0% found this document useful (0 votes)
18 views23 pages

The Journal of Physiology - 2021 - Brooks - Lactate in Contemporary Biology A Phoenix Risen

Uploaded by

Arash Khaknejad
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
0% found this document useful (0 votes)
18 views23 pages

The Journal of Physiology - 2021 - Brooks - Lactate in Contemporary Biology A Phoenix Risen

Uploaded by

Arash Khaknejad
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
You are on page 1/ 23

J Physiol 600.

5 (2022) pp 1229–1251 1229

TOPICAL REVIEW

Lactate in contemporary biology: a phoenix risen


George A. Brooks , Jose A. Arevalo, Adam D. Osmond, Robert G. Leija, Casey C. Curl
and Ashley P. Tovar
Exercise Physiology Laboratory, Department of Integrative Biology, University of California, Berkeley, CA, USA

Edited by: Ian Forsythe & Lykke Sylow

Cell signaling
Cell fueling

Microbiome Adaptation
The Journal of Physiology

Metabolic flexibility Reproduction

Satiety regulation Resuscitation

Lactate

Burn Anaerobic Fatigue Acid


Cramps threshold Waste
O2 debt Metabolic agent
product
poison

Abstract After a century, it’s time to turn the page on understanding of lactate metabolism
and appreciate that lactate shuttling is an important component of intermediary metabolism
in vivo. Cell-cell and intracellular lactate shuttles fulfil purposes of energy substrate production
and distribution, as well as cell signalling under fully aerobic conditions. Recognition of lactate

George A. Brooks is Professor of Integrative Biology at the University of California, Berkeley and
Docteur Honoris Causa de l’Université Montpellier. Dr Brooks has received Honor Awards from
the American College of Sports Medicine and the Exercise and Environmental Physiology Section
of the American Physiological Society. His research interests involve bioenergetics, mitochondrial
morphology, energetics and biogenesis, and the regulation of energy substrate partitioning. His work
on the lactate shuttle has influenced thinking in fields as diverse as brain and cancer metabolism.
When not in the laboratory, his focus centres on interests of family and friends. José Arevalo is a
3rd year PhD student at the University of California Berkeley investigating ageing mitochondrial fragmentation and what drives mitochondrial
dysfunction with age progression in skeletal muscle. José was born in Guatemala and grew up in the Boyle Heights neighbourhood of East Los
Angeles, California. José received his master’s degree in kinesiology from California State University Fullerton. When not in the laboratory, his
interests are football, rugby, trail running, and lifting weights.

© 2021 The Authors. The Journal of Physiology published by John Wiley & Sons Ltd on behalf of The Physiological Society DOI: 10.1113/JP280955

This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution
and reproduction in any medium, provided the original work is properly cited.
14697793, 2022, 5, Downloaded from https://physoc.onlinelibrary.wiley.com/doi/10.1113/JP280955 by Schweizerische Akademie Der, Wiley Online Library on [10/11/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1230 G. A. Brooks and others J Physiol 600.5

shuttling came first in studies of physical exercise where the roles of driver (producer) and
recipient (consumer) cells and tissues were obvious. Moreover, the presence of lactate shuttling as
part of postprandial glucose disposal and satiety signalling has been recognized. Mitochondrial
respiration creates the physiological sink for lactate disposal in vivo. Repeated lactate exposure
from regular exercise results in adaptive processes such as mitochondrial biogenesis and other
healthful circulatory and neurological characteristics such as improved physical work capacity,
metabolic flexibility, learning, and memory. The importance of lactate and lactate shuttling in
healthful living is further emphasized when lactate signalling and shuttling are dysregulated as
occurs in particular illnesses and injuries. Like a phoenix, lactate has risen to major importance
in 21st century biology.
(Received 5 November 2020; accepted after revision 21 January 2021; first published online 10 February 2021)
Corresponding author G. A. Brooks: 5101 Valley Life Sciences Building, Exercise Physiology Laboratory, Berkeley, CA
94720-3140, USA. Email: [email protected]

Abstract figure legend: Roles of lactate in physiology and metabolism elaborated as: cell fuelling (the preferential use
of lactate over other fuel energy substrates such as glucose, fatty and keto acids in cardiac and red skeletal muscle
and brain for cognition); cell signalling (the mechanisms by which lactate affects metabolism via effects on cell redox,
ROS production, lactylation and covalent binding); microbiome (the role of fermentation in health and disease and
the possibility of a gut-soma lactate shuttle); satiety (the role of lactate on the arcuate nucleus of the hypothalamus
and gut-hypothalamic signalling); adaptation (the role of lactate in expression of glycolytic and metabolic enzymes and
insulin sensitivity, the latter by TGF-β); reproduction (roles of lactate in sperm mitochondrial energetics, developing
a favourable microenvironment for blastocysts and secretion of VEGF for uterine angiogenesis), and resuscitation
(treatments of dehydration, acidosis, dengue, pancreatitis and hepatitis, myocardial infarction and wound healing).

Introduction a certain extent despite obvious evidence to the contrary.


Commencing with work of early 20th century Nobel prize
The story of lactate and its role in physiology and medicine winners (Hill, 1914; Meyerhof, 1920), the role of lactate
may be a century old, but has changed dramatically in in metabolism has been misunderstood and perpetuated.
the last three decades (Brooks, 1986, 2002, 2018; Gladden, Even as their cell and tissue cultures incubated in air
2004). No longer conceived of as a dead-end metabolite, often turned acidic overnight, textbook authors (e.g.
a fatigue agent, or metabolic poison, in contemporary Lehninger, 1970) perpetuated the myth of oxygen-limited
physiology, lactate is seen as a major metabolic inter- metabolism giving rise to lactate formation despite the
mediate that has wide ranging impacts in energy substrate fact that the partial pressure of oxygen in air over their
utilization, cell signalling, and adaptation; simply, lactate mitochondrial preparations and culture dishes was 3–5
is at the fulcrum of metabolic integration (Brooks, 1984, times higher than in vivo (Brooks et al. 2019). Hence, a
1986, 2020a). Now, rather than regarded as an oddity of reckoning of minds in biology is necessary to understand
exercise metabolism (Gladden, 2004; Chen et al. 2016; human and mammalian metabolism in a contemporary
Hui et al. 2017; Brooks, 2018; Ferguson et al. 2018), context.
the presence of lactate shuttling is recognized in fields Take for instance conflations of terms ‘glycolysis’ and
as diverse as wound healing (Hunt et al. 2007), cancer ‘fermentation.’ In mammalian tissues, glycolysis (i.e.
biology (San-Millan & Brooks, 2017), insulin secretion conversion of glucose and glycogen to lactate) is very
(Rutter et al. 2015), management of sepsis (Garcia-Alvarez different from aerobic fermentation of sugar to alcohol
et al. 2014a), learning and memory (Suzuki et al. 2011; El by yeast, or the production of swamp gases by anaerobic
Hayek et al. 2019), and treatment of traumatic brain injury bacteria as occurs in the colon, at wound sites or in rotting
(TBI) (Brooks & Martin, 2014). Hence, in contemporary (Brooks, 2018, 2020c; Ferguson et al. 2018). Conflation
biology, the role of lactate in metabolism needs to be of the terms perpetuates the myth of lactate poisoning
understood and viewed as a ‘phoenix risen’ (Brooks, 2002, and thus handicaps understanding of basic biology and
2018) in contrast to a remnant of (1988) early 20th century its translation. Fortunately, textbook authors are noting a
biology (Rabinowitz & Enerback, 2020). distinction between glycolysis as occurs in animals in vivo,
As scholars we have our limitations and, as a and fermentation processes that occur in microbes giving
consequence, in formal education we rely on tradition to rise to ethanol and foul gases (Urry et al. 2020).

© 2021 The Authors. The Journal of Physiology published by John Wiley & Sons Ltd on behalf of The Physiological Society
14697793, 2022, 5, Downloaded from https://physoc.onlinelibrary.wiley.com/doi/10.1113/JP280955 by Schweizerische Akademie Der, Wiley Online Library on [10/11/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J Physiol 600.5 Lactate, phoenix risen 1231

Lactate shuttling: roles of driver and recipient cells beds. For instance, during exercise, fast white fibres can
provide oxidizable substrate to red, oxidative fibres in the
Like other metabolites, lactate flux between cells and
same tissue bed (Baldwin et al. 1972; Hooker & Baldwin,
tissue beds depends on concentration and hydrogen
1979). Conversely, postprandial glucose uptake in red
ion (pH) differences. Lactate exchanges between and
fibres can provide substrate to the body corpus as in the
among cells are facilitated by the presence of cell
glucose paradox (Foster, 1984). Also, working muscle can
membrane lactate transport proteins termed mono-
fuel the beating heart (Gertz et al. 1981), brain (Suzuki
carboxylate transporters (MCTs) (Garcia et al. 1994,
et al. 2011; Glenn et al. 2015a; Steinman et al. 2016) and
1995); MCTs are bidirectional symporters (Roth &
provide gluconeogenic substrates to the splanchnic organs
Brooks, 1990a,b; Brown & Brooks, 1994) sensitive to
(Bergman et al. 2000; Gerich et al. 2001).
trans-stimulation by lactate and hydrogen ion gradients.
Lactate exchanges among muscle, heart, liver and
Hence, we have the concept of glycolytic lactate producing
kidneys are obvious examples of lactate shuttling.
(driver) and lactate consuming (recipient) cells (Fig. 1).
However, are other organs involved? Some might argue
From first observations on dog gracilis muscle pre-
that the integument is the largest organ in the body,
parations that showed net lactate release at rest, an
and it is known to produce lactate under sympathetic
increased release when contractions started followed by
stimulation (Johnson & Fusaro, 1972). Hence, is there
net lactate uptake as contractions continued (Stainsby &
a skin-muscle lactate shuttle during exercise (Brooks,
Welch, 1966), the concepts of lactate driver and recipient
2018)? Similarly, the gut, the only organ of fermentation
cells could have been predicted. Subsequently, similar
in the body, produces lactate because of the presence of
results of net lactate release from resting muscle, followed
bacteria and dietary fibre in the colon may release lactate
by increased release and switch to uptake during exercise
into the systemic circulation. Hence, we can speculate
were confirmed in human muscles (Stanley et al. 1985;
on the presence of a gut to soma lactate shuttle (Brooks,
Bergman et al. 1999b). Again, from such results, the pre-
2018). As there are no obvious venous drainage vessels to
sence of lactate driver and recipient cells as part of lactate
catheterize, other methods to detect lactate exchange in
shuttling could be inferred. Not surprisingly, we now
the integument and gut will need to be found to quantify
know not only that driver and recipient cells can switch
their roles in lactate shuttling under various conditions.
roles depending on conditions, but that some cells can
exchange lactate through the interstitium and vascular

Contributions from muscle and exercise physiology


The lactate shuttle: Lactate the autocrine, (the cell-cell and intracellular lactate shuttles)
paracrine and endocrine link between glycolytic and
oxidative metabolism The cell-cell lactate shuttle. As is often the case, it is
difficult to identify a seminal moment of discovery or
publication, but from this perspective, it was results
on dog muscles made to contract in situ (Stainsby &
Welch, 1966). At rest, those highly red, richly perfused
muscles always released lactate. Then, at the onset of
contractions, lactate release increased, but then switched
to net uptake as contractions continued and oxygen
consumption rose (vide supra). So, in a kernel, we
have it, muscle lactate production under fully aerobic
conditions followed by uptake as oxygen consumption
Glycolytic Oxidative rises to meet metabolic demand. Subsequently, using
metabolism metabolism
NADH fluoroscopy, investigators (Jöbsis & Stainsby, 1968)
Lactate
verified that working muscles were not oxygen limited.
Later efforts to study working muscle lactate metabolism
(Honig et al. 1992) using myoglobin cryomicroscopy
again showed ‘aerobic glycolysis’ in working muscle.
Importantly, Gladden and associates determined effects
Figure 1. The concept of lactate shuttling between producer of exercise intensity, blood lactate concentration, O2
(driver) cells and tissues and consumer (recipient) consumption and pH on lactate production and disposal
By this mechanism lactate has autocrine-, paracrine- and
endocrine-like influences on metabolism that fulfil at least three
using arterial-venous differences ((a-v)), blood flow and
14
purposes. Lactate is: (1) a major energy source; (2) the major C-lactate measurements on resting and contracting
gluconeogenic precursor; and (3) a signalling molecule. Revised from canine muscles in situ (Gladden, 1991; Gladden et al.
Brooks (2018). 1994; Kelley et al. 2002). In their studies investigators

© 2021 The Authors. The Journal of Physiology published by John Wiley & Sons Ltd on behalf of The Physiological Society
14697793, 2022, 5, Downloaded from https://physoc.onlinelibrary.wiley.com/doi/10.1113/JP280955 by Schweizerische Akademie Der, Wiley Online Library on [10/11/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1232 G. A. Brooks and others J Physiol 600.5

showed lactate turnover (production and disposal) in well above the threshold for decreased ATP production
resting and contracting, fully oxygenated and circulated (0.80 mmHg) (Kreutzer & Jue, 1995). Thus, while there
canine muscle (for reviews see Gladden, 2004; Rogatzki is no singular value that would imply a threshold for
et al. 2015). increased lactate production in human muscle or rat heart,
The advent of radiotracers and their use in physio- it is certain that lactate is formed under fully aerobic,
logy and metabolism soon showed continuous lactate normoxic conditions (Connett et al. 1990).
production under fully aerobic conditions in dogs Complimentary to determinations of lactate disposal
(Depocas et al. 1969) and rats (Freminet et al. 1974; via oxidation were those determining lactate disposal
Donovan & Brooks, 1983), with disposal during exercise via gluconeogenesis in resting postabsorptive humans.
by oxidation and conversion to glucose (Brooks & Hence, it is fair to state that while most lactate disposal
Donovan, 1983). Moreover, in studies on rats recovering is accomplished via oxidation, with a minority of
from exercise, the so called ‘oxygen debt’ (now referred to lactate disposal accomplished via gluconeogenesis under
as EPOC: Gasser & Brooks, 1984) period showed oxidative postabsorptive resting conditions, lactate is the most
disposal (4/5) predominating over gluconeogenesis and important gluconeogenic precursor during rest and
glyconeogenesis (1/5) (Brooks et al. 1973; Brooks & exercise (Stanley et al. 1988; Bergman et al. 2000; Gerich
Gaesser, 1980; Gaesser & Brooks, 1980). Compared to et al. 2001)
the classic results of Meyerhof on isolated frog muscle In sum, the above-cited work shows continuous aerobic
preparations ex vivo (Meyerhof, 1920), results on rats in (not oxygen-limited) lactate turnover (production and
vivo upended the long-held idea that after exercise 4/5 of disposal) in humans and mammalian model systems.
the lactate produced during contractions was restored to Further, the work shows two of three features of the lactate
muscle glycogen in situ. shuttle – lactate production in driver cells and disposal in
Initial efforts to study lactate-glucose interactions in recipient cells and tissues; signalling being a third feature
humans involved (a-v) and blood flow measurements of the lactate shuttle (Brooks, 2000, 2002, 2018). Hence,
across working muscle and splanchnic tissue beds we now realize that lactate produced in working muscle is
(Ahlborg & Felig, 1982; Wahren & Ekberg, 2007). a major fuel energy source (Stanley et al. 1986; Bergman
Such experiments are highly invasive and difficult to et al. 1999b), but also that lactate release from working
conduct and interpret. Nonetheless, the significance muscle fuels other organs such as the heart (Gertz et al.
of the technologies employed and lessons learned 1988; Bergman et al. 2009b) and brain (Glenn et al. 2015a)
from those difficult experiments showing inter-organ normally, but also in illness and following injury (Marik
metabolite exchanges in resting and exercising humans & Bellomo, 2013; Brooks & Martin, 2014; Garcia-Alvarez
are noteworthy. et al. 2014b). Moreover, lactate released from working
The advent of stable, non-radioactive tracers and their muscles (Stanley et al. 1986; Bergman et al. 1999b) and
use in physiology and metabolism soon replicated what other driver cells such as the integument is the major
was shown in mammalian models (Stanley et al. 1985, gluconeogenic precursor (Stanley et al. 1988; Bergman
1986; Mazzeo et al. 1986). Subsequently, combined use et al. 2000) (Fig. 2).
of (a-v) differences and blood flow, as well as iso-
topic tracers, showed simultaneous lactate production and
oxidative disposal within resting and working human The intracellular lactate shuttle (ICLS). The above-cited
skeletal muscles (Stanley et al. 1986; Bergman et al. 1999b). studies using infused 13 C-lactate and observing 13 CO2
More recently, the same technologies have been employed excretion from resting and working muscles show
to show lactate turnover (production, net uptake and simultaneous lactate uptake, production, and oxidation
oxidative disposal) in the heart (Gertz et al. 1981, 1988; in vivo (Stanley et al. 1986; Bergman et al. 1999b).
Bergman et al. 2009b) and brain (Glenn et al. 2015a).In However, those studies do not provide information on
their studies of muscle lactate metabolism Richardson the intracellular path of lactate oxidation. Hence, studies
and colleagues made ingenious use of repeated studies of on isolated mitochondria (Brooks et al. 1999c, 1999d;
muscle net balance ((a-v) and blood flow) and nuclear Dubouchaud et al. 2000; Hashimoto et al. 2006, 2008;
magnetic resonance (NMR) to determine intracellular PO2 Atlante et al. 2007; Passarella et al. 2008, 2014) and
from the myoglobin spectrum (Richardson et al. 1998). muscles (Park et al. 2015) were called for.
Their results showed lactate production in, and release To understand mitochondrial lactate oxidation in
from, resting and exercising muscle when intracellular vivo, one needs to appreciate that the cellular respiratory
PO2 was significantly above the critical mitochondrial apparatus is composed not of discrete vesicles (i.e.
oxygen tension as previously determined (Wilson et al. mitochondria), but rather an extensive network, a
1988). Similarly, with their MRS experiments on rat heart mitochondrial reticulum (Bakeeva et al. 1978; Kirkwood
preparations, Kreutzer and Jue demonstrated myocardial et al. 1986, 1987) that extends from the sub-sarcolemmal
lactate formation above an intracellular PO2 that was domain to deep within fibres, that represents an ‘energy

© 2021 The Authors. The Journal of Physiology published by John Wiley & Sons Ltd on behalf of The Physiological Society
14697793, 2022, 5, Downloaded from https://physoc.onlinelibrary.wiley.com/doi/10.1113/JP280955 by Schweizerische Akademie Der, Wiley Online Library on [10/11/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J Physiol 600.5 Lactate, phoenix risen 1233

highway’ (Glancy et al. 2015). To oxidize energy products


of glycolysis, the reticulum contains both lactate (mMCT)
(Brooks et al. 1999c) and pyruvate (mPC) transporters
(Bricker et al. 2012; Herzig et al. 2012). Importantly,
because the product of glycolysis is lactate, not pyruvate
(Rogatzki et al. 2015), mitochondrial uptake and oxidation
far exceeds that of pyruvate. Hence, studies of muscle
lactate oxidation led to discovery of the mitochondrial
Brain lactate oxidation complex (mLOC) in vivo.
In terms of functionality, the mLOC contains several
essential components of lactate oxidation: an MCT, its
membrane chaperone Basigin (BSG or CD147), lactate
Mixed venous
(Pulmonary artery) Lac Gluc dehydrogenase (LDH), and cytochrome oxidase (COx) as
seen in muscle (Hashimoto et al. 2006), liver (De Bari
et al. 2004; Passarella et al. 2014), and brain (Hashimoto
et al. 2008), and various model systems such as brain slices
Lungs (Schurr, 2008), primary neuronal cultures (Atlante et al.
2007; Hashimoto et al. 2008), normal breast and trans-
formed breast cancer cells (Hussien & Brooks, 2011), and
tumours (Sonveaux et al. 2008). Colocalization of MCTs,
LDH and COx has been seen in muscles (Hashimoto et al.
Pulmonary 2005, 2007, 2013), brain (Hashimoto et al. 2008), lung
vein
(Systemic (Johnson et al. 2012), and cancer cells (Hussien & Brooks,
Heart
arterial) 2011).
Confirmation of the preference of mitochondrial lactate
over pyruvate oxidation comes from studies of hyper-
Lac Gluc
Lac Gluc polarized lactate in muscles in situ (Park et al. 2015). So
Coronary Lac Gluc far as mitochondrial pyruvate transport and oxidation are
sinus Lac Gluc concerned, it is known that the putative mPC colocalizes
Lac with mitochondrial MCT1 in L6 myocytes. In preliminary
studies, colocalization analysis of mMCT1 and mPC1
Liver
using Imaris software revealed that both MCT1 and mPC
Gluc Lac are co-localized to the mitochondria (r2 = 0.8) (Brooks,
2020c). However, because muscle lactate concentration
Glycogen exceeds that of pyruvate by one (10×) to two (200–400×)
Lac Lac Gluc orders of magnitude in resting and exercising human
muscles, respectively (Henderson et al. 2004), lactate
Lactate Glycogen + Glucose
oxidation is dominant in vivo.
Integument and muscle As will be discussed in more detail later, an important
feature of lactate shuttling is that it affects cell redox
in both driver and recipient cells, and between cell
compartments. At present there is discussion over
whether the ICLS functions independently of or
cooperatively with longer established malate-aspartate
and glycerol phosphate shuttles (Kane, 2014). Given
Figure 2. Illustration of lactate shuttling during exercise (the redundancies in physiological control systems, it may be
cell-cell lactate shuttle) that cytosolic-to-mitochondrial shuttles work in parallel
Lactate released from muscles, skin and other driver cells provides to manage the lactate load and balance cell redox during
energy for working muscles (Stanley et al. 1986; Bergman et al. exercise and other conditions. Using CRISPR knockout
1999b), heart (Gertz et al. 1981, 1988; Bergman et al. 2009b) and
brain (Glenn et al. 2015a). Moreover, lactate released from working
technology in mouse or other mammalian models may
muscles (Stanley et al. 1986; Bergman et al. 1999b) and other driver provide missing data on the relative uses of specific
cells such as adipose tissue is the major gluconeogenic precursor intracellular redox shuttles under particular physiological
(Stanley et al. 1988; Bergman et al. 2000) and brain fuel, even after circumstances (Kane, 2014).
injury when lactate supplementation may be efficacious (Brooks &
Martin, 2014). Revised from Brooks (2018).

© 2021 The Authors. The Journal of Physiology published by John Wiley & Sons Ltd on behalf of The Physiological Society
14697793, 2022, 5, Downloaded from https://physoc.onlinelibrary.wiley.com/doi/10.1113/JP280955 by Schweizerische Akademie Der, Wiley Online Library on [10/11/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1234 G. A. Brooks and others J Physiol 600.5

Lactate shuttling as a basis for the lactate threshold glycogen synthesis is to be contrasted with the ‘direct’
in physical exercise. It has been half a century since pathway in which dietary glucose from the gut is taken up
introduction of the concept of oxygen-limited metabolism and converted to liver glycogen on first circulatory pass
producing an ‘anaerobic threshold’ (AT), that is, O2 (Fig. 3).
limited metabolism during exercise of a particular The initial concept developed from studies on lab
intensity. The subject has been recently reviewed (Poole animals has been replicated in human subjects showing
et al. 2021). With 1541 citations (Web of Science, 27th both indirect and direct liver glycogen synthesis in healthy,
September, 2020) the paper of (Wasserman & McIlroy, postprandial humans. However, the balance of indirect
1964) is the eighth most cited paper of all time in the and direct glucose conversion appears to be species
Journal of Applied Physiology. As recited in the recent related. Results of Gerich and colleagues in human sub-
review (Poole et al. 2021), in a letter from Karlman jects confirm that glycolysis is the main initial post-
Wasserman to this author, the concept of an ‘anaerobic prandial fate of glucose, accounting for ≈66% of over-
threshold’ was based on the assumption that ‘failure of all disposal, while oxidation and storage each accounted
the heart to transport O2 adequately would result in for ≈45%. However, the majority of hepatic glycogen
lactic acidosis’, an event that would trigger ventilatory synthesis in postprandial humans (≈73%) was formed via
compensation. Subsequently, concepts of anaerobic, the direct pathway (Woerle et al. 2003). In the near future
lactate, ventilatory and other related thresholds (e.g. it should be possible to better understand the issue of
OBLA) are pervasive in the literature and have represented influences of diet and other factors on the balance of direct
key concepts in basic and applied physiology, sports and vs. indirect liver and muscle glycogen synthesis in humans
pulmonary medicine. These thresholds are determined using 13 C-tracers and magnetic resonance spectrometry
by turnpoints in the rise in blood lactate concentration, (Stender et al. 2020).
pulmonary minute ventilation, and heart rate during While considered to be a homogenous ‘organ system,’
graded exercise. In a PubMed (National Library of muscle is in fact a heterogeneous tissue containing
Science) search on 20th October, 2020 the term ‘anaerobic different types of muscle fibres, and circulatory and
threshold’ elicited 5902 citations. In their review, Poole connective tissue networks, all with different metabolic
et al. explain that while the basic tenant of the AT (i.e. characteristics (Barnard et al. 1970, 1971). Postural
oxygen-limited metabolism) in exercise is no longer muscles (e.g. soleus, erector spinae) are alternatively
acceptable, use of the ‘gas exchange threshold’ (GET) termed intermediate (i.e. pink, not red or white), or Type
in graded exercise possesses significant importance I fibres. In many species deep vastus and lateral gastric are
for cardio-pulmonary and other types of evaluation in bright red and termed red or Type IIA fibres. In contrast
patients (Poole et al. 2021). white, fast twitch fibres are termed Type IIX (in humans)
In sum, intracellular lactate disposal is accomplished or IIB (in rodents). Hence, results of the above-cited
by a transport mechanism for direct mitochondrial studies on the glucose paradox are complimented by
uptake and oxidation that involves a mLOC which results of studies showing greater postprandial perfusion
probably works in parallel with malate-aspartate, and glucose uptake in muscles containing predominantly
glycerol-phosphate and other shuttles to balance cell oxidative Type I and IIA fibres than in muscles containing
redox and manage the lactate load from glycolysis under predominantly Type IIB/X fibres (James et al. 1985, 1986).
normal and stressful conditions (Brooks, 2020c). Redundancy of terminology in science is confusing, but
the glucose paradox can also be thought of as a post-
prandial lactate shuttle.
Contributions from studies of postprandial
metabolism: the glucose paradox (indirect pathway
of liver glycogen synthesis) and the postprandial Phasic lactate shuttling: lessons from the heart. Tracer
studies allow us to know that the heart produces lactate
lactate shuttle
even as it acts as a lactate sink displaying net uptake
The glucose paradox. Studies of postprandial glucose from the arterial circulation (Gertz et al. 1988; Bergman
metabolism on rodent models and humans show what has et al. 2009b). Assessing cardiac metabolism in humans is
been termed the ‘glucose paradox,’ or ‘indirect pathway difficult at best and is complicated by the cardiac cycle.
of hepatic glycogen synthesis’ (Foster, 1984). This concept If it is true that even in a healthy heart systole interrupts
states that dietary glucose released into the hepatic portal coronary blood flow, especially in the endocardium, then
vein initially bypasses the liver and goes to the periphery, the presence of a phasic intra-cardiac lactate shuttle is
where glycolysis converts glucose to lactate that is sub- indicated. Assuming a resting heart rate of 60 bpm, then
sequently released into the central venous circulation and systole followed by diastole occurs once per second. This
taken up from the arterial circulation by liver for glycogen could mean that there occurs 200 ms of stopped flow
synthesis. This, paradoxical, ‘indirect’ pathway of hepatic during the isometric phase of systole which is powered

© 2021 The Authors. The Journal of Physiology published by John Wiley & Sons Ltd on behalf of The Physiological Society
14697793, 2022, 5, Downloaded from https://physoc.onlinelibrary.wiley.com/doi/10.1113/JP280955 by Schweizerische Akademie Der, Wiley Online Library on [10/11/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J Physiol 600.5 Lactate, phoenix risen 1235

Brain

Lac Gluc

Postprandial
mixed venous
(Pulmonary artery)

Lungs

Pulmonary vein
(Systemic arterial)
Heart
Lac Gluc
Lac Gluc

Coronary
Lac Gluc
sinus
Lac Gluc
Lac
Gluc Gut Glucose
Liver

Gastrointestinal
tract
Glycogen Lac

Lac

Lactate Glycogen + Glucose


Gluc
Integument and muscle

Figure 3. Illustration of lactate shuttling after consuming dietary carbohydrate: the postprandial period
(the postprandial lactate shuttle)
Depending on liver glycogen content some dietary glucose bypasses liver and enters the systemic circulation. From
there glucose is taken up by non-contracting muscles, particularly red and intermediate fibres (James et al. 1985,
1986). Glycolysis in these and other driver tissues results in lactate release into the central venous circulation
and uptake by the recipient liver from the arterial circulation. Paradoxically, this circuitous, ‘indirect pathway,’ is
preferred over glucose for hepatic glycogen synthesis (Foster, 1984). In contrast, glucose from dietary consumption
or digestion or carbohydrate in the GI tract can be taken up by liver on the first circulatory pass for ‘direct’ glycogen
synthesis. Again as indicated in Fig. 2, lactate released from driver cells and tissues supports cerebral metabolic
needs (Glenn et al. 2015a,b) and functions such as glutamatergic signalling (Pellerin & Magistretti, 1994). Similarly,
systemic lactate and fatty acids from digestion serve as an energy source for the heart (Bergman et al. 2009a,b).
Figures 2 and 3 illustrate the extent of lactate shuttling under diverse conditions.

© 2021 The Authors. The Journal of Physiology published by John Wiley & Sons Ltd on behalf of The Physiological Society
14697793, 2022, 5, Downloaded from https://physoc.onlinelibrary.wiley.com/doi/10.1113/JP280955 by Schweizerische Akademie Der, Wiley Online Library on [10/11/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1236 G. A. Brooks and others J Physiol 600.5

by glycolysis, followed by 800 ms of diastole for oxidative prandial splanchnic lactate release in humans following
recovery and lactate clearance. Then heart rate increases the ingestion of one carbohydrate energy source, fructose.
during exercise, and time for the T-P interval decreases, so In sum, like the event of physical exercise in which
also does the time for restoration of myocardial blood flow lactate plays a prominent role in energy substrate
and intra-cardiac oxidative metabolism decrease. Again, production and disposal, lactate also plays an important
as with the hypothesis of gut lactate shuttling, the pre- role in carbohydrate distribution and disposal after
sence of an intra-cardiac lactate shuttle would be difficult eating. Hence, whether after a meal when glycogen
to prove because even coronary sinus sampling would be storage is prominent, or during post-absorptive exercise
insufficient to determine phasic events. Hopefully in the when glycaemia is supported by hepatic glycogenolysis
future, hyperpolarized MRS (Park et al. 2015) or other and gluconeogenesis, lactate plays important roles in
technologies will allow for detection of lactate production, carbohydrate (CHO) energy substrate distribution
net uptake and oxidative disposal within a cardiac cycle. (Brooks, 2020b). The issue of GI lactate production is
For the present, perhaps the best evidence of a phasic, addressed further in the section on the microbiome
intra-cardiac lactate shuttle may come from the lactate below.
threshold/turn point field in which a steep rise in blood
lactate occurs at 90% of maximal heart rate (Hofmann Lower gut: the microbiome. The possibility of
et al. 1994). Even in the healthy hearts of athletes with participation of the gut microbiome in lactate shuttling
high heart rates, there simply may be inadequate time for and a gut-soma lactate shuttle has previously been
intra-myocardial lactate disposal plus a burden of systemic suggested (Brooks, 2018, 2020a), but we are in the early
blood lactate clearance. stages of discovery. Support for the idea is to be found
in diverse sources. From epidemiology, we know that
regular physical exercise is beneficial for reducing risks
Can the splanchnic bed be a source of lactate? of many common cancers including those of the colon
(Powell et al. 2018). Also, from nutrition science we know
Upper gut: the liver. Classically, the liver removes lactate
that pre- and pro-biotic dietary components favourably
from the circulation and converts it to glucose for release affect gut fermentation and health (Hill et al. 2014) and
into the systemic circulation (Cori & Cori, 1946), or data indicate relationships between microbiota and the
converts lactate to liver glycogen (Nilsson & Hultman, prevalence of insulin resistance and metabolic syndrome
1974). But, can the liver, or splanchnic bed as a whole (Vrieze et al. 2012). The concentration of lactate in
contribute lactate to the systemic circulation? Evidence for human faeces is relatively low (< 5 mm) because of
splanchnic lactate production is sparse. the presence of bacteria that convert lactate to butyrate
Hepatic release from dog liver under glucagon (Duncan et al. 2004). More recently, we have learned that
stimulation was not seen (Wasserman et al. 1984). physical exercise and physical fitness encourage diversity
However, after glucose ingestion in rats with indwelling of species including those that act to convert lactate to
portal vein catheters, a porto-peripheral lactate gradient butyrate (Estaki et al. 2016, 2020). Recently also, the pre-
was present, reflecting the production of lactate in or by sence of ‘performance-enhancing’ gut microbes, members
the intestine (Smadja et al. 1988). Unfortunately, because of the genus Veillonella, in the stools of marathon runners
studies on humans would involve hepatic portal vein (Scheiman et al. 2019) have been found. Unfortunately,
catheterization, an invasive procedure not known to be those investigators were unaware of the importance of
associated with any disease state, and hence probably lactate shuttling during exercise, and did not consider a
ethically unjustified, we know of no report of hepatic scenario in which the gut supplies lactate, a fermentation
or splanchnic net release during postprandial exercise. product that is exported via sodium-mediated mono-
However, because the phenomenon of splanchnic lactate carboxylate transporters (sMCT) (Coady et al. 2004;
release has not been observed, it doesn’t necessarily follow Teramae et al. 2010), thus supporting athletes’ efforts by
that the phenomenon fails to exist. fuelling working muscles as opposed to clearing lactate
In addressing the issue of upper gastrointestinal from the circulation.
(GI) lactate production in humans, consideration of
the carbohydrate energy form may be helpful. Using
combinations of glucose, fructose and lactate tracers to Lactate controls lipid metabolism: the role of lactate
evaluate the use of oral carbohydrate energy sources in
in metabolic flexibility and the crossover concept
sports drinks investigators in the Tappy lab (Lecoultre
et al. 2010; Theytaz et al. 2014) observed carbon Lactate from working muscle (Bergman et al. 1999b) or
atoms from an orally ingested fructose tracer appearing other tissues such as the integument (Johnson & Fusaro,
in the systemic circulation as labelled lactate. Hence, 1972) has short-term effects on lipid metabolism by down-
counter to classic thought, there is evidence for post- regulating both lipolysis and mitochondrial free fatty

© 2021 The Authors. The Journal of Physiology published by John Wiley & Sons Ltd on behalf of The Physiological Society
14697793, 2022, 5, Downloaded from https://physoc.onlinelibrary.wiley.com/doi/10.1113/JP280955 by Schweizerische Akademie Der, Wiley Online Library on [10/11/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J Physiol 600.5 Lactate, phoenix risen 1237

acid entry and oxidation. In contrast, chronic exercise Lactate shuttling and metabolic signalling
exposure, as with endurance training, improves capacities
The effects of increases in cell work and lactate production
to maintain glycaemia and lipid oxidation by improving
on redox, reactive oxygen species (ROS), allosteric
the ability to clear lactate by oxidation (Bergman et al.
binding, and histone lactylation have been recently
1999b) and gluconeogenesis (Bergman et al. 2000), as well
reviewed (Brooks, 2020a). Briefly, cell work leads to lactate
as acting as a pseudo-myokine (Takahashi et al. 2019)
production and changes in the cellular lactate/pyruvate
(vide infra). The role of lactate in affecting energy substrate
ratio (Henderson et al. 2007) that accompanies changes
partitioning in exercise and other conditions is imbedded
in the cellular NAD+ /NADH ratio and subsequent
in concepts of ‘crossover’ (Brooks & Mercier, 1994) and
metabolic and regulatory effects in driver and recipient
metabolic flexibility (Kelley et al. 1999; Goodpaster &
cells (vide supra). Importantly also, additional lactate
Sparks, 2017; San-Millan & Brooks, 2018). In the case of
signalling mechanisms have been revealed.
lactate and lipolysis in adipose tissue, during hard exercise
inverse relationships between blood lactate and plasma
free fatty acid concentration [FFA] in humans (Brooks Cell redox. As described above, muscle contraction
& Mercier, 1994) and other mammals (Issekutz & Miller, causes major changes in the L/P and NADH/NAD+ ratios.
1962; Rodahl et al. 1964) have long been recognized. Also, In driver cells that change allows glycolysis to proceed
lactate infusion into running dogs caused the circulating via providing NAD+ for GAPDH, but also by reducing
[FFA] to decline (Issekutz & Miller, 1962; Gold et al. 1963; pyruvate to lactate in the cytosol, which creates a driving
Miller et al. 1964). In those investigations an effect of force for mitochondrial substrate oxidation. In contrast,
lactate on circulating [FFA] could be clearly observed, in recipient cells uptake of lactate raises the NADH/NAD+
but whether the mechanisms involved hydrogen ions or ratio, thus downregulating glycolysis. This preferential use
lactate anions was not assessed. of lactate over endogenous cellular energy sources reduces
The mechanism by which lactataemia suppresses oxidation of glucose in muscle (Miller et al. 2002a,b) and
circulating FFAs is now known to be due to suppression of heart (Wisneski et al. 1987; Gertz et al. 1988; Bergman
adipose lipolysis by lactate binding to hydroxycarboxylic et al. 2009a).
acid receptor 1 (HCAR-1), formerly known as G-protein Moreover, aside from the major effects of muscle
coupled receptor 81 (GPR-81), independently of changes contraction and glycolysis on energy substrate
in pH (Cai et al. 2008; Ge et al. 2008; Liu et al. 2009; Ahmed partitioning, glycolysis leading to lactate production
et al. 2010). Signalling effects of lactate on HCAR-1 are and resulting change in cytosolic NADH/NAD+ also
further discussed below. effects the status of other redox couples such as the ratio
of reduced to oxidized glutathione (GSH/GSSG) (Gohil
Lactate and pyruvate as inhibitors of mitochondrial et al. 1988; Viguie et al. 1993).
β-oxidation. When glycolysis is accelerated during
muscle contraction, lactate (L) and pyruvate (P) ROS. Lactate can raise cellular production of ROS via
concentrations rise and increase the L/P ratio, (Bergman mitochondrial respiration (Powers et al. 1999; Passarella
et al. 1999b; Henderson et al. 2004) reducing cytosolic et al. 2008) and non-enzymatically via lactate-iron inter-
redox (Brooks, 2020a). At rest, the L/P ratio in muscle actions that are capable of generating ROS (Wagner et al.
and venous effluent from a muscle bed approximates 10, 2004). Another way in which lactate can affect ROS
but the ratio rises more than an order of magnitude (≥ production is via generation of hydrogen peroxide (H2 O2 )
30×) during moderate intensity exercise (Henderson (Passarella et al. 2008). The latter mechanism involves
et al. 2004). By mass action the monocarboxylate generation of H2 O2 via a flavine-dependent lactate oxidase
pair floods into the mitochondrial reticulum (Saddik located in the mitochondrial intermembrane space (de
et al. 1993; Brooks et al. 1999b; Passarella et al. 2008), Bari et al. 2010).
giving rise to acetyl-CoA and, thereby, malonyl-CoA
formation. The rise in malonyl-CoA inhibits the entry of Sirtuins. Sirtuins are deacetylases regulated by the
activated FFAs into the mitochondrial matrix by inhibiting equilibrium between nicotinamide (NAM) and NAD+ .
carnitine-palmitoyl transferase-1 (CPT1) (McGarry et al. Sirtuin activation is accomplished via changes in cell redox
1977; Saddik et al. 1993). As well, the effect of glycolysis (i.e. the NAD+ /NADH) through the concentration of
driving an accumulation of acetyl-CoA downregulates NAM and the activity of the enzyme NAM phosphoribosyl
β-ketothiolase, the terminal and rate-limiting enzyme transferase (Nampt). Many investigators are concerned
of the mitochondrial β-oxidation pathway. Hence, by with how the subtle changes in cell redox affect cell
mass action, allosteric binding and effects on cell redox, homeostasis as occur in apoptosis, inflammation, and
lactate acts to prevent entry of activated fatty acids into other processes. However, more obvious are changes
the matrix of the mitochondrial reticulum, thus limiting in cell redox with increments in cell work as occur in
β-oxidation of fatty acid derivatives. exercise. At present, data are lacking on the change

© 2021 The Authors. The Journal of Physiology published by John Wiley & Sons Ltd on behalf of The Physiological Society
14697793, 2022, 5, Downloaded from https://physoc.onlinelibrary.wiley.com/doi/10.1113/JP280955 by Schweizerische Akademie Der, Wiley Online Library on [10/11/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1238 G. A. Brooks and others J Physiol 600.5

in Nampt activity in working muscle or non-working lactate transporter 2 (Mrs2). Investigators observed
recipient tissues in which changes in NAD+ and NADH that a surge in mitochondrial Mg2+ uptake promotes
concentrations are likely to affect sirtuin regulation in inflammation and organ failure by interfering with
vivo. mitochondrial energetics. In contrast, suppression
of the mMg2+ surge alleviates inflammation-induced
Hydroxycarboxylic acid receptor 1 (HCAR-1). The multi-organ failure. Together, these findings reveal that
mechanism by which lactataemia suppresses circulating lactate mobilizes intracellular Mg2+ and links the mMg2+
FFAs is now known to be due to suppression of adipose transport machinery with major metabolic feedback
lipolysis (Ahmed et al. 2010). Independently of pH, circuits and mitochondrial bioenergetics (Daw et al.
lactate inhibits lipolysis in fat cells through activation of 2020). Hence, on the basis of the work by Daw et al., it is
HCAR-1 (vide supra). In studies of mouse, rat, and human apparent that, in addition to mass action, redox control,
adipocytes, HCAR-1 appears to act as a lactate sensor and ROS generation, lactate may affect mitochondrial
with the inhibitory effect on lipolysis operating through energetics in other ways such as by Mg2+ release from
cyclic AMP (cAMP) and cAMP response element binding endoplasmic reticulum.
(CREB) (Ahmed et al. 2010; Bergersen, 2015). The purported effects of lactate signalling HCAR-1,
TGF-β2, Nampt, mMg2+ and lactylation of histones
observed in rodent and cell models await validation in
Transforming growth factor beta 2 (TGF-β2). The
humans. However, for the present, it is certain that lactate
effects of lactate on inter-organ signalling and dual
both inhibits lipolysis and mitochondrial FFA oxidation
or multiple effects of lactate signalling is illustrated by the
in the short-term and in the long-term stimulates
recent discovery that transforming growth factor beta 2
mitochondrial biogenesis and glucose tolerance and lipid
(TGF-β2) is secreted from adipose tissue in response to
oxidation in in vivo (Takahashi et al. 2019). Therefore, the
lactate released from working muscle (Takahashi et al.
new results are encouraging as the signalling patterns are
2019). Because TGF-β2 improved glucose tolerance
consistent with what is known about the roles of lactate in
in mice, the authors concluded that exercise training
physiology and metabolism.
improves metabolic regulation through an inter-organ
(adipose to liver) communication via a ‘lactate-TGF-β2
signalling cycle.’ If validated on studies of human subjects,
A plethora of lactate shuttles and diverse roles in
TGF-β2 could be classified as an ‘adipokine,’ and lactate
physiology and metabolism
could be thought of as a ‘pseudo myokine’ and the value
of enhancing lactate clearance during exercise by end- Since recognition of the presence of lactate shuttling
urance training further emphasized (Donovan et al. 1983; within and among various cells, tissues and organs such
Bergman et al. 1999b; Messonnier et al. 2013). In this as muscle, heart and liver (Brooks, 1984, 1985, 1986, 2002,
context both short- and long term-effects are illustrated 2018), the concept has been extended to include other
because lactate is known to suppress lipolysis in adipose cells, tissues and organs such as brain (Pellerin et al. 1998;
via HCAR-1 and CREB (vide supra). Liu et al. 2017), lung (Johnson et al. 2011, 2012), sperm
(Storey & Kayne, 1977; Boussouar & Benahmed, 2004),
Lactylation of histones. Regulation of gene expression adipose tissue (Cai et al. 2008; Liu et al. 2009; Ahmed et al.
by lactylation of 28 lysine residues on histones has 2010), and peroxisomes (McClelland et al. 2003). The pre-
been demonstrated (Zhang et al. 2019). The addition sences of multiple lactate shuttles with diverse functions in
of lactate in addition to phosphate, methyl and acetyl physiology has been recently summarized (Brooks, 2009),
tags to histones is yet another epigenetic way by which but for completeness the following are mentioned.
the genome and intermediary metabolism are regulated.
As noted above, lactate release from driver cells into The astrocyte-neuron lactate shuttle and brain
the circulation, as occurs in physical exercise and other functioning. Lactate shuttling between astrocytes and
stressful conditions, has the potential for epigenetic neurons linked to glutamatergic signalling was recognized
modifications in diverse cells around the body during and by Pellerin, Magistretti and colleagues who used the term
after physical exercise. ‘astrocyte-neuron lactate shuttle’ (ANLS) (Magistretti
et al. 1999). Since its introduction and further exposition
Lactate, endoplasmic reticulum Mg2± release and (Pellerin & Magistretti, 2012), the concept has remained
mitochondrial function. Recently, Daw et al. in the controversial (Patel et al. 2014). However, subsequent
Madesh lab (Daw et al. 2020) showed that lactate is an studies show wide expression of brain MCTs (Pellerin
activator of Mg2+ release from endoplasmic reticulum et al. 2005) and support the presence of an ANLS.
(ER). ER Mg2+ release facilitates mitochondrial Mg2+ While cerebral lactate flux may be related to
(mMg2+ ) uptake, a process facilitated by magnesium glutamatergic signalling (Pellerin & Magistretti, 1994),

© 2021 The Authors. The Journal of Physiology published by John Wiley & Sons Ltd on behalf of The Physiological Society
14697793, 2022, 5, Downloaded from https://physoc.onlinelibrary.wiley.com/doi/10.1113/JP280955 by Schweizerische Akademie Der, Wiley Online Library on [10/11/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J Physiol 600.5 Lactate, phoenix risen 1239

there are other processes that are related to lactate downregulates Toll like receptor induction of the pyrin
shuttling. For instance, in health and after injury (Glenn domain-containing protein 3 (NLRP3) inflammasome
et al. 2015a), as well as during physical exercise (van Hall and production of IL1beta, via Arrestin beta 2 (ARRβ2)
et al. 2009; Hashimoto et al. 2018), the brain takes up and and HCAR-1, was the mechanism by which lactate
oxidizes lactate from the systemic circulation. In such suppressed inflammation in patients with acute organ
cases, the lactate shuttle concept holds true and explains injury. Because of the above, the science and translation of
the exchange between muscle lactate driver and brain lactate shuttling in inflammation needs further attention.
recipient cells.
Characteristics of cellular lactate uptake such as
The peroxisomal lactate shuttle and β-oxidation.
stereo-specificity, concentration and pH dependence,
Peroxisomes are cellular organelles found in virtually
saturation, trans-stimulation and inhibition by
all eukaryotic cells that are involved in catabolism of
competitive and non-competitive inhibitors were
substances such as very long chain (i.e. C22 and longer)
recognized early on (Watt et al. 1988; Roth & Brooks,
fatty acids, branched-chain fatty acids, d-amino acids,
1990a,b; Brown & Brooks, 1994). Since MCT iso-
polyamines, and reactive oxygen species such as hydrogen
forms were cloned and sequenced (Garcia et al. 1994,
peroxide (H2 O2 ) (Gladden, 2004). While it was known
1995; Price et al. 1998) and then studied in vitro, it
that β-oxidation of very long-chain fatty acids occurred
has been appreciated that MCTs facilitate movement
in mammalian peroxisomes (Lazarow & De Duve,
of monocarboxylates other than lactate (e.g. pyruvate,
1976), in the absence of enzyme systems such as exist
β-hydroxybutyrate and acetoacetate) across biological
in the mitochondrial matrix, it was not understood how
membranes. However, for at least two sets of reasons,
β-oxidation could occur in peroxisomes. Key findings
these other monocarboxylates are of lesser physiological
were those of Baumgart et al. (1996), who showed the
importance in terms of carbon-energy transfer. First,
presence of peroxisomal LDH isoforms, and McClelland
other monocarboxylates do not compete as well as lactate
et al. (2003), who confirmed the presence of peroxisome
for exchange by MCTs because of Michaelis-Menten
LDH and further demonstrated that rat liver peroxisomal
kinetic properties (Roth & Brooks, 1990a,b). Secondly,
membranes contained MCT1 and MCT2. From there,
physiological levels of lactate exchange competitors are far
it was possible to hypothesize that peroxisomal redox
less abundant (Zinker et al. 1990; Bergman et al. 1999b;
control necessary to support β-oxidation involved a
Henderson et al. 2004). Importantly, from a physio-
lactate-pyruvate shuttle. Discovery of the peroxisomal
logical standpoint, MCTs do not create the conditions for
lactate shuttle involving pyruvate-lactate conversion
lactate exchange; rather, MCTs facilitate carbon exchange
linked to changes in the NADH/NAD+ redox couple
between loci of driver processes such as glycolysis and
emphasizes the critical importance of redox changes in all
glycogenolysis and recipient loci where processes such
forms of lactate shuttles.
as mitochondrial respiration and gluconeogenesis are
responsible for lactate consumption (Brooks, 1984, 2002).
Hence, it is appropriate to recognize the importance of Spermatogenic and Sertoli-germ cell lactate shuttles.
MCTs in cerebral lactate shuttling in cognition, learning, It has been known for decades that mammalian
memory and other executive functions (Holloway et al. spermatozoa use lactate as an aerobic energy source
2007; Suzuki et al. 2011; Steinman et al. 2016; Hashimoto (Storey & Kayne, 1977). Noteworthy also is that in their
et al. 2018; El Hayek et al. 2019). seminal papers on the discovery of MCT1 and MCT2
Garcia et al. (1994, 1995) observed high expression of
Lactate and inflammation. Other than acknowledging MCT1 in sperm heads as they entered the epididymis,
its complexity, the role of lactate in promoting or followed by lower expression of MCT1 expression as
suppressing inflammation is incompletely understood at sperm coursed through the epididymis. It was also noted
present. As mentioned above, Daw et al. have provided that lactate stimulates respiration in ejaculated bovine
evidence that lactate-induced Mg2+ release from ER sperm (Halangk et al. 1985), and further that lactate
affects mitochondrial energetics and could be involved in maintains bovine sperm motility when maintained ex
cell death and organ systems failure (Daw et al. 2020). vivo (Inskeep & Hammerstedt, 1985). By analogy, just as
In contrast, others have provided evidence that -lactate fast twitch-glycolytic driver muscle fibres fuel adjacent
containing solutions have efficacy as resuscitation fluids slow twitch-oxidative muscle fibres, Sertoli cells in testes
for use in a variety of conditions including metabolic secrete lactate to fuel sperm motility in vivo. In effect
acidosis (Brooks, 2018), acute pancreatitis (Wu et al. 2011; then, the Sertoli-sperm cell relationship is the primal
Hoque et al. 2014), hepatitis (Hoque et al. 2014), sepsis demonstration of cell-cell lactate shuttling. In sperm, the
(Garcia-Alvarez et al. 2014b) and dengue fever (Somasetia mitochondrial reticulum is elaborate, large and spiral
et al. 2014). In terms of mechanism, Hoque and colleagues shaped, located at the midpiece (Storey & Kayne, 1977)
(Hoque et al. 2014) found that lactate binding to HCAR-1 at the base of the sperm head. The ability of sperm

© 2021 The Authors. The Journal of Physiology published by John Wiley & Sons Ltd on behalf of The Physiological Society
14697793, 2022, 5, Downloaded from https://physoc.onlinelibrary.wiley.com/doi/10.1113/JP280955 by Schweizerische Akademie Der, Wiley Online Library on [10/11/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1240 G. A. Brooks and others J Physiol 600.5

mitochondria to oxidize exogenously supplied lactate the TCA cycle (Bertocci & Lujan, 1999). Also, using
31
(Jones, 1997) is analogous to capacities for mitochondrial P MRS to study muscle metabolism in patients with
lactate oxidation in other tissues (Baba & Sharma, 1971; McArdle disease, a knock-out experiment of Nature
Kline et al. 1986; Brandt et al. 1987; Brooks et al. 1999a; involving phosphofructokinase (PFK) deficiency, lactate
De Bari et al. 2004). infusion was shown to improve muscle force output and
energy status in terms of higher phosphocreatine (PCr)
Not a few, but many observers and lower adenosine diphosphate (ADP) and inorganic
phosphate (Pi ) levels (Bertocci et al. 1993). Thus, by
Readers may note that the contemporary literature on means of exogenous lactate infusion investigators were
the role of lactate in exercise physiology and metabolism able to bypass the enzymatic block at PFK. Subsequently,
comes from only a few laboratories (Brooks, 2018; in studies using vascular infusion of [1-13 C]lactate
Ferguson et al. 2018), but such is not the case if a wider and arterial-venous difference measurements across
perspective is taken, when it is apparent that support for resting and exercising legs of McArdle disease patients,
the biological roles of lactate come from diverse scientific investigators concluded that ‘lactate formation is
and clinical fields in which solutions to many problems are mandatory for muscle energy generation during exercise.’
addressed utilizing a variety of technologies. In addition In part those results could have been predicted based
to the above identified lactate shuttles, the importance on results of studies on electrically stimulated isolated
of lactate in contemporary biology is illustrated in the rat muscles in which lactate addition to the incubation
following examples. bath helped maintain cell membrane chemical-electrical
gradients and ward off fatigue (de Paoli et al. 2007).
The glycogen shunt. Shulman and colleagues used
proton (1 H) and 13 C magnetic resonance spectroscopy Lactate metabolism in injuries and illnesses. This issue
(MRS) to explain the rapid rate of metabolic transitions has been addressed in recent reviews (Brooks, 2018,
in brain after stimulation; consequently, a glycogen shunt 2020a), but there appear to be both positive and negative
was proposed (Shulman et al. 2001). By this model, aspects to lactate metabolism in illnesses and injuries
shunting a portion of glucose from the circulation into (Table 1).
glycogen with subsequent glycogenolysis provides for
lactate production by two parallel pathways, traditional Positive effects – lactate or supplemental lactate
glycolysis from glucose and glycolysis from glycogen.
treatment
Although ATP energy from the glycogen shunt is
energetically less efficient than glycolysis from glucose, Lactate treatment can be involved in resuscitation
the shunt allows cell energy storage and glial energy to following environmental stresses (dehydration and
be provided in milliseconds for rapid neurotransmitter acidosis), inflammation (hepatitis and pancreatitis),
release when needed. Enlarging on their work on cerebral infectious (Dengue and sepsis) and non-infectious
energy needs, Shulman and colleagues expanded the diseases (hypoglycaemia) and traumatic conditions
model to include the energetics of working skeletal (brain injury, myocardial infarction, burns and wound
muscle (Shulman & Rothman, 2001) The hypothesis is healing).
a forceful reminder of the immediate role of glycogen
over glucose in supplying energy via glycolysis, leading Negative effects – overabundance of lactate
to lactate production in rodents (Brooks & Donovan,
production and accumulation
1983; Donovan et al. 1983) and humans (Bergman et al.
1999a,b). The idea of a glycogen shunt is also a reminder Cancer. Seminal studies have shown that lactate
that everything, including glycogen, turns over during production and accumulation occur in cancer cells
exercise (Azevedo et al. 1998) and that reduced glycogen incubated under fully aerobic condition (Warburg &
depletion in exercising muscle is due, in part, to a Minami, 1923, 1927). Subsequently, accelerated glycolysis
reduction in turnover. leading to lactate production in cancer cells has been
implicated in many stages in carcinogenesis (San-Millan
Muscle lactate disposal and energy status. Bertocci, & Brooks, 2017). More recently, blocking of MCTs in
Haller and colleagues have used MRS and 13 C-lactate driver (glycolytic) and oxidative (recipient) tumour
tracers to evaluate the role of lactate in muscle energy cells can minimize lactate exchange, in effect killing
transduction. By tracking the conversion of 13 C-lactate both cell types by pickling the drivers and starving the
into glutamate in a rat muscle preparation, a TCA cycle recipients, thus arresting tumour development (Semenza,
intermediate in equilibrium with α-ketoglutarate, it was 2008; Sonveaux et al. 2008). To date, however, the use
confirmed that lactate is readily oxidized by skeletal of pharmacological agents to block MCTs in cancer is
muscle during rest and contraction, and directly enters contraindicated because of the ubiquitous expression of

© 2021 The Authors. The Journal of Physiology published by John Wiley & Sons Ltd on behalf of The Physiological Society
14697793, 2022, 5, Downloaded from https://physoc.onlinelibrary.wiley.com/doi/10.1113/JP280955 by Schweizerische Akademie Der, Wiley Online Library on [10/11/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J Physiol 600.5 Lactate, phoenix risen 1241

Table 1. Potential for lactate treatment for illness and injury (from Brooks, 2020a)

Resuscitation (fluid, electrolytes, energy) (Azevedo et al. 2007; Garcia-Alvarez et al. 2014a; Marik & Bellomo, 2016)
Acidosis (exogenous lactate infusion has an alkalotic effect) (Miller et al. 2005; Wu et al. 2011; Marik & Bellomo, 2016)
Regulation of glycaemia (lactate is the major gluconeogenesis (GNG) precursor) (Meyer et al. 1998, 2002; Gerich et al. 2001; Marik,
2009)
Traumatic brain injury (lactate is brain fuel and anti-inflammatory) (Glenn et al. 2015a)
Inflammation (via GPR81 binding down stream signalling lactate inhibit the inflammasome) (Hoque et al. 2014)
Acute pancreatitis and hepatitis (lactate is an energy substrate, a GNG precursor and anti-inflammatory agent) (Hoque et al. 2014)
Myocardial infarction, cardiac surgery and acute heart failure (lactate is heart fuel) (Shapiro et al. 2005; Bergman et al. 2009b)
Burns (lactate is an energy substrate, a GNG precursor and anti-inflammatory agent) (Spitzer, 1979)
Sepsis (lactate incorporation in resuscitation fluids can support maintenance of blood pressure and circulation, and help deliver
antibiotics, as well as being an energy substrate, a GNG precursor and have an anti-inflammatory effect) (Garcia et al. 1995; Marik
& Bellomo, 2016)
Dengue (lactate is an energy substrate, a GNG precursor and anti-inflammatory agent) (Wu et al. 2011; Somasetia et al. 2014)
Cognition (lactate readily crosses the blood-brain barrier, fuels neurons and stimulates secretion of brain-derived neurotrophic
factor (BDNF), improves executive function and memory) (Rice et al. 2002; Holloway et al. 2007; Hashimoto et al. 2018)
Wound healing (Hunt et al. 2007) and muscle regeneration after injury (Tsukamoto et al. 2018; Ohno et al. 2019).

MCTs in most cell types and negative consequences for central role in physiology. Consider for instance several
the host. Additionally, blocking or deleting HCAR-1 leads of the topics discussed above that have been studied
to a reduced transcription of cancer-regulated genes, thus separately, but seldom investigated in an integrated
diminishing cancer cell proliferation in animal and cell fashion. Seemingly, there is need for a more integrative
culture models (Brown et al. 2020; Xie et al. 2020). At pre- approach to develop models of physiological organization.
sent there are no definitive data indicating that aberrant Seemingly also, we may learn from, or contribute to,
lactate metabolism causes cancer, but it has been observed research in other fields of science and medicine. Some
that lactate accumulation is associated with upregulation examples follow.
of cancer promoting genes and downregulation of cancer
suppressor genes (San-Millan et al. 2020).
Nutrition, digestion, muscle physiology and the micro-
Errors in the ability to transport or exclude lactate may
biome. For instance, consider that the consumption of
have serious consequences. For instance, embryologic
deletion of the lactate transporters (monocarboxylate dietary CHO elicits neuroendocrine signals to splanchnic
transporters, MCTs) is lethal. For glucoregulation when tissues and brain. In the upper GI tract, entry of
blood lactate is elevated as in exercise or other conditions, glucose for CHO digestion is disposed of via direct and
MCTs must be excluded from insertion into pancreatic indirect pathways as described in the glucose paradox.
β-cell plasma membranes (Pullen et al. 2010; Rutter Glucose released into the systemic circulation via the
et al. 2015). While MCTs are present on plasma and hepatic portal vein is taken up by red muscle where
mitochondrial membranes of most cells and tissues, glycogen synthesis and glycolysis are stimulated. Sub-
exclusion of MCTs from pancreatic β-cell plasma sequently, in the GI tract glucagon-like peptide-1 (GLP-1)
membranes keeps extracellular lactate from entering and glucose-dependent insulinotropic polypeptide (GIP)
and affecting intracellular redox, thus interfering with are released from enteroendocrine cells in response to
glucose sensing and insulin secretion (Bender et al. 2006). nutrient ingestion and the presence of glucose and
Noteworthy in this regard is that individuals experiencing lactate. GLP-1 receptors (GLP-1Rs) on enteric neurons,
failed suppression of pancreatic β-cell MCT expression including intrinsic afferent neurons, and extrinsic spinal
become hypoglycaemic during hard exercise leading to and vagal sensory afferents, have potentially numerous
lactataemia because the combination of high insulin and effects including enhancement of insulin secretion and
increased glucose disposal through metabolism causes mediation of satiety signalling, via effects on the hypo-
profound hypoglycaemia (Otonkoski et al. 2007). thalamic arcuate and paraventricular nuclei.
It goes without citation that short- and long-term
nutrition and blood glucose regulation is heavily tied to
Lactate shuttling into the future: unresolved issues hunger and satiety signalling. The biochemistry behind
appetite regulation is a complicated and active area of
and future directions
research (Gale et al. 2004; Murphy & Bloom, 2006).
The more lactate shuttling is studied, the more is revealed Suffice it to state that in the arcuate nucleus of the hypo-
about the expansive nature of lactate metabolism and its thalamus effects of the appetite stimulating (feeding)

© 2021 The Authors. The Journal of Physiology published by John Wiley & Sons Ltd on behalf of The Physiological Society
14697793, 2022, 5, Downloaded from https://physoc.onlinelibrary.wiley.com/doi/10.1113/JP280955 by Schweizerische Akademie Der, Wiley Online Library on [10/11/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1242 G. A. Brooks and others J Physiol 600.5

(neuropeptide y (NPY)) centre are balanced Jones and colleagues observed that metabolically
against those of the appetite inhibitory (satiation) inflexible subjects had an increased fasting plasma lactate
[pro-opio-melanocortin (POMC)] centre. Hormones compared to aged matched controls (Jones et al. 2019).
that inform the hypothalamic centres that regulate Similarly, San-Millán and Brooks observed that diabetic
appetite include insulin, ghrelin, leptin, peptide YY and obese subjects showed increased circulating lactate
(PYY), glucagon like peptide-1 (GLP-1) and others and decreased lipid oxidation during rest and exercise
(Ghosal et al. 2013). Results of classic studies relating compared to age matched lean and highly trained athletes
physical activity level and diet (Mayer et al. 1956), as well (San-Millan & Brooks, 2018). Further research is needed
as practical experience, indicate that maximal exercise has to determine the mechanisms by which lactataemia
an appetite inhibiting effect. For instance, few are hungry limits metabolic flexibility in sedentary, obese and older
immediately after maximal exercise to exhaustion. The individuals. Elaboration of those mechanisms could
suppressive effect of lactate on appetite (Schmid et al. lead to behavioural and pharmacological treatments for
2008; Schultes et al. 2012; McCarthy et al. 2020) is metabolic inflexibility in illnesses and ageing.
consistent with data showing that lactataemia suppresses
ghrelin secretion (Islam et al. 2017; Vanderheyden
Epigenetic modifications by lactylation of histones and
et al. 2020). The ghrelin receptor (growth hormone
other potential mechanisms. Glis family zinc finger
secretagogue receptor (GHSR-1α)) is a G-protein coupled
(Glis1) is a transcription factor known to be involved in
receptor expressed throughout both the stomach and GI
cell reprogramming, such as transformation of somatic
tract. Recently, it was found that lactate, short chain fatty
to pluripotent cells. In somatic cells Glis1 binds to
acids and other bacterial excretions in the GI tract are
glycolysis genes to open them and promote transcription
able to attenuate ghrelin mediated signalling through
(Li et al. 2020). This activates glycolysis, without affecting
the GHSR-1α (Torres-Fuentes et al. 2019). Hence, in
OXPHOS, thus increasing lactate and acetyl-CoA levels
combination with lactate produced by gut microbiota,
that result in an increase in histone lactylation and
the high blood lactate of exercise can enter the bowel
acetylation. Further, histone lactylation in some (Pan Kla
via sodium-mediated monocarboxylate transporters
and H3K18la), but not all (H3K9me3 and H3K27me3)
(sMCT) and attenuate ghrelin receptor signalling, perhaps
cultured tumour cells increased substantially after Glis1
revealing how hard exercise attenuates hunger.
overexpression and decreased after Glis1 knockdown.
Continuing with the course of chyme down the
Taken together, the results offer the possibility that Glis1
GI tract, microbiota give rise to lactate that is either
modulates histone acetylation and lactylation during cell
converted to butyrate, released into the systemic
reprogramming. Given these hints in the literature, it
circulation, or excreted (vide supra). However, it is
may be worthwhile to learn from progress in regenerative
clear that more research is needed to better understand
medicine and cancer biology to expand our knowledge of
how upper and lower bowl processes are regulated and
the effects of diet and exercise on histone modulation in
integrated in vivo. Consider for instance, the physiology
disease and ageing processes.
of someone consuming a sports drink during exercise.
Notwithstanding the organoleptic effects of cool beverage
consumption (Jeukendrup & Chambers, 2010), we would Fertilization in vivo and in vitro. It is recognized by
benefit from the development of an overarching model workers in some fields of science (e.g. sepsis: Marik
of nutrient consumption and disposal during rest and & Bellomo, 2016) that progress in understanding
exercise. the biological roles of lactate is attributable to the
contributions of exercise physiology summarized above
and previously (Brooks, 2018; Ferguson et al. 2018).
Clearly all can benefit from greater understanding,
Metabolic flexibility and lactataemia
including those in the fields of in vitro fertilization
Lactataemia limits lipid oxidation by blocking lipolysis (IVF) in which the classic antagonism between glycolytic
and oxidation of FFAs (vide supra). From the fields of and oxidative metabolism is on display. In describing
obesity, diabetes and exercise physiology research come lactate shuttles above, the relationship between lactate
the concepts of metabolic flexibility (Kelley et al. 1999) and secreting Sertoli and sperm cells was described as the
crossover (Brooks & Mercier, 1994). Metabolic flexibility ‘primal demonstration’ of cell-cell lactate shuttling. The
describes the ability of individuals to switch between key ‘good thing, bad thing’ dichotomy of lactate carries
fuel energy substrates in response to changing physio- over to the field of IVF, which is typically conducted
logical conditions such as obesity and diabetes. The cross- on blastocysts in culture media exposed to air, in
over concept additionally considers the effect of metabolic which the PO2 is 5× greater than in vivo. Therefore,
rate on energy substrate partitioning. With regard to an the consequences of hyperoxic ROS generation are a
effect of lactataemia on energy substrate partitioning, major concern because of the potential effects of ROS

© 2021 The Authors. The Journal of Physiology published by John Wiley & Sons Ltd on behalf of The Physiological Society
14697793, 2022, 5, Downloaded from https://physoc.onlinelibrary.wiley.com/doi/10.1113/JP280955 by Schweizerische Akademie Der, Wiley Online Library on [10/11/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J Physiol 600.5 Lactate, phoenix risen 1243

on mitochondrial morphology and function in the homeostasis by oxidative phosphorylation of ADP and
embryos (Belli et al. 2020). Similarly, because a disrupted creatine (Hashimoto et al. 2006). Hence, mitochondrial
microenvironment can lead to problems in postnatal respiration creates the physiological sink for lactate
development (Feuer et al. 2017), high lactate and low pH disposal in vivo. As research progresses important facets
have been matters of concern. Much like the Warburg of lactate shuttling are becoming recognized with regard
effect of high rates of aerobic glycolysis in cancer cells to cell signalling and metabolic regulation (Brooks,
and working muscles, mammalian blastocysts exhibit 2020a). In diverse tissues lactate acts by mass action, cell
high capacities for aerobic glycolysis and lactate secretion redox regulation, ROS generation, allosteric binding and
that may create a favourable microenvironment for lactylation of histones. By inhibiting lipolysis in adipose
uterine implantation and invasion. Beyond providing an tissue, via HCAR-1 binding and CREB activation, and
energy supply, glycolysis in blastocysts creates a micro- muscle mitochondrial fatty acid uptake, via malonyl-CoA
environment around the embryo to contribute to the and CPT1, lactate controls lipid oxidation and overall
disaggregation of uterine tissues to facilitate trophoblast energy substrate partitioning. Repeated lactate exposure
invasion. Further, as in wound healing, it may be that from regular exercise results in adaptive processes
lactate acts as a signalling molecule to elicit vascular end- such as mitochondrial biogenesis and other healthful
othelial growth factor (VEGF) recruitment from uterine circulatory and neurological characteristics such as
cells to promote angiogenesis in the uterus. It has also improved physical work capacity, metabolic flexibility
been suggested that the region of high lactate created by (Brooks, 2018), memory and cognition (Suzuki et al.
the blastocyst modulates the activity of the local immune 2011; El Hayek et al. 2019). The importance of lactate and
response, thereby promoting immune tolerance (Gardner, lactate shuttling in healthful living is further emphasized
2015). Moreover, lactate produced during delivery may when lactate signalling and shuttling are dysregulated
reduce post-partum uterine inflammation via the lactate as occur in cancer (San-Millan & Brooks, 2017) and
receptor HCAR-1 (Madaan et al. 2017). As discussed other conditions such as following traumatic brain injury
above, several sets of independent studies indicate a role (Glenn et al. 2015b), metabolic syndrome (Brooks,
of lactate in mitigating inflammation. Because of the role 2018; San-Millan & Brooks, 2018), inappropriate insulin
of inflammation in chronic diseases, it seems that focused signalling (Rutter et al. 2015) and sepsis (Garcia-Alvarez
efforts on the role of lactate in reducing inflammation et al. 2014a). While much has been done to determine
may be of importance. energetic regulation in steady and transient states of
metabolism, the importance of measuring the dynamics
of metabolism beyond stagnant use of ‘metabolomics’, by
assessing physiological, environmental and age-related
Summary
effects on the turnover rates of lactate, glucose, fatty and
Time is overdue to turn the page on understanding amino acids as well as structural and metabolic proteins
lactate metabolism and consider lactate shuttling as and membranes (i.e. ‘fluxomics’), are research challenges
an important component of intermediary metabolism for the new millennium.
in vivo. Lactate shuttling between producer (driver) Like a phoenix, lactate has again risen to major
and consumer (recipient) cells requires the presence importance in 21st century biology.
of cell-cell and intracellular lactate shuttles that fulfil
at least three purposes; lactate is: (1) a major energy
source, (2) the major gluconeogenic precursor and (3)
a signalling molecule. There is little or no evidence for References
oxygen inadequacy giving rise to lactate production and Ahlborg G & Felig P (1982). Lactate and glucose exchange
accumulation in resting or exercising subjects, even in across the forearm, legs, and splanchnic bed during and
the hypoxia of high altitude (Brooks et al. 1991). Rather, after prolonged leg exercise. J Clin Invest 69, 45–54.
there is abundant evidence that lactate production occurs Ahmed K, Tunaru S, Tang C, Muller M, Gille A, Sassmann A,
in fully aerobic tissues and organs (Gertz et al. 1981; Hanson J & Offermanns S (2010). An autocrine lactate loop
Stanley et al. 1985; Richardson et al. 1998; Park et al. mediates insulin-dependent inhibition of lipolysis through
2015). Recognition for lactate shuttling came first in GPR81. Cell Metab 11, 311–319.
Atlante A, de Bari L, Bobba A, Marra E & Passarella S
studies of physical exercise where roles of driver and
(2007). Transport and metabolism of L-lactate occur in
recipient cells were obvious. However, simultaneously mitochondria from cerebellar granule cells and are modified
and independently the presence of lactate shuttling as in cells undergoing low potassium dependent apoptosis.
part of postprandial glucose disposal was recognized Biochim Biophys Acta 1767, 1285–1299.
in studies of lab animals (Foster, 1984) and humans Azevedo JL Jr, Linderman JK, Lehman SL & Brooks GA
(Woerle et al. 2003). Importantly, lactate (not pyruvate) (1998). Training decreases muscle glycogen turnover during
enters the mitochondrial reticulum to support cell energy exercise. Eur J Appl Physiol Occup Physiol 78, 479–486.

© 2021 The Authors. The Journal of Physiology published by John Wiley & Sons Ltd on behalf of The Physiological Society
14697793, 2022, 5, Downloaded from https://physoc.onlinelibrary.wiley.com/doi/10.1113/JP280955 by Schweizerische Akademie Der, Wiley Online Library on [10/11/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1244 G. A. Brooks and others J Physiol 600.5

Azevedo JL, Tietz E, Two-Feathers T, Paull J & Chapman K Bertocci LA & Lujan BF (1999). Incorporation and utilization
(2007). Lactate, fructose and glucose oxidation profiles in of [3-13C]lactate and [1,2-13C]acetate by rat skeletal
sports drinks and the effect on exercise performance. PLoS muscle. J Appl Physiol 86, 2077–2089.
One 2, e927. Boussouar F & Benahmed M (2004). Lactate and energy
Baba N & Sharma HM (1971). Histochemistry of lactic metabolism in male germ cells. Trends Endocrinol Metab
dehydrogenase in heart and pectoralis muscles of rat. J Cell 15, 345–350.
Biol 51, 621–635. Brandt RB, Laux JE, Spainhour SE & Kline ES (1987). Lactate
Bakeeva LE, Chentsov Yu S & Skulachev VP (1978). dehydrogenase in rat mitochondria. Arch Biochem Biophys
Mitochondrial framework (reticulum mitochondriale) in 259, 412–422.
rat diaphragm muscle. Biochim Biophys Acta 501, 349–369. Bricker DK, Taylor EB, Schell JC, Orsak T, Boutron A, Chen
Baldwin KM, Klinkerfuss GH, Terjung RL, Mole PA & YC, Cox JE, Cardon CM, Van Vranken JG, Dephoure N,
Holloszy JO (1972). Respiratory capacity of white, red, and Redin C, Boudina S, Gygi SP, Brivet M, Thummel CS &
intermediate muscle: adaptative response to exercise. Am J Rutter J (2012). A mitochondrial pyruvate carrier required
Physiol 222, 373–378. for pyruvate uptake in yeast, Drosophila, and humans.
Barnard RJ, Edgerton VR, Furukawa T & Peter JB (1971). Science 337, 96–100.
Histochemical, biochemical, and contractile properties Brooks GA (1985). Lactate:glycolytic end product and
of red, white, and intermediate fibers. Am J Physiol 220, oxidative substrate during sustained exercise in mammals
410–414. — the “lactate shuttle”. In Circulation, Respiration, and
Barnard RJ, Edgerton VR & Peter JB (1970). Effect of exercise Metabolism. Proceedings in Life Sciences. ed. Gilles R,
on skeletal muscle. I. Biochemical and histochemical Springer, Berlin, Heidelberg.
properties. J Appl Physiol 28, 762–766. Brooks GA (1985). Anaerobic threshold: review of the concept
Baumgart E, Fahimi HD, Stich A & Volkl A (1996). L-lactate and directions for future research. Med Sci Sports Exerc 17,
dehydrogenase A4- and A3B isoforms are bona fide 22–34.
peroxisomal enzymes in rat liver. Evidence for involvement Brooks GA (1986). Lactate production under fully aerobic
in intraperoxisomal NADH reoxidation. J Biol Chem 271, conditions: the lactate shuttle during rest and exercise. Fed
3846–3855. Proc 45, 2924–2929.
Belli M, Zhang L, Liu X, Donjacour A, Ruggeri E, Palmerini Brooks GA (2000). Intra- and extra-cellular lactate shuttles.
MG, Nottola SA, Macchiarelli G & Rinaudo P (2020). Med Sci Sports Exerc 32, 790–799.
Oxygen concentration alters mitochondrial structure Brooks GA (2002). Lactate shuttles in nature. Biochem Soc
and function in in vitro fertilized preimplantation mouse Trans 30, 258–264.
embryos. Hum Reprod 35, 1476. Brooks GA (2009). Cell-cell and intracellular lactate shuttles. J
Bender K, Newsholme P, Brennan L & Maechler P (2006). Physiol 587, 5591–5600.
The importance of redox shuttles to pancreatic beta-cell Brooks GA (2018). The science and translation of lactate
energy metabolism and function. Biochem Soc Trans 34, shuttle theory. Cell Metab 27, 757–785.
811–814. Brooks GA (2020a). Lactate as a fulcrum of metabolism.
Bergersen LH (2015). Lactate transport and signaling in the Redox Biol, 35, 101454.
brain: potential therapeutic targets and roles in body-brain Brooks GA (2020b). The precious few grams of glucose during
interaction. J Cereb Blood Flow and Metab 35, 176–185. exercise. Int J Mol Sci 21, 5733.
Bergman BC, Butterfield GE, Wolfel EE, Lopaschuk GD, Brooks GA (2020c). The tortuous path of lactate shuttle
Casazza GA, Horning MA & Brooks GA (1999a). Muscle discovery: from cinders and boards to the lab and ICU. J
net glucose uptake and glucose kinetics after endurance Sport Health Sci 9, 446-460.
training in men. Am J Physiol 277, E81-92. Brooks GA, Brauner KE & Cassens RG (1973). Glycogen
Bergman BC, Horning MA, Casazza GA, Wolfel EE, synthesis and metabolism of lactic acid after exercise. Am
Butterfield GE & Brooks GA (2000). Endurance training J Physiol 224, 1162–1166.
increases gluconeogenesis during rest and exercise in men. Brooks GA, Brown MA, Butz CE, Sicurello JP & Dubouchaud
Am J Physiol Endocrinol Metab 278, E244-E251. H (1999a). Cardiac and skeletal muscle mitochondria have
Bergman BC, Tsvetkova T, Lowes B & Wolfel EE (2009a). a monocarboxylate transporter MCT1. J Appl Physiol (1985)
Myocardial FFA metabolism during rest and atrial pacing in 87, 1713–1718.
humans. Am J Physiol Endocrinol Metab 296, E358-E366. Brooks GA, Brown MA, Butz CE, Sicurello JP & Dubouchaud
Bergman BC, Tsvetkova T, Lowes B & Wolfel EE (2009b). H (1999b). Cardiac and skeletal muscle mitochondria have
Myocardial glucose and lactate metabolism during rest and a monocarboxylate transporter MCT1. J Appl Physiol 87,
atrial pacing in humans. J Physiol 587, 2087–2099. 1713–1718.
Bergman BC, Wolfel EE, Butterfield GE, Lopaschuk GD, Brooks GA, Brown MA, Butz CE, Sicurello JP & Dubouchaud
Casazza GA, Horning MA & Brooks GA (1999b). Active H (1999c). Cardiac and skeletal muscle mitochondria have
muscle and whole body lactate kinetics after endurance a monocarboxylate transporter MCT1. J Appl Physiol 87,
training in men. J Appl Physiol 87, 1684–1696. 1713–1718.
Bertocci LA, Haller RG & Lewis SF (1993). Muscle Brooks GA, Butterfield GE, Wolfe RR, Groves BM, Mazzeo
metabolism during lactate infusion in human RS, Sutton JR, Wolfel EE & Reeves JT (1991). Decreased
phosphofructokinase deficiency. J Appl Physiol 74, reliance on lactate during exercise after acclimatization to
1342–1347. 4,300 m. J Appl Physiol 71, 333–341.

© 2021 The Authors. The Journal of Physiology published by John Wiley & Sons Ltd on behalf of The Physiological Society
14697793, 2022, 5, Downloaded from https://physoc.onlinelibrary.wiley.com/doi/10.1113/JP280955 by Schweizerische Akademie Der, Wiley Online Library on [10/11/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J Physiol 600.5 Lactate, phoenix risen 1245

Brooks GA & Donovan CM (1983). Effect of endurance de Bari L, Valenti D, Atlante A & Passarella S (2010).
training on glucose kinetics during exercise. Am J Physio L-lactate generates hydrogen peroxide in purified rat
Endocrinol Metabl 244, E505–E512. liver mitochondria due to the putative L-lactate oxidase
Brooks GA, Dubouchaud H, Brown M, Sicurello JP & Butz localized in the intermembrane space. FEBS Lett 584,
CE (1999d). Role of mitochondrial lactate dehydrogenase 2285–2290.
and lactate oxidation in the intracellular lactate shuttle. Proc de Paoli FV, Overgaard K, Pedersen TH & Nielsen OB (2007).
Natl Acad Sci U S A 96, 1129–1134. Additive protective effects of the addition of lactic acid and
Brooks GA, Fahey TD & Baldwin KM (2019). The why adrenaline on excitability and force in isolated rat skeletal
of pulmonary ventilation. Exercise Physiology: human muscle depressed by elevated extracellular K+ . J Physiol
bioenergetics and its applications, 5th edn., vol. 1, pp. 581, 829–839.
243–256. Kindle Direct Publishing, Lexington, KY, USA. Depocas F, Minaire Y & Chatonnet J (1969). Rates of
Brooks GA & Gaesser GA (1980). End points of lactate and formation and oxidation of lactic acid in dogs at rest and
glucose metabolism after exhausting exercise. J Appl Physiol during moderate exercise. Can J Physiol Pharmacol 47,
Respir Environ Exerc Physiol 49, 1057–1069. 603–610.
Brooks GA & Martin NA (2014). Cerebral metabolism Donovan CM & Brooks GA (1983). Endurance training affects
following traumatic brain injury: new discoveries with lactate clearance, not lactate production. Am J Physiol End-
implications for treatment. Front Neurosci 8, 408. ocrinol Metab 244, E83–E92.
Brooks GA & Mercier J (1994). Balance of carbohydrate and Donovan CM, Brooks GA, Donovan CM & Brooks GA
lipid utilization during exercise: the “crossover” concept. J (1983). Endurance training affects lactate clearance, not
Appl Physiol 76, 2253–2261. lactate production. Am J Physiol Endocrinol Metab 244,
Brown MA & Brooks GA (1994). Trans-stimulation of lactate E83–E92.
transport from rat sarcolemmal membrane vesicles. Arch Dubouchaud H, Butterfield GE, Wolfel EE, Bergman BC &
Biochem Biophys 313, 22–28. Brooks GA (2000). Endurance training, expression, and
Brown TP, Bhattacharjee P, Ramachandran S, Sivaprakasam S, physiology of LDH, MCT1, and MCT4 in human skeletal
Ristic B, Sikder MOF & Ganapathy V (2020). The lactate muscle. Am J Physiol Endocrinol Metab 278, E571–E579.
receptor GPR81 promotes breast cancer growth via a Duncan SH, Louis P & Flint HJ (2004). Lactate-utilizing
paracrine mechanism involving antigen-presenting cells bacteria, isolated from human feces, that produce butyrate
in the tumor microenvironment. Oncogene, 1–13. as a major fermentation product. Appl Environ Microbiol 70,
Cai TQ, Ren N, Jin L, Cheng K, Kash S, Chen R, Wright 5810–5817.
SD, Taggart AK & Waters MG (2008). Role of GPR81 in El Hayek L, Khalifeh M, Zibara V, Abi Assaad R, Emmanuel
lactate-mediated reduction of adipose lipolysis. Biochem N, Karnib N, El-Ghandour R, Nasrallah P, Bilen M,
Biophys Res Commun 377, 987–991. Ibrahim P, Younes J, Abou Haidar E, Barmo N, Jabre
Chen YJ, Mahieu NG, Huang X, Singh M, Crawford PA, V, Stephan JS & Sleiman SF (2019). Lactate mediates
Johnson SL, Gross RW, Schaefer J & Patti GJ (2016). Lactate the effects of exercise on learning and memory through
metabolism is associated with mammalian mitochondria. SIRT1-dependent activation of hippocampal brain-derived
Nat Chem Biol 12, 937–943. neurotrophic factor (BDNF). J Neurosci 39, 2369–2382.
Coady MJ, Chang MH, Charron FM, Plata C, Wallendorff B, Estaki M, Morck DW, Quin C, Pither J, Barnett JA, Gill
Sah JF, Markowitz SD, Romero MF & Lapointe JY (2004). SK & Gibson DL (2020). Physical activity shapes the
The human tumour suppressor gene SLC5A8 expresses intestinal microbiome and immunity of healthy mice but
a Na+ -monocarboxylate cotransporter. J Physiol 557, has no protective effects against colitis in MUC2 −/− mice.
719–731. mSystems 5. e00515-20.
Connett RJ, Honig CR, Gayeski TE & Brooks GA (1990). Estaki M, Pither J, Baumeister P, Little JP, Gill SK, Ghosh
Defining hypoxia: a systems view of VO2, glycolysis, S, Ahmadi-Vand Z, Marsden KR & Gibson DL (2016).
energetics, and intracellular PO2. J Appl Physiol (1985) 68, Cardiorespiratory fitness as a predictor of intestinal micro-
833–842. bial diversity and distinct metagenomic functions. Micro-
Cori CF & Cori GT (1946). Carbohydrate metabolism. Annu biome 4, 42.
Rev Biochem 15, 193–218. Ferguson BS, Rogatzki MJ, Goodwin ML, Kane DA, Rightmire
Daw C.C., Ramachandran K, Enslow BT, Maity S, Bursic Z & Gladden LB (2018). Lactate metabolism: historical
B, Novello MJ, Rubannelsonkumar CS, Mashal AH, context, prior misinterpretations, and current under-
Ravichandran J, Bakewell TM, Wanf W, Lj K, Madaris TR, standing. Eur J Appl Physiol 118, 691–728.
Shannon CE, Norton L, Kandala S, Caplan J, Srikantan S, Feuer S, Liu X, Donjacour A, Simbulan R, Maltepe E &
Stathopulos PB, Reeves WB & Madesh M (2020). Lactate Rinaudo P (2017). Common and specific transcriptional
elicits ER-mitochondrial Mg2+ dynamics to integrate signatures in mouse embryos and adult tissues induced by
cellular metabolism. Cell 183, 474-489.e17. in vitro procedures. Reproduction 153, 107–122.
De Bari L, Atlante A, Valenti D & Passarella S (2004). Foster DW (1984). Banting lecture 1984. From glycogen to
Partial reconstruction of in vitro gluconeogenesis arising ketones–and back. Diabetes 33, 1188–1199.
from mitochondrial L-lactate uptake/metabolism and Freminet A, Bursaux E & Poyart CF (1974). Effect of elevated
oxaloacetate export via novel L-lactate translocators. lactataemia on the rates of lactate turnover and oxidation in
Biochem J 380, 231–242. rats. Pflugers Arch 346, 75–86.

© 2021 The Authors. The Journal of Physiology published by John Wiley & Sons Ltd on behalf of The Physiological Society
14697793, 2022, 5, Downloaded from https://physoc.onlinelibrary.wiley.com/doi/10.1113/JP280955 by Schweizerische Akademie Der, Wiley Online Library on [10/11/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1246 G. A. Brooks and others J Physiol 600.5

Gaesser GA & Brooks GA (1980). Glycogen repletion Glenn TC, Martin NA, Horning MA, McArthur DL, Hovda
following continuous and intermittent exercise to D, Vespa PM & Brooks GA (2015a). Lactate: brain fuel in
exhaustion. J Appl Physiol Respir Environ Exerc Physiol 49, human traumatic brain injury. A comparison to normal
722–728. healthy control subjects. J Neurotrauma 32, 820–832.
Gaesser GA & Brooks GA (1984). Metabolic bases of excess Glenn TC, Martin NA, McArthur DL, Hovda DA, Vespa P,
post-exercise oxygen consumption: a review. Med Sci Sports Johnson ML, Horning MA & Brooks GA (2015b). End-
Exerc 16, 29–43. ogenous nutritive support after traumatic brain injury:
Gale SM, Castracane VD & Mantzoros CS (2004). Energy peripheral lactate production for glucose supply via
homeostasis, obesity and eating disorders: recent advances gluconeogenesis. J Neurotrauma 32, 811–819.
in endocrinology. J Nutr 134, 295–298. Gohil K, Viguie C, Stanley WC, Brooks GA & Packer L (1988).
Garcia CK, Brown MS, Pathak RK & Goldstein JL (1995). Blood glutathione oxidation during human exercise. J Appl
cDNA cloning of MCT2, a second monocarboxylate trans- Physiol (1985) 64, 115–119.
porter expressed in different cells than MCT1. J Biol Chem Gold M, Miller HI, Issekutz B Jr & Spitzer JJ (1963). Effect
270, 1843–1849. of exercise and lactic acid infusion on individual free fatty
Garcia CK, Goldstein JL, Pathak RK, Anderson RG & Brown acids of plasma. Am J Physiol 205, 902–904.
MS (1994). Molecular characterization of a membrane Goodpaster BH & Sparks LM (2017). Metabolic flexibility in
transporter for lactate, pyruvate, and other mono- health and disease. Cell Metab 25, 1027–1036.
carboxylates: implications for the Cori cycle. Cell 76, Halangk W, Bohnensack R, Frank K & Kunz W (1985).
865–873. Effect of various substrates on mitochondrial and cellular
Garcia-Alvarez M, Marik P & Bellomo R (2014a). energy state of intact spermatozoa. Biomed Biochim Acta 44,
Sepsis-associated hyperlactatemia. Crit Care 18, 503. 411–420.
Garcia-Alvarez M, Marik P & Bellomo R (2014b). Stress Hashimoto T, Hussien R & Brooks GA (2006). Colocalization
hyperlactataemia: present understanding and controversy. of MCT1, CD147, and LDH in mitochondrial inner
Lancet Diabetes Endocrinol 2, 339–347. membrane of L6 muscle cells: evidence of a mitochondrial
Gardner DK (2015). Lactate production by the mammalian lactate oxidation complex. Am J Physiol Endocrinol Metab
blastocyst: manipulating the microenvironment for uterine 290, E1237–E1244.
implantation and invasion? Bioessays 37, 364–371. Hashimoto T, Hussien R, Cho HS, Kaufer D & Brooks GA
Ge H, Weiszmann J, Reagan JD, Gupte J, Baribault H, Gyuris (2008). Evidence for the mitochondrial lactate oxidation
T, Chen JL, Tian H & Li Y (2008). Elucidation of signaling complex in rat neurons: demonstration of an essential
and functional activities of an orphan GPCR, GPR81. J component of brain lactate shuttles. PLoS One 3, e2915.
Lipid Res 49, 797–803. Hashimoto T, Hussien R, Oommen S, Gohil K & Brooks GA
Gerich JE, Meyer C, Woerle HJ & Stumvoll M (2001). Renal (2007). Lactate sensitive transcription factor network in L6
gluconeogenesis: its importance in human glucose homeo- cells: activation of MCT1 and mitochondrial biogenesis.
stasis. Diabetes Care 24, 382–391. FASEB J 21, 2602–2612.
Gertz EW, Wisneski JA, Neese R, Bristow JD, Searle GL & Hashimoto T, Masuda S, Taguchi S & Brooks GA (2005).
Hanlon JT (1981). Myocardial lactate metabolism: evidence Immunohistochemical analysis of MCT1, MCT2 and MCT4
of lactate release during net chemical extraction in man. expression in rat plantaris muscle. J Physiol 567, 121–129.
Circulation 63, 1273–1279. Hashimoto T, Sato K & Iemitsu M (2013). Exercise-inducible
Gertz EW, Wisneski JA, Stanley WC & Neese RA (1988). factors to activate lipolysis in adipocytes. J Appl Physiol
Myocardial substrate utilization during exercise in humans. (1985) 115, 260–267.
Dual carbon-labeled carbohydrate isotope experiments. J Hashimoto T, Tsukamoto H, Takenaka S, Olesen ND, Petersen
Clin Invest 82, 2017–2025. LG, Sorensen H, Nielsen HB, Secher NH & Ogoh S (2018).
Ghosal S, Myers B & Herman JP (2013). Role of central Maintained exercise-enhanced brain executive function
glucagon-like peptide-1 in stress regulation. Physiol Behav related to cerebral lactate metabolism in men. FASEB J 32,
122, 201–207. 1417–1427.
Gladden LB (1991). Net lactate uptake during progressive Henderson GC, Horning MA, Lehman SL, Wolfel EE,
steady-level contractions in canine skeletal muscle. J Appl Bergman BC & Brooks GA (2004). Pyruvate shuttling
Physiol 71, 514–520. during rest and exercise before and after endurance training
Gladden LB (2004). Lactate metabolism: a new paradigm for in men. J Appl Physiol 97, 317–325.
the third millennium. J Physiol 558, 5–30. Henderson GC, Horning MA, Wallis GA & Brooks GA (2007).
Gladden LB, Crawford RE & Webster MJ (1994). Effect of Pyruvate metabolism in working human skeletal muscle.
lactate concentration and metabolic rate on net lactate Am J Physiol Endocrinol Metab 292, E366.
uptake by canine skeletal muscle. Am J Physio Regul Integr Herzig S, Raemy E, Montessuit S, Veuthey JL, Zamboni
Comp Physioll 266, R1095–R1101. N, Westermann B, Kunji ER & Martinou JC (2012).
Glancy B, Hartnell LM, Malide D, Yu ZX, Combs CA, Identification and functional expression of the
Connelly PS, Subramaniam S & Balaban RS (2015). mitochondrial pyruvate carrier. Science 337, 93–96.
Mitochondrial reticulum for cellular energy distribution Hill AV (1914). The oxidative removal of lactic acid. J Physiol
in muscle. Nature 523, 617–620. 48, x-xi.

© 2021 The Authors. The Journal of Physiology published by John Wiley & Sons Ltd on behalf of The Physiological Society
14697793, 2022, 5, Downloaded from https://physoc.onlinelibrary.wiley.com/doi/10.1113/JP280955 by Schweizerische Akademie Der, Wiley Online Library on [10/11/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J Physiol 600.5 Lactate, phoenix risen 1247

Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot Jöbsis FF & Stainsby WN (1968). Oxidation of NADH during
B, Morelli L, Canani RB, Flint HJ, Salminen S, Calder PC contractions of circulated mammalian skeletal muscle.
& Sanders ME (2014). Expert consensus document. The Respir Physiol 4, 292–300.
International Scientific Association for Probiotics and Pre- Johnson JA & Fusaro RM (1972). The role of the skin in
biotics consensus statement on the scope and appropriate carbohydrate metabolism. Adv Metab Disord 60, 1–55.
use of the term probiotic. Nat Rev Gastroenterol Hepatol 11, Johnson ML, Emhoff CA, Horning MA & Brooks GA (2012).
506–514. Transpulmonary lactate shuttle. Am J Physiol Regul Integr
Hofmann P, Bunc V, Leitner H, Pokan R & Gaisl G (1994). Comp Physiol 302, R143–R149.
Heart rate threshold related to lactate turn point and Johnson ML, Hussien R, Horning MA & Brooks GA (2011).
steady-state exercise on a cycle ergometer. Eur J Appl Transpulmonary pyruvate kinetics. Am J Physiol Regul
Physiol Occup Physiol 69, 132–139. Integr Comp Physiol 301, R769–R774.
Holloway R, Zhou Z, Harvey HB, Levasseur JE, Rice AC, Jones AR (1997). Metabolism of lactate by mature boar
Sun D, Hamm RJ & Bullock MR (2007). Effect of lactate spermatozoa. Reprod Fertil Dev 9, 227–232.
therapy upon cognitive deficits after traumatic brain injury Jones TE, Pories WJ, Houmard JA, Tanner CJ, Zheng D, Zou
in the rat. Acta Neurochir (Wien) 149, 919–927; discussion K, Coen PM, Goodpaster BH, Kraus WE & Dohm GL
927. (2019). Plasma lactate as a marker of metabolic health:
Honig CR, Connett RJ & Gayeski TE (1992). O2 transport and Implications of elevated lactate for impairment of aerobic
its interaction with metabolism; a systems view of aerobic metabolism in the metabolic syndrome. Surgery 166,
capacity. Med Sci Sports Exerc 24, 47–53. 861–866,
Hooker AM & Baldwin KM (1979). Substrate oxidation Kane DA (2014). Lactate oxidation at the mitochondria: a
specificity in different types of mammalian muscle. Am J lactate-malate-aspartate shuttle at work. Front Neurosci 8,
Physiol Cell Physiol 236, C66–C69. 366.
Hoque R, Farooq A, Ghani A, Gorelick F & Mehal WZ (2014). Kelley DE, Goodpaster B, Wing RR & Simoneau JA (1999).
Lactate reduces liver and pancreatic injury in Toll-like Skeletal muscle fatty acid metabolism in association with
receptor- and inflammasome-mediated inflammation via insulin resistance, obesity, and weight loss. Am J Physiol
GPR81-mediated suppression of innate immunity. Gastro- Endocrinol Metab 277, E1130–E1141.
enterology 146, 1763–1774. Kelley KM, Hamann JJ, Navarre C & Gladden LB (2002).
Hui S, Ghergurovich JM, Morscher RJ, Jang C, Teng X, Lu Lactate metabolism in resting and contracting canine
W, Esparza LA, Reya T, Le Z, Yanxiang Guo J, White E skeletal muscle with elevated lactate concentration. J Appl
& Rabinowitz JD (2017). Glucose feeds the TCA cycle via Physiol (1985) 93, 865–872.
circulating lactate. Nature 551, 115–118. Kirkwood SP, Munn EA & Brooks GA (1986). Mitochondrial
Hunt TK, Aslam RS, Beckert S, Wagner S, Ghani QP, Hussain reticulum in limb skeletal muscle. Am J Physiol Cell Physiol
MZ, Roy S & Sen CK (2007). Aerobically derived lactate 251, C395–C402.
stimulates revascularization and tissue repair via redox Kirkwood SP, Packer L & Brooks GA (1987). Effects of end-
mechanisms. Antioxid Redox Signal 9, 1115–1124. urance training on a mitochondrial reticulum in limb
Hussien R & Brooks GA (2011). Mitochondrial and plasma skeletal muscle. Arch Biochem Biophys 255, 80–88.
membrane lactate transporter and lactate dehydrogenase Kline ES, Brandt RB, Laux JE, Spainhour SE, Higgins ES,
isoform expression in breast cancer cell lines. Physiol Rogers KS, Tinsley SB & Waters MG (1986). Localization
Genomics 43, 255–264. of L-lactate dehydrogenase in mitochondria. Arch Biochem
Inskeep PB & Hammerstedt RH (1985). Endogenous Biophys 246, 673–680.
metabolism by sperm in response to altered cellular ATP Kreutzer U & Jue T (1995). Critical intracellular O2 in myo-
requirements. J Cell Physiol 123, 180–190. cardium as determined by 1H nuclear magnetic resonance
Islam H, Townsend LK, McKie GL, Medeiros PJ, Gurd signal of myoglobin. Am J Physiol Heart Circ Physiol 268,
BJ & Hazell TJ (2017). Potential involvement of lactate H1675–H1681.
and interleukin-6 in the appetite-regulatory hormonal Lazarow PB & De Duve C (1976). A fatty acyl-CoA oxidizing
response to an acute exercise bout. J Appl Physiol (1985) system in rat liver pEeroxisomes; enhancement by
123, 614–623. clofibrate, a hypolipidemic drug. Proc Natl Acad Sci U S
Issekutz BJ, and & Miller H (1962). Plasma free fatty acids A 73, 2043–2046.
during exercise and the effect of lactic acid. Proc Soc Exp Lecoultre V, Benoit R, Carrel G, Schutz Y, Millet GP, Tappy
Biol Med 110, 237–239. L & Schneiter P (2010). Fructose and glucose co-ingestion
James DE, Kraegen EW & Chisholm DJ (1985). Effects of during prolonged exercise increases lactate and glucose
exercise training on in vivo insulin action in individual fluxes and oxidation compared with an equimolar intake of
tissues of the rat. J Clin Invest 76, 657–666. glucose. Am J Clin Nutr 92, 1071–1079.
James DE, Zorzano A, Boni-Schnetzler M, Nemenoff RA, Lehninger AL (1970). Biochemistry, p. 326. Worth, New York.
Powers A, Pilch PF & Ruderman NB (1986). Intrinsic Li L, Chen K, Wang T, Wu Y, Xing G, Chen M, Hao Z, Zhang
differences of insulin receptor kinase activity in red and C, Zhang J, Ma B, Liu Z, Yuan H, Liu Z, Long Q, Zhou Y,
white muscle. J Biol Chem 261, 14939–14944. Qi J, Zhao D, Gao M, Pei D, Nie J, Ye D, Pan G & Liu X
Jeukendrup AE & Chambers ES (2010). Oral carbohydrate (2020). Glis1 facilitates induction of pluripotency via an
sensing and exercise performance. Curr Opin Clin Nutr epigenome-metabolome-epigenome signalling cascade. Nat
Metab Care 13, 447–451. Metab 2, 882–892.

© 2021 The Authors. The Journal of Physiology published by John Wiley & Sons Ltd on behalf of The Physiological Society
14697793, 2022, 5, Downloaded from https://physoc.onlinelibrary.wiley.com/doi/10.1113/JP280955 by Schweizerische Akademie Der, Wiley Online Library on [10/11/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1248 G. A. Brooks and others J Physiol 600.5

Liu C, Wu J, Zhu J, Kuei C, Yu J, Shelton J, Sutton SW, Li X, Miller BF, Fattor JA, Jacobs KA, Horning MA, Navazio F,
Yun SJ, Mirzadegan T, Mazur C, Kamme F & Lovenberg Lindinger MI & Brooks GA (2002a). Lactate and glucose
TW (2009). Lactate inhibits lipolysis in fat cells through interactions during rest and exercise in men: effect of
activation of an orphan G-protein-coupled receptor, GPR81. exogenous lactate infusion. J Physiol 544, 963–975.
J Biol Chem 284, 2811–2822. Miller BF, Fattor JA, Jacobs KA, Horning MA, Suh SH,
McCarthy SF, Islam H & Hazell TJ (2020). The emerging Navazio F & Brooks GA (2002b). Metabolic and cardio-
role of lactate as a mediator of exercise-induced appetite respiratory responses to “the lactate clamp”. Am J Physiol
suppression. Am J Physiol Endocrinol Metab 319, Endocrinol Metab 283, E889–E898.
E814–E819. Miller BF, Lindinger MI, Fattor JA, Jacobs KA, Leblanc
McClelland GB, Khanna S, Gonzalez GF, Butz CE & Brooks PJ, Duong M, Heigenhauser GJ & Brooks GA (2005).
GA (2003). Peroxisomal membrane monocarboxylate Hematological and acid-base changes in men during
transporters: evidence for a redox shuttle system? Biochem prolonged exercise with and without sodium-lactate
Biophys Res Commun 304, 130–135. infusion. J Appl Physiol 98, 856–865.
McGarry JD, Mannaerts GP & Foster DW (1977). A possible Miller HI, Issekutz B Jr, Rodahl K & Paul P (1964). Effect of
role for malonyl-CoA in the regulation of hepatic fatty acid lactic acid on plasma free fatty acids in pancreatectomized
oxidation and ketogenesis. J Clin Invest 60, 265–270. dogs. Am J Physiol 207, 1226–1230.
Liu L, MacKenzie KR, Putluri N, Maletic-Savatic M & Murphy KG & Bloom SR (2006). Gut hormones and the
Bellen HJ (2017). The glia-neuron lactate shuttle and regulation of energy homeostasis. Nature 444, 854–859.
elevated ROS promote lipid synthesis in neurons and lipid Nilsson LH & Hultman E (1974). Liver and muscle glycogen
droplet accumulation in glia via APOE/D. Cell Metab 26, in man after glucose and fructose infusion. Scand J Clin Lab
719–737.e6. Invest 33, 5–10.
Madaan A, Nadeau-Vallee M, Rivera JC, Obari D, Hou X, Ohno Y, Ando K, Ito T, Suda Y, Matsui Y, Oyama A, Kaneko
Sierra EM, Girard S, Olson DM & Chemtob S (2017). H, Yokoyama S, Egawa T & Goto K (2019). Lactate
Lactate produced during labor modulates uterine stimulates a potential for hypertrophy and regeneration
inflammation via GPR81 (HCA1). Am J Obstet Gynecol of mouse skeletal muscle. Nutrients 869, 11.
216, 60.e1-60.e17. Otonkoski T, Jiao H, Kaminen-Ahola N, Tapia-Paez I, Ullah
Magistretti PJ, Pellerin L, Rothman DL & Shulman RG (1999). MS, Parton LE, Schuit F, Quintens R, Sipila I, Mayatepek
Energy on demand. Science 283, 496–497. E, Meissner T, Halestrap AP, Rutter GA & Kere J (2007).
Marik P & Bellomo R (2013). Lactate clearance as a target of Physical exercise-induced hypoglycemia caused by failed
therapy in sepsis: a flawed paradigm. OA Critical Care 1, silencing of monocarboxylate transporter 1 in pancreatic
3–5. beta cells. Am J Hum Genet 81, 467–474.
Marik P & Bellomo R (2016). A rational approach to fluid Park JM, Josan S, Mayer D, Hurd RE, Chung Y, Bendahan D,
therapy in sepsis. Br J Anaesth 116, 339–349. Spielman DM & Jue T (2015). Hyperpolarized 13C NMR
Marik PE (2009). Glycemic control in critically ill patients: observation of lactate kinetics in skeletal muscle. J Exp Biol
what to do post NICE-SUGAR? World J Gastrointest Surg 1, 218, 3308–3318.
3–5. Passarella S, de Bari L, Valenti D, Pizzuto R, Paventi G &
Mayer J, Roy P & Mitra KP (1956). Relation between caloric Atlante A (2008). Mitochondria and L-lactate metabolism.
intake, body weight, and physical work: studies in an FEBS Lett 582, 3569–3576.
industrial male population in West Bengal. Am J Clin Nutr Passarella S, Paventi G & Pizzuto R (2014). The mitochondrial
4, 169–175. L-lactate dehydrogenase affair. Front Neurosci 8, 407.
Mazzeo RS, Brooks GA, Schoeller DA & Budinger TF (1986). Patel AB, Lai JC, Chowdhury GM, Hyder F, Rothman DL,
Disposal of blood [1-13C]lactate in humans during rest and Shulman RG & Behar KL (2014). Direct evidence for
exercise. J Appl Physiol 60, 232–241. activity-dependent glucose phosphorylation in neurons
Messonnier AL, Emhoff CW, Fattor JA, Horning MA, T.J. C & with implications for the astrocyte-to-neuron lactate shuttle.
Brooks GA (2013). Lactate kinetics at the lactate threshold Proc Natl Acad Sci U S A 111, 5385–5390.
in trained and untrained men. J Appl Physiol 1593-1602, Pellerin L, Bergersen LH, Halestrap AP & Pierre K (2005).
114. Cellular and subcellular distribution of monocarboxylate
Meyer C, Dostou J, Nadkarni V & Gerich J (1998). Effects transporters in cultured brain cells and in the adult brain. J
of physiological hyperinsulinemia on systemic, renal, and Neurosci Res 79, 55–64.
hepatic substrate metabolism. Am J Physiol Renal Physiol Pellerin L & Magistretti PJ (1994). Glutamate uptake into
275, F915–F921. astrocytes stimulates aerobic glycolysis: a mechanism
Meyer C, Dostou JM, Welle SL & Gerich JE (2002). Role of coupling neuronal activity to glucose utilization. Proc Natl
human liver, kidney, and skeletal muscle in postprandial Acad Sci U S A 91, 10625–10629.
glucose homeostasis. Am J Physiol Endocrinol Metab 282, Pellerin L & Magistretti PJ (2012). Sweet sixteen for ANLS. J
E419–E427. Cereb Blood Flow Metab 32, 1152–1166.
Meyerhof O (1920). Die Energieumwandlungen im Muskel II. Pellerin L, Pellegri G, Bittar PG, Charnay Y, Bouras C, Martin
Das Schicksal der Milchsaure in der Erholungsperiode des JL, Stella N & Magistretti PJ (1998). Evidence supporting
Muskels. Pflügers Archiv ges Physiol Mensch Tiere 182-182, the existence of an activity-dependent astrocyte-neuron
284–317. lactate shuttle. Dev Neurosci 20, 291–299.

© 2021 The Authors. The Journal of Physiology published by John Wiley & Sons Ltd on behalf of The Physiological Society
14697793, 2022, 5, Downloaded from https://physoc.onlinelibrary.wiley.com/doi/10.1113/JP280955 by Schweizerische Akademie Der, Wiley Online Library on [10/11/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J Physiol 600.5 Lactate, phoenix risen 1249

Poole DC, Rossiter HB, Brooks GA & Gladden LB (2021). The San-Millan I, Julian CG, Matarazzo C & Brooks GA (2020).
anaerobic threshold: 50+ years of controversy. J Physiol Is lactate an oncometabolite? evidence supporting a role
599, 737–767. for lactate in the regulation of transcriptional activity of
Powell KE, King AC, Buchner DM, Campbell WW, DiPietro cancer-related genes in MCF7 breast cancer cells. Front
L, Erickson KI, Hillman CH, Jakicic JM, Janz KF, Oncol - Cancer Metab 9, 1–10.
Katzmarzyk PT, Kraus WE, Macko RF, Marquez DX, Scheiman J, Luber JM, Chavkin TA, MacDonald T, Tung
McTiernan A, Pate RR, Pescatello LS & Whitt-Glover MC A, Pham LD, Wibowo MC, Wurth RC, Punthambaker S,
(2018). The scientific foundation for the physical activity Tierney BT, Yang Z, Hattab MW, Avila-Pacheco J, Clish CB,
guidelines for Americans, 2nd edition. J Phys Act Health, Lessard S, Church GM & Kostic AD (2019). Meta-omics
1–11. analysis of elite athletes identifies a performance-enhancing
Powers SK, Ji LL & Leeuwenburgh C (1999). Exercise microbe that functions via lactate metabolism. Nat Med 25,
training-induced alterations in skeletal muscle antioxidant 1104–1109.
capacity: a brief review. Med Sci Sports Exerc 31, 987–997. Schmid SM, Jauch-Chara K, Hallschmid M, Oltmanns
Price NT, Jackson VN & Halestrap AP (1998). Cloning and KM, Peters A, Born J & Schultes B (2008). Lactate over-
sequencing of four new mammalian monocarboxylate rides central nervous but not beta-cell glucose sensing in
transporter (MCT) homologues confirms the existence of humans. Metabolism 57, 1733–1739.
a transporter family with an ancient past. Biochem J 329 Schultes B, Schmid SM, Wilms B, Jauch-Chara K, Oltmanns
321–328. KM & Hallschmid M (2012). Lactate infusion during
Pullen TJ, Khan AM, Barton G, Butcher SA, Sun G & Rutter euglycemia but not hypoglycemia reduces subsequent food
GA (2010). Identification of genes selectively disallowed in intake in healthy men. Appetite 58, 818–821.
the pancreatic islet. Islets 2, 89–95. Schurr A (2008). Lactate: a major and crucial player in
Rabinowitz JD & Enerback S (2020). Lactate: the ugly duckling normal function of both muscle and brain. J Physiol 586,
of energy metabolism. Nat Metab 2, 566–571. 2665–2666.
Rice AC, Zsoldos R, Chen T, Wilson MS, Alessandri B, Hamm Semenza GL (2008). Tumor metabolism: cancer cells give and
RJ & Bullock MR (2002). Lactate administration attenuates take lactate. J Clin Invest 118, 3835–3837.
cognitive deficits following traumatic brain injury. Brain Res Shapiro NI, Howell MD, Talmor D, Nathanson LA, Lisbon
928, 156–159. A, Wolfe RE & Weiss JW (2005). Serum lactate as a pre-
Richardson RS, Noyszewski EA, Leigh JS & Wagner PD dictor of mortality in emergency department patients with
(1998). Lactate efflux from exercising human skeletal infection. Ann Emerg Med 45, 524–528.
muscle: role of intracellular PO2. J Appl Physiol 85, Shulman RG, Hyder F & Rothman DL (2001). Cerebral
627–634. energetics and the glycogen shunt: neurochemical basis
Rodahl K, Miller HI & Issekutz B Jr (1964). Plasma free fatty of functional imaging. Proc Natl Acad Sci U S A 98,
acids in exercise. J Appl Physiol 19, 489–492. 6417–6422.
Rogatzki MJ, Ferguson BS, Goodwin ML & Gladden LB Shulman RG & Rothman DL (2001). The “glycogen shunt”
(2015). Lactate is always the end product of glycolysis. Front in exercising muscle: a role for glycogen in muscle
Neurosci 9, 22. energetics and fatigue. Proc Natl Acad Sci U S A 98,
Roth DA & Brooks GA (1990a). Lactate and pyruvate trans- 457–461.
port is dominated by a pH gradient-sensitive carrier in rat Smadja C, Morin J, Ferre P & Girard J (1988). Metabolic fate
skeletal muscle sarcolemmal vesicles. Arch Biochem Biophys of a gastric glucose load in unrestrained rats bearing a
279, 386–394. portal vein catheter. Am J Physiol Endocrinol Metab 254,
Roth DA & Brooks GA (1990b). Lactate transport is mediated E407–E413.
by a membrane-bound carrier in rat skeletal muscle Somasetia DH, Setiati TE, Sjahrodji AM, Idjradinata PS,
sarcolemmal vesicles. Arch Biochem Biophys 279, 377–385. Setiabudi D, Roth H, Ichai C, Fontaine E & Leverve XM
Rutter GA, Pullen TJ, Hodson DJ & Martinez-Sanchez A (2014). Early resuscitation of dengue shock syndrome in
(2015). Pancreatic beta-cell identity, glucose sensing and children with hyperosmolar sodium-lactate: a randomized
the control of insulin secretion. Biochem J 466, 203–218. single-blind clinical trial of efficacy and safety. Crit Care 18,
Saddik M, Gamble J, Witters LA & Lopaschuk GD (1993). 466.
Acetyl-CoA carboxylase regulation of fatty acid oxidation in Sonveaux P, Vegran F, Schroeder T, Wergin MC, Verrax
the heart. J Biol Chem 268, 25836–25845. J, Rabbani ZN, De Saedeleer CJ, Kennedy KM, Diepart
San-Millan I & Brooks GA (2017). Reexamining cancer C, Jordan BF, Kelley MJ, Gallez B, Wahl ML, Feron O &
metabolism: lactate production for carcinogenesis could Dewhirst MW (2008). Targeting lactate-fueled respiration
be the purpose and explanation of the Warburg Effect. selectively kills hypoxic tumor cells in mice. J Clin Invest
Carcinogenesis 38, 119–133. 118, 3930–3942.
San-Millan I & Brooks GA (2018). Assessment of metabolic Spitzer JJ (1979). Gluconeogenesis in the burned patient. J
flexibility by means of measuring blood lactate, fat, and Trauma 19, 899–900.
carbohydrate oxidation responses to exercise in professional Stainsby WN & Welch HG (1966). Lactate metabolism of
endurance athletes and less-fit individuals. Sports Med 48, contracting dog skeletal muscle in situ. Am J Physiol 211,
467–479. 177–183.

© 2021 The Authors. The Journal of Physiology published by John Wiley & Sons Ltd on behalf of The Physiological Society
14697793, 2022, 5, Downloaded from https://physoc.onlinelibrary.wiley.com/doi/10.1113/JP280955 by Schweizerische Akademie Der, Wiley Online Library on [10/11/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1250 G. A. Brooks and others J Physiol 600.5

Stanley WC, Gertz EW, Wisneski JA, Morris DL, Neese RA van Hall G, Stromstad M, Rasmussen P, Jans O, Zaar M, Gam
& Brooks GA (1985). Systemic lactate kinetics during C, Quistorff B, Secher NH & Nielsen HB (2009). Blood
graded exercise in man. Am J Physiol Endocrinol Metab lactate is an important energy source for the human brain. J
249, E595–E602. Cereb Blood Flow Metab 29, 1121–1129.
Stanley WC, Gertz EW, Wisneski JA, Neese RA, Morris DL Vanderheyden LW, McKie GL, Howe GJ & Hazell TJ (2020).
& Brooks GA (1986). Lactate extraction during net lactate Greater lactate accumulation following an acute bout
release in legs of humans during exercise. J Appl Physiol 60, of high-intensity exercise in males suppresses acylated
1116–1120. ghrelin and appetite postexercise. J Appl Physiol 128,
Stanley WC, Wisneski JA, Gertz EW, Neese RA & Brooks 1321–1328.
GA (1988). Glucose and lactate interrelations during Viguie CA, Frei B, Shigenaga MK, Ames BN, Packer L &
moderate-intensity exercise in humans. Metabolism 37, Brooks GA (1993). Antioxidant status and indexes of
850–858. oxidative stress during consecutive days of exercise. J Appl
Steinman MQ, Gao V & Alberini CM (2016). The role of Physiol 75, 566–572.
lactate-mediated metabolic coupling between astrocytes Vrieze A, Van Nood E, Holleman F, Salojarvi J, Kootte RS,
and neurons in long-term memory formation. Front Integr Bartelsman JF, Dallinga-Thie GM, Ackermans MT, Serlie
Neurosci 10, 10. MJ, Oozeer R, Derrien M, Druesne A, Van Hylckama Vlieg
Stender S, Zaha VG, Malloy CR, Sudderth J, DeBerardinis JE, Bloks VW, Groen AK, Heilig HG, Zoetendal EG, Stroes
RJ & Park JM (2020). Assessment of rapid hepatic ES, de Vos WM, Hoekstra JB & Nieuwdorp M (2012).
glycogen synthesis in humans using dynamic (13)C Transfer of intestinal microbiota from lean donors increases
magnetic resonance spectroscopy. Hepatol Commun 4, insulin sensitivity in individuals with metabolic syndrome.
425–433. Gastroenterology 143, 913-916.e7.
Storey BT & Kayne FJ (1977). Energy metabolism of Wagner S, Hussain MZ, Hunt TK, Bacic B & Becker
spermatozoa. VI. Direct intramitochondrial lactate HD (2004). Stimulation of fibroblast proliferation by
oxidation by rabbit sperm mitochondria. Biol Reprod 16, lactate-mediated oxidants. Wound Repair Regen 12,
549–556. 368–373.
Suzuki A, Stern SA, Bozdagi O, Huntley GW, Walker RH, Wahren J & Ekberg K (2007). Splanchnic regulation of glucose
Magistretti PJ & Alberini CM (2011). Astrocyte-neuron production. Annu Rev Nutr 27, 329–345.
lactate transport is required for long-term memory Warburg O & Minami S (1923). Versuche an überlebendem
formation. Cell 144, 810–823. carcinom-gewebe. J Mol Med 2, 776–777.
Takahashi H, Alves CRR, Stanford KI, Middelbeek RJW, Warburg O, Wind F & Negelein E (1927). The metabolism of
Pasquale N, Ryan RE, Xue R, Sakaguchi M, Lynes MD, tumors in the body. J Gen Physiol 8, 519.
So K, Mul JD, Lee MY, Balan E, Pan H, Dreyfuss JM, Wasserman DH, Lickley HL & Vranic M (1984). Interactions
Hirshman MF, Azhar M, Hannukainen JC, Nuutila between glucagon and other counterregulatory hormones
P, Kalliokoski KK, Nielsen S, Pedersen BK, Kahn CR, during normoglycemic and hypoglycemic exercise in dogs. J
Tseng YH & Goodyear LJ (2019). TGF-beta2 is an Clin Invest 74, 1404–1413.
exercise-induced adipokine that regulates glucose and fatty Wasserman K & McIlroy MB (1964). Detecting the threshold
acid metabolism. Nat Metab 1, 291–303. of anaerobic metabolism in cardiac patients during exercise.
Teramae H, Yoshikawa T, Inoue R, Ushida K, Takebe Am J Cardiol 14, 844–852.
K, Nio-Kobayashi J & Iwanaga T (2010). The cellular Watt PW, MacLennan PA, Hundal HS, Kuret CM & Rennie
expression of SMCT2 and its comparison with other trans- MJ (1988). L(+)-lactate transport in perfused rat skeletal
porters for monocarboxylates in the mouse digestive tract. muscle: kinetic characteristics and sensitivity to pH and
Biomed Res 31, 239–249. transport inhibitors. Biochim Biophys Acta 944, 213–222.
Theytaz F, de Giorgi S, Hodson L, Stefanoni N, Rey V, Wilson DF, Rumsey WL, Green TJ & Vanderkooi JM (1988).
Schneiter P, Giusti V & Tappy L (2014). Metabolic fate of The oxygen dependence of mitochondrial oxidative
fructose ingested with and without glucose in a mixed meal. phosphorylation measured by a new optical method
Nutrients 6, 2632–2649. for measuring oxygen concentration. J Biol Chem 263,
Torres-Fuentes C, Golubeva AV, Zhdanov AV, Wallace S, 2712–2718.
Arboleya S, Papkovsky DB, El Aidy S, Ross P, Roy BL, Wisneski JA, Gertz EW, Neese RA & Mayr M (1987).
Stanton C, Dinan TG, Cryan JF & Schellekens H (2019). Myocardial metabolism of free fatty acids. Studies with
Short-chain fatty acids and microbiota metabolites attenuate 14C-labeled substrates in humans. J Clin Invest 79, 359–366.
ghrelin receptor signaling. FASEB J 33, 13546–13559. Woerle HJ, Meyer C, Dostou JM, Gosmanov NR, Islam N,
Tsukamoto S, Shibasaki A, Naka A, Saito H & Iida K (2018). Popa E, Wittlin SD, Welle SL & Gerich JE (2003). Pathways
Lactate promotes myoblast differentiation and myotube for glucose disposal after meal ingestion in humans. Am J
hypertrophy via a pathway involving MyoD in vitro and Physiol Endocrinol Metab 284, E716–E725.
enhances muscle regeneration in vivo. Int J Mol Sci 3649, Wu BU, Hwang JQ, Gardner TH, Repas K, Delee R, Yu S,
19. Smith B, Banks PA & Conwell DL (2011). Lactated Ringer’s
Urry LA, Cain ML, Wasserman SL, Minorsky PV, & Orr R solution reduces systemic inflammation compared with
(2020). Campbell biology. In Campbell Biology, 12 edn, pp. saline in patients with acute pancreatitis. Clin Gastroenterol
164–170.Pearson. Hepatol 9, 710–717.e1.

© 2021 The Authors. The Journal of Physiology published by John Wiley & Sons Ltd on behalf of The Physiological Society
14697793, 2022, 5, Downloaded from https://physoc.onlinelibrary.wiley.com/doi/10.1113/JP280955 by Schweizerische Akademie Der, Wiley Online Library on [10/11/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
J Physiol 600.5 Lactate, phoenix risen 1251

Xie Q, Zhu Z, He Y, Zhang Z, Zhang Y, Wang Y, Luo J, Peng for authorship, and all those who qualify for authorship are
T, Cheng F & Gao J (2020). A lactate-induced Snail/STAT3 listed.
pathway drives GPR81 expression in lung cancer cells.
Biochim Biophys Acta 1866, 165576.
Zhang D, Tang Z, Huang H, Zhou G, Cui C, Weng Y, Liu W, Funding
Kim S, Lee S, Perez-Neut M, Ding J, Czyz D, Hu R, Ye Z,
He M, Zheng YG, Shuman HA, Dai L, Ren B, Roeder RG, Supported by NIH 1 R01 AG059715-01, Pac-12 Conference
Becker L & Zhao Y (2019). Metabolic regulation of gene Grant No. 3-02-Brooks-17 and the UCB Centre for Research
expression by histone lactylation. Nature 574, 575–580. and Education on Aging (CREA) to G.A.B. There are no other
Zinker BA, Britz K & Brooks GA (1990). Effects of a 36-hour funding sources.
fast on human endurance and substrate utilization. J Appl
Physiol 69, 1849–1855.
Acknowledgements

Additional information Michael Horning and Andrea Salvador Pascual are thanked for
reading and commenting on the manuscript.
Competing interests
The authors have no competing interests. Keywords
exercise, fibre type, gene adaptation, gluconeogenesis,
Author contributions glycogenolysis, indirect pathway, lactate shuttle, lactate
All authors contributed to writing of the paper and approve signalling, microbiome, muscle, postabsorptive metabolism,
of the final version. All persons designated as authors qualify postprandial metabolism, satiety

© 2021 The Authors. The Journal of Physiology published by John Wiley & Sons Ltd on behalf of The Physiological Society

You might also like