1
|
Li A, Lin T, Walker A, Tan X, Zhao R, Yao S, Sullivan PF, Hjerling-Leffler J, Wray NR, Zeng J. Benchmarking methods integrating GWAS and single-cell transcriptomic data for mapping trait-cell type associations. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.05.24.25328275. [PMID: 40475144 PMCID: PMC12140538 DOI: 10.1101/2025.05.24.25328275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/16/2025]
Abstract
Genome-wide association studies (GWAS) have discovered numerous trait-associated variants, but their biological context remains unclear. Integrating GWAS summary statistics with single-cell RNA-sequencing expression profiles can help identify the cell types in which these variants influence traits. Two main strategies have been developed to integrate these data types. The "single cell to GWAS" strategy (representing most methods) identifies gene sets with cell-type-specific expression and then follows with enrichment analyses applied to GWAS summary statistics. Conversely, the "GWAS to single cell" strategy begins with a list of trait-associated genes and calculates a cumulative disease score per cell based on gene expression count data. We systematically evaluated 19 approaches verses "ground truth" trait-cell type pairs to assess their statistical power and false positive rates. Based on these analyses, we draw seven key conclusions to guide future studies. We also propose a Cauchy approach to combine the two main strategies to maximize power for detecting trait-cell type associations.
Collapse
Affiliation(s)
- Ang Li
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Tian Lin
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Alicia Walker
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Xiao Tan
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
- Infection and Inflammation Centre, Queensland Institute of Medical Research Berghofer, Australia
| | - Ruolan Zhao
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Shuyang Yao
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Patrick F. Sullivan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Departments of Genetics and Psychiatry, University of North Carolina, Chapel Hill, NC, USA
| | - Jens Hjerling-Leffler
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Naomi R. Wray
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
- Department of Psychiatry, University of Oxford, Oxford, UK
- These authors contributed equally to this work
| | - Jian Zeng
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
- These authors contributed equally to this work
| |
Collapse
|
2
|
Tsukui T, Sheppard D. Stromal heterogeneity in the adult lung delineated by single-cell genomics. Am J Physiol Cell Physiol 2025; 328:C1964-C1972. [PMID: 40353369 DOI: 10.1152/ajpcell.00285.2025] [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: 03/31/2025] [Revised: 04/11/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
Stromal cells in the lung provide structural support to other cells and play critical roles in inflammation, repair, and fibrosis after injury. Recent technological advancements in single-cell genomics have tremendously improved our knowledge of stromal heterogeneity in the lung. Stromal heterogeneity in single-cell RNA sequencing data is often conserved across different studies despite the different annotation strategies. Spatial analyses suggest that each stromal subset is characterized by unique anatomic locations in the adult lung. This review overviews the stromal heterogeneity delineated by single-cell RNA sequencing studies and highlights the functional characteristics and locations of each population.
Collapse
Affiliation(s)
- Tatsuya Tsukui
- Division of Pulmonary, Critical Care, Allergy and Sleep, Department of Medicine, University of California, San Francisco, California, United States
| | - Dean Sheppard
- Division of Pulmonary, Critical Care, Allergy and Sleep, Department of Medicine, University of California, San Francisco, California, United States
| |
Collapse
|
3
|
El-Husseini ZW, Karp T, Lan A, Gillett TE, Qi C, Khalenkow D, van der Molen T, Brightling C, Papi A, Rabe KF, Siddiqui S, Singh D, Kraft M, Beghé B, Joubert P, Bossé Y, Sin D, Cordero AH, Timens W, Brandsma CA, Hao K, Nickle DC, Vonk JM, Nawijn MC, van den Berge M, Gosens R, Faiz A, Koppelman GH. Improved Annotation of Asthma Gene Variants with Cell Type Deconvolution of Nasal and Lung Expression Quantitative Trait Loci. Am J Respir Cell Mol Biol 2025; 72:607-614. [PMID: 39836087 DOI: 10.1165/rcmb.2024-0251ma] [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: 05/30/2024] [Accepted: 01/21/2025] [Indexed: 01/22/2025] Open
Abstract
Asthma is a genetically complex inflammatory airway disease associated with more than 200 SNPs. However, the functional effects of many asthma-associated SNPs in lung and airway epithelial samples are unknown. Here, we aimed to conduct expression quantitative trait loci (eQTL) analysis using a meta-analysis of nasal and lung samples. We hypothesize that incorporating cell type proportions of airway and lung samples enhances eQTL analysis outcomes. Nasal brush (n = 792) and lung tissue (n = 1,087) samples were investigated separately. Initially, a general eQTL analysis identified genetic variants associated with gene expression levels. Estimated cell type proportions were adjusted based on the Human Lung Cell Atlas. In addition, the presence of significant interaction effects between asthma-associated SNPs and each cell type proportion was explored and considered evidence for cell type-associated eQTL. In nasal brush and lung parenchyma samples, 44 and 116 asthma-associated SNPs were identified as eQTL. Adjusting for cell type proportions revealed eQTL for an additional 17 genes (e.g., FCER1G, CD200R1, and GABBR2) and 16 genes (e.g., CYP2C8, SLC9A2, and SGCD) in nose and lung, respectively. Moreover, we identified eQTL for nine SNPs annotated to genes such as VASP, FOXA3, and PCDHB12 displayed significant interactions with cell type proportions of club, goblet, and alveolar macrophages. Our findings demonstrate increased power for identifying eQTL among asthma-associated SNPs by considering cell type proportion of the bulk RNA-sequencing data from nasal and lung tissues. Integration of cell type deconvolution and eQTL analysis enhances our understanding of asthma genetics and cellular mechanisms, uncovering potential therapeutic targets for personalized interventions.
Collapse
Affiliation(s)
- Zaid W El-Husseini
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Molecular Pharmacology, Groningen Research Institute of Pharmacy, Groningen, the Netherlands
| | - Tatiana Karp
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Department of Pulmonary Diseases
| | - Andy Lan
- Respiratory Bioinformatics and Molecular Biology, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Tessa E Gillett
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Department of Pathology and Medical Biology
| | - Cancan Qi
- Microbiome Medicine Center, Division of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Dmitry Khalenkow
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Laboratory of Genome Structure and Ageing, European Research Institute for the Biology of Ageing, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Chris Brightling
- Department of Infection, Immunity, and Inflammation, Institute for Lung Health, University of Leicester, Leicester, United Kingdom
| | - Alberto Papi
- Department of Respiratory Medicine, University of Ferrara, Ferrara, Italy
| | - Klaus F Rabe
- Department of Medicine, Christian Albrechts University Kiel, Kiel and Lungen Clinic Grosshansdorf, Grosshansdorf, Germany (Members of the German Center for Lung Research [DZL])
| | - Salman Siddiqui
- National Heart and Lung Institute, Imperial College and Imperial NIHR Biomedical Research Centre, London, United Kingdom
| | - Dave Singh
- Medicines Evaluation Unit, Manchester University NHS Foundation Hospital Trust, University of Manchester, Manchester, United Kingdom
| | - Monica Kraft
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Bianca Beghé
- Section of Respiratory Diseases, Department of Oncology, Haematology, and Respiratory Diseases, University of Modena and Reggio Emilia, Policlinico di Modena, Modena, Italy
| | - Philippe Joubert
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Yohan Bossé
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Don Sin
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada; and
| | - Ana H Cordero
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada; and
| | - Wim Timens
- Department of Pathology and Medical Biology
| | - Corry-Anke Brandsma
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Department of Pathology and Medical Biology
| | - Ke Hao
- Merck Research Laboratories, Boston, Massachusetts
| | | | - Judith M Vonk
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Department of Epidemiology, and
| | - Martijn C Nawijn
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Department of Pathology and Medical Biology
| | - Maarten van den Berge
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Department of Pulmonary Diseases
| | - Reinoud Gosens
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Molecular Pharmacology, Groningen Research Institute of Pharmacy, Groningen, the Netherlands
| | - Alen Faiz
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Respiratory Bioinformatics and Molecular Biology, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Gerard H Koppelman
- Groningen Research Institute for Asthma and COPD (GRIAC)
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, and
| |
Collapse
|
4
|
Shirazi SP, Negretti NM, Jetter CS, Sharkey AL, Garg S, Kapp ME, Wilkins D, Fortier G, Mallapragada S, Banovich NE, Eldredge LC, Deutsch GH, Wright CVE, Frank DB, Kropski JA, Sucre JMS. Bronchopulmonary dysplasia with pulmonary hypertension associates with semaphorin signaling loss and functionally decreased FOXF1 expression. Nat Commun 2025; 16:5004. [PMID: 40442177 PMCID: PMC12122835 DOI: 10.1038/s41467-025-60371-7] [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: 09/02/2024] [Accepted: 05/20/2025] [Indexed: 06/02/2025] Open
Abstract
Lung injury in preterm infants leads to structural and functional respiratory deficits, with a risk for bronchopulmonary dysplasia (BPD) that in its most severe form is accompanied by pulmonary hypertension (PH). To identify potential cellular and molecular drivers of BPD in humans, we performed single-cell RNA sequencing of preterm infant lungs with evolving BPD and BPD + PH compared to term infants. Examination of endothelial cells reveals a unique, aberrant capillary cell-state in BPD + PH defined by ANKRD1 expression. Within the alveolar parenchyma in infants with BPD/BPD + PH, predictive signaling analysis identifies surprising deficits in the semaphorin guidance-cue pathway, with decreased expression of pro-angiogenic transcription factor FOXF1. Loss of semaphorin signaling is replicated in a murine BPD model and in humans with causal FOXF1 mutations for alveolar capillary dysplasia (ACDMPV), suggesting a mechanistic link between developmental programs underlying BPD and ACDMPV and uncovering a critical role for semaphorin signaling in normal lung development.
Collapse
Affiliation(s)
- Shawyon P Shirazi
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Nicholas M Negretti
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Christopher S Jetter
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alexandria L Sharkey
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shriya Garg
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Meghan E Kapp
- Department of Pathology, Case Western Reserve University Hospitals, Cleveland, OH, USA
| | - Devan Wilkins
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Gabrielle Fortier
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Saahithi Mallapragada
- Division of Bioinnovation and Genome Science, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Nicholas E Banovich
- Division of Bioinnovation and Genome Science, Translational Genomics Research Institute, Phoenix, AZ, USA
- Biodevelopmental Origins of Lung Disease (BOLD) Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Laurie C Eldredge
- Center for Respiratory Biology and Therapeutics, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, USA
| | - Gail H Deutsch
- Center for Respiratory Biology and Therapeutics, Seattle Children's Research Institute, Seattle Children's Hospital, Seattle, WA, USA
| | - Christopher V E Wright
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
- Biodevelopmental Origins of Lung Disease (BOLD) Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Program in Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - David B Frank
- Department of Pediatrics, Division of Cardiology, Children's Hospital of Philadelphia, Penn Cardiovascular Institute, Penn-CHOP Lung Biology Institute, Philadelphia, PA, USA
| | - Jonathan A Kropski
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA.
- Biodevelopmental Origins of Lung Disease (BOLD) Center, Vanderbilt University Medical Center, Nashville, TN, USA.
- Program in Developmental Biology, Vanderbilt University, Nashville, TN, USA.
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Veterans Affairs Medical Center, Nashville, TN, USA.
| | - Jennifer M S Sucre
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA.
- Biodevelopmental Origins of Lung Disease (BOLD) Center, Vanderbilt University Medical Center, Nashville, TN, USA.
- Program in Developmental Biology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
5
|
Lin G, Chancellor SE, Kwon T, Woodbury ME, Doering A, Abdourahman A, Bennett RE, Liao F, Pastika T, Tamm J, Romanul N, Yanamandra K, Hu M, Zhao K, Frosch MP, Grinberg Y, Li H, Das S, Dellovade T, Karran EH, Talanian RV, Biber K, Serrano-Pozo A, Ried JS, Langlois X, Hyman BT. Cell-death pathways and tau-associated neuronal vulnerability in Alzheimer's disease. Cell Rep 2025; 44:115758. [PMID: 40448997 DOI: 10.1016/j.celrep.2025.115758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 11/15/2024] [Accepted: 05/08/2025] [Indexed: 06/02/2025] Open
Abstract
Neuronal loss is the ultimate driver of neural system dysfunction in Alzheimer's disease (AD). We used single-nucleus RNA sequencing and neuropathological phenotyping to elucidate mechanisms of neurodegeneration in AD by identifying vulnerable neuronal populations and probing for their differentially expressed genes. Evidenced by transcriptomic analyses and quantitative tau immunoassays of human AD and non-AD brain tissue, we identified a neuronal population especially vulnerable to tau pathology. Multiplexed immunohistochemistry and in situ hybridization (CBLN2 and LINC00507) validated the presence of the tau-vulnerable neuronal population and revealed a propensity of this population to bear tau pathology. Differentially expressed genes associated with phospho-tau pathology in these neurons revealed genes involved in apoptosis, cell-component dissociation (e.g., autophagosome maturation and actin filament depolymerization), and regulation of vesicle-mediated transport.
Collapse
Affiliation(s)
- Gen Lin
- AbbVie Pte Ltd, North Buona Vista Road #19-01, Singapore 138588, Singapore
| | - Sarah E Chancellor
- AbbVie Inc., Cambridge Research Center, 200 Sidney Street, Cambridge, MA 02139, USA.
| | - Taekyung Kwon
- AbbVie Inc., Cambridge Research Center, 200 Sidney Street, Cambridge, MA 02139, USA
| | - Maya E Woodbury
- AbbVie Inc., Cambridge Research Center, 200 Sidney Street, Cambridge, MA 02139, USA
| | - Astrid Doering
- AbbVie Deutschland GmbH & Co. KG, Knollstraße, 67061 Ludwigshafen, Germany
| | - Aicha Abdourahman
- AbbVie Inc., Cambridge Research Center, 200 Sidney Street, Cambridge, MA 02139, USA
| | - Rachel E Bennett
- Department of Neurology, Harvard Medical School, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Fan Liao
- AbbVie Inc., Cambridge Research Center, 200 Sidney Street, Cambridge, MA 02139, USA
| | - Timothy Pastika
- AbbVie Inc., Cambridge Research Center, 200 Sidney Street, Cambridge, MA 02139, USA
| | - Joseph Tamm
- AbbVie Inc., Cambridge Research Center, 200 Sidney Street, Cambridge, MA 02139, USA
| | - Nandini Romanul
- AbbVie Inc., Cambridge Research Center, 200 Sidney Street, Cambridge, MA 02139, USA
| | - Kiran Yanamandra
- AbbVie Inc., Cambridge Research Center, 200 Sidney Street, Cambridge, MA 02139, USA
| | - Miwei Hu
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Karen Zhao
- AbbVie Inc., Cambridge Research Center, 200 Sidney Street, Cambridge, MA 02139, USA
| | - Matthew P Frosch
- Department of Neurology, Harvard Medical School, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Yelena Grinberg
- AbbVie Inc., Cambridge Research Center, 200 Sidney Street, Cambridge, MA 02139, USA
| | - Huan Li
- Department of Neurology, Harvard Medical School, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Sudeshna Das
- Department of Neurology, Harvard Medical School, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Tammy Dellovade
- AbbVie Inc., Cambridge Research Center, 200 Sidney Street, Cambridge, MA 02139, USA
| | - Eric H Karran
- AbbVie Inc., Cambridge Research Center, 200 Sidney Street, Cambridge, MA 02139, USA
| | - Robert V Talanian
- AbbVie Inc., Cambridge Research Center, 200 Sidney Street, Cambridge, MA 02139, USA
| | - Knut Biber
- AbbVie Deutschland GmbH & Co. KG, Knollstraße, 67061 Ludwigshafen, Germany
| | - Alberto Serrano-Pozo
- Department of Neurology, Harvard Medical School, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Janina S Ried
- AbbVie Deutschland GmbH & Co. KG, Knollstraße, 67061 Ludwigshafen, Germany
| | - Xavier Langlois
- AbbVie Inc., Cambridge Research Center, 200 Sidney Street, Cambridge, MA 02139, USA.
| | - Bradley T Hyman
- Department of Neurology, Harvard Medical School, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, MA 02129, USA.
| |
Collapse
|
6
|
Hewitt RJ, Pearmain L, Lyka E, Dickens J. Epithelial damage and ageing: the perfect storm. Thorax 2025:thorax-2024-222060. [PMID: 40425299 DOI: 10.1136/thorax-2024-222060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 04/14/2025] [Indexed: 05/29/2025]
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a progressive disease of lung parenchymal scarring that is triggered by repeated microinjury to a vulnerable alveolar epithelium. It is increasingly recognised that cellular ageing, whether physiological or accelerated due to telomere dysfunction, renders the epithelium less able to cope with injury and triggers changes in epithelial behaviour that ultimately lead to the development of disease. AIMS This review aims to highlight how, with increasing age, the alveolar epithelium becomes vulnerable to exogenous insults. We discuss the downstream consequences of alveolar epithelial dysfunction on epithelial phenotype, alveolar repair and pro-pathogenic interactions with other alveolar niche-resident cell types which drive IPF pathogenesis. NARRATIVE We highlight how a wide array of cellular mechanisms that maintain cellular homeostasis become dysfunctional with ageing. Waning replicative capacity, genomic stability, mitochondrial function, proteostasis and metabolic function all contribute to a phenotype of vulnerability to 'second hits'. We discuss how in IPF the alveolar epithelium becomes dysfunctional, highlighting changes in repair capacity and fundamental cellular phenotype and how interactions between abnormal epithelium and other alveolar niche-resident cell types perpetuate disease. CONCLUSIONS The ageing epithelium is a vulnerable epithelium which, with the cumulative effects of environmental exposures, fundamentally changes its behaviour towards stalled differentiation, failed repair and profibrotic signalling. Further dissection of aberrant epithelial behaviour, and its impact on other alveolar cell types, will allow identification of novel therapeutic targets aimed at earlier pathogenic events.
Collapse
Affiliation(s)
- Richard J Hewitt
- King's Centre for Lung Health, King's College London, London, UK
| | - Laurence Pearmain
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- ILD Unit, North West Lung Centre, Wythenshawe Hospital, Manchester Foundation Trust, Manchester, UK
| | - Elisavet Lyka
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Jennifer Dickens
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Cambridge Institiute for Medical Research, University of Cambridge, Cambridge, UK
- Royal Papworth NHS Foundation Trust, Cambridge, UK
| |
Collapse
|
7
|
Kawasaki K, Salehi S, Zhan YA, Chen K, Lee JH, Salataj E, Zhong H, Manoj P, Kinyua D, Mello BP, Sridhar H, Tischfield SE, Linkov I, Ceglia N, Zatzman M, Havasov E, Shah NJ, Meng F, Loomis B, Bhanot UK, Redin E, de Stanchina E, Hamard PJ, Koche RP, McPherson A, Quintanal-Villalonga Á, Shah SP, Massagué J, Rudin CM. FOXA2 promotes metastatic competence in small cell lung cancer. Nat Commun 2025; 16:4865. [PMID: 40419484 PMCID: PMC12106783 DOI: 10.1038/s41467-025-60141-5] [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: 10/22/2024] [Accepted: 05/14/2025] [Indexed: 05/28/2025] Open
Abstract
Small cell lung cancer (SCLC) is known for its high metastatic potential, with most patients demonstrating clinically evident metastases in multiple organs at diagnosis. The factors contributing to this exceptional metastatic capacity have not been defined. To bridge this gap, we compare gene expression in SCLC patient samples who never experienced metastasis or relapse throughout their clinical course, versus primary SCLC patient samples from more typical patients who had metastatic disease at diagnosis. This analysis identifies FOXA2 as a transcription factor strongly associated with SCLC metastasis. Subsequent analyses in experimental models demonstrates that FOXA2 induces a fetal neuroendocrine gene expression program and promotes multi-site metastasis. Moreover, we identify ASCL1, a transcription factor known for its initiating role in SCLC tumorigenesis, as a direct binder of the FOXA2 promoter and regulator of FOXA2 expression. Taken together, these data define the ASCL1-FOXA2 axis as a critical driver of multiorgan SCLC metastasis.
Collapse
Affiliation(s)
- Kenta Kawasaki
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sohrab Salehi
- Computational Oncology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yingqian A Zhan
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kevin Chen
- Antitumor Assessment Core, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jun Ho Lee
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Eralda Salataj
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hong Zhong
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Parvathy Manoj
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dennis Kinyua
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Barbara P Mello
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Harsha Sridhar
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sam E Tischfield
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Irina Linkov
- Pathology Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nicholas Ceglia
- Computational Oncology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Matthew Zatzman
- Computational Oncology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Eliyahu Havasov
- Computational Oncology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Neil J Shah
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fanli Meng
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Brian Loomis
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Umesh K Bhanot
- Pathology Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Esther Redin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elisa de Stanchina
- Antitumor Assessment Core, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pierre-Jacques Hamard
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Richard P Koche
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrew McPherson
- Computational Oncology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Sohrab P Shah
- Computational Oncology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joan Massagué
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell Medicine Graduate School of Medical Sciences, New York, NY, USA.
| | - Charles M Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell Medicine Graduate School of Medical Sciences, New York, NY, USA.
| |
Collapse
|
8
|
Duong TE, Diep D, Conklin KY, Bui I, Purkerson JM, Boone E, Olness J, Patel S, Peng B, Kern C, Zhao Z, Misra RS, Huyck HL, Verheyden JM, Borok Z, Zhang Y, Scheuermann RH, Zhu Q, Deutsch G, Hagood J, Sun X, Zhang K, Pryhuber GS. A Multimodal Spatial and Epigenomic Atlas of Human Adult Lung Topography. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.05.23.655666. [PMID: 40475598 PMCID: PMC12140004 DOI: 10.1101/2025.05.23.655666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/18/2025]
Abstract
Developing high-resolution reference maps of disease-susceptible spatial niches is a critical step to mitigating the profound effects of lung disease. Here, we present an integrated multimodal single-nucleus human lung atlas (snHLA) profiling 746,047 nuclei from 49 mapped lung blocks spanning clinically relevant distal airways, alveoli, and interstitium across 11 healthy adults. Integrating snRNA-seq and SNARE-seq2, which co-assays chromatin accessibility and gene expression from the same nucleus, we resolved 70 molecularly distinct populations and captured 332,846 accessible chromatin regions, nominating new transcriptional regulators of human lung cell diversity. Spatial transcriptomics using MERFISH mapped 25 cell populations across 7 structural neighborhoods and multiplexed immunofluorescence localized cell subtypes and distal airway-defining protein markers, expanding and validating distinct lung structure-specific cell populations. This open access snHLA and companion Cell Type and Marker Gene Dictionary with anatomically aligned nomenclature delivers a foundational resource at an unprecedented resolution to interrogate the origins of lung pathophysiology.
Collapse
Affiliation(s)
| | - Dinh Diep
- Department of Bioengineering, University of California San Diego, La Jolla, CA
| | - Kimberly Y Conklin
- Department of Bioengineering, University of California San Diego, La Jolla, CA
| | - Indy Bui
- Department of Pediatrics, University of California San Diego, La Jolla, CA
| | | | - Eric Boone
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA
| | - Jacqueline Olness
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA
| | - Sahil Patel
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA
| | - Beverly Peng
- Department of Informatics, J. Craig Venter Institute, La Jolla, CA
| | - Colin Kern
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA
| | - Zoey Zhao
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA
| | - Ravi S Misra
- Department of Pediatrics, University of Rochester, Rochester, NY
| | - Heidie L Huyck
- Department of Pediatrics, University of Rochester, Rochester, NY
| | - Jamie M Verheyden
- Department of Pediatrics, University of California San Diego, La Jolla, CA
| | - Zea Borok
- Division of Pulmonary, Critical Care, Sleep Medicine and Physiology, Department of Medicine, University of California San Diego, La Jolla, CA
| | - Yun Zhang
- National Library of Medicine, National Institutes of Health, Bethesda, MD
| | | | - Quan Zhu
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA
| | - Gail Deutsch
- Department of Pathology, Seattle Children's Research Center, Seattle, WA
| | - James Hagood
- Department of Pediatrics, Children's Research Institute, University of North Carolina, Chapel Hill, NC
| | - Xin Sun
- Department of Pediatrics, University of California San Diego, La Jolla, CA
| | - Kun Zhang
- Department of Bioengineering, University of California San Diego, La Jolla, CA
| | | |
Collapse
|
9
|
Joulia R, Patti S, Traves WJ, Loewenthal L, Yates L, Walker SA, Puttur F, Al-Sahaf M, Cahill KN, Lai J, Siddiqui S, Boyce JA, Israel E, Lloyd CM. A single-cell spatial chart of the airway wall reveals proinflammatory cellular ecosystems and their interactions in health and asthma. Nat Immunol 2025:10.1038/s41590-025-02161-3. [PMID: 40399607 DOI: 10.1038/s41590-025-02161-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 04/11/2025] [Indexed: 05/23/2025]
Abstract
Determining spatial location of cells within tissues gives vital insight into the interactions between resident and inflammatory cells and is a critical factor for uncoupling the mechanisms driving disease. Here, we apply single-cell spatial transcriptomics to reveal the airway wall landscape in health and during asthma. We identified proinflammatory cellular ecosystems that exist within discrete spatial niches in healthy and asthma samples. These cellular hubs are characterized by a high level of chemokine and alarmin expression, along with unique combinations of stromal cells. Mechanistically, we demonstrated that receptors, such as ACKR1, retain immune mediators locally, while amphiregulin-expressing mast cells are prominent within these proinflammatory hubs. Despite anti-inflammatory treatments, the asthma airway mucosa exhibited a distinct remodeling program within these cellular ecosystems, marked by increased proximity between key cell types. This study provides an unprecedented view of the topography of the airway wall, revealing distinct, specific ecosystems within spatial niches to target for therapeutic intervention.
Collapse
Affiliation(s)
- Régis Joulia
- National Heart and Lung Institute, Imperial College London, London, UK.
| | - Sara Patti
- National Heart and Lung Institute, Imperial College London, London, UK
| | - William J Traves
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Lola Loewenthal
- National Heart and Lung Institute, Imperial College London, London, UK
- Department of Asthma and Allergy, Royal Brompton and Harefield Hospitals, London, UK
- Department of Respiratory Medicine, Royal Brompton and Harefield Hospitals, London, UK
| | - Laura Yates
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Simone A Walker
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Franz Puttur
- National Heart and Lung Institute, Imperial College London, London, UK
| | - May Al-Sahaf
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Katherine N Cahill
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Juying Lai
- Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA, USA
- Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Salman Siddiqui
- National Heart and Lung Institute, Imperial College London, London, UK
- Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Joshua A Boyce
- Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA, USA
- Jeff and Penny Vinik Center for Allergic Disease Research, Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, MA, USA
| | - Elliot Israel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Clare M Lloyd
- National Heart and Lung Institute, Imperial College London, London, UK.
| |
Collapse
|
10
|
Pentimalli TM, Schallenberg S, León-Periñán D, Legnini I, Theurillat I, Thomas G, Boltengagen A, Fritzsche S, Nimo J, Ruff L, Dernbach G, Jurmeister P, Murphy S, Gregory MT, Liang Y, Cordenonsi M, Piccolo S, Coscia F, Woehler A, Karaiskos N, Klauschen F, Rajewsky N. Combining spatial transcriptomics and ECM imaging in 3D for mapping cellular interactions in the tumor microenvironment. Cell Syst 2025; 16:101261. [PMID: 40220761 DOI: 10.1016/j.cels.2025.101261] [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: 07/12/2024] [Revised: 12/13/2024] [Accepted: 03/19/2025] [Indexed: 04/14/2025]
Abstract
Tumors are complex ecosystems composed of malignant and non-malignant cells embedded in a dynamic extracellular matrix (ECM). In the tumor microenvironment, molecular phenotypes are controlled by cell-cell and ECM interactions in 3D cellular neighborhoods (CNs). While their inhibition can impede tumor progression, routine molecular tumor profiling fails to capture cellular interactions. Single-cell spatial transcriptomics (ST) maps receptor-ligand interactions but usually remains limited to 2D tissue sections and lacks ECM readouts. Here, we integrate 3D ST with ECM imaging in serial sections from one clinical lung carcinoma to systematically quantify molecular states, cell-cell interactions, and ECM remodeling in CN. Our integrative analysis pinpointed known immune escape and tumor invasion mechanisms, revealing several druggable drivers of tumor progression in the patient under study. This proof-of-principle study highlights the potential of in-depth CN profiling in routine clinical samples to inform microenvironment-directed therapies. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Tancredi Massimo Pentimalli
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin
| | - Simon Schallenberg
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Daniel León-Periñán
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Ivano Legnini
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Human Technopole, Milan, Italy
| | - Ilan Theurillat
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Gwendolin Thomas
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Anastasiya Boltengagen
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Sonja Fritzsche
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany; Humboldt-Universität zu Berlin, Institute of Biology, 10099 Berlin, Germany
| | - Jose Nimo
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin; Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany; Humboldt-Universität zu Berlin, Institute of Biology, 10099 Berlin, Germany
| | | | - Gabriel Dernbach
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany; Aignostics GmbH, Berlin, Germany; BIFOLD - Berlin Institute for the Foundations of Learning and Data, Berlin, Germany
| | | | | | | | - Yan Liang
- NanoString® Technologies, Inc, Seattle, WA, USA
| | | | - Stefano Piccolo
- Department of Molecular Medicine, University of Padua, Padua, Italy; IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Fabian Coscia
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany
| | - Andrew Woehler
- Systems Biology Imaging Platform, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA, USA
| | - Nikos Karaiskos
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Frederick Klauschen
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin; BIFOLD - Berlin Institute for the Foundations of Learning and Data, Berlin, Germany; Institute of Pathology, Ludwig Maximilians Universität, Munich, Germany
| | - Nikolaus Rajewsky
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin; German Center for Cardiovascular Research (DZHK), Site Berlin, Berlin, Germany; NeuroCure Cluster of Excellence, Berlin, Germany; German Cancer Consortium (DKTK), Berlin, Germany; National Center for Tumor Diseases (NCT), Site Berlin, Berlin, Germany.
| |
Collapse
|
11
|
Kellner MJ, Monteil VM, Zelger P, Pei G, Jiao J, Onji M, Nayak K, Zilbauer M, Balkema-Buschmann A, Dorhoi A, Mirazimi A, Penninger JM. Bat organoids reveal antiviral responses at epithelial surfaces. Nat Immunol 2025:10.1038/s41590-025-02155-1. [PMID: 40399606 DOI: 10.1038/s41590-025-02155-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 04/08/2025] [Indexed: 05/23/2025]
Abstract
Bats can host viruses of pandemic concern without developing disease. The mechanisms underlying their exceptional resilience to viral infections are largely unresolved, necessitating the development of physiologically relevant and genetically tractable research models. Here, we developed respiratory and intestinal organoids that recapitulated the cellular diversity of the in vivo epithelium present in Rousettus aegyptiacus, the natural reservoir for the highly pathogenic Marburg virus (MARV). In contrast to human counterparts, bat organoids and mucosal tissue exhibited elevated constitutive expression of innate immune effectors, including type I interferon-ε (IFNε) and IFN-stimulated genes (ISGs). Upon infection with diverse zoonotic viruses, including MARV, bat organoids strongly induced type I and III IFN responses, which conferred robust antiviral protection. Type III IFNλ3 additionally displayed virus-independent self-amplification, acting as an ISG to enhance antiviral immunity. Our organoid platform reveals key features of bat epithelial antiviral immunity that may inform therapeutic strategies for viral disease resilience.
Collapse
Affiliation(s)
- Max J Kellner
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Vienna, Austria.
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna, Austria.
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria.
- Helmholtz Centre for Infection Research, Braunschweig, Germany.
| | - Vanessa M Monteil
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
- Public Health Agency of Sweden, Solna, Sweden
| | - Patrick Zelger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Gang Pei
- Institute of Immunology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | - Jie Jiao
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Masahiro Onji
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Komal Nayak
- Wellcome - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Matthias Zilbauer
- Wellcome - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Anne Balkema-Buschmann
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | - Anca Dorhoi
- Institute of Immunology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
- Faculty of Mathematics and Natural Sciences, University of Greifswald, Greifswald, Germany
| | - Ali Mirazimi
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
- National Veterinary Institute, Uppsala, Sweden
- Public Health Agency of Sweden, Solna, Sweden
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Vienna, Austria.
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria.
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada.
- Helmholtz Centre for Infection Research, Braunschweig, Germany.
| |
Collapse
|
12
|
Rahal Z, El Darzi R, Moghaddam SJ, Cascone T, Kadara H. Tumour and microenvironment crosstalk in NSCLC progression and response to therapy. Nat Rev Clin Oncol 2025:10.1038/s41571-025-01021-1. [PMID: 40379986 DOI: 10.1038/s41571-025-01021-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2025] [Indexed: 05/19/2025]
Abstract
The treatment landscape of non-small-cell lung cancer (NSCLC) is evolving rapidly, driven by advances in the development of targeted agents and immunotherapies. Despite this progress, some patients have suboptimal responses to treatment, highlighting the need for new therapeutic strategies. In the past decade, the important role of the tumour microenvironment (TME) in NSCLC progression, metastatic dissemination and response to treatment has become increasingly evident. Understanding the complexity of the TME and its interactions with NSCLC can propel efforts to improve current treatment modalities, overcome resistance and develop new treatments, which will ultimately improve the outcomes of patients. In this Review, we provide a comprehensive view of the NSCLC TME, examining its components and highlighting distinct archetypes characterized by spatial niches within and surrounding tumour nests, which form complex neighbourhoods. Next, we explore the interactions within these components, focusing on how inflammation and immunosuppression shape the dynamics of the NSCLC TME. We also address the emerging influences of patient-related factors, such as ageing, sex and health disparities, on the NSCLC-TME crosstalk. Finally, we discuss how various therapeutic strategies interact with and are influenced by the TME in NSCLC. Overall, we emphasize the interconnectedness of these elements and how they influence therapeutic outcomes and tumour progression.
Collapse
Affiliation(s)
- Zahraa Rahal
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Roy El Darzi
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Seyed Javad Moghaddam
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Graduate School of Biomedical Sciences (GSBS), UTHealth Houston, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tina Cascone
- Graduate School of Biomedical Sciences (GSBS), UTHealth Houston, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Thoracic-Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Humam Kadara
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Graduate School of Biomedical Sciences (GSBS), UTHealth Houston, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
13
|
Donoghue LJ, Benner C, Chang D, Irudayanathan FJ, Pendergrass RK, Yaspan BL, Mahajan A, McCarthy MI. Integration of biobank-scale genetics and plasma proteomics reveals evidence for causal processes in asthma risk and heterogeneity. CELL GENOMICS 2025; 5:100840. [PMID: 40187354 DOI: 10.1016/j.xgen.2025.100840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 01/20/2025] [Accepted: 03/07/2025] [Indexed: 04/07/2025]
Abstract
Hundreds of genetic associations for asthma have been identified, yet translating these findings into mechanistic insights remains challenging. We leveraged plasma proteomics from the UK Biobank Pharma Proteomics Project (UKB-PPP) to identify biomarkers and effectors of asthma risk or heterogeneity using genetic causal inference approaches. We identified 609 proteins associated with asthma status (269 proteins after controlling for body mass index [BMI] and smoking). Analysis of genetically predicted protein levels identified 70 proteins with putative causal roles in asthma risk, including known drug targets and proteins without prior genetic evidence in asthma (e.g., GCHFR, TDRKH, and CLEC7A). The genetic architecture of causally associated proteins provided evidence for a Toll-like receptor (TLR)1-interleukin (IL)-27 asthma axis. Lastly, we identified evidence of causal relationships between proteins and heterogeneous aspects of asthma biology, including between TSPAN8 and neutrophil counts. These findings illustrate that integrating biobank-scale genetics and plasma proteomics can provide a framework to identify therapeutic targets and mechanisms underlying disease risk and heterogeneity.
Collapse
Affiliation(s)
| | | | - Diana Chang
- Genentech, Inc., South San Francisco, CA, USA
| | | | | | | | | | | |
Collapse
|
14
|
Zhou Y, Sheng Q, Jin S. Integrating single-cell data with biological variables. Proc Natl Acad Sci U S A 2025; 122:e2416516122. [PMID: 40294274 PMCID: PMC12067276 DOI: 10.1073/pnas.2416516122] [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: 08/15/2024] [Accepted: 03/30/2025] [Indexed: 04/30/2025] Open
Abstract
Constructing single-cell atlases requires preserving differences attributable to biological variables, such as cell types, tissue origins, and disease states, while eliminating batch effects. However, existing methods are inadequate in explicitly modeling these biological variables. Here, we introduce SIGNAL, a general framework that leverages biological variables to disentangle biological and technical effects, thereby linking these metadata to data integration. SIGNAL employs a variant of principal component analysis to align multiple batches, enabling the integration of 1 million cells in approximately 2 min. SIGNAL, despite its computational simplicity, surpasses state-of-the-art methods across multiple integration scenarios: 1) heterogeneous datasets, 2) cross-species datasets, 3) simulated datasets, 4) integration on low-quality cell annotations, and 5) reference-based integration. Furthermore, we demonstrate that SIGNAL accurately transfers knowledge from reference to query datasets. Notably, we propose a self-adjustment strategy to restore annotated cell labels potentially distorted during integration. Finally, we apply SIGNAL to multiple large-scale atlases, including a human heart cell atlas containing 2.7 million cells, identifying tissue- and developmental stage-specific subtypes, as well as condition-specific cell states. This underscores SIGNAL's exceptional capability in multiscale analysis.
Collapse
Affiliation(s)
- Yang Zhou
- School of Mathematics, Harbin Institute of Technology, Harbin150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou450000, China
| | - Qiongyu Sheng
- School of Mathematics, Harbin Institute of Technology, Harbin150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou450000, China
| | - Shuilin Jin
- School of Mathematics, Harbin Institute of Technology, Harbin150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou450000, China
| |
Collapse
|
15
|
Nagai JS, Maié T, Schaub MT, Costa IG. scACCorDiON: a clustering approach for explainable patient level cell-cell communication graph analysis. Bioinformatics 2025; 41:btaf288. [PMID: 40327499 PMCID: PMC12122081 DOI: 10.1093/bioinformatics/btaf288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 04/29/2025] [Accepted: 05/05/2025] [Indexed: 05/08/2025] Open
Abstract
MOTIVATION Combining single-cell sequencing with ligand-receptor (LR) analysis paves the way for the characterization of cell communication events in complex tissues. In particular, directed weighted graphs naturally represent cell-cell communication events. However, current computational methods cannot yet analyze sample-specific cell-cell communication events, as measured in single-cell data produced in large patient cohorts. Cohort-based cell-cell communication analysis presents many challenges, such as the nonlinear nature of cell-cell communication and the high variability given by the patient-specific single-cell RNAseq datasets. RESULTS Here, we present scACCorDiON (single-cell Analysis of Cell-Cell Communication in Disease clusters using Optimal transport in Directed Networks), an optimal transport algorithm exploring node distances on the Markov Chain as the ground metric between directed weighted graphs. Benchmarking indicates that scACCorDiON performs a better clustering of samples according to their disease status than competing methods that use undirected graphs. We provide a case study of pancreas adenocarcinoma, where scACCorDion detects a sub-cluster of disease samples associated with changes in the tumor microenvironment. Our study case corroborates that clusters provide a robust and explainable representation of cell-cell communication events and that the expression of detected LR pairs is predictive of pancreatic cancer survival. AVAILABILITY AND IMPLEMENTATION The code of scACCorDiON is available at https://scaccordion.readthedocs.io/en/latest/. and https://doi.org/10.5281/zenodo.15267648. The survival analysis package can be found at https://github.com/CostaLab/scACCorDiON.su.
Collapse
Affiliation(s)
- James S Nagai
- Institute for Computational Genomics, RWTH Aachen Medical Faculty, Aachen 52074, Germany
| | - Tiago Maié
- Institute for Computational Genomics, RWTH Aachen Medical Faculty, Aachen 52074, Germany
| | - Michael T Schaub
- Department of Computational Science, RWTH Aachen University, Aachen 52074, Germany
| | - Ivan G Costa
- Institute for Computational Genomics, RWTH Aachen Medical Faculty, Aachen 52074, Germany
| |
Collapse
|
16
|
Chen H, Nguyen ND, Ruffalo M, Bar-Joseph Z. A unified analysis of atlas single-cell data. Genome Res 2025; 35:1219-1233. [PMID: 39965934 PMCID: PMC12047537 DOI: 10.1101/gr.279631.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 02/03/2025] [Indexed: 02/20/2025]
Abstract
Recent efforts to generate atlas-scale single-cell data provide opportunities for joint analysis across tissues and modalities. Existing methods use cells as the reference unit, hindering downstream gene-based analysis and removing genuine biological variation. Here we present GIANT, an integration method designed for atlas-scale gene analysis across cell types and tissues. GIANT converts data sets into gene graphs and recursively embeds genes without additional alignment. Applying GIANT to two recent atlas data sets yields unified gene-embedding spaces across human tissues and data modalities. Further evaluations demonstrate GIANT's usefulness in discovering diverse gene functions and underlying gene regulation in cells from different tissues.
Collapse
Affiliation(s)
- Hao Chen
- Ray and Stephanie Lane Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, USA
- Department of Computer Science, University of Illinois Chicago, Chicago, Illinois 60607, USA
| | - Nam D Nguyen
- Ray and Stephanie Lane Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, USA
| | - Matthew Ruffalo
- Ray and Stephanie Lane Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, USA
| | - Ziv Bar-Joseph
- Ray and Stephanie Lane Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, USA;
- Machine Learning Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, USA
| |
Collapse
|
17
|
Theunissen L, Mortier T, Saeys Y, Waegeman W. Evaluation of out-of-distribution detection methods for data shifts in single-cell transcriptomics. Brief Bioinform 2025; 26:bbaf239. [PMID: 40439669 PMCID: PMC12121363 DOI: 10.1093/bib/bbaf239] [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: 01/28/2025] [Revised: 04/01/2025] [Accepted: 05/05/2025] [Indexed: 06/02/2025] Open
Abstract
Automatic cell-type annotation methods assign cell-type labels to new, unlabeled datasets by leveraging relationships from a reference RNA-seq atlas. However, new datasets may include labels absent from the reference dataset or exhibit feature distributions that diverge from it. These scenarios can significantly affect the reliability of cell type predictions, a factor often overlooked in current automatic annotation methods. The field of out-of-distribution detection (OOD), primarily focused on computer vision, addresses the identification of instances that differ from the training distribution. Therefore, the implementation of OOD methods in the context of novel cell type annotation and data shift detection for single-cell transcriptomics may enhance annotation accuracy and trustworthiness. We evaluate six OOD detection methods: LogitNorm, MC dropout, Deep Ensembles, Energy-based OOD, Deep NN, and Posterior networks, for their annotation and OOD detection performance in both synthetical and real-life application settings. We show that OOD detection methods can accurately identify novel cell types and demonstrate potential to detect significant data shifts in non-integrated datasets. Moreover, we find that integration of the OOD datasets does not interfere with OOD detection of novel cell types.
Collapse
Affiliation(s)
- Lauren Theunissen
- Data Mining and Modeling for Biomedicine, VIB Center for Inflammation Research and VIB Center for AI and Computational Biology (VIB.AI), 9000 Ghent, Belgium
- Department of Data-analysis and Mathematical Modeling, Ghent University Faculty of Bioscience Engineering, 9000 Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University Faculty of Sciences, 9000 Ghent, Belgium
| | - Thomas Mortier
- Department of Data-analysis and Mathematical Modeling, Ghent University Faculty of Bioscience Engineering, 9000 Ghent, Belgium
- Department of Environment, Ghent University Faculty of Bioscience Engineering, 9000 Ghent, Belgium
| | - Yvan Saeys
- Data Mining and Modeling for Biomedicine, VIB Center for Inflammation Research and VIB Center for AI and Computational Biology (VIB.AI), 9000 Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University Faculty of Sciences, 9000 Ghent, Belgium
| | - Willem Waegeman
- Department of Data-analysis and Mathematical Modeling, Ghent University Faculty of Bioscience Engineering, 9000 Ghent, Belgium
| |
Collapse
|
18
|
Narke D, Moorthy B. The Central Role of Cytochrome P450 Reductase (CPR) in Hyperoxic Lung Injury. Expert Opin Drug Metab Toxicol 2025; 21:589-598. [PMID: 39992710 DOI: 10.1080/17425255.2025.2470808] [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: 10/28/2024] [Revised: 01/22/2025] [Accepted: 02/17/2025] [Indexed: 02/26/2025]
Abstract
INTRODUCTION Hyperoxic lung injury results from excessive supplemental oxygen therapy in conditions such as bronchopulmonary dysplasia (BPD) in preterm infants and acute respiratory distress syndrome (ARDS) in adults. This review explores the role of cytochrome P450 reductase (CPR) in hyperoxic lung injury. AREAS COVERED Hyperoxia induces the production of reactive oxygen species in excessive amounts, overwhelming the body's antioxidant defenses and exacerbating lung injury in ARDS/BPD. This review examines the differential roles of CPR-dependent enzymes in the context of hyperoxic lung injury. Additionally, we highlight the potential of targeting CPR to study mechanisms of lung injury and leverage gene-editing technologies to deepen our understanding of CPR-mediated pathways. This review consolidates existing knowledge on CPR-dependent processes and their roles in hyperoxic lung injury, based on a literature search conducted in the PubMed database for studies published between 1988 and 2024. EXPERT OPINION This review emphasizes the need for a deeper understanding of disease mechanisms, particularly CPR-mediated pathways. As a regulatory hub for ROS modulation and enzyme activity, CPR represents a promising target, offering a unified strategy to mitigate hyperoxic lung injury and improve outcomes in BPD/ARDS.
Collapse
Affiliation(s)
- Deven Narke
- Department of Pediatrics-Newborn, Baylor College of Medicine, Houston, TX, USA
| | - Bhagavatula Moorthy
- Department of Pediatrics-Newborn, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
19
|
Hagood JS. A Developmental Step Along the 'Omics Journey. Am J Respir Cell Mol Biol 2025; 72:464-465. [PMID: 39565183 PMCID: PMC12051909 DOI: 10.1165/rcmb.2024-0524ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 11/19/2024] [Indexed: 11/21/2024] Open
Affiliation(s)
- James S Hagood
- Department of Pediatrics (Pulmonology) University of North Carolina at Chapel Hill Chapel Hill, North Carolina
| |
Collapse
|
20
|
Conrad C, Magnen M, Tsui J, Wismer H, Naser M, Venkataramani U, Samad B, Cleary SJ, Qiu L, Tian JJ, De Giovanni M, Mende N, Leavitt AD, Passegué E, Laurenti E, Combes AJ, Looney MR. Decoding functional hematopoietic progenitor cells in the adult human lung. Blood 2025; 145:1975-1986. [PMID: 40014797 PMCID: PMC7617544 DOI: 10.1182/blood.2024027884] [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: 12/04/2024] [Revised: 01/10/2025] [Accepted: 01/19/2025] [Indexed: 03/01/2025] Open
Abstract
ABSTRACT Although the bone marrow is the main site of blood cell production in adults, rare pools of hematopoietic stem and progenitor cells have been found in extramedullary organs. In mice, we have previously shown that the lung contains hematopoietic progenitor cells and is a site of platelet production. Here, in the adult human lung, we show that functional hematopoietic precursors reside in the extravascular spaces with a frequency similar to the bone marrow and are capable of proliferation and engraftment in mice. The gene signature of pulmonary and medullary CD34+ hematopoietic progenitors indicates greater baseline activation of immune-, megakaryocyte/platelet-, and erythroid-related pathways in lung progenitors. Spatial transcriptomics mapped blood progenitors in the lung to an alveolar interstitium niche with only a few cells identified in an intravascular location. In human blood samples collected for stem cell transplantation, CD34+ cells with a lung signature enriched the mobilized pool of hematopoietic stem cells. These results identify the lung as a pool for uniquely programmed blood stem and progenitor cells with the potential to support hematopoiesis in humans.
Collapse
Affiliation(s)
- Catharina Conrad
- Department of Medicine, University of California, San Francisco; San Francisco, CA, USA
| | - Mélia Magnen
- Department of Medicine, University of California, San Francisco; San Francisco, CA, USA
| | - Jessica Tsui
- UCSF CoLabs, University of California, San Francisco, CAUSA
| | | | - Mohammad Naser
- UCSF CoLabs, University of California, San Francisco, CAUSA
| | | | - Bushra Samad
- UCSF CoLabs, University of California, San Francisco, CAUSA
| | - Simon J. Cleary
- Department of Medicine, University of California, San Francisco; San Francisco, CA, USA
| | - Longhui Qiu
- Department of Medicine, University of California, San Francisco; San Francisco, CA, USA
| | - Jennifer J. Tian
- Department of Medicine, University of California, San Francisco; San Francisco, CA, USA
| | - Marco De Giovanni
- Department of Microbiology & Immunology, University of California, San Francisco; San Francisco, CA, USA
| | - Nicole Mende
- Wellcome – MRC Cambridge Stem Cell Institute, Department of Hematology, University of Cambridge; Cambridge, UK
| | - Andrew D. Leavitt
- Department of Medicine, University of California, San Francisco; San Francisco, CA, USA
| | - Emmanuelle Passegué
- Columbia Stem Cell Initiative, Columbia University Irving Medical Center; New York, NY, USA
| | - Elisa Laurenti
- Wellcome – MRC Cambridge Stem Cell Institute, Department of Hematology, University of Cambridge; Cambridge, UK
| | - Alexis J. Combes
- Department of Medicine, University of California, San Francisco; San Francisco, CA, USA
- UCSF CoLabs, University of California, San Francisco, CAUSA
- Department of Pathology, University of California, San Francisco; San Francisco, CA, USA
- Bakar ImmunoX Initiative, University of California, San Francisco; San Francisco, CA, USA
- Biomedical Sciences Program, University of California, San Francisco, CA, USA
| | - Mark R. Looney
- Department of Medicine, University of California, San Francisco; San Francisco, CA, USA
- Bakar ImmunoX Initiative, University of California, San Francisco; San Francisco, CA, USA
- Biomedical Sciences Program, University of California, San Francisco, CA, USA
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| |
Collapse
|
21
|
Gómez-López S, Alhendi ASN, Przybilla MJ, Bordeu I, Whiteman ZE, Butler T, Rouhani MJ, Kalinke L, Uddin I, Otter KEJ, Chandrasekharan DP, Lebrusant-Fernandez M, Shurr AYL, Durrenberger PF, Moore DA, Falzon M, Reading JL, Martincorena I, Simons BD, Campbell PJ, Janes SM. Aberrant basal cell clonal dynamics shape early lung carcinogenesis. Science 2025:eads9145. [PMID: 40310937 DOI: 10.1126/science.ads9145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 04/10/2025] [Indexed: 05/03/2025]
Abstract
Preinvasive squamous lung lesions are precursors of lung squamous cell carcinoma (LUSC). The cellular events underlying lesion formation are unknown. Using a carcinogen-induced model of LUSC with no added genetic hits or cell type bias, we find that carcinogen exposure leads to non-neutral competition among basal cells, aberrant clonal expansions, and basal cell mobilization along the airways. Ultimately, preinvasive lesions develop from a few highly mutated clones that dominate most of the bronchial tree. Multi-site sequencing in human patients confirms the presence of clonally related preinvasive lesions across distinct airway regions. Our work identifies a transition in basal cell clonal dynamics, and an associated shift in basal cell fate, as drivers of field cancerization in the lung.
Collapse
Affiliation(s)
- Sandra Gómez-López
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
| | - Ahmed S N Alhendi
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
| | - Moritz J Przybilla
- Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton, UK
| | - Ignacio Bordeu
- Departamento de Física, Facultad de Ciencias Físicas y Matemáticas, Universidad de Chile, Santiago, Chile
| | - Zoe E Whiteman
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
| | - Timothy Butler
- Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton, UK
| | - Maral J Rouhani
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
| | - Lukas Kalinke
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
| | - Imran Uddin
- Cancer Research UK City of London Centre Single Cell Genomics Facility, UCL Cancer Institute, University College London, London, UK
- Genomics Translational Technology Platform, UCL Cancer Institute, University College London, London, UK
| | - Kate E J Otter
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
| | | | - Marta Lebrusant-Fernandez
- Pre-Cancer Immunology Laboratory, UCL Cancer Institute, University College London, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Abigail Y L Shurr
- Pre-Cancer Immunology Laboratory, UCL Cancer Institute, University College London, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Pascal F Durrenberger
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
| | - David A Moore
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Department of Cellular Pathology, University College London Hospitals NHS Trust, London, UK
| | - Mary Falzon
- Department of Cellular Pathology, University College London Hospitals NHS Trust, London, UK
| | - James L Reading
- Pre-Cancer Immunology Laboratory, UCL Cancer Institute, University College London, London, UK
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Iñigo Martincorena
- Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton, UK
| | - Benjamin D Simons
- Department of Applied Mathematics and Theoretical Physics, Centre for Mathematical Sciences, University of Cambridge, Cambridge, UK
- Gurdon Institute, University of Cambridge, Cambridge, UK
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Peter J Campbell
- Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, Hinxton, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Sam M Janes
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
- University College London Hospitals NHS Trust, London, UK
| |
Collapse
|
22
|
Wang J, Ye F, Chai H, Jiang Y, Wang T, Ran X, Xia Q, Xu Z, Fu Y, Zhang G, Wu H, Guo G, Guo H, Ruan Y, Wang Y, Xing D, Xu X, Zhang Z. Advances and applications in single-cell and spatial genomics. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1226-1282. [PMID: 39792333 DOI: 10.1007/s11427-024-2770-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/10/2024] [Indexed: 01/12/2025]
Abstract
The applications of single-cell and spatial technologies in recent times have revolutionized the present understanding of cellular states and the cellular heterogeneity inherent in complex biological systems. These advancements offer unprecedented resolution in the examination of the functional genomics of individual cells and their spatial context within tissues. In this review, we have comprehensively discussed the historical development and recent progress in the field of single-cell and spatial genomics. We have reviewed the breakthroughs in single-cell multi-omics technologies, spatial genomics methods, and the computational strategies employed toward the analyses of single-cell atlas data. Furthermore, we have highlighted the advances made in constructing cellular atlases and their clinical applications, particularly in the context of disease. Finally, we have discussed the emerging trends, challenges, and opportunities in this rapidly evolving field.
Collapse
Affiliation(s)
- Jingjing Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Fang Ye
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Haoxi Chai
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310058, China
| | - Yujia Jiang
- BGI Research, Shenzhen, 518083, China
- BGI Research, Hangzhou, 310030, China
| | - Teng Wang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Xia Ran
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China
| | - Qimin Xia
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China
| | - Ziye Xu
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yuting Fu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Guodong Zhang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Hanyu Wu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Guoji Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Hangzhou, 310058, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China.
| | - Hongshan Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China.
| | - Yijun Ruan
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310058, China.
| | - Yongcheng Wang
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Dong Xing
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China.
- Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, 100871, China.
| | - Xun Xu
- BGI Research, Shenzhen, 518083, China.
- BGI Research, Hangzhou, 310030, China.
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI Research, Shenzhen, 518083, China.
| | - Zemin Zhang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
23
|
Ushakumary MG, Feng S, Bandyopadhyay G, Olson H, Weitz KK, Huyck HL, Poole C, Purkerson JM, Bhattacharya S, Ljungberg MC, Mariani TJ, Deutsch GH, Misra RS, Carson JP, Adkins JN, Pryhuber GS, Clair G. Cell Population-resolved Multiomics Atlas of the Developing Lung. Am J Respir Cell Mol Biol 2025; 72:484-495. [PMID: 39447176 PMCID: PMC12051933 DOI: 10.1165/rcmb.2024-0105oc] [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: 03/04/2024] [Accepted: 10/24/2024] [Indexed: 10/26/2024] Open
Abstract
The lung is a vital organ that undergoes extensive morphological and functional changes during postnatal development. To disambiguate how different cell populations contribute to organ development, we performed proteomic and transcriptomic analyses of four sorted cell populations from the lung of human subjects 0-8 years of age with a focus on early life. The cell populations analyzed included epithelial, endothelial, mesenchymal, and immune cells. Our results revealed distinct molecular signatures for each of the sorted cell populations that enable the description of molecular shifts occurring in these populations during postnatal development. We confirmed that the proteome of the different cell populations was distinct regardless of age and identified functions specific to each population. We identified a series of cell population protein markers, including those located at the cell surface, that show differential expression and distribution on RNA in situ hybridization and immunofluorescence imaging. We validated the spatial distribution of alveolar type 1 and endothelial cell surface markers. Temporal analyses of the proteomes of the four populations revealed processes modulated during postnatal development and clarified the findings obtained from whole-tissue proteome studies. Finally, the proteome was compared with a transcriptomics survey performed on the same lung samples to evaluate processes under post-transcriptional control.
Collapse
Affiliation(s)
- Mereena G. Ushakumary
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington
| | - Song Feng
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington
| | - Gautam Bandyopadhyay
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Heather Olson
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington
| | - Karl K. Weitz
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington
| | - Heidi L. Huyck
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Cory Poole
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Jeffrey M. Purkerson
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Soumyaroop Bhattacharya
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - M. Cecilia Ljungberg
- Department of Pediatrics, College of Medicine, Baylor University, Houston, Texas
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas
| | - Thomas J. Mariani
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Gail H. Deutsch
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Ravi S. Misra
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - James P. Carson
- Texas Advanced Computing Center, University of Texas at Austin, Austin, Texas; and
| | - Joshua N. Adkins
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
| | - Gloria S. Pryhuber
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Geremy Clair
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington
| |
Collapse
|
24
|
Russell DG, Simwela NV, Mattila JT, Flynn J, Mwandumba HC, Pisu D. How macrophage heterogeneity affects tuberculosis disease and therapy. Nat Rev Immunol 2025; 25:370-384. [PMID: 39774813 DOI: 10.1038/s41577-024-01124-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2024] [Indexed: 01/11/2025]
Abstract
Macrophages are the primary host cell type for infection by Mycobacterium tuberculosis in vivo. Macrophages are also key immune effector cells that mediate the control of bacterial growth. However, the specific macrophage phenotypes that are required for optimal immune control of M. tuberculosis infection in vivo remain poorly defined. There are two distinct macrophage lineages in the lung, comprising embryonically derived, tissue-resident alveolar macrophages and recruited, blood monocyte-derived interstitial macrophages. Recent studies have shown that these lineages respond divergently to similar immune environments within the tuberculosis granuloma. Here, we discuss how the differing responses of macrophage lineages might affect the control or progression of tuberculosis disease. We suggest that the ability to reprogramme macrophage responses appropriately, through immunological or chemotherapeutic routes, could help to optimize vaccines and drug regimens for tuberculosis.
Collapse
Affiliation(s)
- David G Russell
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA.
| | - Nelson V Simwela
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Joshua T Mattila
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - JoAnne Flynn
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Henry C Mwandumba
- Malawi Liverpool Wellcome Research Programme, Kamuzu University of Health Sciences, Blantyre, Malawi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Davide Pisu
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
- Department of Microbial Pathogenesis and Immunology, Texas A&M School of Medicine, Bryan, TX, USA
| |
Collapse
|
25
|
Xu W, Yang H, Yao F. Single-cell analysis and machine learning-based integration develop an immune-responsive signature of antigen-presenting cancer-associated fibroblasts in lung adenocarcinoma. J Thorac Dis 2025; 17:2321-2338. [PMID: 40400942 PMCID: PMC12090188 DOI: 10.21037/jtd-2024-2015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/18/2025] [Indexed: 05/23/2025]
Abstract
Background Cancer-associated fibroblasts (CAFs) are pivotal regulators of the tumor immune microenvironment, shaping immune responses and influencing therapeutic outcomes. While previous studies have predominantly focused on CAF subpopulations that impair responses to immune checkpoint inhibitors (ICIs), CAF subsets associated with favorable ICIs responses in lung adenocarcinoma (LUAD) remain underexplored. In this study, we integrated bulk RNA and single-cell RNA sequencing data from LUAD samples to identify CAF subpopulations relevant to ICIs efficacy. Methods Using a machine learning-driven approach, we developed a robust immune response signature based on this antigen-presenting CAFs (apCAFs) subset to predict ICIs responses. Results We uncovered a novel subset of apCAFs exhibiting macrophage-like features, characterized by the expression of major histocompatibility complex (MHC) class II, CD74, and costimulatory molecules (CD80, CD86, CD83, and CD40). This subset, distinct from classic apCAFs described in other cancer types, is strongly associated with favorable ICIs responses across multiple datasets. Notably, these macrophage-like apCAFs are present in LUAD samples prior to treatment, although their abundance varies among individuals. Patients classified as high-risk using signature calculated by a machine learning-driven approach exhibited lower overall survival rates and diminished immune cell infiltration following ICIs therapy. Conclusions Collectively, our findings establish a critical link between macrophage-like apCAFs and ICIs efficacy, offering a clinically applicable signature for patient stratification and guiding therapeutic strategies targeting the tumor microenvironment.
Collapse
Affiliation(s)
- Weijiao Xu
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haitang Yang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Yao
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Thoracic Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
26
|
Svobodová B, Löfdahl A, Kadefors M, Ali SM, Rosmark O, Prabhala P, Magnusson M, Brunnström H, Lundin S, Dellgren G, Müller C, Elowsson L, Westergren-Thorsson G. Collagen VII Is Associated with Airway Remodeling, Honeycombing, and Fibroblast Foci in Usual Interstitial Pneumonia/Idiopathic Pulmonary Fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2025:S0002-9440(25)00140-3. [PMID: 40311757 DOI: 10.1016/j.ajpath.2025.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 02/07/2025] [Accepted: 03/21/2025] [Indexed: 05/03/2025]
Abstract
Collagen VII is an essential anchoring protein in the basement membrane zone, maintaining the attachment of stratified and pseudostratified epithelia to the underlying interstitial matrix. However, collagen VII is largely unexplored in normal lungs and idiopathic pulmonary fibrosis (IPF), a disease characterized by excessive accumulation of extracellular matrix and aberrant re-epithelialization of fibrotic lung parenchyma. Analysis of collagen VII mRNA and protein in IPF distal lungs demonstrated elevated levels compared with normal lungs. To investigate its cellular source and spatial distribution in lung tissue, immunohistochemistry, RNAscope in situ hybridization, and cell culture experiments in combination with analysis of public transcriptomic data sets were performed. In IPF lungs, collagen VII was abundant in pathologic remodeled airways and honeycomb cysts, associated with increased basal cell populations. In contrast, in the control lungs, collagen VII was mainly localized in larger airways. RNA sequencing data revealed that epithelial basal cells and keratin 5-/keratin 17+ aberrant basaloid cells are the primary sources of COL7A1 expression. Furthermore, COL7A1 expression was found in mesenchymal subsets, and both collagen VII mRNA and protein were observed in fibroblast foci, another histopathologic feature of IPF. In vitro, COL7A1 expression was found to be increased in normal human lung fibroblasts treated with transforming growth factor-β1. These findings suggest that collagen VII could be involved in the process of abnormal re-epithelialization in lung fibrosis.
Collapse
Affiliation(s)
- Barbora Svobodová
- Lung Biology, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| | - Anna Löfdahl
- Lung Biology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Måns Kadefors
- Lung Biology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Salad Mohamed Ali
- Lung Biology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Oskar Rosmark
- Lung Biology, Department of Experimental Medical Science, Lund University, Lund, Sweden; Department of Clinical Chemistry and Pharmacology, Office for Medical Services, Region Skåne, Lund, Sweden
| | - Pavan Prabhala
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Mattias Magnusson
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Hans Brunnström
- Department of Pathology, Regional Laboratories Region Skåne, Lund, Sweden; Division of Pathology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Sofia Lundin
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Göran Dellgren
- Department of Cardiothoracic Surgery and Transplant Institute, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Catharina Müller
- Lung Biology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Linda Elowsson
- Lung Biology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | |
Collapse
|
27
|
Gong Y, Yuan X, Jiao Q, Yu Z. Unveiling fine-scale spatial structures and amplifying gene expression signals in ultra-large ST slices with HERGAST. Nat Commun 2025; 16:3977. [PMID: 40295488 PMCID: PMC12037780 DOI: 10.1038/s41467-025-59139-w] [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: 09/11/2024] [Accepted: 04/08/2025] [Indexed: 04/30/2025] Open
Abstract
We propose HERGAST, a system for spatial structure identification and signal amplification in ultra-large-scale and ultra-high-resolution spatial transcriptomics data. To handle ultra-large spatial transcriptomics (ST) data, we consider the divide and conquer strategy and devise a Divide-Iterate-Conquer framework especially for spatial transcriptomics data analysis, which can also be adopted by other computational methods for extending to ultra-large-scale ST data analysis. To tackle the potential over-smoothing problem arising from data splitting, we construct a heterogeneous graph network to incorporate both local and global spatial relationships. In simulations, HERGAST consistently outperforms other methods across all settings with more than a 10% increase in average adjusted rand index (ARI). In real-world datasets, HERGAST's high-precision spatial clustering identifies SPP1+ macrophages intermingled within colorectal tumors, while the enhanced gene expression signals reveal unique spatial expression patterns of key genes in breast cancer.
Collapse
Affiliation(s)
- Yuqiao Gong
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Xin Yuan
- SJTU-Yale Joint Center for Biostatistics and Data Science Organization, Shanghai Jiao Tong University, Shanghai, China
- Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
- National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiong Jiao
- Department of Pathology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zhangsheng Yu
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
- SJTU-Yale Joint Center for Biostatistics and Data Science Organization, Shanghai Jiao Tong University, Shanghai, China.
- Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China.
- Clinical Research Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
28
|
Mwase C, Deng W, Kim HJ, Mitchel J, Phung TK, O'Sullivan MJ, Mathews JA, Crosby J, Turner C, Haber A, Park JA. Hic-5 transduces mechanical force that drives a vicious cycle of bronchoconstriction. RESEARCH SQUARE 2025:rs.3.rs-6498980. [PMID: 40343342 PMCID: PMC12060975 DOI: 10.21203/rs.3.rs-6498980/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Mechanical forces are essential for the function of key organs, including the bladder, bowel, heart, and lung<1/sup>. These organs often encounter excessive or dysregulated mechanical forces, which are associated with pathological conditions. However, the key regulators of mechanotransduction remain poorly understood. As an example of how excessive mechanical force imposed on airway epithelia could lead to mechanotransduction<2/sup> that alters the transcriptome<3/sup> and secretome<4/sup> and induces cell death<5/sup>, all of which contribute to disease progression<6,7/sup>, we used human airway epithelial cells in air-liquid interface culture to mimic bronchoconstriction. We show that Hic-5, a focal adhesion adaptor protein, functions as a key regulator of mechanoresponses in the airway. Hic-5 expression is significantly induced in airway basal cells following mechanical compression or bronchoconstriction. Hic-5 knockdown using antisense oligonucleotides protects against stress fiber formation and abolishes approximately 70% of transcripts differentially regulated by mechanical compression. Moreover, Hic-5 deficiency attenuates secretion of ET-1, a potent bronchoconstrictor. Our data show that during an asthma exacerbation, Hic-5 reinforces a vicious cycle of bronchoconstriction through the secretion of ET-1. We establish Hic-5 as a critical link between mechanical stress and epithelial activation in human disease, implicating dysregulated mechanical forces as active drivers of disease progression with therapeutic relevance.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Adam Haber
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA 02215, USA
| | | |
Collapse
|
29
|
Yamamoto Y, Shirai Y, Sonehara K, Namba S, Ojima T, Yamamoto K, Edahiro R, Suzuki K, Kanai A, Oda Y, Suzuki Y, Morisaki T, Narita A, Takeda Y, Tamiya G, Yamamoto M, Matsuda K, Kumanogoh A, Yamauchi T, Kadowaki T, Okada Y. Dissecting cross-population polygenic heterogeneity across respiratory and cardiometabolic diseases. Nat Commun 2025; 16:3765. [PMID: 40295474 PMCID: PMC12037804 DOI: 10.1038/s41467-025-58149-y] [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: 01/28/2024] [Accepted: 03/11/2025] [Indexed: 04/30/2025] Open
Abstract
Biological mechanisms underlying multimorbidity remain elusive. To dissect the polygenic heterogeneity of multimorbidity in twelve complex traits across populations, we leveraged biobank resources of genome-wide association studies (GWAS) for 232,987 East Asian individuals (the 1st and 2nd cohorts of BioBank Japan) and 751,051 European individuals (UK Biobank and FinnGen). Cross-trait analyses of respiratory and cardiometabolic diseases, rheumatoid arthritis, and smoking identified negative genetic correlations between respiratory and cardiometabolic diseases in East Asian individuals, opposite from the positive associations in European individuals. Associating genome-wide polygenic risk scores (PRS) with 325 blood metabolome and 2917 proteome biomarkers supported the negative cross-trait genetic correlations in East Asian individuals. Bayesian pathway PRS analysis revealed a negative association between asthma and dyslipidemia in a gene set of peroxisome proliferator-activated receptors. The pathway suggested heterogeneity of cell type specificity in the enrichment analysis of the lung single-cell RNA-sequencing dataset. Our study highlights the heterogeneous pleiotropy of immunometabolic dysfunction in multimorbidity.
Collapse
Affiliation(s)
- Yuji Yamamoto
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yuya Shirai
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Kyuto Sonehara
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shinichi Namba
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takafumi Ojima
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Japan
- Center for Advanced Intelligence Project, RIKEN, Tokyo, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Kenichi Yamamoto
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Ryuya Edahiro
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Ken Suzuki
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akinori Kanai
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Yoshiya Oda
- Department of Lipidomics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Japan
| | - Takayuki Morisaki
- Division of Molecular Pathology, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Laboratory of Clinical Genome Sequencing, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Akira Narita
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Yoshito Takeda
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Gen Tamiya
- Graduate School of Medicine, Tohoku University, Sendai, Japan
- Center for Advanced Intelligence Project, RIKEN, Tokyo, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Masayuki Yamamoto
- Graduate School of Medicine, Tohoku University, Sendai, Japan
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Koichi Matsuda
- Laboratory of Clinical Genome Sequencing, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Immunopathology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Japan
- Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Suita, Japan
- Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
- Center for Advanced Modalities and DDS (CAMaD), Osaka University, Suita, Japan
| | - Toshimasa Yamauchi
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
- Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan.
| | - Takashi Kadowaki
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
- Toranomon Hospital, Tokyo, Japan.
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan.
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan.
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
- Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Suita, Japan.
- Center for Advanced Modalities and DDS (CAMaD), Osaka University, Suita, Japan.
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Suita, Japan.
| |
Collapse
|
30
|
Liu G, Shi Y, Huang H, Xiao N, Liu C, Zhao H, Xing Y, Cai L. FPCAM: A Weighted Dictionary-Driven Model for Single-Cell Annotation in Pulmonary Fibrosis. BIOLOGY 2025; 14:479. [PMID: 40427668 PMCID: PMC12108865 DOI: 10.3390/biology14050479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2025] [Revised: 04/23/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025]
Abstract
The groundbreaking development of scRNA-seq has significantly improved cellular resolution. However, accurate cell-type annotation remains a major challenge. Existing annotation tools are often limited by their reliance on reference datasets, the heterogeneity of marker genes, and subjective biases introduced through manual intervention, all of which impact annotation accuracy and reliability. To address these limitations, we developed FPCAM, a fully automated pulmonary fibrosis cell-type annotation model. Built on the R Shiny platform, FPCAM utilizes a matrix of up-regulated marker genes and a manually curated gene-cell association dictionary specific to pulmonary fibrosis. It achieves accurate and efficient cell-type annotation through similarity matrix construction and optimized matching algorithms. To evaluate its performance, we compared FPCAM with state-of-the-art annotation models, including SCSA, SingleR, and SciBet. The results showed that FPCAM and SCSA both achieved an accuracy of 89.7%, outperforming SingleR and SciBet. Furthermore, FPCAM demonstrated high accuracy in annotating the external validation dataset GSE135893, successfully identifying multiple cell subtypes. In summary, FPCAM provides an efficient, flexible, and accurate solution for cell-type identification and serves as a powerful tool for scRNA-seq research in pulmonary fibrosis and other related diseases.
Collapse
Affiliation(s)
- Guojun Liu
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou 014000, China; (G.L.)
- Inner Mongolia Key Laboratory of Life Health and Bioinformatics, Inner Mongolia University of Science and Technology, Baotou 014000, China
| | - Yan Shi
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou 014000, China; (G.L.)
| | - Hongxu Huang
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou 014000, China; (G.L.)
| | - Ningkun Xiao
- Department of Immunochemistry, Institution of Chemical Engineering, Ural Federal University, Yekaterinburg 620000, Russia
| | - Chuncheng Liu
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou 014000, China; (G.L.)
- Inner Mongolia Key Laboratory of Life Health and Bioinformatics, Inner Mongolia University of Science and Technology, Baotou 014000, China
| | - Hongyu Zhao
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou 014000, China; (G.L.)
- Inner Mongolia Key Laboratory of Life Health and Bioinformatics, Inner Mongolia University of Science and Technology, Baotou 014000, China
| | - Yongqiang Xing
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou 014000, China; (G.L.)
- Inner Mongolia Key Laboratory of Life Health and Bioinformatics, Inner Mongolia University of Science and Technology, Baotou 014000, China
| | - Lu Cai
- School of Life Science and Technology, Inner Mongolia University of Science and Technology, Baotou 014000, China; (G.L.)
- Inner Mongolia Key Laboratory of Life Health and Bioinformatics, Inner Mongolia University of Science and Technology, Baotou 014000, China
| |
Collapse
|
31
|
Lee-Ferris RE, Okuda K, Galiger JR, Schworer SA, Rogers TD, Dang H, Gilmore R, Edwards C, Crisp G, Nakano S, Cawley AM, Pickles RJ, Gallant SC, Crisci E, Rivier L, Hagood JS, O’Neal WK, Baric RS, Grubb BR, Boucher RC, Randell SH. Prolonged airway explant culture enables study of health, disease, and viral pathogenesis. SCIENCE ADVANCES 2025; 11:eadp0451. [PMID: 40279421 PMCID: PMC12024639 DOI: 10.1126/sciadv.adp0451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 03/21/2025] [Indexed: 04/27/2025]
Abstract
In vitro models play a major role in studying airway physiology and disease. However, the native lung's complex tissue architecture and nonepithelial cell lineages are not preserved in these models. Ex vivo tissue models could overcome in vitro limitations, but methods for long-term maintenance of ex vivo tissue have not been established. Here, we describe methods to culture human large airway explants, small airway explants, and precision-cut lung slices for at least 14 days. Human airway explants recapitulate genotype-specific electrophysiology; characteristic epithelial, endothelial, stromal, and immune cell populations; and model viral infection after 14 days in culture. These methods also maintain mouse, rabbit, and pig tracheal explants. Notably, intact airway tissue can be cryopreserved, thawed, and used to generate viable explants with recovery of function 14 days postthaw. These studies highlight the broad applications of airway tissue explants and their use as translational intermediates between in vitro and in vivo studies.
Collapse
Affiliation(s)
- Rhianna E. Lee-Ferris
- Marsico Lung Institute/CF Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kenichi Okuda
- Marsico Lung Institute/CF Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jacob R. Galiger
- Marsico Lung Institute/CF Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Stephen A. Schworer
- Marsico Lung Institute/CF Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Troy D. Rogers
- Marsico Lung Institute/CF Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hong Dang
- Marsico Lung Institute/CF Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Rodney Gilmore
- Marsico Lung Institute/CF Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Caitlin Edwards
- Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gillian Crisp
- Marsico Lung Institute/CF Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Satoko Nakano
- Marsico Lung Institute/CF Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Anne M. Cawley
- Marsico Lung Institute/CF Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Raymond J. Pickles
- Marsico Lung Institute/CF Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Samuel C. Gallant
- Marsico Lung Institute/CF Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Elisa Crisci
- College of Veterinary Medicine, Department of Population Health and Pathobiology, North Carolina State University, Raleigh, NC, USA
| | - Lauraine Rivier
- Marsico Lung Institute/CF Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - James S. Hagood
- Marsico Lung Institute/CF Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Pediatric Pulmonology and Program for Rare and Interstitial Lung Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Wanda K. O’Neal
- Marsico Lung Institute/CF Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ralph S. Baric
- Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Barbara R. Grubb
- Marsico Lung Institute/CF Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Richard C. Boucher
- Marsico Lung Institute/CF Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Scott H. Randell
- Marsico Lung Institute/CF Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
32
|
Lee GY, Schaunaman N, Nouri HR, Kraft M, Chu HW. Comprehensive single-cell RNA-sequencing study of Tollip deficiency effect in IL-13-stimulated human airway epithelial cells. BMC Res Notes 2025; 18:194. [PMID: 40269942 PMCID: PMC12020103 DOI: 10.1186/s13104-025-07255-7] [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: 12/28/2024] [Accepted: 04/11/2025] [Indexed: 04/25/2025] Open
Abstract
OBJECTIVE Toll-interacting protein (Tollip) suppresses excessive pro-inflammatory signaling, but its function in airway epithelial responses to IL-13, a key mediator in allergic diseases, remains unclear. This study investigates Tollip knockdown (TKD) effects in primary human airway epithelial cells using single-cell RNA sequencing, providing the first single-cell analysis of TKD and the first exploring its interaction with IL-13. RESULTS IL-13 treatment upregulated key genes, including SPDEF, MUC5AC, POSTN, ALOX15, and CCL26, confirming IL-13's effects and validating our methods. IL-13 reduced TNF-α signaling and epithelial-mesenchymal transition in certain cell types, suggesting a dual role in promoting type 2 inflammation while suppressing Th1-driven inflammation. Tollip deficiency alone significantly amplified TNF-α signaling and inflammatory pathways in goblet, club, and suprabasal cells. Comparisons between TKDIL13 vs IL13 and TKD vs CTR revealed that IL-13 does not substantially alter Tollip deficiency response in most cell types, reinforcing findings in TKD vs CTR. Tollip deficiency alters the response to IL-13 in a cell-type-specific manner, strongly downregulating TNF-α signaling in goblet cells but only weakly in basal and club cells. Tollip deficiency enhances IL-13's suppression of Th1 inflammatory responses in goblet cells. These novel insights in Tollip-IL-13 interactions offer potential therapeutic targets for asthma and related diseases.
Collapse
Affiliation(s)
| | | | | | - Monica Kraft
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hong Wei Chu
- Department of Medicine, National Jewish Health, Denver, CO, USA.
| |
Collapse
|
33
|
Kalfon J, Samaran J, Peyré G, Cantini L. scPRINT: pre-training on 50 million cells allows robust gene network predictions. Nat Commun 2025; 16:3607. [PMID: 40240364 PMCID: PMC12003772 DOI: 10.1038/s41467-025-58699-1] [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: 08/20/2024] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
A cell is governed by the interaction of myriads of macromolecules. Inferring such a network of interactions has remained an elusive milestone in cellular biology. Building on recent advances in large foundation models and their ability to learn without supervision, we present scPRINT, a large cell model for the inference of gene networks pre-trained on more than 50 million cells from the cellxgene database. Using innovative pretraining tasks and model architecture, scPRINT pushes large transformer models towards more interpretability and usability when uncovering the complex biology of the cell. Based on our atlas-level benchmarks, scPRINT demonstrates superior performance in gene network inference to the state of the art, as well as competitive zero-shot abilities in denoising, batch effect correction, and cell label prediction. On an atlas of benign prostatic hyperplasia, scPRINT highlights the profound connections between ion exchange, senescence, and chronic inflammation.
Collapse
Affiliation(s)
- Jérémie Kalfon
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Machine Learning for Integrative Genomics group, F-75015, Paris, France
| | - Jules Samaran
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Machine Learning for Integrative Genomics group, F-75015, Paris, France
| | - Gabriel Peyré
- CNRS and DMA de l'Ecole Normale Supérieure, CNRS, Ecole Normale Supérieure, Université PSL, 75005, Paris, France
| | - Laura Cantini
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Machine Learning for Integrative Genomics group, F-75015, Paris, France.
| |
Collapse
|
34
|
Gerner-Mauro KN, Vila Ellis L, Wang G, Nayak R, Lwigale PY, Poché RA, Chen J. Morphogenic, molecular and cellular adaptations for unidirectional airflow in the chicken lung. Development 2025; 152:dev204346. [PMID: 40177910 PMCID: PMC12070062 DOI: 10.1242/dev.204346] [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: 08/22/2024] [Accepted: 03/27/2025] [Indexed: 04/05/2025]
Abstract
Unidirectional airflow in the avian lung enables gas exchange during both inhalation and exhalation. The underlying developmental process and how it deviates from that of the bidirectional mammalian lung are poorly understood. Sampling key developmental stages with multiscale 3D imaging and single-cell transcriptomics, we delineate morphogenic, molecular and cellular features that accommodate the unidirectional airflow in the chicken lung. Primary termini of hyper-elongated branches undergo proximal-short and distal-long fusions, forming parabronchi for air conduction. Through the parabronchial smooth muscle, neoform termini extend radially to form gas-exchanging alveoli. Supporting this radial alveologenesis, branch stalks halt their proximalization, defined by SOX9-SOX2 transition, and become SOX9low parabronchi. Primary and secondary vascular plexi interface with primary and neoform termini, respectively. Single-cell and Stereo-seq spatial transcriptomics reveal a third, chicken-specific alveolar cell type expressing KRT14, hereby named luminal cells. Luminal, alveolar type 2 and alveolar type 1 cells sequentially occupy concentric zones radiating from the parabronchial lumen. Our study explores the evolutionary space of lung diversification and lays the foundation for functional analysis of species-specific genetic determinants.
Collapse
Affiliation(s)
- Kamryn N. Gerner-Mauro
- Department of Pulmonary Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Development, Disease Models, and Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lisandra Vila Ellis
- Department of Pulmonary Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Cell & Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Guolun Wang
- Department of Pediatrics, Perinatal Institute Division of Pulmonary Biology, University of Cincinnati and Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Richa Nayak
- Department of Pulmonary Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Cancer Biology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Peter Y. Lwigale
- Department of BioSciences, Rice University, Houston, TX 77005, USA
| | - Ross A. Poché
- Development, Disease Models, and Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jichao Chen
- Department of Pulmonary Medicine, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Pediatrics, Perinatal Institute Division of Pulmonary Biology, University of Cincinnati and Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| |
Collapse
|
35
|
Crnkovic S, Thekkekara Puthenparampil H, Mulch S, Biasin V, Radic N, Wilhelm J, Bartkuhn M, Bonyadi Rad E, Wawrzen A, Matzer I, Mitra A, Leib RD, Nagy BM, Sahu-Osen A, Valzano F, Bordag N, Evermann M, Hoetzenecker K, Olschewski A, Ljubojevic-Holzer S, Wygrecka M, Stenmark K, Marsh LM, de Jesus Perez V, Kwapiszewska G. Adventitial fibroblasts direct smooth muscle cell-state transition in pulmonary vascular disease. eLife 2025; 13:RP98558. [PMID: 40208251 PMCID: PMC11984959 DOI: 10.7554/elife.98558] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025] Open
Abstract
Background Pulmonary vascular remodeling is a progressive pathological process characterized by functional alterations within pulmonary artery smooth muscle cells (PASMCs) and adventitial fibroblasts (PAAFs). Mechanisms driving the transition to a diseased phenotype remain elusive. Methods We combined transcriptomic and proteomic profiling with phenotypic characterization of source-matched cells from healthy controls and individuals with idiopathic pulmonary arterial hypertension (IPAH). Bidirectional cellular crosstalk was examined using direct and indirect co-culture models, and phenotypic responses were assessed via transcriptome analysis. Results PASMC and PAAF undergo distinct phenotypic shifts during pulmonary vascular remodeling, with limited shared features, such as reduced mitochondrial content and hyperpolarization. IPAH-PASMC exhibit increased glycosaminoglycan production and downregulation of contractile machinery, while IPAH-PAAF display a hyperproliferative phenotype. We identified alterations in extracellular matrix components, including laminin and collagen, alongside pentraxin-3 and hepatocyte growth factor, as potential regulators of PASMC phenotypic transitions mediated by PAAF. Conclusions While PASMCs and PAAFs retain their core cellular identities, they acquire distinct disease-associated states. These findings provide new insights into the dynamic interplay of pulmonary vascular mesenchymal cells in disease pathogenesis. Funding This work was supported by Cardio-Pulmonary Institute EXC 2026 390649896 (GK) and Austrian Science Fund (FWF) grant I 4651-B (SC).
Collapse
Affiliation(s)
- Slaven Crnkovic
- Ludwig Boltzmann Institute for Lung Vascular ResearchGrazAustria
- Medical University of Graz, Lung Research ClusterGrazAustria
- Institute for Lung Health, Cardiopulmonary Institute, Member of the German Center for Lung Research, Justus-Liebig University GiessenGiessenGermany
| | | | - Shirin Mulch
- Institute for Lung Health, Cardiopulmonary Institute, Member of the German Center for Lung Research, Justus-Liebig University GiessenGiessenGermany
| | - Valentina Biasin
- Ludwig Boltzmann Institute for Lung Vascular ResearchGrazAustria
- Medical University of Graz, Lung Research ClusterGrazAustria
| | - Nemanja Radic
- Medical University of Graz, Lung Research ClusterGrazAustria
| | - Jochen Wilhelm
- Institute for Lung Health, Cardiopulmonary Institute, Member of the German Center for Lung Research, Justus-Liebig University GiessenGiessenGermany
| | - Marek Bartkuhn
- Institute for Lung Health, Cardiopulmonary Institute, Member of the German Center for Lung Research, Justus-Liebig University GiessenGiessenGermany
| | | | - Alicja Wawrzen
- Medical University of Graz, Lung Research ClusterGrazAustria
| | - Ingrid Matzer
- Medical University of Graz, Lung Research ClusterGrazAustria
| | - Ankita Mitra
- Department of Medicine, Stanford University School of MedicineStanfordUnited States
| | - Ryan D Leib
- Mass Spectrometry Laboratory, Stanford University School of MedicineStanfordUnited States
| | | | - Anita Sahu-Osen
- Ludwig Boltzmann Institute for Lung Vascular ResearchGrazAustria
| | | | - Natalie Bordag
- Ludwig Boltzmann Institute for Lung Vascular ResearchGrazAustria
- Medical University of Graz, Lung Research ClusterGrazAustria
| | | | | | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular ResearchGrazAustria
- Medical University of Graz, Lung Research ClusterGrazAustria
| | | | - Malgorzata Wygrecka
- Institute for Lung Health, Cardiopulmonary Institute, Member of the German Center for Lung Research, Justus-Liebig University GiessenGiessenGermany
| | - Kurt Stenmark
- Developmental Lung Biology and Cardiovascular Pulmonary Research Laboratories, University of ColoradoAuroraUnited States
| | - Leigh M Marsh
- Ludwig Boltzmann Institute for Lung Vascular ResearchGrazAustria
- Medical University of Graz, Lung Research ClusterGrazAustria
| | | | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular ResearchGrazAustria
- Medical University of Graz, Lung Research ClusterGrazAustria
- Institute for Lung Health, Cardiopulmonary Institute, Member of the German Center for Lung Research, Justus-Liebig University GiessenGiessenGermany
| |
Collapse
|
36
|
Yates J, Kraft A, Boeva V. Filtering cells with high mitochondrial content depletes viable metabolically altered malignant cell populations in cancer single-cell studies. Genome Biol 2025; 26:91. [PMID: 40205439 PMCID: PMC11983838 DOI: 10.1186/s13059-025-03559-w] [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: 11/05/2024] [Accepted: 03/25/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Single-cell transcriptomics has transformed our understanding of cellular diversity, yet noise from technical artifacts and low-quality cells can obscure key biological signals. A common practice is filtering out cells with a high percentage of mitochondrial RNA counts (pctMT), typically indicative of cell death. However, commonly used filtering thresholds, primarily derived from studies on healthy tissues, may be overly stringent for malignant cells, which often naturally exhibit higher baseline mitochondrial gene expression. RESULTS We examine nine public single-cell RNA-seq datasets from various cancers, including 441,445 cells from 134 patients, and public spatial transcriptomics data, assessing the viability of malignant cells with high pctMT. Our analysis reveals that malignant cells exhibit significantly higher pctMT than nonmalignant cells, without a notable increase in dissociation-induced stress scores. Malignant cells with high pctMT show metabolic dysregulation, including increased xenobiotic metabolism, relevant to therapeutic response. Analysis of pctMT in cancer cell lines further reveals links to drug resistance. We also observe associations between pctMT and malignant cell transcriptional heterogeneity, as well as patient clinical features. CONCLUSIONS This study provides insights into the functional characteristics of malignant cells with elevated pctMT, challenging current quality control practices in tumor single-cell RNA-seq analyses and offering potential improvements in data interpretation for future cancer studies.
Collapse
Affiliation(s)
- Josephine Yates
- Department of Computer Science, Institute for Machine Learning, ETH Zürich, Zurich, CH-8092, Switzerland
- ETH AI Center, Zurich, Switzerland
- Swiss Institute for Bioinformatics (SIB), Lausanne, Switzerland
| | - Agnieszka Kraft
- Department of Computer Science, Institute for Machine Learning, ETH Zürich, Zurich, CH-8092, Switzerland
- Swiss Institute for Bioinformatics (SIB), Lausanne, Switzerland
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Valentina Boeva
- Department of Computer Science, Institute for Machine Learning, ETH Zürich, Zurich, CH-8092, Switzerland.
- ETH AI Center, Zurich, Switzerland.
- Swiss Institute for Bioinformatics (SIB), Lausanne, Switzerland.
- Cochin Institute, Inserm U1016, CNRS UMR 8104, Paris Descartes University UMR-S1016, Paris, 75014, France.
| |
Collapse
|
37
|
Zappia L, Richter S, Ramírez-Suástegui C, Kfuri-Rubens R, Vornholz L, Wang W, Dietrich O, Frishberg A, Luecken MD, Theis FJ. Feature selection methods affect the performance of scRNA-seq data integration and querying. Nat Methods 2025; 22:834-844. [PMID: 40082610 PMCID: PMC11978513 DOI: 10.1038/s41592-025-02624-3] [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: 06/07/2023] [Accepted: 02/08/2025] [Indexed: 03/16/2025]
Abstract
The availability of single-cell transcriptomics has allowed the construction of reference cell atlases, but their usefulness depends on the quality of dataset integration and the ability to map new samples. Previous benchmarks have compared integration methods and suggest that feature selection improves performance but have not explored how best to select features. Here, we benchmark feature selection methods for single-cell RNA sequencing integration using metrics beyond batch correction and preservation of biological variation to assess query mapping, label transfer and the detection of unseen populations. We reinforce common practice by showing that highly variable feature selection is effective for producing high-quality integrations and provide further guidance on the effect of the number of features selected, batch-aware feature selection, lineage-specific feature selection and integration and the interaction between feature selection and integration models. These results are informative for analysts working on large-scale tissue atlases, using atlases or integrating their own data to tackle specific biological questions.
Collapse
Affiliation(s)
- Luke Zappia
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
- School of Computing, Information and Technology, Technical University of Munich, Munich, Germany
| | - Sabrina Richter
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
| | - Ciro Ramírez-Suástegui
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Raphael Kfuri-Rubens
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
- School of Medicine, Technical University of Munich, Munich, Germany
- Klinikum rechts der Isar, IIIrd Medical Department, Munich, Germany
| | - Larsen Vornholz
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
| | - Weixu Wang
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
| | - Oliver Dietrich
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
- Helmholtz Institute for RNA-based Infection Research, Helmholtz Centre for Infection Research, Würzburg, Germany
| | - Amit Frishberg
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
| | - Malte D Luecken
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany
- Institute of Lung Health & Immunity, Helmholtz Munich; Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Neuherberg, Germany.
- School of Computing, Information and Technology, Technical University of Munich, Munich, Germany.
- School of Life Sciences Weihenstephan, Technical University of Munich, Friesing, Germany.
| |
Collapse
|
38
|
Scott IC, Zuydam NV, Cann JA, Negri VA, Tsafou K, Killick H, Liu Z, McCrae C, Rees DG, England E, Guscott MA, Houslay K, McCormick D, Freeman A, Schofield D, Freeman A, Cohen ES, Thwaites R, Brohawn Z, Platt A, Openshaw PJM, Semple MG, Baillie JK, Wilkinson T. IL-33 is associated with alveolar dysfunction in patients with viral lower respiratory tract disease. Mucosal Immunol 2025; 18:312-325. [PMID: 39662674 PMCID: PMC11982439 DOI: 10.1016/j.mucimm.2024.12.001] [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: 09/12/2024] [Revised: 11/29/2024] [Accepted: 12/05/2024] [Indexed: 12/13/2024]
Abstract
Interleukin (IL)-33 is released following tissue damage, causing airway inflammation and remodelling via reduced IL-33 (IL-33red)/serum stimulation-2 (ST2) and oxidised IL-33 (IL-33ox)/receptor for advanced glycation end products (RAGE)/epidermal growth factor receptor (EGFR) pathways. This study aimed to identify associations of IL-33 with clinical outcomes and pathological mechanisms during viral lower respiratory tract disease (LRTD). Ultra-sensitive immunoassays were developed to measure IL-33red, IL-33ox and IL-33/sST2 complexes in samples from patients hospitalised with COVID-19. Immunohistochemistry and multiomics were used to characterise lung samples. Elevated IL-33 in the airway and IL-33/sST2 complex in the circulation correlated with poor clinical outcomes (death, need for intensive care or mechanical ventilation). IL-33 was localised to airway epithelial and endothelial barriers, whereas IL1RL1 was expressed on aerocytes, alveolar endothelial cells specialised for gaseous exchange. IL-33 increased expression of mediators of neutrophilic inflammation, immune cell infiltration, interferon signalling and coagulation in endothelial cell cultures. Endothelial IL-33 signatures were strongly related with signatures associated with viral LRTD. Increased IL-33 release following respiratory viral infections is associated with poor clinical outcomes and might contribute to alveolar dysfunction. Although this does not show a causal relationship with disease, these results provide a rationale to evaluate pathological roles for IL-33 in viral LRTD.
Collapse
Affiliation(s)
- Ian C Scott
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK.
| | - Natalie van Zuydam
- Discovery Sciences, Research and Early Development, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Jennifer A Cann
- Clinical Pharmacology and Safety Sciences, Research and Early Development, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Victor Augusti Negri
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Kalliopi Tsafou
- Discovery Sciences, Research and Early Development, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Helen Killick
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Zhi Liu
- Translational Sciences and Experimental Medicine, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Christopher McCrae
- Translational Sciences and Experimental Medicine, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - D Gareth Rees
- Biologics Engineering, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Elizabeth England
- Biologics Engineering, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Molly A Guscott
- Bioscience Asthma and Skin Immunity, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Kirsty Houslay
- Bioscience Asthma and Skin Immunity, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Dominique McCormick
- Department of Clinical Infection, Microbiology, and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Anna Freeman
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Darren Schofield
- Biologics Engineering, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Adrian Freeman
- Discovery Sciences, Research and Early Development, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - E Suzanne Cohen
- Bioscience Asthma and Skin Immunity, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Ryan Thwaites
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Zach Brohawn
- Translational Sciences and Experimental Medicine, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Adam Platt
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | | | - Malcolm G Semple
- Department of Clinical Infection, Microbiology, and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - J Kenneth Baillie
- Baillie Gifford Pandemic Science Hub, University of Edinburgh, Edinburgh, UK
| | - Tom Wilkinson
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| |
Collapse
|
39
|
Zeng PYF, Lin RJ, Fung K, Khan H, Cecchini MJ, Woo E, Hu A, Anderson J, MacInnis P, Jarycki L, Karimi A, Ying S, Al Jawhri M, Lin S, Shaikh M, Pan H, Coburn B, Mymryk JS, Inculet R, Barrett JW, Nichols AC. Cellular blueprint of healthy and diseased human epiglottis and subglottis-a study of the Canadian Airways Research (CARE) group. EBioMedicine 2025; 114:105631. [PMID: 40048848 PMCID: PMC11929080 DOI: 10.1016/j.ebiom.2025.105631] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 12/02/2024] [Accepted: 02/20/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND The larynx consists of the supraglottis, glottis, and subglottis and each differ in tissue composition, lymphatic drainage, ability to counter infections, and response to injuries. However, the cellular mechanisms driving laryngeal homoeostasis remain largely unexplored. As a result, understanding disease pathogenesis within the larynx including idiopathic subglottic stenosis (iSGS) and intubation-related traumatic stenosis has been challenging. Here, we sought to characterise the cellular processes governing laryngeal health and disease. METHODS As part of the prospective Canadian Airways Research (CARE) iSGS study, we characterised 122,004 high-quality transcriptomes using single nucleus RNA-sequencing to profile 11 human epiglottis and 17 human subglottis biopsies across three different conditions: control, iSGS, and intubation-related traumatic stenosis to define cell populations and pathways associated with disease. We validated our results using cohort-level bulk transcriptomics using 114 human epiglottis and 121 human subglottis. FINDINGS We defined the single-cell taxonomy of the human subglottis and epiglottis using single-nucleus sequencing in both healthy and disease states. Mechanistically, we discovered the presence of unique epithelial and fibroblast progenitor subsets within the control subglottis but not within the anatomically adjacent epiglottis. The uncontrolled proliferation of these cellular subsets exhibited skewed sex hormone signalling and orchestrated a fibro-inflammatory cascade. We leveraged cohort-level bulk transcriptomics to define hallmarks of iSGS associated with disease covariates and introduced the first biomarker associated with recurrent relapse. Longitudinal sampling demonstrated that the subglottic microenvironment in patients with iSGS is changing dynamically with and without therapeutic intervention. INTERPRETATION Together, our data refines our understanding of laryngeal biology, nominates candidate compounds for iSGS treatment, and serves as a transformative platform for future clinical investigations to further precision laryngology. FUNDING This study was funded by a grant from the American Laryngology Association (#1082), an Academic Medical Organisation of Southwestern Ontario innovation fund grant (INN21-016), grant support from the Departments of Otolaryngology-Head and Neck Surgery at University of Toronto, Canada and Western University, Canada. ACN was supported by the Wolfe Surgical Research Professorship in the Biology of Head and Neck Cancers Fund. PYFZ was supported by a Vanier Canada Graduate Scholarship and PSI Foundation fellowship.
Collapse
Affiliation(s)
- Peter Y F Zeng
- Department of Otolaryngology - Head & Neck Surgery, Western University, London, Ontario, Canada; Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada.
| | - R Jun Lin
- Department of Otolaryngology - Head & Neck Surgery, Temerty Faculty of Medicine, University of Toronto, Unity Health Toronto, St. Michael's Hospital, Toronto, Ontario, Canada.
| | - Kevin Fung
- Department of Otolaryngology - Head & Neck Surgery, Western University, London, Ontario, Canada
| | - Halema Khan
- Department of Otolaryngology - Head & Neck Surgery, Western University, London, Ontario, Canada
| | - Matthew J Cecchini
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Elissa Woo
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Amanda Hu
- Division of Otolaryngology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jennifer Anderson
- Department of Otolaryngology - Head & Neck Surgery, Temerty Faculty of Medicine, University of Toronto, Unity Health Toronto, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Patrick MacInnis
- Department of Otolaryngology - Head & Neck Surgery, Temerty Faculty of Medicine, University of Toronto, Unity Health Toronto, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Laura Jarycki
- Department of Otolaryngology - Head & Neck Surgery, Western University, London, Ontario, Canada
| | - Amir Karimi
- Department of Otolaryngology - Head & Neck Surgery, Western University, London, Ontario, Canada; Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Shengjie Ying
- Department of Otolaryngology - Head & Neck Surgery, Western University, London, Ontario, Canada; Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - MohdWessam Al Jawhri
- Department of Otolaryngology - Head & Neck Surgery, Western University, London, Ontario, Canada; Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Sherman Lin
- Department of Otolaryngology - Head & Neck Surgery, Western University, London, Ontario, Canada; Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Mushfiq Shaikh
- Department of Otolaryngology - Head & Neck Surgery, Western University, London, Ontario, Canada; Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Harrison Pan
- Department of Otolaryngology - Head & Neck Surgery, Western University, London, Ontario, Canada; Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Bryan Coburn
- Department of Medicine, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Joe S Mymryk
- Department of Otolaryngology - Head & Neck Surgery, Western University, London, Ontario, Canada; Department of Oncology, Western University, London, Ontario, Canada; Department of Microbiology & Immunology, Western University, London, Ontario, Canada
| | - Richard Inculet
- Division of Thoracic Surgery, Western University, London, Ontario, Canada
| | - John W Barrett
- Department of Otolaryngology - Head & Neck Surgery, Western University, London, Ontario, Canada
| | - Anthony C Nichols
- Department of Otolaryngology - Head & Neck Surgery, Western University, London, Ontario, Canada; Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada; Department of Oncology, Western University, London, Ontario, Canada.
| |
Collapse
|
40
|
Khodaee F, Zandie R, Edelman ER. Multimodal learning for mapping genotype-phenotype dynamics. NATURE COMPUTATIONAL SCIENCE 2025; 5:333-344. [PMID: 39875699 DOI: 10.1038/s43588-024-00765-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 12/20/2024] [Indexed: 01/30/2025]
Abstract
How complex phenotypes emerge from intricate gene expression patterns is a fundamental question in biology. Integrating high-content genotyping approaches such as single-cell RNA sequencing and advanced learning methods such as language models offers an opportunity for dissecting this complex relationship. Here we present a computational integrated genetics framework designed to analyze and interpret the high-dimensional landscape of genotypes and their associated phenotypes simultaneously. We applied this approach to develop a multimodal foundation model to explore the genotype-phenotype relationship manifold for human transcriptomics at the cellular level. Analyzing this joint manifold showed a refined resolution of cellular heterogeneity, uncovered potential cross-tissue biomarkers and provided contextualized embeddings to investigate the polyfunctionality of genes shown for the von Willebrand factor (VWF) gene in endothelial cells. Overall, this study advances our understanding of the dynamic interplay between gene expression and phenotypic manifestation and demonstrates the potential of integrated genetics in uncovering new dimensions of cellular function and complexity.
Collapse
Affiliation(s)
- Farhan Khodaee
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Rohola Zandie
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Elazer R Edelman
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Medicine (Cardiovascular Medicine), Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
41
|
Umeda Y, Izawa T, Kazama K, Arai S, Kamiie J, Nakamura S, Hano K, Takasu M, Hirata A, Rittinghausen S, Yamano S. Comparative anatomy of respiratory bronchioles and lobular structures in mammals. J Toxicol Pathol 2025; 38:113-129. [PMID: 40190622 PMCID: PMC11966126 DOI: 10.1293/tox.2024-0071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 12/19/2024] [Indexed: 04/09/2025] Open
Abstract
Rodents are widely used to study the toxicity of chemicals; however, differences between species indicate that the results from rodents are not always directly transferable to humans. The health of workers exposed to various chemicals and particulates at high doses or for long periods is at risk. Respiratory bronchioles and lobular structures, which are demarcated by interlobular septa, are key sites for occupational lung diseases such as pneumoconiosis; however, these structures vary among animal species. Understanding these differences is crucial for studying the pathology of human occupational lung diseases. However, there is a lack of reviews focusing on these structures in different species. This review explores the lung anatomy of various mammals and its functional importance in disease to connect animal studies with human occupational lung diseases. Our results indicate that artiodactyls, especially small pig breeds and goats, are ideal for research because their respiratory bronchioles and lobular structures are similar to those of humans. This review aims to enhance the use of experimental animal data and improve our understanding of human occupational lung diseases, thereby facilitating early disease detection, treatment, and prevention.
Collapse
Affiliation(s)
- Yumi Umeda
- National Institute of Occupational Safety and Health, Japan,
Organization of Occupational Health and Safety, 2-26-1 Muraoka-higashi, Fujisawa, Kanagawa
251-0015, Japan
| | - Takeshi Izawa
- Laboratory of Veterinary Pathology, Osaka Metropolitan
University Graduate School of Veterinary Science, 1-58 Rinku-Orai-Kita, Izumisano, Osaka
598-8531, Japan
| | - Kei Kazama
- Department of Veterinary Medicine, Azabu University, School
of Veterinary Medicine, 17-71 Fuchinobe 1-chome, Chuo-ku, Sagamihara 252-5201, Japan
| | - Sachiko Arai
- Department of Veterinary Medicine, Azabu University, School
of Veterinary Medicine, 17-71 Fuchinobe 1-chome, Chuo-ku, Sagamihara 252-5201, Japan
| | - Junichi Kamiie
- Laboratory of Veterinary Pathology, School of Veterinary
Medicine, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara, Kanagawa 252-5201,
Japan
| | - Shinichiro Nakamura
- Laboratory of Laboratory Animal Science, School of
Veterinary Medicine, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara, Kanagawa
252-5201, Japan
| | - Kazuki Hano
- Gifu University Institute for Advanced Study, Gifu
University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Masaki Takasu
- Gifu University Institute for Advanced Study, Gifu
University, 1-1 Yanagido, Gifu 501-1193, Japan
- Center for One Medicine Innovative Translational Research
(COMIT), Gifu University, 1-1 Yanagido, Gifu, 501-1193, Gifu, 501-1193, Japan
| | - Akihiro Hirata
- Center for One Medicine Innovative Translational Research
(COMIT), Gifu University, 1-1 Yanagido, Gifu, 501-1193, Gifu, 501-1193, Japan
- Laboratory of Veterinary Pathology, Joint Department of
Veterinary Medicine, Faculty of Applied Biological Sciences, Gifu University, 1-1
Yanagido, Gifu, 501-1193, Japan
| | - Susanne Rittinghausen
- Fraunhofer Institute for Toxicology and Experimental
Medicine (ITEM), Nikolai-Fuchs-Strasse 1, 30625, Hannover, Germany
| | - Shotaro Yamano
- National Institute of Occupational Safety and Health, Japan,
Organization of Occupational Health and Safety, 2-26-1 Muraoka-higashi, Fujisawa, Kanagawa
251-0015, Japan
| |
Collapse
|
42
|
Bian H, Chen Y, Wei L, Zhang X. uHAF: a unified hierarchical annotation framework for cell type standardization and harmonization. Bioinformatics 2025; 41:btaf149. [PMID: 40172934 PMCID: PMC12002906 DOI: 10.1093/bioinformatics/btaf149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/14/2025] [Accepted: 04/01/2025] [Indexed: 04/04/2025] Open
Abstract
SUMMARY In single-cell transcriptomics, inconsistent cell type annotations due to varied naming conventions and hierarchical granularity impede data integration, machine learning applications, and meaningful evaluations. To address this challenge, we developed the unified Hierarchical Annotation Framework (uHAF), which includes organ-specific hierarchical cell type trees (uHAF-T) and a mapping tool (uHAF-Agent) based on large language models. uHAF-T provides standardized hierarchical references for 38 organs, allowing for consistent label unification and analysis at different levels of granularity. uHAF-Agent leverages GPT-4 to accurately map diverse and informal cell type labels onto uHAF-T nodes, streamlining the harmonization process. By simplifying label unification, uHAF enhances data integration, supports machine learning applications, and enables biologically meaningful evaluations of annotation methods. Our framework serves as an essential resource for standardizing cell type annotations and fostering collaborative refinement in the single-cell research community. AVAILABILITY AND IMPLEMENTATION uHAF is publicly available at: https://uhaf.unifiedcellatlas.org and https://github.com/SuperBianC/uhaf.
Collapse
Affiliation(s)
- Haiyang Bian
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division of BNRIST, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Yinxin Chen
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division of BNRIST, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Lei Wei
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division of BNRIST, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Xuegong Zhang
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division of BNRIST, Department of Automation, Tsinghua University, Beijing 100084, China
- Center for Synthetic and Systems Biology, School of Life Sciences and School of Medicine, Tsinghua University, Beijing 100084, China
| |
Collapse
|
43
|
Farhat A, Radhouani M, Deckert F, Zahalka S, Pimenov L, Fokina A, Hakobyan A, Oberndorfer F, Brösamlen J, Hladik A, Lakovits K, Meng F, Quattrone F, Boon L, Vesely C, Starkl P, Boucheron N, Menche J, van der Veeken J, Ellmeier W, Gorki AD, Campbell C, Gawish R, Knapp S. An aging bone marrow exacerbates lung fibrosis by fueling profibrotic macrophage persistence. Sci Immunol 2025; 10:eadk5041. [PMID: 40153488 DOI: 10.1126/sciimmunol.adk5041] [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: 08/29/2023] [Revised: 08/27/2024] [Accepted: 02/19/2025] [Indexed: 03/30/2025]
Abstract
Pulmonary fibrosis is an incurable disease that manifests with advanced age. Yet, how hematopoietic aging influences immune responses and fibrosis progression remains unclear. Using heterochronic bone marrow transplant mouse models, we found that an aged bone marrow exacerbates lung fibrosis irrespective of lung tissue age. Upon lung injury, there was an increased accumulation of monocyte-derived alveolar macrophages (Mo-AMs) driven by cell-intrinsic hematopoietic aging. These Mo-AMs exhibited an enhanced profibrotic profile and stalled maturation into a homeostatic, tissue-resident phenotype. This delay was shaped by cell-extrinsic environmental signals such as reduced pulmonary interleukin-10 (IL-10), perpetuating a profibrotic macrophage state. We identified regulatory T cells (Tregs) as critical providers of IL-10 upon lung injury that promote Mo-AM maturation and attenuate fibrosis progression. Our study highlights the impact of an aging bone marrow on lung immune regulation and identifies Treg-mediated IL-10 signaling as a promising target to mitigate fibrosis and promote tissue repair.
Collapse
Affiliation(s)
- Asma Farhat
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, CeMM, Vienna, Austria
| | - Mariem Radhouani
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, CeMM, Vienna, Austria
| | - Florian Deckert
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, CeMM, Vienna, Austria
| | - Sophie Zahalka
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, CeMM, Vienna, Austria
| | - Lisabeth Pimenov
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Alina Fokina
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Anna Hakobyan
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, CeMM, Vienna, Austria
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Department of Structural and Computational Biology, University of Vienna, Vienna, Austria
| | | | - Jessica Brösamlen
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Anastasiya Hladik
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Karin Lakovits
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Fanzhe Meng
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Federica Quattrone
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, CeMM, Vienna, Austria
| | | | - Cornelia Vesely
- Center for Anatomy and Cell Biology, Medical University of Vienna, Vienna, Austria
| | - Philipp Starkl
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Nicole Boucheron
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Jörg Menche
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, CeMM, Vienna, Austria
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Department of Structural and Computational Biology, University of Vienna, Vienna, Austria
- Faculty of Mathematics, University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Network Medicine at the University of Vienna, Vienna, Austria
| | | | - Wilfried Ellmeier
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Anna-Dorothea Gorki
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Clarissa Campbell
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, CeMM, Vienna, Austria
| | - Riem Gawish
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Sylvia Knapp
- Research Division of Infection Biology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Vienna, Austria
| |
Collapse
|
44
|
Yu Q, Li YY, Chen Y. scMalignantFinder distinguishes malignant cells in single-cell and spatial transcriptomics by leveraging cancer signatures. Commun Biol 2025; 8:504. [PMID: 40148533 PMCID: PMC11950360 DOI: 10.1038/s42003-025-07942-y] [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: 06/30/2024] [Accepted: 03/17/2025] [Indexed: 03/29/2025] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) is a powerful tool for characterizing tumor heterogeneity, yet accurately identifying malignant cells remains challenging. Here, we propose scMalignantFinder, a machine learning tool specifically designed to distinguish malignant cells from their normal counterparts using a data- and knowledge-driven strategy. To develop the tool, multiple cancer datasets were collected, and the initially annotated malignant cells were calibrated using nine carefully curated pan-cancer gene signatures, resulting in over 400,000 single-cell transcriptomes for training. The union of differentially expressed genes across datasets was taken as the features for model construction to comprehensively capture tumor transcriptional diversity. scMalignantFinder outperformed existing automated methods across two gold-standard and eleven patient-derived scRNA-seq datasets. The capability to predict malignancy probability empowers scMalignantFinder to capture dynamic characteristics during tumor progression. Furthermore, scMalignantFinder holds the potential to annotate malignant regions in tumor spatial transcriptomics. Overall, we provide an efficient tool for detecting heterogeneous malignant cell populations.
Collapse
Affiliation(s)
- Qiaoni Yu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
- Shanghai Genbase Biotechnology Co., Ltd, Shanghai, China
| | - Yuan-Yuan Li
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China.
| | - Yunqin Chen
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China.
- Shanghai Genbase Biotechnology Co., Ltd, Shanghai, China.
| |
Collapse
|
45
|
Creyns B, MacKenzie B, Jannini Sa YAP, Coelho AL, Christensen D, Parimon T, Windsor B, Hogaboam CM. Caveolin Scaffolding Domain (CSD) Peptide LTI-2355 Modulates the Phagocytic and Synthetic Activity of Lung-Derived Myeloid Cells in Idiopathic Pulmonary Fibrosis (IPF) and Post-Acute Sequelae of COVID Fibrosis (PASC-F). Biomedicines 2025; 13:796. [PMID: 40299362 PMCID: PMC12024842 DOI: 10.3390/biomedicines13040796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 03/17/2025] [Accepted: 03/21/2025] [Indexed: 04/30/2025] Open
Abstract
Rationale: The role of the innate immune system in idiopathic pulmonary fibrosis (IPF) remains poorly understood. However, a functional myeloid compartment is required to remove dying cells and cellular debris, as well as to mediate innate immune responses against pathogens. Aberrant macrophage activity has been described in patients with post-acute sequelae of COVID fibrosis (PASC-F), and caveolin scaffolding domain (CSD) peptides have been found to attenuate inflammation and fibrosis in mouse lung injury models. Therefore, we examined, for the first time, the effects of CSD peptide LTI-2355 on the functional and synthetic properties of human myeloid cells isolated from lung explant tissue of donor lungs as well as IPF and PASC-F lung explant tissue. Methods and Results: CD45+ myeloid cells isolated from lung explant tissue from IPF and PASC-F patients exhibited an impaired capacity to clear autologous dead cells and cellular debris. The uptake of pathogen-coated bioparticles was impaired in myeloid cells from both fibrotic patient groups independent of the type of pathogen, highlighting an intrinsic functional cell impairment. LTI-2355 improved the phagocytic activity of both IPF and PASC-F myeloid cells, and this improvement was paired with decreased proinflammatory and pro-fibrotic synthetic activity. LTI-2355 was also shown to primarily target CD206-expressing IPF and PASC-F myeloid cells. Conclusions: Primary myeloid cells from IPF and PASC-F patients exhibit dysfunctional phagocytic and synthetic properties that are modulated by LTI-2355. LTI-2355 treatment of IPF myeloid cells resulted in significantly reduced sCD163, IFN-α2, IFN-γ, IL-2, IL-10, IL-12p40, and MMP-1 in the cell supernatant. This study highlights an additional mechanism of action of the CSD peptide in the treatment of IPF and progressive fibrotic lung disease.
Collapse
Affiliation(s)
- Brecht Creyns
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (B.C.); (Y.A.P.J.S.); (A.L.C.); (T.P.)
| | - BreAnne MacKenzie
- Rein Therapeutics, Inc., 12407 N. Mopac Expy., Suite 250 #390, Austin, TX 78758, USA; (B.M.); (B.W.)
| | - Yago Amigo Pinho Jannini Sa
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (B.C.); (Y.A.P.J.S.); (A.L.C.); (T.P.)
| | - Ana Lucia Coelho
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (B.C.); (Y.A.P.J.S.); (A.L.C.); (T.P.)
| | - Dale Christensen
- Division of Hematology, Department of Medicine, Duke University, Durham, NC 27708, USA;
| | - Tanyalak Parimon
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (B.C.); (Y.A.P.J.S.); (A.L.C.); (T.P.)
| | - Brian Windsor
- Rein Therapeutics, Inc., 12407 N. Mopac Expy., Suite 250 #390, Austin, TX 78758, USA; (B.M.); (B.W.)
| | - Cory M. Hogaboam
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, Women’s Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (B.C.); (Y.A.P.J.S.); (A.L.C.); (T.P.)
- Rein Therapeutics, Inc., 12407 N. Mopac Expy., Suite 250 #390, Austin, TX 78758, USA; (B.M.); (B.W.)
| |
Collapse
|
46
|
van der Koog L, Woest ME, Gorter IC, Verschut V, Elferink RAB, Zuidhof AB, Nugraha DF, Koloko Ngassie ML, Bos SIT, Dhakad D, Wolters JC, Horvatovich PL, Prakash YS, Timens W, Yildirim ÖA, Brandsma CA, Frijlink HW, Nagelkerke A, Gosens R. Fibroblast-derived osteoglycin promotes epithelial cell repair. NPJ Regen Med 2025; 10:16. [PMID: 40133363 PMCID: PMC11937367 DOI: 10.1038/s41536-025-00404-3] [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: 12/02/2024] [Accepted: 03/14/2025] [Indexed: 03/27/2025] Open
Abstract
There is an urgent need for innovative therapies targeting defective epithelial repair in chronic diseases like COPD. The mesenchymal niche is a critical regulator in epithelial stem cell activation, suggesting that their secreted factors are possible potent drug targets. Utilizing a proteomics-guided drug discovery strategy, we explored the lung fibroblast secretome to uncover impactful drug targets. Our lung organoid assays identified several regenerative ligands, with osteoglycin (OGN) showing the most profound effects. Transcriptomic analyses revealed that OGN enhances alveolar progenitor differentiation, detoxifies reactive oxygen species, and strengthens fibroblast-epithelial crosstalk. OGN expression was diminished in COPD patients and smoke-exposed mice. An active fragment of OGN (leucine-rich repeat regions 4-7) replicated full-length OGN's regenerative effects, significantly ameliorating elastase-induced lung injury in lung slices and improving lung function in vivo. These findings highlight OGN as a pivotal secreted protein for alveolar epithelial repair, positioning its active fragment as a promising therapeutic for COPD.
Collapse
Affiliation(s)
- Luke van der Koog
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Groningen, The Netherlands
| | | | | | | | - Robin A B Elferink
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Groningen, The Netherlands
| | - Annet B Zuidhof
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Groningen, The Netherlands
| | - Dyan F Nugraha
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Groningen, The Netherlands
| | - Maunick L Koloko Ngassie
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Groningen, The Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Sophie I T Bos
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Groningen, The Netherlands
| | - Deepesh Dhakad
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Justina C Wolters
- Department of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands
| | - Peter L Horvatovich
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Wim Timens
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Groningen, The Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
| | - Önder A Yildirim
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
- Institute of Experimental Pneumology, University Hospital, Ludwig-Maximilians University, Munich, Germany
| | - Corry-Anke Brandsma
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Groningen, The Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
| | - Henderik W Frijlink
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Anika Nagelkerke
- Department of Pharmaceutical Analysis, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Reinoud Gosens
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands.
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
47
|
Ushakumary MG, Chrisler WB, Bandyopadhyay G, Huyck H, Gorman BL, Beishembieva N, Pitonza A, Lai ZJ, Fillmore TL, Attah IK, Dylag AM, Misra R, Carson JP, Adkins JN, Pryhuber GS, Clair G. Sorted-Cell Proteomics Reveals an AT1-Associated Epithelial Cornification Phenotype and Suggests Endothelial Redox Imbalance in Human Bronchopulmonary Dysplasia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.20.644398. [PMID: 40166356 PMCID: PMC11957130 DOI: 10.1101/2025.03.20.644398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Bronchopulmonary dysplasia (BPD) is a neonatal lung disease characterized by inflammation and scarring leading to long-term tissue damage. Previous whole tissue proteomics identified BPD-specific proteome changes and cell type shifts. Little is known about the proteome-level changes within specific cell populations in disease. Here, we sorted epithelial (EPI) and endothelial (ENDO) cell populations based on their differential surface markers from normal and BPD human lungs. Using a low-input compatible sample preparation method (MicroPOT), proteins were extracted and digested into peptides and subjected to Liquid Chromatography-tandem Mass Spectrometry (LC-MS/MS) proteome analysis. Of the 4,970 proteins detected, 293 were modulated in abundance or detection in the EPI population and 422 were modulated in ENDO cells. Modulation of proteins associated with actin-cytoskeletal function such as SCEL, LMO7, and TBA1B were observed in the BPD EPIs. Using confocal imaging and analysis, we validated the presence of aberrant multilayer-like structures comprising SCEL and LMO7, known to be associated with epidermal cornification, in the human BPD lung. This is the first report of accumulation of cornification-associated proteins in BPD. Their localization in the alveolar parenchyma, primarily associated with alveolar type 1 (AT1) cells, suggests a role in the BPD post-injury response. In the ENDOs, redox balance and mitochondrial function pathways were modulated. Alternative mRNA splicing and cell proliferative functions were elevated in both populations suggesting potential dysregulation of cell progenitor fate. This study characterized the proteome of epithelial and endothelial cells from the BPD lung for the first time, identifying population-specific changes in BPD pathogenesis. New & Noteworthy The study is the first to perform proteomics on sorted pulmonary epithelial and endothelial populations from BPD and age-matched control human donors. We identified an increase in cornification-associated proteins in BPD (e.g., SCEL and LMO7), and evidenced the presence of multilayered structures unique to BPD alveolar regions, associated with alveolar type 1 (AT1) cells. By changing the nature and/or biomechanical properties of the epithelium, these structures may alter the behavior of other alveolar cell types potentially contributing to the arrested alveolarization observed in BPD. Lastly, our data suggest the modulation of cell proliferation and redox homeostasis in BPD providing potential mechanisms for the reduced vascular growth associated with BPD.
Collapse
|
48
|
Jain N, Ogbonna EC, Maliga Z, Jacobson C, Zhang L, Shih A, Rosenberg J, Kalam H, Gagné A, Solomon IH, Santagata S, Sorger PK, Aldridge BB, Martinot AJ. Multiomic analysis identifies suppressive myeloid cell populations in human TB granulomas. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.10.642376. [PMID: 40161687 PMCID: PMC11952478 DOI: 10.1101/2025.03.10.642376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Tuberculosis (TB) remains a major global health challenge, particularly in the context of multidrug-resistant (MDR) Mycobacterium tuberculosis (Mtb). Host-directed therapies (HDTs) have been proposed as adjunctive therapy to enhance immune control of infection. Recently, one such HDT, pharmacologic modulation of myeloid-derived suppressor cells (MDSCs), has been proposed to treat MDR-TB. While MDSCs have been well characterized in cancer, their role in TB pathogenesis remains unclear. To investigate whether MDSCs or other myeloid suppressor populations contribute to TB granuloma microenvironments (GME), we performed spatial transcriptional profiling and single-cell immunophenotyping on eighty-four granulomas in lung specimens from three individuals with active disease. Granulomas were histologically classified based on H&E staining, and transcriptional signatures were compared across regions of interest (ROIs) at different states of granuloma maturation. Our analysis revealed that immune suppression within granuloma was not primarily driven by classical MDSCs but rather by multiple myeloid cell subsets, including dendritic cells expressing indoleamine 2,3 dioxygenase-1 expressing (IDO1+ DCs). IDO1+ DCs were the most frequently observed suppressive myeloid cells, particularly in cellular regions, and their spatial proximity to activated T cells suggested localized immunosuppression. Importantly, granulomas at different stages contained distinct proportions of suppressor myeloid cells, with necrotic and cellular regions showing different myeloid phenotypes that may influence granuloma progression. Gene set enrichment analysis (GSEA) further indicated that elevated IDO1 expression was associated with a complex immune response that balanced suppressive signaling, immune activation, and cellular metabolism. These findings suggest that classical MDSCs, as defined in tumor microenvironments, likely play a minor role in TB, whereas IDO1+ DCs may be key regulators of immune suppression in granulomas influencing local Mtb control in infected lung. A deeper understanding of the role of IDO1+ suppressive myeloid cells in TB granulomas is essential to assessing their potential as therapeutic targets in TB treatment.
Collapse
|
49
|
Li Z, Zhang M, Zhang Y, Gan Y, Zhu Z, Wang J, Zhou Y, Yu G, Wang L. Integrative analysis of gene expression and chromatin dynamics multi-omics data in mouse models of bleomycin-induced lung fibrosis. Epigenetics Chromatin 2025; 18:11. [PMID: 40069909 PMCID: PMC11900494 DOI: 10.1186/s13072-025-00579-5] [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: 10/29/2024] [Accepted: 03/01/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Pulmonary fibrosis is a relentless and ultimately fatal lung disorder. Despite a wealth of research, the intricate molecular pathways that contribute to the onset of PF, especially the aspects related to epigenetic modifications and chromatin dynamics, continue to be elusive and not fully understood. METHODS Utilizing a bleomycin-induced pulmonary fibrosis model, we conducted a comprehensive analysis of the interplay between chromatin structure, chromatin accessibility, gene expression patterns, and cellular heterogeneity. Our chromatin structure analysis included 5 samples (2 control and 3 bleomycin-treated), while accessibility and expression analysis included 6 samples each (3 control and 3 bleomycin-treated). RESULTS We found that chromatin architecture, with its alterations in compartmentalization and accessibility, is positively correlated with genome-wide gene expression changes during fibrosis. The importance of immune system inflammation and extracellular matrix reorganization in fibrosis is underscored by these chromatin alterations. Transcription factors such as PU.1, AP-1, and IRF proteins, which are pivotal in immune regulation, are associated with an increased abundance of their motifs in accessible genomic regions and are correlated with highly expressed genes. CONCLUSIONS We identified 14 genes that demonstrated consistent changes in their expression, accessibility, and compartmentalization, suggesting their potential as promising targets for the development of treatments for lung fibrosis.
Collapse
Affiliation(s)
- Zhongzheng Li
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, 46 Jianshe Road, Xinxiang, Henan, 453007, China
| | - Mengke Zhang
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, 46 Jianshe Road, Xinxiang, Henan, 453007, China
| | - Yujie Zhang
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, 46 Jianshe Road, Xinxiang, Henan, 453007, China
| | - Yulong Gan
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, 46 Jianshe Road, Xinxiang, Henan, 453007, China
| | - Zhao Zhu
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, 46 Jianshe Road, Xinxiang, Henan, 453007, China
| | - Jiawei Wang
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, 46 Jianshe Road, Xinxiang, Henan, 453007, China
| | - Yanlin Zhou
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, 46 Jianshe Road, Xinxiang, Henan, 453007, China
| | - Guoying Yu
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, 46 Jianshe Road, Xinxiang, Henan, 453007, China.
| | - Lan Wang
- State Key Laboratory of Cell Differentiation and Regulation, Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Pingyuan Laboratory, College of Life Science, Henan Normal University, 46 Jianshe Road, Xinxiang, Henan, 453007, China.
| |
Collapse
|
50
|
Unjitwattana T, Huang Q, Yang Y, Tao L, Yang Y, Zhou M, Du Y, Garmire LX. Single-cell RNA-seq data have prevalent blood contamination but can be rescued by Originator, a computational tool separating single-cell RNA-seq by genetic and contextual information. Genome Biol 2025; 26:52. [PMID: 40069819 PMCID: PMC11895284 DOI: 10.1186/s13059-025-03495-9] [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: 06/15/2024] [Accepted: 02/05/2025] [Indexed: 03/15/2025] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) data from complex human tissues have prevalent blood cell contamination during the sample preparation process. They may also comprise cells of different genetic makeups. We propose a new computational framework, Originator, which deciphers single cells by genetic origin and separates immune cells of blood contamination from those of expected tissue-resident cells. We demonstrate the accuracy of Originator at separating immune cells from the blood and tissue as well as cells of different genetic origins, using a variety of artificially mixed and real datasets, including pancreatic cancer and placentas as examples.
Collapse
Affiliation(s)
- Thatchayut Unjitwattana
- Department of Biomedical Engineering, University of Michigan, 2200 , Bonisteel, Ann Arbor, MI, 48109, USA
| | - Qianhui Huang
- Department of Computation Medicine and Bioinformatics, University of Michigan, 100 Washtenaw Ave, Ann Arbor, MI, 48109, USA
| | - Yiwen Yang
- Department of Computation Medicine and Bioinformatics, University of Michigan, 100 Washtenaw Ave, Ann Arbor, MI, 48109, USA
| | - Leyang Tao
- Department of Biomedical Engineering, University of Michigan, 2200 , Bonisteel, Ann Arbor, MI, 48109, USA
| | - Youqi Yang
- Department of Biostatistics, University of Michigan, 1415 Washington Heights, Ann Arbor, MI, 48109, USA
| | - Mengtian Zhou
- Department of Statistics, University of Michigan, 1085 S University Ave, Ann Arbor, MI, 48109, USA
| | - Yuheng Du
- Department of Computation Medicine and Bioinformatics, University of Michigan, 100 Washtenaw Ave, Ann Arbor, MI, 48109, USA
| | - Lana X Garmire
- Department of Biomedical Engineering, University of Michigan, 2200 , Bonisteel, Ann Arbor, MI, 48109, USA.
- Department of Computation Medicine and Bioinformatics, University of Michigan, 100 Washtenaw Ave, Ann Arbor, MI, 48109, USA.
- Department of Biostatistics, University of Michigan, 1415 Washington Heights, Ann Arbor, MI, 48109, USA.
| |
Collapse
|