1
|
Berdnikovs S, Newcomb DC, Haruna NF, McKernan KE, Kuehnle SN, Gebretsadik T, McKennan C, Ma S, Cephus JY, Rosas-Salazar C, Anderson LJ, Gern JE, Hartert T. Single-cell profiling demonstrates the combined effect of wheeze phenotype and infant viral infection on airway epithelial development. SCIENCE ADVANCES 2025; 11:eadr9995. [PMID: 40408478 PMCID: PMC12101503 DOI: 10.1126/sciadv.adr9995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 04/21/2025] [Indexed: 05/25/2025]
Abstract
The development of the airway epithelium in asthma is unclear. We characterized nasal airway epithelial cell (NAEC) developmental phenotypes from children aged 2 to 3 years in an a priori designed nested birth cohort from four mutually exclusive groups of wheezers/nonwheezers and respiratory syncytial virus (RSV)-infected/uninfected in the first year of life. NAECs were differentiated, followed by single-cell RNA sequencing analysis and in vitro RSV infection. Gene expression of NAECs from children with a wheeze phenotype indicated abnormal differentiation and basal cell activation of developmental pathways, plasticity in precursor differentiation, delayed onset of maturation, increased diversity of RSV receptors, and blunted antiviral immune responses to in vitro RSV infection. The most marked changes in differentiation were observed in NAECs from children with both wheeze and RSV in the first year of life. Together, this suggests that airway epithelium in children with wheeze is developmentally reprogrammed and characterized by increased barrier permeability, decreased antiviral response, and altered RSV receptor expression.
Collapse
Affiliation(s)
- Sergejs Berdnikovs
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Dawn C. Newcomb
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nana-Fatima Haruna
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Kaitlin E. McKernan
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shelby N. Kuehnle
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Tebeb Gebretsadik
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Siyuan Ma
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | | | - Larry J. Anderson
- Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, Atlanta, GA, USA
| | - James E. Gern
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health-Madison, Madison WI, USA
| | - Tina Hartert
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Dinant S, Castille J, Deloizy C, Bruder E, Sedano L, Daniel-Carlier N, Da Costa B, Passet B, Béringue V, Duchesne A, Chevalier C, Larcher T, Moazami-Goudarzi K, Vilotte JL, Le Goffic R. The prion-family protein Doppel exerts a protective role during influenza virus infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf031. [PMID: 40204637 DOI: 10.1093/jimmun/vkaf031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 01/24/2025] [Indexed: 04/11/2025]
Abstract
The cellular form of the prion protein (PrPC), known for its involvement as a misfolded isoform in transmissible spongiform encephalopathies, has recently been identified to exert a protective effect against viral infections. In this study, we explored the role of 2 other prion family members, Shadoo and Doppel, in protection against influenza A virus infection in mice. Lung expression levels of these genes revealed marked differences, with high expression of PrPC, low expression of Doppel, while Shadoo remained undetectable. Mice genetically knocked out for the genes encoding PrPC, Prnp-/- or Doppel, Prnd-/-, showed increased susceptibility to the virus, resulting in elevated morbidity compared with wild-type mice and mice knocked out for Shadoo, Sprn-/-. Unlike previous results observed in Prnp-/- mice, the absence of Doppel does not show enhancing effect on virus replication levels. Histological analysis of lung tissue from Prnd-/- mice revealed no difference in lesion size and severity compared with wild-type mice. However, transcriptomic analysis on day 7 postinfection revealed distinct signatures in Prnd-/- mice, highlighting the role of specific genes associated with polymorphonuclear neutrophil cells. Bronchoalveolar lavages confirmed a substantial neutrophil influx and increased inflammatory markers in the lungs of Prnd-/- mice. Neutrophil depletion experiments demonstrated a direct link between excessive neutrophil influx and increased susceptibility, mitigating pathology and partially restoring a wild-type phenotype in Prnd-/- mice. These findings underscore the complex role of Doppel in modulating the host immune response to influenza virus infection, particularly in regulating neutrophil recruitment and its implications on disease outcomes.
Collapse
Affiliation(s)
- Soraya Dinant
- UMR892 VIM, UVSQ, INRAE, Université Paris-Saclay, Jouy-en-Josas, F78352, France
| | - Johan Castille
- UMR1313 GABI, AgroParisTech, INRAE, Université Paris-Saclay, Jouy-en-Josas, F78350, France
| | - Charlotte Deloizy
- UMR892 VIM, UVSQ, INRAE, Université Paris-Saclay, Jouy-en-Josas, F78352, France
| | - Elise Bruder
- UMR892 VIM, UVSQ, INRAE, Université Paris-Saclay, Jouy-en-Josas, F78352, France
| | - Laura Sedano
- UMR892 VIM, UVSQ, INRAE, Université Paris-Saclay, Jouy-en-Josas, F78352, France
| | | | - Bruno Da Costa
- UMR892 VIM, UVSQ, INRAE, Université Paris-Saclay, Jouy-en-Josas, F78352, France
| | - Bruno Passet
- UMR1313 GABI, AgroParisTech, INRAE, Université Paris-Saclay, Jouy-en-Josas, F78350, France
| | - Vincent Béringue
- UMR892 VIM, UVSQ, INRAE, Université Paris-Saclay, Jouy-en-Josas, F78352, France
| | - Amandine Duchesne
- UMR1313 GABI, AgroParisTech, INRAE, Université Paris-Saclay, Jouy-en-Josas, F78350, France
| | | | | | | | - Jean-Luc Vilotte
- UMR1313 GABI, AgroParisTech, INRAE, Université Paris-Saclay, Jouy-en-Josas, F78350, France
| | - Ronan Le Goffic
- UMR892 VIM, UVSQ, INRAE, Université Paris-Saclay, Jouy-en-Josas, F78352, France
| |
Collapse
|
5
|
Lu J, Wang Z, Zhang L. Single-cell transcriptome analysis revealing mechanotransduction via the Hippo/YAP pathway in promoting fibroblast-to-myofibroblast transition and idiopathic pulmonary fibrosis development. Gene 2025; 943:149271. [PMID: 39855369 DOI: 10.1016/j.gene.2025.149271] [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: 09/27/2024] [Revised: 12/12/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
OBJECTIVE Idiopathic pulmonary fibrosis (IPF) is an irreversible and fatal interstitial lung disease, characterized by excessive extracellular matrix (ECM) secretion that disrupts normal alveolar structure. This study aims to explore the potential molecular mechanisms underlying the promotion of IPF development. METHODS Firstly, we compared the transcriptome and single-cell sequencing data from lung tissue samples of patients with IPF and healthy individuals. Subsequently, we conducted Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses on the differentially expressed genes (DEGs). Furthermore, we employed sodium alginate hydrogels with varying degrees of crosslinking to provide differential mechanical stress, mimicking the mechanical microenvironment in vivo during lung fibrosis. On this basis, we examined cytoskeletal remodeling in fibroblasts MRC-5, mRNA expression of multiple related genes, immunofluorescence localization, and cellular proliferation capacity. RESULTS Bioinformatics analysis revealed a series of DEGs associated with IPF. Further functional and pathway enrichment analyses indicated that these DEGs were primarily enriched in ECM-related biological processes. Single-cell sequencing data revealed that fibroblasts and myofibroblasts are the main contributors to excessive ECM secretion and suggested activation of mechanotransduction and the Hippo/YAP signaling pathway in myofibroblasts. Cellular experiments demonstrated that sodium alginate hydrogels with different stiffness can simulate different mechanical stress environments, thereby affecting cytoskeletal rearrangement and Hippo/YAP pathway activity in MRC-5 lung fibroblasts. Notably, high levels of mechanical stress promoted YAP nuclear translocation, increased expression of type I collagen and α-SMA, and enhanced proliferative capacity. Additionally, we also found that fibroblasts primarily participate in mechanotransduction through the Rho/ROCK and Integrin/FAK pathways under high mechanical stress conditions, ultimately upregulating the gene expression of CCNE1/2, CTGF, and FGF1. CONCLUSION Our study uncovers the crucial role of cytoskeletal mechanotransduction in myofibroblast transformation and IPF development through activation of the Hippo/YAP pathway, providing new insights into understanding the pathogenesis of IPF.
Collapse
Affiliation(s)
- Jiaqi Lu
- Department of Oncology, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, China.
| | - Zhenhua Wang
- Department of Oncology, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, China
| | - Liguo Zhang
- Department of Oncology, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, China
| |
Collapse
|
6
|
Mestan KK, Sharma AM, Lazar S, Pandey S, Parast MM, Laurent LC, Prince LS, Sahoo D. Bronchopulmonary dysplasia: signatures of monocyte-macrophage reactivity and tolerance define novel placenta-lung endotypes. Pediatr Res 2025:10.1038/s41390-025-04025-w. [PMID: 40175585 DOI: 10.1038/s41390-025-04025-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 03/12/2025] [Accepted: 03/15/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) is a complex disease involving aberrant immune responses across the lifespan, but these mechanisms are challenging to follow in human infants. Leveraging novel Signatures of Macrophage Reactivity and Tolerance (SMaRT), we hypothesized that distinct profiles of immune cell polarization in blood and lung are associated with BPD. METHODS Published transcriptomic datasets of cord blood-derived monocytes (CB-MNC), peripheral blood monocytes (PBMC) and tracheal aspirate-derived lung macrophages were linked to placental inflammatory (PID) and vascular (PVD) disease states using Amsterdam criteria, and BPD outcomes using NIH consensus criteria. Datasets were integrated using SMaRT to investigate monocyte-macrophage polarization tracked over the neonatal course. RESULTS At birth and day 1 (D1), CB-MNCs and lung macrophages exhibited significant reactivity with PID versus PVD. After D14, macrophages from PID versus PVD-exposed infants exhibited reactive phenotypes (p = 0.002), with convergence towards original placental disease. Macrophages exhibited reactivity with BPD on D1-D7 (p = 0.007), but no difference after D14. At birth, CB-MNCs from BPD patients exhibited tolerance, which persisted in PBMCs throughout the neonatal period. CONCLUSION Inflammatory versus vascular-mediated processes in developing lungs are influenced by immune cells programmed by distinct placental disease states. Circulating monocytes may play a role in attenuating macrophage reactivity towards a tolerant phenotype. IMPACT Bronchopulmonary dysplasia is a complex, multifactorial chronic lung disease in which the mechanisms of placenta-lung crosstalk are poorly understood. This study uses novel AI approaches to understand how fetal monocytes and lung macrophages contribute to the pathogenesis of BPD. The study identified changes in macrophage reactivity versus tolerance that could explain the heterogeneity and adaptability of immune cells and the placenta in modulating health and disease in the developing fetus and neonate.
Collapse
Affiliation(s)
- Karen K Mestan
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA.
| | - Abhineet M Sharma
- Department of Pediatrics, Divisions of Neonatology and Pediatric Pulmonology, University of Nebraska College of Medicine, Omaha, NE, USA
| | - Sarah Lazar
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Sonalisa Pandey
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Mana M Parast
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Louise C Laurent
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | | | - Debashis Sahoo
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA.
- Department of Computer Science and Engineering, Jacob's School of Engineering, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
7
|
Börner K, Blood PD, Silverstein JC, Ruffalo M, Satija R, Teichmann SA, Pryhuber GJ, Misra RS, Purkerson JM, Fan J, Hickey JW, Molla G, Xu C, Zhang Y, Weber GM, Jain Y, Qaurooni D, Kong Y, Bueckle A, Herr BW. Human BioMolecular Atlas Program (HuBMAP): 3D Human Reference Atlas construction and usage. Nat Methods 2025; 22:845-860. [PMID: 40082611 PMCID: PMC11978508 DOI: 10.1038/s41592-024-02563-5] [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: 03/27/2024] [Accepted: 11/11/2024] [Indexed: 03/16/2025]
Abstract
The Human BioMolecular Atlas Program (HuBMAP) aims to construct a 3D Human Reference Atlas (HRA) of the healthy adult body. Experts from 20+ consortia collaborate to develop a Common Coordinate Framework (CCF), knowledge graphs and tools that describe the multiscale structure of the human body (from organs and tissues down to cells, genes and biomarkers) and to use the HRA to characterize changes that occur with aging, disease and other perturbations. HRA v.2.0 covers 4,499 unique anatomical structures, 1,195 cell types and 2,089 biomarkers (such as genes, proteins and lipids) from 33 ASCT+B tables and 65 3D Reference Objects linked to ontologies. New experimental data can be mapped into the HRA using (1) cell type annotation tools (for example, Azimuth), (2) validated antibody panels or (3) by registering tissue data spatially. This paper describes HRA user stories, terminology, data formats, ontology validation, unified analysis workflows, user interfaces, instructional materials, application programming interfaces, flexible hybrid cloud infrastructure and previews atlas usage applications.
Collapse
Grants
- OT2 OD026675 NIH HHS
- U54 HL165443 NHLBI NIH HHS
- OT2 OD033759 NIH HHS
- U54 AG075936 NIA NIH HHS
- OT2 OD026671 NIH HHS
- OT2 OD033761 NIH HHS
- RM1HG011014 U.S. Department of Health & Human Services | NIH | National Human Genome Research Institute (NHGRI)
- U24CA268108 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- OT2 OD033760 NIH HHS
- OT2OD033760 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- R03 OD036499 NIH HHS
- U24 DK135157 NIDDK NIH HHS
- U54HL165443 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- OT2OD033759 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- OT2OD026671 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- RM1 HG011014 NHGRI NIH HHS
- U2C DK114886 NIDDK NIH HHS
- OT2OD026673 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- OT2 OD026682 NIH HHS
- OT2 OD033756 NIH HHS
- 3U54AG075936 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- U24DK135157 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- 3OT2OD026682 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- 1R03OD036499 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- U24 CA268108 NCI NIH HHS
- U2CDK114886 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- OT2OD033756 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- OT2OD026675 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- HLU01148861 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- 3OT2OD033760 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- 1OT2OD033761 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- NIH: OT2OD033759
- K.B. is a co-director of and is funded by the CIFAR MacMillan Multiscale Human program.
- S.A.T. is a co-director of and is funded by the CIFAR MacMillan Multiscale Human program. S.A.T. is a remunerated member of the Scientific Advisory Boards of Qiagen, Foresite Labs and Element Biosciences, a co-founder and equity holder of TransitionBio and EnsoCell Therapeutics, and a part-time employee of GlaxoSmithKline since January 2024.
- NIH: U2CDK114886
- U.S. Department of Health & Human Services | NIH | National Human Genome Research Institute (NHGRI)
- In the past 3 years, RS has received compensation from Bristol-Myers Squibb, ImmunAI, Resolve Biosciences, Nanostring, 10X Genomics, Neptune Bio, and the NYC Pandemic Response Lab. RS is a co-founder and equity holder of Neptune Bio.
Collapse
Affiliation(s)
- Katy Börner
- Department of Intelligent Systems Engineering, Luddy School of Informatics, Computing, and Engineering, Indiana University, Bloomington, IN, USA.
- CIFAR MacMillan Multiscale Human program, CIFAR, Toronto, Ontario, Canada.
| | - Philip D Blood
- Pittsburgh Supercomputing Center, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Jonathan C Silverstein
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Matthew Ruffalo
- Ray and Stephanie Lane Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA, USA
| | | | - Sarah A Teichmann
- CIFAR MacMillan Multiscale Human program, CIFAR, Toronto, Ontario, Canada
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | | | - Ravi S Misra
- University of Rochester Medical Center, Rochester, NY, USA
| | | | - Jean Fan
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - John W Hickey
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Chuan Xu
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Yun Zhang
- J. Craig Venter Institute, La Jolla, CA, USA
| | - Griffin M Weber
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Yashvardhan Jain
- Department of Intelligent Systems Engineering, Luddy School of Informatics, Computing, and Engineering, Indiana University, Bloomington, IN, USA
| | - Danial Qaurooni
- Department of Intelligent Systems Engineering, Luddy School of Informatics, Computing, and Engineering, Indiana University, Bloomington, IN, USA
| | - Yongxin Kong
- Department of Intelligent Systems Engineering, Luddy School of Informatics, Computing, and Engineering, Indiana University, Bloomington, IN, USA
| | - Andreas Bueckle
- Department of Intelligent Systems Engineering, Luddy School of Informatics, Computing, and Engineering, Indiana University, Bloomington, IN, USA.
| | - Bruce W Herr
- Department of Intelligent Systems Engineering, Luddy School of Informatics, Computing, and Engineering, Indiana University, Bloomington, IN, USA.
| |
Collapse
|
8
|
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
|
9
|
McSwiggin H, Wang R, Magalhães RDM, Zhu F, Doherty TA, Yan W, Jendzjowsky N. Comprehensive sequencing of the lung neuroimmune landscape in response to asthmatic induction. Front Immunol 2025; 16:1518771. [PMID: 40181989 PMCID: PMC11965707 DOI: 10.3389/fimmu.2025.1518771] [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: 10/28/2024] [Accepted: 02/13/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction Evidence demonstrates that sensory neurons respond to pathogenic/allergic infiltration and mediate immune responses, forming an integral part of host defense that becomes hypersensitized during allergy. Our objective was to investigate how asthmatic induction alters the pulmonary neuroimmune transcriptome. We hypothesized that asthmatic induction would upregulate genes in the vagal ganglia (nodose/jugular ganglia), which would be associated with asthmatic immunity, and that these would be clustered, primarily in nodose neurons. Furthermore, lungs would increase transcripts associated with nerve activation, and these would be centered in neural and neuroendocrine-like cells. Methods Standard RNA sequencing, single nucleus-RNA sequencing, and spatial RNA sequencing of vagal ganglia. Standard RNA-sequencing and spatial RNA-sequencing of lungs in naïve and mice that have undergone asthmatic induction with Alternaria alternata. Results Bulk RNA-seq revealed that genes related to allergen sensing were increased in asthmatic ganglia nodose/jugular ganglia compared to control ganglia. These genes were associated with nodose clusters as shown by single-nucleus RNA sequencing, and a distinct caudal-to-rostral spatial arrangement was presented as delineated by spatial transcriptomics. The distinct clusters closely match previous identification of nodose neuron clusters. Correspondingly, the lung transcriptome was altered with asthmatic induction such that transcripts associated with neural excitation were upregulated. The spatial distribution of these transcripts was revealed by spatial transcriptomics to illustrate that these were expressed in neuroendocrine-like cells/club cells, and neurons. Conclusions These results show that the neuroimmune transcriptome is altered in response to asthmatic induction in a cell cluster and spatially distinct manner.
Collapse
Affiliation(s)
- Hayden McSwiggin
- The Lundquist Institute for Biomedical Innovation at Harbor-University of California, Los Angeles, Medical Center, Torrance, CA, United States
| | - Rui Wang
- The Lundquist Institute for Biomedical Innovation at Harbor-University of California, Los Angeles, Medical Center, Torrance, CA, United States
| | - Rubens Daniel Miserani Magalhães
- The Lundquist Institute for Biomedical Innovation at Harbor-University of California, Los Angeles, Medical Center, Torrance, CA, United States
| | - Fengli Zhu
- The Lundquist Institute for Biomedical Innovation at Harbor-University of California, Los Angeles, Medical Center, Torrance, CA, United States
| | - Taylor A. Doherty
- Division of Allergy and Immunology, Department of Medicine, University of California, San Diego, Veterans Affairs San Diego Healthcare System, La Jolla, CA, United States
| | - Wei Yan
- The Lundquist Institute for Biomedical Innovation at Harbor-University of California, Los Angeles, Medical Center, Torrance, CA, United States
- Division of Endocrinology, Department of Medicine, Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Nicholas Jendzjowsky
- The Lundquist Institute for Biomedical Innovation at Harbor-University of California, Los Angeles, Medical Center, Torrance, CA, United States
- Division of Respiratory and Critical Care Medicine and Physiology, Department of Medicine, Harbor-UCLA Medical Center, Torrance, CA, United States
| |
Collapse
|
10
|
Blackburn JB, Tufenkjian TS, Liu Y, Nichols DS, Blackwell TS, Richmond BW. A Single-Cell RNA Sequencing Atlas of the Chronic Obstructive Pulmonary Disease Distal Lung to Predict Cell-Cell Communication. Am J Respir Cell Mol Biol 2025; 72:332-335. [PMID: 39356793 PMCID: PMC11890073 DOI: 10.1165/rcmb.2024-0232le] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 09/11/2024] [Indexed: 10/04/2024] Open
Affiliation(s)
- Jessica B. Blackburn
- Veterans Affairs Medical CenterNashville, Tennessee
- Vanderbilt University School of MedicineNashville, Tennessee
| | - Tiffany S. Tufenkjian
- Veterans Affairs Medical CenterNashville, Tennessee
- Vanderbilt University School of MedicineNashville, Tennessee
| | - Yang Liu
- Vanderbilt University School of MedicineNashville, Tennessee
| | | | - Timothy S. Blackwell
- Veterans Affairs Medical CenterNashville, Tennessee
- Vanderbilt University School of MedicineNashville, Tennessee
- Vanderbilt UniversityNashville, Tennessee
| | - Bradley W. Richmond
- Veterans Affairs Medical CenterNashville, Tennessee
- Vanderbilt University School of MedicineNashville, Tennessee
- Vanderbilt UniversityNashville, Tennessee
| |
Collapse
|
11
|
Cui H, Banerjee S, Xie N, Hussain M, Jaiswal A, Liu H, Kulkarni T, Antony VB, Liu RM, Colonna M, Liu G. TREM2 promotes lung fibrosis via controlling alveolar macrophage survival and pro-fibrotic activity. Nat Commun 2025; 16:1761. [PMID: 39971937 PMCID: PMC11840137 DOI: 10.1038/s41467-025-57024-0] [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: 07/01/2024] [Accepted: 02/06/2025] [Indexed: 02/21/2025] Open
Abstract
Lung macrophages play a pivotal role in pulmonary fibrosis, with monocyte-derived alveolar macrophages driving disease progression. However, the mechanisms regulating their pro-fibrotic behavior and survival remain unclear, and effective therapeutic strategies are lacking. Here we show that triggering receptors expressed on myeloid cells 2 are predominantly expressed on monocyte-derived alveolar macrophages in fibrotic mouse lungs and are significantly elevated in lung macrophages from patients with idiopathic pulmonary fibrosis. Deletion or knockdown of this receptor disrupts intracellular survival signaling, promotes macrophage apoptosis, and attenuates their pro-fibrotic phenotype. We further demonstrate that a lipid mediator and a high-avidity ligand of this receptor, encountered by macrophages in the alveolar milieu, enhance macrophage survival and activity. Ablation of TREM2 or blocking this receptor with soluble receptors or specific antibodies effectively alleviates lung fibrosis in male mice. These findings identify this receptor as a critical regulator of macrophage-mediated fibrosis and a promising therapeutic target for intervention.
Collapse
Affiliation(s)
- Huachun Cui
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Sami Banerjee
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Na Xie
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Musaddique Hussain
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Ashish Jaiswal
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Hongli Liu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Tejaswini Kulkarni
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Veena B Antony
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Rui-Ming Liu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Gang Liu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
12
|
King EM, Zhao Y, Moore CM, Steinhart B, Anderson KC, Vestal B, Moore PK, McManus SA, Evans CM, Mould KJ, Redente EF, McCubbrey AL, Janssen WJ. Gpnmb and Spp1 mark a conserved macrophage injury response masking fibrosis-specific programming in the lung. JCI Insight 2024; 9:e182700. [PMID: 39509324 PMCID: PMC11665561 DOI: 10.1172/jci.insight.182700] [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: 05/31/2024] [Accepted: 10/30/2024] [Indexed: 11/15/2024] Open
Abstract
Macrophages are required for healthy repair of the lungs following injury, but they are also implicated in driving dysregulated repair with fibrosis. How these 2 distinct outcomes of lung injury are mediated by different macrophage subsets is unknown. To assess this, single-cell RNA-Seq was performed on lung macrophages isolated from mice treated with LPS or bleomycin. Macrophages were categorized based on anatomic location (airspace versus interstitium), developmental origin (embryonic versus recruited monocyte derived), time after inflammatory challenge, and injury model. Analysis of the integrated dataset revealed that macrophage subset clustering was driven by macrophage origin and tissue compartment rather than injury model. Gpnmb-expressing recruited macrophages that were enriched for genes typically associated with fibrosis were present in both injury models. Analogous GPNMB-expressing macrophages were identified in datasets from both fibrotic and nonfibrotic lung disease in humans. We conclude that this subset represents a conserved response to tissue injury and is not sufficient to drive fibrosis. Beyond this conserved response, we identified that recruited macrophages failed to gain resident-like programming during fibrotic repair. Overall, fibrotic versus nonfibrotic tissue repair is dictated by dynamic shifts in macrophage subset programming and persistence of recruited macrophages.
Collapse
Affiliation(s)
- Emily M. King
- Medical Scientist Training Program, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Yifan Zhao
- Center for Genes, Environment, and Health, and
| | | | | | | | | | - Peter K. Moore
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | | | - Christopher M. Evans
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Kara J. Mould
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Elizabeth F. Redente
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | - Alexandra L. McCubbrey
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - William J. Janssen
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
13
|
Lamba R, Paguntalan AM, Petrov PB, Naba A, Izzi V. MatriCom: a scRNA-Seq data mining tool to infer ECM-ECM and cell-ECM communication systems. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.10.627834. [PMID: 39763937 PMCID: PMC11702561 DOI: 10.1101/2024.12.10.627834] [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] [Indexed: 01/14/2025]
Abstract
The ECM is a complex and dynamic meshwork of proteins that forms the framework of all multicellular organisms. Protein interactions within the ECM are critical to building and remodeling the ECM meshwork, while interactions between ECM proteins and cell surface receptors are essential for the initiation of signal transduction and the orchestration of cellular behaviors. Here, we report the development of MatriCom, a web application (https://matrinet.shinyapps.io/matricom) and a companion R package (https://github.com/Izzilab/MatriCom), devised to mine scRNA-Seq datasets and infer communications between ECM components and between different cell populations and the ECM. To impute interactions from expression data, MatriCom relies on a unique database, MatriComDB, that includes over 25,000 curated interactions involving matrisome components, with data on 80% of the ~1,000 genes that compose the mammalian matrisome. MatriCom offers the option to query open-access datasets sourced from large sequencing efforts (Tabula Sapiens, The Human Protein Atlas, HuBMAP) or to process user-generated datasets. MatriCom is also tailored to account for the specific rules governing ECM protein interactions and offers options to customize the output through stringency filters. We illustrate the usability of MatriCom with the example of the human kidney matrisome communication network. Last, we demonstrate how the integration of 46 scRNA-Seq datasets led to the identification of both ubiquitous and tissue-specific ECM communication patterns. We envision that MatriCom will become a powerful resource to elucidate the roles of different cell populations in ECM-ECM and cell-ECM interactions and their dysregulations in the context of diseases such as cancer or fibrosis.
Collapse
Affiliation(s)
- Rijuta Lamba
- Faculty of Biochemistry and Molecular Medicine & Faculty of Medicine, BioIM Unit, University of Oulu, Oulu, FI-90014, Finland
| | - Asia M. Paguntalan
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Petar B. Petrov
- Infotech Institute, University of Oulu, Oulu, FI-90014, Finland
| | - Alexandra Naba
- Department of Physiology and Biophysics, University of Illinois Chicago, Chicago, IL 60612, USA
- University of Illinois Cancer Center, Chicago, IL 60612, USA
| | - Valerio Izzi
- Faculty of Biochemistry and Molecular Medicine & Faculty of Medicine, BioIM Unit, University of Oulu, Oulu, FI-90014, Finland
- Infotech Institute, University of Oulu, Oulu, FI-90014, Finland
| |
Collapse
|
14
|
Jin W, Pei J, Roy JR, Jayaraman S, Ahalliya RM, Kanniappan GV, Mironescu M, Palanisamy CP. Comprehensive review on single-cell RNA sequencing: A new frontier in Alzheimer's disease research. Ageing Res Rev 2024; 100:102454. [PMID: 39142391 DOI: 10.1016/j.arr.2024.102454] [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: 07/08/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 08/16/2024]
Abstract
Alzheimer's disease (AD) is a multifaceted neurodegenerative condition marked by gradual cognitive deterioration and the loss of neurons. While conventional bulk RNA sequencing techniques have shed light on AD pathology, they frequently obscure the cellular diversity within brain tissues. The advent of single-cell RNA sequencing (scRNA-seq) has transformed our capability to analyze the cellular composition of AD, allowing for the detection of unique cell populations, rare cell types, and gene expression alterations at an individual cell level. This review examines the use of scRNA-seq in AD research, focusing on its contributions to understanding cellular diversity, disease progression, and potential therapeutic targets. We discuss key technological innovations, data analysis techniques, and challenges associated with scRNA-seq in studying AD. Furthermore, we highlight recent studies that have utilized scRNA-seq to identify novel biomarkers, uncover disease-associated pathways, and elucidate the role of non-neuronal cells, such as microglia and astrocytes, in AD pathogenesis. By providing a comprehensive overview of advancements in scRNA-seq for unraveling cellular heterogeneity in AD, this review highlights the transformative impact of scRNA-seq on our comprehension of disease mechanisms and the creation of targeted treatments.
Collapse
Affiliation(s)
- Wengang Jin
- Qinba State Key Laboratory of Biological Resources and Ecological Environment, 2011 QinLing-Bashan Mountains Bioresources Comprehensive Development C. I. C, Shaanxi Province Key Laboratory of Bio-Resources, College of Bioscience and Bioengineering, Shaanxi University of Technology, Hanzhong 723001, China
| | - JinJin Pei
- Qinba State Key Laboratory of Biological Resources and Ecological Environment, 2011 QinLing-Bashan Mountains Bioresources Comprehensive Development C. I. C, Shaanxi Province Key Laboratory of Bio-Resources, College of Bioscience and Bioengineering, Shaanxi University of Technology, Hanzhong 723001, China
| | - Jeane Rebecca Roy
- Department of Anatomy, Bhaarath Medical College and hospital, Bharath Institute of Higher Education and Research (BIHER), Chennai, Tamil Nadu 600073, India
| | - Selvaraj Jayaraman
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospital, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai 600077, India
| | - Rathi Muthaiyan Ahalliya
- Department of Biochemistry and Cancer Research Centre, FASCM, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu 641021, India
| | - Gopalakrishnan Velliyur Kanniappan
- Center for Global Health Research, Saveetha Medical College & Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Thandalam, Chennai, Tamil Nadu 602105, India.
| | - Monica Mironescu
- Faculty of Agricultural Sciences Food Industry and Environmental Protection, Lucian Blaga University of Sibiu, Bv. Victoriei 10, Sibiu 550024, Romania.
| | - Chella Perumal Palanisamy
- Department of Chemical Technology, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand.
| |
Collapse
|
15
|
Berdnikovs S, Newcomb DC, Hartert TV. How early life respiratory viral infections impact airway epithelial development and may lead to asthma. Front Pediatr 2024; 12:1441293. [PMID: 39156016 PMCID: PMC11327159 DOI: 10.3389/fped.2024.1441293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 07/25/2024] [Indexed: 08/20/2024] Open
Abstract
Childhood asthma is a common chronic disease of the airways that results from host and environment interactions. Most risk factor studies of asthma point to the first year of life as a susceptibility window of mucosal exposure that directly impacts the airway epithelium and airway epithelial cell development. The development of the airway epithelium, which forms a competent barrier resulting from coordinated interactions of different specialized cell subsets, occurs during a critical time frame in normal postnatal development in the first year of life. Understanding the normal and aberrant developmental trajectory of airway epithelial cells is important in identifying pathways that may contribute to barrier dysfunction and asthma pathogenesis. Respiratory viruses make first contact with and infect the airway mucosa. Human rhinovirus (HRV) and respiratory syncytial virus (RSV) are mucosal pathogens that are consistently identified as asthma risk factors. Respiratory viruses represent a unique early life exposure, different from passive irritant exposures which injure the developing airway epithelium. To replicate, respiratory viruses take over the host cell transcriptional and translational processes and exploit host cell energy metabolism. This takeover impacts the development and differentiation processes of airway epithelial cells. Therefore, delineating the mechanisms through which early life respiratory viral infections alter airway epithelial cell development will allow us to understand the maturation and heterogeneity of asthma and develop tools tailored to prevent disease in specific children. This review will summarize what is understood about the impact of early life respiratory viruses on the developing airway epithelium and define critical gaps in our knowledge.
Collapse
Affiliation(s)
- Sergejs Berdnikovs
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Dawn C. Newcomb
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Tina V. Hartert
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
16
|
Sarfraz I, Wang Y, Shastry A, Teh WK, Sokolov A, Herb BR, Creasy HH, Virshup I, Dries R, Degatano K, Mahurkar A, Schnell DJ, Madrigal P, Hilton J, Gehlenborg N, Tickle T, Campbell JD. MAMS: matrix and analysis metadata standards to facilitate harmonization and reproducibility of single-cell data. Genome Biol 2024; 25:205. [PMID: 39090672 PMCID: PMC11292877 DOI: 10.1186/s13059-024-03349-w] [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: 03/06/2023] [Accepted: 07/24/2024] [Indexed: 08/04/2024] Open
Abstract
Many datasets are being produced by consortia that seek to characterize healthy and disease tissues at single-cell resolution. While biospecimen and experimental information is often captured, detailed metadata standards related to data matrices and analysis workflows are currently lacking. To address this, we develop the matrix and analysis metadata standards (MAMS) to serve as a resource for data centers, repositories, and tool developers. We define metadata fields for matrices and parameters commonly utilized in analytical workflows and developed the rmams package to extract MAMS from single-cell objects. Overall, MAMS promotes the harmonization, integration, and reproducibility of single-cell data across platforms.
Collapse
Affiliation(s)
- Irzam Sarfraz
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Yichen Wang
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Amulya Shastry
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Wei Kheng Teh
- European Bioinformatics Institute, European Molecular Biology Laboratory, Hinxton, Cambridgeshire, UK
| | - Artem Sokolov
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Brian R Herb
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Heather H Creasy
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Isaac Virshup
- Department of Computational Health, Helmholtz Munich, Oberschleißheim, Germany
| | - Ruben Dries
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Kylee Degatano
- Data Sciences Platform, Broad Institute, Cambridge, MA, USA
| | - Anup Mahurkar
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Daniel J Schnell
- Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Pedro Madrigal
- European Bioinformatics Institute, European Molecular Biology Laboratory, Hinxton, Cambridgeshire, UK
| | - Jason Hilton
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Nils Gehlenborg
- Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Timothy Tickle
- Data Sciences Platform, Broad Institute, Cambridge, MA, USA
| | - Joshua D Campbell
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
17
|
Ray JL, Walum J, Jelic D, Barnes R, Bentley ID, Britt RD, Englert JA, Ballinger MN. scRNA-seq identifies unique macrophage population in murine model of ozone induced asthma exacerbation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.23.604740. [PMID: 39211080 PMCID: PMC11361036 DOI: 10.1101/2024.07.23.604740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Ozone (O 3 ) inhalation triggers asthmatic airway hyperresponsiveness (AHR), but the mechanisms by which this occurs are unknown. Previously, we developed a murine model of dust mite, ragweed, and aspergillus (DRA)-induced allergic lung inflammation followed by O 3 exposure for mechanistic investigation. The present study used single cell RNA-sequencing for unbiased profiling of immune cells within the lungs of mice exposed to DRA, O 3 , or DRA+O 3 , to identify the components of the immune cell niche that contribute to AHR. Alveolar macrophages (AMs) had the greatest number of differentially expressed genes following DRA+O 3 , most of which were unique to the 2-hit exposure. Following DRA+O 3 , AMs activated transcriptional pathways related to cholesterol biosynthesis, degradation of the extracellular matrix, endosomal TLR processing, and various cytokine signals. We also identified AM and monocyte subset populations that were unique to the DRA+O 3 group. These unique AMs activated gene pathways related to inflammation, sphingolipid metabolism, and bronchial constriction. The unique monocyte population had a gene signature that suggested phospholipase activation and increased degradation of the extracellular matrix. Flow cytometry analysis of BAL immune cells showed recruited monocyte-derived AMs after DRA and DRA+O 3 , but not after O 3 exposure alone. O 3 alone increased BAL neutrophils but this response was attenuated in DRA+O 3 mice. DRA-induced changes in the airspace immune cell profile were reflected in elevated BAL cytokine/chemokine levels following DRA+O 3 compared to O 3 alone. The present work highlights the role of monocytes and AMs in the response to O 3 and suggests that the presence of distinct subpopulations following allergic inflammation may contribute to O 3 -induced AHR.
Collapse
|
18
|
Berdnikovs S, Newcomb DC, McKernan KE, Kuehnle SN, Haruna NF, Gebretsadik T, McKennan C, Ma S, Cephus JY, Rosas-Salazar C, Anderson LJ, Gern JE, Hartert T. Single cell profiling to determine influence of wheeze and early-life viral infection on developmental programming of airway epithelium. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.08.602506. [PMID: 39026695 PMCID: PMC11257436 DOI: 10.1101/2024.07.08.602506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Although childhood asthma is in part an airway epithelial disorder, the development of the airway epithelium in asthma is not understood. We sought to characterize airway epithelial developmental phenotypes in those with and without recurrent wheeze and the impact of infant infection with respiratory syncytial virus (RSV). Nasal airway epithelial cells (NAECs) were collected at age 2-3 years from an a priori designed nested birth cohort of children from four mutually exclusive groups of wheezers/non-wheezers and RSV-infected/uninfected in the first year of life. NAECs were cultured in air-liquid interface differentiation conditions followed by a combined analysis of single cell RNA sequencing (scRNA-seq) and in vitro infection with respiratory syncytial virus (RSV). NAECs from children with a wheeze phenotype were characterized by abnormal differentiation and basal cell activation of developmental pathways, plasticity in precursor differentiation and a delayed onset of maturation. NAECs from children with wheeze also had increased diversity of currently known RSV receptors and blunted anti-viral immune responses to in vitro infection. The most dramatic changes in differentiation of cultured epithelium were observed in NAECs derived from children that had both wheeze and RSV in the first year of life. Together this suggests that airway epithelium in children with wheeze is developmentally reprogrammed and characterized by increased barrier permeability, decreased antiviral response, and increased RSV receptors, which may predispose to and amplify the effects of RSV infection in infancy and susceptibility to other asthma risk factors that interact with the airway mucosa. SUMMARY Nasal airway epithelial cells from children with wheeze are characterized by altered development and increased susceptibility to RSV infection.
Collapse
|
19
|
Schmitt P, Duval A, Camus M, Lefrançais E, Roga S, Dedieu C, Ortega N, Bellard E, Mirey E, Mouton-Barbosa E, Burlet-Schiltz O, Gonzalez-de-Peredo A, Cayrol C, Girard JP. TL1A is an epithelial alarmin that cooperates with IL-33 for initiation of allergic airway inflammation. J Exp Med 2024; 221:e20231236. [PMID: 38597952 PMCID: PMC11010340 DOI: 10.1084/jem.20231236] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 02/07/2024] [Accepted: 03/11/2024] [Indexed: 04/11/2024] Open
Abstract
Epithelium-derived cytokines or alarmins, such as interleukin-33 (IL-33) and thymic stromal lymphopoietin (TSLP), are major players in type 2 immunity and asthma. Here, we demonstrate that TNF-like ligand 1A (TL1A) is an epithelial alarmin, constitutively expressed in alveolar epithelium at steady state in both mice and humans, which cooperates with IL-33 for early induction of IL-9high ILC2s during the initiation of allergic airway inflammation. Upon synergistic activation by IL-33 and TL1A, lung ILC2s acquire a transient IL-9highGATA3low "ILC9" phenotype and produce prodigious amounts of IL-9. A combination of large-scale proteomic analyses, lung intravital microscopy, and adoptive transfer of ILC9 cells revealed that high IL-9 expression distinguishes a multicytokine-producing state-of-activated ILC2s with an increased capacity to initiate IL-5-dependent allergic airway inflammation. Similar to IL-33 and TSLP, TL1A is expressed in airway basal cells in healthy and asthmatic human lungs. Together, these results indicate that TL1A is an epithelium-derived cytokine and an important cofactor of IL-33 in the airways.
Collapse
Affiliation(s)
- Pauline Schmitt
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Anais Duval
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Mylène Camus
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Emma Lefrançais
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Stéphane Roga
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Cécile Dedieu
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Nathalie Ortega
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Elisabeth Bellard
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Emilie Mirey
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Emmanuelle Mouton-Barbosa
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Odile Burlet-Schiltz
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Anne Gonzalez-de-Peredo
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Corinne Cayrol
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| | - Jean-Philippe Girard
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III—Paul Sabatier (UPS), Toulouse, France
| |
Collapse
|
20
|
Chen K, Han Y, Wang Y, Zhou D, Wu F, Cai W, Zheng S, Xiao Q, Zhang H, Li W. scMoresDB: A comprehensive database of single-cell multi-omics data for human respiratory system. iScience 2024; 27:109567. [PMID: 38617561 PMCID: PMC11015448 DOI: 10.1016/j.isci.2024.109567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/26/2023] [Accepted: 03/22/2024] [Indexed: 04/16/2024] Open
Abstract
The human respiratory system is a complex and important system that can suffer a variety of diseases. Single-cell sequencing technologies, applied in many respiratory disease studies, have enhanced our ability in characterizing molecular and phenotypic features at a single-cell resolution. The exponentially increasing data from these studies have consequently led to difficulties in data sharing and analysis. Here, we present scMoresDB, a single-cell multi-omics database platform with extensive omics types tailored for human respiratory diseases. scMoresDB re-analyzes single-cell multi-omics datasets, providing a user-friendly interface with cross-omics search capabilities, interactive visualizations, and analytical tools for comprehensive data sharing and integrative analysis. Our example applications highlight the potential significance of BSG receptor in SARS-CoV-2 infection as well as the involvement of HHIP and TGFB2 in the development and progression of chronic obstructive pulmonary disease. scMoresDB significantly increases accessibility and utility of single-cell data relevant to human respiratory system and associated diseases.
Collapse
Affiliation(s)
- Kang Chen
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong Province, China
| | - Yutong Han
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong Province, China
| | - Yanni Wang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong Province, China
| | - Dingli Zhou
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong Province, China
| | - Fanjie Wu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong Province, China
| | - Wenhao Cai
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong Province, China
| | - Shikang Zheng
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong Province, China
| | - Qinyuan Xiao
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong Province, China
| | - Haiyue Zhang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong Province, China
| | - Weizhong Li
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong Province, China
- Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, Guangdong Province, China
- Center for Precision Medicine, Sun Yat-sen University, Guangzhou 510080, Guangdong Province, China
| |
Collapse
|
21
|
Yang S, Gaietto K, Chen W. Mapping a New Course to Understand Lung Biology Mechanisms: LungMAP.net. Am J Respir Cell Mol Biol 2024; 70:91-93. [PMID: 38109690 PMCID: PMC10848696 DOI: 10.1165/rcmb.2023-0439ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 12/18/2023] [Indexed: 12/20/2023] Open
Affiliation(s)
- Sheng Yang
- Department of Biostatistics Nanjing Medical University Nanjing, Jiangsu, China
| | - Kristina Gaietto
- Department of Pediatrics University of Pittsburgh School of Medicine Pittsburgh, Pennsylvania
| | - Wei Chen
- Department of Pediatrics University of Pittsburgh School of Medicine Pittsburgh, Pennsylvania
| |
Collapse
|
22
|
Kiley JP, Brown MK, Freemer M, Lin S, Stefan M, Noel P. Research Accomplishments in Pulmonary, Critical Care, and Sleep: A Retrospective Review. Am J Respir Crit Care Med 2023; 208:1268-1270. [PMID: 37966304 PMCID: PMC10765391 DOI: 10.1164/rccm.202311-2047pp] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 11/15/2023] [Indexed: 11/16/2023] Open
Affiliation(s)
- James P Kiley
- Division of Lung Diseases, NHLBI, Bethesda, Maryland
| | | | | | - Sara Lin
- Division of Lung Diseases, NHLBI, Bethesda, Maryland
| | | | - Patricia Noel
- Division of Lung Diseases, NHLBI, Bethesda, Maryland
| |
Collapse
|