Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Genet Med ; 24(11): 2249-2261, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36074124

RESUMO

PURPOSE: The clinical spectrum of motile ciliopathies includes laterality defects, hydrocephalus, and infertility as well as primary ciliary dyskinesia when impaired mucociliary clearance results in otosinopulmonary disease. Importantly, approximately 30% of patients with primary ciliary dyskinesia lack a genetic diagnosis. METHODS: Clinical, genomic, biochemical, and functional studies were performed alongside in vivo modeling of DAW1 variants. RESULTS: In this study, we identified biallelic DAW1 variants associated with laterality defects and respiratory symptoms compatible with motile cilia dysfunction. In early mouse embryos, we showed that Daw1 expression is limited to distal, motile ciliated cells of the node, consistent with a role in left-right patterning. daw1 mutant zebrafish exhibited reduced cilia motility and left-right patterning defects, including cardiac looping abnormalities. Importantly, these defects were rescued by wild-type, but not mutant daw1, gene expression. In addition, pathogenic DAW1 missense variants displayed reduced protein stability, whereas DAW1 loss-of-function was associated with distal type 2 outer dynein arm assembly defects involving axonemal respiratory cilia proteins, explaining the reduced cilia-induced fluid flow in particle tracking velocimetry experiments. CONCLUSION: Our data define biallelic DAW1 variants as a cause of human motile ciliopathy and determine that the disease mechanism involves motile cilia dysfunction, explaining the ciliary beating defects observed in affected individuals.


Assuntos
Transtornos da Motilidade Ciliar , Ciliopatias , Proteínas do Citoesqueleto , Animais , Humanos , Camundongos , Axonema/genética , Cílios/metabolismo , Transtornos da Motilidade Ciliar/genética , Transtornos da Motilidade Ciliar/metabolismo , Transtornos da Motilidade Ciliar/patologia , Ciliopatias/genética , Ciliopatias/metabolismo , Ciliopatias/patologia , Proteínas do Citoesqueleto/genética , Mutação , Proteínas/genética , Peixe-Zebra/genética
2.
Dev Biol ; 395(1): 111-9, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-25173872

RESUMO

The establishment of the coronary circulation is one of the final critical steps during heart development. Despite decades of research, our understanding of how the coronary vasculature develops and connects to the aorta remains limited. This review serves two specific purposes: it addresses recent advances in understanding the origin of the coronary endothelium, and it then focuses on the last crucial step of coronary vasculature development, the connection of the coronary plexus to the aorta. The chick and quail animal models have yielded most of the information for how these connections form, starting with a fine network of vessels that penetrate the aorta and coalesce to form two distinct ostia. Studies in mouse and rat confirm that at least some of these steps are conserved in mammals, but gaps still exist in our understanding of mammalian coronary ostia formation. The signaling cues necessary to guide the coronary plexus to the aorta are also incompletely understood. Hypoxia-inducible transcription factor-1 and its downstream targets are among the few identified genes that promote the formation of the coronary stems. Together, this review summarizes our current knowledge of coronary vascular formation and highlights the significant gaps that remain. In addition, it highlights some of the coronary artery anomalies known to affect human health, demonstrating that even seemingly subtle defects arising from incorrect coronary plexus formation can result in significant health crises.


Assuntos
Vasos Coronários/embriologia , Endotélio Vascular/embriologia , Coração/embriologia , Modelos Anatômicos , Modelos Cardiovasculares , Animais , Vasos Coronários/citologia , Vasos Coronários/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Coração/anatomia & histologia , Humanos , Células-Tronco/citologia , Células-Tronco/metabolismo
3.
Dev Biol ; 386(2): 385-94, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24373957

RESUMO

The connection of the coronary vasculature to the aorta is one of the last essential steps of cardiac development. However, little is known about the signaling events that promote normal coronary artery formation. The bone morphogenetic protein (BMP) signaling pathway regulates multiple aspects of endothelial cell biology but has not been specifically implicated in coronary vascular development. BMP signaling is tightly regulated by numerous factors, including BMP-binding endothelial cell precursor-derived regulator (BMPER), which can both promote and repress BMP signaling activity. In the embryonic heart, BMPER expression is limited to the endothelial cells and the endothelial-derived cushions, suggesting that BMPER may play a role in coronary vascular development. Histological analysis of BMPER(-/-) embryos at early embryonic stages demonstrates that commencement of coronary plexus differentiation is normal and that endothelial apoptosis and cell proliferation are unaffected in BMPER(-/-) embryos compared with wild-type embryos. However, analysis between embryonic days 15.5-17.5 reveals that, in BMPER(-/-) embryos, coronary arteries are either atretic or connected distal to the semilunar valves. In vitro tubulogenesis assays indicate that isolated BMPER(-/-) endothelial cells have impaired tube formation and migratory ability compared with wild-type endothelial cells, suggesting that these defects may lead to the observed coronary artery anomalies seen in BMPER(-/-) embryos. Additionally, recombinant BMPER promotes wild-type ventricular endothelial migration in a dose-dependent manner, with a low concentration promoting and high concentrations inhibiting migration. Together, these results indicate that BMPER-regulated BMP signaling is critical for coronary plexus remodeling and normal coronary artery development.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Transporte/metabolismo , Vasos Coronários/fisiologia , Células Endoteliais/metabolismo , Transdução de Sinais/fisiologia , Análise de Variância , Animais , Proteínas de Transporte/genética , Immunoblotting , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Knockout , Microscopia de Fluorescência
4.
Circ Res ; 111(5): 564-74, 2012 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-22777006

RESUMO

RATIONALE: Among the extracellular modulators of Bmp (bone morphogenetic protein) signaling, Bmper (Bmp endothelial cell precursor-derived regulator) both enhances and inhibits Bmp signaling. Recently we found that Bmper modulates Bmp4 activity via a concentration-dependent, endocytic trap-and-sink mechanism. OBJECTIVE: To investigate the molecular mechanisms required for endocytosis of the Bmper/Bmp4 and signaling complex and determine the mechanism of Bmper's differential effects on Bmp4 signaling. METHODS AND RESULTS: Using an array of biochemical and cell biology techniques, we report that LRP1 (LDL receptor-related protein 1), a member of the LDL receptor family, acts as an endocytic receptor for Bmper and a coreceptor of Bmp4 to mediate the endocytosis of the Bmper/Bmp4 signaling complex. Furthermore, we demonstrate that LRP1-dependent Bmper/Bmp4 endocytosis is essential for Bmp4 signaling, as evidenced by the phenotype of lrp1-deficient zebrafish, which have abnormal cardiovascular development and decreased Smad1/5/8 activity in key vasculogenic structures. CONCLUSIONS: Together, these data reveal a novel role for LRP1 in the regulation of Bmp4 signaling by regulating receptor complex endocytosis. In addition, these data introduce LRP1 as a critical regulator of vascular development. These observations demonstrate Bmper's ability to fine-tune Bmp4 signaling at the single-cell level, unlike the spatial regulatory mechanisms applied by other Bmp modulators.


Assuntos
Proteína Morfogenética Óssea 4/metabolismo , Proteínas de Transporte/metabolismo , Endocitose/fisiologia , Células Endoteliais/fisiologia , Neovascularização Fisiológica/fisiologia , Receptores de LDL/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Proteína Morfogenética Óssea 4/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Proteínas de Transporte/genética , Linhagem Celular , Movimento Celular/fisiologia , Células Endoteliais/citologia , Células HEK293 , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Camundongos , Fenótipo , RNA Interferente Pequeno/genética , Receptores de LDL/genética , Transdução de Sinais/fisiologia , Proteínas Supressoras de Tumor/genética , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
5.
Cells ; 13(2)2024 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-38247840

RESUMO

Besides visceral heterotaxia, Pkd1l1 null mouse embryos exhibit general edema and perinatal lethality. In humans, congenital chylothorax (CCT) is a frequent cause of fetal hydrops. In 2021, Correa and colleagues reported ultrarare compound heterozygous variants in PKD1L1 exhibiting in two consecutive fetuses with severe hydrops, implicating a direct role of PKD1L1 in fetal hydrops formation. Here, we performed an exome survey and identified ultrarare compound heterozygous variants in PKD1L1 in two of the five case-parent trios with CCT. In one family, the affected carried the ultrarare missense variants c.1543G>A(p.Gly515Arg) and c.3845T>A(p.Val1282Glu). In the other family, the affected carried the ultrarare loss-of-function variant (LoF) c.863delA(p.Asn288Thrfs*3) and the ultrarare missense variant c.6549G>T(p.Gln2183His). Investigation of the variants' impact on PKD1L1 protein localization suggests the missense variants cause protein dysfunction and the LoF variant causes protein mislocalization. Further analysis of Pkd1l1 mutant mouse embryos revealed about 20% of Pkd1l1-/- embryos display general edema and pleural effusion at 14.5 dpc. Immunofluorescence staining at 14.5 dpc in Pkd1l1-/- embryos displayed both normal and massively altered lymphatic vessel morphologies. Together, our studies suggest the implication of PKD1L1 in congenital lymphatic anomalies, including CCTs.


Assuntos
Quilotórax , Animais , Feminino , Humanos , Camundongos , Gravidez , Quilotórax/genética , Feto , Doenças Genéticas Ligadas ao Cromossomo X , Hidropisia Fetal , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos Knockout
6.
Arterioscler Thromb Vasc Biol ; 32(9): 2214-22, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22772758

RESUMO

OBJECTIVE: Bone morphogenetic proteins (Bmps) are important mediators of inflammation and atherosclerosis, though their mechanism of action is not fully understood. To better understand the contribution of the Bmp signaling pathway in vascular inflammation, we investigated the role of Bmper (Bmp endothelial cell precursor-derived regulator), an extracellular Bmp modulator, in an induced in vivo model of inflammation and atherosclerosis. METHODS AND RESULTS: We crossed apolipoprotein E-deficient (ApoE(-/-)) mice with mice missing 1 allele of Bmper (Bmper(+/-) mice used in the place of Bmper(-/-) mice that die at birth) and measured the development of atherosclerosis in mice fed a high-fat diet. Bmper haploinsufficiency in ApoE(-/-) mice (Bmper(+/-);ApoE(-/-) mice) led to a more severe phenotype compared with Bmper(+/+);ApoE(-/-) mice. Bmper(+/-);ApoE(-/-) mice also exhibited increased Bmp activity in the endothelial cells in both the greater and lesser curvatures of the aortic arch, suggesting a role for Bmper in regulating Bmp-mediated inflammation associated with laminar and oscillatory shear stress. Small interfering RNA knockdown of Bmper in human umbilical vein endothelial cells caused a dramatic increase in the inflammatory markers intracellular adhesion molecule 1 and vascular cell adhesion molecule 1 at rest and after exposure to oscillatory and laminar shear stress. CONCLUSIONS: We conclude that Bmper is a critical regulator of Bmp-mediated vascular inflammation and that the fine-tuning of Bmp and Bmper levels is essential in the maintenance of normal vascular homeostasis.


Assuntos
Doenças da Aorta/prevenção & controle , Aterosclerose/prevenção & controle , Proteínas de Transporte/metabolismo , Moléculas de Adesão Celular/metabolismo , Células Endoteliais/metabolismo , Mediadores da Inflamação/metabolismo , Inflamação/prevenção & controle , Animais , Doenças da Aorta/genética , Doenças da Aorta/imunologia , Doenças da Aorta/metabolismo , Doenças da Aorta/patologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/imunologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Proteína Morfogenética Óssea 4/metabolismo , Proteínas de Transporte/genética , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/imunologia , Células Endoteliais/patologia , Genótipo , Células Endoteliais da Veia Umbilical Humana/imunologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Inflamação/genética , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Molécula 1 de Adesão Intercelular/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Interferência de RNA , Proteínas Recombinantes/metabolismo , Estresse Mecânico , Fatores de Tempo , Transfecção , Calcificação Vascular/imunologia , Calcificação Vascular/metabolismo , Calcificação Vascular/prevenção & controle , Molécula 1 de Adesão de Célula Vascular/metabolismo
7.
Molecules ; 18(5): 5594-610, 2013 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-23676470

RESUMO

Angiogenesis plays a key role in cancer progression and correlates with disease aggressiveness and poor clinical outcomes. Affinity ligands discovered by screening phage display random peptide libraries can be engineered to molecularly target tumor blood vessels for noninvasive imaging and early detection of tumor aggressiveness. In this study, we tested the ability of a phage-display-selected peptide sequence recognizing specifically bone marrow- derived pro-angiogenic tumor-homing cells, the QFP-peptide, radiolabeled with 64Cu radioisotope to selectively image tumor vasculature in vivo by positron emission tomography (PET). To prepare the targeted PET tracer we modified QFP-phage with the DOTA chelator and radiolabeled the purified QFP-phage-DOTA intermediate with 64Cu to obtain QFP-targeted radioconjugate with high radiopharmaceutical yield and specific activity. We evaluated the new PET tracer in vivo in a subcutaneous (s.c.) Lewis lung carcinoma (LLC) mouse model and conducted tissue distribution, small animal PET/CT imaging study, autoradiography, histology, fluorescence imaging, and dosimetry assessments. The results from this study show that, in the context of the s.c. LLC immunocompetent mouse model, the QFP-tracer can target tumor blood vessels selectively. However, further optimization of the biodistribution and dosimetry profile of the tracer is necessary to ensure efficient radiopharmaceutical applications enabled by the biological specificity of the QFP-peptide.


Assuntos
Carcinoma Pulmonar de Lewis , Neovascularização Patológica , Peptídeos , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos , Animais , Carcinoma Pulmonar de Lewis/diagnóstico por imagem , Carcinoma Pulmonar de Lewis/metabolismo , Cobre/química , Feminino , Isótopos/química , Camundongos , Neovascularização Patológica/diagnóstico por imagem , Neovascularização Patológica/metabolismo , Peptídeos/síntese química , Peptídeos/química , Peptídeos/farmacologia , Radiografia , Compostos Radiofarmacêuticos/síntese química , Compostos Radiofarmacêuticos/química , Compostos Radiofarmacêuticos/farmacologia
8.
Birth Defects Res ; 115(4): 474-487, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36515170

RESUMO

BACKGROUND: Infants with fetal alcohol syndrome exhibit a range of developmental anomalies, many related to the heart (e.g., decreased heart rate variability). However, the baseline heart rate in this population remains unclear. We hypothesized that the age at which heart rate was measured or the age during exposure to alcohol affects the baseline heart rate. METHODS: First, we conducted a systemic review to determine the published heart rate of infants with prenatal alcohol exposure (PAE). Exclusion criteria included potentially confounding factors, including the commonly associated phenotypes of small for gestational age and premature birth. Risk of bias was evaluated based on case study limitations, and data were compared with established heart rate norms. Then, we evaluated the precise age at heart rate measurement using existing datasets from the Collaborative Initiative on Fetal Alcohol Spectrum Disorders and the Maternal Lifestyle Study. RESULTS: Based on the weighted means of six studies, the baseline heart rate was 4.6 bpm higher in infants with PAE (n = 253) than in control infants (n = 152). Using the individual patient data, baseline heart rates were similar between age-matched infants with PAE and control infants who were born full-term and showed no signs of growth restriction (ANOVA, p > .05; n = 49-124 infants per age and exposure). CONCLUSIONS: A systematic literature review suggested that heart rate is elevated in infants with PAE, but these findings are limited by the number of studies and how few studies included control infants. The analysis of individual patient data indicates that infants with PAE have normal baseline heart rates. This knowledge may help clinicians detect changes in cardiac function in infants with PAE. (Registered via PROSPERO, #CRD42020191212.).


Assuntos
Transtornos do Espectro Alcoólico Fetal , Efeitos Tardios da Exposição Pré-Natal , Humanos , Gravidez , Feminino , Frequência Cardíaca , Etanol , Transtornos do Espectro Alcoólico Fetal/diagnóstico , Consumo de Bebidas Alcoólicas/efeitos adversos , Consumo de Bebidas Alcoólicas/epidemiologia
9.
Curr Protoc ; 2(7): e448, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35838628

RESUMO

In this paper, we review lightsheet (selective plane illumination) microscopy for mouse developmental biologists. There are different means of forming the illumination sheet, and we discuss these. We explain how we introduced the lightsheet microscope economically into our core facility and present our results on fixed and living samples. We also describe methods of clearing fixed samples for three-dimensional imaging and discuss the various means of preparing samples with particular reference to mouse cilia, adipose spheroids, and cochleae. © 2022 The Authors. Current Protocols published by Wiley Periodicals LLC.


Assuntos
Imageamento Tridimensional , Iluminação , Animais , Imageamento Tridimensional/métodos , Iluminação/métodos , Camundongos , Microscopia de Fluorescência/métodos
10.
Dev Biol ; 348(2): 167-76, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-20920499

RESUMO

Sonic hedgehog signaling in the secondary heart field has a clear role in cardiac arterial pole development. In the absence of hedgehog signaling, proliferation is reduced in secondary heart field progenitors, and embryos predominantly develop pulmonary atresia. While it is expected that proliferation in the secondary heart field would be increased with elevated hedgehog signaling, this idea has never been tested. We hypothesized that up-regulating hedgehog signaling would increase secondary heart field proliferation, which would lead to arterial pole defects. In culture, secondary heart field explants proliferated up to 6-fold more in response to the hedgehog signaling agonist SAG, while myocardial differentiation and migration were unaffected. Treatment of chick embryos with SAG at HH14, just before the peak in secondary heart field proliferation, resulted unexpectedly in stenosis of both the aortic and pulmonary outlets. We examined proliferation in the secondary heart field and found that SAG-treated embryos exhibited a much milder increase in proliferation than was indicated by the in vitro experiments. To determine the source of other signaling factors that could modulate increased hedgehog signaling, we co-cultured secondary heart field explants with isolated pharyngeal endoderm or outflow tract and found that outflow tract co-cultures prevented SAG-induced proliferation. BMP2 is made and secreted by the outflow tract myocardium. To determine whether BMP signaling could prevent SAG-induced proliferation, we treated explants with SAG and BMP2 and found that BMP2 inhibited SAG-induced proliferation. In vivo, SAG-treated embryos showed up-regulated BMP2 expression and signaling. Together, these results indicate that BMP signaling from the outflow tract modulates hedgehog-induced proliferation in the secondary heart field.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Coração/embriologia , Proteínas Hedgehog/metabolismo , Transdução de Sinais , Animais , Proteínas Morfogenéticas Ósseas/genética , Proliferação de Células , Embrião de Galinha , Cicloexilaminas/farmacologia , Proteínas Hedgehog/genética , Miocárdio/metabolismo , Organogênese , Tiofenos/farmacologia , Regulação para Cima
11.
J Cardiovasc Dev Dis ; 8(8)2021 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-34436232

RESUMO

In congenital heart disease, the presence of structural defects affects blood flow in the heart and circulation. However, because the fetal circulation bypasses the lungs, fetuses with cyanotic heart defects can survive in utero but need prompt intervention to survive after birth. Tetralogy of Fallot and persistent truncus arteriosus are two of the most significant conotruncal heart defects. In both defects, blood access to the lungs is restricted or non-existent, and babies with these critical conditions need intervention right after birth. While there are known genetic mutations that lead to these critical heart defects, early perturbations in blood flow can independently lead to critical heart defects. In this paper, we start by comparing the fetal circulation with the neonatal and adult circulation, and reviewing how altered fetal blood flow can be used as a diagnostic tool to plan interventions. We then look at known factors that lead to tetralogy of Fallot and persistent truncus arteriosus: namely early perturbations in blood flow and mutations within VEGF-related pathways. The interplay between physical and genetic factors means that any one alteration can cause significant disruptions during development and underscore our need to better understand the effects of both blood flow and flow-responsive genes.

13.
Dev Biol ; 330(2): 305-17, 2009 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-19361493

RESUMO

The Sonic hedgehog (Shh)-null mouse was initially described as a phenotypic mimic of Tetralogy of Fallot with pulmonary atresia (Washington Smoak, I., Byrd, N.A., Abu-Issa, R., Goddeeris, M.M., Anderson, R., Morris, J., Yamamura, K., Klingensmith, J., and Meyers, E.N. 2005. Sonic hedgehog is required for cardiac outflow tract and neural crest cell development. Dev. Biol. 283, 357-372.); however, subsequent reports describe only a single outflow tract, leaving the phenotype and its developmental mechanism unclear. We hypothesized that the phenotype that occurs in response to Shh knockdown is pulmonary atresia and is directly related to the abnormal development of the secondary heart field. We found that Shh was expressed by the pharyngeal endoderm adjacent to the secondary heart field and that its receptor Ptc2 was expressed in a gradient in the secondary heart field, with the most robust expression in the caudal secondary heart field, closest to the Shh expression. In vitro culture of secondary heart field with the hedgehog inhibitor cyclopamine significantly reduced proliferation. In ovo, cyclopamine treatment before the secondary heart field adds to the outflow tract reduced proliferation only in the caudal secondary heart field, which coincided with the region of high Ptc2 expression. After outflow tract septation should occur, embryos treated with cyclopamine exhibited pulmonary atresia, pulmonary stenosis, and persistent truncus arteriosus. In hearts with pulmonary atresia, cardiac neural crest-derived cells, which form the outflow tract septum, migrated into the outflow tract and formed a septum. However, this septum divided the outflow tract into two unequal sized vessels and effectively closed off the pulmonary outlet. These experiments show that Shh is necessary for secondary heart field proliferation, which is required for normal pulmonary trunk formation, and that embryos with pulmonary atresia have an outflow tract septum.


Assuntos
Proliferação de Células , Coração/embriologia , Proteínas Hedgehog/fisiologia , Miocárdio/citologia , Animais , Sequência de Bases , Primers do DNA , Coração/efeitos dos fármacos , Proteínas Hedgehog/genética , Hibridização In Situ , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Alcaloides de Veratrum/farmacologia
14.
Dev Biol ; 336(2): 137-44, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19835857

RESUMO

Although de la Cruz and colleagues showed as early as 1977 that the outflow tract was added after the heart tube formed, the source of these secondarily added cells was not identified for nearly 25 years. In 2001, three pivotal publications described a secondary or anterior heart field that contributed to the developing outflow tract. This review details the history of the heart field, the discovery and continuing elucidation of the secondarily adding myocardial cells, and how the different populations identified in 2001 are related to the more recent lineage tracing studies that defined the first and second myocardial heart fields/lineages. Much recent work has focused on secondary heart field progenitors that give rise to the myocardium and smooth muscle at the definitive arterial pole. These progenitors are the last to be added to the arterial pole and are particularly susceptible to abnormal development, leading to conotruncal malformations in children. The major signaling pathways (Wnt, BMP, FGF8, Notch, and Shh) that control various aspects of secondary heart field progenitor behavior are discussed.


Assuntos
Coração/embriologia , Animais , Humanos , Transdução de Sinais
15.
Semin Thromb Hemost ; 36(3): 227-35, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20490975

RESUMO

The endothelium is composed of specialized epithelial cells that line the vasculature, the lymph vessels, and the heart. These endothelial cells are characterized by their stratification and are connected via intercellular junctions that confer specific permeability. Although all endothelium acts as a barrier, considerable heterogeneity exists among different organs and even within vessels. During development, the endothelial cells are specified before they migrate to their final destination, and then they commit to an arterial or venous fate. From the venous endothelial cell population, a subset of cells is further specified as lymphatic endothelium. The endothelium can be highly permeable, as in the lymph vessels, or impenetrable, as in the blood-brain barrier. These differences arise during development and are orchestrated through a series of signaling pathways. This review details how endothelial cells arise and are directed to their specific fate, specifically targeting what differentiates endothelial populations.


Assuntos
Endotélio/crescimento & desenvolvimento , Diferenciação Celular , Endotélio/citologia , Endotélio/embriologia , Endotélio/fisiologia , Endotélio/fisiopatologia , Permeabilidade , Transdução de Sinais
16.
PLoS One ; 13(11): e0207504, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30496196

RESUMO

[This corrects the article DOI: 10.1371/journal.pone.0139209.].

17.
Nat Commun ; 8: 14279, 2017 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-28176794

RESUMO

By moving essential body fluids and molecules, motile cilia and flagella govern respiratory mucociliary clearance, laterality determination and the transport of gametes and cerebrospinal fluid. Primary ciliary dyskinesia (PCD) is an autosomal recessive disorder frequently caused by non-assembly of dynein arm motors into cilia and flagella axonemes. Before their import into cilia and flagella, multi-subunit axonemal dynein arms are thought to be stabilized and pre-assembled in the cytoplasm through a DNAAF2-DNAAF4-HSP90 complex akin to the HSP90 co-chaperone R2TP complex. Here, we demonstrate that large genomic deletions as well as point mutations involving PIH1D3 are responsible for an X-linked form of PCD causing disruption of early axonemal dynein assembly. We propose that PIH1D3, a protein that emerges as a new player of the cytoplasmic pre-assembly pathway, is part of a complementary conserved R2TP-like HSP90 co-chaperone complex, the loss of which affects assembly of a subset of inner arm dyneins.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Dineínas do Axonema/metabolismo , Genes Ligados ao Cromossomo X/genética , Doenças Genéticas Ligadas ao Cromossomo X/genética , Síndrome de Kartagener/genética , Proteínas dos Microtúbulos/genética , Chaperonas Moleculares/genética , Adolescente , Adulto , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Axonema/patologia , Criança , Pré-Escolar , Cílios/patologia , Cílios/ultraestrutura , Citoplasma/patologia , Modelos Animais de Doenças , Feminino , Doenças Genéticas Ligadas ao Cromossomo X/patologia , Células HEK293 , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Recém-Nascido , Peptídeos e Proteínas de Sinalização Intracelular , Síndrome de Kartagener/patologia , Masculino , Microscopia Eletrônica de Transmissão , Linhagem , Filogenia , Mutação Puntual , Dobramento de Proteína , Alinhamento de Sequência , Deleção de Sequência , Motilidade dos Espermatozoides/genética , Sequenciamento do Exoma , Peixe-Zebra
18.
PLoS One ; 10(9): e0139209, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26418455

RESUMO

Formation of the cardiac valves is an essential component of cardiovascular development. Consistent with the role of the bone morphogenetic protein (BMP) signaling pathway in cardiac valve formation, embryos that are deficient for the BMP regulator BMPER (BMP-binding endothelial regulator) display the cardiac valve anomaly mitral valve prolapse. However, how BMPER deficiency leads to this defect is unknown. Based on its expression pattern in the developing cardiac cushions, we hypothesized that BMPER regulates BMP2-mediated signaling, leading to fine-tuned epithelial-mesenchymal transition (EMT) and extracellular matrix deposition. In the BMPER-/- embryo, EMT is dysregulated in the atrioventricular and outflow tract cushions compared with their wild-type counterparts, as indicated by a significant increase of Sox9-positive cells during cushion formation. However, proliferation is not impaired in the developing BMPER-/- valves. In vitro data show that BMPER directly binds BMP2. In cultured endothelial cells, BMPER blocks BMP2-induced Smad activation in a dose-dependent manner. In addition, BMP2 increases the Sox9 protein level, and this increase is inhibited by co-treatment with BMPER. Consistently, in the BMPER-/- embryos, semi-quantitative analysis of Smad activation shows that the canonical BMP pathway is significantly more active in the atrioventricular cushions during EMT. These results indicate that BMPER negatively regulates BMP-induced Smad and Sox9 activity during valve development. Together, these results identify BMPER as a regulator of BMP2-induced cardiac valve development and will contribute to our understanding of valvular defects.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Proteínas de Transporte/metabolismo , Transição Epitelial-Mesenquimal/fisiologia , Valvas Cardíacas/embriologia , Fatores de Transcrição SOX9/metabolismo , Animais , Proteínas de Transporte/biossíntese , Linhagem Celular , Proliferação de Células , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica , Transdução de Sinais
19.
Trends Endocrinol Metab ; 25(9): 472-80, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24908616

RESUMO

The bone morphogenetic protein (BMP) family of proteins has a multitude of roles throughout the body. In embryonic development, BMPs promote endothelial specification and subsequent venous differentiation. The BMP pathway also plays important roles in the adult vascular endothelium, promoting angiogenesis and mediating shear and oxidative stress. The canonical BMP pathway functions through the Smad transcription factors; however, other intracellular signaling cascades can be activated, and receptor complexes beyond the traditional type I and type II receptors add additional layers of regulation. Dysregulated BMP signaling has been linked to vascular diseases including pulmonary hypertension and atherosclerosis. This review addresses recent advances in the roles of BMP signaling in the endothelium and how BMPs affect endothelial dysfunction and human disease.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Morfogenéticas Ósseas/metabolismo , Endotélio Vascular/metabolismo , Modelos Biológicos , Transdução de Sinais , Doenças Vasculares/metabolismo , Animais , Aterosclerose/etiologia , Aterosclerose/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas/agonistas , Receptores de Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/genética , Endotélio Vascular/citologia , Humanos , Hipertensão/metabolismo , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/metabolismo , Camundongos Transgênicos , Neovascularização Patológica/etiologia , Neovascularização Patológica/metabolismo , Neovascularização Fisiológica , Estresse Oxidativo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Resistência ao Cisalhamento , Estresse Fisiológico , Doenças Vasculares/etiologia
20.
J Vis Exp ; (91): 51911, 2014 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-25285454

RESUMO

Western blot analysis is a commonly employed technique for detecting and quantifying protein levels. However, for small tissue samples, this analysis method may not be sufficiently sensitive to detect a protein of interest. To overcome these difficulties, we examined protocols for obtaining protein from adult human cardiac valves and modified these protocols for the developing early embryonic mouse counterparts. In brief, the mouse embryonic aortic valve regions, including the aortic valve and surrounding aortic wall, are collected in the minimal possible volume of a Tris-based lysis buffer with protease inhibitors. If required based on the breeding strategy, embryos are genotyped prior to pooling four embryonic aortic valve regions for homogenization. After homogenization, an SDS-based sample buffer is used to denature the sample for running on an SDS-PAGE gel and subsequent western blot analysis. Although the protein concentration remains too low to quantify using spectrophotometric protein quantification assays and have sample remaining for subsequent analyses, this technique can be used to successfully detect and semi-quantify phosphorylated proteins via western blot from pooled samples of four embryonic day 13.5 mouse aortic valve regions, each of which yields approximately 1 µg of protein. This technique will be of benefit for studying cell signaling pathway activation and protein expression levels during early embryonic mouse valve development.


Assuntos
Valvas Cardíacas/química , Proteínas Musculares/análise , Animais , Western Blotting , Eletroforese em Gel de Poliacrilamida , Feminino , Valvas Cardíacas/embriologia , Valvas Cardíacas/metabolismo , Camundongos , Proteínas Musculares/metabolismo , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA