Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Sci Rep ; 9(1): 2262, 2019 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-30783180

RESUMO

MicroRNAs hold great promise as biomarkers of disease. However, there are few efficient and robust methods for measuring microRNAs from low input samples. Here, we develop a high-throughput sequencing protocol that efficiently captures small RNAs while minimizing inherent biases associated with library production. The protocol is based on early barcoding such that all downstream manipulations can be performed on a pool of many samples thereby reducing reagent usage and workload. We show that the optimization of adapter concentrations along with the addition of nucleotide modifications and random nucleotides increases the efficiency of small RNA capture. We further show, using unique molecular identifiers, that stochastic capture of low input RNA rather than PCR amplification influences the biased quantitation of intermediately and lowly expressed microRNAs. Our improved method allows the processing of tens to hundreds of samples simultaneously while retaining high efficiency quantitation of microRNAs in low input samples from tissues or bodily fluids.


Assuntos
Sequenciamento de Nucleotídeos em Larga Escala , MicroRNAs , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de RNA , Manejo de Espécimes , Humanos , MicroRNAs/química , MicroRNAs/genética , MicroRNAs/isolamento & purificação , MicroRNAs/metabolismo
2.
Stem Cells ; 34(7): 1985-91, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27066911

RESUMO

The embryonic stem cell cycle (ESCC) and let-7 families of miRNAs function antagonistically in the switch between mouse embryonic stem cell self-renewal and somatic differentiation. Here, we report that the human ESCC miRNA miR-372 and let-7 act antagonistically in germline differentiation from human embryonic stem cells (hESCs) and human induced pluripotent stem cells (iPSCs). hESC and iPSC-derived primordial germ cell-like cells (PGCLCs) expressed high levels of miR-372 and conversely, somatic cells expressed high levels of let-7. Manipulation of miRNA levels by introduction of miRNA mimics or knockdown with miRNA sponges demonstrated that miR-372 promotes whereas let-7 antagonizes PGCLC differentiation. Knockdown of the individual miR-372 targets SMARCC1, MECP2, CDKN1, RBL2, RHOC, and TGFBR2 increased PGCLC production, whereas knockdown of the let-7 targets CMYC and NMYC suppressed PGCLC differentiation. These findings uncover a miR-372/let-7 axis regulating human primordial germ cell (PGC) specification. Stem Cells 2016;34:1985-1991.


Assuntos
Linhagem da Célula , Células Germinativas/citologia , Células Germinativas/metabolismo , MicroRNAs/metabolismo , Transdução de Sinais/genética , Biomarcadores/metabolismo , Humanos , MicroRNAs/genética
3.
EMBO J ; 34(9): 1180-94, 2015 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-25715649

RESUMO

The molecular basis of astrocyte differentiation and maturation is poorly understood. As microRNAs have important roles in cell fate transitions, we set out to study their function during the glial progenitor cell (GPC) to astrocyte transition. Inducible deletion of all canonical microRNAs in GPCs in vitro led to a block in the differentiation to astrocytes. In an unbiased screen, the reintroduction of let-7 and miR-125 families of microRNAs rescued differentiation. Let-7 and miR-125 shared many targets and functioned in parallel to JAK-STAT signaling, a known regulator of astrogliogenesis. While individual knockdown of shared targets did not rescue the differentiation phenotype in microRNA-deficient GPCs, overexpression of these targets in wild-type GPCs blocked differentiation. This finding supports the idea that microRNAs simultaneously suppress multiple mRNAs that inhibit differentiation. MicroRNA-regulated transcripts exhibited concordant changes during in vivo differentiation and were enriched for a gene set upregulated in glioblastomas, consistent with validity of using the in vitro model to study in vivo events. These findings provide insight into the microRNAs and the genes they regulate in this important cell fate transition.


Assuntos
Astrócitos/fisiologia , MicroRNAs/metabolismo , Regiões 3' não Traduzidas , Animais , Astrócitos/citologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Diferenciação Celular/fisiologia , Sobrevivência Celular/genética , Células Cultivadas , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Glioma/genética , Glioma/patologia , Janus Quinases/metabolismo , Camundongos , MicroRNAs/genética , Neuroglia/citologia , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Fatores de Transcrição STAT/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Células-Tronco/fisiologia
4.
Nat Rev Mol Cell Biol ; 15(9): 565-76, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25118717

RESUMO

microRNAs (miRNAs) are important modulators of development. Owing to their ability to simultaneously silence hundreds of target genes, they have key roles in large-scale transcriptomic changes that occur during cell fate transitions. In somatic stem and progenitor cells--such as those involved in myogenesis, haematopoiesis, skin and neural development--miRNA function is carefully regulated to promote and stabilize cell fate choice. miRNAs are integrated within networks that form both positive and negative feedback loops. Their function is regulated at multiple levels, including transcription, biogenesis, stability, availability and/or number of target sites, as well as their cooperation with other miRNAs and RNA-binding proteins. Together, these regulatory mechanisms result in a refined molecular response that enables proper cellular differentiation and function.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células , MicroRNAs/metabolismo , Células-Tronco/metabolismo , Animais , Humanos , Células-Tronco/citologia
5.
Curr Opin Genet Dev ; 23(5): 548-55, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23948105

RESUMO

Cells are constantly changing their state of equilibrium in response to internal and external stimuli. These changes in cell identity are driven by highly coordinated modulation of gene expression. This coordinated regulation is achieved in large part due to changes in the structure and composition of the chromatin, driven by epigenetic modulators. Recent discoveries in cellular and genomic reprogramming have highlighted the importance of chromatin modifications to reach and uphold the fidelity of target cell states. In this review, we focus on the latest work addressing the mechanisms surrounding the epigenetic regulation of various types of reprogramming, including somatic cell nuclear transfer (SCNT), cell fusion and transcription factor-induced and microRNA-induced pluripotency. The studies covered herein showcase the interplay between these epigenetic pathways, and highlight the importance of furthering our understanding of these connections to form a clearer picture of the mechanisms underlying stable cell fate transitions.


Assuntos
Reprogramação Celular/genética , Epigênese Genética , Células-Tronco Pluripotentes Induzidas/citologia , Transdução de Sinais/genética , Diferenciação Celular/genética , Fusão Celular , Linhagem da Célula , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Técnicas de Transferência Nuclear
6.
Hepatology ; 51(5): 1735-43, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20432256

RESUMO

UNLABELLED: MicroRNAs (miRNAs) constitute a new class of regulators of gene expression. Among other actions, miRNAs have been shown to control cell proliferation in development and cancer. However, whether miRNAs regulate hepatocyte proliferation during liver regeneration is unknown. We addressed this question by performing 2/3 partial hepatectomy (2/3 PH) on mice with hepatocyte-specific inactivation of DiGeorge syndrome critical region gene 8 (DGCR8), an essential component of the miRNA processing pathway. Hepatocytes of these mice were miRNA-deficient and exhibited a delay in cell cycle progression involving the G(1) to S phase transition. Examination of livers of wildtype mice after 2/3 PH revealed differential expression of a subset of miRNAs, notably an induction of miR-21 and repression of miR-378. We further discovered that miR-21 directly inhibits Btg2, a cell cycle inhibitor that prevents activation of forkhead box M1 (FoxM1), which is essential for DNA synthesis in hepatocytes after 2/3 PH. In addition, we found that miR-378 directly inhibits ornithine decarboxylase (Odc1), which is known to promote DNA synthesis in hepatocytes after 2/3 PH. CONCLUSION: Our results show that miRNAs are critical regulators of hepatocyte proliferation during liver regeneration. Because these miRNAs and target gene interactions are conserved, our findings may also be relevant to human liver regeneration.


Assuntos
Proliferação de Células/efeitos dos fármacos , Hepatócitos/citologia , Regeneração Hepática/genética , MicroRNAs/fisiologia , Animais , Hepatectomia , Proteínas Imediatamente Precoces/antagonistas & inibidores , Masculino , Camundongos , Inibidores da Ornitina Descarboxilase , Proteínas Supressoras de Tumor/antagonistas & inibidores
7.
Proc Natl Acad Sci U S A ; 101(39): 13985-90, 2004 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-15306687

RESUMO

Embryonal carcinoma (EC) cells have served as a model to study the relationship between cancer and cellular differentiation given their potential to produce tumors and, to varying degrees, participate in embryonic development. Here, nuclear transplantation was used to assess the extent to which the tumorigenic and developmental potential of EC cells is governed by epigenetic as opposed to genetic alterations. Nuclei from three independent mouse EC cell lines (F9, P19, and METT-1) with differing developmental and tumorigenic potentials all were able to direct early embryo development, producing morphologically normal blastocysts that gave rise to nuclear transfer (NT)-derived embryonic stem (ES) cell lines at a high efficiency. However, when tested for tumor or chimera formation, the resulting NT ES cells displayed an identical potential as their respective donor EC cells, in stark contrast to previously reported NT ES cells derived from transfer of untransformed cells. Consistent with this finding, comparative genomic hybridization identified previously undescribed genetic lesions in the EC cell lines. Therefore, nonreprogrammable genetic modifications within EC nuclei define the developmental and tumorigenic potential of resulting NT ES cells. Our findings support the notion that cancer results from the deregulation of stem cells and further suggest that the genetics of ECs will reveal genes involved in stem cell self-renewal and pluripotency.


Assuntos
Transformação Celular Neoplásica/genética , Desenvolvimento Embrionário/genética , Epigênese Genética/genética , Células-Tronco Neoplásicas/fisiologia , Animais , Blastocisto/citologia , Blastocisto/fisiologia , Diferenciação Celular , Linhagem Celular , Células Clonais , Clonagem Molecular , Células-Tronco de Carcinoma Embrionário , Feminino , Cariotipagem , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Neoplasias/embriologia , Neoplasias/genética , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/ultraestrutura , Técnicas de Transferência Nuclear , Hibridização de Ácido Nucleico/genética , Hibridização de Ácido Nucleico/métodos , Teratoma/embriologia , Teratoma/genética , Teratoma/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA