1
|
Burgess JK, Gosens R. Mechanotransduction and the extracellular matrix: Key drivers of lung pathologies and drug responsiveness. Biochem Pharmacol 2024; 228:116255. [PMID: 38705536 DOI: 10.1016/j.bcp.2024.116255] [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: 02/02/2024] [Revised: 04/19/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
The lung is a biomechanically active organ, with multiscale mechanical forces impacting the organ, tissue and cellular responses within this microenvironment. In chronic lung diseases, such as chronic obstructive pulmonary disease, pulmonary fibrosis and others, the structure of the lung is drastically altered impeding gas exchange. These changes are, in part, reflected in alterations in the composition, amount and organization of the extracellular matrix within the different lung compartments. The transmission of mechanical forces within lung tissue are broadcast by this complex mix of extracellular matrix components, in particular the collagens, elastin and proteoglycans and the crosslinking of these components. At both a macro and a micro level, the mechanical properties of the microenvironment have a key regulatory role in ascertaining cellular responses and the function of the lung. Cells adhere to, and receive signals from, the extracellular matrix through a number of different surface receptors and complexes which are important for mechanotransduction. This review summarizes the multiscale mechanics in the lung and how the mechanical environment changes in lung disease and aging. We then examine the role of mechanotransduction in driving cell signaling events in lung diseases and finish with a future perspective of the need to consider how such forces may impact pharmacological responsiveness in lung diseases.
Collapse
Affiliation(s)
- Janette K Burgess
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands.
| | - Reinoud Gosens
- University of Groningen, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, the Netherlands; Department of Molecular Pharmacology, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
2
|
Ashique S, Mishra N, Mantry S, Garg A, Kumar N, Gupta M, Kar SK, Islam A, Mohanto S, Subramaniyan V. Crosstalk between ROS-inflammatory gene expression axis in the progression of lung disorders. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03392-1. [PMID: 39196392 DOI: 10.1007/s00210-024-03392-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 08/16/2024] [Indexed: 08/29/2024]
Abstract
A significant number of deaths and disabilities worldwide are brought on by inflammatory lung diseases. Many inflammatory lung disorders, including chronic respiratory emphysema, resistant asthma, resistance to steroids, and coronavirus-infected lung infections, have severe variants for which there are no viable treatments; as a result, new treatment alternatives are needed. Here, we emphasize how oxidative imbalance contributes to the emergence of provocative lung problems that are challenging to treat. Endogenic antioxidant systems are not enough to avert free radical-mediated damage due to the induced overproduction of ROS. Pro-inflammatory mediators are then produced due to intracellular signaling events, which can harm the tissue and worsen the inflammatory response. Overproduction of ROS causes oxidative stress, which causes lung damage and various disease conditions. Invasive microorganisms or hazardous substances that are inhaled repeatedly can cause an excessive amount of ROS to be produced. By starting signal transduction pathways, increased ROS generation during inflammation may cause recurrent DNA damage and apoptosis and activate proto-oncogenes. This review provides information about new targets for conducting research in related domains or target factors to prevent, control, or treat such inflammatory oxidative stress-induced inflammatory lung disorders.
Collapse
Affiliation(s)
- Sumel Ashique
- Department of Pharmaceutics, Bengal College of Pharmaceutical Sciences & Research, Durgapur, West Bengal, 713212, India.
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India.
| | - Neeraj Mishra
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University Madhya Pradesh (AUMP), Gwalior, MP, 474005, India
| | - Shubhrajit Mantry
- Department of Pharmaceutics, Department of Pharmacy, Sarala Birla University, Ranchi, Jharkhand, 835103, India
| | - Ashish Garg
- Department of Pharmaceutics, Guru Ramdas Khalsa Institute of Science and Technology (Pharmacy), Jabalpur, Madhya Pradesh, 483001, India
| | - Nitish Kumar
- SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology (Deemed to Be University), Delhi-NCR Campus, Modinagar, Ghaziabad, Uttar Pradesh, 201204, India
| | - Madhu Gupta
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, Delhi, 110017, India
| | - Sanjeeb Kumar Kar
- Department of Pharmaceutical Chemistry, Department of Pharmacy, Sarala Birla University, Ranchi, Jharkhand, 835103, India
| | - Anas Islam
- Faculty of Pharmacy, Integral University, Lucknow, Uttar Pradesh, 226026, India
| | - Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to Be University), Mangalore, Karnataka, 575018, India.
| | - Vetriselvan Subramaniyan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Subang Jaya, Selangor, Malaysia.
| |
Collapse
|
3
|
Virdee S, Tan WC, Hogg JC, Bourbeau J, Hague CJ, Kirby M. CT Chest Imaging Using Normalized Join-Count: Predicting Emphysema Progression in the CanCOLD Study. Radiology 2024; 312:e233265. [PMID: 39012250 DOI: 10.1148/radiol.233265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
Background Pre-existing emphysema is recognized as an indicator of future worsening in patients with chronic obstructive pulmonary disease (COPD) when observed through CT imaging. However, it remains uncertain whether additional factors, such as the spatial compactness of CT emphysema, might also serve as predictors of disease progression. Purpose To evaluate the relationship between the compactness of CT emphysema voxels and emphysema progression. Materials and Methods This secondary analysis uses data from the prospective Canadian Cohort Obstructive Lung Disease (CanCOLD) study, examining CT images obtained in participants with and without COPD at baseline and a 3-year follow-up time point (November 2009 to November 2018). Measurements of forced expiratory volume in first second of expiration (FEV1) and diffusing capacity of lung for carbon monoxide (DLco) were collected. The normalized join-count (NJC) measurement from baseline CT images and lung density (LD) changes were analyzed. Emphysema progression was defined as an annualized LD change of less than half an SD below the mean of the participants without COPD with no smoking history. Multivariable linear and logistic regression models were used to assess the association between baseline CT NJC measurements and the annualized change in LD, FEV1, DLco, and emphysema progression versus nonprogression. Results A total of 524 participants (mean age, 66 years ± 10 [SD]; 293 male) (FEV1 percent predicted, 88% ± 19; FEV1/FVC, 67% ± 9; DLco percent predicted, 105% ± 25) were analyzed, 187 (36%) of whom had COPD. CT NJC was associated with the annualized change in LD (P < .001), FEV1 (P = .02), and DLco (P = .01). Additionally, CT NJC predicted emphysema progression versus nonprogression (odds ratio, 2.24; 95% CI: 1.37, 3.50; P < .001). Conclusion The spatial distribution, or "compactness," of CT emphysema voxels predicted emphysema progression in individuals with and without COPD. ClinicalTrials.gov Identifier: NCT00920348 © RSNA, 2024 Supplemental material is available for this article.
Collapse
Affiliation(s)
- Sukhraj Virdee
- From the Department of Physics, Toronto Metropolitan University, 350 Victoria St, Kerr Hall South Bldg, Rm KHS-344, Toronto, ON, Canada M5B 2K3 (S.V., M.K.); Center for Heart Lung Innovation, St Paul's Hospital, Vancouver, Canada (W.C.T., J.C.H., C.J.H., M.K.); and McGill University Health Centre, McGill University, Montreal, Canada (J.B.)
| | - Wan C Tan
- From the Department of Physics, Toronto Metropolitan University, 350 Victoria St, Kerr Hall South Bldg, Rm KHS-344, Toronto, ON, Canada M5B 2K3 (S.V., M.K.); Center for Heart Lung Innovation, St Paul's Hospital, Vancouver, Canada (W.C.T., J.C.H., C.J.H., M.K.); and McGill University Health Centre, McGill University, Montreal, Canada (J.B.)
| | - James C Hogg
- From the Department of Physics, Toronto Metropolitan University, 350 Victoria St, Kerr Hall South Bldg, Rm KHS-344, Toronto, ON, Canada M5B 2K3 (S.V., M.K.); Center for Heart Lung Innovation, St Paul's Hospital, Vancouver, Canada (W.C.T., J.C.H., C.J.H., M.K.); and McGill University Health Centre, McGill University, Montreal, Canada (J.B.)
| | - Jean Bourbeau
- From the Department of Physics, Toronto Metropolitan University, 350 Victoria St, Kerr Hall South Bldg, Rm KHS-344, Toronto, ON, Canada M5B 2K3 (S.V., M.K.); Center for Heart Lung Innovation, St Paul's Hospital, Vancouver, Canada (W.C.T., J.C.H., C.J.H., M.K.); and McGill University Health Centre, McGill University, Montreal, Canada (J.B.)
| | - Cameron J Hague
- From the Department of Physics, Toronto Metropolitan University, 350 Victoria St, Kerr Hall South Bldg, Rm KHS-344, Toronto, ON, Canada M5B 2K3 (S.V., M.K.); Center for Heart Lung Innovation, St Paul's Hospital, Vancouver, Canada (W.C.T., J.C.H., C.J.H., M.K.); and McGill University Health Centre, McGill University, Montreal, Canada (J.B.)
| | - Miranda Kirby
- From the Department of Physics, Toronto Metropolitan University, 350 Victoria St, Kerr Hall South Bldg, Rm KHS-344, Toronto, ON, Canada M5B 2K3 (S.V., M.K.); Center for Heart Lung Innovation, St Paul's Hospital, Vancouver, Canada (W.C.T., J.C.H., C.J.H., M.K.); and McGill University Health Centre, McGill University, Montreal, Canada (J.B.)
| |
Collapse
|
4
|
Villa B, Erranz B, Cruces P, Retamal J, Hurtado DE. Mechanical and morphological characterization of the emphysematous lung tissue. Acta Biomater 2024; 181:282-296. [PMID: 38705223 DOI: 10.1016/j.actbio.2024.04.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 04/22/2024] [Accepted: 04/25/2024] [Indexed: 05/07/2024]
Abstract
Irreversible alveolar airspace enlargement is the main characteristic of pulmonary emphysema, which has been extensively studied using animal models. While the alterations in lung mechanics associated with these morphological changes have been documented in the literature, the study of the mechanical behavior of parenchymal tissue from emphysematous lungs has been poorly investigated. In this work, we characterize the mechanical and morphological properties of lung tissue in elastase-induced emphysema rat models under varying severity conditions. We analyze the non-linear tissue behavior using suitable hyperelastic constitutive models that enable to compare different non-linear responses in terms of hyperelastic material parameters. We further analyze the effect of the elastase dose on alveolar morphology and tissue material parameters and study their connection with respiratory-system mechanical parameters. Our results show that while the lung mechanical function is not significantly influenced by the elastase treatment, the tissue mechanical behavior and alveolar morphology are markedly affected by it. We further show a strong association between alveolar enlargement and tissue softening, not evidenced by respiratory-system compliance. Our findings highlight the importance of understanding tissue mechanics in emphysematous lungs, as changes in tissue properties could detect the early stages of emphysema remodeling. STATEMENT OF SIGNIFICANCE: Gas exchange is vital for life and strongly relies on the mechanical function of the lungs. Pulmonary emphysema is a prevalent respiratory disease where alveolar walls are damaged, causing alveolar enlargement that induces harmful changes in the mechanical response of the lungs. In this work, we study how the mechanical properties of lung tissue change during emphysema. Our results from animal models show that tissue properties are more sensitive to alveolar enlargement due to emphysema than other mechanical properties that describe the function of the whole respiratory system.
Collapse
Affiliation(s)
- Benjamín Villa
- Department of Structural and Geotechnical Engineering, School of Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile, Vicuña Mackenna 4860, Santiago, Chile; Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Santiago, Chile
| | - Benjamín Erranz
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Santiago, Chile
| | - Pablo Cruces
- Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile. Avenida Repblica 440, Santiago, Chile
| | - Jaime Retamal
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile, Santiago, Chile
| | - Daniel E Hurtado
- Department of Structural and Geotechnical Engineering, School of Engineering, Pontificia Universidad Católica de Chile, Santiago, Chile, Vicuña Mackenna 4860, Santiago, Chile; Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Vicuña Mackenna 4860, Santiago, Chile; Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, 02140, USA.
| |
Collapse
|
5
|
Lee HW, Lee JK, Kim Y, Jang AS, Hwang YI, Lee JH, Jung KS, Yoo KH, Yoon HK, Kim DK. Differential decline of lung function in COPD patients according to structural abnormality in chest CT. Heliyon 2024; 10:e27683. [PMID: 38560191 PMCID: PMC10980934 DOI: 10.1016/j.heliyon.2024.e27683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 03/02/2024] [Accepted: 03/05/2024] [Indexed: 04/04/2024] Open
Abstract
Background Different progressions or prognoses of chronic obstructive pulmonary disease (COPD) have been reported according to structural abnormalities based on chest computed tomography (CT). This study aimed to investigate whether different structural abnormalities independently affect annual lung function changes and clinical prognosis in patients with COPD. Methods This longitudinal multicenter observational study was conducted using the KOCOSS cohort (NCT02800499) database in Korea from January 2012 to December 2019. For COPD patients with chest CT findings at baseline enrolment and longitudinal spirometric data, annual forced expiratory volume in 1 s (FEV1) decline rate (mL/year) and clinical outcomes were compared according to structural abnormalities, including emphysema, bronchiectasis (BE), and tuberculosis-destroyed lung (TDL). We estimated the adjusted annual FEV1 changes using a mixed-effect linear regression model. Results Among the enrolled 237 patients, 152 showed structural abnormalities. Emphysema, BE, and TDL were observed in 119 (78.3%), 28 (18.4%), and 27 (17.8%) patients, respectively. The annual decline in FEV1 was faster in COPD patients with structural abnormalities than those without (β = -70.6 mL/year, P-value = 0.039). BE/TDL-dominant or emphysema-dominant structural abnormality contributed to an accelerated annual FEV1 decline compared to no structural abnormality (BE/TDL-dominant, β = -103.7 mL/year, P-value = 0.043; emphysema-dominant, β = -84.1 mL/year, P-value = 0.018). Structural abnormalities made no significant differences in acute exacerbation rate and mortality. Conclusion The lung function decline rate in COPD differed according to structural abnormalities on CT. These findings may suggest that more focus should be placed on earlier intervention or regular follow-up with spirometry in COPD patients with BE or TDL on chest CT.
Collapse
Affiliation(s)
- Hyun Woo Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Jung-Kyu Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Youlim Kim
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Konkuk University Hospital, School of Medicine, Konkuk University, Seoul, South Korea
| | - An-Soo Jang
- Department of Pulmonology and Allergy, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, South Korea
| | - Yong il Hwang
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, South Korea
| | - Jae Ha Lee
- Division of Pulmonology, Department of Internal Medicine, Inje University Haeundae Paik Hospital, University of Inje College of Medicine, Busan, South Korea
| | - Ki-Suck Jung
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, South Korea
| | - Kwang Ha Yoo
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Konkuk University Hospital, School of Medicine, Konkuk University, Seoul, South Korea
| | - Hyoung Kyu Yoon
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Yeouido St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, South Korea
| | - Deog Kyeom Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
6
|
Negasi ZH, Nommi N, Liu C, Tesfaigzi Y. Persistence of emphysema following cessation of cigarette smoke exposure requires a susceptibility factor. Am J Physiol Lung Cell Mol Physiol 2024; 326:L431-L439. [PMID: 38349118 PMCID: PMC11281787 DOI: 10.1152/ajplung.00342.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 02/01/2024] [Accepted: 02/02/2024] [Indexed: 02/21/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is caused by cigarette smoke (CS) exposure but can often be progressive even in former smokers. Exposure of mice to CS for 22 wk causes emphysema, but whether emphysema persists after cessation of CS exposure is not clear. The purpose of this study was to determine whether emphysema persists in mice following a recovery period of 22 wk and whether a susceptibility factor, such as deficiency in the Bcl-2-interacting killer (Bik), is required for this persistence. Therefore, bik+/+ and bik-/- mice at 6-10 wk of age were exposed to 250 mg/m3 total particulate matter of CS or filtered air (FA) for 3 or 22 wk and were kept in FA for an additional 22 wk. Lungs were lavaged to quantify inflammatory cells, and sections were stained with hematoxylin and eosin to assess severity of emphysema. Exposure to CS for 3 wk increased the number of inflammatory cells in bik-/- mice compared with bik+/+ mice but not at 22 wk of exposure. At 22 wk of CS exposure, extent of emphysema was similar in bik+/+ and bik-/- mice. However, when mice were exposed to CS over the first 22 wk and were kept in FA for an additional 22 wk, emphysema remained similar in bik+/+ mice but was enhanced in bik-/- mice. These findings link increased inflammation with persistent emphysematous changes even after smoking cessation and demonstrate that a preexisting susceptibility condition is required to sustain enhanced emphysema that was initiated by long-term CS exposure.NEW & NOTEWORTHY Exposure of mice to cigarette smoke (CS) for 22 wk causes emphysema, but whether emphysema persists after an additional period of 6 mo after cessation of CS exposure has not been reported. In addition, the role of preexisting susceptibility in enhancing the persistence of CS-induced emphysema after exposure to CS has stopped has not been shown. The present study shows that a preexisting susceptibility must be present to enhance CS-induced emphysema after cessation of CS exposure.
Collapse
Affiliation(s)
- Zerihun Hailemariam Negasi
- Pulmonary Critical Care Medicine Division, Brigham and Women's Hospital and Harvard Medical School, Harvard University, Boston, Massachusetts, United States
| | - Naomi Nommi
- Pulmonary Critical Care Medicine Division, Brigham and Women's Hospital and Harvard Medical School, Harvard University, Boston, Massachusetts, United States
| | - Congjian Liu
- Pulmonary Critical Care Medicine Division, Brigham and Women's Hospital and Harvard Medical School, Harvard University, Boston, Massachusetts, United States
| | - Yohannes Tesfaigzi
- Pulmonary Critical Care Medicine Division, Brigham and Women's Hospital and Harvard Medical School, Harvard University, Boston, Massachusetts, United States
- Chronic Obstructive Pulmonary Disease Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico, United States
| |
Collapse
|
7
|
Liu H, Fan P, Jin F, Ren H, Xu F, Li J. Targeting biophysical microenvironment for improved treatment of chronic obstructive pulmonary disease. Trends Mol Med 2023; 29:926-938. [PMID: 37704492 DOI: 10.1016/j.molmed.2023.08.007] [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: 06/01/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/15/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is responsible for high disability rates, high death rates, and significant cost to health systems. Growing evidence in recent decades shows significant biophysical microenvironment changes in COPD, impacting lung tissues, cells, and treatment response. Furthermore, such biophysical changes have shown great potential as novel targets for improved therapeutic strategy of COPD, where both pharmacological and non-pharmacological therapies focusing on repairing the biophysical microenvironment of the lung have emerged. We present the first comprehensive review of four distinct biophysical hallmarks [i.e., extracellular matrix (ECM) microarchitecture, stiffness, fluid shear stress, and mechanical stretch] in COPD, the possible involvement of pathological changes, possible effects, and correlated in vitro models and sum up the emerging COPD treatments targeting these biophysical hallmarks.
Collapse
Affiliation(s)
- Han Liu
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province and Education Ministry of China, Zhengzhou, Henan 450046, China
| | - Pengbei Fan
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province and Education Ministry of China, Zhengzhou, Henan 450046, China
| | - Fanli Jin
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province and Education Ministry of China, Zhengzhou, Henan 450046, China
| | - Hui Ren
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, China; Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, China; MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Jiansheng Li
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China; Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province and Education Ministry of China, Zhengzhou, Henan 450046, China.
| |
Collapse
|
8
|
Joshi I, Devine AJ, Joshi R, Smith NJ, Varisco BM. A titratable murine model of progressive emphysema using tracheal porcine pancreatic elastase. Sci Rep 2023; 13:15259. [PMID: 37709810 PMCID: PMC10502133 DOI: 10.1038/s41598-023-41527-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/28/2023] [Indexed: 09/16/2023] Open
Abstract
Progressive emphysema often leads to end-stage lung disease. Most mouse models of emphysema are typically modest (i.e. cigarette smoke exposure), and changes over time are difficult to quantify. The tracheal porcine pancreatic elastase model (PPE) produces severe injury, but the literature is conflicted as to whether emphysema improves, is stable, or progresses over time. We hypothesized a threshold of injury below which repair would occur and above which emphysema would be stable or progress. We treated 8-week-old C57BL6 mixed sex mice with 0, 0.5, 2, or 4 activity units of PPE in 100 µL PBS and performed lung stereology at 21 and 84 days. There were no significant differences in weight gain or mouse health. Despite minimal emphysema at 21-days in the 0.5 units group (2.8 µm increased mean linear intercept, MLI), MLI increased by 4.6 µm between days 21 and 84 (p = 0.0007). In addition to larger MLI at 21 days in 2- and 4-unit groups, MLI increases from day 21 to 84 were 17.2 and 34 µm respectively (p = 0.002 and p = 0.0001). Total lung volume increased, and alveolar surface area decreased with time and injury severity. Contrary to our hypothesis, we found no evidence of alveolar repair over time. Airspace destruction was both progressive and accelerative. Future mechanistic studies in lung immunity, mechano-biology, senescence, and cell-specific changes may lead to novel therapies to slow or halt progressive emphysema in humans.
Collapse
Affiliation(s)
- Imani Joshi
- College of Arts and Sciences, Xavier University, Cincinnati, OH, USA
| | - Andrew J Devine
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Rashika Joshi
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Noah J Smith
- University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Brian M Varisco
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- University of Cincinnati College of Medicine, Cincinnati, OH, USA.
- University of Arkansas for Medical Sciences, 1 Children's Way Slot 663, Little Rock, AR, 72202, USA.
| |
Collapse
|
9
|
Silva LLSD, Barbosa JAS, João JMLG, Fukuzaki S, Camargo LDN, Dos Santos TM, Campos ECD, Costa AS, Saraiva-Romanholo BM, Bezerra SKM, Lopes FTQDS, Bonturi CR, Oliva MLV, Leick EA, Righetti RF, Tibério IDFLC. Effects of a Peptide Derived from the Primary Sequence of a Kallikrein Inhibitor Isolated from Bauhinia bauhinioides (pep-BbKI) in an Asthma-COPD Overlap (ACO) Model. Int J Mol Sci 2023; 24:11261. [PMID: 37511021 PMCID: PMC10379932 DOI: 10.3390/ijms241411261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023] Open
Abstract
(1) There are several patients with asthma-COPD overlap (ACO). A peptide derived from the primary sequence of a kallikrein inhibitor isolated from Bauhinia bauhinioides (pep-BbKI) has potent anti-inflammatory and antioxidant effects. Purpose: To investigate the effects of pep-BbKI treatment in an ACO model and compare them with those of corticosteroids. (2) BALB/c mice were divided into groups: SAL (saline), OVA (ovalbumin), ELA (elastase), ACO (ovalbumin + elastase), ACO-pep-BbKI (treated with inhibitor), ACO-DX (dexamethasone treatment), ACO-DX-pep-BbKI (both treatments), and SAL-pep-BbKI (saline group treated with inhibitor). We evaluated: hyperresponsiveness to methacholine, bronchoalveolar lavage fluid (BALF), exhaled nitric oxide (eNO), IL-1β, IL-4, IL-5, IL-6, IL-10, IL-13, IL-17, IFN-γ, TNF-α, MMP-9, MMP-12, TGF-β, collagen fibers, iNOS, eNO, linear mean intercept (Lm), and NF-κB in airways (AW) and alveolar septa (AS). (3) ACO-pep-BbKI reversed ACO alterations and was similar to SAL in all mechanical parameters, Lm, neutrophils, IL-5, IL-10, IL-17, IFN-γ, TNF-α, MMP-12 (AW), collagen fibers, iNOS (AW), and eNO (p > 0.05). ACO-DX reversed ACO alterations and was similar to SAL in all mechanical parameters, Lm, total cells and differentials, IL-1β(AS), IL-5 (AS), IL-6 (AS), IL-10 (AS), IL-13 (AS), IFN-γ, MMP-12 (AS), TGF-β (AS), collagen fibers (AW), iNOS, and eNO (p > 0.05). SAL was similar to SAL-pep-BbKI for all comparisons (p > 0.05). (4) Pep-BbKI was similar to dexamethasone in reducing the majority of alterations of this ACO model.
Collapse
Affiliation(s)
| | | | | | - Silvia Fukuzaki
- Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo 01246-903, Brazil
| | | | | | | | - Arthur Silva Costa
- Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Beatriz Mangueira Saraiva-Romanholo
- Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo 01246-903, Brazil
- Department of Medicine, University City of São Paulo, São Paulo 03071-000, Brazil
| | | | | | - Camila Ramalho Bonturi
- Departamento de Bioquímica, Universidade Federal de São Paulo (UNIFESP), São Paulo 04039-002, Brazil
| | - Maria Luiza Vilela Oliva
- Departamento de Bioquímica, Universidade Federal de São Paulo (UNIFESP), São Paulo 04039-002, Brazil
| | - Edna Aparecida Leick
- Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Renato Fraga Righetti
- Faculdade de Medicina FMUSP, Universidade de São Paulo, São Paulo 01246-903, Brazil
- Hospital Sírio-Libanês, São Paulo 01308-050, Brazil
| | | |
Collapse
|
10
|
Miserocchi G. Early Endothelial Signaling Transduction in Developing Lung Edema. Life (Basel) 2023; 13:1240. [PMID: 37374024 DOI: 10.3390/life13061240] [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: 04/17/2023] [Revised: 05/17/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
The lung promptly responds to edemagenic conditions through functional adaptations that contrast the increase in microvascular filtration. This review presents evidence for early signaling transduction by endothelial lung cells in two experimental animal models of edema, hypoxia exposure, and fluid overload (hydraulic edema). The potential role of specialized sites of the plasma membranes considered mobile signaling platforms, referred to as membrane rafts, that include caveolae and lipid rafts, is presented. The hypothesis is put forward that early changes in the lipid composition of the bilayer of the plasma membrane might trigger the signal transduction process when facing changes in the pericellular microenvironment caused by edema. Evidence is provided that for an increase in the extravascular lung water volume not exceeding 10%, changes in the composition of the plasma membrane of endothelial cells are evoked in response to mechanical stimuli from the interstitial compartment as well as chemical stimuli relating with changes in the concentration of the disassembled portions of structural macromolecules. In hypoxia, thinning of endothelial cells, a decrease in caveolae and AQP-1, and an increase in lipid rafts are observed. The interpretation of this response is that it favors oxygen diffusion and hinder trans-cellular water fluxes. In hydraulic edema, which generates greater capillary water leakages, an increase in cell volume and opposite changes in membrane rafts were observed; further, the remarkable increase in caveolae suggests a potential abluminal-luminal vesicular-dependent fluid reabsorption.
Collapse
Affiliation(s)
- Giuseppe Miserocchi
- Department of Medicine and Surgery, Università di Milano Bicocca, 20900 Monza, Italy
| |
Collapse
|
11
|
Miserocchi G. The impact of heterogeneity of the air-blood barrier on control of lung extravascular water and alveolar gas exchange. FRONTIERS IN NETWORK PHYSIOLOGY 2023; 3:1142245. [PMID: 37251706 PMCID: PMC10213913 DOI: 10.3389/fnetp.2023.1142245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 04/28/2023] [Indexed: 05/31/2023]
Abstract
The architecture of the air-blood barrier is effective in optimizing the gas exchange as long as it retains its specific feature of extreme thinness reflecting, in turn, a strict control on the extravascular water to be kept at minimum. Edemagenic conditions may perturb this equilibrium by increasing microvascular filtration; this characteristically occurs when cardiac output increases to balance the oxygen uptake with the oxygen requirement such as in exercise and hypoxia (either due to low ambient pressure or reflecting a pathological condition). In general, the lung is well equipped to counteract an increase in microvascular filtration rate. The loss of control on fluid balance is the consequence of disruption of the integrity of the macromolecular structure of lung tissue. This review, merging data from experimental approaches and evidence in humans, will explore how the heterogeneity in morphology, mechanical features and perfusion of the terminal respiratory units might impact on lung fluid balance and its control. Evidence is also provided that heterogeneities may be inborn and they could actually get worse as a consequence of a developing pathological process. Further, data are presented how in humans inter-individual heterogeneities in morphology of the terminal respiratory hinder the control of fluid balance and, in turn, hamper the efficiency of the oxygen diffusion-transport function.
Collapse
|
12
|
Mariano CA, Sattari S, Ramirez GO, Eskandari M. Effects of tissue degradation by collagenase and elastase on the biaxial mechanics of porcine airways. Respir Res 2023; 24:105. [PMID: 37031200 PMCID: PMC10082978 DOI: 10.1186/s12931-023-02376-8] [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: 12/24/2022] [Accepted: 02/22/2023] [Indexed: 04/10/2023] Open
Abstract
BACKGROUND Common respiratory illnesses, such as emphysema and chronic obstructive pulmonary disease, are characterized by connective tissue damage and remodeling. Two major fibers govern the mechanics of airway tissue: elastin enables stretch and permits airway recoil, while collagen prevents overextension with stiffer properties. Collagenase and elastase degradation treatments are common avenues for contrasting the role of collagen and elastin in healthy and diseased states; while previous lung studies of collagen and elastin have analyzed parenchymal strips in animal and human specimens, none have focused on the airways to date. METHODS Specimens were extracted from the proximal and distal airways, namely the trachea, large bronchi, and small bronchi to facilitate evaluations of material heterogeneity, and subjected to biaxial planar loading in the circumferential and axial directions to assess airway anisotropy. Next, samples were subjected to collagenase and elastase enzymatic treatment and tensile tests were repeated. Airway tissue mechanical properties pre- and post-treatment were comprehensively characterized via measures of initial and ultimate moduli, strain transitions, maximum stress, hysteresis, energy loss, and viscoelasticity to gain insights regarding the specialized role of individual connective tissue fibers and network interactions. RESULTS Enzymatic treatment demonstrated an increase in airway tissue compliance throughout loading and resulted in at least a 50% decrease in maximum stress overall. Strain transition values led to significant anisotropic manifestation post-treatment, where circumferential tissues transitioned at higher strains compared to axial counterparts. Hysteresis values and energy loss decreased after enzymatic treatment, where hysteresis reduced by almost half of the untreated value. Anisotropic ratios exhibited axially led stiffness at low strains which transitioned to circumferentially led stiffness when subjected to higher strains. Viscoelastic stress relaxation was found to be greater in the circumferential direction for bronchial airway regions compared to axial counterparts. CONCLUSION Targeted fiber treatment resulted in mechanical alterations across the loading range and interactions between elastin and collagen connective tissue networks was observed. Providing novel mechanical characterization of elastase and collagenase treated airways aids our understanding of individual and interconnected fiber roles, ultimately helping to establish a foundation for constructing constitutive models to represent various states and progressions of pulmonary disease.
Collapse
Affiliation(s)
- Crystal A Mariano
- Department of Mechanical Engineering, University of California at Riverside, Riverside, CA, USA
| | - Samaneh Sattari
- Department of Mechanical Engineering, University of California at Riverside, Riverside, CA, USA
| | - Gustavo O Ramirez
- Department of Mechanical Engineering, University of California at Riverside, Riverside, CA, USA
| | - Mona Eskandari
- Department of Mechanical Engineering, University of California at Riverside, Riverside, CA, USA.
- BREATHE Center, School of Medicine, University of California at Riverside, Riverside, CA, USA.
- Department of Bioengineering, University of California at Riverside, Riverside, CA, USA.
| |
Collapse
|
13
|
Draper M, Bester M, Van Rooy M, Oberholzer H. Adverse pulmonary effects after oral exposure to copper, manganese and mercury, alone and in mixtures, in a Spraque-Dawley rat model. Ultrastruct Pathol 2023; 47:146-159. [PMID: 36857290 DOI: 10.1080/01913123.2023.2184891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
The rise in respiratory disease has been attributed to an increase in environmental pollution. Heavy metals contribute to environmental contamination via air, water, soil and food. The effects of atmospheric exposure to heavy metals on pulmonary structure and function have been researched, but the effects through drinking water have been neglected. The aim of this study was to investigate the potential in vivo alterations in the pulmonary tissue of male Sprague-Dawley rats after a 28-day oral exposure to copper (Cu), manganese (Mn) and mercury (Hg), alone and in mixtures, at 100 times the World Health Organization's (WHO) safety limit for each heavy metal in drinking water. Forty-eight male Sprague-Dawley rats were randomly divided into eight groups (n = 6): control, Cu, Mn, Hg, Cu + Mn, Cu + Hg, Mn + Hg and Cu, Mn + Hg. The morphology of lung tissue and the bronchioles were evaluated using light- and transmission electron microscopy. For all exposed groups, morphological changes included thickened inter- and intra-alveolar spaces, stratified epithelium, disrupted smooth muscle and early fibrosis and desquamation of the epithelia of the bronchioles to varying degrees. In all exposed groups, ultrastructurally, an increase in disarranged collagen and elastin fibers, nuclear membrane detachment, chromatin condensation, indistinct nucleoli and an increase in collagen fiber disarrangement was observed. This study has identified that oral exposure to Cu, Mn and Hg and as part of mixtures caused pathogenesis due to inflammation, cellular damage and fibrosis with Mn + Hg being the most potent heavy metal group.
Collapse
Affiliation(s)
- M Draper
- Department of Anatomy, Faculty of Health Sciences, University of Pretoria, Arcadia, South Africa
| | - Mj Bester
- Department of Anatomy, Faculty of Health Sciences, University of Pretoria, Arcadia, South Africa
| | - M Van Rooy
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia, South Africa
| | - Hm Oberholzer
- Department of Anatomy, Faculty of Health Sciences, University of Pretoria, Arcadia, South Africa
| |
Collapse
|
14
|
Suki B, Bates JH, Bartolák-Suki E. Remodeling of the Aged and Emphysematous Lungs: Roles of Microenvironmental Cues. Compr Physiol 2022; 12:3559-3574. [PMID: 35766835 PMCID: PMC11470990 DOI: 10.1002/cphy.c210033] [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] [Indexed: 11/06/2022]
Abstract
Aging is a slow process that affects all organs, and the lung is no exception. At the alveolar level, aging increases the airspace size with thicker and stiffer septal walls and straighter and thickened collagen and elastic fibers. This creates a microenvironment that interferes with the ability of cells in the parenchyma to maintain normal homeostasis and respond to injury. These changes also make the lung more susceptible to disease such as emphysema. Emphysema is characterized by slow but progressive remodeling of the deep alveolar regions that leads to airspace enlargement and increased but disorganized elastin and collagen deposition. This remodeling has been attributed to ongoing inflammation that involves inflammatory cells and the cytokines they produce. Cellular senescence, another consequence of aging, weakens the ability of cells to properly respond to injury, something that also occurs in emphysema. These factors conspire to make alveolar walls more prone to mechanical failure, which can set emphysema in motion by driving inflammation through immune stimulation by protein fragments. Both aging and emphysema are influenced by microenvironmental conditions such as local inflammation, chemical makeup, tissue stiffness, and mechanical stresses. Although aging and emphysema are not equivalent, they have the potential to influence each other in synergistic ways; aging sets up the conditions for emphysema to develop, while emphysema may accelerate cellular senescence and thus aging itself. This article focuses on the similarities and differences between the remodeled microenvironment of the aging and emphysematous lung, with special emphasis on the alveolar septal wall. © 2022 American Physiological Society. Compr Physiol 12:3559-3574, 2022.
Collapse
Affiliation(s)
- Béla Suki
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | - Jason H.T. Bates
- Depatment of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont
| | | |
Collapse
|
15
|
Yuan Z, Herrmann J, Murthy S, Peters K, Gerard SE, Nia HT, Lutchen KR, Suki B. A Personalized Spring Network Representation of Emphysematous Lungs From CT Images. FRONTIERS IN NETWORK PHYSIOLOGY 2022; 2:828157. [PMID: 36926064 PMCID: PMC10013051 DOI: 10.3389/fnetp.2022.828157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/21/2022] [Indexed: 11/13/2022]
Abstract
Emphysema is a progressive disease characterized by irreversible tissue destruction and airspace enlargement, which manifest as low attenuation area (LAA) on CT images. Previous studies have shown that inflammation, protease imbalance, extracellular matrix remodeling and mechanical forces collectively influence the progression of emphysema. Elastic spring network models incorporating force-based mechanical failure have been applied to investigate the pathogenesis and progression of emphysema. However, these models were general without considering the patient-specific information on lung structure available in CT images. The aim of this work was to develop a novel approach that provides an optimal spring network representation of emphysematous lungs based on the apparent density in CT images, allowing the construction of personalized networks. The proposed method takes into account the size and curvature of LAA clusters on the CT images that correspond to a pre-stressed condition of the lung as opposed to a naïve method that excludes the effects of pre-stress. The main findings of this study are that networks constructed by the new method 1) better preserve LAA cluster sizes and their distribution than the naïve method; and 2) predict different course of emphysema progression compared to the naïve method. We conclude that our new method has the potential to predict patient-specific emphysema progression which needs verification using clinical data.
Collapse
Affiliation(s)
- Ziwen Yuan
- Department of Biomedical Engineering, Boston University, Boston, MA, United States
| | - Jacob Herrmann
- Department of Biomedical Engineering, Boston University, Boston, MA, United States
| | - Samhita Murthy
- Department of Biomedical Engineering, Boston University, Boston, MA, United States
| | - Kevin Peters
- Department of Biomedical Engineering, Boston University, Boston, MA, United States
| | - Sarah E. Gerard
- Department of Radiology, University of Iowa, Iowa City, IA, United States
| | - Hadi T. Nia
- Department of Biomedical Engineering, Boston University, Boston, MA, United States
| | - Kenneth R. Lutchen
- Department of Biomedical Engineering, Boston University, Boston, MA, United States
| | - Béla Suki
- Department of Biomedical Engineering, Boston University, Boston, MA, United States
| |
Collapse
|
16
|
Magnavacca A, Sangiovanni E, Racagni G, Dell'Agli M. The antiviral and immunomodulatory activities of propolis: An update and future perspectives for respiratory diseases. Med Res Rev 2022; 42:897-945. [PMID: 34725836 PMCID: PMC9298305 DOI: 10.1002/med.21866] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/20/2021] [Accepted: 10/20/2021] [Indexed: 12/11/2022]
Abstract
Propolis is a complex natural product that possesses antioxidant, anti-inflammatory, immunomodulatory, antibacterial, and antiviral properties mainly attributed to the high content in flavonoids, phenolic acids, and their derivatives. The chemical composition of propolis is multifarious, as it depends on the botanical sources from which honeybees collect resins and exudates. Nevertheless, despite this variability propolis may have a general pharmacological value, and this review systematically compiles, for the first time, the existing preclinical and clinical evidence of propolis activities as an antiviral and immunomodulatory agent, focusing on the possible application in respiratory diseases. In vitro and in vivo assays have demonstrated propolis broad-spectrum effects on viral infectivity and replication, as well as the modulatory actions on cytokine production and immune cell activation as part of both innate and adaptive immune responses. Clinical trials confirmed propolis undeniable potential as an effective therapeutic agent; however, the lack of rigorous randomized clinical trials in the context of respiratory diseases is tangible. Since propolis is available as a dietary supplement, possible use for the prevention of respiratory diseases and their deleterious inflammatory drawbacks on the respiratory tract in humans is considered and discussed. This review opens up new perspectives on the clinical investigation of neglected propolis biological properties which, now more than ever, are particularly relevant with respect to the recent outbreaks of pandemic respiratory infections.
Collapse
Affiliation(s)
- Andrea Magnavacca
- Department of Pharmacological and Biomolecular SciencesUniversity of MilanMilanItaly
| | - Enrico Sangiovanni
- Department of Pharmacological and Biomolecular SciencesUniversity of MilanMilanItaly
| | - Giorgio Racagni
- Department of Pharmacological and Biomolecular SciencesUniversity of MilanMilanItaly
| | - Mario Dell'Agli
- Department of Pharmacological and Biomolecular SciencesUniversity of MilanMilanItaly
| |
Collapse
|
17
|
Fu HT, Zhang Y, Zhang P, Wu H, Sun XQ, Shen SY, Dou DB. Tumor necrosis factor-α promotes airway mucus hypersecretion by repressing miR-146a-5p and miR-134-5p levels in human airway epithelial cells. Transl Cancer Res 2022; 10:4047-4056. [PMID: 35116702 PMCID: PMC8797934 DOI: 10.21037/tcr-20-3375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 07/14/2021] [Indexed: 12/12/2022]
Abstract
Background Airway mucus acts as an indispensable protective component of innate immune response against invading pathogens. However, airway mucus hypersecretion, largely consisting of mucin 5AC (MUC5AC), is the leading cause of airflow obstruction and airway hyperresponsiveness that contributes to chronic obstructive pulmonary disease (COPD). MicroRNAs (miRNAs) are frequently dysregulated in the pathogenesis of COPD, but the definite role of miRNAs in airway mucus hypersecretion is not well understood. Methods A cell model of mucus hypersecretion was established in 16HBE cells by treatment with TNF-α. Cell viability and apoptosis were assessed using cell counting kit-8 (CCK-8) and flow cytometry, respectively. The aberrant expression of miR-146a-5p and miR-134-5p was assayed in TNF-α-treated 16HBE cells, and the effect of miR-146a-5p and miR-134-5p on regulating MUC5AC expression was evaluated using quantitative real-time PCR (qPCR) and Western blot analysis. Results TNF-α treatment resulted in a significant decrease of cell viability, and increase of cell apoptosis and MUC5AC expression in 16HBE cells. Additionally, the expression of miR-134-5p and miR-146a-5p was markedly decreased in the cell model. Importantly, forced expression of miR-134-5p and miR-146a-5p significantly repressed TNF-α-induced upregulation of MUC5AC. Mechanistically, although miR-134-5p did not affect 16HBE cells viability and apoptosis, miR-134-5p partially blocked TNF-α-induced MUC5AC expression by inhibiting the activation of NF-κB signaling. On the other hand, miR-146a-5p enhanced cell viability and reduced cell apoptosis. miR-146a-5p also repressed TNF-α-induced MUC5AC expression by inhibiting p38 MAPK (mitogen-activated protein kinase) signaling activation. Conclusions The current data demonstrated that both miR-134-5p and miR-146a-5p conferred protection against TNF-α-induced mucus hypersecretion through repressing NF-κB and p38 MAPK signaling, indicating that miR-134-5p and miR-146a-5p may serve as the biomarker for COPD.
Collapse
Affiliation(s)
- Hui-Ting Fu
- Department of Traditional Medicine, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Zhang
- Department of Ultrasound, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ping Zhang
- Department of Traditional Medicine, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huan Wu
- Department of Traditional Medicine, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xuan-Qiu Sun
- Department of Traditional Medicine, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shu-Yang Shen
- Department of Traditional Medicine, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dan-Bo Dou
- Department of Traditional Medicine, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
18
|
Maghsoudi-Ganjeh M, Mariano CA, Sattari S, Arora H, Eskandari M. Developing a Lung Model in the Age of COVID-19: A Digital Image Correlation and Inverse Finite Element Analysis Framework. Front Bioeng Biotechnol 2021; 9:684778. [PMID: 34765590 PMCID: PMC8576180 DOI: 10.3389/fbioe.2021.684778] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 10/04/2021] [Indexed: 02/02/2023] Open
Abstract
Pulmonary diseases, driven by pollution, industrial farming, vaping, and the infamous COVID-19 pandemic, lead morbidity and mortality rates worldwide. Computational biomechanical models can enhance predictive capabilities to understand fundamental lung physiology; however, such investigations are hindered by the lung’s complex and hierarchical structure, and the lack of mechanical experiments linking the load-bearing organ-level response to local behaviors. In this study we address these impedances by introducing a novel reduced-order surface model of the lung, combining the response of the intricate bronchial network, parenchymal tissue, and visceral pleura. The inverse finite element analysis (IFEA) framework is developed using 3-D digital image correlation (DIC) from experimentally measured non-contact strains and displacements from an ex-vivo porcine lung specimen for the first time. A custom-designed inflation device is employed to uniquely correlate the multiscale classical pressure-volume bulk breathing measures to local-level deformation topologies and principal expansion directions. Optimal material parameters are found by minimizing the error between experimental and simulation-based lung surface displacement values, using both classes of gradient-based and gradient-free optimization algorithms and by developing an adjoint formulation for efficiency. The heterogeneous and anisotropic characteristics of pulmonary breathing are represented using various hyperelastic continuum formulations to divulge compound material parameters and evaluate the best performing model. While accounting for tissue anisotropy with fibers assumed along medial-lateral direction did not benefit model calibration, allowing for regional material heterogeneity enabled accurate reconstruction of lung deformations when compared to the homogeneous model. The proof-of-concept framework established here can be readily applied to investigate the impact of assorted organ-level ventilation strategies on local pulmonary force and strain distributions, and to further explore how diseased states may alter the load-bearing material behavior of the lung. In the age of a respiratory pandemic, advancing our understanding of lung biomechanics is more pressing than ever before.
Collapse
Affiliation(s)
- Mohammad Maghsoudi-Ganjeh
- Department of Mechanical Engineering, University of California, Riverside, Riverside, CA, United States
| | - Crystal A Mariano
- Department of Mechanical Engineering, University of California, Riverside, Riverside, CA, United States
| | - Samaneh Sattari
- Department of Mechanical Engineering, University of California, Riverside, Riverside, CA, United States
| | - Hari Arora
- Faculty of Science and Engineering, Swansea University, Swansea, United Kingdom
| | - Mona Eskandari
- Department of Mechanical Engineering, University of California, Riverside, Riverside, CA, United States.,BREATHE Center, School of Medicine, University of California, Riverside, Riverside, CA, United States.,Department of Bioengineering, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
19
|
Wubshet NH, Arreguin-Martinez E, Nail M, Annamalai H, Koerner R, Rousseva M, Tom T, Gillespie RB, Liu AP. Simulating microgravity using a random positioning machine for inducing cellular responses to mechanotransduction in human osteoblasts. THE REVIEW OF SCIENTIFIC INSTRUMENTS 2021; 92:114101. [PMID: 34852501 PMCID: PMC9643046 DOI: 10.1063/5.0056366] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 10/17/2021] [Indexed: 06/13/2023]
Abstract
The mechanotransduction pathways that mediate cellular responses to contact forces are better understood than those that mediate response to distance forces, especially the force of gravity. Removing or reducing gravity for significant periods of time involves either sending samples to space, inducing diamagnetic levitation with high magnetic fields, or continually reorienting samples for a period, all in a manner that supports cell culturing. Undesired secondary effects due to high magnetic fields or shear forces associated with fluid flow while reorienting must be considered in the design of ground-based devices. We have developed a lab-friendly and compact random positioning machine (RPM) that fits in a standard tissue culture incubator. Using a two-axis gimbal, it continually reorients samples in a manner that produces an equal likelihood that all possible orientations are visited. We contribute a new control algorithm by which the distribution of probabilities over all possible orientations is completely uniform. Rather than randomly varying gimbal axis speed and/or direction as in previous algorithms (which produces non-uniform probability distributions of orientation), we use inverse kinematics to follow a trajectory with a probability distribution of orientations that is uniform by construction. Over a time period of 6 h of operation using our RPM, the average gravity is within 0.001 23% of the gravity of Earth. Shear forces are minimized by limiting the angular speed of both gimbal motors to under 42 °/s. We demonstrate the utility of our RPM by investigating the effects of simulated microgravity on adherent human osteoblasts immediately after retrieving samples from our RPM. Cytoskeletal disruption and cell shape changes were observed relative to samples cultured in a 1 g environment. We also found that subjecting human osteoblasts in suspension to simulated microgravity resulted in less filamentous actin and lower cell stiffness.
Collapse
Affiliation(s)
- Nadab H. Wubshet
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | | - Hariprasad Annamalai
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Robert Koerner
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Maria Rousseva
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Tristan Tom
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | - Allen P. Liu
- Author to whom correspondence should be addressed: . Current address: University of Michigan, 2350 Hayward Street, Ann Arbor, Michigan 48109, USA. Tel.: +1 734-764-7719
| |
Collapse
|
20
|
Leslie MN, Chou J, Young PM, Traini D, Bradbury P, Ong HX. How Do Mechanics Guide Fibroblast Activity? Complex Disruptions during Emphysema Shape Cellular Responses and Limit Research. Bioengineering (Basel) 2021; 8:110. [PMID: 34436113 PMCID: PMC8389228 DOI: 10.3390/bioengineering8080110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/28/2021] [Accepted: 08/02/2021] [Indexed: 11/28/2022] Open
Abstract
The emphysema death toll has steadily risen over recent decades, causing the disease to become the third most common cause of death worldwide in 2019. Emphysema is currently incurable and could be due to a genetic condition (Alpha-1 antitrypsin deficiency) or exposure to pollutants/irritants, such as cigarette smoke or poorly ventilated cooking fires. Despite the growing burden of emphysema, the mechanisms behind emphysematous pathogenesis and progression are not fully understood by the scientific literature. A key aspect of emphysematous progression is the destruction of the lung parenchyma extracellular matrix (ECM), causing a drastic shift in the mechanical properties of the lung (known as mechanobiology). The mechanical properties of the lung such as the stiffness of the parenchyma (measured as the elastic modulus) and the stretch forces required for inhalation and exhalation are both reduced in emphysema. Fibroblasts function to maintain the structural and mechanical integrity of the lung parenchyma, yet, in the context of emphysema, these fibroblasts appear incapable of repairing the ECM, allowing emphysema to progress. This relationship between the disturbances in the mechanical cues experienced by an emphysematous lung and fibroblast behaviour is constantly overlooked and consequently understudied, thus warranting further research. Interestingly, the failure of current research models to integrate the altered mechanical environment of an emphysematous lung may be limiting our understanding of emphysematous pathogenesis and progression, potentially disrupting the development of novel treatments. This review will focus on the significance of emphysematous lung mechanobiology to fibroblast activity and current research limitations by examining: (1) the impact of mechanical cues on fibroblast activity and the cell cycle, (2) the potential role of mechanical cues in the diminished activity of emphysematous fibroblasts and, finally, (3) the limitations of current emphysematous lung research models and treatments as a result of the overlooked emphysematous mechanical environment.
Collapse
Affiliation(s)
- Mathew N. Leslie
- Respiratory Technology, The Woolcock Institute of Medical Research, Glebe, Sydney, NSW 2037, Australia; (M.N.L.); (P.M.Y.); (D.T.)
- Department of Biomedical Sciences, Faculty of Medicine, Healthy and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Joshua Chou
- Faculty of Engineering and Information Technology, University of Technology Sydney, Ultimo, Sydney, NSW 2007, Australia;
| | - Paul M. Young
- Respiratory Technology, The Woolcock Institute of Medical Research, Glebe, Sydney, NSW 2037, Australia; (M.N.L.); (P.M.Y.); (D.T.)
- Department of Marketing, Macquarie Business School, Macquarie University, Sydney, NSW 2109, Australia
| | - Daniela Traini
- Respiratory Technology, The Woolcock Institute of Medical Research, Glebe, Sydney, NSW 2037, Australia; (M.N.L.); (P.M.Y.); (D.T.)
- Department of Biomedical Sciences, Faculty of Medicine, Healthy and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Peta Bradbury
- Respiratory Technology, The Woolcock Institute of Medical Research, Glebe, Sydney, NSW 2037, Australia; (M.N.L.); (P.M.Y.); (D.T.)
- Faculty of Engineering and Information Technology, University of Technology Sydney, Ultimo, Sydney, NSW 2007, Australia;
- Mechanics and Genetics of Embryonic and Tumoral Development Group, UMR168—Laboratoire Physico-Chimie Curie, Institut Curie, 75248 Paris, France
| | - Hui Xin Ong
- Respiratory Technology, The Woolcock Institute of Medical Research, Glebe, Sydney, NSW 2037, Australia; (M.N.L.); (P.M.Y.); (D.T.)
- Department of Biomedical Sciences, Faculty of Medicine, Healthy and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia
| |
Collapse
|
21
|
Tanabe N, Kaji S, Sato S, Yokoyama T, Oguma T, Tanizawa K, Handa T, Sakajo T, Hirai T. A homological approach to a mathematical definition of pulmonary fibrosis and emphysema on computed tomography. J Appl Physiol (1985) 2021; 131:601-612. [PMID: 34138650 DOI: 10.1152/japplphysiol.00150.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Three-dimensional imaging is essential to evaluate local abnormalities and understand structure-function relationships in an organ. However, quantifiable and interpretable methods to localize abnormalities remain unestablished. Visual assessments are prone to bias, machine learning methods depend on training images, and the underlying decision principle is usually difficult to interpret. Here, we developed a homological approach to mathematically define emphysema and fibrosis in the lungs on computed tomography (CT). With the use of persistent homology, the density of homological features, including connected components, tunnels, and voids, was extracted from the volumetric CT scans of lung diseases. A pair of CT values at which each homological feature appeared (birth) and disappeared (death) was computed by sweeping the threshold levels from higher to lower CT values. Consequently, fibrosis and emphysema were defined as voxels with dense voids having a longer lifetime (birth-death difference) and voxels with dense connected components having a lower birth, respectively. In an independent dataset including subjects with idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD), and combined pulmonary fibrosis and emphysema (CPFE), the proposed definition enabled accurate segmentation with comparable quality to deep learning in terms of Dice coefficients. Persistent homology-defined fibrosis was closely associated with physiological abnormalities such as impaired diffusion capacity and long-term mortality in subjects with IPF and CPFE, and persistent homology-defined emphysema was associated with impaired diffusion capacity in subjects with COPD. The present persistent homology-based evaluation of structural abnormalities could help explore the clinical and physiological impacts of structural changes and morphological mechanisms of disease progression.NEW & NOTEWORTHY This study proposes a homological approach to mathematically define a three-dimensional texture feature of emphysema and fibrosis on chest computed tomography using persistent homology. The proposed definition enabled accurate segmentation with comparable quality to deep learning while offering higher interpretability than deep learning-based methods.
Collapse
Affiliation(s)
- Naoya Tanabe
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shizuo Kaji
- Institute of Mathematics for Industry, Kyushu University, Fukuoka, Japan
| | - Susumu Sato
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomoo Yokoyama
- Department of Mathematics, Kyoto University of Education, Kyoto, Japan
| | - Tsuyoshi Oguma
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kiminobu Tanizawa
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomohiro Handa
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Advanced Medicine for Respiratory Failure, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Sakajo
- Department of Mathematics, Kyoto University, Kyoto, Japan
| | - Toyohiro Hirai
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
22
|
Tanabe N, Sato S, Suki B, Hirai T. Fractal Analysis of Lung Structure in Chronic Obstructive Pulmonary Disease. Front Physiol 2021; 11:603197. [PMID: 33408642 PMCID: PMC7779609 DOI: 10.3389/fphys.2020.603197] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 11/25/2020] [Indexed: 12/12/2022] Open
Abstract
Chest CT is often used for localizing and quantitating pathologies associated with chronic obstructive pulmonary disease (COPD). While simple measurements of areas and volumes of emphysema and airway structure are common, these methods do not capture the structural complexity of the COPD lung. Since the concept of fractals has been successfully applied to evaluate complexity of the lung, this review is aimed at describing the fractal properties of airway disease, emphysema, and vascular abnormalities in COPD. An object forms a fractal if it exhibits the property of self-similarity at different length scales of evaluations. This fractal property is governed by power-law functions characterized by the fractal dimension (FD). Power-laws can also manifest in other statistical descriptors of structure such as the size distribution of emphysema clusters characterized by the power-law exponent D. Although D is not the same as FD of emphysematous clusters, it is a useful index to characterize the spatial pattern of disease progression and predict clinical outcomes in patients with COPD. The FD of the airway tree shape and the D of the size distribution of airway branches have been proposed indexes of structural assessment and clinical predictions. Simulations are also useful to understand the mechanism of disease progression. Therefore, the power-law and fractal analysis of the parenchyma and airways, especially when combined with computer simulations, could lead to a better understanding of the structural alterations during the progression of COPD and help identify subjects at a high risk of severe COPD.
Collapse
Affiliation(s)
- Naoya Tanabe
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Susumu Sato
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Béla Suki
- Department of Biomedical Engineering, Boston University, Boston, MA, United States
| | - Toyohiro Hirai
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
23
|
Mondoñedo JR, Bartolák-Suki E, Bou Jawde S, Nelson K, Cao K, Sonnenberg A, Obrochta WP, Imsirovic J, Ram-Mohan S, Krishnan R, Suki B. A High-Throughput System for Cyclic Stretching of Precision-Cut Lung Slices During Acute Cigarette Smoke Extract Exposure. Front Physiol 2020; 11:566. [PMID: 32655401 PMCID: PMC7326018 DOI: 10.3389/fphys.2020.00566] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 05/07/2020] [Indexed: 12/31/2022] Open
Abstract
Rationale Precision-cut lung slices (PCLSs) are a valuable tool in studying tissue responses to an acute exposure; however, cyclic stretching may be necessary to recapitulate physiologic, tidal breathing conditions. Objectives To develop a multi-well stretcher and characterize the PCLS response following acute exposure to cigarette smoke extract (CSE). Methods A 12-well stretching device was designed, built, and calibrated. PCLS were obtained from male Sprague-Dawley rats (N = 10) and assigned to one of three groups: 0% (unstretched), 5% peak-to-peak amplitude (low-stretch), and 5% peak-to-peak amplitude superimposed on 10% static stretch (high-stretch). Lung slices were cyclically stretched for 12 h with or without CSE in the media. Levels of Interleukin-1β (IL-1β), matrix metalloproteinase (MMP)-1 and its tissue inhibitor (TIMP1), and membrane type-MMP (MT1-MMP) were assessed via western blot from tissue homogenate. Results The stretcher system produced nearly identical normal Lagrangian strains (Exx and Eyy, p > 0.999) with negligible shear strain (Exy < 0.0005) and low intra-well variability 0.127 ± 0.073%. CSE dose response curve was well characterized by a four-parameter logistic model (R2 = 0.893), yielding an IC50 value of 0.018 cig/mL. Cyclic stretching for 12 h did not decrease PCLS viability. Two-way ANOVA detected a significant interaction between CSE and stretch pattern for IL-1β (p = 0.017), MMP-1, TIMP1, and MT1-MMP (p < 0.001). Conclusion This platform is capable of high-throughput testing of an acute exposure under tightly-regulated, cyclic stretching conditions. We conclude that the acute mechano-inflammatory response to CSE exhibits complex, stretch-dependence in the PCLS.
Collapse
Affiliation(s)
- Jarred R Mondoñedo
- Department of Biomedical Engineering, College of Engineering, Boston University, Boston, MA, United States.,Boston University School of Medicine, Boston, MA, United States
| | - Elizabeth Bartolák-Suki
- Department of Biomedical Engineering, College of Engineering, Boston University, Boston, MA, United States
| | - Samer Bou Jawde
- Department of Biomedical Engineering, College of Engineering, Boston University, Boston, MA, United States
| | - Kara Nelson
- Department of Biomedical Engineering, College of Engineering, Boston University, Boston, MA, United States
| | - Kun Cao
- Department of Biomedical Engineering, College of Engineering, Boston University, Boston, MA, United States
| | - Adam Sonnenberg
- Department of Systems Engineering, College of Engineering, Boston University, Boston, MA, United States
| | - Walter Patrick Obrochta
- Department of Biomedical Engineering, College of Engineering, Boston University, Boston, MA, United States
| | - Jasmin Imsirovic
- Department of Biomedical Engineering, College of Engineering, Boston University, Boston, MA, United States
| | - Sumati Ram-Mohan
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Ramaswamy Krishnan
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Béla Suki
- Department of Biomedical Engineering, College of Engineering, Boston University, Boston, MA, United States
| |
Collapse
|
24
|
Shimizu K, Tanabe N, Tho NV, Suzuki M, Makita H, Sato S, Muro S, Mishima M, Hirai T, Ogawa E, Nakano Y, Konno S, Nishimura M. Per cent low attenuation volume and fractal dimension of low attenuation clusters on CT predict different long-term outcomes in COPD. Thorax 2020; 75:116-122. [PMID: 31896733 DOI: 10.1136/thoraxjnl-2019-213525] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 10/31/2019] [Accepted: 11/17/2019] [Indexed: 11/03/2022]
Abstract
BACKGROUND Fractal dimension (D) characterises the size distribution of low attenuation clusters on CT and assesses the spatial heterogeneity of emphysema that per cent low attenuation volume (%LAV) cannot detect. This study tested the hypothesis that %LAV and D have different roles in predicting decline in FEV1, exacerbation and mortality in patients with COPD. METHODS Chest inspiratory CT scans in the baseline and longitudinal follow-up records for FEV1, exacerbation and mortality prospectively collected over 10 years in the Hokkaido COPD Cohort Study were examined (n=96). The associations between CT measures and long-term outcomes were replicated in the Kyoto University cohort (n=130). RESULTS In the Hokkaido COPD cohort, higher %LAV, but not D, was associated with a greater decline in FEV1 and 10-year mortality, whereas lower D, but not %LAV, was associated with shorter time to first exacerbation. Multivariable analysis for the Kyoto University cohort confirmed that lower D at baseline was independently associated with shorter time to first exacerbation and that higher LAV% was independently associated with increased mortality after adjusting for age, height, weight, FEV1 and smoking status. CONCLUSION These well-established cohorts clarify the different prognostic roles of %LAV and D, whereby lower D is associated with a higher risk of exacerbation and higher %LAV is associated with a rapid decline in lung function and long-term mortality. Combination of %LAV and fractal D may identify COPD subgroups at high risk of a poor clinical outcome more sensitively.
Collapse
Affiliation(s)
- Kaoruko Shimizu
- First Department of Medicine, Hokkaido University, School of Medicine, Sapporo, Japan
| | - Naoya Tanabe
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Nguyen Van Tho
- Division of Respiratory Medicine, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Masaru Suzuki
- First Department of Medicine, Hokkaido University, School of Medicine, Sapporo, Japan
| | - Hironi Makita
- First Department of Medicine, Hokkaido University, School of Medicine, Sapporo, Japan.,Hokkaido Institute of Respiratory Diseases, Sapporo, Japan
| | - Susumu Sato
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shigeo Muro
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Respiratory Medicine, Nara Medical University, Nara, Japan
| | - Michiaki Mishima
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Noe Hospital, Osaka, Japan
| | - Toyohiro Hirai
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Emiko Ogawa
- Division of Respiratory Medicine, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Yasutaka Nakano
- Division of Respiratory Medicine, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Satoshi Konno
- First Department of Medicine, Hokkaido University, School of Medicine, Sapporo, Japan
| | - Masaharu Nishimura
- First Department of Medicine, Hokkaido University, School of Medicine, Sapporo, Japan.,Hokkaido Institute of Respiratory Diseases, Sapporo, Japan
| |
Collapse
|
25
|
Burrowes KS, Iravani A, Kang W. Integrated lung tissue mechanics one piece at a time: Computational modeling across the scales of biology. Clin Biomech (Bristol, Avon) 2019; 66:20-31. [PMID: 29352607 DOI: 10.1016/j.clinbiomech.2018.01.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 12/05/2017] [Accepted: 01/09/2018] [Indexed: 02/07/2023]
Abstract
The lung is a delicately balanced and highly integrated mechanical system. Lung tissue is continuously exposed to the environment via the air we breathe, making it susceptible to damage. As a consequence, respiratory diseases present a huge burden on society and their prevalence continues to rise. Emergent function is produced not only by the sum of the function of its individual components but also by the complex feedback and interactions occurring across the biological scales - from genes to proteins, cells, tissue and whole organ - and back again. Computational modeling provides the necessary framework for pulling apart and putting back together the pieces of the body and organ systems so that we can fully understand how they function in both health and disease. In this review, we discuss models of lung tissue mechanics spanning from the protein level (the extracellular matrix) through to the level of cells, tissue and whole organ, many of which have been developed in isolation. This is a vital step in the process but to understand the emergent behavior of the lung, we must work towards integrating these component parts and accounting for feedback across the scales, such as mechanotransduction. These interactions will be key to unlocking the mechanisms occurring in disease and in seeking new pharmacological targets and improving personalized healthcare.
Collapse
Affiliation(s)
- Kelly S Burrowes
- Department of Chemical and Materials Engineering, University of Auckland, 2-6 Park Avenue, Auckland 1023, New Zealand; Auckland Bioengineering Institute, University of Auckland, 70 Symonds Street, Auckland 1010, New Zealand.
| | - Amin Iravani
- Department of Chemical and Materials Engineering, University of Auckland, 2-6 Park Avenue, Auckland 1023, New Zealand.
| | - Wendy Kang
- Auckland Bioengineering Institute, University of Auckland, 70 Symonds Street, Auckland 1010, New Zealand.
| |
Collapse
|
26
|
de Oliveira MV, Rocha NDN, Santos RS, Rocco MRM, de Magalhães RF, Silva JD, Souza SAL, Capelozzi VL, Pelosi P, Silva PL, Rocco PRM. Endotoxin-Induced Emphysema Exacerbation: A Novel Model of Chronic Obstructive Pulmonary Disease Exacerbations Causing Cardiopulmonary Impairment and Diaphragm Dysfunction. Front Physiol 2019; 10:664. [PMID: 31191356 PMCID: PMC6546905 DOI: 10.3389/fphys.2019.00664] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 05/09/2019] [Indexed: 12/26/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive disorder of the lung parenchyma which also involves extrapulmonary manifestations, such as cardiovascular impairment, diaphragm dysfunction, and frequent exacerbations. The development of animal models is important to elucidate the pathophysiology of COPD exacerbations and enable analysis of possible therapeutic approaches. We aimed to characterize a model of acute emphysema exacerbation and evaluate its consequences on the lung, heart, and diaphragm. Twenty-four Wistar rats were randomly assigned into one of two groups: control (C) or emphysema (ELA). In ELA group, animals received four intratracheal instillations of pancreatic porcine elastase (PPE) at 1-week intervals. The C group received saline under the same protocol. Five weeks after the last instillation, C and ELA animals received saline (SAL) or E. coli lipopolysaccharide (LPS) (200 μg in 200 μl) intratracheally. Twenty-four hours after saline or endotoxin administration, arterial blood gases, lung inflammation and morphometry, collagen fiber content, and lung mechanics were analyzed. Echocardiography, diaphragm ultrasonography (US), and computed tomography (CT) of the chest were done. ELA-LPS animals, compared to ELA-SAL, exhibited decreased arterial oxygenation; increases in alveolar collapse (p < 0.0001), relative neutrophil counts (p = 0.007), levels of cytokine-induced neutrophil chemoattractant-1, interleukin (IL)-1β, tumor necrosis factor-α, IL-6, and vascular endothelial growth factor in lung tissue, collagen fiber deposition in alveolar septa, airways, and pulmonary vessel walls, and dynamic lung elastance (p < 0.0001); reduced pulmonary acceleration time/ejection time ratio, (an indirect index of pulmonary arterial hypertension); decreased diaphragm thickening fraction and excursion; and areas of emphysema associated with heterogeneous alveolar opacities on chest CT. In conclusion, we developed a model of endotoxin-induced emphysema exacerbation that affected not only the lungs but also the heart and diaphragm, thus resembling several features of human disease. This model of emphysema should allow preclinical testing of novel therapies with potential for translation into clinical practice.
Collapse
Affiliation(s)
- Milena Vasconcellos de Oliveira
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Nazareth de Novaes Rocha
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Niterói, Brazil
| | - Raquel Souza Santos
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcella Rieken Macedo Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Raquel Ferreira de Magalhães
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Johnatas Dutra Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sergio Augusto Lopes Souza
- Department of Radiology, Faculty of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vera Luiza Capelozzi
- Department of Pathology, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Paolo Pelosi
- Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa, Genoa, Italy.,San Martino Policlinico Hospital, IRCCS for Oncology and Neurosciences, Genoa, Italy
| | - Pedro Leme Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia Rieken Macedo Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
27
|
Ito JT, Lourenço JD, Righetti RF, Tibério IFLC, Prado CM, Lopes FDTQS. Extracellular Matrix Component Remodeling in Respiratory Diseases: What Has Been Found in Clinical and Experimental Studies? Cells 2019; 8:E342. [PMID: 30979017 PMCID: PMC6523091 DOI: 10.3390/cells8040342] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/03/2019] [Accepted: 04/09/2019] [Indexed: 01/09/2023] Open
Abstract
Changes in extracellular matrix (ECM) components in the lungs are associated with the progression of respiratory diseases, such as asthma, chronic obstructive pulmonary disease (COPD), and acute respiratory distress syndrome (ARDS). Experimental and clinical studies have revealed that structural changes in ECM components occur under chronic inflammatory conditions, and these changes are associated with impaired lung function. In bronchial asthma, elastic and collagen fiber remodeling, mostly in the airway walls, is associated with an increase in mucus secretion, leading to airway hyperreactivity. In COPD, changes in collagen subtypes I and III and elastin, interfere with the mechanical properties of the lungs, and are believed to play a pivotal role in decreased lung elasticity, during emphysema progression. In ARDS, interstitial edema is often accompanied by excessive deposition of fibronectin and collagen subtypes I and III, which can lead to respiratory failure in the intensive care unit. This review uses experimental models and human studies to describe how inflammatory conditions and ECM remodeling contribute to the loss of lung function in these respiratory diseases.
Collapse
Affiliation(s)
- Juliana T Ito
- Department of Clinical Medicine, Laboratory of Experimental Therapeutics/LIM-20, School of Medicine of University of Sao Paulo, Sao Paulo 01246-903, Brazil.
| | - Juliana D Lourenço
- Department of Clinical Medicine, Laboratory of Experimental Therapeutics/LIM-20, School of Medicine of University of Sao Paulo, Sao Paulo 01246-903, Brazil.
| | - Renato F Righetti
- Department of Clinical Medicine, Laboratory of Experimental Therapeutics/LIM-20, School of Medicine of University of Sao Paulo, Sao Paulo 01246-903, Brazil.
- Rehabilitation service, Sírio-Libanês Hospital, Sao Paulo 01308-050, Brazil.
| | - Iolanda F L C Tibério
- Department of Clinical Medicine, Laboratory of Experimental Therapeutics/LIM-20, School of Medicine of University of Sao Paulo, Sao Paulo 01246-903, Brazil.
| | - Carla M Prado
- Department of Bioscience, Laboratory of Studies in Pulmonary Inflammation, Federal University of Sao Paulo, Santos 11015-020, Brazil.
| | - Fernanda D T Q S Lopes
- Department of Clinical Medicine, Laboratory of Experimental Therapeutics/LIM-20, School of Medicine of University of Sao Paulo, Sao Paulo 01246-903, Brazil.
| |
Collapse
|
28
|
Karimi A, Razaghi R. The role of smoking on the mechanical properties of the human lung. Technol Health Care 2018; 26:963-972. [DOI: 10.3233/thc-181340] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Alireza Karimi
- Department of Mechanical Engineering, Kyushu University, Fukuoka 819-0395, Japan
| | | |
Collapse
|
29
|
Aghasafari P, George U, Pidaparti R. A review of inflammatory mechanism in airway diseases. Inflamm Res 2018; 68:59-74. [PMID: 30306206 DOI: 10.1007/s00011-018-1191-2] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 09/12/2018] [Accepted: 09/27/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Inflammation in the lung is the body's natural response to injury. It acts to remove harmful stimuli such as pathogens, irritants, and damaged cells and initiate the healing process. Acute and chronic pulmonary inflammation are seen in different respiratory diseases such as; acute respiratory distress syndrome, chronic obstructive pulmonary disease (COPD), asthma, and cystic fibrosis (CF). FINDINGS In this review, we found that inflammatory response in COPD is determined by the activation of epithelial cells and macrophages in the respiratory tract. Epithelial cells and macrophages discharge transforming growth factor-β (TGF-β), which trigger fibroblast proliferation and tissue remodeling. Asthma leads to airway hyper-responsiveness, obstruction, mucus hyper-production, and airway-wall remodeling. Cytokines, allergens, chemokines, and infectious agents are the main stimuli that activate signaling pathways in epithelial cells in asthma. Mutation of the CF transmembrane conductance regulator (CFTR) gene results in CF. Mutations in CFTR influence the lung epithelial innate immune function that leads to exaggerated and ineffective airway inflammation that fails to abolish pulmonary pathogens. We present mechanistic computational models (based on ordinary differential equations, partial differential equations and agent-based models) that have been applied in studying the complex physiological and pathological mechanisms of chronic inflammation in different airway diseases. CONCLUSION The scope of the present review is to explore the inflammatory mechanism in airway diseases and highlight the influence of aging on airways' inflammation mechanism. The main goal of this review is to encourage research collaborations between experimentalist and modelers to promote our understanding of the physiological and pathological mechanisms that control inflammation in different airway diseases.
Collapse
Affiliation(s)
| | - Uduak George
- College of Engineering, University of Georgia, Athens, GA, USA.,Department of Mathematics and Statistics, San Diego State University, San Diego, CA, USA
| | | |
Collapse
|
30
|
CT Imaging-Based Low-Attenuation Super Clusters in Three Dimensions and the Progression of Emphysema. Chest 2018; 155:79-87. [PMID: 30292758 DOI: 10.1016/j.chest.2018.09.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 08/31/2018] [Accepted: 09/06/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Distributions of low-attenuation areas in two-dimensional (2-D) CT lung slices are used to quantify parenchymal destruction in patients with COPD. However, these segmental approaches are limited and may not reflect the true three-dimensional (3-D) tissue processes that drive emphysematous changes in the lung. The goal of this study was to instead evaluate distributions of 3-D low-attenuation volumes, which we hypothesized would follow a power law distribution and provide a more complete assessment of the mechanisms underlying disease progression. METHODS CT scans and pulmonary function test results were acquired from an observational database for N = 12 patients with COPD and N = 12 control patients. The data set included baseline and two annual follow-up evaluations in patients with COPD. Three-dimensional representations of the lungs were reconstructed from 2-D axial CT slices, with low-attenuation volumes identified as contiguous voxels < -960 Hounsfield units. RESULTS Low-attenuation sizes generally followed a power law distribution, with the exception of large, individual outliers termed "super clusters," which deviated from the expected distribution. Super cluster volume was correlated with disease severity (% total low attenuation, ρ = 0.950) and clinical measures of lung function including FEV1 (ρ = -0.849) and diffusing capacity of the lung for carbon monoxide Dlco (ρ = -0.874). To interpret these results, we developed a personalized computational model of super cluster emergence. Simulations indicated disease progression was more likely to occur near existing emphysematous regions, giving rise to a biomechanical, force-induced mechanism of super cluster growth. CONCLUSIONS Low-attenuation super clusters are defining, quantitative features of parenchymal destruction that dominate disease progression, particularly in advanced COPD.
Collapse
|
31
|
Tanabe N, Muro S, Sato S, Oguma T, Sato A, Hirai T. Fractal analysis of low attenuation clusters on computed tomography in chronic obstructive pulmonary disease. BMC Pulm Med 2018; 18:144. [PMID: 30157833 PMCID: PMC6116481 DOI: 10.1186/s12890-018-0714-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/24/2018] [Indexed: 01/20/2023] Open
Abstract
Background The fractal dimension characterizing the cumulative size distribution of low attenuation area (LAA) clusters, identified with a fixed threshold such as − 950 Hounsfield Units (HU), on computed tomography (CT) sensitively detects parenchymal destruction in chronic obstructive pulmonary disease (COPD) even when the percent LAA (LAA%), a standard emphysema index, is unchanged. This study examines whether the cumulative size distribution of LAA clusters, defined with thresholds of the 15th, 25th, and 35th percentiles of a CT density histogram instead of the fixed-threshold of − 950 HU, exhibits a fractal property and whether its fractal dimension (D’15, D’25, and D’35, respectively) provides additional structural information in emphysematous lungs that is difficult to detect with the conventional − 950-HU-based fractal dimension (D950). Methods Chest inspiratory CT scans and pulmonary functions were cross-sectionally examined in 170 COPD subjects. A proxy for the inspiration level at CT scan was obtained by dividing CT-measured total lung volume (CT-TLV) by physiologically measured total lung capacity. Moreover, long-term (> 5 years) changes in D950 and the new fractal dimensions were longitudinally evaluated in 17 current and 42 former smokers with COPD. Results D950, but not D’15, D’25, or D’35 was weakly correlated with the proxy for the inspiration. D950, D’25, and D’35 but not D’15 correlated with LAA% and diffusion capacity. In the long-term longitudinal study, LAA% was increased and D950 and D’35 were decreased in both current and former smokers, while D’25 was decreased only in current smokers and D’15 was not changed in either group. The longitudinal changes in D’25 but not those in LAA%, D950, D’15, and D’35 were greater in current smokers than in former smokers. This greater change in D’25 in current smokers was confirmed after adjusting the change in CT-TLV and the baseline D’25. Conclusions D’25 reflects diffusion capacity in emphysematous lungs and is robust against inspiration levels during CT scans. This new fractal dimension might provide additional structural information that is difficult to detect with the conventional D950 and LAA% and allow for more sensitive evaluation of emphysema progression over time. Electronic supplementary material The online version of this article (10.1186/s12890-018-0714-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Naoya Tanabe
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Shigeo Muro
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Susumu Sato
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Tsuyoshi Oguma
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Atsuyasu Sato
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Toyohiro Hirai
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|
32
|
Ceresa M, Olivares AL, Fernandez Suelves S, Noailly J, Gonzalez Ballester MA. Multi-scale immunological and biomechanical model of emphysema progression. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2018; 2017:2712-2715. [PMID: 29060459 DOI: 10.1109/embc.2017.8037417] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
This work presents a multi-scale agent-based model of emphysema progression that includes both the slow action of the immune system and the fast action of force redistribution and fracture propagation of the biological tissue. The two scales are coupled because the immune response causes inflammation and adaptation, which affects the biomechanical parameters of the tissue such as his elasticity. During repeated inflammation and breathing cycles, the tissue weakens and breaks down. We found that macrophages lifespan and cytokynes diffusion ratio are the parameters that influence the outcome of the model the most.
Collapse
|
33
|
Ceresa M, Olivares AL, Noailly J, González Ballester MA. Coupled Immunological and Biomechanical Model of Emphysema Progression. Front Physiol 2018; 9:388. [PMID: 29725304 PMCID: PMC5917021 DOI: 10.3389/fphys.2018.00388] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/28/2018] [Indexed: 12/16/2022] Open
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a disabling respiratory pathology, with a high prevalence and a significant economic and social cost. It is characterized by different clinical phenotypes with different risk profiles. Detecting the correct phenotype, especially for the emphysema subtype, and predicting the risk of major exacerbations are key elements in order to deliver more effective treatments. However, emphysema onset and progression are influenced by a complex interaction between the immune system and the mechanical properties of biological tissue. The former causes chronic inflammation and tissue remodeling. The latter influences the effective resistance or appropriate mechanical response of the lung tissue to repeated breathing cycles. In this work we present a multi-scale model of both aspects, coupling Finite Element (FE) and Agent Based (AB) techniques that we would like to use to predict the onset and progression of emphysema in patients. The AB part is based on existing biological models of inflammation and immunological response as a set of coupled non-linear differential equations. The FE part simulates the biomechanical effects of repeated strain on the biological tissue. We devise a strategy to couple the discrete biological model at the molecular /cellular level and the biomechanical finite element simulations at the tissue level. We tested our implementation on a public emphysema image database and found that it can indeed simulate the evolution of clinical image biomarkers during disease progression.
Collapse
Affiliation(s)
- Mario Ceresa
- BCN-Medtech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Barcelona, Spain
| | - Andy L Olivares
- BCN-Medtech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Barcelona, Spain
| | - Jérôme Noailly
- BCN-Medtech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Barcelona, Spain
| | - Miguel A González Ballester
- BCN-Medtech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Barcelona, Spain.,ICREA, Barcelona, Spain
| |
Collapse
|
34
|
Abstract
The structure and function of the lung gradually becomes compromised during the progression of emphysema. In this chapter, we first describe how to assess and evaluate lung function using the forced oscillation technique. Next, we provide details on how to use the Flexivent system to measure respiratory mechanical parameters in mice. We also describe the outlines of how to set up a homemade forced oscillatory system and use it to measure respiratory mechanics. To characterize the structure from standard histological images, we describe a method that is highly sensitive to early emphysema. Correlating structural information such as equivalent alveolar diameter and its variance with respiratory elastance or compliance, provides structure-function relationships that can subsequently reveal novel mechanisms of emphysema progression or be used to track the effectiveness of treatment.
Collapse
|
35
|
Yu X, Akbarzadeh R, Pieper M, Scholzen T, Gehrig S, Schultz C, Zillikens D, König P, Petersen F. Neutrophil Adhesion Is a Prerequisite for Antibody-Mediated Proteolytic Tissue Damage in Experimental Models of Epidermolysis Bullosa Acquisita. J Invest Dermatol 2018; 138:1990-1998. [PMID: 29559343 DOI: 10.1016/j.jid.2018.03.1499] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 02/26/2018] [Accepted: 03/09/2018] [Indexed: 12/17/2022]
Abstract
Although uncontrolled proteolytic activity mediated by activated neutrophils is a major reason for tissue damage, therapeutic approaches using protease inhibitors are inefficient. Here, we investigated the role of the immune complex-induced neutrophil adhesion and protease release in tissue damage. We show both in vitro and in vivo that immune complex-mediated neutrophil adhesion to the target tissue depends on β2 integrins. Without affecting elastase or reactive oxygen species release, blocking of adhesion drastically inhibited tissue damage in an experimental model of autoantibody-mediated skin blistering disease. By using a cell-bound fluorescent resonance energy transfer-based elastase sensor, we detected elastase enzyme activity on the surface of adherent cells resistant to protease inhibitors. Inhibitor resistance was lost by CD18 blockade or deficiency in vitro and in vivo. Immune complex-induced neutrophil adhesion created an enclosed protected space between the cell and its target structure where proteinases and reactive oxygen species can execute their tissue-damaging effect. Because immune complex-induced neutrophil adhesion represents an indispensable step for tissue damage of many diseases, our findings may facilitate the development of strategies for the treatment of such disorders.
Collapse
Affiliation(s)
- Xinhua Yu
- Priority Area Asthma and Allergy, Research Center Borstel, Borstel, Germany; Airway Research Center North, German Center for Lung Research (DZL); Xiamen-Borstel Joint Laboratory of Autoimmunity, The Medical College of Xiamen University, Xiamen University, Xiamen, China
| | - Reza Akbarzadeh
- Priority Area Asthma and Allergy, Research Center Borstel, Borstel, Germany; Airway Research Center North, German Center for Lung Research (DZL); Institute of Anatomy, University of Lübeck, Lübeck, Germany
| | - Mario Pieper
- Airway Research Center North, German Center for Lung Research (DZL); Institute of Anatomy, University of Lübeck, Lübeck, Germany
| | - Thomas Scholzen
- Priority Area Asthma and Allergy, Research Center Borstel, Borstel, Germany
| | - Stefanie Gehrig
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany; Translational Lung Research Center Heidelberg, German Center for Lung Research (DZL)
| | - Carsten Schultz
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany; Translational Lung Research Center Heidelberg, German Center for Lung Research (DZL)
| | - Detlef Zillikens
- Department of Dermatology, University of Lübeck, Lübeck, Germany
| | - Peter König
- Airway Research Center North, German Center for Lung Research (DZL); Institute of Anatomy, University of Lübeck, Lübeck, Germany
| | - Frank Petersen
- Priority Area Asthma and Allergy, Research Center Borstel, Borstel, Germany; Airway Research Center North, German Center for Lung Research (DZL).
| |
Collapse
|
36
|
Bodduluri S, Reinhardt JM, Hoffman EA, Newell JD, Nath H, Dransfield MT, Bhatt SP. Signs of Gas Trapping in Normal Lung Density Regions in Smokers. Am J Respir Crit Care Med 2017; 196:1404-1410. [PMID: 28707983 DOI: 10.1164/rccm.201705-0855oc] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
RATIONALE A substantial proportion of subjects without overt airflow obstruction have significant respiratory morbidity and structural abnormalities as visualized by computed tomography. Whether regions of the lung that appear normal using traditional computed tomography criteria have mild disease is not known. OBJECTIVES To identify subthreshold structural disease in normal-appearing lung regions in smokers. METHODS We analyzed 8,034 subjects with complete inspiratory and expiratory computed tomographic data participating in the COPDGene Study, including 103 lifetime nonsmokers. The ratio of the mean lung density at end expiration (E) to end inspiration (I) was calculated in lung regions with normal density (ND) by traditional thresholds for mild emphysema (-910 Hounsfield units) and gas trapping (-856 Hounsfield units) to derive the ND-E/I ratio. Multivariable regression analysis was used to measure the associations between ND-E/I, lung function, and respiratory morbidity. MEASUREMENTS AND MAIN RESULTS The ND-E/I ratio was greater in smokers than in nonsmokers, and it progressively increased from mild to severe chronic obstructive pulmonary disease severity. A proportion of 26.3% of smokers without airflow obstruction had ND-E/I greater than the 90th percentile of normal. ND-E/I was independently associated with FEV1 (adjusted β = -0.020; 95% confidence interval [CI], -0.032 to -0.007; P = 0.001), St. George's Respiratory Questionnaire scores (adjusted β = 0.952; 95% CI, 0.529 to 1.374; P < 0.001), 6-minute-walk distance (adjusted β = -10.412; 95% CI, -12.267 to -8.556; P < 0.001), and body mass index, airflow obstruction, dyspnea, and exercise capacity index (adjusted β = 0.169; 95% CI, 0.148 to 0.190; P < 0.001), and also with FEV1 change at follow-up (adjusted β = -3.013; 95% CI, -4.478 to -1.548; P = 0.001). CONCLUSIONS Subthreshold gas trapping representing mild small airway disease is prevalent in normal-appearing lung regions in smokers without airflow obstruction, and it is associated with respiratory morbidity. Clinical trial registered with www.clinicaltrials.gov (NCT00608764).
Collapse
Affiliation(s)
- Sandeep Bodduluri
- 1 Division of Pulmonary, Allergy and Critical Care Medicine.,2 UAB Lung Imaging Core.,3 UAB Lung Health Center, and
| | - Joseph M Reinhardt
- 4 Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa; and
| | - Eric A Hoffman
- 4 Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa; and.,5 Department of Radiology, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - John D Newell
- 4 Department of Biomedical Engineering, University of Iowa, Iowa City, Iowa; and.,5 Department of Radiology, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Hrudaya Nath
- 2 UAB Lung Imaging Core.,6 Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Mark T Dransfield
- 1 Division of Pulmonary, Allergy and Critical Care Medicine.,2 UAB Lung Imaging Core.,3 UAB Lung Health Center, and
| | - Surya P Bhatt
- 1 Division of Pulmonary, Allergy and Critical Care Medicine.,2 UAB Lung Imaging Core.,3 UAB Lung Health Center, and
| | | |
Collapse
|
37
|
Tobino K, Hirai T, Johkoh T, Fujimoto K, Kawaguchi A, Tomiyama N, Takahashi K, Seyama K. Difference of the progression of pulmonary cysts assessed by computed tomography among COPD, lymphangioleiomyomatosis, and Birt-Hogg-Dubé syndrome. PLoS One 2017; 12:e0188771. [PMID: 29220357 PMCID: PMC5722335 DOI: 10.1371/journal.pone.0188771] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 11/13/2017] [Indexed: 11/19/2022] Open
Abstract
Many groups developed the methods to quantitatively analyze low attenuation area (LAA) on chest CT in patients with cystic lung diseases. Especially in COPD, it was reported that the cumulative size distribution of LAA clusters follows a power law characterized by the exponent D, which reflect the fractal dimension of terminal airspace geometry. We hypoyhesized that the quantitative charateristics of LAA clusters including fractal property might indicate the different features of the progression of cysts in cystic lung diseases. The aim of this study was to apply the CT image-based method of characterizing the size distribution of LAA clusters for lymphangioleiomyomatosis (LAM) and Birt-Hogg-Dubé syndrome (BHDS) to disclose their features of the progression of pulmonary cysts. 40 patients with COPD, 52 patients with LAM, and 18 patients with BHDS who had undergone CT scans at our institute between January 2002 and August 2009 were included. Differences among these diseases in the quantitative characteristics of LAA clusters {i.e., extent, number, size, fractal property, and the relationship between these quantitatives} were assessed. The Chi-sqsuare test, unpaired t-test, and one-way analyses of variance with Tukey post-hoc tests were used to compare groups, spline model with an interaction terms were used to assess the relationship between extent and number, and exponential regression model was used to assess the relationship between extent and size. Statistically significant differences separated the three diseases in extent and number (P < 0.001). Number was significantly correlated with extent in COPD (P < 0.001), but was not so in LAM and BHDS when extent exceeded 11.5% and 20.8%, respectively. Size was significantly correlated with extent in COPD and LAM (P < 0.001), but was not so in BHDS. The percentage of CT images with fractal property was higher in COPD than that in LAM and BHDS (95.8%, 92.9% and 63.0%, respectively). In conclusion, our study has demonstrated for the first time the different characteristics of the size distribution of LAA clusters among COPD, LAM and BHDS, and indicated that this method is useful for exploration of the pathophysiology in cystic lung diseases.
Collapse
Affiliation(s)
- Kazunori Tobino
- Department of Respiratory Medicine, Iizuka Hospital, Iizuka, Fukuoka, Japan
- Divisions of Respiratory Medicine, Juntendo University Faculty of Medicine & Graduate School of Medicine, Bunkyo-Ku, Tokyo, Japan
- The Study Group of Pneumothorax and Cystic Lung Diseases, Setagaya-Ku, Tokyo, Japan
- * E-mail:
| | - Toyohiro Hirai
- Department of Respiratory Medicine, Kyoto University, Graduate School of Medicine, Sakyo-Ku, Kyoto, Japan
| | - Takeshi Johkoh
- Department of Radiology, Kinki Central Hospital of Mutual Aid Association of Public School Teachers, Itami, Hyogo, Japan
| | - Kiminori Fujimoto
- Department of Radiology, Kurume University School of Medicine and Center for Diagnostic Imaging, Kurume University Hospital, Kurume, Fukuoka, Japan
| | - Atsushi Kawaguchi
- Center for Comprehensive Community Medicine Faculty of Medicine, Saga University, Saga, Saga, Japan
| | - Noriyuki Tomiyama
- Department of Radiology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kazuhisa Takahashi
- Divisions of Respiratory Medicine, Juntendo University Faculty of Medicine & Graduate School of Medicine, Bunkyo-Ku, Tokyo, Japan
| | - Kuniaki Seyama
- Divisions of Respiratory Medicine, Juntendo University Faculty of Medicine & Graduate School of Medicine, Bunkyo-Ku, Tokyo, Japan
- The Study Group of Pneumothorax and Cystic Lung Diseases, Setagaya-Ku, Tokyo, Japan
| |
Collapse
|
38
|
Rocha NDN, de Oliveira MV, Braga CL, Guimarães G, Maia LDA, Padilha GDA, Silva JD, Takiya CM, Capelozzi VL, Silva PL, Rocco PRM. Ghrelin therapy improves lung and cardiovascular function in experimental emphysema. Respir Res 2017; 18:185. [PMID: 29100513 PMCID: PMC5670513 DOI: 10.1186/s12931-017-0668-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 10/30/2017] [Indexed: 12/21/2022] Open
Abstract
Background Emphysema is a progressive disease characterized by irreversible airspace enlargement followed by a decline in lung function. It also causes extrapulmonary effects, such as loss of body mass and cor pulmonale, which are associated with shorter survival and worse clinical outcomes. Ghrelin, a growth-hormone secretagogue, stimulates muscle anabolism, has anti-inflammatory effects, promotes vasodilation, and improves cardiac performance. Therefore, we hypothesized that ghrelin might reduce lung inflammation and remodelling as well as improve lung mechanics and cardiac function in experimental emphysema. Methods Forty female C57BL/6 mice were randomly assigned into two main groups: control (C) and emphysema (ELA). In the ELA group (n=20), animals received four intratracheal instillations of pancreatic porcine elastase (PPE) at 1-week intervals. C animals (n=20) received saline alone (50 μL) using the same protocol. Two weeks after the last instillation of saline or PPE, C and ELA animals received ghrelin or saline (n=10/group) intraperitoneally (i.p.) daily, during 3 weeks. Dual-energy X-ray absorptiometry (DEXA), echocardiography, lung mechanics, histology, and molecular biology were analysed. Results In elastase-induced emphysema, ghrelin treatment decreased alveolar hyperinflation and mean linear intercept, neutrophil infiltration, and collagen fibre content in the alveolar septa and pulmonary vessel wall; increased elastic fibre content; reduced M1-macrophage populations and increased M2 polarization; decreased levels of keratinocyte-derived chemokine (KC, a mouse analogue of interleukin-8), tumour necrosis factor-α, and transforming growth factor-β, but increased interleukin-10 in lung tissue; augmented static lung elastance; reduced arterial pulmonary hypertension and right ventricular hypertrophy on echocardiography; and increased lean mass. Conclusion In the elastase-induced emphysema model used herein, ghrelin not only reduced lung damage but also improved cardiac function and increased lean mass. These findings should prompt further studies to evaluate ghrelin as a potential therapy for emphysema. Electronic supplementary material The online version of this article (10.1186/s12931-017-0668-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nazareth de Novaes Rocha
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,Department of Physiology and Pharmacology, Biomedical Institute, Fluminense Federal University, Rio de Janeiro, Brazil
| | - Milena Vasconcellos de Oliveira
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Cássia Lisboa Braga
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Gabriela Guimarães
- Laboratory of Immunopathology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lígia de Albuquerque Maia
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Gisele de Araújo Padilha
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Johnatas Dutra Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Christina Maeda Takiya
- Laboratory of Immunopathology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vera Luiza Capelozzi
- Department of Pathology, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Pedro Leme Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Patricia Rieken Macedo Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Centro de Ciências da Saúde, Avenida Carlos Chagas Filho, s/n, Bloco G-014, Ilha do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil. .,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil.
| |
Collapse
|
39
|
Abstract
Lung densitometry assesses with computed tomography (CT) the X-ray attenuation of the pulmonary tissue which reflects both the degree of inflation and the structural lung abnormalities implying decreased attenuation, as in emphysema and cystic diseases, or increased attenuation, as in fibrosis. Five reasons justify replacement with lung densitometry of semi-quantitative visual scales used to measure extent and severity of diffuse lung diseases: (I) improved reproducibility; (II) complete vs. discrete assessment of the lung tissue; (III) shorter computation times; (IV) better correlation with pathology quantification of pulmonary emphysema; (V) better or equal correlation with pulmonary function tests (PFT). Commercially and open platform software are available for lung densitometry. It requires attention to technical and methodological issues including CT scanner calibration, radiation dose, and selection of thickness and filter to be applied to sections reconstructed from whole-lung CT acquisition. Critical is also the lung volume reached by the subject at scanning that can be measured in post-processing and represent valuable information per se. The measurements of lung density include mean and standard deviation, relative area (RA) at -970, -960 or -950 Hounsfield units (HU) and 1st and 15th percentile for emphysema in inspiratory scans, and RA at -856 HU for air trapping in expiratory scans. Kurtosis and skewness are used for evaluating pulmonary fibrosis in inspiratory scans. The main indication for lung densitometry is assessment of emphysema component in the single patient with chronic obstructive pulmonary diseases (COPD). Additional emerging applications include the evaluation of air trapping in COPD patients and in subjects at risk of emphysema and the staging in patients with lymphangioleiomyomatosis (LAM) and with pulmonary fibrosis. It has also been applied to assess prevalence of smoking-related emphysema and to monitor progression of smoking-related emphysema, alpha1 antitrypsin deficiency emphysema, and pulmonary fibrosis. Finally, it is recommended as end-point in pharmacological trials of emphysema and lung fibrosis.
Collapse
Affiliation(s)
- Mario Mascalchi
- "Mario Serio" Department of Experimental and Clinical Biomedical Sciences
| | - Gianna Camiciottoli
- "Mario Serio" Department of Experimental and Clinical Biomedical Sciences.,Section of Respiratory Medicine, Careggi University Hospital, Florence, Italy
| | - Stefano Diciotti
- Department of Electrical, Electronic, and Information Engineering "Guglielmo Marconi", University of Bologna, Cesena, Italy
| |
Collapse
|
40
|
van Agteren JEM, Hnin K, Grosser D, Carson KV, Smith BJ. Bronchoscopic lung volume reduction procedures for chronic obstructive pulmonary disease. Cochrane Database Syst Rev 2017; 2:CD012158. [PMID: 28230230 PMCID: PMC6464526 DOI: 10.1002/14651858.cd012158.pub2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND In the recent years, a variety of bronchoscopic lung volume reduction (BLVR) procedures have emerged that may provide a treatment option to participants suffering from moderate to severe chronic obstructive pulmonary disease (COPD). OBJECTIVES To assess the effects of BLVR on the short- and long-term health outcomes in participants with moderate to severe COPD and determine the effectiveness and cost-effectiveness of each individual technique. SEARCH METHODS Studies were identified from the Cochrane Airways Group Specialised Register (CAGR) and by handsearching of respiratory journals and meeting abstracts. All searches are current until 07 December 2016. SELECTION CRITERIA We included randomized controlled trials (RCTs). We included studies reported as full text, those published as abstract only and unpublished data, if available. DATA COLLECTION AND ANALYSIS Two independent review authors assessed studies for inclusion and extracted data. Where possible, data from more than one study were combined in a meta-analysis using RevMan 5 software. MAIN RESULTS AeriSealOne RCT of 95 participants found that AeriSeal compared to control led to a significant median improvement in forced expiratory volume in one second (FEV1) (18.9%, interquartile range (IQR) -0.7% to 41.9% versus 1.3%, IQR -8.2% to 12.9%), and higher quality of life, as measured by the St Georges Respiratory Questionnaire (SGRQ) (-12 units, IQR -22 units to -5 units, versus -3 units, IQR -5 units to 1 units), P = 0.043 and P = 0.0072 respectively. Although there was no significant difference in mortality (Odds Ratio (OR) 2.90, 95% CI 0.14 to 62.15), adverse events were more common for participants treated with AeriSeal (OR 3.71, 95% CI 1.34 to 10.24). The quality of evidence found in this prematurely terminated study was rated low to moderate. Airway bypass stentsTreatment with airway bypass stents compared to control did not lead to significant between-group changes in FEV1 (0.95%, 95% CI -0.16% to 2.06%) or SGRQ scores (-2.00 units, 95% CI -5.58 units to 1.58 units), as found by one study comprising 315 participants. There was no significant difference in mortality (OR 0.76, 95% CI 0.21 to 2.77), nor were there significant differences in adverse events (OR 1.33, 95% CI 0.65 to 2.73) between the two groups. The quality of evidence was rated moderate to high. Endobronchial coilsThree studies comprising 461 participants showed that treatment with endobronchial coils compared to control led to a significant between-group mean difference in FEV1 (10.88%, 95% CI 5.20% to 16.55%) and SGRQ (-9.14 units, 95% CI -11.59 units to -6.70 units). There were no significant differences in mortality (OR 1.49, 95% CI 0.67 to 3.29), but adverse events were significantly more common for participants treated with coils (OR 2.14, 95% CI 1.41 to 3.23). The quality of evidence ranged from low to high. Endobronchial valvesFive studies comprising 703 participants found that endobronchial valves versus control led to significant improvements in FEV1 (standardized mean difference (SMD) 0.48, 95% CI 0.32 to 0.64) and scores on the SGRQ (-7.29 units, 95% CI -11.12 units to -3.45 units). There were no significant differences in mortality between the two groups (OR 1.07, 95% CI 0.47 to 2.43) but adverse events were more common in the endobronchial valve group (OR 5.85, 95% CI 2.16 to 15.84). Participant selection plays an important role as absence of collateral ventilation was associated with superior clinically significant improvements in health outcomes. The quality of evidence ranged from low to high. Intrabronchial valvesIn the comparison of partial bilateral placement of intrabronchial valves to control, one trial favoured control in FEV1 (-2.11% versus 0.04%, P = 0.001) and one trial found no difference between the groups (0.9 L versus 0.87 L, P = 0.065). There were no significant differences in SGRQ scores (MD 2.64 units, 95% CI -0.28 units to 5.56 units) or mortality rates (OR 4.95, 95% CI 0.85 to 28.94), but adverse events were more frequent (OR 3.41, 95% CI 1.48 to 7.84) in participants treated with intrabronchial valves. The lack of functional benefits may be explained by the procedural strategy used, as another study (22 participants) compared unilateral versus partial bilateral placement, finding significant improvements in FEV1 and SGRQ when using the unilateral approach. The quality of evidence ranged between moderate to high. Vapour ablationOne study of 69 participants found significant mean between-group differences in FEV1 (14.70%, 95% CI 7.98% to 21.42%) and SGRQ (-9.70 units, 95% CI -15.62 units to -3.78 units), favouring vapour ablation over control. There was no significant between-group difference in mortality (OR 2.82, 95% CI 0.13 to 61.06), but vapour ablation led to significantly more adverse events (OR 3.86, 95% CI 1.00 to 14.97). The quality of evidence ranged from low to moderate. AUTHORS' CONCLUSIONS Results for selected BLVR procedures indicate they can provide significant and clinically meaningful short-term (up to one year) improvements in health outcomes, but this was at the expense of increased adverse events. The currently available evidence is not sufficient to assess the effect of BLVR procedures on mortality. These findings are limited by the lack of long-term follow-up data, limited availability of cost-effectiveness data, significant heterogeneity in results, presence of skew and high CIs, and the open-label character of a number of the studies.
Collapse
Affiliation(s)
| | - Khin Hnin
- Flinders UniversityAdelaideAustralia
| | | | | | - Brian J Smith
- The University of AdelaideSchool of MedicineAdelaideAustralia
| |
Collapse
|
41
|
Takayanagi S, Kawata N, Tada Y, Ikari J, Matsuura Y, Matsuoka S, Matsushita S, Yanagawa N, Kasahara Y, Tatsumi K. Longitudinal changes in structural abnormalities using MDCT in COPD: do the CT measurements of airway wall thickness and small pulmonary vessels change in parallel with emphysematous progression? Int J Chron Obstruct Pulmon Dis 2017; 12:551-560. [PMID: 28243075 PMCID: PMC5315203 DOI: 10.2147/copd.s121405] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Recent advances in multidetector computed tomography (MDCT) facilitate acquiring important clinical information for managing patients with COPD. MDCT can detect the loss of lung tissue associated with emphysema as a low-attenuation area (LAA) and the thickness of airways as the wall area percentage (WA%). The percentage of small pulmonary vessels <5 mm2 (% cross-sectional area [CSA] <5) has been recently recognized as a parameter for expressing pulmonary perfusion. We aimed to analyze the longitudinal changes in structural abnormalities using these CT parameters and analyze the effect of exacerbation and smoking cessation on structural changes in COPD patients. Methods We performed pulmonary function tests (PFTs), an MDCT, and a COPD assessment test (CAT) in 58 patients with COPD at the time of their enrollment at the hospital and 2 years later. We analyzed the change in clinical parameters including CT indices and examined the effect of exacerbations and smoking cessation on the structural changes. Results The CAT score and forced expiratory volume in 1 second (FEV1) did not significantly change during the follow-up period. The parameters of emphysematous changes significantly increased. On the other hand, the WA% at the distal airways significantly decreased or tended to decrease, and the %CSA <5 slightly but significantly increased over the same period, especially in ex-smokers. The parameters of emphysematous change were greater in patients with exacerbations and continued to progress even after smoking cessation. In contrast, the WA% and %CSA <5 did not change in proportion to emphysema progression. Conclusion The WA% at the distal bronchi and the %CSA <5 did not change in parallel with parameters of LAA over the same period. We propose that airway disease and vascular remodeling may be reversible to some extent by smoking cessation and appropriate treatment. Optimal management may have a greater effect on pulmonary vascularity and airway disease than parenchymal deconstruction in the early stage of COPD.
Collapse
Affiliation(s)
- Shin Takayanagi
- Department of Respirology, Graduate School of Medicine, Chiba University, Inohana, Chuo-ku, Chiba-shi, Chiba
| | - Naoko Kawata
- Department of Respirology, Graduate School of Medicine, Chiba University, Inohana, Chuo-ku, Chiba-shi, Chiba
| | - Yuji Tada
- Department of Respirology, Graduate School of Medicine, Chiba University, Inohana, Chuo-ku, Chiba-shi, Chiba
| | - Jun Ikari
- Department of Respirology, Graduate School of Medicine, Chiba University, Inohana, Chuo-ku, Chiba-shi, Chiba
| | - Yukiko Matsuura
- Department of Respirology, Graduate School of Medicine, Chiba University, Inohana, Chuo-ku, Chiba-shi, Chiba
| | - Shin Matsuoka
- Department of Radiology, St Marianna University School of Medicine, Sugao, Miyamae-ku, Kawasaki-shi, Kanagawa, Japan
| | - Shoichiro Matsushita
- Department of Radiology, St Marianna University School of Medicine, Sugao, Miyamae-ku, Kawasaki-shi, Kanagawa, Japan
| | - Noriyuki Yanagawa
- Department of Respirology, Graduate School of Medicine, Chiba University, Inohana, Chuo-ku, Chiba-shi, Chiba
| | - Yasunori Kasahara
- Department of Respirology, Graduate School of Medicine, Chiba University, Inohana, Chuo-ku, Chiba-shi, Chiba
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of Medicine, Chiba University, Inohana, Chuo-ku, Chiba-shi, Chiba
| |
Collapse
|
42
|
Mondoñedo JR, Suki B. Predicting Structure-Function Relations and Survival following Surgical and Bronchoscopic Lung Volume Reduction Treatment of Emphysema. PLoS Comput Biol 2017; 13:e1005282. [PMID: 28182686 PMCID: PMC5300131 DOI: 10.1371/journal.pcbi.1005282] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 12/06/2016] [Indexed: 11/26/2022] Open
Abstract
Lung volume reduction surgery (LVRS) and bronchoscopic lung volume reduction (bLVR) are palliative treatments aimed at reducing hyperinflation in advanced emphysema. Previous work has evaluated functional improvements and survival advantage for these techniques, although their effects on the micromechanical environment in the lung have yet to be determined. Here, we introduce a computational model to simulate a force-based destruction of elastic networks representing emphysema progression, which we use to track the response to lung volume reduction via LVRS and bLVR. We find that (1) LVRS efficacy can be predicted based on pre-surgical network structure; (2) macroscopic functional improvements following bLVR are related to microscopic changes in mechanical force heterogeneity; and (3) both techniques improve aspects of survival and quality of life influenced by lung compliance, albeit while accelerating disease progression. Our model predictions yield unique insights into the microscopic origins underlying emphysema progression before and after lung volume reduction. Surgical and, more recently, bronchoscopic lung volume reduction is the only available treatments for patients with advanced stage emphysema. Several large-scale, clinical studies have outlined appropriate selection criteria based on patient outcomes; however, the underlying mechanisms determining disease progression and response to these treatments are not well-understood. To answer this question, we have developed a network model of the lung to compare immediate and long-term response to each treatment. This approach allows us to directly study macroscopic changes in function related to microscopic changes in the local structural and mechanical environment. In addition, it facilitates direct comparisons between surgical and bronchoscopic lung volume reduction given identical initial conditions, which is not feasible in a clinical study. We propose here a mechanism suggesting that lung volume reduction efficacy is intimately linked to changes in microscopic force heterogeneity within the lung. Such an understanding of the mechanisms driving emphysema has the potential to greatly improve current therapies for this condition through more rationalized, patient-specific treatment strategies.
Collapse
Affiliation(s)
- Jarred R. Mondoñedo
- Department of Biomedical Engineering, Boston University, Boston, MA, United States of America
- School of Medicine, Boston University, Boston, MA, United States of America
| | - Béla Suki
- Department of Biomedical Engineering, Boston University, Boston, MA, United States of America
- * E-mail:
| |
Collapse
|
43
|
Diciotti S, Nobis A, Ciulli S, Landini N, Mascalchi M, Sverzellati N, Innocenti B. Development of digital phantoms based on a finite element model to simulate low-attenuation areas in CT imaging for pulmonary emphysema quantification. Int J Comput Assist Radiol Surg 2016; 12:1561-1570. [PMID: 27838881 DOI: 10.1007/s11548-016-1500-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 10/31/2016] [Indexed: 11/28/2022]
Abstract
PURPOSE To develop an innovative finite element (FE) model of lung parenchyma which simulates pulmonary emphysema on CT imaging. The model is aimed to generate a set of digital phantoms of low-attenuation areas (LAA) images with different grades of emphysema severity. METHODS Four individual parameter configurations simulating different grades of emphysema severity were utilized to generate 40 FE models using ten randomizations for each setting. We compared two measures of emphysema severity (relative area (RA) and the exponent D of the cumulative distribution function of LAA clusters size) between the simulated LAA images and those computed directly on the models output (considered as reference). RESULTS The LAA images obtained from our model output can simulate CT-LAA images in subjects with different grades of emphysema severity. Both RA and D computed on simulated LAA images were underestimated as compared to those calculated on the models output, suggesting that measurements in CT imaging may not be accurate in the assessment of real emphysema extent. CONCLUSIONS Our model is able to mimic the cluster size distribution of LAA on CT imaging of subjects with pulmonary emphysema. The model could be useful to generate standard test images and to design physical phantoms of LAA images for the assessment of the accuracy of indexes for the radiologic quantitation of emphysema.
Collapse
Affiliation(s)
- Stefano Diciotti
- Department of Electrical, Electronic, and Information Engineering "Guglielmo Marconi", University of Bologna, Via Venezia 52, 47521, Cesena, Italy.
| | - Alessandro Nobis
- Department of Clinical and Experimental Biomedical Sciences, University of Florence, Florence, Italy
| | - Stefano Ciulli
- Department of Clinical and Experimental Biomedical Sciences, University of Florence, Florence, Italy.,School of Computer Science and Electronic Engineering, University of Essex, Colchester, UK.,Medical Physics Section, Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Nicholas Landini
- Department of Clinical and Experimental Biomedical Sciences, University of Florence, Florence, Italy
| | - Mario Mascalchi
- Department of Clinical and Experimental Biomedical Sciences, University of Florence, Florence, Italy
| | - Nicola Sverzellati
- Section of Radiology, Department of Surgical Sciences, University of Parma, Parma, Italy
| | - Bernardo Innocenti
- BEAMS Department, École polytechnique de Bruxelles, ULB - Université Libre de Bruxelles, Bruxelles, Belgium
| |
Collapse
|
44
|
Moreira Gomes MD, Carvalho GMC, Casquilho NV, Araújo ACP, Valença SS, Leal-Cardoso JH, Zin WA. 2,2'-Azobis (2-Amidinopropane) Dihydrochloride Is a Useful Tool to Impair Lung Function in Rats. Front Physiol 2016; 7:475. [PMID: 27812337 PMCID: PMC5071333 DOI: 10.3389/fphys.2016.00475] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 10/03/2016] [Indexed: 01/24/2023] Open
Abstract
Recently, several studies have reported that respiratory disease may be associated with an increased production of free radicals. In this context, 2,2′-azobis (2-amidinopropane) dihydrochloride (AAPH) is a free radical-generating compound widely used to mimic the oxidative stress state. We aimed to investigate whether AAPH can generate lung functional, inflammatory, histological and biochemical impairments in the lung. Wistar rats were divided into five groups and instilled with saline solution (714 μL/kg, CTRL group) or different amounts of AAPH (25, 50, 100, and 200 mg/kg, 714 μL/kg, AAPH groups). Seventy-two hours later the animals were anesthetized, paralyzed, intubated and static elastance (Est), viscoelastic component of elastance (ΔE), resistive (ΔP1) and viscoelastic (ΔP2) pressures were measured. Oxidative damage, inflammatory markers and lung morphometry were analyzed. ΔP1 and Est were significantly higher in AAPH100 and AAPH200 than in the other groups. The bronchoconstriction indexes were larger in AAPH groups than in CTRL. The area occupied by collagen and elastic fibers, polymorpho- and mononuclear cells, malondialdehyde and carbonyl groups levels were significantly higher in AAPH200 than in CTRL. In comparison to CTRL, AAPH200 showed significant decrease and increase in the activities of superoxide dismutase and catalase, respectively. AAPH augmented the release of pro-inflammatory cytokines IL-1β, IL-6 e TNF-α. Hence, exposure to AAPH caused significant inflammatory alterations and redox imbalance accompanied by altered lung mechanics and histology. Furthermore, we disclosed that exposure to AAPH may represent a useful in vivo tool to trigger lung lesions.
Collapse
Affiliation(s)
- Maria D Moreira Gomes
- Laboratory of Respiration Physiology, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil; Electrophysiology Laboratory, Superior Institute of Biomedical Sciences, State University of CearáFortaleza, Brazil
| | - Giovanna M C Carvalho
- Laboratory of Respiration Physiology, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| | - Natalia V Casquilho
- Laboratory of Respiration Physiology, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| | - Andressa C P Araújo
- Laboratory of Respiration Physiology, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| | - Samuel S Valença
- Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| | - Jose H Leal-Cardoso
- Electrophysiology Laboratory, Superior Institute of Biomedical Sciences, State University of Ceará Fortaleza, Brazil
| | - Walter A Zin
- Laboratory of Respiration Physiology, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| |
Collapse
|
45
|
Abstract
BACKGROUND Lung volume reduction surgery (LVRS) performed to treat patients with severe diffuse emphysema was reintroduced in the nineties. Lung volume reduction surgery aims to resect damaged emphysematous lung tissue, thereby increasing elastic properties of the lung. This treatment is hypothesised to improve long-term daily functioning and quality of life, although it may be costly and may be associated with risks of morbidity and mortality. Ten years have passed since the last version of this review was prepared, prompting us to perform an update. OBJECTIVES The objective of this review was to gather all available evidence from randomised controlled trials comparing the effectiveness of lung volume reduction surgery (LVRS) versus non-surgical standard therapy in improving health outcomes for patients with severe diffuse emphysema. Secondary objectives included determining which subgroup of patients benefit from LVRS and for which patients LVRS is contraindicated, to establish the postoperative complications of LVRS and its morbidity and mortality, to determine which surgical approaches for LVRS are most effective and to calculate the cost-effectiveness of LVRS. SEARCH METHODS We identified RCTs by using the Cochrane Airways Group Chronic Obstructive Pulmonary Disease (COPD) register, in addition to the online clinical trials registers. Searches are current to April 2016. SELECTION CRITERIA We included RCTs that studied the safety and efficacy of LVRS in participants with diffuse emphysema. We excluded studies that investigated giant or bullous emphysema. DATA COLLECTION AND ANALYSIS Two independent review authors assessed trials for inclusion and extracted data. When possible, we combined data from more than one study in a meta-analysis using RevMan 5 software. MAIN RESULTS We identified two new studies (89 participants) in this updated review. A total of 11 studies (1760 participants) met the entry criteria of the review, one of which accounted for 68% of recruited participants. The quality of evidence ranged from low to moderate owing to an unclear risk of bias across many studies, lack of blinding and low participant numbers for some outcomes. Eight of the studies compared LVRS versus standard medical care, one compared two closure techniques (stapling vs laser ablation), one looked at the effect of buttressing the staple line on the effectiveness of LVRS and one compared traditional 'resectional' LVRS with a non-resectional surgical approach. Participants completed a mandatory course of pulmonary rehabilitation/physical training before the procedure commenced. Short-term mortality was higher for LVRS (odds ratio (OR) 6.16, 95% confidence interval (CI) 3.22 to 11.79; 1489 participants; five studies; moderate-quality evidence) than for control, but long-term mortality favoured LVRS (OR 0.76, 95% CI 0.61 to 0.95; 1280 participants; two studies; moderate-quality evidence). Participants identified post hoc as being at high risk of death from surgery were those with particularly impaired lung function, poor diffusing capacity and/or homogenous emphysema. Participants with upper lobe-predominant emphysema and low baseline exercise capacity showed the most favourable outcomes related to mortality, as investigators reported no significant differences in early mortality between participants treated with LVRS and those in the control group (OR 0.87, 95% CI 0.23 to 3.29; 290 participants; one study), as well as significantly lower mortality at the end of follow-up for LVRS compared with control (OR 0.45, 95% CI 0.26 to 0.78; 290 participants; one study). Trials in this review furthermore provided evidence of low to moderate quality showing that improvements in lung function parameters other than forced expiratory volume in one second (FEV1), quality of life and exercise capacity were more likely with LVRS than with usual follow-up. Adverse events were more common with LVRS than with control, specifically the occurrence of (persistent) air leaks, pulmonary morbidity (e.g. pneumonia) and cardiovascular morbidity. Although LVRS leads to an increase in quality-adjusted life-years (QALYs), the procedure is relatively costly overall. AUTHORS' CONCLUSIONS Lung volume reduction surgery, an effective treatment for selected patients with severe emphysema, may lead to better health status and lung function outcomes, specifically for patients who have upper lobe-predominant emphysema with low exercise capacity, but the procedure is associated with risks of early mortality and adverse events.
Collapse
Affiliation(s)
| | | | - Leong Ung Tiong
- The Queen Elizabeth HospitalDepartment of SurgeryAdelaideAustralia
| | - Brian J Smith
- The University of AdelaideSchool of MedicineAdelaideAustralia
| | | |
Collapse
|
46
|
Oliveira MV, Abreu SC, Padilha GA, Rocha NN, Maia LA, Takiya CM, Xisto DG, Suki B, Silva PL, Rocco PRM. Characterization of a Mouse Model of Emphysema Induced by Multiple Instillations of Low-Dose Elastase. Front Physiol 2016; 7:457. [PMID: 27774071 PMCID: PMC5054025 DOI: 10.3389/fphys.2016.00457] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 09/22/2016] [Indexed: 01/05/2023] Open
Abstract
Many experimental models have been proposed to study the pathophysiological features of emphysema, as well as to search for new therapeutic approaches for acute or chronically injured lung parenchyma. We aimed to characterize an emphysema model induced by multiple instillations of elastase by tracking changes in inflammation, remodeling, and cardiac function after each instillation. Forty-eight C57BL/6 mice were randomly assigned across two groups. Emphysema (ELA) animals received 1, 2, 3, or 4 intratracheal instillations of pancreatic porcine elastase (PPE, 0.2 IU) with a 1-week interval between them. Controls (C) received saline following the same protocol. Before and after implementation of the protocol, animals underwent echocardiographic analysis. After the first instillation of PPE, the percentage of mononuclear cells in the lung parenchyma increased compared to C (p = 0.0001). The second instillation resulted in hyperinflated alveoli, increased mean linear intercept, and reduced elastic fiber content in lung parenchyma compared to C (p = 0.0197). Following the third instillation, neutrophils and collagen fiber content in alveolar septa and airways increased, whereas static lung elastance was reduced compared to C (p = 0.0094). After the fourth instillation, the percentage of M1 macrophages in lungs; levels of interleukin-1β (IL-1β), keratinocyte-derived chemokine, hepatocyte growth factor (HGF), and vascular endothelial growth factor (VEGF); and collagen fiber content in the pulmonary vessel wall were increased compared to C (p = 0.0096). At this time point, pulmonary arterial hypertension was apparent, with increased diastolic right ventricular wall thickness. In conclusion, the initial phase of emphysema was characterized by lung inflammation with predominance of mononuclear cells, whereas at the late stage, impairment of pulmonary and cardiovascular functions was observed. This model enables analysis of therapies at different time points during controlled progression of emphysema. Accordingly, early interventions could focus on the inflammatory process, while late interventions should focus on restoring cardiorespiratory function.
Collapse
Affiliation(s)
- Milena V Oliveira
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Soraia C Abreu
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Gisele A Padilha
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Nazareth N Rocha
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de JaneiroRio de Janeiro, Brazil; Department of Physiology and Pharmacology, Fluminense Federal UniversityNiteroi, Brazil
| | - Lígia A Maia
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Christina M Takiya
- Laboratory of Cellular Pathology, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Debora G Xisto
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Bela Suki
- Department of Biomedical Engineering, Boston University Boston, MA, USA
| | - Pedro L Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| |
Collapse
|
47
|
Henriques I, Lopes-Pacheco M, Padilha GA, Marques PS, Magalhães RF, Antunes MA, Morales MM, Rocha NN, Silva PL, Xisto DG, Rocco PRM. Moderate Aerobic Training Improves Cardiorespiratory Parameters in Elastase-Induced Emphysema. Front Physiol 2016; 7:329. [PMID: 27536247 PMCID: PMC4971418 DOI: 10.3389/fphys.2016.00329] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 07/18/2016] [Indexed: 12/31/2022] Open
Abstract
Aim: We investigated the therapeutic effects of aerobic training on lung mechanics, inflammation, morphometry and biological markers associated with inflammation, and endothelial cell damage, as well as cardiac function in a model of elastase-induced emphysema. Methods: Eighty-four BALB/c mice were randomly allocated to receive saline (control, C) or 0.1 IU porcine pancreatic elastase (emphysema, ELA) intratracheally once weekly for 4 weeks. After the end of administration period, once cardiorespiratory impairment associated with emphysema was confirmed, each group was further randomized into sedentary (S) and trained (T) subgroups. Trained mice ran on a motorized treadmill, at moderate intensity, 30 min/day, 3 times/week for 4 weeks. Results: Four weeks after the first instillation, ELA animals, compared to C, showed: (1) reduced static lung elastance (Est,L) and levels of vascular endothelial growth factor (VEGF) in lung tissue, (2) increased elastic and collagen fiber content, dynamic elastance (E, in vitro), alveolar hyperinflation, and levels of interleukin-1β and tumor necrosis factor (TNF)-α, and (3) increased right ventricular diastolic area (RVA). Four weeks after aerobic training, ELA-T group, compared to ELA-S, was associated with reduced lung hyperinflation, elastic and collagen fiber content, TNF-α levels, and RVA, as well as increased Est,L, E, and levels of VEGF. Conclusion: Four weeks of regular and moderate intensity aerobic training modulated lung inflammation and remodeling, thus improving pulmonary function, and reduced RVA and pulmonary arterial hypertension in this animal model of elastase-induced emphysema.
Collapse
Affiliation(s)
- Isabela Henriques
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Miquéias Lopes-Pacheco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de JaneiroRio de Janeiro, Brazil; Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de JaneiroRio de Janeiro, Brazil
| | - Gisele A Padilha
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Patrícia S Marques
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Raquel F Magalhães
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Mariana A Antunes
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Marcelo M Morales
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Nazareth N Rocha
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de JaneiroRio de Janeiro, Brazil; Department of Physiology, Fluminense Federal UniversityNiterói, Brazil
| | - Pedro L Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Débora G Xisto
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro, Brazil
| |
Collapse
|
48
|
Yi E, Sato S, Takahashi A, Parameswaran H, Blute TA, Bartolák-Suki E, Suki B. Mechanical Forces Accelerate Collagen Digestion by Bacterial Collagenase in Lung Tissue Strips. Front Physiol 2016; 7:287. [PMID: 27462275 PMCID: PMC4940411 DOI: 10.3389/fphys.2016.00287] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 06/24/2016] [Indexed: 11/13/2022] Open
Abstract
Most tissues in the body are under mechanical tension, and while enzymes mediate many cellular and extracellular processes, the effects of mechanical forces on enzyme reactions in the native extracellular matrix (ECM) are not fully understood. We hypothesized that physiological levels of mechanical forces are capable of modifying the activity of collagenase, a key remodeling enzyme of the ECM. To test this, lung tissue Young's modulus and a nonlinearity index characterizing the shape of the stress-strain curve were measured in the presence of bacterial collagenase under static uniaxial strain of 0, 20, 40, and 80%, as well as during cyclic mechanical loading with strain amplitudes of ±10 or ±20% superimposed on 40% static strain, and frequencies of 0.1 or 1 Hz. Confocal and electron microscopy was used to determine and quantify changes in ECM structure. Generally, mechanical loading increased the effects of enzyme activity characterized by an irreversible decline in stiffness and tissue deterioration seen on both confocal and electron microscopic images. However, a static strain of 20% provided protection against digestion compared to both higher and lower strains. The decline in stiffness during digestion positively correlated with the increase in equivalent alveolar diameters and negatively correlated with the nonlinearity index. These results suggest that the decline in stiffness results from rupture of collagen followed by load transfer and subsequent rupture of alveolar walls. This study may provide new understanding of the role of collagen degradation in general tissue remodeling and disease progression.
Collapse
Affiliation(s)
- Eunice Yi
- Cell and Tissue Mechanics, Department of Biomedical Engineering, Boston University Boston, MA, USA
| | - Susumu Sato
- Cell and Tissue Mechanics, Department of Biomedical Engineering, Boston University Boston, MA, USA
| | - Ayuko Takahashi
- Cell and Tissue Mechanics, Department of Biomedical Engineering, Boston University Boston, MA, USA
| | | | - Todd A Blute
- Cell and Tissue Mechanics, Department of Biomedical Engineering, Boston University Boston, MA, USA
| | - Erzsébet Bartolák-Suki
- Cell and Tissue Mechanics, Department of Biomedical Engineering, Boston University Boston, MA, USA
| | - Béla Suki
- Cell and Tissue Mechanics, Department of Biomedical Engineering, Boston University Boston, MA, USA
| |
Collapse
|
49
|
Rizzi M, Tarsia P, La Spina T, Cristiano A, Frassanito F, Macaluso C, Airoldi A, Vanni S, Legnani D. A new approach to detect early lung functional impairment in very light smokers. Respir Physiol Neurobiol 2016; 231:1-6. [PMID: 27224237 DOI: 10.1016/j.resp.2016.05.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/28/2016] [Accepted: 05/16/2016] [Indexed: 10/21/2022]
Abstract
PURPOSE The aim of our study is to investigate if lung carbon monoxide diffusing capacity (DLCO) measured during effort is able to detect early respiratory functional impairment. METHODS We enrolled 25 very light smokers and 20 healthy non smokers. Subjects underwent plethysmography, DLCO (single breath technique) and calculated effective pulmonary blood flow (Qc) by rebreathing method. During exercise by cycle ergometer (duration 10±2min; recovery 11±3min) DLCO and Qc were calculated at 25% and 50% of theoretical maximum workload. RESULTS At baseline lung function and Qc did not differ between groups. DLCO and DLCO/Qc measured during exercise were significantly greater in non smokers (p<0.001); Qc was not statistically different. In very light smokers, DLCO, DLCO/Qc measured during exercise significantly correlated with the number of pack years (r=-0.60 p<0.001; r=-0.58 p<0.05; r=-0.55 p<0.05, respectively). CONCLUSIONS In very light smokers there is lung function impairment and our data show that DLCO during exercise may reveal this underlying early damage.
Collapse
Affiliation(s)
- Maurizio Rizzi
- Pulmonary Departement, "Luigi Sacco" University Hospital, Milan, Italy.
| | - Paolo Tarsia
- Pulmonary Departement IRCCS Policlinico Hospital, Milan, Italy, Italy.
| | - Tiziana La Spina
- Pulmonary Departement, "Luigi Sacco" University Hospital, Milan, Italy.
| | - Andrea Cristiano
- Pulmonary Departement, "Luigi Sacco" University Hospital, Milan, Italy.
| | | | - Claudio Macaluso
- Pulmonary Departement, "Luigi Sacco" University Hospital, Milan, Italy.
| | - Andrea Airoldi
- Pulmonary Departement, "Luigi Sacco" University Hospital, Milan, Italy.
| | - Silvia Vanni
- Pulmonary Departement, "Luigi Sacco" University Hospital, Milan, Italy.
| | - Delfino Legnani
- Pulmonary Departement, "Luigi Sacco" University Hospital, Milan, Italy.
| |
Collapse
|
50
|
M'Saad S, Kammoun K, Yangui I, Fourati H, Feki W, Marouen F, Daoud E, Kammoun S. Micropolyangéite, syndrome d’emphysème des sommets et fibrose pulmonaire des bases. Rev Mal Respir 2016; 33:391-6. [DOI: 10.1016/j.rmr.2015.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 07/01/2015] [Indexed: 11/26/2022]
|