246 results on '"Embryo, Mammalian blood supply"'
Search Results
152. Transient bowel ischaemia of the fetus.
- Author
-
Ghi T, Tani G, Carletti A, Basile B, Rizzo N, Pilu G, and Bovicelli L
- Subjects
- Embryo, Mammalian blood supply, Female, Humans, Infant, Newborn, Ischemia diagnosis, Magnetic Resonance Imaging, Prenatal Diagnosis, Ultrasonography, Prenatal, Intestines embryology, Ischemia embryology, Ischemia physiopathology, Recovery of Function
- Abstract
Objective: To discover the different underlying conditions in 2 fetuses suffering from temporary bowel ischaemia., Methods: Abnormal bowel findings were detected using antenatal sonography., Results: The abnormal bowel findings disappeared postnatally. Transient ischaemia of the fetal bowel due to different causes has been advocated antenatally to explain the abnormal findings. When a normal blood supply to the bowel has been restored, either in utero or after birth, the abnormal findings disappear., Conclusions: Whenever gut dilatation is detected in a fetus at risk of bowel ischaemia the possibility of a transient functional finding must be considered.
- Published
- 2005
- Full Text
- View/download PDF
153. Functional significance of developmental changes in placental microvascular architecture.
- Author
-
Reynolds LP, Biondini ME, Borowicz PP, Vonnahme KA, Caton JS, Grazul-Bilska AT, and Redmer DA
- Subjects
- Animals, Capillaries cytology, Capillaries physiology, Embryo, Mammalian cytology, Embryo, Mammalian physiology, Female, Placenta cytology, Placenta physiology, Sheep, Embryo, Mammalian blood supply, Placenta blood supply, Pregnancy physiology, Pregnancy, Animal physiology
- Published
- 2005
- Full Text
- View/download PDF
154. Role of oxygen and vascular development in epithelial branching morphogenesis of the developing mouse lung.
- Author
-
van Tuyl M, Liu J, Wang J, Kuliszewski M, Tibboel D, and Post M
- Subjects
- Animals, Embryo, Mammalian blood supply, Embryo, Mammalian metabolism, Embryonic Development physiology, Epithelium embryology, Mice, Mice, Transgenic, Organ Culture Techniques, Platelet Endothelial Cell Adhesion Molecule-1 metabolism, Vascular Endothelial Growth Factor A metabolism, Blood Vessels embryology, Lung embryology, Morphogenesis physiology, Oxygen physiology
- Abstract
Recent investigations have suggested an active role for endothelial cells in organ development, including the lung. Herein, we investigated some of the molecular mechanisms underlying normal pulmonary vascular development and their influence on epithelial branching morphogenesis. Because the lung in utero develops in a relative hypoxic environment, we first investigated the influence of low oxygen on epithelial and vascular branching morphogenesis. Two transgenic mouse models, the C101-LacZ (epithelial-LacZ marker) and the Tie2-LacZ (endothelial-LacZ marker), were used. At embryonic day 11.5, primitive lung buds were dissected and cultured at either 20 or 3% oxygen. At 24-h intervals, epithelial and endothelial LacZ gene expression was visualized by X-galactosidase staining. The rate of branching of both tissue elements was increased in explants cultured at 3% oxygen compared with 20% oxygen. Low oxygen increased expression of VEGF, but not that of the VEGF receptor (Flk-1). Expression of two crucial epithelial branching factors, fibroblast growth factor-10 and bone morphogenetic protein-4, were not affected by low oxygen. Epithelial differentiation was maintained at low oxygen as shown by surfactant protein C in situ hybridization. To explore epithelial-vascular interactions, we inhibited vascular development with antisense oligonucleotides targeted against either hypoxia inducible factor-1 alpha or VEGF. Epithelial branching morphogenesis in vitro was dramatically abrogated when pulmonary vascular development was inhibited. Collectively, the in vitro data show that a low-oxygen environment enhances branching of both distal lung epithelium and vascular tissue and that pulmonary vascular development appears to be rate limiting for epithelial branching morphogenesis.
- Published
- 2005
- Full Text
- View/download PDF
155. Distribution patterns of the anti-angiogenic protein ADAMTS-1 during rat development.
- Author
-
Günther W, Skaftnesmo KO, Arnold H, Bjerkvig R, and Terzis AJ
- Subjects
- ADAM Proteins chemistry, ADAM Proteins genetics, ADAMTS1 Protein, Amino Acid Sequence, Animals, Blotting, Western, Cell Line, Embryo, Mammalian blood supply, Humans, Immunohistochemistry, Molecular Sequence Data, RNA, Messenger genetics, Rats, ADAM Proteins metabolism, Embryo, Mammalian embryology, Embryo, Mammalian metabolism, Embryonic Development genetics, Gene Expression Regulation, Developmental
- Abstract
A disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS-1) belongs to the large ADAM family of proteins. ADAMTS-1 contains a metalloproteinase domain, a disintegrin domain and three thrombospondin-like repeats but unlike ADAMs lacks a transmembrane domain. For the elucidation of the biological functions of ADAMTS-1, we raised new antibodies against ADAMTS-1. We show an accumulation of ADAMTS-1 protein at the basal lamina of rat embryonal epithelia of intestines, nasal cavity, choroid plexus, skin and in intracellular storage vesicles of epithelial cells. ADAMTS-1 protein seems to play a role in the development of the neuronal system, adipose tissue, muscle, heart, liver and adrenal glands. At the time of birth its presence is reduced in most organs. However, in the developing bone as well as in the skin, labelling increased towards late embryonal development. Immunoblots revealed a strong presence of a 62 kDa ADAMTS-1 fragment in kidneys, adrenal glands, lungs, intestines and heart. ADAMTS-1 was also present in the corresponding adult rat organs, but in more restricted distribution patterns. It was typically found in principal cells of collecting ducts, of the renal medulla, in ependymal cells of the ventricles and in some neurons. The results were confirmed by real-time PCR. The specific distribution pattern of ADAMTS-1 in a variety of organs during embryogenesis suggests a role of the molecule in tissue remodelling, vasculogenesis and angiogenesis.
- Published
- 2005
- Full Text
- View/download PDF
156. Three-dimensional porous alginate scaffolds provide a conducive environment for generation of well-vascularized embryoid bodies from human embryonic stem cells.
- Author
-
Gerecht-Nir S, Cohen S, Ziskind A, and Itskovitz-Eldor J
- Subjects
- Bioartificial Organs, Biocompatible Materials chemistry, Cell Differentiation, Cell Line, Cell Proliferation, Cell Size, Cell Survival physiology, Humans, Materials Testing, Membranes, Artificial, Neovascularization, Physiologic physiology, Porosity, Alginates chemistry, Cell Culture Techniques methods, Embryo, Mammalian blood supply, Embryo, Mammalian cytology, Embryonic Development physiology, Glucuronic Acid chemistry, Hexuronic Acids chemistry, Stem Cells cytology, Stem Cells physiology, Tissue Engineering methods
- Abstract
Differentiation of human embryonic stem cells (hESCs) can be instigated through the formation of embryo-like aggregates in suspension, termed human embryoid bodies (hEBs). Controlling cell aggregation and agglomeration during hEBs formation has a profound effect on the extent of cell proliferation and differentiation. In a previous work, we showed that control over hEBs formation and differentiation can be achieved via cultivation of hESC suspensions in a rotating bioreactor system. We now report that hEBs can be generated directly from hESC suspensions within three-dimensional (3D) porous alginate scaffolds. The confining environments of the alginate scaffold pores enabled efficient formation of hEBs with a relatively high degree of cell proliferation and differentiation; encouraged round, small-sized hEBs; and induced vasculogenesis in the forming hEBs to a greater extent than in static or rotating cultures. We therefore conclude that differentiation of hEBs can be induced and directed by physical constraints in addition to chemical cues., (Copyright 2004 Wiley Periodicals, Inc.)
- Published
- 2004
- Full Text
- View/download PDF
157. Anti-angiogenic effect of pyridoxal 5'-phosphate, pyridoxal and pyridoxamine on embryoid bodies derived from mouse embryonic stem cells.
- Author
-
Matsubara K, Mori M, Akagi R, and Kato N
- Subjects
- Animals, Embryo, Mammalian blood supply, Hematopoietic Stem Cells drug effects, Mice, Neovascularization, Pathologic prevention & control, Neovascularization, Physiologic drug effects, Angiogenesis Inhibitors pharmacology, Hematopoietic Stem Cells physiology, Pyridoxal pharmacology, Pyridoxal Phosphate pharmacology, Pyridoxamine pharmacology
- Abstract
We previously demonstrated anti-angiogenic activity of pyridoxal 5'-phosphate (PLP) and pyridoxal (PL) using in vitro and ex vivo angiogenesis models and anti-colon tumor effect of vitamin B(6) in mice. There is growing evidence that endothelial progenitor cells (EPCs) in circulation contribute to tumor-induced angiogenesis. It is of importance to clarify whether EPC differentiation is involved in the mechanism of anti-angiogenic effect of vitamin B(6). In this study, we investigated the effect of vitamin B(6) on angiogenesis in embryonic stem cell-derived embryoid bodies (EBs). PLP suppressed angiogenesis in EBs at >or=100 microM. Among the vitamin B(6) compounds at the concentration of 200 microM, PL, as well as PLP, was the most effective suppressor of vasculogenesis, while pyridoxine was inactive. These results were consistent with their effects in rat aorta and endothelial cell assays. Interestingly, pyridoxamine (PM), which had no effect in rat aorta and endothelial cell assays, also exerted a significant suppressive effect in this model. This study demonstrated an inhibitory effect of PM on vasculogenesis in this EB model as well as PLP and PL, and suggests that suppression of EPC differentiation is at least in part responsible for the mechanism of the anti-angiogenesis effect of vitamin B(6).
- Published
- 2004
158. The hemangioblast: cradle to clinic.
- Author
-
Cogle CR and Scott EW
- Subjects
- Animals, Blood Vessels embryology, Blood Vessels growth & development, Embryo, Mammalian blood supply, Embryo, Mammalian cytology, Endothelium, Vascular cytology, Hematopoietic Stem Cell Transplantation, Hematopoietic Stem Cells cytology, Humans, Neovascularization, Physiologic, Blood Vessels cytology, Mesoderm cytology, Stem Cells cytology
- Abstract
In the embryo, the mesodermal precursor cell, the hemangioblast, gives rise to blood and blood vessels. During adult life, the hematopoietic stem cell (HSC) also exhibits this bipotential hemangioblast activity, serving as a rich source for circulating endothelial progenitor cells (EPCs). As a result of this finding, many questions have arisen as to whether the adult HSC is involved in the day-to-day maintenance of tissues, what mechanisms influence this adult hemangioblast activity, and whether blood vessels harbor hematopoietic capability. In answering these questions, the power of adult hemangioblast activity could be harnessed to evaluate and treat diseases such as myocardial infarction, stroke, cancer, and blindness. Enumeration of activated EPCs aims to alert the patient as to the severity of their disease, predict response to therapy, and gauge for relapse potential. Identification of hemangioblast stimulatory or inhibitory cues would allow physicians to regulate neovascularization in their patient, augmenting vessel production in situations of hypo-proliferation such as wound healing and inhibiting vessel production in situations of hyper-proliferation such as cancer. Finally, given that EPCs home to sites of new blood vessel growth, genetic engineering of harvested HSC or EPC offers the potential to deliver vasoregulatory factors directly to sites of neovascularization.
- Published
- 2004
- Full Text
- View/download PDF
159. Identification of genes involved in VEGF-mediated vascular morphogenesis using embryonic stem cell-derived cystic embryoid bodies.
- Author
-
Ng YS, Ramsauer M, Loureiro RM, and D'Amore PA
- Subjects
- Animals, Caveolin 1, Caveolins genetics, Caveolins metabolism, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Cells, Cultured, Embryo, Mammalian blood supply, Embryonic and Fetal Development, Immunohistochemistry, Mice, Mice, Knockout, Models, Animal, Oligonucleotide Array Sequence Analysis, RNA, Messenger analysis, RNA, Messenger biosynthesis, Reverse Transcriptase Polymerase Chain Reaction, Vascular Endothelial Growth Factor A deficiency, Vascular Endothelial Growth Factor A metabolism, Embryo, Mammalian physiology, Gene Expression Regulation, Developmental, Neovascularization, Physiologic genetics, Stem Cells physiology, Vascular Endothelial Growth Factor A genetics
- Abstract
The vasculature forms during development via two processes, vasculogenesis and angiogenesis, in which vessels form de novo from angioblast precursors or as sprouts from pre-existing vessels, respectively. A common and critical aspect of both processes is vascular morphogenesis, which includes branching of endothelial cell cords and lumen formation. Although ample evidence support the central role of vascular endothelial growth factor (VEGF) in both vasculogenesis and angiogenesis, the role of VEGF in vascular morphogenesis is unclear and little is known about the regulation of vascular morphogenesis, in general. We have used the in vitro vessel differentiation system of embryonic stem (ES) cell-derived cystic embryonic bodies (CEB) as a model for studying VEGF-mediated vessel formation. Whereas CEB formed from wild-type ES cells make well-formed vessel-like structures, CEB derived from VEGF-null ES cells contain PECAM-1-positive endothelial cells, but these cells do not participate in vascular morphogenesis. Using gene expression microarray analysis to compare gene expression in these two systems, we have been able to identify many genes and novel ESTs that are downstream of VEGF function, and which may be involved in VEGF-mediated vascular morphogenesis including caveolin-1 and HEY-1. These results support using the CEB model, in combination with gene knockout ES cells, for studying vascular morphogenesis.
- Published
- 2004
- Full Text
- View/download PDF
160. Choroideremia gene product affects trophoblast development and vascularization in mouse extra-embryonic tissues.
- Author
-
Shi W, van den Hurk JA, Alamo-Bethencourt V, Mayer W, Winkens HJ, Ropers HH, Cremers FP, and Fundele R
- Subjects
- Alkyl and Aryl Transferases metabolism, Animals, Blood Vessels physiology, Breeding, Eye pathology, Female, Fetal Death genetics, Male, Mice, Mice, Inbred C57BL, Mice, Mutant Strains, Placenta pathology, Polyploidy, Pregnancy, X Chromosome, Y Chromosome, Yolk Sac blood supply, Yolk Sac pathology, Alkyl and Aryl Transferases genetics, Blood Vessels embryology, Embryo, Mammalian blood supply, Gene Expression Regulation, Developmental, Trophoblasts physiology
- Abstract
Choroideremia (CHM) is a hereditary eye disease caused by mutations in the X-linked CHM gene. Disruption of the Chm gene in mice resulted in prenatal death of Chm-/Y males and Chm-/Chm+ females that had inherited the mutation from their mothers. Male chimeras and Chm+/Chm- females with paternal transmission of the mutation were viable and had photoreceptor degeneration reminiscent of human choroideremia. Here, we show that Chm-/Y males and Chm-/Chm+ females were retarded at e7.5 and died before e11.5 due to multiple defects of the extra-embryonic tissues. Mutant embryos exhibited deficiency of diploid trophoblasts associated with overabundance of giant cells. In yolk sac and placenta, severe defects in vasculogenesis were obvious. Chm-/Y males exhibited more pronounced phenotypes than Chm-/Chm+ females. The lethal genotypes could be rescued by tetraploid aggregation. Chm-/Chm+ females, but not Chm-/Y males, could also be rescued when their Chm+/Chm- mothers were mated with Mus spretus males. Backcross analysis suggested that the viability of interspecies hybrid Chm-/Chm+ females may be due to expression from the Chm allele on the M. spretus X-chromosome rather than a modifier effect. Our results demonstrate that Chm is essential for diploid trophoblast development and plays a role in the vascularization in placenta and yolk sac.
- Published
- 2004
- Full Text
- View/download PDF
161. Yolk sac-derived primitive erythroblasts enucleate during mammalian embryogenesis.
- Author
-
Kingsley PD, Malik J, Fantauzzo KA, and Palis J
- Subjects
- Animals, Cell Count methods, Cell Differentiation, Cell Size, Embryo, Mammalian blood supply, Erythroblasts cytology, Erythroblasts ultrastructure, Erythrocytes cytology, Erythropoiesis physiology, Female, Gene Expression, Globins biosynthesis, Globins chemistry, Globins immunology, Immunohistochemistry, Microscopy, Phase-Contrast, Pregnancy, Yolk Sac cytology, Erythroblasts physiology, Mice embryology, Yolk Sac physiology
- Abstract
The enucleated definitive erythrocytes of mammals are unique in the animal kingdom. The observation that yolk sac-derived primitive erythroid cells in mammals circulate as nucleated cells has led to the conjecture that they are related to the red cells of fish, amphibians, and birds that remain nucleated throughout their life span. In mice, primitive red cells express both embryonic and adult hemoglobins, whereas definitive erythroblasts accumulate only adult hemoglobins. We investigated the terminal differentiation of murine primitive red cells with use of antibodies raised to embryonic beta H1-globin. Primitive erythroblasts progressively enucleate between embryonic days 12.5 and 16.5, generating mature primitive erythrocytes that are similar in size to their nucleated counterparts. These enucleated primitive erythrocytes circulate as late as 5 days after birth. The enucleation of primitive red cells in the mouse embryo has not previously been well recognized because it coincides with the emergence of exponentially expanding numbers of definitive erythrocytes from the fetal liver. Our studies establish a new paradigm in the understanding of primitive erythropoiesis and support the concept that primitive erythropoiesis in mice shares many similarities with definitive erythropoiesis of mammals.
- Published
- 2004
- Full Text
- View/download PDF
162. Egfl7, a novel epidermal growth factor-domain gene expressed in endothelial cells.
- Author
-
Fitch MJ, Campagnolo L, Kuhnert F, and Stuhlmann H
- Subjects
- Aging physiology, Amino Acid Sequence, Animals, Calcium-Binding Proteins, Cell Line, Cloning, Molecular, DNA, Complementary genetics, EGF Family of Proteins, Embryo, Mammalian blood supply, Embryo, Mammalian cytology, Embryo, Mammalian embryology, Embryo, Mammalian metabolism, Embryonic Development, Endoplasmic Reticulum metabolism, Endothelial Cells cytology, Gene Expression Profiling, Golgi Apparatus metabolism, Humans, Mesoderm metabolism, Mice, Molecular Sequence Data, Proteins chemistry, Proteins metabolism, Sequence Alignment, Yolk Sac metabolism, Endothelial Cells metabolism, Gene Expression Regulation, Proteins genetics
- Abstract
We report the cloning and characterization of a novel epidermal growth factor (EGF) domain gene that was identified in a retroviral gene entrapment screen and is expressed in endothelial cells. This gene encodes a protein of 278 amino acids with an amino-terminal signal peptide and two centrally located EGF-like domains. We have named this novel gene in accordance with the guidelines of the Mouse Genome Informatics group Egfl7, for EGF-like domain 7. Egfl7 mRNA is expressed in highly vascularized adult tissues such as the lung, heart, uterus, and ovary. In addition, Egfl7 is expressed early during mouse embryogenesis and in undifferentiated murine embryonic stem cells. The analysis of Egfl7 expression in embryonic day 9.5 embryos by in situ hybridization indicates that Egfl7 is expressed in vascular structures in both the embryo proper and the yolk sac and at sites of mesodermal precursors of angioblasts. Within the cell, EGFL7 protein is localized to the endoplasmic reticulum and Golgi apparatus, suggesting that the protein is targeted for secretion. Indeed, recombinant EGFL7 is readily detectable in the supernatant media of transiently transfected HEK293 cells. We also report the identification of an Egfl7 paralog, Egfl8, and show that EGFL8 protein shares similar domains and molecular weight with EGFL7., (Copyright 2004 Wiley-Liss, Inc.)
- Published
- 2004
- Full Text
- View/download PDF
163. The endothelial-cell-derived secreted factor Egfl7 regulates vascular tube formation.
- Author
-
Parker LH, Schmidt M, Jin SW, Gray AM, Beis D, Pham T, Frantz G, Palmieri S, Hillan K, Stainier DY, De Sauvage FJ, and Ye W
- Subjects
- Animals, Blood Vessels cytology, Calcium-Binding Proteins, Cell Adhesion, Cell Count, EGF Family of Proteins, Embryo, Mammalian abnormalities, Embryo, Mammalian cytology, Embryo, Nonmammalian abnormalities, Embryo, Nonmammalian blood supply, Embryo, Nonmammalian cytology, Endothelial Cells cytology, In Situ Hybridization, Mice, Oligonucleotides, Antisense genetics, Oligonucleotides, Antisense metabolism, Proteins genetics, RNA, Messenger genetics, RNA, Messenger metabolism, Zebrafish abnormalities, Zebrafish genetics, Zebrafish Proteins genetics, Blood Vessels embryology, Embryo, Mammalian blood supply, Endothelial Cells metabolism, Proteins metabolism, Zebrafish embryology, Zebrafish Proteins metabolism
- Abstract
Vascular development is a complex but orderly process that is tightly regulated. A number of secreted factors produced by surrounding cells regulate endothelial cell (EC) differentiation, proliferation, migration and coalescence into cord-like structures. Vascular cords then undergo tubulogenesis to form vessels with a central lumen. But little is known about how tubulogenesis is regulated in vivo. Here we report the identification and characterization of a new EC-derived secreted factor, EGF-like domain 7 (Egfl7). Egfl7 is expressed at high levels in the vasculature associated with tissue proliferation, and is downregulated in most of the mature vessels in normal adult tissues. Loss of Egfl7 function in zebrafish embryos specifically blocks vascular tubulogenesis. We uncover a dynamic process during which gradual separation and proper spatial arrangement of the angioblasts allow subsequent assembly of vascular tubes. This process fails to take place in Egfl7 knockdown embryos, leading to the failure of vascular tube formation. Our study defines a regulator that controls a specific and important step in vasculogenesis.
- Published
- 2004
- Full Text
- View/download PDF
164. Kidney, blood, and endothelium: developmental expression of stem cell leukemia during nephrogenesis.
- Author
-
Dekel B, Hochman E, Sanchez MJ, Maharshak N, Amariglio N, Green AR, and Izraeli S
- Subjects
- Aging metabolism, Animals, Animals, Newborn, Basic Helix-Loop-Helix Transcription Factors, Cell Line, Chlorocebus aethiops, DNA-Binding Proteins blood, Dogs, Embryo, Mammalian blood supply, Embryo, Mammalian metabolism, Embryonic and Fetal Development, Humans, Immunohistochemistry, Leukemia genetics, Mice, Mice, Inbred BALB C, Mice, Transgenic, Proto-Oncogene Proteins blood, RNA, Messenger metabolism, T-Cell Acute Lymphocytic Leukemia Protein 1, Transcription Factors blood, DNA-Binding Proteins metabolism, Endothelium, Vascular embryology, Fetal Blood, Kidney embryology, Proto-Oncogene Proteins metabolism, Transcription Factors metabolism
- Abstract
Background: In vertebrates the hematopoietic and renal tissues share a common mesodermal origin. Recently, we have analyzed global gene expression during human nephrogenesis and observed up-regulation of stem cell leukemia (SCL), a transcription factor critical for hematopoietic and endothelial lineage specification. Here we characterize the expression of SCL along with its distinct 3' hematopoietic and endothelial enhancer (SCL 3'En) during kidney development., Methods: mRNA and protein expression of SCL were examined in developing murine and human kidneys by quantitative reverse transcription-polymerase chain reaction (RT-PCR) and immunohistochemistry. The activity of SCL 3'En was examined by X-galactosidase (X-gal) staining of embryonic kidneys obtained from SCL +6E5/lacZ/3'En transgenic mice and by reporter lacZ assay in various renal cell lines., Results: We found developmental regulation of SCL mRNA with highest levels of expression in embryonic day 17 (E17) mouse kidneys and lowest in postnatal and adult kidneys. Immunostaining of human fetal kidneys demonstrated the protein predominantly in the nephrogenic cortex and particularly in mesenchymal cells and developing glomeruli. Similarly, SCL +6E5/lacZ/3'En transgenic kidneys showed prominent lacZ staining in cells resembling undifferentiated mesoderm cells in close proximity to S and comma-shaped primitive nephrons and in peritubular and glomerular vessel endothelium. The SCL 3'En was activated in the human embryonic kidney cell line (HEK 293), but not in cell lines derived from adult kidney., Conclusion: These observations suggest a possible role for SCL in renal vasculogenesis. Undifferentiated mesenchymal cells expressing SCL during early nephrogenesis might represent putative progenitors that can simultaneously give rise to kidney, blood, and endothelium.
- Published
- 2004
- Full Text
- View/download PDF
165. Hematopoietic, angiogenic and eye defects in Meis1 mutant animals.
- Author
-
Hisa T, Spence SE, Rachel RA, Fujita M, Nakamura T, Ward JM, Devor-Henneman DE, Saiki Y, Kutsuna H, Tessarollo L, Jenkins NA, and Copeland NG
- Subjects
- Animals, Cell Transplantation, Embryo, Mammalian blood supply, Embryo, Mammalian cytology, Erythroid Precursor Cells cytology, Female, Fetus cytology, Gene Targeting, Germ-Line Mutation, Hemorrhage etiology, Homeodomain Proteins genetics, Liver cytology, Liver embryology, Megakaryocytes cytology, Mice, Mice, Inbred C57BL, Mice, Knockout, Myeloid Ecotropic Viral Integration Site 1 Protein, Myeloid Progenitor Cells cytology, Neoplasm Proteins genetics, Eye Abnormalities etiology, Hematopoiesis, Homeodomain Proteins physiology, Neoplasm Proteins physiology, Neovascularization, Pathologic etiology
- Abstract
Meis1 and Hoxa9 expression is upregulated by retroviral integration in murine myeloid leukemias and in human leukemias carrying MLL translocations. Both genes also cooperate to induce leukemia in a mouse leukemia acceleration assay, which can be explained, in part, by their physical interaction with each other as well as the PBX family of homeodomain proteins. Here we show that Meis1-deficient embryos have partially duplicated retinas and smaller lenses than normal. They also fail to produce megakaryocytes, display extensive hemorrhaging, and die by embryonic day 14.5. In addition, Meis1-deficient embryos lack well-formed capillaries, although larger blood vessels are normal. Definitive myeloerythroid lineages are present in the mutant embryos, but the total numbers of colony-forming cells are dramatically reduced. Mutant fetal liver cells also fail to radioprotect lethally irradiated animals and they compete poorly in repopulation assays even though they can repopulate all hematopoietic lineages. These and other studies showing that Meis1 is expressed at high levels in hematopoietic stem cells (HSCs) suggest that Meis1 may also be required for the proliferation/self-renewal of the HSC.
- Published
- 2004
- Full Text
- View/download PDF
166. Mathematical modelling of tumour-induced angiogenesis: network growth and structure.
- Author
-
Chaplain M and Anderson A
- Subjects
- Animals, Cell Division, Cell Movement, Chemotaxis, Embryo, Mammalian blood supply, Embryo, Mammalian metabolism, Endothelial Cells, Humans, Models, Biological, Models, Theoretical, Neoplasms blood supply, Time Factors, Neoplasms pathology, Neovascularization, Pathologic
- Abstract
Angiogenesis, the formation of blood vessels from a pre-existing vasculature, is a process whereby capillary sprouts are formed in response to externally supplied chemical stimuli. The sprouts then grow and develop, driven initially by endothelial cell migration, and organise themselves into a branched, connected network. Subsequent cell proliferation near the sprout-tips permits further extension of the capillaries and ultimately completes the process. Angiogenesis occurs during embryogenesis, wound healing, arthritis and during the growth of solid tumours. In this chapter we first of all present a review of a variety of mathematical models which have been used to describe the formation of capillary networks and then focus on a specific recent model which uses novel mathematical modelling techniques to generate both 2 and 3 dimensional vascular structures. The modelling focusses on key events of angiogenesis such as the migratory response of endothelial cells to exogenous cytokines (tumour angiogenic factors, TAF) secreted by a solid tumour; endothelial cell proliferation; endothelial cell interactions with extracellular matrix macromolecules such as fibronectin; matrix degradation; capillary sprout branching and anastomosis. Numerical simulations of the model, using parameter values based on experimental data, are presented and the theoretical structures generated by the model are compared with the morphology of actual capillary networks observed in in vivo experiments. A final section discusses the use of the mathematical model as a possible angiogenesis assay and implications for chemotherapy regimes.
- Published
- 2004
- Full Text
- View/download PDF
167. Knockout of ERK5 causes multiple defects in placental and embryonic development.
- Author
-
Yan L, Carr J, Ashby PR, Murry-Tait V, Thompson C, and Arthur JS
- Subjects
- Abdomen abnormalities, Abdomen embryology, Animals, Crosses, Genetic, Embryo, Mammalian blood supply, Embryo, Mammalian enzymology, Female, Gene Expression Regulation, Developmental physiology, Gene Expression Regulation, Enzymologic physiology, Genes, Lethal physiology, Head abnormalities, Head embryology, Lower Extremity embryology, Lower Extremity Deformities, Congenital genetics, Male, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Inbred Strains, Mice, Knockout, Mitogen-Activated Protein Kinase 7, Mitogen-Activated Protein Kinase Kinases biosynthesis, Mitogen-Activated Protein Kinases biosynthesis, Mitogen-Activated Protein Kinases genetics, Neovascularization, Physiologic physiology, Placenta embryology, Placenta enzymology, Embryo, Mammalian abnormalities, Mitogen-Activated Protein Kinases deficiency, Mitogen-Activated Protein Kinases physiology, Placenta abnormalities
- Abstract
Background: ERK5 is a member of the mitogen activated protein kinase family activated by certain mitogenic or stressful stimuli in cells, but whose physiological role is largely unclear., Results: To help determine the function of ERK5 we have used gene targeting to inactivate this gene in mice. Here we report that ERK5 knockout mice die at approximately E10.5. In situ hybridisation for ERK5, and its upstream activator MKK5, showed strong expression in the head and trunk of the embryo at this stage of development. Between E9.5 and E10.5, multiple developmental problems are seen in the ERK5-/- embryos, including an increase in apoptosis in the cephalic mesenchyme tissue, abnormalities in the hind gut, as well as problems in vascular remodelling, cardiac development and placental defects., Conclusion: Erk5 is essential for early embryonic development, and is required for normal development of the vascular system and cell survival.
- Published
- 2003
- Full Text
- View/download PDF
168. The antimalaria agent artemisinin exerts antiangiogenic effects in mouse embryonic stem cell-derived embryoid bodies.
- Author
-
Wartenberg M, Wolf S, Budde P, Grünheck F, Acker H, Hescheler J, Wartenberg G, and Sauer H
- Subjects
- Animals, Cell Line, Dose-Response Relationship, Drug, Down-Regulation, Doxorubicin metabolism, Embryo, Mammalian blood supply, Embryo, Mammalian metabolism, Free Radical Scavengers pharmacology, Hypoxia-Inducible Factor 1, alpha Subunit, Laminin metabolism, Matrix Metalloproteinases metabolism, Mice, Reactive Oxygen Species metabolism, Stem Cells metabolism, Transcription Factors metabolism, Vascular Endothelial Growth Factor A metabolism, Anti-Infective Agents pharmacology, Antimalarials pharmacology, Artemisinins pharmacology, Embryo, Mammalian drug effects, Neovascularization, Pathologic, Sesquiterpenes pharmacology, Stem Cells drug effects
- Abstract
Artemisinin is widely used as an agent to treat malaria; the possible antiangiogenic effects of this compound are unknown. In the present study, the antiangiogenic effects of artemisinin were investigated in mouse embryonic stem cell-derived embryoid bodies, which are a model system for early postimplantation embryos and which efficiently differentiate capillaries. Artemisinin dose dependently inhibited angiogenesis in embryoid bodies and raised the level of intracellular reactive oxygen species. Furthermore impaired organization of the extracellular matrix component laminin and altered expression patterns of matrix metalloproteinases 1, 2, and 9 were observed during the time course of embryoid body differentiation. Consequently accelerated penetration kinetics of the fluorescent anthracycline doxorubicin occurred within the tissue, indicating increased tissue permeability. Artemisinin down-regulated hypoxia-inducible factor-1alpha and vascular endothelial growth factor (VEGF) expression, which control endothelial cell growth. The antiangiogenic effects and the inhibition of hypoxia-inducible factor-1alpha and VEGF were reversed upon cotreatment with the free radical scavengers mannitol and vitamin E, indicating that artemisinin may act via reactive oxygen species generation. Furthermore, capillary formation was restored upon coadministration of exogenous VEGF. The data of the present study suggest that the antiangiogenic activity of artemisinin and the increase in tissue permeability for cytostatics may be exploited for anticancer treatment.
- Published
- 2003
- Full Text
- View/download PDF
169. Identification of novel roles of the cytochrome p450 system in early embryogenesis: effects on vasculogenesis and retinoic Acid homeostasis.
- Author
-
Otto DM, Henderson CJ, Carrie D, Davey M, Gundersen TE, Blomhoff R, Adams RH, Tickle C, and Wolf CR
- Subjects
- Animals, Blood Vessels abnormalities, Embryo, Mammalian blood supply, Fetal Death genetics, Fibroblast Growth Factor 8, Fibroblast Growth Factors genetics, Gene Expression Regulation, Developmental, Homeostasis genetics, Mice, Mice, Mutant Strains, Microsomes physiology, NADPH-Ferrihemoprotein Reductase metabolism, Phenotype, Receptors, Retinoic Acid genetics, Tretinoin pharmacology, Vitamin A metabolism, Blood Vessels embryology, Cytochrome P-450 Enzyme System physiology, Embryo, Mammalian enzymology, NADPH-Ferrihemoprotein Reductase genetics, Tretinoin metabolism
- Abstract
The cytochrome P450-dependent monooxygenase system catalyzes the metabolism of xenobiotics and endogenous compounds, including hormones and retinoic acid. In order to establish the role of these enzymes in embryogenesis, we have inactivated the system through the deletion of the gene for the electron donor to all microsomal P450 proteins, cytochrome P450 reductase (Cpr). Mouse embryos homozygous for this deletion died in early to middle gestation (approximately 9.5 days postcoitum [dpc]) and exhibited a number of novel phenotypes, including the severe inhibition of vasculogenesis and hematopoiesis. In addition, defects in the brain, limbs, and cell types where CPR was shown to be expressed were observed. Some of the observed abnormalities have been associated with perturbations in retinoic acid homeostasis in later embryogenesis. Consistent with this possibility, embryos at 9.5 dpc had significantly elevated levels of retinoic acid and reduced levels of retinol. Further, some of the observed phenotypes could be either reversed or exacerbated by decreasing or increasing maternal retinoic acid exposure, respectively. Detailed analysis demonstrated a close relationship between the observed phenotype and the expression of genes controlling vasculogenesis. These data demonstrate that the cytochrome P450 system plays a key role in early embryonic development; this process appears to be, at least in part, controlled by regional concentrations of retinoic acid and has profound effects on blood vessel formation.
- Published
- 2003
- Full Text
- View/download PDF
170. Complications of monochorionic twins.
- Author
-
Feldstein VA and Filly RA
- Subjects
- Diseases in Twins embryology, Embryo, Mammalian blood supply, Female, Humans, Placenta blood supply, Placental Circulation, Placentation physiology, Pregnancy, Pregnancy, Multiple physiology, Syndrome, Diseases in Twins diagnosis, Twins, Monozygotic, Ultrasonography, Prenatal
- Abstract
Sonography has made a dramatic impact on the obstetric management of complicated twin pregnancies. This is based in part on the ability to use prenatal US to diagnose syndromes and complications of MC twinning. All twin pregnancies are at high risk for perinatal morbidity and mortality compared with singleton gestations, but when one of the described complications is recognized, the difficulties in management are compounded dramatically. Despite the relative rarity of some of the entities described, it is vitally important to be familiar with these problems and their sonographic evaluation and diagnosis.
- Published
- 2003
- Full Text
- View/download PDF
171. E-Tmod capping of actin filaments at the slow-growing end is required to establish mouse embryonic circulation.
- Author
-
Chu X, Chen J, Reedy MC, Vera C, Sung KL, and Sung LA
- Subjects
- Animals, Biomarkers, Embryo, Mammalian blood supply, Embryo, Mammalian physiology, Erythrocytes physiology, Gene Expression Regulation, Developmental, Heart embryology, Heart physiology, Hematopoiesis genetics, Lac Operon, Mice, Mice, Knockout, Microscopy, Electron, Myocardial Contraction genetics, Myocytes, Cardiac ultrastructure, Phenotype, Rotation, Stress, Mechanical, Tropomodulin, Yolk Sac blood supply, Yolk Sac physiology, Actin Cytoskeleton metabolism, Carrier Proteins genetics, Carrier Proteins metabolism, Erythroid Precursor Cells metabolism, Microfilament Proteins, Neovascularization, Physiologic physiology
- Abstract
Tropomodulins are a family of proteins that cap the slow-growing end of actin filaments. Erythrocyte tropomodulin (E-Tmod) stabilizes short actin protofilaments in erythrocytes and caps longer sarcomeric actin filaments in striated muscles. We report the knockin of the beta-galactosidase gene (LacZ) under the control of the endogenous E-Tmod promoter and the knockout of E-Tmod in mouse embryonic stem cells. E-Tmod(-/-) embryos die around embryonic day 10 and exhibit a noncontractile heart tube with disorganized myofibrils and underdevelopment of the right ventricle, accumulation of mechanically weakened primitive erythroid cells in the yolk sac, and failure of primary capillary plexuses to remodel into vitelline vessels, all required to establish blood circulation between the yolk sac and the embryo proper. We propose a hemodynamic "plexus channel selection" mechanism as the basis for vitelline vascular remodeling. The defects in cardiac contractility, vitelline circulation, and hematopoiesis reflect an essential role for E-Tmod capping of the actin filaments in both assembly of cardiac sarcomeres and of the membrane skeleton in erythroid cells that is not compensated for by other proteins.
- Published
- 2003
- Full Text
- View/download PDF
172. The embryo makes red blood cell progenitors in every tissue simultaneously with blood vessel morphogenesis.
- Author
-
Sequeira Lopez ML, Chernavvsky DR, Nomasa T, Wall L, Yanagisawa M, and Gomez RA
- Subjects
- Animals, Gene Expression Regulation, Developmental, Kidney blood supply, Kidney cytology, Kidney embryology, Mice, Mice, Transgenic, Morphogenesis, Time Factors, Blood Vessels embryology, Embryo, Mammalian blood supply, Embryo, Mammalian embryology, Erythroid Precursor Cells cytology, Erythropoiesis, Hematopoiesis
- Abstract
During embryonic life, hematopoiesis occurs first in the yolk sac, followed by the aorto-gonado-mesonephric region, the fetal liver, and the bone marrow. The possibility of hematopoiesis in other embryonic sites has been suspected for a long time. With the use of different methodologies (transgenic mice, electron microscopy, laser capture microdissection, organ culture, and cross-transplant experiments), we show that multiple regions within the embryo are capable of forming blood before and during organogenesis. This widespread phenomenon occurs by hemo-vasculogenesis, the formation of blood vessels accompanied by the simultaneous generation of red blood cells. Erythroblasts develop within aggregates of endothelial cell precursors. When the lumen forms, the erythroblasts "bud" from endothelial cells into the forming vessel. The extensive hematopoietic capacity found in the embryo helps explain why, under pathological circumstances such as severe anemia, extramedullary hematopoiesis can occur in any adult tissue. Understanding the intrinsic ability of tissues to manufacture their own blood cells and vessels has the potential to advance the fields of organogenesis, regeneration, and tissue engineering.
- Published
- 2003
- Full Text
- View/download PDF
173. Circulation is established in a stepwise pattern in the mammalian embryo.
- Author
-
McGrath KE, Koniski AD, Malik J, and Palis J
- Subjects
- Animals, Blood Cell Count, Blood Vessels embryology, Embryonic and Fetal Development, Erythroblasts cytology, Erythrocyte Count, Gestational Age, Hematopoietic Stem Cells cytology, Liver embryology, Mice, Mice, Inbred ICR, Neovascularization, Physiologic, Yolk Sac cytology, Blood Circulation, Embryo, Mammalian blood supply
- Abstract
To better understand the relationship between the embryonic hematopoietic and vascular systems, we investigated the establishment of circulation in mouse embryos by examining the redistribution of yolk sac-derived primitive erythroblasts and definitive hematopoietic progenitors. Our studies revealed that small numbers of erythroblasts first enter the embryo proper at 4 to 8 somite pairs (sp) (embryonic day 8.25 [E8.25]), concomitant with the proposed onset of cardiac function. Hours later (E8.5), most red cells remained in the yolk sac. Although the number of red cells expanded rapidly in the embryo proper, a steady state of approximately 40% red cells was not reached until 26 to 30 sp (E10). Additionally, erythroblasts were unevenly distributed within the embryo's vasculature before 35 sp. These data suggest that fully functional circulation is established after E10. This timing correlated with vascular remodeling, suggesting that vessel arborization, smooth muscle recruitment, or both are required. We also examined the distribution of committed hematopoietic progenitors during early embryogenesis. Before E8.0, all progenitors were found in the yolk sac. When normalized to circulating erythroblasts, there was a significant enrichment (20- to 5-fold) of progenitors in the yolk sac compared with the embryo proper from E9.5 to E10.5. These results indicated that the yolk sac vascular network remains a site of progenitor production and preferential adhesion even as the fetal liver becomes a hematopoietic organ. We conclude that a functional vascular system develops gradually and that specialized vascular-hematopoietic environments exist after circulation becomes fully established.
- Published
- 2003
- Full Text
- View/download PDF
174. Developmental biology: A twist in a mouse tale.
- Author
-
Dyson N
- Subjects
- Animals, Cell Division, Cell Lineage, Embryo Loss genetics, Embryo Loss metabolism, Embryo Loss pathology, Embryo, Mammalian blood supply, Embryo, Mammalian cytology, Mice, Mutation, Phenotype, Placenta blood supply, Placenta cytology, Retinoblastoma Protein genetics, Embryo, Mammalian embryology, Embryo, Mammalian metabolism, Placenta abnormalities, Placenta metabolism, Retinoblastoma Protein metabolism
- Published
- 2003
- Full Text
- View/download PDF
175. Extra-embryonic function of Rb is essential for embryonic development and viability.
- Author
-
Wu L, de Bruin A, Saavedra HI, Starovic M, Trimboli A, Yang Y, Opavska J, Wilson P, Thompson JC, Ostrowski MC, Rosol TJ, Woollett LA, Weinstein M, Cross JC, Robinson ML, and Leone G
- Subjects
- Animals, Cell Division, Embryo Loss metabolism, Embryo Loss pathology, Embryo, Mammalian blood supply, Embryo, Mammalian cytology, Fatty Acids metabolism, Female, Fetus blood supply, Fetus cytology, Fetus embryology, Fetus metabolism, Gene Deletion, Immunohistochemistry, Male, Mice, Mice, Knockout, Placenta abnormalities, Placenta blood supply, Placenta cytology, RNA, Messenger genetics, RNA, Messenger metabolism, Retinoblastoma Protein genetics, Trophoblasts cytology, Trophoblasts metabolism, Cell Lineage, Embryo, Mammalian embryology, Embryo, Mammalian metabolism, Placenta metabolism, Retinoblastoma Protein metabolism
- Abstract
The retinoblastoma (Rb) gene was the first tumour suppressor identified. Inactivation of Rb in mice results in unscheduled cell proliferation, apoptosis and widespread developmental defects, leading to embryonic death by day 14.5 (refs 2-4). However, the actual cause of the embryonic lethality has not been fully investigated. Here we show that loss of Rb leads to excessive proliferation of trophoblast cells and a severe disruption of the normal labyrinth architecture in the placenta. This is accompanied by a decrease in vascularization and a reduction in placental transport function. We used two complementary techniques-tetraploid aggregation and conditional knockout strategies-to demonstrate that Rb-deficient embryos supplied with a wild-type placenta can be carried to term, but die soon after birth. Most of the neurological and erythroid abnormalities thought to be responsible for the embryonic lethality of Rb-null animals were virtually absent in rescued Rb-null pups. These findings identify and define a key function of Rb in extra-embryonic cell lineages that is required for embryonic development and viability, and provide a mechanism for the cell autonomous versus non-cell autonomous roles of Rb in development.
- Published
- 2003
- Full Text
- View/download PDF
176. Focal adhesion kinase is required for blood vessel morphogenesis.
- Author
-
Ilic D, Kovacic B, McDonagh S, Jin F, Baumbusch C, Gardner DG, and Damsky CH
- Subjects
- Animals, Blood Vessels pathology, Cell Differentiation genetics, Cell Differentiation physiology, Cells, Cultured, Embryo, Mammalian abnormalities, Embryo, Mammalian blood supply, Embryo, Mammalian enzymology, Endothelium, Vascular cytology, Endothelium, Vascular enzymology, Focal Adhesion Kinase 1, Focal Adhesion Protein-Tyrosine Kinases, Humans, In Vitro Techniques, Mice, Mice, Inbred CBA, Mice, Knockout, Morphogenesis genetics, Platelet Endothelial Cell Adhesion Molecule-1 genetics, Platelet Endothelial Cell Adhesion Molecule-1 metabolism, Protein-Tyrosine Kinases genetics, RNA, Messenger metabolism, Vascular Endothelial Growth Factor Receptor-2 genetics, Vascular Endothelial Growth Factor Receptor-2 metabolism, Blood Vessels abnormalities, Blood Vessels enzymology, Protein-Tyrosine Kinases deficiency, Protein-Tyrosine Kinases metabolism
- Abstract
The nonreceptor tyrosine kinase focal adhesion kinase (FAK) is a point of convergence for signals from extracellular matrix, soluble factors, and mechanical stimuli. Targeted disruption of the fak gene in mice leads to death at embryonic day 8.5 (E8.5). FAK-/- embryos have severely impaired blood vessel development. Gene expression and in vitro differentiation studies revealed that endothelial cell differentiation was comparable in FAK-/- and wild-type E8.5 embryos. We examined the role of FAK in blood vessel morphogenesis using an in vitro tubulogenesis assay and three different culture systems: FAK+/+ and FAK-/- embryoid bodies, FAK+/+ and FAK-/- endothelial cells, and human umbilical vein endothelial cells expressing antisense FAK, a dominant-negative fragment of FAK, or wild-type FAK. In all of these systems, endothelial cells deficient in FAK expression or function displayed a severely reduced ability to form tubules in Matrigel. These studies demonstrate clearly that the vascular defects in FAK-/- mice result from the inability of FAK-deficient endothelial cells to organize themselves into vascular networks, rather than from defects in tissue-specific differentiation.
- Published
- 2003
- Full Text
- View/download PDF
177. Onset of cardiac function during early mouse embryogenesis coincides with entry of primitive erythroblasts into the embryo proper.
- Author
-
Ji RP, Phoon CK, Aristizábal O, McGrath KE, Palis J, and Turnbull DH
- Subjects
- Animals, Aorta embryology, Aorta physiology, Blood Circulation physiology, Blood Flow Velocity physiology, Cell Movement physiology, Embryo, Mammalian blood supply, Heart Rate, Fetal, In Situ Hybridization, Mice, Microscopy, Acoustic methods, Time Factors, Ultrasonography, Doppler, Ultrasonography, Prenatal, Erythroblasts cytology, Heart embryology, Heart physiology, Myocardium cytology, Organogenesis physiology
- Abstract
When cardiac function and blood flow are first established are fundamental questions in mammalian embryogenesis. The earliest erythroblasts arise in yolk sac blood islands and subsequently enter the embryo proper to initiate circulation. Embryos staged 0 to 30 somites (S) were examined in utero with 40- to 50-MHz ultrasound biomicroscopy (UBM)-Doppler, to determine onset of embryonic heartbeat and blood flow and to characterize basic physiology of the very early mouse embryonic circulation. A heartbeat was first detected at 5 S, and blood vascular flow at 7 S. Heart rate, peak arterial velocity, and velocity-time integral showed progressive increases that indicated a dramatically increasing cardiac output from even the earliest stages. In situ hybridization revealed an onset of the heartbeat coincident with the appearance of yolk sac-derived erythroblasts in the embryo proper at 5 S. Early maturation of the circulation follows a tightly coordinated program.
- Published
- 2003
- Full Text
- View/download PDF
178. A fluorescent Tie1 reporter allows monitoring of vascular development and endothelial cell isolation from transgenic mouse embryos.
- Author
-
Iljin K, Petrova TV, Veikkola T, Kumar V, Poutanen M, and Alitalo K
- Subjects
- Animals, Blood Vessels embryology, Cell Line, Embryo, Mammalian blood supply, Embryo, Mammalian cytology, Endothelium, Vascular cytology, Female, Green Fluorescent Proteins, Humans, Luminescent Proteins genetics, Luminescent Proteins metabolism, Male, Mice, Mice, Inbred Strains, Mice, Transgenic, Neoplasms, Experimental blood supply, Neoplasms, Experimental genetics, Neoplasms, Experimental metabolism, Neovascularization, Pathologic genetics, Neovascularization, Pathologic metabolism, Promoter Regions, Genetic genetics, Receptor Protein-Tyrosine Kinases genetics, Receptor, TIE-1, Receptors, Cell Surface genetics, Receptors, TIE, Skin metabolism, Time Factors, Transfection, Tumor Cells, Cultured, Blood Vessels metabolism, Embryo, Mammalian metabolism, Endothelium, Vascular metabolism, Receptor Protein-Tyrosine Kinases metabolism, Receptors, Cell Surface metabolism
- Abstract
Tie1 is an endothelial receptor tyrosine kinase essential for development and maintenance of the vascular system. Here we report generation of transgenic mice expressing enhanced green fluorescent protein (EGFP) or a chimeric protein consisting of a Zeosin resistance marker and EGFP under the control of mouse Tie1 promoter. Intravital monitoring of fluorescence showed that the EGFP reporter recapitulates the Tie1 expression pattern in the developing vasculature, and flow cytometry using EGFP allowed the isolation of essentially pure Tie1-expressing endothelial cells from transgenic mouse embryos. However, EGFP and LacZ transgenic markers were strongly down-regulated in the adult vasculature; unlike the Tie1-LacZ knock-in locus, the promoter was not reactivated during tumor neovascularization, indicating the presence of additional regulatory elements in the Tie1 locus. Starting at midgestation, Tie1 promoter activity became stronger in the arterial than in the venous endothelium; in adult mice, promoter activity was observed in arterioles, capillaries, and lymphatic vessels, indicating a significant degree of specificity in different types of endothelial cells. Our results establish Tie1-Z/EGFP transgenic mice as a useful model to study embryonic vascular development and a convenient source for the isolation of primary endothelial cells.
- Published
- 2002
- Full Text
- View/download PDF
179. Plastic casting of embryonic, placental, and tumor vasculature in the mouse.
- Author
-
Ozerdem U, Charbono WL, and Stallcup WB
- Subjects
- Animals, Melanoma, Experimental, Mice, Mice, Inbred C57BL, Neoplasm Transplantation, Plastics, Polymers, Time Factors, Umbilical Cord physiology, Blood Vessels ultrastructure, Corrosion Casting methods, Embryo, Mammalian blood supply, Microcirculation, Neoplasms blood supply, Placenta blood supply
- Published
- 2002
- Full Text
- View/download PDF
180. Spatial velocity profile in mouse embryonic aorta and Doppler-derived volumetric flow: a preliminary model.
- Author
-
Phoon CK, Aristizábal O, and Turnbull DH
- Subjects
- Animals, Embryo, Mammalian blood supply, Female, Male, Mice, Models, Cardiovascular, Pregnancy, Reproducibility of Results, Rheology instrumentation, Rheology standards, Ultrasonography instrumentation, Ultrasonography standards, Aorta embryology, Aorta physiology, Blood Flow Velocity physiology, Rheology methods, Ultrasonography methods
- Abstract
Characterizing embryonic circulatory physiology requires accurate cardiac output and flow data. Despite recent applications of high-frequency ultrasound Doppler to the study of embryonic circulation, current Doppler analysis of volumetric flow is relatively crude. To improve Doppler derivation of volumetric flow, we sought a preliminary model of the spatial velocity profile in the mouse embryonic dorsal aorta using ultrasound biomicroscopy (UBM)-Doppler data. Embryonic hematocrit is 0.05-0.10 so rheologic properties must be insignificant. Low Reynolds numbers (<500) and Womersley parameters (<0.76) suggest laminar flow. UBM demonstrated a circular dorsal aortic cross section with no significant tapering. Low Dean numbers (<100) suggest the presence of minimal skewing of the spatial velocity profile. The inlet length allows for fully developed flow. There is no apparent aortic wall pulsatility. Extrapolation of prior studies to these vessel diameters (300-350 microm) and flow velocities (~50-200 mm/s) suggests parabolic spatial velocity profiles. Therefore, mouse embryonic dorsal aortic blood flow may correspond to Poiseuille flow in a straight rigid tube with parabolic spatial velocity profiles. As a first approximation, these results are an important step toward precise in utero ultrasound characterization of blood flow within the developing mammalian circulation.
- Published
- 2002
- Full Text
- View/download PDF
181. Blood-forming potential of vascular endothelium in the human embryo.
- Author
-
Oberlin E, Tavian M, Blazsek I, and Péault B
- Subjects
- Antigens, CD34 metabolism, Bone Marrow physiology, Cells, Cultured, Coculture Techniques, Embryo, Mammalian physiology, Endothelium, Vascular embryology, Flow Cytometry, Humans, Immunohistochemistry, Leukocyte Common Antigens metabolism, Liver physiology, Platelet Endothelial Cell Adhesion Molecule-1 metabolism, Stromal Cells, Yolk Sac physiology, Embryo, Mammalian blood supply, Endothelium, Vascular physiology, Hematopoiesis physiology
- Abstract
Hematopoietic cells arise first in the third week of human ontogeny inside yolk sac developing blood vessels, then, one week later and independently, from the wall of the embryonic aorta and vitelline artery. To address the suggested derivation of emerging hematopoietic stem cells from the vessel endothelium, endothelial cells have been sorted by flow cytometry from the yolk sac and aorta and cultured in the presence of stromal cells that support human multilineage hematopoiesis. Embryonic endothelial cells were most accurately selected on CD34 or CD31 surface expression and absence of CD45, which guaranteed the absence of contaminating hematopoietic cells. Yet, rigorously selected endothelial cells yielded a progeny of myelo-lymphoid cells in culture. The frequency of hemogenic endothelial cells in the yolk sac and aorta reflected the actual blood-forming activity of these tissues, as a function of developmental age. Even less expected, a subset of endothelial cells sorted similarly from the embryonic liver and fetal bone marrow also exhibited blood-forming potential. These results suggest that a part at least of emerging hematopoietic cells in the human embryo and fetus originate in vascular walls.
- Published
- 2002
- Full Text
- View/download PDF
182. Distribution of endothelial cell protein C/activated protein C receptor (EPCR) during mouse embryo development.
- Author
-
Crawley JT, Gu JM, Ferrell G, and Esmon CT
- Subjects
- Animals, Aorta cytology, Aorta embryology, Blood Vessels cytology, Blood Vessels embryology, Blood Vessels growth & development, Embryo, Mammalian blood supply, Endothelium, Vascular cytology, Immunohistochemistry, Mice, Placenta cytology, Tissue Distribution, Trophoblasts chemistry, Endothelium, Vascular chemistry, Endothelium, Vascular embryology
- Abstract
The endothelial cell protein C receptor (EPCR) augments protein C activation by the thrombomodulin.thrombin complex. Deletion of the EPCR gene in mice has been reported to lead to embryonic lethality before embryonic day 10 (E10.0). To identify potential mechanisms responsible for this lethality, we performed an immunohistological analysis of EPCR distribution during mouse embryogenesis. EPCR was detected in the trophoblast giant cells at the feto-maternal boundary from E7.5 and at later time points in the trophoblasts of the placenta, suggesting a role in the haemostatic regulation of the maternal blood that irrigates these surfaces. In the embryo, EPCR was weakly detected in aortic endothelial cells from E13.5. Thereafter, EPCR levels increased in certain large blood vessels endothelial cells suggesting that the specificity of EPCR to large vessels is conferred in utero. However, not until postnatal day 7 did the intensity and distribution of EPCR staining mimic that observed in adult mice.
- Published
- 2002
183. Central roles of alpha5beta1 integrin and fibronectin in vascular development in mouse embryos and embryoid bodies.
- Author
-
Francis SE, Goh KL, Hodivala-Dilke K, Bader BL, Stark M, Davidson D, and Hynes RO
- Subjects
- Animals, Blood Vessels embryology, Blood Vessels pathology, Blood Vessels physiology, Cell Differentiation genetics, Cells, Cultured, Embryo, Mammalian pathology, Embryo, Mammalian physiology, Embryonic Structures pathology, Embryonic Structures physiology, Endothelium, Vascular pathology, Endothelium, Vascular physiology, Mice, Phenotype, Platelet Endothelial Cell Adhesion Molecule-1 metabolism, Receptors, Fibronectin biosynthesis, Receptors, Fibronectin deficiency, Receptors, Fibronectin genetics, Stem Cells chemistry, Stem Cells pathology, Stem Cells physiology, Embryo, Mammalian blood supply, Embryonic Structures blood supply, Endothelium, Vascular embryology, Receptors, Fibronectin physiology
- Abstract
Vascular development and maturation are dependent on the interactions of endothelial cell integrins with surrounding extracellular matrix. Previous investigations of the primacy of certain integrins in vascular development have not addressed whether this could also be a secondary effect due to poor embryonic nutrition. Here, we show that the alpha5 integrin subunit and fibronectin have critical roles in blood vessel development in mouse embryos and in embryoid bodies (EBs) differentiated from embryonic stem cells (a situation in which there is no nutritional deficit caused by the mutations). In contrast, vascular development in vivo and in vitro is not strongly dependent on alpha(v) or beta3 integrin subunits. In mouse embryos lacking alpha5 integrin, greatly distended blood vessels are seen in the vitelline yolk sac and in the embryo itself. Additionally, overall blood vessel pattern complexity is reduced in alpha5-null tissues. This defective vascular phenotype is correlated with a decrease in the ligand for alpha5 integrin, fibronectin (FN), in the endothelial basement membranes. A striking and significant reduction in early capillary plexus formation and maturation was apparent in EBs formed from embryonic stem cells lacking alpha5 integrin or FN compared with wild-type EBs or EBs lacking alpha(v) or beta3 integrin subunits. Vessel phenotype could be partially restored to FN-null EBs by the addition of whole FN to the culture system. These findings confirm a clear role for alpha5 and FN in early blood vessel development not dependent on embryo nutrition or alpha(v) or beta3 integrin subunits. Thus, successful early vasculogenesis and angiogenesis require alpha5-FN interactions.
- Published
- 2002
- Full Text
- View/download PDF
184. Targeting of both mouse neuropilin-1 and neuropilin-2 genes severely impairs developmental yolk sac and embryonic angiogenesis.
- Author
-
Takashima S, Kitakaze M, Asakura M, Asanuma H, Sanada S, Tashiro F, Niwa H, Miyazaki Ji J, Hirota S, Kitamura Y, Kitsukawa T, Fujisawa H, Klagsbrun M, and Hori M
- Subjects
- Animals, Embryo, Mammalian embryology, Genotype, Hemorrhage, Mice, Mice, Knockout, Nerve Tissue Proteins genetics, Neuropilin-1, Phenotype, Platelet Endothelial Cell Adhesion Molecule-1 analysis, RNA, Messenger genetics, RNA, Messenger metabolism, Staining and Labeling, Yolk Sac embryology, Embryo, Mammalian blood supply, Gene Deletion, Neovascularization, Physiologic, Nerve Tissue Proteins deficiency, Nerve Tissue Proteins metabolism, Yolk Sac blood supply
- Abstract
Neuropilins (NP1 and NP2) are vascular endothelial growth factor (VEGF) receptors that mediate developmental and tumor angiogenesis. Transgenic mice, in which both NP1 and NP2 were targeted (NP1(-/-)NP2(-/-)) died in utero at E8.5. Their yolk sacs were totally avascular. Mice deficient for NP2 but heterozygous for NP1 (NP1(+/-)NP2(-/-)) or deficient for NP1 but heterozygous for NP2 (NP1(-/-)NP2(+/-)) were also embryonic lethal and survived to E10-E10.5. The E10 yolk sacs and embryos were easier to analyze for vascular phenotype than the fragile poorly formed 8.5 embryos. The vascular phenotypes of these E10 mice were very abnormal. The yolk sacs, although of normal size, lacked the larger collecting vessels and had less dense capillary networks. PECAM staining of yolk sac endothelial cells showed the absence of branching arteries and veins, the absence of a capillary bed, and the presence of large avascular spaces between the blood vessels. The embryos displayed blood vessels heterogeneous in size, large avascular regions in the head and trunk, and blood vessel sprouts that were unconnected. The embryos were about 50% the length of wild-type mice and had multiple hemorrhages. These double NP1/NP2 knockout mice had a more severe abnormal vascular phenotype than either NP1 or NP2 single knockouts. Their abnormal vascular phenotype resembled those of VEGF and VEGFR-2 knockouts. These results suggest that NRPs are early genes in embryonic vessel development and that both NP1 and NP2 are required.
- Published
- 2002
- Full Text
- View/download PDF
185. Histological and histochemical analysis of embryoid bodies.
- Author
-
Karbanová J and Mokrý J
- Subjects
- Actins analysis, Animals, Biomarkers analysis, Cell Differentiation, Cells, Cultured, Coculture Techniques, Desmin analysis, Embryo, Mammalian blood supply, Embryo, Mammalian chemistry, Mice, Myogenin analysis, Neovascularization, Physiologic, Totipotent Stem Cells chemistry, alpha-Fetoproteins analysis, Embryo, Mammalian cytology, Totipotent Stem Cells cytology
- Abstract
We examined the histological structure of embryoid bodies arising from aggregation of mouse embryonic stem (ES D3) cells after 7, 12, 18 and 26 days of in vitro culture. Morphology of originally solid embryoid bodies was affected by the process of cavitation that resulted in formation of cystic embryoid bodies and by spontaneous differentiation of the ES D3 cells. We applied in situ immunophenotyping to characterise cell populations that spontaneously differentiated inside the embryoid bodies in the various stages. The most distinct cell populations that were found inside embryoid bodies were alpha-fetoprotein-positive endodermal cells and myogenic cells that expressed desmin, myogenin or smooth muscle actin. ES D3-derived endothelial cells generated during vasculogenesis inside the embryoid bodies differed from mature endothelial cells because they did not stain for von Willebrand factor. These cells also differed from endothelial cell that were generated during angiogenesis since they did not stain for the intermediate filament nestin. Our results demonstrate the usefulness of this in vitro model for studying early embryogenesis.
- Published
- 2002
- Full Text
- View/download PDF
186. Three-dimensional power Doppler in study of embryonic vasculogenesis.
- Author
-
Kurjak A, Hafner T, Kupesić S, and Kostovic L
- Subjects
- Embryonic and Fetal Development, Female, Gestational Age, Humans, Pregnancy, Embryo, Mammalian blood supply, Embryo, Mammalian diagnostic imaging, Ultrasonography, Prenatal methods
- Abstract
Three-dimensional power Doppler sonography plays an important role in obstetrics, predominantly for assessing the relationship between fetal vascular system and surrounding organs. Presenting volume data in a standard anatomic orientation is valuable for assisting both ultrasonographers and pregnant patients to recognize anatomy more readily. This investigation was designed as an observational cross-sectional study. A group of 25 patients in gestational age of five to eleven weeks were recruited for the study. Architecture of the embryonic vessels was depicted by 3D power flow mapping and analyzed visually. Vascular 3D measurements were done through 3D color/power histogram and expressed by Vascularization Index (VI) and Vascularization Flow Index (VFI). Volume of the embryo increased exponentially throughout the observation period. The VI and VFI showed no change despite an exponential growth of embryonic volume. The findings of our study suggest that in cases of physiologic embryonic vasculogenesis there is a homeostatic steady state between tissue demands and blood supply given through the vascular network.
- Published
- 2002
- Full Text
- View/download PDF
187. Syk expression in endothelial cells and their morphologic defects in embryonic Syk-deficient mice.
- Author
-
Yanagi S, Inatome R, Ding J, Kitaguchi H, Tybulewicz VL, and Yamamura H
- Subjects
- Animals, Aorta cytology, Aorta enzymology, Cattle, Embryo, Mammalian blood supply, Embryo, Mammalian enzymology, Embryo, Mammalian pathology, Endothelium, Vascular embryology, Endothelium, Vascular pathology, Enzyme Precursors deficiency, Humans, Immunohistochemistry, Intracellular Signaling Peptides and Proteins, Mice, Mice, Mutant Strains embryology, Microscopy, Electron, Protein-Tyrosine Kinases deficiency, Purpura embryology, Purpura enzymology, Purpura etiology, Syk Kinase, Umbilical Veins cytology, Umbilical Veins enzymology, Endothelium, Vascular enzymology, Enzyme Precursors metabolism, Protein-Tyrosine Kinases metabolism
- Abstract
Mice deficient in the Syk tyrosine kinase showed severe petechiae in utero and died shortly after birth. The mechanism of this bleeding, however, remains unknown. Here it is shown that this bleeding is caused by morphologic defects of Syk-deficient endothelial cells during embryogenesis. Immunoblot and reverse transcriptase-polymerase chain reaction Northern blot analysis indicated that Syk is expressed in several endothelial cell lines. Immunocytochemical analysis also confirmed that Syk is expressed in the normal embryonic endothelial cells and is absent in Syk-deficient mice. Furthermore, electron microscopic analysis of Syk-deficient mice revealed an abnormal morphogenesis and a decreased number of endothelial cells. The results indicate a critical role for Syk in endothelial cell function and in maintaining vascular integrity in vivo.
- Published
- 2001
- Full Text
- View/download PDF
188. Role of hematopoietic stem cells in angiogenesis.
- Author
-
Suda T and Takakura N
- Subjects
- Angiopoietin-1, Animals, Core Binding Factor Alpha 2 Subunit, DNA-Binding Proteins genetics, DNA-Binding Proteins pharmacology, DNA-Binding Proteins physiology, Embryo, Mammalian blood supply, Embryo, Mammalian cytology, Embryo, Mammalian metabolism, Endothelium, Vascular cytology, Endothelium, Vascular embryology, Humans, Membrane Glycoproteins metabolism, Membrane Glycoproteins pharmacology, Membrane Glycoproteins physiology, Mice, Transcription Factors genetics, Transcription Factors pharmacology, Transcription Factors physiology, Hematopoietic Stem Cells physiology, Neovascularization, Physiologic physiology, Proto-Oncogene Proteins
- Abstract
Angiogenesis is an important event for embryonic organogenesis as well as for tissue repair in the adult. Here we show that hematopoietic stem cells (HSCs) are essential for angiogenesis during embryogenesis. To investigate the role of HSCs in endothelial cell (EC) development, we analyzed AML1-deficient embryos, which lack definitive hematopoiesis. These embryos showed defective angiogenesis in the head, pericardium, and fetal liver. Para-aortic splanchnopleural (P-Sp) explant cultures on stromal cells (P-Sp cultures) did not generate definitive hematopoietic cells and showed defective angiogenesis in the AML1-null embryo. Disrupted angiogenesis in P-Sp cultures from AML1-null embryos was rescued by addition of HSCs. HSCsspecifically produce angiopoietin-1 (Ang1). Thus HSCs,which expressAng1, directly promoted migration of ECs. These findings suggest that HSCs alone prepare the hematopoietic microenvironment.
- Published
- 2001
- Full Text
- View/download PDF
189. Molecular regulation of embryonic hematopoiesis and vascular development: a novel pathway.
- Author
-
Baron MH
- Subjects
- Animals, Embryo, Mammalian blood supply, Embryo, Mammalian metabolism, Gene Expression Regulation, Developmental, Hedgehog Proteins, Hematopoiesis genetics, Humans, Models, Biological, Neovascularization, Physiologic genetics, Signal Transduction, Trans-Activators genetics, Trans-Activators physiology, Hematopoiesis physiology, Hematopoietic System embryology, Neovascularization, Physiologic physiology
- Abstract
In all vertebrate animals, the first blood and vascular endothelial cells are formed during gastrulation, a process in which the mesoderm of the embryo is induced and then patterned by molecules whose identity is still largely unknown. Clusters of developing blood cells surrounded by a layer of endothelial cells comprise the "blood islands" and form in the visceral yolk sac, external to the developing embryo proper. Despite the identification of genes, such as Flk1, SCL/tal-1, Cbfa2/Runx1/AML1, and CD34, that are expressed during the induction of primitive hematopoiesis and vasculogenesis, the early molecular and cellular events involved in these processes are not well understood. Recent work has demonstrated that extracellular signals secreted by a layer of visceral endoderm surrounding the embryo are essential for the initiation of these events. A member of the Hedgehog family of signaling molecules is produced by visceral endoderm and is required for formation of blood and endothelial cells in explant cultures. Hedgehog proteins also stimulate proliferation of definitive hematopoietic stem/progenitor cells. Therefore, these findings may have important medical implications for regulating hematopoiesis and vascular development for therapeutic purposes and for the development of new sources of hematopoietic stem cells for transplantation and as targets for gene therapy.
- Published
- 2001
- Full Text
- View/download PDF
190. Circulatory physiology in the developing embryo.
- Author
-
Phoon CK
- Subjects
- Biomechanical Phenomena, Blood Flow Velocity, Diastole physiology, Heart Rate physiology, Heart Valves physiology, Humans, Systole physiology, Ventricular Function, Coronary Circulation physiology, Embryo, Mammalian blood supply, Embryo, Mammalian physiology, Fetal Heart physiology
- Abstract
Knowledge of early developmental circulatory physiology has lagged behind advances in molecular cardiology. Cardiovascular physiology changes during embryonic development in a highly complex and carefully orchestrated manner, tightly correlated with structural development. Circulatory changes in early development include increasing heart rate, preload, and cardiac output; decreasing peripheral resistance; and increasing ventricular compliance, paralleling the increasing metabolic needs of the growing embryo. Newer techniques and the recent ability to study mammalian models of development have led to further insight into changes in myocardial and peripheral vascular physiology. The next major challenges include understanding the mechanisms regulating cardiovascular hemodynamics, normal physiologic adaptation of the growing embryo, and the physiology of abnormal cardiovascular development.
- Published
- 2001
- Full Text
- View/download PDF
191. In vivo requirements for GATA-1 functional domains during primitive and definitive erythropoiesis.
- Author
-
Shimizu R, Takahashi S, Ohneda K, Engel JD, and Yamamoto M
- Subjects
- Animals, Cell Line, DNA-Binding Proteins genetics, Embryo, Mammalian anatomy & histology, Embryo, Mammalian blood supply, Embryo, Mammalian cytology, Erythrocytes physiology, Erythroid-Specific DNA-Binding Factors, GATA1 Transcription Factor, Heme metabolism, Male, Mice, Mice, Transgenic, Protein Structure, Tertiary, RNA, Messenger biosynthesis, Sequence Deletion, Trans-Activators chemistry, Trans-Activators genetics, Trans-Activators physiology, Transcription Factors genetics, Zinc Fingers, DNA-Binding Proteins chemistry, DNA-Binding Proteins physiology, Erythropoiesis genetics, Gene Expression Regulation, Developmental, Hematopoietic System embryology, Transcription Factors chemistry, Transcription Factors physiology
- Abstract
GATA-1 is a transcription factor essential for erythroid/megakaryocytic cell differentiation. To investigate the contribution of individual domains of GATA-1 to its activity, transgenic mice expressing either an N-terminus, or an N- or C-terminal zinc finger deletion of GATA-1 (Delta NT, Delta NF or Delta CF, respectively) were generated and crossed to GATA-1 germline mutant (GATA-1.05) mice. Since the GATA-1 gene is located on the X-chromosome, male GATA-1 mutants die by embryonic day 12.5. Both Delta NF and Delta CF transgenes failed to rescue the GATA-1.05/Y pups. However, transgenic mice expressing Delta NT, but not the Delta NF protein, were able to rescue definitive hematopoiesis. In embryos, while neither the Delta CF protein nor a mutant missing both N-terminal domains (Delta NTNF) was able to support primitive erythropoiesis, the two independent Delta NT and Delta NF mutants could support primitive erythropoiesis. Thus, lineage-specific transgenic rescue of the GATA-1 mutant mouse revealed novel properties that are conferred by specific domains of GATA-1 during primitive and definitive erythropoiesis, and demonstrate that the NT and NF moieties lend complementary, but distinguishable properties to the function of GATA-1.
- Published
- 2001
- Full Text
- View/download PDF
192. Targeted deletion of the cytosolic domain of tissue factor in mice does not affect development.
- Author
-
Melis E, Moons L, De Mol M, Herbert JM, Mackman N, Collen D, Carmeliet P, and Dewerchin M
- Subjects
- Animals, Cells, Cultured, Embryo, Mammalian blood supply, Factor VIIa pharmacology, Factor Xa pharmacology, Fibroblasts drug effects, Fibroblasts metabolism, Gene Deletion, Gene Targeting, Hemostasis, Mice, Mitogen-Activated Protein Kinase 1 metabolism, Mitogen-Activated Protein Kinase 3, Mitogen-Activated Protein Kinases metabolism, Neovascularization, Physiologic, Protein Structure, Tertiary, Thromboplastin chemistry, Thrombosis etiology, Embryonic and Fetal Development, Thromboplastin genetics, Thromboplastin physiology
- Abstract
The role of the cytosolic domain of tissue factor (TF) in signal transduction and gene regulation was studied in mice with a targeted deletion of the 18 carboxy-terminal intracellular amino acids. This deletion was introduced in exon 6 along with a floxed neo(R) selection cassette in intron 5 using homologous recombination in embryonic stem cells. Removal of the floxed neo(R) cassette by in vivo Cre-mediated loxP recombination yielded TF(+/deltaCT) and TF(deltaCT/deltaCT) mice. In contrast to TF(-/-) mice, TF(+/deltaCT) and TF(deltaCT/deltaCT) mice displayed normal embryonic development, survival, fertility, and blood coagulation. Factor VIIa or factor Xa stimulation produced similar p44/42 MAPK activation in TF(+/+) and TF(deltaCT/deltaCT) fibroblasts. These data, based on expression of a TF(deltaCT) molecule from the endogenous TF locus, provide conclusive proof that the cytosolic domain of TF is not essential for signal transduction in embryogenesis and in physiological postnatal processes., (Copyright 2001 Academic Press.)
- Published
- 2001
- Full Text
- View/download PDF
193. Mast cells and angiogenesis.
- Author
-
Levi-Schaffer F and Pe'er J
- Subjects
- Animals, Embryo, Mammalian blood supply, Endothelial Growth Factors immunology, Endothelial Growth Factors pharmacology, Fibroblast Growth Factor 2 immunology, Fibroblast Growth Factor 2 pharmacology, Humans, Lymphokines immunology, Lymphokines pharmacology, Vascular Endothelial Growth Factor A, Vascular Endothelial Growth Factors, Mast Cells physiology, Neovascularization, Pathologic physiopathology, Neovascularization, Physiologic physiology
- Published
- 2001
- Full Text
- View/download PDF
194. First trimester transabdominal embryo fetoscopy.
- Author
-
Miliou-Paouleskou D, Antsaklis A, Papantoniou N, Koutra P, and Daskalakis G
- Subjects
- Embryo, Mammalian blood supply, Female, Humans, Pregnancy, Pregnancy Trimester, First, Fetoscopy methods
- Abstract
The aim of this study was the visualization of the embryo and blood sampling through transabdominal embryoscopy during the first 12 week of pregnancy. Twenty women attending Alexandra Maternity Hospital, Athens for pregnancy termination were invited to participate in this study. Informed consent was obtained before participation. The gestational age ranged between 9 and 12 weeks from LMP. We used a Karl Storz semirigid fiberoptic endoscope 20 cm in length, with a diameter of 1 mm and an angle of view of 70 degrees. Under local anesthesia the endoscope was introduced transabdominally into the amniotic sac, using continuous ultrasound guidance. Complete examination of the embryo including head, face, limbs ventral and dorsal walls, genitalia, placenta, umbilical cord and yolk sac were performed. A 0,6 mm diameter puncture needle was inserted into the blood vessels of the chorionic plate or umbilical cord through the side arm of the canulla to obtain a small amount of blood. The complete anatomical survey was possible in all cases (100%). Access to the embryonic circulation was attempted in 14 cases. A small quantity of blood was obtained in 10 cases (71,4%). The average length of the procedure was 15 min. No maternal complications were recorded. D & C were performed 24 hours after the embryoscopy. Embryoscopy represent a diagnostic tool to confirm a diagnosis suspected through noninvasive means. Gaining access to the embryonic circulation when embryos are immunologically naïve will allow perhaps gene or cell therapy. Finally embryoscopy may be helpful to study the biology of embryonic development.
- Published
- 2001
195. Comparison of in vitro and in utero ethanol exposure on indices of oxidative stress.
- Author
-
Akella SS, Beck MJ, Philbert MA, and Harris C
- Subjects
- Administration, Oral, Animals, Culture Techniques, Embryo, Mammalian blood supply, Embryo, Mammalian drug effects, Embryonic and Fetal Development drug effects, Ethanol administration & dosage, Ethanol blood, Female, Neural Tube Defects blood, Neural Tube Defects etiology, Rats, Rats, Sprague-Dawley, Uterus blood supply, Ethanol toxicity, Oxidative Stress drug effects, Uterus drug effects
- Abstract
Prenatal ethanol exposure produces neural tube defects and growth retardation in experimental animals. Because ethanol's teratogenic effects may involve oxidative stress and effects may differ in vitro and in utero, glutathione, cysteine and ATP were evaluated in gestational day 10 rat conceptuses exposed to ethanol. Cultured embryos exposed to ethanol (1.5 or 3.0 mg/mL) maintained a concentration-dependent decrease in glutathione of 21 or 35%, respectively, at 6 h; visceral yolk sac (VYS) glutathione (GSH) decreased by 22 or 18%, respectively, at 3 h. Maternal ethanol exposure (4.5 g/kg) decreased glutathione by 30% in embryos and VYSs at 3 h, but values rebounded. Cultured embryonic cysteine decreased after 30 min by 42% with both doses and after 6 h by 32 or 38% with 1.5 or 3.0 mg/mL, respectively. Ethanol (1.5 mg/mL) increased VYS cysteine by 35% after 30 min. In utero ethanol exposure decreased embryonic cysteine by 58% at 3 h. Ethanol (1.5 mg/mL) decreased adenosine triphosphate (ATP) by 30-60% in embryos and VYSs at 30 min. After 6 h, embryonic ATP decreased by 41 and 30% with 1.5 and 3.0 mg/mL, respectively, while VYS ATP decreased by 38% with 1.5 mg/mL. In utero ethanol exposure decreased ATP by 31% at 3 h in VYSs. While decreases in GSH and cysteine were evident earlier in utero than in vitro, values returned to control suggesting embryos exposed in utero respond rapidly to chemical-induced oxidative stress due to maternal protective mechanisms. Differences between in vitro and in utero responses to ethanol have important implications for interpretation of in vitro developmental studies.
- Published
- 2000
196. VEGF-C signaling pathways through VEGFR-2 and VEGFR-3 in vasculoangiogenesis and hematopoiesis.
- Author
-
Hamada K, Oike Y, Takakura N, Ito Y, Jussila L, Dumont DJ, Alitalo K, and Suda T
- Subjects
- Animals, Blood Vessels abnormalities, Blood Vessels drug effects, Cell Differentiation drug effects, Cell Line, Coculture Techniques, Embryo, Mammalian blood supply, Embryo, Mammalian chemistry, Endothelial Growth Factors pharmacology, Humans, Immunohistochemistry, Mesoderm chemistry, Mesoderm cytology, Mice, Mice, Mutant Strains, Receptor Protein-Tyrosine Kinases metabolism, Receptor Protein-Tyrosine Kinases pharmacology, Receptor Protein-Tyrosine Kinases physiology, Receptors, Growth Factor metabolism, Receptors, Vascular Endothelial Growth Factor, Signal Transduction drug effects, Splanchnic Circulation, Stromal Cells chemistry, Stromal Cells cytology, Vascular Endothelial Growth Factor C, Vascular Endothelial Growth Factor Receptor-3, Yolk Sac blood supply, Endothelial Growth Factors physiology, Hematopoiesis drug effects, Neovascularization, Physiologic drug effects, Receptors, Growth Factor physiology
- Abstract
Signaling by vascular endothelial growth factors (VEGFs) through VEGF receptors (VEGFRs) plays important roles in vascular development and hematopoiesis. The authors analyzed the function of VEGF-C signaling through both VEGFR-2 and VEGFR-3 in vasculoangiogenesis and hematopoiesis using a coculture of para-aortic splanchnopleural mesoderm (P-Sp) explants from mouse embryos with stromal cells (OP9). Vasculogenesis and angiogenesis were evaluated by the extent of vascular bed and network formation, respectively. Addition of VEGF-C to the P-Sp culture enhanced vascular bed formation and suppressed definitive hematopoiesis. Both vascular bed and network formations were completely suppressed by addition of soluble VEGFR-1-Fc competitor protein. Formation of vascular beds but not networks could be rescued by VEGF-C in the presence of the competitor, while both were rescued by VEGF-A. VEGFR-3-deficient embryos show the abnormal vasculature and severe anemia. Consistent with these in vivo findings, vascular bed formation in the P-Sp from the VEGFR-3-deficient embryos was enhanced to that in wild-type or heterozygous embryos, and hematopoiesis was severely suppressed. When VEGFR-3-Fc chimeric protein was added to trap endogenous VEGF-C in the P-Sp culture of the VEGFR-3-deficient embryos, vascular bed formation was suppressed and hematopoiesis was partially rescued. These results demonstrate that because VEGF-C signaling through VEGFR-2 works synergistically with VEGF-A, the binding of VEGF-C to VEGFR-3 consequently regulates VEGFR-2 signaling. In VEGFR-3-deficient embryos, an excess of VEGF-C signals through VEGFR-2 induced the disturbance of vasculogenesis and hematopoiesis during embryogenesis. This indicates that elaborated control through VEGFR-3 signaling is critical in vasculoangiogenesis and hematopoiesis. (Blood. 2000;96:3793-3800)
- Published
- 2000
197. Cytokeratins 8 and 19 in the mouse placental development.
- Author
-
Tamai Y, Ishikawa T, Bösl MR, Mori M, Nozaki M, Baribault H, Oshima RG, and Taketo MM
- Subjects
- Animals, Crosses, Genetic, Embryo, Mammalian blood supply, Embryo, Mammalian embryology, Embryo, Mammalian metabolism, Female, Fetal Death, Fetal Growth Retardation, Fluorescent Antibody Technique, Galactosidases genetics, Galactosidases metabolism, Gene Deletion, Gene Targeting, Genes, Reporter, Genotype, Germ-Line Mutation genetics, In Situ Hybridization, Keratins genetics, Male, Mice, Mice, Inbred C57BL, Mice, Inbred Strains, Mice, Knockout, Phenotype, Placenta blood supply, Placenta pathology, Placental Circulation, Pregnancy, RNA, Messenger analysis, RNA, Messenger genetics, Recombination, Genetic, Stem Cells metabolism, Trophoblasts metabolism, Trophoblasts pathology, Keratins metabolism, Placenta embryology, Placenta metabolism
- Abstract
To investigate the expression and biological roles of cytokeratin 19 (K19) in development and in adult tissues, we inactivated the mouse K19 gene (Krt1-19) by inserting a bacterial beta-galactosidase gene (lacZ) by homologous recombination in embryonic stem cells, and established germ line mutant mice. Both heterozygous and homozygous mutant mice were viable, fertile, and appeared normal. By 7.5-8.0 days post coitum (dpc), heterozygous mutant embryos expressed lacZ in the notochordal plate and hindgut diverticulum, reflecting the fact that the notochord and the gut endoderm are derived from the axial mesoderm-originated cells. In the adult mutant, lacZ was expressed mainly in epithelial tissues. To investigate the possible functional cooperation and synergy between K19 and K8, we then constructed compound homozygous mutants, whose embryos died approximately 10 dpc. The lethality resulted from defects in the placenta where both K19 and K8 are normally expressed. As early as 9. 5 dpc, the compound mutant placenta had an excessive number of giant trophoblasts, but lacked proper labyrinthine trophoblast or spongiotrophoblast development, which apparently caused flooding of the maternal blood into the embryonic placenta. These results indicate that K19 and K8 cooperate in ensuring the normal development of placental tissues.
- Published
- 2000
- Full Text
- View/download PDF
198. Maintenance of vascular integrity in the embryo requires signaling through the fibroblast growth factor receptor.
- Author
-
Lee SH, Schloss DJ, and Swain JL
- Subjects
- Animals, Apoptosis, Blotting, Western, Cell Movement, Cells, Cultured, Humans, Mice, Mitogen-Activated Protein Kinases metabolism, Phosphorylation, Receptors, Fibroblast Growth Factor genetics, Transgenes, Yolk Sac blood supply, Embryo, Mammalian blood supply, Receptors, Fibroblast Growth Factor physiology
- Abstract
Basic fibroblast growth factor (FGF)-2 is important for vessel formation and/or maintenance of vascular integrity in the embryo. FGF signaling may be mediated through transmembrane tyrosine kinase receptors or directly through intracellular pathways that do not involve receptor activation. To determine the role of receptor-mediated signaling in endothelial cells, an adenovirus encoding truncated FGF receptor (FGFR)-1, under the control of the cytomegalovirus promoter, was expressed in endothelial cells. FGF signaling was impaired, as indicated by inhibition of MAPK phosphorylation. Functional consequences included inhibition of endothelial cell migration and induction of apoptosis. To address the role of endothelial FGFR signaling in vascular development, recombinant adenovirus encoding a dominant-negative FGFR was injected into the sinus venosus of embryonic day 9.0 cultured mouse embryos. Previous studies demonstrated that transgenes delivered via adenovirus, under the control of the cytomegalovirus promoter, are expressed selectively in the developing vasculature. Embryos expressing a control adenovirus developed normally, whereas those expressing the FGFR-1 mutant exhibited abnormal embryonic and extra-embryonic vascular development. These data demonstrate that FGF, by signaling through the FGFR, plays a pivotal role in the development and maintenance of a mature vascular network in the embryo.
- Published
- 2000
- Full Text
- View/download PDF
199. Essential role for p38alpha mitogen-activated protein kinase in placental angiogenesis.
- Author
-
Mudgett JS, Ding J, Guh-Siesel L, Chartrain NA, Yang L, Gopal S, and Shen MM
- Subjects
- Animals, Base Sequence, DNA Primers, Embryo, Mammalian blood supply, In Situ Nick-End Labeling, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, p38 Mitogen-Activated Protein Kinases, Mitogen-Activated Protein Kinases physiology, Neovascularization, Physiologic physiology, Placenta blood supply
- Abstract
The p38 family of mitogen-activated protein kinases (MAPKs) mediates signaling in response to environmental stresses and inflammatory cytokines, but the requirements for the p38 MAPK pathway in normal mammalian development have not been elucidated. Here, we show that targeted disruption of the p38alpha MAPK gene results in homozygous embryonic lethality because of severe defects in placental development. Although chorioallantoic placentation is initiated appropriately in p38alpha null homozygotes, placental defects are manifest at 10.5 days postcoitum as nearly complete loss of the labyrinth layer and significant reduction of the spongiotrophoblast. In particular, p38alpha mutant placentas display lack of vascularization of the labyrinth layer as well as increased rates of apoptosis, consistent with a defect in placental angiogenesis. Furthermore, p38alpha mutants display abnormal angiogenesis in the embryo proper as well as in the visceral yolk sac. Thus, our results indicate a requirement for p38alpha MAPK in diploid trophoblast development and placental vascularization and suggest a more general role for p38 MAPK signaling in embryonic angiogenesis.
- Published
- 2000
- Full Text
- View/download PDF
200. Does fetal antigen 1 (FA1) identify cells with regenerative, endocrine and neuroendocrine potentials? A study of FA1 in embryonic, fetal, and placental tissue and in maternal circulation.
- Author
-
Floridon C, Jensen CH, Thorsen P, Nielsen O, Sunde L, Westergaard JG, Thomsen SG, and Teisner B
- Subjects
- Biomarkers analysis, Biomarkers blood, Ectoderm metabolism, Embryo, Mammalian chemistry, Embryo, Mammalian cytology, Endoderm metabolism, Female, Fetus blood supply, Fetus chemistry, Fetus cytology, Fetus metabolism, Glycoproteins analysis, Glycoproteins blood, Humans, Immunohistochemistry, Mesoderm metabolism, Molecular Sequence Data, Placenta blood supply, Placenta chemistry, Placenta cytology, Placenta metabolism, Pregnancy, Embryo, Mammalian blood supply, Embryo, Mammalian metabolism, Endocrine System metabolism, Glycoproteins metabolism, Placental Circulation, Regeneration
- Abstract
Fetal antigen 1 (FA1) is a circulating EGF multidomain glycoprotein. FA1 and its membrane-associated precursor is defined by the mRNAs referred to as delta-like (dlk), preadipocyte factor 1 (pref-1) or zona glomerulosa-specific factor (ZOG). Using a polyclonal antibody recognising both forms, the localisation of FA1/dlk was analysed in embryonic and fetal tissues between week 5 to 25 of gestation and related to germinal origin and development. FA1 was observed in endodermally derived hepatocytes, glandular cells of the pancreas anlage, and in respiratory epithelial cells. FA1 was also present in mesodermally derived cells of the renal proximal tubules, adrenal cortex, Leydig and Hilus cells of the testes and ovaries, fetal chondroblasts, and skeletal myotubes. Ectodermally derived neuro- and adenohypophysial cells, cells in the floor of the 3rd ventricle and plexus choroideus were also FA1 positive. The number of cells expressing FA1 decreased during fetal development where the expression became restricted to specific functional cells. Epidermis, gut epithelium, gall bladder, blood cells, spleen, thyroid gland, salivary glands, and smooth muscle cells were FA1 negative. Analysis of extra-embryonic tissues from normal and pathological pregnancies revealed FA1 in stromal cells surrounding the blood islands of the yolk sac as well as in placental fibroblasts where the expression was most pronounced in diploid, androgenic complete hydatidiform moles. However, as measured by ELISA, the circulating maternal FA1 levels in complete moles were not different from normal pregnancies. The results presented suggest that FA1 is a growth and/or differentiation factor extensively expressed in immature cells and down-regulated during fetal development. FA1 down-regulation was associated with a shift in the subcellular localisation indicating differential post-translational/post-transcriptional modifications during fetal development. FA1 may be a new marker of cellular subtypes with a regenerative potential and of specific cells with endocrine or neuroendocrine functions.
- Published
- 2000
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.