Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Nature ; 619(7971): 860-867, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37468622

RESUMEN

Many cancers originate from stem or progenitor cells hijacked by somatic mutations that drive replication, exemplified by adenomatous transformation of pulmonary alveolar epithelial type II (AT2) cells1. Here we demonstrate a different scenario: expression of KRAS(G12D) in differentiated AT1 cells reprograms them slowly and asynchronously back into AT2 stem cells that go on to generate indolent tumours. Like human lepidic adenocarcinoma, the tumour cells slowly spread along alveolar walls in a non-destructive manner and have low ERK activity. We find that AT1 and AT2 cells act as distinct cells of origin and manifest divergent responses to concomitant WNT activation and KRAS(G12D) induction, which accelerates AT2-derived but inhibits AT1-derived adenoma proliferation. Augmentation of ERK activity in KRAS(G12D)-induced AT1 cells increases transformation efficiency, proliferation and progression from lepidic to mixed tumour histology. Overall, we have identified a new cell of origin for lung adenocarcinoma, the AT1 cell, which recapitulates features of human lepidic cancer. In so doing, we also uncover a capacity for oncogenic KRAS to reprogram a differentiated and quiescent cell back into its parent stem cell en route to adenomatous transformation. Our work further reveals that irrespective of a given cancer's current molecular profile and driver oncogene, the cell of origin exerts a pervasive and perduring influence on its subsequent behaviour.


Asunto(s)
Adenocarcinoma del Pulmón , Reprogramación Celular , Neoplasias Pulmonares , Proteínas Proto-Oncogénicas p21(ras) , Células Madre , Humanos , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/patología , Reprogramación Celular/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Células Madre/metabolismo , Células Madre/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo
2.
Nature ; 588(7839): 670-675, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33238290

RESUMEN

The distal lung contains terminal bronchioles and alveoli that facilitate gas exchange. Three-dimensional in vitro human distal lung culture systems would strongly facilitate the investigation of pathologies such as interstitial lung disease, cancer and coronavirus disease 2019 (COVID-19) pneumonia caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Here we describe the development of a long-term feeder-free, chemically defined culture system for distal lung progenitors as organoids derived from single adult human alveolar epithelial type II (AT2) or KRT5+ basal cells. AT2 organoids were able to differentiate into AT1 cells, and basal cell organoids developed lumens lined with differentiated club and ciliated cells. Single-cell analysis of KRT5+ cells in basal organoids revealed a distinct population of ITGA6+ITGB4+ mitotic cells, whose offspring further segregated into a TNFRSF12Ahi subfraction that comprised about ten per cent of KRT5+ basal cells. This subpopulation formed clusters within terminal bronchioles and exhibited enriched clonogenic organoid growth activity. We created distal lung organoids with apical-out polarity to present ACE2 on the exposed external surface, facilitating infection of AT2 and basal cultures with SARS-CoV-2 and identifying club cells as a target population. This long-term, feeder-free culture of human distal lung organoids, coupled with single-cell analysis, identifies functional heterogeneity among basal cells and establishes a facile in vitro organoid model of human distal lung infections, including COVID-19-associated pneumonia.


Asunto(s)
COVID-19/virología , Pulmón/citología , Modelos Biológicos , Organoides/citología , Organoides/virología , SARS-CoV-2/fisiología , Técnicas de Cultivo de Tejidos , Células Epiteliales Alveolares/citología , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/virología , COVID-19/metabolismo , COVID-19/patología , Diferenciación Celular , División Celular , Células Clonales/citología , Células Clonales/metabolismo , Células Clonales/virología , Humanos , Técnicas In Vitro , Subtipo H1N1 del Virus de la Influenza A/crecimiento & desarrollo , Subtipo H1N1 del Virus de la Influenza A/fisiología , Integrina alfa6/análisis , Integrina beta4/análisis , Queratina-5/análisis , Organoides/metabolismo , Neumonía Viral/metabolismo , Neumonía Viral/patología , Neumonía Viral/virología , SARS-CoV-2/crecimiento & desarrollo , Análisis de la Célula Individual , Receptor de TWEAK/análisis
3.
Indian J Med Res ; 159(1): 71-77, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-38293841

RESUMEN

BACKGROUND OBJECTIVES: This study aimed to compare the admission characteristics and outcomes of tribal and non-tribal neonates admitted to a level II special newborn care unit (SNCU) in rural Gujarat. METHODS: This was a retrospective observational study that looked at all neonates admitted to a high-volume SNCU between 2013 and 2021. A series of quality improvement measures were introduced over the study period. Admission characteristics, such as birth weight, gestational age, gender and outcomes for tribal and non-tribal neonates, were compared. RESULTS: Six thousand nine hundred and ninety neonates [4829 tribal (69.1%) and 2161 (30.9%) non-tribal] were admitted to the SNCU. Tribal neonates had lower mean birth weight (2047 vs . 2311 g, P <0.01) and gestational week at birth (35.8 vs . 36.7 weeks, P <0.01) compared to non-tribal neonates. Common causes of admissions were neonatal jaundice (1990, 28.4%), low birth weight (1308, 18.7%) and neonatal sepsis (843, 12%). Six hundred and thirty-eight (9.1%) neonates died during the treatment in the SNCU. The odds of death among tribal neonates was similar to non-tribal neonates [adjusted odds ratio: 1.12 (95% confidence interval [CI]: 0.89, 1.42)]. The tribal neonates had significantly higher cause-specific case fatality rate from sepsis [relative risk (RR): 2.18 (95% CI: 1.41, 3.37)], prematurity [RR: 1.98 (95% CI: 1.23, 3.17)] and low birth weight [RR: 1.83 (95% CI: 1.17, 2.85)]. The overall case fatality rate in the SNCU decreased from 18.2 per cent during the year 2013-2014 to 2.1 per cent in the year 2020-2021. INTERPRETATION CONCLUSIONS: There was a reduction in the case fatality rate over the study period. Tribal and non-tribal neonates had similar risk of death. Sepsis prevention and management, mechanical respiratory support and timely referral to a higher centre might help further reduction in mortality for these neonates.


Asunto(s)
Hospitalización , Sepsis , Recién Nacido , Humanos , Peso al Nacer , Recien Nacido Prematuro , Recién Nacido de Bajo Peso , Sepsis/epidemiología
4.
Mol Ther ; 30(1): 223-237, 2022 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33794364

RESUMEN

Cystic fibrosis (CF) is a monogenic disease caused by impaired production and/or function of the CF transmembrane conductance regulator (CFTR) protein. Although we have previously shown correction of the most common pathogenic mutation, there are many other pathogenic mutations throughout the CF gene. An autologous airway stem cell therapy in which the CFTR cDNA is precisely inserted into the CFTR locus may enable the development of a durable cure for almost all CF patients, irrespective of the causal mutation. Here, we use CRISPR-Cas9 and two adeno-associated viruses (AAVs) carrying the two halves of the CFTR cDNA to sequentially insert the full CFTR cDNA along with a truncated CD19 (tCD19) enrichment tag in upper airway basal stem cells (UABCs) and human bronchial epithelial cells (HBECs). The modified cells were enriched to obtain 60%-80% tCD19+ UABCs and HBECs from 11 different CF donors with a variety of mutations. Differentiated epithelial monolayers cultured at air-liquid interface showed restored CFTR function that was >70% of the CFTR function in non-CF controls. Thus, our study enables the development of a therapy for almost all CF patients, including patients who cannot be treated using recently approved modulator therapies.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística , Fibrosis Quística , Sistemas CRISPR-Cas , Fibrosis Quística/genética , Fibrosis Quística/metabolismo , Fibrosis Quística/terapia , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Células Epiteliales/metabolismo , Humanos , Mutación , Células Madre/metabolismo
5.
Environ Toxicol ; 38(1): 39-48, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36124540

RESUMEN

Parkinson's disease (PD) is a common neurodegenerative disorder associated with striatal dopaminergic neuronal loss in the Substantia nigra. Oxidative stress plays a significant role in several neurodegenerative diseases. Paraquat (PQ) is considered a potential neurotoxin that affects the brain leading to the death of dopaminergic neurons mimicking the PD phenotype. Various scientific reports have proven that cryptotanshinone possesses antioxidant and anti-inflammatory properties. We hypothesized that cryptotanshinone could extend its neuroprotective activity by exerting antioxidant effects. This study was designed to evaluate the effects of cryptotanshinone in both cellular and animal models of PQ-induced PD. Annexin V-PI double staining and immunoblotting were used to detect apoptosis and oxidative stress proteins, respectively. Reactive oxygen species kits were used to evaluate oxidative stress in cells. For in vivo studies, 18 B6 mice were divided into three groups. The rotarod data revealed the motor function and immunostaining showed the survival of TH+ neurons in SNpc region. Our study showed that cryptotanshinone attenuated paraquat-induced oxidative stress by upregulating anti-oxidant markers in vitro, and restored behavioral deficits and survival of dopaminergic neurons in vivo, demonstrating its therapeutic potential.


Asunto(s)
Fármacos Neuroprotectores , Enfermedad de Parkinson , Animales , Ratones , Paraquat/toxicidad , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Neuronas Dopaminérgicas/metabolismo , Estrés Oxidativo , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Modelos Animales de Enfermedad
6.
Am J Respir Cell Mol Biol ; 67(3): 284-308, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35679511

RESUMEN

Clinical and molecular heterogeneity are common features of human disease. Understanding the basis for heterogeneity has led to major advances in therapy for many cancers and pulmonary diseases such as cystic fibrosis and asthma. Although heterogeneity of risk factors, disease severity, and outcomes in survivors are common features of the acute respiratory distress syndrome (ARDS), many challenges exist in understanding the clinical and molecular basis for disease heterogeneity and using heterogeneity to tailor therapy for individual patients. This report summarizes the proceedings of the 2021 Aspen Lung Conference, which was organized to review key issues related to understanding clinical and molecular heterogeneity in ARDS. The goals were to review new information about ARDS phenotypes, to explore multicellular and multisystem mechanisms responsible for heterogeneity, and to review how best to account for clinical and molecular heterogeneity in clinical trial design and assessment of outcomes. The report concludes with recommendations for future research to understand the clinical and basic mechanisms underlying heterogeneity in ARDS to advance the development of new treatments for this life-threatening critical illness.


Asunto(s)
Síndrome de Dificultad Respiratoria , Humanos , Pulmón , Factores de Riesgo , Índice de Severidad de la Enfermedad , Tórax
7.
Thorax ; 77(12): 1176-1186, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35580897

RESUMEN

INTRODUCTION: Chronic lung disease, that is, bronchopulmonary dysplasia (BPD) is the most common complication in preterm infants and develops as a consequence of the misguided formation of the gas-exchange area undergoing prenatal and postnatal injury. Subsequent vascular disease and its progression into pulmonary arterial hypertension critically determines long-term outcome in the BPD infant but lacks identification of early, disease-defining changes. METHODS: We link impaired bone morphogenetic protein (BMP) signalling to the earliest onset of vascular pathology in the human preterm lung and delineate the specific effects of the most prevalent prenatal and postnatal clinical risk factors for lung injury mimicking clinically relevant conditions in a multilayered animal model using wild-type and transgenic neonatal mice. RESULTS: We demonstrate (1) the significant reduction in BMP receptor 2 (BMPR2) expression at the onset of vascular pathology in the lung of preterm infants, later mirrored by reduced plasma BMP protein levels in infants with developing BPD, (2) the rapid impairment (and persistent change) of BMPR2 signalling on postnatal exposure to hyperoxia and mechanical ventilation, aggravated by prenatal cigarette smoke in a preclinical mouse model and (3) a link to defective alveolar septation and matrix remodelling through platelet derived growth factor-receptor alpha deficiency. In a treatment approach, we partially reversed vascular pathology by BMPR2-targeted treatment with FK506 in vitro and in vivo. CONCLUSION: We identified impaired BMP signalling as a hallmark of early vascular disease in the injured neonatal lung while outlining its promising potential as a future biomarker or therapeutic target in this growing, high-risk patient population.


Asunto(s)
Displasia Broncopulmonar , Hiperoxia , Lesiones del Sistema Vascular , Lactante , Recién Nacido , Humanos , Ratones , Animales , Recien Nacido Prematuro , Lesiones del Sistema Vascular/complicaciones , Lesiones del Sistema Vascular/patología , Displasia Broncopulmonar/etiología , Hiperoxia/complicaciones , Hiperoxia/metabolismo , Hiperoxia/patología , Pulmón , Ratones Transgénicos , Factores de Riesgo , Animales Recién Nacidos
8.
Cytotherapy ; 24(8): 774-788, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35613962

RESUMEN

The ISCT Scientific Signature Series Symposium "Advances in Cell and Gene Therapies for Lung Diseases and Critical Illnesses" was held as an independent symposium in conjunction with the biennial meeting, "Stem Cells, Cell Therapies, and Bioengineering in Lung Biology and Diseases," which took place July 12-15, 2021, at the University of Vermont. This is the third Respiratory System-based Signature Series event; the first 2, "Tracheal Bioengineering, the Next Steps" and "Cellular Therapies for Pulmonary Diseases and Critical Illnesses: State of the Art of European Science," took place in 2014 and 2015, respectively. Cell- and gene-based therapies for respiratory diseases and critical illnesses continue to be a source of great promise and opportunity. This reflects ongoing advancements in understanding of the mechanisms by which cell-based therapies, particularly those using mesenchymal stromal cells (MSCs), can mitigate different lung injuries and the increasing sophistication with which preclinical data is translated into clinical investigations. This also reflects continuing evolution in gene transfer vectors, including those designed for in situ gene editing in parallel with those targeting gene or cell replacement. Therefore, this symposium convened global thought leaders in a forum designed to catalyze communication and collaboration to bring the greatest possible innovation and value of cell- and gene-based therapies for patients with respiratory diseases and critical illnesses.


Asunto(s)
Enfermedad Crítica , Enfermedades Pulmonares , Tratamiento Basado en Trasplante de Células y Tejidos , Enfermedad Crítica/terapia , Terapia Genética , Humanos , Enfermedades Pulmonares/genética , Enfermedades Pulmonares/terapia , Células Madre
9.
Respir Res ; 22(1): 265, 2021 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-34666752

RESUMEN

RATIONALE: αv integrins, key regulators of transforming growth factor-ß activation and fibrogenesis in in vivo models of pulmonary fibrosis, are expressed on abnormal epithelial cells (αvß6) and fibroblasts (αvß1) in fibrotic lungs. OBJECTIVES: We evaluated multiple αv integrin inhibition strategies to assess which most effectively reduced fibrogenesis in explanted lung tissue from patients with idiopathic pulmonary fibrosis. METHODS: Selective αvß6 and αvß1, dual αvß6/αvß1, and multi-αv integrin inhibitors were characterized for potency, selectivity, and functional activity by ligand binding, cell adhesion, and transforming growth factor-ß cell activation assays. Precision-cut lung slices generated from lung explants from patients with idiopathic pulmonary fibrosis or bleomycin-challenged mouse lungs were treated with integrin inhibitors or standard-of-care drugs (nintedanib or pirfenidone) and analyzed for changes in fibrotic gene expression or TGF-ß signaling. Bleomycin-challenged mice treated with dual αvß6/αvß1 integrin inhibitor, PLN-74809, were assessed for changes in pulmonary collagen deposition and Smad3 phosphorylation. MEASUREMENTS AND MAIN RESULTS: Inhibition of integrins αvß6 and αvß1 was additive in reducing type I collagen gene expression in explanted lung tissue slices from patients with idiopathic pulmonary fibrosis. These data were replicated in fibrotic mouse lung tissue, with no added benefit observed from inhibition of additional αv integrins. Antifibrotic efficacy of dual αvß6/αvß1 integrin inhibitor PLN-74809 was confirmed in vivo, where dose-dependent inhibition of pulmonary Smad3 phosphorylation and collagen deposition was observed. PLN-74809 also, more potently, reduced collagen gene expression in fibrotic human and mouse lung slices than clinically relevant concentrations of nintedanib or pirfenidone. CONCLUSIONS: In the fibrotic lung, dual inhibition of integrins αvß6 and αvß1 offers the optimal approach for blocking fibrogenesis resulting from integrin-mediated activation of transforming growth factor-ß.


Asunto(s)
Antifibróticos/farmacología , Células Epiteliales/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Integrina alfa6beta1/antagonistas & inhibidores , Pulmón/efectos de los fármacos , Receptores de Vitronectina/antagonistas & inhibidores , Animales , Bleomicina , Línea Celular , Técnicas de Cocultivo , Cadena alfa 1 del Colágeno Tipo I/genética , Cadena alfa 1 del Colágeno Tipo I/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Integrina alfa6beta1/metabolismo , Pulmón/metabolismo , Pulmón/patología , Ratones Endogámicos C57BL , Fosforilación , Receptores de Vitronectina/metabolismo , Transducción de Señal , Proteína smad3/metabolismo
10.
Nature ; 507(7491): 190-4, 2014 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-24499815

RESUMEN

Alveoli are gas-exchange sacs lined by squamous alveolar type (AT) 1 cells and cuboidal, surfactant-secreting AT2 cells. Classical studies suggested that AT1 arise from AT2 cells, but recent studies propose other sources. Here we use molecular markers, lineage tracing and clonal analysis to map alveolar progenitors throughout the mouse lifespan. We show that, during development, AT1 and AT2 cells arise directly from a bipotent progenitor, whereas after birth new AT1 cells derive from rare, self-renewing, long-lived, mature AT2 cells that produce slowly expanding clonal foci of alveolar renewal. This stem-cell function is broadly activated by AT1 injury, and AT2 self-renewal is selectively induced by EGFR (epidermal growth factor receptor) ligands in vitro and oncogenic Kras(G12D) in vivo, efficiently generating multifocal, clonal adenomas. Thus, there is a switch after birth, when AT2 cells function as stem cells that contribute to alveolar renewal, repair and cancer. We propose that local signals regulate AT2 stem-cell activity: a signal transduced by EGFR-KRAS controls self-renewal and is hijacked during oncogenesis, whereas another signal controls reprogramming to AT1 fate.


Asunto(s)
Neoplasias Pulmonares/patología , Pulmón/citología , Pulmón/crecimiento & desarrollo , Células Madre Multipotentes/citología , Células Madre Multipotentes/patología , Alveolos Pulmonares/citología , Regeneración , Animales , Diferenciación Celular , División Celular , Linaje de la Célula , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Células Cultivadas , Reprogramación Celular , Células Clonales/citología , Receptores ErbB/metabolismo , Femenino , Pulmón/embriología , Pulmón/patología , Neoplasias Pulmonares/metabolismo , Masculino , Ratones , Modelos Biológicos , Células Madre Multipotentes/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Transducción de Señal
11.
Nature ; 509(7500): 371-5, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24739965

RESUMEN

The mammalian lung is a highly branched network in which the distal regions of the bronchial tree transform during development into a densely packed honeycomb of alveolar air sacs that mediate gas exchange. Although this transformation has been studied by marker expression analysis and fate-mapping, the mechanisms that control the progression of lung progenitors along distinct lineages into mature alveolar cell types are still incompletely known, in part because of the limited number of lineage markers and the effects of ensemble averaging in conventional transcriptome analysis experiments on cell populations. Here we show that single-cell transcriptome analysis circumvents these problems and enables direct measurement of the various cell types and hierarchies in the developing lung. We used microfluidic single-cell RNA sequencing (RNA-seq) on 198 individual cells at four different stages encompassing alveolar differentiation to measure the transcriptional states which define the developmental and cellular hierarchy of the distal mouse lung epithelium. We empirically classified cells into distinct groups by using an unbiased genome-wide approach that did not require a priori knowledge of the underlying cell types or the previous purification of cell populations. The results confirmed the basic outlines of the classical model of epithelial cell-type diversity in the distal lung and led to the discovery of many previously unknown cell-type markers, including transcriptional regulators that discriminate between the different populations. We reconstructed the molecular steps during maturation of bipotential progenitors along both alveolar lineages and elucidated the full life cycle of the alveolar type 2 cell lineage. This single-cell genomics approach is applicable to any developing or mature tissue to robustly delineate molecularly distinct cell types, define progenitors and lineage hierarchies, and identify lineage-specific regulatory factors.


Asunto(s)
Linaje de la Célula/genética , Células Epiteliales/citología , Células Epiteliales/metabolismo , Pulmón/citología , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Animales , Bronquios/citología , Diferenciación Celular/genética , Células Epiteliales/clasificación , Femenino , Marcadores Genéticos , Genoma/genética , Genómica , Pulmón/embriología , Ratones , Ratones Endogámicos C57BL , Alveolos Pulmonares/citología , Intercambio Gaseoso Pulmonar , Células Madre/citología , Transcriptoma/genética
12.
Dev Biol ; 430(1): 214-223, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28811219

RESUMEN

GW182 (also known asTNRC6) family members are critically involved in the final effector phase of miRNA-mediated mRNA repression. The three mammalian paralogs, TNRC6a, b and c, are thought to be redundant based on Argonaute (Ago) binding, tethering assays, and RNAi silencing of individual members in cell lines. To test this idea, we generated TNRC6a, b and c knockout mice. TNRC6a mutants die at mid-gestation, while b- and c- deleted mice are born at a Mendelian ratio. However, the majority of TNRC6b and all TNRC6c mutants die within 24h after birth, the latter with respiratory failure. Necropsy of TNRC6c mutants revealed normal-appearing airways that give rise to abnormally thick-walled distal gas exchange sacs. Immunohistological analysis of mutant lungs demonstrated a normal distribution of bronchiolar and alveolar cells, indicating that loss of TNRC6c did not abrogate epithelial cell differentiation. The cellular kinetics and relative proportions of endothelial, epithelial, and mesenchymal cells were also not altered. However, the underlying capillary network was simplified and endothelial cells had failed to become tightly apposed to the surface epithelium in TNRC6c mutants, presumably causing the observed respiratory failure. TGFß family mutant mice exhibit a similar lung phenotype of thick-walled air sacs and neonatal lethality, and qRT-PCR confirmed dynamic downregulation of TGFß1 and TGFßR2 in TNRC6c mutant lungs during sacculation. VEGFR, but not VEGF-A ligand, was also lower, likely reflecting the overall reduced capillary density in TNRC6c mutants. Together, these results demonstrate that GW182 paralogs are not functionally redundant in vivo. Surprisingly, despite regulating a general cellular process, TNRC6c is selectively required only in the distal lung and not until late in gestation for proper expression of the TGFß family genes that drive sacculation. These results imply a complex and indirect mode of regulation of sacculation by TNRC6c, mediated in part by dynamic transcriptional repression of an inhibitor of TGFß family gene expression.


Asunto(s)
Autoantígenos/metabolismo , Pulmón/irrigación sanguínea , Pulmón/embriología , Microvasos/embriología , Microvasos/metabolismo , Organogénesis , Proteínas de Unión al ARN/metabolismo , Repeticiones de Trinucleótidos/genética , Animales , Autoantígenos/genética , Diferenciación Celular , Células Epiteliales/citología , Células Epiteliales/metabolismo , Gases/metabolismo , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Células Germinativas/metabolismo , Pulmón/metabolismo , Mesodermo/embriología , Mesodermo/metabolismo , Ratones , Ratones Noqueados , Organogénesis/genética , Proteínas de Unión al ARN/genética , Reproducibilidad de los Resultados , Homología de Secuencia de Aminoácido , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Grabación en Video
13.
Exp Cell Res ; 340(1): 43-52, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26460176

RESUMEN

Hedgehog (Hh) pathway activation in R26-SmoM2;CAGGS-CreER mice, which carry a tamoxifen-inducible activated Smoothened allele (SmoM2), results in numerous microscopic tumor foci in mouse skeletal muscle. These tumors exhibit a highly differentiated myogenic phenotype and resemble human fetal rhabdomyomas. This study sought to apply previously established strategies to isolate lineally distinct populations of normal mouse myofiber-associated cells in order to examine cellular heterogeneity in SmoM2 tumors. We demonstrate that established SmoM2 tumors are composed of cells expressing myogenic, adipocytic and hematopoietic lineage markers and differentiation capacity. SmoM2 tumors thus recapitulate the phenotypic and functional hetereogeneity observed in normal mouse skeletal muscle. SmoM2 tumors also contain an expanded population of PAX7+ and MyoD+ satellite-like cells with extremely low clonogenic activity. Selective activation of Hh signaling in freshly isolated muscle satellite cells enhanced terminal myogenic differentiation without stimulating proliferation. Our findings support the conclusion that SmoM2 tumors represent an aberrant skeletal muscle state and demonstrate that, similar to normal muscle, myogenic tumors contain functionally distinct cell subsets, including cells lacking myogenic differentiation potential.


Asunto(s)
Proteínas Hedgehog/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Neoplasias de Tejido Muscular/metabolismo , Neoplasias de Tejido Muscular/patología , Alelos , Animales , Diferenciación Celular , Proliferación Celular , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Fenotipo
14.
Bioessays ; 37(9): 1028-37, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26201286

RESUMEN

The application of in vivo genetic lineage tracing has advanced our understanding of cellular mechanisms for tissue renewal in organs with slow turnover, like the lung. These studies have identified an adult stem cell with very different properties than classically understood ones that maintain continuously cycling tissues such as the intestine. A portrait has emerged of an ensemble of cellular programs that replenish the cells that line the gas exchange (alveolar) surface, enabling a response tailored to the extent of cell loss. A capacity for differentiated cells to undergo direct lineage transitions allows for local restoration of proper cell balance at sites of injury. We present these recent findings as a paradigm for how a relatively quiescent tissue compartment can maintain homeostasis throughout a lifetime punctuated by injuries ranging from mild to life-threatening, and discuss how dysfunction or insufficiency of alveolar repair programs produce serious health consequences like cancer and fibrosis.


Asunto(s)
Alveolos Pulmonares/citología , Alveolos Pulmonares/fisiología , Animales , Bronquios/fisiología , Gases/metabolismo , Humanos , Neoplasias/patología , Fenotipo , Células Madre/citología
15.
Curr Opin Pediatr ; 27(3): 341-7, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25888154

RESUMEN

PURPOSE OF REVIEW: Childhood interstitial lung diseases (ILDs) are a diverse class of disorders affecting the alveolar gas exchange region that lack specific treatments and are usually fatal. Here, we integrate recent insights into alveolar cell biology with histopathology from well characterized mutations of surfactant-associated genes. We take a reductionist approach by parsing discrete histological features and correlating each to perturbation of a particular function of the alveolar epithelial type II (AT2) cell, the central driver of disease, to generate a working model for the cellular mechanisms of disease pathogenesis. RECENT FINDINGS: The application of genetically modified mice and single cell genomics has yielded new insights into lung biology, including the identification of a bipotent alveolar progenitor in development, mapping of adult AT2 stem cells in vivo, and demonstration that latent cooperative interactions with fibroblasts can be pathologically activated by targeted injury of the AT2 cell. SUMMARY: As we learn more about individual and cooperative roles for alveolar cells in health, we can dissect how perturbations of specific cellular functions contribute to disease in childhood ILDs. We hope our updated model centered around the AT2 cell as the initiator of disease provides a cellular framework that researchers can build upon and revise as they identify the specific molecular signals within and between alveolar cells that mediate the diverse pathologic features, so that targeted pharmacologic and cell-based treatments for patients can ultimately be engineered.


Asunto(s)
Enfermedades Pulmonares Intersticiales/patología , Pulmón/patología , Alveolos Pulmonares/patología , Animales , Diferenciación Celular , Células Cultivadas , Reprogramación Celular , Niño , Modelos Animales de Enfermedad , Humanos , Pulmón/citología , Enfermedades Pulmonares Intersticiales/genética , Ratones , Alveolos Pulmonares/citología , Transducción de Señal
16.
Am J Emerg Med ; 33(7): 992.e3-6, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25684743

RESUMEN

Electrical injuries are most commonly due to household accidents.Various factors determine the severity of electric injury, including type of current, amperage, voltage, tissue resistance, pathway of current,and duration of contact with the body. Various types of neurologic damage due to electrical injury have been described in literature. It may manifest as peripheral nerve injury, spinal cord damage, seizures, cerebellarataxia, hypoxic encephalopathy, and intracerebral hemorrhage. Acute ischemic stroke is an infrequent complication of electrical injury. Herein,we report a case of middle-aged man, who accidentally sustained high voltage electrical injury followed by acute vertebrobasilar ischemic stroke. Magnetic resonance imaging of the brain showed acute infarctin bilateral cerebellar and medial occipital regions. Computed tomographic angiogram of the brain and neck vessels was normal. Possibly,in our patient, the mechanism could be related to direct vascular injury due to electric current.


Asunto(s)
Traumatismos por Electricidad/complicaciones , Accidente Cerebrovascular/etiología , Insuficiencia Vertebrobasilar/etiología , Enfermedad Aguda , Adulto , Humanos , Masculino , Accidente Cerebrovascular/diagnóstico , Insuficiencia Vertebrobasilar/diagnóstico
19.
J Cell Biol ; 222(12)2023 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-37843535

RESUMEN

With each breath, oxygen diffuses across remarkably thin alveolar type I (AT1) cells into underlying capillaries. Interspersed cuboidal AT2 cells produce surfactant and act as stem cells. Even transient disruption of this delicate barrier can promote capillary leak. Here, we selectively ablated AT1 cells, which uncovered rapid AT2 cell flattening with near-continuous barrier preservation, culminating in AT1 differentiation. Proliferation subsequently restored depleted AT2 cells in two phases, mitosis of binucleated AT2 cells followed by replication of mononucleated AT2 cells. M phase entry of binucleated and S phase entry of mononucleated cells were both triggered by AT1-produced hbEGF signaling via EGFR to Wnt-active AT2 cells. Repeated AT1 cell killing elicited exuberant AT2 proliferation, generating aberrant daughter cells that ceased surfactant function yet failed to achieve AT1 differentiation. This hyperplasia eventually resolved, yielding normal-appearing alveoli. Overall, this specialized regenerative program confers a delicate simple epithelium with functional resiliency on par with the physical durability of thicker, pseudostratified, or stratified epithelia.


Asunto(s)
Pulmón , Células Madre , Tensoactivos , Diferenciación Celular , División Celular , Células Cultivadas , Células Madre/citología
20.
Exp Mol Med ; 55(8): 1831-1842, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37582976

RESUMEN

We present an in-depth single-cell atlas of in vitro multiculture systems on human primary airway epithelium derived from normal and diseased lungs of 27 individual donors. Our large-scale single-cell profiling identified new cell states and differentiation trajectories of rare airway epithelial cell types in human distal lungs. By integrating single-cell datasets of human lung tissues, we discovered immune-primed subsets enriched in lungs and organoids derived from patients with chronic respiratory disease. To demonstrate the full potential of our platform, we further illustrate transcriptomic responses to various respiratory virus infections in vitro airway models. Our work constitutes a single-cell roadmap for the cellular and molecular characteristics of human primary lung cells in vitro and their relevance to human tissues in vivo.


Asunto(s)
Células Epiteliales , Pulmón , Humanos , Células Epiteliales/metabolismo , Epitelio , Diferenciación Celular/fisiología , Organoides
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda