Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 10(1): 9011, 2020 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-32514154

RESUMEN

Autism spectrum disorder (ASD) is a group of developmental pathologies that impair social communication and cause repetitive behaviors. The suggested roles of noncoding RNAs in pathology led us to perform a comparative analysis of the microRNAs expressed in the serum of human ASD patients. The analysis of a cohort of 45 children with ASD revealed that six microRNAs (miR-19a-3p, miR-361-5p, miR-3613-3p, miR-150-5p, miR-126-3p, and miR-499a-5p) were expressed at low to very low levels compared to those in healthy controls. A similar but less pronounced decrease was registered in the clinically unaffected parents of the sick children and in their siblings but never in any genetically unrelated control. Results consistent with these observations were obtained in the blood, hypothalamus and sperm of two of the established mouse models of ASD: valproic acid-treated animals and Cc2d1a+/- heterozygotes. In both instances, the same characteristic miRNA profile was evidenced in the affected individuals and inherited together with disease symptoms in the progeny of crosses with healthy animals. The consistent association of these genetic regulatory changes with the disease provides a starting point for evaluating the changes in the activity of the target genes and, thus, the underlying mechanism(s). From the applied societal and medical perspectives, once properly confirmed in large cohorts, these observations provide tools for the very early identification of affected children and progenitors.


Asunto(s)
Trastorno del Espectro Autista/sangre , Perfilación de la Expresión Génica , MicroARNs/sangre , Adolescente , Adulto , Animales , Ansiedad/genética , Trastorno del Espectro Autista/diagnóstico , Trastorno del Espectro Autista/genética , Trastorno Autístico/sangre , Trastorno Autístico/inducido químicamente , Trastorno Autístico/genética , Niño , Preescolar , Depresión/genética , Modelos Animales de Enfermedad , Diagnóstico Precoz , Conducta Exploratoria , Femenino , Humanos , Hipotálamo/química , Lactante , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Mutantes Neurológicos , MicroARNs/análisis , MicroARNs/genética , Padres , Proteínas Represoras/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Hermanos , Conducta Social , Espermatozoides/química , Ácido Valproico/toxicidad , Adulto Joven
2.
Cells ; 8(10)2019 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-31623338

RESUMEN

Mature spermatozoa contain a whole repertoire of the various classes of cellular RNAs, both coding and non-coding. It was hypothesized that after fertilization they might impact development, a claim supported by experimental evidence in various systems. Despite the current increasing interest in the transgenerational maintenance of epigenetic traits and their possible determination by RNAs, little remains known about conservation in sperm and across generations and the specificities and mechanisms involved in transgenerational maintenance. We identified two distinct fractions of RNAs in mature mouse sperm, one readily extracted in the aqueous phase of the classical TRIzol procedure and a distinct fraction hybridized with homologous DNA in DNA-RNA complexes recovered from the interface, purified after DNase hydrolysis and analyzed by RNA-seq methodology. This DNA-associated RNA (D RNA) was found to represent as much as half of the cell contents in differentiated sperm, in which a major part of the cytoplasmic material has been discarded. Stable complexes were purified free of proteins and identified as hybrids (R-loops) on the basis of their sensitivity to RNase H hydrolysis. Further analysis by RNA-seq identified transcripts from all the coding and non-coding regions of the genome, thus revealing an extensive wave of transcription, prior to or concomitant with the terminal compaction of the chromatin.


Asunto(s)
ADN/genética , Perfilación de la Expresión Génica/métodos , ARN/genética , Espermatozoides/química , Animales , Cromatina/genética , Epigénesis Genética , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Masculino , Ratones , ARN Largo no Codificante/genética , Análisis de Secuencia de ARN/métodos , Transcripción Genética
4.
Environ Epigenet ; 4(2): dvy006, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29732170

RESUMEN

RNA-based inheritance provides a reasonable hypothesis to explain multigenerational maintenance of the disease in the progeny of either a male or female parent suffering from the metabolic syndrome (obesity and type 2 diabetes) induced by abnormal diet. Although, it is still difficult to formulate a complete rational mechanism, study of inheritance is a most direct way to learn about the epigenetic control of gene expression and we wished to summarised our current approach along this line.

5.
Sci Rep ; 7: 41799, 2017 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-28165496

RESUMEN

Microinjection of small noncoding RNAs in one-cell embryos was reported in several instances to result in transcriptional activation of target genes. To determine the molecular mechanisms involved and to explore whether such epigenetic regulations could play a role in early development, we used a cell culture system as close as possible to the embryonic state. We report efficient cardiac differentiation of embryonic stem (ES) cells induced by small non-coding RNAs with sequences of Cdk9, a key player in cardiomyocyte differentiation. Transfer of oligoribonucleotides representing parts of the Cdk9 mRNA into ES and mouse embryo fibroblast cultures resulted in upregulation of transcription. Dependency on Argonaute proteins and endogenous antisense transcripts indicated that the inducer oligoribonucleotides were processed by the RNAi machinery. Upregulation of Cdk9 expression resulted in increased efficiency of cardiac differentiation suggesting a potential tool for stem cell-based regenerative medicine.


Asunto(s)
Diferenciación Celular/genética , Células Madre Embrionarias/metabolismo , Epigénesis Genética , Miocitos Cardíacos/citología , ARN Pequeño no Traducido/genética , Animales , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Quinasa 9 Dependiente de la Ciclina/genética , Sitios Genéticos , Ratones , ARN sin Sentido , Factor de Transcripción SOX9/genética , Transcripción Genética , Activación Transcripcional
6.
PLoS One ; 11(6): e0156953, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27270731

RESUMEN

Dnmt2/Trdmt1 is a methyltransferase, which has been shown to methylate tRNAs. Deficient mutants were reported to exhibit various, seemingly unrelated, defects in development and RNA-mediated epigenetic heredity. Here we report a role in a distinct developmental regulation effected by a noncoding RNA. We show that Dnmt2-deficiency in mice results in cardiac hypertrophy. Echocardiographic measurements revealed that cardiac function is preserved notwithstanding the increased dimensions of the organ due to cardiomyocyte enlargement. Mechanistically, activation of the P-TEFb complex, a critical step for cardiac growth, results from increased dissociation of the negatively regulating Rn7sk non-coding RNA component in Dnmt2-deficient cells. Our data suggest that Dnmt2 plays an unexpected role for regulation of cardiac growth by modulating activity of the P-TEFb complex.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/fisiología , Corazón/embriología , Organogénesis/genética , ARN Polimerasa II/metabolismo , Transcripción Genética , Animales , Células Cultivadas , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica , Corazón/crecimiento & desarrollo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor B de Elongación Transcripcional Positiva/metabolismo , Transactivadores/fisiología , Transcripción Genética/genética
7.
Semin Cell Dev Biol ; 44: 47-50, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26335266

RESUMEN

Epigenetic variation, a stable alteration in gene expression, occurs at multiple moments during development. Several instances of non-Mendelian transmission to the progenies modes are very intriguing. Studies of the mode of hereditary transmission revealed in a series of such cases a role of noncoding RNA molecules as inducers. While still an enigmatic mechanism, emerging models pinpoint to a more general roles of these variations, initiated as a response to genetic and environmental variation. Here we compare the known modes of transgenerational epigenetic variation in mice and humans.


Asunto(s)
Enfermedad/genética , Epigénesis Genética , Mutación , ARN no Traducido/genética , Animales , Herencia , Humanos
8.
Ann N Y Acad Sci ; 1341: 172-5, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25726734

RESUMEN

In addition to the Mendelian mutations, several instances of heritable phenotypic variation have been reported. We have observed, in mice, a role for sperm RNAs in the induction of such stable phenotypic variation. When experimentally transferred by RNA microinjection into fertilized mouse eggs, the noncoding RNAs homologous in sequence to the target locus are efficient inducers of variation at the transcriptional level. Transmission of the phenotypic variation to progeny is highly efficient and independent of gender. Here, we have summarized these finding and how they relate to other reports of epigenetic variation.


Asunto(s)
Epigénesis Genética/genética , Mutación , ARN/genética , Espermatozoides/metabolismo , Animales , Masculino , Ratones , Modelos Genéticos , Fenotipo , ARN no Traducido/genética
10.
PLoS Genet ; 9(5): e1003498, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23717211

RESUMEN

RNA-mediated transmission of phenotypes is an important way to explain non-Mendelian heredity. We have previously shown that small non-coding RNAs can induce hereditary epigenetic variations in mice and act as the transgenerational signalling molecules. Two prominent examples for these paramutations include the epigenetic modulation of the Kit gene, resulting in altered fur coloration, and the modulation of the Sox9 gene, resulting in an overgrowth phenotype. We now report that expression of the Dnmt2 RNA methyltransferase is required for the establishment and hereditary maintenance of both paramutations. Our data show that the Kit paramutant phenotype was not transmitted to the progeny of Dnmt2(-/-) mice and that the Sox9 paramutation was also not established in Dnmt2(-/-) embryos. Similarly, RNA from Dnmt2-negative Kit heterozygotes did not induce the paramutant phenotype when microinjected into Dnmt2-deficient fertilized eggs and microinjection of the miR-124 microRNA failed to induce the characteristic giant phenotype. In agreement with an RNA-mediated mechanism of inheritance, no change was observed in the DNA methylation profiles of the Kit locus between the wild-type and paramutant mice. RNA bisulfite sequencing confirmed Dnmt2-dependent tRNA methylation in mouse sperm and also indicated Dnmt2-dependent cytosine methylation in Kit RNA in paramutant embryos. Together, these findings uncover a novel function of Dnmt2 in RNA-mediated epigenetic heredity.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN/genética , Epigénesis Genética/genética , ARN Pequeño no Traducido/genética , Animales , Color , Citosina/metabolismo , Femenino , Cabello , Herencia , Secuenciación de Nucleótidos de Alto Rendimiento , Masculino , Ratones , Fenotipo , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Espermatozoides/citología , Espermatozoides/metabolismo
11.
Front Genet ; 2: 95, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22303389

RESUMEN

Part of the heterodimeric P-TEF-b element of the Pol II transcription machinery, the cyclin-dependent kinase 9 plays a critical role in gene expression. Phosphorylation of several residues in the polymerase is required for elongation of transcript. It determines the rates of transcription and thus, plays a critical role in several differentiation pathways, best documented in heart development. The synthesis and activity of the protein are tightly regulated in a coordinated manner by at least three non-coding RNAs. First, its kinase activity is reversibly inhibited by formation of a complex with the 334 nt 7SK RNA, from which it is released under conditions of stress. Then, heart development requires a maximal rate of synthesis during cardiomyocyte differentiation, followed by a decrease in the differentiated state. The latter is insured by microRNA-mediated translational inhibition. In a third mode of RNA control, increased levels of transcription are induced by small non-coding RNA molecules with sequences homologous to the transcript. Designated paramutation, this epigenetic variation, stable during development, and hereditarily transmitted in a non-Mendelian manner over several generations, is thought to be a response to the inactivation of one of the two alleles by an abnormal recombination event such as insertion of a transposon.

12.
Essays Biochem ; 48(1): 101-6, 2010 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-20822489

RESUMEN

Inheritance of epigenetic variations may account for a significant part of heritability in human and in mammalian models. Heritable epigenetic variations were reported in plants under the name 'paramutation' more than 50 years ago. Reports by E. Whitelaw and her colleagues and by our laboratory now describe a variety of situations resulting in epigenetic inheritance in mouse systems. In the three cases that we have analysed, a transcriptional increase is initiated by RNAs related to the locus, either microRNAs or transcript fragments. RNAs carried by the spermatozoon appear as the transgenerational signals responsible for paternal transmission. Extension from mouse models to human heredity, obviously speculative at present, is encouraged by the high load of RNA in human sperm.


Asunto(s)
Epigénesis Genética , ARN/genética , Transducción de Señal , Animales , Humanos , Ratones
13.
Organogenesis ; 6(1): 33-6, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20592863

RESUMEN

Based initially on the observation of inheritance patterns at variance with Mendel's first law, hereditary epigenetic variations were evidenced in the mouse. Modulating the transcription of a locus, they are induced by RNAs with sequence homology to the transcript. RNAs transferred by the gamete, including sperm, to the fertilized egg appeared to be responsible for transgenerational maintenance of the variant phenotypes. Instances of RNA-dependent variations so far analyzed in the mouse-a pathological deviation of heart development and a syndrome of gigantism initiated by hyperproliferation of embryonic stem cells-suggest a general dependence of organogenesis on epigenetic controls of gene expression. "I conclude it is impossible to say we know the limit of variation."-Charles Darwin. One of the most fascinating visions offered to the biologist is to watch the fertilized egg ingeniously unfolding a program to create a novel being. Development takes place by activating networks of gene activation that result in the proper adjustment of cell growth and functional differentiation. How is the whole process started? Thoughts are generally centered on the activation of critical genes at the early stages due to a newly acquired organization of their chromatin structures. Is the embryo induced to start a given program by molecules contributed by the maternal and paternal gametes? While genetic determinants are clearly essential, the epigenetic landscape largely dominates our current way of thinking. In this essay, we will focus on the evidence showing that RNA molecules are present in the gametes and that RNA can modulate the robust genetic program of organ formation in the mouse.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Organogénesis/genética , ARN/genética , Animales , Embrión de Mamíferos/fisiología , Epigénesis Genética , Ratones
14.
Development ; 136(21): 3647-55, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19820183

RESUMEN

The size of the mammalian body is determined by genetic and environmental factors differentially modulating pre- and postnatal growth. We now report a control of growth acting in the mouse from the first cleavages to the postnatal stages. It was evidenced by a hereditary epigenetic modification (paramutation) created by injection of a miR-124 microRNA into fertilized eggs. From the blastocyst to the adult, mouse pups born after microinjection of this miRNA showed a 30% increase in size. At the blastocyst stage, frequent duplication of the inner cell mass resulted in twin pregnancies. A role of sperm RNA as a transgenerational signal was confirmed by the giant phenotype of the progeny of transgenic males expressing miR-124 during spermiogenesis. In E2.5 to E8.5 embryos, increased levels of several transcripts with sequence homology to the microRNA were noted, including those of Sox9, a gene known for its crucial role in the progenitors of several adult tissues. A role in embryonic growth was confirmed by the large size of embryos expressing a Sox9 DNA transgene. Increased expression in the paramutants was not related to a change in miR-124 expression, but to the establishment of a distinct, heritable chromatin structure in the promoter region of Sox9. While the heritability of body size is not readily accounted for by Mendelian genetics, our results suggest the alternate model of RNA-mediated heritable epigenetic modifications.


Asunto(s)
Tamaño Corporal/genética , Epigénesis Genética , Ratones/embriología , MicroARNs/metabolismo , Factor de Transcripción SOX9/metabolismo , Animales , Masculino , Ratones/genética
15.
Dev Cell ; 14(6): 962-9, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18539123

RESUMEN

Epigenetic regulation shapes normal and pathological mammalian development and physiology. Our previous work showed that Kit RNAs injected into fertilized mouse eggs can produce heritable epigenetic defects, or paramutations, with relevant loss-of-function pigmentation phenotypes, which affect adult phenotypes in multiple succeeding generations of mice. Here, we illustrate the relevance of paramutation to pathophysiology by injecting fertilized mouse eggs with RNAs targeting Cdk9, a key regulator of cardiac growth. Microinjecting fragments of either the coding region or the related microRNA miR-1 led to high levels of expression of homologous RNA, resulting in an epigenetic defect, cardiac hypertrophy, whose efficient hereditary transmission correlated with the presence of miR-1 in the sperm nucleus. In this case, paramutation increased rather than decreased expression of Cdk9. These results highlight the diversity of RNA-mediated epigenetic effects and may provide a paradigm for clinical cases of familial diseases whose inheritance is not fully explained in Mendelian terms.


Asunto(s)
Cardiomegalia/genética , Epigénesis Genética , ARN/biosíntesis , Animales , Peso Corporal , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Núcleo Celular/metabolismo , Quinasa 9 Dependiente de la Ciclina/biosíntesis , Quinasa 9 Dependiente de la Ciclina/ultraestructura , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Epidídimo/citología , Masculino , Ratones , Ratones Mutantes , MicroARNs/metabolismo , Microinyecciones , Mutación , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/ultraestructura , Oligonucleótidos/administración & dosificación , Oligonucleótidos/química , Oocitos/metabolismo , Tamaño de los Órganos/genética , Fosforilación , ARN Polimerasa II/metabolismo , Espermatozoides/citología
16.
Curr Opin Genet Dev ; 18(2): 193-6, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18280137

RESUMEN

In contrast with a wide definition of the 'epigenetic variation', including all changes in gene expression that do not result from the alteration of the gene structure, a more restricted class had been defined, initially in plants, under the name 'paramutation'. It corresponds to epigenetic modifications distinct from the regulatory interactions of the cell differentiation pathways, mitotically stable and sexually transmitted with non-Mendelian patterns. This class of epigenetic changes appeared for some time restricted to the plant world, but examples progressively accumulated of epigenetic inheritance in organisms ranging from mice to humans. Occurrence of paramutation in the mouse and possible mechanisms were then established in the paradigmatic case of a mutant phenotype maintained and hereditarily transmitted by wild-type homozygotes. Together with the recent findings in plants indicative of a necessary step of RNA amplification in the reference maize paramutation, the mouse studies point to a new role of RNA, as an inducer and hereditary determinant of epigenetic variation. Given the known presence of a wide range of RNAs in human spermatozoa, as well as a number of unexplained cases of familial disease predisposition and transgenerational maintenance, speculations can be extended to possible roles of RNA-mediated inheritance in human biology and pathology.


Asunto(s)
Epigénesis Genética/genética , Plantas/genética , Animales , Evolución Biológica , Humanos , Ratones , Mutación/genética
17.
PLoS One ; 2(11): e1136, 2007 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-17989773

RESUMEN

Site-specific methylation of cytosines is a key epigenetic mark of vertebrate DNA. While a majority of the methylated residues are in the symmetrical (meC)pG:Gp(meC) configuration, a smaller, but significant fraction is found in the CpA, CpT and CpC asymmetric (non-CpG) dinucleotides. CpG methylation is reproducibly maintained by the activity of the DNA methyltransferase 1 (Dnmt1) on the newly replicated hemimethylated substrates (meC)pG:GpC. On the other hand, establishment and hereditary maintenance of non-CpG methylation patterns have not been analyzed in detail. We previously reported the occurrence of site- and allele-specific methylation at both CpG and non-CpG sites. Here we characterize a hereditary complex of non-CpG methylation, with the transgenerational maintenance of three distinct profiles in a constant ratio, associated with extensive CpG methylation. These observations raised the question of the signal leading to the maintenance of the pattern of asymmetric methylation. The complete non-CpG pattern was reinstated at each generation in spite of the fact that the majority of the sperm genomes contained either none or only one methylated non-CpG site. This observation led us to the hypothesis that the stable CpG patterns might act as blueprints for the maintenance of non-CpG DNA methylation. As predicted, non-CpG DNA methylation profiles were abrogated in a mutant lacking Dnmt1, the enzymes responsible for CpG methylation, but not in mutants defective for either Dnmt3a or Dnmt2.


Asunto(s)
Islas de CpG , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Epigénesis Genética , Alelos , Animales , ADN (Citosina-5-)-Metiltransferasa 1 , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal
18.
Development ; 134(21): 3815-25, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17913791

RESUMEN

In the adult mammalian testis, spermatogenic differentiation starts from a minute population of spermatogonial stem cells (SSCs). SSCs are generated after birth from the fetal gonocytes, themselves derived from the primordial germ cells (PGCs), which are specified during the first days after implantation. Transcriptome profiling of purified preparations evidenced the preferential accumulation in SSCs of transcripts of PU.1 (Sfpi1), a regulatory gene previously identified in hematopoietic progenitors. In situ immunolabeling and RNA determination showed a complex pattern of expression in the adult testis, first in SSCs and early spermatogonia followed by de novo expression in pachytene spermatocytes. Spermatogenesis in a null mutant (PU.1(G/G)) was arrested at the prenatal stage, with reduced numbers of gonocytes owing to a defect in proliferation already noticeable at E12.5. Transcripts of several germinal markers, including vasa (Mvh, Ddx4), Oct4 (Pou5f1), Dazl and Taf4b, were detected, whereas stella (PGC7, Dppa3) was not. Germ cells of PU.1(G/G) newborn testes grafted in nude mice did not initiate the postnatal replicative stage, whereas grafts of their wild-type littermates underwent complete spermatogenesis. During embryonic development, PU.1 transcription was initiated as early as the blastocyst stage, with a generalized expression at E6.5 in the embryonic ectoderm. PU.1 therefore appears to play a determinant role in at least two distinct lineages and, given its wide range of expression, possibly in other stem cells.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Células Germinativas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Células Madre/metabolismo , Transactivadores/metabolismo , Envejecimiento/fisiología , Animales , Biomarcadores , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular , Femenino , Células Germinativas/citología , Hematopoyesis , Humanos , Masculino , Ratones , Proteínas Proto-Oncogénicas/genética , Testículo/embriología , Testículo/metabolismo , Factores de Tiempo , Transactivadores/genética , Transcripción Genética/genética
19.
J Soc Biol ; 201(4): 397-9, 2007.
Artículo en Francés | MEDLINE | ID: mdl-18533100

RESUMEN

By contrast with a wide definition of the 'epigenetic variation', including all changes in gene expression that do not result from alteration of the gene structure, a more restricted class had been defined, initially in plants, under the name 'paramutation'. It corresponds to epigenetic modifications distinct from the regulatory interactions of the cell differentiation pathways, mitotically stable and sexually transmitted with non-Mendelian patterns. This class of epigenetic changes appeared for some time restricted to the plant world, but examples progressively accumulated of epigenetic inheritance in organisms ranging from mice to humans. Occurrence of paramutation in the mouse and possible mechanisms were then established in the paradigmatic case of a mutant phenotype maintained and hereditarily transmitted by wild type homozygotes. Together with recent findings in plants indicative of a necessary step of RNA amplification in the reference maize paramutation, the mouse studies point to a new role of RNA, as an inducer and hereditary determinant of epigenetic variation. Given the known presence of a wide range of RNAs in human spermatozoa, as well as a number of unexplained cases of familial disease predisposition and transgenerational maintenance, speculations can be extended to possible roles of RNA-mediated inheritance in human biology and pathology.


Asunto(s)
Epigénesis Genética/genética , Patrón de Herencia/genética , Ratones/genética , ARN , Animales , Regulación de la Expresión Génica de las Plantas , Enfermedades Genéticas Congénitas/genética , Genotipo , Humanos , Masculino , MicroARNs , Modelos Anatómicos , Mutagénesis Insercional , Plantas/genética , Proteínas Proto-Oncogénicas c-kit/genética , ARN de Planta , Especificidad de la Especie , Espermatozoides/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...