2020 - Lactate The Ugly Duckling of Energy Metabolism

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

Perspective

https://doi.org/10.1038/s42255-020-0243-4

Lactate: the ugly duckling of energy metabolism


Joshua D. Rabinowitz   1 ✉ and Sven Enerbäck   2 ✉

Lactate, perhaps the best-known metabolic waste product, was first isolated from sour milk, in which it is produced by lactoba-
cilli. Whereas microbes also generate other fermentation products, such as ethanol or acetone, lactate dominates in mammals.
Lactate production increases when the demand for ATP and oxygen exceeds supply, as occurs during intense exercise and isch-
aemia. The build-up of lactate in stressed muscle and ischaemic tissues has established lactate’s reputation as a deleterious
waste product. In this Perspective, we summarize emerging evidence that, in mammals, lactate also serves as a major circu-
lating carbohydrate fuel. By providing mammalian cells with both a convenient source and sink for three-carbon compounds,
circulating lactate enables the uncoupling of carbohydrate-driven mitochondrial energy generation from glycolysis. Lactate
and pyruvate together serve as a circulating redox buffer that equilibrates the NADH/NAD ratio across cells and tissues. This
reconceptualization of lactate as a fuel—analogous to how Hans Christian Andersen’s ugly duckling is actually a beautiful
swan—has the potential to reshape the field of energy metabolism.

C
arbohydrates account for approximately half the caloric yielding valuable energy. When oxygen is not available, however,
intake in humans. Most are eaten as starch, which is broken mitochondria can no longer effectively clear electrons. Thus, under
down into glucose in the small intestinal lumen. Glucose is anaerobic conditions, fermentation is the only metabolic option.
then absorbed into the portal circulation and passed to the liver. Even when oxygen is available, ATP production by oxidative phos-
The liver takes up a portion of dietary glucose and stores it as gly- phorylation can be limited by the oxygen uptake rate. Thus, under
cogen for future release during fasting. The remainder is distributed high-demand conditions, such as intense exercise, fermentation
throughout the body for use as fuel. Some of this glucose is con- provides a way to accelerate energy generation. Accordingly, when
verted into lactate, and glucose and lactate are the two most abun- muscle is pushed beyond the aerobic threshold, such as during a
dant circulating carbon carriers in mammals. short sprint or toward the end of a long hard run, lactate is released
as metabolic waste5 (Fig. 1b).
The classic view: glucose as fuel, lactate as waste
Energy can be derived from glucose through two processes: fermen- Lactate excretion by cultured cells
tation and respiration. Both begin with catabolism of glucose via Lactate production does not occur in only oxygen-limited muscle.
glycolysis into two molecules of pyruvate, with associated produc- Most cultured mammalian cells produce copious lactate even when
tion of two ATP and two NADH molecules. The NADH is made at oxygen is abundant. Such aerobic glycolysis was initially noted by
the glyceraldehyde-3-phosphate dehydrogenase (GAPDH) step in Warburg nearly a century ago, and rapid lactate production was
the middle of glycolysis. During fermentation, the NADH is used to the first molecular phenotype associated with cancer6. This obser-
reduce pyruvate to lactate, which is then excreted. This process results vation led to the hypothesis that cancer is a disease of defective
in a net yield of two ATP and two lactate molecules per glucose, with- mitochondria7. But although mitochondrial defects occasionally
out consuming any oxygen. In respiration, the NADH electrons and occur in cancer, they are not a major cause8–10. Instead, fermenta-
pyruvate generated by glycolysis are shuttled into the mitochondria, tion is a common feature of mammalian cells in culture, includ-
where they are consumed and subsequently produce copious usable ing non-cancerous cells, such as proliferating T cells and even
energy (approximately 25 ATP molecules per glucose). non-growing cells, such as quiescent fibroblasts11,12.
The ability to ferment glucose into lactate reflects that two lac- The reasons why cultured cells avidly ferment glucose remain
tic acid molecules contain the same atoms as one glucose molecule incompletely understood, but media formulation plays a role.
(Fig. 1a). Thus, although the chemical bonds are rearranged, lactic Classical tissue medium contains serum growth factors, supraphysi-
acid is half of glucose. ologic glucose and no lactate13. In such conditions, glucose is the
Pyruvate, in contrast, is more oxidized than either glucose or only available carbohydrate fuel. Moreover, both glucose availabil-
lactate. Specifically, each lactate molecule carries two more hydro- ity and lactate secretion are unconstrained, and glucose is refreshed
gen atoms than pyruvate. These two hydrogen atoms consist of and lactate is removed every time the culture medium is changed.
two protons and two electrons. To convert either glucose or lactate These factors, together with growth-factor signals that trigger glu-
into pyruvate, these electrons must be disposed of, in a process that cose uptake14, favour avid glucose fermentation. The release of lac-
requires transporting the electrons, which are stored in NADH, into tate by both cells in vitro and stressed muscle in vivo supports the
mitochondria. Although direct lactate import into the mitochon- paradigm of glucose as fuel and lactate as waste.
drial matrix is a possibility1, there are two better-established routes
to transport the electrons: the malate–aspartate and glycerol phos- An emerging possibility: glucose as specific fuel, lactate as
phate shuttles2–4 (Fig. 1b). universal fuel
When oxygen is available, NADH’s electrons can be quickly Mammals evolved under different metabolic pressures from cells in
used by the electron-transport chain in the mitochondria, thereby culture, including a frequent risk of starvation. Because lactate is

Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA. 2Department of Medical Biochemistry and Cell Biology, Institute
1

of Biomedicine, University of Gothenburg, Gothenburg, Sweden. ✉e-mail: joshr@princeton.edu; sven.enerback@medgen.gu.se

566 Nature Metabolism | VOL 2 | July 2020 | 566–571 | www.nature.com/natmetab


NATure MeTABoLIsm Perspective
a b c
Lactate-producing cell Lactate-consuming cell
O H
Glucose Glucose
H OH

HO H
O O ×2 ×2
H OH
H3C H3C GAP Malate– GAP Malate–
H OH OH aspartate aspartate
OH GAPDH shuttle GAPDH shuttle
CH2OH OH O Glycerol Glycerol
Glucose 2 Lactic acids 2 Pyruvic acids NADH phosphate NADH phosphate
shuttle shuttle
C, O 6 6 6
H 12 12 8 Pyruvate Lactate Pyruvate Lactate
LDH LDH

Fig. 1 | Lactate—waste and fuel. a, Lactate is half of glucose, whereas pyruvate is more oxidized. b, Lactate as waste. Glycolytic flux from glucose to
pyruvate generates NADH from NAD at the GAPDH reaction. NADH’s electrons can be transported into mitochondria via the malate–aspartate or
glycerol phosphate shuttles, regenerating cytosolic NAD. Alternatively, NADH can be used by LDH to reduce pyruvate to lactate, which is secreted as
waste. c, Lactate as fuel. The reactions are the same, but the direction of LDH flux is reversed. The resulting extra NADH electrons are transported into
mitochondria. GAP, glyceraldehyde 3-phosphate.

energy rich, mammals do not excrete it. Indeed, CO2 is the only equivalent on a carbon-atom basis (because two lactates are equal to
carbonaceous waste that we excrete in substantial quantities. The one glucose). The straightforward interpretation of these measure-
full oxidation of dietary carbon to CO2 maximizes the extraction of ments is that pyruvate generated by glycolysis rarely flows within a
usable energy from food. How is this achieved? cell directly into the tricarboxylic acid (TCA) cycle but instead is
The two most abundant circulating carbon metabolites are glu- converted to lactate and released into the bloodstream. This process
cose (5 mM) and lactate (1 mM). Glucose and lactate can be inter- requires lactate dehydrogenase (LDH) and monocarboxylic trans-
converted by the processes of glycolysis and gluconeogenesis. One porter (MCT) activity.
logical way to coordinate these processes is as follows: (1) most cells Rapid production of circulating lactate must be offset by compa-
extract energy from carbohydrate by taking up glucose and fully rably rapid consumption (Fig. 1c). Recent work has examined the
oxidizing it to CO2; (2) cells facing particularly urgent energy needs fate of infused 13C-labelled lactate, finding that it is a major fuel in
take up extra glucose and release some lactate as waste—in addition, the TCA cycle. Specifically, [13C]lactate labels TCA intermediates in
as shown by Gerty and Cori, fasting muscle releases glycogen stores every tissue of the body, even in tumors22,23. With the exception of
as lactate15; and (3) the liver ‘cleans up’ this lactate, reconverting it the brain, tissue TCA labelling from lactate is typically greater than
to glucose. In this scenario, lactate is valuable only as a substrate for that from infused [13C]glucose.
generating glucose. A challenge in interpreting these isotope-tracer studies is that
The above strategy yields clear expectations regarding mamma- lactate can be converted into glucose, and glucose can be converted
lian metabolic flux: tissue glucose consumption should far exceed into lactate. Thus, infusion of either metabolite also labels the
lactate consumption, and the whole-body lactate production rate other. The relative contributions of circulating glucose and lactate
should approximately equal the lactate use by the liver and kidneys to tissue TCA-cycle metabolism can be deconvoluted mathemati-
to support gluconeogenesis. cally. Such analysis has shown that, in agreement with textbook
The relevant metabolic fluxes can be measured from two per- knowledge, the brain directly oxidizes glucose as its primary fuel.
spectives: arterial–venous differences in metabolite concentration Especially in the fasted state, however, the main route through
and isotope tracing. Differences in arterial–venous metabolite con- which glucose labels the TCA cycle in the other major organs is via
centrations reveal the net production or consumption of a metabo- circulating lactate22.
lite across a vascular bed. In some cases, such as the renal artery and How can these observations be reconciled with the arterial–
vein, the vascular bed aligns relatively neatly with a single organ venous measurements, which show modest net lactate consumption
(the kidney). In other cases, such as the femoral artery and vein, the or production across most vascular beds? One possibility is that tis-
vascular bed drains multiple tissue types (skin, adipose, bone and sue pyruvate is rapidly exchanged for circulating lactate, but with
various types of muscle) whose activities may offset one another. only modest net flux of three-carbon (3C) units. This possibility is
With this caveat, such measurements generally conform to the biochemically reasonable, because MCTs exchange one carboxylic
above expectations regarding metabolic flux16,17. acid for another more rapidly than they catalyse net excretion or
Isotope-tracing measurements, however, paint a different pic- uptake24
ture. At pseudo–steady state, the total flux of a metabolite from The more radical possibility is that, at the cellular level, glucose
tissues into the bloodstream is balanced with the total tissue con- uptake may be dissociated from carbohydrate burning, and lactate
sumption of the metabolite, thereby defining the metabolite’s ‘cir- may serve as a universal carbohydrate fuel.
culatory turnover flux’. For non-perturbative isotope infusions,
circulatory turnover flux is synonymous with the ‘rate of appear- Uncoupling of glycolysis and TCA
ance’, ‘rate of disappearance’ and, in the fasted state, the ‘endogenous In the absence of lactate, glycolysis must operate in lockstep with
production rate’. It can be measured by infusing an isotope-labelled the TCA cycle, such that every glycolysis-produced NADH and
metabolite and monitoring its dilution by endogenous production pyruvate is cleared by mitochondrial metabolism. Correspondingly,
and the diet. Such measurements have commonly been made since cells would need to break down glucose via glycolysis to generate
the 1970s (refs. 18–21). The fasted circulatory turnover flux of lactate energy from carbohydrate. Lactate’s fundamental role is to uncouple
has consistently been shown, in both rodents and humans, to be these pathways25 (Fig. 2). Interestingly, pancreatic alpha and beta
approximately twice that of glucose on a molar basis, and thus is cells, whose function is to secrete glucagon and insulin in response

Nature Metabolism | VOL 2 | July 2020 | 566–571 | www.nature.com/natmetab 567


Perspective NATure MeTABoLIsm

a b
Traditional Redox buffering

Pyruvate
Glucose Lactate Glucose Lactate MCT
GLUT MCT GLUT MCT

Glucose Glucose
Lactate Lactate
NAD NAD

LDH LDH

NADH NADH
Pyruvate Pyruvate

TCA TCA

Fig. 2 | Redox buffering by circulating lactate and pyruvate. a, Traditional perspective. Glycolytic flux exceeds pyruvate and lactate exchange between
the cell and the circulation. Intracellular metabolic reactions (glycolytic flux and transport of NADH’s electrons into mitochondria) determine the cellular
NADH/NAD ratio and thereby the cellular lactate/pyruvate ratio. b, Redox buffering perspective. Exchange in pyruvate and lactate between cells and the
circulation is more rapid than glycolysis. These MCT-catalysed exchange reactions determine the cellular lactate/pyruvate ratio and thereby the cellular
NADH/NAD ratio. In cases in which excessive NADH begins to build up, net uptake of pyruvate and excretion of lactate alleviates the redox imbalance.
GLUT, glucose transporter.

to circulating glucose levels, lack MCTs and therefore have tight gly- In contrast, lactate rapidly exchanges throughout the body. This
colysis–TCA coupling26,27. Most mammalian cells, however, express advantagenously tends to minimize local lactate build-up. When
both LDH and MCTs and therefore can independently run glycoly- lactate does accumulate, this accumulation often reflects underlying
sis and the TCA cycle. pathology, such as acute inflammation or inadequate perfusion36,37.
How extensive is such uncoupling? Given the modest net lactate Such accumulation can be read out to drive proper physiological
exchange across major vascular beds, this depends on the extent responses, such as inflammation resolution through favouring the
of lactate by exchange within vascular beds. Such exchange might development of regulatory T cells38, a propensity that can backfire
occur between different tissues sharing the same vascular supply or in the case of cancer by suppressing anticancer immune responses39.
between cell types within a tissue. For example, in the kidney, lactate
uptake by the gluconeogenic cortex may be offset by lactate produc- Redox robustness
tion by the glycolytic medulla28. Or, within the leg, a subset of highly Both lactate and pyruvate circulate, and lactate is approximately 20
glycolytic cells might rapidly ferment circulating glucose to circu- times more abundant in the bloodstream. MCTs can transport both
lating lactate, while other cells consume circulating lactate as their lactate and pyruvate, and one can be exchanged for the other. Once
carbohydrate fuel. For example, adipocytes have been suggested to inside cells, pyruvate and lactate are rapidly interconverted via the
be highly glycolytic and to secrete copious lactate29–33. action of LDH. The direction of net LDH flux depends on the reac-
In agreement with glucose use being relatively restricted, fluoro- tion quotient (Q) relative to the equilibrium constant (Keq) of LDH,
deoxyglucose positron emission tomography (PET) imaging stud- as shown in the following equation:
ies show intense glucose uptake in the brain, tumours and areas of
inflammation, but little uptake in many other parts of the body34. Q ¼ ½lactate½NAD=½pyruvate½NADH
Although this finding partially reflects PET imaging studies being
conducted in fasted resting individuals, the PET data align with with square brackets indicating concentration, and Q > Keq imply-
glucose-transporter expression, which is strongest in the brain and ing lactate consumption. Both lactate consumption and glycolysis
activated immune cells. require NAD as a substrate. Because GAPDH’s Michaelis constant
In contrast to the restricted expression of glucose transporters, for NAD is lower than LDH’s, glycolysis has priority for NAD40,41.
which renders glucose uptake a key gating step in metabolism, the On the basis of the LDH reaction approaching equilibrium, the
nearly universal expression of MCTs makes lactate freely available cellular lactate-to-pyruvate ratio has often been used as a proxy for
to all cells of the body. the cytosolic NADH-to-NAD ratio42–44. If most cytosolic NADH,
Use of lactate as the primary circulating carbohydrate energy pyruvate and lactate were from glycolysis, then intracellular met-
source advantageously reserves glucose for particularly vital systems abolic reactions would indeed control, via NADH, the balance
(such as the brain and immune system) and for biochemical func- between cellular lactate and pyruvate (Fig. 2a). Given the rapidity
tions that cannot be readily achieved by using other substrates (gly- of the pyruvate–lactate exchange between cells and the circulation,
cogen storage, glycosylation, NADPH and ribose generation by the however, causality may flow in the other direction. Specifically, the
pentose-phosphate pathway). By dissociating these processes from abundance of lactate and pyruvate in the circulation may deter-
the housekeeping function of aerobic ATP generation from carbo- mine their intracellular concentrations, which in turn may deter-
hydrate, glucose use can be regulated in tune with more advanced mine the cellular NADH-to-NAD ratio. In this case, the MCT–LDH
organismal needs. For example, in lymphocytes, glucose entry is reaction sequence would function to align every cell’s cytosolic
coordinately regulated with activation and proliferation. Exclusion NAD-to-NADH ratio to the systemic pyruvate-to-lactate ratio,
of glucose in the quiescent state may help prevent autoimmunity thus effectively buffering NAD and NADH in each cell against the
and cancer35. whole-body pyruvate–lactate pool (Fig. 2b).

568 Nature Metabolism | VOL 2 | July 2020 | 566–571 | www.nature.com/natmetab


NATure MeTABoLIsm Perspective

Blood lactate

Glycolysis Tissue TCA


Glucose CO2
lactate

Glycogen

Fig. 3 | Whole-body lactate homeostasis. At the tissue level, lactate exchange with the circulation allows for independent operation of glycolysis and
the TCA cycle. At the organismal level, however, lactate production (largely via glycolysis) and consumption (largely via the TCA cycle) must balance to
maintain lactate homeostasis. Some tissues, such as kidney and muscle, both produce and consume circulating lactate.

Similar redox equilibration can also occur across compartments sis and the TCA cycle at the cellular level, glycolysis and the TCA
of a cell. Mitochondria are notable for lacking MCT transporters on cycle must be balanced at the whole-body level (Fig. 3). TCA entry
their inner membranes, thereby excluding lactate, and instead using via pyruvate carboxylase generates a four-carbon compound, which
mitochondrial pyruvate carriers to import pyruvate. However, per- can be reconverted into 3C compounds via malic enzyme or phos-
oxisomes express MCT2, which may buffer their NAD and NADH phoenolpyruvate carboxykinase47. In contrast, TCA entry via PDH
pools with the cytosol45. generates a two-carbon unit in the form of acetyl-CoA, which in
Redox buffering is based on cells (or compartments) with mammals cannot be reconverted into 3C units (Fig. 4). Thus, the
excess NADH taking in pyruvate and excreting lactate (Fig. 2b). PDH reaction irreversibly clears lactate. Therefore, the whole-body
Importantly, whereas glycolysis terminating in lactate is redox balance between glycolysis and PDH flux is likely to be the key
neutral, uptake of pyruvate and excretion of lactate results in a net determinant of lactate levels.
dumping of electrons. Physiologically, the kidneys clear lactate elec- PDH is a multimeric enzyme whose catalytic activity is regu-
trons, releasing pyruvate and thereby helping to maintain an appro- lated by the phosphorylation status of the E1a subunit and by
priately oxidized circulating lactate-to-pyruvate ratio17. NADH48,49. High phosphorylation, mediated by pyruvate dehydro-
Direct support for the ability of circulating pyruvate and lac- genase kinases (PDK 1–4), impairs mitochondrial pyruvate clear-
tate to control tissue NADH-to-NAD ratio comes from the ance and thereby promotes cellular lactate excretion and systemic
Mootha laboratory, which has recently developed an engineered 3C-compound accumulation. NADH activates PDK and thereby
enzyme that uses molecular oxygen to irreversibly convert circu- inhibits PDH, thus contributing to the elevated circulating lactate in
lating lactate to pyruvate. Administration of this enzyme to mice patients with impaired mitochondrial activity or respiration.
with lactate and NADH build-up due to genetic mitochondrial
deficiency resulted, as desired, in a more oxidized circulating Future perspectives
lactate-to-pyruvate ratio. Importantly, although the enzyme was Given that 3C compounds are mainly produced by glycolysis (Fig.
localized to the bloodstream, both the brain and heart showed 4), mechanisms of lactate homeostasis may substantially overlap
a decreased NADH-to-NAD ratio46. Thus, lactate–pyruvate with those for glucose homeostasis. In insulin resistance, circulat-
exchange renders tissue redox maintenance robust by equilibrat- ing lactate may play a critical role as an energy substrate in cells
ing redox status across the organism. that are carbon limited because of deficient insulin-mediated glu-
cose uptake. Whether interindividual differences in lactate handling
Regulation of lactate homeostasis contribute to diabetes pathogenesis or robustness to diabetic com-
Compared with some other important fuel molecules such as fatty plications is an important topic for future study.
acids, lactate is notable for tight homeostasis of its serum concen- Control of lactate clearance ties into the well-studied regulatory
trations, and elevated serum lactate is a serious medical condition biology of PDK. Despite PDK’s importance, manifested by the exis-
known as lactic acidosis. How are circulating lactate levels regu- tence of four different kinase isozymes, its physiological purpose
lated? Lactate’s entry and exit from cells are governed by MCTs remains unclear. One likely role is redox homeostasis, promot-
1–4 (Slc16a1, Slc16a7, Slc16a3 and Slc16a4). Both the expression ing lactate export instead of catabolism when NADH builds up.
and activity of these proteins can potentially be regulated to con- Another possibility is that, during starvation, PDK serves to shut
trol lactate localization in the body. MCT flux, however, is fast, and off PDH to preserve 3C units50. Conversion of these 3C units into
therefore lactate is typically well mixed between tissues and the cir- glucose would then preserve the availability of glucose for the brain
culation. Accordingly, regulation of lactate concentration occurs and immune system.
substantially at the level of the whole-body lactate pool. Because Given the particularly tight control of circulating lactate levels,
lactate is always much more abundant in the circulation than pyru- a novel regulatory system for lactate homeostasis is likely to exist.
vate, lactate homeostasis depends on controlling the total pool size Lactate can potentially be cleared by urinary excretion in addition
of these 3C carboxylic acids. to PDH. Renal lactate loss is typically prevented by urinary reab-
To maintain tight concentration homeostasis, overall production sorption via the sodium/lactate co-transporters Slc5a12 and Slc5A8
and consumption of lactate must be synchronized. The main route (ref. 51). Whether these enzymes are sometimes downregulated to
of 3C-compound production is glycolysis. The major consumption release lactate is unclear. Circulating lactate can also enter the faeces
route is via the TCA cycle. Thus, whereas lactate decouples glycoly- and thereby feed the microbiome52. A quantitative understanding

Nature Metabolism | VOL 2 | July 2020 | 566–571 | www.nature.com/natmetab 569


Perspective NATure MeTABoLIsm

Glycogen Glucose 12. Lemons, J. M. et al. Quiescent fibroblasts exhibit high metabolic activity. PLoS
Biol. 8, e1000514 (2010).
13. Cantor, J. R. et al. Physiologic medium rewires cellular metabolism and
reveals uric acid as an endogenous inhibitor of UMP synthase. Cell 169,
Glycolysis makes 3C units 258–272.e17 (2017).
14. Vander Heiden, M. G. et al. Growth factors can influence cell growth and
survival through effects on glucose metabolism. Mol. Cell. Biol. 21, 5899–5912
(2001).
3C (lactate) 15. Cori, C. F. Glycogen breakdown and synthesis in animal tissues.
PDH clears 3C units Endocrinology 26, 285–296 (1940).
16. Wahren, J., Felig, P., Ahlborg, G. & Jorfeldt, L. Glucose metabolism during leg
2C (Acetyl-CoA) exercise in man. J. Clin. Invest. 50, 2715–2725 (1971).
17. Jang, C. et al. Metabolite exchange between mammalian organs quantified in
pigs. Cell Metab. 30, 594–606.e3 (2019).
18. Dunn, A., Katz, J., Golden, S. & Chenoweth, M. Estimation of glucose
4C Citrate turnover and recycling in rabbits using various [3H, 14C]glucose labels. Am. J.
Physiol. 230, 1159–1162 (1976).
19. Katz, J., Okajima, F., Chenoweth, M. & Dunn, A. The determination of lactate
turnover in vivo with 3H- and 14C-labelled lactate. The significance of sites of
tracer administration and sampling. Biochem. J. 194, 513–524 (1981).
20. Stanley, W. C. et al. Lactate extraction during net lactate release in legs of
humans during exercise. J. Appl. Physiol. 60, 1116–1120 (1986).
21. Wolfe, R. R. Isotopic measurement of glucose and lactate kinetics. Ann. Med.
22, 163–170 (1990).
22. Hui, S. et al. Glucose feeds the TCA cycle via circulating lactate. Nature 551,
115–118 (2017).
Fig. 4 | Production and consumption of 3C units. At the organismal level, 23. Faubert, B. et al. Lactate metabolism in human lung tumors. Cell 171,
3C-unit production (largely via glycolysis) and consumption (largely 358–371.e9 (2017).
via PDH) must balance to maintain lactate homeostasis. Bidirectional 24. Halestrap, A. P. The monocarboxylate transporter family: structure and
connections indicate that the metabolites can interconvert (typically with functional characterization. IUBMB Life 64, 1–9 (2012).
25. Brooks, G. A. The science and translation of lactate shuttle theory. Cell Metab.
energy input, for example, via glycolysis or gluconeogenesis). 27, 757–785 (2018).
26. Zhao, C., Wilson, M. C., Schuit, F., Halestrap, A. P. & Rutter, G. A. Expression
and distribution of lactate/monocarboxylate transporter isoforms in
of the importance and regulation of microbiome-mediated lactate pancreatic islets and the exocrine pancreas. Diabetes 50, 361–366 (2001).
clearance merits further investigation. 27. Sekine, N. et al. Low lactate dehydrogenase and high mitochondrial glycerol
Major metabolic enzymes and transporters, including glycolytic phosphate dehydrogenase in pancreatic beta-cells: potential role in nutrient
enzymes, MCTs, LDH, mitochondrial pyruvate carriers and PDH, sensing. J. Biol. Chem. 269, 4895–4902 (1994).
28. Wirthensohn, G. & Guder, W. G. Renal substrate metabolism. Physiol. Rev.
are also potential targets for novel regulatory circuits controlling 66, 469–497 (1986).
lactate homeostasis. Such circuits would presumably involve lactate 29. Mårin, P., Rebuffé-Scrive, M., Smith, U. & Björntorp, P. Glucose uptake in
sensors such as G-protein-coupled receptor 81 (ref. 53). Given the human adipose tissue. Metabolism 36, 1154–1160 (1987).
role of lactate as a universal fuel, the elucidation of such a system— 30. Gustafsson, J., Eriksson, J. & Marcus, C. Glucose metabolism in human
for ensuring a beautiful but not overcrowded ‘pool of swans’—has adipose tissue studied by 13C-glucose and microdialysis. Scand. J. Clin. Lab.
Invest. 67, 155–164 (2007).
the potential to reshape understanding of mammalian metabolism 31. DiGirolamo, M., Newby, F. D. & Lovejoy, J. Lactate production in adipose
and disease treatment. tissue: a regulated function with extra-adipose implications. FASEB J. 6,
2405–2412 (1992).
Received: 23 April 2020; Accepted: 16 June 2020; 32. Tozzo, E., Shepherd, P. R., Gnudi, L. & Kahn, B. B. Transgenic GLUT-4
Published online: 20 July 2020 overexpression in fat enhances glucose metabolism: preferential effect on fatty
acid synthesis. Am. J. Physiol. 268, E956–E964 (1995).
33. Jansson, P. A., Smith, U. & Lönnroth, P. Evidence for lactate production by
References human adipose tissue in vivo. Diabetologia 33, 253–256 (1990).
1. Chen, Y.-J. et al. Lactate metabolism is associated with mammalian 34. Boellaard, R. et al. FDG PET/CT: EANM procedure guidelines for tumour
mitochondria. Nat. Chem. Biol. 12, 937–943 (2016). imaging: version 2.0. Eur. J. Nucl. Med. Mol. Imaging 42, 328–354 (2015).
2. Barron, J. T., Gu, L. & Parrillo, J. E. Malate-aspartate shuttle, cytoplasmic 35. Macintyre, A. N. et al. The glucose transporter Glut1 is selectively essential
NADH redox potential, and energetics in vascular smooth muscle. J. Mol. for CD4 T cell activation and effector function. Cell Metab. 20, 61–72
Cell. Cardiol. 30, 1571–1579 (1998). (2014).
3. LaNoue, K. F. & Williamson, J. R. Interrelationships between malate-aspartate 36. Pucino, V. et al. Lactate buildup at the site of chronic inflammation promotes
shuttle and citric acid cycle in rat heart mitochondria. Metabolism 20, disease by inducing CD4+ T cell metabolic rewiring. Cell Metab. 30,
119–140 (1971). 1055–1074.e8 (2019).
4. Houstĕk, J., Cannon, B. & Lindberg, O. Gylcerol-3-phosphate shuttle and its 37. Zhang, J. et al. Endothelial lactate controls muscle regeneration from ischemia
function in intermediary metabolism of hamster brown-adipose tissue. Eur. J. by inducing M2-like macrophage polarization. Cell Metab. 31, 1136–1153.e7
Biochem. 54, 11–18 (1975). (2020).
5. Jorfeldt, L., Juhlin-Dannfelt, A. & Karlsson, J. Lactate release in relation to 38. Angelin, A. et al. Foxp3 reprograms T cell metabolism to function in
tissue lactate in human skeletal muscle during exercise. J. Appl. Physiol. 44, low-glucose, high-lactate environments. Cell Metab. 25, 1282–1293.e7 (2017).
350–352 (1978). 39. Brand, A. et al. LDHA-associated lactic acid production blunts tumor
6. Warburg, O., Wind, F. & Negelein, E. The metabolism of tumors in the body. immunosurveillance by T and NK cells. Cell Metab. 24, 657–671 (2016).
J. Gen. Physiol. 8, 519–530 (1927). 40. Mountassif, D. et al. Immunoaffinity purification and characterization of
7. Warburg, O. On the origin of cancer cells. Science 123, 309–314 (1956). glyceraldehyde-3-phosphate dehydrogenase from human erythrocytes. Acta
8. Vyas, S., Zaganjor, E. & Haigis, M. C. Mitochondria and cancer. Cell 166, Biochim. Biophys. Sin. (Shanghai) 41, 399–406 (2009).
555–566 (2016). 41. Talaiezadeh, A., Shahriari, A., Tabandeh, M. R., Fathizadeh, P. & Mansouri, S.
9. Wallace, D. C. Mitochondria and cancer. Nat. Rev. Cancer 12, 685–698 (2012). Kinetic characterization of lactate dehydrogenase in normal and malignant
10. Zong, W. X., Rabinowitz, J. D. & White, E. Mitochondria and cancer. Mol. human breast tissues. Cancer Cell Int. 15, 19 (2015).
Cell 61, 667–676 (2016). 42. Mintun, M. A., Vlassenko, A. G., Rundle, M. M. & Raichle, M. E. Increased
11. Rathmell, J. C., Vander Heiden, M. G., Harris, M. H., Frauwirth, K. A. & lactate/pyruvate ratio augments blood flow in physiologically activated human
Thompson, C. B. In the absence of extrinsic signals, nutrient utilization by brain. Proc. Natl Acad. Sci. USA 101, 659–664 (2004).
lymphocytes is insufficient to maintain either cell size or viability. Mol. Cell 6, 43. Lambeth, M. J. & Kushmerick, M. J. A computational model for
683–692 (2000). glycogenolysis in skeletal muscle. Ann. Biomed. Eng. 30, 808–827 (2002).

570 Nature Metabolism | VOL 2 | July 2020 | 566–571 | www.nature.com/natmetab


NATure MeTABoLIsm Perspective
44. Williamson, D. H., Lund, P. & Krebs, H. A. The redox state of free Acknowledgements
nicotinamide-adenine dinucleotide in the cytoplasm and mitochondria of rat S.E. is supported by the Swedish Research Council (2019-00773, 2018-02537), The Knut
liver. Biochem. J. 103, 514–527 (1967). and Alice Wallenberg Foundation, Sahlgrenska’s University Hospital (LUA-ALF) and
45. McClelland, G. B., Khanna, S., González, G. F., Butz, C. E. & Brooks, G. A. Novo Nordisk Foundation. J.D.R. is supported by NIH Pioneer award 1DP1DK113643,
Peroxisomal membrane monocarboxylate transporters: evidence for a redox Diabetes Research Center grant P30 DK019525 and Pfizer, Inc.
shuttle system? Biochem. Biophys. Res. Commun. 304, 130–135 (2003).
46. Patgiri, A. et al. An engineered enzyme that targets circulating lactate to alleviate
intracellular NADH:NAD+ imbalance. Nat. Biotechnol. 38, 309–313 (2020). Author contributions
47. Jitrapakdee, S., Vidal-Puig, A. & Wallace, J. C. Anaplerotic roles of pyruvate J.R. and S.E. equally conceived the idea, selected content and wrote the manuscript.
carboxylase in mammalian tissues. Cell. Mol. Life Sci. 63, 843–854 (2006).
48. Holness, M. J. & Sugden, M. C. Regulation of pyruvate dehydrogenase
complex activity by reversible phosphorylation. Biochem. Soc. Trans. 31, Competing interests
1143–1151 (2003). J.D.R. is a cofounder and stockholder in VL54 and Raze Therapeutics, and an advisor
49. Bowker-Kinley, M. M., Davis, W. I., Wu, P., Harris, R. A. & Popov, K. M. and stockholder in Agios Pharmaceuticals, Kadmon Pharmaceuticals, Bantam
Evidence for existence of tissue-specific regulation of the mammalian Pharmaceuticals, Colorado Research Partners, Rafael Pharmaceuticals and L.E.A.F.
pyruvate dehydrogenase complex. Biochem. J. 329, 191–196 (1998). Pharmaceuticals. S.E. declares no competing interests.
50. Sukonina, V. et al. FOXK1 and FOXK2 regulate aerobic glycolysis. Nature
566, 279–283 (2019).
51. Gopal, E. et al. Expression of slc5a8 in kidney and its role in Na+-coupled Additional information
transport of lactate. J. Biol. Chem. 279, 44522–44532 (2004). Correspondence should be addressed to J.D.R. or S.E.
52. Scheiman, J. et al. Meta-omics analysis of elite athletes identifies a Peer review information Primary Handling Editor: Christoph Schmitt.
performance-enhancing microbe that functions via lactate metabolism. Nat. Reprints and permissions information is available at www.nature.com/reprints.
Med. 25, 1104–1109 (2019).
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in
53. Madaan, A. et al. Lactate produced during labor modulates uterine
published maps and institutional affiliations.
inflammation via GPR81 (HCA1). Am. J. Obstet. Gynecol. 216,
60.e1–60.e17 (2017). © Springer Nature Limited 2020

Nature Metabolism | VOL 2 | July 2020 | 566–571 | www.nature.com/natmetab 571

You might also like