Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 7.224
Filtrer
1.
Methods Mol Biol ; 2856: 241-262, 2025.
Article de Anglais | MEDLINE | ID: mdl-39283456

RÉSUMÉ

Single-cell Hi-C (scHi-C) is a collection of protocols for studying genomic interactions within individual cells. Although data analysis for scHi-C resembles data analysis for bulk Hi-C, the unique challenges of scHi-C, such as high noise and protocol-specific biases, require specialized data processing strategies. In this tutorial chapter, we focus on using pairtools, a suite of tools optimized for scHi-C data, demonstrating its application on a Drosophila snHi-C dataset. While centered on pairtools for snHi-C data, the principles outlined are applicable across scHi-C variants with minor adjustments. This educational chapter aims to guide researchers in using open-source tools for scHi-C analysis, emphasizing critical steps of contact pair extraction, detection of ligation junctions, filtration, and deduplication.


Sujet(s)
Génomique , Analyse sur cellule unique , Logiciel , Flux de travaux , Analyse sur cellule unique/méthodes , Animaux , Génomique/méthodes , Drosophila/génétique , Séquençage nucléotidique à haut débit/méthodes , Biologie informatique/méthodes
2.
Methods Mol Biol ; 2856: 263-268, 2025.
Article de Anglais | MEDLINE | ID: mdl-39283457

RÉSUMÉ

We describe an approach for reconstructing three-dimensional (3D) structures from single-cell Hi-C data. This approach has been inspired by a method of recurrence plots and visualization tools for nonlinear time series data. Some examples are also presented.


Sujet(s)
Analyse sur cellule unique , Analyse sur cellule unique/méthodes , Imagerie tridimensionnelle/méthodes , Humains , Logiciel , Chromosomes/génétique , Algorithmes
3.
Methods Mol Biol ; 2854: 83-91, 2025.
Article de Anglais | MEDLINE | ID: mdl-39192121

RÉSUMÉ

Transcriptomics is an extremely important area of molecular biology and is a powerful tool for studying all RNA molecules in an organism. Conventional transcriptomic technologies include microarrays and RNA sequencing, and the rapid development of single-cell sequencing and spatial transcriptomics in recent years has provided an enormous scope for research in this field. This chapter describes the application, significance, and experimental procedures of a variety of transcriptomic technologies in antiviral natural immunity.


Sujet(s)
Analyse de profil d'expression de gènes , Immunité innée , Transcriptome , Immunité innée/génétique , Humains , Analyse de profil d'expression de gènes/méthodes , Animaux , Maladies virales/immunologie , Maladies virales/génétique , Analyse de séquence d'ARN/méthodes , Analyse sur cellule unique/méthodes , Séquençage par oligonucléotides en batterie/méthodes
4.
Methods Mol Biol ; 2848: 85-103, 2025.
Article de Anglais | MEDLINE | ID: mdl-39240518

RÉSUMÉ

Recent technological advances in single-cell RNA sequencing (scRNA-Seq) have enabled scientists to answer novel questions in biology with unparalleled precision. Indeed, in the field of ocular development and regeneration, scRNA-Seq studies have resulted in a number of exciting discoveries that have begun to revolutionize the way we think about these processes. Despite the widespread success of scRNA-Seq, many scientists are wary to perform scRNA-Seq experiments due to the uncertainty of obtaining high-quality viable cell populations that are necessary for the generation of usable data that enable rigorous computational analyses. Here, we describe methodology to reproducibility generate high-quality single-cell suspensions from embryonic zebrafish eyes. These single-cell suspensions served as inputs to the 10× Genomics v3.1 system and yielded high-quality scRNA-Seq data in proof-of-principle studies. In describing methodology to quantitatively assess cell yields, cell viability, and other critical quality control parameters, this protocol can serve as a useful starting point for others in designing their scRNA-Seq experiments in the zebrafish eye and in other developing or regenerating tissues in zebrafish or other model systems.


Sujet(s)
Rétine , Analyse de séquence d'ARN , Analyse sur cellule unique , Danio zébré , Animaux , Danio zébré/génétique , Danio zébré/embryologie , Analyse sur cellule unique/méthodes , Rétine/cytologie , Rétine/embryologie , Rétine/métabolisme , Analyse de séquence d'ARN/méthodes , Séparation cellulaire/méthodes
5.
Methods Mol Biol ; 2848: 105-116, 2025.
Article de Anglais | MEDLINE | ID: mdl-39240519

RÉSUMÉ

The generation of quality data from a single-nucleus profiling experiment requires nuclei to be isolated from tissues in a gentle and efficient manner. Nuclei isolation must be carefully optimized across tissue types to preserve nuclear architecture, prevent nucleic acid degradation, and remove unwanted contaminants. Here, we present an optimized workflow for generating a single-nucleus suspension from ocular tissues of the embryonic chicken that is compatible with various downstream workflows. The described protocol enables the rapid isolation of a high yield of aggregate-free nuclei from the embryonic chicken eye without compromising nucleic acid integrity, and the nuclei suspension is compatible with single-nucleus RNA and ATAC sequencing. We detail several stopping points, either via cryopreservation or fixation, to enhance workflow adaptability. Further, we provide a guide through multiple QC points and demonstrate proof-of-principle using two commercially available kits. Finally, we demonstrate that existing in silico genotyping methods can be adopted to computationally derive biological replicates from a single pool of chicken nuclei, greatly reducing the cost of biological replication and allowing researchers to consider sex as a variable during analysis. Together, this tutorial represents a cost-effective, simple, and effective approach to single-nucleus profiling of embryonic chicken eye tissues and is likely to be easily modified to be compatible with similar tissue types.


Sujet(s)
Noyau de la cellule , Poulets , Analyse sur cellule unique , Animaux , Noyau de la cellule/métabolisme , Noyau de la cellule/génétique , Embryon de poulet , Analyse sur cellule unique/méthodes , Oeil/embryologie , Oeil/métabolisme , Cryoconservation/méthodes , Séquençage après immunoprécipitation de la chromatine/méthodes
6.
Methods Mol Biol ; 2848: 117-134, 2025.
Article de Anglais | MEDLINE | ID: mdl-39240520

RÉSUMÉ

Retinal degenerative diseases including age-related macular degeneration and glaucoma are estimated to currently affect more than 14 million people in the United States, with an increased prevalence of retinal degenerations in aged individuals. An expanding aged population who are living longer forecasts an increased prevalence and economic burden of visual impairments. Improvements to visual health and treatment paradigms for progressive retinal degenerations slow vision loss. However, current treatments fail to remedy the root cause of visual impairments caused by retinal degenerations-loss of retinal neurons. Stimulation of retinal regeneration from endogenous cellular sources presents an exciting treatment avenue for replacement of lost retinal cells. In multiple species including zebrafish and Xenopus, Müller glial cells maintain a highly efficient regenerative ability to reconstitute lost cells throughout the organism's lifespan, highlighting potential therapeutic avenues for stimulation of retinal regeneration in humans. Here, we describe how the application of single-cell RNA-sequencing (scRNA-seq) has enhanced our understanding of Müller glial cell-derived retinal regeneration, including the characterization of gene regulatory networks that facilitate/inhibit regenerative responses. Additionally, we provide a validated experimental framework for cellular preparation of mouse retinal cells as input into scRNA-seq experiments, including insights into experimental design and analyses of resulting data.


Sujet(s)
Cellules épendymogliales , Rétine , Analyse sur cellule unique , Animaux , Souris , Analyse sur cellule unique/méthodes , Rétine/métabolisme , Cellules épendymogliales/métabolisme , Régénération/génétique , Analyse de séquence d'ARN/méthodes , Dégénérescence de la rétine/génétique , Dégénérescence de la rétine/thérapie , RNA-Seq/méthodes , Modèles animaux de maladie humaine
7.
Front Immunol ; 15: 1419126, 2024.
Article de Anglais | MEDLINE | ID: mdl-39234248

RÉSUMÉ

Background: Bladder cancer, a highly fatal disease, poses a significant threat to patients. Positioned at 19q13.2-13.3, LIG1, one of the four DNA ligases in mammalian cells, is frequently deleted in tumour cells of diverse origins. Despite this, the precise involvement of LIG1 in BLCA remains elusive. This pioneering investigation delves into the uncharted territory of LIG1's impact on BLCA. Our primary objective is to elucidate the intricate interplay between LIG1 and BLCA, alongside exploring its correlation with various clinicopathological factors. Methods: We retrieved gene expression data of para-carcinoma tissues and bladder cancer (BLCA) from the GEO repository. Single-cell sequencing data were processed using the "Seurat" package. Differential expression analysis was then performed with the "Limma" package. The construction of scale-free gene co-expression networks was achieved using the "WGCNA" package. Subsequently, a Venn diagram was utilized to extract genes from the positively correlated modules identified by WGCNA and intersect them with differentially expressed genes (DEGs), isolating the overlapping genes. The "STRINGdb" package was employed to establish the protein-protein interaction (PPI) network.Hub genes were identified through the PPI network using the Betweenness Centrality (BC) algorithm. We conducted KEGG and GO enrichment analyses to uncover the regulatory mechanisms and biological functions associated with the hub genes. A machine-learning diagnostic model was established using the R package "mlr3verse." Mutation profiles between the LIG1^high and LIG1^low groups were visualized using the BEST website. Survival analyses within the LIG1^high and LIG1^low groups were performed using the BEST website and the GENT2 website. Finally, a series of functional experiments were executed to validate the functional role of LIG1 in BLCA. Results: Our investigation revealed an upregulation of LIG1 in BLCA specimens, with heightened LIG1 levels correlating with unfavorable overall survival outcomes. Functional enrichment analysis of hub genes, as evidenced by GO and KEGG enrichment analyses, highlighted LIG1's involvement in critical function such as the DNA replication, cellular senescence, cell cycle and the p53 signalling pathway. Notably, the mutational landscape of BLCA varied significantly between LIG1high and LIG1low groups.Immune infiltrating analyses suggested a pivotal role for LIG1 in immune cell recruitment and immune regulation within the BLCA microenvironment, thereby impacting prognosis. Subsequent experimental validations further underscored the significance of LIG1 in BLCA pathogenesis, consolidating its functional relevance in BLCA samples. Conclusions: Our research demonstrates that LIG1 plays a crucial role in promoting bladder cancer malignant progression by heightening proliferation, invasion, EMT, and other key functions, thereby serving as a potential risk biomarker.


Sujet(s)
Marqueurs biologiques tumoraux , DNA ligase ATP , Apprentissage machine , Analyse sur cellule unique , Tumeurs de la vessie urinaire , Tumeurs de la vessie urinaire/génétique , Tumeurs de la vessie urinaire/mortalité , Tumeurs de la vessie urinaire/anatomopathologie , Humains , Analyse sur cellule unique/méthodes , Marqueurs biologiques tumoraux/génétique , DNA ligase ATP/génétique , DNA ligase ATP/métabolisme , Pronostic , Mâle , Régulation de l'expression des gènes tumoraux , Femelle , Réseaux de régulation génique , Cartes d'interactions protéiques , Adulte d'âge moyen , Analyse de profil d'expression de gènes , Biologie informatique/méthodes , Lignée cellulaire tumorale , Sujet âgé
9.
Clin Transl Med ; 14(9): e1818, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39308059

RÉSUMÉ

With rapid development and mature of single-cell measurements, single-cell biology and pathology become an emerging discipline to understand the disease. However, it is important to address concerns raised by clinicians as to how to apply single-cell measurements for clinical practice, translate the signals of single-cell systems biology into determination of clinical phenotype, and predict patient response to therapies. The present Perspective proposes a new system coined as the clinical artificial intelligent single-cell (caiSC) with the dynamic generator of clinical single-cell informatics, artificial intelligent analyzers, molecular multimodal reference boxes, clinical inputs and outs, and AI-based computerization. This system provides reliable and rapid information for impacting clinical diagnoses, monitoring, and prediction of the disease at the single-cell level. The caiSC represents an important step and milestone to translate the single-cell measurement into clinical application, assist clinicians' decision-making, and improve the quality of medical services. There is increasing evidence to support the possibility of the caiSC proposal, since the corresponding biotechnologies associated with caiSCs are rapidly developed. Therefore, we call the special attention and efforts from various scientists and clinicians on the caiSCs and believe that the appearance of the caiSCs can shed light on the future of clinical molecular medicine.


Sujet(s)
Intelligence artificielle , Analyse sur cellule unique , Analyse sur cellule unique/méthodes , Humains
10.
Brief Bioinform ; 25(6)2024 Sep 23.
Article de Anglais | MEDLINE | ID: mdl-39311699

RÉSUMÉ

The inference of gene regulatory networks (GRNs) is crucial to understanding the regulatory mechanisms that govern biological processes. GRNs may be represented as edges in a graph, and hence, it have been inferred computationally for scRNA-seq data. A wisdom of crowds approach to integrate edges from several GRNs to create one composite GRN has demonstrated improved performance when compared with individual algorithm implementations on bulk RNA-seq and microarray data. In an effort to extend this approach to scRNA-seq data, we present COFFEE (COnsensus single cell-type speciFic inFerence for gEnE regulatory networks), a Borda voting-based consensus algorithm that integrates information from 10 established GRN inference methods. We conclude that COFFEE has improved performance across synthetic, curated, and experimental datasets when compared with baseline methods. Additionally, we show that a modified version of COFFEE can be leveraged to improve performance on newer cell-type specific GRN inference methods. Overall, our results demonstrate that consensus-based methods with pertinent modifications continue to be valuable for GRN inference at the single cell level. While COFFEE is benchmarked on 10 algorithms, it is a flexible strategy that can incorporate any set of GRN inference algorithms according to user preference. A Python implementation of COFFEE may be found on GitHub: https://github.com/lodimk2/coffee.


Sujet(s)
Algorithmes , Réseaux de régulation génique , Analyse sur cellule unique , Analyse sur cellule unique/méthodes , Biologie informatique/méthodes , Humains , Logiciel
11.
Proc Natl Acad Sci U S A ; 121(40): e2402781121, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39312655

RÉSUMÉ

While considerable knowledge exists about the enzymes pivotal for C4 photosynthesis, much less is known about the cis-regulation important for specifying their expression in distinct cell types. Here, we use single-cell-indexed ATAC-seq to identify cell-type-specific accessible chromatin regions (ACRs) associated with C4 enzymes for five different grass species. This study spans four C4 species, covering three distinct photosynthetic subtypes: Zea mays and Sorghum bicolor (NADP-dependent malic enzyme), Panicum miliaceum (NAD-dependent malic enzyme), Urochloa fusca (phosphoenolpyruvate carboxykinase), along with the C3 outgroup Oryza sativa. We studied the cis-regulatory landscape of enzymes essential across all C4 species and those unique to C4 subtypes, measuring cell-type-specific biases for C4 enzymes using chromatin accessibility data. Integrating these data with phylogenetics revealed diverse co-option of gene family members between species, showcasing the various paths of C4 evolution. Besides promoter proximal ACRs, we found that, on average, C4 genes have two to three distal cell-type-specific ACRs, highlighting the complexity and divergent nature of C4 evolution. Examining the evolutionary history of these cell-type-specific ACRs revealed a spectrum of conserved and novel ACRs, even among closely related species, indicating ongoing evolution of cis-regulation at these C4 loci. This study illuminates the dynamic and complex nature of cis-regulatory elements evolution in C4 photosynthesis, particularly highlighting the intricate cis-regulatory evolution of key loci. Our findings offer a valuable resource for future investigations, potentially aiding in the optimization of C3 crop performance under changing climatic conditions.


Sujet(s)
Régulation de l'expression des gènes végétaux , Photosynthèse , Poaceae , Photosynthèse/génétique , Poaceae/génétique , Poaceae/métabolisme , Analyse sur cellule unique/méthodes , Chromatine/métabolisme , Chromatine/génétique , Oryza/génétique , Oryza/métabolisme , Phylogenèse , Zea mays/génétique , Zea mays/métabolisme , Malate dehydrogenase/métabolisme , Malate dehydrogenase/génétique , Protéines végétales/génétique , Protéines végétales/métabolisme , Sorghum/génétique , Sorghum/métabolisme
12.
JCI Insight ; 9(18)2024 Sep 24.
Article de Anglais | MEDLINE | ID: mdl-39315546

RÉSUMÉ

Therapies against cell-surface targets (CSTs) represent an emerging treatment class in solid malignancies. However, high-throughput investigations of CST expression across cancer types have been reliant on data sets of mostly primary tumors, despite therapeutic use most commonly in metastatic disease. We identified a total of 818 clinical trials of CST therapies with 78 CSTs. We assembled a data set spanning RNA-seq and microarrays in 7,927 benign samples, 16,866 primary tumor samples, and 6,124 metastatic tumor samples. We also utilized single-cell RNA-seq data from 36 benign tissues and 558 primary and metastatic tumor samples, and matched RNA versus protein expression in 29 benign tissue samples, 1,075 tumor samples, and 942 cell lines. High RNA expression accurately predicted high protein expression across CST therapies in benign tissues, tumor samples, and cell lines. We compared metastatic versus primary tumor expression, identified potential opportunities for repositioning, and matched cell lines to tumor types based on CST and global RNA expression. We evaluated single-cell heterogeneity across tumors, and identified rare normal cell subpopulations that may contribute to toxicity. Finally, we identified combinations of CST therapies for which bispecific approaches could improve tumor specificity. This study helps better define the landscape of CST expression in metastatic and primary cancers.


Sujet(s)
Métastase tumorale , Tumeurs , Humains , Tumeurs/anatomopathologie , Tumeurs/génétique , Lignée cellulaire tumorale , Analyse sur cellule unique/méthodes , Régulation de l'expression des gènes tumoraux , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , Thérapie moléculaire ciblée , RNA-Seq
13.
Sci Rep ; 14(1): 21751, 2024 09 18.
Article de Anglais | MEDLINE | ID: mdl-39294296

RÉSUMÉ

Gastric cancer (GC) is a prevalent malignancy with high mortality rates. Immunogenic cell death (ICD) is a unique form of programmed cell death that is closely linked to antitumor immunity and plays a critical role in modulating the tumor microenvironment (TME). Nevertheless, elucidating the precise effect of ICD on GC remains a challenging endeavour. ICD-related genes were identified in single-cell sequencing datasets and bulk transcriptome sequencing datasets via the AddModuleScore function, weighted gene co-expression network (WGCNA), and differential expression analysis. A robust signature associated with ICD was constructed using a machine learning computational framework incorporating 101 algorithms. Furthermore, multiomics analysis, including single-cell sequencing analysis, bulk transcriptomic analysis, and proteomics analysis, was conducted to verify the correlation of these hub genes with the immune microenvironment features of GC and with GC invasion and metastasis. We screened 59 genes associated with ICD and developed a robust ICD-related gene signature (ICDRS) via a machine learning computational framework that integrates 101 different algorithms. Furthermore, we identified five key hub genes (SMAP2, TNFAIP8, LBH, TXNIP, and PIK3IP1) from the ICDRS. Through single-cell analysis of GC tumor s, we confirmed the strong correlations of the hub genes with immune microenvironment features. Among these five genes, LBH exhibited the most significant associations with a poor prognosis and with the invasion and metastasis of GC. Finally, our findings were validated through immunohistochemical staining of a large clinical sample set, and the results further supported that LBH promotes GC cell invasion by activating the epithelial-mesenchymal transition (EMT) pathway.


Sujet(s)
Mort cellulaire immunogène , Apprentissage machine , Analyse sur cellule unique , Tumeurs de l'estomac , Microenvironnement tumoral , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/immunologie , Tumeurs de l'estomac/mortalité , Humains , Analyse sur cellule unique/méthodes , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/génétique , Régulation de l'expression des gènes tumoraux , Analyse de profil d'expression de gènes , Protéomique/méthodes , Transcriptome , Biologie informatique/méthodes , Réseaux de régulation génique , Multi-omique
14.
Bioinformatics ; 40(9)2024 Sep 02.
Article de Anglais | MEDLINE | ID: mdl-39226185

RÉSUMÉ

MOTIVATION: The growing number of single-cell RNA-seq (scRNA-seq) studies highlights the potential benefits of integrating multiple datasets, such as augmenting sample sizes and enhancing analytical robustness. Inherent diversity and batch discrepancies within samples or across studies continue to pose significant challenges for computational analyses. Questions persist in practice, lacking definitive answers: Should we use a specific integration method or opt for simply merging the datasets during joint analysis? Among all the existing data integration methods, which one is more suitable in specific scenarios? RESULT: To fill the gap, we introduce SCIntRuler, a novel statistical metric for guiding the integration of multiple scRNA-seq datasets. SCIntRuler helps researchers make informed decisions regarding the necessity of data integration and the selection of an appropriate integration method. Our simulations and real data applications demonstrate that SCIntRuler streamlines decision-making processes and facilitates the analysis of diverse scRNA-seq datasets under varying contexts, thereby alleviating the complexities associated with the integration of heterogeneous scRNA-seq datasets. AVAILABILITY AND IMPLEMENTATION: The implementation of our method is available on CRAN as an open-source R package with a user-friendly manual available: https://cloud.r-project.org/web/packages/SCIntRuler/index.html.


Sujet(s)
RNA-Seq , Analyse sur cellule unique , Analyse sur cellule unique/méthodes , RNA-Seq/méthodes , Logiciel , Humains , Analyse de séquence d'ARN/méthodes , Algorithmes , Biologie informatique/méthodes , Analyse de l'expression du gène de la cellule unique
16.
Front Immunol ; 15: 1438962, 2024.
Article de Anglais | MEDLINE | ID: mdl-39281674

RÉSUMÉ

γδ T-cells are a rare population of T-cells with both adaptive and innate-like properties. Despite their low prevalence, they have been found to be implicated various human diseases. γδ T-cell infiltration has been associated with improved clinical outcomes in solid cancers, prompting renewed interest in understanding their biology. To date, their biology remains elusive due to their low prevalence. The introduction of high-resolution single-cell sequencing has allowed various groups to characterize key effector subsets in various contexts, as well as begin to elucidate key regulatory mechanisms directing the differentiation and activity of these cells. In this review, we will review some of insights obtained from single-cell studies of γδ T-cells across various malignancies and highlight some important questions that remain unaddressed.


Sujet(s)
Tumeurs , Récepteur lymphocytaire T antigène, gamma-delta , Analyse sur cellule unique , Humains , Tumeurs/immunologie , Analyse sur cellule unique/méthodes , Récepteur lymphocytaire T antigène, gamma-delta/métabolisme , Récepteur lymphocytaire T antigène, gamma-delta/immunologie , Sous-populations de lymphocytes T/immunologie , Sous-populations de lymphocytes T/métabolisme , Animaux , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Microenvironnement tumoral/immunologie , Lymphocytes T/immunologie
17.
Int J Med Sci ; 21(12): 2348-2364, 2024.
Article de Anglais | MEDLINE | ID: mdl-39310264

RÉSUMÉ

Recent advancements have elucidated the multifaceted roles of the Schlafen (SLFN) family, including SLFN5, SLFN11, SLFN12, SLFN13, and SLFN14, which are implicated in immunological responses. However, little is known about the roles of this gene family in relation to malignancy development. The current study aimed to explore the diagnostic and prognostic potential of Schlafen family genes in colorectal adenocarcinoma (COAD) through bioinformatics analysis. Leveraging advanced bioinformatics tools of bulk RNA-sequencing and single-cell sequencing, we conducted in-depth analyses of gene expressions, functional enrichment, and survival patterns of patients with colorectal cancer compared to normal tissue. Among Schlafen family genes, the transcription levels of SLFN5 in COAD tissues were significantly elevated and correlated with poor survival outcomes. Furthermore, SLFN5 regulated the immune response via Janus kinase (JAK)/signal transduction and activator of transcription (STAT)/interferon (IFN)-alpha/beta signaling. These chemokines in inflammation are associated with diabetes and metabolism, suggesting their involvement in altered cellular energetics for COAD progress. In addition, an immune cell deconvolution analysis indicated a correlation between SLFN5 expression and immune-related cell populations, such as regulatory T cells (Tregs). These findings highlighted the potential clinical significance of SLFN5 in COAD and provided insights into its involvement in the tumor microenvironment and immune regulation. Meanwhile, the drug discovery data of SFLN5 with potential targeted small molecules suggested its therapeutic potential for COAD. Collectively, the current research demonstrated that SFLN5 play crucial roles in tumor development and serve as a prospective biomarker for COAD.


Sujet(s)
Tumeurs colorectales , Régulation de l'expression des gènes tumoraux , Analyse sur cellule unique , Humains , Tumeurs colorectales/génétique , Tumeurs colorectales/immunologie , Tumeurs colorectales/anatomopathologie , Analyse sur cellule unique/méthodes , Pronostic , Marqueurs biologiques tumoraux/génétique , Biologie informatique/méthodes , Analyse de séquence d'ARN , Adénocarcinome/génétique , Adénocarcinome/immunologie , Adénocarcinome/anatomopathologie , Adénocarcinome/mortalité , Analyse de profil d'expression de gènes , Transduction du signal/génétique , Transduction du signal/immunologie , Protéines du cycle cellulaire
18.
BMC Med ; 22(1): 383, 2024 Sep 12.
Article de Anglais | MEDLINE | ID: mdl-39267041

RÉSUMÉ

BACKGROUND: The development of the human vermiform appendix at the cellular level, as well as its function, is not well understood. Appendicitis in preschool children, although uncommon, is associated with a high perforation rate and increased morbidity. METHODS: We performed single-cell RNA sequencing (scRNA-seq) on the human appendix during fetal and pediatric stages as well as preschool-age inflammatory appendices. Transcriptional features of each cell compartment were discussed in the developing appendix. Cellular interactions and differentiation trajectories were also investigated. We compared scRNA-seq profiles from preschool appendicitis to those of matched healthy controls to reveal disease-associated changes. Bulk transcriptomic data, immunohistochemistry, and real-time quantitative PCR were used to validate the findings. RESULTS: Our analysis identified 76 cell types in total and described the cellular atlas of the developing appendix. We discovered the potential role of the BMP signaling pathway in appendiceal epithelium development and identified HOXC8 and PITX2 as the specific regulons of appendix goblet cells. Higher pericyte coverage, endothelial angiogenesis, and goblet mucus scores together with lower epithelial and endothelial tight junction scores were found in the preschool appendix, which possibly contribute to the clinical features of preschool appendicitis. Preschool appendicitis scRNA-seq profiles revealed that the interleukin-17 signaling pathway may participate in the inflammation process. CONCLUSIONS: Our study provides new insights into the development of the appendix and deepens the understanding of appendicitis in preschool children.


Sujet(s)
Appendicite , Appendice vermiforme , Analyse sur cellule unique , Humains , Appendicite/génétique , Appendicite/anatomopathologie , Enfant d'âge préscolaire , Analyse sur cellule unique/méthodes , Femelle , Mâle , Analyse de séquence d'ARN/méthodes , Nourrisson , Protéines à homéodomaine/génétique
19.
BMC Cancer ; 24(1): 1138, 2024 Sep 12.
Article de Anglais | MEDLINE | ID: mdl-39267056

RÉSUMÉ

PURPOSE: Lung adenocarcinoma (LUAD) significantly contributes to cancer-related mortality worldwide. The heterogeneity of the tumor immune microenvironment in LUAD results in varied prognoses and responses to immunotherapy among patients. Consequently, a clinical stratification algorithm is necessary and inevitable to effectively differentiate molecular features and tumor microenvironments, facilitating personalized treatment approaches. METHODS: We constructed a comprehensive single-cell transcriptional atlas using single-cell RNA sequencing data to reveal the cellular diversity of malignant epithelial cells of LUAD and identified a novel signature through a computational framework coupled with 10 machine learning algorithms. Our study further investigates the immunological characteristics and therapeutic responses associated with this prognostic signature and validates the predictive efficacy of the model across multiple independent cohorts. RESULTS: We developed a six-gene prognostic model (MYO1E, FEN1, NMI, ZNF506, ALDOA, and MLLT6) using the TCGA-LUAD dataset, categorizing patients into high- and low-risk groups. This model demonstrates robust performance in predicting survival across various LUAD cohorts. We observed distinct molecular patterns and biological processes in different risk groups. Additionally, analysis of two immunotherapy cohorts (N = 317) showed that patients with a high-risk signature responded more favorably to immunotherapy compared to those in the low-risk group. Experimental validation further confirmed that MYO1E enhances the proliferation and migration of LUAD cells. CONCLUSION: We have identified malignant cell-associated ligand-receptor subtypes in LUAD cells and developed a robust prognostic signature by thoroughly analyzing genomic, transcriptomic, and immunologic data. This study presents a novel method to assess the prognosis of patients with LUAD and provides insights into developing more effective immunotherapies.


Sujet(s)
Adénocarcinome pulmonaire , Tumeurs du poumon , Microenvironnement tumoral , Humains , Adénocarcinome pulmonaire/génétique , Adénocarcinome pulmonaire/anatomopathologie , Adénocarcinome pulmonaire/mortalité , Adénocarcinome pulmonaire/immunologie , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/mortalité , Pronostic , Microenvironnement tumoral/génétique , Microenvironnement tumoral/immunologie , Marqueurs biologiques tumoraux/génétique , Immunothérapie , Régulation de l'expression des gènes tumoraux , Analyse de profil d'expression de gènes , Femelle , Analyse sur cellule unique/méthodes , Mâle , Transcriptome , Apprentissage machine , Multi-omique
20.
Bioinformatics ; 40(9)2024 Sep 02.
Article de Anglais | MEDLINE | ID: mdl-39240328

RÉSUMÉ

SUMMARY: To address the challenges in single-cell metabolomics (SCM) research, we have developed an open-source Python-based modular library, named SCMeTA, for SCM data processing. We designed standardized pipeline and inter-container communication format and have developed modular components to adapt to the diverse needs of SCM studies. The validation was carried out on multiple SCM experiment data. The results demonstrated significant improvements in batch effects, accuracy of results, metabolic extraction rate, cell matching rate, as well as processing speed. This library is of great significance in advancing the practical application of SCM analysis and makes a foundation for wide-scale adoption in biological studies. AVAILABILITY AND IMPLEMENTATION: SCMeTA is freely available on https://github.com/SCMeTA/SCMeTA and https://doi.org/10.5281/zenodo.13569643.


Sujet(s)
Métabolomique , Analyse sur cellule unique , Logiciel , Analyse sur cellule unique/méthodes , Métabolomique/méthodes , Humains
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE