Skip to content
BY-NC-ND 3.0 license Open Access Published by De Gruyter May 7, 2023

Animal models: An essential tool to dissect the heterogeneity of chronic obstructive pulmonary disease

  • Jingxian Jiang , Shuanglan Xu , Zi Chen , Weihua Liu , Liuchao Zhang , Jianmin Li , Zhou Zhu and Linfu Zhou EMAIL logo

Chronic obstructive pulmonary disease (COPD) is a heterogeneous disease characterized by persistent respiratory symptoms and progressive airflow obstruction. Its main pathophysiology includes chronic airway inflammation, mucus hypersecretion, airway remodeling, and emphysema.[1] Environment-gene-immune interactions drive the occurrence and development of COPD (Figure 1). Environmental and genetic factors may affect baseline lung function, causing abnormal lung development and disrupting lung homeostasis by activating type 1 and type 3 immunity.[2] COPD is predicted to be the third leading cause of death by 2030, and its prevalence continues to increase.[3] Mechanisms underlying the development and progression of COPD and its potential targets remain to be explored.

Figure 1 Environment-gene-immune interaction drives the occurrence and development of COPD, wherein combined risk factors induce the novel gene editing animals, and molecular biology and imaging tools are used to explore potential targets for diagnosis and treatment of the disease.
Figure 1

Environment-gene-immune interaction drives the occurrence and development of COPD, wherein combined risk factors induce the novel gene editing animals, and molecular biology and imaging tools are used to explore potential targets for diagnosis and treatment of the disease.

Animal models on the way to dissect heterogeneity

Human studies are complicated by individual genetic backgrounds, environmental factors, and limitations in sample collection. Animal models are valuable tools for therapeutic testing before progression to humans (Table 1). Several animal species have been used as a model of COPD, including mice, guinea pigs, rats, hamsters, ferrets, and monkeys. Each animal model exhibits advantages and disadvantages. Mice have become the most popular model owing to their low cost, availability of molecular reagents, mature gene editing, and sequencing technologies.[4] Guinea pigs are superior to other animals in the development of mucus hypersecretion and emphysema;[5] however, there are interspecies differences in lung anatomy, lung development, and maturity. Progressive narrowing of terminal bronchioles accompanied by emphysema typically starts in the respiratory bronchioles, whereas rodents often lack these and have far fewer bronchial branches than humans. Guinea pigs and monkeys have well-developed alveoli at birth, whereas those of rats and mice develop after birth[6] Therefore, it is critical to recognize the limitations of current animal models.

Table 1

Different methods of inducing COPD animal models

Stimulant Species Methods Ref.
CS Mice Acute model: 5 cigarettes (with filter); for 20 min [30, 31, 32]
Chronic model: 3 cigarettes per day; 5 days per week; for 7 months
Nose only: 75 min per time; twice per day; 5 days per week; for 8 or 12 weeks Whole-body exposure: 5 cigarettes; 4 times per day; 30 min per interval; 5 days per week; for 4 or 24 weeks
Nose only: 12 cigarettes per run; twice per day; 5 days per week; for 8 weeks
Whole-body exposure: 5 cigarettes (without filter); 4 times per day; 5 days per week; for 4 weeks
Rat 7 h per day; 5 days per week; for 7 or 13 months [33, 34, 35]
1 mL PBS per cigarette; injected intraperitoneally on days 1, 8, and 15
5 cigarettes; 30 min per time; twice per day; 6 days per week; for 4 weeks
Guinea pig 10 cigarettes per day; 5 days per week; for 1, 3, and 6 months [36, 37, 38]
7 cigarettes per day; 5 days per week; for 3, 4, and 6 months
Nose noly: 7 cigarettes per day (without filter); 5 days per week; for 12 weeks
LPS Mice 5 μg LPS (it); twice per week; for 12 weeks [12, 13]
2 mg nebulized LPS; 15 min per day; 5 days per week; for 1, 4, and 8 weeks
Rat 200 μg LPS (it); twice per week; for 3 weeks [39]
Guinea pig Single exposure: 30 μg/mL nebulized LPS, for 1 h [14, 40]
Chronic exposure: 30 μg/mL nebulized LPS, for 1 h, 47 h per interval; for 9 times
30 μg/mL nebulized LPS, for 1 h, 47 h per interval; for 15 times
Elastase Mice 100 U/kg PPE (it) [41, 42]
100 U/kg PPE (it)
Rat 75 U PPE (it) [17, 43]
25 U/kg PPE, on days 1, and 10
Hamster 40 U PPE [44]
Ozone Mice 2.5 ppm ozone; 3 h per day; twice per week; for over 6 weeks [18]
PM2.5 Mice 10 h per day; 7 days per week; for 3 months [45]
Rat 2 h per time; twice per day; 5 days per week; for 7 months [46]
  1. COPD: chronic obstructive pulmonary disease; CS: cigarette smoking; LPS: lipopolysaccharide. PPE: porcine pancreatic elastase; it: intratracheal; ppm: parts per million.

Risk factors MIMIC the real world

Cigarette smoking (CS) is a major risk factor for COPD in high-income countries, whereas exposure to cooking biomass fuels is a major precipitant in low-income countries.[7] CS was the most common COPD-inducer used in previous studies. A variety of animals have been used to establish COPD models with CS, wherein guinea pigs are perhaps the most susceptible species owing to their significant airspace enlargement.[8] Rats appear to be the most resistant to emphysema and develop nonspecific particle overload effects, whereas susceptibility in mice appears to be strain-dependent. Similar to that in humans, COPD features are not suppressed by corticosteroid treatment in animal models. However, the method of smoke generation, constituents of CS, delivery systems used, and dosage varied among studies, leading to poor homogeneity of Results.[9] Pathological phenotypes, such as emphysema, airway remodeling, and pulmonary hypertension, can only be induced in chronic CS models, as it is time-consuming. One month after CS exposure, an acute phase dominated with maximal neutrophil infiltration during the first week, followed by a progressive chronic phase consisting of infiltrating neutrophils, macrophages, and lymphocytes in the lungs.[10] Another limitation is the degree of induced COPD equivalent to Global Initiative for Obstructive Lung Disease (GOLD) stage 1 or 2 in CS models, whereas most exacerbations and deaths occur during GOLD stage 3 or 4, and inflammation persists after smoking cessation in patients with COPD.[11] Therefore, the CS model differs from the real COPD phenotype to some extent.

Lipopolysaccharide (LPS) is present in CS, air pollutants, and organic dust as a major component of the outer cell wall of gram-negative bacteria. Chronic exposure of animals to LPS has been shown to induce pathological features of COPD, including pronounced neutrophilic inflammation, enlarged air spaces, airway remodeling, and decreased lung function.[12, 13] However, robust inflammation can be inhibited by glucocorticoids, and repeated administration induces immune tolerance, which can alleviate neutrophilic inflammation.[14]

Excessive elastase secretion can induce matrix destruction, and the elastase model can induce tissue damage and emphysema. The development of emphysema following the instillation of elastases, including neutrophil elastase (NE), porcine pancreatic elastase (PPE), and papain, Results in rapid and significant airspace enlargement, followed by acute neutrophil and macrophage accumulation and mucus cell metaplasia.[15] COPD can be induced by a single instillation and its severity is adjusted to the dosage of elastase,[16] which makes it cheaper and easier to induce than chronic smoke exposure. This model is ideal for testing the efficacy of therapeutic agents, particularly those with the capacity to reverse or repair emphysematous damage to the lungs.[17] However, detailed mechanisms of the COPD proteolytic process are quite different from those of the elastase model. In addition, inflammation is transient and resolves within one week of elastase administration.

Pollutants (PM2.5), ozone, and other irritants were also applied to the experimental animals. Ozone initiates intracellular oxidative stress through the formation of ozonide and hydrogen peroxide, which induces airway inflammation, airway hyperresponsiveness, lung destruction, and a steroid-insensitive phenotype within six weeks.[18] Combination inducers, such as CS, PPE,LPS, bacteria, viruses, and pollutants, are widely used to simulate the different stages of COPD, which is of great value in exploring how environmental risk factors affect the development and deterioration of COPD.[19,20] For example, mice were intranasally injected with PPE and LPS for 4 weeks, which can induce COPD-like lung inflammation and alveolar enlargement.[21]

However, the existing single or combined risk factor-induced models can only partially mimic the characteristics of COPD, and more risk factors should be considered to refine the animal models.

Genetic factors reveal susceptibility

CS is a major risk factor for COPD; however, only 15%– 20% of smokers are susceptible to developing COPD and 25%–45% of patients with COPD have never smoked. Therefore, it is being increasingly recognized that COPD is not merely caused by smoking.[7] The heterogeneity of COPD indicates that individuals have different susceptibilities to it, and biological networks consisting of genes and proteins are important determinants of COPD.

Severe alpha-1 antitrypsin deficiency was the first documented genetic determinant of COPD.[22] Over the past decade, a large number of studies have focused on the role of genetic variants in COPD using classical genome-wide association studies (GWAS) that have successfully identified many genomic regions in association with COPD susceptibility, as listed in Table 2.[23, 24] Studies of candidate genes have linked specific loci to phenotypes of COPD;[25,26] therefore, the newly discovered susceptibility genes could provide novel insights into COPD pathogenesis. However, identifying functional variants and key genes within these regions remains a major challenge. Therefore, an integrated approach combining GWAS and other omics sequencing data (transcriptomics, epigenetic analysis, proteomics, metabolomics, etc.) may be required to provide a more comprehensive view of the genetic architecture of COPD.[27]

Table 2

Candidate genes for COPD suggested by genome-wide association studies

Gene Locus rsID Function Phenotype Ref.
FAM13A 4q22.1 rs2013701 rs7671167 rs10007590 rs2869966 rs2869967 rs2045517 Rho GTPase signaling Fatty acid oxidation Mitochondrial function Alveolar epithelial cells repair and regeneration Severe COPD Lung function (FEV1/FVC) Emphysema [23, 25, 47, 48]
TGFB2 1q41 rs1690789 rs4846480 rs6684205 Lung development Tissue repair and remodeling Severe COPD Lung function (FEV1/FVC) Emphysema [23, 49]
HHIP 4q31.21 rs1828591 rs13118928 rs6537296 rs1542725 rs6817273 rs10519717 rs1980057 rs1032295 Hedgehog signaling pathway Embryonic development Lung organogenesis Oxidative stress alleviation Airway remodeling repression Severe COPD COPD exacerbation Lung function (FEV1/FVC, FEVAirflow obstruction Emphysema Fat-free body mass Body mass index [23, 25, 48, 50, 51, 52] 1)
HTR4 5q32 rs7733088 rs11168048 rs7735184 A major neurotransmitter in the CNS Learning, memory, depression, anxiety and feeding behaviour Lung development Lung function (FEV1/FVC, FEVAirflow obstruction 1) [48, 51, 53]
CHRNA3/5 15q25.1 rs8034191 rs1051730 rs17486278 rs16969968 rs8040863 rs55853698 rs6495308 rs13141641 Nicotinic acetylcholine receptor Cell-cycle regulation Severe COPD Lung function (FEV1/FVC, FEVAirflow obstruction Emphysema Nicotine addiction [23, 25, 50, 51] 1)
MMP12 11q22 rs2276109 rs652438 rs626750 Tissue repair and remodelling Protease - antiprotease imbalance Severe/very severe COPD Lung function (FEV1) Emphysema [23, 54]
AGER 6p21.32 rs2070600 Epithelium–extracellular matrix interaction Cell surface receptor receptor Homeostasis, development and inflammation Lung function (FEV1/FVC) Emphysema [24, 48]
SFTPD 10q22.3 rs721917 rs2245121 rs911887 rs6413520 rs7078012 Host defence Surfactant metabolism A promising COPD biomarker Lung function ( FEV1) Emphysema [55]
ADAM19 5q33.3 rs1422795 Immune defense Inflammatory process Extracellular matrix breakdown and reconstruction Lung function (FEV1/FVC) Airflow obstruction [48]
RARB 3p24.2 rs1529672 Active form of vitamin A Embryonic morphogenesis Cell growth and differentiation Tumor suppression Lung function (FEV1/FVC) [56]
  1. FAM13A: family with sequence similarity 13 member A; TGFB2: transforming growth factor beta 2; HHIP: hedgehog interacting protein; HTR4: hydroxytryptamine receptor 4; CHRNA3/5: cholinergic receptor nicotinic alpha 3/5; MMP12: matrix metalloprotein 12; AGER: advanced glycosylation end-product specific receptor; SFTPD: surfactant protein D; ADAM19: a disintegrin and metalloprotease 19; RARB: retinoic acid receptor beta; FEV1: forced expiratory volume in 1 second; FVC: predicted forced vital capacity.

Drug targets for personalized medicine

COPD, characterized by non-type 2 inflammation, is usually corticosteroid insensitive. Smoking cessation attenuates the accelerated decline in lung function in patients with COPD. Long-acting β2-adrenergic receptor agonists and long-acting muscarinic acetylcholine are beneficial for alleviating symptoms.[11] However, although drugs produce effective bronchodilation, no drugs are available to considerably reduce disease progression or mortality. This has prompted a concerted search for new treatments and biomarkers that can monitor the responses of specific patients. Many potential drug targets have been implicated in the development of COPD animal models. These targets include inflammation medium inhibitors, antioxidants, and kinase inhibitors, in particular, drugs targeting cell regeneration, microbial colonization, and corticosteroid resistance.[28,29] However, some targets have so far not shown clear clinical benefits in COPD, albeit they are limited by side effects owing to the widespread distribution of intracellular targets.[57]

In summary, animal models should be developed to accurately simulate the distinct clinical features of COPD. It is necessary to optimize the existing modeling program from an array of aspects, such as the choice of combined factors, species and susceptibility genes, construction of multi-locus gene-editing animals, and application of advanced imaging tools. Based on analysis of phenotypes, mechanisms of the disease should be further investigated in combination with multi-omics analyses. With a better understanding of the disease, more potent targets may be discovered in the future, which can reduce the burden of comorbidities and treatment costs. The development of biomarkers could optimize the selection of patient populations for clinical trials, and therapies can be tailored to an individual patient’s disease profile for personalized medicine.


#Jingxian Jiang, Shuanglan Xu, and Zi Chen contributed equally to this work.

Prof. Linfu Zhou, Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, Jiangsu Province, China.

Funding statement: This work was supported by grants from the National Key Research and Development Program of China 2022YFF0710800 and 2018YFC1313600, Major International (Regional) Joint Research Project of China 81820108001, National Natural Science Foundation of China 81670029, Jiangsu Key Principal Investigator of Medicine ZDRCA2016018, and Jiangsu Provincial Project 333 for Cultivation of High-Level Talents (Leading Talents of the Young and Middle-Aged) BRA2019078 (to L. Zhou).

  1. Conflict of Interest

    The authors have nothing to disclose.

References

1 McDonough JE, Yuan R, Suzuki M, Seyednejad N, Elliott WM, Sanchez PG, et al. Small-airway obstruction and emphysema in chronic obstructive pulmonary disease. N Engl J Med 2011; 365:1567-7510.1056/NEJMoa1106955Search in Google Scholar PubMed PubMed Central

2 Brusselle GG, Joos GF, Bracke KR. New insights into the immunology of chronic obstructive pulmonary disease. Lancet 2011; 378:1015-2610.1016/S0140-6736(11)60988-4Search in Google Scholar PubMed

3 Lozano R, Naghavi M, Foreman K, Lim S, Shibuya K, Aboyans V, et al. Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet 2012; 380:2095-12810.1016/S0140-6736(12)61728-0Search in Google Scholar PubMed

4 Serban KA,Petrache I. Mouse models of COPD. Methods Mol Biol 2018; 1809:379-9410.1007/978-1-4939-8570-8_25Search in Google Scholar PubMed

5 Wright JL, Ngai T, Churg A. Effect of long-term exposure to cigarette smoke on the small airways of the guinea pig. Exp Lung Res 1992; 18:1051410.3109/01902149209020654Search in Google Scholar PubMed

6 Massaro D, Massaro GD. Developmental alveologenesis: longer, differential regulation and perhaps more danger. Am J Physiol Lung Cell Mol Physiol 2007; 293:L568-910.1152/ajplung.00258.2007Search in Google Scholar PubMed

7 Salvi SS, Barnes PJ. Chronic obstructive pulmonary disease in nonsmokers. Lancet 2009; 374:733-4310.1016/S0140-6736(09)61303-9Search in Google Scholar PubMed

8 Wright JL, Churg A. Cigarette smoke causes physiologic and morphologic changes of emphysema in the guinea pig. Am Rev Respir Dis 1990; 142:1422-810.1164/ajrccm/142.6_Pt_1.1422Search in Google Scholar PubMed

9 Leberl M, Kratzer A, Taraseviciene-Stewart L. Tobacco smoke induced COPD/emphysema in the animal model: are we all on the same page? Front Physiol 2013; 4:9110.3389/fphys.2013.00091Search in Google Scholar PubMed PubMed Central

10 D’Hulst AI, Vermaelen KY, Brusselle GG, Joos GF, Pauwels RA. Time course of cigarette smoke-induced pulmonary inflammation in mice. Eur Respir J 2005; 26:204-1310.1183/09031936.05.00095204Search in Google Scholar PubMed

11 Singh D, Agusti A, Anzueto A, Barnes PJ, Bourbeau J, Celli BR, et al. Global Strategy for the Diagnosis, Management, and Prevention of Chronic Obstructive Lung Disease: the GOLD science committee report 2019. Eur Respir J 2019; 5310.1183/13993003.00164-2019Search in Google Scholar PubMed

12 Vernooy JH, Dentener MA, van Suylen RJ, Buurman WA, Wouters EF. Long-term intratracheal lipopolysaccharide exposure in mice results in chronic lung inflammation and persistent pathology. Am J Respir Cell Mol Biol 2002; 26:152-910.1165/ajrcmb.26.1.4652Search in Google Scholar PubMed

13 Lai PS, Fresco JM, Pinilla MA, Macias AA, Brown RD, Englert JA, et al. Chronic endotoxin exposure produces airflow obstruction and lung dendritic cell expansion. Am J Respir Cell Mol Biol 2012; 47:209-1710.1165/rcmb.2011-0447OCSearch in Google Scholar PubMed PubMed Central

14 Kaneko Y, Takashima K, Suzuki N, Yamana K. Effects of theophylline on chronic inflammatory lung injury induced by LPS exposure in guinea pigs. Allergol Int 2007; 56:445-5610.2332/allergolint.O-07-490Search in Google Scholar PubMed

15 Morichika D, Taniguchi A, Oda N, Fujii U, Senoo S, Itano J, et al. Loss of IL-33 enhances elastase-induced and cigarette smoke extract-induced emphysema in mice. Respir Res 2021; 22:15010.1186/s12931-021-01705-zSearch in Google Scholar PubMed PubMed Central

16 Antunes MA,Rocco PR. Elastase-induced pulmonary emphysema: insights from experimental models. An Acad Bras Cienc 2011; 83:1385-9610.1590/S0001-37652011005000039Search in Google Scholar PubMed

17 Rubio ML, Martin-Mosquero MC, Ortega M, Peces-Barba G, Gonzalez-Mangado N. Oral N-acetylcysteine attenuates elastase-induced pulmonary emphysema in rats. Chest 2004; 125:1500-610.1378/chest.125.4.1500Search in Google Scholar PubMed

18 Li F, Xu M, Wang M, Wang L, Wang H, Zhang H, et al. Roles of mitochondrial ROS and NLRP3 inflammasome in multiple ozone-induced lung inflammation and emphysema. Respir Res 2018; 19:23010.1186/s12931-018-0931-8Search in Google Scholar PubMed PubMed Central

19 Sajjan U, Ganesan S, Comstock AT, Shim J, Wang Q, Nagarkar DR, et al. Elastase- and LPS-exposed mice display altered responses to rhinovirus infection. Am J Physiol Lung Cell Mol Physiol 2009; 297:L931-4410.1152/ajplung.00150.2009Search in Google Scholar PubMed PubMed Central

20 Ganesan S, Faris AN, Comstock AT, Sonstein J, Curtis JL, Sajjan US. Elastase/LPS-exposed mice exhibit impaired innate immune responses to bacterial challenge: role of scavenger receptor A. Am J Pathol 2012; 180:61-7210.1016/j.ajpath.2011.09.029Search in Google Scholar PubMed PubMed Central

21 Yadava K, Pattaroni C, Sichelstiel AK, Trompette A, Gollwitzer ES, Salami O, et al. Microbiota promotes chronic pulmonary inflammation by enhancing IL-17A and autoantibodies. Am J Respir Crit Care Med 2016; 193:975-8710.1164/rccm.201504-0779OCSearch in Google Scholar PubMed

22 Shigeoka JW, Hall WJ, Hyde RW, Schwartz RH, Mudholkar GS, Speers DM, et al. The prevalence of alpha-antitrypsin heterozygotes (Pi MZ) in patients with obstructive pulmonary disease. Am Rev Respir Dis 1976; 114:1077-84Search in Google Scholar

23 Cho MH, McDonald ML, Zhou X, Mattheisen M, Castaldi PJ, Hersh CP, et al. Risk loci for chronic obstructive pulmonary disease: a genome-wide association study and meta-analysis. Lancet Respir Med 2014; 2:214-2510.1016/S2213-2600(14)70002-5Search in Google Scholar PubMed PubMed Central

24 Repapi E, Sayers I, Wain LV, Burton PR, Johnson T, Obeidat M, et al. Genome-wide association study identifies five loci associated with lung function. Nat Genet 2010; 42:36-4410.1164/ajrccm-conference.2010.181.1_MeetingAbstracts.A1312Search in Google Scholar

25 Pillai SG, Kong X, Edwards LD, Cho MH, Anderson WH, Coxson HO, et al. Loci identified by genome-wide association studies influence different disease-related phenotypes in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2010; 182:1498-150510.1164/rccm.201002-0151OCSearch in Google Scholar PubMed PubMed Central

26 Sakornsakolpat P, Prokopenko D, Lamontagne M, Reeve NF, Guyatt AL, Jackson VE, et al. Genetic landscape of chronic obstructive pulmonary disease identifies heterogeneous cell-type and phenotype associations. Nat Genet 2019; 51:494-50510.1038/s41588-018-0342-2Search in Google Scholar PubMed PubMed Central

27 Kan M, Shumyatcher M,Himes BE. Using omics approaches to understand pulmonary diseases. Respir Res 2017; 18:14910.1186/s12931-017-0631-9Search in Google Scholar PubMed PubMed Central

28 Ti H, Zhou Y, Liang X, Li R, Ding K,Zhao X. Targeted treatments for chronic obstructive pulmonary disease (COPD) using low-molecular-weight drugs (LMWDs). J Med Chem 2019; 62:5944-7810.1021/acs.jmedchem.8b01520Search in Google Scholar PubMed

29 Barnes PJ. Emerging pharmacotherapies for COPD. Chest 2008; 134:1278-8610.1378/chest.08-1385Search in Google Scholar PubMed

30 Martorana PA, Beume R, Lucattelli M, Wollin L,Lungarella G. Roflumi-last fully prevents emphysema in mice chronically exposed to cigarette smoke. Am J Respir Crit Care Med 2005; 172:848-5310.1164/rccm.200411-1549OCSearch in Google Scholar PubMed

31 Kim RY, Sunkara KP, Bracke KR, Jarnicki AG, Donovan C, Hsu AC, et al. A microRNA-21-mediated SATB1/S100A9/NF-kappaB axis promotes chronic obstructive pulmonary disease pathogenesis. Sci Transl Med 2021; 13:eaav722310.1126/scitranslmed.aav7223Search in Google Scholar PubMed

32 Lu Z, Van Eeckhoutte HP, Liu G, Nair PM, Jones B, Gillis CM, et al. Necroptosis signaling promotes inflammation, airway remodeling, and emphysema in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2021; 204:667-8110.1164/rccm.202009-3442OCSearch in Google Scholar PubMed

33 March TH, Barr EB, Finch GL, Hahn FF, Hobbs CH, Menache MG, et al. Cigarette smoke exposure produces more evidence of emphysema in B6C3F1 mice than in F344 rats. Toxicol Sci 1999; 51:289-9910.1093/toxsci/51.2.289Search in Google Scholar PubMed

34 Chen Y, Hanaoka M, Droma Y, Chen P, Voelkel NF,Kubo K. Endothelin-1 receptor antagonists prevent the development of pulmonary emphysema in rats. Eur Respir J 2010; 35:904-1210.1183/09031936.00003909Search in Google Scholar PubMed

35 Chen L, Sun BB, Wang T, Wang X, Li JQ, Wang HX, et al. Cigarette smoke enhances beta-defensin 2 expression in rat airways via nuclear factor-kappaB activation. Eur Respir J 2010; 36:638-4510.1183/09031936.00029409Search in Google Scholar PubMed

36 Churg A, Wang R, Wang X, Onnervik PO, Thim K,Wright JL. Effect of an MMP-9/MMP-12 inhibitor on smoke-induced emphysema and airway remodelling in guinea pigs. Thorax 2007; 62:706-1310.1136/thx.2006.068353Search in Google Scholar PubMed PubMed Central

37 Barreiro E, Peinado VI, Galdiz JB, Ferrer E, Marin-Corral J, Sanchez F, et al. Cigarette smoke-induced oxidative stress: a role in chronic obstructive pulmonary disease skeletal muscle dysfunction. Am J Respir Crit Care Med 2010; 182:477-8810.1164/rccm.200908-1220OCSearch in Google Scholar PubMed

38 Dominguez-Fandos D, Valdes C, Ferrer E, Puig-Pey R, Blanco I, Tura-Ceide O, et al. Sildenafil in a cigarette smoke-induced model of COPD in the guinea-pig. Eur Respir J 2015; 46:346-5410.1183/09031936.00139914Search in Google Scholar PubMed

39 Zhang F, Guo F, Liu Y, Zhang Y, Li D,Yang H. Shema oral liquid ameliorates the severity of LPS-induced COPD via regulating DNMT1. Front Pharmacol 2022; 13:90359310.3389/fphar.2022.903593Search in Google Scholar PubMed PubMed Central

40 Johnson FJ, Reynolds LJ, Toward TJ. Elastolytic activity and alveolar epithelial type-1 cell damage after chronic LPS inhalation: effects of dexamethasone and rolipram. Toxicol Appl Pharmacol 2005; 207:257-6510.1016/j.taap.2005.01.006Search in Google Scholar PubMed

41 Kneidinger N, Yildirim AO, Callegari J, Takenaka S, Stein MM, Dumitrascu R, et al. Activation of the WNT/beta-catenin pathway attenuates experimental emphysema. Am J Respir Crit Care Med 2011; 183:723-3310.1164/rccm.200910-1560OCSearch in Google Scholar PubMed

42 Hou HH, Cheng SL, Chung KP, Kuo MY, Yeh CC, Chang BE, et al. Elastase induces lung epithelial cell autophagy through placental growth factor: a new insight of emphysema pathogenesis. Autophagy 2014; 10:1509-2110.4161/auto.29190Search in Google Scholar PubMed PubMed Central

43 Sohrabi F, Dianat M, Badavi M, Radan M,Mard SA. Gallic acid suppresses inflammation and oxidative stress through modulating Nrf2-HO-1-NF-kappaB signaling pathways in elastase-induced emphysema in rats. Environ Sci Pollut Res Int 2021; 28:56822-3410.1007/s11356-021-14513-1Search in Google Scholar PubMed

44 Degens H, Swisher AK, Heijdra YF, Siu PM, Dekhuijzen PN, Alway SE. Apoptosis and Id2 expression in diaphragm and soleus muscle from the emphysematous hamster. Am J Physiol Regul Integr Comp Physiol 2007; 293:R135-4410.1152/ajpregu.00046.2007Search in Google Scholar PubMed

45 Li M, Hua Q, Shao Y, Zeng H, Liu Y, Diao Q, et al. Circular RNA circBbs9 promotes PM(2.5)-induced lung inflammation in mice via NLRP3 inflammasome activation. Environ Int 2020; 143:10597610.1016/j.envint.2020.105976Search in Google Scholar PubMed

46 Guo X, Lin Y, Lin Y, Zhong Y, Yu H, Huang Y, et al. PM2.5 induces pulmonary microvascular injury in COPD via METTL16-mediated m6A modification. Environ Pollut 2022; 303:11911510.1016/j.envpol.2022.119115Search in Google Scholar PubMed

47 Castaldi PJ, Guo F, Qiao D, Du F, Naing ZZC, Li Y, et al. Identification of functional variants in the FAM13A chronic obstructive pulmonary disease genome-wide association study locus by massively parallel reporter assays. Am J Respir Crit Care Med 2019; 199:52-6110.1164/rccm.201802-0337OCSearch in Google Scholar PubMed PubMed Central

48 Hancock DB, Eijgelsheim M, Wilk JB, Gharib SA, Loehr LR, Marciante KD, et al. Meta-analyses of genome-wide association studies identify multiple loci associated with pulmonary function. Nat Genet 2010; 42:45-5210.1038/ng.500Search in Google Scholar PubMed PubMed Central

49 Parker MM, Hao Y, Guo F, Pham B, Chase R, Platig J, et al. Identification of an emphysema-associated genetic variant near TGFB2 with regulatory effects in lung fibroblasts. Elife 2019; 810.7554/eLife.42720Search in Google Scholar PubMed PubMed Central

50 Pillai SG, Ge D, Zhu G, Kong X, Shianna KV, Need AC, et al. A genomewide association study in chronic obstructive pulmonary disease (COPD): identification of two major susceptibility loci. PLoS Genet 2009; 5:e100042110.1371/journal.pgen.1000421Search in Google Scholar PubMed PubMed Central

51 Wilk JB, Shrine NR, Loehr LR, Zhao JH, Manichaikul A, Lopez LM, et al. Genome-wide association studies identify CHRNA5/3 and HTR4 in the development of airflow obstruction. Am J Respir Crit Care Med 2012; 186:622-3210.1164/rccm.201202-0366OCSearch in Google Scholar PubMed PubMed Central

52 Van Durme YM, Eijgelsheim M, Joos GF, Hofman A, Uitterlinden AG, Brusselle GG, et al. Hedgehog-interacting protein is a COPD susceptibility gene: the Rotterdam Study. Eur Respir J 2010; 36:89-9510.1183/09031936.00129509Search in Google Scholar PubMed

53 Hodge E, Nelson CP, Miller S, Billington CK, Stewart CE, Swan C, et al. HTR4 gene structure and altered expression in the developing lung. Respir Res 2013; 14:7710.1186/1465-9921-14-77Search in Google Scholar PubMed PubMed Central

54 Haq I, Chappell S, Johnson SR, Lotya J, Daly L, Morgan K, et al. Association of MMP-2 polymorphisms with severe and very severe COPD: a case control study of MMPs-1, 9 and 12 in a European population. BMC Med Genet 2010; 11:710.1186/1471-2350-11-7Search in Google Scholar PubMed PubMed Central

55 Foreman MG, Kong X, DeMeo DL, Pillai SG, Hersh CP, Bakke P, et al. Polymorphisms in surfactant protein-D are associated with chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol 2011; 44:316-2210.1165/rcmb.2009-0360OCSearch in Google Scholar PubMed PubMed Central

56 Soler Artigas M, Loth DW, Wain LV, Gharib SA, Obeidat M, Tang W, et al. Genome-wide association and large-scale follow up identifies 16 new loci influencing lung function. Nat Genet 2011; 43:1082-9010.1038/ng.941Search in Google Scholar PubMed PubMed Central

57 Sun L, Chen Y. Interpretation of the key issues of expert consensus on immunomodulatory therapies for chronic obstructive pulmonary disease. J Transl Intern Med 2022;10:277-28010.2478/jtim-2022-0069Search in Google Scholar PubMed PubMed Central

Published Online: 2023-05-07

© 2023 Jingxian Jiang, Shuanglan Xu, Zi Chen, Weihua Liu, published by Sciendo

This work is licensed under the Creative Commons Attribution-NonCommercial-NoDerivatives 3.0 License.

Downloaded on 31.5.2024 from https://www.degruyter.com/document/doi/10.2478/jtim-2023-0007/html
Scroll to top button