Anat Cell Biol.  2014 Mar;47(1):18-27. 10.5115/acb.2014.47.1.18.

Expression and localization of Rdd proteins in Xenopus embryo

Affiliations
  • 1Graduate School of Science and Technology, Niigata University, Niigata, Japan. maenobio@bio.sc.niigata-u.ac.jp

Abstract

The previous study has shown that repeated D domain-like (Rdd) proteins, a group of novel secretory proteins consisting of repeated domains of a cysteine-rich sequence, are involved in the process of blood vessel formation in Xenopus embryo. We performed further experiments to examine the localization of Rdd proteins in embryogenesis. Detection of tagged Rdd proteins expressed in blastomeres showed that Rdd proteins formed a high molecular weight complex and existed in the extracellular space. A rabbit antibody against the Rdd synthetic peptide identified a single band of 28 kD in embryonic tissue extract. By whole-mount immunostaining analysis, signal was detected in the regions of inter-somites, vitelline veins, and branchial arches at the tailbud stage. Staining of Rdd was remarkably reduced in the embryos injected with vascular endothelial growth factor Morpholino. We suggest that Rdd proteins interact with a molecule(s) associated with vascular precursor cells.

Keyword

Vascular cells; Frog; Vascular endothelial growth factor; Antibody; Cysteine-rich

MeSH Terms

Blastomeres
Blood Vessels
Branchial Region
Embryonic Development
Embryonic Structures*
Extracellular Space
Female
Molecular Weight
Pregnancy
Vascular Endothelial Growth Factor A
Veins
Vitellins
Xenopus*
Vascular Endothelial Growth Factor A
Vitellins

Figure

  • Fig. 1 Biochemical properties of Rdd proteins. (A) rdd3-ha mRNA was injected into 2-cell-stage embryos. The embryos were cultured until the late neurula stage. Western blot analysis was performed with extracted proteins after treatment with N-glycosidase F (lane 2) or no treatment (lane 1). The signal of Rdd3-HA was shifted by N-glycosidase F treatment, indicating that recombinant Rdd3 protein was N-glycosylated. (B) Western blot analysis was performed with extracted proteins from uninjected or rdd4-flag mRNA-injected embryos under the reducing (lanes 1 and 2) or non-reducing (lanes 3 and 4) condition. Rdd4-Flag appeared as a single band under the reducing condition (lane 2), but it showed a smear at the high molecular weight range under the non-reducing condition (lane 4).

  • Fig. 2 Localization of recombinant Rdd3 protein in the intercellular region. val-flag mRNA (Flag-tagged Val protein) (A-C) or rdd3-flag mRNA (Flag-tagged Rdd3 protein) (D-I) together with mif-myc mRNA (coding for Myc-tagged Mif protein) were injected into a single blastomere at the 16-cell stage. Animal cap explants were isolated and cultured until the early tailbud stage (st. 20). Explants were stained with anti-Myc and anti-Flag antibodies simultaneously and the reaction was visualized with Alexa488-based fluorescence (A, D, H) and Cy3-based fluorescence (B, E, G). Positive signal for Mif protein shows descendant cells of the RNA-injected single cell at the 16-cell stage (A, D, G). The border between the injected and uninjected cells is indicated by a white dotted line. Merged images of the double staining are also shown (C, F, I). Val-Flag protein was detected in the nuclei of cells that had been injected with mif-myc mRNA (C, C'). On the other hand, Rdd3-Flag protein was stained in the cytoplasm of Mif-positive cells and also stained in the intercellular region of Mif-negative cells (F, F'). Exchange of the second antibodies between Cy3-conjugated IgG and Alexa488-conjugated IgG gave the same results (I, I'). Scale bars in panels I and I'=50 µm (A-I), 50 µm (C', F', I').

  • Fig. 3 Production of anti-sera against a synthetic peptide from the Rdd3 amino acid sequence. (A) Western blot analysis was performed with anti-HA antibody or anti-Rdd antibody to detect HA-tagged recombinant Rdd proteins. Anti-HA antibody reacted with Rdd2, Rdd3, and Rdd4 proteins (lanes 1-3), while anti-Rdd antibody reacted with Rdd3 and Rdd4 (lanes 5 and 6). (B) Absorption of anti-serum with Rdd (lane2) or control (lane 3) peptide indicated specific reaction of the antibody with Rdd4 protein in Western blot analysis.

  • Fig. 4 Detection of endogenous Rdd proteins in the embryo. (A) Western blot analysis was performed using anti-Rdd antibody to detect the Rdd protein in extracts from pooled embryos at various stages. A single band at 28 kDa was detected at the gastrula and tailbud stages. The signal for detecting glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is shown as a loading control. (B) Whole-mount immunostaining analysis showed the localization of endogenous Rdd protein in intact (a-d) or Rdd3/4 Morpholino oligo (MO) (9.2 pmol/embryo)-injected (e-h) albino embryos at the tailbud stage (st. 32). Staining was found in the vitelline vein (b, arrowheads), intersomitic region (c, white arrowheads), hypaxial muscle segment (c, arrowheads) and branchial arches (d, arrowheads). The intensity of staining was reduced in the rdd3/4 MO-injected embryo (e-h). (C) Another experiment shows the staining of Rdd protein in control MO (a)- or Rdd3/4 MO (b)-injected wild-type embryos at the tailbud stage (st. 32). (D) Histological views of the lateral (a-c), ventral (d-f ), and pharyngeal (g-i) parts of albino embryos are shown. Sections were stained by hematoxylin and eosin (H&E) (a, d, g) or immunostained using anti-Rdd antibody in the absence (b, e, h) or presence (c, f, i) of Rdd peptide. a, b, c and d, e, f are distinct but neighboring cross sections. g, h, i are horizontal sections. Endogenous Rdd protein was detected in the intermediate mesoderm (b, arrowheads), in the ventral mesoderm (e, arrowheads), and in the branchial arches (h, arrowheads). en, endoderm; pt, pronephric tube. Scale bars=0.5 mm (B, C), 50 µm (D).

  • Fig. 5 Concomitant localization of Rdd3 protein with blood vessel formation. Whole-mount in situ hybridization (A) and whole-mount immunostaining (B) analyses were performed to show tie-2 expression and Rdd protein localization in tailbud (st. 32) embryos. Embryos were injected with vascular endothelial growth factor Morpholino oligo (VEGF MO) (9.2 pmol/embryo) (d-f ) or H2O (a-c) in the marginal zone at the 4-cell stage. (A) In the control embryo, tie-2 was expressed in the vitelline vein (b, arrowheads), intermediate mesoderm (c, white arrowheads) and branchial arches (c, arrowheads). Localization of tie-2 expression was lost in the VEGF MO-injected embryo (d-f ). (B) Rdd protein was detected in the vitelline vein (b, arrowheads), intersomite region (c, white arrowheads), and branchial arches (c, arrowheads). Localization of Rdd protein was lost in the VEGF MO-injected embryo (d-f ). Scale bars=0.5 mm (A, B).


Reference

1. Maéno M, Todate A, Katagiri C. The localization of precursor cells for larval and adult hemopoietic cells of Xenopus laevis in two regions of embryos. Dev Growth Differ. 1985; 27:137–148.
2. Maéno M, Tochinai S, Katagiri C. Differential participation of ventral and dorsolateral mesoderms in the hemopoiesis of Xenopus, as revealed in diploid-triploid or interspecific chimeras. Dev Biol. 1985; 110:503–508.
3. Dale L, Howes G, Price BM, Smith JC. Bone morphogenetic protein 4: a ventralizing factor in early Xenopus development. Development. 1992; 115:573–585.
4. Jones CM, Lyons KM, Lapan PM, Wright CV, Hogan BL. DVR-4 (bone morphogenetic protein-4) as a posterior-ventralizing factor in Xenopus mesoderm induction. Development. 1992; 115:639–647.
5. Maeno M, Ong RC, Suzuki A, Ueno N, Kung HF. A truncated bone morphogenetic protein 4 receptor alters the fate of ventral mesoderm to dorsal mesoderm: roles of animal pole tissue in the development of ventral mesoderm. Proc Natl Acad Sci U S A. 1994; 91:10260–10264.
6. Suzuki A, Thies RS, Yamaji N, Song JJ, Wozney JM, Murakami K, Ueno N. A truncated bone morphogenetic protein receptor affects dorsal-ventral patterning in the early Xenopus embryo. Proc Natl Acad Sci U S A. 1994; 91:10255–10259.
7. Tran HT, Sekkali B, Van Imschoot G, Janssens S, Vleminckx K. Wnt/beta-catenin signaling is involved in the induction and maintenance of primitive hematopoiesis in the vertebrate embryo. Proc Natl Acad Sci U S A. 2010; 107:16160–16165.
8. Mead PE, Kelley CM, Hahn PS, Piedad O, Zon LI. SCL specifies hematopoietic mesoderm in Xenopus embryos. Development. 1998; 125:2611–2620.
9. Mead PE, Deconinck AE, Huber TL, Orkin SH, Zon LI. Primitive erythropoiesis in the Xenopus embryo: the synergistic role of LMO-2, SCL and GATA-binding proteins. Development. 2001; 128:2301–2308.
10. Walmsley M, Ciau-Uitz A, Patient R. Adult and embryonic blood and endothelium derive from distinct precursor populations which are differentially programmed by BMP in Xenopus. Development. 2002; 129:5683–5695.
11. Kelley C, Yee K, Harland R, Zon LI. Ventral expression of GATA-1 and GATA-2 in the Xenopus embryo defines induction of hematopoietic mesoderm. Dev Biol. 1994; 165:193–205.
12. Maeno M, Mead PE, Kelley C, Xu RH, Kung HF, Suzuki A, Ueno N, Zon LI. The role of BMP-4 and GATA-2 in the induction and differentiation of hematopoietic mesoderm in Xenopus laevis. Blood. 1996; 88:1965–1972.
13. Dalgin G, Goldman DC, Donley N, Ahmed R, Eide CA, Christian JL. GATA-2 functions downstream of BMPs and CaM KIV in ectodermal cells during primitive hematopoiesis. Dev Biol. 2007; 310:454–469.
14. Chen Y, Costa RM, Love NR, Soto X, Roth M, Paredes R, Amaya E. C/EBPalpha initiates primitive myelopoiesis in pluripotent embryonic cells. Blood. 2009; 114:40–48.
15. Costa RM, Soto X, Chen Y, Zorn AM, Amaya E. spib is required for primitive myeloid development in Xenopus. Blood. 2008; 112:2287–2296.
16. Liu F, Walmsley M, Rodaway A, Patient R. Fli1 acts at the top of the transcriptional network driving blood and endothelial development. Curr Biol. 2008; 18:1234–1240.
17. Salanga MC, Meadows SM, Myers CT, Krieg PA. ETS family protein ETV2 is required for initiation of the endothelial lineage but not the hematopoietic lineage in the Xenopus embryo. Dev Dyn. 2010; 239:1178–1187.
18. Meadows SM, Salanga MC, Krieg PA. Kruppel-like factor 2 cooperates with the ETS family protein ERG to activate Flk1 expression during vascular development. Development. 2009; 136:1115–1125.
19. Ciau-Uitz A, Pinheiro P, Gupta R, Enver T, Patient R. Tel1/ETV6 specifies blood stem cells through the agency of VEGF signaling. Dev Cell. 2010; 18:569–578.
20. Carmeliet P, Ferreira V, Breier G, Pollefeyt S, Kieckens L, Gertsenstein M, Fahrig M, Vandenhoeck A, Harpal K, Eberhardt C, Declercq C, Pawling J, Moons L, Collen D, Risau W, Nagy A. Abnormal blood vessel development and lethality in embryos lacking a single VEGF allele. Nature. 1996; 380:435–439.
21. Ferrara N, Carver-Moore K, Chen H, Dowd M, Lu L, O'Shea KS, Powell-Braxton L, Hillan KJ, Moore MW. Heterozygous embryonic lethality induced by targeted inactivation of the VEGF gene. Nature. 1996; 380:439–442.
22. Koibuchi N, Taniyama Y, Nagao K, Ogihara T, Kaneda Y, Morishita R. The effect of VEGF on blood vessels and blood cells during Xenopus development. Biochem Biophys Res Commun. 2006; 344:339–345.
23. Nagao K, Taniyama Y, Koibuchi N, Morishita R. Constitutive over-expression of VEGF results in reduced expression of Hand-1 during cardiac development in Xenopus. Biochem Biophys Res Commun. 2007; 359:431–437.
24. Cleaver O, Tonissen KF, Saha MS, Krieg PA. Neovascularization of the Xenopus embryo. Dev Dyn. 1997; 210:66–77.
25. Cleaver O, Krieg PA. VEGF mediates angioblast migration during development of the dorsal aorta in Xenopus. Development. 1998; 125:3905–3914.
26. Iraha F, Saito Y, Yoshida K, Kawakami M, Izutsu Y, Daar IO, Maéno M. Common and distinct signals specify the distribution of blood and vascular cell lineages in Xenopus laevis embryos. Dev Growth Differ. 2002; 44:395–407.
27. Walmsley M, Cleaver D, Patient R. Fibroblast growth factor controls the timing of Scl, Lmo2, and Runx1 expression during embryonic blood development. Blood. 2008; 111:1157–1166.
28. Maéno M, Komiyama K, Matsuzaki Y, Hosoya J, Kurihara S, Sakata H, Izutsu Y. Distinct mechanisms control the timing of differentiation of two myeloid populations in Xenopus ventral blood islands. Dev Growth Differ. 2012; 54:187–201.
29. Kazanskaya O, Ohkawara B, Heroult M, Wu W, Maltry N, Augustin HG, Niehrs C. The Wnt signaling regulator R-spondin 3 promotes angioblast and vascular development. Development. 2008; 135:3655–3664.
30. Shibata T, Takahashi Y, Tasaki J, Saito Y, Izutsu Y, Maéno M. A role of D domain-related proteins in differentiation and migration of embryonic cells in Xenopus laevis. Mech Dev. 2008; 125:284–298.
31. Saito Y, Takahashi Y, Izutsu Y, Maéno M. Identification and expression of ventrally associated leucine-zipper (VAL) in Xenopus embryo. Int J Dev Biol. 2010; 54:203–208.
32. Niuewkoop PD, Faber J. Normal table of Xenopus laevis (Daudin). Amsterdam: Elsevier;1994.
33. Suzuki M, Takamura Y, Maéno M, Tochinai S, Iyaguchi D, Tanaka I, Nishihira J, Ishibashi T. Xenopus laevis macrophage migration inhibitory factor is essential for axis formation and neural development. J Biol Chem. 2004; 279:21406–21414.
34. Shain DH, Zuber MX. Sodium dodecyl sulfate (SDS)-based whole-mount in situ hybridization of Xenopus laevis embryos. J Biochem Biophys Methods. 1996; 31:185–188.
35. Martin BL, Harland RM. Hypaxial muscle migration during primary myogenesis in Xenopus laevis. Dev Biol. 2001; 239:270–280.
36. Verweij CL, Diergaarde PJ, Hart M, Pannekoek H. Full-length von Willebrand factor (vWF) cDNA encodes a highly repetitive protein considerably larger than the mature vWF subunit. EMBO J. 1986; 5:1839–1847.
37. Hardy DM, Garbers DL. A sperm membrane protein that binds in a species-specific manner to the egg extracellular matrix is homologous to von Willebrand factor. J Biol Chem. 1995; 270:26025–26028.
38. Gao Z, Garbers DL. Species diversity in the structure of zonadhesin, a sperm-specific membrane protein containing multiple cell adhesion molecule-like domains. J Biol Chem. 1998; 273:3415–3421.
Full Text Links
  • ACB
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr