1
|
Zhou H, Zhang Q, Liu C, Fan J, Huang W, Li N, Yang M, Wang H, Xie W, Kong H. NLRP3 inflammasome mediates abnormal epithelial regeneration and distal lung remodeling in silica‑induced lung fibrosis. Int J Mol Med 2024; 53:25. [PMID: 38240085 PMCID: PMC10836498 DOI: 10.3892/ijmm.2024.5349] [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: 09/06/2023] [Accepted: 12/28/2023] [Indexed: 01/23/2024] Open
Abstract
NOD-like receptor protein 3 (NLRP3) inflammasome is closely related to silica particle‑induced chronic lung inflammation but its role in epithelial remodeling, repair and regeneration in the distal lung during development of silicosis remains to be elucidated. The present study aimed to determine the effects of the NLRP3 inflammasome on epithelial remodeling and cellular regeneration and potential mechanisms in the distal lung of silica‑treated mice at three time points. Pulmonary function assessment, inflammatory cell counting, enzyme‑linked immunosorbent assay, histological and immunological analyses, hydroxyproline assay and western blotting were used in the study. Single intratracheal instillation of a silica suspension caused sustained NLRP3 inflammasome activation in the distal lung. Moreover, a time‑dependent increase in airway resistance and a decrease in lung compliance accompanied progression of pulmonary fibrosis. In the terminal bronchiole, lung remodeling including pyroptosis (membrane‑distributed GSDMD+), excessive proliferation (Ki67+), mucus overproduction (mucin 5 subtype AC and B) and epithelial‑mesenchymal transition (decreased E‑Cadherin+ and increased Vimentin+), was observed by immunofluorescence analysis. Notably, aberrant spatiotemporal expression of the embryonic lung stem/progenitor cell markers SOX2 and SOX9 and ectopic distribution of bronchioalveolar stem cells were observed in the distal lung only on the 7th day after silica instillation (the early inflammatory phase of silicosis). Western blotting revealed that the Sonic hedgehog/Glioma‑associated oncogene (Shh/Gli) and Wnt/β‑catenin pathways were involved in NLRP3 inflammasome activation‑mediated epithelial remodeling and dysregulated regeneration during the inflammatory and fibrotic phases. Overall, sustained NLRP3 inflammasome activation led to epithelial remodeling in the distal lung of mice. Moreover, understanding the spatiotemporal profile of dysregulated epithelial repair and regeneration may provide a novel therapeutic strategy for inhalable particle‑related chronic inflammatory and fibrotic lung disease.
Collapse
Affiliation(s)
- Hong Zhou
- Department of Pulmonary and Critical Care Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Qun Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Chenyang Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Jiahao Fan
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Wen Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Nan Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Mingxia Yang
- Department of Pulmonary and Critical Care Medicine, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu 213003, P.R. China
| | - Hong Wang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Weiping Xie
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Hui Kong
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
2
|
Tsikis ST, Klouda T, Hirsch TI, Fligor SC, Liu T, Kim Y, Pan A, Quigley M, Mitchell PD, Puder M, Yuan K. A pneumonectomy model to study flow-induced pulmonary hypertension and compensatory lung growth. CELL REPORTS METHODS 2023; 3:100613. [PMID: 37827157 PMCID: PMC10626210 DOI: 10.1016/j.crmeth.2023.100613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 08/01/2023] [Accepted: 09/20/2023] [Indexed: 10/14/2023]
Abstract
In newborns, developmental disorders such as congenital diaphragmatic hernia (CDH) and specific types of congenital heart disease (CHD) can lead to defective alveolarization, pulmonary hypoplasia, and pulmonary arterial hypertension (PAH). Therapeutic options for these patients are limited, emphasizing the need for new animal models representative of disease conditions. In most adult mammals, compensatory lung growth (CLG) occurs after pneumonectomy; however, the underlying relationship between CLG and flow-induced pulmonary hypertension (PH) is not fully understood. We propose a murine model that involves the simultaneous removal of the left lung and right caval lobe (extended pneumonectomy), which results in reduced CLG and exacerbated reproducible PH. Extended pneumonectomy in mice is a promising animal model to study the cellular response and molecular mechanisms contributing to flow-induced PH, with the potential to identify new treatments for patients with CDH or PAH-CHD.
Collapse
Affiliation(s)
- Savas T Tsikis
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Fegan 3, Boston, MA 02115, USA
| | - Timothy Klouda
- Division of Pulmonary Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Thomas I Hirsch
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Fegan 3, Boston, MA 02115, USA
| | - Scott C Fligor
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Fegan 3, Boston, MA 02115, USA
| | - Tiffany Liu
- Division of Pulmonary Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Yunhye Kim
- Division of Pulmonary Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Amy Pan
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Fegan 3, Boston, MA 02115, USA
| | - Mikayla Quigley
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Fegan 3, Boston, MA 02115, USA
| | - Paul D Mitchell
- Institutional Centers for Clinical and Translational Research, Boston Children's Hospital, Boston, MA 02115, USA
| | - Mark Puder
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Surgery, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Fegan 3, Boston, MA 02115, USA.
| | - Ke Yuan
- Division of Pulmonary Medicine, Boston Children's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
3
|
Redente EF, Kopf KW, Bahadur AN, Robichaud A, Lundblad LK, McDonald LT. Application-specific approaches to MicroCT for evaluation of mouse models of pulmonary disease. PLoS One 2023; 18:e0281452. [PMID: 36757935 PMCID: PMC9910664 DOI: 10.1371/journal.pone.0281452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 01/23/2023] [Indexed: 02/10/2023] Open
Abstract
The advent of micro-computed tomography (microCT) has provided significant advancement in our ability to generate clinically relevant assessments of lung health and disease in small animal models. As microCT use to generate outcomes analysis in pulmonary preclinical models has increased there have been substantial improvements in image quality and resolution, and data analysis software. However, there are limited published methods for standardized imaging and automated analysis available for investigators. Manual quantitative analysis of microCT images is complicated by the presence of inflammation and parenchymal disease. To improve the efficiency and limit user-associated bias, we have developed an automated pulmonary air and tissue segmentation (PATS) task list to segment lung air volume and lung tissue volume for quantitative analysis. We demonstrate the effective use of the PATS task list using four distinct methods for imaging, 1) in vivo respiration controlled scanning using a flexiVent, 2) longitudinal breath-gated in vivo scanning in resolving and non-resolving pulmonary disease initiated by lipopolysaccharide-, bleomycin-, and silica-exposure, 3) post-mortem imaging, and 4) ex vivo high-resolution scanning. The accuracy of the PATS task list was compared to manual segmentation. The use of these imaging techniques and automated quantification methodology across multiple models of lung injury and fibrosis demonstrates the broad applicability and adaptability of microCT to various lung diseases and small animal models and presents a significant advance in efficiency and standardization of preclinical microCT imaging and analysis for the field of pulmonary research.
Collapse
Affiliation(s)
- Elizabeth F. Redente
- Department of Pediatrics, National Jewish Health, Denver, Colorado, United States of America
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
- * E-mail:
| | - Katrina W. Kopf
- Department of Academic Affairs, National Jewish Health, Denver, Colorado, United States of America
| | - Ali N. Bahadur
- Bruker BioSpin Corporation, Billerica, Massachusetts, United States of America
| | | | | | - Lindsay T. McDonald
- Ralph H. Johnson VA Medical Center, Charleston, South Carolina, United States of America
| |
Collapse
|
4
|
Zhuang Y, Yang W, Zhang L, Fan C, Qiu L, Zhao Y, Chen B, Chen Y, Shen H, Dai J. A novel leptin receptor binding peptide tethered-collagen scaffold promotes lung injury repair. Biomaterials 2022; 291:121884. [DOI: 10.1016/j.biomaterials.2022.121884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 10/10/2022] [Accepted: 10/23/2022] [Indexed: 11/06/2022]
|
5
|
Jones-Freeman B, Starkey MR. Bronchioalveolar stem cells in lung repair, regeneration and disease. J Pathol 2020; 252:219-226. [PMID: 32737996 DOI: 10.1002/path.5527] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/08/2020] [Accepted: 07/26/2020] [Indexed: 12/17/2022]
Abstract
Bronchioalveolar stem cells (BASCs) are a lung resident stem cell population located at bronchioalveolar duct junctions that contribute to the maintenance of bronchiolar club cells and alveolar epithelial cells of the distal lung. Their transformed counterparts are considered to be likely progenitors of lung adenocarcinomas, which has been a major area of research in relation to BASCs. A critical limitation in addressing the function of BASCs in vivo has been the lack of a unique BASC marker, which has prevented specific targeting of BASCs in animal models of respiratory conditions. Recently, there have been several studies describing genetically modified mice that allow in vivo quantification, tracing, and functional analysis of BASCs to address this long-standing issue. These cutting-edge experimental tools will likely have significant implications for future experimental studies involving BASCs and the elucidation of their role in various lung diseases. To date, this has been largely explored in models of lung injury including naphthalene-induced airway injury, bleomycin-induced alveolar injury, hyperoxia-induced models of bronchopulmonary dysplasia, and influenza virus infection. These novel experimental mouse tools will facilitate the assessment of the impact of BASC loss on additional respiratory conditions including infection-induced severe asthma and chronic obstructive pulmonary disease, as well as respiratory bacterial infections, both in early life and adulthood. These future studies may shed light on the potential broad applicability of targeting BASCs for a diverse range of respiratory conditions during lung development and in promoting effective regeneration and repair of the lung in respiratory diseases. © 2020 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Bernadette Jones-Freeman
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Australia
| | - Malcolm R Starkey
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Australia
| |
Collapse
|
6
|
Dane DM, Cao K, Zhang YA, H Kernstine K, Gazdhar A, Geiser T, Hsia CCW. Inhalational delivery of induced pluripotent stem cell secretome improves postpneumonectomy lung structure and function. J Appl Physiol (1985) 2020; 129:1051-1061. [PMID: 32909918 DOI: 10.1152/japplphysiol.00205.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cell-free secretory products (secretome) of human induced pluripotent stem cells (iPSCs) have been shown to attenuate tissue injury and facilitate repair and recovery. To examine whether iPSC secretome facilitates mechanically induced compensatory responses following unilateral pneumonectomy (PNX), litter-matched young adult female hounds underwent right PNX (removing 55%-58% of lung units), followed by inhalational delivery of either the nebulized-conditioned media containing induced pluripotent stem cell secretome (iPSC CM) or control cell-free media (CFM); inhalation was repeated every 5 days for 10 treatments. Lung function was measured under anesthesia pre-PNX and 10 days after the last treatment (8 wk post-PNX); detailed quantitative analysis of lung ultrastructure was performed postmortem. Pre-PNX lung function was similar between groups. Compared with CFM control, treatment with iPSC CM attenuated the post-PNX decline in lung diffusing capacity for carbon monoxide and membrane diffusing capacity, accompanied by a 24% larger postmortem lobar volume and distal air spaces. Alveolar double-capillary profiles were 39% more prevalent consistent with enhanced intussusceptive angiogenesis. Frequency distribution of the harmonic mean thickness of alveolar blood-gas barrier shifted toward the lowest values, whereas alveolar septal tissue volume and arithmetic septal thickness were similar, indicating septal remodeling and reduced diffusive resistance of the blood-gas barrier. Thus, repetitive inhalational delivery of iPSC secretome enhanced post-PNX alveolar angiogenesis and septal remodeling that are associated with improved gas exchange compensation. Results highlight the plasticity of the remaining lung units following major loss of lung mass that are responsive to broad-based modulation provided by the iPSC secretome.NEW & NOTEWORTHY To examine whether the secreted products of human induced pluripotent stem cells (iPSCs) facilitate innate adaptive responses following loss of lung tissue, adult dogs underwent surgical removal of one lung, then received repeated administration of iPSC secretory products via inhalational delivery compared with control treatment. Inhalation of iPSC secretory products enhanced capillary formation and beneficial structural remodeling in the remaining lung, leading to improved lung function.
Collapse
Affiliation(s)
- D Merrill Dane
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Khoa Cao
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Yu-An Zhang
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Kemp H Kernstine
- Department of Cardiothoracic and Thoracic Surgery, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Amiq Gazdhar
- Department of Pulmonary Medicine, University of Bern, Bern, Switzerland.,Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Thomas Geiser
- Department of Pulmonary Medicine, University of Bern, Bern, Switzerland.,Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Connie C W Hsia
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
7
|
Morales-Hernández A, Nacarino-Palma A, Moreno-Marín N, Barrasa E, Paniagua-Quiñones B, Catalina-Fernández I, Alvarez-Barrientos A, Bustelo XR, Merino JM, Fernández-Salguero PM. Lung regeneration after toxic injury is improved in absence of dioxin receptor. Stem Cell Res 2017; 25:61-71. [PMID: 29107893 DOI: 10.1016/j.scr.2017.10.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 08/07/2017] [Accepted: 10/09/2017] [Indexed: 12/26/2022] Open
Abstract
Recent experimental evidences from cellular systems and from mammalian and non-mammalian animal models highlight novel functions for the aryl hydrocarbon/dioxin receptor (AhR) in maintaining cell differentiation and tissue homeostasis. Notably, AhR depletion stimulates an undifferentiated and pluripotent phenotype likely associated to a mesenchymal transition in epithelial cells and to increased primary tumorigenesis and metastasis in melanoma. In this work, we have used a lung model of epithelial regeneration to investigate whether AhR regulates proper tissue repair by adjusting the expansion of undifferentiated stem-like cells. AhR-null mice developed a faster and more efficient repair of the lung bronchiolar epithelium upon naphthalene injury that required increased cell proliferation and the earlier activation of stem-like Clara, Basal and neuroepithelial cells precursors. Increased basal content in multipotent Sca1+/CD31-/CD4- cells and in cells expressing pluripotency factors NANOG and OCT4 could also improve re-epithelialization in AhR-null lungs. The reduced response of AhR-deficient lungs to Sonic Hedgehog (Shh) repression shortly after injury may also help their improved bronchiolar epithelium repair. These results support a role for AhR in the regenerative response against toxins, and open the possibility of modulating its activation level to favor recovery from lesions caused by environmental contaminants.
Collapse
Affiliation(s)
| | - Ana Nacarino-Palma
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, Universidad de Extremadura, 06071 Badajoz, Spain
| | - Nuria Moreno-Marín
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, Universidad de Extremadura, 06071 Badajoz, Spain
| | - Eva Barrasa
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, Universidad de Extremadura, 06071 Badajoz, Spain
| | - Beroé Paniagua-Quiñones
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, Universidad de Extremadura, 06071 Badajoz, Spain
| | | | | | - Xosé R Bustelo
- Centro de Investigación del Cáncer and CIBERONC, CSIC-Universidad de Salamanca, 37007 Salamanca, Spain
| | - Jaime M Merino
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, Universidad de Extremadura, 06071 Badajoz, Spain.
| | - Pedro M Fernández-Salguero
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias, Universidad de Extremadura, 06071 Badajoz, Spain.
| |
Collapse
|
8
|
Dioxin Receptor Adjusts Liver Regeneration After Acute Toxic Injury and Protects Against Liver Carcinogenesis. Sci Rep 2017; 7:10420. [PMID: 28874739 PMCID: PMC5585208 DOI: 10.1038/s41598-017-10984-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 08/17/2017] [Indexed: 12/15/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR) has roles in cell proliferation, differentiation and organ homeostasis, including the liver. AhR depletion induces undifferentiation and pluripotency in normal and transformed cells. Here, AhR-null mice (AhR-/-) were used to explore whether AhR controls liver regeneration and carcinogenesis by restricting the expansion of stem-like cells and the expression of pluripotency genes. Short-term CCl4 liver damage was earlier and more efficiently repaired in AhR-/- than in AhR+/+ mice. Stem-like CK14 + and TBX3 + and pluripotency-expressing OCT4 + and NANOG + cells expanded sooner in AhR-/- than in AhR+/+ regenerating livers. Stem-like side population cells (SP) isolated from AhR-/- livers had increased β-catenin (β-Cat) signaling with overexpression of Axin2, Dkk1 and Cyclin D1. Interestingly, β-Cat, Axin2 and Dkk1 also increased during regeneration but more notably in AhR-null livers. Liver carcinogenesis induced by diethylnitrosamine (DEN) produced large carcinomas in all AhR-/- mice but mostly premalignant adenomas in less than half of AhR+/+ mice. AhR-null tumoral tissue, but not their surrounding non-tumoral parenchyma, had nuclear β-Cat and Axin2 overexpression. OCT4 and NANOG were nevertheless similarly expressed in AhR+/+ and AhR-/- lesions. We suggest that AhR may serve to adjust liver repair and to block tumorigenesis by modulating stem-like cells and β-Cat signaling.
Collapse
|
9
|
Dinh PUC, Cores J, Hensley MT, Vandergriff AC, Tang J, Allen TA, Caranasos TG, Adler KB, Lobo LJ, Cheng K. Derivation of therapeutic lung spheroid cells from minimally invasive transbronchial pulmonary biopsies. Respir Res 2017; 18:132. [PMID: 28666430 PMCID: PMC5493087 DOI: 10.1186/s12931-017-0611-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 06/12/2017] [Indexed: 12/21/2022] Open
Abstract
Background Resident stem and progenitor cells have been identified in the lung over the last decade, but isolation and culture of these cells remains a challenge. Thus, although these lung stem and progenitor cells provide an ideal source for stem-cell based therapy, mesenchymal stem cells (MSCs) remain the most popular cell therapy product for the treatment of lung diseases. Surgical lung biopsies can be the tissue source but such procedures carry a high risk of mortality. Methods In this study we demonstrate that therapeutic lung cells, termed “lung spheroid cells” (LSCs) can be generated from minimally invasive transbronchial lung biopsies using a three-dimensional culture technique. The cells were then characterized by flow cytometry and immunohistochemistry. Angiogenic potential was tested by in-vitro HUVEC tube formation assay. In-vivo bio- distribution of LSCs was examined in athymic nude mice after intravenous delivery. Results From one lung biopsy, we are able to derive >50 million LSC cells at Passage 2. These cells were characterized by flow cytometry and immunohistochemistry and were shown to represent a mixture of lung stem cells and supporting cells. When introduced systemically into nude mice, LSCs were retained primarily in the lungs for up to 21 days. Conclusion Here, for the first time, we demonstrated that direct culture and expansion of human lung progenitor cells from pulmonary tissues, acquired through a minimally invasive biopsy, is possible and straightforward with a three-dimensional culture technique. These cells could be utilized in long-term expansion of lung progenitor cells and as part of the development of cell-based therapies for the treatment of lung diseases such as chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF). Electronic supplementary material The online version of this article (doi:10.1186/s12931-017-0611-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Phuong-Uyen C Dinh
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, 1060 William Moore Drive, RB306, Raleigh, NC 27607, NC, USA
| | - Jhon Cores
- Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh/Chapel Hill, NC, USA
| | - M Taylor Hensley
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, 1060 William Moore Drive, RB306, Raleigh, NC 27607, NC, USA
| | - Adam C Vandergriff
- Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh/Chapel Hill, NC, USA
| | - Junnan Tang
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, 1060 William Moore Drive, RB306, Raleigh, NC 27607, NC, USA.,Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Tyler A Allen
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, 1060 William Moore Drive, RB306, Raleigh, NC 27607, NC, USA
| | - Thomas G Caranasos
- Divisions of Cardiothoracic Surgery, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kenneth B Adler
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, 1060 William Moore Drive, RB306, Raleigh, NC 27607, NC, USA
| | - Leonard J Lobo
- Pulmonary Diseases and Critical Care Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, 1060 William Moore Drive, RB306, Raleigh, NC 27607, NC, USA. .,Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh/Chapel Hill, NC, USA.
| |
Collapse
|
10
|
Lung Regeneration: Endogenous and Exogenous Stem Cell Mediated Therapeutic Approaches. Int J Mol Sci 2016; 17:ijms17010128. [PMID: 26797607 PMCID: PMC4730369 DOI: 10.3390/ijms17010128] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 01/07/2016] [Accepted: 01/11/2016] [Indexed: 12/25/2022] Open
Abstract
The tissue turnover of unperturbed adult lung is remarkably slow. However, after injury or insult, a specialised group of facultative lung progenitors become activated to replenish damaged tissue through a reparative process called regeneration. Disruption in this process results in healing by fibrosis causing aberrant lung remodelling and organ dysfunction. Post-insult failure of regeneration leads to various incurable lung diseases including chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis. Therefore, identification of true endogenous lung progenitors/stem cells, and their regenerative pathway are crucial for next-generation therapeutic development. Recent studies provide exciting and novel insights into postnatal lung development and post-injury lung regeneration by native lung progenitors. Furthermore, exogenous application of bone marrow stem cells, embryonic stem cells and inducible pluripotent stem cells (iPSC) show evidences of their regenerative capacity in the repair of injured and diseased lungs. With the advent of modern tissue engineering techniques, whole lung regeneration in the lab using de-cellularised tissue scaffold and stem cells is now becoming reality. In this review, we will highlight the advancement of our understanding in lung regeneration and development of stem cell mediated therapeutic strategies in combating incurable lung diseases.
Collapse
|
11
|
Hogan BLM, Barkauskas CE, Chapman HA, Epstein JA, Jain R, Hsia CCW, Niklason L, Calle E, Le A, Randell SH, Rock J, Snitow M, Krummel M, Stripp BR, Vu T, White ES, Whitsett JA, Morrisey EE. Repair and regeneration of the respiratory system: complexity, plasticity, and mechanisms of lung stem cell function. Cell Stem Cell 2014; 15:123-38. [PMID: 25105578 PMCID: PMC4212493 DOI: 10.1016/j.stem.2014.07.012] [Citation(s) in RCA: 638] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Respiratory disease is the third leading cause of death in the industrialized world. Consequently, the trachea, lungs, and cardiopulmonary vasculature have been the focus of extensive investigations. Recent studies have provided new information about the mechanisms driving lung development and differentiation. However, there is still much to learn about the ability of the adult respiratory system to undergo repair and to replace cells lost in response to injury and disease. This Review highlights the multiple stem/progenitor populations in different regions of the adult lung, the plasticity of their behavior in injury models, and molecular pathways that support homeostasis and repair.
Collapse
Affiliation(s)
- Brigid L M Hogan
- Department of Cell Biology, Duke Medicine, Durham, NC 27705, USA.
| | - Christina E Barkauskas
- Division of Pulmonary, Allergy and Critical Care Medicine, Duke Medicine, Durham, NC 27705, USA
| | - Harold A Chapman
- Division of Pulmonary and Critical Care, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jonathan A Epstein
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rajan Jain
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Connie C W Hsia
- Department of Internal Medicine, University of Texas, Southwestern Medical Center, Dallas, TX 75390, USA
| | - Laura Niklason
- Departments of Anesthesiology and Biomedical Engineering, Yale University, New Haven, CT 06520, USA
| | - Elizabeth Calle
- Department of Cell Biology, Duke Medicine, Durham, NC 27705, USA
| | - Andrew Le
- Department of Cell Biology, Duke Medicine, Durham, NC 27705, USA
| | - Scott H Randell
- Department of Cell Biology and Physiology, The University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Jason Rock
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Melinda Snitow
- Perleman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matthew Krummel
- Division of Pulmonary and Critical Care, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Barry R Stripp
- Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Thiennu Vu
- Division of Pulmonary and Critical Care, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Eric S White
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jeffrey A Whitsett
- Section of Neonatology, Perinatal and Pulmonary Biology, Department of Pediatrics, Cincinnati Children's Hospital Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Edward E Morrisey
- Departments of Medicine and Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
12
|
Kotton DN, Morrisey EE. Lung regeneration: mechanisms, applications and emerging stem cell populations. Nat Med 2014; 20:822-32. [PMID: 25100528 PMCID: PMC4229034 DOI: 10.1038/nm.3642] [Citation(s) in RCA: 372] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 06/24/2014] [Indexed: 12/15/2022]
Abstract
Recent studies have shown that the respiratory system has an extensive ability to respond to injury and regenerate lost or damaged cells. The unperturbed adult lung is remarkably quiescent, but after insult or injury progenitor populations can be activated or remaining cells can re-enter the cell cycle. Techniques including cell-lineage tracing and transcriptome analysis have provided novel and exciting insights into how the lungs and trachea regenerate in response to injury and have allowed the identification of pathways important in lung development and regeneration. These studies are now informing approaches for modulating the pathways that may promote endogenous regeneration as well as the generation of exogenous lung cell lineages from pluripotent stem cells. The emerging advances, highlighted in this Review, are providing new techniques and assays for basic mechanistic studies as well as generating new model systems for human disease and strategies for cell replacement.
Collapse
Affiliation(s)
- Darrell N Kotton
- 1] Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, Massachusetts, USA. [2] Pulmonary Center, Boston University, Boston, Massachusetts, USA. [3] Department of Medicine, Boston University, Boston, Massachusetts, USA
| | - Edward E Morrisey
- 1] Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA. [2] Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA. [3] Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA. [4] Institute for Regenerative Medicine, University of Pennsylvania Philadelphia, Pennsylvania, USA
| |
Collapse
|
13
|
Volckaert T, De Langhe S. Lung epithelial stem cells and their niches: Fgf10 takes center stage. FIBROGENESIS & TISSUE REPAIR 2014; 7:8. [PMID: 24891877 PMCID: PMC4041638 DOI: 10.1186/1755-1536-7-8] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 04/04/2014] [Indexed: 12/20/2022]
Abstract
Throughout life adult animals crucially depend on stem cell populations to maintain and repair their tissues to ensure life-long organ function. Stem cells are characterized by their capacity to extensively self-renew and give rise to one or more differentiated cell types. These powerful stem cell properties are key to meet the changing demand for tissue replacement during normal lung homeostasis and regeneration after lung injury. Great strides have been made over the last few years to identify and characterize lung epithelial stem cells as well as their lineage relationships. Unfortunately, knowledge on what regulates the behavior and fate specification of lung epithelial stem cells is still limited, but involves communication with their microenvironment or niche, a local tissue environment that hosts and influences the behaviors or characteristics of stem cells and that comprises other cell types and extracellular matrix. As such, an intimate and dynamic epithelial-mesenchymal cross-talk, which is also essential during lung development, is required for normal homeostasis and to mount an appropriate regenerative response after lung injury. Fibroblast growth factor 10 (Fgf10) signaling in particular seems to be a well-conserved signaling pathway governing epithelial-mesenchymal interactions during lung development as well as between different adult lung epithelial stem cells and their niches. On the other hand, disruption of these reciprocal interactions leads to a dysfunctional epithelial stem cell-niche unit, which may culminate in chronic lung diseases such as chronic obstructive pulmonary disease (COPD), chronic asthma and idiopathic pulmonary fibrosis (IPF).
Collapse
Affiliation(s)
- Thomas Volckaert
- Department of Pediatrics, Division of Cell Biology, National Jewish Health, 1400 Jackson St, Denver, CO 80206, USA ; The Inflammation Research Center, Unit of Molecular Signal Transduction in Inflammation, VIB, Technologiepark 927, 9052 Ghent, Belgium ; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, 9052 Ghent, Belgium
| | - Stijn De Langhe
- Department of Pediatrics, Division of Cell Biology, National Jewish Health, 1400 Jackson St, Denver, CO 80206, USA ; Department of Cellular and Developmental Biology, School of Medicine, University of Colorado Denver, 12605 E 16th Avenue, Aurora CO 80045, USA
| |
Collapse
|
14
|
Zamora IJ, Sheikh F, Olutoye OO, Cassady CI, Lee TC, Ruano R, Cass DL. Mainstem bronchial atresia: a lethal anomaly amenable to fetal surgical treatment. J Pediatr Surg 2014; 49:706-11. [PMID: 24851752 DOI: 10.1016/j.jpedsurg.2014.02.051] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Accepted: 02/13/2014] [Indexed: 12/25/2022]
Abstract
PURPOSE The purpose of this study was to review the unique imaging characteristics, prenatal course, and outcomes for fetuses with mainstem bronchial atresia (MBA). METHODS The records of all patients referred for a fetal lung malformation from 2001 to 2012 and the medical literature were reviewed to identify cases of MBA. RESULTS Of 129 fetuses evaluated, 3 were diagnosed prenatally with right-sided MBA. The first had a CCAM-volume ratio (CVR) of 9, hydrops, mirror syndrome, and preterm delivery of a nonviable fetus. The second (CVR 2.6) had ascites, preterm delivery at 34-weeks, and neonatal demise. The third fetus (CVR 5.7) presented with hydrops at 21-weeks, prompting fetal pneumonectomy. Postoperatively, hydrops resolved, and the contralateral lung grew dramatically, but preterm delivery occurred 3 weeks later. Ventilation could not be sustained, and the infant died. Four similar cases of MBA were in the literature, all right-sided. Two fetuses with hydrops delivered at 25-weeks and died immediately. One pregnancy was terminated. One fetus underwent pneumonectomy at 24-weeks but died intraoperatively. CONCLUSION MBA is a rare and lethal lesion that must be distinguished from other right-sided lung masses. Fetal pneumonectomy can be performed with resolution of hydrops and compensatory contralateral lung growth, but remains limited by complications of preterm birth.
Collapse
Affiliation(s)
- Irving J Zamora
- Texas Children's Fetal Center, Texas Children's Hospital, Houston, TX; Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX
| | - Fariha Sheikh
- Texas Children's Fetal Center, Texas Children's Hospital, Houston, TX; Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX
| | - Oluyinka O Olutoye
- Texas Children's Fetal Center, Texas Children's Hospital, Houston, TX; Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX; Department Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX
| | - Christopher I Cassady
- Texas Children's Fetal Center, Texas Children's Hospital, Houston, TX; Department of Radiology, Baylor College of Medicine, Houston, TX
| | - Timothy C Lee
- Texas Children's Fetal Center, Texas Children's Hospital, Houston, TX; Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX
| | - Rodrigo Ruano
- Texas Children's Fetal Center, Texas Children's Hospital, Houston, TX; Department Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX
| | - Darrell L Cass
- Texas Children's Fetal Center, Texas Children's Hospital, Houston, TX; Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX; Department Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX.
| |
Collapse
|
15
|
Thane K, Ingenito EP, Hoffman AM. Lung regeneration and translational implications of the postpneumonectomy model. Transl Res 2014; 163:363-76. [PMID: 24316173 DOI: 10.1016/j.trsl.2013.11.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 10/30/2013] [Accepted: 11/18/2013] [Indexed: 10/26/2022]
Abstract
Lung regeneration research is yielding data with increasing translational value. The classical models of lung development, postnatal alveolarization, and postpneumonectomy alveolarization have contributed to a broader understanding of the cellular participants including stem-progenitor cells, cell-cell signaling pathways, and the roles of mechanical deformation and other physiologic factors that have the potential to be modulated in human and animal patients. Although recent information is available describing the lineage fate of lung fibroblasts, genetic fate mapping, and clonal studies are lacking in the study of lung regeneration and deserve further examination. In addition to increasing knowledge concerning classical alveolarization (postnatal, postpneumonectomy), there is increasing evidence for remodeling of the adult lung after partial pneumonectomy. Though limited in scope, compelling data have emerged describing restoration of lung tissue mass in the adult human and in large animal models. The basis for this long-term adaptation to pneumonectomy is poorly understood, but investigations into mechanisms of lung regeneration in older animals that have lost their capacity for rapid re-alveolarization are warranted, as there would be great translational value in modulating these mechanisms. In addition, quantitative morphometric analysis has progressed in conjunction with developments in advanced imaging, which allow for longitudinal and nonterminal evaluation of pulmonary regenerative responses in animals and humans. This review focuses on the cellular and molecular events that have been observed in animals and humans after pneumonectomy because this model is closest to classical regeneration in other mammalian systems and has revealed several new fronts of translational research that deserve consideration.
Collapse
Affiliation(s)
- Kristen Thane
- Department of Clinical Sciences, Regenerative Medicine Laboratory, Tufts University Cummings School of Veterinary Medicine, North Grafton, Mass
| | - Edward P Ingenito
- Division of Pulmonary, Critical Care, and Sleep Medicine, Brigham and Women's Hospital, Boston, Mass
| | - Andrew M Hoffman
- Department of Clinical Sciences, Regenerative Medicine Laboratory, Tufts University Cummings School of Veterinary Medicine, North Grafton, Mass.
| |
Collapse
|
16
|
Herriges M, Morrisey EE. Lung development: orchestrating the generation and regeneration of a complex organ. Development 2014; 141:502-13. [PMID: 24449833 DOI: 10.1242/dev.098186] [Citation(s) in RCA: 399] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The respiratory system, which consists of the lungs, trachea and associated vasculature, is essential for terrestrial life. In recent years, extensive progress has been made in defining the temporal progression of lung development, and this has led to exciting discoveries, including the derivation of lung epithelium from pluripotent stem cells and the discovery of developmental pathways that are targets for new therapeutics. These discoveries have also provided new insights into the regenerative capacity of the respiratory system. This Review highlights recent advances in our understanding of lung development and regeneration, which will hopefully lead to better insights into both congenital and acquired lung diseases.
Collapse
Affiliation(s)
- Michael Herriges
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | |
Collapse
|
17
|
Hematopoietic and mesenchymal stem cells for the treatment of chronic respiratory diseases: role of plasticity and heterogeneity. ScientificWorldJournal 2014; 2014:859817. [PMID: 24563632 PMCID: PMC3916026 DOI: 10.1155/2014/859817] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 10/20/2013] [Indexed: 12/21/2022] Open
Abstract
Chronic lung diseases, such as cystic fibrosis (CF), asthma, and chronic obstructive pulmonary disease (COPD) are incurable and represent a very high social burden. Stem cell-based treatment may represent a hope for the cure of these diseases. In this paper, we revise the overall knowledge about the plasticity and engraftment of exogenous marrow-derived stem cells into the lung, as well as their usefulness in lung repair and therapy of chronic lung diseases. The lung is easily accessible and the pathophysiology of these diseases is characterized by injury, inflammation, and eventually by remodeling of the airways. Bone marrow-derived stem cells, including hematopoietic stem/progenitor cells (HSPCs) and mesenchymal stromal (stem) cells (MSCs), encompass a wide array of cell subsets with different capacities of engraftment and injured tissue regenerating potential. Proof-of-principle that marrow cells administered locally may engraft and give rise to specialized epithelial cells has been given, but the efficiency of this conversion is too limited to give a therapeutic effect. Besides the identification of plasticity mechanisms, the characterization/isolation of the stem cell subpopulations represents a major challenge to improving the efficacy of transplantation protocols used in regenerative medicine for lung diseases.
Collapse
|