1
|
Kary AD, Noelle H, Magin CM. Tissue-Informed Biomaterial Innovations Advance Pulmonary Regenerative Engineering. ACS Macro Lett 2025; 14:434-447. [PMID: 40102038 DOI: 10.1021/acsmacrolett.5c00075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Irreversible progressive pulmonary diseases drastically reduce the patient quality of life, while transplantation remains the only definitive cure. Research into lung regeneration pathways holds significant potential to expand and promote the discovery of new treatment options. Polymeric biomaterials designed to replicate key tissue characteristics (i.e., biochemical composition and mechanical cues) show promise for creating environments in which to study chronic lung diseases and initiate lung tissue regeneration. In this Viewpoint, we explore how naturally derived materials can be employed alone or combined with engineered polymer systems to create advanced tissue culture platforms. Pulmonary tissue models have historically leveraged natural materials, including basement membrane extracts and a decellularized extracellular matrix, as platforms for lung regeneration studies. Here, we provide an overview of the progression of pulmonary regenerative engineering, exploring how innovations in the growing field of tissue-informed biomaterials have the potential to advance lung regeneration research by bridging the gap between biological relevance and mechanical precision.
Collapse
Affiliation(s)
- Anton D Kary
- Department of Bioengineering, University of Colorado, Denver | Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Haley Noelle
- Department of Bioengineering, University of Colorado, Denver | Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Chelsea M Magin
- Department of Bioengineering, University of Colorado, Denver | Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Department of Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045, United States
| |
Collapse
|
2
|
Elliot SJ, Anderson-Terhune D, Roos B, Rubio GA, Xia X, Pereira-Simon S, Catanuto P, Civettini G, Hagen ES, Arvanitis C, Shahzeidi S, Glassberg MK. Ratio of miRNA-29 to miRNA-199 expression coordinates mesenchymal stem cell repair of bleomycin-induced pulmonary injury. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102461. [PMID: 40124162 PMCID: PMC11930095 DOI: 10.1016/j.omtn.2025.102461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 01/17/2025] [Indexed: 03/25/2025]
Abstract
Our previous work demonstrated the anti-fibrotic effects of infusion of adipose-derived mesenchymal stem cells (ASCs) to prevent or repair bleomycin (BLM)-induced lung injury. The present study investigates mechanisms driving these anti-fibrotic effects. Pulmonary fibrosis developed at day 12 in 22-month-old C57BL/6 male mice after intratracheal BLM instillation. There was a decrease in indices of pulmonary fibrosis, including collagen content, AKT activation, collagen types I and III, αV-integrin, tumor necrosis factor alpha, and transforming growth factor β mRNA after infusion of ASCs 12 days post-BLM treatment compared to BLM alone. Infusion of ASCs increased the population of alveolar types I and II epithelial cells that had been reduced after BLM treatment. miRNAscope technology and reverse-transcription polymerase chain reaction revealed that ASC-treated mice demonstrated increased miR-29a, decreased miR-199, and increased telomere length, telomerase RNA component, and telomerase reverse transcriptase compared to BLM alone. In vitro and ex vivo experiments using double-transfected mouse or human myofibroblasts (miR-29 mimic, and miR-199 inhibitor) confirmed that alterations of these miRNAs regulate downstream effectors of fibrosis. These data suggest that alteration of the ratio of anti-fibrotic to fibrotic miRNAs and increase in telomere length are critical mechanisms of ASC-mediated repair of BLM-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Sharon J. Elliot
- Stritch School of Medicine, Department of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - Dustin Anderson-Terhune
- Department of Pulmonary and Critical Care, University of Utah, Salt Lake City, UT 84112, USA
| | - Benjamin Roos
- Stritch School of Medicine, Department of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - Gustavo A. Rubio
- Associate Medical Director, Jackson Health System, 1611 NW 12 Avenue, Miami, FL 33136, USA
| | - Xiaomei Xia
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep, University of Arizona College of Medicine-Phoenix University Medical Center-Phoenix, Phoenix, AZ 85004, USA
| | - Simone Pereira-Simon
- Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33101, USA
| | - Paola Catanuto
- Department of Ophthalmology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33101, USA
| | - Gina Civettini
- Stritch School of Medicine, Department of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - Emily S. Hagen
- Stritch School of Medicine, Department of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - Constadina Arvanitis
- Director of Center for Advanced Microscopy & Nikon Imaging Center, Northwestern University, Chicago, IL 60611, USA
| | - Shahriar Shahzeidi
- Grand Health Institute, 1717 N. Bayshore Drive, Suite R244, Miami, FL 33132, USA
| | - Marilyn K. Glassberg
- Stritch School of Medicine, Department of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| |
Collapse
|
3
|
Melo-Narvaez MC, Gölitz F, Jain E, Gote-Schniering J, Stoleriu MG, Bertrams W, Schmeck B, Yildirim AÖ, Rauen U, Wille T, Lehmann M. Cold storage of human precision-cut lung slices in TiProtec preserves cellular composition and transcriptional responses and enables on-demand mechanistic studies. Respir Res 2025; 26:57. [PMID: 39962456 PMCID: PMC11834602 DOI: 10.1186/s12931-025-03132-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 01/31/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Human precision-cut lung slices (hPCLS) are a unique platform for functional, mechanistic, and drug discovery studies in the field of respiratory research. However, tissue availability, generation, and cultivation time represent important challenges for their usage. Therefore, the present study evaluated the efficacy of a specifically designed tissue preservation solution, TiProtec, complete or in absence (-) of iron chelators, for long-term cold storage of hPCLS. METHODS hPCLS were generated from peritumor control tissues and stored in DMEM/F-12, TiProtec, or TiProtec (-) for up to 28 days. Viability, metabolic activity, and tissue structure were determined. Moreover, bulk-RNA sequencing was used to study transcriptional changes, regulated signaling pathways, and cellular composition after cold storage. Induction of cold storage-associated senescence was determined by transcriptomics and immunofluorescence (IF). Finally, cold-stored hPCLS were exposed to a fibrotic cocktail and early fibrotic changes were assessed by RT-qPCR and IF. RESULTS Here, we found that TiProtec preserves the viability, metabolic activity, transcriptional profile, as well as cellular composition of hPCLS for up to 14 days. Cold storage did not significantly induce cellular senescence in hPCLS. Moreover, TiProtec downregulated pathways associated with cell death, inflammation, and hypoxia while activating pathways protective against oxidative stress. Cold-stored hPCLS remained responsive to fibrotic stimuli and upregulated extracellular matrix-related genes such as fibronectin and collagen 1 as well as alpha-smooth muscle actin, a marker for myofibroblasts. CONCLUSIONS Optimized long-term cold storage of hPCLS preserves their viability, metabolic activity, transcriptional profile, and cellular composition for up to 14 days, specifically in TiProtec. Finally, our study demonstrated that cold-stored hPCLS can be used for on-demand mechanistic studies relevant for respiratory research.
Collapse
Affiliation(s)
- M Camila Melo-Narvaez
- Comprehensive Pneumology Center with the CPC-M bioArchive and Institute of Lung Health and Immunity, Helmholtz Center Munich, German Center for Lung Research (DZL), Munich, Germany
- Institute for Lung Research, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany
| | - Fee Gölitz
- Bundeswehr Institute of Pharmacology and Toxicology, Munich, Germany
- Walther-Straub-Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University, Munich, Germany
| | - Eshita Jain
- Comprehensive Pneumology Center with the CPC-M bioArchive and Institute of Lung Health and Immunity, Helmholtz Center Munich, German Center for Lung Research (DZL), Munich, Germany
| | - Janine Gote-Schniering
- Department of Rheumatology and Immunology, Department of Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Mircea Gabriel Stoleriu
- Comprehensive Pneumology Center with the CPC-M bioArchive and Institute of Lung Health and Immunity, Helmholtz Center Munich, German Center for Lung Research (DZL), Munich, Germany
- Division for Thoracic Surgery Munich, Ludwig-Maximilians-University of Munich (LMU) and Asklepios Lung Clinic Munich-Gauting, Gauting, Germany
| | - Wilhelm Bertrams
- Institute for Lung Research, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany
| | - Bernd Schmeck
- Institute for Lung Research, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany
- Core Facility Flow Cytometry - Bacterial Vesicles, Philipps-University Marburg, Marburg, Germany
- Department of Medicine, Pulmonary and Critical Care Medicine, University Medical Center Marburg, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany
- Center for Synthetic Microbiology (Synmikro), Philipps-University Marburg, Marburg, Germany
- Member of the German Center of Infectious Disease Research, Marburg, Germany
- Institute for Lung Health (ILH), German Center for Lung Research (DZL), Giessen, Germany
| | - Ali Önder Yildirim
- Comprehensive Pneumology Center with the CPC-M bioArchive and Institute of Lung Health and Immunity, Helmholtz Center Munich, German Center for Lung Research (DZL), Munich, Germany
- Institute of Experimental Pneumology (IEP), Ludwig-Maximilians University of Munich (LMU), Munich, Germany
| | - Ursula Rauen
- Institute of Physiological Chemistry, University Hospital Essen, Essen, Germany
| | - Timo Wille
- Bundeswehr Institute of Pharmacology and Toxicology, Munich, Germany.
- Department of CBRN Medical Defense, Bundeswehr Medical Academy, Munich, Germany.
| | - Mareike Lehmann
- Comprehensive Pneumology Center with the CPC-M bioArchive and Institute of Lung Health and Immunity, Helmholtz Center Munich, German Center for Lung Research (DZL), Munich, Germany.
- Institute for Lung Research, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany.
- Institute for Lung Health (ILH), German Center for Lung Research (DZL), Giessen, Germany.
| |
Collapse
|
4
|
Vierhout M, Ayoub A, Ali P, Kumaran V, Naiel S, Isshiki T, Koenig JFE, Kolb MRJ, Ask K. A novel ex vivo approach for investigating profibrotic macrophage polarization using murine precision-cut lung slices. Biochem Biophys Res Commun 2024; 741:151038. [PMID: 39603027 DOI: 10.1016/j.bbrc.2024.151038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/28/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is fatal interstitial lung disease characterized by excessive scarring of the lung tissue and declining respiratory function. Given its short prognosis and limited treatment options, novel strategies to investigate emerging experimental treatments are urgently needed. Macrophages, as the most abundant immune cell in the lung, have key implications in wound healing and lung fibrosis. However, they are highly plastic and adaptive to their surrounding microenvironment, and thus to maximize translation of research to lung disease, there is a need to study macrophages in multifaceted, complex systems that are representative of the lung. Precision-cut lung slices (PCLS) are living tissue preparations derived from the lung that are cultured ex vivo, which bypass the need for artificial recapitulation of the lung milieu and architecture. Macrophage programming studies are traditionally conducted using isolated cells in vitro, thus our objective was to establish and validate a moderate-throughput, biologically-translational, viable model to study profibrotic polarization of pulmonary-resident macrophages using murine PCLS. To achieve this, we used a polarization cocktail (PC), consisting of IL-4, IL-13, and IL-6, over a 72-h time course. We first demonstrated no adverse effects of the PC on PCLS viability and architecture. Next, we showed that multiple markers of macrophage profibrotic polarization, including Arginase-1, CD206, YM1, and CCL17 were induced in PCLS following PC treatment. Through tissue microarray-based histological assessments, we directly visualized and quantified Arginase-1 and CD206 staining in PCLS in a moderate-throughput manner. We further delineated phenotype of polarized macrophages, and using high-plex immunolabelling with the Iterative Bleaching Extends Multiplexity (IBEX) method, showed that the PC effects both interstitial and alveolar macrophages. Substantiating the profibrotic properties of the system, we also showed expression of extracellular matrix components and fibrotic markers in stimulated PCLS. Finally, we demonstrated that clodronate treatment diminishes the PC effects on profibrotic macrophage readouts. Overall, our findings support a suitable complex model for studying ex vivo profibrotic macrophage programming in the lung, with future capacity for investigating experimental therapeutic candidates and disease mechanisms in pulmonary fibrosis.
Collapse
Affiliation(s)
- Megan Vierhout
- Firestone Institute for Respiratory Health, Department of Medicine, McMaster University and the Research Institute of St. Joe's Hamilton, 50 Charlton Avenue East, Hamilton, Ontario, L8N 4A6, Canada; McMaster Immunology Research Centre, Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L8, Canada
| | - Anmar Ayoub
- Firestone Institute for Respiratory Health, Department of Medicine, McMaster University and the Research Institute of St. Joe's Hamilton, 50 Charlton Avenue East, Hamilton, Ontario, L8N 4A6, Canada; McMaster Immunology Research Centre, Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L8, Canada
| | - Pareesa Ali
- Firestone Institute for Respiratory Health, Department of Medicine, McMaster University and the Research Institute of St. Joe's Hamilton, 50 Charlton Avenue East, Hamilton, Ontario, L8N 4A6, Canada; McMaster Immunology Research Centre, Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L8, Canada
| | - Vaishnavi Kumaran
- Firestone Institute for Respiratory Health, Department of Medicine, McMaster University and the Research Institute of St. Joe's Hamilton, 50 Charlton Avenue East, Hamilton, Ontario, L8N 4A6, Canada; McMaster Immunology Research Centre, Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L8, Canada
| | - Safaa Naiel
- Firestone Institute for Respiratory Health, Department of Medicine, McMaster University and the Research Institute of St. Joe's Hamilton, 50 Charlton Avenue East, Hamilton, Ontario, L8N 4A6, Canada; McMaster Immunology Research Centre, Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L8, Canada
| | - Takuma Isshiki
- Firestone Institute for Respiratory Health, Department of Medicine, McMaster University and the Research Institute of St. Joe's Hamilton, 50 Charlton Avenue East, Hamilton, Ontario, L8N 4A6, Canada; McMaster Immunology Research Centre, Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L8, Canada; Department of Respiratory Medicine, Toho University School of Medicine, 6-11-1 Omorinishi, Ota-ku, Tokyo, 143-8540, Japan
| | - Joshua F E Koenig
- Department of Respiratory Medicine, Toho University School of Medicine, 6-11-1 Omorinishi, Ota-ku, Tokyo, 143-8540, Japan; Schroeder Allergy and Immunology Research Institute, Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L8, Canada
| | - Martin R J Kolb
- Firestone Institute for Respiratory Health, Department of Medicine, McMaster University and the Research Institute of St. Joe's Hamilton, 50 Charlton Avenue East, Hamilton, Ontario, L8N 4A6, Canada; McMaster Immunology Research Centre, Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L8, Canada
| | - Kjetil Ask
- Firestone Institute for Respiratory Health, Department of Medicine, McMaster University and the Research Institute of St. Joe's Hamilton, 50 Charlton Avenue East, Hamilton, Ontario, L8N 4A6, Canada; McMaster Immunology Research Centre, Department of Medicine, McMaster University, 1280 Main Street West, Hamilton, Ontario, L8S 4L8, Canada.
| |
Collapse
|
5
|
Liu G, Fang Y, Zhang Y, Zhu M. Dihydroquercetin improves the proliferation of porcine intestinal epithelial cells via the Wnt/β-catenin pathway. Biochem Biophys Res Commun 2024; 734:150460. [PMID: 39083968 DOI: 10.1016/j.bbrc.2024.150460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024]
Abstract
Dihydroquercetin (DHQ), also known as Taxifolin (TA), is a flavanonol with various biological activities, such as anticancer, anti-inflammatory, and antioxidative properties. It has been found to effectively increase the viability of porcine intestinal epithelial cells (IPEC-J2). However, the precise mechanism by which DHQ increases the proliferation of IPEC-J2 cells is not entirely understood. This study aimed to explore the potential pathways through which DHQ encourages the proliferation of IPEC-J2 cells. The findings indicated that DHQ significantly improved the protein expression of tight junction proteins (ZO-1, Occludin, and Claudin1) and a molecular biomarker of proliferation (PCNA) in IPEC-J2 cells. Furthermore, DHQ was found to increase the Wnt/β-catenin pathway-associated β-catenin, c-Myc, and cyclin D1 mRNA expression, and promote the protein expression of β-catenin and TCF4. To confirm the involvement of the Wnt/β-catenin signaling pathway in the DHQ-promoted proliferation of IPEC-J2 cells, the inhibitor LF3, which targets β-catenin/TCF4 interaction, was used. It was found that LF3 inhibited the protein expressions upregulated by DHQ and blocked the promotion of cell proliferation. These results indicate that DHQ positively regulates IPEC-J2 cell proliferation through the Wnt/β-catenin pathway, providing constructive insights into the role of DHQ in regulating intestine development.
Collapse
Affiliation(s)
- Guowei Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, 550025, China; Institute of Animal Nutrition and Feed Science, Guizhou University, Guiyang, 550025, China
| | - Yongxia Fang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, 550025, China; Institute of Animal Nutrition and Feed Science, Guizhou University, Guiyang, 550025, China
| | - Yiyu Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, 550025, China; Institute of Animal Nutrition and Feed Science, Guizhou University, Guiyang, 550025, China
| | - Min Zhu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, 550025, China; Institute of Animal Nutrition and Feed Science, Guizhou University, Guiyang, 550025, China.
| |
Collapse
|
6
|
Lehmann M, Krishnan R, Sucre J, Kulkarni HS, Pineda RH, Anderson C, Banovich NE, Behrsing HP, Dean CH, Haak A, Gosens R, Kaminski N, Zagorska A, Koziol-White C, Metcalf JP, Kim YH, Loebel C, Neptune E, Noel A, Raghu G, Sewald K, Sharma A, Suki B, Sperling A, Tatler A, Turner S, Rosas IO, van Ry P, Wille T, Randell SH, Pryhuber G, Rojas M, Bourke J, Königshoff M. Precision Cut Lung Slices: Emerging Tools for Preclinical and Translational Lung Research. An Official American Thoracic Society Workshop Report. Am J Respir Cell Mol Biol 2024; 72:16-31. [PMID: 39499861 PMCID: PMC11707673 DOI: 10.1165/rcmb.2024-0479st] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Indexed: 11/07/2024] Open
Abstract
The urgent need for effective treatments for acute and chronic lung diseases underscores the significance of developing innovative preclinical human research tools. The 2023 ATS Workshop on Precision Cut Lung Slices (PCLS) brought together 35 experts to discuss and address the role of human tissue-derived PCLS as a unique tool for target and drug discovery and validation in pulmonary medicine. With increasing interest and usage, along with advancements in methods and technology, there is a growing need for consensus on PCLS methodology and readouts. The current document recommends standard reporting criteria and emphasizes the requirement for careful collection and integration of clinical metadata. We further discuss current clinically relevant readouts that can be applied to PCLS and highlight recent developments and future steps for implementing novel technologies for PCLS modeling and analysis. The collection and correlation of clinical metadata and multiomic analysis will further advent the integration of this preclinical platform into patient endotyping and the development of tailored therapies for lung disease patients.
Collapse
Affiliation(s)
- Mareike Lehmann
- Philipps University Marburg, Institute for Lung Research, Marburg, Germany
- Helmholtz Center Munich, Institute for Lung Health and Immunity, Munich, Germany;
| | - Ramaswamy Krishnan
- Beth Israel Deaconess Medical Center, Emergency Medicine, Boston, United States
| | - Jennifer Sucre
- Vanderbilt University Medical Center, Pediatrics, Nashville, Tennessee, United States
| | - Hrishikesh S Kulkarni
- Washington University in Saint Louis, Division of Pulmonary and Critical Care Medicine, Saint Louis, Missouri, United States
| | - Ricardo H Pineda
- University of Pittsburgh, Division of Pulmonary, Allergy and Critical Care Medicine, Pittsburgh, Pennsylvania, United States
| | | | | | - Holger P Behrsing
- Institute for In Vitro Sciences Inc, Gaithersburg, Maryland, United States
| | - Charlotte H Dean
- Imperial College, National Heart and Lung Institute, London, United Kingdom of Great Britain and Northern Ireland
| | - Andrew Haak
- Mayo Clinic College of Medicine, Rochester, Minnesota, United States
| | - Reinoud Gosens
- University of Groningen, Molecular Pharmacology, Groningen, Netherlands
| | - Naftali Kaminski
- Yale School of Medicine , Pulmonary, Critical Care and Sleep Mediine , New Haven, Connecticut, United States
| | - Anna Zagorska
- Gilead Sciences Inc, Foster City, California, United States
| | - Cynthia Koziol-White
- Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, New Jersey, United States
| | - Jordan P Metcalf
- The University of Oklahoma Health Sciences Center, Medicine, Oklahoma City, Oklahoma, United States
| | - Yong Ho Kim
- U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, United States
| | | | - Enid Neptune
- Johns Hopkins, Medicine/Pulmonary and Critical Care, Baltimore, Maryland, United States
| | - Alexandra Noel
- Louisiana State University, Baton Rouge, Louisiana, United States
| | - Ganesh Raghu
- University of Washington Medical Center, Division of Pulmonary and Critical Care Medicine, Seattle, Washington, United States
| | | | - Ashish Sharma
- University of Florida, Gainesville, Florida, United States
| | - Bela Suki
- Boston University, Biomedical Engineering, Boston, Massachusetts, United States
| | - Anne Sperling
- University of Virginia School of Medicine, Charlottesville, Virginia, United States
| | - Amanda Tatler
- University of Nottingham, Respiratory Medicine , Nottingham, United Kingdom of Great Britain and Northern Ireland
| | - Scott Turner
- Pliant Therapeutics, South San Francisco, California, United States
| | - Ivan O Rosas
- Brigham and Women's Hospital, Department of Medicine, Division of Pulmonary and Critical Care Medicine, Boston, Massachusetts, United States
| | - Pam van Ry
- Brigham Young University, Chemistry and Biochemistry, Provo, Utah, United States
| | - Timo Wille
- Bundeswehr Institute of Pharmacology and Toxicology, Bundeswehr Medical Academy, Germany, Munich, Germany
| | - Scott H Randell
- University of North Carolina, Department of Cell Biology & Physiology, Chapel Hill, North Carolina, United States
| | - Gloria Pryhuber
- University of Rochester, Pediatrics, Rochester, New York, United States
| | - Mauricio Rojas
- Ohio State University, Columbus, OH, Pulmonary, Critical Care and Sleep Medicine, College of Medicine, , Columbus, Ohio, United States
| | - Jane Bourke
- Monash University, Department of Pharmacology, Biomedicine Discovery Institute, Clayton, Victoria, Australia
| | - Melanie Königshoff
- University of Pittsburgh, Medicine, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
7
|
Nowakowska J, Gvazava N, Langwiński W, Ziarniak K, da Silva IAN, Stegmayr J, Wagner DE, Szczepankiewicz A. Optimizing miRNA transfection for screening in precision cut lung slices. Am J Physiol Lung Cell Mol Physiol 2024; 327:L712-L723. [PMID: 39254091 PMCID: PMC11563635 DOI: 10.1152/ajplung.00138.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/31/2024] [Accepted: 08/30/2024] [Indexed: 09/11/2024] Open
Abstract
Precision cut lung slices (PCLS) are complex three-dimensional (3-D) lung tissue models, which preserve the native microenvironment, including cell diversity and cell-matrix interactions. They are an innovative ex vivo platform that allows studying disease as well as the effects of therapeutic agents or regulatory molecules [e.g., microRNA (miRNA)]. The aim of our study was to develop a protocol to transfect PCLS with miRNA using lipid nanoparticles (LNPs) to enable higher throughput screening of miRNA, obviating the need for custom stabilization and internalization approaches. PCLS of 4 mm diameter were generated using agarose-filled rodent lungs and a vibratome. TYE665-labeled scrambled miRNA was used to evaluate transfection efficacy of six different commercially available LNPs. Transfection efficacy was visualized using live high-content fluorescence microscopy, followed by higher-resolution confocal fluorescence microscopy in fixed PCLS. Metabolic activity and cellular damage were assessed using water-soluble tetrazolium salt (WST-1) and lactate dehydrogenase (LDH) release. Using a live staining kit containing a cell membrane impermeant nuclear dye, RedDot2, we established that cellular membranes in PCLS are permeable in the initial 24 h of slicing but diminished thereafter. Therefore, all transfection experiments occurred at least 24 h after slicing. All six commercially available LNPs enabled transfection without inducing significant cytotoxicity or impaired metabolic function. However, RNAiMAX and INTERFERin led to increases in transfection efficacy as compared with other LNPs, with detection possible as low as 25 nM. Therefore, LNP-based transfection of miRNA is possible and can be visualized in live or fixed PCLS, enabling future higher throughput studies using diverse miRNAs.NEW & NOTEWORTHY RNA-based therapeutics hold significant promise for disease treatment; however, limited research exists on miRNA transfection specifically within PCLS. miRNA transfection has thus far required custom functionalization for stabilization and internalization. We aimed to optimize a transfection protocol for rapid screening approaches of miRNA sequences. We show that transfecting miRNA in PCLS is possible using lipid nanoparticles. In addition, we show that 25 nM of TYE665-miRNA is sufficient for detection in a high-content imaging system.
Collapse
Affiliation(s)
- Joanna Nowakowska
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
- Doctoral School, Poznan University of Medical Sciences, Poznan, Poland
| | - Nika Gvazava
- Lung Bioengineering and Regeneration, Department of Experimental Medical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Faculty of Medicine, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
- NanoLund, Lund University, Lund, Sweden
| | - Wojciech Langwiński
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Kamil Ziarniak
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland
| | - Iran Augusto N da Silva
- Lung Bioengineering and Regeneration, Department of Experimental Medical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Faculty of Medicine, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
- NanoLund, Lund University, Lund, Sweden
| | - John Stegmayr
- Lung Bioengineering and Regeneration, Department of Experimental Medical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Faculty of Medicine, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
- NanoLund, Lund University, Lund, Sweden
| | - Darcy E Wagner
- Lung Bioengineering and Regeneration, Department of Experimental Medical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
- Lund Stem Cell Center, Faculty of Medicine, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
- NanoLund, Lund University, Lund, Sweden
- Meakins-Christie Laboratories, The Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Department of Biomedical Engineering, McGill University, Montreal, Quebec, Canada
| | | |
Collapse
|
8
|
Nicoletto RE, Holdcraft CJ, Yin AC, Retzbach EP, Sheehan SA, Greenspan AA, Laugier CM, Trama J, Zhao C, Zheng H, Goldberg GS. Effects of cadherin mediated contact normalization on oncogenic Src kinase mediated gene expression and protein phosphorylation. Sci Rep 2024; 14:23942. [PMID: 39397108 PMCID: PMC11471763 DOI: 10.1038/s41598-024-75449-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 10/04/2024] [Indexed: 10/15/2024] Open
Abstract
Nontransformed cells form heterotypic cadherin junctions with adjacent transformed cells to inhibit tumor cell growth and motility. Transformed cells must override this form of growth control, called "contact normalization", to invade and metastasize during cancer progression. Heterocellular cadherin junctions between transformed and nontransformed cells are needed for this process. However, specific mechanisms downstream of cadherin signaling have not been clearly elucidated. Here, we utilized a β-catenin reporter construct to determine if contact normalization affects Wnt signaling in transformed cells. β-catenin driven GFP expression in Src transformed mouse embryonic cells was decreased when cultured with cadherin competent nontransformed cells compared to transformed cells cultured with themselves, but not when cultured with cadherin deficient nontransformed cells. We also utilized a layered culture system to investigate the effects of oncogenic transformation and contact normalization on gene expression and oncogenic Src kinase mediated phosphorylation events. RNA-Seq analysis found that cadherin dependent contact normalization inhibited the expression of 22 transcripts that were induced by Src transformation, and increased the expression of 78 transcripts that were suppressed by Src transformation. Phosphoproteomic analysis of cells expressing a temperature sensitive Src kinase construct found that contact normalization decreased phosphorylation of 10 proteins on tyrosine residues that were phosphorylated within 1 h of Src kinase activation in transformed cells. Taken together, these results indicate that cadherin dependent contact normalization inhibits Wnt signaling to regulate oncogenic kinase activity and gene expression, particularly PDPN expression, in transformed cells in order to control tumor progression.
Collapse
Affiliation(s)
- Rachel E Nicoletto
- Rowan-Virtua School of Osteopathic Medicine, Rowan University, B330 Science Center, 2 Medical Center Dr., Stratford, NJ, 08084, USA
| | - Cayla J Holdcraft
- Rowan-Virtua School of Osteopathic Medicine, Rowan University, B330 Science Center, 2 Medical Center Dr., Stratford, NJ, 08084, USA
| | - Ariel C Yin
- Rowan-Virtua School of Osteopathic Medicine, Rowan University, B330 Science Center, 2 Medical Center Dr., Stratford, NJ, 08084, USA
| | - Edward P Retzbach
- Rowan-Virtua School of Osteopathic Medicine, Rowan University, B330 Science Center, 2 Medical Center Dr., Stratford, NJ, 08084, USA
| | - Stephanie A Sheehan
- Rowan-Virtua School of Osteopathic Medicine, Rowan University, B330 Science Center, 2 Medical Center Dr., Stratford, NJ, 08084, USA
| | - Amanda A Greenspan
- Rowan-Virtua School of Osteopathic Medicine, Rowan University, B330 Science Center, 2 Medical Center Dr., Stratford, NJ, 08084, USA
| | - Christopher M Laugier
- Rowan-Virtua School of Osteopathic Medicine, Rowan University, B330 Science Center, 2 Medical Center Dr., Stratford, NJ, 08084, USA
| | - Jason Trama
- Medical Diagnostic Laboratories, 2439 Kuser Rd, Hamilton Township, NJ, 08690, USA
| | - Caifeng Zhao
- Biological Mass Spectrometry Resources, Robert Wood Johnson Medical School, Rutgers University, 174 Frelinghuysen Road, Piscataway, NJ, 08854, USA
| | - Haiyan Zheng
- Biological Mass Spectrometry Resources, Robert Wood Johnson Medical School, Rutgers University, 174 Frelinghuysen Road, Piscataway, NJ, 08854, USA
| | - Gary S Goldberg
- Rowan-Virtua School of Osteopathic Medicine, Rowan University, B330 Science Center, 2 Medical Center Dr., Stratford, NJ, 08084, USA.
| |
Collapse
|
9
|
Jheng JR, Bai Y, Noda K, Huot JR, Cook T, Fisher A, Chen YY, Goncharov DA, Goncharova EA, Simon MA, Zhang Y, Forman DE, Rojas M, Machado RF, Auwerx J, Gladwin MT, Lai YC. Skeletal Muscle SIRT3 Deficiency Contributes to Pulmonary Vascular Remodeling in Pulmonary Hypertension Due to Heart Failure With Preserved Ejection Fraction. Circulation 2024; 150:867-883. [PMID: 38804138 PMCID: PMC11384544 DOI: 10.1161/circulationaha.124.068624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 04/30/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND Pulmonary hypertension (PH) is a major complication linked to adverse outcomes in heart failure with preserved ejection fraction (HFpEF), yet no specific therapies exist for PH associated with HFpEF (PH-HFpEF). We have recently reported on the role of skeletal muscle SIRT3 (sirtuin-3) in modulation of PH-HFpEF, suggesting a novel endocrine signaling pathway for skeletal muscle modulation of pulmonary vascular remodeling. METHODS Using skeletal muscle-specific Sirt3 knockout mice (Sirt3skm-/-) and mass spectrometry-based comparative secretome analysis, we attempted to define the processes by which skeletal muscle SIRT3 defects affect pulmonary vascular health in PH-HFpEF. RESULTS Sirt3skm-/- mice exhibited reduced pulmonary vascular density accompanied by pulmonary vascular proliferative remodeling and elevated pulmonary pressures. Comparative analysis of secretome by mass spectrometry revealed elevated secretion levels of LOXL2 (lysyl oxidase homolog 2) in SIRT3-deficient skeletal muscle cells. Elevated circulation and protein expression levels of LOXL2 were also observed in plasma and skeletal muscle of Sirt3skm-/- mice, a rat model of PH-HFpEF, and humans with PH-HFpEF. In addition, expression levels of CNPY2 (canopy fibroblast growth factor signaling regulator 2), a known proliferative and angiogenic factor, were increased in pulmonary artery endothelial cells and pulmonary artery smooth muscle cells of Sirt3skm-/- mice and animal models of PH-HFpEF. CNPY2 levels were also higher in pulmonary artery smooth muscle cells of subjects with obesity compared with nonobese subjects. Moreover, treatment with recombinant LOXL2 protein promoted pulmonary artery endothelial cell migration/proliferation and pulmonary artery smooth muscle cell proliferation through regulation of CNPY2-p53 signaling. Last, skeletal muscle-specific Loxl2 deletion decreased pulmonary artery endothelial cell and pulmonary artery smooth muscle cell expression of CNPY2 and improved pulmonary pressures in mice with high-fat diet-induced PH-HFpEF. CONCLUSIONS This study demonstrates a systemic pathogenic impact of skeletal muscle SIRT3 deficiency in remote pulmonary vascular remodeling and PH-HFpEF. This study suggests a new endocrine signaling axis that links skeletal muscle health and SIRT3 deficiency to remote CNPY2 regulation in the pulmonary vasculature through myokine LOXL2. Our data also identify skeletal muscle SIRT3, myokine LOXL2, and CNPY2 as potential targets for the treatment of PH-HFpEF.
Collapse
MESH Headings
- Animals
- Sirtuin 3/metabolism
- Sirtuin 3/deficiency
- Sirtuin 3/genetics
- Heart Failure/metabolism
- Heart Failure/physiopathology
- Heart Failure/genetics
- Heart Failure/pathology
- Heart Failure/etiology
- Vascular Remodeling
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/physiopathology
- Hypertension, Pulmonary/etiology
- Hypertension, Pulmonary/genetics
- Hypertension, Pulmonary/pathology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscle, Skeletal/physiopathology
- Mice, Knockout
- Mice
- Humans
- Stroke Volume
- Male
- Rats
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Pulmonary Artery/physiopathology
- Disease Models, Animal
- Female
Collapse
Affiliation(s)
- Jia-Rong Jheng
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine (J.-R.J., Y.B., T.C., A.F., R.F.M., Y.-C.L.), Indiana University School of Medicine, Indianapolis
| | - Yang Bai
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine (J.-R.J., Y.B., T.C., A.F., R.F.M., Y.-C.L.), Indiana University School of Medicine, Indianapolis
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang (Y.B.)
| | - Kentaro Noda
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center, PA (K.N.)
| | - Joshua R Huot
- Department of Anatomy, Cell Biology and Physiology (J.R.H., R.F.M., Y.-C.L.), Indiana University School of Medicine, Indianapolis
| | - Todd Cook
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine (J.-R.J., Y.B., T.C., A.F., R.F.M., Y.-C.L.), Indiana University School of Medicine, Indianapolis
| | - Amanda Fisher
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine (J.-R.J., Y.B., T.C., A.F., R.F.M., Y.-C.L.), Indiana University School of Medicine, Indianapolis
| | - Yi-Yun Chen
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan (Y.-Y.C.)
| | - Dmitry A Goncharov
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California, Davis (D.A.G., E.A.G.)
| | - Elena A Goncharova
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California, Davis (D.A.G., E.A.G.)
| | - Marc A Simon
- Division of Cardiology, University of California, San Francisco (M.A.S.)
| | - Yingze Zhang
- Division of Pulmonary, Allergy and Critical Care Medicine (Y.Z.), University of Pittsburgh, PA
| | - Daniel E Forman
- Department of Medicine, Divisions of Geriatrics and Cardiology (D.E.F.), University of Pittsburgh, PA
- Geriatric Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, PA (D.E.F.)
| | - Mauricio Rojas
- Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University, Columbus (M.R.)
| | - Roberto F Machado
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine (J.-R.J., Y.B., T.C., A.F., R.F.M., Y.-C.L.), Indiana University School of Medicine, Indianapolis
- Department of Anatomy, Cell Biology and Physiology (J.R.H., R.F.M., Y.-C.L.), Indiana University School of Medicine, Indianapolis
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, Ecole Polytechnique Fédérale de Lausanne, Switzerland (J.A.)
| | - Mark T Gladwin
- Department of Medicine, University of Maryland, Baltimore (M.T.G.)
| | - Yen-Chun Lai
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine (J.-R.J., Y.B., T.C., A.F., R.F.M., Y.-C.L.), Indiana University School of Medicine, Indianapolis
- Department of Anatomy, Cell Biology and Physiology (J.R.H., R.F.M., Y.-C.L.), Indiana University School of Medicine, Indianapolis
| |
Collapse
|
10
|
Post Y, Lu C, Fletcher RB, Yeh WC, Nguyen H, Lee SJ, Li Y. Design principles and therapeutic applications of novel synthetic WNT signaling agonists. iScience 2024; 27:109938. [PMID: 38832011 PMCID: PMC11145361 DOI: 10.1016/j.isci.2024.109938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024] Open
Abstract
Wingless-related integration site or Wingless and Int-1 or Wingless-Int (WNT) signaling is crucial for embryonic development, and adult tissue homeostasis and regeneration, through its essential roles in cell fate, patterning, and stem cell regulation. The biophysical characteristics of WNT ligands have hindered efforts to interrogate ligand activity in vivo and prevented their development as therapeutics. Recent breakthroughs have enabled the generation of synthetic WNT signaling molecules that possess characteristics of natural ligands and potently activate the pathway, while also providing distinct advantages for therapeutic development and manufacturing. This review provides a detailed discussion of the protein engineering of these molecular platforms for WNT signaling agonism. We discuss the importance of WNT signaling in several organs and share insights from the initial application of these new classes of molecules in vitro and in vivo. These molecules offer a unique opportunity to enhance our understanding of how WNT signaling agonism promotes tissue repair, enabling targeted development of tailored therapeutics.
Collapse
Affiliation(s)
- Yorick Post
- Surrozen, Inc., 171 Oyster Point Blvd, Suite 400, South San Francisco, CA 94080, USA
| | - Chenggang Lu
- Surrozen, Inc., 171 Oyster Point Blvd, Suite 400, South San Francisco, CA 94080, USA
| | - Russell B. Fletcher
- Surrozen, Inc., 171 Oyster Point Blvd, Suite 400, South San Francisco, CA 94080, USA
| | - Wen-Chen Yeh
- Surrozen, Inc., 171 Oyster Point Blvd, Suite 400, South San Francisco, CA 94080, USA
| | - Huy Nguyen
- Surrozen, Inc., 171 Oyster Point Blvd, Suite 400, South San Francisco, CA 94080, USA
| | - Sung-Jin Lee
- Surrozen, Inc., 171 Oyster Point Blvd, Suite 400, South San Francisco, CA 94080, USA
| | - Yang Li
- Surrozen, Inc., 171 Oyster Point Blvd, Suite 400, South San Francisco, CA 94080, USA
| |
Collapse
|
11
|
Roger I, Montero P, Milara J, Cortijo J. Pirfenidone and nintedanib attenuates pulmonary artery endothelial and smooth muscle cells transformations induced by IL-11. Eur J Pharmacol 2024; 972:176547. [PMID: 38561103 DOI: 10.1016/j.ejphar.2024.176547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 03/21/2024] [Accepted: 03/27/2024] [Indexed: 04/04/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) associated to pulmonary hypertension (PH) portends a poor prognosis, characterized by lung parenchyma fibrosis and pulmonary artery remodeling. Serum and parenchyma levels of Interleukin 11 (IL-11) are elevated in IPF-PH patients and contributes to pulmonary artery remodeling and PH. However, the effect of current approved therapies against IPF in pulmonary artery remodeling induced by IL-11 is unknown. The aim of this study is to analyze the effects of nintedanib and pirfenidone on pulmonary artery endothelial and smooth muscle cell remodeling induced by IL-11 in vitro. Our results show that nintedanib (NTD) and pirfenidone (PFD) ameliorates endothelial to mesenchymal transition (EnMT), pulmonary artery smooth muscle cell to myofibroblast-like transformation and pulmonary remodeling in precision lung cut slices. This study provided also evidence of the inhibitory effect of PFD and NTD on IL-11-induced endothelial and muscle cells proliferation and senescence. The inhibitory effect of these drugs on monocyte arrest and angiogenesis was also studied. Finally, we observed that IL-11 induced canonical signal transducer and activator of transcription 3 (STAT3) and non-canonical mitogen-activated protein kinase 1/2 (ERK1/2) phosphorylation, but, PFD and NTD only inhibited ERK1/2 phosphorylation. Therefore, this study provided evidence of the inhibitory effect of NTD and PFD on markers of pulmonary artery remodeling induced by IL-11.
Collapse
Affiliation(s)
- Inés Roger
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Health Institute Carlos III, 28029, Madrid, Spain; Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010, Valencia, Spain; Faculty of Health Sciences, Universidad Europea de Valencia, 46010, Valencia, Spain.
| | - Paula Montero
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Health Institute Carlos III, 28029, Madrid, Spain; Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010, Valencia, Spain; Faculty of Health Sciences, Universidad Europea de Valencia, 46010, Valencia, Spain
| | - Javier Milara
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Health Institute Carlos III, 28029, Madrid, Spain; Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010, Valencia, Spain; Pharmacy Unit, University General Hospital Consortium, 46014, Valencia, Spain
| | - Julio Cortijo
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Health Institute Carlos III, 28029, Madrid, Spain; Department of Pharmacology, Faculty of Medicine, University of Valencia, 46010, Valencia, Spain; Research and Teaching Unit, University General Hospital Consortium, 46014, Valencia, Spain
| |
Collapse
|
12
|
Koziol-White C, Gebski E, Cao G, Panettieri RA. Precision cut lung slices: an integrated ex vivo model for studying lung physiology, pharmacology, disease pathogenesis and drug discovery. Respir Res 2024; 25:231. [PMID: 38824592 PMCID: PMC11144351 DOI: 10.1186/s12931-024-02855-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 05/18/2024] [Indexed: 06/03/2024] Open
Abstract
Precision Cut Lung Slices (PCLS) have emerged as a sophisticated and physiologically relevant ex vivo model for studying the intricacies of lung diseases, including fibrosis, injury, repair, and host defense mechanisms. This innovative methodology presents a unique opportunity to bridge the gap between traditional in vitro cell cultures and in vivo animal models, offering researchers a more accurate representation of the intricate microenvironment of the lung. PCLS require the precise sectioning of lung tissue to maintain its structural and functional integrity. These thin slices serve as invaluable tools for various research endeavors, particularly in the realm of airway diseases. By providing a controlled microenvironment, precision-cut lung slices empower researchers to dissect and comprehend the multifaceted interactions and responses within lung tissue, thereby advancing our understanding of pulmonary pathophysiology.
Collapse
Affiliation(s)
- Cynthia Koziol-White
- Rutgers Institute for Translational Medicine and Science, The State University of NJ, 08901, Rutgers, New Brunswick, NJ, USA.
| | - Eric Gebski
- Rutgers Institute for Translational Medicine and Science, The State University of NJ, 08901, Rutgers, New Brunswick, NJ, USA
| | - Gaoyaun Cao
- Rutgers Institute for Translational Medicine and Science, The State University of NJ, 08901, Rutgers, New Brunswick, NJ, USA
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, The State University of NJ, 08901, Rutgers, New Brunswick, NJ, USA
| |
Collapse
|
13
|
Li P, Han X, Li J, Wang Y, Cao Y, Wu W, Liu X. Aerobic exercise training engages the canonical wnt pathway to improve pulmonary function and inflammation in COPD. BMC Pulm Med 2024; 24:236. [PMID: 38745304 PMCID: PMC11095004 DOI: 10.1186/s12890-024-03048-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 05/06/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND We studied whether the exercise improves cigarette smoke (CS) induced chronic obstructive pulmonary disease (COPD) in mice through inhibition of inflammation mediated by Wnt/β-catenin-peroxisome proliferator-activated receptor (PPAR) γ signaling. METHODS Firstly, we observed the effect of exercise on pulmonary inflammation, lung function, and Wnt/β-catenin-PPARγ. A total of 30 male C57BL/6J mice were divided into the control group (CG), smoke group (SG), low-intensity exercise group (LEG), moderate-intensity exercise group (MEG), and high-intensity exercise group (HEG). All the groups, except for CG, underwent whole-body progressive exposure to CS for 25 weeks. Then, we assessed the maximal exercise capacity of mice from the LEG, MEG, and HEG, and performed an 8-week treadmill exercise intervention. Then, we used LiCl (Wnt/β-catenin agonist) and XAV939 (Wnt/β-catenin antagonist) to investigate whether Wnt/β-catenin-PPARγ pathway played a role in the improvement of COPD via exercise. Male C57BL/6J mice were randomly divided into six groups (n = 6 per group): CG, SG, LiCl group, LiCl and exercise group, XAV939 group, and XAV939 and exercise group. Mice except those in the CG were exposed to CS, and those in the exercise groups were subjected to moderate-intensity exercise training. All the mice were subjected to lung function test, lung histological assessment, and analysis of inflammatory markers in the bronchoalveolar lavage fluid, as well as detection of Wnt1, β-catenin and PPARγ proteins in the lung tissue. RESULTS Exercise of various intensities alleviated lung structural changes, pulmonary function and inflammation in COPD, with moderate-intensity exercise exhibiting significant and comprehensive effects on the alleviation of pulmonary inflammation and improvement of lung function. Low-, moderate-, and high-intensity exercise decreased β-catenin levels and increased those of PPARγ significantly, and only moderate-intensity exercise reduced the level of Wnt1 protein. Moderate-intensity exercise relieved the inflammation aggravated by Wnt agonist. Wnt antagonist combined with moderate-intensity exercise increased the levels of PPARγ, which may explain the highest improvement of pulmonary function observed in this group. CONCLUSIONS Exercise effectively decreases COPD pulmonary inflammation and improves pulmonary function. The beneficial role of exercise may be exerted through Wnt/β-catenin-PPARγ pathway.
Collapse
Affiliation(s)
- Peijun Li
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China
| | - Xiaoyu Han
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai, 200438, P.R. China
| | - Jian Li
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai, 200438, P.R. China
- Faculty of Traditional Chinese Medicine, Naval Medical University (Second Military Medical University), Shanghai, 200433, P.R. China
| | - Yingqi Wang
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China
| | - Yuanyuan Cao
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai, 200438, P.R. China
| | - Weibing Wu
- Department of Sports Rehabilitation, Shanghai University of Sport, Shanghai, 200438, P.R. China.
| | - Xiaodan Liu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, P.R. China.
- Institute of Rehabilitation Medicine, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, P.R. China.
- Engineering Research Center of Traditional Chinese Medicine Intelligent Rehabilitation, Ministry of Education, Shanghai, 201203, P.R. China.
| |
Collapse
|
14
|
Burt M, Angelidou G, Mais CN, Preußer C, Glatter T, Heimerl T, Groß R, Serrania J, Boosarpu G, Pogge von Strandmann E, Müller JA, Bange G, Becker A, Lehmann M, Jonigk D, Neubert L, Freitag H, Paczia N, Schmeck B, Jung AL. Lipid A in outer membrane vesicles shields bacteria from polymyxins. J Extracell Vesicles 2024; 13:e12447. [PMID: 38766978 PMCID: PMC11103557 DOI: 10.1002/jev2.12447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/18/2024] [Accepted: 04/23/2024] [Indexed: 05/22/2024] Open
Abstract
The continuous emergence of multidrug-resistant bacterial pathogens poses a major global healthcare challenge, with Klebsiella pneumoniae being a prominent threat. We conducted a comprehensive study on K. pneumoniae's antibiotic resistance mechanisms, focusing on outer membrane vesicles (OMVs) and polymyxin, a last-resort antibiotic. Our research demonstrates that OMVs protect bacteria from polymyxins. OMVs derived from Polymyxin B (PB)-stressed K. pneumoniae exhibited heightened protective efficacy due to increased vesiculation, compared to OMVs from unstressed Klebsiella. OMVs also shield bacteria from different bacterial families. This was validated ex vivo and in vivo using precision cut lung slices (PCLS) and Galleria mellonella. In all models, OMVs protected K. pneumoniae from PB and reduced the associated stress response on protein level. We observed significant changes in the lipid composition of OMVs upon PB treatment, affecting their binding capacity to PB. The altered binding capacity of single OMVs from PB stressed K. pneumoniae could be linked to a reduction in the lipid A amount of their released vesicles. Although the amount of lipid A per vesicle is reduced, the overall increase in the number of vesicles results in an increased protection because the sum of lipid A and therefore PB binding sites have increased. This unravels the mechanism of the altered PB protective efficacy of OMVs from PB stressed K. pneumoniae compared to control OMVs. The lipid A-dependent protective effect against PB was confirmed in vitro using artificial vesicles. Moreover, artificial vesicles successfully protected Klebsiella from PB ex vivo and in vivo. The findings indicate that OMVs act as protective shields for bacteria by binding to polymyxins, effectively serving as decoys and preventing antibiotic interaction with the cell surface. Our findings provide valuable insights into the mechanisms underlying antibiotic cross-protection and offer potential avenues for the development of novel therapeutic interventions to address the escalating threat of multidrug-resistant bacterial infections.
Collapse
Affiliation(s)
- Marie Burt
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL)Philipps‐University MarburgMarburgGermany
| | - Georgia Angelidou
- Core Facility for Metabolomics and Small Molecules Mass SpectrometryMax Planck Institute for Terrestrial MicrobiologyMarburgGermany
- Core Facility for Mass Spectrometry and ProteomicsMax Planck Institute for terrestrial MicrobiologyMarburgGermany
| | - Christopher Nils Mais
- Center for Synthetic Microbiology (SYNMIKRO)Philipps‐University MarburgMarburgGermany
| | - Christian Preußer
- Institute for Tumor ImmunologyPhilipps‐University MarburgMarburgGermany
- Core Facility ‐ Extracellular VesiclesPhilipps‐University MarburgMarburgGermany
| | - Timo Glatter
- Core Facility for Mass Spectrometry and ProteomicsMax Planck Institute for terrestrial MicrobiologyMarburgGermany
| | - Thomas Heimerl
- Center for Synthetic Microbiology (SYNMIKRO)Philipps‐University MarburgMarburgGermany
| | - Rüdiger Groß
- Institute of Molecular VirologyUlm University Medical CenterUlmGermany
| | - Javier Serrania
- Center for Synthetic Microbiology (SYNMIKRO)Philipps‐University MarburgMarburgGermany
| | - Gowtham Boosarpu
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL)Philipps‐University MarburgMarburgGermany
| | - Elke Pogge von Strandmann
- Institute for Tumor ImmunologyPhilipps‐University MarburgMarburgGermany
- Core Facility ‐ Extracellular VesiclesPhilipps‐University MarburgMarburgGermany
| | - Janis A. Müller
- Institute of VirologyPhilipps‐University MarburgMarburgGermany
| | - Gert Bange
- Center for Synthetic Microbiology (SYNMIKRO)Philipps‐University MarburgMarburgGermany
| | - Anke Becker
- Center for Synthetic Microbiology (SYNMIKRO)Philipps‐University MarburgMarburgGermany
| | - Mareike Lehmann
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL)Philipps‐University MarburgMarburgGermany
- Comprehensive Pneumology Center (CPC), Institute of Lung Health and ImmunityHelmholtz Zentrum MünchenGerman Center for Lung Research (DZL)MunichGermany
- Institute for Lung Health (ILH)GiessenGermany
| | - Danny Jonigk
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH)German Center of Lung Research (DZL)HannoverGermany
- Institute of PathologyUniversity Medical Center RWTH University of AachenAachenGermany
| | - Lavinia Neubert
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH)German Center of Lung Research (DZL)HannoverGermany
- Institute of PathologyHannover Medical SchoolHannoverGermany
| | - Hinrich Freitag
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH)German Center of Lung Research (DZL)HannoverGermany
- Institute of PathologyHannover Medical SchoolHannoverGermany
| | - Nicole Paczia
- Core Facility for Metabolomics and Small Molecules Mass SpectrometryMax Planck Institute for Terrestrial MicrobiologyMarburgGermany
| | - Bernd Schmeck
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL)Philipps‐University MarburgMarburgGermany
- Center for Synthetic Microbiology (SYNMIKRO)Philipps‐University MarburgMarburgGermany
- Institute for Lung Health (ILH)GiessenGermany
- Department of Medicine, Pulmonary and Critical Care MedicineUniversity Medical Center MarburgUniversities of Giessen and Marburg Lung CenterPhilipps‐University MarburgMarburgGermany
- Member of the German Center for Infectious Disease Research (DZIF)MarburgGermany
- Core Facility Flow Cytometry – Bacterial VesiclesPhilipps‐University MarburgMarburgGermany
| | - Anna Lena Jung
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL)Philipps‐University MarburgMarburgGermany
- Core Facility Flow Cytometry – Bacterial VesiclesPhilipps‐University MarburgMarburgGermany
| |
Collapse
|
15
|
Basil MC, Alysandratos KD, Kotton DN, Morrisey EE. Lung repair and regeneration: Advanced models and insights into human disease. Cell Stem Cell 2024; 31:439-454. [PMID: 38492572 PMCID: PMC11070171 DOI: 10.1016/j.stem.2024.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/07/2024] [Accepted: 02/22/2024] [Indexed: 03/18/2024]
Abstract
The respiratory system acts as both the primary site of gas exchange and an important sensor and barrier to the external environment. The increase in incidences of respiratory disease over the past decades has highlighted the importance of developing improved therapeutic approaches. This review will summarize recent research on the cellular complexity of the mammalian respiratory system with a focus on gas exchange and immunological defense functions of the lung. Different models of repair and regeneration will be discussed to help interpret human and animal data and spur the investigation of models and assays for future drug development.
Collapse
Affiliation(s)
- Maria C Basil
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn, Children's Hospital of Philadelphia (CHOP) Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Konstantinos-Dionysios Alysandratos
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA.
| | - Darrell N Kotton
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA.
| | - Edward E Morrisey
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn, Children's Hospital of Philadelphia (CHOP) Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
16
|
Delgado-Ortiz L, Ranciati S, Arbillaga-Etxarri A, Balcells E, Buekers J, Demeyer H, Frei A, Gimeno-Santos E, Hopkinson NS, de Jong C, Karlsson N, Louvaris Z, Palmerini L, Polkey MI, Puhan MA, Rabinovich RA, Rodríguez Chiaradia DA, Rodriguez-Roisin R, Toran-Montserrat P, Vogiatzis I, Watz H, Troosters T, Garcia-Aymerich J. Real-world walking cadence in people with COPD. ERJ Open Res 2024; 10:00673-2023. [PMID: 38444656 PMCID: PMC10910309 DOI: 10.1183/23120541.00673-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 12/05/2023] [Indexed: 03/07/2024] Open
Abstract
Introduction The clinical validity of real-world walking cadence in people with COPD is unsettled. Our objective was to assess the levels, variability and association with clinically relevant COPD characteristics and outcomes of real-world walking cadence. Methods We assessed walking cadence (steps per minute during walking bouts longer than 10 s) from 7 days' accelerometer data in 593 individuals with COPD from five European countries, and clinical and functional characteristics from validated questionnaires and standardised tests. Severe exacerbations during a 12-month follow-up were recorded from patient reports and medical registries. Results Participants were mostly male (80%) and had mean±sd age of 68±8 years, post-bronchodilator forced expiratory volume in 1 s (FEV1) of 57±19% predicted and walked 6880±3926 steps·day-1. Mean walking cadence was 88±9 steps·min-1, followed a normal distribution and was highly stable within-person (intraclass correlation coefficient 0.92, 95% CI 0.90-0.93). After adjusting for age, sex, height and number of walking bouts in fractional polynomial or linear regressions, walking cadence was positively associated with FEV1, 6-min walk distance, physical activity (steps·day-1, time in moderate-to-vigorous physical activity, vector magnitude units, walking time, intensity during locomotion), physical activity experience and health-related quality of life and negatively associated with breathlessness and depression (all p<0.05). These associations remained after further adjustment for daily steps. In negative binomial regression adjusted for multiple confounders, walking cadence related to lower number of severe exacerbations during follow-up (incidence rate ratio 0.94 per step·min-1, 95% CI 0.91-0.99, p=0.009). Conclusions Higher real-world walking cadence is associated with better COPD status and lower severe exacerbations risk, which makes it attractive as a future prognostic marker and clinical outcome.
Collapse
Affiliation(s)
- Laura Delgado-Ortiz
- ISGlobal, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Saverio Ranciati
- Department of Statistical Sciences, Università di Bologna, Bologna, Italy
| | - Ane Arbillaga-Etxarri
- Deusto Physical TherapIker, Physical Therapy Department, Faculty of Health Sciences, University of Deusto, San Sebastian, Spain
| | - Eva Balcells
- Universitat Pompeu Fabra, Barcelona, Spain
- Respiratory Medicine Department, Hospital del Mar, Barcelona, Spain
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Instituto de Salud Carlos III, Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Joren Buekers
- ISGlobal, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Heleen Demeyer
- Department of Rehabilitation Sciences, KULeuven, Leuven, Belgium
- Department of Rehabilitation Sciences, Ghent University, Ghent, Belgium
| | - Anja Frei
- Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - Elena Gimeno-Santos
- ISGlobal, Barcelona, Spain
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Instituto de Salud Carlos III, Barcelona, Spain
- Hospital Clínic de Barcelona, Barcelona, Spain
| | | | - Corina de Jong
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | | | - Luca Palmerini
- Department of Electrical, Electronic, and Information Engineering “Guglielmo Marconi”, Università di Bologna, Bologna, Italy
| | - Michael I. Polkey
- National Heart and Lung Institute, Imperial College London, London, UK
- Respiratory Medicine, Royal Brompton and Harefield NHS Foundation Trust, London, UK
| | - Milo A. Puhan
- Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - Roberto A. Rabinovich
- Respiratory Medicine Department, Royal Infirmary of Edinburgh, Centre for Inflammation Research, QMRI, The University of Edinburgh, Scotland, UK
| | - Diego A. Rodríguez Chiaradia
- Respiratory Medicine Department, Hospital del Mar, Barcelona, Spain
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Instituto de Salud Carlos III, Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Robert Rodriguez-Roisin
- Biomedical Research Networking Centre on Respiratory Diseases (CIBERES), Instituto de Salud Carlos III, Barcelona, Spain
- Universitat de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clínic of Barcelona, Barcelona, Spain
| | - Pere Toran-Montserrat
- Unitat de Suport a la Recerca Metropolitana Nord, Institut Universitari d'Investigació en Atenció Primària Jordi Gol, Mataró, Spain
- Germans Trias i Pujol Research Institute, Badalona, Spain
| | - Ioannis Vogiatzis
- Department of Sport, Exercise and Rehabilitation, Northumbria University Newcastle, Newcastle upon Tyne, UK
| | - Henrik Watz
- Pulmonary Research Institute at Lungen Clinic Grosshansdorf, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Grosshansdorf, Germany
| | | | - Judith Garcia-Aymerich
- ISGlobal, Barcelona, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| |
Collapse
|
17
|
Blomberg R, Sompel K, Hauer C, Smith AJ, Peña B, Driscoll J, Hume PS, Merrick DT, Tennis MA, Magin CM. Hydrogel-Embedded Precision-Cut Lung Slices Model Lung Cancer Premalignancy Ex Vivo. Adv Healthc Mater 2024; 13:e2302246. [PMID: 37953708 PMCID: PMC10872976 DOI: 10.1002/adhm.202302246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/17/2023] [Indexed: 11/14/2023]
Abstract
Lung cancer is the leading global cause of cancer-related deaths. Although smoking cessation is the best prevention, 50% of lung cancer diagnoses occur in people who have quit smoking. Research into treatment options for high-risk patients is constrained to rodent models, which are time-consuming, expensive, and require large cohorts. Embedding precision-cut lung slices (PCLS) within an engineered hydrogel and exposing this tissue to vinyl carbamate, a carcinogen from cigarette smoke, creates an in vitro model of lung cancer premalignancy. Hydrogel formulations are selected to promote early lung cancer cellular phenotypes and extend PCLS viability to six weeks. Hydrogel-embedded PCLS are exposed to vinyl carbamate, which induces adenocarcinoma in mice. Analysis of proliferation, gene expression, histology, tissue stiffness, and cellular content after six weeks reveals that vinyl carbamate induces premalignant lesions with a mixed adenoma/squamous phenotype. Putative chemoprevention agents diffuse through the hydrogel and induce tissue-level changes. The design parameters selected using murine tissue are validated with hydrogel-embedded human PCLS and results show increased proliferation and premalignant lesion gene expression patterns. This tissue-engineered model of human lung cancer premalignancy is the foundation for more sophisticated ex vivo models that enable the study of carcinogenesis and chemoprevention strategies.
Collapse
Affiliation(s)
- Rachel Blomberg
- Department of Bioengineering, University of Colorado, Denver |Anschutz, Aurora, CO, 80045, USA
| | - Kayla Sompel
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Caroline Hauer
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Alex J Smith
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Brisa Peña
- Department of Bioengineering, University of Colorado, Denver |Anschutz, Aurora, CO, 80045, USA
- Cardiovascular Institute & Adult Medical Genetics, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Jennifer Driscoll
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, CO, 80206, USA
| | - Patrick S Hume
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, CO, 80206, USA
| | - Daniel T Merrick
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Meredith A Tennis
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Chelsea M Magin
- Department of Bioengineering, University of Colorado, Denver |Anschutz, Aurora, CO, 80045, USA
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| |
Collapse
|
18
|
Lin L, Yang L, Wang N, Chen S, Du X, Chen R, Zhang H, Kong X. FGF10 protects against LPS-induced epithelial barrier injury and inflammation by inhibiting SIRT1-ferroptosis pathway in acute lung injury in mice. Int Immunopharmacol 2024; 127:111426. [PMID: 38147776 DOI: 10.1016/j.intimp.2023.111426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 12/14/2023] [Accepted: 12/18/2023] [Indexed: 12/28/2023]
Abstract
Pulmonary alveolar epithelial cell injury is considered the main pathological and physiological change in acute lung injury. Ferroptosis in alveolar epithelial cells is one of crucial factors contributing to acute lung injury (ALI). Therefore, reducing ferroptosis and repair epithelial barrier is very necessary. More and more evidence suggested that FGF10 plays an important role in lung development and repair after injury. However, the relationship between FGF10 and ferroptosis remains unclear. This study aims to explore the regulatory role of FGF10 on ferroptosis in ALI. Differential gene expression analysis indicated that genes associated with ferroptosis showed that FGF10 can significantly alleviate LPS induced lung injury and epithelial barrier damage by decreasing levels of malonaldehyde(MDA), and lipid ROS. SIRT1 activator (Resveratrol) and inhibitor (EX527) are used in vivo showed that FGF10 protects ferroptosis of pulmonary epithelial cells through SIRT1 signal. Furthermore, knockdown of FGFR2 gene reduced the protective effect of FGF10 on acute lung injury in mice and SIRT1 activation. After the application of NRF2 inhibitor ML385 in vitro, the results showed that SIRT1 regulated the expression of ferroptosis related proteins NRF2, GPX4 and FTH1 are related to activation of NRF2. These data indicate that SIRT-ferroptosis was one of the critical mechanisms contributing to LPS-induced ALI. FGF10 is promising as a therapeutic candidate against ALI through inhibiting ferroptosis.
Collapse
Affiliation(s)
- Lidan Lin
- School of Basic Medical Sciences, Institute of Hypoxia Research, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Li Yang
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Nan Wang
- School of Basic Medical Sciences, Institute of Hypoxia Research, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Siyue Chen
- Department of Children's Respiration disease, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Xiaotong Du
- School of Pharmaceutical Sciences, Wenzhou Medical University, Zhejiang 315302, China
| | - Ran Chen
- School of Basic Medical Sciences, Institute of Hypoxia Research, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Hongyu Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Zhejiang 315302, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang 315302, China; Department of Pharmacy, Zhuji People's Hospital, Wenzhou Medical University, Zhuji, Shaoxing, Zhejiang, 311800, China
| | - Xiaoxia Kong
- School of Basic Medical Sciences, Institute of Hypoxia Research, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang 315302, China.
| |
Collapse
|
19
|
Kaasalainen M, Zhang R, Vashisth P, Birjandi AA, S'Ari M, Martella DA, Isaacs M, Mäkilä E, Wang C, Moldenhauer E, Clarke P, Pinna A, Zhang X, Mustfa SA, Caprettini V, Morrell AP, Gentleman E, Brauer DS, Addison O, Zhang X, Bergholt M, Al-Jamal K, Volponi AA, Salonen J, Hondow N, Sharpe P, Chiappini C. Lithiated porous silicon nanowires stimulate periodontal regeneration. Nat Commun 2024; 15:487. [PMID: 38216556 PMCID: PMC10786831 DOI: 10.1038/s41467-023-44581-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 12/20/2023] [Indexed: 01/14/2024] Open
Abstract
Periodontal disease is a significant burden for oral health, causing progressive and irreversible damage to the support structure of the tooth. This complex structure, the periodontium, is composed of interconnected soft and mineralised tissues, posing a challenge for regenerative approaches. Materials combining silicon and lithium are widely studied in periodontal regeneration, as they stimulate bone repair via silicic acid release while providing regenerative stimuli through lithium activation of the Wnt/β-catenin pathway. Yet, existing materials for combined lithium and silicon release have limited control over ion release amounts and kinetics. Porous silicon can provide controlled silicic acid release, inducing osteogenesis to support bone regeneration. Prelithiation, a strategy developed for battery technology, can introduce large, controllable amounts of lithium within porous silicon, but yields a highly reactive material, unsuitable for biomedicine. This work debuts a strategy to lithiate porous silicon nanowires (LipSiNs) which generates a biocompatible and bioresorbable material. LipSiNs incorporate lithium to between 1% and 40% of silicon content, releasing lithium and silicic acid in a tailorable fashion from days to weeks. LipSiNs combine osteogenic, cementogenic and Wnt/β-catenin stimuli to regenerate bone, cementum and periodontal ligament fibres in a murine periodontal defect.
Collapse
Affiliation(s)
- Martti Kaasalainen
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, UK
| | - Ran Zhang
- Department of Oral Pathology, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Priya Vashisth
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, UK
| | - Anahid Ahmadi Birjandi
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, UK
| | - Mark S'Ari
- School of Chemical and Process Engineering, University of Leeds, Leeds, LS2 9JT, UK
| | | | - Mark Isaacs
- Department of Chemistry, University College London, London, WC1H 0AJ, UK
- HarwellXPS, Research Complex at Harwell, Rutherford Appleton Labs, Didcot, OX11 0DE, UK
| | - Ermei Mäkilä
- Department of Physics and Astronomy, University of Turku, Turku, 20014, Finland
| | - Cong Wang
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, UK
| | - Evelin Moldenhauer
- Postnova Analytics GmbH, Rankinestr. 1, Landsberg am Lech, 86899, Germany
| | - Paul Clarke
- Postnova Analytics GmbH, Rankinestr. 1, Landsberg am Lech, 86899, Germany
| | - Alessandra Pinna
- Department of Materials, Imperial College London, London, SW72AZ, UK
- The Francis Crick Institute, London, NW11AT, UK
- School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Xuechen Zhang
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, UK
| | - Salman A Mustfa
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, UK
| | - Valeria Caprettini
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, UK
| | - Alexander P Morrell
- Centre for Oral Clinical & Translational Sciences, King's College London, London, SE1 9RT, UK
| | - Eileen Gentleman
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, UK
| | - Delia S Brauer
- Otto Schott Institute of Materials Research, Friedrich Schiller University Jena, Jena, 07743, Germany
| | - Owen Addison
- Centre for Oral Clinical & Translational Sciences, King's College London, London, SE1 9RT, UK
| | - Xuehui Zhang
- Department of Dental Materials & NMPA Key Laboratory for Dental Materials, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Mads Bergholt
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, UK
| | - Khuloud Al-Jamal
- Institute of Pharmaceutical Science, King's College London, London, SE1 9NH, UK
| | - Ana Angelova Volponi
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, UK
| | - Jarno Salonen
- Department of Physics and Astronomy, University of Turku, Turku, 20014, Finland
| | - Nicole Hondow
- School of Chemical and Process Engineering, University of Leeds, Leeds, LS2 9JT, UK
| | - Paul Sharpe
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, UK
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, 602 00, Czech Republic
| | - Ciro Chiappini
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, UK.
- London Centre for Nanotechnology, King's College London, London, WC2R 2LS, UK.
| |
Collapse
|
20
|
Hamdon S, Fernandez-Gonzalez P, Omar MY, González-Sepúlveda M, Ortiz J, Gil C. CHIR99021 causes inactivation of Tyrosine Hydroxylase and depletion of dopamine in rat brain striatum. Neuropharmacology 2024; 242:109759. [PMID: 37844866 DOI: 10.1016/j.neuropharm.2023.109759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/06/2023] [Accepted: 10/09/2023] [Indexed: 10/18/2023]
Abstract
CHIR99021, also known as laduviglusib or CT99021, is a Glycogen-synthase kinase 3β (GSK3β) inhibitor, which has been reported as a promising drug for cardiomyocyte regeneration or treatment of sensorial hearing loss. Since the activation of dopamine (DA) receptors regulates dopamine synthesis and they can signal through the β-arrestin pathway and GSK3β, we decided to check the effect of GSK3β inhibitors (CHIR99021, SB216763 and lithium ion) on the control of DA synthesis. Using ex vivo experiments with minces from rat brain striatum, we observed that CHIR99021, but not SB216763 or lithium, causes complete abrogation of both DA synthesis and accumulation, pointing to off-target effects of CHIR99021. This decrease can be attributed to tyrosine hydroxylase (TH) inhibition since the accumulation of l-DOPA in the presence of a DOPA decarboxylase inhibitor was similarly decreased. On the other hand, CHIR99021 caused a dramatic increase in the DOPAC/DA ratio, an indicator of DA metabolization, and hindered DA incorporation into striatum tissue. Tetrabenazine, an inhibitor of DA vesicular transport, also caused DA depletion and DOPAC/DA ratio increase to the same extent as CHIR99021. In addition, both CHIR99021 or SB216763, but not lithium, decreased TH phosphorylation in Ser19, but not in Ser31 or Ser40. These results demonstrate that CHIR99021 can lead to TH inactivation and DA depletion in brain striatum, opening the possibility of its use in DA-related disorders, and shows effects to be considered in future clinical trials. More work is needed to find the mechanism exerted by CHIR99021 on DA accumulation.
Collapse
Affiliation(s)
- Sally Hamdon
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Spain; Institut de Neurociències, Universitat Autònoma de Barcelona, Spain
| | - Pol Fernandez-Gonzalez
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Spain
| | - Muhammad Yusof Omar
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Spain; Institut de Neurociències, Universitat Autònoma de Barcelona, Spain
| | - Marta González-Sepúlveda
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute (VHIR) - Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), 08035, Barcelona, Spain; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Jordi Ortiz
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Spain; Institut de Neurociències, Universitat Autònoma de Barcelona, Spain; Centro Investigación Biomédica en Red de Salud Mental, CIBERSAM, and Translational Neuroscience Unit, Parc Taulí University Hospital and Universitat Autònoma de Barcelona, Spain
| | - Carles Gil
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Spain; Institut de Neurociències, Universitat Autònoma de Barcelona, Spain.
| |
Collapse
|
21
|
Diwan R, Bhatt HN, Beaven E, Nurunnabi M. Emerging delivery approaches for targeted pulmonary fibrosis treatment. Adv Drug Deliv Rev 2024; 204:115147. [PMID: 38065244 PMCID: PMC10787600 DOI: 10.1016/j.addr.2023.115147] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 11/02/2023] [Accepted: 11/29/2023] [Indexed: 01/01/2024]
Abstract
Pulmonary fibrosis (PF) is a progressive, and life-threatening interstitial lung disease which causes scarring in the lung parenchyma and thereby affects architecture and functioning of lung. It is an irreversible damage to lung functioning which is related to epithelial cell injury, immense accumulation of immune cells and inflammatory cytokines, and irregular recruitment of extracellular matrix. The inflammatory cytokines trigger the differentiation of fibroblasts into activated fibroblasts, also known as myofibroblasts, which further increase the production and deposition of collagen at the injury sites in the lung. Despite the significant morbidity and mortality associated with PF, there is no available treatment that efficiently and effectively treats the disease by reversing their underlying pathologies. In recent years, many therapeutic regimens, for instance, rho kinase inhibitors, Smad signaling pathway inhibitors, p38, BCL-xL/ BCL-2 and JNK pathway inhibitors, have been found to be potent and effective in treating PF, in preclinical stages. However, due to non-selectivity and non-specificity, the therapeutic molecules also result in toxicity mediated severe side effects. Hence, this review demonstrates recent advances on PF pathology, mechanism and targets related to PF, development of various drug delivery systems based on small molecules, RNAs, oligonucleotides, peptides, antibodies, exosomes, and stem cells for the treatment of PF and the progress of various therapeutic treatments in clinical trials to advance PF treatment.
Collapse
Affiliation(s)
- Rimpy Diwan
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX 79902, United States; Department of Biomedical Engineering, College of Engineering, The University of Texas El Paso, El Paso, TX 79968, United States
| | - Himanshu N Bhatt
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX 79902, United States; Department of Biomedical Engineering, College of Engineering, The University of Texas El Paso, El Paso, TX 79968, United States
| | - Elfa Beaven
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX 79902, United States; Department of Biomedical Engineering, College of Engineering, The University of Texas El Paso, El Paso, TX 79968, United States
| | - Md Nurunnabi
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX 79902, United States; Department of Biomedical Engineering, College of Engineering, The University of Texas El Paso, El Paso, TX 79968, United States; The Border Biomedical Research Center, The University of Texas El Paso, El Paso, TX 79968, United States.
| |
Collapse
|
22
|
Gvazava N, Konings SC, Cepeda-Prado E, Skoryk V, Umeano CH, Dong J, Silva IAN, Ottosson DR, Leigh ND, Wagner DE, Klementieva O. Label-Free
High-Resolution Photothermal Optical Infrared
Spectroscopy for Spatiotemporal Chemical Analysis in Fresh, Hydrated
Living Tissues and Embryos. J Am Chem Soc 2023; 145. [PMCID: PMC10655180 DOI: 10.1021/jacs.3c08854] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/11/2023] [Accepted: 10/11/2023] [Indexed: 06/24/2024]
Abstract
Label-free chemical imaging of living and functioning systems is the holy grail of biochemical research. However, existing techniques often require extensive sample preparation to remove interfering molecules such as water, rendering many molecular imaging techniques unsuitable for in situ structural studies. Here, we examined freshly extracted tissue biopsies and living small vertebrates at submicrometer resolution using optical photothermal infrared (O-PTIR) microspectroscopy and demonstrated the following major advances: (1) O-PTIR can be used for submicrometer structural analysis of unprocessed, fully hydrated tissue biopsies extracted from diverse organs, including living brain and lung tissues. (2) O-PTIR imaging can be performed on living organisms, such as salamander embryos, without compromising their further development. (3) Using O-PTIR, we tracked the structural changes of amyloids in functioning brain tissues over time, observing the appearance of newly formed amyloids for the first time. (4) Amyloid structures appeared altered following standard fixation and dehydration procedures. Thus, we demonstrate that O-PTIR enables time-resolved submicrometer in situ investigation of chemical and structural changes in diverse biomolecules in their native conditions, representing a technological breakthrough for in situ molecular imaging of biological samples.
Collapse
Affiliation(s)
- Nika Gvazava
- Department
of Experimental Medical Science, Lund University, 22180 Lund, Sweden
- NanoLund, Lund University, 22180 Lund, Sweden
- Lund
Stem Cell Center, Lund University, 22100 Lund, Sweden
- Wallenberg
Centre for Molecular Medicine, Lund University, 22184 Lund, Sweden
| | - Sabine C. Konings
- Department
of Experimental Medical Science, Lund University, 22180 Lund, Sweden
- MultiPark, Lund University, 22180 Lund, Sweden
- NanoLund, Lund University, 22180 Lund, Sweden
| | - Efrain Cepeda-Prado
- Department
of Experimental Medical Science, Lund University, 22180 Lund, Sweden
- MultiPark, Lund University, 22180 Lund, Sweden
- Lund
Stem Cell Center, Lund University, 22100 Lund, Sweden
| | - Valeriia Skoryk
- Department
of Experimental Medical Science, Lund University, 22180 Lund, Sweden
- MultiPark, Lund University, 22180 Lund, Sweden
- NanoLund, Lund University, 22180 Lund, Sweden
| | - Chimezie H. Umeano
- Department
of Laboratory Medicine, Molecular Medicine
and Gene Therapy, 22184 Lund, Sweden
- Lund
Stem Cell Center, Lund University, 22100 Lund, Sweden
- Wallenberg
Centre for Molecular Medicine, Lund University, 22184 Lund, Sweden
| | - Jiao Dong
- NanoLund, Lund University, 22180 Lund, Sweden
- Lund
Stem Cell Center, Lund University, 22100 Lund, Sweden
- Wallenberg
Centre for Molecular Medicine, Lund University, 22184 Lund, Sweden
| | - Iran A. N. Silva
- NanoLund, Lund University, 22180 Lund, Sweden
- Lund
Stem Cell Center, Lund University, 22100 Lund, Sweden
- Wallenberg
Centre for Molecular Medicine, Lund University, 22184 Lund, Sweden
| | - Daniella Rylander Ottosson
- Department
of Experimental Medical Science, Lund University, 22180 Lund, Sweden
- MultiPark, Lund University, 22180 Lund, Sweden
- Lund
Stem Cell Center, Lund University, 22100 Lund, Sweden
| | - Nicholas D. Leigh
- Department
of Laboratory Medicine, Molecular Medicine
and Gene Therapy, 22184 Lund, Sweden
- Lund
Stem Cell Center, Lund University, 22100 Lund, Sweden
- Wallenberg
Centre for Molecular Medicine, Lund University, 22184 Lund, Sweden
| | - Darcy Elizabeth Wagner
- Department
of Experimental Medical Science, Lund University, 22180 Lund, Sweden
- NanoLund, Lund University, 22180 Lund, Sweden
- Lund
Stem Cell Center, Lund University, 22100 Lund, Sweden
- Wallenberg
Centre for Molecular Medicine, Lund University, 22184 Lund, Sweden
| | - Oxana Klementieva
- Department
of Experimental Medical Science, Lund University, 22180 Lund, Sweden
- MultiPark, Lund University, 22180 Lund, Sweden
- NanoLund, Lund University, 22180 Lund, Sweden
| |
Collapse
|
23
|
Milara J, Roger I, Montero P, Artigues E, Escrivá J, Perez-Vizcaino F, Cortijo J. Targeting IL-11 system as a treatment of pulmonary arterial hypertension. Pharmacol Res 2023; 197:106985. [PMID: 37949331 DOI: 10.1016/j.phrs.2023.106985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/02/2023] [Accepted: 11/06/2023] [Indexed: 11/12/2023]
Abstract
IL-11 is linked to fibrotic diseases, but its role in pulmonary hypertension is unclear. We examined IL-11's involvement in idiopathic pulmonary arterial hypertension (iPAH). Using samples from control (n = 20) and iPAH (n = 6) subjects, we assessed IL-11 and IL-11Rα expression and localization through RT-qPCR, ELISA, immunohistochemistry, and immunofluorescence. A monocrotaline-induced PAH model helped evaluate the impact of siRNA-IL-11 on pulmonary artery remodeling and PH. The effects of recombinant human IL-11 and IL-11Rα on human pulmonary artery smooth muscle cell (HPASMC) proliferation, pulmonary artery endothelial cell (HPAEC) mesenchymal transition, monocyte interactions, endothelial tube formation, and precision cut lung slice (PCLS) pulmonary artery remodeling and contraction were evaluated. IL-11 and IL-11Rα were over-expressed in pulmonary arteries (3.2-fold and 75-fold respectively) and serum (1.5-fold and 2-fold respectively) of patients with iPAH. Therapeutic transient transfection with siRNA targeting IL-11 resulted in a significant reduction in pulmonary artery remodeling (by 98%), right heart hypertrophy (by 66%), and pulmonary hypertension (by 58%) in rats exposed to monocrotaline treatment. rhIL-11 and soluble rhIL-11Rα induce HPASMC proliferation and HPAEC to monocyte interactions, mesenchymal transition, and tube formation. Neutralizing monoclonal IL-11 and IL-11Rα antibodies inhibited TGFβ1 and EDN-1 induced HPAEC to mesenchymal transition and HPASMC proliferation. In 3D PCLS, rhIL-11 and soluble rhIL-11Rα do not promote pulmonary artery contraction but sensitize PCLS pulmonary artery contraction induced by EDN-1. In summary, IL-11 and IL-11Rα are more highly expressed in the pulmonary arteries of iPAH patients and contribute to pulmonary artery remodeling and the development of PH.
Collapse
Affiliation(s)
- Javier Milara
- CIBER de enfermedades respiratorias, Health Institute Carlos III, Valencia, Spain; Department of Pharmacology, Faculty of Medicine, University of Valencia, Spain; Pharmacy Unit, University General Hospital Consortium of Valencia, Spain.
| | - Inés Roger
- CIBER de enfermedades respiratorias, Health Institute Carlos III, Valencia, Spain; Department of Pharmacology, Faculty of Medicine, University of Valencia, Spain
| | - Paula Montero
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Spain
| | - Enrique Artigues
- Surgery Unit, University General Hospital Consortium, Valencia, Spain
| | - Juan Escrivá
- Thoracic Surgery Unit, University and Polytechnic Hospital La Fe, Valencia, Spain
| | - Francisco Perez-Vizcaino
- CIBER de enfermedades respiratorias, Health Institute Carlos III, Valencia, Spain; Dept of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Julio Cortijo
- CIBER de enfermedades respiratorias, Health Institute Carlos III, Valencia, Spain; Department of Pharmacology, Faculty of Medicine, University of Valencia, Spain; Research and Teaching Unit, University General Hospital Consortium, Valencia, Spain
| |
Collapse
|
24
|
Kapellos TS, Conlon TM, Yildirim AÖ, Lehmann M. The impact of the immune system on lung injury and regeneration in COPD. Eur Respir J 2023; 62:2300589. [PMID: 37652569 DOI: 10.1183/13993003.00589-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 08/17/2023] [Indexed: 09/02/2023]
Abstract
COPD is a devastating respiratory condition that manifests via persistent inflammation, emphysema development and small airway remodelling. Lung regeneration is defined as the ability of the lung to repair itself after injury by the proliferation and differentiation of progenitor cell populations, and becomes impaired in the COPD lung as a consequence of cell intrinsic epithelial stem cell defects and signals from the micro-environment. Although the loss of structural integrity and lung regenerative capacity are critical for disease progression, our understanding of the cellular players and molecular pathways that hamper regeneration in COPD remains limited. Intriguingly, despite being a key driver of COPD pathogenesis, the role of the immune system in regulating lung regenerative mechanisms is understudied. In this review, we summarise recent evidence on the contribution of immune cells to lung injury and regeneration. We focus on four main axes: 1) the mechanisms via which myeloid cells cause alveolar degradation; 2) the formation of tertiary lymphoid structures and the production of autoreactive antibodies; 3) the consequences of inefficient apoptotic cell removal; and 4) the effects of innate and adaptive immune cell signalling on alveolar epithelial proliferation and differentiation. We finally provide insight on how recent technological advances in omics technologies and human ex vivo lung models can delineate immune cell-epithelium cross-talk and expedite precision pro-regenerative approaches toward reprogramming the alveolar immune niche to treat COPD.
Collapse
Affiliation(s)
- Theodore S Kapellos
- Comprehensive Pneumology Center, Institute of Lung Health and Immunity, Helmholtz Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Thomas M Conlon
- Comprehensive Pneumology Center, Institute of Lung Health and Immunity, Helmholtz Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Ali Önder Yildirim
- Comprehensive Pneumology Center, Institute of Lung Health and Immunity, Helmholtz Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
- Institute of Experimental Pneumology, University Hospital, Ludwig Maximilians University of Munich, Munich, Germany
| | - Mareike Lehmann
- Comprehensive Pneumology Center, Institute of Lung Health and Immunity, Helmholtz Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
- Institute for Lung Research, Philipps University of Marburg, Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research (DZL), Marburg, Germany
| |
Collapse
|
25
|
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
|
26
|
Sucre JM, Bock F, Negretti NM, Benjamin JT, Gulleman PM, Dong X, Ferguson KT, Jetter CS, Han W, Liu Y, Kook S, Gokey JJ, Guttentag SH, Kropski JA, Blackwell TS, Zent R, Plosa EJ. Alveolar repair following LPS-induced injury requires cell-ECM interactions. JCI Insight 2023; 8:e167211. [PMID: 37279065 PMCID: PMC10443799 DOI: 10.1172/jci.insight.167211] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 05/31/2023] [Indexed: 06/07/2023] Open
Abstract
During alveolar repair, alveolar type 2 (AT2) epithelial cell progenitors rapidly proliferate and differentiate into flat AT1 epithelial cells. Failure of normal alveolar repair mechanisms can lead to loss of alveolar structure (emphysema) or development of fibrosis, depending on the type and severity of injury. To test if β1-containing integrins are required during repair following acute injury, we administered E. coli lipopolysaccharide (LPS) by intratracheal injection to mice with a postdevelopmental deletion of β1 integrin in AT2 cells. While control mice recovered from LPS injury without structural abnormalities, β1-deficient mice had more severe inflammation and developed emphysema. In addition, recovering alveoli were repopulated with an abundance of rounded epithelial cells coexpressing AT2 epithelial, AT1 epithelial, and mixed intermediate cell state markers, with few mature type 1 cells. AT2 cells deficient in β1 showed persistently increased proliferation after injury, which was blocked by inhibiting NF-κB activation in these cells. Lineage tracing experiments revealed that β1-deficient AT2 cells failed to differentiate into mature AT1 epithelial cells. Together, these findings demonstrate that functional alveolar repair after injury with terminal alveolar epithelial differentiation requires β1-containing integrins.
Collapse
Affiliation(s)
- Jennifer M.S. Sucre
- Department of Pediatrics, Division of Neonatology
- Department of Cell and Developmental Biology
| | - Fabian Bock
- Department of Medicine, Division of Nephrology and Hypertension; and
| | | | | | | | - Xinyu Dong
- Department of Medicine, Division of Nephrology and Hypertension; and
| | | | | | - Wei Han
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Yang Liu
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Jason J. Gokey
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Jonathan A. Kropski
- Department of Cell and Developmental Biology
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Nashville Veterans Affairs Medical Center, Nashville, Tennessee, USA
| | - Timothy S. Blackwell
- Department of Cell and Developmental Biology
- Department of Medicine, Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Nashville Veterans Affairs Medical Center, Nashville, Tennessee, USA
| | - Roy Zent
- Department of Cell and Developmental Biology
- Department of Medicine, Division of Nephrology and Hypertension; and
- Nashville Veterans Affairs Medical Center, Nashville, Tennessee, USA
| | | |
Collapse
|
27
|
Saini M, Schmidleitner L, Moreno HD, Donato E, Falcone M, Bartsch JM, Klein C, Vogel V, Würth R, Pfarr N, Espinet E, Lehmann M, Königshoff M, Reitberger M, Haas S, Graf E, Schwarzmayr T, Strom TM, Spaich S, Sütterlin M, Schneeweiss A, Weichert W, Schotta G, Reichert M, Aceto N, Sprick MR, Trumpp A, Scheel CH. Resistance to mesenchymal reprogramming sustains clonal propagation in metastatic breast cancer. Cell Rep 2023; 42:112533. [PMID: 37257449 DOI: 10.1016/j.celrep.2023.112533] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 10/04/2022] [Accepted: 05/03/2023] [Indexed: 06/02/2023] Open
Abstract
The acquisition of mesenchymal traits is considered a hallmark of breast cancer progression. However, the functional relevance of epithelial-to-mesenchymal transition (EMT) remains controversial and context dependent. Here, we isolate epithelial and mesenchymal populations from human breast cancer metastatic biopsies and assess their functional potential in vivo. Strikingly, progressively decreasing epithelial cell adhesion molecule (EPCAM) levels correlate with declining disease propagation. Mechanistically, we find that persistent EPCAM expression marks epithelial clones that resist EMT induction and propagate competitively. In contrast, loss of EPCAM defines clones arrested in a mesenchymal state, with concomitant suppression of tumorigenicity and metastatic potential. This dichotomy results from distinct clonal trajectories impacting global epigenetic programs that are determined by the interplay between human ZEB1 and its target GRHL2. Collectively, our results indicate that susceptibility to irreversible EMT restrains clonal propagation, whereas resistance to mesenchymal reprogramming sustains disease spread in multiple models of human metastatic breast cancer, including patient-derived cells in vivo.
Collapse
Affiliation(s)
- Massimo Saini
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), and DKFZ-ZMBH Alliance, Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany; Institute of Stem Cell Research, Helmholtz Center Munich, Neuherberg, Germany; Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland.
| | - Laura Schmidleitner
- Institute of Stem Cell Research, Helmholtz Center Munich, Neuherberg, Germany; Translational Pancreatic Cancer Research Center, Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; Center for Functional Protein Assemblies (CPA), Technical University of Munich (TUM), Garching, Germany; Center for Organoid Systems (COS), Technical University of Munich (TUM), Garching, Germany; Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), Garching, Germany
| | - Helena Domínguez Moreno
- Division of Molecular Biology, Biomedical Center, Faculty of Medicine, Ludwig-Maximilian University of Munich (LMU), Munich, Germany
| | - Elisa Donato
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), and DKFZ-ZMBH Alliance, Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
| | - Mattia Falcone
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), and DKFZ-ZMBH Alliance, Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
| | - Johanna M Bartsch
- Institute of Stem Cell Research, Helmholtz Center Munich, Neuherberg, Germany
| | - Corinna Klein
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), and DKFZ-ZMBH Alliance, Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
| | - Vanessa Vogel
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), and DKFZ-ZMBH Alliance, Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
| | - Roberto Würth
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), and DKFZ-ZMBH Alliance, Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
| | - Nicole Pfarr
- Institute of Pathology, Technical University of Munich (TUM), Munich, Germany
| | - Elisa Espinet
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), and DKFZ-ZMBH Alliance, Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
| | - Mareike Lehmann
- Institute for Lung Health and Immunity (LHI) and Comprehensive Pneumology Center (CPC), Helmholtz Center Munich, Member of the German Center for Lung Research (DZL), Munich, Germany; Institute for Lung Research, Philipps-University Marburg, Member of the German Center for Lung Research (DZL), Marburg, Germany
| | - Melanie Königshoff
- Research Unit Lung Repair and Regeneration, Helmholtz Center Munich, Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Manuel Reitberger
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), and DKFZ-ZMBH Alliance, Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
| | - Simon Haas
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), and DKFZ-ZMBH Alliance, Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany; Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Elisabeth Graf
- Institute of Human Genetics, Helmholtz Center Munich, Neuherberg, Germany
| | - Thomas Schwarzmayr
- Institute of Human Genetics, Helmholtz Center Munich, Neuherberg, Germany
| | - Tim-Matthias Strom
- Institute of Human Genetics, Helmholtz Center Munich, Neuherberg, Germany
| | - Saskia Spaich
- Department of Gynaecology and Obstetrics, University Women's Clinic, University Medical Centre Mannheim, Mannheim, Germany
| | - Marc Sütterlin
- Department of Gynaecology and Obstetrics, University Women's Clinic, University Medical Centre Mannheim, Mannheim, Germany
| | - Andreas Schneeweiss
- National Center for Tumor Diseases, University Hospital and German Cancer Research Center, Heidelberg, Germany
| | - Wilko Weichert
- Institute of Pathology, Technical University of Munich (TUM), Munich, Germany; German Cancer Consortium (DKTK), Germany
| | - Gunnar Schotta
- Division of Molecular Biology, Biomedical Center, Faculty of Medicine, Ludwig-Maximilian University of Munich (LMU), Munich, Germany
| | - Maximilian Reichert
- Translational Pancreatic Cancer Research Center, Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany; Center for Functional Protein Assemblies (CPA), Technical University of Munich (TUM), Garching, Germany; Center for Organoid Systems (COS), Technical University of Munich (TUM), Garching, Germany; Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), Garching, Germany; German Cancer Consortium (DKTK), Germany
| | - Nicola Aceto
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Martin R Sprick
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), and DKFZ-ZMBH Alliance, Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany; German Cancer Consortium (DKTK), Germany.
| | - Andreas Trumpp
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), and DKFZ-ZMBH Alliance, Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany; German Cancer Consortium (DKTK), Germany.
| | - Christina H Scheel
- Institute of Stem Cell Research, Helmholtz Center Munich, Neuherberg, Germany.
| |
Collapse
|
28
|
Zhao J, Xia H, Wu Y, Lu L, Cheng C, Sun J, Xiang Q, Bian T, Liu Q. CircRNA_0026344 via miR-21 is involved in cigarette smoke-induced autophagy and apoptosis of alveolar epithelial cells in emphysema. Cell Biol Toxicol 2023; 39:929-944. [PMID: 34524572 DOI: 10.1007/s10565-021-09654-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 09/03/2021] [Indexed: 12/17/2022]
Abstract
Cigarette smoke (CS), a main source of indoor air pollution, is a primary risk factor for emphysema, and aberrant cellular autophagy is related to the pathogenesis of emphysema. Circular RNAs (circRNAs) affect the expression of mRNAs via acting as microRNA (miRNA) sponges, but their role in emphysema progression is not established. In the present investigation, CS, acting on alveolar epithelial cells, caused higher levels of miR-21, p-ERK, and cleaved-caspase 3 and led to lower levels of circRNA_0026344 and PTEN, which induced autophagy and apoptosis. miR-21 suppressed the expression of PTEN, which was involved in the regulation of autophagy and apoptosis. Further, in alveolar epithelial cells, overexpression of circRNA_0026344 blocked cigarette smoke extract (CSE)-induced autophagy and apoptosis, but this blockage was reversed by upregulation of miR-21 with a mimic. These results demonstrated that, in alveolar epithelial cells, CS decreases circRNA_0026344 levels, which sponge miR-21 to inhibit the miR-21 target, PTEN, which, in turn, activates ERK and thereby promotes autophagy and apoptosis, leading to emphysema. Thus, for emphysema, circRNA_0026344 regulates the PTEN/ERK axis by sponging miR-21, which is associated with the CS-induced autophagy and apoptosis of alveolar epithelial cells. In sum, the present investigation identifies a novel mechanism for CS-induced emphysema and provides information useful for the diagnosis and treatment of CS-induced emphysema.
Collapse
Affiliation(s)
- Jing Zhao
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
- China International Cooperation Center for Environment and Human Health, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Haibo Xia
- School of Public Health, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Yan Wu
- Department of Respiratory and Critical Care Medicine, Wuxi People's Hospital, Affiliated to Nanjing Medical University, Wuxi, 214023, Jiangsu, People's Republic of China
| | - Lu Lu
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
- China International Cooperation Center for Environment and Human Health, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Cheng Cheng
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
- China International Cooperation Center for Environment and Human Health, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Jing Sun
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
- China International Cooperation Center for Environment and Human Health, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China
| | - Quanyong Xiang
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Tao Bian
- Department of Respiratory and Critical Care Medicine, Wuxi People's Hospital, Affiliated to Nanjing Medical University, Wuxi, 214023, Jiangsu, People's Republic of China.
| | - Qizhan Liu
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China.
- China International Cooperation Center for Environment and Human Health, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China.
| |
Collapse
|
29
|
Siwczak F, Hiller C, Pfannkuche H, Schneider MR. Culture of vibrating microtome tissue slices as a 3D model in biomedical research. J Biol Eng 2023; 17:36. [PMID: 37264444 DOI: 10.1186/s13036-023-00357-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/21/2023] [Indexed: 06/03/2023] Open
Abstract
The basic idea behind the use of 3-dimensional (3D) tools in biomedical research is the assumption that the structures under study will perform at the best in vitro if cultivated in an environment that is as similar as possible to their natural in vivo embedding. Tissue slicing fulfills this premise optimally: it is an accessible, unexpensive, imaging-friendly, and technically rather simple procedure which largely preserves the extracellular matrix and includes all or at least most supportive cell types in the correct tissue architecture with little cellular damage. Vibrating microtomes (vibratomes) can further improve the quality of the generated slices because of the lateral, saw-like movement of the blade, which significantly reduces tissue pulling or tearing compared to a straight cut. In spite of its obvious advantages, vibrating microtome slices are rather underrepresented in the current discussion on 3D tools, which is dominated by methods as organoids, organ-on-chip and bioprinting. Here, we review the development of vibrating microtome tissue slices, the major technical features underlying its application, as well as its current use and potential advances, such as a combination with novel microfluidic culture chambers. Once fully integrated into the 3D toolbox, tissue slices may significantly contribute to decrease the use of laboratory animals and is likely to have a strong impact on basic and translational research as well as drug screening.
Collapse
Affiliation(s)
- Fatina Siwczak
- Institute of Veterinary Physiology, University of Leipzig, An den Tierkliniken 7, 04103, Leipzig, Germany
| | - Charlotte Hiller
- Institute of Veterinary Physiology, University of Leipzig, An den Tierkliniken 7, 04103, Leipzig, Germany
| | - Helga Pfannkuche
- Institute of Veterinary Physiology, University of Leipzig, An den Tierkliniken 7, 04103, Leipzig, Germany
| | - Marlon R Schneider
- Institute of Veterinary Physiology, University of Leipzig, An den Tierkliniken 7, 04103, Leipzig, Germany.
| |
Collapse
|
30
|
The Wnt/β-catenin pathway regulates inflammation and apoptosis in ventilator-induced lung injury. Biosci Rep 2023; 43:232596. [PMID: 36825682 PMCID: PMC10011329 DOI: 10.1042/bsr20222429] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 02/25/2023] Open
Abstract
Ventilator-induced lung injury (VILI) may be caused by incorrect mechanical ventilation (MV), and its progression is mainly related to inflammatory reaction, apoptosis, and oxidative stress. The Wnt/β-catenin pathway can modulate inflammation and apoptosis; however, its role in VILI is unknown. This research aims to explore the role of the Wnt/β-catenin pathway in VILI. VILI models were established using rats and type II alveolar epithelial (ATII) cells. Glycogen synthase kinase 3β (GSK-3β), β-catenin, and cyclin D1 were determined using western blotting and immunofluorescence. Apoptosis of lung tissues was evaluated using TUNEL, flow cytometry, Bax, and Bcl2 protein. Interleukin-1β (IL-1β), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α) were detected via enzyme-linked immunosorbent assay (ELISA). Lung pathological injury was evaluated through hematoxylin and eosin (H&E) staining. Lung permeability was evaluated by the ratio of dry to wet weight of lung tissue and the total protein level of bronchoalveolar lavage fluid (BALF). The results showed that GSK-3β expression was enhanced and β-catenin expression was diminished in lung tissue under MV. SB216763 increased β-catenin and cyclin D1 expression by inhibiting GSK-3β expression and inhibited the inflammatory response and apoptosis of lung, alleviated pulmonary edema and lung tissue permeability, and significantly mitigated lung injury. However, inhibition of β-catenin expression by MSAB attenuated the anti-inflammatory and antiapoptotic effects of SB216763 in VILI. Overall, the present study demonstrates that the Wnt/β-catenin pathway activation in MV may play an anti-inflammatory and antiapoptotic role, thereby alleviating lung injury and delaying VILI progression, which may be a key point of intervention in VILI.
Collapse
|
31
|
Blomberg R, Sompel K, Hauer C, Pe A B, Driscoll J, Hume PS, Merrick DT, Tennis MA, Magin CM. Tissue-engineered models of lung cancer premalignancy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.15.532835. [PMID: 36993773 PMCID: PMC10055140 DOI: 10.1101/2023.03.15.532835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Lung cancer is the leading global cause of cancer-related deaths. Although smoking cessation is the best preventive action, nearly 50% of all lung cancer diagnoses occur in people who have already quit smoking. Research into treatment options for these high-risk patients has been constrained to rodent models of chemical carcinogenesis, which are time-consuming, expensive, and require large numbers of animals. Here we show that embedding precision-cut lung slices within an engineered hydrogel and exposing this tissue to a carcinogen from cigarette smoke creates an in vitro model of lung cancer premalignancy. Hydrogel formulations were selected to promote early lung cancer cellular phenotypes and extend PCLS viability up to six weeks. In this study, hydrogel-embedded lung slices were exposed to the cigarette smoke derived carcinogen vinyl carbamate, which induces adenocarcinoma in mice. At six weeks, analysis of proliferation, gene expression, histology, tissue stiffness, and cellular content revealed that vinyl carbamate induced the formation of premalignant lesions with a mixed adenoma/squamous phenotype. Two putative chemoprevention agents were able to freely diffuse through the hydrogel and induce tissue-level changes. The design parameters selected using murine tissue were validated with hydrogel-embedded human PCLS and results showed increased proliferation and premalignant lesion gene expression patterns. This tissue-engineered model of human lung cancer premalignancy is the starting point for more sophisticated ex vivo models and a foundation for the study of carcinogenesis and chemoprevention strategies.
Collapse
|
32
|
Ahangari F, Price NL, Malik S, Chioccioli M, Bärnthaler T, Adams TS, Kim J, Pradeep SP, Ding S, Cosmos C, Rose KAS, McDonough JE, Aurelien NR, Ibarra G, Omote N, Schupp JC, DeIuliis G, Villalba Nunez JA, Sharma L, Ryu C, Dela Cruz CS, Liu X, Prasse A, Rosas I, Bahal R, Fernández-Hernando C, Kaminski N. microRNA-33 deficiency in macrophages enhances autophagy, improves mitochondrial homeostasis, and protects against lung fibrosis. JCI Insight 2023; 8:e158100. [PMID: 36626225 PMCID: PMC9977502 DOI: 10.1172/jci.insight.158100] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and ultimately fatal disease. Recent findings have shown a marked metabolic reprogramming associated with changes in mitochondrial homeostasis and autophagy during pulmonary fibrosis. The microRNA-33 (miR-33) family of microRNAs (miRNAs) encoded within the introns of sterol regulatory element binding protein (SREBP) genes are master regulators of sterol and fatty acid (FA) metabolism. miR-33 controls macrophage immunometabolic response and enhances mitochondrial biogenesis, FA oxidation, and cholesterol efflux. Here, we show that miR-33 levels are increased in bronchoalveolar lavage (BAL) cells isolated from patients with IPF compared with healthy controls. We demonstrate that specific genetic ablation of miR-33 in macrophages protects against bleomycin-induced pulmonary fibrosis. The absence of miR-33 in macrophages improves mitochondrial homeostasis and increases autophagy while decreasing inflammatory response after bleomycin injury. Notably, pharmacological inhibition of miR-33 in macrophages via administration of anti-miR-33 peptide nucleic acids (PNA-33) attenuates fibrosis in different in vivo and ex vivo mice and human models of pulmonary fibrosis. These studies elucidate a major role of miR-33 in macrophages in the regulation of pulmonary fibrosis and uncover a potentially novel therapeutic approach to treat this disease.
Collapse
Affiliation(s)
- Farida Ahangari
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Nathan L. Price
- Vascular Biology and Therapeutics Program, Yale Center for Molecular and System Metabolism, Department of Comparative Medicine, and Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - Shipra Malik
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut, USA
| | - Maurizio Chioccioli
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Thomas Bärnthaler
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Taylor S. Adams
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jooyoung Kim
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sai Pallavi Pradeep
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut, USA
| | - Shuizi Ding
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Carlos Cosmos
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Kadi-Ann S. Rose
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - John E. McDonough
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Nachelle R. Aurelien
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Internal Medicine, Weill Cornell Hospital Medicine, New York, New York, USA
| | - Gabriel Ibarra
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Life Span Medical Group, Department of Internal Medicine, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Norihito Omote
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jonas C. Schupp
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Giuseppe DeIuliis
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Julian A. Villalba Nunez
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lokesh Sharma
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Changwan Ryu
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Charles S. Dela Cruz
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Xinran Liu
- Center for Cellular and Molecular Imaging (CCMI), Department of Cell Biology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Antje Prasse
- Department of Pneumology, University of Hannover, Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Ivan Rosas
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Raman Bahal
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, Connecticut, USA
| | - Carlos Fernández-Hernando
- Vascular Biology and Therapeutics Program, Yale Center for Molecular and System Metabolism, Department of Comparative Medicine, and Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
33
|
Ahangari F, Becker C, Foster DG, Chioccioli M, Nelson M, Beke K, Wang X, Justet A, Adams T, Readhead B, Meador C, Correll K, Lili LN, Roybal HM, Rose KA, Ding S, Barnthaler T, Briones N, DeIuliis G, Schupp JC, Li Q, Omote N, Aschner Y, Sharma L, Kopf KW, Magnusson B, Hicks R, Backmark A, Dela Cruz CS, Rosas I, Cousens LP, Dudley JT, Kaminski N, Downey GP. Saracatinib, a Selective Src Kinase Inhibitor, Blocks Fibrotic Responses in Preclinical Models of Pulmonary Fibrosis. Am J Respir Crit Care Med 2022; 206:1463-1479. [PMID: 35998281 PMCID: PMC9757097 DOI: 10.1164/rccm.202010-3832oc] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/23/2022] [Indexed: 12/24/2022] Open
Abstract
Rationale: Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, and often fatal disorder. Two U.S. Food and Drug Administration-approved antifibrotic drugs, nintedanib and pirfenidone, slow the rate of decline in lung function, but responses are variable and side effects are common. Objectives: Using an in silico data-driven approach, we identified a robust connection between the transcriptomic perturbations in IPF disease and those induced by saracatinib, a selective Src kinase inhibitor originally developed for oncological indications. Based on these observations, we hypothesized that saracatinib would be effective at attenuating pulmonary fibrosis. Methods: We investigated the antifibrotic efficacy of saracatinib relative to nintedanib and pirfenidone in three preclinical models: 1) in vitro in normal human lung fibroblasts; 2) in vivo in bleomycin and recombinant Ad-TGF-β (adenovirus transforming growth factor-β) murine models of pulmonary fibrosis; and 3) ex vivo in mice and human precision-cut lung slices from these two murine models as well as patients with IPF and healthy donors. Measurements and Main Results: In each model, the effectiveness of saracatinib in blocking fibrogenic responses was equal or superior to nintedanib and pirfenidone. Transcriptomic analyses of TGF-β-stimulated normal human lung fibroblasts identified specific gene sets associated with fibrosis, including epithelial-mesenchymal transition, TGF-β, and WNT signaling that was uniquely altered by saracatinib. Transcriptomic analysis of whole-lung extracts from the two animal models of pulmonary fibrosis revealed that saracatinib reverted many fibrogenic pathways, including epithelial-mesenchymal transition, immune responses, and extracellular matrix organization. Amelioration of fibrosis and inflammatory cascades in human precision-cut lung slices confirmed the potential therapeutic efficacy of saracatinib in human lung fibrosis. Conclusions: These studies identify novel Src-dependent fibrogenic pathways and support the study of the therapeutic effectiveness of saracatinib in IPF treatment.
Collapse
Affiliation(s)
- Farida Ahangari
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Christine Becker
- Institute for Next Generation Healthcare, Department of Genetics and Genomic Sciences, and
- Division of Clinical Immunology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Daniel G. Foster
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Department of Pediatrics, and Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado
| | - Maurizio Chioccioli
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Meghan Nelson
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Department of Pediatrics, and Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado
| | - Keriann Beke
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Department of Pediatrics, and Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado
| | - Xing Wang
- Institute for Next Generation Healthcare, Department of Genetics and Genomic Sciences, and
- Division of Clinical Immunology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Aurelien Justet
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
- Service de Pneumologie, UNICAEN, Normandie University, Caen, France
| | - Taylor Adams
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Benjamin Readhead
- Institute for Next Generation Healthcare, Department of Genetics and Genomic Sciences, and
- ASU-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, Arizona
| | - Carly Meador
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Department of Pediatrics, and Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado
| | - Kelly Correll
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Department of Pediatrics, and Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado
| | - Loukia N. Lili
- Institute for Next Generation Healthcare, Department of Genetics and Genomic Sciences, and
| | - Helen M. Roybal
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Department of Pediatrics, and Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado
| | - Kadi-Ann Rose
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Shuizi Ding
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Thomas Barnthaler
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
- Section of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Natalie Briones
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Department of Pediatrics, and Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado
| | - Giuseppe DeIuliis
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Jonas C. Schupp
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Qin Li
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Norihito Omote
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Yael Aschner
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Department of Pediatrics, and Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado
| | - Lokesh Sharma
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Katrina W. Kopf
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Department of Pediatrics, and Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado
| | - Björn Magnusson
- Discovery Biology, Discovery Sciences, Research & Development, AstraZeneca, Gothenburg, Sweden
| | - Ryan Hicks
- BioPharmaceuticals Research & Development Cell Therapy, Research, and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), AstraZeneca, Gothenburg, Sweden
| | - Anna Backmark
- Discovery Biology, Discovery Sciences, Research & Development, AstraZeneca, Gothenburg, Sweden
| | - Charles S. Dela Cruz
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Ivan Rosas
- Department of Medicine, Baylor College of Medicine, Houston, Texas; and
| | - Leslie P. Cousens
- Emerging Innovations, Discovery Sciences, Research & Development, AstraZeneca, Boston, Massachusetts
| | - Joel T. Dudley
- Institute for Next Generation Healthcare, Department of Genetics and Genomic Sciences, and
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Gregory P. Downey
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Department of Pediatrics, and Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado
| |
Collapse
|
34
|
Elliot S, Catanuto P, Pereira-simon S, Xia X, Shahzeidi S, Roberts E, Ludlow J, Hamdan S, Daunert S, Parra J, Stone R, Pastar I, Tomic-Canic M, Glassberg MK. Urine-derived exosomes from individuals with IPF carry pro-fibrotic cargo. eLife 2022; 11:e79543. [PMID: 36454035 PMCID: PMC9714968 DOI: 10.7554/elife.79543] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 10/31/2022] [Indexed: 12/03/2022] Open
Abstract
Background MicroRNAs (miRNA) and other components contained in extracellular vesicles may reflect the presence of a disease. Lung tissue, sputum, and sera of individuals with idiopathic pulmonary fibrosis (IPF) show alterations in miRNA expression. We designed this study to test whether urine and/or tissue derived exosomal miRNAs from individuals with IPF carry cargo that can promote fibrosis. Methods Exosomes were isolated from urine (U-IPFexo), lung tissue myofibroblasts (MF-IPFexo), serum from individuals with IPF (n=16) and age/sex-matched controls without lung disease (n=10). We analyzed microRNA expression of isolated exosomes and their in vivo bio-distribution. We investigated the effect on ex vivo skin wound healing and in in vivo mouse lung models. Results U-IPFexo or MF-IPFexo expressed miR-let-7d, miR-29a-5p, miR-181b-3p and miR-199a-3p consistent with previous reports of miRNA expression obtained from lung tissue/sera from patients with IPF. In vivo bio-distribution experiments detected bioluminescent exosomes in the lung of normal C57Bl6 mice within 5 min after intravenous infusion, followed by distribution to other organs irrespective of exosome source. Exosomes labeled with gold nanoparticles and imaged by transmission electron microscopy were visualized in alveolar epithelial type I and type II cells. Treatment of human and mouse lung punches obtained from control, non-fibrotic lungs with either U-IPFexo or MF-IPFexo produced a fibrotic phenotype. A fibrotic phenotype was also induced in a human ex vivo skin model and in in vivo lung models. Conclusions Our results provide evidence of a systemic feature of IPF whereby exosomes contain pro-fibrotic miRNAs when obtained from a fibrotic source and interfere with response to tissue injury as measured in skin and lung models. Funding This work was supported in part by Lester and Sue Smith Foundation and The Samrick Family Foundation and NIH grants R21 AG060338 (SE and MKG), U01 DK119085 (IP, RS, MTC).
Collapse
Affiliation(s)
- Sharon Elliot
- DeWitt Daughtry Family Department of Surgery, University of Miami Leonard M. Miller School of MedicineMiamiUnited States
| | - Paola Catanuto
- DeWitt Daughtry Family Department of Surgery, University of Miami Leonard M. Miller School of MedicineMiamiUnited States
| | - Simone Pereira-simon
- DeWitt Daughtry Family Department of Surgery, University of Miami Leonard M. Miller School of MedicineMiamiUnited States
| | - Xiaomei Xia
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep, University of MiamiMiamiUnited States
| | | | - Evan Roberts
- Cancer Modeling Shared Resource Sylvester Comprehensive Cancer Center, University of MiamiMiamiUnited States
| | | | - Suzana Hamdan
- Department of Biochemistry and Molecular Biology, University of Miami, Miller School of MedicineMiamiUnited States
- Dr. JT Macdonald Foundation Biomedical Nanotechnology Institute, University of Miami Miller School of MedicineMiamiUnited States
| | - Sylvia Daunert
- Department of Biochemistry and Molecular Biology, University of Miami, Miller School of MedicineMiamiUnited States
- Dr. JT Macdonald Foundation Biomedical Nanotechnology Institute, University of Miami Miller School of MedicineMiamiUnited States
- Miami Clinical and Translational Science Institute, University of Miami Miller School of MedicineMiamiUnited States
| | - Jennifer Parra
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep, University of MiamiMiamiUnited States
| | - Rivka Stone
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous Surgery, University of MiamiMiamiUnited States
| | - Irena Pastar
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous Surgery, University of MiamiMiamiUnited States
| | - Marjana Tomic-Canic
- Wound Healing and Regenerative Medicine Research Program, Dr Phillip Frost Department of Dermatology and Cutaneous Surgery, University of MiamiMiamiUnited States
| | - Marilyn K Glassberg
- DeWitt Daughtry Family Department of Surgery, University of Miami Leonard M. Miller School of MedicineMiamiUnited States
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep, University of MiamiMiamiUnited States
- Department of Medicine, Stritch School of Medicine, Loyola University ChicagoChicagoUnited States
| |
Collapse
|
35
|
Macrophages and Wnts in Tissue Injury and Repair. Cells 2022; 11:cells11223592. [PMID: 36429021 PMCID: PMC9688352 DOI: 10.3390/cells11223592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/07/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022] Open
Abstract
Macrophages are important players in the immune system that sense various tissue challenges and trigger inflammation. Tissue injuries are followed by inflammation, which is tightly coordinated with tissue repair processes. Dysregulation of these processes leads to chronic inflammation or tissue fibrosis. Wnt ligands are present both in homeostatic and pathological conditions. However, their roles and mechanisms regulating inflammation and tissue repair are being investigated. Here we aim to provide an overview of overarching themes regarding Wnt and macrophages by reviewing the previous literature. We aim to gain future insights into how tissue inflammation, repair, regeneration, and fibrosis events are regulated by macrophages.
Collapse
|
36
|
Zimmermann CM, Baldassi D, Chan K, Adams NBP, Neumann A, Porras-Gonzalez DL, Wei X, Kneidinger N, Stoleriu MG, Burgstaller G, Witzigmann D, Luciani P, Merkel OM. Spray drying siRNA-lipid nanoparticles for dry powder pulmonary delivery. J Control Release 2022; 351:137-150. [PMID: 36126785 PMCID: PMC7613708 DOI: 10.1016/j.jconrel.2022.09.021] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 11/17/2022]
Abstract
While all the siRNA drugs on the market target the liver, the lungs offer a variety of currently undruggable targets which could potentially be treated with RNA therapeutics. Hence, local, pulmonary delivery of RNA nanoparticles could finally enable delivery beyond the liver. The administration of RNA drugs via dry powder inhalers offers many advantages related to physical, chemical and microbial stability of RNA and nanosuspensions. The present study was therefore designed to test the feasibility of engineering spray dried lipid nanoparticle (LNP) powders. Spray drying was performed using 5% lactose solution (m/V), and the targets were set to obtain nanoparticle sizes after redispersion of spray-dried powders around 150 nm, a residual moisture level below 5%, and RNA loss below 15% at maintained RNA bioactivity. The LNPs consisted of an ionizable cationic lipid which is a sulfur-containing analog of DLin-MC3-DMA, a helper lipid, cholesterol, and PEG-DMG encapsulating siRNA. Prior to the spray drying, the latter process was simulated with a novel dual emission fluorescence spectroscopy method to preselect the highest possible drying temperature and excipient solution maintaining LNP integrity and stability. Through characterization of physicochemical and aerodynamic properties of the spray dried powders, administration criteria for delivery to the lower respiratory tract were fulfilled. Spray dried LNPs penetrated the lung mucus layer and maintained bioactivity for >90% protein downregulation with a confirmed safety profile in a lung adenocarcinoma cell line. Additionally, the spray dried LNPs successfully achieved up to 50% gene silencing of the house keeping gene GAPDH in ex vivo human precision-cut lung slices at without increasing cytokine levels. This study verifies the successful spray drying procedure of LNP-siRNA systems maintaining their integrity and mediating strong gene silencing efficiency on mRNA and protein levels both in vitro and ex vivo. The successful spray drying procedure of LNP-siRNA formulations in 5% lactose solution creates a novel siRNA-based therapy option to target respiratory diseases such as lung cancer, asthma, COPD, cystic fibrosis and viral infections.
Collapse
Affiliation(s)
- Christoph M Zimmermann
- Department of Pharmacy, Pharmaceutical Technology and Biopharmacy, Ludwig-Maximilians Universität München, 81377 Munich, Germany; Department of Chemistry, Biochemistry and Pharmacy, University Bern, Freiestrasse 3, Bern, Switzerland
| | - Domizia Baldassi
- Department of Pharmacy, Pharmaceutical Technology and Biopharmacy, Ludwig-Maximilians Universität München, 81377 Munich, Germany
| | - Karen Chan
- Department of Biochemistry and Molecular Biology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; NanoMedicines Innovation Network (NMIN), 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Nathan B P Adams
- Nanotemper Technologies GmbH, Flößergasse 4, 81369 Munich, Germany
| | - Alina Neumann
- Nanotemper Technologies GmbH, Flößergasse 4, 81369 Munich, Germany
| | - Diana Leidy Porras-Gonzalez
- Institute of Lung Health and Immunity (LHI) and Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive, Helmholtz Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Xin Wei
- Institute of Lung Health and Immunity (LHI) and Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive, Helmholtz Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Nikolaus Kneidinger
- Department of Medicine V, University Hospital, LMU Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Mircea Gabriel Stoleriu
- Center for Thoracic Surgery Munich, Ludwig-Maximilians-University of Munich (LMU) and Asklepios Pulmonary Hospital, Marchioninistraße 15, 81377 Munich and Robert-Koch-Allee 2, 82131 Gauting, Germany
| | - Gerald Burgstaller
- Institute of Lung Health and Immunity (LHI) and Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive, Helmholtz Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Dominik Witzigmann
- Department of Biochemistry and Molecular Biology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; NanoMedicines Innovation Network (NMIN), 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada; NanoVation Therapeutics Inc., 2405 Wesbrook Mall 4th Floor, Vancouver V6T 1Z3, Canada.
| | - Paola Luciani
- Department of Chemistry, Biochemistry and Pharmacy, University Bern, Freiestrasse 3, Bern, Switzerland.
| | - Olivia M Merkel
- Department of Pharmacy, Pharmaceutical Technology and Biopharmacy, Ludwig-Maximilians Universität München, 81377 Munich, Germany; Institute of Lung Health and Immunity (LHI) and Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive, Helmholtz Munich, Member of the German Center for Lung Research (DZL), Munich, Germany.
| |
Collapse
|
37
|
Nußbaum SM, Krabbe J, Böll S, Babendreyer A, Martin C. Functional changes in long-term incubated rat precision-cut lung slices. Respir Res 2022; 23:261. [PMID: 36127699 PMCID: PMC9490993 DOI: 10.1186/s12931-022-02169-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 09/02/2022] [Indexed: 11/10/2022] Open
Abstract
Background Respiratory diseases represent a global health burden. Because research on therapeutic strategies of airway diseases is essential, the technique of precision-cut lung slices (PCLS) has been developed and widely studied. PCLS are an alternative ex vivo model and have the potential to replace and reduce in vivo animal models. So far, the majority of studies was conducted with short-term cultivated PCLS (≤ 72 h). As there is large interest in research of chronic diseases and chronic toxicity, feasibility of cultivating human PCLS long-term over 2 weeks and recently over 4 weeks was investigated by another research group with successful results. Our aim was to establish a model of long-term cultivated rat PCLS over a period of 29 days. Methods Rat PCLS were cultured for 29 days and analysed regarding viability, histopathology, reactivity and gene expression at different time points during cultivation. Results Cultivation of rat PCLS over a 29-day time period was successful with sustained viability. Furthermore, the ability of bronchoconstriction was maintained between 13 and 25 days, depending on the mediator. However, reduced relaxation, altered sensitivity and increased respiratory tone were observed. Regarding transcription, alteration in gene expression pattern of the investigated target genes was ascertained during long-term cultivation with mixed results. Furthermore, the preparation of PCLS seems to influence messenger ribonucleic acid (mRNA) expression of most target genes. Moreover, the addition of fetal bovine serum (FBS) to the culture medium did not improve viability of PCLS. In contrast to medium without FBS, FBS seems to affect measurements and resulted in marked cellular changes of metaplastic and/or regenerative origin. Conclusions Overall, a model of long-term cultivated rat PCLS which stays viable for 29 days and reactive for at least 13 days could be established. Before long-term cultivated PCLS can be used for in-depth study of chronic diseases and chronic toxicity, further investigations have to be made. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-02169-5.
Collapse
Affiliation(s)
- Sarah Marie Nußbaum
- Institute of Pharmacology and Toxicology, Medical Faculty, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Julia Krabbe
- Institute of Occupational, Social and Environmental Medicine, Medical Faculty, RWTH Aachen University, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Svenja Böll
- Department of Pediatrics, Medical Faculty, RWTH Aachen University, University Hospital Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Aaron Babendreyer
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Christian Martin
- Institute of Pharmacology and Toxicology, Medical Faculty, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.
| |
Collapse
|
38
|
Qiao X, Hou G, He YL, Song DF, An Y, Altawil A, Zhou XM, Wang QY, Kang J, Yin Y. The Novel Regulatory Role of the lncRNA–miRNA–mRNA Axis in Chronic Inflammatory Airway Diseases. Front Mol Biosci 2022; 9:927549. [PMID: 35769905 PMCID: PMC9234692 DOI: 10.3389/fmolb.2022.927549] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 05/19/2022] [Indexed: 12/28/2022] Open
Abstract
Chronic inflammatory airway diseases, characterized by airway inflammation and airway remodelling, are increasing as a cause of morbidity and mortality for all age groups and races across the world. The underlying molecular mechanisms involved in chronic inflammatory airway diseases have not been fully explored. MicroRNAs (miRNAs) and long noncoding RNAs (lncRNAs) have recently attracted much attention for their roles in the regulation of a variety of biological processes. A number of studies have confirmed that both lncRNAs and miRNAs can regulate the initiation and progression of chronic airway diseases by targeting mRNAs and regulating different cellular processes, such as proliferation, apoptosis, inflammation, migration, and epithelial–mesenchymal transition (EMT). Recently, accumulative evidence has shown that the novel regulatory mechanism underlying the interaction among lncRNAs, miRNAs and messenger RNAs (mRNAs) plays a critical role in the pathophysiological processes of chronic inflammatory airway diseases. In this review, we comprehensively summarized the regulatory roles of the lncRNA–miRNA–mRNA network in different cell types and their potential roles as biomarkers, indicators of comorbidities or therapeutic targets for chronic inflammatory airway diseases, particularly chronic obstructive pulmonary disease (COPD) and asthma.
Collapse
Affiliation(s)
- Xin Qiao
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Gang Hou
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Yu-Lin He
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Dong-Fang Song
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yi An
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Abdullah Altawil
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiao-Ming Zhou
- Respiratory Department, Center for Pulmonary Vascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- *Correspondence: Xiao-Ming Zhou, ; Yan Yin,
| | - Qiu-Yue Wang
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jian Kang
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yan Yin
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of China Medical University, Shenyang, China
- *Correspondence: Xiao-Ming Zhou, ; Yan Yin,
| |
Collapse
|
39
|
Fernandez RJ, Gardner ZJG, Slovik KJ, Liberti DC, Estep KN, Yang W, Chen Q, Santini GT, Perez JV, Root S, Bhatia R, Tobias JW, Babu A, Morley MP, Frank DB, Morrisey EE, Lengner CJ, Johnson FB. GSK3 inhibition rescues growth and telomere dysfunction in dyskeratosis congenita iPSC-derived type II alveolar epithelial cells. eLife 2022; 11:64430. [PMID: 35559731 PMCID: PMC9200405 DOI: 10.7554/elife.64430] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 05/11/2022] [Indexed: 11/27/2022] Open
Abstract
Dyskeratosis congenita (DC) is a rare genetic disorder characterized by deficiencies in telomere maintenance leading to very short telomeres and the premature onset of certain age-related diseases, including pulmonary fibrosis (PF). PF is thought to derive from epithelial failure, particularly that of type II alveolar epithelial (AT2) cells, which are highly dependent on Wnt signaling during development and adult regeneration. We use human induced pluripotent stem cell-derived AT2 (iAT2) cells to model how short telomeres affect AT2 cells. Cultured DC mutant iAT2 cells accumulate shortened, uncapped telomeres and manifest defects in the growth of alveolospheres, hallmarks of senescence, and apparent defects in Wnt signaling. The GSK3 inhibitor, CHIR99021, which mimics the output of canonical Wnt signaling, enhances telomerase activity and rescues the defects. These findings support further investigation of Wnt agonists as potential therapies for DC-related pathologies.
Collapse
Affiliation(s)
- Rafael Jesus Fernandez
- Medical Scientist Training Program, University of Pennsylvania, Philadelphia, United States
| | - Zachary J G Gardner
- Medical Scientist Training Program, University of Pennsylvania, Philadelphia, United States
| | - Katherine J Slovik
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, United States
| | - Derek C Liberti
- Cell and Molecular Biology Graduate Group, University of Pennsylvania, Philadelphia, United States
| | - Katrina N Estep
- Cell and Molecular Biology Graduate Group, University of Pennsylvania, Philadelphia, United States
| | - Wenli Yang
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, United States
| | - Qijun Chen
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, United States
| | - Garrett T Santini
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Javier V Perez
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, United States
| | - Sarah Root
- College of Arts and Sciences and Vagelos Scholars Program, University of Pennsylvania, Philadelphia, United States
| | - Ranvir Bhatia
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - John W Tobias
- Penn Genomic Analysis Core, University of Pennsylvania, Philadelphia, United States
| | - Apoorva Babu
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, United States
| | - Michael P Morley
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, United States
| | - David B Frank
- Penn-CHOP Lung Biology Institute, Children's Hospital of Philadelphia, Philadelphia, United States
| | - Edward E Morrisey
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, United States
| | - Christopher J Lengner
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, United States
| | - F Brad Johnson
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, United States
| |
Collapse
|
40
|
Baarsma HA, Van der Veen CHTJ, Lobee D, Mones N, Oosterhout E, Cattani-Cavalieri I, Schmidt M. Epithelial 3D-spheroids as a tool to study air pollutant-induced lung pathology. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2022; 27:185-190. [PMID: 35227934 DOI: 10.1016/j.slasd.2022.02.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 02/02/2022] [Accepted: 02/22/2022] [Indexed: 06/14/2023]
Abstract
Cigarette smoke (CS) and air pollutants (AP) activate pathological processes in bronchial epithelial cells resulting in lung function decline which severely impacts human health. Knowledge about the molecular mechanism(s) by which CS and AP induce pathology is limited. Our previous studies in 2D cultures of human bronchial epithelial (BEAS-2B) cells showed that CS exposure activates transforming growth factor-β1 (TGF-β1) release and signaling. Furthermore, CS exposure reduced the expression of E-cadherin, which was prevented by applying a TGF-β1 neutralizing antibody. Exposure of BEAS-2B cells cultured in 2D to diesel exhaust particles (DEP) increased TGF-β1 protein expression and reduced the expression of epithelial cell markers, whereas mesenchymal markers are upregulated. Conventional 2D cell culture may, however, not fully reflect the physiology of bronchial epithelial cells in vivo. To simulate the in vivo situation more closely we cultured the bronchial epithelial cells in a 3D environment in the current study. Treatment of epithelial spheroids with TGF-β resulted in reduced E-cadherin and increased collagen I expression, indicating the activation of epithelial-to-mesenchymal transition (EMT). Similarly, exposure of spheroids to DEP induced and EMT-like phenotype. Collectively, our data indicate AP induces an EMT-like phenotype of BEAS-2B cells in 3D spheroid cultures. This opens new avenues for drug development for the treatment of lung diseases induced by AP. The 3D spheroid cell culture is a novel, innovative and physiologically relevant model for culturing a variety of cells. It is a versatile tool for both high-throughput studies and for identifying molecular mechanisms involved in bronchial epithelial cell (patho)physiology.
Collapse
Affiliation(s)
- Hoeke A Baarsma
- Department of Molecular Pharmacology, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, the Netherland; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, the Netherland.
| | - Christina H T J Van der Veen
- Department of Molecular Pharmacology, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, the Netherland; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, the Netherland
| | - Danique Lobee
- Department of Molecular Pharmacology, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, the Netherland
| | - Nienke Mones
- Department of Molecular Pharmacology, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, the Netherland
| | - Emily Oosterhout
- Department of Molecular Pharmacology, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, the Netherland
| | - Isabella Cattani-Cavalieri
- Department of Molecular Pharmacology, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, the Netherland; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, the Netherland; Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, the Netherland; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, the Netherland
| |
Collapse
|
41
|
Wang H, Zhong Y, Li N, Yu M, Zhu L, Wang L, Chen F, Xu Y, Liu J, Huang H. Transcriptomic analysis and validation reveal the pathogenesis and a novel biomarker of acute exacerbation of chronic obstructive pulmonary disease. Respir Res 2022; 23:27. [PMID: 35151329 PMCID: PMC8840779 DOI: 10.1186/s12931-022-01950-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 02/06/2022] [Indexed: 11/29/2022] Open
Abstract
Background Acute exacerbation of chronic obstructive pulmonary disease (AECOPD) is the main factor that leads to the deterioration of the disease. Currently, the diagnosis of AECOPD mainly relies on clinical manifestations, good predictors or biomarkers are lacking. We aim to reveal specific biomarkers and potential pathogenesis of AECOPD and provide a research basis for the diagnosis and treatment. Methods Four patients with AECOPD, four patients with stable COPD, and five control subjects were enrolled for RNA sequencing and KEGG analysis. The mRNA level of target genes was verified by quantitative real-time PCR (qPCR) with an expanded sample size (30 patients with AECOPD, 27 patients with stable COPD, and 35 control subjects). ELISA and immunofluorescence were used to identify the target proteins. Furthermore, the expression and function of WNT/β-catenin signaling pathway were assessed in animal models of COPD. Results RNA sequencing showed that 54 genes were up-regulated and 111 genes were down-regulated in the AECOPD. Differentially expressed genes were mainly enriched in WNT signaling pathway, et al. QPCR revealed that multi-genes of the WNT/β-catenin signaling were significantly down-regulated in AECOPD (P < 0.05), and β-catenin protein was significantly decreased in plasma of AECOPD and stable COPD (P < 0.01), while phosphorylated β-catenin was significantly up-regulated in peripheral blood mononuclear cells of AECOPD (P < 0.05). Similarly, WNT ligands, WNT receptors, and downstream signaling molecules were down-regulated, with an increased phosphorylated β-catenin protein in animal models of COPD. Activation of WNT/β-catenin signaling pathway by lithium chloride reduced the expression of phosphorylated β-catenin and ameliorated the COPD-like airway inflammation in mice. Conclusion WNT/β-catenin signaling pathway is down-regulated in AECOPD patients and in animal models of COPD. Increased expression of phosphorylated β-catenin in the blood might be a potential biomarker of AECOPD. Activation of WNT/β-catenin pathway may also represent a therapeutic target for AECOPD. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-01950-w.
Collapse
|
42
|
Xie T, Lynn H, Parks WC, Stripp B, Chen P, Jiang D, Noble PW. Abnormal respiratory progenitors in fibrotic lung injury. Stem Cell Res Ther 2022; 13:64. [PMID: 35130980 PMCID: PMC8822870 DOI: 10.1186/s13287-022-02737-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/18/2022] [Indexed: 12/19/2022] Open
Abstract
Recent advances in single-cell RNA sequencing (scRNA-seq) and epithelium lineage labeling have yielded identification of multiple abnormal epithelial progenitor populations during alveolar type 2 (ATII) cell differentiation into alveolar type 1 (ATI) cells during regenerative lung post-fibrotic injury. These abnormal cells include basaloid/basal-like cells, ATII transition cells, and persistent epithelial progenitors (PEPs). These cells occurred and accumulated during the regeneration of distal airway and alveoli in response to both chronic and acute pulmonary injury. Among the alveolar epithelial progenitors, PEPs express a distinct Krt8+ phenotype that is rarely found in intact alveoli. However, post-injury, the Krt8+ phenotype is seen in dysplastic epithelial cells. Fully understanding the characteristics and functions of these newly found, injury-induced abnormal behavioral epithelial progenitors and the signaling pathways regulating their phenotype could potentially point the way to unique therapeutic targets for fibrosing lung diseases. This review summarizes recent advances in understanding these epithelial progenitors as they relate to uncovering regenerative mechanisms.
Collapse
Affiliation(s)
- Ting Xie
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Heather Lynn
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - William C Parks
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Barry Stripp
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Peter Chen
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Dianhua Jiang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Paul W Noble
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
43
|
Wnt/β-catenin signalling: function, biological mechanisms, and therapeutic opportunities. Signal Transduct Target Ther 2022; 7:3. [PMID: 34980884 PMCID: PMC8724284 DOI: 10.1038/s41392-021-00762-6] [Citation(s) in RCA: 1031] [Impact Index Per Article: 343.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 06/28/2021] [Accepted: 07/07/2021] [Indexed: 02/06/2023] Open
Abstract
The Wnt/β-catenin pathway comprises a family of proteins that play critical roles in embryonic development and adult tissue homeostasis. The deregulation of Wnt/β-catenin signalling often leads to various serious diseases, including cancer and non-cancer diseases. Although many articles have reviewed Wnt/β-catenin from various aspects, a systematic review encompassing the origin, composition, function, and clinical trials of the Wnt/β-catenin signalling pathway in tumour and diseases is lacking. In this article, we comprehensively review the Wnt/β-catenin pathway from the above five aspects in combination with the latest research. Finally, we propose challenges and opportunities for the development of small-molecular compounds targeting the Wnt signalling pathway in disease treatment.
Collapse
|
44
|
Soni DK, Biswas R. Role of Non-Coding RNAs in Post-Transcriptional Regulation of Lung Diseases. Front Genet 2021; 12:767348. [PMID: 34819948 PMCID: PMC8606426 DOI: 10.3389/fgene.2021.767348] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/25/2021] [Indexed: 12/16/2022] Open
Abstract
Non-coding RNAs (ncRNAs), notably microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), have recently gained increasing consideration because of their versatile role as key regulators of gene expression. They adopt diverse mechanisms to regulate transcription and translation, and thereby, the function of the protein, which is associated with several major biological processes. For example, proliferation, differentiation, apoptosis, and metabolic pathways demand fine-tuning for the precise development of a specific tissue or organ. The deregulation of ncRNA expression is concomitant with multiple diseases, including lung diseases. This review highlights recent advances in the post-transcriptional regulation of miRNAs and lncRNAs in lung diseases such as asthma, chronic obstructive pulmonary disease, cystic fibrosis, and idiopathic pulmonary fibrosis. Further, we also discuss the emerging role of ncRNAs as biomarkers as well as therapeutic targets for lung diseases. However, more investigations are required to explore miRNAs and lncRNAs interaction, and their function in the regulation of mRNA expression. Understanding these mechanisms might lead to early diagnosis and the development of novel therapeutics for lung diseases.
Collapse
Affiliation(s)
- Dharmendra Kumar Soni
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Roopa Biswas
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
45
|
Atkinson SP. A preview of selected articles. Stem Cells Transl Med 2021. [DOI: 10.1002/sct3.13034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
46
|
Decaris ML, Schaub JR, Chen C, Cha J, Lee GG, Rexhepaj M, Ho SS, Rao V, Marlow MM, Kotak P, Budi EH, Hooi L, Wu J, Fridlib M, Martin SP, Huang S, Chen M, Muñoz M, Hom TF, Wolters PJ, Desai TJ, Rock F, Leftheris K, Morgans DJ, Lepist EI, Andre P, Lefebvre EA, Turner SM. Dual inhibition of α vβ 6 and α vβ 1 reduces fibrogenesis in lung tissue explants from patients with IPF. Respir Res 2021; 22:265. [PMID: 34666752 PMCID: PMC8524858 DOI: 10.1186/s12931-021-01863-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 10/10/2021] [Indexed: 12/11/2022] Open
Abstract
RATIONALE αv integrins, key regulators of transforming growth factor-β activation and fibrogenesis in in vivo models of pulmonary fibrosis, are expressed on abnormal epithelial cells (αvβ6) and fibroblasts (αvβ1) in fibrotic lungs. OBJECTIVES We evaluated multiple αv integrin inhibition strategies to assess which most effectively reduced fibrogenesis in explanted lung tissue from patients with idiopathic pulmonary fibrosis. METHODS Selective αvβ6 and αvβ1, dual αvβ6/αvβ1, and multi-αv integrin inhibitors were characterized for potency, selectivity, and functional activity by ligand binding, cell adhesion, and transforming growth factor-β cell activation assays. Precision-cut lung slices generated from lung explants from patients with idiopathic pulmonary fibrosis or bleomycin-challenged mouse lungs were treated with integrin inhibitors or standard-of-care drugs (nintedanib or pirfenidone) and analyzed for changes in fibrotic gene expression or TGF-β signaling. Bleomycin-challenged mice treated with dual αvβ6/αvβ1 integrin inhibitor, PLN-74809, were assessed for changes in pulmonary collagen deposition and Smad3 phosphorylation. MEASUREMENTS AND MAIN RESULTS Inhibition of integrins αvβ6 and αvβ1 was additive in reducing type I collagen gene expression in explanted lung tissue slices from patients with idiopathic pulmonary fibrosis. These data were replicated in fibrotic mouse lung tissue, with no added benefit observed from inhibition of additional αv integrins. Antifibrotic efficacy of dual αvβ6/αvβ1 integrin inhibitor PLN-74809 was confirmed in vivo, where dose-dependent inhibition of pulmonary Smad3 phosphorylation and collagen deposition was observed. PLN-74809 also, more potently, reduced collagen gene expression in fibrotic human and mouse lung slices than clinically relevant concentrations of nintedanib or pirfenidone. CONCLUSIONS In the fibrotic lung, dual inhibition of integrins αvβ6 and αvβ1 offers the optimal approach for blocking fibrogenesis resulting from integrin-mediated activation of transforming growth factor-β.
Collapse
Affiliation(s)
| | | | - Chun Chen
- Pliant Therapeutics, South San Francisco, CA, USA
| | - Jacob Cha
- Pliant Therapeutics, South San Francisco, CA, USA
| | - Gail G Lee
- Pliant Therapeutics, South San Francisco, CA, USA
| | | | - Steve S Ho
- Pliant Therapeutics, South San Francisco, CA, USA
| | - Vikram Rao
- Pliant Therapeutics, South San Francisco, CA, USA
| | | | - Prerna Kotak
- Pliant Therapeutics, South San Francisco, CA, USA
| | - Erine H Budi
- Pliant Therapeutics, South San Francisco, CA, USA
| | - Lisa Hooi
- Pliant Therapeutics, South San Francisco, CA, USA
| | - Jianfeng Wu
- Pliant Therapeutics, South San Francisco, CA, USA
| | | | | | - Shaoyi Huang
- Pliant Therapeutics, South San Francisco, CA, USA
| | - Ming Chen
- Pliant Therapeutics, South San Francisco, CA, USA
| | - Manuel Muñoz
- Pliant Therapeutics, South San Francisco, CA, USA
| | | | - Paul J Wolters
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Tushar J Desai
- Department of Medicine, Stanford University, Stanford, CA, USA
| | | | | | - David J Morgans
- Pliant Therapeutics, South San Francisco, CA, USA
- Maze Therapeutics, South San Francisco, CA, USA
| | | | - Patrick Andre
- Pliant Therapeutics, South San Francisco, CA, USA
- Acceleron Pharma, Cambridge, MA, USA
| | | | | |
Collapse
|
47
|
Selo MA, Sake JA, Kim KJ, Ehrhardt C. In vitro and ex vivo models in inhalation biopharmaceutical research - advances, challenges and future perspectives. Adv Drug Deliv Rev 2021; 177:113862. [PMID: 34256080 DOI: 10.1016/j.addr.2021.113862] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/02/2021] [Accepted: 07/06/2021] [Indexed: 12/11/2022]
Abstract
Oral inhalation results in pulmonary drug targeting and thereby reduces systemic side effects, making it the preferred means of drug delivery for the treatment of respiratory disorders such as asthma, chronic obstructive pulmonary disease or cystic fibrosis. In addition, the high alveolar surface area, relatively low enzymatic activity and rich blood supply of the distal airspaces offer a promising pathway to the systemic circulation. This is particularly advantageous when a rapid onset of pharmacological action is desired or when the drug is suffering from stability issues or poor biopharmaceutical performance following oral administration. Several cell and tissue-based in vitro and ex vivo models have been developed over the years, with the intention to realistically mimic pulmonary biological barriers. It is the aim of this review to critically discuss the available models regarding their advantages and limitations and to elaborate further which biopharmaceutical questions can and cannot be answered using the existing models.
Collapse
|
48
|
Chen DY, Sun NH, Chen X, Gong JJ, Yuan ST, Hu ZZ, Lu NN, Körbelin J, Fukunaga K, Liu QH, Lu YM, Han F. Endothelium-derived semaphorin 3G attenuates ischemic retinopathy by coordinating β-catenin-dependent vascular remodeling. J Clin Invest 2021; 131:135296. [PMID: 33586674 DOI: 10.1172/jci135296] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 12/10/2020] [Indexed: 12/14/2022] Open
Abstract
Abnormal angiogenesis and regression of the diseased retinal vasculature are key processes associated with ischemic retinopathies, but the underlying mechanisms that regulate vascular remodeling remain poorly understood. Here, we confirmed the specific expression of semaphorin 3G (Sema3G) in retinal endothelial cells (ECs), which was required for vascular remodeling and the amelioration of ischemic retinopathy. We found that Sema3G was elevated in the vitreous fluid of patients with proliferative diabetic retinopathy (PDR) and in the neovascularization regression phase of oxygen-induced retinopathy (OIR). Endothelial-specific Sema3G knockout mice exhibited decreased vessel density and excessive matrix deposition in the retinal vasculature. Moreover, loss of Sema3G aggravated pathological angiogenesis in mice with OIR. Mechanistically, we demonstrated that HIF-2α directly regulated Sema3G transcription in ECs under hypoxia. Sema3G coordinated the functional interaction between β-catenin and VE-cadherin by increasing β-catenin stability in the endothelium through the neuropilin-2 (Nrp2)/PlexinD1 receptor. Furthermore, Sema3G supplementation enhanced healthy vascular network formation and promoted diseased vasculature regression during blood vessel remodeling. Overall, we deciphered the endothelium-derived Sema3G-dependent events involved in modulating physiological vascular remodeling and regression of pathological blood vessels for reparative vascular regeneration. Our findings shed light on the protective effect of Sema3G in ischemic retinopathies.
Collapse
Affiliation(s)
- Dan-Yang Chen
- Department of Physiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China.,Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, China.,Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ning-He Sun
- Department of Physiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China.,Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, China.,Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xiang Chen
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Jun-Jie Gong
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Song-Tao Yuan
- Department of Ophthalmology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zi-Zhong Hu
- Department of Ophthalmology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Nan-Nan Lu
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Jakob Körbelin
- Department of Oncology and Hematology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Qing-Huai Liu
- Department of Ophthalmology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ying-Mei Lu
- Department of Physiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Feng Han
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, China.,Institute of Brain Science, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
49
|
Burgstaller G, Gerckens M, Eickelberg O, Königshoff M. Decellularized Human Lung Scaffolds as Complex Three-Dimensional Tissue Culture Models to Study Functional Behavior of Fibroblasts. Methods Mol Biol 2021; 2299:447-456. [PMID: 34028760 DOI: 10.1007/978-1-0716-1382-5_30] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
In vitro culturing of cells in two-dimensional (2D) environments is a widespread used methodology in biomedical research. Most commonly, cells are cultured on artificial plastic dish surfaces, which lead to abnormal functional behaviors, as plastic does not reflect the native microenvironment found in vivo or in situ. Therefore, a multitude of three-dimensional (3D) cell culture systems were developed in the past years, which aim to bridge the gap between 2D cell culture dishes and the in vivo situation. One of the more recent development in the field, the generation of viable precision-cut tissue slices from various organs emerged as an exciting approach to study complex interactions and biological processes ex vivo in 3D. Decellularization of such tissue slices leads to the removal of all functional cells, and leaves behind a scaffold of extracellular matrix (ECM), which closely recapitulates the molecular composition, mechanical properties, topology, and microarchitecture of native ECM. Subsequently, decellularized precision-cut lung slices (PCLS), also called 3D lung tissue culture (3D-LTCs), can be successfully reseeded with a variety of cell types, including fibroblasts, which attach to and engraft into the matrix. Here, we describe the generation of PCLS from resected human lung tissue and their decellularization and recellularization with primary human fibroblasts. This novel 3D tissue culture model allows for various functional studies of fibroblast behavior on native ECM composition and topology.
Collapse
Affiliation(s)
- Gerald Burgstaller
- Institute of Lung Biology and Disease (ILBD) and Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany. .,CPC-M bioArchive, Helmholtz Zentrum München, Comprehensive Pneumology Center Munich DZL/CPC-M, München, Germany.
| | - Michael Gerckens
- Institute of Lung Biology and Disease (ILBD) and Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany.,CPC-M bioArchive, Helmholtz Zentrum München, Comprehensive Pneumology Center Munich DZL/CPC-M, München, Germany
| | - Oliver Eickelberg
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Melanie Königshoff
- CPC-M bioArchive, Helmholtz Zentrum München, Comprehensive Pneumology Center Munich DZL/CPC-M, München, Germany.,Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Comprehensive Pneumology Center (CPC), Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| |
Collapse
|
50
|
Tsoyi K, Liang X, De Rossi G, Ryter SW, Xiong K, Chu SG, Liu X, Ith B, Celada LJ, Romero F, Robertson MJ, Esposito AJ, Poli S, El-Chemaly S, Perrella MA, Shi Y, Whiteford J, Rosas IO. CD148 Deficiency in Fibroblasts Promotes the Development of Pulmonary Fibrosis. Am J Respir Crit Care Med 2021; 204:312-325. [PMID: 33784491 DOI: 10.1164/rccm.202008-3100oc] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Rationale: CD148/PTRJ (receptor-like protein tyrosine phosphatase η) exerts antifibrotic effects in experimental pulmonary fibrosis via interactions with its ligand syndecan-2; however, the role of CD148 in human pulmonary fibrosis remains incompletely characterized.Objectives: We investigated the role of CD148 in the profibrotic phenotype of fibroblasts in idiopathic pulmonary fibrosis (IPF).Methods: Conditional CD148 fibroblast-specific knockout mice were generated and exposed to bleomycin and then assessed for pulmonary fibrosis. Lung fibroblasts (mouse lung and human IPF lung), and precision-cut lung slices from human patients with IPF were isolated and subjected to experimental treatments. A CD148-activating 18-aa mimetic peptide (SDC2-pep) derived from syndecan-2 was evaluated for its therapeutic potential.Measurements and Main Results: CD148 expression was downregulated in IPF lungs and fibroblasts. In human IPF lung fibroblasts, silencing of CD148 increased extracellular matrix production and resistance to apoptosis, whereas overexpression of CD148 reversed the profibrotic phenotype. CD148 fibroblast-specific knockout mice displayed increased pulmonary fibrosis after bleomycin challenge compared with control mice. CD148-deficient fibroblasts exhibited hyperactivated PI3K/Akt/mTOR signaling, reduced autophagy, and increased p62 accumulation, which induced NF-κB activation and profibrotic gene expression. SDC2-pep reduced pulmonary fibrosis in vivo and inhibited IPF-derived fibroblast activation. In precision-cut lung slices from patients with IPF and control patients, SDC2-pep attenuated profibrotic gene expression in IPF and normal lungs stimulated with profibrotic stimuli.Conclusions: Lung fibroblast CD148 activation reduces p62 accumulation, which exerts antifibrotic effects by inhibiting NF-κB-mediated profibrotic gene expression. Targeting the CD148 phosphatase with activating ligands such as SDC2-pep may represent a potential therapeutic strategy in IPF.
Collapse
Affiliation(s)
- Konstantin Tsoyi
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Xiaoliang Liang
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Giulia De Rossi
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Stefan W Ryter
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Kevin Xiong
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Sarah G Chu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Xiaoli Liu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Bonna Ith
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Lindsay J Celada
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Freddy Romero
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Matthew J Robertson
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Anthony J Esposito
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Sergio Poli
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Souheil El-Chemaly
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Mark A Perrella
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - YuanYuan Shi
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - James Whiteford
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Ivan O Rosas
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Baylor College of Medicine, Houston, Texas.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| |
Collapse
|