Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
1.
Nature ; 604(7904): 120-126, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35355013

RESUMEN

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


Asunto(s)
Bronquiolos , Hurones , Células Madre Multipotentes , Alveolos Pulmonares , Animales , Bronquiolos/citología , Linaje de la Célula , Humanos , Pulmón/patología , Ratones , Células Madre Multipotentes/citología , Alveolos Pulmonares/citología , Enfermedad Pulmonar Obstructiva Crónica
2.
Am J Respir Cell Mol Biol ; 70(4): 247-258, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38117250

RESUMEN

BCL-2 family members are known to be implicated in survival in numerous biological settings. Here, we provide evidence that in injury and repair processes in lungs, BCL-2 mainly acts to attenuate endoplasmic reticulum (ER) stress and limit extracellular matrix accumulation. Days after an intratracheal bleomycin challenge, mice lose a fraction of their alveolar type II epithelium from terminal ER stress driven by activation of the critical ER sensor and stress effector IRE1α. This fraction is dramatically increased by BCL-2 inhibition, because IRE1α activation is dependent on its physical association with the BCL-2-proapoptotic family member BAX, and we found BCL-2 to disrupt this association in vitro. In vivo, navitoclax (a BCL-2/BCL-xL inhibitor) given 15-21 days after bleomycin challenge evoked strong activation of IRE-1α in mesenchymal cells and markers of ER stress, but not apoptosis. Remarkably, after BCL-2 inhibition, bleomycin-exposed mice demonstrated persistent collagen accumulation at Day 42, compared with resolution in controls. Enhanced fibrosis proved to be due to the RNAase activity of IRE1α downregulating MRC2 mRNA and protein, a mediator of collagen turnover. The critical role of MRC2 was confirmed in precision-cut lung slice cultures of Day-42 lungs from bleomycin-exposed wild-type and MRC2 null mice. Soluble and tissue collagen accumulated in precision-cut lung slice cultures from navitoclax-treated, bleomycin-challenged mice compared with controls, in a manner nearly identical to that of challenged but untreated MRC2 null mice. Thus, apart from mitochondrial-based antiapoptosis, BCL-2 functions to attenuate ER stress responses, fostering tissue homeostasis and injury repair.


Asunto(s)
Compuestos de Anilina , Fibrosis Pulmonar , Sulfonamidas , Ratones , Animales , Fibrosis Pulmonar/metabolismo , Endorribonucleasas , Proteínas Serina-Treonina Quinasas , Estrés del Retículo Endoplásmico , Ratones Noqueados , Colágeno/metabolismo , Bleomicina/farmacología
3.
Mol Cell ; 56(6): 723-37, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25526531

RESUMEN

TGF-ß signaling is essential in many processes, including immune surveillance, and its dysregulation controls various diseases, including cancer, fibrosis, and inflammation. Studying the innate host defense, which functions in most cell types, we found that RLR signaling represses TGF-ß responses. This regulation is mediated by activated IRF3, using a dual mechanism of IRF3-directed suppression. Activated IRF3 interacts with Smad3, thus inhibiting TGF-ß-induced Smad3 activation and, in the nucleus, disrupts functional Smad3 transcription complexes by competing with coregulators. Consequently, IRF3 activation by innate antiviral signaling represses TGF-ß-induced growth inhibition, gene regulation and epithelial-mesenchymal transition, and the generation of Treg effector lymphocytes from naive CD4(+) lymphocytes. Conversely, silencing IRF3 expression enhances epithelial-mesenchymal transition, TGF-ß-induced Treg cell differentiation upon virus infection, and Treg cell generation in vivo. We present a mechanism of regulation of TGF-ß signaling by the antiviral defense, with evidence for its role in immune tolerance and cancer cell behavior.


Asunto(s)
Factor 3 Regulador del Interferón/fisiología , Virus Sendai/inmunología , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/fisiología , Animales , Diferenciación Celular , Transición Epitelial-Mesenquimal , Células HEK293 , Células Hep G2 , Humanos , Inmunidad Innata , Ratones Endogámicos C57BL , Transducción de Señal , Linfocitos T Reguladores/inmunología , Transcripción Genética , Activación Transcripcional/inmunología
4.
Thorax ; 76(7): 729-732, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33472968

RESUMEN

We recently identified epigallocatechin gallate (EGCG), a trihydroxyphenolic compound, as a dual inhibitor of lysyl oxidase-like2 and transforming growth factor-ß1 (TGFß1) receptor kinase that when given orally to patients with idiopathic pulmonary fibrosis (IPF) reversed profibrotic biomarkers in their diagnostic biopsies. Here, we extend these findings to advanced pulmonary fibrosis using cultured precision-cut lung slices from explants of patients with IPF undergoing transplantation. During these experiments, we were surprised to discover that not only did EGCG attenuate TGFß1 signalling and new collagen accumulation but also activated matrix metalloproteinase-dependent collagen I turnover, raising the possibility of slow fibrosis resolution with continued treatment.


Asunto(s)
Aminoácido Oxidorreductasas/metabolismo , Colágeno Tipo I/metabolismo , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Células Cultivadas , Humanos , Fibrosis Pulmonar Idiopática/patología , Immunoblotting , Pulmón/patología , Transducción de Señal
5.
Nature ; 517(7536): 621-5, 2015 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-25533958

RESUMEN

Broadly, tissue regeneration is achieved in two ways: by proliferation of common differentiated cells and/or by deployment of specialized stem/progenitor cells. Which of these pathways applies is both organ- and injury-specific. Current models in the lung posit that epithelial repair can be attributed to cells expressing mature lineage markers. By contrast, here we define the regenerative role of previously uncharacterized, rare lineage-negative epithelial stem/progenitor (LNEP) cells present within normal distal lung. Quiescent LNEPs activate a ΔNp63 (a p63 splice variant) and cytokeratin 5 remodelling program after influenza or bleomycin injury in mice. Activated cells proliferate and migrate widely to occupy heavily injured areas depleted of mature lineages, at which point they differentiate towards mature epithelium. Lineage tracing revealed scant contribution of pre-existing mature epithelial cells in such repair, whereas orthotopic transplantation of LNEPs, isolated by a definitive surface profile identified through single-cell sequencing, directly demonstrated the proliferative capacity and multipotency of this population. LNEPs require Notch signalling to activate the ΔNp63 and cytokeratin 5 program, and subsequent Notch blockade promotes an alveolar cell fate. Persistent Notch signalling after injury led to parenchymal 'micro-honeycombing' (alveolar cysts), indicative of failed regeneration. Lungs from patients with fibrosis show analogous honeycomb cysts with evidence of hyperactive Notch signalling. Our findings indicate that distinct stem/progenitor cell pools repopulate injured tissue depending on the extent of the injury, and the outcomes of regeneration or fibrosis may depend in part on the dynamics of LNEP Notch signalling.


Asunto(s)
Células Epiteliales/citología , Células Epiteliales/patología , Lesión Pulmonar/patología , Pulmón/citología , Pulmón/patología , Repitelización , Células Madre/citología , Animales , Bleomicina , Linaje de la Célula , Proliferación Celular , Separación Celular , Quistes/metabolismo , Quistes/patología , Células Epiteliales/metabolismo , Femenino , Humanos , Queratina-5/metabolismo , Pulmón/fisiología , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/virología , Masculino , Ratones , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Trasplante de Células Madre , Células Madre/metabolismo , Transactivadores/genética , Transactivadores/metabolismo
7.
Am J Pathol ; 187(8): 1772-1786, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28618253

RESUMEN

During the acute respiratory distress syndrome, epithelial cells, primarily alveolar type (AT) I cells, die and slough off, resulting in enhanced permeability. ATII cells proliferate and spread onto the denuded basement membrane to reseal the barrier. Repair of the alveolar epithelium is critical for clinical recovery; however, mechanisms underlying ATII cell proliferation and spreading are not well understood. We hypothesized that hypoxia-inducible factor (HIF)1α promotes proliferation and spreading of ATII cells during repair after lung injury. Mice were treated with lipopolysaccharide or hydrochloric acid. HIF activation in ATII cells after injury was demonstrated by increased luciferase activity in oxygen degradation domain-Luc (HIF reporter) mice and expression of the HIF1α target gene GLUT1. ATII cell proliferation during repair was attenuated in ATII cell-specific HIF1α knockout (SftpcCreERT2+/-;HIF1αf/f) mice. The HIF target vascular endothelial growth factor promoted ATII cell proliferation in vitro and after lung injury in vivo. In the scratch wound assay of cell spreading, HIF stabilization accelerated, whereas HIF1α shRNA delayed wound closure. SDF1 and its receptor, CXCR4, were found to be HIF1α-regulated genes in ATII cells and were up-regulated during lung injury. Stromal cell-derived factor 1/CXCR4 inhibition impaired cell spreading and delayed the resolution of permeability after lung injury. We conclude that HIF1α is activated in ATII cells after lung injury and promotes proliferation and spreading during repair.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Células Epiteliales Alveolares/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Alveolos Pulmonares/metabolismo , Transducción de Señal/fisiología , Animales , Línea Celular , Proliferación Celular/fisiología , Quimiocina CXCL12/metabolismo , Modelos Animales de Enfermedad , Ratones , Permeabilidad , Ratas , Receptores CXCR4/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Cicatrización de Heridas/fisiología
8.
J Biol Chem ; 289(18): 12791-804, 2014 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-24644284

RESUMEN

The urokinase-type plasminogen activator receptor (uPAR) is a glycosylphosphatidylinositol-linked membrane protein with no cytosolic domain that localizes to lipid raft microdomains. Our laboratory and others have documented that lung fibroblasts from patients with idiopathic pulmonary fibrosis (IPF) exhibit a hypermotile phenotype. This study was undertaken to elucidate the molecular mechanism whereby uPAR ligation with its cognate ligand, urokinase, induces a motile phenotype in human lung fibroblasts. We found that uPAR ligation with the urokinase receptor binding domain (amino-terminal fragment) leads to enhanced migration of fibroblasts on fibronectin in a protease-independent, lipid raft-dependent manner. Ligation of uPAR with the amino-terminal fragment recruited α5ß1 integrin and the acylated form of the Src family kinase, Fyn, to lipid rafts. The biological consequences of this translocation were an increase in fibroblast motility and a switch of the integrin-initiated signal pathway for migration away from the lipid raft-independent focal adhesion kinase pathway and toward a lipid raft-dependent caveolin-Fyn-Shc pathway. Furthermore, an integrin homologous peptide as well as an antibody that competes with ß1 for uPAR binding have the ability to block this effect. In addition, its relative insensitivity to cholesterol depletion suggests that the interactions of α5ß1 integrin and uPAR drive the translocation of α5ß1 integrin-acylated Fyn signaling complexes into lipid rafts upon uPAR ligation through protein-protein interactions. This signal switch is a novel pathway leading to the hypermotile phenotype of IPF patient-derived fibroblasts, seen with uPAR ligation. This uPAR dependent, fibrotic matrix-selective, and profibrotic fibroblast phenotype may be amenable to targeted therapeutics designed to ameliorate IPF.


Asunto(s)
Movimiento Celular , Fibroblastos/metabolismo , Integrina alfa5beta1/metabolismo , Microdominios de Membrana/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Animales , Western Blotting , Caveolinas/genética , Caveolinas/metabolismo , Células Cultivadas , Fibroblastos/citología , Fibronectinas/metabolismo , Humanos , Fibrosis Pulmonar Idiopática/sangre , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Integrina alfa5beta1/genética , Ratones , Microscopía Fluorescente , Unión Proteica , Proteínas Proto-Oncogénicas c-fyn/genética , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Interferencia de ARN , Receptores del Activador de Plasminógeno Tipo Uroquinasa/sangre , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Índice de Severidad de la Enfermedad , Proteínas Adaptadoras de la Señalización Shc/genética , Proteínas Adaptadoras de la Señalización Shc/metabolismo , Transducción de Señal , Activador de Plasminógeno de Tipo Uroquinasa/genética , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
9.
Hum Mol Genet ; 22(7): 1417-23, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23297359

RESUMEN

Kufs disease, an adult-onset neuronal ceroid lipofuscinosis, is challenging to diagnose and genetically heterogeneous. Mutations in CLN6 were recently identified in recessive Kufs disease presenting as progressive myoclonus epilepsy (Type A), whereas the molecular basis of cases presenting with dementia and motor features (Type B) is unknown. We performed genome-wide linkage mapping of two families with recessive Type B Kufs disease and identified a single region on chromosome 11 to which both families showed linkage. Exome sequencing of five samples from the two families identified homozygous and compound heterozygous missense mutations in CTSF within this linkage region. We subsequently sequenced CTSF in 22 unrelated individuals with suspected recessive Kufs disease, and identified an additional patient with compound heterozygous mutations. CTSF encodes cathepsin F, a lysosomal cysteine protease, dysfunction of which is a highly plausible candidate mechanism for a storage disorder like ceroid lipofuscinosis. In silico modeling suggested the missense mutations would alter protein structure and function. Moreover, re-examination of a previously published mouse knockout of Ctsf shows that it recapitulates the light and electron-microscopic pathological features of Kufs disease. Although CTSF mutations account for a minority of cases of type B Kufs, CTSF screening should be considered in cases with early-onset dementia and may avoid the need for invasive biopsies.


Asunto(s)
Catepsina F/genética , Mutación Missense , Lipofuscinosis Ceroideas Neuronales/genética , Adulto , Animales , Células del Asta Anterior/patología , Estudios de Casos y Controles , Catepsina F/metabolismo , Mapeo Cromosómico , Consanguinidad , Análisis Mutacional de ADN , Exoma , Femenino , Estudios de Asociación Genética , Humanos , Escala de Lod , Ratones , Ratones Noqueados , Persona de Mediana Edad , Modelos Moleculares , Lipofuscinosis Ceroideas Neuronales/enzimología , Lipofuscinosis Ceroideas Neuronales/patología , Linaje , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Análisis de Secuencia de ARN
10.
Am J Respir Crit Care Med ; 189(2): 214-22, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24160862

RESUMEN

The median survival of patients with idiopathic pulmonary fibrosis (IPF) continues to be approximately 3 years from the time of diagnosis, underscoring the lack of effective medical therapies for this disease. In the United States alone, approximately 40,000 patients die of this disease annually. In November 2012, the NHLBI held a workshop aimed at coordinating research efforts and accelerating the development of IPF therapies. Basic, translational, and clinical researchers gathered with representatives from the NHLBI, patient advocacy groups, pharmaceutical companies, and the U.S. Food and Drug Administration to review the current state of IPF research and identify priority areas, opportunities for collaborations, and directions for future research. The workshop was organized into groups that were tasked with assessing and making recommendations to promote progress in one of the following six critical areas of research: (1) biology of alveolar epithelial injury and aberrant repair; (2) role of extracellular matrix; (3) preclinical modeling; (4) role of inflammation and immunity; (5) genetic, epigenetic, and environmental determinants; (6) translation of discoveries into diagnostics and therapeutics. The workshop recommendations provide a basis for directing future research and strategic planning by scientific, professional, and patient communities and the NHLBI.


Asunto(s)
Fibrosis Pulmonar Idiopática , Animales , Investigación Biomédica/tendencias , Modelos Animales de Enfermedad , Matriz Extracelular/patología , Predisposición Genética a la Enfermedad , Humanos , Fibrosis Pulmonar Idiopática/diagnóstico , Fibrosis Pulmonar Idiopática/fisiopatología , Fibrosis Pulmonar Idiopática/terapia , Inflamación/inmunología , Ratones , Alveolos Pulmonares/patología , Mucosa Respiratoria/patología
11.
Annu Rev Physiol ; 73: 413-35, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21054168

RESUMEN

Lung epithelial cells have emerged as a frequent target of injury, a driver of normal repair, and a key element in the pathobiology of fibrotic lung diseases. An important aspect of epithelial cells is their capacity to respond to microenvironmental cues by undergoing epithelial-mesenchymal transition (EMT). EMT is not simply widespread conversion of epithelial cells to fibroblasts but a graded response whereby epithelial cells reversibly acquire mesenchymal features and enhanced capacity for mesenchymal cross-talk. Recent studies elucidate distinct integrin-sensing systems that coordinate activity of TGFß1, a critical signaling element regulating EMT, with the presence of proinflammatory signals and cell injury. Repeated injury superimposes persistent inflammation and hypoxia onto these highly regulated repair pathways, potentially overwhelming orderly repair and creating sustained fibrogenesis. Understanding specific signaling mechanisms driving the mesenchymal response to TGFß1 may reveal therapeutics to attenuate fibrogenesis yet preserve the important homeostatic functions of TGFß1.


Asunto(s)
Transición Epitelial-Mesenquimal , Fibrosis Pulmonar/patología , Animales , Carcinoma/patología , Femenino , Homeostasis/fisiología , Humanos , Integrinas/fisiología , Neoplasias Pulmonares/patología , Masculino , Ratones , Transducción de Señal/fisiología , Células Madre/fisiología , Factor de Crecimiento Transformador beta1/fisiología
12.
Am J Respir Cell Mol Biol ; 50(1): 51-60, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23944988

RESUMEN

A high-throughput small-molecule screen was conducted to identify inhibitors of epithelial-mesenchymal transition (EMT) that could be used as tool compounds to test the importance of EMT signaling in vivo during fibrogenesis. Transforming growth factor (TGF)-ß1-induced fibronectin expression and E-cadherin repression in A549 cells were used as 48-hour endpoints in a cell-based imaging screen. Compounds that directly blocked Smad2/3 phosphorylation were excluded. From 2,100 bioactive compounds, methacycline was identified as an inhibitor of A549 EMT with the half maximal inhibitory concentration (IC50) of roughly 5 µM. In vitro, methacycline inhibited TGF-ß1-induced α-smooth muscle actin, Snail1, and collagen I of primary alveolar epithelial cells . Methacycline inhibited TGF-ß1-induced non-Smad pathways, including c-Jun N-terminal kinase, p38, and Akt activation, but not Smad or ß-catenin transcriptional activity. Methacycline had no effect on baseline c-Jun N-terminal kinase, p38, or Akt activities or lung fibroblast responses to TGF-ß1. In vivo, 100 mg/kg intraperitoneal methacycline delivered daily beginning 10 days after intratracheal bleomycin improved survival at Day 17 (P < 0.01). Bleomycin-induced canonical EMT markers, Snail1, Twist1, collagen I, as well as fibronectin protein and mRNA, were attenuated by methacycline (Day 17). Methacycline did not attenuate inflammatory cell accumulation or alter TGF-ß1-responsive genes in alveolar macrophages. These studies identify a novel inhibitor of EMT as a potent suppressor of fibrogenesis, further supporting the concept that EMT signaling is important to lung fibrosis. The findings also provide support for testing the impact of methacycline or doxycycline, an active analog, on progression of human pulmonary fibrosis.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Metaciclina/farmacología , Alveolos Pulmonares/efectos de los fármacos , Fibrosis Pulmonar/tratamiento farmacológico , Actinas/metabolismo , Animales , Cadherinas/metabolismo , Línea Celular , Colágeno Tipo I/metabolismo , Células Epiteliales/metabolismo , Femenino , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-akt/metabolismo , Alveolos Pulmonares/metabolismo , Fibrosis Pulmonar/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Smad/metabolismo , Factores de Transcripción de la Familia Snail , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
Biochim Biophys Acta ; 1832(7): 922-30, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23219956

RESUMEN

Lung epithelial cells use remarkably adaptive sensing and signaling systems to maintain a physiological state supporting gas exchange and minimizing environmental insults. One facet of epithelial adaptability is the reversible acquisition of mesenchymal features, a process termed epithelial-mesenchymal transition (EMT). Although in the adult, permanent and complete EMT appears rare or non-existent, a growing body of evidence implicates a critical role for the activation of EMT signaling in tissue remodeling, including fibrotic lung disease. The specific phenotypes of cells undergoing EMT re-programming during epithelial responses to injury continue to be defined and are reviewed here. Several recent studies implicate epithelial expression of canonical EMT transcription factors, such as Snail and Twist1, with the acquisition of a less differentiated, more proliferative stem-like state, providing an additional link between activation of EMT signaling and tissue repair. In lung airways, proliferating variant clara cells rely upon Snail for effective epithelial repair, and in the breast, cells possessing the greatest regenerative capacity also express Snail2. The ongoing elucidation of signaling underlying epithelial stem/progenitor expansion coincides with recent discoveries implicating regenerative activity in the lung, possibly including de novo regeneration of airway and alveolar units. It remains largely unknown what signals drive organization of epithelial progenitor cells that expand after lung injury, to what degree such organization is ever functionally relevant, and whether the lung regenerative potential recently observed in mouse models extends to humans. Yet these unknowns with clinical potential bring future mechanistic studies of EMT and lung repair directly into the field of regenerative medicine. This article is part of a Special Issue entitled: Fibrosis: Translation of basic research to human disease.


Asunto(s)
Transición Epitelial-Mesenquimal , Pulmón , Animales , Diferenciación Celular , Células Epiteliales/metabolismo , Humanos , Células Madre/metabolismo , Factores de Transcripción/metabolismo
14.
Am J Pathol ; 183(5): 1559-1570, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24012677

RESUMEN

Fibrosis is characterized by accumulation of activated fibroblasts and pathological deposition of fibrillar collagens. Activated fibroblasts overexpress matrix proteins and release factors that promote further recruitment of activated fibroblasts, leading to progressive fibrosis. The contribution of epithelial cells to this process remains unknown. Epithelium-directed injury may lead to activation of epithelial cells with phenotypes and functions similar to activated fibroblasts. Prior reports that used a reporter gene fate-mapping strategy are limited in their ability to investigate the functional significance of epithelial cell-derived mesenchymal proteins during fibrogenesis. We found that lung epithelial cell-derived collagen I activates fibroblast collagen receptor discoidin domain receptor-2, contributes significantly to fibrogenesis, and promotes resolution of lung inflammation. Alveolar epithelial cells undergoing transforming growth factor-ß-mediated mesenchymal transition express several other secreted profibrotic factors and are capable of activating lung fibroblasts. These studies provide direct evidence that activated epithelial cells produce mesenchymal proteins that initiate a cycle of fibrogenic effector cell activation, leading to progressive fibrosis. Therapy targeted at epithelial cell production of type I collagen offers a novel pathway for abrogating this progressive cycle and for limiting tissue fibrosis but may lead to sustained lung injury/inflammation.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/patología , Mesodermo/metabolismo , Proteínas/metabolismo , Animales , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis , Eliminación de Gen , Humanos , Ratones , Especificidad de Órganos , Neumonía/metabolismo , Neumonía/patología , Reproducibilidad de los Resultados
15.
Nature ; 456(7222): 658-62, 2008 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-18820679

RESUMEN

Mammalian Toll-like receptors (TLRs) 3, 7, 8 and 9 initiate immune responses to infection by recognizing microbial nucleic acids; however, these responses come at the cost of potential autoimmunity owing to inappropriate recognition of self nucleic acids. The localization of TLR9 and TLR7 to intracellular compartments seems to have a role in facilitating responses to viral nucleic acids while maintaining tolerance to self nucleic acids, yet the cell biology regulating the transport and localization of these receptors remains poorly understood. Here we define the route by which TLR9 and TLR7 exit the endoplasmic reticulum and travel to endolysosomes in mouse macrophages and dendritic cells. The ectodomains of TLR9 and TLR7 are cleaved in the endolysosome, such that no full-length protein is detectable in the compartment where ligand is recognized. Notably, although both the full-length and cleaved forms of TLR9 are capable of binding ligand, only the processed form recruits MyD88 on activation, indicating that this truncated receptor, rather than the full-length form, is functional. Furthermore, conditions that prevent receptor proteolysis, including forced TLR9 surface localization, render the receptor non-functional. We propose that ectodomain cleavage represents a strategy to restrict receptor activation to endolysosomal compartments and prevent TLRs from responding to self nucleic acids.


Asunto(s)
Procesamiento Proteico-Postraduccional , Receptor Toll-Like 9/química , Receptor Toll-Like 9/metabolismo , Animales , Línea Celular , Células Cultivadas , Células Dendríticas/citología , Células Dendríticas/metabolismo , Retículo Endoplásmico/metabolismo , Femenino , Aparato de Golgi/metabolismo , Ligandos , Lisosomas/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Masculino , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Ratones , Factor 88 de Diferenciación Mieloide/metabolismo , Fagosomas/metabolismo , Estructura Terciaria de Proteína , Transporte de Proteínas , Receptor Toll-Like 7/química , Receptor Toll-Like 7/metabolismo
16.
Am J Respir Crit Care Med ; 188(3): 370-5, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23713908

RESUMEN

The National Heart, Lung, and Blood Institute (NHLBI) of the National Institutes of Health convened the Cell Therapy for Lung Disease Working Group on November 13-14, 2012, to review and formulate recommendations for future research directions. The workshop brought together investigators studying basic mechanisms and the roles of cell therapy in preclinical models of lung injury and pulmonary vascular disease, with clinical trial experts in cell therapy for cardiovascular diseases and experts from the NHLBI Production Assistance for Cell Therapy program. The purpose of the workshop was to discuss the current status of basic investigations in lung cell therapy, to identify some of the scientific gaps in current knowledge regarding the potential roles and mechanisms of cell therapy in the treatment of lung diseases, and to develop recommendations to the NHLBI and the research community on scientific priorities and practical steps that would lead to first-in-human trials of lung cell therapy.


Asunto(s)
Investigación Biomédica/métodos , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Enfermedades Pulmonares/terapia , National Heart, Lung, and Blood Institute (U.S.) , Humanos , Estados Unidos
17.
J Clin Invest ; 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38980870

RESUMEN

Reciprocal interactions between alveolar fibroblasts and epithelial cells are crucial for lung homeostasis, injury repair, and fibrogenesis, but underlying mechanisms remain unclear. To investigate, we administered the fibroblast-selective TGFß1 signaling inhibitor, epigallocatechin gallate (EGCG), to Interstitial Lung Disease (ILD) patients undergoing diagnostic lung biopsy and conducted single-cell RNA sequencing on spare tissue. Biopsies from untreated patients showed higher fibroblast TGFß1 signaling compared to non-disease donor or end-stage ILD tissues. In vivo, EGCG downregulated TGFß1 signaling and several pro-inflammatory and stress pathways in biopsy samples. Notably, EGCG reduced fibroblast secreted frizzle-like receptor protein 2 (sFRP2), an unrecognized TGFß1 fibroblast target gene induced near type II alveolar epithelial cells (AEC2s) in situ. Using AEC2-fibroblast coculture organoids and precision cut lung slices (PCLS) from non-diseased donors, we found TGFß1 signaling promotes a spread AEC2 KRT17+ basaloid state, whereupon sFRP2 then activates a mature Krt5+ basal cell program. Wnt-receptor Frizzled 5 (Fzd5) expression and downstream calcineurin signaling were required for sFRP2-induced nuclear NFATc3 accumulation and KRT5 expression. These findings highlight stage-specific TGFß1 signaling in ILD, the therapeutic potential of EGCG in reducing IPF-related transcriptional changes, and identify TGFß1-non-canonical Wnt pathway crosstalk via sFRP2 as a novel mechanism for dysfunctional epithelial signaling in Idiopathic Pulmonary Fibrosis/ILD.

19.
J Biol Chem ; 287(7): 5164-72, 2012 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-22203675

RESUMEN

Epithelial to mesenchymal transition (EMT) and pulmonary fibrogenesis require epithelial integrin α3ß1-mediated cross-talk between TGFß1 and Wnt signaling pathways. One hallmark of this cross-talk is pY654-ß-catenin accumulation, but whether pY654-ß-catenin is a biomarker of fibrogenesis or functionally important is unknown. To clarify further the role of ß-catenin in fibrosis, we explored pY654-ß-catenin generation and function. α3ß1 was required for TGFß1-mediated activation of Src family kinases, and Src inhibition blocked both pY654 and EMT in primary alveolar epithelial cells (AECs). TGFß1 stimulated ß-catenin/Lef1-dependent promoter activity comparably in immortalized AECs stably expressing WT ß-catenin as well as Y654E or Y654F ß-catenin point mutants. But EMT was abrogated in the Tyr to Phe mutant. pY654-ß-catenin was sensitive to the axin ß-catenin turnover pathway as inhibition of tankyrase 1 led to high AEC axin levels, loss of pY654-ß-catenin, and inhibition of EMT ex vivo. Mice given a tankyrase inhibitor (50 mg/kg orally) daily for 7 days beginning 10 days after intratracheal bleomycin had improved survival over controls. Treated mice developed raised axin levels in the lung that abrogated pY654-ß-catenin and attenuated lung Snail1, Twist1, α-smooth muscle actin, and type I collagen accumulation. Total ß-catenin levels were unaltered. These findings identify Src kinase(s) as a mediator of TGFß1-induced pY654-ß-catenin, provide evidence that pY654-ß-catenin levels are a critical determinant of EMT and fibrogenesis, and suggest regulation of axin levels as a novel therapeutic approach to fibrotic disorders.


Asunto(s)
Sustitución de Aminoácidos , Colágeno Tipo I/biosíntesis , Células Epiteliales/metabolismo , Mutación Missense , Alveolos Pulmonares/metabolismo , Fibrosis Pulmonar/metabolismo , Animales , Antibióticos Antineoplásicos/efectos adversos , Antibióticos Antineoplásicos/farmacología , Proteína Axina , Bleomicina/efectos adversos , Bleomicina/farmacología , Línea Celular Transformada , Colágeno Tipo I/genética , Inhibidores Enzimáticos/farmacología , Células Epiteliales/patología , Transición Epitelial-Mesenquimal/genética , Integrina alfa3beta1/genética , Integrina alfa3beta1/metabolismo , Ratones , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Alveolos Pulmonares/patología , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología , Factores de Transcripción de la Familia Snail , Tanquirasas/antagonistas & inhibidores , Tanquirasas/genética , Tanquirasas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Proteína 1 Relacionada con Twist/genética , Proteína 1 Relacionada con Twist/metabolismo , beta Catenina
20.
bioRxiv ; 2023 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-37577522

RESUMEN

Reciprocal interactions between alveolar fibroblasts and epithelial cells are crucial for lung homeostasis, injury repair, and fibrogenesis, but underlying mechanisms remain unclear. To investigate this, we administered the fibroblast-selective TGFß1 signaling inhibitor, epigallocatechin gallate (EGCG), to Interstitial Lung Disease (ILD) patients undergoing diagnostic lung biopsy and conducted single-cell RNA sequencing on spare tissue. Unexposed biopsy samples showed higher fibroblast TGFß1 signaling compared to non-disease donor or end-stage ILD tissues. In vivo, EGCG significantly downregulated TGFß1 signaling and several pro-inflammatory and stress pathways in biopsy samples. Notably, EGCG reduced fibroblast secreted Frizzle-like Receptor Protein 2 (sFRP2), an unrecognized TGFß1 fibroblast target gene induced near type II alveolar epithelial cells (AEC2s). In human AEC2-fibroblast coculture organoids, sFRP2 was essential for AEC2 trans-differentiation to basal cells. Precision cut lung slices (PCLS) from normal donors demonstrated that TGFß1 promoted KRT17 expression and AEC2 morphological change, while sFRP2 was necessary for KRT5 expression in AEC2-derived basaloid cells. Wnt-receptor Frizzled 5 (Fzd5) expression and downstream calcineurin-related signaling in AEC2s were required for sFRP2-induced KRT5 expression. These findings highlight stage-specific TGFß1 signaling in ILD, the therapeutic potential of EGCG in reducing IPF-related transcriptional changes, and identify the TGFß1-non-canonical Wnt pathway crosstalk via sFRP2 as a novel mechanism for dysfunctional epithelial signaling in Idiopathic Pulmonary Fibrosis/ILD.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda