1
|
Radiofrequency therapy improves exercise capacity of mice with emphysema. Sci Rep 2021; 11:20056. [PMID: 34625605 PMCID: PMC8501094 DOI: 10.1038/s41598-021-99474-8] [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: 03/12/2021] [Accepted: 09/14/2021] [Indexed: 11/08/2022] Open
Abstract
Emphysema is a common phenotype of chronic obstructive pulmonary disease (COPD). Although resection of emphysematous tissue can improve lung mechanics, it is invasive and fraught with adverse effects. Meanwhile, radiofrequency (RF) treatment is an extracorporeal method that leads to tissue destruction and remodeling, resulting in “volume reduction” and overall improvement in lung compliance of emphysematous lungs. Whether these changes lead to improved exercise tolerance is unknown. Here, we investigated the effectiveness of RF treatment to improve the exercise capacity of mice with emphysema. Fifty-two mice (7 weeks of age) were used in this experiment. A bilateral emphysema model was created by intratracheally instilling porcine pancreatic elastase (PPE) (1.5U/100 g body weight). RF treatment (0.5 W/ g body weight) was administered extracorporeally 14 days later and mice were sacrificed after another 21 days. The exercise capacity of mice was measured using a treadmill. Treadmill runs were performed just before PPE instillation (baseline), before RF treatment and before sacrifice. Following sacrifice, lung compliance and mean linear intercept (Lm) were measured and fibrosis was assessed using a modified Ashcroft score. There were 3 experimental groups: controls (instilled with saline, n = 12), emphysema (instilled with porcine pancreatic elastase, PPE, n = 11) and emphysema + treatment (instilled with PPE and given RF, n = 9). At endpoint, the maximum velocity of the emphysema + treatment group was significantly higher than that of the emphysema group, indicating improved exercise tolerance (86.29% of baseline vs 61.69% of baseline, p = 0.01). Histological analysis revealed a significant reduction in emphysema as denoted by Lm between the two groups (median 29.60 µm vs 35.68 µm, p = 0.03). The emphysema + treatment group also demonstrated a higher prevalence of lung fibrosis (≧Grade 3) compared with the emphysema group (11.7% vs 5.4%, p < 0.01). No severe adverse events from RF were observed. RF treatment improved the exercise capacity of mice with emphysema. These data highlight the therapeutic potential of RF treatment in improving the functional status of patients with COPD.
Collapse
|
2
|
Gülhan PY, Ekici MS, Niyaz M, Gülhan M, Erçin ME, Ekici A, Aksoy N. Therapeutic Treatment with Abdominal Adipose Mesenchymal Cells Does Not Prevent Elastase-Induced Emphysema in Rats. Turk Thorac J 2020; 21:14-20. [PMID: 32163359 PMCID: PMC7020897 DOI: 10.5152/turkthoracj.2019.180136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 02/06/2019] [Indexed: 11/22/2022]
Abstract
OBJECTIVES Emphysema and chronic bronchitis have different pathophysiologies but both are significant components of chronic obstructive lung disease (COPD). The levels of Matrix metalloproteinase (MMP)-9 in the bronchoalveloar lavage fluid (BALF) and in serum indicate the presence of emphysema. Intratracheal administration of elastase has been used to create a rat model of emphysema. Adipose tissue-derived mesenchymal stem cells (MSC) have been postulated to prevent or reverse emphysema, however, this has not been examined in the rat model of elastase-induced emphysema. MATERIALS AND METHODS In this study, 31 Wistar albino rats aged 6-8 weeks and weighing 250-300 g were assessed. On day 1, the animals were treated intratracheally with 0.5 mL saline (control group, n=10), i.e., 0.5 mL saline solution containing 0.1 IU porcine pancreatic elastase (PPE) (Elastase group, n=12) or PPE plus MSC (Elastase-MSC group, n=9) was adminstered per animal. MSCs suspended in serum were injected via the caudal vein on day 21. At least 106 cells were injected. All animals were sacrificed on day 42 and the emphysema index (EI) was calculated, along with measuring the BALF and serum MMP-9 concentrations. RESULTS Porcine pancreatic elastase induced a significant degree of emphysema in the PPE groups as compared to the control group, which was determined by the EI index (p=0.008). This was not reversed by MSC treatment. The EI remained significantly low in comprison with the controls (p=0.001) and measured no different from the Elastase-treated animals. There was no statistically significant difference between the BALF and serum MMP-9 levels between the control and treatment groups. CONCLUSION Our findings suggest that therapeutic treatment with adipose tissue-derived MSC in rats has no effect on emphysema or on MMP9 expression, which is a known marker of emphysema.
Collapse
Affiliation(s)
- Pınar Yıldız Gülhan
- Department of Chest Diseases, Düzce University School of Medicine, Düzce, Turkey
| | - Mehmet Savaş Ekici
- Department of Chest Diseases, Kırıkkale University School of Medicine, Kırıkkale, Turkey
| | - Mehmet Niyaz
- Clinic of Cardiovascular Surgery, Bartın State Hospital, Bartın, Turkey
| | - Muhammet Gülhan
- Clinic of Infectious Diseases and Clinical Microbiology, Tosya State Hospital, Kastamonu, Turkey
| | - Mustafa Emre Erçin
- Department of Pathology, Karadeniz Technical University School of Medicine, Trabzon, Turkey
| | - Aydanur Ekici
- Department of Chest Diseases, Kırıkkale University School of Medicine, Kırıkkale, Turkey
| | - Nurkan Aksoy
- Clinic of Biochemistry, Yenimahalle State Hospital, Ankara, Turkey
| |
Collapse
|
3
|
Oki H, Yazawa T, Baba Y, Kanegae Y, Sato H, Sakamoto S, Goto T, Saito I, Kurahashi K. Adenovirus vector expressing keratinocyte growth factor using CAG promoter impairs pulmonary function of mice with elastase-induced emphysema. Microbiol Immunol 2017; 61:264-271. [PMID: 28543309 DOI: 10.1111/1348-0421.12492] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 05/15/2017] [Accepted: 05/21/2017] [Indexed: 11/29/2022]
Abstract
Pulmonary emphysema impairs quality of life and increases mortality. It has previously been shown that administration of adenovirus vector expressing murine keratinocyte growth factor (KGF) before elastase instillation prevents pulmonary emphysema in mice. We therefore hypothesized that therapeutic administration of KGF would restore damage to lungs caused by elastase instillation and thus improve pulmonary function in an animal model. KGF expressing adenovirus vector, which prevented bleomycin-induced pulmonary fibrosis in a previous study, was constructed. Adenovirus vector (1.0 × 109 plaque-forming units) was administered intratracheally one week after administration of elastase into mouse lungs. One week after administration of KGF-vector, exercise tolerance testing and blood gas analysis were performed, after which the lungs were removed under deep anesthesia. KGF-positive pneumocytes were more numerous, surfactant protein secretion in the airspace greater and mean linear intercept of lungs shorter in animals that had received KGF than in control animals. Unexpectedly, however, arterial blood oxygenation was worse in the KGF group and maximum running speed, an indicator of exercise capacity, had not improved after KGF in mice with elastase-induced emphysema, indicating that KGF-expressing adenovirus vector impaired pulmonary function in these mice. Notably, vector lacking KGF-expression unit did not induce such impairment, implying that the KGF expression unit itself may cause the damage to alveolar cells. Possible involvement of the CAG promoter used for KGF expression in impairing pulmonary function is discussed.
Collapse
Affiliation(s)
- Hiroshi Oki
- Department of Anesthesiology and Critical Care Medicine, Yokohama City University Graduate School of Medicine, Yokohama
| | - Takuya Yazawa
- Department of Pathology, Yokohama City University Graduate School of Medicine, Yokohama
| | - Yasuko Baba
- Department of Anesthesiology and Critical Care Medicine, Yokohama City University Graduate School of Medicine, Yokohama
| | - Yumi Kanegae
- Laboratory of Molecular Genetics, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Hanako Sato
- Department of Pathology, Yokohama City University Graduate School of Medicine, Yokohama
| | - Seiko Sakamoto
- Department of Anesthesiology and Critical Care Medicine, Yokohama City University Graduate School of Medicine, Yokohama
| | - Takahisa Goto
- Department of Anesthesiology and Critical Care Medicine, Yokohama City University Graduate School of Medicine, Yokohama
| | - Izumu Saito
- Laboratory of Molecular Genetics, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Kiyoyasu Kurahashi
- Department of Anesthesiology and Critical Care Medicine, Yokohama City University Graduate School of Medicine, Yokohama
| |
Collapse
|
4
|
Pourteymour S, Eckardt K, Holen T, Langleite T, Lee S, Jensen J, Birkeland KI, Drevon CA, Hjorth M. Global mRNA sequencing of human skeletal muscle: Search for novel exercise-regulated myokines. Mol Metab 2017; 6:352-365. [PMID: 28377874 PMCID: PMC5369209 DOI: 10.1016/j.molmet.2017.01.007] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 01/16/2017] [Accepted: 01/19/2017] [Indexed: 12/19/2022] Open
Abstract
Objective Skeletal muscle is an important secretory organ, producing and releasing numerous myokines, which may be involved in mediating beneficial health effects of physical activity. More than 100 myokines have been identified by different proteomics approaches, but these techniques may not detect all myokines. We used mRNA sequencing as an untargeted approach to study gene expression of secreted proteins in skeletal muscle upon acute as well as long-term exercise. Methods Twenty-six middle-aged, sedentary men underwent combined endurance and strength training for 12 weeks. Skeletal muscle biopsies from m. vastus lateralis and blood samples were taken before and after an acute bicycle test, performed at baseline as well as after 12 weeks of training intervention. We identified transcripts encoding secretory proteins that were changed more than 1.5-fold in muscle after exercise. Secretory proteins were defined based on either curated UniProt annotations or predictions made by multiple bioinformatics methods. Results This approach led to the identification of 161 candidate secretory transcripts that were up-regulated after acute exercise and 99 that where increased after 12 weeks exercise training. Furthermore, 92 secretory transcripts were decreased after acute and/or long-term physical activity. From these responsive transcripts, we selected 17 candidate myokines sensitive to short- and/or long-term exercise that have not been described as myokines before. The expression of these transcripts was confirmed in primary human skeletal muscle cells during in vitro differentiation and electrical pulse stimulation (EPS). One of the candidates we identified was macrophage colony-stimulating factor-1 (CSF1), which influences macrophage homeostasis. CSF1 mRNA increased in skeletal muscle after acute and long-term exercise, which was accompanied by a rise in circulating CSF1 protein. In cultured muscle cells, EPS promoted a significant increase in the expression and secretion of CSF1. Conclusion We identified 17 new, exercise-responsive transcripts encoding secretory proteins. We further identified CSF1 as a novel myokine, which is secreted from cultured muscle cells and up-regulated in muscle and plasma after acute exercise. Numerous transcripts were identified that were regulated in human skeletal muscle after acute and/or long-term exercise. These transcripts encode potential myokines, which may play key roles in local and systemic adaptations to exercise. CSF1 was identified as a novel myokine. CSF1 was increased after acute exercise, and secreted from cultured human myotubes in response to EPS.
Collapse
Affiliation(s)
- S Pourteymour
- Department of Nutrition, Institute for Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - K Eckardt
- Department of Nutrition, Institute for Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - T Holen
- Department of Nutrition, Institute for Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - T Langleite
- Department of Nutrition, Institute for Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Sindre Lee
- Department of Nutrition, Institute for Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - J Jensen
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| | - K I Birkeland
- Department of Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital and Faculty of Medicine, University of Oslo, Oslo, Norway
| | - C A Drevon
- Department of Nutrition, Institute for Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - M Hjorth
- Department of Nutrition, Institute for Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway; Diabetes and Metabolism Division, Garvan Institute of Medical Research, Darlinghurst, NSW, 2010, Australia.
| |
Collapse
|
5
|
Vaz M, Rajasekaran S, Potteti HR, Reddy SP. Myeloid-specific Fos-related antigen-1 regulates cigarette smoke-induced lung inflammation, not emphysema, in mice. Am J Respir Cell Mol Biol 2015; 53:125-34. [PMID: 25489966 DOI: 10.1165/rcmb.2014-0118oc] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Heightened lung inflammation is a cardinal feature of chronic obstructive pulmonary disease (COPD). Cigarette smoke (CS)-induced macrophage recruitment and activation, accompanied by abnormal secretion of a number of inflammatory cytokines and matrix metalloproteinases, play a major role in the pathophysiology of COPD. The Fos-related antigen-1 (Fra-1) transcription factor differentially regulates several cellular processes that are implicated in COPD, such as inflammation and immune responses, cell proliferation and death, and extracellular remodeling. Although CS stimulates Fra-1 expression in the lung, the precise role of this transcription factor in the regulation of CS-induced lung inflammation in vivo is poorly understood. Here, we report that myeloid-specific Fra-1 signaling is important for CS-induced lung macrophagic inflammatory response. In response to chronic CS exposure, mice with Fra-1 specifically deleted in myeloid cells showed reduced levels of CS-induced lung macrophagic inflammation, accompanied by decreased expression levels of proinflammatory cytokines compared with their wild-type counterparts. Consistent with this result, bone marrow-derived Fra-1-null macrophages treated with CS showed decreased levels of proinflammatory mediators and matrix metalloproteinases. Interestingly, deletion of Fra-1 in myeloid cells did not affect the severity of emphysema. We propose that Fra-1 plays a key role in promoting chronic CS-induced lung macrophagic inflammation in vivo, and that targeting this transcription factor may be useful in dampening persistent lung inflammation in patients with COPD.
Collapse
Affiliation(s)
- Michelle Vaz
- 1 Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University, Baltimore, Maryland; and
| | - Subbiah Rajasekaran
- 2 Division of Developmental Biology and Basic Research, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois
| | - Haranatha R Potteti
- 2 Division of Developmental Biology and Basic Research, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois
| | - Sekhar P Reddy
- 2 Division of Developmental Biology and Basic Research, Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
6
|
Aida Y, Shibata Y, Abe S, Inoue S, Kimura T, Igarashi A, Yamauchi K, Nunomiya K, Kishi H, Nemoto T, Sato M, Sato-Nishiwaki M, Nakano H, Sato K, Kubota I. Inhibition of elastase-pulmonary emphysema in dominant-negative MafB transgenic mice. Int J Biol Sci 2014; 10:882-94. [PMID: 25170302 PMCID: PMC4147222 DOI: 10.7150/ijbs.8737] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Accepted: 07/30/2014] [Indexed: 11/06/2022] Open
Abstract
Alveolar macrophages (AMs) play important roles in the pathogenesis of chronic obstructive pulmonary disease (COPD). We previously demonstrated upregulation of the transcription factor MafB in AMs of mice exposed to cigarette smoke. The aim of this study was to elucidate the roles of MafB in the development of pulmonary emphysema. Porcine pancreatic elastase was administered to wild-type (WT) and dominant-negative (DN)-MafB transgenic (Tg) mice in which MafB activity was suppressed only in macrophages. We measured the mean linear intercept and conducted cell differential analysis of bronchoalveolar lavage (BAL) cells, surface marker analysis using flow cytometry, and immunohistochemical staining using antibodies to matrix metalloproteinase (MMP)-9 and MMP-12. Airspace enlargement of the lungs was suppressed significantly in elastase-treated DN-MafB Tg mice compared with treated WT mice. AMs with projected pseudopods were decreased in DN-MafB Tg mice. The number of cells intermediately positive for F4/80 and weakly or intermediately positive for CD11b, which are considered cell subsets of matured AMs, decreased in the BAL of DN-MafB Tg mice. Furthermore, MMP-9 and -12 were significantly downregulated in BAL cells of DN-MafB Tg mice. Because MMPs exacerbate emphysema, MafB may be involved in pulmonary emphysema development through altered maturation of macrophages and MMP expression.
Collapse
Affiliation(s)
- Yasuko Aida
- Department of Cardiology, Pulmonology, and Nephrology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Yoko Shibata
- Department of Cardiology, Pulmonology, and Nephrology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Shuichi Abe
- Department of Cardiology, Pulmonology, and Nephrology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Sumito Inoue
- Department of Cardiology, Pulmonology, and Nephrology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Tomomi Kimura
- Department of Cardiology, Pulmonology, and Nephrology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Akira Igarashi
- Department of Cardiology, Pulmonology, and Nephrology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Keiko Yamauchi
- Department of Cardiology, Pulmonology, and Nephrology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Keiko Nunomiya
- Department of Cardiology, Pulmonology, and Nephrology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Hiroyuki Kishi
- Department of Cardiology, Pulmonology, and Nephrology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Takako Nemoto
- Department of Cardiology, Pulmonology, and Nephrology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Masamichi Sato
- Department of Cardiology, Pulmonology, and Nephrology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Michiko Sato-Nishiwaki
- Department of Cardiology, Pulmonology, and Nephrology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Hiroshi Nakano
- Department of Cardiology, Pulmonology, and Nephrology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Kento Sato
- Department of Cardiology, Pulmonology, and Nephrology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| | - Isao Kubota
- Department of Cardiology, Pulmonology, and Nephrology, School of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata 990-9585, Japan
| |
Collapse
|
7
|
Arimura K, Aoshiba K, Tsuji T, Tamaoki J. Chronic low-grade systemic inflammation causes DNA damage in the lungs of mice. Lung 2012; 190:613-20. [PMID: 22965854 DOI: 10.1007/s00408-012-9414-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Accepted: 08/26/2012] [Indexed: 12/19/2022]
Abstract
BACKGROUND Whether systemic inflammation compromises the pulmonary system is largely unknown. We tested the hypothesis that chronic low-grade systemic inflammation damages alveolar wall cells. METHODS A chronic low-grade systemic inflammatory state was induced in 8-week-old male C57/BL6J mice by administration of lipopolysaccharide (LPS, 44.4 μg/day) for a 90-day period by subcutaneous implantation of a delayed-release pellet system. Acute systemic inflammation was induced in another group of mice by a single intraperitoneal injection of LPS (125 μg/body). The lungs of mice were examined for histologic changes and genetic damage to alveolar wall cells. RESULTS Chronic LPS exposure for a 30-day period or a 90-day period did not cause any obvious architectural changes in the lungs except for a mild level of alveolar macrophage infiltration. Despite the lack of architectural changes in the lung, immunofluorescence staining for γH2AX and phosphorylated 53BP1 showed that chronic LPS exposure resulted in an almost doubling of the number of DNA double-strand breaks (DSBs) in type 1 and type 2 alveolar epithelial cells and in alveolar endothelial cells. Acute LPS exposure also resulted in a doubling of the number of DSBs in type 1 and type 2 alveolar epithelial cells and in alveolar endothelial cells at 24 h, but the increased number of DSBs returned to the baseline level by 48 h. CONCLUSIONS These results suggest that chronic systemic low-grade inflammation induces persistent DNA damage in alveolar epithelial and endothelial cells before architectural changes in the lung become evident.
Collapse
Affiliation(s)
- Ken Arimura
- Pulmonary Division, Graduate School of Medical Science, Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | | | | | | |
Collapse
|
8
|
Roulet A, Armand L, Dagouassat M, Rogerieux F, Simon-Deckers A, Belade E, Van Nhieu JT, Lanone S, Pairon JC, Lacroix G, Boczkowski J. Intratracheally administered titanium dioxide or carbon black nanoparticles do not aggravate elastase-induced pulmonary emphysema in rats. BMC Pulm Med 2012; 12:38. [PMID: 22849372 PMCID: PMC3499434 DOI: 10.1186/1471-2466-12-38] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Accepted: 07/26/2012] [Indexed: 11/22/2022] Open
Abstract
Background Titanium dioxide (TiO2) and carbon black (CB) nanoparticles (NPs) have biological effects that could aggravate pulmonary emphysema. The aim of this study was to evaluate whether pulmonary administration of TiO2 or CB NPs in rats could induce and/or aggravate elastase-induced emphysema, and to investigate the underlying molecular mechanisms. Methods On day 1, Sprague-Dawley rats were intratracheally instilled with 25 U kg−1 pancreatic porcine elastase or saline. On day 7, they received an intratracheal instillation of TiO2 or CB (at 100 and 500 μg) dispersed in bovine serum albumin or bovine serum albumin alone. Animals were sacrificed at days 8 or 21, and bronchoalveolar lavage (BAL) cellularity, histological analysis of inflammation and emphysema, and lung mRNA expression of heme oxygenase-1 (HO-1), interleukin-1β (IL-1β), macrophage inflammatory protein-2, monocyte chemotactic protein-1, and matrix metalloprotease (MMP)-1, and -12 were measured. In addition, pulmonary MMP-12 expression was also analyzed at the protein level by immunohistochemistry. Results TiO2 NPs per se did not modify the parameters investigated, but CB NPs increased perivascular/peribronchial infiltration, and macrophage MMP-12 expression, without inducing emphysema. Elastase administration increased BAL cellularity, histological inflammation, HO-1, IL-1β and macrophage MMP-12 expression and induced emphysema. Exposure to TiO2 NPs did not modify pulmonary responses to elastase, but exposure to CB NPs aggravated elastase-induced histological inflammation without aggravating emphysema. Conclusions TiO2 and CB NPs did not aggravate elastase-induced emphysema. However, CB NPs induced histological inflammation and MMP-12 mRNA and protein expression in macrophages.
Collapse
|
9
|
Lee JH, Hanaoka M, Kitaguchi Y, Kraskauskas D, Shapiro L, Voelkel NF, Taraseviciene-Stewart L. Imbalance of apoptosis and cell proliferation contributes to the development and persistence of emphysema. Lung 2012; 190:69-82. [PMID: 22015802 DOI: 10.1007/s00408-011-9326-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Accepted: 09/12/2011] [Indexed: 01/13/2023]
Abstract
BACKGROUND We postulate that in adults there is an established lung structure maintenance program and that lung alveolar septal cells are undergoing both continuous apoptosis and proliferation. Whereas lung cell apoptosis has been recognized in human emphysema, little is known about cell proliferation. METHODS Using a novel rat model of emphysema, induced by intratracheal instillation of cigarette smoke extract (CSE), we investigated the dynamics of emphysematous lung destruction. Emphysematous lung destruction was determined by measuring mean linear intercept and destructive index. Lung injury and repair were assessed by immunohistochemistry and Western blot analysis for active caspase-3 and proliferating cell nuclear antigen (PCNA) after 4, 8, and 12 weeks of CSE instillations. RESULTS The emphysematous lung tissue destruction was present at 4 weeks of CSE treatment and progressed to 8 weeks. Spontaneous repair began at 12 weeks. Treatment with a peroxisome proliferator activated receptor (PPAR)α+γ agonist or granulocyte and macrophage-colony stimulating factor (GM-CSF) for 4 weeks prevented the progression of emphysematous lung destruction and decreased the number of caspase-3-positive cells. CONCLUSION Apoptosis and cell proliferation occur in this new model of emphysema. Treatment with a PPARα+γ agonist or GM-CSF can inhibit the progression of emphysematous alveolar septal destruction by decreasing alveolar cell apoptosis.
Collapse
Affiliation(s)
- Ji-Hyun Lee
- Division of Pulmonary Science and Critical Care Medicine, University of Colorado School of Medicine, Denver, CO 80262, USA.
| | | | | | | | | | | | | |
Collapse
|
10
|
Tasaka S, Inoue KI, Miyamoto K, Nakano Y, Kamata H, Shinoda H, Hasegawa N, Miyasho T, Satoh M, Takano H, Ishizaka A. Role of interleukin-6 in elastase-induced lung inflammatory changes in mice. Exp Lung Res 2010; 36:362-72. [PMID: 20653471 DOI: 10.3109/01902141003678590] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Interleukin-6 (IL-6) is known to be involved in the pathogenesis of various inflammatory diseases, but its role in the development of pulmonary emphysema remains unclear. Wild-type (WT) and IL-6-deficient mice received either phosphate-buffered saline (PBS) or porcine pancreatic elastase (PPE) intratracheally. The development of emphysema was determined by measuring the mean linear intercept (Lm). The lung specimens were also subjected to immunohistochemistry for single-stranded DNA to detect apoptotic cells. Lung mechanics and airway responsiveness to inhaled methacholine were analyzed. Bronchoalveolar lavage (BAL) fluid was subjected to evaluation of inflammatory cell accumulation and cytokine measurement. PPE treatment caused significant increases in Lm and lung compliance, which was attenuated by IL-6 deficiency. The increases in apoptotic cells in the lung were attenuated in IL-6 null mice. Airway responsiveness was not affected by PPE challenge or IL-6 deficiency. Intratracheal PPE increased the cell counts in BAL fluid throughout the observation, which was suppressed in IL-6 null mice. In BAL fluid, PPE-induced increases in the levels of macrophage inflammatory protein (MIP)-1alpha and eotaxin were mitigated by IL-6 deficiency. PPE-induced up-regulation of matrix metalloproteinase (MMP)-12 in the lung was attenuated by IL-6 deficiency. These results indicate that IL-6 may play an important role in the development of elastase-induced lung inflammatory changes.
Collapse
Affiliation(s)
- Sadatomo Tasaka
- Division of Pulmonary Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Lüthje L, Raupach T, Michels H, Unsöld B, Hasenfuss G, Kögler H, Andreas S. Exercise intolerance and systemic manifestations of pulmonary emphysema in a mouse model. Respir Res 2009; 10:7. [PMID: 19175913 PMCID: PMC2644670 DOI: 10.1186/1465-9921-10-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2008] [Accepted: 01/28/2009] [Indexed: 08/30/2023] Open
Abstract
Background Systemic effects of chronic obstructive pulmonary disease (COPD) significantly contribute to severity and mortality of the disease. We aimed to develop a COPD/emphysema model exhibiting systemic manifestations of the disease. Methods Female NMRI mice were treated 5 times intratracheally with porcine pancreatic elastase (emphysema) or phosphate-buffered saline (control). Emphysema severity was quantified histologically by mean linear intercept, exercise tolerance by treadmill running distance, diaphragm dysfunction using isolated muscle strips, pulmonary hypertension by measuring right ventricular pressure, and neurohumoral activation by determining urinary norepinephrine concentration. Results Mean linear intercept was higher in emphysema (260.7 ± 26.8 μm) than in control lungs (24.7 ± 1.7 μm). Emphysema mice lost body weight, controls gained weight. Running distance was shorter in emphysema than in controls. Diaphragm muscle length was shorter in controls compared to emphysema. Fatigue tests of muscle strips revealed impaired relaxation in emphysema diaphragms. Maximum right ventricular pressure and norepinephrine were elevated in emphysema compared to controls. Linear correlations were observed between running distance changes and intercept, right ventricular weight, norepinephrine, and diaphragm length. Conclusion The elastase mouse model exhibited severe emphysema with consecutive exercise limitation, and neurohumoral activation. The model may deepen our understanding of systemic aspects of COPD.
Collapse
Affiliation(s)
- Lars Lüthje
- Kardiologie und Pneumologie, Georg-August-Universität, Göttingen, Germany.
| | | | | | | | | | | | | |
Collapse
|
12
|
Abstract
Although they were originally defined as haematopoietic-cell growth factors, colony-stimulating factors (CSFs) have been shown to have additional functions by acting directly on mature myeloid cells. Recent data from animal models indicate that the depletion of CSFs has therapeutic benefit in many inflammatory and/or autoimmune conditions and as a result, early-phase clinical trials targeting granulocyte/macrophage colony-stimulating factor and macrophage colony-stimulating factor have now commenced. The distinct biological features of CSFs offer opportunities for specific targeting, but with some associated risks. Here, I describe these biological features, discuss the probable specific outcomes of targeting CSFs in vivo and highlight outstanding questions that need to be addressed.
Collapse
|
13
|
Intranasal HGF administration ameliorates the physiologic and morphologic changes in lung emphysema. Mol Ther 2008; 16:1417-26. [PMID: 18560414 DOI: 10.1038/mt.2008.137] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Hepatocyte growth factor (HGF) has multiple biological effects on stem cells, epithelial proliferation, and wound healing. In this study, we investigated a possible therapeutic benefit of intranasal HGF on elastase-induced emphysema, and assessed the role of stem/progenitor cells in this process. HGF was given twice a week for 1-4 weeks after the establishment of emphysema in mice. HGF inhalation significantly ameliorated the enlargement of airspaces and alveolar wall destruction. Also, elevated static lung compliance returned to control levels within 2 weeks of HGF treatment. The expressions of stem-cell markers, c-kit, stem-cell antigen 1 (Sca-1), and CD34 were also significantly influenced by HGF. Most of the c-kit(+) cells were bone marrow derived, while most Sca-1(+) were lung endogenous cells. CD34(+) cells were from both sources, and a portion of the endogenous CD34(+) cells was also Sca-1(+). Further, HGF increased the expression levels of proliferating cell nuclear antigen (PCNA) and cytokeratin-19. Also, their immunohistochemical staining patterns were colocalized, indicative of epithelial multiplication. The results of the study show that intranasal treatment with HGF reverses both the physiological and morphometric changes of lung emphysema, possibly through stem-cell mobilization and alveolar regeneration, providing a nonsurgical treatment and suggesting the possibility of achieving a similar effect in humans.
Collapse
|
14
|
Wright JL, Cosio M, Churg A. Animal models of chronic obstructive pulmonary disease. Am J Physiol Lung Cell Mol Physiol 2008; 295:L1-15. [PMID: 18456796 DOI: 10.1152/ajplung.90200.2008] [Citation(s) in RCA: 300] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The mechanisms involved in the genesis of chronic obstructive pulmonary disease (COPD) are poorly defined. This area is complicated and difficult to model because COPD consists of four separate anatomic lesions (emphysema, small airway remodeling, pulmonary hypertension, and chronic bronchitis) and a functional lesion, acute exacerbation; moreover, the disease in humans develops over decades. This review discusses the various animal models that have been used to attempt to recreate human COPD and the advantages and disadvantages of each. None of the models reproduces the exact changes seen in humans, but cigarette smoke-induced disease appears to come the closest, and genetically modified animals also, in some instances, shed light on processes that appear to play a role.
Collapse
Affiliation(s)
- Joanne L Wright
- Department of Pathology, University of British Columbia, Vancouver, British Columbia, Canada.
| | | | | |
Collapse
|