1
|
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
|
2
|
Li Q, Liao Y, Zeng J, Hu S, Li C, Whitsett JA, Zheng Y, Luo F, Xu C, He T, Lin X, Wan H. KLF5 Shapes Developing Respiratory Tubules by Inhibiting Actin Asymmetry in Epithelial Cells. Am J Respir Cell Mol Biol 2025; 72:663-677. [PMID: 39556252 DOI: 10.1165/rcmb.2024-0140oc] [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: 03/23/2024] [Accepted: 11/18/2024] [Indexed: 11/19/2024] Open
Abstract
Tubulogenesis depends on precise cell shape changes driven by asymmetric tension from the actin cytoskeleton. How actin asymmetry is dynamically controlled to coordinate epithelial cell shape changes required for respiratory tubulogenesis remains unknown. Herein, we unveiled a critical role for the transcription factor KLF5, regulating actin asymmetry, inducing epithelial cell shape changes by balancing RHOA and CDC42 GTPase activity via RICH2. Conditional Klf5 expression or deletion in pulmonary epithelial cells affected apical actin organization and the positioning of apical polarity proteins in cell membranes, disrupting branching and sacculation of respiratory tubules during mouse lung morphogenesis. Increased KLF5 concentrations were observed in epithelial cells lining dilated tubules in lungs from patients with congenital pulmonary airway malformation. Together, our results demonstrate that dynamic regulation of apical actin organization by KLF5 is essential for respiratory tubulogenesis, providing a mechanistic framework for comprehending the morphogenesis of respiratory tubules.
Collapse
Affiliation(s)
- Qing Li
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Greater Bay Area Institute of Precision Medicine (Guangzhou), Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Lung Inflammation and Injury, Shanghai, China
- Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University, Chengdu, China
- School of Life Sciences, Fudan University, Chengdu, China; and
| | - Yong Liao
- Chengdu Newgenegle Biotech Co. Ltd., Chengdu, China
| | - Junwei Zeng
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Greater Bay Area Institute of Precision Medicine (Guangzhou), Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Lung Inflammation and Injury, Shanghai, China
- Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University, Chengdu, China
- School of Life Sciences, Fudan University, Chengdu, China; and
| | - Silu Hu
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity and
| | - Chunjie Li
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity and
| | | | - Yi Zheng
- Division of Experimental Hematology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Fengming Luo
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity and
| | - Chang Xu
- Department of Pediatric Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Taozhen He
- Department of Pediatric Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xinhua Lin
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Greater Bay Area Institute of Precision Medicine (Guangzhou), Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Lung Inflammation and Injury, Shanghai, China
- Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University, Chengdu, China
- School of Life Sciences, Fudan University, Chengdu, China; and
| | - Huajing Wan
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity and
| |
Collapse
|
3
|
Zhao B, Socha J, Toth A, Fernandes S, Warheit-Niemi H, Ruff B, Khurana Hershey GK, VanDussen KL, Swarr D, Zacharias WJ. The Homeobox Transcription Factor CUX1 Coordinates Postnatal Epithelial Developmental Timing but Is Dispensable for Lung Organogenesis and Regeneration. Am J Respir Cell Mol Biol 2025; 72:678-687. [PMID: 39589256 DOI: 10.1165/rcmb.2024-0147oc] [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: 11/26/2024] [Indexed: 11/27/2024] Open
Abstract
Lung epithelial progenitors use a complex network of known and predicted transcriptional regulators to influence early lung development. In this study, we evaluated the function of one predicted regulator, CUX1, that we identified from transcriptional regulatory analysis of the SOX9+ distal lung progenitor network. We generated a new Cux1-floxed mouse model and created an epithelium-specific knockout of CUX1 using Shh-Cre (Cux1ShhCre-LOF). Postnatal Cux1ShhCre-LOF animals recapitulated key skin phenotypic features found in prior constitutive CUX1 knockout animals, confirming the functionality of our new floxed model. Postnatal Cux1ShhCre-LOF mice displayed subtle alveolar simplification and a transient delay in alveologenesis and alveolar type 1 cell development without persistent lung phenotypes. Cux1ShhCre-LOF mice developed failure to thrive in their second and third weeks of life because of delayed ileal maturation, which similarly resolves by Postnatal Day 35. Finally, we challenged Cux1ShhCre-LOF with influenza-mediated lung injury to demonstrate that Cux1ShhCre-LOF mice undergo productive alveolar regeneration that is indistinguishable from that in wild-type animals. Together, these findings indicate that epithelium-specific loss of CUX1 leads to transient developmental delays in the skin, lung, and intestine without defects in definitive organogenesis. We conclude that CUX1 function is required for temporal optimization of developmental maturation in multiple organs with implications for susceptibility windows in developmental disease pathogenesis.
Collapse
Affiliation(s)
- Barbara Zhao
- Perinatal Institute
- Division of Pulmonary Biology
- Division of Developmental Biology
- Medical Scientist Training Program
- Molecular and Developmental Biology Graduate Program
| | - Jacob Socha
- Division of Gastroenterology, Hepatology, and Nutrition, and
- Molecular and Developmental Biology Graduate Program
| | - Andrea Toth
- Perinatal Institute
- Division of Pulmonary Biology
- Division of Developmental Biology
- Medical Scientist Training Program
- Molecular and Developmental Biology Graduate Program
| | | | | | - Brandy Ruff
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; and
| | - Gurjit K Khurana Hershey
- Division of Asthma Research, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; and
- Department of Pediatrics
| | - Kelli L VanDussen
- Division of Developmental Biology
- Division of Gastroenterology, Hepatology, and Nutrition, and
- Department of Pediatrics
| | - Daniel Swarr
- Perinatal Institute
- Division of Pulmonary Biology
- Department of Pediatrics
| | - William J Zacharias
- Perinatal Institute
- Division of Pulmonary Biology
- Division of Developmental Biology
- Department of Pediatrics
- Division of Pulmonary and Critical Care Medicine, and
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
4
|
Sakurai M, Hosokawa S, Yamaguchi Y, Kirimura S, Ihara K, Ohashi K, Furukawa T, Sasano T, Kashimada K, Ishii T. Cyclopamine Attenuates Pulmonary Arterial Hypertension Development: Implications of Hedgehog Signaling Involvement for the Pathophysiology. FASEB J 2025; 39:e70628. [PMID: 40353829 DOI: 10.1096/fj.202403350r] [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: 12/24/2024] [Revised: 03/24/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Pulmonary arterial hypertension (PAH) is one of the most severe pulmonary diseases. Although combination therapies of the key drugs have improved survival rates, unmet needs remain in its management. We examined the effects of cyclopamine, a Hedgehog (HH) signaling inhibitor, as a potential novel therapeutic approach for PAH. C57BL/6J male mice were exposed to 10% oxygen for 3 weeks to induce pulmonary hypertension. One week after hypoxia exposure, these mice were treated with cyclopamine or vehicle. Cyclopamine significantly attenuated right ventricular (RV) systolic pressure (H + C: 31 mmHg vs. H: 38 mmHg, p < 0.01) and RV hypertrophy (H + C: 0.28 vs. H: 0.37, p < 0.01). The fully muscularized small pulmonary arteries significantly decreased with cyclopamine (H + C: 30% vs. H: 80%, p < 0.01), suggesting a mediation by vascular remodeling inhibition. In vitro, human pulmonary arterial smooth muscle cells (HPASMC) exposed to hypoxia revealed that the inhibitory action of cyclopamine was limited to hypoxia-promoted cell proliferation. In single-cell RNA sequencing analysis of mice lungs treated with cyclopamine, the signaling pathways of vascular smooth muscle contraction and cGMP-PKG, that is, key regulators in PAH development through vascular remodeling, were suppressed in cells with the characteristics of vascular endothelial and smooth muscle cells. RNA sequencing analysis of hypoxia-exposed hPASMCs revealed that the pathways related to extracellular matrix regulation were particularly recovered. Our animal model-based data revealed that HH signaling inhibition would improve PAH development by suppressing pulmonary vascular remodeling.
Collapse
MESH Headings
- Animals
- Veratrum Alkaloids/pharmacology
- Hedgehog Proteins/metabolism
- Male
- Mice
- Signal Transduction/drug effects
- Mice, Inbred C57BL
- Humans
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Pulmonary Arterial Hypertension/drug therapy
- Pulmonary Arterial Hypertension/metabolism
- Pulmonary Artery/drug effects
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/drug therapy
- Hypoxia
- Hypertrophy, Right Ventricular/metabolism
- Hypertrophy, Right Ventricular/drug therapy
- Vascular Remodeling/drug effects
- Cell Proliferation/drug effects
Collapse
Affiliation(s)
- Makito Sakurai
- Department of Pediatrics and Developmental Biology, Institute of Science Tokyo, Tokyo, Japan
| | - Susumu Hosokawa
- Department of Pediatrics and Developmental Biology, Institute of Science Tokyo, Tokyo, Japan
- Department of Pediatrics, Japanese Red Cross Musashino Hospital, Tokyo, Japan
| | - Yohei Yamaguchi
- Department of Pediatrics and Developmental Biology, Institute of Science Tokyo, Tokyo, Japan
| | - Susumu Kirimura
- Department of Pathology, Institute of Science Tokyo, Tokyo, Japan
| | - Kensuke Ihara
- Department of Cardiovascular Medicine, Institute of Science Tokyo, Tokyo, Japan
| | - Kenichi Ohashi
- Department of Human Pathology, Institute of Science Tokyo, Tokyo, Japan
| | - Tetsushi Furukawa
- Department of Bio-Informational Pharmacology, Medical Research Institute, Institute of Science Tokyo, Tokyo, Japan
| | - Tetsuo Sasano
- Department of Cardiovascular Medicine, Institute of Science Tokyo, Tokyo, Japan
| | - Kenichi Kashimada
- Department of Pediatrics and Developmental Biology, Institute of Science Tokyo, Tokyo, Japan
- Department of Pediatric Endocrinology, National Center for Child Health and Development, Tokyo, Japan
| | - Taku Ishii
- Department of Pediatrics and Developmental Biology, Institute of Science Tokyo, Tokyo, Japan
| |
Collapse
|
5
|
Mellors PW, Lange AN, Casino Remondo B, Shestov M, Planer JD, Peterson AR, Ying Y, Zhou S, Christie JD, Diamond JM, Cantu E, Basil MC, Gill S. Shared roles of immune and stromal cells in the pathogenesis of human bronchiolitis obliterans syndrome. JCI Insight 2025; 10:e176596. [PMID: 40232854 DOI: 10.1172/jci.insight.176596] [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: 09/20/2024] [Accepted: 04/11/2025] [Indexed: 04/17/2025] Open
Abstract
Bronchiolitis obliterans syndrome (BOS) is a progressive, fatal obstructive lung disease that occurs following lung transplant, where it is termed chronic lung allograft dysfunction BOS (CLAD-BOS), or as the primary manifestation of pulmonary chronic graft versus host disease (cGVHD-BOS) following allogeneic hematopoietic stem cell transplant. Disease pathogenesis is poorly understood; however, chronic alloreactivity is common to both conditions, suggesting a shared pathophysiology. We performed single-cell RNA-Seq (scRNA-Seq) on explanted human lungs from 4 patients with CLAD-BOS, 3 patients with cGVHD-BOS, and 3 deceased controls to identify cell types, genes, and pathways enriched in BOS to better understand disease mechanisms. In both forms of BOS, we found an expanded population of CD8+ tissue resident memory T cells (TRM), which was distinct to BOS compared with other chronic lung diseases. In addition, BOS samples expressed genes and pathways associated with macrophage chemotaxis and proliferation, including in nonimmune cell populations. We also identified dysfunctional stromal cells in BOS, characterized by pro- and antifibrotic gene programs. These data suggest substantial cellular and molecular overlap between CLAD- and cGVHD-BOS and, therefore, common pathways for possible therapeutic intervention.
Collapse
Affiliation(s)
- Patrick W Mellors
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania (Penn), Philadelphia, Pennsylvania, USA
- Center for Cellular Immunotherapies and
| | - Ana N Lange
- Department of Medicine, Perelman School of Medicine, Penn, Philadelphia, Pennsylvania, USA
- Penn-CHOP Lung Biology Institute and
- Penn Cardiovascular Institute, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | | | | | - Joseph D Planer
- Department of Medicine, Perelman School of Medicine, Penn, Philadelphia, Pennsylvania, USA
- Penn-CHOP Lung Biology Institute and
- Penn Cardiovascular Institute, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Andrew R Peterson
- Department of Medicine, Perelman School of Medicine, Penn, Philadelphia, Pennsylvania, USA
- Penn-CHOP Lung Biology Institute and
- Penn Cardiovascular Institute, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Yun Ying
- Department of Medicine, Perelman School of Medicine, Penn, Philadelphia, Pennsylvania, USA
- Penn-CHOP Lung Biology Institute and
- Penn Cardiovascular Institute, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Su Zhou
- Department of Medicine, Perelman School of Medicine, Penn, Philadelphia, Pennsylvania, USA
- Penn-CHOP Lung Biology Institute and
- Penn Cardiovascular Institute, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jason D Christie
- Department of Medicine, Perelman School of Medicine, Penn, Philadelphia, Pennsylvania, USA
- Penn-CHOP Lung Biology Institute and
| | - Joshua M Diamond
- Department of Medicine, Perelman School of Medicine, Penn, Philadelphia, Pennsylvania, USA
- Penn-CHOP Lung Biology Institute and
| | - Edward Cantu
- Penn-CHOP Lung Biology Institute and
- Department of Surgery, Perelman School of Medicine, Penn, Philadelphia, Pennsylvania, USA
| | - Maria C Basil
- Department of Medicine, Perelman School of Medicine, Penn, Philadelphia, Pennsylvania, USA
- Penn-CHOP Lung Biology Institute and
- Penn Cardiovascular Institute, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Saar Gill
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania (Penn), Philadelphia, Pennsylvania, USA
- Center for Cellular Immunotherapies and
| |
Collapse
|
6
|
Scherer M, Singh I, Braun MM, Szu-Tu C, Sanchez Sanchez P, Lindenhofer D, Jakobsen NA, Körber V, Kardorff M, Nitsch L, Kautz P, Rühle J, Bianchi A, Cozzuto L, Frömel R, Beneyto-Calabuig S, Lareau C, Satpathy AT, Beekman R, Steinmetz LM, Raffel S, Ludwig LS, Vyas P, Rodriguez-Fraticelli A, Velten L. Clonal tracing with somatic epimutations reveals dynamics of blood ageing. Nature 2025:10.1038/s41586-025-09041-8. [PMID: 40399669 DOI: 10.1038/s41586-025-09041-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 04/17/2025] [Indexed: 05/23/2025]
Abstract
Current approaches used to track stem cell clones through differentiation require genetic engineering1,2 or rely on sparse somatic DNA variants3,4, which limits their wide application. Here we discover that DNA methylation of a subset of CpG sites reflects cellular differentiation, whereas another subset undergoes stochastic epimutations and can serve as digital barcodes of clonal identity. We demonstrate that targeted single-cell profiling of DNA methylation5 at single-CpG resolution can accurately extract both layers of information. To that end, we develop EPI-Clone, a method for transgene-free lineage tracing at scale. Applied to mouse and human haematopoiesis, we capture hundreds of clonal differentiation trajectories across tens of individuals and 230,358 single cells. In mouse ageing, we demonstrate that myeloid bias and low output of old haematopoietic stem cells6 are restricted to a small number of expanded clones, whereas many functionally young-like clones persist in old age. In human ageing, clones with and without known driver mutations of clonal haematopoieis7 are part of a spectrum of age-related clonal expansions that display similar lineage biases. EPI-Clone enables accurate and transgene-free single-cell lineage tracing on hematopoietic cell state landscapes at scale.
Collapse
Affiliation(s)
- Michael Scherer
- Computational Biology and Health Genomics, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Indranil Singh
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Martina Maria Braun
- Computational Biology and Health Genomics, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Chelsea Szu-Tu
- Computational Biology and Health Genomics, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Pedro Sanchez Sanchez
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Dominik Lindenhofer
- Genome Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Niels Asger Jakobsen
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Verena Körber
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Michael Kardorff
- Department of Medicine, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Lena Nitsch
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
- Department of Biology, Chemistry, Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Pauline Kautz
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
- Technische Universität Berlin, Institute of Biotechnology, Berlin, Germany
| | - Julia Rühle
- Computational Biology and Health Genomics, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Agostina Bianchi
- Computational Biology and Health Genomics, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Luca Cozzuto
- Computational Biology and Health Genomics, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Robert Frömel
- Computational Biology and Health Genomics, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Sergi Beneyto-Calabuig
- Computational Biology and Health Genomics, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Caleb Lareau
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ansuman T Satpathy
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Renée Beekman
- Computational Biology and Health Genomics, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Lars M Steinmetz
- Genome Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Heidelberg/Mannheim, Heidelberg, Germany
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Genome Technology Center, Palo Alto, CA, USA
| | - Simon Raffel
- Department of Medicine, Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Leif S Ludwig
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
| | - Paresh Vyas
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Alejo Rodriguez-Fraticelli
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| | - Lars Velten
- Computational Biology and Health Genomics, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain.
- Universitat Pompeu Fabra (UPF), Barcelona, Spain.
| |
Collapse
|
7
|
Hu H, Fan Y, Wang J, Zhang J, Lyu Y, Hou X, Cui J, Zhang Y, Gao J, Zhang T, Nan K. Single-cell technology for cell-based drug delivery and pharmaceutical research. J Control Release 2025; 381:113587. [PMID: 40032008 DOI: 10.1016/j.jconrel.2025.113587] [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: 10/16/2024] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/05/2025]
Abstract
Leveraging the capacity to precisely manipulate and analyze individual cells, single-cell technology has rapidly become an indispensable tool in the advancement of cell-based drug delivery systems and innovative cell therapies. This technology offers powerful means to address cellular heterogeneity and significantly enhance therapeutic efficacy. Recent breakthroughs in techniques such as single-cell electroporation, mechanical perforation, and encapsulation, particularly when integrated with microfluidics and bioelectronics, have led to remarkable improvements in drug delivery efficiency, reductions in cytotoxicity, and more precise targeting of therapeutic effects. Moreover, single-cell analyses, including advanced sequencing and high-resolution sensing, offer profound insights into complex disease mechanisms, the development of drug resistance, and the intricate processes of stem cell differentiation. This review summarizes the most significant applications of these single-cell technologies, highlighting their impact on the landscape of modern biomedicine. Furthermore, it provides a forward-looking perspective on future research directions aimed at further optimizing drug delivery strategies and enhancing therapeutic outcomes in the treatment of various diseases.
Collapse
Affiliation(s)
- Huihui Hu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China
| | - Yunlong Fan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China; MicroTech Medical (Hangzhou) Co., Hangzhou 311100, China
| | - Jiawen Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China
| | - Jialu Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China
| | - Yidan Lyu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China
| | - Xiaoqi Hou
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Jizhai Cui
- Department of Materials Science, Fudan University, Shanghai 200438, China; International Institute of Intelligent Nanorobots and Nanosystems, Fudan University, Shanghai 200438, China
| | - Yamin Zhang
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 117585, Singapore
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China
| | - Tianyuan Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China.
| | - Kewang Nan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China.
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Polkoff KM, Lampe R, Gupta NK, Murphy Y, Chung J, Carter A, Simon JM, Gleason K, Moatti A, Murthy PK, Edwards L, Greenbaum A, Tata A, Tata PR, Piedrahita JA. Novel Porcine Model Reveals Two Distinct LGR5 Cell Types during Lung Development and Homeostasis. Am J Respir Cell Mol Biol 2025; 72:496-509. [PMID: 39499850 PMCID: PMC12051919 DOI: 10.1165/rcmb.2024-0040oc] [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: 11/05/2024] [Indexed: 11/07/2024] Open
Abstract
Cells expressing leucine-rich repeat-containing G-protein-coupled receptor 5 (LGR5) play a pivotal role in homeostasis, repair, and regeneration in multiple organs, including skin and gastrointestinal tract, but little is known about their role in the lung. Findings from mice, a widely used animal model, suggest that lung LGR5 expression differs from that of humans. In this work, using a new transgenic pig model, we identify two main populations of LGR5+ cells in the lung that are conserved in human but not mouse lungs. Using RNA sequencing, three-dimensional imaging, and organoid models, we determine that in the fetal lung, epithelial LGR5 expression is transient in a subpopulation of SOX9+/ETV5+/SFTPC+ progenitor lung tip cells. In contrast, epithelial LGR5 expression is absent from postnatal lung but is reactivated in bronchioalveolar organoids derived from basal airway cells. We also describe a separate population of mesenchymal LGR5+ cells that surrounds developing and mature airways, lies adjacent to airway basal cells, and is closely associated with nerve fibers. Transcriptionally, mesenchymal LGR5+ cells include a subset of peribronchial fibroblasts that express unique patterns of SHH, FGF, WNT, and TGF-β signaling pathway genes. These results support distinct roles for LGR5+ cells in the lung and describe a physiologically relevant animal model for further studies on the function of these cells in repair and regeneration.
Collapse
Affiliation(s)
- Kathryn M. Polkoff
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, and
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
| | - Ross Lampe
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, and
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
| | - Nithin K. Gupta
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
- School of Osteopathic Medicine, Campbell University, Lillington, North Carolina
| | - Yanet Murphy
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, and
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
| | - Jaewook Chung
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, and
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
| | - Amber Carter
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, and
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
| | - Jeremy M. Simon
- Department of Genetics, School of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Katherine Gleason
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, and
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
| | - Adele Moatti
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh and Chapel Hill, North Carolina; and
| | - Preetish K. Murthy
- Department of Cell Biology, School of Medicine, Duke University, Durham, North Carolina
| | - Laura Edwards
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, and
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
| | - Alon Greenbaum
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh and Chapel Hill, North Carolina; and
| | - Aleksandra Tata
- Department of Cell Biology, School of Medicine, Duke University, Durham, North Carolina
| | - Purushothama Rao Tata
- Department of Cell Biology, School of Medicine, Duke University, Durham, North Carolina
| | - Jorge A. Piedrahita
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, and
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina
| |
Collapse
|
11
|
Varricchi G, Poto R, Criscuolo G, Strisciuglio C, Nair P, Marone G. TL1A, a novel alarmin in airway, intestinal, and autoimmune disorders. J Allergy Clin Immunol 2025; 155:1420-1434. [PMID: 40010414 DOI: 10.1016/j.jaci.2025.02.018] [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: 12/11/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 02/28/2025]
Abstract
The term alarmin denotes a broad class of molecules rapidly released to alert the immune system through the engagement of specific receptors on immune cells. Three alarmin cytokines-thymic stromal lymphopoietin, IL-33, and IL-25-are released from epithelial and certain stromal cells. TNF-like cytokine 1A (TL1A) is a member of the TNF cytokine superfamily, first identified in human endothelial cells. TL1A is now considered a novel alarmin expressed by human and mouse bronchial and intestinal epithelial cells. TL1A exerts its biological activities by binding to a trimeric receptor DR3 (death receptor 3), expressed on a wide spectrum of immune and structural cells, including lung fibroblasts, endothelial cells, and bronchial epithelial cells. TL1A has been implicated in experimental and human inflammatory bowel diseases as well as in airway inflammation and remodeling in severe asthma. A monoclonal antibody anti-TL1A (tulisokibart) is effective in inducing clinical remission in ulcerative colitis patients. Increasing evidence suggests that TL1A is also involved in certain autoimmune disorders, such as rheumatoid arthritis and psoriasis. These emerging findings broaden the role of TL1A in various human inflammatory conditions. Several clinical trials are currently evaluating the safety and efficacy of monoclonal antibodies targeting TL1A in asthma or inflammatory bowel disease patients.
Collapse
Affiliation(s)
- Gilda Varricchi
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy; World Allergy Organization (WAO) Center of Excellence (CoE), Naples, Italy; Istituto Endotipi in Oncologia, Metabolismo e Immunologia "G. Salvatore" (IEOMI), National Research Council (CNR), Naples, Italy.
| | - Remo Poto
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy; World Allergy Organization (WAO) Center of Excellence (CoE), Naples, Italy; Istituti Clinici Scientifici Maugeri-IRCCS Scientific Institute of Telese Terme, Benevento, Italy
| | - Gjada Criscuolo
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy; World Allergy Organization (WAO) Center of Excellence (CoE), Naples, Italy
| | - Caterina Strisciuglio
- Department of Woman, Child and General and Specialistic Surgery, University of Campania "L. Vanvitelli," Naples, Italy
| | - Parameswaran Nair
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada; Research Institute of St Joe's Hamilton, St Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
| | - Gianni Marone
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy; Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy; World Allergy Organization (WAO) Center of Excellence (CoE), Naples, Italy
| |
Collapse
|
12
|
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
|
13
|
Pearce EM, Evans E, Mayday MY, Reyes G, Simon MR, Blum J, Kim H, Mu J, Shaw PJ, Rowan CM, Auletta JJ, Martin PL, Hurley C, Kreml EM, Qayed M, Abdel-Azim H, Keating AK, Cuvelier GDE, Hume JR, Killinger JS, Godder K, Hanna R, Duncan CN, Quigg TC, Castillo P, Lalefar NR, Fitzgerald JC, Mahadeo KM, Satwani P, Moore TB, Hanisch B, Abdel-Mageed A, Davis DB, Hudspeth MP, Yanik GA, Pulsipher MA, Dvorak CCJL, Zinter MS. Integrating Pulmonary and Systemic Transcriptomic Profiles to Characterize Lung Injury after Pediatric Hematopoietic Stem Cell Transplant. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.31.25324969. [PMID: 40236411 PMCID: PMC11998824 DOI: 10.1101/2025.03.31.25324969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Hematopoietic stem cell transplantation (HCT) is potentially curative for numerous malignant and non-malignant diseases but can lead to lung injury due to chemoradiation toxicity, infection, and immune dysregulation. Bronchoalveolar lavage (BAL) is the most commonly used procedure for diagnostic sampling of the lung but is invasive, cannot be performed in medically fragile patients, and is challenging to perform serially. We previously showed that BAL transcriptomes representing pulmonary inflammation and cellular injury can phenotype post-HCT lung injury and predict mortality outcomes. However, whether peripheral blood testing is a suitable minimally-invasive surrogate for pulmonary sampling in the HCT population remains unknown. To address this question, we compared 210 paired BAL and peripheral blood transcriptomes obtained from 166 pediatric HCT patients at 27 children's hospitals. BAL and blood mRNA abundance showed minimal overall correlation at the level of individual genes, gene set enrichment scores, imputed cell fractions, and T- and B-cell receptor clonotypes. Instead, we identified significant site-specific transcriptional programs. In BAL, expression of innate and adaptive immune pathways was tightly co-regulated with expression of epithelial mesenchymal transition and hypoxia pathways, and these signatures were associated with mortality. In contrast, in blood, expression of endothelial injury, DNA repair, and cellular metabolism pathways was associated with mortality. Integration of paired BAL and blood transcriptomes dichotomized patients into two groups, of which one group showed twice the rate of hypoxia and significantly worse outcomes within 7 days of enrollment. These findings reveal a compartmentalized injury response, where BAL and peripheral blood transcriptomes provide distinct but complementary insights into local and systemic mechanisms of post-HCT lung injury.
Collapse
Affiliation(s)
- Emma M Pearce
- Division of Critical Care Medicine, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Erica Evans
- Division of Critical Care Medicine, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Madeline Y Mayday
- Division of Critical Care Medicine, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
- Departments of Laboratory Medicine and Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Gustavo Reyes
- Division of Critical Care Medicine, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Miriam R Simon
- Division of Critical Care Medicine, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Jacob Blum
- Division of Critical Care Medicine, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Hanna Kim
- Division of Critical Care Medicine, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Jessica Mu
- Division of Critical Care Medicine, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Peter J Shaw
- The Children`s Hospital at Westmead, Westmead, NSW, Australia
| | - Courtney M Rowan
- Indiana University, Department of Pediatrics, Division of Critical Care Medicine, Indianapolis, IN, USA
| | - Jeffrey J Auletta
- Hematology/Oncology/BMT and Infectious Diseases, Nationwide Children's Hospital, Columbus, OH, USA
- CIBMTR (Center for International Blood and Marrow Transplant Research), National Marrow Donor Program/Be The Match, Minneapolis, MN, USA
| | - Paul L Martin
- Division of Pediatric and Cellular Therapy, Duke University Medical Center, Durham, NC, USA
| | - Caitlin Hurley
- Division of Critical Care, Department of Pediatric Medicine, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Erin M Kreml
- Department of Child Health, Division of Critical Care Medicine, University of Arizona, Phoenix, AZ, USA
| | - Muna Qayed
- Aflac Cancer & Blood Disorders Center, Children's Healthcare of Atlanta and Emory University, Atlanta, GA, USA
| | - Hisham Abdel-Azim
- Department of Pediatrics, Division of Hematology/Oncology and Transplant and Cell Therapy, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Loma Linda University School of Medicine, Cancer Center, Children Hospital and Medical Center, Loma Linda, CA, USA
| | - Amy K Keating
- Harvard Medical School, Boston, Massachusetts; Division of Pediatric Oncology, Department of Pediatrics, Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, MA, USA
- Center for Cancer and Blood Disorders, Children's Hospital Colorado and University of Colorado, Aurora, CO, USA
| | - Geoffrey D E Cuvelier
- CancerCare Manitoba, Manitoba Blood and Marrow Transplant Program, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Janet R Hume
- University of Minnesota, Department of Pediatrics, Division of Critical Care Medicine, Minneapolis, MN, USA
| | - James S Killinger
- Division of Pediatric Critical Care, Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA
| | - Kamar Godder
- Cancer and Blood Disorders Center, Nicklaus Children's Hospital, Miami, FL, USA
| | - Rabi Hanna
- Department of Pediatric Hematology, Oncology and Blood and Marrow Transplantation, Pediatric Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Christine N Duncan
- Harvard Medical School, Boston, Massachusetts; Division of Pediatric Oncology, Department of Pediatrics, Dana-Farber Cancer Institute and Boston Children's Hospital, Boston, MA, USA
| | - Troy C Quigg
- Pediatric Blood and Marrow Transplantation Program, Texas Transplant Institute, Methodist Children's Hospital, San Antonio, TX, USA
- Section of Pediatric BMT and Cellular Therapy, Helen DeVos Children's Hospital, Grand Rapids, MI, USA
| | - Paul Castillo
- University of Florida, Gainesville, UF Health Shands Children's Hospital, Gainesville, FL, USA
| | - Nahal R Lalefar
- Division of Pediatric Hematology/Oncology, UCSF Benioff Children's Hospital Oakland, University of California San Francisco, Oakland, CA, USA
| | - Julie C Fitzgerald
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
| | - Kris M Mahadeo
- Department of Pediatrics, Division of Hematology/Oncology, MD Anderson Cancer Center, Houston, TX, USA
- Division of Pediatric and Cellular Therapy, Duke University Medical Center, Durham, NC, USA
| | - Prakash Satwani
- Division of Pediatric Hematology, Oncology and Stem Cell Transplantation, Department of Pediatrics, Columbia University, New York, NY, USA
| | - Theodore B Moore
- Department of Pediatric Hematology-Oncology, Mattel Children's Hospital, University of California, Los Angeles, CA, USA
| | - Benjamin Hanisch
- Children's National Hospital, Washington, District of Columbia, USA
| | - Aly Abdel-Mageed
- Section of Pediatric BMT and Cellular Therapy, Helen DeVos Children's Hospital, Grand Rapids, MI, USA
| | - Dereck B Davis
- Department of Pediatrics, Hematology/Oncology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Michelle P Hudspeth
- Adult and Pediatric Blood & Marrow Transplantation, Pediatric Hematology/Oncology, Medical University of South Carolina Children's Hospital/Hollings Cancer Center, Charleston, SC, USA
| | - Greg A Yanik
- Pediatric Blood and Bone Marrow Transplantation, Michigan Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Michael A Pulsipher
- Division of Pediatric Hematology and Oncology, Intermountain Primary Children's Hospital, Huntsman Cancer Institute, Spencer Fox Eccles School of Medicine at the University of Utah, Salt Lake City, UT, USA
| | - Christopher C Joseph L Dvorak
- Division of Allergy, Immunology, and Bone Marrow Transplantation, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Matt S Zinter
- Division of Critical Care Medicine, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
- Division of Allergy, Immunology, and Bone Marrow Transplantation, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
14
|
Kalailingam P, Ngan SC, Iyappan R, Nehchiri A, Mohd‐Kahliab K, Lee BST, Sharma B, Machan R, Bo ST, Chambers ES, Fajardo VA, Macpherson REK, Liu J, Klentrou P, Tsiani EL, Lim KL, Su IH, Gao Y, Richar AM, Kalaria RN, Chen CP, Balion C, de Kleijn D, McCarthy NE, Sze SK. Immunotherapeutic targeting of aging-associated isoDGR motif in chronic lung inflammation. Aging Cell 2025; 24:e14425. [PMID: 39757428 PMCID: PMC11984686 DOI: 10.1111/acel.14425] [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] [Revised: 08/18/2024] [Accepted: 11/04/2024] [Indexed: 01/07/2025] Open
Abstract
Accumulation of damaged biomolecules in body tissues is the primary cause of aging and age-related chronic diseases. Since this damage often occurs spontaneously, it has traditionally been regarded as untreatable, with typical therapeutic strategies targeting genes or enzymes being ineffective in this domain. In this report, we demonstrate that an antibody targeting the isoDGR damage motif in lung tissue can guide immune clearance of harmful damaged proteins in vivo, effectively reducing age-linked lung inflammation. We observed age-dependent accumulation of the isoDGR motif in human lung tissues, as well as an 8-fold increase in isoDGR-damaged proteins in lung fibrotic tissues compared with healthy tissue. This increase was accompanied by marked infiltration of CD68+/CD11b + macrophages, consistent with a role for isoDGR in promoting chronic inflammation. We therefore assessed isoDGR function in mice that were either naturally aged or lacked the isoDGR repair enzyme. IsoDGR-protein accumulation in mouse lung tissue was strongly correlated with chronic inflammation, pulmonary edema, and hypoxemia. This accumulation also induced mitochondrial and ribosomal dysfunction, in addition to features of cellular senescence, thereby contributing to progressive lung damage over time. Importantly, treatment with anti-isoDGR antibody was able to reduce these molecular features of disease and significantly reduced lung pathology in vivo.
Collapse
Affiliation(s)
- Pazhanichamy Kalailingam
- Center for Genomic MedicineMassachusetts General Hospital and Harvard Medical SchoolBostonMassachusettsUSA
- School of Biological SciencesNanyang Technological UniversitySingaporeSingapore
| | - SoFong Cam Ngan
- Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | - Ranjith Iyappan
- Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | - Afra Nehchiri
- Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | | | | | - Bhargy Sharma
- School of Biological SciencesNanyang Technological UniversitySingaporeSingapore
| | - Radek Machan
- SCELSENanyang Technological UniversitySingaporeSingapore
| | - Sint Thida Bo
- School of Biological SciencesNanyang Technological UniversitySingaporeSingapore
| | - Emma S. Chambers
- Centre for Immunobiology, the Blizard Institute, Bart's and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Val A. Fajardo
- Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | | | - Jian Liu
- Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | - Panagiota Klentrou
- Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | | | - Kah Leong Lim
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingaporeSingapore
| | - I. Hsin Su
- School of Biological SciencesNanyang Technological UniversitySingaporeSingapore
| | - Yong‐Gui Gao
- School of Biological SciencesNanyang Technological UniversitySingaporeSingapore
| | - A. Mark Richar
- Cardiovascular Research InstituteNational University Health SystemSingaporeSingapore
| | - Raj N. Kalaria
- Institute of Neuroscience, Campus for Ageing and VitalityNewcastle UniversityNewcastle upon TyneUK
| | - Christopher P. Chen
- Memory, Aging and Cognition CentreNational University Health SystemSingaporeSingapore
| | - Cynthia Balion
- Department of Pathology and Molecular MedicineMcMaster UniversityHamiltonOntarioCanada
| | | | - Neil E. McCarthy
- Centre for Immunobiology, the Blizard Institute, Bart's and the London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Siu Kwan Sze
- School of Biological SciencesNanyang Technological UniversitySingaporeSingapore
- Faculty of Applied Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| |
Collapse
|
15
|
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
|
16
|
Nguyen DH, Tian J, Shanahan SL, Wang CK, Jacks T, Wang X, Li P. A tissue-scale strategy for sensing threats in barrier organs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.19.644134. [PMID: 40166266 PMCID: PMC11957033 DOI: 10.1101/2025.03.19.644134] [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
Barrier organs rely on a limited set of pattern recognition receptors (PRRs) to detect diverse immunogenic challenges. How organs assess threats and adjust immune responses to balance host protection with collateral tissue damage remains unclear. Here, by analyzing influenza infection in the lung using single-molecule imaging and spatial transcriptomics, we discovered a tiered threat-sensing strategy at the tissue scale, where the probability of detecting and responding to infection is lowest in the outermost epithelia and highest in the inner stroma. This strategy emerges from spatially graded PRR expression that results in cell-type-specific probabilities of threat-sensing across the tissue, a design broadly adopted by barrier organs. Selectively increasing PRR expression in lung epithelia in vivo exacerbated tissue damage upon inflammatory challenge. These results reveal a spatially tiered strategy to tolerate threats restricted within the epithelia, and yet enable progressively potent immune responses as threats invade deeper into the tissue.
Collapse
|
17
|
Rizzoli E, Fievez L, Fastrès A, Roels E, Marichal T, Clercx C. A single-cell RNA sequencing atlas of the healthy canine lung: a foundation for comparative studies. Front Immunol 2025; 16:1501603. [PMID: 40114924 PMCID: PMC11922831 DOI: 10.3389/fimmu.2025.1501603] [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: 09/25/2024] [Accepted: 02/13/2025] [Indexed: 03/22/2025] Open
Abstract
Single cell RNA sequencing (scRNA-seq) can be used to resolve the cellular and molecular heterogeneity within a tissue by identifying cell populations with an unprecedented granularity along with their transcriptional signatures. Yet, the single cell gene expression profiles of cell populations in the healthy canine lung tissue remain unexplored and such analysis could reveal novel cell populations or markers lacking in dogs and facilitate comparisons with lung diseases. Using fresh healthy lung biopsies from four dogs, we conducted droplet-based scRNA-seq on 26,278 cells. We characterized 46 transcriptionally distinct cell subpopulations across all lung tissue compartments including 23 immune, 13 mesenchymal, five epithelial and five endothelial cell subpopulations. Of note, we captured rare cells such as unconventional T cells or Schwann cells. Differential gene expression profiles identified specific markers across all cell subpopulations. Fibroblasts clusters exhibited a marked transcriptional heterogeneity, some of which might exert immune regulatory functions. Finally, the integration of canine lung cells with an annotated human lung atlas highlighted many similarities in gene expression profiles between species. This study thus provides an extensive molecular cell atlas of the healthy canine lung, expanding our knowledge of lung cell diversity in dogs, and providing the molecular foundation for investigating lung cell identities and functions in canine lung diseases. Besides, the occurrence of spontaneous lung diseases in pet dogs, with phenotypes closely resembling those in humans, may provide a relevant model for advancing research into human lung diseases.
Collapse
Affiliation(s)
- Elodie Rizzoli
- Department of Companion Animals Clinical Sciences, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Laurence Fievez
- Department of Functional Sciences, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
- Laboratory of Cellular and Molecular Immunology, GIGA Institute, University of Liège, Liège, Belgium
| | - Aline Fastrès
- Department of Companion Animals Clinical Sciences, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Elodie Roels
- Department of Companion Animals Clinical Sciences, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Thomas Marichal
- Department of Functional Sciences, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
- Laboratory of Immunophysiology, GIGA Institute, University of Liège, Liège, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO) Department, WEL Research Institute, Wavre, Belgium
| | - Cécile Clercx
- Department of Companion Animals Clinical Sciences, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| |
Collapse
|
18
|
Kim DK, Synn CB, Lee W, Jo HN, Lee CY, Lee S, Hwang JY, Kim Y, Kang SS, Baek S, Na K, Yang SM, Kim MH, Han H, Han YJ, Kim JH, Park SY, Park YJ, Lee GT, Choi SJ, Sohn JO, Ye SK, Lee JB, Lim SM, Hong MH, Pyo KH, Cho BC. Denfivontinib Activates Effector T Cells Through the NLRP3 Inflammasome, Yielding Potent Anticancer Effects by Combination with Pembrolizumab. Mol Cancer Ther 2025; 24:354-369. [PMID: 39632711 PMCID: PMC11876964 DOI: 10.1158/1535-7163.mct-24-0501] [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: 06/23/2024] [Revised: 08/30/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
Various combination therapies have been investigated to overcome the limitations of using immune checkpoint inhibitors. However, determining the optimal combination therapy remains challenging. To overcome the therapeutic limitation, we conducted a translational research to elucidate the mechanisms by which AXL inhibition enhances antitumor effects when combined with anti-PD-1 antibody therapy. Herein, we demonstrated improved antitumor effects through combination treatment with denfivontinib and pembrolizumab which resulted in enhanced differentiation into effector CD4+ and CD8+ memory T cells, accompanied by an increase in IFN-γ expression in the YHIM-2004 xenograft model derived from patients with non-small cell lung cancer. Concurrently, a reduction in the number of immunosuppressive M2 macrophages and myeloid-derived suppressor cells was observed. Mechanistically, denfivontinib potentiated the NOD-like receptor pathway, thereby facilitating NLRP3 inflammasome formation. This leads to macrophage activation via NF-κB signaling pathway activation. We have confirmed that the positive interaction between macrophages and T cells arises from the enhanced antigen-presenting machinery of activated macrophages. Furthermore, the observed tumor effects in AXL knockout mice confirmed that AXL inhibition by denfivontinib enhances the antitumor effects, thus opening new avenues for therapeutic interventions aimed at overcoming limitations in immunotherapy. To demonstrate the extent to which our findings reflect clinical results, we analyzed bulk RNA sequencing data from 21 patients with non-small cell lung cancer undergoing anti-PD-1 immunotherapy. The NLRP3 inflammasome score influenced enhanced immune responses in patient data undergoing anti-PD-1 immunotherapy, suggesting a role for the NLRP3 inflammasome in activating immune responses during treatment.
Collapse
Affiliation(s)
- Dong Kwon Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Chun-Bong Synn
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Wongeun Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ha-Ni Jo
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Chai Young Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seul Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Joon Yeon Hwang
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Youngtaek Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seong-san Kang
- JEUK Institute for Cancer Research, JEUK Co., Ltd., Gumi-City, Republic of Korea
| | - Sujeong Baek
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kwangmin Na
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung Min Yang
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Mi Hyun Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Heekyung Han
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yu Jin Han
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae Hwan Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - So Young Park
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young Joon Park
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Gang-Taik Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Su-Jin Choi
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jie-Ohn Sohn
- Wide River Institute of Immunology, Seoul National University, Hongcheon, Republic of Korea
| | - Sang-Kyu Ye
- Wide River Institute of Immunology, Seoul National University, Hongcheon, Republic of Korea
- Department of Pharmacology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jii Bum Lee
- Division of Medical Oncology, Department of Internal Medicine and Yonsei Cancer Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sun Min Lim
- Division of Medical Oncology, Department of Internal Medicine and Yonsei Cancer Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Min Hee Hong
- Division of Medical Oncology, Department of Internal Medicine and Yonsei Cancer Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyoung-Ho Pyo
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
- Yonsei New Il Han Institute for Integrative Lung Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Research Support, Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Byoung Chul Cho
- Division of Medical Oncology, Department of Internal Medicine and Yonsei Cancer Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
- Yonsei New Il Han Institute for Integrative Lung Cancer Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
19
|
Shorey-Kendrick LE, Crosland BA, Schabel MC, Messaoudi I, Guo M, Drake MG, Nie Z, Edenfield RC, Cinco I, Davies MH, Graham JA, Hagen OL, McCarty OJT, McEvoy CT, Spindel ER, Lo JO. Effects of maternal edible THC consumption on offspring lung growth and function in a rhesus macaque model. Am J Physiol Lung Cell Mol Physiol 2025; 328:L463-L477. [PMID: 39903192 DOI: 10.1152/ajplung.00360.2024] [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: 11/07/2024] [Revised: 12/19/2024] [Accepted: 01/30/2025] [Indexed: 02/06/2025] Open
Abstract
Prenatal cannabis use is rising, in part due to legalization and perceptions of safety. The impact of prenatal cannabis exposure on offspring development, especially respiratory health, remains largely unknown. The objective of this study was to determine whether in utero exposure to delta-9-tetrahydrocannabinol (THC), the main psychoactive component of cannabis, is deleterious to offspring lung development and function using a rhesus macaque model. Female rhesus macaques received a daily edible containing either THC (2.5 mg/7 kg/day, equivalent to a heavy medical cannabis dose) or placebo during gestation and postnatally. Serial in utero magnetic resonance imaging (MRI) was performed during pregnancy at approximately gestational days (G)110 and G150. At 6 mo of age, infants underwent pulmonary function testing, followed by tissue collection for molecular analysis (bulk RNAseq, whole genome bisulfite sequencing, and spatial RNAseq). THC-exposed infants displayed significantly reduced forced residual capacity, which correlated with nonsignificant decreases in total lung capacity, lung diffusion capacity and lower fetal lung perfusion, oxygen availability, and lung volume measured by MRI. Consistent with these decreases in volume indices, levels of pulmonary growth factors were decreased in bronchial alveolar lavage at 6 mo. Molecular analysis of infant lungs revealed altered epigenetic regulation of gene expression, including at genes involved in extracellular matrix organization and lung development, and activation of immune signaling. Our study suggests that exposure to prenatal edible THC alters epigenetic regulation of lung gene expression and may negatively affect offspring lung development and function. Data from this study will help guide healthcare provider counseling on cannabis use in pregnancy.NEW & NOTEWORTHY In a translational rhesus macaque model, chronic prenatal delta-9-tetrahydrocannabinol exposure resulted in decreased lung volumes in offspring measured at 6 mo of age. These decreases correlated with altered DNA methylation in the lung, including at genes involved in extracellular matrix organization, lung development, and activation of immune signaling, and changes in lung cell composition as measured by spatial transcriptomics. These findings add to the growing evidence that prenatal cannabis exposure may adversely affect offspring development.
Collapse
Affiliation(s)
- Lyndsey E Shorey-Kendrick
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States
| | - B Adam Crosland
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, Oregon, United States
| | - Matthias C Schabel
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon, United States
| | - Ilhem Messaoudi
- Department of Microbiology, Immunology, and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, Kentucky, United States
| | - Minzhe Guo
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
| | - Matthew G Drake
- Division of Pulmonary and Critical Care Medicine, Oregon Health & Science University, Portland, Oregon, United States
| | - Zhenying Nie
- Division of Pulmonary and Critical Care Medicine, Oregon Health & Science University, Portland, Oregon, United States
| | - R Clayton Edenfield
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Sciences University, Beaverton, Oregon, United States
| | - Issac Cinco
- Department of Microbiology, Immunology, and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, Kentucky, United States
| | - Michael H Davies
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States
| | - Jason A Graham
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Sciences University, Beaverton, Oregon, United States
| | - Olivia L Hagen
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Sciences University, Beaverton, Oregon, United States
| | - Owen J T McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, United States
| | - Cindy T McEvoy
- Division of Neonatology, Department of Pediatrics, Oregon Health & Science University, Portland, Oregon, United States
| | - Eliot R Spindel
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States
| | - Jamie O Lo
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, Oregon, United States
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Sciences University, Beaverton, Oregon, United States
| |
Collapse
|
20
|
Joulia R, Lloyd CM. Location, Location, Location: Spatial Immune-Stroma Crosstalk Drives Pathogenesis in Asthma. Immunol Rev 2025; 330:e70013. [PMID: 39991870 PMCID: PMC11848993 DOI: 10.1111/imr.70013] [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: 10/08/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 02/25/2025]
Abstract
Chronic lung diseases including asthma are characterized by an abnormal immune response and active tissue remodeling. These changes in the architecture of the tissue are a fundamental part of the pathology across the life course of patients suffering from asthma. Current treatments aim at dampening the immune system hyperactivation, but effective drugs targeting stromal or acellular structures are still lacking. This is mainly due to the lack of a detailed understanding of the composition of the large airways and the cellular interactions taking place in this niche. We and others have revealed multiple aspects of the spatial architecture of the airway wall in response to airborne insults. In this review, we discuss four elements that we believe should be the focus of future asthma research across the life course, to increase understanding and improve therapies: (i) specialized lung niches, (ii) the 3D architecture of the epithelium, (iii) the extracellular matrix, and (iv) the vasculature. These components comprise the main stromal structures at the airway wall, each playing a key role in the development of asthma and directing the immune response. We summarize promising future directions that will enhance lung research, ultimately benefiting patients with asthma.
Collapse
Affiliation(s)
- Régis Joulia
- National Heart and Lung InstituteImperial College LondonLondonUK
| | - Clare M. Lloyd
- National Heart and Lung InstituteImperial College LondonLondonUK
| |
Collapse
|
21
|
Vannan A, Lyu R, Williams AL, Negretti NM, Mee ED, Hirsh J, Hirsh S, Hadad N, Nichols DS, Calvi CL, Taylor CJ, Polosukhin VV, Serezani APM, McCall AS, Gokey JJ, Shim H, Ware LB, Bacchetta MJ, Shaver CM, Blackwell TS, Walia R, Sucre JMS, Kropski JA, McCarthy DJ, Banovich NE. Spatial transcriptomics identifies molecular niche dysregulation associated with distal lung remodeling in pulmonary fibrosis. Nat Genet 2025; 57:647-658. [PMID: 39901013 PMCID: PMC11906353 DOI: 10.1038/s41588-025-02080-x] [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: 01/22/2024] [Accepted: 01/08/2025] [Indexed: 02/05/2025]
Abstract
Large-scale changes in the structure and cellular makeup of the distal lung are a hallmark of pulmonary fibrosis (PF), but the spatial contexts that contribute to disease pathogenesis have remained uncertain. Using image-based spatial transcriptomics, we analyzed the gene expression of 1.6 million cells from 35 unique lungs. Through complementary cell-based and innovative cell-agnostic analyses, we characterized the localization of PF-emergent cell types, established the cellular and molecular basis of classical PF histopathologic features and identified a diversity of distinct molecularly defined spatial niches in control and PF lungs. Using machine learning and trajectory analysis to segment and rank airspaces on a gradient of remodeling severity, we identified compositional and molecular changes associated with progressive distal lung pathology, beginning with alveolar epithelial dysregulation and culminating with changes in macrophage polarization. Together, these results provide a unique, spatially resolved view of PF and establish methods that could be applied to other spatial transcriptomic studies.
Collapse
Grants
- R01HL145372 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- U01HL175444 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL158906 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- HL126176 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL160551 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- P01HL092870 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HG011886 U.S. Department of Health & Human Services | NIH | National Human Genome Research Institute (NHGRI)
- W81XWH1910415 United States Department of Defense | United States Army | Army Medical Command | Congressionally Directed Medical Research Programs (CDMRP)
- GNT1195595 Department of Health | National Health and Medical Research Council (NHMRC)
- GNT1162829 Department of Health | National Health and Medical Research Council (NHMRC)
- U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- U.S. Department of Health & Human Services | NIH | National Human Genome Research Institute (NHGRI)
Collapse
Affiliation(s)
- Annika Vannan
- Division of Bioinnovation and Genome Sciences, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Ruqian Lyu
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
- Melbourne Integrative Genomics, University of Melbourne, Parkville, Victoria, Australia
- School of Mathematics and Statistics, Faculty of Science, University of Melbourne, Parkville, Victoria, Australia
| | - Arianna L Williams
- Division of Bioinnovation and Genome Sciences, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Nicholas M Negretti
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Evan D Mee
- Division of Bioinnovation and Genome Sciences, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Joseph Hirsh
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Samuel Hirsh
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Niran Hadad
- Division of Bioinnovation and Genome Sciences, Translational Genomics Research Institute, Phoenix, AZ, USA
| | - David S Nichols
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Carla L Calvi
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Chase J Taylor
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Vasiliy V Polosukhin
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ana P M Serezani
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - A Scott McCall
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jason J Gokey
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Heejung Shim
- Melbourne Integrative Genomics, University of Melbourne, Parkville, Victoria, Australia
- School of Mathematics and Statistics, Faculty of Science, University of Melbourne, Parkville, Victoria, Australia
| | - Lorraine B Ware
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Matthew J Bacchetta
- Department of Cardiac Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ciara M Shaver
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Timothy S Blackwell
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
- Department of Veterans Affairs Medical Center, Nashville, TN, USA
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Rajat Walia
- Department of Thoracic Disease and Transplantation, Norton Thoracic Institute, Phoenix, AZ, USA
| | - Jennifer M S Sucre
- Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Jonathan A Kropski
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
- Department of Veterans Affairs Medical Center, Nashville, TN, USA
| | - Davis J McCarthy
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
- Melbourne Integrative Genomics, University of Melbourne, Parkville, Victoria, Australia
- School of Mathematics and Statistics, Faculty of Science, University of Melbourne, Parkville, Victoria, Australia
| | - Nicholas E Banovich
- Division of Bioinnovation and Genome Sciences, Translational Genomics Research Institute, Phoenix, AZ, USA.
| |
Collapse
|
22
|
Banaschewski BJH, Michki SN, Sitaraman S, Pan R, Wang JY, Stewart D, Goldy MK, Lin SM, Cantu E, Katzen JB, Basil MC, Emtiazjoo AM, Todd JL, Gokey JJ, Kropski JA, Frank DB, Zepp JA, Young LR. Emergence of inflammatory fibroblasts with aging in Hermansky-Pudlak syndrome associated pulmonary fibrosis. Commun Biol 2025; 8:284. [PMID: 39987372 PMCID: PMC11846979 DOI: 10.1038/s42003-025-07589-9] [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: 08/05/2024] [Accepted: 01/21/2025] [Indexed: 02/24/2025] Open
Abstract
The longitudinal cellular interactions that drive pulmonary fibrosis are not well understood. To investigate the disease underpinnings associated with fibrosis onset and progression, we generated a scRNA-seq atlas of lungs from young and aged mouse models of multiple subtypes of Hermansky-Pudlak syndrome (HPS), a collection of rare autosomal recessive diseases associated with albinism, platelet dysfunction, and pulmonary fibrosis. We have identified an age-dependent increase in SAA3+ inflammatory lung fibroblasts in HPS mice, including in double-mutant HPS1-2 mice which develop spontaneous fibrosis. HPS1 fibroblasts show increased expression of IL-1R1, whereas alveolar type II epithelial cells from HPS2 mice induce the inflammatory gene signature in co-cultured fibroblasts. scRNA-seq of lung tissue from three HPS1 patients similarly shows the presence of inflammatory fibroblasts and increased IL1R1 expression on fibroblasts. These data posit complex interactions between dysfunctional epithelial cells, inflammatory fibroblasts, and recruited immune cells, suggesting potential opportunities for mitigation of the fibrotic cascade.
Collapse
Affiliation(s)
- Brandon J H Banaschewski
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sylvia N Michki
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Cardiology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sneha Sitaraman
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ruby Pan
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Joanna Y Wang
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Dominique Stewart
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Mary Kate Goldy
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Susan M Lin
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
- Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward Cantu
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jeremy B Katzen
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
- Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Maria C Basil
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
- Lung Biology Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Amir M Emtiazjoo
- Deparment of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville, FL, USA
| | - Jamie L Todd
- Duke Clinical Research Institute, Durham, NC, USA
- Duke University Medical Center, Durham, NC, USA
| | - Jason J Gokey
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jonathan A Kropski
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
- Department of Veterans Affairs Medical Center, Nashville, TN, USA
| | - David B Frank
- Division of Cardiology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jarod A Zepp
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Lisa R Young
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
23
|
He H, Ma C, Wei W, Wang H, Lai Y, Liu M, Sun S, Ma Q, Lai J, Liu H, Liu H, Sun F, Lin X. Heparan sulfate regulates myofibroblast heterogeneity and function to mediate niche homeostasis during alveolar morphogenesis. Nat Commun 2025; 16:1834. [PMID: 39979343 PMCID: PMC11842828 DOI: 10.1038/s41467-025-57163-4] [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: 05/06/2024] [Accepted: 02/13/2025] [Indexed: 02/22/2025] Open
Abstract
Postnatal respiration requires bulk formation of alveoli that produces extensive surface area for gas diffusion from epithelium to the circulatory system. Alveolar morphogenesis initiates at late gestation or postnatal stage during mammalian development and is mediated by coordination among multiple cell types. Here we show that fibroblast-derived Heparan Sulfate Glycosaminoglycan (HS-GAG) is essential for maintaining a niche that supports alveolar formation by modulating both biophysical and biochemical cues. Gli1-CreER mediated deletion of HS synthase gene Ext1 in lung fibroblasts results in enlarged and simplified alveolar structures. Ablation of HS results in loss of a subset of PDGFRαhi αSMA+ alveolar myofibroblasts residing in the distal alveolar region, which exhibit contractile properties and maintain WNT signaling activity to support normal proliferation and differentiation of alveolar epithelial cells. HS is essential for proliferation while preventing precocious apoptosis of alveolar myofibroblasts. We show that these processes are dependent upon FGF/MAPK signaling and forced activation of MAPK/ERK signaling partially corrected alveolar simplification and restored alveolar myofibroblast number and AT2 cell proliferation in HS deficient mice. These data reveal HS-dependent myofibroblast heterogeneity and function as an essential orchestrator for developing alveolar niche critical for the generation of gas exchange units.
Collapse
Affiliation(s)
- Hua He
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, China.
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, Sichuan, China.
| | - Chong Ma
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, Sichuan, China
| | - Wei Wei
- The State Key Laboratory of Genetic Engineering, School of Life Sciences, Greater Bay Area Institute of Precision Medicine (Guangzhou), Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haonan Wang
- The State Key Laboratory of Genetic Engineering, School of Life Sciences, Greater Bay Area Institute of Precision Medicine (Guangzhou), Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yutian Lai
- Department of Lung Cancer, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ming Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, Sichuan, China
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Shenfei Sun
- The State Key Laboratory of Genetic Engineering, School of Life Sciences, Greater Bay Area Institute of Precision Medicine (Guangzhou), Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qing Ma
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, Sichuan, China
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Jiashuang Lai
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, Sichuan, China
| | - Hanxiang Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, Sichuan, China
| | - Hanmin Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, China.
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, Sichuan, China.
| | - Fei Sun
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, China.
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, Sichuan, China.
| | - Xinhua Lin
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, Chengdu, Sichuan, China.
- The State Key Laboratory of Genetic Engineering, School of Life Sciences, Greater Bay Area Institute of Precision Medicine (Guangzhou), Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
24
|
Manji A, Wang L, Pape CM, McCaig LA, Troitskaya A, Batnyam O, McDonald LJ, Appleton CT, Veldhuizen RA, Gill SE. Effect of aging on pulmonary cellular responses during mechanical ventilation. JCI Insight 2025; 10:e185834. [PMID: 39946196 PMCID: PMC11949020 DOI: 10.1172/jci.insight.185834] [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: 08/13/2024] [Accepted: 01/28/2025] [Indexed: 03/25/2025] Open
Abstract
Acute respiratory distress syndrome (ARDS) results in substantial morbidity and mortality, especially in elderly people. Mechanical ventilation, a common supportive treatment for ARDS, is necessary for maintaining gas exchange but can also propagate injury. We hypothesized that aging leads to alterations in surfactant function, inflammatory signaling, and microvascular permeability within the lung during mechanical ventilation. Young and aged male mice were mechanically ventilated, and surfactant function, inflammation, and vascular permeability were assessed. Additionally, single-cell RNA-Seq was used to delineate cell-specific transcriptional changes. The results showed that, in aged mice, surfactant dysfunction and vascular permeability were significantly augmented, while inflammation was less pronounced. Differential gene expression and pathway analyses revealed that alveolar macrophages in aged mice showed a blunted inflammatory response, while aged endothelial cells exhibited altered cell-cell junction formation. In vitro functional analysis revealed that aged endothelial cells had an impaired ability to form a barrier. These results highlight the complex interplay between aging and mechanical ventilation, including an age-related predisposition to endothelial barrier dysfunction, due to altered cell-cell junction formation, and decreased inflammation, potentially due to immune exhaustion. It is concluded that age-related vascular changes may underlie the increased susceptibility to injury during mechanical ventilation in elderly patients.
Collapse
Affiliation(s)
- Aminmohamed Manji
- Centre for Critical Illness Research, London Health Sciences Centre Research Institute, London, Ontario, Canada
- Department of Physiology and Pharmacology
| | - Lefeng Wang
- Centre for Critical Illness Research, London Health Sciences Centre Research Institute, London, Ontario, Canada
- Department of Medicine, and
| | - Cynthia M. Pape
- Centre for Critical Illness Research, London Health Sciences Centre Research Institute, London, Ontario, Canada
- Department of Medicine, and
| | - Lynda A. McCaig
- Centre for Critical Illness Research, London Health Sciences Centre Research Institute, London, Ontario, Canada
- Department of Medicine, and
| | - Alexandra Troitskaya
- Centre for Critical Illness Research, London Health Sciences Centre Research Institute, London, Ontario, Canada
- Department of Physiology and Pharmacology
| | - Onon Batnyam
- Centre for Critical Illness Research, London Health Sciences Centre Research Institute, London, Ontario, Canada
| | - Leah J.J. McDonald
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | | | - Ruud A.W. Veldhuizen
- Centre for Critical Illness Research, London Health Sciences Centre Research Institute, London, Ontario, Canada
- Department of Physiology and Pharmacology
- Department of Medicine, and
| | - Sean E. Gill
- Centre for Critical Illness Research, London Health Sciences Centre Research Institute, London, Ontario, Canada
- Department of Physiology and Pharmacology
- Department of Medicine, and
| |
Collapse
|
25
|
Diao S, Zou JY, Wang S, Ghaddar N, Chan JE, Kim H, Poulain N, Koumenis C, Hatzoglou M, Walter P, Sonenberg N, Le Quesne J, Tammela T, Koromilas AE. Lineage plasticity of the integrated stress response is a hallmark of cancer evolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.10.637516. [PMID: 39990365 PMCID: PMC11844398 DOI: 10.1101/2025.02.10.637516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
The link between the "stress phenotype"-a well-established hallmark of cancer-and its role in tumor progression and intratumor heterogeneity remains poorly defined. The integrated stress response (ISR) is a key adaptive pathway that enables tumor survival under oncogenic stress. While ISR has been implicated in promoting tumor growth, its precise role in driving tumor evolution and heterogeneity has not been elucidated. In this study, using a genetically engineered mouse models, we demonstrate that ISR activation-indicated by elevated levels of phosphorylated eIF2 (p-eIF2) and ATF4-is essential for the emergence of dedifferentiated, therapy-resistant cell states. ISR, through the coordinated actions of ATF4 and MYC, facilitates the development of tumor cell populations characterized by high plasticity, stemness, and an epithelial-mesenchymal transition (EMT)-prone phenotype. This process is driven by ISR-mediated expression of genes that maintain mitochondrial integrity and function, critical for sustaining tumor progression. Importantly, genetic, or pharmacological inhibition of the p-eIF2-ATF4 signaling axis leads to mitochondrial dysfunction and significantly impairs tumor growth in mouse models of lung adenocarcinoma (LUAD). Moreover, ISR-driven dedifferentiation is associated with poor prognosis and therapy resistance in advanced human LUAD, underscoring ISR inhibition as a promising therapeutic strategy to disrupt tumor evolution and counteract disease progression.
Collapse
|
26
|
Kim J, Yuan Y, Agaronyan K, Zhao A, Wang VD, Gau D, Toosi N, Gupta G, Essayas H, Kaminski A, McGovern J, Yu S, Woo S, Lee CJ, Gandhi S, Saber T, Saleh T, Hu B, Sun Y, Ishikawa G, Bain W, Evankovich J, Chen L, Yun H, Herzog EL, Dela Cruz CS, Ryu C, Sharma L. Damage sensing through TLR9 regulates inflammatory and antiviral responses during influenza infection. Mucosal Immunol 2025:S1933-0219(25)00008-X. [PMID: 39884393 DOI: 10.1016/j.mucimm.2025.01.008] [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: 09/02/2024] [Revised: 12/05/2024] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
Host response aimed at eliminating the infecting pathogen, as well as the pathogen itself, can cause tissue injury. Tissue injury leads to the release of a myriad of cellular components including mitochondrial DNA (mtDNA), which the host senses through pattern recognition receptors. How the sensing of tissue injury by the host shapes the anti-pathogen response remains poorly understood. In this study, we utilized mice that are deficient in toll-like receptor-9 (TLR9), which binds to unmethylated CpG DNA sequences such as those present in bacterial and mtDNA. To avoid direct pathogen sensing by TLR9, we utilized the influenza virus, which lacks ligands for TLR9, to determine how damage sensing by TLR9 contributes to anti-influenza immunity. Our data showed that TLR9-mediated sensing of tissue damage promoted an inflammatory response during early infection, driven by epithelial and myeloid cells. Along with the diminished inflammatory response, the absence of TLR9 led to impaired viral clearance manifested as higher and prolonged influenza components in myeloid cells, including monocytes and macrophages, rendering them highly inflammatory. The persistent inflammation driven by infected myeloid cells led to persistent lung injury and impaired recovery in influenza-infected TLR9-/- mice. Further, we found elevated TLR9 ligands in the plasma samples of patients with influenza infection and its association with the disease severity in hospitalized patients, demonstrating its clinical relevance. Overall, we demonstrated an essential role of damage sensing through TLR9 in promoting anti-influenza immunity and inflammatory response. AUTHOR SUMMARY: Tissue damage is an inevitable outcome of clinically relevant lung infections, but the host mechanisms for detecting such damage during infection are not well understood. We investigated the role of Toll-like receptor 9 (TLR9) in sensing tissue damage caused by influenza. Since influenza lacks TLR9 ligands, we hypothesized that TLR9 signaling is driven by tissue damage molecules like mitochondrial DNA (mtDNA). Our data revealed that TLR9 deficiency reduces early inflammatory lung injury but impairs viral clearance, resulting in extensive infection of immune cells, persistent inflammation, and delayed recovery. Myeloid-specific TLR9 deletion ameliorated late-stage inflammatory responses. In humans, influenza-infected individuals exhibited elevated TLR9 activity and mtDNA levels in plasma compared to healthy controls, with higher TLR9 activation potential correlating with severe disease requiring ICU admission. These findings suggest that TLR9-mediated damage sensing triggers both inflammatory tissue injury and viral clearance. These data indicate that TLR9 activity can serve as a crucial biomarker and therapeutic target to limit influenza-induced tissue injury.
Collapse
Affiliation(s)
- Jooyoung Kim
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center Pittsburgh PA USA; Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine New Haven CT USA
| | - Yifan Yuan
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine New Haven CT USA; University of Maryland MD USA
| | - Karen Agaronyan
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine New Haven CT USA; Howard Hughes Medical Institute, USA
| | - Amy Zhao
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine New Haven CT USA
| | - Victoria D Wang
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center Pittsburgh PA USA
| | - David Gau
- Department of Pathology, University of Pittsburgh Pittsburgh PA USA; Department of Bioengineering, University of Pittsburgh Pittsburgh PA USA
| | - Nicholas Toosi
- Department of Bioengineering, University of Pittsburgh Pittsburgh PA USA
| | - Gayatri Gupta
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine New Haven CT USA
| | - Heran Essayas
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine New Haven CT USA
| | - Ayelet Kaminski
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine New Haven CT USA
| | - John McGovern
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine New Haven CT USA
| | - Sheeline Yu
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine New Haven CT USA
| | - Samuel Woo
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine New Haven CT USA
| | - Chris J Lee
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine New Haven CT USA
| | - Shifa Gandhi
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine New Haven CT USA
| | - Tina Saber
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine New Haven CT USA
| | - Tayebeh Saleh
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center Pittsburgh PA USA
| | - Buqu Hu
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine New Haven CT USA
| | - Ying Sun
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine New Haven CT USA
| | - Genta Ishikawa
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine New Haven CT USA
| | - William Bain
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center Pittsburgh PA USA; VA Medical Center Pittsburgh PA USA
| | - John Evankovich
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center Pittsburgh PA USA
| | - Lujia Chen
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15206, USA
| | - HongDuck Yun
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center Pittsburgh PA USA
| | - Erica L Herzog
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine New Haven CT USA
| | - Charles S Dela Cruz
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center Pittsburgh PA USA; Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine New Haven CT USA; VA Medical Center Pittsburgh PA USA
| | - Changwan Ryu
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine New Haven CT USA.
| | - Lokesh Sharma
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center Pittsburgh PA USA; Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine New Haven CT USA.
| |
Collapse
|
27
|
Eigenbrood J, Wong N, Mallory P, Pereira J, Morris-II DW, Beck JA, Cronk JC, Sayers CM, Mendez M, Kaiser L, Galindo J, Singh J, Cardamone A, Pore M, Kelly M, LeBlanc AK, Cotter J, Kaplan RN, McEachron TA. Spatial profiling identifies regionally distinct microenvironments and targetable immunosuppressive mechanisms in pediatric osteosarcoma pulmonary metastases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.22.631350. [PMID: 39896512 PMCID: PMC11785069 DOI: 10.1101/2025.01.22.631350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Osteosarcoma is the most common malignant bone tumor in young patients and remains a significant clinical challenge, particularly in the context of metastatic disease. Despite extensive documentation of genomic alterations in osteosarcoma, studies detailing the immunosuppressive mechanisms within the metastatic osteosarcoma microenvironment are lacking. Our objective was to characterize the spatial transcriptional landscape of metastatic osteosarcoma to reveal these immunosuppressive mechanisms and identify promising therapeutic targets. Here, we performed spatial transcriptional profiling on a cohort of osteosarcoma pulmonary metastases from pediatric patients. We reveal a conserved spatial gene expression pattern resembling a foreign body granuloma, characterized by peripheral inflammatory signaling, fibrocollagenous encapsulation, lymphocyte exclusion, and peritumoral macrophage accumulation. We also show that the intratumoral microenvironment of these lesions lack inflammatory signaling. Additionally, we identified CXCR4 as an actionable immunomodulatory target that bridges both the intratumoral and extratumoral microenvironments and highlights the spatial heterogeneity and complexity of this pathway. Collectively, this study reveals that metastatic osteosarcoma specimens are comprised of multiple regionally distinct immunosuppressive microenvironments.
Collapse
Affiliation(s)
- Jason Eigenbrood
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- Current Address: University of Cambridge, Cancer Research UK Cambridge Institute, Cambridge, UK
- These authors contributed equally to this manuscript
| | - Nathan Wong
- Collaborative Bioinformatics Resource, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
- These authors contributed equally to this manuscript
| | - Paul Mallory
- Imaging Mass Cytometry Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Janice Pereira
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Douglass W Morris-II
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jessica A Beck
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - James C Cronk
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Carly M Sayers
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Monica Mendez
- Department of Pathology and Laboratory Medicine, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Linus Kaiser
- Department of Pathology and Laboratory Medicine, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Julie Galindo
- Department of Pathology and Laboratory Medicine, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Jatinder Singh
- Center for Cancer Research Single Cell Analysis Facility, Cancer Research Technology Program, Frederick National Laboratory, Bethesda, MD, USA
| | - Ashley Cardamone
- Imaging Mass Cytometry Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Milind Pore
- Imaging Mass Cytometry Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Michael Kelly
- Center for Cancer Research Single Cell Analysis Facility, Cancer Research Technology Program, Frederick National Laboratory, Bethesda, MD, USA
| | - Amy K LeBlanc
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jennifer Cotter
- Department of Pathology and Laboratory Medicine, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Rosandra N Kaplan
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Troy A McEachron
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
28
|
Hrovatin K, Sikkema L, Shitov VA, Heimberg G, Shulman M, Oliver AJ, Mueller MF, Ibarra IL, Wang H, Ramírez-Suástegui C, He P, Schaar AC, Teichmann SA, Theis FJ, Luecken MD. Considerations for building and using integrated single-cell atlases. Nat Methods 2025; 22:41-57. [PMID: 39672979 DOI: 10.1038/s41592-024-02532-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 10/22/2024] [Indexed: 12/15/2024]
Abstract
The rapid adoption of single-cell technologies has created an opportunity to build single-cell 'atlases' integrating diverse datasets across many laboratories. Such atlases can serve as a reference for analyzing and interpreting current and future data. However, it has become apparent that atlasing approaches differ, and the impact of these differences are often unclear. Here we review the current atlasing literature and present considerations for building and using atlases. Importantly, we find that no one-size-fits-all protocol for atlas building exists, but rather we discuss context-specific considerations and workflows, including atlas conceptualization, data collection, curation and integration, atlas evaluation and atlas sharing. We further highlight the benefits of integrated atlases for analyses of new datasets and deriving biological insights beyond what is possible from individual datasets. Our overview of current practices and associated recommendations will improve the quality of atlases to come, facilitating the shift to a unified, reference-based understanding of single-cell biology.
Collapse
Affiliation(s)
- Karin Hrovatin
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Lisa Sikkema
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Vladimir A Shitov
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive / Institute of Lung Health and Immunity (LHI), Helmholtz Zentrum München; Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Graham Heimberg
- Department of OMNI Bioinformatics, Genentech, South San Francisco, CA, USA
- Department of Biological Research | AI Development, Genentech, South San Francisco, CA, USA
| | - Maiia Shulman
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Amanda J Oliver
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Michaela F Mueller
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
| | - Ignacio L Ibarra
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
| | - Hanchen Wang
- Department of Biological Research | AI Development, Genentech, South San Francisco, CA, USA
- Department of Computer Science, Stanford University, Palo Alto, CA, USA
| | - Ciro Ramírez-Suástegui
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Peng He
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Anna C Schaar
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
- TUM School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Theory of Condensed Matter Group, Department of Physics, Cavendish Laboratory, University of Cambridge, Cambridge, UK
- Cambridge Stem Cell Institute and Department of Medicine, University of Cambridge, Cambridge, UK
- CIFAR MacMillan Multiscale Human Programme, Toronto, Ontario, Canada
| | - Fabian J Theis
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany.
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
- Department of Mathematics, Technical University of Munich, Garching, Germany.
| | - Malte D Luecken
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany.
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive / Institute of Lung Health and Immunity (LHI), Helmholtz Zentrum München; Member of the German Center for Lung Research (DZL), Munich, Germany.
| |
Collapse
|
29
|
Nishimura T, Kouwaki T, Takashima K, Ochi A, Mtali YS, Oshiumi H. Cholesterol restriction primes antiviral innate immunity via SREBP1-driven noncanonical type I IFNs. EMBO Rep 2025; 26:560-592. [PMID: 39668245 PMCID: PMC11772592 DOI: 10.1038/s44319-024-00346-9] [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: 05/22/2024] [Revised: 11/28/2024] [Accepted: 11/28/2024] [Indexed: 12/14/2024] Open
Abstract
Cholesterol metabolism is associated with innate immune responses; however, the underlying mechanism remains unclear. Here, we perform chemical screening to isolate small molecules influencing RIG-I activity, a cytoplasmic viral RNA sensor. We find that statins, which inhibit cholesterol synthesis, dramatically enhance RIG-I-dependent antiviral responses in specific cell types. Since statins exhibit pleiotropic effects on type I interferon (IFN) responses, we further focus on their effects on RIG-I signaling. The restriction of cholesterol synthesis induces expression of noncanonical type I IFNs, such as IFN-ω, in an SREBP1 transcription factor-dependent manner. This pathway subsequently enhances RIG-I-mediated signaling following viral infection. Administration of statins augments RIG-I-dependent cytokine expression in the lungs of mice. Conversely, a mouse obesity model shows a diminished RIG-I response. Single-cell transcriptome analyses reveal a subset of alveolar macrophages that increase RIG-I expression in response to inhibited cholesterol synthesis in vivo. This study reveals SREBP1-mediated noncanonical type I IFN expression, linking cholesterol metabolism and RIG-I signaling.
Collapse
Affiliation(s)
- Tasuku Nishimura
- Department of Immunology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Takahisa Kouwaki
- Department of Immunology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan.
| | - Ken Takashima
- Department of Immunology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Akie Ochi
- School of Medicine, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Yohana S Mtali
- Department of Immunology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Hiroyuki Oshiumi
- Department of Immunology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan.
| |
Collapse
|
30
|
Eastburn DJ, White KS, Jayne ND, Camiolo S, Montis G, Ha S, Watson KG, Yeakley JM, McComb J, Seligmann B. High-throughput gene expression analysis with TempO-LINC sensitively resolves complex brain, lung and kidney heterogeneity at single-cell resolution. Sci Rep 2024; 14:31285. [PMID: 39732835 PMCID: PMC11682069 DOI: 10.1038/s41598-024-82736-6] [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: 08/19/2024] [Accepted: 12/09/2024] [Indexed: 12/30/2024] Open
Abstract
We report the development and performance of a novel genomics platform, TempO-LINC, for conducting high-throughput transcriptomic analysis on single cells and nuclei. TempO-LINC works by adding cell-identifying molecular barcodes onto highly selective and high-sensitivity gene expression probes within fixed cells, without having to first generate cDNA. Using an instrument-free combinatorial indexing approach, all probes within the same fixed cell receive an identical barcode, enabling the reconstruction of single-cell gene expression profiles across as few as several hundred cells and up to 100,000 + cells per sample. The TempO-LINC approach is easily scalable based on the number of barcodes and rounds of barcoding performed; however, for the experiments reported in this study, the assay utilized over 5.3 million unique barcodes. TempO-LINC offers a robust protocol for fixing and banking cells and displays high-sensitivity gene detection from multiple diverse sample types. We show that TempO-LINC has a multiplet rate of less than 1.1% and a cell capture rate of ~ 50%. Although the assay can accurately profile the whole transcriptome (19,683 human, 21,400 mouse and 21,119 rat genes), it can be targeted to measure only actionable/informative genes and molecular pathways of interest - thereby reducing sequencing requirements. In this study, we applied TempO-LINC to profile the transcriptomes of more than 90,000 cells across multiple species and sample types, including nuclei from mouse lung, kidney and brain tissues. The data demonstrated the ability to identify and annotate more than 50 unique cell populations and positively correlate expression of cell type-specific molecular markers within them. TempO-LINC is a robust new single-cell technology that is ideal for large-scale applications/studies with high data quality.
Collapse
Affiliation(s)
| | | | | | | | | | - Seungeun Ha
- BioSpyder Technologies, Inc., Carlsbad, CA, USA
| | | | | | - Joel McComb
- BioSpyder Technologies, Inc., Carlsbad, CA, USA
| | | |
Collapse
|
31
|
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
|
32
|
Yan K, Dumenil T, Stewart R, Bishop CR, Tang B, Nguyen W, Suhrbier A, Rawle DJ. TMEM106B-mediated SARS-CoV-2 infection allows for robust ACE2-independent infection in vitro but not in vivo. Cell Rep 2024; 43:114921. [PMID: 39480813 DOI: 10.1016/j.celrep.2024.114921] [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: 06/16/2024] [Revised: 09/17/2024] [Accepted: 10/14/2024] [Indexed: 11/02/2024] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2) is the primary entry receptor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), but ACE2-independent entry has been observed in vitro for strains with the spike-E484D substitution. Here, we conduct a whole-genome CRISPR-Cas9 knockout screen using SARS-CoV-2 mouse adapted 1 (SARS-CoV-2MA1), which carries spike-E484D, to identify the ACE2-independent entry mechanisms. SARS-CoV-2MA1 infection in HEK293T cells relies on heparan sulfate and endocytic pathways, with TMEM106B, a transmembrane lysosomal protein, the most significant contributor. While SARS-CoV-2MA1 productively infects human brain organoids and K18-hACE2 mouse brains, it does not infect C57BL/6J or Ifnar-/- mouse brains. This suggests that ACE2-independent entry via TMEM106B, which is predominantly expressed in the brain, does not overtly increase the risk of SARS-CoV-2 neuroinvasiveness in mice with endogenous Ace2 expression. Importantly, SARS-CoV-2MA1 does not replicate in the Ace2-/- mouse respiratory tract. Overall, this suggests that robust ACE2-independent infection by SARS-CoV-2MA1 is likely an in vitro phenomenon with no apparent implications for infection in vivo.
Collapse
Affiliation(s)
- Kexin Yan
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Troy Dumenil
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Romal Stewart
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Cameron R Bishop
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Bing Tang
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Wilson Nguyen
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Andreas Suhrbier
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia; Australian Infectious Disease Research Centre, GVN Center of Excellence, Brisbane, QLD, Australia
| | - Daniel J Rawle
- Infection and Inflammation Department, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia.
| |
Collapse
|
33
|
Park HB, Kim KH, Kim JH, Kim SI, Oh YM, Kang M, Lee S, Hwang S, Lee H, Lee T, Park S, Lee JE, Jeong GR, Lee DH, Youn H, Choi EY, Son WC, Chung SJ, Chung J, Choi K. Improved safety of chimeric antigen receptor T cells indirectly targeting antigens via switchable adapters. Nat Commun 2024; 15:9917. [PMID: 39557825 PMCID: PMC11574259 DOI: 10.1038/s41467-024-53996-7] [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: 08/30/2023] [Accepted: 10/28/2024] [Indexed: 11/20/2024] Open
Abstract
Chimeric antigen receptor T (CAR-T) cells show remarkable efficacy for some hematological malignancies. However, CAR targets that are expressed at high level and selective to tumors are scarce. Several strategies have been proposed to tackle the on-target off-tumor toxicity of CAR-T cells that arise from suboptimal selectivity, but these are complicated, with many involving dual gene expression for specificity. In this study, we show that switchable CAR-T cells with a tumor targeting adaptor can mitigate on-target off-tumor toxicity against a low selectivity tumor antigen that cannot be targeted by conventional CAR-T cells, such as CD40. Our system is composed of anti-cotinine murine CAR-T cells and cotinine-labeled anti-CD40 single chain variable fragments (scFv), with which we show selective tumor killing while sparing CD40-expressing normal cells including macrophages in a mouse model of lymphoma. Simple replacement of the tumor-targeting adaptor with a suicidal drug-conjugated tag may further enhance safety by enabling permanent in vivo depletion of the switchable CAR-T cells when necessary. In summary, our switchable CAR system can control CAR-T cell toxicity while maintaining therapeutic efficacy, thereby expanding the range of CAR targets.
Collapse
MESH Headings
- Animals
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Receptors, Chimeric Antigen/genetics
- Mice
- Humans
- Immunotherapy, Adoptive/methods
- CD40 Antigens/immunology
- CD40 Antigens/metabolism
- T-Lymphocytes/immunology
- Cell Line, Tumor
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Single-Chain Antibodies/immunology
- Single-Chain Antibodies/genetics
- Lymphoma/immunology
- Lymphoma/therapy
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/genetics
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Hyung Bae Park
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ki Hyun Kim
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ju Hwan Kim
- AbTis Co. Ltd., Suwon, Gyeonggi-do, Republic of Korea
| | - Sang Il Kim
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yu Mi Oh
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Miseung Kang
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seoho Lee
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Siwon Hwang
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyeonmin Lee
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - TaeJin Lee
- AbTis Co. Ltd., Suwon, Gyeonggi-do, Republic of Korea
- Department of Biopharmaceutical Convergence, School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, Republic of Korea
| | - Seungbin Park
- Department of Biopharmaceutical Convergence, School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, Republic of Korea
| | - Ji Eun Lee
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ga Ram Jeong
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Ticaros Inc., Seoul, Republic of Korea
| | - Dong Hyun Lee
- Department of Medical Science, AMIST, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Hyewon Youn
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Nuclear Medicine, Cancer Imaging Center, Seoul National University Hospital, Seoul, Republic of Korea
| | - Eun Young Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Institute of Human Environment Interface Biology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Woo Chan Son
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Sang J Chung
- Department of Biopharmaceutical Convergence, School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, Republic of Korea.
| | - Junho Chung
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| | - Kyungho Choi
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
34
|
Na M, Kang H, Kim N, Jo A, Lee HO. Characterization of an orthotopic mouse transplant model reveals early changes in the tumor microenvironment of lung cancer. BMB Rep 2024; 57:484-489. [PMID: 39044458 PMCID: PMC11608852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 02/13/2024] [Accepted: 03/23/2024] [Indexed: 07/25/2024] Open
Abstract
To understand the cellular and molecular dynamics in the early stages of lung cancer, we explored a mouse model of orthotopic tumor transplant created from the Lewis Lung Carcinoma (LLC) cell line. Employing single-cell RNA sequencing, we analyzed the cellular landscape during tumor engraftment, focusing particularly on LLC cells harboring the Kras G12C mutation. This allowed us to identify LLC tumor cells via the detection of mutant Kras transcripts and observe elevated levels of Myc and mesenchymal gene expression. Moreover, our study revealed significant alterations in the lung microenvironment, including the activation of tissue remodeling genes in fibroblasts and the downregulation of MHC class II genes in myeloid subsets. Additionally, T/NK cell subsets displayed more regulatory phenotypes, coupled with reduced proliferation in CD8+ T cells. Collectively, these findings enhance our understanding of lung cancer progression, particularly in a tumor microenvironment with low immunogenicity. [BMB Reports 2024; 57(11): 484-489].
Collapse
Affiliation(s)
- Minsu Na
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Department of Biomedicine and Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
| | - Huiram Kang
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Department of Biomedicine and Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
| | - Nayoung Kim
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Department of Biomedicine and Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
| | - Areum Jo
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Department of Biomedicine and Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
| | - Hae-Ock Lee
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Department of Biomedicine and Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|
35
|
Kaur G, Wang Q, Tjitropranoto A, Unwalla H, Rahman I. Cold ischemia time alters cell-type specific senescence leading to loss of cellular integrity in mouse lungs. Exp Lung Res 2024; 50:184-198. [PMID: 39427288 PMCID: PMC11513191 DOI: 10.1080/01902148.2024.2414974] [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: 06/19/2024] [Revised: 09/11/2024] [Accepted: 10/01/2024] [Indexed: 10/22/2024]
Abstract
Purpose: Ischemia-reperfusion injury (IRI) is a major challenge in lung transplantation often causing graft dysfunction and chronic airway illnesses in recipients. To prevent potential transplant related complications, strict guidelines were put in place to choose viable donor lungs with minimal risk of IRI. These regulations deem most of the donor organs unfit for transplant which then are donated for research to understand the mechanisms of health and diseases in human. However, resected organs that are being transported undergo cold ischemia that can negatively affect the tissue architecture and other cellular functions under study. Thus, it is important to assess how cold ischemia time (CIT) affects the physiological mechanism. In this respect, we are interested in studying how CIT affects cellular senescence in normal aging and various pulmonary pathologies. We thus hypothesized that prolonged CIT exhibits cell-type specific changes in lung cellular senescence in mice. Methods: Lung lobes from C57BL/6J (n = 5-8) mice were harvested and stored in UW Belzer cold storage solution for 0, 4-, 9-, 12-, 24-, and 48-h CIT. Lung cellular senescence was determined using fluorescence (C12FdG) assay and co-immunolabelling was performed to identify changes in individual cell types. Results: We found a rapid decline in the overall lung cellular senescence after 4-h of CIT in our study. Co-immunolabelling revealed the endothelial cells to be most affected by cold ischemia, demonstrating significant decrease in the endothelial cell senescence immediately after harvest. Annexin V-PI staining further revealed a prominent increase in the number of necrotic cells at 4-h CIT, thus suggesting that most of the cells undergo cell death within a few hours of cold ischemic injury. Conclusions: We thus concluded that CIT significantly lowers the cellular senescence in lung tissues and must be considered as a confounding factor for mechanistic studies in the future.
Collapse
Affiliation(s)
- Gagandeep Kaur
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY
| | - Qixin Wang
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY
| | - Ariel Tjitropranoto
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY
| | - Hoshang Unwalla
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY
| |
Collapse
|
36
|
Xu Z, Chen L, Lin X, Lyu Y, Zhou M, Chen H, Zhang H, Zhang T, Chen Y, Suo Y, Liang Q, Qin Z, Wang Y. Single Nucleus Total RNA Sequencing of Formalin-Fixed Paraffin-Embedded Gliomas. SMALL METHODS 2024; 8:e2301801. [PMID: 38958078 DOI: 10.1002/smtd.202301801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 06/20/2024] [Indexed: 07/04/2024]
Abstract
Gliomas, the predominant form of brain cancer, comprise diverse malignant subtypes with limited curative therapies available. The insufficient understanding of their molecular diversity and evolutionary processes hinders the advancement of new treatments. Technical complexities associated with formalin-fixed paraffin-embedded (FFPE) clinical samples hinder molecular-level analyses of gliomas. Current single-cell RNA sequencing (scRNA-seq) platforms are inadequate for large-scale clinical applications. In this study, automated snRandom-seq is developed, a high-throughput single-nucleus total RNA sequencing platform optimized for archival FFPE samples. This platform integrates automated single-nucleus isolation and droplet barcoding systems with the random primer-based scRNA-seq chemistry, accommodating a broad spectrum of sample types. The automated snRandom-seq is applied to analyze 116 492 single nuclei from 17 FFPE samples of various glioma subtypes, including rare clinical samples and matched primary-recurrent glioblastomas (GBMs). The study provides comprehensive insights into the molecular characteristics of gliomas at the single-cell level. Abundant non-coding RNAs (ncRNAs) with distinct expression profiles across different glioma clusters and uncovered promising recurrence-related targets and pathways in primary-recurrent GBMs are identified. These findings establish automated snRandom-seq as a robust tool for scRNA-seq of FFPE samples, enabling exploration of molecular diversities and tumor evolution. This platform holds significant implications for large-scale integrative and retrospective clinical research.
Collapse
Affiliation(s)
- Ziye Xu
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Lingchao Chen
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Xin Lin
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yuexiao Lyu
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | | | - Haide Chen
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | | | | | - Yu Chen
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Key Laboratory of Clinical In Vitro Diagnostic Techniques, Hangzhou, 310003, China
| | - Yuanzhen Suo
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Jiangsu Healthy Life Innovation Medical Technology Co., Ltd, Wuxi, 214174, China
| | | | - Zhiyong Qin
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yongcheng Wang
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310003, China
| |
Collapse
|
37
|
He H, Bell SM, Davis AK, Zhao S, Sridharan A, Na CL, Guo M, Xu Y, Snowball J, Swarr DT, Zacharias WJ, Whitsett JA. PRDM3/16 regulate chromatin accessibility required for NKX2-1 mediated alveolar epithelial differentiation and function. Nat Commun 2024; 15:8112. [PMID: 39284798 PMCID: PMC11405758 DOI: 10.1038/s41467-024-52154-3] [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: 01/08/2024] [Accepted: 08/27/2024] [Indexed: 09/20/2024] Open
Abstract
While the critical role of NKX2-1 and its transcriptional targets in lung morphogenesis and pulmonary epithelial cell differentiation is increasingly known, mechanisms by which chromatin accessibility alters the epigenetic landscape and how NKX2-1 interacts with other co-activators required for alveolar epithelial cell differentiation and function are not well understood. Combined deletion of the histone methyl transferases Prdm3 and Prdm16 in early lung endoderm causes perinatal lethality due to respiratory failure from loss of AT2 cells and the accumulation of partially differentiated AT1 cells. Combination of single-cell RNA-seq, bulk ATAC-seq, and CUT&RUN data demonstrate that PRDM3 and PRDM16 regulate chromatin accessibility at NKX2-1 transcriptional targets critical for perinatal AT2 cell differentiation and surfactant homeostasis. Lineage specific deletion of PRDM3/16 in AT2 cells leads to lineage infidelity, with PRDM3/16 null cells acquiring partial AT1 fate. Together, these data demonstrate that NKX2-1-dependent regulation of alveolar epithelial cell differentiation is mediated by epigenomic modulation via PRDM3/16.
Collapse
Affiliation(s)
- Hua He
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, Sichuan, China.
| | - Sheila M Bell
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Ashley Kuenzi Davis
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Shuyang Zhao
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Anusha Sridharan
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Cheng-Lun Na
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Minzhe Guo
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Yan Xu
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - John Snowball
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Daniel T Swarr
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - William J Zacharias
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jeffrey A Whitsett
- Perinatal Institute, Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
38
|
Dandavate V, Bolshette N, Van Drunen R, Manella G, Bueno-Levy H, Zerbib M, Kawano I, Golik M, Adamovich Y, Asher G. Hepatic BMAL1 and HIF1α regulate a time-dependent hypoxic response and prevent hepatopulmonary-like syndrome. Cell Metab 2024; 36:2038-2053.e5. [PMID: 39106859 DOI: 10.1016/j.cmet.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/28/2024] [Accepted: 07/05/2024] [Indexed: 08/09/2024]
Abstract
The transcriptional response to hypoxia is temporally regulated, yet the molecular underpinnings and physiological implications are unknown. We examined the roles of hepatic Bmal1 and Hif1α in the circadian response to hypoxia in mice. We found that the majority of the transcriptional response to hypoxia is dependent on either Bmal1 or Hif1α, through shared and distinct roles that are daytime determined. We further show that hypoxia-inducible factor (HIF)1α accumulation upon hypoxia is temporally regulated and Bmal1 dependent. Unexpectedly, mice lacking both hepatic Bmal1 and Hif1α are hypoxemic and exhibit increased mortality upon hypoxic exposure in a daytime-dependent manner. These mice display mild liver dysfunction with pulmonary vasodilation likely due to extracellular signaling regulated kinase (ERK) activation, endothelial nitric oxide synthase, and nitric oxide accumulation in lungs, suggestive of hepatopulmonary syndrome. Our findings indicate that hepatic BMAL1 and HIF1α are key time-dependent regulators of the hypoxic response and can provide molecular insights into the pathophysiology of hepatopulmonary syndrome.
Collapse
Affiliation(s)
- Vaishnavi Dandavate
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Nityanand Bolshette
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Rachel Van Drunen
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Gal Manella
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Hanna Bueno-Levy
- Department of the Veterinary Resources, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Mirie Zerbib
- Department of the Veterinary Resources, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Ippei Kawano
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Marina Golik
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Yaarit Adamovich
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Gad Asher
- Department of Biomolecular Sciences, Weizmann Institute of Science, 7610001 Rehovot, Israel.
| |
Collapse
|
39
|
Milman Krentsis I, Zheng Y, Rosen C, Shin SY, Blagdon C, Shoshan E, Qi Y, Wang J, Yadav SK, Bachar Lustig E, Shetzen E, Dickey BF, Karmouty-Quintana H, Reisner Y. Lung cell transplantation for pulmonary fibrosis. SCIENCE ADVANCES 2024; 10:eadk2524. [PMID: 39178253 PMCID: PMC11343030 DOI: 10.1126/sciadv.adk2524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 07/19/2024] [Indexed: 08/25/2024]
Abstract
Idiopathic pulmonary fibrosis is a major cause of death with few treatment options. Here, we demonstrate the therapeutic efficacy for lung fibrosis of adult lung cell transplantation using a single-cell suspension of the entire lung in two distinct mouse systems: bleomycin treatment and mice lacking telomeric repeat-binding factor 1 expression in alveolar type 2 (AT2) cells (SPC-Cre TRF1fl/fl), spontaneously developing fibrosis. In both models, the progression of fibrosis was associated with reduced levels of host lung progenitors, enabling engraftment of donor progenitors without any additional conditioning, in contrast to our previous studies. Two months after transplantation, engrafted progenitors expanded to form numerous donor-derived patches comprising AT1 and AT2 alveolar cells, as well as donor-derived mesenchymal and endothelial cells. This lung chimerism was associated with attenuation of fibrosis, as demonstrated histologically, biochemically, by computed tomography imaging, and by lung function measurements. Our study provides a strong rationale for the treatment of lung fibrosis using lung cell transplantation.
Collapse
Affiliation(s)
- Irit Milman Krentsis
- Department of Stem Cell Transplantation and Cell Therapy, MD Anderson Cancer Center, Houston, TX, USA
| | - Yangxi Zheng
- Department of Stem Cell Transplantation and Cell Therapy, MD Anderson Cancer Center, Houston, TX, USA
| | - Chava Rosen
- Department of Stem Cell Transplantation and Cell Therapy, MD Anderson Cancer Center, Houston, TX, USA
- Department of Neonatology, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel-Hashomer, Israel
| | - Sarah Y. Shin
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Christa Blagdon
- Department of Stem Cell Transplantation and Cell Therapy, MD Anderson Cancer Center, Houston, TX, USA
| | - Einav Shoshan
- Department of Stem Cell Transplantation and Cell Therapy, MD Anderson Cancer Center, Houston, TX, USA
| | - Yuan Qi
- Department of Bioinformatics and Computational Biology, MD Anderson Cancer, Houston, TX, USA
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, MD Anderson Cancer, Houston, TX, USA
| | - Sandeep K. Yadav
- Department of Stem Cell Transplantation and Cell Therapy, MD Anderson Cancer Center, Houston, TX, USA
| | - Esther Bachar Lustig
- Department of Stem Cell Transplantation and Cell Therapy, MD Anderson Cancer Center, Houston, TX, USA
| | - Elias Shetzen
- Department of Stem Cell Transplantation and Cell Therapy, MD Anderson Cancer Center, Houston, TX, USA
| | - Burton F. Dickey
- Department of Pulmonary Medicine, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Divisions of Critical Care, Pulmonary and Sleep Medicine, Department of Internal Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yair Reisner
- Department of Stem Cell Transplantation and Cell Therapy, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
40
|
Kim J, Yuan Y, Agaronyan K, Zhao A, D Wang V, Gupta G, Essayas H, Kaminski A, McGovern J, Yu S, Woo S, Lee CJ, Gandhi S, Saber T, Saleh T, Hu B, Sun Y, Ishikawa G, Bain W, Evankovich J, Chen L, Yun H, Herzog EL, Dela Cruz CS, Ryu C, Sharma L. Damage sensing through TLR9 Regulates Inflammatory and Antiviral Responses During Influenza Infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.04.583378. [PMID: 38496452 PMCID: PMC10942338 DOI: 10.1101/2024.03.04.583378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Host response aimed at eliminating the infecting pathogen, as well as the pathogen itself, can cause tissue injury. Tissue injury leads to the release of a myriad of cellular components including mitochondrial DNA, which the host senses through pattern recognition receptors. How the sensing of tissue injury by the host shapes the anti-pathogen response remains poorly understood. In this study, we utilized mice that are deficient in toll-like receptor-9 (TLR9), which binds to unmethylated CpG DNA sequences such as those present in bacterial and mitochondrial DNA. To avoid direct pathogen sensing by TLR9, we utilized the influenza virus, which lacks ligands for TLR9, to determine how damage sensing by TLR9 contributes to anti-influenza immunity. Our data show that TLR9-mediated sensing of tissue damage promotes an inflammatory response during early infection, driven by the epithelial and myeloid cells. Along with the diminished inflammatory response, the absence of TLR9 led to impaired viral clearance manifested as a higher and prolonged influenza components in myeloid cells including monocytes and macrophages rendering them highly inflammatory. The persistent inflammation driven by infected myeloid cells led to persistent lung injury and impaired recovery in influenza-infected TLR9-/- mice. Further, we show elevated TLR9 activation in the plasma samples of patients with influenza and its association with the disease severity in hospitalized patients, demonstrating its clinical relevance. Overall, we demonstrate an essential role of damage sensing through TLR9 in promoting anti-influenza immunity and inflammatory response.
Collapse
Affiliation(s)
- Jooyoung Kim
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Yifan Yuan
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
- University of Maryland, MD
| | - Karen Agaronyan
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
- Howard Hughes Medical Institute
| | - Amy Zhao
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Victoria D Wang
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Gayatri Gupta
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Heran Essayas
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Ayelet Kaminski
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - John McGovern
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Sheeline Yu
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Samuel Woo
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Chris J. Lee
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Shifa Gandhi
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Tina Saber
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Tayebeh Saleh
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Buqu Hu
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Ying Sun
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Genta Ishikawa
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - William Bain
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
- VA Medical Center, Pittsburgh, PA
| | - John Evankovich
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Lujia Chen
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA 15206, USA
| | - HongDuck Yun
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Erica L. Herzog
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Charles S. Dela Cruz
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
- VA Medical Center, Pittsburgh, PA
| | - Changwan Ryu
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| | - Lokesh Sharma
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, CT
| |
Collapse
|
41
|
Eastburn DJ, White KS, Jayne ND, Camiolo S, Montis G, Ha S, Watson KG, Yeakley JM, McComb J, Seligmann B. High-throughput gene expression analysis with TempO-LINC sensitively resolves complex brain, lung and kidney heterogeneity at single-cell resolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.03.606484. [PMID: 39149288 PMCID: PMC11326174 DOI: 10.1101/2024.08.03.606484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
We report the development and performance of a novel genomics platform, TempO-LINC, for conducting high-throughput transcriptomic analysis on single cells and nuclei. TempO-LINC works by adding cell-identifying molecular barcodes onto highly selective and high-sensitivity gene expression probes within fixed cells, without having to first generate cDNA. Using an instrument-free combinatorial-indexing approach, all probes within the same fixed cell receive an identical barcode, enabling the reconstruction of single-cell gene expression profiles across as few as several hundred cells and up to 100,000+ cells per run. The TempO-LINC approach is easily scalable based on the number of barcodes and rounds of barcoding performed; however, for the experiments reported in this study, the assay utilized over 5.3 million unique barcodes. TempO-LINC has a robust protocol for fixing and banking cells and displays high-sensitivity gene detection from multiple diverse sample types. We show that TempO-LINC has an observed multiplet rate of less than 1.1% and a cell capture rate of ~50%. Although the assay can accurately profile the whole transcriptome (19,683 human or 21,400 mouse genes), it can be targeted to measure only actionable/informative genes and molecular pathways of interest - thereby reducing sequencing requirements. In this study, we applied TempO-LINC to profile the transcriptomes of 89,722 cells across multiple sample types, including nuclei from mouse lung, kidney and brain tissues. The data demonstrated the ability to identify and annotate at least 50 unique cell populations and positively correlate expression of cell type-specific molecular markers within them. TempO-LINC is a robust new single-cell technology that is ideal for large-scale applications/studies across thousands of samples with high data quality.
Collapse
|
42
|
Luo S, Rollins S, Schmitz-Abe K, Tam A, Li Q, Shi J, Lin J, Wang R, Agrawal PB. The solute carrier family 26 member 9 modifies rapidly progressing cystic fibrosis associated with homozygous F508del CFTR mutation. Clin Chim Acta 2024; 561:119765. [PMID: 38852790 DOI: 10.1016/j.cca.2024.119765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/14/2024] [Accepted: 06/04/2024] [Indexed: 06/11/2024]
Abstract
BACKGROUND AND AIMS Cystic fibrosis (CF) is an autosomal recessive disease caused by mutations to the CF transmembrane conductance regulator (CFTR). Symptoms and severity of the disease can be quite variable suggesting modifier genes play an important role. MATERIALS AND METHODS Exome sequencing was performed on six individuals carrying homozygous deltaF508 for CFTR genotype but present with rapidly progressing CF (RPCF). Data was analyzed using an unbiased genome-wide genetic burden test against 3076 controls. Single cell RNA sequencing data from LungMAP was utilized to evaluate unique and co-expression of candidate genes, and structural modeling to evaluate the deleterious effects of identified candidate variants. RESULTS We have identified solute carrier family 26 member 9 (SLC26A9) as a modifier gene to be associated with RPCF. Two rare missense SLC26A9 variants were discovered in three of six individuals deemed to have RPCF: c.229G > A; p.G77S (present in two patients), and c.1885C > T; p.P629S. Co-expression of SLC26A9 and CFTR mRNA is limited across different lung cell types, with the highest level of co-expression seen in human (6.3 %) and mouse (9.0 %) alveolar type 2 (AT2) cells. Structural modeling suggests deleterious effects of these mutations as they are in critical protein domains which might affect the anion transport capability of SLC26A9. CONCLUSION The enrichment of rare and potentially deleterious SLC26A9 mutations in patients with RPCF suggests SLC26A9 may act as an alternative anion transporter in CF and is a modifier gene associated with this lung phenotype.
Collapse
Affiliation(s)
- Shiyu Luo
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine and Holtz Children's Hospital, Jackson Health System, Miami, FL 33136, USA; Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; The Manton Center for Orphan Disease Research, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Stuart Rollins
- Division of Pulmonary Medicine, Boston Children's Hospital, USA; Department of Medicine, Harvard Medical School, USA
| | - Klaus Schmitz-Abe
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine and Holtz Children's Hospital, Jackson Health System, Miami, FL 33136, USA; Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; The Manton Center for Orphan Disease Research, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Amy Tam
- Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; The Manton Center for Orphan Disease Research, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Qifei Li
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine and Holtz Children's Hospital, Jackson Health System, Miami, FL 33136, USA; Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; The Manton Center for Orphan Disease Research, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Jiahai Shi
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jasmine Lin
- Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; The Manton Center for Orphan Disease Research, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Ruobing Wang
- Division of Pulmonary Medicine, Boston Children's Hospital, USA; Department of Medicine, Harvard Medical School, USA; Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA 02115, USA.
| | - Pankaj B Agrawal
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine and Holtz Children's Hospital, Jackson Health System, Miami, FL 33136, USA; Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; The Manton Center for Orphan Disease Research, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
43
|
Yang M, Shulkin N, Gonzalez E, Castillo J, Yan C, Zhang K, Arvanitis L, Borok Z, Wallace WD, Raz D, Torres ETR, Marconett CN. Cell of origin alters myeloid-mediated immunosuppression in lung adenocarcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.19.599651. [PMID: 38948812 PMCID: PMC11213232 DOI: 10.1101/2024.06.19.599651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Solid carcinomas are often highly heterogenous cancers, arising from multiple epithelial cells of origin. Yet, how the cell of origin influences the response of the tumor microenvironment is poorly understood. Lung adenocarcinoma (LUAD) arises in the distal alveolar epithelium which is populated primarily by alveolar epithelial type I (AT1) and type II (AT2) cells. It has been previously reported that Gramd2 + AT1 cells can give rise to a histologically-defined LUAD that is distinct in pathology and transcriptomic identity from that arising from Sftpc + AT2 cells1,2. To determine how cells of origin influence the tumor immune microenvironment (TIME) landscape, we comprehensively characterized transcriptomic, molecular, and cellular states within the TIME of Gramd2 + AT1 and Sftpc + AT2-derived LUAD using KRASG12D oncogenic driver mouse models. Myeloid cells within the Gramd2 + AT1-derived LUAD TIME were increased, specifically, immunoreactive monocytes and tumor associated macrophages (TAMs). In contrast, the Sftpc + AT2 LUAD TIME was enriched for Arginase-1+ myeloid derived suppressor cells (MDSC) and TAMs expressing profiles suggestive of immunosuppressive function. Validation of immune infiltration was performed using flow cytometry, and intercellular interaction analysis between the cells of origin and major myeloid cell populations indicated that cell-type specific markers SFTPD in AT2 cells and CAV1 in AT1 cells mediated unique interactions with myeloid cells of the differential immunosuppressive states within each cell of origin mouse model. Taken together, Gramd2 + AT1-derived LUAD presents with an anti-tumor, immunoreactive TIME, while the TIME of Sftpc + AT2-derived LUAD has hallmarks of immunosuppression. This study suggests that LUAD cell of origin influences the composition and suppression status of the TIME landscape and may hold critical implications for patient response to immunotherapy.
Collapse
Affiliation(s)
- Minxiao Yang
- Department of Integrative Translational Sciences, Beckman Research Institute, City of Hope, Duarte, CA USA 91010
- Department of Surgery, University of Southern California, Los Angeles, CA USA 90089
- Department of Translational Genomics, University of Southern California, Los Angeles, CA USA 90089
| | - Noah Shulkin
- Department of Integrative Translational Sciences, Beckman Research Institute, City of Hope, Duarte, CA USA 91010
| | - Edgar Gonzalez
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA USA 90089
| | - Jonathan Castillo
- Department of Integrative Translational Sciences, Beckman Research Institute, City of Hope, Duarte, CA USA 91010
| | - Chunli Yan
- Department of Surgery, University of Southern California, Los Angeles, CA USA 90089
| | - Keqiang Zhang
- Division of Thoracic Surgery, Department of Surgery, City of Hope National Medical Center, City of Hope, Duarte, CA USA 91010
| | - Leonidas Arvanitis
- Department of Pathology, City of Hope National Medical Center, City of Hope, Duarte, CA USA 91010
| | - Zea Borok
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA USA 92093
| | - W. Dean Wallace
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA USA 90089
| | - Dan Raz
- Division of Thoracic Surgery, Department of Surgery, City of Hope National Medical Center, City of Hope, Duarte, CA USA 91010
| | - Evanthia T. Roussos Torres
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA USA 90089
| | - Crystal N. Marconett
- Department of Integrative Translational Sciences, Beckman Research Institute, City of Hope, Duarte, CA USA 91010
- Department of Surgery, University of Southern California, Los Angeles, CA USA 90089
- Department of Translational Genomics, University of Southern California, Los Angeles, CA USA 90089
| |
Collapse
|
44
|
Palit S, Shrestha AK, Thapa S, L. Grimm S, Coarfa C, Theis F, Simon LM, Shivanna B. Leveraging Integrated RNA Sequencing to Decipher Adrenomedullin's Protective Mechanisms in Experimental Bronchopulmonary Dysplasia. Genes (Basel) 2024; 15:806. [PMID: 38927741 PMCID: PMC11202456 DOI: 10.3390/genes15060806] [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: 05/06/2024] [Revised: 06/12/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease commonly affecting premature infants, with limited therapeutic options and increased long-term consequences. Adrenomedullin (Adm), a proangiogenic peptide hormone, has been found to protect rodents against experimental BPD. This study aims to elucidate the molecular and cellular mechanisms through which Adm influences BPD pathogenesis using a lipopolysaccharide (LPS)-induced model of experimental BPD in mice. Bulk RNA sequencing of Adm-sufficient (wild-type or Adm+/+) and Adm-haplodeficient (Adm+/-) mice lungs, integrated with single-cell RNA sequencing data, revealed distinct gene expression patterns and cell type alterations associated with Adm deficiency and LPS exposure. Notably, computational integration with cell atlas data revealed that Adm-haplodeficient mouse lungs exhibited gene expression signatures characteristic of increased inflammation, natural killer (NK) cell frequency, and decreased endothelial cell and type II pneumocyte frequency. Furthermore, in silico human BPD patient data analysis supported our cell type frequency finding, highlighting elevated NK cells in BPD infants. These results underscore the protective role of Adm in experimental BPD and emphasize that it is a potential therapeutic target for BPD infants with an inflammatory phenotype.
Collapse
Affiliation(s)
- Subarna Palit
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany
| | - Amrit Kumar Shrestha
- Division of Neonatology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA (B.S.)
| | - Shyam Thapa
- Division of Neonatology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA (B.S.)
| | - Sandra L. Grimm
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Fabian Theis
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Department of Mathematics, Technical University of Munich, 85748 Garching, Germany
| | - Lukas M. Simon
- Therapeutic Innovation Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Binoy Shivanna
- Division of Neonatology, Department of Pediatrics, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX 77030, USA (B.S.)
| |
Collapse
|
45
|
Kong Y, Börner K. Publication, funding, and experimental data in support of Human Reference Atlas construction and usage. Sci Data 2024; 11:574. [PMID: 38834597 PMCID: PMC11150433 DOI: 10.1038/s41597-024-03416-8] [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: 01/31/2024] [Accepted: 05/24/2024] [Indexed: 06/06/2024] Open
Abstract
Experts from 18 consortia are collaborating on the Human Reference Atlas (HRA) which aims to map the 37 trillion cells in the healthy human body. Information relevant for HRA construction and usage is held by experts, published in scholarly papers, and captured in experimental data. However, these data sources use different metadata schemas and cannot be cross-searched efficiently. This paper documents the compilation of a dataset, named HRAlit, that links the 136 HRA v1.4 digital objects (31 organs with 4,279 anatomical structures, 1,210 cell types, 2,089 biomarkers) to 583,117 experts; 7,103,180 publications; 896,680 funded projects, and 1,816 experimental datasets. The resulting HRAlit has 22 tables with 20,939,937 records including 6 junction tables with 13,170,651 relationships. The HRAlit can be mined to identify leading experts, major papers, funding trends, or alignment with existing ontologies in support of systematic HRA construction and usage.
Collapse
Affiliation(s)
- Yongxin Kong
- Department of Intelligent Systems Engineering, Luddy School of Informatics, Computing, and Engineering, Indiana University, Bloomington, IN, 47408, USA.
- School of Information Management, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Katy Börner
- Department of Intelligent Systems Engineering, Luddy School of Informatics, Computing, and Engineering, Indiana University, Bloomington, IN, 47408, USA.
| |
Collapse
|
46
|
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: 8] [Impact Index Per Article: 8.0] [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
|
47
|
Yin Y, Koenitzer JR, Patra D, Dietmann S, Bayguinov P, Hagan AS, Ornitz DM. Identification of a myofibroblast differentiation program during neonatal lung development. Development 2024; 151:dev202659. [PMID: 38602479 PMCID: PMC11165721 DOI: 10.1242/dev.202659] [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: 12/26/2023] [Accepted: 03/25/2024] [Indexed: 04/12/2024]
Abstract
Alveologenesis is the final stage of lung development in which the internal surface area of the lung is increased to facilitate efficient gas exchange in the mature organism. The first phase of alveologenesis involves the formation of septal ridges (secondary septae) and the second phase involves thinning of the alveolar septa. Within secondary septa, mesenchymal cells include a transient population of alveolar myofibroblasts (MyoFBs) and a stable but poorly described population of lipid-rich cells that have been referred to as lipofibroblasts or matrix fibroblasts (MatFBs). Using a unique Fgf18CreER lineage trace mouse line, cell sorting, single-cell RNA sequencing and primary cell culture, we have identified multiple subtypes of mesenchymal cells in the neonatal lung, including an immature progenitor cell that gives rise to mature MyoFB. We also show that the endogenous and targeted ROSA26 locus serves as a sensitive reporter for MyoFB maturation. These studies identify a MyoFB differentiation program that is distinct from other mesenchymal cell types and increases the known repertoire of mesenchymal cell types in the neonatal lung.
Collapse
Affiliation(s)
- Yongjun Yin
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jeffrey R. Koenitzer
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Debabrata Patra
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sabine Dietmann
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Institute for Informatics, Data Science and Biostatistics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Peter Bayguinov
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Andrew S. Hagan
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David M. Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
48
|
Ohnishi Y, Masui A, Suezawa T, Mikawa R, Hirai T, Hagiwara M, Gotoh S. Screening of factors inducing alveolar type 1 epithelial cells using human pluripotent stem cells. Stem Cell Reports 2024; 19:529-544. [PMID: 38552636 PMCID: PMC11096435 DOI: 10.1016/j.stemcr.2024.02.009] [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: 09/03/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 04/12/2024] Open
Abstract
Alveolar type 2 (AT2) epithelial cells are tissue stem cells capable of differentiating into alveolar type 1 (AT1) cells for injury repair and maintenance of lung homeostasis. However, the factors involved in human AT2-to-AT1 cell differentiation are not fully understood. Here, we established SFTPCGFP and AGERmCherry-HiBiT dual-reporter induced pluripotent stem cells (iPSCs), which detected AT2-to-AT1 cell differentiation with high sensitivity and identified factors inducing AT1 cell differentiation from AT2 and their progenitor cells. We also established an "on-gel" alveolar epithelial spheroid culture suitable for medium-throughput screening. Among the 274 chemical compounds, several single compounds, including LATS-IN-1, converted AT1 cells from AT2 and their progenitor cells. Moreover, YAP/TAZ signaling activation and AKT signaling suppression synergistically recapitulated the induction of transcriptomic, morphological, and functionally mature AT1 cells. Our findings provide novel insights into human lung development and lung regenerative medicine.
Collapse
Affiliation(s)
- Yuko Ohnishi
- Department of Drug Discovery for Lung Diseases, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan; Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Atsushi Masui
- Department of Drug Discovery for Lung Diseases, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan; Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Takahiro Suezawa
- Department of Drug Discovery for Lung Diseases, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Ryuta Mikawa
- Department of Drug Discovery for Lung Diseases, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan; Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Toyohiro Hirai
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Masatoshi Hagiwara
- Department of Anatomy and Developmental Biology, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Shimpei Gotoh
- Department of Drug Discovery for Lung Diseases, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan; Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan.
| |
Collapse
|
49
|
Basil MC, Alysandratos KD, Kotton DN, Morrisey EE. Lung repair and regeneration: Advanced models and insights into human disease. Cell Stem Cell 2024; 31:439-454. [PMID: 38492572 PMCID: PMC11070171 DOI: 10.1016/j.stem.2024.02.009] [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: 12/05/2023] [Revised: 02/07/2024] [Accepted: 02/22/2024] [Indexed: 03/18/2024]
Abstract
The respiratory system acts as both the primary site of gas exchange and an important sensor and barrier to the external environment. The increase in incidences of respiratory disease over the past decades has highlighted the importance of developing improved therapeutic approaches. This review will summarize recent research on the cellular complexity of the mammalian respiratory system with a focus on gas exchange and immunological defense functions of the lung. Different models of repair and regeneration will be discussed to help interpret human and animal data and spur the investigation of models and assays for future drug development.
Collapse
Affiliation(s)
- Maria C Basil
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn, Children's Hospital of Philadelphia (CHOP) Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Konstantinos-Dionysios Alysandratos
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA.
| | - Darrell N Kotton
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA.
| | - Edward E Morrisey
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn, Children's Hospital of Philadelphia (CHOP) Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
50
|
Zhang H, Mulqueen RM, Iannuzo N, Farrera DO, Polverino F, Galligan JJ, Ledford JG, Adey AC, Cusanovich DA. txci-ATAC-seq: a massive-scale single-cell technique to profile chromatin accessibility. Genome Biol 2024; 25:78. [PMID: 38519979 PMCID: PMC10958877 DOI: 10.1186/s13059-023-03150-1] [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: 05/12/2023] [Accepted: 12/20/2023] [Indexed: 03/25/2024] Open
Abstract
We develop a large-scale single-cell ATAC-seq method by combining Tn5-based pre-indexing with 10× Genomics barcoding, enabling the indexing of up to 200,000 nuclei across multiple samples in a single reaction. We profile 449,953 nuclei across diverse tissues, including the human cortex, mouse brain, human lung, mouse lung, mouse liver, and lung tissue from a club cell secretory protein knockout (CC16-/-) model. Our study of CC16-/- nuclei uncovers previously underappreciated technical artifacts derived from remnant 129 mouse strain genetic material, which cause profound cell-type-specific changes in regulatory elements near many genes, thereby confounding the interpretation of this commonly referenced mouse model.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
- Asthma & Airway Disease Research Center, University of Arizona, Tucson, AZ, USA
| | - Ryan M Mulqueen
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA
| | - Natalie Iannuzo
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Dominique O Farrera
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA
| | - Francesca Polverino
- Asthma & Airway Disease Research Center, University of Arizona, Tucson, AZ, USA
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, University of Arizona, Tucson, AZ, USA
- Banner - University Medicine North, Pulmonary - Clinic F, Tucson, AZ, USA
| | - James J Galligan
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA
| | - Julie G Ledford
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
- Asthma & Airway Disease Research Center, University of Arizona, Tucson, AZ, USA
| | - Andrew C Adey
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA.
- Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, USA.
- Oregon Health & Science University, Knight Cancer Institute, Portland, OR, USA.
- Oregon Health & Science University, Knight Cardiovascular Institute, Portland, OR, USA.
| | - Darren A Cusanovich
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA.
- Asthma & Airway Disease Research Center, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|