• No results found

University of Groningen Regenerative Pharmacology for COPD Wu, Xinhui

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Regenerative Pharmacology for COPD Wu, Xinhui"

Copied!
21
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

University of Groningen

Regenerative Pharmacology for COPD

Wu, Xinhui

DOI:

10.33612/diss.157528459

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date: 2021

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Wu, X. (2021). Regenerative Pharmacology for COPD. University of Groningen. https://doi.org/10.33612/diss.157528459

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

XINHUI

WU

Chapter 1

(3)

Preface

Chronic obstructive pulmonary disease (COPD) is characterized by progressive and incompletely reversible airway obstruction, in some patients associated with emphysema caused by enhanced tissue destruction in combination with defective airway and alveolar repair1–3. The main causes of COPD are tobacco smoke and air

pollution resulting in accelerated lung function decline in susceptible individuals. Current treatment options are solely reducing symptoms, but do not reverse or slow down the progression of the disease. The objective of this thesis is to unravel the mechanisms underlying cigarette smoke and air pollution-induced dysfunctional alveolar epithelial repair, thereby aiming to discover novel therapeutic targets that could be beneficial to lung repair in COPD.

COPD prevalence and perspective

Chronic obstructive pulmonary disease (COPD) is a complex and heterogenous syndrome characterized by persistent respiratory symptoms and airflow limitation that is due to airway and/or alveolar abnormalities. Globally, there are around three million deaths annually, the estimated number of COPD cases was 384 million in 2010, with a global prevalence of 11.7% and resulting in significant social as well as economic burden 4.

Pathogenesis of COPD

The growing number of COPD cases in recent years is due to the pandemic of tobacco smoking, due to environmental pollution and due to ageing of the world population5,6.

Though the pathobiological mechanisms of COPD remain incompletely understood, various contributors are suggested to play essential roles in COPD pathogenesis, such as the protease-antiprotease imbalance, the oxidant-antioxidant imbalance, cellular senescence, autoimmunity, chronic inflammation, deficient lung growth and development, metaplastic epithelial injuries, and ineffective lung repair5,7.

(4)

1

Airway remodeling

Airway remodeling8–10 is one of the major pathological features of COPD (Fig.1).

It is defined as structural alterations of both small and large airways and includes epithelial abnormalities, extracellular matrix abnormalities and increased smooth muscle mass of airways. The obstruction of the small airways in COPD is associated with the airway wall thickening by means of a remodeling process related to aberrant tissue repair and a malfunction of the mucociliary clearance of the innate host defense system, which results in the accumulation of inflammation11. There is

no available therapy proven to be prevent or reverse airway remodeling, although reversibility of airway remodeling is suggested by studies in animal models12. Airway

remodeling is caused by aberrant wound healing in response to injuries by such as cigarette smoke, viruses and bacteria, and wound healing is tightly regulated by the interaction between an immune response to these stimuli and remodeling of the extracellular matrix (ECM) by myofibroblasts13,14. Structural alterations in COPD

airways occur and involve dysregulation of the epithelial-mesenchymal unit, with epithelial modification and peribronchial fibrosis15-17.

Alveolar destruction (Emphysema)

Emphysema18,19 is another important characteristic of COPD, which is characterised

by alveolar wall destruction and loss of surface area in the lung parenchyma (Fig.1) where gas exchange occurs. However, the pathogenesis of emphysema is not yet well understood, and there is no effective pharmacological treatment against it. For patients with severe emphysema20, the limited therapeutic options available

(5)

Figure 1. Overview of pulmonary pathologies of the COPD lung.

Causes of COPD

Tobacco smoking

Tobacco smoking is the main risk factor for COPD, which results in a rapid decline in lung function in susceptible individuals. Smoking induces epithelial injury and repeated injuries of the epithelium trigger a pathophysiologic response resulting in tissue remodeling in the conducting airways and destruction of the respiratory bronchioles and alveoli21. Furthermore, the defective epithelium increases the

risk for bacterial and viral infections and subsequent exacerbations. The current SARS-Cov-2 pandemic illustrates this as an example22. The virus has infected more

than 15 million people globally22 by July 2020 with around 650,000 deaths, and

several studies23,24 showed that cigarette smokers are more susceptible to develop

Covid-19 and have more severe disease. This is associated with consistently higher levels of the SARS-Cov-2 receptor ACE2 (angiotensin-converting enzyme 2) in their respiratory tracts, and chronic smokers may exhibit a number of co-morbidities, including emphysema, atherosclerosis, and immune dysregulations that predispose to disease severity25.

Oxidative stress has been suggested to be a key driver of accelerated pulmonary ageing and abnormal repair in COPD. Increased reactive oxygen species (ROS)

(6)

1

and reactive nitrogen species (NRS) contribute to the occurrence of oxidative and nitrosative stress in the lungs26. Mitochondria are the major cellular oxidative stress

sensors that regulate metabolism, signaling, and energetics to maintain cellular and tissue homeostasis. Both cigarette smoke and air pollution are known to cause mitochondrial dysfunction contributing to the pathogenesis of chronic inflammatory lung diseases27–29.

Air pollution

Outdoor air pollution is a significantly risky factor to global morbidity and is responsible for 3.3 million premature deaths annually worldwide. Traffic-related air pollution is one of the most significant sources of outdoor air pollution exposure in many countries30,31. Emerging epidemiological studies30,32,33 demonstrate a strong

association between airborne pollutant levels and COPD exacerbations requiring hospitalization, indicating that COPD patients may be more susceptible to the harmful effects of pollutant exposure than the general population. According to World Health Organization (WHO), the major outdoor air pollution sources include vehicles, power generation, building heating systems, agriculture waste and industry4,34–39. Although ambient air pollution has well-documented adverse

effects on patients with COPD40, the evidence for air pollution causing COPD is not

conclusive41. Moreover, the emphysema seen with tobacco smoking has not been

described as being causally linked to ambient air pollution, thus, it is essential to further establish any links between air pollution and COPD.

Current treatments for COPD

As COPD is affecting around 300 million people globally and representing a leading cause of death worldwide, COPD is a clinical problem with increasing interests. The therapeutic intervention with the greatest impact on COPD is smoking cessation. Inhaled corticosteroids (ICS) and acting bronchodilators, including long-acting b-agonists (LABA) and long-long-acting muscarinic antagonists (LAMA) are the mainly used as pharmacological therapy to prevent acute exacerbations of COPD, reduce symptoms and minimize the rate of lung function decline. A combination of inhaled drugs is often required for symptomatic patients, and the combination of LAMA/LABA is proven to be more effective at reducing exacerbations than either bronchodilator alone. The combination of ICS/LABA inhaler also has been used in patients with exacerbations42. Additionally, triple inhaler therapy (ICS/LABA/LAMA)

(7)

is shown to be superior compared with dual combinations in reducing the risk of moderate or severe COPD exacerbations. At present, Budesonide43–45 is a commonly

used medication to treat lung inflammation that is believed to remain primarily in the airways, however, it is associated with side effects such as bruising easily, headache and diarrhea increase. Roflumilast, the only phosphodiesterase 4 (PDE4) inhibitor46 currently approved for the treatment of COPD has also been approved as

an alternative maintenance treatment in COPD patients47. It is an oral medication

that decreases inflammation in the lungs, but still is associated with adverse effects such as headaches and gastrointestinal side effects. In addition, GOLD (Global Initiative for Chronic Obstructive Disease) recommends as alternative option to use an antioxidant agent such as N-acetylcysteine, which is a thiol-containing drug that functions as a mucolytic and an anti-inflammatory drug and for which modest effects on reducing COPD exacerbation frequency have been reported48–51. All these

pharmacological therapies for COPD are used to reduce symptoms, the frequency and severity of exacerbations, and improve exercise tolerance and health status; however, there is no medication that effectively modifies the long-term decline in lung function currently.

Lung repair in COPD

Humans can survive only for several minutes without oxygen that is contained in the air and the lungs are the main organ of the respiratory system and function to exchange gas, taking up oxygen and removing carbon dioxide52. The respiratory

system is proximally to distally anatomically composed of the trachea, bronchi, bronchioles and alveoli, which is the main site for gas exchange and host defense functions. The upper airway epithelium consists mainly of ciliated cells, club cells, goblet cells and undifferentiated basal cells, whereas the alveolar epithelium consists mainly of a layer of alveolar epithelial type 1 and type 2 (AT1 and AT2) cells53,54. The

pulmonary anatomical and cellular composition of mice and humans share both similarities and critical differences with respect to structure and function55. Based

on the same cell specific marker genes, many cellular counterparts of the murine lung have been identified in the human lung such as AT2 cells55–57.

The lung epithelial cells are in direct contact with the outside environment. They are exposed to inhaled particles as well as pathogens and once damaged, these epithelial cells must be replaced rapidly by the resident stem or progenitor

(8)

1

populations. Understanding the populations of stem cells activated under different injury conditions and understanding the role of lung resident stem or progenitor cells in the maintenance of the epithelium during lung homeostasis and repair should provide novel insights into therapeutic approaches for treating COPD. Alveolar repair

Alveolar epithelial type 2 (AT2) cells are playing essential roles in response to alveolar injuries as the main progenitors within the alveolar niche. About 95% of the alveolar surface area is covered by flat AT1 cells that die by apoptosis upon injury and the cuboidal AT2 cells are more resistant to injury. Once AT1 cells are injured, adjacent AT2 cells are triggered to self-renewal and transdifferentiate into AT1 cells58. In addition to AT2 cells, the alveolus also contains a myriad of mesenchymal

and myeloid cells that help alveolar repair. Both alveolar epithelial cells and the surrounding mesenchymal cells (Figure 2) seem to upregulate ligands and receptors in response to injury, allowing for increased, directed cell-cell interactions in the alveolar niche58–60.

Figure 2. Scheme of lung alveolar structure covered by different types of cells.

Cellular signaling pathways regulating lung repair

It is likely that activation of multiple molecular signaling pathways is required to stimulate lung regeneration61. Molecular signaling pathways, such as WNT, FGF,

TGF-b/BMP signaling pathways, have been identified to promote alveolar epithelial cell proliferation and differentiation62,63. Understanding the molecular regulation of

alveolar regeneration is of high clinical importance as it may generate strategies for drug development.

(9)

WNT signaling pathway in lung repair

WNT signaling has emerged as a fundamental growth control pathway that drives most types of tissue stem cells in adult mammals64. This signaling pathway contains

a large family of conserved secreted proteins activating intracellular signaling cascades, which is classically categorized in two types based on the downstream effectors of those proteins: 1) The canonical WNT signaling pathway (b-catenin dependent); and 2) The non-canonical WNT signaling pathway (b-catenin independent)62,65,66. WNT proteins (19 in human) comprise an important class of

glycoproteins that are essential ligands for repair activating the signaling by binding specific cell surface receptors including frizzled (FZD), and lipoprotein receptor related proteins (LRP), as well as atypical receptors, such as the receptor tyrosine kinase-like orphan receptors (ROR)67–69.

Accumulating evidence demonstrates that WNT/b-catenin signaling regulates progenitor cell fate decision during lung development as well as after lung injury62,70.

Specifically in the distal lung, WNT signaling plays a critical role in the development of both the surfactant-producing alveolar epithelial type 2 (AT2) cells and the AT1 cells that for the gas-exchange surface of the lung alveolus56,71–73. Although the WNT

signaling pathway is involved in the promotion of the proliferation and expansion of regenerative cell lineages, and is extensively indicated to be activated in response to injury, however, the regeneration of the injured lung seems to be insufficient to repulse some pulmonary pathological conditions74. The canonical

WNT/b-catenin signaling is reduced in the lung epithelium linked to COPD, and enhanced expression of non-canonical WNT-5A has been reported in experimental models of COPD and human COPD tissue specimens75. In addition to WNT-5A, WNT-5B has

been reported increased in airway epithelium of COPD patients76, and the WNT-5B

signaling induces inflammatory responses in human lung fibroblasts77. Moreover,

distal lung epithelial progenitor cells derived from the mouse model of elastase-induced emphysema exhibited reduced organoid forming capacity, whereas the activation of WNT/b-catenin signaling largely restored the reduction78.

FGF signaling pathway in lung repair

Fibroblast growth factor (FGF) signaling is another critical signaling pathway involved in a wide range of biological processes including homeostasis, repair and metabolism. Moreover, niche-derived paracrine signals modulate the behavior of AT2 cells, and FGF signaling is of particular importance. There are 23 known FGFs in both human and mouse that function in a paracrine or autocrine manner binding

(10)

1

one of the FGFRs (FGFR1-4), thereby initiate the signaling pathway79. Among all

FGFs, FGF2, 7, 10 are mostly studied in respiratory studies. FGF2, also known as basic fibroblast growth factor (bFGF), which is a potent mitogen for fibroblasts that has been shown to be overexpressed in severe asthma and COPD80. Conversely,

FGF2 was decreased in the lungs of cigarette smoke-exposed mice in line with a significant decrease in COPD patients81. FGF7 tends to be upregulated in lung tissues

from COPD patients82 but has been reported together with FGF10 as growth factors

secreted from fibroblasts preventing fibrosis and promoting the differentiation and maintenance of the alveolar epithelium83. Intriguingly, it has been demonstrated

that FGF7 alone is insufficient to induce alveolar organoid formation in mesenchymal cell-free organoid culture system, implying additional factors are needed84.

TGF-β/ROCK signaling in lung repair

Aberrant tissue repair and remodeling leading to a disturbed extracellular matrix (ECM) homeostasis are features of COPD development. Lung fibroblasts are the principal cells in ECM homeostasis regulation and transforming growth factor TGF-b is the main inducer of ECM production in lung fibroblasts, which has been reported elevated levels in COPD85. TGF-b has been reported as the most potent factor for

inducing myofibroblast differentiation in fibrotic lungs86–88. Myofibroblasts express

a-smooth muscle actin (a-SMA), which is a marker of activated fibroblasts, and are capable of ECM production including collagen, laminin, and fibronectin86,87.

TGF-b not only regulates fibroblast differentiation, but also regulates epithelial differentiation as well as the epithelial-mesenchymal interactions86. TGF-b induced

myofibroblast differentiation results in WNT/b-catenin pathway skewing and impairs fibroblast ability to support epithelial repair88. Interestingly, overexpressed

TGF-b can activate the Rho/ROCK signaling pathway, which was observed in the occurrence and development of pulmonary diseases89. ROCK1 and ROCK2 are two

isoforms of the Rho-associated coiled-coil-forming protein kinase (ROCK), which play essential roles in many cellular responses to injury such as rearrangements of actin cytoskeleton. Pharmacological inhibition of ROCK using nonselective ROCK inhibitors has been shown to prevent fibrosis in animal models, however, little is known about the specific roles of each ROCK isoform90.

(11)

Pulmonary drug discovery

Many of the new pulmonary drugs that have failed in the clinical due to safety or efficacy. The absence of predictive models and tools to model lung progenitor cell function resulted in the failure of promising drug candidates from animal models to humans91. Most in vitro drug screenings were done on 2D cell-based assays,

however, although cultured cell systems can provide some information, they do not recapitulate the complex interactions between different cell types and tissues in vivo. A considerable unmet medical need remains for defective lung repair in COPD and the current approach to drug discovery is insufficient to meet the needs of the industry and patients. Recently, several new methods have been added to the portfolio of the pharmacologist and cell biologist including lung slices and the organoid, which better recapitulate the progenitor cell and its functions.

Precision-cut lung slices to model lung repair

Precision-cut lung slices (PCLS)92–94 represents an in vitro technology that is becoming

increasingly used in laboratories as an assay to study pulmonary physiology and pathogenesis. The cell types and extracellular matrix composition in the slice accurately reflect the physiological situation, and for that reason, PCLS have an advantage over cell models that are often used in drug development95. PCLS provide

emerging and exciting opportunities to advance chronic lung diseases research and to validate novel therapies, and they can be generated from explanted human lung, diseased human and animal models of disease to explore molecular and cellular interactions in disease, which also reduces the number of animal required95,96.

Crucially, PCLS contain intact alveoli rather than monolayers of one or two cells types; and cell types are present in the same ratios and with the same cell-cell and cell-matrix interactions as in vivo97. To improve translatability understanding,

human PCLS form healthy and diseased patients have been utilized as an ex vivo tool to study pulmonary diseases including asthma, COPD and Idiopathic pulmonary fibrosis (IPF)94. Recently, precision-cut lung slices imaging (PCLSi) is also widely

used, and when in combination with pharmacological treatments, more dynamic interactions between cell-cell and cell-matrix can be easily visualized97.

(12)

1

Organoids model lung repair

Organoids are stem cell derived 3D structures that are supported by an extracellular matrix and contain multiple cell types whose spatial arrangement and interactions mimic those of the native organ98–101. Organoids are self-organizing units that mimic

the physiological state of the organ of origin at the morphological and molecular levels, offering a convenient means to study development, organogenesis and stem niches102–105. The stem cell niche is the microenvironment where stem cells reside,

and it assists stem cells to self-renewal and differentiation for tissue homeostasis and restoration from injury106. Moreover, organoids represent a powerful tool to

study cell-cell interactions107 within the stem cell niche and allow the modeling

of various pathological states of environmental or genetic origin that can be easily manipulated in vitro for the exploration and validation of new therapeutic approaches100,102,108,109. Organoids provide a platform to investigate the stem cell

function in vitro and have been utilized for drug screening as well103. Organoid

drug-screening assays generate reproducible high quality drug sensitivity data that positively correlate with physiological responses and achieve reproducible activity of compounds inhibiting the same target105.

In this thesis, we used a lung organoid assay88,110 of alveolar epithelial cells (EpCAM+/

CD31-/CD45-/cells) co-cultured with mesenchymal fibroblasts embedded in Matrigel based on previous publications60,111,112. The presence of mesenchymal cells

is essential not only for supporting the growth of alveolar epithelial progenitors but also determines whether the effects of added treatments on the progenitor cells behavior are direct or indirect.

Scope of the thesis

As mentioned above, current COPD medications provide insufficient inhibition of lung function decline, and a regenerative pharmacology approach with the potential to slow down lung function decline by promoting repair would be a major step forward. This thesis uses different disease models to mimic individual features of COPD pathology to investigate the mechanisms of either cigarette smoke or air pollution induced defective alveolar epithelial repair. The ultimate goal of this work is to discover novel therapeutic approaches.

(13)

Chapter 2 provides an overview of the methodology as well as the application of Precision-cut lung slices (PCLS) in mice. As PCLS can be treated ex vivo to mimic changes associated with pathological conditions, this model creates an important platform for drug screening and represents a strategy to reduce and refine animal studies.

In addition to the PCLS model, recent advances in 3D culturing technology have demonstrated that organoids represent an ideal system to study repair and regeneration56,108,109,113. Canonical WNT signaling plays a critical role in tissue

development and homeostasis, and WNT-5A signaling as well as WNT-5B signaling have been linked to COPD respecitvely75–77. Thus, in Chapter 3, we established an

organoid assay to investigate the impact of non-canonical WNT signaling mediated by WNT-5A/WNT-5B on both alveolar epithelial progenitors, on fibroblasts and on their crosstalk.

In the organoid co-culture system, we previously found88 that the role of

mesenchymal fibroblasts is essential to support alveolar epithelial growth, and multiple molecular signaling pathways are involved in the crosstalk. Accordingly, myofibroblast differentiation induced by TGF-b negatively impacts their support to epithelial cells89. In chapter 4, we examined two novel ROCK inhibitors on the

organoid model consisting of epithelial cells and TGF-b-induced myofibroblasts with the aim of reversing these negative effects of TGF-b.

Cigarette smoke (CS) has a profound impact on lung functions and therefore we aimed to establish a cigarette smoke exposed lung organoid model in chapter 5. To find novel drug targets for COPD in an unbiased manner, we utilized a transcriptomic-guided drug target strategy to identify potential novel targets that restore lung repair.

As cigarette smoke and air pollution represent the most relevant etiologic factors of COPD, we established a diesel exhaust particle (DEP) exposed organoid model in vitro in chapter 6 and aimed to uncover the mechanisms via which DEP results in defective organoid growth. Moreover, we examined the effects of two antioxidants, NAC and MitoQ on the organoid model after exposure to DEP.

Finally, in chapter 7, the findings within this thesis are summarized and discussed, thereby identify open questions and perspectives for future studies.

(14)

1

References

1. Vasilescu, D. M. et al. Noninvasive imaging biomarker identifies small airway damage in severe chronic obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 200, 575–581 (2019). 2. Hoffmann, R. F. et al. Mitochondrial dysfunction increases pro-inflammatory cytokine

production and impairs repair and corticosteroid responsiveness in lung epithelium. Sci. Rep.

9, 1–10 (2019).

3. Olajuyin, A. M., Zhang, X. & Ji, H. L. Alveolar type 2 progenitor cells for lung injury repair. Cell

Death Discov. 5, (2019).

4. COPD Global Initiative. 2020 Report. Glob. Initiat. Chronic Obstr. Lung Dis. (2020).

5. Rao, W. et al. Regenerative Metaplastic Clones in COPD Lung Drive Inflammation and Fibrosis.

Cell 181, 848-864.e18 (2020).

6. Jamieson, E. et al. Smoking, DNA Methylation, and Lung Function: a Mendelian Randomization Analysis to Investigate Causal Pathways. Am. J. Hum. Genet. 106, 315–326 (2020).

7. Paci, P. et al. Integrated transcriptomic correlation network analysis identifies COPD molecular determinants. Sci. Rep. 10, 1–18 (2020).

8. Jones, R. L., Noble, P. B., Elliot, J. G. & James, A. L. Airway remodelling in COPD: It’s not asthma!

Respirology 21, 1347–1356 (2016).

9. Barnes, P. J. Small airway fibrosis in COPD. Int. J. Biochem. Cell Biol. 116, 105598 (2019). 10. Barnes, P. J. Chronic obstructive pulmonary disease. in Genomic and Precision Medicine:

Infectious and Inflammatory Disease (2019).

11. Bershad, L., Mesmer Jr, D. R. B. & Foley, S. J. M. New England Journal on. Heinonlinebackup.Com 2481–2488 (2005).

12. Hirota, N. & Martin, J. G. Mechanisms of airway remodeling. Chest 144, 1026–1032 (2013). 13. Higham, A., Quinn, A. M., Cançado, J. E. D. & Singh, D. The pathology of small airways disease

in COPD: Historical aspects and future directions. Respir. Res. 20, 1–11 (2019).

14. Habiel, D. M. et al. Divergent roles for Clusterin in Lung Injury and Repair. Sci. Rep. 7, 1–14 (2017).

15. Gohy, S. et al. Altered generation of ciliated cells in chronic obstructive pulmonary disease. Sci.

Rep. 9, 1–12 (2019).

16. Penn, R. B., Nayak, A. P. & Deshpande, D. A. New targets for resolution of airway remodeling in obstructive lung diseases [version 1; referees: 2 approved]. F1000Research 7, 1–14 (2018). 17. Lim, T. K., Ko, F. W. S., Benton, M. J., Berge, M. Van den & Mak, J. Year in review 2016: Chronic

obstructive pulmonary disease and asthma. Respirology 22, 820–828 (2017).

18. Vij, N., Chandramani-Shivalingappa, P., Van Westphal, C., Hole, R. & Bodas, M. Cigarette smoke-induced autophagy impairment accelerates lung aging, copd-emphysema exacerbations and pathogenesis. Am. J. Physiol. - Cell Physiol. 314, C73–C87 (2018).

(15)

19. Cantor, J. O. & Turino, G. M. COPD Pathogenesis: Finding the Common in the Complex. Chest

155, 266–271 (2019).

20. Lin, C. R. et al. S100A8 protects human primary alveolar type II cells against injury and emphysema. Am. J. Respir. Cell Mol. Biol. 60, 299–307 (2019).

21. Gindele, J. A. et al. Intermittent exposure to whole cigarette smoke alters the differentiation of primary small airway epithelial cells in the air-liquid interface culture. Sci. Rep. 10, 1–17 (2020). 22. Scudellari, B. M. THE PANDEMIC ’ S. (2020).

23. Wang, Q. et al. Structural and Functional Basis of SARS-CoV-2 Entry by Using Human ACE2. Cell

181, 894-904.e9 (2020).

24. Kaur, G., Lungarella, G. & Rahman, I. SARS-CoV-2 COVID-19 susceptibility and lung inflammatory storm by smoking and vaping. J. Inflamm. 17, 1–8 (2020).

25. Smith, J. C. et al. Cigarette Smoke Exposure and Inflammatory Signaling Increase the Expression of the SARS-CoV-2 Receptor ACE2 in the Respiratory Tract. Dev. Cell 53, 514-529.e3 (2020). 26. Seimetz, M. et al. NADPH oxidase subunit NOXO1 is a target for emphysema treatment in

COPD. Nat. Metab. (2020).

27. Sundar, I. K., Maremanda, K. P. & Rahman, I. Mitochondrial dysfunction is associated with Miro1 reduction in lung epithelial cells by cigarette smoke. Toxicol. Lett. 317, 92–101 (2019).

28. Manevski, M. et al. Cellular stress responses and dysfunctional Mitochondrial–cellular senescence, and therapeutics in chronic respiratory diseases. Redox Biol. 33, 101443 (2020). 29. Maremanda, K. P., Sundar, I. K. & Rahman, I. Protective role of mesenchymal stem cells and

mesenchymal stem cell-derived exosomes in cigarette smoke-induced mitochondrial dysfunction in mice. Toxicol. Appl. Pharmacol. 385, 114788 (2019).

30. Wooding, D. J. et al. Acute air pollution exposure alters neutrophils in never-smokers and at-risk humans. Eur. Respir. J. 55, (2020).

31. Lee, Y. M., Lee, J. H., Kim, H. C. & Ha, E. Effects of PM10 on mortality in pure COPD and asthma-COPD overlap: difference in exposure duration, gender, and smoking status. Sci. Rep. 10, 1–10 (2020).

32. Kim, S.H.,et al. Herbal Combinational Medication of Glycyrrhiza glabra, A gastache rugosa Containing Glycyrrhizic Acid, Tilianin Inhibits Neutrophilic Lung Inflammation by Affecting CXCL2, Interleukin-17 / STAT3 Signal Pathways in a Murine Model of COPD. Nutients 12, 926 (2020).

33. Moreira, A. R. et al. Chronic exposure to diesel particles worsened emphysema and increased M2-like phenotype macrophages in a PPE-induced model. PLoS One 15, 1–18 (2020).

34. Chau, T. T. & Wang, K. Y. An association between air pollution and daily most frequently visits of eighteen outpatient diseases in an industrial city. Sci. Rep. 10, 1–21 (2020).

35. Bayram, H. et al. Effect of serum on diesel exhaust particles (DEP)-induced apoptosis of airway epithelial cells in vitro. Toxicol. Lett. 218, 215–223 (2013).

(16)

1

36. Gao, N. et al. Lung function and systemic inflammation associated with short-term air pollution exposure in chronic obstructive pulmonary disease patients in Beijing, China. Environ. Health

19, 12 (2020).

37. Gao, N. et al. Short-term effects of ambient air pollution on chronic obstructive pulmonary disease admissions in Beijing, China (2013–2017). Int. J. COPD 14, 297–309 (2019).

38. Song, Q., Christiani, D. C., XiaorongWang & Ren, J. The global contribution of outdoor air pollution to the incidence, prevalence, mortality and hospital admission for chronic obstructive pulmonary disease: a systematic review and meta-analysis. Int. J. Environ. Res.

Public Health 11, 11822–11832 (2014).

39. Qu, F. et al. The hospitalization attributable burden of acute exacerbations of chronic obstructive pulmonary disease due to ambient air pollution in Shijiazhuang, China. Environ.

Sci. Pollut. Res. 26, 30866–30875 (2019).

40. Cattani-Cavalieri, I., Valença, S. dos S. & Schmidt, M. Nanodomains in cardiopulmonary disorders and the impact of air pollution. Biochem. Soc. Trans. 48, 799-811(2020).

41. Berend, N. Contribution of air pollution to COPD and small airway dysfunction. Respirology 21, 237–244 (2016).

42. Duffy, S. P. & Criner, G. J. Chronic Obstructive Pulmonary Disease: Evaluation and Management.

Med. Clin. North Am. 103, 453–461 (2019).

43. Ricci, F. et al. In vitro and in vivo characterization of poractant alfa supplemented with budesonide for safe and effective intratracheal administration. Pediatr. Res. 82, 1056–1063 (2017).

44. Waters, R. C. & Hochhaus, G. Characterization of a dextran-budesonide prodrug for inhalation therapy. Eur. J. Pharm. Sci. 129, 58–67 (2019).

45. Tashkin, D. P., Lipworth, B. & Brattsand, R. Benefit:Risk Profile of Budesonide in Obstructive Airways Disease. Drugs 79, 1757–1775 (2019).

46. Franssen, F. M. E. et al. Personalized medicine for patients with COPD: Where are we? Int. J.

COPD 14, 1465–1484 (2019).

47. Schmidt, M.,et al. Phosphodiesterase isoforms and cAMP compartments in the development of new therapies for obstructive pulmonary diseases. Current Opinion in Pharmacology 51, 34-42 (2020).

48. Ansari, S. F., Memon, M., Brohi, N. & Tahir, A. N-acetylcysteine in the Management of Acute Exacerbation of Chronic Obstructive Pulmonary Disease. Cureus 11, (2019).

49. Breau, M. et al. The antioxidant N-acetylcysteine protects from lung emphysema but induces lung adenocarcinoma in mice. JCI Insight 4, 1–11 (2019).

50. Moitra, S. N-acetylcysteine (NAC) in COPD: Benefits often lost in trials. Qjm 112, 387–388 (2019).

(17)

51. Cazzola, M. et al. Influence of N-acetylcysteine on chronic bronchitis or COPD exacerbations: A meta-analysis. Eur. Respir. Rev. 24, 451–461 (2015).

52. Andrzej C., Mariusz R., ed. Stem cells: Therapeutic Applications. Springer. 13, 261 (2019). 53. Zepp, J. A. & Morrisey, E. E. Cellular crosstalk in the development and regeneration of the

respiratory system. Nat. Rev. Mol. Cell Biol. 20, 551–566 (2019).

54. Basil, M. C. et al. The Cellular and Physiological Basis for Lung Repair and Regeneration: Past, Present, and Future. Cell Stem Cell 26, 482–502 (2020).

55. Basil, M. C. & Morrisey, E. E. Lung regeneration: a tale of mice and men. Semin. Cell Dev. Biol.

100, 88–100 (2020).

56. Zacharias, W. J. et al. Regeneration of the lung alveolus by an evolutionarily conserved epithelial progenitor. Nature 555, 251–255 (2018).

57. Mammoto, A. & Mammoto, T. Vascular Niche in Lung Alveolar Development, Homeostasis, and Regeneration. Front. Bioeng. Biotechnol. 7, 1–16 (2019).

58. Leeman, K. T., Pessina, P., Lee, J. H. & Kim, C. F. Mesenchymal Stem Cells Increase Alveolar Differentiation in Lung Progenitor Organoid Cultures. Sci. Rep. 9, 1–10 (2019).

59. Chu, X. et al. Evidence for lung repair and regeneration in humans: key stem cells and therapeutic functions of fibroblast growth factors. Front. Med. 14, 262–272 (2020).

60. McQualter, J. L., Yuen, K., Williams, B. & Bertoncello, I. Evidence of an epithelial stem/progenitor cell hierarchy in the adult mouse lung. Proc. Natl. Acad. Sci. U. S. A. 107, 1414–1419 (2010). 61. Ng-Blichfeldt, J.-P., Gosens, R., Dean, C., Griffiths, M. & Hind, M. Regenerative pharmacology for

COPD: breathing new life into old lungs. Thorax. 74, 890-897 (2019).

62. Hu, H. H., Cao, G., Wu, X. Q., Vaziri, N. D. & Zhao, Y. Y. Wnt signaling pathway in aging-related tissue fibrosis and therapies. Ageing Res. Rev. 60, (2020).

63. Yan, J. et al. PM2.5 collecting in a tire manufacturing plant affects epithelial differentiation of human umbilical cord derived mesenchymal stem cells by Wnt/β-catenin pathway.

Chemosphere 244, 125441 (2020).

64. Nusse, R. & Clevers, H. Wnt/β-Catenin Signaling, Disease, and Emerging Therapeutic Modalities.

Cell 169, 985–999 (2017).

65. Gómez-Orte, E., Sáenz-Narciso, B., Moreno, S. & Cabello, J. Multiple functions of the noncanonical Wnt pathway. Trends Genet. 29, 545–553 (2013).

66. Katoh, M. Canonical and non-canonical WNT signaling in cancer stem cells and their niches: Cellular heterogeneity, omics reprogramming, targeted therapy and tumor plasticity (Review).

Int. J. Oncol. 51, 1357–1369 (2017).

67. Burgy, O. & Königshoff, M. The WNT signaling pathways in wound healing and fibrosis. Matrix

Biol. 68–69, 67–80 (2018).

68. Kumawat, K. & Gosens, R. WNT-5A: Signaling and functions in health and disease. Cell. Mol. Life

(18)

1

69. Skronska-Wasek, W., Gosens, R., Königshoff, M. & Baarsma, H. A. WNT receptor signalling in lung physiology and pathology. Pharmacol. Ther. 187, 150–166 (2018).

70. Jacob, A. et al. Differentiation of Human Pluripotent Stem Cells into Functional Lung Alveolar Epithelial Cells. Cell Stem Cell 21, 472-488.e10 (2017).

71. Aspal, M. & Zemans, R. L. Mechanisms of ATII-to-ATI cell differentiation during lung regeneration. Int. J. Mol. Sci. 21, (2020).

72. Juul, N. H., Stockman, C. A. & Desai, T. J. Niche Cells and Signals that Regulate Lung Alveolar Stem Cells In Vivo. Cold Spring Harb. Perspect. Biol. a035717 (2020).

73. Abdelwahab, E. M. M. et al. Wnt signaling regulates trans-differentiation of stem cell like type 2 alveolar epithelial cells to type 1 epithelial cells. Respir. Res. 20, 1–9 (2019).

74. Majidinia, M., Aghazadeh, J., Jahanban-Esfahlani, R. & Yousefi, B. The roles of Wnt/β-catenin pathway in tissue development and regenerative medicine. J. Cell. Physiol. 233, 5598–5612 (2018).

75. Baarsma, H. A. et al. Noncanonical WNT-5A signaling impairs endogenous lung repair in COPD.

J. Exp. Med. 214, 143–163 (2017).

76. Heijink, I. H. et al. Cigarette smoke-induced epithelial expression of WNT-5B: Implications for COPD. Eur. Respir. J. 48, 504–515 (2016).

77. van Dijk, E. M. et al. Noncanonical WNT-5B signaling induces inflammatory responses in human lung fibroblasts. Am. J. Physiol. - Lung Cell. Mol. Physiol. 310, L1166–L1176 (2016). 78. Hu, Y. et al. Wnt/β-catenin signaling is critical for regenerative potential of distal lung epithelial

progenitor cells in homeostasis and emphysema. Stem Cells. (2020).

79. Katoh, M. FGFR inhibitors: Effects on cancer cells, tumor microenvironment and whole-body homeostasis (Review). Int. J. Mol. Med. 38, 3–15 (2016).

80. Tan, Y. et al. FGF2, an Immunomodulatory Factor in Asthma and Chronic Obstructive Pulmonary Disease (COPD). Front. Cell Dev. Biol. 8, 1–12 (2020).

81. Kim, Y. S. et al. The role of FGF-2 in smoke-induced emphysema and the therapeutic potential of recombinant FGF-2 in patients with COPD. Exp. Mol. Med. 50, (2018).

82. El Agha, E. et al. Is the fibroblast growth factor signaling pathway a victim of receptor tyrosine kinase inhibition in pulmonary parenchymal and vascular remodeling? Am. J. Physiol. - Lung

Cell. Mol. Physiol. 315, L248–L252 (2018).

83. Correll, K. A. et al. TGF beta inhibits HGF, FGF7, and FGF10 expression in normal and IPF lung fibroblasts. Physiol. Rep. 6, 1–11 (2018).

84. Shiraishi, K. et al. Mesenchymal-Epithelial Interactome Analysis Reveals Essential Factors Required for Fibroblast-Free Alveolosphere Formation. iScience 11, 318–333 (2019).

85. Ong, J. et al. Marked TGF-β-regulated miRNA expression changes in both COPD and control lung fibroblasts. Sci. Rep. 9, 1–11 (2019).

(19)

86. Saito, A., Horie, M. & Nagase, T. TGF-β signaling in lung health and disease. Int. J. Mol. Sci. 19, 1–18 (2018).

87. Kim, K. K., Sheppard, D. & Chapman, H. A. TGF-β1 signaling and tissue fibrosis. Cold Spring Harb.

Perspect. Biol. 10, 1–34 (2018).

88. Ng-Blichfeldt, J. P. et al. Tgf-β activation impairs fibroblast ability to support adult lung epithelial progenitor cell organoid formation. Am. J. Physiol. - Lung Cell. Mol. Physiol. 317, L14– L28 (2019).

89. Zhang, Y., Yuan, R. X. & Bao, D. TGF-β1 promotes pulmonary arterial hypertension in rats via activating RhoA/ROCK signaling pathway. Eur. Rev. Med. Pharmacol. Sci. 24, 4988–4996 (2020). 90. Knipe, R. S. et al. The rho kinase isoforms ROCK1 and ROCK2 each contribute to the development

of experimental pulmonary fibrosis. Am. J. Respir. Cell Mol. Biol. 58, 471–481 (2018).

91. Barnes, P. J. et al. Barriers to new drug development in respiratory disease. Eur. Respir. J. 45, 1197–1207 (2015).

92. Alsafadi, H. N. et al. An ex vivo model to induce early fibrosis-like changes in human precision-cut lung slices. Am. J. Physiol. - Lung Cell. Mol. Physiol. 312, L896–L902 (2017).

93. Lehmann, M. et al. Differential effects of Nintedanib and Pirfenidone on lung alveolar epithelial cell function in ex vivo murine and human lung tissue cultures of pulmonary fibrosis 11 Medical and Health Sciences 1102 Cardiorespiratory Medicine and Haematology 06 Biologica.

Respir. Res. 19, 1–12 (2018).

94. Rosales Gerpe, M. C. et al. Use of Precision-Cut Lung Slices as an Ex Vivo Tool for Evaluating Viruses and Viral Vectors for Gene and Oncolytic Therapy. Mol. Ther. - Methods Clin. Dev. 10, 245–256 (2018).

95. Cedilak, M. et al. Precision-cut lung slices from bleomycin treated animals as a model for testing potential therapies for idiopathic pulmonary fibrosis. Pulm. Pharmacol. Ther. 55, 75–83 (2019).

96. Liu, G. et al. Use of precision cut lung slices as a translational model for the study of lung biology. Respir. Res. 20, 1–14 (2019).

97. Akram, K. M. et al. Live imaging of alveologenesis in precision-cut lung slices reveals dynamic epithelial cell behaviour. Nat. Commun. 10, (2019).

98. Nadkarni, R. R., Abed, S. & Draper, J. S. Organoids as a model system for studying human lung development and disease. Biochem. Biophys. Res. Commun. 473, 675–682 (2016).

99. Sachs, N. et al. Long‐term expanding human airway organoids for disease modeling. EMBO J.

38, 1–20 (2019).

100. Xian, W., Duleba, M., Yamamoto, Y., Vincent, M. & McKeon, F. Biobanking Organoids or Ground-State Stem Cells? J. Clin. Med. 7, 555 (2018).

101. Kim, C. F. Intersections of lung progenitor cells, lung disease and lung cancer. Eur. Respir. Rev.

(20)

1

102. Paschini, M. & Kim, C. F. An airway organoid is forever. EMBO J. 38, 2–4 (2019).

103. Node, T. L., Site, N. & Organogenesis, K. Regenerating a kidney in a lymph node. Pediatr Nephrol.

31, (2015).

104. Schilders, K. A. A. et al. Regeneration of the lung: Lung stem cells and the development of lung mimicking devices. Respir. Res. 17, 1–16 (2016).

105. Wang, J., Li, X. & Chen, H. Organoid models in lung regeneration and cancer. Cancer Lett. 475, 129–135 (2020).

106. Liao, D. & Li, H. Dissecting the Niche for Alveolar Type II Cells With Alveolar Organoids. Front.

Cell Dev. Biol. 8, 1–6 (2020).

107. Nikolić, M. Z. & Rawlins, E. L. Lung Organoids and Their Use To Study Cell-Cell Interaction. Curr.

Pathobiol. Rep. 5, 223–231 (2017).

108. Sundarakrishnan, A., Chen, Y., Black, L. D., Aldridge, B. B. & Kaplan, D. L. Engineered cell and tissue models of pulmonary fibrosis. Adv. Drug Deliv. Rev. 129, 78–94 (2018).

109. Tan, Q., Choi, K. M., Sicard, D. & Tschumperlin, D. J. Human airway organoid engineering as a step toward lung regeneration and disease modeling. Biomaterials 113, 118–132 (2017). 110. Ng-Blichfeldt, J. P. et al. Retinoic acid signaling balances adult distal lung epithelial progenitor

cell growth and differentiation. EBioMedicine. 36, 461-474 (2018).

111. Barkauskas, C. E. et al. Type 2 alveolar cells are stem cells in adult lung. J. Clin. Invest. 123, 3025–3036 (2013).

112. Teisanu, R. M. et al. Functional analysis of two distinct bronchiolar progenitors during lung injury and repair. Am. J. Respir. Cell Mol. Biol. 44, 794–803 (2011).

(21)

XINHUI

Referenties

GERELATEERDE DOCUMENTEN

The studies described in this thesis were performed within the framework of the Groningen University Institute for Drug Exploration (GUIDE), the Groningen Research Institute for

Elastase-induced parenchymal disruption and airway hyper responsiveness in mouse precision cut lung slices: Toward an ex vivo COPD model. Airway and extracellular matrix mechanics

Ex vivo co-culture organoid of murine fibroblasts and EpCAM + cells from TCF/Lef: H2B-GFP mice treated with WNT-5A/5B revealed that both WNT-5A and WNT-5B negatively regulate

We conclude that dual pharmacological inhibition of ROCK 1 and 2 counteracts TGF-β induced effects on remodeling and alveolar epithelial progenitor function, suggesting this to be

Moreover, an adult organoid model exposed to either CS or DEP was established in chapter 5 and chapter 6, which revealed different molecular signaling pathways are regulating

Haoxiao, thanks for inviting me a dinner at your place in Groningen, and also shared life experience and working experience to me.. Congrats on the second princess coming to

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.. Downloaded

Hierdie tipies gereformeerde siening van belydenis, wat ’n direkte uit- vloeisel is van die gereformeerde siening van die lewe voor die aangesig van die lewende, sprekende God en