Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 625(7996): 788-796, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38029793

RESUMEN

The expansion of the neocortex, a hallmark of mammalian evolution1,2, was accompanied by an increase in cerebellar neuron numbers3. However, little is known about the evolution of the cellular programmes underlying the development of the cerebellum in mammals. In this study we generated single-nucleus RNA-sequencing data for around 400,000 cells to trace the development of the cerebellum from early neurogenesis to adulthood in human, mouse and the marsupial opossum. We established a consensus classification of the cellular diversity in the developing mammalian cerebellum and validated it by spatial mapping in the fetal human cerebellum. Our cross-species analyses revealed largely conserved developmental dynamics of cell-type generation, except for Purkinje cells, for which we observed an expansion of early-born subtypes in the human lineage. Global transcriptome profiles, conserved cell-state markers and gene-expression trajectories across neuronal differentiation show that cerebellar cell-type-defining programmes have been overall preserved for at least 160 million years. However, we also identified many orthologous genes that gained or lost expression in cerebellar neural cell types in one of the species or evolved new expression trajectories during neuronal differentiation, indicating widespread gene repurposing at the cell-type level. In sum, our study unveils shared and lineage-specific gene-expression programmes governing the development of cerebellar cells and expands our understanding of mammalian brain evolution.


Asunto(s)
Cerebelo , Evolución Molecular , Mamíferos , Neurogénesis , Animales , Humanos , Ratones , Linaje de la Célula/genética , Cerebelo/citología , Cerebelo/embriología , Cerebelo/crecimiento & desarrollo , Feto/citología , Feto/embriología , Regulación del Desarrollo de la Expresión Génica , Neurogénesis/genética , Neuronas/citología , Neuronas/metabolismo , Zarigüeyas/embriología , Zarigüeyas/crecimiento & desarrollo , Células de Purkinje/citología , Células de Purkinje/metabolismo , Análisis de Expresión Génica de una Sola Célula , Especificidad de la Especie , Transcriptoma , Mamíferos/embriología , Mamíferos/crecimiento & desarrollo
2.
Nature ; 613(7943): 308-316, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36544022

RESUMEN

The testis produces gametes through spermatogenesis and evolves rapidly at both the morphological and molecular level in mammals1-6, probably owing to the evolutionary pressure on males to be reproductively successful7. However, the molecular evolution of individual spermatogenic cell types across mammals remains largely uncharacterized. Here we report evolutionary analyses of single-nucleus transcriptome data for testes from 11 species that cover the three main mammalian lineages (eutherians, marsupials and monotremes) and birds (the evolutionary outgroup), and include seven primates. We find that the rapid evolution of the testis was driven by accelerated fixation rates of gene expression changes, amino acid substitutions and new genes in late spermatogenic stages, probably facilitated by reduced pleiotropic constraints, haploid selection and transcriptionally permissive chromatin. We identify temporal expression changes of individual genes across species and conserved expression programs controlling ancestral spermatogenic processes. Genes predominantly expressed in spermatogonia (germ cells fuelling spermatogenesis) and Sertoli (somatic support) cells accumulated on X chromosomes during evolution, presumably owing to male-beneficial selective forces. Further work identified transcriptomal differences between X- and Y-bearing spermatids and uncovered that meiotic sex-chromosome inactivation (MSCI) also occurs in monotremes and hence is common to mammalian sex-chromosome systems. Thus, the mechanism of meiotic silencing of unsynapsed chromatin, which underlies MSCI, is an ancestral mammalian feature. Our study illuminates the molecular evolution of spermatogenesis and associated selective forces, and provides a resource for investigating the biology of the testis across mammals.


Asunto(s)
Evolución Molecular , Mamíferos , Espermatogénesis , Testículo , Animales , Masculino , Cromatina/genética , Mamíferos/genética , Meiosis/genética , Espermatogénesis/genética , Testículo/citología , Transcriptoma , Análisis de la Célula Individual , Aves/genética , Primates/genética , Regulación de la Expresión Génica , Espermatogonias/citología , Células de Sertoli/citología , Cromosoma X/genética , Cromosoma Y/genética , Compensación de Dosificación (Genética) , Silenciador del Gen
3.
Nature ; 592(7856): 756-762, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33408411

RESUMEN

Egg-laying mammals (monotremes) are the only extant mammalian outgroup to therians (marsupial and eutherian animals) and provide key insights into mammalian evolution1,2. Here we generate and analyse reference genomes of the platypus (Ornithorhynchus anatinus) and echidna (Tachyglossus aculeatus), which represent the only two extant monotreme lineages. The nearly complete platypus genome assembly has anchored almost the entire genome onto chromosomes, markedly improving the genome continuity and gene annotation. Together with our echidna sequence, the genomes of the two species allow us to detect the ancestral and lineage-specific genomic changes that shape both monotreme and mammalian evolution. We provide evidence that the monotreme sex chromosome complex originated from an ancestral chromosome ring configuration. The formation of such a unique chromosome complex may have been facilitated by the unusually extensive interactions between the multi-X and multi-Y chromosomes that are shared by the autosomal homologues in humans. Further comparative genomic analyses unravel marked differences between monotremes and therians in haptoglobin genes, lactation genes and chemosensory receptor genes for smell and taste that underlie the ecological adaptation of monotremes.


Asunto(s)
Evolución Biológica , Genoma , Ornitorrinco/genética , Tachyglossidae/genética , Animales , Femenino , Masculino , Mamíferos/genética , Filogenia , Cromosomas Sexuales/genética
4.
Nature ; 588(7839): 642-647, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33177713

RESUMEN

Gene-expression programs define shared and species-specific phenotypes, but their evolution remains largely uncharacterized beyond the transcriptome layer1. Here we report an analysis of the co-evolution of translatomes and transcriptomes using ribosome-profiling and matched RNA-sequencing data for three organs (brain, liver and testis) in five mammals (human, macaque, mouse, opossum and platypus) and a bird (chicken). Our within-species analyses reveal that translational regulation is widespread in the different organs, in particular across the spermatogenic cell types of the testis. The between-species divergence in gene expression is around 20% lower at the translatome layer than at the transcriptome layer owing to extensive buffering between the expression layers, which especially preserved old, essential and housekeeping genes. Translational upregulation specifically counterbalanced global dosage reductions during the evolution of sex chromosomes and the effects of meiotic sex-chromosome inactivation during spermatogenesis. Despite the overall prevalence of buffering, some genes evolved faster at the translatome layer-potentially indicating adaptive changes in expression; testis tissue shows the highest fraction of such genes. Further analyses incorporating mass spectrometry proteomics data establish that the co-evolution of transcriptomes and translatomes is reflected at the proteome layer. Together, our work uncovers co-evolutionary patterns and associated selective forces across the expression layers, and provides a resource for understanding their interplay in mammalian organs.


Asunto(s)
Evolución Molecular , Mamíferos/genética , Biosíntesis de Proteínas , Transcriptoma/genética , Animales , Encéfalo/metabolismo , Pollos/genética , Femenino , Genes Ligados a X/genética , Humanos , Hígado/metabolismo , Macaca/genética , Masculino , Ratones , Zarigüeyas/genética , Especificidad de Órganos/genética , Ornitorrinco/genética , Biosíntesis de Proteínas/genética , RNA-Seq , Ribosomas/metabolismo , Cromosomas Sexuales/genética , Especificidad de la Especie , Espermatogénesis/genética , Testículo/metabolismo , Regulación hacia Arriba
5.
Nature ; 571(7766): 510-514, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31243368

RESUMEN

Although many long noncoding RNAs (lncRNAs) have been identified in human and other mammalian genomes, there has been limited systematic functional characterization of these elements. In particular, the contribution of lncRNAs to organ development remains largely unexplored. Here we analyse the expression patterns of lncRNAs across developmental time points in seven major organs, from early organogenesis to adulthood, in seven species (human, rhesus macaque, mouse, rat, rabbit, opossum and chicken). Our analyses identified approximately 15,000 to 35,000 candidate lncRNAs in each species, most of which show species specificity. We characterized the expression patterns of lncRNAs across developmental stages, and found many with dynamic expression patterns across time that show signatures of enrichment for functionality. During development, there is a transition from broadly expressed and conserved lncRNAs towards an increasing number of lineage- and organ-specific lncRNAs. Our study provides a resource of candidate lncRNAs and their patterns of expression and evolutionary conservation across mammalian organ development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Especificidad de Órganos/genética , Organogénesis/genética , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Especificidad de la Especie , Animales , Atlas como Asunto , Pollos/genética , Evolución Molecular , Femenino , Humanos , Macaca mulatta/genética , Masculino , Ratones , Zarigüeyas/genética , Proteínas/genética , ARN Largo no Codificante/análisis , Conejos , Ratas
6.
Nature ; 571(7766): 505-509, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31243369

RESUMEN

The evolution of gene expression in mammalian organ development remains largely uncharacterized. Here we report the transcriptomes of seven organs (cerebrum, cerebellum, heart, kidney, liver, ovary and testis) across developmental time points from early organogenesis to adulthood for human, rhesus macaque, mouse, rat, rabbit, opossum and chicken. Comparisons of gene expression patterns identified correspondences of developmental stages across species, and differences in the timing of key events during the development of the gonads. We found that the breadth of gene expression and the extent of purifying selection gradually decrease during development, whereas the amount of positive selection and expression of new genes increase. We identified differences in the temporal trajectories of expression of individual genes across species, with brain tissues showing the smallest percentage of trajectory changes, and the liver and testis showing the largest. Our work provides a resource of developmental transcriptomes of seven organs across seven species, and comparative analyses that characterize the development and evolution of mammalian organs.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Organogénesis/genética , Transcriptoma/genética , Animales , Evolución Biológica , Pollos/genética , Femenino , Humanos , Macaca mulatta/genética , Masculino , Ratones , Zarigüeyas/genética , Conejos , Ratas
7.
Proc Natl Acad Sci U S A ; 117(42): 26307-26317, 2020 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-33020272

RESUMEN

Reptiles exhibit a spectacular diversity of skin colors and patterns brought about by the interactions among three chromatophore types: black melanophores with melanin-packed melanosomes, red and yellow xanthophores with pteridine- and/or carotenoid-containing vesicles, and iridophores filled with light-reflecting platelets generating structural colors. Whereas the melanosome, the only color-producing endosome in mammals and birds, has been documented as a lysosome-related organelle, the maturation paths of xanthosomes and iridosomes are unknown. Here, we first use 10x Genomics linked-reads and optical mapping to assemble and annotate a nearly chromosome-quality genome of the corn snake Pantherophis guttatus The assembly is 1.71 Gb long, with an N50 of 16.8 Mb and L50 of 24. Second, we perform mapping-by-sequencing analyses and identify a 3.9-Mb genomic interval where the lavender variant resides. The lavender color morph in corn snakes is characterized by gray, rather than red, blotches on a pink, instead of orange, background. Third, our sequencing analyses reveal a single nucleotide polymorphism introducing a premature stop codon in the lysosomal trafficking regulator gene (LYST) that shortens the corresponding protein by 603 amino acids and removes evolutionary-conserved domains. Fourth, we use light and transmission electron microscopy comparative analyses of wild type versus lavender corn snakes and show that the color-producing endosomes of all chromatophores are substantially affected in the LYST mutant. Our work provides evidence characterizing xanthosomes in xanthophores and iridosomes in iridophores as lysosome-related organelles.


Asunto(s)
Colubridae/genética , Pigmentación de la Piel/genética , Proteínas de Transporte Vesicular/genética , Animales , Evolución Biológica , Cromatóforos/metabolismo , Mapeo Cromosómico , Color , Colubridae/metabolismo , Genoma , Lisosomas/metabolismo , Melaninas/metabolismo , Melanóforos/metabolismo , Melanosomas/metabolismo , Mutación , Piel/metabolismo , Serpientes/genética , Vertebrados/metabolismo , Proteínas de Transporte Vesicular/metabolismo
8.
Genomics ; 114(4): 110434, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35863675

RESUMEN

Advances in RNA high-throughput sequencing and large-scale functional assays yield new insights into the multifaceted activities of transposed elements (TE) and many other previously undiscovered sequence elements. Currently, no tool for easy access, analysis, quantification, and visualization of alternatively spliced exons across multiple tissues or developmental stages is available. Also, analysis pipelines demand computational skills or hardware requirements, which often are hard to meet by wet-lab scientists. We developed ExoPLOT to enable simplified access to massive RNA high throughput sequencing datasets to facilitate the analysis of alternative splicing across many biological samples. To demonstrate the functonality of ExoPLOT, we analyzed the contributon of exonized TEs to human coding sequences (CDS). mRNA splice variants containing the TE-derived exon were quantified and compared to expression levels of TE-free splice variants. For analysis, we utilized 313 human cerebrum, cerebellum, heart, kidney, liver, ovary, and testis transcriptomes, representing various pre- and postnatal developmental stages. ExoPLOT visualizes the relative expression levels of alternative transcripts, e.g., caused by the insertion of new TE-derived exons, across different developmental stages of and among multiple tissues. This tool also provides a unique link between evolution and function during exonization (gain of a new exon) and exaptation (recruitment/co-optation) of a new exon. As input for analysis, we derived a database of 1151 repeat-masked, exonized TEs, representing all prominent families of transposons in the human genome and the collection of human consensus coding sequences (CCDS). ExoPLOT screened preprocessed RNA high-throughput sequencing datasets from seven human tissues to quantify and visualize the dynamics in RNA splicing for these 1151 TE-derived exons during the entire human organ development. In addition, we successfully mapped and analyzed 993 recently described exonized sequences from the human frontal cortex onto these 313 transcriptome libraries. ExoPLOT's approach to preprocessing RNA deep sequencing datasets facilitates alternative splicing analysis and significantly reduces processing times. In addition, ExoPLOT's design allows studying alternative RNA isoforms other than TE-derived in a customized - coordinate-based manner and is available at http://retrogenomics3.uni-muenster.de:3838/exz-plot-d/.


Asunto(s)
Empalme Alternativo , Elementos Transponibles de ADN , Exones , Humanos , ARN Mensajero/genética , Análisis de Secuencia de ARN
9.
Genome Res ; 27(9): 1461-1474, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28743766

RESUMEN

Gene duplications generate genomic raw material that allows the emergence of novel functions, likely facilitating adaptive evolutionary innovations. However, global assessments of the functional and evolutionary relevance of duplicate genes in mammals were until recently limited by the lack of appropriate comparative data. Here, we report a large-scale study of the expression evolution of DNA-based functional gene duplicates in three major mammalian lineages (placental mammals, marsupials, egg-laying monotremes) and birds, on the basis of RNA sequencing (RNA-seq) data from nine species and eight organs. We observe dynamic changes in tissue expression preference of paralogs with different duplication ages, suggesting differential contribution of paralogs to specific organ functions during vertebrate evolution. Specifically, we show that paralogs that emerged in the common ancestor of bony vertebrates are enriched for genes with brain-specific expression and provide evidence for differential forces underlying the preferential emergence of young testis- and liver-specific expressed genes. Further analyses uncovered that the overall spatial expression profiles of gene families tend to be conserved, with several exceptions of pronounced tissue specificity shifts among lineage-specific gene family expansions. Finally, we trace new lineage-specific genes that may have contributed to the specific biology of mammalian organs, including the little-studied placenta. Overall, our study provides novel and taxonomically broad evidence for the differential contribution of duplicate genes to tissue-specific transcriptomes and for their importance for the phenotypic evolution of vertebrates.


Asunto(s)
Evolución Molecular , Duplicación de Gen/genética , Regulación de la Expresión Génica/genética , Mamíferos/genética , Animales , Genoma/genética , Especificidad de Órganos/genética , Filogenia , Análisis de Secuencia de ARN
10.
Genome Res ; 27(12): 1961-1973, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29079676

RESUMEN

Sexual dimorphism depends on sex-biased gene expression, but the contributions of microRNAs (miRNAs) have not been globally assessed. We therefore produced an extensive small RNA sequencing data set to analyze male and female miRNA expression profiles in mouse, opossum, and chicken. Our analyses uncovered numerous cases of somatic sex-biased miRNA expression, with the largest proportion found in the mouse heart and liver. Sex-biased expression is explained by miRNA-specific regulation, including sex-biased chromatin accessibility at promoters, rather than piggybacking of intronic miRNAs on sex-biased protein-coding genes. In mouse, but not opossum and chicken, sex bias is coordinated across tissues such that autosomal testis-biased miRNAs tend to be somatically male-biased, whereas autosomal ovary-biased miRNAs are female-biased, possibly due to broad hormonal control. In chicken, which has a Z/W sex chromosome system, expression output of genes on the Z Chromosome is expected to be male-biased, since there is no global dosage compensation mechanism that restores expression in ZW females after almost all genes on the W Chromosome decayed. Nevertheless, we found that the dominant liver miRNA, miR-122-5p, is Z-linked but expressed in an unbiased manner, due to the unusual retention of a W-linked copy. Another Z-linked miRNA, the male-biased miR-2954-3p, shows conserved preference for dosage-sensitive genes on the Z Chromosome, based on computational and experimental data from chicken and zebra finch, and acts to equalize male-to-female expression ratios of its targets. Unexpectedly, our findings thus establish miRNA regulation as a novel gene-specific dosage compensation mechanism.


Asunto(s)
Pollos/genética , Compensación de Dosificación (Genética)/genética , MicroARNs/genética , Monodelphis/genética , Caracteres Sexuales , Animales , Conjuntos de Datos como Asunto , Femenino , Pinzones/genética , Perfilación de la Expresión Génica , Masculino , Ratones , MicroARNs/biosíntesis , Proteínas/genética , Secuencias Reguladoras de Ácidos Nucleicos
11.
Genome Res ; 27(12): 1974-1987, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29133310

RESUMEN

Sex chromosomes differentiated from different ancestral autosomes in various vertebrate lineages. Here, we trace the functional evolution of the XY Chromosomes of the green anole lizard (Anolis carolinensis), on the basis of extensive high-throughput genome, transcriptome and histone modification sequencing data and revisit dosage compensation evolution in representative mammals and birds with substantial new expression data. Our analyses show that Anolis sex chromosomes represent an ancient XY system that originated at least ≈160 million years ago in the ancestor of Iguania lizards, shortly after the separation from the snake lineage. The age of this system approximately coincides with the ages of the avian and two mammalian sex chromosomes systems. To compensate for the almost complete Y Chromosome degeneration, X-linked genes have become twofold up-regulated, restoring ancestral expression levels. The highly efficient dosage compensation mechanism of Anolis represents the only vertebrate case identified so far to fully support Ohno's original dosage compensation hypothesis. Further analyses reveal that X up-regulation occurs only in males and is mediated by a male-specific chromatin machinery that leads to global hyperacetylation of histone H4 at lysine 16 specifically on the X Chromosome. The green anole dosage compensation mechanism is highly reminiscent of that of the fruit fly, Drosophila melanogaster Altogether, our work unveils the convergent emergence of a Drosophila-like dosage compensation mechanism in an ancient reptilian sex chromosome system and highlights that the evolutionary pressures imposed by sex chromosome dosage reductions in different amniotes were resolved in fundamentally different ways.


Asunto(s)
Compensación de Dosificación (Genética) , Drosophila/genética , Evolución Molecular , Lagartos/genética , Animales , Epigénesis Genética , Femenino , Genoma , Humanos , Masculino , Procesos de Determinación del Sexo , Transcriptoma , Cromosoma X , Cromosoma Y
12.
Nat Rev Genet ; 15(11): 734-48, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25297727

RESUMEN

Gene expression changes may underlie much of phenotypic evolution. The development of high-throughput RNA sequencing protocols has opened the door to unprecedented large-scale and cross-species transcriptome comparisons by allowing accurate and sensitive assessments of transcript sequences and expression levels. Here, we review the initial wave of the new generation of comparative transcriptomic studies in mammals and vertebrate outgroup species in the context of earlier work. Together with various large-scale genomic and epigenomic data, these studies have unveiled commonalities and differences in the dynamics of gene expression evolution for various types of coding and non-coding genes across mammalian lineages, organs, developmental stages, chromosomes and sexes. They have also provided intriguing new clues to the regulatory basis and phenotypic implications of evolutionary gene expression changes.


Asunto(s)
Epigenómica , Evolución Molecular , Genómica , Transcriptoma/genética , Vertebrados/genética , Animales , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Mamíferos , Fenotipo , ARN/química , ARN/genética , ARN no Traducido/química , ARN no Traducido/genética , Análisis de Secuencia de ADN , Especificidad de la Especie
13.
Nature ; 508(7497): 488-93, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-24759410

RESUMEN

Y chromosomes underlie sex determination in mammals, but their repeat-rich nature has hampered sequencing and associated evolutionary studies. Here we trace Y evolution across 15 representative mammals on the basis of high-throughput genome and transcriptome sequencing. We uncover three independent sex chromosome originations in mammals and birds (the outgroup). The original placental and marsupial (therian) Y, containing the sex-determining gene SRY, emerged in the therian ancestor approximately 180 million years ago, in parallel with the first of five monotreme Y chromosomes, carrying the probable sex-determining gene AMH. The avian W chromosome arose approximately 140 million years ago in the bird ancestor. The small Y/W gene repertoires, enriched in regulatory functions, were rapidly defined following stratification (recombination arrest) and erosion events and have remained considerably stable. Despite expression decreases in therians, Y/W genes show notable conservation of proto-sex chromosome expression patterns, although various Y genes evolved testis-specificities through differential regulatory decay. Thus, although some genes evolved novel functions through spatial/temporal expression shifts, most Y genes probably endured, at least initially, because of dosage constraints.


Asunto(s)
Evolución Molecular , Mamíferos/genética , Cromosoma Y/genética , Animales , Aves/genética , Secuencia Conservada/genética , Femenino , Dosificación de Gen/genética , Genes sry/genética , Genómica , Secuenciación de Nucleótidos de Alto Rendimiento , Masculino , Marsupiales/genética , Receptores de Péptidos/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Selección Genética/genética , Cromosomas Sexuales/genética , Análisis Espacio-Temporal , Espermatogénesis/genética , Testículo/metabolismo , Transcriptoma/genética
14.
Nature ; 505(7485): 635-40, 2014 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-24463510

RESUMEN

Only a very small fraction of long noncoding RNAs (lncRNAs) are well characterized. The evolutionary history of lncRNAs can provide insights into their functionality, but the absence of lncRNA annotations in non-model organisms has precluded comparative analyses. Here we present a large-scale evolutionary study of lncRNA repertoires and expression patterns, in 11 tetrapod species. We identify approximately 11,000 primate-specific lncRNAs and 2,500 highly conserved lncRNAs, including approximately 400 genes that are likely to have originated more than 300 million years ago. We find that lncRNAs, in particular ancient ones, are in general actively regulated and may function predominantly in embryonic development. Most lncRNAs evolve rapidly in terms of sequence and expression levels, but tissue specificities are often conserved. We compared expression patterns of homologous lncRNA and protein-coding families across tetrapods to reconstruct an evolutionarily conserved co-expression network. This network suggests potential functions for lncRNAs in fundamental processes such as spermatogenesis and synaptic transmission, but also in more specific mechanisms such as placenta development through microRNA production.


Asunto(s)
Evolución Molecular , ARN Largo no Codificante/genética , Animales , Anuros/genética , Pollos/genética , Secuencia Conservada/genética , Regulación del Desarrollo de la Expresión Génica/genética , Genómica , Humanos , Ratones , MicroARNs/genética , Familia de Multigenes , Primates/genética , Proteínas/genética , Precursores del ARN/genética , Transcriptoma
15.
Genome Res ; 26(3): 301-14, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26728716

RESUMEN

New genes contribute substantially to adaptive evolutionary innovation, but the functional evolution of new mammalian genes has been little explored at a broad scale. Previous work established mRNA-derived gene duplicates, known as retrocopies, as models for the study of new gene origination. Here we combine mammalian transcriptomic and epigenomic data to unveil the processes underlying the evolution of stripped-down retrocopies into complex new genes. We show that although some robustly expressed retrocopies are transcribed from preexisting promoters, most evolved new promoters from scratch or recruited proto-promoters in their genomic vicinity. In particular, many retrocopy promoters emerged from ancestral enhancers (or bivalent regulatory elements) or are located in CpG islands not associated with other genes. We detected 88-280 selectively preserved retrocopies per mammalian species, illustrating that these mechanisms facilitated the birth of many functional retrogenes during mammalian evolution. The regulatory evolution of originally monoexonic retrocopies was frequently accompanied by exon gain, which facilitated co-option of distant promoters and allowed expression of alternative isoforms. While young retrogenes are often initially expressed in the testis, increased regulatory and structural complexities allowed retrogenes to functionally diversify and evolve somatic organ functions, sometimes as complex as those of their parents. Thus, some retrogenes evolved the capacity to temporarily substitute for their parents during the process of male meiotic X inactivation, while others rendered parental functions superfluous, allowing for parental gene loss. Overall, our reconstruction of the "life history" of mammalian retrogenes highlights retroposition as a general model for understanding new gene birth and functional evolution.


Asunto(s)
Evolución Molecular , Mamíferos/genética , Retroelementos , Animales , Biología Computacional/métodos , Epigénesis Genética , Perfilación de la Expresión Génica , Genes , Humanos , Anotación de Secuencia Molecular , Filogenia , Regiones Promotoras Genéticas , Secuencias Reguladoras de Ácidos Nucleicos , Transcriptoma , Vertebrados/genética
16.
Nature ; 478(7369): 343-8, 2011 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-22012392

RESUMEN

Changes in gene expression are thought to underlie many of the phenotypic differences between species. However, large-scale analyses of gene expression evolution were until recently prevented by technological limitations. Here we report the sequencing of polyadenylated RNA from six organs across ten species that represent all major mammalian lineages (placentals, marsupials and monotremes) and birds (the evolutionary outgroup), with the goal of understanding the dynamics of mammalian transcriptome evolution. We show that the rate of gene expression evolution varies among organs, lineages and chromosomes, owing to differences in selective pressures: transcriptome change was slow in nervous tissues and rapid in testes, slower in rodents than in apes and monotremes, and rapid for the X chromosome right after its formation. Although gene expression evolution in mammals was strongly shaped by purifying selection, we identify numerous potentially selectively driven expression switches, which occurred at different rates across lineages and tissues and which probably contributed to the specific organ biology of various mammals.


Asunto(s)
Evolución Molecular , Perfilación de la Expresión Génica , ARN Mensajero/genética , Animales , Humanos , Filogenia , Análisis de Componente Principal , Cromosoma X/genética
17.
Genome Res ; 23(1): 34-45, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23034410

RESUMEN

MicroRNAs (miRNAs) are major post-transcriptional regulators of gene expression, yet their origins and functional evolution in mammals remain little understood due to the lack of appropriate comparative data. Using RNA sequencing, we have generated extensive and comparable miRNA data for five organs in six species that represent all main mammalian lineages and birds (the evolutionary outgroup) with the aim to unravel the evolution of mammalian miRNAs. Our analyses reveal an overall expansion of miRNA repertoires in mammals, with threefold accelerated birth rates of miRNA families in placentals and marsupials, facilitated by the de novo emergence of miRNAs in host gene introns. Generally, our analyses suggest a high rate of miRNA family turnover in mammals with many newly emerged miRNA families being lost soon after their formation. Selectively preserved mammalian miRNA families gradually evolved higher expression levels, as well as altered mature sequences and target gene repertoires, and were apparently mainly recruited to exert regulatory functions in nervous tissues. However, miRNAs that originated on the X chromosome evolved high expression levels and potentially diverse functions during spermatogenesis, including meiosis, through selectively driven duplication-divergence processes. Overall, our study thus provides detailed insights into the birth and evolution of mammalian miRNA genes and the associated selective forces.


Asunto(s)
Evolución Molecular , Expresión Génica/genética , MicroARNs/genética , Animales , Pollos , Humanos , Intrones , Macaca , Ratones , MicroARNs/clasificación , Familia de Multigenes , Zarigüeyas , Filogenia , Ornitorrinco , Cromosoma X/genética
18.
Mol Microbiol ; 92(5): 903-9, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24865634

RESUMEN

On 19 January 2014 Rolf ('Roffe') Bernander passed away unexpectedly. Rolf was a dedicated scientist; his research aimed at unravelling the cell biology of the archaeal domain of life, especially cell cycle-related questions, but he also made important contributions in other areas of microbiology. Rolf had a professor position in the Molecular Evolution programme at Uppsala University, Sweden for about 8 years, and in January 2013 he became chair professor at the Department of Molecular Biosciences, The Wenner-Gren Institute at Stockholm University in Sweden. Rolf was an exceptional colleague and will be deeply missed by his family and friends, and the colleagues and co-workers that he leaves behind in the scientific community. He will be remembered for his endless enthusiasm for science, his analytical mind, and his quirky sense of humour.


Asunto(s)
Archaea/citología , Ciclo Celular/fisiología , Historia del Siglo XX , Historia del Siglo XXI , Suecia
19.
PLoS Biol ; 10(5): e1001328, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22615540

RESUMEN

As a result of sex chromosome differentiation from ancestral autosomes, male mammalian cells only contain one X chromosome. It has long been hypothesized that X-linked gene expression levels have become doubled in males to restore the original transcriptional output, and that the resulting X overexpression in females then drove the evolution of X inactivation (XCI). However, this model has never been directly tested and patterns and mechanisms of dosage compensation across different mammals and birds generally remain little understood. Here we trace the evolution of dosage compensation using extensive transcriptome data from males and females representing all major mammalian lineages and birds. Our analyses suggest that the X has become globally upregulated in marsupials, whereas we do not detect a global upregulation of this chromosome in placental mammals. However, we find that a subset of autosomal genes interacting with X-linked genes have become downregulated in placentals upon the emergence of sex chromosomes. Thus, different driving forces may underlie the evolution of XCI and the highly efficient equilibration of X expression levels between the sexes observed for both of these lineages. In the egg-laying monotremes and birds, which have partially homologous sex chromosome systems, partial upregulation of the X (Z in birds) evolved but is largely restricted to the heterogametic sex, which provides an explanation for the partially sex-biased X (Z) expression and lack of global inactivation mechanisms in these lineages. Our findings suggest that dosage reductions imposed by sex chromosome differentiation events in amniotes were resolved in strikingly different ways.


Asunto(s)
Aves/genética , Compensación de Dosificación (Genética) , Evolución Molecular , Mamíferos/genética , Animales , Secuencia de Bases , Mapeo Cromosómico , Simulación por Computador , Femenino , Duplicación de Gen , Regulación de la Expresión Génica , Genes Ligados a X , Masculino , Análisis de Secuencia de ARN , Cromosomas Sexuales , Testículo/citología , Transcriptoma
20.
Nat Rev Genet ; 10(1): 19-31, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19030023

RESUMEN

Gene copies that stem from the mRNAs of parental source genes have long been viewed as evolutionary dead-ends with little biological relevance. Here we review a range of recent studies that have unveiled a significant number of functional retroposed gene copies in both mammalian and some non-mammalian genomes. These studies have not only revealed previously unknown mechanisms for the emergence of new genes and their functions but have also provided fascinating general insights into molecular and evolutionary processes that have shaped genomes. For example, analyses of chromosomal gene movement patterns via RNA-based gene duplication have shed fresh light on the evolutionary origin and biology of our sex chromosomes.


Asunto(s)
Evolución Molecular , Duplicación de Gen , ARN/genética , Animales , Genes Duplicados , Humanos , Modelos Genéticos , Filogenia , ARN Mensajero/genética , Elementos Reguladores de la Transcripción/genética , Retroelementos/genética , Cromosomas Sexuales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA