Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
Medicine (Baltimore) ; 97(26): e11043, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29952943

ABSTRACT

The traditional surgical approach for removing a symptomatic urachal remnant is via a lower midline laparotomy and infraumbilical incision or a laparoscopic approach with umbilicoplasty. We reviewed our experience with umbilicus-sparing laparoscopic urachal remnant excision in a single-center study and evaluated its efficacy versus open approach (OA). This study was a retrospective study. Between March 2012 and September 2016, 32 consecutive patients with symptomatic urachal remnants underwent the umbilicus-sparing laparoscopic approach (USLA) (n = 17) or OA (n = 15). The efficacy, recovery, and long-term outcomes were reviewed. Our Results showed that the clinical characteristics of the patients in each group, such as age, gender, body mass index (BMI), and disease type, had no significant differences (P > .05). No significant difference was found in the surgical procedure times (76.1 ±â€Š15.4 vs 69.2 ±â€Š13.9 minutes, P = .189) and intraoperative blood loss (29.4 ±â€Š13.3 vs 32.2 ±â€Š12.9 mL, P = .543) between the USLA groups and OA groups. However, the mean postoperative hospital stay (patients with bladder cuff excision: 4.1 ±â€Š1.8 vs 6.1 ±â€Š1.4 days, P = .040 and patients without bladder cuff excision: 1.8 ±â€Š0.5 vs 3.6 ±â€Š0.8 days, P < .001) and the time of full recovery (11.2 ±â€Š1.9 vs 15.6 ±â€Š3.1 days, P < .001), the USLA group were both significantly shorter than that of the OA group. No infected recurrence and malignant transformation had occurred at a mean follow-up of 32.4 ±â€Š8.1 and 34.1 ±â€Š8.8 months in USLA group and OA group, respectively. In conclusion, to minimize the morbidity of radical excision, umbilicus-sparing management of benign urachal remnants in adults is a safe and efficacious alternative with superior cosmetic outcomes, postoperative recovery compared with an OA or umbilicoplasty.


Subject(s)
Allantois/embryology , Laparoscopy/methods , Umbilicus/surgery , Urachal Cyst/surgery , Urachus/surgery , Adolescent , Adult , Aftercare , Blood Loss, Surgical/statistics & numerical data , Cystoscopy/methods , Female , Humans , Length of Stay/statistics & numerical data , Male , Minimally Invasive Surgical Procedures/methods , Operative Time , Retrospective Studies , Tomography, X-Ray Computed/methods , Treatment Outcome , Urachal Cyst/epidemiology , Urachus/abnormalities , Young Adult
2.
Dev Biol ; 425(2): 208-222, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28389228

ABSTRACT

How the fetal-placental arterial connection is made and positioned relative to the embryonic body axis, thereby ensuring efficient and directed blood flow to and from the mother during gestation, is not known. Here we use a combination of genetics, timed pharmacological inhibition in living mouse embryos, and three-dimensional modeling to link two novel architectural features that, at present, have no status in embryological atlases. The allantoic core domain (ACD) is the extraembryonic extension of the primitive streak into the allantois, or pre-umbilical tissue; the vessel of confluence (VOC), situated adjacent to the ACD, is an extraembryonic vessel that marks the site of fetal-placental arterial union. We show that genesis of the fetal-placental connection involves the ACD and VOC in a series of steps, each one dependent upon the last. In the first, Brachyury (T) ensures adequate extension of the primitive streak into the allantois, which in turn designates the allantoic-yolk sac junction. Next, the streak-derived ACD organizes allantoic angioblasts to the axial junction; upon signaling from Fibroblast Growth Factor Receptor-1 (FGFR1), these endothelialize and branch, forming a sprouting VOC that unites the umbilical and omphalomesenteric arteries with the fetal dorsal aortae. Arterial union is followed by the appearance of the medial umbilical roots within the VOC, which in turn designate the correct axial placement of the lateral umbilical roots/common iliac arteries. In addition, we show that the ACD and VOC are conserved across Placentalia, including humans, underscoring their fundamental importance in mammalian biology. We conclude that T is required for correct axial positioning of the VOC via the primitive streak/ACD, while FGFR1, through its role in endothelialization and branching, further patterns it. Together, these genetic, molecular and structural elements safeguard the fetus against adverse outcomes that can result from vascular mispatterning of the fetal-placental arterial connection.


Subject(s)
Arteries/embryology , Fetal Proteins/metabolism , Fetus/embryology , Gastrula/blood supply , Gastrula/metabolism , Morphogenesis , Placenta/embryology , T-Box Domain Proteins/metabolism , Allantois/embryology , Allantois/metabolism , Animals , Arteries/metabolism , Endothelium, Vascular/metabolism , Female , Fetus/metabolism , Gastrula/embryology , Mice , Models, Biological , Placenta/metabolism , Pregnancy , Primitive Streak/embryology , Primitive Streak/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Umbilical Arteries/embryology , Umbilical Arteries/metabolism , Vascular Remodeling , Yolk Sac/metabolism
3.
Dev Biol ; 425(1): 44-57, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28322735

ABSTRACT

The allantois-derived umbilical component of the chorio-allantoic placenta shuttles fetal blood to and from the chorion, thereby ensuring fetal-maternal exchange. The progenitor populations that establish and supply the fetal-umbilical interface lie, in part, within the base of the allantois, where the germ line is claimed to segregate from the soma. Results of recent studies in the mouse have reported that STELLA (DPPA-3, PGC7) co-localizes with PRDM1 (BLIMP1), the bimolecular signature of putative primordial germ cells (PGCs) throughout the fetal-placental interface. Thus, if PGCs form extragonadally within the posterior region of the mammal, they cannot be distinguished from the soma on the basis of these proteins. We used immunohistochemistry, immunofluorescence, and confocal microscopy of the mouse gastrula to co-localize STELLA with a variety of gene products, including pluripotency factor OCT-3/4, mesendoderm-associated T and MIXl1, mesendoderm- and endoderm-associated FOXa2 and hematopoietic factor Runx1. While a subpopulation of cells localizing OCT-3/4 was always found independently of STELLA, STELLA always co-localized with OCT-3/4. Despite previous reports that T is involved in specification of the germ line, co-localization of STELLA and T was detected only in a small subset of cells in the base of the allantois. Slightly later in the hindgut lip, STELLA+/(OCT-3/4+) co-localized with FOXa2, as well as with RUNX1, indicative of definitive endoderm and hemangioblasts, respectively. STELLA was never found with MIXl1. On the basis of these and previous results, we conclude that STELLA identifies at least five distinct cell subpopulations within the allantois and hindgut, where they may be involved in mesendodermal differentiation and hematopoiesis at the posterior embryonic-extraembryonic interface. These data provide a new point of departure for understanding STELLA's potential roles in building the fetal-placental connection.


Subject(s)
Embryo, Mammalian/metabolism , Endoderm/metabolism , Gastrula/metabolism , Repressor Proteins/metabolism , Allantois/cytology , Allantois/embryology , Allantois/metabolism , Animals , Chromosomal Proteins, Non-Histone , Core Binding Factor Alpha 2 Subunit/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Endoderm/cytology , Endoderm/embryology , Female , Fetal Proteins/metabolism , Fetus/embryology , Fetus/metabolism , Gastrula/embryology , Hepatocyte Nuclear Factor 3-beta/metabolism , Homeodomain Proteins/metabolism , Immunohistochemistry , Mice , Microscopy, Confocal , Octamer Transcription Factor-3/metabolism , Placenta/embryology , Placenta/metabolism , Positive Regulatory Domain I-Binding Factor 1 , Pregnancy , T-Box Domain Proteins/metabolism , Transcription Factors/metabolism
4.
J Morphol ; 278(5): 675-688, 2017 05.
Article in English | MEDLINE | ID: mdl-28145561

ABSTRACT

Ultrastructure of the placental tissues from redbelly watersnakes (Nerodia erythrogaster) was analyzed during late pregnancy to provide insight into placental development and function. Examination of the chorioallantoic placenta with transmission electron microscopy reveals that chorionic and uterine epithelia are extremely attenuated but intact and that the eggshell membrane is vestigial and lacks a calcareous layer. These features minimize the interhemal diffusion distance across the placenta. Scanning electron microscopy reveals that fetal and maternal components of the placentas are richly vascularized by dense networks of capillaries. Although the yolk sac omphalopleure has largely been replaced by chorioallantois by late gestation, it retains patches of yolk droplets and regions of absorptive cells with microvilli and abundant mitochondria. Transmission electron microscopy reveals that yolk material is taken up for digestion by endodermal cells. As yolk is removed, allantoic capillaries invade to occupy positions just beneath the epithelium, forming regions of chorioallantoic placentation. Ultrastructural features indicate that the chorioallantoic placenta is specialized for gas exchange, while the omphalallantoic ("yolk sac") placenta shows evidence of functions in yolk digestion and maternal-fetal nutrient transfer. Placental features of this species are consistent with those of other thamnophines, and are evolutionarily convergent on snakes of other viviparous clades.


Subject(s)
Colubridae/embryology , Placenta/ultrastructure , Placentation/physiology , Allantois/embryology , Allantois/ultrastructure , Animals , Biological Evolution , Chorion/embryology , Chorion/ultrastructure , Female , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Pregnancy , Yolk Sac/embryology , Yolk Sac/ultrastructure
5.
J Morphol ; 278(5): 665-674, 2017 05.
Article in English | MEDLINE | ID: mdl-28145572

ABSTRACT

As part of a broad survey of placental structure, function, and evolution in reptilian sauropsids paraffin-section histology was used to study microscopic anatomy of the uterus and fetal membranes of three species of North American watersnakes (Nerodia: Colubridae). The pre-ovulatory uterus is poorly vascularized with inactive shell glands. These shell glands are activated during vitellogenesis but regress during pregnancy. Two placentas develop through apposition of the uterine lining to the chorioallantois and the yolk sac omphalopleure. Fetal and maternal components of the chorioallantoic placenta are progressively vascularized during development. Their epithelia are attenuated, but (contrary to a previous report), epithelia of neither the uterus nor the chorion are eroded. The fetal portion of the yolk sac placenta is an omphalallantois, formed of avascular omphalopleure, isolated yolk mass, and allantois. This placenta is progressively replaced by chorioallantoic placenta during mid- to late-development through depletion of the isolated yolk mass. The chorioallantoic placenta is anatomically specialized for maternal-fetal gas exchange, and its expansion during development reflects the growing needs of the fetus for gas exchange. The yolk sac placenta is morphologically unsuited for gas exchange, but may serve other functions in maternal-fetal exchange.


Subject(s)
Colubridae/anatomy & histology , Placenta/embryology , Placentation/physiology , Allantois/embryology , Allantois/ultrastructure , Animals , Biological Evolution , Chorion/embryology , Chorion/ultrastructure , Extraembryonic Membranes/ultrastructure , Female , Mammals , Placenta/ultrastructure , Pregnancy , United States , Yolk Sac/embryology , Yolk Sac/ultrastructure
6.
J Morphol ; 278(5): 600-620, 2017 May.
Article in English | MEDLINE | ID: mdl-28165148

ABSTRACT

Despite its conserved role in placenta and umbilical cord formation, the mammalian allantois shows remarkable diversity in size and form as well as in the timing of its appearance and attachment to the chorion. In the mouse, the common allantoic diverticulum is lacking; instead, the allantoic core domain is defined as a progenitor center for allantoic development. In this study, the allantoises of the pig and the rabbit as two nonrodent mammals of increasing significance in biomedical research are compared (1) morphologically using high resolution light and electron microscopy and (2) molecularly using brachyury mRNA expression as a mesodermal marker. Multiple small allantoic diverticula in the rabbit contrast with a single large cavity filling the entire allantois of the pig, but neither pig nor rabbit allantois expresses brachyury. The mesothelium on the allantois surface shows regional variability of cell contacts and microvilli, while blood vessels appear randomly around the allantoic diverticula in a mesodermal layer of variable thickness. Primordial germ cell-like cells are found in the allantois of the pig but not of the rabbit. To understand further the relevance of this developmental and morphological diversity, we compare the allantois development of pig and rabbit with early developmental landmarks of mouse and man. Our findings suggest that (1) tissue interaction between endoderm and mesoderm is important for allantoic development and vascular differentiation in species with a rudimentary allantoic diverticulum, (2) allantoic mesothelium plays a specific role in chorioallantoic attachment, allantoic differentiation and vascularization, and (3) there is a pronounced diversity in the extraembryonic migratory pathways of primordial germ cells among mammals. Finally, the phylogenetically basal characteristics of the pig allantois are suggestive of a functional similarity in mammals with a large allantois before placentation and in (aplacental) sauropsids with a chorioallantoic membrane well-adjusted to material exchange function.


Subject(s)
Allantois/embryology , Animals , Female , Mice , Pregnancy , Rabbits , Swine
7.
Dev Biol ; 418(1): 89-97, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27514653

ABSTRACT

The embryonic-maternal interface of the placental labyrinth, allantois, and yolk sac are vital during embryogenesis; however, the precise mechanism underlying the vascularization of these structures remains unknown. Herein we focus on the role of inositol 1,4,5-trisphosphate (IP3) receptors (IP3R), which are intracellular Ca(2+) release channels, in placentation. Double knockout (DKO) of type 1 and 3 IP3Rs (IP3R1 and IP3R3, respectively) in mice resulted in embryonic lethality around embryonic day (E) 11.5. Because IP3R1 and IP3R3 were co-expressed in endothelial cells in the labyrinth, allantois, and yolk sac, we investigated extra-embryonic vascular development in IP3R1- and IP3R3-DKO mice. The formation of chorionic plates and yolk sac vessels seemed dysregulated around the timing of the chorio-allantoic attachment, immediately followed by the disorganization of allantoic vessels, the decreased expression of the spongiotrophoblast cell marker Tpbpa and the growth retardation of the embryos in DKO mice. Fluorescent immunohistochemistry demonstrated downregulation of a vascular endothelial marker, CD31, in labyrinth embryonic vessels and poor elongation of extra-embryonic mesoderm into the labyrinth layer in DKO placenta, whereas the branching of the DKO chorionic trophoblast was initiated. In addition, allantoic and yolk sac vessels in extra-embryonic tissues were less remodeled in DKO mice. In vitro endothelial cord formation and migration activities of cultured vascular endothelial cells derived from human umbilical vein were downregulated under the inhibition of IP3R. Our results suggest that IP3R1 and IP3R3 are required for extra-embryonic vascularization in the placenta, allantois, and yolk sac. This is the first demonstration of the essential role of IP3/IP3Rs signaling in the development of the vasculature at the embryonic-maternal interface.


Subject(s)
Allantois/blood supply , Inositol 1,4,5-Trisphosphate Receptors/genetics , Neovascularization, Physiologic/genetics , Placenta/blood supply , Placentation/genetics , Umbilical Veins/embryology , Yolk Sac/blood supply , Allantois/embryology , Animals , Cell Line , Embryonic Development , Endothelial Cells/metabolism , Female , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Knockout , Placenta/embryology , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , Pregnancy , Trophoblasts/cytology , Umbilical Veins/cytology , Yolk Sac/embryology
8.
J Exp Zool B Mol Dev Evol ; 326(5): 290-302, 2016 07.
Article in English | MEDLINE | ID: mdl-27373551

ABSTRACT

In eggs of oviparous reptiles, fetal membranes maintain developing embryos through the exchange of respiratory gases and provision of water and calcium. As part of a survey of reptilian fetal membranes, we used scanning electron microscopy to study fetal membrane morphology in the oviparous Pueblan milksnake, Lampropeltis triangulum campbelli. The chorioallantois initially is an avascular structure lined by enlarged chorionic and allantoic epithelia. Upon vascularization, the chorionic epithelium becomes greatly attenuated, enhancing the potential for gas exchange; the allantoic epithelium also flattens. The bilaminar omphalopleure of the yolk sac lacks blood vessels, but it becomes vascularized by allantoic capillaries and transformed into an omphalallantois. Upon regression of the isolated yolk mass, this membrane is converted to chorioallantois, equipping it for gas exchange. Allantoic fluid serves as a water reservoir, and we postulate that it facilitates water uptake by establishing an osmotic gradient. Early in development, epithelia of both the chorion and the omphalopleure show apical microvilli that greatly increase the cell surface area available for water uptake. However, these features are incompatible with gas exchange and are lost as oxygen needs take precedence. A comparison of the fetal membranes to those of other squamate species (both oviparous and viviparous) reveals characteristics that are probably ancestral for snakes, some of which are plesiomorphic for Squamata. The widespread phylogenetic distribution of these features reflects their utility as adaptations that serve functional requirements of squamate embryos.


Subject(s)
Chorioallantoic Membrane/ultrastructure , Colubridae/embryology , Extraembryonic Membranes/ultrastructure , Allantois/embryology , Allantois/ultrastructure , Animals , Biological Evolution , Chorioallantoic Membrane/blood supply , Chorioallantoic Membrane/embryology , Extraembryonic Membranes/embryology , Oviparity , Ovum/growth & development , Ovum/ultrastructure , Yolk Sac/embryology , Yolk Sac/ultrastructure
9.
Dev Growth Differ ; 55(1): 1-14, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23278669

ABSTRACT

The chick embryo is as ancient a source of knowledge on animal development as the very beginning of embryology. Already, at the time of Caspar Friedrich Wolff, contemplating the strikingly beautiful scenario of the germ deploying on the yellow background of the yolk inspired and supported the tenants of epigenesis at the expense of the preformation theory. In this article, we shall mention some of the many problems of developmental biology that were successfully clarified by research on chick embryos. Two topics, the development of the neural system and that of blood and blood vessels, familiar to the authors, will be discussed in more detail.


Subject(s)
Chick Embryo/embryology , Embryo, Nonmammalian/blood supply , Hematopoietic Stem Cells/cytology , Nerve Net/embryology , Allantois/embryology , Allantois/metabolism , Animals , Biological Evolution , Brain/embryology , Brain/metabolism , Cell Lineage , Cell Movement , Developmental Biology/methods , Embryo, Nonmammalian/immunology , Embryo, Nonmammalian/metabolism , Hematopoietic Stem Cells/metabolism , Nerve Net/blood supply , Nerve Net/metabolism , Neural Crest/embryology , Neural Crest/metabolism , Species Specificity , T-Lymphocytes, Regulatory/immunology , Yolk Sac/blood supply , Yolk Sac/metabolism
10.
Dev Biol ; 371(2): 227-34, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22960234

ABSTRACT

Cdx gene products regulate the extent of axial elongation from the posterior growth zone. These transcription factors sustain the emergence of trunk and tail tissues by providing a suitable niche in the axial progenitor zone, via regulation of Wnt signaling. Cdx genes are expressed in and along the complete primitive streak including its posterior part wherefrom the extraembryonic mesoderm of the allantois emerges. Cdx genes are required for the full development of the allantois and its derivatives in the placental labyrinth. The mouse germ cell lineage also originates from the proximo-posterior epiblast of the primitive streak, and is established within the extraembryonic mesoderm that generates the allantois. We asked whether the expression of Cdx genes around the newly specified PGCs is necessary for the maintenance and expansion of this population, as it is for the allantois and axial progenitors. We observed a significantly lower number of PGCs in Cdx2(null) embryos than in controls. We found that Wnt3a loss of function decreases the PGC population to the same extent as Cdx2 inactivation. Moreover, exogenous Wnt3a corrects the lower PGC number in Cdx2(null) posterior embryonic tissues cultured in vitro. Cdx2 is not expressed in PGCs themselves, and we propose that the expression of Cdx2 in posterior extraembryonic tissues contributes to the proper niche of the germ cell progenitors by stimulating canonical Wnt signaling. Since PGC residence within the posterior growth zone is a mouse-specific feature, our data suggest that mouse PGCs opportunistically became dependent on the axial progenitor niche.


Subject(s)
Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Germ Cells/cytology , Homeodomain Proteins/genetics , Transcription Factors/genetics , Allantois/cytology , Allantois/embryology , Allantois/metabolism , Animals , CDX2 Transcription Factor , Embryo, Mammalian/cytology , Germ Cells/metabolism , Homeodomain Proteins/metabolism , Mice , Mice, Knockout , Transcription Factors/metabolism
11.
BMJ Case Rep ; 20122012 Aug 27.
Article in English | MEDLINE | ID: mdl-22927270

ABSTRACT

Allantiod cysts are true cysts in the umbilical cord. They arise from persistent structures of allantois, and the cysts are filled with urine because of a connection to the urinary bladder. Allantoid cysts are located centrally in the umbilical cord and separate the umbilical cord vessels. B-flow ultrasound is a new technique illustrating blood flow. This method is independent of the insonation angle and therefore superior to Doppler ultrasound visualising long segments of vessels with a winding course. The authors present a case with allantoid cysts diagnosed with B-flow ultrasound.


Subject(s)
Allantois/diagnostic imaging , Image Processing, Computer-Assisted , Imaging, Three-Dimensional , Ultrasonography, Prenatal , Umbilical Cord/diagnostic imaging , Urachal Cyst/diagnostic imaging , Adult , Allantois/embryology , Allantois/pathology , Cesarean Section , Diagnosis, Differential , Endothelium/pathology , Epithelium/pathology , Female , Humans , Infant, Newborn , Male , Placenta/diagnostic imaging , Pregnancy , Pregnancy Trimester, First , Pregnancy Trimester, Second , Rupture, Spontaneous , Umbilical Cord/embryology , Umbilical Cord/pathology , Urachal Cyst/embryology , Urachal Cyst/pathology
12.
Blood ; 120(13): 2562-72, 2012 Sep 27.
Article in English | MEDLINE | ID: mdl-22855605

ABSTRACT

The allantois is the embryonic precursor of the umbilical cord in mammals and is one of several embryonic regions, including the yolk sac and dorsal aorta, that undergoes vasculogenesis, the de novo formation of blood vessels. Despite its importance in establishing the chorioallantoic placenta and umbilical circulation, the allantois frequently is overlooked in embryologic studies. Nonetheless, recent studies demonstrate that vasculogenesis, vascular remodeling, and angiogenesis are essential allantois functions in the establishment of the chorioallantoic placenta. Here, we review blood vessel formation in the murine allantois, highlighting the expression of genes and involvement of pathways common to vasculogenesis or angiogenesis in other parts of the embryo. We discuss experimental techniques available for manipulation of the allantois that are unavailable for yolk sac or dorsal aorta, and review how this system has been used as a model system to discover new genes and mechanisms involved in vessel formation. Finally, we discuss the potential of the allantois as a model system to provide insights into disease and therapeutics.


Subject(s)
Allantois/blood supply , Allantois/embryology , Blood Vessels/growth & development , Models, Animal , Neovascularization, Physiologic , Allantois/metabolism , Animals , Mice
13.
J Morphol ; 273(10): 1163-84, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22730070

ABSTRACT

Development of the yolk sac of squamate reptiles (lizards and snakes) differs from other amniote lineages in the pattern of growth of extraembryonic mesoderm, which produces a cavity, the yolk cleft, within the yolk. The structure of the yolk cleft and the accompanying isolated yolk mass influence development of the allantois and chorioallantoic membrane. The yolk cleft of viviparous species of the Eugongylus group of scincid lizards is the foundation for an elaborate yolk sac placenta; development of the yolk cleft of oviparous species has not been studied. We used light microscopy to describe the yolk sac and chorioallantoic membrane in a developmental series of an oviparous member of this species group, Oligosoma lichenigerum. Topology of the extraembryonic membranes of late stage embryos differs from viviparous species as a result of differences in development of the yolk sac. The chorioallantoic membrane encircles the egg of O. lichenigerum but is confined to the embryonic hemisphere of the egg in viviparous species. Early development of the yolk cleft is similar for both modes of parity, but in contrast to viviparous species, the yolk cleft of O. lichenigerum is transformed into a tube-like structure, which fills with cells. The yolk cleft originates as extraembryonic mesoderm is diverted from the periphery of the egg into the yolk sac cavity. As a result, a bilaminar omphalopleure persists over the abembryonic surface of the yolk. The bilaminar omphalopleure is ultimately displaced by intrusion of allantoic mesoderm between ectodermal and endodermal layers. The resulting chorioallantoic membrane has a similar structure but different developmental history to the chorioallantoic membrane of the embryonic hemisphere of the egg.


Subject(s)
Lizards/embryology , Viviparity, Nonmammalian , Yolk Sac/cytology , Allantois/embryology , Animals , Chorioallantoic Membrane/ultrastructure , Chorion/embryology , Ectoderm , Endoderm , Extraembryonic Membranes/ultrastructure , Female , Mesoderm , Microscopy , Oviparity , Ovum , Reptiles , Snakes/embryology , Yolk Sac/embryology
14.
Dev Biol ; 363(1): 201-18, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22019303

ABSTRACT

The developmental relationship between the posterior embryonic and extraembryonic regions of the mammalian gastrula is poorly understood. Although many different cell types are deployed within this region, only the primordial germ cells (PGCs) have been closely studied. Recent evidence has suggested that the allantois, within which the PGCs temporarily take up residence, contains a pool of cells, called the Allantoic Core Domain (ACD), critical for allantoic elongation to the chorion. Here, we have asked whether the STELLA-positive cells found within this region, thought to be specified PGCs, are actually part of the ACD and to what extent they, and other ACD cells, contribute to the allantois and fetal tissues. To address these hypotheses, STELLA was immunolocalized to the mouse gastrula between Early Streak (ES) and 12-somite pair (-s) stages (~6.75-9.0 days post coitum, dpc) in histological sections. STELLA was found in both the nucleus and cytoplasm in a variety of cell types, both within and outside of the putative PGC trajectory. Fate-mapping the headfold-stage (~7.75-8.0 dpc) posterior region, by which time PGCs are thought to be segregated into a distinct lineage, revealed that the STELLA-positive proximal ACD and intraembryonic posterior primitive streak (IPS) contributed to a wide range of somatic tissues that encompassed derivatives of the three primary germ layers. This contribution included STELLA-positive cells localizing to tissues both within and outside of the putative PGC trajectory. Thus, while STELLA may identify a subpopulation of cells destined for the PGC lineage, our findings reveal that it may be part of a broader niche that encompasses the ACD and through which the STELLA population may contribute cells to a wide variety of posterior tissues of the mouse gastrula.


Subject(s)
Allantois/metabolism , Embryo, Mammalian/metabolism , Primitive Streak/metabolism , Repressor Proteins/metabolism , Allantois/cytology , Allantois/embryology , Animals , Cell Lineage , Cell Nucleus/metabolism , Chromosomal Proteins, Non-Histone , Cytoplasm/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Female , Gastrula/cytology , Gastrula/embryology , Gastrula/metabolism , Germ Cells/metabolism , Immunohistochemistry , Male , Mice , Pregnancy , Primitive Streak/embryology , Time Factors
15.
Dev Dyn ; 240(10): 2290-300, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21932311

ABSTRACT

T-box gene Tbx4 is critical for the formation of the umbilicus and the initiation of the hindlimb. Previous studies show broad expression in the allantois, hindlimb, lung and proctodeum. We have examined the expression of Tbx4 in detail and used a Tbx4-Cre line to trace the fates of Tbx4-expressing cells. Tbx4 expression and lineage reveal that various distinct appendages, such as the allantois, hindlimb, and external genitalia, all arise from a single mesenchymal expression domain. Additionally, although Tbx4 is associated primarily with the hindlimb, we find two forelimb expression domains. Most notably, we find that, despite the requirement for Tbx4 in allantoic vasculogenesis, the presumptive endothelial cells of the allantois do not express Tbx4 and lineage tracing reveals that the umbilical vasculature never expresses Tbx4. These results suggest that endothelial lineages are segregated before the onset of vasculogenesis, and demonstrate a role for the peri-vascular tissue in vasculogenesis.


Subject(s)
Allantois/cytology , Allantois/embryology , Extremities/embryology , Genitalia/embryology , Morphogenesis , T-Box Domain Proteins/metabolism , Allantois/physiology , Animals , Cell Lineage , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Extremities/anatomy & histology , Extremities/physiology , Female , Gene Expression Regulation, Developmental , Genitalia/cytology , Genitalia/physiology , Male , Mesoderm/cytology , Mesoderm/physiology , Mice , Pregnancy , T-Box Domain Proteins/genetics , Tissue Distribution
16.
J Morphol ; 272(4): 465-84, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21290418

ABSTRACT

Placental membranes mediate maternal-fetal exchange in all viviparous reptilian sauropsids. We used scanning electron microscopy to examine the placental interface in the mountain spiny lizard, Sceloporus jarrovi (Phrynosomatidae). From the late limb bud stage until birth, the conceptus is surrounded by placental membranes formed from the chorioallantois and yolk sac omphalopleure. The chorioallantois lies directly apposed to the uterine lining with no intervening shell membrane. Both fetal and maternal sides of the chorioallantoic placenta are lined by continuous layers of flattened epithelial cells that overlie dense capillary networks. The chorioallantoic placenta shows specializations that enhance respiratory exchange, as well as ultrastructural evidence of maternal secretion and fetal absorption. The yolk sac placenta contains enlarged fetal and maternal epithelia with specializations for histotrophic nutrient transfer. This placenta lacks intrinsic vascularity, although the vascular allantois lies against its inner face, contributing to an omphallantoic placenta. In a specialized region at the abembryonic pole, uterine and fetal tissues are separated by a compact mass of shed shell membrane, yolk droplets, and cellular debris. The omphalopleure in this region develops elongate folds that may contribute to sequestration and absorption of this material. Fetal membrane morphogenesis and composition in S. jarrovi are consistent with those of typical squamates. However, this species exhibits unusual placental specializations characteristic of highly placentotrophic lizards.


Subject(s)
Extraembryonic Membranes/ultrastructure , Lizards/anatomy & histology , Allantois/embryology , Allantois/ultrastructure , Animals , Chorioallantoic Membrane/anatomy & histology , Chorioallantoic Membrane/ultrastructure , Extraembryonic Membranes/anatomy & histology , Female , Lizards/embryology , Microscopy, Electron, Scanning , Uterus/embryology , Uterus/ultrastructure , Viviparity, Nonmammalian , Yolk Sac/anatomy & histology , Yolk Sac/ultrastructure
17.
Microcirculation ; 17(6): 447-57, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20690983

ABSTRACT

OBJECTIVE: To assess the impact of vascular endothelial growth factor (VEGF) on intussusceptive angiogenesis. METHODS AND RESULTS: Polyurethane casts of the microvasculature of chicken chorioallantoic membrane (CAM) were prepared on embryonic days (E) 8, 10, 12, and 14. At light microscopy level, minute holes (<2 microm in diameter) and hollows (>2 microm) were observed in the casts. Transmission electron microscopy disclosed the minute holes to mainly represent transluminal pillars characteristic for intussusceptive angiogenesis. The numerical density of the holes/pillars was highest at an early (E8) and a late (E12-E14) stage. Only mRNA of VEGF-A-122 and VEGF-A-166 isoforms was detected in the CAM. The transcription rate of VEGF-A mRNA peaked on E8/9 and E12, while VEGF-A protein expression increased on E8/9 and E11/12 to rapidly decrease thereafter as determined by immunoblotting. At all time points investigated, VEGF-A immunohistochemical reactivity was restricted to cells of the chorionic epithelium in direct contact to the capillary plexus. When the VEGF-R-inhibitor PTK787/ZK222584 (0.1 mg/mL) was applied on E9 CAM, the microvasculature topology on E12 was similar to that on E10. CONCLUSIONS: The temporal course of intussusception corresponded to the expression of VEGF-A in CAM microvasculature. Inhibition of VEGF-signaling retarded intussusceptive-dependent capillary maturation. These data suggest that VEGF promotes intussusception.


Subject(s)
Allantois/blood supply , Allantois/embryology , Chorion/blood supply , Chorion/embryology , Neovascularization, Physiologic , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Allantois/metabolism , Animals , Chick Embryo , Chorion/metabolism , Corrosion Casting , Microcirculation , Neovascularization, Physiologic/genetics , Phthalazines/pharmacology , Pyridines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors
18.
Bioessays ; 31(8): 892-902, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19609969

ABSTRACT

The primitive streak establishes the antero-posterior body axis in all amniote species. It is thought to be the conduit through which mesoderm and endoderm progenitors ingress and migrate to their ultimate destinations. Despite its importance, the streak remains poorly defined and one of the most enigmatic structures of the animal kingdom. In particular, the posterior end of the primitive streak has not been satisfactorily identified in any species. Unexpectedly, and contrary to prevailing notions, recent evidence suggests that the murine posterior primitive streak extends beyond the embryo proper. In its extraembryonic site, the streak creates a node-like cell reservoir from which the allantois, a universal caudal appendage of all amniotes and the future umbilical cord of placental mammals, emerges. This new insight into the fetal/umbilical relationship may explain the etiology of a large number of umbilical-associated birth defects, many of which are correlated with abnormalities of the embryonic midline.


Subject(s)
Body Patterning , Mammals/embryology , Primitive Streak/embryology , Allantois/cytology , Allantois/embryology , Animals , Mesoderm/embryology , Stem Cells/cytology
19.
Development ; 136(8): 1295-303, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19279135

ABSTRACT

Steel factor is an essential survival and proliferation factor for primordial germ cells (PGCs) during their migration in the early mouse embryo. PGCs arise during gastrulation, and migrate into the posterior endoderm that becomes the hindgut. Previous reports have suggested that PGCs become dependent on Steel factor when they colonize the hindgut. However, in the absence of a good marker for living PGCs, their behavior before hindgut colonization has not been previously studied. We report here the normal behavior of PGCs in live embryos before hindgut colonization, and the roles of Steel factor, using a reporter line in which GFP is driven by the promoter of the Stella gene, whose activation accompanies the initial specification of PGCs. We show first that PGCs are surrounded by Steel factor-expressing cells from their first appearance in the allantois to the time they enter the genital ridges. Second, fewer PGCs are found in the allantois in Steel-null embryos, but this is not due to a failure of PGC specification. Third, the analysis of cultured Steel-null early embryos shows that Steel factor is required for normal PGC motility, both in the allantois and in the hindgut. Germ cells migrate actively in the allantois, and move directionally from the allantois into the proximal epiblast. In the absence of Steel factor, caused by either null mutation or antibody blockade, PGC motility is dramatically decreased, but directionality is maintained, demonstrating a primary role for Steel factor in PGC motility. This was found both before and after colonization of the hindgut. These data, together with previously published data, show that PGCs are Steel factor dependent from their initial specification until they colonize the genital ridges, and suggest the existence of a ;spatio-temporal niche' that travels with this important pluripotential cell population in the embryo.


Subject(s)
Allantois/cytology , Allantois/metabolism , Germ Cells/cytology , Germ Cells/metabolism , Stem Cell Factor/metabolism , Stem Cell Niche/cytology , Stem Cell Niche/metabolism , Allantois/embryology , Animals , Cell Death , Cell Movement , Cell Survival , Digestive System/cytology , Digestive System/embryology , Digestive System/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Stem Cell Factor/genetics , Time Factors
20.
Dev Dyn ; 238(3): 532-53, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19191225

ABSTRACT

The whereabouts and properties of the posterior end of the primitive streak have not been identified in any species. In the mouse, the streak's posterior terminus is assumed to be confined to the embryonic compartment, and to give rise to the allantois, which links the embryo to its mother during pregnancy. In this study, we have refined our understanding of the biology of the murine posterior primitive streak and its relation to the allantois. Through a combination of immunostaining and morphology, we demonstrate that the primitive streak spans the posterior extraembryonic and embryonic regions at the onset of the neural plate stage ( approximately 7.0 days postcoitum, dpc). Several hours later, the allantoic bud emerges from the extraembryonic component of the primitive streak (XPS). Then, possibly in collaboration with overlying allantois-associated extraembryonic visceral endoderm, the XPS establishes a germinal center within the allantois, named here the Allantoic Core Domain (ACD). Microsurgical removal of the ACD beyond headfold (HF) stages resulted in the formation of allantoic regenerates that lacked the ACD and failed to elongate; nevertheless, vasculogenesis and vascular patterning proceeded. In situ and transplantation fate mapping demonstrated that, from HF stages onward, the ACD's progenitor pool contributed to the allantois exclusive of the proximal flanks. By contrast, the posterior intraembryonic primitive streak (IPS) provided the flanks. Grafting the ACD into T(C)/T(C) hosts, whose allantoises are significantly foreshortened, restored allantoic elongation. These results revealed that the ACD is essential for allantoic elongation, but the cues required for vascularization lie outside of it. On the basis of these and previous findings, we conclude that the posterior primitive streak of the mouse conceptus is far more complex than was previously believed. Our results provide new directives for addressing the origin and development of the umbilical cord, and establish a novel paradigm for investigating the fetal/placental relationship.


Subject(s)
Allantois/embryology , Primitive Streak/embryology , Allantois/blood supply , Allantois/metabolism , Allantois/transplantation , Animals , Body Patterning , Mice , Microscopy, Electron, Transmission , Vascular Endothelial Growth Factor Receptor-2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...