1
|
Nizamoglu M, Joglekar MM, Almeida CR, Larsson Callerfelt AK, Dupin I, Guenat OT, Henrot P, van Os L, Otero J, Elowsson L, Farre R, Burgess JK. Innovative three-dimensional models for understanding mechanisms underlying lung diseases: powerful tools for translational research. Eur Respir Rev 2023; 32:230042. [PMID: 37495250 PMCID: PMC10369168 DOI: 10.1183/16000617.0042-2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 05/04/2023] [Indexed: 07/28/2023] Open
Abstract
Chronic lung diseases result from alteration and/or destruction of lung tissue, inevitably causing decreased breathing capacity and quality of life for patients. While animal models have paved the way for our understanding of pathobiology and the development of therapeutic strategies for disease management, their translational capacity is limited. There is, therefore, a well-recognised need for innovative in vitro models to reflect chronic lung diseases, which will facilitate mechanism investigation and the advancement of new treatment strategies. In the last decades, lungs have been modelled in healthy and diseased conditions using precision-cut lung slices, organoids, extracellular matrix-derived hydrogels and lung-on-chip systems. These three-dimensional models together provide a wide spectrum of applicability and mimicry of the lung microenvironment. While each system has its own limitations, their advantages over traditional two-dimensional culture systems, or even over animal models, increases the value of in vitro models. Generating new and advanced models with increased translational capacity will not only benefit our understanding of the pathobiology of lung diseases but should also shorten the timelines required for discovery and generation of new therapeutics. This article summarises and provides an outline of the European Respiratory Society research seminar "Innovative 3D models for understanding mechanisms underlying lung diseases: powerful tools for translational research", held in Lisbon, Portugal, in April 2022. Current in vitro models developed for recapitulating healthy and diseased lungs are outlined and discussed with respect to the challenges associated with them, efforts to develop best practices for model generation, characterisation and utilisation of models and state-of-the-art translational potential.
Collapse
Affiliation(s)
- Mehmet Nizamoglu
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
- Both authors contributed equally
| | - Mugdha M Joglekar
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
- Both authors contributed equally
| | - Catarina R Almeida
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| | | | - Isabelle Dupin
- Centre de Recherche Cardio-thoracique de Bordeaux, Université de Bordeaux, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, Pessac, France
| | - Olivier T Guenat
- Organs-on-Chip Technologies, ARTORG Center for Biomedical Engineering Research, 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
| | - Pauline Henrot
- Centre de Recherche Cardio-thoracique de Bordeaux, Université de Bordeaux, Pessac, France
- INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, Pessac, France
- Service d'exploration fonctionnelle respiratoire, CHU de Bordeaux, Pessac, France
| | - Lisette van Os
- Organs-on-Chip Technologies, ARTORG Center for Biomedical Engineering Research, University of Bern, Bern, Switzerland
| | - Jorge Otero
- Unit of Biophysics and Bioengineering, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Madrid, Spain
| | - Linda Elowsson
- Lung Biology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Ramon Farre
- Unit of Biophysics and Bioengineering, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Madrid, Spain
- Institut Investigacions Biomediques August Pi Sunyer, Barcelona, Spain
| | - Janette K Burgess
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, W.J. Kolff Institute for Biomedical Engineering and Materials Science-FB41, Groningen, The Netherlands
| |
Collapse
|
2
|
Humbert MV, Spalluto CM, Bell J, Blume C, Conforti F, Davies ER, Dean LSN, Elkington P, Haitchi HM, Jackson C, Jones MG, Loxham M, Lucas JS, Morgan H, Polak M, Staples KJ, Swindle EJ, Tezera L, Watson A, Wilkinson TMA. Towards an artificial human lung: modelling organ-like complexity to aid mechanistic understanding. Eur Respir J 2022; 60:2200455. [PMID: 35777774 DOI: 10.1183/13993003.00455-2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/11/2022] [Indexed: 11/05/2022]
Abstract
Respiratory diseases account for over 5 million deaths yearly and are a huge burden to healthcare systems worldwide. Murine models have been of paramount importance to decode human lung biology in vivo, but their genetic, anatomical, physiological and immunological differences with humans significantly hamper successful translation of research into clinical practice. Thus, to clearly understand human lung physiology, development, homeostasis and mechanistic dysregulation that may lead to disease, it is essential to develop models that accurately recreate the extraordinary complexity of the human pulmonary architecture and biology. Recent advances in micro-engineering technology and tissue engineering have allowed the development of more sophisticated models intending to bridge the gap between the native lung and its replicates in vitro Alongside advanced culture techniques, remarkable technological growth in downstream analyses has significantly increased the predictive power of human biology-based in vitro models by allowing capture and quantification of complex signals. Refined integrated multi-omics readouts could lead to an acceleration of the translational pipeline from in vitro experimental settings to drug development and clinical testing in the future. This review highlights the range and complexity of state-of-the-art lung models for different areas of the respiratory system, from nasal to large airways, small airways and alveoli, with consideration of various aspects of disease states and their potential applications, including pre-clinical drug testing. We explore how development of optimised physiologically relevant in vitro human lung models could accelerate the identification of novel therapeutics with increased potential to translate successfully from the bench to the patient's bedside.
Collapse
Affiliation(s)
- Maria Victoria Humbert
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Cosma Mirella Spalluto
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- M.V. Humbert and C.M. Spalluto are co-first authors and contributed equally to this work
| | - Joseph Bell
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Cornelia Blume
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Franco Conforti
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Elizabeth R Davies
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| | - Lareb S N Dean
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Paul Elkington
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Hans Michael Haitchi
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Claire Jackson
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Mark G Jones
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Matthew Loxham
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Jane S Lucas
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Hywel Morgan
- Institute for Life Sciences, University of Southampton, Southampton, UK
- Electronics and Computer Science, Faculty of Physical Sciences and Engineering, University of Southampton, Southampton, UK
| | - Marta Polak
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Karl J Staples
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| | - Emily J Swindle
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Liku Tezera
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Department of Infection and Immunity, Faculty of Medicine, University College London, London, UK
| | - Alastair Watson
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Tom M A Wilkinson
- School of Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton, UK
| |
Collapse
|
3
|
Herminghaus A, Kozlov AV, Szabó A, Hantos Z, Gylstorff S, Kuebart A, Aghapour M, Wissuwa B, Walles T, Walles H, Coldewey SM, Relja B. A Barrier to Defend - Models of Pulmonary Barrier to Study Acute Inflammatory Diseases. Front Immunol 2022; 13:895100. [PMID: 35874776 PMCID: PMC9300899 DOI: 10.3389/fimmu.2022.895100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 06/20/2022] [Indexed: 12/04/2022] Open
Abstract
Pulmonary diseases represent four out of ten most common causes for worldwide mortality. Thus, pulmonary infections with subsequent inflammatory responses represent a major public health concern. The pulmonary barrier is a vulnerable entry site for several stress factors, including pathogens such as viruses, and bacteria, but also environmental factors e.g. toxins, air pollutants, as well as allergens. These pathogens or pathogen-associated molecular pattern and inflammatory agents e.g. damage-associated molecular pattern cause significant disturbances in the pulmonary barrier. The physiological and biological functions, as well as the architecture and homeostatic maintenance of the pulmonary barrier are highly complex. The airway epithelium, denoting the first pulmonary barrier, encompasses cells releasing a plethora of chemokines and cytokines, and is further covered with a mucus layer containing antimicrobial peptides, which are responsible for the pathogen clearance. Submucosal antigen-presenting cells and neutrophilic granulocytes are also involved in the defense mechanisms and counterregulation of pulmonary infections, and thus may directly affect the pulmonary barrier function. The detailed understanding of the pulmonary barrier including its architecture and functions is crucial for the diagnosis, prognosis, and therapeutic treatment strategies of pulmonary diseases. Thus, considering multiple side effects and limited efficacy of current therapeutic treatment strategies in patients with inflammatory diseases make experimental in vitro and in vivo models necessary to improving clinical therapy options. This review describes existing models for studyying the pulmonary barrier function under acute inflammatory conditions, which are meant to improve the translational approaches for outcome predictions, patient monitoring, and treatment decision-making.
Collapse
Affiliation(s)
- Anna Herminghaus
- Department of Anaesthesiology, University of Duesseldorf, Duesseldorf, Germany
| | - Andrey V. Kozlov
- L Boltzmann Institute for Traumatology in Cooperation with AUVA and Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Department of Human Pathology , IM Sechenov Moscow State Medical University, Moscow, Russia
| | - Andrea Szabó
- Institute of Surgical Research, University of Szeged, Szeged, Hungary
| | - Zoltán Hantos
- Department of Anaesthesiology and Intensive Therapy, Semmelweis University, Budapest, Hungary
| | - Severin Gylstorff
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto-von-Guericke University, Magdeburg, Germany
- Research Campus STIMULATE, Otto-von-Guericke University, Magdeburg, Germany
| | - Anne Kuebart
- Department of Anaesthesiology, University of Duesseldorf, Duesseldorf, Germany
| | - Mahyar Aghapour
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto-von-Guericke University, Magdeburg, Germany
| | - Bianka Wissuwa
- Department of Anaesthesiology and Intensive Care Medicine, Septomics Research Centre, Centre for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Thorsten Walles
- Department of Thoracic Surgery, Magdeburg University Medicine, Magdeburg, Germany
| | - Heike Walles
- Research Campus STIMULATE, Otto-von-Guericke University, Magdeburg, Germany
- Core Facility Tissue Engineering, Otto-von-Guericke-University, Magdeburg, Germany
| | - Sina M. Coldewey
- Department of Anaesthesiology and Intensive Care Medicine, Septomics Research Centre, Centre for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| | - Borna Relja
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto-von-Guericke University, Magdeburg, Germany
- Research Campus STIMULATE, Otto-von-Guericke University, Magdeburg, Germany
- *Correspondence: Borna Relja,
| |
Collapse
|
4
|
Rothbauer M, Bachmann BE, Eilenberger C, Kratz SR, Spitz S, Höll G, Ertl P. A Decade of Organs-on-a-Chip Emulating Human Physiology at the Microscale: A Critical Status Report on Progress in Toxicology and Pharmacology. MICROMACHINES 2021; 12:470. [PMID: 33919242 PMCID: PMC8143089 DOI: 10.3390/mi12050470] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 12/22/2022]
Abstract
Organ-on-a-chip technology has the potential to accelerate pharmaceutical drug development, improve the clinical translation of basic research, and provide personalized intervention strategies. In the last decade, big pharma has engaged in many academic research cooperations to develop organ-on-a-chip systems for future drug discoveries. Although most organ-on-a-chip systems present proof-of-concept studies, miniaturized organ systems still need to demonstrate translational relevance and predictive power in clinical and pharmaceutical settings. This review explores whether microfluidic technology succeeded in paving the way for developing physiologically relevant human in vitro models for pharmacology and toxicology in biomedical research within the last decade. Individual organ-on-a-chip systems are discussed, focusing on relevant applications and highlighting their ability to tackle current challenges in pharmacological research.
Collapse
Affiliation(s)
- Mario Rothbauer
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9/163-164, 1060 Vienna, Austria; (B.E.M.B.); (C.E.); (S.R.A.K.); (S.S.); (G.H.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- Karl Chiari Lab for Orthopaedic Biology, Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Währinger Gürtel 18-22, 1090 Vienna, Austria
| | - Barbara E.M. Bachmann
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9/163-164, 1060 Vienna, Austria; (B.E.M.B.); (C.E.); (S.R.A.K.); (S.S.); (G.H.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Allgemeine Unfallversicherungsanstalt (AUVA) Research Centre, Donaueschingenstraße 13, 1200 Vienna, Austria
| | - Christoph Eilenberger
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9/163-164, 1060 Vienna, Austria; (B.E.M.B.); (C.E.); (S.R.A.K.); (S.S.); (G.H.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Sebastian R.A. Kratz
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9/163-164, 1060 Vienna, Austria; (B.E.M.B.); (C.E.); (S.R.A.K.); (S.S.); (G.H.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- Drug Delivery and 3R-Models Group, Buchmann Institute for Molecular Life Sciences & Institute for Pharmaceutical Technology, Goethe University Frankfurt Am Main, 60438 Frankfurt, Germany
| | - Sarah Spitz
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9/163-164, 1060 Vienna, Austria; (B.E.M.B.); (C.E.); (S.R.A.K.); (S.S.); (G.H.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Gregor Höll
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9/163-164, 1060 Vienna, Austria; (B.E.M.B.); (C.E.); (S.R.A.K.); (S.S.); (G.H.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | - Peter Ertl
- Faculty of Technical Chemistry, Institute of Applied Synthetic Chemistry and Institute of Chemical Technologies and Analytics, Vienna University of Technology, Getreidemarkt 9/163-164, 1060 Vienna, Austria; (B.E.M.B.); (C.E.); (S.R.A.K.); (S.S.); (G.H.)
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| |
Collapse
|
5
|
Dang H, Polineni D, Pace RG, Stonebraker JR, Corvol H, Cutting GR, Drumm ML, Strug LJ, O’Neal WK, Knowles MR. Mining GWAS and eQTL data for CF lung disease modifiers by gene expression imputation. PLoS One 2020; 15:e0239189. [PMID: 33253230 PMCID: PMC7703903 DOI: 10.1371/journal.pone.0239189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 09/02/2020] [Indexed: 12/18/2022] Open
Abstract
Genome wide association studies (GWAS) have identified several genomic loci with candidate modifiers of cystic fibrosis (CF) lung disease, but only a small proportion of the expected genetic contribution is accounted for at these loci. We leveraged expression data from CF cohorts, and Genotype-Tissue Expression (GTEx) reference data sets from multiple human tissues to generate predictive models, which were used to impute transcriptional regulation from genetic variance in our GWAS population. The imputed gene expression was tested for association with CF lung disease severity. By comparing and combining results from alternative approaches, we identified 379 candidate modifier genes. We delved into 52 modifier candidates that showed consensus between approaches, and 28 of them were near known GWAS loci. A number of these genes are implicated in the pathophysiology of CF lung disease (e.g., immunity, infection, inflammation, HLA pathways, glycosylation, and mucociliary clearance) and the CFTR protein biology (e.g., cytoskeleton, microtubule, mitochondrial function, lipid metabolism, endoplasmic reticulum/Golgi, and ubiquitination). Gene set enrichment results are consistent with current knowledge of CF lung disease pathogenesis. HLA Class II genes on chr6, and CEP72, EXOC3, and TPPP near the GWAS peak on chr5 are most consistently associated with CF lung disease severity across the tissues tested. The results help to prioritize genes in the GWAS regions, predict direction of gene expression regulation, and identify new candidate modifiers throughout the genome for potential therapeutic development.
Collapse
Affiliation(s)
- Hong Dang
- Marsico Lung Institute, University of North Carolina at Chapel Hill School of Medicine Cystic Fibrosis/Pulmonary Research & Treatment Center, Chapel Hill, North Carolina, United States of America
| | - Deepika Polineni
- University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Rhonda G. Pace
- Marsico Lung Institute, University of North Carolina at Chapel Hill School of Medicine Cystic Fibrosis/Pulmonary Research & Treatment Center, Chapel Hill, North Carolina, United States of America
| | - Jaclyn R. Stonebraker
- Marsico Lung Institute, University of North Carolina at Chapel Hill School of Medicine Cystic Fibrosis/Pulmonary Research & Treatment Center, Chapel Hill, North Carolina, United States of America
| | - Harriet Corvol
- Pediatric Pulmonary Department, Assistance Publique-Hôpitaux sde Paris (AP-HP), Hôpital Trousseau, Institut National de la Santé et la Recherche Médicale (INSERM) U938, Paris, France
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC), Paris 6, Paris, France
| | - Garry R. Cutting
- McKusick-Nathans Institute of Genetic Medicine, Baltimore, Maryland, United States of America
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Mitchell L. Drumm
- Department of Pediatrics, School of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Lisa J. Strug
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Wanda K. O’Neal
- Marsico Lung Institute, University of North Carolina at Chapel Hill School of Medicine Cystic Fibrosis/Pulmonary Research & Treatment Center, Chapel Hill, North Carolina, United States of America
| | - Michael R. Knowles
- Marsico Lung Institute, University of North Carolina at Chapel Hill School of Medicine Cystic Fibrosis/Pulmonary Research & Treatment Center, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
6
|
Peng Y, Xu AR, Chen SY, Huang Y, Han XR, Guan WJ, Wang DY, Zhong NS. Aberrant Epithelial Cell Proliferation in Peripheral Airways in Bronchiectasis. Front Cell Dev Biol 2020; 8:88. [PMID: 32154248 PMCID: PMC7044270 DOI: 10.3389/fcell.2020.00088] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 01/31/2020] [Indexed: 12/13/2022] Open
Abstract
Dilation of bronchi and bronchioles caused by destruction and excessive epithelial remodeling is a characteristic feature of bronchiectasis. It is not known how epithelial progenitor cells contribute to these pathologic conditions in peripheral airways (bronchioles) in bronchiectasis. We aimed to explore the expression levels of signature airway progenitor cells in the dilated bronchioles in patients with bronchiectasis. We obtained the surgically resected peripheral lung tissues from 43 patients with bronchiectasis and 33 control subjects. Immunostaining was performed to determine the expression patterns of thyroid transcription factor-1 (TTF-1, for labeling progenitor cells in distal airways), P63 (basal cells), club cell 10 kDa protein (CC10, club cells), and surfactant protein C (SPC, alveolar type II epithelial cells) in epithelium or sub-epithelium. Here, we reported significantly lower percentage of TTF-1+ cells and CC10+ cells, and higher percentage of P63+ cells within the epithelium of dilated bronchioles compared with control bronchioles. In airway sub-epithelium of the dilated bronchioles, epithelial hyperplasia with disarrangement of TTF-1+ cells yielded cuboidal (100%) and columnar (93.0%) type among bronchiectasis patients. Most progenitor cell markers co-localized with TTF-1. The median (the 1st, 3rd quartile) percentage of P63+TTF-1+, CC10+TTF-1+, and SPC+TTF-1+ cells was 16.0% (8.9, 24.0%), 14.5% (7.1, 20.8%), and 52% (40.3, 64.4%), respectively. For cuboidal epithelial hyperplasia, 91.0% (86.5, 94.0%) of areas co-stained with SPC and TTF-1. Columnar epithelial hyperplasia was characterized by TTF-1 co-staining with P63+TTF-1+ and CC10+TTF-1+ cells. Taken together, aberrant proliferation of airway progenitor cells in both epithelium and sub-epithelium are implicated in bronchiectasis.
Collapse
Affiliation(s)
- Yang Peng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China.,Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ai-Ru Xu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Shi-Ying Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Yan Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Xiao-Rong Han
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Wei-Jie Guan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - De-Yun Wang
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Nan-Shan Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
7
|
Calvert BA, Ryan Firth AL. Application of iPSC to Modelling of Respiratory Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1237:1-16. [PMID: 31468358 PMCID: PMC8274633 DOI: 10.1007/5584_2019_430] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Respiratory disease is one of the leading causes of morbidity and mortality world-wide with an increasing incidence as the aged population prevails. Many lung diseases are treated for symptomatic relief, with no cure available, indicating a critical need for novel therapeutic strategies. Such advances are hampered by a lack of understanding of how human lung pathologies initiate and progress. Research on human lung disease relies on the isolation of primary cells from explanted lungs or the use of immortalized cells, both are limited in their capacity to represent the genomic and phenotypic variability among the population. In an era where we are progressing toward precision medicine the use of patient specific induced pluripotent cells (iPSC) to generate models, where sufficient primary cells and tissues are scarce, has increased our capacity to understand human lung pathophysiology. Directed differentiation of iPSC toward lung presented the initial challenge to overcome in generating iPSC-derived lung epithelial cells. Since then major advances have been made in defining protocols to specify and isolate specific lung lineages, with the generation of airway spheroids and multi cellular organoids now possible. This technological advance has opened up our capacity for human lung research and prospects for autologous cell therapy. This chapter will focus on the application of iPSC to studying human lung disease.
Collapse
Affiliation(s)
- Ben A Calvert
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Amy L Ryan Firth
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles, CA, USA.
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
8
|
Ainslie GR, Davis M, Ewart L, Lieberman LA, Rowlands DJ, Thorley AJ, Yoder G, Ryan AM. Microphysiological lung models to evaluate the safety of new pharmaceutical modalities: a biopharmaceutical perspective. LAB ON A CHIP 2019; 19:3152-3161. [PMID: 31469131 DOI: 10.1039/c9lc00492k] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The lung is a complex organ; it is both the initial barrier for inhaled agents and the site of metabolism and therapeutic effect for a subset of systemically administered drugs. Comprised of more than 40 cell types that are responsible for various important functions, the lung's complexity contributes to the subsequent challenges in developing complex in vitro co-culture models (also called microphysiological systems (MPS), complex in vitro models or organs-on-a-chip). Although there are multiple considerations and limitations in the development and qualification of such in vitro systems, MPS exhibit great promise in the fields of pharmacology and toxicology. Successful development and implementation of MPS models may enable mechanistic bridging between non-clinical species and humans, and increase clinical relevance of safety endpoints, while decreasing overall animal use. This article summarizes, from a biopharmaceutical industry perspective, essential elements for the development and qualification of lung MPS models. Its purpose is to guide MPS developers and manufacturers to expedite MPS utilization for safety assessment in the biopharmaceutical industry.
Collapse
Affiliation(s)
- Garrett R Ainslie
- Drug Metabolism and Pharmacokinetics, Theravance Biopharma US, Inc, South San Francisco, CA, USA.
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Miller AJ, Dye BR, Ferrer-Torres D, Hill DR, Overeem AW, Shea LD, Spence JR. Generation of lung organoids from human pluripotent stem cells in vitro. Nat Protoc 2019; 14:518-540. [PMID: 30664680 DOI: 10.1038/s41596-018-0104-8] [Citation(s) in RCA: 258] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The lung epithelium is derived from the endodermal germ layer, which undergoes a complex series of endoderm-mesoderm-mediated signaling events to generate the final arborized network of conducting airways (bronchi, bronchioles) and gas-exchanging units (alveoli). These stages include endoderm induction, anterior-posterior and dorsal-ventral patterning, lung specification, lung budding, branching morphogenesis, and, finally, maturation. Here we describe a protocol that recapitulates several of these milestones in order to differentiate human pluripotent stem cells (hPSCs) into ventral-anterior foregut spheroids and further into two distinct types of organoids: human lung organoids and bud tip progenitor organoids. The resulting human lung organoids possess cell types and structures that resemble the bronchi/bronchioles of the developing human airway surrounded by lung mesenchyme and cells expressing alveolar-cell markers. The bud tip progenitor organoids possess a population of highly proliferative multipotent cells with in vitro multilineage differentiation potential and in vivo engraftment potential. Human lung organoids can be generated from hPSCs in 50-85 d, and bud tip progenitor organoids can be generated in 22 d. The two hPSC-derived models presented here have been benchmarked with human fetal tissue and found to be representative of human fetal-like tissue. The bud tip progenitor organoids are thus ideal for exploring epithelial fate decisions, while the human lung organoids can be used to model epithelial-mesenchymal cross-talk during human lung development. In addition to their applications in developmental biology, human lung organoids and bud tip progenitor organoids may be implemented in regenerative medicine, tissue engineering, and pharmaceutical safety and efficacy testing.
Collapse
Affiliation(s)
- Alyssa J Miller
- Program in Cell and Molecular Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Briana R Dye
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI, USA
| | - Daysha Ferrer-Torres
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - David R Hill
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Arend W Overeem
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI, USA
| | - Jason R Spence
- Program in Cell and Molecular Biology, University of Michigan Medical School, Ann Arbor, MI, USA. .,Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI, USA. .,Division of Gastroenterology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA. .,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA. .,Center for Organogenesis, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
10
|
Gkatzis K, Taghizadeh S, Huh D, Stainier DYR, Bellusci S. Use of three-dimensional organoids and lung-on-a-chip methods to study lung development, regeneration and disease. Eur Respir J 2018; 52:13993003.00876-2018. [PMID: 30262579 DOI: 10.1183/13993003.00876-2018] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 09/16/2018] [Indexed: 11/05/2022]
Abstract
Differences in lung anatomy between mice and humans, as well as frequently disappointing results when using animal models for drug discovery, emphasise the unmet need for in vitro models that can complement animal studies and improve our understanding of human lung physiology, regeneration and disease. Recent papers have highlighted the use of three-dimensional organoids and organs-on-a-chip to mimic tissue morphogenesis and function in vitro Here, we focus on the respiratory system and provide an overview of these in vitro models, which can be derived from primary lung cells and pluripotent stem cells, as well as healthy or diseased lungs. We emphasise their potential application in studies of respiratory development, regeneration and disease modelling.
Collapse
Affiliation(s)
- Konstantinos Gkatzis
- Dept of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Sara Taghizadeh
- Dept of Internal Medicine, Justus-Liebig University Giessen, Giessen, Germany
| | - Dongeun Huh
- Dept of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Didier Y R Stainier
- Dept of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Saverio Bellusci
- Dept of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.,Excellence Cluster Cardio-Pulmonary System, Justus-Liebig University Giessen, Giessen, Germany and German Center for Lung Research (DZL)
| |
Collapse
|
11
|
Klay D, Hoffman TW, Harmsze AM, Grutters JC, van Moorsel CHM. Systematic review of drug effects in humans and models with surfactant-processing disease. Eur Respir Rev 2018; 27:27/149/170135. [PMID: 29997245 DOI: 10.1183/16000617.0135-2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 04/12/2018] [Indexed: 12/14/2022] Open
Abstract
Fibrotic interstitial pneumonias are a group of rare diseases characterised by distortion of lung interstitium. Patients with mutations in surfactant-processing genes, such as surfactant protein C (SFTPC), surfactant protein A1 and A2 (SFTPA1 and A2), ATP binding cassette A3 (ABCA3) and Hermansky-Pudlak syndrome (HPS1, 2 and 4), develop progressive pulmonary fibrosis, often culminating in fatal respiratory insufficiency. Although many mutations have been described, little is known about the optimal treatment strategy for fibrotic interstitial pneumonia patients with surfactant-processing mutations.We performed a systematic literature review of studies that described a drug effect in patients, cell or mouse models with a surfactant-processing mutation. In total, 73 articles were selected, consisting of 55 interstitial lung disease case reports/series, two clinical trials and 16 cell or mouse studies. Clinical effect parameters included lung function, radiological characteristics and clinical symptoms, while experimental outcome parameters included chemokine/cytokine expression, surfactant trafficking, necrosis and apoptosis. SP600125, a c-jun N-terminal kinase (JNK) inhibitor, hydroxychloroquine and 4-phenylbutyric acid were most frequently studied in disease models and lead to variable outcomes, suggesting that outcome is mutation dependent.This systematic review summarises effect parameters for future studies on surfactant-processing disorders in disease models and provides directions for future trials in affected patients.
Collapse
Affiliation(s)
- Dymph Klay
- Interstitial Lung Disease Center of Excellence, Dept of Pulmonology, St Antonius Hospital, Nieuwegein, The Netherlands
| | - Thijs W Hoffman
- Interstitial Lung Disease Center of Excellence, Dept of Pulmonology, St Antonius Hospital, Nieuwegein, The Netherlands
| | - Ankie M Harmsze
- Dept of Clinical Pharmacy, St Antonius Hospital, Nieuwegein, The Netherlands
| | - Jan C Grutters
- Interstitial Lung Disease Center of Excellence, Dept of Pulmonology, St Antonius Hospital, Nieuwegein, The Netherlands.,Division of Heart and Lung, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Coline H M van Moorsel
- Interstitial Lung Disease Center of Excellence, Dept of Pulmonology, St Antonius Hospital, Nieuwegein, The Netherlands .,Division of Heart and Lung, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
12
|
Surate Solaligue DE, Rodríguez-Castillo JA, Ahlbrecht K, Morty RE. Recent advances in our understanding of the mechanisms of late lung development and bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2017; 313:L1101-L1153. [PMID: 28971976 DOI: 10.1152/ajplung.00343.2017] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 09/21/2017] [Accepted: 09/23/2017] [Indexed: 02/08/2023] Open
Abstract
The objective of lung development is to generate an organ of gas exchange that provides both a thin gas diffusion barrier and a large gas diffusion surface area, which concomitantly generates a steep gas diffusion concentration gradient. As such, the lung is perfectly structured to undertake the function of gas exchange: a large number of small alveoli provide extensive surface area within the limited volume of the lung, and a delicate alveolo-capillary barrier brings circulating blood into close proximity to the inspired air. Efficient movement of inspired air and circulating blood through the conducting airways and conducting vessels, respectively, generates steep oxygen and carbon dioxide concentration gradients across the alveolo-capillary barrier, providing ideal conditions for effective diffusion of both gases during breathing. The development of the gas exchange apparatus of the lung occurs during the second phase of lung development-namely, late lung development-which includes the canalicular, saccular, and alveolar stages of lung development. It is during these stages of lung development that preterm-born infants are delivered, when the lung is not yet competent for effective gas exchange. These infants may develop bronchopulmonary dysplasia (BPD), a syndrome complicated by disturbances to the development of the alveoli and the pulmonary vasculature. It is the objective of this review to update the reader about recent developments that further our understanding of the mechanisms of lung alveolarization and vascularization and the pathogenesis of BPD and other neonatal lung diseases that feature lung hypoplasia.
Collapse
Affiliation(s)
- David E Surate Solaligue
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| | - José Alberto Rodríguez-Castillo
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| | - Katrin Ahlbrecht
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and.,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| | - Rory E Morty
- Department of Lung Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and .,Department of Internal Medicine (Pulmonology), University of Giessen and Marburg Lung Center, German Center for Lung Research, Giessen, Germany
| |
Collapse
|
13
|
Miller AJ, Spence JR. In Vitro Models to Study Human Lung Development, Disease and Homeostasis. Physiology (Bethesda) 2017; 32:246-260. [PMID: 28404740 DOI: 10.1152/physiol.00041.2016] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 01/23/2017] [Accepted: 01/23/2017] [Indexed: 01/08/2023] Open
Abstract
The main function of the lung is to support gas exchange, and defects in lung development or diseases affecting the structure and function of the lung can have fatal consequences. Most of what we currently understand about human lung development and disease has come from animal models. However, animal models are not always fully able to recapitulate human lung development and disease, highlighting an area where in vitro models of the human lung can compliment animal models to further understanding of critical developmental and pathological mechanisms. This review will discuss current advances in generating in vitro human lung models using primary human tissue, cell lines, and human pluripotent stem cell derived lung tissue, and will discuss crucial next steps in the field.
Collapse
Affiliation(s)
- Alyssa J Miller
- PhD Program in Cell and Molecular Biology, University of Michigan Medical School, Ann Arbor, Michigan.,Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Jason R Spence
- PhD Program in Cell and Molecular Biology, University of Michigan Medical School, Ann Arbor, Michigan; .,PhD Program in Cell and Molecular Biology, University of Michigan Medical School, Ann Arbor, Michigan.,PhD Program in Cell and Molecular Biology, University of Michigan Medical School, Ann Arbor, Michigan.,Center for Organogenesis, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
14
|
Directed differentiation of human induced pluripotent stem cells into functional cholangiocyte-like cells. Nat Protoc 2017; 12:814-827. [PMID: 28333915 DOI: 10.1038/nprot.2017.011] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The difficulty in isolating and propagating functional primary cholangiocytes is a major limitation in the study of biliary disorders and the testing of novel therapeutic agents. To overcome this problem, we have developed a platform for the differentiation of human pluripotent stem cells (hPSCs) into functional cholangiocyte-like cells (CLCs). We have previously reported that our 26-d protocol closely recapitulates key stages of biliary development, starting with the differentiation of hPSCs into endoderm and subsequently into foregut progenitor (FP) cells, followed by the generation of hepatoblasts (HBs), cholangiocyte progenitors (CPs) expressing early biliary markers and mature CLCs displaying cholangiocyte functionality. Compared with alternative protocols for biliary differentiation of hPSCs, our system does not require coculture with other cell types and relies on chemically defined conditions up to and including the generation of CPs. A complex extracellular matrix is used for the maturation of CLCs; therefore, experience in hPSC culture and 3D organoid systems may be necessary for optimal results. Finally, the capacity of our platform for generating large amounts of disease-specific functional cholangiocytes will have broad applications for cholangiopathies, in disease modeling and for screening of therapeutic compounds.
Collapse
|
15
|
|
16
|
Cai WF, Huang W, Wang L, Wang JP, Zhang L, Ashraf M, Wu S, Wang Y. Induced Pluripotent Stem Cells derived Muscle Progenitors Effectively Mitigate Muscular Dystrophy through Restoring the Dystrophin Distribution. ACTA ACUST UNITED AC 2016; 6. [PMID: 28979820 PMCID: PMC5624556 DOI: 10.4172/2157-7633.1000361] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Background Duchenne Muscular Dystrophy (DMD) is a recessive form of muscular disorder, resulting from the dystrophin gene mutations in X-chromosome. Application of embryonic stem cells or adult stem cells has demonstrated the therapeutic effects on DMD through both cell-based and non-cell based mechanisms. In this study, we proposed that Myogenic Progenitor Cells from Induced Pluripotent Stem Cells (iPSC-MPCs) would be more effective in repairing muscle damage caused by muscular dystrophy. Methods and results Mouse iPSCs were cultured in myogenic differentiation culture medium and the MPCs were characterized using Reverse Transcription Polymerase Chain Reaction (RT-PCR) and flow cytometry. iPSCs were successfully converted into MPCs, as evidenced by the distinct expression of myogenic genes and cell surface markers. The muscle injury was induced in tibialis muscle of mdx mouse by cardiotoxin injection, and the iPSC-MPCs were then engrafted into the damage site. Firefly luciferase expression vector was transduced into iPSC-MPCs and the in vivo bioluminescence imaging analysis revealed that these progenitor cells survived even at 30-days post transplantation. Importantly, histological analysis revealed that the central nuclei percentage, as well as fibrosis, was significantly reduced in the iPSC-MPCs treated muscle. In addition,the transplantation of progenitor cells restored the distributions of dystrophin and nicotinic acetylcholine receptors together with up-regulation of pair box protein 7(Pax7), a myogenic transcription factor. Conclusion iPSCs-derived MPCs exert strong therapeutic effects on muscular dystrophy by restoring dystrophin expression and acetylcholine receptor distribution.
Collapse
Affiliation(s)
- Wen-Feng Cai
- Department of Pathology and Lab Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA
| | - Wei Huang
- Department of Pathology and Lab Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA
| | - Lei Wang
- Department of Pathology and Lab Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA
| | - Jia-Peng Wang
- Department of Pathology and Lab Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA
| | - Lu Zhang
- Department of Pathology and Lab Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA.,Key Laboratory of Functional Proteomics of Guangdong Province, Department of Pathophysiology, Southern Medical University, Guangzhou 510515, China
| | - Muhammad Ashraf
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Shizheng Wu
- Qinghai Provincial People's Hospital, 2 Gonghe Rd, Xining, Qinghai, 810007, China
| | - Yigang Wang
- Department of Pathology and Lab Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267-0529, USA
| |
Collapse
|
17
|
Grapin-Botton A. Three-dimensional pancreas organogenesis models. Diabetes Obes Metab 2016; 18 Suppl 1:33-40. [PMID: 27615129 PMCID: PMC5021194 DOI: 10.1111/dom.12720] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 05/02/2016] [Indexed: 01/07/2023]
Abstract
A rediscovery of three-dimensional culture has led to the development of organ biogenesis, homeostasis and disease models applicable to human tissues. The so-called organoids that have recently flourished serve as valuable models bridging between cell lines or primary cells grown on the bottom of culture plates and experiments performed in vivo. Though not recapitulating all aspects of organ physiology, the miniature organs generated in a dish are useful models emerging for the pancreas, starting from embryonic progenitors, adult cells, tumour cells and stem cells. This review focusses on the currently available systems and their relevance to the study of the pancreas, of β-cells and of several pancreatic diseases including diabetes. We discuss the expected future developments for studying human pancreas development and function, for developing diabetes models and for producing therapeutic cells.
Collapse
|
18
|
Aurora M, Spence JR. hPSC-derived lung and intestinal organoids as models of human fetal tissue. Dev Biol 2016; 420:230-238. [PMID: 27287882 DOI: 10.1016/j.ydbio.2016.06.006] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 05/23/2016] [Accepted: 06/04/2016] [Indexed: 02/07/2023]
Abstract
In vitro human pluripotent stem cell (hPSC) derived tissues are excellent models to study certain aspects of normal human development. Current research in the field of hPSC derived tissues reveals these models to be inherently fetal-like on both a morphological and gene expression level. In this review we briefly discuss current methods for differentiating lung and intestinal tissue from hPSCs into individual 3-dimensional units called organoids. We discuss how these methods mirror what is known about in vivo signaling pathways of the developing embryo. Additionally, we will review how the inherent immaturity of these models lends them to be particularly valuable in the study of immature human tissues in the clinical setting of premature birth. Human lung organoids (HLOs) and human intestinal organoids (HIOs) not only model normal development, but can also be utilized to study several important diseases of prematurity such as respiratory distress syndrome (RDS), bronchopulmonary dysplasia (BPD), and necrotizing enterocolitis (NEC).
Collapse
Affiliation(s)
- Megan Aurora
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Jason R Spence
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, MI, United States; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States; Center for Organogenesis, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
19
|
Konar D, Devarasetty M, Yildiz DV, Atala A, Murphy SV. Lung-On-A-Chip Technologies for Disease Modeling and Drug Development. Biomed Eng Comput Biol 2016; 7:17-27. [PMID: 27127414 PMCID: PMC4839966 DOI: 10.4137/becb.s34252] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 03/15/2016] [Accepted: 03/31/2016] [Indexed: 01/13/2023] Open
Abstract
Animal and two-dimensional cell culture models have had a profound impact on not only lung research but also medical research at large, despite inherent flaws and differences when compared with in vivo and clinical observations. Three-dimensional (3D) tissue models are a natural progression and extension of existing techniques that seek to plug the gaps and mitigate the drawbacks of two-dimensional and animal technologies. In this review, we describe the transition of historic models to contemporary 3D cell and organoid models, the varieties of current 3D cell and tissue culture modalities, the common methods for imaging these models, and finally, the applications of these models and imaging techniques to lung research.
Collapse
Affiliation(s)
- Dipasri Konar
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Mahesh Devarasetty
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Didem V Yildiz
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Sean V Murphy
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
20
|
Dye BR, Miller AJ, Spence JR. How to Grow a Lung: Applying Principles of Developmental Biology to Generate Lung Lineages from Human Pluripotent Stem Cells. CURRENT PATHOBIOLOGY REPORTS 2016; 4:47-57. [PMID: 27340610 PMCID: PMC4882378 DOI: 10.1007/s40139-016-0102-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The number and severity of diseases affecting human lung development and adult respiratory function has stimulated great interest in new in vitro models to study the human lung. This review summarizes the most recent breakthroughs deriving lung lineages in a dish by directing the differentiation of human pluripotent stem cells. A variety of culturing platforms have been developed, including two-dimensional and three-dimensional (organoid) culture platforms, to derive specific cell types and structures of the lung. These stem cell-derived lung models will further our understanding of human lung development, disease, and regeneration.
Collapse
Affiliation(s)
- Briana R. Dye
- />Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109 USA
| | - Alyssa J. Miller
- />Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48109 USA
- />Department of Cell and Molecular Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109 USA
| | - Jason R. Spence
- />Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109 USA
- />Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48109 USA
- />Department of Cell and Molecular Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109 USA
- />Center for Organogenesis, University of Michigan Medical School, Ann Arbor, Michigan 48109 USA
| |
Collapse
|
21
|
Sampaziotis F, de Brito MC, Madrigal P, Bertero A, Saeb-Parsy K, Soares FAC, Schrumpf E, Melum E, Karlsen TH, Bradley JA, Gelson WTH, Davies S, Baker A, Kaser A, Alexander GJ, Hannan NR, Vallier L. Cholangiocytes derived from human induced pluripotent stem cells for disease modeling and drug validation. Nat Biotechnol 2015; 33:845-852. [PMID: 26167629 PMCID: PMC4768345 DOI: 10.1038/nbt.3275] [Citation(s) in RCA: 279] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 06/05/2015] [Indexed: 12/20/2022]
Abstract
The study of biliary disease has been constrained by a lack of primary human cholangiocytes. Here we present an efficient, serum-free protocol for directed differentiation of human induced pluripotent stem cells into cholangiocyte-like cells (CLCs). CLCs show functional characteristics of cholangiocytes, including bile acids transfer, alkaline phosphatase activity, γ-glutamyl-transpeptidase activity and physiological responses to secretin, somatostatin and vascular endothelial growth factor. We use CLCs to model in vitro key features of Alagille syndrome, polycystic liver disease and cystic fibrosis (CF)-associated cholangiopathy. Furthermore, we use CLCs generated from healthy individuals and patients with polycystic liver disease to reproduce the effects of the drugs verapamil and octreotide, and we show that the experimental CF drug VX809 rescues the disease phenotype of CF cholangiopathy in vitro. Our differentiation protocol will facilitate the study of biological mechanisms controlling biliary development, as well as disease modeling and drug screening.
Collapse
Affiliation(s)
- Fotios Sampaziotis
- Wellcome Trust-Medical Research Council Stem Cell Institute, Anne McLaren Laboratory for Regenerative Medicine, Department of Surgery, University of Cambridge, Cambridge, UK
- Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Miguel Cardoso de Brito
- Wellcome Trust-Medical Research Council Stem Cell Institute, Anne McLaren Laboratory for Regenerative Medicine, Department of Surgery, University of Cambridge, Cambridge, UK
| | - Pedro Madrigal
- Wellcome Trust-Medical Research Council Stem Cell Institute, Anne McLaren Laboratory for Regenerative Medicine, Department of Surgery, University of Cambridge, Cambridge, UK
- Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Alessandro Bertero
- Wellcome Trust-Medical Research Council Stem Cell Institute, Anne McLaren Laboratory for Regenerative Medicine, Department of Surgery, University of Cambridge, Cambridge, UK
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - Filipa A. C. Soares
- Wellcome Trust-Medical Research Council Stem Cell Institute, Anne McLaren Laboratory for Regenerative Medicine, Department of Surgery, University of Cambridge, Cambridge, UK
| | - Elisabeth Schrumpf
- Norwegian PSC Research Center, Division of Cancer, Surgery and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- K.G. Jebsen Inflammation Research Centre, Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Espen Melum
- Norwegian PSC Research Center, Division of Cancer, Surgery and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- K.G. Jebsen Inflammation Research Centre, Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Tom H. Karlsen
- Norwegian PSC Research Center, Division of Cancer, Surgery and Transplantation, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- K.G. Jebsen Inflammation Research Centre, Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - J. Andrew Bradley
- Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - William TH Gelson
- Department of Hepatology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Susan Davies
- Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Alastair Baker
- Child Health Clinical Academic Grouping, King’s Health Partners, Denmark Hill Campus, London, United Kingdom
| | - Arthur Kaser
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - Graeme J. Alexander
- Department of Medicine, School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Nicholas R.F. Hannan
- Wellcome Trust-Medical Research Council Stem Cell Institute, Anne McLaren Laboratory for Regenerative Medicine, Department of Surgery, University of Cambridge, Cambridge, UK
| | - Ludovic Vallier
- Wellcome Trust-Medical Research Council Stem Cell Institute, Anne McLaren Laboratory for Regenerative Medicine, Department of Surgery, University of Cambridge, Cambridge, UK
- Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| |
Collapse
|