Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Cells ; 13(2)2024 01 12.
Article in English | MEDLINE | ID: mdl-38247840

ABSTRACT

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.


Subject(s)
Chylothorax , Animals , Female , Humans , Mice , Pregnancy , Chylothorax/genetics , Fetus , Genetic Diseases, X-Linked , Hydrops Fetalis , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Knockout
2.
Birth Defects Res ; 115(4): 474-487, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36515170

ABSTRACT

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.).


Subject(s)
Fetal Alcohol Spectrum Disorders , Prenatal Exposure Delayed Effects , Humans , Pregnancy , Female , Heart Rate , Ethanol , Fetal Alcohol Spectrum Disorders/diagnosis , Alcohol Drinking/adverse effects , Alcohol Drinking/epidemiology
3.
Genet Med ; 24(11): 2249-2261, 2022 11.
Article in English | MEDLINE | ID: mdl-36074124

ABSTRACT

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.


Subject(s)
Ciliary Motility Disorders , Ciliopathies , Cytoskeletal Proteins , Animals , Humans , Mice , Axoneme/genetics , Cilia/metabolism , Ciliary Motility Disorders/genetics , Ciliary Motility Disorders/metabolism , Ciliary Motility Disorders/pathology , Ciliopathies/genetics , Ciliopathies/metabolism , Ciliopathies/pathology , Cytoskeletal Proteins/genetics , Mutation , Proteins/genetics , Zebrafish/genetics
4.
Curr Protoc ; 2(7): e448, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35838628

ABSTRACT

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.


Subject(s)
Imaging, Three-Dimensional , Lighting , Animals , Imaging, Three-Dimensional/methods , Lighting/methods , Mice , Microscopy, Fluorescence/methods
5.
J Cardiovasc Dev Dis ; 8(8)2021 Jul 30.
Article in English | MEDLINE | ID: mdl-34436232

ABSTRACT

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.

6.
PLoS One ; 13(11): e0207504, 2018.
Article in English | MEDLINE | ID: mdl-30496196

ABSTRACT

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

7.
Nat Commun ; 8: 14279, 2017 02 08.
Article in English | MEDLINE | ID: mdl-28176794

ABSTRACT

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.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Axonemal Dyneins/metabolism , Genes, X-Linked/genetics , Genetic Diseases, X-Linked/genetics , Kartagener Syndrome/genetics , Microtubule Proteins/genetics , Molecular Chaperones/genetics , Adolescent , Adult , Animals , Apoptosis Regulatory Proteins/metabolism , Axoneme/pathology , Child , Child, Preschool , Cilia/pathology , Cilia/ultrastructure , Cytoplasm/pathology , Disease Models, Animal , Female , Genetic Diseases, X-Linked/pathology , HEK293 Cells , HSP90 Heat-Shock Proteins/metabolism , Humans , Infant, Newborn , Intracellular Signaling Peptides and Proteins , Kartagener Syndrome/pathology , Male , Microscopy, Electron, Transmission , Pedigree , Phylogeny , Point Mutation , Protein Folding , Sequence Alignment , Sequence Deletion , Sperm Motility/genetics , Exome Sequencing , Zebrafish
8.
PLoS One ; 10(9): e0139209, 2015.
Article in English | MEDLINE | ID: mdl-26418455

ABSTRACT

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.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Carrier Proteins/metabolism , Epithelial-Mesenchymal Transition/physiology , Heart Valves/embryology , SOX9 Transcription Factor/metabolism , Animals , Carrier Proteins/biosynthesis , Cell Line , Cell Proliferation , Mice , Mice, Inbred C57BL , Protein Binding , Signal Transduction
10.
J Vis Exp ; (91): 51911, 2014 Sep 23.
Article in English | MEDLINE | ID: mdl-25285454

ABSTRACT

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.


Subject(s)
Heart Valves/chemistry , Muscle Proteins/analysis , Animals , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Female , Heart Valves/embryology , Heart Valves/metabolism , Mice , Muscle Proteins/metabolism , Pregnancy
11.
Dev Biol ; 395(1): 111-9, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25173872

ABSTRACT

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.


Subject(s)
Coronary Vessels/embryology , Endothelium, Vascular/embryology , Heart/embryology , Models, Anatomic , Models, Cardiovascular , Animals , Coronary Vessels/cytology , Coronary Vessels/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Gene Expression Regulation, Developmental , Heart/anatomy & histology , Humans , Stem Cells/cytology , Stem Cells/metabolism
12.
Trends Endocrinol Metab ; 25(9): 472-80, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24908616

ABSTRACT

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.


Subject(s)
Bone Morphogenetic Protein Receptors/metabolism , Bone Morphogenetic Proteins/metabolism , Endothelium, Vascular/metabolism , Models, Biological , Signal Transduction , Vascular Diseases/metabolism , Animals , Atherosclerosis/etiology , Atherosclerosis/metabolism , Bone Morphogenetic Protein Receptors/agonists , Bone Morphogenetic Protein Receptors/genetics , Bone Morphogenetic Proteins/genetics , Endothelium, Vascular/cytology , Humans , Hypertension/metabolism , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/metabolism , Mice, Transgenic , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic , Oxidative Stress , Protein Isoforms/genetics , Protein Isoforms/metabolism , Shear Strength , Stress, Physiological , Vascular Diseases/etiology
13.
Dev Biol ; 386(2): 385-94, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24373957

ABSTRACT

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.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Carrier Proteins/metabolism , Coronary Vessels/physiology , Endothelial Cells/metabolism , Signal Transduction/physiology , Analysis of Variance , Animals , Carrier Proteins/genetics , Immunoblotting , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Knockout , Microscopy, Fluorescence
14.
J Vis Exp ; (77)2013 Jul 20.
Article in English | MEDLINE | ID: mdl-23912902

ABSTRACT

Cell culture has greatly enhanced our ability to assess individual populations of cells under myriad culture conditions. While immortalized cell lines offer significant advantages for their ease of use, these cell lines are unavailable for all potential cell types. By isolating primary cells from a specific region of interest, particularly from a transgenic mouse, more nuanced studies can be performed. The basic technique involves dissecting the organ or partial organ of interest (e.g. the heart or a specific region of the heart) and dissociating the organ to single cells. These cells are then incubated with magnetic beads conjugated to an antibody that recognizes the cell type of interest. The cells of interest can then be isolated with the use of a magnet, with a short trypsin incubation dissociating the cells from the beads. These isolated cells can then be cultured and analyzed as desired. This technique was originally designed for adult mouse organs but can be easily scaled down for use with embryonic organs, as demonstrated herein. Because our interest is in the developing coronary vasculature, we wanted to study this population of cells during specific embryonic stages. Thus, the original protocol had to be modified to be compatible with the small size of the embryonic ventricles and the low potential yield of endothelial cells at these developmental stages. Utilizing this scaled-down approach, we have assessed coronary plexus remodeling in transgenic embryonic ventricular endothelial cells.


Subject(s)
Endothelial Cells/cytology , Heart/embryology , Immunomagnetic Separation/methods , Myocardium/cytology , Animals , Female , Heart Ventricles/embryology , Mice , Mice, Transgenic , Pregnancy
15.
J Vis Exp ; (75): e50359, 2013 May 24.
Article in English | MEDLINE | ID: mdl-23728379

ABSTRACT

Developmental studies in the mouse are hampered by the inaccessibility of the embryo during gestation. Thus, protocols to isolate and culture individual organs of interest are essential to provide a method of both visualizing changes in development and allowing novel treatment strategies. To promote the long-term culture of the embryonic heart at late stages of gestation, we developed a protocol in which the excised heart is cultured in a semi-solid, dilute Matrigel. This substrate provides enough support to maintain the three-dimensional structure but is flexible enough to allow continued contraction. In brief, hearts are excised from the embryo and placed in a mixture of cold Matrigel diluted 1:1 with growth medium. After the diluted Matrigel solidifies, growth medium is added to the culture dish. Hearts excised as late as embryonic day 16.5 were viable for four days post-dissection. Analysis of the coronary plexus shows that this method does not disrupt coronary vascular development. Thus, we present a novel method for long-term culture of embryonic hearts.


Subject(s)
Embryo Culture Techniques/methods , Heart/embryology , Mice/embryology , Animals , Collagen , Drug Combinations , Embryo, Mammalian , Female , Laminin , Pregnancy , Proteoglycans
16.
Molecules ; 18(5): 5594-610, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23676470

ABSTRACT

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.


Subject(s)
Carcinoma, Lewis Lung , Neovascularization, Pathologic , Peptides , Positron-Emission Tomography , Radiopharmaceuticals , Animals , Carcinoma, Lewis Lung/diagnostic imaging , Carcinoma, Lewis Lung/metabolism , Copper/chemistry , Female , Isotopes/chemistry , Mice , Neovascularization, Pathologic/diagnostic imaging , Neovascularization, Pathologic/metabolism , Peptides/chemical synthesis , Peptides/chemistry , Peptides/pharmacology , Radiography , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacology
17.
Cardiovasc Pathol ; 22(3): 228-40, 2013.
Article in English | MEDLINE | ID: mdl-23200275

ABSTRACT

BACKGROUND: BMPER, an orthologue of Drosophila melanogaster Crossveinless-2, is a secreted factor that regulates bone morphogenetic protein activity in endothelial cell precursors and during early cardiomyocyte differentiation. Although previously described in the heart, the role of BMPER in cardiac development and function remain unknown. METHODS: BMPER-deficient hearts were phenotyped histologically and functionally using echocardiography and Doppler analysis. Since BMPER -/- mice die perinatally, adult BMPER +/- mice were challenged to pressure-overload-induced cardiac hypertrophy and hindlimb ischemia to determine changes in angiogenesis and regulation of cardiomyocyte size. RESULTS: We identify for the first time the cardiac phenotype associated with BMPER haploinsufficiency. BMPER messenger RNA and protein are present in the heart during cardiac development through at least E14.5 but is lost by E18.5. BMPER +/- ventricles are thinner and less compact than sibling wild-type hearts. In the adult, BMPER +/- hearts present with decreased anterior and posterior wall thickness, decreased cardiomyocyte size and an increase in cardiac vessel density. Despite these changes, BMPER +/- mice respond to pressure-overload-induced cardiac hypertrophy challenge largely to the same extent as wild-type mice. CONCLUSION: BMPER appears to play a role in regulating both vessel density and cardiac development in vivo; however, BMPER haploinsufficiency does not result in marked effects on cardiac function or adaptation to pressure overload hypertrophy.


Subject(s)
Carrier Proteins/metabolism , Heart/growth & development , Myocytes, Cardiac/cytology , Neovascularization, Physiologic/physiology , Animals , Blotting, Western , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cell Enlargement , Coronary Vessels/metabolism , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/metabolism , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction
18.
Circ Res ; 111(5): 564-74, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22777006

ABSTRACT

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.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Carrier Proteins/metabolism , Endocytosis/physiology , Endothelial Cells/physiology , Neovascularization, Physiologic/physiology , Receptors, LDL/metabolism , Tumor Suppressor Proteins/metabolism , Zebrafish Proteins/metabolism , Animals , Bone Morphogenetic Protein 4/genetics , Bone Morphogenetic Protein Receptors, Type I/genetics , Bone Morphogenetic Protein Receptors, Type I/metabolism , Carrier Proteins/genetics , Cell Line , Cell Movement/physiology , Endothelial Cells/cytology , HEK293 Cells , Humans , Low Density Lipoprotein Receptor-Related Protein-1 , Mice , Phenotype , RNA, Small Interfering/genetics , Receptors, LDL/genetics , Signal Transduction/physiology , Tumor Suppressor Proteins/genetics , Zebrafish , Zebrafish Proteins/genetics
19.
Arterioscler Thromb Vasc Biol ; 32(9): 2214-22, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22772758

ABSTRACT

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.


Subject(s)
Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Carrier Proteins/metabolism , Cell Adhesion Molecules/metabolism , Endothelial Cells/metabolism , Inflammation Mediators/metabolism , Inflammation/prevention & control , Animals , Aortic Diseases/genetics , Aortic Diseases/immunology , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Bone Morphogenetic Protein 4/metabolism , Carrier Proteins/genetics , Cells, Cultured , Disease Models, Animal , Endothelial Cells/immunology , Endothelial Cells/pathology , Genotype , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Intercellular Adhesion Molecule-1/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , RNA Interference , Recombinant Proteins/metabolism , Stress, Mechanical , Time Factors , Transfection , Vascular Calcification/immunology , Vascular Calcification/metabolism , Vascular Calcification/prevention & control , Vascular Cell Adhesion Molecule-1/metabolism
20.
Dev Biol ; 348(2): 167-76, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-20920499

ABSTRACT

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.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Heart/embryology , Hedgehog Proteins/metabolism , Signal Transduction , Animals , Bone Morphogenetic Proteins/genetics , Cell Proliferation , Chick Embryo , Cyclohexylamines/pharmacology , Hedgehog Proteins/genetics , Myocardium/metabolism , Organogenesis , Thiophenes/pharmacology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...