Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
2.
Elife ; 112022 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-36178196

RESUMO

Basal cells are multipotent stem cells of a variety of organs, including the respiratory tract, where they are major components of the airway epithelium. However, it remains unclear how diverse basal cells are and how distinct subpopulations respond to airway challenges. Using single cell RNA-sequencing and functional approaches, we report a significant and previously underappreciated degree of heterogeneity in the basal cell pool, leading to identification of six subpopulations in the adult murine trachea. Among these, we found two major subpopulations, collectively comprising the most uncommitted of all the pools, but with distinct gene expression signatures. Notably, these occupy distinct ventral and dorsal tracheal niches and differ in their ability to self-renew and initiate a program of differentiation in response to environmental perturbations in primary cultures and in mouse injury models in vivo. We found that such heterogeneity is acquired prenatally, when the basal cell pool and local niches are still being established, and depends on the integrity of these niches, as supported by the altered basal cell phenotype of tracheal cartilage-deficient mouse mutants. Finally, we show that features that distinguish these progenitor subpopulations in murine airways are conserved in humans. Together, the data provide novel insights into the origin and impact of basal cell heterogeneity on the establishment of regionally distinct responses of the airway epithelium during injury-repair and in disease conditions.


Assuntos
Células Epiteliais , Mucosa Respiratória , Humanos , Adulto , Camundongos , Animais , Células Epiteliais/metabolismo , Diferenciação Celular/fisiologia , Traqueia/metabolismo , RNA/metabolismo
3.
Elife ; 112022 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-36129169

RESUMO

Viral infection often causes severe damage to the lungs, leading to the appearance of ectopic basal cells (EBCs) and tuft cells in the lung parenchyma. Thus far, the roles of these ectopic epithelial cells in alveolar regeneration remain controversial. Here, we confirm that the ectopic tuft cells are originated from EBCs in mouse models and COVID-19 lungs. The differentiation of tuft cells from EBCs is promoted by Wnt inhibition while suppressed by Notch inhibition. Although progenitor functions have been suggested in other organs, pulmonary tuft cells don't proliferate or give rise to other cell lineages. Consistent with previous reports, Trp63CreERT2 and KRT5-CreERT2-labeled ectopic EBCs do not exhibit alveolar regeneration potential. Intriguingly, when tamoxifen was administrated post-viral infection, Trp63CreERT2 but not KRT5-CreERT2 labels islands of alveolar epithelial cells that are negative for EBC biomarkers. Furthermore, germline deletion of Trpm5 significantly increases the contribution of Trp63CreERT2-labeled cells to the alveolar epithelium. Although Trpm5 is known to regulate tuft cell development, complete ablation of tuft cell production fails to improve alveolar regeneration in Pou2f3-/- mice, implying that Trpm5 promotes alveolar epithelial regeneration through a mechanism independent of tuft cells.


Assuntos
COVID-19 , Animais , Biomarcadores , Diferenciação Celular , Linhagem da Célula , Células Epiteliais , Camundongos , Tamoxifeno/farmacologia , Transativadores
4.
Chest ; 162(6): 1328-1337, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35753385

RESUMO

BACKGROUND: Intravitreal bevacizumab (IVB), an anti-vascular endothelial growth factor (VEGF) antibody, is a widely adopted treatment for retinopathy of prematurity (ROP). Although animal studies have demonstrated that IVB inhibits alveologenesis in neonatal rat lung, the clinical influence of IVB on respiratory outcomes has not been studied. RESEARCH QUESTION: Does IVB affect the respiratory outcome in preterm infants with bronchopulmonary dysplasia? STUDY DESIGN AND METHODS: We retrospectively assessed very low birth weight (VLBW) preterm infants admitted to our neonatal ICU between January 2016 and June 2021. Furthermore, we evaluated the short-term respiratory outcomes after IVB therapy in VLBW preterm infants requiring ventilatory support at 36 weeks' postmenstrual age (PMA). RESULTS: One hundred seventy-four VLBW preterm infants with bronchopulmonary dysplasia were recruited. Eighty-eight infants showed ROP onset before being ventilator free, and 78 infants received a diagnosis of the most severe ROP before being ventilator free. Among them, 32 received a diagnosis with type 1 ROP and received IVB treatment. After adjusting for gestational age, birth body weight, and baseline respiratory status, we discovered that IVB is associated significantly with prolonged ventilatory support and a lower likelihood of becoming ventilator free (hazard ratio, 0.53; P = .03). INTERPRETATION: IVB may have a short-term respiratory adverse effect in patients requiring ventilatory support at 36 weeks' PMA. Therefore, long-term follow-up for respiratory outcomes may be considered in VLBW infants who receive IVB treatment.


Assuntos
Displasia Broncopulmonar , Retinopatia da Prematuridade , Recém-Nascido , Humanos , Bevacizumab/uso terapêutico , Displasia Broncopulmonar/terapia , Injeções Intravítreas , Inibidores da Angiogênese/uso terapêutico , Recém-Nascido Prematuro , Estudos Retrospectivos , Retinopatia da Prematuridade/diagnóstico , Retinopatia da Prematuridade/tratamento farmacológico , Idade Gestacional
5.
Mol Biol Cell ; 32(20): ar1, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34260288

RESUMO

Multiciliated cells play critical roles in the airway, reproductive organs, and brain. Generation of multiple cilia requires both activation of a specialized transcriptional program and subsequent massive amplification of centrioles within the cytoplasm. The E2F4 transcription factor is required for both roles and consequently for multiciliogenesis. Here we establish that E2F4 associates with two distinct components of the centriole replication machinery, Deup1 and SAS6, targeting nonhomologous domains in these proteins. We map Deup1 and SAS6 binding to E2F4's N-terminus and show that this domain is sufficient to mediate E2F4's cytoplasmic role in multiciliogenesis. This sequence is highly conserved across the E2F family, but the ability to bind Deup1 and SAS6 is specific to E2F4 and E2F5, consistent with their shared roles in multiciliogenesis. By generating E2F4/E2F1 chimeras, we identify a six-residue motif that is critical for Deup1 and SAS6 binding. We propose that the ability of E2F4 and E2F5 to recruit Deup1 and/or SAS6, and enable centriole replication, contributes to their cytoplasmic roles in multiciliogenesis.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Fator de Transcrição E2F4/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Comunicação Celular/fisiologia , Ciclo Celular/fisiologia , Centríolos/metabolismo , Cílios/metabolismo , Citoplasma/metabolismo , Citosol/metabolismo , Células HEK293 , Humanos , Ligação Proteica , Domínios Proteicos
6.
Sci Rep ; 11(1): 10799, 2021 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-34031475

RESUMO

The impact of prematurity on human development and neonatal diseases, such as bronchopulmonary dysplasia, has been widely reported. However, little is known about the effects of prematurity on the programs of stem cell self-renewal and differentiation of the upper respiratory epithelium, which is key for adaptation to neonatal life. We developed a minimally invasive methodology for isolation of neonatal basal cells from nasopharyngeal (NP) aspirates and performed functional analysis in organotypic cultures to address this issue. We show that preterm NP progenitors have a markedly distinct molecular signature of abnormal proliferation and mitochondria quality control compared to term progenitors. Preterm progenitors had lower oxygen consumption at baseline and were unable to ramp up consumption to the levels of term cells when challenged. Although they formed a mucociliary epithelium, ciliary function tended to decline in premature cells as they differentiated, compared to term cells. Together, these differences suggested increased sensitivity of preterm progenitors to environmental stressors under non-homeostatic conditions.


Assuntos
Displasia Broncopulmonar/patologia , Perfilação da Expressão Gênica/métodos , Nasofaringe/citologia , Oxigênio/metabolismo , Células-Tronco/citologia , Displasia Broncopulmonar/genética , Técnicas de Cultura de Células , Diferenciação Celular , Células Cultivadas , Regulação da Expressão Gênica , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Nasofaringe/metabolismo , Análise de Sequência de RNA , Células-Tronco/metabolismo
7.
Cell Rep ; 35(3): 109011, 2021 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-33882306

RESUMO

Pulmonary neuroendocrine cells (PNECs) have crucial roles in airway physiology and immunity by producing bioactive amines and neuropeptides (NPs). A variety of human diseases exhibit PNEC hyperplasia. Given accumulated evidence that PNECs represent a heterogenous population of cells, we investigate how PNECs differ, whether the heterogeneity is similarly present in mouse and human cells, and whether specific disease involves discrete PNECs. Herein, we identify three distinct types of PNECs in human and mouse airways based on single and double positivity for TUBB3 and the established NP markers. We show that the three PNEC types exhibit significant differences in NP expression, homeostatic turnover, and response to injury and disease. We provide evidence that these differences parallel their distinct cell of origin from basal stem cells (BSCs) or other airway epithelial progenitors.


Assuntos
Linhagem da Célula/genética , Células Epiteliais/patologia , Células Neuroendócrinas/patologia , Células-Tronco/patologia , Tubulina (Proteína)/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , Células Epiteliais/classificação , Células Epiteliais/metabolismo , Feminino , Regulação da Expressão Gênica , Humanos , Hiperplasia/genética , Hiperplasia/metabolismo , Hiperplasia/patologia , Lactente , Vírus da Influenza A Subtipo H1N1/crescimento & desenvolvimento , Vírus da Influenza A Subtipo H1N1/patogenicidade , Pulmão , Masculino , Camundongos , Camundongos Transgênicos , Células Neuroendócrinas/classificação , Células Neuroendócrinas/metabolismo , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/virologia , Transdução de Sinais , Células-Tronco/classificação , Células-Tronco/metabolismo , Morte Súbita do Lactente/genética , Morte Súbita do Lactente/patologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Tubulina (Proteína)/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
8.
Wiley Interdiscip Rev Dev Biol ; 9(3): e371, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31828974

RESUMO

The Hippo pathway has emerged as a crucial integrator of signals in biological events from development to adulthood and in diseases. Although extensively studied in Drosophila and in cell cultures, major gaps of knowledge still remain on how this pathway functions in mammalian systems. The pathway consists of a growing number of components, including core kinases and adaptor proteins, which control the subcellular localization of the transcriptional co-activators Yap and Taz through phosphorylation of serines at key sites. When localized to the nucleus, Yap/Taz interact with TEAD transcription factors to induce transcriptional programs of proliferation, stemness, and growth. In the cytoplasm, Yap/Taz interact with multiple pathways to regulate a variety of cellular functions or are targeted for degradation. The Hippo pathway receives cues from diverse intracellular and extracellular inputs, including growth factor and integrin signaling, polarity complexes, and cell-cell junctions. This review highlights the mechanisms of regulation of Yap/Taz nucleocytoplasmic shuttling and their implications for epithelial cell behavior using the lung as an intriguing example of this paradigm. This article is categorized under: Gene Expression and Transcriptional Hierarchies > Regulatory Mechanisms Signaling Pathways > Cell Fate Signaling Establishment of Spatial and Temporal Patterns > Cytoplasmic Localization.


Assuntos
Proteínas de Drosophila/metabolismo , Células Epiteliais/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transativadores/metabolismo , Animais , Diferenciação Celular , Proteínas de Drosophila/genética , Drosophila melanogaster , Células Epiteliais/citologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas Nucleares/genética , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais , Transativadores/genética , Proteínas de Sinalização YAP
9.
Nat Med ; 25(11): 1691-1698, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31700187

RESUMO

Millions of people worldwide with incurable end-stage lung disease die because of inadequate treatment options and limited availability of donor organs for lung transplantation1. Current bioengineering strategies to regenerate the lung have not been able to replicate its extraordinary cellular diversity and complex three-dimensional arrangement, which are indispensable for life-sustaining gas exchange2,3. Here we report the successful generation of functional lungs in mice through a conditional blastocyst complementation (CBC) approach that vacates a specific niche in chimeric hosts and allows for initiation of organogenesis by donor mouse pluripotent stem cells (PSCs). We show that wild-type donor PSCs rescued lung formation in genetically defective recipient mouse embryos unable to specify (due to Ctnnb1cnull mutation) or expand (due to Fgfr2cnull mutation) early respiratory endodermal progenitors. Rescued neonates survived into adulthood and had lungs functionally indistinguishable from those of wild-type littermates. Efficient chimera formation and lung complementation required newly developed culture conditions that maintained the developmental potential of the donor PSCs and were associated with global DNA hypomethylation and increased H4 histone acetylation. These results pave the way for the development of new strategies for generating lungs in large animals to enable modeling of human lung disease as well as cell-based therapeutic interventions4-6.


Assuntos
Pneumopatias/terapia , Pulmão/crescimento & desenvolvimento , Células-Tronco Pluripotentes/metabolismo , Regeneração/genética , Acilação/genética , Animais , Blastocisto/metabolismo , Diferenciação Celular/genética , Metilação de DNA/genética , Modelos Animais de Doenças , Histonas/genética , Humanos , Pulmão/patologia , Pneumopatias/patologia , Camundongos , Organogênese/genética , Células-Tronco Pluripotentes/transplante , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , beta Catenina/genética
10.
Elife ; 82019 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-31631837

RESUMO

Notch signaling regulates cell fate selection during development in multiple organs including the lung. Previous studies on the role of Notch in the lung focused mostly on Notch pathway core components or receptor-specific functions. It is unclear, however, how Jagged or Delta-like ligands collectively or individually (Jag1, Jag2, Dll1, Dll4) influence differentiation of airway epithelial progenitors. Using mouse genetic models we show major differences in Jag and Dll in regulation and establishment of cell fate. Jag ligands had a major impact in balancing distinct cell populations in conducting airways, but had no role in the establishment of domains and cellular abundance in the neuroendocrine (NE) microenvironment. Surprisingly, Dll ligands were crucial in restricting cell fate and size of NE bodies and showed an overlapping role with Jag in differentiation of NE-associated secretory (club) cells. These mechanisms may potentially play a role in human conditions that result in aberrant NE differentiation, including NE hyperplasias and cancer.


Assuntos
Diferenciação Celular , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Pulmão/citologia , Proteínas de Membrana/metabolismo , Proteínas Serrate-Jagged/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Microambiente Celular , Ligantes , Camundongos , Sistemas Neurossecretores/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais
11.
Mol Cell Biol ; 39(18)2019 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-31262999

RESUMO

Cellular communication network factor 1 (CCN1) is a dynamically expressed, matricellular protein required for vascular development and tissue repair. The CCN1 gene is a presumed target of Yes-associated protein (YAP), a transcriptional coactivator that regulates cell growth and organ size. Herein, we demonstrate that the CCN1 promoter is indeed a direct genomic target of YAP in endothelial cells (ECs) of new blood vessel sprouts and that YAP deficiency in mice downregulates CCN1 and alters cytoskeletal and mitogenic gene expression. Interestingly, CCN1 overexpression in cultured ECs inactivates YAP in a negative feedback and causes its nuclear exclusion. Accordingly, EC-specific deletion of the CCN1 gene in mice mimics a YAP gain-of-function phenotype, characterized by EC hyperproliferation and blood vessel enlargement. CCN1 brings about its effect by providing cells with a soft compliant matrix that creates YAP-repressive cytoskeletal states. Concordantly, pharmacological inhibition of cell stiffness recapitulates the CCN1 deletion vascular phenotype. Furthermore, adeno-associated virus-mediated expression of CCN1 reversed the pathology of YAP hyperactivation and the subsequent aberrant growth of blood vessels in mice with ischemic retinopathy. Our studies unravel a new paradigm of functional interaction between CCN1 and YAP and underscore the significance of their interplay in the pathogenesis of neovascular diseases.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteína Rica em Cisteína 61/genética , Doenças Retinianas/metabolismo , Vasos Retinianos/patologia , Fatores de Transcrição/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Linhagem Celular , Proliferação de Células , Proteína Rica em Cisteína 61/metabolismo , Modelos Animais de Doenças , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Retroalimentação Fisiológica , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos , Regiões Promotoras Genéticas , Doenças Retinianas/genética , Doenças Retinianas/patologia , Vasos Retinianos/citologia , Vasos Retinianos/metabolismo , Fatores de Transcrição/genética , Proteínas de Sinalização YAP
12.
Cell Stem Cell ; 22(5): 613-614, 2018 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-29727674

RESUMO

Submucosal glands contribute to the luminal secretions of conducting airways in the respiratory tract. In this issue of Cell Stem Cell, both Tata et al. (2018) and Lynch et al. (2018) report that myoepithelial cells of submucosal glands serve as reserve stem cells to regenerate the damaged surface epithelium following severe airway injury.


Assuntos
Células Epiteliais , Traqueia , Animais , Epitélio , Camundongos , Mucosa , Regeneração , Células-Tronco
13.
Proc Natl Acad Sci U S A ; 115(5): E974-E981, 2018 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-29339516

RESUMO

Susceptibility to chronic obstructive pulmonary disease (COPD) beyond cigarette smoking is incompletely understood, although several genetic variants associated with COPD are known to regulate airway branch development. We demonstrate that in vivo central airway branch variants are present in 26.5% of the general population, are unchanged over 10 y, and exhibit strong familial aggregation. The most common airway branch variant is associated with COPD in two cohorts (n = 5,054), with greater central airway bifurcation density, and with emphysema throughout the lung. The second most common airway branch variant is associated with COPD among smokers, with narrower airway lumens in all lobes, and with genetic polymorphisms within the FGF10 gene. We conclude that central airway branch variation, readily detected by computed tomography, is a biomarker of widely altered lung structure with a genetic basis and represents a COPD susceptibility factor.


Assuntos
Brônquios/fisiopatologia , Fator 10 de Crescimento de Fibroblastos/genética , Doença Pulmonar Obstrutiva Crônica/fisiopatologia , Traqueia/fisiopatologia , Idoso , Idoso de 80 Anos ou mais , Brônquios/anatomia & histologia , Suscetibilidade a Doenças , Feminino , Genótipo , Humanos , Processamento de Imagem Assistida por Computador , Pulmão/fisiopatologia , Masculino , Pessoa de Meia-Idade , Fenótipo , Polimorfismo de Nucleotídeo Único , Estudos Prospectivos , Doença Pulmonar Obstrutiva Crônica/genética , Enfisema Pulmonar/fisiopatologia , Respiração , Fumar , Tomografia Computadorizada por Raios X , Traqueia/anatomia & histologia
14.
Nat Commun ; 8: 15857, 2017 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-28675157

RESUMO

Abnormal development of multiciliated cells is a hallmark of a variety of human conditions associated with chronic airway diseases, hydrocephalus and infertility. Multiciliogenesis requires both activation of a specialized transcriptional program and assembly of cytoplasmic structures for large-scale centriole amplification that generates basal bodies. It remains unclear, however, what mechanism initiates formation of these multiprotein complexes in epithelial progenitors. Here we show that this is triggered by nucleocytoplasmic translocation of the transcription factor E2f4. After inducing a transcriptional program of centriole biogenesis, E2f4 forms apical cytoplasmic organizing centres for assembly and nucleation of deuterosomes. Using genetically altered mice and E2F4 mutant proteins we demonstrate that centriole amplification is crucially dependent on these organizing centres and that, without cytoplasmic E2f4, deuterosomes are not assembled, halting multiciliogenesis. Thus, E2f4 integrates nuclear and previously unsuspected cytoplasmic events of centriole amplification, providing new perspectives for the understanding of normal ciliogenesis, ciliopathies and cancer.


Assuntos
Centríolos/metabolismo , Cílios/metabolismo , Citoplasma/metabolismo , Fator de Transcrição E2F4/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Autoantígenos/metabolismo , Corpos Basais/metabolismo , Proteínas de Ciclo Celular/metabolismo , Grânulos Citoplasmáticos/metabolismo , Feminino , Humanos , Masculino , Camundongos Endogâmicos C57BL , Transcrição Gênica
15.
Cell Rep ; 19(2): 246-254, 2017 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-28402849

RESUMO

There is evidence that certain club cells (CCs) in the murine airways associated with neuroepithelial bodies (NEBs) and terminal bronchioles are resistant to the xenobiotic naphthalene (Nap) and repopulate the airways after Nap injury. The identity and significance of these progenitors (variant CCs, v-CCs) have remained elusive. A recent screen for CC markers identified rare Uroplakin3a (Upk3a)-expressing cells (U-CCs) with a v-CC-like distribution. Here, we employ lineage analysis in the uninjured and chemically injured lungs to investigate the role of U-CCs as epithelial progenitors. U-CCs proliferate and generate CCs and ciliated cells in uninjured airways long-term and, like v-CCs, after Nap. U-CCs have a higher propensity to generate ciliated cells than non-U-CCs. Although U-CCs do not contribute to alveolar maintenance long-term, they generate alveolar type I and type II cells after Bleomycin (Bleo)-induced alveolar injury. Finally, we report that Upk3a+ cells exist in the NEB microenvironment of the human lung and are aberrantly expanded in conditions associated with neuroendocrine hyperplasias.


Assuntos
Bronquíolos/metabolismo , Microambiente Celular/genética , Células-Tronco/metabolismo , Uroplaquina III/biossíntese , Animais , Bleomicina/toxicidade , Bronquíolos/efeitos dos fármacos , Bronquíolos/lesões , Linhagem da Célula/efeitos dos fármacos , Microambiente Celular/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Humanos , Camundongos , Naftalenos/toxicidade , Corpos Neuroepiteliais/metabolismo , Corpos Neuroepiteliais/patologia , Alvéolos Pulmonares/lesões , Células-Tronco/efeitos dos fármacos , Uroplaquina III/genética , Cicatrização
16.
Ann Am Thorac Soc ; 13(8): S259-78, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27509163

RESUMO

The University of Vermont College of Medicine, in collaboration with the NHLBI, Alpha-1 Foundation, American Thoracic Society, Cystic Fibrosis Foundation, European Respiratory Society, International Society for Cellular Therapy, and the Pulmonary Fibrosis Foundation, convened a workshop, "Stem Cells and Cell Therapies in Lung Biology and Lung Diseases," held July 27 to 30, 2015, at the University of Vermont. The conference objectives were to review the current understanding of the role of stem and progenitor cells in lung repair after injury and to review the current status of cell therapy and ex vivo bioengineering approaches for lung diseases. These are all rapidly expanding areas of study that both provide further insight into and challenge traditional views of mechanisms of lung repair after injury and pathogenesis of several lung diseases. The goals of the conference were to summarize the current state of the field, discuss and debate current controversies, and identify future research directions and opportunities for both basic and translational research in cell-based therapies for lung diseases. This 10th anniversary conference was a follow up to five previous biennial conferences held at the University of Vermont in 2005, 2007, 2009, 2011, and 2013. Each of those conferences, also sponsored by the National Institutes of Health, American Thoracic Society, and respiratory disease foundations, has been important in helping guide research and funding priorities. The major conference recommendations are summarized at the end of the report and highlight both the significant progress and major challenges in these rapidly progressing fields.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Pneumopatias/terapia , Células-Tronco/citologia , Bioengenharia , Humanos , Sociedades Médicas , Estados Unidos
17.
Proc Natl Acad Sci U S A ; 113(29): 8242-7, 2016 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-27364009

RESUMO

Abnormal enlargement of the alveolar spaces is a hallmark of conditions such as chronic obstructive pulmonary disease and bronchopulmonary dysplasia. Notch signaling is crucial for differentiation and regeneration and repair of the airway epithelium. However, how Notch influences the alveolar compartment and integrates this process with airway development remains little understood. Here we report a prominent role of Notch signaling in the epithelial-mesenchymal interactions that lead to alveolar formation in the developing lung. We found that alveolar type II cells are major sites of Notch2 activation and show by Notch2-specific epithelial deletion (Notch2(cNull)) a unique contribution of this receptor to alveologenesis. Epithelial Notch2 was required for type II cell induction of the PDGF-A ligand and subsequent paracrine activation of PDGF receptor-α signaling in alveolar myofibroblast progenitors. Moreover, Notch2 was crucial in maintaining the integrity of the epithelial and smooth muscle layers of the distal conducting airways. Our data suggest that epithelial Notch signaling regulates multiple aspects of postnatal development in the distal lung and may represent a potential target for intervention in pulmonary diseases.


Assuntos
Pulmão/metabolismo , Receptor Notch2/metabolismo , Mucosa Respiratória/metabolismo , Animais , Linhagem Celular , Proliferação de Células , Células Epiteliais/metabolismo , Fucosiltransferases/genética , Pulmão/anatomia & histologia , Camundongos Transgênicos , Músculo Liso/anatomia & histologia , Músculo Liso/metabolismo , Receptor Notch1/genética , Receptor Notch2/genética , Mucosa Respiratória/anatomia & histologia , Transdução de Sinais
18.
Dev Cell ; 34(3): 283-96, 2015 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-26235047

RESUMO

Epithelial cells undergo dynamic polarity changes as organs pattern, but the relationship between epithelial polarity and cell fate is poorly understood. Using the developing lung as a model, we found that distinct alterations in apical-basal polarity dictate airway epithelial differentiation. We demonstrate that Crb3, a Crumbs isoform that determines epithelial apical domain identity, is required for airway differentiation by controlling the localization of the transcriptional regulator Yap. We show that Crb3 promotes the interaction between Yap and the Hippo pathway kinases Lats1/2 at apical cell junctions to induce Yap phosphorylation and cytoplasmic retention, which drive cell differentiation. Loss of Crb3 in developing mouse airways or isolated adult airway progenitors results in unrestricted nuclear Yap activity and consequent cell differentiation defects. Our findings demonstrate that polarity-dependent cues control airway cell differentiation, offering important molecular insights into organ patterning.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Polaridade Celular , Proteínas de Membrana/metabolismo , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Proteínas de Ciclo Celular , Diferenciação Celular/genética , Células Epiteliais/citologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Pulmão/embriologia , Glicoproteínas de Membrana , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Interferência de RNA , RNA Interferente Pequeno , Mucosa Respiratória/citologia , Células-Tronco/citologia , Transativadores/metabolismo , Proteínas de Sinalização YAP
19.
Development ; 142(2): 258-67, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25564622

RESUMO

Basal cells are multipotent airway progenitors that generate distinct epithelial cell phenotypes crucial for homeostasis and repair of the conducting airways. Little is known about how these progenitor cells expand and transition to differentiation to form the pseudostratified airway epithelium in the developing and adult lung. Here, we show by genetic and pharmacological approaches that endogenous activation of Notch3 signaling selectively controls the pool of undifferentiated progenitors of upper airways available for differentiation. This mechanism depends on the availability of Jag1 and Jag2, and is key to generating a population of parabasal cells that later activates Notch1 and Notch2 for secretory-multiciliated cell fate selection. Disruption of this mechanism resulted in aberrant expansion of basal cells and altered pseudostratification. Analysis of human lungs showing similar abnormalities and decreased NOTCH3 expression in subjects with chronic obstructive pulmonary disease suggests an involvement of NOTCH3-dependent events in the pathogenesis of this condition.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Diferenciação Celular/fisiologia , Células Epiteliais/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Pulmão/embriologia , Proteínas de Membrana/metabolismo , Receptores Notch/metabolismo , Mucosa Respiratória/embriologia , Transdução de Sinais/fisiologia , Animais , Western Blotting , Técnicas de Cultura de Células , Imunofluorescência , Humanos , Imuno-Histoquímica , Hibridização In Situ , Proteína Jagged-1 , Camundongos , Microscopia Confocal , Reação em Cadeia da Polimerase em Tempo Real , Receptor Notch3 , Mucosa Respiratória/citologia , Proteínas Serrate-Jagged , Especificidade da Espécie
20.
Dev Cell ; 30(2): 137-50, 2014 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-25043473

RESUMO

How epithelial progenitor cells integrate local signals to balance expansion with differentiation during organogenesis is still little understood. Here, we provide evidence that the Hippo pathway effector Yap is a key regulator of this process in the developing lung. We show that when epithelial tubules are forming and branching, a nucleocytoplasmic shift in Yap localization marks the boundary between the airway and the distal lung compartments. At this transition zone, Yap specifies a transcriptional program that controls Sox2 expression and ultimately generates the airway epithelium. Without Yap, epithelial progenitors are unable to properly respond to local TGF-ß-induced cues and control levels and distribution of Sox2 to form airways. Yap levels and subcellular localization also markedly influence Sox2 expression and differentiation of adult airway progenitors. Our data reveal a role for the Hippo-Yap pathway in integrating growth-factor-induced cues in the developing and adult lung potentially key for homeostasis and regeneration repair.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Padronização Corporal , Diferenciação Celular , Células-Tronco Embrionárias/citologia , Células Epiteliais/citologia , Pulmão/citologia , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proteínas de Ciclo Celular , Células-Tronco Embrionárias/metabolismo , Células Epiteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Via de Sinalização Hippo , Pulmão/embriologia , Pulmão/metabolismo , Camundongos , Fosfoproteínas/genética , Proteínas Serina-Treonina Quinases/genética , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Transcrição Gênica , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Proteínas de Sinalização YAP
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA