, Six gestating females of each genotype were used at each analyzed stages. Individual RNA extractions were performed using the RNeasy Lipid Tissue Mini kit, RNA purifications and RT-(q)PCR

, Reverse transcription was performed on 5?g of total RNA, using Invitrogen SuperScript TMIII reverse transcriptase kit (18080051) or RTVilo (11754050) and random primers, according to the manufacturer's instructions. Amplification of the mouse Sprn cDNA ORF was performed using oligonucleotides 5?-taggcttgtgaccaattcttgcc-3? and 5?-ggagtttagcctggtctaaggc-3?, that are located on the two separate Sprn exons to avoid amplification of potential contaminating genomic DNA. PCR conditions were made of 40 amplifications cycles; 94 °C-30 s, 60 °C-60 s and 72 °C-30 s, using GC-rich adapted PCR amplification kit (KAPA2G Robust PCR kit, SigmaAldrich). Two WT and two Sprn 0/0 mouse brains were analyzed. For RT-qPCR, quantification was performed on triplicates, using 3 different RNA samples for each genotype, using the ABsolute Blue QPCR SYBR Green ROX Mix (Thermo Scientific) and standard PCR conditions. Primers were designed on separate exons, RNA concentration was calculated by electro-spectrophotometry and the RNA integrity checked with the Agilent Bioanalyser

S. B. Prusiner, Early evidence that a protease-resistant protein is an active component of the infectious prion, Cell, vol.116, issue.2, p.109, 2004.

H. Büeler, Normal development and behaviour of mice lacking the neuronal cell-surface PrP protein, Nature, vol.356, issue.6370, pp.577-582, 1992.

J. A. Richt, Production of cattle lacking prion protein, Nat. Biotechnol, vol.25, issue.1, pp.132-138, 2007.

G. Yu, Generation of goats lacking prion protein, Mol. Reprod. Dev, vol.76, issue.1, p.3, 2009.

S. L. Benestad, L. Austbø, M. A. Tranulis, A. Espenes, and I. Olsaker, Healthy goats naturally devoid of prion protein, Vet. Res, vol.43, p.87, 2012.

P. L. Leighton, R. Kanyo, G. J. Neil, N. M. Pollock, and W. T. Allison, Prion gene paralogs are dispensable for early zebrafish development but have non-additive roles in seizure susceptibility, J Biol Chem, 2018.

D. Westaway, N. Daude, S. Wohlgemuth, and P. Harrison, The PrP-like proteins Shadoo and Doppel, Top. Curr. Chem, vol.305, pp.225-256, 2011.

D. Paisley, Male infertility and DNA damage in Doppel knockout and prion protein/Doppel double-knockout mice, Am. J. Pathol, vol.4, issue.6, pp.2279-2288, 2004.

N. Daude and D. Westaway, Biological properties of the PrP-like Shadoo protein, Front. Biosci, vol.16, pp.1505-1516, 2011.

R. Young, The prion or the related Shadoo protein is required for early mouse embryogenesis, FEBS Lett, vol.583, pp.3296-3300, 2009.
URL : https://hal.archives-ouvertes.fr/hal-01193399

B. Passet, Prion protein and Shadoo are involved in overlapping embryonic pathways and trophoblastic development, PLoS One, vol.7, issue.7, p.41959, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00999993

N. Daude, Knockout of the prion protein (PrP)-like Sprn gene does not produce embryonic lethality in combination with PrP(C)-deficiency, Proc. Natl Acad. Sci. USA, vol.109, issue.23, pp.9035-9040, 2012.

A. Rossi, Genetic compensation induced by deleterious mutations but not gene knockdowns, Nature, vol.524, issue.7564, pp.230-233, 2015.

N. Alfaidy, Prion protein expression and functional importance in developmental angiogenesis: role in oxidative stress and copper homeostasis, Antioxid. Redox Signal, vol.18, issue.4, pp.400-411, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-00734867

A. Kishor, S. E. Fritz, and J. R. Hogg, Nonsense-mediated mRNA decay: The challenge of telling right from wrong in a complexe transcriptome, Wiley Interdiscip. Rev. RNA, vol.1548, pp.1-22, 2019.

L. Wu, J. Zheng, P. Chen, Q. Liu, and Y. Yuan, Small nucleolar RNA ACA11 promotes proliferation, migration and invasion in hepatocellular carcinoma by targeting the PI3K/AKT signaling pathway, Biomed. Pharmacother, vol.90, pp.705-712, 2017.

G. Li, Small nucleolar RNA 47 promotes tumorigenesis by regulating EMT markers in hepatocellular carcinoma, Minerva Med, vol.108, pp.396-404, 2017.

M. Mehrabian, S. Ehsani, and G. Schmitt-ulms, An emerging role of the cellular prion protein as a modulator of a morphogenetic program underlying epithelial-to-mesenchymal transition, Front. Cell Dev. Biol, vol.18, issue.2, p.53, 2014.

F. Ghodrati, The prion protein is embedded in a molecular environment that modulates transforming growth factor ? and integrin signaling, Sci. Rep, vol.8, issue.1, p.8654, 2018.

N. Daude, H. Gapeshina, B. Dong, I. Winship, and D. Westaway, Neuroprotective properties of the PrP-like Shadoo glycoprotein assessed in the middle cerebral artery occlusion model of ischemia, Prion, vol.9, issue.5, p.6765, 2015.

A. Nyeste, Expression of the Prion Protein Family Member Shadoo Causes Drug Hypersensitivity That Is Diminished by the Coexpression of the Wild Type Prion Protein, J. Biol. Chem, vol.291, issue.9, pp.4473-4486, 2016.

M. Cardoso-moreira, Gene expression across mammalian organ development, Nature, vol.571, issue.7766, pp.505-509, 2019.

T. Iwaki, Fibrinogen stabilizes placental-maternal attachment during embryonic development in the mouse, Am. J. Pathol, vol.160, issue.3, pp.1021-1034, 2002.

F. Charlton, The protective effect of apolipoprotein in models of trophoblast invasion and preeclampsia, Am. J. Physiol. Regul. Integr. Comp. Physiol, vol.312, issue.1, pp.40-48, 2017.

M. J. Liao, Enrichment of a population of mammary gland cells that form mammospheres and have in vivo repopulating activity, Cancer Res, vol.67, issue.17, pp.8131-8138, 2007.

E. Honvo-houéto, The endoplasmic reticulum and casein-containing vesicles contribute to milk fat globule membrane, Mol. Biol. Cell, vol.27, pp.2946-64, 2016.

R. Mesilati-stahy, K. Mida, and N. Argov-argaman, Size-dependent lipid content of bovine milk fat globule and membrane phospholipids, J. Agric. Food Chem, vol.59, issue.13, pp.7427-7462, 2011.

Y. Yamada, Impact of novel oncogenic pathways regulated by antitumor miR-451a in renal cell carcinoma, Cancer Sci, vol.109, issue.4, pp.1239-1253, 2018.

J. C. Watts, Protease-resistant prions selectively decrease Shadoo protein, PLoS Pathog, vol.7, issue.11, p.1002382, 2011.

H. Wang, Overexpression of Shadoo protein in transgenic mice does not impact the pathogenesis of scrapie, Neurosci. Lett, vol.496, issue.1, pp.1-4, 2011.

A. Miranda, E. Pericuesta, M. Á. Ramírez, and A. Gutierrez-adan, Prion protein expression regulates embryonic stem cell pluripotency and differentiation, PLoS One, vol.6, issue.4, p.18422, 2011.

G. Miele, Embryonic activation and developmental expression of the murine prion protein gene, Gene Expr, vol.11, issue.1, pp.1-12, 2003.