Download as pdf or txt
Download as pdf or txt
You are on page 1of 19

Archives of Toxicology (2021) 95:1141–1159

https://doi.org/10.1007/s00204-021-02976-7

REVIEW ARTICLE

Carbon monoxide‑triggered health effects: the important role


of the inflammasome and its possible crosstalk with autophagy
and exosomes
Rong‑Jane Chen1 · Yu‑Hsuan Lee2   · Tzu‑Hao Chen3,4 · Yu‑Ying Chen3 · Ya‑Ling Yeh3 · Ching‑Ping Chang4 ·
Chien‑Cheng Huang3,5,6 · How‑Ran Guo3,7,8   · Ying‑Jan Wang3,9 

Received: 27 September 2020 / Accepted: 4 January 2021 / Published online: 8 February 2021
© The Author(s), under exclusive licence to Springer-Verlag GmbH, DE part of Springer Nature 2021

Abstract
Carbon monoxide (CO) has long been known as a “silent killer” because of its ability to bind hemoglobin (Hb), leading
to reduced oxygen carrying capacity of Hb, which is the main cause of CO poisoning (COP) in humans. Emerging stud-
ies suggest that mitochondria is a key target of CO action that can impact key biological processes, including apoptosis,
cellular proliferation, inflammation, and autophagy. Despite its toxicity at high concentrations, CO also exhibits cyto- and
tissue-protective effects at low concentrations in animal models of organ injury and disease. Specifically, CO modulates the
production of pro- or anti-inflammatory cytokines and mediators by regulating the NLRP3 inflammasome. Given that human
diseases are strongly associated with inflammation, a deep understanding of the exact mechanism is helpful for treatment.
Autophagic factors and inflammasomes interact in various situations, including inflammatory disease, and exosomes might
function as the bridge between the inflammasome and autophagy activation. Thus, the interplay among autophagy, mito-
chondrial dysfunction, exosomes, and the inflammasome may play pivotal roles in the health effects of CO. In this review,
we summarize the latest research on the beneficial and toxic effects of CO and their underlying mechanisms, focusing on the
important role of the inflammasome and its possible crosstalk with autophagy and exosomes. This knowledge may lead to
the development of new therapies for inflammation-related diseases and is essential for the development of new therapeutic
strategies and biomarkers of COP.

Keywords  Carbon monoxide · Mitochondria · NLRP3 inflammasome · Autophagy · Exosome

Abbreviations COHb Carboxyhemoglobin


CO Carbon monoxide ROS Reactive oxygen species
Hb Hemoglobin HO Heme oxygenase
BV Biliverdin
BR Bilirubin
Rong-Jane Chen and Yu-Hsuan Lee contributed equally to this IL-1β Interleukin-1β
work.

4
* How‑Ran Guo Department of Medical Research, Chi Mei Medical Center,
hrguo@mail.ncku.edu.tw Tainan, Taiwan
5
* Ying‑Jan Wang Department of Emergency Medicine, Chi Mei Medical
yjwang@mail.ncku.edu.tw Center, Tainan, Taiwan
6
1 Department of Senior Services, Southern Taiwan University
Department of Food Safety/Hygiene and Risk Management,
of Science and Technology, Tainan, Taiwan
College of Medicine, National Cheng Kung University,
7
Tainan, Taiwan Department of Occupational and Environmental Medicine,
2 National Cheng Kung University Hospital, Tainan, Taiwan
Department of Cosmeceutics, China Medical University,
8
Taichung, Taiwan Occupational Safety, Health and Medicine Research Center,
3 National Cheng Kung University Hospital, Tainan, Taiwan
Department of Environmental and Occupational Health,
9
College of Medicine, National Cheng Kung University, 138 Department of Medical Research, China Medical University
Sheng‑Li Road, Tainan 70428, Taiwan Hospital, China Medical University, Taichung, Taiwan

13
Vol.:(0123456789)

1142 Archives of Toxicology (2021) 95:1141–1159

miRs MicroRNAs have emerged describing the physiological and cellular con-
COP Carbon monoxide poisoning sequences of CO exposure in vitro and in vivo as well as
DM Diabetes mellitus the mechanisms responsible for these effects. In addition
IRR Incidence rate ratios to the effects of exogenous CO, a variety of biologic and
CI Confidence interval physiologic functions could be regulated by endogenous CO,
CK Creatine kinase including vascular tone, mitochondrial homeostasis and bio-
BNP Brain natriuretic peptide genesis, neurotransmission, inflammation and programmed
FABP Fatty acid-binding protein cell death (Borzelleca 2000). Basically, CO can evolve
TLR4 Toll-like receptor 4 endogenously in living organisms as the result of enzymatic
LPS Lipopolysaccharide heme degradation, which is catalyzed by the heme oxyge-
MAPK Mitogen-activated protein kinase nase (HO) enzymes (Maines et al. 1986; Tenhunen et al.
ASC Apoptosis-associated speck-like protein contain- 1968). It has been well demonstrated that HO activity cata-
ing caspase-recruitment domain lyzes the oxidative cleavage of heme to biliverdin (BV), in
NO Nitric oxide a reaction that liberates ferrous iron and CO. BV is further
mtROS Mitochondrial ROS enzymatically reduced to bilirubin (Keyse and Tyrrell 1989;
COPD Chronic obstruction pulmonary disease Maines et al. 1986; Tenhunen et al. 1970).
The success of CO as a therapeutic candidate in preclini-
cal models suggests its potential of further clinical appli-
Introduction: dark side and bright side of CO cation to inflammatory and proliferative disorders, which
is currently under evaluation in clinical trials (Goebel and
Carbon monoxide (CO) is a colorless, tasteless, odorless Wollborn 2020; Kim and Choi 2018). Additional studies
gas generated by incomplete combustion of carbon com- are also needed to identify how discrete signaling targets of
pounds. Exposure to high concentrations of CO can cause CO are integrated in a context-specific fashion to coordinate
asphyxiation, severe neurocognitive effects, cardiac dysfunc- cyto- and tissue-protection.
tion, and death during accidental or occupational inhalation The mechanisms by which HO-1 expression are associ-
(Von Burg 1999). In the United States, CO poisoning (COP) ated with cyto- and tissue-protection have not been com-
affects approximately 50,000 people per year with mortality pletely understood, but may involve the combined effects
rates ranging from 1 to 3% (Rose et al. 2017). The binding of the removal of heme (a pro-oxidant iron-chelate) with
affinity of CO to hemoglobin (Hb) is 250-fold greater than the enzymatic generation of CO and the generation of BV/
that of oxygen, and so CO can reduce the oxygen carrying BR from heme catabolism (Ryter and Tyrrell 2000). Recent
capacity of Hb (Hall 2010). However, neither the clinical studies have demonstrated that induction of HO-1 can reduce
severity of CO-poisoned patients nor the patients’ improve- interleukin-1β (IL-1β) maturation and downregulate inflam-
ment directly correlates with the blood carboxyhemoglobin masome activation in a lung injury model (Luo et al. 2014).
(COHb) level or COHb clearance (Hampson and Hauff Several molecular mechanisms and regulators have been
2008; Weaver et al. 2008). Thus, the toxic effects of CO proposed for NLRP3 inflammasome activation, includ-
involve inhibition of oxygen delivery as well as binding to ing potassium efflux and calcium influx, lysosome desta-
other cellular heme-containing proteins, such as myoglo- bilization and rupture, mitochondrial ROS (mtROS), and
bin, mitochondrial cytochrome c oxidase, soluble guanylyl mitochondrial DNA damage. On the other hand, negative
cyclase, cytochrome p-450, inducible nitric oxide synthase regulators include autophagy, cytokines, CO, and miRNAs
(NOS) and others (Borzelleca 2000; Rose et  al. 2017). (Jo et al. 2016; Rathinam and Fitzgerald 2016). A balance
Thus, mitochondria may represent a key target of CO, and between activation and inactivation of the NLRP3 inflam-
exposure to CO results in changes in membrane potential, masome is critical for maintaining normal homeostasis and
generation of reactive oxygen species (ROS), release of pro- physiological function (Mangan et al. 2018). Activation of
apoptotic and pro-inflammatory mediators, and inhibition of the NLRP3 inflammasome plays a pivotal role in the patho-
respiration at high concentrations (Ryter et al. 2018). genesis of a variety of diseases such as atherosclerosis, myo-
The dose determines the toxicity is a basic principle of cardial ischemia, ischemic stroke, hypertension, and diabe-
toxicology; thus, the same substance could act as a thera- tes mellitus (DM) (Liu et al. 2020). Therefore, studies on
peutic at a lower dosage (Borzelleca 2000). In this regard, NLRP3 inflammasome, a core and important component of
when applied at low concentrations, CO has been used innate immunity, may indicate new targets for the treatment
as a gaseous therapeutic in experimental models of organ of various inflammatory diseases.
injury and disease, as well as in conditions involving aber- Autophagy is an evolutionarily conservative pathway
rant inflammatory or proliferative states (Ryter et al. 2006; important to cellular homeostasis that is induced when
Ryter and Choi 2016). From these studies, accumulated data eukaryotic cells encounter various types of stimulation. It is

13
Archives of Toxicology (2021) 95:1141–1159 1143

involved in the process of recycling of damaged organelles regulation that exists among inflammasome, autophagy, and
(e.g., mitochondria) and proteins as well as the destruction exosome. By analyzing these intriguing interconnections, we
of intracellular pathogens (Deretic 2011; Yin et al. 2016). hope to unveil the mechanisms involved in these processes
Autophagy and inflammasomes interact in many pathologi- and provide potential therapeutic targets for certain diseases.
cal situations, including infectious disease, cancer, DM,
and neurodegeneration (Pu et al. 2019). Via crosstalk with
autophagic pathways, inflammasomes affect the effector Updated knowledge regarding CO
cells of innate and adaptive immunity during inflamma- intoxication
tory response (Rao and Eissa 2020). The interplay between
autophagy and inflammasome activation can be attributed COP and major long‑term consequences
to multiple layers of molecular regulators, and the details
of such interplay remain largely unknown. Recent studies People suffering from COP manifest acute syndromes,
have uncovered novel anti-inflammatory targets of HO-1/ including headache, nausea, fatigue, dizziness, and chest
CO, including regulation of autophagy and inflammasome pain within a short period of time (Hampson et al. 2012).
pathways. HO-1 may localize to mitochondria in response Nonetheless, long term consequences such as neurologi-
to stress, whereas CO moderates mitochondrial dysfunction cal disorders also need to be taken seriously. In addition,
and regulates autophagy and mitochondrial biogenesis. The Huang et al. (2014) reported that COP patients exhibited a
interplay among autophagy, mitochondrial dysfunction, and significantly increased risk of long-term mortality, which
the regulation of inflammation may make important contri- is particularly high in the first month after COP and then
butions to the protection afforded by HO-1/CO in cellular decreased slowly at 6 months to 1 year. Persistent neuro-
and organ injury models (Ryter 2019). Interestingly, emerg- logical sequelae and delayed neurological sequelae are the
ing studies suggest the critical involvement of microRNAs most common syndromes following COP, and both condi-
(miRs) in the regulation of HO-1 gene expression either by tions are triggered by white matter injury (Ning et al. 2020).
decreasing mRNA stability or by modulating the expres- Chang et al. (2009) found that the fractional anisotropy value
sion of upstream regulatory factors, such as Nrf2/Keap1 of white matter on diffusion tensor imaging by CT or MRI
and Bach1 (Cheng et al. 2013). These findings indicate an in the presence of acute brain lesions can predict delayed
emerging complexity of HO-1 regulation and function that neurological sequelae. Morphological changes in subcor-
may involve a network of miR-dependent regulatory events. tical structures and the pallidum have also been observed
They also indicate that miRs may participate in the crosstalk after COP (Hsiao et al. 2004). However, Ozcan et al. (2016)
between inflammasomes and autophagy in physiological or claimed that white matter lesions that progress to demyeli-
disease conditions (Pu et al. 2019). nation and result in neuropsychological sequelae could not
Recent studies have revealed that exosomal and always be diagnosed by early CT or MRI in COP. Addition-
autophagic pathways can be regulated by each other, and ally, neurocognitive deficits concomitant with depression,
the effects of this cross-regulation play important roles in anxiety, mental and psychiatric disorders, and violence con-
the occurrence and progression of various diseases (Lin tribute to the increased risk for long-term mortality (Huang
et al. 2019). Autophagy can regulate endosomal secretion et al. 2014). Apart from neurological disorder, COP has also
to form exosomes, which subsequently regulate autophagy been demonstrated to cause long-term effects on the cardio-
in a paracrine manner (Papandreou and Tavernarakis 2017). vascular system, including myocardial dysfunction, ischemia
In addition, growing evidence indicates that inflammasome and infarction, arrhythmias, and cardiac arrest. Similarly, a
activity correlates with enhanced secretion of exosomes and cohort study showed an increased long-term risk of major
modulation of their protein cargo through the effector pro- adverse cardiovascular events in patients with COP (Wong
tease caspase-1 to alert and guide neighboring cells (Cypryk et al. 2017). It is believed that the direct damage of myocar-
et al. 2018). Accordingly, the present review summarizes the dial mitochondria from tissue hypoxia by COP increases the
most recent studies on the critical roles of exogenous CO and risk of developing myocardial diseases (Garg et al. 2018). In
the HO-1/CO system in inflammation and their underlying addition, COP could damage pancreatic function and lead
mechanisms. We emphasize both organ injuries induced by to a higher risk of DM. Takahashi et al. (1982) found that
high-dose CO exposure and modulation of HO-1 expression patients with 39.5% COHb manifested hyperamylasemia,
in cellular and organ protection, which implicate inflam- and the time course of amylase levels in COP patients is
mation as a key mediator of pathogenesis. In this regard, similar to that in acute pancreatitis. Another cohort study
studies on the NLRP3 inflammasome, the core mediator of also demonstrated that COP is a potential risk factor for
the inflammatory response, may indicate new targets for the developing subsequent DM, which is supported by the con-
treatment of various inflammatory diseases. We also dis- cept that the brain and endocrine system are tightly linked.
cuss currently available information about the interplay of Once the brain receives damage from COP, the endocrine

13

1144 Archives of Toxicology (2021) 95:1141–1159

system is likely to have problems producing or secreting in vivo and in vitro models (Gandini et al. 2001). Huang,
hormones; therefore, DM may develop (Huang et al. 2014; et al. (2019) found that patients with COP had an increased
Mimura et al. 1999). risk of myocardial injury (incidence rate ratio: 1.45; 95%
confidence interval [CI] 1.06–1.98). Histological findings,
Toxic mechanisms of CO: inflammation including focal areas of myocardial fiber degeneration with
and oxidative stress vacuolization and mononuclear cell infiltration, have also
been reported. The main cardiotoxic effects of CO are as
It is widely believed that the mechanism of CO toxicity follows: (1) left ventricular systolic and diastolic dysfunc-
involves rapid and strong binding of CO to Hb, thus decreas- tion, and wall motion abnormalities (Kim et al. 2020), (2)
ing the ability of H­ bO2 to unload O ­ 2 and the subsequent ECG changes, including arrhythmia, disturbances of repo-
oxygen-carrying capacity of the blood. However, tissue larization (e.g., T wave flattening and inversion), ischemic
hypoxia may not be the only mechanism of the toxic effects. changes (e.g., ST-segment depression or elevation), QT
Studies have indicated that CO may directly affect cellular interval prolongation, P wave elevation, QRS widening,
changes involving immunological, inflammatory, and oxi- and R wave depression (Cetin et al. 2011; DeBias et al.
dative stress-triggered damage via a variety of mechanisms 1976), and (3) myocardial injury (Huang et al. 2019). In
including: (1) binding to intracellular proteins such as heme- addition, a positive correlation was observed between
containing proteins, (2) nitric oxide generation and perox- ambient CO levels and hospitalization for congestive heart
ynitrite production, (3) lipid peroxidation by neutrophils, (4) failure (Gandini et al. 2001).
mitochondrial oxidative stress, (5) apoptosis (programmed
cell death), (6) immune-mediated injury, and (7) delayed
inflammation (Hampson et al. 2012). In addition, Akyol Cerebral injuries following COP
et al. (2014) dissected the involvement of a series of ROS
pathways in COP in distinct experimental studies. Accord- Hypoxia has been suspected to be the major mechanism of
ing to these studies, CO-induced damage is more widely brain injury in patients with COP. However, animal studies
derived from inflammation and oxidative stress instead of revealed that cerebral blood flow increases within minutes
hypoxia. CO promotes the formation of ROS throughout during CO exposure initially and remains elevated until
the body, possibly playing a critical role in the mechanism loss of consciousness (Lo et al. 2007). This indicates that
of CO toxicity (Han et al. 2007; Roderique et al. 2015; hypoxia may not explain all the effects of COP. Persistent
Tofighi et al. 2006). The burst in ROS may be accompanied neurological sequelae are noted in 46% of the COP survi-
by damages attributable to free radicals in patients (Thom vors. Delayed neurological sequelae, including cognitive and
1985). Indeed, it has been found that COP patients exhibited personality changes, incontinence, psychosis, and Parkinson-
increased serum total oxidative stress levels compared to ism, are found in 10–30% of the survivors between 2 days
healthy controls (17.14 vs. 8.47 μmol H ­ 2O2 equivalent/L) and 8 months following COP (Yogaratnam et al. 2011).
(Kavakli et al. 2011). Brain injuries following COP as noted by imaging include
diffuse hypoxic-ischemic encephalopathy and focal cortical
Target organs of CO after acute and chronic injury; necrosis of the globus pallidus; injury to other basal
exposures ganglia, thalamus, brainstem, and cerebellum; diffused brain
atrophy; and cerebral white matter demyelination (Lo et al.
Although cardiac and brain dysfunctions are recognized 2007). In addition to indirect effects through hypoxia, CO
as the major insults after COP, subsequent complications, can also affect the tissues in the brain directly. CO-induced
including endocrine disease, connective tissue disease, and mitochondrial oxidative phosphorylation may also produce
gastrointestinal disease, are exacerbated following COP. ischemic and anoxic brain injuries, leading to cognitive
Moreover, the respiratory system, the first target of CO deficits in survivors. Furthermore, COP enhanced the pro-
inhalation contact, may also be significantly affected during duction of nitric oxide by vascular endothelium, which may
COP. The following sections discuss the currently known form oxygen free radicals at proximal sites. Overall, these
clinical or experimental animal studies with regard to CO effects lead to mitochondrial dysfunction, capillary leakage,
intoxication. leukocyte sequestration, and apoptosis in brain (Oh and Choi
2015). Moreover, brain ischemia can result from excitotox-
Cardiotoxicity following COP icity, acidosis, calcium influx and depolarization, oxidative
stress, nitrative stress, inflammation, and increases in the
CO has cardiotoxic effects, including myocardial injury levels of excitatory amino acids such as glutamine, lead-
and subsequent cardiac dysfunction, which have been ing to elevated nitrate levels and subsequent injury to the
described in clinical and experimental studies using cerebral cortex (Piantadosi et al. 1997; Rose et al. 2017).

13
Archives of Toxicology (2021) 95:1141–1159 1145

COP‑induced gastrointestinal injury limited. A case report demonstrated that a 15-year-old


child suffering from COP manifested acute cardiopulmo-
The gut-brain axis has already been used to explain the rela- nary edema observed by chest X-ray (Wu et al. 2009).
tionship between gut dysfunction and neuropsychiatric dys- Many environmental studies showed that ambient CO
function (Singh et al. 2018a; b). Recent studies showed that may increase the risk of pulmonary disease (Losacco and
the incidence of gastrointestinal disease potentially increases Perillo 2018). A study in the US reported that CO was
via autonomic dysfunction following brain injury (Katzen- associated with emergency department visits for respira-
berger et al. 2015). Balzan et al. (1993) reported that intes- tory diseases overall and visits for asthma with a 3-day
tinal vasoconstriction due to COP made the gut much more lag (Slaughter et al. 2005). Another large study in China
vulnerable to the hypoxic effects of CO than the brain and reported that ambient CO contributed to pulmonary, car-
heart. In addition, Gennari and Alexander (1996) reported diovascular, genitourinary, gastrointestinal, and neuropsy-
that the ­pO2 levels of bowel might be an important regu- chiatric diseases, especially in the warm seasons (Wang
lator of gut barrier function. It is plausible that COP may et al. 2019). A meta-analysis of 87 eligible studies showed
contribute to the increased risk of gastrointestinal disease that 42 of them observed a positive association between
through hypoxia as well as immunological and inflammatory short-term CO exposure from ambient air pollutant and
damages (Weaver 2009). There have been case reports of emergency department visits/hospitalization for asthma
gastrointestinal diseases following CO exposure: a 24-year- with a pooled risk ratio of 1.045 (95% CI 1.029, 1.061;
old female who suffered from intestinal and hepatic ischemia I2 = 85.7%) associated with 1 mg/m3 increase and a popu-
(COHb: 34%) (Watson and Williams 1984), a 34-year-old lation attributable fraction of 4.3 (95% CI 2.8, 5.7) (Zheng
female who had diffuse colonic mural thickening and edema- et al. 2015). Cigarette smoke is another common source
tous friable pale mucosa from rectum to distal sigmoid colon of CO exposure. Aside from the numerous toxic chemi-
(Weaver and Deru 2016), an 83-year-old female who devel- cals in cigarette smoke, such as nicotine, tar, toluene, free
oped ischemic colitis (COHb: 19.7%) (Duenas-Laita et al. radicals, and pesticides, CO is a health hazard that should
2008), and a 61-year-old female with upper gastrointestinal not be overlooked. In smokers, COHb levels are generally
bleeding (COHb: 33.6%) (Tekin et al. 2019). The frequency in the 3–5% range; for heavy smokers, the COHb levels
of gastrointestinal bleeding is 103/100,000 per year in COP increase by approximately 20% (Hess 2017). In addition
patients. More importantly, there is solid evidence show- to the high levels of COHb in serum, cigarette smokers
ing increased intestinal permeability during gut damage via often exhibit pulmonary edema, barrier dysfunction, epi-
tight junction disintegration (Arrieta et al. 2006). Numer- thelial and endothelial cells changes, neutrophil infiltra-
ous studies have demonstrated intestinal damage following tion, and upregulation of IL-6 and CXCL-9 (Heijink et al.
COP via ischemia, inflammation, and gut junction disrup- 2012; Zeglinski et al. 2019). In addition, wood smoke has
tion, implying that microbiota manifestation and distribution been found to disrupt barrier dysfunction in the alveolar
may be an important feature during COP. On the other hand, epithelial cells via the mitogen-activated protein kinase
an experimental study in mice indicated that gastrointestinal (MAPK) signaling pathway (Zeglinski et al. 2019). These
inflammation could induce anxiety-like behavior and altered phenomena were directly involved in increasing systemic
biomarkers related to brain dysfunction, which may be due pulmonary capillary permeability after CO exposure.
to inflammation in the brain (Bercik et al. 2010). It seems Although CO is a fatal toxin with some potential proper-
that the brain-gut axis is bidirectional and involves more ties that require verification, few studies have explored
than just the sensory input that positively influences mood the underlying mechanisms through which CO leading to
and behavior. Therefore, the brain-gut and HPA axes may pulmonary dysfunction. Thus, pulmonary diseases caused
also play specific roles in the development of gut dysfunc- by COP should be studied more extensively.
tion following COP. However, the underlying mechanism In summary, studies have revealed that cognitive seque-
remains unclear. lae are not the only complications of COP. In addition to
the brain, other organs affected by COP include the heart,
Pulmonary diseases following CO inhalation lung, and intestines, as noted in clinical reports and ani-
mal experiments (Fig. 1). Whether the injured tissue can
Inhalation is the main route of CO exposure. CO diffuses transmit impaired signals to other locations or transmit
from alveoli to lung capillaries and subsequently competes the “eat me” signal to immune cells to reinforce the repair
with oxygen to bind Hb. Environmental CO comes from system is an important issue that should be further inves-
various sources, including industrial fuel-burning, motor tigated. Another issue of concern is whether organ-organ
vehicles, and cigarette smoke (5.9–17.4 mg CO/cigarette). communication is impaired by neuroendocrine or hormone
Studies on pulmonary diseases following acute COP are disruption after COP.

13

1146 Archives of Toxicology (2021) 95:1141–1159

Fig. 1  Schematic diagram of multiorgan damages induced by carbon with immune cell infiltration and lung integrity disruption, which is
monoxide poisoning (COP). Several organs manifest deficits after medicated by ERK pathway activation. As to the heart, a variety of
COP. The main target organs that are most often discussed in the cur- clinical syndromes of COP result in heart failure, including cardiac
rent literature include brain, heart, lung, and bowel (included intes- contractility exacerbation and myocardial injury. ECG abnormali-
tine and colon). In the brain, the most commonsyndrome is delayed ties have been observed following COP. In the bowel, ischemia and
neurological sequelae combining cognitive and personality disorders. hypoxia accompanied with inflammation are noted, and three clini-
These patients also exhibit necrosis and/or apoptosis in specific brain cal cases also revealed gut damage, even bleeding, after COP. Once
regions, such as the globus pallidus, thalamus, brainstem, cerebellum, inflamed, intestinal integrity is disrupted, and enteric microflora and
and basal ganglia. COP-induced hypoxia–ischemia damage causes their metabolites may enter the vascular or lymphatic system. In
encephalopathy. The mechanism involves CO directly binding to summary, hypoxia–ischemia, ROS generation, immune cell relative
cytochrome c in mitochondria, thereby generating ROS, and reducing responses, cell apoptosis, and necrosis are commonly observed after
ATP production. Fewer studies have assessed the underlying molecu- COP. The crosslink between target organs may influence each other
lar mechanism of damages to the lung after COP; nevertheless, a few through distinct pathways. Crosstalk between hormones and neuroen-
case reports indicated that COP patients exhibit acute cardiopulmo- docrine factors via the hypothalamus-pituitary gland-adrenal gland
nary edema. Prospective/retrospective studies using large data also (H–P–A) or hypothalamus-pituitary gland-thyroid gland (H–P–T)
demonstrated that ambient CO may increase the risk of pulmonary axis may play a vital role in organ-organ communication
diseases. Moreover, cigarette smokers have higher COHb blood levels

General biomarkers in COP level increased (> 0.1 mg/L) in 30% of patients with COP
and was positively correlated with the level of COHb.
Many investigators have searched for biomarkers that can Endogenous natriuretic peptides, such as brain natriuretic
predict complications and severity following COP. Some peptide (BNP), are cardiac hormones that act as markers
biomarkers, including creatine kinase (CK), CK-MB, tro- of left ventricle function. Because there are positive cor-
ponins, natriuretic peptides (NT-proANP, NT-proBNP, etc.), relations between BNP and COHb and between COHb and
and S100B, have been used to predict the toxicity of CO CK, BNP may be used as an early marker of cardiotoxicity
to the brain or heart (Gawlikowski et al. 2014). The most in COP (Davutoglu et al. 2006). Fatty acid-binding protein
frequently used enzymes are CK-MB and troponin I. Eren- (FABP), which is found in myocyte cytoplasm and released
ler et al. (2013) reported that troponin I and CK-MB lev- from injured myocardium, has been reported as a biomarker
els increased to 0.2 ng/mL (range 0–17.9 ng/mL; reference of COP. Erenler et al. (2013) reported that serum heart-type
value, 0.0–1.0 ng/mL) and 3.1 ng/mL (range 0.2–167 ng/ FABP increased in patients with severe COP. They also
mL; reference value, 0–3.2 ng/mL), respectively, in COP reported positive correlations between heart-type FABP
patients. Lippi et al. (2012) also found that the troponin I and the levels of COHb, troponin I, and CK-MB. However,

13
Archives of Toxicology (2021) 95:1141–1159 1147

these candidate biomarkers also increased in other brain genes. These effects collectively contribute to the anti-
and heart diseases, such as encephalopathy and ischemic/ inflammatory effect of CO (Knauert et al. 2013). (2) Anti-
hemorrhage shock, not specifically for COP. Another major apoptotic reactions that inhibit the cellular apoptosis path-
problem associated with these candidate COP biomarkers is way in mitochondria (Kim and Choi 2018). CO activates
that they typically only change in the first 12–24 h after CO the PI3K/Akt pathway to inhibit GSK-3β activity, which is
exposure and return to normal levels after 24 h (Kim et al. involved in numerous apoptotic signaling pathways (Kim
2016). Therefore, they are not suitable for predicting long- and Choi 2018; Kim et al. 2013; Zhang et al. 2005). (3)
term sequelae following COP. Vasodilatory effects that regulate blood vessel smooth mus-
cle tone via activation of the sGC/cGMP pathway (Kim and
Choi 2018). After activation of sGC, the subsequent increase
Therapeutic potential of CO and its in cGMP in smooth muscle cells leads to PKG activation,
underlying mechanisms induces the reuptake of ­Ca2+ by the sarcoplasmic reticulum,
and leads to reduced intracellular ­[Ca2+] and smooth muscle
The overall beneficial effects and mechanisms relaxation (Kim and Choi 2018).
triggered by CO In recent decades, many researchers have tried to use the
unique characteristics of CO to treat diseases. Most stud-
Although inhalation of high concentrations of CO may cause ies indicate that endogenous CO and exogenous CO gas
certain toxic effects, studies have discovered that CO has inhaled at doses such that the oxygen-carrying capacity of
certain beneficial effects and confirmed that CO exhibits hemoglobin is not severely compromised (COHb < 20%) can
potential benefits in medical applications. Initially, some provide protective and beneficial effects against organ injury,
clinical data revealed increased levels of CO in diseased inflammation disease, ischemia–reperfusion injury, and car-
patients, and the induction of HO-1 under stress conditions diovascular disease (Bauer and Pannen 2009).
strongly suggested that endogenously produced CO has
beneficial or therapeutic effects (Romao et al. 2012). This Application of CO in human diseases
hypothesis was later confirmed in various animal models
of human diseases using inhaled CO (Romao et al. 2012). In 1999, a model using hyperoxia-induced lung injury rats
For more convenient use of CO in medical applications, showed improved survival and alleviated lung injury after
CO-releasing molecules (CORMs) have been developed as exposure to low concentrations of CO (Otterbein et  al.
a new strategy that can provide CO in therapeutic applica- 1999). The study is probably the first in vivo evidence dem-
tions without significantly affecting COHb levels (Ismailova onstrating that low-dose gaseous CO exhibits therapeutic
et al. 2018). CORMs can release CO and mimic the medi- potential. CO exposure inhibits pro-inflammatory cytokines
cal effects of gaseous CO in animals (Romao et al. 2012). involved in the MKK3/p38 MAPK pathway to achieve anti-
Inhalation-based and CORM-based exogenous CO can be inflammatory and anti-apoptotic effects (Bauer and Pannen
used to induce HO-1 to metabolize heme into BV, ­Fe2+ and 2009). In addition, other types of lung injury (such as acute
CO (Duvigneau et al. 2019), and induction of HO-1 gene lung injury, asthma, and acute respiratory distress syndrome)
expression by genetic or pharmacological (e.g., heme deriva- have been assessed, and the protective effect of inhaled CO
tives and anesthetics) approaches represent new strategies to has been confirmed (Bauer and Pannen 2009; Foresti et al.
induce CO production (Abraham et al. 2016; Levy 2016). 2008; Ismailova et al. 2018).
CO is recognized as an important signal messenger that Given its anti-inflammatory effect, CO exhibits the poten-
can affect multiple pathways. Currently, the best-known tial as a treatment for inflammation-related diseases. For
therapeutic effects of CO are classified into three catego- example, systemic inflammatory response syndrome is an
ries: (1) Anti-inflammatory effects that inhibit the produc- inflammatory condition that affects the entire body. Inflam-
tion of pro-inflammatory cytokines (e.g., IL-6 and TNF-α) matory responses and organ damages induced by lipopoly-
and increase anti-inflammatory cytokines (e.g., IL-10) via saccharide (LPS) are models of systemic inflammation that
regulation of MAPK cascades, such as ERK1/2, JNK and have been used in multiple studies to verify the therapeutic
p38 pathways (Otterbein et al. 2000). CO also activates effect of CO (Bauer and Pannen 2009; Foresti et al. 2008;
Stat1/Stat3, PPAR-γ, and HIF-1α to inhibit the TGF-β and Ismailova et al. 2018; Nakahira and Choi 2015). In pigs and
Egr-1 genes (Hoetzel et al. 2008). Egr-1 is considered to be rodents treated with LPS, inhaled CO can reduce the LPS-
a multi-effect inflammatory trans-activator, regulating hun- induced cytokine responses and improve long-term survival
dreds of target genes, including IL-1β, IL-6, TNF-α, and through antioxidant, anti-inflammatory, and anti-apoptotic
ICAM-1. In addition, CO inactivates Toll-like receptor 4 effects (Bauer and Pannen 2009). Another study also showed
(TLR4) by downregulating NADPH oxidase activity. TLR4 that CORM-2 suppressed the thioredoxin-interacting pro-
induces the expression of inflammatory response-related tein/NLRP3 inflammasome pathway and protected against

13

1148 Archives of Toxicology (2021) 95:1141–1159

LPS-induced lung injury (Jiang et al. 2016). Wang et al. their metabolism in the context of processed glucose and
(2015) provided evidence that CORM-2 protected against other energy sources to promote survival and proliferation
sepsis-induced acute kidney injury and reduced TNF-α and (Schwartz et al. 2017). This change in metabolism is called
IL-1β levels, oxidative stress, and NLRP3 inflammasome the Warburg effect. CO can induce the anti-Warburg effect by
activation. forcing cancer cells to increase respiration, quickly fueling
Regarding ischemia–reperfusion injury, including the cancer cell’s bioenergy and eventually leading to meta-
transplantation, CO is considered to be a very promising bolic failure (Wegiel et al. 2013). As the role of the HO-1/
treatment given its anti-inflammatory and anti-apoptotic CO system in angiogenesis and metabolism in tumor growth
effects (Cheng and Rong 2017; Ismailova et al. 2018). In has been clarified, CO has also started to be perceived as an
experimental models of heart transplantation, CO reduced anti-tumor therapy strategy (Loboda et al. 2015).
ischemia–reperfusion injury and cardiac rejection of mouse Specifically, inhibition of tumor angiogenesis can prevent
to rat cardiac transplants via anti-apoptotic, anti-inflamma- tumor growth and even to cause tumor regression in various
tory, and vasodilatory mechanisms as well as suppression experimental models. CO influences the tumor angiogen-
of platelet aggregation and fibrinolysis (Bauer and Pannen esis response that depends on the production of cGMP and
2009; Soni et al. 2012). In the recipient, low-dose CO inha- the modulation of MAPK pathways (Pae et al. 2008). In
lation after transplantation effectively ameliorated heart addition, CO is involved in the angiogenic pathways driven
allograft rejection via downregulation of pro-inflammatory by the two major angiogenic mediators, VEGF and stromal
mediators (Cheng and Rong 2017). cell-derived factor-1, and improves endothelial cell viability
Additionally, CO has an inhibitory effect on the initia- and reactions (Dulak et al. 2008; Kourti et al. 2019; Lv et al.
tion and progression of heart and blood vessel diseases. CO- 2019).
induced vasodilation ameliorates acute coronary syndrome CO is also involved in the modulation of immune reac-
by expanding coronary arteries, especially in the case of tions. The effects of CO on tumor stroma and reprogram-
angina pectoris, and reducing myocardial cell death through ming of macrophages towards the anti-tumoral phenotype
anti-apoptotic and anti-inflammatory properties (Kim and are mediated by ROS-dependent activation of the MAPK/
Choi 2018). CO also stabilizes blood pressure in patients Erk1/2-c-myc pathway and Notch 1-dependent negative
with hypertension by regulating vascular tone. In patients feedback on the metabolic enzyme HO-1 (Nemeth et al.
with pulmonary hypertension and heart failure, CO relieves 2016). Furthermore, HO-1 and CO regulate T cell responses
pressure in the pulmonary and systemic vasculature (Kim by inhibiting dendritic cell function. CO pretreatment mark-
and Choi 2018). CO inhibits the proliferation of smooth edly suppressed intracellular ROS generation and the activi-
muscle cells in coronary arteries to avoid the formation of ties of NADPH oxidase and mitochondrial complexes I–IV
coronary atherosclerosis and prevents the occurrence of in T cells after stimulation. Moreover, increasing intracel-
coronary obstructive diseases (Kim and Choi 2018). lular ROS via hydrogen peroxide supplementation largely
In summary, the positive effects of CO are evident. To reversed the inhibitory effect of CO on T cell proliferation.
date, several clinical trials have been conducted to evalu- The inhibitory effects of CO on both cell proliferation and
ate the therapeutic effects of exogenous CO, e.g., treatment intracellular ROS production were also shown in a T cell
of acute respiratory distress syndrome caused by sepsis, proliferation model involving stimulation by allogeneic den-
reduction of postoperative ileus after colon resection, and dritic cells. In addition, CO treatment significantly inhibited
improvement of chronic inflammation in chronic obstructive T cell activation in vivo (Yan et al. 2020).
pulmonary disease (COPD) (Kim and Choi 2018). However, Various studies have explored the therapeutic effects of
it must be noted that due to the dose-dependent dual nature CO in various types of cancer, including breast cancer, non-
of CO, when CO and CORM are used for therapeutic pur- small cell lung carcinoma, pancreatic cancer, and prostate
poses, it is important to continuously monitor CO concen- cancer (Loboda et al. 2015). In this context, CO inhibited
trations and establish an accurate therapeutic range for safe ERK/AP-1 activation; HSP90 client protein activity, such
treatment (Ismailova et al. 2018). as AKT, ERα and cyclin D1; and the expression of cyclin
D1, CDK4, and hTERT. CO blocked the growth of breast
Application of CO in cancer therapy cancer cells independently of p53, which was destabilized,
ubiquitinated, and degraded in response to CO (Lee et al.
The dual role of CO transforms it from a toxic substance 2014). CO also significantly inhibits proliferation and inva-
to a therapeutic agent, and more interestingly, studies have sion and induces apoptosis in prostate cancer cells. LKB1
demonstrated that CO has anti-cancer effects. In contrast expression is upregulated after CO treatment, which also
to the mechanism triggered by CO to treat diseases, such increases p-AMPK levels and decreases p-mTOR. Further-
as inflammation, the application of CO in anti-cancer treat- more, LKB1 knockdown could weaken the effect of CO on
ment is based on other mechanisms. Cancer cells can change prostate cancer cells (Yan et al. 2018).

13
Archives of Toxicology (2021) 95:1141–1159 1149

Overall, the anti-proliferative properties of CO may Role of the HO‑1/CO system


indeed have chemotherapeutic potential against certain can- in inflammasome regulation
cers. In controlled doses, CO not only mimicked the effects
of chemotherapy agents by blocking proliferation of cancer Detoxification effects of the heme oxygenase family
cells, but also amplified the toxic effects of the chemother- and interacting regulators
apy drugs to accelerate cancer cell death (Li et al. 2019;
Loboda et al. 2015; Wegiel et al. 2013). More importantly, HO is the primary and rate-determining enzyme in the heme
a common side effect of cancer treatment is the injury of catabolic pathway, which plays a crucial role in protecting
normal tissues by chemotherapy agents, and CO can pro- cells from heme-induced oxidative stimuli. In general, HO
tect normal tissues while fighting cancer cells (Wegiel et al. breaks down the heme group and produces endogenous
2013). This finding adds to the feasibility of using CO for CO to ameliorate oxidative stress and the inflammatory
anti-cancer treatment (Fig. 2). response, including regulation of the inflammasome system
(Gomperts et al. 2017; Liu et al. 2019; Waza et al. 2018;
Wu et al. 2019). There are three distinct isoforms of HO:
HO-1, HO-2, and HO-3 (Gáll et al. 2019; Waza et al. 2018).
Among them, HO-1 is the focus of most research, because
its expression is enhanced under various pathophysiologi-
cal conditions (Waza et al. 2018). HO-1 can be induced by
a wide variety of stimuli, such as hypoxia, oxidative stress,
cytokines, LPS, and heavy metals in biological systems

Fig. 2  Protective effects and potential therapeutic applications of inflammatory responses by several established mechanisms, including
carbon monoxide. CO is a heme ligand that can bind to and poten- the p38 MAPK-dependent downregulation of inflammation and apop-
tially modulate the cellular activity several heme-containing proteins. tosis effects. CO can activate PPAR-γ to inhibit Egr-1 gene to down-
Regarding anti-cancer effects, many studies have discussed the thera- regulate inflammation. Recent studies also indicate that low-level CO
peutic effects of CO on various cancer types. HO-1 expression inhib- exposure affects mitochondria inhibition and can alter the autophagy
its cancer cell proliferation. This effect might be mediated by CO, pathway induction. Activated autophagy flux might downregulate the
which inhibits ERK/AP-1 activation and HSP90 client protein activ- NLRP3 inflammasome and pyroptosis pathway in response to pro-
ity, such as cyclin D1, CDK, and hTERT. Inhibition of p53 protein in inflammatory stimuli
response to CO treatment was also reported. CO can also modulate

13

1150 Archives of Toxicology (2021) 95:1141–1159

(Waza et al. 2018). The alterations in the HO-1 level are consists of pattern recognition receptors, the adapter protein,
associated with the pathogenesis of some age-dependent apoptosis-associated speck-like protein containing caspase-
disorders, including neurodegeneration, inflammatory dis- recruitment domain (ASC), and the protease caspase-1.
orders, and cancer (Wegiel et al. 2014). Since HO-1 induc- NLRP3 (NACHT, LRR and PYD domains-containing pro-
tion is widely recognized as a mechanism used by cells to tein 3) is an intracellular recognition receptor that can detect a
neutralize stress conditions, the induction of this powerful broad range of microbial factors, endogenous danger signals,
enzyme becomes a therapeutic strategy against different dis- and environmental irritants, resulting in the formation and acti-
eases, arising as a result of inappropriate immune response vation of the NLRP3 inflammasome (Swanson et al. 2019).
and oxidative dysregulation. Particularly, the identifica- Assembly of the NLRP3 inflammasome leads to release of
tion of noncytotoxic HO-1 inducers may represent a novel caspase 1, maturation of pro-inflammatory cytokines IL-1β
approach to cope against various oxidative and inflammatory and IL-18, and gasdermin D-mediated pyroptotic cell death
responses (Son et al. 2013). (Shao et al. 2015). In innate immune activation, the inflamma-
Earlier studies indicate that different HO-1 inducers acti- some plays a critical role by activating cytosolic multiprotein
vate diverse protein phosphorylation-dependent signaling complexes that induce inflammatory cytokine production after
pathways that ultimately regulate HO-1 gene expression by sensing invading pathogens (Shao et al. 2015). Recent studies
activating a wide variety of transcription factors, such as demonstrated that heme preconditioning of HO-1 induction
PI3K/Akt, MAPK, and Nrf2 (Chen et al. 2013). For exam- reduced IL-1β maturation and downregulated inflammasome
ple, Hemin is the substrate and a well-known inducer of activation in a model of sepsis-associated lung injury (Luo
HO-1. A recent study showed that it provided protection et al. 2014; Ryter 2019).
against lead neurotoxicity by HO-1/CO activation (Ye et al. The actual underlying mechanism of the anti-inflammatory
2018). Multiple functions of HO-1 have been confirmed in properties of HO-1 in CO exposure is not fully understood.
biological systems, and its protective effects could involve However, the signaling action of CO together with the anti-
its ability to degrade heme (Gáll et al. 2019; Waza et al. oxidant properties of BV/BR and iron sequestration by fer-
2018), whereby the byproducts of heme degradation (mostly ritin contributes to the amelioration of inflammation (Loboda
CO and BR) mediate its cytoprotective effects. Interestingly, et al. 2016). HO-1 expression inhibits ovalbumin-induced air-
evidence indicates that extensive exogenous CO can also way inflammation in asthma via downregulation of NLRP3-
disrupt the HO-1/CO system. Acute COP in rats reduced dependent regulation of retinoid-X receptor (RXRa/b/g) acti-
HO enzyme activity, whereas HO-1 protein expression vation (Lv et al. 2018). Moreover, in an NLRP3 inflammasome
was consistently upregulated in a time-dependent manner. activation model, the application of CO gas inhibits activation
Endogenous CO produced by activated HO-1 is increased in of caspase-1 and secretion of IL-1β and IL-18 in bone marrow-
the hippocampus of the rat brain, which is potentially associ- derived macrophages in response to LPS and ATP treatment
ated with brain injury following COP (Guan et al. 2011). In (Cronje et al. 2004; Lee et al. 2017a). Although the precise
general conditions, normal brain tissue exhibits basal expres- mechanisms still require further clarification, HO-1 and CO
sion of HO-1 and Nrf-2 proteins; however, COP activates exhibit the potential of functioning as novel regulators of the
the HO-1/Nrf-2 pathway and induces oxidative stress, caus- NLRP3 inflammasome and secretion of IL-1β and IL-18 in
ing brain lesions. Various drugs can be used to upregulate inflammatory conditions (Chen et al. 2013).
the expression of HO-1 and Nrf-2, including those that may CO exposure at different concentrations exerts both anti-
increase endogenous biological CO, inhibit oxidative stress inflammation and inflammation effects via cellular responses.
and apoptosis, or activate the Keap1-Nrf/ARE pathway to Exposure to high concentrations of CO might activate the
improve neurophysiologic changes following COP (Guan inflammation through the NLRP3 inflammasome by affect-
et al. 2009; Li et al. 2016). ing mitochondria and upregulating mtROS, thereby inducing
the HO-1 system to alleviate the inflammation. In contrast, at
The HO‑1/CO system exerts cytoprotective effects low concentrations, CO acts as an efficient anti-inflammatory
by regulating the inflammasome system agent through inducing the HO-1 system.

CO exposure rapidly increases brain cytosolic heme levels


through three mechanisms: (1) alteration in heme synthesis, a
process that is regulated by CO; (2) release of heme from dam-
aged cellular proteins; and (3) disturbance in mitochondrial
heme storage by CO (Rose et al. 2017) (Fig. 3). In addition
to these three major mechanisms, research suggests a poten-
tial regulatory role of HO-1/CO in the inflammasome system,
especially the NLRP3 inflammasome. The inflammasome

13
Archives of Toxicology (2021) 95:1141–1159 1151

Fig. 3  Mitochondrial effects of CO poisoning. Under normal condi- petitively binds to Hb with O­ 2, reducing its total O
­ 2 carrying capacity
tions, hemoglobin (Hb) binds oxygen (­O2) and delivers it to periph- and affecting cytochrome proteins. This action causes the following:
eral tissues. In the cytosol, mitochondria produce energy by con- (1) inhibition of the reduction of ­O2 to water (the end destination of
suming oxygen. Complex IV receives the electron transferred by electrons in the electron transport chain), (2) inhibition of the transfer
cytochrome c. Electrons are first transferred to cytochrome c oxidase of ­H+ into the intermembrane space, shutting down ATP generation
(COX) subunit 1 (­Cua). The electron reduces O ­ 2 at subunit 2 (Cyta3 through ATP synthase, and (3) accumulation of electrons entering the
and ­Cub), forming water and transporting a proton (­H+) through the electron transport chain through complexes I and III, which generates
mitochondrial intermembrane space. Upon exposure to CO, CO com- ROS and leads to deleterious effects

The possible role of CO in the interplay human alveolar (A549) and human bronchial epithelial cells
among the inflammasome, autophagy (Lee et al. 2011). Formation of mtROS might be an activa-
and exosomes tion signal of the inflammasome (Shao et al. 2015). Given
that these two pathways exhibit distinct effects and occasion-
Crosstalk between autophagy and inflammasome ally crosstalk, we argue that CO-mediated cellular responses
triggered by CO could be mediated by the interaction between autophagy and
the inflammasome.
Growing evidence has shown that autophagy plays important Many studies have indicated that autophagy inhibits
roles in various biological processes. Autophagy is induced inflammasome activation, thereby inhibiting inflammatory
at a low frequency under steady-state conditions and con- responses and reducing inflammatory injuries in a variety
tributes to cellular homeostasis by promoting the turnover of diseases (Chen et al. 2016). The mechanism through
of organelles, such as mitochondria (Dikic and Elazar 2018). which autophagy inhibits NLRP3 inflammasome could be
CO exposure might activate autophagy through mtROS for- related to the reduction of NLRP3 components (NLRP3,
mation (Lee et al. 2011). It has been demonstrated that CO ASC), sequestration and degradation of pro-IL-1β, and the
exposure time-dependently increased the expression and clearance of mtROS (Cao et al. 2019; Liu et al. 2018). A
activation of an autophagic protein, microtubule-associated recent study indicated that immunity-related GTPase M,
protein-1 light chain-3B, in mouse lung and in both cultured which belongs to a family of interferon-inducible GTPases

13

1152 Archives of Toxicology (2021) 95:1141–1159

involved in resistance to intracellular pathogens, could medi- modulating the expression of NLRP3 and ASC (Xue et al.
ate selective autophagic degradation of NLRP3 and ASC 2019). Taken together, the crosstalk between the inflam-
to protect gut inflammation in a Crohn’s disease model masome and autophagy is complex and bidirectional with
(Mehto et al. 2019). Moreover, autophagy was also reported context-dependent features. The relationship and underlying
to limit inflammation by downregulating caspase 1-depend- mechanisms between autophagy and the inflammasome in
ent inflammasome cleavage of pro-IL-1β to the active form response to CO exposure need to be further investigated.
(Ilyas et al. 2016). HO-1 generated CO is required for the
induction of autophagy and killing of pathogens resid- Exosomes exhibit interplay
ing in macrophages in response to immunomodulation by with both the inflammasome and autophagy
interferon-γ (Singh et al. 2018a, b). In addition, the accu-
mulation of higher concentrations of mitochondrial damage- In addition to its well-known role in processing pro-inflam-
induced mtROS in response to autophagy/mitophagy inhibi- matory cytokines, the inflammasome can also serve as a
tion was associated with NLRP3 inflammasome activation universal regulator of protein secretion, which allows the
and the release of IL-1β by macrophages (Rocha et al. 2020). release of various proteins from cells exposed to danger
Notably, a recent study indicated a novel interaction pathway signals to alert and guide neighboring cells. These pro-
among actin assembly, autophagy, and the inflammasome teins are mostly segregated in membrane-enclosed com-
activation (Lee et al. 2017a, b). Lee et al. (2017a, b) showed partment and secreted in nano-sized extracellular vesicles
that deficiency of Wiskott-Aldrich syndrome protein, which named exosomes (Cypryk et al. 2018). The impact of this
signals to the actin cytoskeleton and causes defective bac- type of intercellular communication in the clinical setting
terial clearance, excessive inflammasome activation, and has recently been recognized. For instance, exosomes iso-
host cell death, plays a crucial role in impaired autophagic lated from body fluids of patients have been proposed to
p62/LC3 recruitment and defective formation of canonical serve as disease or early diagnosis biomarkers of inflamma-
autophagosomes. Taken together, autophagy is generally tory-related diseases, such as atherosclerosis, Alzheimer’s
recognized as a negative feedback regulator of inflammation. disease, and rheumatoid arthritis (Cypryk et al. 2018). It
Thus, in the context of autophagic dysfunction, damaged has been shown that the immune response-related pro-
mitochondria, proteins, and ROS can be accumulated and teins and signaling pathways were specifically enriched in
then trigger the novel signaling pathways described above, inflammasome-derived exosomes and directly activated the
which may be induced to promote inflammasome activation. NF-κB signaling pathway (Zhang et al. 2017). Inflamma-
A mutual relationship between autophagy and the inflam- tory signaling could be amplified in neighbor cells in an
masome has been reported recently. Although autophagy exosome-dependent manner. Thus, inflammasome-derived
was assumed to act as a negative regulator of the NLRP3 exosomes might be used to augment the immune response,
inflammasome, the NLRP3 inflammasome also positively and prevention of the transfer of these exosomes might
regulates the activation of autophagy. Deng et al. (2016) ameliorate immune-related diseases. In addition, inflamma-
showed that, probably for the first time, Pseudomonas some-derived exosomes are preferentially taken up by mac-
aeruginosa infection induced the assembly of the NLRP3 rophages, leading to the upregulation of NLRP3, process-
inflammasome and secretion of caspase 1 and IL-1β. Silenc- ing of caspase-1/IL-1β, and triggering of pyroptosis (Abal
ing of the NLRP3 inflammasome impaired the elimination 2017; Wu et al. 1866). Growing evidence also indicates
of P. aeruginosa in macrophages by reducing the level of that exosome-delivered miRs undergo functional transfer
IL-1β/IL-18 secretion and autophagy (Deng et al. 2016). among immune cells and constitute a mechanism of regula-
The finding is consistent with a previous study indicat- tion of the inflammatory response (Alexander et al. 2015).
ing that activation of caspase 1 could promote adaptive A recent report demonstrated that M2 macrophage-derived
responses to oxidative stress and increase autophagic exosomes could carry miR-148a to alleviate myocardial
influx by upregulating Beclin 1 (Sun et al. 2013). Similar ischemia/reperfusion injury through inhibition of the TLR4/
to the abovementioned study, a study using human mono- NF-κB/NLRP3 (Dai et al. 2020). It has also been shown
cytic cell line (THP-1)-derived macrophages showed that that miR-9 inhibits NLRP3 inflammasome activation via the
activation of the NLRP3 inflammasome may serve as an JAK1/STAT1 pathway, and inhibition of miR-9 upregulates
immune defense strategy against T. pallidum and mycobac- NLRP3 inflammasome and caspase-1 expression (Jeyabal
teria infection through modulation of autophagy (Xu et al. et al. 2016; Wang et al. 2017).
2020). In addition, it was found that ncRNAs (including Recent studies have revealed that exosome biogenesis
lincRNA) played an important role in the crosstalk between and autophagy share similar molecular machinery, and
the NLRP3 inflammasome and autophagy (Xue et al. 2019). these processes can be regulated by each other and play
It is also reported that lincRNA-Cox2 could bind to NF-κB important roles in health and disease. However, the cross-
p65 and promote its nuclear translocation and transcription, regulation between these pathways is highly complicated

13
Archives of Toxicology (2021) 95:1141–1159 1153

(Xu et al. 2018). For instance, IL-1β secretion is inhibited cellular proliferation programs. An emerging concept is
under basal autophagy and can be activated when autophagy that mitochondria may represent a common target of CO
is induced in macrophages (Nakahira et al. 2011). Under and lead to subsequent mitochondria-derived mediators
starvation conditions, IL-1β secretion, which lacks the signal and mtROS generation, which may be associated with
peptide sequence to follow the conventional ER-regulated the regulation of downstream signaling effects of CO (Lo
secretion pathway, is dramatically increased by the coor- Iacono et al. 2011; Ryter et al. 2018) (Fig. 4). Mitochon-
dinated action of ATG5, the inflammasome, GRASP, and dria play a central role in many cellular functions, ranging
Rab8a via a mechanism known as autophagy-based uncon- from energy status to cell fate. Thus, once mitochondria
ventional secretion (Villarroya-Beltri et al. 2016). A recent dysfunction occurs, ROS are induced, and various path-
review article concluded that exosomal trafficking can play ways are activated, including the NLRP3 inflammasome,
a beneficial or harmful role in disease through the regulation pyroptosis, apoptosis, autophagy, and exosome release
of autophagy (Lin et al. 2019). In addition, it showed that pathways. Thus, modulation of cell mitochondria by CO
autophagy plays a vital role in disease by regulating the gen- is a relevant subject for studying CO toxicity through the
eration of exosomes and that the cross-regulation between downstream effects of mitochondria. This notion high-
exosomal and autophagic pathways may represent a promis- lights the need for further research into the signaling
ing target for disease prevention and treatment. Accordingly, pathways which activate the NLRP3 inflammasome that
the exosome might serve as a bridge between the inflamma- can, therefore, be targeted by appropriate inhibitors. More
some and autophagy activation under the stress conditions, studies are urgently needed.
including CO exposure. However, the detailed mechanisms The successful demonstration of inhaled CO in preclini-
remain unclear and require further investigation. cal animal models has led to continued efforts toward the
clinical application of CO, either through inhaled CO or
pharmacological delivery by CORMs. A recent clinical
Conclusions and perspectives trial demonstrated the feasibility of administering inhaled
CO to patients with COPD without any adverse effects. In
CO intoxication is the most common human poisoning addition, the HO-1/CO system is implicated in numerous
caused by gases and has a high mortality rate. In addition, cellular protective pathways. Therefore, there is a need
many survivors suffer from long-term morbidity involv- to reveal the link between the HO-1 and candidate genes
ing multiple organ systems. The pathophysiology of COP involved in various diseases to identify the actual role of
involves both the reduction of oxygen delivery and the HO-1. This information will eventually help in making
inhibition of mitochondrial respiration (Borzelleca 2000; HO-1 a potential therapeutic target for the amelioration
Rose et al. 2017). Excessive CO exposure disrupts the elec- of various stress-related diseases.
tron transport chain in mitochondria and produces ROS to In the context of the three-party interplay described
cause damages to cells or tissues (Fig. 3). COP-induced tis- above, the exosome might have a pivotal impact on disease
sue hypoxia is recognized as the major etiology of related by mediating autophagy and the inflammasome. However,
sequelae such as neurological symptoms, pulmonary edema in contrast to the mechanisms involved in autophagy and
with respiratory failure, decreased myocardial contractil- inflammasomes, much less is known about how exosome
ity, arrhythmias, heart failure, and renal failure (Kinoshita regulates these processes under inflammatory conditions
et al. 2020). In addition, CO may directly lead to cellular related to CO exposure. Therefore, novel findings involving
changes involving immunological, inflammatory, and oxida- mechanistic understanding of the modes of action for CO-
tive stress-triggered damage via several defined mechanisms regulated pathways, including the abovementioned mecha-
(Hampson et al. 2012). Although cardiac and brain dysfunc- nisms of NLRP3 inflammasome activation, autophagy, and
tions are recognized as the major health effects occurring exosome release, are essential for the development of new
after COP, subsequent complications are also noted in other therapeutic strategies and biomarkers for COP.
organ systems, including endocrine, gastrointestinal, and We have thoroughly reviewed the beneficial and toxic
respiratory systems. However, current COP biomarkers only effects of CO, as well as the molecular pathways regulated
reflect acute CO exposure, and some of them are lack of by CO in both scenarios. Given that the mitochondrion is
specificity. Thus, the identification of proper biomarkers to the major cellular target of CO exposure, it is worth men-
predict complications and their severity following COP is a tioning again that further evaluating its downstream sign-
main research goal in this field in the near future. aling pathways could provide a solid basis for CO-based
The cellular signaling networks modulated by CO therapy against diseases, a solution for COP treatment, and
include several signal transduction pathways associ- a biomarker for COP monitoring (Fig. 4).
ated with the regulation of inflammation, cell death, and

13

1154 Archives of Toxicology (2021) 95:1141–1159

Fig. 4  Possible mechanisms of regulation of inflammation in CO Mitochondria play a central role in many cellular functions. When
toxicity. CO exerts direct effects by inhibiting mitochondria and mitochondria dysfunction occurs, ROS could be induced, and vari-
heme-like proteins, such as cytochrome c oxidase, promoting mito- ous signaling pathways are activated. Potential mechanisms involve
chondria ROS production and modulating mitochondria metabolism. NLRP3 inflammasome activation, autophagy, and exosome release

Acknowledgements This work was supported by the Minis- Abraham NG, Junge JM, Drummond GS (2016) Translational signifi-
try of Science and Technology, Taiwan through Grants MOST cance of heme oxygenase in obesity and metabolic syndrome.
108-2638-B-006-001-MY2, MOST 107-2311-B-006-004-MY3, MOST Trends Pharmacol Sci 37(1):17–36. https​://doi.org/10.1016/j.
108-2314-B-384-010, MOST 109-2314-B-384-005-MY3, MOST 108- tips.2015.09.003
2314-B-039-061–MY3, MOST 109-2314-B-006-051-MY3, and MOST Akyol S, Erdogan S, Idiz N et al (2014) The role of reactive oxy-
108-2314-B-006-057. Figure 1 were created using BioRender.com. gen species and oxidative stress in carbon monoxide toxicity:
an in-depth analysis. Redox Rep 19(5):180–189. https​://doi.
Author contributions  R-JC writing—original draft, writing—review org/10.1179/13510​00214​Y.00000​00094​
and editing. Y-HL writing—original draft, writing—review and edit- Alexander M, Hu R, Runtsch MC et al (2015) Exosome-delivered
ing. T-HC writing—original draft. Y-YC writing—original draft. Y-LY microRNAs modulate the inflammatory response to endotoxin.
writing—original draft. C-PC conceptualization. C-CH conceptual- Nat Commun 6:7321. https​://doi.org/10.1038/ncomm​s8321​
ization. H-RG conceptualization, writing—review and editing. Y-JW Arrieta MC, Bistritz L, Meddings JB (2006) Alterations in intestinal
conceptualization, writing—original draft, writing—review and edit- permeability. Gut 55(10):1512–1520. https​://doi.org/10.1136/
ing, supervision. gut.2005.08537​3
Balzan M, Cacciottolo JM, Casha A (1993) Intestinal infarction follow-
ing carbon monoxide poisoning. Postgrad Med J 69(810):302–
Compliance with ethical standards  303. https​://doi.org/10.1136/pgmj.69.810.302
Bauer I, Pannen BH (2009) Bench-to-bedside review: carbon monox-
Conflict of interest  The authors declare that they have no known com- ide-from mitochondrial poisoning to therapeutic use. Crit Care
peting financial interests or personal relationships that could have ap- 13(4):220. https​://doi.org/10.1186/cc788​7
peared to influence the work reported in this paper. Bercik P, Verdu EF, Foster JA et  al (2010) Chronic gastrointesti-
nal inflammation induces anxiety-like behavior and alters
central nervous system biochemistry in mice. Gastroenter-
ology 139(6):2102-2112.e1. https​: //doi.org/10.1053/j.gastr​
References o.2010.06.063
Borzelleca JF (2000) Paracelsus: herald of modern toxicology. Toxicol
Abal M (2017) Characterizing the contribution of inflammasome- Sci 53(1):2–4. https​://doi.org/10.1093/toxsc​i/53.1.2
derived exosomes in the activation of the immune response. Ann Cao Z, Wang Y, Long Z, He G (2019) Interaction between autophagy
Transl Med 5(7):172. https​://doi.org/10.21037​/atm.2017.03.48 and the NLRP3 inflammasome. Acta Biochim Biophys Sin

13
Archives of Toxicology (2021) 95:1141–1159 1155

(Shanghai) 51(11):1087–1095. https​://doi.org/10.1093/abbs/ Duvigneau JC, Esterbauer H, Kozlov AV (2019) Role of heme oxyge-
gmz09​8 nase as a modulator of heme-mediated pathways. Antioxidants
Cetin M, Ornek E, Murat SN, Cetin ZG, Oksuz F, Gokcen E (2011) A (Basel) 8(10):475. https​://doi.org/10.3390/antio​x8100​475
case of carbon monoxide poisoning presenting with supraven- Erenler AK, Yardan T, Baydin A, Gunay M, Amanvermez R (2013)
tricular tachycardia. Intern Med 50(21):2607–2609. https​://doi. Heart-type fatty acid-binding protein as a potential biomarker of
org/10.2169/inter​nalme​dicin​e.50.5929 acute carbon monoxide poisoning. Am J Emerg Med 31(8):1165–
Chang CC, Lee YC, Chang WN et al (2009) Damage of white mat- 1169. https​://doi.org/10.1016/j.ajem.2013.04.005
ter tract correlated with neuropsychological deficits in carbon Foresti R, Bani-Hani MG, Motterlini R (2008) Use of carbon monoxide
monoxide intoxication after hyperbaric oxygen therapy. J Neuro- as a therapeutic agent: promises and challenges. Intensive Care
trauma 26(8):1263–1270. https:​ //doi.org/10.1089/neu.2008-0619 Med 34(4):649–658. https:​ //doi.org/10.1007/s00134​ -008-1011-1
Chen H-G, Xie K-L, Han H-Z et al (2013) Heme oxygenase-1 mediates Gáll T, Balla G, Balla J (2019) Heme, heme oxygenase, and endo-
the anti-inflammatory effect of molecular hydrogen in LPS-stim- plasmic reticulum stress—a new insight into the pathophysiol-
ulated RAW 264.7 macrophages. Int J Surg 11(10):1060–1066. ogy of vascular diseases. Int J Mol Sci 20(15):3675. https​://doi.
https​://doi.org/10.1016/j.ijsu.2013.10.007 org/10.3390/ijms2​01536​75
Chen RJ, Lee YH, Yeh YL, Wang YJ, Wang BJ (2016) The roles of Gandini C, Castoldi AF, Candura SM et al (2001) Carbon monoxide
autophagy and the inflammasome during environmental stress- cardiotoxicity. J Toxicol Clin Toxicol 39(1):35–44. https​://doi.
triggered skin inflammation. Int J Mol Sci 17(12):2063. https​:// org/10.1081/clt-10010​2878
doi.org/10.3390/ijms1​71220​63 Garg J, Krishnamoorthy P, Palaniswamy C et al (2018) Cardiovas-
Cheng Y, Rong J (2017) Therapeutic potential of heme oxyge- cular abnormalities in carbon monoxide poisoning. Am J Ther
nase-1/carbon monoxide system against ischemia-reperfu- 25(3):e339–e348. https​://doi.org/10.1097/MJT.00000​00000​
sion injury. Curr Pharm Des 23(26):3884–3898. https​://doi. 00001​6
org/10.2174/13816​12823​66617​04131​22439​ Gawlikowski T, Golasik M, Gomolka E, Piekoszewski W (2014)
Cheng X, Ku CH, Siow RC (2013) Regulation of the Nrf2 antioxi- Proteins as biomarkers of carbon monoxide neurotoxicity.
dant pathway by microRNAs: new players in micromanaging Inhal Toxicol 26(14):885–890. https​://doi.org/10.3109/08958​
redox homeostasis. Free Radic Biol Med 64:4–11. https​://doi. 378.2014.97078​6
org/10.1016/j.freer​adbio​med.2013.07.025 Gennari R, Alexander JW (1996) Effects of hyperoxia on bacterial
Cronje FJ, Carraway MS, Freiberger JJ, Suliman HB, Piantadosi CA translocation and mortality during gut-derived sepsis. Arch Surg
(2004) Carbon monoxide actuates ­O2-limited heme degradation 131(1):57–62. https​://doi.org/10.1001/archs​urg.1996.01430​
in the rat brain. Free Radic Biol Med 37(11):1802–1812. https​ 13005​9010
://doi.org/10.1016/j.freer​adbio​med.2004.08.022 Goebel U, Wollborn J (2020) Carbon monoxide in intensive care med-
Cypryk W, Nyman TA, Matikainen S (2018) From inflammasome icine-time to start the therapeutic application?! Intensive Care
to exosome-does extracellular vesicle secretion constitute an Med Exp 8(1):2. https​://doi.org/10.1186/s4063​5-020-0292-8
inflammasome-dependent immune response? Front Immunol Gomperts E, Belcher JD, Otterbein LE et al (2017) The role of carbon
9:2188. https​://doi.org/10.3389/fimmu​.2018.02188​ monoxide and heme oxygenase in the prevention of sickle cell
Dai Y, Wang S, Chang S et al (2020) M2 macrophage-derived exosomes disease vaso-occlusive crises. Am J Hematol 92(6):569–582.
carry microRNA-148α to alleviate myocardial ischemia/reper- https​://doi.org/10.1002/ajh.24750​
fusion injury via inhibiting TXNIP and the TLR4/NF-kappaB/ Guan L, Wen T, Zhang Y, Wang X, Zhao J (2009) Induction of heme
NLRP3 inflammasome signaling pathway. J Mol Cell Cardiol oxygenase-1 with hemin attenuates hippocampal injury in rats
142:65–79. https​://doi.org/10.1016/j.yjmcc​.2020.02.007 after acute carbon monoxide poisoning. Toxicology 262(2):146–
Davutoglu V, Gunay N, Kocoglu H et al (2006) Serum levels of NT- 152. https​://doi.org/10.1016/j.tox.2009.06.001
ProBNP as an early cardiac marker of carbon monoxide poison- Guan L, Zhang YL, Wen T, Wang XF, Zhu MX, Zhao JY (2011)
ing. Inhal Toxicol 18(2):155–158. https:​ //doi.org/10.1080/08958​ Dynamic changes of heme oxygenase-1 in the hippocampus
37050​03058​85 of rats after acute carbon monoxide poisoning. Arch Environ
DeBias DA, Banerjee CM, Birkhead NC, Greene CH, Scott SD, Harrer Contam Toxicol 60(1):165–172. https​://doi.org/10.1007/s0024​
WV (1976) Effects of carbon monoxide inhalation on ventricu- 4-010-9524-3
lar fibrillation. Arch Environ Health 31(1):42–46. https​://doi. Hall JE (2010) Guyton and hall textbook of medical physiology e-book,
org/10.1080/00039​896.1976.10667​188 12th edn. Elsevier Health Sciences, Jordan
Deng Q, Wang Y, Zhang Y et al (2016) Pseudomonas aeruginosa trig- Hampson NB, Hauff NM (2008) Risk factors for short-term mortality
gers macrophage autophagy to escape intracellular killing by from carbon monoxide poisoning treated with hyperbaric oxy-
activation of the NLRP3 inflammasome. Infect Immun 84(1):56– gen. Crit Care Med 36(9):2523–2527. https​://doi.org/10.1097/
66. https​://doi.org/10.1128/iai.00945​-15 CCM.0b013​e3181​8419d​8
Deretic V (2011) Autophagy in immunity and cell-autonomous defense Hampson NB, Piantadosi CA, Thom SR, Weaver LK (2012) Prac-
against intracellular microbes. Immunol Rev 240:92–104. https​ tice recommendations in the diagnosis, management, and pre-
://doi.org/10.1111/j.1600-065X.2010.00995​.x vention of carbon monoxide poisoning. Am J Respir Crit Care
Dikic I, Elazar Z (2018) Mechanism and medical implications of mam- Med 186(11):1095–1101. https​://doi.org/10.1164/rccm.20120​
malian autophagy. Nat Rev Mol Cell Biol 19(6):349–364. https​ 7-1284C​I
://doi.org/10.1038/s4158​0-018-0003-4 Han ST, Bhopale VM, Thom SR (2007) Xanthine oxidoreductase
Duenas-Laita A, Mena-Martin FJ, Roquelai-Ruiz P, Gil-Simon P, Bar- and neurological sequelae of carbon monoxide poisoning.
rio-Andres J, Martin-Escudero JC (2008) Ischemic colitis associ- Toxicol Lett 170(2):111–115. https​://doi.org/10.1016/j.toxle​
ated with acute carbon monoxide poisoning. Clin Toxicol (Phila) t.2007.02.006
46(8):780–781. https​://doi.org/10.1080/15563​65070​15462​27 Heijink IH, Brandenburg SM, Postma DS, van Oosterhout AJ (2012)
Dulak J, Deshane J, Jozkowicz A, Agarwal A (2008) Heme oxyge- Cigarette smoke impairs airway epithelial barrier function and
nase-1 and carbon monoxide in vascular pathobiology: focus cell-cell contact recovery. Eur Respir J 39(2):419–428. https​://
on angiogenesis. Circulation 117(2):231–241. https ​ : //doi. doi.org/10.1183/09031​936.00193​810
org/10.1161/CIRCU​LATIO​NAHA.107.69831​6

13

1156 Archives of Toxicology (2021) 95:1141–1159

Hess DR (2017) Inhaled carbon monoxide: from toxin to therapy. Kinoshita H, Türkan H, Vucinic S et al (2020) Carbon monoxide poi-
Respir Care 62(10):1333–1342. https​://doi.org/10.4187/respc​ soning. Toxicol Rep 7:169–173. https​://doi.org/10.1016/j.toxre​
are.05781​ p.2020.01.005
Hoetzel A, Dolinay T, Vallbracht S et al (2008) Carbon monoxide pro- Knauert M, Vangala S, Haslip M, Lee PJ (2013) Therapeutic applica-
tects against ventilator-induced lung injury via PPAR-gamma and tions of carbon monoxide. Oxid Med Cell Longev 2013:360815.
inhibition of Egr-1. Am J Respir Crit Care Med 177(11):1223– https​://doi.org/10.1155/2013/36081​5
1232. https​://doi.org/10.1164/rccm.20070​8-1265O​C Kourti M, Westwell A, Jiang W, Cai J (2019) Repurposing old car-
Hsiao CL, Kuo HC, Huang CC (2004) Delayed encephalopathy after bon monoxide-releasing molecules towards the anti-angiogenic
carbon monoxide intoxication–long-term prognosis and corre- therapy of triple-negative breast cancer. Oncotarget 10(10):1132.
lation of clinical manifestations and neuroimages. Acta Neurol https​://doi.org/10.18632​/oncot​arget​.26638​
Taiwan 13(2):64–70. https:​ //doi.org/10.29819/​ ANT.200406​ .0004 Lee SJ, Ryter SW, Xu JF et al (2011) Carbon monoxide activates
Huang CC, Chung MH, Weng SF et al (2014) Long-term prognosis of autophagy via mitochondrial reactive oxygen species forma-
patients with carbon monoxide poisoning: a nationwide cohort tion. Am J Respir Cell Mol Biol 45(4):867–873. https​://doi.
study. PLoS ONE 9(8):e105503. https​://doi.org/10.1371/journ​ org/10.1165/rcmb.2010-0352O​C
al.pone.01055​03 Lee W-Y, Chen Y-C, Shih C-M et al (2014) The induction of heme
Huang CC, Ho CH, Chen YC et al (2019) Risk of myocardial infarc- oxygenase-1 suppresses heat shock protein 90 and the prolifera-
tion after carbon monoxide poisoning: a nationwide population- tion of human breast cancer cells through its byproduct carbon
based cohort study. Cardiovasc Toxicol 19(2):147–155. https​:// monoxide. Toxicol Appl Pharmacol 274(1):55–62. https​://doi.
doi.org/10.1007/s1201​2-018-9484-9 org/10.1016/j.taap.2013.10.027
Ilyas G, Zhao E, Liu K et  al (2016) Macrophage autophagy lim- Lee DW, Shin HY, Jeong JH, Han J, Ryu S, Nakahira K, Moon JS
its acute toxic liver injury in mice through down regula- (2017a) Carbon monoxide regulates glycolysis-dependent
tion of interleukin-1β. J Hepatol 64(1):118–127. https​://doi. NLRP3 inflammasome activation in macrophages. Biochem
org/10.1016/j.jhep.2015.08.019 Biophys Res Commun 493(2):957–963. https:​ //doi.org/10.1016/j.
Ismailova A, Kuter D, Bohle DS, Butler IS (2018) An overview of bbrc.2017.09.111
the potential therapeutic applications of CO-releasing mol- Lee PP, Lobato-Márquez D, Pramanik N et al (2017b) Wiskott-Aldrich
ecules. Bioinorg Chem Appl 2018:8547364. https ​ : //doi. syndrome protein regulates autophagy and inflammasome activ-
org/10.1155/2018/85473​64 ity in innate immune cells. Nat Commun 8(1):1576. https​://doi.
Jeyabal P, Thandavarayan RA, Joladarashi D et  al (2016) Micro- org/10.1038/s4146​7-017-01676​-0
RNA-9 inhibits hyperglycemia-induced pyroptosis in human Levy RJ (2016) Anesthesia-related carbon monoxide exposure: toxicity
ventricular cardiomyocytes by targeting ELAVL1. Biochem and potential therapy. Anesth Analg 123(3):670–681. https:​ //doi.
Biophys Res Commun 471(4):423–429. https:​ //doi.org/10.1016/j. org/10.1213/ANE.00000​00000​00146​1
bbrc.2016.02.065 Li Q, Bi MJ, Bi WK, Kang H, le Yan J, Guo YL (2016) Edaravone
Jiang L, Fei D, Gong R et al (2016) CORM-2 inhibits TXNIP/NLRP3 attenuates brain damage in rats after acute CO poisoning through
inflammasome pathway in LPS-induced acute lung injury. inhibiting apoptosis and oxidative stress. Environ Toxicol
Inflamm Res 65(11):905–915. https​://doi.org/10.1007/s0001​ 31(3):372–379. https​://doi.org/10.1002/tox.22052​
1-016-0973-7 Li Y, Dang J, Liang Q, Yin L (2019) Thermal-responsive carbon mon-
Jo EK, Kim JK, Shin DM, Sasakawa C (2016) Molecular mechanisms oxide (CO) delivery expedites metabolic exhaustion of cancer
regulating NLRP3 inflammasome activation. Cell Mol Immunol cells toward reversal of chemotherapy resistance. ACS Cent Sci
13(2):148–159. https​://doi.org/10.1038/cmi.2015.95 5(6):1044–1058. https​://doi.org/10.1021/acsce​ntsci​.9b002​16
Katzenberger RJ, Ganetzky B, Wassarman DA (2015) The gut reaction Lin JP, Lu X, Liao SZ et al (2019) Cross-regulation between exosomal
to traumatic brain injury. Fly (Austin) 9(2):68–74. https​://doi. and autophagic pathways: promising therapy targets in disease.
org/10.1080/19336​934.2015.10856​23 Discov Med 27(149):201–210
Kavakli HS, Erel O, Delice O, Gormez G, Isikoglu S, Tanriverdi F Lippi G, Rastelli G, Meschi T, Borghi L, Cervellin G (2012) Patho-
(2011) Oxidative stress increases in carbon monoxide poison- physiology, clinics, diagnosis and treatment of heart involvement
ing patients. Hum Exp Toxicol 30(2):160–164. https​://doi. in carbon monoxide poisoning. Clin Biochem 45(16–17):1278–
org/10.1177/09603​27110​38853​9 1285. https​://doi.org/10.1016/j.clinb​ioche​m.2012.06.004
Keyse SM, Tyrrell RM (1989) Heme oxygenase is the major 32-kDa Liu P, Huang G, Wei T et al (2018) Sirtuin 3-induced macrophage
stress protein-induced in human-skin fibroblasts by UVA radia- autophagy in regulating NLRP3 inflammasome activation. Bio-
tion, hydrogen-peroxide, and sodium arsenite. Proc Natl Acad Sci chim Biophys Acta Mol Basis Dis 1864(3):764–777. https​://doi.
USA 86(1):99–103. https​://doi.org/10.1073/pnas.86.1.99 org/10.1016/j.bbadi​s.2017.12.027
Kim HH, Choi S (2018) Therapeutic aspects of carbon monoxide in Liu W-H, Shi L-S, Chung M-C, Chang T-C, Lee S-Y (2019) Ant-
cardiovascular disease. Int J Mol Sci 19(8):2381. https​://doi. camphin M inhibits TLR4-mediated inflammatory responses
org/10.3390/ijms1​90823​81 by upregulating the Nrf2/HO-1 pathway and suppressing the
Kim HJ, Joe Y, Kong JS et al (2013) Carbon monoxide protects against NLRP3 inflammasome pathway in macrophages. Am J Chin Med
hepatic ischemia/reperfusion injury via ROS-dependent Akt sign- 47(7):1611–1626. https​://doi.org/10.1142/s0192​415x1​95008​24
aling and inhibition of glycogen synthase kinase 3β. Oxid Med Liu DL, Zeng X, Li X et al (2020) Advances in the molecular mecha-
Cell Longev 2013:306421. https​://doi.org/10.1155/2013/30642​1 nisms of NLRP3 inflammasome activators and inactivators.
Kim K, Chini N, Fairchild DG et al (2016) Evaluation of cardiac toxic- Biochem Pharmacol 175:113863. https​://doi.org/10.1016/j.
ity biomarkers in rats from different laboratories. Toxicol Pathol bcp.2020.11386​3
44(8):1072–1083. https​://doi.org/10.1177/01926​23316​66827​6 Lo CP, Chen SY, Lee KW et al (2007) Brain injury after acute car-
Kim JS, Ko BS, Sohn CH, Kim YJ, Kim WY (2020) High-sensitivity bon monoxide poisoning: early and late complications. AJR
troponin i and creatinine kinase-myocardial band in screening Am J Roentgenol 189(4):W205-211. https​://doi.org/10.2214/
for myocardial injury in patients with carbon monoxide poison- AJR.07.2425
ing. Diagnostics (Basel) 10(4):242. https:​ //doi.org/10.3390/diagn​ Lo Iacono L, Boczkowski J, Zini R et al (2011) A carbon monox-
ostic​s1004​0242 ide-releasing molecule (CORM-3) uncouples mitochondrial
respiration and modulates the production of reactive oxygen

13
Archives of Toxicology (2021) 95:1141–1159 1157

species. Free Radic Biol Med 50(11):1556–1564. https​://doi. J Physiol 276(4):L688–L694. https​: //doi.org/10.1152/ajplu​
org/10.1016/j.freer​adbio​med.2011.02.033 ng.1999.276.4.L688
Loboda A, Jozkowicz A, Dulak J (2015) HO-1/CO system in tumor Otterbein LE, Bach FH, Alam J et al (2000) Carbon monoxide has
growth, angiogenesis and metabolism—targeting HO-1 as an anti-inflammatory effects involving the mitogen-activated
anti-tumor therapy. Vascul Pharmacol 74:11–22. https​://doi. protein kinase pathway. Nat Med 6(4):422–428. https​://doi.
org/10.1016/j.vph.2015.09.004 org/10.1038/74680​
Loboda A, Damulewicz M, Pyza E, Jozkowicz A, Dulak J (2016) Role Ozcan N, Ozcam G, Kosar P, Ozcan A, Basar H, Kaymak C (2016)
of Nrf2/HO-1 system in development, oxidative stress response Correlation of computed tomography, magnetic resonance
and diseases: an evolutionarily conserved mechanism. Cell imaging and clinical outcome in acute carbon monoxide
Mol Life Sci 73(17):3221–3247. https​://doi.org/10.1007/s0001​ poisoning. Braz J Anesthesiol 66(5):529–532. https​: //doi.
8-016-2223-0 org/10.1016/j.bjane​.2014.05.006
Losacco C, Perillo A (2018) Particulate matter air pollution and res- Pae H-O, Kim E-C, Chung H-T (2008) Integrative survival response
piratory impact on humans and animals. Environ Sci Pollut evoked by heme oxygenase-1 and heme metabolites. J Clin
Res Int 25(34):33901–33910. https​://doi.org/10.1007/s1135​ Biochem Nutr 42(3):197. https​://doi.org/10.3164/jcbn.20080​29
6-018-3344-9 Papandreou ME, Tavernarakis N (2017) Autophagy and the endo/
Luo YP, Jiang L, Kang K et al (2014) Hemin inhibits NLRP3 inflam- exosomal pathways in health and disease. Biotechnol J
masome activation in sepsis-induced acute lung injury, involving 12(1):1600175. https​://doi.org/10.1002/biot.20160​0175
heme oxygenase-1. Int Immunopharmacol 20(1):24–32. https​:// Piantadosi CA, Zhang J, Levin ED, Folz RJ, Schmechel DE (1997)
doi.org/10.1016/j.intim​p.2014.02.017 Apoptosis and delayed neuronal damage after carbon monoxide
Lv J, Su W, Yu Q et al (2018) Heme oxygenase-1 protects airway poisoning in the rat. Exp Neurol 147(1):103–114. https​://doi.
epithelium against apoptosis by targeting the proinflammatory org/10.1006/exnr.1997.6584
NLRP3-RXR axis in asthma. J Biol Chem 293(48):18454– Pu Q, Lin P, Wang Z et al (2019) Interaction among inflammasome,
18465. https​://doi.org/10.1074/jbc.RA118​.00495​0 autophagy and non-coding RNAs: new horizons for drug. Pre-
Lv C, Su Q, Fang J, Yin H (2019) Styrene-maleic acid copolymer- cis Clin Med 2(3):166–182. https​://doi.org/10.1093/pcmed​i/
encapsulated carbon monoxide releasing molecule-2 (SMA/ pbz01​9
CORM-2) suppresses proliferation, migration and invasion Rao L, Eissa NT (2020) Autophagy in pulmonary innate immunity. J
of colorectal cancer cells in vitro and in vivo. Biochem Bio- Innate Immun 12(1):21–30. https​://doi.org/10.1159/00049​7414
phys Res Commun 520(2):320–326. https​://doi.org/10.1016/j. Rathinam VAK, Fitzgerald KA (2016) Inflammasome complexes:
bbrc.2019.09.112 emerging mechanisms and effector functions. Cell 165(4):792–
Maines MD, Trakshel GM, Kutty RK (1986) Characterization of two 800. https​://doi.org/10.1016/j.cell.2016.03.046
constitutive forms of rat liver microsomal heme oxygenase. Only Rocha M, Apostolova N, Diaz-Rua R, Muntane J, Victor VM (2020)
one molecular species of the enzyme is inducible. J Biol Chem Mitochondria and T2D: Role of autophagy, ER Stress, and
261(1):411–419 inflammasome. Trends Endocrinol Metab 31(10):725–741.
Mangan MSJ, Olhava EJ, Roush WR, Seidel HM, Glick GD, Latz https​://doi.org/10.1016/j.tem.2020.03.004
E (2018) Targeting the NLRP3 inflammasome in inflamma- Roderique JD, Josef CS, Feldman MJ, Spiess BD (2015) A modern
tory diseases. Nat Rev Drug Discov 17(8):588–606. https​://doi. literature review of carbon monoxide poisoning theories, thera-
org/10.1038/nrd.2018.97 pies, and potential targets for therapy advancement. Toxicology
Mehto S, Jena KK, Nath P et al (2019) The Crohn’s disease risk factor 334:45–58. https​://doi.org/10.1016/j.tox.2015.05.004
IRGM limits NLRP3 inflammasome activation by impeding its Romao CC, Blattler WA, Seixas JD, Bernardes GJ (2012) Develop-
assembly and by mediating its selective autophagy. Mol Cell ing drug molecules for therapy with carbon monoxide. Chem
73(3):429-445.e7. https​://doi.org/10.1016/j.molce​l.2018.11.018 Soc Rev 41(9):3571–3583. https​://doi.org/10.1039/c2cs1​5317c​
Mimura K, Harada M, Sumiyoshi S et al (1999) Long-term follow-up Rose JJ, Wang L, Xu Q et al (2017) Carbon monoxide poisoning:
study on sequelae of carbon monoxide poisoning; serial inves- pathogenesis, management, and future directions of therapy.
tigation 33 years after poisoning. Seishin Shinkeigaku Zasshi Am J Respir Crit Care Med 195(5):596–606. https​: //doi.
101:592–618 org/10.1164/rccm.20160​6-1275C​I
Nakahira K, Choi AMK (2015) Carbon monoxide in the treatment of Ryter SW (2019) Heme oxygenase-1/carbon monoxide as modula-
sepsis. Am J Physiol Lung C 309(12):L1387–L1393. https​://doi. tors of autophagy and inflammation. Arch Biochem Biophys
org/10.1152/ajplu​ng.00311​.2015 678:108186. https​://doi.org/10.1016/j.abb.2019.10818​6
Nakahira K, Haspel JA, Rathinam VA et al (2011) Autophagy proteins Ryter SW, Choi AMK (2016) Targeting heme oxygenase-1 and
regulate innate immune responses by inhibiting the release of carbon monoxide for therapeutic modulation of inflamma-
mitochondrial DNA mediated by the NALP3 inflammasome. Nat tion. Transl Res 167(1):7–34. https ​ : //doi.org/10.1016/j.
immunol 12(3):222. https​://doi.org/10.1038/ni.1980 trsl.2015.06.011
Nemeth Z, Csizmadia E, Vikstrom L et al (2016) Alterations of tumor Ryter SW, Tyrrell RM (2000) The heme synthesis and degradation
microenvironment by carbon monoxide impedes lung cancer pathways: role in oxidant sensitivity. Heme oxygenase has both
growth. Oncotarget 7(17):23919. https​://doi.org/10.18632​/oncot​ pro- and antioxidant properties. Free Radic Biol Med 28(2):289–
arget​.8081 309. https​://doi.org/10.1016/s0891​-5849(99)00223​-3
Ning K, Zhou YY, Zhang N, Sun XJ, Liu WW, Han CH (2020) Neuro- Ryter SW, Alam J, Choi AMK (2006) Heme oxygenase-1/carbon mon-
cognitive sequelae after carbon monoxide poisoning and hyper- oxide: From basic science to therapeutic applications. Physiol
baric oxygen therapy. Med Gas Res 10(1):30–36. https​://doi. Rev 86(2):583–650. https:​ //doi.org/10.1152/physre​ v.00011.​ 2005
org/10.4103/2045-9912.27998​1s Ryter SW, Ma KC, Choi AMK (2018) Carbon monoxide in lung cell
Oh S, Choi SC (2015) Acute carbon monoxide poisoning and physiology and disease. Am J Physiol Cell Physiol 314(2):C211–
delayed neurological sequelae: a potential neuroprotection C227. https​://doi.org/10.1152/ajpce​ll.00022​.2017
bundle therapy. Neural Regen Res 10(1):36–38. https​://doi. Schwartz L, Supuran CT, Alfarouk KO (2017) The Warburg effect
org/10.4103/1673-5374.15064​4 and the hallmarks of cancer. Anticancer Agents Med Chem
Otterbein LE, Mantell LL, Choi AM (1999) Carbon monox- 17(2):164–170. https:​ //doi.org/10.2174/187152​ 06166​ 66161​ 0311​
ide provides protection against hyperoxic lung injury. Am 43301​

13

1158 Archives of Toxicology (2021) 95:1141–1159

Shao B-Z, Xu Z-Q, Han B-Z, Su D-F, Liu C (2015) NLRP3 inflamma- Wang Y, Han ZH, Fan YQ et al (2017) MicroRNA-9 inhibits NLRP3
some and its inhibitors: a review. Front Pharmacol 6:262. https​ inflammasome activation in human atherosclerosis inflam-
://doi.org/10.3389/fphar​.2015.00262​ mation cell models through the JAK1/STAT signaling path-
Singh RB, Fedacko J, Pella D et al (2018a) The gut-brain-axis and the way. Cell Physiol Biochem 41(4):1555–1571. https ​ : //doi.
heart. MOJ Public Health 7(3):129–138. https:​ //doi.org/10.15406​ org/10.1159/00047​0822
/mojph​.2018.07.00218​ Wang Y, Yao C, Xu C et al (2019) Carbon monoxide and risk of
Singh N, Ahmad Z, Baid N, Kumar A (2018b) Host heme oxygenase-1: outpatient visits due to cause-specific diseases: a time-series
friend or foe in tackling pathogens? IUBMB Life 70(9):869–880. study in Yichang, China. Environ Health 18(1):36. https​://doi.
https​://doi.org/10.1002/iub.1868 org/10.1186/s1294​0-019-0477-3
Slaughter JC, Kim E, Sheppard L, Sullivan JH, Larson TV, Claiborn Watson A, Williams R (1984) Anoxic hepatic and intestinal injury
C (2005) Association between particulate matter and emergency from carbon monoxide poisoning. Br Med J (Clin Res Ed)
room visits, hospital admissions and mortality in Spokane, Wash- 289(6452):1113. https​://doi.org/10.1136/bmj.289.6452.1113
ington. J Expo Anal Environ Epidemiol 15(2):153–159. https​:// Waza AA, Hamid Z, Ali S, Bhat SA, Bhat MA (2018) A review
doi.org/10.1038/sj.jea.75003​82 on heme oxygenase-1 induction: is it a necessary evil.
Son Y, Lee JH, Chung H-T, Pae H-O (2013) Therapeutic roles of heme Inflamm Res 67(7):579–588. https​: //doi.org/10.1007/s0001​
oxygenase-1 in metabolic diseases: curcumin and resveratrol ana- 1-018-1151-x
logues as possible inducers of heme oxygenase-1. Oxid Med Cell Weaver LK (2009) Clinical practice. Carbon monoxide poisoning.
Longev 2013:639541. https​://doi.org/10.1155/2013/63954​1 N Engl J Med 360(12):1217–1225. https​: //doi.org/10.1056/
Soni HM, Jain MR, Mehta AA (2012) Mechanism(s) involved in NEJMc​p0808​891
carbon monoxide-releasing molecule-2-mediated cardioprotec- Weaver LK, Deru K (2016) Ischemic colitis associated with acute
tion during ischaemia-reperfusion injury in isolated rat heart. carbon monoxide poisoning–a case report. Undersea Hyperb
Indian J Pharm Sci 74(4):281–291. https:​ //doi.org/10.4103/0250- Med 43(2):175–180
474X.10704​7 Weaver L, Hopkins R, Churchill S, Deru K (2008) Neurological
Sun Q, Gao W, Loughran P et al (2013) Caspase 1 activation is protec- outcomes 6 years after acute carbon monoxide poisoning. In:
tive against hepatocyte cell death by up-regulating beclin 1 pro- Abstract of the undersea & hyperbaric medical society 2008
tein and mitochondrial autophagy in the setting of redox stress. annual scientific meeting
J Biol Chem 288(22):15947–15958. https​://doi.org/10.1074/jbc. Wegiel B, Gallo D, Csizmadia E et al (2013) Carbon monoxide expe-
M112.42679​1 dites metabolic exhaustion to inhibit tumor growth. Cancer
Swanson KV, Deng M, Ting JP-Y (2019) The NLRP3 inflamma- Res 73(23):7009–7021. https​: //doi.org/10.1158/0008-5472.
some: molecular activation and regulation to therapeutics. Nat CAN-13-1075
Rev Immunol 19(8):477–489. https​://doi.org/10.1038/s4157​ Wegiel B, Larsen R, Gallo D et al (2014) Macrophages sense and
7-019-0165-0 kill bacteria through carbon monoxide-dependent inflamma-
Takahashi M, Maemura K, Sawada Y et al (1982) Hyperamylasemia some activation. J Clin Invest 124(11):4926–4940. https​://doi.
in acute carbon monoxide poisoning. J Trauma 22(4):311–314. org/10.1172/JCI72​853
https​://doi.org/10.1097/00005​373-19820​4000-00008​ Wong CS, Lin YC, Sung LC et al (2017) Increased long-term risk
Tekin YK, Korkmaz I, Demirtas E, Gunday B, Tekin G (2019) Car- of major adverse cardiovascular events in patients with car-
bon monoxide poisoning-induced upper gastrointestinal bleed- bon monoxide poisoning: a population-based study in Taiwan.
ing. Notfall Rettungsmed 23:132–133. https​://doi.org/10.1007/ PLoS ONE 12(4):e0176465. https​: //doi.org/10.1371/journ​
s1004​9-019-00640​-3 al.pone.01764​65
Tenhunen R, Marver HS, Schmid R (1968) The enzymatic conver- Wu XB, Sun HY, Luo ZL, Cheng L, Duan XM (1866) Ren JD (2020)
sion of heme to bilirubin by microsomal heme oxygenase. Proc Plasma-derived exosomes contribute to pancreatitis-associated
Natl Acad Sci USA 61(2):748–755. https​://doi.org/10.1073/ lung injury by triggering NLRP3-dependent pyroptosis in
pnas.61.2.748 alveolar macrophages. Biochim Biophys Acta Mol Basis Dis
Tenhunen R, Ross ME, Marver HS, Schmid R (1970) Reduced nico- 5:165685. https​://doi.org/10.1016/j.bbadi​s.2020.16568​5
tinamide-adenine dinucleotide phosphate dependent biliverdin Wu CT, Huang JL, Hsia SH (2009) Acute carbon monoxide poisoning
reductase: partial purification and characterization. Biochemistry with severe cardiopulmonary compromise: a case report. Cases
9(2):298–303. https​://doi.org/10.1021/bi008​04a01​6 J 2(1):52. https​://doi.org/10.1186/1757-1626-2-52
Thom SR (1985) (1990) Carbon monoxide-mediated brain lipid per- Wu B, Wu Y, Tang W (2019) Heme catabolic pathway in inflamma-
oxidation in the rat. J Appl Physiol 68(3):997–1003. https​://doi. tion and immune disorders. Front Pharmacol 10:825. https​://doi.
org/10.1152/jappl​.1990.68.3.997 org/10.3389/fphar​.2019.00825​
Tofighi R, Tillmark N, Dare E, Aberg AM, Larsson JE, Ceccatelli S Xu J, Camfield R, Gorski SM (2018) The interplay between exosomes
(2006) Hypoxia-independent apoptosis in neural cells exposed and autophagy-partners in crime. J Cell Sci 131(15):jcs215210.
to carbon monoxide in vitro. Brain Res 1098(1):1–8. https​://doi. https​://doi.org/10.1242/jcs.21521​0
org/10.1016/j.brain​res.2006.04.095 Xu SL, Lin Y, Zhu XZ et al (2020) Autophagy promotes phagocytosis
Villarroya-Beltri C, Baixauli F, Mittelbrunn M et al (2016) ISGyla- and clearance of Treponema pallidum via the NLRP3 inflamma-
tion controls exosome secretion by promoting lysosomal degra- some in macrophages. J Eur Acad Dermatol Venereol. https​://
dation of MVB proteins. Nat commun 7(1):13588. https​://doi. doi.org/10.1111/jdv.16463​
org/10.1038/ncomm​s1358​8 Xue Z, Zhang Z, Liu H et al (2019) lincRNA-Cox2 regulates NLRP3
Von Burg R (1999) Carbon monoxide. J Appl Toxicol 19(5):379–386. inflammasome and autophagy mediated neuroinflammation.
https:​ //doi.org/10.1002/(sici)1099-1263(199909​ /10)19:5%3c379​ Cell Death Differ 26(1):130–145. https​://doi.org/10.1038/s4141​
::aid-jat56​3%3e3.0.co;2-8 8-018-0105-8
Wang P, Huang J, Li Y et al (2015) Exogenous carbon monoxide Yan Y, Du C, Li G et al (2018) CO suppresses prostate cancer cell
decreases sepsis-induced acute kidney injury and inhibits NLRP3 growth by directly targeting LKB1/AMPK/mTOR pathway
inflammasome activation in rats. Int J Mol Sci 16(9):20595– in vitro and in vivo. Urol Oncol 36(6):312.e1-312.e8. https​://
20608. https​://doi.org/10.3390/ijms1​60920​595 doi.org/10.1016/j.urolo​nc.2018.02.013

13
Archives of Toxicology (2021) 95:1141–1159 1159

Yan Y, Wang L, Chen S et al (2020) Carbon monoxide inhibits T cell Zhang X, Shan P, Alam J, Fu XY, Lee PJ (2005) Carbon monoxide
proliferation by suppressing reactive oxygen species signaling. differentially modulates STAT1 and STAT3 and inhibits apop-
Antioxid Redox Signal 32(7):429–446. https​://doi.org/10.1089/ tosis via a phosphatidylinositol 3-kinase/Akt and p38 kinase-
ars.2019.7814 dependent STAT3 pathway during anoxia-reoxygenation injury.
Ye F, Li X, Liu Y et al (2018) Hemin provides protection against J Biol Chem 280(10):8714–8721. https​://doi.org/10.1074/jbc.
lead neurotoxicity through heme oxygenase 1/carbon monox- M4080​92200​
ide activation. J Appl Toxicol 38(10):1353–1364. https​://doi. Zhang Y, Liu F, Yuan Y et al (2017) Inflammasome-derived exosomes
org/10.1002/jat.3646 activate NF-κB signaling in macrophages. J Proteome Res
Yin ZY, Pascual C, Klionsky DJ (2016) Autophagy: machinery and 16(1):170–178. https​://doi.org/10.1021/acs.jprot​eome.6b005​99
regulation. Microb Cell 3(12):457–465. https:​ //doi.org/10.15698​ Zheng XY, Ding H, Jiang LN et al (2015) Association between air
/mic20​16.12.546 pollutants and asthma emergency room visits and hospital admis-
Yogaratnam J, Hariram J, Lee DS et al (2011) Delayed neuropsychiatric sions in time series studies: a systematic review and meta-anal-
sequelae and recovery following carbon monoxide poisoning. ysis. PLoS ONE 10(9):e0138146. https​://doi.org/10.1371/journ​
Ann Acad Med Singapore 40(11):516–517 al.pone.01381​46
Zeglinski MR, Turner CT, Zeng R et al (2019) Soluble wood smoke
extract promotes barrier dysfunction in alveolar epithelial cells Publisher’s Note Springer Nature remains neutral with regard to
through a MAPK signaling pathway. Sci Rep 9(1):10027. https​ jurisdictional claims in published maps and institutional affiliations.
://doi.org/10.1038/s4159​8-019-46400​-8

13

You might also like