1
|
Abbasi M, Zarei-Hanzaki A, Baghaei K, Abedi HR, Haghighipour N. Compression-induced apoptosis of fibroblasts and myofibroblasts in an in vitro model of pulmonary fibrosis by alginate/gelatin scaffold. Int J Biol Macromol 2024; 280:135875. [PMID: 39307498 DOI: 10.1016/j.ijbiomac.2024.135875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/03/2024]
Abstract
Pulmonary fibrosis leads to increased mortality but is poorly understood. Fibrotic progression is associated with abnormal wound repair and an increase in myofibroblast cell populations. Here we investigate how the myofibroblast population is impacted by unique compression-induced apoptosis derived from mechanical strain characteristic of asthma. Using a mechanical device, both static and dynamic mechanical strains were applied to alginate/gelatin/CaCl2 scaffolds containing fibroblasts and myofibroblasts. As cell groups were stimulated with 30 % static strain for 12 h, fibroblast and myofibroblast cell groups showed increased cell apoptosis by 5.55 % and 19.56 %, respectively, compared to control groups. Additionally, myofibroblasts exhibited higher susceptibility to apoptosis induction than did fibroblasts. Comparing dynamic and static loading modes, dynamic loading resulted in a higher apoptosis rate of fibroblast and myofibroblast cells, indicating its potential to induce apoptosis effectively. These findings suggest that mechanical stimulation can be considered a promising approach to induce apoptosis in myofibroblasts, thus offering the potential for future approaches to treating pulmonary fibrosis. Moreover, mechanical loads can be designed for other diseases, selectively reducing or increasing apoptosis in either hard or soft cell groups, based on specific application needs.
Collapse
Affiliation(s)
- Mahla Abbasi
- School of Metallurgy and Materials Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Abbas Zarei-Hanzaki
- School of Metallurgy and Materials Engineering, College of Engineering, University of Tehran, Tehran, Iran.
| | - Kaveh Baghaei
- Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Abedi
- School of Metallurgy & Materials Engineering, Iran University of Science and Technology (IUST), Tehran, Iran
| | | |
Collapse
|
2
|
Varricchi G, Brightling CE, Grainge C, Lambrecht BN, Chanez P. Airway remodelling in asthma and the epithelium: on the edge of a new era. Eur Respir J 2024; 63:2301619. [PMID: 38609094 PMCID: PMC11024394 DOI: 10.1183/13993003.01619-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 02/15/2024] [Indexed: 04/14/2024]
Abstract
Asthma is a chronic, heterogeneous disease of the airways, often characterised by structural changes known collectively as airway remodelling. In response to environmental insults, including pathogens, allergens and pollutants, the epithelium can initiate remodelling via an inflammatory cascade involving a variety of mediators that have downstream effects on both structural and immune cells. These mediators include the epithelial cytokines thymic stromal lymphopoietin, interleukin (IL)-33 and IL-25, which facilitate airway remodelling through cross-talk between epithelial cells and fibroblasts, and between mast cells and airway smooth muscle cells, as well as through signalling with immune cells such as macrophages. The epithelium can also initiate airway remodelling independently of inflammation in response to the mechanical stress present during bronchoconstriction. Furthermore, genetic and epigenetic alterations to epithelial components are believed to influence remodelling. Here, we review recent advances in our understanding of the roles of the epithelium and epithelial cytokines in driving airway remodelling, facilitated by developments in genetic sequencing and imaging techniques. We also explore how new and existing therapeutics that target the epithelium and epithelial cytokines could modify airway remodelling.
Collapse
Affiliation(s)
- Gilda Varricchi
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), School of Medicine, University of Naples Federico II, WAO Center of Excellence, Naples, Italy
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, Naples, Italy
- G. Varricchi and C.E. Brightling contributed equally
| | - Christopher E. Brightling
- Institute for Lung Health, NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
- G. Varricchi and C.E. Brightling contributed equally
| | - Christopher Grainge
- School of Medicine and Public Health, University of Newcastle, Callaghan, Australia
| | - Bart N. Lambrecht
- Center for Inflammation Research, Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Ghent, Belgium
| | - Pascal Chanez
- Department of Respiratory Diseases, Aix-Marseille University, Marseille, France
| |
Collapse
|
3
|
Park S, Newton J, Hidjir T, Young EWK. Bidirectional airflow in lung airway-on-a-chip with matrix-derived membrane elicits epithelial glycocalyx formation. LAB ON A CHIP 2023; 23:3671-3682. [PMID: 37462986 DOI: 10.1039/d3lc00259d] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/09/2023]
Abstract
Organ-on-a-chip systems are rapidly advancing as a viable alternative to existing experimental models in respiratory research. To date, however, epithelial cell cultures within lung airway-on-a-chip devices have yet to demonstrate the presence of an epithelial glycocalyx, a thin layer of proteoglycans, glycoproteins, and glycolipids known to play an important role in regulating epithelial function. Here, we demonstrate that an airway-on-a-chip device that incorporates bidirectional flow mimicking breathing cycles in combination with an ultra-thin matrix-derived membrane (UMM) layer can generate a glycocalyx layer comprised of heparan sulfate. Results with this device and airflow system showed dramatic differences of airway epithelial cell viability and expression of tight junctions, cilia, and mucus over a wide range of flow rates when cultured under oscillatory flow. More importantly, for the first time in a microfluidic organ-on-a-chip setting, we achieved the visualization of an airflow-induced epithelial glycocalyx layer. Our experiments highlight the importance of physiological mimicry in developing in vitro models, as bidirectional airflow showed more representative mucociliary differentiation compared to continuous unidirectional airflow. Thus, the lung airway-on-a-chip platform demonstrated in this study holds great potential as a lung epithelial barrier model for studying the mechanisms of various respiratory diseases and for testing the efficacy of therapeutic candidates in the presence of bidirectional airflow and the glycocalyx.
Collapse
Affiliation(s)
- Siwan Park
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada.
| | - Jeremy Newton
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON, M5S 3G8, Canada
| | - Tesnime Hidjir
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON, M5S 3G8, Canada
| | - Edmond W K Young
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, M5S 3G9, Canada.
- Department of Mechanical & Industrial Engineering, University of Toronto, Toronto, ON, M5S 3G8, Canada
| |
Collapse
|
4
|
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
|
5
|
Tang X, Zhou J, Koski TM, Liu S, Zhao L, Sun J. Hypoxia-induced tracheal elasticity in vector beetle facilitates the loading of pinewood nematode. eLife 2023; 12:84621. [PMID: 36995744 PMCID: PMC10063229 DOI: 10.7554/elife.84621] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 02/21/2023] [Indexed: 03/31/2023] Open
Abstract
Many pathogens rely on their insect vectors for transmission. Such pathogens are under selection to improve vector competence for their transmission by employing various tissue or cellular responses of vectors. However, whether pathogens can actively cause hypoxia in vectors and exploit hypoxia responses to promote their vector competence is still unknown. Fast dispersal of pinewood nematode (PWN), the causal agent for the destructive pine wilt disease and subsequent infection of pine trees, is characterized by the high vector competence of pine sawyer beetles (Monochamus spp.), and a single beetle can harbor over 200,000 PWNs in its tracheal system. Here, we demonstrate that PWN loading activates hypoxia in tracheal system of the vector beetles. Both PWN loading and hypoxia enhanced tracheal elasticity and thickened the apical extracellular matrix (aECM) of the tracheal tubes while a notable upregulated expression of a resilin-like mucin protein Muc91C was observed at the aECM layer of PWN-loaded and hypoxic tracheal tubes. RNAi knockdown of Muc91C reduced tracheal elasticity and aECM thickness under hypoxia conditions and thus decreasing PWN loading. Our study suggests a crucial role of hypoxia-induced developmental responses in shaping vector tolerance to the pathogen and provides clues for potential molecular targets to control pathogen dissemination.
Collapse
Affiliation(s)
- Xuan Tang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jiao Zhou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Tuuli-Marjaana Koski
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- College of Life Science/Hebei Basic Science Center for Biotic Interactions, Institute of Life Science and Green Development, Hebei University, Baoding, China
| | - Shiyao Liu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lilin Zhao
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jianghua Sun
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- College of Life Science/Hebei Basic Science Center for Biotic Interactions, Institute of Life Science and Green Development, Hebei University, Baoding, China
| |
Collapse
|
6
|
Baguma-Nibasheka M, Kablar B. Mechanics of Lung Development. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2023; 236:131-150. [PMID: 37955774 DOI: 10.1007/978-3-031-38215-4_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
We summarize how skeletal muscle and lung developmental biology fields have been bridged to benefit from mouse genetic engineering technologies and to explore the role of fetal breathing-like movements (FBMs) in lung development, by using skeletal muscle-specific mutant mice. It has been known for a long time that FBMs are essential for the lung to develop properly. However, the cellular and molecular mechanisms transducing the mechanical forces of muscular activity into specific genetic programs that propel lung morphogenesis (development of the shape, form and size of the lung, its airways, and gas exchange surface) as well as its differentiation (acquisition of specialized cell structural and functional features from their progenitor cells) are only starting to be revealed. This chapter is a brief synopsis of the cumulative findings from that ongoing quest. An update on and the rationale for our recent International Mouse Phenotyping Consortium (IMPC) search is also provided.
Collapse
Affiliation(s)
- Mark Baguma-Nibasheka
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada.
| | - Boris Kablar
- Department of Medical Neuroscience, Anatomy and Pathology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
7
|
Gautam LK, Harriott NC, Caceres AM, Ryan AL. Basic Science Perspective on Engineering and Modeling the Large Airways. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1413:73-106. [PMID: 37195527 DOI: 10.1007/978-3-031-26625-6_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
The airway epithelium provides a physical and biochemical barrier playing a key role in protecting the lung from infiltration of pathogens and irritants and is, therefore, crucial in maintaining tissue homeostasis and regulating innate immunity. Due to continual inspiration and expiration of air during breathing, the epithelium is exposed to a plethora of environmental insults. When severe or persistent, these insults lead to inflammation and infection. The effectiveness of the epithelium as a barrier is reliant upon its capacity for mucociliary clearance, immune surveillance, and regeneration upon injury. These functions are accomplished by the cells that comprise the airway epithelium and the niche in which they reside. Engineering of new physiological and pathological models of the proximal airways requires the generation of complex structures comprising the surface airway epithelium, submucosal gland epithelium, extracellular matrix, and niche cells, including smooth muscle cells, fibroblasts, and immune cells. This chapter focuses on the structure-function relationships in the airways and the challenges of developing complex engineered models of the human airway.
Collapse
Affiliation(s)
- Lalit K Gautam
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Noa C Harriott
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Adrian M Caceres
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Amy L Ryan
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
8
|
De Leon N, Tse WH, Ameis D, Keijzer R. Embryology and anatomy of congenital diaphragmatic hernia. Semin Pediatr Surg 2022; 31:151229. [PMID: 36446305 DOI: 10.1016/j.sempedsurg.2022.151229] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Prenatal and postnatal treatment modalities for congenital diaphragmatic hernia (CDH) continue to improve, however patients still face high rates of morbidity and mortality caused by severe underlying persistent pulmonary hypertension and pulmonary hypoplasia. Though the majority of CDH cases are idiopathic, it is believed that CDH is a polygenic developmental defect caused by interactions between candidate genes, as well as environmental and epigenetic factors. However, the origin and pathogenesis of these developmental insults are poorly understood. Further, connections between disrupted lung development and the failure of diaphragmatic closure during embryogenesis have not been fully elucidated. Though several animal models have been useful in identifying candidate genes and disrupted signalling pathways, more studies are required to understand the pathogenesis and to develop effective preventative care. In this article, we summarize the most recent litterature on disrupted embryological lung and diaphragmatic development associated with CDH.
Collapse
Affiliation(s)
- Nolan De Leon
- Departments of Surgery, Division of Pediatric Surgery, Pediatrics & Child Health and Physiology and Pathophysiology, University of Manitoba and Biology of Breathing Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Wai Hei Tse
- Departments of Surgery, Division of Pediatric Surgery, Pediatrics & Child Health and Physiology and Pathophysiology, University of Manitoba and Biology of Breathing Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Dustin Ameis
- Departments of Surgery, Division of Pediatric Surgery, Pediatrics & Child Health and Physiology and Pathophysiology, University of Manitoba and Biology of Breathing Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Richard Keijzer
- Departments of Surgery, Division of Pediatric Surgery, Pediatrics & Child Health and Physiology and Pathophysiology, University of Manitoba and Biology of Breathing Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
9
|
Lacombe J, Zenhausern F. Effect of mechanical forces on cellular response to radiation. Radiother Oncol 2022; 176:187-198. [PMID: 36228760 DOI: 10.1016/j.radonc.2022.10.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/08/2022] [Accepted: 10/05/2022] [Indexed: 12/14/2022]
Abstract
While the cellular interactions and biochemical signaling has been investigated for long and showed to play a major role in the cell's fate, it is now also evident that mechanical forces continuously applied to the cells in their microenvironment are as important for tissue homeostasis. Mechanical cues are emerging as key regulators of cellular drug response and we aimed to demonstrate in this review that such effects should also be considered vital for the cellular response to radiation. In order to explore the mechanobiology of the radiation response, we reviewed the main mechanoreceptors and transducers, including integrin-mediated adhesion, YAP/TAZ pathways, Wnt/β-catenin signaling, ion channels and G protein-coupled receptors and showed their implication in the modulation of cellular radiosensitivity. We then discussed the current studies that investigated a direct effect of mechanical stress, including extracellular matrix stiffness, shear stress and mechanical strain, on radiation response of cancer and normal cells and showed through preliminary results that such stress effectively can alter cell response after irradiation. However, we also highlighted the limitations of these studies and emphasized some of the contradictory data, demonstrating that the effect of mechanical cues could involve complex interactions and potential crosstalk with numerous cellular processes also affected by irradiation. Overall, mechanical forces alter radiation response and although additional studies are required to deeply understand the underlying mechanisms, these effects should not be neglected in radiation research as they could reveal new fundamental knowledge for predicting radiosensitivity or understanding resistance to radiotherapy.
Collapse
Affiliation(s)
- Jerome Lacombe
- Center for Applied NanoBioscience and Medicine, College of Medicine Phoenix, University of Arizona, 475 North 5th Street, Phoenix, AZ 85004, USA; Department of Basic Medical Sciences, College of Medicine Phoenix, University of Arizona, 425 N 5th St, Phoenix, AZ 85004, USA.
| | - Frederic Zenhausern
- Center for Applied NanoBioscience and Medicine, College of Medicine Phoenix, University of Arizona, 475 North 5th Street, Phoenix, AZ 85004, USA; Department of Basic Medical Sciences, College of Medicine Phoenix, University of Arizona, 425 N 5th St, Phoenix, AZ 85004, USA; Department of Biomedical Engineering, College of Engineering, University of Arizona, 1127 E. James E. Rogers Way, Tucson, AZ 85721, USA.
| |
Collapse
|
10
|
Naumann J, Koppe N, Thome UH, Laube M, Zink M. Mechanical properties of the premature lung: From tissue deformation under load to mechanosensitivity of alveolar cells. Front Bioeng Biotechnol 2022; 10:964318. [PMID: 36185437 PMCID: PMC9523442 DOI: 10.3389/fbioe.2022.964318] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
Many preterm infants require mechanical ventilation as life-saving therapy. However, ventilation-induced overpressure can result in lung diseases. Considering the lung as a viscoelastic material, positive pressure inside the lung results in increased hydrostatic pressure and tissue compression. To elucidate the effect of positive pressure on lung tissue mechanics and cell behavior, we mimic the effect of overpressure by employing an uniaxial load onto fetal and adult rat lungs with different deformation rates. Additionally, tissue expansion during tidal breathing due to a negative intrathoracic pressure was addressed by uniaxial tension. We found a hyperelastic deformation behavior of fetal tissues under compression and tension with a remarkable strain stiffening. In contrast, adult lungs exhibited a similar response only during compression. Young’s moduli were always larger during tension compared to compression, while only during compression a strong deformation-rate dependency was found. In fact, fetal lung tissue under compression showed clear viscoelastic features even for small strains. Thus, we propose that the fetal lung is much more vulnerable during inflation by mechanical ventilation compared to normal inspiration. Electrophysiological experiments with different hydrostatic pressure gradients acting on primary fetal distal lung epithelial cells revealed that the activity of the epithelial sodium channel (ENaC) and the sodium-potassium pump (Na,K-ATPase) dropped during pressures of 30 cmH2O. Thus, pressures used during mechanical ventilation might impair alveolar fluid clearance important for normal lung function.
Collapse
Affiliation(s)
- Jonas Naumann
- Research Group Biotechnology and Biomedicine, Peter-Debye-Institute for Soft Matter Physics, Leipzig University, Leipzig, Germany
| | - Nicklas Koppe
- Research Group Biotechnology and Biomedicine, Peter-Debye-Institute for Soft Matter Physics, Leipzig University, Leipzig, Germany
| | - Ulrich H. Thome
- Center for Pediatric Research Leipzig, Department of Pediatrics, Division of Neonatology, Leipzig University, Leipzig, Germany
| | - Mandy Laube
- Center for Pediatric Research Leipzig, Department of Pediatrics, Division of Neonatology, Leipzig University, Leipzig, Germany
| | - Mareike Zink
- Research Group Biotechnology and Biomedicine, Peter-Debye-Institute for Soft Matter Physics, Leipzig University, Leipzig, Germany
- *Correspondence: Mareike Zink,
| |
Collapse
|
11
|
Ferreira JMC, Huhle R, Müller S, Schnabel C, Mehner M, Koch T, Gama de Abreu M. Static Stretch Increases the Pro-Inflammatory Response of Rat Type 2 Alveolar Epithelial Cells to Dynamic Stretch. Front Physiol 2022; 13:838834. [PMID: 35480037 PMCID: PMC9035495 DOI: 10.3389/fphys.2022.838834] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 03/28/2022] [Indexed: 11/21/2022] Open
Abstract
Background: Mechanical ventilation (MV) inflicts stress on the lungs, initiating or increasing lung inflammation, so-called ventilator-induced lung injury (VILI). Besides overdistention, cyclic opening-and-closing of alveoli (atelectrauma) is recognized as a potential mechanism of VILI. The dynamic stretch may be reduced by positive end-expiratory pressure (PEEP), which in turn increases the static stretch. We investigated whether static stretch modulates the inflammatory response of rat type 2 alveolar epithelial cells (AECs) at different levels of dynamic stretch and hypothesized that static stretch increases pro-inflammatory response of AECs at given dynamic stretch. Methods: AECs, stimulated and not stimulated with lipopolysaccharide (LPS), were subjected to combinations of static (10, 20, and 30%) and dynamic stretch (15, 20, and 30%), for 1 and 4 h. Non-stretched AECs served as control. The gene expression and secreted protein levels of interleukin-6 (IL-6), monocyte chemoattractant protein-1 (MCP-1), and macrophage inflammatory protein 2 (MIP-2) were studied by real-time polymerase chain reaction (RT-qPCR) and enzyme-linked immunosorbent assay (ELISA), respectively. The effects of static and dynamic stretch were assessed by two-factorial ANOVA with planned effects post-hoc comparison according to Šidák. Statistical significance was considered for p < 0.05. Results: In LPS-stimulated, but not in non-stimulated rat type 2 AECs, compared to non-stretched cells: 1) dynamic stretch increased the expression of amphiregulin (AREG) (p < 0.05), MCP-1 (p < 0.001), and MIP-2 (<0.05), respectively, as well as the protein secretion of IL-6 (p < 0.001) and MCP-1 (p < 0.05); 2) static stretch increased the gene expression of MCP-1 (p < 0.001) and MIP-2, but not AREG, and resulted in higher secretion of IL-6 (p < 0.001), but not MCP-1, while MIP-2 was not detectable in the medium. Conclusion: In rat type 2 AECs stimulated with LPS, static stretch increased the pro-inflammatory response to dynamic stretch, suggesting a potential pro-inflammatory effect of PEEP during mechanical ventilation at the cellular level.
Collapse
Affiliation(s)
- Jorge M. C. Ferreira
- Department of Anesthesiology and Intensive Care Medicine, Pulmonary Engineering Group, University Hospital Carl Gustav Carus Dresden at Technische Universität Dresden, Dresden, Germany
- *Correspondence: Jorge M. C. Ferreira,
| | - Robert Huhle
- Department of Anesthesiology and Intensive Care Medicine, Pulmonary Engineering Group, University Hospital Carl Gustav Carus Dresden at Technische Universität Dresden, Dresden, Germany
| | - Sabine Müller
- Department of Anesthesiology and Intensive Care Medicine, Pulmonary Engineering Group, University Hospital Carl Gustav Carus Dresden at Technische Universität Dresden, Dresden, Germany
| | - Christian Schnabel
- Department of Anesthesiology and Intensive Care Medicine, Clinical Sensoring and Monitoring Group, University Hospital Carl Gustav Carus Dresden at Technische Universität Dresden, Dresden, Germany
| | - Mirko Mehner
- Department of Anesthesiology and Intensive Care Medicine, Clinical Sensoring and Monitoring Group, University Hospital Carl Gustav Carus Dresden at Technische Universität Dresden, Dresden, Germany
| | - Thea Koch
- Department of Anesthesiology and Intensive Care Medicine, Pulmonary Engineering Group, University Hospital Carl Gustav Carus Dresden at Technische Universität Dresden, Dresden, Germany
| | - Marcelo Gama de Abreu
- Department of Anesthesiology and Intensive Care Medicine, Pulmonary Engineering Group, University Hospital Carl Gustav Carus Dresden at Technische Universität Dresden, Dresden, Germany
- Department of Intensive Care and Resuscitation, Anesthesiology Institute, Cleveland Clinic, Cleveland, OH, United States
- Department of Outcomes Research, Anesthesiology Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
12
|
Boraldi F, Lofaro FD, Cossarizza A, Quaglino D. The "Elastic Perspective" of SARS-CoV-2 Infection and the Role of Intrinsic and Extrinsic Factors. Int J Mol Sci 2022; 23:ijms23031559. [PMID: 35163482 PMCID: PMC8835950 DOI: 10.3390/ijms23031559] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/20/2022] [Accepted: 01/28/2022] [Indexed: 02/07/2023] Open
Abstract
Elastin represents the structural component of the extracellular matrix providing elastic recoil to tissues such as skin, blood vessels and lungs. Elastogenic cells secrete soluble tropoelastin monomers into the extracellular space where these monomers associate with other matrix proteins (e.g., microfibrils and glycoproteins) and are crosslinked by lysyl oxidase to form insoluble fibres. Once elastic fibres are formed, they are very stable, highly resistant to degradation and have an almost negligible turnover. However, there are circumstances, mainly related to inflammatory conditions, where increased proteolytic degradation of elastic fibres may lead to consequences of major clinical relevance. In severely affected COVID-19 patients, for instance, the massive recruitment and activation of neutrophils is responsible for the profuse release of elastases and other proteolytic enzymes which cause the irreversible degradation of elastic fibres. Within the lungs, destruction of the elastic network may lead to the permanent impairment of pulmonary function, thus suggesting that elastases can be a promising target to preserve the elastic component in COVID-19 patients. Moreover, intrinsic and extrinsic factors additionally contributing to damaging the elastic component and to increasing the spread and severity of SARS-CoV-2 infection are reviewed.
Collapse
Affiliation(s)
- Federica Boraldi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (F.B.); (F.D.L.)
| | - Francesco Demetrio Lofaro
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (F.B.); (F.D.L.)
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, 41125 Modena, Italy;
| | - Daniela Quaglino
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (F.B.); (F.D.L.)
- Correspondence:
| |
Collapse
|
13
|
Lai Y, Huang Y. Mechanisms of Mechanical Force Induced Pulmonary Vascular Endothelial Hyperpermeability. Front Physiol 2021; 12:714064. [PMID: 34671268 PMCID: PMC8521004 DOI: 10.3389/fphys.2021.714064] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 08/30/2021] [Indexed: 12/27/2022] Open
Abstract
Mechanical ventilation is a supportive therapy for patients with acute respiratory distress syndrome (ARDS). However, it also inevitably produces or aggravates the original lung injury with pathophysiological changes of pulmonary edema caused by increased permeability of alveolar capillaries which composed of microvascular endothelium, alveolar epithelium, and basement membrane. Vascular endothelium forms a semi-selective barrier to regulate body fluid balance. Mechanical ventilation in critically ill patients produces a mechanical force on lung vascular endothelium when the endothelial barrier was destructed. This review aims to provide a comprehensive overview of molecular and signaling mechanisms underlying the endothelial barrier permeability in ventilator-induced lung jury (VILI).
Collapse
Affiliation(s)
- Yan Lai
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Critical Care Medicine, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yongbo Huang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Critical Care Medicine, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
14
|
Poole K. The Diverse Physiological Functions of Mechanically Activated Ion Channels in Mammals. Annu Rev Physiol 2021; 84:307-329. [PMID: 34637325 DOI: 10.1146/annurev-physiol-060721-100935] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Many aspects of mammalian physiology are mechanically regulated. One set of molecules that can mediate mechanotransduction are the mechanically activated ion channels. These ionotropic force sensors are directly activated by mechanical inputs, resulting in ionic flux across the plasma membrane. While there has been much research focus on the role of mechanically activated ion channels in touch sensation and hearing, recent data have highlighted the broad expression pattern of these molecules in mammalian cells. Disruption of mechanically activated channels has been shown to impact (a) the development of mechanoresponsive structures, (b) acute mechanical sensing, and (c) mechanically driven homeostatic maintenance in multiple tissue types. The diversity of processes impacted by these molecules highlights the importance of mechanically activated ion channels in mammalian physiology. Expected final online publication date for the Annual Review of Physiology, Volume 84 is February 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Kate Poole
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia; .,Cellular and Systems Physiology, School of Medical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| |
Collapse
|
15
|
Vasse GF, Nizamoglu M, Heijink IH, Schlepütz M, van Rijn P, Thomas MJ, Burgess JK, Melgert BN. Macrophage-stroma interactions in fibrosis: biochemical, biophysical, and cellular perspectives. J Pathol 2021; 254:344-357. [PMID: 33506963 PMCID: PMC8252758 DOI: 10.1002/path.5632] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/18/2020] [Accepted: 01/08/2021] [Indexed: 12/16/2022]
Abstract
Fibrosis results from aberrant wound healing and is characterized by an accumulation of extracellular matrix, impairing the function of an affected organ. Increased deposition of extracellular matrix proteins, disruption of matrix degradation, but also abnormal post-translational modifications alter the biochemical composition and biophysical properties of the tissue microenvironment - the stroma. Macrophages are known to play an important role in wound healing and tissue repair, but the direct influence of fibrotic stroma on macrophage behaviour is still an under-investigated element in the pathogenesis of fibrosis. In this review, the current knowledge on interactions between macrophages and (fibrotic) stroma will be discussed from biochemical, biophysical, and cellular perspectives. Furthermore, we provide future perspectives with regard to how macrophage-stroma interactions can be examined further to ultimately facilitate more specific targeting of these interactions in the treatment of fibrosis. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Gwenda F Vasse
- University of Groningen, University Medical Center GroningenBiomedical Engineering Department‐FB40GroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials ScienceGroningenThe Netherlands
- University of Groningen, Department of Molecular PharmacologyGroningen Research Institute for PharmacyGroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
| | - Mehmet Nizamoglu
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
- University of Groningen, University Medical Center GroningenDepartment of Pathology and Medical BiologyGroningenThe Netherlands
| | - Irene H Heijink
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
- University of Groningen, University Medical Center GroningenDepartment of Pathology and Medical BiologyGroningenThe Netherlands
- University of Groningen, University Medical Center GroningenDepartment of PulmonologyGroningenThe Netherlands
| | - Marco Schlepütz
- Immunology & Respiratory Diseases ResearchBoehringer Ingelheim Pharma GmbH & Co KGBiberach an der RissGermany
| | - Patrick van Rijn
- University of Groningen, University Medical Center GroningenBiomedical Engineering Department‐FB40GroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials ScienceGroningenThe Netherlands
| | - Matthew J Thomas
- Immunology & Respiratory Diseases ResearchBoehringer Ingelheim Pharma GmbH & Co KGBiberach an der RissGermany
| | - Janette K Burgess
- University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials ScienceGroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
- University of Groningen, University Medical Center GroningenDepartment of Pathology and Medical BiologyGroningenThe Netherlands
| | - Barbro N Melgert
- University of Groningen, Department of Molecular PharmacologyGroningen Research Institute for PharmacyGroningenThe Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC)GroningenThe Netherlands
| |
Collapse
|
16
|
Nossa R, Costa J, Cacopardo L, Ahluwalia A. Breathing in vitro: Designs and applications of engineered lung models. J Tissue Eng 2021; 12:20417314211008696. [PMID: 33996022 PMCID: PMC8107677 DOI: 10.1177/20417314211008696] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 03/22/2021] [Indexed: 12/11/2022] Open
Abstract
The aim of this review is to provide a systematic design guideline to users, particularly engineers interested in developing and deploying lung models, and biologists seeking to identify a suitable platform for conducting in vitro experiments involving pulmonary cells or tissues. We first discuss the state of the art on lung in vitro models, describing the most simplistic and traditional ones. Then, we analyze in further detail the more complex dynamic engineered systems that either provide mechanical cues, or allow for more predictive exposure studies, or in some cases even both. This is followed by a dedicated section on microchips of the lung. Lastly, we present a critical discussion of the different characteristics of each type of system and the criteria which may help researchers select the most appropriate technology according to their specific requirements. Readers are encouraged to refer to the tables accompanying the different sections where comprehensive and quantitative information on the operating parameters and performance of the different systems reported in the literature is provided.
Collapse
|
17
|
Yang J, Zhang Y, Qin M, Cheng W, Wang W, Cao Y. Understanding and Regulating Cell-Matrix Interactions Using Hydrogels of Designable Mechanical Properties. J Biomed Nanotechnol 2021; 17:149-168. [PMID: 33785089 DOI: 10.1166/jbn.2021.3026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Similar to natural tissues, hydrogels contain abundant water, so they are considered as promising biomaterials for studying the influence of the mechanical properties of extracellular matrices (ECM) on various cell functions. In recent years, the growing research on cellular mechanical response has revealed that many cell functions, including cell spreading, migration, tumorigenesis and differentiation, are related to the mechanical properties of ECM. Therefore, how cells sense and respond to the extracellular mechanical environment has gained considerable attention. In these studies, hydrogels are widely used as the in vitro model system. Hydrogels of tunable stiffness, viscoelasticity, degradability, plasticity, and dynamical properties have been engineered to reveal how cells respond to specific mechanical features. In this review, we summarize recent process in this research direction and specifically focus on the influence of the mechanical properties of the ECM on cell functions, how cells sense and respond to the extracellular mechanical environment, and approaches to adjusting the stiffness of hydrogels.
Collapse
Affiliation(s)
- Jiapeng Yang
- Key Laboratory of Intelligent Optical Sensing and Integration, National Laboratory of Solid State Microstructure, and Department of Physics, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, China
| | - Yu Zhang
- Key Laboratory of Intelligent Optical Sensing and Integration, National Laboratory of Solid State Microstructure, and Department of Physics, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, China
| | - Meng Qin
- Key Laboratory of Intelligent Optical Sensing and Integration, National Laboratory of Solid State Microstructure, and Department of Physics, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, China
| | - Wei Cheng
- Department of Oral Implantology Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing 210008, China
| | - Wei Wang
- Key Laboratory of Intelligent Optical Sensing and Integration, National Laboratory of Solid State Microstructure, and Department of Physics, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, China
| | - Yi Cao
- Key Laboratory of Intelligent Optical Sensing and Integration, National Laboratory of Solid State Microstructure, and Department of Physics, Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing 210093, China
| |
Collapse
|
18
|
Veerati PC, Mitchel JA, Reid AT, Knight DA, Bartlett NW, Park JA, Grainge CL. Airway mechanical compression: its role in asthma pathogenesis and progression. Eur Respir Rev 2020; 29:190123. [PMID: 32759373 PMCID: PMC8008491 DOI: 10.1183/16000617.0123-2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 01/30/2020] [Indexed: 12/22/2022] Open
Abstract
The lung is a mechanically active organ, but uncontrolled or excessive mechanical forces disrupt normal lung function and can contribute to the development of disease. In asthma, bronchoconstriction leads to airway narrowing and airway wall buckling. A growing body of evidence suggests that pathological mechanical forces induced by airway buckling alone can perpetuate disease processes in asthma. Here, we review the data obtained from a variety of experimental models, including in vitro, ex vivo and in vivo approaches, which have been used to study the impact of mechanical forces in asthma pathogenesis. We review the evidence showing that mechanical compression alters the biological and biophysical properties of the airway epithelium, including activation of the epidermal growth factor receptor pathway, overproduction of asthma-associated mediators, goblet cell hyperplasia, and a phase transition of epithelium from a static jammed phase to a mobile unjammed phase. We also define questions regarding the impact of mechanical forces on the pathology of asthma, with a focus on known triggers of asthma exacerbations such as viral infection.
Collapse
Affiliation(s)
- Punnam Chander Veerati
- School of Medicine and Public Health, University of Newcastle, Callaghan, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, Australia
| | - Jennifer A Mitchel
- Molecular and Integrative Physiological Sciences Program, Dept of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Andrew T Reid
- School of Medicine and Public Health, University of Newcastle, Callaghan, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, Australia
| | - Darryl A Knight
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, Australia
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, Australia
- Dept of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, Canada
- Research and Academic Affairs, Providence Health Care Research Institute, Vancouver, Canada
| | - Nathan W Bartlett
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, Australia
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, Australia
| | - Jin-Ah Park
- Molecular and Integrative Physiological Sciences Program, Dept of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Chris L Grainge
- School of Medicine and Public Health, University of Newcastle, Callaghan, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, University of Newcastle, New Lambton Heights, Australia
- Dept of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, Australia
| |
Collapse
|
19
|
|
20
|
Wirsching E, Fauler M, Fois G, Frick M. P2 Purinergic Signaling in the Distal Lung in Health and Disease. Int J Mol Sci 2020; 21:E4973. [PMID: 32674494 PMCID: PMC7404078 DOI: 10.3390/ijms21144973] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/07/2020] [Accepted: 07/10/2020] [Indexed: 12/13/2022] Open
Abstract
The distal lung provides an intricate structure for gas exchange in mammalian lungs. Efficient gas exchange depends on the functional integrity of lung alveoli. The cells in the alveolar tissue serve various functions to maintain alveolar structure, integrity and homeostasis. Alveolar epithelial cells secrete pulmonary surfactant, regulate the alveolar surface liquid (ASL) volume and, together with resident and infiltrating immune cells, provide a powerful host-defense system against a multitude of particles, microbes and toxicants. It is well established that all of these cells express purinergic P2 receptors and that purinergic signaling plays important roles in maintaining alveolar homeostasis. Therefore, it is not surprising that purinergic signaling also contributes to development and progression of severe pathological conditions like pulmonary inflammation, acute lung injury/acute respiratory distress syndrome (ALI/ARDS) and pulmonary fibrosis. Within this review we focus on the role of P2 purinergic signaling in the distal lung in health and disease. We recapitulate the expression of P2 receptors within the cells in the alveoli, the possible sources of ATP (adenosine triphosphate) within alveoli and the contribution of purinergic signaling to regulation of surfactant secretion, ASL volume and composition, as well as immune homeostasis. Finally, we summarize current knowledge of the role for P2 signaling in infectious pneumonia, ALI/ARDS and idiopathic pulmonary fibrosis (IPF).
Collapse
Affiliation(s)
| | | | | | - Manfred Frick
- Institute of General Physiology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany; (E.W.); (M.F.); (G.F.)
| |
Collapse
|
21
|
Quantitative Phosphoproteomics Reveals Cell Alignment and Mitochondrial Length Change under Cyclic Stretching in Lung Cells. Int J Mol Sci 2020; 21:ijms21114074. [PMID: 32517296 PMCID: PMC7312583 DOI: 10.3390/ijms21114074] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 12/14/2022] Open
Abstract
Lung cancer is a leading cause of death. Most previous studies have been based on traditional cell-culturing methods. However, lung cells are periodically subjected to mechanical forces during breathing. Understanding the mechanisms underlying the cyclic stretching induced in lung cells may be important for lung cancer therapy. Here, we applied cyclic stretching to stimulate the continual contraction that is present under physiological conditions in lung cells. We first uncovered the stretching-induced phosphoproteome in lung cancer cell line A549 and fibroblast cell line IMR-90. We identified 2048 and 2604 phosphosites corresponding to 837 and 1008 phosphoproteins in A549 and IMR-90, respectively. Furthermore, we combined our phosphoproteomics and public gene expression data to identify the biological functions in response to cyclic stretching. Interestingly, cytoskeletal and mitochondrial reorganization were enriched. We further used cell imaging analysis to validate the profiling results and found that this physical force changed cell alignment and mitochondrial length. This study not only reveals the molecular mechanism of cyclic stretching but also provides evidence that cell stretching causes cellular rearrangement and mitochondrial length change.
Collapse
|
22
|
Domingo-Gonzalez R, Zanini F, Che X, Liu M, Jones RC, Swift MA, Quake SR, Cornfield DN, Alvira CM. Diverse homeostatic and immunomodulatory roles of immune cells in the developing mouse lung at single cell resolution. eLife 2020; 9:e56890. [PMID: 32484158 PMCID: PMC7358008 DOI: 10.7554/elife.56890] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/13/2020] [Indexed: 12/20/2022] Open
Abstract
At birth, the lungs rapidly transition from a pathogen-free, hypoxic environment to a pathogen-rich, rhythmically distended air-liquid interface. Although many studies have focused on the adult lung, the perinatal lung remains unexplored. Here, we present an atlas of the murine lung immune compartment during early postnatal development. We show that the late embryonic lung is dominated by specialized proliferative macrophages with a surprising physical interaction with the developing vasculature. These macrophages disappear after birth and are replaced by a dynamic mixture of macrophage subtypes, dendritic cells, granulocytes, and lymphocytes. Detailed characterization of macrophage diversity revealed an orchestration of distinct subpopulations across postnatal development to fill context-specific functions in tissue remodeling, angiogenesis, and immunity. These data both broaden the putative roles for immune cells in the developing lung and provide a framework for understanding how external insults alter immune cell phenotype during a period of rapid lung growth and heightened vulnerability.
Collapse
Affiliation(s)
- Racquel Domingo-Gonzalez
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
- Center for Excellence in Pulmonary Biology, Stanford University School of MedicineStanfordUnited States
| | - Fabio Zanini
- Department of Bioengineering, Stanford UniversityStanfordUnited States
- Prince of Wales Clinical School, Lowy Cancer Research Centre, University of New South WalesSydneyAustralia
| | - Xibing Che
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
- Center for Excellence in Pulmonary Biology, Stanford University School of MedicineStanfordUnited States
- Division of Pulmonary, Asthma and Sleep Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
| | - Min Liu
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
- Center for Excellence in Pulmonary Biology, Stanford University School of MedicineStanfordUnited States
| | - Robert C Jones
- Department of Bioengineering, Stanford UniversityStanfordUnited States
| | - Michael A Swift
- Department of Chemical and Systems Biology, Stanford UniversityStanfordUnited States
| | - Stephen R Quake
- Department of Bioengineering, Stanford UniversityStanfordUnited States
- Chan Zuckerberg BiohubSan FranciscoUnited States
- Department of Applied Physics, Stanford UniversityStanfordUnited States
| | - David N Cornfield
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
- Center for Excellence in Pulmonary Biology, Stanford University School of MedicineStanfordUnited States
- Division of Pulmonary, Asthma and Sleep Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
| | - Cristina M Alvira
- Division of Critical Care Medicine, Department of Pediatrics, Stanford University School of MedicineStanfordUnited States
- Center for Excellence in Pulmonary Biology, Stanford University School of MedicineStanfordUnited States
| |
Collapse
|
23
|
Simpson LJ, Reader JS, Tzima E. Mechanical Regulation of Protein Translation in the Cardiovascular System. Front Cell Dev Biol 2020; 8:34. [PMID: 32083081 PMCID: PMC7006472 DOI: 10.3389/fcell.2020.00034] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 01/15/2020] [Indexed: 12/12/2022] Open
Abstract
The cardiovascular system can sense and adapt to changes in mechanical stimuli by remodeling the physical properties of the heart and blood vessels in order to maintain homeostasis. Imbalances in mechanical forces and/or impaired sensing are now not only implicated but are, in some cases, considered to be drivers for the development and progression of cardiovascular disease. There is now growing evidence to highlight the role of mechanical forces in the regulation of protein translation pathways. The canonical mechanism of protein synthesis typically involves transcription and translation. Protein translation occurs globally throughout the cell to maintain general function but localized protein synthesis allows for precise spatiotemporal control of protein translation. This Review will cover studies on the role of biomechanical stress -induced translational control in the heart (often in the context of physiological and pathological hypertrophy). We will also discuss the much less studied effects of mechanical forces in regulating protein translation in the vasculature. Understanding how the mechanical environment influences protein translational mechanisms in the cardiovascular system, will help to inform disease pathogenesis and potential areas of therapeutic intervention.
Collapse
Affiliation(s)
- Lisa J Simpson
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - John S Reader
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Ellie Tzima
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
24
|
Viola H, Chang J, Grunwell JR, Hecker L, Tirouvanziam R, Grotberg JB, Takayama S. Microphysiological systems modeling acute respiratory distress syndrome that capture mechanical force-induced injury-inflammation-repair. APL Bioeng 2019; 3:041503. [PMID: 31768486 PMCID: PMC6874511 DOI: 10.1063/1.5111549] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 11/08/2019] [Indexed: 12/14/2022] Open
Abstract
Complex in vitro models of the tissue microenvironment, termed microphysiological systems, have enormous potential to transform the process of discovering drugs and disease mechanisms. Such a paradigm shift is urgently needed in acute respiratory distress syndrome (ARDS), an acute lung condition with no successful therapies and a 40% mortality rate. Here, we consider how microphysiological systems could improve understanding of biological mechanisms driving ARDS and ultimately improve the success of therapies in clinical trials. We first discuss how microphysiological systems could explain the biological mechanisms underlying the segregation of ARDS patients into two clinically distinct phenotypes. Then, we contend that ARDS-mimetic microphysiological systems should recapitulate three critical aspects of the distal airway microenvironment, namely, mechanical force, inflammation, and fibrosis, and we review models that incorporate each of these aspects. Finally, we recognize the substantial challenges associated with combining inflammation, fibrosis, and/or mechanical force in microphysiological systems. Nevertheless, complex in vitro models are a novel paradigm for studying ARDS, and they could ultimately improve patient care.
Collapse
Affiliation(s)
| | - Jonathan Chang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, Georgia 30332, USA
| | - Jocelyn R. Grunwell
- Department of Pediatrics, Division of Critical Care Medicine, Children's Healthcare of Atlanta at Egleston, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Louise Hecker
- Division of Pulmonary, Allergy and Critical Care and Sleep Medicine, University of Arizona, Tucson, Arizona 85724, USA and Southern Arizona Veterans Affairs Health Care System, Tucson, Arizona 85723, USA
| | - Rabindra Tirouvanziam
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia 30322, USA and Center for CF and Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, Georgia 30322, USA
| | - James B. Grotberg
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | |
Collapse
|
25
|
Marrese M, Lonardoni D, Boi F, van Hoorn H, Maccione A, Zordan S, Iannuzzi D, Berdondini L. Investigating the Effects of Mechanical Stimulation on Retinal Ganglion Cell Spontaneous Spiking Activity. Front Neurosci 2019; 13:1023. [PMID: 31611765 PMCID: PMC6776634 DOI: 10.3389/fnins.2019.01023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 09/09/2019] [Indexed: 01/03/2023] Open
Abstract
Mechanical forces are increasingly recognized as major regulators of several physiological processes at both the molecular and cellular level; therefore, a deep understanding of the sensing of these forces and their conversion into electrical signals are essential for studying the mechanosensitive properties of soft biological tissues. To contribute to this field, we present a dual-purpose device able to mechanically stimulate retinal tissue and to record the spiking activity of retinal ganglion cells (RGCs). This new instrument relies on combining ferrule-top micro-indentation, which provides local measurements of viscoelasticity, with high-density multi-electrode array (HD-MEAs) to simultaneously record the spontaneous activity of the retina. In this paper, we introduce this instrument, describe its technical characteristics, and present a proof-of-concept experiment that shows how RGC spiking activity of explanted mice retinas respond to mechanical micro-stimulations of their photoreceptor layer. The data suggest that, under specific conditions of indentation, the retina perceive the mechanical stimulation as modulation of the visual input, besides the longer time-scale of activation, and the increase in spiking activity is not only localized under the indentation probe, but it propagates across the retinal tissue.
Collapse
Affiliation(s)
- Marica Marrese
- LaserLab, Department of Physics and Astronomy, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Davide Lonardoni
- NetS3 Laboratory, Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia, Genova, Italy
| | - Fabio Boi
- NetS3 Laboratory, Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia, Genova, Italy
| | - Hedde van Hoorn
- LaserLab, Department of Physics and Astronomy, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Alessandro Maccione
- NetS3 Laboratory, Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia, Genova, Italy
| | - Stefano Zordan
- NetS3 Laboratory, Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia, Genova, Italy
| | - Davide Iannuzzi
- LaserLab, Department of Physics and Astronomy, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Luca Berdondini
- NetS3 Laboratory, Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia, Genova, Italy
| |
Collapse
|
26
|
Kelly GT, Faraj R, Zhang Y, Maltepe E, Fineman JR, Black SM, Wang T. Pulmonary Endothelial Mechanical Sensing and Signaling, a Story of Focal Adhesions and Integrins in Ventilator Induced Lung Injury. Front Physiol 2019; 10:511. [PMID: 31105595 PMCID: PMC6498899 DOI: 10.3389/fphys.2019.00511] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 04/11/2019] [Indexed: 12/17/2022] Open
Abstract
Patients with critical illness such as acute lung injury often undergo mechanical ventilation in the intensive care unit. Though lifesaving in many instances, mechanical ventilation often results in ventilator induced lung injury (VILI), characterized by overdistension of lung tissue leading to release of edemagenic agents, which further damage the lung and contribute to the mortality and progression of pulmonary inflammation. The endothelium is particularly sensitive, as VILI associated mechanical stress results in endothelial cytoskeletal rearrangement, stress fiber formation, and integrity loss. At the heart of these changes are integrin tethered focal adhesions (FAs) which participate in mechanosensing, structure, and signaling. Here, we present the known roles of FA proteins including c-Src, talin, FAK, paxillin, vinculin, and integrins in the sensing and response to cyclic stretch and VILI associated stress. Attention is given to how stretch is propagated from the extracellular matrix through integrins to talin and other FA proteins, as well as signaling cascades that include FA proteins, leading to stress fiber formation and other cellular responses. This unifying picture of FAs aids our understanding in an effort to prevent and treat VILI.
Collapse
Affiliation(s)
- Gabriel T Kelly
- Department of Internal Medicine, College of Medicine Phoenix, The University of Arizona, Phoenix, AZ, United States
| | - Reem Faraj
- Department of Internal Medicine, College of Medicine Phoenix, The University of Arizona, Phoenix, AZ, United States
| | - Yao Zhang
- Department of Internal Medicine, College of Medicine Phoenix, The University of Arizona, Phoenix, AZ, United States
| | - Emin Maltepe
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| | - Jeffrey R Fineman
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| | - Stephen M Black
- Department of Medicine, College of Medicine, The University of Arizona, Tucson, AZ, United States
| | - Ting Wang
- Department of Internal Medicine, College of Medicine Phoenix, The University of Arizona, Phoenix, AZ, United States
| |
Collapse
|
27
|
Elhebeary M, Emon MAB, Aydin O, Saif MTA. A novel technique for in situ uniaxial tests of self-assembled soft biomaterials. LAB ON A CHIP 2019; 19:1153-1161. [PMID: 30776038 PMCID: PMC6437030 DOI: 10.1039/c8lc01273c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
We introduce a novel method to form 3D biomimetic tissues from a droplet of a cell-extracellular matrix (ECM) mixture on a sensor stage and to quantify tissue force and stiffness as a function of time under optical microscopes. This method exploits advances in micro-nano fabrication and capillarity for self-assembly and self-alignment of tissues on the stage. It allows simultaneous investigation of the microstructure of the tissue in situ while its mechanical response is quantified, thus linking tissue biophysics with physiology and revealing structural-functional properties of 3D tissues. We demonstrate the functionality of the stage by studying the mechanical behavior of different cell-collagen mixtures under mechanical, chemical and electrical stimulation. This includes force evolution in cell-free collagen during curing, myotubes differentiated from muscle cell-collagen/Matrigel ECM subjected to electrical stimulation, and fibroblast-collagen tissue subjected to cancer cell conditioned media (CM) and a Rho-kinase inhibitor, Y27632. Muscle contraction decreases with increasing frequency of electrical stimulation, and fibroblasts respond to CM by increasing contractility for a short time and completely relax in the presence of Y27632 but restore force with Y27632 washout.
Collapse
Affiliation(s)
- Mohamed Elhebeary
- University of Illinois at Urbana-Champaign, 1206 W. Green St, Urbana, IL 61801, USA.
| | | | | | | |
Collapse
|
28
|
Fois G, Winkelmann VE, Bareis L, Staudenmaier L, Hecht E, Ziller C, Ehinger K, Schymeinsky J, Kranz C, Frick M. ATP is stored in lamellar bodies to activate vesicular P2X 4 in an autocrine fashion upon exocytosis. J Gen Physiol 2017; 150:277-291. [PMID: 29282210 PMCID: PMC5806682 DOI: 10.1085/jgp.201711870] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 10/12/2017] [Accepted: 11/17/2017] [Indexed: 12/14/2022] Open
Abstract
P2X4 receptor activation facilitates secretion of pulmonary surfactant from secretory vesicles called lamellar bodies in alveolar epithelial cells. Fois et al. reveal that P2X4 receptors on the lamellar body membranes are activated by ATP stored within the vesicles themselves upon vesicle exocytosis. Vesicular P2X4 receptors are known to facilitate secretion and activation of pulmonary surfactant in the alveoli of the lungs. P2X4 receptors are expressed in the membrane of lamellar bodies (LBs), large secretory lysosomes that store lung surfactant in alveolar type II epithelial cells, and become inserted into the plasma membrane after exocytosis. Subsequent activation of P2X4 receptors by adenosine triphosphate (ATP) results in local fusion-activated cation entry (FACE), facilitating fusion pore dilation, surfactant secretion, and surfactant activation. Despite the importance of ATP in the alveoli, and hence lung function, the origin of ATP in the alveoli is still elusive. In this study, we demonstrate that ATP is stored within LBs themselves at a concentration of ∼1.9 mM. ATP is loaded into LBs by the vesicular nucleotide transporter but does not activate P2X4 receptors because of the low intraluminal pH (5.5). However, the rise in intravesicular pH after opening of the exocytic fusion pore results in immediate activation of vesicular P2X4 by vesicular ATP. Our data suggest a new model in which agonist (ATP) and receptor (P2X4) are located in the same intracellular compartment (LB), protected from premature degradation (ATP) and activation (P2X4), and ideally placed to ensure coordinated and timely receptor activation as soon as fusion occurs to facilitate surfactant secretion.
Collapse
Affiliation(s)
- Giorgio Fois
- Institute of General Physiology, Ulm University, Ulm, Germany
| | | | - Lara Bareis
- Institute of General Physiology, Ulm University, Ulm, Germany
| | | | - Elena Hecht
- Institute of General Physiology, Ulm University, Ulm, Germany
| | - Charlotte Ziller
- Institute of Analytical and Bioanalytical Chemistry, Ulm University, Ulm, Germany
| | | | - Jürgen Schymeinsky
- Immunology and Respiratory Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Christine Kranz
- Institute of Analytical and Bioanalytical Chemistry, Ulm University, Ulm, Germany
| | - Manfred Frick
- Institute of General Physiology, Ulm University, Ulm, Germany
| |
Collapse
|
29
|
Saluzzo S, Gorki AD, Rana BMJ, Martins R, Scanlon S, Starkl P, Lakovits K, Hladik A, Korosec A, Sharif O, Warszawska JM, Jolin H, Mesteri I, McKenzie ANJ, Knapp S. First-Breath-Induced Type 2 Pathways Shape the Lung Immune Environment. Cell Rep 2017; 18:1893-1905. [PMID: 28228256 PMCID: PMC5329122 DOI: 10.1016/j.celrep.2017.01.071] [Citation(s) in RCA: 191] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 12/27/2016] [Accepted: 01/26/2017] [Indexed: 01/07/2023] Open
Abstract
From birth onward, the lungs are exposed to the external environment and therefore harbor a complex immunological milieu to protect this organ from damage and infection. We investigated the homeostatic role of the epithelium-derived alarmin interleukin-33 (IL-33) in newborn mice and discovered the immediate upregulation of IL-33 from the first day of life, closely followed by a wave of IL-13-producing type 2 innate lymphoid cells (ILC2s), which coincided with the appearance of alveolar macrophages (AMs) and their early polarization to an IL-13-dependent anti-inflammatory M2 phenotype. ILC2s contributed to lung quiescence in homeostasis by polarizing tissue resident AMs and induced an M2 phenotype in transplanted macrophage progenitors. ILC2s continued to maintain the M2 AM phenotype during adult life at the cost of a delayed response to Streptococcus pneumoniae infection in mice. These data highlight the homeostatic role of ILC2s in setting the activation threshold in the lung and underline their implications in anti-bacterial defenses. The first breath triggers IL-33 induction by AEC2 in lungs of newborn mice IL-33 promotes the perinatal expansion and activation of ST2-expressing ILC2s ILC2-derived IL-13 polarizes newborn’s AMs into an M2 phenotype This homeostatic type 2 pathway delays antibacterial effector responses
Collapse
Affiliation(s)
- Simona Saluzzo
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria; MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Anna-Dorothea Gorki
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Batika M J Rana
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Rui Martins
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Seth Scanlon
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Philipp Starkl
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Karin Lakovits
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Anastasiya Hladik
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Ana Korosec
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Omar Sharif
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Joanna M Warszawska
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Helen Jolin
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Ildiko Mesteri
- Institute of Pathology Überlingen, Überlingen 88662, Germany
| | - Andrew N J McKenzie
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK.
| | - Sylvia Knapp
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria.
| |
Collapse
|
30
|
Bovard D, Iskandar A, Luettich K, Hoeng J, Peitsch MC. Organs-on-a-chip. TOXICOLOGY RESEARCH AND APPLICATION 2017. [DOI: 10.1177/2397847317726351] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In the last few years, considerable attention has been given to in vitro models in an attempt to reduce the use of animals and to decrease the rate of preclinical failure associated with the development of new drugs. Simple two-dimensional cultures grown in a dish are now frequently replaced by organotypic cultures with three-dimensional (3-D) architecture, which enables interactions between cells, promoting their differentiation and increasing their in vivo likeness. Microengineering now enables the incorporation of small devices into 3-D culture models to reproduce the complex microenvironment of the modeled organ, often referred to as organs-on-a-chip (OoCs). This review describes various OoCs developed to mimic liver, brain, kidney, and lung tissues. Current challenges encountered in attempts to recreate the in vivo environment are described, as well as some examples of OoCs. Finally, attention is given to the ongoing evolution of OoCs with the aim of solving one of the major limitations in that they can only represent a single organ. Multi-organ-on-a-chip (MOC) systems mimic organ interactions observed in the human body and aim to provide the features of compound uptake, metabolism, and excretion, while simultaneously allowing for insights into biological effects. MOCs might therefore represent a new paradigm in drug development, providing a better understanding of dose responses and mechanisms of toxicity, enabling the detection of drug resistance and supporting the evaluation of pharmacokinetic–pharmacodynamics parameters.
Collapse
Affiliation(s)
- David Bovard
- Philip Morris Products SA, Neuchatel, Switzerland
| | | | | | - Julia Hoeng
- Philip Morris Products SA, Neuchatel, Switzerland
| | | |
Collapse
|
31
|
A microengineered model of RBC transfusion-induced pulmonary vascular injury. Sci Rep 2017; 7:3413. [PMID: 28611413 PMCID: PMC5469736 DOI: 10.1038/s41598-017-03597-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 05/02/2017] [Indexed: 11/08/2022] Open
Abstract
Red blood cell (RBC) transfusion poses significant risks to critically ill patients by increasing their susceptibility to acute respiratory distress syndrome. While the underlying mechanisms of this life-threatening syndrome remain elusive, studies suggest that RBC-induced microvascular injury in the distal lung plays a central role in the development of lung injury following blood transfusion. Here we present a novel microengineering strategy to model and investigate this key disease process. Specifically, we created a microdevice for culturing primary human lung endothelial cells under physiological flow conditions to recapitulate the morphology and hemodynamic environment of the pulmonary microvascular endothelium in vivo. Perfusion of the microengineered vessel with human RBCs resulted in abnormal cytoskeletal rearrangement and release of intracellular molecules associated with regulated necrotic cell death, replicating the characteristics of acute endothelial injury in transfused lungs in vivo. Our data also revealed the significant effect of hemodynamic shear stress on RBC-induced microvascular injury. Furthermore, we integrated the microfluidic endothelium with a computer-controlled mechanical stretching system to show that breathing-induced physiological deformation of the pulmonary microvasculature may exacerbate vascular injury during RBC transfusion. Our biomimetic microsystem provides an enabling platform to mechanistically study transfusion-associated pulmonary vascular complications in susceptible patient populations.
Collapse
|
32
|
Liu AP, Chaudhuri O, Parekh SH. New advances in probing cell-extracellular matrix interactions. Integr Biol (Camb) 2017; 9:383-405. [PMID: 28352896 PMCID: PMC5708530 DOI: 10.1039/c6ib00251j] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Accepted: 03/20/2017] [Indexed: 12/17/2022]
Abstract
The extracellular matrix (ECM) provides structural and biochemical support to cells within tissues. An emerging body of evidence has established that the ECM plays a key role in cell mechanotransduction - the study of coupling between mechanical inputs and cellular phenotype - through either mediating transmission of forces to the cells, or presenting mechanical cues that guide cellular behaviors. Recent progress in cell mechanotransduction research has been facilitated by advances of experimental tools, particularly microtechnologies, engineered biomaterials, and imaging and analytical methods. Microtechnologies have enabled the design and fabrication of controlled physical microenvironments for the study and measurement of cell-ECM interactions. Advances in engineered biomaterials have allowed researchers to develop synthetic ECMs that mimic tissue microenvironments and investigate the impact of altered physicochemical properties on various cellular processes. Finally, advanced imaging and spectroscopy techniques have facilitated the visualization of the complex interaction between cells and ECM in vitro and in living tissues. This review will highlight the application of recent innovations in these areas to probing cell-ECM interactions. We believe cross-disciplinary approaches, combining aspects of the different technologies reviewed here, will inspire innovative ideas to further elucidate the secrets of ECM-mediated cell control.
Collapse
Affiliation(s)
- Allen P. Liu
- Department of Mechanical Engineering , University of Michigan , Ann Arbor , MI 48109 , USA .
- Department of Biomedical Engineering , University of Michigan , Ann Arbor , MI 48109 , USA
- Cellular and Molecular Biology Program , University of Michigan , Ann Arbor , MI 48109 , USA
- Biophysics Program , University of Michigan , Ann Arbor , MI 48109 , USA
| | - Ovijit Chaudhuri
- Department of Mechanical Engineering , Stanford University , Stanford , CA 94305 , USA .
| | - Sapun H. Parekh
- Department of Molecular Spectroscopy , Max Planck Institute for Polymer Research , Mainz 55128 , Germany .
| |
Collapse
|
33
|
Hsia CCW. Comparative analysis of the mechanical signals in lung development and compensatory growth. Cell Tissue Res 2017; 367:687-705. [PMID: 28084523 PMCID: PMC5321790 DOI: 10.1007/s00441-016-2558-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Accepted: 12/13/2016] [Indexed: 12/16/2022]
Abstract
This review compares the manner in which physical stress imposed on the parenchyma, vasculature and thorax and the thoraco-pulmonary interactions, drive both developmental and compensatory lung growth. Re-initiation of anatomical lung growth in the mature lung is possible when the loss of functioning lung units renders the existing physiologic-structural reserves insufficient for maintaining adequate function and physical stress on the remaining units exceeds a critical threshold. The appropriate spatial and temporal mechanical interrelationships and the availability of intra-thoracic space, are crucial to growth initiation, follow-on remodeling and physiological outcome. While the endogenous potential for compensatory lung growth is retained and may be pharmacologically augmented, supra-optimal mechanical stimulation, unbalanced structural growth, or inadequate remodeling may limit functional gain. Finding ways to optimize the signal-response relationships and resolve structure-function discrepancies are major challenges that must be overcome before the innate compensatory ability could be fully realized. Partial pneumonectomy reproducibly removes a known fraction of functioning lung units and remains the most robust model for examining the adaptive mechanisms, structure-function consequences and plasticity of the remaining functioning lung units capable of regeneration. Fundamental mechanical stimulus-response relationships established in the pneumonectomy model directly inform the exploration of effective approaches to maximize compensatory growth and function in chronic destructive lung diseases, transplantation and bioengineered lungs.
Collapse
Affiliation(s)
- Connie C W Hsia
- Department of Internal Medicine, Pulmonary and Critical Care Medicine, University of Texas Southwestern Medical Center, Dallas, 5323 Harry Hines Blvd., Dallas, TX, 75390-9034, USA.
| |
Collapse
|
34
|
Wang T, Gross C, Desai AA, Zemskov E, Wu X, Garcia AN, Jacobson JR, Yuan JXJ, Garcia JGN, Black SM. Endothelial cell signaling and ventilator-induced lung injury: molecular mechanisms, genomic analyses, and therapeutic targets. Am J Physiol Lung Cell Mol Physiol 2016; 312:L452-L476. [PMID: 27979857 DOI: 10.1152/ajplung.00231.2016] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 12/08/2016] [Accepted: 12/11/2016] [Indexed: 12/13/2022] Open
Abstract
Mechanical ventilation is a life-saving intervention in critically ill patients with respiratory failure due to acute respiratory distress syndrome (ARDS). Paradoxically, mechanical ventilation also creates excessive mechanical stress that directly augments lung injury, a syndrome known as ventilator-induced lung injury (VILI). The pathobiology of VILI and ARDS shares many inflammatory features including increases in lung vascular permeability due to loss of endothelial cell barrier integrity resulting in alveolar flooding. While there have been advances in the understanding of certain elements of VILI and ARDS pathobiology, such as defining the importance of lung inflammatory leukocyte infiltration and highly induced cytokine expression, a deep understanding of the initiating and regulatory pathways involved in these inflammatory responses remains poorly understood. Prevailing evidence indicates that loss of endothelial barrier function plays a primary role in the development of VILI and ARDS. Thus this review will focus on the latest knowledge related to 1) the key role of the endothelium in the pathogenesis of VILI; 2) the transcription factors that relay the effects of excessive mechanical stress in the endothelium; 3) the mechanical stress-induced posttranslational modifications that influence key signaling pathways involved in VILI responses in the endothelium; 4) the genetic and epigenetic regulation of key target genes in the endothelium that are involved in VILI responses; and 5) the need for novel therapeutic strategies for VILI that can preserve endothelial barrier function.
Collapse
Affiliation(s)
- Ting Wang
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Christine Gross
- Vascular Biology Center, Augusta University, Augusta, Georgia
| | - Ankit A Desai
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Evgeny Zemskov
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Xiaomin Wu
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Alexander N Garcia
- Department of Pharmacology University of Illinois at Chicago, Chicago, Illinois; and
| | - Jeffrey R Jacobson
- Department of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Jason X-J Yuan
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Joe G N Garcia
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona
| | - Stephen M Black
- Department of Medicine, The University of Arizona Health Sciences, Tucson, Arizona;
| |
Collapse
|
35
|
Majeski HE, Yang J. The 2016 John J. Abel Award Lecture: Targeting the Mechanical Microenvironment in Cancer. Mol Pharmacol 2016; 90:744-754. [PMID: 27742780 DOI: 10.1124/mol.116.106765] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 10/13/2016] [Indexed: 12/14/2022] Open
Abstract
Past decades of cancer research have mainly focused on the role of various extracellular and intracellular biochemical signals on cancer progression and metastasis. Recent studies suggest an important role of mechanical forces in regulating cellular behaviors. This review first provides an overview of the mechanobiology research field. Then we specially focus on mechanotransduction pathways in cancer progression and describe in detail the key signaling components of such mechanotransduction pathways and extracellular matrix components that are altered in cancer. Although our understanding of mechanoregulation in cancer is still in its infancy, some agents against key mechanoregulators have been developed and will be discussed to explore the potential of pharmacologically targeting mechanotransduction in cancer.
Collapse
Affiliation(s)
- Hannah E Majeski
- Department of Pharmacology (H.E.M., J.Y.), Department of Pediatrics (J.Y.), and Biomedical Sciences Graduate Program (H.E.M., J.Y.), Moores Cancer Center, University of California, San Diego, La Jolla, California
| | - Jing Yang
- Department of Pharmacology (H.E.M., J.Y.), Department of Pediatrics (J.Y.), and Biomedical Sciences Graduate Program (H.E.M., J.Y.), Moores Cancer Center, University of California, San Diego, La Jolla, California
| |
Collapse
|
36
|
The Cerebral Surfactant System and Its Alteration in Hydrocephalic Conditions. PLoS One 2016; 11:e0160680. [PMID: 27656877 PMCID: PMC5033422 DOI: 10.1371/journal.pone.0160680] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 07/24/2016] [Indexed: 01/06/2023] Open
Abstract
Introduction Pulmonary Surfactant reduces surface tension in the terminal airways thus facilitating breathing and contributes to host’s innate immunity. Surfactant Proteins (SP) A, B, C and D were recently identified as inherent proteins of the CNS. Aim of the study was to investigate cerebrospinal fluid (CSF) SP levels in hydrocephalus patients compared to normal subjects. Patients and Methods CSF SP A-D levels were quantified using commercially available ELISA kits in 126 patients (0–84 years, mean 39 years). 60 patients without CNS pathologies served as a control group. Hydrocephalus patients were separated in aqueductal stenosis (AQS, n = 24), acute hydrocephalus without aqueductal stenosis (acute HC w/o AQS, n = 16) and idiopathic normal pressure hydrocephalus (NPH, n = 20). Furthermore, six patients with pseudotumor cerebri were investigated. Results SP A—D are present under physiological conditions in human CSF. SP-A is elevated in diseases accompanied by ventricular enlargement (AQS, acute HC w/o AQS) in a significant manner (0.67, 1.21 vs 0.38 ng/ml in control, p<0.001). SP-C is also elevated in hydrocephalic conditions (AQS, acute HC w/o AQS; 0.87, 1.71 vs. 0.48 ng/ml in controls, p<0.001) and in Pseudotumor cerebri (1.26 vs. 0.48 ng/ml in controls, p<0.01). SP-B and SP-D did not show significant alterations. Conclusion The present study confirms the presence of SPs in human CSF. There are significant changes of SP-A and SP-C levels in diseases affecting brain water circulation and elevation of intracranial pressure. Cause of the alterations, underlying regulatory mechanisms, as well as diagnostic and therapeutic consequences of cerebral SP’s requires further thorough investigations.
Collapse
|
37
|
Abstract
Structural and functional complexities of the mammalian lung evolved to meet a unique set of challenges, namely, the provision of efficient delivery of inspired air to all lung units within a confined thoracic space, to build a large gas exchange surface associated with minimal barrier thickness and a microvascular network to accommodate the entire right ventricular cardiac output while withstanding cyclic mechanical stresses that increase several folds from rest to exercise. Intricate regulatory mechanisms at every level ensure that the dynamic capacities of ventilation, perfusion, diffusion, and chemical binding to hemoglobin are commensurate with usual metabolic demands and periodic extreme needs for activity and survival. This article reviews the structural design of mammalian and human lung, its functional challenges, limitations, and potential for adaptation. We discuss (i) the evolutionary origin of alveolar lungs and its advantages and compromises, (ii) structural determinants of alveolar gas exchange, including architecture of conducting bronchovascular trees that converge in gas exchange units, (iii) the challenges of matching ventilation, perfusion, and diffusion and tissue-erythrocyte and thoracopulmonary interactions. The notion of erythrocytes as an integral component of the gas exchanger is emphasized. We further discuss the signals, sources, and limits of structural plasticity of the lung in alveolar hypoxia and following a loss of lung units, and the promise and caveats of interventions aimed at augmenting endogenous adaptive responses. Our objective is to understand how individual components are matched at multiple levels to optimize organ function in the face of physiological demands or pathological constraints.
Collapse
Affiliation(s)
- Connie C.W. Hsia
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Dallas M. Hyde
- California National Primate Research Center, University of California at Davis, Davis, California, USA
| | | |
Collapse
|
38
|
Kilpatrick JI, Revenko I, Rodriguez BJ. Nanomechanics of Cells and Biomaterials Studied by Atomic Force Microscopy. Adv Healthc Mater 2015. [PMID: 26200464 DOI: 10.1002/adhm.201500229] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The behavior and mechanical properties of cells are strongly dependent on the biochemical and biomechanical properties of their microenvironment. Thus, understanding the mechanical properties of cells, extracellular matrices, and biomaterials is key to understanding cell function and to develop new materials with tailored mechanical properties for tissue engineering and regenerative medicine applications. Atomic force microscopy (AFM) has emerged as an indispensable technique for measuring the mechanical properties of biomaterials and cells with high spatial resolution and force sensitivity within physiologically relevant environments and timescales in the kPa to GPa elastic modulus range. The growing interest in this field of bionanomechanics has been accompanied by an expanding array of models to describe the complexity of indentation of hierarchical biological samples. Furthermore, the integration of AFM with optical microscopy techniques has further opened the door to a wide range of mechanotransduction studies. In recent years, new multidimensional and multiharmonic AFM approaches for mapping mechanical properties have been developed, which allow the rapid determination of, for example, cell elasticity. This Progress Report provides an introduction and practical guide to making AFM-based nanomechanical measurements of cells and surfaces for tissue engineering applications.
Collapse
Affiliation(s)
- Jason I. Kilpatrick
- Conway Institute of Biomolecular and Biomedical Research; University College Dublin; Belfield Dublin 4 Ireland
| | - Irène Revenko
- Asylum Research an Oxford Instruments Company; 6310 Hollister Avenue Santa Barbara CA 93117 USA
| | - Brian J. Rodriguez
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin; Belfield, Dublin 4, Ireland; School of Physics; University College Dublin; Belfield Dublin 4 Ireland
| |
Collapse
|
39
|
Vitzthum C, Clauss WG, Fronius M. Mechanosensitive activation of CFTR by increased cell volume and hydrostatic pressure but not shear stress. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:2942-51. [PMID: 26357939 DOI: 10.1016/j.bbamem.2015.09.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 09/03/2015] [Accepted: 09/05/2015] [Indexed: 12/20/2022]
Abstract
The cystic fibrosis transmembrane conductance regulator (CFTR) is a Cl(-) channel that is essential for electrolyte and fluid homeostasis. Preliminary evidence indicates that CFTR is a mechanosensitive channel. In lung epithelia, CFTR is exposed to different mechanical forces such as shear stress (Ss) and membrane distention. The present study questioned whether Ss and/or stretch influence CFTR activity (wild type, ∆F508, G551D). Human CFTR (hCFTR) was heterologously expressed in Xenopus oocytes and the response to the mechanical stimulus and forskolin/IBMX (FI) was measured by two-electrode voltage-clamp experiments. Ss had no influence on hCFTR activity. Injection of an intracellular analogous solution to increase cell volume alone did not affect hCFTR activity. However, hCFTR activity was augmented by injection after pre-stimulation with FI. The response to injection was similar in channels carrying the common mutations ∆F508 and G551D compared to wild type hCFTR. Stretch-induced CFTR activation was further assessed in Ussing chamber measurements using Xenopus lung preparations. Under control conditions increased hydrostatic pressure (HP) decreased the measured ion current including activation of a Cl(-) secretion that was unmasked by the CFTR inhibitor GlyH-101. These data demonstrate activation of CFTR in vitro and in a native pulmonary epithelium in response to mechanical stress. Mechanosensitive regulation of CFTR is highly relevant for pulmonary physiology that relies on ion transport processes facilitated by pulmonary epithelial cells.
Collapse
Affiliation(s)
- Constanze Vitzthum
- Institute of Animal Physiology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Wolfgang G Clauss
- Institute of Animal Physiology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Martin Fronius
- Department of Physiology, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
40
|
Geng Y, Wang Z. Review of cellular mechanotransduction on micropost substrates. Med Biol Eng Comput 2015; 54:249-71. [PMID: 26245253 DOI: 10.1007/s11517-015-1343-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Accepted: 07/07/2015] [Indexed: 01/09/2023]
Abstract
As physical entities, living cells can sense and respond to various stimulations within and outside the body through cellular mechanotransduction. Any deviation in cellular mechanotransduction will not only undermine the orchestrated regulation of mechanical responses, but also lead to the breakdown of their physiological function. Therefore, a quantitative study of cellular mechanotransduction needs to be conducted both in experiments and in computational simulations to investigate the underlying mechanisms of cellular mechanotransduction. In this review, we present an overview of the current knowledge and significant progress in cellular mechanotransduction via micropost substrates. In the aspect of experimental studies, we summarize significant experimental progress and place an emphasis on the coupled relationship among cellular spreading, focal adhesion and contractility as well as the influence of substrate properties on force-involved cellular behaviors. In the other aspect of computational investigations, we outline a coupled framework including the biochemically motivated stress fiber model and thermodynamically motivated adhesion model and present their predicted biomechanical responses and then compare predicted simulation results with experimental observations to further explore the mechanisms of cellular mechanotransduction. At last, we discuss the future perspectives both in experimental technologies and in computational models, as well as facing challenges in the area of cellular mechanotransduction.
Collapse
Affiliation(s)
- Yuxu Geng
- State Key Laboratory of Mechanical Transmission, Chongqing University, Chongqing, 400030, China
| | - Zhanjiang Wang
- State Key Laboratory of Mechanical Transmission, Chongqing University, Chongqing, 400030, China.
| |
Collapse
|
41
|
Hogan BLM, Barkauskas CE, Chapman HA, Epstein JA, Jain R, Hsia CCW, Niklason L, Calle E, Le A, Randell SH, Rock J, Snitow M, Krummel M, Stripp BR, Vu T, White ES, Whitsett JA, Morrisey EE. Repair and regeneration of the respiratory system: complexity, plasticity, and mechanisms of lung stem cell function. Cell Stem Cell 2014; 15:123-38. [PMID: 25105578 PMCID: PMC4212493 DOI: 10.1016/j.stem.2014.07.012] [Citation(s) in RCA: 641] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Respiratory disease is the third leading cause of death in the industrialized world. Consequently, the trachea, lungs, and cardiopulmonary vasculature have been the focus of extensive investigations. Recent studies have provided new information about the mechanisms driving lung development and differentiation. However, there is still much to learn about the ability of the adult respiratory system to undergo repair and to replace cells lost in response to injury and disease. This Review highlights the multiple stem/progenitor populations in different regions of the adult lung, the plasticity of their behavior in injury models, and molecular pathways that support homeostasis and repair.
Collapse
Affiliation(s)
- Brigid L M Hogan
- Department of Cell Biology, Duke Medicine, Durham, NC 27705, USA.
| | - Christina E Barkauskas
- Division of Pulmonary, Allergy and Critical Care Medicine, Duke Medicine, Durham, NC 27705, USA
| | - Harold A Chapman
- Division of Pulmonary and Critical Care, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jonathan A Epstein
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rajan Jain
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Connie C W Hsia
- Department of Internal Medicine, University of Texas, Southwestern Medical Center, Dallas, TX 75390, USA
| | - Laura Niklason
- Departments of Anesthesiology and Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Elizabeth Calle
- Department of Cell Biology, Duke Medicine, Durham, NC 27705, USA
| | - Andrew Le
- Department of Cell Biology, Duke Medicine, Durham, NC 27705, USA
| | - Scott H Randell
- Department of Cell Biology and Physiology, The University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Jason Rock
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Melinda Snitow
- Perleman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matthew Krummel
- Division of Pulmonary and Critical Care, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Barry R Stripp
- Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Thiennu Vu
- Division of Pulmonary and Critical Care, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Eric S White
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jeffrey A Whitsett
- Section of Neonatology, Perinatal and Pulmonary Biology, Department of Pediatrics, Cincinnati Children's Hospital Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Edward E Morrisey
- Departments of Medicine and Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
42
|
Mustafa SB, Isaac J, Seidner SR, Dixon PS, Henson BM, DiGeronimo RJ. Mechanical stretch induces lung α-epithelial Na(+) channel expression. Exp Lung Res 2014; 40:380-91. [PMID: 25058750 DOI: 10.3109/01902148.2014.934410] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
ABSTRACT During fetal development physiological stretching helps drive lung growth and maturation. At birth, the α-subunit of the alveolar epithelial sodium channel (α-ENaC) is a critical factor in helping to facilitate clearance of lung fluid during the perinatal period. The effects of stretch, however, on α-ENaC expression in the fetal lung have yet to be elucidated. In an effort to explore this question, we used both an in vitro cell culture model that exposes cells to repetitive cyclic stretch (CS) as well as an in vivo preterm animal model of mechanical ventilation (MV). We found that murine lung epithelial (MLE-12) cells exposed to repetitive CS showed a significant rise in α-ENaC mRNA expression. Total and cell-surface protein abundance of α-ENaC were also elevated after 24 h of CS. Stretch-induced increases in α-ENaC expression were suppressed in the presence of either actinomycin D or cycloheximide. Pharmacological inhibition of the extracellular signal-regulated protein kinase (ERK1/2) did not attenuate stretch-induced increases in α-ENaC protein, whereas inhibition of p38 MAPK or c-Jun NH2-terminal kinase (JNK) did. In 29-day preterm rabbits, alveolar stretching secondary to postnatal MV markedly elevated fetal lung α-ENaC expression compared to spontaneously breathing counterparts. In summary, our findings indicate that mechanical stretch promotes α-ENaC expression.
Collapse
Affiliation(s)
- Shamimunisa B Mustafa
- 1Department of Pediatrics, University of Texas Health Science Center , San Antonio Texas , USA
| | | | | | | | | | | |
Collapse
|
43
|
Treatment of acute lung injury by targeting MG53-mediated cell membrane repair. Nat Commun 2014; 5:4387. [PMID: 25034454 PMCID: PMC4109002 DOI: 10.1038/ncomms5387] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 06/13/2014] [Indexed: 12/27/2022] Open
Abstract
Injury to lung epithelial cells has a role in multiple lung diseases. We previously identified mitsugumin 53 (MG53) as a component of the cell membrane repair machinery in striated muscle cells. Here we show that MG53 also has a physiological role in the lung and may be used as a treatment in animal models of acute lung injury. Mice lacking MG53 show increased susceptibility to ischemia-reperfusion and over-ventilation induced injury to the lung when compared with wild type mice. Extracellular application of recombinant human MG53 (rhMG53) protein protects cultured lung epithelial cells against anoxia/reoxygenation-induced injuries. Intravenous delivery or inhalation of rhMG53 reduces symptoms in rodent models of acute lung injury and emphysema. Repetitive administration of rhMG53 improves pulmonary structure associated with chronic lung injury in mice. Our data indicate a physiological function for MG53 in the lung and suggest that targeting membrane repair may be an effective means for treatment or prevention of lung diseases.
Collapse
|
44
|
Calle EA, Ghaedi M, Sundaram S, Sivarapatna A, Tseng MK, Niklason LE. Strategies for whole lung tissue engineering. IEEE Trans Biomed Eng 2014; 61:1482-96. [PMID: 24691527 PMCID: PMC4126648 DOI: 10.1109/tbme.2014.2314261] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Recent work has demonstrated the feasibility of using decellularized lung extracellular matrix scaffolds to support the engineering of functional lung tissue in vitro. Rendered acellular through the use of detergents and other reagents, the scaffolds are mounted in organ-specific bioreactors where cells in the scaffold are provided with nutrients and appropriate mechanical stimuli such as ventilation and perfusion. Though initial studies are encouraging, a great deal remains to be done to advance the field and transition from rodent lungs to whole human tissue engineered lungs. To do so, a variety of hurdles must be overcome. In particular, a reliable source of human-sized scaffolds, as well as a method of terminal sterilization of scaffolds, must be identified. Continued research in lung cell and developmental biology will hopefully help identify the number and types of cells that will be required to regenerate functional lung tissue. Finally, bioreactor designs must be improved in order to provide more precise ventilation stimuli and vascular perfusion in order to avoid injury to or death of the cells cultivated within the scaffold. Ultimately, the success of efforts to engineer a functional lung in vitro will critically depend on the ability to create a fully endothelialized vascular network that provides sufficient barrier function and alveolar-capillary surface area to exchange gas at rates compatible with healthy lung function.
Collapse
Affiliation(s)
- Elizabeth A. Calle
- Department of Biomedical Engineering, Yale University, New Haven, CT 06519 USA
| | - Mahboobe Ghaedi
- Department of Anesthesia, Yale University, New Haven, CT 06519 USA
| | - Sumati Sundaram
- Department of Anesthesia, Yale University, New Haven, CT 06519 USA
| | - Amogh Sivarapatna
- Department of Biomedical Engineering, Yale University, New Haven, CT 06519 USA
| | - Michelle K. Tseng
- Department of Biomedical Engineering, Yale University, New Haven, CT 06519 USA
| | - Laura E. Niklason
- Department of Anesthesia and Department of Biomedical Engineering, Yale University, New Haven, CT 06519 USA
| |
Collapse
|
45
|
Guo J, Sachs F, Meng F. Fluorescence-based force/tension sensors: a novel tool to visualize mechanical forces in structural proteins in live cells. Antioxid Redox Signal 2014; 20:986-99. [PMID: 24205787 PMCID: PMC3924807 DOI: 10.1089/ars.2013.5708] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
SIGNIFICANCE Three signaling systems, chemical, electrical, and mechanical, ubiquitously contribute to cellular activities. There is limited information on the mechanical signaling system because of a lack of tools to measure stress in specific proteins. Although significant advances in methodologies such as atomic force microscopy and laser tweezers have achieved great success in single molecules and measuring the mean properties of cells and tissues, they cannot deal with specific proteins in live cells. RECENT ADVANCES To remedy the situation, we developed a family of genetically encoded optical force sensors to measure the stress in structural proteins in living cells. The sensors can be incorporated into specific proteins and are not harmful in transgenic animals. The chimeric proteins distribute and function as their wild-type counterparts, and local stress can be read out from changes in Förster resonance energy transfer (FRET). CRITICAL ISSUES Our original sensor used two mutant green fluorescence proteins linked by an alpha helix that served as a linking spring. Ever since, we have improved the probe design in a number of ways. For example, we replaced the helical linker with more common elastic protein domains to better match the compliance of the wild-type hosts. We greatly improved sensitivity by using the angular dependence of FRET rather than the distance dependence as the transduction mechanism, because that has nearly 100% efficiency at rest and nearly zero when stretched. FUTURE DIRECTIONS These probes enable researchers to investigate the roles of mechanical force in cellular activities at the level of single molecules, cells, tissues, and whole animals.
Collapse
Affiliation(s)
- Jun Guo
- 1 Department of Biochemistry, Nanjing Medical University , Nanjing, People's Republic of China
| | | | | |
Collapse
|
46
|
Sanchez-Esteban J. Mechanical forces in fetal lung development: opportunities for translational research. Front Pediatr 2013; 1:51. [PMID: 24400295 PMCID: PMC3872295 DOI: 10.3389/fped.2013.00051] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 12/11/2013] [Indexed: 12/29/2022] Open
Affiliation(s)
- Juan Sanchez-Esteban
- Department of Pediatrics, Alpert Medical School of Brown University , Providence, RI , USA
| |
Collapse
|
47
|
Cell shape-dependent early responses of fibroblasts to cyclic strain. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:3415-3425. [DOI: 10.1016/j.bbamcr.2013.10.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 10/04/2013] [Accepted: 10/10/2013] [Indexed: 01/16/2023]
|
48
|
Liu C, Li Q, Zhou X, Kolosov VP, Perelman JM. Cortactin mediates elevated shear stress-induced mucin hypersecretion via actin polymerization in human airway epithelial cells. Int J Biochem Cell Biol 2013; 45:2756-63. [DOI: 10.1016/j.biocel.2013.09.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 09/12/2013] [Accepted: 09/28/2013] [Indexed: 11/25/2022]
|
49
|
Sanchez-Esteban J. Growth factors and fetal lung development mediated by mechanical forces. World J Respirol 2013; 3:44-47. [DOI: 10.5320/wjr.v3.i3.44] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Revised: 07/04/2013] [Accepted: 07/19/2013] [Indexed: 02/06/2023] Open
Abstract
Incomplete development of the lung secondary to extreme prematurity or pulmonary hypoplasia causes significant morbidity and mortality during the neonatal period. Currently, the management is primarily supportive with no specific treatment to stimulate the growth and development of the lung. Mechanical forces generated inside the fetal lung by constant distention pressure and “breathing-like movements” are a major determinant of fetal lung development. However, the mechanisms by which lung cells sense these mechanical signals to promote lung development are not well-defined. Tracheal ligation has been used not only experimentally but also in human fetuses affected by severe congenital diaphragmatic hernia to stimulate lung growth and decrease the degree of pulmonary hypoplasia. Past investigations suggested that the increase of intratracheal pressure after tracheal ligation releases soluble factors that are critical for lung development. Studies from our laboratory have shown that mechanical strain of fetal type II epithelial cells, simulating mechanical forces in utero, promotes differentiation via release of epidermal growth factor receptor ligands heparin binding epidermal growth factor-like growth factor and transforming growth factor alpha. The identification of growth factors released by mechanical forces that are important for normal lung development could lead to novel treatments to accelerate lung development.
Collapse
|
50
|
Giordani VM, DeBenedictus CM, Wang Y, Sanchez-Esteban J. Epidermal growth factor receptor (EGFR) contributes to fetal lung fibroblast injury induced by mechanical stretch. J Recept Signal Transduct Res 2013; 34:58-63. [DOI: 10.3109/10799893.2013.862270] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|