Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cell Metab ; 35(10): 1688-1703.e10, 2023 10 03.
Article in English | MEDLINE | ID: mdl-37793345

ABSTRACT

Metastasis causes breast cancer-related mortality. Tumor-infiltrating neutrophils (TINs) inflict immunosuppression and promote metastasis. Therapeutic debilitation of TINs may enhance immunotherapy, yet it remains a challenge to identify therapeutic targets highly expressed and functionally essential in TINs but under-expressed in extra-tumoral neutrophils. Here, using single-cell RNA sequencing to compare TINs and circulating neutrophils in murine mammary tumor models, we identified aconitate decarboxylase 1 (Acod1) as the most upregulated metabolic enzyme in mouse TINs and validated high Acod1 expression in human TINs. Activated through the GM-CSF-JAK/STAT5-C/EBPß pathway, Acod1 produces itaconate, which mediates Nrf2-dependent defense against ferroptosis and upholds the persistence of TINs. Acod1 ablation abates TIN infiltration, constrains metastasis (but not primary tumors), bolsters antitumor T cell immunity, and boosts the efficacy of immune checkpoint blockade. Our findings reveal how TINs escape from ferroptosis through the Acod1-dependent immunometabolism switch and establish Acod1 as a target to offset immunosuppression and improve immunotherapy against metastasis.


Subject(s)
Breast Neoplasms , Carboxy-Lyases , Ferroptosis , Humans , Mice , Animals , Female , Breast Neoplasms/metabolism , Neutrophils , Carboxy-Lyases/metabolism , Melanoma, Cutaneous Malignant
2.
Nucleic Acids Res ; 51(W1): W180-W190, 2023 07 05.
Article in English | MEDLINE | ID: mdl-37216602

ABSTRACT

Quantitative assessment of single cell fluxome is critical for understanding the metabolic heterogeneity in diseases. Unfortunately, laboratory-based single cell fluxomics is currently impractical, and the current computational tools for flux estimation are not designed for single cell-level prediction. Given the well-established link between transcriptomic and metabolomic profiles, leveraging single cell transcriptomics data to predict single cell fluxome is not only feasible but also an urgent task. In this study, we present FLUXestimator, an online platform for predicting metabolic fluxome and variations using single cell or general transcriptomics data of large sample-size. The FLUXestimator webserver implements a recently developed unsupervised approach called single cell flux estimation analysis (scFEA), which uses a new neural network architecture to estimate reaction rates from transcriptomics data. To the best of our knowledge, FLUXestimator is the first web-based tool dedicated to predicting cell-/sample-wise metabolic flux and metabolite variations using transcriptomics data of human, mouse and 15 other common experimental organisms. The FLUXestimator webserver is available at http://scFLUX.org/, and stand-alone tools for local use are available at https://github.com/changwn/scFEA. Our tool provides a new avenue for studying metabolic heterogeneity in diseases and has the potential to facilitate the development of new therapeutic strategies.


Subject(s)
Software , Transcriptome , Animals , Humans , Mice , Metabolic Networks and Pathways , Metabolomics , Models, Biological
3.
Comput Struct Biotechnol J ; 21: 2160-2171, 2023.
Article in English | MEDLINE | ID: mdl-37013005

ABSTRACT

The cells of colorectal cancer (CRC) in their microenvironment experience constant stress, leading to dysregulated activity in the tumor niche. As a result, cancer cells acquire alternative pathways in response to the changing microenvironment, posing significant challenges for the design of effective cancer treatment strategies. While computational studies on high-throughput omics data have advanced our understanding of CRC subtypes, characterizing the heterogeneity of this disease remains remarkably complex. Here, we present a novel computational Pipeline for Characterizing Alternative Mechanisms (PCAM) based on biclustering to gain a more detailed understanding of cancer heterogeneity. Our application of PCAM to large-scale CRC transcriptomics datasets suggests that PCAM can generate a wealth of information leading to new biological understanding and predictive markers of alternative mechanisms. Our key findings include: 1) A comprehensive collection of alternative pathways in CRC, associated with biological and clinical factors. 2) Full annotation of detected alternative mechanisms, including their enrichment in known pathways and associations with various clinical outcomes. 3) A mechanistic relationship between known clinical subtypes and outcomes on a consensus map, visualized by the presence of alternative mechanisms. 4) Several potential novel alternative drug resistance mechanisms for Oxaliplatin, 5-Fluorouracil, and FOLFOX, some of which were validated on independent datasets. We believe that gaining a deeper understanding of alternative mechanisms is a critical step towards characterizing the heterogeneity of CRC. The hypotheses generated by PCAM, along with the comprehensive collection of biologically and clinically associated alternative pathways in CRC, could provide valuable insights into the underlying mechanisms driving cancer progression and drug resistance, which could aid in the development of more effective cancer therapies and guide experimental design towards more targeted and personalized treatment strategies. The computational pipeline of PCAM is available in GitHub (https://github.com/changwn/BC-CRC).

4.
Front Endocrinol (Lausanne) ; 14: 1063083, 2023.
Article in English | MEDLINE | ID: mdl-36777346

ABSTRACT

Introduction: Due to a lack of spatial-temporal resolution at the single cell level, the etiologies of the bone dysfunction caused by diseases such as normal aging, osteoporosis, and the metabolic bone disease associated with chronic kidney disease (CKD) remain largely unknown. Methods: To this end, flow cytometry and scRNAseq were performed on long bone cells from Sost-cre/Ai9+ mice, and pure osteolineage transcriptomes were identified, including novel osteocyte-specific gene sets. Results: Clustering analysis isolated osteoblast precursors that expressed Tnc, Mmp13, and Spp1, and a mature osteoblast population defined by Smpd3, Col1a1, and Col11a1. Osteocytes were demarcated by Cd109, Ptprz1, Ramp1, Bambi, Adamts14, Spns2, Bmp2, WasI, and Phex. We validated our in vivo scRNAseq using integrative in vitro promoter occupancy via ATACseq coupled with transcriptomic analyses of a conditional, temporally differentiated MSC cell line. Further, trajectory analyses predicted osteoblast-to-osteocyte transitions via defined pathways associated with a distinct metabolic shift as determined by single-cell flux estimation analysis (scFEA). Using the adenine mouse model of CKD, at a time point prior to major skeletal alterations, we found that gene expression within all stages of the osteolineage was disturbed. Conclusion: In sum, distinct populations of osteoblasts/osteocytes were defined at the single cell level. Using this roadmap of gene assembly, we demonstrated unrealized molecular defects across multiple bone cell populations in a mouse model of CKD, and our collective results suggest a potentially earlier and more broad bone pathology in this disease than previously recognized.


Subject(s)
Renal Insufficiency, Chronic , Transcriptome , Mice , Animals , Bone and Bones/metabolism , Osteoblasts/metabolism , Cortical Bone/metabolism , Renal Insufficiency, Chronic/pathology , Membrane Proteins/metabolism , Sphingomyelin Phosphodiesterase/metabolism
5.
Article in English | MEDLINE | ID: mdl-35787947

ABSTRACT

Acid-base homeostasis is a fundamental property of living cells and its persistent disruption in human cells can lead to a wide range of diseases. We have conducted computational modeling and analysis of transcriptomic data of 4750 human tissue samples of nine cancer types in the Cancer Genome Atlas (TCGA) database. Built on our previous study, we have quantitatively estimated the (average) production rate of OH- by cytosolic Fenton reactions, which continuously disrupt the intracellular pH homeostasis. Our predictions indicate that all or a subset of 43 reprogrammed metabolisms (RMs) are induced to produce net protons (H+) at comparable rates of Fenton reactions to keep the intracellular pH stable. We have then discovered that a number of well-known phenotypes of cancers, including increased growth rate, metastasis rate, and local immune cell composition, can be naturally explained in terms of the Fenton reaction level and the induced RMs. This study strongly suggests the possibility to have a unified framework for studies of cancer-inducing stressors, adaptive metabolic reprogramming, and cancerous behaviors. In addition, strong evidence is provided to demonstrate that a popular view of that Na+/H+ exchangers, along with lactic acid exporters and carbonic anhydrases are responsible for the intracellular alkalization and extracellular acidification in cancer may not be justified.

7.
Blood ; 140(11): 1263-1277, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35772013

ABSTRACT

Hematopoietic stem cells (HSCs) manifest impaired recovery and self-renewal with a concomitant increase in differentiation when exposed to ambient air as opposed to physioxia. Mechanism(s) behind this distinction are poorly understood but have the potential to improve stem cell transplantation. Single-cell RNA sequencing of HSCs in physioxia revealed upregulation of HSC self-renewal genes and downregulation of genes involved in inflammatory pathways and HSC differentiation. HSCs under physioxia also exhibited downregulation of the epigenetic modifier Tet2. Tet2 is α-ketoglutarate, iron- and oxygen-dependent dioxygenase that converts 5-methylcytosine to 5-hydroxymethylcytosine, thereby promoting active transcription. We evaluated whether loss of Tet2 affects the number and function of HSCs and hematopoietic progenitor cells (HPCs) under physioxia and ambient air. In contrast to wild-type HSCs (WT HSCs), a complete nonresponsiveness of Tet2-/- HSCs and HPCs to changes in oxygen tension was observed. Unlike WT HSCs, Tet2-/- HSCs and HPCs exhibited similar numbers and function in either physioxia or ambient air. The lack of response to changes in oxygen tension in Tet2-/- HSCs was associated with similar changes in self-renewal and quiescence genes among WT HSC-physioxia, Tet2-/- HSC-physioxia and Tet2-/- HSC-air. We define a novel molecular program involving Tet2 in regulating HSCs under physioxia.


Subject(s)
5-Methylcytosine , Dioxygenases , 5-Methylcytosine/metabolism , Cell Differentiation/physiology , Dioxygenases/metabolism , Down-Regulation , Hematopoietic Stem Cells/metabolism , Iron/metabolism , Ketoglutaric Acids , Oxygen/metabolism
8.
PLoS Comput Biol ; 18(3): e1009956, 2022 03.
Article in English | MEDLINE | ID: mdl-35349572

ABSTRACT

Metastatic cancer accounts for over 90% of all cancer deaths, and evaluations of metastasis potential are vital for minimizing the metastasis-associated mortality and achieving optimal clinical decision-making. Computational assessment of metastasis potential based on large-scale transcriptomic cancer data is challenging because metastasis events are not always clinically detectable. The under-diagnosis of metastasis events results in biased classification labels, and classification tools using biased labels may lead to inaccurate estimations of metastasis potential. This issue is further complicated by the unknown metastasis prevalence at the population level, the small number of confirmed metastasis cases, and the high dimensionality of the candidate molecular features. Our proposed algorithm, called Positive and unlabeled Learning from Unbalanced cases and Sparse structures (PLUS), is the first to use a positive and unlabeled learning framework to account for the under-detection of metastasis events in building a classifier. PLUS is specifically tailored for studying metastasis that deals with the unbalanced instance allocation as well as unknown metastasis prevalence, which are not considered by other methods. PLUS achieves superior performance on synthetic datasets compared with other state-of-the-art methods. Application of PLUS to The Cancer Genome Atlas Pan-Cancer gene expression data generated metastasis potential predictions that show good agreement with the clinical follow-up data, in addition to predictive genes that have been validated by independent single-cell RNA-sequencing datasets.


Subject(s)
Algorithms , Neoplasms , Humans
9.
Brief Bioinform ; 22(4)2021 07 20.
Article in English | MEDLINE | ID: mdl-34293851

ABSTRACT

Identifying relationships between genetic variations and their clinical presentations has been challenged by the heterogeneous causes of a disease. It is imperative to unveil the relationship between the high-dimensional genetic manifestations and the clinical presentations, while taking into account the possible heterogeneity of the study subjects.We proposed a novel supervised clustering algorithm using penalized mixture regression model, called component-wise sparse mixture regression (CSMR), to deal with the challenges in studying the heterogeneous relationships between high-dimensional genetic features and a phenotype. The algorithm was adapted from the classification expectation maximization algorithm, which offers a novel supervised solution to the clustering problem, with substantial improvement on both the computational efficiency and biological interpretability. Experimental evaluation on simulated benchmark datasets demonstrated that the CSMR can accurately identify the subspaces on which subset of features are explanatory to the response variables, and it outperformed the baseline methods. Application of CSMR on a drug sensitivity dataset again demonstrated the superior performance of CSMR over the others, where CSMR is powerful in recapitulating the distinct subgroups hidden in the pool of cell lines with regards to their coping mechanisms to different drugs. CSMR represents a big data analysis tool with the potential to resolve the complexity of translating the clinical representations of the disease to the real causes underpinning it. We believe that it will bring new understanding to the molecular basis of a disease and could be of special relevance in the growing field of personalized medicine.


Subject(s)
Algorithms , Genetic Variation , Models, Genetic , Humans
10.
Genome Res ; 31(10): 1867-1884, 2021 10.
Article in English | MEDLINE | ID: mdl-34301623

ABSTRACT

The metabolic heterogeneity and metabolic interplay between cells are known as significant contributors to disease treatment resistance. However, with the lack of a mature high-throughput single-cell metabolomics technology, we are yet to establish systematic understanding of the intra-tissue metabolic heterogeneity and cooperative mechanisms. To mitigate this knowledge gap, we developed a novel computational method, namely, single-cell flux estimation analysis (scFEA), to infer the cell-wise fluxome from single-cell RNA-sequencing (scRNA-seq) data. scFEA is empowered by a systematically reconstructed human metabolic map as a factor graph, a novel probabilistic model to leverage the flux balance constraints on scRNA-seq data, and a novel graph neural network-based optimization solver. The intricate information cascade from transcriptome to metabolome was captured using multilayer neural networks to capitulate the nonlinear dependency between enzymatic gene expressions and reaction rates. We experimentally validated scFEA by generating an scRNA-seq data set with matched metabolomics data on cells of perturbed oxygen and genetic conditions. Application of scFEA on this data set showed the consistency between predicted flux and the observed variation of metabolite abundance in the matched metabolomics data. We also applied scFEA on five publicly available scRNA-seq and spatial transcriptomics data sets and identified context- and cell group-specific metabolic variations. The cell-wise fluxome predicted by scFEA empowers a series of downstream analyses including identification of metabolic modules or cell groups that share common metabolic variations, sensitivity evaluation of enzymes with regards to their impact on the whole metabolic flux, and inference of cell-tissue and cell-cell metabolic communications.


Subject(s)
Single-Cell Analysis , Transcriptome , Gene Expression Profiling/methods , Neural Networks, Computer , RNA-Seq , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Exome Sequencing
11.
Brief Bioinform ; 22(4)2021 07 20.
Article in English | MEDLINE | ID: mdl-33230549

ABSTRACT

Deconvolution of mouse transcriptomic data is challenged by the fact that mouse models carry various genetic and physiological perturbations, making it questionable to assume fixed cell types and cell type marker genes for different data set scenarios. We developed a Semi-Supervised Mouse data Deconvolution (SSMD) method to study the mouse tissue microenvironment. SSMD is featured by (i) a novel nonparametric method to discover data set-specific cell type signature genes; (ii) a community detection approach for fixing cell types and their marker genes; (iii) a constrained matrix decomposition method to solve cell type relative proportions that is robust to diverse experimental platforms. In summary, SSMD addressed several key challenges in the deconvolution of mouse tissue data, including: (i) varied cell types and marker genes caused by highly divergent genotypic and phenotypic conditions of mouse experiment; (ii) diverse experimental platforms of mouse transcriptomics data; (iii) small sample size and limited training data source and (iv) capable to estimate the proportion of 35 cell types in blood, inflammatory, central nervous or hematopoietic systems. In silico and experimental validation of SSMD demonstrated its high sensitivity and accuracy in identifying (sub) cell types and predicting cell proportions comparing with state-of-the-arts methods. A user-friendly R package and a web server of SSMD are released via https://github.com/xiaoyulu95/SSMD.


Subject(s)
Antigens, Differentiation , Cellular Microenvironment , Computational Biology , Databases, Genetic , Gene Expression Profiling , Transcriptome , Animals , Antigens, Differentiation/biosynthesis , Antigens, Differentiation/genetics , Mice , Organ Specificity
12.
Cancers (Basel) ; 12(11)2020 Oct 29.
Article in English | MEDLINE | ID: mdl-33138184

ABSTRACT

Tumor immune infiltration plays a key role in the progression of solid tumors, including ovarian cancer, and immunotherapies are rapidly emerging as effective treatment modalities. However, the role of cancer-associated fibroblasts (CAFs), a predominant stromal constituent, in determining the tumor-immune microenvironment and modulating efficacy of immunotherapies remains poorly understood. We have conducted an extensive bioinformatic analysis of our and other publicly available ovarian cancer datasets (GSE137237, GSE132289 and GSE71340), to determine the correlation of fibroblast subtypes within the tumor microenvironment (TME) with the characteristics of tumor-immune infiltration. We identified (1) four functional modules of CAFs in ovarian cancer that are associated with the TME and metastasis of ovarian cancer, (2) immune-suppressive function of the collagen 1,3,5-expressing CAFs in primary ovarian cancer and omental metastases, and (3) consistent positive correlations between the functional modules of CAFs with anti-immune response genes and negative correlation with pro-immune response genes. Our study identifies a specific fibroblast subtype, fibroblast functional module (FFM)2, in the ovarian cancer tumor microenvironment that can potentially modulate a tumor-promoting immune microenvironment, which may be detrimental toward the effectiveness of ovarian cancer immunotherapies.

13.
BMC Bioinformatics ; 20(Suppl 24): 672, 2019 Dec 20.
Article in English | MEDLINE | ID: mdl-31861972

ABSTRACT

BACKGROUND: Various statistical models have been developed to model the single cell RNA-seq expression profiles, capture its multimodality, and conduct differential gene expression test. However, for expression data generated by different experimental design and platforms, there is currently lack of capability to determine the most proper statistical model. RESULTS: We developed an R package, namely Multi-Modal Model Selection (M3S), for gene-wise selection of the most proper multi-modality statistical model and downstream analysis, useful in a single-cell or large scale bulk tissue transcriptomic data. M3S is featured with (1) gene-wise selection of the most parsimonious model among 11 most commonly utilized ones, that can best fit the expression distribution of the gene, (2) parameter estimation of a selected model, and (3) differential gene expression test based on the selected model. CONCLUSION: A comprehensive evaluation suggested that M3S can accurately capture the multimodality on simulated and real single cell data. An open source package and is available through GitHub at https://github.com/zy26/M3S.


Subject(s)
RNA/genetics , Sequence Analysis, RNA , High-Throughput Nucleotide Sequencing , Models, Genetic , Single-Cell Analysis , Transcriptome
14.
Nucleic Acids Res ; 47(18): e111, 2019 10 10.
Article in English | MEDLINE | ID: mdl-31372654

ABSTRACT

A key challenge in modeling single-cell RNA-seq data is to capture the diversity of gene expression states regulated by different transcriptional regulatory inputs across individual cells, which is further complicated by largely observed zero and low expressions. We developed a left truncated mixture Gaussian (LTMG) model, from the kinetic relationships of the transcriptional regulatory inputs, mRNA metabolism and abundance in single cells. LTMG infers the expression multi-modalities across single cells, meanwhile, the dropouts and low expressions are treated as left truncated. We demonstrated that LTMG has significantly better goodness of fitting on an extensive number of scRNA-seq data, comparing to three other state-of-the-art models. Our biological assumption of the low non-zero expressions, rationality of the multimodality setting, and the capability of LTMG in extracting expression states specific to cell types or functions, are validated on independent experimental data sets. A differential gene expression test and a co-regulation module identification method are further developed. We experimentally validated that our differential expression test has higher sensitivity and specificity, compared with other five popular methods. The co-regulation analysis is capable of retrieving gene co-regulation modules corresponding to perturbed transcriptional regulations. A user-friendly R package with all the analysis power is available at https://github.com/zy26/LTMGSCA.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , RNA/genetics , Single-Cell Analysis/methods , Software , Algorithms , Gene Expression Profiling , Gene Expression Regulation/genetics , Models, Statistical , Sequence Analysis, RNA/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...