Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 179(2): 403-416.e23, 2019 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-31585080

RESUMEN

Pulmonary neuroendocrine (NE) cells are neurosensory cells sparsely distributed throughout the bronchial epithelium, many in innervated clusters of 20-30 cells. Following lung injury, NE cells proliferate and generate other cell types to promote epithelial repair. Here, we show that only rare NE cells, typically 2-4 per cluster, function as stem cells. These fully differentiated cells display features of classical stem cells. Most proliferate (self-renew) following injury, and some migrate into the injured area. A week later, individual cells, often just one per cluster, lose NE identity (deprogram), transit amplify, and reprogram to other fates, creating large clonal repair patches. Small cell lung cancer (SCLC) tumor suppressors regulate the stem cells: Rb and p53 suppress self-renewal, whereas Notch marks the stem cells and initiates deprogramming and transit amplification. We propose that NE stem cells give rise to SCLC, and transformation results from constitutive activation of stem cell renewal and inhibition of deprogramming.


Asunto(s)
Transformación Celular Neoplásica/patología , Neoplasias Pulmonares/patología , Pulmón/patología , Células Madre Neoplásicas/patología , Células Neuroendocrinas/patología , Receptores Notch/metabolismo , Proteína de Retinoblastoma/metabolismo , Carcinoma Pulmonar de Células Pequeñas/patología , Proteína p53 Supresora de Tumor/metabolismo , Animales , Diferenciación Celular , Transformación Celular Neoplásica/metabolismo , Lesión Pulmonar/patología , Neoplasias Pulmonares/metabolismo , Ratones , Células Madre Neoplásicas/metabolismo , Células Neuroendocrinas/metabolismo , Análisis de la Célula Individual/métodos , Carcinoma Pulmonar de Células Pequeñas/metabolismo
2.
Cell ; 179(5): 1129-1143.e23, 2019 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-31730854

RESUMEN

Energy homeostasis requires precise measurement of the quantity and quality of ingested food. The vagus nerve innervates the gut and can detect diverse interoceptive cues, but the identity of the key sensory neurons and corresponding signals that regulate food intake remains unknown. Here, we use an approach for target-specific, single-cell RNA sequencing to generate a map of the vagal cell types that innervate the gastrointestinal tract. We show that unique molecular markers identify vagal neurons with distinct innervation patterns, sensory endings, and function. Surprisingly, we find that food intake is most sensitive to stimulation of mechanoreceptors in the intestine, whereas nutrient-activated mucosal afferents have no effect. Peripheral manipulations combined with central recordings reveal that intestinal mechanoreceptors, but not other cell types, potently and durably inhibit hunger-promoting AgRP neurons in the hypothalamus. These findings identify a key role for intestinal mechanoreceptors in the regulation of feeding.


Asunto(s)
Conducta Alimentaria/fisiología , Fenómenos Genéticos , Células Receptoras Sensoriales/fisiología , Nervio Vago/fisiología , Proteína Relacionada con Agouti/metabolismo , Animales , Encéfalo/fisiología , Tracto Gastrointestinal/inervación , Marcadores Genéticos , Mecanorreceptores/metabolismo , Ratones , Nervio Vago/anatomía & histología , Vísceras/inervación
3.
Cell ; 160(1-2): 313-23, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25557078

RESUMEN

To meet the extreme oxygen demand of insect flight muscle, tracheal (respiratory) tubes ramify not only on its surface, as in other tissues, but also within T-tubules and ultimately surrounding every mitochondrion. Although this remarkable physiological specialization has long been recognized, its cellular and molecular basis is unknown. Here, we show that Drosophila tracheoles invade flight muscle T-tubules through transient surface openings. Like other tracheal branching events, invasion requires the Branchless FGF pathway. However, localization of the FGF chemoattractant changes from all muscle membranes to T-tubules as invasion begins. Core regulators of epithelial basolateral membrane identity localize to T-tubules, and knockdown of AP-1γ, required for basolateral trafficking, redirects FGF from T-tubules to surface, increasing tracheal surface ramification and preventing invasion. We propose that tracheal invasion is controlled by an AP-1-dependent switch in FGF trafficking. Thus, subcellular targeting of a chemoattractant can direct outgrowth to specific domains, including inside the cell.


Asunto(s)
Drosophila melanogaster/crecimiento & desarrollo , Factores de Crecimiento de Fibroblastos/metabolismo , Alas de Animales/embriología , Animales , Membrana Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Embrión no Mamífero/metabolismo , Larva/metabolismo , Proteínas de la Membrana/metabolismo , Células Musculares/citología , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/metabolismo , Tráquea/metabolismo
4.
Cell ; 163(2): 394-405, 2015 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-26435104

RESUMEN

Epithelial cells are normally stably anchored, maintaining their relative positions and association with the basement membrane. Developmental rearrangements occur through cell intercalation, and cells can delaminate during epithelial-mesenchymal transitions and metastasis. We mapped the formation of lung neuroepithelial bodies (NEBs), innervated clusters of neuroendocrine/neurosensory cells within the bronchial epithelium, revealing a targeted mode of cell migration that we named "slithering," in which cells transiently lose epithelial character but remain associated with the membrane while traversing neighboring epithelial cells to reach cluster sites. Immunostaining, lineage tracing, clonal analysis, and live imaging showed that NEB progenitors, initially distributed randomly, downregulate adhesion and polarity proteins, crawling over and between neighboring cells to converge at diametrically opposed positions at bronchial branchpoints, where they reestablish epithelial structure and express neuroendocrine genes. There is little accompanying progenitor proliferation or apoptosis. Activation of the slithering program may explain why lung cancers arising from neuroendocrine cells are highly metastatic.


Asunto(s)
Movimiento Celular , Pulmón/citología , Células Neuroendocrinas/citología , Células Neuroendocrinas/metabolismo , Cuerpos Neuroepiteliales/citología , Animales , Linaje de la Célula , Regulación hacia Abajo , Transición Epitelial-Mesenquimal , Pulmón/embriología , Pulmón/metabolismo , Ratones , Cuerpos Neuroepiteliales/metabolismo , Células Madre/citología , Células Madre/metabolismo
5.
Nature ; 606(7915): 739-746, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35650438

RESUMEN

The sympathetic and parasympathetic nervous systems regulate the activities of internal organs1, but the molecular and functional diversity of their constituent neurons and circuits remains largely unknown. Here we use retrograde neuronal tracing, single-cell RNA sequencing, optogenetics and physiological experiments to dissect the cardiac parasympathetic control circuit in mice. We show that cardiac-innervating neurons in the brainstem nucleus ambiguus (Amb) are comprised of two molecularly, anatomically and functionally distinct subtypes. The first, which we call ambiguus cardiovascular (ACV) neurons (approximately 35 neurons per Amb), define the classical cardiac parasympathetic circuit. They selectively innervate a subset of cardiac parasympathetic ganglion neurons and mediate the baroreceptor reflex, slowing heart rate and atrioventricular node conduction in response to increased blood pressure. The other, ambiguus cardiopulmonary (ACP) neurons (approximately 15 neurons per Amb) innervate cardiac ganglion neurons intermingled with and functionally indistinguishable from those innervated by ACV neurons. ACP neurons also innervate most or all lung parasympathetic ganglion neurons-clonal labelling shows that individual ACP neurons innervate both organs. ACP neurons mediate the dive reflex, the simultaneous bradycardia and bronchoconstriction that follows water immersion. Thus, parasympathetic control of the heart is organized into two parallel circuits, one that selectively controls cardiac function (ACV circuit) and another that coordinates cardiac and pulmonary function (ACP circuit). This new understanding of cardiac control has implications for treating cardiac and pulmonary diseases and for elucidating the control and coordination circuits of other organs.


Asunto(s)
Sistema Cardiovascular , Corazón , Pulmón , Vías Nerviosas , Sistema Nervioso Parasimpático , Animales , Corazón/fisiología , Pulmón/fisiología , Bulbo Raquídeo/citología , Bulbo Raquídeo/fisiología , Ratones , Técnicas de Trazados de Vías Neuroanatómicas , Optogenética , Sistema Nervioso Parasimpático/citología , Sistema Nervioso Parasimpático/fisiología , RNA-Seq , Análisis de la Célula Individual
6.
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
7.
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
8.
Nature ; 530(7590): 293-297, 2016 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-26855425

RESUMEN

Sighs are long, deep breaths expressing sadness, relief or exhaustion. Sighs also occur spontaneously every few minutes to reinflate alveoli, and sighing increases under hypoxia, stress, and certain psychiatric conditions. Here we use molecular, genetic, and pharmacologic approaches to identify a peptidergic sigh control circuit in murine brain. Small neural subpopulations in a key breathing control centre, the retrotrapezoid nucleus/parafacial respiratory group (RTN/pFRG), express bombesin-like neuropeptide genes neuromedin B (Nmb) or gastrin-releasing peptide (Grp). These project to the preBötzinger Complex (preBötC), the respiratory rhythm generator, which expresses NMB and GRP receptors in overlapping subsets of ~200 neurons. Introducing either neuropeptide into preBötC or onto preBötC slices, induced sighing or in vitro sigh activity, whereas elimination or inhibition of either receptor reduced basal sighing, and inhibition of both abolished it. Ablating receptor-expressing neurons eliminated basal and hypoxia-induced sighing, but left breathing otherwise intact initially. We propose that these overlapping peptidergic pathways comprise the core of a sigh control circuit that integrates physiological and perhaps emotional input to transform normal breaths into sighs.


Asunto(s)
Péptido Liberador de Gastrina/metabolismo , Neuroquinina B/análogos & derivados , Neuronas/fisiología , Receptores de Bombesina/metabolismo , Respiración , Transducción de Señal/fisiología , Animales , Bombesina/farmacología , Emociones/fisiología , Femenino , Péptido Liberador de Gastrina/deficiencia , Péptido Liberador de Gastrina/genética , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Neuroquinina B/deficiencia , Neuroquinina B/genética , Neuroquinina B/metabolismo , Neuroquinina B/farmacología , Neuronas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Respiración/efectos de los fármacos , Centro Respiratorio/citología , Centro Respiratorio/efectos de los fármacos , Centro Respiratorio/fisiología , Proteínas Inactivadoras de Ribosomas Tipo 1/farmacología , Saporinas , Transducción de Señal/efectos de los fármacos
9.
Nature ; 527(7577): 240-4, 2015 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-26560302

RESUMEN

Animals have evolved homeostatic responses to changes in oxygen availability that act on different timescales. Although the hypoxia-inducible factor (HIF) transcriptional pathway that controls long-term responses to low oxygen (hypoxia) has been established, the pathway that mediates acute responses to hypoxia in mammals is not well understood. Here we show that the olfactory receptor gene Olfr78 is highly and selectively expressed in oxygen-sensitive glomus cells of the carotid body, a chemosensory organ at the carotid artery bifurcation that monitors blood oxygen and stimulates breathing within seconds when oxygen declines. Olfr78 mutants fail to increase ventilation in hypoxia but respond normally to hypercapnia. Glomus cells are present in normal numbers and appear structurally intact, but hypoxia-induced carotid body activity is diminished. Lactate, a metabolite that rapidly accumulates in hypoxia and induces hyperventilation, activates Olfr78 in heterologous expression experiments, induces calcium transients in glomus cells, and stimulates carotid sinus nerve activity through Olfr78. We propose that, in addition to its role in olfaction, Olfr78 acts as a hypoxia sensor in the breathing circuit by sensing lactate produced when oxygen levels decline.


Asunto(s)
Ácido Láctico/metabolismo , Neuronas Receptoras Olfatorias/metabolismo , Oxígeno/metabolismo , Receptores Odorantes/metabolismo , Respiración , Animales , Señalización del Calcio , Cuerpo Carotídeo/citología , Cuerpo Carotídeo/efectos de los fármacos , Cuerpo Carotídeo/metabolismo , Seno Carotídeo/inervación , Femenino , Células HEK293 , Humanos , Hipercapnia/genética , Hipercapnia/metabolismo , Hipoxia/genética , Hipoxia/metabolismo , Ácido Láctico/farmacología , Ratones , Oxígeno/sangre , Receptores Odorantes/deficiencia
10.
Nature ; 507(7491): 190-4, 2014 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-24499815

RESUMEN

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


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

RESUMEN

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


Asunto(s)
Linaje de la Célula/genética , Células Epiteliales/citología , Células Epiteliales/metabolismo , Pulmón/citología , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Animales , Bronquios/citología , Diferenciación Celular/genética , Células Epiteliales/clasificación , Femenino , Marcadores Genéticos , Genoma/genética , Genómica , Pulmón/embriología , Ratones , Ratones Endogámicos C57BL , Alveolos Pulmonares/citología , Intercambio Gaseoso Pulmonar , Células Madre/citología , Transcriptoma/genética
12.
Development ; 143(8): 1318-27, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26952982

RESUMEN

Macrophages are specialized phagocytic cells, present in all tissues, which engulf and digest pathogens, infected and dying cells, and debris, and can recruit and regulate other immune cells and the inflammatory response and aid in tissue repair. Macrophage subpopulations play distinct roles in these processes and in disease, and are typically recognized by differences in marker expression, immune function, or tissue of residency. Although macrophage subpopulations in the brain have been found to have distinct developmental origins, the extent to which development contributes to macrophage diversity between tissues and within tissues is not well understood. Here, we investigate the development and maintenance of mouse lung macrophages by marker expression patterns, genetic lineage tracing and parabiosis. We show that macrophages populate the lung in three developmental waves, each giving rise to a distinct lineage. These lineages express different markers, reside in different locations, renew in different ways, and show little or no interconversion. Thus, development contributes significantly to lung macrophage diversity and targets each lineage to a different anatomical domain.


Asunto(s)
Pulmón/embriología , Macrófagos Alveolares/citología , Animales , Antígenos de Diferenciación , Circulación Sanguínea , Linaje de la Célula , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/citología , Pulmón/citología , Ratones , Ratones Endogámicos C57BL , Parabiosis , Saco Vitelino/citología
15.
Development ; 140(20): 4277-86, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24048590

RESUMEN

The transcriptional control of primary cilium formation and ciliary motility are beginning to be understood, but little is known about the transcriptional programs that control cilium number and other structural and functional specializations. One of the most intriguing ciliary specializations occurs in multiciliated cells (MCCs), which amplify their centrioles to nucleate hundreds of cilia per cell, instead of the usual monocilium. Here we report that the transcription factor MYB, which promotes S phase and drives cycling of a variety of progenitor cells, is expressed in postmitotic epithelial cells of the mouse airways and ependyma destined to become MCCs. MYB is expressed early in multiciliogenesis, as progenitors exit the cell cycle and amplify their centrioles, then switches off as MCCs mature. Conditional inactivation of Myb in the developing airways blocks or delays centriole amplification and expression of FOXJ1, a transcription factor that controls centriole docking and ciliary motility, and airways fail to become fully ciliated. We provide evidence that MYB acts in a conserved pathway downstream of Notch signaling and multicilin, a protein related to the S-phase regulator geminin, and upstream of FOXJ1. MYB can activate endogenous Foxj1 expression and stimulate a cotransfected Foxj1 reporter in heterologous cells, and it can drive the complete multiciliogenesis program in Xenopus embryonic epidermis. We conclude that MYB has an early, crucial and conserved role in multiciliogenesis, and propose that it promotes a novel S-like phase in which centriole amplification occurs uncoupled from DNA synthesis, and then drives later steps of multiciliogenesis through induction of Foxj1.


Asunto(s)
Centriolos/metabolismo , Cilios/metabolismo , Factores de Transcripción Forkhead/metabolismo , Proteínas Proto-Oncogénicas c-myb/metabolismo , Animales , Encéfalo/embriología , Encéfalo/metabolismo , Diferenciación Celular , Movimiento Celular , Células Cultivadas , Centriolos/genética , Cilios/genética , Epéndimo/embriología , Epéndimo/metabolismo , Células Epiteliales/metabolismo , Factores de Transcripción Forkhead/biosíntesis , Pulmón/embriología , Pulmón/metabolismo , Ratones/embriología , Ratones Transgénicos , Transducción de Señal , Tráquea/embriología , Tráquea/metabolismo , Xenopus laevis/embriología
16.
Nature ; 464(7288): 549-53, 2010 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-20336138

RESUMEN

Coronary artery disease is the leading cause of death worldwide. Determining the coronary artery developmental program could aid understanding of the disease and lead to new treatments, but many aspects of the process, including their developmental origin, remain obscure. Here we show, using histological and clonal analysis in mice and cardiac organ culture, that coronary vessels arise from angiogenic sprouts of the sinus venosus-the vein that returns blood to the embryonic heart. Sprouting venous endothelial cells dedifferentiate as they migrate over and invade the myocardium. Invading cells differentiate into arteries and capillaries; cells on the surface redifferentiate into veins. These results show that some differentiated venous cells retain developmental plasticity, and indicate that position-specific cardiac signals trigger their dedifferentiation and conversion into coronary arteries, capillaries and veins. Understanding this new reprogramming process and identifying the endogenous signals should suggest more natural ways of engineering coronary bypass grafts and revascularizing the heart.


Asunto(s)
Diferenciación Celular , Vasos Coronarios/citología , Vasos Coronarios/embriología , Animales , Biomarcadores/metabolismo , Linaje de la Célula , Regulación hacia Abajo , Células Endoteliales/citología , Regulación del Desarrollo de la Expresión Génica , Técnicas de Sustitución del Gen , Ratones , Técnicas de Cultivo de Órganos
18.
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
19.
PLoS Genet ; 7(7): e1002087, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21750678

RESUMEN

Many signaling proteins and transcription factors that induce and pattern organs have been identified, but relatively few of the downstream effectors that execute morphogenesis programs. Because such morphogenesis genes may function in many organs and developmental processes, mutations in them are expected to be pleiotropic and hence ignored or discarded in most standard genetic screens. Here we describe a systematic screen designed to identify all Drosophila third chromosome genes (∼40% of the genome) that function in development of the tracheal system, a tubular respiratory organ that provides a paradigm for branching morphogenesis. To identify potentially pleiotropic morphogenesis genes, the screen included analysis of marked clones of homozygous mutant tracheal cells in heterozygous animals, plus a secondary screen to exclude mutations in general "house-keeping" genes. From a collection including more than 5,000 lethal mutations, we identified 133 mutations representing ∼70 or more genes that subdivide the tracheal terminal branching program into six genetically separable steps, a previously established cell specification step plus five major morphogenesis and maturation steps: branching, growth, tubulogenesis, gas-filling, and maintenance. Molecular identification of 14 of the 70 genes demonstrates that they include six previously known tracheal genes, each with a novel function revealed by clonal analysis, and two well-known growth suppressors that establish an integral role for cell growth control in branching morphogenesis. The rest are new tracheal genes that function in morphogenesis and maturation, many through cytoskeletal and secretory pathways. The results suggest systematic genetic screens that include clonal analysis can elucidate the full organogenesis program and that over 200 patterning and morphogenesis genes are required to build even a relatively simple organ such as the Drosophila tracheal system.


Asunto(s)
Proteínas de Drosophila/genética , Drosophila melanogaster , Genes de Insecto , Tráquea/crecimiento & desarrollo , Factores de Transcripción/genética , Animales , Animales Modificados Genéticamente , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Genes Letales , Heterocigoto , Homocigoto , Morfogénesis , Mutación , Factores de Transcripción/metabolismo
20.
Nat Commun ; 15(1): 2188, 2024 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-38467625

RESUMEN

Hormones mediate long-range cell communication and play vital roles in physiology, metabolism, and health. Traditionally, endocrinologists have focused on one hormone or organ system at a time. Yet, hormone signaling by its very nature connects cells of different organs and involves crosstalk of different hormones. Here, we leverage the organism-wide single cell transcriptional atlas of a non-human primate, the mouse lemur (Microcebus murinus), to systematically map source and target cells for 84 classes of hormones. This work uncovers previously-uncharacterized sites of hormone regulation, and shows that the hormonal signaling network is densely connected, decentralized, and rich in feedback loops. Evolutionary comparisons of hormonal genes and their expression patterns show that mouse lemur better models human hormonal signaling than mouse, at both the genomic and transcriptomic levels, and reveal primate-specific rewiring of hormone-producing/target cells. This work complements the scale and resolution of classical endocrine studies and sheds light on primate hormone regulation.


Asunto(s)
Cheirogaleidae , Animales , Cheirogaleidae/genética , Cheirogaleidae/metabolismo , Transcriptoma/genética , Evolución Biológica , Hormonas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA