Download as pdf or txt
Download as pdf or txt
You are on page 1of 21

Received: 17 October 2018 | Revised: 10 February 2019 | Accepted: 14 February 2019

DOI: 10.1002/jcp.28482

MINI‐REVIEW

Identification of biomarkers and genetic approaches toward


chronic obstructive pulmonary disease

Ridhima Wadhwa1 | Taru Aggarwal2* | Vamshikrishna Malyla3,12 | Nitesh Kumar4 |


Gaurav Gupta5 | Dinesh Kumar Chellappan6 | Harish Dureja7 | Meenu Mehta8 |
Saurabh Satija8 | Monica Gulati8 | Pawan Kumar Maurya9 | Trudi Collet10 |
Philip Michael Hansbro11,12,13 | Kamal Dua3,11,12
1
Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi, India
2
Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
3
Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, New South Wales, Australia
4
Amity Institute for Advanced Research & Studies (M&D), Amity University, Noida, Uttar Pradesh, India
5
School of Pharmaceutical Sciences, Jaipur National University, Jagatpura, Jaipur, Rajasthan, India
6
Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
7
Department of Pharmaceutical Sciences, Maharishi Dayanand University, Rohtak, Haryana, India
8
School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
9
Department of Biochemistry, Central University of Haryana, Mahendergarh, Haryana, India
10
Innovative Medicines Group, Institute of Health & Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
11
Priority Research Centre for Healthy Lungs, University of Newcastle & Hunter Medical Research Institute, Newcastle, New South Wales, Australia
12
Centre for Inflammation, Centenary Institute, Sydney, New South Wales, Australia
13
School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia

Correspondence
Ridhima Wadhwa, Faculty of Life Abstract
Sciences and Biotechnology, South Asian Chronic obstructive pulmonary disease accounts as the leading cause of mortality
University, Akbar Bhawan, Chanakya-
worldwide prominently affected by genetic and environmental factors. The disease is
puri, New Delhi 110021, India.
Email: rw4565@gmail.com; characterized by persistent coughing, breathlessness airways inflammation followed
Kamal Dua, Discipline of Pharmacy, by a decrease in forced expiratory volume1 and exacerbations, which affect the
Graduate School of Health, University of
quality of life. Determination of genetic, epigenetic, and oxidant biomarkers to
Technology Sydney, P.O. Box: 123
Broadway, New South Wales 2007, evaluate the progression of disease has proved complicated and challenging.
Australia. Approaches including exome sequencing, genome‐wide association studies, linkage
Email: Kamal.Dua@uts.edu.au
studies, and inheritance and segregation studies played a crucial role in the
Funding information identification of genes, their pathways and variation in genes. This review highlights
National Health Medical Research Council,
multiple approaches for biomarker and gene identification, which can be used for
NHMRC, Grant/Award Number: 1079187
differential diagnosis along with the genome editing tools to study genes associated
with the development of disease and models their function. Further, we have
discussed the approaches to rectify the abnormal gene functioning of respiratory
tissues and various novel gene editing techniques like Zinc finger nucleases (ZFN),
transcription activator‐like effector nucleases (TALEN), and clustered regulatory
interspaced short palindromic repeats/CRISPR‐associated protein 9 (CRISPR/Cas9).

*Taru Aggarwal contributed equally.

J Cell Physiol. 2019;1–21. wileyonlinelibrary.com/journal/jcp © 2019 Wiley Periodicals, Inc. | 1


2 | WADHWA ET AL.

KEYWORDS
biomarkers, cellular therapy, chronic obstructive pulmonary disease, CRISPR/Cas‐9, genome
editing, oxidative stress

1 | INTRODUCTION Bracke, 2011; Ezzati Givi, A Redegeld, Folkerts, & Mortaz, 2012).
Thus, the disease is characterized by both innate and adaptive
According to the World Health Organization (WHO), chronic immune response. Spirometry based diagnoses are used for
obstructive pulmonary disease (COPD) is defined by chronic identification of COPD that gives permanent decline in forced
obstruction of airways interfering with normal lung breathing expiratory volume (FEV) per second along with the ratio of FEV1 to
which is not completely reversible. It is characterized by forced vital capacity (FVC) that is FEV1/ FVC. The FEV1 and FVC
emphysema, airway fibrosis, and chronic bronchitis, which ob- give a relationship to dose‐response relationship to exposure to
struct the normal physiological breathing due to multiple factors smoke. Depending on post‐bronchodilator FEV1 GOLD classifies
like genetic, environmental, and cigarette smoking (World Health COPD into 4 stages namely: GOLD1 is mild COPD with FEV1 > =
Organization, 2019). Global obstruction for chronic lung disease 80%, GOLD2 is moderate COPD with 50% = < FEV1 < 80%, GOLD3
(GOLD) report 2019, defines COPD as prevalent, preventable, and is severe COPD with 30% = < FEV1 < 50% and GOLD4 is very severe
treatable disease distinguished by persistent respiratory symp- COPD with FEV1 < 30% (Mapel et al., 2015). However, this indicates
toms and airflow limitation because of airways/alveolar anomaly that the environmental factors and cigarette smoke and genetic
due to noxious particles and gases exposure (Global Initiative for constituents are responsible to cause COPD. The genetic variations
Chronic Obstructive Lung Disease, 2019).The disease is associated of α‐1 anti‐trypsin (AAT) gene serpin peptidase inhibitor, clade A,
with dyspnea, cough or sputum production obstruction of airflow, member 1 (SERPINA1) have been regarded to be the major
inflammation in the lungs, emphysema, and tissue tear down contributing factor for disease pathogenesis but AAT deficiency
(Global Initiative for Chronic Obstructive Lung Disease, 2019; has been interpreted up to 2% of all the causes of COPD over 45
Mapel, Dalal, Johnson, Becker, & Hunter, 2015). Worldwide, COPD years (Berndt, Leme, & Shapiro, 2012). Therefore, it is essential to
is the fourth leading cause of mortality and by 2020, it is expected decipher the genetic and environmental causes of COPD as it
to be the third major cause of death (Global Initiative for Chronic involves both epigenetic factors along with somatic level mutations
Obstructive Lung Disease, 2019). According to the WHO report in (Tzortzaki et al., 2012; Yao & Rahman, 2012).
2016, 251 million cases of COPD along with 3.15 million deaths This review intent to emphasize the role of genes and biomarkers
per year have been reported (Mathers & Loncar, 2006). In low and involved in COPD along with the current approaches used to treat
middle income countries the burden of disease is much higher due the disease and how the genome editing approach can be used as
to environmental pollution and smoking, which have significant therapeutic for treating the disease.
association in progressive symptoms and functional impairment of
airways due to accumulation of oxidative and carbonyl stress
(Banerjee, 2014; Lopez et al., 2006; McGuinness & Sapey, 2017; 2 | AIRWA YS B IOMA RK ERS O F
World Health Organization, 2017). O XI D A T I V E S T R E S S I N CO P D
COPD is not only caused by environmental factors but also the
genetic predisposition of an individual (Kennedy, Chambers, Du, & The environmental risk factors including air pollution, dust, and
Dimich‐Ward, 2007). It is systematic inflammation predominantly fumes, smoke from biomass combustion increases the probability of
marked in lung parenchyma and peripheral airways (Barnes, 2016). disease in nonsmokers (Mannino & Buist, 2007). While cigarette
The chronic obstruction of lung airways is majorly due to smokers are highly susceptible to disease as smoke from a cigarette
disintegration and fibrosis of airways owing to lung parenchyma contains 1017 oxidants per puff (Pryor & Stone, 1993). This leads to
rigidity which traps air in an irreversible manner (Barnes, 2014). direct exposure of lung epithelial cells to inflammation in neutrophils,
Only, the cholinergic contractions in the small airways are macrophages, and lymphocytes. In response to the inflammation
reversible in nature. Hypersecretion of mucus is also responsible caused, antiproteases and antioxidant factors are released to
for the obstruction of airways, thereafter occupying the lumen counterbalance the damage by reactive oxygen species (ROS) and
space rendering ciliary dysfunction causing accelerated seizure of proteolytic enzymes (Larsson, 2007). ROS includes superoxide anion
lung function (McDonough et al., 2011). The inflammatory mechan- (O2−), hydroxyl radical (OH−) along with other derivatives of oxygen
ism is found similar to that of smokers but is amplified by airways with unpaired electrons like hydrogen peroxide (H2O2). ROS
obstruction in patients with COPD. In such conditions, airway formation is a continuous process as a result of metabolic activities
lumen recruits a large number of macrophages and neutrophils in the cell. However, a great amount of ROS is produced by
while airway walls & parenchyma renders increased levels of neutrophils and macrophages to encounter the inhaled particles
macrophages, T lymphocytes, and B lymphocytes (Brusselle, Joos, & (Rahman, Biswas, & Kode, 2006). The oxidative stress produced
WADHWA ET AL. | 3

F I G U R E 1 Source and mechanism of oxidative stress in chronic obstructive pulmonary disease. Oxidative stress (OS) can be due to the
endogenous and exogenous source. Oxidative stress is a major factor leading to chronic obstructive pulmonary disease (COPD). The exogenous
sources of OS are air pollutants (NO2, SO2), cigarette smoke and infectious agents including bacteria, fungus, and viruses. While the endogenous
sources of OS are inflammatory cells and mitochondrial dysfunction. In case of COPD, oxidative stress stimulates mucus hypersecretion,
inflammation, oxidation of extracellular matrix (ECM), cellular leakage, upregulation of factors including chemokines, tumor necrosis factor–ß (TNF‐ß),
nuclear factor kappa‐light‐chain‐enhancer of activated B cells (NFκB) therefore leading to cell senescence. Furthermore, disintegrates the
homeostasis within the airways by elevated secretion of proteases, elastase, cytokines, perforins, and granozymes, which causes the production of
carbonyl modified autoantibodies, cytotoxicity, and decline in phagocytosis activity [Color figure can be viewed at wileyonlinelibrary.com]

damages lipids, DNA, and protein as represented in Figure 1. ROS are 2.2 | Protein oxidation products
highly reactive in nature so they have a short half‐life, their level
The most abundant oxidative damage product of proteins is protein
can be directly estimated in the body and tissue fluids by
carbonylation (Berlett & Stadtman, 1997). These carbonyl derivatives
measuring target products of oxidation such as oxidized proteins,
are formed by oxidation of lysine, arginine, threonine, and proline
lipid peroxidation, antioxidants, and so forth (Dalle‐Donne, Rossi,
residue side chains. Proteins can react directly to ROS to produce
Colombo, Giustarini, & Milzani, 2006).
peptide fragments with a carbonyl group. The common method of
detection of carbonylation of proteins is with 2,4‐dinitrophenylhy-
2.1 | Lipid peroxidation drazine (DNPH) which forms dinitrophenylhydrazone (DNP; Levine,
2002). DNP can be estimated spectroscopically at 370 nm or by
Oxidative stress leads to lipid peroxidation causing oxidative damage
immunochemistry with a specific antigen (DNP)‐antibody reaction.
(Niki, 2009). Oxidation of lipids occurs readily by nonenzymatic and
Studies have identified an increase in protein carbonyl level in plasma
enzymatic oxidants. Polyunsaturated fatty acids are prone to free
of patients with COPD as compared to healthy control. The level of
radical oxygen producing aldehydes such as lipid hydroperoxides as
protein carbonyl increases with disease progression (Santos
the product (Iuliano et al., 2003). Malondialdehyde (MDA) and
et al., 2004).
thiobarbituric acid reactive substances (TBARS) are the most
commonly used biomarkers for lipid peroxidation (Iuliano et al.,
2003). In the TBARS method, MDA reacts with thiobarbituric acid
2.3 | Reactive oxygen species
(TBA) at acidic pH a stable chromophore with maximum absorbance
at 586 nm (Jammes, Steinberg, Ba, Delliaux, & Brégeon, 2008). Also, Several authors have studied the level of O2− in ROS production.
the product of the reaction can be examined by using high Chemiluminescence‐based method of estimation of O2− has been
performance liquid chromatography (HPLC). Spectroscopic and used to identify the diseased state. An enzymatic method involving
HPLC analysis infers that the level of lipid peroxidation is high in superoxide dismutase inhibitable reduction of cytochrome c‐ and
COPD cases compared to healthy controls (Sünnetçioğlu, Alp, nitro‐blue tetrazoliun for reduction of O2− to form diformazan,
Sertogullarından, Balaharoglu, & Gunbatar, 2015). which can be measured spectroscopically at 550 nm (Hanta,
4 | WADHWA ET AL.

Kocabas, Canacankatan, Kuleci, & Seydaoglu, 2006; Nadeem, Raj, GSH/GSSH ratio significantly decreases during COPD exacerbation
& Chhabra, 2005). in comparison to stable COPD (ben Anes et al., 2014).

2.4 | Oxidatively damaged DNA 2.8 | Total antioxidant capacity

Oxidation of DNA can be studied by a single cell gel electrophoresis Most commonly the ferric reducing ability of plasma (FRAP) and the
or comet assay that allows the visualization of DNA breaks. DNA trolox equivalent antioxidant capacity (TEAC) assay have been used
breaks move out of supercoiled DNA loops to form a comet‐like tail. to determine the total antioxidant capacity in the biological samples.
The DNA tail gives an estimate of breaks in DNA. This assay is The principle of the FRAP assay is the reduction of ferric
reliable for the detection of COPD as a significant increase in DNA tripyridyltriazine (Fe3 + ‐TPTZ) complex to the ferrous tripyridyltria-
damage is observed in COPD compared with the healthy individual zine (Fe2 + ‐TPTZ) with the formation of blue color, observed at
(ben Anes et al., 2014; Lin, Wu, Chen, Hsieh, & Yeh, 2010). 593 nm. The TEAC assay works by inhibiting antioxidants of the
absorbance of the radical cation of 2, 2‐azinobis (3‐ethylbenzothiazo-
line 6‐sulfonate; ABTS), which has maximal absorption at 660, 734,
2.5 | Uric acid and 820 nm. ABTS radical cation is formed by reaction of ABTS with
peroxidase metmyoglobin and H2O2 and on the addition of plasma,
Uric acid is an essential antioxidant for the protection of lipoproteins
the color change reaction is avoided by suppressed antioxidant
from oxidation as it acts scavenger of hydroxyl and oxygen free
activity. In COPD, both the assays infer a significant decrease in the
radicals. The levels can be estimated by an enzymatic method using
total antioxidant capacity (ben Anes et al., 2014).
spectroscopy and HPLC, which suggests a significant decrease of uric
acid in COPD (Aggarwal, Wadhwa, Rohil, & Maurya, 2018; Nicks,
O'Brien, & Bowler, 2011). 2.9 | Enzymatic antioxidant activity
Antioxidant enzyme activity includes peroxidase (GSH‐Px), glu-
2.6 | Antioxidant nutrients tathione‐S‐transferase (GST), paraoxonase 1 (PON1), and ceruloplas-
min ferroxidase have been studied in patients with COPD. Refer
Plasma levels for antioxidants like vitamin A, C, E, and carotenes can be
Table 1 for a detailed list of antioxidant activity in COPD.
estimated spectrophotometric methods. A significant decrease in
vitamin A, C, and E levels have been found in case of COPD
exacerbation, these levels can be restored after exacerbation treatment
3 | TH E G ENE TIC A PPROAC H
(ben Anes et al., 2014; Woźniak, Górecki, Szpinda, Mila‐Kierzenkowska,
TOWARDS C OPD
& Woźniak, 2013). Along with, a significant decrease in α and ß
carotenes is also found in patients with COPD in comparison to healthy
COPD is a complex disease, which is caused due to environmental
controls. Trace elements are also an important component to maintain
influence or cigarette smoke, but the relation between genes
oxidant‐antioxidant balance. The composition can be studied by X‐ray
interacting with the environment is partially known. Multiple factors
emission and mass spectroscopy. Studies reveal that plasma levels of
or genes contribute to COPD development where few alterations can
zinc (Zn), selenium (Se), and potassium (K) are reduced while an increase
be due to epigenetic modification and others can be due to the
in iron (Fe), calcium (Ca), copper (Cu), and rubidium (Rb) has been
genetic predisposition. The disease progression or pathophysiology is
observed (Santos et al., 2004).
difficult to study as it is complex to identify genes which are
epigenetically modified and predisposed genetically. Therefore, to
identify predisposed genes to epigenetic modifications several
2.7 | Protein and nonprotein thiols
studies are carried out among various population groups which
Thiols are organic compounds containing sulfide group (−SH) which include family and segregation studies, linkage studies, candidate
are important for maintaining the redox balance. Intracellular thiols gene association studies, meta‐analysis studies, genome wide analysis
like glutathione (GSH) maintain a highly reduced niche in the cell and studies, and whole genome and exome sequencing. With the help of
the extracellular thiols maintain antioxidant defense system (Turell, developing technology for diagnosis such as computer tomography
Radi, & Alvarez, 2013). Human serum albumin has highest –SH in a (Kim et al., 2009) or the spirometry, these genotypes can be
plasma comprising of 80% of the reduced thiol groups and is essential correlated phenotypic changes in a precise manner as referred in
in free radical oxygen and nitrogen scavenging (Quinlan, Martin, & Table S1 which summarize the reported and mapped genes
Evans, 2005). Thiols are estimated by Ellman's reagent where free associated with COPD phenotype (DeMeo et al., 2009).
thiols are reduced to form 5‐thionitrobenzoic acid, which is spectro- With the help of high‐throughput techniques such as whole genome
scopically determined at 412 nm (Turell et al., 2013). A significant sequencing, proteomic studies can correlate disease phenotypes to
reduction in protein and non‐ protein thiols have been observed in all genotype. These studies help to identify the proteins which interact in the
stages of COPD in comparison to healthy controls. The total biological system (Von Mering et al., 2006). Figure 2 explains the
WADHWA
ET AL.

T A B L E 1 Antioxidant activity involved in COPD

Enzymatic antioxidant COPD


S.No activity Mode of estimation Mode of action condition Reference
1. Superoxide ELISA, Mc Cord, and Fridovich assay Dismutation of the O2 to molecular oxygen and H2O2. Decline in Quinlan et al., 2005
dismutase (SOD) Xanthine and xanthine oxidase are used to generate O2 and levels
2‐(4‐ iodophenyl)‐3‐(4‐nitrophenol)‐5‐phenyltetrazolium chloride
which reacts with O2 to form a red formazan dye
2. Catalase Spectrophotometry (240 nm), ELISA Detoxification of H2O2 into molecular oxygen and water Not significant ben Anes et al., 2014; Quinlan
et al., 2005
3. GSHPx Oxidation of NADPH Reduction of organic peroxides into alcohols or H2O2 into water Decline in Vibhuti, Arif, Deepak, Singh,
Oxidation of nicotinamide adenine dinucleotide phosphate levels and Pasha (2007)
(NADPH), a coenzyme in the reaction catalyzed by glutathione
reductase that reduces GSSG formed during the activities
catalized by GSHPx
4. GST 1‐Chloro‐ 2,4 dinitrobenzene asa substrate, Inactivates reactive electrophiles by conjugating with GSH Decline in Lakhdar et al. (2011)
spectrophotometry at 340 nm levels
5. PON1 Two‐substrate activity (paraoxon/diazoxon) Hydrolysis of paraoxon protects against the toxicity of some Decline in Isik, Isik, Ceylan, and
method organophosphates and contributes to the antioxidant levels Calik (2005)
protection conferred by HDL on low‐density lipoprotein
oxidation
6. Ceruloplasmin Spectrophotometry (376 nm), Oxidize ferrous ion to ferric ion, complexing with a Elevated levels Healy and Tipton (2007);
ferroxidase Immunoelectrophotometry chromogen Milevoj Kopčinović
et al. (2016)
Abbreviations: COPD: chronic obstructive pulmonary disease; ELISA: enzyme‐linked immunosorbent assay; GST: glutathione‐S‐transferase; PON1: paraoxonase 1
|
5
6 | WADHWA ET AL.

F I G U R E 2 Protein‐protein interaction network in COPD. Genes involved in COPD (as mentioned in Supplementary Table 2) are used to plot
the protein‐protein interaction network using STRING v10: protein‐protein interaction networks, integrated over the tree of life. Szklarczyk D,
Franceschini A, Wyder S, Forslund K, Heller D, Huerta‐Cepas J, Simonovic M, Roth A, Santos A, Tsafou KP, Kuhn M, Bork P, Jensen LJ, von
Mering C. The network created contains 49 nodes, 57 edges, with a PPI enrichment p‐value: < 1.0e‐16 [Color figure can be viewed at
wileyonlinelibrary.com]

interaction of various proteins encoded by the crucial gene involved in 3.1 | Genetic predisposition in COPD
COPD. The interaction network represents protein–protein association
COPD aggregates within families, resulting in genetic predisposition
for both epigenetic and predisposed genes based on experiment,
of several genes as evident from Table 3.
predicted expression, coexpression, text mining, and protein homology
One of the majorly discussed genes in this category is IREB2. IREB2
(Table S2: Represents gene name associated with protein). These
is located on 15q25.1, which encodes for protein iron responsive
reported genes can be mapped for airways measurement and COPD
element binding protein 2, a RNA binding protein involved in iron
diagnosis (Table 2).
metabolism inside the human cell (Rouault, 2006). In COPD situations
Genes involved in COPD (as mentioned in Table S2) are used to plot
such as hypoxemia can arise any time; whereas IREB2 is known as iron
the protein‐protein interaction network using STRING v10: protein–
binding protein hence it remains in the active state in lower oxygen
protein interaction networks, integrated over the tree of life. Szklarczyk
conditions (Rouault, 2006). In COPD the expression of IREB in family
D, Franceschini A, Wyder S, Forslund K, Heller D, Huerta‐Cepas J,
cohorts was found to be differentially expressed, according to DeMeo
Simonovic M, Roth A, Santos A, Tsafou KP, Kuhn M, Bork P, Jensen LJ,
et al (2009) five IREB2 SNPs (rs1964678, rs2656069, rs12593229,
von Mering C. The network created contains 49 nodes, 57 edges, with a
rs965604, rs13180) have been significantly associated with COPD. Risk
PPI enrichment p < 1.0e−16.
T A B L E 2 Pathogenesis of COPD due to genetic predisposition

Gene Study Chromosomal


WADHWA

S.No symbol Gene name Protein name Sample analyzed population location Mechanism genes involved Reference
ET AL.

1. MMP12 Matrix Macrophage Blood 28 11q22.2 Hydrolysis of soluble and insoluble elastin. Aggarwal
metalloproteinase 12 metalloelastase Specific cleavages are also produced at et al. (2018)
14‐Ala‐|‐Leu‐15 and 16‐Tyr‐|‐Leu‐17 in
the B chain of insulin.
2. FAM13A Family with sequence Fam13a protein Lung tissue sample 5 4q22.1 COPD susceptibility by promoting β‐ Jiang
similarity 13, human + 5 catenin degradation et al. (2016)
member A mice
3. IREB2 Iron‐responsive element‐ Iron‐responsive element‐ Blood 248 15q25.1 IREB2 registers cytosolic iron status Ziółkowska‐
binding protein 2 binding protein 2 mainly through an iron‐sulfur switch Suchanek
mechanism et al. (2015)
4. HHIP Hedgehog‐interacting Hedgehog‐interacting Plasma, 20 Mice 4q31.21 Hhiphaploinsufficiency leads to increased Wan et al. (2017)
protein protein urine, and lung susceptibility towards the development
tissue of emphysema following exposure to
chronic cigarette smoke (CS).
5. AGER Advanced glycosylation end Receptor for advanced NETT/NAS, – 6p21.32 RAGE has systemic anti‐inflammatory Kim and Do
product‐specific receptor glycan end GenKOLS, activity Lee (2015)
products (RAGE) ECLIPSE, and
COPD Gene
study cohort
6. CXCL8 C‐X‐C motif chemokine Interleukin‐8 – – 4q13.3 CXCL8 is a member of the chemokine Gilowska (2014)
ligand 8 family and is a major chemoattractant to
neutrophils
7. KCNE2 Potassium voltage‐gated Potassium Blood 9338 21q22.11 Modulates the gating kinetics and Cho et al. (2015);
channel subfamily E voltage‐gated channel enhances the stability of the channel Yang
regulatory subunit 2 subfamily E member 2 complex et al. (2004)
8. HTR4 5‐Hydroxytryptamine 5‐Hydroxytryptamine Lung tissue; fetal 3;12 5q32 This receptor is mediated by G proteins Hodge
receptor 4 receptor 4 tissue that stimulate adenylate cyclase et al. (2013)
9. GSTCD Glutathione Glutathione S‐transferase C‐ Human airway – 4q24 GSTCD lacks key functional domains Obeidat
S‐transferase C‐terminal terminal domain‐containing smooth muscle important for GST (Glutathione S‐ et al. (2013)
domain‐containing protein cells transferase) activity
10. TNS1 Tensin 1 Tensin‐1 Human airway – 2q35 TNS1 gene is associated with airflow Stylianou, Clark,
smooth muscle obstruction in GWAS and
cells Bradding,
(2016)
(Continues)
|
7
8 | WADHWA ET AL.

can be prevented by utilizing gene editing tools by modifying the genes

Cho et al. (2014)


which are involved in the diseased state.

et al., (2011)
et al. (2011)
Reference

Hersh

Kong
3.2 | Epigenetic modifications in COPD
Epigenetic modifications refer to alteration in the gene expression

the effect of BICD1 in emphysema could


interleukin‐2 dependent T‐cell growth.

pattern which may be due to histone modification, DNA methylation,


SOX5 SNPs have been associated with
pharmacogenetic potentially relevant

be related to shortened telomeres


TGF‐β 2 has suppressive effects on

gene silencing by Noncoding RNA, an aberration in chromatin and


DNA occurring due to environmental influences (Handy, Castro, &
Loscalzo, 2011). Evidence suggested that epigenetic changes asso-
Mechanism genes involved

ciated with COPD include DNA methylation at specific CpG Islands


as mentioned in Table 3, these studies are based on analysis of blood
sample for genotyping (Aggarwal, Wadhwa, Thapliyal et al., 2018). A
major cause of epigenetic changes in bronchial epithelium genes is
for COPD

suggested to be due to environmental stress or cigarette smoke


(Schamberger, Mise, Meiners, & Eickelberg, 2014) and the epigenetic
pathways regulate inflammation in the airways. Features of COPD
can be highlighted by epigenetic variation study, which could not be
Chromosomal

fully inferred by DNA sequencing technology, GWAS or meta‐


12p11.21

analysis. This study could help to identify susceptibility developed in


12p12.1
location
1q41

smokers, identification of post smoking cessation and continued risk


to develop COPD.
population
6663; 15
Study

3.2.1 | Epigenetic mechanism of COPD


2380
386

There are three levels of epigenetic gene regulation: DNA modifica-


Blood; Lung tissue
Sample analyzed

tion, histone modification and transcriptionally packed chromatin or


noncoding RNAs. These modifications are encountered and studied in
COPD models or patients (Schamberger et al., 2014).
Blood

Blood

DNA methylation in COPD


Transforming growth factor

Transcription factor SOX‐5

Typically, DNA methylation act by either gene activation or


repression. GWAS of large scale DNA methylation study indicates
Protein bicaudal D

349CpGs, to be significantly associated with COPD (Qiu et al., 2012).


It also suggests that several genes involved in COPD, are also in the
Protein name

homolog 1

inflammation pathway, stress response and healing wound. Gene


SERPIN 1 studied in a population of 1481 subjects where the
β‐2

genomic analysis DNA was extracted from white blood cells is found
in hypomethylation state in COPD patients, indicating gene activa-
Transforming growth factor

tion or over expression (Qiu et al., 2012). Several other studies


BICD cargo adapter 1

established DNA methylation in COPD for a variety of genes. One


such example is for hypomethylation of gene encoding‐1‐antitrypsin,
which is in significant correlated in COPD (Qiu et al., 2012; Siedlinski
Gene name

SRY‐box 5

et al., 2012). Gene expression level alters when DNA methylation


occurs in smoking related blood peripheral leukocytes and small cell
β‐2

airways, this leads to changes in GPCR15 (G Protein Coupled


(Continued)

Receptor) and NRF 2 (nuclear factor erythroid 2‐related Factor 2;


Vucic et al., 2014; Wan et al., 2012). Smoke related animal model
symbol
TGFB2

BICD1
SOX5
Gene

study on mice lung tissue sample and human BEAS‐2B lung epithelial
TABLE 2

cells reveals that the expression of HDAC6 increased hypomethyla-


tion in COPD, which can be linked to several other diseases as well
S.No
11.

12.

13.

(Lam et al., 2013).


T A B L E 3 Pathogenesis of COPD due to epigenetic modifications

Modifica-
WADHWA

tion Gene Population Chromosomal Mechanism genes


S.No Gene reported expression Protein Approach with COPD SNPs location involved Reference
ET AL.

1. SERPINA Hypome- Under α 1‐ antitrypsin Pi system 2; 1086 rs8004738 14q32.13 Protease inhibition DeMeo and Silverman,
1 thylation Expressed (electrophoresis), rs28929474 (2004); Yuan, Chang
GWAS and Deng (2009);
Eriksson, (2018); Li
et al. (2017)
2. SERPIN 2 N.A. Under Protease inhibitor Illumina array‐based 973 rs6734100 2q36.1 Protease inhibition Zhu et al. (2007)
Expressed 7; PI7 custom SNP rs729631
genotyping platform rs975278
rs7583463
rs6748795
3. CHRNA7 N.A. Over Neuronal GWAS 2614 4‐copy of CNV‐3956 15q13.3 Ligand‐gated ion Wang, Shi, Zhu, Gao,
Expressed acetylcholine increased COPD risk channel and Zhang, (2017)
receptor subunit
α‐7
4. CHRNA5 Hyperme- N.A. Neuronal GWAS and meta‐ 2614 rs17486278 15q25.1 Causes opening of Morrow et al. (2016);
thylation acetylcholine analysis. an ion‐conducting Nedeljkovic
receptor subunit channel across the et al. (2018)
α‐5 plasma membrane.
5. CHRNA3 Hyperme- N.A. Neuronal GWAS and meta‐ 2614 rs12914385 15q25.1 Causes opening of Cho et al. (2014); Lee
thylation acetylcholine analysis. an ion‐conducting et al. (2015);
receptor subunit channel across the Nedeljkovic
α‐3 plasma membrane. et al. (2018)
6. EGLN2 N.A N.A Egl nine GWAS 724 subjects, rs7937 19q13.2 Regulates Nedeljkovic
homolog 2 both blood and posttranscriptional et al. (2017)
lung tissue modifications of
sample hypoxia induced
factor
7. MMP1 N.A N.A Collagenase I Expression analysis in 63 N.A. 11q22.2 Cigarette smoke Gosselink et al. (2010);
transgenic mice responsive (CSR) Houghton (2015);
element within the Woode, Shiomi, and
promoter region of D'Armiento, (2015)
mmp‐1
8. MMP12 N.A N.A Fragments of Gene‐association 8300 rs626750 11q22.2 Elastase activity Houghton, (2015);
elastin study Hunninghake
et al., (2009)
(Continues)
|
9
10

TABLE 3 (Continued)
|

Modifica-
tion Gene Population Chromosomal Mechanism genes
S.No Gene reported expression Protein Approach with COPD SNPs location involved Reference
9. VHL Hypome- Over von Hippel‐ Gene polymorphism 164;15 rs11549467 3p25.3 Involved in the Vucic et al. (2014); Yu,
thylation expressed Lindau disease study rs11549465 ubiquitination and Wang, and
tumor subsequent Cheng (2017)
suppressor proteasomal
degradation via the
von Hippel‐Lindau
ubiquitination
complex
10. MUC13 Hypome- Over Mucin−13 Illumina 15 N.A. 3q21.2 Epithelial and Vucic et al. (2014)
thylation expressed Infinium 27 K hemopoietic
Methylation Arrays, transmembrane
San Diego, CA mucin that may play
a role in cell
signaling.
11 NFE2L2 Hyperme- Under Nuclear factor genotyped at K‐ 15;1390 rs1806649 2q31.2 Transcription Figarska, Vonk, and
thylation expressed erythroid 2‐ Bioscience (UK) activator that binds Boezen, (2014); Vucic
related Factor 2 to antioxidant et al. (2014)
response (ARE)
elements in the
promoter regions of
target genes
12 MMP14 Hyperme- Under matrix TaqMan Genotyping 15;284 rs1003349rs1042703- 14q11.2 Endopeptidase that Hadchouel et al.
thylation expressed metallopepti- rs2236302r- degrades various (2008); Vucic
dase 14 s1042704rs2236303- components of the et al. (2014)
rs2236307 extracellular matrix
such as collagen.
Activates
progelatinase A.
13 CD9 Hyperme- Under CD9 antigen Illumina 15 N.A. 12p13.31 Involved in platelet Vucic et al. (2014)
thylation expressed Infinium 27 K activation and
Methylation Arrays, aggregation.
San Diego, CA Regulates paranodal
junction formation.
Involved in cell
adhesion, cell
motility and tumor
metastasis.
Required for sperm‐
egg fusion.
(Continues)
WADHWA
ET AL.
TABLE 3 (Continued)

Modifica-
WADHWA

tion Gene Population Chromosomal Mechanism genes


ET AL.

S.No Gene reported expression Protein Approach with COPD SNPs location involved Reference
14. LGALS3 Hyperme- Under Galectin‐3 Illumina 15 N.A. 14q22.3 Galactose‐specific Vucic et al. (2014)
thylation expressed Infinium 27 K lectin which
Methylation Arrays, binds IgE.
San Diego, CA
15. TFF3 Hyperme- Under Trefoil Factor 3 Illumina 15 N.A. 21q22.3 Involved in the Vucic et al. (2014)
thylation expressed Infinium 27 K maintenance and
Methylation Arrays, repair of the
San Diego, CA intestinal mucosa.
Promotes the
mobility of epithelial
cells in healing
processes (motogen)
16. PTEN Hyperme- Under Phosphatidylino- Oligo Pool (OPA) by 15;53 rs701848rs1903858 10q23.31 Tumor suppressor. Hosgood III, Menashe,
thylation expressed sitol 3,4,5‐ the Illumina Acts as a dual‐ He, Chanock, and Lan,
trisphosphate 3‐ GoldenGate Assay specificity protein (2009); Shi et al.
phosphatase and phosphatase, (2012); Vucic
dual‐specificity dephosphorylating et al. (2014)
protein tyrosine‐, serine‐ ,
phosphatase and threonine‐
PTEN phosphorylated
proteins
17. MUC1 Hyperme- Under Mucin‐1 Genome‐wide 15 rs4971100 1q22 The α subunit has cell Vucic et al. (2014)
thylation expressed association adhesive
studies (GWAS) properties.The β
subunit contains a
C‐terminal domain
which is involved in
cell signaling,
through
phosphorylations
and protein‐protein
interactions.
18. CKB Hyperme- Under Creatine Kinase B Illumina 15 N.A. 14q32.33 Reversibly catalyzes Vucic et al. (2014)
thylation expressed Infinium 27 K the transfer of
Methylation Arrays, phosphate between
San Diego, CA ATP and various
phosphogens
(Continues)
|
11
12

TABLE 3 (Continued)
|

Modifica-
tion Gene Population Chromosomal Mechanism genes
S.No Gene reported expression Protein Approach with COPD SNPs location involved Reference
19. BNIP3 Hyperme- Under BCL2/adenovirus Illumina 15 N.A. 10q26.3 Apoptosis‐inducing Vucic et al. (2014)
thylation expressed E1B 19 kDa Infinium 27 K protein that can
protein‐ Methylation Arrays, overcome BCL2
interacting San Diego, CA suppression
protein 3
20. TTC3 Hyperme- Under E3 ubiquitin‐ Illumina 15 N.A. 21q22.13 Mediates the Vucic et al. (2014)
thylation expressed protein Infinium 27 K ubiquitination and
ligase TTC3 Methylation Arrays, subsequent
San Diego, CA degradation of
phosphorylated Akt
(AKT1, AKT2 and
AKT3) in the
nucleus
21. SMPD3 Hyperme- Under Sphingomyelin Illumina 15 N.A. 16q22.1 Catalyzes the Vucic et al. (2014)
thylation expressed phosphodiester- Infinium 27 K hydrolysis of
ase 3 Methylation Arrays, sphingomyelin to
San Diego, CA form ceramide and
phosphocholine.
22. CXCL1 Hyperme- Under Growth‐ Illumina 15 N.A. 4q13.3 Chemotactic activity Vucic et al. (2014)
thylation expressed regulated α Infinium 27 K for neutrophils
protein Methylation Arrays,
San Diego, CA
23. EPHX1 Hyperme- Under Epoxide Geneservices Ltd 15; 1017 rs2854450 rs2854451 1q42.12 Biotransformation Chappell et al. (2008);
thylation expressed hydrolase 1 (Cambridge, UK) rs3753658rs1051740 enzyme that Vucic et al. (2014)
using Taqman rs2234922 catalyzes the
assays hydrolysis of arene
and aliphatic
epoxides
WADHWA
ET AL.
WADHWA ET AL. | 13

Histone modification et al., 2012; Ito et al., 2005; Szulakowski et al., 2006). Reduced HDAC is
Inside the cell nucleus, the DNA strand is folded and wrapped around an related to the severity of COPD, with a low reduction in HDAC3 and
octamer complex of a protein consisting histone protein, these combine HDAC5 (Ito et al., 2005). In small airways, the reduced levels of HDAC2
and form a nucleosome. Amongst several histone modifications, the most were found to be more significant in cigarette smoking COPD patients
important is in the amino terminal of H3 and H4 through changes in (Isajevs, Taivans, Svirina, Strazda, & Kopeika, 2011). Accordingly, CXCL8
lysine residue by acetylation and methylation (Grewal & Moazed, 2003). histone acetylation increases in the airway biopsies sample of COPD
Mostly, increased acetylation corresponds with transcriptional activity, patients as a promoting factor for proinflammatory cascade initiation
while decreased acetylation connects with transcriptional inactivation (Isajevs et al., 2011; Ito et al., 2005). One of the reason for emphysema
(Grewal & Moazed, 2003). Moreover, heterochromatin assembly is can be HDAC inhibition, due to the fact that HDAC mechanism play a
strongly associated with methylation of histone H3 lysine 9 (Grewal & pivotal role in adult lungs (Mizuno et al., 2010). When CXCL8 expression
Moazed, 2003). However, few post translational modifications are in is increased, HDAC activity decreases due to oxidative stress with the
connection with gene expression either can be activated or repressed. help of HDAC2 tyrosine nitration and serine phosphorylation(Kode et al.,
2006; Marwick et al., 2004; Marwick et al., 2009) or by S‐nitrosylation of
Histone acetylation/deacetylation HDAC2 cysteine (Malhotra et al., 2011). One of the inhibitors of HDAC,
Gene transcription can be initiated by allowing chromatin remodeling MS‐275 when exposed to smoke and associate with anti‐inflammatory
which regulates gene expression by acetylation of histone in the nucleus cytokine IL‐10 (Leus et al., 2017). In another study, Clarke et al (2008)
to unfold the chromatin structure so that transcription factors and RNA reported that protein kinase C (PKC) β increases TNF‐α‐induced NF‐κB
polymerase can bind to available unfolded DNA (Kouzarides, 2007; transcriptional activity at the promoter of CCL11 through recruiting
Shahbazian & Grunstein, 2007). Multiple inflammatory mediators, p300/CBP‐associated factor and acetylation of histone H4 in human
cytokines, and chemokines are involved in COPD inflammatory response, airway smooth muscle cells. Hence, a response to cigarette smoke
which is generated by inflammatory cascade due to gene response corresponds to the upregulate acetylation of histone (H4) and in
(Aggarwal, Wadhwa, Thapliyal et al., 2018). Histone acetylation control exsmokers upregulated histone H3. Therefore, a misbalance created
these proinflammatory genes through regulating promoters of activator between acetylation and deacetylation increases the probability of COPD
protein (AP)‐1 and transcription factors NF‐kB. These pro‐inflammatory development (P. J. Barnes, 2009).
cytokines play an important role by activating NF‐kB, activated NF‐kB Sirtuins comprise of seven family of proteins SIRT1‐SIRT7, SIRT
has the potential to bind to DNA at specific recognition sequence belonging to NAD + family which are dependent deacetylase, SIRT is
enabling interaction with large coactivator molecules (Lawrence, 2009). known to regulate the mitochondrial functions and proteins while
Coactivator molecule for instance, CRE binding protein, the p300 existing in multiple subcellular locations (Finkel, Deng, & Mostoslavsky,
controls intrinsic activator of HAT. Therefore, the outcome of NF‐kB 2009; Houtkooper, Pirinen, & Auwerx, 2012). Much is not known about
activation is the increased rate of histone acetylation which leads to SIRT, but few proteins are studied for interaction, including transcription
altered inflammatory gene expression, similar is the case with gene factor, cellular proteins, signaling molecule and histone (Finkel et al.,
CXCL8 (P. Barnes, Adcock, & Ito, 2005). Several studies that reported, 2009; Houtkooper et al., 2012). Cigarette smoke causes stress because of
NF‐kB is upregulated in smokers with COPD in different tissue samples the presence of oxidants or aldehydes, this stress causes covalent
and bio‐fluids such as in lung homogenate (Szulakowski et al., 2006), rat modification of SIRT1 which reduces the enzymatic activity and initiates
lungs exposed to smoke (Marwick et al., 2004), blood sample proteasomal degradation (Arunachalam, Yao, Sundar, Caito, & Rahman,
(Zaynagetdinov et al., 2016), BALF fluid (Zaynagetdinov et al., 2016), 2010; Caito et al., 2010). SIRT1 is also known to interact with NF‐kB
and so forth. Moreover, NF‐kB can be activated by oxidative stress and subunit RelA/p65, diminished levels of SIRT1 in the macrophages and
tumor necrosis factor‐α (TNF‐α), over expressing the gene CXCL8 which lungs of cigarette smokers are in correlation to COPD which represents
in turn increases the release of IL‐8 (Kode, Yang, & Rahman, 2006; an increased rate of acetylation and further, increased IL8 release
Rahman, Gilmour, Jimenez, & MacNee, 2002). Cigarette smoke activates (Rajendrasozhan, Yang, Kinnula, & Rahman, 2008; Yeung et al., 2004).
Mitogen and stress activated kinase 1 (MSK1), this MSK1 gets in contact Decreased expression of SIRT1 is observed in the large airways of
with a family member of NF‐kB RelA/p65, leading to acetylation and smokers and in both large and small airways of COPD patients (Isajevs,
phosphorylation which is studied on human bronchial epithelial cells. This Strazda, Kopeika, & Taivans, 2012). Hence, SIRT1 plays a crucial role in
causes post translational modification of histones, phosphoacetylation of the regulation of NF‐kB dependent inflammatory cascade.
histone H3, and acetylation of histone H4 (Sundar et al., 2012).
HDACs regulates proinflammatory cytokines negatively (Adcock, Histone methylation
Tsaprouni, Bhavsar, & Ito, 2007). Histone deacetylation may occur by Apart from histone acetylation and deacetylation, histone methyla-
blocking the transcription of inflammatory genes. This blocking is tion is a complex mechanism where histone methyltransferase (HMT)
achieved as a result of interaction between NF‐kB family member and histone demethylase (HDM) play a pivotal role in the regulation
RelA/p65 with HDAC1 and HDAC2 (Ashburner, Westerheide, & Baldwin, of histone methylation (D. D. Wu et al., 2017). The complexity of
2001). In both cigarette smoker and excigarette smoker patients with histone methylation can be studied as different and opposite
COPD, the activity of HDAC is reduced in samples of alveoli containing functional outcomes of histone methylation of residues such as
macrophages, peripheral blood monocytes, and airway biopsies (Chen lysine or arginine can be obtained, additionally, the intensity of
14 | WADHWA ET AL.

methylation may vary distinctly for the same residues (Mosamma- miRNAs, including miR‐30c, miR‐181d, and miR‐638, whose expres-
parast & Shi, 2010). For for example, Histone H3 lysine 4 sion levels were associated with those of their mRNA targets
trimethylation (H3K4me3) is usually present in active genes while (Schamberger et al., 2014). They also propose that miR‐638 may play
methylations of H3K9 and H3K27 are in close proximity to gene an important role in COPD pathology by responding to oxidative
repression. Till now, limited research is focused on histone methyla- stress, and contribute to accelerated lung aging.
tion mechanism in COPD. In COPD studies on histones, smoke
exposed mouse lungs have been collected and studied using the
3.2.2 | Smoking independent epigenetic variants
spectroscopic method to recognize novel histone markers, it includes
of COPD
acetylation, monomethylation, and dimethylation of histone H3 and
H4 significant residues lysine and arginine (Sundar, Nevid, Friedman, As COPD is a complex disease, several genes are differentially expressed
& Rahman, 2013). Andresen, Günther, Bullwinkel, Lange, and Heine due to epigenetic modification irrespective of no smoking cases. These
(2011) reported the pathophysiological progression of the disease is genes are smoking independent epigenetic variants of COPD. For
in tight coordination with elevated β‐Defensin 1 (DEFB1) mRNA example, Nedeljkovic et al (2017) genotyped 724 individuals on whole
levels are found in association to H3K4me3 significantly identified in blood genomic DNA for association with DNA methylation data in the
bronchial epithelium and BALF obtained from 34 COPD subjects. In region 19q13.2 and conducted epigenome wide association analysis. They
addition, Yildirim et al (2006) found protein arginine methyltransfer- found that gene Egl‐nine homolog 2 (EGLN2) and RAB4B (member RAS
ase 2 (PRMT2) protein level and mRNA are upregulated in the lung oncogene family), are associated with COPD and not associated with
tissue of hypoxic mouse lung tissue that is developing COPD. smoking, consistently associated with differential DNA methylation in
Similarly Hussain et al (2009) reported histone H3K27me1 and blood at four CpG sites. They additionally found methylation is also
histone H3K27me2 can be recognized in the group exposed to associated with gene expression level in blood and in lung tissues
cigarette smoke. Hence, further studies and research is required to (Nedeljkovic et al., 2017). This suggests the importance of epigenetic wide
identify the mechanisms involved in the pathogenesis of COPD. analysis to recognize the epigenetic changes in various genes, which can
be used for diagnosis or as a therapeutic target.
Noncoding RNA
Several recent reports provided experimental evidence suggesting
the involvement of miRNA in inflammatory response via innate and 4 | G E N E E D I T I N G TE C H N O L O G Y
acquired immune response, additionally miRNA is also known to have
a pivotal role in sustaining homeostasis during lung development, The phenomenon by which DNA can be engineered (insertion, deletion or
suggesting indirect association of miRNA in inflammatory lung replacement) to understand the function of the gene. Tools for genome
diseases, including COPD (Ezzie et al., 2011; Oglesby, McElvaney, editing includes conventional homologous recombination (HR) and
& Greene, 2010; O'neill, Sheedy, & McCoy, 2011). Recently several advanced tools including zinc finger nucleases (ZFNs), transcription
authors reported that miRNA generally downregulates as an effect of activator like effector nucleases (TALENs) and clustered regularly
cigarette smoke (De Flora et al., 2012; Izzotti, Calin, Steele, Croce, & interspaced short palindromic repeats (CRISPR)—Cas system which
De Flora, 2009; Pottelberge et al., 2011). Several studies identify potentially can be applied to human pluripotent stem cells (hPSCs)
differentially expressed genes in the lung tissue of COPD patients in wherein the template DNA can be exposed to natural mutations or the
comparison with healthy individuals (De Flora et al., 2012; Ezzie mutations present can be repaired for creating isogenic control (Gaj,
et al., 2011; A. C. Hsu et al., 2017). A. C. Hsu et al (2017) identified Gersbach & Barbas, 2013). ZFNs and TALENs are simple to engineer and
miRNA 125a and 125b involved in inflammation in vivo and ex vivo have low cytotoxicity (Mussolino et al., 2011). CRISRP/Cas‐9 is favorable
studies on 15 COPD subjects, similarly, De Flora et al (2012) because it is based on RNA to guide DNA in comparison to ZFNs and
identified 28 miRNAs in smokers, these miRNAs could be related to TALENs which rely on proteins for specific target DNA. CRISPR/Cas‐9 is
the pathophysiology of COPD (Ezzie et al., 2011). A general more efficient, cost effective, and easy to use technology (Ding et al.,
downregulation of miRNA expression in alveolar macrophages 2013). We shall briefly discuss the gene editing approaches as:
derived from smokers compared to non‐smokers. Significantly lower
expression of miR‐452 has been linked to increased expression of
4.1 | Homologous recombination
matrix metalloproteinase 12 (MMP12), analyzed from BALF of 24
COPD individuals which is known to be an important effector of The method of introducing a specific gene sequence into the host
smoking‐related diseases (Graff et al., 2012; Hunninghake et al., genome is based on homologous recombination or gene targeting. In
2009). Recently Christenson et al. (2013) reported miRNA profiles a cell, when a double stranded DNA breaks, it can be repaired by
are altered with regional emphysema severity and can modulate homologous or nonhomologous end joining (San Filippo, Sung, &
disease‐associated gene expression networks. The authors found that Klein, 2008). Homologous recombination results in crossing over
the pathway gene sets involved in cell surface signaling and ECM leading to genetic recombination or can occur by the introduction of
maintenance are inversely correlated with the expression of multiple a transgene that integrates into the genomic DNA without the
upregulated miRNAs. Particularly, this study identified a subset of assistance of nucleases.
WADHWA ET AL. | 15

4.2 | Zinc figure nucleases (ZFNs) spacer DNA. The unique system was observed in bacteria and archea
as a means of adaptive immunity (Barrangou et al., 2007). Once the
ZFNs consist of DNA binding domain and a DNA cleaving domain. The
bacterium is attacked by phage, initially the bacterium transcribes
binding domain is composed of three–six Cys2‐His2 repeats called zinc
the spacer and palindromic DNA into RNA. RNA III along with Cas 9
finger, which recognizes cumulatively recognizes 9–18 bps of the DNA
cleaves the RNA. The CRISPR RNA forms a dimer with transactivat-
(Porteus & Carroll, 2005). This leads to activation of dimerized
ing crRNA, thus forming a guide RNA (gRNA; Jinek et al., 2012; Mali,
restriction enzyme II, Fok I which cleaves DNA with sticky ends.
Yang et al., 2013). The gRNA directs Cas‐9 for endonuclease activity
Further, ZFNs stimulates DNA repair mechanism endogenously in the
by recognizing 20 bp target which further breaks the double
presence of template donor DNA, which can lead to substitution of
stranded DNA. The Cas‐9 comprises of two domains, a HNH domain
target allele (Urnov, Rebar, Holmes, Zhang, & Gregory, 2010).
with breaks the complementary strand of DNA and RuvC‐like domain
that breaks the noncomplementary DNA strand (Jinek et al., 2012;
4.3 | Transcription activator –like effector Mali, Yang et al., 2013). Thus, by using this technology gRNA can be
nucleases (TALENs) engineered complimentary to DNA sequence allowing simple and
rapid correction of defective genes to restore normal functioning.
TALENs are also composed of DNA binding domain called the TAL
repeats and a cleavage domain of Fok I dimer which cleaves the
double stranded DNA (Boch et al., 2009). TAL repeats contains 33–
35 highly conserved amino acids that target DNA. 12th and 13th 5 | A P P L IC A T I O N S O F G E N E E D I T I N G
amino acids differ and are termed as repeat variable residues (RVD) TECH NOL O GY IN COP D
so that one repeat identifies one nucleotide (Boch et al., 2009).
Variation in RVD of the TAL repeats recognizes DNA sequence Till now very limited research studies are available on the application
specifically. On fusion with DNA generates TALE nucleases that of gene editing technology in COPD (Figure 3).
introduces foreign DNA for gene repair or disrupt endogenous gene A major risk factor for COPD is due to genetic variation in genes
function (Cermak et al., 2011). aggregated in families. Growth differentiation factor 15 (GDF15),
which is a member of TGF‐β family, increases in COPD cigarette
smoker group in airway epithelium and human primary airway
4.4 | Clustered regularly interspaced short epithelial cells (HAEC; Q. Wu, Jiang, & Chu, 2012). Human trachea‐
palindromic repeats (CRISPR)‐Cas system bronchial epithelial cells packaged with CRISPR/Cas9 targeting exon
Clustered regularly interspaced short palindromic repeats is based 1infected by lentivirus, for human the GDF15 gene, results in small
on a repetitive pattern (24–48 base pairs) wherein the DNA mutations such as deletion or insertion in exon 1 of the GDF15 gene
sequence would repeat itself in the reverse orientation after a and this significantly reduces GDF15 mRNA, protein and cellular

F I G U R E 3 Strategies of the implication of genome editing technology in chronic obstructive pulmonary disease. Lung tissues,
bronchoalveolar lavage, and bronchial brushings obtained from patients with COPD are extrapolated for the identification of novel genes by
using microarray and proteomics technology. The identified novel gene can be rectified further by recently developed genome editing
technologies including Zinc finger nucleases (ZFN), transcription activator‐like effector nucleases (TALEN) and clustered regulatory interspaced
short palindromic repeats/CRISPR‐associated protein 9 (CRISPR/Cas9) systems for clinical applications for treatment of COPD. COPD: chronic
obstructive pulmonary disease [Color figure can be viewed at wileyonlinelibrary.com]
16 | WADHWA ET AL.

senescence (Q. Wu et al., 2012). The first application of CRISPR/ utilizing gene editing technology as therapeutics are a delivery
Cas9‐mediated gene editing in HAECs had depicted the possibility of strategy, clinical settings, dose optimization, immune response,
increased research on COPD with gene editing. Another major pharmacokinetic analysis, biochemical analysis, long term effect,
symptom of COPD pathogenesis is airway mucus hypersecretion and so forth laboratory systems are designed such that the targeting
regulated by Mucus secretion regulator SAM‐pointed domain‐ could be studied in the more efficient way. For for example, IREB2 a
containing Ets‐like factor (SPDEF) as key regulator of mucus significant iron binding protein producing gene is dysregulated in
secretion by silencing its expression. Song et al (2017) reported that COPD, using technology such as CRISPR/CAS9 knockouts for
gene SPDEF silencing can be reduced by using ZFPs and CRISPR/ identification of gene importance and metabolism. The genome
dCas platforms, hence decrease in the mucus production in lung editing approach for COPD can be used by targeting the exon in
epithelial cells. Therefore, we can say that epigenetic downregulation 15q25 locus having CHRNA3/CHRNA5 located on the same locus as
of SPDEF possesses a vital role in the generation of a constant effort of IREB2. CHRNA3/CHRNA5 are usually found to be epigenetically
of reduction in mucus, this may act as an upcoming therapeutic modified in COPD (Hardin et al., 2012). These studies may lead to the
approach for patients with excessive mucus secretion. Further DNA development of therapeutically applicable solutions. The authors
targeting systems had been applied for genome editing approach, for would like to mention that gene editing technology has a huge
for example, V64 fused with zinc finger primers as an activator of potential in therapeutic applications of COPD and other respiratory
artificial transcription, which binds to gene promoter of γ‐globin, for disease. Recognition, optimization, and delivery are the three key
developmental silencing of fetal γ‐globin in primary human adult components for the development of gene editing technology
erythroblasts (Wilber et al., 2010). Furthermore, TALENs are for COPD.
designed such that TALEs fuses with DNMT3A or TET1 and targets
Ascl1 in neural stem cells in the promoter region, affecting the
variation in gene expression, and methylation (Lo, Choudhury, 7 | CO NCL USION
Irudayaraj, & Zhou, 2017).
COPD with huge global burden becomes an important disease for
the development of novel therapeutics and diagnostics. In this
6 | LIMITATIONS AND FUTURE review, we discussed the crucial genes and airway stress
PE RSP ECTIVE biomarkers in detail to identify the genes or targets, which can
be used for genome editing approach. Genome editing approach
In recent years, ZFNs, TALENs, and CRISPR/Cas9 are the gene includes upcoming pioneer technologies ZFNs, TALENs, and
editing technologies discovered with increased efficiency. Due to CRISPR/Cas9 to develop the system in which the disease can be
active challenges related to engineering associated with ZFNs and cured by targeting DNA. There are several drawbacks in these
TALENs, CRISPR/Cas9 is considered to be an efficient gene editing technologies which are being worked to improve and irradiate off‐
tools since its discovery in 2013. The simplicity and rapid application target risks. So we can conclude that, before moving towards RNA
of CRISPR/CAS9 highlighted and overshadowed ZFNs and TALENs. editing, gene, and protein targets are important for the develop-
Despite possessing wide application of gene editing technology there ment of therapeutics and diagnostics.
are several limitations associated with ZFNs, TALENs, and CRISPR/
Cas9 such as off‐target effects, due to off target unwanted gene
mutation, or rearrangement may occur, which can cause risk of AC KNO WL EDG EM E NT
oncogenesis, dysregulation in cell functioning, affecting the prolifera-
PMH is funded by a Fellowship from the National Health Medical
tion ability, and may lead to cell death. Hence, researchers are
Research Council, NHMRC (1079187). The authors acknowledge
working to improve technology by avoiding the problem of off‐target
NHMRC and the Graduate School of Health (GSH), University of
effect, it is overcome by considering mismatch tolerance and NGG
Technology Sydney (UTS), NSW, Australia for the funding.
mutations (P. D. Hsu et al., 2013; Pattanayak et al., 2013) and with
the introduction of offset nicking through Cas9 nickases and paired
gRNA (Mali, Aach et al., 2013; Ran et al., 2013), dose standardization CON F LI CT OF IN TE RES T S
of Cas9, and gRNA are also to be accomplished (P. D. Hsu et al.,
2013) by swapping protospacer adjacent motif (PAM) specificity to The authors declare that there are no conflict of interests.
noncanonical sequences (Kleinstiver et al., 2015; Zhang et al., 2014)
and merging catalytically inactive Cas9 to Fok I (fCas9; Guilinger,
AU TH OR CO NTRIB UTION S
Thompson, & Liu, 2014). These methods can act in prevention of off‐
target risks but cannot completely eradicate them. Therefore it is R. W., K. D., P. M. H., and P. K. M. conceived the idea, and all the
crucial to identify off‐site targets, which can be detected using high authors contributed to the preparation and editing of this manuscript
throughput technologies such as genome wide association studies, for intellectual content. All the authors read and approved the final
next generation sequencing, and so forth. Other challenges in version of the manuscript.
WADHWA ET AL. | 17

OR CID and other TAL effector‐based constructs for DNA targeting. Nucleic
Acids Research, 39(12), e82–e82.
Ridhima Wadhwa http://orcid.org/0000-0001-8074-1543 Chappell, S., Daly, L., Morgan, K., Guetta‐Baranes, T., Roca, J., Rabinovich, R., &
Keatings, V. (2008). Genetic variants of microsomal epoxide hydrolase and
glutamate‐cysteine ligase in COPD. European Respiratory Journal, 32(4),
REFERENC ES 931–937. https://doi.org/10.1183/09031936.00065308
Chen, Y., Huang, P., Ai, W., Li, X., Guo, W., Zhang, J., & Yang, J. (2012).
Adcock, I. M., Tsaprouni, L., Bhavsar, P., & Ito, K. (2007). Epigenetic Histone deacetylase activity is decreased in peripheral blood
regulation of airway inflammation. Current Opinion in Immunology, monocytes in patients with COPD. Journal of Inflammation, 9(1), 10.
19(6), 694–700. Cho, M. H., Castaldi, P. J., Hersh, C. P., Hobbs, B. D., Barr, R. G., Tal‐Singer,
Aggarwal, T., Wadhwa, R., Rohil, V., & Maurya, P. K. (2018). Biomarkers of R., & Coxson, H. O. (2015). A genome‐wide association study of
oxidative stress and protein–protein interaction in chronic obstruc- emphysema and airway quantitative imaging phenotypes. American
tive pulmonary disease. Archives of Physiology and Biochemistry, 124(3), journal of respiratory and critical care medicine, 192(5), 559–569.
226–231. Cho, M. H., McDonald, Merry‐L. N., Zhou, X., Mattheisen, M., Castaldi, P.
Aggarwal, T., Wadhwa, R., Thapliyal, N., Sharma, K., Rani, V., & Maurya, P. J., Hersh, C. P., DeMeo, D. L., Sylvia, J. S., Ziniti, J., Laird, N. M., Lange,
K. (2018). Oxidative, inflammatory, genetic, and epigenetic biomar- C., Litonjua, A. A., Sparrow, D., Casaburi, R., Barr, R. G., Regan, E. A.,
kers associated with chronic obstructive pulmonary disorder. Journal Make, B. J., Hokanson, J. E., Lutz, S., Dudenkov, T. M., Farzadegan, H.,
of Cellular Physiology, 234, 2067–2082. Hetmanski, J. B., Tal‐Singer, R., Lomas, D. A., Bakke, P., Gulsvik, A.,
Andresen, E., Günther, G., Bullwinkel, J., Lange, C., & Heine, H. (2011). Crapo, J. D., Silverman, E. K., & Beaty, T. H. (2014). Risk loci for
Increased expression of beta‐defensin 1 (DEFB1) in chronic obstruc- chronic obstructive pulmonary disease: a genome‐wide association
tive pulmonary disease. PLOS One, 6(7), e21898. study and meta‐analysis. The Lancet Respiratory Medicine, 2(3), 214–
Arunachalam, G., Yao, H., Sundar, I. K., Caito, S., & Rahman, I. (2010). 225. https://doi.org/10.1016/s2213‐2600(14)70002‐5
SIRT1 regulates oxidant‐and cigarette smoke‐induced eNOS acetyla- Christenson, S. A., Brandsma, C. ‐A., Campbell, J. D., Knight, D. A.,
tion in endothelial cells: Role of resveratrol. Biochemical and Pechkovsky, D. V., Hogg, J. C., & Spira, A. (2013). miR‐638 regulates
Biophysical Research Communications, 393(1), 66–72. gene expression networks associated with emphysematous lung
Ashburner, B. P., Westerheide, S. D., & Baldwin, A. S. (2001). The p65 destruction. Genome Medicine, 5(12), 114.
(RelA) subunit of NF‐κB interacts with the histone deacetylase Clarke, D. L., Sutcliffe, A., Deacon, K., Bradbury, D., Corbett, L., & Knox, A. J.
(HDAC) corepressors HDAC1 and HDAC2 to negatively regulate (2008). PKCβΙΙ Augments NF‐κB‐dependent transcription at the CCL11
gene expression. Molecular and Cellular Biology, 21(20), 7065–7077. promoter via p300/CBP‐associated factor recruitment and histone H4
Banerjee, E. R. (2014). Perspectives in inflammation biology. Springer. acetylation. The Journal of Immunology, 181(5), 3503–3514.
Barnes, P., Adcock, I., & Ito, K. (2005). Histone acetylation and Dalle‐Donne, I., Rossi, R., Colombo, R., Giustarini, D., & Milzani, A. (2006).
deacetylation: Importance in inflammatory lung diseases. European Biomarkers of oxidative damage in human disease. Clinical Chemistry,
Respiratory Journal, 25(3), 552–563. 52(4), 601–623.
Barnes, P. J. (2009). Role of HDAC2 in the pathophysiology of COPD. De Flora, S., Balansky, R., D'agostini, F., Cartiglia, C., Longobardi, M.,
Annual Review of Physiology, 71, 451–464. Steele, V. E., & Izzotti, A. (2012). Smoke‐induced microRNA and
Barnes, P. J. (2014). Cellular and molecular mechanisms of chronic related proteome alterations. Modulation by chemopreventive agents.
obstructive pulmonary disease. Clinics in Chest Medicine, 35(1), 71–86. International Journal of Cancer, 131(12), 2763–2773.
Barnes, P. J. (2016). Inflammatory mechanisms in patients with chronic DeMeo, D. L., Mariani, T., Bhattacharya, S., Srisuma, S., Lange, C., Litonjua,
obstructive pulmonary disease. Journal of Allergy and Clinical Immunol- A., & Sparrow, D. (2009). Integration of genomic and genetic
ogy, 138(1), 16–27. approaches implicates IREB2 as a COPD susceptibility gene. The
Barrangou, R., Fremaux, C., Deveau, H., Richards, M., Boyaval, P., Moineau, American Journal of Human Genetics, 85(4), 493–502.
S., & Horvath, P. (2007). CRISPR provides acquired resistance against DeMeo, D., & Silverman, E. (2004). à1‐Antitrypsin deficiencyú 2: Genetic
viruses in prokaryotes. Science, 315(5819), 1709–1712. aspects of à1‐antitrypsin deficiency: phenotypes and genetic modi-
ben Anes, A., Fetoui, H., Bchir, S., ben Nasr, H., Chahdoura, H., Chabchoub, fiers of emphysema risk. Thorax, 59(3), 259–264.
E., & Tabka, Z. (2014). Increased oxidative stress and altered levels of Ding, Q., Regan, S. N., Xia, Y., Oostrom, L. A., Cowan, C. A., & Musunuru, K.
nitric oxide and peroxynitrite in Tunisian patients with chronic (2013). Enhanced efficiency of human pluripotent stem cell genome
obstructive pulmonary disease: Correlation with disease severity and editing through replacing TALENs with CRISPRs. Cell Stem Cell, 12(4),
airflow obstruction. Biological Trace Element Research, 161(1), 20–31. 393–394.
Berlett, B. S., & Stadtman, E. R. (1997). Protein oxidation in aging, disease, and Eriksson, S. (2009). Pulmonary Emphysema and Alpha1‐Antitrypsin
oxidative stress. Journal of Biological Chemistry, 272(33), 20313–20316. Deficiency. Acta Medica Scandinavica, 175(2), 197–205. https://doi.
Berndt, A., Leme, A. S., & Shapiro, S. D. (2012). Emerging genetics of org/10.1111/j.0954‐6820.1964.tb00567.x
COPD. EMBO molecular medicine. EMBO Molecular Medicine, 4(11), Ezzati Givi, M., A Redegeld, F., Folkerts, G., & Mortaz, E. (2012). Dendritic
1144–1155. cells in pathogenesis of COPD. Current Pharmaceutical Design, 18(16),
Boch, J., Scholze, H., Schornack, S., Landgraf, A., Hahn, S., Kay, S., & Bonas, 2329–2335.
U. (2009). Breaking the code of DNA binding specificity of TAL‐type Ezzie, M. E., Crawford, M., Cho, J. ‐H., Orellana, R., Zhang, S., Gelinas, R., &
III effectors. Science, 326(5959), 1509–1512. Galas, D. (2011). Gene expression networks in COPD: MicroRNA and
Brusselle, G. G., Joos, G. F., & Bracke, K. R. (2011). New insights into the mRNA regulation. Thorax, thoraxjnl‐, 67, 2011–200089.
immunology of chronic obstructive pulmonary disease. The Lancet, Figarska, S. M., Vonk, J. M., & Boezen, H. M. (2014). NFE2L2
378(9795), 1015–1026. polymorphisms, mortality and metabolism in the general population.
Caito, S., Rajendrasozhan, S., Cook, S., Chung, S., Yao, H., Friedman, A. E., & American Journal of Physiology‐Heart and Circulatory Physiology.
Rahman, I. (2010). SIRT1 is a redox‐sensitive deacetylase that is post‐ Finkel, T., Deng, C. ‐X., & Mostoslavsky, R. (2009). Recent progress in the
translationally modified by oxidants and carbonyl stress. The FASEB biology and physiology of sirtuins. Nature, 460(7255), 587–591.
Journal, 24(9), 3145–3159. Gaj, T., Gersbach, C. A., & Barbas, C. F., III (2013). ZFN, TALEN, and
Cermak, T., Doyle, E. L., Christian, M., Wang, L., Zhang, Y., Schmidt, C., & CRISPR/Cas‐based methods for genome engineering. Trends in
Voytas, D. F. (2011). Efficient design and assembly of custom TALEN Biotechnology, 31(7), 397–405.
18 | WADHWA ET AL.

Gilowska, I. (2014). CXCL8 (interleukin 8) the key inflammatory mediator Hsu, P. D., Scott, D. A., Weinstein, J. A., Ran, F. A., Konermann, S.,
in chronic obstructive pulmonary disease? Advances in Hygiene & Agarwala, V., & Shalem, O. (2013). DNA targeting specificity of RNA‐
Experimental Medicine/Postepy Higieny i Medycyny Doswiadczalnej, 68, guided Cas9 nucleases. Nature Biotechnology, 31(9), 827–832.
842–850. Hunninghake, G. M., Cho, M. H., Tesfaigzi, Y., Soto‐Quiros, M. E., Avila, L.,
Global Initiative for Chronic Obstructive Lung Disease. (2019). Global Lasky‐Su, J., & Hallberg, J. (2009). MMP12, lung function, and COPD
strategy for the diagnosis, management, and prevention of COPD. in high‐risk populations. New England Journal of Medicine, 361(27),
Retrieved from https://goldcopd.org/wp‐content/uploads/2018/11/ 2599–2608.
GOLD‐2019‐v1.7‐FINAL‐14Nov2018‐WMS.pdf. Hussain, M., Rao, M., Humphries, A. E., Hong, J. A., Liu, F., Yang, M., &
Gosselink, J. V., Hayashi, S., Elliott, W. M., Xing, L., Chan, B., Yang, L., & Schrump, D. S. (2009). Tobacco smoke induces polycomb‐mediated
Pierce, J. A. (2010). Differential expression of tissue repair genes in repression of Dickkopf‐1 in lung cancer cells. Cancer Research, 69(8),
the pathogenesis of chronic obstructive pulmonary disease. American 3570–3578.
journal of respiratory and critical care medicine, 181(12), 1329–1335. Isajevs, S., Strazda, G., Kopeika, U., & Taivans, I. (2012). Different patterns
https://doi.org/10.1164/rccm.200812‐1902OC of lung sirtuin expression in smokers with and without chronic
Graff, J. W., Powers, L. S., Dickson, A. M., Kim, J., Reisetter, A. C., Hassan, I. obstructive pulmonary disease. Medicina, 48(10), 80.
H., & Monick, M. M. (2012). Cigarette smoking decreases global Isajevs, S., Taivans, I., Svirina, D., Strazda, G., & Kopeika, U. (2011).
microRNA expression in human alveolar macrophages. PLoS One, 7(8), Patterns of inflammatory responses in large and small airways in
e44066. smokers with and without chronic obstructive pulmonary disease.
Grewal, S. I., & Moazed, D. (2003). Heterochromatin and epigenetic Respiration, 81(5), 362–371.
control of gene expression. Science, 301(5634), 798–802. Isik, B., Isik, R. S., Ceylan, A., & Calik, O. (2005). Trace elements and
Guilinger, J. P., Thompson, D. B., & Liu, D. R. (2014). Fusion of catalytically oxidative stress in chronic obstructive pulmonary disease. Saudi
inactive Cas9 to FokI nuclease improves the specificity of genome medical journal, 26(12), 1882–1885.
modification. Nature Biotechnology, 32(6), 577–582. Ito, K., Ito, M., Elliott, W. M., Cosio, B., Caramori, G., Kon, O. M., & Hogg, J.
Hadchouel, A., Decobert, F., Franco‐Montoya, M.‐L., Halphen, I., Jarreau, C. (2005). Decreased histone deacetylase activity in chronic obstruc-
P.‐H., Boucherat, O., & Bourbon, J. (2008). Matrix metalloproteinase tive pulmonary disease. New England Journal of Medicine, 352(19),
gene polymorphisms and bronchopulmonary dysplasia: identification 1967–1976.
of MMP16 as a new player in lung development. PloS one, 3(9), e3188. Iuliano, L., Micheletta, F., Natoli, S., Corradini, S. G., Iappelli, M., Elisei, W.,
https://doi.org/10.1371/journal.pone.0003188 & Diczfalusy, U. (2003). Measurement of oxysterols and α‐tocopherol
Handy, D. E., Castro, R., & Loscalzo, J. (2011). Epigenetic modifications: in plasma and tissue samples as indices of oxidant stress status.
Basic mechanisms and role in cardiovascular disease. Circulation, Analytical Biochemistry, 312(2), 217–223.
123(19), 2145–2156. Izzotti, A., Calin, G. A., Steele, V. E., Croce, C. M., & De Flora, S. (2009).
Hanta, I., Kocabas, A., Canacankatan, N., Kuleci, S., & Seydaoglu, G. (2006). Relationships of microRNA expression in mouse lung with age and
Oxidant–antioxidant balance in patients with COPD. Lung, 184(2), exposure to cigarette smoke and light. The FASEB Journal, 23(9),
51–55. 3243–3250.
Hardin, M., Zielinski, J., Wan, E. S., Hersh, C. P., Castaldi, P. J., Schwinder, Jammes, Y., Steinberg, J. G., Ba, A., Delliaux, S., & Brégeon, F. (2008).
E., & Silverman, E. K. (2012). CHRNA3/5, IREB2, and ADCY2 are Enhanced exercise‐induced plasma cytokine response and oxidative
associated with severe chronic obstructive pulmonary disease in stress in COPD patients depend on blood oxygenation. Clinical
Poland. American Journal of Respiratory Cell and Molecular Biology, physiology and functional imaging, 28(3), 182–188.
47(2), 203–208. Jiang, Z., Lao, T., Qiu, W., Polverino, F., Gupta, K., Guo, F., & Castaldi, P. J.
Healy, J., & Tipton, K. (2007). Ceruloplasmin and what it might do. Journal (2016). A chronic obstructive pulmonary disease susceptibility gene,
of neural transmission, 114(6), 777. FAM13A, regulates protein stability of á‐catenin. American journal of
Hersh, C. P., Silverman, E. K., Gascon, J., Bhattacharya, S., Klanderman, B. respiratory and critical care medicine, 194(2), 185–197. https://doi.org/
J., Litonjua, A. A., Lefebvre, V., Sparrow, D., Reilly, J. J., Anderson, W. 10.1164/rccm.201505‐0999OC
H., Lomas, D. A., & Mariani, T. J. (2011). SOX5 Is a Candidate Gene for Jinek, M., Chylinski, K., Fonfara, I., Hauer, M., Doudna, J. A., & Charpentier,
Chronic Obstructive Pulmonary Disease Susceptibility and Is Neces- E. (2012). A programmable dual‐RNA–guided DNA endonuclease in
sary for Lung Development. American Journal of Respiratory and Critical adaptive bacterial immunity. Science, 337. 1225829‐21
Care Medicine, 183(11), 1482–1489. https://doi.org/10.1164/rccm. Kennedy, S. M., Chambers, R., Du, W., & Dimich‐Ward, H. (2007).
201010‐1751oc Environmental and occupational exposures: Do they affect chronic
Hodge, Emily, Nelson, Carl P, Miller, Suzanne, Billington, Charlotte K, obstructive pulmonary disease differently in women and men?
Stewart, Ceri E, Swan, Caroline, Malarstig, Anders, Henry, Amanda P, Proceedings of the American Thoracic Society, 4(8), 692–694.
Gowland, Catherine, Melén, Erik, Hall, Ian P, & Sayers, Ian (2013). Kim, W. J., & Lee, S. D. (2015). Candidate genes for COPD: current
HTR4 gene structure and altered expression in the developing lung. evidence and research. International journal of chronic obstructive
Respiratory Research, 14(1), 77. https://doi.org/10.1186/1465‐9921‐ pulmonary disease, 10, 2249–2255.
14‐77 Kim, W. J., Silverman, E. K., Hoffman, E., Criner, G. J., Mosenifar, Z.,
Hosgood, H. D., Menashe, I., He, X., Chanock, S., & Lan, Q. (2009). PTEN Sciurba, F. C., & Diaz, A. (2009). CT metrics of airway disease and
identified as important risk factor of chronic obstructive pulmonary emphysema in severe COPD. Chest, 136(2), 396–404.
disease. Respiratory medicine, 103(12), 1866–1870. Kleinstiver, B. P., Prew, M. S., Tsai, S. Q., Topkar, V. V., Nguyen, N. T.,
Houghton, A. M. (2015). Matrix metalloproteinases in destructive lung Zheng, Z., & Yeh, J. ‐R. J. (2015). Engineered CRISPR‐Cas9 nucleases
disease. Matrix biology, 44, 167–174. with altered PAM specificities. Nature, 523(7561), 481–485.
Houtkooper, R. H., Pirinen, E., & Auwerx, J. (2012). Sirtuins as regulators Kode, A., Yang, S. ‐R., & Rahman, I. (2006). Differential effects of cigarette
of metabolism and healthspan. Nature Reviews Molecular Cell Biology, smoke on oxidative stress and proinflammatory cytokine release in
13(4), 225–238. primary human airway epithelial cells and in a variety of transformed
Hsu, A. C., Dua, K., Starkey, M. R., Haw, T. ‐J., Nair, P. M., Nichol, K., & alveolar epithelial cells. Respiratory Research, 7(1), 132.
Foster, P. S. (2017). MicroRNA‐125a and‐b inhibit A20 and MAVS to Kong, X., Cho, M. H., Anderson, W., Coxson, H. O., Muller, N., Washko, G.,
promote inflammation and impair antiviral response in COPD. JCI Hoffman, E. A., Bakke, P., Gulsvik, A., Lomas, D. A., Silverman, E. K., &
Insight, 2(7), 90443. Pillai, S. G. (2011). Genome‐wide Association Study IdentifiesBICD1as
WADHWA ET AL. | 19

a Susceptibility Gene for Emphysema. American Journal of Respiratory impressions of severity. International Journal of Chronic Obstructive
and Critical Care Medicine, 183(1), 43–49. https://doi.org/10.1164/ Pulmonary Disease, 10, 1477.
rccm.201004‐0541oc Marwick, J. A., Kirkham, P. A., Stevenson, C. S., Danahay, H., Giddings, J.,
Kouzarides, T. (2007). Chromatin modifications and their function. Cell, Butler, K., & Rahman, I. (2004). Cigarette smoke alters chromatin
128(4), 693–705. remodeling and induces proinflammatory genes in rat lungs. American
Lakhdar, R., Denden, S., Mouhamed, M. H., Chalgoum, A., Leban, N., Knani, Journal of Respiratory Cell and Molecular Biology, 31(6), 633–642.
J., & Khelil, A. H. (2011). Correlation of EPHX1, GSTP1, GSTM1, and Marwick, J. A., Caramori, G., Stevenson, C. S., Casolari, P., Jazrawi, E.,
GSTT1 genetic polymorphisms with antioxidative stress markers in Barnes, P. J., & Papi, A. (2009). Inhibition of PI3Kδ restores
chronic obstructive pulmonary disease. Experimental lung research, glucocorticoid function in smoking‐induced airway inflammation in
37(4), 195–204. https://doi.org/10.3109/01902148.2010.535093 mice. American Journal of Respiratory and Critical Care Medicine, 179(7),
Lam, H. C., Cloonan, S. M., Bhashyam, A. R., Haspel, J. A., Singh, A., 542–548.
Sathirapongsasuti, J. F., & Mizumura, K. (2013). Histone deacetylase Mathers, C. D., & Loncar, D. (2006). Projections of global mortality and
6–mediated selective autophagy regulates COPD‐associated cilia burden of disease from 2002 to 2030. PLOS Medicine, 3(11), e442.
dysfunction. The Journal of Clinical Investigation, 123(12), 5212–5230. McDonough, J. E., Yuan, R., Suzuki, M., Seyednejad, N., Elliott, W. M.,
Larsson, K. (2007). Aspects on pathophysiological mechanisms in COPD. Sanchez, P. G., & Coxson, H. O. (2011). Small‐airway obstruction and
Journal of Internal Medicine, 262(3), 311–340. emphysema in chronic obstructive pulmonary disease. New England
Lawrence, T. (2009). The nuclear factor NF‐κB pathway in inflammation. Journal of Medicine, 365(17), 1567–1575.
Cold Spring Harbor Perspectives in Biology, 1, a001651. McGuinness, A. J. A., & Sapey, E. (2017). Oxidative stress in COPD:
Lee, J. H., Cho, M. H., Hersh, C. P., McDonald, M.‐L. N., Wells, J. M., Sources, markers, and potential mechanisms. Journal of Clinical
Dransfield, M. T., & Crapo, J. D. (2015). IREB2 and GALC are Medicine, 6(2), 21.
associated with pulmonary artery enlargement in chronic obstructive Milevoj Kopcinovic, L., Domijan, A.‐M., Posavac, K., Cepelak, I., Zanic
pulmonary disease. American journal of respiratory cell and molecular Grubisic, T., & Rumora, L. (2016). Systemic redox imbalance in stable
biology, 52(3), 365–376. https://doi.org/10.1165/rcmb.2014‐0210OC chronic obstructive pulmonary disease. Biomarkers, 21(8), 692–698.
Leus, N. G., Van Den Bosch, T., Van Der Wouden, P. E., Krist, K., Mizuno, S., Yasuo, M., Bogaard, H. J., Kraskauskas, D., Natarajan, R., &
Ourailidou, M. E., Eleftheriadis, N., & Mekonnen, S. A. (2017). HDAC1‐ Voelkel, N. F. (2010). Inhibition of histone deacetylase causes
3 inhibitor MS‐275 enhances IL10 expression in RAW264. 7 emphysema. American Journal of Physiology‐Lung Cellular and Molecular
macrophages and reduces cigarette smoke‐induced airway inflamma- Physiology, 300(3), L402–L413.
tion in mice. Scientific Reports, 7, 45047. Morrow, J. D., Cho, M. H., Hersh, C. P., Pinto‐Plata, V., Celli, B., Marchetti,
Levine, R. L. (2002). Carbonyl modified proteins in cellular regulation, N., & Glass, K. (2016). DNA methylation profiling in human lung tissue
aging, and disease2, 3. Free Radical Biology and Medicine, 32(9), identifies genes associated with COPD. Epigenetics, 11(10), 730–739.
790–796. https://doi.org/10.1080/15592294.2016.1226451
Li, X., Ortega, V. E., Ampleford, E. J., Barr, R. G., Christenson, S. A., Cooper, Mosammaparast, N., & Shi, Y. (2010). Reversal of histone methylation:
C. B., & Hansel, N. N. (2018). Genome‐wide association study of lung Biochemical and molecular mechanisms of histone demethylases.
function and clinical implication in heavy smokers. BMC medical Annual Review of Biochemistry, 79, 155–179.
genetics, 19(1), 134. https://doi.org/10.1186/s12881‐018‐0656‐z Mussolino, C., Morbitzer, R., Lütge, F., Dannemann, N., Lahaye, T., &
Lin, Y. ‐C., Wu, T. ‐C., Chen, P. ‐Y., Hsieh, L. ‐Y., & Yeh, S. ‐L. (2010). Cathomen, T. (2011). A novel TALE nuclease scaffold enables high
Comparison of plasma and intake levels of antioxidant nutrients in genome editing activity in combination with low toxicity. Nucleic Acids
patients with chronic obstructive pulmonary disease and healthy Research, 39(21), 9283–9293.
people in Taiwan: A case‐control study. Asia Pacific Journal of Clinical Nadeem, A., Raj, H. G., & Chhabra, S. K. (2005). Increased oxidative stress
Nutrition, 19(3), 393–401. and altered levels of antioxidants in chronic obstructive pulmonary
Lo, C. ‐L., Choudhury, S. R., Irudayaraj, J., & Zhou, F. C. (2017). Epigenetic disease. Inflammation, 29(1), 23–32.
editing of Ascl1 gene in neural stem cells by optogenetics. Scientific Nedeljkovic, I., Carnero‐Montoro, E., Lahousse, L., van der Plaat, D. A., de
Reports, 7, 42047. Jong, K., Vonk, J. M., & Boss‚, Y. (2018). Understanding the role of the
Lopez, A., Shibuya, K., Rao, C., Mathers, C., Hansell, A., Held, L., & Buist, S. chromosome 15q25. 1 in COPD through epigenetics and transcrip-
(2006). Chronic obstructive pulmonary disease: Current burden and tomics. European Journal of Human Genetics, 26(5), 709. https://doi.org/
future projections. European Respiratory Journal, 27(2), 397–412. 10.1038/s41431‐017‐0089‐8
Malhotra, D., Thimmulappa, R. K., Mercado, N., Ito, K., Kombairaju, P., Nedeljkovic, I., Lahousse, L., Carnero‐Montoro, E., Faiz, A., Vonk, J. M., de
Kumar, S., & Barnes, P. (2011). Denitrosylation of HDAC2 by targeting Jong, K., & Obeidat, M. e (2017). COPD GWAS variant at 19q13. 2 in
Nrf2 restores glucocorticosteroid sensitivity in macrophages from relation with DNA methylation and gene expression. Human Molecular
COPD patients. The Journal of Clinical Investigation, 121(11), Genetics, 27(2), 396–405.
4289–4302. Nicks, M. E., O'Brien, M. M., & Bowler, R. P. (2011). Plasma antioxidants
Mali, P., Yang, L., Esvelt, K. M., Aach, J., Guell, M., DiCarlo, J. E., & Church, are associated with impaired lung function and COPD exacerbations
G. M. (2013). RNA‐guided human genome engineering via Cas9. in smokers. COPD: Journal of Chronic Obstructive Pulmonary Disease,
Science, 339(6121), 823–826. 8(4), 264–269.
Mali, P., Aach, J., Stranges, P. B., Esvelt, K. M., Moosburner, M., Kosuri, S., Niki, E. (2009). Lipid peroxidation: Physiological levels and dual biological
& Church, G. M. (2013). CAS9 transcriptional activators for target effects. Free Radical Biology and Medicine, 47(5), 469–484.
specificity screening and paired nickases for cooperative genome Obeidat, Ma’en, Miller, Suzanne, Probert, Kelly, Billington, Charlotte K.,
engineering. Nature Biotechnology, 31(9), 833–838. Henry, Amanda P., Hodge, Emily, Nelson, Carl P., Stewart, Ceri E.,
Mannino, D. M., & Buist, A. S. (2007). Global burden of COPD: Risk Swan, Caroline, Wain, Louise V., Artigas, María Soler, Melén, Erik,
factors, prevalence, and future trends. The Lancet, 370(9589), Ushey, Kevin, Hao, Ke, Lamontagne, Maxime, Bossé, Yohan,
765–773. Postma, Dirkje S., Tobin, Martin D., Sayers, Ian, & Hall, Ian P.
Mapel, D. W., Dalal, A. A., Johnson, P. T., Becker, L. K., & Hunter, A. G. (2013). GSTCD and INTS12 Regulation and Expression in the
(2015). Application of the new GOLD COPD staging system to a US Human Lung. PLoS ONE, 8(9), e74630. https://doi.org/10.1371/
primary care cohort, with comparison to physician and patient journal.pone.0074630
20 | WADHWA ET AL.

Oglesby, I. K., McElvaney, N. G., & Greene, C. M. (2010). MicroRNAs in Song, J., Heijink, I., Kistemaker, L., Reinders‐Luinge, M., Kooistra, W.,
inflammatory lung disease‐master regulators or target practice? Noordhoek, J., & Hiemstra, P. (2017). Aberrant DNA methylation and
Respiratory Research, 11(1), 148. expression of SPDEF and FOXA2 in airway epithelium of patients with
O'neill, L. A., Sheedy, F. J., & McCoy, C. E. (2011). MicroRNAs: The fine‐ COPD. Clinical Epigenetics, 9(1), 42.
tuners of Toll‐like receptor signalling. Nature Reviews Immunology, Stylianou, P., Clark, K., & Bradding, P. (2016). Investigating tensin1
11(3), 163–175. expression and the presence of SNP identified in GWAS in COPD.
Pattanayak, V., Lin, S., Guilinger, J. P., Ma, E., Doudna, J. A., & Liu, D. R. European Respiratory Journal, 48(suppl 60PA4650). https://doi.org/10.
(2013). High‐throughput profiling of off‐target DNA cleavage reveals 1183/13993003.congress‐2016.PA4650
RNA‐programmed Cas9 nuclease specificity. Nature Biotechnology, Sundar, I. K., Nevid, M. Z., Friedman, A. E., & Rahman, I. (2013). Cigarette
31(9), 839–843. smoke induces distinct histone modifications in lung cells: Implica-
Porteus, M. H., & Carroll, D. (2005). Gene targeting using zinc finger tions for the pathogenesis of COPD and lung cancer. Journal of
nucleases. Nature Biotechnology, 23(8), 967–973. Proteome Research, 13(2), 982–996.
Pottelberge, G. R. V., Mestdagh, P., Bracke, K. R., Thas, O., Durme, Y. M. v, Sundar, I. K., Chung, S., Hwang, J. ‐w, Lapek Jr, J. D., Bulger, M., Friedman,
Joos, G. F., & Brusselle, G. G. (2011). MicroRNA expression in induced A. E., & Rahman, I. (2012). Mitogen‐and stress‐activated kinase 1
sputum of smokers and patients with chronic obstructive pulmonary (MSK1) regulates cigarette smoke‐induced histone modifications on
disease. American Journal of Respiratory and Critical Care Medicine, NF‐κB‐dependent genes. PLOS One, 7(2), e31378.
183(7), 898–906. Sünnetçioğlu, A., Alp, H. H., Sertogullarından, B., Balaharoglu, R., &
Pryor, W. A., & Stone, K. (1993). Oxidants in cigarette smoke radicals, Gunbatar, H. (2015). Evaluation of oxidative damage and antioxidant
hydrogen peroxide, peroxynitrate, and peroxynitrite. Annals of the mechanisms in COPD, lung cancer, and obstructive sleep apnea
New York Academy of Sciences, 686(1), 12–27. syndrome. Respiratory Care, 04209.
Qiu, W., Baccarelli, A., Carey, V. J., Boutaoui, N., Bacherman, H., Szulakowski, P., Crowther, A. J., Jiménez, L. A., Donaldson, K., Mayer, R.,
Klanderman, B., & Lomas, D. A. (2012). Variable DNA methylation is Leonard, T. B., & Drost, E. M. (2006). The effect of smoking on the
associated with chronic obstructive pulmonary disease and lung transcriptional regulation of lung inflammation in patients with
function. American Journal of Respiratory and Critical Care Medicine, chronic obstructive pulmonary disease. American Journal of Respiratory
185(4), 373–381. and Critical Care Medicine, 174(1), 41–50.
Quinlan, G. J., Martin, G. S., & Evans, T. W. (2005). Albumin: Biochemical Turell, L., Radi, R., & Alvarez, B. (2013). The thiol pool in human plasma:
properties and therapeutic potential. Hepatology, 41(6), 1211–1219. The central contribution of albumin to redox processes. Free Radical
Rahman, I., Biswas, S. K., & Kode, A. (2006). Oxidant and antioxidant Biology and Medicine, 65, 244–253.
balance in the airways and airway diseases. European Journal of Tzortzaki, E. G., Dimakou, K., Neofytou, E., Tsikritsaki, K., Samara, K.,
Pharmacology, 533(1‐3), 222–239. Avgousti, M., & Siafakas, N. M. (2012). Oxidative DNA damage and
Rahman, I., Gilmour, P. S., Jimenez, L. A., & MacNee, W. (2002). Oxidative somatic mutations: A link to the molecular pathogenesis of chronic
stress and TNF‐a induce histone acetylation and NF‐кB/AP‐1 inflammatory airway diseases. Chest, 141(5), 1243–1250.
activation in alveolar epithelial cells: Potential mechanism In gene Urnov, F. D., Rebar, E. J., Holmes, M. C., Zhang, H. S., & Gregory, P. D.
transcription in lung inflammation, In Oxygen/Nitrogen Radicals: Cell (2010). Genome editing with engineered zinc finger nucleases. Nature
Injury and Disease (239–248). Springer. Reviews Genetics, 11(9), 636–646.
Rajendrasozhan, S., Yang, S. ‐R., Kinnula, V. L., & Rahman, I. (2008). SIRT1, Vibhuti, A., Arif, E., Deepak, D., Singh, B., & Pasha, M. Q. (2007).
an antiinflammatory and antiaging protein, is decreased in lungs of Correlation of oxidative status with BMI and lung function in COPD.
patients with chronic obstructive pulmonary disease. American Journal Clinical biochemistry, 40(13‐14), 958–963.
of Respiratory and Critical Care Medicine, 177(8), 861–870. Von Mering, C., Jensen, L. J., Kuhn, M., Chaffron, S., Doerks, T., Krüger, B.,
Ran, F. A., Hsu, P. D., Lin, C. ‐Y., Gootenberg, J. S., Konermann, S., Trevino, & Bork, P. (2006). STRING 7—recent developments in the integration
A. E., & Zhang, Y. (2013). Double nicking by RNA‐guided CRISPR Cas9 and prediction of protein interactions. Nucleic Acids Research,
for enhanced genome editing specificity. Cell, 154(6), 1380–1389. 35(suppl_1), D358–D362.
Rouault, T. A. (2006). The role of iron regulatory proteins in mammalian Vucic, E. A., Chari, R., Thu, K. L., Wilson, I. M., Cotton, A. M., Kennett, J. Y.,
iron homeostasis and disease. Nature Chemical Biology, 2(8), 406–414. & Brown, C. J. (2014). DNA methylation is globally disrupted and
San Filippo, J., Sung, P., & Klein, H. (2008). Mechanism of eukaryotic associated with expression changes in chronic obstructive pulmonary
homologous recombination. Annual Review of Biochemistry, 77, disease small airways. American Journal of Respiratory Cell and
229–257. Molecular Biology, 50(5), 912–922.
Santos, M., Oliveira, A., Viegas‐Crespo, A., Vicente, L., Barreiros, A., Wan, E. S., Li, Y., Lao, T., Qiu, W., Jiang, Z., Mancini, J. D., & Zhou, X. (2017).
Monteiro, P., & De Almeida, A. B. (2004). Systemic markers of the Metabolomic profiling in a Hedgehog Interacting Protein (Hhip)
redox balance in chronic obstructive pulmonary disease. Biomarkers, murine model of chronic obstructive pulmonary disease. Scientific
9(6), 461–469. reports, 7(1), 2504. https://doi.org/10.1038/s41598‐017‐02701‐4
Schamberger, A. C., Mise, N., Meiners, S., & Eickelberg, O. (2014). Wan, E. S., Qiu, W., Baccarelli, A., Carey, V. J., Bacherman, H., Rennard,
Epigenetic mechanisms in COPD: Implications for pathogenesis and S. I., & DeMeo, D. L. (2012). Cigarette smoking behaviors and time
drug discovery. Expert Opinion on Drug Discovery, 9(6), 609–628. since quitting are associated with differential DNA methylation
Shahbazian, M. D., & Grunstein, M. (2007). Functions of site‐specific across the human genome. Human Molecular Genetics, 21(13),
histone acetylation and deacetylation. Annual Review of Biochemistry, 3073–3082.
76, 75–100. Wang, D. C., Shi, L., Zhu, Z., Gao, D., & Zhang, Y. (2017). Genomic
Shi, Y., Paluch, B. E., Wang, X., & Jiang, X. (2012). PTEN at a glance. Journal mechanisms of transformation from chronic obstructive pulmonary
of Cell Science, 125(20), 4687–4692. https://doi.org/10.1242/jcs. disease to lung cancer. Seminars in cancer biology.
093765 Wilber, A., Tschulena, U., Hargrove, P. W., Kim, Y. ‐S., Persons, D. A.,
Siedlinski, M., Klanderman, B., Sandhaus, R. A., Barker, A. F., Brantly, M. L., Barbas, C. F., & Nienhuis, A. W. (2010). A zinc‐finger transcriptional
Eden, E., & Stoller, J. K. (2012). Association of cigarette smoking and activator designed to interact with the γ‐globin gene promoters
CRP levels with DNA methylation in α‐1 antitrypsin deficiency. enhances fetal hemoglobin production in primary human adult
Epigenetics, 7(7), 720–728. erythroblasts. Blood, 115, 3033–3041. blood‐2009‐2008‐240556
WADHWA ET AL. | 21

Woode, D., Shiomi, T., & D'Armiento, J. (2015). Collagenolytic matrix Yu, Z.‐G., Wang, B.‐Z., & Cheng, Z.‐Z. (2017). The association of genetic
metalloproteinases in chronic obstructive lung disease and cancer. polymorphisms of hypoxia inducible factor‐1 alpha and vascular
Cancers, 7(1), 329–341. endothelial growth factor with increased risk of chronic obstructive
World Health Organization. (2017). Asthma. Retrieved from http://www. pulmonary disease: A case‐control study. The Kaohsiung journal of
who.int/mediacentre/factsheets/fs307/en/. medical sciences, 33(9), 433–441.
World Health Organization. (2019). Chronic obstructive pulmonary Yuan, C., De Chang, G. L., & Deng, X. (2017). Genetic polymorphism and
disease (COPD). Retrieved from https://www.who.int/respiratory/ chronic obstructive pulmonary disease. International journal of chronic
copd/en/. obstructive pulmonary disease, 12, 1385.
Woźniak, A., Górecki, D., Szpinda, M., Mila‐Kierzenkowska, C., & Woźniak, Zaynagetdinov, R., Sherrill, T. P., Gleaves, L. A., Hunt, P., Han, W., McLoed,
B. (2013). Oxidant‐antioxidant balance in the blood of patients with A. G., & Young, L. R. (2016). Chronic NF‐κB activation links COPD and
chronic obstructive pulmonary disease after smoking cessation. lung cancer through generation of an immunosuppressive micro-
Oxidative Medicine and Cellular Longevity, 2013, 2013–2019. environment in the lungs. Oncotarget, 7(5), 5470.
Wu, D.‐D., Song, J., Bartel, S., Krauss‐Etschmann, S., Rots, M. G., & Zhang, Y., Ge, X., Yang, F., Zhang, L., Zheng, J., Tan, X., & Gu, F. (2014).
Hylkema, M. N. (2017). The potential for targeted rewriting of Comparison of non‐canonical PAMs for CRISPR/Cas9‐mediated DNA
epigenetic marks in COPD as a new therapeutic approach. Pharma- cleavage in human cells. Scientific Reports, 4, 5405.
cology & Therapeutics, 182, 1–14. Zhu, G., Warren, L., Aponte, J., Gulsvik, A., Bakke, P., Investigators, I. C. G.
Wu, Q., Jiang, D., & Chu, H. W. (2012). Cigarette smoke induces growth N., & Pillai, S. G. (2007). The SERPINE2 gene is associated with chronic
differentiation factor 15 production in human lung epithelial cells: obstructive pulmonary disease in two large populations. American
Implication in mucin over‐expression. Innate Immunity, 18(4), journal of respiratory and critical care medicine, 176(2), 167–173.
617–626. https://doi.org/10.1164/rccm.200611‐1723OC
Yang, Yiqing, Xia, Min, Jin, Qingfeng, Bendahhou, Saïd, Shi, Jingyi, Chen, Zi¢lkowska‐Suchanek, I., Mosor, M., Gabryel, P., Grabicki, M., Zurawek,
Yiping, Liang, Bo, Lin, Jie, Liu, Yi, Liu, Ban, Zhou, Qinshu, Zhang, M., Fichna, M., & Nowak, J. (2015). Susceptibility loci in lung cancer
Dongwei, Wang, Rong, Ma, Ning, Su, Xiaoyan, Niu, Kaiya, Pei, Yan, Xu, and COPD: association of IREB2 and FAM13A with pulmonary
Wenyuan, Chen, Zhaopeng, Wan, Haiying, Cui, Jianmin, Barhanin, diseases. Scientific reports, 5(1), 13502. https://doi.org/10.1038/
Jacques, & Chen, Yihan (2004). Identification of a KCNE2 Gain‐of‐ srep13502
Function Mutation in Patients with Familial Atrial Fibrillation. The
American Journal of Human Genetics, 75(5), 899–905. https://doi.org/
10.1086/425342
Yao, H., & Rahman, I. (2012). Role of histone deacetylase 2 in epigenetics SU PP ORT IN G IN FOR M ATI O N
and cellular senescence: Implications in lung inflammaging and COPD.
American Journal of Physiology‐Lung Cellular and Molecular Physiology, Additional supporting information may be found online in the
303(7), L557–L566. Supporting Information section at the end of the article.
Yeung, F., Hoberg, J. E., Ramsey, C. S., Keller, M. D., Jones, D. R., Frye, R.
A., & Mayo, M. W. (2004). Modulation of NF‐κB‐dependent
transcription and cell survival by the SIRT1 deacetylase. The EMBO
Journal, 23(12), 2369–2380. How to cite this article: Wadhwa R, Aggarwal T, Malyla V,
Yildirim, A. O., Bulau, P., Zakrzewicz, D., Kitowska, K. E., Weissmann, N., et al. Identification of biomarkers and genetic approaches
Grimminger, F., & Eickelberg, O. (2006). Increased protein arginine toward chronic obstructive pulmonary disease. J Cell Physiol.
methylation in chronic hypoxia: Role of protein arginine methyltrans-
2019;1–21. https://doi.org/10.1002/jcp.28482
ferases. American Journal of Respiratory Cell and Molecular Biology,
35(4), 436–443.

You might also like