Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Clin Genet ; 105(2): 190-195, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-37821225

RESUMO

Congenital alveolar dysplasia (CAD) belongs to rare lethal lung developmental disorders (LLDDs) in neonates, manifesting with acute respiratory failure and pulmonary arterial hypertension refractory to treatment. The majority of CAD cases have been associated with copy-number variant (CNV) deletions at 17q23.1q23.2 or 5p12. Most CNV deletions at 17q23.1q23.2 were recurrent and encompassed two closely located genes, TBX4 and TBX2. In a few CAD cases, intragenic frameshifting deletions or single-nucleotide variants (SNVs) involved TBX4 but not TBX2. Here, we describe a male neonate who died at 27 days of life from acute respiratory failure caused by lung growth arrest along the spectrum of CAD confirmed by histopathological assessment. Trio-based genome sequencing revealed in the proband a novel non-recurrent ~1.07 Mb heterozygous CNV deletion at 17q23.2, encompassing TBX4 that arose de novo on the paternal chromosome. This is the first report of a larger-sized CNV deletion in a CAD patient involving TBX4 and leaving TBX2 intact. Our results, together with previous reports, indicate that perturbations of TBX4, rather than TBX2, cause severe lung phenotypes in humans.


Assuntos
Síndrome do Desconforto Respiratório do Recém-Nascido , Insuficiência Respiratória , Humanos , Recém-Nascido , Masculino , Hipertensão Pulmonar Primária Familiar , Pulmão , Fenótipo , Proteínas com Domínio T/genética
2.
Am J Respir Crit Care Med ; 208(6): 709-725, 2023 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-37463497

RESUMO

Rationale: Alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV) is a lethal developmental disorder of lung morphogenesis caused by insufficiency of FOXF1 (forkhead box F1) transcription factor function. The cellular and transcriptional mechanisms by which FOXF1 deficiency disrupts human lung formation are unknown. Objectives: To identify cell types, gene networks, and cell-cell interactions underlying the pathogenesis of ACDMPV. Methods: We used single-nucleus RNA and assay for transposase-accessible chromatin sequencing, immunofluorescence confocal microscopy, and RNA in situ hybridization to identify cell types and molecular networks influenced by FOXF1 in ACDMPV lungs. Measurements and Main Results: Pathogenic single-nucleotide variants and copy-number variant deletions involving the FOXF1 gene locus in all subjects with ACDMPV (n = 6) were accompanied by marked changes in lung structure, including deficient alveolar development and a paucity of pulmonary microvasculature. Single-nucleus RNA and assay for transposase-accessible chromatin sequencing identified alterations in cell number and gene expression in endothelial cells (ECs), pericytes, fibroblasts, and epithelial cells in ACDMPV lungs. Distinct cell-autonomous roles for FOXF1 in capillary ECs and pericytes were identified. Pathogenic variants involving the FOXF1 gene locus disrupt gene expression in EC progenitors, inhibiting the differentiation or survival of capillary 2 ECs and cell-cell interactions necessary for both pulmonary vasculogenesis and alveolar type 1 cell differentiation. Loss of the pulmonary microvasculature was associated with increased VEGFA (vascular endothelial growth factor A) signaling and marked expansion of systemic bronchial ECs expressing COL15A1 (collagen type XV α 1 chain). Conclusions: Distinct FOXF1 gene regulatory networks were identified in subsets of pulmonary endothelial and fibroblast progenitors, providing both cellular and molecular targets for the development of therapies for ACDMPV and other diffuse lung diseases of infancy.


Assuntos
Síndrome da Persistência do Padrão de Circulação Fetal , Recém-Nascido , Humanos , Síndrome da Persistência do Padrão de Circulação Fetal/genética , Síndrome da Persistência do Padrão de Circulação Fetal/patologia , Redes Reguladoras de Genes/genética , Fator A de Crescimento do Endotélio Vascular/genética , Células Endoteliais/patologia , Multiômica , Pulmão/patologia , RNA , Fatores de Transcrição Forkhead/genética
3.
Am J Respir Cell Mol Biol ; 65(4): 442-460, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34101541

RESUMO

Alveolar epithelial type 2 cells (AEC2s), the facultative progenitors of lung alveoli, are typically identified through the use of the canonical markers, SFTPC and ABCA3. Self-renewing AEC2-like cells have been generated from human induced pluripotent stem cells (iPSCs) through the use of knock-in SFTPC fluorochrome reporters. However, developmentally, SFTPC expression onset begins in the fetal distal lung bud tip and thus is not specific to mature AEC2s. Furthermore, SFTPC reporters appear to identify only those iPSC-derived AEC2s (iAEC2s) expressing the highest SFTPC levels. Here, we generate an ABCA3 knock-in GFP fusion reporter (ABCA3:GFP) that enables the purification of iAEC2s while allowing visualization of lamellar bodies, organelles associated with AEC2 maturation. Using an SFTPCtdTomato and ABCA3:GFP bifluorescent line for in vitro distal lung-directed differentiation, we observe later onset of ABCA3:GFP expression and broader identification of the subsequently emerging iAEC2 population based on ABCA3:GFP expression compared with SFTPCtdTomato expression. Comparing ABCA3:GFP/SFTPCtdTomato double-positive with ABCA3:GFP single-positive (SP) cells by RNA sequencing and functional studies reveals iAEC2 cellular heterogeneity with both populations functionally processing surfactant proteins but the SP cells exhibiting faster growth kinetics, increased clonogenicity, increased expression of progenitor markers, lower levels of SFTPC expression, and lower levels of AEC2 maturation markers. Over time, we observe that each population (double-positive and SP) gives rise to the other and each can serve as the parents of indefinitely self-renewing iAEC2 progeny. Our results indicate that iAEC2s are a heterogeneous population of cells with differing proliferation versus maturation properties, the majority of which can be tracked and purified using the ABCA3:GFP reporter or surrogate cell surface proteins, such as SLC34A2 and CPM.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Células Epiteliais Alveolares/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Alvéolos Pulmonares/citologia , Proteína C Associada a Surfactante Pulmonar/metabolismo , Diferenciação Celular/fisiologia , Células Epiteliais/metabolismo , Humanos , Pulmão/metabolismo , Proteínas Associadas a Surfactantes Pulmonares/metabolismo
4.
J Pediatr ; 206: 286-292.e1, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30413314

RESUMO

Variable lung disease was documented in 2 infants with heterozygous TBX4 mutations; their clinical presentations, pathology, and outcomes were distinct. These findings demonstrate that TBX4 gene mutations are associated with neonatal respiratory failure and highlight the wide spectrum of clinicopathological outcomes that have implications for patient diagnosis and management.


Assuntos
Mutação/genética , Insuficiência Respiratória/genética , Insuficiência Respiratória/patologia , Proteínas com Domínio T/genética , Feminino , Humanos , Recém-Nascido , Masculino
5.
J Immunol ; 197(11): 4228-4239, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27794000

RESUMO

KIF3A, the gene encoding kinesin family member 3A, is a susceptibility gene locus associated with asthma; however, mechanisms by which KIF3A might influence the pathogenesis of the disorder are unknown. In this study, we deleted the mouse Kif3a gene in airway epithelial cells. Both homozygous and heterozygous Kif3a gene-deleted mice were highly susceptible to aeroallergens from Aspergillus fumigatus and the house dust mite, resulting in an asthma-like pathology characterized by increased goblet cell metaplasia, airway hyperresponsiveness, and Th2-mediated inflammation. Deletion of the Kif3a gene increased the severity of pulmonary eosinophilic inflammation and expression of cytokines (Il-4, Il-13, and Il-17a) and chemokine (Ccl11) RNAs following pulmonary exposure to Aspergillus extract. Inhibition of Kif3a disrupted the structure of motile cilia and impaired mucociliary clearance, barrier function, and epithelial repair, demonstrating additional mechanisms by which deficiency of KIF3A in respiratory epithelial cells contributes to pulmonary pathology. Airway epithelial KIF3A suppresses Th2 pulmonary inflammation and airway hyperresponsiveness following aeroallergen exposure, implicating epithelial microtubular functions in the pathogenesis of Th2-mediated lung pathology.


Assuntos
Alérgenos/imunologia , Aspergillus fumigatus/imunologia , Asma/imunologia , Células Epiteliais/imunologia , Cinesinas/imunologia , Mucosa Respiratória/imunologia , Células Th2/imunologia , Animais , Asma/induzido quimicamente , Asma/genética , Asma/patologia , Citocinas/genética , Citocinas/imunologia , Células Epiteliais/patologia , Cinesinas/genética , Pulmão/imunologia , Pulmão/patologia , Camundongos , Camundongos Transgênicos , Mucosa Respiratória/patologia , Células Th2/patologia
6.
J Clin Invest ; 125(5): 2021-31, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25866971

RESUMO

Epithelial cells that line the conducting airways provide the initial barrier and innate immune responses to the abundant particles, microbes, and allergens that are inhaled throughout life. The transcription factors SPDEF and FOXA3 are both selectively expressed in epithelial cells lining the conducting airways, where they regulate goblet cell differentiation and mucus production. Moreover, these transcription factors are upregulated in chronic lung disorders, including asthma. Here, we show that expression of SPDEF or FOXA3 in airway epithelial cells in neonatal mice caused goblet cell differentiation, spontaneous eosinophilic inflammation, and airway hyperresponsiveness to methacholine. SPDEF expression promoted DC recruitment and activation in association with induction of Il33, Csf2, thymic stromal lymphopoietin (Tslp), and Ccl20 transcripts. Increased Il4, Il13, Ccl17, and Il25 expression was accompanied by recruitment of Th2 lymphocytes, group 2 innate lymphoid cells, and eosinophils to the lung. SPDEF was required for goblet cell differentiation and pulmonary Th2 inflammation in response to house dust mite (HDM) extract, as both were decreased in neonatal and adult Spdef(-/-) mice compared with control animals. Together, our results indicate that SPDEF causes goblet cell differentiation and Th2 inflammation during postnatal development and is required for goblet cell metaplasia and normal Th2 inflammatory responses to HDM aeroallergen.


Assuntos
Antígenos de Dermatophagoides/toxicidade , Células Epiteliais/metabolismo , Células Caliciformes/fisiologia , Pulmão/imunologia , Proteínas Proto-Oncogênicas c-ets/fisiologia , Eosinofilia Pulmonar/imunologia , Células Th2/imunologia , Fatores Etários , Animais , Animais Recém-Nascidos , Diferenciação Celular , Quimiocina CCL20/biossíntese , Quimiocina CCL20/genética , Quimiotaxia de Leucócito , Citocinas/antagonistas & inibidores , Citocinas/biossíntese , Citocinas/genética , Células Dendríticas/imunologia , Eosinófilos/fisiologia , Fator 3-gama Nuclear de Hepatócito/fisiologia , Interleucinas/biossíntese , Interleucinas/genética , Metaplasia , Cloreto de Metacolina/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-ets/genética , Eosinofilia Pulmonar/etiologia , Eosinofilia Pulmonar/metabolismo , Eosinofilia Pulmonar/patologia , Proteínas Recombinantes de Fusão/metabolismo , Transgenes , Linfopoietina do Estroma do Timo
7.
Am J Respir Cell Mol Biol ; 49(5): 845-54, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23795648

RESUMO

Pulmonary surfactant protein-C (SP-C) gene-targeted mice (Sftpc(-/-)) develop progressive lung inflammation and remodeling. We hypothesized that SP-C deficiency reduces the ability to suppress repetitive inflammatory injury. Sftpc(+/+) and Sftpc(-/-) mice given three doses of bacterial LPS developed airway and airspace inflammation, which was more intense in the Sftpc(-/-) mice at 3 and 5 days after the final dose. Compared with Sftpc(+/+)mice, inflammatory injury persisted in the lungs of Sftpc(-/-) mice 30 days after the final LPS challenge. Sftpc(-/-) mice showed LPS-induced airway goblet cell hyperplasia with increased detection of Sam pointed Ets domain and FoxA3 transcription factors. Sftpc(-/-) type II alveolar epithelial cells had increased cytokine expression after LPS exposure relative to Sftpc(+/+) cells, indicating that type II cell dysfunction contributes to inflammatory sensitivity. Microarray analyses of isolated type II cells identified a pattern of enhanced expression of inflammatory genes consistent with an intrinsic low-level inflammation resulting from SP-C deficiency. SP-C-containing clinical surfactant extract (Survanta) or SP-C/phospholipid vesicles blocked LPS signaling through the LPS receptor (Toll-like receptor [TLR] 4/CD14/MD2) in human embryonic kidney 293T cells, indicating that SP-C blocks LPS-induced cytokine production by a TLR4-dependent mechanism. Phospholipid vesicles alone did not modify the TLR4 response. In vivo deficiency of SP-C leads to inflammation, increased cytokine production by type II cells, and persistent inflammation after repetitive LPS stimulation.


Assuntos
Endotoxinas , Pulmão/metabolismo , Peptídeos/deficiência , Pneumonia/metabolismo , Células Epiteliais Alveolares/imunologia , Células Epiteliais Alveolares/metabolismo , Células Epiteliais Alveolares/patologia , Animais , Produtos Biológicos/farmacologia , Citocinas/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica , Células Caliciformes/imunologia , Células Caliciformes/metabolismo , Células Caliciformes/patologia , Células HEK293 , Fator 3-gama Nuclear de Hepatócito/metabolismo , Humanos , Hiperplasia , Imunidade Inata , Mediadores da Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Receptores de Lipopolissacarídeos/metabolismo , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/patologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Peptídeos/genética , Pneumonia/induzido quimicamente , Pneumonia/genética , Pneumonia/imunologia , Pneumonia/patologia , Proteínas Proto-Oncogênicas c-ets/metabolismo , Proteína C Associada a Surfactante Pulmonar , Transdução de Sinais , Fatores de Tempo , Receptor 4 Toll-Like/metabolismo
8.
J Clin Invest ; 117(4): 978-88, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17347682

RESUMO

Goblet cell hyperplasia and mucous hypersecretion contribute to the pathogenesis of chronic pulmonary diseases including cystic fibrosis, asthma, and chronic obstructive pulmonary disease. In the present work, mouse SAM pointed domain-containing ETS transcription factor (SPDEF) mRNA and protein were detected in subsets of epithelial cells lining the trachea, bronchi, and tracheal glands. SPDEF interacted with the C-terminal domain of thyroid transcription factor 1, activating transcription of genes expressed selectively in airway epithelial cells, including Sftpa, Scgb1a1, Foxj1, and Sox17. Expression of Spdef in the respiratory epithelium of adult transgenic mice caused goblet cell hyperplasia, inducing both acidic and neutral mucins in vivo, and stainined for both acidic and neutral mucins in vivo. SPDEF expression was increased at sites of goblet cell hyperplasia caused by IL-13 and dust mite allergen in a process that was dependent upon STAT-6. SPDEF was induced following intratracheal allergen exposure and after Th2 cytokine stimulation and was sufficient to cause goblet cell differentiation of Clara cells in vivo.


Assuntos
Células Caliciformes/fisiologia , Hiperplasia/fisiopatologia , Proteínas Proto-Oncogênicas c-ets/genética , Mucosa Respiratória/fisiologia , Animais , Sítios de Ligação , Diferenciação Celular , Linhagem Celular , Citocinas/fisiologia , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/fisiologia , Regulação da Expressão Gênica , Variação Genética , Células Caliciformes/citologia , História do Século XVI , Humanos , Pulmão/crescimento & desenvolvimento , Pulmão/fisiologia , Neoplasias Pulmonares , Camundongos , Proteínas Proto-Oncogênicas c-ets/metabolismo , Proteínas Proto-Oncogênicas c-ets/fisiologia , RNA Mensageiro/genética , Mucosa Respiratória/patologia , Mucosa Respiratória/fisiopatologia , Células Th2/fisiologia , Traqueia/fisiologia , Fatores de Transcrição , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA