201
|
Busch SM, Lorenzana Z, Ryan AL. Implications for Extracellular Matrix Interactions With Human Lung Basal Stem Cells in Lung Development, Disease, and Airway Modeling. Front Pharmacol 2021; 12:645858. [PMID: 34054525 PMCID: PMC8149957 DOI: 10.3389/fphar.2021.645858] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 04/29/2021] [Indexed: 12/18/2022] Open
Abstract
The extracellular matrix (ECM) is not simply a quiescent scaffold. This three-dimensional network of extracellular macromolecules provides structural, mechanical, and biochemical support for the cells of the lung. Throughout life, the ECM forms a critical component of the pulmonary stem cell niche. Basal cells (BCs), the primary stem cells of the airways capable of differentiating to all luminal cell types, reside in close proximity to the basolateral ECM. Studying BC-ECM interactions is important for the development of therapies for chronic lung diseases in which ECM alterations are accompanied by an apparent loss of the lung's regenerative capacity. The complexity and importance of the native ECM in the regulation of BCs is highlighted as we have yet to create an in vitro culture model that is capable of supporting the long-term expansion of multipotent BCs. The interactions between the pulmonary ECM and BCs are, therefore, a vital component for understanding the mechanisms regulating BC stemness during health and disease. If we are able to replicate these interactions in airway models, we could significantly improve our ability to maintain basal cell stemness ex vivo for use in in vitro models and with prospects for cellular therapies. Furthermore, successful, and sustained airway regeneration in an aged or diseased lung by small molecules, novel compounds or via cellular therapy will rely upon both manipulation of the airway stem cells and their immediate niche within the lung. This review will focus on the current understanding of how the pulmonary ECM regulates the basal stem cell function, how this relationship changes in chronic disease, and how replicating native conditions poses challenges for ex vivo cell culture.
Collapse
Affiliation(s)
- Shana M. Busch
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Zareeb Lorenzana
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, United States
| | - 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, United States
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
202
|
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
|
203
|
Goliwas KF, Ashraf HM, Wood AM, Wang Y, Hough KP, Bodduluri S, Athar M, Berry JL, Ponnazhagan S, Thannickal VJ, Deshane JS. Extracellular Vesicle Mediated Tumor-Stromal Crosstalk Within an Engineered Lung Cancer Model. Front Oncol 2021; 11:654922. [PMID: 33968758 PMCID: PMC8103208 DOI: 10.3389/fonc.2021.654922] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 04/06/2021] [Indexed: 12/14/2022] Open
Abstract
Tumor-stromal interactions within the tumor microenvironment (TME) influence lung cancer progression and response to therapeutic interventions, yet traditional in vitro studies fail to replicate the complexity of these interactions. Herein, we developed three-dimensional (3D) lung tumor models that mimic the human TME and demonstrate tumor-stromal crosstalk mediated by extracellular vesicles (EVs). EVs released by tumor cells, independent of p53 status, and fibroblasts within the TME mediate immunomodulatory effects; specifically, monocyte/macrophage polarization to a tumor-promoting M2 phenotype within this 3D-TME. Additionally, immune checkpoint inhibition in a 3D model that included T cells showed an inhibition of tumor growth and reduced hypoxia within the TME. Thus, perfused 3D tumor models incorporating diverse cell types provide novel insights into EV-mediated tumor-immune interactions and immune-modulation for existing and emerging cancer therapies.
Collapse
Affiliation(s)
- Kayla F Goliwas
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Hannah M Ashraf
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Anthony M Wood
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Yong Wang
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Kenneth P Hough
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Sandeep Bodduluri
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mohammad Athar
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Joel L Berry
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Selvarangan Ponnazhagan
- Department of Pathology, Division of Molecular and Cellular Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Victor J Thannickal
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jessy S Deshane
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
204
|
Mould KJ, Moore CM, McManus SA, McCubbrey AL, McClendon JD, Griesmer CL, Henson PM, Janssen WJ. Airspace Macrophages and Monocytes Exist in Transcriptionally Distinct Subsets in Healthy Adults. Am J Respir Crit Care Med 2021; 203:946-956. [PMID: 33079572 PMCID: PMC8048748 DOI: 10.1164/rccm.202005-1989oc] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 10/15/2020] [Indexed: 01/06/2023] Open
Abstract
Rationale: Macrophages are the most abundant immune cell in the alveoli and small airways and are traditionally viewed as a homogeneous population during health. Whether distinct subsets of airspace macrophages are present in healthy humans is unknown. Single-cell RNA sequencing allows for examination of transcriptional heterogeneity between cells and between individuals. Understanding the conserved repertoire of airspace macrophages during health is essential to understanding cellular programing during disease.Objectives: We sought to determine the transcriptional heterogeneity of human cells obtained from BAL of healthy adults.Methods: Ten subjects underwent bronchoscopy with BAL. Cells from lavage were subjected to single-cell RNA sequencing. Unique cell populations and putative functions were identified. Transcriptional profiles were compared across individuals.Measurements and Main Results: We identify two novel subgroups of resident airspace macrophages-defined by proinflammatory and metallothionein gene expression profiles. We define subsets of monocyte-like cells and compare them with peripheral blood mononuclear cells. Finally, we compare global macrophage and monocyte programing between males and females.Conclusions: Healthy human airspaces contain multiple populations of myeloid cells that are highly conserved between individuals and between sexes. Resident macrophages make up the largest population and include novel subsets defined by inflammatory and metal-binding profiles. Monocyte-like cells within the airspaces are transcriptionally aligned with circulating blood cells and include a rare population defined by expression of cell-matrix interaction genes. This study is the first to delineate the conserved heterogeneity of airspace immune cells during health and identifies two previously unrecognized macrophage subsets.
Collapse
Affiliation(s)
- Kara J. Mould
- Department of Medicine
- Department of Biomedical Research, and
| | - Camille M. Moore
- Department of Pediatrics, National Jewish Health, Denver, Colorado
- Department of Medicine, University of Colorado, Aurora, Colorado; and
| | | | | | | | | | - Peter M. Henson
- Department of Biomedical Research, and
- Department of Biostatistics and Informatics, University of Colorado, Denver, Colorado
| | | |
Collapse
|
205
|
Wanczyk H, Jensen T, Weiss DJ, Finck C. Advanced single-cell technologies to guide the development of bioengineered lungs. Am J Physiol Lung Cell Mol Physiol 2021; 320:L1101-L1117. [PMID: 33851545 DOI: 10.1152/ajplung.00089.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Lung transplantation remains the only viable option for individuals suffering from end-stage lung failure. However, a number of current limitations exist including a continuing shortage of suitable donor lungs and immune rejection following transplantation. To address these concerns, engineering a decellularized biocompatible lung scaffold from cadavers reseeded with autologous lung cells to promote tissue regeneration is being explored. Proof-of-concept transplantation of these bioengineered lungs into animal models has been accomplished. However, these lungs were incompletely recellularized with resulting epithelial and endothelial leakage and insufficient basement membrane integrity. Failure to repopulate lung scaffolds with all of the distinct cell populations necessary for proper function remains a significant hurdle for the progression of current engineering approaches and precludes clinical translation. Advancements in 3D bioprinting, lung organoid models, and microfluidic device and bioreactor development have enhanced our knowledge of pulmonary lung development, as well as important cell-cell and cell-matrix interactions, all of which will help in the path to a bioengineered transplantable lung. However, a significant gap in knowledge of the spatiotemporal interactions between cell populations as well as relative quantities and localization within each compartment of the lung necessary for its proper growth and function remains. This review will provide an update on cells currently used for reseeding decellularized scaffolds with outcomes of recent lung engineering attempts. Focus will then be on how data obtained from advanced single-cell analyses, coupled with multiomics approaches and high-resolution 3D imaging, can guide current lung bioengineering efforts for the development of fully functional, transplantable lungs.
Collapse
Affiliation(s)
- Heather Wanczyk
- Department of Pediatrics, University of Connecticut Health Center, Farmington, Connecticut
| | - Todd Jensen
- Department of Pediatrics, University of Connecticut Health Center, Farmington, Connecticut
| | - Daniel J Weiss
- Department of Medicine, University of Vermont, Burlington, Vermont
| | - Christine Finck
- Department of Pediatrics, University of Connecticut Health Center, Farmington, Connecticut.,Department of Surgery, Connecticut Children's Medical Center, Hartford, Connecticut
| |
Collapse
|
206
|
Karnati S, Seimetz M, Kleefeldt F, Sonawane A, Madhusudhan T, Bachhuka A, Kosanovic D, Weissmann N, Krüger K, Ergün S. Chronic Obstructive Pulmonary Disease and the Cardiovascular System: Vascular Repair and Regeneration as a Therapeutic Target. Front Cardiovasc Med 2021; 8:649512. [PMID: 33912600 PMCID: PMC8072123 DOI: 10.3389/fcvm.2021.649512] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a major cause of morbidity and mortality worldwide and encompasses chronic bronchitis and emphysema. It has been shown that vascular wall remodeling and pulmonary hypertension (PH) can occur not only in patients with COPD but also in smokers with normal lung function, suggesting a causal role for vascular alterations in the development of emphysema. Mechanistically, abnormalities in the vasculature, such as inflammation, endothelial dysfunction, imbalances in cellular apoptosis/proliferation, and increased oxidative/nitrosative stress promote development of PH, cor pulmonale, and most probably pulmonary emphysema. Hypoxemia in the pulmonary chamber modulates the activation of key transcription factors and signaling cascades, which propagates inflammation and infiltration of neutrophils, resulting in vascular remodeling. Endothelial progenitor cells have angiogenesis capabilities, resulting in transdifferentiation of the smooth muscle cells via aberrant activation of several cytokines, growth factors, and chemokines. The vascular endothelium influences the balance between vaso-constriction and -dilation in the heart. Targeting key players affecting the vasculature might help in the development of new treatment strategies for both PH and COPD. The present review aims to summarize current knowledge about vascular alterations and production of reactive oxygen species in COPD. The present review emphasizes on the importance of the vasculature for the usually parenchyma-focused view of the pathobiology of COPD.
Collapse
Affiliation(s)
- Srikanth Karnati
- Institute of Anatomy and Cell Biology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Michael Seimetz
- Excellence Cluster Cardio-Pulmonary System (ECCPS), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Florian Kleefeldt
- Institute of Anatomy and Cell Biology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Avinash Sonawane
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India
| | - Thati Madhusudhan
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Akash Bachhuka
- UniSA Science, Technology, Engineering and Mathematics, University of South Australia, Mawson Lakes Campus, Adelaide, SA, Australia
| | - Djuro Kosanovic
- Excellence Cluster Cardio-Pulmonary System (ECCPS), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany.,Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Norbert Weissmann
- Excellence Cluster Cardio-Pulmonary System (ECCPS), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Karsten Krüger
- Department of Exercise Physiology and Sports Therapy, University of Giessen, Giessen, Germany
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| |
Collapse
|
207
|
Biomimetic lipid Nanocomplexes incorporating STAT3-inhibiting peptides effectively infiltrate the lung barrier and ameliorate pulmonary fibrosis. J Control Release 2021; 332:160-170. [PMID: 33631224 DOI: 10.1016/j.jconrel.2021.02.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/14/2021] [Accepted: 02/18/2021] [Indexed: 01/09/2023]
Abstract
Activation of signal transducer and activator of transcription 3 (STAT3) under conditions of inflammation plays a crucial role in the pathogenesis of life-threatening pulmonary fibrosis (PF), initiating pro-fibrotic signaling following its phosphorylation. While there have been attempts to interfere with STAT3 activation and associated signaling as a strategy for ameliorating PF, potent inhibitors with minimal systemic toxicity have yet to be developed. Here, we assessed the in vitro and in vivo therapeutic effectiveness of a cell-permeable peptide inhibitor of STAT3 phosphorylation, designated APTstat3-9R, for ameliorating the indications of pulmonary fibrosis. Our results demonstrate that APTstat3-9R formulated with biomimetic disc-shaped lipid nanoparticles (DLNPs) markedly enhanced the penetration of pulmonary surfactant barrier and alleviated clinical symptoms of PF while causing negligible systemic cytotoxicity. Taken together, our findings suggest that biomimetic lipid nanoparticle-assisted pulmonary delivery of APTstat3-9R may be a feasible therapeutic option for PF in the clinic, and could be applied to treat other fibrotic diseases.
Collapse
|
208
|
Hoyer N, Jessen H, Prior TS, Sand JMB, Leeming DJ, Karsdal MA, Åttingsberg EKA, Vangsgaard GKM, Bendstrup E, Shaker SB. High turnover of types III and VI collagen in progressive idiopathic pulmonary fibrosis. Respirology 2021; 26:582-589. [PMID: 33834579 DOI: 10.1111/resp.14056] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/13/2021] [Accepted: 03/03/2021] [Indexed: 01/19/2023]
Abstract
BACKGROUND AND OBJECTIVE Prediction of idiopathic pulmonary fibrosis (IPF) progression is vital for the choice and timing of treatment and patient follow-up. This could potentially be achieved by prognostic blood biomarkers of extracellular matrix (ECM) remodelling. METHODS Neoepitope biomarkers of types III and VI collagen turnover (C3M, C6M, PRO-C3 and PRO-C6) were measured in 185 patients with newly diagnosed IPF. Disease severity at baseline and progression over 6 months was assessed by lung function tests and 6-min walk tests. All-cause mortality was assessed over a 3-year follow-up period. RESULTS High baseline levels of C3M, C6M, PRO-C3 and PRO-C6 were associated with more advanced disease at the time of diagnosis. Baseline levels of C6M and PRO-C3 were also associated with mortality over 3 years of follow-up (hazard ratio [HR]: 2.3, 95% CI: 1.3-3.9, p = 0.002 and HR: 1.8, 95% CI: 1.1-3.0, p = 0.03). Patients with several increased biomarkers at baseline, representing a high ECM remodelling phenotype, had more advanced disease at baseline, higher risk of progression or death at 6 months (OR: 1.4, 95% CI: 1.1-1.8, p = 0.002) and higher mortality over 3 years of follow-up (HR: 2.4, 95% CI: 1.3-4.5, p = 0.007). CONCLUSION Blood biomarkers of types III and VI collagen turnover, assessed at the time of diagnosis, are associated with several indices of disease severity, short-term progression and long-term mortality. These biomarkers can help to identify patients with a high ECM remodelling phenotype at high risk of disease progression and death.
Collapse
Affiliation(s)
- Nils Hoyer
- Department of Respiratory Medicine, Herlev and Gentofte University Hospital, Copenhagen, Denmark
| | - Henrik Jessen
- Nordic Bioscience, Biomarkers and Research, Herlev, Denmark
| | - Thomas S Prior
- Centre for Rare Lung Diseases, Department of Respiratory Disease and Allergy, Aarhus University Hospital, Aarhus, Denmark
| | | | | | | | - Emilia K A Åttingsberg
- Department of Respiratory Medicine, Herlev and Gentofte University Hospital, Copenhagen, Denmark
| | - Gustav K M Vangsgaard
- Department of Respiratory Medicine, Herlev and Gentofte University Hospital, Copenhagen, Denmark
| | - Elisabeth Bendstrup
- Centre for Rare Lung Diseases, Department of Respiratory Disease and Allergy, Aarhus University Hospital, Aarhus, Denmark
| | - Saher B Shaker
- Department of Respiratory Medicine, Herlev and Gentofte University Hospital, Copenhagen, Denmark
| |
Collapse
|
209
|
Campbell DR, Senger CN, Ryan AL, Magin CM. Engineering Tissue-Informed Biomaterials to Advance Pulmonary Regenerative Medicine. Front Med (Lausanne) 2021; 8:647834. [PMID: 33898484 PMCID: PMC8060451 DOI: 10.3389/fmed.2021.647834] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/09/2021] [Indexed: 11/13/2022] Open
Abstract
Biomaterials intentionally designed to support the expansion, differentiation, and three-dimensional (3D) culture of induced-pluripotent stem cells (iPSCs) may pave the way to cell-based therapies for chronic respiratory diseases. These conditions are endured by millions of people worldwide and represent a significant cause of morbidity and mortality. Currently, there are no effective treatments for the majority of advanced lung diseases and lung transplantation remains the only hope for many chronically ill patients. Key opinion leaders speculate that the novel coronavirus, COVID-19, may lead to long-term lung damage, further exacerbating the need for regenerative therapies. New strategies for regenerative cell-based therapies harness the differentiation capability of human iPSCs for studying pulmonary disease pathogenesis and treatment. Excitingly, biomaterials are a cell culture platform that can be precisely designed to direct stem cell differentiation. Here, we present a closer look at the state-of-the-art of iPSC differentiation for pulmonary engineering, offer evidence supporting the power of biomaterials to improve stem cell differentiation, and discuss our perspective on the potential for tissue-informed biomaterials to transform pulmonary regenerative medicine.
Collapse
Affiliation(s)
- Donald R. Campbell
- Department of Bioengineering, Denver, Anschutz Medical Campus, University of Colorado, Aurora, CO, United States
| | - Christiana N. Senger
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Hastings Center for Pulmonary Research, University of Southern California, Los Angeles, CA, United States
| | - Amy L. Ryan
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Hastings Center for Pulmonary Research, University of Southern California, Los Angeles, CA, United States
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, United States
| | - Chelsea M. Magin
- Department of Bioengineering, Denver, Anschutz Medical Campus, University of Colorado, Aurora, CO, United States
- Department of Pediatrics, Anschutz Medical Campus, University of Colorado, Aurora, CO, United States
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO, United States
| |
Collapse
|
210
|
Waeijen-Smit K, Houben-Wilke S, DiGiandomenico A, Gehrmann U, Franssen FME. Unmet needs in the management of exacerbations of chronic obstructive pulmonary disease. Intern Emerg Med 2021; 16:559-569. [PMID: 33616876 PMCID: PMC7897880 DOI: 10.1007/s11739-020-02612-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 12/17/2020] [Indexed: 12/11/2022]
Abstract
Exacerbations of chronic obstructive pulmonary disease (COPD) are episodes of acute worsening of respiratory symptoms that require additional therapy. These events play a pivotal role in the natural course of the disease and are associated with a progressive decline in lung function, reduced health status, a low physical activity level, tremendous health care costs, and increased mortality. Although most exacerbations have an infectious origin, the underlying mechanisms are heterogeneous and specific predictors of their occurrence in individual patients are currently unknown. Accurate prediction and early diagnosis of exacerbations is essential to develop novel targets for prevention and personalized treatments to reduce the impact of these events. Several potential biomarkers have previously been studied, these however lack specificity, accuracy and do not add value to the available clinical predictors. At present, microbial composition and host-microbiome interactions in the lung are increasingly recognized for their role in affecting the susceptibility to exacerbations, and may steer towards a novel direction in the management of COPD exacerbations. This narrative review describes the current challenges and unmet needs in the management of acute exacerbations of COPD. Exacerbation triggers, biological clusters, current treatment strategies, and their limitations, previously studied biomarkers and prediction tools, the lung microbiome and its role in COPD exacerbations as well as future directions are discussed.
Collapse
Affiliation(s)
- Kiki Waeijen-Smit
- Department of Research and Education, Ciro, Horn, NM, 6085, The Netherlands.
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht, The Netherlands.
- Department of Respiratory Medicine, Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands.
| | - Sarah Houben-Wilke
- Department of Research and Education, Ciro, Horn, NM, 6085, The Netherlands
| | - Antonio DiGiandomenico
- Discovery Microbiome, Microbial Sciences, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, USA
| | - Ulf Gehrmann
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Frits M E Franssen
- Department of Research and Education, Ciro, Horn, NM, 6085, The Netherlands
- NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht, The Netherlands
- Department of Respiratory Medicine, Maastricht University Medical Centre (MUMC+), Maastricht, The Netherlands
| |
Collapse
|
211
|
Liu G, Philp AM, Corte T, Travis MA, Schilter H, Hansbro NG, Burns CJ, Eapen MS, Sohal SS, Burgess JK, Hansbro PM. Therapeutic targets in lung tissue remodelling and fibrosis. Pharmacol Ther 2021; 225:107839. [PMID: 33774068 DOI: 10.1016/j.pharmthera.2021.107839] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 03/03/2021] [Indexed: 02/07/2023]
Abstract
Structural changes involving tissue remodelling and fibrosis are major features of many pulmonary diseases, including asthma, chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF). Abnormal deposition of extracellular matrix (ECM) proteins is a key factor in the development of tissue remodelling that results in symptoms and impaired lung function in these diseases. Tissue remodelling in the lungs is complex and differs between compartments. Some pathways are common but tissue remodelling around the airways and in the parenchyma have different morphologies. Hence it is critical to evaluate both common fibrotic pathways and those that are specific to different compartments; thereby expanding the understanding of the pathogenesis of fibrosis and remodelling in the airways and parenchyma in asthma, COPD and IPF with a view to developing therapeutic strategies for each. Here we review the current understanding of remodelling features and underlying mechanisms in these major respiratory diseases. The differences and similarities of remodelling are used to highlight potential common therapeutic targets and strategies. One central pathway in remodelling processes involves transforming growth factor (TGF)-β induced fibroblast activation and myofibroblast differentiation that increases ECM production. The current treatments and clinical trials targeting remodelling are described, as well as potential future directions. These endeavours are indicative of the renewed effort and optimism for drug discovery targeting tissue remodelling and fibrosis.
Collapse
Affiliation(s)
- Gang Liu
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Ashleigh M Philp
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia; St Vincent's Medical School, UNSW Medicine, UNSW, Sydney, NSW, Australia
| | - Tamera Corte
- Royal Prince Alfred Hospital, Camperdown, NSW, Australia; Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Mark A Travis
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre and Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, United Kingdom
| | - Heidi Schilter
- Pharmaxis Ltd, 20 Rodborough Road, Frenchs Forest, Sydney, NSW, Australia
| | - Nicole G Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia
| | - Chris J Burns
- Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Mathew S Eapen
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Launceston, TAS, Australia
| | - Sukhwinder S Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Launceston, TAS, Australia
| | - Janette K Burgess
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Department of Pathology and Medical Biology, Groningen, The Netherlands; Woolcock Institute of Medical Research, Discipline of Pharmacology, The University of Sydney, Sydney, NSW, Australia
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, NSW, Australia.
| |
Collapse
|
212
|
Acinetobacter baumannii: An Ancient Commensal with Weapons of a Pathogen. Pathogens 2021; 10:pathogens10040387. [PMID: 33804894 PMCID: PMC8063835 DOI: 10.3390/pathogens10040387] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/19/2021] [Accepted: 03/23/2021] [Indexed: 12/22/2022] Open
Abstract
Acinetobacter baumannii is regarded as a life-threatening pathogen associated with community-acquired and nosocomial infections, mainly pneumonia. The rise in the number of A. baumannii antibiotic-resistant strains reduces effective therapies and increases mortality. Bacterial comparative genomic studies have unraveled the innate and acquired virulence factors of A. baumannii. These virulence factors are involved in antibiotic resistance, environmental persistence, host-pathogen interactions, and immune evasion. Studies on host–pathogen interactions revealed that A. baumannii evolved different mechanisms to adhere to in order to invade host respiratory cells as well as evade the host immune system. In this review, we discuss current data on A. baumannii genetic features and virulence factors. An emphasis is given to the players in host–pathogen interaction in the respiratory tract. In addition, we report recent investigations into host defense systems using in vitro and in vivo models, providing new insights into the innate immune response to A. baumannii infections. Increasing our knowledge of A. baumannii pathogenesis may help the development of novel therapeutic strategies based on anti-adhesive, anti-virulence, and anti-cell to cell signaling pathways drugs.
Collapse
|
213
|
NOX4-Derived ROS Promotes Collagen I Deposition in Bronchial Smooth Muscle Cells by Activating Noncanonical p38MAPK/Akt-Mediated TGF- β Signaling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6668971. [PMID: 33824697 PMCID: PMC8007363 DOI: 10.1155/2021/6668971] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 02/10/2021] [Accepted: 02/18/2021] [Indexed: 01/17/2023]
Abstract
Background Airway smooth muscle (ASM) remodeling is a hallmark in chronic obstructive pulmonary disease (COPD). NADPH oxidase 4- (NOX4-) mediated reactive oxygen species (ROS) production plays a crucial role in cell differentiation and extracellular matrix (ECM) synthesis in ASM remodeling. However, the precise mechanisms underpinning its pathogenic roles remain elusive. Methods The expression of NOX4 and TGF-β1 in the airway of the lung was measured in COPD patients and the control group. Cigarette smoke- (CS-) induced emphysema mice were generated, and the alteration of α-SMA, NOX4, TGF-β1, and collagen I was accessed. The changes of the expression of ECM markers, NOX4, components of TGF-β/Smad, and MAPK/Akt signaling in human bronchial smooth muscle cells (HBSMCs) were ascertained for delineating mechanisms of NOX4-mediated ROS production on cell differentiation and remodeling in human ASM cells. Results An increased abundance of NOX4 and TGF-β1 proteins in the epithelial cells and ASM of lung was observed in COPD patients compared with the control group. Additionally, an increased abundance expression of NOX4 and α-SMA was observed in the lungs of the CS-induced emphysema mouse model. TGF-β1 displayed abilities to increase the oxidative burden and collagen I production, along with enhanced phosphorylation of ERK, p38MAPK, and p-Akt473 in HBSMCs. These effects of TGF-β1 could be inhibited by the ROS scavenger N-acetylcysteine (NAC), siRNA-mediated knockdown of Smad3 and NOX4, and pharmacological inhibitors SB203580 (p38MAPK inhibitor) and LY294002 (Akt inhibitor). Conclusions NOX4-mediated ROS production alters TGF-β1-induced cell differentiation and collagen I protein synthesis in HBSMCs in part through the p38MAPK/Akt signaling pathway in a Smad-dependent manner.
Collapse
|
214
|
Baruffaldi D, Palmara G, Pirri C, Frascella F. 3D Cell Culture: Recent Development in Materials with Tunable Stiffness. ACS APPLIED BIO MATERIALS 2021; 4:2233-2250. [PMID: 35014348 DOI: 10.1021/acsabm.0c01472] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
It is widely accepted that three-dimensional cell culture systems simulate physiological conditions better than traditional 2D systems. Although extracellular matrix components strongly modulate cell behavior, several studies underlined the importance of mechanosensing in the control of different cell functions such as growth, proliferation, differentiation, and migration. Human tissues are characterized by different degrees of stiffness, and various pathologies (e.g., tumor or fibrosis) cause changes in the mechanical properties through the alteration of the extracellular matrix structure. Additionally, these modifications have an impact on disease progression and on therapy response. Hence, the development of platforms whose stiffness could be modulated may improve our knowledge of cell behavior under different mechanical stress stimuli. In this review, we have analyzed the mechanical diversity of healthy and diseased tissues, and we have summarized recently developed materials with a wide range of stiffness.
Collapse
Affiliation(s)
- Désirée Baruffaldi
- Dipartimento di Scienza Applicata e Tecnologia, Politecnico di Torino, C.so Duca degli Abruzzi 24, Turin 10129, Italy.,PolitoBIOMed Lab, Politecnico di Torino, C.so Duca degli Abruzzi 24, Turin 10129, Italy
| | - Gianluca Palmara
- Dipartimento di Scienza Applicata e Tecnologia, Politecnico di Torino, C.so Duca degli Abruzzi 24, Turin 10129, Italy.,PolitoBIOMed Lab, Politecnico di Torino, C.so Duca degli Abruzzi 24, Turin 10129, Italy
| | - Candido Pirri
- Dipartimento di Scienza Applicata e Tecnologia, Politecnico di Torino, C.so Duca degli Abruzzi 24, Turin 10129, Italy.,PolitoBIOMed Lab, Politecnico di Torino, C.so Duca degli Abruzzi 24, Turin 10129, Italy.,Center for Sustainable Futures@Polito, Istituto Italiano di Tecnologia, Via Livorno 60, Turin 10144, Italy
| | - Francesca Frascella
- Dipartimento di Scienza Applicata e Tecnologia, Politecnico di Torino, C.so Duca degli Abruzzi 24, Turin 10129, Italy.,PolitoBIOMed Lab, Politecnico di Torino, C.so Duca degli Abruzzi 24, Turin 10129, Italy
| |
Collapse
|
215
|
Zamprogno P, Thoma G, Cencen V, Ferrari D, Putz B, Michler J, Fantner GE, Guenat OT. Mechanical Properties of Soft Biological Membranes for Organ-on-a-Chip Assessed by Bulge Test and AFM. ACS Biomater Sci Eng 2021; 7:2990-2997. [PMID: 33651947 DOI: 10.1021/acsbiomaterials.0c00515] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Advanced in vitro models called "organ-on-a-chip" can mimic the specific cellular environment found in various tissues. Many of these models include a thin, sometimes flexible, membrane aimed at mimicking the extracellular matrix (ECM) scaffold of in vivo barriers. These membranes are often made of polydimethylsiloxane (PDMS), a silicone rubber that poorly mimics the chemical and physical properties of the basal membrane. However, the ECM and its mechanical properties play a key role in the homeostasis of a tissue. Here, we report about biological membranes with a composition and mechanical properties similar to those found in vivo. Two types of collagen-elastin (CE) membranes were produced: vitrified and nonvitrified (called "hydrogel membrane"). Their mechanical properties were characterized using the bulge test method. The results were compared using atomic force microscopy (AFM), a standard technique used to evaluate the Young's modulus of soft materials at the nanoscale. Our results show that CE membranes with stiffnesses ranging from several hundred of kPa down to 1 kPa can be produced by tuning the CE ratio, the production mode (vitrified or not), and/or certain parameters such as temperature. The Young's modulus can easily be determined using the bulge test. This method is a robust and reproducible to determine membrane stiffness, even for soft membranes, which are more difficult to assess by AFM. Assessment of the impact of substrate stiffness on the spread of human fibroblasts on these surfaces showed that cell spread is lower on softer surfaces than on stiffer surfaces.
Collapse
Affiliation(s)
- Pauline Zamprogno
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern 3008, Switzerland
| | - Giuditta Thoma
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern 3008, Switzerland
| | - Veronika Cencen
- Laboratory for Bio- and Nano- Instrumentation, Ecole Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Dario Ferrari
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern 3008, Switzerland
| | - Barbara Putz
- Laboratory for Mechanics of Materials and Nanostructures, EMPA Swiss Federal Laboratories for Materials Science and Technology, Thun 3602, Switzerland
| | - Johann Michler
- Laboratory for Mechanics of Materials and Nanostructures, EMPA Swiss Federal Laboratories for Materials Science and Technology, Thun 3602, Switzerland
| | - Georg E Fantner
- Laboratory for Bio- and Nano- Instrumentation, Ecole Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Olivier T Guenat
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern 3008, Switzerland.,Department of Pulmonary Medicine, University Hospital of Bern, Bern 3008, Switzerland.,Department of General Thoracic Surgery, University Hospital of Bern, Bern 3008, Switzerland
| |
Collapse
|
216
|
Yeoman B, Shatkin G, Beri P, Banisadr A, Katira P, Engler AJ. Adhesion strength and contractility enable metastatic cells to become adurotactic. Cell Rep 2021; 34:108816. [PMID: 33691109 PMCID: PMC7997775 DOI: 10.1016/j.celrep.2021.108816] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 01/10/2021] [Accepted: 02/10/2021] [Indexed: 11/05/2022] Open
Abstract
Significant changes in cell stiffness, contractility, and adhesion, i.e., mechanotype, are observed during a variety of biological processes. Whether cell mechanics merely change as a side effect of or driver for biological processes is still unclear. Here, we sort genotypically similar metastatic cancer cells into strongly adherent (SA) versus weakly adherent (WA) phenotypes to study how contractility and adhesion differences alter the ability of cells to sense and respond to gradients in material stiffness. We observe that SA cells migrate up a stiffness gradient, or durotax, while WA cells largely ignore the gradient, i.e., adurotax. Biophysical modeling and experimental validation suggest that differences in cell migration and durotaxis between weakly and strongly adherent cells are driven by differences in intra-cellular actomyosin activity. These results provide a direct relationship between cell phenotype and durotaxis and suggest how, unlike other senescent cells, metastatic cancer cells navigate against stiffness gradients.
Collapse
Affiliation(s)
- Benjamin Yeoman
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Department of Mechanical Engineering, San Diego State University, San Diego, CA 92182, USA
| | - Gabriel Shatkin
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Pranjali Beri
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Afsheen Banisadr
- Biomedical Sciences Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Parag Katira
- Department of Mechanical Engineering, San Diego State University, San Diego, CA 92182, USA; Computational Sciences Research Center, San Diego State University, San Diego, CA 92182, USA.
| | - Adam J Engler
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Biomedical Sciences Program, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
217
|
Eosinophil Responses at the Airway Epithelial Barrier during the Early Phase of Influenza A Virus Infection in C57BL/6 Mice. Cells 2021; 10:cells10030509. [PMID: 33673645 PMCID: PMC7997358 DOI: 10.3390/cells10030509] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/09/2021] [Accepted: 02/12/2021] [Indexed: 02/07/2023] Open
Abstract
Eosinophils, previously considered terminally differentiated effector cells, have multifaceted functions in tissues. We previously found that allergic mice with eosinophil-rich inflammation were protected from severe influenza and discovered specialized antiviral effector functions for eosinophils including promoting cellular immunity during influenza. In this study, we hypothesized that eosinophil responses during the early phase of influenza contribute to host protection. Using in vitro and in vivo models, we found that eosinophils were rapidly and dynamically regulated upon influenza A virus (IAV) exposure to gain migratory capabilities to traffic to lymphoid organs after pulmonary infection. Eosinophils were capable of neutralizing virus upon contact and combinations of eosinophil granule proteins reduced virus infectivity through hemagglutinin inactivation. Bi-directional crosstalk between IAV-exposed epithelial cells and eosinophils occurred after IAV infection and cross-regulation promoted barrier responses to improve antiviral defenses in airway epithelial cells. Direct interactions between eosinophils and airway epithelial cells after IAV infection prevented virus-induced cytopathology in airway epithelial cells in vitro, and eosinophil recipient IAV-infected mice also maintained normal airway epithelial cell morphology. Our data suggest that eosinophils are important in the early phase of IAV infection providing immediate protection to the epithelial barrier until adaptive immune responses are deployed during influenza.
Collapse
|
218
|
Garrison CM, Schwarzbauer JE. Fibronectin fibril alignment is established upon initiation of extracellular matrix assembly. Mol Biol Cell 2021; 32:739-752. [PMID: 33625865 PMCID: PMC8108514 DOI: 10.1091/mbc.e20-08-0533] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The physical structure of the extracellular matrix (ECM) is tissue-specific and fundamental to normal tissue function. Proper alignment of ECM fibers is essential for the functioning of a variety of tissues. While matrix assembly in general has been intensively investigated, little is known about the mechanisms required for formation of aligned ECM fibrils. We investigated the initiation of fibronectin (FN) matrix assembly using fibroblasts that assemble parallel ECM fibrils and found that matrix assembly sites, where FN fibrillogenesis is initiated, were oriented in parallel at the cell poles. We show that these polarized matrix assembly sites progress into fibrillar adhesions and ultimately into aligned FN fibrils. Cells that assemble an unaligned meshwork matrix form matrix assembly sites around the cell periphery, but the distribution of matrix assembly sites in these cells could be modulated through micropatterning or mechanical stretch. While an elongated cell shape corresponds with a polarized matrix assembly site distribution, these two features are not absolutely linked, since we discovered that transforming growth factor beta (TGF-β1) enhances matrix assembly site polarity and assembly of aligned fibrils independent of cell elongation. We conclude that the ultimate orientation of FN fibrils is determined by the alignment and distribution of matrix assembly sites that form during the initial stages of cell–FN interactions.
Collapse
Affiliation(s)
- Carly M Garrison
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | | |
Collapse
|
219
|
Santos MP, Gonçalves-Santos E, Gonçalves RV, Santos EC, Campos CC, Bastos DSS, Marques MJ, Souza RLM, Novaes RD. Doxycycline aggravates granulomatous inflammation and lung microstructural remodeling induced by Schistosoma mansoni infection. Int Immunopharmacol 2021; 94:107462. [PMID: 33611055 DOI: 10.1016/j.intimp.2021.107462] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 12/20/2022]
Abstract
Although doxycycline exhibits immunomodulatory properties, its effects on pulmonary infection by Schistosoma mansoni remain overlooked. Thus, we investigated the impact of this drug on lung granulomatous inflammation and microstructural remodeling in a murine model of schistosomiasis. Swiss mice were randomized in four groups: (i) uninfected, (ii) infected with S. mansoni and untreated, (iii) infected treated with praziquantel (Pzq; 200 mg/kg), and (iv) infected treated with Dox (50 mg/kg). Pz was administered in a single dose, and Dox for 60 days. S. mansoni induced marked granulomatous lung inflammation, which was associated to cytokines upregulation (IL-2, IL-4, IL-10, IFN-γ, TNF-α, and TGF-β), neutrophils and macrophages recruitment, alveolar collapse, lung fibrosis, and extensive depletion of elastic fibers. These parameters were attenuated by Pzq and aggravated by Dox. Exudative/productive granulomas were predominant in untreated and Dox-treated animals, while fibrotic granulomas were more frequent in Pzq-treated mice. The number and size of granulomas in Dox-treated animals was higher than untreated and Pzq-treated mice. Dox treatment inhibited the increase in MMP-1 and MMP-2 activity but upregulated myeloperoxidase and N-acetylglucosaminidase activity compared to untreated and Pzq-treated animals. Dox and Pzq exerted no effect on elastin depletion and upregulation of elastase activity. Together, our findings indicated that Dox aggravated granulomatous inflammation, accelerating lung microstructural remodeling by downregulating MMP-1 and MMP-2 activity without impair neutrophils and macrophages recruitment or elastase activity. Thus, Dox potentiates inflammatory damage associated with lung fibrosis, elastin depletion and massive alveolar collapse, profoundly subverting lung structure in S. mansoni-infected mice.
Collapse
Affiliation(s)
- Margarida P Santos
- Institute of Biomedical Sciences, Department of Structural Biology, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | - Elda Gonçalves-Santos
- Institute of Biomedical Sciences, Department of Structural Biology, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | - Reggiani V Gonçalves
- Department of Animal Biology, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | - Eliziária C Santos
- School of Medicine, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Minas Gerais, Brazil
| | - Camila C Campos
- Institute of Biomedical Sciences, Department of Structural Biology, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | - Daniel S S Bastos
- Department of General Biology, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | - Marcos J Marques
- Institute of Biomedical Sciences, Department of Pathology and Parasitology, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | - Raquel L M Souza
- Institute of Biomedical Sciences, Department of Pathology and Parasitology, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | - Rômulo D Novaes
- Institute of Biomedical Sciences, Department of Structural Biology, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil.
| |
Collapse
|
220
|
Zamprogno P, Wüthrich S, Achenbach S, Thoma G, Stucki JD, Hobi N, Schneider-Daum N, Lehr CM, Huwer H, Geiser T, Schmid RA, Guenat OT. Second-generation lung-on-a-chip with an array of stretchable alveoli made with a biological membrane. Commun Biol 2021; 4:168. [PMID: 33547387 PMCID: PMC7864995 DOI: 10.1038/s42003-021-01695-0] [Citation(s) in RCA: 138] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 01/12/2021] [Indexed: 02/07/2023] Open
Abstract
The air-blood barrier with its complex architecture and dynamic environment is difficult to mimic in vitro. Lung-on-a-chips enable mimicking the breathing movements using a thin, stretchable PDMS membrane. However, they fail to reproduce the characteristic alveoli network as well as the biochemical and physical properties of the alveolar basal membrane. Here, we present a lung-on-a-chip, based on a biological, stretchable and biodegradable membrane made of collagen and elastin, that emulates an array of tiny alveoli with in vivo-like dimensions. This membrane outperforms PDMS in many ways: it does not absorb rhodamine-B, is biodegradable, is created by a simple method, and can easily be tuned to modify its thickness, composition and stiffness. The air-blood barrier is reconstituted using primary lung alveolar epithelial cells from patients and primary lung endothelial cells. Typical alveolar epithelial cell markers are expressed, while the barrier properties are preserved for up to 3 weeks.
Collapse
Affiliation(s)
- Pauline Zamprogno
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Simon Wüthrich
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Sven Achenbach
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Giuditta Thoma
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Janick D Stucki
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
- AlveoliX AG, Bern, Switzerland
| | - Nina Hobi
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
- AlveoliX AG, Bern, Switzerland
| | - Nicole Schneider-Daum
- Drug Delivery (DDEL), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Germany
| | - Claus-Michael Lehr
- Drug Delivery (DDEL), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Germany
| | - Hanno Huwer
- SHG Clinics, Department of Cardiothoracic Surgery, Völklingen Heart Center, Völklingen, Germany
| | - Thomas Geiser
- Department of Pulmonary Medicine, University Hospital of Bern, Bern, Switzerland
| | - Ralph A Schmid
- Department of General Thoracic Surgery, University Hospital of Bern, Bern, Switzerland
| | - Olivier T Guenat
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland.
- Department of Pulmonary Medicine, University Hospital of Bern, Bern, Switzerland.
- Department of General Thoracic Surgery, University Hospital of Bern, Bern, Switzerland.
| |
Collapse
|
221
|
McDonald LT. Healing after COVID-19: are survivors at risk for pulmonary fibrosis? Am J Physiol Lung Cell Mol Physiol 2021; 320:L257-L265. [PMID: 33355522 PMCID: PMC7900916 DOI: 10.1152/ajplung.00238.2020] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 12/18/2020] [Accepted: 12/20/2020] [Indexed: 12/16/2022] Open
Abstract
The novel SARS-CoV-2 coronavirus, which is responsible for COVID-19 disease, was first reported in Wuhan, China, in December of 2019. The virus rapidly spread, and the World Health Organization declared a pandemic by March 2020. With millions of confirmed cases worldwide, there is growing concern and considerable debate regarding the potential for coronavirus infection to contribute to an appreciable burden of chronic respiratory symptoms or fibrotic disease among recovered individuals. Because the first case of COVID-19 was documented less than one year ago, data regarding long-term clinical outcomes are not yet available, and predictions for long-term outcome are speculative at best. However, due to the staggering number of cases and the severity of disease in many individuals, there is a critical need to consider the potential long-term implications of COVID-19. This review examines current basic and clinical data regarding fibrogenic mechanisms of viral injury in the context of SARS-CoV-2. Several intersecting mechanisms between coronavirus infection and fibrotic pathways are discussed to highlight factors and processes that may be targetable to improve patient outcome. Reports of post-infection sequelae from previous coronavirus outbreaks are presented toward the goal of improved recognition of potential contributing risk factors for fibrotic disease.
Collapse
Affiliation(s)
- Lindsay T McDonald
- Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| |
Collapse
|
222
|
Abstract
PURPOSE OF REVIEW 'Biomarkers of remodeling' represent a loose collection of features referring to several biological adaptations of the lung to cope with stressing factors. In addition, remodel-'ing' infers a dynamic process that would require a spatiotemporal resolution. This review focuses on different aspects of remodeling in pediatric and adult care. RECENT FINDINGS This review will cover aspects of pediatric remodeling, adult remodeling and techniques and procedures to adequately assess remodeling across different age spectra. In pediatrics, the onset and first features of remodeling are discussed and the continuation into adolescence is addressed. For adults, this review addresses predominant features of remodeling throughout the adult life span and whether there are currently interventions available to treat or reverse remodeling. SUMMARY The term 'remodeling' is often referred to via biomarkers that reflect the endstage of a process, although it rather reflects a continuous process starting in childhood and progressing to all age-levels in patients with asthma. Hence, only few biomarkers or surrogates are able to 'capture' its spatiotemporal component, and hardly any are ready for routine use in clinical practice. Given the clinical impact of the remodeling processes, new biomarkers are needed to adequately treat patients with asthma and objectively monitor treatment response beyond symptom control and lung function.
Collapse
|
223
|
Liu L, Stephens B, Bergman M, May A, Chiang T. Role of Collagen in Airway Mechanics. Bioengineering (Basel) 2021; 8:13. [PMID: 33467161 PMCID: PMC7830870 DOI: 10.3390/bioengineering8010013] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 01/06/2021] [Accepted: 01/09/2021] [Indexed: 12/13/2022] Open
Abstract
Collagen is the most abundant airway extracellular matrix component and is the primary determinant of mechanical airway properties. Abnormal airway collagen deposition is associated with the pathogenesis and progression of airway disease. Thus, understanding how collagen affects healthy airway tissue mechanics is essential. The impact of abnormal collagen deposition and tissue stiffness has been an area of interest in pulmonary diseases such as cystic fibrosis, asthma, and chronic obstructive pulmonary disease. In this review, we discuss (1) the role of collagen in airway mechanics, (2) macro- and micro-scale approaches to quantify airway mechanics, and (3) pathologic changes associated with collagen deposition in airway diseases. These studies provide important insights into the role of collagen in airway mechanics. We summarize their achievements and seek to provide biomechanical clues for targeted therapies and regenerative medicine to treat airway pathology and address airway defects.
Collapse
Affiliation(s)
- Lumei Liu
- Center of Regenerative Medicine, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43215, USA;
| | - Brooke Stephens
- College of Medicine, The Ohio State University, Columbus, OH 43210, USA;
| | - Maxwell Bergman
- Department of Otolaryngology-Head & Neck Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA;
| | - Anne May
- Section of Pulmonary Medicine, Nationwide Children’s Hospital, Columbus, OH 43205, USA;
- Department of Pediatrics, The Ohio State University Wexner Medical Center, Columbus, OH 43205, USA
| | - Tendy Chiang
- Center of Regenerative Medicine, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43215, USA;
- Department of Pediatric Otolaryngology, Nationwide Children’s Hospital, Columbus, OH 43205, USA
| |
Collapse
|
224
|
Bagher M, Rosmark O, Elowsson Rendin L, Nybom A, Wasserstrom S, Müller C, Zhou XH, Dellgren G, Hallgren O, Bjermer L, Larsson-Callerfelt AK, Westergren-Thorsson G. Crosstalk between Mast Cells and Lung Fibroblasts Is Modified by Alveolar Extracellular Matrix and Influences Epithelial Migration. Int J Mol Sci 2021; 22:ijms22020506. [PMID: 33419174 PMCID: PMC7825515 DOI: 10.3390/ijms22020506] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/29/2020] [Accepted: 01/04/2021] [Indexed: 02/06/2023] Open
Abstract
Mast cells play an important role in asthma, however, the interactions between mast cells, fibroblasts and epithelial cells in idiopathic pulmonary fibrosis (IPF) are less known. The objectives were to investigate the effect of mast cells on fibroblast activity and migration of epithelial cells. Lung fibroblasts from IPF patients and healthy individuals were co-cultured with LAD2 mast cells or stimulated with the proteases tryptase and chymase. Human lung fibroblasts and mast cells were cultured on cell culture plastic plates or decellularized human lung tissue (scaffolds) to create a more physiological milieu by providing an alveolar extracellular matrix. Released mediators were analyzed and evaluated for effects on epithelial cell migration. Tryptase increased vascular endothelial growth factor (VEGF) release from fibroblasts, whereas co-culture with mast cells increased IL-6 and hepatocyte growth factor (HGF). Culture in scaffolds increased the release of VEGF compared to culture on plastic. Migration of epithelial cells was reduced by IL-6, while HGF and conditioned media from scaffold cultures promoted migration. In conclusion, mast cells and tryptase increased fibroblast release of mediators that influenced epithelial migration. These data indicate a role of mast cells and tryptase in the interplay between fibroblasts, epithelial cells and the alveolar extracellular matrix in health and lung disease.
Collapse
Affiliation(s)
- Mariam Bagher
- Unit of Lung Biology, Department of Experimental Medical Sciences, Lund University, 221 84 Lund, Sweden; (M.B.); (O.R.); (L.E.R.); (A.N.); (C.M.); (G.W.-T.)
- Department of Respiratory Medicine and Allergology, Skåne University Hospital, Lund University, 221 85 Lund, Sweden; (O.H.); (L.B.)
| | - Oskar Rosmark
- Unit of Lung Biology, Department of Experimental Medical Sciences, Lund University, 221 84 Lund, Sweden; (M.B.); (O.R.); (L.E.R.); (A.N.); (C.M.); (G.W.-T.)
| | - Linda Elowsson Rendin
- Unit of Lung Biology, Department of Experimental Medical Sciences, Lund University, 221 84 Lund, Sweden; (M.B.); (O.R.); (L.E.R.); (A.N.); (C.M.); (G.W.-T.)
| | - Annika Nybom
- Unit of Lung Biology, Department of Experimental Medical Sciences, Lund University, 221 84 Lund, Sweden; (M.B.); (O.R.); (L.E.R.); (A.N.); (C.M.); (G.W.-T.)
| | | | - Catharina Müller
- Unit of Lung Biology, Department of Experimental Medical Sciences, Lund University, 221 84 Lund, Sweden; (M.B.); (O.R.); (L.E.R.); (A.N.); (C.M.); (G.W.-T.)
| | - Xiao-Hong Zhou
- Bioscience Department, Respiratory, Inflammation and Autoimmunity, IMED Biotech Unit, AstraZeneca, 431 53 Mölndal, Sweden;
| | - Göran Dellgren
- Department of Cardiothoracic Surgery and Transplant Institute, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden;
| | - Oskar Hallgren
- Department of Respiratory Medicine and Allergology, Skåne University Hospital, Lund University, 221 85 Lund, Sweden; (O.H.); (L.B.)
| | - Leif Bjermer
- Department of Respiratory Medicine and Allergology, Skåne University Hospital, Lund University, 221 85 Lund, Sweden; (O.H.); (L.B.)
| | - Anna-Karin Larsson-Callerfelt
- Unit of Lung Biology, Department of Experimental Medical Sciences, Lund University, 221 84 Lund, Sweden; (M.B.); (O.R.); (L.E.R.); (A.N.); (C.M.); (G.W.-T.)
- Correspondence: ; Tel.: +46-46-222-8580 or +46-733-525420
| | - Gunilla Westergren-Thorsson
- Unit of Lung Biology, Department of Experimental Medical Sciences, Lund University, 221 84 Lund, Sweden; (M.B.); (O.R.); (L.E.R.); (A.N.); (C.M.); (G.W.-T.)
| |
Collapse
|
225
|
Mezu-Ndubuisi OJ, Maheshwari A. The role of integrins in inflammation and angiogenesis. Pediatr Res 2021; 89:1619-1626. [PMID: 33027803 PMCID: PMC8249239 DOI: 10.1038/s41390-020-01177-9] [Citation(s) in RCA: 171] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/18/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023]
Abstract
Integrins are heterodimeric transmembrane cell adhesion molecules made up of alpha (α) and beta (β) subunits arranged in numerous dimeric pairings. These complexes have varying affinities to extracellular ligands. Integrins regulate cellular growth, proliferation, migration, signaling, and cytokine activation and release and thereby play important roles in cell proliferation and migration, apoptosis, tissue repair, as well as in all processes critical to inflammation, infection, and angiogenesis. This review presents current evidence from human and animal studies on integrin structure and molecular signaling, with particular emphasis on signal transduction in infants. We have included evidence from our own laboratory studies and from an extensive literature search in databases PubMed, EMBASE, Scopus, and the electronic archives of abstracts presented at the annual meetings of the Pediatric Academic Societies. To avoid bias in identification of existing studies, key words were short-listed prior to the actual search both from anecdotal experience and from PubMed's Medical Subject Heading (MeSH) thesaurus. IMPACT: Integrins are a family of ubiquitous αβ heterodimeric receptors that interact with numerous ligands in physiology and disease. Integrins play a key role in cell proliferation, tissue repair, inflammation, infection, and angiogenesis. This review summarizes current evidence from human and animal studies on integrin structure and molecular signaling and promising role in diseases of inflammation, infection, and angiogenesis in infants. This review shows that integrin receptors and ligands are novel therapeutic targets of clinical interest and hold promise as novel therapeutic targets in the management of several neonatal diseases.
Collapse
Affiliation(s)
- Olachi J. Mezu-Ndubuisi
- grid.14003.360000 0001 2167 3675Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI USA
| | - Akhil Maheshwari
- grid.21107.350000 0001 2171 9311Department of Pediatrics, Johns Hopkins University, Baltimore, MD USA
| |
Collapse
|
226
|
Jandl K, Kwapiszewska G. Stiffness of the Extracellular Matrix: A Regulator of Prostaglandins in Pulmonary Fibrosis? Am J Respir Cell Mol Biol 2020; 63:721-722. [PMID: 32946267 PMCID: PMC7790137 DOI: 10.1165/rcmb.2020-0398ed] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- Katharina Jandl
- Ludwig Boltzmann Institute for Lung Vascular Research Graz, Austria.,Otto Loewi Research Center, Division of Pharmacology Medical University of Graz Graz, Austria and
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular Research Graz, Austria.,Otto Loewi Research Center, Division of Physiology Medical University of Graz Graz, Austria
| |
Collapse
|
227
|
Schmidt AF, Kannan PS, Bridges J, Presicce P, Jackson CM, Miller LA, Kallapur SG, Chougnet CA, Jobe AH. Prenatal inflammation enhances antenatal corticosteroid-induced fetal lung maturation. JCI Insight 2020; 5:139452. [PMID: 33328385 PMCID: PMC7819743 DOI: 10.1172/jci.insight.139452] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 11/11/2020] [Indexed: 11/17/2022] Open
Abstract
Respiratory complicˆations are the major cause of morbidity and mortality among preterm infants, which is partially prevented by the administration of antenatal corticosteroids (ACS). Most very preterm infants are exposed to chorioamnionitis, but short- and long-term effects of ACS treatment in this setting are not well defined. In low-resource settings, ACS increased neonatal mortality by perhaps increasing infection. We report that treatment with low-dose ACS in the setting of inflammation induced by intraamniotic lipopolysaccharide (LPS) in rhesus macaques improves lung compliance and increases surfactant production relative to either exposure alone. RNA sequencing shows that these changes are mediated by suppression of proliferation and induction of mesenchymal cellular death via TP53. The combined exposure results in a mature-like transcriptomic profile with inhibition of extracellular matrix development by suppression of collagen genes COL1A1, COL1A2, and COL3A1 and regulators of lung development FGF9 and FGF10. ACS and inflammation also suppressed signature genes associated with proliferative mesenchymal progenitors similar to the term gestation lung. Treatment with ACS in the setting of inflammation may result in early respiratory advantage to preterm infants, but this advantage may come at a risk of abnormal extracellular matrix development, which may be associated with increased risk of chronic lung disease.
Collapse
Affiliation(s)
- Augusto F. Schmidt
- Division of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Paranthaman S. Kannan
- Division of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - James Bridges
- Division of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Pietro Presicce
- Division of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Courtney M. Jackson
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Lisa A. Miller
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, UCD, Davis, California, USA
| | - Suhas G. Kallapur
- Division of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Claire A. Chougnet
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
| | - Alan H. Jobe
- Division of Neonatology and Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
228
|
Erben A, Hörning M, Hartmann B, Becke T, Eisler SA, Southan A, Cranz S, Hayden O, Kneidinger N, Königshoff M, Lindner M, Tovar GEM, Burgstaller G, Clausen‐Schaumann H, Sudhop S, Heymann M. Precision 3D-Printed Cell Scaffolds Mimicking Native Tissue Composition and Mechanics. Adv Healthc Mater 2020; 9:e2000918. [PMID: 33025765 DOI: 10.1002/adhm.202000918] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 08/29/2020] [Indexed: 12/20/2022]
Abstract
Cellular dynamics are modeled by the 3D architecture and mechanics of the extracellular matrix (ECM) and vice versa. These bidirectional cell-ECM interactions are the basis for all vital tissues, many of which have been investigated in 2D environments over the last decades. Experimental approaches to mimic in vivo cell niches in 3D with the highest biological conformity and resolution can enable new insights into these cell-ECM interactions including proliferation, differentiation, migration, and invasion assays. Here, two-photon stereolithography is adopted to print up to mm-sized high-precision 3D cell scaffolds at micrometer resolution with defined mechanical properties from protein-based resins, such as bovine serum albumin or gelatin methacryloyl. By modifying the manufacturing process including two-pass printing or post-print crosslinking, high precision scaffolds with varying Young's moduli ranging from 7-300 kPa are printed and quantified through atomic force microscopy. The impact of varying scaffold topographies on the dynamics of colonizing cells is observed using mouse myoblast cells and a 3D-lung microtissue replica colonized with primary human lung fibroblast. This approach will allow for a systematic investigation of single-cell and tissue dynamics in response to defined mechanical and bio-molecular cues and is ultimately scalable to full organs.
Collapse
Affiliation(s)
- Amelie Erben
- Center for Applied Tissue Engineering and Regenerative Medicine Munich University of Applied Sciences Lothstr. 34 Munich 80533 Germany
- Heinz‐Nixdorf‐Chair of Biomedical Electronics, TranslaTUM, Campus Klinikum rechts der Isar Technical University of Munich Einsteinstraße 25 Munich 81675 Germany
- Center for NanoScience (CeNS) Ludwig‐Maximilians‐University Geschwister‐Scholl Platz 1 Munich 80539 Germany
| | - Marcel Hörning
- Institute of Biomaterials and Biomolecular Systems University of Stuttgart Pfaffenwaldring 57 Stuttgart 70569 Germany
| | - Bastian Hartmann
- Center for Applied Tissue Engineering and Regenerative Medicine Munich University of Applied Sciences Lothstr. 34 Munich 80533 Germany
- Center for NanoScience (CeNS) Ludwig‐Maximilians‐University Geschwister‐Scholl Platz 1 Munich 80539 Germany
| | - Tanja Becke
- Center for Applied Tissue Engineering and Regenerative Medicine Munich University of Applied Sciences Lothstr. 34 Munich 80533 Germany
- Center for NanoScience (CeNS) Ludwig‐Maximilians‐University Geschwister‐Scholl Platz 1 Munich 80539 Germany
| | - Stephan A. Eisler
- Stuttgart Research Center Systems Biology University of Stuttgart Nobelstr. 15 Stuttgart 70569 Germany
| | - Alexander Southan
- Institute of Interfacial Process Engineering and Plasma Technology IGVP University of Stuttgart Nobelstr. 12 Stuttgart 70569 Germany
| | - Séverine Cranz
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC‐M bioArchive, Helmholtz Zentrum München Member of the German Center for Lung Research (DZL) Max‐Lebsche‐Platz 31 Munich 81377 Germany
- Research Unit Lung Repair and Regeneration Helmholtz Zentrum München Max‐Lebsche‐Platz 31 Munich 81377 Germany
| | - Oliver Hayden
- Heinz‐Nixdorf‐Chair of Biomedical Electronics, TranslaTUM, Campus Klinikum rechts der Isar Technical University of Munich Einsteinstraße 25 Munich 81675 Germany
| | - Nikolaus Kneidinger
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC‐M bioArchive, Helmholtz Zentrum München Member of the German Center for Lung Research (DZL) Max‐Lebsche‐Platz 31 Munich 81377 Germany
- Department of Internal Medicine V Ludwig‐Maximillians‐University Munich Marchioninistr. 15 Munich 81377 Germany
| | - Melanie Königshoff
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC‐M bioArchive, Helmholtz Zentrum München Member of the German Center for Lung Research (DZL) Max‐Lebsche‐Platz 31 Munich 81377 Germany
- Research Unit Lung Repair and Regeneration Helmholtz Zentrum München Max‐Lebsche‐Platz 31 Munich 81377 Germany
- University of Colorado Department of Pulmonary Sciences and Critical Care Medicine 13001 E. 17th Pl. Aurora CO 80045 USA
| | - Michael Lindner
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC‐M bioArchive, Helmholtz Zentrum München Member of the German Center for Lung Research (DZL) Max‐Lebsche‐Platz 31 Munich 81377 Germany
- University Department of Visceral and Thoracic Surgery Salzburg Paracelsus Medical University Müllner Hauptstraße 48 Salzburg A‐5020 Austria
| | - Günter E. M. Tovar
- Institute of Interfacial Process Engineering and Plasma Technology IGVP University of Stuttgart Nobelstr. 12 Stuttgart 70569 Germany
| | - Gerald Burgstaller
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC‐M bioArchive, Helmholtz Zentrum München Member of the German Center for Lung Research (DZL) Max‐Lebsche‐Platz 31 Munich 81377 Germany
- Institute of Lung Biology and Disease (ILBD) Helmholtz Zentrum München Max‐Lebsche‐Platz 31 Munich 81377 Germany
| | - Hauke Clausen‐Schaumann
- Center for Applied Tissue Engineering and Regenerative Medicine Munich University of Applied Sciences Lothstr. 34 Munich 80533 Germany
- Center for NanoScience (CeNS) Ludwig‐Maximilians‐University Geschwister‐Scholl Platz 1 Munich 80539 Germany
| | - Stefanie Sudhop
- Center for Applied Tissue Engineering and Regenerative Medicine Munich University of Applied Sciences Lothstr. 34 Munich 80533 Germany
- Center for NanoScience (CeNS) Ludwig‐Maximilians‐University Geschwister‐Scholl Platz 1 Munich 80539 Germany
| | - Michael Heymann
- Center for NanoScience (CeNS) Ludwig‐Maximilians‐University Geschwister‐Scholl Platz 1 Munich 80539 Germany
- Institute of Biomaterials and Biomolecular Systems University of Stuttgart Pfaffenwaldring 57 Stuttgart 70569 Germany
- Department of Cellular and Molecular Biophysics MPI of Biochemistry Martinsried Am Klopferspitz 18 Planegg 82152 Germany
| |
Collapse
|
229
|
Marino GE, Weeraratna AT. A glitch in the matrix: Age-dependent changes in the extracellular matrix facilitate common sites of metastasis. AGING AND CANCER 2020; 1:19-29. [PMID: 35694033 PMCID: PMC9187055 DOI: 10.1002/aac2.12013] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 09/07/2020] [Indexed: 12/15/2022]
Abstract
People over 55 years old represent the majority of cancer patients and suffer from increased metastatic burden compared to the younger patient population. As the aging population increases globally, it is prudent to understand how the intrinsic aging process contributes to cancer progression. As we age, we incur aberrant changes in the extracellular matrix (ECM) of our organs, which contribute to numerous pathologies, including cancer. Notably, the lung, liver, and bone represent the most common sites of distal metastasis for all cancer types. In this review, we describe how age-dependent changes in the ECM of these organs influence cancer progression. Further, we outline how these alterations prime the premetastatic niche and why these may help explain the disparity in outcome for older cancer patients.
Collapse
Affiliation(s)
- Gloria E. Marino
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Ashani T. Weeraratna
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
- Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
230
|
Karakioulaki M, Papakonstantinou E, Stolz D. Extracellular matrix remodelling in COPD. Eur Respir Rev 2020; 29:29/158/190124. [PMID: 33208482 DOI: 10.1183/16000617.0124-2019] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 05/16/2020] [Indexed: 12/30/2022] Open
Abstract
The extracellular matrix (ECM) of the lung plays several important roles in lung function, as it offers a low resistant pathway that allows the exchange of gases, provides compressive strength and elasticity that supports the fragile alveolar-capillary intersection, controls the binding of cells with growth factors and cell surface receptors and acts as a buffer against retention of water.COPD is a chronic inflammatory respiratory condition, characterised by various conditions that result in progressive airflow limitation. At any stage in the course of the disease, acute exacerbations of COPD may occur and lead to accelerated deterioration of pulmonary function. A key factor of COPD is airway remodelling, which refers to the serious alterations of the ECM affecting airway wall thickness, resistance and elasticity. Various studies have shown that serum biomarkers of ECM turnover are significantly associated with disease severity in patients with COPD and may serve as potential targets to control airway inflammation and remodelling in COPD. Unravelling the complete molecular composition of the ECM in the diseased lungs will help to identify novel biomarkers for disease progression and therapy.
Collapse
Affiliation(s)
- Meropi Karakioulaki
- Clinic of Pulmonary Medicine and Respiratory Cell Research, University Hospital, Basel, Switzerland
| | - Eleni Papakonstantinou
- Clinic of Pulmonary Medicine and Respiratory Cell Research, University Hospital, Basel, Switzerland.,Dept of Pharmacology, Faculty of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Daiana Stolz
- Clinic of Pulmonary Medicine and Respiratory Cell Research, University Hospital, Basel, Switzerland
| |
Collapse
|
231
|
Interleukin-1β Modulation of the Mechanobiology of Primary Human Pulmonary Fibroblasts: Potential Implications in Lung Repair. Int J Mol Sci 2020; 21:ijms21228417. [PMID: 33182538 PMCID: PMC7696791 DOI: 10.3390/ijms21228417] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/05/2020] [Accepted: 11/08/2020] [Indexed: 12/16/2022] Open
Abstract
Pro-inflammatory cytokines like interleukin-1β (IL-1β) are upregulated during early responses to tissue damage and are expected to transiently compromise the mechanical microenvironment. Fibroblasts are key regulators of tissue mechanics in the lungs and other organs. However, the effects of IL-1β on fibroblast mechanics and functions remain unclear. Here we treated human pulmonary fibroblasts from control donors with IL-1β and used Atomic Force Microscopy to unveil that IL-1β significantly reduces the stiffness of fibroblasts concomitantly with a downregulation of filamentous actin (F-actin) and alpha-smooth muscle (α-SMA). Likewise, COL1A1 mRNA was reduced, whereas that of collagenases MMP1 and MMP2 were upregulated, favoring a reduction of type-I collagen. These mechanobiology changes were functionally associated with reduced proliferation and enhanced migration upon IL-1β stimulation, which could facilitate lung repair by drawing fibroblasts to sites of tissue damage. Our observations reveal that IL-1β may reduce local tissue rigidity by acting both intracellularly and extracellularly through the downregulation of fibroblast contractility and type I collagen deposition, respectively. These IL-1β-dependent mechanical effects may enhance lung repair further by locally increasing pulmonary tissue compliance to preserve normal lung distension and function. Moreover, our results support that IL-1β provides innate anti-fibrotic protection that may be relevant during the early stages of lung repair.
Collapse
|
232
|
Gardelli C, Russo L, Cipolla L, Moro M, Andriani F, Rondinone O, Nicotra F, Sozzi G, Bertolini G, Roz L. Differential glycosylation of collagen modulates lung cancer stem cell subsets through β1 integrin-mediated interactions. Cancer Sci 2020; 112:217-230. [PMID: 33068069 PMCID: PMC7780011 DOI: 10.1111/cas.14700] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/11/2020] [Accepted: 10/12/2020] [Indexed: 12/16/2022] Open
Abstract
In lung cancer, CD133+ cells represent the subset of cancer stem cells (CSC) able to sustain tumor growth and metastatic dissemination. CSC function is tightly regulated by specialized niches composed of both stromal cells and extracellular matrix (ECM) proteins, mainly represented by collagen. The relevance of collagen glycosylation, a fundamental post-translational modification controlling several biological processes, in regulating tumor cell phenotype remains, however, largely unexplored. To investigate the bioactive effects of differential ECM glycosylation on lung cancer cells, we prepared collagen films functionalized with glucose (Glc-collagen) and galactose (Gal-collagen) exploiting a neoglycosylation approach based on a reductive amination of maltose and lactose with the amino residues of collagen lysines. We demonstrate that culturing of tumor cells on collagen determines a glycosylation-dependent positive selection of CSC and triggers their expansion/generation. The functional relevance of CD133+ CSC increase was validated in vivo, proving an augmented tumorigenic and metastatic potential. High expression of integrin β1 in its active form is associated with an increased proficiency of tumor cells to sense signaling from glycosylated matrices (glyco-collagen) and to acquire stemness features. Accordingly, inhibition of integrin β1 in tumor cells prevents CSC enrichment, suggesting that binding of integrin β1 to Glc-collagen subtends CSC expansion/generation. We provide evidence suggesting that collagen glycosylation could play an essential role in modulating the creation of a niche favorable for the generation and selection/survival of lung CSC. Interfering with this crosstalk may represent an innovative therapeutic strategy for lung cancer treatment.
Collapse
Affiliation(s)
- Cecilia Gardelli
- Tumor Genomics Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Laura Russo
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Laura Cipolla
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Massimo Moro
- Tumor Genomics Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Francesca Andriani
- Tumor Genomics Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Ornella Rondinone
- Tumor Genomics Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Francesco Nicotra
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Gabriella Sozzi
- Tumor Genomics Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giulia Bertolini
- Tumor Genomics Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Luca Roz
- Tumor Genomics Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
233
|
Blokland K, Pouwels S, Schuliga M, Knight D, Burgess J. Regulation of cellular senescence by extracellular matrix during chronic fibrotic diseases. Clin Sci (Lond) 2020; 134:2681-2706. [PMID: 33084883 PMCID: PMC7578566 DOI: 10.1042/cs20190893] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 02/07/2023]
Abstract
The extracellular matrix (ECM) is a complex network of macromolecules surrounding cells providing structural support and stability to tissues. The understanding of the ECM and the diverse roles it plays in development, homoeostasis and injury have greatly advanced in the last three decades. The ECM is crucial for maintaining tissue homoeostasis but also many pathological conditions arise from aberrant matrix remodelling during ageing. Ageing is characterised as functional decline of tissue over time ultimately leading to tissue dysfunction, and is a risk factor in many diseases including cardiovascular disease, diabetes, cancer, dementia, glaucoma, chronic obstructive pulmonary disease (COPD) and fibrosis. ECM changes are recognised as a major driver of aberrant cell responses. Mesenchymal cells in aged tissue show signs of growth arrest and resistance to apoptosis, which are indicative of cellular senescence. It was recently postulated that cellular senescence contributes to the pathogenesis of chronic fibrotic diseases in the heart, kidney, liver and lung. Senescent cells negatively impact tissue regeneration while creating a pro-inflammatory environment as part of the senescence-associated secretory phenotype (SASP) favouring disease progression. In this review, we explore and summarise the current knowledge around how aberrant ECM potentially influences the senescent phenotype in chronic fibrotic diseases. Lastly, we will explore the possibility for interventions in the ECM-senescence regulatory pathways for therapeutic potential in chronic fibrotic diseases.
Collapse
Affiliation(s)
- Kaj E.C. Blokland
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
- University of Newcastle, School of Biomedical Sciences and Pharmacy, Callaghan, NSW, Australia
- National Health and Medical Research Council Centre of Research Excellence in Pulmonary Fibrosis, Sydney, NSW, Australia
| | - Simon D. Pouwels
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
- Department of Lung Diseases, University Medical Center Groningen, Groningen, The Netherlands
| | - Michael Schuliga
- University of Newcastle, School of Biomedical Sciences and Pharmacy, Callaghan, NSW, Australia
| | - Darryl A. Knight
- University of Newcastle, School of Biomedical Sciences and Pharmacy, Callaghan, NSW, Australia
- National Health and Medical Research Council Centre of Research Excellence in Pulmonary Fibrosis, Sydney, NSW, Australia
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Providence Health Care Research Institute, Vancouver, BC, Canada
| | - Janette K. Burgess
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
| |
Collapse
|
234
|
Lung gene expression signatures suggest pathogenic links and molecular markers for pulmonary tuberculosis, adenocarcinoma and sarcoidosis. Commun Biol 2020; 3:604. [PMID: 33097805 PMCID: PMC7584606 DOI: 10.1038/s42003-020-01318-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 09/25/2020] [Indexed: 12/22/2022] Open
Abstract
Previous reports have suggested a link between pulmonary tuberculosis (TB), which is caused by Mycobacterium tuberculosis (Mtb), and the development of lung adenocarcinoma (LUAD) and sarcoidosis. Furthermore, these lung diseases share certain clinical similarities that can challenge differential diagnosis in some cases. Here, through comparison of lung transcriptome-derived molecular signatures of TB, LUAD and sarcoidosis patients, we identify certain shared disease-related expression patterns. We also demonstrate that MKI67, an over-expressed gene shared by TB and LUAD, is a key mediator in Mtb-promoted tumor cell proliferation, migration, and invasion. Moreover, we reveal a distinct ossification-related TB lung signature, which may be associated with the activation of the BMP/SMAD/RUNX2 pathway in Mtb-infected macrophages that can restrain mycobacterial survival and promote osteogenic differentiation of mesenchymal stem cells. Taken together, these findings provide novel pathogenic links and potential molecular markers for better understanding and differential diagnosis of pulmonary TB, LUAD and sarcoidosis. Previous work has suggested potential links between Mycobacterium tuberculosis infection and the development of both lung cancer and sarcoidosis, in addition to tuberculosis. Here, Qiyao Chai, Zhe Lu, Zhidong Liu and colleagues report a transcriptomic analysis of lung tissue from tuberculosis, lung adenocarcinoma, and sarcoidosis patients and find that while many disease-linked expression changes are shared between the three diseases, each also has distinct transcriptional signatures that could be useful as molecular markers.
Collapse
|
235
|
Sattari S, Mariano CA, Vittalbabu S, Velazquez JV, Postma J, Horst C, Teh E, Nordgren TM, Eskandari M. Introducing a Custom-Designed Volume-Pressure Machine for Novel Measurements of Whole Lung Organ Viscoelasticity and Direct Comparisons Between Positive- and Negative-Pressure Ventilation. Front Bioeng Biotechnol 2020; 8:578762. [PMID: 33195138 PMCID: PMC7643401 DOI: 10.3389/fbioe.2020.578762] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/17/2020] [Indexed: 12/13/2022] Open
Abstract
Asthma, emphysema, COVID-19 and other lung-impacting diseases cause the remodeling of tissue structural properties and can lead to changes in conducting pulmonary volume, viscoelasticity, and air flow distribution. Whole organ experimental inflation tests are commonly used to understand the impact of these modifications on lung mechanics. Here we introduce a novel, automated, custom-designed device for measuring the volume and pressure response of lungs, surpassing the capabilities of traditional machines and built to range size-scales to accommodate both murine and porcine tests. The software-controlled system is capable of constructing standardized continuous volume-pressure curves, while accounting for air compressibility, yielding consistent and reproducible measures while eliminating the need for pulmonary degassing. This device uses volume-control to enable viscoelastic whole lung macromechanical insights from rate dependencies and pressure-time curves. Moreover, the conceptual design of this device facilitates studies relating the phenomenon of diaphragm breathing and artificial ventilation induced by pushing air inside the lungs. System capabilities are demonstrated and validated via a comparative study between ex vivo murine lungs and elastic balloons, using various testing protocols. Volume-pressure curve comparisons with previous pressure-controlled systems yield good agreement, confirming accuracy. This work expands the capabilities of current lung experiments, improving scientific investigations of healthy and diseased pulmonary biomechanics. Ultimately, the methodologies demonstrated in the manufacturing of this system enable future studies centered on investigating viscoelasticity as a potential biomarker and improvements to patient ventilators based on direct assessment and comparisons of positive- and negative-pressure mechanics.
Collapse
Affiliation(s)
- Samaneh Sattari
- Department of Mechanical Engineering, University of California, Riverside, Riverside, CA, United States
| | - Crystal A Mariano
- Department of Mechanical Engineering, University of California, Riverside, Riverside, CA, United States
| | - Swathi Vittalbabu
- Department of Mechanical Engineering, University of California, Riverside, Riverside, CA, United States
| | - Jalene V Velazquez
- BREATHE Center at the School of Medicine, University of California, Riverside, Riverside, CA, United States.,Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | | | - Caleb Horst
- CellScale Biomaterials Testing, Waterloo, ON, Canada
| | - Eric Teh
- CellScale Biomaterials Testing, Waterloo, ON, Canada
| | - Tara M Nordgren
- BREATHE Center at the School of Medicine, University of California, Riverside, Riverside, CA, United States.,Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| | - Mona Eskandari
- Department of Mechanical Engineering, University of California, Riverside, Riverside, CA, United States.,BREATHE Center at the School of Medicine, University of California, Riverside, Riverside, CA, United States.,Department of Bioengineering, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
236
|
Freeberg MAT, Perelas A, Rebman JK, Phipps RP, Thatcher TH, Sime PJ. Mechanical Feed-Forward Loops Contribute to Idiopathic Pulmonary Fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 191:18-25. [PMID: 33031756 PMCID: PMC7768346 DOI: 10.1016/j.ajpath.2020.09.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/25/2020] [Accepted: 09/08/2020] [Indexed: 12/11/2022]
Abstract
Idiopathic pulmonary fibrosis is a progressive scarring disease characterized by extracellular matrix accumulation and altered mechanical properties of lung tissue. Recent studies support the hypothesis that these compositional and mechanical changes create a progressive feed-forward loop in which enhanced matrix deposition and tissue stiffening contribute to fibroblast and myofibroblast differentiation and activation, which further perpetuates matrix production and stiffening. The biomechanical properties of tissues are sensed and responded to by mechanotransduction pathways that facilitate sensing of changes in mechanical cues by tissue resident cells and convert the mechanical signals into downstream biochemical signals. Although our understanding of mechanotransduction pathways associated with pulmonary fibrosis remains incomplete, recent progress has allowed us to begin to elucidate the specific mechanisms supporting fibrotic feed-forward loops. The mechanosensors discussed here include integrins, Piezo channels, transient receptor potential channels, and nonselective ion channels. Also discussed are downstream transcription factors, including myocardin-related transcription factor and Yes-associated protein/transcriptional coactivator with PDZ-binding motif. This review describes mechanosensors and mechanotransduction pathways associated with fibrosis progression and highlights promising therapeutic insights.
Collapse
Affiliation(s)
- Margaret A T Freeberg
- Division of Pulmonary Disease and Critical Care Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Apostolos Perelas
- Division of Pulmonary Disease and Critical Care Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Jane K Rebman
- Division of Pulmonary Disease and Critical Care Medicine, Virginia Commonwealth University, Richmond, Virginia
| | | | - Thomas H Thatcher
- Division of Pulmonary Disease and Critical Care Medicine, Virginia Commonwealth University, Richmond, Virginia
| | - Patricia J Sime
- Division of Pulmonary Disease and Critical Care Medicine, Virginia Commonwealth University, Richmond, Virginia.
| |
Collapse
|
237
|
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
|
238
|
Gouarderes S, Mingotaud AF, Vicendo P, Gibot L. Vascular and extracellular matrix remodeling by physical approaches to improve drug delivery at the tumor site. Expert Opin Drug Deliv 2020; 17:1703-1726. [PMID: 32838565 DOI: 10.1080/17425247.2020.1814735] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Modern comprehensive studies of tumor microenvironment changes allowed scientists to develop new and more efficient strategies that will improve anticancer drug delivery on site. The tumor microenvironment, especially the dense extracellular matrix, has a recognized capability to hamper the penetration of conventional drugs. Development and co-applications of strategies aiming at remodeling the tumor microenvironment are highly demanded to improve drug delivery at the tumor site in a therapeutic prospect. AREAS COVERED Increasing indications suggest that classical physical approaches such as exposure to ionizing radiations, hyperthermia or light irradiation, and emerging ones as sonoporation, electric field or cold plasma technology can be applied as standalone or associated strategies to remodel the tumor microenvironment. The impacts on vasculature and extracellular matrix remodeling of these physical approaches will be discussed with the goal to improve nanotherapeutics delivery at the tumor site. EXPERT OPINION Physical approaches to modulate vascular properties and remodel the extracellular matrix are of particular interest to locally control and improve drug delivery and thus increase its therapeutic index. They are particularly powerful as adjuvant to nanomedicine delivery; the development of these technologies could have extremely widespread implications for cancer treatment.[Figure: see text].
Collapse
Affiliation(s)
- Sara Gouarderes
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III - Paul Sabatier , Toulouse, France
| | - Anne-Françoise Mingotaud
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III - Paul Sabatier , Toulouse, France
| | - Patricia Vicendo
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III - Paul Sabatier , Toulouse, France
| | - Laure Gibot
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III - Paul Sabatier , Toulouse, France
| |
Collapse
|
239
|
Virumbrales-Muñoz M, Ayuso JM, Gong MM, Humayun M, Livingston MK, Lugo-Cintrón KM, McMinn P, Álvarez-García YR, Beebe DJ. Microfluidic lumen-based systems for advancing tubular organ modeling. Chem Soc Rev 2020; 49:6402-6442. [PMID: 32760967 PMCID: PMC7521761 DOI: 10.1039/d0cs00705f] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Microfluidic lumen-based systems are microscale models that recapitulate the anatomy and physiology of tubular organs. These technologies can mimic human pathophysiology and predict drug response, having profound implications for drug discovery and development. Herein, we review progress in the development of microfluidic lumen-based models from the 2000s to the present. The core of the review discusses models for mimicking blood vessels, the respiratory tract, the gastrointestinal tract, renal tubules, and liver sinusoids, and their application to modeling organ-specific diseases. We also highlight emerging application areas, such as the lymphatic system, and close the review discussing potential future directions.
Collapse
Affiliation(s)
- María Virumbrales-Muñoz
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - José M Ayuso
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA and Morgridge Institute for Research, Madison, WI, USA
| | - Max M Gong
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA and Department of Biomedical Engineering, Trine University, Angola, IN, USA
| | - Mouhita Humayun
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Megan K Livingston
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Karina M Lugo-Cintrón
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Patrick McMinn
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - Yasmín R Álvarez-García
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - David J Beebe
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA. and University of Wisconsin Carbone Cancer Center, Madison, WI, USA and Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
240
|
Fane M, Weeraratna AT. Normal Aging and Its Role in Cancer Metastasis. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a037341. [PMID: 31615864 DOI: 10.1101/cshperspect.a037341] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Metastasis is the most common cause of death, with treatments failing to provide a durable response. Aging is a key prognostic factor in many cancers. Emerging data suggest that normal age-related changes in the tumor microenvironment can contribute to metastatic progression. These changes encompass secreted factors, biophysical changes, and changes in both stromal and immune cell populations. These data also highlight the importance of conducting studies in preclinical models of appropriate age. Ultimately, therapies may also need to be tailored to reflect patient age, as markers of metastatic disease differ in young and aged populations. In this review, we will discuss some of the changes that occur during aging that increase the metastatic capacity of tumor cells.
Collapse
Affiliation(s)
- Mitchell Fane
- The Wistar Institute, Immunology, Microenvironment and Metastasis Program, Philadelphia, Pennsylvania 19104, USA.,Department of Biochemistry and Molecular Biology, Johns Hopkins School of Public Health, Baltimore, Maryland 21205, USA
| | - Ashani T Weeraratna
- The Wistar Institute, Immunology, Microenvironment and Metastasis Program, Philadelphia, Pennsylvania 19104, USA.,Department of Biochemistry and Molecular Biology, Johns Hopkins School of Public Health, Baltimore, Maryland 21205, USA.,Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, USA
| |
Collapse
|
241
|
Barenholz-Cohen T, Merkher Y, Haj J, Shechter D, Kirchmeier D, Shaked Y, Weihs D. Lung mechanics modifications facilitating metastasis are mediated in part by breast cancer-derived extracellular vesicles. Int J Cancer 2020; 147:2924-2933. [PMID: 32700789 DOI: 10.1002/ijc.33229] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 07/10/2020] [Accepted: 07/14/2020] [Indexed: 12/20/2022]
Abstract
Tumor microenvironment-mechanics greatly affect tumor-cell characteristics such as invasion and proliferation. We and others have previously shown that after chemotherapy, tumor cells shed more extracellular vesicles (EVs), leading to tumor growth and even spread, via angiogenesis and the mobilization of specific bone-marrow-derived cells contributing to metastasis. However, physical, mechanobiological and mechanostructural changes at premetastatic sites that may support tumor cell seeding, have yet to be determined. Here, we collected tumor-derived extracellular vesicles (tEV) from breast carcinoma cells exposed to paclitaxel chemotherapy, and tested their effects on tissue mechanics (eg, elasticity and stiffness) of likely metastatic organs in cancer-free mice, using shear rheometry. Cancer-free mice were injected with saline or with tEVs from untreated cells and lung tissue demonstrated widely variable, viscoelastic mechanics, being more elastic than viscous. Contrastingly, tEVs from chemotherapy-exposed cells induced more uniform, viscoelastic lung mechanics, with lower stiffness and viscosity; interestingly, livers were significantly stiffer than both controls. We observe statistically significant differences in softening of lung samples from all three groups under increasing strain-amplitudes and in their stiffening under increasing strain-frequencies; the groups reach similar values at high strain amplitudes and frequencies, indicating local changes in tissue microstructure. Evaluation of genes associated with the extracellular matrix and fibronectin protein-expression revealed potential compositional changes underlying the altered mechanics. Thus, we propose that tEVs, even without cancer cells, contribute to metastasis by changing microstructures at distant organs. This is done partially by altering the composition and mechanostructure of tissues to support tumor cell invasion and seeding.
Collapse
Affiliation(s)
- Tamar Barenholz-Cohen
- Faculty of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Yulia Merkher
- Faculty of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Jozafina Haj
- Rappaport Faculty of Medicine, Technion-Integrated Cancer Center, Technion - Israel Institute of Technology, Haifa, Israel
| | - Dvir Shechter
- Rappaport Faculty of Medicine, Technion-Integrated Cancer Center, Technion - Israel Institute of Technology, Haifa, Israel
| | - Daniela Kirchmeier
- Faculty of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel.,Medical and Pharmaceutical Biotechnology, IMC University of Applied Sciences Krems, Krems an der Donau, Austria
| | - Yuval Shaked
- Rappaport Faculty of Medicine, Technion-Integrated Cancer Center, Technion - Israel Institute of Technology, Haifa, Israel
| | - Daphne Weihs
- Faculty of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
242
|
Bhosle VK, Mukherjee T, Huang YW, Patel S, Pang BWF, Liu GY, Glogauer M, Wu JY, Philpott DJ, Grinstein S, Robinson LA. SLIT2/ROBO1-signaling inhibits macropinocytosis by opposing cortical cytoskeletal remodeling. Nat Commun 2020; 11:4112. [PMID: 32807784 PMCID: PMC7431850 DOI: 10.1038/s41467-020-17651-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 07/08/2020] [Indexed: 01/06/2023] Open
Abstract
Macropinocytosis is essential for myeloid cells to survey their environment and for growth of RAS-transformed cancer cells. Several growth factors and inflammatory stimuli are known to induce macropinocytosis, but its endogenous inhibitors have remained elusive. Stimulation of Roundabout receptors by Slit ligands inhibits directional migration of many cell types, including immune cells and cancer cells. We report that SLIT2 inhibits macropinocytosis in vitro and in vivo by inducing cytoskeletal changes in macrophages. In mice, SLIT2 attenuates the uptake of muramyl dipeptide, thereby preventing NOD2-dependent activation of NF-κB and consequent secretion of pro-inflammatory chemokine, CXCL1. Conversely, blocking the action of endogenous SLIT2 enhances CXCL1 secretion. SLIT2 also inhibits macropinocytosis in RAS-transformed cancer cells, thereby decreasing their survival in nutrient-deficient conditions which resemble tumor microenvironment. Our results identify SLIT2 as a physiological inhibitor of macropinocytosis and challenge the conventional notion that signals that enhance macropinocytosis negatively regulate cell migration, and vice versa.
Collapse
Affiliation(s)
- Vikrant K Bhosle
- Program in Cell Biology, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
| | - Tapas Mukherjee
- Department of Immunology, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Yi-Wei Huang
- Program in Cell Biology, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
| | - Sajedabanu Patel
- Program in Cell Biology, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
| | - Bo Wen Frank Pang
- Program in Cell Biology, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
- Institute of Medical Science, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
- BenchSci, Suite 201, 559 College Street, Toronto, ON, M6G 1A9, Canada
| | - Guang-Ying Liu
- Program in Cell Biology, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
| | - Michael Glogauer
- Faculty of Dentistry, University of Toronto, 101 Elm Street, Toronto, ON, M5G 2L3, Canada
- Department of Dental Oncology and Maxillofacial Prosthetics, University Health Network, Princess Margaret Cancer Centre, 610 University Avenue, Toronto, ON, M5G 2C1, Canada
- Centre for Advanced Dental Research and Care, Mount Sinai Hospital, 600 University Avenue, Toronto, ON, M5G 1X5, Canada
| | - Jane Y Wu
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
- Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - Sergio Grinstein
- Program in Cell Biology, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Toronto, ON, M5G 0A4, Canada
- Department of Biochemistry, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
- Keenan Research Centre of the Li Ka Shing Knowledge Institute, St. Michael's Hospital, 290 Victoria Street, Toronto, ON, M5C 1N8, Canada
| | - Lisa A Robinson
- Program in Cell Biology, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, 686 Bay Street, Toronto, ON, M5G 0A4, Canada.
- Institute of Medical Science, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.
- Division of Nephrology, The Hospital for Sick Children, 555 University Avenue, Toronto, ON, M5G 1X8, Canada.
- Department of Paediatrics, Faculty of Medicine, University of Toronto, 555 University Avenue, Toronto, ON, M5G 1X8, Canada.
| |
Collapse
|
243
|
Vieira CP, de Oliveira LP, Da Silva MB, Majolli Andre D, Tavares EBG, Pimentel ER, Antunes E. Role of metalloproteinases and TNF-α in obesity-associated asthma in mice. Life Sci 2020; 259:118191. [PMID: 32777302 DOI: 10.1016/j.lfs.2020.118191] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/24/2020] [Accepted: 07/30/2020] [Indexed: 02/06/2023]
Abstract
Numerous population studies conducted worldwide indicate that the prevalence of asthma is higher in obese versus lean individuals. It has been reported that sensitized lean mice has a better recovery of lung inflammation in asthma. Extracellular matrix (ECM) plays an essential role in the structural support of the lungs regulating the airways diameter, thus preventing its collapse during expiration. ECM renewal by metalloproteinase (MMPs) enzymes is critical for pulmonary biology. There seems to be an imbalance of MMPs activity in asthma and obesity, which can impair the lung remodeling process. In this study, we characterized the pulmonary ECM of obese and lean mice, non-sensitized and sensitized with ovalbumin (OVA). Pharmacological intervention was performed by using anti-TNF-α, and MMP-8 and MMP-9 inhibitors in obese and lean sensitized mice. Activity of MMPs was assessed by gelatinase electrophorese, western blotting and zymogram in situ. Unbalance of MMP-2, MMP-8, MMP-9 and MMP-12 was detected in lung tissue of OVA-sensitized obese mice, which was accompanied by high degradation, corroborating an excessive deposition of types I and III collagen in pulmonary matrix of obese animals. Inhibitions of TNF-α and MMP-9 reduced this MMP imbalance, clearly suggesting a positive effect on pulmonary ECM. Obese and lean mice presented diverse phenotype of asthma regarding the ECM compounds and the inhibition of MMPs pathway could be a good alternative to regulate the activity in ECM lungs of asthmatic obese individuals.
Collapse
Affiliation(s)
- Cristiano Pedrozo Vieira
- Department of Pharmacology, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.
| | - Leticia Prado de Oliveira
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, 13083-863 CP 6109, Campinas, Sao Paulo, Brazil
| | - Mariana Bombardi Da Silva
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, 13083-863 CP 6109, Campinas, Sao Paulo, Brazil
| | - Diana Majolli Andre
- Department of Pharmacology, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Edith Bastos Gandra Tavares
- Department of Pharmacology, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Edson Rosa Pimentel
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, 13083-863 CP 6109, Campinas, Sao Paulo, Brazil
| | - Edson Antunes
- Department of Pharmacology, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| |
Collapse
|
244
|
Fabris L, Cadamuro M, Cagnin S, Strazzabosco M, Gores GJ. Liver Matrix in Benign and Malignant Biliary Tract Disease. Semin Liver Dis 2020; 40:282-297. [PMID: 32162285 DOI: 10.1055/s-0040-1705109] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The extracellular matrix is a highly reactive scaffold formed by a wide array of multifunctional molecules, encompassing collagens and noncollagenous glycoproteins, proteoglycans, glycosaminoglycans, and polysaccharides. Besides outlining the tissue borders, the extracellular matrix profoundly regulates the behavior of resident cells by transducing mechanical signals, and by integrating multiple cues derived from the microenvironment. Evidence is mounting that changes in the biostructure of the extracellular matrix are instrumental for biliary repair. Following biliary damage and eventually, malignant transformation, the extracellular matrix undergoes several quantitative and qualitative modifications, which direct interactions among hepatic progenitor cells, reactive ductular cells, activated myofibroblasts and macrophages, to generate the ductular reaction. Herein, we will give an overview of the main molecular factors contributing to extracellular matrix remodeling in cholangiopathies. Then, we will discuss the structural alterations in terms of biochemical composition and physical stiffness featuring the "desmoplastic matrix" of cholangiocarcinoma along with their pro-oncogenic effects.
Collapse
Affiliation(s)
- Luca Fabris
- Department of Molecular Medicine, University of Padua, Padua, Italy.,Liver Center, Department of Medicine, Yale University, New Haven, Connecticut
| | | | - Silvia Cagnin
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Mario Strazzabosco
- Liver Center, Department of Medicine, Yale University, New Haven, Connecticut
| | - Gregory J Gores
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, Michigan
| |
Collapse
|
245
|
Rønnow SR, Langholm LL, Karsdal MA, Manon-Jensen T, Tal-Singer R, Miller BE, Vestbo J, Leeming DJ, Sand JMB. Endotrophin, an extracellular hormone, in combination with neoepitope markers of von Willebrand factor improves prediction of mortality in the ECLIPSE COPD cohort. Respir Res 2020; 21:202. [PMID: 32731895 PMCID: PMC7393910 DOI: 10.1186/s12931-020-01461-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 07/20/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Lung epithelial damage, activation of the wound healing cascade, and remodeling of the extracellular matrix (ECM) play a major role in chronic obstructive pulmonary disease (COPD). The pro-peptide of type VI collagen has been identified as the hormone endotrophin. Endotrophin has been shown to promote fibrosis and inflammation, whereas von Willebrand factor (VWF) is a crucial part of wound healing initiation. Here, we assessed the released and activated form of VWF and endotrophin, the pro-peptide of type VI collagen, serologically to investigate their association with mortality in COPD subjects alone or in combination. METHODS One thousand COPD patients with 3 years of clinical follow-up from the Evaluation of COPD Longitudinally to Identify Predictive Surrogate Endpoints (ECLIPSE) cohort were included. Serum and heparin plasma were collected at 6 months and 1 year, respectively. Competitive ELISA utilizing specific monoclonal antibodies assessed endotrophin/type VI collagen formation (PRO-C6), VWF release (VWF-N), and activated VWF (VWF-A). Biomarker levels were dichotomized into high and low as defined by receiver operating characteristic (ROC) curves based on mortality data. Kaplan-Meier analysis was used to determine hazard ratios for all-cause mortality for biomarkers alone or in combination. RESULTS High levels of PRO-C6, VWF-A, and VWF-N have previously been shown to be individually associated with a higher risk of mortality with hazard ratios of 5.6 (95% CI 2.4-13.1), 3.7 (1.8-7.6), and 4.6 (2.2-9.6), respectively. The hazard ratios increased when combining the biomarkers: PRO-C6*VWFA 8.8 (2.8-27.7) and PRO-C6*VWFN 13.3 (5.6-32.0). Notably, PRO-C6*VWF-N increased more than 2-fold. CONCLUSION We demonstrated that by combining two pathological relevant aspects of COPD, tissue remodeling, and wound healing, the predictive value of biomarkers for mortality increased notably.
Collapse
Affiliation(s)
| | | | | | | | | | - Bruce E Miller
- R&D Respiratory Therapy Area Unit, GlaxoSmithKline, King Of Prussia, PA, USA
| | - Jørgen Vestbo
- Division of Infection, Immunity and Respiratory Medicine, University of Manchester, Manchester, UK
| | | | | |
Collapse
|
246
|
Uludağ H, Parent K, Aliabadi HM, Haddadi A. Prospects for RNAi Therapy of COVID-19. Front Bioeng Biotechnol 2020; 8:916. [PMID: 32850752 PMCID: PMC7409875 DOI: 10.3389/fbioe.2020.00916] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/15/2020] [Indexed: 12/12/2022] Open
Abstract
COVID-19 caused by the SARS-CoV-2 virus is a fast emerging disease with deadly consequences. The pulmonary system and lungs in particular are most prone to damage caused by the SARS-CoV-2 infection, which leaves a destructive footprint in the lung tissue, making it incapable of conducting its respiratory functions and resulting in severe acute respiratory disease and loss of life. There were no drug treatments or vaccines approved for SARS-CoV-2 at the onset of pandemic, necessitating an urgent need to develop effective therapeutics. To this end, the innate RNA interference (RNAi) mechanism can be employed to develop front line therapies against the virus. This approach allows specific binding and silencing of therapeutic targets by using short interfering RNA (siRNA) and short hairpin RNA (shRNA) molecules. In this review, we lay out the prospect of the RNAi technology for combatting the COVID-19. We first summarize current understanding of SARS-CoV-2 virology and the host response to viral entry and duplication, with the purpose of revealing effective RNAi targets. We then summarize the past experience with nucleic acid silencers for SARS-CoV, the predecessor for current SARS-CoV-2. Efforts targeting specific protein-coding regions within the viral genome and intragenomic targets are summarized. Emphasizing non-viral delivery approaches, molecular underpinnings of design of RNAi agents are summarized with comparative analysis of various systems used in the past. Promising viral targets as well as host factors are summarized, and the possibility of modulating the immune system are presented for more effective therapies. We place special emphasis on the limitations of past studies to propel the field faster by focusing on most relevant models to translate the promising agents to a clinical setting. Given the urgency to address lung failure in COVID-19, we summarize the feasibility of delivering promising therapies by the inhalational route, with the expectation that this route will provide the most effective intervention to halt viral spread. We conclude with the authors' perspectives on the future of RNAi therapeutics for combatting SARS-CoV-2. Since time is of the essence, a strong perspective for the path to most effective therapeutic approaches are clearly articulated by the authors.
Collapse
Affiliation(s)
- Hasan Uludağ
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB, Canada
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Kylie Parent
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB, Canada
| | | | - Azita Haddadi
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
247
|
Deng Z, Fear MW, Suk Choi Y, Wood FM, Allahham A, Mutsaers SE, Prêle CM. The extracellular matrix and mechanotransduction in pulmonary fibrosis. Int J Biochem Cell Biol 2020; 126:105802. [PMID: 32668329 DOI: 10.1016/j.biocel.2020.105802] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/06/2020] [Accepted: 07/10/2020] [Indexed: 12/11/2022]
Abstract
Pulmonary fibrosis is characterised by excessive scarring in the lung which leads to compromised lung function, serious breathing problems and in some diseases, death. It includes several lung disorders with idiopathic pulmonary fibrosis (IPF) the most common and most severe. Pulmonary fibrosis is considered to be perpetuated by aberrant wound healing which leads to fibroblast accumulation, differentiation and activation, and deposition of excessive amounts of extracellular matrix (ECM) components, in particular, collagen. Recent studies have identified the importance of changes in the composition and structure of lung ECM during the development of pulmonary fibrosis and the interaction between ECM and lung cells. There is strong evidence that increased matrix stiffness induces changes in cell function including proliferation, migration, differentiation and activation. Understanding how changes in the ECM microenvironment influence cell behaviour during fibrogenesis, and the mechanisms regulating these changes, will provide insight for developing new treatments.
Collapse
Affiliation(s)
- Zhenjun Deng
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Nedlands, 6009, WA, Australia
| | - Mark W Fear
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Nedlands, 6009, WA, Australia; Institute for Respiratory Health, Nedlands, WA, Australia
| | - Yu Suk Choi
- School of Human Sciences, The University of Western Australia, Crawley, WA, Australia
| | - Fiona M Wood
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Nedlands, 6009, WA, Australia; Burns Service of Western Australia, Perth Children's Hospital, Nedlands, WA, Australia; Fiona Stanley Hospital, Murdoch, WA, Australia
| | - Amira Allahham
- Burn Injury Research Unit, School of Biomedical Sciences, The University of Western Australia, Nedlands, 6009, WA, Australia
| | - Steven E Mutsaers
- Institute for Respiratory Health, Nedlands, WA, Australia; Centre for Respiratory Health, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia; Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| | - Cecilia M Prêle
- Institute for Respiratory Health, Nedlands, WA, Australia; Centre for Respiratory Health, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia; Centre for Cell Therapy and Regenerative Medicine, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia.
| |
Collapse
|
248
|
Hong Y, Liang X, Gilhus NE. AChR antibodies show a complex interaction with human skeletal muscle cells in a transcriptomic study. Sci Rep 2020; 10:11230. [PMID: 32641696 PMCID: PMC7343820 DOI: 10.1038/s41598-020-68185-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 06/17/2020] [Indexed: 01/26/2023] Open
Abstract
Acetylcholine receptor (AChR) antibodies are the most important pathogenic marker in patients with myasthenia gravis (MG). The antibodies bind to AChRs on the postsynaptic membrane, and this leads to receptor degradation, destruction, or functional blocking with impaired signal at the neuromuscular junction. In this study, we have explored the effects of AChR antibodies binding to mature human myotubes with agrin-induced AChR clusters and pathways relevant for AChR degradation using bulk RNA sequencing. Protein-coding RNAs and lncRNAs were examined by RNA sequencing analysis. AChR antibodies induced marked changes of the transcriptomic profiles, with over 400 genes differentially expressed. Cholesterol metabolic processes and extracellular matrix organization gene sets were influenced and represent AChR-trafficking related pathways. Muscle contraction and cellular homeostasis gene sets were also affected, and independently of AChR trafficking. Furthermore, we found changes in a protein-coding RNA and lncRNA network, where expression of lncRNA MEG3 correlated closely with protein-coding genes for cellular homeostasis. We conclude that AChR antibodies induce an active response in human skeletal muscle cells which affects key intra- and extracellular pathways.
Collapse
Affiliation(s)
- Yu Hong
- Department of Clinical Medicine, University of Bergen, Bergen, Norway.
| | - Xiao Liang
- Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Nils Erik Gilhus
- Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Department of Neurology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
249
|
Harikrishnan K, Joshi O, Madangirikar S, Balasubramanian N. Cell Derived Matrix Fibulin-1 Associates With Epidermal Growth Factor Receptor to Inhibit Its Activation, Localization and Function in Lung Cancer Calu-1 Cells. Front Cell Dev Biol 2020; 8:522. [PMID: 32719793 PMCID: PMC7348071 DOI: 10.3389/fcell.2020.00522] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 06/02/2020] [Indexed: 12/11/2022] Open
Abstract
Epidermal Growth Factor Receptor (EGFR) is a known promoter of tumor progression and is overexpressed in lung cancers. Growth factor receptors (including EGFR) are known to interact with extracellular matrix (ECM) proteins, which regulate their activation and function. Fibulin-1 (FBLN1) is a major component of the ECM in lung tissue, and its levels are known to be downregulated in non-small cell lung cancers (NSCLC). To test the possible role FBLN1 isoforms could have in regulating EGFR signaling and function in lung cancer, we performed siRNA mediated knockdown of FBLN1C and FBLN1D in NSCLC Calu-1 cells. Their loss significantly increased basal (with serum) and EGF (Epidermal Growth Factor) mediated EGFR activation without affecting net EGFR levels. Overexpression of FBLN1C and FBLN1D also inhibits EGFR activation confirming their regulatory crosstalk. Loss of FBLN1C and FBLN1D promotes EGFR-dependent cell migration, inhibited upon Erlotinib treatment. Mechanistically, both FBLN1 isoforms interact with EGFR, their association not dependent on its activation. Notably, cell-derived matrix (CDM) enriched FBLN1 binds EGFR. Calu-1 cells plated on CDM derived from FBLN1C and FBLN1D knockdown cells show a significant increase in EGF mediated EGFR activation. This promotes cell adhesion and spreading with active EGFR enriched at membrane ruffles. Both adhesion and spreading on CDMs is significantly reduced by Erlotinib treatment. Together, these findings show FBLN1C/1D, as part of the ECM, can bind and regulate EGFR activation and function in NSCLC Calu-1 cells. They further highlight the role tumor ECM composition could have in influencing EGFR dependent lung cancers.
Collapse
Affiliation(s)
| | - Omkar Joshi
- Indian Institute of Science Education and Research, Pune, India
| | | | | |
Collapse
|
250
|
Marini JJ, Rocco PRM, Gattinoni L. Static and Dynamic Contributors to Ventilator-induced Lung Injury in Clinical Practice. Pressure, Energy, and Power. Am J Respir Crit Care Med 2020; 201:767-774. [PMID: 31665612 PMCID: PMC7124710 DOI: 10.1164/rccm.201908-1545ci] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Ventilation is inherently a dynamic process. The present-day clinical practice of concentrating on the static inflation characteristics of the individual tidal cycle (plateau pressure, positive end-expiratory pressure, and their difference [driving pressure, the ratio of Vt to compliance]) does not take into account key factors shown experimentally to influence ventilator-induced lung injury (VILI). These include rate of airway pressure change (influenced by flow amplitude, inspiratory time fraction, and inspiratory inflation contour) and cycling frequency. Energy must be expended to cause injury, and the product of applied stress and resulting strain determines the energy delivered to the lungs per breathing cycle. Understanding the principles of VILI energetics may provide valuable insights and guidance to intensivists for safer clinical practice. In this interpretive review, we highlight that the injuring potential of the inflation pattern depends upon tissue vulnerability, the number of intolerable high-energy cycles applied in unit time (mechanical power), and the duration of that exposure. Yet, as attractive as this energy/power hypothesis for encapsulating the drivers of VILI may be for clinical applications, we acknowledge that even these all-inclusive and measurable ergonomic parameters (energy per cycle and power) are still too bluntly defined to pinpoint the precise biophysical link between ventilation strategy and tissue injury.
Collapse
Affiliation(s)
- John J Marini
- University of Minnesota and Regions Hospital, Minneapolis/St. Paul, Minnesota
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; and
| | - Luciano Gattinoni
- Department of Anaesthesiology, Emergency and Intensive Care Medicine, University of Göttingen, Göttingen, Germany
| |
Collapse
|