Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Cell ; 171(2): 398-413.e21, 2017 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-28942919

RESUMEN

A fundamental challenge in immunology is to decipher the principles governing immune responses at the whole-organism scale. Here, using a comparative infection model, we observe immune signal propagation within and between organs to obtain a dynamic map of immune processes at the organism level. We uncover two inter-organ mechanisms of protective immunity mediated by soluble and cellular factors. First, analyzing ligand-receptor connectivity across tissues reveals that type I IFNs trigger a whole-body antiviral state, protecting the host within hours after skin vaccination. Second, combining parabiosis, single-cell analyses, and gene knockouts, we uncover a multi-organ web of tissue-resident memory T cells that functionally adapt to their environment to stop viral spread across the organism. These results have implications for manipulating tissue-resident memory T cells through vaccination and open up new lines of inquiry for the analysis of immune responses at the organism level.


Asunto(s)
Memoria Inmunológica , Interferón Tipo I/inmunología , Virus Vaccinia/fisiología , Vaccinia/inmunología , Vaccinia/prevención & control , Vacunas Virales/inmunología , Administración Cutánea , Animales , Femenino , Perfilación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Especificidad de Órganos , Organismos Libres de Patógenos Específicos , Linfocitos T/inmunología , Vacunas Virales/administración & dosificación
2.
Cell ; 162(2): 412-424, 2015 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-26186193

RESUMEN

Induced pluripotency is a promising avenue for disease modeling and therapy, but the molecular principles underlying this process, particularly in human cells, remain poorly understood due to donor-to-donor variability and intercellular heterogeneity. Here, we constructed and characterized a clonal, inducible human reprogramming system that provides a reliable source of cells at any stage of the process. This system enabled integrative transcriptional and epigenomic analysis across the human reprogramming timeline at high resolution. We observed distinct waves of gene network activation, including the ordered re-activation of broad developmental regulators followed by early embryonic patterning genes and culminating in the emergence of a signature reminiscent of pre-implantation stages. Moreover, complementary functional analyses allowed us to identify and validate novel regulators of the reprogramming process. Altogether, this study sheds light on the molecular underpinnings of induced pluripotency in human cells and provides a robust cell platform for further studies. PAPERCLIP.


Asunto(s)
Reprogramación Celular , Células Madre Pluripotentes Inducidas/citología , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Epigénesis Genética , Perfilación de la Expresión Génica , Histona Demetilasas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo
3.
Immunity ; 42(5): 965-76, 2015 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-25992865

RESUMEN

Colonization by Lactobacillus in the female genital tract is thought to be critical for maintaining genital health. However, little is known about how genital microbiota influence host immune function and modulate disease susceptibility. We studied a cohort of asymptomatic young South African women and found that the majority of participants had genital communities with low Lactobacillus abundance and high ecological diversity. High-diversity communities strongly correlated with genital pro-inflammatory cytokine concentrations in both cross-sectional and longitudinal analyses. Transcriptional profiling suggested that genital antigen-presenting cells sense gram-negative bacterial products in situ via Toll-like receptor 4 signaling, contributing to genital inflammation through activation of the NF-κB signaling pathway and recruitment of lymphocytes by chemokine production. Our study proposes a mechanism by which cervicovaginal microbiota impact genital inflammation and thereby might affect a woman's reproductive health, including her risk of acquiring HIV.


Asunto(s)
Interacciones Huésped-Patógeno/inmunología , Lactobacillus/inmunología , Vagina/inmunología , Vagina/microbiología , Adolescente , Adulto , África , Bacterias/genética , Bacterias/inmunología , Biodiversidad , Citocinas/inmunología , Femenino , Humanos , Lactobacillus/genética , ARN Ribosómico 16S/genética , Análisis de Secuencia , Sudáfrica , Adulto Joven
4.
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
5.
Hepatology ; 63(1): 35-48, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26224662

RESUMEN

UNLABELLED: Chronic hepatitis B and D infections are major causes of liver disease and hepatocellular carcinoma worldwide. Efficient therapeutic approaches for cure are absent. Sharing the same envelope proteins, hepatitis B virus and hepatitis delta virus use the sodium/taurocholate cotransporting polypeptide (a bile acid transporter) as a receptor to enter hepatocytes. However, the detailed mechanisms of the viral entry process are still poorly understood. Here, we established a high-throughput infectious cell culture model enabling functional genomics of hepatitis delta virus entry and infection. Using a targeted RNA interference entry screen, we identified glypican 5 as a common host cell entry factor for hepatitis B and delta viruses. CONCLUSION: These findings advance our understanding of virus cell entry and open new avenues for curative therapies. As glypicans have been shown to play a role in the control of cell division and growth regulation, virus-glypican 5 interactions may also play a role in the pathogenesis of virus-induced liver disease and cancer.


Asunto(s)
Glipicanos/fisiología , Virus de la Hepatitis B/patogenicidad , Virus de la Hepatitis Delta/patogenicidad , ARN no Traducido/fisiología , Internalización del Virus , Células Cultivadas , Humanos
6.
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
7.
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
8.
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
9.
Cell Stem Cell ; 28(9): 1625-1640.e6, 2021 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-34004179

RESUMEN

Understanding lineage specification during human pre-implantation development is a gateway to improving assisted reproductive technologies and stem cell research. Here we employ pseudotime analysis of single-cell RNA sequencing (scRNA-seq) data to reconstruct early mouse and human embryo development. Using time-lapse imaging of annotated embryos, we provide an integrated, ordered, and continuous analysis of transcriptomics changes throughout human development. We reveal that human trophectoderm/inner cell mass transcriptomes diverge at the transition from the B2 to the B3 blastocyst stage, just before blastocyst expansion. We explore the dynamics of the fate markers IFI16 and GATA4 and show that they gradually become mutually exclusive upon establishment of epiblast and primitive endoderm fates, respectively. We also provide evidence that NR2F2 marks trophectoderm maturation, initiating from the polar side, and subsequently spreads to all cells after implantation. Our study pinpoints the precise timing of lineage specification events in the human embryo and identifies transcriptomics hallmarks and cell fate markers.


Asunto(s)
Desarrollo Embrionario , Transcriptoma , Animales , Blastocisto , Linaje de la Célula/genética , Desarrollo Embrionario/genética , Estratos Germinativos , Humanos , Ratones , Transcriptoma/genética
11.
Commun Biol ; 1: 41, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30271925

RESUMEN

Despite improvements in the CRISPR molecular toolbox, identifying and purifying properly edited clones remains slow, laborious, and low-yield. Here, we establish a method to enable clonal isolation, selection, and expansion of properly edited cells, using OptoElectroPositioning technology for single-cell manipulation on a nanofluidic device. Briefly, after electroporation of primary T cells with CXCR4-targeting Cas9 ribonucleoproteins, single T cells are isolated on a chip and expanded into colonies. Phenotypic consequences of editing are rapidly assessed on-chip with cell-surface staining for CXCR4. Furthermore, individual colonies are identified based on their specific genotype. Each colony is split and sequentially exported for on-target sequencing and further off-chip clonal expansion of the validated clones. Using this method, single-clone editing efficiencies, including the rate of mono- and bi-allelic indels or precise nucleotide replacements, can be assessed within 10 days from Cas9 ribonucleoprotein introduction in cells.

12.
Nat Commun ; 9(1): 360, 2018 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-29367672

RESUMEN

Induced pluripotent stem cells (iPSCs) have considerably impacted human developmental biology and regenerative medicine, notably because they circumvent the use of cells of embryonic origin and offer the potential to generate patient-specific pluripotent stem cells. However, conventional reprogramming protocols produce developmentally advanced, or primed, human iPSCs (hiPSCs), restricting their use to post-implantation human development modeling. Hence, there is a need for hiPSCs resembling preimplantation naive epiblast. Here, we develop a method to generate naive hiPSCs directly from somatic cells, using OKMS overexpression and specific culture conditions, further enabling parallel generation of their isogenic primed counterparts. We benchmark naive hiPSCs against human preimplantation epiblast and reveal remarkable concordance in their transcriptome, dependency on mitochondrial respiration and X-chromosome status. Collectively, our results are essential for the understanding of pluripotency regulation throughout preimplantation development and generate new opportunities for disease modeling and regenerative medicine.


Asunto(s)
Blastocisto/citología , Células Madre Embrionarias/citología , Estratos Germinativos/citología , Células Madre Pluripotentes Inducidas/citología , Animales , Blastocisto/metabolismo , Células Cultivadas , Reprogramación Celular/genética , Técnicas de Reprogramación Celular , Desarrollo Embrionario/genética , Células Madre Embrionarias/metabolismo , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Estratos Germinativos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Ratones , Transcriptoma
13.
Nat Commun ; 8(1): 1096, 2017 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-29061959

RESUMEN

Gene expression heterogeneity in the pluripotent state of mouse embryonic stem cells (mESCs) has been increasingly well-characterized. In contrast, exit from pluripotency and lineage commitment have not been studied systematically at the single-cell level. Here we measure the gene expression dynamics of retinoic acid driven mESC differentiation from pluripotency to lineage commitment, using an unbiased single-cell transcriptomics approach. We find that the exit from pluripotency marks the start of a lineage transition as well as a transient phase of increased susceptibility to lineage specifying signals. Our study reveals several transcriptional signatures of this phase, including a sharp increase of gene expression variability and sequential expression of two classes of transcriptional regulators. In summary, we provide a comprehensive analysis of the exit from pluripotency and lineage commitment at the single cell level, a potential stepping stone to improved lineage manipulation through timing of differentiation cues.


Asunto(s)
Células Madre Embrionarias/citología , Análisis de la Célula Individual , Transcriptoma , Animales , Diferenciación Celular , Linaje de la Célula , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ratones , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Proteínas/genética , Proteínas/metabolismo
14.
Sci Rep ; 7(1): 14626, 2017 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-29116112

RESUMEN

Creating a cDNA library for deep mRNA sequencing (mRNAseq) is generally done by random priming, creating multiple sequencing fragments along each transcript. A 3'-end-focused library approach cannot detect differential splicing, but has potentially higher throughput at a lower cost, along with the ability to improve quantification by using transcript molecule counting with unique molecular identifiers (UMI) that correct PCR bias. Here, we compare an implementation of such a 3'-digital gene expression (3'-DGE) approach with "conventional" random primed mRNAseq. Given our particular datasets on cultured human cardiomyocyte cell lines, we find that, while conventional mRNAseq detects ~15% more genes and needs ~500,000 fewer reads per sample for equivalent statistical power, the resulting differentially expressed genes, biological conclusions, and gene signatures are highly concordant between two techniques. We also find good quantitative agreement at the level of individual genes between two techniques for both read counts and fold changes between given conditions. We conclude that, for high-throughput applications, the potential cost savings associated with 3'-DGE approach are likely a reasonable tradeoff for modest reduction in sensitivity and inability to observe alternative splicing, and should enable many larger scale studies focusing on not only differential expression analysis, but also quantitative transcriptome profiling.


Asunto(s)
Biblioteca de Genes , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Células Madre Pluripotentes Inducidas/metabolismo , Atrofia Muscular Espinal/genética , Miocitos Cardíacos/metabolismo , ARN Mensajero/genética , Análisis de Secuencia de ARN/métodos , Estudios de Casos y Controles , Células Cultivadas , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/citología , Modelos Estadísticos , Miocitos Cardíacos/citología , ARN Mensajero/análisis
15.
Elife ; 52016 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-27177420

RESUMEN

Certain neuron types fire spontaneously at high rates, an ability that is crucial for their function in brain circuits. The spontaneously active GABAergic neurons of the substantia nigra pars reticulata (SNr), a major output of the basal ganglia, provide tonic inhibition of downstream brain areas. A depolarizing 'leak' current supports this firing pattern, but its molecular basis remains poorly understood. To understand how SNr neurons maintain tonic activity, we used single-cell RNA sequencing to determine the transcriptome of individual mouse SNr neurons. We discovered that SNr neurons express the sodium leak channel, NALCN, and that SNr neurons lacking NALCN have impaired spontaneous firing. In addition, NALCN is involved in the modulation of excitability by changes in glycolysis and by activation of muscarinic acetylcholine receptors. Our findings suggest that disruption of NALCN could impair the basal ganglia circuit, which may underlie the severe motor deficits in humans carrying mutations in NALCN.


Asunto(s)
Potenciales de Acción , Neuronas GABAérgicas/fisiología , Glucólisis , Canales Iónicos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Porción Reticular de la Sustancia Negra/fisiología , Animales , Neuronas GABAérgicas/metabolismo , Perfilación de la Expresión Génica , Proteínas de la Membrana , Ratones , Análisis de Secuencia de ARN , Análisis de la Célula Individual
16.
Cell Rep ; 3(6): 2179-90, 2013 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-23791531

RESUMEN

Understanding the extent of genomic transcription and its functional relevance is a central goal in genomics research. However, detailed genome-wide investigations of transcriptome complexity in major mammalian organs have been scarce. Here, using extensive RNA-seq data, we show that transcription of the genome is substantially more widespread in the testis than in other organs across representative mammals. Furthermore, we reveal that meiotic spermatocytes and especially postmeiotic round spermatids have remarkably diverse transcriptomes, which explains the high transcriptome complexity of the testis as a whole. The widespread transcriptional activity in spermatocytes and spermatids encompasses protein-coding and long noncoding RNA genes but also poorly conserves intergenic sequences, suggesting that it may not be of immediate functional relevance. Rather, our analyses of genome-wide epigenetic data suggest that this prevalent transcription, which most likely promoted the birth of new genes during evolution, is facilitated by an overall permissive chromatin in these germ cells that results from extensive chromatin remodeling.


Asunto(s)
ARN/genética , Testículo/fisiología , Transcripción Genética , Transcriptoma , Animales , Evolución Biológica , Humanos , Masculino , Mamíferos , Espermatocitos/citología , Espermatocitos/fisiología , Testículo/citología
17.
Discov Med ; 12(63): 153-8, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21878192

RESUMEN

The Enterovirus genus encompasses several species and various serotypes, like coxsackievirus-B1 (CV-B1) to CV-B6, and many variants. The role of these viruses, especially CV-B4, in the pathogenesis of type 1 diabetes is strongly suspected. It has been reported that antibodies directed towards the region of amino acids 11-30 of the VP4 capsid protein enhance the infection of human peripheral blood mononuclear cells with CV-B4. In order to predict the inter- and intra-serotype specificity of the region 11-30 of CV-B4 VP4, 362 available protein sequences of CV-B1 to -B6, CV-A9, and swine vesicular disease virus (SVDV) have been aligned and levels of homology have been calculated. Serine residue substitutions in this region of VP4 were observed without predictable subsequent modification of conformation or charge. The amino acids 16-24 region was the most variable. The sequence of amino acids 16-24 of the CV-B4E2 VP4 protein was highly homologous to those of other CV-B4 (64.4%) whereas there was no homology with CV-B3 and B5 and very low levels of homology with CV-B1 and B2 (3.3% and 9.9%, respectively). In conclusion, the bioinformatic analysis suggests that the region 16-24 of the VP4 capsid protein is the feature of the specificity of the target of infection-enhancing antibodies directed towards CV-B.


Asunto(s)
Proteínas de la Cápside/química , Proteínas de la Cápside/metabolismo , Enterovirus Humano B/metabolismo , Secuencia de Aminoácidos , Biología Computacional , Enterovirus Humano B/fisiología , Humanos , Leucocitos Mononucleares/virología , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA