Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Front Bioinform ; 4: 1336135, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38690527

RESUMO

Background: Understanding how cells and tissues respond to stress factors and perturbations during disease processes is crucial for developing effective prevention, diagnosis, and treatment strategies. Single-cell RNA sequencing (scRNA-seq) enables high-resolution identification of cells and exploration of cell heterogeneity, shedding light on cell differentiation/maturation and functional differences. Recent advancements in multimodal sequencing technologies have focused on improving access to cell-specific subgroups for functional genomics analysis. To facilitate the functional annotation of cell groups and characterization of molecular mechanisms underlying cell trajectories, we introduce the Pathways, Annotated Gene Lists, and Gene Signatures Electronic Repository for Single-Cell Functional Genomics Analysis (PAGER-scFGA). Results: We have developed PAGER-scFGA, which integrates cell functional annotations and gene-set enrichment analysis into popular single-cell analysis pipelines such as Scanpy. Using differentially expressed genes (DEGs) from pairwise cell clusters, PAGER-scFGA infers cell functions through the enrichment of potential cell-marker genesets. Moreover, PAGER-scFGA provides pathways, annotated gene lists, and gene signatures (PAGs) enriched in specific cell subsets with tissue compositions and continuous transitions along cell trajectories. Additionally, PAGER-scFGA enables the construction of a gene subcellular map based on DEGs and allows examination of the gene functional compartments (GFCs) underlying cell maturation/differentiation. In a real-world case study of mouse natural killer (mNK) cells, PAGER-scFGA revealed two major stages of natural killer (NK) cells and three trajectories from the precursor stage to NK T-like mature stage within blood, spleen, and bone marrow tissues. As the trajectories progress to later stages, the DEGs exhibit greater divergence and variability. However, the DEGs in different trajectories still interact within a network during NK cell maturation. Notably, PAGER-scFGA unveiled cell cytotoxicity, exocytosis, and the response to interleukin (IL) signaling pathways and associated network models during the progression from precursor NK cells to mature NK cells. Conclusion: PAGER-scFGA enables in-depth exploration of functional insights and presents a comprehensive knowledge map of gene networks and GFCs, which can be utilized for future studies and hypothesis generation. It is expected to become an indispensable tool for inferring cell functions and detecting molecular mechanisms within cell trajectories in single-cell studies. The web app (accessible at https://au-singlecell.streamlit.app/) is publicly available.

2.
J Geriatr Oncol ; 15(5): 101772, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38676976

RESUMO

INTRODUCTION: Older adults with prostate cancer (PC) are at risk of polypharmacy, which further complicates disease management and health-related quality of life (HRQoL). This study evaluated the association between polypharmacy and HRQoL among Medicare beneficiaries with PC. MATERIALS AND METHODS: This observational, retrospective study analyzed data from the Surveillance, Epidemiology, and End Results (SEER) Medicare Health Outcomes Survey (MHOS) data resource. Beneficiaries aged ≥65 and enrolled in Medicare Advantage Organizations were included if they had a PC diagnosis and continuously enrolled in Part D for 12 months prior to the completion of MHOS. Polypharmacy was determined based on the unique number of concurrent Part D prescriptions during 12 months before survey: no polypharmacy (NP, n = 0-4), polypharmacy (PP, n = 5-9), and excessive polypharmacy (EPP, n ≥ 10). HRQoL was assessed using the Physical and Mental Component Summary T-scores (PCS and MCS, respectively) in MHOS. ANOVA and Pearson's Chi-Square tests were performed to assess variances between polypharmacy and continuous/categorical variables. Multivariate linear regression models with generalized estimating equations were used to assess the association between polypharmacy and HRQoL. The severely impaired HRQoL cohort was identified based on normalized z-scores of PCS and MCS. Odds ratios were calculated to prioritize drug-drug and class-class pairs associated with patients with severely impaired HRQoL. RESULTS: Data from 16,573 beneficiaries (24,126 records) showed that 44.4% had PP and 10.1% had EPP. Beneficiaries with PP and EPP had significantly lower mean PCS and MCS scores compared to those without polypharmacy (p < 0.001). After adjusting for covariates, beneficiaries with EPP had clinically significantly lower PCS (adjusted marginal difference: -8.47 [-9.00, -7.94]) and MCS (adjusted marginal difference: -4.32 [-4.89, -3.75]) compared to the NP group. Top-ranked drug-drug pairs like tiotropium bromide and oxycodone/acetaminophen exhibited significant associations with HRQoL decline. Analysis of class-class pairs highlighted (1) corticosteroid hormone receptor agonists and opioid agonists and (2) benzodiazepines and adrenergic beta2-agonists as having significant associations with HRQoL decline. DISCUSSION: Polypharmacy exhibits a significant association with HRQoL declines among older adults with PC.


Assuntos
Polimedicação , Neoplasias da Próstata , Qualidade de Vida , Programa de SEER , Humanos , Masculino , Idoso , Estudos Retrospectivos , Estados Unidos , Idoso de 80 Anos ou mais , Neoplasias da Próstata/tratamento farmacológico , Medicare Part D/estatística & dados numéricos , Medicare
3.
bioRxiv ; 2024 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-38617348

RESUMO

This study introduces the GeneTerrain Knowledge Map Representation (GTKM), a novel method for visualizing gene expression data in cancer research. GTKM leverages protein-protein interactions to graphically display differentially expressed genes (DEGs) on a 2-dimensional contour plot, offering a more nuanced understanding of gene interactions and expression patterns compared to traditional heatmap methods. The research demonstrates GTKM's utility through four case studies on glioblastoma (GBM) datasets, focusing on survival analysis, subtype identification, IDH1 mutation analysis, and drug sensitivities of different tumor cell lines. Additionally, a prototype website has been developed to showcase these findings, indicating the method's adaptability for various cancer types. The study reveals that GTKM effectively identifies gene patterns associated with different clinical outcomes in GBM, and its profiles enable the identification of sub-gene signature patterns crucial for predicting survival. The methodology promises significant advancements in precision medicine, providing a powerful tool for understanding complex gene interactions and identifying potential therapeutic targets in cancer treatment.

4.
Cancer Res ; 84(5): 757-770, 2024 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-38190709

RESUMO

Overweight and obesity are identified by a high body mass index (BMI) and carry significant health risks due to associated comorbidities. Although epidemiologic data connect overweight/obesity with 13 cancer types, a better understanding of the molecular mechanisms underlying this correlation is needed to improve prevention and treatment strategies. In this study, we conducted a comprehensive analysis of molecular differences between overweight or obese patients and normal weight patients across 14 different cancer types from The Cancer Genome Atlas. Using the propensity score weighting algorithm to control for confounding factors, obesity-specific mutational features were identified, such as higher mutation burden in rectal cancer and biased mutational signatures in other cancers. Differentially expressed genes (DEG) in tumors from patients with overweight/obesity were predominantly upregulated and enriched in inflammatory and hormone-related pathways. These DEGs were significantly associated with survival rates in various cancer types, highlighting the impact of elevated body fat on gene expression profiles and clinical outcomes in patients with cancer. Interestingly, while high BMI seemed to have a negative impact on most cancer types, the normal weight-biased mutational and gene expression patterns indicated overweight/obesity may be beneficial in endometrial cancer, suggesting the presence of an "obesity paradox" in this context. Body fat also significantly impacted the tumor microenvironment by modulating immune cell infiltration, underscoring the importance of understanding the interplay between weight and immune response in cancer progression. Together, this study systematically elucidates the molecular differences corresponding to body weight in multiple cancer types, offering potentially critical insights for developing precision therapy for patients with cancer. SIGNIFICANCE: Elucidation of the complex interplay between body weight and the molecular landscape of cancer could potentially guide tailored therapies and improve patient management amid the global obesity crisis.


Assuntos
Neoplasias , Sobrepeso , Humanos , Sobrepeso/epidemiologia , Obesidade/complicações , Obesidade/genética , Obesidade/epidemiologia , Neoplasias/epidemiologia , Índice de Massa Corporal , Comorbidade , Microambiente Tumoral
5.
Pharmaceutics ; 16(1)2023 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-38258072

RESUMO

The tumor microenvironment (TME) is pivotal in tumor growth and metastasis, aligning with the "Seed and Soil" theory. Within the TME, tumor-associated macrophages (TAMs) play a central role, profoundly influencing tumor progression. Strategies targeting TAMs have surfaced as potential therapeutic avenues, encompassing interventions to block TAM recruitment, eliminate TAMs, reprogram M2 TAMs, or bolster their phagocytic capabilities via specific pathways. Nanomaterials including inorganic materials, organic materials for small molecules and large molecules stand at the forefront, presenting significant opportunities for precise targeting and modulation of TAMs to enhance therapeutic efficacy in cancer treatment. This review provides an overview of the progress in designing nanoparticles for interacting with and influencing the TAMs as a significant strategy in cancer therapy. This comprehensive review presents the role of TAMs in the TME and various targeting strategies as a promising frontier in the ever-evolving field of cancer therapy. The current trends and challenges associated with TAM-based therapy in cancer are presented.

6.
Front Big Data ; 5: 1016606, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36407327

RESUMO

Background and contribution: In network biology, molecular functions can be characterized by network-based inference, or "guilt-by-associations." PageRank-like tools have been applied in the study of biomolecular interaction networks to obtain further the relative significance of all molecules in the network. However, there is a great deal of inherent noise in widely accessible data sets for gene-to-gene associations or protein-protein interactions. How to develop robust tests to expand, filter, and rank molecular entities in disease-specific networks remains an ad hoc data analysis process. Results: We describe a new biomolecular characterization and prioritization tool called Weighted In-Network Node Expansion and Ranking (WINNER). It takes the input of any molecular interaction network data and generates an optionally expanded network with all the nodes ranked according to their relevance to one another in the network. To help users assess the robustness of results, WINNER provides two different types of statistics. The first type is a node-expansion p-value, which helps evaluate the statistical significance of adding "non-seed" molecules to the original biomolecular interaction network consisting of "seed" molecules and molecular interactions. The second type is a node-ranking p-value, which helps evaluate the relative statistical significance of the contribution of each node to the overall network architecture. We validated the robustness of WINNER in ranking top molecules by spiking noises in several network permutation experiments. We have found that node degree-preservation randomization of the gene network produced normally distributed ranking scores, which outperform those made with other gene network randomization techniques. Furthermore, we validated that a more significant proportion of the WINNER-ranked genes was associated with disease biology than existing methods such as PageRank. We demonstrated the performance of WINNER with a few case studies, including Alzheimer's disease, breast cancer, myocardial infarctions, and Triple negative breast cancer (TNBC). In all these case studies, the expanded and top-ranked genes identified by WINNER reveal disease biology more significantly than those identified by other gene prioritizing software tools, including Ingenuity Pathway Analysis (IPA) and DiAMOND. Conclusion: WINNER ranking strongly correlates to other ranking methods when the network covers sufficient node and edge information, indicating a high network quality. WINNER users can use this new tool to robustly evaluate a list of candidate genes, proteins, or metabolites produced from high-throughput biology experiments, as long as there is available gene/protein/metabolic network information.

7.
JCI Insight ; 7(16)2022 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-35852875

RESUMO

Key molecular regulators of acquired radiation resistance in recurrent glioblastoma (GBM) are largely unknown, with a dearth of accurate preclinical models. To address this, we generated 8 GBM patient-derived xenograft (PDX) models of acquired radiation therapy-selected (RTS) resistance compared with same-patient, treatment-naive (radiation-sensitive, unselected; RTU) PDXs. These likely unique models mimic the longitudinal evolution of patient recurrent tumors following serial radiation therapy. Indeed, while whole-exome sequencing showed retention of major genomic alterations in the RTS lines, we did detect a chromosome 12q14 amplification that was associated with clinical GBM recurrence in 2 RTS models. A potentially novel bioinformatics pipeline was applied to analyze phenotypic, transcriptomic, and kinomic alterations, which identified long noncoding RNAs (lncRNAs) and targetable, PDX-specific kinases. We observed differential transcriptional enrichment of DNA damage repair pathways in our RTS models, which correlated with several lncRNAs. Global kinomic profiling separated RTU and RTS models, but pairwise analyses indicated that there are multiple molecular routes to acquired radiation resistance. RTS model-specific kinases were identified and targeted with clinically relevant small molecule inhibitors. This cohort of in vivo RTS patient-derived models will enable future preclinical therapeutic testing to help overcome the treatment resistance seen in patients with GBM.


Assuntos
Glioblastoma , RNA Longo não Codificante , Animais , Modelos Animais de Doenças , Genômica , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/radioterapia , Humanos , Recidiva Local de Neoplasia , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Bioinformatics ; 38(Suppl 1): i359-i368, 2022 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-35758816

RESUMO

SUMMARY: In biology, graph layout algorithms can reveal comprehensive biological contexts by visually positioning graph nodes in their relevant neighborhoods. A layout software algorithm/engine commonly takes a set of nodes and edges and produces layout coordinates of nodes according to edge constraints. However, current layout engines normally do not consider node, edge or node-set properties during layout and only curate these properties after the layout is created. Here, we propose a new layout algorithm, distance-bounded energy-field minimization algorithm (DEMA), to natively consider various biological factors, i.e., the strength of gene-to-gene association, the gene's relative contribution weight and the functional groups of genes, to enhance the interpretation of complex network graphs. In DEMA, we introduce a parameterized energy model where nodes are repelled by the network topology and attracted by a few biological factors, i.e., interaction coefficient, effect coefficient and fold change of gene expression. We generalize these factors as gene weights, protein-protein interaction weights, gene-to-gene correlations and the gene set annotations-four parameterized functional properties used in DEMA. Moreover, DEMA considers further attraction/repulsion/grouping coefficient to enable different preferences in generating network views. Applying DEMA, we performed two case studies using genetic data in autism spectrum disorder and Alzheimer's disease, respectively, for gene candidate discovery. Furthermore, we implement our algorithm as a plugin to Cytoscape, an open-source software platform for visualizing networks; hence, it is convenient. Our software and demo can be freely accessed at http://discovery.informatics.uab.edu/dema. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Algoritmos , Transtorno do Espectro Autista , Fatores Biológicos , Humanos , Software
9.
Front Genet ; 13: 820361, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35495152

RESUMO

Functional genomics studies have helped researchers annotate differentially expressed gene lists, extract gene expression signatures, and identify biological pathways from omics profiling experiments conducted on biological samples. The current geneset, network, and pathway analysis (GNPA) web servers, e.g., DAVID, EnrichR, WebGestaltR, or PAGER, do not allow automated integrative functional genomic downstream analysis. In this study, we developed a new web-based interactive application, "PAGER Web APP", which supports online R scripting of integrative GNPA. In a case study of melanoma drug resistance, we showed that the new PAGER Web APP enabled us to discover highly relevant pathways and network modules, leading to novel biological insights. We also compared PAGER Web APP's pathway analysis results retrieved among PAGER, EnrichR, and WebGestaltR to show its advantages in integrative GNPA. The interactive online web APP is publicly accessible from the link, https://aimed-lab.shinyapps.io/PAGERwebapp/.

10.
Immunity ; 55(3): 494-511.e11, 2022 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-35263568

RESUMO

Interleukin (IL)-22 is central to immune defense at barrier sites. We examined the contributions of innate lymphoid cell (ILC) and T cell-derived IL-22 during Citrobacter rodentium (C.r) infection using mice that both report Il22 expression and allow lineage-specific deletion. ILC-derived IL-22 activated STAT3 in C.r-colonized surface intestinal epithelial cells (IECs) but only temporally restrained bacterial growth. T cell-derived IL-22 induced a more robust and extensive activation of STAT3 in IECs, including IECs lining colonic crypts, and T cell-specific deficiency of IL-22 led to pathogen invasion of the crypts and increased mortality. This reflected a requirement for T cell-derived IL-22 for the expression of a host-protective transcriptomic program that included AMPs, neutrophil-recruiting chemokines, and mucin-related molecules, and it restricted IFNγ-induced proinflammatory genes. Our findings demonstrate spatiotemporal differences in the production and action of IL-22 by ILCs and T cells during infection and reveal an indispensable role for IL-22-producing T cells in the protection of the intestinal crypts.


Assuntos
Citrobacter rodentium , Infecções por Enterobacteriaceae , Animais , Antibacterianos , Imunidade Inata , Interleucinas/metabolismo , Mucosa Intestinal , Linfócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T/metabolismo , Interleucina 22
12.
Front Big Data ; 4: 725276, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34604741

RESUMO

Unsupervised learning techniques, such as clustering and embedding, have been increasingly popular to cluster biomedical samples from high-dimensional biomedical data. Extracting clinical data or sample meta-data shared in common among biomedical samples of a given biological condition remains a major challenge. Here, we describe a powerful analytical method called Statistical Enrichment Analysis of Samples (SEAS) for interpreting clustered or embedded sample data from omics studies. The method derives its power by focusing on sample sets, i.e., groups of biological samples that were constructed for various purposes, e.g., manual curation of samples sharing specific characteristics or automated clusters generated by embedding sample omic profiles from multi-dimensional omics space. The samples in the sample set share common clinical measurements, which we refer to as "clinotypes," such as age group, gender, treatment status, or survival days. We demonstrate how SEAS yields insights into biological data sets using glioblastoma (GBM) samples. Notably, when analyzing the combined The Cancer Genome Atlas (TCGA)-patient-derived xenograft (PDX) data, SEAS allows approximating the different clinical outcomes of radiotherapy-treated PDX samples, which has not been solved by other tools. The result shows that SEAS may support the clinical decision. The SEAS tool is publicly available as a freely available software package at https://aimed-lab.shinyapps.io/SEAS/.

13.
Methods Mol Biol ; 2328: 139-151, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34251623

RESUMO

In this book chapter, we introduce a pipeline to mine significant biomedical entities (or bioentities) in biological networks. Our focus is on prioritizing both bioentities themselves and the associations between bioentities in order to reveal their biological functions. We will introduce three tools BEERE, WIPER, and PAGER 2.0 that can be used together for network analysis and function interpretation: (1) BEERE is a network analysis tool for "Biomedical Entity Expansion, Ranking and Explorations," (2) WIPER is an entity-to-entity association ranking tool, and (3) PAGER 2.0 is a service for gene enrichment analysis.


Assuntos
Mineração de Dados/métodos , Redes Reguladoras de Genes/genética , Mapas de Interação de Proteínas/genética , Algoritmos , Humanos , Linguagens de Programação
14.
Leukemia ; 35(12): 3371-3382, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34120146

RESUMO

Leukemic stem cells (LSCs) can acquire non-mutational resistance following drug treatment leading to therapeutic failure and relapse. However, oncogene-independent mechanisms of drug persistence in LSCs are incompletely understood, which is the primary focus of this study. We integrated proteomics, transcriptomics, and metabolomics to determine the contribution of STAT3 in promoting metabolic changes in tyrosine kinase inhibitor (TKI) persistent chronic myeloid leukemia (CML) cells. Proteomic and transcriptional differences in TKI persistent CML cells revealed BCR-ABL-independent STAT3 activation in these cells. While knockout of STAT3 inhibited the CML cells from developing drug-persistence, inhibition of STAT3 using a small molecule inhibitor sensitized the persistent CML cells to TKI treatment. Interestingly, given the role of phosphorylated STAT3 as a transcription factor, it localized uniquely to genes regulating metabolic pathways in the TKI-persistent CML stem and progenitor cells. Subsequently, we observed that STAT3 dysregulated mitochondrial metabolism forcing the TKI-persistent CML cells to depend on glycolysis, unlike TKI-sensitive CML cells, which are more reliant on oxidative phosphorylation. Finally, targeting pyruvate kinase M2, a rate-limiting glycolytic enzyme, specifically eradicated the TKI-persistent CML cells. By exploring the role of STAT3 in altering metabolism, we provide critical insight into identifying potential therapeutic targets for eliminating TKI-persistent LSCs.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Metaboloma , Células-Tronco Neoplásicas/efeitos dos fármacos , Fator de Transcrição STAT3/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Transcriptoma , Animais , Apoptose , Feminino , Glicólise , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Masculino , Camundongos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Inibidores de Proteínas Quinases/farmacologia , Fator de Transcrição STAT3/genética
15.
Nucleic Acids Res ; 49(D1): D589-D599, 2021 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-33245774

RESUMO

PAGER-CoV (http://discovery.informatics.uab.edu/PAGER-CoV/) is a new web-based database that can help biomedical researchers interpret coronavirus-related functional genomic study results in the context of curated knowledge of host viral infection, inflammatory response, organ damage, and tissue repair. The new database consists of 11 835 PAGs (Pathways, Annotated gene-lists, or Gene signatures) from 33 public data sources. Through the web user interface, users can search by a query gene or a query term and retrieve significantly matched PAGs with all the curated information. Users can navigate from a PAG of interest to other related PAGs through either shared PAG-to-PAG co-membership relationships or PAG-to-PAG regulatory relationships, totaling 19 996 993. Users can also retrieve enriched PAGs from an input list of COVID-19 functional study result genes, customize the search data sources, and export all results for subsequent offline data analysis. In a case study, we performed a gene set enrichment analysis (GSEA) of a COVID-19 RNA-seq data set from the Gene Expression Omnibus database. Compared with the results using the standard PAGER database, PAGER-CoV allows for more sensitive matching of known immune-related gene signatures. We expect PAGER-CoV to be invaluable for biomedical researchers to find molecular biology mechanisms and tailored therapeutics to treat COVID-19 patients.


Assuntos
Algoritmos , COVID-19/prevenção & controle , Biologia Computacional/métodos , Coronavirus/genética , Bases de Dados Genéticas , SARS-CoV-2/genética , COVID-19/epidemiologia , COVID-19/virologia , Coronavirus/metabolismo , Curadoria de Dados/métodos , Epidemias , Redes Reguladoras de Genes , Humanos , Armazenamento e Recuperação da Informação/métodos , Internet , Anotação de Sequência Molecular/métodos , SARS-CoV-2/metabolismo , SARS-CoV-2/fisiologia , Interface Usuário-Computador
16.
Artigo em Inglês | MEDLINE | ID: mdl-31406955

RESUMO

PURPOSE: As a tumor immunotherapy, allogeneic hematopoietic cell transplantation with subsequent donor lymphocyte injection (DLI) aims to induce the graft-versus-tumor (GVT) effect but often also leads to acute graft-versus-host disease (GVHD). Plasma tests that can predict the likelihood of GVT without GVHD are still needed. PATIENTS AND METHODS: We first used an intact-protein analysis system to profile the plasma proteome post-DLI of patients who experienced GVT and acute GVHD for comparison with the proteome of patients who experienced GVT without GVHD in a training set. Our novel six-step systems biology analysis involved removing common proteins and GVHD-specific proteins, creating a protein-protein interaction network, calculating relevance and penalty scores, and visualizing candidate biomarkers in gene networks. We then performed a second proteomics experiment in a validation set of patients who experienced GVT without acute GVHD after DLI for comparison with the proteome of patients before DLI. We next combined the two experiments to define a biologically relevant signature of GVT without GVHD. An independent experiment with single-cell profiling in tumor antigen-activated T cells from a patient with post-hematopoietic cell transplantation relapse was performed. RESULTS: The approach provided a list of 46 proteins in the training set, and 30 proteins in the validation set were associated with GVT without GVHD. The combination of the two experiments defined a unique 61-protein signature of GVT without GVHD. Finally, the single-cell profiling in activated T cells found 43 of the 61 genes. Novel markers, such as RPL23, ILF2, CD58, and CRTAM, were identified and could be extended to other antitumoral responses. CONCLUSION: Our multiomic analysis provides, to our knowledge, the first human plasma signature for GVT without GVHD. Risk stratification on the basis of this signature would allow for customized treatment plans.

17.
Nucleic Acids Res ; 47(W1): W578-W586, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31114876

RESUMO

BEERE (Biomedical Entity Expansion, Ranking and Explorations) is a new web-based data analysis tool to help biomedical researchers characterize any input list of genes/proteins, biomedical terms or their combinations, i.e. 'biomedical entities', in the context of existing literature. Specifically, BEERE first aims to help users examine the credibility of known entity-to-entity associative or semantic relationships supported by database or literature references from the user input of a gene/term list. Then, it will help users uncover the relative importance of each entity-a gene or a term-within the user input by computing the ranking scores of all entities. At last, it will help users hypothesize new gene functions or genotype-phenotype associations by an interactive visual interface of constructed global entity relationship network. The output from BEERE includes: a list of the original entities matched with known relationships in databases; any expanded entities that may be generated from the analysis; the ranks and ranking scores reported with statistical significance for each entity; and an interactive graphical display of the gene or term network within data provenance annotations that link to external data sources. The web server is free and open to all users with no login requirement and can be accessed at http://discovery.informatics.uab.edu/beere/.


Assuntos
Genes , Proteínas , Software , Mineração de Dados , Bases de Dados Genéticas , Internet , Mapeamento de Interação de Proteínas , PubMed
18.
Quant Biol ; 7(4): 313-326, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38525413

RESUMO

Background: In network biology researchers generate biomolecular networks with candidate genes or proteins experimentally-derived from high-throughput data and known biomolecular associations. Current bioinformatics research focuses on characterizing candidate genes/proteins, or nodes, with network characteristics, e.g., betweenness centrality. However, there have been few research reports to characterize and prioritize biomolecular associations ("edges"), which can represent gene regulatory events essential to biological processes. Method: We developed Weighted In-Path Edge Ranking (WIPER), a new computational algorithm which can help evaluate all biomolecular interactions/associations ("edges") in a network model and generate a rank order of every edge based on their in-path traversal scores and statistical significance test result. To validate whether WIPER worked as we designed, we tested the algorithm on synthetic network models. Results: Our results showed WIPER can reliably discover both critical "well traversed in-path edges", which are statistically more traversed than normal edges, and "peripheral in-path edges", which are less traversed than normal edges. Compared with other simple measures such as betweenness centrality, WIPER provides better biological interpretations. In the case study of analyzing postanal pig hearts gene expression, WIPER highlighted new signaling pathways suggestive of cardiomyocyte regeneration and proliferation. In the case study of Alzheimer's disease genetic disorder association, WIPER reports SRC:APP, AR:APP, APP:FYN, and APP:NES edges (gene-gene associations) both statistically and biologically important from PubMed co-citation. Conclusion: We believe that WIPER will become an essential software tool to help biologists discover and validate essential signaling/regulatory events from high-throughput biology data in the context of biological networks. Availability: The free WIPER API is described at discovery.informatics.uab.edu/wiper/.

19.
IEEE/ACM Trans Comput Biol Bioinform ; 15(6): 1991-1998, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30040650

RESUMO

In this article, we present a computational framework to identify "causal relationships" among super gene sets. For "causal relationships," we refer to both stimulatory and inhibitory regulatory relationships, regardless of through direct or indirect mechanisms. For super gene sets, we refer to "pathways, annotated lists, and gene signatures," or PAGs. To identify causal relationships among PAGs, we extend the previous work on identifying PAG-to-PAG regulatory relationships by further requiring them to be significantly enriched with gene-to-gene co-expression pairs across the two PAGs involved. This is achieved by developing a quantitative metric based on PAG-to-PAG Co-expressions (PPC), which we use to infer the likelihood that PAG-to-PAG relationships under examination are causal-either stimulatory or inhibitory. Since true causal relationships are unknown, we approximate the overall performance of inferring causal relationships with the performance of recalling known r-type PAG-to-PAG relationships from causal PAG-to-PAG inference, using a functional genomics benchmark dataset from the GEO database. We report the area-under-curve (AUC) performance for both precision and recall being 0.81. By applying our framework to a myeloid-derived suppressor cells (MDSC) dataset, we further demonstrate that this framework is effective in helping build multi-scale biomolecular systems models with new insights on regulatory and causal links for downstream biological interpretations.


Assuntos
Redes Reguladoras de Genes/genética , Genômica/métodos , Biologia de Sistemas/métodos , Algoritmos , Animais , Linhagem Celular Tumoral , Bases de Dados Genéticas , Perfilação da Expressão Gênica , Humanos , Camundongos
20.
Nucleic Acids Res ; 46(D1): D668-D676, 2018 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-29126216

RESUMO

Integrative Gene-set, Network and Pathway Analysis (GNPA) is a powerful data analysis approach developed to help interpret high-throughput omics data. In PAGER 1.0, we demonstrated that researchers can gain unbiased and reproducible biological insights with the introduction of PAGs (Pathways, Annotated-lists and Gene-signatures) as the basic data representation elements. In PAGER 2.0, we improve the utility of integrative GNPA by significantly expanding the coverage of PAGs and PAG-to-PAG relationships in the database, defining a new metric to quantify PAG data qualities, and developing new software features to simplify online integrative GNPA. Specifically, we included 84 282 PAGs spanning 24 different data sources that cover human diseases, published gene-expression signatures, drug-gene, miRNA-gene interactions, pathways and tissue-specific gene expressions. We introduced a new normalized Cohesion Coefficient (nCoCo) score to assess the biological relevance of genes inside a PAG, and RP-score to rank genes and assign gene-specific weights inside a PAG. The companion web interface contains numerous features to help users query and navigate the database content. The database content can be freely downloaded and is compatible with third-party Gene Set Enrichment Analysis tools. We expect PAGER 2.0 to become a major resource in integrative GNPA. PAGER 2.0 is available at http://discovery.informatics.uab.edu/PAGER/.


Assuntos
Bases de Dados Genéticas , Redes Reguladoras de Genes , Genoma Humano , Transcriptoma , Epistasia Genética , Perfilação da Expressão Gênica , Ontologia Genética , Humanos , Armazenamento e Recuperação da Informação , Interface Usuário-Computador
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA