Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Front Cell Dev Biol ; 10: 864765, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35706902

RESUMO

Background: The cardiac interstitial cellular fraction is composed of multiple cell types. Some of these cells are known to express some well-known stem cell markers such as c-Kit and Sca1, but they are no longer accepted to be true cardiac stem cells. Although their existence in the cardiac interstitium has not been disputed, their dynamic throughout development, specific embryonic origin, and potential heterogeneity remain unknown. In this study, we hypothesized that both c-KitPOS and Sca1POS cardiac interstitial cell (CIC) subpopulations are related to the Wilms' tumor 1 (Wt1) epicardial lineage. Methods: In this study, we have used genetic cell lineage tracing methods, immunohistochemistry, and FACS techniques to characterize cardiac c-KitPOS and Sca1POS cells. Results: Our data show that approximately 50% of cardiac c-KitPOS cells are derived from the Wt1-lineage at E15.5. This subpopulation decreased along with embryonic development, disappearing from P7 onwards. We found that a large proportion of cardiac c-KitPOS cells express specific markers strongly suggesting they are blood-borne cells. On the contrary, the percentage of Sca1POS cells within the Wt1-lineage increases postnatally. In accordance with these findings, 90% of adult epicardial-derived endothelial cells and 60% of mEFSK4POS cardiac fibroblasts expressed Sca1. Conclusion: Our study revealed a minor contribution of the Wt1-epicardial lineage to c-KitPOS CIC from embryonic stages to adulthood. Remarkably, a major part of the adult epicardial-derived cell fraction is enriched in Sca1, suggesting that this subpopulation of CICs is heterogeneous from their embryonic origin. The study of this heterogeneity can be instrumental to the development of diagnostic and prognostic tests for the evaluation of cardiac homeostasis and cardiac interstitium response to pathologic stimuli.

2.
J Cell Mol Med ; 15(5): 1200-9, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-20477904

RESUMO

The peritoneal mesothelium exhibits a high regenerative ability. Peritoneal regeneration is concomitant with the appearance, in the coelomic cavity, of a free-floating population of cells whose origin and functions are still under discussion. We have isolated and characterized this cell population and we have studied the process of mesothelial regeneration through flow cytometry and confocal microscopy in a murine model lethally irradiated and reconstituted with GFP-expressing bone marrow cells. In unoperated control mice, most free cells positive for mesothelin, a mesothelial marker, are green fluorescent protein (GFP). However, 24 hrs after peritoneal damage, free mesothelin(+)/GFP(+) cells appear in peritoneal lavages. Cultured lavage peritoneal cells show colocalization of GFP with mesothelial (mesothelin, cytokeratin) and fibroblastic markers. Immunohistochemical staining of the peritoneal wall also revealed colocalization of GFP with mesothelial markers and with procollagen-1 and smooth muscle α-actin. This was observed in the injured area as well as in the surrounding not-injured peritoneal surfaces. These cells, which we herein call peritoneal repairing cells (PRC), are very abundant 1 week after surgery covering both the damaged peritoneal wall and the surrounding uninjured area. However, they become very scarce 1 month later, when the mesothelium has completely healed. We suggest that PRC constitute a type of monocyte-derived cells, closely related with the tissue-repairing cells known as 'fibrocytes' and specifically involved in peritoneal reparation. Thus, our results constitute a synthesis of the different scenarios hitherto proposed about peritoneal regeneration, particularly recruitment of circulating progenitor cells and adhesion of free-floating coelomic cells.


Assuntos
Células da Medula Óssea/citologia , Peritônio/fisiologia , Regeneração , Células-Tronco/citologia , Actinas/biossíntese , Animais , Células da Medula Óssea/metabolismo , Células Cultivadas , Colágeno Tipo I/biossíntese , Epitélio/fisiologia , Queratinas/biossíntese , Mesotelina , Camundongos , Monócitos , Lavagem Peritoneal , Peritônio/citologia , Pró-Colágeno/biossíntese , Coloração e Rotulagem , Células-Tronco/metabolismo
3.
Artigo em Inglês | MEDLINE | ID: mdl-20884305

RESUMO

Nitric oxide synthases (NOS), the enzymes responsible for the NO synthesis, are present in all eukaryotes. Three isoforms (neuronal, inducible and endothelial), encoded by different loci, have been described in vertebrates, although the endothelial isoform seems to be restricted to tetrapods. In invertebrates, a variety of NOS isoforms have been variably annotated as "inducible" or "neuronal", while others lack precise annotation. We have performed an exhaustive collection of the available NOS amino-acid sequences in order to perform a phylogenetic analysis. We hypothesized that the NOS isoforms reported in vertebrates derive from 1) different invertebrate NOS, 2) a single invertebrate ancestral gene, through an event related to the double whole genomic duplication that occurred at the origin of vertebrates, and 3) the endothelial form of NOS appeared late in the evolution of vertebrates, after the split of tetrapods and fishes. Our molecular evolution analysis strongly supports the second scenario, the three vertebrate NOS isoforms derived from a single ancestral invertebrate gene. Thus, the diverse NOS isoforms in invertebrates can be explained by events of gene duplication, but their characterization as "inducible" or "neuronal" should only be justified by physiological features, since they are evolutionarily unrelated to the homonym isoforms of vertebrates.


Assuntos
Evolução Molecular , Invertebrados/genética , Óxido Nítrico Sintase/genética , Vertebrados/genética , Sequência de Aminoácidos , Animais , Biologia Computacional/métodos , Bases de Dados Genéticas , Bases de Dados de Proteínas , Duplicação Gênica , Genoma , Invertebrados/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo I , Óxido Nítrico Sintase Tipo II , Óxido Nítrico Sintase Tipo III , Filogenia , Estrutura Terciária de Proteína/genética , Alinhamento de Sequência , Vertebrados/metabolismo
4.
J Cell Mol Med ; 14(8): 2066-72, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20477903

RESUMO

The epicardium has recently been identified as an active and essential element of cardiac development. Recent reports have unveiled a variety of functions performed by the embryonic epicardium, as well as the cellular and molecular mechanisms regulating them. However, despite its developmental importance, a number of unsolved issues related to embryonic epicardial biology persist. In this review, we will summarize our current knowledge about (i) the ontogeny and evolution of the epicardium, including a discussion on the evolutionary origins of the proepicardium (the epicardial primordium), (ii) the nature of epicardial-myocardial interactions during development, known to be essential for myocardial growth and maturation, and (iii) the contribution of epicardially derived cells to the vascular and connective tissue of the heart. We will finish with a note on the relationships existing between the primordia of the viscera and their coelomic epithelial lining. We would like to suggest that at least a part of the properties of the embryonic epicardium are shared by many other coelomic cell types, such that the role of epicardium in cardiac development is a particular example of a more general mechanism for the contribution of coelomic and coelomic-derived cells to the morphogenesis of organs such as the liver, kidneys, gonads or spleen.


Assuntos
Mesoderma/embriologia , Organogênese , Pericárdio/embriologia , Animais , Diferenciação Celular , Proliferação de Células , Humanos , Mesoderma/citologia , Modelos Cardiovasculares , Pericárdio/citologia
5.
Dev Biol ; 312(1): 157-70, 2007 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18028902

RESUMO

Previous studies of knock-out mouse embryos have shown that the Wilms' tumor suppressor gene (Wt1) is indispensable for the development of kidneys, gonads, heart, adrenals and spleen. Using OPT (Optical Projection Tomography) we have found a new role for Wt1 in mouse liver development. In the absence of Wt1, the liver is reduced in size, and shows lobing abnormalities. In normal embryos, coelomic cells expressing Wt1, GATA-4, RALDH2 and RXRalpha delaminate from the surface of the liver, intermingle with the hepatoblasts and incorporate to the sinusoidal walls. Some of these cells express desmin, suggesting a contribution to the stellate cell population. Other cells, keeping high levels of RXRalpha immunoreactivity, are negative for stellate or smooth muscle cell markers. However, coelomic cells lining the liver of Wt1-null embryos show decreased or absent RALDH2 expression, the population of cells expressing high levels of RXRalpha is much reduced and the proliferation of hepatoblasts and RXRalpha-positive cells is significantly decreased. On the other hand, the expression of smooth muscle cell specific alpha-actin increases throughout the liver, suggesting an accelerated and probably anomalous differentiation of stellate cell progenitors. We describe a similar retardation of liver growth in RXRalpha-null mice as well as in chick embryos after inhibition of retinoic acid synthesis. We propose that Wt1 expression in cells delaminating from the coelomic epithelium is essential for the expansion of the progenitor population of liver stellate cells and for liver morphogenesis. Mechanistically, at least part of this effect is mediated via the retinoic acid signaling pathway.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Hepatócitos/citologia , Fígado/embriologia , Morfogênese/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Tretinoína/farmacologia , Proteínas WT1/metabolismo , Monoterpenos Acíclicos , Aldeído Oxirredutases/antagonistas & inibidores , Animais , Biomarcadores/metabolismo , Proliferação de Células/efeitos dos fármacos , Embrião de Galinha , Embrião de Mamíferos/citologia , Embrião de Mamíferos/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Hepatócitos/enzimologia , Fígado/efeitos dos fármacos , Fígado/enzimologia , Fígado/patologia , Camundongos , Camundongos Knockout , Modelos Biológicos , Monoterpenos/farmacologia , Fenótipo , Codorniz , Receptor X Retinoide alfa/metabolismo , Estômago/anatomia & histologia , Estômago/efeitos dos fármacos , Proteínas WT1/deficiência
6.
J Microsc ; 225(Pt 1): 96-9, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17286699

RESUMO

Colocalization of fluorescent signals in confocal microscopy is usually evaluated by inspecting merged images from different colour channels or by using commercially available software packages. We describe in this paper a simple method for assessment of nuclear localization of proteins in tissue sections through confocal immunolocalization, propidium iodide counterstaining and image analysis. Through a macro command developed for the public domain, Java-based software imagej, red, green, blue (RGB) images are automatically split in the red and green channels and a new image composed of the nonblack pixels coincident in both channels is created and inverted for better visualization. This method renders images devoid of both, extranuclear staining and background, thus emphasizing the nuclear signal. The resulting images can easily be used for comparison or quantification of the results. Given the simplicity of the technique and the worldwide diffusion of the software utilized, we think that this method could be useful in order to define standards of colocalization in confocal microscopy.


Assuntos
Processamento de Imagem Assistida por Computador/métodos , Imuno-Histoquímica/métodos , Proteínas Nucleares/metabolismo , Animais , Anticorpos Monoclonais , Embrião de Galinha , Endocárdio/metabolismo , Fator 1 Nuclear de Hepatócito/metabolismo , Camundongos , Microscopia Confocal/métodos , Antígeno Nuclear de Célula em Proliferação/metabolismo , Fatores de Transcrição da Família Snail , Fatores de Transcrição/metabolismo , beta Catenina/metabolismo
7.
Evol Dev ; 7(4): 351-8, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15982372

RESUMO

Circulatory systems of vertebrate and invertebrate metazoans are very different. Large vessels of invertebrates are constituted of spaces and lacunae located between the basement membranes of endodermal and mesodermal epithelia, and they lack an endothelial lining. Myoepithelial differentation of the coelomic cells covering hemal spaces is a frequent event, and myoepithelial cells often form microvessels in some large invertebrates. There is no phylogenetic theory about the origin of the endothelial cells in vertebrates. We herein propose that endothelial cells originated from a type of specialized blood cells, called amoebocytes, that adhere to the vascular basement membrane. The transition between amoebocytes and endothelium involved the acquisition of an epithelial phenotype. We suggest that immunological cooperation was the earliest function of these protoendothelial cells. Furthermore, their ability to transiently recover the migratory, invasive phenotype of amoebocytes (i.e., the angiogenic phenotype) allowed for vascular growth from the original visceral areas to the well-developed somatic areas of vertebrates (especially the tail, head, and neural tube). We also hypothesize that pericytes and smooth muscle cells derived from myoepithelial cells detached from the coelomic lining. As the origin of blood cells in invertebrates is probably coelomic, our hypothesis relates the origin of all the elements of the circulatory system with the coelomic wall. We have collected from the literature a number of comparative and developmental data supporting our hypothesis, for example the localization of the vascular endothelial growth factor receptor-2 ortholog in hemocytes of Drosophila or the fact that circulating progenitors can differentiate into endothelial cells even in adult vertebrates.


Assuntos
Endotélio Vascular/fisiologia , Invertebrados/fisiologia , Músculo Liso Vascular/fisiologia , Sistema Nervoso Simpático/fisiologia , Vertebrados/fisiologia , Animais , Endotélio Vascular/inervação , Humanos , Músculo Liso Vascular/inervação
8.
Cell Mol Life Sci ; 61(17): 2224-43, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15338053

RESUMO

Endothelial cells receive multiple information from their environment that eventually leads them to progress along all the stages of the process of formation of new vessels. Angiogenic signals promote endothelial cell proliferation, increased resistance to apoptosis, changes in proteolytic balance, cytoskeletal reorganization, migration and, finally, differentiation and formation of a new vascular lumen. We aim to review herein the main signaling cascades that become activated in angiogenic endothelial cells as well as the opportunities of modulating angiogenesis through pharmacological interference with these signaling mechanisms. We will deal mainly with the mitogen-activated protein kinases pathway, which is very important in the transduction of proliferation signals; the phosphatidylinositol-3-kinase/protein kinase B signaling system, particularly essential for the survival of the angiogenic endothelium; the small GTPases involved in cytoskeletal reorganization and migration; and the kinases associated to focal adhesions which contribute to integrate the pathways from the two main sources of angiogenic signals, i.e. growth factors and the extracellular matrix.


Assuntos
Células Endoteliais/fisiologia , Neovascularização Fisiológica , Transdução de Sinais , Animais , Diferenciação Celular , Movimento Celular , Sobrevivência Celular , Receptores ErbB/fisiologia , Matriz Extracelular/metabolismo , GTP Fosfo-Hidrolases/fisiologia , Humanos , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Proto-Oncogênicas c-met/fisiologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia
9.
Dev Dyn ; 229(3): 465-74, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14991702

RESUMO

The developing liver is vascularized through a complex process of vasculogenesis that leads to the differentiation of the sinusoids. The main structural elements of the sinusoidal wall are endothelial and stellate (Ito) cells. We have studied the differentiation of the hepatic sinusoids in avian embryos through confocal colocalization of differentiation markers, in ovo direct labeling of the liver mesothelium, induced invasion of the developing chick liver by quail proepicardial cells, and in vitro culture of chimeric aggregates. Our results show that liver mesothelial cells give rise to mesenchymal cells which intermingle between the growing hepatoblast cords and become incorporated to the sinusoidal wall, contributing to both endothelial and stellate cell populations. We have also shown that the proepicardium, a mesothelial tissue anatomically continuous with liver mesothelium, is able to form sinusoid-like vessels into the hepatic primordium as well as in cultured aggregates of hepatoblasts. Thus, both intrinsic or extrinsic mesothelium-derived cells have the developmental potential to contribute to the establishment of liver sinusoids.


Assuntos
Epitélio/metabolismo , Fígado/embriologia , Animais , Aves , Diferenciação Celular , Embrião de Galinha , Técnicas de Cocultura , Biologia do Desenvolvimento , Embrião não Mamífero , Corantes Fluorescentes/farmacologia , Imuno-Histoquímica , Fígado/citologia , Fígado/metabolismo , Modelos Biológicos , Pericárdio/citologia , Fatores de Tempo
10.
J Mol Cell Cardiol ; 35(7): 795-802, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12818570

RESUMO

Transposition of great arteries in humans is associated with a wide spectrum of coronary artery patterns. However, no information is available about how this pattern diversity develops. We have studied the development of the coronary arteries in mouse embryos with a targeted mutation of perlecan, a mutation that leads to ventriculo-arterial discordance and complete transposition in about 70% of the embryos. The perlecan-deficient embryos bearing complete transposition showed a coronary artery pattern consisting of right and left coronary arteries arising from the morphologically dorsal and ventral sinuses of Valsalva, respectively. The left coronary artery gives rise to a large septal artery and runs along the ventral margin of the pulmonary root. In the earliest embryos where transposition could be confirmed (12.5 d post coitum), a dense subepicardial vascular plexus is located in this ventral margin. In wild-type mice, however, capillaries are very scarce on the ventral surface of the pulmonary root and the left coronary artery runs dorsally to this root. We suggest that the establishment of the diverse coronary artery patterns is determined by the anatomical arrangement and the capillary density of the peritruncal vascular plexus, a plexus that spreads from the atrio-ventricular groove and grows around the aortic or pulmonary roots depending on the degree of the short-axis aortopulmonary rotation. This simple model, based on very few assumptions, might explain all the observed variation of the coronary artery patterns in humans with transposition, as well as our observations on the perlecan-deficient and the normal mice.


Assuntos
Vasos Coronários/embriologia , Modelos Animais de Doenças , Transposição dos Grandes Vasos/embriologia , Animais , Vasos Coronários/anatomia & histologia , Embrião de Mamíferos/anormalidades , Camundongos
11.
Dev Biol ; 247(2): 307-26, 2002 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-12086469

RESUMO

Epicardially derived cells (EPDCs) delaminate from the primitive epicardium through an epithelial-to-mesenchymal transformation (EMT). After this transformation, a subpopulation of cells progressively invades myocardial and valvuloseptal tissues. The first aim of the study was to determine the tissue-specific distribution of two molecules that are thought to play a crucial function in the interaction between EPDCs and other cardiac tissues, namely the Wilms' Tumor transcription factor (WT1) and retinaldehyde-dehydrogenase2 (RALDH2). This study was performed in normal avian and in quail-to-chick chimeric embryos. It was found that EPDCs that maintain the expression of WT1 and RALDH2 initially populate the subepicardial space and subsequently invade the ventricular myocardium. As EPDCs differentiate into the smooth muscle and endothelial cell lineage of the coronary vessels, the expression of WT1 and RALDH2 becomes downregulated. This process is accompanied by the upregulation of lineage-specific markers. We also observed EPDCs that continued to express WT1 (but very little RALDH2) which did not contribute to the formation of the coronary system. A subset of these cells eventually migrates into the atrioventricular (AV) cushions, at which point they no longer express WT1. The WT1/RALDH2-negative EPDCs in the AV cushions do, however, express the smooth muscle cell marker caldesmon. The second aim of this study was to determine the impact of abnormal epicardial growth on cardiac development. Experimental delay of epicardial growth distorted normal epicardial development, reduced the number of invasive WT1/RALDH2-positive EPDCs, and provoked anomalies in the coronary vessels, the ventricular myocardium, and the AV cushions. We suggest that the proper development of ventricular myocardium is dependent on the invasion of undifferentiated, WT1-positive, retinoic acid-synthesizing EPDCs. Furthermore, we propose that an interaction between EPDCs and endocardial (derived) cells is imperative for correct development of the AV cushions.


Assuntos
Aldeído Oxirredutases/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Coração/embriologia , Pericárdio/metabolismo , Proteínas WT1/biossíntese , Animais , Diferenciação Celular , Linhagem da Célula , Embrião de Galinha , Quimera/metabolismo , Imuno-Histoquímica , Queratinas/metabolismo , Modelos Biológicos , Miocárdio/metabolismo , Fenótipo , Codorniz , Retinal Desidrogenase , Fatores de Tempo
12.
Ital J Anat Embryol ; 106(2 Suppl 1): 187-96, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11729954

RESUMO

The primitive epicardium of the vertebrate embryo has traditionally been regarded as a rather passive mesothelium, lining the embryonic myocardium and forming the adult visceral pericardium. However, in recent years, there is an increasing evidence that the primitive epicardium is a highly dynamic element which supplies cells to the developing heart through a process of epithelial-mesenchymal transition. This process seems to be more active at the atrioventricular canal and outflow tract, i.e. the cardiac segments where the endothelium transforms into mesenchyme. In this paper we review the current evidence which supports such epicardial-mesenchymal transition, namely: 1) morphological features, 2) colocalization of cytokeratin and vimentin in the epicardial and subepicardial mesenchymal cells, 3) presence of common antigens in the transforming epicardium and endocardial cushions (fibrillin-2/JB3, ES/130, Ets-1). Recendy, we have immunolocated the transcription factor Slug in the developing avian heart. Slug is a zinc-finger protein involved in the formation of the neural crest, a developmental event which implies an epithelial-mesenchymal transition. All cells of the primitive epicardium are Slug+ from their differentiation until the stage HH24. However, only a fraction of the endothelial cells from the endocardial cushions are Slug+. We speculate that the expression of Slug marks competence of the epicardial cells to transform into mesenchyme, although this transformation is only achieved where an inducing signal is produced. Regarding the developmental fate of the epicardial-derived cell population, there is strong evidence of its differentiation in fibroblasts and vascular smooth muscle cells, although a contribution to the coronary endothelium cannot be discarded.


Assuntos
Proteínas Aviárias , Diferenciação Celular/fisiologia , Epitélio/embriologia , Coração/embriologia , Mesoderma/citologia , Miocárdio/citologia , Pericárdio/embriologia , Fatores de Transcrição/metabolismo , Animais , Embrião de Galinha , Quimera/embriologia , Quimera/metabolismo , Cação (Peixe)/embriologia , Cação (Peixe)/metabolismo , Epitélio/metabolismo , Epitélio/ultraestrutura , Proteínas da Matriz Extracelular/metabolismo , Fibrilinas , Coração/fisiologia , Imuno-Histoquímica , Queratinas/metabolismo , Mesoderma/metabolismo , Proteínas dos Microfilamentos/metabolismo , Miocárdio/metabolismo , Pericárdio/citologia , Pericárdio/metabolismo , Codorniz/embriologia , Codorniz/metabolismo , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética , Vimentina/metabolismo
13.
Cells Tissues Organs ; 169(2): 89-103, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11399849

RESUMO

Questions on the embryonic origin and developmental significance of the epicardium did not receive much recognition for more than a century. It was generally thought that the epicardium was derived from the outermost layer of the primitive myocardium of the early embryonic heart tube. During the past few years, however, there has been an increasing interest in the development of the epicardium. This was caused by a series of new embryological data. The first data showed that the epicardium did not derive from the primitive myocardium but from a primarily extracardiac primordium, called the proepicardial serosa. Subsequent data then suggested that the proepicardial serosa and the newly formed epicardium provided nearly all cellular elements of the subepicardial and intermyocardial connective tissue, and of the coronary vasculature. Recent data even suggest important modulatory roles of the epicardium and of other proepicardium-derived cells in the differentiation of the embryonic myocardium and cardiac conduction system. The present paper reviews our current knowledge on the origin and embryonic development of the epicardium.


Assuntos
Coração/embriologia , Coração/fisiologia , Miocárdio/metabolismo , Pericárdio/embriologia , Pericárdio/fisiologia , Animais , Linhagem da Célula , Embrião de Galinha , Humanos , Miocárdio/patologia , Pericárdio/patologia , Fatores de Transcrição/metabolismo
14.
Cell Tissue Res ; 303(2): 173-86, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11291764

RESUMO

The Wilms' tumour suppressor gene WT1 encodes a zinc-finger transcription factor which is essential for the development of kidney, gonads, spleen and adrenals. WT1-null embryos lack all of these viscerae and they also show a thin ventricular myocardium and unexpectedly die from cardiac failure between 13 and 15 days post coitum. We studied the localization of the WT1 protein in chick and quail embryos between stages HH18 and HH35. In early embryos, WT1 protein was located in specific areas of the coelomic mesothelium adjacent to the nephric ducts, the myocardium or the primordia of the endodermal organs (gut, liver and lungs). These mesothelial areas also showed localized expression of Slug, a zinc-finger transcription factor involved in epithelial-mesenchymal transitions. WT1+ mesenchymal cells were always found below the immunoreactive mesothelial areas, either forming a narrow band on the surface of the endodermal organs (gut, liver and lungs) or migrating throughout the mesodermal organs (mesonephros, metanephros, gonads, spleen and heart). In the developing heart, the invasion of WTI+ cells started at stage HH26, and all the ventricular myocardium was pervaded by these cells, presumably derived from the epicardium, at HH30. We suggest that WT1 is not required for the epithelial-mesenchymal transition of the coelomic mesothelium, but it might be a marker of the mesothelial-derived cells, where this protein would be acting as a repressor of the differentiation.


Assuntos
Proteínas de Ligação a DNA/análise , Mesonefro/química , Pericárdio/química , Fatores de Transcrição/análise , Animais , Especificidade de Anticorpos , Embrião de Galinha , Galinhas , Proteínas de Ligação a DNA/imunologia , Epitélio/química , Feto/química , Feto/embriologia , Gônadas/química , Gônadas/embriologia , Mesonefro/embriologia , Pericárdio/embriologia , Codorniz , Fatores de Transcrição da Família Snail , Baço/química , Baço/embriologia , Fatores de Transcrição/imunologia , Proteínas WT1
15.
FEBS Lett ; 477(3): 237-43, 2000 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-10908727

RESUMO

The major isoenzyme of glutamine synthetase found in leaves of angiosperms is the chloroplastic form. However, pine seedlings contain two cytosolic glutamine synthetases in green cotyledons: GS1a, the predominant isoform, and GS1b, a minor enzyme whose relative amount is increased following phosphinotricin treatment. We have cloned a GS1b cDNA, and comparison with the previously reported GS1a cDNA sequence indicated that they correspond to separate cytosolic GS genes encoding distinct protein products. Phylogenetic analysis showed that the newly reported sequence is closer to cytosolic angiosperm GS than to GS1a, suggesting therefore that GS1a could be a divergent gymnospermous GS1 gene. Gene mapping using a F2 family of maritime pine showed co-localization of both GS genes on group 2 of the genetic linkage map. This result supports the proposed origin of different members of the GS1 family by adjacent gene duplication. The implications for gymnosperm genome organization are discussed.


Assuntos
Citosol/enzimologia , Genoma de Planta , Glutamato-Amônia Ligase/genética , Isoenzimas/genética , Árvores/genética , Sequência de Bases , DNA Complementar , Dados de Sequência Molecular , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência do Ácido Nucleico
16.
Anat Embryol (Berl) ; 201(2): 103-9, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10672362

RESUMO

Slug is a transcription factor involved in processes such as the formation of mesoderm and neural crest, two developmental events that imply a transition from an epithelial to a mesenchymal phenotype. During late cardiac morphogenesis, mesenchymal cells originate from two epithelia--epicardial mesothelium and cushion endocardium. We aimed to check if Slug is expressed in these systems of epithelial-mesenchymal transition. We have immuno-located the Slug protein in the heart of quail embryos between Hamburger and Hamilton stages HH16 and HH30. In the proepicardium (the epicardial primordium), Slug was detected in most cells, mesothelial as well as mesenchymal. Slug immunoreactivity was strong in the mesenchyme of the endocardial cushions and subepicardium from its inception until HH24, but the immunoreactivity disappeared in later embryos. Only a small portion of the endocardial cells located in the areas of epithelial-mesenchymal transition (atrioventricular groove and outflow tract) were immuno-labelled, mainly between HH16 and HH20. Endocardial cells from other cardiac segments were always negative, except for a transient, weak immunoreactivity that coincided with the development of the intertrabecular sinusoids of the ventricle. In contrast, virtually all cells of the epicardial mesothelium were immunoreactive until stage HH24. The mesenchymal cells that migrate to the heart through the spina vestibuli were also conspicuously immunoreactive. The myocardium was not labelled in the stages studied. Our results stress the involvement of Slug in the epithelial to mesenchymal transition. We suggest that Slug can constitute a reliable marker of the cardiac epithelial cells that are competent to transform into mesenchyme as well as a transient marker of the epithelial-derived mesenchymal cells in the developing heart.


Assuntos
Coturnix/embriologia , Coração/embriologia , Miocárdio/química , Fatores de Transcrição/análise , Animais , Anticorpos Monoclonais , Endocárdio/química , Endocárdio/embriologia , Fatores de Transcrição da Família Snail , Fatores de Tempo , Dedos de Zinco
17.
Differentiation ; 64(3): 133-41, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10234810

RESUMO

The existence of the hemangioblast, a common progenitor of the endothelial and hematopoietic cell lineages, was proposed at the beginning of the century. Although recent findings seem to confirm its existence, it is still unknown when and how the hemangioblasts differentiate. We propose a hypothesis about the origin of hemangioblasts from the embryonic splanchnic mesothelium. The model is based on observations collected from the literature and from our own studies. These observations include: (1) the extensive population of the splanchnic mesoderm by mesothelial-derived cells coinciding with the emergence of the endothelial and hematopoietic progenitors; (2) the transient localization of cytokeratin, the main mesothelial intermediate filament protein, in some embryonic vessels and endothelial progenitors; (3) the possible origin of cardiac vessels from epicardial-derived cells; (4) the origin of endocardial cells from the splanchnic mesoderm when this mesoderm is an epithelium; (5) the evidence that mesothelial cells migrate to the hemogenic areas of the dorsal aorta. (6) Biochemical and antigenic similarities between mesothelial and endothelial cells. We suggest that the endothelium-lined vascular system arose as a specialization of the phylogenetically older coelomic cavities. The origin of the hematopoietic cells might be related to the differentiation, reported in some invertebrates, of coelomocytes from the coelomic epithelium. Some types of coelomocytes react against microbial invasion and other types transport respiratory pigments. We propose that this phylogenetic origin is recapitulated in the vertebrate ontogeny and explains the differentiation of endothelial and blood cells from a common mesothelial-derived progenitor.


Assuntos
Sistema Cardiovascular/embriologia , Células Epiteliais/fisiologia , Epitélio/embriologia , Células-Tronco Hematopoéticas/fisiologia , Modelos Biológicos , Vertebrados/embriologia , Animais , Aorta/citologia , Aorta/embriologia , Vasos Sanguíneos/citologia , Vasos Sanguíneos/embriologia , Sistema Cardiovascular/citologia , Diferenciação Celular/fisiologia , Embrião de Galinha , Cricetinae , Indução Embrionária , Endotélio Vascular/citologia , Endotélio Vascular/embriologia , Células-Tronco Hematopoéticas/citologia , Queratinas/imunologia , Mesoderma/citologia , Filogenia
18.
Histochem J ; 31(12): 771-9, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10661320

RESUMO

We report morphological and immunohistochemical evidence for a translocation of cells from the coelomic mesothelium to the aortic wall between the developmental stages HH16 and HH22 of the quail embryos. The coelomic mesothelial cells closest to the aorta showed, at these stages, increased mitotic activity, reduced intercellular adhesion, loss of tight junctions, and long basal cytoplasmic processes. Coinciding with these morphological traits, cytokeratin immunoreactivity was found in the mesothelium, in cells of the aortic wall and throughout the ventral periaortic mesenchyme (but not in the lateral and dorsal aortic regions). Vimentin immunoreactivity colocalized with cytokeratin in the mesothelial cells adjacent to the aorta. In the ventral aortic wall, cytokeratin colocalized with smooth muscle alpha-actin and with the 1E12 antigen (a smooth muscle-specific alpha-actinin isoform). We think that the morphological and immunolocalization data observed are compatible with an epithelial-mesenchymal transition by which mesothelial-derived cells contribute to the splanchnic mesoderm and aortic wall. The precise coincidence between the mesothelial contribution and the emergence of the aortic smooth muscle cells progenitors, as well as the immunolocalization data, suggest a potential relationship of the mesothelial-derived cells with this cell lineage. This may explain the observed ventrodorsal asymmetry in the distribution of smooth muscle cells progenitors in the aortic wall.


Assuntos
Aorta/citologia , Embrião não Mamífero/irrigação sanguínea , Endotélio Vascular/citologia , Células Epiteliais/citologia , Codorniz/embriologia , Actinas/metabolismo , Animais , Aorta/embriologia , Aorta/metabolismo , Movimento Celular , Endotélio Vascular/embriologia , Endotélio Vascular/metabolismo , Células Epiteliais/metabolismo , Queratinas/metabolismo , Músculo Liso Vascular/citologia , Músculo Liso Vascular/embriologia , Músculo Liso Vascular/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Vimentina/metabolismo
19.
Histochem J ; 30(9): 627-34, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9870763

RESUMO

The earliest evidence of the development of the cardiac vessels in mammals is the emergence of subepicardial blood islands, which are thought to originate from mesenchymal progenitors. In order to identify these progenitor cells, we have studied the immunohistochemical localization in the heart of Syrian hamster embryos of the type 2 vascular endothelial growth factor receptor, the earliest molecule known to be expressed in the vasculogenic cell lineage. Only a few immunoreactive subepicardial mesenchymal cells were present by 10 days post coitum. By 11 days post coitum, the subepicardial mesenchymal cells became abundant at the dorsal part of the ventricle, the atrioventricular and the conoventricular grooves. About 20% of cells were labelled with the antibody. Immunoreactive cells were isolated or formed pairs, short cords, rounded clusters or ring-like structures at the subepicardium or, occasionally, within the ventricular myocardium. Other labelled cells were simultaneously cytokeratin immunoreactive. By 12 days post coitum, most immunoreactive mesenchymal cells have been replaced by a capillary network. We propose that an active process of vascular differentiation occurs between 10 and 12 days post coitum in the subepicardium of this species, and it might be a suitable model for the study of vasculogenetic mechanisms.


Assuntos
Vasos Coronários/citologia , Mesoderma/citologia , Receptores Proteína Tirosina Quinases/análise , Receptores de Fatores de Crescimento/análise , Células-Tronco , Animais , Vasos Coronários/embriologia , Cricetinae , Mesocricetus/embriologia , Receptores de Fatores de Crescimento do Endotélio Vascular , Células-Tronco/química
20.
Anat Embryol (Berl) ; 198(4): 307-15, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9764544

RESUMO

Cardiac morphogenesis involves substantial remodeling processes that include cell transdifferentiation and migration. The c-ets-1 protooncogene codes for a transcription factor that can transactivate a number of genes involved in developmental processes such as degradation of extracellular matrices and cell migration. We have immunolocated the ets-1 protein in the heart of quail and chick embryos between the Hamburger and Hamilton stages HH16 and HH37. In HH16-17 embryos, the ets-1 transcription factor was only detected in some endocardial cells and in most mesothelial and mesenchymal cells of the proepicardium. Ets-1 immunoreactivity increased markedly in the developing endocardial cushions, myocardium, epicardium and early subepicardial mesenchyme of HH18-19 embryos. By HH20-24 the immunoreactivity was found throughout the heart, with a stronger intensity in the areas of epithelial-mesenchymal transition of the endocardium and epicardium. In embryos between HH26 and HH33, ets-1 immunoreactivity increased in the cushion mesenchyme, atrioventricular endocardium, ventricular epicardium and subepicardial mesenchyme cells, but not in other areas of the heart. The immunoreactivity declined in the innermost part of the endocardial cushions. The subepicardial mesenchyme was particularly immunoreactive in these stages, coinciding with the development of the subepicardial vascular network. In fact, ets-1 colocalized with the quail vascular marker QH1 in the subepicardial mesenchymal cells. Ets-1-negative cells were abundant in the subepicardium and valvuloseptal tissue of the HH37 embryos. The results suggest that ets-1, probably through transactivation of genes such as urokinase-type plasminogen activator and matrix metalloproteinases, might play a crucial role in the differentiation of the cushion and subepicardial mesenchyme, the formation of the intratrabecular sinusoids and the early development of the cardiac vessels.


Assuntos
Células Epiteliais/fisiologia , Coração/embriologia , Mesoderma/fisiologia , Miocárdio/química , Proteínas Proto-Oncogênicas/análise , Fatores de Transcrição/análise , Animais , Embrião de Galinha , Coturnix , Células Epiteliais/química , Técnica Indireta de Fluorescência para Anticorpo , Coração/fisiologia , Mesoderma/química , Microscopia Confocal , Proteína Proto-Oncogênica c-ets-1 , Proteínas Proto-Oncogênicas c-ets , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA