Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Cardiovasc Dev Dis ; 9(8)2022 Aug 16.
Article in English | MEDLINE | ID: mdl-36005436

ABSTRACT

Calcific aortic valve disease (CAVD) is a common cardiac defect, particularly in the aging population. While several risk factors, such as bi-leaflet valve structure and old age, have been identified in CAVD pathogenesis, molecular mechanisms resulting in this condition are still under active investigation. Bone morphogenetic protein signaling via the activin type I receptor (AcvRI) plays an important role during physiological and pathological processes involving calcification, e.g., bone formation and heterotopic ossification. In addition, AcvRI is required for normal cardiac valve development, yet its role in aortic valve disease, if any, is currently unknown. Here, we induced the expression of constitutively active AcvRI in developing mouse embryos in the endocardium and in cells at the valve leaflet-wall junction that are not of endocardium origin using the Nfac1Cre transgene. The mutant mice were born alive, but showed thickened aortic and pulmonary valve leaflets during the early postnatal period. Adult mutant mice developed aortic stenosis with high frequency, sclerotic aortic valves, and displayed Alcian Blue-positive hypertrophic chondrocyte-like cells at the leaflet-wall junction. Calcification was only seen with low penetrance. In addition, we observed that the expression levels of gene sets associated with inflammation-related cytokine signaling, smooth muscle cell contraction, and cGMP signaling were altered in the mutants when compared with those of the controls. This work shows that, in a mouse model, such continuous AcvRI activity in the Nfatc1Cre recombination domain results in pathological changes in the aortic valve structure and function.

2.
Nat Commun ; 13(1): 4065, 2022 07 13.
Article in English | MEDLINE | ID: mdl-35831318

ABSTRACT

Developmental etiologies causing complex congenital aortic root abnormalities are unknown. Here we show that deletion of Sox17 in aortic root endothelium in mice causes underdeveloped aortic root leading to a bicuspid aortic valve due to the absence of non-coronary leaflet and mispositioned left coronary ostium. The respective defects are associated with reduced proliferation of non-coronary leaflet mesenchyme and aortic root smooth muscle derived from the second heart field cardiomyocytes. Mechanistically, SOX17 occupies a Pdgfb transcriptional enhancer to promote its transcription and Sox17 deletion inhibits the endothelial Pdgfb transcription and PDGFB growth signaling to the non-coronary leaflet mesenchyme. Restoration of PDGFB in aortic root endothelium rescues the non-coronary leaflet and left coronary ostium defects in Sox17 nulls. These data support a SOX17-PDGFB axis underlying aortic root development that is critical for aortic valve and coronary ostium patterning, thereby informing a potential shared disease mechanism for concurrent anomalous aortic valve and coronary arteries.


Subject(s)
Bicuspid Aortic Valve Disease , Heart Defects, Congenital , Heart Valve Diseases , Animals , Aortic Valve/abnormalities , HMGB Proteins , Mice , Proto-Oncogene Proteins c-sis , SOXF Transcription Factors/genetics
3.
Adv Exp Med Biol ; 1132: 177-191, 2019.
Article in English | MEDLINE | ID: mdl-31037635

ABSTRACT

Although periostin plays a significant role in adult cardiac remodeling diseases, the focus of this review is on periostin as a valvulogenic gene. Periostin is expressed throughout valvular development, initially being expressed in endocardial endothelial cells that have been activated to transform into prevalvular mesenchyme termed "cushion tissues" that sustain expression of periostin throughout their morphogenesis into mature (compacted) valve leaflets. The phenotype of periostin null indicates that periostin is not required for endocardial transformation nor the proliferation of its mesenchymal progeny but rather promotes cellular behaviors that promote migration, survival (anti-apoptotic), differentiation into fibroblastic lineages, collagen secretion and postnatal remodeling/maturation. These morphogenetic activities are promoted or coordinated by periostin signaling through integrin receptors activating downstream kinases in cushion cells that activate hyaluronan synthetase II (Akt/PI3K), collagen synthesis (Erk/MapK) and changes in cytoskeletal organization (Pak1) which regulate postnatal remodeling of cells and associated collagenous matrix into a trilaminar (zonal) histoarchitecture. Pak1 binding to filamin A is proposed as one mechanism by which periostin supports remodeling. The failure to properly remodel cushions sets up a trajectory of degenerative (myxomatous-like) changes that over time reduce biomechanical properties and increase chances for prolapse, regurgitation or calcification of the leaflets. Included in the review are considerations of lineage diversity and the role of periostin as a determinant of mesenchymal cell fate.


Subject(s)
Cell Adhesion Molecules/physiology , Heart Valves/growth & development , Organogenesis , Cell Differentiation , Endothelial Cells/cytology , Humans , Integrins , Mesoderm/cytology
4.
Nat Commun ; 10(1): 1929, 2019 04 26.
Article in English | MEDLINE | ID: mdl-31028265

ABSTRACT

Genetically modified mice have advanced our understanding of valve development and disease. Yet, human pathophysiological valvulogenesis remains poorly understood. Here we report that, by combining single cell sequencing and in vivo approaches, a population of human pre-valvular endocardial cells (HPVCs) can be derived from pluripotent stem cells. HPVCs express gene patterns conforming to the E9.0 mouse atrio-ventricular canal (AVC) endocardium signature. HPVCs treated with BMP2, cultured on mouse AVC cushions, or transplanted into the AVC of embryonic mouse hearts, undergo endothelial-to-mesenchymal transition and express markers of valve interstitial cells of different valvular layers, demonstrating cell specificity. Extending this model to patient-specific induced pluripotent stem cells recapitulates features of mitral valve prolapse and identified dysregulation of the SHH pathway. Concurrently increased ECM secretion can be rescued by SHH inhibition, thus providing a putative therapeutic target. In summary, we report a human cell model of valvulogenesis that faithfully recapitulates valve disease in a dish.


Subject(s)
Endothelial Cells/pathology , Hedgehog Proteins/genetics , Mitral Valve Prolapse/pathology , Mitral Valve/pathology , Pluripotent Stem Cells/pathology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Biomarkers/metabolism , Bone Morphogenetic Protein 2/pharmacology , Cadherin Related Proteins , Cadherins/genetics , Cadherins/metabolism , Cell Differentiation/drug effects , Embryo, Mammalian , Endocardium/metabolism , Endocardium/pathology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/transplantation , Epithelial-Mesenchymal Transition/drug effects , GATA5 Transcription Factor/genetics , GATA5 Transcription Factor/metabolism , Gene Expression Profiling , Gene Expression Regulation , Heart Atria/metabolism , Heart Atria/pathology , Hedgehog Proteins/metabolism , Humans , Mice , Mitral Valve/metabolism , Mitral Valve Prolapse/genetics , Mitral Valve Prolapse/metabolism , Mitral Valve Prolapse/therapy , Models, Biological , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Primary Cell Culture , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Wnt3A Protein/pharmacology
5.
Endocrinology ; 159(2): 1062-1073, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29272356

ABSTRACT

Deficiencies in pancreatic ß-cell mass contribute to both type 1 and type 2 diabetes. We investigated the role of the glucose-regulated protein (GRP) 94, an endoplasmic reticulum protein abundantly expressed in the pancreatic acini and islets, in ß-cell development, survival, and function. We used a conditional knockout (KO) mouse in which the GRP94 gene, Hsp90b1, was specifically deleted in pancreatic and duodenal homeobox 1 (Pdx1)-expressing cells. These Hsp90b1 flox/flox;Pdx1Cre KO mice exhibited pancreatic hypoplasia at embryonic day (E) 16.5 to E18.5 and had significantly reduced ß-cell mass at 4 weeks after birth. Further mechanistic studies showed that deletion of GRP94 reduced ß-cell proliferation with increased cell apoptosis in both Pdx1+ endocrine progenitor cells and differentiated ß cells. Although Hsp90b1 flox/flox;Pdx1Cre KO mice remained euglycemic at 8 weeks of age, they exhibited impaired glucose tolerance. In aggregate, these findings indicate that GRP94 is an essential regulator of pancreatic ß-cell development, mass, and function.


Subject(s)
Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/physiology , Membrane Glycoproteins/physiology , Pancreas/embryology , Animals , Cell Count , Cell Differentiation/genetics , Cell Proliferation/genetics , Cells, Cultured , Embryo, Nonmammalian , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , HEK293 Cells , Humans , Male , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Size/genetics , Pancreas/growth & development , Pancreas/physiology
6.
Dev Biol ; 430(1): 113-128, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28790014

ABSTRACT

Distal outgrowth, maturation and remodeling of the endocardial cushion mesenchyme in the atrioventricular (AV) canal are the essential morphogenetic events during four-chambered heart formation. Mesenchymalized AV endocardial cushions give rise to the AV valves and the membranous ventricular septum (VS). Failure of these processes results in several human congenital heart defects. Despite this clinical relevance, the mechanisms governing how mesenchymalized AV endocardial cushions mature and remodel into the membranous VS and AV valves have only begun to be elucidated. The role of BMP signaling in the myocardial and secondary heart forming lineage has been well studied; however, little is known about the role of BMP2 expression in the endocardial lineage. To fill this knowledge gap, we generated Bmp2 endocardial lineage-specific conditional knockouts (referred to as Bmp2 cKOEndo) by crossing conditionally-targeted Bmp2flox/flox mice with a Cre-driver line, Nfatc1Cre, wherein Cre-mediated recombination was restricted to the endocardial cells and their mesenchymal progeny. Bmp2 cKOEndo mouse embryos did not exhibit failure or delay in the initial AV endocardial cushion formation at embryonic day (ED) 9.5-11.5; however, significant reductions in AV cushion size were detected in Bmp2 cKOEndo mouse embryos when compared to control embryos at ED13.5 and ED16.5. Moreover, deletion of Bmp2 from the endocardial lineage consistently resulted in membranous ventricular septal defects (VSDs), and mitral valve deficiencies, as evidenced by the absence of stratification of mitral valves at birth. Muscular VSDs were not found in Bmp2 cKOEndo mouse hearts. To understand the underlying morphogenetic mechanisms leading to a decrease in cushion size, cell proliferation and cell death were examined for AV endocardial cushions. Phospho-histone H3 analyses for cell proliferation and TUNEL assays for apoptotic cell death did not reveal significant differences between control and Bmp2 cKOEndo in AV endocardial cushions. However, mRNA expression of the extracellular matrix components, versican, Has2, collagen 9a1, and periostin was significantly reduced in Bmp2 cKOEndo AV cushions. Expression of transcription factors implicated in the cardiac valvulogenesis, Snail2, Twist1 and Sox9, was also significantly reduced in Bmp2 cKOEndo AV cushions. These data provide evidence that BMP2 expression in the endocardial lineage is essential for the distal outgrowth, maturation and remodeling of AV endocardial cushions into the normal membranous VS and the stratified AV valves.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Cell Lineage , Endocardial Cushions/cytology , Endocardial Cushions/growth & development , Animals , Animals, Newborn , Bone Morphogenetic Protein 2/genetics , Cell Adhesion Molecules/metabolism , Cell Death , Cell Proliferation , Collagen/metabolism , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Endocardial Cushions/metabolism , Gene Deletion , Heart Septal Defects, Ventricular/metabolism , Heart Septal Defects, Ventricular/pathology , Imaging, Three-Dimensional , Immunohistochemistry , Mesoderm/cytology , Mice, Knockout , Mitral Valve/pathology , NFATC Transcription Factors/metabolism , Proteoglycans/metabolism , Transcription Factors/metabolism , Transformation, Genetic
7.
Mech Dev ; 136: 123-32, 2015 May.
Article in English | MEDLINE | ID: mdl-25575930

ABSTRACT

Endothelial to mesenchymal transition (EMT) that occurs during cardiac outflow tract (OFT) development is critical for formation of the semilunar valves. Fibulin-1 (Fbln1) is an extracellular matrix protein that is present at several sites of EMT, including the OFT (i.e., E9.5-10.5). The aim of this study was to determine the role of Fbln1 in EMT during the earliest events of OFT development. Examination of proximal OFT cushions in Fbln1 null embryos detected hypercellularity at both E9.5 (93% increase; p = 0.002) and E10.5 (43% increase; p = 0.01) as compared to wild type, suggesting that Fbln1 normally suppresses OFT endocardial cushion EMT. This was supported by studies of proximal OFT cushion explants, which showed that explants from Fbln1 null embryos displayed a 58% increase in cells migrating from the explants as compared to wild type (p = 0.005). We next evaluated the effects of Fbln1 deficiency on the expression of factors that regulate proximal OFT EMT. At E9.5, Fbln1 null proximal OFT endocardium and EMT-derived mesenchyme showed increased TGFß2 (58% increase; p = 0.01) and increased Snail1-positive nuclei (27% increase; p = 0.0003). Histological examination of OFT cushions in Fbln1 null embryos (E9.5) also detected cells present in the cushion that were determined to be erythrocytes based on round morphology, autofluorescence, and positive staining for hemoglobin. Erythrocytes were also detected in Fbln1 null OFT cushions at E10.5. Together, the findings indicate that Fbln1 normally suppresses proximal OFT EMT preventing proximal cushion hypercellularity and blood cell accumulation.


Subject(s)
Calcium-Binding Proteins/metabolism , Endocardial Cushions/metabolism , Endocardium/metabolism , Extracellular Matrix Proteins/metabolism , Myocardium/metabolism , Animals , Apoptosis , Calcium-Binding Proteins/genetics , Cell Proliferation , Endocardial Cushions/cytology , Endocardium/cytology , Extracellular Matrix Proteins/genetics , Mice , Mice, Knockout , Myocardium/cytology
8.
Circ Cardiovasc Genet ; 7(4): 423-433, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25028484

ABSTRACT

BACKGROUND: Heterozygous human mutations of NKX2-5 are highly penetrant and associated with varied congenital heart defects. The heterozygous knockout of murine Nkx2-5, in contrast, manifests less profound cardiac malformations, with low disease penetrance. We sought to study this apparent discrepancy between human and mouse genetics. Because missense mutations in the NKX2-5 homeodomain (DNA-binding domain) are the most frequently reported type of human mutation, we replicated this genetic defect in a murine knockin model. METHODS AND RESULTS: We generated a murine model in a 129/Sv genetic background by knocking-in an Nkx2-5 homeodomain missense mutation previously identified in humans. The mutation was located at homeodomain position 52Arg→Gly (R52G). All the heterozygous neonatal Nkx2-5(+/R52G) mice demonstrated a prominent trabecular layer in the ventricular wall, so called noncompaction, along with diverse cardiac anomalies, including atrioventricular septal defects, Ebstein malformation of the tricuspid valve, and perimembranous and muscular ventricular septal defects. In addition, P10 Nkx2-5(+/R52G) mice demonstrated atrial sepal anomalies, with significant increase in the size of the interatrial communication and fossa ovalis, and decrease in the length of the flap valve compared with control Nkx2-5(+/+) or Nkx2-5(+/-) mice. CONCLUSIONS: The results of our study demonstrate that heterozygous missense mutation in the murine Nkx2-5 homeodomain (R52G) is highly penetrant and result in pleiotropic cardiac effects. Thus, in contrast to heterozygous Nkx2-5 knockout mice, the effects of the heterozygous knockin mimic findings in humans with heterozygous missense mutation in NKX2-5 homeodomain.


Subject(s)
Heart Defects, Congenital/genetics , Homeodomain Proteins/genetics , Transcription Factors/genetics , Amino Acid Sequence , Animals , Disease Models, Animal , Gene Knock-In Techniques , Heart Defects, Congenital/pathology , Heart Ventricles/pathology , Heterozygote , Homeobox Protein Nkx-2.5 , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Mutation, Missense , Phenotype , Protein Structure, Tertiary
9.
PLoS One ; 8(10): e77593, 2013.
Article in English | MEDLINE | ID: mdl-24147033

ABSTRACT

Distal outgrowth and maturation of mesenchymalized endocardial cushions are critical morphogenetic events during post-EMT atrioventricular (AV) valvuloseptal morphogenesis. We explored the role of BMP-2 in the regulation of valvulogenic extracellular matrix (ECM) components, versican and hyaluronan (HA), and cell migration during post-EMT AV cushion distal outgrowth/expansion. We observed intense staining of versican and HA in AV cushion mesenchyme from the early cushion expansion stage, Hamburger and Hamilton (HH) stage-17 to the cushion maturation stage, HH stage-29 in the chick. Based on this expression pattern we examined the role of BMP-2 in regulating versican and HA using 3D AV cushion mesenchymal cell (CMC) aggregate cultures on hydrated collagen gels. BMP-2 induced versican expression and HA deposition as well as mRNA expression of versican and Has2 by CMCs in a dose dependent manner. Noggin, an antagonist of BMP, abolished BMP-2-induced versican and HA as well as mRNA expression of versican and Has2. We further examined whether BMP-2-promoted cell migration was associated with expression of versican and HA. BMP-2- promoted cell migration was significantly impaired by treatments with versican siRNA and HA oligomer. In conclusion, we provide evidence that BMP-2 induces expression of versican and HA by AV CMCs and that these ECM components contribute to BMP-2-induced CMC migration, indicating critical roles for BMP-2 in distal outgrowth/expansion of mesenchymalized AV cushions.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Cell Movement , Endocardial Cushions/embryology , Endocardial Cushions/metabolism , Epithelial-Mesenchymal Transition , Hyaluronic Acid/metabolism , Versicans/metabolism , Animals , Bone Morphogenetic Protein 2/pharmacology , Cell Movement/drug effects , Cell Movement/genetics , Chick Embryo , Endocardial Cushions/drug effects , Gene Expression , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Hyaluronic Acid/genetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mesoderm/cytology , Mesoderm/metabolism , RNA Interference , RNA, Messenger/genetics , Versicans/genetics
10.
J Neonatal Biol ; 12012 Jan 21.
Article in English | MEDLINE | ID: mdl-25383261

ABSTRACT

Smad6 is known to predominantly inhibit BMP signaling by negatively regulating the BMP signaling process. Therefore, Smad6 mutation potentially provides an important genetic model for investigating the role of BMP signaling in vivo. Periostin is a 90-kDA secreted extracellular matrix (ECM) protein and implicated in cardiac valve progenitor cell differentiation, maturation and adult aortic valve calcification in mice. We have previously reported periostin expression patterns during AV valve development in mice. Because periostin can play critical roles in aortic valve interstitial cell differentiation and can be correlated with adult valve disease pathogenesis, in the present study we specifically focused on periostin expression during outflow tract (OT) development and its expression within the adult mouse valves. We previously reported that periostin expression in valve progenitor cells was altered by exogenously adding BMP-2 in culture. In this study, we investigated whether expression of periostin and other valvulogenic ECM proteins was altered in Smad6-mutant newborn mice in vivo. Periostin protein was localized within OT during embryonic development in mice. At embryonic day (ED) 13.5, robust periostin expression was detected within the developing pulmonary trunk and developing pulmonary and aortic valves. Periostin expression remained intense in pulmonary and aortic valves up to the adult stage. Our immunohistochemical and immunointensity analyses revealed that periostin expression was significantly reduced in the aortic valves in Smad6-/- neonatal hearts. Versican expression was also significantly reduced in Smad6-/- aortic valves, whereas, hyaluronan deposition was not significantly altered in the Smad6-/- neonatal valves. Expression of periostin and versican was less prominently affected in AV valves compared to the aortic valves, suggesting that a cell lineage/origin-dependent response to regulatory molecules may play a critical role in valve interstitial cell development and ECM protein expression.

11.
Dev Biol ; 316(2): 200-13, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18313657

ABSTRACT

Cardiac valve leaflets develop from rudimentary structures termed endocardial cushions. These pre-valve tissues arise from a complex interplay of signals between the myocardium and endocardium whereby secreted cues induce the endothelial cells to transform into migratory mesenchyme through an endothelial to mesenchymal transformation (EMT). Even though much is currently known regarding the initial EMT process, the mechanisms by which these undifferentiated cushion mesenchymal tissues are remodeled "post-EMT" into mature fibrous valve leaflets remains one of the major, unsolved questions in heart development. Expression analyses, presented in this report, demonstrate that periostin, a component of the extracellular matrix, is predominantly expressed in post-EMT valve tissues and their supporting apparatus from embryonic to adult life. Analyses of periostin gene targeted mice demonstrate that it is within these regions that significant defects are observed. Periostin null mice exhibit atrial septal defects, structural abnormalities of the AV valves and their supporting tensile apparatus, and aberrant differentiation of AV cushion mesenchyme. Rescue experiments further demonstrate that periostin functions as a hierarchical molecular switch that can promote the differentiation of mesenchymal cells into a fibroblastic lineage while repressing their transformation into other mesodermal cell lineages (e.g. myocytes). This is the first report of an extracellular matrix protein directly regulating post-EMT AV valve differentiation, a process foundational and indispensable for the morphogenesis of a cushion into a leaflet.


Subject(s)
Atrioventricular Node/embryology , Cell Adhesion Molecules/genetics , Gene Expression Regulation, Developmental , Heart Valves/embryology , Heart/embryology , Heart/physiology , Animals , Atrioventricular Node/ultrastructure , Cell Adhesion Molecules/deficiency , Embryonic Development , Heart Valves/ultrastructure , Mice , Mice, Knockout , Microscopy, Atomic Force
12.
Dev Biol ; 315(2): 383-96, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18261719

ABSTRACT

Atrioventricular (AV) endocardium transforms into the cushion mesenchyme, the primordia of the valves and membranous septa, through epithelial-mesenchymal transformation (EMT). While bone morphogenetic protein (BMP)-2 is known to be critical for AV EMT, the role of BMP-2 in post-EMT AV valvulogenesis remains to be elucidated. To find BMP signaling loops, we first localized Type I BMP receptors (BMPRs), BMPR-1A (ALK3), -1B (ALK6) and ALK2 in AV cushion mesenchyme in stage-24 chick embryos. Based on the BMP receptor expression pattern, we examined the functional roles of BMP-2 and BMP signaling in post-EMT valvulogenesis by using stage-24 AV cushion mesenchymal cell aggregates cultured on 3D-collagen gels. Exogenous BMP-2 or constitutively active (ca) BMPR-1B (ALK6)-virus treatments induced migration of the mesenchymal cells into the collagen gels, whereas noggin, an antagonist of BMPs, or dominant-negative (dn) BMPR-1 B (ALK6)-virus treatments reduced cell migration from the mesenchymal cell aggregates. Exogenous BMP-2 or caBMPR-1B (ALK6) treatments significantly promoted expression of an extracellular matrix (ECM) protein, periostin, a known valvulogenic matrix maturation mediator, at both mRNA and protein levels, whereas periostin expression was repressed by adding noggin or dnBMPR-1B (ALK6)-virus to the culture. Moreover, transcripts of Twist and Id1, which have been implicated in cell migration in embryogenesis and activation of the periostin promoter, were induced by BMP-2 but repressed by noggin in cushion mesenchymal cell cultures. These data provide evidence that BMP-2 and BMP signaling induce biological processes involved in early AV valvulogenesis, i.e. mesenchymal cell migration and expression of periostin, indicating critical roles for BMP signaling in post-EMT AV cushion tissue maturation and differentiation.


Subject(s)
Bone Morphogenetic Proteins/physiology , Cell Adhesion Molecules/metabolism , Endocardial Cushions/embryology , Transforming Growth Factor beta/physiology , Animals , Avian Proteins/genetics , Avian Proteins/metabolism , Base Sequence , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein Receptors, Type I/genetics , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bone Morphogenetic Proteins/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Movement/drug effects , Cell Movement/physiology , Chick Embryo , DNA Primers/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Endocardial Cushions/cytology , Endocardial Cushions/drug effects , Endocardial Cushions/metabolism , In Situ Hybridization , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Models, Biological , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta/pharmacology , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism
13.
Dev Biol ; 306(1): 179-92, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17449024

ABSTRACT

Transformation of atrioventricular (AV) canal endocardium into invasive mesenchyme correlates spatially and temporally with the expression of bone morphogenetic protein (BMP)-2 in the AV myocardium. We revealed the presence of mRNA of Type I BMP receptors, BMPR-1A (ALK3), BMPR-1B (ALK6) and ALK2 in chick AV endocardium at stage-14(-), the onset of epithelial to mesenchymal transformation (EMT), by RT-PCR and localized BMPR-1B mRNA in the endocardium by in situ hybridization. To circumvent the functional redundancies among the Type I BMP receptors, we applied dominant-negative (dn) BMPR-1B-viruses to chick AV explants and whole-chick embryo cultures to specifically block BMP signaling in AV endocardium during EMT. dnBMPR-1B-virus infection of AV endocardial cells abolished BMP-2-supported AV endocardial EMT. Conversely, caBMPR-1B-virus infection promoted AV endocardial EMT in the absence of AV myocardium. Moreover, dnBMPR-1B-virus treatments significantly reduced myocardially supported EMT in AV endocardial-myocardial co-culture. AV cushion mesenchymal cell markers, alpha-smooth muscle actin (SMA), and TGFbeta3 in the endocardial cells were promoted by caBMPR-1B and reduced by dnBMPR-1B infection. Microinjection of the virus into the cardiac jelly in the AV canal at stage-13 in vivo (ovo) revealed that the dnBMPR-1B-virus-infected cells remained in the endocardial epithelium, whereas caBMPR-1B-infected cells invaded deep into the cushions. These results provide evidence that BMP signaling through the AV endocardium is required for the EMT and the activation of the BMP receptor in the endocardium can promote AV EMT in the chick.


Subject(s)
Bone Morphogenetic Protein Receptors, Type I/metabolism , Endocardium/embryology , Endocardium/metabolism , Heart/embryology , Mesoderm/cytology , Animals , Bone Morphogenetic Protein Receptors, Type I/genetics , Carrier Proteins/metabolism , Chick Embryo , Coculture Techniques , Collagen/chemistry , Endocardium/cytology , Gels/chemistry , Heart Atria/cytology , Heart Atria/embryology , Heart Atria/metabolism , Heart Ventricles/cytology , Heart Ventricles/embryology , Heart Ventricles/metabolism , Mesoderm/metabolism , Microinjections , Myocardium/cytology , Myocardium/metabolism , Signal Transduction , Transforming Growth Factor beta3/metabolism
14.
J Cell Biochem ; 101(3): 695-711, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17226767

ABSTRACT

Periostin is predominantly expressed in collagen-rich fibrous connective tissues that are subjected to constant mechanical stresses including: heart valves, tendons, perichondrium, cornea, and the periodontal ligament (PDL). Based on these data we hypothesize that periostin can regulate collagen I fibrillogenesis and thereby affect the biomechanical properties of connective tissues. Immunoprecipitation and immunogold transmission electron microscopy experiments demonstrate that periostin is capable of directly interacting with collagen I. To analyze the potential role of periostin in collagen I fibrillogenesis, gene targeted mice were generated. Transmission electron microscopy and morphometric analyses demonstrated reduced collagen fibril diameters in skin dermis of periostin knockout mice, an indication of aberrant collagen I fibrillogenesis. In addition, differential scanning calorimetry (DSC) demonstrated a lower collagen denaturing temperature in periostin knockout mice, reflecting a reduced level of collagen cross-linking. Functional biomechanical properties of periostin null skin specimens and atrioventricular (AV) valve explant experiments provided direct evidence of the role that periostin plays in regulating the viscoelastic properties of connective tissues. Collectively, these data demonstrate for the first time that periostin can regulate collagen I fibrillogenesis and thereby serves as an important mediator of the biomechanical properties of fibrous connective tissues.


Subject(s)
Cell Adhesion Molecules/metabolism , Connective Tissue/metabolism , Fibrillar Collagens/metabolism , Adenoviridae/genetics , Adenoviridae/growth & development , Animals , Biomechanical Phenomena , Blotting, Western , Calorimetry, Differential Scanning , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/physiology , Cell Line , Chick Embryo , Chickens , Collagen Type I/metabolism , Connective Tissue/growth & development , Female , Fibrillar Collagens/ultrastructure , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry , Immunoprecipitation , Male , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Mutation , Protein Binding , Skin/metabolism , Skin/ultrastructure
15.
Dev Biol ; 269(2): 505-18, 2004 May 15.
Article in English | MEDLINE | ID: mdl-15110716

ABSTRACT

Transformation of endocardial endothelial cells into invasive mesenchyme is a critical antecedent of cardiac cushion tissue formation. The message for bone morphogenetic protein (BMP)-2 is known to be expressed in myocardial cells in a manner consistent with the segmental pattern of cushion formation [Development 109(1990) 833]. In the present work, we localized BMP-2 protein in atrioventricular (AV) myocardium in mice at embryonic day (ED) 8.5 (12 somite stage) before the onset of AV mesenchymal cell formation at ED 9.5. BMP-2 protein expression was absent from ventricular myocardium throughout the stages examined. After cellularization of the AV cushion at ED 10.5, myocardial BMP-2 protein expression was diminished in AV myocardium, whereas cushion mesenchymal cells started expressing BMP protein. Expression of BMP-2 in cushion mesenchyme persisted during later stages of development, ED 13.5-16, during valuvulogenesis. Intense expression of BMP-2 persisted in the valve tissue in adult mice. Based on the expression pattern, we performed a series of experiments to test the hypothesis that BMP-2 mediates myocardial regulation of cardiac cushion tissue formation in mice. When BMP-2 protein was added to the 16-18 somite stage (ED 9.25) AV endocardial endothelium in culture, cushion mesenchymal cells were formed in the absence of AV myocardium, which invaded into collagen gels and expressed the mesenchymal marker, smooth muscle (SM) alpha-actin; whereas the endothelial marker, PECAM-1, was lost from the invaded cells. In contrast, when noggin, a specific antagonist to BMPs, was applied together with BMP-2 to the culture medium, AV endothelial cells remained as an epithelial monolayer with little expression of SM alpha-actin, and expression of PECAM-1 was retained in the endocardial cells. When noggin was added to AV endothelial cells cocultured with associated myocardium, it blocked endothelial transformation to mesenchyme. AV endothelium treated with BMP-2 expressed elevated levels of TGFbeta-2 in the absence of myocardium, as observed in the endothelium cocultured with myocardium. BMP-2-supported elevation of TGFbeta-2 expression in endocardial cells was abolished by noggin treatment. These data indicated that BMP signaling is required in and BMP-2 is sufficient for myocardial segmental regulation of AV endocardial cushion mesenchymal cell formation in mice.


Subject(s)
Atrioventricular Node/embryology , Bone Morphogenetic Proteins/physiology , Mesoderm/cytology , Animals , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/analysis , Carrier Proteins , Endothelial Cells/cytology , Heart Valves/embryology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Proteins/physiology , Signal Transduction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta2
16.
Dev Biol ; 263(1): 35-49, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-14568545

ABSTRACT

We previously demonstrated that the initial emergence of endocardial precursor cells (endocardial angioblasts) occurred within the precardiac mesoderm and that the endodermal secretory products promoted delamination of cells from the precardiac mesoderm and expression of endothelial lineage markers [Dev. Biol. 175 (1996), 66]. In this study, we sought to extend our original study to the identification of candidate molecules derived from the endoderm that might have induced endocardial precursor cell formation. We have detected expression of transforming growth factors beta (TGFbeta) 2, 3, and 4 in anterior endoderm at Hamburger and Hamilton (H-H) stage 5 by RT-PCR. To address the role of growth factors known to be present in the endoderm, precardiac mesodermal explants were isolated from H-H stage 5 quail embryos and cultured on the surface of collagen gels with serum-free defined medium 199. Similar to the effect of explants cocultured with anterior endoderm, when cultured with TGFbetas 1-3 (3 ng/ml each), explants formed QH-1 (anti-quail endothelial marker)-positive mesenchymal cells, which invaded the gel and expressed the extracellular marker, cytotactin (tenascin). Another member of the TGFbeta superfamily, bone morphogenetic protein-2 (BMP-2; 100 ng/ml), did not induce QH-1-positive mesenchymal cell formation but promoted formation of an epithelial monolayer on the surface of the collagen gel; this monolayer did not express QH-1. Explants treated with vascular endothelial growth factor (VEGF(165), 100 ng/ml) also did not invade the gel but formed an epithelial-like outgrowth on the surface of the gel. However, this monolayer did express the QH-1 marker. Fibroblast growth factor-2 (FGF-2; 250 ng/ml)-treated explants expressed QH-1 and exhibited separation of the cells on the surface of the gel. Finally, a combination of TGFbetas and VEGF enhanced formation of QH-1-positive cord-like structures within the gel from mesenchyme that had previously invaded the gel. Luminization of the cords, however, was not clearly evident. These findings suggest that TGFbetas, among the growth factors tested, mediate the initial step of endocardial formation, i.e., delamination of endothelial precursor cells from precardiac mesoderm, whereas VEGF may primarily effect early vasculogenesis (cord-like structure formation).


Subject(s)
Endoderm/physiology , Growth Substances/pharmacology , Heart/embryology , Mesoderm/physiology , Nuclear Proteins , Stem Cells/physiology , Animals , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/pharmacology , Coturnix , DNA-Binding Proteins/genetics , Fibroblast Growth Factor 2/pharmacology , NFATC Transcription Factors , Tenascin/analysis , Transcription Factors/genetics , Transforming Growth Factor beta/pharmacology
17.
Dev Biol ; 258(2): 252-63, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12798286

ABSTRACT

While much has been learned about how endothelial cells transform to mesenchyme during cardiac cushion formation, there remain fundamental questions about the developmental fate of cushions. In the present work, we focus on the growth and development of cushion mesenchyme. We hypothesize that proliferative expansion and distal elongation of cushion mesenchyme mediated by growth factors are the basis of early valve leaflet formation. As a first step to test this hypothesis, we have localized fibroblast growth factor (FGF)-4 protein in cushion mesenchymal cells at the onset of prevalve leaflet formation in chick embryos (Hamburger and Hamilton stage 20-25). Ligand distribution was correlated with FGF receptor (FGFR) expression. In situ hybridization data indicated that FGFR3 mRNA was confined to the endocardial rim of the atrioventricular (AV) cushion pads, whereas FGFR2 was expressed exclusively in cushion mesenchymal cells. FGFR1 expression was detected in both endocardium and cushion mesenchyme as well as in myocardium. To determine whether the FGF pathways play regulatory roles in cushion mesenchymal cell proliferation and elongation into prevalvular structure, FGF-4 protein was added to the cushion mesenchymal cells explanted from stage 24-25 chick embryos. A significant increase in proliferative ability was strongly suggested in FGF-4-treated mesenchymal cells as judged by the incorporation of 5'-bromodeoxyuridine (BrdU). To determine whether cushion cells responded similarly in vivo, a replication-defective retrovirus encoding FGF-4 with the reporter, bacterial beta-galactosidase was microinjected into stage 18 chick cardiac cushion mesenchyme along the inner curvature where AV and outflow cushions converge. As compared with vector controls, overexpression of FGF-4 clearly induced expansion of cushion mesenchyme toward the lumen. To further test the proliferative effect of FGF-4 in cardiac cushion expansion in vivo (ovo), FGF-4 protein was microinjected into stage 18 chick inner curvature. An assay for BrdU incorporation indicated a significant increase in proliferative ability in FGF-4 microinjected cardiac cushion mesenchyme as compared with BSA-microinjected controls. Together, these results suggest a role of FGF-4 for cardiac valve leaflet formation through proliferative expansion of cushion mesenchyme.


Subject(s)
Fibroblast Growth Factors/physiology , Heart Valves/embryology , Protein-Tyrosine Kinases , Proto-Oncogene Proteins/physiology , Animals , Bromodeoxyuridine/metabolism , Cell Division/drug effects , Cells, Cultured , Chick Embryo , Fibroblast Growth Factor 4 , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/pharmacology , Gene Expression Regulation, Developmental , Heart Valves/cytology , Heart Valves/drug effects , Immunohistochemistry , In Situ Hybridization , In Vitro Techniques , Mesoderm/cytology , Mesoderm/drug effects , Microinjections , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor, Fibroblast Growth Factor, Type 1 , Receptor, Fibroblast Growth Factor, Type 2 , Receptor, Fibroblast Growth Factor, Type 3 , Receptors, Fibroblast Growth Factor/genetics , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology
18.
J Histochem Cytochem ; 31(1A_suppl): 219-221, 1983 Jan.
Article in English | MEDLINE | ID: mdl-28054842

ABSTRACT

Secretion granules in the shell gland, isthmus, and albumin-secreting region of the hen oviduct were analyzed with WET-scanning electron microscopy (SEM) and EDX, a combination of wide-angle backscattered electron detector (BED) and energy-dispersive X-ray microanalyzer (EDX). Glutaraldehyde-fixed but unhydrated, unstained, and uncoated samples were analyzed; Ca was localized in all secretion granules in all three sections of the hen oviduct studied.

SELECTION OF CITATIONS
SEARCH DETAIL
...