Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Tmem88 confines ectodermal Wnt2bb signaling in pharyngeal arch artery progenitors for balancing cell cycle progression and cell fate decision

Abstract

Pharyngeal arch artery (PAA) progenitors undergo proliferative expansion and angioblast differentiation to build vessels connecting the heart with the dorsal aortae. However, it remains unclear whether and how these two processes are orchestrated. Here we demonstrate that Tmem88 is required to fine-tune PAA progenitor proliferation and differentiation. Loss of zebrafish tmem88a/b leads to an excessive expansion and a failure of differentiation of PAA progenitors. Moreover, tmem88a/b deficiency enhances cyclin D1 expression in PAA progenitors via aberrant Wnt signal activation. Mechanistically, cyclin D1-CDK4/6 promotes progenitor proliferation through accelerating the G1/S transition while suppressing angioblast differentiation by phosphorylating Nkx2.5/Smad3. Ectodermal Wnt2bb signaling is confined by Tmem88 in PAA progenitors to ensure a balance between proliferation and differentiation. Therefore, the proliferation and angioblast differentiation of PAA progenitors manifest an inverse relationship and are delicately regulated by cell cycle machinery downstream of the Tmem88-Wnt pathway.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: tmem88a and tmem88b are expressed in PAA progenitors.
Fig. 2: Genetic depletion of tmem88a and tmem88b impairs PAA morphogenesis.
Fig. 3: Loss of tmem88a/b promotes PAA progenitor proliferation but disrupts its angioblast differentiation.
Fig. 4: Tmem88a/b modulates PAA morphogenesis via suppression of Wnt/β-catenin signaling.
Fig. 5: Tmem88 restricts Wnt/β-catenin signaling to balance the proliferation and differentiation of PAA progenitors.
Fig. 6: CDK4 inhibits Nkx2.5 transcriptional activity by phosphorylation.
Fig. 7: CDK4-mediated phosphorylation of Smad3 inhibits PAA progenitor angioblast differentiation.
Fig. 8: Ectodermal Wnt2bb signaling is confined by Tmem88 to balance the proliferation and differentiation of PAA progenitors.

Similar content being viewed by others

Data availability

All data supporting the findings in this study are included in the main article and associated files.

References

  1. Kodo, K. & Yamagishi, H. A decade of advances in the molecular embryology and genetics underlying congenital heart defects. Circ. J. 75, 2296–2304 (2011).

    Article  CAS  PubMed  Google Scholar 

  2. Psillas, G. et al. Subclavian steal syndrome: neurotological manifestations. Acta Otorhinolaryngol. Ital. 27, 33–37 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Hoffman, J. I. & Kaplan, S. The incidence of congenital heart disease. J. Am. Coll. Cardiol. 39, 1890–1900 (2002).

    Article  PubMed  Google Scholar 

  4. Wang, X. et al. Endothelium in the pharyngeal arches 3, 4 and 6 is derived from the second heart field. Dev. Biol. 421, 108–117 (2017).

    Article  CAS  PubMed  Google Scholar 

  5. Paffett-Lugassy, N. et al. Heart field origin of great vessel precursors relies on nkx2.5-mediated vasculogenesis. Nat. Cell Biol. 15, 1362–1369 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Hardwick, L. J. A., Azzarelli, R. & Philpott, A. Cell cycle-dependent phosphorylation and regulation of cellular differentiation. Biochem. Soc. Trans. 46, 1083–1091 (2018).

    Article  CAS  PubMed  Google Scholar 

  7. Lauridsen, F. K. B. et al. Differences in cell cycle status underlie transcriptional heterogeneity in the HSC compartment. Cell Rep. 24, 766–780 (2018).

    Article  CAS  PubMed  Google Scholar 

  8. Lu, Y. C. et al. The molecular signature of megakaryocyte-erythroid progenitors reveals a role for the cell cycle in fate specification. Cell Rep. 25, 2083–2093 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Pauklin, S. & Vallier, L. The cell-cycle state of stem cells determines cell fate propensity. Cell 155, 135–147 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Dalton, S. Linking the cell cycle to cell fate decisions. Trends Cell Biol. 25, 592–600 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Mao, A. et al. Pharyngeal pouches provide a niche microenvironment for arch artery progenitor specification. Development 148, dev192658 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Mao, A. et al. PDGF signaling from pharyngeal pouches promotes arch artery morphogenesis. J. Genet. Genomics 46, 551–559 (2019).

    Article  PubMed  Google Scholar 

  13. Abrial, M. et al. TGF-β signaling is necessary and sufficient for pharyngeal arch artery angioblast formation. Cell Rep. 20, 973–983 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Meng, Z. Z. et al. The pro-inflammatory signalling regulator Stat4 promotes vasculogenesis of great vessels derived from endothelial precursors. Nat. Commun. 8, 14640 (2017).

  15. Gao, J. et al. Wnt/β-catenin signaling in neural stem cell homeostasis and neurological diseases. Neuroscientist 27, 58–72 (2021).

    Article  CAS  PubMed  Google Scholar 

  16. Pinto, D. & Clevers, H. Wnt control of stem cells and differentiation in the intestinal epithelium. Exp. Cell Res. 306, 357–363 (2005).

    Article  CAS  PubMed  Google Scholar 

  17. Logan, C. Y. & Nusse, R. The Wnt signaling pathway in development and disease. Annu. Rev. Cell Dev. Biol. 20, 781–810 (2004).

    Article  CAS  PubMed  Google Scholar 

  18. Angers, S. & Moon, R. T. Proximal events in Wnt signal transduction. Nat. Rev. Mol. Cell Biol. 10, 468–477 (2009).

    Article  CAS  PubMed  Google Scholar 

  19. Lee, H. J. et al. Identification of transmembrane protein 88 (TMEM88) as a Dishevelled-binding protein. J. Biol. Chem. 285, 41549–41556 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Lee, H. & Evans, T. TMEM88 inhibits Wnt signaling by promoting Wnt signalosome localization to multivesicular bodies. iScience 19, 267–280 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Palpant, N. J. et al. Transmembrane protein 88: a Wnt regulatory protein that specifies cardiomyocyte development. Development 140, 3799–3808 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Eve, A. M., Place, E. S. & Smith, J. C. Comparison of zebrafish tmem88a mutant and morpholino knockdown phenotypes. PLoS ONE 12, e0172227 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Cannon, J. E. et al. Global analysis of the haematopoietic and endothelial transcriptome during zebrafish development. Mech. Dev. 130, 122–131 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Gomez, G. et al. Identification of vascular and hematopoietic genes downstream of etsrp by deep sequencing in zebrafish. PLoS ONE 7, e31658 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Novikov, N. & Evans, T. Tmem88a mediates GATA-dependent specification of cardiomyocyte progenitors by restricting WNT signaling. Development 140, 3787–3798 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Sugiyama, M. et al. Illuminating cell-cycle progression in the developing zebrafish embryo. Proc. Natl Acad. Sci. USA 106, 20812–20817 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Dorsky, R. I., Sheldahl, L. C. & Moon, R. T. A transgenic Lef1/β-catenin-dependent reporter is expressed in spatially restricted domains throughout zebrafish development. Dev. Biol. 241, 229–237 (2002).

    Article  CAS  PubMed  Google Scholar 

  28. Ewan, K. et al. A useful approach to identify novel small-molecule inhibitors of Wnt-dependent transcription. Cancer Res. 70, 5963–5973 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Kuure, S. et al. Glycogen synthase kinase-3 inactivation and stabilization of β-catenin induce nephron differentiation in isolated mouse and rat kidney mesenchymes. J. Am. Soc. Nephrol. 18, 1130–1139 (2007).

    Article  CAS  PubMed  Google Scholar 

  30. Murray, A. W. & Kirschner, M. W. Cyclin synthesis drives the early embryonic cell cycle. Nature 339, 275–280 (1989).

    Article  CAS  PubMed  Google Scholar 

  31. Davidson, G. & Niehrs, C. Emerging links between CDK cell cycle regulators and Wnt signaling. Trends Cell Biol. 20, 453–460 (2010).

    Article  CAS  PubMed  Google Scholar 

  32. Fry, D. W. et al. Specific inhibition of cyclin-dependent kinase 4/6 by PD 0332991 and associated antitumor activity in human tumor xenografts. Mol. Cancer Ther. 3, 1427–1438 (2004).

    Article  CAS  PubMed  Google Scholar 

  33. Sherr, C. J. & Roberts, J. M. CDK inhibitors: positive and negative regulators of G1-phase progression. Genes Dev. 13, 1501–1512 (1999).

    Article  CAS  PubMed  Google Scholar 

  34. McElhinney, D. B. et al. NKX2.5 mutations in patients with congenital heart disease. J. Am. Coll. Cardiol. 42, 1650–1655 (2003).

    Article  CAS  PubMed  Google Scholar 

  35. Tang, X. et al. SIRT1 deacetylates the cardiac transcription factor Nkx2.5 and inhibits its transcriptional activity. Sci Rep. 6, 36576 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Matsushime, H. et al. Identification and properties of an atypical catalytic subunit (p34PSK-J3/cdk4) for mammalian D type G1 cyclins. Cell 71, 323–334 (1992).

    Article  CAS  PubMed  Google Scholar 

  37. Kitagawa, M. et al. The consensus motif for phosphorylation by cyclin D1-Cdk4 is different from that for phosphorylation by cyclin A/E-Cdk2. EMBO J. 15, 7060–7069 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Wei, S. et al. The guanine nucleotide exchange factor Net1 facilitates the specification of dorsal cell fates in zebrafish embryos by promoting maternal β-catenin activation. Cell Res. 27, 202–225 (2017).

    Article  CAS  PubMed  Google Scholar 

  39. Nagelberg, D. et al. Origin, specification, and plasticity of the great vessels of the heart. Curr. Biol. 25, 2099–2110 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Rossi, A. et al. Genetic compensation induced by deleterious mutations but not gene knockdowns. Nature 524, 230–233 (2015).

    Article  CAS  PubMed  Google Scholar 

  41. Matsuura, I. et al. Cyclin-dependent kinases regulate the antiproliferative function of Smads. Nature 430, 226–231 (2004).

    Article  CAS  PubMed  Google Scholar 

  42. Sumanas, S. & Lin, S. Ets1-related protein is a key regulator of vasculogenesis in zebrafish. PLoS Biol. 4, e10 (2006).

    Article  PubMed  Google Scholar 

  43. Drake, C. J. et al. TAL1/SCL is expressed in endothelial progenitor cells/angioblasts and defines a dorsal-to-ventral gradient of vasculogenesis. Dev. Biol. 192, 17–30 (1997).

    Article  CAS  PubMed  Google Scholar 

  44. Schupp, M. O. et al. Transcriptional inhibition of etv2 expression is essential for embryonic cardiac development. Dev. Biol. 393, 71–83 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Jin, H. et al. The 5′ zebrafish scl promoter targets transcription to the brain, spinal cord, and hematopoietic and endothelial progenitors. Dev. Dyn. 235, 60–67 (2006).

    Article  CAS  PubMed  Google Scholar 

  46. Ma, J. et al. MCP-1 mediates TGF-β-induced angiogenesis by stimulating vascular smooth muscle cell migration. Blood 109, 987–994 (2007).

    Article  CAS  PubMed  Google Scholar 

  47. Ober, E. A. et al. Mesodermal Wnt2b signalling positively regulates liver specification. Nature 442, 688–691 (2006).

    Article  CAS  PubMed  Google Scholar 

  48. Nakajima, K. et al. Coordinated regulation of differentiation and proliferation of embryonic cardiomyocytes by a jumonji (Jarid2)-cyclin D1 pathway. Development 138, 1771–1782 (2011).

    Article  CAS  PubMed  Google Scholar 

  49. Friedrichs, M. et al. BMP signaling balances proliferation and differentiation of muscle satellite cell descendants. BMC Cell Biol. 12, 26 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Singh, A. M. et al. Cell-cycle control of developmentally regulated transcription factors accounts for heterogeneity in human pluripotent cells. Stem Cell Rep. 1, 532–544 (2013).

    Article  CAS  Google Scholar 

  51. Azzarelli, R. et al. Multi-site Neurogenin3 phosphorylation controls pancreatic endocrine differentiation. Dev. Cell 41, 274–286. (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Lu, H. et al. EpCAM is an endoderm-specific Wnt derepressor that licenses hepatic development. Dev. Cell 24, 543–553 (2013).

    Article  CAS  PubMed  Google Scholar 

  53. Zorn, A. M. & Wells, J. M. Vertebrate endoderm development and organ formation. Annu. Rev. Cell Dev. Biol. 25, 221–251 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Kimmel, C. B. et al. Stages of embryonic development of the zebrafish. Dev. Dyn. 203, 253–310 (1995).

    Article  CAS  PubMed  Google Scholar 

  55. Reischauer, S. et al. Cloche is a bHLH-PAS transcription factor that drives haemato-vascular specification. Nature 535, 294–298 (2016).

    Article  CAS  PubMed  Google Scholar 

  56. Dickmeis, T. et al. A crucial component of the endoderm formation pathway, CASANOVA, is encoded by a novel sox-related gene. Genes Dev. 15, 1487–1492 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Ning, G. et al. MicroRNA-92a upholds Bmp signaling by targeting noggin3 during pharyngeal cartilage formation. Dev. Cell 24, 283–295 (2013).

    Article  CAS  PubMed  Google Scholar 

  58. Yan, Y. et al. The BMP ligand Pinhead together with Admp supports the robustness of embryonic patterning. Sci. Adv. 5, eaau6455 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We acknowledge the financial support of the National Key Research and Development Program of China (2018YFA0800200 (Q.W.) and 2020YFA0804000 (Q.W.)) and the National Natural Science Foundation of China (32025014 (Q.W.) and 81921006 (Q.W.)).

Author information

Authors and Affiliations

Authors

Contributions

Q.W. conceived the study, designed experiments and created the figures. M.M.Z., J.L., A.H.M., G.Z.N. and Y.C. performed experiments and collected data. M.M.Z., W.Q.Z. and Q.W. analyzed the data and discussed results and strategy. M.M.Z. and Q.W. wrote the manuscript, with input from all authors. Q.W. reviewed and edited the manuscript.

Corresponding author

Correspondence to Qiang Wang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cardiovascular Research thanks Todd Evans, Lluis Fajas and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Expression patterns of tmem88a and tmem88b during embryo development.

a,b, Expression of tmem88a and tmem88b in wild-type embryos at the indicated developmental stages was analyzed using whole-mount in situ hybridization. Arrowheads indicate pharynx expression of tmem88a (a) and tmem88b (b). Lateral views, anterior to the left.

Extended Data Fig. 2 Generation of tmem88a and tmem88b mutants using the CRISPR/Cas9 system.

a, The tmem88a mutant had a 17-base deletion and 1-base substitution that resulted in a truncated protein lacking the TM domain and PDZ-binding motif. Red arrowhead indicates the position of the gRNA target site. b, The tmem88b mutant had a 166-base deletion, which was predicted to encode a truncated protein lacking the TM domain and PDZ-binding motif. c, The tmem88a mutant was identified using HpyCH4III digestion. d, The tmem88b homozygous mutant was identified using PCR-based genotyping analyses. M indicates the DNA marker. e, The expression of tmem88a and tmem88b in wild-type and the relevant mutant embryos was examined using in situ hybridization. f, Morphological observation of wild-type, tmem88a/ and tmem88b/ embryos at the indicated stages. Scale bars, 600 μm. g,h, tmem88a/ and tmem88b/ embryos have no apparent defects on the formation of the heart and blood cells. Wild-type, tmem88a/ and tmem88b/ embryos were harvested at 48 hpf for in situ hybridization (g) or O-dianisidine staining (h). Ventral views, anterior to the top.

Extended Data Fig. 3 tmem88a/b−/− mutants exhibit evident pericardial edema and obvious defects in heart and blood cell development.

a-c, The expression of tmem88a and tmem88b in wild-type embryos and the indicated mutants was examined using in situ hybridization (a) and qRT-PCR (b and c). The expression levels of β-actin were used as a reference to normalize the amount of mRNAs in each sample. Data represent the mean ± SD of three independent experiments. Student’s t-test was used. NS, non-significant. d, The expression of Tmem88a and Tmem88b proteins in wild-type embryos and tmem88a/, tmem88b/ and tmem88a/b/ mutants at 28 hpf was examined using Western blot. e, Morphological defects in tmem88a/b/ embryos at the indicated stages. Scale bars, 600 μm. f, Wild-type and tmem88a/b/ embryos were harvested at 48 hpf for in situ hybridization with a cmlc2 probe. Ventral views, anterior to the top. g, O-dianisidine staining of erythrocytes in wild-type and tmem88a/b/ embryos at 48 hpf. Ventral views, anterior to the top. h, Wild-type and tmem88a/b/ embryos were harvested at 18 hpf and 20 hpf for in situ hybridization with a nkx2.5 probe. Dorsal views, anterior to the top.

Source data

Extended Data Fig. 4 Ectopic activation of Wnt/β-catenin signaling impairs the PAA progenitor to angioblast transition.

a,b, Tg(hsp70l:wnt8a-EGFP) embryos were heat shocked at 20 hpf for 30 min, and then harvested at 38 hpf for in situ hybridization with etv2 and scl probes (a). The average numbers of PAA angioblast clusters were quantified in (b). n ≥ 22 per group. Student’s t-test was used. Data are mean ± SD.

Source data

Extended Data Fig. 5 The expression of cyclin D1 is increased in tmem88a/b-depleted PAA progenitors.

a-c, qRT-PCR analysis of the expression of cyclin D1 and cdk4 in ZsYellow+ cells sorted from the pharynx region of Tg(nkx2.5:ZsYellow) and tmem88a/b-deficient Tg(nkx2.5:ZsYellow) embryos at 28 hpf (a), 48 hpf (b) and 60 hpf (c) using fluorescence activated cell sorting purification. The expression levels of β-actin were used as a reference to normalize the amount of mRNAs in each sample. Data represent the mean ± SD of three independent experiments. Student’s t-test was used. NS, non-significant. d, tmem88a/b/ embryos were treated with either 10 μM Ro-3306 or PHA-767491 from 20 hpf, and then harvested at 38 hpf for in situ hybridization with nkx2.5 and etv2 probes.

Source data

Extended Data Fig. 6 CDK4-mediated phosphorylation on threonine 11 of zNkx2.5 restrained its transcriptional activity.

a, HEK293T cells were co-transfected with Flag-zNkx2.5 and increasing amounts of Myc-CDK4, then harvested for Western blot. Tubulin was used as the loading control. b, HeLa cells transfected with the indicated plasmids were immunostained with anti-Flag and anti-Myc antibodies. The nuclei were counterstained with DAPI. Scale bar, 10 µm. c-e, HEK293T cells co-transfected with ANF-luciferase plasmid and the indicated plasmids were harvested 36 h after transfection for luciferase assays. Data represent the mean ± SD of three independent experiments. Student’s t-test was used. NS, no significant. WT, wild-type; KD, kinase dead. f,g, HEK293T cells were co-transfected with ANF-luciferase plasmid and a construct expressing either Flag-zNkx2.5 or its T11A mutant. Cells were then harvested for luciferase assays (f). Wild-type embryos were injected with 100 pg of the ANF-luciferase reporter constructs and 200 pg Flag-nkx2.5 mRNA or Flag-nkx2.5-T11A mRNA at the one-cell stage, and then these embryos were subjected to luciferase assays at 24 hpf (g). Data represent the mean ± SD of three independent experiments. Student’s t-test was used.

Source data

Extended Data Fig. 7 CDK4 interacts with and phosphorylates Smad3a and Smad3b.

a, Conserved CDK substrate motifs in Smad3 proteins from different species. Red stars indicate the critical residues in CDK substrate motifs. b,c, Both Smad3a and Smad3b interact with CDK4. HEK293T cells transfected with the indicated plasmids were subjected to immunoprecipitation with an anti-Flag antibody. d, CDK4 phosphorylates Smad3a and Smad3b. HEK293T cells were transfected with the indicated plasmids, and then treated with or without 0.5 μM PD0332991 for 5 h prior to harvest for Western blot. CDK4 substrates were blotted with an antibody against phosphorylated human Smad3 (Thr178). Tubulin was used as the loading control. WT, wild-type; KD, kinase dead.

Source data

Extended Data Fig. 8 CDK4-dependent phosphorylation of Smad3 suppresses etv2 transcription.

a, HEK293T cells co-transfected with increasing amounts of Smad3a expression plasmids and etv2-Luc or scl-Luc reporter, then harvested for luciferase assays. Data represent the mean ± SD of three independent experiments. Student’s t-test was used. b,c, CDK4-mediated phosphorylation of Smad3 represses etv2 transcription. Note that the treatment with CDK4/6 inhibitor PD0332991 (b) or mutation of the three CDK phosphorylation sites in Smad3a (Smad3a-3A) (c) relieved the CDK4-induced suppression of etv2-Luc reporter expression. Data represent the mean ± SD of three independent experiments. Student’s t-test was used. d, The −2516 to −2270 bp region of the etv2 promoter is important to respond to Smad3. HEK293T cells were transfected with various constructs as indicated. The cells were then harvested for luciferase assays 36 h later. Data represent the mean ± SD of three independent experiments. Student’s t-test was used. e, The potential Smad-binding elements in the etv2 promoter. There are two potential Smad-binding elements (SBEs) in the −2516 to −2270 bp region. The SBE in probe A was mutated with G-2396 replaced by A to generate MT probe A, and the SBE in probe B was mutated with G-2312 replaced by A to generate MT probe B. f, Smad3 directly bound to probe A and probe B as shown by EMSA. Cold A, unlabeled probe A; Cold B, unlabeled probe B. MTPA and MTPB, mutants of probe A and B. g,h, MT1-etv2-Luc and MT2-etv2-Luc contained the same mutations in MTPA and MTPB, respectively, while MT-etv2-Luc contained both of the mutations. HEK293T cells were co-transfected with various constructs as indicated. Cells were then harvested for luciferase assays 36 h later (g). Wild-type embryos were injected with 100 pg of the indicated reporter constructs together with or without 150 pg Flag-smad3a mRNA at the one-cell stage, and then these embryos were subjected to luciferase assays at 24 hpf (h). Data represent the mean ± SD of three independent experiments. Student’s t-test was used.

Source data

Extended Data Fig. 9 The expression of p21 remains unchanged during PAA development.

Tg(nkx2.5:ZsYellow) embryos stained with anti-ZsYellow antibody were hybridized with a fluorescence-labeled p21 probe at 24, 32, and 42 hpf. PAA clusters are indicated by white dotted lines. This experiment was repeated independently three times. Dorsal views, anterior to the left. Scale bar, 50 μm.

Extended Data Fig. 10 Reducing the expression of wnt2bb rebalances the proliferation and differentiation of PAA progenitors in tmem88a/b−/− mutants.

a, Wild-type, wnt2bb+/− and wnt2bb−/− embryos in the Tg(nkx2.5:ZsYellow) background were coimmunostained with anti-ZsYellow and anti-β-catenin antibodies. Nuclei were counterstained with DAPI. PAA progenitor clusters are indicated by the white dotted lines. This experiment was repeated independently three times. Dorsal views, anterior to the left. Scale bar, 50 μm. b, The PAAs 3–6 morphology of wild-type, wnt2bb+/− and wnt2bb−/− embryos in the Tg(flk:EGFP;gata1:DsRed) background at 60 hpf. This experiment was repeated independently three times. Lateral views, anterior to the left. Scale bar, 50 µm. c,d, tmem88a/b-deficient Tg(nkx2.5:ZsYellow) embryos were injected with 3 ng cMO or wnt2bb MO at the one-cell stage, then harvested at 28 hpf. PAA progenitors were labeled with anti-ZsYellow antibody. Proliferating cells were visualized by BrdU immunofluorescence. This experiment was repeated independently three times. Representative pictures are shown in (c). Dorsal views, anterior to the left. Scale bar, 50 μm. The percentage of BrdU-positive cells in PAA progenitors is shown in (d). n ≥ 4 per group. Student’s t-test was used. Data are mean ± SD. e,f, The expression changes of nkx2.5 and etv2 in tmem88a/b/ embryos injected with 3 ng cMO or wnt2bb MO (e). The average numbers of etv2+ PAA angioblast clusters were quantified in (f). n ≥ 19 per group. Student’s t-test was used. Data are mean ± SD.

Source data

Supplementary information

Source data

Source Data Fig. 2

Statistical Source Data

Source Data Fig. 3

Statistical Source Data

Source Data Fig. 4

Statistical Source Data

Source Data Fig. 4

Unprocessed western blots

Source Data Fig. 5

Statistical Source Data

Source Data Fig. 6

Statistical Source Data

Source Data Fig. 6

Unprocessed western blots

Source Data Fig. 7

Statistical Source Data

Source Data Fig. 8

Statistical Source Data

Source Data Extended Data Fig./Table 3

Statistical Source Data

Source Data Extended Data Fig./Table 3

Unprocessed western blots

Source Data Extended Data Fig./Table 4

Statistical Source Data

Source Data Extended Data Fig./Table 5

Statistical Source Data

Source Data Extended Data Fig./Table 6

Statistical Source Data

Source Data Extended Data Fig./Table 6

Unprocessed western blots

Source Data Extended Data Fig./Table 7

Unprocessed western blots

Source Data Extended Data Fig./Table 8

Statistical Source Data

Source Data Extended Data Fig./Table 8

Unprocessed EMSA gels

Source Data Extended Data Fig./Table 10

Statistical Source Data

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, M., Liu, J., Mao, A. et al. Tmem88 confines ectodermal Wnt2bb signaling in pharyngeal arch artery progenitors for balancing cell cycle progression and cell fate decision. Nat Cardiovasc Res 2, 234–250 (2023). https://doi.org/10.1038/s44161-023-00215-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s44161-023-00215-z

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing