Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Ano de publicação
Tipo de documento
Intervalo de ano de publicação
1.
NPJ Syst Biol Appl ; 10(1): 92, 2024 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-39169016

RESUMO

Complex signaling pathways are believed to be responsible for drug resistance. Drug combinations perturbing multiple signaling targets have the potential to reduce drug resistance. The large-scale multi-omic datasets and experimental drug combination synergistic score data are valuable resources to study mechanisms of synergy (MoS) to guide the development of precision drug combinations. However, signaling patterns of MoS are complex and remain unclear, and thus it is challenging to identify synergistic drug combinations in clinical. Herein, we proposed a novel integrative and interpretable graph AI model, DeepSignalingFlow, to uncover the MoS by integrating and mining multi-omic data. The major innovation is that we uncover MoS by modeling the signaling flow from multi-omic features of essential disease proteins to the drug targets, which has not been introduced by the existing models. The model performance was assessed utilizing four distinct drug combination synergy evaluation datasets, i.e., NCI ALMANAC, O'Neil, DrugComb, and DrugCombDB. The comparison results showed that the proposed model outperformed existing graph AI models in terms of synergy score prediction, and can interpret MoS using the core signaling flows. The code is publicly accessible via Github: https://github.com/FuhaiLiAiLab/DeepSignalingFlow.


Assuntos
Sinergismo Farmacológico , Transdução de Sinais , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Humanos , Biologia Computacional/métodos
2.
bioRxiv ; 2024 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-39149314

RESUMO

Generative pretrained models represent a significant advancement in natural language processing and computer vision, which can generate coherent and contextually relevant content based on the pre-training on large general datasets and fine-tune for specific tasks. Building foundation models using large scale omic data is promising to decode and understand the complex signaling language patterns within cells. Different from existing foundation models of omic data, we build a foundation model, mosGraphGPT , for multi-omic signaling (mos) graphs, in which the multi-omic data was integrated and interpreted using a multi-level signaling graph. The model was pretrained using multi-omic data of cancers in The Cancer Genome Atlas (TCGA), and fine-turned for multi-omic data of Alzheimer's Disease (AD). The experimental evaluation results showed that the model can not only improve the disease classification accuracy, but also is interpretable by uncovering disease targets and signaling interactions. And the model code are uploaded via GitHub with link: https://github.com/mosGraph/mosGraphGPT.

3.
Front Cell Neurosci ; 18: 1369242, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38846640

RESUMO

Recently, large-scale scRNA-seq datasets have been generated to understand the complex signaling mechanisms within the microenvironment of Alzheimer's Disease (AD), which are critical for identifying novel therapeutic targets and precision medicine. However, the background signaling networks are highly complex and interactive. It remains challenging to infer the core intra- and inter-multi-cell signaling communication networks using scRNA-seq data. In this study, we introduced a novel graph transformer model, PathFinder, to infer multi-cell intra- and inter-cellular signaling pathways and communications among multi-cell types. Compared with existing models, the novel and unique design of PathFinder is based on the divide-and-conquer strategy. This model divides complex signaling networks into signaling paths, which are then scored and ranked using a novel graph transformer architecture to infer intra- and inter-cell signaling communications. We evaluated the performance of PathFinder using two scRNA-seq data cohorts. The first cohort is an APOE4 genotype-specific AD, and the second is a human cirrhosis cohort. The evaluation confirms the promising potential of using PathFinder as a general signaling network inference model.

4.
bioRxiv ; 2024 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-38798349

RESUMO

Multi-omic data, i.e., genomics, epigenomics, transcriptomics, proteomics, characterize cellular complex signaling systems from multi-level and multi-view and provide a holistic view of complex cellular signaling pathways. However, it remains challenging to integrate and interpret multi-omics data. Graph neural network (GNN) AI models have been widely used to analyze graph-structure datasets and are ideal for integrative multi-omics data analysis because they can naturally integrate and represent multi-omics data as a biologically meaningful multi-level signaling graph and interpret multi-omics data by node and edge ranking analysis for signaling flow/cascade inference. However, it is non-trivial for graph-AI model developers to pre-analyze multi-omics data and convert them into graph-structure data for individual samples, which can be directly fed into graph-AI models. To resolve this challenge, we developed mosGraphGen (multi-omics signaling graph generator), a novel computational tool that generates multi-omics signaling graphs of individual samples by mapping the multi-omics data onto a biologically meaningful multi-level background signaling network. With mosGraphGen, AI model developers can directly apply and evaluate their models using these mos-graphs. We evaluated the mosGraphGen using both multi-omics datasets of cancer and Alzheimer's disease (AD) samples. The code of mosGraphGen is open-source and publicly available via GitHub: https://github.com/Multi-OmicGraphBuilder/mosGraphGen.

5.
BMC Cardiovasc Disord ; 24(1): 278, 2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38811882

RESUMO

BACKGROUND: Left ventricular thrombus (LVT) is a serious complication after myocardial infarction. However, due to its asymptomatic nature, early detection is challenging. We aimed to explore the differences in clinical correlates of LVT found in acute to subacute and chronic phases of myocardial infarction. METHODS: We collected data from 153 patients who were diagnosed with LVT after myocardial infarction at the Affiliated Hospital of Qingdao University from January 2013 to December 2022. Baseline information, inflammatory markers, transthoracic echocardiograph (TTE) data and other clinical correlates were collected. Patients were categorized into acute to subacute phase group (< 30 days) and chronic phase group (30 days and after) according to the time at which echocardiograph was performed. The resolution of thrombus within 90 days is regarded as the primary endpoint event. We fitted logistic regression models to relating clinical correlates with phase-specific thrombus resolution. RESULTS: For acute to subacute phase thrombus patients: C-reactive protein levels (OR: 0.95, 95% CI: 0.918-0.983, p = 0.003) were significantly associated with thrombus resolution. For chronic phase thrombus patients: anticoagulant treatment was associated with 5.717-fold odds of thrombus resolution (OR: 5.717, 95% CI: 1.543-21.18, p = 0.009). CONCLUSIONS: Higher levels of CRP were associated with lower likelihood of LVT resolution in acute phase myocardial infarction; Anticoagulant therapy is still needed for thrombus in the chronic stage of myocardial infarction.


Assuntos
Trombose , Humanos , Masculino , Feminino , Pessoa de Meia-Idade , Fatores de Tempo , Trombose/diagnóstico por imagem , Trombose/etiologia , Idoso , Fatores de Risco , Anticoagulantes/uso terapêutico , Proteína C-Reativa/análise , Proteína C-Reativa/metabolismo , Estudos Retrospectivos , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/diagnóstico , Biomarcadores/sangue , Resultado do Tratamento , Cardiopatias/diagnóstico por imagem , Cardiopatias/etiologia , Cardiopatias/diagnóstico , Ventrículos do Coração/diagnóstico por imagem , Ventrículos do Coração/fisiopatologia , China , Ecocardiografia , Função Ventricular Esquerda
6.
Mol Neurodegener ; 19(1): 1, 2024 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-38172904

RESUMO

Triggering receptor expressed on myeloid cells 2 (TREM2) plays a critical role in microglial activation, survival, and apoptosis, as well as in Alzheimer's disease (AD) pathogenesis. We previously reported the MS4A locus as a key modulator for soluble TREM2 (sTREM2) in cerebrospinal fluid (CSF). To identify additional novel genetic modifiers of sTREM2, we performed the largest genome-wide association study (GWAS) and identified four loci for CSF sTREM2 in 3,350 individuals of European ancestry. Through multi-ethnic fine mapping, we identified two independent missense variants (p.M178V in MS4A4A and p.A112T in MS4A6A) that drive the association in MS4A locus and showed an epistatic effect for sTREM2 levels and AD risk. The novel TREM2 locus on chr 6 contains two rare missense variants (rs75932628 p.R47H, P=7.16×10-19; rs142232675 p.D87N, P=2.71×10-10) associated with sTREM2 and AD risk. The third novel locus in the TGFBR2 and RBMS3 gene region (rs73823326, P=3.86×10-9) included a regulatory variant with a microglia-specific chromatin loop for the promoter of TGFBR2. Using cell-based assays we demonstrate that overexpression and knock-down of TGFBR2, but not RBMS3, leads to significant changes of sTREM2. The last novel locus is located on the APOE region (rs11666329, P=2.52×10-8), but we demonstrated that this signal was independent of APOE genotype. This signal colocalized with cis-eQTL of NECTIN2 in the brain cortex and cis-pQTL of NECTIN2 in CSF. Overexpression of NECTIN2 led to an increase of sTREM2 supporting the genetic findings. To our knowledge, this is the largest study to date aimed at identifying genetic modifiers of CSF sTREM2. This study provided novel insights into the MS4A and TREM2 loci, two well-known AD risk genes, and identified TGFBR2 and NECTIN2 as additional modulators involved in TREM2 biology.


Assuntos
Doença de Alzheimer , Humanos , Doença de Alzheimer/patologia , Receptor do Fator de Crescimento Transformador beta Tipo II/genética , Estudo de Associação Genômica Ampla , Microglia/patologia , Apolipoproteínas E/genética , Biomarcadores/líquido cefalorraquidiano , Glicoproteínas de Membrana/genética , Receptores Imunológicos/genética
7.
bioRxiv ; 2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-38293243

RESUMO

Recently, large-scale scRNA-seq datasets have been generated to understand the complex and poorly understood signaling mechanisms within microenvironment of Alzheimer's Disease (AD), which are critical for identifying novel therapeutic targets and precision medicine. Though a set of targets have been identified, however, it remains a challenging to infer the core intra- and inter-multi-cell signaling communication networks using the scRNA-seq data, considering the complex and highly interactive background signaling network. Herein, we introduced a novel graph transformer model, PathFinder, to infer multi-cell intra- and inter-cellular signaling pathways and signaling communications among multi-cell types. Compared with existing models, the novel and unique design of PathFinder is based on the divide-and-conquer strategy, which divides the complex signaling networks into signaling paths, and then score and rank them using a novel graph transformer architecture to infer the intra- and inter-cell signaling communications. We evaluated PathFinder using scRNA-seq data of APOE4-genotype specific AD mice models and identified novel APOE4 altered intra- and inter-cell interaction networks among neurons, astrocytes, and microglia. PathFinder is a general signaling network inference model and can be applied to other omics data-driven signaling network inference.

8.
PLoS Comput Biol ; 20(1): e1011785, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38181047

RESUMO

Single-cell RNA sequencing (scRNA-seq) is a powerful technology to investigate the transcriptional programs in stromal, immune, and disease cells, like tumor cells or neurons within the Alzheimer's Disease (AD) brain or tumor microenvironment (ME) or niche. Cell-cell communications within ME play important roles in disease progression and immunotherapy response and are novel and critical therapeutic targets. Though many tools of scRNA-seq analysis have been developed to investigate the heterogeneity and sub-populations of cells, few were designed for uncovering cell-cell communications of ME and predicting the potentially effective drugs to inhibit the communications. Moreover, the data analysis processes of discovering signaling communication networks and effective drugs using scRNA-seq data are complex and involve a set of critical analysis processes and external supportive data resources, which are difficult for researchers who have no strong computational background and training in scRNA-seq data analysis. To address these challenges, in this study, we developed a novel open-source computational tool, sc2MeNetDrug (https://fuhaililab.github.io/sc2MeNetDrug/). It was specifically designed using scRNA-seq data to identify cell types within disease MEs, uncover the dysfunctional signaling pathways within individual cell types and interactions among different cell types, and predict effective drugs that can potentially disrupt cell-cell signaling communications. sc2MeNetDrug provided a user-friendly graphical user interface to encapsulate the data analysis modules, which can facilitate the scRNA-seq data-based discovery of novel inter-cell signaling communications and novel therapeutic regimens.


Assuntos
Análise de Célula Única , Software , RNA-Seq , Análise de Sequência de RNA , Perfilação da Expressão Gênica , Transdução de Sinais/genética
9.
Europace ; 26(1)2023 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-38099508

RESUMO

AIMS: Patients with heart failure with preserved ejection fraction (HFpEF) and atrial fibrillation (AF) have worse clinical outcomes than those with sinus rhythm (SR). We aim to investigate whether maintaining SR in patients with HFpEF through a strategy such as AF ablation would improve outcomes. METHODS AND RESULTS: This is a cohort study that analysed 1034 patients (median age 69 [63-76] years, 46.2% [478/1034] female) with HFpEF and AF. Of these, 392 patients who underwent first-time AF ablation were assigned to the ablation group, and the remaining 642 patients, who received only medical therapy, were assigned to the no ablation group. The primary endpoint was a composite of all-cause death or rehospitalization for worsening heart failure. After a median follow-up of 39 months, the cumulative incidence of the primary endpoint was significantly lower in the ablation group compared to the no ablation group (adjusted hazard ratio [HR], 0.55 [95% CI, 0.37-0.82], P = 0.003) in the propensity score-matched model. Secondary endpoint analysis showed that the benefit of AF ablation was mainly driven by a reduction in rehospitalization for worsening heart failure (adjusted HR, 0.52 [95% CI, 0.34-0.80], P = 0.003). Patients in the ablation group showed a 33% relative decrease in atrial tachycardia/AF recurrence compared to the no ablation group (adjusted HR, 0.67 [95% CI, 0.54-0.84], P < 0.001). CONCLUSION: Among patients with HFpEF and AF, the strategy of AF ablation to maintain SR was associated with a lower risk of the composite outcome of all-cause death or rehospitalization for worsening heart failure.


Assuntos
Fibrilação Atrial , Insuficiência Cardíaca , Humanos , Feminino , Idoso , Fibrilação Atrial/diagnóstico , Fibrilação Atrial/cirurgia , Fibrilação Atrial/complicações , Estudos de Coortes , Volume Sistólico/fisiologia , Insuficiência Cardíaca/diagnóstico , Insuficiência Cardíaca/cirurgia , Insuficiência Cardíaca/complicações , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA