Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Development ; 148(24)2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-34927678

RESUMO

Lung organogenesis requires precise timing and coordination to effect spatial organization and function of the parenchymal cells. To provide a systematic broad-based view of the mechanisms governing the dynamic alterations in parenchymal cells over crucial periods of development, we performed a single-cell RNA-sequencing time-series yielding 102,571 epithelial, endothelial and mesenchymal cells across nine time points from embryonic day 12 to postnatal day 14 in mice. Combining computational fate-likelihood prediction with RNA in situ hybridization and immunofluorescence, we explore lineage relationships during the saccular to alveolar stage transition. The utility of this publicly searchable atlas resource (www.sucrelab.org/lungcells) is exemplified by discoveries of the complexity of type 1 pneumocyte function and characterization of mesenchymal Wnt expression patterns during the saccular and alveolar stages - wherein major expansion of the gas-exchange surface occurs. We provide an integrated view of cellular dynamics in epithelial, endothelial and mesenchymal cell populations during lung organogenesis.


Assuntos
Desenvolvimento Embrionário/genética , Pulmão/crescimento & desenvolvimento , Células-Tronco Mesenquimais/citologia , Organogênese/genética , Animais , Diferenciação Celular/genética , Linhagem da Célula/genética , Embrião de Mamíferos/ultraestrutura , Células Epiteliais/citologia , Células Epiteliais/ultraestrutura , Regulação da Expressão Gênica no Desenvolvimento/genética , Pulmão/ultraestrutura , Células-Tronco Mesenquimais/ultraestrutura , Camundongos , RNA-Seq , Análise de Célula Única , Transcriptoma/genética
2.
Proc Natl Acad Sci U S A ; 118(20)2021 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-33990468

RESUMO

Lamellar bodies (LBs) are lysosome-related organelles (LROs) of surfactant-producing alveolar type 2 (AT2) cells of the distal lung epithelium. Trafficking pathways to LBs have been understudied but are likely critical to AT2 cell homeostasis given associations between genetic defects of endosome to LRO trafficking and pulmonary fibrosis in Hermansky Pudlak syndrome (HPS). Our prior studies uncovered a role for AP-3, defective in HPS type 2, in trafficking Peroxiredoxin-6 to LBs. We now show that the P4-type ATPase ATP8A1 is sorted by AP-3 from early endosomes to LBs through recognition of a C-terminal dileucine-based signal. Disruption of the AP-3/ATP8A1 interaction causes ATP8A1 accumulation in early sorting and/or recycling endosomes, enhancing phosphatidylserine exposure on the cytosolic leaflet. This in turn promotes activation of Yes-activating protein, a transcriptional coactivator, augmenting cell migration and AT2 cell numbers. Together, these studies illuminate a mechanism whereby loss of AP-3-mediated trafficking contributes to a toxic gain-of-function that results in enhanced and sustained activation of a repair pathway associated with pulmonary fibrosis.


Assuntos
Complexo 3 de Proteínas Adaptadoras/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Adenosina Trifosfatases/genética , Células Epiteliais Alveolares/metabolismo , Síndrome de Hermanski-Pudlak/genética , Proteínas de Transferência de Fosfolipídeos/genética , Fibrose Pulmonar/genética , Fatores de Transcrição/genética , Complexo 3 de Proteínas Adaptadoras/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adenosina Trifosfatases/metabolismo , Células Epiteliais Alveolares/citologia , Animais , Transporte Biológico , Linhagem Celular , Movimento Celular , Modelos Animais de Doenças , Endossomos/metabolismo , Feminino , Regulação da Expressão Gênica , Síndrome de Hermanski-Pudlak/metabolismo , Síndrome de Hermanski-Pudlak/patologia , Humanos , Pulmão/metabolismo , Pulmão/patologia , Lisossomos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Peroxirredoxina VI/genética , Peroxirredoxina VI/metabolismo , Fosfatidilserinas/metabolismo , Proteínas de Transferência de Fosfolipídeos/metabolismo , Cultura Primária de Células , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , Transdução de Sinais , Fatores de Transcrição/metabolismo , Proteínas de Sinalização YAP , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo
3.
Immunity ; 36(5): 782-94, 2012 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-22560444

RESUMO

Effective major histocompatibility complex-II (MHC-II) antigen presentation from phagocytosed particles requires phagosome-intrinsic Toll-like receptor (TLR) signaling, but the molecular mechanisms underlying TLR delivery to phagosomes and how signaling regulates antigen presentation are incompletely understood. We show a requirement in dendritic cells (DCs) for adaptor protein-3 (AP-3) in efficient TLR recruitment to phagosomes and MHC-II presentation of antigens internalized by phagocytosis but not receptor-mediated endocytosis. DCs from AP-3-deficient pearl mice elicited impaired CD4(+) T cell activation and Th1 effector cell function to particulate antigen in vitro and to recombinant Listeria monocytogenes infection in vivo. Whereas phagolysosome maturation and peptide:MHC-II complex assembly proceeded normally in pearl DCs, peptide:MHC-II export to the cell surface was impeded. This correlated with reduced TLR4 recruitment and proinflammatory signaling from phagosomes by particulate TLR ligands. We propose that AP-3-dependent TLR delivery from endosomes to phagosomes and subsequent signaling mobilize peptide:MHC-II export from intracellular stores.


Assuntos
Complexo 3 de Proteínas Adaptadoras/imunologia , Apresentação de Antígeno/imunologia , Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Fagossomos/imunologia , Receptores Toll-Like/imunologia , Complexo 3 de Proteínas Adaptadoras/metabolismo , Animais , Antígenos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Diferenciação Celular/imunologia , Membrana Celular/imunologia , Membrana Celular/metabolismo , Células Cultivadas , Células Dendríticas/metabolismo , Endocitose/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Ligantes , Listeria monocytogenes/imunologia , Listeriose/imunologia , Listeriose/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fator 88 de Diferenciação Mieloide/imunologia , Fator 88 de Diferenciação Mieloide/metabolismo , Ovalbumina/imunologia , Ovalbumina/metabolismo , Peptídeos/imunologia , Peptídeos/metabolismo , Fagocitose/imunologia , Fagossomos/metabolismo , Transdução de Sinais/imunologia , Células Th1/imunologia , Células Th1/metabolismo , Receptores Toll-Like/metabolismo
4.
Am J Respir Crit Care Med ; 201(10): 1249-1262, 2020 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-32023086

RESUMO

Rationale: Bronchopulmonary dysplasia (BPD) is a leading complication of preterm birth that affects infants born in the saccular stage of lung development at <32 weeks of gestation. Although the mechanisms driving BPD remain uncertain, exposure to hyperoxia is thought to contribute to disease pathogenesis.Objectives: To determine the effects of hyperoxia on epithelial-mesenchymal interactions and to define the mediators of activated Wnt/ß-catenin signaling after hyperoxia injury.Methods: Three hyperoxia models were used: A three-dimensional organotypic coculture using primary human lung cells, precision-cut lung slices (PCLS), and a murine in vivo hyperoxia model. Comparisons of normoxia- and hyperoxia-exposed samples were made by real-time quantitative PCR, RNA in situ hybridization, quantitative confocal microscopy, and lung morphometry.Measurements and Main Results: Examination of an array of Wnt ligands in the three-dimensional organotypic coculture revealed increased mesenchymal expression of WNT5A. Inhibition of Wnt5A abrogated the BPD transcriptomic phenotype induced by hyperoxia. In the PCLS model, Wnt5A inhibition improved alveolarization following hyperoxia exposure, and treatment with recombinant Wnt5a reproduced features of the BPD phenotype in PCLS cultured in normoxic conditions. Chemical inhibition of NF-κB with BAY11-7082 reduced Wnt5a expression in the PCLS hyperoxia model and in vivo mouse hyperoxia model, with improved alveolarization in the PCLS model.Conclusions: Increased mesenchymal Wnt5A during saccular-stage hyperoxia injury contributes to the impaired alveolarization and septal thickening observed in BPD. Precise targeting of Wnt5A may represent a potential therapeutic strategy for the treatment of BPD.


Assuntos
Células Epiteliais Alveolares/metabolismo , Fibroblastos/metabolismo , Hiperóxia/genética , Pulmão/metabolismo , Células-Tronco Mesenquimais/metabolismo , Proteína Wnt-5a/genética , Animais , Displasia Broncopulmonar , Técnicas de Cocultura , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Hiperóxia/metabolismo , Hibridização In Situ , Pulmão/crescimento & desenvolvimento , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Microscopia Confocal , NF-kappa B/antagonistas & inibidores , Nitrilas/farmacologia , Técnicas de Cultura de Órgãos , Reação em Cadeia da Polimerase em Tempo Real , Sulfonas/farmacologia , Proteína Wnt-5a/efeitos dos fármacos , Proteína Wnt-5a/metabolismo
5.
Am J Pathol ; 188(4): 853-862, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29355514

RESUMO

Wnt/ß-catenin signaling is necessary for normal lung development, and abnormal Wnt signaling contributes to the pathogenesis of both bronchopulmonary dysplasia (BPD) and idiopathic pulmonary fibrosis (IPF), fibrotic lung diseases that occur during infancy and aging, respectively. Using a library of human normal and diseased human lung samples, we identified a distinct signature of nuclear accumulation of ß-catenin phosphorylated at tyrosine 489 and epithelial cell cytosolic localization of ß-catenin phosphorylated at tyrosine 654 in early normal lung development and fibrotic lung diseases BPD and IPF. Furthermore, this signature was recapitulated in murine models of BPD and IPF. Image analysis of immunofluorescence colocalization demonstrated a consistent pattern of elevated nuclear phosphorylated ß-catenin in the lung epithelium and surrounding mesenchyme in BPD and IPF, closely resembling the pattern observed in 18-week fetal lung. Nuclear ß-catenin phosphorylated at tyrosine 489 associated with an increased expression of Wnt target gene AXIN2, suggesting that the observed ß-catenin signature is of functional significance during normal development and injury repair. The association of specific modifications of ß-catenin during normal lung development and again in response to lung injury supports the widely held concept that repair of lung injury involves the recapitulation of developmental programs. Furthermore, these observations suggest that ß-catenin phosphorylation has potential as a therapeutic target for the treatment and prevention of both BPD and IPF.


Assuntos
Displasia Broncopulmonar/metabolismo , Fibrose Pulmonar Idiopática/metabolismo , beta Catenina/metabolismo , Células A549 , Adulto , Animais , Animais Recém-Nascidos , Proteína Axina/metabolismo , Displasia Broncopulmonar/patologia , Núcleo Celular/metabolismo , Células Epiteliais/metabolismo , Feminino , Feto/metabolismo , Humanos , Fibrose Pulmonar Idiopática/patologia , Pulmão/metabolismo , Pulmão/patologia , Camundongos Endogâmicos C57BL , Fosforilação , Gravidez , Segundo Trimestre da Gravidez , Processamento de Proteína Pós-Traducional , Transdução de Sinais , Tirosina/metabolismo
6.
Am J Respir Cell Mol Biol ; 58(5): 566-574, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29190429

RESUMO

Defining the mechanisms of cellular pathogenesis in rare lung diseases such as Hermansky-Pudlak syndrome (HPS) is often complicated by loss of the differentiated phenotype of cultured primary alveolar type 2 (AT2) cells, as well as by a lack of durable cell lines that are faithful to both AT2-cell and rare disease phenotypes. We used CRISPR/Cas9 gene editing to generate a series of HPS-specific mutations in the MLE-15 cell line. The resulting MLE-15/HPS cell lines exhibit preservation of AT2 cellular functions, including formation of lamellar body-like organelles, complete processing of surfactant protein B, and known features of HPS specific to each trafficking complex, including loss of protein targeting to lamellar bodies. MLE-15/HPS1 and MLE-15/HPS2 (with a mutation in Ap3ß1) express increased macrophage chemotactic protein-1, a well-described mediator of alveolitis in patients with HPS and in mouse models. We show that MLE-15/HPS9 and pallid AT2 cells (with a mutation in Bloc1s6) also express increased macrophage chemotactic protein-1, suggesting that mice and humans with BLOC-1 mutations may also be susceptible to alveolitis. In addition to providing a flexible platform to examine the role of HPS-specific mutations in trafficking AT2 cells, MLE-15/HPS cell lines provide a durable resource for high-throughput screening and studies of cellular pathophysiology that are likely to accelerate progress toward developing novel therapies for this rare lung disease.


Assuntos
Células Epiteliais Alveolares/metabolismo , Proteína 9 Associada à CRISPR/genética , Sistemas CRISPR-Cas , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Edição de Genes/métodos , Síndrome de Hermanski-Pudlak/genética , Mutação , Células Epiteliais Alveolares/patologia , Animais , Proteína 9 Associada à CRISPR/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Marcadores Genéticos , Predisposição Genética para Doença , Síndrome de Hermanski-Pudlak/metabolismo , Síndrome de Hermanski-Pudlak/patologia , Humanos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo
7.
Am J Respir Cell Mol Biol ; 59(2): 158-166, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29625013

RESUMO

Alveolar type II (AT2) epithelial cells are uniquely specialized to produce surfactant in the lung and act as progenitor cells in the process of repair after lung injury. AT2 cell injury has been implicated in several lung diseases, including idiopathic pulmonary fibrosis and bronchopulmonary dysplasia. The inability to maintain primary AT2 cells in culture has been a significant barrier in the investigation of pulmonary biology. We have addressed this knowledge gap by developing a three-dimensional (3D) organotypic coculture using primary human fetal AT2 cells and pulmonary fibroblasts. Grown on top of matrix-embedded fibroblasts, the primary human AT2 cells establish a monolayer and have direct contact with the underlying pulmonary fibroblasts. Unlike conventional two-dimensional (2D) culture, the structural and functional phenotype of the AT2 cells in our 3D organotypic culture was preserved over 7 days of culture, as evidenced by the presence of lamellar bodies and by production of surfactant proteins B and C. Importantly, the AT2 cells in 3D cocultures maintained the ability to replicate, with approximately 60% of AT2 cells staining positive for the proliferation marker Ki67, whereas no such proliferation is evident in 2D cultures of the same primary AT2 cells. This organotypic culture system enables interrogation of AT2 epithelial biology by providing a reductionist in vitro model in which to investigate the response of AT2 epithelial cells and AT2 cell-fibroblast interactions during lung injury and repair.


Assuntos
Comunicação Celular/fisiologia , Células Epiteliais/metabolismo , Lesão Pulmonar/patologia , Pulmão/patologia , Células Cultivadas , Técnicas de Cocultura , Fibroblastos/metabolismo , Humanos , Fenótipo
8.
J Biol Chem ; 291(16): 8414-27, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26907692

RESUMO

The Hermansky Pudlak syndromes (HPS) constitute a family of disorders characterized by oculocutaneous albinism and bleeding diathesis, often associated with lethal lung fibrosis. HPS results from mutations in genes of membrane trafficking complexes that facilitate delivery of cargo to lysosome-related organelles. Among the affected lysosome-related organelles are lamellar bodies (LB) within alveolar type 2 cells (AT2) in which surfactant components are assembled, modified, and stored. AT2 from HPS patients and mouse models of HPS exhibit enlarged LB with increased phospholipid content, but the mechanism underlying these defects is unknown. We now show that AT2 in the pearl mouse model of HPS type 2 lacking the adaptor protein 3 complex (AP-3) fails to accumulate the soluble enzyme peroxiredoxin 6 (PRDX6) in LB. This defect reflects impaired AP-3-dependent trafficking of PRDX6 to LB, because pearl mouse AT2 cells harbor a normal total PRDX6 content. AP-3-dependent targeting of PRDX6 to LB requires the transmembrane protein LIMP-2/SCARB2, a known AP-3-dependent cargo protein that functions as a carrier for lysosomal proteins in other cell types. Depletion of LB PRDX6 in AP-3- or LIMP-2/SCARB2-deficient mice correlates with phospholipid accumulation in lamellar bodies and with defective intraluminal degradation of LB disaturated phosphatidylcholine. Furthermore, AP-3-dependent LB targeting is facilitated by protein/protein interaction between LIMP-2/SCARB2 and PRDX6 in vitro and in vivo Our data provide the first evidence for an AP-3-dependent cargo protein required for the maturation of LB in AT2 and suggest that the loss of PRDX6 activity contributes to the pathogenic changes in LB phospholipid homeostasis found HPS2 patients.


Assuntos
Complexo 3 de Proteínas Adaptadoras/metabolismo , Antígenos CD36/metabolismo , Síndrome de Hermanski-Pudlak/metabolismo , Proteínas de Membrana Lisossomal/metabolismo , Peroxirredoxina VI/metabolismo , Fosfatidilcolinas/metabolismo , Alvéolos Pulmonares/metabolismo , Complexo 3 de Proteínas Adaptadoras/genética , Animais , Antígenos CD36/genética , Feminino , Síndrome de Hermanski-Pudlak/genética , Síndrome de Hermanski-Pudlak/patologia , Proteínas de Membrana Lisossomal/genética , Masculino , Camundongos , Peroxirredoxina VI/genética , Fosfatidilcolinas/genética , Alvéolos Pulmonares/patologia
9.
Am J Physiol Lung Cell Mol Physiol ; 312(2): L186-L195, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-27941077

RESUMO

Bronchopulmonary dysplasia (BPD) is a common complication of premature birth. The histopathology of BPD is characterized by an arrest of alveolarization with fibroblast activation. The Wnt/ß-catenin signaling pathway is important in early lung development. When Wnt signaling is active, phosphorylation of ß-catenin by tyrosine kinases at activating sites, specifically at tyrosine 489 (Y489), correlates with nuclear localization of ß-catenin. We examined fetal lung tissue, lung tissue from term newborns, and lung tissue from infants who died with BPD; we found nuclear ß-catenin phosphorylation at Y489 in epithelial and mesenchymal cells in fetal tissue and BPD tissue, but not in the lungs of term infants. Using a 3D human organoid model, we found increased nuclear localization of ß-catenin phosphorylated at Y489 (p-ß-cateninY489) after exposure to alternating hypoxia and hyperoxia compared with organoids cultured in normoxia. Exogenous stimulation of the canonical Wnt pathway in organoids was sufficient to cause nuclear localization of p-ß-cateninY489 in normoxia and mimicked the pattern of α-smooth muscle actin (α-SMA) expression seen with fibroblastic activation from oxidative stress. Treatment of organoids with a tyrosine kinase inhibitor prior to cyclic hypoxia-hyperoxia inhibited nuclear localization of p-ß-cateninY489 and prevented α-SMA expression by fibroblasts. Posttranslational phosphorylation of ß-catenin is a transient feature of normal lung development. Moreover, the persistence of p-ß-cateninY489 is a durable marker of fibroblast activation in BPD and may play an important role in BPD disease pathobiology.


Assuntos
Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/patologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Processamento de Proteína Pós-Traducional , beta Catenina/metabolismo , Actinas/metabolismo , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Dasatinibe/farmacologia , Fibroblastos/efeitos dos fármacos , Humanos , Hiperóxia/complicações , Hiperóxia/metabolismo , Hiperóxia/patologia , Hipóxia/complicações , Hipóxia/metabolismo , Hipóxia/patologia , Recém-Nascido , Pulmão/efeitos dos fármacos , Pulmão/crescimento & desenvolvimento , Pulmão/metabolismo , Pulmão/patologia , Organoides/efeitos dos fármacos , Organoides/metabolismo , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Via de Sinalização Wnt/efeitos dos fármacos
10.
Am J Physiol Lung Cell Mol Physiol ; 308(1): L33-47, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25344067

RESUMO

Mutation of threonine for isoleucine at codon 73 (I73T) in the human surfactant protein C (hSP-C) gene (SFTPC) accounts for a significant portion of SFTPC mutations associated with interstitial lung disease (ILD). Cell lines stably expressing tagged primary translation product of SP-C isoforms were generated to test the hypothesis that deposition of hSP-C(I73T) within the endosomal system promotes disruption of a key cellular quality control pathway, macroautophagy. By fluorescence microscopy, wild-type hSP-C (hSP-C(WT)) colocalized with exogenously expressed human ATP binding cassette class A3 (hABCA3), an indicator of normal trafficking to lysosomal-related organelles. In contrast, hSP-C(I73T) was dissociated from hABCA3 but colocalized to the plasma membrane as well as the endosomal network. Cells expressing hSP-C(I73T) exhibited increases in size and number of cytosolic green fluorescent protein/microtubule-associated protein 1 light-chain 3 (LC3) vesicles, some of which colabeled with red fluorescent protein from the gene dsRed/hSP-C(I73T). By transmission electron microscopy, hSP-C(I73T) cells contained abnormally large autophagic vacuoles containing organellar and proteinaceous debris, which phenocopied ultrastructural changes in alveolar type 2 cells in a lung biopsy from a SFTPC I73T patient. Biochemically, hSP-C(I73T) cells exhibited increased expression of Atg8/LC3, SQSTM1/p62, and Rab7, consistent with a distal block in autophagic vacuole maturation, confirmed by flux studies using bafilomycin A1 and rapamycin. Functionally, hSP-C(I73T) cells showed an impaired degradative capacity for an aggregation-prone huntingtin-1 reporter substrate. The disruption of autophagy-dependent proteostasis was accompanied by increases in mitochondria biomass and parkin expression coupled with a decrease in mitochondrial membrane potential. We conclude that hSP-C(I73T) induces an acquired block in macroautophagy-dependent proteostasis and mitophagy, which could contribute to the increased vulnerability of the lung epithelia to second-hit injury as seen in ILD.


Assuntos
Autofagia , Doenças Genéticas Inatas/metabolismo , Doenças Pulmonares Intersticiais/metabolismo , Mutação de Sentido Incorreto , Proteína C Associada a Surfactante Pulmonar/metabolismo , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Proteínas Adaptadoras de Transdução de Sinal/genética , Substituição de Aminoácidos , Família da Proteína 8 Relacionada à Autofagia , Feminino , Regulação da Expressão Gênica/genética , Doenças Genéticas Inatas/genética , Doenças Genéticas Inatas/patologia , Células HEK293 , Humanos , Lactente , Doenças Pulmonares Intersticiais/genética , Doenças Pulmonares Intersticiais/patologia , Lisossomos/genética , Lisossomos/metabolismo , Lisossomos/ultraestrutura , Potencial da Membrana Mitocondrial/genética , Proteínas dos Microfilamentos/biossíntese , Proteínas dos Microfilamentos/genética , Proteínas Associadas aos Microtúbulos/biossíntese , Proteínas Associadas aos Microtúbulos/genética , Mitocôndrias/genética , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Deficiências na Proteostase/genética , Deficiências na Proteostase/metabolismo , Deficiências na Proteostase/patologia , Proteína C Associada a Surfactante Pulmonar/genética , Proteína Sequestossoma-1 , Ubiquitina-Proteína Ligases/biossíntese , Ubiquitina-Proteína Ligases/genética , Vacúolos/genética , Vacúolos/metabolismo , Vacúolos/ultraestrutura , Proteínas rab de Ligação ao GTP/biossíntese , Proteínas rab de Ligação ao GTP/genética , proteínas de unión al GTP Rab7
11.
Traffic ; 12(9): 1196-210, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21707890

RESUMO

Interstitial lung disease in both children and adults has been linked to mutations in the lung-specific surfactant protein C (SFTPC) gene. Among these, the missense mutation [isoleucine to threonine at codon 73 = human surfactant protein C (hSP-C(I73T) )] accounts for ∼30% of all described SFTPC mutations. We reported previously that unlike the BRICHOS misfolding SFTPC mutants, expression of hSP-C(I73T) induces lung remodeling and alveolar lipoproteinosis without a substantial Endoplasmic Reticulum (ER) stress response or ER-mediated intrinsic apoptosis. We show here that, in contrast to its wild-type counterpart that is directly routed to lysosomal-like organelles for processing, SP-C(I73T) is misdirected to the plasma membrane and subsequently internalized to the endocytic pathway via early endosomes, leading to the accumulation of abnormally processed proSP-C isoforms. Functionally, cells expressing hSP-C(I73T) demonstrated both impaired uptake and degradation of surfactant phospholipid, thus providing a molecular mechanism for the observed lipid accumulation in patients expressing hSP-C(I73T) through the disruption of normal phospholipid recycling. Our data provide evidence for a novel cellular mechanism for conformational protein-associated diseases and suggest a paradigm for mistargeted proteins involved in the disruption of the endosomal/lysosomal sorting machinery.


Assuntos
Endossomos/metabolismo , Doenças Pulmonares Intersticiais/genética , Mutação , Fosfolipídeos/metabolismo , Proteína C Associada a Surfactante Pulmonar/genética , Proteína C Associada a Surfactante Pulmonar/metabolismo , Adulto , Membrana Celular/metabolismo , Células Cultivadas , Criança , Endocitose/fisiologia , Retículo Endoplasmático/metabolismo , Endossomos/química , Humanos , Doenças Pulmonares Intersticiais/patologia , Doenças Pulmonares Intersticiais/fisiopatologia , Lisossomos/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Transporte Proteico/fisiologia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transferrina/metabolismo , Vesículas Transportadoras/metabolismo
12.
Am J Physiol Lung Cell Mol Physiol ; 305(12): L970-80, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24142515

RESUMO

The lipid transport protein, ABCA3, expressed in alveolar type 2 (AT2) cells, is critical for surfactant homeostasis. The first luminal loop of ABCA3 contains three putative N-linked glycosylation sites at residues 53, 124, and 140. A common cotranslational modification, N-linked glycosylation, is critical for the proper expression of glycoproteins by enhancing folding, trafficking, and stability through augmentation of the endoplasmic reticulum (ER) folding cycle. To understand its role in ABCA3 biosynthesis, we utilized EGFP-tagged fusion constructs with either wild-type or mutant ABCA3 cDNAs that contained glutamine for asparagine substitutions at the putative glycosylation motifs. In A549 cells, inhibition of glycosylation by tunicamycin increased the electrophoretic mobility (Mr) and reduced the expression level of wild-type ABCA3 in a dose-dependent manner. Fluorescence imaging of transiently transfected A549 or primary human AT2 cells showed that although single motif mutants exhibited a vesicular distribution pattern similar to wild-type ABCA3, mutation of N124 and N140 residues resulted in a shift toward an ER-predominant distribution. By immunoblotting, the N53 mutation exhibited no effect on either the Mr or ABCA3 expression level. In contrast, substitutions at N124 or N140, as well a N124/N140 double mutation, resulted in increased electrophoretic mobility indicative of a glycosylation deficiency accompanied by reduced overall expression levels. Diminished steady-state levels of glycan-deficient ABCA3 isoforms were rescued by treatment with the proteasome inhibitor MG132. These results suggest that cotranslational N-linked glycosylation at N124 and N140 is critical for ABCA3 stability, and its disruption results in protein destabilization and proteasomal degradation.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Retículo Endoplasmático/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Transportadores de Cassetes de Ligação de ATP/genética , Células Cultivadas , Retículo Endoplasmático/genética , Glicosilação/efeitos dos fármacos , Humanos , Mutação/genética , Complexo de Endopeptidases do Proteassoma/genética , Transporte Proteico/genética , Tunicamicina/farmacologia
13.
Pediatr Res ; 74(6): 646-51, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24002332

RESUMO

BACKGROUND: Surfactant protein B (SP-B) is essential for normal lung function, and decreased concentrations of SP-B have a deleterious effect on pulmonary outcome. SP-B levels may correlate with variations in the encoding gene (SFTPB). SFTPB single-nucleotide polymorphism Ile131Thr affects proSP-B N-glycosylation in humans and the glycosylated Thr variant associates with pulmonary diseases. METHODS: We analyzed SP-B levels in amniotic fluid samples for associations with SFTPB polymorphisms and generated cell lines expressing either proSP-B/131Ile or proSP-B/131Thr for examining the effect of glycosylation on proSP-B secretion kinetics. To determine any transcription preference between Ile131Thr allelic variants, we used heterozygous human lungs for allelic expression imbalance assays. RESULTS: Protein levels correlated with Ile131Thr genotype and the lowest SP-B levels were observed in Thr/Thr homozygotes. Our results suggest that Ile131Thr variation-dependent N-glycosylation associates with decreased levels of SP-B, which is secreted from fetal lung to amniotic fluid. Glycosylated proSP-B/131Thr was secreted from transfected cells at a lower rate than nonglycosylated proSP-B/131Ile. Expression levels of the mRNA variants were equal. Secretion of the glycosylated variant was thus delayed in vitro by a posttranscriptional mechanism. CONCLUSION: These data support the hypothesis that proSP-B glycosylation due to Ile131Thr variation may have a causal role in genetic susceptibility to acute respiratory distress.


Assuntos
Alelos , Proteína B Associada a Surfactante Pulmonar/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , Glicosilação , Humanos , Isoleucina/metabolismo , Polimorfismo de Nucleotídeo Único , Proteína B Associada a Surfactante Pulmonar/genética , Treonina/metabolismo
14.
J Immunol ; 186(5): 3197-205, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21282514

RESUMO

CXCL5, a member of the CXC family of chemokines, contributes to neutrophil recruitment during lung inflammation, but its regulation is poorly understood. Because the T cell-derived cytokine IL-17A enhances host defense by triggering production of chemokines, particularly in combination with TNF-α, we hypothesized that IL-17A would enhance TNF-α-induced expression of CXCL5. Intratracheal coadministration of IL-17A and TNF-α in mice induced production of CXCL1, CXCL2, and CXCL5, which was associated with increased neutrophil influx in the lung at 8 and 24 h. The synergistic effects of TNF-α and IL17A were greatly attenuated in Cxcl5(-/-) mice at 24 h, but not 8 h, after exposure, a time when CXCL5 expression was at its peak in wild-type mice. Bone marrow chimeras produced using Cxcl5(-/-) donors and recipients demonstrated that lung-resident cells were the source of CXCL5. Using differentiated alveolar epithelial type II (ATII) cells derived from human fetal lung, we found that IL-17A enhanced TNF-α-induced CXCL5 transcription and stabilized TNF-α-induced CXCL5 transcripts. Whereas expression of CXCL5 required activation of NF-κB, IL-17A did not increase TNF-α-induced NF-κB activation. Apical costimulation of IL-17A and TNF-α provoked apical secretion of CXCL5 by human ATII cells in a transwell system, whereas basolateral costimulation led to both apical and basolateral secretion of CXCL5. The observation that human ATII cells secrete CXCL5 in a polarized fashion may represent a mechanism to recruit neutrophils in host defense in a fashion that discriminates the site of initial injury.


Assuntos
Quimiocina CXCL5/biossíntese , Interleucina-17/fisiologia , Alvéolos Pulmonares/imunologia , Alvéolos Pulmonares/metabolismo , Fator de Necrose Tumoral alfa/fisiologia , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/patologia , Animais , Inibição de Migração Celular/genética , Inibição de Migração Celular/imunologia , Células Cultivadas , Quimiocina CXCL1/biossíntese , Quimiocina CXCL2/biossíntese , Quimiocina CXCL5/deficiência , Quimiocina CXCL5/metabolismo , Quimiotaxia de Leucócito/genética , Quimiotaxia de Leucócito/imunologia , Modelos Animais de Doenças , Quimioterapia Combinada , Humanos , Mediadores da Inflamação/metabolismo , Mediadores da Inflamação/fisiologia , Interleucina-17/administração & dosagem , Interleucina-17/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/imunologia , Neutrófilos/patologia , Pneumonia Bacteriana/imunologia , Pneumonia Bacteriana/metabolismo , Pneumonia Bacteriana/patologia , Alvéolos Pulmonares/patologia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/biossíntese , Índice de Gravidade de Doença , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Fator de Necrose Tumoral alfa/administração & dosagem , Fator de Necrose Tumoral alfa/biossíntese
15.
JCI Insight ; 8(14)2023 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-37279065

RESUMO

During alveolar repair, alveolar type 2 (AT2) epithelial cell progenitors rapidly proliferate and differentiate into flat AT1 epithelial cells. Failure of normal alveolar repair mechanisms can lead to loss of alveolar structure (emphysema) or development of fibrosis, depending on the type and severity of injury. To test if ß1-containing integrins are required during repair following acute injury, we administered E. coli lipopolysaccharide (LPS) by intratracheal injection to mice with a postdevelopmental deletion of ß1 integrin in AT2 cells. While control mice recovered from LPS injury without structural abnormalities, ß1-deficient mice had more severe inflammation and developed emphysema. In addition, recovering alveoli were repopulated with an abundance of rounded epithelial cells coexpressing AT2 epithelial, AT1 epithelial, and mixed intermediate cell state markers, with few mature type 1 cells. AT2 cells deficient in ß1 showed persistently increased proliferation after injury, which was blocked by inhibiting NF-κB activation in these cells. Lineage tracing experiments revealed that ß1-deficient AT2 cells failed to differentiate into mature AT1 epithelial cells. Together, these findings demonstrate that functional alveolar repair after injury with terminal alveolar epithelial differentiation requires ß1-containing integrins.


Assuntos
Enfisema , Lipopolissacarídeos , Camundongos , Animais , Lipopolissacarídeos/toxicidade , Escherichia coli , Pulmão , Integrinas
16.
J Biol Chem ; 286(24): 21239-53, 2011 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-21525008

RESUMO

Sodium 4-phenylbutyrate (4PBA) improves the intracellular trafficking of ΔF508-CFTR in cystic fibrosis (CF) epithelial cells. The underlying mechanism is uncertain, but 4PBA modulates the expression of some cytosolic molecular chaperones. To identify other 4PBA-regulated proteins that might regulate ΔF508-CFTR trafficking, we performed a differential display RT-PCR screen on IB3-1 CF bronchiolar epithelial cells exposed to 4PBA. One transcript up-regulated by 4PBA encoded ERp29, a luminal resident of the endoplasmic reticulum (ER) thought to be a novel molecular chaperone. We tested the hypothesis that ERp29 is a 4PBA-regulated ER chaperone that influences ΔF508-CFTR trafficking. ERp29 mRNA and protein expression was significantly increased (∼1.5-fold) in 4PBA-treated IB3-1 cells. In Xenopus oocytes, ERp29 overexpression increased the functional expression of both wild-type and ΔF508-CFTR over 3-fold and increased wild-type cystic fibrosis transmembrane conductance regulator (CFTR) plasma membrane expression. In CFBE41o- WT-CFTR cells, expression of and short circuit currents mediated by CFTR decreased upon depletion of ERp29 as did maturation of newly synthesized CFTR. In IB3-1 cells, ΔF508-CFTR co-immunoprecipitated with endogenous ERp29, and overexpression of ERp29 led to increased ΔF508-CFTR expression at the plasma membrane. These data suggest that ERp29 is a 4PBA-regulated ER chaperone that regulates WT-CFTR biogenesis and can promote ΔF508-CFTR trafficking in CF epithelial cells.


Assuntos
Membrana Celular/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Fibrose Cística/metabolismo , Retículo Endoplasmático/metabolismo , Células Epiteliais/metabolismo , Proteínas de Choque Térmico/metabolismo , Animais , Biotinilação , Eletrofisiologia/métodos , Humanos , Íons/química , Oócitos/metabolismo , Fenilbutiratos/farmacologia , Transporte Proteico , Xenopus
17.
Am J Respir Crit Care Med ; 184(4): 449-58, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21616998

RESUMO

RATIONALE: The pulmonary phenotype of Hermansky-Pudlak syndrome (HPS) in adults includes foamy alveolar type 2 cells, inflammation, and lung remodeling, but there is no information about ontogeny or early disease mediators. OBJECTIVES: To establish the ontogeny of HPS lung disease in an animal model, examine disease mediators, and relate them to patients with HPS1. METHODS: Mice with mutations in both HPS1/pale ear and HPS2/AP3B1/pearl (EPPE mice) were studied longitudinally. Total lung homogenate, lung tissue sections, and bronchoalveolar lavage (BAL) were examined for phospholipid, collagen, histology, cell counts, chemokines, surfactant protein D (SP-D), and S-nitrosylated SP-D. Isolated alveolar epithelial cells were examined for expression of inflammatory mediators, and chemotaxis assays were used to assess their importance. Pulmonary function test results and BAL from patients with HPS1 and normal volunteers were examined for clinical correlation. MEASUREMENTS AND MAIN RESULTS: EPPE mice develop increased total lung phospholipid, followed by a macrophage-predominant pulmonary inflammation, and lung remodeling including fibrosis. BAL fluid from EPPE animals exhibited early accumulation of both SP-D and S-nitrosylated SP-D. BAL fluid from patients with HPS1 exhibited similar changes in SP-D that correlated inversely with pulmonary function. Alveolar epithelial cells demonstrated expression of both monocyte chemotactic protein (MCP)-1 and inducible nitric oxide synthase in juvenile EPPE mice. Last, BAL from EPPE mice and patients with HPS1 enhanced migration of RAW267.4 cells, which was attenuated by immunodepletion of SP-D and MCP-1. CONCLUSIONS: Inflammation is initiated from the abnormal alveolar epithelial cells in HPS, and S-nitrosylated SP-D plays a significant role in amplifying pulmonary inflammation.


Assuntos
Modelos Animais de Doenças , Síndrome de Hermanski-Pudlak , Camundongos , Pneumonia/etiologia , Alvéolos Pulmonares/fisiopatologia , Proteína D Associada a Surfactante Pulmonar/metabolismo , Mucosa Respiratória/fisiopatologia , Envelhecimento/metabolismo , Animais , Movimento Celular , Quimiocina CCL2/metabolismo , Fatores Quimiotáticos/metabolismo , Citocinas/metabolismo , Fibrose , Síndrome de Hermanski-Pudlak/fisiopatologia , Humanos , Pulmão/metabolismo , Macrófagos/patologia , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Compostos Nitrosos/metabolismo , Fosfolipídeos/metabolismo , Alvéolos Pulmonares/patologia , Índice de Gravidade de Doença , Fatores de Tempo
18.
Nat Med ; 8(5): 514-7, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11984597

RESUMO

The only proven requirement for ascorbic acid (vitamin C) is in preventing scurvy, presumably because it is a cofactor for hydroxylases required for post-translational modifications that stabilize collagen. We have created mice deficient in the mouse ortholog (solute carrier family 23 member 1 or Slc23a1) of a rat ascorbic-acid transporter, Svct2 (ref. 4). Cultured embryonic fibroblasts from homozygous Slc23a1(-/-) mice had less than 5% of normal ascorbic-acid uptake. Ascorbic-acid levels were undetectable or markedly reduced in the blood and tissues of Slc23a1(-/-) mice. Prenatal supplementation of pregnant females did not elevate blood ascorbic acid in Slc23a1(-/-) fetuses, suggesting Slc23a1 is important in placental ascorbic-acid transport. Slc23a1(-/-) mice died within a few minutes of birth with respiratory failure and intraparenchymal brain hemorrhage. Lungs showed no postnatal expansion but had normal surfactant protein B levels. Brain hemorrhage was unlikely to be simply a form of scurvy since Slc23a1(-/-) mice showed no hemorrhage in any other tissues and their skin had normal skin 4-hydroxyproline levels despite low ascorbic-acid content. We conclude that Slc23a1 is required for transport of ascorbic acid into many tissues and across the placenta. Deficiency of the transporter is lethal in newborn mice, thereby revealing a previously unrecognized requirement for ascorbic acid in the perinatal period.


Assuntos
Ácido Ascórbico/metabolismo , Encéfalo/metabolismo , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Simportadores , Animais , Desenvolvimento Embrionário e Fetal , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Genes Essenciais , Humanos , Camundongos , Camundongos Knockout , Transportadores de Ânions Orgânicos Dependentes de Sódio/deficiência , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Mapeamento por Restrição , Transportadores de Sódio Acoplados à Vitamina C
19.
Pediatrics ; 147(5)2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33863843

RESUMO

BACKGROUND: In preterm infants who require mechanical ventilation (MV), volume-targeted ventilation (VTV) modes are associated with lower rates of bronchopulmonary dysplasia compared with pressure-limited ventilation. Bronchopulmonary dysplasia rates in our NICU were higher than desired, prompting quality improvement initiatives to improve MV by increasing the use of VTV. METHODS: We implemented and tested interventions over a 3-year period. Primary outcomes were the percentage of conventional MV hours when any-VTV mode was used and the percentage of conventional MV hours when an exclusively VTV mode was used. Exclusively VTV modes were modes in which all breaths were volume targeted. We evaluated outcomes during 3 project periods: baseline (May 2016-December 2016); epoch 1 (December 2016-October 2018), increasing the use of any-VTV mode; and epoch 2 (October 2018-November 2019), increasing the use of exclusively VTV modes. RESULTS: Use of any-VTV mode increased from 18 694 of 22 387 (83%) MV hours during baseline to 72 846 of 77 264 (94%) and 58 174 of 60 605 (96%) MV hours during epochs 1 and 2, respectively (P < .001). Use of exclusively VTV increased from 5967 of 22 387 (27%) during baseline to 47 364 of 77 264 (61%) and 46 091 of 60 605 (76%) of all conventional MV hours during epochs 1 and 2, respectively (P < .001). In statistical process control analyses, multiple interventions were associated with improvements in primary outcomes. Measured clinical outcomes were unchanged. CONCLUSIONS: Quality improvement interventions were associated with improved use of VTV but no change in measured clinical outcomes.


Assuntos
Displasia Broncopulmonar/prevenção & controle , Unidades de Terapia Intensiva Neonatal , Melhoria de Qualidade , Respiração Artificial/métodos , Displasia Broncopulmonar/etiologia , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Respiração Artificial/efeitos adversos , Respiração Artificial/estatística & dados numéricos , Fatores de Tempo
20.
J Clin Invest ; 131(1)2021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-33180746

RESUMO

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) novel coronavirus 2019 (COVID-19) global pandemic has led to millions of cases and hundreds of thousands of deaths. While older adults appear at high risk for severe disease, hospitalizations and deaths due to SARS-CoV-2 among children have been relatively rare. Integrating single-cell RNA sequencing (scRNA-seq) of developing mouse lung with temporally resolved immunofluorescence in mouse and human lung tissue, we found that expression of SARS-CoV-2 Spike protein primer TMPRSS2 was highest in ciliated cells and type I alveolar epithelial cells (AT1), and TMPRSS2 expression increased with aging in mice and humans. Analysis of autopsy tissue from fatal COVID-19 cases detected SARS-CoV-2 RNA most frequently in ciliated and secretory cells in airway epithelium and AT1 cells in peripheral lung. SARS-CoV-2 RNA was highly colocalized in cells expressing TMPRSS2. Together, these data demonstrate the cellular spectrum infected by SARS-CoV-2 in lung epithelium and suggest that developmental regulation of TMPRSS2 may underlie the relative protection of infants and children from severe respiratory illness.


Assuntos
Células Epiteliais Alveolares/enzimologia , COVID-19/enzimologia , COVID-19/metabolismo , Regulação Enzimológica da Expressão Gênica , SARS-CoV-2/metabolismo , Serina Endopeptidases/biossíntese , Adulto , Envelhecimento , Células Epiteliais Alveolares/patologia , Células Epiteliais Alveolares/virologia , Animais , COVID-19/patologia , Pré-Escolar , Modelos Animais de Doenças , Feminino , Humanos , Lactente , Masculino , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA