE | ID: mdl-25114215

ABSTRACT

Changing the morphology of a simple epithelial tube to form a highly ramified branching network requires changes in cell behavior that lead to tissue-wide changes in organ shape. How epithelial cells in branched organs modulate their shape and behavior to promote bending and sculpting of the epithelial sheet is not well understood, and the mechanisms underlying this process remain obscure. We show that the Wnt receptor Frizzled 2 (Fzd2) is required for domain branch formation during the initial establishment of the respiratory tree. Live imaging and transcriptome analysis of lung-branching morphogenesis demonstrate that Fzd2 promotes changes in epithelial cell length and shape. These changes in cell morphology deform the developing epithelial tube to generate and maintain new domain branches. Fzd2 controls branch formation and the shape of the epithelial tube by regulating Rho signaling and by the localization of phospho-myosin light chain 2, in turn controlling the changes in the shape of epithelial cells during morphogenesis. This study demonstrates the importance of Wnt/Fzd2 signaling in promoting and maintaining changes in epithelial cell shape that affect development of a branching network.


Subject(s)
Frizzled Receptors/metabolism , Lung/embryology , Animals , Cell Shape , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Frizzled Receptors/deficiency , Frizzled Receptors/genetics , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Ligands , Lung/cytology , Lung/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Morphogenesis , Pregnancy , Signal Transduction , Wnt Signaling Pathway , beta Catenin/metabolism , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein
20.
Arterioscler Thromb Vasc Biol ; 34(4): 902-12, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24504737

ABSTRACT

OBJECTIVE: Inappropriate transcriptional activation of innate immunity is a pathological feature of several cardiometabolic disorders, but little is known about inflammatory modulation of long intergenic noncoding RNAs (lincRNAs) in disease-relevant human tissues. APPROACH AND RESULTS: We applied deep RNA sequencing (>500 million filtered reads per sample) to blood and adipose during low-dose experimental endotoxemia (lipopolysaccharide) in a healthy human, with targeted replication in separate individuals undergoing endotoxemia (n=6), to identify inflammatory lincRNAs. A subset of these lincRNAs was examined for expression in adipocytes and monocytes, modulation in adipose of obese humans, and overlap with genome-wide association study signals for inflammatory and cardiometabolic traits. Of a stringent set of 4284 lincRNAs, ≈11% to 22% were expressed with 201 and 56 lincRNAs modulated by lipopolysaccharide in blood or adipose, respectively. Tissue-specific expression of a subset of 6 lipopolysaccharide-lincRNAs was replicated with lipopolysaccharide modulation confirmed for all 3 expressed in blood and 2 of 4 expressed in adipose. The broader generalizability of findings in blood of subject A was confirmed by RNA sequencing in 7 additional subjects. We confirmed adipocytes and monocytes as potential cell-sources of selective lipopolysaccharide-regulated lincRNAs, and 2 of these, linc-DMRT2 (P=0.002) and linc-TP53I13 (P=0.01), were suppressed in adipose of obese humans. Finally, we provide examples of lipopolysaccharide-modulated lincRNAs that overlap single nucleotide polymorphisms that are associated with cardiometabolic traits. CONCLUSIONS: Our findings provide novel insights into tissue-level, inflammatory transcriptome regulation in cardiometabolic diseases. These are complementary to more usual approaches limited to interrogation of DNA variations.


Subject(s)
Endotoxemia/genetics , Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing , Inflammation/genetics , Metabolic Syndrome/genetics , RNA, Long Noncoding/blood , Sequence Analysis, RNA/methods , Subcutaneous Fat/metabolism , Adipocytes/metabolism , Adult , Binding Sites , Case-Control Studies , Cells, Cultured , Endotoxemia/blood , Female , Gene Expression Regulation , Genetic Markers , Genome-Wide Association Study , Humans , Inflammation/blood , Inflammation Mediators/blood , Lipopolysaccharides/pharmacology , Male , Metabolic Syndrome/blood , Monocytes/metabolism , Obesity/blood , Obesity/genetics , Reproducibility of Results , Subcutaneous Fat/drug effects , Transcription Factors/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL