1
|
Roth D, Şahin AT, Ling F, Tepho N, Senger CN, Quiroz EJ, Calvert BA, van der Does AM, Güney TG, Glasl S, van Schadewijk A, von Schledorn L, Olmer R, Kanso E, Nawroth JC, Ryan AL. Structure and function relationships of mucociliary clearance in human and rat airways. Nat Commun 2025; 16:2446. [PMID: 40069153 PMCID: PMC11897160 DOI: 10.1038/s41467-025-57667-z] [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: 04/10/2024] [Accepted: 02/27/2025] [Indexed: 03/15/2025] Open
Abstract
Mucociliary clearance is a vital defense mechanism of the human airways, protecting against harmful particles and infections. When this process fails, it contributes to respiratory diseases like chronic obstructive pulmonary disease (COPD) and asthma. While advances in single-cell transcriptomics have revealed the complexity of airway composition, much of what we know about how airway structure impacts clearance relies on animal studies. This limits our ability to create accurate human-based models of airway diseases. Here we show that the airways in female rats and in humans exhibit species-specific differences in the distribution of ciliated and secretory cells as well as in ciliary beat, resulting in significantly higher clearance effectiveness in humans. We further reveal that standard lab-grown cultures exhibit lower clearance effectiveness compared to human airways, and we identify the underlying structural differences. By combining diverse experiments and physics-based modeling, we establish universal benchmarks to assess human airway function, interpret preclinical models, and better understand disease-specific impairments in mucociliary clearance.
Collapse
Affiliation(s)
- Doris Roth
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, Technical University of Munich, Munich, Germany
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Ayşe Tuğçe Şahin
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, Technical University of Munich, Munich, Germany
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Feng Ling
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, Technical University of Munich, Munich, Germany
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Niels Tepho
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, Technical University of Munich, Munich, Germany
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Christiana N Senger
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Hastings Center for Pulmonary Research, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Erik J Quiroz
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Hastings Center for Pulmonary Research, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Ben A Calvert
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Hastings Center for Pulmonary Research, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Anne M van der Does
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | - Tankut G Güney
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, Technical University of Munich, Munich, Germany
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Sarah Glasl
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Annemarie van Schadewijk
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | - Laura von Schledorn
- Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany
- Biomedical Research in End stage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
- REBIRTH-Research Center for Translational and Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Ruth Olmer
- Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany
- Biomedical Research in End stage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
- REBIRTH-Research Center for Translational and Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Eva Kanso
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Janna C Nawroth
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany.
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg, Germany.
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, Technical University of Munich, Munich, Germany.
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany.
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, CA, USA.
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Hastings Center for Pulmonary Research, University of Southern California, Los Angeles, CA, USA.
| | - Amy L Ryan
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Hastings Center for Pulmonary Research, University of Southern California, Los Angeles, CA, USA.
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA.
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
2
|
Roth D, Şahin AT, Ling F, Tepho N, Senger CN, Quiroz EJ, Calvert BA, van der Does AM, Güney TG, Glasl S, van Schadewijk A, von Schledorn L, Olmer R, Kanso E, Nawroth JC, Ryan AL. Structure and Function Relationships of Mucociliary Clearance in Human and Rat Airways. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.12.24.572054. [PMID: 38187619 PMCID: PMC10769450 DOI: 10.1101/2023.12.24.572054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Mucociliary clearance is a vital defense mechanism of the human airways, protecting against harmful particles and infections. When this process fails, it contributes to respiratory diseases like chronic obstructive pulmonary disease (COPD) and asthma. While advances in single-cell transcriptomics have revealed the complexity of airway composition, much of what we know about how airway structure impacts clearance relies on animal studies. This limits our ability to create accurate human-based models of airway diseases. Here we show that the airways in female rats and in humans exhibit species-specific differences in the distribution of ciliated and secretory cells as well as in ciliary beat, resulting in significantly higher clearance effectiveness in humans. We further reveal that standard lab-grown cultures exhibit lower clearance effectiveness compared to human airways, and we identify the underlying structural differences. By combining diverse experiments and physics-based modeling, we establish universal benchmarks to assess human airway function, interpret preclinical models, and better understand disease-specific impairments in mucociliary clearance.
Collapse
Affiliation(s)
- Doris Roth
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, D-85764, Germany
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg D-85764, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, Technical University of Munich, Munich, Munich, D-81675, Germany
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Ayşe Tuğçe Şahin
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, D-85764, Germany
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg D-85764, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, Technical University of Munich, Munich, Munich, D-81675, Germany
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Feng Ling
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, D-85764, Germany
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg D-85764, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, Technical University of Munich, Munich, Munich, D-81675, Germany
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Niels Tepho
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, D-85764, Germany
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg D-85764, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, Technical University of Munich, Munich, Munich, D-81675, Germany
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Christiana N. Senger
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Erik J. Quiroz
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Ben A. Calvert
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Anne M. van der Does
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | - Tankut G. Güney
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, D-85764, Germany
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg D-85764, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, Technical University of Munich, Munich, Munich, D-81675, Germany
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Sarah Glasl
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, D-85764, Germany
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg D-85764, Germany
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Annemarie van Schadewijk
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | - Laura von Schledorn
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, Hannover, D-30625, Germany
- Biomedical Research in End stage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover, D-30625, Germany
- REBIRTH-Research Center for Translational and Regenerative Medicine, Hannover Medical School, Hannover, D-30625, Germany
| | - Ruth Olmer
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, Hannover, D-30625, Germany
- Biomedical Research in End stage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover, D-30625, Germany
- REBIRTH-Research Center for Translational and Regenerative Medicine, Hannover Medical School, Hannover, D-30625, Germany
| | - Eva Kanso
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Janna C. Nawroth
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, D-85764, Germany
- Institute of Biological and Medical Imaging, Bioengineering Center, Helmholtz Zentrum München, Neuherberg D-85764, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, Technical University of Munich, Munich, Munich, D-81675, Germany
- Comprehensive Pneumology Center Munich, German Center for Lung Research (DZL), Munich, Germany
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, CA 90089, USA
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Amy L. Ryan
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, IA 52242, USA
| |
Collapse
|
3
|
Makrides N, Sun E, Mir H, Jiang Z, Wu Y, Serra C, Cardoso WV, Shah NH, Zhang X. Allosteric inhibition rescues hydrocephalus caused by catalytically inactive Shp2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.28.635289. [PMID: 39974929 PMCID: PMC11838390 DOI: 10.1101/2025.01.28.635289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
SHP2, a protein tyrosine phosphatase (PTP) crucial in Ras-MAPK signaling, is associated with various human congenital diseases and cancers. Here, we show that the catalytically inactive Shp2 C459S mutation results in communicating hydrocephalus, similar to the catalytically activating Shp2 E76K and Mek1 DD mutants. Unlike previous mutants, however, Shp2 C459S/+ mutation uniquely affects ciliary development rather than neurogenesis, leading to reduced cilia density and impaired ciliary motility. Differential scanning fluorimetry revealed that SHP2 C459S , SHP2 E76K and SHP2 C459S/E76K mutations all induce an open SHP2 conformation, but only SHP2 C459S leads to aberrant GAB1 phosphorylation in cells expressing wild-type SHP2. This distinctive signaling pattern correlates with our observations in brain ventricular tissues of Shp2 C459S/+ mice, where Erk and Stat3 activities remain normal but Gab1 phosphorylation is elevated. Critically, we show that the hydrocephalus phenotype in Shp2 C459S mice can be mitigated by allosteric inhibition of Shp2. These findings suggest that Shp2-associated hydrocephalus is driven by conformational changes rather than altered catalytic activity. Our results underscore the therapeutic potential of conformation-specific allosteric inhibitors in targeting both catalytically active and inactive SHP2 mutants.
Collapse
Affiliation(s)
- Neoklis Makrides
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Emily Sun
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Hilal Mir
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Ziyuan Jiang
- Department of Chemistry, Columbia University, New York, NY 10027, USA
| | - Yihua Wu
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| | - Carlos Serra
- Columbia Center for Human Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Wellington V Cardoso
- Columbia Center for Human Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Neel H. Shah
- Department of Chemistry, Columbia University, New York, NY 10027, USA
| | - Xin Zhang
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| |
Collapse
|
4
|
Roth D, Şahin AT, Ling F, Senger CN, Quiroz EJ, Calvert BA, van der Does AM, Güney TG, Tepho N, Glasl S, van Schadewijk A, von Schledorn L, Olmer R, Kanso E, Nawroth JC, Ryan AL. STRUCTURE-FUNCTION RELATIONSHIPS OF MUCOCILIARY CLEARANCE IN HUMAN AIRWAYS. RESEARCH SQUARE 2024:rs.3.rs-4164522. [PMID: 38746209 PMCID: PMC11092836 DOI: 10.21203/rs.3.rs-4164522/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Our study focuses on the intricate connection between tissue-level organization and ciliated organ function in humans, particularly in understanding the morphological organization of airways and their role in mucociliary clearance. Mucociliary clearance is a key mechanical defense mechanism of human airways, and clearance failure is associated with many respiratory diseases, including chronic obstructive pulmonary disease (COPD) and asthma. While single-cell transcriptomics have unveiled the cellular complexity of the human airway epithelium, our understanding of the mechanics that link epithelial structure to clearance function mainly stem from animal models. This reliance on animal data limits crucial insights into human airway barrier function and hampers the human-relevant in vitro modeling of airway diseases. This study, for the first time, maps the distribution of ciliated and secretory cell types along the airway tree in both rats and humans, noting species-specific differences in ciliary function and elucidates structural parameters of airway epithelia that predict clearance function in both native and in vitro tissues alike. By uncovering how tissue organization influences ciliary function, we can better understand disruptions in mucociliary clearance, which could have implications for various ciliated organs beyond the airways.
Collapse
Affiliation(s)
- Doris Roth
- Helmholtz Pioneer Campus, Institute of Biological and Medical Imaging, and Member of the German Lung Research Center (DZL CPC-M), Helmholtz Zentrum München, Neuherberg, D-85764, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, D-81675, Germany
| | - Ayşe Tuğçe Şahin
- Helmholtz Pioneer Campus, Institute of Biological and Medical Imaging, and Member of the German Lung Research Center (DZL CPC-M), Helmholtz Zentrum München, Neuherberg, D-85764, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, D-81675, Germany
| | - Feng Ling
- Helmholtz Pioneer Campus, Institute of Biological and Medical Imaging, and Member of the German Lung Research Center (DZL CPC-M), Helmholtz Zentrum München, Neuherberg, D-85764, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, D-81675, Germany
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Christiana N. Senger
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA 90033
| | - Erik J. Quiroz
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA 90033
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Ben A. Calvert
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA 90033
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Anne M. van der Does
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | - Tankut G. Güney
- Helmholtz Pioneer Campus, Institute of Biological and Medical Imaging, and Member of the German Lung Research Center (DZL CPC-M), Helmholtz Zentrum München, Neuherberg, D-85764, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, D-81675, Germany
| | - Niels Tepho
- Helmholtz Pioneer Campus, Institute of Biological and Medical Imaging, and Member of the German Lung Research Center (DZL CPC-M), Helmholtz Zentrum München, Neuherberg, D-85764, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, D-81675, Germany
| | - Sarah Glasl
- Helmholtz Pioneer Campus, Institute of Biological and Medical Imaging, and Member of the German Lung Research Center (DZL CPC-M), Helmholtz Zentrum München, Neuherberg, D-85764, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, D-81675, Germany
| | - Annemarie van Schadewijk
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | - Laura von Schledorn
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, Hannover, D-30625, Germany
- Biomedical Research in End stage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover, D-30625, Germany
- REBIRTH-Research Center for Translational and Regenerative Medicine, Hannover Medical School, Hannover, D-30625, Germany
| | - Ruth Olmer
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), Hannover Medical School, Hannover, D-30625, Germany
- Biomedical Research in End stage and Obstructive Lung Disease (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover, D-30625, Germany
- REBIRTH-Research Center for Translational and Regenerative Medicine, Hannover Medical School, Hannover, D-30625, Germany
| | - Eva Kanso
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Janna C. Nawroth
- Helmholtz Pioneer Campus, Institute of Biological and Medical Imaging, and Member of the German Lung Research Center (DZL CPC-M), Helmholtz Zentrum München, Neuherberg, D-85764, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, D-81675, Germany
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA 90033
- Department of Aerospace and Mechanical Engineering, University of Southern California, Los Angeles, CA 90089, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Amy L. Ryan
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA 90033
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, IA 52242, USA
| |
Collapse
|
5
|
Jain A, Kim BR, Yu W, Moninger TO, Karp PH, Wagner BA, Welsh MJ. Mitochondrial uncoupling proteins protect human airway epithelial ciliated cells from oxidative damage. Proc Natl Acad Sci U S A 2024; 121:e2318771121. [PMID: 38416686 PMCID: PMC10927548 DOI: 10.1073/pnas.2318771121] [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: 10/27/2023] [Accepted: 01/12/2024] [Indexed: 03/01/2024] Open
Abstract
Apical cilia on epithelial cells defend the lung by propelling pathogens and particulates out of the respiratory airways. Ciliated cells produce ATP that powers cilia beating by densely grouping mitochondria just beneath the apical membrane. However, this efficient localization comes at a cost because electrons leaked during oxidative phosphorylation react with molecular oxygen to form superoxide, and thus, the cluster of mitochondria creates a hotspot for oxidant production. The relatively high oxygen concentration overlying airway epithelia further intensifies the risk of generating superoxide. Thus, airway ciliated cells face a unique challenge of producing harmful levels of oxidants. However, surprisingly, highly ciliated epithelia produce less reactive oxygen species (ROS) than epithelia with few ciliated cells. Compared to other airway cell types, ciliated cells express high levels of mitochondrial uncoupling proteins, UCP2 and UCP5. These proteins decrease mitochondrial protonmotive force and thereby reduce production of ROS. As a result, lipid peroxidation, a marker of oxidant injury, decreases. However, mitochondrial uncoupling proteins exact a price for decreasing oxidant production; they decrease the fraction of mitochondrial respiration that generates ATP. These findings indicate that ciliated cells sacrifice mitochondrial efficiency in exchange for safety from damaging oxidation. Employing uncoupling proteins to prevent oxidant production, instead of relying solely on antioxidants to decrease postproduction oxidant levels, may offer an advantage for targeting a local area of intense ROS generation.
Collapse
Affiliation(s)
- Akansha Jain
- Department of Internal Medicine, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA52242
- Department of Molecular Physiology and Biophysics, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA52242
| | - Bo Ram Kim
- Department of Internal Medicine, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA52242
- HHMI, Department of Internal Medicine, University of Iowa, Iowa City, IA52242
| | - Wenjie Yu
- Department of Internal Medicine, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA52242
- HHMI, Department of Internal Medicine, University of Iowa, Iowa City, IA52242
| | - Thomas O. Moninger
- Department of Internal Medicine, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA52242
| | - Philip H. Karp
- Department of Internal Medicine, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA52242
- HHMI, Department of Internal Medicine, University of Iowa, Iowa City, IA52242
| | - Brett A. Wagner
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA52242
| | - Michael J. Welsh
- Department of Internal Medicine, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA52242
- Department of Molecular Physiology and Biophysics, Pappajohn Biomedical Institute, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA52242
- HHMI, Department of Internal Medicine, University of Iowa, Iowa City, IA52242
| |
Collapse
|
6
|
Petit LMG, Belgacemi R, Ancel J, Saber Cherif L, Polette M, Perotin JM, Spassky N, Pilette C, Al Alam D, Deslée G, Dormoy V. Airway ciliated cells in adult lung homeostasis and COPD. Eur Respir Rev 2023; 32:230106. [PMID: 38056888 PMCID: PMC10698550 DOI: 10.1183/16000617.0106-2023] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 10/07/2023] [Indexed: 12/08/2023] Open
Abstract
Cilia are organelles emanating from the cell surface, consisting of an axoneme of microtubules that extends from a basal body derived from the centrioles. They are either isolated and nonmotile (primary cilia), or grouped and motile (motile cilia). Cilia are at the centre of fundamental sensory processes and are involved in a wide range of human disorders. Pulmonary cilia include motile cilia lining the epithelial cells of the conductive airways to orchestrate mucociliary clearance, and primary cilia found on nondifferentiated epithelial and mesenchymal cells acting as sensors and cell cycle keepers. Whereas cilia are essential along the airways, their regulatory molecular mechanisms remain poorly understood, resulting in a lack of therapeutic strategies targeting their structure or functions. This review summarises the current knowledge on cilia in the context of lung homeostasis and COPD to provide a comprehensive overview of the (patho)biology of cilia in respiratory medicine with a particular emphasis on COPD.
Collapse
Affiliation(s)
- Laure M G Petit
- Université de Reims Champagne-Ardenne, INSERM, P3Cell UMR-S1250, SFR CAP-SANTE, Reims, France
| | - Randa Belgacemi
- Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Julien Ancel
- Université de Reims Champagne-Ardenne, INSERM, P3Cell UMR-S1250, SFR CAP-SANTE, Reims, France
- CHU Reims, Hôpital Maison Blanche, Service de Pneumologie, Reims, France
| | - Lynda Saber Cherif
- Université de Reims Champagne-Ardenne, INSERM, P3Cell UMR-S1250, SFR CAP-SANTE, Reims, France
| | - Myriam Polette
- Université de Reims Champagne-Ardenne, INSERM, P3Cell UMR-S1250, SFR CAP-SANTE, Reims, France
- CHU Reims, Hôpital Maison Blanche, Laboratoire de Biopathologie, Reims, France
| | - Jeanne-Marie Perotin
- Université de Reims Champagne-Ardenne, INSERM, P3Cell UMR-S1250, SFR CAP-SANTE, Reims, France
- CHU Reims, Hôpital Maison Blanche, Service de Pneumologie, Reims, France
| | - Nathalie Spassky
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, PSL Université Paris, Paris, France
| | - Charles Pilette
- Université Catholique de Louvain (UCL), Institute of Experimental and Clinical Research - Pole of Pneumology, ENT, Dermatology and Pulmonology Department, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Denise Al Alam
- Lundquist Institute for Biomedical Innovation, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Gaëtan Deslée
- Université de Reims Champagne-Ardenne, INSERM, P3Cell UMR-S1250, SFR CAP-SANTE, Reims, France
- CHU Reims, Hôpital Maison Blanche, Service de Pneumologie, Reims, France
| | - Valérian Dormoy
- Université de Reims Champagne-Ardenne, INSERM, P3Cell UMR-S1250, SFR CAP-SANTE, Reims, France
| |
Collapse
|
7
|
Nawroth JC, Roth D, van Schadewijk A, Ravi A, Maulana TI, Senger CN, van Riet S, Ninaber DK, de Waal AM, Kraft D, Hiemstra PS, Ryan AL, van der Does AM. Breathing on chip: Dynamic flow and stretch accelerate mucociliary maturation of airway epithelium in vitro. Mater Today Bio 2023; 21:100713. [PMID: 37455819 PMCID: PMC10339259 DOI: 10.1016/j.mtbio.2023.100713] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 07/18/2023] Open
Abstract
Human lung function is intricately linked to blood flow and breathing cycles, but it remains unknown how these dynamic cues shape human airway epithelial biology. Here we report a state-of-the-art protocol for studying the effects of dynamic medium and airflow as well as stretch on human primary airway epithelial cell differentiation and maturation, including mucociliary clearance, using an organ-on-chip device. Perfused epithelial cell cultures displayed accelerated maturation and polarization of mucociliary clearance, and changes in specific cell-types when compared to traditional (static) culture methods. Additional application of airflow and stretch to the airway chip resulted in an increase in polarization of mucociliary clearance towards the applied flow, reduced baseline secretion of interleukin-8 and other inflammatory proteins, and reduced gene expression of matrix metalloproteinase (MMP) 9, fibronectin, and other extracellular matrix factors. These results indicate that breathing-like mechanical stimuli are important modulators of airway epithelial cell differentiation and maturation and that their fine-tuned application could generate models of specific epithelial pathologies, including mucociliary (dys)function.
Collapse
Affiliation(s)
- Janna C. Nawroth
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, USA
- Emulate Inc., Boston, MA, USA
- Helmholtz Pioneer Campus and Institute for Biological and Medical Imaging, Helmholtz Zentrum München (GmbH), Neuherberg, Germany
- Chair of Biological Imaging at the Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
| | | | - Annemarie van Schadewijk
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | - Abilash Ravi
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Christiana N. Senger
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Sander van Riet
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | - Dennis K. Ninaber
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | - Amy M. de Waal
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Dorothea Kraft
- Helmholtz Pioneer Campus and Institute for Biological and Medical Imaging, Helmholtz Zentrum München (GmbH), Neuherberg, Germany
| | - Pieter S. Hiemstra
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | - Amy L. Ryan
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cells and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Anne M. van der Does
- PulmoScience Lab, Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
8
|
Horani A, Gupta DK, Xu J, Xu H, del Carmen Puga-Molina L, Santi CM, Ramagiri S, Brennan SK, Pan J, Koenitzer JR, Huang T, Hyland RM, Gunsten SP, Tzeng SC, Strahle JM, Mill P, Mahjoub MR, Dutcher SK, Brody SL. The effect of Dnaaf5 gene dosage on primary ciliary dyskinesia phenotypes. JCI Insight 2023; 8:e168836. [PMID: 37104040 PMCID: PMC10393236 DOI: 10.1172/jci.insight.168836] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 04/20/2023] [Indexed: 04/28/2023] Open
Abstract
DNAAF5 is a dynein motor assembly factor associated with the autosomal heterogenic recessive condition of motile cilia, primary ciliary dyskinesia (PCD). The effects of allele heterozygosity on motile cilia function are unknown. We used CRISPR-Cas9 genome editing in mice to recreate a human missense variant identified in patients with mild PCD and a second, frameshift-null deletion in Dnaaf5. Litters with Dnaaf5 heteroallelic variants showed distinct missense and null gene dosage effects. Homozygosity for the null Dnaaf5 alleles was embryonic lethal. Compound heterozygous animals with the missense and null alleles showed severe disease manifesting as hydrocephalus and early lethality. However, animals homozygous for the missense mutation had improved survival, with partially preserved cilia function and motor assembly observed by ultrastructure analysis. Notably, the same variant alleles exhibited divergent cilia function across different multiciliated tissues. Proteomic analysis of isolated airway cilia from mutant mice revealed reduction in some axonemal regulatory and structural proteins not previously reported in DNAAF5 variants. Transcriptional analysis of mouse and human mutant cells showed increased expression of genes coding for axonemal proteins. These findings suggest allele-specific and tissue-specific molecular requirements for cilia motor assembly that may affect disease phenotypes and clinical trajectory in motile ciliopathies.
Collapse
Affiliation(s)
- Amjad Horani
- Department of Pediatrics
- Department of Cell Biology and Physiology
| | | | | | | | | | | | - Sruthi Ramagiri
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | | | | | | | | | | | - Jennifer M. Strahle
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Pleasantine Mill
- MRC Human Genetics Unit, University of Edinburgh, Edinburgh, United Kingdom
| | - Moe R. Mahjoub
- Department of Cell Biology and Physiology
- Department of Medicine
| | - Susan K. Dutcher
- Department of Cell Biology and Physiology
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | | |
Collapse
|
9
|
Lee C, Ma Y, Tu F, Wallingford JB. Ordered deployment of distinct ciliary beating machines in growing axonemes of vertebrate multiciliated cells. Differentiation 2023; 131:49-58. [PMID: 37120964 PMCID: PMC10523804 DOI: 10.1016/j.diff.2023.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 02/14/2023] [Accepted: 03/11/2023] [Indexed: 05/02/2023]
Abstract
The beating of motile cilia requires the coordinated action of diverse machineries that include not only the axonemal dynein arms, but also the central apparatus, the radial spokes, and the microtubule inner proteins. These machines exhibit complex radial and proximodistal patterns in mature axonemes, but little is known about the interplay between them during motile ciliogenesis. Here, we describe and quantify the relative rates of axonemal deployment for these diverse cilia beating machineries during the final stages of differentiation of Xenopus epidermal multiciliated cells.
Collapse
Affiliation(s)
- Chanjae Lee
- Dept. of Molecular Biosciences, University of Texas at Austin, USA
| | - Yun Ma
- Dept. of Molecular Biosciences, University of Texas at Austin, USA
| | - Fan Tu
- Dept. of Molecular Biosciences, University of Texas at Austin, USA
| | | |
Collapse
|
10
|
Wong SL, Kardia E, Vijayan A, Umashankar B, Pandzic E, Zhong L, Jaffe A, Waters SA. Molecular and Functional Characteristics of Airway Epithelium under Chronic Hypoxia. Int J Mol Sci 2023; 24:ijms24076475. [PMID: 37047450 PMCID: PMC10095024 DOI: 10.3390/ijms24076475] [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: 03/03/2023] [Revised: 03/20/2023] [Accepted: 03/24/2023] [Indexed: 04/14/2023] Open
Abstract
Localized and chronic hypoxia of airway mucosa is a common feature of progressive respiratory diseases, including cystic fibrosis (CF). However, the impact of prolonged hypoxia on airway stem cell function and differentiated epithelium is not well elucidated. Acute hypoxia alters the transcription and translation of many genes, including the CF transmembrane conductance regulator (CFTR). CFTR-targeted therapies (modulators) have not been investigated in vitro under chronic hypoxic conditions found in CF airways in vivo. Nasal epithelial cells (hNECs) derived from eight CF and three non-CF participants were expanded and differentiated at the air-liquid interface (26-30 days) at ambient and 2% oxygen tension (hypoxia). Morphology, global proteomics (LC-MS/MS) and function (barrier integrity, cilia motility and ion transport) of basal stem cells and differentiated cultures were assessed. hNECs expanded at chronic hypoxia, demonstrating epithelial cobblestone morphology and a similar proliferation rate to hNECs expanded at normoxia. Hypoxia-inducible proteins and pathways in stem cells and differentiated cultures were identified. Despite the stem cells' plasticity and adaptation to chronic hypoxia, the differentiated epithelium was significantly thinner with reduced barrier integrity. Stem cell lineage commitment shifted to a more secretory epithelial phenotype. Motile cilia abundance, length, beat frequency and coordination were significantly negatively modulated. Chronic hypoxia reduces the activity of epithelial sodium and CFTR ion channels. CFTR modulator drug response was diminished. Our findings shed light on the molecular pathophysiology of hypoxia and its implications in CF. Targeting hypoxia can be a strategy to augment mucosal function and may provide a means to enhance the efficacy of CFTR modulators.
Collapse
Affiliation(s)
- Sharon L Wong
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
- Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales, Sydney, NSW 2052, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Egi Kardia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
- Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales, Sydney, NSW 2052, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Abhishek Vijayan
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
- Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales, Sydney, NSW 2052, Australia
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Bala Umashankar
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
- Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales, Sydney, NSW 2052, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Elvis Pandzic
- Katharina Gaus Light Microscopy Facility, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW 2052, Australia
| | - Ling Zhong
- Bioanalytical Mass Spectrometry Facility, University of New South Wales, Sydney, NSW 2052, Australia
| | - Adam Jaffe
- Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales, Sydney, NSW 2052, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
- Department of Respiratory Medicine, Sydney Children's Hospital, Sydney, NSW 2052, Australia
| | - Shafagh A Waters
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
- Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales, Sydney, NSW 2052, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
- Department of Respiratory Medicine, Sydney Children's Hospital, Sydney, NSW 2052, Australia
| |
Collapse
|
11
|
Rationalized deep learning super-resolution microscopy for sustained live imaging of rapid subcellular processes. Nat Biotechnol 2023; 41:367-377. [PMID: 36203012 DOI: 10.1038/s41587-022-01471-3] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 08/12/2022] [Indexed: 12/12/2022]
Abstract
The goal when imaging bioprocesses with optical microscopy is to acquire the most spatiotemporal information with the least invasiveness. Deep neural networks have substantially improved optical microscopy, including image super-resolution and restoration, but still have substantial potential for artifacts. In this study, we developed rationalized deep learning (rDL) for structured illumination microscopy and lattice light sheet microscopy (LLSM) by incorporating prior knowledge of illumination patterns and, thereby, rationally guiding the network to denoise raw images. Here we demonstrate that rDL structured illumination microscopy eliminates spectral bias-induced resolution degradation and reduces model uncertainty by five-fold, improving the super-resolution information by more than ten-fold over other computational approaches. Moreover, rDL applied to LLSM enables self-supervised training by using the spatial or temporal continuity of noisy data itself, yielding results similar to those of supervised methods. We demonstrate the utility of rDL by imaging the rapid kinetics of motile cilia, nucleolar protein condensation during light-sensitive mitosis and long-term interactions between membranous and membrane-less organelles.
Collapse
|
12
|
Ringers C, Bialonski S, Ege M, Solovev A, Hansen JN, Jeong I, Friedrich BM, Jurisch-Yaksi N. Novel analytical tools reveal that local synchronization of cilia coincides with tissue-scale metachronal waves in zebrafish multiciliated epithelia. eLife 2023; 12:77701. [PMID: 36700548 PMCID: PMC9940908 DOI: 10.7554/elife.77701] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 01/25/2023] [Indexed: 01/27/2023] Open
Abstract
Motile cilia are hair-like cell extensions that beat periodically to generate fluid flow along various epithelial tissues within the body. In dense multiciliated carpets, cilia were shown to exhibit a remarkable coordination of their beat in the form of traveling metachronal waves, a phenomenon which supposedly enhances fluid transport. Yet, how cilia coordinate their regular beat in multiciliated epithelia to move fluids remains insufficiently understood, particularly due to lack of rigorous quantification. We combine experiments, novel analysis tools, and theory to address this knowledge gap. To investigate collective dynamics of cilia, we studied zebrafish multiciliated epithelia in the nose and the brain. We focused mainly on the zebrafish nose, due to its conserved properties with other ciliated tissues and its superior accessibility for non-invasive imaging. We revealed that cilia are synchronized only locally and that the size of local synchronization domains increases with the viscosity of the surrounding medium. Even though synchronization is local only, we observed global patterns of traveling metachronal waves across the zebrafish multiciliated epithelium. Intriguingly, these global wave direction patterns are conserved across individual fish, but different for left and right noses, unveiling a chiral asymmetry of metachronal coordination. To understand the implications of synchronization for fluid pumping, we used a computational model of a regular array of cilia. We found that local metachronal synchronization prevents steric collisions, i.e., cilia colliding with each other, and improves fluid pumping in dense cilia carpets, but hardly affects the direction of fluid flow. In conclusion, we show that local synchronization together with tissue-scale cilia alignment coincide and generate metachronal wave patterns in multiciliated epithelia, which enhance their physiological function of fluid pumping.
Collapse
Affiliation(s)
- Christa Ringers
- Department of Clinical and Molecular Medicine, Norwegian University of Science and TechnologyTrondheimNorway
- Kavli Institute for Systems, Neuroscience and Centre for Neural Computation, Norwegian University of Science and TechnologyTrondheimNorway
- Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala UniversityUppsalaSweden
| | - Stephan Bialonski
- Institute for Data-Driven Technologies, Aachen University of Applied SciencesJülichGermany
- Center for Advancing Electronics, Technical University DresdenDresdenGermany
| | - Mert Ege
- Department of Clinical and Molecular Medicine, Norwegian University of Science and TechnologyTrondheimNorway
| | - Anton Solovev
- Center for Advancing Electronics, Technical University DresdenDresdenGermany
- Cluster of Excellence 'Physics of Life', Technical University DresdenDresdenGermany
| | - Jan Niklas Hansen
- Kavli Institute for Systems, Neuroscience and Centre for Neural Computation, Norwegian University of Science and TechnologyTrondheimNorway
| | - Inyoung Jeong
- Department of Clinical and Molecular Medicine, Norwegian University of Science and TechnologyTrondheimNorway
| | - Benjamin M Friedrich
- Center for Advancing Electronics, Technical University DresdenDresdenGermany
- Cluster of Excellence 'Physics of Life', Technical University DresdenDresdenGermany
| | - Nathalie Jurisch-Yaksi
- Department of Clinical and Molecular Medicine, Norwegian University of Science and TechnologyTrondheimNorway
- Kavli Institute for Systems, Neuroscience and Centre for Neural Computation, Norwegian University of Science and TechnologyTrondheimNorway
| |
Collapse
|
13
|
Horani A, Gupta DK, Xu J, Xu H, Del Carmen Puga-Molina L, Santi CM, Ramagiri S, Brennen SK, Pan J, Huang T, Hyland RM, Gunsten SP, Tzeng SC, Strahle JM, Mill P, Mahjoub MR, Dutcher SK, Brody SL. The effect of Dnaaf5 gene dosage on primary ciliary dyskinesia phenotypes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.13.523966. [PMID: 36712068 PMCID: PMC9882222 DOI: 10.1101/2023.01.13.523966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
DNAAF5 is a dynein motor assembly factor associated with the autosomal heterogenic recessive condition of motile cilia, primary ciliary dyskinesia (PCD). The effects of allele heterozygosity on motile cilia function are unknown. We used CRISPR-Cas9 genome editing in mice to recreate a human missense variant identified in patients with mild PCD and a second, frameshift null deletion in Dnaaf5 . Litters with Dnaaf5 heteroallelic variants showed distinct missense and null gene dosage effects. Homozygosity for the null Dnaaf5 alleles was embryonic lethal. Compound heterozygous animals with the missense and null alleles showed severe disease manifesting as hydrocephalus and early lethality. However, animals homozygous for the missense mutation had improved survival, with partial preserved cilia function and motor assembly observed by ultrastructure analysis. Notably, the same variant alleles exhibited divergent cilia function across different multiciliated tissues. Proteomic analysis of isolated airway cilia from mutant mice revealed reduction in some axonemal regulatory and structural proteins not previously reported in DNAAF5 variants. While transcriptional analysis of mouse and human mutant cells showed increased expression of genes coding for axonemal proteins. Together, these findings suggest allele-specific and tissue-specific molecular requirements for cilia motor assembly that may affect disease phenotypes and clinical trajectory in motile ciliopathies. Brief Summary A mouse model of human DNAAF5 primary ciliary dyskinesia variants reveals gene dosage effects of mutant alleles and tissue-specific molecular requirements for cilia motor assembly.
Collapse
|
14
|
Cho JH, Li ZA, Zhu L, Muegge BD, Roseman HF, Lee EY, Utterback T, Woodhams LG, Bayly PV, Hughes JW. Islet primary cilia motility controls insulin secretion. SCIENCE ADVANCES 2022; 8:eabq8486. [PMID: 36149960 PMCID: PMC9506710 DOI: 10.1126/sciadv.abq8486] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 07/13/2022] [Indexed: 06/16/2023]
Abstract
Primary cilia are specialized cell-surface organelles that mediate sensory perception and, in contrast to motile cilia and flagella, are thought to lack motility function. Here, we show that primary cilia in human and mouse pancreatic islets exhibit movement that is required for glucose-dependent insulin secretion. Islet primary cilia contain motor proteins conserved from those found in classic motile cilia, and their three-dimensional motion is dynein-driven and dependent on adenosine 5'-triphosphate and glucose metabolism. Inhibition of cilia motion blocks beta cell calcium influx and insulin secretion. Human beta cells have enriched ciliary gene expression, and motile cilia genes are altered in type 2 diabetes. Our findings redefine primary cilia as dynamic structures having both sensory and motile function and establish that pancreatic islet cilia movement plays a regulatory role in insulin secretion.
Collapse
Affiliation(s)
- Jung Hoon Cho
- Department of Medicine, Washington University School of Medicine, 660 South Euclid Ave, St. Louis, MO, USA
| | - Zipeng A. Li
- Department of Medicine, Washington University School of Medicine, 660 South Euclid Ave, St. Louis, MO, USA
| | - Lifei Zhu
- Department of Medicine, Washington University School of Medicine, 660 South Euclid Ave, St. Louis, MO, USA
| | - Brian D. Muegge
- Department of Medicine, Washington University School of Medicine, 660 South Euclid Ave, St. Louis, MO, USA
- Department of Medicine, VA Medical Center, 915 North Grand Blvd, St. Louis, MO, USA
| | - Henry F. Roseman
- Department of Medicine, Washington University School of Medicine, 660 South Euclid Ave, St. Louis, MO, USA
| | - Eun Young Lee
- Department of Medicine, Washington University School of Medicine, 660 South Euclid Ave, St. Louis, MO, USA
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Toby Utterback
- Department of Mechanical Engineering and Materials Science, Washington University McKelvey School of Engineering, 1 Brookings Drive, St. Louis, MO, USA
| | - Louis G. Woodhams
- Department of Mechanical Engineering and Materials Science, Washington University McKelvey School of Engineering, 1 Brookings Drive, St. Louis, MO, USA
| | - Philip V. Bayly
- Department of Mechanical Engineering and Materials Science, Washington University McKelvey School of Engineering, 1 Brookings Drive, St. Louis, MO, USA
| | - Jing W. Hughes
- Department of Medicine, Washington University School of Medicine, 660 South Euclid Ave, St. Louis, MO, USA
| |
Collapse
|
15
|
Serra CF, Liu H, Qian J, Mori M, Lu J, Cardoso WV. Prominin 1 and Notch regulate ciliary length and dynamics in multiciliated cells of the airway epithelium. iScience 2022; 25:104751. [PMID: 35942101 PMCID: PMC9356082 DOI: 10.1016/j.isci.2022.104751] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 06/06/2022] [Accepted: 07/08/2022] [Indexed: 11/29/2022] Open
Abstract
Differences in ciliary morphology and dynamics among multiciliated cells of the respiratory tract contribute to efficient mucociliary clearance. Nevertheless, little is known about how these phenotypic differences are established. We show that Prominin 1 (Prom1), a transmembrane protein widely used as stem cell marker, is crucial to this process. During airway differentiation, Prom1 becomes restricted to multiciliated cells, where it is expressed at distinct levels along the proximal-distal axis of the airways. Prom1 is induced by Notch in multiciliated cells, and Notch inactivation abolishes this gradient of expression. Prom1 was not required for multicilia formation, but when inactivated resulted in longer cilia that beat at a lower frequency. Disruption of Notch resulted in opposite effects and suggested that Notch fine-tunes Prom1 levels to regulate the multiciliated cell phenotype and generate diversity among these cells. This mechanism could contribute to the innate defense of the lung and help prevent pulmonary disease.
Collapse
Affiliation(s)
- Carlos F.H. Serra
- Columbia Center for Human Development, Department of Medicine, Pulmonary Allergy Critical Care, Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA,Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal,ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Helu Liu
- Columbia Center for Human Development, Department of Medicine, Pulmonary Allergy Critical Care, Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Jun Qian
- Columbia Center for Human Development, Department of Medicine, Pulmonary Allergy Critical Care, Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Munemasa Mori
- Columbia Center for Human Development, Department of Medicine, Pulmonary Allergy Critical Care, Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Jining Lu
- Columbia Center for Human Development, Department of Medicine, Pulmonary Allergy Critical Care, Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Wellington V. Cardoso
- Columbia Center for Human Development, Department of Medicine, Pulmonary Allergy Critical Care, Department of Genetics and Development, Columbia University Medical Center, New York, NY 10032, USA,Corresponding author
| |
Collapse
|
16
|
Bearce EA, Irons ZH, Craig SB, Kuhns CJ, Sabazali C, Farnsworth DR, Miller AC, Grimes DT. Daw1 regulates the timely onset of cilia motility during development. Development 2022; 149:275714. [PMID: 35708608 PMCID: PMC9270974 DOI: 10.1242/dev.200017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 05/05/2022] [Indexed: 12/15/2022]
Abstract
Motile cilia generate cell propulsion and extracellular fluid flows that are crucial for airway clearance, fertility and left-right patterning. Motility is powered by dynein arm complexes that are assembled in the cytoplasm then imported into the cilium. Studies in Chlamydomonas reinhardtii showed that ODA16 is a cofactor which promotes dynein arm import. Here, we demonstrate that the zebrafish homolog of ODA16, Daw1, facilitates the onset of robust cilia motility during development. Without Daw1, cilia showed markedly reduced motility during early development; however, motility subsequently increased to attain close to wild-type levels. Delayed motility onset led to differential effects on early and late cilia-dependent processes. Remarkably, abnormal body axis curves, which formed during the first day of development due to reduced cilia motility, self-corrected when motility later reached wild-type levels. Zebrafish larva therefore possess the ability to survey and correct body shape abnormalities. This work defines Daw1 as a factor which promotes the onset of timely cilia motility and can explain why human patients harboring DAW1 mutations exhibit significant laterality perturbations but mild airway and fertility complications. Summary: Daw1 promotes the onset of timely cilia motility for robust axial straightening during zebrafish development.
Collapse
Affiliation(s)
- Elizabeth A Bearce
- Institute of Molecular Biology, Department of Biology, University of Oregon, Eugene, OR 97403, USA
| | - Zoe H Irons
- Institute of Molecular Biology, Department of Biology, University of Oregon, Eugene, OR 97403, USA
| | - Samuel B Craig
- Institute of Molecular Biology, Department of Biology, University of Oregon, Eugene, OR 97403, USA
| | - Colin J Kuhns
- Institute of Molecular Biology, Department of Biology, University of Oregon, Eugene, OR 97403, USA
| | - Cynthia Sabazali
- Institute of Molecular Biology, Department of Biology, University of Oregon, Eugene, OR 97403, USA
| | - Dylan R Farnsworth
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, OR 97403, USA
| | - Adam C Miller
- Institute of Neuroscience, Department of Biology, University of Oregon, Eugene, OR 97403, USA
| | - Daniel T Grimes
- Institute of Molecular Biology, Department of Biology, University of Oregon, Eugene, OR 97403, USA
| |
Collapse
|
17
|
Jackson CL, Bottier M. Methods for the assessment of human airway ciliary function. Eur Respir J 2022; 60:13993003.02300-2021. [PMID: 35595315 DOI: 10.1183/13993003.02300-2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 04/19/2022] [Indexed: 11/05/2022]
Affiliation(s)
- Claire L Jackson
- Primary Ciliary Dyskinesia Centre, NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, UK .,School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| | - Mathieu Bottier
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| |
Collapse
|
18
|
Katsumata M, Fujisawa T, Kamiya Y, Tanaka Y, Kamiya C, Inoue Y, Hozumi H, Karayama M, Suzuki Y, Furuhashi K, Enomoto N, Nakamura Y, Inui N, Maekawa M, Setou M, Watanabe H, Ikegami K, Suda T. Effects of long-acting muscarinic antagonists on promoting ciliary function in airway epithelium. BMC Pulm Med 2022; 22:186. [PMID: 35527239 PMCID: PMC9080152 DOI: 10.1186/s12890-022-01983-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 05/05/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Mucociliary clearance (MCC) is an essential defense mechanism in airway epithelia for removing pathogens from the respiratory tract. Impaired ciliary functions and MCC have been demonstrated in asthma and chronic obstructive pulmonary disease (COPD). Long-acting muscarinic antagonists (LAMAs) are a major class of inhaled bronchodilators, which are used for treating asthma and COPD; however, the effects of LAMAs on ciliary function remain unclear. This study aimed to identify the effects of LAMAs on airway ciliary functions.
Methods
Wild-type BALB/c mice were treated with daily intranasal administrations of glycopyrronium for 7 days, and tracheal samples were collected. Cilia-driven flow and ciliary activity, including ciliary beat frequency (CBF), ciliary beating amplitude, effective stroke velocity, recovery stroke velocity and the ratio of effective stroke velocity to recovery stroke velocity, were analyzed by imaging techniques. Using in vitro murine models, tracheal tissues were transiently cultured in media with/without LAMAs, glycopyrronium or tiotropium, for 60 min. Cilia-driven flow and ciliary activity were then analyzed. Well-differentiated normal human bronchial epithelial (NHBE) cells were treated with glycopyrronium, tiotropium, or vehicle for 60 min, and CBF was evaluated. Several mechanistic analyses were performed.
Results
Intranasal glycopyrronium administration for 7 days significantly increased cilia-driven flow and ciliary activity in murine airway epithelium. In the murine tracheal organ culture models, treatment with glycopyrronium or tiotropium for 60 min significantly increased cilia-driven flow and ciliary activity in airway epithelium. Further, we confirmed that 60-min treatment with glycopyrronium or tiotropium directly increased CBF in well-differentiated NHBE cells. In the mechanistic analyses, neither treatment with glycopyrronium nor tiotropium affected intracellular calcium ion concentrations in well-differentiated NHBE cells. Glycopyrronium did not increase protein kinase A activity in well-differentiated NHBE cells. Moreover, glycopyrronium had no effect on extracellular adenosine triphosphate concentration.
Conclusions
LAMAs exert a direct effect on airway epithelium to enhance ciliary function, which may improve impaired MCC in asthma and COPD. Further investigations are warranted to elucidate the underlying mechanisms of the effects of LAMAs on the promotion of airway ciliary function.
Collapse
|
19
|
Choi WJ, Yoon JK, Paulson B, Lee CH, Yim JJ, Kim JI, Kim JK. Image Correlation-Based Method to Assess Ciliary Beat Frequency in Human Airway Organoids. IEEE TRANSACTIONS ON MEDICAL IMAGING 2022; 41:374-382. [PMID: 34524956 DOI: 10.1109/tmi.2021.3112992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Ciliary movements within the human airway are essential for maintaining a clean lung environment. Motile cilia have a characteristic ciliary beat frequency (CBF). However, CBF measurement with current video microscopic techniques can be error-prone due to the use of the single-point Fourier transformation, which is often biased for ciliary measurements. Herein, we describe a new video microscopy technique that harnesses a metric of motion-contrast imaging and image correlation for CBF analysis. It can provide objective and selective CBF measurements for individual motile cilia and generate CBF maps for the imaged area. The measurement performance of our methodology was validated with in vitro human airway organoid models that simulated an actual human airway epithelium. The CBF determined for the region of interest (ROI) was equal to that obtained with manual counting. The signal redundancy problem of conventional methods was not observed. Moreover, the obtained CBF measurements were robust to optical focal shifts, and exhibited spatial heterogeneity and temperature dependence. This technique can be used to evaluate ciliary movement in respiratory tracts and determine whether it is non-synchronous or aperiodic in patients. Therefore, our observations suggest that the proposed method can be clinically adapted as a screening tool to diagnose ciliopathies.
Collapse
|
20
|
Haward F, Maslon MM, Yeyati PL, Bellora N, Hansen JN, Aitken S, Lawson J, von Kriegsheim A, Wachten D, Mill P, Adams IR, Caceres JF. Nucleo-cytoplasmic shuttling of splicing factor SRSF1 is required for development and cilia function. eLife 2021; 10:e65104. [PMID: 34338635 PMCID: PMC8352595 DOI: 10.7554/elife.65104] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 07/30/2021] [Indexed: 12/02/2022] Open
Abstract
Shuttling RNA-binding proteins coordinate nuclear and cytoplasmic steps of gene expression. The SR family proteins regulate RNA splicing in the nucleus and a subset of them, including SRSF1, shuttles between the nucleus and cytoplasm affecting post-splicing processes. However, the physiological significance of this remains unclear. Here, we used genome editing to knock-in a nuclear retention signal (NRS) in Srsf1 to create a mouse model harboring an SRSF1 protein that is retained exclusively in the nucleus. Srsf1NRS/NRS mutants displayed small body size, hydrocephalus, and immotile sperm, all traits associated with ciliary defects. We observed reduced translation of a subset of mRNAs and decreased abundance of proteins involved in multiciliogenesis, with disruption of ciliary ultrastructure and motility in cells and tissues derived from this mouse model. These results demonstrate that SRSF1 shuttling is used to reprogram gene expression networks in the context of high cellular demands, as observed here, during motile ciliogenesis.
Collapse
Affiliation(s)
- Fiona Haward
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Magdalena M Maslon
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Patricia L Yeyati
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Nicolas Bellora
- Institute of Nuclear Technologies for Health (Intecnus), National Scientific and Technical Research Council (CONICET)BarilocheArgentina
| | - Jan N Hansen
- Institute of Innate Immunity, Biophysical Imaging, Medical Faculty, University of BonnBonnGermany
| | - Stuart Aitken
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Jennifer Lawson
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Alex von Kriegsheim
- Edinburgh Cancer Research United Kingdom Centre, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Dagmar Wachten
- Institute of Innate Immunity, Biophysical Imaging, Medical Faculty, University of BonnBonnGermany
| | - Pleasantine Mill
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Ian R Adams
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Javier F Caceres
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| |
Collapse
|
21
|
Shui JE, Wang W, Liu H, Stepanova A, Liao G, Qian J, Ai X, Ten V, Lu J, Cardoso WV. Prematurity alters the progenitor cell program of the upper respiratory tract of neonates. Sci Rep 2021; 11:10799. [PMID: 34031475 PMCID: PMC8144386 DOI: 10.1038/s41598-021-90093-x] [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: 01/06/2021] [Accepted: 04/29/2021] [Indexed: 11/09/2022] Open
Abstract
The impact of prematurity on human development and neonatal diseases, such as bronchopulmonary dysplasia, has been widely reported. However, little is known about the effects of prematurity on the programs of stem cell self-renewal and differentiation of the upper respiratory epithelium, which is key for adaptation to neonatal life. We developed a minimally invasive methodology for isolation of neonatal basal cells from nasopharyngeal (NP) aspirates and performed functional analysis in organotypic cultures to address this issue. We show that preterm NP progenitors have a markedly distinct molecular signature of abnormal proliferation and mitochondria quality control compared to term progenitors. Preterm progenitors had lower oxygen consumption at baseline and were unable to ramp up consumption to the levels of term cells when challenged. Although they formed a mucociliary epithelium, ciliary function tended to decline in premature cells as they differentiated, compared to term cells. Together, these differences suggested increased sensitivity of preterm progenitors to environmental stressors under non-homeostatic conditions.
Collapse
Affiliation(s)
- Jessica E Shui
- Division of Neonatology and Newborn Medicine, Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Wei Wang
- Division of Neonatology and Newborn Medicine, Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Helu Liu
- Columbia Center for Human Development, Pulmonary Allergy & Critical Care Medicine, Department of Medicine, Columbia University Irving Medical Center, 650 West 168th Street, BB 8-812, New York, NY, 10032, USA
| | - Anna Stepanova
- Division of Neonatology, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Grace Liao
- Columbia Center for Human Development, Pulmonary Allergy & Critical Care Medicine, Department of Medicine, Columbia University Irving Medical Center, 650 West 168th Street, BB 8-812, New York, NY, 10032, USA
- Division of Neonatology, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Jun Qian
- Columbia Center for Human Development, Pulmonary Allergy & Critical Care Medicine, Department of Medicine, Columbia University Irving Medical Center, 650 West 168th Street, BB 8-812, New York, NY, 10032, USA
| | - Xingbin Ai
- Division of Neonatology and Newborn Medicine, Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Vadim Ten
- Division of Neonatology, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Jining Lu
- Division of Lung Diseases, NHLBI, NIH, Bethesda, MD, USA
| | - Wellington V Cardoso
- Columbia Center for Human Development, Pulmonary Allergy & Critical Care Medicine, Department of Medicine, Columbia University Irving Medical Center, 650 West 168th Street, BB 8-812, New York, NY, 10032, USA.
| |
Collapse
|
22
|
Haisma S, Weersma RK, Joosse ME, de Koning BAE, de Meij T, Koot BGP, Wolters V, Norbruis O, Daly MJ, Stevens C, Xavier RJ, Koskela J, Rivas MA, Visschedijk MC, Verkade HJ, Barbieri R, Jansen DBH, Festen EAM, van Rheenen PF, van Diemen CC. Exome sequencing in patient-parent trios suggests new candidate genes for early-onset primary sclerosing cholangitis. Liver Int 2021; 41:1044-1057. [PMID: 33590606 PMCID: PMC8252477 DOI: 10.1111/liv.14831] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 01/29/2021] [Accepted: 02/07/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND & AIMS Primary sclerosing cholangitis (PSC) is a rare bile duct disease strongly associated with inflammatory bowel disease (IBD). Whole-exome sequencing (WES) has contributed to understanding the molecular basis of very early-onset IBD, but rare protein-altering genetic variants have not been identified for early-onset PSC. We performed WES in patients diagnosed with PSC ≤ 12 years to investigate the contribution of rare genetic variants to early-onset PSC. METHODS In this multicentre study, WES was performed on 87 DNA samples from 29 patient-parent trios with early-onset PSC. We selected rare (minor allele frequency < 2%) coding and splice-site variants that matched recessive (homozygous and compound heterozygous variants) and dominant (de novo) inheritance in the index patients. Variant pathogenicity was predicted by an in-house developed algorithm (GAVIN), and PSC-relevant variants were selected using gene expression data and gene function. RESULTS In 22 of 29 trios we identified at least 1 possibly pathogenic variant. We prioritized 36 genes, harbouring a total of 54 variants with predicted pathogenic effects. In 18 genes, we identified 36 compound heterozygous variants, whereas in the other 18 genes we identified 18 de novo variants. Twelve of 36 candidate risk genes are known to play a role in transmembrane transport, adaptive and innate immunity, and epithelial barrier function. CONCLUSIONS The 36 candidate genes for early-onset PSC need further verification in other patient cohorts and evaluation of gene function before a causal role can be attributed to its variants.
Collapse
Affiliation(s)
- Sjoukje‐Marije Haisma
- Department of Paediatric Gastroenterology Hepatology and NutritionUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Rinse K. Weersma
- Department of Gastroenterology and HepatologyUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Maria E. Joosse
- Department of Paediatric GastroenterologyErasmus University Medical CenterSophia Children's HospitalRotterdamThe Netherlands
| | - Barbara A. E. de Koning
- Department of Paediatric GastroenterologyErasmus University Medical CenterSophia Children's HospitalRotterdamThe Netherlands
| | - Tim de Meij
- Department of Pediatric GastroenterologyVU University Medical CenterAmsterdamThe Netherlands
| | - Bart G. P. Koot
- Pediatric GastroenterologyEmma Children's HospitalAmsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Victorien Wolters
- Department of Pediatric GastroenterologyUniversity Medical Center Utrecht – Wilhelmina Children's HospitalUtrechtThe Netherlands
| | - Obbe Norbruis
- Department of PediatricsIsala HospitalZwolleThe Netherlands
| | - Mark J. Daly
- Broad Institute of Harvard and Massachusetts Institute of TechnologyBostonMAUSA
| | - Christine Stevens
- Broad Institute of Harvard and Massachusetts Institute of TechnologyBostonMAUSA
| | | | - Jukka Koskela
- Massachusetts General Hospital, GastroenterologyBostonMAUSA,Institute for Molecular Medicine Finland (FIMM)University of HelsinkiHelsinkiFinland,Clinic of Gastroenterology HelsinkiHelsinki University and Helsinki University HospitalHelsinkiFinland
| | | | - Marijn C. Visschedijk
- Department of Gastroenterology and HepatologyUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Henkjan J. Verkade
- Department of Paediatric Gastroenterology Hepatology and NutritionUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Ruggero Barbieri
- Department of Gastroenterology and HepatologyUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands,Department of GeneticsUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Dianne B. H. Jansen
- Department of Gastroenterology and HepatologyUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Eleonora A. M. Festen
- Department of Gastroenterology and HepatologyUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands,Department of GeneticsUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Patrick F. van Rheenen
- Department of Paediatric Gastroenterology Hepatology and NutritionUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Cleo C. van Diemen
- Department of GeneticsUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| |
Collapse
|
23
|
Sears PR, Bustamante-Marin XM, Gong H, Markovetz MR, Superfine R, Hill DB, Ostrowski LE. Induction of ciliary orientation by matrix patterning and characterization of mucociliary transport. Biophys J 2021; 120:1387-1395. [PMID: 33705757 PMCID: PMC8105732 DOI: 10.1016/j.bpj.2021.01.041] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/13/2021] [Accepted: 01/20/2021] [Indexed: 11/17/2022] Open
Abstract
Impaired mucociliary clearance (MCC) is a key feature of many airway diseases, including asthma, bronchiectasis, chronic obstructive pulmonary disease, cystic fibrosis, and primary ciliary dyskinesia. To improve MCC and develop new treatments for these diseases requires a thorough understanding of how mucus concentration, mucus composition, and ciliary activity affect MCC, and how different therapeutics impact this process. Although differentiated cultures of human airway epithelial cells are useful for investigations of MCC, the extent of ciliary coordination in these cultures varies, and the mechanisms controlling ciliary orientation are not completely understood. By introducing a pattern of ridges and grooves into the underlying collagen substrate, we demonstrate for the first time, to our knowledge, that changes in the extracellular matrix can induce ciliary alignment. Remarkably, 90% of human airway epithelial cultures achieved continuous directional mucociliary transport (MCT) when grown on the patterned substrate. These cultures maintain transport for months, allowing carefully controlled investigations of MCC over a wide range of normal and pathological conditions. To characterize the system, we measured the transport of bovine submaxillary gland mucin (BSM) under several conditions. Transport of 5% BSM was significantly reduced compared with that of 2% BSM, and treatment of 5% BSM with the reducing agent tris(2-carboxyethyl)phosphine (TCEP) reduced viscosity and increased the rate of MCT by approximately twofold. Addition of a small amount of high-molecular-weight DNA increased mucus viscosity and reduced MCT by ∼75%, demonstrating that the composition of mucus, as well as the concentration, can have significant effects on MCT. Our results demonstrate that a simple patterning of the collagen substrate results in highly coordinated ciliated cultures that develop directional MCT, and can be used to investigate the mechanisms controlling the regulation of ciliary orientation. Furthermore, the results demonstrate that this method provides an improved system for studying the effects of mucus composition and therapeutic agents on MCC.
Collapse
Affiliation(s)
- Patrick R Sears
- Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina
| | | | - Henry Gong
- Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina
| | - Matthew R Markovetz
- Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina
| | - Richard Superfine
- Department of Applied Physical Sciences, University of North Carolina, Chapel Hill, North Carolina
| | - David B Hill
- Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina; Department of Physics and Astronomy, University of North Carolina, Chapel Hill, North Carolina
| | - Lawrence E Ostrowski
- Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina; Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina.
| |
Collapse
|
24
|
Bonser LR, Koh KD, Johansson K, Choksi SP, Cheng D, Liu L, Sun DI, Zlock LT, Eckalbar WL, Finkbeiner WE, Erle DJ. Flow-Cytometric Analysis and Purification of Airway Epithelial-Cell Subsets. Am J Respir Cell Mol Biol 2021; 64:308-317. [PMID: 33196316 PMCID: PMC7909335 DOI: 10.1165/rcmb.2020-0149ma] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 11/16/2020] [Indexed: 12/16/2022] Open
Abstract
The human airway epithelium is essential in homeostasis, and epithelial dysfunction contributes to chronic airway disease. Development of flow-cytometric methods to characterize subsets of airway epithelial cells will enable further dissection of airway epithelial biology. Leveraging single-cell RNA-sequencing data in combination with known cell type-specific markers, we developed panels of antibodies to characterize and isolate the major airway epithelial subsets (basal, ciliated, and secretory cells) from human bronchial epithelial-cell cultures. We also identified molecularly distinct subpopulations of secretory cells and demonstrated cell subset-specific expression of low-abundance transcripts and microRNAs that are challenging to analyze with current single-cell RNA-sequencing methods. These new tools will be valuable for analyzing and separating airway epithelial subsets and interrogating airway epithelial biology.
Collapse
Affiliation(s)
| | - Kyung Duk Koh
- Lung Biology Center
- Cardiovascular Research Institute
| | - Kristina Johansson
- Department of Microbiology and Immunology
- Division of Pulmonary, Critical Care, Sleep, and Allergy
- Sandler Asthma Basic Research Center
| | | | - Dan Cheng
- Lung Biology Center
- Cardiovascular Research Institute
- Department of Respiratory and Critical Care Medicine, Renmin Hospital, Wuhan University, Wuhan, China
| | - Leqian Liu
- Department of Bioengineering and Therapeutic Sciences
| | | | | | - Walter L. Eckalbar
- Lung Biology Center
- University of California, San Francisco CoLabs, University of California San Francisco, San Francisco, California; and
| | | | - David J. Erle
- Lung Biology Center
- Cardiovascular Research Institute
- University of California, San Francisco CoLabs, University of California San Francisco, San Francisco, California; and
| |
Collapse
|
25
|
Legendre M, Zaragosi LE, Mitchison HM. Motile cilia and airway disease. Semin Cell Dev Biol 2020; 110:19-33. [PMID: 33279404 DOI: 10.1016/j.semcdb.2020.11.007] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/10/2020] [Accepted: 11/14/2020] [Indexed: 01/10/2023]
Abstract
A finely regulated system of airway epithelial development governs the differentiation of motile ciliated cells of the human respiratory tract, conferring the body's mucociliary clearance defence system. Human cilia dysfunction can arise through genetic mutations and this is a cause of debilitating disease morbidities that confer a greatly reduced quality of life. The inherited human motile ciliopathy disorder, primary ciliary dyskinesia (PCD), can arise from mutations in genes affecting various aspects of motile cilia structure and function through deficient production, transport and assembly of cilia motility components or through defective multiciliogenesis. Our understanding about the development of the respiratory epithelium, motile cilia biology and the implications for human pathology has expanded greatly over the past 20 years since isolation of the first PCD gene, rising to now nearly 50 genes. Systems level insights about cilia motility in health and disease have been made possible through intensive molecular and omics (genomics, transcriptomics, proteomics) research, applied in ciliate organisms and in animal and human disease modelling. Here, we review ciliated airway development and the genetic stratification that underlies PCD, for which the underlying genotype can increasingly be connected to biological mechanism and disease prognostics. Progress in this field can facilitate clinical translation of research advances, with potential for great medical impact, e.g. through improvements in ciliopathy disease diagnosis, management, family counselling and by enhancing the potential for future genetically tailored approaches to disease therapeutics.
Collapse
Affiliation(s)
- Marie Legendre
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Childhood Genetic Disorders, Département de Génétique Médicale, Hôpital Armand-Trousseau, Assistance Publique-Hôpitaux de Paris, Paris 75012, France
| | | | - Hannah M Mitchison
- Genetics and Genomic Medicine, University College London, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK; NIHR Biomedical Research Centre at Great Ormond Street Hospital, London, UK.
| |
Collapse
|
26
|
Liu H, Wei X, Sha Y, Liu W, Gao H, Lin J, Li Y, Tang Y, Wang Y, Wang Y, Su Z. Whole-exome sequencing in patients with premature ovarian insufficiency: early detection and early intervention. J Ovarian Res 2020; 13:114. [PMID: 32962729 PMCID: PMC7510158 DOI: 10.1186/s13048-020-00716-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 09/11/2020] [Indexed: 02/08/2023] Open
Abstract
Background The loss of ovarian function in women, referred to as premature ovarian insufficiency (POI), is associated with a series of concomitant diseases. POI is genetically heterogeneous, and in most cases, the etiology is unknown. Methods Whole-exome sequencing (WES) was performed on DNA samples obtained from patients with POI, and Sanger sequencing was used to validate the detected potentially pathogenic variants. An in silico analysis was carried out to predict the pathogenicity of the variants. Results We recruited 24 patients with POI and identified variants in POI-related genes in 14 patients, including bi-allelic mutations in DNAH6, HFM1, EIF2B2, BNC, and LRPPRC and heterozygous variants in BNC1, EIF2B4, FOXL2, MCM9, FANCA, ATM, EIF2B3, and GHR. No variants in the above genes were detected in the WES data obtained from 29 women in a control group without POI. Determining a clear genetic etiology could significantly increase patient compliance with appropriate intervention strategies. Conclusions Our study confirmed that POI is a genetically heterogeneous condition and that whole-exome sequencing is a powerful tool for determining its genetic etiology. The results of this study will aid researchers and clinicians in genetic counseling and suggests the potential of WES for the detection of POI and thus early interventions for patients with POI.
Collapse
Affiliation(s)
- Hongli Liu
- Department of Gynecology, Key Clinical Discipline of Fujian province, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, 361005, Fujian, China
| | - Xiaoli Wei
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361005, Fujian, China
| | - Yanwei Sha
- Department of Reproductive Medicine, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, 361005, Fujian, China
| | - Wensheng Liu
- Department of Gynecology and Obstetrics, Zhujiang Hospital, Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Haijie Gao
- Department of Reproductive Medicine, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, 361005, Fujian, China
| | - Jin Lin
- Department of Reproductive Medicine, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, 361005, Fujian, China
| | - Youzhu Li
- Reproductive Medicine Center, the First Affiliated Hospital of Xiamen University, Xiamen, 361003, Fujian, China
| | - Yaling Tang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Xiamen University, Xiamen, 361003, China
| | - Yifeng Wang
- Department of Gynecology and Obstetrics, Zhujiang Hospital, Southern Medical University, Guangzhou, 510000, Guangdong, China.
| | - Yanlong Wang
- Department of Gynecology, Key Clinical Discipline of Fujian province, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, 361005, Fujian, China.
| | - Zhiying Su
- Department of Reproductive Medicine, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, 361005, Fujian, China.
| |
Collapse
|
27
|
Nakamura R, Katsuno T, Kishimoto Y, Kaba S, Yoshimatsu M, Kitamura M, Suehiro A, Hiwatashi N, Yamashita M, Tateya I, Omori K. A novel method for live imaging of human airway cilia using wheat germ agglutinin. Sci Rep 2020; 10:14417. [PMID: 32879324 PMCID: PMC7468155 DOI: 10.1038/s41598-020-71049-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 08/06/2020] [Indexed: 11/17/2022] Open
Abstract
Multiciliated epithelial cells in the airway are essential for mucociliary clearance. Their function relies on coordinated, metachronal and directional ciliary beating, appropriate mucus secretion and airway surface hydration. However, current conventional methods for observing human airway ciliary movement require ciliated cells to be detached from airway tissues. Determining the directionality of cilia is difficult. We developed a novel method to stain airway epithelial cilia to observe their movement without releasing ciliated cells. Human tracheae were obtained from patients (n = 13) who underwent laryngectomies to treat malignancies or swallowing disorders. The tracheae were treated with fluorescently labeled wheat germ agglutinin, which interacts with the acidic mucopolysaccharides present on the cilia. Epithelial surfaces were observed using an epi-fluorescence microscope equipped with a water-immersion objective lens and a high-speed camera. Ciliary movement was observable at 125 fps (13/13 samples). Ciliated cells in close proximity mostly exhibited well-coordinated ciliary beats with similar directionalities. These findings indicated that wheat germ agglutinin renders ciliary beats visible, which is valuable for observing human airway ciliary movements in situ.
Collapse
Affiliation(s)
- Ryosuke Nakamura
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Tatsuya Katsuno
- Center of Anatomical, Pathological and Forensic Medical Researches, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yo Kishimoto
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Shinji Kaba
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Masayoshi Yoshimatsu
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Morimasa Kitamura
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Atsushi Suehiro
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Nao Hiwatashi
- Department of Otolaryngology, Kyoto-Katsura Hospital, Kyoto, Japan
| | - Masaru Yamashita
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Ichiro Tateya
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Fujita Health University, Toyoake, Japan
| | - Koichi Omori
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|
28
|
Bustamante-Marin XM, Horani A, Stoyanova M, Charng WL, Bottier M, Sears PR, Yin WN, Daniels LA, Bowen H, Conrad DF, Knowles MR, Ostrowski LE, Zariwala MA, Dutcher SK. Mutation of CFAP57, a protein required for the asymmetric targeting of a subset of inner dynein arms in Chlamydomonas, causes primary ciliary dyskinesia. PLoS Genet 2020; 16:e1008691. [PMID: 32764743 PMCID: PMC7444499 DOI: 10.1371/journal.pgen.1008691] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 08/19/2020] [Accepted: 02/22/2020] [Indexed: 01/10/2023] Open
Abstract
Primary ciliary dyskinesia (PCD) is characterized by chronic airway disease, reduced fertility, and randomization of the left/right body axis. It is caused by defects of motile cilia and sperm flagella. We screened a cohort of affected individuals that lack an obvious axonemal defect for pathogenic variants using whole exome capture, next generation sequencing, and bioinformatic analysis assuming an autosomal recessive trait. We identified one subject with an apparently homozygous nonsense variant [(c.1762C>T), p.(Arg588*)] in the uncharacterized CFAP57 gene. Interestingly, the variant results in the skipping of exon 11 (58 amino acids), which may be due to disruption of an exonic splicing enhancer. In normal human nasal epithelial cells, CFAP57 localizes throughout the ciliary axoneme. Nasal cells from the PCD patient express a shorter, mutant version of CFAP57 and the protein is not incorporated into the axoneme. The missing 58 amino acids include portions of WD repeats that may be important for loading onto the intraflagellar transport (IFT) complexes for transport or docking onto the axoneme. A reduced beat frequency and an alteration in ciliary waveform was observed. Knockdown of CFAP57 in human tracheobronchial epithelial cells (hTECs) recapitulates these findings. Phylogenetic analysis showed that CFAP57 is highly conserved in organisms that assemble motile cilia. CFAP57 is allelic with the BOP2/IDA8/FAP57 gene identified previously in Chlamydomonas reinhardtii. Two independent, insertional fap57 Chlamydomonas mutant strains show reduced swimming velocity and altered waveforms. Tandem mass tag (TMT) mass spectroscopy shows that FAP57 is missing, and the "g" inner dyneins (DHC7 and DHC3) and the "d" inner dynein (DHC2) are reduced, but the FAP57 paralog FBB7 is increased. Together, our data identify a homozygous variant in CFAP57 that causes PCD that is likely due to a defect in the inner dynein arm assembly process.
Collapse
Affiliation(s)
- Ximena M. Bustamante-Marin
- Department of Medicine, Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Amjad Horani
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Mihaela Stoyanova
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Wu-Lin Charng
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Mathieu Bottier
- Department of Mechanical Engineering, Washington University, St. Louis, Missouri, United States of America
| | - Patrick R. Sears
- Department of Medicine, Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Wei-Ning Yin
- Department of Medicine, Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Leigh Anne Daniels
- Department of Medicine, Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Hailey Bowen
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Donald F. Conrad
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Michael R. Knowles
- Department of Medicine, Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Lawrence E. Ostrowski
- Department of Medicine, Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Maimoona A. Zariwala
- Department of Pathology and Laboratory Medicine and the Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Susan K. Dutcher
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
29
|
Chivukula RR, Montoro DT, Leung HM, Yang J, Shamseldin HE, Taylor MS, Dougherty GW, Zariwala MA, Carson J, Daniels MLA, Sears PR, Black KE, Hariri LP, Almogarri I, Frenkel EM, Vinarsky V, Omran H, Knowles MR, Tearney GJ, Alkuraya FS, Sabatini DM. A human ciliopathy reveals essential functions for NEK10 in airway mucociliary clearance. Nat Med 2020; 26:244-251. [PMID: 31959991 PMCID: PMC7018620 DOI: 10.1038/s41591-019-0730-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 12/06/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Raghu R Chivukula
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA. .,Whitehead Institute for Biomedical Research, Cambridge, MA, USA. .,Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA. .,Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA. .,Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Daniel T Montoro
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Hui Min Leung
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Jason Yang
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA.,Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.,Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA.,Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Hanan E Shamseldin
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Martin S Taylor
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA.,Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.,Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA.,Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Gerard W Dougherty
- Department of General Pediatrics, University Children's Hospital Muenster, Münster, Germany
| | - Maimoona A Zariwala
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Johnny Carson
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - M Leigh Anne Daniels
- Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Patrick R Sears
- Cystic Fibrosis/Pulmonary Research and Treatment Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Katharine E Black
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Lida P Hariri
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Ibrahim Almogarri
- Department of Pediatrics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Evgeni M Frenkel
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA.,Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.,Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA.,Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Vladimir Vinarsky
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Heymut Omran
- Department of General Pediatrics, University Children's Hospital Muenster, Münster, Germany
| | - Michael R Knowles
- Cystic Fibrosis/Pulmonary Research and Treatment Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Guillermo J Tearney
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Department of Pathology, Massachusetts General Hospital, Boston, MA, USA.,Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
| | - Fowzan S Alkuraya
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.
| | - David M Sabatini
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA.,Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.,Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA.,Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
30
|
Chioccioli M, Feriani L, Kotar J, Bratcher PE, Cicuta P. Phenotyping ciliary dynamics and coordination in response to CFTR-modulators in Cystic Fibrosis respiratory epithelial cells. Nat Commun 2019; 10:1763. [PMID: 30992452 PMCID: PMC6467870 DOI: 10.1038/s41467-019-09798-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 03/29/2019] [Indexed: 12/22/2022] Open
Abstract
Personalized approaches for systematically assessing ciliary beat dynamics and for drug testing would improve the challenging task of diagnosing and treating respiratory disorders. In this pilot study, we show how multiscale differential dynamic microscopy (multi-DDM) can be used to characterize collective ciliary beating in a non-biased automated manner. We use multi-DDM to assess the efficacy of different CFTR-modulating drugs in human airway epithelial cells derived from subjects with cystic fibrosis (ΔF508/ΔF508 and ∆F508/-) based on ciliary beat frequency and coordination. Similar to clinical observations, drug efficacy is variable across donors, even within the same genotype. We show how our assay can quantitatively identify the most efficient drugs for restoring ciliary beating for each individual donor. Multi-DDM provides insight into ciliary beating responses following treatment with drugs, and has application in the broader context of respiratory disease and for drug screening.
Collapse
Affiliation(s)
- M Chioccioli
- Biological and Soft Systems Sector, Cavendish Laboratory, University of Cambridge, Cambridge, CB3 0HE, UK
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, 06510, USA
| | - L Feriani
- Biological and Soft Systems Sector, Cavendish Laboratory, University of Cambridge, Cambridge, CB3 0HE, UK
- Institute of Clinical Sciences, Imperial College London, London, SW7 2AZ, UK
- MRC London Institute of Medical Sciences, London, W12 0NN, UK
| | - J Kotar
- Biological and Soft Systems Sector, Cavendish Laboratory, University of Cambridge, Cambridge, CB3 0HE, UK
| | - P E Bratcher
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, CO, 80206, USA.
| | - P Cicuta
- Biological and Soft Systems Sector, Cavendish Laboratory, University of Cambridge, Cambridge, CB3 0HE, UK.
| |
Collapse
|
31
|
Bottier M, Thomas KA, Dutcher SK, Bayly PV. How Does Cilium Length Affect Beating? Biophys J 2019; 116:1292-1304. [PMID: 30878201 PMCID: PMC6451027 DOI: 10.1016/j.bpj.2019.02.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/23/2019] [Accepted: 02/13/2019] [Indexed: 12/21/2022] Open
Abstract
The effects of cilium length on the dynamics of cilia motion were investigated by high-speed video microscopy of uniciliated mutants of the swimming alga, Chlamydomonas reinhardtii. Cells with short cilia were obtained by deciliating cells via pH shock and allowing cilia to reassemble for limited times. The frequency of cilia beating was estimated from the motion of the cell body and of the cilium. Key features of the ciliary waveform were quantified from polynomial curves fitted to the cilium in each image frame. Most notably, periodic beating did not emerge until the cilium reached a critical length between 2 and 4 μm. Surprisingly, in cells that exhibited periodic beating, the frequency of beating was similar for all lengths with only a slight decrease in frequency as length increased from 4 μm to the normal length of 10-12 μm. The waveform average curvature (rad/μm) was also conserved as the cilium grew. The mechanical metrics of ciliary propulsion (force, torque, and power) all increased in proportion to length. The mechanical efficiency of beating appeared to be maximal at the normal wild-type length of 10-12 μm. These quantitative features of ciliary behavior illuminate the biophysics of cilia motion and, in future studies, may help distinguish competing hypotheses of the underlying mechanism of oscillation.
Collapse
Affiliation(s)
- Mathieu Bottier
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, Missouri; Department of Genetics, Washington University in St. Louis, St. Louis, Missouri
| | - Kyle A Thomas
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, Missouri
| | - Susan K Dutcher
- Department of Genetics, Washington University in St. Louis, St. Louis, Missouri
| | - Philip V Bayly
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, Missouri.
| |
Collapse
|
32
|
Fassad MR, Shoemark A, Legendre M, Hirst RA, Koll F, le Borgne P, Louis B, Daudvohra F, Patel MP, Thomas L, Dixon M, Burgoyne T, Hayes J, Nicholson AG, Cullup T, Jenkins L, Carr SB, Aurora P, Lemullois M, Aubusson-Fleury A, Papon JF, O’Callaghan C, Amselem S, Hogg C, Escudier E, Tassin AM, Mitchison HM. Mutations in Outer Dynein Arm Heavy Chain DNAH9 Cause Motile Cilia Defects and Situs Inversus. Am J Hum Genet 2018; 103:984-994. [PMID: 30471717 DOI: 10.1016/j.ajhg.2018.10.016] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 10/15/2018] [Indexed: 12/30/2022] Open
Abstract
Motile cilia move body fluids and gametes and the beating of cilia lining the airway epithelial surfaces ensures that they are kept clear and protected from inhaled pathogens and consequent respiratory infections. Dynein motor proteins provide mechanical force for cilia beating. Dynein mutations are a common cause of primary ciliary dyskinesia (PCD), an inherited condition characterized by deficient mucociliary clearance and chronic respiratory disease coupled with laterality disturbances and subfertility. Using next-generation sequencing, we detected mutations in the ciliary outer dynein arm (ODA) heavy chain gene DNAH9 in individuals from PCD clinics with situs inversus and in one case male infertility. DNAH9 and its partner heavy chain DNAH5 localize to type 2 ODAs of the distal cilium and in DNAH9-mutated nasal respiratory epithelial cilia we found a loss of DNAH9/DNAH5-containing type 2 ODAs that was restricted to the distal cilia region. This confers a reduced beating frequency with a subtle beating pattern defect affecting the motility of the distal cilia portion. 3D electron tomography ultrastructural studies confirmed regional loss of ODAs from the distal cilium, manifesting as either loss of whole ODA or partial loss of ODA volume. Paramecium DNAH9 knockdown confirms an evolutionarily conserved function for DNAH9 in cilia motility and ODA stability. We find that DNAH9 is widely expressed in the airways, despite DNAH9 mutations appearing to confer symptoms restricted to the upper respiratory tract. In summary, DNAH9 mutations reduce cilia function but some respiratory mucociliary clearance potential may be retained, widening the PCD disease spectrum.
Collapse
|
33
|
Loges NT, Antony D, Maver A, Deardorff MA, Güleç EY, Gezdirici A, Nöthe-Menchen T, Höben IM, Jelten L, Frank D, Werner C, Tebbe J, Wu K, Goldmuntz E, Čuturilo G, Krock B, Ritter A, Hjeij R, Bakey Z, Pennekamp P, Dworniczak B, Brunner H, Peterlin B, Tanidir C, Olbrich H, Omran H, Schmidts M. Recessive DNAH9 Loss-of-Function Mutations Cause Laterality Defects and Subtle Respiratory Ciliary-Beating Defects. Am J Hum Genet 2018; 103:995-1008. [PMID: 30471718 PMCID: PMC6288205 DOI: 10.1016/j.ajhg.2018.10.020] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 10/23/2018] [Indexed: 11/29/2022] Open
Abstract
Dysfunction of motile monocilia, altering the leftward flow at the embryonic node essential for determination of left-right body asymmetry, is a major cause of laterality defects. Laterality defects are also often associated with reduced mucociliary clearance caused by defective multiple motile cilia of the airway and are responsible for destructive airway disease. Outer dynein arms (ODAs) are essential for ciliary beat generation, and human respiratory cilia contain different ODA heavy chains (HCs): the panaxonemally distributed γ-HC DNAH5, proximally located β-HC DNAH11 (defining ODA type 1), and the distally localized β-HC DNAH9 (defining ODA type 2). Here we report loss-of-function mutations in DNAH9 in five independent families causing situs abnormalities associated with subtle respiratory ciliary dysfunction. Consistent with the observed subtle respiratory phenotype, high-speed video microscopy demonstrates distally impaired ciliary bending in DNAH9 mutant respiratory cilia. DNAH9-deficient cilia also lack other ODA components such as DNAH5, DNAI1, and DNAI2 from the distal axonemal compartment, demonstrating an essential role of DNAH9 for distal axonemal assembly of ODAs type 2. Yeast two-hybrid and co-immunoprecipitation analyses indicate interaction of DNAH9 with the ODA components DNAH5 and DNAI2 as well as the ODA-docking complex component CCDC114. We further show that during ciliogenesis of respiratory cilia, first proximally located DNAH11 and then distally located DNAH9 is assembled in the axoneme. We propose that the β-HC paralogs DNAH9 and DNAH11 achieved specific functional roles for the distinct axonemal compartments during evolution with human DNAH9 function matching that of ancient β-HCs such as that of the unicellular Chlamydomonas reinhardtii.
Collapse
|
34
|
Kim MD, Salathe M. Catch the Wave: Quantitatively Assessing Airway Ciliary Function as a Diagnostic Tool. Am J Respir Cell Mol Biol 2018; 59:415-416. [PMID: 30040472 DOI: 10.1165/rcmb.2018-0208ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Michael D Kim
- 1 Department of Internal Medicine University of Kansas Medical School Kansas City, Kansas
| | - Matthias Salathe
- 1 Department of Internal Medicine University of Kansas Medical School Kansas City, Kansas
| |
Collapse
|