1
|
van der Koog L, Woest ME, Gorter IC, Verschut V, Elferink RAB, Zuidhof AB, Nugraha DF, Koloko Ngassie ML, Bos SIT, Dhakad D, Wolters JC, Horvatovich PL, Prakash YS, Timens W, Yildirim ÖA, Brandsma CA, Frijlink HW, Nagelkerke A, Gosens R. Fibroblast-derived osteoglycin promotes epithelial cell repair. NPJ Regen Med 2025; 10:16. [PMID: 40133363 PMCID: PMC11937367 DOI: 10.1038/s41536-025-00404-3] [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: 12/02/2024] [Accepted: 03/14/2025] [Indexed: 03/27/2025] Open
Abstract
There is an urgent need for innovative therapies targeting defective epithelial repair in chronic diseases like COPD. The mesenchymal niche is a critical regulator in epithelial stem cell activation, suggesting that their secreted factors are possible potent drug targets. Utilizing a proteomics-guided drug discovery strategy, we explored the lung fibroblast secretome to uncover impactful drug targets. Our lung organoid assays identified several regenerative ligands, with osteoglycin (OGN) showing the most profound effects. Transcriptomic analyses revealed that OGN enhances alveolar progenitor differentiation, detoxifies reactive oxygen species, and strengthens fibroblast-epithelial crosstalk. OGN expression was diminished in COPD patients and smoke-exposed mice. An active fragment of OGN (leucine-rich repeat regions 4-7) replicated full-length OGN's regenerative effects, significantly ameliorating elastase-induced lung injury in lung slices and improving lung function in vivo. These findings highlight OGN as a pivotal secreted protein for alveolar epithelial repair, positioning its active fragment as a promising therapeutic for COPD.
Collapse
Affiliation(s)
- Luke van der Koog
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Groningen, The Netherlands
| | | | | | | | - Robin A B Elferink
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Groningen, The Netherlands
| | - Annet B Zuidhof
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Groningen, The Netherlands
| | - Dyan F Nugraha
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Groningen, The Netherlands
| | - Maunick L Koloko Ngassie
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Groningen, The Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Sophie I T Bos
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Groningen, The Netherlands
| | - Deepesh Dhakad
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Justina C Wolters
- Department of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands
| | - Peter L Horvatovich
- Department of Analytical Biochemistry, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Wim Timens
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Groningen, The Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
| | - Önder A Yildirim
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
- Institute of Experimental Pneumology, University Hospital, Ludwig-Maximilians University, Munich, Germany
| | - Corry-Anke Brandsma
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Groningen, The Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
| | - Henderik W Frijlink
- Department of Pharmaceutical Technology and Biopharmacy, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Anika Nagelkerke
- Department of Pharmaceutical Analysis, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Reinoud Gosens
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, The Netherlands.
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
2
|
van der Koog L, Boerrigter MJ, Gorter IC, Gosens R, Nagelkerke A. Lung fibroblast-derived extracellular vesicles and soluble factors alleviate elastase-induced lung injury. Eur J Pharmacol 2024; 974:176612. [PMID: 38677537 DOI: 10.1016/j.ejphar.2024.176612] [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: 11/17/2023] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 04/29/2024]
Abstract
One of the main pathological features of chronic obstructive pulmonary disease (COPD) is the loss of functional alveolar tissue as a consequence of impaired regenerative capacities (emphysema). Recent research suggests that the secretome from mesenchymal cells, particularly extracellular vesicles (EVs), may possess regenerative properties beneficial for lung repair. However, the regenerative potential of the soluble factors (SFs) within the secretome remains largely unexplored in COPD. To this extent, we purified EVs and SFs secreted by lung fibroblasts to generate EV-enriched and SF-enriched fractions, and evaluated their effects on elastase-induced lung injury in both precision-cut lung slices (PCLS) and a mouse model. EV- and SF-enriched fractions were concentrated and purified from the conditioned medium of cultured MRC-5 lung fibroblasts using a combination of ultrafiltration and size exclusion chromatography, and were subsequently characterized according to the MISEV guidelines. Treatment with EV- or SF-enriched concentrates prevented and improved elastase-induced emphysema in PCLS, leading to reduced lung injury and upregulated markers of alveolar epithelial cells (aquaporin 5 and surfactant protein C), indicating potential parenchymal regeneration. Accordingly, prophylactic intratracheal treatment with lung fibroblast-derived EV- and SF-enriched concentrates in vivo attenuated elastase-induced lung tissue destruction, improved lung function, and enhanced gene expression of alveolar epithelial cell markers. Here, alveolar repair not only serves the purpose of facilitating gas exchange, but also by reinstating the essential parenchymal tethering required for optimal airway mechanics. In conclusion, this study highlights the therapeutic potential of both lung fibroblast-derived EV- and SF-enriched concentrates for the treatment of lung injury and emphysema.
Collapse
Affiliation(s)
- Luke van der Koog
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, Faculty of Science and Engineering, University of Groningen, Groningen, the Netherlands; GRIAC, Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Groningen, the Netherlands
| | - Myrthe J Boerrigter
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, Faculty of Science and Engineering, University of Groningen, Groningen, the Netherlands; GRIAC, Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Reinoud Gosens
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy, Faculty of Science and Engineering, University of Groningen, Groningen, the Netherlands; GRIAC, Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, Groningen, the Netherlands
| | - Anika Nagelkerke
- Department of Pharmaceutical Analysis, Groningen Research Institute of Pharmacy, Faculty of Science and Engineering, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
3
|
Huang WJ, Ko CY. Systematic review and meta-analysis of nutrient supplements for treating sarcopenia in people with chronic obstructive pulmonary disease. Aging Clin Exp Res 2024; 36:69. [PMID: 38483650 PMCID: PMC10940388 DOI: 10.1007/s40520-024-02722-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: 11/09/2023] [Accepted: 02/09/2024] [Indexed: 03/17/2024]
Abstract
Individuals with chronic obstructive pulmonary disease (COPD) are prone to malnutrition and sarcopenia as a result of nutritional deficiencies and increased energy metabolism. However, the effects of nutrient supplements (NS) on treating sarcopenia in patients with COPD are not well established from systematic evidence. This meta-analysis examined the effect of NS on sarcopenia in patients with COPD. A systematic search of multiple databases was conducted, and 29 randomized controlled trials involving 1625 participants (age, mean [SD] = 67.9 [7.8] years) were analyzed. NS demonstrated significant improvements in body weight (MD,1.33 kg; 95% CI, 0.60, 2.05 kg; P = 0.0003; I2 = 87%), fat-free mass index (MD, 0.74 kg/m2; 95% CI, 0.21, 1.27 kg/m2; P = 0.007; I2 = 75%), and 6-min walk test (MD, 19.43 m; 95% CI, 4.91, 33.94 m; P = 0.009; I2 = 81%) compared with control. However, NS had nonsignificant effects on handgrip strength (SMD, 0.36; 95% CI, - 0.15, 0.88; P = 0.16; I2 = 87%) and quadriceps muscle strength (SMD, 0.11; 95% CI, - 0.06, 0.27; P = 0.20; I2 = 25%) compared with the control. In conclusion, NS may be an effective treatment for improving body composition and physical performance in COPD. Future studies should explore the effects of intervention durations, specific NS types, or combined training in patients with COPD and sarcopenia.
Collapse
Affiliation(s)
- Wen-Jian Huang
- Department of Clinical Nutrition, the Second Affiliated Hospital of Fujian Medical University, No. 34, Zhongshanbei Rd, Licheng District, Quanzhou, 362000, Fujian, China
- Huidong Center for Chronic Disease Control, Huizhou, 516300, Guangdong, China
| | - Chih-Yuan Ko
- Department of Clinical Nutrition, the Second Affiliated Hospital of Fujian Medical University, No. 34, Zhongshanbei Rd, Licheng District, Quanzhou, 362000, Fujian, China.
- School of Public Health, Fujian Medical University, Fuzhou, Fujian, China.
| |
Collapse
|
4
|
Fuentes-Mateos R, Gosens R. Disinherit your Descendants by Rewriting the Chronic Pulmonary Obstructive Disease Epigenetic Script. Am J Respir Cell Mol Biol 2024; 70:153-154. [PMID: 38060824 PMCID: PMC10914766 DOI: 10.1165/rcmb.2023-0387ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024] Open
Affiliation(s)
- Rocío Fuentes-Mateos
- Department of Molecular Pharmacology University of Groningen Groningen, the Netherlands
| | - Reinoud Gosens
- Department of Molecular Pharmacology University of Groningen Groningen, the Netherlands
| |
Collapse
|
5
|
Fabbri LM, Celli BR, Agustí A, Criner GJ, Dransfield MT, Divo M, Krishnan JK, Lahousse L, Montes de Oca M, Salvi SS, Stolz D, Vanfleteren LEGW, Vogelmeier CF. COPD and multimorbidity: recognising and addressing a syndemic occurrence. THE LANCET. RESPIRATORY MEDICINE 2023; 11:1020-1034. [PMID: 37696283 DOI: 10.1016/s2213-2600(23)00261-8] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 06/21/2023] [Accepted: 06/30/2023] [Indexed: 09/13/2023]
Abstract
Most patients with chronic obstructive pulmonary disease (COPD) have at least one additional, clinically relevant chronic disease. Those with the most severe airflow obstruction will die from respiratory failure, but most patients with COPD die from non-respiratory disorders, particularly cardiovascular diseases and cancer. As many chronic diseases have shared risk factors (eg, ageing, smoking, pollution, inactivity, and poverty), we argue that a shift from the current paradigm in which COPD is considered as a single disease with comorbidities, to one in which COPD is considered as part of a multimorbid state-with co-occurring diseases potentially sharing pathobiological mechanisms-is needed to advance disease prevention, diagnosis, and management. The term syndemics is used to describe the co-occurrence of diseases with shared mechanisms and risk factors, a novel concept that we propose helps to explain the clustering of certain morbidities in patients diagnosed with COPD. A syndemics approach to understanding COPD could have important clinical implications, in which the complex disease presentations in these patients are addressed through proactive diagnosis, assessment of severity, and integrated management of the COPD multimorbid state, with a patient-centred rather than a single-disease approach.
Collapse
Affiliation(s)
- Leonardo M Fabbri
- Section of Respiratory Medicine, Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Bartolome R Celli
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Alvar Agustí
- Cátedra Salud Respiratoria, Universitat de Barcelona, Barcelona, Spain; Institut Respiratori, Clínic Barcelona, Barcelona, Spain; Institut d'Investigacions Biomédicas August Pi i Sunyer, Barcelona, Spain; Centro de Investigación Biomédica en Red Enfermedades Respiratorias, Spain
| | - Gerard J Criner
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, USA
| | - Mark T Dransfield
- Lung Health Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Miguel Divo
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jamuna K Krishnan
- Division of Pulmonary and Critical Care, Weill Cornell Medicine, New York, NY, USA
| | - Lies Lahousse
- Department of Bioanalysis, Ghent University, Ghent, Belgium
| | - Maria Montes de Oca
- School of Medicine, Universidad Central de Venezuela, Caracas, Venezuela; Hospital Centro Medico de Caracas, Caracas, Venezuela
| | - Sundeep S Salvi
- Pulmocare Research and Education (PURE) Foundation, Pune, India; School of Health Sciences, Symbiosis International Deemed University, Pune, India
| | - Daiana Stolz
- Clinic of Respiratory Medicine and Pulmonary Cell Research, University Hospital Basel, Basel, Switzerland; Department of Clinical Research, University Hospital Basel, Basel, Switzerland; Clinic of Respiratory Medicine and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lowie E G W Vanfleteren
- COPD Center, Department of Respiratory Medicine and Allergology, Sahlgrenska University Hospital, Gothenburg, Sweden; Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Claus F Vogelmeier
- Department of Medicine, Pulmonary and Critical Care Medicine, University Medical Centre Giessen and Marburg, Philipps University of Marburg, Member of the German Centre for Lung Research, Marburg, Germany.
| |
Collapse
|
6
|
Upadhyay P, Wu CW, Pham A, Zeki AA, Royer CM, Kodavanti UP, Takeuchi M, Bayram H, Pinkerton KE. Animal models and mechanisms of tobacco smoke-induced chronic obstructive pulmonary disease (COPD). JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2023; 26:275-305. [PMID: 37183431 PMCID: PMC10718174 DOI: 10.1080/10937404.2023.2208886] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is the third leading cause of death worldwide, and its global health burden is increasing. COPD is characterized by emphysema, mucus hypersecretion, and persistent lung inflammation, and clinically by chronic airflow obstruction and symptoms of dyspnea, cough, and fatigue in patients. A cluster of pathologies including chronic bronchitis, emphysema, asthma, and cardiovascular disease in the form of hypertension and atherosclerosis variably coexist in COPD patients. Underlying causes for COPD include primarily tobacco use but may also be driven by exposure to air pollutants, biomass burning, and workplace related fumes and chemicals. While no single animal model might mimic all features of human COPD, a wide variety of published models have collectively helped to improve our understanding of disease processes involved in the genesis and persistence of COPD. In this review, the pathogenesis and associated risk factors of COPD are examined in different mammalian models of the disease. Each animal model included in this review is exclusively created by tobacco smoke (TS) exposure. As animal models continue to aid in defining the pathobiological mechanisms of and possible novel therapeutic interventions for COPD, the advantages and disadvantages of each animal model are discussed.
Collapse
Affiliation(s)
- Priya Upadhyay
- Center for Health and the Environment, University of California, Davis, Davis, CA 95616 USA
| | - Ching-Wen Wu
- Center for Health and the Environment, University of California, Davis, Davis, CA 95616 USA
| | - Alexa Pham
- Center for Health and the Environment, University of California, Davis, Davis, CA 95616 USA
| | - Amir A. Zeki
- Department of Internal Medicine; Division of Pulmonary, Critical Care, and Sleep Medicine, Center for Comparative Respiratory Biology and Medicine, School of Medicine; University of California, Davis, School of Medicine; U.C. Davis Lung Center; Davis, CA USA
| | - Christopher M. Royer
- California National Primate Research Center, University of California, Davis, Davis, CA 95616 USA
| | - Urmila P. Kodavanti
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Minoru Takeuchi
- Department of Animal Medical Science, Kyoto Sangyo University, Kyoto, Japan
| | - Hasan Bayram
- Koc University Research Center for Translational Medicine (KUTTAM), School of Medicine, Istanbul, Turkey
| | - Kent E. Pinkerton
- Center for Health and the Environment, University of California, Davis, Davis, CA 95616 USA
| |
Collapse
|
7
|
Tanino R, Tsubata Y, Hotta T, Okimoto T, Amano Y, Takechi M, Tanaka T, Akita T, Nagase M, Yamashita C, Wada K, Isobe T. Characterization of a spontaneous mouse model of mild, accelerated aging via ECM degradation in emphysematous lungs. Sci Rep 2023; 13:10740. [PMID: 37400563 DOI: 10.1038/s41598-023-37638-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 06/25/2023] [Indexed: 07/05/2023] Open
Abstract
Emphysema limits airflow and causes irreversible progression of chronic obstructive pulmonary disease (COPD). Strain differences must be considered when selecting mouse models of COPD, owing to disease complexity. We previously reported that a novel C57BL/6JJcl substrain, the Mayumi-Emphysema (ME) mouse, exhibits spontaneous emphysema; however, the other characteristics remain unknown. We aimed to characterize the lungs of ME mice and determine their experimental availability as a model. ME mice had a lower body weight than the control C57BL/6JJcl mice, with a median survival time of ~80 weeks. ME mice developed diffused emphysema with respiratory dysfunction from 8 to 26 weeks of age, but did not develop bronchial wall thickening. Proteomic analyses revealed five extracellular matrix-related clusters in downregulated lung proteins in ME mice. Moreover, EFEMP2/fibulin-4, an essential extracellular matrix protein, was the most downregulated protein in the lungs of ME mice. Murine and human EFEMP2 were detected in the pulmonary artery. Furthermore, patients with mild COPD showed decreased EFEMP2 levels in the pulmonary artery when compared to those without COPD. The ME mouse is a model of mild, accelerated aging with low-inflammatory emphysema and respiratory dysfunction that progresses with age and pulmonary EFEMP2 decrease, similar to that observed in patients with mild COPD.
Collapse
Affiliation(s)
- Ryosuke Tanino
- Department of Internal Medicine, Division of Respiratory Medicine and Medical Oncology, Faculty of Medicine, Shimane University, 89-1 Enya, Izumo, Shimane, 693-8501, Japan
| | - Yukari Tsubata
- Department of Internal Medicine, Division of Respiratory Medicine and Medical Oncology, Faculty of Medicine, Shimane University, 89-1 Enya, Izumo, Shimane, 693-8501, Japan.
| | - Takamasa Hotta
- Department of Internal Medicine, Division of Respiratory Medicine and Medical Oncology, Faculty of Medicine, Shimane University, 89-1 Enya, Izumo, Shimane, 693-8501, Japan
| | - Tamio Okimoto
- Department of Internal Medicine, Division of Respiratory Medicine and Medical Oncology, Faculty of Medicine, Shimane University, 89-1 Enya, Izumo, Shimane, 693-8501, Japan
| | - Yoshihiro Amano
- Department of Internal Medicine, Division of Respiratory Medicine and Medical Oncology, Faculty of Medicine, Shimane University, 89-1 Enya, Izumo, Shimane, 693-8501, Japan
| | - Mayumi Takechi
- Department of Experimental Animals, Interdisciplinary Center for Science Research, Organization for Research and Academic Information, Shimane University, Izumo, Japan
| | - Tetsuya Tanaka
- Department of Human Nutrition, Faculty of Contemporary Life Science, Chugoku Gakuen University, Okayama, Japan
| | - Tomomi Akita
- Department of Pharmaceutics and Drug Delivery, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Mamiko Nagase
- Department of Organ Pathology, Faculty of Medicine, Shimane University, Izumo, Japan
| | - Chikamasa Yamashita
- Department of Pharmaceutics and Drug Delivery, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Koichiro Wada
- Department of Pharmacology, Faculty of Medicine, Shimane University, Izumo, Japan
| | - Takeshi Isobe
- Department of Internal Medicine, Division of Respiratory Medicine and Medical Oncology, Faculty of Medicine, Shimane University, 89-1 Enya, Izumo, Shimane, 693-8501, Japan
| |
Collapse
|
8
|
Altalhi W, Wu T, Wojtkiewicz GR, Jeffs S, Miki K, Ott HC. Intratracheally injected human-induced pluripotent stem cell-derived pneumocytes and endothelial cells engraft in the distal lung and ameliorate emphysema in a rat model. J Thorac Cardiovasc Surg 2023; 166:e23-e37. [PMID: 36933786 DOI: 10.1016/j.jtcvs.2023.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/01/2023] [Accepted: 03/05/2023] [Indexed: 03/20/2023]
Abstract
OBJECTIVES Pulmonary emphysema is characterized by the destruction of alveolar units and reduced gas exchange capacity. In the present study, we aimed to deliver induced pluripotent stem cell-derived endothelial cells and pneumocytes to repair and regenerate distal lung tissue in an elastase-induced emphysema model. METHODS We induced emphysema in athymic rats via intratracheal injection of elastase as previously reported. At 21 and 35 days after elastase treatment, we suspended 80 million induced pluripotent stem cell-derived endothelial cells and 20 million induced pluripotent stem cell-derived pneumocytes in hydrogel and injected the mixture intratracheally. On day 49 after elastase treatment, we performed imaging, functional analysis, and collected lungs for histology. RESULTS Using immunofluorescence detection of human-specific human leukocyte antigen 1, human-specific CD31, and anti--green fluorescent protein for the reporter labeled pneumocytes, we found that transplanted cells engrafted in 14.69% ± 0.95% of the host alveoli and fully integrated to form vascularized alveoli together with host cells. Transmission electron microscopy confirmed the incorporation of the transplanted human cells and the formation of a blood-air barrier. Human endothelial cells formed perfused vasculature. Computed tomography scans revealed improved vascular density and decelerated emphysema progression in cell-treated lungs. Proliferation of both human and rat cell was higher in cell-treated versus nontreated controls. Cell treatment reduced alveolar enlargement, improved dynamic compliance and residual volume, and improved diffusion capacity. CONCLUSIONS Our findings suggest that human induced pluripotent stem cell-derived distal lung cells can engraft in emphysematous lungs and participate in the formation of functional distal lung units to ameliorate the progression of emphysema.
Collapse
Affiliation(s)
- Wafa Altalhi
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Mass; Clinical Laboratory Medicine, Faculty of Medical Sciences, Taif University, Taif, Makkah, Saudi Arabia
| | - Tong Wu
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| | | | - Sydney Jeffs
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| | - Kenji Miki
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| | - Harald C Ott
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Mass.
| |
Collapse
|
9
|
Ciminieri C, Woest ME, Reynaert NL, Heijink IH, Wardenaar R, Spierings DCJ, Brandsma CA, Königshoff M, Gosens R. IL-1β Induces a Proinflammatory Fibroblast Microenvironment that Impairs Lung Progenitors' Function. Am J Respir Cell Mol Biol 2023; 68:444-455. [PMID: 36608844 PMCID: PMC12042164 DOI: 10.1165/rcmb.2022-0209oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 01/06/2023] [Indexed: 01/08/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by a persistent inflammatory state in the lungs and defective tissue repair. Although the inflammatory response in patients with COPD is well characterized and known to be exaggerated during exacerbations, its contribution to lung injury and abnormal repair is still unclear. In this study, we aimed to investigate how the inflammatory microenvironment affects the epithelial progenitors and their supporting mesenchymal niche cells involved in tissue repair of the distal lung. We focused on IL-1β, a key inflammatory mediator that is increased during exacerbations of COPD, and used an organoid model of lung epithelial cells and fibroblasts to assess the effect of IL-1β treatment on these cells' transcriptome and secreted factors. Whereas direct treatment of the lung organoids with IL-1β promoted organoid growth, this switched toward inhibition when it was added as fibroblast pretreatment followed by organoid treatment. We then investigated the IL-1β-driven mechanisms in the fibroblasts and found an inflammatory response related to (C-X-C motif) ligand (CXCL) chemokines; we confirmed that these chemokines were responsible for the impaired organoid growth and found that targeting their C-X-C chemokine receptors 1/2 (CXCR1/2) receptors or the IL-1β intracellular signaling reduced the proinflammatory response and restored organoid growth. These data demonstrate that IL-1β alters the fibroblasts' state by promoting a distinct inflammatory response, switching their supportive function on epithelial progenitors toward an inhibitory one in an organoid assay. These results imply that chronic inflammation functions as a shift toward inhibition of repair, thereby contributing to chronic inflammatory diseases like COPD.
Collapse
Affiliation(s)
- Chiara Ciminieri
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD
| | - Manon E Woest
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD
- Aquilo BV, Groningen, The Netherlands
| | - Niki L Reynaert
- Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht University Medical Center, Maastricht, The Netherlands; and
| | - Irene H Heijink
- Groningen Research Institute for Asthma and COPD
- Groningen Department of Pathology and Medical Biology
- Groningen Department of Pulmonary Diseases, and
| | - René Wardenaar
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Diana C J Spierings
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Corry-Anke Brandsma
- Groningen Research Institute for Asthma and COPD
- Groningen Department of Pathology and Medical Biology
| | - Melanie Königshoff
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Reinoud Gosens
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
- Groningen Research Institute for Asthma and COPD
| |
Collapse
|
10
|
Deskin B. IL-1β Signaling in the Alveolar Niche: Context Is Everything. Am J Respir Cell Mol Biol 2023; 68:356-357. [PMID: 36893326 PMCID: PMC10112426 DOI: 10.1165/rcmb.2023-0008ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023] Open
Affiliation(s)
- Brian Deskin
- The John W. Deming Department of Medicine Tulane University School of Medicine New Orleans, Louisiana
| |
Collapse
|
11
|
Tracy EP, Stielberg V, Rowe G, Benson D, Nunes SS, Hoying JB, Murfee WL, LeBlanc AJ. State of the field: cellular and exosomal therapeutic approaches in vascular regeneration. Am J Physiol Heart Circ Physiol 2022; 322:H647-H680. [PMID: 35179976 PMCID: PMC8957327 DOI: 10.1152/ajpheart.00674.2021] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 01/19/2023]
Abstract
Pathologies of the vasculature including the microvasculature are often complex in nature, leading to loss of physiological homeostatic regulation of patency and adequate perfusion to match tissue metabolic demands. Microvascular dysfunction is a key underlying element in the majority of pathologies of failing organs and tissues. Contributing pathological factors to this dysfunction include oxidative stress, mitochondrial dysfunction, endoplasmic reticular (ER) stress, endothelial dysfunction, loss of angiogenic potential and vascular density, and greater senescence and apoptosis. In many clinical settings, current pharmacologic strategies use a single or narrow targeted approach to address symptoms of pathology rather than a comprehensive and multifaceted approach to address their root cause. To address this, efforts have been heavily focused on cellular therapies and cell-free therapies (e.g., exosomes) that can tackle the multifaceted etiology of vascular and microvascular dysfunction. In this review, we discuss 1) the state of the field in terms of common therapeutic cell population isolation techniques, their unique characteristics, and their advantages and disadvantages, 2) common molecular mechanisms of cell therapies to restore vascularization and/or vascular function, 3) arguments for and against allogeneic versus autologous applications of cell therapies, 4) emerging strategies to optimize and enhance cell therapies through priming and preconditioning, and, finally, 5) emerging strategies to bolster therapeutic effect. Relevant and recent clinical and animal studies using cellular therapies to restore vascular function or pathologic tissue health by way of improved vascularization are highlighted throughout these sections.
Collapse
Affiliation(s)
- Evan Paul Tracy
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Virginia Stielberg
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Gabrielle Rowe
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Daniel Benson
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
- Department of Bioengineering, University of Louisville, Louisville, Kentucky
| | - Sara S Nunes
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Heart & Stroke/Richard Lewar Centre of Excellence, University of Toronto, Toronto, Ontario, Canada
| | - James B Hoying
- Advanced Solutions Life Sciences, Manchester, New Hampshire
| | - Walter Lee Murfee
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida
| | - Amanda Jo LeBlanc
- Cardiovascular Innovation Institute and the Department of Physiology, University of Louisville, Louisville, Kentucky
| |
Collapse
|
12
|
Yu H, Lin Y, Zhong Y, Guo X, Lin Y, Yang S, Liu J, Xie X, Sun Y, Wang D, Li B, Ran P, Dai J. Impaired AT2 to AT1 cell transition in PM2.5-induced mouse model of chronic obstructive pulmonary disease. Respir Res 2022; 23:70. [PMID: 35337337 PMCID: PMC8957194 DOI: 10.1186/s12931-022-01996-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/16/2022] [Indexed: 12/12/2022] Open
Abstract
Background Particular matter 2.5 (PM2.5) is one of the most important air pollutant, and it is positively associated with the development of chronic obstructive pulmonary disease (COPD). However, the precise underlying mechanisms through which PM2.5 promotes the development of COPD remains largely unknown.
Methods Mouse alveolar destruction were determined by histological analysis of lung tissues and lung function test. Alveolar type II cells (AT2) to alveolar type I cells (AT1) transition in PM2.5-induced COPD mouse model was confirmed via immunofluorescence staining and qPCR analysis. The differentially expressed genes in PM2.5-induced COPD mouse model were identified by RNA-sequencing of alveolar epithelial organoids and generated by bioinformatics analysis. Results In this study, we found that 6 months exposure of PM2.5 induced a significantly decreased pulmonary compliance and resulted in pulmonary emphysema in mice. We showed that PM2.5 exposure significantly reduced the AT2 to AT1 cell transition in vitro and in vivo. In addition, we found a reduced expression of the intermediate AT2-AT1 cell process marker claudin 4 (CLDN4) at day 4 of differentiation in mouse alveolar organoids treated with PM2.5, suggesting that PM2.5 exposure inhibited AT2 cells from entering the transdifferentiation process. RNA-sequencing of mouse alveolar organoids showed that several key signaling pathways that involved in the AT2 to AT1 cell transition were significantly altered including the Wnt signaling, MAPK signaling and signaling pathways regulating pluripotency of stem cells following PM2.5 exposure. Conclusions In summary, these data demonstrate a critical role of AT2 to AT1 cell transition in PM2.5-induced COPD mouse model and reveal the signaling pathways that potentially regulate AT2 to AT1 cell transition during this process. Our findings therefore advance the current knowledge of PM2.5-induced COPD and may lead to a novel therapeutic strategy to treat this disease. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-01996-w.
Collapse
Affiliation(s)
- Hongjiao Yu
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510000, People's Republic of China
| | - Yingnan Lin
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510000, People's Republic of China
| | - Yue Zhong
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510000, People's Republic of China
| | - Xiaolan Guo
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510000, People's Republic of China
| | - Yuyin Lin
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510000, People's Republic of China
| | - Siqi Yang
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510000, People's Republic of China
| | - Jinglin Liu
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510000, People's Republic of China
| | - Xinran Xie
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510000, People's Republic of China
| | - Yaowei Sun
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510000, People's Republic of China
| | - Dong Wang
- State Key Lab of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, People's Republic of China
| | - Bing Li
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510000, People's Republic of China
| | - Pixin Ran
- State Key Lab of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, People's Republic of China.
| | - Jianwei Dai
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 510000, People's Republic of China. .,State Key Lab of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, People's Republic of China. .,The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Qingyuan, 511500, People's Republic of China.
| |
Collapse
|
13
|
Bernhagen J. A new cytokine target for chronic obstructive pulmonary disease? EBioMedicine 2021; 69:103479. [PMID: 34252695 PMCID: PMC8324801 DOI: 10.1016/j.ebiom.2021.103479] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 11/25/2022] Open
Affiliation(s)
- Jürgen Bernhagen
- Chair of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München (KUM), Ludwig-Maximilians-University (LMU), 81377 Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany; Munich Heart Alliance, 80802 Munich, Germany.
| |
Collapse
|
14
|
Khedoe PPPSJ, Wu X, Gosens R, Hiemstra PS. Repairing damaged lungs using regenerative therapy. Curr Opin Pharmacol 2021; 59:85-94. [PMID: 34161852 PMCID: PMC9188766 DOI: 10.1016/j.coph.2021.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 02/14/2021] [Accepted: 05/12/2021] [Indexed: 12/19/2022]
Abstract
There is an urgent need for better treatment of lung diseases that are a major cause of morbidity and mortality worldwide. This urgency is illustrated by the current COVID-19 health crisis. Moderate-to-extensive lung injury characterizes several lung diseases, and not only therapies that reduce such lung injury are needed but also those that regenerate lung tissue and repair existing lung injury. At present, such therapies are not available, but as a result of a rapid increase in our understanding of lung development and repair, lung regenerative therapies are on the horizon. Here, we discuss existing targets for treatment, as well as novel strategies for development of pharmacological and cell therapy-based regenerative treatment for a variety of lung diseases and clinical studies. We discuss how both patient-relevant in vitro disease models using innovative culture techniques and other advanced new technologies aid in the development of pulmonary regenerative medicine.
Collapse
Affiliation(s)
| | - Xinhui Wu
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Reinoud Gosens
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Groningen, the Netherlands; Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Pieter S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
15
|
Costa R, Wagner DE, Doryab A, De Santis MM, Schorpp K, Rothenaigner I, Lehmann M, Baarsma HA, Liu X, Schmid O, Campillos M, Yildirim AÖ, Hadian K, Königshoff M. A drug screen with approved compounds identifies amlexanox as a novel Wnt/β-catenin activator inducing lung epithelial organoid formation. Br J Pharmacol 2021; 178:4026-4041. [PMID: 34089180 PMCID: PMC8965750 DOI: 10.1111/bph.15581] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 05/04/2021] [Accepted: 05/19/2021] [Indexed: 11/30/2022] Open
Abstract
Background and Purpose: Emphysema is an incurable disease characterized by loss of lung tissue leading to impaired gas exchange. Wnt/β-catenin signalling is reduced in emphysema, and exogenous activation of the pathway in experimental models in vivo and in human ex vivo lung tissue improves lung function and structure. We sought to identify a pharmaceutical able to activate Wnt/β-catenin signalling and assess its potential to activate lung epithelial cells and repair. Experimental Approach: We screened 1216 human-approved compounds for Wnt/β-catenin signalling activation using luciferase reporter cells and selected candidates based on their computationally predicted protein targets. We further performed confirmatory luciferase reporter and metabolic activity assays. Finally, we studied the regenerative potential in murine adult epithelial cell-derived lung organoids and in vivo using a murine elastase-induced emphysema model. Key Results: The primary screen identified 16 compounds that significantly induced Wnt/β-catenin-dependent luciferase activity. Selected compounds activated Wnt/β-catenin signalling without inducing cell toxicity or proliferation. Two compounds were able to promote organoid formation, which was reversed by pharmacological Wnt/β-catenin inhibition, confirming the Wnt/β-catenin-dependent mechanism of action. Amlexanox was used for in vivo evaluation, and preventive treatment resulted in improved lung function and structure in emphysematous mouse lungs. Moreover, gene expression of Hgf, an important alveolar repair marker, was increased, whereas disease marker Eln was decreased, indicating that amlexanox induces proregenerative signalling in emphysema. Conclusion and Implications: Using a drug screen based on Wnt/β-catenin activity, organoid assays and a murine emphysema model, amlexanox was identified as a novel potential therapeutic agent for emphysema.
Collapse
Affiliation(s)
- Rita Costa
- Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München-German Research Center for Environmental Health, Ludwig Maximilian University of Munich, University Hospital Großhadern, Member of the German Center for Lung Research (DZL), Munich, Germany.,Institute of Virology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Darcy E Wagner
- Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München-German Research Center for Environmental Health, Ludwig Maximilian University of Munich, University Hospital Großhadern, Member of the German Center for Lung Research (DZL), Munich, Germany.,Department of Experimental Medical Sciences, Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Stem Cell Centre, Lund University, Lund, Sweden
| | - Ali Doryab
- Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München-German Research Center for Environmental Health, Ludwig Maximilian University of Munich, University Hospital Großhadern, Member of the German Center for Lung Research (DZL), Munich, Germany.,Pulmonary Aerosol Delivery, Institute of Lung Biology and Disease, Comprehensive Pneumology Center, Helmholtz Zentrum München-German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Martina M De Santis
- Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München-German Research Center for Environmental Health, Ludwig Maximilian University of Munich, University Hospital Großhadern, Member of the German Center for Lung Research (DZL), Munich, Germany.,Department of Experimental Medical Sciences, Wallenberg Centre for Molecular Medicine, Faculty of Medicine, Stem Cell Centre, Lund University, Lund, Sweden
| | - Kenji Schorpp
- Assay Development and Screening Platform, Institute for Molecular Toxicology and Pharmacology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Ina Rothenaigner
- Assay Development and Screening Platform, Institute for Molecular Toxicology and Pharmacology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Mareike Lehmann
- Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München-German Research Center for Environmental Health, Ludwig Maximilian University of Munich, University Hospital Großhadern, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Hoeke A Baarsma
- Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München-German Research Center for Environmental Health, Ludwig Maximilian University of Munich, University Hospital Großhadern, Member of the German Center for Lung Research (DZL), Munich, Germany.,Department of Molecular Pharmacology, Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Xueping Liu
- Institute of Structural Biology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Otmar Schmid
- Pulmonary Aerosol Delivery, Institute of Lung Biology and Disease, Comprehensive Pneumology Center, Helmholtz Zentrum München-German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Monica Campillos
- Institute of Structural Biology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Ali Önder Yildirim
- Immunopathology of COPD, Institute of Lung Biology and Disease, Comprehensive Pneumology Center, Helmholtz Zentrum München-German Research Center for Environmental Health, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Kamyar Hadian
- Assay Development and Screening Platform, Institute for Molecular Toxicology and Pharmacology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Melanie Königshoff
- Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München-German Research Center for Environmental Health, Ludwig Maximilian University of Munich, University Hospital Großhadern, Member of the German Center for Lung Research (DZL), Munich, Germany.,Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
16
|
AbdelMassih AF, Ashraf A, Ismail HA, AbdelAzeim B, Barsoum IH, Girgis S, Afdal G, AbdelAzeim N, Afdal P, Menshawey E, Menshawey R, Badr K, Arsanyous M. Effect of pioglitazone, as antidiabetic agent, on atheroma regression in type 2 diabetic patients: a systematic review and meta-analysis. BENI-SUEF UNIVERSITY JOURNAL OF BASIC AND APPLIED SCIENCES 2021. [DOI: 10.1186/s43088-021-00096-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Pioglitazone’s role in the induction of atheroma regression in diabetics was suggested by several RCT. The aim of our study was to evaluate this role through a systematic review of all RCT conducted on this subject.
Methods
Literature was searched for relevant studies. We included all RCT that compared pioglitazone versus other antidiabetic agents. Mean differences of either AV or CIMT, HbA1C, HDL, and LDL between the two groups were used to assess the effect of pioglitazone versus alternative therapies.
Results
Six RCT were included with a total of 1180 patients. Pioglitazone was significantly superior to glimepiride and gliclazide in improving IMT. No significant difference was observed in overall AV, HbA1C, and LDL.
Conclusion
The latter findings confirm that anti-atheroma action of pioglitazone is not achieved through its antiglycemic or antidyslipidemia effects, but probably through a DNA-mediated effect, and may lead to its repurposing for reversal of organ fibrosis.
Collapse
|
17
|
Barnes PJ, Anderson GP, Fagerås M, Belvisi MG. Chronic lung diseases: prospects for regeneration and repair. Eur Respir Rev 2021; 30:30/159/200213. [PMID: 33408088 PMCID: PMC9488945 DOI: 10.1183/16000617.0213-2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/17/2020] [Indexed: 12/17/2022] Open
Abstract
COPD and idiopathic pulmonary fibrosis (IPF) together represent a considerable unmet medical need, and advances in their treatment lag well behind those of other chronic conditions. Both diseases involve maladaptive repair mechanisms leading to progressive and irreversible damage. However, our understanding of the complex underlying disease mechanisms is incomplete; with current diagnostic approaches, COPD and IPF are often discovered at an advanced stage and existing definitions of COPD and IPF can be misleading. To halt or reverse disease progression and achieve lung regeneration, there is a need for earlier identification and treatment of these diseases. A precision medicine approach to treatment is also important, involving the recognition of disease subtypes, or endotypes, according to underlying disease mechanisms, rather than the current “one-size-fits-all” approach. This review is based on discussions at a meeting involving 38 leading global experts in chronic lung disease mechanisms, and describes advances in the understanding of the pathology and molecular mechanisms of COPD and IPF to identify potential targets for reversing disease degeneration and promoting tissue repair and lung regeneration. We also discuss limitations of existing disease measures, technical advances in understanding disease pathology, and novel methods for targeted drug delivery. Treatment outcomes with COPD and IPF are suboptimal. Better understanding of the diseases, such as targetable repair mechanisms, may generate novel therapies, and earlier diagnosis and treatment is needed to stop or even reverse disease progression.https://bit.ly/2Ga8J1g
Collapse
Affiliation(s)
- Peter J Barnes
- National Heart & Lung Institute, Imperial College London, London, UK
| | - Gary P Anderson
- Lung Health Research Centre, University of Melbourne, Melbourne, Australia
| | | | - Maria G Belvisi
- National Heart & Lung Institute, Imperial College London, London, UK.,Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
18
|
Nguyen JMK, Robinson DN, Sidhaye VK. Why new biology must be uncovered to advance therapeutic strategies for chronic obstructive pulmonary disease. Am J Physiol Lung Cell Mol Physiol 2021; 320:L1-L11. [PMID: 33174444 PMCID: PMC7847061 DOI: 10.1152/ajplung.00367.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/22/2020] [Accepted: 11/06/2020] [Indexed: 12/13/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by the destruction of alveolar tissue (in emphysema) and airway remodeling (leading to chronic bronchitis), which cause difficulties in breathing. It is a growing public health concern with few therapeutic options that can reverse disease progression or mortality. This is in part because current treatments mainly focus on ameliorating symptoms induced by inflammatory pathways as opposed to curing disease. Hence, emerging research focused on upstream pathways are likely to be beneficial in the development of efficient therapeutics to address the root causes of disease. Some of these pathways include mitochondrial function, cytoskeletal structure and maintenance, and airway hydration, which are all affected by toxins that contribute to COPD. Because of the complexity of COPD and unknown targets for disease onset, simpler model organisms have proved to be useful tools in identifying disease-relevant pathways and targets. This review summarizes COPD pathology, current treatments, and therapeutic discovery research, with a focus on the aforementioned pathways that can advance the therapeutic landscape of COPD.
Collapse
Affiliation(s)
- Jennifer M K Nguyen
- Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Douglas N Robinson
- Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | | |
Collapse
|
19
|
Extracorporeal Shock Waves Increase Markers of Cellular Proliferation in Bronchial Epithelium and in Primary Bronchial Fibroblasts of COPD Patients. Can Respir J 2020; 2020:1524716. [PMID: 32831979 PMCID: PMC7429777 DOI: 10.1155/2020/1524716] [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: 12/12/2019] [Revised: 04/02/2020] [Accepted: 04/08/2020] [Indexed: 11/18/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is due to structural changes and narrowing of small airways and parenchymal destruction (loss of the alveolar attachment as a result of pulmonary emphysema), which all lead to airflow limitation. Extracorporeal shock waves (ESW) increase cell proliferation and differentiation of connective tissue fibroblasts. To date no studies are available on ESW treatment of human bronchial fibroblasts and epithelial cells from COPD and control subjects. We obtained primary bronchial fibroblasts from bronchial biopsies of 3 patients with mild/moderate COPD and 3 control smokers with normal lung function. 16HBE cells were also studied. Cells were treated with a piezoelectric shock wave generator at low energy (0.3 mJ/mm2, 500 pulses). After treatment, viability was evaluated and cells were recultured and followed up for 4, 24, 48, and 72 h. Cell growth (WST-1 test) was assessed, and proliferation markers were analyzed by qRT-PCR in cell lysates and by ELISA tests in cell supernatants and cell lysates. After ESW treatment, we observed a significant increase of cell proliferation in all cell types. C-Kit (CD117) mRNA was significantly increased in 16HBE cells at 4 h. Protein levels were significantly increased for c-Kit (CD117) at 4 h in 16HBE (p < 0.0001) and at 24 h in COPD-fibroblasts (p = 0.037); for PCNA at 4 h in 16HBE (p = 0.046); for Thy1 (CD90) at 24 and 72 h in CS-fibroblasts (p = 0.031 and p = 0.041); for TGFβ1 at 72 h in CS-fibroblasts (p = 0.038); for procollagen-1 at 4 h in COPD-fibroblasts (p = 0.020); and for NF-κB-p65 at 4 and 24 h in 16HBE (p = 0.015 and p = 0.0002). In the peripheral lung tissue of a representative COPD patient, alveolar type II epithelial cells (TTF‐1+) coexpressing c-Kit (CD117) and PCNA were occasionally observed. These data show an increase of cell proliferation induced by a low dosage of extracorporeal shock waves in 16HBE cells and primary bronchial fibroblasts of COPD and control smoking subjects.
Collapse
|
20
|
Manevski M, Muthumalage T, Devadoss D, Sundar IK, Wang Q, Singh KP, Unwalla HJ, Chand HS, Rahman I. Cellular stress responses and dysfunctional Mitochondrial-cellular senescence, and therapeutics in chronic respiratory diseases. Redox Biol 2020; 33:101443. [PMID: 32037306 PMCID: PMC7251248 DOI: 10.1016/j.redox.2020.101443] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 01/14/2020] [Accepted: 01/22/2020] [Indexed: 02/06/2023] Open
Abstract
The abnormal inflammatory responses due to the lung tissue damage and ineffective repair/resolution in response to the inhaled toxicants result in the pathological changes associated with chronic respiratory diseases. Investigation of such pathophysiological mechanisms provides the opportunity to develop the molecular phenotype-specific diagnostic assays and could help in designing the personalized medicine-based therapeutic approaches against these prevalent diseases. As the central hubs of cell metabolism and energetics, mitochondria integrate cellular responses and interorganellar signaling pathways to maintain cellular and extracellular redox status and the cellular senescence that dictate the lung tissue responses. Specifically, as observed in chronic obstructive pulmonary disease (COPD) and pulmonary fibrosis, the mitochondria-endoplasmic reticulum (ER) crosstalk is disrupted by the inhaled toxicants such as the combustible and emerging electronic nicotine-delivery system (ENDS) tobacco products. Thus, the recent research efforts have focused on understanding how the mitochondria-ER dysfunctions and oxidative stress responses can be targeted to improve inflammatory and cellular dysfunctions associated with these pathologic illnesses that are exacerbated by viral infections. The present review assesses the importance of these redox signaling and cellular senescence pathways that describe the role of mitochondria and ER on the development and function of lung epithelial responses, highlighting the cause and effect associations that reflect the disease pathogenesis and possible intervention strategies.
Collapse
Affiliation(s)
- Marko Manevski
- Department of Immunology and NanoMedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Thivanka Muthumalage
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Dinesh Devadoss
- Department of Immunology and NanoMedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Isaac K Sundar
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Qixin Wang
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Kameshwar P Singh
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Hoshang J Unwalla
- Department of Immunology and NanoMedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Hitendra S Chand
- Department of Immunology and NanoMedicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
| | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
21
|
Basil MC, Morrisey EE. Lung regeneration: a tale of mice and men. Semin Cell Dev Biol 2020; 100:88-100. [PMID: 31761445 PMCID: PMC7909713 DOI: 10.1016/j.semcdb.2019.11.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 11/06/2019] [Accepted: 11/11/2019] [Indexed: 01/11/2023]
Abstract
The respiratory system is the main site of gas exchange with the external environment in complex terrestrial animals. Within the trachea and lungs are multiple different tissue niches each consisting of a myriad of cells types with critical roles in air conduction, gas exchange, providing important niche specific cell-cell interactions, connection to the cardiovascular system, and immune surveillance. How the respiratory system responds to external insults and executes the appropriate regenerative response remains challenging to study given the plethora of cell and tissue interactions for this to occur properly. This review will examine the various cell types and tissue niches found within the respiratory system and provide a comparison between mouse and human lungs and trachea to highlight important similarities and differences. Defining the critical gaps in knowledge in human lung and tracheal regeneration is critical for future development of therapies directed towards respiratory diseases.
Collapse
Affiliation(s)
- Maria C Basil
- Department of Medicine; Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Edward E Morrisey
- Department of Medicine; Department of Cell and Developmental Biology; Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, United States.
| |
Collapse
|
22
|
Fernandes-Silva H, Araújo-Silva H, Correia-Pinto J, Moura RS. Retinoic Acid: A Key Regulator of Lung Development. Biomolecules 2020; 10:biom10010152. [PMID: 31963453 PMCID: PMC7022928 DOI: 10.3390/biom10010152] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/14/2020] [Accepted: 01/15/2020] [Indexed: 12/14/2022] Open
Abstract
Retinoic acid (RA) is a key molecular player in embryogenesis and adult tissue homeostasis. In embryo development, RA plays a crucial role in the formation of different organ systems, namely, the respiratory system. During lung development, there is a spatiotemporal regulation of RA levels that assures the formation of a fully functional organ. RA signaling influences lung specification, branching morphogenesis, and alveolarization by regulating the expression of particular target genes. Moreover, cooperation with other developmental pathways is essential to shape lung organogenesis. This review focuses on the events regulated by retinoic acid during lung developmental phases and pulmonary vascular development; also, it aims to provide a snapshot of RA interplay with other well-known regulators of lung development.
Collapse
Affiliation(s)
- Hugo Fernandes-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (H.F.-S.); (H.A.-S.); (J.C.-P.)
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
- PhDOC PhD Program, ICVS/3B’s, School of Medicine, University of Minho, 4710-057 Braga, Portugal
| | - Henrique Araújo-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (H.F.-S.); (H.A.-S.); (J.C.-P.)
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Jorge Correia-Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (H.F.-S.); (H.A.-S.); (J.C.-P.)
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
- Department of Pediatric Surgery, Hospital of Braga, 4710-243 Braga, Portugal
| | - Rute S Moura
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal; (H.F.-S.); (H.A.-S.); (J.C.-P.)
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
- Correspondence: ; Tel.: +35-12-5360-4911
| |
Collapse
|
23
|
Hadzic S, Wu CY, Avdeev S, Weissmann N, Schermuly RT, Kosanovic D. Lung epithelium damage in COPD - An unstoppable pathological event? Cell Signal 2020; 68:109540. [PMID: 31953012 DOI: 10.1016/j.cellsig.2020.109540] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 01/11/2020] [Accepted: 01/11/2020] [Indexed: 10/25/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a common term for alveolar septal wall destruction resulting in emphysema, and chronic bronchitis accompanied by conductive airway remodelling. In general, this disease is characterized by a disbalance of proteolytic/anti-proteolytic activity, augmented inflammatory response, increased oxidative/nitrosative stress, rise in number of apoptotic cells and decreased proliferation. As the first responder to the various environmental stimuli, epithelium occupies an important position in different lung pathologies, including COPD. Epithelium sequentially transitions from the upper airways in the direction of the gas exchange surface in the alveoli, and every cell type possesses a distinct role in the maintenance of the homeostasis. Basically, a thick ciliated structure of the airway epithelium has a major function in mucus secretion, whereas, alveolar epithelium which forms a thin barrier covered by surfactant has a function in gas exchange. Following this line, we will try to reveal whether or not the chronic bronchitis and emphysema, being two pathological phenotypes in COPD, could originate in two different types of epithelium. In addition, this review focuses on the role of lung epithelium in COPD pathology, and summarises underlying mechanisms and potential therapeutics.
Collapse
Affiliation(s)
- Stefan Hadzic
- Department of Internal Medicine, Cardio-Pulmonary Institute (CPI), German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany
| | - Cheng-Yu Wu
- Department of Internal Medicine, Cardio-Pulmonary Institute (CPI), German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany
| | - Sergey Avdeev
- Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Norbert Weissmann
- Department of Internal Medicine, Cardio-Pulmonary Institute (CPI), German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany
| | - Ralph Theo Schermuly
- Department of Internal Medicine, Cardio-Pulmonary Institute (CPI), German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany
| | - Djuro Kosanovic
- Department of Internal Medicine, Cardio-Pulmonary Institute (CPI), German Center for Lung Research (DZL), Justus-Liebig University, Giessen, Germany; Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia.
| |
Collapse
|
24
|
Wu X, van Dijk EM, Ng-Blichfeldt JP, Bos IST, Ciminieri C, Königshoff M, Kistemaker LEM, Gosens R. Mesenchymal WNT-5A/5B Signaling Represses Lung Alveolar Epithelial Progenitors. Cells 2019; 8:cells8101147. [PMID: 31557955 PMCID: PMC6829372 DOI: 10.3390/cells8101147] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 09/17/2019] [Accepted: 09/25/2019] [Indexed: 01/23/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) represents a worldwide concern with high morbidity and mortality, and is believed to be associated with accelerated ageing of the lung. Alveolar abnormalities leading to emphysema are a key characteristic of COPD. Pulmonary alveolar epithelial type 2 cells (AT2) produce surfactant and function as progenitors for type 1 cells. Increasing evidence shows elevated WNT-5A/B expression in ageing and in COPD that may contribute to the disease process. However, supportive roles for WNT-5A/B in lung regeneration were also reported in different studies. Thus, we explored the role of WNT-5A/B on alveolar epithelial progenitors (AEPs) in more detail. We established a Precision-Cut-Lung Slices (PCLS) model and a lung organoid model by co-culturing epithelial cells (EpCAM+/CD45-/CD31-) with fibroblasts in matrigel in vitro to study the impact of WNT-5A and WNT-5B. Our results show that WNT-5A and WNT-5B repress the growth of epithelial progenitors with WNT-5B preferentially restraining the growth and differentiation of alveolar epithelial progenitors. We provide evidence that both WNT-5A and WNT-5B negatively regulate the canonical WNT signaling pathway in alveolar epithelium. Taken together, these findings reveal the functional impact of WNT-5A/5B signaling on alveolar epithelial progenitors in the lung, which may contribute to defective alveolar repair in COPD.
Collapse
Affiliation(s)
- Xinhui Wu
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713AV Groningen, The Netherlands.
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands.
| | - Eline M van Dijk
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713AV Groningen, The Netherlands.
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands.
| | - John-Poul Ng-Blichfeldt
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713AV Groningen, The Netherlands.
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands.
| | - I Sophie T Bos
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713AV Groningen, The Netherlands.
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands.
| | - Chiara Ciminieri
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713AV Groningen, The Netherlands.
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands.
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado, CO 80045 Aurora, USA.
| | - Melanie Königshoff
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado, CO 80045 Aurora, USA.
| | - Loes E M Kistemaker
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713AV Groningen, The Netherlands.
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands.
- Aquilo BV, 9713 AV Groningen, The Netherlands.
| | - Reinoud Gosens
- Department of Molecular Pharmacology, Faculty of Science and Engineering, University of Groningen, Antonius Deusinglaan 1, 9713AV Groningen, The Netherlands.
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands.
| |
Collapse
|
25
|
Barreiro E, Wang X, Tang J. COPD: preclinical models and emerging therapeutic targets. Expert Opin Ther Targets 2019; 23:829-838. [DOI: 10.1080/14728222.2019.1667976] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Esther Barreiro
- Respiratory Medicine Department, Muscle Wasting and Cachexia in Chronic Respiratory Diseases and Lung Cancer Research Group, Institute of Medical Research of Hospital del Mar (IMIM)-Hospital del Mar, Parc de Salut Mar, Barcelona Biomedical Research Park (PRBB), Barcelona, Spain
- Department of Health Sciences (CEXS), Universitat Pompeu Fabra, Barcelona, Spain
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), Barcelona, Spain
| | - Xuejie Wang
- Respiratory Medicine Department, Muscle Wasting and Cachexia in Chronic Respiratory Diseases and Lung Cancer Research Group, Institute of Medical Research of Hospital del Mar (IMIM)-Hospital del Mar, Parc de Salut Mar, Barcelona Biomedical Research Park (PRBB), Barcelona, Spain
- Department of Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Jun Tang
- Respiratory Medicine Department, Muscle Wasting and Cachexia in Chronic Respiratory Diseases and Lung Cancer Research Group, Institute of Medical Research of Hospital del Mar (IMIM)-Hospital del Mar, Parc de Salut Mar, Barcelona Biomedical Research Park (PRBB), Barcelona, Spain
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), Barcelona, Spain
- Department of Medicine, Autonomous University of Barcelona, Barcelona, Spain
| |
Collapse
|