Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 156(3): 440-55, 2014 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-24485453

RESUMEN

Lung stem cells are instructed to produce lineage-specific progeny through unknown factors in their microenvironment. We used clonal 3D cocultures of endothelial cells and distal lung stem cells, bronchioalveolar stem cells (BASCs), to probe the instructive mechanisms. Single BASCs had bronchiolar and alveolar differentiation potential in lung endothelial cell cocultures. Gain- and loss-of-function experiments showed that BMP4-Bmpr1a signaling triggers calcineurin/NFATc1-dependent expression of thrombospondin-1 (Tsp1) in lung endothelial cells to drive alveolar lineage-specific BASC differentiation. Tsp1 null mice exhibited defective alveolar injury repair, confirming a crucial role for the BMP4-NFATc1-TSP1 axis in lung epithelial differentiation and regeneration in vivo. Discovery of this pathway points to methods to direct the derivation of specific lung epithelial lineages from multipotent cells. These findings elucidate a pathway that may be a critical target in lung diseases and provide tools to understand the mechanisms of respiratory diseases at the single-cell level.


Asunto(s)
Bronquiolos/citología , Diferenciación Celular , Células Endoteliales/metabolismo , Alveolos Pulmonares/citología , Transducción de Señal , Células Madre/metabolismo , Animales , Proteína Morfogenética Ósea 4/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Bronquiolos/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Ratones , Factores de Transcripción NFATC/metabolismo , Alveolos Pulmonares/metabolismo , Células Madre/citología , Trombospondina 1/genética , Trombospondina 1/metabolismo
2.
Am J Respir Cell Mol Biol ; 70(3): 159-164, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38207122

RESUMEN

Efferocytosis is a process whereby apoptotic cells are cleared to maintain tissue homeostasis. In the lungs, efferocytosis has been implicated in several acute and chronic inflammatory diseases. A long-standing method to study efferocytosis in vivo is to instill apoptotic cells into the lungs to evaluate macrophage uptake. However, this approach provides nonphysiologic levels of cells to the airspaces, where there is preferential access to the alveolar macrophages. To circumvent this limitation, we developed a new method to study efferocytosis of damaged alveolar type 2 (AT2) epithelial cells in vivo. A reporter mouse that expresses TdTomato in AT2 epithelial cells was injured with influenza (strain PR8) to induce apoptosis of AT2 cells. We were able to identify macrophages that acquire red fluorescence after influenza injury, indicating efferocytosis of AT2 cells. Furthermore, evaluation of macrophage populations led to the surprising finding that lung interstitial macrophages were the primary efferocyte in vivo. In summary, we present a novel finding that the interstitial macrophage, not the alveolar macrophage, primarily mediates clearance of AT2 cells in the lungs after influenza infection. Our method of studying efferocytosis provides a more physiologic approach in evaluating the spatiotemporal dynamics of apoptotic cell clearance in vivo and opens new avenues to study the mechanisms by which efferocytosis regulates inflammation.


Asunto(s)
Eferocitosis , Gripe Humana , Proteína Fluorescente Roja , Animales , Ratones , Humanos , Macrófagos , Epitelio
3.
Am J Physiol Cell Physiol ; 325(2): C483-C495, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37458437

RESUMEN

Pulmonary fibrosis comprises a range of chronic interstitial lung diseases (ILDs) that impose a significant burden on patients and public health. Among these, idiopathic pulmonary fibrosis (IPF), a disease of aging, is the most common and most severe form of ILD and is treated largely by lung transplantation. The lack of effective treatments to stop or reverse lung fibrosis-in fact, fibrosis in most organs-has sparked the need to understand causative mechanisms with the goal of identifying critical points for potential therapeutic intervention. Findings from many groups have indicated that repeated injury to the alveolar epithelium-where gas exchange occurs-leads to stem cell exhaustion and impaired alveolar repair that, in turn, triggers the onset and progression of fibrosis. Cellular senescence of alveolar epithelial progenitors is a critical cause of stemness failure. Hence, senescence impairs repair and thus contributes significantly to fibrosis. In this review, we discuss recent evidence indicating that senescence of epithelial progenitor cells impairs alveolar homeostasis and repair creating a profibrotic environment. Moreover, we discuss the impact of senescent alveolar epithelial progenitors, alveolar type 2 (AT2) cells, and AT2-derived transitional epithelial cells in fibrosis. Emerging evidence indicates that transitional epithelial cells are prone to senescence and, hence, are a new player involved in senescence-associated lung fibrosis. Understanding the complex interplay of cell types and cellular regulatory factors contributing to alveolar epithelial progenitor senescence will be crucial to developing targeted therapies to mitigate their downstream profibrotic sequelae and to promote normal alveolar repair.NEW & NOTEWORTHY With an aging population, lung fibrotic diseases are becoming a global health burden. Dysfunctional repair of the alveolar epithelium is a key causative process that initiates lung fibrosis. Normal alveolar regeneration relies on functional progenitor cells; however, the senescence of these cells, which increases with age, hinders their ability to contribute to repair. Here, we discuss studies on the control and consequence of progenitor cell senescence in fibrosis and opportunities for research.


Asunto(s)
Células Epiteliales Alveolares , Fibrosis Pulmonar Idiopática , Humanos , Anciano , Células Epiteliales Alveolares/metabolismo , Fibrosis Pulmonar Idiopática/metabolismo , Senescencia Celular , Envejecimiento , Células Madre/metabolismo , Células Epiteliales/metabolismo , Pulmón/metabolismo
4.
J Pathol ; 257(4): 494-500, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35608561

RESUMEN

The human lung is a relatively quiescent organ in the normal healthy state but contains stem/progenitor cells that contribute to normal tissue maintenance and either repair or remodeling in response to injury and disease. Maintenance or repair lead to proper restoration of functional lung tissue and maintenance of physiological functions, with remodeling resulting in altered structure and function that is typically associated with disease. Knowledge of cell types contributing to lung tissue maintenance and repair/remodeling have largely relied on mouse models of injury-repair and lineage tracing of local progenitors. Therefore, many of the functional alterations underlying remodeling in human lung disease have remained poorly defined. However, the advent of advanced genomics approaches to define the molecular phenotype of lung cells at single-cell resolution has paved the way for rapid advances in our understanding of cell types present within the normal human lung and changes that accompany disease. Here we summarize recent advances in our understanding of disease-related changes in the molecular phenotype of human lung epithelium that have emerged from single-cell transcriptomic studies. We focus attention on emerging concepts of epithelial transitional states that characterize the pathological remodeling that accompanies chronic lung diseases, including idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, cystic fibrosis, and asthma. Concepts arising from these studies are actively evolving and require corroborative studies to improve our understanding of disease mechanisms. Whenever possible, we highlight opportunities for providing a unified nomenclature in this rapidly advancing field of research. © 2022 The Pathological Society of Great Britain and Ireland.


Asunto(s)
Fibrosis Pulmonar Idiopática , Enfermedades Pulmonares , Enfermedad Pulmonar Obstructiva Crónica , Animales , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/patología , Enfermedades Pulmonares/patología , Ratones , Enfermedad Pulmonar Obstructiva Crónica/patología , Transcriptoma
5.
Am J Respir Cell Mol Biol ; 67(6): 623-631, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36036918

RESUMEN

The epithelium lining airspaces of the human lung is maintained by regional stem cells, including basal cells of pseudostratified airways and alveolar type 2 (AT2) pneumocytes of the gas-exchange region. Despite effective techniques for long-term preservation of airway basal cells, procedures for efficient preservation of functional epithelial cell types of the distal gas-exchange region are lacking. Here we detail a method for cryobanking of epithelial cells from either mouse or human lung tissue for preservation of their phenotypic and functional characteristics. Flow cytometric profiling, epithelial organoid-forming efficiency, and single-cell transcriptomic analysis were used to compare cells recovered from cryobanked tissue with those of freshly dissociated tissue. AT2 cells within single-cell suspensions of enzymatically digested cryobanked distal lung tissue retained expression of the pan-epithelial marker CD326 and the AT2 cell surface antigen recognized by monoclonal antibody HT II-280, allowing antibody-mediated enrichment and downstream analysis. Isolated AT2 cells from cryobanked tissue were comparable with those of freshly dissociated tissue both in their single-cell transcriptome and their capacity for in vitro organoid formation in three-dimensional cultures. We conclude that the cryobanking method described herein allows long-term preservation of distal human lung tissue for downstream analysis of lung cell function and molecular phenotype and is ideally suited for the creation of an easily accessible tissue resource for the research community.


Asunto(s)
Células Epiteliales , Pulmón , Humanos , Ratones , Animales , Diferenciación Celular/fisiología , Células Epiteliales/metabolismo , Células Epiteliales Alveolares/metabolismo , Fenotipo
6.
Am J Transplant ; 22(2): 565-573, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34464505

RESUMEN

Despite the common detection of non-donor specific anti-HLA antibodies (non-DSAs) after lung transplantation, their clinical significance remains unclear. In this retrospective single-center cohort study of 325 lung transplant recipients, we evaluated the association between donor-specific HLA antibodies (DSAs) and non-DSAs with subsequent CLAD development. DSAs were detected in 30% of recipients and were associated with increased CLAD risk, with higher HRs for both de novo and high MFI (>5000) DSAs. Non-DSAs were detected in 56% of recipients, and 85% of DSA positive tests had concurrent non-DSAs. In general, non-DSAs did not increase CLAD risk in multivariable models accounting for DSAs. However, non-DSAs in conjunction with high BAL CXCL9 levels were associated with increased CLAD risk. Multivariable proportional hazards models demonstrate the importance of the HLA antibody-CXCL9 interaction: CLAD risk increases when HLA antibodies (both DSAs and non-DSAs) are detected in conjunction with high CXCL9. Conversely, CLAD risk is not increased when HLA antibodies are detected with low CXCL9. This study supports the potential utility of BAL CXCL9 measurement as a biomarker to risk stratify HLA antibodies for future CLAD. The ability to discriminate between high versus low-risk HLA antibodies may improve management by allowing for guided treatment decisions.


Asunto(s)
Antígenos HLA , Trasplante de Pulmón , Aloinjertos , Biomarcadores , Quimiocina CXCL9 , Estudios de Cohortes , Rechazo de Injerto/diagnóstico , Rechazo de Injerto/etiología , Supervivencia de Injerto , Humanos , Isoanticuerpos , Trasplante de Pulmón/efectos adversos , Pronóstico , Estudios Retrospectivos , Donantes de Tejidos
7.
Eur Respir J ; 60(2)2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35086840

RESUMEN

BACKGROUND: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection of the respiratory system can progress to a multisystemic disease with aberrant inflammatory response. Cellular senescence promotes chronic inflammation, named senescence-associated secretory phenotype (SASP). We investigated whether coronavirus disease 2019 (COVID-19) is associated with cellular senescence and SASP. METHODS: Autopsy lung tissue samples from 11 COVID-19 patients and 43 age-matched non-COVID-19 controls with similar comorbidities were analysed by immunohistochemistry for SARS-CoV-2, markers of senescence and key SASP cytokines. Virally induced senescence was functionally recapitulated in vitro, by infecting epithelial Vero-E6 cells and a three-dimensional alveosphere system of alveolar type 2 (AT2) cells with SARS-CoV-2 strains isolated from COVID-19 patients. RESULTS: SARS-CoV-2 was detected by immunocytochemistry and electron microscopy predominantly in AT2 cells. Infected AT2 cells expressed angiotensin-converting enzyme 2 and exhibited increased senescence (p16INK4A and SenTraGor positivity) and interleukin (IL)-1ß and IL-6 expression. In vitro, infection of Vero-E6 cells with SARS-CoV-2 induced senescence (SenTraGor), DNA damage (γ-H2AX) and increased cytokine (IL-1ß, IL-6, CXCL8) and apolipoprotein B mRNA-editing (APOBEC) enzyme expression. Next-generation sequencing analysis of progenies obtained from infected/senescent Vero-E6 cells demonstrated APOBEC-mediated SARS-CoV-2 mutations. Dissemination of the SARS-CoV-2-infection and senescence was confirmed in extrapulmonary sites (kidney and liver) of a COVID-19 patient. CONCLUSIONS: We demonstrate that in severe COVID-19, AT2 cells infected by SARS-CoV-2 exhibit senescence and a proinflammatory phenotype. In vitro, SARS-CoV-2 infection induces senescence and inflammation. Importantly, infected senescent cells may act as a source of SARS-CoV-2 mutagenesis mediated by APOBEC enzymes. Therefore, SARS-CoV-2-induced senescence may be an important molecular mechanism of severe COVID-19, disease persistence and mutagenesis.


Asunto(s)
COVID-19 , SARS-CoV-2 , Senescencia Celular , Citocinas/metabolismo , Humanos , Inflamación , Interleucina-6 , Pulmón/metabolismo , Mutagénesis , Fenotipo
8.
Am J Respir Crit Care Med ; 203(6): 707-717, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-32991815

RESUMEN

Rationale: Idiopathic pulmonary fibrosis (IPF) is an insidious and fatal interstitial lung disease associated with declining pulmonary function. Accelerated aging, loss of epithelial progenitor cell function and/or numbers, and cellular senescence are implicated in the pathogenies of IPF.Objectives: We sought to investigate the role of alveolar type 2 (AT2) cellular senescence in initiation and/or progression of pulmonary fibrosis and therapeutic potential of targeting senescence-related pathways and senescent cells.Methods: Epithelial cells of 9 control donor proximal and distal lung tissues and 11 IPF fibrotic lung tissues were profiled by single-cell RNA sequencing to assesses the contribution of epithelial cells to the senescent cell fraction for IPF. A novel mouse model of conditional AT2 cell senescence was generated to study the role of cellular senescence in pulmonary fibrosis.Measurements and Main Results: We show that AT2 cells isolated from IPF lung tissue exhibit characteristic transcriptomic features of cellular senescence. We used conditional loss of Sin3a in adult mouse AT2 cells to initiate a program of p53-dependent cellular senescence, AT2 cell depletion, and spontaneous, progressive pulmonary fibrosis. We establish that senescence rather than loss of AT2 cells promotes progressive fibrosis and show that either genetic or pharmacologic interventions targeting p53 activation or senescence block fibrogenesis.Conclusions: Senescence of AT2 cells is sufficient to drive progressive pulmonary fibrosis. Early attenuation of senescence-related pathways and elimination of senescent cells are promising therapeutic approaches to prevent pulmonary fibrosis.


Asunto(s)
Envejecimiento/patología , Células Epiteliales Alveolares/patología , Senescencia Celular , Fibrosis Pulmonar Idiopática/patología , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino
9.
Am J Respir Crit Care Med ; 203(10): 1275-1289, 2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-33321047

RESUMEN

Rationale: Identification of the specific cell types expressing CFTR (cystic fibrosis [CF] transmembrane conductance regulator) is required for precision medicine therapies for CF. However, a full characterization of CFTR expression in normal human airway epithelia is missing. Objectives: To identify the cell types that contribute to CFTR expression and function within the proximal-distal axis of the normal human lung. Methods: Single-cell RNA (scRNA) sequencing (scRNA-seq) was performed on freshly isolated human large and small airway epithelial cells. scRNA in situ hybridization (ISH) and single-cell qRT-PCR were performed for validation. In vitro culture systems correlated CFTR function with cell types. Lentiviruses were used for cell type-specific transduction of wild-type CFTR in CF cells. Measurements and Main Results: scRNA-seq identified secretory cells as dominating CFTR expression in normal human large and, particularly, small airway superficial epithelia, followed by basal cells. Ionocytes expressed the highest CFTR levels but were rare, whereas the expression in ciliated cells was infrequent and low. scRNA ISH and single-cell qRT-PCR confirmed the scRNA-seq findings. CF lungs exhibited distributions of CFTR and ionocytes similar to those of normal control subjects. CFTR mediated Cl- secretion in cultures tracked secretory cell, but not ionocyte, densities. Furthermore, the nucleotide-purinergic regulatory system that controls CFTR-mediated hydration was associated with secretory cells and not with ionocytes. Lentiviral transduction of wild-type CFTR produced CFTR-mediated Cl- secretion in CF airway secretory cells but not in ciliated cells. Conclusions: Secretory cells dominate CFTR expression and function in human airway superficial epithelia. CFTR therapies may need to restore CFTR function to multiple cell types, with a focus on secretory cells.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Fibrosis Quística/metabolismo , Fibrosis Quística/patología , Células Epiteliales/metabolismo , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Estudios de Casos y Controles , Técnicas de Cultivo de Célula , Humanos
10.
Am J Respir Cell Mol Biol ; 65(1): 22-29, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33625958

RESUMEN

The National Heart, Lung, and Blood Institute of the National Institutes of Health, together with the Longfonds BREATH consortium, convened a working group to review the field of lung regeneration and suggest avenues for future research. The meeting took place on May 22, 2019, at the American Thoracic Society 2019 conference in Dallas, Texas, United States, and brought together investigators studying lung development, adult stem-cell biology, induced pluripotent stem cells, biomaterials, and respiratory disease. The purpose of the working group was 1) to examine the present status of basic science approaches to tackling lung disease and promoting lung regeneration in patients and 2) to determine priorities for future research in the field.


Asunto(s)
Células Madre Pluripotentes Inducidas , Enfermedades Pulmonares , Pulmón/fisiología , Regeneración , Mucosa Respiratoria/fisiología , Animales , Tratamiento Basado en Trasplante de Células y Tejidos , Congresos como Asunto , Educación , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/trasplante , Enfermedades Pulmonares/metabolismo , Enfermedades Pulmonares/terapia , National Heart, Lung, and Blood Institute (U.S.) , Estados Unidos
11.
Am J Respir Crit Care Med ; 202(11): 1540-1550, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32692579

RESUMEN

Rationale: Declining lung function in patients with interstitial lung disease is accompanied by epithelial remodeling and progressive scarring of the gas-exchange region. There is a need to better understand the contribution of basal cell hyperplasia and associated mucosecretory dysfunction to the development of idiopathic pulmonary fibrosis (IPF).Objectives: We sought to decipher the transcriptome of freshly isolated epithelial cells from normal and IPF lungs to discern disease-dependent changes within basal stem cells.Methods: Single-cell RNA sequencing was used to map epithelial cell types of the normal and IPF human airways. Organoid and air-liquid interface cultures were used to investigate functional properties of basal cell subtypes.Measurements and Main Results: We found that basal cells included multipotent and secretory primed subsets in control adult lung tissue. Secretory primed basal cells include an overlapping molecular signature with basal cells obtained from the distal lung tissue of IPF lungs. We confirmed that NOTCH2 maintains undifferentiated basal cells and restricts basal-to-ciliated differentiation, and we present evidence that NOTCH3 functions to restrain secretory differentiation.Conclusions: Basal cells are dynamically regulated in disease and are specifically biased toward the expansion of the secretory primed basal cell subset in IPF. Modulation of basal cell plasticity may represent a relevant target for therapeutic intervention in IPF.


Asunto(s)
Plasticidad de la Célula , Proliferación Celular/genética , Autorrenovación de las Células/genética , Células Epiteliales/citología , Fibrosis Pulmonar Idiopática/genética , Mucosa Respiratoria/citología , Anciano , Células Epiteliales Alveolares/citología , Células Epiteliales Alveolares/metabolismo , Membrana Basal , Estudios de Casos y Controles , Células Epiteliales/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Masculino , Persona de Mediana Edad , RNA-Seq , Mucosa Respiratoria/metabolismo , Análisis de la Célula Individual , Transcriptoma , Adulto Joven
12.
Development ; 144(14): 2618-2628, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28619823

RESUMEN

Mechanisms that regulate tissue-specific progenitors for maintenance and differentiation during development are poorly understood. Here, we demonstrate that the co-repressor protein Sin3a is crucial for lung endoderm development. Loss of Sin3a in mouse early foregut endoderm led to a specific and profound defect in lung development with lung buds failing to undergo branching morphogenesis and progressive atrophy of the proximal lung endoderm with complete epithelial loss at later stages of development. Consequently, neonatal pups died at birth due to respiratory insufficiency. Further analysis revealed that loss of Sin3a resulted in embryonic lung epithelial progenitor cells adopting a senescence-like state with permanent cell cycle arrest in G1 phase. This was mediated at least partially through upregulation of the cell cycle inhibitors Cdkn1a and Cdkn2c. At the same time, loss of endodermal Sin3a also disrupted cell differentiation of the mesoderm, suggesting aberrant epithelial-mesenchymal signaling. Together, these findings reveal that Sin3a is an essential regulator for early lung endoderm specification and differentiation.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Pulmón/embriología , Pulmón/metabolismo , Proteínas Represoras/metabolismo , Animales , Animales Recién Nacidos , Puntos de Control del Ciclo Celular , Diferenciación Celular , Linaje de la Célula/genética , Linaje de la Célula/fisiología , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Endodermo/citología , Endodermo/embriología , Endodermo/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Pulmón/citología , Ratones , Ratones Noqueados , Organogénesis/genética , Organogénesis/fisiología , Embarazo , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Transducción de Señal , Complejo Correpresor Histona Desacetilasa y Sin3
13.
Am J Respir Crit Care Med ; 200(2): 220-234, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-30973754

RESUMEN

Rationale: The goal was to connect elements of idiopathic pulmonary fibrosis (IPF) pathogenesis, including chronic endoplasmic reticulum stress in respiratory epithelia associated with injury/inflammation and remodeling, distal airway mucus obstruction and honeycomb cyst formation with accumulation of MUC5B (mucin 5B), and associations between IPF risk and polymorphisms in the MUC5B promoter. Objectives: To test whether the endoplasmic reticulum (ER) stress sensor protein ERN2 (ER-to-nucleus signaling 2) and its downstream effector, the spliced form of XBP1S (X-box-binding protein 1), regulate MUC5B expression and differentially activate the MUC5B promoter variant in respiratory epithelia. Methods: Primary human airway epithelial (HAE) cells, transgenic mouse models, human IPF lung tissues, and cell lines expressing XBP1S and MUC5B promoters were used to explore relationships between the ERN2/XBP1S pathway and MUC5B. An inhibitor of the pathway, KIRA6, and XBP1 CRISPR-Cas9 were used in HAE cells to explore therapeutic potential. Measurements and Main Results: ERN2 regulated MUC5B and MUC5AC mRNAs. Downstream XBP1S selectively promoted MUC5B expression in vitro and in distal murine airway epithelia in vivo. XBP1S bound to the proximal region of the MUC5B promoter and differentially upregulated MUC5B expression in the context of the MUC5B promoter rs35705950 variant. High levels of ERN2 and XBP1S were associated with excessive MUC5B mRNAs in distal airways of human IPF lungs. Cytokine-induced MUC5B expression in HAE cells was inhibited by KIRA6 and XBP1 CRISPR-Cas9. Conclusions: A positive feedback bistable ERN2-XBP1S pathway regulates MUC5B-dominated mucus obstruction in IPF, providing an unfolded protein response-dependent mechanism linking the MUC5B promoter rs35705950 polymorphism with IPF pathogenesis. Inhibiting ERN2-dependent pathways/elements may provide a therapeutic option for IPF.


Asunto(s)
Endorribonucleasas/genética , Fibrosis Pulmonar Idiopática/genética , Proteínas de la Membrana/genética , Proteínas Serina-Treonina Quinasas/genética , Mucosa Respiratoria/metabolismo , Proteína 1 de Unión a la X-Box/genética , Animales , Sistemas CRISPR-Cas , Línea Celular , Estrés del Retículo Endoplásmico/genética , Endorribonucleasas/metabolismo , Regulación de la Expresión Génica , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Polimorfismo Genético , Cultivo Primario de Células , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína 1 de Unión a la X-Box/metabolismo
14.
Int J Mol Sci ; 21(7)2020 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-32218238

RESUMEN

: Alveolar epithelial type II cells (AT2) are a heterogeneous population that have critical secretory and regenerative roles in the alveolus to maintain lung homeostasis. However, impairment to their normal functional capacity and development of a pro-fibrotic phenotype has been demonstrated to contribute to the development of idiopathic pulmonary fibrosis (IPF). A number of factors contribute to AT2 death and dysfunction. As a mucosal surface, AT2 cells are exposed to environmental stresses that can have lasting effects that contribute to fibrogenesis. Genetical risks have also been identified that can cause AT2 impairment and the development of lung fibrosis. Furthermore, aging is a final factor that adds to the pathogenic changes in AT2 cells. Here, we will discuss the homeostatic role of AT2 cells and the studies that have recently defined the heterogeneity of this population of cells. Furthermore, we will review the mechanisms of AT2 death and dysfunction in the context of lung fibrosis.


Asunto(s)
Células Epiteliales Alveolares , Fibrosis Pulmonar Idiopática , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Animales , Muerte Celular , Autorrenovación de las Células , Fibroblastos , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Transducción de Señal
16.
J Cell Sci ; 125(Pt 4): 932-42, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22421361

RESUMEN

Wnt-ß-catenin signaling regulates cell fate during organ development and postnatal tissue maintenance, but its contribution to specification of distinct lung epithelial lineages is still unclear. To address this question, we used a Cre recombinase (Cre)-LoxP approach to activate canonical Wnt signaling ectopically in developing lung endoderm. We found that persistent activation of canonical Wnt signaling within distal lung endoderm was permissive for normal development of alveolar epithelium, yet led to the loss of developing bronchiolar epithelium and ectasis of distal conducting airways. Activation of canonical Wnt led to ectopic expression of a lymphoid-enhancing factor and a T-cell factor (LEF and TCF, respectively) and absence of SRY (sex-determining region Y)-box 2 (SOX2) and tumor protein p63 (p63) expression in proximal derivatives. Conditional loss of SOX2 in airways phenocopied epithelial differentiation defects observed with ectopic activation of canonical Wnt. Our data suggest that Wnt negatively regulates a SOX2-dependent signaling program required for developmental progression of the bronchiolar lineage.


Asunto(s)
Células Epiteliales/citología , Células Epiteliales/metabolismo , Pulmón/citología , Factores de Transcripción SOXB1/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo , Animales , Apoptosis , Bronquiolos/citología , Bronquiolos/metabolismo , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Endodermo/metabolismo , Femenino , Regulación de la Expresión Génica , Genes Reporteros , Pulmón/metabolismo , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Masculino , Ratones , Ratones Transgénicos , Fosfoproteínas/metabolismo , Estabilidad Proteica , Alveolos Pulmonares/citología , Alveolos Pulmonares/metabolismo , Factor de Transcripción SOX9/metabolismo , Factores de Transcripción SOXB1/deficiencia , Factores de Transcripción TCF/metabolismo , Transactivadores/metabolismo , Transcripción Genética , Proteínas Wnt/metabolismo , beta Catenina/biosíntesis , beta Catenina/genética
17.
Am J Respir Cell Mol Biol ; 48(3): 288-98, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23204392

RESUMEN

The regeneration of alveolar epithelial cells is a critical aspect of alveolar reorganization after lung injury. Although alveolar Type II (AT2) cells have been described as progenitor cells for alveolar epithelia, more remains to be understood about how their progenitor cell properties are regulated. A nuclear, chromatin-bound green fluorescence protein reporter (H2B-GFP) was driven from the murine surfactant protein-C (SPC) promoter to generate SPC H2B-GFP transgenic mice. The SPC H2B-GFP allele allowed the FACS-based enrichment and gene expression profiling of AT2 cells. Approximately 97% of AT2 cells were GFP-labeled on Postnatal Day 1, and the percentage of GFP-labeled AT2 cells decreased to approximately 63% at Postnatal Week 8. Isolated young adult SPC H2B-GFP(+) cells displayed proliferation, differentiation, and self-renewal capacity in the presence of lung fibroblasts in a Matrigel-based three-dimensional culture system. Heterogeneity within the GFP(+) population was revealed, because cells with distinct alveolar and bronchiolar gene expression arose in three-dimensional cultures. CD74, a surface marker highly enriched on GFP(+) cells, was identified as a positive selection marker, providing 3-fold enrichment for AT2 cells. In vivo, GFP expression was induced within other epithelial cell types during maturation of the distal lung. The utility of the SPC H2B-GFP murine model for the identification of AT2 cells was greatest in early postnatal lungs and more limited with age, when some discordance between SPC and GFP expression was observed. In adult mice, this allele may allow for the enrichment and future characterization of other SPC-expressing alveolar and bronchiolar cells, including putative stem/progenitor cell populations.


Asunto(s)
Cromatina/metabolismo , Células Epiteliales/metabolismo , Fibroblastos/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Pulmón/metabolismo , Proteína C Asociada a Surfactante Pulmonar/metabolismo , Alelos , Animales , Antígenos de Diferenciación de Linfocitos B/genética , Antígenos de Diferenciación de Linfocitos B/metabolismo , Bronquiolos/citología , Bronquiolos/metabolismo , Diferenciación Celular/genética , Procesos de Crecimiento Celular/genética , Células Cultivadas , Cromatina/genética , Células Epiteliales/citología , Femenino , Fibroblastos/citología , Expresión Génica , Perfilación de la Expresión Génica/métodos , Proteínas Fluorescentes Verdes/genética , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/metabolismo , Pulmón/citología , Lesión Pulmonar/genética , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas , Alveolos Pulmonares/citología , Alveolos Pulmonares/metabolismo , Proteína C Asociada a Surfactante Pulmonar/biosíntesis , Proteína C Asociada a Surfactante Pulmonar/genética , Regeneración/genética , Mucosa Respiratoria/citología , Mucosa Respiratoria/metabolismo
18.
Stem Cells ; 30(9): 1948-60, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22696116

RESUMEN

Mechanisms that regulate regional epithelial cell diversity and pathologic remodeling in airways are poorly understood. We hypothesized that regional differences in cell composition and injury-related tissue remodeling result from the type and composition of local progenitors. We used surface markers and the spatial expression pattern of an SFTPC-GFP transgene to subset epithelial progenitors by airway region. Green fluorescent protein (GFP) expression ranged from undetectable to high in a proximal-to-distal gradient. GFP(hi) cells were subdivided by CD24 staining into alveolar (CD24(neg)) and conducting airway (CD24(low)) populations. This allowed for the segregation of three types of progenitors displaying distinct clonal behavior in vitro. GFP(neg) and GFP(low) progenitors both yielded lumen containing colonies but displayed transcriptomes reflective of pseudostratified and distal conducting airways, respectively. CD24(low)GFP(hi) progenitors were present in an overlapping distribution with GFP(low) progenitors in distal airways, yet expressed lower levels of Sox2 and expanded in culture to yield undifferentiated self-renewing progeny. Colony-forming ability was reduced for each progenitor cell type after in vivo bleomycin exposure, but only CD24(low) GFP(hi) progenitors showed robust expansion during tissue remodeling. These data reveal intrinsic differences in the properties of regional progenitors and suggest that their unique responses to tissue damage drive local tissue remodeling.


Asunto(s)
Lesión Pulmonar/patología , Células Madre/efectos de los fármacos , Células Madre/patología , Animales , Bleomicina , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Análisis por Micromatrices , Sistema Respiratorio/efectos de los fármacos , Sistema Respiratorio/patología , Células Madre/metabolismo , Uteroglobina/biosíntesis
19.
J Immunol ; 187(1): 382-90, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21632717

RESUMEN

Epithelial antimicrobial activity may protect the lung against inhaled pathogens. The bactericidal/permeability-increasing protein family has demonstrated antimicrobial activity in vitro. PLUNC (palate, lung, and nasal epithelium associated) is a 25-kDa secreted protein that shares homology with bactericidal/permeability-increasing proteins and is expressed in nasopharyngeal and respiratory epithelium. The objective of this study was to determine whether PLUNC can limit Pseudomonas aeruginosa infection in mice. Transgenic mice (Scgb1a1-hPLUNC) were generated in which human PLUNC (hPLUNC) was directed to the airway epithelium with the Scgb1a1 promoter. The hPLUNC protein (hPLUNC) was detected in the epithelium throughout the trachea and bronchial airways and in bronchoalveolar lavage fluid. Bronchoalveolar lavage fluid from transgenic mice exhibited higher antibacterial activity than that from wild type littermates in vitro. After in vivo P. aeruginosa challenge, Scgb1a1-hPLUNC transgenic mice displayed enhanced bacterial clearance. This was accompanied by a decrease in neutrophil infiltration and cytokine levels. More importantly, the overexpressed hPLUNC in Scgb1a1-hPLUNC transgenic mouse airway significantly enhanced mouse survival against P. aeruginosa-induced respiratory infection. These data indicate that PLUNC is a novel antibacterial protein that likely plays a critical role in airway epithelium-mediated innate immune response.


Asunto(s)
Antibacterianos/farmacología , Glicoproteínas/fisiología , Fosfoproteínas/fisiología , Infecciones por Pseudomonas/prevención & control , Pseudomonas aeruginosa/inmunología , Animales , Antibacterianos/administración & dosificación , Femenino , Glicoproteínas/biosíntesis , Glicoproteínas/genética , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/fisiología , Ratones , Ratones Endogámicos , Ratones Transgénicos , Fosfoproteínas/biosíntesis , Fosfoproteínas/genética , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/patología , Neumonía Bacteriana/prevención & control , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/patología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/microbiología , Mucosa Respiratoria/patología , Uteroglobina/biosíntesis , Uteroglobina/genética , Uteroglobina/fisiología
20.
Elife ; 122023 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-37314162

RESUMEN

Aging is a critical risk factor in idiopathic pulmonary fibrosis (IPF). Dysfunction and loss of type 2 alveolar epithelial cells (AEC2s) with failed regeneration is a seminal causal event in the pathogenesis of IPF, although the precise mechanisms for their regenerative failure and demise remain unclear. To systematically examine the genomic program changes of AEC2s in aging and after lung injury, we performed unbiased single-cell RNA-seq analyses of lung epithelial cells from uninjured or bleomycin-injured young and old mice, as well as from lungs of IPF patients and healthy donors. We identified three AEC2 subsets based on their gene signatures. Subset AEC2-1 mainly exist in uninjured lungs, while subsets AEC2-2 and AEC2-3 emerged in injured lungs and increased with aging. Functionally, AEC2 subsets are correlated with progenitor cell renewal. Aging enhanced the expression of the genes related to inflammation, stress responses, senescence, and apoptosis. Interestingly, lung injury increased aging-related gene expression in AEC2s even in young mice. The synergistic effects of aging and injury contributed to impaired AEC2 recovery in aged mouse lungs after injury. In addition, we also identified three subsets of AEC2s from human lungs that formed three similar subsets to mouse AEC2s. IPF AEC2s showed a similar genomic signature to AEC2 subsets from bleomycin-injured old mouse lungs. Taken together, we identified synergistic effects of aging and AEC2 injury in transcriptomic and functional analyses that promoted fibrosis. This study provides new insights into the interactions between aging and lung injury with interesting overlap with diseased IPF AEC2 cells.


Asunto(s)
Lesión Pulmonar , Fibrosis Pulmonar , Humanos , Ratones , Animales , Fibrosis Pulmonar/patología , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/metabolismo , Pulmón/patología , Envejecimiento , Bleomicina/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA