Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Nature ; 614(7947): 343-348, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36697821

RESUMEN

Transcriptional enhancer elements are responsible for orchestrating the temporal and spatial control over gene expression that is crucial for programming cell identity during development1-3. Here we describe a novel enhancer element that is important for regulating the expression of Prox1 in lymphatic endothelial cells. This evolutionarily conserved enhancer is bound by key lymphatic transcriptional regulators including GATA2, FOXC2, NFATC1 and PROX1. Genome editing of the enhancer to remove five nucleotides encompassing the GATA2-binding site resulted in perinatal death of homozygous mutant mice due to profound lymphatic vascular defects. Lymphatic endothelial cells in enhancer mutant mice exhibited reduced expression of genes characteristic of lymphatic endothelial cell identity and increased expression of genes characteristic of haemogenic endothelium, and acquired the capacity to generate haematopoietic cells. These data not only reveal a transcriptional enhancer element important for regulating Prox1 expression and lymphatic endothelial cell identity but also demonstrate that the lymphatic endothelium has haemogenic capacity, ordinarily repressed by Prox1.


Asunto(s)
Células Endoteliales , Elementos de Facilitación Genéticos , Hematopoyesis , Vasos Linfáticos , Animales , Ratones , Células Endoteliales/metabolismo , Elementos de Facilitación Genéticos/genética , Hematopoyesis/genética , Proteínas de Homeodominio/metabolismo , Vasos Linfáticos/citología , Vasos Linfáticos/metabolismo , Factores de Transcripción/metabolismo
2.
Proc Natl Acad Sci U S A ; 119(46): e2213308119, 2022 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-36346842

RESUMEN

Invasive rodents are a major cause of environmental damage and biodiversity loss, particularly on islands. Unlike insects, genetic biocontrol strategies including population-suppressing gene drives with biased inheritance have not been developed in mice. Here, we demonstrate a gene drive strategy (tCRISPR) that leverages super-Mendelian transmission of the t haplotype to spread inactivating mutations in a haplosufficient female fertility gene (Prl). Using spatially explicit individual-based in silico modeling, we show that tCRISPR can eradicate island populations under a range of realistic field-based parameter values. We also engineer transgenic tCRISPR mice that, crucially, exhibit biased transmission of the modified t haplotype and Prl mutations at levels our modeling predicts would be sufficient for eradication. This is an example of a feasible gene drive system for invasive alien rodent population control.


Asunto(s)
Biodiversidad , Tecnología de Genética Dirigida , Ratones , Femenino , Animales , Roedores , Genética de Población , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas
3.
Nucleic Acids Res ; 49(18): 10785-10795, 2021 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-34534334

RESUMEN

Precise genomic modification using prime editing (PE) holds enormous potential for research and clinical applications. In this study, we generated all-in-one prime editing (PEA1) constructs that carry all the components required for PE, along with a selection marker. We tested these constructs (with selection) in HEK293T, K562, HeLa and mouse embryonic stem (ES) cells. We discovered that PE efficiency in HEK293T cells was much higher than previously observed, reaching up to 95% (mean 67%). The efficiency in K562 and HeLa cells, however, remained low. To improve PE efficiency in K562 and HeLa, we generated a nuclease prime editor and tested this system in these cell lines as well as mouse ES cells. PE-nuclease greatly increased prime editing initiation, however, installation of the intended edits was often accompanied by extra insertions derived from the repair template. Finally, we show that zygotic injection of the nuclease prime editor can generate correct modifications in mouse fetuses with up to 100% efficiency.


Asunto(s)
Proteína 9 Asociada a CRISPR , Edición Génica , Animales , Proteína 9 Asociada a CRISPR/genética , Células Cultivadas , Células Madre Embrionarias/metabolismo , Células HEK293 , Células HeLa , Humanos , Células K562 , Ratones , Plásmidos/genética , Cigoto
4.
Reproduction ; 161(2): 135-144, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33434162

RESUMEN

Animal models are needed to develop interventions to prevent or treat intrauterine growth restriction (IUGR). Foetal growth rates and effects of in utero exposures differ between sexes, but little is known about sex-specific effects of increasing litter size. We established a murine IUGR model using pregnancies generated by multiple embryo transfers, and evaluated sex-specific responses to increasing litter size. CBAF1 embryos were collected at gestation day 0.5 (GD0.5) and 6, 8, 10 or 12 embryos were transferred into each uterine horn of pseudopregnant female CD1 mice (n = 32). Foetal and placental outcomes were measured at GD18.5. In the main experiment, foetuses were genotyped (Sry) for analysis of sex-specific outcomes. The number of implantation sites (P = 0.033) and litter size (number of foetuses, P = 0.008) correlated positively with the number of embryos transferred, while placental weight correlated negatively with litter size (both P < 0.01). The relationship between viable litter size and foetal weight differed between sexes (interaction P = 0.002), such that foetal weights of males (P = 0.002), but not females (P = 0.233), correlated negatively with litter size. Placental weight decreased with increasing litter size (P < 0.001) and was lower in females than males (P = 0.020). Our results suggest that male foetuses grow as fast as permitted by nutrient supply, whereas the female maintains placental reserve capacity. This strategy reflecting sex-specific gene expression is likely to place the male foetus at greater risk of death in the event of a 'second hit'.


Asunto(s)
Retardo del Crecimiento Fetal , Placenta , Animales , Modelos Animales de Enfermedad , Transferencia de Embrión , Femenino , Peso Fetal , Tamaño de la Camada , Masculino , Ratones , Embarazo
5.
Neurobiol Dis ; 134: 104640, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31639411

RESUMEN

Mutations in the GAP activity toward RAGs 1 (GATOR1) complex genes (DEPDC5, NPRL2 and NPRL3) have been associated with focal epilepsy and focal cortical dysplasia (FCD). GATOR1 functions as an inhibitor of the mTORC1 signalling pathway, indicating that the downstream effects of mTORC1 deregulation underpin the disease. However, the vast majority of putative disease-causing variants have not been functionally assessed for mTORC1 repression activity. Here, we develop a novel in vitro functional assay that enables rapid assessment of GATOR1-gene variants. Surprisingly, of the 17 variants tested, we show that only six showed significantly impaired mTORC1 inhibition. To further investigate variant function in vivo, we generated a conditional Depdc5 mouse which modelled a 'second-hit' mechanism of disease. Generation of Depdc5 null 'clones' in the embryonic brain resulted in mTORC1 hyperactivity and modelled epilepsy and FCD symptoms including large dysmorphic neurons, defective migration and lower seizure thresholds. Using this model, we validated DEPDC5 variant F164del to be loss-of-function. We also show that Q542P is not functionally compromised in vivo, consistent with our in vitro findings. Overall, our data show that mTORC1 deregulation is the central pathological mechanism for GATOR1 variants and also indicates that a significant proportion of putative disease variants are pathologically inert, highlighting the importance of GATOR1 variant functional assessment.


Asunto(s)
Epilepsias Parciales/metabolismo , Epilepsia/metabolismo , Proteínas Activadoras de GTPasa/genética , Malformaciones del Desarrollo Cortical de Grupo I/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Animales , Epilepsias Parciales/genética , Epilepsia/genética , Proteínas Activadoras de GTPasa/metabolismo , Técnicas Genéticas , Células HEK293 , Humanos , Malformaciones del Desarrollo Cortical de Grupo I/genética , Ratones , Ratones Noqueados , Mutación
7.
Neurobiol Dis ; 116: 106-119, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29763708

RESUMEN

PCDH19-Girls Clustering Epilepsy (PCDH19-GCE) is a childhood epileptic encephalopathy characterised by a spectrum of neurodevelopmental problems. PCDH19-GCE is caused by heterozygous loss-of-function mutations in the X-chromosome gene, Protocadherin 19 (PCDH19) encoding a cell-cell adhesion molecule. Intriguingly, hemizygous males are generally unaffected. As PCDH19 is subjected to random X-inactivation, heterozygous females are comprised of a mosaic of cells expressing either the normal or mutant allele, which is thought to drive pathology. Despite being the second most prevalent monogeneic cause of epilepsy, little is known about the role of PCDH19 in brain development. In this study we show that PCDH19 is highly expressed in human neural stem and progenitor cells (NSPCs) and investigate its function in vitro in these cells of both mouse and human origin. Transcriptomic analysis of mouse NSPCs lacking Pcdh19 revealed changes to genes involved in regulation of neuronal differentiation, and we subsequently show that loss of Pcdh19 causes increased NSPC neurogenesis. We reprogramed human fibroblast cells harbouring a pathogenic PCDH19 mutation into human induced pluripotent stem cells (hiPSC) and employed neural differentiation of these to extend our studies into human NSPCs. As in mouse, loss of PCDH19 function caused increased neurogenesis, and furthermore, we show this is associated with a loss of human NSPC polarity. Overall our data suggests a conserved role for PCDH19 in regulating mammalian cortical neurogenesis and has implications for the pathogenesis of PCDH19-GCE. We propose that the difference in timing or "heterochrony" of neuronal cell production originating from PCDH19 wildtype and mutant NSPCs within the same individual may lead to downstream asynchronies and abnormalities in neuronal network formation, which in-part predispose the individual to network dysfunction and epileptic activity.


Asunto(s)
Cadherinas/biosíntesis , Epilepsia/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Animales , Cadherinas/genética , Células Cultivadas , Análisis por Conglomerados , Epilepsia/patología , Femenino , Humanos , Células Madre Pluripotentes Inducidas/patología , Masculino , Ratones , Ratones Noqueados , Células-Madre Neurales/patología , Protocadherinas
8.
PLoS Genet ; 9(3): e1003290, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23505376

RESUMEN

Polyalanine expansions in transcription factors have been associated with eight distinct congenital human diseases. It is thought that in each case the polyalanine expansion causes misfolding of the protein that abrogates protein function. Misfolded proteins form aggregates when expressed in vitro; however, it is less clear whether aggregation is of relevance to these diseases in vivo. To investigate this issue, we used targeted mutagenesis of embryonic stem (ES) cells to generate mice with a polyalanine expansion mutation in Sox3 (Sox3-26ala) that is associated with X-linked Hypopituitarism (XH) in humans. By investigating both ES cells and chimeric mice, we show that endogenous polyalanine expanded SOX3 does not form protein aggregates in vivo but rather is present at dramatically reduced levels within the nucleus of mutant cells. Importantly, the residual mutant protein of chimeric embryos is able to rescue a block in gastrulation but is not sufficient for normal development of the hypothalamus, a region that is functionally compromised in Sox3 null embryos and individuals with XH. Together, these data provide the first definitive example of a disease-relevant PA mutant protein that is both nuclear and functional, thereby manifesting as a partial loss-of-function allele.


Asunto(s)
Enfermedades Genéticas Ligadas al Cromosoma X , Hipopituitarismo/genética , Péptidos , Factores de Transcripción SOXB1/genética , Alelos , Animales , Núcleo Celular , Modelos Animales de Enfermedad , Desarrollo Embrionario/genética , Células Madre Embrionarias/citología , Humanos , Hipopituitarismo/patología , Ratones , Mutagénesis , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Péptidos/genética , Péptidos/metabolismo , Deficiencias en la Proteostasis/genética , Deficiencias en la Proteostasis/metabolismo , Factores de Transcripción SOXB1/metabolismo
9.
Sci Rep ; 14(1): 13466, 2024 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-38866815

RESUMEN

CRISPR-Cas9 technology has facilitated development of strategies that can potentially provide more humane and effective methods to control invasive vertebrate species, such as mice. One promising strategy is X chromosome shredding which aims to bias offspring towards males, resulting in a gradual and unsustainable decline of females. This method has been explored in insects with encouraging results. Here, we investigated this strategy in Mus musculus by targeting repeat DNA sequences on the X chromosome with the aim of inducing sufficient DNA damage to specifically eliminate X chromosome-bearing sperm during gametogenesis. We tested three different guide RNAs (gRNAs) targeting different repeats on the X chromosome, together with three male germline-specific promoters for inducing Cas9 expression at different stages of spermatogenesis. A modest bias towards mature Y-bearing sperm was detected in some transgenic males, although this did not translate into significant male-biasing of offspring. Instead, cleavage of the X chromosome during meiosis typically resulted in a spermatogenic block, manifest as small testes volume, empty tubules, low sperm concentration, and sub/infertility. Our study highlights the importance of controlling the timing of CRISPR-Cas9 activity during mammalian spermatogenesis and the sensitivity of spermatocytes to X chromosome disruption.


Asunto(s)
Sistemas CRISPR-Cas , Espermatogénesis , Cromosoma X , Animales , Masculino , Ratones , Espermatogénesis/genética , Cromosoma X/genética , Femenino , ARN Guía de Sistemas CRISPR-Cas/genética , Espermatozoides/metabolismo , Ratones Transgénicos , Meiosis/genética
10.
Nat Commun ; 15(1): 1210, 2024 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-38331934

RESUMEN

We implicated the X-chromosome THOC2 gene, which encodes the largest subunit of the highly-conserved TREX (Transcription-Export) complex, in a clinically complex neurodevelopmental disorder with intellectual disability as the core phenotype. To study the molecular pathology of this essential eukaryotic gene, we generated a mouse model based on a hypomorphic Thoc2 exon 37-38 deletion variant of a patient with ID, speech delay, hypotonia, and microcephaly. The Thoc2 exon 37-38 deletion male (Thoc2Δ/Y) mice recapitulate the core phenotypes of THOC2 syndrome including smaller size and weight, and significant deficits in spatial learning, working memory and sensorimotor functions. The Thoc2Δ/Y mouse brain development is significantly impacted by compromised THOC2/TREX function resulting in R-loop accumulation, DNA damage and consequent cell death. Overall, we suggest that perturbed R-loop homeostasis, in stem cells and/or differentiated cells in mice and the patient, and DNA damage-associated functional alterations are at the root of THOC2 syndrome.


Asunto(s)
Discapacidad Intelectual , Factores de Transcripción , Humanos , Masculino , Ratones , Animales , Factores de Transcripción/metabolismo , Estructuras R-Loop , Transporte Activo de Núcleo Celular , Discapacidad Intelectual/genética , Daño del ADN , Fenotipo , ARN Mensajero/metabolismo
11.
Methods Mol Biol ; 2495: 203-230, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35696035

RESUMEN

Gene drives are genetic elements that are transmitted to greater than 50% of offspring and have potential for population modification or suppression. While gene drives are known to occur naturally, the recent emergence of CRISPR-Cas9 genome-editing technology has enabled generation of synthetic gene drives in a range of organisms including mosquitos, flies, and yeast. For example, studies in Anopheles mosquitos have demonstrated >95% transmission of CRISPR-engineered gene drive constructs, providing a possible strategy for malaria control. Recently published studies have also indicated that it may be possible to develop gene drive technology in invasive rodents such as mice. Here, we discuss the prospects for gene drive development in mice, including synthetic "homing drive" and X-shredder strategies as well as modifications of the naturally occurring t haplotype. We also provide detailed protocols for generation of gene drive mice through incorporation of plasmid-based transgenes in a targeted and non-targeted manner. Importantly, these protocols can be used for generating transgenic mice for any project that requires insertion of kilobase-scale transgenes such as knock-in of fluorescent reporters, gene swaps, overexpression/ectopic expression studies, and conditional "floxed" alleles.


Asunto(s)
Tecnología de Genética Dirigida , Animales , Sistemas CRISPR-Cas/genética , Edición Génica/métodos , Especies Introducidas , Ratones , Ratones Transgénicos , Transgenes
12.
JCI Insight ; 7(23)2022 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-36173683

RESUMEN

Developmental and epileptic encephalopathies (DEEs) are characterized by pharmaco-resistant seizures with concomitant intellectual disability. Epilepsy of infancy with migrating focal seizures (EIMFS) is one of the most severe of these syndromes. De novo variants in ion channels, including gain-of-function variants in KCNT1, which encodes for sodium activated potassium channel protein KNa1.1, have been found to play a major role in the etiology of EIMFS. Here, we test a potential precision therapeutic approach in KCNT1-associated DEE using a gene-silencing antisense oligonucleotide (ASO) approach. We generated a mouse model carrying the KCNT1 p.P924L pathogenic variant; only the homozygous animals presented with the frequent, debilitating seizures and developmental compromise that are seen in patients. After a single intracerebroventricular bolus injection of a Kcnt1 gapmer ASO in symptomatic mice at postnatal day 40, seizure frequency was significantly reduced, behavioral abnormalities improved, and overall survival was extended compared with mice treated with a control ASO (nonhybridizing sequence). ASO administration at neonatal age was also well tolerated and effective in controlling seizures and extending the life span of treated animals. The data presented here provide proof of concept for ASO-based gene silencing as a promising therapeutic approach in KCNT1-associated epilepsies.


Asunto(s)
Encefalopatías , Ratones , Animales , Convulsiones/genética , Convulsiones/terapia
13.
Sci Transl Med ; 14(634): eabm4869, 2022 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-35235341

RESUMEN

Central conducting lymphatic anomaly (CCLA), characterized by the dysfunction of core collecting lymphatic vessels including the thoracic duct and cisterna chyli, and presenting as chylothorax, pleural effusions, chylous ascites, and lymphedema, is a severe disorder often resulting in fetal or perinatal demise. Although pathogenic variants in RAS/mitogen activated protein kinase (MAPK) signaling pathway components have been documented in some patients with CCLA, the genetic etiology of the disorder remains uncharacterized in most cases. Here, we identified biallelic pathogenic variants in MDFIC, encoding the MyoD family inhibitor domain containing protein, in seven individuals with CCLA from six independent families. Clinical manifestations of affected fetuses and children included nonimmune hydrops fetalis (NIHF), pleural and pericardial effusions, and lymphedema. Generation of a mouse model of human MDFIC truncation variants revealed that homozygous mutant mice died perinatally exhibiting chylothorax. The lymphatic vasculature of homozygous Mdfic mutant mice was profoundly mispatterned and exhibited major defects in lymphatic vessel valve development. Mechanistically, we determined that MDFIC controls collective cell migration, an important early event during the formation of lymphatic vessel valves, by regulating integrin ß1 activation and the interaction between lymphatic endothelial cells and their surrounding extracellular matrix. Our work identifies MDFIC variants underlying human lymphatic disease and reveals a crucial, previously unrecognized role for MDFIC in the lymphatic vasculature. Ultimately, understanding the genetic and mechanistic basis of CCLA will facilitate the development and implementation of new therapeutic approaches to effectively treat this complex disease.


Asunto(s)
Quilotórax , Vasos Linfáticos , Linfedema , Factores Reguladores Miogénicos , Animales , Quilotórax/genética , Quilotórax/metabolismo , Células Endoteliales , Femenino , Humanos , Hidropesía Fetal/genética , Hidropesía Fetal/metabolismo , Vasos Linfáticos/patología , Linfedema/genética , Linfedema/metabolismo , Ratones , Factores Reguladores Miogénicos/genética , Embarazo
14.
BMC Genomics ; 12: 176, 2011 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-21466694

RESUMEN

BACKGROUND: MicroRNAs (miRNAs) are small non-coding RNAs that can exert multilevel inhibition/repression at a post-transcriptional or protein synthesis level during disease or development. Characterisation of miRNAs in adult mammalian brains by deep sequencing has been reported previously. However, to date, no small RNA profiling of the developing brain has been undertaken using this method. We have performed deep sequencing and small RNA analysis of a developing (E15.5) mouse brain. RESULTS: We identified the expression of 294 known miRNAs in the E15.5 developing mouse brain, which were mostly represented by let-7 family and other brain-specific miRNAs such as miR-9 and miR-124. We also discovered 4 putative 22-23 nt miRNAs: mm_br_e15_1181, mm_br_e15_279920, mm_br_e15_96719 and mm_br_e15_294354 each with a 70-76 nt predicted pre-miRNA. We validated the 4 putative miRNAs and further characterised one of them, mm_br_e15_1181, throughout embryogenesis. Mm_br_e15_1181 biogenesis was Dicer1-dependent and was expressed in E3.5 blastocysts and E7 whole embryos. Embryo-wide expression patterns were observed at E9.5 and E11.5 followed by a near complete loss of expression by E13.5, with expression restricted to a specialised layer of cells within the developing and early postnatal brain. Mm_br_e15_1181 was upregulated during neurodifferentiation of P19 teratocarcinoma cells. This novel miRNA has been identified as miR-3099. CONCLUSIONS: We have generated and analysed the first deep sequencing dataset of small RNA sequences of the developing mouse brain. The analysis revealed a novel miRNA, miR-3099, with potential regulatory effects on early embryogenesis, and involvement in neuronal cell differentiation/function in the brain during late embryonic and early neonatal development.


Asunto(s)
Encéfalo/metabolismo , MicroARNs/genética , Animales , Encéfalo/crecimiento & desarrollo , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario/genética , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Ratones , MicroARNs/metabolismo
15.
PLoS One ; 16(11): e0258538, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34739481

RESUMEN

Enhancers are vitally important during embryonic development to control the spatial and temporal expression of genes. Recently, large scale genome projects have identified a vast number of putative developmental regulatory elements. However, the proportion of these that have been functionally assessed is relatively low. While enhancers have traditionally been studied using reporter assays, this approach does not characterise their contribution to endogenous gene expression. We have studied the murine Nestin (Nes) intron 2 enhancer, which is widely used to direct exogenous gene expression within neural progenitor cells in cultured cells and in vivo. We generated CRISPR deletions of the enhancer region in mice and assessed their impact on Nes expression during embryonic development. Loss of the Nes neural enhancer significantly reduced Nes expression in the developing CNS by as much as 82%. By assessing NES protein localization, we also show that this enhancer region contains repressor element(s) that inhibit Nes expression within the vasculature. Previous reports have stated that Nes is an essential gene, and its loss causes embryonic lethality. We also generated 2 independent Nes null lines and show that both develop without any obvious phenotypic effects. Finally, through crossing of null and enhancer deletion mice we provide evidence of trans-chromosomal interaction of the Nes enhancer and promoter.


Asunto(s)
Sistema Nervioso Central/metabolismo , Desarrollo Embrionario/genética , Nestina/genética , Animales , Sistema Nervioso Central/embriología , Elementos de Facilitación Genéticos , Femenino , Regulación del Desarrollo de la Expresión Génica , Intrones/genética , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Embarazo
16.
CRISPR J ; 3(5): 388-397, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33095043

RESUMEN

CRISPR-based synthetic gene drives have the potential to deliver a more effective and humane method of invasive vertebrate pest control than current strategies. Relatively efficient CRISPR gene drive systems have been developed in insects and yeast but not in mammals. Here, we investigated the efficiency of CRISPR-Cas9-based gene drives in Mus musculus by constructing "split drive" systems where gRNA expression occurs on a separate chromosome to Cas9, which is under the control of either a zygotic (CAG) or germline (Vasa) promoter. While both systems generated double-strand breaks at their intended target site in vivo, no homology-directed repair between chromosomes ("homing") was detectable. Our data indicate that robust and specific Cas9 expression during meiosis is a critical requirement for the generation of efficient CRISPR-based synthetic gene drives in rodents.


Asunto(s)
Proteína 9 Asociada a CRISPR/metabolismo , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Tecnología de Genética Dirigida , Genes Sintéticos , Meiosis , Cigoto , Animales , Proteína 9 Asociada a CRISPR/genética , Femenino , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Transgénicos , Modelos Animales , Regiones Promotoras Genéticas , ARN Guía de Kinetoplastida/metabolismo , Reparación del ADN por Recombinación
17.
Genes Brain Behav ; 19(4): e12637, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31909872

RESUMEN

Sotos syndrome is a developmental disorder characterized by a suite of clinical features. In children, the three cardinal features of Sotos syndrome are a characteristic facial appearance, learning disability and overgrowth (height and/or head circumference > 2 SDs above average). These features are also evident in adults with this syndrome. Over 90% of Sotos syndrome patients are haploinsufficient for the gene encoding nuclear receptor-binding Su(var)3-9, Enhancer-of-zesteand Trithorax domain-containing protein 1 (NSD1). NSD1 is a histone methyltransferase that catalyzes the methylation of lysine residue 36 on histone H3. However, although the symptomology of Sotos syndrome is well established, many aspects of NSD1 biology remain unknown. Here, we assessed the expression of Nsd1 within the mouse brain, and showed a predominantly neuronal pattern of expression for this histone-modifying factor. We also generated a mouse strain lacking one allele of Nsd1 and analyzed morphological and behavioral characteristics in these mice, showing behavioral characteristics reminiscent of some of the deficits seen in Sotos syndrome patients.


Asunto(s)
Corteza Cerebral/patología , N-Metiltransferasa de Histona-Lisina/genética , Síndrome de Sotos/genética , Animales , Corteza Cerebral/metabolismo , Femenino , Heterocigoto , N-Metiltransferasa de Histona-Lisina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Síndrome de Sotos/patología
18.
CRISPR J ; 1: 431-439, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-31021242

RESUMEN

The RNA-guided endonuclease CRISPR-Cas system from Streptococcus pyogenes (SpCas9) is widely used for generating genetically modified mice via zygotic microinjection. Although SpCas9 is a potent mutagen, it requires an NGG proto-spacer adjacent motif (PAM) at the target site, restricting sequence targetability. Here, we show that RNA-guided endonucleases that utilize a range of alternative PAM sequences can edit the mouse genome at the neurog3 (Ngn3) locus: SpCas9 VQR (NGAN PAM), SpCas9 VRER (NGCG), AsCas12a (TTTN), SaCas9 (NNGRRT), and SaCas9 KKH (NNNRRT). Additional experiments targeting tyrosinase and frizzled3 with SaCas9 KKH and its parent protein demonstrated that these endonucleases generated mutations in up to 100% of embryos across three loci. Remarkably, in contrast to wild-type SpCas9, these endonucleases frequently generated mutant embryos that retain unmodified alleles in both template-free and HDR-repair experiments. Our findings broaden PAM recognition options for mouse genome editing and identify SaCas9/SaCas9 KKH as useful alternatives when targeting genes with null lethal phenotypes.

19.
Neuron ; 97(1): 59-66.e5, 2018 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-29301106

RESUMEN

X-linked diseases typically exhibit more severe phenotypes in males than females. In contrast, protocadherin 19 (PCDH19) mutations cause epilepsy in heterozygous females but spare hemizygous males. The cellular mechanism responsible for this unique pattern of X-linked inheritance is unknown. We show that PCDH19 contributes to adhesion specificity in a combinatorial manner such that mosaic expression of Pcdh19 in heterozygous female mice leads to striking sorting between cells expressing wild-type (WT) PCDH19 and null PCDH19 in the developing cortex, correlating with altered network activity. Complete deletion of PCDH19 in heterozygous mice abolishes abnormal cell sorting and restores normal network activity. Furthermore, we identify variable cortical malformations in PCDH19 epilepsy patients. Our results highlight the role of PCDH19 in determining cell adhesion affinities during cortical development and the way segregation of WT and null PCDH19 cells is associated with the unique X-linked inheritance of PCDH19 epilepsy.


Asunto(s)
Cadherinas/genética , Movimiento Celular/genética , Corteza Cerebral/anomalías , Epilepsia/genética , Animales , Corteza Cerebral/embriología , Epilepsia/embriología , Femenino , Genes Ligados a X , Humanos , Masculino , Ratones , Células-Madre Neurales/metabolismo , Protocadherinas
20.
Sci Rep ; 7(1): 12618, 2017 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-28974734

RESUMEN

DEPDC5 mutations have recently been shown to cause epilepsy in humans. Evidence from in vitro studies has implicated DEPDC5 as a negative regulator of mTORC1 during amino acid insufficiency as part of the GATOR1 complex. To investigate the role of DEPDC5 in vivo we generated a null mouse model using targeted CRISPR mutagenesis. Depdc5 homozygotes display severe phenotypic defects between 12.5-15.5 dpc, including hypotrophy, anaemia, oedema, and cranial dysmorphology as well as blood and lymphatic vascular defects. mTORC1 hyperactivity was observed in the brain of knockout embryos and in fibroblasts and neurospheres isolated from knockout embryos and cultured in nutrient deprived conditions. Heterozygous mice appeared to be normal and we found no evidence of increased susceptibility to seizures or tumorigenesis. Together, these data support mTORC1 hyperactivation as the likely pathogenic mechanism that underpins DEPDC5 loss of function in humans and highlights the potential utility of mTORC1 inhibitors in the treatment of DEPDC5-associated epilepsy.


Asunto(s)
Epilepsia/genética , Proteínas Activadoras de GTPasa/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Convulsiones/genética , Animales , Encéfalo/fisiopatología , Sistemas CRISPR-Cas/genética , Modelos Animales de Enfermedad , Epilepsia/fisiopatología , Fibroblastos/patología , Regulación de la Expresión Génica , Heterocigoto , Humanos , Ratones , Ratones Noqueados , Complejos Multiproteicos/química , Complejos Multiproteicos/genética , Mutación , Convulsiones/fisiopatología , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA