You are on page 1of 10

Acta Physiol 2010, 199, 499–508

REVIEW
Lactate kinetics in human tissues at rest and during exercise

Gerrit van Hall


Metabolic Mass-Spectrometry Facility, Rigshospitalet and Department of Biomedical Sciences, Faculty of Health Sciences, University of
Copenhagen, Copenhagen, Denmark

Received 5 July 2009, Abstract


accepted 7 September 2009 Lactate production in skeletal muscle has now been studied for nearly two
Correspondence: G. van Hall,
centuries and still its production and functional role at rest and during
Metabolic Mass-Spectrometry
Facility, Rigshospitalet, Section exercise is much debated. In the early days skeletal muscle was mainly seen as
7652, 9 Blegdamsvej, DK-2100 the site of lactate production during contraction and lactate production
Copenhagen Ø, Denmark. associated with a lack of muscle oxygenation and fatigue. Later it was rec-
E-mail: gvanhall@cmrc.dk ognized that skeletal muscle not only played an important role in lactate
production but also in lactate clearance and this led to a renewed interest,
not the least from the Copenhagen School in the 1930s, in the metabolic role
of lactate in skeletal muscle. With the introduction of lactate isotopes muscle
lactate kinetics and oxidation could be studied and a simultaneous lactate
uptake and release was observed, not only in muscle but also in other tissues.
Therefore, this review will discuss in vivo human: (1) skeletal muscle lactate
metabolism at rest and during exercise and suggestions are put forward to
explain the simultaneous lactate uptake and release; and (2) lactate metab-
olism in the heart, liver, kidneys, brain, adipose tissue and lungs will be
discussed and its potential importance in these tissues.
Keywords glycogenolysis, glycolysis, intracellular compartmentation,
lactate dehydrogenase, lactate shuttle.

Skeletal muscle lactate metabolism has been studied for lactate is burned up in the oxidative processes in the
nearly two centuries and its production and functional active muscle) until there is no more excess lactate left’
role at rest and in muscle contraction is still a subject of (Bang 1936). This conclusion has magnificently stood
debate (for a historical overview, see Karlsson 1971). the test of time as will be discussed in this review;
The Copenhagen School in the 1930s with Einar however, it seems to be forgotten by many in sport
Lundsgaard and Ole Bang also contributed to the debate physiology. Einar Lundsgaard was more active on the
but not as much as Lindhard and Krogh. However, Ole international scene. In his Harvey lecture in New York
Bang at the Department of Medical Physiology at the 1937 (Lundsgaard 1938) he attacked Meyerhof’s con-
University of Copenhagen with Christian Bohr’s succes- clusion that a constant lactic acid production occurs
sor as mentor, Professor V. Henriques, was closely from the start to finish of exercise and that it was
connected with Krogh and Lindhard and they were both essential for muscle contraction. The constant lactic acid
mentioned in his thesis (Bang 1936). Moreover, Ole production was supposed to lead, in relative few
Bang worked much on the improvement of the lactate minutes, to the attainment of a constant, increased
analysis but the samples originated from Erik Hohwü- lactate level in the active muscle, subsequently main-
Christensen’s experiments. Ole Bang was the first to tained throughout the ‘steady state’ exercise. Lundsg-
notice that blood lactate levels increased with exercise aard argued that lactate was not essential for muscle
but decreased again after 10–20 min of continuous biochemistry; based on his observation that iodo-acetic
exercise and he concluded ‘The removal of lactate during acid poisoning muscle was able to contract. However, he
the initial anaerobic muscular activity is going on during also mentioned that the amount of work performed by
steady state as an accessory process (possibly some of the isolated poisoned muscle was much smaller than that of

Ó 2010 The Author


Journal compilation Ó 2010 Scandinavian Physiological Society, doi: 10.1111/j.1748-1716.2010.02122.x 499
Tissue lactate kinetics Æ G van Hall Acta Physiol 2010, 199, 499–508

a normal one. This review will follow up on this debate lactate levels are increased by lactate infusion (van Hall
as to whether or not lactate is an essential metabolite in et al. 2009b) or exercise (Richter et al. 1988). In fact,
skeletal muscle metabolism with the extension to the the active leg consumes more lactate than the resting leg
relevance of lactate metabolism in other tissues. when arterial lactate levels are increased markedly when
the two-arm ergometer is added to leg exercise (Richter
et al. 1988). The reason that the increase in arterial
Skeletal muscle lactate metabolism at rest
lactate concentration can cause the muscle to switch
and during continuous moderate intensity
from net release to uptake is that muscle unidirectional
exercise
lactate uptake is tightly correlated with arterial con-
Upon the start of moderate intensity exercise blood centration/lactate delivery (van Hall et al. 2003, 2009b)
lactate accumulation increases but after some time, and the lactate taken up is oxidized (van Hall et al.
depending on the intensity, it starts to decrease as 2002, 2003), whereas the unidirectional lactate release
already noticed by Ole Bang in the mid-1930s (Fig. 1a). is related to metabolic rate of the muscle (van Hall et al.
However, if one measures directly across the muscle 2003). How to explain these observations of (1) the
with arterial–venous differences, it can be seen that a high initial high rate of glycogenolysis/glycolysis and
small muscle net lactate release under resting conditions lactate production that immediately decreases with
increases instantaneously many fold where after imme- duration of continuous exercise and (2) the simulta-
diately a steady decrease in net lactate release occurs neous lactate uptake and release by skeletal muscle and
(Fig. 1b) and a similar but quantitatively far lower net that these two processes both increase with muscle
release of pyruvate and alanine, suggesting that during contraction and not just the lactate release.
the initial transition from rest to exercise the muscle Upon the start of exercise muscle adenosine triphos-
exhibits a very high rate of glycogenolysis/glycolysis and phate (ATP) requirement for contraction is instanta-
a release of excessive pyruvate and shunting to lactate neously many-fold increased whereas the increase in
via lactate dehydrogenase and alanine via alanine muscle oxygen utilization is somewhat delayed (Bang-
aminotransferase. However, the picture is more com- sbo et al. 2000, Krustrup et al. 2004, Grassi 2005).
plex as a simultaneous unidirectional lactate uptake and Muscle ATP levels are virtually unchanged, suggesting
release occurs (Jorfeldt 1970, van Hall et al. 2003). The that ATP hydrolysis is matched by re-synthesis via PCr
decrease in net lactate release with exercise duration is (creative phosphate) and glycogenolysis/glycolysis, the
brought about by a decrease in lactate release (produc- latter quantitatively most important. These two fast
tion) with an unchanged lactate uptake (utilization) as processes are crucial to match ATP utilization until
shown for the active forearm when arterial lactate oxidative phosphorylation, energy transport of NADH
concentration is similar over time (Jorfeldt & Wahren (nicotinamide adenine dinucleotide) to and into the
1970), implying that the decrease in net lactate release mitochondria via the malate-aspartate shuttle system as
with exercise duration is mainly caused by decreased the most important one, and adenosine diphosphate
production, not utilization. However, skeletal muscle (ADP) via creatine kinase convection to the mitochon-
has a far more flexible lactate metabolism than can be dria and ATP back to the myofibrils are optimal again
seen during normal continuous arm or leg exercise. The (Hochachka 2003, Hochachka & Beatty 2003, Kaasik
muscle has the ability to switch quickly from a net et al. 2003, Grassi 2005) (Fig. 2). An immediate
lactate producer to a net lactate consumer when arterial adjustment of glycolytic rate is thought to be regulated

Figure 1 Arterial concentration and net leg release of lactate, pyruvate and alanine at rest and during one-leg knee-extensor
exercise at 70% of maximal power output of the knee-extensors (G. van Hall, B. Saltin & F. Dela, unpublished data).

Ó 2010 The Author


500 Journal compilation Ó 2010 Scandinavian Physiological Society, doi: 10.1111/j.1748-1716.2010.02122.x
Acta Physiol 2010, 199, 499–508 G van Hall Æ Tissue lactate kinetics

Figure 2 Scheme for skeletal muscle lactate metabolism. The myocyte, indifferent whether fast or slow twitch fibre, is suggested to
consist of a glycolytic and an oxidative compartment. The glycolytic compartment close to the myofibrils and their glycogen stores
associated with glycogenolysis/glycolysis and lactate release into the circulation. The other oxidative compartment is in close
proximity of the mitochondria. The mitochondria form a sink for both pyruvate and NADH causing low local concentration and
thus lactate utilization via mass action of the equilibrium enzyme lactate dehydrogenase and associated with lactate uptake from the
circulation and subsequent oxidation. Possibly, the subsarcolemmal mitochondria play an important role due to their localization in
close proximities of the arterioles (Hoppeler & Fluck 2003).

at the site of 6-phosphofructo-1-kinase, activated by dehydrogenase reaction. Thus, lactate formation via de
free ADP, AMP and Pi. Moreover, it is mandatory for a equilibrium enzyme lactate dehydrogenase is caused by
high myofibrillar glycogenolytic/glycolytic rate to main- a local mass action of high pyruvate and NADH
tain NAD+ levels for the glyceraldehydes 3-phosphate concentrations; possibly also essential to re-synthesize

Ó 2010 The Author


Journal compilation Ó 2010 Scandinavian Physiological Society, doi: 10.1111/j.1748-1716.2010.02122.x 501
Tissue lactate kinetics Æ G van Hall Acta Physiol 2010, 199, 499–508

NAD+ to maintain a high glycogenolytic rate and as tity of heart type lactate dehydrogenase is virtually the
argued to prevent an acute local low pH as the protons same (for an extensive review, see van Hall 2000).
are utilized in the lactate dehydrogenase reaction Furthermore, the instantaneous switch from a resting or
towards lactate formation (Robergs et al. 2004). When active limb, that maintains the same workload, from a
exercise duration at the same workload continues net large net lactate release to an uptake (Richter et al.
lactate formation ceases as oxidative phosphorylation 1988, van Hall et al. 2003) suggests that lactate
and/or ATP transport from mitochondria to myofibril is dehydrogenase isoform or fibre type recruitment cannot
matched with demand. Lactate formation will decrease be important but the local environmental milieu around
due to lowering of glycogenolytic rate and thus pyru- the lactate dehydrogenase of pyruvate/NADH vs.
vate and NADH concentration. When workload will be lactate/NAD+. Therefore, a simple two-pool model for
increased like in for example an incremental exercise myocellular lactate compartmentation is suggested.
protocol an increased mismatch will occur between the A glycolytic compartment close to the myofibrils and
glycogenolytic and oxidative phosphorylation rate, their glycogen stores associated with glycogenolysis/
hence a higher lactate formation in a linear fashion glycolysis, and lactate release into the circulation. The
(van Hall et al. 2009b). other oxidative compartment is in close proximity of
How to explain the simultaneous lactate uptake and the mitochondria. The mitochondria form a sink for
release by skeletal muscle and the fact that these two both pyruvate and NADH causing low local concen-
processes both increase with muscle contraction? Orig- trations and thus lactate utilization via mass action of
inally, it has been suggested that the slow twitch fibre the equilibrium enzyme lactate dehydrogenase and is
took up lactate from the circulation or lactate produced associated with lactate uptake from the circulation and
by neighbouring fast twitch fibres. This was mainly subsequent oxidation. Observations made in creatine
based on the higher content of heart type lactate kinase-deficient mice underscore the concept of com-
dehydrogenase isoform in slow twitch fibres that was partmentation and intramyocellular diffusion limitation
assumed to promote lactate utilization vs. the higher for energy transfer that makes local high rates of
lactate production capacity of fast twitch fibres with a glycolysis and thus lactate formation mandatory. Major
higher content of the muscle lactate dehydrogenase changes are seen in the glycolytic network and mito-
isoform suggested to promote lactate formation. How- chondrial design when creatine kinase is absent. A large
ever, myocellular compartmentation of both the slow number of mitochondria is found dispersed between the
and fast twitch fibres (Fig. 2) is a more likely explana- myofibrils near the longitudinal sarcoplasmatic reticu-
tion for the simultaneous lactate production and utili- lum and the myosin filaments (de Groof et al. 2001,
zation. Thus, lactate uptake and subsequent oxidation Kaasik et al. 2003, Novotova et al. 2006) reducing the
vs. production takes place in the same cell depending on distance for energy transfer within the myocyte. It
the local intracellular milieu, i.e. myocellular compart- should be emphasized that the glycolytic and oxidative
mentation (Brooks 1991, van Hall 2000) as suggested compartment should not be seen as separated units for
previously for vascular smooth muscle (Lynch & Paul lactate release into and lactate uptake from the circu-
1983) and heart (Peuhkurinen et al. 1983, Schadewaldt lation respectively. The lactate produced in the glyco-
et al. 1983, Bunger 1985, Chatham et al. 2001). In lytic compartment can diffuse to the oxidative
addition, the overall picture of skeletal muscle lactate compartment for oxidation within the cell without
dehydrogenase activity and isoform patterns is that the being released into the circulation, most likely the
heart type lactate dehydrogenase does not necessarily intramyofibrillar mitochondria are important in that
favour pyruvate production and the muscle type lactate process. The subsarcolemmal mitochondria on the other
dehydrogenase lactate production but that all lactate hand may play an important role in the lactate taken up
dehydrogenase isoforms in vivo have the ability to from the circulation and its subsequent oxidation due to
produce or consume lactate depending on the very local their localization in close proximities of arterioles
lactate and pyruvate concentrations and redox state (Hoppeler & Eliich 2003). Of note is that in the
(van Hall 2000, Newsholme 2003). Convincing evi- proposed model for skeletal muscle lactate metabolism
dence is that patients with only the heart type LDH1 there is no role or necessity for mitochondrial lactate
isoform in skeletal muscles do accumulate lactate dehydrogenase and thus mitochondrial lactate oxida-
during ‘semi ischaemic’ exercise and, in fact, in excess tion as suggested by Szczesna-Kaczmarek (1990a,b) and
of their absolute lactate dehydrogenase activity com- pressed by Brooks and co-workers (Brooks et al. 1999,
pared with healthy individuals (Kanno & Maekawa Hashimoto et al. 2006, 2008). To date, the over-
1995, Miyajima et al. 1995a,b). Moreover, the relative whelming theoretical (Sahlin et al. 2002) and practical
quantities of lactate dehydrogenase isoforms may vary evidence is against lactate oxidation by skeletal muscle
considerably between fibre types or in endurance vs. mitochondria (Popinigis et al. 1991, Rasmussen et al.
strength or sprint-trained athletes – the absolute quan- 2002, Sahlin et al. 2002, Ponsot et al. 2005, Yoshida

Ó 2010 The Author


502 Journal compilation Ó 2010 Scandinavian Physiological Society, doi: 10.1111/j.1748-1716.2010.02122.x
Acta Physiol 2010, 199, 499–508 G van Hall Æ Tissue lactate kinetics
et al. 2007) and it should be concluded that it does not intensity exercise (Gertz et al. 1988), which is consid-
occur as summarized by Gladden (2007) and Bonen and erably higher than the contribution of exogenous
co-workers (Bonen et al. 2007, Yoshida et al. 2007). glucose with 8–14% at rest and exercise respectively.
The important role of lactate in heart energy metabo-
lism is underpinned by a recent study showing that
Tissue contribution to systemic lactate
systemic lactate deprivation is detrimental to myocar-
turnover
dial energetic, cardiovascular performance and outcome
in rats (Levy et al. 2007).
Heart
The heart exhibits a net lactate uptake under resting
Brain
conditions; however, similar to skeletal muscle a mutual
unidirectional lactate uptake and release is observed The brain exhibits a net lactate release of about
(Gertz et al. 1981, Wisneski et al. 1985a,b, Neglia et al. 50 lmol min)1 in post-absorptive healthy individuals
2007) (Fig. 3). The lactate taken up by the heart is (van Hall et al. 2009b). Nevertheless, lactate as a fuel
almost entirely oxidized (Wisneski et al. 1985a,b, Gertz for the brain has been debated for many years between
et al. 1988). The unidirectional lactate uptake is highly those who claim that glucose is the sole energy substrate
positively correlated with the arterial lactate concentra- for brain cells and the others who support the concept
tion (Gertz et al. 1988), similar to that seen for skeletal that lactate could be an important fuel for neurones
muscle. However, in contrast to the skeletal muscle, the (Pellerin et al. 2007). The lactate for neuronal oxidation
heart does not seem to increase much its lactate release is suggested to originate mainly from astrocytes, i.e. the
with enhanced metabolic rate during pacing in patients astrocyte-to-neurone lactate shuttle (Pellerin & Magist-
with coronary artery disease (Wisneski et al. 1985a,b) retti 1994). Less emphasis has been on a potential brain
and during exercise in healthy individuals (Gertz et al. lactate uptake from the circulation and subsequent
1981, 1988). As already discussed for skeletal muscle, metabolism despite observation of a net lactate uptake
the explanation of the simultaneous lactate uptake and during exercise when arterial lactate levels are markedly
release by the heart is intracellular compartmentation of increased with insulin-induced hypoglycaemia (Lubow
lactate metabolism. Interestingly, carbohydrate oxida- et al. 2006), cardiopulmonary resuscitation (Rivers
tion by the heart via lactate is higher than via exogenous et al. 1991) and high-intensity exercise (Ide et al.
glucose at rest and during exercise. The contribution of 2000). Moreover, during hypoglycaemia cognitive func-
lactate to oxidative energy production is reported to be tion is improved when lactate is infused similar to
about 10–15% at rest (Gertz et al. 1988, Neglia et al. hyperketonaemia (Maran et al. 1994, Veneman et al.
2007) and increases to about 30% during moderate 1994, King et al. 1997, 1998). Recently, it was shown

Figure 3 Scheme of tissue contributions to systemic lactate turnover in healthy humans under post-absorptive conditions (a) and
during moderate intensity exercise (b). This scheme is compiled from data of different studies and should be considered as
estimates also in view of extrapolations of local skeletal muscle (leg or forearm) and adipose tissue (abdominal wall) to
whole-body skeletal muscle and adipose tissue and of somewhat different exercise intensities, durations and tracer use
(Ahlborg 1974, Gertz et al. 1988, Consoli et al. 1990, van der Merwe et al. 1999, Cersosimo et al. 2000a,b, Mulla et al. 2000,
van Hall et al. 2002, 2003, 2009a,b, Meyer et al. 2002, Woerle et al. 2003, Neglia et al. 2007, Qvisth et al. 2007).

Ó 2010 The Author


Journal compilation Ó 2010 Scandinavian Physiological Society, doi: 10.1111/j.1748-1716.2010.02122.x 503
Tissue lactate kinetics Æ G van Hall Acta Physiol 2010, 199, 499–508

that despite a brain net lactate release a substantial Qvisth et al. 2007). Estimates of net lactate release from
simultaneous unidirectional lactate uptake was observed lactate concentration differences between an artery and
similar to that in the skeletal muscle (van Hall et al. a subcutaneous fat vein on the anterior abdominal wall
2009b). However, in contrast to the skeletal muscle but have reported substantial lower net lactate values with
similar to the heart, virtually all lactate that is taken up 2 lmol kg)1 fat tissue min)1 or 30–40 lmol min)1 for
is oxidized. When lactate levels are elevated due to total body fat mass and an increase during moderate
lactate infusion or exercise the unidirectional lactate intensity exercise (Mulla et al. 2000). Despite the
uptake increases tightly positively correlated with arte- differences in net lactate release, it can be concluded
rial lactate concentration as seen for the heart and the that adipose tissue significantly contributes to systemic
skeletal muscle. Moreover, cerebral energy requirement lactate turnover. Whether adipose tissue also takes up
via lactate could account for about 7% under basal lactate is unknown.
conditions and up to 25% during exercise and lactate
infusion reaching systemic lactate levels of 7 mmol L)1.
Kidney
The brain contributes substantially to systemic lactate
production and utilization with 13% and 8% under Under post-absorptive conditions the kidneys exhibit a
basal conditions respectively. During exercise the active net lactate uptake of approx. 160 lmol min)1 (Ekberg
skeletal muscle is the predominant tissue in lactate et al. 1999, Meyer et al. 2002, Woerle et al. 2003). The
turnover; however, due to the substantial increase in unidirectional lactate uptake can account for 20–30%
cerebral lactate uptake and subsequent oxidation the of systemic lactate clearance. Moreover, a small but
brain relative contribution to lactate clearance increased consistent unidirectional lactate release is found
to 11%. It can be concluded that lactate is an essential accounting for 4–5% of systemic lactate production
part of cerebral energy metabolism either for neurones (Meyer et al. 2002, Woerle et al. 2003). The lactate
or astrocytes. Astrocytes have also been suggested to taken up can account for more than 50% of renal
utilize lactate based on their capacity to oxidize lactate gluconeogenesis and is several fold higher than for the
(Peng et al. 1994, Zielke et al. 2007), brain anatomy other gluconeogenic precursors glutamine, glycerol and
and potential astrocytic compartmentation (van Hall alanine (Meyer et al. 2002). The contribution of renal
et al. 2009b). An astrocyte has three major anatomical gluconeogensis to whole-body glucose appearance and
areas – the soma (cell body), the larger processes thus the contribution of lactate as a gluconeogenic
(including those associated with arterioles and venules precursor shows large variability between studies: 5%
as endfeets), and the very fine surface extensions (Ekberg et al. 1999), 16% (Cersosimo et al. 2000a,b,
lamellae and filopodia, also called peripheral astrocytic Woerle et al. 2003) and up to 40% (Meyer et al. 2002).
processes (PAPs), that are in close apposition with Moreover, the contribution of the kidneys to systemic
neuronal elements (including synapses) (Nedergaard glucose appearance can increase considerably upon
et al. 2003, Hertz et al. 2007). Therefore, in analogy fasting (Ekberg et al. 1999), hypoglycaemia (Cersosimo
with the suggestions for skeletal muscle, subcellular et al. 2000a,b) and adrenaline infusion (Meyer et al.
regions within astrocytes may differ in their spatial and 2003) with markedly increased lactate concentrations.
temporal dependence on the glycolytic and oxidative It can be concluded that the kidney plays a very
pathways. The astrocytic oxidative capabilities reside in important role in net lactate clearance under the post-
the soma and larger cell processes that contain the absorptive resting condition, quantitatively nearly
mitochondria (Wolff & Chao 2004). The threadlike equally important as the liver. Whether the lactate
PAPs cannot accommodate mitochondria that are uptake and release changes during exercise is unknown
approximately twice as wide. However, they may but an increase in lactate uptake may be expected based
respond rapidly to an increase in energy demand from upon the observations that interventions causing an
glycolytic or glycogenolytic-derived ATP because they increase in arterial lactate levels concomitantly find an
have access to glucose and contain glycogen deposits in increase in renal net lactate uptake (Meyer et al. 1999,
contrast to neurones (Hertz et al. 2007). 2003) and in that respect the values for renal lactate
uptake in Figure 3 may be underestimated considering
the higher levels of lactate that can be reached during
Adipose tissue
moderate intensity exercise.
In the post-absorptive state the net lactate release
estimated from arterial and interstitial lactate concen-
Liver
tration differences is reported to range from 4 to
10 lmol kg)1 fat tissue min)1 in the abdominal and Next to the kidneys the liver is the most important
femoral region or 60–150 lmol min)1 if extrapolated tissue in net lactate clearance under resting conditions.
to total body fat mass (van der Merwe et al. 1999, The actual liver lactate uptake and potential release in

Ó 2010 The Author


504 Journal compilation Ó 2010 Scandinavian Physiological Society, doi: 10.1111/j.1748-1716.2010.02122.x
Acta Physiol 2010, 199, 499–508 G van Hall Æ Tissue lactate kinetics
humans is difficult to determine and the estimates to systemic lactate release (Kellum et al. 1997, Walsh
shown in Figure 3 are estimated from measurements et al. 1999), even if one assumes a very much lower
across the splanchnic tissues via arterial and hepatic contribution in healthy individuals. Therefore, the lungs
venous concentration differences. A net lactate uptake may definitely contribute to the lacking net lactate
by the splanchnic bed of 200 lmol min)1 at rest under release under resting conditions. The gut is most likely
the post-absorptive condition has been reported (Ahl- also a substantial net lactate producer, the major
borg 1974, Capaldo et al. 1999) and net lactate uptake substrate being glutamine but glucose is also used as a
increased with continuous moderate intensity exercise substrate. The major fate of glucose is thought not to be
to 300–400 lmol min)1 at 30% VO2max (Ahlborg oxidation but lactate and alanine formation (Mithieux
1974) and 50% VO2max (Ahlborg & Juhlin-Dannfelt 2001, Murphy et al. 2001). However, the liver will
1994) bicycling exercise for 4 and 3 h with very small take up most of this lactate produced by the gut
increases in arterial lactate concentration compared to without entering the main circulation and this is
rest respectively. From these studies it is difficult to why the gut is not represented in the systemic lactate
establish whether increased lactate concentration causes turnover scheme.
an increased splanchnic lactate extraction as the range It is clear that lactate plays an important, not to say
of lactate concentration was small over the entire mandatory, role in human metabolism in spite of its
exercise period. bad rumour from the past as being responsible for
fatigue during exercise or being produced in response
to tissue hypoxia. The importance of lactate for the
Other tissues that may contribute to lactate turnover
liver and the kidneys seems primarily as a carbon
When adding up the major tissues net lactate release to precursor for gluconeogensis. However, lactate is a
– or net uptake from – the circulation it seems that crucial intermediate metabolite in energy turnover in
under post-absorptive resting conditions a substantial the skeletal muscle, heart and brain, and potentially in
net lactate release is lacking. Or to put it in other words, the lungs and gut. Due to intracellular compartmenta-
if tissues clear more lactate (liver, kidney, heart) than tion, and possibly intercellular compartmentation
they add (skeletal muscle, adipose tissue, brain), the between neurones and astrocytes in the brain, energy
arterial lactate concentration would decrease to zero. production from glucose and most importantly glyco-
The reason for this discrepancy may in part originate gen to lactate enables these tissues to respond fast to
from the fact that data from different studies have been local increases in energy demands without limitations
compiled. Furthermore, under resting conditions net in intracellular transport of oxygen, NADH, and
arterial-tissue venous differences can have a consider- pyruvate to the mitochondria and mitochondrially
able variability. Moreover, the extrapolation of arm or produced ATP back to the site of utilization. The great
leg skeletal muscle net lactate release to whole-body advantage of lactate metabolism in these tissues is that
muscle mass and abdominal adipose tissue net lactate the valuable glucose and glycogen is not wasted in the
release to total fat mass may also cause substantial process of glycogenolysis/glycolysis with its fast but
variability. In addition, different fat depots have limited ATP yield but that these tissues can take up
reported to have rather different metabolic activity lactate in large quantities from the circulation and have
and possibly lactate metabolism. Nevertheless, a tissue the ability to be directly oxidized without first being
that has not been discussed but that may quantitatively transformed to glucose via gluconeogensis in the
be very important in net lactate release are the lungs. kidney and liver. All tissues seem to have a large
The lactate fluxes across the lungs are difficult to capacity for lactate transport, both uptake and release.
quantify due to the high blood flow and thereby small In vivo, within the physiological lactate concentration
lactate concentration gradient across the lungs. No net range, the lactate uptake appears dependent on the
exchange, a release and even uptake of lactate by the extra- and intracellular concentration gradient. In that
lungs have been reported, which means that it is respect it has an advantage over glucose that can have
generally assumed that the lungs do not contribute limitation in transport. Also the capacity of lactate
significantly to systemic lactate turnover (Mitchell & dehydrogenase activity in the direction of pyruvate
Cournand 1954). However, most studies are performed formation does not seem limited in view of the high
on a variety of patients. Generally, it seems that patients capacity for lactate oxidation. Therefore, lactate has
with low systemic lactate concentrations exhibit a very properties that make it worthwhile to be used as an
much lower net lactate release compared with patients energy source under certain conditions possibly pref-
with high systemic lactate levels. Nevertheless, the erably over glucose. Thus, after more than 150 years of
reported net lactate release of 80–300 lmol min)1 in lactate research the area is very much alive and in the
patients with near-normal arterial lactate levels would last decades a renewed interest has been witnessed, less
imply that the lungs can make a substantial contribution in the classical field of (exercise) physiology as in the

Ó 2010 The Author


Journal compilation Ó 2010 Scandinavian Physiological Society, doi: 10.1111/j.1748-1716.2010.02122.x 505
Tissue lactate kinetics Æ G van Hall Acta Physiol 2010, 199, 499–508

years of the Copenhagen School but more in medical lactate release during net chemical extraction in man. Cir-
research. culation 63, 1273–1279.
Gertz, E., Wisneski, J., Stanley, W. & Neese, R. 1988. Myo-
cardial substrate utilization during exercise in humans. Dual
Conflict of interest carbon-labeled carbohydrate isotope experiments. J Clin
Invest 82, 2017–2025.
There is no conflict of interest.
Gladden, L.B. 2007. Is there an intracellular lactate shuttle in
skeletal muscle? J Physiol 582, 899.
References Grassi, B. 2005. Delayed metabolic activation of oxidative
phosphorylation in skeletal muscle at exercise onset. Med Sci
Ahlborg, G. 1974. Substrate turnover during prolonged exer- Sports Exerc 37, 1567–1573.
cise in man splanchnic and leg metabolism of glucose, free de Groof, A., Oerlemans, F., Jost, C. & Wieringa, B. 2001.
fatty acids, and amino acids. J Clin Invest 53, 1080–1090. Changes in glycolytic network and mitochondrial design in
Ahlborg, G. & Juhlin-Dannfelt, A. 1994. Effect of beta-receptor creatine kinase-deficient muscles. Muscle Nerve 24, 1188–
blockade on splanchnic and muscle metabolism during 1196.
prolonged exercise in men. J Appl Physiol 76, 1037–1042. van Hall, G. 2000. Lactate as a fuel for mitochondrial respi-
Bang, O. 1936. The lactate content of blood during and after ration. Acta Physiol Scand 168, 643–656.
muscular exercise in man. Skand Arch Physiol 74, 5182. van Hall, G., Calbet, J.A.L., Sondergaard, H. & Saltin, B.
Bangsbo, J., Krustrup, P., Gonzalez-Alonso, J., Boushel, R. & 2002. Similar carbohydrate but enhanced lactate utilization
Saltin, B. 2000. Muscle oxygen kinetics at onset of intense during exercise after 9 wk of acclimatization to 5,620 m.
dynamic exercise in humans. Am J Physiol Regul Integr Am J Physiol Endocrinol Metab 283, E1203–E1213.
Comp Physiol 279, R899–R906. van Hall, G., Jensen-Urstad, M., Rosdahl, H., Holmberg, H.-C.,
Bonen, A., Hatta, H., Holloway, G., Spriet, L. & Yoshida, Y. Saltin, B. & Calbet, J. 2003. Leg and arm lactate and
2007. Reply from Arend Bonen, Hideo Hatta, Graham P. substrate kinetics during exercise. Am J Physiol Endocrinol
Holloway, Lawrence L. Spriet and Yuko Yoshida. J Physiol Metab 284, E193–E205.
584, 707–708. van Hall, G., Lundby, C., Araoz, M., Calbet, J.A.L., Sander,
Brooks, G.A. 1991. Current concepts in lactate exchange. Med M. & Saltin, B. 2009a. The lactate paradox revisited in
Sci Sports Exerc 23, 895–906. lowlanders during acclimatization to 4100 m and in high-
Brooks, G.A., Dubouchaud, H., Brown, M., Sicurello, J.P. & altitude natives. J Physiol 587, 1117–1129.
Butz, C.E. 1999. Role of mitochondrial lactate dehydroge- van Hall, G., Stromstad, M., Rasmussen, P., Jans, O., Zaar,
nase and lactate oxidation in the intracellular lactate shuttle. M., Gam, C., Quistorff, B., Secher, N.H. & Nielsen, H.B.
Proc Natl Acad Sci USA 96, 1129–1134. 2009b. Blood lactate is an important energy source for
Bunger, R. 1985. Compartmented pyruvate in perfused work- the human brain. J Cereb Blood Flow Metab 29, 1121–1129.
ing heart. Am J Physiol Heart Circ Physiol 249, H439– Hashimoto, T., Hussien, R. & Brooks, G.A. 2006. Co-locali-
H449. zation of MCT1, CD147, and LDH in mitochondrial inner
Capaldo, B., Gastaldelli, A., Antoniello, S., Auletta, M., Pardo, membrane of L6 muscle cells: evidence of a mitochondrial
F., Ciociaro, D., Guida, R., Ferrannini, E. & Sacca, L. 1999. lactate oxidation complex. Am J Physiol Endocrinol Metab
Splanchnic and leg substrate exchange after ingestion of a 290, E1237–E1244.
natural mixed meal in humans. Diabetes 48, 958–966. Hashimoto, T., Hussien, R., Cho, H.-S., Kaufer, D. &
Cersosimo, E., Garlick, P. & Ferretti, J. 2000a. Regulation of Brooks, G.A. 2008. Evidence for the mitochondrial lactate
splanchnic and renal substrate supply by insulin in humans. oxidation complex in rat neurons: demonstration of an
Metabolism 49, 676–683. essential component of brain lactate shuttles. PLoS ONE 3,
Cersosimo, E., Garlick, P. & Ferretti, J. 2000b. Renal substrate e2915.
metabolism and gluconeogenesis during hypoglycemia in Hertz, L., Peng, L. & Dienel, G.A. 2007. Energy metabolism in
humans. Diabetes 49, 1186–1193. astrocytes: high rate of oxidative metabolism and spatio-
Chatham, J.C., Des Rosiers, C. & Forder, J.R. 2001. Evidence temporal dependence on glycolysis/glycogenolysis. J Cereb
of separate pathways for lactate uptake and release by the Blood Flow Metab 27, 219–249.
perfused rat heart. Am J Physiol Endocrinol Metab 281, Hochachka, P.W. 2003. Intracellular convection, homeostasis
E794–E802. and metabolic regulation. J Exp Biol 206, 2001–2009.
Consoli, A., Nurjhan, N., Reilly, J.J., Jr, Bier, D.M. & Gerich, Hochachka, P.W. & Beatty, C.L. 2003. Patterns of control of
J.E. 1990. Contribution of liver and skeletal muscle to ala- maximum metabolic rate in humans. Comp Biochem Physiol
nine and lactate metabolism in humans. Am J Physiol A Mol Integr Physiol 136, 215–225.
Endocrinol Metab 259, E677–E684. Hoppeler, H. & Fluck, M. 2003. Plasticity of skeletal muscle
Ekberg, K., Landau, B.R., Wajngot, A., Chandramouli, V., mitochondria: structure and function. Med Sci Sports Exerc
Efendic, S., Brunengraber, H. & Wahren, J. 1999. Contri- 35, 95–104.
butions by kidney and liver to glucose production in the Ide, K., Schmalbruch, I.K., Quistorff, B., Horn, A. & Secher,
postabsorptive state and after 60 h of fasting. Diabetes 48, N.H. 2000. Lactate, glucose and O2 uptake in human brain
292–298. during recovery from maximal exercise. J Physiol 522, 159–
Gertz, E., Wisneski, J., Neese, R., Bristow, J., Searle, G. & 164.
Hanlon, J. 1981. Myocardial lactate metabolism: evidence of

Ó 2010 The Author


506 Journal compilation Ó 2010 Scandinavian Physiological Society, doi: 10.1111/j.1748-1716.2010.02122.x
Acta Physiol 2010, 199, 499–508 G van Hall Æ Tissue lactate kinetics
Jorfeldt, L. 1970. Metabolism L (+)-Lactate in human skeletal Meyer, C., Stumvoll, M., Dostou, J., Welle, S., Haymond, M.
muscle during exercise. Acta Physiol Scand 338(Suppl.), & Gerich, J. 2002. Renal substrate exchange and gluco-
1–67. neogenesis in normal postabsorptive humans. Am J Physiol
Jorfeldt, L. & Wahren, J. 1970. Human forearm muscle Endocrinol Metab 282, E428–E434.
metabolism during exercise. V. Quantitative aspects of Meyer, C., Stumvoll, M., Welle, S., Woerle, H.J., Haymond,
glucose uptake and lactate production during prolonged M. & Gerich, J. 2003. Relative importance of liver, kidney,
exercise. Scand J Clin Lab Invest 26, 73–81. and substrates in epinephrine-induced increased gluconeo-
Kaasik, A., Veksler, V., Boehm, E., Novotova, M. & Ventura- genesis in humans. Am J Physiol Endocrinol Metab 285,
Clapier, R. 2003. From energy store to energy flux: a study E819–E826.
in creatine kinase deficient fast skeletal muscle. FASEB J 17, Mitchell, A. & Cournand, A. 1954. The fate of circulating
708–710. lactic acid in the human lung. J Clin Invest 34, 471–476.
Kanno, T. & Maekawa, M. 1995. Lactate dehydrogenase Mithieux, G. 2001. New data and concepts on glutamine and
M-subunit deficiencies: clinical features, metabolic back- glucose metabolism in the gut. Curr Opin Clin Nutr Metab
ground, and genetic heterogeneities. Muscle Nerve 3(Suppl.), Care 4, 267–271.
S54–S60. Miyajima, H., Takahashi, Y. & Kaneko, E. 1995a. Charac-
Karlsson, J. 1971. Lactate and phosphagen concentrations in terization of the glycolysis in lactate dehydrogenase-A defi-
working muscle of man. Acta Physiol Scand 358(Suppl.), ciency. Muscle Nerve 18, 874–878.
1–72. Miyajima, H., Takahashi, Y. & Kaneko, E. 1995b. Charac-
Kellum, J.A., Kramer, D.J., Lee, K., Mankad, S., Bellomo, R. terization of the oxidative metabolism in lactate dehydro-
& Pinsky, M.R. 1997. Release of lactate by the lung in acute genase A deficiency. Int Med 34, 502–506.
lung injury. Chest 111, 1301–1305. Mulla, N., Simonsen, L. & Bülow, J. 2000. Post-exercise
King, P., Parkin, H., Macdonald, I., Barber, C. & Tattersall, R. adipose tissue and skeletal muscle lipid metabolism in
1997. The effect of intravenous lactate on cerebral function humans: the effects of exercise intensity. J Physiol 524,
during hypoglycaemia. Diabet Med 14, 19–28. 919–928.
King, P., Kong, M., Parkin, H., MacDonald, I., Barber, C. & Murphy, N., Kodakat, S., Wendon, J., Jooste, C., Muiesan, P.,
Tattersall, R. 1998. Intravenous lactate prevents cerebral Rela, M. & Heaton, N. 2001. Liver and intestinal lactate
dysfunction during hypoglycaemia in insulin-dependent metabolism in patients with acute hepatic failure undergoing
diabetes mellitus. Clin Sci 94, 157–163. liver transplantation. Crit Care Med 29, 2111–2118.
Krustrup, P., Hellsten, Y. & Bangsbo, J. 2004. Intense interval Nedergaard, M., Ransom, B. & Goldman, S. 2003. New roles
training enhances human skeletal muscle oxygen uptake in for astrocytes: redefining the functional architecture of the
the initial phase of dynamic exercise at high but not at low brain. Trends Neurosci 26, 523–530.
intensities. J Physiol 559, 335–345. Neglia, D., De Caterina, A., Marraccini, P., Natali, A.,
Levy, B., Mansart, A., Montemont, C., Gibot, S., Mallie, J.-P., Ciardetti, M., Vecoli, C., Gastaldelli, A., Ciociaro, D.,
Regnault, V., Lecompte, T. & Lacolley, P. 2007. Myocardial Pellegrini, P., Testa, R., Menichetti, L., L’Abbate, A.,
lactate deprivation is associated with decreased cardiovascu- Stanley, W.C. & Recchia, F.A. 2007. Impaired myocardial
lar performance, decreased myocardial energetics, and early metabolic reserve and substrate selection flexibility during
death in endotoxic shock. Intensive Care Med 33, 495–502. stress in patients with idiopathic dilated cardiomyopathy.
Lubow, J.M., Pinon, I.G., Avogaro, A., Cobelli, C., Treeson, Am J Physiol Heart Circ Physiol 293, H3270–H3278.
D.M., Mandeville, K.A., Toffolo, G. & Boyle, P.J. 2006. Newsholme, E. 2003. Enzymes, energy and endurance. In:
Brain oxygen utilization is unchanged by hypoglycemia in J. Poortmans (ed.) Principles of Exercise Biochemistry, pp.
normal humans: lactate, alanine, and leucine uptake are not 1–35. Karger, Basel.
sufficient to offset energy deficit. Am J Physiol Endocrinol Novotova, M., Pavlovicova, M., Veksler, V., Ventura-Clapier,
Metab 290, E149–E153. R. & Zahradnik, I. 2006. Ultrastructural remodeling of fast
Lundsgaard, E. 1938. The Pasteur–Meyerhof reaction in skel- skeletal muscle fibers induced by invalidation of creatine
etal muscle. Bull N Y Acad Med 14, 163–182. kinase. Am J Physiol Cell Physiol 291, C1279–C1285.
Lynch, R. & Paul, R. 1983. Compartmentation of glycolytic Pellerin, L. & Magistretti, P. 1994. Glutamate uptake into
and glycogenolytic metabolism in vascular smooth muscle. astrocytes stimulates aerobic glycolysis: a mechanism cou-
Science 222, 1344–1346. pling neuronal activity to glucose utilization. Proc Natl Acad
Maran, A., Cranston, I., Lomas, J., Macdonald, I. & Amiel, Sci USA 91, 10625–10629.
S.A. 1994. Protection by lactate of cerebral function during Pellerin, L., Bouzier- Sore, A.-K., Aubert, A., Serres, S., Merle,
hyperglycaemia. Lancet 343, 16–20. M., Costalat, R. & Magistretti, P. 2007. Activity-dependent
van der Merwe, M.-T., Jansson, P.-A., Crowther, N.J., Boyd, regulation of energy metabolism by astrocytes: an update.
I.H., Gray, I.P., Joffe, B.I. & Lonnroth, P.N. 1999. Lactate Glia 55, 1251–1262.
and glycerol release from subcutaneous adipose tissue in Peng, L., Zhang, X. & Hertz, L. 1994. High extracellular
black and white lean men. J Clin Endocrinol Metab 84, potassium concentrations stimulate oxidative metabolism in
2888–2895. a glutamatergic neuronal culture and glycolysis in cultured
Meyer, C., Dostou, J.M. & Gerich, J.E. 1999. Role of the astrocytes but have no stimulatory effect in a GABAergic
human kidney in glucose counterregulation. Diabetes 48, neuronal culture. Brain Res 663, 168–172.
943–948.

Ó 2010 The Author


Journal compilation Ó 2010 Scandinavian Physiological Society, doi: 10.1111/j.1748-1716.2010.02122.x 507
Tissue lactate kinetics Æ G van Hall Acta Physiol 2010, 199, 499–508

Peuhkurinen, K., Hiltunen, J. & Hassinen, I. 1983. Metabolic R. Terjung, H. Kaciuba-U’scilko, L. Budohoski (eds) Inter-
compartmentation of pyruvate in the isolated perfused rat national Perspectives in Exercise Physiology, pp. 130–131.
heart. Biochem J 210, 193–198. Human Kinetics Books, Champaign, IL.
Ponsot, E., Zoll, J., N’Guessan, B., Ribera, F., Lampert, E., Szczesna-Kaczmarek, A. 1990b. L-lactate oxidation by skeletal
Richard, R., Veksler, V., Ventura-Clapier, R. & Mettauer, B. muscle mitochondria. Int J Biochem 22, 617–620.
2005. Mitochondrial tissue specificity of substrates Veneman, T., Mitrakou, A., Mokan, M., Cryer, P. & Gerich, J.
utilization in rat cardiac and skeletal muscles. J Cell Physiol 1994. Effect of hyperketonemia and hyperlacticacidemia on
203, 479–486. symptoms, cognitive dysfunction, and counter regulatory
Popinigis, J., Antosiewicz, J., Crimi, M., Lenaz, G. & Waka- hormone responses during hypoglycemia in normal humans.
bayashi, T. 1991. Human skeletal muscle: participation of Diabetes 43, 1311–1317.
different metabolic activities in oxidation of L-lactate. Acta Walsh, T.S., McLellan, S., Mackenzie, S.J. & Lee, A. 1999.
Biochim Pol 38, 169–175. Hyperlactatemia and pulmonary lactate production in
Qvisth, V., Hagstrom-Toft, E., Moberg, E., Sjoberg, S. & patients with fulminant hepatic failure. Chest 116, 471–
Bolinder, J. 2007. Lactate release from adipose tissue and 476.
skeletal muscle in vivo: defective insulin regulation in insu- Wisneski, J., Gertz, E., Neese, R., Gruenke, L. & Craig, J.
lin-resistant obese women. Am J Physiol Endocrinol Metab 1985a. Dual carbon-labeled isotope experiments using
292, E709–E714. D-[6-14C] glucose and L-[1,2,3-13C3] lactate: a new
Rasmussen, H., van Hall, G. & Rasmussen, U. 2002. Lactate approach for investigating human myocardial metabolism
dehydrogenase is not a mitochondrial enzyme in human and during ischemia. J Am Coll Cardiol 5, 1138–1346.
mouse vastus lateralis muscle. J Physiol 541, 575–580. Wisneski, J., Gertz, E., Neese, R., Gruenke, L., Morris, D. &
Richter, E.A., Kiens, B., Saltin, B., Christensen, N.J. & Savard, Craig, J. 1985b. Metabolic fate of extracted glucose in
G. 1988. Skeletal muscle glucose uptake during dynamic normal human myocardium. J Clin Invest 76, 819–1827.
exercise in humans: role of muscle mass. Am J Physiol Woerle, H.J., Meyer, C., Popa, E.M., Cryer, P.E. & Gerich,
Endocrinol Metab 254, E555–E561. J.E. 2003. Renal compensation for impaired hepatic glucose
Rivers, E., Paradis, N., Martin, G., Goetting, M., Rosenberg, release during hypoglycemia in type 2 diabetes. Diabetes 52,
J., Smithline, H., Appleton, T. & Nowak, R. 1991. Cerebral 1386–1392.
lactate uptake during cardiopulmonary resuscitation in hu- Wolff, J. & Chao, T. 2004. Architectonics of non-neural cells
mans. J Cereb Blood Flow Metab 11, 479–484. of the nervous system. In: L. Hertz (ed.) Non-Neuronal Cell
Robergs, R.A., Ghiasvand, F. & Parker, D. 2004. Biochemistry of the Nervous System: Function and Dysfunction, pp. 1–52.
of exercise-induced metabolic acidosis. Am J Physiol Regul Elsevier, Amsterdam.
Integr Comp Physiol 287, R502–R516. Yoshida, Y., Holloway, G.P., Ljubicic, V., Hatta, H., Spriet,
Sahlin, K., Fernström, M., Svensson, M. & Tonkonogi, M. L.L., Hood, D.A. & Bonen, A. 2007. Negligible direct lac-
2002. No evidence of an intracellular lactate shuttle in rat tate oxidation in subsarcolemmal and intermyofibrillar
skeletal muscle. J Physiol 541, 569–574. mitochondria obtained from red and white rat skeletal
Schadewaldt, P., Münch, U. & Staib, W. 1983. Evidence for muscle. J Physiol 582, 1317–1335.
the compartmentation of pyruvate metabolism in perfused Zielke, H., Zielke, C., Baab, P. & Tildon, J. 2007. Effect of
rat skeletal muscle. Biochem J 216, 761–764. fluorocitrate on cerebral oxidation of lactate and glucose in
Szczesna-Kaczmarek, A. 1990a. Direct oxidation of L-lactate freely moving rats. J Neurochem 101, 9–16.
by mitochondria isolated from different tissues. In: K. Nazar,

Ó 2010 The Author


508 Journal compilation Ó 2010 Scandinavian Physiological Society, doi: 10.1111/j.1748-1716.2010.02122.x

You might also like