Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 250
Filtrar
1.
Development ; 148(10)2021 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-34015093

RESUMO

Congenital diaphragmatic hernia (CDH) is a developmental disorder associated with diaphragm defects and lung hypoplasia. The etiology of CDH is complex and its clinical presentation is variable. We investigated the role of the pulmonary mesothelium in dysregulated lung growth noted in the Wt1 knockout mouse model of CDH. Loss of WT1 leads to intrafetal effusions, altered lung growth, and branching defects prior to normal closure of the diaphragm. We found significant differences in key genes; however, when Wt1 null lungs were cultured ex vivo, growth and branching were indistinguishable from wild-type littermates. Micro-CT imaging of embryos in situ within the uterus revealed a near absence of space in the dorsal chest cavity, but no difference in total chest cavity volume in Wt1 null embryos, indicating a redistribution of pleural space. The altered space and normal ex vivo growth suggest that physical constraints are contributing to the CDH lung phenotype observed in this mouse model. These studies emphasize the importance of examining the mesothelium and chest cavity as a whole, rather than focusing on single organs in isolation to understand early CDH etiology.


Assuntos
Diafragma/embriologia , Epitélio/patologia , Hérnias Diafragmáticas Congênitas/genética , Pulmão/embriologia , Proteínas WT1/genética , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Knockout , Tórax/anatomia & histologia
2.
Dev Biol ; 467(1-2): 30-38, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32827499

RESUMO

The mammalian muscularized diaphragm is essential for respiration and defects in the developing diaphragm cause a common and frequently lethal birth defect, congenital diaphragmatic hernia (CDH). Human genetic studies have implicated more than 150 genes and multiple molecular pathways in CDH, but few of these have been validated because of the expense and time to generate mouse mutants. The pleuroperitoneal folds (PPFs) are transient embryonic structures in diaphragm development and defects in PPFs lead to CDH. We have developed a system to culture PPF fibroblasts from E12.5 mouse embryos and show that these fibroblasts, in contrast to the commonly used NIH 3T3 fibroblasts, maintain expression of key genes in normal diaphragm development. Using pharmacological and genetic manipulations that result in CDH in vivo, we also demonstrate that differences in proliferation provide a rapid means of distinguishing healthy and impaired PPF fibroblasts. Thus, the PPF fibroblast cell culture system is an efficient tool for assaying the functional significance of CDH candidate genes and molecular pathways and will be an important resource for elucidating the complex etiology of CDH.


Assuntos
Técnicas de Cultura de Células , Diafragma/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Hérnias Diafragmáticas Congênitas/embriologia , Animais , Feminino , Humanos , Masculino , Camundongos , Células NIH 3T3
3.
Int J Mol Sci ; 22(12)2021 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-34198563

RESUMO

Congenital diaphragmatic hernia (CDH) is a relatively common major life-threatening birth defect that results in significant mortality and morbidity depending primarily on lung hypoplasia, persistent pulmonary hypertension, and cardiac dysfunction. Despite its clinical relevance, CDH multifactorial etiology is still not completely understood. We reviewed current knowledge on normal diaphragm development and summarized genetic mutations and related pathways as well as cellular mechanisms involved in CDH. Our literature analysis showed that the discovery of harmful de novo variants in the fetus could constitute an important tool for the medical team during pregnancy, counselling, and childbirth. A better insight into the mechanisms regulating diaphragm development and genetic causes leading to CDH appeared essential to the development of new therapeutic strategies and evidence-based genetic counselling to parents. Integrated sequencing, development, and bioinformatics strategies could direct future functional studies on CDH; could be applied to cohorts and consortia for CDH and other birth defects; and could pave the way for potential therapies by providing molecular targets for drug discovery.


Assuntos
Hérnias Diafragmáticas Congênitas/genética , Diafragma/embriologia , Diafragma/patologia , Predisposição Genética para Doença , Hérnias Diafragmáticas Congênitas/classificação , Hérnias Diafragmáticas Congênitas/diagnóstico , Hérnias Diafragmáticas Congênitas/patologia , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Prognóstico
4.
Radiographics ; 40(5): 1205-1218, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32706612

RESUMO

The diaphragm serves as an anatomic border between the abdominal and thoracic cavities. Pathologic conditions traversing the diaphragm are often incompletely described and may be overlooked, resulting in diagnostic delays. Several routes allow abdominal contents or pathologic processes to spread into the thorax, including along normal transphrenic structures, through congenital defects in the diaphragm, through inherent areas of weakness between muscle groups, or by pathways created by tissue destruction, trauma, or iatrogenic injuries. A thorough knowledge of the anatomy of the diaphragm can inform an accurate differential diagnosis. Often, intraperitoneal pathologic conditions crossing the diaphragm may be overlooked if axial imaging is the only approach to this complex region because of the horizontal orientation of much of the diaphragm. Multiplanar capabilities of volumetric CT and MRI provide insight into the pathways where pathologic conditions may traverse this border. Knowledge of these characteristic routes and use of multiplanar imaging are critical for depiction of specific transdiaphragmatic pathologic conditions.©RSNA, 2020.


Assuntos
Abdome/diagnóstico por imagem , Diafragma/anatomia & histologia , Hérnia Diafragmática/diagnóstico por imagem , Doenças Torácicas/diagnóstico por imagem , Abdome/patologia , Diafragma/embriologia , Humanos , Imageamento por Ressonância Magnética , Doenças Torácicas/patologia , Tomografia Computadorizada por Raios X
5.
Dev Biol ; 440(2): 64-73, 2018 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-29679560

RESUMO

The diaphragm is a mammalian skeletal muscle essential for respiration and for separating the thoracic and abdominal cavities. Development of the diaphragm requires the coordinated development of muscle, muscle connective tissue, tendon, nerves, and vasculature that derive from different embryonic sources. However, defects in diaphragm development are common and the cause of an often deadly birth defect, Congenital Diaphragmatic Hernia (CDH). Here we comprehensively describe the normal developmental origin and complex spatial-temporal relationship between the different developing tissues to form a functional diaphragm using a developmental series of mouse embryos genetically and immunofluorescently labeled and analyzed in whole mount. We find that the earliest developmental events are the emigration of muscle progenitors from cervical somites followed by the projection of phrenic nerve axons from the cervical neural tube. Muscle progenitors and phrenic nerve target the pleuroperitoneal folds (PPFs), transient pyramidal-shaped structures that form between the thoracic and abdominal cavities. Subsequently, the PPFs expand across the surface of the liver to give rise to the muscle connective tissue and central tendon, and the leading edge of their expansion precedes muscle morphogenesis, formation of the vascular network, and outgrowth and branching of the phrenic nerve. Thus development and morphogenesis of the PPFs is critical for diaphragm formation. In addition, our data indicate that the earliest events in diaphragm development are critical for the etiology of CDH and instrumental to the evolution of the diaphragm. CDH initiates prior to E12.5 in mouse and suggests that defects in the early PPF formation or their ability to recruit muscle are an important source of CDH. Also, the recruitment of muscle progenitors from cervical somites to the nascent PPFs is uniquely mammalian and a key developmental innovation essential for the evolution of the muscularized diaphragm.


Assuntos
Diafragma/embriologia , Diafragma/fisiologia , Animais , Tecido Conjuntivo/embriologia , Tecido Conjuntivo/fisiologia , Modelos Animais de Doenças , Regulação da Expressão Gênica no Desenvolvimento/genética , Genes Controladores do Desenvolvimento/genética , Mamíferos , Camundongos , Camundongos Endogâmicos C57BL , Morfogênese , Desenvolvimento Muscular/fisiologia , Músculo Esquelético/embriologia , Músculo Esquelético/crescimento & desenvolvimento , Músculo Esquelético/fisiologia
6.
Fetal Pediatr Pathol ; 38(4): 335-339, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30912683

RESUMO

Introduction: Congenital diaphragmatic eventration (CDE) is defined as the abnormal elevation of the diaphragm, due to incomplete muscularization of the diaphragm with a thin membranous sheet replacing normal diaphragmatic muscle. Case report: We report a prenatal case with a diaphragmatic mesothelial cyst combined with CDE. Conclusion: A large cystic mass between the thoracic wall and the liver in early pregnancy is highly suggestive of cystic diaphragm.


Assuntos
Diafragma/anormalidades , Diafragma/embriologia , Eventração Diafragmática/diagnóstico , Adulto , Anormalidades Congênitas , Diagnóstico Diferencial , Epitélio/patologia , Feminino , Feto , Humanos , Fígado/embriologia , Masculino , Gravidez , Diagnóstico Pré-Natal , Parede Torácica/embriologia , Ultrassonografia
7.
J Cell Sci ; 129(17): 3295-308, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27466379

RESUMO

Correct innervation of the main respiratory muscle in mammals, namely the thoracic diaphragm, is a crucial pre-requisite for the functionality of this muscle and the viability of the entire organism. Systemic impairment of Sema3A-Npn-1 (Npn-1 is also known as NRP1) signaling causes excessive branching of phrenic nerves in the diaphragm and into the central tendon region, where the majority of misguided axons innervate ectopic musculature. To elucidate whether these ectopic muscles are a result of misguidance of myoblast precursors due to the loss of Sema3A-Npn-1 signaling, we conditionally ablated Npn-1 in somatic motor neurons, which led to a similar phenotype of phrenic nerve defasciculation and, intriguingly, also formation of innervated ectopic muscles. We therefore hypothesize that ectopic myocyte fusion is caused by additional factors released by misprojecting growth cones. Slit2 and its Robo receptors are expressed by phrenic motor axons and migrating myoblasts, respectively, during innervation of the diaphragm. In vitro analyses revealed a chemoattractant effect of Slit2 on primary diaphragm myoblasts. Thus, we postulate that factors released by motor neuron growth cones have an influence on the migration properties of myoblasts during establishment of the diaphragm.


Assuntos
Diafragma/inervação , Diafragma/metabolismo , Desenvolvimento Muscular , Neuropilina-1/metabolismo , Semaforina-3A/metabolismo , Transdução de Sinais , Animais , Fasciculação Axônica , Diafragma/embriologia , Embrião de Mamíferos/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Neurônios Motores/metabolismo , Mioblastos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Nervo Frênico/metabolismo , Receptores Imunológicos/metabolismo , Células-Tronco/metabolismo , Tendões/metabolismo , Proteínas Roundabout
8.
Pediatr Surg Int ; 34(3): 315-321, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29196881

RESUMO

PURPOSE: Congenital diaphragmatic hernia (CDH) and associated pulmonary hypoplasia are thought to be caused by a malformation of the underlying diaphragmatic and airway mesenchyme. GATA binding protein 6 (Gata-6) is a zinc finger-containing transcription factor that plays a crucial role during diaphragm and lung development. In the primordial diaphragm, Gata-6 expression is restricted to mesenchymal compartments of the pleuroperitoneal folds (PPFs). In addition, Gata-6 is essential for airway branching morphogenesis through upregulation of mesenchymal signaling. Recently, mutations in Gata-6 have been linked to human CDH. We hypothesized that diaphragmatic and pulmonary Gata-6 expression is decreased in the nitrofen-induced CDH model. METHODS: Time-mated rats were exposed to either nitrofen or vehicle on gestational day 9 (D9). Fetal diaphragms (n = 72) and lungs (n = 48) were microdissected on selected timepoints D13, D15 and D18, and divided into control and nitrofen-exposed specimens (n = 12 per sample, timepoint and experimental group, respectively). Diaphragmatic and pulmonary gene expression of Gata-6 was analyzed by qRT-PCR. Immunofluorescence-double staining for Gata-6 was combined with the diaphragmatic mesenchymal marker Gata-4 and the pulmonary mesenchymal marker Fgf-10 to evaluate protein expression and localization in fetal diaphragms and lungs. RESULTS: Relative mRNA expression levels of Gata-6 were significantly decreased in PPFs on D13 (0.57 ± 0.21 vs. 2.27 ± 1.30; p < 0.05), developing diaphragms (0.94 ± 0.59 vs. 2.28 ± 1.89; p < 0.05) and lungs (0.56 ± 0.16 vs. 0.71 ± 0.39; p < 0.05) on D15 and fully muscularized diaphragms (1.20 ± 1.10 vs. 2.52 ± 1.86; p < 0.05) and differentiated lungs (0.56 ± 0.05 vs. 0.77 ± 0.14; p < 0.05) on D18 of nitrofen-exposed fetuses compared to controls. Confocal laser scanning microscopy demonstrated markedly diminished immunofluorescence of Gata-6 mainly in diaphragmatic and pulmonary mesenchyme, which was associated with a reduction of proliferating mesenchymal cells in nitrofen-exposed fetuses on D13, D15, and D18 compared to controls. CONCLUSION: Decreased Gata-6 expression during diaphragmatic development and lung branching morphogenesis may disrupt mesenchymal cell proliferation, causing malformed PPFs and reduced airway branching, thus leading to diaphragmatic defects and pulmonary hypoplasia in the nitrofen-induced CDH model.


Assuntos
Diafragma/metabolismo , Feto/metabolismo , Fator de Transcrição GATA6/metabolismo , Hérnias Diafragmáticas Congênitas , Pulmão/metabolismo , Mesoderma/metabolismo , Animais , Diafragma/embriologia , Feminino , Fator de Transcrição GATA6/genética , Regulação da Expressão Gênica no Desenvolvimento , Pulmão/embriologia , Microscopia Confocal , Éteres Fenílicos , Gravidez , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley
9.
Am J Pathol ; 186(10): 2532-43, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27565037

RESUMO

Congenital diaphragmatic hernia (CDH) is one of the most common and lethal congenital anomalies, and significant evidence is available in support of a genetic contribution to its etiology, including single-gene knockout mice associated with diaphragmatic defects, rare monogenetic disorders in humans, familial aggregation, and association of CDH with chromosomal abnormalities. Structural lung defects in the form of lung hypoplasia are almost invariably seen in patients with CDH and frequently in animal models of this condition. Better understanding of the mechanisms of pulmonary defects in CDH has the potential for creating targeted therapies, particularly in postnatal stages, when therapeutics can have maximum clinical impact on the surviving cohorts. Successful treatment of CDH is dependent on the integration of human genomic and genetic data with developmental expression profiling, mouse knockouts, and gene network and pathway modeling, which have generated a large number of candidate genes and pathways for follow-up studies. In particular, defective alveolarization appears to be a common and potentially actionable phenotype in both patients and animal models.


Assuntos
Aberrações Cromossômicas , Hérnias Diafragmáticas Congênitas/genética , Hérnias Diafragmáticas Congênitas/patologia , Animais , Diafragma/embriologia , Diafragma/patologia , Modelos Animais de Doenças , Feminino , Hérnias Diafragmáticas Congênitas/terapia , Humanos , Lactente , Pulmão/embriologia , Pulmão/patologia , Camundongos , Camundongos Knockout
10.
Proc Natl Acad Sci U S A ; 111(34): 12450-5, 2014 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-25107291

RESUMO

Congenital diaphragmatic hernia (CDH) is a common and severe birth defect. Despite its clinical significance, the genetic and developmental pathways underlying this disorder are incompletely understood. In this study, we report a catalog of variants detected by a whole exome sequencing study on 275 individuals with CDH. Predicted pathogenic variants in genes previously identified in either humans or mice with diaphragm defects are enriched in our CDH cohort compared with 120 size-matched random gene sets. This enrichment was absent in control populations. Variants in these critical genes can be found in up to 30.9% of individuals with CDH. In addition, we filtered variants by using genes derived from regions of recurrent copy number variations in CDH, expression profiles of the developing diaphragm, protein interaction networks expanded from the known CDH-causing genes, and prioritized genes with ultrarare and highly disruptive variants, in 11.3% of CDH patients. These strategies have identified several high priority genes and developmental pathways that likely contribute to the CDH phenotype. These data are valuable for comparison of candidate genes generated from whole exome sequencing of other CDH cohorts or multiplex kindreds and provide ideal candidates for further functional studies. Furthermore, we propose that these genes and pathways will enhance our understanding of the heterogeneous molecular etiology of CDH.


Assuntos
Hérnias Diafragmáticas Congênitas/etiologia , Hérnias Diafragmáticas Congênitas/genética , Animais , Estudos de Coortes , Biologia Computacional , Variações do Número de Cópias de DNA , Diafragma/embriologia , Exoma , Variação Genética , Hérnias Diafragmáticas Congênitas/embriologia , Humanos , Camundongos , Mapas de Interação de Proteínas
11.
Proc Natl Acad Sci U S A ; 111(46): 16556-61, 2014 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-25368159

RESUMO

The motoneural control of skeletal muscle contraction requires the neuromuscular junction (NMJ), a midmuscle synapse between the motor nerve and myotube. The formation and maintenance of NMJs are orchestrated by the muscle-specific receptor tyrosine kinase (MuSK). Motor neuron-derived agrin activates MuSK via binding to MuSK's coreceptor Lrp4, and genetic defects in agrin underlie a congenital myasthenic syndrome (an NMJ disorder). However, MuSK-dependent postsynaptic differentiation of NMJs occurs in the absence of a motor neuron, indicating a need for nerve/agrin-independent MuSK activation. We previously identified the muscle protein Dok-7 as an essential activator of MuSK. Although NMJ formation requires agrin under physiological conditions, it is dispensable for NMJ formation experimentally in the absence of the neurotransmitter acetylcholine, which inhibits postsynaptic specialization. Thus, it was hypothesized that MuSK needs agrin together with Lrp4 and Dok-7 to achieve sufficient activation to surmount inhibition by acetylcholine. Here, we show that forced expression of Dok-7 in muscle enhanced MuSK activation in mice lacking agrin or Lrp4 and restored midmuscle NMJ formation in agrin-deficient mice, but not in Lrp4-deficient mice, probably due to the loss of Lrp4-dependent presynaptic differentiation. However, these NMJs in agrin-deficient mice rapidly disappeared after birth, and postsynaptic specializations emerged ectopically throughout myotubes whereas exogenous Dok-7-mediated MuSK activation was maintained. These findings demonstrate that the MuSK activator agrin plays another role essential for the postnatal maintenance, but not for embryonic formation, of NMJs and also for the postnatal, but not prenatal, midmuscle localization of postsynaptic specializations, providing physiological and pathophysiological insight into NMJ homeostasis.


Assuntos
Agrina/fisiologia , Junção Neuromuscular/enzimologia , Receptores Proteína Tirosina Quinases/metabolismo , Agrina/deficiência , Agrina/genética , Processamento Alternativo , Animais , Diafragma/embriologia , Diafragma/crescimento & desenvolvimento , Ativação Enzimática , Feminino , Proteínas Relacionadas a Receptor de LDL , Longevidade/genética , Masculino , Camundongos , Camundongos Transgênicos , Fibras Musculares Esqueléticas/enzimologia , Fibras Musculares Esqueléticas/fisiologia , Proteínas Musculares/deficiência , Proteínas Musculares/fisiologia , Junção Neuromuscular/embriologia , Junção Neuromuscular/crescimento & desenvolvimento , Doenças da Junção Neuromuscular/enzimologia , Doenças da Junção Neuromuscular/genética , Doenças da Junção Neuromuscular/fisiopatologia , Fosforilação , Densidade Pós-Sináptica/fisiologia , Processamento de Proteína Pós-Traducional , Receptores Colinérgicos/fisiologia , Receptores de LDL/deficiência , Receptores de LDL/fisiologia , Proteínas Recombinantes de Fusão/metabolismo , Teste de Desempenho do Rota-Rod
12.
Dev Biol ; 407(1): 40-56, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26278035

RESUMO

The developing diaphragm consists of various differentiating cell types, many of which are not well characterized during organogenesis. One important but incompletely understood tissue, the diaphragmatic mesothelium, is distinctively present from early stages of development. Congenital Diaphragmatic Hernia (CDH) occurs in humans when diaphragm tissue is lost during development, resulting in high morbidity and mortality postnatally. We utilized a Wilms Tumor 1 (Wt1) mutant mouse model to investigate the involvement of the mesothelium in normal diaphragm signaling and development. Additionally, we developed and characterized a Wt1(CreERT2)-driven ß-catenin loss-of-function model of CDH after finding that canonical Wnt signaling and ß-catenin are reduced in Wt1 mutant mesothelium. Mice with ß-catenin loss or constitutive activation induced in the Wt1 lineage are only affected when tamoxifen injection occurs between E10.5 and E11.5, revealing a critical time-frame for Wt1/ ß-catenin activity. Conditional ß-catenin loss phenocopies the Wt1 mutant diaphragm defect, while constitutive activation of ß-catenin on the Wt1 mutant background is sufficient to close the diaphragm defect. Proliferation and apoptosis are affected, but primarily these genetic manipulations appear to lead to a change in normal diaphragm differentiation. Our data suggest a fundamental role for mesothelial signaling in proper formation of the diaphragm.


Assuntos
Diafragma/embriologia , Proteínas Repressoras/fisiologia , Transdução de Sinais , beta Catenina/fisiologia , Animais , Apoptose , Diferenciação Celular , Proliferação de Células , Transição Epitelial-Mesenquimal , Feminino , Hérnias Diafragmáticas Congênitas/etiologia , Humanos , Camundongos , Especificidade de Órgãos , Proteínas WT1
13.
Development ; 140(6): 1231-9, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23406902

RESUMO

The myogenic regulatory factor Myod and insulin-like growth factor 2 (Igf2) have been shown to interact in vitro during myogenic differentiation. In order to understand how they interact in vivo, we produced double-mutant mice lacking both the Myod and Igf2 genes. Surprisingly, these mice display neonatal lethality due to severe diaphragm atrophy. Alteration of diaphragm muscle development occurs as early as 15.5 days post-coitum in the double-mutant embryos and leads to a defect in the terminal differentiation of muscle progenitor cells. A negative-feedback loop was detected between Myod and Igf2 in embryonic muscles. Igf2 belongs to the imprinted H19-Igf2 locus. Molecular analyses show binding of Myod on a mesodermal enhancer (CS9) of the H19 gene. Chromatin conformation capture experiments reveal direct interaction of CS9 with the H19 promoter, leading to increased H19 expression in the presence of Myod. In turn, the non-coding H19 RNA represses Igf2 expression in trans. In addition, Igf2 also negatively regulates Myod expression, possibly by reducing the expression of the Srf transcription factor, a known Myod activator. In conclusion, Igf2 and Myod are tightly co-regulated in skeletal muscles and act in parallel pathways in the diaphragm, where they affect the progression of myogenic differentiation. Igf2 is therefore an essential player in the formation of a functional diaphragm in the absence of Myod.


Assuntos
Diafragma/embriologia , Epistasia Genética/fisiologia , Fator de Crescimento Insulin-Like II/genética , Proteína MyoD/genética , RNA Longo não Codificante/genética , Animais , Animais Recém-Nascidos , Diafragma/crescimento & desenvolvimento , Diafragma/metabolismo , Embrião de Mamíferos , Feminino , Loci Gênicos , Fator de Crescimento Insulin-Like II/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Desenvolvimento Muscular/genética , Proteína MyoD/fisiologia , Organogênese/genética , Gravidez , RNA Longo não Codificante/fisiologia
14.
Proc Natl Acad Sci U S A ; 110(21): E1898-905, 2013 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-23650387

RESUMO

Congenital diaphragmatic hernia (CDH) is a common birth defect that results in a high degree of neonatal morbidity and mortality, but its pathological mechanisms are largely unknown. Therefore, we performed a forward genetic screen in mice to identify unique genes, models, and mechanisms of abnormal diaphragm development. We identified a mutant allele of kinesin family member 7 (Kif7), the disorganized diaphragm (dd). Embryos homozygous for the dd allele possess communicating diaphragmatic hernias, central tendon patterning defects, and increased cell proliferation with diaphragmatic tissue hyperplasia. Because the patterning of the central tendon is undescribed, we analyzed the expression of genes regulating tendonogenesis in dd/dd mutant embryos, and we determined that retinoic acid (RA) signaling was misregulautted. To further investigate the role of Kif7 and RA signaling in the development of the embryonic diaphragm, we established primary mesenchymal cultures of WT embryonic day 13.5 diaphragmatic cells. We determined that RA signaling is necessary for the expression of tendon markers as well as the expression of other CDH-associated genes. Knockdown of Kif7, and retinoic acid receptors alpha (Rara), beta (Rarb), and gamma (Rarg) indicated that RA signaling is dependent on these genes to promote tendonogenesis within the embryonic diaphragm. Taken together, our results provide evidence for a model in which inhibition of RA receptor signaling promotes CDH pathogenesis through a complex gene network.


Assuntos
Padronização Corporal , Diferenciação Celular , Diafragma/embriologia , Hérnias Diafragmáticas Congênitas , Cinesinas/metabolismo , Proteínas Musculares/metabolismo , Transdução de Sinais , Alelos , Animais , Linhagem Celular , Diafragma/patologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Hérnia Diafragmática/embriologia , Hérnia Diafragmática/genética , Hérnia Diafragmática/patologia , Cinesinas/genética , Camundongos , Camundongos Mutantes , Proteínas Musculares/genética , Receptores do Ácido Retinoico/genética , Receptores do Ácido Retinoico/metabolismo , Receptor alfa de Ácido Retinoico , Tendões/embriologia , Tendões/patologia , Tretinoína/metabolismo , Receptor gama de Ácido Retinoico
15.
Pediatr Surg Int ; 32(12): 1127-1132, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27651373

RESUMO

PURPOSE: Congenital diaphragmatic hernia (CDH) is presumed to originate from defects in the primordial diaphragmatic mesenchyme, mainly comprising of muscle connective tissue (MCT). Thus, normal diaphragmatic morphogenesis depends on the structural integrity of the underlying MCT. Developmental mutations that inhibit normal formation of diaphragmatic MCT have been shown to result in CDH. Desmin (DES) is a major filament protein in the MCT, which is essential for the tensile strength of the developing diaphragm muscle. DES -/- knockout mice exhibit significant reductions in stiffness and elasticity of the developing diaphragmatic muscle tissue. Furthermore, sequence changes in the DES gene have recently been identified in human cases of CDH, suggesting that alterations in DES expression may lead to diaphragmatic defects. This study was designed to investigate the hypothesis that diaphragmatic DES expression is decreased in fetal rats with nitrofen-induced CDH. METHODS: Time-mated Sprague-Dawley rats were exposed to either nitrofen or vehicle on gestational day 9 (D9). Fetuses were harvested on selected time-points D13, D15 and D18, and dissected diaphragms (n = 72) were divided into control and nitrofen-exposed specimens (n = 12 per time-point and experimental group, respectively). Laser-capture microdissection was used to obtain diaphragmatic tissue elements. Diaphragmatic gene expression of DES was analyzed by quantitative real-time polymerase chain reaction. Immunofluorescence double staining for DES was combined with the mesenchymal marker GATA4 to evaluate protein expression and localization in developing fetal diaphragms. RESULTS: Relative mRNA expression levels of DES were significantly decreased in pleuroperitoneal folds on D13 (1.49 ± 1.79 vs. 3.47 ± 2.32; p < 0.05), developing diaphragms on D15 (1.49 ± 1.41 vs. 3.94 ± 3.06; p < 0.05) and fully muscularized diaphragms on D18 (2.45 ± 1.47 vs. 5.12 ± 3.37; p < 0.05) of nitrofen-exposed fetuses compared to controls. Confocal laser scanning microscopy demonstrated markedly diminished immunofluorescence of DES mainly in diaphragmatic MCT, which was associated with a reduction of proliferating mesenchymal cells in nitrofen-exposed fetuses on D13, D15 and D18 compared to controls. CONCLUSION: Decreased expression of DES in the fetal diaphragm may disturb the basic integrity of myofibrils and the cytoskeletal network during myogenesis, causing malformed MCT and leading to diaphragmatic defects in the nitrofen-induced CDH model.


Assuntos
Desmina/metabolismo , Diafragma/embriologia , Hérnias Diafragmáticas Congênitas/embriologia , Hérnias Diafragmáticas Congênitas/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Desenvolvimento Fetal , Imunofluorescência , Éteres Fenílicos , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real
16.
Pediatr Surg Int ; 32(2): 135-40, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26519041

RESUMO

PURPOSE: Developmental mutations that inhibit normal formation of extracellular matrix (ECM) in fetal diaphragms have been identified in congenital diaphragmatic hernia (CDH). FRAS1 and FRAS1-related extracellular matrix 2 (FREM2), which encode important ECM proteins, are secreted by mesenchymal cells during diaphragmatic development. The FRAS1/FREM2 gene unit has been shown to form a ternary complex with FREM1, which plays a crucial role during formation of human and rodent diaphragms. Furthermore, it has been demonstrated that the diaphragmatic expression of FREM1 is decreased in the nitrofen-induced CDH model. We hypothesized that FRAS1 and FREM2 expression is decreased in the developing diaphragms of fetal rats with nitrofen-induced CDH. METHODS: Pregnant rats were exposed to either nitrofen or vehicle on gestational day 9 (D9), and fetuses were harvested on D13, D15 and D18. Microdissected diaphragms were divided into nitrofen-exposed/CDH and control samples (n = 12 per time-point and experimental group, respectively). Diaphragmatic gene expression levels of FRAS1 and FREM2 were analyzed by qRT-PCR. Immunofluorescence double staining for FRAS1 and FREM2 was combined with the mesenchymal marker GATA4 in order to evaluate protein expression and localization in pleuroperitoneal folds (PPFs) and fetal diaphragmatic tissue. RESULTS: Relative mRNA expression of FRAS1 and FREM2 were significantly reduced in PPFs of nitrofen-exposed fetuses on D13 (1.76 ± 0.86 vs. 3.09 ± 1.15; p < 0.05 and 0.47 ± 0.26 vs. 0.82 ± 0.36; p < 0.05), developing diaphragms of nitrofen-exposed fetuses on D15 (1.45 ± 0.80 vs. 2.63 ± 0.84; p < 0.05 and 0.41 ± 0.16 vs. 1.02 ± 0.49; p < 0.05) and fully muscularized diaphragms of CDH fetuses on D18 (1.35 ± 0.75 vs. 2.32 ± 0.92; p < 0.05 and 0.37 ± 0.24 vs. 0.70 ± 0.32; p < 0.05) compared to controls. Confocal laser scanning microscopy revealed markedly diminished FRAS1 and FREM2 immunofluorescence in diaphragmatic mesenchyme, which was associated with reduced proliferation of mesenchymal cells in nitrofen-exposed PPFs and fetal CDH diaphragms on D13, D15 and D18 compared to controls. CONCLUSION: Decreased mesenchymal expression of FRAS1 and FREM2 in the nitrofen-induced CDH model may cause failure of the FRAS1/FREM2 gene unit to activate FREM1 signaling, disturbing the formation of diaphragmatic ECM and thus contributing to the development of diaphragmatic defects in CDH.


Assuntos
Diafragma/metabolismo , Desenvolvimento Fetal/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Hérnias Diafragmáticas Congênitas/genética , Hérnias Diafragmáticas Congênitas/metabolismo , Mesoderma/metabolismo , Animais , Diafragma/embriologia , Modelos Animais de Doenças , Expressão Gênica/genética , Éteres Fenílicos , Ratos , Ratos Sprague-Dawley
17.
Pediatr Surg Int ; 32(10): 967-73, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27480986

RESUMO

PURPOSE: Pleuroperitoneal folds (PPFs) are essential for normal diaphragmatic development, representing the only source of the diaphragm's muscle connective tissue. Hepatocyte growth factor (Hgf), which is secreted in PPFs, plays a crucial role in the formation of the muscular diaphragmatic components by regulating the migration of myogenic progenitor cells into the primordial diaphragm. Hgf is also a known downstream target of Gata4 and it has been demonstrated that the expression of Hgf was significantly downregulated in PPF cells of Gata4 knockouts with congenital diaphragmatic hernia (CDH). Furthermore, mutations in PPF-derived cells have been shown to result in CDH. We hypothesized that Hgf expression is decreased in developing diaphragms of fetal rats with nitrofen-induced CDH. METHODS: Timed-pregnant rats were exposed to either nitrofen or vehicle on gestational day 9 (D9). Fetuses were harvested on selected time-points D13, D15 and D18. Dissected diaphragms (n = 72) were divided into control and nitrofen-exposed specimens (n = 12 per time-point and experimental group, respectively). Diaphragmatic gene expression of Hgf was analyzed by qRT-PCR. Immunofluorescence double staining for Hgf and the mesenchymal marker Gata4 or muscular progenitor marker Myogenin was performed to evaluate protein expression and localization in fetal diaphragms. RESULTS: Relative mRNA expression of Hgf was significantly downregulated in PPFs of nitrofen-exposed fetuses on D13 (3.08 ± 1.46 vs. 5.24 ± 1.93; p < 0.05), developing diaphragms of nitrofen-exposed fetuses on D15 (2.01 ± 0.79 vs. 4.10 ± 1.50; p < 0.05) and fully muscularized diaphragms of nitrofen-exposed fetuses on D18 (1.60 ± 0.78 vs. 3.21 ± 1.89; p < 0.05) compared to controls. Confocal laser scanning microscopy revealed markedly diminished diaphragmatic immunofluorescence of Hgf in nitrofen-exposed fetuses on D13, D15 and D18 compared to controls, which was associated with disruptions in muscle connective tissue formation and reduced myogenic progenitor cell invasion. CONCLUSION: Decreased diaphragmatic expression of Hgf may disturb the formation of muscle connective tissue in PPFs and thus prevent essential migration of muscle progenitor cells into the developing diaphragm, leading to diaphragmatic defects in the nitrofen CDH model.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/genética , Fator de Crescimento de Hepatócito/sangue , Fator de Crescimento de Hepatócito/genética , Hérnias Diafragmáticas Congênitas/genética , Hérnias Diafragmáticas Congênitas/metabolismo , Animais , Diafragma/embriologia , Modelos Animais de Doenças , Desenvolvimento Embrionário , Feminino , Feto/metabolismo , Imunofluorescência , Éteres Fenílicos , Gravidez , Ratos , Ratos Sprague-Dawley
18.
Dev Biol ; 394(2): 228-41, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25172430

RESUMO

FgfrL1, which interacts with Fgf ligands and heparin, is a member of the fibroblast growth factor receptor (Fgfr) family. FgfrL1-deficient mice show two significant alterations when compared to wildtype mice: They die at birth due to a malformed diaphragm and they lack metanephric kidneys. Utilizing gene arrays, qPCR and in situ hybridization we show here that the diaphragm of FgfrL1 knockout animals lacks any slow muscle fibers at E18.5 as indicated by the absence of slow fiber markers Myh7, Myl2 and Myl3. Similar lesions are also found in other skeletal muscles that contain a high proportion of slow fibers at birth, such as the extraocular muscles. In contrast to the slow fibers, fast fibers do not appear to be affected as shown by expression of fast fiber markers Myh3, Myh8, Myl1 and MylPF. At early developmental stages (E10.5, E15.5), FgfrL1-deficient animals express slow fiber genes at normal levels. The loss of slow fibers cannot be attributed to the lack of kidneys, since Wnt4 knockout mice, which also lack metanephric kidneys, show normal expression of Myh7, Myl2 and Myl3. Thus, FgfrL1 is specifically required for embryonic development of slow muscle fibers.


Assuntos
Diafragma/embriologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Desenvolvimento Muscular/genética , Fibras Musculares de Contração Lenta/fisiologia , Receptor Tipo 5 de Fator de Crescimento de Fibroblastos/metabolismo , Animais , Northern Blotting , Diafragma/citologia , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Knockout , Análise em Microsséries , Desenvolvimento Muscular/fisiologia , Cadeias Pesadas de Miosina/metabolismo , Cadeias Leves de Miosina/metabolismo , Reação em Cadeia da Polimerase
19.
Am J Physiol Lung Cell Mol Physiol ; 308(2): L147-57, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25416379

RESUMO

Congenital diaphragmatic hernia (CDH) is a common life-threatening congenital anomaly resulting in high rates of perinatal death and neonatal respiratory distress. Some of the nonisolated forms are related to single-gene mutations or genomic rearrangements, but the genetics of the isolated forms (60% of cases) still remains a challenging issue. Retinoid signaling (RA) is critical for both diaphragm and lung development, and it has been hypothesized that subtle disruptions of this pathway could contribute to isolated CDH etiology. Here we used time series of normal and CDH lungs in humans, in nitrofen-exposed rats, and in surgically induced hernia in rabbits to perform a systematic transcriptional analysis of the RA pathway key components. The results point to CRPBP2, CY26B1, and ALDH1A2 as deregulated RA signaling genes in human CDH. Furthermore, the expression profile comparisons suggest that ALDH1A2 overexpression is not a primary event, but rather a consequence of the CDH-induced lung injury. Taken together, these data show that RA signaling disruption is part of CDH pathogenesis, and also that dysregulation of this pathway should be considered organ specifically.


Assuntos
Sistema Enzimático do Citocromo P-450/biossíntese , Diafragma/embriologia , Hérnias Diafragmáticas Congênitas/metabolismo , Retinal Desidrogenase/biossíntese , Vitamina A/metabolismo , Família Aldeído Desidrogenase 1 , Animais , Linhagem Celular , Sistema Enzimático do Citocromo P-450/genética , Diafragma/patologia , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Hérnias Diafragmáticas Congênitas/genética , Hérnias Diafragmáticas Congênitas/patologia , Humanos , Pulmão/embriologia , Masculino , Coelhos , Ratos , Retinal Desidrogenase/genética , Ácido Retinoico 4 Hidroxilase , Proteínas Celulares de Ligação ao Retinol/biossíntese , Proteínas Celulares de Ligação ao Retinol/genética , Transdução de Sinais/genética , Vitamina A/genética
20.
Proc Natl Acad Sci U S A ; 109(8): 2978-83, 2012 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-22315423

RESUMO

Congenital diaphragmatic hernia (CDH) is a common (1 in 3,000 live births) major congenital malformation that results in significant morbidity and mortality. The discovery of CDH loci using standard genetic approaches has been hindered by its genetic heterogeneity. We hypothesized that gene expression profiling of developing embryonic diaphragms would help identify genes likely to be associated with diaphragm defects. We generated a time series of whole-transcriptome expression profiles from laser captured embryonic mouse diaphragms at embryonic day (E)11.5 and E12.5 when experimental perturbations lead to CDH phenotypes, and E16.5 when the diaphragm is fully formed. Gene sets defining biologically relevant pathways and temporal expression trends were identified by using a series of bioinformatic algorithms. These developmental sets were then compared with a manually curated list of genes previously shown to cause diaphragm defects in humans and in mouse models. Our integrative filtering strategy identified 27 candidates for CDH. We examined the diaphragms of knockout mice for one of the candidate genes, pre-B-cell leukemia transcription factor 1 (Pbx1), and identified a range of previously undetected diaphragmatic defects. Our study demonstrates the utility of genetic characterization of normal development as an integral part of a disease gene identification and prioritization strategy for CDH, an approach that can be extended to other diseases and developmental anomalies.


Assuntos
Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Estudos de Associação Genética , Hérnias Diafragmáticas Congênitas , Transcriptoma/genética , Animais , Diafragma/embriologia , Diafragma/metabolismo , Diafragma/patologia , Regulação da Expressão Gênica no Desenvolvimento , Hérnia Diafragmática/genética , Hérnia Diafragmática/patologia , Proteínas de Homeodomínio/metabolismo , Lasers , Mesoderma/embriologia , Mesoderma/metabolismo , Mesoderma/patologia , Camundongos , Camundongos Knockout , Modelos Biológicos , Fator de Transcrição 1 de Leucemia de Células Pré-B , Transdução de Sinais/genética , Fatores de Tempo , Fatores de Transcrição/deficiência , Fatores de Transcrição/metabolismo , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA