Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 589(7842): 462-467, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33328628

RESUMO

Mechanical deformations of DNA such as bending are ubiquitous and have been implicated in diverse cellular functions1. However, the lack of high-throughput tools to measure the mechanical properties of DNA has limited our understanding of how DNA mechanics influence chromatin transactions across the genome. Here we develop 'loop-seq'-a high-throughput assay to measure the propensity for DNA looping-and determine the intrinsic cyclizabilities of 270,806 50-base-pair DNA fragments that span Saccharomyces cerevisiae chromosome V, other genomic regions, and random sequences. We found sequence-encoded regions of unusually low bendability within nucleosome-depleted regions upstream of transcription start sites (TSSs). Low bendability of linker DNA inhibits nucleosome sliding into the linker by the chromatin remodeller INO80, which explains how INO80 can define nucleosome-depleted regions in the absence of other factors2. Chromosome-wide, nucleosomes were characterized by high DNA bendability near dyads and low bendability near linkers. This contrast increases for deeper gene-body nucleosomes but disappears after random substitution of synonymous codons, which suggests that the evolution of codon choice has been influenced by DNA mechanics around gene-body nucleosomes. Furthermore, we show that local DNA mechanics affect transcription through TSS-proximal nucleosomes. Overall, this genome-scale map of DNA mechanics indicates a 'mechanical code' with broad functional implications.


Assuntos
Fenômenos Biomecânicos , DNA Fúngico/química , DNA Fúngico/genética , Genoma Fúngico , Saccharomyces cerevisiae/genética , Montagem e Desmontagem da Cromatina , Códon/genética , DNA Fúngico/metabolismo , Nucleossomos/química , Nucleossomos/genética , Nucleossomos/metabolismo , Maleabilidade , Proteínas de Saccharomyces cerevisiae/metabolismo , Sítio de Iniciação de Transcrição
2.
Nature ; 540(7631): 119-123, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27880763

RESUMO

Cultured pluripotent stem cells are a cornerstone of regenerative medicine owing to their ability to give rise to all cell types of the body. Although pluripotent stem cells can be propagated indefinitely in vitro, pluripotency is paradoxically a transient state in vivo, lasting 2-3 days around the time of blastocyst implantation. The exception to this rule is embryonic diapause, a reversible state of suspended development triggered by unfavourable conditions. Diapause is a physiological reproductive strategy widely employed across the animal kingdom, including in mammals, but its regulation remains poorly understood. Here we report that the partial inhibition of mechanistic target of rapamycin (mTOR), a major nutrient sensor and promoter of growth, induces reversible pausing of mouse blastocyst development and allows their prolonged culture ex vivo. Paused blastocysts remain pluripotent and competent-able to give rise to embryonic stem (ES) cells and live, fertile mice. We show that both naturally diapaused blastocysts in vivo and paused blastocysts ex vivo display pronounced reductions in mTOR activity, translation, histone modifications associated with gene activity and transcription. Pausing can be induced directly in cultured ES cells and sustained for weeks without appreciable cell death or deviations from cell cycle distributions. We show that paused ES cells display a remarkable global suppression of transcription, maintain a gene expression signature of diapaused blastocysts and remain pluripotent. These results uncover a new pluripotent stem cell state corresponding to the epiblast of the diapaused blastocyst and indicate that mTOR regulates developmental timing at the peri-implantation stage. Our findings have implications in the fields of assisted reproduction, regenerative medicine, cancer, metabolic disorders and ageing.


Assuntos
Blastocisto/citologia , Células-Tronco Pluripotentes/citologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Animais , Células Cultivadas , Regulação para Baixo , Desenvolvimento Embrionário , Feminino , Camadas Germinativas/citologia , Histonas/metabolismo , Técnicas In Vitro , Masculino , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Biossíntese de Proteínas , Serina-Treonina Quinases TOR/metabolismo , Transcrição Gênica
3.
Proc Natl Acad Sci U S A ; 110(33): 13250-4, 2013 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-23898166

RESUMO

We report the results of highly sensitive transmission X-ray scattering measurements performed at the Advanced Photon Source, Argonne National Laboratory, on nearly fully dense high-purity amorphous-silicon (a-Si) samples for the purpose of determining their degree of hyperuniformity. A perfectly hyperuniform structure has complete suppression of infinite-wavelength density fluctuations, or, equivalently, the structure factor S(q→0) = 0; the smaller the value of S(0), the higher the degree of hyperuniformity. Annealing was observed to increase the degree of hyperuniformity in a-Si where we found S(0) = 0.0075 (±0.0005), which is significantly below the computationally determined lower bound recently suggested by de Graff and Thorpe [de Graff AMR, Thorpe MF (2010) Acta Crystallogr A 66(Pt 1):22-31] based on studies of continuous random network models, but consistent with the recently proposed nearly hyperuniform network picture of a-Si. Increasing hyperuniformity is correlated with narrowing of the first diffraction peak and extension of the range of oscillations in the pair distribution function.


Assuntos
Modelos Químicos , Silício/química , Microscopia Eletrônica , Difração de Raios X/métodos
4.
Cancer Res ; 81(24): 6131-6141, 2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-34728538

RESUMO

Loss-of-function mutations of JAK1/2 impair cancer cell responsiveness to IFNγ and immunogenicity. Therefore, an understanding of compensatory pathways to activate IFNγ signaling in cancer cells is clinically important for the success of immunotherapy. Here we demonstrate that the transcription factor SOX10 hinders immunogenicity of melanoma cells through the IRF4-IRF1 axis. Genetic and pharmacologic approaches revealed that SOX10 repressed IRF1 transcription via direct induction of a negative regulator, IRF4. The SOX10-IRF4-IRF1 axis regulated PD-L1 expression independently of JAK-STAT pathway activity, and suppression of SOX10 increased the efficacy of combination therapy with an anti-PD-1 antibody and histone deacetylase inhibitor against a clinically relevant melanoma model. Thus, the SOX10-IRF4-IRF1 axis serves as a potential target that can bypass JAK-STAT signaling to immunologically warm up melanoma with a "cold" tumor immune microenvironment. SIGNIFICANCE: This study identifies a novel SOX10/IRF4 pathway that regulates noncanonical induction of IRF1 independent of the JAK-STAT pathway and can be targeted to improve the efficacy of anti-PD-1 therapy in melanoma.


Assuntos
Inibidores de Histona Desacetilases/farmacologia , Inibidores de Checkpoint Imunológico/farmacologia , Fator Regulador 1 de Interferon/metabolismo , Fatores Reguladores de Interferon/metabolismo , Melanoma/tratamento farmacológico , Melanoma/imunologia , Fatores de Transcrição SOXE/metabolismo , Animais , Apoptose , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/imunologia , Antígeno B7-H1/metabolismo , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Proliferação de Células , Quimioterapia Combinada , Regulação Neoplásica da Expressão Gênica , Humanos , Imunoterapia , Fator Regulador 1 de Interferon/genética , Fatores Reguladores de Interferon/genética , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Prognóstico , Fatores de Transcrição SOXE/genética , Taxa de Sobrevida , Células Tumorais Cultivadas
5.
Nat Commun ; 12(1): 4859, 2021 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-34381042

RESUMO

Stem and progenitor cells undergo a global elevation of nascent transcription, or hypertranscription, during key developmental transitions involving rapid cell proliferation. The chromatin remodeler Chd1 mediates hypertranscription in pluripotent cells but its mechanism of action remains poorly understood. Here we report a novel role for Chd1 in protecting genome integrity at promoter regions by preventing DNA double-stranded break (DSB) accumulation in ES cells. Chd1 interacts with several DNA repair factors including Atm, Parp1, Kap1 and Topoisomerase 2ß and its absence leads to an accumulation of DSBs at Chd1-bound Pol II-transcribed genes and rDNA. Genes prone to DNA breaks in Chd1 KO ES cells are longer genes with GC-rich promoters, a more labile nucleosomal structure and roles in chromatin regulation, transcription and signaling. These results reveal a vulnerability of hypertranscribing stem cells to accumulation of endogenous DNA breaks, with important implications for developmental and cancer biology.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Células-Tronco Embrionárias Murinas/metabolismo , Regiões Promotoras Genéticas , Transcrição Gênica , Animais , Cromatina/metabolismo , Quebras de DNA de Cadeia Dupla , Reparo do DNA , DNA Topoisomerases Tipo II/metabolismo , DNA Ribossômico/metabolismo , Proteínas de Ligação a DNA/genética , Camundongos , Proteínas de Ligação a Poli-ADP-Ribose/metabolismo , Transdução de Sinais , Sítio de Iniciação de Transcrição
6.
Cell Rep ; 34(13): 108927, 2021 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-33789109

RESUMO

Understanding the epigenomic evolution and specificity of disease subtypes from complex patient data remains a major biomedical problem. We here present DeCET (decomposition and classification of epigenomic tensors), an integrative computational approach for simultaneously analyzing hierarchical heterogeneous data, to identify robust epigenomic differences among tissue types, differentiation states, and disease subtypes. Applying DeCET to our own data from 21 uterine benign tumor (leiomyoma) patients identifies distinct epigenomic features discriminating normal myometrium and leiomyoma subtypes. Leiomyomas possess preponderant alterations in distal enhancers and long-range histone modifications confined to chromatin contact domains that constrain the evolution of pathological epigenomes. Moreover, we demonstrate the power and advantage of DeCET on multiple publicly available epigenomic datasets representing different cancers and cellular states. Epigenomic features extracted by DeCET can thus help improve our understanding of disease states, cellular development, and differentiation, thereby facilitating future therapeutic, diagnostic, and prognostic strategies.


Assuntos
Epigenoma , Leiomioma/classificação , Leiomioma/genética , Neoplasias Uterinas/classificação , Neoplasias Uterinas/genética , Diferenciação Celular/genética , Cromatina/metabolismo , Análise por Conglomerados , Elementos Facilitadores Genéticos/genética , Epigênese Genética , Matriz Extracelular/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Genes Homeobox , Células HEK293 , Humanos , Leiomioma/patologia , Miométrio/patologia , Motivos de Nucleotídeos/genética , Fatores de Transcrição/metabolismo , Neoplasias Uterinas/patologia
7.
J Invest Dermatol ; 140(1): 164-173.e7, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31580842

RESUMO

The use of microRNAs as biomarkers has been proposed for many diseases, including the diagnosis of melanoma. Although hundreds of microRNAs have been identified as differentially expressed in melanomas as compared to benign melanocytic lesions, a limited consensus has been achieved across studies, constraining the effective use of these potentially useful markers. In this study, we applied a machine learning-based pipeline to a dataset consisting of genetic features, clinical features, and next-generation microRNA sequencing from micro-dissected formalin-fixed paraffin embedded melanomas and their adjacent benign precursor nevi. We identified patient age and tumor cellularity as variables that frequently confound the measured expression of potentially diagnostic microRNAs. By employing the ratios of microRNAs that were either enriched or depleted in melanoma compared to the nevi as a normalization strategy, we developed a model that classified all the available published cohorts with an area under the receiver operating characteristic curve of 0.98. External validation on an independent cohort classified lesions with 81% sensitivity and 88% specificity and was uninfluenced by the tumor content of the sample or patient age.


Assuntos
Biomarcadores Tumorais/genética , Melanócitos/fisiologia , Melanoma/diagnóstico , MicroRNAs/genética , Nevo/diagnóstico , Neoplasias Cutâneas/diagnóstico , Conjuntos de Dados como Assunto , Diagnóstico Diferencial , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Aprendizado de Máquina , Prognóstico , Curva ROC , Sensibilidade e Especificidade , Análise de Sequência de RNA
8.
Pigment Cell Melanoma Res ; 32(4): 500-509, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30548162

RESUMO

MITF and MYC are well-known oncoproteins and members of the basic helix-loop-helix leucine zipper (bHLH-Zip) family of transcription factors (TFs) recognizing hexamer E-box motifs. MITF and MYC not only share the core binding motif, but are also the two most highly expressed bHLH-Zip transcription factors in melanocytes, raising the possibility that they may compete for the same binding sites in select oncogenic targets. Mechanisms determining the distinct and potentially overlapping binding modes of these critical oncoproteins remain uncharacterized. We introduce computational predictive models using local sequence features, including a boosted convolutional decision tree framework, to distinguish MITF versus MYC-MAX binding sites with up to 80% accuracy genomewide. Select E-box locations that can be bound by both MITF and MYC-MAX form a separate class of MITF binding sites characterized by differential sequence content in the flanking region, diminished interaction with SOX10, higher evolutionary conservation, and less tissue-specific chromatin organization.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Genoma Humano , Fator de Transcrição Associado à Microftalmia/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Motivos de Aminoácidos , Área Sob a Curva , Composição de Bases/genética , Sequência de Bases , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Sítios de Ligação , Epigênese Genética , Humanos , Fator de Transcrição Associado à Microftalmia/genética , Ligação Proteica , Proteínas Proto-Oncogênicas c-myc/genética , Curva ROC , Fatores de Transcrição SOXE/metabolismo
9.
Sci Rep ; 7(1): 11943, 2017 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-28931937

RESUMO

Digital holographic cytometry (DHC) permits label-free visualization of adherent cells. Dozens of cellular features can be derived from segmentation of hologram-derived images. However, the accuracy of single cell classification by these features remains limited for most applications, and lack of standardization metrics has hindered independent experimental comparison and validation. Here we identify twenty-six DHC-derived features that provide biologically independent information across a variety of mammalian cell state transitions. When trained on these features, machine-learning algorithms achieve blind single cell classification with up to 95% accuracy. Using classification accuracy to guide platform optimization, we develop methods to standardize holograms for the purpose of kinetic single cell cytometry. Applying our approach to human melanoma cells treated with a panel of cancer therapeutics, we track dynamic changes in cellular behavior and cell state over time. We provide the methods and computational tools for optimizing DHC for kinetic single adherent cell classification.


Assuntos
Técnicas Citológicas/métodos , Holografia/métodos , Melanoma/patologia , Análise de Célula Única/métodos , Algoritmos , Linhagem Celular Tumoral , Humanos , Aprendizado de Máquina
10.
Nat Commun ; 7: 11025, 2016 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-27008915

RESUMO

Methyl-CpG binding protein 2 (MeCP2) is critical for proper brain development and expressed at near-histone levels in neurons, but the mechanism of its genomic localization remains poorly understood. Using high-resolution MeCP2-binding data, we show that DNA sequence features alone can predict binding with 88% accuracy. Integrating MeCP2 binding and DNA methylation in a probabilistic graphical model, we demonstrate that previously reported genome-wide association with methylation is in part due to MeCP2's affinity to GC-rich chromatin, a result replicated using published data. Furthermore, MeCP2 co-localizes with nucleosomes. Finally, MeCP2 binding downstream of promoters correlates with increased expression in Mecp2-deficient neurons.


Assuntos
Cromatina/metabolismo , Metilação de DNA/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteína 2 de Ligação a Metil-CpG/genética , Mucosa Olfatória/metabolismo , Animais , Sequência de Bases , Sítios de Ligação , Imunoprecipitação da Cromatina , Sequência Rica em GC , Proteína 2 de Ligação a Metil-CpG/metabolismo , Camundongos , Neurônios , Nucleossomos/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Análise de Sequência de DNA , Análise de Sequência de RNA
11.
Cell Rep ; 14(10): 2301-12, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26947063

RESUMO

The human naive pluripotent stem cell (PSC) state, corresponding to a pre-implantation stage of development, has been difficult to capture and sustain in vitro. We report that the Hippo pathway effector YAP is nuclearly localized in the inner cell mass of human blastocysts. Overexpression of YAP in human embryonic stem cells (ESCs) and induced PSCs (iPSCs) promotes the generation of naive PSCs. Lysophosphatidic acid (LPA) can partially substitute for YAP to generate transgene-free human naive PSCs. YAP- or LPA-induced naive PSCs have a rapid clonal growth rate, a normal karyotype, the ability to form teratomas, transcriptional similarities to human pre-implantation embryos, reduced heterochromatin levels, and other hallmarks of the naive state. YAP/LPA act in part by suppressing differentiation-inducing effects of GSK3 inhibition. CRISPR/Cas9-generated YAP(-/-) cells have an impaired ability to form colonies in naive but not primed conditions. These results uncover an unexpected role for YAP in the human naive state, with implications for early human embryology.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Fosfoproteínas/metabolismo , Células-Tronco Pluripotentes/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/genética , Benzamidas/farmacologia , Blastocisto/citologia , Blastocisto/metabolismo , Blastocisto/patologia , Sistemas CRISPR-Cas/genética , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Difenilamina/análogos & derivados , Difenilamina/farmacologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/metabolismo , Heterocromatina/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Cariótipo , Lisofosfolipídeos/farmacologia , Masculino , Fosfoproteínas/antagonistas & inibidores , Fosfoproteínas/genética , Células-Tronco Pluripotentes/citologia , Piridinas/farmacologia , Pirimidinas/farmacologia , Interferência de RNA , Teratoma/metabolismo , Teratoma/patologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos , Proteínas de Sinalização YAP
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA