Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 22(12): 1577-1589, 2021 12.
Article in English | MEDLINE | ID: mdl-34811546

ABSTRACT

Single-cell genomics technology has transformed our understanding of complex cellular systems. However, excessive cost and a lack of strategies for the purification of newly identified cell types impede their functional characterization and large-scale profiling. Here, we have generated high-content single-cell proteo-genomic reference maps of human blood and bone marrow that quantitatively link the expression of up to 197 surface markers to cellular identities and biological processes across all main hematopoietic cell types in healthy aging and leukemia. These reference maps enable the automatic design of cost-effective high-throughput cytometry schemes that outperform state-of-the-art approaches, accurately reflect complex topologies of cellular systems and permit the purification of precisely defined cell states. The systematic integration of cytometry and proteo-genomic data enables the functional capacities of precisely mapped cell states to be measured at the single-cell level. Our study serves as an accessible resource and paves the way for a data-driven era in cytometry.


Subject(s)
Blood Cells/metabolism , Bone Marrow Cells/metabolism , Cell Separation , Flow Cytometry , Gene Expression Profiling , Proteome , Proteomics , Single-Cell Analysis , Transcriptome , Age Factors , Blood Cells/immunology , Blood Cells/pathology , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Cells, Cultured , Databases, Genetic , Healthy Aging/genetics , Healthy Aging/immunology , Healthy Aging/metabolism , Humans , Leukemia/genetics , Leukemia/immunology , Leukemia/metabolism , Leukemia/pathology , RNA-Seq , Systems Biology
2.
Immunity ; 56(7): 1578-1595.e8, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37329888

ABSTRACT

It is currently not well known how necroptosis and necroptosis responses manifest in vivo. Here, we uncovered a molecular switch facilitating reprogramming between two alternative modes of necroptosis signaling in hepatocytes, fundamentally affecting immune responses and hepatocarcinogenesis. Concomitant necrosome and NF-κB activation in hepatocytes, which physiologically express low concentrations of receptor-interacting kinase 3 (RIPK3), did not lead to immediate cell death but forced them into a prolonged "sublethal" state with leaky membranes, functioning as secretory cells that released specific chemokines including CCL20 and MCP-1. This triggered hepatic cell proliferation as well as activation of procarcinogenic monocyte-derived macrophage cell clusters, contributing to hepatocarcinogenesis. In contrast, necrosome activation in hepatocytes with inactive NF-κB-signaling caused an accelerated execution of necroptosis, limiting alarmin release, and thereby preventing inflammation and hepatocarcinogenesis. Consistently, intratumoral NF-κB-necroptosis signatures were associated with poor prognosis in human hepatocarcinogenesis. Therefore, pharmacological reprogramming between these distinct forms of necroptosis may represent a promising strategy against hepatocellular carcinoma.


Subject(s)
Liver Neoplasms , NF-kappa B , Humans , NF-kappa B/metabolism , Protein Kinases/metabolism , Necroptosis , Inflammation/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Apoptosis
3.
PLoS Biol ; 21(8): e3002198, 2023 08.
Article in English | MEDLINE | ID: mdl-37594988

ABSTRACT

Pathogenic bacteria proliferating inside mammalian host cells need to rapidly adapt to the intracellular environment. How they achieve this and scavenge essential nutrients from the host has been an open question due to the difficulties in distinguishing between bacterial and host metabolites in situ. Here, we capitalized on the inability of mammalian cells to metabolize mannitol to develop a stable isotopic labeling approach to track Salmonella enterica metabolites during intracellular proliferation in host macrophage and epithelial cells. By measuring label incorporation into Salmonella metabolites with liquid chromatography-mass spectrometry (LC-MS), and combining it with metabolic modeling, we identify relevant carbon sources used by Salmonella, uncover routes of their metabolization, and quantify relative reaction rates in central carbon metabolism. Our results underline the importance of the Entner-Doudoroff pathway (EDP) and the phosphoenolpyruvate carboxylase for intracellularly proliferating Salmonella. More broadly, our metabolic labeling strategy opens novel avenues for understanding the metabolism of pathogens inside host cells.


Subject(s)
Salmonella enterica , Salmonella , Animals , Carbon , Chromatography, Liquid , Isotopes , Mammals
4.
J Am Soc Nephrol ; 2024 May 21.
Article in English | MEDLINE | ID: mdl-38771634

ABSTRACT

BACKGROUND: Diabetes is expected to directly impact renal glycosylation, yet to date, there has not been a comprehensive evaluation of alterations in N-glycan composition in the glomeruli of patients with diabetic kidney disease (DKD). METHODS: We used untargeted mass spectrometry imaging to identify N-glycan structures in healthy and sclerotic glomeruli in FFPE sections from needle biopsies of five patients with DKD and three healthy kidney samples. Regional proteomics was performed on glomeruli from additional biopsies from the same patients to compare the abundances of enzymes involved in glycosylation. Secondary analysis of single nuclei transcriptomics (snRNAseq) data was used to inform on transcript levels of glycosylation machinery in different cell types and states. RESULTS: We detected 120 N-glycans, and among them identified twelve of these protein post-translated modifications that were significantly increased in glomeruli. All glomeruli-specific N-glycans contained an N-acetyllactosamine (LacNAc) epitope. Five N-glycan structures were highly discriminant between sclerotic and healthy glomeruli. Sclerotic glomeruli had an additional set of glycans lacking fucose linked to their core, and they did not show tetra-antennary structures that are common in healthy glomeruli. Orthogonal omics analyses revealed lower protein abundance and lower gene expression involved in synthesizing fucosylated and branched N-glycans in sclerotic podocytes. In snRNAseq and regional proteomics analyses, we observed that genes and/or proteins involved in sialylation and LacNAc synthesis were also downregulated in DKD glomeruli, but this alteration remained undetectable by our spatial N-glycomics assay. CONCLUSIONS: Integrative spatial glycomics, proteomics, and transcriptomics revealed protein N-glycosylation characteristic of sclerotic glomeruli in DKD.

5.
Nat Methods ; 18(7): 799-805, 2021 07.
Article in English | MEDLINE | ID: mdl-34226721

ABSTRACT

A growing appreciation of the importance of cellular metabolism and revelations concerning the extent of cell-cell heterogeneity demand metabolic characterization of individual cells. We present SpaceM, an open-source method for in situ single-cell metabolomics that detects >100 metabolites from >1,000 individual cells per hour, together with a fluorescence-based readout and retention of morpho-spatial features. We validated SpaceM by predicting the cell types of cocultured human epithelial cells and mouse fibroblasts. We used SpaceM to show that stimulating human hepatocytes with fatty acids leads to the emergence of two coexisting subpopulations outlined by distinct cellular metabolic states. Inducing inflammation with the cytokine interleukin-17A perturbs the balance of these states in a process dependent on NF-κB signaling. The metabolic state markers were reproduced in a murine model of nonalcoholic steatohepatitis. We anticipate SpaceM to be broadly applicable for investigations of diverse cellular models and to democratize single-cell metabolomics.


Subject(s)
Image Processing, Computer-Assisted/methods , Metabolomics/methods , Single-Cell Analysis/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Animals , Coculture Techniques , Epithelial Cells , Fatty Acids/pharmacology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Inflammation/metabolism , Interleukin-17/metabolism , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , NIH 3T3 Cells , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Signal Transduction , Stress, Physiological
6.
Nat Methods ; 18(11): 1370-1376, 2021 11.
Article in English | MEDLINE | ID: mdl-34725482

ABSTRACT

Comprehensive metabolome analyses are essential for biomedical, environmental, and biotechnological research. However, current MS1- and MS2-based acquisition and data analysis strategies in untargeted metabolomics result in low identification rates of metabolites. Here we present HERMES, a molecular-formula-oriented and peak-detection-free method that uses raw LC/MS1 information to optimize MS2 acquisition. Investigating environmental water, Escherichia coli, and human plasma extracts with HERMES, we achieved an increased biological specificity of MS2 scans, leading to improved mass spectral similarity scoring and identification rates when compared with a state-of-the-art data-dependent acquisition (DDA) approach. Thus, HERMES improves sensitivity, selectivity, and annotation of metabolites. HERMES is available as an R package with a user-friendly graphical interface for data analysis and visualization.


Subject(s)
Algorithms , Escherichia coli/metabolism , Metabolome , Metabolomics/methods , Plasma/metabolism , Water Pollutants, Chemical/metabolism , Chromatography, Liquid/methods , Humans , Plasma/chemistry , Tandem Mass Spectrometry/methods , Water Pollutants, Chemical/analysis
7.
Mol Syst Biol ; 19(11): e10571, 2023 Nov 09.
Article in English | MEDLINE | ID: mdl-37842805

ABSTRACT

Spatial omics has emerged as a rapidly growing and fruitful field with hundreds of publications presenting novel methods for obtaining spatially resolved information for any omics data type on spatial scales ranging from subcellular to organismal. From a technology development perspective, spatial omics is a highly interdisciplinary field that integrates imaging and omics, spatial and molecular analyses, sequencing and mass spectrometry, and image analysis and bioinformatics. The emergence of this field has not only opened a window into spatial biology, but also created multiple novel opportunities, questions, and challenges for method developers. Here, we provide the perspective of technology developers on what makes the spatial omics field unique. After providing a brief overview of the state of the art, we discuss technological enablers and challenges and present our vision about the future applications and impact of this melting pot.


Subject(s)
Genomics , Proteomics , Genomics/methods , Proteomics/methods , Metabolomics/methods , Computational Biology , Mass Spectrometry
8.
Anal Chem ; 94(25): 8983-8991, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35708227

ABSTRACT

On-tissue chemical derivatization is a valuable tool for expanding compound coverage in untargeted metabolomic studies with matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI). Applying multiple derivatization agents in parallel increases metabolite coverage even further but results in large and more complex datasets that can be challenging to analyze. In this work, we present a pipeline to provide rigorous annotations for on-tissue derivatized MSI data using Metaspace. To test and validate the pipeline, maize roots were used as a model system to obtain MSI datasets after chemical derivatization with four different reagents, Girard's T and P for carbonyl groups, coniferyl aldehyde for primary amines, and 2-picolylamine for carboxylic acids. Using this pipeline helped us annotate 631 unique metabolites from the CornCyc/BraChem database compared to 256 in the underivatized dataset, yet, at the same time, shortening the processing time compared to manual processing and providing robust and systematic scoring and annotation. We have also developed a method to remove false derivatized annotations, which can clean 5-25% of false derivatized annotations from the derivatized data, depending on the reagent. Taken together, our pipeline facilitates the use of broadly targeted spatial metabolomics using multiple derivatization reagents.


Subject(s)
Metabolomics , Zea mays , Indicators and Reagents , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods
9.
Mol Syst Biol ; 17(7): e9833, 2021 07.
Article in English | MEDLINE | ID: mdl-34309190

ABSTRACT

Human intestinal epithelial cells form a primary barrier protecting us from pathogens, yet only limited knowledge is available about individual contribution of each cell type to mounting an immune response against infection. Here, we developed a framework combining single-cell RNA-Seq and highly multiplex RNA FISH and applied it to human intestinal organoids infected with human astrovirus, a model human enteric virus. We found that interferon controls the infection and that astrovirus infects all major cell types and lineages and induces expression of the cell proliferation marker MKI67. Intriguingly, each intestinal epithelial cell lineage exhibits a unique basal expression of interferon-stimulated genes and, upon astrovirus infection, undergoes an antiviral transcriptional reprogramming by upregulating distinct sets of interferon-stimulated genes. These findings suggest that in the human intestinal epithelium, each cell lineage plays a unique role in resolving virus infection. Our framework is applicable to other organoids and viruses, opening new avenues to unravel roles of individual cell types in viral pathogenesis.


Subject(s)
Transcriptome , Virus Diseases , Humans , Immunity , Intestinal Mucosa , Intestines
10.
Mol Syst Biol ; 17(4): e10232, 2021 04.
Article in English | MEDLINE | ID: mdl-33904651

ABSTRACT

Exacerbated pro-inflammatory immune response contributes to COVID-19 pathology. However, despite the mounting evidence about SARS-CoV-2 infecting the human gut, little is known about the antiviral programs triggered in this organ. To address this gap, we performed single-cell transcriptomics of SARS-CoV-2-infected intestinal organoids. We identified a subpopulation of enterocytes as the prime target of SARS-CoV-2 and, interestingly, found the lack of positive correlation between susceptibility to infection and the expression of ACE2. Infected cells activated strong pro-inflammatory programs and produced interferon, while expression of interferon-stimulated genes was limited to bystander cells due to SARS-CoV-2 suppressing the autocrine action of interferon. These findings reveal that SARS-CoV-2 curtails the immune response and highlights the gut as a pro-inflammatory reservoir that should be considered to fully understand SARS-CoV-2 pathogenesis.


Subject(s)
Intestines/immunology , SARS-CoV-2/physiology , Single-Cell Analysis , COVID-19/virology , Gastrointestinal Microbiome , Humans , In Situ Hybridization, Fluorescence , Organoids/metabolism , Sequence Analysis, RNA
11.
J Proteome Res ; 20(4): 2089-2097, 2021 04 02.
Article in English | MEDLINE | ID: mdl-33529026

ABSTRACT

Liquid chromatography-mass spectrometry (LC-MS)-based untargeted metabolomics studies require high-quality spectral libraries for reliable metabolite identification. We have constructed EMBL-MCF (European Molecular Biology Laboratory-Metabolomics Core Facility), an open LC-MS/MS spectral library that currently contains over 1600 fragmentation spectra from 435 authentic standards of endogenous metabolites and lipids. The unique features of the library include the presence of chromatographic profiles acquired with different LC-MS methods and coverage of different adduct ions. The library covers many biologically important metabolites with some unique metabolites and lipids as compared with other public libraries. The EMBL-MCF spectral library is created and shared using an in-house-developed web application at https://curatr.mcf.embl.de/. The library is freely available online and also integrated with other mass spectral repositories.


Subject(s)
Metabolomics , Tandem Mass Spectrometry , Chromatography, Liquid , Gene Library , Ions
12.
Physiol Genomics ; 53(1): 1-11, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33197228

ABSTRACT

Comprehensive and spatially mapped molecular atlases of organs at a cellular level are a critical resource to gain insights into pathogenic mechanisms and personalized therapies for diseases. The Kidney Precision Medicine Project (KPMP) is an endeavor to generate three-dimensional (3-D) molecular atlases of healthy and diseased kidney biopsies by using multiple state-of-the-art omics and imaging technologies across several institutions. Obtaining rigorous and reproducible results from disparate methods and at different sites to interrogate biomolecules at a single-cell level or in 3-D space is a significant challenge that can be a futile exercise if not well controlled. We describe a "follow the tissue" pipeline for generating a reliable and authentic single-cell/region 3-D molecular atlas of human adult kidney. Our approach emphasizes quality assurance, quality control, validation, and harmonization across different omics and imaging technologies from sample procurement, processing, storage, shipping to data generation, analysis, and sharing. We established benchmarks for quality control, rigor, reproducibility, and feasibility across multiple technologies through a pilot experiment using common source tissue that was processed and analyzed at different institutions and different technologies. A peer review system was established to critically review quality control measures and the reproducibility of data generated by each technology before their being approved to interrogate clinical biopsy specimens. The process established economizes the use of valuable biopsy tissue for multiomics and imaging analysis with stringent quality control to ensure rigor and reproducibility of results and serves as a model for precision medicine projects across laboratories, institutions and consortia.


Subject(s)
Guidelines as Topic , Kidney/pathology , Precision Medicine , Biopsy , Humans , Reproducibility of Results
13.
Anal Chem ; 93(40): 13421-13425, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34581565

ABSTRACT

Imaging N-glycan spatial distribution in tissues using mass spectrometry imaging (MSI) is emerging as a promising tool in biological and clinical applications. However, there is currently no high-throughput tool for visualization and molecular annotation of N-glycans in MSI data, which significantly slows down data processing and hampers the applicability of this approach. Here, we present how METASPACE, an open-source cloud engine for molecular annotation of MSI data, can be used to automatically annotate, visualize, analyze, and interpret high-resolution mass spectrometry-based spatial N-glycomics data. METASPACE is an emerging tool in spatial metabolomics, but the lack of compatible glycan databases has limited its application for comprehensive N-glycan annotations from MSI data sets. We created NGlycDB, a public database of N-glycans, by adapting available glycan databases. We demonstrate the applicability of NGlycDB in METASPACE by analyzing MALDI-MSI data from formalin-fixed paraffin-embedded (FFPE) human kidney and mouse lung tissue sections. We added NGlycDB to METASPACE for public use, thus, facilitating applications of MSI in glycobiology.


Subject(s)
Glycomics , Polysaccharides , Animals , Diagnostic Imaging , Mice , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tissue Fixation
14.
Anal Chem ; 93(8): 4066-4074, 2021 03 02.
Article in English | MEDLINE | ID: mdl-33583182

ABSTRACT

Mass spectrometry imaging (MSI) is a powerful and convenient method for revealing the spatial chemical composition of different biological samples. Molecular annotation of the detected signals is only possible if a high mass accuracy is maintained over the entire image and the m/z range. However, the change in the number of ions from pixel-to-pixel of the biological samples could lead to small fluctuations in the detected m/z-values, called mass shift. The use of internal calibration is known to offer the best solution to avoid, or at least to reduce, mass shifts. Their "a priori" selection for a global MSI acquisition is prone to false positive detection and therefore to poor recalibration. To fill this gap, this work describes an algorithm that recalibrates each spectrum individually by estimating its mass shift with the help of a list of pixel-specific internal calibrating ions, automatically generated in a data-adaptive manner (https://github.com/LaRoccaRaphael/MSI_recalibration). Through a practical example, we applied the methodology to a zebrafish whole-body section acquired at a high mass resolution to demonstrate the impact of mass shift on data analysis and the capability of our algorithm to recalibrate MSI data. In addition, we illustrate the broad applicability of the method by recalibrating 31 different public MSI data sets from METASPACE from various samples and types of MSI and show that our recalibration significantly increases the numbers of METASPACE annotations (gaining from 20 up to 400 additional annotations), particularly the high-confidence annotations with a low false discovery rate.


Subject(s)
Histological Techniques , Zebrafish , Animals , Calibration , Ions , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
15.
Bioinformatics ; 36(10): 3215-3224, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32049317

ABSTRACT

MOTIVATION: Imaging mass spectrometry (imaging MS) is a prominent technique for capturing distributions of molecules in tissue sections. Various computational methods for imaging MS rely on quantifying spatial correlations between ion images, referred to as co-localization. However, no comprehensive evaluation of co-localization measures has ever been performed; this leads to arbitrary choices and hinders method development. RESULTS: We present ColocML, a machine learning approach addressing this gap. With the help of 42 imaging MS experts from nine laboratories, we created a gold standard of 2210 pairs of ion images ranked by their co-localization. We evaluated existing co-localization measures and developed novel measures using term frequency-inverse document frequency and deep neural networks. The semi-supervised deep learning Pi model and the cosine score applied after median thresholding performed the best (Spearman 0.797 and 0.794 with expert rankings, respectively). We illustrate these measures by inferring co-localization properties of 10 273 molecules from 3685 public METASPACE datasets. AVAILABILITY AND IMPLEMENTATION: https://github.com/metaspace2020/coloc. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Machine Learning , Neural Networks, Computer , Mass Spectrometry , Software , Supervised Machine Learning
16.
Mol Syst Biol ; 16(10): e9474, 2020 10.
Article in English | MEDLINE | ID: mdl-33022142

ABSTRACT

The advent of single-cell methods is paving the way for an in-depth understanding of the cell cycle with unprecedented detail. Due to its ramifications in nearly all biological processes, the evaluation of cell cycle progression is critical for an exhaustive cellular characterization. Here, we present DeepCycle, a deep learning method for estimating a cell cycle trajectory from unsegmented single-cell microscopy images, relying exclusively on the brightfield and nuclei-specific fluorescent signals. DeepCycle was evaluated on 2.6 million single-cell microscopy images of MDCKII cells with the fluorescent FUCCI2 system. DeepCycle provided a latent representation of cell images revealing a continuous and closed trajectory of the cell cycle. Further, we validated the DeepCycle trajectories by showing its nearly perfect correlation with real time measured from live-cell imaging of cells undergoing an entire cell cycle. This is the first model able to resolve the closed cell cycle trajectory, including cell division, solely based on unsegmented microscopy data from adherent cell cultures.


Subject(s)
Cell Cycle , Image Processing, Computer-Assisted/methods , Single-Cell Analysis/methods , Time-Lapse Imaging/methods , Animals , Cell Line , Dogs , Microscopy, Fluorescence , Neural Networks, Computer
17.
BMC Bioinformatics ; 21(1): 129, 2020 Apr 03.
Article in English | MEDLINE | ID: mdl-32245392

ABSTRACT

BACKGROUND: Imaging mass spectrometry (imaging MS) is an enabling technology for spatial metabolomics of tissue sections with rapidly growing areas of applications in biology and medicine. However, imaging MS data is polluted with off-sample ions caused by sample preparation, particularly by the MALDI (matrix-assisted laser desorption/ionization) matrix application. Off-sample ion images confound and hinder statistical analysis, metabolite identification and downstream analysis with no automated solutions available. RESULTS: We developed an artificial intelligence approach to recognize off-sample ion images. First, we created a high-quality gold standard of 23,238 expert-tagged ion images from 87 public datasets from the METASPACE knowledge base. Next, we developed several machine and deep learning methods for recognizing off-sample ion images. The following methods were able to reproduce expert judgements with a high agreement: residual deep learning (F1-score 0.97), semi-automated spatio-molecular biclustering (F1-score 0.96), and molecular co-localization (F1-score 0.90). In a test-case study, we investigated off-sample images corresponding to the most common MALDI matrix (2,5-dihydroxybenzoic acid, DHB) and characterized properties of matrix clusters. CONCLUSIONS: Overall, our work illustrates how artificial intelligence approaches enabled by open-access data, web technologies, and machine and deep learning open novel avenues to address long-standing challenges in imaging MS.


Subject(s)
Machine Learning , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Deep Learning , Gentisates/chemistry
18.
Nat Methods ; 14(1): 57-60, 2017 01.
Article in English | MEDLINE | ID: mdl-27842059

ABSTRACT

High-mass-resolution imaging mass spectrometry promises to localize hundreds of metabolites in tissues, cell cultures, and agar plates with cellular resolution, but it is hampered by the lack of bioinformatics tools for automated metabolite identification. We report pySM, a framework for false discovery rate (FDR)-controlled metabolite annotation at the level of the molecular sum formula, for high-mass-resolution imaging mass spectrometry (https://github.com/alexandrovteam/pySM). We introduce a metabolite-signal match score and a target-decoy FDR estimate for spatial metabolomics.


Subject(s)
Brain/metabolism , Computational Biology/methods , Mass Spectrometry/methods , Metabolome , Metabolomics/methods , Molecular Imaging/methods , Software , Animals , Brain/cytology , Chromatography, Liquid , False Positive Reactions , Female , Mice , Mice, Inbred C57BL
19.
Cytometry A ; 97(3): 288-295, 2020 03.
Article in English | MEDLINE | ID: mdl-31872957

ABSTRACT

Technologies such as microscopy, sequential hybridization, and mass spectrometry enable quantitative single-cell phenotypic and molecular measurements in situ. Deciphering spatial phenotypic and molecular effects on the single-cell level is one of the grand challenges and a key to understanding the effects of cell-cell interactions and microenvironment. However, spatial information is usually overlooked by downstream data analyses, which usually consider single-cell read-out values as independent measurements for further averaging or clustering, thus disregarding spatial locations. With this work, we attempt to fill this gap. We developed a toolbox that allows one to test for the presence of a spatial effect in microscopy images of adherent cells and estimate the spatial scale of this effect. The proposed Python module can be used for any light microscopy images of cells as well as other types of single-cell data such as in situ transcriptomics or metabolomics. The input format of our package matches standard output formats from image analysis tools such as CellProfiler, Fiji, or Icy and thus makes our toolbox easy and straightforward to use, yet offering a powerful statistical approach for a wide range of applications. © 2019 International Society for Advancement of Cytometry.


Subject(s)
Image Processing, Computer-Assisted , Microscopy , Cluster Analysis , Mass Spectrometry , Spatial Analysis
20.
Metabolomics ; 16(1): 11, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31925564

ABSTRACT

INTRODUCTION: Diabetic kidney disease (DKD) is the most prevalent complication in diabetic patients, which contributes to high morbidity and mortality. Urine and plasma metabolomics studies have been demonstrated to provide valuable insights for DKD. However, limited information on spatial distributions of metabolites in kidney tissues have been reported. OBJECTIVES: In this work, we employed an ambient desorption electrospray ionization-mass spectrometry imaging (DESI-MSI) coupled to a novel bioinformatics platform (METASPACE) to characterize the metabolome in a mouse model of DKD. METHODS: DESI-MSI was performed for spatial untargeted metabolomics analysis in kidneys of mouse models (F1 C57BL/6J-Ins2Akita male mice at 17 weeks of age) of type 1 diabetes (T1D, n = 5) and heathy controls (n = 6). RESULTS: Multivariate analyses (i.e., PCA and PLS-DA (a 2000 permutation test: P < 0.001)) showed clearly separated clusters for the two groups of mice on the basis of 878 measured m/z's in kidney cortical tissues. Specifically, mice with T1D had increased relative abundances of pseudouridine, accumulation of free polyunsaturated fatty acids (PUFAs), and decreased relative abundances of cardiolipins in cortical proximal tubules when compared with healthy controls. CONCLUSION: Results from the current study support potential key roles of pseudouridine and cardiolipins for maintaining normal RNA structure and normal mitochondrial function, respectively, in cortical proximal tubules with DKD. DESI-MSI technology coupled with METASPACE could serve as powerful new tools to provide insight on fundamental pathways in DKD.


Subject(s)
Diabetic Nephropathies/metabolism , Kidney Tubules, Proximal/metabolism , Metabolome , Mitochondrial Membranes/metabolism , Animals , Cardiolipins/metabolism , Computational Biology , Fatty Acids, Omega-3/metabolism , Male , Metabolic Networks and Pathways , Mice , Mice, Inbred C57BL , Pseudouridine/metabolism , Spectrometry, Mass, Electrospray Ionization
SELECTION OF CITATIONS
SEARCH DETAIL