ArticlePDF Available

The role of 15 lipoxygenase 1 in asthma comes into focus

Authors:

Abstract

IL-4- and IL-13-driven epithelial cell expression of 15 lipoxygenase 1 (15LO1) is a consistent feature of eosinophil-dominated asthma known as type 2-high (T2-high) asthma. The abundant soluble products of arachidonic acid (AA) metabolized by 15LO1 reflect a high level of enzymatic activity in asthma and chronic rhinosinusitis. However, the precise role of 15LO1 and its products in disease pathogenesis remains enigmatic. In this issue of the JCI, Nagasaki and colleagues demonstrate a role for 15LO1 in controlling redox balance and epithelial homeostasis in T2-high asthma by metabolizing AA that is esterified to membrane phospholipids. The findings may pave the way toward the development of 15LO1 inhibitors as asthma treatments.
The Journal of Clinical Investigation COMMENTARY
1
The role of 15 lipoxygenase 1 in asthma comes into focus
Joshua A. Boyce
Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA.
Altered epithelial cell
differentiation in chronic
respiratory tract inflammation
Asthma and chronic rhinosinusitis with
nasal polyposis CRSwNP are prevalent
immune-mediated diseases of the respira-
tory tract that frequently coexist and cause
substantial morbidity . CRSwNP and
asthma are most often associated with
eosinophilic respiratory tissue iniltration,
mast cell hyperplasia, goblet cell metapla-
sia, and disrupted epithelial integrity ,
processes that are linked to the actions of
type  T cytokines type  inflammation;
ref. . Speciically, cytokines IL and IL
alter epithelial cell differentiation trajecto-
ries and function while strongly inducing
the expression of a cassette of characteristic
transcripts e.g., MUCAC, CLCA, CCL,
POSTN, and INOS in local stromal cells
by signaling though the α subunit of the
IL receptor ILRα , . The strength
of expression of these ILRα–driven tran-
scripts in bronchial and nasal mucosal epi-
thelial cells correlates with indices of dis-
ease severity , . Moreover, the eficacy of
a humanized monoclonal antibody against
ILRα dupilumab in the treatment of
severe T-high asthma and CRSwNP vali-
dates the biological importance of ILRα
inducible products in disease patho-
physiology , , although the relative
contributions of each remain less clear.
Pro- and antiinflammatory
functions of 15LO1 products
ALOX, encoding  lipoxygenase 
LO, is one of the strongly and con-
sistently expressed ILRα–inducible
transcripts by mucosal epithelial cells
in T-high asthma and CRSwNP , ,
. Its expression is especially strong in
aspirin-exacerbated respiratory disease
AERD, a disease phenotype that is over-
represented in the most severely affected
patients with asthma and CRSwNP .
ALOX is also expressed inducibly by
macrophages and constitutively by eosin-
ophils . LO catalyzes the oxidation
of arachidonic acid AA and other poly-
unsaturated fatty acids PUFAs, forming
peroxy derivatives that are precursors of
diverse mediators. In contrast to other
lipoxygenase enzymes e.g., LO, LO,
LO introduces molecular oxygen spe-
cies into both free PUFAs and into PUFAs
that are esteriied in phosphatidyletha-
nolamine PE in cell membranes .
LO converts free AA to an unstable
hydroperoxide, OOH eicosatetraeno-
ic acid HpETE, which is rapidly reduced
to S-HETE. These intermediates are
converted in turn to diverse metabolites,
including -oxo-S-hydroxyeicosa-
tetraenoic acid -oxo-S-HETE, a
chemoattractant for human eosinophils
, eoxin C, a conjugate of HETE
to reduced glutathione GSH that is
generated by eosinophils , and other
less-well-characterized products. LO
also converts the granulocyte-derived
LO product leukotriene A to lipoxin A,
a mediator that resolves inflammation
. LO products abound in biologi-
cal fluids e.g., nasal lavage, bronchoal-
veolar lavage BAL from patients with
asthma and CRSwNP; ref. , reflecting
LO activity in vivo, and correlating
with disease severity. Understanding the
balance of pro- and antiinflammatory
functions of LO products is essential
to therapeutic targeting of this enzyme.
While soluble products of LO
activity reflect peroxidation of free
PUFAs, oxidative products of esteriied
PUFAs remain cell associated and are
therefore more challenging to study. Both
LO and its homologue, LO which
is constitutively expressed in several
organs, interact with cell membrane–
associated PE-binding protein PEBP, a
scaffolding protein that regulates mito-
gen-activated protein kinase MAPK cas-
cades . The interaction between PEBP
and LO permits IL to induce the
activation of extracellular signal–regu-
lated kinase ERK . Additionally, the
association with PEBP switches the sub-
strate preference of LO from free to
esteriied AA, resulting in the formation
of HpETE esteriied onto PE HpETE-
PE. Importantly, LO-derived HpETE-
PE is a potent trigger of ferroptosis ,
a form of iron-dependent programmed
cell death involving lipid peroxidation
Related Article: https://doi.org/10.1172/JCI151685
Conflict of interest: JAB is a member of the Scientific Advisory Boards of Sanofi/Aventis, Third Harmonic Bio, and Siolta
Therapeutics.
Copyright: © 2022, Boyce. This is an open access article published under the terms of the Creative Commons Attribution 4.0
International License.
Reference information: J Clin Invest. 2022;132(1):e155884. https://doi.org/10.1172/JCI155884.
IL-4– and IL-13–driven epithelial cell expression of 15 lipoxygenase 1 (15LO1)
is a consistent feature of eosinophil-dominated asthma known as type
2–high (T2-high) asthma. The abundant soluble products of arachidonic
acid (AA) metabolized by 15LO1 reflect a high level of enzymatic activity in
asthma and chronic rhinosinusitis. However, the precise role of 15LO1 and its
products in disease pathogenesis remains enigmatic. In this issue of the JCI,
Nagasaki and colleagues demonstrate a role for 15LO1 in controlling redox
balance and epithelial homeostasis in T2-high asthma by metabolizing AA
that is esterified to membrane phospholipids. The findings may pave the
way toward the development of 15LO1 inhibitors as asthma treatments.
The Journal of Clinical Investigation
COMMENTARY
2J Clin Invest. 2022;132(1):e155884 https://doi.org/10.1172/JCI155884
upregulated LO protein expression.
Unexpectedly, IL also upregulated the
expression of both GPX and SLCA, a
glutamine transporter necessary to main-
tain intracellular GSH, suggesting a coor-
dinated cellular response to preserve redox
homeostasis. Stimulation of the cultured
cells with IL decreased both intra-
cellular and extracellular levels of GSH,
increased extracellular GSSH levels, and
decreased GSH/GSSH ratios in both com-
partments. Based on studies using small
interfering RNA knockdown and pharma-
cologic inhibition of LO in IL–stimu-
lated epithelial cells, the changes involved
LO. Intracellular GSH levels and GSH/
GSSH ratios in freshly obtained bronchial
epithelial cells correlated inversely with
LO expression levels but positively
with the SLCA/LO expression ratio
. Treatment of the IL–stimulated
epithelial cells with erastin, an inhibitor of
SLCA that depletes intracellular levels
of GSH, induced cell death while modest-
gators found higher glutathione disulide
GSSH reflecting the consumption of
GSH and lower GSH/GSSH ratios in BAL
fluids from subjects with severe asthma
than those with mild/moderate disease and
healthy controls. Intracellular GSH levels
and GSH/GSSH ratios were lowest in epi-
thelial cells from severe asthmatic subjects,
consistent with increased utilization of GSH
to maintain homeostasis. Both BAL fluid
and intracellular GSH/GSSH levels correlat-
ed inversely with the levels of exhaled nitric
oxide, a surrogate marker of T-driven INOS
expression, and correlated positively with
measures of lung function. These observa-
tions are consistent with altered redox bal-
ance in severe asthma that correlates with
both physiologic impairment and with sur-
rogate measures of T inflammation.
To understand the potential role of
ILRα–driven LO activity in altering
epithelial redox balance, the authors treat-
ed cultured bronchial epithelial cells with
IL ex vivo. As expected, IL strongly
. Glutathione peroxidase  GPX
reduces PE-associated lipid peroxides
 and prevents ferroptosis by convert-
ing HpETEPE to HETEPE, con-
suming GSH in the process. Diminished
expression of GPX or insuficient GSH
levels alter redox balance, favoring fer-
roptosis in the context of LOPEBP
interactions , a mechanism that may
potentially disrupt epithelial function and
barrier integrity Figure .
Controlling redox balance
in the airway
In this issue of the JCI, Nagasaki and
colleagues provide direct evidence that
LO-derived HpETEPE plays a role
in controlling redox balance in the airway
of patients with asthma, with potential
pathophysiologic consequences . Using
measures of redox balance in BAL fluids
and freshly harvested bronchial epithelial
cells from subjects with asthma who were
enrolled in two cohort studies, the investi-
Figure 1. Model for dual role of 15LO1 in asthma
related to type 2 inflammation. IL-4 and IL-13
induce IL-4Rα–dependent signaling to drive
ALOX15 (15LO1) expression in human airway
epithelial cells (HAECs). 15LO1 can add molecular
oxygen to free AA, forming HpETE that is rapidly
reduced to 15(S)-HETE, which in turn can be
converted into 5-oxo-15(S)-HETE, eoxin C4, and
other detectable mediators found in biological
fluids. 15LO1 also associates with PEBP in the
cell membrane, activating ERK (leading to
potentiated expression of additional type 2
inflammatory proteins) and switching the 15LO1
substrate preference to PE-esterified AA. The
resultant HpETE-PE triggers ferroptosis unless
it is reduced by GPX. This reduction requires
GSH, which is maintained by the glutamic acid
transporter protein SLC7A11. IL-13 increases both
GPX and SLC7A11 expression, suggesting that
a coordinated system maintains cellular redox
balance. Perturbations in this system, such as
diminished GSH availability, may favor ferro-
ptosis over cytoprotection, leading to epithelial
damage. Red color indicates proteins that are
upregulated in epithelial cells by IL-4 and IL-13.
The Journal of Clinical Investigation COMMENTARY
3
J Clin Invest. 2022;132(1):e155884 https://doi.org/10.1172/JCI155884
arachidonic acid metabolites produced via the
-lipoxygenase- pathway in human eosino-
phils and mast cells. Proc Natl Acad Sci U S A.
;:.
. Kuhn H, et al. Oxygenation of biological mem-
branes by the pure reticulocyte lipoxygenase.
J Biol Chem. ;:.
. Schwenk U, Schroder JM. Oxo- eicosanoids are
potent eosinophil chemotactic factors. Function-
al characterization and structural requirements.
J Biol Chem. ;: .
. Fiore S, Serhan CN. Formation of lipoxins and leu-
kotrienes during receptor-mediated interactions of
human platelets and recombinant human granulo-
cyte/macrophage colony-stimulating factor-primed
neutrophils. J Exp Med. ;:.
. Zhao J, et al. Lipoxygenase  interacts with
phosphatidylethanolamine-binding protein
to regulate MAPK signaling in human air-
way epithelial cells. Proc Natl Acad Sci U S A.
;:.
. Li Z, et al. Lipoxygenase  in nasal polyps
promotes CCL/eotaxin  expression through
extracellular signal-regulated kinase activation.
J Allerg Clin Immunol. ;:.
. Wenzel SE, et al. PEBP wardens ferroptosis by
enabling lipoxygenase generation of lipid death
signals. Cell. ;: .
. Dixon Scott J, et al. Ferroptosis: an iron-
dependent form of nonapoptotic cell death.
Cell. ;:.
. Yang WS, et al. Regulation of ferroptotic cancer
cell death by GPX. Cell. ;:.
. Nagasaki T, et al. LO dictates glutathione
redox changes in asthmatic airway epithelium
to worsen type  inflammation. J Clin Invest.
;:e.
. Zhao J, et al. Interleukin--induced MUCAC
is regulated by -lipoxygenase  pathway in
human bronchial epithelial cells. Am J Respir Crit
Care Me d. ;:.
. Zhao J, et al. Preferential generation of HETE-
PE induced by IL regulates goblet cell differ-
entiation in human airway epithelial cells. Am J
Respir Cell Mol Biol. ;:.
. Kristjansson RP, et al. A loss-of-function
variant in ALOX protects against nasal
polyps and chronic rhinosinusitis. Nat Genet.
;:.
. Smith KA, et al. Cost of adult chronic rhinosi-
nusitis: a systematic review. Laryngoscope.
;:.
. Stevens WW, et al. Clinical characteristics of
patients with chronic rhinosinusitis with nasal
polyps, asthma, and aspirin-exacerbated respi-
ratory disease. J Allerg Clin Immunol Pract.
;:.
. Wu W, et al. Unsupervised phenotyping of
Severe Asthma Research Program participants
using expanded lung data. J Allerg Clin Immu-
nol. ;:.
. Hellings PW, Steelant B. Epithelial barriers in
allergy and asthma. J Allerg Clin Immunol.
;:.
. Fahy JV. Type  inflammation in asthma--pres-
ent in most, absent in many. Nat Rev Immunol.
;:.
. Woodruff PG, et al. Genome-wide proiling
identiies epithelial cell genes associated
with asthma and with treatment response
to corticosteroids. Proc Natl Acad Sci U S A.
;:.
. Ordovas-Montanes J, et al. Allergic inflammato-
ry memory in human respiratory epithelial pro-
genitor cells. Nature. ; :.
. Dougherty RH, et al. Accumulation of intraepi-
thelial mast cells with a unique protease pheno-
type in TH-high asthma. J Allerg Clin Immu -
nol. ;:.
. Stevens WW, et al. Activation of the -lip-
oxygenase pathway in aspirin-exacerbated
respiratory disease. J Allerg Clin Immunol.
;:.
. Castro M, et al. Dupilumab eficacy and safety in
moderate-to-severe uncontrolled asthma.
N Engl J Med. ;:.
. Bachert C, et al. Eficacy and safety of dupilum-
ab in patients with severe chronic rhinosinusitis
with nasal polyps LIBERTY NP SINUS and
LIBERTY NP SINUS: results from two multi-
centre, randomised, double-blind, placebo-con-
trolled, parallel-group phase  trials. Lancet.
;:.
. Chu HW, et al. Expression and activation of -lip-
oxygenase pathway in severe asthma: relationship
to eosinophilic phenotype and collagen deposition.
Clin Exp Allerg. ;: .
. Feltenmark S, et al. Eoxins are proinflammatory
ly increasing expression and secretion of
CCL, periostin, and MUCAC, each of
which had previously been linked to LO
activity based on ex vivo studies , , ,
. Thus, perturbations in redox homeo-
stasis may substantially influence the phys-
iological consequences of induced LO
expression in asthma, CRSwNP, and other
diseases associated with T inflammation.
The study by Nagasaki et al. 
directly implicates altered redox balance
in severe asthma, and links this altered bal-
ance to the induced expression and func-
tion of LO. Although the authors do not
directly demonstrate LO-mediated fer-
roptosis in vivo, it is tempting to speculate
that the altered barrier integrity observed
in both asthma and CRSwNP may at least
partly reflect this process. While no cur-
rently available drugs block LO activity
in humans, a loss-of-function mutation of
ALOX confers strong protection against
the development of CRSwNP in cohorts
from Sweden and the United Kingdom ,
strongly suggesting a key role for LO
and its products in ILRα–driven epithelial
functional changes, and potential for ther-
apeutic targeting of LO with inhibitors.
It is tempting to speculate that the success
of dupilumab as a treatment for asthma and
CRSwNP could at least partly reflect a res-
toration of epithelial function by preventing
ferroptosis and other epithelial responses
that LO may control or influence.
Address correspondence to: Joshua A.
Boyce, Hale Building for Transformative
Medicine, Room V,  Fenwood
Road, Boston, Massachusetts , USA.
Phone: ..; Email: jboyce@rics.
bwh.harvard.edu.
... This indicates their importance in a sensitive response to allergic attacks and drug treatment in IL-13-treated lung epithelial cells. We also identified three highly upregulated proteins (with fold changes of over 25), FCGBP, ALOX15, and SER-PINB2, which play critical roles in human allergy, immune responses, and asthma [54][55][56]. The roles of increased ribosomal activity and histones were strongly reflected in our studies, as the proteomics pathways shown by the upregulated proteins in the Treated group revealed highly enriched pathway activity. ...
Article
Full-text available
Asthma is a chronic respiratory disease with one of the largest numbers of cases in the world; thus, constant investigation and technical development are needed to unravel the underlying biochemical mechanisms. In this study, we aimed to develop a nano-DESI MS method for the in vivo characterization of the cellular metabolome. Using air–liquid interface (ALI) cell layers, we studied the role of Interleukin-13 (IL-13) on differentiated lung epithelial cells acting as a lung tissue model. We demonstrate the feasibility of nano-DESI MS for the in vivo monitoring of basal–apical molecular transport, and the subsequent endogenous metabolic response, for the first time. Conserving the integrity of the ALI lung-cell layer enabled us to perform temporally resolved metabolomic characterization followed by “bottom-up” proteomics on the same population of cells. Metabolic remodeling was observed upon histamine and corticosteroid treatment of the IL-13-exposed lung cell monolayers, in correlation with alterations in the proteomic profile. This proof of principle study demonstrates the utility of in vivo nano-DESI MS for characterizing ALI tissue layers, and the new markers identified in our study provide a good starting point for future, larger-scale studies.
... Nevertheless, allergens-induced canonical ferroptosis in airway epithelial cells is involved in the development of asthma. [15][16][17] The usage of FIAs may aggravate ferroptosis in airway epithelial cells, thereby having adverse effects on asthma. Accordingly, figuring out the mechanism of the FIAs-induced noncanonical ferroptosis in eosinophils is of great value because it lays the foundation for targeted ferroptosis induction in asthma patients' eosinophils. ...
Article
Full-text available
Ferroptosis-inducing agents (FIAs) induced lipid-peroxidation-independent ferroptosis in eosinophils, thus ameliorating airway inflammation in asthmatic mice. Differences in ferroptosis-related genes (FerrGs) between eosinophils and cells in which FIAs induce canonical ferroptosis are supposed to contribute to this noncanonical ferroptosis but remain unclear. This study aims to explore these differences. This study used gastric cancer cells (GCCs) in stomach adenocarcinoma as the representative of cells in which FIAs induce canonical ferroptosis. FerrGs in Ferroptosis Database V2 respectively intersected with differentially expressed genes (DEGs) of eosinophils (E-MTAB-4660 dataset) and GCCs (GEPIA2 Stomach adenocarcinoma dataset) to obtain original ferroptosis DEGs (FerrDEGs). Then, they were subjected to Venn analysis to identify FerrDEGs shared by them and FerrDEGs exclusively expressed in eosinophils or GCCs. Identified genes were subjected to functional enrichment analysis, protein-protein interactions analysis, Hub genes analysis, and construction of the LncRNA-mediated ceRNA network. Sixty-six original FerrDEGs in eosinophils and 110 original FerrDEGs in GCCs were obtained. Venn analysis identified that eosinophils and GCCs shared 19 FerrDEGs that presented opposite expression directions and were involved in the ferroptosis pathway. Four upregulated and 20 downregulated FerrDEGs were exclusively expressed in eosinophils and GCCs, respectively. The former were enriched only in glycerolipid metabolism, while the latter were not enriched in pathways. Forty downregulated and 68 upregulated FerrDEGs were solely expressed in eosinophils and GCCs, respectively. The former was associated with the FoxO signaling pathway; the latter was related to glutathione metabolism and they were all implicated in autophagy. PPI analysis shows that the top 10 Hub genes of 66 original FerrDEGs and 44 exclusive FerrDEGs in eosinophils shared 9 genes ( STAT3, NFE2L2, MAPK8, PTEN, MAPK3, TLR4, SIRT1, BECN1 , and PTGS2 ) and they were also involved in the FoxO signaling pathway and autophagy pathway. Among them, PTEN is involved in forming a ceRNA network containing 3 LncRNAs, 3 miRNAs and 3 mRNAs. In contrast to FerrGs in cells in which FIAs induce canonical ferroptosis, the FerrGs in eosinophils differ in expression and in the regulation of ferroptosis, FoxO signaling pathway, and autophagy. It lays the groundwork for targeted induction of eosinophils lipid-peroxidation-independent ferroptosis in asthma.
... Increased 15-LOX activity has been reported in chronic sinusitis and asthma. Here, 15-LOX plays an important role in controlling the redox balance and epithelial homeostasis in asthma; therefore, the discoveries of 15-LOX inhibitors may pave the way for the creation of a new class of drugs used in the treatment of asthma [74]. ...
Article
Full-text available
In the present study, we characterize the biological activity of a newly designed and synthesized series of 15 compounds 2-[2-hydroxy-3-(4-substituted-1-piperazinyl)propyl] derivatives of pyrrolo[3,4-c]pyrrole 3a–3o. The compounds were obtained with good yields of pyrrolo[3,4-c]pyrrole scaffold 2a–2c with secondary amines in C2H5OH. The chemical structures of the compounds were characterized by 1H-NMR, 13C-NMR, FT-IR, and MS. All the new compounds were investigated for their potencies to inhibit the activity of three enzymes, i.e., COX-1, COX-2, and LOX, by a colorimetric inhibitor screening assay. In order to analyze the structural basis of interactions between the ligands and cyclooxygenase/lipooxygenase, experimental data were supported by the results of molecular docking simulations. The data indicate that all of the tested compounds influence the activity of COX-1, COX-2, and LOX.
Article
Full-text available
Traditional nutraceutical therapy is a promising approach in the management of inflammatory disorders. Fenugreek, bottle gourd, and betel leaf are rich sources of polyphenolic secondary metabolites. They are abundantly found in the Indian subcontinent for the preparation of healthy traditional functional food. Traditional evidence recommended that these herbs could control inflammation. The study was conducted with those herbs to validate their protective role against inflammatory mediators of arthritis and asthma through in‐silico and in‐vitro models. Phytochemical tests revealed the presence of flavonoids, amino acids, polyphenols, and saponins. Molecular docking was performed against six potential inflammatory bio‐marker proteins, like, LOX‐5, 15‐LOX, PLA2, IL‐6, TNFR1 & TNF‐α with the phyto‐marker compounds of test herbs and found satisfactory binding scores against inflammatory bio‐markers. Extended studies with Fenugreek and betel leaf aqueous fractions showcased 87 % and 91 % inhibition of protein denaturation at 200 μg/ml. Bottle‐gourd and betel‐leaf demonstrated 96 % and 98 % inhibition against 15‐Lipoxygenase at 100 μg/ml. TNF‐α and IL‐6 were dropped significantly by 49 % and 36 % upon treatment of betel leaf at 200 μg/ml from the LPS‐stimulated PBMC cultured cells. Findings may recommend the development of commercial anti‐inflammatory polyherbal phytopharmaceuticals for controlling polyarthritis and inflammatory complications subject to preclinical in‐vivo and clinical investigation.
Article
Full-text available
The persisting presence of opportunistic pathogens like Pseudomonas aeruginosa poses a significant threat to many immunocompromised cancer patients with pulmonary infections. This review highlights the complexity of interactions in the host’s defensive eicosanoid signaling network and its hijacking by pathogenic bacteria to their own advantage. Human lipoxygenases (ALOXs) and their mouse counterparts are integral elements of the innate immune system, mostly operating in the pro-inflammatory mode. Taking into account the indispensable role of inflammation in carcinogenesis, lipoxygenases have counteracting roles in this process. In addition to describing the structure-function of lipoxygenases in this review, we discuss their roles in such critical processes as cancer cell signaling, metastases, death of cancer and immune cells through ferroptosis, as well as the roles of ALOXs in carcinogenesis promoted by pathogenic infections. Finally, we discuss perspectives of novel oncotherapeutic approaches to harness lipoxygenase signaling in tumors.
Article
Full-text available
Altered redox biology challenges all cells, with compensatory responses often determining a cell's fate. When 15 lipoxygenase 1 (15LO1), a lipid-peroxidizing enzyme abundant in asthmatic human airway epithelial cells (HAECs), binds phosphatidylethanolamine-binding protein 1 (PEBP1), hydroperoxy-phospholipids, which drive ferroptotic cell death, are generated. Peroxidases, including glutathione peroxidase 4 (GPX4), metabolize hydroperoxy-phospholipids to hydroxy derivatives to prevent ferroptotic death, but consume reduced glutathione (GSH). The cystine transporter SLC7A11 critically restores/maintains intracellular GSH. We hypothesized that high 15LO1, PEBP1, and GPX4 activity drives abnormal asthmatic redox biology, evidenced by lower bronchoalveolar lavage (BAL) fluid and intraepithelial cell GSH:oxidized GSH (GSSG) ratios, to enhance type 2 (T2) inflammatory responses. GSH, GSSG (enzymatic assays), 15LO1, GPX4, SLC7A11, and T2 biomarkers (Western blot and RNA-Seq) were measured in asthmatic and healthy control (HC) cells and fluids, with siRNA knockdown as appropriate. GSSG was higher and GSH:GSSG lower in asthmatic compared with HC BAL fluid, while intracellular GSH was lower in asthma. In vitro, a T2 cytokine (IL-13) induced 15LO1 generation of hydroperoxy-phospholipids, which lowered intracellular GSH and increased extracellular GSSG. Lowering GSH further by inhibiting SLC7A11 enhanced T2 inflammatory protein expression and ferroptosis. Ex vivo, redox imbalances corresponded to 15LO1 and SLC7A11 expression, T2 biomarkers, and worsened clinical outcomes. Thus, 15LO1 pathway-induced redox biology perturbations worsen T2 inflammation and asthma control, supporting 15LO1 as a therapeutic target.
Article
Full-text available
Nasal polyps (NP) are lesions on the nasal and paranasal sinus mucosa and are a risk factor for chronic rhinosinusitis (CRS). We performed genome-wide association studies on NP and CRS in Iceland and the UK (using UK Biobank data) with 4,366 NP cases, 5,608 CRS cases, and >700,000 controls. We found 10 markers associated with NP and 2 with CRS. We also tested 210 markers reported to associate with eosinophil count, yielding 17 additional NP associations. Of the 27 NP signals, 7 associate with CRS and 13 with asthma. Most notably, a missense variant in ALOX15 that causes a p.Thr560Met alteration in arachidonate 15-lipoxygenase (15-LO) confers large genome-wide significant protection against NP (P = 8.0 × 10⁻²⁷, odds ratio = 0.32; 95% confidence interval = 0.26, 0.39) and CRS (P = 1.1 × 10⁻⁸, odds ratio = 0.64; 95% confidence interval = 0.55, 0.75). p.Thr560Met, carried by around 1 in 20 Europeans, was previously shown to cause near total loss of 15-LO enzymatic activity. Our findings identify 15-LO as a potential target for therapeutic intervention in NP and CRS. © 2019, The Author(s), under exclusive licence to Springer Nature Limited.
Article
Full-text available
Barrier tissue dysfunction is a fundamental feature of chronic human inflammatory diseases1. Specialized subsets of epithelial cells-including secretory and ciliated cells-differentiate from basal stem cells to collectively protect the upper airway2-4. Allergic inflammation can develop from persistent activation5 of type 2 immunity6 in the upper airway, resulting in chronic rhinosinusitis, which ranges in severity from rhinitis to severe nasal polyps7. Basal cell hyperplasia is a hallmark of severe disease7-9, but it is not known how these progenitor cells2,10,11 contribute to clinical presentation and barrier tissue dysfunction in humans. Here we profile primary human surgical chronic rhinosinusitis samples (18,036 cells, n = 12) that span the disease spectrum using Seq-Well for massively parallel single-cell RNA sequencing12, report transcriptomes for human respiratory epithelial, immune and stromal cell types and subsets from a type 2 inflammatory disease, and map key mediators. By comparison with nasal scrapings (18,704 cells, n = 9), we define signatures of core, healthy, inflamed and polyp secretory cells. We reveal marked differences between the epithelial compartments of the non-polyp and polyp cellular ecosystems, identifying and validating a global reduction in cellular diversity of polyps characterized by basal cell hyperplasia, concomitant decreases in glandular cells, and phenotypic shifts in secretory cell antimicrobial expression. We detect an aberrant basal progenitor differentiation trajectory in polyps, and propose cell-intrinsic13, epigenetic14,15 and extrinsic factors11,16,17 that lock polyp basal cells into this uncommitted state. Finally, we functionally demonstrate that ex vivo cultured basal cells retain intrinsic memory of IL-4/IL-13 exposure, and test the potential for clinical blockade of the IL-4 receptor α-subunit to modify basal and secretory cell states in vivo. Overall, we find that reduced epithelial diversity stemming from functional shifts in basal cells is a key characteristic of type 2 immune-mediated barrier tissue dysfunction. Our results demonstrate that epithelial stem cells may contribute to the persistence of human disease by serving as repositories for allergic memories.
Article
The respiratory epithelium provides a physical, functional, and immunologic barrier to protect the host from the potential harming effects of inhaled environmental particles and to guarantee maintenance of a healthy state of the host. When compromised, activation of immune/inflammatory responses against exogenous allergens, microbial substances, and pollutants might occur, rendering individuals prone to develop chronic inflammation as seen in allergic rhinitis, chronic rhinosinusitis, and asthma. The airway epithelium in asthma and upper airway diseases is dysfunctional due to disturbed tight junction formation. By putting the epithelial barrier to the forefront of the pathophysiology of airway inflammation, different approaches to diagnose and target epithelial barrier defects are currently being developed. Using single-cell transcriptomics, novel epithelial cell types are being unraveled that might play a role in chronicity of respiratory diseases. We here review and discuss the current understandings of epithelial barrier defects in type 2–driven chronic inflammation of the upper and lower airways, the estimated contribution of these novel identified epithelial cells to disease, and the current clinical challenges in relation to diagnosis and treatment of allergic rhinitis, chronic rhinosinusitis, and asthma.
Article
Background Aspirin Exacerbated Respiratory Disease (AERD) is characterized by asthma, chronic rhinosinusitis with nasal polyps (CRSwNP), and an intolerance to medications that inhibit cyclooxygenase-1. AERD patients have more severe upper and lower respiratory tract disease compared to aspirin-tolerant CRSwNP patients. A dysregulation in arachidonic acid metabolism is thought to contribute to the enhanced sinonasal inflammation in AERD. Objective To utilize an unbiased approach investigating arachidonic acid metabolic pathways in AERD. Methods Single-cell RNA sequencing (10X Genomics) was utilized to compare the transcriptional profile of NP cells from AERD and CRSwNP patients and map differences in the expression of select genes among identified cell types. Findings were confirmed by traditional RT-PCR. Lipid mediators were measured in sinonasal tissue by mass spectrometry. Localization of various proteins within NP was assessed by immunofluorescence. Results The gene encoding for 15-LO, ALOX15, was significantly elevated in NP of AERD compared to CRSwNP (p<0.05) or control (p<0.001). ALOX15 was predominantly expressed by epithelial cells. Expression levels significantly correlated with radiographic sinus disease severity (r=0.56, p<0.001) and were associated with asthma. 15-oxo-ETE, a downstream product of 15-LO, was significantly elevated in CRSwNP (27.93pg/mg tissue) and AERD (61.03pg/mg tissue) NP compared to control (7.17pg/mg tissue, p<0.001). Hydroxyprostaglandin dehydrogenase, an enzyme required for 15-oxo-ETE synthesis, was predominantly expressed in mast cells and localized near 15-LO⁺ epithelium in AERD NP. Conclusions Epithelial and mast cell interactions, leading to the synthesis of 15-oxo-ETE, may contribute to the dysregulation of arachidonic acid metabolism via the 15-LO pathway, and to the enhanced sinonasal disease severity observed in AERD.
Article
Background: Patients with chronic rhinosinusitis with nasal polyps (CRSwNP) generally have a high symptom burden and poor health-related quality of life, often requiring recurring systemic corticosteroid use and repeated sinus surgery. Dupilumab is a fully human monoclonal antibody that inhibits signalling of interleukin (IL)-4 and IL-13, key drivers of type 2 inflammation, and has been approved for use in atopic dermatitis and asthma. In these two studies, we aimed to assess efficacy and safety of dupilumab in patients with CRSwNP despite previous treatment with systemic corticosteroids, surgery, or both. Methods: LIBERTY NP SINUS-24 and LIBERTY NP SINUS-52 were two multinational, multicentre, randomised, double-blind, placebo-controlled, parallel-group studies assessing dupilumab added to standard of care in adults with severe CRSwNP. SINUS-24 was done in 67 centres in 13 countries, and SINUS-52 was done in 117 centres in 14 countries. Eligible patients were 18 years or older with bilateral CRSwNP and symptoms despite intranasal corticosteroid use, receiving systemic corticosteroids in the preceding 2 years, or having had sinonasal surgery. Patients in SINUS-24 were randomly assigned (1:1) to subcutaneous dupilumab 300 mg or placebo every 2 weeks for 24 weeks. Patients in SINUS-52 were randomly assigned (1:1:1) to dupilumab 300 mg every 2 weeks for 52 weeks, dupilumab every 2 weeks for 24 weeks and then every 4 weeks for the remaining 28 weeks, or placebo every 2 weeks for 52 weeks. All patients were randomly assigned centrally with a permuted block randomisation schedule. Randomisation was stratified by asthma or non-steroidal anti-inflammatory drug-exacerbated respiratory disease status at screening, previous surgery at screening, and country. Patients with or without comorbid asthma were included. Coprimary endpoints were changes from baseline to week 24 in nasal polyp score (NPS), nasal congestion or obstruction, and sinus Lund-Mackay CT scores (a coprimary endpoint in Japan), done in an intention-to-treat population. Safety was assessed in a pooled population of both dupilumab groups in SINUS-52 up to week 24 and the dupilumab group in SINUS-24 and the placebo groups in both studies until week 24. The trials are complete and registered at ClinicalTrials.gov, NCT02912468 and NCT02898454. Findings: Between Dec 5, 2016, and Aug 3, 2017, 276 patients were enrolled in SINUS-24, with 143 in the dupilumab group and 133 in the placebo group receiving at least one study drug dose. Between Nov 28, 2016, and Aug 28, 2017, 448 patients were enrolled in SINUS-52, with 150 receiving at least one dose of dupilumab every 2 weeks, 145 receiving at least one dose of dupilumab every 2 weeks for 24 weeks and every 4 weeks until week 52, and 153 receiving at least one dose of placebo. Dupilumab significantly improved the coprimary endpoints in both studies. At 24 weeks, least squares mean difference in NPS of dupilumab treatment versus placebo was -2·06 (95% CI -2·43 to -1·69; p<0·0001) in SINUS-24 and -1·80 (-2·10 to -1·51; p<0·0001) in SINUS-52; difference in nasal congestion or obstruction score was -0·89 (-1·07 to -0·71; p<0·0001) in SINUS-24 and -0·87 (-1·03 to -0·71; p<0·0001) in SINUS-52; and difference in Lund-Mackay CT scores was -7·44 (-8·35 to -6·53; p<0·0001) in SINUS-24 and -5·13 (-5·80 to -4·46; p<0·0001) in SINUS-52. The most common adverse events (nasopharyngitis, worsening of nasal polyps and asthma, headache, epistaxis, and injection-site erythema) were more frequent with placebo. Interpretation: In adult patients with severe CRSwNP, dupilumab reduced polyp size, sinus opacification, and severity of symptoms and was well tolerated. These results support the benefits of adding dupilumab to daily standard of care for patients with severe CRSwNP who otherwise have few therapeutic options. Funding: Sanofi and Regeneron Pharmaceuticals.
Article
Background 15-Lipoxygenase 1 (15LO1) is expressed in airway epithelial cells in patients with type 2–high asthma in association with eosinophilia. Chronic rhinosinusitis with nasal polyps (CRSwNP) is also associated with type 2 inflammation and eosinophilia. CCL26/eotaxin 3 has been reported to be regulated by 15LO1 in lower airway epithelial cells. However, its relation to 15LO1 in patients with CRSwNP or mechanisms for its activation are unclear. Objective We sought to evaluate 15LO1 and CCL26 expression in nasal epithelial cells (NECs) from patients with CRSwNP and healthy control subjects (HCs) and determine whether 15LO1 regulates CCL26 in NECs through extracellular signal-regulated kinase (ERK) activation. Methods 15LO1, CCL26, and phosphorylated ERK were evaluated in NECs from patients with CRSwNP and HCs. 15LO1/CCL26 and CCL26/cytokeratin 5 were colocalized by means of immunofluorescence. IL-13–stimulated NECs were cultured at an air-liquid interface with or without 15-lipoxygenase 1 gene (ALOX15) Dicer-substrate short interfering RNAs (DsiRNA) transfection, a specific 15LO1 enzymatic inhibitor, and 2 ERK inhibitors. Expression of 15LO1 and CCL26 mRNA and protein was analyzed by using quantitative RT-PCR, Western blotting, and ELISA. Results 15LO1 expression was increased in nasal polyp (NP) epithelial cells compared with middle turbinate epithelial cells from patients with CRSwNP and HCs. 15LO1 expression correlated with CCL26 expression and colocalized with CCL26 expression in basal cells of the middle turbinate and NPs from patients with CRSwNP. In primary NECs in vitro, IL-13 induced 15LO1 and CCL26 expression. 15LO1 knockdown and inhibition decreased IL-13–induced ERK phosphorylation and CCL26 expression. ERK inhibition (alone) similarly decreased IL-13-induced CCL26. Phosphorylated ERK expression was increased in NECs from CRSwNP subjects and positively correlated with both 15LO1 and CCL26 expression. Conclusions 15LO1 expression is increased in NP epithelial cells and contributes to CCL26 expression through ERK activation. 15LO1 could be considered a novel therapeutic target for CRSwNP.
Article
Background Dupilumab is a fully human anti–interleukin-4 receptor α monoclonal antibody that blocks both interleukin-4 and interleukin-13 signaling. We assessed its efficacy and safety in patients with uncontrolled asthma. Methods We randomly assigned 1902 patients 12 years of age or older with uncontrolled asthma in a 2:2:1:1 ratio to receive add-on subcutaneous dupilumab at a dose of 200 or 300 mg every 2 weeks or matched-volume placebos for 52 weeks. The primary end points were the annualized rate of severe asthma exacerbations and the absolute change from baseline to week 12 in the forced expiratory volume in 1 second (FEV1) before bronchodilator use in the overall trial population. Secondary end points included the exacerbation rate and FEV1 in patients with a blood eosinophil count of 300 or more per cubic millimeter. Asthma control and dupilumab safety were also assessed. Results The annualized rate of severe asthma exacerbations was 0.46 (95% confidence interval [CI], 0.39 to 0.53) among patients assigned to 200 mg of dupilumab every 2 weeks and 0.87 (95% CI, 0.72 to 1.05) among those assigned to a matched placebo, for a 47.7% lower rate with dupilumab than with placebo (P<0.001); similar results were seen with the dupilumab dose of 300 mg every 2 weeks. At week 12, the FEV1 had increased by 0.32 liters in patients assigned to the lower dose of dupilumab (difference vs. matched placebo, 0.14 liters; P<0.001); similar results were seen with the higher dose. Among patients with a blood eosinophil count of 300 or more per cubic millimeter, the annualized rate of severe asthma exacerbations was 0.37 (95% CI, 0.29 to 0.48) among those receiving lower-dose dupilumab and 1.08 (95% CI, 0.85 to 1.38) among those receiving a matched placebo (65.8% lower rate with dupilumab than with placebo; 95% CI, 52.0 to 75.6); similar results were observed with the higher dose. Blood eosinophilia occurred after the start of the intervention in 52 patients (4.1%) who received dupilumab as compared with 4 patients (0.6%) who received placebo. Conclusions In this trial, patients who received dupilumab had significantly lower rates of severe asthma exacerbation than those who received placebo, as well as better lung function and asthma control. Greater benefits were seen in patients with higher baseline levels of eosinophils. Hypereosinophilia was observed in some patients. (Funded by Sanofi and Regeneron Pharmaceuticals; LIBERTY ASTHMA QUEST ClinicalTrials.gov number, NCT02414854.)
Article
Ferroptosis is a form of programmed cell death that is pathogenic to several acute and chronic diseases and executed via oxygenation of polyunsaturated phosphatidylethanolamines (PE) by 15-lipoxygenases (15-LO) that normally use free polyunsaturated fatty acids as substrates. Mechanisms of the altered 15-LO substrate specificity are enigmatic. We sought a common ferroptosis regulator for 15LO. We discovered that PEBP1, a scaffold protein inhibitor of protein kinase cascades, complexes with two 15LO isoforms, 15LO1 and 15LO2, and changes their substrate competence to generate hydroperoxy-PE. Inadequate reduction of hydroperoxy-PE due to insufficiency or dysfunction of a selenoperoxidase, GPX4, leads to ferroptosis. We demonstrated the importance of PEBP1-dependent regulatory mechanisms of ferroptotic death in airway epithelial cells in asthma, kidney epithelial cells in renal failure, and cortical and hippocampal neurons in brain trauma. As master regulators of ferroptotic cell death with profound implications for human disease, PEBP1/15LO complexes represent a new target for drug discovery.
Article
Rationale: Type-2-associated goblet cell hyperplasia and mucus hypersecretion are well known features of asthma. 15-Lipoxygenase (15LO1) is induced by the Type-2 cytokines/IL-13 in human airway epithelial cells (HAEC) in vitro and is increased in fresh asthmatic HAECs ex vivo. 15LO1 generates a variety of products, including 15-hydroxyeicosatetraenoic acid (15-HETE), 15-HETE-phosphatidyethanoloamine (PE) (15-HETE-PE) and 13-hydroxyoctadecadienoic acid (13-HODE). The current study investigated the 15LO1 metabolite profile at baseline and after IL-13 treatment, and the influence on goblet cell differentiation in HAECs. Methods: Primary HAECs obtained from bronchial brushings of asthmatic and healthy subjects were cultured under air-liquid interface (ALI) culture supplemented with arachidonic acid (AA) and linoleic acid (LA) (10 µM each) and exposed to IL-13 for 7 days. siRNA transfection and 15LO1 inhibition were applied to suppress 15LO1 expression and activity. Results: IL-13 stimulation induced 15LO1 expression and preferentially generated 15-HETE-PE in vitro, both of which persist after removal of IL-13. 15LO1 inhibition (siRNA and chemical inhibitor) decreased IL-13 induced FOXA3 expression, while enhancing FOXA2 expression. These changes were associated with reductions in both MUC5AC and periostin. Exogenous 15-HETE-PE stimulation (alone) recapitulated IL-13 induced FOXA3, MUC5AC and periostin expression. Conclusions: The results from this study confirm the central importance of 15LO1 and its primary product 15-HETE-PE to epithelial cell remodeling in HAECs.