Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Adv Exp Med Biol ; 1441: 155-166, 2024.
Article in English | MEDLINE | ID: mdl-38884710

ABSTRACT

Congenital anomalies and acquired diseases of the coronary blood vessels are of great clinical relevance. The early diagnosis of these conditions remains, however, challenging. In order to improve our knowledge of these ailments, progress has to be achieved in the research of the molecular and cellular mechanisms that control development of the coronary vascular bed. The aim of this chapter is to provide a succint account of the key elements of coronary blood vessel development, especially in the context of the role played by the epicardium and epicardial cellular derivatives. We will discuss the importance of the epicardium in coronary blood vessel morphogenesis, from the contribution of the epicardially derived mesenchyme to these blood vessels to its role as an instructive signaling center, attempting to relate these concepts to the origin of coronary disease.


Subject(s)
Coronary Vessels , Pericardium , Pericardium/embryology , Humans , Coronary Vessels/embryology , Animals , Signal Transduction , Mesoderm , Morphogenesis
2.
Adv Exp Med Biol ; 1441: 811-816, 2024.
Article in English | MEDLINE | ID: mdl-38884750

ABSTRACT

The genetics of human congenital coronary vascular anomalies (hCCVA) remains largely underresearched. This is surprising, because although coronary vascular defects represent a relatively small proportion of human congenital heart disease (CHD), hCCVAs are clinically significant conditions. Indeed, hCCVA frequently associate to other congenital cardiac structural defects and may even result in sudden cardiac death in the adult. In this brief chapter, we will attempt to summarize our current knowledge on the topic, also proposing a rationale for the development of novel approaches to the genetics of hCCVA.


Subject(s)
Coronary Vessel Anomalies , Humans , Coronary Vessel Anomalies/genetics , Genetic Predisposition to Disease/genetics , Heart Defects, Congenital/genetics
3.
Adv Exp Med Biol ; 1441: 817-831, 2024.
Article in English | MEDLINE | ID: mdl-38884751

ABSTRACT

Coronary blood vessels are in charge of sustaining cardiac homeostasis. It is thus logical that coronary congenital anomalies (CCA) directly or indirectly associate with multiple cardiac conditions, including sudden death. The coronary vascular system is a sophisticated, highly patterned anatomical entity, and therefore a wide range of congenital malformations of the coronary vasculature have been described. Despite the clinical interest of CCA, very few attempts have been made to relate specific embryonic developmental mechanisms to the congenital anomalies of these blood vessels. This is so because developmental data on the morphogenesis of the coronary vascular system derive from complex studies carried out in animals (mostly transgenic mice), and are not often accessible to the clinician, who, in turn, possesses essential information on the significance of CCA. During the last decade, advances in our understanding of normal embryonic development of coronary blood vessels have provided insight into the cellular and molecular mechanisms underlying coronary arteries anomalies. These findings are the base for our attempt to offer plausible embryological explanations to a variety of CCA as based on the analysis of multiple animal models for the study of cardiac embryogenesis, and present them in an organized manner, offering to the reader developmental mechanistic explanations for the pathogenesis of these anomalies.


Subject(s)
Coronary Vessel Anomalies , Coronary Vessels , Animals , Humans , Mice , Coronary Vessel Anomalies/pathology , Coronary Vessel Anomalies/genetics , Coronary Vessel Anomalies/embryology , Coronary Vessels/embryology , Coronary Vessels/pathology , Coronary Vessels/physiopathology , Disease Models, Animal
4.
J Pers Med ; 12(2)2022 Feb 04.
Article in English | MEDLINE | ID: mdl-35207702

ABSTRACT

Cardiomyocytes derived from human pluripotent stem cells (hPSC-CMs) hold a great potential as human in vitro models for studying heart disease and for drug safety screening. Nevertheless, their associated immaturity relative to the adult myocardium limits their utility in cardiac research. In this study, we describe the development of a platform for generating three-dimensional engineered heart tissues (EHTs) from hPSC-CMs for the measurement of force while under mechanical and electrical stimulation. The modular and versatile EHT platform presented here allows for the formation of three tissues per well in a 12-well plate format, resulting in 36 tissues per plate. We compared the functional performance of EHTs and their histology in three different media and demonstrated that tissues cultured and maintained in maturation medium, containing triiodothyronine (T3), dexamethasone, and insulin-like growth factor-1 (TDI), resulted in a higher force of contraction, sarcomeric organization and alignment, and a higher and lower inotropic response to isoproterenol and nifedipine, respectively. Moreover, in this study, we highlight the importance of integrating a serum-free maturation medium in the EHT platform, making it a suitable tool for cardiovascular research, disease modeling, and preclinical drug testing.

5.
Front Cell Dev Biol ; 9: 645276, 2021.
Article in English | MEDLINE | ID: mdl-34055776

ABSTRACT

During the last decade, extensive efforts have been made to comprehend cardiac cell genetic and functional diversity. Such knowledge allows for the definition of the cardiac cellular interactome as a reasonable strategy to increase our understanding of the normal and pathologic heart. Previous experimental approaches including cell lineage tracing, flow cytometry, and bulk RNA-Seq have often tackled the analysis of cardiac cell diversity as based on the assumption that cell types can be identified by the expression of a single gene. More recently, however, the emergence of single-cell RNA-Seq technology has led us to explore the diversity of individual cells, enabling the cardiovascular research community to redefine cardiac cell subpopulations and identify relevant ones, and even novel cell types, through their cell-specific transcriptomic signatures in an unbiased manner. These findings are changing our understanding of cell composition and in consequence the identification of potential therapeutic targets for different cardiac diseases. In this review, we provide an overview of the continuously changing cardiac cellular landscape, traveling from the pre-single-cell RNA-Seq times to the single cell-RNA-Seq revolution, and discuss the utilities and limitations of this technology.

6.
Semin Cell Dev Biol ; 112: 16-26, 2021 04.
Article in English | MEDLINE | ID: mdl-32591270

ABSTRACT

Cardiac chamber walls contain large numbers of non-contractile interstitial cells, including fibroblasts, endothelial cells, pericytes and significant populations of blood lineage-derived cells. Blood cells first colonize heart tissues a few days before birth, although their recruitment from the bloodstream to the cardiac interstitium is continuous and extends throughout adult life. The bone marrow, as the major hematopoietic site of adult individuals, is in charge of renewing all circulating cell types, and it therefore plays a pivotal role in the incorporation of blood cells to the heart. Bone marrow-derived cells are instrumental to tissue homeostasis in the steady-state heart, and are major effectors in cardiac disease progression. This review will provide a comprehensive approach to bone marrow-derived blood cell functions in the heart, and discuss aspects related to hot topics in the cardiovascular field like cell-based heart regeneration strategies.


Subject(s)
Bone Marrow/physiology , Heart/growth & development , Hematopoietic Stem Cells/physiology , Regeneration/physiology , Bone Marrow Cells/physiology , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Lineage/physiology , Endothelial Cells/physiology , Heart/physiopathology , Heart Diseases/genetics , Heart Diseases/physiopathology , Humans
7.
ACS Sens ; 5(4): 1068-1074, 2020 04 24.
Article in English | MEDLINE | ID: mdl-32227860

ABSTRACT

Novel pH probes based on 2-(6-methoxynaphthalen-2-yl)-3,3-dimethyl-3H-indole have been synthesized and characterized. These compounds display excellent "off-on" fluorescence responses to acidic pH especially under two-photon (TP) excitation conditions as well as strong selectivity and sensitivity toward H+. These features are supported by fluorescence quantum yields over 35%, TP cross sections ∼60 GM, and good resistance to photodegradation under acidic conditions. The synthetic versatility of this model allows subcellular targets to be tuned through minor scaffold modifications without affecting its optical characteristics. The effectiveness of the probes' innate photophysical properties and the structural modifications for different pH-related applications are demonstrated in mouse embryonic fibroblast cells.


Subject(s)
Biosensing Techniques/methods , Fluorescent Dyes/therapeutic use , Humans , Hydrogen-Ion Concentration , Photons
8.
ACS Omega ; 4(8): 13027-13033, 2019 Aug 20.
Article in English | MEDLINE | ID: mdl-31460429

ABSTRACT

Herein, we present a water-soluble dendritric Pt(II) complex as a phosphorescent label for bacterial cells. The dendritic moiety endows the Pt(II) complex with unique properties such as water solubility, shielding from quenching by dioxygen, and binding to bacterial surfaces. The new biosensor was employed for two-photon excitation microscopy, and the binding was confirmed by electron microscopy, which demonstrates that such hybrid arrays can provide orthogonal yet complementary readouts.

9.
J Org Chem ; 84(16): 10197-10208, 2019 08 16.
Article in English | MEDLINE | ID: mdl-31310119

ABSTRACT

Herein, we present an easy and efficient synthesis of amino terminal dendrons, combining protection/deprotection reactions with copper-catalyzed azide alkyne cycloaddition in a convergent way. This new approach affords dendrons in gram scale with excellent yields and easy purification. By choosing the appropriate azido-functionalized core, these dendrons lead to a more efficient and controlled convergent synthesis of dendrimers with different sizes and shapes and multivalence. The amino terminal dendrimers were analyzed by diffusion-ordered spectroscopy experiments. The observed dendrimer size is in excellent correlation with the expected size and shape by molecular dynamic simulations. The construction of these kinds of nanostructures, in a simple and efficient way, opens new opportunities for biomedical applications. Moreover, by choosing the appropriate core, these versatile macromolecules become an excellent fluorescent biomarker.


Subject(s)
Dendrimers/chemistry , Dendrimers/chemical synthesis , Alkynes/chemistry , Azides/chemistry , Biomarkers/chemistry , Catalysis , Copper/chemistry , Cycloaddition Reaction , Molecular Dynamics Simulation , Molecular Structure , Particle Size
10.
Anat Rec (Hoboken) ; 302(1): 58-68, 2019 01.
Article in English | MEDLINE | ID: mdl-30288955

ABSTRACT

The space between cardiac myocytes is commonly referred-to as the cardiac interstitium (CI). The CI is a unique, complex and dynamic microenvironment in which multiple cell types, extracellular matrix molecules, and instructive signals interact to crucially support heart homeostasis and promote cardiac responses to normal and pathologic stimuli. Despite the biomedical and clinical relevance of the CI, its detailed cellular structure remains to be elucidated. In this review, we will dissect the organization of the cardiac interstitium by following its changing cellular and molecular composition from embryonic developmental stages to adulthood, providing a systematic analysis of the biological components of the CI. The main goal of this review is to contribute to our understanding of the CI roles in health and disease. Anat Rec, 302:58-68, 2019. © 2018 Wiley Periodicals, Inc.


Subject(s)
Embryonic Development , Extracellular Space/chemistry , Myocardium/cytology , Animals , Humans
11.
Dev Dyn ; 247(5): 686-698, 2018 05.
Article in English | MEDLINE | ID: mdl-29226547

ABSTRACT

BACKGROUND: Coronary vasculature irrigates the myocardium and is crucial to late embryonic and adult heart function. Despite the developmental significance and clinical relevance of these blood vessels, the embryonic origin and the cellular and molecular mechanisms that regulate coronary arterio-venous patterning are not known in detail. In this study, we have used the avian embryo to dissect the ontogenetic origin and morphogenesis of coronary vasculature. RESULTS: We show that sinus venosus endocardial sprouts and proepicardial angioblasts pioneer coronary vascular formation, invading the developing heart simultaneously. We also report that avian ventricular endocardium has the potential to contribute to coronary vessels, and describe the incorporation of cardiac distal outflow tract endothelial cells to the peritruncal endothelial plexus to participate in coronary vascular formation. Finally, our findings indicate that large sinus venosus-independent sections of the forming coronary vasculature develop without connection to the systemic circulation and that coronary arterio-venous shunts form a few hours before peritruncal arterial endothelium connects to the aortic root. CONCLUSIONS: Embryonic coronary vasculature is a developmental mosaic, formed by the integration of vascular cells from, at least, four different embryological origins, which assemble in a coordinated manner to complete coronary vascular development. Developmental Dynamics 247:686-698, 2018. © 2017 Wiley Periodicals, Inc.


Subject(s)
Endothelial Cells/cytology , Endothelium, Vascular/embryology , Myocardium/cytology , Animals , Chick Embryo , Coronary Vessels/embryology , Coronary Vessels/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Immunohistochemistry , In Situ Hybridization , Myocardium/metabolism , Quail
12.
Stem Cell Reports ; 9(6): 1754-1764, 2017 12 12.
Article in English | MEDLINE | ID: mdl-29173898

ABSTRACT

Human pluripotent stem cells (hPSCs) are widely used to study cardiovascular cell differentiation and function. Here, we induced differentiation of hPSCs (both embryonic and induced) to proepicardial/epicardial progenitor cells that cover the heart during development. Addition of retinoic acid (RA) and bone morphogenetic protein 4 (BMP4) promoted expression of the mesodermal marker PDGFRα, upregulated characteristic (pro)epicardial progenitor cell genes, and downregulated transcription of myocardial genes. We confirmed the (pro)epicardial-like properties of these cells using in vitro co-culture assays and in ovo grafting of hPSC-epicardial cells into chick embryos. Our data show that RA + BMP4-treated hPSCs differentiate into (pro)epicardial-like cells displaying functional properties (adhesion and spreading over the myocardium) of their in vivo counterpart. The results extend evidence that hPSCs are an excellent model to study (pro)epicardial differentiation into cardiovascular cells in human development and evaluate their potential for cardiac regeneration.


Subject(s)
Cell Differentiation/genetics , Embryonic Development/genetics , Heart/growth & development , Induced Pluripotent Stem Cells/cytology , Animals , Bone Morphogenetic Protein 4/administration & dosage , Cardiovascular System/cytology , Cardiovascular System/growth & development , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Culture Techniques/methods , Cell Differentiation/drug effects , Chick Embryo , Gene Expression Regulation, Developmental/drug effects , Heart/drug effects , Humans , Induced Pluripotent Stem Cells/drug effects , Myocardium/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Pericardium/cytology , Pericardium/growth & development , Receptor, Platelet-Derived Growth Factor alpha/genetics , Stem Cells/cytology , Tretinoin/administration & dosage
13.
Proc Natl Acad Sci U S A ; 113(46): E7250-E7259, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27799555

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS) is a rare genetic disease caused by defective prelamin A processing, leading to nuclear lamina alterations, severe cardiovascular pathology, and premature death. Prelamin A alterations also occur in physiological aging. It remains unknown how defective prelamin A processing affects the cardiac rhythm. We show age-dependent cardiac repolarization abnormalities in HGPS patients that are also present in the Zmpste24-/- mouse model of HGPS. Challenge of Zmpste24-/- mice with the ß-adrenergic agonist isoproterenol did not trigger ventricular arrhythmia but caused bradycardia-related premature ventricular complexes and slow-rate polymorphic ventricular rhythms during recovery. Patch-clamping in Zmpste24-/- cardiomyocytes revealed prolonged calcium-transient duration and reduced sarcoplasmic reticulum calcium loading and release, consistent with the absence of isoproterenol-induced ventricular arrhythmia. Zmpste24-/- progeroid mice also developed severe fibrosis-unrelated bradycardia and PQ interval and QRS complex prolongation. These conduction defects were accompanied by overt mislocalization of the gap junction protein connexin43 (Cx43). Remarkably, Cx43 mislocalization was also evident in autopsied left ventricle tissue from HGPS patients, suggesting intercellular connectivity alterations at late stages of the disease. The similarities between HGPS patients and progeroid mice reported here strongly suggest that defective cardiac repolarization and cardiomyocyte connectivity are important abnormalities in the HGPS pathogenesis that increase the risk of arrhythmia and premature death.


Subject(s)
Arrhythmias, Cardiac/physiopathology , Cardiac Conduction System Disease/physiopathology , Progeria/physiopathology , Adolescent , Adult , Animals , Arrhythmias, Cardiac/metabolism , Calcium/physiology , Cardiac Conduction System Disease/metabolism , Child , Child, Preschool , Connexin 43/metabolism , Connexin 43/physiology , Female , Heart/physiology , Humans , Male , Membrane Proteins/genetics , Membrane Proteins/physiology , Metalloendopeptidases/genetics , Metalloendopeptidases/physiology , Mice, Inbred C57BL , Mice, Knockout , Myocardium/metabolism , Nuclear Lamina/physiology , Progeria/metabolism , Sarcoplasmic Reticulum/physiology , Young Adult
14.
Proc Natl Acad Sci U S A ; 113(3): 656-61, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26739565

ABSTRACT

Recent reports suggest that mammalian embryonic coronary endothelium (CoE) originates from the sinus venosus and ventricular endocardium. However, the contribution of extracardiac cells to CoE is thought to be minor and nonsignificant for coronary formation. Using classic (Wt1(Cre)) and previously undescribed (G2-Gata4(Cre)) transgenic mouse models for the study of coronary vascular development, we show that extracardiac septum transversum/proepicardium (ST/PE)-derived endothelial cells are required for the formation of ventricular coronary arterio-venous vascular connections. Our results indicate that at least 20% of embryonic coronary arterial and capillary endothelial cells derive from the ST/PE compartment. Moreover, we show that conditional deletion of the ST/PE lineage-specific Wilms' tumor suppressor gene (Wt1) in the ST/PE of G2-Gata4(Cre) mice and in the endothelium of Tie2(Cre) mice disrupts embryonic coronary transmural patterning, leading to embryonic death. Taken together, our results demonstrate that ST/PE-derived endothelial cells contribute significantly to and are required for proper coronary vascular morphogenesis.


Subject(s)
Coronary Vessels/embryology , Embryo, Mammalian/cytology , Endothelial Cells/cytology , Heart Septum/cytology , Pericardium/cytology , Animals , Biomarkers/metabolism , Cell Lineage , Coronary Vessels/cytology , Embryonic Development , Enhancer Elements, Genetic/genetics , Epithelial-Mesenchymal Transition , GATA4 Transcription Factor/metabolism , Gene Deletion , Genes, Reporter , Green Fluorescent Proteins/metabolism , Integrases/metabolism , Mice , Models, Biological , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Phenotype , WT1 Proteins/metabolism
15.
Differentiation ; 91(4-5): 72-7, 2016.
Article in English | MEDLINE | ID: mdl-26558986

ABSTRACT

Tissue ablation is a classic experimental approach to study early embryo patterning. However, ablation methods are less frequently used to assess the reparative or regenerative properties of embryonic tissues during organogenesis. Surgical procedures based on the removal of a significant amount of tissue during organ formation very much depend on the skills of the researcher, are difficult to reproduce, and often result in extensive tissue disruption leading to embryonic death. In this paper, we present a new protocol to generate discrete, locally-restricted and highly reproducible wounds in the developing chick embryo using a liquid N2-cooled metallic probe. This in ovo procedure allows for the study of organ-specific tissue responses to damage, such as compensatory cell growth, cell differentiation, and reparative/regenerative mechanisms throughout the embryonic lifespan.


Subject(s)
Cell Differentiation/genetics , Chick Embryo/growth & development , Embryonic Development/genetics , Organogenesis/genetics , Animals , Wound Healing
16.
J Am Coll Cardiol ; 65(19): 2057-66, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25975467

ABSTRACT

BACKGROUND: Although efforts continue to find new therapies to regenerate infarcted heart tissue, knowledge of the cellular and molecular mechanisms involved remains poor. OBJECTIVES: This study sought to identify the origin of cardiac fibroblasts (CFs) in the infarcted heart to better understand the pathophysiology of ventricular remodeling following myocardial infarction (MI). METHODS: Permanent genetic tracing of epicardium-derived cell (EPDC) and bone marrow-derived blood cell (BMC) lineages was established using Cre/LoxP technology. In vivo gene and protein expression studies, as well as in vitro cell culture assays, were developed to characterize EPDC and BMC interaction and properties. RESULTS: EPDCs, which colonize the cardiac interstitium during embryogenesis, massively differentiate into CFs after MI. This response is disease-specific, because angiotensin II-induced pressure overload does not trigger significant EPDC fibroblastic differentiation. The expansion of epicardial-derived CFs follows BMC infiltration into the infarct site; the number of EPDCs equals that of BMCs 1 week post-infarction. BMC-EPDC interaction leads to cell polarization, packing, massive collagen deposition, and scar formation. Moreover, epicardium-derived CFs display stromal properties with respect to BMCs, contributing to the sustained recruitment of circulating cells to the damaged zone and the cardiac persistence of hematopoietic progenitors/stem cells after MI. CONCLUSIONS: EPDCs, but not BMCs, are the main origin of CFs in the ischemic heart. Adult resident EPDC contribution to the CF compartment is time- and disease-dependent. Our findings are relevant to the understanding of post-MI ventricular remodeling and may contribute to the development of new therapies to treat this disease.


Subject(s)
Bone Marrow Cells/pathology , Cicatrix/pathology , Fibroblasts/pathology , Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , Ventricular Remodeling , Animals , Cell Differentiation , Cells, Cultured , Cicatrix/etiology , Cicatrix/physiopathology , Disease Models, Animal , Mice , Mice, Inbred C57BL , Myocardial Infarction/complications , Myocardial Infarction/physiopathology , Pericardium/pathology
17.
PLoS One ; 8(1): e53694, 2013.
Article in English | MEDLINE | ID: mdl-23349729

ABSTRACT

The non-muscular cells that populate the space found between cardiomyocyte fibers are known as 'cardiac interstitial cells' (CICs). CICs are heterogeneous in nature and include different cardiac progenitor/stem cells, cardiac fibroblasts and other cell types. Upon heart damage CICs soon respond by initiating a reparative response that transforms with time into extensive fibrosis and heart failure. Despite the biomedical relevance of CICs, controversy remains on the ontogenetic relationship existing between the different cell kinds homing at the cardiac interstitium, as well as on the molecular signals that regulate their differentiation, maturation, mutual interaction and role in adult cardiac homeostasis and disease. Our work focuses on the analysis of epicardial-derived cells, the first cell type that colonizes the cardiac interstitium. We present here a characterization and an experimental analysis of the differentiation potential and mobilization properties of a new cell line derived from mouse embryonic epicardium (EPIC). Our results indicate that these cells express some markers associated with cardiovascular stemness and retain part of the multipotent properties of embryonic epicardial derivatives, spontaneously differentiating into smooth muscle, and fibroblast/myofibroblast-like cells. Epicardium-derived cells are also shown to initiate a characteristic response to different growth factors, to display a characteristic proteolytic expression profile and to degrade biological matrices in 3D in vitro assays. Taken together, these data indicate that EPICs are relevant to the analysis of epicardial-derived CICs, and are a god model for the research on cardiac fibroblasts and the role these cells play in ventricular remodeling in both ischemic or non/ischemic myocardial disease.


Subject(s)
Cell Differentiation , Cell Movement , Embryonic Stem Cells/cytology , Myofibroblasts/cytology , Pericardium/cytology , Animals , Biomarkers/metabolism , Cell Line , Mice , Mice, Inbred C57BL , Multipotent Stem Cells/cytology , Pericardium/embryology , Pericardium/metabolism , Proteolysis , Stem Cell Niche
18.
Curr Opin Pediatr ; 24(5): 569-76, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22890066

ABSTRACT

PURPOSE OF REVIEW: In this review, we aim at presenting and discussing the cellular and molecular mechanisms of embryonic epicardial development that may underlie the origin of congenital heart disease (CHD). RECENT FINDINGS: New discoveries on the multiple cell lineages that form part of the original pool of epicardial progenitors and the roles played by epicardial transcription factors and morphogens in the regulation of epicardial epithelial-to-mesenchymal transition, epicardial-derived cell (EPDCs) differentiation, coronary blood vessel morphogenesis and cardiac interstitium formation are presented in a comprehensive manner. SUMMARY: We have provided evidence on the critical participation of epicardial cells and EPDCs in normal and abnormal cardiac development, suggesting the implication of defective epicardial development in various forms of CHD.


Subject(s)
Epithelial-Mesenchymal Transition , Heart Defects, Congenital/metabolism , Pericardium/cytology , Stem Cells/cytology , Animals , Cell Differentiation , Heart Defects, Congenital/embryology , Humans , Myocardium/cytology , Pericardium/embryology
19.
Dev Biol ; 366(2): 111-24, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22546693

ABSTRACT

The importance of the epicardium for myocardial and valvuloseptal development has been well established; perturbation of epicardial development results in cardiac abnormalities, including thinning of the ventricular myocardial wall and malformations of the atrioventricular valvuloseptal complex. To determine the spatiotemporal contribution of epicardially derived cells to the developing fibroblast population in the heart, we have used a mWt1/IRES/GFP-Cre mouse to trace the fate of EPDCs from embryonic day (ED)10 until birth. EPDCs begin to populate the compact ventricular myocardium around ED12. The migration of epicardially derived fibroblasts toward the interface between compact and trabecular myocardium is completed around ED14. Remarkably, epicardially derived fibroblasts do not migrate into the trabecular myocardium until after ED17. Migration of EPDCs into the atrioventricular cushion mesenchyme commences around ED12. As development progresses, the number of EPDCs increases significantly, specifically in the leaflets which derive from the lateral atrioventricular cushions. In these developing leaflets the epicardially derived fibroblasts eventually largely replace the endocardially derived cells. Importantly, the contribution of EPDCs to the leaflets derived from the major AV cushions is very limited. The differential contribution of EPDCs to the various leaflets of the atrioventricular valves provides a new paradigm in valve development and could lead to new insights into the pathogenesis of abnormalities that preferentially affect individual components of this region of the heart. The notion that there is a significant difference in the contribution of epicardially and endocardially derived cells to the individual leaflets of the atrioventricular valves has also important pragmatic consequences for the use of endocardial and epicardial cre-mouse models in studies of heart development.


Subject(s)
Fibroblasts/cytology , Heart Valves/embryology , Heart/embryology , Pericardium/cytology , Animals , Embryonic Development , Heart Valves/cytology , Heart Ventricles/cytology , Heart Ventricles/embryology , Mice , Organogenesis
20.
PLoS One ; 5(11): e15071, 2010 Nov 30.
Article in English | MEDLINE | ID: mdl-21151498

ABSTRACT

BACKGROUND: Mast cell secretory granules accommodate a large number of components, many of which interact with highly sulfated serglycin proteoglycan (PG) present within the granules. Polyamines (putrescine, spermidine and spermine) are absolutely required for the survival of the vast majority of living cells. Given the reported ability of polyamines to interact with PGs, we investigated the possibility that polyamines may be components of mast cell secretory granules. METHODOLOGY/PRINCIPAL FINDINGS: Spermidine was released by mouse bone marrow derived mast cells (BMMCs) after degranulation induced by IgE/anti-IgE or calcium ionophore A23187. Additionally, both spermidine and spermine were detected in isolated mouse mast cell granules. Further, depletion of polyamines by culturing BMMCs with α-difluoromethylornithine (DFMO) caused aberrant secretory granule ultrastructure, impaired histamine storage, reduced serotonin levels and increased ß-hexosaminidase content. A proteomic approach revealed that DFMO-induced polyamine depletion caused an alteration in the levels of a number of proteins, many of which are connected either with the regulated exocytosis or with the endocytic system. CONCLUSIONS/SIGNIFICANCE: Taken together, our results show evidence that polyamines are present in mast cell secretory granules and, furthermore, indicate an essential role of these polycations during the biogenesis and homeostasis of these organelles.


Subject(s)
Homeostasis , Mast Cells/metabolism , Polyamines/metabolism , Secretory Vesicles/metabolism , Animals , Blotting, Western , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Marrow Cells/physiology , Calcimycin/pharmacology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Degranulation , Cells, Cultured , Electrophoresis, Gel, Two-Dimensional , Female , Ionophores/pharmacology , Mast Cells/cytology , Mast Cells/physiology , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Perilipin-3 , Proteomics/methods , Reverse Transcriptase Polymerase Chain Reaction , Secretory Vesicles/drug effects , Secretory Vesicles/ultrastructure , Spermidine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...