Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Respir Cell Mol Biol ; 71(2): 229-241, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38635761

RESUMEN

Aging poses a global public health challenge, which is linked to the rise of age-related lung diseases. The precise understanding of the molecular and genetic changes in the aging lung that elevate the risk of acute and chronic lung diseases remains incomplete. Alveolar type II (AT2) cells are stem cells that maintain epithelial homeostasis and repair the lung after injury. AT2 progenitor function decreases with aging. The maintenance of AT2 function requires niche support from other cell types, but little has been done to characterize alveolar alterations with aging in the AT2 niche. To systematically profile the genetic changes associated with age, we present a single-cell transcriptional atlas comprising nearly half a million cells from the healthy lungs of human subjects spanning various ages, sexes, and smoking statuses. Most annotated cell lineages in aged lungs exhibit dysregulated genetic programs. Specifically, the aged AT2 cells demonstrate loss of epithelial identities, heightened inflammaging characterized by increased expression of AP-1 (Activator Protein-1) transcription factor and chemokine genes, and significantly increased cellular senescence. Furthermore, the aged mesenchymal cells display a remarkable decrease in collagen and elastin transcription and a loss of support to epithelial cell stemness. The decline of the AT2 niche is further exacerbated by a dysregulated genetic program in macrophages and dysregulated communications between AT2 and macrophages in aged human lungs. These findings highlight the dysregulations observed in both AT2 stem cells and their supportive niche cells, potentially contributing to the increased susceptibility of aged populations to lung diseases.


Asunto(s)
Envejecimiento , Células Epiteliales Alveolares , Pulmón , Nicho de Células Madre , Transcriptoma , Humanos , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Envejecimiento/genética , Pulmón/metabolismo , Pulmón/patología , Transcriptoma/genética , Anciano , Persona de Mediana Edad , Masculino , Senescencia Celular/genética , Perfilación de la Expresión Génica , Femenino , Adulto , Células Madre/metabolismo
2.
Am J Respir Cell Mol Biol ; 71(2): 242-253, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38657143

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is an aging-associated interstitial lung disease resulting from repeated epithelial injury and inadequate epithelial repair. Alveolar type II cells (AEC2s) are progenitor cells that maintain epithelial homeostasis and repair the lung after injury. In the current study, we assessed lipid metabolism in AEC2s from human lungs of patients with IPF and healthy donors, as well as AEC2s from bleomycin-injured young and old mice. Through single-cell RNA sequencing, we observed that lipid metabolism-related genes were downregulated in IPF AEC2s and bleomycin-injured mouse AEC2s. Aging aggravated this decrease and hindered recovery of lipid metabolism gene expression in AEC2s after bleomycin injury. Pathway analyses revealed downregulation of genes related to lipid biosynthesis and fatty acid ß-oxidation in AEC2s from IPF lungs and bleomycin-injured, old mouse lungs compared with the respective controls. We confirmed decreased cellular lipid content in AEC2s from IPF lungs and bleomycin-injured, old mouse lungs using immunofluorescence staining and flow cytometry. Futhermore, we show that lipid metabolism was associated with AEC2 progenitor function. Lipid supplementation and PPARγ (peroxisome proliferator activated receptor γ) activation promoted progenitor renewal capacity of both human and mouse AEC2s in three-dimensional organoid cultures. Lipid supplementation also increased AEC2 proliferation and expression of SFTPC in AEC2s. In summary, we identified a lipid metabolism deficiency in AEC2s from lungs of patients with IPF and bleomycin-injured old mice. Restoration of lipid metabolism homeostasis in AEC2s might promote AEC2 progenitor function and offer new opportunities for therapeutic approaches to IPF.


Asunto(s)
Envejecimiento , Células Epiteliales Alveolares , Bleomicina , Fibrosis Pulmonar Idiopática , Metabolismo de los Lípidos , Células Madre , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Animales , Humanos , Ratones , Células Madre/metabolismo , Células Madre/patología , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Envejecimiento/metabolismo , Envejecimiento/patología , PPAR gamma/metabolismo , Masculino , Ratones Endogámicos C57BL , Femenino
3.
Am J Physiol Lung Cell Mol Physiol ; 327(2): L160-L172, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38771132

RESUMEN

The alveolar type II epithelial cells (AEC2s) act as stem cells in the lung for alveolar epithelial maintenance and repair. Chemokine C-X-C motif chemokine 10 (CXCL10) is expressed in injured tissues, modulating multiple cellular functions. AEC2s, previously reported to release chemokines to recruit leukocytes, were found in our study to secrete CXCL10 after bleomycin injury. We found that Sftpc-Cxcl10 transgenic mice were protected from bleomycin injury. The transgenic mice showed an increase in the AEC2 population in the lung by flow cytometry analysis. Both endogenous and exogenous CXCL10 promoted the colony formation efficiency of AEC2s in a three-dimensional (3-D) organoid growth assay. We identified that the regenerative effect of CXCL10 was CXCR3 independent using Cxcr3-deficient mice, but it was related to the TrkA pathway. Binding experiments showed that CXCL10 interacted with TrkA directly and reversibly. This study demonstrates a previously unidentified AEC2 autocrine signaling of CXCL10 to promote their regeneration and proliferation, probably involving a CXCR3-independent TrkA pathway.NEW & NOTEWORTHY CXCL10 may aid in lung injury recovery by promoting the proliferation of alveolar stem cells and using a distinct regulatory pathway from the classical one.


Asunto(s)
Células Epiteliales Alveolares , Quimiocina CXCL10 , Receptores CXCR3 , Animales , Ratones , Células Epiteliales Alveolares/metabolismo , Proliferación Celular , Quimiocina CXCL10/metabolismo , Quimiocina CXCL10/genética , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Ratones Endogámicos C57BL , Ratones Transgénicos , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología , Receptores CXCR3/metabolismo , Receptores CXCR3/genética , Regeneración , Transducción de Señal
4.
Am J Respir Cell Mol Biol ; 69(1): 45-56, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36927333

RESUMEN

Progressive pulmonary fibrosis results from a dysfunctional tissue repair response and is characterized by fibroblast proliferation, activation, and invasion and extracellular matrix accumulation. Lung fibroblast heterogeneity is well recognized. With single-cell RNA sequencing, fibroblast subtypes have been reported by recent studies. However, the roles of fibroblast subtypes in effector functions in lung fibrosis are not well understood. In this study, we incorporated the recently published single-cell RNA-sequencing datasets on murine lung samples of fibrosis models and human lung samples of fibrotic diseases and analyzed fibroblast gene signatures. We identified and confirmed the novel fibroblast subtypes we reported recently across all samples of both mouse models and human lung fibrotic diseases, including idiopathic pulmonary fibrosis, systemic sclerosis-associated interstitial lung disease, and coronavirus disease (COVID-19). Furthermore, we identified specific cell surface proteins for each fibroblast subtype through differential gene expression analysis, which enabled us to isolate primary cells representing distinct fibroblast subtypes by flow cytometry sorting. We compared matrix production, including fibronectin, collagen, and hyaluronan, after profibrotic factor stimulation and assessed the invasive capacity of each fibroblast subtype. Our results suggest that in addition to myofibroblasts, lipofibroblasts and Ebf1+ (Ebf transcription factor 1+) fibroblasts are two important fibroblast subtypes that contribute to matrix deposition and also have enhanced invasive, proliferative, and contraction phenotypes. The histological locations of fibroblast subtypes are identified in healthy and fibrotic lungs by these cell surface proteins. This study provides new insights to inform approaches to targeting lung fibroblast subtypes to promote the development of therapeutics for lung fibrosis.


Asunto(s)
COVID-19 , Fibrosis Pulmonar Idiopática , Humanos , Ratones , Animales , COVID-19/metabolismo , Fibroblastos/metabolismo , Pulmón/patología , Fibrosis Pulmonar Idiopática/patología , Fibrosis , Proteínas de la Membrana/metabolismo
5.
Am J Respir Cell Mol Biol ; 68(3): 302-313, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36318668

RESUMEN

Loss of epithelial integrity, bronchiolarization, and fibroblast activation are key characteristics of idiopathic pulmonary fibrosis (IPF). Prolonged accumulation of basal-like cells in IPF may impact the fibrotic niche to promote fibrogenesis. To investigate their role in IPF, basal cells were isolated from IPF explant and healthy donor lung tissues. Single-cell RNA sequencing was used to assess differentially expressed genes in basal cells. Basal cell and niche interaction was demonstrated with the sLP-mCherry niche labeling system. Luminex assays were used to assess cytokines secreted by basal cells. The role of basal cells in fibroblast activation was studied. Three-dimensional organoid culture assays were used to interrogate basal cell effects on AEC2 (type 2 alveolar epithelial cell) renewal capacity. Perturbation was used to investigate WNT7A function in vitro and in a repetitive bleomycin model in vivo. We found that WNT7A is highly and specifically expressed in basal-like cells. Proteins secreted by basal cells can be captured by neighboring fibroblasts and AEC2s. Basal cells or basal cell-conditioned media activate fibroblasts through WNT7A. Basal cell-derived WNT7A inhibits AEC2 progenitor cell renewal in three-dimensional organoid cultures. Neutralizing antibodies against WNT7A or a small molecule inhibitor of Frizzled signaling abolished basal cell-induced fibroblast activation and attenuated lung fibrosis in mice. In summary, basal cells and basal cell-derived WNT7A are key components of the fibrotic niche, providing a unique non-stem cell function of basal cells in IPF progression and a novel targeting strategy for IPF.


Asunto(s)
Fibrosis Pulmonar Idiopática , Animales , Ratones , Bleomicina/farmacología , Fibroblastos/metabolismo , Fibrosis , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/patología , Transducción de Señal
6.
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
7.
Mol Ther ; 29(1): 347-364, 2021 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-33007201

RESUMEN

Follistatin-like 1 (FSTL1) is a matricellular protein that is upregulated during development and disease, including idiopathic pulmonary fibrosis (IPF), keloid, and arthritis. The profibrotic and pro-inflammatory roles of FSTL1 have been intensively studied during the last several years, as well as in this report. We screened and identified epitope-specific monoclonal neutralizing antibodies (nAbs) to functionally block FSTL1. FSTL1 nAbs attenuated bleomycin-induced pulmonary and dermal fibrosis in vivo and transforming growth factor (TGF)-ß1-induced dermal fibrosis ex vivo in human skin. In addition, FSTL1 nAbs significantly reduced existing lung fibrosis and skin fibrosis in experimental models. FSTL1 nAbs exerted their potent antifibrotic effects via reduced TGF-ß1 responsiveness and subsequent myofibroblast activation and extracellular matrix production. We also observed that FSTL1 nAbs attenuated the severity of collagen-induced arthritis in mice, which was accompanied by reduced inflammatory responses in vitro. Our findings suggest that FSTL1 nAbs are a promising new therapeutic strategy for the treatment of multiple organ fibrosis and systemic autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/metabolismo , Biomarcadores , Susceptibilidad a Enfermedades , Proteínas Relacionadas con la Folistatina/metabolismo , Fibrosis Pulmonar Idiopática/etiología , Fibrosis Pulmonar Idiopática/metabolismo , Animales , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/patología , Modelos Animales de Enfermedad , Descubrimiento de Drogas , Fibrosis , Proteínas Relacionadas con la Folistatina/antagonistas & inhibidores , Proteínas Relacionadas con la Folistatina/genética , Expresión Génica , Humanos , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Fibrosis Pulmonar Idiopática/patología , Ratones , Terapia Molecular Dirigida , Factor de Crecimiento Transformador beta1/metabolismo
8.
Am J Respir Cell Mol Biol ; 60(1): 41-48, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30130411

RESUMEN

Severe pulmonary fibrosis such as idiopathic pulmonary fibrosis (IPF) is characterized by the accumulation of extracellular matrix and fibroblast activation. Targeting fibroblast activation has contributed to the development of antifibrotic therapeutics for patients with IPF. Mitogen-activated protein kinase-activated protein kinase 2 (MK2), downstream in the transforming growth factor-ß/p38 mitogen-activated protein kinase pathway, has been implicated in inflammatory and fibrosing diseases. Increased concentrations of activated MK2 were expressed in IPF lung and in the mouse bleomycin model of lung fibrosis. The aim of the present study was to determine the role and the mechanisms of MK2 in fibroblast invasion and lung fibrosis. Our results showed that an MK2 inhibitor (MMI-0100) was able to inhibit the invasive capacity of lung fibroblasts isolated from patients with IPF, as well as fibroblasts isolated from both wild-type mice and mice with overexpressing hyaluronan synthase 2 (HAS2) in the myofibroblast compartment. We previously showed that hyaluronan and HAS2 regulate fibroblast invasion and lung fibrosis in vivo. The results of the present study showed that MMI-0100 reduced transforming growth factor-ß-induced hyaluronan production in human and mouse fibroblasts in vitro and that HAS2 mediated MK2 activation, suggesting a feed-forward loop in fibroblast activation. More importantly, MK2 inhibition attenuated hyaluronan accumulation and reduced collagen content in bleomycin-injured mouse lungs in vivo. Conditional deletion of MK2 in fibroblasts attenuated bleomycin-induced lung fibrosis. These data provide evidence that MK2 has a role in fibroblast invasion and fibrosis and may be a novel therapeutic target in pulmonary fibrosis.


Asunto(s)
Fibroblastos/patología , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Fibrosis Pulmonar/prevención & control , Índice de Severidad de la Enfermedad , Animales , Antibióticos Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Bleomicina/toxicidad , Células Cultivadas , Fibroblastos/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología
11.
Physiol Rev ; 91(1): 221-64, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21248167

RESUMEN

Accumulation and turnover of extracellular matrix components are the hallmarks of tissue injury. Fragmented hyaluronan stimulates the expression of inflammatory genes by a variety of immune cells at the injury site. Hyaluronan binds to a number of cell surface proteins on various cell types. Hyaluronan fragments signal through both Toll-like receptor (TLR) 4 and TLR2 as well as CD44 to stimulate inflammatory genes in inflammatory cells. Hyaluronan is also present on the cell surface of epithelial cells and provides protection against tissue damage from the environment by interacting with TLR2 and TLR4. Hyaluronan and hyaluronan-binding proteins regulate inflammation, tissue injury, and repair through regulating inflammatory cell recruitment, release of inflammatory cytokines, and cell migration. This review focuses on the role of hyaluronan as an immune regulator in human diseases.


Asunto(s)
Ácido Hialurónico/inmunología , Sistema Inmunológico/metabolismo , Animales , Artritis/metabolismo , Lesiones Encefálicas/metabolismo , Diabetes Mellitus/metabolismo , Cardiopatías/metabolismo , Humanos , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/biosíntesis , Hialuronoglucosaminidasa/metabolismo , Enfermedades Renales/metabolismo , Hepatopatías/metabolismo , Enfermedades Pulmonares/metabolismo , Transducción de Señal , Células Madre/metabolismo , Receptores Toll-Like/metabolismo , Heridas y Lesiones/metabolismo
12.
Am J Respir Cell Mol Biol ; 57(6): 721-732, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28799781

RESUMEN

Successful repair and renewal of alveolar epithelial cells (AECs) are critical in prohibiting the accumulation of myofibroblasts in pulmonary fibrogenesis. MicroRNAs (miRNAs) are multifocal regulators involved in lung injury and repair. However, the contribution of miRNAs to AEC2 renewal and apoptosis is incompletely understood. We report that miRNA-29c (miR-29c) expression is lower in AEC2s of individuals with idiopathic pulmonary fibrosis than in healthy lungs. Epithelial cells overexpressing miR-29c show higher proliferative rates and viability. miR-29c protects epithelial cells from apoptosis by targeting forkhead box O3a (Foxo3a). Both overexpression of miR-29c conventionally and AEC2s specifically lead to less fibrosis and better recovery in vivo. Furthermore, deficiency of miR-29c in AEC2s results in higher apoptosis and reduced epithelial renewal. Interestingly, a gene network including a subset of apoptotic genes was coregulated by both Toll-like receptor 4 and miR-29c. Taken together, miR-29c maintains epithelial integrity and promotes recovery from lung injury, thereby attenuating lung fibrosis in mice.


Asunto(s)
Apoptosis , Células Epiteliales/metabolismo , Fibrosis Pulmonar Idiopática/metabolismo , MicroARNs/metabolismo , Mucosa Respiratoria/metabolismo , Animales , Células Epiteliales/patología , Femenino , Humanos , Fibrosis Pulmonar Idiopática/inducido químicamente , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/patología , Masculino , Ratones , MicroARNs/genética , Mucosa Respiratoria/patología , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
13.
Am J Respir Cell Mol Biol ; 57(6): 702-710, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28787175

RESUMEN

Hyaluronan (HA), a major component of the extracellular matrix, is secreted by airway structural cells. Airway fibroblasts in allergic asthma secrete elevated levels of HA in association with increased HA synthase 2 (HAS2) expression. Thus, we hypothesized that HA accumulation in the airway wall may contribute to airway remodeling and hyperresponsiveness in allergic airways disease. To examine this hypothesis, transgenic mice in which the α-smooth muscle actin (α-SMA) promoter drives HAS2 expression were generated. Mixed male and female α-SMA-HAS2 mice (HAS2+ mice, n = 16; HAS2- mice, n = 13) were sensitized via intraperitoneal injection and then chronically challenged with aerosolized ovalbumin (OVA) for 6 weeks. To test airway responsiveness, increasing doses of methacholine were delivered intravenously and airway resistance was measured using the forced oscillation technique. HA, cytokines, and cell types were analyzed in bronchoalveolar lavage fluid, serum, and whole lung homogenates. Lung sections were stained using antibodies specific for HA-binding protein (HABP) and α-SMA, as well as Masson's trichrome stain. Staining of lung tissue demonstrated significantly increased peribronchial HA, α-SMA, and collagen deposition in OVA-challenged α-SMA-HAS2+ mice compared with α-SMA-HAS2- mice. Unexpectedly, OVA-challenged α-SMA-HAS2+ mice displayed significantly reduced airway responsiveness to methacholine compared with similarly treated α-SMA-HAS2- mice. The total numbers of inflammatory cell types in the bronchoalveolar lavage fluid did not differ significantly between OVA-challenged α-SMA-HAS2+ mice and α-SMA-HAS2- mice. We conclude that allergen-challenged mice that overexpress HAS2 in myofibroblasts and smooth muscle cells develop increased airway fibrosis, which lessens airway hyperresponsiveness to bronchoconstrictors.


Asunto(s)
Asma/enzimología , Regulación Enzimológica de la Expresión Génica , Hialuronano Sintasas/biosíntesis , Pulmón/enzimología , Miocitos del Músculo Liso/enzimología , Miofibroblastos/enzimología , Actinas/biosíntesis , Actinas/genética , Alérgenos/toxicidad , Animales , Asma/inducido químicamente , Asma/genética , Broncoconstricción/efectos de los fármacos , Broncoconstricción/genética , Enfermedad Crónica , Humanos , Hialuronano Sintasas/genética , Pulmón/patología , Ratones , Ratones Noqueados , Miocitos del Músculo Liso/patología , Miofibroblastos/patología
14.
Am J Physiol Lung Cell Mol Physiol ; 313(1): L41-L51, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28473330

RESUMEN

The alveolar epithelium is composed of type I cells covering most of the gas-blood exchange surface and type II cells secreting surfactant that lowers surface tension of alveoli to prevent alveolar collapse. Here, we have identified a subgroup of type II cells expressing a higher level of cell surface molecule CD44 (CD44high type II cells) that composed ~3% of total type II cells in 5-10-wk-old mice. These cells were preferentially apposed to lung capillaries. They displayed a higher proliferation rate and augmented differentiation capacity into type I cells and the ability to form alveolar organoids compared with CD44low type II cells. Moreover, in aged mice, 18-24 mo old, the percentage of CD44high type II cells among all type II cells was increased, but these cells showed decreased progenitor properties. Thus CD44high type II cells likely represent a type II cell subpopulation important for constitutive regulation of alveolar homeostasis.


Asunto(s)
Células Epiteliales Alveolares/citología , Células Epiteliales Alveolares/metabolismo , Homeostasis , Receptores de Hialuranos/metabolismo , Células Madre/citología , Envejecimiento/metabolismo , Animales , Capilares/metabolismo , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Células Cultivadas , Integrinas/metabolismo , Pulmón/citología , Ratones Endogámicos C57BL , Organoides/metabolismo , Proteína C Asociada a Surfactante Pulmonar/metabolismo , Células Madre/metabolismo
15.
Respir Res ; 16: 124, 2015 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-26453058

RESUMEN

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a fatal disease characterized by fibroblastic foci and progressive scarring of the pulmonary parenchyma. IPF fibroblasts display increased proliferation and enhanced migration and invasion, analogous to cancer cells. This transformation-like phenotype of fibroblasts plays an important role in the development of pulmonary fibrosis, but the mechanism for this is not well understood. METHODS: In this study, we compared gene expression profiles in fibrotic lung tissues from IPF patients and normal lung tissues from patients with primary spontaneous pneumothorax using a cDNA microarray to examine the mechanisms involved in the pathogenesis of IPF. In a cDNA microarray, we found that USP13 was decreased in lung tissues from patients with IPF, which was further confirmed by results from immunohistochemistry and western blot assays. Then, we used RNA interference in MRC-5 cells to inhibit USP13 and evaluated its effects by western blot, real-time RT-PCR, bromodeoxyuridine incorporation, and transwell assays. We also used co-immunoprecipitation and immunofluorescence staining to identify the correlation between USP13 and PTEN in IPF. RESULTS: USP13 expression levels were markedly reduced in fibroblastic foci and primary IPF fibroblast lines. The depletion of USP13 resulted in the transformation of fibroblasts into an aggressive phenotype with enhanced proliferative, migratory, and invasive capacities. Additionally, USP13 interacted with PTEN and mediated PTEN ubiquitination and degradation in lung fibroblasts. CONCLUSIONS: Down-regulation of USP13 mediates PTEN protein loss and fibroblast phenotypic change, and thereby plays a crucial role in IPF pathogenesis.


Asunto(s)
Diferenciación Celular , Endopeptidasas/metabolismo , Transición Epitelial-Mesenquimal , Fibroblastos/enzimología , Fibrosis Pulmonar Idiopática/enzimología , Pulmón/enzimología , Adulto , Western Blotting , Estudios de Casos y Controles , Línea Celular , Movimiento Celular , Proliferación Celular , Regulación hacia Abajo , Endopeptidasas/genética , Fibroblastos/patología , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica/métodos , Humanos , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/patología , Inmunoprecipitación , Pulmón/patología , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfohidrolasa PTEN/metabolismo , Fenotipo , Proteolisis , Interferencia de ARN , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Factores de Tiempo , Transfección , Proteasas Ubiquitina-Específicas , Ubiquitinación
17.
J Immunol ; 188(5): 2437-44, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22287715

RESUMEN

The molecular mechanisms of acute lung injury are incompletely understood. MicroRNAs (miRNAs) are crucial biological regulators that act by suppressing their target genes and are involved in a variety of pathophysiologic processes. miR-127 appears to be downregulated during lung injury. We set out to investigate the role of miR-127 in lung injury and inflammation. Expression of miR-127 significantly reduced cytokine release by macrophages. Looking into the mechanisms of regulation of inflammation by miR-127, we found that IgG FcγRI (CD64) was a target of miR-127, as evidenced by reduced CD64 protein expression in macrophages overexpressing miR-127. Furthermore, miR-127 significantly reduced the luciferase activity with a reporter construct containing the native 3' untranslated region of CD64. Importantly, we demonstrated that miR-127 attenuated lung inflammation in an IgG immune complex model in vivo. Collectively, these data show that miR-127 targets macrophage CD64 expression and promotes the reduction of lung inflammation. Understanding how miRNAs regulate lung inflammation may represent an attractive way to control inflammation induced by infectious or noninfectious lung injury.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/prevención & control , Marcación de Gen/métodos , Mediadores de Inflamación/fisiología , MicroARNs/fisiología , Receptores de IgG/metabolismo , Lesión Pulmonar Aguda/patología , Animales , Complejo Antígeno-Anticuerpo/toxicidad , Línea Celular , Citocinas/antagonistas & inhibidores , Citocinas/metabolismo , Femenino , Células HEK293 , Humanos , Mediadores de Inflamación/metabolismo , Infecciones por Lentivirus/inmunología , Infecciones por Lentivirus/patología , Infecciones por Lentivirus/prevención & control , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , MicroARNs/biosíntesis , MicroARNs/genética , Datos de Secuencia Molecular , Neumonía Viral/inmunología , Neumonía Viral/patología , Neumonía Viral/prevención & control , Receptores de IgG/antagonistas & inhibidores , Receptores de IgG/biosíntesis , Receptores de IgG/genética , Células U937
18.
Proc Natl Acad Sci U S A ; 108(52): E1475-83, 2011 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-22123957

RESUMEN

There are currently few treatment options for pulmonary fibrosis. Innovations may come from a better understanding of the cellular origin of the characteristic fibrotic lesions. We have analyzed normal and fibrotic mouse and human lungs by confocal microscopy to define stromal cell populations with respect to several commonly used markers. In both species, we observed unexpected heterogeneity of stromal cells. These include numerous cells with molecular and morphological characteristics of pericytes, implicated as a source of myofibroblasts in other fibrotic tissues. We used mouse genetic tools to follow the fates of specific cell types in the bleomcyin-induced model of pulmonary fibrosis. Using inducible transgenic alleles to lineage trace pericyte-like cells in the alveolar interstitium, we show that this population proliferates in fibrotic regions. However, neither these cells nor their descendants express high levels of the myofibroblast marker alpha smooth muscle actin (Acta2, aSMA). We then used a Surfactant protein C-CreER(T2) knock-in allele to follow the fate of Type II alveolar cells (AEC2) in vivo. We find no evidence at the cellular or molecular level for epithelial to mesenchymal transition of labeled cells into myofibroblasts. Rather, bleomycin accelerates the previously reported conversion of AEC2 into AEC1 cells. Similarly, epithelial cells labeled with our Scgb1a1-CreER allele do not give rise to fibroblasts but generate both AEC2 and AEC1 cells in response to bleomycin-induced lung injury. Taken together, our results show a previously unappreciated heterogeneity of cell types proliferating in fibrotic lesions and exclude pericytes and two epithelial cell populations as the origin of myofibroblasts.


Asunto(s)
Diferenciación Celular/fisiología , Alveolos Pulmonares/citología , Fibrosis Pulmonar/patología , Células del Estroma/citología , Actinas/metabolismo , Animales , Biomarcadores/metabolismo , Bleomicina/toxicidad , Bromodesoxiuridina , Proliferación Celular , Transición Epitelial-Mesenquimal/fisiología , Proteínas de la Matriz Extracelular/metabolismo , Citometría de Flujo , Humanos , Inmunohistoquímica , Ratones , Miofibroblastos/citología , Pericitos/metabolismo , Alveolos Pulmonares/patología , Fibrosis Pulmonar/inducido químicamente , Reacción en Cadena en Tiempo Real de la Polimerasa , Células del Estroma/metabolismo
19.
Chin Med J Pulm Crit Care Med ; 2(2): 63-71, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-39169931

RESUMEN

Cell-cell interactions are essential components of coordinated cell function in lung homeostasis. Lung diseases involve altered cell-cell interactions and communication between different cell types, as well as between subsets of cells of the same type. The identification and understanding of intercellular signaling in lung fibrosis offer insights into the molecular mechanisms underlying these interactions and their implications in the development and progression of lung fibrosis. A comprehensive cell atlas of the human lung, established with the facilitation of single-cell RNA transcriptomic analysis, has enabled the inference of intercellular communications using ligand-receptor databases. In this review, we provide a comprehensive overview of the modified cell-cell communications in lung fibrosis. We highlight the intricate interactions among the major cell types within the lung and their contributions to fibrogenesis. The insights presented in this review will contribute to a better understanding of the molecular mechanisms underlying lung fibrosis and may guide future research efforts in developing targeted therapies for this debilitating disease.

20.
Artículo en Inglés | MEDLINE | ID: mdl-39220636

RESUMEN

Human alveolar type I (AT1) cells are specialized epithelial cells that line the alveoli in the lungs where gas exchange occurs. The primary function of AT1 cells is not only to facilitate efficient gas exchange between the air and the blood in the lungs, but also to contribute to the structural integrity of the alveoli to maintain lung function and homeostasis. Aging has notable effects on the structure, function, and regenerative capacity of human AT1 cells. However, our understanding of the molecular mechanisms driving these age-related changes in AT1 cells remains limited. Leveraging a recent single-cell transcriptomics dataset we generated on healthy human lungs, we identified a series of significant molecular alterations in AT1 cells from aged lungs. Notably, the aged AT1 cells exhibited increased cellular senescence and chemokine gene expression, alongside diminished epithelial features such as decreases in cell junctions, endocytosis, and pulmonary matrisome gene expression. Gene set analyses also indicated that aged AT1 cells were resistant to apoptosis, a crucial mechanism for turnover and renewal of AT1 cells, thereby ensuring alveolar integrity and function. Further research on these alterations is imperative to fully elucidate the impact on AT1 cells and is indispensable for developing effective therapies to preserve lung function and promote healthy aging.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA