1
|
Liu F, Ding Y, Xu Z, Hao X, Pan T, Miles G, Wang S, Wu YH, Liu J, Bado IL, Zhang W, Wu L, Gao Y, Yu L, Edwards DG, Chan HL, Aguirre S, Dieffenbach MW, Chen E, Shen Y, Hoffman D, Becerra Dominguez L, Rivas CH, Chen X, Wang H, Gugala Z, Satcher RL, Zhang XHF. Single-cell profiling of bone metastasis ecosystems from multiple cancer types reveals convergent and divergent mechanisms of bone colonization. CELL GENOMICS 2025:100888. [PMID: 40412393 DOI: 10.1016/j.xgen.2025.100888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 02/26/2025] [Accepted: 04/29/2025] [Indexed: 05/27/2025]
Abstract
Bone is a common site for metastasis of solid cancers. The diversity of histological and molecular characteristics of bone metastases (BMs) remains poorly studied. Here, we performed single-cell RNA sequencing on 42 BMs from eight cancer types, identifying three distinct ecosystem archetypes, each characterized by an enrichment of specific immune cells: macrophages/osteoclasts, regulatory/exhausted T cells, or monocytes. We validated these archetypes by immunostaining on tissue sections and bioinformatic analysis of bulk RNA sequencing/microarray data from 158 BMs across more than 10 cancer types. Interestingly, we found only a modest correlation between the BM archetypes and the tissues of origin; BMs from the same cancer type often fell into different archetypes, while BMs from different cancer types sometimes converged on the same archetype. Additional analyses revealed parallel immunosuppression and bone remodeling mechanisms, some of which were experimentally validated. Overall, we discovered unappreciated heterogeneity of BMs across different cancers.
Collapse
Affiliation(s)
- Fengshuo Liu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Graduate Program in Cancer and Cell Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yunfeng Ding
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Zhan Xu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Xiaoxin Hao
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Tianhong Pan
- Department of Orthopedic Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - George Miles
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Siyue Wang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Graduate Program in Immunology and Microbiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yi-Hsuan Wu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Graduate Program in Cancer and Cell Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Jun Liu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Igor L Bado
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Weijie Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Ling Wu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Yang Gao
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Liqun Yu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - David G Edwards
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Hilda L Chan
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Medical Scientist Training Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sergio Aguirre
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Graduate Program in Cancer and Cell Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Michael Warren Dieffenbach
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Graduate Program in Development, Disease Models, and Therapeutics, Baylor College of Medicine, One Baylor Plaza, Houston TX 77030, USA
| | - Elina Chen
- College of Natural Sciences, University of Texas at Austin, 110 Inner Campus Drive, Austin, TX 78706, USA
| | - Yichao Shen
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Graduate Program in Cancer and Cell Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Graduate Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Dane Hoffman
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Graduate Program in Cancer and Cell Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Luis Becerra Dominguez
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Graduate Program in Immunology and Microbiology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Charlotte Helena Rivas
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Graduate Program in Cancer and Cell Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Xiang Chen
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Hai Wang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Zbigniew Gugala
- Department of Orthopedic Surgery and Rehabilitation, University of Texas Medical Branch, Galveston, TX, USA
| | - Robert L Satcher
- Department of Orthopedic Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
| | - Xiang H-F Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; McNair Medical Institute, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA.
| |
Collapse
|
2
|
Mihalas AB, Arora S, O'Connor SA, Feldman HM, Cucinotta CE, Mitchell K, Bassett J, Kim D, Jin K, Hoellerbauer P, Delegard J, Ling M, Jenkins W, Kufeld M, Corrin P, Carter L, Tsukiyama T, Aronow B, Plaisier CL, Patel AP, Paddison PJ. KAT5 regulates neurodevelopmental states associated with G0-like populations in glioblastoma. Nat Commun 2025; 16:4327. [PMID: 40346033 PMCID: PMC12064679 DOI: 10.1038/s41467-025-59503-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 04/22/2025] [Indexed: 05/11/2025] Open
Abstract
Quiescence cancer stem-like cells may play key roles in promoting tumor cell heterogeneity and recurrence for many tumors, including glioblastoma (GBM). Here we show that the protein acetyltransferase KAT5 is a key regulator of transcriptional, epigenetic, and proliferative heterogeneity impacting transitions into G0-like states in GBM. KAT5 activity suppresses the emergence of quiescent subpopulations with neurodevelopmental progenitor characteristics, while promoting GBM stem-like cell (GSC) self-renewal through coordinately regulating E2F- and MYC- transcriptional networks with protein translation. KAT5 inactivation significantly decreases tumor progression and invasive behavior while increasing survival after standard of care. Further, increasing MYC expression in human neural stem cells stimulates KAT5 activity and protein translation, as well as confers sensitivity to homoharringtonine, to similar levels to those found in GSCs and high-grade gliomas. These results suggest that the dynamic behavior of KAT5 plays key roles in G0 ingress/egress, adoption of quasi-neurodevelopmental states, and aggressive tumor growth in gliomas.
Collapse
Affiliation(s)
- Anca B Mihalas
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Sonali Arora
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Samantha A O'Connor
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85281, USA
| | - Heather M Feldman
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Christine E Cucinotta
- College of Arts and Sciences, Department of Molecular Genetics, Ohio State University, Columbus, OH, 43210, USA
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Kelly Mitchell
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - John Bassett
- Department of Medicine, Karolinska Institute, Huddinge, Sweden
| | - Dayoung Kim
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Kang Jin
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Pia Hoellerbauer
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Jennifer Delegard
- Department of Neurosurgery, University of Washington, Seattle, WA, 98195, USA
| | - Melissa Ling
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA, 98195, USA
| | - Wesley Jenkins
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA, 98195, USA
| | - Megan Kufeld
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Philip Corrin
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Lucas Carter
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Toshio Tsukiyama
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Bruce Aronow
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Christopher L Plaisier
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85281, USA
| | - Anoop P Patel
- Department of Neurosurgery, Duke University, Durham, NC, 27710, USA.
- Preston Robert Tisch Brain Tumor Center, Duke University, Durham, NC, 27710, USA.
- Center for Advanced Genomic Technologies, Duke University, Durham, NC, 27710, USA.
| | - Patrick J Paddison
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA.
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
3
|
Jogani S, Pol AS, Prajapati M, Samal A, Bhatia K, Parmar J, Patel U, Shah F, Vyas N, Gupta S. scaLR: a low-resource deep neural network-based platform for single cell analysis and biomarker discovery. Brief Bioinform 2025; 26:bbaf243. [PMID: 40439670 PMCID: PMC12121358 DOI: 10.1093/bib/bbaf243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 04/14/2025] [Accepted: 05/02/2025] [Indexed: 06/02/2025] Open
Abstract
Single-cell ribonucleic acid (RNA) sequencing (scRNA-seq) produces vast amounts of individual cell profiling data. Its analysis presents a significant challenge in accurately annotating cell types and their associated biomarkers. Different pipelines based on deep neural network (DNN) methods have been employed to tackle these issues. These pipelines have arisen as a promising resource and can extract meaningful and concise features from noisy, diverse, and high-dimensional data to enhance annotations and subsequent analysis. Existing tools require high computational resources to execute large sample datasets. We have developed a cutting-edge platform known as scaLR (Single-cell analysis using low resource) that efficiently processes data into feature subsets, samples in batches to reduce the required memory for processing large datasets, and running DNN models in multiple central processing units. scaLR is equipped with data processing, feature extraction, training, evaluation, and downstream analysis. Its novel feature extraction algorithm first trains the model on a feature subset and stores the importance of the features for all the features in that subset. At the end of the training of all subsets, the top-K features are selected based on their importance. The final model is trained on top-K features; its performance evaluation and associated downstream analysis provide significant biomarkers for different cell types and diseases/traits. Our findings indicate that scaLR offers comparable prediction accuracy and requires less model training time and computational resources than existing Python-based pipelines. We present scaLR, a Python-based platform, engineered to utilize minimal computational resources while maintaining comparable execution times and analysis costs to existing frameworks.
Collapse
Affiliation(s)
- Saiyam Jogani
- Department of Generative AI & Bioinformatics, Infocusp Innovations, Laxman Nagar Baner, Pune 411045, Maharashtra, India
| | - Anand Santosh Pol
- Department of Generative AI & Bioinformatics, Infocusp Innovations, Laxman Nagar Baner, Pune 411045, Maharashtra, India
| | - Mayur Prajapati
- Department of Generative AI & Bioinformatics, Infocusp Innovations, Gala-hub, Bopal, Ahmedabad 380058, Gujarat, India
| | - Amit Samal
- Department of Generative AI & Bioinformatics, Infocusp Innovations, Gala-hub, Bopal, Ahmedabad 380058, Gujarat, India
| | - Kriti Bhatia
- Department of Generative AI & Bioinformatics, Infocusp Innovations, Laxman Nagar Baner, Pune 411045, Maharashtra, India
| | - Jayendra Parmar
- Department of Generative AI & Bioinformatics, Infocusp Innovations, Gala-hub, Bopal, Ahmedabad 380058, Gujarat, India
| | - Urvik Patel
- Department of Generative AI & Bioinformatics, Infocusp Innovations, Gala-hub, Bopal, Ahmedabad 380058, Gujarat, India
| | - Falak Shah
- Department of Generative AI & Bioinformatics, Infocusp Innovations, Gala-hub, Bopal, Ahmedabad 380058, Gujarat, India
| | - Nisarg Vyas
- Department of Generative AI & Bioinformatics, Infocusp Innovations, Gala-hub, Bopal, Ahmedabad 380058, Gujarat, India
| | - Saurabh Gupta
- Department of Generative AI & Bioinformatics, Infocusp Innovations, Gala-hub, Bopal, Ahmedabad 380058, Gujarat, India
| |
Collapse
|
4
|
CZI Cell Science Program, Abdulla S, Aevermann B, Assis P, Badajoz S, Bell SM, Bezzi E, Cakir B, Chaffer J, Chambers S, Cherry J, Chi T, Chien J, Dorman L, Garcia-Nieto P, Gloria N, Hastie M, Hegeman D, Hilton J, Huang T, Infeld A, Istrate AM, Jelic I, Katsuya K, Kim YJ, Liang K, Lin M, Lombardo M, Marshall B, Martin B, McDade F, Megill C, Patel N, Predeus A, Raymor B, Robatmili B, Rogers D, Rutherford E, Sadgat D, Shin A, Small C, Smith T, Sridharan P, Tarashansky A, Tavares N, Thomas H, Tolopko A, Urisko M, Yan J, Yeretssian G, Zamanian J, Mani A, Cool J, Carr A. CZ CELLxGENE Discover: a single-cell data platform for scalable exploration, analysis and modeling of aggregated data. Nucleic Acids Res 2025; 53:D886-D900. [PMID: 39607691 PMCID: PMC11701654 DOI: 10.1093/nar/gkae1142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 10/28/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024] Open
Abstract
Hundreds of millions of single cells have been analyzed using high-throughput transcriptomic methods. The cumulative knowledge within these datasets provides an exciting opportunity for unlocking insights into health and disease at the level of single cells. Meta-analyses that span diverse datasets building on recent advances in large language models and other machine-learning approaches pose exciting new directions to model and extract insight from single-cell data. Despite the promise of these and emerging analytical tools for analyzing large amounts of data, the sheer number of datasets, data models and accessibility remains a challenge. Here, we present CZ CELLxGENE Discover (cellxgene.cziscience.com), a data platform that provides curated and interoperable single-cell data. Available via a free-to-use online data portal, CZ CELLxGENE hosts a growing corpus of community-contributed data of over 93 million unique cells. Curated, standardized and associated with consistent cell-level metadata, this collection of single-cell transcriptomic data is the largest of its kind and growing rapidly via community contributions. A suite of tools and features enables accessibility and reusability of the data via both computational and visual interfaces to allow researchers to explore individual datasets, perform cross-corpus analysis, and run meta-analyses of tens of millions of cells across studies and tissues at the resolution of single cells.
Collapse
Affiliation(s)
| | - Shibla Abdulla
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, UK
| | - Brian Aevermann
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Pedro Assis
- Department of Genetics, Stanford University School of Medicine, 291 Campus Drive, Li Ka Shing Building, Stanford, CA 94305, USA
| | - Seve Badajoz
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Sidney M Bell
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Emanuele Bezzi
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Batuhan Cakir
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, UK
| | - Jim Chaffer
- Department of Genetics, Stanford University School of Medicine, 291 Campus Drive, Li Ka Shing Building, Stanford, CA 94305, USA
| | - Signe Chambers
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - J Michael Cherry
- Department of Genetics, Stanford University School of Medicine, 291 Campus Drive, Li Ka Shing Building, Stanford, CA 94305, USA
| | - Tiffany Chi
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Jennifer Chien
- Department of Genetics, Stanford University School of Medicine, 291 Campus Drive, Li Ka Shing Building, Stanford, CA 94305, USA
| | - Leah Dorman
- Chan Zuckerberg, Biohub, SF, 499 Illinois St, San Francisco, CA 94158, USA
| | - Pablo Garcia-Nieto
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Nayib Gloria
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Mim Hastie
- Clever Canary, 850 Front St. #1491, Santa Cruz, CA, USA
| | - Daniel Hegeman
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Jason Hilton
- Department of Genetics, Stanford University School of Medicine, 291 Campus Drive, Li Ka Shing Building, Stanford, CA 94305, USA
| | - Timmy Huang
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Amanda Infeld
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Ana-Maria Istrate
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Ivana Jelic
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Kuni Katsuya
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Yang Joon Kim
- Chan Zuckerberg, Biohub, SF, 499 Illinois St, San Francisco, CA 94158, USA
| | - Karen Liang
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Mike Lin
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | | | - Bailey Marshall
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Bruce Martin
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Fran McDade
- Clever Canary, 850 Front St. #1491, Santa Cruz, CA, USA
| | - Colin Megill
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Nikhil Patel
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Alexander Predeus
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, UK
| | - Brian Raymor
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Behnam Robatmili
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Dave Rogers
- Clever Canary, 850 Front St. #1491, Santa Cruz, CA, USA
| | - Erica Rutherford
- Department of Genetics, Stanford University School of Medicine, 291 Campus Drive, Li Ka Shing Building, Stanford, CA 94305, USA
| | - Dana Sadgat
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Andrew Shin
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Corinn Small
- Department of Genetics, Stanford University School of Medicine, 291 Campus Drive, Li Ka Shing Building, Stanford, CA 94305, USA
| | - Trent Smith
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Prathap Sridharan
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | | | - Norbert Tavares
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Harley Thomas
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Andrew Tolopko
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Meghan Urisko
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Joyce Yan
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Garabet Yeretssian
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Jennifer Zamanian
- Department of Genetics, Stanford University School of Medicine, 291 Campus Drive, Li Ka Shing Building, Stanford, CA 94305, USA
| | - Arathi Mani
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Jonah Cool
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| | - Ambrose Carr
- Chan Zuckerberg Initiative, 1180 Main Street, Redwood City, CA 94063, USA
| |
Collapse
|
5
|
Yin D, Cao Y, Chen J, Mak CLY, Yu KHO, Zhang J, Li J, Lin Y, Ho JWK, Yang JYH. Scope+: an open source generalizable architecture for single-cell RNA-seq atlases at sample and cell levels. Bioinformatics 2024; 41:btae727. [PMID: 39705183 PMCID: PMC11755096 DOI: 10.1093/bioinformatics/btae727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 11/18/2024] [Accepted: 12/13/2024] [Indexed: 12/22/2024] Open
Abstract
SUMMARY With the recent advancement in single-cell RNA-sequencing technologies and the increased availability of integrative tools, challenges arise in easy and fast access to large collections of cell atlas. Existing cell atlas portals rarely are open sourced and adaptable, and do not support meta-analysis at cell level. Here, we present an open source, highly optimized and scalable architecture, named Scope+, to allow quick access, meta-analysis and cell-level selection of the atlas data. We applied this architecture to our well-curated 5 million COVID-19 blood and immune cells, as a portal called Covidscope. We achieved efficient access to atlas-scale data via three strategies, such as cell-as-unit data modelling, novel database optimization techniques and innovative software architectural design. Scope+ serves as an open source architecture for researchers to build on with their own atlas. AVAILABILITY AND IMPLEMENTATION The COVID-19 web portal, data and meta-analysis are available on Covidscope (https://covidsc.d24h.hk/). User tutorials on how to implement Scope+ architecture with their atlases can be found at https://hiyin.github.io/scopeplus-user-tutorial/. Scope+ source code can be found at https://doi.org/10.5281/zenodo.14174632 and https://github.com/hiyin/scopeplus.
Collapse
Affiliation(s)
- Danqing Yin
- Laboratory of Data Discovery for Health Limited (D24H), Pak Shek Kok, Hong Kong SAR, 999077, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, 999077, China
| | - Yue Cao
- Laboratory of Data Discovery for Health Limited (D24H), Pak Shek Kok, Hong Kong SAR, 999077, China
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, 2006, Australia
- School of Mathematics and Statistics, University of Sydney, Camperdown, NSW, 2006, Australia
- Sydney Precision Data Science Centre, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Junyi Chen
- Laboratory of Data Discovery for Health Limited (D24H), Pak Shek Kok, Hong Kong SAR, 999077, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, 999077, China
| | - Candice L Y Mak
- Laboratory of Data Discovery for Health Limited (D24H), Pak Shek Kok, Hong Kong SAR, 999077, China
| | - Ken H O Yu
- Laboratory of Data Discovery for Health Limited (D24H), Pak Shek Kok, Hong Kong SAR, 999077, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, 999077, China
| | - Jiaxuan Zhang
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong Province 510005, China
| | - Jia Li
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong Province 510005, China
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province, 510005, China
| | - Yingxin Lin
- Laboratory of Data Discovery for Health Limited (D24H), Pak Shek Kok, Hong Kong SAR, 999077, China
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, 2006, Australia
- School of Mathematics and Statistics, University of Sydney, Camperdown, NSW, 2006, Australia
- Sydney Precision Data Science Centre, University of Sydney, Camperdown, NSW, 2006, Australia
| | - Joshua W K Ho
- Laboratory of Data Discovery for Health Limited (D24H), Pak Shek Kok, Hong Kong SAR, 999077, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, 999077, China
| | - Jean Y H Yang
- Laboratory of Data Discovery for Health Limited (D24H), Pak Shek Kok, Hong Kong SAR, 999077, China
- Charles Perkins Centre, University of Sydney, Camperdown, NSW, 2006, Australia
- School of Mathematics and Statistics, University of Sydney, Camperdown, NSW, 2006, Australia
- Sydney Precision Data Science Centre, University of Sydney, Camperdown, NSW, 2006, Australia
| |
Collapse
|
6
|
Ma Y, Ji J, Liu X, Zheng X, Xu L, Zhou Q, Li Z, Yang L. Integrative Analysis by Mendelian Randomization and Large-Scale Single-Cell Transcriptomics Reveals Causal Links between B Cell Subtypes and Diabetic Kidney Disease. KIDNEY DISEASES (BASEL, SWITZERLAND) 2024; 10:327-345. [PMID: 39430286 PMCID: PMC11488840 DOI: 10.1159/000539689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 06/03/2024] [Indexed: 10/22/2024]
Abstract
Introduction The increasing incidence of diabetic kidney disease (DKD) and the challenges in its management highlight the necessity for a deeper understanding of its pathogenesis. While recent studies have underscored the substantial impact of circulating immunity on the development of diabetic microvascular complications such as retinopathy and neuropathy, research on circulating immunity in DKD remains limited. Methods This study utilized Mendelian randomization analysis to explore the potential independent causal relationships between circulating immune cells and DKD pathogenesis. Additionally, a combination of single-cell disease relevance score (scDRS) and immune cell infiltration analysis was employed to map the circulating immunity landscape in DKD patients. Results Ten immune traits, including 5 of B cells, 2 of T cells, 2 of granulocytes, and one of monocytes, were defined to be associated with the pathogenesis of DKD. Notably, IgD - CD27 - B cell Absolute Count (IVW: OR, 1.102 [1.023-1.189], p = 0.011) and IgD - CD24 - B cell Absolute Count (IVW: OR, 1.106 [1.030-1.188], p = 0.005) were associated with promoting DKD pathogenesis, while CD24 + CD27 + B cell %B cell (IVW: OR, 0.943 [0.898-0.989], p = 0.016) demonstrated a protective effect against DKD onset. The presence of B cell-activating factor receptor (BAFF-R) on CD20 - CD38 - B cell (IVW: OR, 0.946 [0.904-0.989], p = 0.015) and BAFF-R on IgD - CD38 + B cell (IVW: OR, 0.902 [0.834-0.975], p = 0.009) also indicated a potential role in preventing DKD. scDRS analysis revealed that two main subsets of B cells, naïve B and memory B cells, had a higher proportion of DKD-related cells or a higher scDRS score of DKD phenotype, indicating their strong association with DKD. Furthermore, immune infiltrate deconvolution analysis showed a notable decrease in the circulating memory B cells and class-switched memory B cells in DKD patients compared to those of DM patients without DKD. Conclusion Our study revealed the causal relations between circulating immunity and DKD susceptibility, particularly highlighted the potential roles of B cell subtypes in DKD development. Further studies addressing the related mechanisms would broaden the current understanding of DKD pathogenesis.
Collapse
Affiliation(s)
- Yuan Ma
- Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Jing Ji
- Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Department of Nephrology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xintong Liu
- Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Xizi Zheng
- Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Lingyi Xu
- Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Qingqing Zhou
- Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Zehua Li
- Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Li Yang
- Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
7
|
Karp JM, Modrek AS, Ezhilarasan R, Zhang ZY, Ding Y, Graciani M, Sahimi A, Silvestro M, Chen T, Li S, Wong KK, Ramkhelawon B, Bhat KP, Sulman EP. Deconvolution of the tumor-educated platelet transcriptome reveals activated platelet and inflammatory cell transcript signatures. JCI Insight 2024; 9:e178719. [PMID: 39190500 PMCID: PMC11466191 DOI: 10.1172/jci.insight.178719] [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: 12/21/2023] [Accepted: 08/13/2024] [Indexed: 08/29/2024] Open
Abstract
Tumor-educated platelets (TEPs) are a potential method of liquid biopsy for the diagnosis and monitoring of cancer. However, the mechanism underlying tumor education of platelets is not known, and transcripts associated with TEPs are often not tumor-associated transcripts. We demonstrated that direct tumor transfer of transcripts to circulating platelets is an unlikely source of the TEP signal. We used CDSeq, a latent Dirichlet allocation algorithm, to deconvolute the TEP signal in blood samples from patients with glioblastoma. We demonstrated that a substantial proportion of transcripts in the platelet transcriptome are derived from nonplatelet cells, and the use of this algorithm allows the removal of contaminant transcripts. Furthermore, we used the results of this algorithm to demonstrate that TEPs represent a subset of more activated platelets, which also contain transcripts normally associated with nonplatelet inflammatory cells, suggesting that these inflammatory cells, possibly in the tumor microenvironment, transfer transcripts to platelets that are then found in circulation. Our analysis suggests a useful and efficient method of processing TEP transcriptomic data to enable the isolation of a unique TEP signal associated with specific tumors.
Collapse
Affiliation(s)
- Jerome M. Karp
- Department of Radiation Oncology, NYU Grossman School of Medicine, New York, New York, USA
| | - Aram S. Modrek
- Department of Radiation Oncology, Keck School of Medicine of University of Southern California, Los Angeles, California, USA
| | - Ravesanker Ezhilarasan
- Department of Radiation Oncology, NYU Grossman School of Medicine, New York, New York, USA
| | - Ze-Yan Zhang
- Department of Radiation Oncology, NYU Grossman School of Medicine, New York, New York, USA
| | - Yingwen Ding
- Department of Radiation Oncology, NYU Grossman School of Medicine, New York, New York, USA
| | - Melanie Graciani
- Department of Radiation Oncology, NYU Grossman School of Medicine, New York, New York, USA
| | - Ali Sahimi
- Department of Radiation Oncology, NYU Grossman School of Medicine, New York, New York, USA
| | | | - Ting Chen
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, New York, USA
| | - Shuai Li
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, New York, USA
| | - Kwok-Kin Wong
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, New York, USA
| | | | | | - Erik P. Sulman
- Department of Radiation Oncology, NYU Grossman School of Medicine, New York, New York, USA
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
8
|
Mangiola S, Milton M, Ranathunga N, Li-Wai-Suen C, Odainic A, Yang E, Hutchison W, Garnham A, Iskander J, Pal B, Yadav V, Rossello J, Carey VJ, Morgan M, Bedoui S, Kallies A, Papenfuss AT. A multi-organ map of the human immune system across age, sex and ethnicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.08.542671. [PMID: 38746418 PMCID: PMC11092463 DOI: 10.1101/2023.06.08.542671] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Understanding tissue biology's heterogeneity is crucial for advancing precision medicine. Despite the centrality of the immune system in tissue homeostasis, a detailed and comprehensive map of immune cell distribution and interactions across human tissues and demographics remains elusive. To fill this gap, we harmonised data from 12,981 single-cell RNA sequencing samples and curated 29 million cells from 45 anatomical sites to create a comprehensive compositional and transcriptional healthy map of the healthy immune system. We used this resource and a novel multilevel modelling approach to track immune ageing and test differences across sex and ethnicity. We uncovered conserved and tissue-specific immune-ageing programs, resolved sex-dependent differential ageing and identified ethnic diversity in clinically critical immune checkpoints. This study provides a quantitative baseline of the immune system, facilitating advances in precision medicine. By sharing our immune map, we hope to catalyse further breakthroughs in cancer, infectious disease, immunology and precision medicine.
Collapse
Affiliation(s)
- S Mangiola
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - M Milton
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - N Ranathunga
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Csn Li-Wai-Suen
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - A Odainic
- The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127 Bonn, Germany
| | - E Yang
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - W Hutchison
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - A Garnham
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - J Iskander
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - B Pal
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia
| | - V Yadav
- Systems Biology of Aging Laboratory, Columbia University; New York, USA
| | - Jfj Rossello
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC 3052, Australia
- Novo Nordisk Foundation Center for Stem Cell Medicine, Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Victoria, Australia
| | - V J Carey
- Channing Division of Network Medicine, Mass General Brigham, Harvard Medical School, Harvard University, Boston, USA
| | - M Morgan
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, NY, USA
| | - S Bedoui
- The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - A Kallies
- The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - A T Papenfuss
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
9
|
Yang S, Gaietto K, Chen W. Mapping a New Course to Understand Lung Biology Mechanisms: LungMAP.net. Am J Respir Cell Mol Biol 2024; 70:91-93. [PMID: 38109690 PMCID: PMC10848696 DOI: 10.1165/rcmb.2023-0439ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 12/18/2023] [Indexed: 12/20/2023] Open
Affiliation(s)
- Sheng Yang
- Department of Biostatistics Nanjing Medical University Nanjing, Jiangsu, China
| | - Kristina Gaietto
- Department of Pediatrics University of Pittsburgh School of Medicine Pittsburgh, Pennsylvania
| | - Wei Chen
- Department of Pediatrics University of Pittsburgh School of Medicine Pittsburgh, Pennsylvania
| |
Collapse
|
10
|
Chu CY, Kim SY, Pryhuber GS, Mariani TJ, McGraw MD. Single-cell resolution of human airway epithelial cells exposed to bronchiolitis obliterans-associated chemicals. Am J Physiol Lung Cell Mol Physiol 2024; 326:L135-L148. [PMID: 38084407 PMCID: PMC11279737 DOI: 10.1152/ajplung.00304.2023] [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: 09/25/2023] [Revised: 10/31/2023] [Accepted: 11/23/2023] [Indexed: 01/24/2024] Open
Abstract
Bronchiolitis obliterans (BO) is a fibrotic lung disease characterized by progressive luminal narrowing and obliteration of the small airways. In the nontransplant population, inhalation exposure to certain chemicals is associated with BO; however, the mechanisms contributing to disease induction remain poorly understood. This study's objective was to use single-cell RNA sequencing for the identification of transcriptomic signatures common to primary human airway epithelial cells after chemical exposure to BO-associated chemicals-diacetyl or nitrogen mustard-to help explain BO induction. Primary airway epithelial cells were cultured at air-liquid interface and exposed to diacetyl, nitrogen mustard, or control vapors. Cultures were dissociated and sequenced for single-cell RNA. Differential gene expression and functional pathway analyses were compared across exposures. In total, 75,663 single cells were captured and sequenced from all exposure conditions. Unbiased clustering identified 11 discrete phenotypes, including 5 basal, 2 ciliated, and 2 secretory cell clusters. With chemical exposure, the proportion of cells assigned to keratin 5+ basal cells decreased, whereas the proportion of cells aligned to secretory cell clusters increased compared with control exposures. Functional pathway analysis identified interferon signaling and antigen processing/presentation as pathways commonly upregulated after diacetyl or nitrogen mustard exposure in a ciliated cell cluster. Conversely, the response of airway basal cells differed significantly with upregulation of the unfolded protein response in diacetyl-exposed basal cells, not seen in nitrogen mustard-exposed cultures. These new insights provide early identification of airway epithelial signatures common to BO-associated chemical exposures.NEW & NOTEWORTHY Bronchiolitis obliterans (BO) is a devastating fibrotic lung disease of the small airways, or bronchioles. This original manuscript uses single-cell RNA sequencing for identifying common signatures of chemically exposed airway epithelial cells in BO induction. Chemical exposure reduced the proportion of keratin 5+ basal cells while increasing the proportion of keratin 4+ suprabasal cells. Functional pathways contributory to these shifts differed significantly across exposures. These new results highlight similarities and differences in BO induction across exposures.
Collapse
Affiliation(s)
- Chin-Yi Chu
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
| | - So-Young Kim
- Division of Pediatric Pulmonology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York, United States
| | - Gloria S Pryhuber
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York, United States
| | - Thomas J Mariani
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York, United States
| | - Matthew D McGraw
- Division of Pediatric Pulmonology, Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, United States
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York, United States
| |
Collapse
|
11
|
Prebensen C, Lefol Y, Myhre PL, Lüders T, Jonassen C, Blomfeldt A, Omland T, Nilsen H, Berdal JE. Longitudinal whole blood transcriptomic analysis characterizes neutrophil activation and interferon signaling in moderate and severe COVID-19. Sci Rep 2023; 13:10368. [PMID: 37365222 PMCID: PMC10293211 DOI: 10.1038/s41598-023-37606-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 06/24/2023] [Indexed: 06/28/2023] Open
Abstract
A maladaptive inflammatory response has been implicated in the pathogenesis of severe COVID-19. This study aimed to characterize the temporal dynamics of this response and investigate whether severe disease is associated with distinct gene expression patterns. We performed microarray analysis of serial whole blood RNA samples from 17 patients with severe COVID-19, 15 patients with moderate disease and 11 healthy controls. All study subjects were unvaccinated. We assessed whole blood gene expression patterns by differential gene expression analysis, gene set enrichment, two clustering methods and estimated relative leukocyte abundance using CIBERSORT. Neutrophils, platelets, cytokine signaling, and the coagulation system were activated in COVID-19, and this broad immune activation was more pronounced in severe vs. moderate disease. We observed two different trajectories of neutrophil-associated genes, indicating the emergence of a more immature neutrophil phenotype over time. Interferon-associated genes were strongly enriched in early COVID-19 before falling markedly, with modest severity-associated differences in trajectory. In conclusion, COVID-19 necessitating hospitalization is associated with a broad inflammatory response, which is more pronounced in severe disease. Our data suggest a progressively more immature circulating neutrophil phenotype over time. Interferon signaling is enriched in COVID-19 but does not seem to drive severe disease.
Collapse
Affiliation(s)
- Christian Prebensen
- Department of Infectious Diseases, Oslo University Hospital, Kirkeveien 166, 0450, Oslo, Norway.
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Yohan Lefol
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Microbiology, University of Oslo, Oslo, Norway
| | - Peder L Myhre
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Akershus University Hospital, Lørenskog, Norway
| | - Torben Lüders
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Clinical Molecular Biology, Akershus University Hospital, Lørenskog, Norway
| | | | - Anita Blomfeldt
- Department of Microbiology and Infection Control, Akershus University Hospital, Lørenskog, Norway
| | - Torbjørn Omland
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Akershus University Hospital, Lørenskog, Norway
| | - Hilde Nilsen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Microbiology, University of Oslo, Oslo, Norway
| | - Jan-Erik Berdal
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Infectious Diseases, Akershus University Hospital, Lørenskog, Norway
| |
Collapse
|
12
|
Sebastian R, Jin K, Pavon N, Bansal R, Potter A, Song Y, Babu J, Gabriel R, Sun Y, Aronow B, Pak C. Schizophrenia-associated NRXN1 deletions induce developmental-timing- and cell-type-specific vulnerabilities in human brain organoids. Nat Commun 2023; 14:3770. [PMID: 37355690 PMCID: PMC10290702 DOI: 10.1038/s41467-023-39420-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 06/13/2023] [Indexed: 06/26/2023] Open
Abstract
De novo mutations and copy number deletions in NRXN1 (2p16.3) pose a significant risk for schizophrenia (SCZ). It is unclear how NRXN1 deletions impact cortical development in a cell type-specific manner and disease background modulates these phenotypes. Here, we leveraged human pluripotent stem cell-derived forebrain organoid models carrying NRXN1 heterozygous deletions in isogenic and SCZ patient genetic backgrounds and conducted single-cell transcriptomic analysis over the course of brain organoid development from 3 weeks to 3.5 months. Intriguingly, while both deletions similarly impacted molecular pathways associated with ubiquitin-proteasome system, alternative splicing, and synaptic signaling in maturing glutamatergic and GABAergic neurons, SCZ-NRXN1 deletions specifically perturbed developmental trajectories of early neural progenitors and accumulated disease-specific transcriptomic signatures. Using calcium imaging, we found that both deletions led to long-lasting changes in spontaneous and synchronous neuronal networks, implicating synaptic dysfunction. Our study reveals developmental-timing- and cell-type-dependent actions of NRXN1 deletions in unique genetic contexts.
Collapse
Affiliation(s)
- Rebecca Sebastian
- Graduate Program in Neuroscience & Behavior, UMass Amherst, Amherst, MA, 01003, USA
- Department of Biochemistry and Molecular Biology, UMass Amherst, Amherst, MA, 01003, USA
| | - Kang Jin
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Biomedical Informatics, University of Cincinnati, Cincinnati, OH, 45229, USA
| | - Narciso Pavon
- Department of Biochemistry and Molecular Biology, UMass Amherst, Amherst, MA, 01003, USA
| | - Ruby Bansal
- Department of Biochemistry and Molecular Biology, UMass Amherst, Amherst, MA, 01003, USA
| | - Andrew Potter
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Yoonjae Song
- Department of Biochemistry and Molecular Biology, UMass Amherst, Amherst, MA, 01003, USA
| | - Juliana Babu
- Department of Biochemistry and Molecular Biology, UMass Amherst, Amherst, MA, 01003, USA
| | - Rafael Gabriel
- Department of Biochemistry and Molecular Biology, UMass Amherst, Amherst, MA, 01003, USA
| | - Yubing Sun
- Department of Mechanical and Industrial Engineering, UMass Amherst, Amherst, MA, 01003, USA
| | - Bruce Aronow
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Biomedical Informatics, University of Cincinnati, Cincinnati, OH, 45229, USA
- Department of Electrical Engineering and Computer Science, University of Cincinnati, Cincinnati, OH, 45221, USA
- Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, 45256, USA
| | - ChangHui Pak
- Department of Biochemistry and Molecular Biology, UMass Amherst, Amherst, MA, 01003, USA.
| |
Collapse
|
13
|
Arriaga-Canon C, Contreras-Espinosa L, Rebollar-Vega R, Montiel-Manríquez R, Cedro-Tanda A, García-Gordillo JA, Álvarez-Gómez RM, Jiménez-Trejo F, Castro-Hernández C, Herrera LA. Transcriptomics and RNA-Based Therapeutics as Potential Approaches to Manage SARS-CoV-2 Infection. Int J Mol Sci 2022; 23:11058. [PMID: 36232363 PMCID: PMC9570475 DOI: 10.3390/ijms231911058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 11/24/2022] Open
Abstract
SARS-CoV-2 is a coronavirus family member that appeared in China in December 2019 and caused the disease called COVID-19, which was declared a pandemic in 2020 by the World Health Organization. In recent months, great efforts have been made in the field of basic and clinical research to understand the biology and infection processes of SARS-CoV-2. In particular, transcriptome analysis has contributed to generating new knowledge of the viral sequences and intracellular signaling pathways that regulate the infection and pathogenesis of SARS-CoV-2, generating new information about its biology. Furthermore, transcriptomics approaches including spatial transcriptomics, single-cell transcriptomics and direct RNA sequencing have been used for clinical applications in monitoring, detection, diagnosis, and treatment to generate new clinical predictive models for SARS-CoV-2. Consequently, RNA-based therapeutics and their relationship with SARS-CoV-2 have emerged as promising strategies to battle the SARS-CoV-2 pandemic with the assistance of novel approaches such as CRISPR-CAS, ASOs, and siRNA systems. Lastly, we discuss the importance of precision public health in the management of patients infected with SARS-CoV-2 and establish that the fusion of transcriptomics, RNA-based therapeutics, and precision public health will allow a linkage for developing health systems that facilitate the acquisition of relevant clinical strategies for rapid decision making to assist in the management and treatment of the SARS-CoV-2-infected population to combat this global public health problem.
Collapse
Affiliation(s)
- Cristian Arriaga-Canon
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Avenida San Fernando No. 22 ColC. Sección XVI, Tlalpan. C.P., Mexico City 14080, Mexico
| | - Laura Contreras-Espinosa
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Avenida San Fernando No. 22 ColC. Sección XVI, Tlalpan. C.P., Mexico City 14080, Mexico
| | - Rosa Rebollar-Vega
- Genomics Laboratory, Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México, Vasco de Quiroga 15, Belisario Domínguez Secc 16, Tlalpan, Mexico City 14080, Mexico
| | - Rogelio Montiel-Manríquez
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Avenida San Fernando No. 22 ColC. Sección XVI, Tlalpan. C.P., Mexico City 14080, Mexico
| | - Alberto Cedro-Tanda
- Instituto Nacional de Medicina Genómica, Periférico Sur 4809, Arenal Tepepan, Tlalpan. C.P., Mexico City 14610, Mexico
| | - José Antonio García-Gordillo
- Oncología Médica, Instituto Nacional de Cancerología, Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan. C.P., Mexico City 14080, Mexico
| | - Rosa María Álvarez-Gómez
- Clínica de Cáncer Hereditario, Instituto Nacional de Cancerología, Avenida San Fernando No. 22 Col. Sección XVI, Tlalpan. C.P., Mexico City 14080, Mexico
| | - Francisco Jiménez-Trejo
- Instituto Nacional de Pediatría, Insurgentes Sur No. 3700-C, Coyoacán. C.P., Mexico City 04530, Mexico
| | - Clementina Castro-Hernández
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Avenida San Fernando No. 22 ColC. Sección XVI, Tlalpan. C.P., Mexico City 14080, Mexico
| | - Luis A. Herrera
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Avenida San Fernando No. 22 ColC. Sección XVI, Tlalpan. C.P., Mexico City 14080, Mexico
- Instituto Nacional de Medicina Genómica, Periférico Sur 4809, Arenal Tepepan, Tlalpan. C.P., Mexico City 14610, Mexico
| |
Collapse
|
14
|
Zoran MA, Savastru RS, Savastru DM, Tautan MN. Impacts of exposure to air pollution, radon and climate drivers on the COVID-19 pandemic in Bucharest, Romania: A time series study. ENVIRONMENTAL RESEARCH 2022; 212:113437. [PMID: 35594963 PMCID: PMC9113773 DOI: 10.1016/j.envres.2022.113437] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 05/05/2023]
Abstract
During the ongoing global COVID-19 pandemic disease, like several countries, Romania experienced a multiwaves pattern over more than two years. The spreading pattern of SARS-CoV-2 pathogens in the Bucharest, capital of Romania is a multi-factorial process involving among other factors outdoor environmental variables and viral inactivation. Through descriptive statistics and cross-correlation analysis applied to daily time series of observational and geospatial data, this study aims to evaluate the synergy of COVID-19 incidence and lethality with air pollution and radon under different climate conditions, which may exacerbate the coronavirus' effect on human health. During the entire analyzed period 1 January 2020-21 December 2021, for each of the four COVID-19 waves were recorded different anomalous anticyclonic synoptic meteorological patterns in the mid-troposphere, and favorable stability conditions during fall-early winter seasons for COVID-19 disease fast-spreading, mostly during the second, and the fourth waves. As the temporal pattern of airborne SARS-CoV-2 and its mutagen variants is affected by seasonal variability of the main air pollutants and climate parameters, this paper found: 1) the daily outdoor exposures to air pollutants (particulate matter PM2.5 and PM10, nitrogen dioxide-NO2, sulfur dioxide-SO2, carbon monoxide-CO) and radon - 222Rn, are directly correlated with the daily COVID-19 incidence and mortality, and may contribute to the spread and the severity of the pandemic; 2) the daily ground ozone-O3 levels, air temperature, Planetary Boundary Layer height, and surface solar irradiance are anticorrelated with the daily new COVID-19 incidence and deaths, averageingful for spring-summer periods. Outdoor exposure to ambient air pollution associated with radon is a non-negligible driver of COVID-19 transmission in large metropolitan areas, and climate variables are risk factors in spreading the viral infection. The findings of this study provide useful information for public health authorities and decision-makers to develop future pandemic diseases strategies in high polluted metropolitan environments.
Collapse
Affiliation(s)
- Maria A Zoran
- National Institute of R&D for Optoelectronics, Bucharest, Magurele, Romania.
| | - Roxana S Savastru
- National Institute of R&D for Optoelectronics, Bucharest, Magurele, Romania
| | - Dan M Savastru
- National Institute of R&D for Optoelectronics, Bucharest, Magurele, Romania
| | - Marina N Tautan
- National Institute of R&D for Optoelectronics, Bucharest, Magurele, Romania
| |
Collapse
|