Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Exp Med ; 221(6)2024 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-38597954

RESUMO

Early stages of deadly respiratory diseases including COVID-19 are challenging to elucidate in humans. Here, we define cellular tropism and transcriptomic effects of SARS-CoV-2 virus by productively infecting healthy human lung tissue and using scRNA-seq to reconstruct the transcriptional program in "infection pseudotime" for individual lung cell types. SARS-CoV-2 predominantly infected activated interstitial macrophages (IMs), which can accumulate thousands of viral RNA molecules, taking over 60% of the cell transcriptome and forming dense viral RNA bodies while inducing host profibrotic (TGFB1, SPP1) and inflammatory (early interferon response, CCL2/7/8/13, CXCL10, and IL6/10) programs and destroying host cell architecture. Infected alveolar macrophages (AMs) showed none of these extreme responses. Spike-dependent viral entry into AMs used ACE2 and Sialoadhesin/CD169, whereas IM entry used DC-SIGN/CD209. These results identify activated IMs as a prominent site of viral takeover, the focus of inflammation and fibrosis, and suggest targeting CD209 to prevent early pathology in COVID-19 pneumonia. This approach can be generalized to any human lung infection and to evaluate therapeutics.


Assuntos
COVID-19 , Humanos , SARS-CoV-2 , Macrófagos , Inflamação , RNA Viral , Pulmão
2.
bioRxiv ; 2023 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-36711554

RESUMO

Neural activity is increasingly recognized as a critical regulator of cancer growth. In the brain, neuronal activity robustly influences glioma growth both through paracrine mechanisms and through electrochemical integration of malignant cells into neural circuitry via neuron-to-glioma synapses, while perisynaptic neurotransmitter signaling drives breast cancer brain metastasis growth. Outside of the CNS, innervation of tumors such as prostate, breast, pancreatic and gastrointestinal cancers by peripheral nerves similarly regulates cancer progression. However, the extent to which the nervous system regulates lung cancer progression, either in the lung or when metastatic to brain, is largely unexplored. Small cell lung cancer (SCLC) is a lethal high-grade neuroendocrine tumor that exhibits a strong propensity to metastasize to the brain. Here we demonstrate that, similar to glioma, metastatic SCLC cells in the brain co-opt neuronal activity-regulated mechanisms to stimulate growth and progression. Optogenetic stimulation of cortical neuronal activity drives proliferation and invasion of SCLC brain metastases. In the brain, SCLC cells exhibit electrical currents and consequent calcium transients in response to neuronal activity, and direct SCLC cell membrane depolarization is sufficient to promote the growth of SCLC tumors. In the lung, vagus nerve transection markedly inhibits primary lung tumor formation, progression and metastasis, highlighting a critical role for innervation in overall SCLC initiation and progression. Taken together, these studies illustrate that neuronal activity plays a crucial role in dictating SCLC pathogenesis in both primary and metastatic sites.

3.
Elife ; 112022 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-36469459

RESUMO

Pulmonary neuroendocrine cells (PNECs) are sensory epithelial cells that transmit airway status to the brain via sensory neurons and locally via calcitonin gene-related peptide (CGRP) and γ- aminobutyric acid (GABA). Several other neuropeptides and neurotransmitters have been detected in various species, but the number, targets, functions, and conservation of PNEC signals are largely unknown. We used scRNAseq to profile hundreds of the rare mouse and human PNECs. This revealed over 40 PNEC neuropeptide and peptide hormone genes, most cells expressing unique combinations of 5-18 genes. Peptides are packaged in separate vesicles, their release presumably regulated by the distinct, multimodal combinations of sensors we show are expressed by each PNEC. Expression of the peptide receptors predicts an array of local cell targets, and we show the new PNEC signal angiotensin directly activates one subtype of innervating sensory neuron. Many signals lack lung targets so may have endocrine activity like those of PNEC-derived carcinoid tumors. PNECs are an extraordinarily rich and diverse signaling hub rivaling the enteroendocrine system.


Assuntos
Pulmão , Células Neuroendócrinas , Neuropeptídeos , Animais , Humanos , Camundongos , Células Epiteliais/metabolismo , Pulmão/patologia , Células Neuroendócrinas/metabolismo , Neuropeptídeos/metabolismo , Análise de Sequência de RNA
4.
Arthritis Rheumatol ; 74(7): 1271-1283, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35189047

RESUMO

OBJECTIVE: Recent observations in systemic juvenile idiopathic arthritis (JIA) suggest an increasing incidence of high-mortality interstitial lung disease often characterized by a variant of pulmonary alveolar proteinosis (PAP). Co-occurrence of macrophage activation syndrome (MAS) and PAP in systemic JIA suggests a shared pathology, but patients with lung disease associated with systemic JIA (designated SJIA-LD) also commonly experience features of drug reaction such as atypical rashes and eosinophilia. This study was undertaken to investigate immunopathology and identify biomarkers in systemic JIA, MAS, and SJIA-LD. METHODS: We used SOMAscan to measure ~1,300 analytes in sera from healthy controls and patients with systemic JIA, MAS, SJIA-LD, or other related diseases. We verified selected findings by enzyme-linked immunosorbent assay and lung immunostaining. Because the proteome of a sample may reflect multiple states (systemic JIA, MAS, or SJIA-LD), we used regression modeling to identify subsets of altered proteins associated with each state. We tested key findings in a validation cohort. RESULTS: Proteome alterations in active systemic JIA and MAS overlapped substantially, including known systemic JIA biomarkers such as serum amyloid A and S100A9, and novel elevations in the levels of heat-shock proteins and glycolytic enzymes. Interleukin-18 levels were elevated in all systemic JIA groups, particularly MAS and SJIA-LD. We also identified an MAS-independent SJIA-LD signature notable for elevated levels of intercellular adhesion molecule 5 (ICAM-5), matrix metalloproteinase 7 (MMP-7), and allergic/eosinophilic chemokines, which have been previously associated with lung damage. Immunohistochemistry localized ICAM-5 and MMP-7 in the lungs of patients with SJIA-LD. The ability of ICAM-5 to distinguish SJIA-LD from systemic JIA/MAS was independently validated. CONCLUSION: Serum proteins support a systemic JIA-to-MAS continuum; help distinguish systemic JIA, systemic JIA/MAS, and SJIA-LD; and suggest etiologic hypotheses. Select biomarkers, such as ICAM-5, could aid in early detection and management of SJIA-LD.


Assuntos
Artrite Juvenil , Pneumopatias , Síndrome de Ativação Macrofágica , Artrite Juvenil/complicações , Biomarcadores , Humanos , Pneumopatias/epidemiologia , Metaloproteinase 7 da Matriz , Proteoma
5.
Comp Med ; 71(3): 256-266, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-34082858

RESUMO

The gray mouse lemur (Microcebus murinus, GML) is a nocturnal, arboreal, prosimian primate that is native to Madagascar. Captive breeding colonies of GMLs have been established primarily for noninvasive studies on questions related to circadian rhythms and metabolism. GMLs are increasingly considered to be a strong translational model for neurocognitive aging due to overlapping histopathologic features shared with aged humans. However, little information is available describing the clinical presentations, naturally occurring diseases, and histopathology of aged GMLs. In our colony, a 9 y-old, male, GML was euthanized after sudden onset of weakness, lethargy, and tibial fracture. Evaluation of this animal revealed widespread fibrous osteodystrophy (FOD) of the mandible, maxilla, cranium, appendicular, and vertebral bones. FOD and systemic metastatic mineralization were attributed to underlying chronic renal disease. Findings in this GML prompted periodic colony-wide serum biochemical screenings for azotemia and electrolyte abnormalities. Subsequently, 3 additional GMLs (2 females and 1 male) were euthanized due to varying clinical and serum biochemical presentations. Common to all 4 animals were FOD, chronic renal disease, uterine adenocarcinoma (females only), cataracts, and osteoarthritis. This case study highlights the concurrent clinical and histopathologic abnormalities that are relevant to use of GMLs in the expanding field of aging research.


Assuntos
Adenocarcinoma , Cheirogaleidae , Insuficiência Renal Crônica , Adenocarcinoma/veterinária , Envelhecimento , Animais , Ritmo Circadiano , Feminino , Masculino , Camundongos
6.
Nature ; 587(7835): 619-625, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33208946

RESUMO

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.


Assuntos
Células/classificação , Células/metabolismo , Imunidade , Pulmão/citologia , Análise de Sequência de RNA , Análise de Célula Única , Transcriptoma/genética , Idoso , Animais , Atlas como Assunto , Biomarcadores , Comunicação Celular , Células/imunologia , Quimiocinas/metabolismo , Células Endoteliais/metabolismo , Células Epiteliais/metabolismo , Feminino , Humanos , Pulmão/imunologia , Masculino , Camundongos , Pessoa de Meia-Idade , Receptores de Retorno de Linfócitos/metabolismo , Transdução de Sinais , Células Estromais/metabolismo
7.
J Thorac Oncol ; 15(4): 520-540, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32018053

RESUMO

The outcomes of patients with SCLC have not yet been substantially impacted by the revolution in precision oncology, primarily owing to a paucity of genetic alterations in actionable driver oncogenes. Nevertheless, systemic therapies that include immunotherapy are beginning to show promise in the clinic. Although, these results are encouraging, many patients do not respond to, or rapidly recur after, current regimens, necessitating alternative or complementary therapeutic strategies. In this review, we discuss ongoing investigations into the pathobiology of this recalcitrant cancer and the therapeutic vulnerabilities that are exposed by the disease state. Included within this discussion, is a snapshot of the current biomarker and clinical trial landscapes for SCLC. Finally, we identify key knowledge gaps that should be addressed to advance the field in pursuit of reduced SCLC mortality. This review largely summarizes work presented at the Third Biennial International Association for the Study of Lung Cancer SCLC Meeting.


Assuntos
Neoplasias Pulmonares , Carcinoma de Pequenas Células do Pulmão , Humanos , Laboratórios , Neoplasias Pulmonares/terapia , Recidiva Local de Neoplasia , Medicina de Precisão , Carcinoma de Pequenas Células do Pulmão/terapia
8.
Ann N Y Acad Sci ; 1462(1): 27-36, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31655007

RESUMO

Adult stem cells are rare, undifferentiated cells found in all tissues of the body. Although normally kept in a quiescent, nondividing state, these cells can proliferate and differentiate to replace naturally dying cells within their tissue and to repair its wounds in response to injury. Due to their proliferative nature and ability to regenerate tissue, adult stem cells have the potential to treat a variety of degenerative diseases as well as aging. In addition, since stem cells are often thought to be the source of malignant tumors, understanding the mechanisms that keep their proliferative abilities in check can pave the way for new cancer therapies. While adult stem cells have had limited practical and clinical applications to date, several clinical trials of stem cell-based therapies are underway. This report details recent research presented at the New York Academy of Sciences on March 14, 2019 on understanding the factors that regulate stem cell activity and differentiation, with the hope of translating these findings into the clinic.


Assuntos
Células-Tronco Adultas/transplante , Medicina Regenerativa/tendências , Relatório de Pesquisa/tendências , Transplante de Células-Tronco/tendências , Adulto , Células-Tronco Adultas/fisiologia , Envelhecimento/patologia , Animais , Diferenciação Celular/fisiologia , Humanos , Neoplasias/patologia , Neoplasias/terapia , Cidade de Nova Iorque , Medicina Regenerativa/métodos , Transplante de Células-Tronco/métodos
9.
Cell ; 179(2): 403-416.e23, 2019 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-31585080

RESUMO

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.


Assuntos
Transformação Celular Neoplásica/patologia , Neoplasias Pulmonares/patologia , Pulmão/patologia , Células-Tronco Neoplásicas/patologia , Células Neuroendócrinas/patologia , Receptores Notch/metabolismo , Proteína do Retinoblastoma/metabolismo , Carcinoma de Pequenas Células do Pulmão/patologia , Proteína Supressora de Tumor p53/metabolismo , Animais , Diferenciação Celular , Transformação Celular Neoplásica/metabolismo , Lesão Pulmonar/patologia , Neoplasias Pulmonares/metabolismo , Camundongos , Células-Tronco Neoplásicas/metabolismo , Células Neuroendócrinas/metabolismo , Análise de Célula Única/métodos , Carcinoma de Pequenas Células do Pulmão/metabolismo
11.
Science ; 359(6380): 1118-1123, 2018 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-29420258

RESUMO

Alveoli, the lung's respiratory units, are tiny sacs where oxygen enters the bloodstream. They are lined by flat alveolar type 1 (AT1) cells, which mediate gas exchange, and AT2 cells, which secrete surfactant. Rare AT2s also function as alveolar stem cells. We show that AT2 lung stem cells display active Wnt signaling, and many of them are near single, Wnt-expressing fibroblasts. Blocking Wnt secretion depletes these stem cells. Daughter cells leaving the Wnt niche transdifferentiate into AT1s: Maintaining Wnt signaling prevents transdifferentiation, whereas abrogating Wnt signaling promotes it. Injury induces AT2 autocrine Wnts, recruiting "bulk" AT2s as progenitors. Thus, individual AT2 stem cells reside in single-cell fibroblast niches providing juxtacrine Wnts that maintain them, whereas injury induces autocrine Wnts that transiently expand the progenitor pool. This simple niche maintains the gas exchange surface and is coopted in cancer.


Assuntos
Transdiferenciação Celular , Alvéolos Pulmonares/citologia , Nicho de Células-Tronco/fisiologia , Células-Tronco/citologia , Via de Sinalização Wnt , Animais , Fibroblastos/citologia , Fibroblastos/metabolismo , Pulmão/fisiologia , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Alvéolos Pulmonares/metabolismo , Células-Tronco/metabolismo
12.
Cell Rep ; 20(7): 1533-1542, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28813666

RESUMO

In the developing brain, neurons expressing VEGF-A and blood vessels grow in close apposition, but many of the molecular pathways regulating neuronal VEGF-A and neurovascular system development remain to be deciphered. Here, we show that miR-9 links neurogenesis and angiogenesis through the formation of neurons expressing VEGF-A. We found that miR-9 directly targets the transcription factors TLX and ONECUTs to regulate VEGF-A expression. miR-9 inhibition leads to increased TLX and ONECUT expression, resulting in VEGF-A overexpression. This untimely increase of neuronal VEGF-A signal leads to the thickening of blood vessels at the expense of the normal formation of the neurovascular network in the brain and retina. Thus, this conserved transcriptional cascade is critical for proper brain development in vertebrates. Because of this dual role on neural stem cell proliferation and angiogenesis, miR-9 and its downstream targets are promising factors for cellular regenerative therapy following stroke and for brain tumor treatment.


Assuntos
Córtex Cerebral/metabolismo , MicroRNAs/genética , Neovascularização Fisiológica/genética , Células-Tronco Neurais/metabolismo , Neurogênese/genética , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Sequência de Bases , Sítios de Ligação , Diferenciação Celular , Proliferação de Células , Córtex Cerebral/crescimento & desenvolvimento , Embrião não Mamífero , Feto , Regulação da Expressão Gênica no Desenvolvimento , Fator 6 Nuclear de Hepatócito/genética , Fator 6 Nuclear de Hepatócito/metabolismo , Humanos , MicroRNAs/metabolismo , Morfogênese/genética , Células-Tronco Neurais/citologia , Neurônios/metabolismo , Neurônios/patologia , Receptores Nucleares Órfãos , Cultura Primária de Células , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Retina/crescimento & desenvolvimento , Retina/metabolismo , Transdução de Sinais , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Peixe-Zebra
13.
Development ; 143(8): 1318-27, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26952982

RESUMO

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.


Assuntos
Pulmão/embriologia , Macrófagos Alveolares/citologia , Animais , Antígenos de Diferenciação , Circulação Sanguínea , Linhagem da Célula , Perfilação da Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Pulmão/citologia , Camundongos , Camundongos Endogâmicos C57BL , Parabiose , Saco Vitelino/citologia
15.
Cell ; 163(2): 394-405, 2015 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-26435104

RESUMO

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.


Assuntos
Movimento Celular , Pulmão/citologia , Células Neuroendócrinas/citologia , Células Neuroendócrinas/metabolismo , Corpos Neuroepiteliais/citologia , Animais , Linhagem da Célula , Regulação para Baixo , Transição Epitelial-Mesenquimal , Pulmão/embriologia , Pulmão/metabolismo , Camundongos , Corpos Neuroepiteliais/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo
16.
Science ; 346(6211): 1258810, 2014 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-25395543

RESUMO

Most vertebrate organs are composed of epithelium surrounded by support and stromal tissues formed from mesenchyme cells, which are not generally thought to form organized progenitor pools. Here, we use clonal cell labeling with multicolor reporters to characterize individual mesenchymal progenitors in the developing mouse lung. We observe a diversity of mesenchymal progenitor populations with different locations, movements, and lineage boundaries. Airway smooth muscle (ASM) progenitors map exclusively to mesenchyme ahead of budding airways. Progenitors recruited from these tip pools differentiate into ASM around airway stalks; flanking stalk mesenchyme can be induced to form an ASM niche by a lateral bud or by an airway tip plus focal Wnt signal. Thus, mesenchymal progenitors can be organized into localized and carefully controlled domains that rival epithelial progenitor niches in regulatory sophistication.


Assuntos
Pulmão/crescimento & desenvolvimento , Células-Tronco Mesenquimais/fisiologia , Nicho de Células-Tronco/fisiologia , Animais , Diferenciação Celular , Linhagem da Célula , Movimento Celular , Proliferação de Células , Rastreamento de Células , Células Clonais , Pulmão/citologia , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Transgênicos , Análise de Célula Única/métodos , Via de Sinalização Wnt
17.
Nat Commun ; 5: 3923, 2014 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-24879355

RESUMO

The lung is a branched tubular network with two distinct compartments--the proximal conducting airways and the peripheral gas exchange region--separated by a discrete boundary termed the bronchoalveolar duct junction (BADJ). Here we image the developing mouse lung in three-dimensions (3D) and show that two nested developmental waves demarcate the BADJ under the control of a global hormonal signal. A first wave of branching morphogenesis progresses throughout embryonic development, generating branches for both compartments. A second wave of conducting airway differentiation follows the first wave but terminates earlier, specifying the proximal compartment and setting the BADJ. The second wave is terminated by a glucocorticoid signalling: premature activation or loss of glucocorticoid signalling causes a proximal or distal shift, respectively, in BADJ location. The results demonstrate a new mechanism of boundary formation in complex, 3D organs and provide new insights into glucocorticoid therapies for lung defects in premature birth.


Assuntos
Brônquios/embriologia , Alvéolos Pulmonares/embriologia , Actinas/metabolismo , Animais , Brônquios/anatomia & histologia , Brônquios/metabolismo , Caderinas/metabolismo , Diferenciação Celular , Glucocorticoides/metabolismo , Imageamento Tridimensional , Imuno-Histoquímica , Peptídeos e Proteínas de Sinalização Intercelular , Pulmão/embriologia , Pulmão/metabolismo , Camundongos , Morfogênese , Peptídeos/metabolismo , Alvéolos Pulmonares/anatomia & histologia , Alvéolos Pulmonares/metabolismo , Proteína C Associada a Surfactante Pulmonar , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Transdução de Sinais , Uteroglobina/metabolismo
18.
Nature ; 509(7500): 371-5, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24739965

RESUMO

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.


Assuntos
Linhagem da Célula/genética , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Pulmão/citologia , Análise de Sequência de RNA/métodos , Análise de Célula Única/métodos , Animais , Brônquios/citologia , Diferenciação Celular/genética , Células Epiteliais/classificação , Feminino , Marcadores Genéticos , Genoma/genética , Genômica , Pulmão/embriologia , Camundongos , Camundongos Endogâmicos C57BL , Alvéolos Pulmonares/citologia , Troca Gasosa Pulmonar , Células-Tronco/citologia , Transcriptoma/genética
19.
Nature ; 507(7491): 190-4, 2014 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-24499815

RESUMO

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.


Assuntos
Neoplasias Pulmonares/patologia , Pulmão/citologia , Pulmão/crescimento & desenvolvimento , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/patologia , Alvéolos Pulmonares/citologia , Regeneração , Animais , Diferenciação Celular , Divisão Celular , Linhagem da Célula , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Células Cultivadas , Reprogramação Celular , Células Clonais/citologia , Receptores ErbB/metabolismo , Feminino , Pulmão/embriologia , Pulmão/patologia , Neoplasias Pulmonares/metabolismo , Masculino , Camundongos , Modelos Biológicos , Células-Tronco Multipotentes/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA