You are on page 1of 11

Respiratory Physiology & Neurobiology 132 (2002) 81 – 91

www.elsevier.com/locate/resphysiol

O2 sensing in hypoxic pulmonary vasoconstriction: the


mitochondrial door re-opens
Gregory B. Waypa, Paul T. Schumacker *
Department of Medicine MC6026, The Uni6ersity of Chicago, 5841 South Maryland A6enue, Chicago, IL 60637, USA
Accepted 9 April 2002

Abstract

The identity of the O2 sensor underlying the hypoxic pulmonary vasoconstriction (HPV) response has been sought
for more than 50 years. Recently, the mitochondria have again come into sharp focus as the cellular organelle
responsible for triggering the events that culminate in pulmonary artery constriction. Studies from different
laboratories propose two disparate models to explain how mitochondria react to a decrease in PO2. One model
proposes that hypoxia slows or inhibits mitochondrial electron transport resulting in the accumulation of reducing
equivalents and a decrease in the generation of reactive oxygen species (ROS). This is proposed to activate a
redox-sensitive pathway leading to pulmonary vasoconstriction. A second and opposing model suggests that hypoxia
triggers a paradoxical increase in mitochondrial ROS generation. This increase would then lead to the activation of
an oxidant-sensitive signaling transduction pathway leading to HPV. This article summarizes the potential involve-
ment of mitochondria in these two very different models. © 2002 Elsevier Science B.V. All rights reserved.

Keywords: Blood, flow, pulmonary; Hypoxia, pulmonary vasoconstriction; Mitochondria, reactive oxygen species; Oxygen, reactive
species, HPV; Perfusion, pulmonary

1. Introduction in health and disease states, and toward unravel-


ing the underlying cellular mechanism by which
Hypoxic pulmonary vasoconstriction (HPV) di- the cells in the pulmonary artery activate the
verts blood flow away from the lung during in response. A paramount and still unresolved ques-
utero development and it helps to optimize lung tion relates to the underlying mechanism by which
gas exchange after birth by enhancing the match- cells detect low PO2 conditions and convert this
ing of blood flow and ventilation. Much effort has into a biological signal. It is rightly believed that
been directed toward characterizing this response an understanding of this mechanism will enhance
the base of knowledge related to the physiology
and pathophysiology of the pulmonary circula-
tion. An understanding of the mechanism of tis-
* Corresponding author. Tel.: +1-773-702-6790; fax: + 1-
773-702-4736 sue oxygen sensing in the lung could also help to
E-mail address: pschumac@medicine.bsd.uchicago.edu (P.T. lay the foundation for our understanding of oxy-
Schumacker). gen sensing in other tissues. Arguably, progress

1569-9048/02/$ - see front matter © 2002 Elsevier Science B.V. All rights reserved.
PII: S1569-9048(02)00051-4
82 G.B. Waypa, P.T. Schumacker / Respiratory Physiology & Neurobiology 132 (2002) 81–91

has been made toward identifying the O2 sensor in rent views on mitochondrial O2 sensing and to
HPV. However, a diverse collection of candidate reconcile these points.
O2 sensors are still being evaluated by different
laboratories, and definitive studies in support of
any single model have not yet appeared. Not 2. The HPV response
surprisingly, no consensus of opinion has emerged
behind any single mechanism. Several excellent HPV was first described in 1946 by von Euler
review articles dealing with HPV have appeared and Liljestrand, in a paper characterizing the
recently, and the reader is directed to those for a effects of interventions on pulmonary arterial
more comprehensive analysis of the field (Mar- pressure in the cat (von Euler and Liljestrand,
shall et al., 1994; Michelakis et al., 1995; Ward 1946). They found that pulmonary arterial pres-
and Robertson, 1995; Weir and Archer, 1995; sure increased when the lungs were ventilated with
Michelakis et al., 1997; Ward and Aaronson, low inspired O2 mixtures and decreased when
1999; Archer et al., 2000; Jones et al., 2000; Jones ventilated with pure O2. They recognized that
and Morice, 2000; Sweeney and Yuan, 2000; these responses reflected changes in pulmonary
Coppock et al., 2001; Michelakis and Weir, 2001; vascular resistance, and they speculated that a
Sylvester, 2001; Weissmann et al., 2001). vasoconstrictor response to hypoxia could help to
The present report is intended to provide a improve gas exchange efficiency by diverting
more narrow analysis of information and ideas blood flow away from poorly ventilated lung re-
pointing to mitochondria as the O2 sensor during gions and toward areas with better oxygenation.
HPV. The notion that mitochondria might func- Much later, this prediction was shown to be true,
tion as O2 sensors is an old one. From a teleolog- especially in damaged or diseased lungs where
ical standpoint it is reasonable that an organelle hypoxic and normoxic regions co-exist. We now
where most of the O2 is consumed would be the know that the HPV response is retained in excised
site of O2 sensing. However, smooth muscle cells lungs (Rounds and McMurtry, 1981; McMurtry,
do not contain large numbers of mitochondria. 1984; Archer et al., 1993; Grimminger et al., 1995;
Also, certain mitochondrial inhibitors failed to Weissmann et al., 1995, 1998) and hypoxia-in-
block the HPV response in early experiments. duced contraction can even be seen in rings of
This led investigators to wonder how that or- pulmonary arteries subjected to low O2 conditions
ganelle could be the site of O2 sensing if the in an organ bath (Marshall et al., 1996; Zhao et
downstream responses were preserved in experi- al., 1996). Constriction is preserved if the vessels
ments where mitochondrial function was crippled. are denuded of endothelium (Gelband and Gel-
Nevertheless, more recent studies have provided a band, 1997; Archer et al., 1998), although the
clearer explanation of how mitochondria might be response is quite markedly attenuated. This has
involved. In addition, they provide an explanation led to the conclusion that the endothelium am-
of why certain mitochondrial inhibitors block plifies, but does not initiate HPV. Given that the
HPV while others do not. Two different models of pulmonary endothelium can amplify the HPV re-
mitochondrial O2 sensing recently have come to sponse, particularly during conditions of sus-
the forefront. One model proposes that hypoxia tained hypoxia (Ward and Robertson, 1995;
slows or inhibits mitochondrial electron transport Robertson et al., 2001), it is probably more ap-
resulting in the accumulation of reducing equiva- propriate to consider the intact pulmonary artery
lents and a decrease in the generation of reactive when studying HPV than to focus narrowly on
oxygen species (ROS). A second and contrasting the pulmonary arterial myocyte. Nevertheless, iso-
model suggests that hypoxia triggers a paradoxi- lated pulmonary arterial (PA) myocytes do con-
cal increase in mitochondrial ROS generation. It tract under hypoxia (Zhang et al., 1997),
therefore appears that the mitochondrial door is indicating that PA smooth muscle cells possess
re-opening in the field of O2 sensing in HPV. The the ability to detect hypoxia and to activate a
present article will attempt to summarize the cur- contractile response. If both smooth muscle cells
G.B. Waypa, P.T. Schumacker / Respiratory Physiology & Neurobiology 132 (2002) 81–91 83

and endothelium contribute to the HPV response, trast, phase 2 occurs within minutes after hypoxic
it is interesting to ask whether the same O2 sensor challenge and is associated with chronic hypoxia.
is functioning in both cell types, or whether the Phase 2 requires an intact endothelium which
endothelium is merely permissive for the normal suggests that an endothelial-derived promoting
vasoconstriction response to hypoxia. factor may be involved (Liu et al., 2001). En-
It has now been well established that the HPV dothelin-1 (ET-1) is a potent vasoconstrictor that
response in pulmonary arteries is biphasic (Dipp may function as the amplifier of hypoxia-induced
et al., 2001; Leach et al., 2001). An initial tran- vasoconstriction. In this regard, the expression
sient constriction response (phase 1) is followed and release of ET-1 by endothelial cells has been
by a slow sustained constriction response (phase shown to increase during hypoxia (Bodi et al.,
2) (Fig. 1). Phase 1 has been shown to occur 1995; Hu et al., 1998; Yamashita et al., 2001).
independently of an intact endothelium (Zhang et Furthermore, some studies suggest that phase 2
al., 1997) and is thought to be initiated by the can be attenuated with BQ-123, an antagonist of
release of calcium from intracellular stores in PA endothelin-1 receptor subtype A (ETA-receptor)
myocytes, resulting in the closure of voltage-de- (Liu et al., 2001; Sato et al., 2001). However,
pendent potassium channels (KV), membrane de- other studies suggest that phase 2 is not affected
polarization, and the opening of voltage-gated by endothelin-1 receptor blockade with bosentan,
L-type calcium channels (Gelband and Gelband, a antagonist of both the ETA- and ETB-receptor
1997; Archer et al., 1998; Morio and McMurtry, subtypes, and that phase 2 contraction of small
2002). The influx of Ca2 + through the voltage- pulmonary arteries in rats does not involve en-
gated L-type Ca2 + channels initiates the calmod- dothelin-1 (Lazor et al., 1996). In either case,
ulin-mediated activation of myosin light chain phase 1 and phase 2 are mechanistically distinct
kinase, resulting in actin– myosin interaction and contributors to the HPV response. It is conceiv-
PA myocyte contraction. Phase 1 is also referred able that a similar O2 sensor mechanism underlies
to as the acute HPV response because it begins the different responses to hypoxia in PA myocytes
within seconds after hypoxic challenge. By con- and PA endothelium, but this has not been
demonstrated.

3. Historical basis for implicating mitochondria as


the O2 sensor

The idea that mitochondria may function as the


O2 sensor during HPV is an old one. Early studies
focused on the premise that O2, acting as a sub-
strate for biochemical reactions, could impart a
rate-limiting effect during hypoxia that somehow
resulted in the contraction of PA myocytes. This
was appealing from a teleological standpoint since
the mitochondria are the primary consumers of
O2 in the cell. In accordance with this reasoning,
it was suggested that the inhibition of mitochon-
drial electron transport and oxidative phosphory-
Fig. 1. An overview of hypoxic pulmonary vasoconstriction lation could mimic the HPV response. Indeed, in
integrating phase 1 and phase 2 of the response. The trace 1981 Rounds and McMurtry reported that certain
shows a typical example of the biphasic response of the
pulmonary vasculature to hypoxia. This figure is based on
inhibitors of electron transport and oxidative
previously published work (Ward and Aaronson, 1999), with phosphorylation, including azide, cyanide, dini-
permission. trophenol, antimycin A, and rotenone, all mim-
84 G.B. Waypa, P.T. Schumacker / Respiratory Physiology & Neurobiology 132 (2002) 81–91

icked the hypoxic pressor response in isolated, et al., 1993). They suggested that the decrease in
blood-perfused lungs (Rounds and McMurtry, available O2 during hypoxia resulted in the inhibi-
1981). They suggested that hypoxia-induced de- tion of mitochondrial electron transport, an accu-
creases in mitochondrial electron transport could mulation of reducing equivalents such as
compromise the energy state of the cell, thereby NAD(P)H, and a decrease in ROS production.
initiating HPV. However, later studies in isolated According to this model, inhibitors of mitochon-
lungs by Buescher et al. (1991) and in isolated drial electron transport ought to mimic HPV by
pulmonary arteries by Leach et al. (2000) showed augmenting NAD(P)H levels and decreasing
that the energy state of the cell does not decrease ROS. In support of this idea, Reeve et al. re-
during hypoxia in either of these systems ported that the mitochondrial complex I inhibitor
(Buescher et al., 1991; Leach et al., 2000). There- rotenone decreased chemilumenescence and in-
fore, one may conclude that HPV is not triggered creased vascular tone in isolated perfused lungs
by a decrease in the energy state as a consequence (Reeve et al., 2001). Their data is consistent with
of hypoxia-induced inhibition of mitochondrial a model in which complex I (NADH:ubiquinone
electron transport. Classically, mitochondrial res- oxidoreductase) would function as an O2 sensor
piration has been found to remain unchanged by maintaining a basal level of superoxide and
until intracellular PO2 falls below a critical H2O2 production during normoxia. During hy-
threshold of  5 mmHg (Jones and Kennedy, poxia, the ROS produced by this system would
1982, 1986). It therefore seems unlikely that en- decrease and NADH concentrations would rise.
ergy supply in PA myocytes would become O2- These changes would shift the state of cytosolic
limited at oxygen tensions in the range of 10– 40 redox toward a more reduced level (Mohazzab et
mmHg where HPV is activated. Some investiga- al., 1995; Zhu et al., 1999). The change in redox
tors have underscored this by point by noting that state would then result in the inhibition of K+
mitochondria cannot serve as the an O2 sensor current through redox-sensitive 4-aminopyradine
because the Km of cytochrome oxidase is too low (4-AP)-sensitive KV channels (Reeve et al., 1995).
to permit detection of hypoxia in the physiologi- Inhibition of currents through KV-channels would
cal range (Bunn and Poyton, 1996). Such reason- then trigger membrane depolarization, opening of
ing appeared to shut the door on the L-type, voltage gated Ca2 + channels and subse-
mitochondria as a putative oxygen sensor in the quent myocyte contraction (Archer et al., 1998,
response to hypoxia 2000). However, it has been reported that antago-
nism of KV channels did not prevent HPV, sug-
gesting that this scheme may not be correct
4. Does hypoxia decrease mitochondrial ROS (Hasunuma et al., 1991; Sham et al., 2000). More-
generation? over, the importance of the change in ROS rela-
tive to the change in cytosolic NAD/NADH ratio
Renewed interest in the mitochondria as an O2 is not clear in this model.
sensor has been stimulated by speculation that
hypoxia could act by slowing down or inhibiting
the rate of ROS production. In 1993, Archer et al. 5. Does hypoxia increase mitochondrial ROS
reported that hypoxia increased PA pressure and generation?
simultaneously caused a decrease in whole lung
chemiluminescence. Their study therefore sug- In opposition to the theory that hypoxia results
gested that HPV and hypoxia could be linked by in a shift towards a reduced state, other labs have
a decrease in ROS production. The same labora- suggested that a paradoxical increase in ROS
tory reported that antimycin A and rotenone, generation occurs during hypoxia. For example,
agents that inhibit mitochondrial electron trans- Marshall et al. (1996) found an increase
port, caused a decrease in measured chemilume- chemilumenescence during hypoxia in isolated PA
nescence and an increase in PA pressure (Archer myocytes (Marshall et al., 1996). Similar increases
G.B. Waypa, P.T. Schumacker / Respiratory Physiology & Neurobiology 132 (2002) 81–91 85

in oxidant signaling have also been reported by Marshall et al. (1996) suggested that hypoxia
other laboratories in a variety of cell types (Chan- accelerates ROS generation by a membrane-
del et al., 1998; Duranteau et al., 1998; Chandel et bound NADPH oxidase, based on their observa-
al., 2000; Killilea et al., 2000; Liu et al., 2001; tion that diphenyleneiodonium (DPI) inhibited
Waypa et al., 2001). In cultured PA myocytes, we the oxidant signal and the contractile response to
observed an increase in oxidant signaling during hypoxia (Marshall et al., 1996). Grimminger et al.
hypoxia using the intracellular probe 2%,7%-dichlor- (1995) also found that DPI attenuated HPV
ofluorescin-diacetate (DCFH-DA). This probe en- (Grimminger et al., 1995). DPI inhibits a wide
ters cells and can be oxidized to the fluorescent range of flavoproteins including NADPH oxidase,
compound 2%,7%-dichlorofluorescein (DCF) in the mitochondrial complex I, glutathione reductase,
presence of ROS including H2O2, hydroxyl radi- nitric oxide synthase, and prostaglandin syn-
cals and nitric oxide, but not by superoxide di- thetase (Holland et al., 1973; Majander et al.,
rectly. It should therefore be regarded as a 1994). Therefore, the site of inhibition by DPI
sensitive but nonspecific indicator of oxidant responsible for the attenuation of HPV is not
stress. known. To clarify the possible involvement of an
Although some confusion exists with respect to NAD(P)H-oxidase in HPV, two separate groups
the cellular mechanisms of DCFH oxidation dur- used apocynin, an inhibitor of the neutrophil
ing hypoxia, other studies provide independent form of this enzyme (Stolk et al., 1994; Grim-
support for the conclusion that hypoxia-induced minger et al., 1995; Waypa et al., 2001). Apocynin
increases in ROS are involved in HPV. For exam- failed to attenuate either HPV or PA myocyte
ple, pharmacological antioxidants block the hy- contraction during hypoxia. The possible involve-
poxic response in isolated, buffer-perfused lungs ment of NADPH-oxidase was further explored by
(Waypa et al., 2001), suggesting that increases in Archer et al. (1999), who studied HPV in ho-
ROS generation are required for HPV. In addi- mozygous knockout animals lacking the
tion, when H2O2 formation in the cytosol was gp91phox subunit of the NADPH oxidase com-
blocked during hypoxia by inhibiting Cu,Zn su- plex. They found that the response to hypoxia
peroxide dismutase (SOD), the HPV response was was preserved, suggesting a lack of involvement of
abrogated indicating that H2O2 is an important that system in HPV (Archer et al., 1999). How-
downstream signaling agent (Waypa et al., 2001). ever, because of the specificity of this knockout,
Other studies also implicate increased H2O2 gener- these results do not rule out that possibility that
ation in the HPV response. For example, Monaco other NAD(P)H-like oxidase systems could still
and Burke-Wolin (1995) showed that HPV was be involved in HPV.
augmented when catalase was inhibited with
aminotriazole. Catalase normally degrades H2O2,
so this supports the notion that hypoxia-induced 6. Mitochondrial as the source of ROS
ROS generation mediates HPV. Weissmann et al. generation during hypoxia
(1998) observed that SOD and 4,5-dihydroxy-1,3-
benzenedisulfonic acid (to accelerate H2O2 genera- Mitochondria have long been known to gener-
tion from superoxide) did not affect HPV, ate ROS (Parsons et al., 1966) although these
confirming that H2O2, rather than superoxide, is oxidants have classically been viewed as toxic
involved (Weissmann et al., 1998). Furthermore, byproducts of the electron transport pathway,
nitrobluetetrazolium, which traps superoxide and possibly contributing to the effects of aging (So-
prevents H2O2 formation, attenuated HPV. Other hal and Weindruch, 1996). More recently, mito-
studies show that H2O2 constricts the pulmonary chondrial ROS have been implicated as
circulation during normoxia (Waypa et al., 2000). intracellular signaling agents. Our laboratory pre-
Collectively, these findings are consistent with a viously reported that mitochondria increase ROS
role for increased H2O2 as a signaling molecule generation during hypoxia and that site-specific
involved in HPV. inhibition of electron transport could attenuate
86 G.B. Waypa, P.T. Schumacker / Respiratory Physiology & Neurobiology 132 (2002) 81–91

Fig. 2. Schematic diagram showing mitochondrial ROS production at complex III. Solid arrows show electron transfer steps; solid
bars show sites of electron transport inhibition. Normal mitochondrial electron transport involves the movement of reducing
equivalents generated in the Krebs cycle through complex I or II, then through complex III and IV (cytochrome oxidase). The Q
cycle converts the dual electron transfer in complex I and II into single electron transfer steps in complex IV. Ubisemiquinone (a
free radical) created in this process can generate superoxide. This process appears to be amplified during hypoxia.

hypoxia-induced ROS generation (Chandel et al., menting ROS production. This conclusion is sup-
1998; Duranteau et al., 1998). These observations ported by the observation that the
suggest that a similar mechanism could be occur- vasoconstriction in response to cyanide could be
ring in the pulmonary arteries during HPV. Dur- inhibited by ebselen (an antioxidant) or myxothia-
ing normoxia, mitochondrial electron transport zol (a more proximal ETC inhibitor).
inhibitors such as cyanide or antimycin A, which Recently, Leach et al. (2001) reported an ele-
inhibit sites distal to complex III, tend to augment gant series of studies in isolated pulmonary arter-
ROS generation (Fig. 2). The likely explanation ies that are consistent with this model. They
for these findings is that ROS production occurs found that rotenone and myxothiazol could atten-
primarily within complex III. In the Q cycle, a uate hypoxia-induced increases in intracellular
free radical (ubisemiquinone) is normally gener- calcium ([Ca2 + ]i) activation and subsequent vaso-
ated during the electron transport process. This constriction. By contrast, inhibition of complex
radical can potentially donate its unpaired elec- IV with cyanide augmented the hypoxic response
tron to O2, thereby generating superoxide. During but had no effect on [Ca2 + ]. Interestingly, in PA
hypoxia, our model suggests that the process of vessels that had been treated with rotenone, they
ROS generation from that site is amplified. By were able to restore HPV by using succinate to
contrast, when mitochondrial electron transport is shuttle electrons into complex III via complex II,
inhibited at a more proximal site by inhibitors effectively bypassing the rotenone inhibition of
such as rotenone, DPI, or myxothiazol, the Complex I (Leach et al., 2001) (Fig. 2). This was
ubiquinone pool becomes fully oxidized and ROS consistent with earlier data from Rounds and
generation at complex III is abrogated. According McMurtry, who had found that antimycin A elic-
to this model, the ROS production at Complex III its vasoconstriction in normoxic lungs (Rounds
increases during hypoxia, and these oxidants trig- and McMurtry, 1981). Also, Archer et al. (1993)
ger signal pathways culminating in Ca2 + activa- found that cyanide induced vasoconstriction dur-
tion and contraction during HPV. Distal ing normoxia and augmented HPV without af-
inhibitors of the ETC such as cyanide induce fecting the response to angiotensin II or KCl
constriction during normoxia, presumably by aug- (Archer et al., 1993). Collectively, these findings
G.B. Waypa, P.T. Schumacker / Respiratory Physiology & Neurobiology 132 (2002) 81–91 87

are consistent with a role for increased mitochon- myocytes independently of the endothelium. This
drial ROS in HPV. A caveat is that these studies indicates that the O2 sensor responsible for trig-
rely on pharmacological inhibitors, which may gering phase 1 must be intrinsic to the PA my-
have unexpected effects. However, r0 PA my- ocyte. The HPV response may also involve one or
ocytes, which lack a functional mitochondrial more 4-AP-sensitive KV channels. Inhibition of
electron transport chain, selectively lost the re- Kv channels would promote membrane depolar-
sponse to hypoxia while they retained the ability ization, the opening of voltage-gated L-type Ca2 +
to contract in response to exogenous H2O2 or channels, and subsequent vasoconstriction
U46619 in our study (Waypa et al., 2001). These (Archer et al., 1993, 1998, 2000; Urena et al.,
observations are consistent with the proposed 1996; Gelband and Gelband, 1997; Zhang et al.,
model, but the mechanism by which hypoxia am- 1997; Barman, 1998; Weir et al., 1998). However,
plifies ROS generation at complex III is not yet other studies bring into question the requirements
known. of the KV channel for the HPV response. As
An alternative explanation for the increase in mentioned previously, HPV was shown to remain
ROS signaling during hypoxia involves the regula- intact after inhibition of KV channels with 4-AP
tion of superoxide egress from mitochondria to (Hasunuma et al., 1991; Sham et al., 2000). Fur-
cytosol. Superoxide generated inside the mito- thermore, HPV was significantly blunted, but not
chondria would presumably require an anion completely abolished, in SWAP mice which lack
channel to escape to the cytosol, because charged the KV1.5 channel (Archer et al., 2001). This
species cannot easily diffuse through the mem- suggests that KV channels may function to aug-
brane lipid bilayer. The mitochondrial inner mem-
ment the HPV response, rather than to trigger it
brane anion channel (IMAC) is a nonspecific
directly.
anion channel that could conceivably function as
It is possible that hypoxia causes the activation
such a pathway. If the IMAC were O2-sensitive
of Ca2 + as an early step in phase I. The release of
such that it increased its conductance during hy-
Ca2 + from intracellular stores seems to be re-
poxia, the egress of superoxide from the matrix
quired for HPV (Gelband and Gelband, 1997;
could increase even if the rate of mitochondrial
Morio and McMurtry, 2002) and it may be the
superoxide generation were to decrease during
hypoxia. This might explain why ROS signaling in source of that initial signal. Post et al. (1995)
the cytosol is increased during moderate hypoxia, reported that release of Ca2 + from internal stores
and why 4,4%-diisothiocyanatostilbene-2.2%-disul- causes inhibition of 4-AP-sensitive KV channels
ponic acid (DIDS), an anion channel inhibitor and subsequent membrane depolarization (Post et
which inhibits mitochondrial IMAC (Beavis and al., 1995). These results are consistent with a
Davatol-Hag, 1996; Terada, 1996), attenuated model of HPV in which hypoxia triggers the
HPV and abolished PA myocyte contraction dur- release of Ca2 + from internal stores, resulting in
ing hypoxia (Waypa et al., 2001). It should be KV channel inhibition, membrane depolarization,
noted that DIDS is also a thiol-reactive com- and the opening of voltage-gated L-type Ca2 +
pound; however, pretreatment with DIDS had no channels.
effect on H2O2-induced vasoconstriction in the Possible sources of intracellular Ca2 + during
lung. In either case, it appears that superoxide hypoxia included inositol 1,4,5-triphosphate (IP3)-
enters the cytosol where it is dismuted to H2O2 by sensitive sarcoplamic reticulum (SR) stores and
cytosolic Cu,Zn SOD. ryanodine-sensitive SR stores. Gelband and Gel-
band (1997) and Robertson et al. (2001) found
that hypoxic contraction of isolated pulmonary
7. Role of mitochondria in phase 1 of the HPV arteries was inhibited by thapsigargin, which
response blocks the uptake of Ca2 + into intracellular
stores. These observations suggest that hypoxia
Phase 1 of HPV is transient and occurs in PA triggers the release of Ca2 + from IP3-sensitive SR
88 G.B. Waypa, P.T. Schumacker / Respiratory Physiology & Neurobiology 132 (2002) 81–91

stores. Along similar lines, exogenous H2O2 has tured endothelium increases during hypoxia (Bodi
been shown to activate phospholipase C (PLC) in et al., 1995; Hu et al., 1998; Yamashita et al.,
a time- and concentration-dependent manner 2001), and BQ-123, an ETA antagonist, attenu-
(Wang et al., 2001). This suggests the possibility ates phase 2 of HPV in isolated vessels and whole
that mitochondrial ROS could trigger intracellu- lungs (Sato et al., 2000; Liu et al., 2001). This
lar Ca2 + release by activating PLC. PLC in turn suggests that an O2 sensor must also exist within
would metabolize phosphatidylinositol 4,5-bis- the PA endothelium. However, the relationship
phosphate (PIP2) to inositol 1,4,5-trisphosphate between the O2 sensor in the myocytes and the
(IP3) and diacylglycerol (DAG). IP3 could then sensor in endothelium is not known.
release Ca2 + from IP3-sensative Ca2 + stores. Recently, Hu et al. (1998) reported that the
However, studies demonstrating this mechanism proximal promoter of the ET-1 gene contains a
have not been published. hypoxia-inducible factor 1 (HIF-1) binding site on
Hypoxia has been shown to trigger the release the antisense DNA strand positioned at −118 to
of Ca2 + from ryanodine-sensitive SR stores (Gel- − 125 base pairs upstream from the transcription
band and Gelband, 1997; Dipp and Evans, 2001; start site (Hu et al., 1998). HIF-1 has been shown
Dipp et al., 2001; Liu et al., 2001; Wilson et al., to be the principal O2-responsive factor underly-
2001; Morio and McMurtry, 2002). Recently, ing increased expression of glycolytic enzymes,
Wilson et al. (2001) reported that hypoxia in- glucose transporters, and other genes during hy-
creases cyclic ADP-ribose accumulation in iso- poxia (Semenza, 2001a,b,c). This suggests a mech-
lated rabbit PA and Ca2 + release from anism in which hypoxia could regulate ET-1
ryanodine-sensitive SR stores. Richter et al. expression and release by endothelial cells
(1995) reported that H2O2 could cause oxidation through HIF-1 activation. Several groups have
of mitochondrial pyridine nucleotides, resulting in found evidence that hypoxia-induced mitochon-
increased ADP-ribose production, monoADP-ri- drial ROS generation activates HIF-1 during hy-
bosylation of mitochondrial proteins, and calcium poxia (Chandel et al., 1998, 2000; Agani et al.,
release (Richter et al., 1995). These results suggest 2000; Semenza, 2000). Furthermore, hypoxia in-
a mechanism in which mitochondrial ROS gener- creases ROS generation at complex III of the
ation could trigger Ca2 + release from IP3- and/or mitochondria in endothelial cells (Pearlstein et al.,
ryanodine-sensitive SR stores. The resulting in- 2002). This suggests that the mitochondria in PA
crease in intracellular Ca2 + could then trigger endothelium could contribute to phase 2 by in-
HPV by inhibiting KV-channels. The increase in creasing ROS generation, resulting in increased
[Ca2 + ]i resulting from membrane depolarization ET-1 expression and release, which in turn would
would trigger calmodulin-mediated activation of contribute to PA myocyte contraction. It seems
myosin light chain kinase, actin– myosin interac- reasonable to predict that such a response could
tion, and contraction. be important for the enhanced contraction and
even vascular remodeling during periods of sus-
tained or chronic hypoxia, but more definitive
8. Role of mitochondria in phase 2 of the HPV experiments are needed to test this hypothesis
response directly.

Phase 2 of HPV involves a slow sustained vaso-


constriction that is dependent on the endothelium. 9. Conclusion
An endothelium-derived vasoconstrictor may
therefore be required for phase 2. ET-1, a 21- It is clear that a consensus has yet to be reached
residue peptide synthesized and secreted by en- regarding the O2 sensing mechanism underlying
dothelial cells, has been proposed to be the the HPV response. However, mitochondria have
vasoconstrictor involved in phase 2 of the HPV once again stepped into the spotlight as a likely
response. ET-1 expression and release from cul- O2 sensor involved in the initiation of HPV. This
G.B. Waypa, P.T. Schumacker / Respiratory Physiology & Neurobiology 132 (2002) 81–91 89

model is attractive given that the mitochondria dothelin-1 gene regulation by hypoxia. Cardiovasc. Res.
are the primary consumers of oxygen in the cell. 30, 975 – 984.
Buescher, P.C., Pearse, D.B., Pillai, R.P., Litt, M.C., Mitchell,
However, recent studies by different laboratories M.C., Sylvester, J.T., 1991. Energy state and vasomotor
suggest two opposing explanations of how the tone in hypoxic pig lungs. J. Appl. Physiol. 70, 1874 – 1881.
mitochondria could trigger a biological response Bunn, H.F., Poyton, R.O., 1996. Oxygen sensing and molecu-
to hypoxia. At this point, more definitive experi- lar adaptation to hypoxia. Physiol. Rev. 76, 839 – 885.
ments are needed to provide a resolution to these Chandel, N.S., Maltepe, E., Goldwasser, E., Mathieu, C.E.,
Simon, M.C., Schumacker, P.T., 1998. Mitochondrial reac-
differences. tive oxygen species trigger hypoxia-induced transcription.
Proc. Natl. Acad. Sci. USA 95, 11715 – 11720.
Chandel, N.S., McClintock, D.S., Feliciano, C.E., Wood,
Acknowledgements T.M., Melendez, J.A., Rodriguez, A.M., Schumacker, P.T.,
2000. Reactive oxygen species generated at mitochondrial
complex III stabilize hypoxia-inducible factor-1alpha dur-
This work was supported by NHLBI grants
ing hypoxia: a mechanism of O2 sensing. J. Biol. Chem.
HL66315 and HL10405. 275, 25130 – 25138.
Coppock, E.A., Martens, J.R., Tamkun, M.M., 2001. Molecu-
lar basis of hypoxia-induced pulmonary vasoconstriction:
References role of voltage-gated K+ channels. Am. J. Physiol. 281,
L1– 12.
Agani, F.H., Pichiule, P., Chavez, J.C., LaManna, J.C., 2000. Dipp, M., Evans, A.M., 2001. Cyclic ADP-ribose is the pri-
The role of mitochondria in the regulation of hypoxia-in- mary trigger for hypoxic pulmonary vasoconstriction in the
ducible factor 1 expression during hypoxia. J. Biol. Chem. rat lung in situ. Circ. Res. 89, 77 – 83.
275, 35863 – 35867. Dipp, M., Nye, P.C., Evans, A.M., 2001. Hypoxic release of
Archer, S.L., Huang, J., Henry, T., Peterson, D., Weir, E.K., calcium from the sarcoplasmic reticulum of pulmonary
1993. A redox-based O2 sensor in rat pulmonary vascula- artery smooth muscle. Am. J. Physiol. 281, L318 – 325.
ture. Circ. Res. 73, 1100 –1112. Duranteau, J., Chandel, N.S., Kulisz, A., Shao, Z., Schu-
Archer, S.L., Souil, E., Dinh-Xuan, A.T., Schremmer, B., macker, P.T., 1998. Intracellular signaling by reactive oxy-
Mercier, J.C., El Yaagoubi, A., Nguyen-Huu, L., Reeve, gen species during hypoxia in cardiomyocytes. J. Biol.
H.L., Hampl, V., 1998. Molecular identification of the role Chem. 273, 11619 – 11624.
of voltage-gated K+ channels, KV1.5 and KV2.1, in hy- Gelband, C.H., Gelband, H., 1997. Ca2 + release from intra-
poxic pulmonary vasoconstriction and control of resting cellular stores is an initial step in hypoxic pulmonary
membrane potential in rat pulmonary artery myocytes. J. vasoconstriction of rat pulmonary artery resistance vessels.
Clin. Invest. 101, 2319 –2330. Circulation 96, 3647 – 3654.
Archer, S.L., Reeve, H.L., Michelakis, E., Puttagunta, L., Grimminger, F., Weissmann, N., Spriestersbach, R., Becker,
Waite, R., Nelson, D.P., Dinauer, M.C., Weir, E.K., 1999. E., Rosseau, S., Seeger, W., 1995. Effects of NADPH
O2 sensing is preserved in mice lacking the gp91 phox oxidase inhibitors on hypoxic vasoconstriction in buffer-
subunit of NADPH oxidase. Proc. Natl. Acad. Sci. USA perfused rabbit lungs. Am. J. Physiol. 268, L747 – 752.
96, 7944 – 7949. Hasunuma, K., Rodman, D.M., McMurtry, I.F., 1991. Effects
Archer, S.L., Weir, E.K., Reeve, H.L., Michelakis, E., 2000. of K+ channel blockers on vascular tone in the perfused
Molecular identification of O2 sensors and O2-sensitive rat lung. Am. Rev. Respir. Dis. 144, 884 – 887.
potassium channels in the pulmonary circulation. Adv. Holland, P.C., Clark, M.G., Bloxham, D.P., Lardy, H.A.,
Exp. Med. Biol. 475, 219 –240. 1973. Mechanism of action of the hypoglycemic agent
Archer, S.L., London, B., Hampl, V., Wu, X., Nsair, A., diphenyleneiodonium. J. Biol. Chem. 248, 6050 – 6056.
Puttagunta, L., Hashimoto, K., Waite, R.E., Michelakis, Hu, J., Discher, D.J., Bishopric, N.H., Webster, K.A., 1998.
E.D., 2001. Impairment of hypoxic pulmonary vasocon- Hypoxia regulates expression of the endothelin-1 gene
striction in mice lacking the voltage-gated potassium chan- through a proximal hypoxia-inducible factor-1 binding site
nel KV1.5. FASEB J. 15, 1801 –1803. on the antisense strand. Biochem. Biophys. Res. Commun.
Barman, S.A., 1998. Potassium channels modulate hypoxic 245, 894 – 899.
pulmonary vasoconstriction. Am. J. Physiol. 275, L64 – Jones, D.P., Kennedy, F.G., 1982. Intracellular oxygen supply
L70. during hypoxia. Am. J. Physiol. 243, C247 – 253.
Beavis, A.D., Davatol-Hag, H., 1996. The mitochondrial inner Jones, D.P., Kennedy, F.G., 1986. Analysis of intracellular
membrane anion channel is inhibited by DIDS. J. Bioen- oxygenation of isolated adult cardiac myocytes. Am. J.
erg. Biomembr. 28, 207 –214. Physiol. 250, C384 – 390.
Bodi, I., Bishopric, N.H., Discher, D.J., Wu, X., Webster, Jones, R.D., Morice, A.H., 2000. Hydrogen peroxide — an
K.A., 1995. Cell-specificity and signaling pathway of en- intracellular signal in the pulmonary circulation: involve-
90 G.B. Waypa, P.T. Schumacker / Respiratory Physiology & Neurobiology 132 (2002) 81–91

ment in hypoxic pulmonary vasoconstriction. Pharmacol. Morio, Y., McMurtry, I.F., 2002. Ca2 + release from ryan-
Ther. 88, 153 – 161. odine-sensitive store contributes to mechanism of hypoxic
Jones, R.D., Hancock, J.T., Morice, A.H., 2000. NADPH vasoconstriction in rat lungs. J. Appl. Physiol. 92, 527 –
oxidase: a universal oxygen sensor? Free Radic. Biol. Med. 534.
29, 416 – 424. Parsons, D.F., Williams, G.R., Chance, B., 1966. Characteris-
Killilea, D.W., Hester, R., Balczon, R., Babal, P., Gillespie, tics of isolated and purified preparations of the outer and
M.N., 2000. Free radical production in hypoxic pulmonary inner membranes of mitochondria. Ann. N. Y. Acad. Sci.
artery smooth muscle cells. Am. J. Physiol. 279, L408 –412. 137, 643 – 666.
Lazor, R., Feihl, F., Waeber, B., Kucera, P., Perret, C., 1996. Pearlstein D.P., Ali, M.H., Mungai, P.T., Hynes, K.L., Gew-
Endothelin-1 does not mediate the endothelium-dependent ertz, B.L., Schumacker, P.T., 2002. Role of mitochondrial
hypoxic contractions of small pulmonary arteries in rats. oxidant generation in endothelial cell responses to hypoxia.
Chest 110, 189 – 197. Arterioscler. Thromb. Vasc. Biol. 566 – 573.
Leach, R.M., Sheehan, D.W., Chacko, V.P., Sylvester, J.T., Post, J.M., Gelband, C.H., Hume, J.R., 1995. [Ca2 + ]i inhibi-
2000. Energy state, pH, and vasomotor tone during hy- tion of K+ channels in canine pulmonary artery. Novel
poxia in precontracted pulmonary and femoral arteries. mechanism for hypoxia-induced membrane depolarization.
Am. J. Physiol. 278, L294 –304. Circ. Res. 77, 131 – 139.
Leach, R.M., Hill, H.M., Snetkov, V.A., Robertson, T.P., Reeve, H.L., Weir, E.K., Nelson, D.P., Peterson, D.A.,
Ward, J.P., 2001. Divergent roles of glycolysis and the Archer, S.L., 1995. Opposing effects of oxidants and an-
mitochondrial electron transport chain in hypoxic pul- tioxidants on K+ channel activity and tone in rat vascular
monary vasoconstriction of the rat: identity of the hypoxic tissue. Exp. Physiol. 80, 825 – 834.
sensor. J. Physiol., Lond. 536, 211 –224. Reeve, H.L., Michelakis, E., Nelson, D.P., Weir, E.K., Archer,
Liu, Q., Sham, J.S., Shimoda, L.A., Sylvester, J.T., 2001. S.L., 2001. Alterations in a redox oxygen sensing mecha-
Hypoxic constriction of porcine distal pulmonary arteries: nism in chronic hypoxia. J. Appl. Physiol. 90, 2249 – 2256.
endothelium and endothelin dependence. Am. J. Physiol.
Richter, C., Gogvadze, V., Laffranchi, R., Schlapbach, R.,
280, L856 – 865.
Schweizer, M., Suter, M., Walter, P., Yaffee, M., 1995.
Majander, A., Finel, M., Wikstrom, M., 1994.
Oxidants in mitochondria: from physiology to diseases.
Diphenyleneiodonium inhibits reduction of iron –sulfur
Biochim. Biophys. Acta 1271, 67 – 74.
clusters in the mitochondrial NADH –ubiquinone oxidore-
Robertson, T.P., Ward, J.P., Aaronson, P.I., 2001. Hypoxia
ductase (complex I). J. Biol. Chem. 269, 21037 – 21042.
induces the release of a pulmonary-selective, Ca2 + -sensitis-
Marshall, B.E., Marshall, C., Frasch, F., Hanson, C.W., 1994.
ing, vasoconstrictor from the perfused rat lung. Cardio-
Role of hypoxic pulmonary vasoconstriction in pulmonary
vasc. Res. 50, 145 – 150.
gas exchange and blood flow distribution. 1. Physiologic
Rounds, S., McMurtry, I.F., 1981. Inhibitors of oxidative
concepts. Intensive Care Med. 20, 291 –297.
ATP production cause transient vasoconstriction and
Marshall, C., Mamary, A.J., Verhoeven, A.J., Marshall, B.E.,
block subsequent pressor responses in rat lungs. Circ. Res.
1996. Pulmonary artery NADPH-oxidase is activated in
hypoxic pulmonary vasoconstriction. Am. J. Respir. Cell 48, 393 – 400.
Mol. Biol. 15, 633 – 644. Sato, K., Morio, Y., Morris, K.G., Rodman, D.M., Mc-
McMurtry, I.F., 1984. Angiotensin is not required for hypoxic Murtry, I.F., 2000. Mechanism of hypoxic pulmonary
constriction in salt solution-perfused rat lungs. J. Appl. vasoconstriction involves ET(A) receptor-mediated inhibi-
Physiol. 56, 375 – 380. tion of K(ATP) channel. Am. J. Physiol. 278, L434 – 442.
Michelakis, E.D., Weir, E.K., 2001. The pathobiology of Sato, K., Ogawa, K., Tokmakov, A.A., Iwasaki, T., Fukami,
pulmonary hypertension. Smooth muscle cells and ion Y., 2001. Hydrogen peroxide induces Src family tyrosine
channels. Clin. Chest Med. 22, 419 –432. kinase-dependent activation of Xenopus eggs. Dev.
Michelakis, E.D., Archer, S.L., Weir, E.K., 1995. Acute hy- Growth Differ. 43, 55 – 72.
poxic pulmonary vasoconstriction: a model of oxygen sens- Semenza, G.L., 2000. Expression of hypoxia-inducible factor
ing. Physiol. Res. 44, 361 –367. 1: mechanisms and consequences. Biochem. Pharmacol. 59,
Michelakis, E.D., Reeve, H.L., Huang, J.M., Tolarova, S., 47 – 53.
Nelson, D.P., Weir, E.K., Archer, S.L., 1997. Potassium Semenza, G.L., 2001a. HIF-1, O2 and the 3 PHDs: how
channel diversity in vascular smooth muscle cells. Can. J. animal cells signal hypoxia to the nucleus. Cell 107, 1 – 3.
Physiol. Pharmacol. 75, 889 –897. Semenza, G.L., 2001b. Hypoxia-inducible factor 1: control of
Mohazzab, K.M., Fayngersh, R.P., Kaminski, P.M., Wolin, oxygen homeostasis in health and disease. Pediatr. Res. 49,
M.S., 1995. Potential role of NADH oxidoreductase- 614 – 617.
derived reactive O2 species in calf pulmonary arterial PO2- Semenza, G.L., 2001c. HIF-1 and mechanisms of hypoxia
elicited responses. Am. J. Physiol. 269, L637 –644. sensing. Curr. Opin. Cell Biol. 13, 167 – 171.
Monaco, J.A., Burke-Wolin, T., 1995. NO and H2O2 mecha- Sham, J.S., Crenshaw, B.R. Jr, Deng, L.H., Shimoda, L.A.,
nisms of guanylate cyclase activation in oxygen-dependent Sylvester, J.T., 2000. Effects of hypoxia in porcine pul-
responses of rat pulmonary circulation. Am. J. Physiol. monary arterial myocytes: roles of KV channel and en-
268, L546 – 550. dothelin-1. Am. J. Physiol. 279, L262 – L272.
G.B. Waypa, P.T. Schumacker / Respiratory Physiology & Neurobiology 132 (2002) 81–91 91

Sohal, R.S., Weindruch, R., 1996. Oxidative stress, caloric Weir, E.K., Archer, S.L., 1995. The mechanism of acute
restriction and aging. Science 273, 59 –63. hypoxic pulmonary vasoconstriction: the tale of two chan-
Stolk, J., Hiltermann, T.J., Dijkman, J.H., Verhoeven, A.J., nels. FASEB J. 9, 183 – 189.
1994. Characteristics of the inhibition of NADPH oxidase Weir, E.K., Reeve, H.L., Peterson, D.A., Michelakis, E.D.,
activation in neutrophils by apocynin, a methoxy-substi- Nelson, D.P., Archer, S.L., 1998. Pulmonary vasoconstric-
tuted catechol. Am. J. Respir. Cell Mol. Biol. 11, 95 –102. tion, oxygen sensing, and the role of ion channels: Thomas
Sweeney, M., Yuan, J.X., 2000. Hypoxic pulmonary vasocon- A. Neff lecture. Chest 114, 17S – 22S.
striction: role of voltage-gated potassium channels. Respir. Weissmann, N., Grimminger, F., Walmrath, D., Seeger, W.,
Res. 1, 40 – 48. 1995. Hypoxic vasoconstriction in buffer-perfused rabbit
Sylvester, J.T., 2001. Hypoxic pulmonary vasoconstriction: a lungs. Respir. Physiol. 100, 159 – 169.
radical view. Circ. Res. 88, 1228 –1230. Weissmann, N., Grimminger, F., Voswinckel, R., Conzen, J.,
Terada, L.S., 1996. Hypoxia-reoxygenation increases O ’− 2 Seeger, W., 1998. Nitro blue tetrazolium inhibits but does
efflux which injures endothelial cells by an extracellular not mimic hypoxic vasoconstriction in isolated rabbit
mechanism. Am. J. Physiol. 270, H945 –H950. lungs. Am. J. Physiol. 274, L721 – 727.
Urena, J., Franco-Obregon, A., Lopez-Barneo, J., 1996. Con- Weissmann, N., Grimminger, F., Olschewski, A., Seeger, W.,
trasting effects of hypoxia on cytosolic Ca2 + spikes in 2001. Hypoxic pulmonary vasoconstriction: a multifacto-
conduit and resistance myocytes of the rabbit pulmonary rial response? Am. J. Physiol. 281, L314 – 317.
artery. J. Physiol., Lond. 496, 103 –109. Wilson, H.L., Dipp, M., Thomas, J.M., Lad, C., Galione, A.,
von Euler, U., Liljestrand, G., 1946. Observations on the Evans, A.M., 2001. Adp-ribosyl cyclase and cyclic ADP-ri-
pulmonary arterial blood pressure of the cat. Acta Physiol. bose hydrolase act as a redox sensor. a primary role for
Scand. 12, 301 – 320. cyclic ADP-ribose in hypoxic pulmonary vasoconstriction.
Wang, X.T., McCullough, K.D., Wang, X.J., Carpenter, G., J. Biol. Chem. 276, 11180 – 11188.
Holbrook, N.J., 2001. Oxidative stress-induced phospholi- Yamashita, K., Discher, D.J., Hu, J., Bishopric, N.H., Web-
pase C-gamma 1 activation enhances cell survival. J. Biol. ster, K.A., 2001. Molecular regulation of the endothelin-1
Chem. 276, 28364 – 28371. gene by hypoxia. Contributions of hypoxia-inducible fac-
Ward, J.P., Aaronson, P.I., 1999. Mechanisms of hypoxic tor-1, activator protein-1, GATA-2 and p300/CBP. J. Biol.
pulmonary vasoconstriction: can anyone be right? Respir. Chem. 276, 12645 – 12653.
Physiol. 115, 261 – 271. Zhang, F., Carson, R.C., Zhang, H., Gibson, G., Thomas,
Ward, J.P., Robertson, T.P., 1995. The role of the endothe- H.M. III, 1997. Pulmonary artery smooth muscle cell
lium in hypoxic pulmonary vasoconstriction. Exp. Physiol. [Ca2 +]i and contraction: responses to diphenyleneiodo-
80, 793 – 801. nium and hypoxia. Am. J. Physiol. 273, L603 – 611.
Waypa, G.B., Morton, C.A., Vincent, P.A., Mahoney, J.R. Jr, Zhao, Y., Rhoades, R.A., Packer, C.S., 1996. Hypoxia-in-
Johnston, W.K. III, Minnear, F.L., 2000. Oxidant-in- duced pulmonary arterial contraction appears to be depen-
creased endothelial permeability: prevention with phospho- dent on myosin light chain phosphorylation. Am. J.
diesterase inhibition vs. cAMP production. J. Appl. Physiol. 271, L768 – 774.
Physiol. 88, 835 – 842. Zhu, H., Qiu, H., Yoon, H.W., Huang, S., Bunn, H.F., 1999.
Waypa, G.B., Chandel, N.S., Schumacker, P.T., 2001. Model Identification of a cytochrome b-type NAD(P)H oxidore-
for hypoxic pulmonary vasoconstriction involving mito- ductase ubiquitously expressed in human cells. Proc. Natl.
chondrial oxygen sensing. Circ. Res. 88, 1259 –1266. Acad. Sci. USA 96, 14742 – 14747.

You might also like