Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Johan Castille is active.

Publication


Featured researches published by Johan Castille.


PLOS ONE | 2012

Overexpression of miR-30b in the Developing Mouse Mammary Gland Causes a Lactation Defect and Delays Involution

Sandrine Le Guillou; Nezha Sdassi; Johann Laubier; Bruno Passet; Marthe Vilotte; Johan Castille; Denis Laloë; Jacqueline Polyte; Stephan Bouet; Florence Jaffrézic; E. P. Cribiu; Jean-Luc Vilotte; Fabienne Le Provost

Background MicroRNA (miRNA) are negative regulators of gene expression, capable of exerting pronounced influences upon the translation and stability of mRNA. They are potential regulators of normal mammary gland development and of the maintenance of mammary epithelial progenitor cells. This study was undertaken to determine the role of miR-30b on the establishment of a functional mouse mammary gland. miR-30b is a member of the miR-30 family, composed of 6 miRNA that are highly conserved in vertebrates. It has been suggested to play a role in the differentiation of several cell types. Methodology/Principal Findings The expression of miR-30b was found to be regulated during mammary gland development. Transgenic mice overexpressing miR-30b in mammary epithelial cells were used to investigate its role. During lactation, mammary histological analysis of the transgenic mice showed a reduction in the size of alveolar lumen, a defect of the lipid droplets and a growth defect of pups fed by transgenic females. Moreover some mammary epithelial differentiated structures persisted during involution, suggesting a delay in the process. The genes whose expression was affected by the overexpression of miR-30b were characterized by microarray analysis. Conclusion/Significance Our data suggests that miR-30b is important for the biology of the mammary gland and demonstrates that the deregulation of only one miRNA could affect lactation and involution.


RNA Biology | 2015

No effect of an elevated miR-30b level in mouse milk on its level in pup tissues.

Johann Laubier; Johan Castille; Sandrine Le Guillou; Fabienne Le Provost

Recent reports have shown that ingested microRNAs may be transferred to blood, accumulate in tissues and exert canonical regulation on endogenous transcripts. In spite of several attempts to replicate these findings, they have not been confirmed and several questions remain. By using a transgenic mouse model presenting a high level of miR-30b in milk, the horizontal delivery of this microRNA via oral ingestion was studied in pups. Our findings demonstrated that, although very high levels of miR-30b were found in milk and in stomach contents of the pups, we did not detect an increase in miR-30b in tissues of pups fed by transgenic females compared to pups fed by wild-type females.


PLOS ONE | 2011

Fidgetin-like1 is a strong candidate for a dynamic impairment of male meiosis leading to reduced testis weight in mice.

David L'Hôte; Magalie Vatin; Jana Auer; Johan Castille; Bruno Passet; Xavier Montagutelli; Catherine Serres; Daniel Vaiman

Background In a previous work, using an interspecific recombinant congenic mouse model, we reported a genomic region of 23 Mb on mouse chromosome 11 implicated in testis weight decrease and moderate teratozoospermia (∼20–30%), a Quantitative Trait Locus (QTL) called Ltw1. The objective of the present study is to identify the gene underlying this phenotype. Results In the present study, we refined the QTL position to a 5 Mb fragment encompassing only 11 genes. We showed that the low testis weight phenotype was due to kinetic alterations occurring during the first wave of the spermatogenesis where we could point out to an abnormal lengthening of spermatocyte prophase. We identify Fidgetin-like 1 (Fignl1) as the gene underlying the phenotype, since if fulfilled both the physiological and molecular characteristics required. Indeed, amongst the 11 positional candidates it is the only gene that is expressed during meiosis at the spermatocyte stage, and that presents with non-synonymous coding variations differentiating the two mouse strains at the origin of the cross. Conclusions This work prompted us to propose Fignl1 as a novel actor in mammals male meiosis dynamics which has fundamental interest. Besides, this gene is a new potential candidate for human infertilities caused by teratozoospermia and blockades of spermatogenesis. In addition this study demonstrates that interspecific models may be useful for understanding complex quantitative traits.


Frontiers in Cell and Developmental Biology | 2014

The Prion protein family: a view from the placenta

Samira Makzhami; Bruno Passet; Sophie Halliez; Johan Castille; Katayoun Moazami-Goudarzi; Amandine Duchesne; Marthe Vilotte; Hubert Laude; Sophie Mouillet-Richard; Vincent Béringue; Daniel Vaiman; Jean Luc Vilotte

Based on its developmental pattern of expression, early studies suggested the implication of the mammalian Prion protein PrP, a glycosylphosphatidylinositol-anchored ubiquitously expressed and evolutionary conserved glycoprotein encoded by the Prnp gene, in early embryogenesis. However, gene invalidation in several species did not result in obvious developmental abnormalities and it was only recently that it was associated in mice with intra-uterine growth retardation and placental dysfunction. A proposed explanation for this lack of easily detectable developmental-related phenotype is the existence in the genome of one or more gene (s) able to compensate for the absence of PrP. Indeed, two other members of the Prnp gene family have been recently described, Doppel and Shadoo, and the consequences of their invalidation alongside that of PrP tested in mice. No embryonic defect was observed in mice depleted for Doppel and PrP. Interestingly, the co-invalidation of PrP and Shadoo in two independent studies led to apparently conflicting observations, with no apparent consequences in one report and the observation of a developmental defect of the ectoplacental cone that leads to early embryonic lethality in the other. This short review aims at summarizing these recent, apparently conflicting data highlighting the related biological questions and associated implications in terms of animal and human health.


Biochemical and Biophysical Research Communications | 2011

Expression of the prion-like protein Shadoo in the developing mouse embryo

Rachel Young; Stephan Bouet; Jacqueline Polyte; Sandrine Le Guillou; Bruno Passet; Marthe Vilotte; Johan Castille; Vincent Béringue; Fabienne Le Provost; Hubert Laude; Jean Luc Vilotte

The prion-like protein Shadoo has been suggested to compensate for the lack of PrP in Prnp-knockout mice, explaining their lack of extreme phenotype. In adult mice, both PrP and Shadoo have shown overlapping expression patterns and shared functions. Their expression in the mouse embryo has also been suggested to be complementary, as invalidation of both genes results in embryonic lethality. The developmental expression profile of PrP has been described from post-implantation stages up until birth. However the spatial expression pattern of Shadoo in the developing mouse embryo is not known. We previously described the expression profile of the prion-like protein Shadoo in adult mice using Sprn reporter mice (Sprn-GFP and Sprn-LacZ). Here we used these mice to describe the developmental expression of Shadoo between 10.5 and 14.5 dpc. The observed pattern in specific embryonic cell lineages and in extra-embryonic tissues is consistent with the previously reported phenotype resulting from its knockdown.


PLOS Genetics | 2017

Bovine and murine models highlight novel roles for SLC25A46 in mitochondrial dynamics and metabolism, with implications for human and animal health

Amandine Duchesne; Anne Vaiman; Johan Castille; Christian Beauvallet; Pauline Gaignard; Sandrine Floriot; Sabrina Rodriguez; Marthe Vilotte; Laurent Boulanger; Bruno Passet; Olivier Albaric; François Guillaume; Abdelhak Boukadiri; Laurence Richard; Maud Bertaud; Edouard Timsit; Raphaël Guatteo; Florence Jaffrézic; Pierre Calvel; Louise Helary; Rachid Mahla; Diane Esquerre; Christine Péchoux; Sophie Liuu; Jean-Michel Vallat; Didier Boichard; Abdelhamid Slama; Jean-Luc Vilotte; Edmund Kunji

Neuropathies are neurodegenerative diseases affecting humans and other mammals. Many genetic causes have been identified so far, including mutations of genes encoding proteins involved in mitochondrial dynamics. Recently, the “Turning calves syndrome”, a novel sensorimotor polyneuropathy was described in the French Rouge-des-Prés cattle breed. In the present study, we determined that this hereditary disease resulted from a single nucleotide substitution in SLC25A46, a gene encoding a protein of the mitochondrial carrier family. This mutation caused an apparent damaging amino-acid substitution. To better understand the function of this protein, we knocked out the Slc25a46 gene in a mouse model. This alteration affected not only the nervous system but also altered general metabolism, resulting in premature mortality. Based on optic microscopy examination, electron microscopy and on biochemical, metabolic and proteomic analyses, we showed that the Slc25a46 disruption caused a fusion/fission imbalance and an abnormal mitochondrial architecture that disturbed mitochondrial metabolism. These data extended the range of phenotypes associated with Slc25a46 dysfunction. Moreover, this Slc25a46 knock-out mouse model should be useful to further elucidate the role of SLC25A46 in mitochondrial dynamics.


Journal of Virology | 2016

Mutated but not deleted ovine PrPC N-terminal polybasic region strongly interferes with prion propagation in transgenic mice

Manal Khalifé; Fabienne Reine; Sophie Paquet-Fifield; Johan Castille; Laetitia Herzog; Marthe Vilotte; Mohammed Moudjou; Katayoun Moazami-Goudarzi; Samira Makhzami; Bruno Passet; Olivier Andreoletti; Didier Vilette; Hubert Laude; Vincent Béringue; Jean-Luc Vilotte

ABSTRACT Mammalian prions are proteinaceous infectious agents composed of misfolded assemblies of the host-encoded, cellular prion protein (PrP). Physiologically, the N-terminal polybasic region of residues 23 to 31 of PrP has been shown to be involved in its endocytic trafficking and interactions with glycosaminoglycans or putative ectodomains of membrane-associated proteins. Several recent reports also describe this PrP region as important for the toxicity of mutant prion proteins and the efficiency of prion propagation, both in vitro and in vivo. The question remains as to whether the latter observations made with mouse PrP and mouse prions would be relevant to other PrP species/prion strain combinations given the dramatic impact on prion susceptibility of minimal amino acid substitutions and structural variations in PrP. Here, we report that transgenic mouse lines expressing ovine PrP with a deletion of residues 23 to 26 (KKRP) or mutated in this N-terminal region (KQHPH instead of KKRPK) exhibited a variable, strain-dependent susceptibility to prion infection with regard to the proportion of affected mice and disease tempo relative to findings in their wild-type counterparts. Deletion has no major effect on 127S scrapie prion pathogenesis, whereas mutation increased by almost 3-fold the survival time of the mice. Deletion marginally affected the incubation time of scrapie LA19K and ovine bovine spongiform encephalopathy (BSE) prions, whereas mutation caused apparent resistance to disease. IMPORTANCE Recent reports suggested that the N-terminal polybasic region of the prion protein could be a therapeutic target to prevent prion propagation or toxic signaling associated with more common neurodegenerative diseases such as Alzheimers disease. Mutating or deleting this region in ovine PrP completes the data previously obtained with the mouse protein by identifying the key amino acid residues involved.


FEBS Open Bio | 2016

A specific role for PRND in goat foetal Leydig cells is suggested by prion family gene expression during gonad development in goats and mice

Aurélie Allais-Bonnet; Johan Castille; Maëlle Pannetier; Bruno Passet; Maëva Elzaiat; Marjolaine André; Fatemeh Montazer-Torbati; Katayoun Moazami-Goudarzi; Jean-Luc Vilotte; Eric Pailhoux

Three genes of the prion protein gene family are expressed in gonads. Comparative analyses of their expression patterns in mice and goats revealed constant expression of PRNP and SPRN in both species and in both male and female gonads, but with a weaker expression of SPRN. By contrast, expression of PRND was found to be sex‐dimorphic, in agreement with its role in spermatogenesis. More importantly, our study revealed that PRND seems to be a key marker of foetal Leydig cells specifically in goats, suggesting a yet unknown role for its encoded protein Doppel during gonadal differentiation in nonrodent mammals.


Scientific Reports | 2015

Prion protein localizes at the ciliary base during neural and cardiovascular development, and its depletion affects α-tubulin post-translational modifications

Sophie Halliez; Séverine Martin-Lannerée; Bruno Passet; Julia Hernandez-Rapp; Johan Castille; Céline Urien; Sophie Chat; Hubert Laude; Jean-Luc Vilotte; Sophie Mouillet-Richard; Vincent Béringue

Although conversion of the cellular form of the prion protein (PrPC) into a misfolded isoform is the underlying cause of prion diseases, understanding PrPC physiological functions has remained challenging. PrPC depletion or overexpression alters the proliferation and differentiation properties of various types of stem and progenitor cells in vitro by unknown mechanisms. Such involvement remains uncertain in vivo in the absence of any drastic phenotype of mice lacking PrPC. Here, we report PrPC enrichment at the base of the primary cilium in stem and progenitor cells from the central nervous system and cardiovascular system of developing mouse embryos. PrPC depletion in a neuroepithelial cell line dramatically altered key cilium-dependent processes, such as Sonic hedgehog signalling and α-tubulin post-translational modifications. These processes were also affected over a limited time window in PrPC–ablated embryos. Thus, our study reveals PrPC as a potential actor in the developmental regulation of microtubule dynamics and ciliary functions.


PLOS ONE | 2016

Phenotypic and Molecular Alterations in the Mammary Tissue of R-Spondin1 Knock-Out Mice during Pregnancy.

Sead Chadi; Jacqueline Polyte; Lucas Lefevre; Johan Castille; Aude Ehanno; Johann Laubier; Florence Jaffrézic; Fabienne Le Provost

R-spondin1 (Rspo1) is a member of a secreted protein family which has pleiotropic functions in development and stem cell growth. Rspo1 knock-out mice are sex-reversed, but some remain sub-fertile, so they fail to nurse their pups. A lack of Rspo1 expression in the mammary gland results in an absence of duct side-branching development and defective alveolar formation. The aim of this study was to characterize the phenotypic and molecular alterations of mammary gland due to Rspo1 knock-out. Using the transcriptional profiling of mammary tissues, we identified misregulated genes in the mammary gland of Rspo1 knock-out mice during pregnancy. A stronger expression of mesenchymal markers was observed, without modifications to the structure of mammary epithelial tissue. Mammary epithelial cell immunohistochemical analysis revealed a persistence of virgin markers, which signify a delay in cell differentiation. Moreover, serial transplantation experiments showed that Rspo1 is associated with a regenerative potential of mammary epithelial cell control. Our finding also highlights the negatively regulated expression of Rspo1’s partners, Lgr4 and RNF43, in the mammary gland during pregnancy. Moreover, we offer evidence that Tgf-β signalling is modified in the absence of Rspo1. Taken together, our results show an abrupt halt or delay to mammary development during pregnancy due to the loss of a further differentiated function.

Collaboration


Dive into the Johan Castille's collaboration.

Top Co-Authors

Avatar

Bruno Passet

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar

Marthe Vilotte

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar

Sandrine Le Guillou

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar

Hubert Laude

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar

Jacqueline Polyte

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar

Jean Luc Vilotte

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar

Jean-Luc Vilotte

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar

Johann Laubier

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar

Vincent Béringue

Institut national de la recherche agronomique

View shared research outputs
Top Co-Authors

Avatar

Fabienne Le Provost

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge