Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Philos Trans R Soc Lond B Biol Sci ; 377(1865): 20210251, 2022 12 05.
Article in English | MEDLINE | ID: mdl-36252214

ABSTRACT

During the early development of Placentalia, a distinctive projection emerges at the posterior embryonic-extraembryonic interface of the conceptus; its fingerlike shape presages maturation into the placental umbilical cord, whose major role is to shuttle fetal blood to and from the chorion for exchange with the mother during pregnancy. Until recently, the biology of the cord's vital vascular anlage, called the body stalk/allantois in humans and simply the allantois in rodents, has been largely unknown. Here, new insights into the development of the mouse allantois are featured, from its origin and mechanism of arterial patterning through its union with the chorion. Key to generating the allantois and its critical functions are the primitive streak and visceral endoderm, which together are sufficient to create the entire fetal-placental connection. Their newly discovered roles at the embryonic-extraembryonic interface challenge conventional wisdom, including the physical limits of the primitive streak, its function as sole purveyor of mesoderm in the mouse, potency of visceral endoderm, and the putative role of the allantois in the germ line. With this working model of allantois development, understanding a plethora of hitherto poorly understood orphan diseases in humans is now within reach. This article is part of the theme issue 'Extraembryonic tissues: exploring concepts, definitions and functions across the animal kingdom'.


Subject(s)
Allantois , Placenta , Allantois/blood supply , Animals , Embryo, Mammalian , Female , Humans , Mesoderm , Mice , Pregnancy , Primitive Streak
2.
Elife ; 92020 10 12.
Article in English | MEDLINE | ID: mdl-33044167

ABSTRACT

High levels of proteins called proteoglycans in the walls of umbilical arteries enable these arteries to close rapidly after birth and thus prevent blood loss in newborns.


Subject(s)
Proteoglycans , Umbilical Arteries , Cell Division , Humans , Infant, Newborn , Sex Characteristics
3.
Exp Hematol ; 89: 37-42, 2020 09.
Article in English | MEDLINE | ID: mdl-32735907

ABSTRACT

The extra-embryonic hypoblast/visceral endoderm of Placentalia carries out a variety of functions during gestation, including hematopoietic induction. Results of decades-old and recent experiments have provided compelling evidence that, in addition to its inducing properties, hypoblast/visceral endoderm itself is a source of placental blood cells. Those observations that highlight extra-embryonic endoderm's role as an overlooked source of placental blood cells across species are briefly discussed here, with suggestions for future exploration.


Subject(s)
Allantois/cytology , Blood Cells/cytology , Endoderm/cytology , Erythroblasts/cytology , Placenta/cytology , Yolk Sac/cytology , Allantois/growth & development , Allantois/metabolism , Animals , Blood Cells/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Endoderm/growth & development , Endoderm/metabolism , Erythroblasts/metabolism , Female , Fetal Proteins/genetics , Fetal Proteins/metabolism , Gene Expression Regulation, Developmental , Mice , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Patched-1 Receptor/genetics , Patched-1 Receptor/metabolism , Placenta/metabolism , Pregnancy , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Yolk Sac/growth & development , Yolk Sac/metabolism
4.
Wiley Interdiscip Rev Dev Biol ; 9(2): e362, 2020 03.
Article in English | MEDLINE | ID: mdl-31622045

ABSTRACT

In Placentalia, the fetus depends upon an organized vascular connection with its mother for survival and development. Yet, this connection was, until recently, obscure. Here, we summarize how two unrelated tissues, the primitive streak, or body axis, and extraembryonic visceral endoderm collaborate to create and organize the fetal-placental arterial connection in the mouse gastrula. The primitive streak reaches into the extraembryonic space, where it marks the site of arterial union and creates a progenitor cell pool. Through contact with the streak, associated visceral endoderm undergoes an epithelial-to-mesenchymal transition, contributing extraembryonic mesoderm to the placental arterial vasculature, and to the allantois, or pre-umbilical tissue. In addition, visceral endoderm bifurcates into the allantois where, with the primitive streak, it organizes the nascent umbilical artery and promotes allantoic elongation to the chorion, the site of fetal-maternal exchange. Brachyury mediates streak extension and vascular patterning, while Hedgehog is involved in visceral endoderm's conversion to mesoderm. A unique CASPASE-3-positive cell separates streak- and non-streak-associated domains in visceral endoderm. Based on these new insights at the posterior embryonic-extraembryonic interface, we conclude by asking whether so-called primordial germ cells are truly antecedents to the germ line that segregate within the allantois, or whether they are placental progenitor cells. Incorporating these new working hypotheses into mutational analyses in which the placentae are affected will aid understanding a spectrum of disorders, including orphan diseases, which often include abnormalities of the umbilical cord, yolk sac, and hindgut, whose developmental relationship to each other has, until now, been poorly understood. This article is categorized under: Birth Defects > Associated with Preimplantation and Gastrulation Early Embryonic Development > Gastrulation and Neurulation.


Subject(s)
Arteries/embryology , Embryo, Mammalian/cytology , Embryo, Mammalian/physiology , Endoderm/embryology , Fetus/embryology , Placenta/cytology , Primitive Streak/embryology , Animals , Female , Gastrula/cytology , Gastrula/physiology , Humans , Mice , Pregnancy
5.
Reprod Biomed Online ; 36(1): 6-11, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29079195

ABSTRACT

Primordial germ cells (PGCs), the precursors of the gametes, are now claimed to segregate within the extra-embryonic tissues of three species of placental mammals. In this brief Commentary, I raise the question of whether the so-called PGCs are not PGCs at all, but rather, progenitor cells that build the fetal-placental interface in Placentalia.


Subject(s)
Germ Cells/physiology , Placenta/embryology , Animals , Female , Gene Expression , Pregnancy
6.
Dev Biol ; 432(1): 98-124, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28882402

ABSTRACT

Hypoblast/visceral endoderm assists in amniote nutrition, axial positioning and formation of the gut. Here, we provide evidence, currently limited to humans and non-human primates, that hypoblast is a purveyor of extraembryonic mesoderm in the mouse gastrula. Fate mapping a unique segment of axial extraembryonic visceral endoderm associated with the allantoic component of the primitive streak, and referred to as the "AX", revealed that visceral endoderm supplies the placentae with extraembryonic mesoderm. Exfoliation of the AX was dependent upon contact with the primitive streak, which modulated Hedgehog signaling. Resolution of the AX's epithelial-to-mesenchymal transition (EMT) by Hedgehog shaped the allantois into its characteristic projectile and individualized placental arterial vessels. A unique border cell separated the delaminating AX from the yolk sac blood islands which, situated beyond the limit of the streak, were not formed by an EMT. Over time, the AX became the hindgut lip, which contributed extensively to the posterior interface, including both embryonic and extraembryonic tissues. The AX, in turn, imparted antero-posterior (A-P) polarity on the primitive streak and promoted its elongation and differentiation into definitive endoderm. Results of heterotopic grafting supported mutually interactive functions of the AX and primitive streak, showing that together, they self-organized into a complete version of the fetal-placental interface, forming an elongated structure that exhibited A-P polarity and was composed of the allantois, an AX-derived rod-like axial extension reminiscent of the embryonic notochord, the placental arterial vasculature and visceral endoderm/hindgut.


Subject(s)
Gastrula/embryology , Placenta/embryology , Primitive Streak/cytology , Primitive Streak/embryology , Animals , Cell Differentiation/physiology , Developmental Biology/methods , Endoderm/embryology , Epithelial-Mesenchymal Transition , Female , Gastrula/metabolism , Hedgehog Proteins/metabolism , Mesoderm/embryology , Mice , Notochord/embryology , Placenta/metabolism , Pregnancy , Signal Transduction
7.
Nat Protoc ; 12(5): 1029-1054, 2017 05.
Article in English | MEDLINE | ID: mdl-28426026

ABSTRACT

Here we describe a protocol to generate expandable and multipotent induced cardiac progenitor cells (iCPCs) from mouse adult fibroblasts using forced expression of Mesp1, Tbx5, Gata4, Nkx2.5 and Baf60c (MTGNB) along with activation of Wnt and JAK/STAT signaling. This method does not use iPS cell factors and thus differs from cell activation and signaling-directed (CASD) reprogramming to cardiac progenitors. Our method is specific to direct CPC reprogramming, whereas CASD reprogramming can generate various cell types depending on culture conditions and raises the possibility of transitioning through a pluripotent cell state. The protocol describes how to isolate and infect primary fibroblasts; induce reprogramming and observe iCPC colonies; expand and characterize reprogrammed iCPCs by immunostaining, flow cytometry and gene expression; differentiate iCPCs in vitro into cardiac-lineage cells; and test the embryonic potency of iCPCs via injection into the cardiac crescent of mouse embryos. A scientist experienced in molecular cell biology and embryology can reproduce this protocol in 12-16 weeks. iCPCs can be used for studying cardiac biology, drug discovery and regenerative medicine.


Subject(s)
Cell Differentiation , Cytological Techniques/methods , Fibroblasts/physiology , Stem Cells/physiology , Animals , Cell Proliferation , Flow Cytometry , Gene Expression , Gene Expression Profiling , Immunohistochemistry , Mice , Signal Transduction
8.
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
9.
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
10.
Dev Dyn ; 246(1): 50-71, 2017 01.
Article in English | MEDLINE | ID: mdl-27696611

ABSTRACT

BACKGROUND: PRDM1 is a transcriptional repressor that contributes to primordial germ cell (PGC) development. During early gastrulation, epiblast-derived PRDM1 is thought to be restricted to a lineage-segregated germ line in the allantois. However, given recent findings that PGCs overlap an allantoic progenitor pool that contributes widely to the fetal-umbilical interface, posterior PRDM1 may also contribute to soma. RESULTS: Within the posterior mouse gastrula (early streak, 12-s stages, embryonic days ∼6.75-9.0), PRDM1 localized to all tissues containing putative PGCs; however, PRDM1 was also found in all three primary germ layers, their derivatives, and two presumptive growth centers, the allantoic core domain and ventral ectodermal ridge. While PRDM1 and STELLA colocalized predominantly within the hindgut, where putative PGCs reside, other colocalizing cells were found in non-PGC sites. Additional PRDM1 and STELLA cells were found independent of each other throughout the posterior region, including the hindgut. The Prdm1-Cre-driven reporter supported PRDM1 localization in the majority of sites; however, some Prdm1 descendants were found in sites independent of PRDM1 protein, including allantoic mesothelium and hindgut endoderm. CONCLUSIONS: Posterior PRDM1 contributes more broadly to the developing fetal-maternal connection than previously recognized, and PRDM1 and STELLA, while overlapping in putative PGCs, also co-localize in several other tissues. Developmental Dynamics 246:50-71, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Gastrula/metabolism , Positive Regulatory Domain I-Binding Factor 1/analysis , Allantois/chemistry , Animals , Chromosomal Proteins, Non-Histone , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryonic Germ Cells , Endoderm/chemistry , Endoderm/embryology , Female , Fetus/metabolism , Gastrula/cytology , Mice , Placenta/metabolism , Pregnancy , Repressor Proteins/analysis
11.
Stem Cells ; 34(12): 2875-2888, 2016 12.
Article in English | MEDLINE | ID: mdl-27570947

ABSTRACT

While much progress has been made in the resolution of the cellular hierarchy underlying cardiogenesis, our understanding of chamber-specific myocardium differentiation remains incomplete. To better understand ventricular myocardium differentiation, we targeted the ventricle-specific gene, Irx4, in mouse embryonic stem cells to generate a reporter cell line. Using an antibiotic-selection approach, we purified Irx4+ cells in vitro from differentiating embryoid bodies. The isolated Irx4+ cells proved to be highly proliferative and presented Cxcr4, Pdgfr-alpha, Flk1, and Flt1 on the cell surface. Single Irx4+ ventricular progenitor cells (VPCs) exhibited cardiovascular potency, generating endothelial cells, smooth muscle cells, and ventricular myocytes in vitro. The ventricular specificity of the Irx4+ population was further demonstrated in vivo as VPCs injected into the cardiac crescent subsequently produced Mlc2v+ myocytes that exclusively contributed to the nascent ventricle at E9.5. These findings support the existence of a newly identified ventricular myocardial progenitor. This is the first report of a multipotent cardiac progenitor that contributes progeny specific to the ventricular myocardium. Stem Cells 2016;34:2875-2888.


Subject(s)
Heart Ventricles/cytology , Homeodomain Proteins/metabolism , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Animals , Biomarkers/metabolism , Cell Line , Cell Membrane/metabolism , Cell Proliferation , Cell Separation , Clone Cells , Embryonic Development , Endothelial Cells/metabolism , Gene Expression Regulation, Developmental , Genes, Reporter , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Smooth Muscle/cytology , Organ Specificity , Single-Cell Analysis , Time Factors
12.
Cell Stem Cell ; 18(3): 354-67, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26877223

ABSTRACT

Several studies have reported reprogramming of fibroblasts into induced cardiomyocytes; however, reprogramming into proliferative induced cardiac progenitor cells (iCPCs) remains to be accomplished. Here we report that a combination of 11 or 5 cardiac factors along with canonical Wnt and JAK/STAT signaling reprogrammed adult mouse cardiac, lung, and tail tip fibroblasts into iCPCs. The iCPCs were cardiac mesoderm-restricted progenitors that could be expanded extensively while maintaining multipotency to differentiate into cardiomyocytes, smooth muscle cells, and endothelial cells in vitro. Moreover, iCPCs injected into the cardiac crescent of mouse embryos differentiated into cardiomyocytes. iCPCs transplanted into the post-myocardial infarction mouse heart improved survival and differentiated into cardiomyocytes, smooth muscle cells, and endothelial cells. Lineage reprogramming of adult somatic cells into iCPCs provides a scalable cell source for drug discovery, disease modeling, and cardiac regenerative therapy.


Subject(s)
Cell Proliferation , Cellular Reprogramming Techniques/methods , Cellular Reprogramming , Fibroblasts/metabolism , Myoblasts, Cardiac/metabolism , Transcription Factors/biosynthesis , Animals , Cell Survival , Fibroblasts/cytology , Mice , Mice, Transgenic , Myoblasts, Cardiac/cytology , Transcription Factors/genetics
13.
Gene Expr Patterns ; 15(1): 8-20, 2014 May.
Article in English | MEDLINE | ID: mdl-24632399

ABSTRACT

Mixl1 is thought to play important roles in formation of mesoderm and endoderm. Previously, Mixl1 expression was reported in the posterior primitive streak and allantois, but the precise spatiotemporal whereabouts of Mixl1 protein throughout gastrulation have not been elucidated. To localize Mixl1 protein, immunohistochemistry was carried out at 2-4 h intervals on mouse gastrulae between primitive streak and 16-somite pair (s) stages (~E6.5-9.5). Mixl1 localized to the entire primitive streak early in gastrulation. However, by headfold stages (~E7.75-8.0), Mixl1 diminished within the mid-streak but remained concentrated at either end of the streak, and localized throughout midline posterior visceral endoderm. At the streak's anterior end, Mixl1 was confined to the posterior crown cells of Hensen's node, which contribute to dorsal hindgut endoderm, and the posterior notochord. In the posterior streak, Mixl1 localized to the Allantoic Core Domain (ACD), which is the source of most of the allantois and contributes to the posterior embryonic-extraembryonic interface. In addition, Mix1 co-localized with the early hematopoietic marker, Runx1, in the allantois and visceral yolk sac blood islands. During hindgut invagination (4-16s, ~E8.5-9.5), Mixl1 localized to the hindgut lip, becoming concentrated within the midline anastomosis of the splanchnopleure, which appears to create the ventral component of the hindgut and omphalomesenteric artery. Surrounding the distal hindgut, Mixl1 identified midline cells within tailbud mesoderm. Mixl1 was also found in the posterior notochord. These findings provide a critical systematic, and tissue-level understanding of embryonic Mixl1 localization, and support its role in regulation of crucial posterior axial mesendodermal stem cell niches during embryogenesis.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Gastrula/embryology , Homeodomain Proteins/metabolism , Animals , Embryo, Mammalian/metabolism , Endoderm/embryology , Gene Expression Regulation, Developmental , Mice , Primitive Streak/embryology , Somites/embryology
14.
Int Rev Cell Mol Biol ; 309: 1-57, 2014.
Article in English | MEDLINE | ID: mdl-24529721

ABSTRACT

Current dogma is that mouse primordial germ cells (PGCs) segregate within the allantois, or source of the umbilical cord, and translocate to the gonads, differentiating there into sperm and eggs. In light of emerging data on the posterior embryonic-extraembryonic interface, and the poorly studied but vital fetal-umbilical connection, we have reviewed the past century of experiments on mammalian PGCs and their relation to the allantois. We demonstrate that, despite best efforts and valuable data on the pluripotent state, what is and is not a PGC in vivo is obscure. Furthermore, sufficient experimental evidence has yet to be provided either for an extragonadal origin of mammalian PGCs or for their segregation within the posterior region. Rather, most evidence points to an alternative hypothesis that PGCs in the mouse allantois are part of a stem/progenitor cell pool that exhibits all known PGC "markers" and that builds/reinforces the fetal-umbilical interface, common to amniotes. We conclude by suggesting experiments to distinguish the mammalian germ line from the soma.


Subject(s)
Germ Cells/cytology , Allantois/cytology , Animals , Embryo, Mammalian/cytology , Mammals/embryology , Mice , Models, Biological
15.
Dev Dyn ; 243(3): 381-92, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24123507

ABSTRACT

BACKGROUND: The ventricular myocardium is the most prominent layer of the heart, and the most important for mediating cardiac physiology. Although the ventricular myocardium is critical for heart function, the cellular hierarchy responsible for ventricle-specific myocardium development remains unresolved. RESULTS: To determine the pattern and time course of ventricular myocardium development, we investigated IRX4 protein expression, which has not been previously reported. We identified IRX4+ cells in the cardiac crescent, and these cells were positive for markers of the first or second heart fields. From the onset of chamber formation, IRX4+ cells were restricted to the ventricular myocardium. This expression pattern persisted into adulthood. Of interest, we observed that IRX4 exhibits developmentally regulated dynamic intracellular localization. Throughout prenatal cardiogenesis, and up to postnatal day 4, IRX4 was detected in the cytoplasm of ventricular myocytes. However, between postnatal days 5­6, IRX4 translocated to the nucleus of ventricular myocytes. CONCLUSIONS: Given the ventricle-specific expression of Irx4 in later stages of heart development, we hypothesize that IRX4+ cells in the cardiac crescent represent the earliest cell population in the cellular hierarchy underlying ventricular myocardium development.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Heart Ventricles , Homeodomain Proteins/biosynthesis , Myocardium , Myocytes, Cardiac , Organogenesis/physiology , Animals , Heart Ventricles/cytology , Heart Ventricles/embryology , Mice , Mice, Inbred BALB C , Myocardium/cytology , Myocardium/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Organ Specificity
16.
Gene Expr Patterns ; 13(7): 225-39, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23639725

ABSTRACT

Interferon-induced transmembrane protein 3 (IFITM3; FRAGILIS; MIL-1) is part of a larger family of important small interferon-induced transmembrane genes and proteins involved in early development, cell adhesion, and cell proliferation, and which also play a major role in response to bacterial and viral infections and, more recently, in pronounced malignancies. IFITM3, together with tissue-nonspecific alkaline phosphatase (TNAP), PRDM1, and STELLA, has been claimed to be a hallmark of segregated primordial germ cells (PGCs) (Saitou et al., 2002). However, whether IFITM3, like STELLA, is part of a broader stem/progenitor pool that builds the posterior region of the mouse conceptus (Mikedis and Downs, 2012) is obscure. To discover the whereabouts of IFITM3 during mouse gastrulation (~E6.5-9.0), systematic immunohistochemical analysis was carried out at closely spaced 2-4-h intervals. Results revealed diverse, yet consistent, profiles of IFITM3 localization throughout the gastrula. Within the putative PGC trajectory and surrounding posterior tissues, IFITM3 localized as a large cytoplasmic spot with or without staining in the plasma membrane. IFITM3, like STELLA, was also found in the ventral ectodermal ridge (VER), a posterior progenitor pool that builds the tailbud. The large cytoplasmic spot with plasma membrane staining was exclusive to the posterior region; the visceral yolk sac, non-posterior tissues, and epithelial tissues exhibited spots of IFITM3 without cell surface staining. Colocalization of the intracellular IFITM3 spot with the endoplasmic reticulum, Golgi apparatus, or endolysosomes was not observed. That relatively high levels of IFITM3 were found throughout the posterior primitive streak and its derivatives is consistent with evidence that IFITM3, like STELLA, is part of a larger stem/progenitor cell pool at the posterior end of the primitive streak that forms the base of the allantois and builds the fetal-umbilical connection, thus further obfuscating practical phenotypic distinctions between so-called PGCs and surrounding soma.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Embryo, Mammalian/metabolism , Gastrula/metabolism , Gene Expression Regulation, Developmental , Membrane Proteins/metabolism , Animals , Core Binding Factor Alpha 2 Subunit/metabolism , Core Binding Factor Alpha 2 Subunit/physiology , Cytoplasm/metabolism , Embryo, Mammalian/cytology , Gastrula/cytology , Interferons/genetics , Interferons/metabolism , Membrane Proteins/genetics , Mice , Organelles/genetics , Organelles/metabolism , Primitive Streak , Trophoblasts/metabolism
17.
Development ; 139(6): 1059-69, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22354839

ABSTRACT

When amniotes appeared during evolution, embryos freed themselves from intracellular nutrition; development slowed, the mid-blastula transition was lost and maternal components became less important for polarity. Extra-embryonic tissues emerged to provide nutrition and other innovations. One such tissue, the hypoblast (visceral endoderm in mouse), acquired a role in fixing the body plan: it controls epiblast cell movements leading to primitive streak formation, generating bilateral symmetry. It also transiently induces expression of pre-neural markers in the epiblast, which also contributes to delay streak formation. After gastrulation, the hypoblast might protect prospective forebrain cells from caudalizing signals. These functions separate mesendodermal and neuroectodermal domains by protecting cells against being caught up in the movements of gastrulation.


Subject(s)
Biological Evolution , Body Patterning/genetics , Gastrulation , Germ Layers/physiology , Amnion/metabolism , Amnion/physiology , Animals , Chick Embryo , Embryonic Induction/genetics , Endoderm/embryology , Gene Expression Regulation, Developmental , Mice , Nervous System/embryology , Nutritional Physiological Phenomena , Primitive Streak/physiology
18.
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
19.
Dev Dyn ; 240(9): 2175-93, 2011 Sep.
Article in English | MEDLINE | ID: mdl-22016185

ABSTRACT

Although many fetal birth defects, particularly those of the body wall and gut, are associated with abnormalities of the umbilical cord, the developmental relationship between these structures is largely obscure. Recently, genetic analysis of mid-gestation mouse embryos revealed that defects in Hedgehog signaling led to omphalocoele, or failure of the body wall to close at the umbilical ring (Matsumaru et al. [ 2011] PLos One 6:e16260). However, systematic spatiotemporal localization of Hedgehog signaling in the allantois, or umbilical precursor tissue, and the surrounding regions has not been documented. Here, a combination of reagents, including the Ptc1:lacZ and Runx1:lacZ reporter mice, immunohistochemistry for Smoothened (Smo), Sonic Hedgehog (Shh), and Indian hedgehog (Ihh), and detailed PECAM-1/Flk-1/Runx-1 analysis, revealed robust Hedgehog signaling in previously undocumented posterior sites over an extended period of time (∼7.0-9.75 dpc). These included the recently described proximal walls of the allantois (Ventral and Dorsal Cuboidal Mesothelia; VCM and DCM, respectively); the ventral embryonic surface continuous with them; hemogenic arterial endothelia; hematopoietic cells; the hindgut; ventral ectodermal ridge (VER); chorionic ectoderm; and the intraplacental yolk sac (IPY), which appeared to be a site of placental hematopoiesis. This map of Hedgehog signaling in the posterior region of the mouse conceptus will provide a valuable foundation upon which to elucidate the origin of many posterior midline abnormalities, especially those of the umbilical cord and associated fetal defects. Developmental Dynamics 240:2175-2193, 2011. © 2011 Wiley-Liss, Inc.


Subject(s)
Embryo, Mammalian/metabolism , Fetus/physiology , Gastrula/metabolism , Hedgehog Proteins/metabolism , Umbilical Cord/physiology , Animals , Female , Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Immunohistochemistry , Male , Mice , Mice, Mutant Strains , Morphogenesis/genetics , Morphogenesis/physiology , Signal Transduction
20.
EMBO Rep ; 12(10): 987-90, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21941298

ABSTRACT

The EMBO Workshop on 'Lineage Commitments: Emphasis on Embryonic-Extraembryonic Interfaces', held in May 2011, demonstrated that embryonic and extraembryonic tissues play early and significant interacting roles that mutually promote each other's further and correct deployment within the mammalian conceptus. Highlighted here are those presentations that directly addressed embryonic-extraembryonic interfaces in building the mammalian fetus.


Subject(s)
Cell Lineage , Embryonic Induction , Amnion/embryology , Animals , Biotechnology , Chorion/embryology , Gene Expression Regulation, Developmental , Germ Layers/metabolism , Mice , Trophoblasts/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...