Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Circ Res ; 135(1): 60-75, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38770652

RESUMEN

BACKGROUND: Pathogenic concepts of right ventricular (RV) failure in pulmonary arterial hypertension focus on a critical loss of microvasculature. However, the methods underpinning prior studies did not take into account the 3-dimensional (3D) aspects of cardiac tissue, making accurate quantification difficult. We applied deep-tissue imaging to the pressure-overloaded RV to uncover the 3D properties of the microvascular network and determine whether deficient microvascular adaptation contributes to RV failure. METHODS: Heart sections measuring 250-µm-thick were obtained from mice after pulmonary artery banding (PAB) or debanding PAB surgery and properties of the RV microvascular network were assessed using 3D imaging and quantification. Human heart tissues harvested at the time of transplantation from pulmonary arterial hypertension cases were compared with tissues from control cases with normal RV function. RESULTS: Longitudinal 3D assessment of PAB mouse hearts uncovered complex microvascular remodeling characterized by tortuous, shorter, thicker, highly branched vessels, and overall preserved microvascular density. This remodeling process was reversible in debanding PAB mice in which the RV function recovers over time. The remodeled microvasculature tightly wrapped around the hypertrophied cardiomyocytes to maintain a stable contact surface to cardiomyocytes as an adaptation to RV pressure overload, even in end-stage RV failure. However, microvasculature-cardiomyocyte contact was impaired in areas with interstitial fibrosis where cardiomyocytes displayed signs of hypoxia. Similar to PAB animals, microvascular density in the RV was preserved in patients with end-stage pulmonary arterial hypertension, and microvascular architectural changes appeared to vary by etiology, with patients with pulmonary veno-occlusive disease displaying a lack of microvascular complexity with uniformly short segments. CONCLUSIONS: 3D deep tissue imaging of the failing RV in PAB mice, pulmonary hypertension rats, and patients with pulmonary arterial hypertension reveals complex microvascular changes to preserve the microvascular density and maintain a stable microvascular-cardiomyocyte contact. Our studies provide a novel framework to understand microvascular adaptation in the pressure-overloaded RV that focuses on cell-cell interaction and goes beyond the concept of capillary rarefaction.


Asunto(s)
Hipertensión Pulmonar , Imagenología Tridimensional , Ratones Endogámicos C57BL , Animales , Humanos , Ratones , Hipertensión Pulmonar/fisiopatología , Hipertensión Pulmonar/diagnóstico por imagen , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/patología , Masculino , Ventrículos Cardíacos/fisiopatología , Ventrículos Cardíacos/diagnóstico por imagen , Ventrículos Cardíacos/patología , Microvasos/fisiopatología , Microvasos/diagnóstico por imagen , Microvasos/patología , Remodelación Vascular , Arteria Pulmonar/fisiopatología , Arteria Pulmonar/diagnóstico por imagen , Arteria Pulmonar/patología , Disfunción Ventricular Derecha/fisiopatología , Disfunción Ventricular Derecha/etiología , Disfunción Ventricular Derecha/diagnóstico por imagen , Función Ventricular Derecha , Remodelación Ventricular , Modelos Animales de Enfermedad , Miocitos Cardíacos/patología
2.
Elife ; 112022 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-36173678

RESUMEN

In allergic asthma, allergen inhalation leads to local Th2 cell activation and peribronchial inflammation. However, the mechanisms for local antigen capture and presentation remain unclear. By two-photon microscopy of the mouse lung, we established that soluble antigens in the bronchial airway lumen were efficiently captured and presented by a population of CD11c+ interstitial macrophages with high CX3CR1-GFP and MHC class II expression. We refer to these cells as Bronchus-Associated Macrophages (BAMs) based on their localization underneath the bronchial epithelium. BAMs were enriched in collagen-rich regions near some airway branchpoints, where inhaled antigens are likely to deposit. BAMs engaged in extended interactions with effector Th2 cells and promoted Th2 cytokine production. BAMs were also often in contact with dendritic cells (DCs). After exposure to inflammatory stimuli, DCs migrated to draining lymph nodes, whereas BAMs remained lung resident. We propose that BAMs act as local antigen presenting cells in the lung and also transfer antigen to DCs.


Asunto(s)
Células Dendríticas , Células Th2 , Alérgenos , Animales , Bronquios , Citocinas , Pulmón/patología , Macrófagos , Ratones
3.
Nat Commun ; 13(1): 4941, 2022 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-35999210

RESUMEN

Physiologic laminar shear stress (LSS) induces an endothelial gene expression profile that is vasculo-protective. In this report, we delineate how LSS mediates changes in the epigenetic landscape to promote this beneficial response. We show that under LSS, KLF4 interacts with the SWI/SNF nucleosome remodeling complex to increase accessibility at enhancer sites that promote the expression of homeostatic endothelial genes. By combining molecular and computational approaches we discover enhancers that loop to promoters of KLF4- and LSS-responsive genes that stabilize endothelial cells and suppress inflammation, such as BMPR2, SMAD5, and DUSP5. By linking enhancers to genes that they regulate under physiologic LSS, our work establishes a foundation for interpreting how non-coding DNA variants in these regions might disrupt protective gene expression to influence vascular disease.


Asunto(s)
Cromatina , Células Endoteliales , Cromatina/genética , Ensamble y Desensamble de Cromatina/genética , Nucleosomas/genética , Secuencias Reguladoras de Ácidos Nucleicos
4.
Cell Rep ; 36(3): 109408, 2021 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-34289374

RESUMEN

The molecular mechanisms that govern the choreographed timing of organ development remain poorly understood. Our investigation of the role of the Lin28a and Lin28b paralogs during the developmental process of branching morphogenesis establishes that dysregulation of Lin28a/b leads to abnormal branching morphogenesis in the lung and other tissues. Additionally, we find that the Lin28 paralogs, which regulate post-transcriptional processing of both mRNAs and microRNAs (miRNAs), predominantly control mRNAs during the initial phases of lung organogenesis. Target mRNAs include Sox2, Sox9, and Etv5, which coordinate lung development and differentiation. Moreover, we find that functional interactions between Lin28a and Sox9 are capable of bypassing branching defects in Lin28a/b mutant lungs. Here, we identify Lin28a and Lin28b as regulators of early embryonic lung development, highlighting the importance of the timing of post-transcriptional regulation of both miRNAs and mRNAs at distinct stages of organogenesis.


Asunto(s)
Pulmón/embriología , Pulmón/metabolismo , Morfogénesis , Proteínas de Unión al ARN/metabolismo , Homología de Secuencia de Aminoácido , Embrión de Mamíferos/metabolismo , Retroalimentación Fisiológica , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Proteínas Hedgehog/metabolismo , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Modelos Biológicos , Morfogénesis/genética , Proteínas de Unión al ARN/genética , Factor de Transcripción SOX9/metabolismo , Transducción de Señal/genética
5.
Am J Respir Cell Mol Biol ; 65(3): 272-287, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33938785

RESUMEN

Right ventricular (RV) function is the predominant determinant of survival in patients with pulmonary arterial hypertension (PAH). In preclinical models, pharmacological activation of BMP (bone morphogenetic protein) signaling with FK506 (tacrolimus) improved RV function by decreasing RV afterload. FK506 therapy further stabilized three patients with end-stage PAH. Whether FK506 has direct effects on the pressure-overloaded right ventricle is yet unknown. We hypothesized that increasing cardiac BMP signaling with FK506 improves RV structure and function in a model of fixed RV afterload after pulmonary artery banding (PAB). Direct cardiac effects of FK506 on the microvasculature and RV fibrosis were studied after surgical PAB in wild-type and heterozygous Bmpr2 mutant mice. RV function and strain were assessed longitudinally via cardiac magnetic resonance imaging during continuous FK506 infusion. Genetic lineage tracing of endothelial cells (ECs) was performed to assess the contribution of ECs to fibrosis. Molecular mechanistic studies were performed in human cardiac fibroblasts and ECs. In mice, low BMP signaling in the right ventricle exaggerated PAB-induced RV fibrosis. FK506 therapy restored cardiac BMP signaling, reduced RV fibrosis in a BMP-dependent manner independent from its immunosuppressive effect, preserved RV capillarization, and improved RV function and strain over the time course of disease. Endothelial mesenchymal transition was a rare event and did not significantly contribute to cardiac fibrosis after PAB. Mechanistically, FK506 required ALK1 in human cardiac fibroblasts as a BMPR2 co-receptor to reduce TGFß1-induced proliferation and collagen production. Our study demonstrates that increasing cardiac BMP signaling with FK506 improves RV structure and function independent from its previously described beneficial effects on pulmonary vascular remodeling.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Hipertensión Arterial Pulmonar/metabolismo , Transducción de Señal/efectos de los fármacos , Tacrolimus/farmacología , Función Ventricular Derecha/efectos de los fármacos , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Proteínas Morfogenéticas Óseas/genética , Fibroblastos/metabolismo , Fibrosis , Humanos , Masculino , Ratones , Ratones Mutantes , Miocardio/metabolismo , Hipertensión Arterial Pulmonar/tratamiento farmacológico , Hipertensión Arterial Pulmonar/genética , Transducción de Señal/genética , Función Ventricular Derecha/genética
6.
Nature ; 587(7835): 619-625, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33208946

RESUMEN

Although single-cell RNA sequencing studies have begun to provide compendia of cell expression profiles1-9, it has been difficult to systematically identify and localize all molecular cell types in individual organs to create a full molecular cell atlas. Here, using droplet- and plate-based single-cell RNA sequencing of approximately 75,000 human cells across all lung tissue compartments and circulating blood, combined with a multi-pronged cell annotation approach, we create an extensive cell atlas of the human lung. We define the gene expression profiles and anatomical locations of 58 cell populations in the human lung, including 41 out of 45 previously known cell types and 14 previously unknown ones. This comprehensive molecular atlas identifies the biochemical functions of lung cells and the transcription factors and markers for making and monitoring them; defines the cell targets of circulating hormones and predicts local signalling interactions and immune cell homing; and identifies cell types that are directly affected by lung disease genes and respiratory viruses. By comparing human and mouse data, we identified 17 molecular cell types that have been gained or lost during lung evolution and others with substantially altered expression profiles, revealing extensive plasticity of cell types and cell-type-specific gene expression during organ evolution including expression switches between cell types. This atlas provides the molecular foundation for investigating how lung cell identities, functions and interactions are achieved in development and tissue engineering and altered in disease and evolution.


Asunto(s)
Células/clasificación , Células/metabolismo , Inmunidad , Pulmón/citología , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Transcriptoma/genética , Anciano , Animales , Atlas como Asunto , Biomarcadores , Comunicación Celular , Células/inmunología , Quimiocinas/metabolismo , Células Endoteliales/metabolismo , Células Epiteliales/metabolismo , Femenino , Humanos , Pulmón/inmunología , Masculino , Ratones , Persona de Mediana Edad , Receptores Mensajeros de Linfocitos/metabolismo , Transducción de Señal , Células del Estroma/metabolismo
7.
Nature ; 586(7831): 785-789, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33057196

RESUMEN

In the mammalian lung, an apparently homogenous mesh of capillary vessels surrounds each alveolus, forming the vast respiratory surface across which oxygen transfers to the blood1. Here we use single-cell analysis to elucidate the cell types, development, renewal and evolution of the alveolar capillary endothelium. We show that alveolar capillaries are mosaics; similar to the epithelium that lines the alveolus, the alveolar endothelium is made up of two intermingled cell types, with complex 'Swiss-cheese'-like morphologies and distinct functions. The first cell type, which we term the 'aerocyte', is specialized for gas exchange and the trafficking of leukocytes, and is unique to the lung. The other cell type, termed gCap ('general' capillary), is specialized to regulate vasomotor tone, and functions as a stem/progenitor cell in capillary homeostasis and repair. The two cell types develop from bipotent progenitors, mature gradually and are affected differently in disease and during ageing. This cell-type specialization is conserved between mouse and human lungs but is not found in alligator or turtle lungs, suggesting it arose during the evolution of the mammalian lung. The discovery of cell type specialization in alveolar capillaries transforms our understanding of the structure, function, regulation and maintenance of the air-blood barrier and gas exchange in health, disease and evolution.


Asunto(s)
Capilares/citología , Alveolos Pulmonares/irrigación sanguínea , Alveolos Pulmonares/citología , Intercambio Gaseoso Pulmonar , Envejecimiento , Caimanes y Cocodrilos/anatomía & histología , Animales , Evolución Biológica , Capilares/metabolismo , División Celular , Autorrenovación de las Células , Senescencia Celular , Humanos , Masculino , Ratones , Alveolos Pulmonares/metabolismo , Células Madre/clasificación , Células Madre/citología , Tortugas/anatomía & histología
8.
Circulation ; 142(16): 1545-1561, 2020 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-32794408

RESUMEN

BACKGROUND: Pulmonary arterial hypertension (PAH) is a fatal disease characterized by profound vascular remodeling in which pulmonary arteries narrow because of medial thickening and occlusion by neointimal lesions, resulting in elevated pulmonary vascular resistance and right heart failure. Therapies targeting the neointima would represent a significant advance in PAH treatment; however, our understanding of the cellular events driving neointima formation, and the molecular pathways that control them, remains limited. METHODS: We comprehensively map the stepwise remodeling of pulmonary arteries in a robust, chronic inflammatory mouse model of pulmonary hypertension. This model demonstrates pathological features of the human disease, including increased right ventricular pressures, medial thickening, neointimal lesion formation, elastin breakdown, increased anastomosis within the bronchial circulation, and perivascular inflammation. Using genetic lineage tracing, clonal analysis, multiplexed in situ hybridization, immunostaining, deep confocal imaging, and staged pharmacological inhibition, we define the cell behaviors underlying each stage of vascular remodeling and identify a pathway required for neointima formation. RESULTS: Neointima arises from smooth muscle cells (SMCs) and not endothelium. Medial SMCs proliferate broadly to thicken the media, after which a small number of SMCs are selected to establish the neointima. These neointimal founder cells subsequently undergoing massive clonal expansion to form occlusive neointimal lesions. The normal pulmonary artery SMC population is heterogeneous, and we identify a Notch3-marked minority subset of SMCs as the major neointimal cell of origin. Notch signaling is specifically required for the selection of neointimal founder cells, and Notch inhibition significantly improves pulmonary artery pressure in animals with pulmonary hypertension. CONCLUSIONS: This work describes the first nongenetically driven murine model of pulmonary hypertension (PH) that generates robust and diffuse occlusive neointimal lesions across the pulmonary vascular bed and does so in a stereotyped timeframe. We uncover distinct cellular and molecular mechanisms underlying medial thickening and neointima formation and highlight novel transcriptional, behavioral, and pathogenic heterogeneity within pulmonary artery SMCs. In this model, inflammation is sufficient to generate characteristic vascular pathologies and physiological measures of human PAH. We hope that identifying the molecular cues regulating each stage of vascular remodeling will open new avenues for therapeutic advancements in the treatment of PAH.


Asunto(s)
Hipertensión Pulmonar/fisiopatología , Miocitos del Músculo Liso/metabolismo , Receptor Notch3/metabolismo , Remodelación Vascular/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Músculo Liso Vascular/metabolismo
9.
Cardiovasc Res ; 116(10): 1700-1709, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31738411

RESUMEN

AIMS: The temporal sequence of events underlying functional right ventricular (RV) recovery after improvement of pulmonary hypertension-associated pressure overload is unknown. We sought to establish a novel mouse model of gradual RV recovery from pressure overload and use it to delineate RV reverse-remodelling events. METHODS AND RESULTS: Surgical pulmonary artery banding (PAB) around a 26-G needle induced RV dysfunction with increased RV pressures, reduced exercise capacity and caused liver congestion, hypertrophic, fibrotic, and vascular myocardial remodelling within 5 weeks of chronic RV pressure overload in mice. Gradual reduction of the afterload burden through PA band absorption (de-PAB)-after RV dysfunction and structural remodelling were established-initiated recovery of RV function (cardiac output and exercise capacity) along with rapid normalization in RV hypertrophy (RV/left ventricular + S and cardiomyocyte area) and RV pressures (right ventricular systolic pressure). RV fibrotic (collagen, elastic fibres, and vimentin+ fibroblasts) and vascular (capillary density) remodelling were equally reversible; however, reversal occurred at a later timepoint after de-PAB, when RV function was already completely restored. Microarray gene expression (ClariomS, Thermo Fisher Scientific, Waltham, MA, USA) along with gene ontology analyses in RV tissues revealed growth factors, immune modulators, and apoptosis mediators as major cellular components underlying functional RV recovery. CONCLUSION: We established a novel gradual de-PAB mouse model and used it to demonstrate that established pulmonary hypertension-associated RV dysfunction is fully reversible. Mechanistically, we link functional RV improvement to hypertrophic normalization that precedes fibrotic and vascular reverse-remodelling events.


Asunto(s)
Hipertrofia Ventricular Derecha/fisiopatología , Arteria Pulmonar/cirugía , Disfunción Ventricular Derecha/fisiopatología , Función Ventricular Derecha , Remodelación Ventricular , Animales , Presión Arterial , Modelos Animales de Enfermedad , Tolerancia al Ejercicio , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis , Hipertrofia Ventricular Derecha/etiología , Hipertrofia Ventricular Derecha/metabolismo , Hipertrofia Ventricular Derecha/patología , Masculino , Ratones Endogámicos C57BL , Miocardio/metabolismo , Miocardio/patología , Hipertensión Arterial Pulmonar/etiología , Hipertensión Arterial Pulmonar/fisiopatología , Arteria Pulmonar/fisiopatología , Recuperación de la Función , Técnicas de Sutura , Factores de Tiempo , Disfunción Ventricular Derecha/etiología , Disfunción Ventricular Derecha/metabolismo , Disfunción Ventricular Derecha/patología
10.
Cell Syst ; 9(3): 221-227, 2019 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-31557453

RESUMEN

Many metazoan organs are comprised of branching trees of epithelial tubes; how patterning occurs in these trees is a fundamental problem of development. Commonly, branch tips fill the volume of the organ approximately uniformly, e.g., in mammalian lung, airway branch tips are dispersed roughly uniformly throughout the volume of the lung. In contrast, in the developing metanephric kidney, the tips of the ureteric bud tree are located close to the outer surface of the kidney rather than filling the kidney. Here, we describe a simple alteration in the branching rules that accounts for the difference between the kidney pattern that leads to tips near the organ surface versus previously known patterns that lead to the branch tips being dispersed throughout the organ. We further use a simple toy model to deduce from first principles how this rule change accounts for the differences in the two types of trees.


Asunto(s)
Epitelio/embriología , Riñón/embriología , Pulmón/embriología , Animales , Tipificación del Cuerpo , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica , Humanos , Riñón/anatomía & histología , Pulmón/anatomía & histología , Modelos Biológicos , Morfogénesis
11.
Am J Respir Crit Care Med ; 199(1): 83-98, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30107138

RESUMEN

RATIONALE: Pulmonary arterial hypertension (PAH) is characterized by progressive narrowing of pulmonary arteries, resulting in right heart failure and death. BMPR2 (bone morphogenetic protein receptor type 2) mutations account for most familial PAH forms whereas reduced BMPR2 is present in many idiopathic PAH forms, suggesting dysfunctional BMPR2 signaling to be a key feature of PAH. Modulating BMPR2 signaling is therapeutically promising, yet how BMPR2 is downregulated in PAH is unclear. OBJECTIVES: We intended to identify and pharmaceutically target BMPR2 modifier genes to improve PAH. METHODS: We combined siRNA high-throughput screening of >20,000 genes with a multicohort analysis of publicly available PAH RNA expression data to identify clinically relevant BMPR2 modifiers. After confirming gene dysregulation in tissue from patients with PAH, we determined the functional roles of BMPR2 modifiers in vitro and tested the repurposed drug enzastaurin for its propensity to improve experimental pulmonary hypertension (PH). MEASUREMENTS AND MAIN RESULTS: We discovered FHIT (fragile histidine triad) as a novel BMPR2 modifier. BMPR2 and FHIT expression were reduced in patients with PAH. FHIT reductions were associated with endothelial and smooth muscle cell dysfunction, rescued by enzastaurin through a dual mechanism: upregulation of FHIT as well as miR17-5 repression. Fhit-/- mice had exaggerated hypoxic PH and failed to recover in normoxia. Enzastaurin reversed PH in the Sugen5416/hypoxia/normoxia rat model, by improving right ventricular systolic pressure, right ventricular hypertrophy, cardiac fibrosis, and vascular remodeling. CONCLUSIONS: This study highlights the importance of the novel BMPR2 modifier FHIT in PH and the clinical value of the repurposed drug enzastaurin as a potential novel therapeutic strategy to improve PAH.


Asunto(s)
Ácido Anhídrido Hidrolasas/genética , Hipertensión Pulmonar Primaria Familiar/genética , Genes Modificadores/genética , Proteínas de Neoplasias/genética , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Modelos Animales de Enfermedad , Hipertensión Pulmonar Primaria Familiar/metabolismo , Femenino , Humanos , Indoles/farmacología , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
12.
Development ; 143(19): 3582-3590, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27702786

RESUMEN

cAMP-dependent protein kinase A (PKA) is a ubiquitously expressed serine/threonine kinase that regulates a variety of cellular functions. Here, we demonstrate that endothelial PKA activity is essential for vascular development, specifically regulating the transition from sprouting to stabilization of nascent vessels. Inhibition of endothelial PKA by endothelial cell-specific expression of dominant-negative PKA in mice led to perturbed vascular development, hemorrhage and embryonic lethality at mid-gestation. During perinatal retinal angiogenesis, inhibition of PKA resulted in hypersprouting as a result of increased numbers of tip cells. In zebrafish, cell autonomous PKA inhibition also increased and sustained endothelial cell motility, driving cells to become tip cells. Although these effects of PKA inhibition were highly reminiscent of Notch inhibition effects, our data demonstrate that PKA and Notch independently regulate tip and stalk cell formation and behavior.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Neovascularización Fisiológica/fisiología , Receptores Notch/metabolismo , Retina/citología , Retina/metabolismo , Animales , Movimiento Celular/genética , Movimiento Celular/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Ratones , Ratones Mutantes , Neovascularización Fisiológica/genética , Reacción en Cadena de la Polimerasa , Transducción de Señal/genética , Transducción de Señal/fisiología , Pez Cebra
13.
Cell Host Microbe ; 15(5): 636-43, 2014 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-24832456

RESUMEN

The mucosal epithelium consists of polarized cells with distinct apical and basolateral membranes that serve as functional and physical barriers to external pathogens. The apical surface of the epithelium constitutes the first point of contact between mucosal pathogens, such as Pseudomonas aeruginosa, and their host. We observed that binding of P. aeruginosa aggregates to the apical surface of polarized cells led to the striking formation of an actin-rich membrane protrusion with inverted polarity, containing basolateral lipids and membrane components. Such protrusions were associated with a spatially localized host immune response to P. aeruginosa aggregates that required bacterial flagella and a type III secretion system apparatus. Host protrusions formed de novo underneath bacterial aggregates and involved the apical recruitment of a Par3/Par6α/aPKC/Rac1 signaling module for a robust, spatially localized host NF-κB response. Our data reveal a role for spatiotemporal epithelial polarity changes in the activation of innate immune responses.


Asunto(s)
Polaridad Celular , Inmunidad Innata , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Línea Celular , Células Epiteliales/citología , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Humanos , FN-kappa B/genética , FN-kappa B/inmunología , Proteínas del Tejido Nervioso , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/inmunología , Infecciones por Pseudomonas/enzimología , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/fisiopatología , Pseudomonas aeruginosa/fisiología , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/inmunología
14.
PLoS Genet ; 8(8): e1002866, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22876201

RESUMEN

Normal development of the respiratory system is essential for survival and is regulated by multiple genes and signaling pathways. Both Tbx4 and Tbx5 are expressed throughout the mesenchyme of the developing lung and trachea; and, although multiple genes are known to be required in the epithelium, only Fgfs have been well studied in the mesenchyme. In this study, we investigated the roles of Tbx4 and Tbx5 in lung and trachea development using conditional mutant alleles and two different Cre recombinase transgenic lines. Loss of Tbx5 leads to a unilateral loss of lung bud specification and absence of tracheal specification in organ culture. Mutants deficient in Tbx4 and Tbx5 show severely reduced lung branching at mid-gestation. Concordant with this defect, the expression of mesenchymal markers Wnt2 and Fgf10, as well as Fgf10 target genes Bmp4 and Spry2, in the epithelium is downregulated. Lung branching undergoes arrest ex vivo when Tbx4 and Tbx5 are both completely lacking. Lung-specific Tbx4 heterozygous;Tbx5 conditional null mice die soon after birth due to respiratory distress. These pups have small lungs and show severe disruptions in tracheal/bronchial cartilage rings. Sox9, a master regulator of cartilage formation, is expressed in the trachea; but mesenchymal cells fail to condense and consequently do not develop cartilage normally at birth. Tbx4;Tbx5 double heterozygous mutants show decreased lung branching and fewer tracheal cartilage rings, suggesting a genetic interaction. Finally, we show that Tbx4 and Tbx5 interact with Fgf10 during the process of lung growth and branching but not during tracheal/bronchial cartilage development.


Asunto(s)
Pulmón/metabolismo , Transducción de Señal/genética , Proteínas de Dominio T Box/genética , Tráquea/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Biomarcadores/metabolismo , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Cartílago/anatomía & histología , Cartílago/embriología , Cartílago/metabolismo , Embrión de Mamíferos , Femenino , Factor 10 de Crecimiento de Fibroblastos/genética , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular , Pulmón/anatomía & histología , Pulmón/embriología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mesodermo/anatomía & histología , Mesodermo/embriología , Mesodermo/metabolismo , Ratones , Morfogénesis/genética , Técnicas de Cultivo de Órganos , Proteínas Serina-Treonina Quinasas , Proteínas de Dominio T Box/deficiencia , Tráquea/anatomía & histología , Tráquea/embriología , Proteína wnt2
15.
Science ; 333(6040): 342-345, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21764747

RESUMEN

During early lung development, airway tubes change shape. Tube length increases more than circumference as a large proportion of lung epithelial cells divide parallel to the airway longitudinal axis. We show that this bias is lost in mutants with increased extracellular signal-regulated kinase 1 (ERK1) and ERK2 activity, revealing a link between the ERK1/2 signaling pathway and the control of mitotic spindle orientation. Using a mathematical model, we demonstrate that change in airway shape can occur as a function of spindle angle distribution determined by ERK1/2 signaling, independent of effects on cell proliferation or cell size and shape. We identify sprouty genes, which encode negative regulators of fibroblast growth factor 10 (FGF10)-mediated RAS-regulated ERK1/2 signaling, as essential for controlling airway shape change during development through an effect on mitotic spindle orientation.


Asunto(s)
Pulmón/embriología , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Morfogénesis , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Mucosa Respiratoria/embriología , Huso Acromático/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Polaridad Celular , Proliferación Celular , Forma de la Célula , Tamaño de la Célula , Células Epiteliales/citología , Factor 10 de Crecimiento de Fibroblastos/genética , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Pulmón/citología , Pulmón/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Mitosis , Modelos Biológicos , Mutación , Organogénesis , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas p21(ras)/genética , Mucosa Respiratoria/citología , Huso Acromático/ultraestructura
16.
Nature ; 453(7196): 745-50, 2008 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-18463632

RESUMEN

Mammalian lungs are branched networks containing thousands to millions of airways arrayed in intricate patterns that are crucial for respiration. How such trees are generated during development, and how the developmental patterning information is encoded, have long fascinated biologists and mathematicians. However, models have been limited by a lack of information on the normal sequence and pattern of branching events. Here we present the complete three-dimensional branching pattern and lineage of the mouse bronchial tree, reconstructed from an analysis of hundreds of developmental intermediates. The branching process is remarkably stereotyped and elegant: the tree is generated by three geometrically simple local modes of branching used in three different orders throughout the lung. We propose that each mode of branching is controlled by a genetically encoded subroutine, a series of local patterning and morphogenesis operations, which are themselves controlled by a more global master routine. We show that this hierarchical and modular programme is genetically tractable, and it is ideally suited to encoding and evolving the complex networks of the lung and other branched organs.


Asunto(s)
Tipificación del Cuerpo/fisiología , Pulmón/anatomía & histología , Pulmón/embriología , Organogénesis/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Tipificación del Cuerpo/genética , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Pulmón/citología , Pulmón/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Modelos Biológicos , Organogénesis/genética , Proteínas Serina-Treonina Quinasas , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...