Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Genome Res ; 32(7): 1367-1384, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35705328

RESUMO

Changes in transcriptional regulatory networks can significantly alter cell fate. To gain insight into transcriptional dynamics, several studies have profiled bulk multi-omic data sets with parallel transcriptomic and epigenomic measurements at different stages of a developmental process. However, integrating these data to infer cell type-specific regulatory networks is a major challenge. We present dynamic regulatory module networks (DRMNs), a novel approach to infer cell type-specific cis-regulatory networks and their dynamics. DRMN integrates expression, chromatin state, and accessibility to predict cis-regulators of context-specific expression, where context can be cell type, developmental stage, or time point, and uses multitask learning to capture network dynamics across linearly and hierarchically related contexts. We applied DRMNs to study regulatory network dynamics in three developmental processes, each showing different temporal relationships and measuring a different combination of regulatory genomic data sets: cellular reprogramming, liver dedifferentiation, and forward differentiation. DRMN identified known and novel regulators driving cell type-specific expression patterns, showing its broad applicability to examine dynamics of gene regulatory networks from linearly and hierarchically related multi-omic data sets.


Assuntos
Redes Reguladoras de Genes , Genoma , Cromatina/genética , Genômica , Transcriptoma
2.
J Immunol ; 211(1): 154-162, 2023 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-37195197

RESUMO

Immunological tolerance toward the semiallogeneic fetus is one of many maternal adaptations required for a successful pregnancy. T cells are major players of the adaptive immune system and balance tolerance and protection at the maternal-fetal interface; however, their repertoire and subset programming are still poorly understood. Using emerging single-cell RNA sequencing technologies, we simultaneously obtained transcript, limited protein, and receptor repertoire at the single-cell level, from decidual and matched maternal peripheral human T cells. The decidua maintains a tissue-specific distribution of T cell subsets compared with the periphery. We find that decidual T cells maintain a unique transcriptome programming, characterized by restraint of inflammatory pathways by overexpression of negative regulators (DUSP, TNFAIP3, ZFP36) and expression of PD-1, CTLA-4, TIGIT, and LAG3 in some CD8 clusters. Finally, analyzing TCR clonotypes demonstrated decreased diversity in specific decidual T cell populations. Overall, our data demonstrate the power of multiomics analysis in revealing regulation of fetal-maternal immune coexistence.


Assuntos
Decídua , Proteogenômica , Gravidez , Feminino , Humanos , Subpopulações de Linfócitos T , Transcriptoma , Feto
3.
J Allergy Clin Immunol ; 153(6): 1647-1654, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38309597

RESUMO

BACKGROUND: Farm exposures in early life reduce the risks for childhood allergic diseases and asthma. There is less information about how farm exposures relate to respiratory illnesses and mucosal immune development. OBJECTIVE: We hypothesized that children raised in farm environments have a lower incidence of respiratory illnesses over the first 2 years of life than nonfarm children. We also analyzed whether farm exposures or respiratory illnesses were related to patterns of nasal cell gene expression. METHODS: The Wisconsin Infant Study Cohort included farm (n = 156) and nonfarm (n = 155) families with children followed to age 2 years. Parents reported prenatal farm and other environmental exposures. Illness frequency and severity were assessed using illness diaries and periodic surveys. Nasopharyngeal cell gene expression in a subset of 64 children at age 2 years was compared to farm exposure and respiratory illness history. RESULTS: Farm versus nonfarm children had nominally lower rates of respiratory illnesses (rate ratio 0.82 [95% CI, 0.69, 0.97]) with a stepwise reduction in illness rates in children exposed to 0, 1, or ≥2 animal species, but these trends were nonsignificant in a multivariable model. Farm exposures and preceding respiratory illnesses were positively related to nasal cell gene signatures for mononuclear cells and innate and antimicrobial responses. CONCLUSIONS: Maternal and infant exposure to farms and farm animals was associated with nonsignificant trends for reduced respiratory illnesses. Nasal cell gene expression in a subset of children suggests that farm exposures and respiratory illnesses in early life are associated with distinct patterns of mucosal immune expression.


Assuntos
Exposição Ambiental , Fazendas , Mucosa Nasal , Doenças Respiratórias , Humanos , Feminino , Animais , Masculino , Lactente , Exposição Ambiental/efeitos adversos , Pré-Escolar , Mucosa Nasal/imunologia , Doenças Respiratórias/imunologia , Doenças Respiratórias/epidemiologia , Doenças Respiratórias/genética , Animais Domésticos/imunologia , Recém-Nascido , Wisconsin/epidemiologia
4.
Genome Res ; 30(3): 361-374, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32179589

RESUMO

RNA-binding proteins (RNA-BPs) play critical roles in development and disease to regulate gene expression. However, genome-wide identification of their targets in primary human cells has been challenging. Here, we applied a modified CLIP-seq strategy to identify genome-wide targets of the FMRP translational regulator 1 (FMR1), a brain-enriched RNA-BP, whose deficiency leads to Fragile X Syndrome (FXS), the most prevalent inherited intellectual disability. We identified FMR1 targets in human dorsal and ventral forebrain neural progenitors and excitatory and inhibitory neurons differentiated from human pluripotent stem cells. In parallel, we measured the transcriptomes of the same four cell types upon FMR1 gene deletion. We discovered that FMR1 preferentially binds long transcripts in human neural cells. FMR1 targets include genes unique to human neural cells and associated with clinical phenotypes of FXS and autism. Integrative network analysis using graph diffusion and multitask clustering of FMR1 CLIP-seq and transcriptional targets reveals critical pathways regulated by FMR1 in human neural development. Our results demonstrate that FMR1 regulates a common set of targets among different neural cell types but also operates in a cell type-specific manner targeting distinct sets of genes in human excitatory and inhibitory neural progenitors and neurons. By defining molecular subnetworks and validating specific high-priority genes, we identify novel components of the FMR1 regulation program. Our results provide new insights into gene regulation by a critical neuronal RNA-BP in human neurodevelopment.


Assuntos
Proteína do X Frágil da Deficiência Intelectual/metabolismo , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Transtorno Autístico/genética , Linhagem Celular , Sequenciamento de Cromatina por Imunoprecipitação , Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/genética , Deleção de Genes , Redes Reguladoras de Genes , Humanos , Masculino , Células-Tronco Neurais/citologia , Neurogênese , Células-Tronco Pluripotentes/citologia , Prosencéfalo/citologia , Prosencéfalo/metabolismo , Transcriptoma
5.
PLoS Comput Biol ; 15(6): e1006758, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31246951

RESUMO

Many biological studies involve either (i) manipulating some aspect of a cell or its environment and then simultaneously measuring the effect on thousands of genes, or (ii) systematically manipulating each gene and then measuring the effect on some response of interest. A common challenge that arises in these studies is to explain how genes identified as relevant in the given experiment are organized into a subnetwork that accounts for the response of interest. The task of inferring a subnetwork is typically dependent on the information available in publicly available, structured databases, which suffer from incompleteness. However, a wealth of potentially relevant information resides in the scientific literature, such as information about genes associated with certain concepts of interest, as well as interactions that occur among various biological entities. We contend that by exploiting this information, we can improve the explanatory power and accuracy of subnetwork inference in multiple applications. Here we propose and investigate several ways in which information extracted from the scientific literature can be used to augment subnetwork inference. We show that we can use literature-extracted information to (i) augment the set of entities identified as being relevant in a subnetwork inference task, (ii) augment the set of interactions used in the process, and (iii) support targeted browsing of a large inferred subnetwork by identifying entities and interactions that are closely related to concepts of interest. We use this approach to uncover the pathways involved in interactions between a virus and a host cell, and the pathways that are regulated by a transcription factor associated with breast cancer. Our experimental results demonstrate that these approaches can provide more accurate and more interpretable subnetworks. Integer program code, background network data, and pathfinding code are available at https://github.com/Craven-Biostat-Lab/subnetwork_inference.


Assuntos
Biologia Computacional/métodos , Mineração de Dados/métodos , Redes Reguladoras de Genes/genética , Mapeamento de Interação de Proteínas/métodos , Mapas de Interação de Proteínas/genética , Bases de Dados Genéticas , HIV , Infecções por HIV/genética , Infecções por HIV/virologia , Humanos
6.
PLoS Comput Biol ; 13(5): e1006088, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29738528

RESUMO

Cells respond to stressful conditions by coordinating a complex, multi-faceted response that spans many levels of physiology. Much of the response is coordinated by changes in protein phosphorylation. Although the regulators of transcriptome changes during stress are well characterized in Saccharomyces cerevisiae, the upstream regulatory network controlling protein phosphorylation is less well dissected. Here, we developed a computational approach to infer the signaling network that regulates phosphorylation changes in response to salt stress. We developed an approach to link predicted regulators to groups of likely co-regulated phospho-peptides responding to stress, thereby creating new edges in a background protein interaction network. We then use integer linear programming (ILP) to integrate wild type and mutant phospho-proteomic data and predict the network controlling stress-activated phospho-proteomic changes. The network we inferred predicted new regulatory connections between stress-activated and growth-regulating pathways and suggested mechanisms coordinating metabolism, cell-cycle progression, and growth during stress. We confirmed several network predictions with co-immunoprecipitations coupled with mass-spectrometry protein identification and mutant phospho-proteomic analysis. Results show that the cAMP-phosphodiesterase Pde2 physically interacts with many stress-regulated transcription factors targeted by PKA, and that reduced phosphorylation of those factors during stress requires the Rck2 kinase that we show physically interacts with Pde2. Together, our work shows how a high-quality computational network model can facilitate discovery of new pathway interactions during osmotic stress.


Assuntos
Proteômica/métodos , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Cloreto de Sódio/química , Ciclo Celular , Biologia Computacional , Simulação por Computador , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Imunoprecipitação , Espectrometria de Massas , Modelos Biológicos , Pressão Osmótica , Fosforilação , Mapeamento de Interação de Proteínas , Proteínas Serina-Treonina Quinases/metabolismo , Proteoma , Transdução de Sinais , Fatores de Transcrição/metabolismo
7.
Plant Physiol ; 173(3): 1811-1823, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28159827

RESUMO

Arbuscular mycorrhizal (AM) associations enhance the phosphorous and nitrogen nutrition of host plants, but little is known about their role in potassium (K+) nutrition. Medicago truncatula plants were cocultured with the AM fungus Rhizophagus irregularis under high and low K+ regimes for 6 weeks. We determined how K+ deprivation affects plant development and mineral acquisition and how these negative effects are tempered by the AM colonization. The transcriptional response of AM roots under K+ deficiency was analyzed by whole-genome RNA sequencing. K+ deprivation decreased root biomass and external K+ uptake and modulated oxidative stress gene expression in M. truncatula roots. AM colonization induced specific transcriptional responses to K+ deprivation that seem to temper these negative effects. A gene network analysis revealed putative key regulators of these responses. This study confirmed that AM associations provide some tolerance to K+ deprivation to host plants, revealed that AM symbiosis modulates the expression of specific root genes to cope with this nutrient stress, and identified putative regulators participating in these tolerance mechanisms.


Assuntos
Regulação da Expressão Gênica de Plantas , Redes Reguladoras de Genes , Medicago truncatula/genética , Raízes de Plantas/genética , Potássio/metabolismo , Análise por Conglomerados , Perfilação da Expressão Gênica/métodos , Interações Hospedeiro-Patógeno , Medicago truncatula/metabolismo , Medicago truncatula/microbiologia , Micorrizas/fisiologia , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Raízes de Plantas/metabolismo , Raízes de Plantas/microbiologia , Simbiose/fisiologia , Fatores de Tempo
8.
Bioinformatics ; 32(10): 1509-17, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-26801959

RESUMO

MOTIVATION: Identifying the shared and pathogen-specific components of host transcriptional regulatory programs is important for understanding the principles of regulation of immune response. Recent efforts in systems biology studies of infectious diseases have resulted in a large collection of datasets measuring host transcriptional response to various pathogens. Computational methods to identify and compare gene expression modules across different infections offer a powerful way to identify strain-specific and shared components of the regulatory program. An important challenge is to identify statistically robust gene expression modules as well as to reliably detect genes that change their module memberships between infections. RESULTS: We present MULCCH (MULti-task spectral Consensus Clustering for Hierarchically related tasks), a consensus extension of a multi-task clustering algorithm to infer high-confidence strain-specific host response modules under infections from multiple virus strains. On simulated data, MULCCH more accurately identifies genes exhibiting pathogen-specific patterns compared to non-consensus and nonmulti-task clustering approaches. Application of MULCCH to mammalian transcriptional response to a panel of influenza viruses showed that our method identifies clusters with greater coherence compared to non-consensus methods. Further, MULCCH derived clusters are enriched for several immune system-related processes and regulators. In summary, MULCCH provides a reliable module-based approach to identify molecular pathways and gene sets characterizing commonality and specificity of host response to viruses of different pathogenicities. AVAILABILITY AND IMPLEMENTATION: The source code is available at https://bitbucket.org/roygroup/mulcch CONTACT: sroy@biostat.wisc.edu SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Genoma , Transcriptoma , Algoritmos , Animais , Análise por Conglomerados , Biologia Computacional , Consenso , Perfilação da Expressão Gênica , Redes Reguladoras de Genes
9.
PLoS Comput Biol ; 12(7): e1005013, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27403523

RESUMO

Mammalian host response to pathogenic infections is controlled by a complex regulatory network connecting regulatory proteins such as transcription factors and signaling proteins to target genes. An important challenge in infectious disease research is to understand molecular similarities and differences in mammalian host response to diverse sets of pathogens. Recently, systems biology studies have produced rich collections of omic profiles measuring host response to infectious agents such as influenza viruses at multiple levels. To gain a comprehensive understanding of the regulatory network driving host response to multiple infectious agents, we integrated host transcriptomes and proteomes using a network-based approach. Our approach combines expression-based regulatory network inference, structured-sparsity based regression, and network information flow to infer putative physical regulatory programs for expression modules. We applied our approach to identify regulatory networks, modules and subnetworks that drive host response to multiple influenza infections. The inferred regulatory network and modules are significantly enriched for known pathways of immune response and implicate apoptosis, splicing, and interferon signaling processes in the differential response of viral infections of different pathogenicities. We used the learned network to prioritize regulators and study virus and time-point specific networks. RNAi-based knockdown of predicted regulators had significant impact on viral replication and include several previously unknown regulators. Taken together, our integrated analysis identified novel module level patterns that capture strain and pathogenicity-specific patterns of expression and helped identify important regulators of host response to influenza infection.


Assuntos
Perfilação da Expressão Gênica/métodos , Redes Reguladoras de Genes/genética , Interações Hospedeiro-Patógeno/genética , Proteoma/genética , Proteômica/métodos , Transcriptoma/genética , Animais , Humanos , Influenza Humana/genética , Camundongos , Modelos Biológicos , Biologia de Sistemas
10.
Nucleic Acids Res ; 43(18): 8694-712, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26338778

RESUMO

Long range regulatory interactions among distal enhancers and target genes are important for tissue-specific gene expression. Genome-scale identification of these interactions in a cell line-specific manner, especially using the fewest possible datasets, is a significant challenge. We develop a novel computational approach, Regulatory Interaction Prediction for Promoters and Long-range Enhancers (RIPPLE), that integrates published Chromosome Conformation Capture (3C) data sets with a minimal set of regulatory genomic data sets to predict enhancer-promoter interactions in a cell line-specific manner. Our results suggest that CTCF, RAD21, a general transcription factor (TBP) and activating chromatin marks are important determinants of enhancer-promoter interactions. To predict interactions in a new cell line and to generate genome-wide interaction maps, we develop an ensemble version of RIPPLE and apply it to generate interactions in five human cell lines. Computational validation of these predictions using existing ChIA-PET and Hi-C data sets showed that RIPPLE accurately predicts interactions among enhancers and promoters. Enhancer-promoter interactions tend to be organized into subnetworks representing coordinately regulated sets of genes that are enriched for specific biological processes and cis-regulatory elements. Overall, our work provides a systematic approach to predict and interpret enhancer-promoter interactions in a genome-wide cell-type specific manner using a few experimentally tractable measurements.


Assuntos
Elementos Facilitadores Genéticos , Genômica/métodos , Modelos Genéticos , Regiões Promotoras Genéticas , Algoritmos , Fator de Ligação a CCCTC , Proteínas de Ciclo Celular/análise , Linhagem Celular , Cromatina/química , Cromatina/metabolismo , Proteínas Cromossômicas não Histona/análise , Código das Histonas , Humanos , Proteínas Repressoras/análise , Proteína de Ligação a TATA-Box/análise , Coesinas
11.
Plant Physiol ; 169(1): 233-65, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26175514

RESUMO

The legume-rhizobium symbiosis is initiated through the activation of the Nodulation (Nod) factor-signaling cascade, leading to a rapid reprogramming of host cell developmental pathways. In this work, we combine transcriptome sequencing with molecular genetics and network analysis to quantify and categorize the transcriptional changes occurring in roots of Medicago truncatula from minutes to days after inoculation with Sinorhizobium medicae. To identify the nature of the inductive and regulatory cues, we employed mutants with absent or decreased Nod factor sensitivities (i.e. Nodulation factor perception and Lysine motif domain-containing receptor-like kinase3, respectively) and an ethylene (ET)-insensitive, Nod factor-hypersensitive mutant (sickle). This unique data set encompasses nine time points, allowing observation of the symbiotic regulation of diverse biological processes with high temporal resolution. Among the many outputs of the study is the early Nod factor-induced, ET-regulated expression of ET signaling and biosynthesis genes. Coupled with the observation of massive transcriptional derepression in the ET-insensitive background, these results suggest that Nod factor signaling activates ET production to attenuate its own signal. Promoter:ß-glucuronidase fusions report ET biosynthesis both in root hairs responding to rhizobium as well as in meristematic tissue during nodule organogenesis and growth, indicating that ET signaling functions at multiple developmental stages during symbiosis. In addition, we identified thousands of novel candidate genes undergoing Nod factor-dependent, ET-regulated expression. We leveraged the power of this large data set to model Nod factor- and ET-regulated signaling networks using MERLIN, a regulatory network inference algorithm. These analyses predict key nodes regulating the biological process impacted by Nod factor perception. We have made these results available to the research community through a searchable online resource.


Assuntos
Etilenos/metabolismo , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Medicago truncatula/genética , Medicago truncatula/microbiologia , Proteínas de Plantas/metabolismo , Raízes de Plantas/genética , Transdução de Sinais/efeitos dos fármacos , Transcriptoma/genética , Vias Biossintéticas/efeitos dos fármacos , Vias Biossintéticas/genética , Análise por Conglomerados , Etilenos/farmacologia , Retroalimentação Fisiológica , Regulação da Expressão Gênica de Plantas/efeitos dos fármacos , Ontologia Genética , Redes Reguladoras de Genes , Genes de Plantas , Medicago truncatula/efeitos dos fármacos , Proteínas de Plantas/genética , Raízes de Plantas/efeitos dos fármacos , Raízes de Plantas/crescimento & desenvolvimento , Raízes de Plantas/microbiologia , Rhizobium/efeitos dos fármacos , Rhizobium/fisiologia , Transdução de Sinais/genética , Simbiose/genética , Fatores de Tempo , Fatores de Transcrição/metabolismo , Transcrição Gênica/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos
12.
Mol Syst Biol ; 10: 759, 2014 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-25411400

RESUMO

Stressed cells coordinate a multi-faceted response spanning many levels of physiology. Yet knowledge of the complete stress-activated regulatory network as well as design principles for signal integration remains incomplete. We developed an experimental and computational approach to integrate available protein interaction data with gene fitness contributions, mutant transcriptome profiles, and phospho-proteome changes in cells responding to salt stress, to infer the salt-responsive signaling network in yeast. The inferred subnetwork presented many novel predictions by implicating new regulators, uncovering unrecognized crosstalk between known pathways, and pointing to previously unknown 'hubs' of signal integration. We exploited these predictions to show that Cdc14 phosphatase is a central hub in the network and that modification of RNA polymerase II coordinates induction of stress-defense genes with reduction of growth-related transcripts. We find that the orthologous human network is enriched for cancer-causing genes, underscoring the importance of the subnetwork's predictions in understanding stress biology.


Assuntos
Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Ciclo Celular/metabolismo , Biologia Computacional/métodos , Perfilação da Expressão Gênica , Regulação Fúngica da Expressão Gênica , Aptidão Genética , Proteínas Tirosina Fosfatases/metabolismo , RNA Polimerase II/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Transdução de Sinais , Cloreto de Sódio/metabolismo , Estresse Fisiológico
13.
PLoS Comput Biol ; 10(5): e1003626, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24874113

RESUMO

Systematic, genome-wide loss-of-function experiments can be used to identify host factors that directly or indirectly facilitate or inhibit the replication of a virus in a host cell. We present an approach that combines an integer linear program and a diffusion kernel method to infer the pathways through which those host factors modulate viral replication. The inputs to the method are a set of viral phenotypes observed in single-host-gene mutants and a background network consisting of a variety of host intracellular interactions. The output is an ensemble of subnetworks that provides a consistent explanation for the measured phenotypes, predicts which unassayed host factors modulate the virus, and predicts which host factors are the most direct interfaces with the virus. We infer host-virus interaction subnetworks using data from experiments screening the yeast genome for genes modulating the replication of two RNA viruses. Because a gold-standard network is unavailable, we assess the predicted subnetworks using both computational and qualitative analyses. We conduct a cross-validation experiment in which we predict whether held-aside test genes have an effect on viral replication. Our approach is able to make high-confidence predictions more accurately than several baselines, and about as well as the best baseline, which does not infer mechanistic pathways. We also examine two kinds of predictions made by our method: which host factors are nearest to a direct interaction with a viral component, and which unassayed host genes are likely to be involved in viral replication. Multiple predictions are supported by recent independent experimental data, or are components or functional partners of confirmed relevant complexes or pathways. Integer program code, background network data, and inferred host-virus subnetworks are available at http://www.biostat.wisc.edu/~craven/chasman_host_virus/.


Assuntos
Transformação Celular Viral/fisiologia , Proteínas Fúngicas/metabolismo , Vírus de RNA/fisiologia , Transdução de Sinais/fisiologia , Replicação Viral/fisiologia , Leveduras/metabolismo , Leveduras/virologia , Regulação Fúngica da Expressão Gênica/fisiologia , Genes Virais
15.
Front Genet ; 12: 788318, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35087569

RESUMO

Cancer risk by environmental exposure is modulated by an individual's genetics and age at exposure. This age-specific period of susceptibility is referred to as the "Window of Susceptibility" (WOS). Rats have a similar WOS for developing breast cancer. A previous study in rat identified an age-specific long-range regulatory interaction for the cancer gene, Pappa, that is associated with breast cancer susceptibility. However, the global role of three-dimensional genome organization and downstream gene expression programs in the WOS is not known. Therefore, we generated Hi-C and RNA-seq data in rat mammary epithelial cells within and outside the WOS. To systematically identify higher-order changes in 3D genome organization, we developed NE-MVNMF that combines network enhancement followed by multitask non-negative matrix factorization. We examined three-dimensional genome organization dynamics at the level of individual loops as well as higher-order domains. Differential chromatin interactions tend to be associated with differentially up-regulated genes with the WOS and recapitulate several human SNP-gene interactions associated with breast cancer susceptibility. Our approach identified genomic blocks of regions with greater overall differences in contact count between the two time points when the cluster assignments change and identified genes and pathways implicated in early carcinogenesis and cancer treatment. Our results suggest that WOS-specific changes in 3D genome organization are linked to transcriptional changes that may influence susceptibility to breast cancer.

16.
Am J Reprod Immunol ; 86(6): e13495, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34411378

RESUMO

PROBLEM: Mucosal-Associated Invariant T (MAIT) cells have been recently identified at the maternal-fetal interface. However, transcriptional programming of decidual MAIT cells in pregnancy remains poorly understood. METHOD OF STUDY: We employed a multiomic approach to address this question. Mononuclear cells from the decidua basalis and parietalis, and control PBMCs, were analyzed via flow cytometry to investigate MAIT cells in the decidua and assess their transcription factor expression. In a separate study, both decidual and matched peripheral MAIT cells were analyzed using Cellular Indexing of Transcriptomes and Epitopes by Sequencing (CITE-seq) coupled with gene expression analysis. Lastly, decidual MAIT cells were stimulated with E.coli and expression of MR1 by antigen presenting cells was measured to evaluate decidual MAIT cell function. RESULTS: First, we identified MAIT cells in both the decidua basalis and parietalis. CITE-seq, coupled with scRNA-seq gene expression analysis, highlighted transcriptional programming differences between decidual and matched peripheral MAIT cells at a single cell resolution. Transcription factor expression analysis further highlighted transcriptional differences between decidual MAIT cells and non-matched peripheral MAIT cells. Functionally, MAIT cells are skewed towards IFNγ and TNFα production upon stimulation, with E.coli leading to IFNγ production. Lastly, we demonstrate that MR1, the antigen presenting molecule restricting MAIT cells, is expressed by decidual APCs. CONCLUSION: MAIT cells are present in the decidua basalis and obtain a unique gene expression profile. The presence of MR1 on APCs coupled with in vitro activation by E.coli suggests that MAIT cells might be involved in tissue-repair mechanisms at the maternal-fetal interface.


Assuntos
Decídua/metabolismo , Células T Invariantes Associadas à Mucosa/metabolismo , Placenta/metabolismo , Decídua/imunologia , Feminino , Citometria de Fluxo , Humanos , Leucócitos/imunologia , Células T Invariantes Associadas à Mucosa/imunologia , Placenta/imunologia , Gravidez
17.
Methods Mol Biol ; 1883: 161-194, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30547400

RESUMO

Transcriptional regulatory networks specify the regulatory proteins of target genes that control the context-specific expression levels of genes. With our ability to profile the different types of molecular components of cells under different conditions, we are now uniquely positioned to infer regulatory networks in diverse biological contexts such as different cell types, tissues, and time points. In this chapter, we cover two main classes of computational methods to integrate different types of information to infer genome-scale transcriptional regulatory networks. The first class of methods focuses on integrative methods for specifically inferring connections between transcription factors and target genes by combining gene expression data with regulatory edge-specific knowledge. The second class of methods integrates upstream signaling networks with transcriptional regulatory networks by combining gene expression data with protein-protein interaction networks and proteomic datasets. We conclude with a section on practical applications of a network inference algorithm to infer a genome-scale regulatory network.


Assuntos
Biologia Computacional/métodos , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Modelos Genéticos , Algoritmos , Biologia Computacional/instrumentação , Conjuntos de Dados como Assunto , Perfilação da Expressão Gênica/instrumentação , Perfilação da Expressão Gênica/métodos , Genoma/genética , Mapas de Interação de Proteínas/genética , Proteômica/instrumentação , Proteômica/métodos , Software , Fatores de Transcrição/metabolismo
18.
Nat Commun ; 10(1): 5449, 2019 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-31811132

RESUMO

The three-dimensional (3D) organization of the genome plays an important role in gene regulation bringing distal sequence elements in 3D proximity to genes hundreds of kilobases away. Hi-C is a powerful genome-wide technique to study 3D genome organization. Owing to experimental costs, high resolution Hi-C datasets are limited to a few cell lines. Computational prediction of Hi-C counts can offer a scalable and inexpensive approach to examine 3D genome organization across multiple cellular contexts. Here we present HiC-Reg, an approach to predict contact counts from one-dimensional regulatory signals. HiC-Reg predictions identify topologically associating domains and significant interactions that are enriched for CCCTC-binding factor (CTCF) bidirectional motifs and interactions identified from complementary sources. CTCF and chromatin marks, especially repressive and elongation marks, are most important for HiC-Reg's predictive performance. Taken together, HiC-Reg provides a powerful framework to generate high-resolution profiles of contact counts that can be used to study individual locus level interactions and higher-order organizational units of the genome.


Assuntos
Cromossomos/química , Biologia Computacional/métodos , Simulação por Computador , Genoma , Genômica/métodos , Fator de Ligação a CCCTC/metabolismo , Linhagem Celular , Cromatina/química , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Aprendizado de Máquina , Modelos Genéticos , Regiões Promotoras Genéticas/genética
19.
Front Immunol ; 10: 3065, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-32038619

RESUMO

A successful pregnancy requires many physiological adaptations from the mother, including the establishment of tolerance toward the semiallogeneic fetus. Innate lymphoid cells (ILCs) have arisen as important players in immune regulation and tissue homeostasis at mucosal and barrier surfaces. Dimensionality reduction and transcriptomic analysis revealed the presence of two novel CD56Bright decidual ILCs that express low T-bet and divergent Eomes levels. Transcriptional correlation with recently identified first trimester decidual dNKs suggests that these novel decidual ILCs might be present throughout pregnancy. Functional testing with permutation analysis revealed production of multiple factors by individual cells, with a preference for IFNγ and VEGF. Overall, our data suggests continuity of a unique decidual innate lymphocytes across pregnancy with a polyfunctional functional profile conducive for pregnancy.


Assuntos
Decídua/fisiologia , Células Matadoras Naturais/fisiologia , Gravidez/imunologia , Adulto , Antígeno CD56/metabolismo , Feminino , Perfilação da Expressão Gênica , Homeostase , Humanos , Tolerância Imunológica/genética , Imunidade Inata , Interferon gama/metabolismo , Proteínas com Domínio T/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
20.
Cell Syst ; 9(2): 167-186.e12, 2019 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-31302154

RESUMO

Neuroepithelial stem cells (NSC) from different anatomical regions of the embryonic neural tube's rostrocaudal axis can differentiate into diverse central nervous system tissues, but the transcriptional regulatory networks governing these processes are incompletely understood. Here, we measure region-specific NSC gene expression along the rostrocaudal axis in a human pluripotent stem cell model of early central nervous system development over a 72-h time course, spanning the hindbrain to cervical spinal cord. We introduce Escarole, a probabilistic clustering algorithm for non-stationary time series, and combine it with prior-based regulatory network inference to identify genes that are regulated dynamically and predict their upstream regulators. We identify known regulators of patterning and neural development, including the HOX genes, and predict a direct regulatory connection between the transcription factor POU3F2 and target gene STMN2. We demonstrate that POU3F2 is required for expression of STMN2, suggesting that this regulatory connection is important for region specificity of NSCs.


Assuntos
Células-Tronco Neurais/metabolismo , Rombencéfalo/embriologia , Medula Espinal/embriologia , Diferenciação Celular/genética , Linhagem Celular , Regulação da Expressão Gênica no Desenvolvimento/genética , Redes Reguladoras de Genes/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Células-Tronco Neurais/fisiologia , Células Neuroepiteliais , Neurogênese , Neurônios/metabolismo , Fatores do Domínio POU/genética , Fatores do Domínio POU/metabolismo , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/fisiologia , Estatmina/genética , Estatmina/metabolismo , Transcriptoma/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA