Está en la página 1de 5

Exp Physiol 101.

1 (2016) pp 23–27 23

Symposium Report

Energy metabolism and the high-altitude environment


Andrew J. Murray
Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK

New Findings
r What is the topic of this review?
This report describes changes in cardiac and skeletal muscle energy metabolism that occur
with exposure to high altitude and considers possible underlying mechanisms.
r What advances does it highlight?
In the human heart, sustained hypoxia at high altitude or shorter-term normobaric hypoxia
result in a loss of cardiac energetic reserve. In the hypoxic rat heart, fatty acid oxidation
and respiratory capacity fall. In skeletal muscle, prolonged exposure to extreme high altitude
Experimental Physiology

results in the loss of mitochondrial density, but even at more moderate high altitude the
respiratory capacity may be suppressed. Evidence from cells, genetically modified mice and
high-altitude-adapted Tibetans suggests a possible mechanistic role for the hypoxia-inducible
factor pathway.

At high altitude the barometric pressure falls, challenging oxygen delivery to the tissues. Thus,
whilst hypoxia is not the only physiological stress encountered at high altitude, low arterial
P O2 is a sustained feature, even after allowing adequate time for acclimatization. Cardiac and
skeletal muscle energy metabolism is altered in subjects at, or returning from, high altitude.
In the heart, energetic reserve falls, as indicated by lower phosphocreatine-to-ATP ratios. The
underlying mechanism is unknown, but in the hypoxic rat heart fatty acid oxidation and
respiratory capacity are decreased, whilst pyruvate oxidation is also lower after sustained hypoxic
exposure. In skeletal muscle, there is not a consensus. With prolonged exposure to extreme
high altitude (>5500 m) a loss of muscle mitochondrial density is seen, but this was not
observed in a simulated ascent of Everest in hypobaric chambers. At more moderate high
altitude, decreased respiratory capacity may occur without changes in mitochondrial volume
density, and fat oxidation may be downregulated, although this is not seen in all studies. The
underlying mechanisms, including the possible role of hypoxia-signalling pathways, remain to
be resolved, particularly in light of confounding factors in the high-altitude environment. In
high-altitude-adapted Tibetan natives, however, there is evidence of natural selection centred
around the hypoxia-inducible factor pathway, and metabolic features in this population (e.g.
low cardiac phosphocreatine-to-ATP ratios, increased cardiac glucose uptake and lower muscle
mitochondrial densities) share similarities with those in acclimatized lowlanders, supporting a
possible role for the hypoxia-inducible factor pathway in the metabolic response of cardiac and
skeletal muscle energy metabolism to high altitude.

(Received 12 July 2015; accepted after revision 20 August 2015; first published online 28 August 2015)
Corresponding author A. J. Murray: Department of Physiology, Development and Neuroscience, University of
Cambridge, Downing Street, Cambridge CB2 3EG, UK. Email: ajm267@cam.ac.uk


C 2015 The Authors. Experimental Physiology 
C 2015 The Physiological Society DOI: 10.1113/EP085317
24 A. J. Murray Exp Physiol 101.1 (2016) pp 23–27

Introduction and a consideration of the limits of human tolerance is a


stimulating intellectual challenge in its own right and adds
The defining feature of the high-altitude environment to our understanding of homeostatic regulation. Third,
is sustained hypobaric hypoxia. Whilst the atmosphere exposure to sustained physiological challenge in extreme
is 21% oxygen at all altitudes, barometric pressure falls environments has been proposed as a possible analogue
upon ascent, and with it inspired P O2 , challenging oxygen of human disease (Grocott et al. 2007), and whilst this
delivery to the tissues. The majority of the world’s approach risks overinterpretation of similarities between
population inhabits regions below 1000 m elevation, but settings it does allow for the study of preselected and
nevertheless, our species has a remarkable capacity for otherwise healthy cohorts in adverse conditions. Finally,
hypoxia tolerance. Most notably, over tens of thousands the natural experiment of high-altitude adaptation in
of years some high-altitude populations have adapted East Africa, the Tibetan Plateau and the Andes offers an
to life in this environment, with genetic signatures invaluable scenario in which to study human genetics and
revealing natural selection around hypoxia-sensing natural selection (Bigham & Lee, 2014).
pathways (Bigham & Lee, 2014). Approximately 140
million people live above 2500 m, but around 40 million
others venture into high-altitude regions each year for
Cardiac energy metabolism at high altitude
work or leisure (West et al. 2007). These lowlanders
experience a physiological response upon ascent, which In subjects returning from Everest Base Camp
has been well documented elsewhere and includes (5300 m), cardiac phosphocreatine-to-ATP ratios
ventilatory, cardiovascular and erythropoietic aspects (PCr/ATP; measured using 31 P NMR spectroscopy)
(West et al. 2007). Even with adequate acclimatization, decreased by 18% (Holloway et al. 2011b), indicating a
energy metabolism is altered in human heart and skeletal loss of energetic reserve. This was accompanied by altered
muscle. This might indicate a failure to compensate fully diastolic function and a loss of left ventricular (LV) mass,
or may itself form part of the acclimatization process, but it but all measures returned to normal after 6 months back
involves changes in gene expression and thus appears to be at sea level. In an accompanying study, shorter term
a regulated response. The positive aspects of high-altitude exposure to normobaric hypoxia (20 h, 50–60 mmHg
acclimatization, most notably decreased susceptibility atmospheric P O2 ) also elicited a loss of cardiac energetics
to acute mountain sickness, also contrast with the (Holloway et al. 2011a). The PCr/ATP fell by 15%,
less well-understood phenomenon of high-altitude and echocardiographic measurements suggested a mild
deterioration, which occurs with prolonged exposure to alteration in diastolic function. Whilst different cellular
extreme high altitude (>5500 m) and is characterized by mechanisms may underlie the observations in these
lethargy, fatigue and muscle wasting (Ward, 1954). studies, in both instances exposure to a hypoxic stimulus
Hypoxia is not, however, the only stress encountered resulted in a loss of cardiac energetic reserve. This is
at altitude (West et al. 2007). Temperature falls with reminiscent of findings in the hearts of highland-dwelling
increasing elevation, whilst absolute humidity is extremely Sherpas, who had lower a cardiac PCr/ATP than
low and exposure to solar/ultraviolet radiation high. lowlanders, which persisted even after 4 weeks spent
Visitors frequently experience gastrointestinal upset and de-acclimatizing at low altitude (Hochachka et al. 1996).
appetite loss, which could result from the hypoxia itself, Studies of rats in hypoxic chambers have helped to
but may be exacerbated by infection, particularly in elucidate the cardiac metabolic response to sustained
developing countries. In addition, activity levels are environmental hypoxia. In rats, exposure to 11% O2
often altered, as oxygen delivery limits exercise capacity for 1 week resulted in decreased LV expression of genes
and motivation falls; thus, individuals may undergo regulated by the fatty acid (FA)-activated transcription
detraining. factor peroxisome proliferator-activated receptor α
In light of these confounding factors, the high-altitude (PPARα), including carnitine palmitoyl-transferase 1,
environment is not a perfect setting in which to study pyruvate dehydrogenase kinase 4 and uncoupling
the physiological response to hypoxia alone, although protein 3, suggesting downregulation of FA oxidation
other contexts (e.g. confinement to normobaric hypoxia and increased pyruvate oxidation (Essop et al. 2004).
chambers) have limitations too. The study of humans Whilst this was associated with decreased O2 consumption
(and other animals) at altitude is nevertheless a valuable and ATP synthesis, mitochondrial coupling was preserved
research endeavour. First, with increasing numbers of (Essop et al. 2004). After 12 weeks, expression of FA
people visiting mountainous regions, an understanding oxidation genes decreased in both ventricles, whereas
of the integrated response to altitude is vital, particularly expression of pyruvate dehydrogenase kinase 4 mRNA
the interindividual variation in this response, which is not increased in the LV, suggesting inhibition of pyruvate
fully explained by classic descriptions of acclimatization. oxidation with more sustained exposure (Adrogue et al.
Second, the study of extreme environment physiology 2005).


C 2015 The Authors. Experimental Physiology 
C 2015 The Physiological Society
Exp Physiol 101.1 (2016) pp 23–27 Energy metabolism at high altitude 25

In support of this suggestion, 2 weeks at 11% O2 co-activator 1 alpha (PGC1α), and corresponding changes
decreased respiration rates with FA substrates (palmitoyl- in protein levels of metabolic regulators and enzymes
carnitine or palmitoyl-CoA) and pyruvate in both (Levett et al. 2012).
subsarcolemmal and intermyofibrillar subpopulations During Operation Everest II (a gradual decompression
of mitochondria from rat LV/septum. Curiously, to the equivalent of 8840 m in a chamber), however,
O2 consumption with the electron transport chain no such change in mitochondrial volume density was
complex I substrate, glutamate, was lowered in hypoxic observed (Green et al. 1989). This simulation took place
subsarcolemmal but not intermyofibrillar mitochondria, over a slightly shorter time frame than a classic Everest
alongside decreased complex I protein (Heather et al. ascent, but with the subjects confined to a chamber a
2012). Work from our laboratory confirmed a loss of greater loss of mitochondria might have been expected
complex I-supported respiration, FA oxidation capacity due to detraining. The discrepancy has not been resolved,
and ATP levels in the LV of hypoxic rats (13% O2 , but whatever the cause the loss of mitochondria at
2 weeks), alongside increased oxidative stress. All changes extreme altitude fits neatly alongside Ward’s description
were prevented, however, by dietary supplementation of high-altitude deterioration (Ward, 1954).
with a moderate dose of nitrate, which improved tissue At more moderate high altitude, even with prolonged
oxygenation, possibly via improved blood flow and/or exposure, no such loss in mitochondrial volume density
tissue metabolic efficiency (Ashmore et al. 2014). occurs, although notably, lower muscle mitochondrial
In the hypoxic rat heart, therefore, FA oxidation densities have been reported in some high-altitude natives
is downregulated, with a loss of respiratory capacity (Kayser et al. 1991, 1996). Changes in muscle respiratory
in the LV. Pyruvate oxidation may also be inhibited, function occur at more moderate altitudes, but again
suggesting increased glycolysis. Whilst little is known this may be dependent on the extent of exposure. Two
about substrate metabolism in the hypoxic human heart, similar high-resolution respirometry studies by Lundby
glucose uptake was higher in the hearts of highland natives and co-workers described a loss of respiratory capacity
than lowlanders (Holden et al. 1995). and improved coupling following 28 days at 3454 m
(Jacobs et al. 2012), but no changes after 9–11 days at
4559 m (Jacobs et al. 2013). Meanwhile, some studies have
Skeletal muscle energy metabolism at high altitude
reported the downregulation of FA oxidative enzymes
In comparison with studies of cardiac energy metabolism, with prolonged exposure to altitudes between 4300 and
more numerous investigations have been carried out 8848 m (Roberts et al. 1996; Levett et al. 2012), although
into the metabolic response of human skeletal muscle no such differences were found in another study at
at altitude, perhaps owing to the relative ease of 5250 m (Mizuno et al. 2008). Despite changes in resting
obtaining biopsies. This has not, however, resulted in metabolites, however, muscle PCr recovery half-times
a consistent picture, perhaps because of varied ascent following an exercise challenge were remarkably well
profiles, training status and exposure to confounding preserved in subjects returning either from Everest Base
features of the high-altitude environment. In a recent Camp or the summit, indicating that muscle capacity for
review, we attempted to find consensus amongst studies ATP synthesis may in fact be spared (Edwards et al. 2010).
of metabolism in human muscle at altitude, plus humans The recent discovery of genetic selection on the
and rodents in hypoxia chambers (Horscroft & Murray, PPARA gene in Tibetan populations (Simonson et al.
2014). Considering only human studies, a picture emerges 2010) suggests that altered FA metabolism may be a
in which the duration and degree of exposure to hypoxia feature of long-term adaptation to high altitude, because
may be crucial. the gene encodes the FA-activated transcription factor
With prolonged exposure to extreme high altitude, as and regulator of FA oxidation, PPARα. The functional
experienced on Himalayan mountaineering expeditions, a significance of this polymorphism is currently unresolved,
loss of muscle mitochondrial volume density occurs, with but recent work carried out by our group, in collaboration
the greatest loss from the subsarcolemmal population of with Professor Erich Gnaiger and Dr Julian Griffin, has
mitochondria (Hoppeler et al. 1990; Levett et al. 2012). used high-resolution respirometry and metabolic profiling
It is not clear whether this is caused by hypoxia (or the to investigate FA metabolism in the muscles of lowlanders
accompanying oxidative stress) or is instead a detraining and Sherpas at altitudes up to 5300 m (Gilbert-Kawai et al.
response, but notably, the subsarcolemmal population of 2015).
mitochondria is more susceptible to changes in volume
density during training studies at sea level (Desplanches Mechanisms, unanswered questions and future
et al. 1993). The response does, however, appear to be
directions
regulated, with altered expression of genes including
a 50% loss of mRNA of the mitochondrial biogenesis What mechanisms might underlie the changes in heart
factor, peroxisome proliferator-activated receptor gamma and muscle metabolism at altitude? Hypoxia-signalling


C 2015 The Authors. Experimental Physiology 
C 2015 The Physiological Society
26 A. J. Murray Exp Physiol 101.1 (2016) pp 23–27

pathways are an intuitive possibility. Many of the metabolic are probably distinct. Confounding factors such as changes
changes reported in humans at altitude or in the hypoxic in activity or diet at altitude cannot, however, be ruled
rat heart (e.g. pyruvate dehydrogenase inhibition and out. Future studies combining different hypoxic protocols
decreased mitochondrial respiration) have also been with controlled activity/inactivity and diet could thus
observed in hypoxic cells in culture and are associated be revealing and have the potential to elucidate the
with stabilization of the hypoxia-inducible factor (HIF) mechanisms underlying the metabolic changes in human
family of transcription factors (reviewed by Murray, 2009). heart and muscle at high altitude.
Adding support to a HIF-driven mechanism, oxidative
metabolism, and thus exercise capacity, were augmented in
References
the gastrocnemius muscle of mice in which skeletal muscle
HIF-1α had been selectively deleted (Mason et al. 2007). Adrogue JV, Sharma S, Ngumbela K, Essop MF & Taegtmeyer
The effects of the HIF pathway on muscle metabolism are H (2005). Acclimatization to chronic hypobaric hypoxia is
outlined elsewhere in this issue of Experimental Physiology associated with a differential transcriptional profile between
by Dr Helene Rundqvist (Lindholm et al. 2016). the right and left ventricle. Mol Cell Biochem 278, 71–78.
An alternative possibility to low muscle P O2 per se Ashmore T, Fernandez BO, Branco-Price C, West JA, Cowburn
AS, Heather LC, Griffin JL, Johnson RS, Feelisch M &
could be reactive oxygen species (ROS)-mediated effects,
Murray AJ (2014). Dietary nitrate increases arginine
because mitochondrial ROS production increases in availability and protects mitochondrial complex I and
hypoxia (Guzy & Schumacker, 2006) and is modelled to energetics in the hypoxic rat heart. J Physiol 592,
increase sharply in muscle above altitudes corresponding 4715–4731.
to those of the highest permanent human settlements Bigham AW & Lee FS (2014). Human high-altitude adaptation:
(Cano et al. 2014). Reactive oxygen species have sometimes forward genetics meets the HIF pathway. Genes Dev 28,
been described as indiscriminate mediators of damage 2189–2204.
to lipids, protein and DNA, and this may be the case Cano I, Selivanov V, Gomez-Cabrero D, Tegnér J, Roca J,
when generated in large quantities, but at more moderate Wagner PD & Cascante M (2014). Oxygen pathway modeling
concentrations they play an important signalling estimates high reactive oxygen species production above the
role within the cell and can, for instance, bring about highest permanent human habitation. PLoS One 9, e111068.
Desplanches D, Hoppeler H, Linossier MT, Denis C, Claassen
stabilization of HIF-1α (Guzy & Schumacker, 2006).
H, Dormois D, Lacour JR & Geyssant A (1993). Effects of
Reactive oxygen species-mediated effects, particularly training in normoxia and normobaric hypoxia on human
those that occur via interactions with HIF, are difficult to muscle ultrastructure. Pflugers Arch 425, 263–267.
separate from hypoxia-mediated responses, although the Edwards LM, Murray AJ, Tyler DJ, Kemp GJ, Holloway CJ,
use of antioxidants in studies of hypoxia can help. Indeed, Robbins PA, Neubauer S, Levett D, Montgomery HE,
in contrast to the notion of ROS-mediated suppression of Grocott MP & Clarke K; Caudwell Xtreme Everest Research
mitochondrial function, antioxidant therapy was found to Group (2010). The effect of high-altitude on human skeletal
suppress muscle mitochondrial biogenesis in response to muscle energetics: P-MRS results from the Caudwell Xtreme
training (Gomez-Cabrera et al. 2008). This might suggest Everest expedition. PLoS One 5, e10681.
that transient production of ROS during training (possibly Essop MF, Razeghi P, McLeod C, Young ME, Taegtmeyer H &
as a result of acute hypoxia due to high rates of muscle Sack MN (2004). Hypoxia-induced decrease of UCP3 gene
expression in rat heart parallels metabolic gene switching
O2 consumption) may elicit training-induced changes, in
but fails to affect mitochondrial respiratory coupling.
agreement with a signalling role. Moreover, as outlined Biochem Biophys Res Commun 314, 561–564.
elsewhere in this issue of Experimental Physiology by Prof. Gilbert-Kawai E, Sheperdigian A, Adams T, Mitchell K, Feelisch
Carsten Lundby (Lundby & Jacobs, 2016), the response to M, Murray A, Peters M, Gilbert-Kawai G, Montgomery H,
hypoxia may in fact mediate some aspects of endurance Levett D, Kumar R, Mythen M, Grocott M & Martin D
training in muscle. (2015). Design and conduct of Xtreme Everest 2: an
observational cohort study of Sherpa and lowlander
responses to graduated hypobaric hypoxia. F1000Res 4, 90.
Gomez-Cabrera MC, Domenech E, Romagnoli M, Arduini A,
Conclusions Borras C, Pallardo FV, Sastre J & Viña J (2008). Oral
The duration and degree of hypoxic exposure are critical administration of vitamin C decreases muscle mitochondrial
biogenesis and hampers training-induced adaptations in
to the metabolic response of skeletal muscle, with the
endurance performance. Am J Clin Nutr 87, 142–149.
response to acute hypoxia during exercise or relatively Green HJ, Sutton JR, Cymerman A, Young PM & Houston CS
mild hypoxia at lower altitudes differing from that (1989). Operation Everest II: adaptations in human skeletal
seen with more sustained and severe hypoxia following muscle. J Appl Physiol 66, 2454–2461.
prolonged stays at higher altitudes. In the human heart, Grocott M, Montgomery H & Vercueil A (2007). High-altitude
a loss of energetics is common to both short-term and physiology and pathophysiology: implications and relevance
long-term exposure, although the underlying mechanisms for intensive care medicine. Crit Care 11, 203.


C 2015 The Authors. Experimental Physiology 
C 2015 The Physiological Society
Exp Physiol 101.1 (2016) pp 23–27 Energy metabolism at high altitude 27

Guzy RD & Schumacker PT (2006). Oxygen sensing by Lindholm ME & Rundqvist H (2016). Skeletal muscle
mitochondria at complex III: the paradox of increased hypoxia-inducible factor-1 and exercise. Exp Physiol 101,
reactive oxygen species during hypoxia. Exp Physiol 91, 28–32.
807–819. Lundby C & Jacobs RA (2016). Adaptations of skeletal muscle
Heather LC, Cole MA, Tan JJ, Ambrose LJ, Pope S, Abd-Jamil mitochondria to exercise training. Exp Physiol 101,
AH, Carter EE, Dodd MS, Yeoh KK, Schofield CJ & Clarke K 17–22.
(2012). Metabolic adaptation to chronic hypoxia in cardiac Mason SD, Rundqvist H, Papandreou I, Duh R, McNulty WJ,
mitochondria. Basic Res Cardiol 107, 268. Howlett RA, Olfert IM, Sundberg CJ, Denko NC, Poellinger
Hochachka PW, Clark CM, Holden JE, Stanley C, Ugurbil K & L & Johnson RS (2007). HIF-1α in endurance training:
Menon RS (1996). 31 P magnetic resonance spectroscopy of suppression of oxidative metabolism. Am J Physiol Regul
the Sherpa heart: a phosphocreatine/adenosine triphosphate Integr Comp Physiol 293, R2059–R2069.
signature of metabolic defense against hypobaric hypoxia. Mizuno M, Savard GK, Areskog NH, Lundby C & Saltin B
Proc Natl Acad Sci USA 93, 1215–1220. (2008). Skeletal muscle adaptations to prolonged exposure
Holden JE, Stone CK, Clark CM, Brown WD, Nickles RJ, to extreme altitude: a role of physical activity? High Alt Med
Stanley C & Hochachka PW (1995). Enhanced cardiac Biol 9, 311–317.
metabolism of plasma glucose in high-altitude natives: Murray AJ (2009). Metabolic adaptation of skeletal muscle to
adaptation against chronic hypoxia. J Appl Physiol 79, high altitude hypoxia: how new technologies could resolve
222–228. the controversies. Genome Med 1, 117.
Holloway C, Cochlin L, Codreanu I, Bloch E, Fatemian M, Roberts AC, Butterfield GE, Cymerman A, Reeves JT, Wolfel EE
Szmigielski C, Atherton H, Heather L, Francis J, Neubauer S, & Brooks GA (1996). Acclimatization to 4,300-m altitude
Robbins P, Montgomery H & Clarke K (2011a). Normobaric decreases reliance on fat as a substrate. J Appl Physiol 81,
hypoxia impairs human cardiac energetics. FASEB J 25, 1762–1771.
3130–3135. Simonson TS, Yang Y, Huff CD, Yun H, Qin G, Witherspoon
Holloway CJ, Montgomery HE, Murray AJ, Cochlin LE, DJ, Bai Z, Lorenzo FR, Xing J, Jorde LB, Prchal JT & Ge R
Codreanu I, Hopwood N, Johnson AW, Rider OJ, Levett DZ, (2010). Genetic evidence for high-altitude adaptation in
Tyler DJ, Francis JM, Neubauer S, Grocott MP & Clarke K; Tibet. Science 329, 72–75.
Caudwell Xtreme Everest Research Group (2011b). Cardiac Ward M (1954). High altitude deterioration. Proc R Soc Lond B
response to hypobaric hypoxia: persistent changes in cardiac Biol Sci 143, 40–42.
mass, function, and energy metabolism after a trek to Mt. West JB, Schoene RB & Milledge JS (2007). High Altitude
Everest Base Camp. FASEB J 25, 792–796. Medicine and Physiology. Hodder Arnold, London.
Hoppeler H, Howald H & Cerretelli P (1990). Human muscle
structure after exposure to extreme altitude. Experientia 46,
1185–1187.
Horscroft JA & Murray AJ (2014). Skeletal muscle energy Additional information
metabolism in environmental hypoxia: climbing towards
consensus. Extrem Physiol Med 3, 19. Competing interests
Jacobs RA, Boushel R, Wright-Paradis C, Calbet JA, Robach P,
None declared.
Gnaiger E & Lundby C (2013). Mitochondrial function in
human skeletal muscle following high-altitude exposure. Exp
Physiol 98, 245–255. Funding
Jacobs RA, Siebenmann C, Hug M, Toigo M, Meinild AK &
Lundby C (2012). Twenty-eight days at 3454-m altitude This report received no direct funding.
diminishes respiratory capacity but enhances efficiency in
human skeletal muscle mitochondria. FASEB J 26,
5192–5200. Acknowledgements
Kayser B, Hoppeler H, Claassen H & Cerretelli P (1991).
Muscle structure and performance capacity of Himalayan The author wishes to thank the many colleagues and
Sherpas. J Appl Physiol 70, 1938–1942. collaborators with whom he has had the pleasure of carrying
Kayser B, Hoppeler H, Desplanches D, Marconi C, Broers B & out research in this area, but particularly Tom Ashmore,
Cerretelli P (1996). Muscle ultrastructure and biochemistry Kieran Clarke, Lindsay Edwards, Martin Feelisch, Edward
of lowland Tibetans. J Appl Physiol 81, 419–425. Gilbert-Kawai, Erich Gnaiger, Julian Griffin, Mike Grocott,
Levett DZ, Radford EJ, Menassa DA, Graber EF, Morash AJ, Cameron Holloway, Hans Hoppeler, James Horscroft, Randall
Hoppeler H, Clarke K, Martin DS, Ferguson-Smith AC, Johnson, Aleksandra Kotwica, Verena Laner, Denny Levett,
Montgomery HE, Grocott MP & Murray AJ; Caudwell Daniel Martin and Hugh Montgomery. The author also thanks
Xtreme Everest Research Group (2012). Acclimatization of the British Heart Foundation, Research Councils UK, BBSRC,
skeletal muscle mitochondria to high-altitude hypoxia Action Medical Research, Oroboros Instruments and the WYNG
during an ascent of Everest. FASEB J 26, 1431–1441. Foundation for supporting research in his laboratory.


C 2015 The Authors. Experimental Physiology 
C 2015 The Physiological Society

También podría gustarte