Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Biol Reprod ; 110(1): 78-89, 2024 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-37776549

RESUMEN

The kinesin light chain 3 protein (KLC3) is the only member of the kinesin light chain protein family that was identified in post-meiotic mouse male germ cells. It plays a role in the formation of the sperm midpiece through its association with both spermatid mitochondria and outer dense fibers (ODF). Previous studies showed a significant correlation between its expression level and sperm motility and quantitative semen parameters in humans, while the overexpression of a KLC3-mutant protein unable to bind ODF also affected the same traits in mice. To further assess the role of KLC3 in fertility, we used CRISPR/Cas9 genome editing in mice and investigated the phenotypes induced by the invalidation of the gene or of a functional domain of the protein. Both approaches gave similar results, i.e. no detectable change in male or female fertility. Testis histology, litter size and sperm count were not altered. Apart from the line-dependent alterations of Klc3 mRNA levels, testicular transcriptome analysis did not reveal any other changes in the genes tested. Western analysis supported the absence of KLC3 in the gonads of males homozygous for the inactivating mutation and a strong decrease in expression in males homozygous for the allele lacking one out of the five tetratricopeptide repeats. Overall, these observations raise questions about the supposedly critical role of this kinesin in reproduction, at least in mice where its gene mutation or inactivation did not translate into fertility impairment.


Asunto(s)
Cinesinas , Motilidad Espermática , Animales , Femenino , Humanos , Masculino , Ratones , Fertilidad/genética , Cinesinas/genética , Cinesinas/metabolismo , Ratones Noqueados , Mutación , Proteínas/metabolismo , Semen , Motilidad Espermática/genética , Espermatogénesis/fisiología , Espermatozoides/metabolismo , Testículo/metabolismo
2.
J Transl Med ; 22(1): 337, 2024 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-38589873

RESUMEN

BACKGROUND: The mesenchymal subtype of colorectal cancer (CRC), associated with poor prognosis, is characterized by abundant expression of the cellular prion protein PrPC, which represents a candidate therapeutic target. How PrPC is induced in CRC remains elusive. This study aims to elucidate the signaling pathways governing PrPC expression and to shed light on the gene regulatory networks linked to PrPC. METHODS: We performed in silico analyses on diverse datasets of in vitro, ex vivo and in vivo models of mouse CRC and patient cohorts. We mined ChIPseq studies and performed promoter analysis. CRC cell lines were manipulated through genetic and pharmacological approaches. We created mice combining conditional inactivation of Apc in intestinal epithelial cells and overexpression of the human prion protein gene PRNP. Bio-informatic analyses were carried out in two randomized control trials totalizing over 3000 CRC patients. RESULTS: In silico analyses combined with cell-based assays identified the Wnt-ß-catenin and glucocorticoid pathways as upstream regulators of PRNP expression, with subtle differences between mouse and human. We uncover multiple feedback loops between PrPC and these two pathways, which translate into an aggravation of CRC pathogenesis in mouse. In stage III CRC patients, the signature defined by PRNP-CTNNB1-NR3C1, encoding PrPC, ß-catenin and the glucocorticoid receptor respectively, is overrepresented in the poor-prognosis, mesenchymal subtype and associates with reduced time to recurrence. CONCLUSIONS: An unleashed PrPC-dependent vicious circle is pathognomonic of poor prognosis, mesenchymal CRC. Patients from this aggressive subtype of CRC may benefit from therapies targeting the PRNP-CTNNB1-NR3C1 axis.


Asunto(s)
Neoplasias del Colon , Neoplasias Colorrectales , Humanos , Ratones , Animales , Proteínas Priónicas/genética , Proteínas Priónicas/metabolismo , beta Catenina/metabolismo , Glucocorticoides , Neoplasias del Colon/genética , Neoplasias Colorrectales/genética , Fenotipo , Pronóstico , Vía de Señalización Wnt , Regulación Neoplásica de la Expresión Génica , Línea Celular Tumoral
3.
Genet Sel Evol ; 55(1): 40, 2023 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-37308849

RESUMEN

BACKGROUND: Inherited epidermolysis bullosa (EB) is a group of painful and life-threatening genetic disorders that are characterized by mechanically induced blistering of the skin and mucous membranes. Congenital skin fragility resembling EB was recently reported in three Charolais calves born in two distinct herds from unaffected parents. Phenotypic and genetic analyses were carried out to describe this condition and its molecular etiology. RESULTS: Genealogical, pathological and histological investigations confirmed the diagnosis of recessive EB. However, the affected calves showed milder clinical signs compared to another form of EB, which was previously reported in the same breed and is caused by a homozygous deletion of the ITGB4 gene. Homozygosity mapping followed by analysis of the whole-genome sequences of two cases and 5031 control individuals enabled us to prioritize a splice donor site of ITGA6 (c.2160 + 1G > T; Chr2 g.24112740C > A) as the most compelling candidate variant. This substitution showed a perfect genotype-phenotype correlation in the two affected pedigrees and was found to segregate only in Charolais, and at a very low frequency (f = 1.6 × 10-4) after genotyping 186,154 animals from 15 breeds. Finally, RT-PCR analyses revealed increased retention of introns 14 and 15 of the ITGA6 gene in a heterozygous mutant cow compared with a matched control. The mutant mRNA is predicted to cause a frameshift (ITGA6 p.I657Mfs1) that affects the assembly of the integrin α6ß4 dimer and its correct anchoring to the cell membrane. This dimer is a key component of the hemidesmosome anchoring complex, which ensures the attachment of basal epithelial cells to the basal membrane. Based on these elements, we arrived at a diagnosis of junctional EB. CONCLUSIONS: We report a rare example of partial phenocopies observed in the same breed and due to mutations that affect two members of the same protein dimer, and provide the first evidence of an ITGA6 mutation that causes EB in livestock species.


Asunto(s)
Epidermólisis Ampollosa de la Unión , Femenino , Bovinos , Animales , Homocigoto , Eliminación de Secuencia , Mutación , Mutación del Sistema de Lectura
4.
Biochem Biophys Res Commun ; 551: 1-6, 2021 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-33713980

RESUMEN

Shadoo and PrP belongs to the same protein family, whose biological function remains poorly understood. Previous experiments reported potential functional redundancies or antagonisms between these two proteins, depending on the tissue analysed. While knockdown experiments suggested the requirement of Shadoo in the absence of PrP during early mouse embryogenesis, knockout ones, on the contrary, highlighted little impact, if any, of the double-knockout of these two loci. In the present study, we reinvestigated the phenotype associated with the concomitant knockout of these two genes using newly produced FVB/N Sprn knockout mice. In this genetic background, the combined two genes' knockout induces intra-uterine growth retardations, likely resulting from placental failures highlighted by transcriptomic analyses that revealed potential redundant or antagonist roles of these two proteins in different developmental-related pathways. It also induced an increased perinatal-lethality and ascertained the role of these two loci in the lactation process.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , Proteínas Priónicas/metabolismo , Reproducción/fisiología , Animales , Animales Recién Nacidos/crecimiento & desarrollo , Desarrollo Embrionario , Femenino , Proteínas Ligadas a GPI , Genes Letales , Lactancia/genética , Lactancia/fisiología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Fenotipo , Placentación , Embarazo , Proteínas Priónicas/deficiencia , Proteínas Priónicas/genética , Reproducción/genética , Transcriptoma
5.
Antimicrob Agents Chemother ; 64(10)2020 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-32660994

RESUMEN

Respiratory syncytial virus (RSV) is the main cause of severe respiratory infection in young children worldwide, and no therapies have been approved for the treatment of RSV infection. Data from recent clinical trials of fusion or L polymerase inhibitors for the treatment of RSV-infected patients revealed the emergence of escape mutants, highlighting the need for the discovery of inhibitors with novel mechanisms of action. Here we describe stapled peptides derived from the N terminus of the phosphoprotein (P) that act as replication inhibitors. We demonstrate that these peptides inhibit RSV replication in vitro and in vivo by preventing the formation of the N0-P complex. The present strategy provides a novel means of targeting RSV replication with constrained macrocyclic peptides or small molecules and is broadly applicable to other viruses of the Mononegavirales order.


Asunto(s)
Antivirales , Péptidos , Conformación Proteica en Hélice alfa , Virus Sincitial Respiratorio Humano , Animales , Antivirales/farmacología , Humanos , Ratones , Péptidos/farmacología , Fosfoproteínas/farmacología , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Replicación Viral
6.
PLoS Genet ; 13(4): e1006597, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28376083

RESUMEN

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.


Asunto(s)
Dinámicas Mitocondriales/genética , Proteínas Mitocondriales/genética , Proteínas de Transporte de Fosfato/genética , Polineuropatías/genética , Proteómica , Sustitución de Aminoácidos/genética , Animales , Bovinos , Humanos , Ratones , Mitocondrias/genética , Mitocondrias/patología , Mutación , Fenotipo , Polineuropatías/patología , Polineuropatías/veterinaria
7.
Am J Hum Genet ; 96(4): 631-9, 2015 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-25772937

RESUMEN

Congenital microcoria (MCOR) is a rare autosomal-dominant disorder characterized by inability of the iris to dilate owing to absence of dilator pupillae muscle. So far, a dozen MCOR-affected families have been reported worldwide. By using whole-genome oligonucleotide array CGH, we have identified deletions at 13q32.1 segregating with MCOR in six families originating from France, Japan, and Mexico. Breakpoint sequence analyses showed nonrecurrent deletions in 5/6 families. The deletions varied from 35 kbp to 80 kbp in size, but invariably encompassed or interrupted only two genes: TGDS encoding the TDP-glucose 4,6-dehydratase and GPR180 encoding the G protein-coupled receptor 180, also known as intimal thickness-related receptor (ITR). Unlike TGDS which has no known function in muscle cells, GPR180 is involved in the regulation of smooth muscle cell growth. The identification of a null GPR180 mutation segregating over two generations with iridocorneal angle dysgenesis, which can be regarded as a MCOR endophenotype, is consistent with the view that deletions of this gene, with or without the loss of elements regulating the expression of neighboring genes, are the cause of MCOR.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 13/genética , Trastornos de la Pupila/congénito , Receptores de Superficie Celular/genética , Secuencia de Bases , Hibridación Genómica Comparativa , Componentes del Gen , Genes Dominantes/genética , Humanos , Hidroliasas/genética , Datos de Secuencia Molecular , Mutación/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Linaje , Trastornos de la Pupila/genética , Trastornos de la Pupila/patología , Receptores Acoplados a Proteínas G , Análisis de Secuencia de ADN
8.
J Virol ; 90(3): 1638-46, 2016 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-26608316

RESUMEN

UNLABELLED: 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 Alzheimer's 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.


Asunto(s)
Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas PrPC/genética , Proteínas PrPC/metabolismo , Enfermedades por Prión/patología , Animales , Modelos Animales de Enfermedad , Ratones Transgénicos , Mutación Missense , Eliminación de Secuencia , Ovinos
9.
Sci Rep ; 14(1): 9240, 2024 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-38649394

RESUMEN

In MeLiM minipigs, melanomas develop around birth, can metastasize, and have histopathologic characteristics similar to humans. Interestingly, MeLiM melanomas eventually regress. This favorable outcome raises the question of their malignancy, which we investigated. We clinically followed tens of tumors from onset to first signs of regression. Transcriptome analysis revealed an enrichment of all cancer hallmarks in melanomas, although no activating or suppressing somatic mutation were found in common driver genes. Analysis of tumor cell genomes revealed high mutation rates without UV signature. Canonical proliferative, survival and angiogenic pathways were detected in MeLiM tumor cells all along progression stages. Functionally, we show that MeLiM melanoma cells are capable to grow in immunocompromised mice, with serial passages and for a longer time than in MeLiM pigs. Pigs set in place an immune response during progression with dense infiltration by myeloid cells while melanoma cells are deficient in B2M expression. To conclude, our data on MeLiM melanomas reveal several malignancy characteristics. The combination of these features with the successful spontaneous regression of these tumors make it an outstanding model to study an efficient anti-tumor immune response.


Asunto(s)
Melanoma , Regresión Neoplásica Espontánea , Porcinos Enanos , Animales , Porcinos , Melanoma/patología , Melanoma/genética , Ratones , Regresión Neoplásica Espontánea/patología , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/genética , Mutación , Regulación Neoplásica de la Expresión Génica , Perfilación de la Expresión Génica , Modelos Animales de Enfermedad
10.
J Gen Virol ; 94(Pt 6): 1435-1440, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23388201

RESUMEN

In naturally acquired transmissible spongiform encephalopathies, the pathogenic agents or prions spread from the sites of initial peripheral uptake or replication to the brain where they cause progressive and fatal neurodegeneration. Routing via the peripheral nervous system is considered to be one of the main pathways to the central nervous system. Replication of prions in Schwann cells is viewed as a potentially important mechanism for efficient prion spread along nerves. Here we used a Cre-loxP mouse transgenetic approach to disrupt host-encoded prion protein (PrP(C)) specifically in myelinating Schwann cells. Despite the use of infection routes targeting highly myelinated nerves, there was no alteration in mouse prion pathogenesis, suggesting that conversion-dependent, centripetal spread of prions does not crucially rely on PrP(C) expressed by myelinating Schwann cells.


Asunto(s)
Marcación de Gen , Vaina de Mielina/metabolismo , Proteínas PrPC/genética , Proteínas PrPC/metabolismo , Enfermedades por Prión/metabolismo , Enfermedades por Prión/patología , Células de Schwann/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Ratones Transgénicos , Vaina de Mielina/patología
11.
Biomolecules ; 13(9)2023 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-37759696

RESUMEN

Intestinal barrier integrity is essential in order to maintain the homeostasis of mucosal functions and efficient defensive reactions against chemical and microbial challenges. An impairment of the intestinal barrier has been observed in several chronic diseases. The gut microbiota and its impact on intestinal homeostasis is well described and numerous studies suggest the ability of some probiotic strains to protect the intestinal epithelial integrity and host homeostasis. In this work, we aimed to assess the beneficial effects of three Lactobacillus strains (Lacticaseibacillus rhamnosus LR04, Lacticaseibacillus casei LC03, and Lactiplantibacillus plantarum CNCM I-4459) and their mechanism of action in low-grade inflammation or neonatal maternal separation models in mice. We compared the impact of these strains to that of the well-known probiotic Lacticaseibacillus rhamnosus GG. Our results demonstrated that the three strains have the potential to restore the barrier functions by (i) increasing mucus production, (ii) restoring normal permeability, and (iii) modulating colonic hypersensitivity. Moreover, gene expression analysis of junctional proteins revealed the implication of Claudin 2 and Cingulin in the mechanisms that underlie the interactions between the strains and the host. Taken together, our data suggest that LR04, CNCM I-4459, and LC03 restore the functions of an impaired intestinal barrier.


Asunto(s)
Lacticaseibacillus rhamnosus , Lactobacillus , Animales , Ratones , Privación Materna , Homeostasis , Inflamación
12.
Biochem Biophys Res Commun ; 412(4): 752-6, 2011 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-21871438

RESUMEN

The protein Shadoo (Sho) is a paralogue of prion protein, and encoded by the gene Sprn. Like prion protein it is primarily expressed in central nervous system, and has been shown to have a similar expression pattern in certain regions of the brain. We have generated reporter mice carrying a transgene encompassing the Sprn promoter, exon 1, intron 1 and the 5'-end of exon 2 driving expression of either the LacZ or GFP reporter gene to study the expression profile of Shadoo in mice. Expression of the reporter genes was analysed in brains of these transgenic mice and was shown to mimic that of the endogenous gene expression, previously described by Watts et al. [1]. Consequently, the Sprn-LacZ mice were used to study the spatial expression of Sho in other tissues of the adult mouse. Several tissues were collected and stained for ß-gal activity, including the thymus, heart, lung, liver, kidney, spleen, intestine, muscle, and gonads. From this array of tissues, the transgene was consistently expressed only in specific cell types of the testicle and ovary, suggesting a role for Shadoo in fertility and reproduction. These mice may serve as a useful tool in deciphering the regulation of the prion-like gene Sprn and thus, indirectly, of the Shadoo protein.


Asunto(s)
Gónadas/metabolismo , Proteínas del Tejido Nervioso/genética , Priones/genética , Animales , Proteínas Ligadas a GPI , Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Ratones , Ratones Transgénicos , Distribución Tisular , beta-Galactosidasa/genética
13.
Biochem Biophys Res Commun ; 416(1-2): 184-7, 2011 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-22093825

RESUMEN

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.


Asunto(s)
Embrión de Mamíferos/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Animales , Linaje de la Célula , Embrión de Mamíferos/citología , Proteínas Ligadas a GPI , Técnicas de Silenciamiento del Gen , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas Priónicas , Priones/biosíntesis , Priones/genética , Transgenes , beta-Galactosidasa/biosíntesis , beta-Galactosidasa/genética
14.
Front Cell Dev Biol ; 9: 754054, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35127699

RESUMEN

Male gametogenesis involves both mitotic divisions to amplify germ cell progenitors that gradually differentiate and meiotic divisions. Centrosomal regulation is essential for both types of divisions, with centrioles remaining tightly paired during the interphase. Here, we generated and characterized the phenotype of mutant mice devoid of Cep250/C-Nap1, a gene encoding for a docking protein for fibers linking centrioles, and characterized their phenotype. The Cep250 -/- mice presented with no major defects, apart from male infertility due to a reduction in the spermatogonial pool and the meiotic blockade. Spermatogonial stem cells expressing Zbtb16 were not affected, whereas the differentiating spermatogonia were vastly lost. These cells displayed abnormal γH2AX-staining, accompanied by an increase in the apoptotic rate. The few germ cells that survived at this stage, entered the meiotic prophase I and were arrested at a pachytene-like stage, likely due to synapsis defects and the unrepaired DNA double-strand breaks. In these cells, centrosomes split up precociously, with γ-tubulin foci being separated whereas these were closely associated in wild-type cells. Interestingly, this lack of cohesion was also observed in wild-type female meiocytes, likely explaining the normal fertility of Cep250 -/- female mice. Taken together, this study proposes a specific requirement of centrosome cohesion in the male germline, with a crucial role of CEP250 in both differentiating spermatogonia and meiotic spermatocytes.

15.
BMC Genomics ; 11: 448, 2010 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-20649983

RESUMEN

BACKGROUND: The physiological function of the prion protein remains largely elusive while its key role in prion infection has been expansively documented. To potentially assess this conundrum, we performed a comparative transcriptomic analysis of the brain of wild-type mice with that of transgenic mice invalidated at this locus either at the zygotic or at the adult stages. RESULTS: Only subtle transcriptomic differences resulting from the Prnp knockout could be evidenced, beside Prnp itself, in the analyzed adult brains following microarray analysis of 24 109 mouse genes and QPCR assessment of some of the putatively marginally modulated loci. When performed at the adult stage, neuronal Prnp disruption appeared to sequentially induce a response to an oxidative stress and a remodeling of the nervous system. However, these events involved only a limited number of genes, expression levels of which were only slightly modified and not always confirmed by RT-qPCR. If not, the qPCR obtained data suggested even less pronounced differences. CONCLUSIONS: These results suggest that the physiological function of PrP is redundant at the adult stage or important for only a small subset of the brain cell population under classical breeding conditions. Following its early reported embryonic developmental regulation, this lack of response could also imply that PrP has a more detrimental role during mouse embryogenesis and that potential transient compensatory mechanisms have to be searched for at the time this locus becomes transcriptionally activated.


Asunto(s)
Envejecimiento/genética , Encéfalo/metabolismo , Perfilación de la Expresión Génica , Silenciador del Gen , Priones/genética , Transcripción Genética , Cigoto/metabolismo , Animales , Encéfalo/citología , Femenino , Técnicas de Inactivación de Genes , Sitios Genéticos/genética , Masculino , Ratones , Neuronas/metabolismo
16.
Sci Rep ; 10(1): 6765, 2020 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-32317725

RESUMEN

Shadoo belongs to the prion protein family, an evolutionary conserved and extensively studied family due to the implication of PrP in Transmissible Spongiform Encephalopathies. However, the biological function of these genes remains poorly understood. While Sprn-knockdown experiments suggested an involvement of Shadoo during mouse embryonic development, Sprn-knockout experiments in 129Pas/C57BL/6J or 129Pas/FVB/NCr mice did not confirm it. In the present study, we analyzed the impact of Sprn gene invalidation in a pure FVB/NJ genetic background, using a zinc finger nuclease approach. The in-depth analysis of the derived knockout transgenic mice revealed a significant increase in embryonic lethality at early post-implantation stages, a growth retardation of young Sprn-knockout pups fed by wild type mice and a lactation defect of Sprn-knockout females. Histological and transcriptional analyses of knockout E7.5 embryos, E14.5 placentas and G7.5 mammary glands revealed specific roles of the Shadoo protein in mouse early embryogenesis, tissue development and differentiation with a potential antagonist action between PrP and Shadoo. This study thus highlights the entanglement between the proteins of the prion family.


Asunto(s)
Diferenciación Celular/genética , Desarrollo Embrionario/genética , Proteínas del Tejido Nervioso/genética , Proteínas Priónicas/genética , Animales , Proteínas Ligadas a GPI , Humanos , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/metabolismo , Ratones , Ratones Noqueados , Células Madre Embrionarias de Ratones/metabolismo , Organogénesis/genética , Enfermedades por Prión/genética , Enfermedades por Prión/patología
17.
Transgenic Res ; 18(4): 649-54, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19184501

RESUMEN

RSPO1 is a newly discovered gene involved in sex differentiation. Two goat BAC clones encompassing the RSPO1 gene (gRSPO1) were injected into mouse oocytes and several transgenic lines derived. Both clones induced gRSPO1 over-expression in various tissues, including male and female gonads, with no obvious phenotype and normal sex-ratios. Introgression of the gRSPO1 transgene into a mouse RSPO1 knockout genotype resulted in the rescue of the fertility and the disappearance of the masculinized gonadic features of the females, demonstrating the functionality of the goat protein in a mouse context. On the contrary, over-expression of gRSPO1 within a mSRY or a gSRY-XX genotypes did not interfere with the SRY-induced male phenotype.


Asunto(s)
Trastornos del Desarrollo Sexual , Cabras/genética , Testículo/crecimiento & desarrollo , Trombospondinas/fisiología , Animales , Animales Modificados Genéticamente , Diferenciación Celular/genética , Cromosomas Artificiales Bacterianos/genética , Femenino , Fertilidad/genética , Cabras/fisiología , Humanos , Masculino , Ratones , Ratones Noqueados , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Testículo/citología , Trombospondinas/genética , Transgenes , Cromosoma X/genética , Cromosoma Y/genética
18.
FEBS Lett ; 582(15): 2219-24, 2008 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-18501713

RESUMEN

Spatial and temporal control of ovine prion protein (Prnp) gene expression was achieved in mice using two transgenes: a Prnp minigene with tet-operator sequences inserted 5' to exon 1 and a mouse neurofilament genomic clone carrying the chimeric-repressor TRSID cDNA. In bi-transgenic mice, ovine PrP(C) expression could be reversibly controlled in neuronal cells by doxycycline treatment whereas it remains constant in other cell types. Overall, this model opens opportunities to assess the involvement of cell types in prion diseases and PrP physiological function. It demonstrates the potentiality of the TRSID-silencer to precisely control temporal and spatial gene expression in vivo.


Asunto(s)
Proteínas PrPC/genética , Ovinos/genética , Elementos Silenciadores Transcripcionales , Transgenes , Animales , Regulación hacia Abajo , Expresión Génica , Ratones , Ratones Transgénicos , Neuronas/metabolismo
19.
Front Cell Dev Biol ; 2: 35, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25364742

RESUMEN

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.

20.
Hypertension ; 61(3): 662-8, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23357179

RESUMEN

Preeclampsia (PE) is a common human-specific pregnancy disorder defined by hypertension and proteinuria during gestation and responsible for maternal and fetal morbimortality. STOX1, encoding a transcription factor, was the first gene associated with PE as identified by positional cloning approaches. Its overexpression in choriocarcinoma cells mimics the transcriptional consequences of PE in the human placenta. Here, we created transgenic mouse strains overexpressing human STOX1. Wild-type female mice crossed with transgenic male mice reproduce accurately the symptoms of severe PE: gestational hypertension, proteinuria, and elevated plasma levels of soluble fms-like tyrosine kinase 1 and soluble endoglin. Placental and kidney histology were altered. Symptoms were prevented or alleviated by aspirin treatment. STOX1-overexpressing mice constitute a unique model for studying PE, allow testing therapeutic approaches, and assessing the long-term effects of the preeclamptic syndrome.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Antihipertensivos/uso terapéutico , Aspirina/uso terapéutico , Proteínas Portadoras/biosíntesis , Placenta/metabolismo , Preeclampsia/tratamiento farmacológico , Animales , Proteínas Portadoras/efectos adversos , Proteínas Portadoras/genética , Modelos Animales de Enfermedad , Endoglina , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular/sangre , Riñón/patología , Masculino , Ratones , Ratones Transgénicos , Placenta/patología , Preeclampsia/etiología , Preeclampsia/genética , Embarazo , Índice de Severidad de la Enfermedad , Receptor 1 de Factores de Crecimiento Endotelial Vascular/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA