Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
NAR Genom Bioinform ; 6(1): lqae017, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38486887

RESUMO

Latest advancements in the high-throughput single-cell genome (scDNA) and transcriptome (scRNA) sequencing technologies enabled cell-resolved investigation of tissue clones. However, it remains challenging to cluster and couple single cells for heterogeneous scRNA and scDNA data generated from the same specimen. In this study, we present a computational framework called CCNMF, which employs a novel Coupled-Clone Non-negative Matrix Factorization technique to jointly infer clonal structure for matched scDNA and scRNA data. CCNMF couples multi-omics single cells by linking copy number and gene expression profiles through their general concordance. It successfully resolved the underlying coexisting clones with high correlations between the clonal genome and transcriptome from the same specimen. We validated that CCNMF can achieve high accuracy and robustness using both simulated benchmarks and real-world applications, including an ovarian cancer cell lines mixture, a gastric cancer cell line, and a primary gastric cancer. In summary, CCNMF provides a powerful tool for integrating multi-omics single-cell data, enabling simultaneous resolution of genomic and transcriptomic clonal architecture. This computational framework facilitates the understanding of how cellular gene expression changes in conjunction with clonal genome alternations, shedding light on the cellular genomic difference of subclones that contributes to tumor evolution.

2.
Food Chem ; 441: 138358, 2024 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-38266315

RESUMO

Protein microgels are emerging as versatile soft particles due to their desirable interfacial activities and functional properties. In this study, pea protein isolate microgel particles (PPIMP) were prepared by heat treatment and transglutaminase crosslinking, and PPIMP were non-covalently and covalently modified with sodium alginate (SA). The effects of polymer ratio and pH on the formation of PPIMP-SA mixtures and conjugates were investigated. The optimal ratio of PPIMP and SA was found to be 20:1, with the optimal pH being 7 and 10, respectively. PPIMP-SA conjugates were prepared by Maillard reaction. It was found that ultrasound (195 W, 40 min) enhanced the degree of glycation of PPIMP, with a highest value of 37.21 ± 0.71 %. SDS-PAGE, browning intensity and FTIR data also confirmed the formation of PPIMP-SA conjugates. Compared with PPIMP and PPIMP-SA mixtures, PPIMP-SA conjugates exhibited better thermal stability, antioxidant, emulsifying and foaming properties, which opens up opportunities for protein microgel in various food applications.


Assuntos
Microgéis , Proteínas de Ervilha , Emulsões/química , Alginatos , Proteínas de Ervilha/química , Antioxidantes/química
3.
Genome Med ; 15(1): 100, 2023 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-38008725

RESUMO

BACKGROUND: Understanding the mechanistic effects of novel immunotherapy agents is critical to improving their successful clinical translation. These effects need to be studied in preclinical models that maintain the heterogenous tumor microenvironment (TME) and dysfunctional cell states found in a patient's tumor. We investigated immunotherapy perturbations targeting co-stimulatory molecule GITR and co-inhibitory immune checkpoint TIGIT in a patient-derived ex vivo system that maintains the TME in its near-native state. Leveraging single-cell genomics, we identified cell type-specific transcriptional reprogramming in response to immunotherapy perturbations. METHODS: We generated ex vivo tumor slice cultures from fresh surgical resections of gastric and colon cancer and treated them with GITR agonist or TIGIT antagonist antibodies. We applied paired single-cell RNA and TCR sequencing to the original surgical resections, control, and treated ex vivo tumor slice cultures. We additionally confirmed target expression using multiplex immunofluorescence and validated our findings with RNA in situ hybridization. RESULTS: We confirmed that tumor slice cultures maintained the cell types, transcriptional cell states and proportions of the original surgical resection. The GITR agonist was limited to increasing effector gene expression only in cytotoxic CD8 T cells. Dysfunctional exhausted CD8 T cells did not respond to GITR agonist. In contrast, the TIGIT antagonist increased TCR signaling and activated both cytotoxic and dysfunctional CD8 T cells. This included cells corresponding to TCR clonotypes with features indicative of potential tumor antigen reactivity. The TIGIT antagonist also activated T follicular helper-like cells and dendritic cells, and reduced markers of immunosuppression in regulatory T cells. CONCLUSIONS: We identified novel cellular mechanisms of action of GITR and TIGIT immunotherapy in the patients' TME. Unlike the GITR agonist that generated a limited transcriptional response, TIGIT antagonist orchestrated a multicellular response involving CD8 T cells, T follicular helper-like cells, dendritic cells, and regulatory T cells. Our experimental strategy combining single-cell genomics with preclinical models can successfully identify mechanisms of action of novel immunotherapy agents. Understanding the cellular and transcriptional mechanisms of response or resistance will aid in prioritization of targets and their clinical translation.


Assuntos
Neoplasias Gastrointestinais , Microambiente Tumoral , Humanos , Imunoterapia , Receptores de Antígenos de Linfócitos T/genética , Receptores Imunológicos/genética , RNA
4.
bioRxiv ; 2023 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-37786704

RESUMO

Objective: Gastric intestinal metaplasia (GIM) is a precancerous lesion that increases gastric cancer (GC) risk. The Operative Link on GIM (OLGIM) is a combined clinical-histopathologic system to risk-stratify patients with GIM. The identification of molecular biomarkers that are indicators for advanced OLGIM lesions may improve cancer prevention efforts. Methods: This study was based on clinical and genomic data from four cohorts: 1) GAPS, a GIM cohort with detailed OLGIM severity scoring (N=303 samples); 2) the Cancer Genome Atlas (N=198); 3) a collation of in-house and publicly available scRNA-seq data (N=40), and 4) a spatial validation cohort (N=5) consisting of annotated histology slides of patients with either GC or advanced GIM. We used a multi-omics pipeline to identify, validate and sequentially parse a highly-refined signature of 26 genes which characterize high-risk GIM. Results: Using standard RNA-seq, we analyzed two separate, non-overlapping discovery (N=88) and validation (N=215) sets of GIM. In the discovery phase, we identified 105 upregulated genes specific for high-risk GIM (defined as OLGIM III-IV), of which 100 genes were independently confirmed in the validation set. Spatial transcriptomic profiling revealed 36 of these 100 genes to be expressed in metaplastic foci in GIM. Comparison with bulk GC sequencing data revealed 26 of these genes to be expressed in intestinal-type GC. Single-cell profiling resolved the 26-gene signature to both mature intestinal lineages (goblet cells, enterocytes) and immature intestinal lineages (stem-like cells). A subset of these genes was further validated using single-molecule multiplex fluorescence in situ hybridization. We found certain genes (TFF3 and ANPEP) to mark differentiated intestinal lineages, whereas others (OLFM4 and CPS1) localized to immature cells in the isthmic/crypt region of metaplastic glands, consistent with the findings from scRNAseq analysis. Conclusions: using an integrated multi-omics approach, we identified a novel 26-gene expression signature for high-OLGIM precursors at increased risk for GC. We found this signature localizes to aberrant intestinal stem-like cells within the metaplastic microenvironment. These findings hold important translational significance for future prevention and early detection efforts.

5.
NAR Cancer ; 5(3): zcad034, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37435532

RESUMO

In this proof-of-concept study, we developed a single-cell method that provides genotypes of somatic alterations found in coding regions of messenger RNAs and integrates these transcript-based variants with their matching cell transcriptomes. We used nanopore adaptive sampling on single-cell complementary DNA libraries to validate coding variants in target gene transcripts, and short-read sequencing to characterize cell types harboring the mutations. CRISPR edits for 16 targets were identified using a cancer cell line, and known variants in the cell line were validated using a 352-gene panel. Variants in primary cancer samples were validated using target gene panels ranging from 161 to 529 genes. A gene rearrangement was also identified in one patient, with the rearrangement occurring in two distinct tumor sites.

6.
Genome Med ; 15(1): 33, 2023 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-37138315

RESUMO

Epigenetic characterization of cell-free DNA (cfDNA) is an emerging approach for detecting and characterizing diseases such as cancer. We developed a strategy using nanopore-based single-molecule sequencing to measure cfDNA methylomes. This approach generated up to 200 million reads for a single cfDNA sample from cancer patients, an order of magnitude improvement over existing nanopore sequencing methods. We developed a single-molecule classifier to determine whether individual reads originated from a tumor or immune cells. Leveraging methylomes of matched tumors and immune cells, we characterized cfDNA methylomes of cancer patients for longitudinal monitoring during treatment.


Assuntos
Ácidos Nucleicos Livres , Sequenciamento por Nanoporos , Neoplasias , Humanos , Ácidos Nucleicos Livres/genética , Neoplasias/genética , DNA , Metilação de DNA
7.
bioRxiv ; 2023 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-36993756

RESUMO

Understanding the cellular mechanisms of novel immunotherapy agents in the human tumor microenvironment (TME) is critical to their clinical success. We examined GITR and TIGIT immunotherapy in gastric and colon cancer patients using ex vivo slice tumor slice cultures derived from cancer surgical resections. This primary culture system maintains the original TME in a near-native state. We applied paired single-cell RNA and TCR sequencing to identify cell type specific transcriptional reprogramming. The GITR agonist was limited to increasing effector gene expression only in cytotoxic CD8 T cells. The TIGIT antagonist increased TCR signaling and activated both cytotoxic and dysfunctional CD8 T cells, including clonotypes indicative of potential tumor antigen reactivity. The TIGIT antagonist also activated T follicular helper-like cells and dendritic cells, and reduced markers of immunosuppression in regulatory T cells. Overall, we identified cellular mechanisms of action of these two immunotherapy targets in the patients' TME.

8.
Food Chem ; 417: 135889, 2023 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-36933430

RESUMO

Aqueous probiotic suspensions were dispersed in an oil phase consisting of fish oil and medium chain triglycerides to form W1/O emulsions. These emulsions were then homogenized with an aqueous solution containing soybean protein isolate and sodium alginate to form W1/O/W2 emulsions. Fish oil was used to promote the growth of the probiotics and increase their ability to adhere to the intestinal mucosa. Sodium alginate increased the viscosity, stability, and probiotic encapsulation efficiency of the double emulsions, which was mainly attributed to its interactions with adsorbed soy proteins. The encapsulation efficiency of the probiotics in the double emulsions was relatively high (>96%). In vitro simulated digestion experiments showed that the double emulsions significantly increased the number of viable probiotics remaining after passing through the entire gastrointestinal tract. This study suggests that encapsulation of probiotics in double emulsions may increase their viability under gastrointestinal conditions, thereby enhancing their efficacy in functional foods.


Assuntos
Óleos de Peixe , Probióticos , Emulsões , Água , Alginatos , Proteínas de Soja
9.
Clin Cancer Res ; 29(1): 244-260, 2023 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-36239989

RESUMO

PURPOSE: The liver is the most frequent metastatic site for colorectal cancer. Its microenvironment is modified to provide a niche that is conducive for colorectal cancer cell growth. This study focused on characterizing the cellular changes in the metastatic colorectal cancer (mCRC) liver tumor microenvironment (TME). EXPERIMENTAL DESIGN: We analyzed a series of microsatellite stable (MSS) mCRCs to the liver, paired normal liver tissue, and peripheral blood mononuclear cells using single-cell RNA sequencing (scRNA-seq). We validated our findings using multiplexed spatial imaging and bulk gene expression with cell deconvolution. RESULTS: We identified TME-specific SPP1-expressing macrophages with altered metabolism features, foam cell characteristics, and increased activity in extracellular matrix (ECM) organization. SPP1+ macrophages and fibroblasts expressed complementary ligand-receptor pairs with the potential to mutually influence their gene-expression programs. TME lacked dysfunctional CD8 T cells and contained regulatory T cells, indicative of immunosuppression. Spatial imaging validated these cell states in the TME. Moreover, TME macrophages and fibroblasts had close spatial proximity, which is a requirement for intercellular communication and networking. In an independent cohort of mCRCs in the liver, we confirmed the presence of SPP1+ macrophages and fibroblasts using gene-expression data. An increased proportion of TME fibroblasts was associated with the worst prognosis in these patients. CONCLUSIONS: We demonstrated that mCRC in the liver is characterized by transcriptional alterations of macrophages in the TME. Intercellular networking between macrophages and fibroblasts supports colorectal cancer growth in the immunosuppressed metastatic niche in the liver. These features can be used to target immune-checkpoint-resistant MSS tumors.


Assuntos
Neoplasias do Colo , Leucócitos Mononucleares , Neoplasias Hepáticas , Humanos , Neoplasias do Colo/patologia , Fibroblastos , Imunossupressores , Fígado , Macrófagos , Osteopontina , Microambiente Tumoral/genética , Neoplasias Hepáticas/secundário
10.
Food Res Int ; 157: 111451, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35761690

RESUMO

Zein nanoparticles are commonly used as colloidal emulsifiers to form and stabilize Pickering emulsions. However, the strong surface hydrophobicity of zein nanoparticles limits their widespread application. In this study, composite colloidal emulsifiers were fabricated from zein, sodium caseinate (NaCas), and epigallocatechin gallate (EGCG). Initially, NaCas-EGCG conjugates were formed using either an alkaline or enzymatic method. The enzymatic method led to conjugates containing more EGCG and with a higher thermal stability and surface hydrophilicity. Colloidal emulsifiers were prepared using an antisolvent precipitation method that involved titrating an ethanolic zein solution into an aqueous NaCas-EGCG conjugate solution. The potential application of these emulsifiers for forming and stabilizing high internal phase emulsions (HIPEs) was then explored. The emulsification properties of the zein nanoparticles were improved after they were complexed with NaCas-EGCG conjugates. Pickering HIPEs containing closely packed polygon oil droplets were formed from the colloidal emulsifiers, even at low particle concentrations (0.3% w/v). Overall, our results show that the functional performance of zein nanoparticles can be improved by complexing them with NaCas-EGCG conjugates. The novel colloidal emulsifiers developed in this study may therefore have useful applications in the food and other industries.


Assuntos
Zeína , Caseínas/química , Catequina/análogos & derivados , Emulsificantes/química , Emulsões/química , Interações Hidrofóbicas e Hidrofílicas , Zeína/química
11.
Bioinformatics ; 36(Suppl_1): i48-i56, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32657382

RESUMO

MOTIVATION: Single-cell multi-omics data provide a comprehensive molecular view of cells. However, single-cell multi-omics datasets consist of unpaired cells measured with distinct unmatched features across modalities, making data integration challenging. RESULTS: In this study, we present a novel algorithm, termed UnionCom, for the unsupervised topological alignment of single-cell multi-omics integration. UnionCom does not require any correspondence information, either among cells or among features. It first embeds the intrinsic low-dimensional structure of each single-cell dataset into a distance matrix of cells within the same dataset and then aligns the cells across single-cell multi-omics datasets by matching the distance matrices via a matrix optimization method. Finally, it projects the distinct unmatched features across single-cell datasets into a common embedding space for feature comparability of the aligned cells. To match the complex non-linear geometrical distorted low-dimensional structures across datasets, UnionCom proposes and adjusts a global scaling parameter on distance matrices for aligning similar topological structures. It does not require one-to-one correspondence among cells across datasets, and it can accommodate samples with dataset-specific cell types. UnionCom outperforms state-of-the-art methods on both simulated and real single-cell multi-omics datasets. UnionCom is robust to parameter choices, as well as subsampling of features. AVAILABILITY AND IMPLEMENTATION: UnionCom software is available at https://github.com/caokai1073/UnionCom. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Algoritmos , Software
12.
Artigo em Inglês | MEDLINE | ID: mdl-29994127

RESUMO

The advance in single-cell profiling technologies and the development in computational algorithms provide the opportunity to reconstruct pseudo temporal trajectory with branch point of cellular development. On the other hand, theories such as dynamical network biomarkers (DNB) theory have been recently proposed to characterize the pre-transition state in biological systems. Few studies have validated whether the branch point identified in pseudo time is the critical point in dynamical system. In this study, the dynamical behavior of the branch point on the pseudo trajectory has been investigated. We study the pseudo temporal trajectories reconstructed by Wishbone and diffusion pseudotime analysis (DPT) algorithms, as well as the simulated trajectory. DNB theory is applied to justify the bifurcating event on the pseudo trajectories. Our results demonstrate that the branch point recovered by Wishbone and DPT algorithms is confirmed as a transition state in cell differentiation process by DNB theory. Furthermore, we show that an appropriate DNB group will amplify the comprehensive index of critical event as defined in DNB theory. Our study provides biological insights on pseudo trajectory with branch point in a dynamical view and also indicates that DNB theory may serve as a benchmark to check the validity of branch point.


Assuntos
Algoritmos , Biomarcadores/análise , Diferenciação Celular/fisiologia , Biologia Computacional/métodos , Análise de Célula Única/métodos , Animais , Bases de Dados Genéticas , Camundongos
13.
Bioinformatics ; 35(23): 4962-4970, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31116393

RESUMO

MOTIVATION: Cell fate determination is a continuous process in which one cell type diversifies to other cell types following a hierarchical path. Advancements in single-cell technologies provide the opportunity to reveal the continuum of cell progression which forms a structured continuous tree (SCTree). Computational algorithms, which are usually based on a priori assumptions on the hidden structures, have previously been proposed as a means of recovering pseudo trajectory along cell differentiation process. However, there still lack of statistical framework on the assessments of intrinsic structure embedded in high-dimensional gene expression profile. Inherit noise and cell-to-cell variation underlie the single-cell data, however, pose grand challenges to testing even basic structures, such as linear versus bifurcation. RESULTS: In this study, we propose an adaptive statistical framework, termed SCTree, to test the intrinsic structure of a high-dimensional single-cell dataset. SCTree test is conducted based on the tools derived from metric geometry and random matrix theory. In brief, by extending the Gromov-Farris transform and utilizing semicircular law, we formulate the continuous tree structure testing problem into a signal matrix detection problem. We show that the SCTree test is most powerful when the signal-to-noise ratio exceeds a moderate value. We also demonstrate that SCTree is able to robustly detect linear, single and multiple branching events with simulated datasets and real scRNA-seq datasets. Overall, the SCTree test provides a unified statistical assessment of the significance of the hidden structure of single-cell data. AVAILABILITY AND IMPLEMENTATION: SCTree software is available at https://github.com/XQBai/SCTree-test. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Software , Algoritmos , Perfilação da Expressão Gênica , Análise de Sequência de RNA , Análise de Célula Única
14.
Bioinformatics ; 35(15): 2593-2601, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-30535348

RESUMO

MOTIVATION: Visualizing and reconstructing cell developmental trajectories intrinsically embedded in high-dimensional expression profiles of single-cell RNA sequencing (scRNA-seq) snapshot data are computationally intriguing, but challenging. RESULTS: We propose DensityPath, an algorithm allowing (i) visualization of the intrinsic structure of scRNA-seq data on an embedded 2-d space and (ii) reconstruction of an optimal cell state-transition path on the density landscape. DensityPath powerfully handles high dimensionality and heterogeneity of scRNA-seq data by (i) revealing the intrinsic structures of data, while adopting a non-linear dimension reduction algorithm, termed elastic embedding, which can preserve both local and global structures of the data; and (ii) extracting the topological features of high-density, level-set clusters from a single-cell multimodal density landscape of transcriptional heterogeneity, as the representative cell states. DensityPath reconstructs the optimal cell state-transition path by finding the geodesic minimum spanning tree of representative cell states on the density landscape, establishing a least action path with the minimum-transition-energy of cell fate decisions. We demonstrate that DensityPath can ably reconstruct complex trajectories of cell development, e.g. those with multiple bifurcating and trifurcating branches, while maintaining computational efficiency. Moreover, DensityPath has high accuracy for pseudotime calculation and branch assignment on real scRNA-seq, as well as simulated datasets. DensityPath is robust to parameter choices, as well as permutations of data. AVAILABILITY AND IMPLEMENTATION: DensityPath software is available at https://github.com/ucasdp/DensityPath. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
RNA/genética , Algoritmos , Perfilação da Expressão Gênica , Análise de Sequência de RNA , Análise de Célula Única
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA