Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Clin Invest ; 134(6)2024 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-38488000

RESUMEN

Premature birth disrupts normal lung development and places infants at risk for bronchopulmonary dysplasia (BPD), a disease disrupting lung health throughout the life of an individual and that is increasing in incidence. The TGF-ß superfamily has been implicated in BPD pathogenesis, however, what cell lineage it impacts remains unclear. We show that TGFbr2 is critical for alveolar epithelial (AT1) cell fate maintenance and function. Loss of TGFbr2 in AT1 cells during late lung development leads to AT1-AT2 cell reprogramming and altered pulmonary architecture, which persists into adulthood. Restriction of fetal lung stretch and associated AT1 cell spreading through a model of oligohydramnios enhances AT1-AT2 reprogramming. Transcriptomic and proteomic analyses reveal the necessity of TGFbr2 expression in AT1 cells for extracellular matrix production. Moreover, TGF-ß signaling regulates integrin transcription to alter AT1 cell morphology, which further impacts ECM expression through changes in mechanotransduction. These data reveal the cell intrinsic necessity of TGF-ß signaling in maintaining AT1 cell fate and reveal this cell lineage as a major orchestrator of the alveolar matrisome.


Asunto(s)
Displasia Broncopulmonar , Alveolos Pulmonares , Humanos , Ratones , Animales , Recién Nacido , Receptor Tipo II de Factor de Crecimiento Transformador beta/genética , Receptor Tipo II de Factor de Crecimiento Transformador beta/metabolismo , Alveolos Pulmonares/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Mecanotransducción Celular , Proteómica , Células Epiteliales Alveolares , Pulmón/patología , Diferenciación Celular , Matriz Extracelular/metabolismo , Displasia Broncopulmonar/patología , Transcripción Genética
2.
bioRxiv ; 2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-38529490

RESUMEN

Severe lung injury causes basal stem cells to migrate and outcompete alveolar stem cells resulting in dysplastic repair and a loss of gas exchange function. This "stem cell collision" is part of a multistep process that is now revealed to generate an injury-induced tissue niche (iTCH) containing Keratin 5+ epithelial cells and plastic Pdgfra+ mesenchymal cells. Temporal and spatial single cell analysis reveals that iTCHs are governed by mesenchymal proliferation and Notch signaling, which suppresses Wnt and Fgf signaling in iTCHs. Conversely, loss of Notch in iTCHs rewires alveolar signaling patterns to promote euplastic regeneration and gas exchange. The signaling patterns of iTCHs can differentially phenotype fibrotic from degenerative human lung diseases, through apposing flows of FGF and WNT signaling. These data reveal the emergence of an injury and disease associated iTCH in the lung and the ability of using iTCH specific signaling patterns to discriminate human lung disease phenotypes.

3.
medRxiv ; 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38464240

RESUMEN

MTSS1 (metastasis suppressor 1) is an I-BAR protein that regulates cytoskeleton dynamics through interactions with actin, Rac, and actin-associated proteins. In a prior study, we identified genetic variants in a cardiac-specific enhancer upstream of MTSS1 that reduce human left ventricular (LV) MTSS1 expression and associate with protection against dilated cardiomyopathy (DCM). We sought to probe these effects further using population genomics and in vivo murine models. We crossed Mtss1-/- mice with a transgenic (Tg) murine model of human DCM caused by the D230N pathogenic mutation in Tpm1 (tropomyosin 1). In females, Tg/Mtss1+/- mice had significantly increased LV ejection fraction and reduced LV volumes relative to their Tg/Mtss1+/+ counterparts, signifying partial rescue of DCM due to Mtss1 haploinsufficiency. No differences were observed in males. To study effects in humans, we fine-mapped the MTSS1 locus with 82 cardiac magnetic resonance (CMR) traits in 22,381 UK Biobank participants. MTSS1 enhancer variants showed interaction with biological sex in their associations with several CMR traits. After stratification by biological sex, associations with CMR traits and colocalization with MTSS1 expression in the Genotype-Tissue Expression (GTEx) Project were observed principally in women and were substantially weaker in men. These findings suggest sex dimorphism in the effects of MTSS1-lowering alleles, and parallel the increased LV ejection fraction and reduced LV volumes observed female Tg/Mtss1+/- mice. Together, our findings at the MTSS1 locus suggest a genetic basis for sex dimorphism in cardiac remodeling and motivate sex-specific study of common variants associated with cardiac traits and disease.

4.
NPJ Regen Med ; 9(1): 2, 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-38182591

RESUMEN

Maintenance of the cellular boundary between airway and alveolar compartments during homeostasis and after injury is essential to prohibit pathological plasticity which can reduce respiratory function. Lung injury and disease can induce either functional alveolar epithelial regeneration or dysplastic formation of keratinized epithelium which does not efficiently contribute to gas exchange. Here we show that Sox2 preserves airway cell identity and prevents fate changes into either functional alveolar tissue or pathological keratinization following lung injury. Loss of Sox2 in airway epithelium leads to a loss of airway epithelial identity with a commensurate gain in alveolar and basal cell identity, in part due to activation of Wnt signaling in secretory cells and increased Trp63 expression in intrapulmonary basal-like progenitors. In idiopathic pulmonary fibrosis, loss of SOX2 expression correlates with increased WNT signaling activity in dysplastic keratinized epithelium. SOX2-deficient dysplastic epithelial cells are also observed in COVID-19 damaged lungs. Thus, Sox2 provides a molecular barrier that suppresses airway epithelial plasticity to prevent acquisition of alveolar or basal cell identity after injury and help guide proper epithelial fate and regeneration.

5.
bioRxiv ; 2023 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-37214932

RESUMEN

Premature birth disrupts normal lung development and places infants at risk for bronchopulmonary dysplasia (BPD), a disease increasing in incidence which disrupts lung health throughout the lifespan. The TGFß superfamily has been implicated in BPD pathogenesis, however, what cell lineage it impacts remains unclear. We show that Tgfbr2 is critical for AT1 cell fate maintenance and function. Loss of Tgfbr2 in AT1 cells during late lung development leads to AT1-AT2 cell reprogramming and altered pulmonary architecture, which persists into adulthood. Restriction of fetal lung stretch and associated AT1 cell spreading through a model of oligohydramnios enhances AT1-AT2 reprogramming. Transcriptomic and proteomic analysis reveal the necessity of Tgfbr2 expression in AT1 cells for extracellular matrix production. Moreover, TGFß signaling regulates integrin transcription to alter AT1 cell morphology, which further impacts ECM expression through changes in mechanotransduction. These data reveal the cell intrinsic necessity of TGFß signaling in maintaining AT1 cell fate and reveal this cell lineage as a major orchestrator of the alveolar matrisome.

6.
Adv Mater ; 34(28): e2202992, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35522531

RESUMEN

Epithelial cell organoids have increased opportunities to probe questions on tissue development and disease in vitro and for therapeutic cell transplantation. Despite their potential, current protocols to grow these organoids almost exclusively depend on culture within 3D Matrigel, which limits defined culture conditions, introduces animal components, and results in heterogenous organoids (i.e., shape, size, composition). Here, a method is described that relies on hyaluronic acid hydrogels for the generation and expansion of lung alveolar organoids (alveolospheres). Using synthetic hydrogels with defined chemical and physical properties, human-induced pluripotent stem cell (iPSC)-derived alveolar type 2 cells (iAT2s) self-assemble into alveolospheres and propagate in Matrigel-free conditions. By engineering predefined microcavities within these hydrogels, the heterogeneity of alveolosphere size and structure is reduced when compared to 3D culture, while maintaining the alveolar type 2 cell fate of human iAT2-derived progenitor cells. This hydrogel system is a facile and accessible system for the culture of iPSC-derived lung progenitors and the method can be expanded to the culture of primary mouse tissue derived AT2 and other epithelial progenitor and stem cell aggregates.


Asunto(s)
Hidrogeles , Células Madre Pluripotentes Inducidas , Animales , Humanos , Ácido Hialurónico/metabolismo , Hidrogeles/química , Células Madre Pluripotentes Inducidas/metabolismo , Pulmón , Ratones , Organoides/metabolismo
7.
Circ Heart Fail ; 15(6): e009521, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35543134

RESUMEN

BACKGROUND: Defects in energetics are thought to be central to the pathophysiology of hypertrophic cardiomyopathy (HCM); yet, the determinants of ATP availability are not known. The purpose of this study is to ascertain the nature and extent of metabolic reprogramming in human HCM, and its potential impact on contractile function. METHODS: We conducted proteomic and targeted, quantitative metabolomic analyses on heart tissue from patients with HCM and from nonfailing control human hearts. RESULTS: In the proteomic analysis, the greatest differences observed in HCM samples compared with controls were increased abundances of extracellular matrix and intermediate filament proteins and decreased abundances of muscle creatine kinase and mitochondrial proteins involved in fatty acid oxidation. These differences in protein abundance were coupled with marked reductions in acyl carnitines, byproducts of fatty acid oxidation, in HCM samples. Conversely, the ketone body 3-hydroxybutyrate, branched chain amino acids, and their breakdown products, were all significantly increased in HCM hearts. ATP content, phosphocreatine, nicotinamide adenine dinucleotide and its phosphate derivatives, NADP and NADPH, and acetyl CoA were also severely reduced in HCM compared with control hearts. Functional assays performed on human skinned myocardial fibers demonstrated that the magnitude of observed reduction in ATP content in the HCM samples would be expected to decrease the rate of cross-bridge detachment. Moreover, left atrial size, an indicator of diastolic compliance, was inversely correlated with ATP content in hearts from patients with HCM. CONCLUSIONS: HCM hearts display profound deficits in nucleotide availability with markedly reduced capacity for fatty acid oxidation and increases in ketone bodies and branched chain amino acids. These results have important therapeutic implications for the future design of metabolic modulators to treat HCM.


Asunto(s)
Cardiomiopatía Hipertrófica , Insuficiencia Cardíaca , Adenosina Trifosfato/metabolismo , Aminoácidos de Cadena Ramificada/metabolismo , Ácidos Grasos/metabolismo , Insuficiencia Cardíaca/metabolismo , Humanos , Metaboloma , Miocitos Cardíacos/metabolismo , Proteoma , Proteómica
8.
Nature ; 604(7904): 120-126, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35355013

RESUMEN

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.


Asunto(s)
Bronquiolos , Hurones , Células Madre Multipotentes , Alveolos Pulmonares , Animales , Bronquiolos/citología , Linaje de la Célula , Humanos , Pulmón/patología , Ratones , Células Madre Multipotentes/citología , Alveolos Pulmonares/citología , Enfermedad Pulmonar Obstructiva Crónica
9.
Nat Commun ; 11(1): 5640, 2020 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-33159078

RESUMEN

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.


Asunto(s)
Pulmón/metabolismo , Linfangioleiomiomatosis/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Mesodermo/metabolismo , Factores de Edad , Anciano , Animales , Femenino , Humanos , Pulmón/efectos de los fármacos , Pulmón/fisiopatología , Linfangioleiomiomatosis/tratamiento farmacológico , Linfangioleiomiomatosis/genética , Linfangioleiomiomatosis/fisiopatología , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Mesodermo/efectos de los fármacos , Ratones , Factores Sexuales , Sirolimus/administración & dosificación , Proteína 2 del Complejo de la Esclerosis Tuberosa/genética , Proteína 2 del Complejo de la Esclerosis Tuberosa/metabolismo , Vía de Señalización Wnt
10.
Cell Rep ; 30(9): 3105-3116.e4, 2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-32130910

RESUMEN

The mammalian heart is incapable of regenerating a sufficient number of cardiomyocytes to ameliorate the loss of contractile muscle after acute myocardial injury. Several reports have demonstrated that mononucleated cardiomyocytes are more responsive than are binucleated cardiomyocytes to pro-proliferative stimuli. We have developed a strategy to isolate and characterize highly enriched populations of mononucleated and binucleated cardiomyocytes at various times of development. Our results suggest that an E2f/Rb transcriptional network is central to the divergence of these two populations and that remnants of the differences acquired during the neonatal period remain in adult cardiomyocytes. Moreover, inducing binucleation by genetically blocking the ability of cardiomyocytes to complete cytokinesis leads to a reduction in E2f target gene expression, directly linking the E2f pathway with nucleation. These data identify key molecular differences between mononucleated and binucleated mammalian cardiomyocytes that can be used to leverage cardiomyocyte proliferation for promoting injury repair in the heart.


Asunto(s)
Núcleo Celular/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Animales , Animales Recién Nacidos , Secuencia de Bases , Núcleo Celular/ultraestructura , Proliferación Celular , Separación Celular , Regulación hacia Abajo/genética , Factores de Transcripción E2F/metabolismo , Citometría de Flujo , Fase G1 , Ratones Noqueados , Miocitos Cardíacos/ultraestructura , Proteínas Proto-Oncogénicas/metabolismo , Regeneración , Proteína de Retinoblastoma/metabolismo , Fase S
11.
Nature ; 573(7774): 430-433, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31511695

RESUMEN

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.


Asunto(s)
Linfocitos T CD8-positivos , Fibrosis Endomiocárdica/terapia , Inmunoterapia Adoptiva , Animales , Antígenos de Superficie/inmunología , Linfocitos T CD8-positivos/inmunología , Fibrosis Endomiocárdica/inmunología , Fibroblastos/inmunología , Humanos , Masculino , Ratones , Ovalbúmina/inmunología , Cicatrización de Heridas
12.
Dev Biol ; 454(2): 108-117, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31242446

RESUMEN

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.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1/genética , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Animales , Diferenciación Celular/genética , Linaje de la Célula/fisiología , Polaridad Celular , Proliferación Celular/genética , ADN (Citosina-5-)-Metiltransferasa 1/fisiología , Metilación de ADN/genética , Endodermo/metabolismo , Epigénesis Genética/genética , Células Epiteliales/citología , Transición Epitelial-Mesenquimal , Femenino , Regulación del Desarrollo de la Expresión Génica , Pulmón/citología , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Morfogénesis , Organogénesis/fisiología , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo
14.
Science ; 364(6438)2019 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-31023895

RESUMEN

Metabolic health depends on the capacity of adipose tissue progenitor cells to undergo de novo adipogenesis. The cellular hierarchy and mechanisms governing adipocyte progenitor differentiation are incompletely understood. Through single-cell RNA sequence analyses, we show that the lineage hierarchy of adipocyte progenitors consists of distinct mesenchymal cell types that are present in both mouse and human adipose tissues. Cells marked by dipeptidyl peptidase-4 (DPP4)/CD26 expression are highly proliferative, multipotent progenitors. During the development of subcutaneous adipose tissue in mice, these progenitor cells give rise to intercellular adhesion molecule-1 (ICAM1)/CD54-expressing (CD54+) committed preadipocytes and a related adipogenic cell population marked by Clec11a and F3/CD142 expression. Transforming growth factor-ß maintains DPP4+ cell identity and inhibits adipogenic commitment of DPP4+ and CD142+ cells. Notably, DPP4+ progenitors reside in the reticular interstitium, a recently appreciated fluid-filled space within and between tissues, including adipose depots.


Asunto(s)
Adipocitos/citología , Adipogénesis , Tejido Adiposo/citología , Células Madre Mesenquimatosas/citología , Adipocitos/enzimología , Animales , Dipeptidil Peptidasa 4/metabolismo , Factores de Crecimiento de Célula Hematopoyética/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Lectinas Tipo C/metabolismo , Células Madre Mesenquimatosas/enzimología , Ratones , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Tromboplastina/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
15.
Nature ; 555(7695): 251-255, 2018 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-29489752

RESUMEN

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.


Asunto(s)
Células Epiteliales/citología , Evolución Molecular , Alveolos Pulmonares/citología , Regeneración , Células Madre/citología , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/cirugía , Animales , Antígenos de Superficie/metabolismo , Proteína Axina/metabolismo , Biomarcadores/metabolismo , Ciclo Celular , Linaje de la Célula , Cromatina/genética , Cromatina/metabolismo , Epigenómica , Células Epiteliales/metabolismo , Femenino , Factores de Crecimiento de Fibroblastos/metabolismo , Humanos , Masculino , Ratones , Proteínas de Neoplasias/metabolismo , Organoides/citología , Organoides/metabolismo , Células Madre/metabolismo , Transcriptoma , Vía de Señalización Wnt
16.
Cell ; 170(6): 1134-1148.e10, 2017 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-28886382

RESUMEN

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.


Asunto(s)
Pulmón/citología , Mesodermo/citología , Algoritmos , Animales , Células Epiteliales/metabolismo , Fibrosis/metabolismo , Perfilación de la Expresión Génica , Pulmón/patología , Pulmón/fisiología , Lesión Pulmonar/patología , Ratones , Organoides/citología , Comunicación Paracrina , Regeneración , Transducción de Señal , Análisis de la Célula Individual , Células Madre/metabolismo
17.
Genes Dev ; 31(9): 889-903, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28546511

RESUMEN

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.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Homeostasis , Pulmón/crecimiento & desarrollo , Pulmón/fisiología , Proteínas Nucleares/metabolismo , ARN Largo no Codificante/metabolismo , Factores de Transcripción/genética , Animales , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Humanos , Inmunidad Celular , Pulmón/inmunología , Ratones , Proteínas Nucleares/genética , ARN Largo no Codificante/genética , Factor Nuclear Tiroideo 1 , Factores de Transcripción/metabolismo
18.
Cell Rep ; 17(9): 2312-2325, 2016 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-27880906

RESUMEN

Alveologenesis is the culmination of lung development and involves the correct temporal and spatial signals to generate the delicate gas exchange interface required for respiration. Using a Wnt-signaling reporter system, we demonstrate the emergence of a Wnt-responsive alveolar epithelial cell sublineage, which arises during alveologenesis, called the axin2+ alveolar type 2 cell, or AT2Axin2. The number of AT2Axin2 cells increases substantially during late lung development, correlating with a wave of Wnt signaling during alveologenesis. Transcriptome analysis, in vivo clonal analysis, and ex vivo lung organoid assays reveal that AT2sAxin2 promote enhanced AT2 cell growth during generation of the alveolus. Activating Wnt signaling results in the expansion of AT2s, whereas inhibition of Wnt signaling inhibits AT2 cell development and shunts alveolar epithelial development toward the alveolar type 1 cell lineage. These findings reveal a wave of Wnt-dependent AT2 expansion required for lung alveologenesis and maturation.


Asunto(s)
Diferenciación Celular , Autorrenovación de las Células , Células Epiteliales/citología , Pulmón/embriología , Organogénesis , Alveolos Pulmonares/embriología , Vía de Señalización Wnt , Animales , Proteína Axina/metabolismo , Linaje de la Célula , Proliferación Celular , Células Clonales , Células Epiteliales/metabolismo , Epitelio/embriología , Genes Reporteros , Integrasas/metabolismo , Ratones , Modelos Biológicos , Organogénesis/genética , Organoides , Alveolos Pulmonares/citología , Vía de Señalización Wnt/genética
19.
Dev Cell ; 36(3): 303-15, 2016 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-26832331

RESUMEN

The terminal stages of pulmonary development, called sacculation and alveologenesis, involve both differentiation of distal lung endoderm progenitors and extensive cellular remodeling of the resultant epithelial lineages. These processes are coupled with dramatic expansion of distal airspace and surface area. Despite the importance of these late developmental processes and their relation to neonatal respiratory diseases, little is understood about the molecular and cellular pathways critical for their successful completion. We show that a histone deacetylase 3 (Hdac3)-mediated epigenetic pathway is critical for the proper remodeling and expansion of the distal lung saccules into primitive alveoli. Loss of Hdac3 in the developing lung epithelium leads to a reduction of alveolar type 1 cell spreading and a disruption of lung sacculation. Hdac3 represses miR-17-92 expression, a microRNA cluster that regulates transforming growth factor ß (TGF-ß) signaling. De-repression of miR-17-92 in Hdac3-deficient lung epithelium results in decreased TGF-ß signaling activity. Importantly, inhibition of TGF-ß signaling and overexpression of miR-17-92 can phenocopy the defects observed in Hdac3 null lungs. Conversely, loss of miR-17-92 expression rescues many of the defects caused by loss of Hdac3 in the lung. These studies reveal an intricate epigenetic pathway where Hdac3 is required to repress miR-17-92 expression to allow for proper TGF-ß signaling during lung sacculation.


Asunto(s)
Epigénesis Genética , Células Epiteliales/citología , Histona Desacetilasas/metabolismo , Pulmón/metabolismo , MicroARNs/genética , Transducción de Señal , Animales , Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica , Pulmón/embriología , Ratones , Factor de Crecimiento Transformador beta/metabolismo
20.
Nature ; 526(7574): 578-82, 2015 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-26436454

RESUMEN

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.


Asunto(s)
Proteínas Hedgehog/metabolismo , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Pulmón/citología , Pulmón/metabolismo , Regeneración , Cicatrización de Heridas , Animales , Proliferación Celular , Regulación hacia Abajo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Retroalimentación Fisiológica , Proteínas Hedgehog/deficiencia , Proteínas Hedgehog/genética , Homeostasis , Pulmón/patología , Masculino , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Comunicación Paracrina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA