1
|
Kettawan A, Ruangklai S, Rungruang T, Thongam J, Kettawan AK, Nirmal N, Srisuma S. Rice Bran Oil Improves Emphysema in Cigarette Smoke Extract-Induced Mice through Anti-Inflammatory and Antioxidative Effects. Nutrients 2024; 16:433. [PMID: 38337717 PMCID: PMC10857134 DOI: 10.3390/nu16030433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/10/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
Lung inflammation and alveolar enlargement are the major pathological conditions of chronic obstructive pulmonary disease (COPD) patients. Rice bran oil (RBO), a natural anti-inflammatory and antioxidative agent, has been used for therapeutic purposes in several inflammatory diseases. This study aimed to investigate the anti-inflammatory and antioxidative effect of RBO on a cigarette smoke extract (CSE)-induced emphysema model in mice. The results indicated that CSE significantly induced airspace enlargement in mouse lung. Increased inflammatory cells, macrophage, and TNF-alpha levels in bronchoalveolar lavage fluid (BALF) were noticed in CSE-treated mice. RBO (low and high dose)-supplemented mice showed decreased total BALF inflammatory cell, macrophage, and neutrophil numbers and TNF-alpha levels (p < 0.05). Additionally, the administration of RBO decreased the mean linear alveolar intercept (MLI) in the CSE-treated group. Additionally, RBO treatment significantly increased the total antioxidant capacity in both mouse BALF and serum. However, RBO did not have an effect on the malondialdehyde (MDA) level. These findings suggested that RBO treatment ameliorates lung inflammation in a CSE-induced emphysema mice model through anti-inflammatory and antioxidant pathways. Therefore, the supplementation of RBO could be a new potential therapeutic to relieve the severity of COPD.
Collapse
Affiliation(s)
- Aikkarach Kettawan
- Institute of Nutrition, Mahidol University, Nakhon Pathom 73170, Thailand; (A.K.); (A.K.K.); (N.N.)
| | - Sukpattaraporn Ruangklai
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand (J.T.)
| | - Thanaporn Rungruang
- Department of Anatomy, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand;
| | - Julalux Thongam
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand (J.T.)
| | | | - Nilesh Nirmal
- Institute of Nutrition, Mahidol University, Nakhon Pathom 73170, Thailand; (A.K.); (A.K.K.); (N.N.)
| | - Sorachai Srisuma
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand (J.T.)
| |
Collapse
|
2
|
Tiendrébéogo AJF, Soumagne T, Pellegrin F, Dagouassat M, Tran Van Nhieu J, Caramelle P, Paul EN, Even B, Zysman M, Julé Y, Samb A, Boczkowski J, Lanone S, Schlemmer F. The telomerase activator TA-65 protects from cigarette smoke-induced small airway remodeling in mice through extra-telomeric effects. Sci Rep 2023; 13:25. [PMID: 36646720 PMCID: PMC9842758 DOI: 10.1038/s41598-022-25993-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 12/07/2022] [Indexed: 01/18/2023] Open
Abstract
Small airway remodeling (SAR) is a key phenomenon of airflow obstruction in smokers, leading to chronic obstructive pulmonary disease (COPD). SAR results in an increased thickness of small airway walls, with a combination of peribronchiolar fibrosis with increased fibrous tissue and accumulation of mesenchymal and epithelial cells. SAR pathogenesis is still unclear but recent data suggest that alterations in telomerase activity could represent a possible underlying mechanism of SAR. Our study was dedicated to identify a potential protective role of TA-65, a pharmacological telomerase activator, in a cigarette smoke (CS) model of SAR in mice, and to further precise if extra-telomeric effects of telomerase, involving oxidative stress modulation, could explain it. C57BL/6J mice were daily exposed to air or CS during 4 weeks with or without a concomitant administration of TA-65 starting 7 days before CS exposure. Morphological analyses were performed, and mucus production, myofibroblast differentiation, collagen deposition, as well as transforming growth factor-β1 (TGF-β1) expression in the small airway walls were examined. In addition, the effects of TA-65 treatment on TGF-β expression, fibroblast-to-myofibroblast differentiation, reactive oxygen species (ROS) production and catalase expression and activity were evaluated in primary cultures of pulmonary fibroblasts and/or mouse embryonic fibroblasts in vitro. Exposure to CS during 4 weeks induced SAR in mice, characterized by small airway walls thickening and peribronchiolar fibrosis (increased deposition of collagen, expression of α-SMA in small airway walls), without mucus overproduction. Treatment of mice with TA-65 protected them from CS-induced SAR. This effect was associated with the prevention of CS-induced TGF-β expression in vivo, the blockade of TGF-β-induced myofibroblast differentiation, and the reduction of TGF-β-induced ROS production that correlates with an increase of catalase expression and activity. Our findings demonstrate that telomerase is a critical player of SAR, probably through extra-telomeric anti-oxidant effects, and therefore provide new insights in the understanding and treatment of COPD pathogenesis.
Collapse
Affiliation(s)
- Arnaud Jean Florent Tiendrébéogo
- grid.462410.50000 0004 0386 3258IMRB, INSERM U955, 94000 Créteil, France ,grid.410511.00000 0001 2149 7878Université Paris Est-Créteil, Faculté de Santé, 94000 Créteil, France ,Laboratoire de physiologie et d’explorations fonctionnelles physiologiques, Université Cheik Anta Diop, Dakar, Senegal
| | - Thibaud Soumagne
- grid.462410.50000 0004 0386 3258IMRB, INSERM U955, 94000 Créteil, France ,grid.410511.00000 0001 2149 7878Université Paris Est-Créteil, Faculté de Santé, 94000 Créteil, France
| | - François Pellegrin
- grid.462410.50000 0004 0386 3258IMRB, INSERM U955, 94000 Créteil, France ,grid.410511.00000 0001 2149 7878Université Paris Est-Créteil, Faculté de Santé, 94000 Créteil, France
| | - Maylis Dagouassat
- grid.462410.50000 0004 0386 3258IMRB, INSERM U955, 94000 Créteil, France ,grid.410511.00000 0001 2149 7878Université Paris Est-Créteil, Faculté de Santé, 94000 Créteil, France
| | - Jeanne Tran Van Nhieu
- grid.462410.50000 0004 0386 3258IMRB, INSERM U955, 94000 Créteil, France ,grid.410511.00000 0001 2149 7878Université Paris Est-Créteil, Faculté de Santé, 94000 Créteil, France ,grid.412116.10000 0004 1799 3934Assistance Publique Hôpitaux de Paris, Hôpitaux Universitaires Henri Mondor, Département de Pathologie, 94000 Créteil, France
| | - Philippe Caramelle
- grid.462410.50000 0004 0386 3258IMRB, INSERM U955, 94000 Créteil, France ,grid.410511.00000 0001 2149 7878Université Paris Est-Créteil, Faculté de Santé, 94000 Créteil, France
| | - Emmanuel N. Paul
- grid.462410.50000 0004 0386 3258IMRB, INSERM U955, 94000 Créteil, France ,grid.410511.00000 0001 2149 7878Université Paris Est-Créteil, Faculté de Santé, 94000 Créteil, France
| | - Benjamin Even
- grid.462410.50000 0004 0386 3258IMRB, INSERM U955, 94000 Créteil, France ,grid.410511.00000 0001 2149 7878Université Paris Est-Créteil, Faculté de Santé, 94000 Créteil, France
| | - Maeva Zysman
- grid.462410.50000 0004 0386 3258IMRB, INSERM U955, 94000 Créteil, France ,grid.410511.00000 0001 2149 7878Université Paris Est-Créteil, Faculté de Santé, 94000 Créteil, France
| | | | - Abdoulaye Samb
- Laboratoire de physiologie et d’explorations fonctionnelles physiologiques, Université Cheik Anta Diop, Dakar, Senegal
| | - Jorge Boczkowski
- grid.462410.50000 0004 0386 3258IMRB, INSERM U955, 94000 Créteil, France ,grid.410511.00000 0001 2149 7878Université Paris Est-Créteil, Faculté de Santé, 94000 Créteil, France ,grid.412116.10000 0004 1799 3934Assistance Publique Hôpitaux de Paris, Hôpitaux Universitaires Henri Mondor, Service d’explorations fonctionnelles respiratoires, DHU A-TVB, FHU Senec, 94000 Créteil, France
| | - Sophie Lanone
- grid.462410.50000 0004 0386 3258IMRB, INSERM U955, 94000 Créteil, France ,grid.410511.00000 0001 2149 7878Université Paris Est-Créteil, Faculté de Santé, 94000 Créteil, France
| | - Frédéric Schlemmer
- grid.462410.50000 0004 0386 3258IMRB, INSERM U955, 94000 Créteil, France ,grid.410511.00000 0001 2149 7878Université Paris Est-Créteil, Faculté de Santé, 94000 Créteil, France ,grid.412116.10000 0004 1799 3934Assistance Publique Hôpitaux de Paris, Hôpitaux Universitaires Henri Mondor, Unité de Pneumologie, DHU A-TVB, FHU Senec, 94000 Créteil, France
| |
Collapse
|
3
|
Saraiva-Romanholo BM, de Genaro IS, de Almeida FM, Felix SN, Lopes MRC, Amorim TS, Vieira RP, Arantes-Costa FM, Martins MA, de Fátima Lopes Calvo Tibério I, Prado CM. Exposure to Sodium Hypochlorite or Cigarette Smoke Induces Lung Injury and Mechanical Impairment in Wistar Rats. Inflammation 2022; 45:1464-1483. [PMID: 35501465 DOI: 10.1007/s10753-022-01625-0] [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: 12/11/2020] [Revised: 12/11/2020] [Accepted: 01/11/2022] [Indexed: 11/05/2022]
Abstract
Pulmonary irritants, such as cigarette smoke (CS) and sodium hypochlorite (NaClO), are associated to pulmonary diseases in cleaning workers. We examined whether their association affects lung mechanics and inflammation in Wistar rats. Exposure to these irritants alone induced alterations in the lung mechanics, inflammation, and remodeling. The CS increased airway cell infiltration, acid mucus production, MMP-12 expression, and alveolar enlargement. NaClO increased the number of eosinophils and macrophages in the bronchoalveolar lavage fluid, with cells expressing IL-13, MMP-12, MMP-9, TIMP-1, and iNOS in addition to increased IL-1β and TNF-α levels. Co-exposure to both irritants increased epithelial and smooth muscle cell area, acid mucus production, and IL-13 expression in the airways, while it reduced the lung inflammation. In conclusion, the co-exposure of CS with NaClO reduced the pulmonary inflammation, but increased the acidity of mucus, which may protect lungs from more injury. A cross-resistance in people exposed to multiple lung irritants should also be considered.
Collapse
Affiliation(s)
- Beatriz Mangueira Saraiva-Romanholo
- Sao Paulo Hospital (IAMSPE), Sao Paulo, Brazil.
- Department of Medicine, School of Medicine, University of Sao Paulo, LIM 20 Av. Dr. Arnaldo, 455 - Sala 1210, 1º andar, CEP: 01246903, Sao Paulo, Brazil.
- University City of Sao Paulo (UNICID), Sao Paulo, Brazil.
- Laboratory of Studies in Pulmonary Inflammation, Department of Biosciences, Federal University of Sao Paulo (UNIFESP), Santos, Brazil.
| | - Isabella Santos de Genaro
- Sao Paulo Hospital (IAMSPE), Sao Paulo, Brazil
- Department of Medicine, School of Medicine, University of Sao Paulo, LIM 20 Av. Dr. Arnaldo, 455 - Sala 1210, 1º andar, CEP: 01246903, Sao Paulo, Brazil
| | - Francine Maria de Almeida
- Department of Medicine, School of Medicine, University of Sao Paulo, LIM 20 Av. Dr. Arnaldo, 455 - Sala 1210, 1º andar, CEP: 01246903, Sao Paulo, Brazil
| | - Soraia Nogueira Felix
- Sao Paulo Hospital (IAMSPE), Sao Paulo, Brazil
- Department of Medicine, School of Medicine, University of Sao Paulo, LIM 20 Av. Dr. Arnaldo, 455 - Sala 1210, 1º andar, CEP: 01246903, Sao Paulo, Brazil
| | | | | | - Rodolfo Paula Vieira
- Post-Graduation Program in Bioengineering and in Biomedical Engineering, Brazil University, Sao Paulo, Brazil
- Brazilian Institute of Teaching and Research in Pulmonary and Exercise Immunology (IBEPIPE), Sao Jose dos Campos, Brazil
- Post-Graduation Program in Sciences of Human Movement and Rehabilitation, Federal University of São Paulo (UNIFESP), Santos, Brazil
- School of Medicine, Anhembi Morumbi University, Sao Jose dos Campos, SP, Brazil
| | - Fernanda Magalhães Arantes-Costa
- Department of Medicine, School of Medicine, University of Sao Paulo, LIM 20 Av. Dr. Arnaldo, 455 - Sala 1210, 1º andar, CEP: 01246903, Sao Paulo, Brazil
| | - Milton Arruda Martins
- Department of Medicine, School of Medicine, University of Sao Paulo, LIM 20 Av. Dr. Arnaldo, 455 - Sala 1210, 1º andar, CEP: 01246903, Sao Paulo, Brazil
| | - Iolanda de Fátima Lopes Calvo Tibério
- Department of Medicine, School of Medicine, University of Sao Paulo, LIM 20 Av. Dr. Arnaldo, 455 - Sala 1210, 1º andar, CEP: 01246903, Sao Paulo, Brazil
| | - Carla Máximo Prado
- Laboratory of Studies in Pulmonary Inflammation, Department of Biosciences, Federal University of Sao Paulo (UNIFESP), Santos, Brazil
| |
Collapse
|
4
|
Buccheri R, Duarte-Neto AN, Silva FLB, Haddad GC, Silva LBRD, Azevedo Netto R, Ledesma FL, Taborda CP, Benard G. Chronic exposure to cigarette smoke transiently worsens the disease course in a mouse model of pulmonary paracoccidioidomycosis. Rev Inst Med Trop Sao Paulo 2022; 64:e71. [DOI: 10.1590/s1678-9946202264071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
|
5
|
Sousa MV, Amaral AG, Freitas JA, Murata GM, Watanabe EH, Balbo BE, Tavares MD, Hortegal RA, Rocon C, Souza LE, Irigoyen MC, Salemi VM, Onuchic LF. Smoking accelerates renal cystic disease and worsens cardiac phenotype in Pkd1-deficient mice. Sci Rep 2021; 11:14443. [PMID: 34262092 PMCID: PMC8280209 DOI: 10.1038/s41598-021-93633-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 06/23/2021] [Indexed: 11/08/2022] Open
Abstract
Smoking has been associated with renal disease progression in ADPKD but the underlying deleterious mechanisms and whether it specifically worsens the cardiac phenotype remain unknown. To investigate these matters, Pkd1-deficient cystic mice and noncystic littermates were exposed to smoking from conception to 18 weeks of age and, along with nonexposed controls, were analyzed at 13-18 weeks. Renal cystic index and cyst-lining cell proliferation were higher in cystic mice exposed to smoking than nonexposed cystic animals. Smoking increased serum urea nitrogen in cystic and noncystic mice and independently enhanced tubular cell proliferation and apoptosis. Smoking also increased renal fibrosis, however this effect was much higher in cystic than in noncystic animals. Pkd1 deficiency and smoking showed independent and additive effects on reducing renal levels of glutathione. Systolic function and several cardiac structural parameters were also negatively affected by smoking and the Pkd1-deficient status, following independent and additive patterns. Smoking did not increase, however, cardiac apoptosis or fibrosis in cystic and noncystic mice. Notably, smoking promoted a much higher reduction in body weight in Pkd1-deficient than in noncystic animals. Our findings show that smoking aggravated the renal and cardiac phenotypes of Pkd1-deficient cystic mice, suggesting that similar effects may occur in human ADPKD.
Collapse
Affiliation(s)
- Marciana V Sousa
- Divisions of Nephrology and Molecular Medicine, Department of Medicine, University of São Paulo School of Medicine, Avenida Dr. Arnaldo, 455 - Sala 4304, São Paulo, SP, 01246-903, Brazil
| | - Andressa G Amaral
- Divisions of Nephrology and Molecular Medicine, Department of Medicine, University of São Paulo School of Medicine, Avenida Dr. Arnaldo, 455 - Sala 4304, São Paulo, SP, 01246-903, Brazil
| | - Jessica A Freitas
- Divisions of Nephrology and Molecular Medicine, Department of Medicine, University of São Paulo School of Medicine, Avenida Dr. Arnaldo, 455 - Sala 4304, São Paulo, SP, 01246-903, Brazil
| | - Gilson M Murata
- Divisions of Nephrology and Molecular Medicine, Department of Medicine, University of São Paulo School of Medicine, Avenida Dr. Arnaldo, 455 - Sala 4304, São Paulo, SP, 01246-903, Brazil
| | - Elieser H Watanabe
- Divisions of Nephrology and Molecular Medicine, Department of Medicine, University of São Paulo School of Medicine, Avenida Dr. Arnaldo, 455 - Sala 4304, São Paulo, SP, 01246-903, Brazil
| | - Bruno E Balbo
- Divisions of Nephrology and Molecular Medicine, Department of Medicine, University of São Paulo School of Medicine, Avenida Dr. Arnaldo, 455 - Sala 4304, São Paulo, SP, 01246-903, Brazil
| | - Marcelo D Tavares
- Heart Institute, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Renato A Hortegal
- Heart Institute, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Camila Rocon
- Heart Institute, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Leandro E Souza
- Heart Institute, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Maria C Irigoyen
- Heart Institute, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Vera M Salemi
- Heart Institute, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Luiz F Onuchic
- Divisions of Nephrology and Molecular Medicine, Department of Medicine, University of São Paulo School of Medicine, Avenida Dr. Arnaldo, 455 - Sala 4304, São Paulo, SP, 01246-903, Brazil.
| |
Collapse
|
6
|
ul Fatima N, Tursun B. Conversion of Germ Cells to Somatic Cell Types in C. elegans. J Dev Biol 2020; 8:E24. [PMID: 33036439 PMCID: PMC7712076 DOI: 10.3390/jdb8040024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/02/2020] [Accepted: 10/06/2020] [Indexed: 12/11/2022] Open
Abstract
The potential of a cell to produce all types of differentiated cells in an organism is termed totipotency. Totipotency is an essential property of germ cells, which constitute the germline and pass on the parental genetic material to the progeny. The potential of germ cells to give rise to a whole organism has been the subject of intense research for decades and remains important in order to better understand the molecular mechanisms underlying totipotency. A better understanding of the principles of totipotency in germ cells could also help to generate this potential in somatic cell lineages. Strategies such as transcription factor-mediated reprogramming of differentiated cells to stem cell-like states could benefit from this knowledge. Ensuring pluripotency or even totipotency of reprogrammed stem cells are critical improvements for future regenerative medicine applications. The C. elegans germline provides a unique possibility to study molecular mechanisms that maintain totipotency and the germ cell fate with its unique property of giving rise to meiotic cells Studies that focused on these aspects led to the identification of prominent chromatin-repressing factors such as the C. elegans members of the Polycomb Repressive Complex 2 (PRC2). In this review, we summarize different factors that were recently identified, which use molecular mechanisms such as control of protein translation or chromatin repression to ensure maintenance of totipotency and the germline fate. Additionally, we focus on recently identified factors involved in preventing transcription-factor-mediated conversion of germ cells to somatic lineages. These so-called reprogramming barriers have been shown in some instances to be conserved with regard to their function as a cell fate safeguarding factor in mammals. Overall, continued studies assessing the different aspects of molecular pathways involved in maintaining the germ cell fate in C. elegans may provide more insight into cell fate safeguarding mechanisms also in other species.
Collapse
Affiliation(s)
- Nida ul Fatima
- Berlin Institute of Medical Systems Biology, 10115 Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Baris Tursun
- Berlin Institute of Medical Systems Biology, 10115 Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| |
Collapse
|
7
|
Camargo CP, Pfann RZ, Kubrusly MS, Silva MFR, Guimarães ET, Leite MS, Saldiva PHN, Gemperli R. Study of the Effect of Hyperbaric Oxygen Therapy on the Viability of Dorsal Cutaneous Flaps in Tobacco-Exposed Rats. Aesthetic Plast Surg 2020; 44:979-985. [PMID: 32193614 DOI: 10.1007/s00266-020-01677-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 03/01/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND Smoking causes a threefold increase in the risk of surgical complications in flaps. Hyperbaric oxygen therapy (HBOT) increases the viability of chronic wounds. However, there are few studies concerning the effects of HBOT on surgical flaps in patients who smoke. This study aimed to analyze the effect of HBOT on the viability of cutaneous flaps in tobacco-exposed rats. METHODS Twenty Wistar rats were exposed to tobacco smoke for two months. Following this period, all animals underwent a dorsal cutaneous flap (3 × 10 cm) surgery and were divided into two groups: control (n = 10) and HBOT (n = 10). HBOT was performed in seven daily sessions (2 ATA, 90 min). After seven days, the animals were euthanized. The outcomes were total area, viable area, viable area/total area rate, analysis of dermal appendages and angiogenesis (hematoxylin-eosin), and gene expression analysis of iNOS and VEGF-a biomarkers. RESULTS The HBOT group showed an increase in viable area compared with the control group (84% versus 47%, p = 0.009, respectively). The HBOT group also showed an increase in appendage units (1.69 ± 0.54 versus 1.87 ± 0.58, p = 0.04) and angiogenesis density (1.29 ± 0.45 versus 1.82 ± 0.64, p < 0.001) compared to the control group. There was a difference between the control and HBOT groups in iNOS levels (0.926 ± 1.4 versus 0.04 ± 0.1 p = 0.002, respectively). However, this study did not show a difference between the groups concerning the gene expression of VEGF-a. CONCLUSION The use of hyperbaric oxygen therapy increased the viability of cutaneous flaps in tobacco-exposed rats and decreased iNOS mRNA levels; however, it did not change VEGF-a levels. Level of Evidence IV This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266.
Collapse
Affiliation(s)
- C P Camargo
- Laboratório de Microcirurgia E Cirurgia Plástica, Faculdade de Medicina, Universidade de São Paulo, Avenida Dr. Arnaldo, 455/1363, São Paulo, SP, 01246903, Brazil.
| | - R Z Pfann
- School of Medicine, Faculdade de Medicina, Universidade de São Paulo, Avenida Dr. Arnaldo, 455, São Paulo, SP, 01246903, Brazil
| | - M S Kubrusly
- Laboratório de Transplante de Cirurgia Do Fígado, Faculdade de Medicina, Universidade de São Paulo, Avenida Dr. Arnaldo, 455/2302, São Paulo, SP, 01246903, Brazil
| | - M F R Silva
- Laboratório de Poluição Atmosférica Experimental, Faculdade de Medicina, Universidade de São Paulo, Avenida Dr. Arnaldo, 455/1120, São Paulo, SP, 01246903, Brazil
| | - E T Guimarães
- Laboratório de Poluição Atmosférica Experimental, Faculdade de Medicina, Universidade de São Paulo, Avenida Dr. Arnaldo, 455/1120, São Paulo, SP, 01246903, Brazil
| | - M S Leite
- Laboratório de Poluição Atmosférica Experimental, Faculdade de Medicina, Universidade de São Paulo, Avenida Dr. Arnaldo, 455/1120, São Paulo, SP, 01246903, Brazil
| | - P H N Saldiva
- Laboratório de Poluição Atmosférica Experimental, Faculdade de Medicina, Universidade de São Paulo, Avenida Dr. Arnaldo, 455/1120, São Paulo, SP, 01246903, Brazil
| | - R Gemperli
- Laboratório de Microcirurgia E Cirurgia Plástica, Faculdade de Medicina, Universidade de São Paulo, Avenida Dr. Arnaldo, 455/1363, São Paulo, SP, 01246903, Brazil
| |
Collapse
|
8
|
Kohler JB, Cervilha DADB, Riani Moreira A, Santana FR, Farias TM, Alonso Vale MIC, Martins MDA, Prado CM, Tibério IC, Ito JT, Lopes FDTQDS. Microenvironmental stimuli induce different macrophage polarizations in experimental models of emphysema. Biol Open 2019; 8:bio.040808. [PMID: 30971412 PMCID: PMC6504010 DOI: 10.1242/bio.040808] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Macrophages play a pivotal role in the development of emphysema and depending on the microenvironment stimuli can be polarized into M1- or M2-like macrophage phenotypes. We compared macrophage polarizations in cigarette smoke (CS)- and porcine pancreatic elastase (PPE)-induced emphysema models. C57BL/6 mice were subdivided into four experimental groups. In the PPE group, animals received an intranasal instillation of PPE (0.677 IU); in the saline group, animals received an intranasal instillation of saline (0.9%). Animals from both groups were euthanized on day 28. In the CS group, animals were exposed to CS for 30 min, twice a day, 5 days per week for 12 weeks. In the control group, animals received filtered air. We observed an increase in total macrophages for both experimental models. For M1-like macrophage markers, we observed an increase in TNF-α+ and IFN-γ+ cells, Cxcl-9 and Cxcl-10 expressions in PPE and CS groups. Only in the CS group, we detected an increased expression of IL-12b For M2-like macrophages markers we observed a down regulation in IL-10, IL-4, IL-13, Arg1 and Fizz1 and an increase of TGF-β+ cells in the PPE group, while for the CS group there was an increase in TGF-β+ cells and IL-10 expression. All exposure groups were compared to their respective controls. In summary, we demonstrated that CS- and PPE-induced models resulted in different microenvironmental stimuli. CS exposure induced an environmental stimulus related to M1- and M2-like macrophage phenotypes similar to previous results described in COPD patients, whereas the elastase-induced model provided an environmental stimulus related only to the M1 phenotype.
Collapse
Affiliation(s)
- Júlia Benini Kohler
- Department of Clinical Medicine, Laboratory of Experimental Therapeutics (LIM-20), School of Medicine, University of São Paulo, São Paulo 01246-903, Brazil
| | - Daniela Aparecida de Brito Cervilha
- Department of Clinical Medicine, Laboratory of Experimental Therapeutics (LIM-20), School of Medicine, University of São Paulo, São Paulo 01246-903, Brazil
| | - Alyne Riani Moreira
- Department of Clinical Medicine, Laboratory of Experimental Therapeutics (LIM-20), School of Medicine, University of São Paulo, São Paulo 01246-903, Brazil
| | | | - Talita M Farias
- Department of Bioscience, Federal University of São Paulo, Diadema 09961-400, Brazil
| | | | - Milton de Arruda Martins
- Department of Clinical Medicine, Laboratory of Experimental Therapeutics (LIM-20), School of Medicine, University of São Paulo, São Paulo 01246-903, Brazil
| | - Carla Máximo Prado
- Department of Biological Science, Federal University of São Paulo, Santos 09972-270, Brazil
| | - Iolanda Calvo Tibério
- Department of Clinical Medicine, Laboratory of Experimental Therapeutics (LIM-20), School of Medicine, University of São Paulo, São Paulo 01246-903, Brazil
| | - Juliana Tiyaki Ito
- Department of Clinical Medicine, Laboratory of Experimental Therapeutics (LIM-20), School of Medicine, University of São Paulo, São Paulo 01246-903, Brazil
| | | |
Collapse
|
9
|
iNOS Inhibition Reduces Lung Mechanical Alterations and Remodeling Induced by Particulate Matter in Mice. Pulm Med 2019; 2019:4781528. [PMID: 30984425 PMCID: PMC6432736 DOI: 10.1155/2019/4781528] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 02/07/2019] [Indexed: 12/14/2022] Open
Abstract
Background. The epidemiologic association between pulmonary exposure to ambient particulate matter (PM) and acute lung damage is well known. However, the mechanism involved in the effects of repeated exposures of PM in the lung injury is poorly documented. This study tested the hypotheses that chronic nasal instillation of residual oil fly ash (ROFA) induced not only distal lung and airway inflammation but also remodeling. In addition, we evaluated the effects of inducible nitric oxide inhibition in these responses. For this purpose, airway and lung parenchyma were evaluated by quantitative analysis of collagen and elastic fibers, immunohistochemistry for macrophages, neutrophils, inducible nitric oxide synthase (iNOS), neuronal nitric oxide synthase (nNOS), and alveolar septa 8-iso prostaglandin F2α (8-iso-PGF-2α) detection. Anesthetized in vivo (airway resistance, elastance, H, G, and Raw) respiratory mechanics were also analyzed. C57BL6 mice received daily 60ul of ROFA (intranasal) for five (ROFA-5d) or fifteen days (ROFA-15d). Controls have received saline (SAL). Part of the animals has received 1400W (SAL+1400W and ROFA-15d+1400W), an iNOS inhibitor, for four days before the end of the protocol. A marked neutrophil and macrophage infiltration and an increase in the iNOS, nNOS, and 8-iso-PGF2 α expression was observed in peribronchiolar and alveolar wall both in ROFA-5d and in ROFA-15d groups. There was an increment of the collagen and elastic fibers in alveolar and airway walls in ROFA-15d group. The iNOS inhibition reduced all alterations induced by ROFA, except for the 8-iso-PGF2 α expression. In conclusion, repeated particulate matter exposures induce extracellular matrix remodeling of airway and alveolar walls, which could contribute to the pulmonary mechanical changes observed. The mechanism involved is, at least, dependent on the inducible nitric oxide activation.
Collapse
|
10
|
Camargo CP, Frassei RD, Silva DIDSBCE, Pfann RZ, da Silva LDCM, Morais-Besteiro J, Gemperli R. Hyaluronic acid in tobacco-exposed rats. Inflammatory reaction, and duration of effect1. Acta Cir Bras 2019; 34:e201900202. [PMID: 30843935 PMCID: PMC6585909 DOI: 10.1590/s0102-8650201900202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 12/21/2018] [Accepted: 01/22/2019] [Indexed: 11/26/2022] Open
Abstract
PURPOSE To evaluate the hyaluronic acid (HA) inflammatory reaction, fibroblasts, fibrosis and duration of effect in the dorsal region of tobacco-exposed rats. METHODS Ten Wistar rats were divided into two groups: tobacco-exposed-group (TEG;n=5) and air-control-group (CG;n=5). The TEG animals were tobacco-exposed twice a day, 30-minutes/session, during 60 days. After this period, all animals received 0.1 mL HA subcutaneous injection in the dorsal area. The volume of HA was measured immediately after HA injection and weekly using a hand-caliper in nine weeks. After this period, all the animals were euthanized, and a specimen of was collected to evaluate inflammatory cells, fibroblasts, and fibrosis by HE. RESULTS This study showed a higher inflammatory reaction in TEG than CG: inflammatory cell-count (CG: 1.07±0.9; TEG: 8.61±0.36, p<0.001); fibroblast count (CG: 2.92±0.17; TEG: 19.14±0.62, p<0.001), and fibrosis quantification (CG: 2.0; TEG: 3.75, p<0.001). The analysis of the HA volume in nine weeks in the dorsal region did not show a difference between groups (p=0.39). CONCLUSIONS This study suggested that the HA injection in the TEG caused an increase in inflammatory cell count, fibroblast, and fibrosis quantification when compared to the CG. There was no difference in the duration of effect of HA between the groups.
Collapse
Affiliation(s)
- Cristina Pires Camargo
- MD, Division of Plastic Surgery, Hospital das Clínicas, Laboratory
of Microsurgery and Plastic Surgery (LIM-04), Medical School, Universidade de São
Paulo (USP), Brazil. Intellectual and scientific content of the study,
interpretation of data, statistics analysis, manuscript writing, critical
revision
| | - Renan Dias Frassei
- Graduate student, Laboratory of Microsurgery and Plastic Surgery
(LIM-04), Medical School, USP, Sao Paulo-SP, Brazil. Acquisition, analysis and
interpretation of data; technical procedures
| | | | - Robert Zawadzki Pfann
- Graduate student, Laboratory of Microsurgery and Plastic Surgery
(LIM-04), Medical School, USP, Sao Paulo-SP, Brazil. Acquisition, analysis and
interpretation of data; technical procedures
| | - Luiza de Campos Moreira da Silva
- Graduate student, Laboratory of Microsurgery and Plastic Surgery
(LIM-04), Medical School, USP, Sao Paulo-SP, Brazil. Acquisition, analysis and
interpretation of data; technical procedures
| | - Julio Morais-Besteiro
- MD, Division of Plastic Surgery, Hospital das Clínicas, Laboratory
of Microsurgery and Plastic Surgery (LIM-04), Medical School, Universidade de São
Paulo (USP), Brazil. Intellectual and scientific content of the study,
interpretation of data, statistics analysis, manuscript writing, critical
revision
| | - Rolf Gemperli
- PhD, Division of Plastic Surgery, Hospital das Clínicas,
Laboratory of Microsurgery and Plastic Surgery (LIM-04), Medical School, USP, Sao
Paulo-SP, Brazil. Intellectual and scientific content of the study, interpretation
of data, critical revision
| |
Collapse
|
11
|
Strzelak A, Ratajczak A, Adamiec A, Feleszko W. Tobacco Smoke Induces and Alters Immune Responses in the Lung Triggering Inflammation, Allergy, Asthma and Other Lung Diseases: A Mechanistic Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2018; 15:E1033. [PMID: 29883409 PMCID: PMC5982072 DOI: 10.3390/ijerph15051033] [Citation(s) in RCA: 353] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 05/08/2018] [Accepted: 05/09/2018] [Indexed: 02/06/2023]
Abstract
Many studies have been undertaken to reveal how tobacco smoke skews immune responses contributing to the development of chronic obstructive pulmonary disease (COPD) and other lung diseases. Recently, environmental tobacco smoke (ETS) has been linked with asthma and allergic diseases in children. This review presents the most actual knowledge on exact molecular mechanisms responsible for the skewed inflammatory profile that aggravates inflammation, promotes infections, induces tissue damage, and may promote the development of allergy in individuals exposed to ETS. We demonstrate how the imbalance between oxidants and antioxidants resulting from exposure to tobacco smoke leads to oxidative stress, increased mucosal inflammation, and increased expression of inflammatory cytokines (such as interleukin (IL)-8, IL-6 and tumor necrosis factor α ([TNF]-α). Direct cellular effects of ETS on epithelial cells results in increased permeability, mucus overproduction, impaired mucociliary clearance, increased release of proinflammatory cytokines and chemokines, enhanced recruitment of macrophages and neutrophils and disturbed lymphocyte balance towards Th2. The plethora of presented phenomena fully justifies a restrictive policy aiming at limiting the domestic and public exposure to ETS.
Collapse
Affiliation(s)
- Agnieszka Strzelak
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warszawa, Poland.
| | - Aleksandra Ratajczak
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warszawa, Poland.
| | - Aleksander Adamiec
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warszawa, Poland.
| | - Wojciech Feleszko
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, Zwirki i Wigury 61, 02-091 Warszawa, Poland.
| |
Collapse
|
12
|
Drummond D, Baravalle-Einaudi M, Lezmi G, Vibhushan S, Franco-Montoya ML, Hadchouel A, Boczkowski J, Delacourt C. Combined Effects of in Utero and Adolescent Tobacco Smoke Exposure on Lung Function in C57Bl/6J Mice. ENVIRONMENTAL HEALTH PERSPECTIVES 2017; 125:392-399. [PMID: 27814244 PMCID: PMC5332197 DOI: 10.1289/ehp54] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 10/10/2016] [Accepted: 10/13/2016] [Indexed: 05/30/2023]
Abstract
BACKGROUND Fetal determinants of airway function, such as in utero exposure to maternal cigarette smoke (CS), may create a predisposition to adult airflow obstruction and chronic obstructive pulmonary disease (COPD) in adulthood. It has been suggested that active smoking in adolescence and preexisting airflow obstruction have synergistic deleterious effects. OBJECTIVE We used a mouse model to investigate whether there is a synergistic effect of exposure to CS in utero and during adolescence on lung function. METHODS Female C57Bl/6J mice were exposed to CS or to filtered room air during pregnancy. Exposure to CS began 2 weeks before mating and continued until delivery. After birth, the pups were not exposed to CS until day 21 (D21). Between D21 and D49, corresponding to "adolescence," litters were randomized for an additional 4 weeks of exposure to CS. Lung morphometry, lung mechanics, and the expression of genes involved in senescence were evaluated in different subsets of mice on D21 and D49. RESULTS In utero exposure to CS induced significant lung function impairment by D21. CS exposure between D21 and D49 induced significant functional impairment only in mice exposed to CS prenatally. On D49, no difference was observed between subgroups in terms of lung p53, p16, p21, and Bax mRNA levels. CONCLUSIONS Our findings suggest that prenatal and adolescent CS exposure have a synergistic effect on lung function in mice. The combined effect did not appear to be a consequence of early pulmonary senescence. Citation: Drummond D, Baravalle-Einaudi M, Lezmi G, Vibhushan S, Franco-Montoya ML, Hadchouel A, Boczkowski J, Delacourt C. 2017. Combined effects of in utero and adolescent tobacco smoke exposure on lung function in C57Bl/6J mice. Environ Health Perspect 125:392-399; http://dx.doi.org/10.1289/EHP54.
Collapse
Affiliation(s)
- David Drummond
- INSERM (Institut National de la Santé et de la Recherche Médicale), U955, Equipe 04, IMRB (Institut Mondor de Recherche Biomédicale), Créteil, France
| | - Mélissa Baravalle-Einaudi
- INSERM (Institut National de la Santé et de la Recherche Médicale), U955, Equipe 04, IMRB (Institut Mondor de Recherche Biomédicale), Créteil, France
| | - Guillaume Lezmi
- INSERM (Institut National de la Santé et de la Recherche Médicale), U955, Equipe 04, IMRB (Institut Mondor de Recherche Biomédicale), Créteil, France
| | - Shamila Vibhushan
- INSERM (Institut National de la Santé et de la Recherche Médicale), U955, Equipe 04, IMRB (Institut Mondor de Recherche Biomédicale), Créteil, France
| | - Marie-Laure Franco-Montoya
- INSERM (Institut National de la Santé et de la Recherche Médicale), U955, Equipe 04, IMRB (Institut Mondor de Recherche Biomédicale), Créteil, France
| | - Alice Hadchouel
- INSERM (Institut National de la Santé et de la Recherche Médicale), U955, Equipe 04, IMRB (Institut Mondor de Recherche Biomédicale), Créteil, France
- Pneumologie Pédiatrique, Necker, AP-HP (Assistance Publique-Hôpitaux de Paris), France; Centre de Référence des Maladies Respiratoires Rares, Paris, France
- Université Paris-Descartes, Paris, France
| | - Jorge Boczkowski
- INSERM (Institut National de la Santé et de la Recherche Médicale), U955, Equipe 04, IMRB (Institut Mondor de Recherche Biomédicale), Créteil, France
| | - Christophe Delacourt
- INSERM (Institut National de la Santé et de la Recherche Médicale), U955, Equipe 04, IMRB (Institut Mondor de Recherche Biomédicale), Créteil, France
- Pneumologie Pédiatrique, Necker, AP-HP (Assistance Publique-Hôpitaux de Paris), France; Centre de Référence des Maladies Respiratoires Rares, Paris, France
- Université Paris-Descartes, Paris, France
| |
Collapse
|
13
|
Camargo CP, Fernandes FA, Lee MHM, Silva LCM, Besteiro JM, Gemperli R. The positive effect of Botulinum toxin type A on the viability of random flap in tobacco exposed in rats. Acta Cir Bras 2016; 31:720-723. [DOI: 10.1590/s0102-865020160110000003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 10/13/2016] [Indexed: 11/21/2022] Open
|
14
|
Mazzoli-Rocha F, Oliveira VR, Barcellos BC, Moreira DKM, Saldiva PHN, Faffe DS, Zin WA. Time-dependency of mice lung recovery after a 4-week exposure to traffic or biomass air pollutants. Respir Physiol Neurobiol 2016; 230:16-21. [PMID: 27179431 DOI: 10.1016/j.resp.2016.05.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 05/07/2016] [Accepted: 05/10/2016] [Indexed: 10/21/2022]
Abstract
The time-dependency of lung recovery after 3 intranasal instillations per week during four weeks of distilled water (C groups) or particles (15μg) from traffic (U groups) or biomass burning (B groups) was observed in BALB/c mice. Lung mechanics [static elastance (Est), viscoelastic component of elastance (ΔE), lung resistive (ΔP1) and viscoelastic/inhomogeneous (ΔP2) pressures] and histology were analyzed 1 (C1, U1, B1), 2 (C2, U2, B2), 7 (C7, U7, B7) or 14 days (C14, U14, B14) after the last instillation. Est, ΔE, ΔP1 and ΔP2 were higher in U1 and B1 than in C1, returning to control values at day 2, except for ΔP1 that normalized after 7 days. Alveolar collapse, bronchoconstriction index and alveolar lesion were larger in U1 and B1 than in C1, however collapse returned to baseline at 7 days, while the others normalized in 2 days. A 4-week exposure to U and B induced lung impairment that resolved 7 days after the last exposure.
Collapse
Affiliation(s)
- Flavia Mazzoli-Rocha
- Laboratory of Respiration Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of do Rio de Janeiro, Brazil
| | - Vinícius Rosa Oliveira
- Laboratory of Respiration Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of do Rio de Janeiro, Brazil
| | - Bárbara Chaves Barcellos
- Laboratory of Respiration Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of do Rio de Janeiro, Brazil
| | - Dayse Kelly Molina Moreira
- Laboratory of Respiration Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of do Rio de Janeiro, Brazil
| | - Paulo Hilário Nascimento Saldiva
- Laboratory of Experimental Air Pollution, Department of Pathology, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Débora Souza Faffe
- Laboratory of Respiration Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of do Rio de Janeiro, Brazil
| | - Walter Araújo Zin
- Laboratory of Respiration Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of do Rio de Janeiro, Brazil.
| |
Collapse
|
15
|
Zou W, Liu S, Hu J, Sheng Q, He F, Li B, Ran P. Nicotine reduces the levels of surfactant proteins A and D via Wnt/β-catenin and PKC signaling in human airway epithelial cells. Respir Physiol Neurobiol 2016; 221:1-10. [DOI: 10.1016/j.resp.2015.10.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 10/07/2015] [Accepted: 10/08/2015] [Indexed: 12/12/2022]
|
16
|
Camargo CP, Jacomo AL, Battlehner CN, Lemos M, Saldiva PH, Martins MA, Munhoz AM, Gemperli R. Botulinum toxin type A on cutaneous flap viability in diabetic and tobacco-exposed rats. Acta Cir Bras 2015; 30:639-45. [DOI: 10.1590/s0102-865020150090000009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 08/17/2015] [Indexed: 11/22/2022] Open
|
17
|
Peron JPS, de Brito AA, Pelatti M, Brandão WN, Vitoretti LB, Greiffo FR, da Silveira EC, Oliveira-Junior MC, Maluf M, Evangelista L, Halpern S, Nisenbaum MG, Perin P, Czeresnia CE, Câmara NOS, Aimbire F, Vieira RDP, Zatz M, Ligeiro de Oliveira AP. Human Tubal-Derived Mesenchymal Stromal Cells Associated with Low Level Laser Therapy Significantly Reduces Cigarette Smoke-Induced COPD in C57BL/6 mice. PLoS One 2015; 10:e0136942. [PMID: 26322981 PMCID: PMC4554986 DOI: 10.1371/journal.pone.0136942] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 08/11/2015] [Indexed: 01/06/2023] Open
Abstract
Cigarette smoke-induced chronic obstructive pulmonary disease is a very debilitating disease, with a very high prevalence worldwide, which results in a expressive economic and social burden. Therefore, new therapeutic approaches to treat these patients are of unquestionable relevance. The use of mesenchymal stromal cells (MSCs) is an innovative and yet accessible approach for pulmonary acute and chronic diseases, mainly due to its important immunoregulatory, anti-fibrogenic, anti-apoptotic and pro-angiogenic. Besides, the use of adjuvant therapies, whose aim is to boost or synergize with their function should be tested. Low level laser (LLL) therapy is a relatively new and promising approach, with very low cost, no invasiveness and no side effects. Here, we aimed to study the effectiveness of human tube derived MSCs (htMSCs) cell therapy associated with a 30mW/3J-660 nm LLL irradiation in experimental cigarette smoke-induced chronic obstructive pulmonary disease. Thus, C57BL/6 mice were exposed to cigarette smoke for 75 days (twice a day) and all experiments were performed on day 76. Experimental groups receive htMSCS either intraperitoneally or intranasally and/or LLL irradiation either alone or in association. We show that co-therapy greatly reduces lung inflammation, lowering the cellular infiltrate and pro-inflammatory cytokine secretion (IL-1β, IL-6, TNF-α and KC), which were followed by decreased mucus production, collagen accumulation and tissue damage. These findings seemed to be secondary to the reduction of both NF-κB and NF-AT activation in lung tissues with a concomitant increase in IL-10. In summary, our data suggests that the concomitant use of MSCs + LLLT may be a promising therapeutic approach for lung inflammatory diseases as COPD.
Collapse
Affiliation(s)
- Jean Pierre Schatzmann Peron
- Neuroimmune Interactions Laboratory, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, SP, Brazil
| | - Auriléia Aparecida de Brito
- Laboratory of Pulmonary and Exercise Immunology–LABPEI, Nove de Julho University (UNINOVE), São Paulo, SP, Brazil
| | - Mayra Pelatti
- Division of Human Genome Research Center, Biosciences Institute, University of São Paulo, São Paulo, SP, Brazil
| | - Wesley Nogueira Brandão
- Neuroimmune Interactions Laboratory, Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, SP, Brazil
| | - Luana Beatriz Vitoretti
- Laboratory of Pulmonary and Exercise Immunology–LABPEI, Nove de Julho University (UNINOVE), São Paulo, SP, Brazil
| | - Flávia Regina Greiffo
- Laboratory of Pulmonary and Exercise Immunology–LABPEI, Nove de Julho University (UNINOVE), São Paulo, SP, Brazil
| | - Elaine Cristina da Silveira
- Laboratory of Pulmonary and Exercise Immunology–LABPEI, Nove de Julho University (UNINOVE), São Paulo, SP, Brazil
| | | | - Mariangela Maluf
- CEERH—Specialized Center for Human Reproduction, Division of Reproductive Medicine, São Paulo, SP, Brazil
| | | | - Silvio Halpern
- Division of Reproductive Medicine—Célula Mater, São Paulo, SP, Brazil
| | | | - Paulo Perin
- CEERH—Specialized Center for Human Reproduction, Division of Reproductive Medicine, São Paulo, SP, Brazil
| | | | - Niels Olsen Saraiva Câmara
- Laboratory of Transplantation Immunobiology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Flávio Aimbire
- Department of Science and Technology, Federal University of São Paulo, São José dos Campos, SP, Brazil
| | - Rodolfo de Paula Vieira
- Division of Human Genome Research Center, Biosciences Institute, University of São Paulo, São Paulo, SP, Brazil
| | - Mayana Zatz
- Laboratory of Pulmonary and Exercise Immunology–LABPEI, Nove de Julho University (UNINOVE), São Paulo, SP, Brazil
| | | |
Collapse
|
18
|
Taguchi L, Pinheiro NM, Olivo CR, Choqueta-Toledo A, Grecco SS, Lopes FDTQS, Caperuto LC, Martins MA, Tiberio IFLC, Câmara NO, Lago JHG, Prado CM. A flavanone from Baccharis retusa (Asteraceae) prevents elastase-induced emphysema in mice by regulating NF-κB, oxidative stress and metalloproteinases. Respir Res 2015; 16:79. [PMID: 26122092 PMCID: PMC4489216 DOI: 10.1186/s12931-015-0233-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 06/08/2015] [Indexed: 01/02/2023] Open
Abstract
Background Pulmonary emphysema is characterized by irreversible airflow obstruction, inflammation, oxidative stress imbalance and lung remodeling, resulting in reduced lung function and a lower quality of life. Flavonoids are plant compounds with potential anti-inflammatory and antioxidant effects that have been used in folk medicine. Our aim was to determine whether treatment with sakuranetin, a flavonoid extracted from the aerial parts of Baccharis retusa, interferes with the development of lung emphysema. Methods Intranasal saline or elastase was administered to mice; the animals were then treated with sakuranetin or vehicle 2 h later and again on days 7, 14 and 28. We evaluated lung function and the inflammatory profile in bronchoalveolar lavage fluid (BALF). The lungs were removed to evaluate alveolar enlargement, extracellular matrix fibers and the expression of MMP-9, MMP-12, TIMP-1, 8-iso-PGF-2α and p65-NF-κB in the fixed tissues as well as to evaluate cytokine levels and p65-NF-κB protein expression. Results In the elastase-treated animals, sakuranetin treatment reduced the alveolar enlargement, collagen and elastic fiber deposition and the number of MMP-9- and MMP-12-positive cells but increased TIMP-1 expression. In addition, sakuranetin treatment decreased the inflammation and the levels of TNF-α, IL-1β and M-CSF in the BALF as well as the levels of NF-κB and 8-iso-PGF-2α in the lungs of the elastase-treated animals. However, this treatment did not affect the changes in lung function. Conclusion These data emphasize the importance of oxidative stress and metalloproteinase imbalance in the development of emphysema and suggest that sakuranetin is a potent candidate that should be further investigated as an emphysema treatment. This compound may be useful for counteracting lung remodeling and oxidative stress and thus attenuating the development of emphysema. Electronic supplementary material The online version of this article (doi:10.1186/s12931-015-0233-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Laura Taguchi
- Department of Biological Science, Universidade Federal de São Paulo, Rua Artur Riedel, 275 - Eldorado, Diadema, SP, Brazil
| | - Nathalia M Pinheiro
- Department of Medicine, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Clarice R Olivo
- Department of Medicine, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | | | - Simone S Grecco
- Department of Exact and Earth Sciences, Universidade Federal de São Paulo, Diadema, Brazil
| | - Fernanda D T Q S Lopes
- Department of Medicine, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Luciana C Caperuto
- Department of Biological Science, Universidade Federal de São Paulo, Rua Artur Riedel, 275 - Eldorado, Diadema, SP, Brazil
| | - Mílton A Martins
- Department of Medicine, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Iolanda F L C Tiberio
- Department of Medicine, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Niels O Câmara
- Department of Immunology, Biological Institute, Universidade de São Paulo, São Paulo, Brazil
| | - João Henrique G Lago
- Department of Exact and Earth Sciences, Universidade Federal de São Paulo, Diadema, Brazil
| | - Carla M Prado
- Department of Biological Science, Universidade Federal de São Paulo, Rua Artur Riedel, 275 - Eldorado, Diadema, SP, Brazil. .,Department of Medicine, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil.
| |
Collapse
|
19
|
Yan H, Zhao L, Wu X, Liu H, Wu C, Li Y, Zheng W, Jiang H. Inflammation and pathological damage to the lungs of mice are only partially reversed following smoking cessation on subacute exposure to cigarette smoke. Mol Med Rep 2015; 11:4246-54. [PMID: 25672547 PMCID: PMC4394953 DOI: 10.3892/mmr.2015.3337] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 01/21/2015] [Indexed: 02/07/2023] Open
Abstract
The present study aimed to observe the level of inflammation and the number of lesions in the airways and parenchyma of mouse lungs subsequent to smoking cessation following 4 weeks exposure to cigarette smoke. Enlargement of the regional airspaces, deposition of peribronchial collagen fibers and macrophage infiltration were assessed. In addition, the expression levels of matrix metalloproteinase (MMP)‑12 and transforming growth factor (TGF)‑β1 were detected in the airways and lung parenchyma of C57BL/6 J mice. Mice, which were exposed to filtered air for 4 weeks or cigarette smoke for 8 weeks were used as control groups. A 4 week duration of smoke exposure induced the expansion of alveolar spaces ~100 µm from the terminal bronchioles, but without increased deposition of collagen around the small airways, which was not reversed following smoking cessation. Pulmonary infiltration of macrophages and the protein expression levels of MMP‑12 and TGF‑β1 increased in the airways following 4 weeks smoke exposure, however, there was no further increase at 8 weeks, and the expression levels of TGF‑β1 in the lung parenchyma decreased. At 4 weeks post‑smoking cessation, the expression levels of TGF‑β1 in the airways and lung parenchyma returned to normal; whereas, 1 week after smoking cessation, the expression levels of MMP‑12 were higher compared with the normal control group. Subacute exposure to cigarette smoke induced an inflammatory response and regional damage to the lung parenchyma, prior to deposition of collagen around the airways. Following smoking cessation, the pulmonary inflammatory reaction was partially reversed, however, macrophage infiltration and the expression levels of MMP‑12 remained significantly higher compared with the control mice. These results suggested that regulation of the expression of MMP‑12 and TGF‑β1, particularly in the distribution in the airways and lung parenchyma, may be a strategy for the early treatment of chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Hengyi Yan
- Department of First Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Li Zhao
- Department of First Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Xiaojie Wu
- Department of First Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Hongbo Liu
- Department of First Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Cen Wu
- Department of First Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Yu Li
- Department of First Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Wei Zheng
- Department of First Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Hongfang Jiang
- Department of First Respiratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| |
Collapse
|
20
|
Yoshizaki K, Brito JM, Moriya HT, Toledo AC, Ferzilan S, Ligeiro de Oliveira AP, Machado ID, Farsky SHP, Silva LFF, Martins MA, Saldiva PHN, Mauad T, Macchione M. Chronic exposure of diesel exhaust particles induces alveolar enlargement in mice. Respir Res 2015; 16:18. [PMID: 25848680 PMCID: PMC4345004 DOI: 10.1186/s12931-015-0172-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Accepted: 01/17/2015] [Indexed: 12/21/2022] Open
Abstract
Background Diesel exhaust particles (DEPs) are deposited into the respiratory tract and are thought to be a risk factor for the development of diseases of the respiratory system. In healthy individuals, the timing and mechanisms of respiratory tract injuries caused by chronic exposure to air pollution remain to be clarified. Methods We evaluated the effects of chronic exposure to DEP at doses below those found in a typical bus corridor in Sao Paulo (150 μg/m3). Male BALB/c mice were divided into mice receiving a nasal instillation: saline (saline; n = 30) and 30 μg/10 μL of DEP (DEP; n = 30). Nasal instillations were performed five days a week, over a period of 90 days. Bronchoalveolar lavage (BAL) was performed, and the concentrations of interleukin (IL)-4, IL-10, IL-13 and interferon-gamma (INF-γ) were determined by ELISA-immunoassay. Assessment of respiratory mechanics was performed. The gene expression of Muc5ac in lung was evaluated by RT-PCR. The presence of IL-13, MAC2+ macrophages, CD3+, CD4+, CD8+ T cells and CD20+ B cells in tissues was analysed by immunohistochemistry. Bronchial thickness and the collagen/elastic fibers density were evaluated by morphometry. We measured the mean linear intercept (Lm), a measure of alveolar distension, and the mean airspace diameter (D0) and statistical distribution (D2). Results DEP decreased IFN-γ levels in BAL (p = 0.03), but did not significantly alter IL-4, IL-10 and IL-13 levels. MAC2+ macrophage, CD4+ T cell and CD20+ B cell numbers were not altered; however, numbers of CD3+ T cells (p ≤ 0.001) and CD8+ T cells (p ≤ 0.001) increased in the parenchyma. Although IL-13 (p = 0.008) expression decreased in the bronchiolar epithelium, Muc5ac gene expression was not altered in the lung of DEP-exposed animals. Although respiratory mechanics, elastic and collagen density were not modified, the mean linear intercept (Lm) was increased in the DEP-exposed animals (p ≤ 0.001), and the index D2 was statistically different (p = 0.038) from the control animals. Conclusion Our data suggest that nasal instillation of low doses of DEP over a period of 90 days results in alveolar enlargement in the pulmonary parenchyma of healthy mice. Electronic supplementary material The online version of this article (doi:10.1186/s12931-015-0172-z) contains supplementary material, which is available to authorized users.
Collapse
|
21
|
Eurlings IMJ, Reynaert NL, van den Beucken T, Gosker HR, de Theije CC, Verhamme FM, Bracke KR, Wouters EFM, Dentener MA. Cigarette smoke extract induces a phenotypic shift in epithelial cells; involvement of HIF1α in mesenchymal transition. PLoS One 2014; 9:e107757. [PMID: 25329389 PMCID: PMC4199572 DOI: 10.1371/journal.pone.0107757] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 08/11/2014] [Indexed: 12/22/2022] Open
Abstract
In COPD, matrix remodeling contributes to airflow limitation. Recent evidence suggests that next to fibroblasts, the process of epithelial-mesenchymal transition can contribute to matrix remodeling. CSE has been shown to induce EMT in lung epithelial cells, but the signaling mechanisms involved are largely unknown and subject of this study. EMT was assessed in A549 and BEAS2B cells stimulated with CSE by qPCR, Western blotting and immunofluorescence for epithelial and mesenchymal markers, as were collagen production, cell adhesion and barrier integrity as functional endpoints. Involvement of TGF-β and HIF1α signaling pathways were investigated. In addition, mouse models were used to examine the effects of CS on hypoxia signaling and of hypoxia per se on mesenchymal expression. CSE induced EMT characteristics in A549 and BEAS2B cells, evidenced by decreased expression of epithelial markers and a concomitant increase in mesenchymal marker expression after CSE exposure. Furthermore cells that underwent EMT showed increased production of collagen, decreased adhesion and disrupted barrier integrity. The induction of EMT was found to be independent of TGF-β signaling. On the contrary, CS was able to induce hypoxic signaling in A549 and BEAS2B cells as well as in mice lung tissue. Importantly, HIF1α knock-down prevented induction of mesenchymal markers, increased collagen production and decreased adhesion after CSE exposure, data that are in line with the observed induction of mesenchymal marker expression by hypoxia in vitro and in vivo. Together these data provide evidence that both bronchial and alveolar epithelial cells undergo a functional phenotypic shift in response to CSE exposure which can contribute to increased collagen deposition in COPD lungs. Moreover, HIF1α signaling appears to play an important role in this process.
Collapse
Affiliation(s)
- Irene M. J. Eurlings
- Department of Respiratory Medicine, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Niki L. Reynaert
- Department of Respiratory Medicine, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Twan van den Beucken
- Department of Radiation Oncology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Harry R. Gosker
- Department of Respiratory Medicine, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - C. C. de Theije
- Department of Respiratory Medicine, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Fien M. Verhamme
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Ken R. Bracke
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Emiel F. M. Wouters
- Department of Respiratory Medicine, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Mieke A. Dentener
- Department of Respiratory Medicine, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| |
Collapse
|
22
|
Carvalho GMC, Nagato LKDS, Fagundes SDS, Dos Santos FB, Calheiros AS, Malm O, Bozza PT, Saldiva PHN, Faffe DS, Rocco PRM, Zin WA. Time course of pulmonary burden in mice exposed to residual oil fly ash. Front Physiol 2014; 5:366. [PMID: 25309454 PMCID: PMC4174882 DOI: 10.3389/fphys.2014.00366] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 09/07/2014] [Indexed: 01/15/2023] Open
Abstract
Residual oil fly ash (ROFA) is a common pollutant in areas where oil is burned. This particulate matter (PM) with a broad distribution of particle diameters can be inhaled by human beings and putatively damage their respiratory system. Although some studies deal with cultured cells, animals, and even epidemiological issues, so far a comprehensive analysis of respiratory outcomes as a function of the time elapsed after exposure to a low dose of ROFA is wanted. Thus, we aimed to investigate the time course of mechanical, histological, and inflammatory lung changes, as well as neutrophils in the blood, in mice exposed to ROFA until 5 days after exposure. BALB/c mice (25 ± 5 g) were randomly divided into 7 groups and intranasally instilled with either 10 μL of sterile saline solution (0.9% NaCl, CTRL) or ROFA (0.2 μg in 10 μL of saline solution). Pulmonary mechanics, histology (normal and collapsed alveoli, mononuclear and polymorphonuclear cells, and ultrastructure), neutrophils (in blood and bronchoalveolar lavage fluid) were determined at 6 h in CTRL and at 6, 24, 48, 72, 96, and 120 h after ROFA exposure. ROFA contained metal elements, especially iron, polycyclic aromatic hydrocarbons (PAHs), and organochlorines. Lung resistive pressure augmented early (6 h) in the course of lung injury and other mechanical, histological and inflammatory parameters increased at 24 h, returning to control values at 120 h. Blood neutrophilia was present only at 24 and 48 h after exposure. Swelling of endothelial cells with adherent neutrophils was detected after ROFA instillation. No neutrophils were present in the lavage fluid. In conclusion, the exposure to ROFA, even in low doses, induced early changes in pulmonary mechanics, lung histology and accumulation of neutrophils in blood of mice that lasted for 4 days and disappeared spontaneously.
Collapse
Affiliation(s)
| | - Lilian Katiê da Silva Nagato
- Laboratory of Respiration Physiology, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| | - Sheila da Silva Fagundes
- Laboratory of Respiration Physiology, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| | - Flávia Brandão Dos Santos
- Laboratory of Respiration Physiology, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| | - Andrea Surrage Calheiros
- Laboratory of Immunopharmacology, Department of Physiology and Pharmacodynamics, Oswaldo Cruz Institute, Fundação Oswaldo Cruz Rio de Janeiro, Brazil
| | - Olaf Malm
- Laboratory of Radioisotopes, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| | - Patricia Torres Bozza
- Laboratory of Immunopharmacology, Department of Physiology and Pharmacodynamics, Oswaldo Cruz Institute, Fundação Oswaldo Cruz Rio de Janeiro, Brazil
| | - Paulo Hilário N Saldiva
- Laboratory of Experimental Air Pollution, Department of Pathology, School of Medicine, Universidade de São Paulo São Paulo, Brazil
| | - Débora Souza Faffe
- Laboratory of Macromolecular Metabolism Firmino Torres de Castro, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| | - Patricia Rieken Macedo Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| | - Walter Araujo Zin
- Laboratory of Respiration Physiology, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| |
Collapse
|
23
|
The regulation of 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone-induced lung tumor promotion by estradiol in female A/J mice. PLoS One 2014; 9:e93152. [PMID: 24682076 PMCID: PMC3969372 DOI: 10.1371/journal.pone.0093152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 03/02/2014] [Indexed: 11/25/2022] Open
Abstract
Epidemiological studies indicate that women are at a higher risk developing lung cancer than men are. It is suggested that estrogen is one of the most important factors in lung cancer development in females. Additionally, cigarette smoke, and environmental pollutants, such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), may play salient roles in female lung carcinogenesis. However, the mechanisms responsible for the interaction of these factors in the promotion of lung cancer are still poorly understood. The present study was designed to explore two ideas: first, the synergistic lung tumorigenic effects of 4-(methylnitrosamino)-1-(3-pyridyl)-butanol (NNK) combined with TCDD, 17β-estradiol (E2) or both through a long-term treatment experiment, and second, to identify early changes in the inflammatory and signaling pathways through short-term treatment experiments. The results indicate that A/J mice given E2 had strong effects in potentiating NNK-induced activation of MAPK signaling, NFκB, and COX-2 expression. In the long-term exposure model, E2 had a strong tumor promoting effect, whereas TCDD antagonized this effect in A/J mice. We conclude that treatment with NNK combined with either E2 or TCDD induces lung carcinogenesis and the promotion effects could be correlated with lung inflammation. E2 was shown to potentiate NNK-induced inflammation, cell proliferation, thereby leading to lung tumorigenesis.
Collapse
|
24
|
Mazzoli-Rocha F, Carvalho GMC, Lanzetti M, Valença SS, Silva LFF, Saldiva PHN, Zin WA, Faffe DS. Respiratory toxicity of repeated exposure to particles produced by traffic and sugar cane burning. Respir Physiol Neurobiol 2013; 191:106-13. [PMID: 24280381 DOI: 10.1016/j.resp.2013.11.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 11/18/2013] [Accepted: 11/19/2013] [Indexed: 11/25/2022]
Abstract
We compared the toxicity of subchronic exposure to equivalent masses of particles from sugar cane burning and traffic. BALB/c mice received 3 intranasal instillations/week during 1, 2 or 4 weeks of either distilled water (C1, C2, C4) or particles (15μg) from traffic (UP1, UP2, UP4) or biomass burning (BP1, BP2, BP4). Lung mechanics, histology and oxidative stress were analyzed 24h after the last instillation. In all instances UP and BP groups presented worse pulmonary elastance, airway and tissue resistance, alveolar collapse, bronchoconstriction and macrophage influx into the lungs than controls. UP4, BP2 and BP4 presented more alveolar collapse than UP1 and BP1, respectively. UP and BP had worse bronchial and alveolar lesion scores than their controls; BP4 had greater bronchial lesion scores than UP4. Catalase was higher in UP4 and BP4 than in C4. In conclusion, biomass particles were more toxic than those from traffic after repeated exposures.
Collapse
Affiliation(s)
- Flavia Mazzoli-Rocha
- Laboratory of Respiration Physiology, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, São Paulo, Brazil
| | - Giovanna M C Carvalho
- Laboratory of Respiration Physiology, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, São Paulo, Brazil
| | - Manuella Lanzetti
- Laboratory of Inflammation, Fundação Oswaldo Cruz, São Paulo, Brazil
| | - Samuel S Valença
- Laboratory of Inflammation, Institute of Biomedical Sciences, Universidade Federal do Rio de Janeiro, São Paulo, Brazil
| | - Luiz F F Silva
- Laboratory of Experimental Air Pollution, Department of Pathology, School of Medicine, Universidade de São Paulo, São Paulo, Brazil
| | - Paulo H N Saldiva
- Laboratory of Experimental Air Pollution, Department of Pathology, School of Medicine, Universidade de São Paulo, São Paulo, Brazil
| | - Walter A Zin
- Laboratory of Respiration Physiology, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, São Paulo, Brazil.
| | - Débora S Faffe
- Laboratory of Respiration Physiology, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, São Paulo, Brazil
| |
Collapse
|
25
|
Zou W, Zou Y, Zhao Z, Li B, Ran P. Nicotine-induced epithelial-mesenchymal transition via Wnt/β-catenin signaling in human airway epithelial cells. Am J Physiol Lung Cell Mol Physiol 2012. [PMID: 23204070 DOI: 10.1152/ajplung.00094.2012] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) has been proposed to be a mechanism in airway remodeling, which is a characteristic of chronic obstructive pulmonary disease (COPD). Studies have shown that cigarette smoke and nicotine are factors that induce Wnt/β-catenin activation, which is a pathway that has also been implicated in EMT. The main aim of this study was to test whether human bronchial epithelial cells are able to undergo EMT in vitro following nicotine stimulation via the Wnt3a/β-catenin signaling pathway. We show that nicotine activates the Wnt3a signal pathway, which leads to the translocation of β-catenin into the nucleus and activation of β-catenin/Tcf-dependent transcription in the human bronchial epithelial cell (HBEC) line. This accumulation was accompanied by an increase in smooth muscle actin, vimentin, matrix metalloproteinases-9, and type I collagen expression as well as downregulation of E-cadherin, which are typical characteristics of EMT. We also noted that the release of TGF-β(1) from these cells was stimulated by nicotine. Knockdown of Wnt3a with small interfering RNA (siRNA) prevented these effects, implying that β-catenin activation in these responses is Wnt3a dependent. Furthermore, specific knockdown of TGF-β(1) with TGF-β(1) siRNA partially prevented nicotine-induced EMT, suggesting that TGF-β(1) has a role in nicotine-mediated EMT in HBECs. These results suggest that HBECs are able to undergo EMT in vitro upon nicotine stimulation via the Wnt3a/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Weifeng Zou
- The State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | | | | | | | | |
Collapse
|
26
|
Xue H, Sun K, Xie W, Hu G, Kong H, Wang Q, Wang H. Etanercept attenuates short-term cigarette-smoke-exposure-induced pulmonary arterial remodelling in rats by suppressing the activation of TNF-a/NF-kB signal and the activities of MMP-2 and MMP-9. Pulm Pharmacol Ther 2012; 25:208-15. [PMID: 22724137 DOI: 10.1016/j.pupt.2012.02.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The pathogenesis of cigarette-smoke-exposure-induced pulmonary vasculature impairment is unclear. Cigarette-smoke-exposure-induced the accumulation of tumour necrosis factor-alpha (TNF-α) and upregulates the expression and activities of matrix metalloproteinases (MMPs) involved in smoke-induced vascular remodelling, which are important processes in the pathogenesis of vasculature impairment. The TNF-α antagonist Etanercept is an anti-inflammatory drug with a potential role in regulating MMP expression. To determine the effect of Etanercept on short-term smoke-induced pulmonary arteriole impairment and investigate its possible mechanism, male Sprague-Dawley rats were exposed to cigarette-smoke daily for two weeks in both the absence and presence of Etanercept. Cigarette-smoke-exposure-induced elevation of mean pulmonary artery pressures and medial hypertrophy of pulmonary arterioles were partially reduced by Etanercept. Up-regulation of the expression and activities of MMP-2 and MMP-9, induced by cigarette-smoke, were also suppressed significantly by Etanercept. Furthermore, Etanercept treatment significantly attenuated cigarette-smoke-induced TNFα accumulation and activation of nuclear factor NF-kB signal. These results suggest that Etanercept have the protective effects in cigarette-smoke-induced pulmonary vascular remodelling, with the attenuation of the up-regulated expression and activities of MMP-2 and MMP-9 and activation of TNF-α/NF-kB signal pathway probably being involved as part of its mechanism. Our study might provide insight into the development of new interventions for vasculature impairment.
Collapse
Affiliation(s)
- Hong Xue
- Department of respiratory medicine, The First Affiliated Hospital of Nanjing Medical University, and Department of Pharmacology and Neurobiology, Nanjing Medical University, Nanjing, Jiangsu, PR China.
| | | | | | | | | | | | | |
Collapse
|
27
|
Hizume DC, Toledo AC, Moriya HT, Saraiva-Romanholo BM, Almeida FM, Arantes-Costa FM, Vieira RP, Dolhnikoff M, Kasahara DI, Martins MA. Cigarette smoke dissociates inflammation and lung remodeling in OVA-sensitized and challenged mice. Respir Physiol Neurobiol 2012; 181:167-76. [PMID: 22446562 DOI: 10.1016/j.resp.2012.03.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Revised: 03/08/2012] [Accepted: 03/08/2012] [Indexed: 01/29/2023]
Abstract
We evaluated the effects of cigarette smoke (CS) on lung inflammation and remodeling in a model of ovalbumin (OVA)-sensitized and OVA-challenged mice. Male BALB/c mice were divided into 4 groups: non-sensitized and air-exposed (control); non-sensitized and exposed to cigarette smoke (CS), sensitized and air-exposed (OVA) (50 μg+OVA 1% 3 times/week for 3 weeks) and sensitized and cigarette smoke exposed mice (OVA+CS). IgE levels were not affected by CS exposure. The increases in total bronchoalveolar fluid cells in the OVA group were attenuated by co-exposure to CS, as were the changes in IL-4, IL-5, and eotaxin levels as well as tissue elastance (p<0.05). In contrast, only the OVA+CS group showed a significant increase in the protein expression of IFN-γ, VEGF, GM-CSF and collagen fiber content (p<0.05). In our study, exposure to cigarette smoke in OVA-challenged mice resulted in an attenuation of pulmonary inflammation but led to an increase in pulmonary remodeling and resulted in the dissociation of airway inflammation from lung remodeling.
Collapse
Affiliation(s)
- Deborah C Hizume
- Department of Medicine (LIM-20), School of Medicine, University of Sao Paulo, Sao Paulo, Brazil.
| | | | | | | | | | | | | | | | | | | |
Collapse
|