Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38895226

RESUMEN

The directed differentiation of pluripotent stem cells (PSCs) from panels of genetically diverse individuals is emerging as a powerful experimental system for characterizing the impact of natural genetic variation on developing cell types and tissues. Here, we establish new PSC lines and experimental approaches for modeling embryonic development in a genetically diverse, outbred mouse stock (Diversity Outbred mice). We show that a range of inbred and outbred PSC lines can be stably maintained in the primed pluripotent state (epiblast stem cells -- EpiSCs) and establish the contribution of genetic variation to phenotypic differences in gene regulation and directed differentiation. Using pooled in vitro fertilization, we generate and characterize a genetic reference panel of Diversity Outbred PSCs (n = 230). Finally, we demonstrate the feasibility of pooled culture of Diversity Outbred EpiSCs as "cell villages", which can facilitate the differentiation of large numbers of EpiSC lines for forward genetic screens. These data can complement and inform similar efforts within the stem cell biology and human genetics communities to model the impact of natural genetic variation on phenotypic variation and disease-risk.

2.
Elife ; 122024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38669177

RESUMEN

Gene expression is known to be affected by interactions between local genetic variation and DNA accessibility, with the latter organized into three-dimensional chromatin structures. Analyses of these interactions have previously been limited, obscuring their regulatory context, and the extent to which they occur throughout the genome. Here, we undertake a genome-scale analysis of these interactions in a genetically diverse population to systematically identify global genetic-epigenetic interaction, and reveal constraints imposed by chromatin structure. We establish the extent and structure of genotype-by-epigenotype interaction using embryonic stem cells derived from Diversity Outbred mice. This mouse population segregates millions of variants from eight inbred founders, enabling precision genetic mapping with extensive genotypic and phenotypic diversity. With 176 samples profiled for genotype, gene expression, and open chromatin, we used regression modeling to infer genetic-epigenetic interactions on a genome-wide scale. Our results demonstrate that statistical interactions between genetic variants and chromatin accessibility are common throughout the genome. We found that these interactions occur within the local area of the affected gene, and that this locality corresponds to topologically associated domains (TADs). The likelihood of interaction was most strongly defined by the three-dimensional (3D) domain structure rather than linear DNA sequence. We show that stable 3D genome structure is an effective tool to guide searches for regulatory elements and, conversely, that regulatory elements in genetically diverse populations provide a means to infer 3D genome structure. We confirmed this finding with CTCF ChIP-seq that revealed strain-specific binding in the inbred founder mice. In stem cells, open chromatin participating in the most significant regression models demonstrated an enrichment for developmental genes and the TAD-forming CTCF-binding complex, providing an opportunity for statistical inference of shifting TAD boundaries operating during early development. These findings provide evidence that genetic and epigenetic factors operate within the context of 3D chromatin structure.


Asunto(s)
Cromatina , Epigénesis Genética , Genoma , Animales , Ratones , Cromatina/metabolismo , Cromatina/genética , Variación Genética , Células Madre Embrionarias/metabolismo
3.
Cell Genom ; 3(5): 100291, 2023 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-37228752

RESUMEN

Diverse inbred mouse strains are important biomedical research models, yet genome characterization of many strains is fundamentally lacking in comparison with humans. In particular, catalogs of structural variants (SVs) (variants ≥ 50 bp) are incomplete, limiting the discovery of causative alleles for phenotypic variation. Here, we resolve genome-wide SVs in 20 genetically distinct inbred mice with long-read sequencing. We report 413,758 site-specific SVs affecting 13% (356 Mbp) of the mouse reference assembly, including 510 previously unannotated coding variants. We substantially improve the Mus musculus transposable element (TE) callset, and we find that TEs comprise 39% of SVs and account for 75% of altered bases. We further utilize this callset to investigate how TE heterogeneity affects mouse embryonic stem cells and find multiple TE classes that influence chromatin accessibility. Our work provides a comprehensive analysis of SVs found in diverse mouse genomes and illustrates the role of TEs in epigenetic differences.

4.
EMBO J ; 41(2): e109445, 2022 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-34931323

RESUMEN

Genetically diverse pluripotent stem cells display varied, heritable responses to differentiation cues. Here, we harnessed these disparities through derivation of mouse embryonic stem cells from the BXD genetic reference panel, along with C57BL/6J (B6) and DBA/2J (D2) parental strains, to identify loci regulating cell state transitions. Upon transition to formative pluripotency, B6 stem cells quickly dissolved naïve networks adopting gene expression modules indicative of neuroectoderm lineages, whereas D2 retained aspects of naïve pluripotency. Spontaneous formation of embryoid bodies identified divergent differentiation where B6 showed a propensity toward neuroectoderm and D2 toward definitive endoderm. Genetic mapping identified major trans-acting loci co-regulating chromatin accessibility and gene expression in both naïve and formative pluripotency. These loci distally modulated occupancy of pluripotency factors at hundreds of regulatory elements. One trans-acting locus on Chr 12 primarily impacted chromatin accessibility in embryonic stem cells, while in epiblast-like cells, the same locus subsequently influenced expression of genes enriched for neurogenesis, suggesting early chromatin priming. These results demonstrate genetically determined biases in lineage commitment and identify major regulators of the pluripotency epigenome.


Asunto(s)
Diferenciación Celular , Epigenoma , Células Madre Embrionarias de Ratones/metabolismo , Animales , Linaje de la Célula , Ensamble y Desensamble de Cromatina , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Ratones , Ratones Endogámicos DBA , Células Madre Embrionarias de Ratones/citología , Secuencias Reguladoras de Ácidos Nucleicos
5.
Cell Stem Cell ; 27(3): 470-481.e6, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32795399

RESUMEN

Variability among pluripotent stem cell (PSC) lines is a prevailing issue that hampers not only experimental reproducibility but also large-scale applications and personalized cell-based therapy. This variability could result from epigenetic and genetic factors that influence stem cell behavior. Naive culture conditions minimize epigenetic fluctuation, potentially overcoming differences in PSC line differentiation potential. Here we derived PSCs from distinct mouse strains under naive conditions and show that lines from distinct genetic backgrounds have divergent differentiation capacity, confirming a major role for genetics in PSC phenotypic variability. This is explained in part through inconsistent activity of extra-cellular signaling, including the Wnt pathway, which is modulated by specific genetic variants. Overall, this study shows that genetic background plays a dominant role in driving phenotypic variability of PSCs.


Asunto(s)
Células Madre Pluripotentes Inducidas , Células Madre Pluripotentes , Animales , Variación Biológica Poblacional , Diferenciación Celular/genética , Variación Genética , Ratones , Reproducibilidad de los Resultados
6.
Cell Stem Cell ; 27(3): 459-469.e8, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32795400

RESUMEN

Mouse embryonic stem cells (mESCs) cultured in the presence of LIF occupy a ground state with highly active pluripotency-associated transcriptional and epigenetic circuitry. However, ground state pluripotency in some inbred strain backgrounds is unstable in the absence of ERK1/2 and GSK3 inhibition. Using an unbiased genetic approach, we dissect the basis of this divergent response to extracellular cues by profiling gene expression and chromatin accessibility in 170 genetically heterogeneous mESCs. We map thousands of loci affecting chromatin accessibility and/or transcript abundance, including 10 QTL hotspots where genetic variation at a single locus coordinates the regulation of genes throughout the genome. For one hotspot, we identify a single enhancer variant ∼10 kb upstream of Lifr associated with chromatin accessibility and mediating a cascade of molecular events affecting pluripotency. We validate causation through reciprocal allele swaps, demonstrating the functional consequences of noncoding variation in gene regulatory networks that stabilize pluripotent states in vitro.


Asunto(s)
Cromatina , Células Madre Pluripotentes , Animales , Diferenciación Celular , Cromatina/genética , Expresión Génica , Variación Genética , Glucógeno Sintasa Quinasa 3 , Ratones
7.
J Cell Biol ; 218(4): 1148-1163, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30733233

RESUMEN

Chromosome alignment at the equator of the mitotic spindle is a highly conserved step during cell division; however, its importance to genomic stability and cellular fitness is not understood. Normal mammalian somatic cells lacking KIF18A function complete cell division without aligning chromosomes. These alignment-deficient cells display normal chromosome copy numbers in vitro and in vivo, suggesting that chromosome alignment is largely dispensable for maintenance of euploidy. However, we find that loss of chromosome alignment leads to interchromosomal compaction defects during anaphase, abnormal organization of chromosomes into a single nucleus at mitotic exit, and the formation of micronuclei in vitro and in vivo. These defects slow cell proliferation and are associated with impaired postnatal growth and survival in mice. Our studies support a model in which the alignment of mitotic chromosomes promotes proper organization of chromosomes into a single nucleus and continued proliferation by ensuring that chromosomes segregate as a compact mass during anaphase.


Asunto(s)
Anafase , Segregación Cromosómica , Cromosomas Humanos , Huso Acromático/fisiología , Animales , Línea Celular , Proliferación Celular , Células Epiteliales/fisiología , Humanos , Cinesinas/genética , Cinesinas/metabolismo , Ratones Noqueados , Epitelio Pigmentado de la Retina/fisiología , Huso Acromático/genética , Huso Acromático/metabolismo , Factores de Tiempo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
8.
Nat Genet ; 50(11): 1574-1583, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30275530

RESUMEN

We report full-length draft de novo genome assemblies for 16 widely used inbred mouse strains and find extensive strain-specific haplotype variation. We identify and characterize 2,567 regions on the current mouse reference genome exhibiting the greatest sequence diversity. These regions are enriched for genes involved in pathogen defence and immunity and exhibit enrichment of transposable elements and signatures of recent retrotransposition events. Combinations of alleles and genes unique to an individual strain are commonly observed at these loci, reflecting distinct strain phenotypes. We used these genomes to improve the mouse reference genome, resulting in the completion of 10 new gene structures. Also, 62 new coding loci were added to the reference genome annotation. These genomes identified a large, previously unannotated, gene (Efcab3-like) encoding 5,874 amino acids. Mutant Efcab3-like mice display anomalies in multiple brain regions, suggesting a possible role for this gene in the regulation of brain development.


Asunto(s)
Mapeo Cromosómico , Sitios Genéticos , Genoma , Haplotipos , Ratones Endogámicos/genética , Animales , Animales de Laboratorio , Mapeo Cromosómico/veterinaria , Haplotipos/genética , Ratones , Ratones Endogámicos BALB C/genética , Ratones Endogámicos C3H/genética , Ratones Endogámicos C57BL/genética , Ratones Endogámicos CBA/genética , Ratones Endogámicos DBA/genética , Ratones Endogámicos NOD/genética , Ratones Endogámicos/clasificación , Anotación de Secuencia Molecular , Filogenia , Polimorfismo de Nucleótido Simple , Especificidad de la Especie
9.
Genome Res ; 25(7): 948-57, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25917818

RESUMEN

Spontaneously arising mouse mutations have served as the foundation for understanding gene function for more than 100 years. We have used exome sequencing in an effort to identify the causative mutations for 172 distinct, spontaneously arising mouse models of Mendelian disorders, including a broad range of clinically relevant phenotypes. To analyze the resulting data, we developed an analytics pipeline that is optimized for mouse exome data and a variation database that allows for reproducible, user-defined data mining as well as nomination of mutation candidates through knowledge-based integration of sample and variant data. Using these new tools, putative pathogenic mutations were identified for 91 (53%) of the strains in our study. Despite the increased power offered by potentially unlimited pedigrees and controlled breeding, about half of our exome cases remained unsolved. Using a combination of manual analyses of exome alignments and whole-genome sequencing, we provide evidence that a large fraction of unsolved exome cases have underlying structural mutations. This result directly informs efforts to investigate the similar proportion of apparently Mendelian human phenotypes that are recalcitrant to exome sequencing.


Asunto(s)
Exoma , Mutación , Animales , Femenino , Enfermedades Genéticas Congénitas/genética , Ligamiento Genético , Variación Genética , Estudio de Asociación del Genoma Completo , Genómica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento , Masculino , Ratones , Fenotipo , Reproducibilidad de los Resultados
10.
Dev Biol ; 402(2): 253-262, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25824710

RESUMEN

Genome integrity in the developing germ line is strictly required for fecundity. In proliferating somatic cells and in germ cells, there are mitotic checkpoint mechanisms that ensure accurate chromosome segregation and euploidy. There is growing evidence of mitotic cell cycle components that are uniquely required in the germ line to ensure genome integrity. We previously showed that the primary phenotype of germ cell deficient 2 (gcd2) mutant mice is infertility due to germ cell depletion during embryogenesis. Here we show that the underlying mutation is a mis-sense mutation, R308K, in the motor domain of the kinesin-8 family member, KIF18A, a protein that is expressed in a variety of proliferative tissues and is a key regulator of chromosome alignment during mitosis. Despite the conservative nature of the mutation, we show that its functional consequences are equivalent to KIF18A deficiency in HeLa cells. We also show that somatic cells progress through mitosis, despite having chromosome alignment defects, while germ cells with similar chromosome alignment defects undergo mitotic arrest and apoptosis. Our data provide evidence for differential requirements for chromosome alignment in germ and somatic cells and show that Kif18a is one of a growing number of genes that are specifically required for cell cycle progression in proliferating germ cells.


Asunto(s)
Proteínas de Ciclo Celular/genética , Células Germinativas/fisiología , Cinesinas/genética , Mitosis/fisiología , Animales , Apoptosis/fisiología , Western Blotting , Proteínas de Ciclo Celular/metabolismo , Clonación Molecular , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Silenciador del Gen , Vectores Genéticos/genética , Células HeLa , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Inmunohistoquímica , Hibridación in Situ , Etiquetado Corte-Fin in Situ , Cinesinas/metabolismo , Ratones , Mitosis/genética , Mutación Missense/genética , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Nat Protoc ; 9(3): 559-74, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24504480

RESUMEN

Mouse embryonic stem cells (mESCs) are key tools for genetic engineering, development of stem cell-based therapies and basic research on pluripotency and early lineage commitment. However, successful derivation of germline-competent embryonic stem cell lines has, until recently, been limited to a small number of inbred mouse strains. Recently, there have been considerable advances in the field of embryonic stem cell biology, particularly in the area of pluripotency maintenance in the epiblast from which the mESCs are derived. Here we describe a protocol for efficient derivation of germline-competent mESCs from any mouse strain, including strains previously deemed nonpermissive. We provide a protocol that is generally applicable to most inbred strains, as well as a variant for nonpermissive strains. By using this protocol, mESCs can be derived in 3 weeks and fully characterized after an additional 12 weeks, at efficiencies as high as 90% and in any strain background.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Línea Celular/citología , Células Madre Embrionarias/citología , Células Madre Pluripotentes/citología , Animales , Línea Celular/fisiología , Células Madre Embrionarias/fisiología , Femenino , Ratones , Células Madre Pluripotentes/fisiología , Embarazo
12.
PLoS One ; 7(11): e50081, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23209647

RESUMEN

Mouse embryonic stem (ES) cells are derived from the inner cell mass of blastocyst stage embryos and are used primarily for the creation of genetically engineered strains through gene targeting. While some inbred strains of mice are permissive to the derivation of embryonic stem cell lines and are therefore easily engineered, others are nonpermissive or recalcitrant. Genetic engineering of recalcitrant strain backgrounds requires gene targeting in a permissive background followed by extensive backcrossing of the engineered allele into the desired strain background. The inbred mouse strain DBA/2J is a recalcitrant strain that is used as a model of many human diseases, including glaucoma, deafness and schizophrenia. Here, we describe the generation of germ-line competent ES cell lines derived from DBA/2J mice. We also demonstrate the utility of DBA/2J ES cells with the creation of conditional knockout allele for Endothelin-2 (Edn2) directly on the DBA/2J strain background.


Asunto(s)
Técnicas de Cultivo de Célula , Modelos Animales de Enfermedad , Células Madre Embrionarias/citología , Glaucoma/genética , Alelos , Animales , Células Cultivadas , Medios de Cultivo/farmacología , Sordera/genética , Endotelina-2/genética , Femenino , Ingeniería Genética/métodos , Técnicas Genéticas , Genoma , Humanos , Cariotipificación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Microscopía Fluorescente/métodos , Modelos Genéticos , Esquizofrenia/genética
13.
Nature ; 477(7364): 289-94, 2011 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-21921910

RESUMEN

We report genome sequences of 17 inbred strains of laboratory mice and identify almost ten times more variants than previously known. We use these genomes to explore the phylogenetic history of the laboratory mouse and to examine the functional consequences of allele-specific variation on transcript abundance, revealing that at least 12% of transcripts show a significant tissue-specific expression bias. By identifying candidate functional variants at 718 quantitative trait loci we show that the molecular nature of functional variants and their position relative to genes vary according to the effect size of the locus. These sequences provide a starting point for a new era in the functional analysis of a key model organism.


Asunto(s)
Regulación de la Expresión Génica/genética , Variación Genética/genética , Genoma/genética , Ratones Endogámicos/genética , Ratones/genética , Fenotipo , Alelos , Animales , Animales de Laboratorio/genética , Genómica , Ratones/clasificación , Ratones Endogámicos C57BL/genética , Filogenia , Sitios de Carácter Cuantitativo/genética
14.
Mamm Genome ; 22(11-12): 685-91, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21953412

RESUMEN

Ts65Dn is a mouse model of Down syndrome: a syndrome that results from chromosome (Chr) 21 trisomy and is associated with congenital defects, cognitive impairment, and ultimately Alzheimer's disease. Ts65Dn mice have segmental trisomy for distal mouse Chr 16, a region sharing conserved synteny with human Chr 21. As a result, this strain harbors three copies of over half of the human Chr 21 orthologs. The trisomic segment of Chr 16 is present as a translocation chromosome (Mmu17(16)), with breakpoints that have not been defined previously. To molecularly characterize the Chrs 16 and 17 breakpoints on the translocation chromosome in Ts65Dn mice, we used a selective enrichment and high-throughput paired-end sequencing approach. Analysis of paired-end reads flanking the Chr 16, Chr 17 junction on Mmu17(16) and de novo assembly of the reads directly spanning the junction provided the precise locations of the Chrs 16 and 17 breakpoints at 84,351,351 and 9,426,822 bp, respectively. These data provide the basis for low-cost, highly efficient genotyping of Ts65Dn mice. More importantly, these data provide, for the first time, complete characterization of gene dosage in Ts65Dn mice.


Asunto(s)
Modelos Animales de Enfermedad , Síndrome de Down/genética , Translocación Genética , Trisomía , Animales , Secuencia de Bases , Síndrome de Down/patología , Femenino , Dosificación de Gen , Genotipo , Secuenciación de Nucleótidos de Alto Rendimiento , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Polimorfismo de Nucleótido Simple , Alineación de Secuencia , Análisis de Secuencia de ADN
15.
Chromosoma ; 118(6): 723-36, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19639331

RESUMEN

Aneuploidy, which leads to unpaired chromosomal axes during meiosis, is frequently accompanied by infertility. We previously showed, using three mouse models of Down syndrome, that it is an extra chromosome, but not extra gene dose, that is associated with male infertility and virtual absence of post-meiotic gem cells. Here, we test the hypothesis that aneuploid segments are differentially modified and expressed during meiosis, depending on whether they are present as an extra chromosome or not. In all three models examined, the trisomic region lacks a pairing partner, but in one case, spermatocytes have an extra (and unpaired) chromosome, while the two other models involve translocation of the trisomic region rather than an extra chromosome. An extra unpaired chromosome was always modified by phosphorylation of histone H2AX and lacked RNA PolII. But in the case of trisomic regions attached to a paired chromosome, assembly of these protein modifications was affected by the position of a trisomic region relative to a centromere and the physical extent of the unpaired chromatin. Analysis of gene expression in testes revealed that extra copy number alone was not sufficient for meiotic upregulation of genes in the trisomic interval. Additionally and unexpectedly, presence of meiotic gene silencing chromatin modifications was not sufficient for downregulation of genes in unpaired trisomic chromatin. Thus, the meiotic chromatin modifications that are cytologically visible are unlikely to be directly involved in sterility versus fertility of DS models. Finally, the presence of an extra unpaired chromosome, but not the presence of extra (trisomic) genes, caused global deregulation of transcription in spermatocytes. These results reveal mechanisms by which an extra chromosome, but not trisomic gene dose, impact on meiotic progress and infertility.


Asunto(s)
Aneuploidia , Cromatina/genética , Síndrome de Down/genética , Síndrome de Down/patología , Meiosis/genética , Animales , Cromosomas de los Mamíferos/genética , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Silenciador del Gen , Histonas/metabolismo , Hibridación Fluorescente in Situ , Masculino , Ratones , Transporte de Proteínas , ARN Polimerasa II/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espermatozoides/citología , Espermatozoides/metabolismo , Transcripción Genética , Trisomía/genética , Trisomía/patología
16.
Mamm Genome ; 20(7): 424-36, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19629596

RESUMEN

Forward genetics (phenotype-driven approaches) remain the primary source for allelic variants in the mouse. Unfortunately, the gap between observable phenotype and causative genotype limits the widespread use of spontaneous and induced mouse mutants. As alternatives to traditional positional cloning and mutation detection approaches, sequence capture and next-generation sequencing technologies can be used to rapidly sequence subsets of the genome. Application of these technologies to mutation detection efforts in the mouse has the potential to significantly reduce the time and resources required for mutation identification by abrogating the need for high-resolution genetic mapping, long-range PCR, and sequencing of individual PCR amplimers. As proof of principle, we used array-based sequence capture and pyrosequencing to sequence an allelic series from the classically defined Kit locus (approximately 200 kb) from each of five noncomplementing Kit mutants (one known allele and four unknown alleles) and have successfully identified and validated a nonsynonymous coding mutation for each allele. These data represent the first documentation and validation that these new technologies can be used to efficiently discover causative mutations. Importantly, these data also provide a specific methodological foundation for the development of large-scale mutation detection efforts in the laboratory mouse.


Asunto(s)
Análisis Mutacional de ADN/métodos , Ratones/genética , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Alelos , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Datos de Secuencia Molecular , Alineación de Secuencia
17.
Artículo en Inglés | MEDLINE | ID: mdl-18407792

RESUMEN

The organic solute and steroid transporter (OST/Ost) is a unique membrane transport protein heterodimer composed of subunits designated alpha and beta, that transports conjugated steroids and prostaglandin E(2) across the plasma membrane. Ost was first identified in the liver of the cartilaginous fish Leucoraja erinacea, the little skate, and subsequently was found in many other species, including humans and rodents. The present study describes the isolation of a new cell line, LEE-1, derived from an early embryo of L. erinacea, and characterizes the expression of Ost in these cells. The mRNA size and amino acid sequence of Ost-beta in LEE-1 were identical to that previously reported for Ost-beta from skate liver, and the primary structure was identical to that of the spiny dogfish shark (Squalus acanthias) with the exception of a single amino acid. Ost-beta was found both on the plasma membrane and intracellularly in LEE-1 cells, consistent with its localization in other cell types. Interestingly, arachidonic acid, the precursor to eicosanoids, strongly induced Ost-beta expression in LEE-1 cells and a lipid mixture containing arachidonic acid also induced Ost-alpha. Overall, the present study describes the isolation of a novel marine cell line, and shows that this cell line expresses relatively high levels of Ost when cultured in the presence of arachidonic acid. Although the function of this transport protein in embryo-derived cells is unknown, it may play a role in the disposition of eicosanoids or steroid-derived molecules.


Asunto(s)
Ácido Araquidónico/metabolismo , Proteínas de Peces/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Rajidae/metabolismo , Esteroides/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Proteínas de Peces/genética , Proteínas de Transporte de Membrana/genética , Datos de Secuencia Molecular , ARN Mensajero/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Rajidae/embriología , Regulación hacia Arriba
19.
Zebrafish ; 4(4): 261-75, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18284333

RESUMEN

The multidrug resistance-associated protein 3 (MRP3/Mrp3) is a member of the ATP-binding cassette (ABC) protein family of membrane transporters and related proteins that act on a variety of xenobiotic and anionic molecules to transfer these substrates in an ATP-dependent manner. In recent years, useful comparative information regarding evolutionarily conserved structure and transport functions of these proteins has accrued through the use of primitive marine animals such as cartilaginous fish. Until recently, one missing tool in comparative studies with cartilaginous fish was cell culture. We have derived from the embryo of Squalus acanthias, the spiny dogfish shark, the S. acanthias embryo (SAE) mesenchymal stem cell line. This is the first continuously proliferating cell line from a cartilaginous fish. We identified expression of Mrp3 in this cell line, cloned the molecule, and examined molecular and cellular physiological aspects of the protein. Shark Mrp3 is characterized by three membrane-spanning domains and two nucleotide-binding domains. Multiple alignments with other species showed that the shark Mrp3 amino acid sequence was well conserved. The shark sequence was overall 64% identical to human MRP3, 72% identical to chicken Mrp3, and 71% identical to frog and stickleback Mrp3. Highest identity between shark and human amino acid sequence (82%) was seen in the carboxyl-terminal nucleotide-binding domain of the proteins. Cell culture experiments showed that mRNA for the protein was induced as much as 25-fold by peptide growth factors, fetal bovine serum, and lipid nutritional components, with the largest effect mediated by a combination of lipids including unsaturated and saturated fatty acids, cholesterol, and vitamin E.


Asunto(s)
Embrión no Mamífero/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Células Madre Mesenquimatosas/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/biosíntesis , Squalus acanthias/fisiología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Clonación Molecular , Medios de Cultivo , ADN/química , Embrión no Mamífero/citología , Insulina/fisiología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Lípidos/fisiología , Células Madre Mesenquimatosas/citología , Datos de Secuencia Molecular , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/química , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Mutación , Polimorfismo Genético , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Squalus acanthias/embriología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...