1
|
Prabhakaran S, Yapp C, Baker GJ, Beyer J, Chang YH, Creason AL, Krueger R, Muhlich J, Patterson NH, Sidak K, Sudar D, Taylor AJ, Ternes L, Troidl J, Yubin X, Sokolov A, Tyson DR. Addressing persistent challenges in digital image analysis of cancer tissue: resources developed from a hackathon. Mol Oncol 2025. [PMID: 39927650 DOI: 10.1002/1878-0261.13783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 10/17/2024] [Accepted: 11/11/2024] [Indexed: 02/11/2025] Open
Abstract
The National Cancer Institute (NCI) supports numerous research consortia that rely on imaging technologies to study cancerous tissues. To foster collaboration and innovation in this field, the Image Analysis Working Group (IAWG) was created in 2019. As multiplexed imaging techniques grow in scale and complexity, more advanced computational methods are required beyond traditional approaches like segmentation and pixel intensity quantification. In 2022, the IAWG held a virtual hackathon focused on addressing challenges in analyzing complex, high-dimensional datasets from fixed cancer tissues. The hackathon addressed key challenges in three areas: (1) cell type classification and assessment, (2) spatial data visualization and translation, and (3) scaling image analysis for large, multi-terabyte datasets. Participants explored the limitations of current automated analysis tools, developed potential solutions, and made significant progress during the hackathon. Here we provide a summary of the efforts and resultant resources and highlight remaining challenges facing the research community as emerging technologies are integrated into diverse imaging modalities and data analysis platforms.
Collapse
Affiliation(s)
| | - Clarence Yapp
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Gregory J Baker
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Johanna Beyer
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Young Hwan Chang
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Allison L Creason
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | | | | | | | | | - Damir Sudar
- Quantitative Imaging Systems, Monroeville, PA, USA
| | | | - Luke Ternes
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Jakob Troidl
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Xie Yubin
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Artem Sokolov
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Darren R Tyson
- Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
2
|
Liu Y, Li Y, Wu R, Wang Y, Li P, Jiang T, Wang K, Liu Y, Cheng Z. Epithelial and immune transcriptomic characteristics and possible regulatory mechanisms in asthma exacerbation: insights from integrated studies. Front Immunol 2025; 16:1512053. [PMID: 39917297 PMCID: PMC11798785 DOI: 10.3389/fimmu.2025.1512053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/02/2025] [Indexed: 02/09/2025] Open
Abstract
Background Asthma exacerbation significantly contribute to disease mortality and result in heightened health care expenditures. This study was aimed at gaining important new insights into the heterogeneity of epithelial and immune cells and elucidating key regulatory genes involved in the pathogenesis of asthma exacerbation. Methods Functional enrichment, pseudotime, metabolism and cell-cell communication analyses of epithelial cells and immune cells in single-cell RNA sequencing (scRNA-seq) dataset were applied. Immune infiltration analysis was performed in bulk RNA sequencing (bulk RNA-seq) dataset. Key regulatory genes were obtained by taking the intersection of the differentially expressed genes (DEGs) between control and asthma group in epithelial cells, immune cells and bulk RNA-seq data. Asthma animal and in vitro cell line models were established to verify the key regulatory genes expression by employing quantitative reverse transcription polymerase chain reaction (qRT-PCR). Results ScRNA-seq analysis identified 7 epithelial subpopulations and 14 distinct immune cell types based on gene expression profiles. Further analysis demonstrated that these cells manifested high heterogeneity at the levels of functional variations, dynamics, communication patterns and metabolic changes. Notably, TMPRSS11A, TUBA1A, SCEL, ICAM4, TMPRSS11B, IGFBP2, CLC, NFAM1 and F13A1 were identified as key regulatory genes of asthma. The results of the qRT-PCR demonstrated that the 9 key regulatory genes were involved in asthma. Conclusions We systematically explored epithelial and immune characteristics in asthma exacerbation and identified 9 key regulatory genes underlying asthma occurrence and progression, which may be valuable for providing new insights into the cellular and molecular mechanisms driving asthma exacerbations.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Zhe Cheng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of
Zhengzhou University, Zhengzhou, He’nan, China
| |
Collapse
|
3
|
Strober BJ, Tayeb K, Popp J, Qi G, Gordon MG, Perez R, Ye CJ, Battle A. SURGE: uncovering context-specific genetic-regulation of gene expression from single-cell RNA sequencing using latent-factor models. Genome Biol 2024; 25:28. [PMID: 38254214 PMCID: PMC10801966 DOI: 10.1186/s13059-023-03152-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 12/20/2023] [Indexed: 01/24/2024] Open
Abstract
Genetic regulation of gene expression is a complex process, with genetic effects known to vary across cellular contexts such as cell types and environmental conditions. We developed SURGE, a method for unsupervised discovery of context-specific expression quantitative trait loci (eQTLs) from single-cell transcriptomic data. This allows discovery of the contexts or cell types modulating genetic regulation without prior knowledge. Applied to peripheral blood single-cell eQTL data, SURGE contexts capture continuous representations of distinct cell types and groupings of biologically related cell types. We demonstrate the disease-relevance of SURGE context-specific eQTLs using colocalization analysis and stratified LD-score regression.
Collapse
Affiliation(s)
- Benjamin J Strober
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Karl Tayeb
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Joshua Popp
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Guanghao Qi
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - M Grace Gordon
- Biological and Medical Informatics Graduate Program, University of California, San Francisco, CA, USA
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA
| | - Richard Perez
- Institute for Human Genetics, University of California, San Francisco, CA, USA
| | - Chun Jimmie Ye
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA
- Institute for Computational Health Sciences, University of California, San Francisco, San Francisco, CA, USA
- Chan-Zuckerberg Biohub, San Francisco, CA, USA
| | - Alexis Battle
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
- Department of Computer Science, Johns Hopkins University, Baltimore, MD, USA.
- Department of Genetic Medicine, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
4
|
Neufeld A, Gao LL, Popp J, Battle A, Witten D. Inference after latent variable estimation for single-cell RNA sequencing data. Biostatistics 2023; 25:270-287. [PMID: 36511385 DOI: 10.1093/biostatistics/kxac047] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/17/2022] [Accepted: 11/26/2022] [Indexed: 12/14/2022] Open
Abstract
In the analysis of single-cell RNA sequencing data, researchers often characterize the variation between cells by estimating a latent variable, such as cell type or pseudotime, representing some aspect of the cell's state. They then test each gene for association with the estimated latent variable. If the same data are used for both of these steps, then standard methods for computing p-values in the second step will fail to achieve statistical guarantees such as Type 1 error control. Furthermore, approaches such as sample splitting that can be applied to solve similar problems in other settings are not applicable in this context. In this article, we introduce count splitting, a flexible framework that allows us to carry out valid inference in this setting, for virtually any latent variable estimation technique and inference approach, under a Poisson assumption. We demonstrate the Type 1 error control and power of count splitting in a simulation study and apply count splitting to a data set of pluripotent stem cells differentiating to cardiomyocytes.
Collapse
Affiliation(s)
- Anna Neufeld
- Department of Statistics, University of Washington, Seattle, WA 98195, USA
| | - Lucy L Gao
- Department of Statistics, University of British Columbia, BC V6T 1Z4, Canada
| | - Joshua Popp
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Alexis Battle
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA and Department of Computer Science, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Daniela Witten
- Department of Statistics, University of Washington, Seattle, WA 98195, USA and Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
5
|
Cao F, Liu Y, Cheng Y, Wang Y, He Y, Xu Y. Multi-omics characteristics of tumor-associated macrophages in the tumor microenvironment of gastric cancer and their exploration of immunotherapy potential. Sci Rep 2023; 13:18265. [PMID: 37880233 PMCID: PMC10600170 DOI: 10.1038/s41598-023-38822-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/15/2023] [Indexed: 10/27/2023] Open
Abstract
The incidence and mortality rate of gastric cancer (GC) have remained high worldwide. Although some progress has been made in immunotargeted therapy, the treatment effect remains limited. With more attention has been paid to the immune potential of tumor-associated macrophages (TAMs), but the specific mechanisms of tumor immunity are still unclear. Thus, we screened marker genes in TAMs differentiation (MDMs) through single-cell RNA sequencing, and combined with GC transcriptome data from TCGA and GEO databases, the clinical and TME characteristics, prognostic differences, immune infiltration, and drug sensitivity among different subtypes of patients with GC in different data sets were analyzed. A prognostic model of GC was constructed to evaluate the prognosis and immunotherapy response of patients with GC. In this study, we extensively studied the mutations in MDMs such as CGN, S100A6, and C1QA, and found differences in the infiltration of immune cells and immune checkpoints including M2 TAMs, T cells, CD274, and CTLA4 in different GC subtypes. In the model, we constructed a predictive scoring system with high accuracy and screened out key MDMs-related genes associated with prognosis and M2 TAMs, among which VKORC1 may be involved in GC progression and iron death in tumor cells. Therefore, this study explores the therapeutic strategy of TAMs reprogramming in-depth, providing new ideas for the clinical diagnosis, treatment, and prognosis assessment of GC.
Collapse
Affiliation(s)
- Feng Cao
- Department of General Surgery, University Hospital RWTH Aachen, 52074, Aachen, Germany
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yanwei Liu
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yunsheng Cheng
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yong Wang
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| | - Yan He
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| | - Yanyan Xu
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| |
Collapse
|
6
|
Deng Y, Tang S, Cheng J, Zhang X, Jing D, Lin Z, Zhou J. Integrated analysis reveals Atf3 promotes neuropathic pain via orchestrating JunB mediated release of inflammatory cytokines in DRG macrophage. Life Sci 2023; 329:121939. [PMID: 37451398 DOI: 10.1016/j.lfs.2023.121939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/08/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
The dorsal root ganglion (DRG) is actively involved in the development of neuropathic pain (NP), serving as an intermediate station for pain signals from the peripheral nervous system to the central nervous system. The mechanism by which DRG is involved in NP regulation is not fully understood. The immune system plays a pivotal role in the physiological and pathological states of the human body. In recent years, the immune system has been thought to play an increasingly important role in the pathogenesis of NP. The immune system plays a key role in pain through specific immune cells and their immune-related genes (IRGs). However, the mechanism by which IRGs of DRG regulate NP action has not been fully elucidated. Here, we performed Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses of IRGs in DRG bulk-RNA sequencing data from spared nerve injury (SNI) model mice and found that their IRGs were enriched in many pathways, especially in the immune response pathway. Subsequently, we analyzed single-cell RNA sequencing (scRNA-seq) data from DRGs extracted from the SNI model and identified eight cell populations. Among them, the highest IRG activity was presented in macrophages. Next, we analyzed the scRNA and bulk-sequencing data and deduced five common transcription factors (TFs) from differentially expressed genes (DEGs). The protein-protein interaction (PPI) network suggested that Atf3 and JunB are closely related. In vitro experiments, we verified that the protein and mRNA expressions of Atf3 and JunB were up-regulated in macrophages after lipopolysaccharide (LPS) stimulation. Moreover, the down-regulation of Atf3 reduced the release of inflammatory cytokines and decreased the protein and mRNA expression levels of JunB. The down-regulation of JunB also reduced the release of inflammatory cytokines. Furthermore, overexpression of JunB attenuated the effect of Atf3 down-regulation in reducing the release of inflammatory cytokines. Therefore, we speculated that Atf3 might promote NP through JunB-mediated release of inflammatory factors in DRG macrophages.
Collapse
Affiliation(s)
- Yingdong Deng
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province 510000, China
| | - Simin Tang
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province 510000, China
| | - Jiurong Cheng
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province 510000, China
| | - Xiangsheng Zhang
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province 510000, China
| | - Danqin Jing
- College of Anesthesiology, Shanxi Medical University, Taiyuan, Shanxi Province 030001, China
| | - Ziqiang Lin
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province 510000, China
| | - Jun Zhou
- Department of Anesthesiology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province 510000, China.
| |
Collapse
|
7
|
Yin Y, Wu S, Niu L, Huang S. Atonal homolog 7 (ATOH7) confers neuroprotection for photoreceptor cells in glaucoma via inhibition of the notch pathway. J Neurochem 2023; 166:847-861. [PMID: 37526008 DOI: 10.1111/jnc.15905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/13/2023] [Accepted: 06/20/2023] [Indexed: 08/02/2023]
Abstract
Single-cell RNA sequencing (scRNA-seq) technologies enable the profiling and analysis of the transcriptomes of single cells and hold promise for clarifying gene mechanisms at single-cell resolution. We based this study on scRNA-seq data to reveal glaucoma-related genes and downstream pathways with neuroprotection effects. The scRNA-seq datasets related to glaucoma of retinal tissue samples of human beings and Atonal Homolog 7 (ATOH7)-null mice were obtained from the GEO database. The 74 top marker genes and 20 cell clusters were obtained in human retinal tissue samples. The key gene ATOH7 was found after the intersection with genes from GeneCards data. In the ATOH7-null mouse retinal tissue samples, pseudotime inference demonstrated significant changes in cell differentiation. Moreover, mouse retinal photoreceptor cells (PRCs) were cultured and treated with lentivirus carrying oe-ATOH7 alone or in combination with Notch signaling pathway activator Jagged-1/FC, after which cell biological functions were determined. The involvement of ATOH7 in glaucoma was identified through regulating PRCs. Furthermore, ATOH7 conferred neuroprotection in PRCs in glaucoma by mediating the Notch signaling pathway. In vitro data confirmed that ATOH7 overexpression promoted the differentiation of PRCs and inhibited their apoptosis by suppressing the Notch signaling pathway. The evidence provided by our study highlighted the involvement of ATOH7 in the blockade of the Notch signaling pathway, resulting in the neuroprotection for PRCs in glaucoma.
Collapse
Affiliation(s)
- Yuan Yin
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, People's Republic of China
| | - Shuai Wu
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, People's Republic of China
| | - Lingzhi Niu
- Department of Ophthalmology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, People's Republic of China
| | - Shiwei Huang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, People's Republic of China
| |
Collapse
|
8
|
Prabhakaran S, Yapp C, Baker GJ, Beyer J, Chang YH, Creason AL, Krueger R, Muhlich J, Patterson NH, Sidak K, Sudar D, Taylor AJ, Ternes L, Troidl J, Xie Y, Sokolov A, Tyson DR. Addressing persistent challenges in digital image analysis of cancerous tissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.21.548450. [PMID: 37547011 PMCID: PMC10401923 DOI: 10.1101/2023.07.21.548450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
The National Cancer Institute (NCI) supports many research programs and consortia, many of which use imaging as a major modality for characterizing cancerous tissue. A trans-consortia Image Analysis Working Group (IAWG) was established in 2019 with a mission to disseminate imaging-related work and foster collaborations. In 2022, the IAWG held a virtual hackathon focused on addressing challenges of analyzing high dimensional datasets from fixed cancerous tissues. Standard image processing techniques have automated feature extraction, but the next generation of imaging data requires more advanced methods to fully utilize the available information. In this perspective, we discuss current limitations of the automated analysis of multiplexed tissue images, the first steps toward deeper understanding of these limitations, what possible solutions have been developed, any new or refined approaches that were developed during the Image Analysis Hackathon 2022, and where further effort is required. The outstanding problems addressed in the hackathon fell into three main themes: 1) challenges to cell type classification and assessment, 2) translation and visual representation of spatial aspects of high dimensional data, and 3) scaling digital image analyses to large (multi-TB) datasets. We describe the rationale for each specific challenge and the progress made toward addressing it during the hackathon. We also suggest areas that would benefit from more focus and offer insight into broader challenges that the community will need to address as new technologies are developed and integrated into the broad range of image-based modalities and analytical resources already in use within the cancer research community.
Collapse
|
9
|
Chen YT, Witten DM. Selective inference for k-means clustering. JOURNAL OF MACHINE LEARNING RESEARCH : JMLR 2023; 24:152. [PMID: 38264325 PMCID: PMC10805457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
We consider the problem of testing for a difference in means between clusters of observations identified via k -means clustering. In this setting, classical hypothesis tests lead to an inflated Type I error rate. In recent work, Gao et al. (2022) considered a related problem in the context of hierarchical clustering. Unfortunately, their solution is highly-tailored to the context of hierarchical clustering, and thus cannot be applied in the setting of k -means clustering. In this paper, we propose a p-value that conditions on all of the intermediate clustering assignments in the k -means algorithm. We show that the p-value controls the selective Type I error for a test of the difference in means between a pair of clusters obtained using k -means clustering in finite samples, and can be efficiently computed. We apply our proposal on hand-written digits data and on single-cell RNA-sequencing data.
Collapse
Affiliation(s)
- Yiqun T Chen
- Data Science Institute and Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
| | - Daniela M Witten
- Departments of Statistics and Biostatistics, University of Washington, Seattle, WA 98195-4322, USA
| |
Collapse
|
10
|
Dreishpoon MB, Bick NR, Petrova B, Warui DM, Cameron A, Booker SJ, Kanarek N, Golub TR, Tsvetkov P. FDX1 regulates cellular protein lipoylation through direct binding to LIAS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.03.526472. [PMID: 36778498 PMCID: PMC9915701 DOI: 10.1101/2023.02.03.526472] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Ferredoxins are a family of iron-sulfur (Fe-S) cluster proteins that serve as essential electron donors in numerous cellular processes that are conserved through evolution. The promiscuous nature of ferredoxins as electron donors enables them to participate in many metabolic processes including steroid, heme, vitamin D and Fe-S cluster biosynthesis in different organisms. However, the unique natural function(s) of each of the two human ferredoxins (FDX1 and FDX2) are still poorly characterized. We recently reported that FDX1 is both a crucial regulator of copper ionophore induced cell death and serves as an upstream regulator of cellular protein lipoylation, a mitochondrial lipid-based post translational modification naturally occurring on four mitochondrial enzymes that are crucial for TCA cycle function. Here we show that FDX1 regulates protein lipoylation by directly binding to the lipoyl synthase (LIAS) enzyme and not through indirect regulation of cellular Fe-S cluster biosynthesis. Metabolite profiling revealed that the predominant cellular metabolic outcome of FDX1 loss-of-function is manifested through the regulation of the four lipoylation-dependent enzymes ultimately resulting in loss of cellular respiration and sensitivity to mild glucose starvation. Transcriptional profiling of cells growing in either normal or low glucose conditions established that FDX1 loss-of-function results in the induction of both compensatory metabolism related genes and the integrated stress response, consistent with our findings that FDX1 loss-of-functions is conditionally lethal. Together, our findings establish that FDX1 directly engages with LIAS, promoting cellular protein lipoylation, a process essential in maintaining cell viability under low glucose conditions.
Collapse
Affiliation(s)
| | | | - Boryana Petrova
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Boston Children’s Hospital, Boston, MA USA
| | - Douglas M. Warui
- Department of Chemistry and Biochemistry and Molecular Biology and the Howard Hughes Medical Institute, The Pennsylvania State University, PA, United States
| | | | - Squire J. Booker
- Department of Chemistry and Biochemistry and Molecular Biology and the Howard Hughes Medical Institute, The Pennsylvania State University, PA, United States
| | - Naama Kanarek
- Broad Institute of Harvard and MIT, Cambridge, USA
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Boston Children’s Hospital, Boston, MA USA
| | - Todd R. Golub
- Broad Institute of Harvard and MIT, Cambridge, USA
- Harvard Medical School, Boston, MA, USA
- Department of Pediatric Oncology, Dana Farber Cancer Institute, Boston, MA, USA
- Division of Pediatric Hematology/Oncology, Boston Children’s Hospital, Boston, MA, USA
| | | |
Collapse
|
11
|
Freeman TC, Horsewell S, Patir A, Harling-Lee J, Regan T, Shih BB, Prendergast J, Hume DA, Angus T. Graphia: A platform for the graph-based visualisation and analysis of high dimensional data. PLoS Comput Biol 2022; 18:e1010310. [PMID: 35877685 PMCID: PMC9352203 DOI: 10.1371/journal.pcbi.1010310] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/04/2022] [Accepted: 06/16/2022] [Indexed: 01/04/2023] Open
Abstract
Graphia is an open-source platform created for the graph-based analysis of the huge amounts of quantitative and qualitative data currently being generated from the study of genomes, genes, proteins metabolites and cells. Core to Graphia’s functionality is support for the calculation of correlation matrices from any tabular matrix of continuous or discrete values, whereupon the software is designed to rapidly visualise the often very large graphs that result in 2D or 3D space. Following graph construction, an extensive range of measurement algorithms, routines for graph transformation, and options for the visualisation of node and edge attributes are available, for graph exploration and analysis. Combined, these provide a powerful solution for the interpretation of high-dimensional data from many sources, or data already in the form of a network or equivalent adjacency matrix. Several use cases of Graphia are described, to showcase its wide range of applications in the analysis biological data. Graphia runs on all major desktop operating systems, is extensible through the deployment of plugins and is freely available to download from https://graphia.app/. Graphia is a new visual analytics platform specifically created for the network-based analysis of large and complex data, such as that generated in huge amounts by modern biological analyses. It works in a data agnostic, hypothesis-free manner to generate correlation networks from any table of numerical or discrete values, thereafter providing a means to rapidly visualise the often very large networks that result, in either 2D or 3D space. Following network construction, the tool offers an extensive range of analysis algorithms, routines for network transformation, and options for the visualisation of metadata. This provides a powerful analysis solution for the exploration and interpretation of high-dimensional data from any source, as well as any data already defined as a network. Several use cases of Graphia are described to showcase its wide range of applications in the analysis biological data. Graphia is open source and free to all.
Collapse
Affiliation(s)
- Tom C. Freeman
- The Roslin Institute, Easter Bush Campus, The University of Edinburgh, Edinburgh, United Kingdom
- Kajeka Limited, Roslin Innovation Centre, Easter Bush Campus, The University of Edinburgh, Edinburgh, United Kingdom
- * E-mail:
| | - Sebastian Horsewell
- Kajeka Limited, Roslin Innovation Centre, Easter Bush Campus, The University of Edinburgh, Edinburgh, United Kingdom
| | - Anirudh Patir
- The Roslin Institute, Easter Bush Campus, The University of Edinburgh, Edinburgh, United Kingdom
| | - Josh Harling-Lee
- The Roslin Institute, Easter Bush Campus, The University of Edinburgh, Edinburgh, United Kingdom
| | - Tim Regan
- The Roslin Institute, Easter Bush Campus, The University of Edinburgh, Edinburgh, United Kingdom
| | - Barbara B. Shih
- The Roslin Institute, Easter Bush Campus, The University of Edinburgh, Edinburgh, United Kingdom
| | - James Prendergast
- The Roslin Institute, Easter Bush Campus, The University of Edinburgh, Edinburgh, United Kingdom
| | - David A. Hume
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Tim Angus
- Kajeka Limited, Roslin Innovation Centre, Easter Bush Campus, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
12
|
Abstract
Spatial transcriptomic technologies have been developed rapidly in recent years. The addition of spatial context to expression data holds the potential to revolutionize many fields in biology. However, the lack of computational tools remains a bottleneck that is preventing the broader utilization of these technologies. Recently, we have developed Giotto as a comprehensive, generally applicable, and user-friendly toolbox for spatial transcriptomic data analysis and visualization. Giotto implements a rich set of algorithms to enable robust spatial data analysis. To help users get familiar with the Giotto environment and apply it effectively in analyzing new datasets, we will describe the detailed protocols for applying Giotto without any advanced programming skills. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Getting Giotto set up for use Basic Protocol 2: Pre-processing Basic Protocol 3: Clustering and cell-type identification Basic Protocol 4: Cell-type enrichment and deconvolution analyses Basic Protocol 5: Spatial structure analysis tools Basic Protocol 6: Spatial domain detection by using a hidden Markov random field model Support Protocol 1: Spatial proximity-associated cell-cell interactions Support Protocol 2: Assembly of a registered 3D Giotto object from 2D slices.
Collapse
Affiliation(s)
- Natalie Del Rossi
- Department of Genetics and Genomic Sciences. Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Jiaji G. Chen
- Section of Hematology and Medical Oncology, School of Medicine, Boston University, Boston, Massachusetts 02138, USA
| | - Guo-Cheng Yuan
- Department of Genetics and Genomic Sciences. Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Ruben Dries
- Section of Hematology and Medical Oncology, School of Medicine, Boston University, Boston, Massachusetts 02138, USA
- Division of Computational Biomedicine, School of Medicine, Boston University, Boston, Massachusetts 02138, USA
| |
Collapse
|
13
|
Saturation variant interpretation using CRISPR prime editing. Nat Biotechnol 2022; 40:885-895. [DOI: 10.1038/s41587-021-01201-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 12/20/2021] [Indexed: 12/13/2022]
|
14
|
Clarke ZA, Andrews TS, Atif J, Pouyabahar D, Innes BT, MacParland SA, Bader GD. Tutorial: guidelines for annotating single-cell transcriptomic maps using automated and manual methods. Nat Protoc 2021; 16:2749-2764. [PMID: 34031612 DOI: 10.1038/s41596-021-00534-0] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 03/12/2021] [Indexed: 11/09/2022]
Abstract
Single-cell transcriptomics can profile thousands of cells in a single experiment and identify novel cell types, states and dynamics in a wide variety of tissues and organisms. Standard experimental protocols and analysis workflows have been developed to create single-cell transcriptomic maps from tissues. This tutorial focuses on how to interpret these data to identify cell types, states and other biologically relevant patterns with the objective of creating an annotated map of cells. We recommend a three-step workflow including automatic cell annotation (wherever possible), manual cell annotation and verification. Frequently encountered challenges are discussed, as well as strategies to address them. Guiding principles and specific recommendations for software tools and resources that can be used for each step are covered, and an R notebook is included to help run the recommended workflow. Basic familiarity with computer software is assumed, and basic knowledge of programming (e.g., in the R language) is recommended.
Collapse
Affiliation(s)
- Zoe A Clarke
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.,The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Tallulah S Andrews
- The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada.,Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, Ontario, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Jawairia Atif
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, Ontario, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Delaram Pouyabahar
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.,The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Brendan T Innes
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada.,The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Sonya A MacParland
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, Toronto, Ontario, Canada. .,Department of Immunology, University of Toronto, Toronto, Ontario, Canada. .,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.
| | - Gary D Bader
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada. .,The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada. .,Department of Computer Science, University of Toronto, Toronto, Ontario, Canada. .,Lunenfeld-Tanenbaum Research Institute, Toronto, Ontario, Canada.
| |
Collapse
|
15
|
Mimouna S, Rollins DA, Shibu G, Tharmalingam B, Deochand DK, Chen X, Oliver D, Chinenov Y, Rogatsky I. Transcription cofactor GRIP1 differentially affects myeloid cell-driven neuroinflammation and response to IFN-β therapy. J Exp Med 2021; 218:e20192386. [PMID: 33045064 PMCID: PMC7555412 DOI: 10.1084/jem.20192386] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 07/29/2020] [Accepted: 09/04/2020] [Indexed: 11/18/2022] Open
Abstract
Macrophages (MФ) and microglia (MG) are critical in the pathogenesis of multiple sclerosis (MS) and its mouse model, experimental autoimmune encephalomyelitis (EAE). Glucocorticoids (GCs) and interferon β (IFN-β) are frontline treatments for MS, and disrupting each pathway in mice aggravates EAE. Glucocorticoid receptor-interacting protein 1 (GRIP1) facilitates both GR and type I IFN transcriptional actions; hence, we evaluated the role of GRIP1 in neuroinflammation. Surprisingly, myeloid cell-specific loss of GRIP1 dramatically reduced EAE severity, immune cell infiltration of the CNS, and MG activation and demyelination specifically during the neuroinflammatory phase of the disease, yet also blunted therapeutic properties of IFN-β. MФ/MG transcriptome analyses at the bulk and single-cell levels revealed that GRIP1 deletion attenuated nuclear receptor, inflammatory and, interestingly, type I IFN pathways and promoted the persistence of a homeostatic MG signature. Together, these results uncover the multifaceted function of type I IFN in MS/EAE pathogenesis and therapy, and an unexpectedly permissive role of myeloid cell GRIP1 in neuroinflammation.
Collapse
Affiliation(s)
- Sanda Mimouna
- The David Z. Rosensweig Genomics Center, Hospital for Special Surgery Research Institute, New York, NY
| | - David A. Rollins
- The David Z. Rosensweig Genomics Center, Hospital for Special Surgery Research Institute, New York, NY
- Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, NY
| | - Gayathri Shibu
- The David Z. Rosensweig Genomics Center, Hospital for Special Surgery Research Institute, New York, NY
- Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, NY
| | - Bowranigan Tharmalingam
- The David Z. Rosensweig Genomics Center, Hospital for Special Surgery Research Institute, New York, NY
| | - Dinesh K. Deochand
- The David Z. Rosensweig Genomics Center, Hospital for Special Surgery Research Institute, New York, NY
| | - Xi Chen
- The David Z. Rosensweig Genomics Center, Hospital for Special Surgery Research Institute, New York, NY
- Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, NY
| | - David Oliver
- The David Z. Rosensweig Genomics Center, Hospital for Special Surgery Research Institute, New York, NY
| | - Yurii Chinenov
- The David Z. Rosensweig Genomics Center, Hospital for Special Surgery Research Institute, New York, NY
| | - Inez Rogatsky
- The David Z. Rosensweig Genomics Center, Hospital for Special Surgery Research Institute, New York, NY
- Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, NY
| |
Collapse
|
16
|
Gustafsson J, Robinson J, Inda-Díaz JS, Björnson E, Jörnsten R, Nielsen J. DSAVE: Detection of misclassified cells in single-cell RNA-Seq data. PLoS One 2020; 15:e0243360. [PMID: 33270740 PMCID: PMC7714356 DOI: 10.1371/journal.pone.0243360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 11/19/2020] [Indexed: 11/19/2022] Open
Abstract
Single-cell RNA sequencing has become a valuable tool for investigating cell types in complex tissues, where clustering of cells enables the identification and comparison of cell populations. Although many studies have sought to develop and compare different clustering approaches, a deeper investigation into the properties of the resulting populations is lacking. Specifically, the presence of misclassified cells can influence downstream analyses, highlighting the need to assess subpopulation purity and to detect such cells. We developed DSAVE (Down-SAmpling based Variation Estimation), a method to evaluate the purity of single-cell transcriptome clusters and to identify misclassified cells. The method utilizes down-sampling to eliminate differences in sampling noise and uses a log-likelihood based metric to help identify misclassified cells. In addition, DSAVE estimates the number of cells needed in a population to achieve a stable average gene expression profile within a certain gene expression range. We show that DSAVE can be used to find potentially misclassified cells that are not detectable by similar tools and reveal the cause of their divergence from the other cells, such as differing cell state or cell type. With the growing use of single-cell RNA-seq, we foresee that DSAVE will be an increasingly useful tool for comparing and purifying subpopulations in single-cell RNA-Seq datasets.
Collapse
Affiliation(s)
- Johan Gustafsson
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
- Wallenberg Center for Protein Research, Chalmers University of Technology, Gothenburg, Sweden
| | - Jonathan Robinson
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
- Wallenberg Center for Protein Research, Chalmers University of Technology, Gothenburg, Sweden
| | - Juan S. Inda-Díaz
- Mathematical Sciences, University of Gothenburg and Chalmers University of Technology, Gothenburg, Sweden
| | - Elias Björnson
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory for Cardiovascular and Metabolic Research, University of Gothenburg, Gothenburg, Sweden
| | - Rebecka Jörnsten
- Mathematical Sciences, University of Gothenburg and Chalmers University of Technology, Gothenburg, Sweden
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
- Wallenberg Center for Protein Research, Chalmers University of Technology, Gothenburg, Sweden
- BioInnovation Institute, Copenhagen, Denmark
- * E-mail:
| |
Collapse
|
17
|
Chung NC, Choi H, Wang D, Mirza B, Pelletier AR, Sigdel D, Wang W, Ping P. Identifying temporal molecular signatures underlying cardiovascular diseases: A data science platform. J Mol Cell Cardiol 2020; 145:54-58. [PMID: 32504647 PMCID: PMC7583079 DOI: 10.1016/j.yjmcc.2020.05.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/18/2020] [Accepted: 05/31/2020] [Indexed: 11/26/2022]
Abstract
OBJECTIVE During cardiovascular disease progression, molecular systems of myocardium (e.g., a proteome) undergo diverse and distinct changes. Dynamic, temporally-regulated alterations of individual molecules underlie the collective response of the heart to pathological drivers and the ultimate development of pathogenesis. Advances in high-throughput omics technologies have enabled cost-effective, temporal profiling of targeted systems in animal models of human diseases. However, computational analysis of temporal patterns from omics data remains challenging. In particular, bioinformatic pipelines involving unsupervised statistical approaches to support cardiovascular investigations are lacking, which hinders one's ability to extract biomedical insights from these complex datasets. APPROACH AND RESULTS We developed a non-parametric data analysis platform to resolve computational challenges unique to temporal omics datasets. Our platform consists of three modules. Module I preprocesses the temporal data using either cubic splines or principal component analysis (PCA), and it simultaneously accomplishes the tasks on missing data imputation and denoising. Module II performs an unsupervised classification by K-means or hierarchical clustering. Module III evaluates and identifies biological entities (e.g., molecular events) that exhibit strong associations to specific temporal patterns. The jackstraw method for cluster membership has been applied to estimate p-values and posterior inclusion probabilities (PIPs), both of which guided feature selection. To demonstrate the utility of the analysis platform, we employed a temporal proteomics dataset that captured the proteome-wide dynamics of oxidative stress induced post-translational modifications (O-PTMs) in mouse hearts undergoing isoproterenol (ISO)-induced hypertrophy. CONCLUSION We have created a platform, CV.Signature.TCP, to identify distinct temporal clusters in omics datasets. We presented a cardiovascular use case to demonstrate its utility in unveiling biological insights underlying O-PTM regulations in cardiac remodeling. This platform is implemented in an open source R package (https://github.com/UCLA-BD2K/CV.Signature.TCP).
Collapse
Affiliation(s)
- Neo Christopher Chung
- NHLBI Integrated Cardiovascular Data Science Training Program at University of California (UCLA), Los Angeles, USA; Departments of Physiology and Medicine (Cardiology) at UCLA School of Medicine, USA; Institute of Informatics, Faculty of Mathematics, Informatics and Mechanics University of Warsaw, Warsaw, Poland.
| | - Howard Choi
- NHLBI Integrated Cardiovascular Data Science Training Program at University of California (UCLA), Los Angeles, USA; Departments of Physiology and Medicine (Cardiology) at UCLA School of Medicine, USA; Bioinformatics and Medical Informatics at UCLA School of Engineering, Los Angeles, CA 90095, USA; Scalable Analytics Institute (ScAi) at UCLA School of Engineering, Los Angeles, CA 90095, USA
| | - Ding Wang
- Departments of Physiology and Medicine (Cardiology) at UCLA School of Medicine, USA
| | - Bilal Mirza
- Departments of Physiology and Medicine (Cardiology) at UCLA School of Medicine, USA
| | - Alexander R Pelletier
- NHLBI Integrated Cardiovascular Data Science Training Program at University of California (UCLA), Los Angeles, USA; Bioinformatics and Medical Informatics at UCLA School of Engineering, Los Angeles, CA 90095, USA; Scalable Analytics Institute (ScAi) at UCLA School of Engineering, Los Angeles, CA 90095, USA
| | - Dibakar Sigdel
- NHLBI Integrated Cardiovascular Data Science Training Program at University of California (UCLA), Los Angeles, USA; Departments of Physiology and Medicine (Cardiology) at UCLA School of Medicine, USA
| | - Wei Wang
- NHLBI Integrated Cardiovascular Data Science Training Program at University of California (UCLA), Los Angeles, USA; Bioinformatics and Medical Informatics at UCLA School of Engineering, Los Angeles, CA 90095, USA; Scalable Analytics Institute (ScAi) at UCLA School of Engineering, Los Angeles, CA 90095, USA
| | - Peipei Ping
- NHLBI Integrated Cardiovascular Data Science Training Program at University of California (UCLA), Los Angeles, USA; Departments of Physiology and Medicine (Cardiology) at UCLA School of Medicine, USA; Bioinformatics and Medical Informatics at UCLA School of Engineering, Los Angeles, CA 90095, USA; Scalable Analytics Institute (ScAi) at UCLA School of Engineering, Los Angeles, CA 90095, USA.
| |
Collapse
|