ArticlePDF AvailableLiterature Review

Pyruvate in the Correction of Intracellular Acidosis: A Metabolic Basis as a Novel Superior Buffer

Authors:
  • Fresenius Med Care

Abstract

The review focuses on biochemical metabolisms of conventional buffers and emphasizes advantages of sodium pyruvate (Pyr) in the correction of intracellular acidosis. Exogenous lactate (Lac) as an alternative of natural buffer, bicarbonate, consumes intracellular protons on an equimolar basis, regenerating bicarbonate anions in plasma while the completion of gluconeogenesis and/or oxidation occurs via tricarboxylic-acid cycle in mitochondria mainly in liver and kidney, or heart. The general assumption that Lac is 'metabolized to bicarbonate' in liver to serve as a buffer has been questioned. Pyr as a novel buffer would be superior to conventional ones in the correction of metabolic acidosis. Several likely biochemical mechanisms of Pyr action are discussed. Experimental evidence, in vivo, strongly suggested that Pyr would be particularly efficient in the correction of severe acidemia: type A lactic acidosis, hypercapnia with cardiac arrest, and diabetic and alcoholic ketoacidosis in animal experiments and clinic settings. Because of its multi-cytoprotection, Pyrs not only correct acidosis, but also benefit theunderlying dysfunction of vital organs. In addition, Pyr is also a potential buffer component of dialysis solutions. However, the instability of Pyr in aqueous solutions restricts its clinical applications as a therapeutic agent. Attempts to create a stable Pyr preparation are needed.
Fax +41 61 306 12 34
E-Mail karger@karger.ch
www.karger.com
In-Depth Topic Review
Am J Nephrol 2005;25:55–63
DOI: 10.1159/000084141
Pyruvate in the Correction of
Intracellular Acidosis: A Metabolic Basis
as a Novel Superior Buffer
Fang Qiang Zhou
Fresenius Dialysis Centers, Chicago, Ill. , USA
tion, Pyrs not only correct acidosis, but also benefi t the
underlying dysfunction of vital organs. In addition, Pyr is
also a potential buffer component of dialysis solutions.
However, the instability of Pyr in aqueous solutions re-
stricts its clinical applications as a therapeutic agent. At-
tempts to create a stable Pyr preparation are needed.
Copyright © 2005 S. Karger AG, Basel
Since 1970s, pyruvate (Pyr) has become increasingly
attractive in the protection of dysfunctional vital organs,
particularly in myocardial ischemia and reperfusion in-
jury, pointing to a potential therapeutic value for the dys-
functional myocardium [1–3] . Also, fi ndings strongly in-
dicated that Pyr would be a novel buffer in many clinic
settings [4–6] . This review discusses its distinctive effects
on intracellular pH (pHi), proposing that Pyr may be su-
perior to lactate (Lac) and bicarbonate in the correction
of severe intracellular acidosis.
Lactate Metabolism and Effects on pHi and
Lactic Acidosis
Lac has been traditionally accepted as the best replace-
ment for bicarbonate in the treatment of metabolic aci-
dosis since early 1930s [7]
; it has since become part of
solutions for dialysis and continuous renal replacement
therapy for several decades.
Key Words
Pyruvate Lactate Bicarbonate Glucose Ischemia
Intracellular pH Acidosis
Abstract
The review focuses on biochemical metabolisms of con-
ventional buffers and emphasizes advantages of sodium
pyruvate (Pyr) in the correction of intracellular acidosis.
Exogenous lactate (Lac) as an alternative of natural buf-
fer, bicarbonate, consumes intracellular protons on an
equimolar basis, regenerating bicarbonate anions in
plasma while the completion of gluconeogenesis and/or
oxidation occurs via tricarboxylic-acid cycle in mitochon-
dria mainly in liver and kidney, or heart. The general as-
sumption that Lac is ‘metabolized to bicarbonate’ in liver
to serve as a buffer has been questioned. Pyr as a novel
buffer would be superior to conventional ones in the cor-
rection of metabolic acidosis. Several likely biochemical
mechanisms of Pyr action are discussed. Experimental
evidence, in vivo, strongly suggested that Pyr would be
particularly effi cient in the correction of severe acidemia:
type A lactic acidosis, hypercapnia with cardiac arrest,
and diabetic and alcoholic ketoacidosis in animal experi-
ments and clinic settings. Because of its multi-cytoprotec-
Received: June 24, 2004
Accepted: January 6, 2005
Published online: February 23, 2005
Nephrolo
gy
American Journal of
Dr. Fang Q. Zhou
Fresenius Dialysis Centers at Chicago
4180 Winnetka Avenue
Rolling Meadows, IL 60008 (USA)
Tel. +1 847 394 6250, Fax +1 847 394 4621, E-Mail fqz@hotmail.com
© 2005 S. Karger AG, Basel
0250–8095/05/0251–0055$22.00/0
Accessible online at:
www.karger.com/ajn
The opinions or assertions contained herein are the author’s pri-
vate view and are not to be construed as refl ecting the view of Fre-
senius Dialysis Centers at Chicago, USA .
Downloaded by:
68.75.176.9 - 6/26/2014 8:37:00 AM
Zhou
Am J Nephrol 2005;25:55–63
56
However, the widely propagated theory that Lac is
‘metabolized to bicarbonate’ on an equimolar basis in
liver represents a simplifi cation. In fact, Lac is an end
product of anaerobic glycolysis with Pyr as its only outlet.
As an alkalizer, it must be converted to Pyr by lactic de-
hydrogenase (LDH) in most mammalian tissues, which
in turn can either be completely oxidized to CO
2
and H
2
O
in the mitochondria mainly in liver, kidney, heart and
brain for the titration of a proton or enter gluconeogen-
esis from two Lac anions in the mitochondria and cytosol
of liver and kidney for the titration of two protons. Both
Lac oxidation and Lac-based gluconeogenesis are a mito-
chondrial tricarboxylic-acid cycle (TCA cycle)-dependent
pathway. The overall reactions can be described as shown
in equations 1 and 2; whichever route of Lac disposal is
followed, one proton (H
+
) is consumed per Lac anion me-
tabolized (Lac
:
H
+
= 1: 1):
oxidation) ATP, 17.5 ( OH 3 CO 3 O 3 H
lactate
CHOHCOOCH 22
cycle TCA
23
_
enesis)(gluconeog OHC
H 2
lactate
CHOHCOOCH 2 61263
_
o
o
1)
(2)
(
The consumption of a proton in cytosol effectively
means the regeneration of a bicarbonate anion, thus rais-
ing pHi and preserving the blood bicarbonate level stoi-
chiometrically [8, 9] .
The uptake of Lac or Pyr by cells and the entry of Pyr
into mitochondria depend on the Lac
/H
+
cotransport
system, monocarboxylate transporters (MCT, g. 1 ) with
H
+
([H
+
] in g. 1 ) fl ux in symport. The system is located
in the cytoplasma membrane and inner membrane of mi-
tochondria. The oxidation of exogenous Lac to Pyr by
LDH is NAD
+
-dependent in cytosol, which releases two
hydrogen ions with one (H
+
in g.1 ) left in the hydrogen
pool of cytosol as shown in equation 3. Both MCT-trans-
ported and Lac-released protons initially decrease pHi
[10, 11] , while the other hydrogen ion (negative hydrogen
or hydride ion) released from Lac reduces cytosolic NAD
+
([NAD
+
]c) to NADH ([NADH]c). Such a decrease of re-
ducing equivalents has been called cellular ‘pseudohy-
poxia’. In aerobic conditions, [NADH
2
]c ([NADH +
H
+
]c) may be transferred into mitochondria and oxidized
with the generation of 1 H
2
O and 2.5 ATP. Thus, the
proton released from Lac is eventually consumed. The
administration of Lac raises the Lac/Pyr ratio and de-
creases the [NAD
+
/NADH]c ratio in cytosol, which may
critically inhibit glycolysis at the level of glyceraldehyde-
3-phosphate dehydrogenase (G-3-PD) [12] . It also reduc-
es the glycolytically derived ATP production that is vital
for sustaining cellular functions in anaerobic conditions.
Pyr oxidative decarboxylation, then, is catalyzed by the
Pyr dehydrogenase complex (PDC) in mitochondria,
coupled with the reduction of mitochondrial NAD
+
([NAD
+
]m) to [NADH]m, yielding acetyl-CoA. It de-
pends on oxygen as an electron acceptor in mitochondria
that one pair of [NADH
2
]m ([NADH + H
+
]m) as well as
transferred [NADH
2
]c is oxidized through the electron
transfer chain, consuming the two hydrogen ions of
[NADH
2
]m with 1 H
2
O and 3 ATP generations.
Catalyzed by PDC, one hydrogen (hydride) ion of
[NADH + H
+
]m is transferred via [FADH
2
] from the
reactive thiol group (-SH) of coenzyme A, HS-CoA,
which does not affect pHi; the other hydrogen ion (pro-
ton) is picked up in the hydrogen pool in the mitochon-
dria matrix [13] . The overall reaction is the oxidation of
imported proton via MCT and raises pHi as reported in
cardiomyocytes of ischemic reperfused rabbit hearts and
failing human myocardium [10, 11] . Thus, for every ex-
ogenous Lac anion metabolized in aerobic conditions
either by oxidative phosphorylation or with glyconeo-
genesis, one endogenous proton is consumed, leading to
the preservation of blood bicarbonate levels on a 1: 1 mo-
lar basis.
In the mitochondria of many cell lines, mainly hepa-
tocytes, carbonic anhydrase isoenzyme (CA V) exists,
which catalyzes hydration (-HCO
3
or H
2
CO
3
) of CO
2
and
H
2
O.
The bicarbonate so generated in the mitochondria
is vital for the activity of bicarbonate-requiring Pyr car-
boxylase (PC) that is essential for the TCA cycle and glu-
coneogenesis [14, 15] . Also, acetazolamide, an inhibitor
of most CA isoenzymes including CA V, may decrease
Lac-induced blood alkalinization and induce lactic aci-
dosis [15] . Although these facts support that CA V plays
an important role in the pHi modulation, they do not
prove the general assumption that ‘the irreversible oxida-
tion of Lac generates alkali from the conversion of CO
2
to bicarbonate by CA’, which then corrects metabolic ac-
idosis [16, 17] . The traditional concept that Lac as a buf-
fer is ‘metabolized to bicarbonate’ in liver mitochondria
catalyzed by CA V to correct acidosis lacks direct evi-
dence. It is well known that the Lac oxidation induces
cytotoxic effects in tissue ischemia or hypoxia, such as the
depletion of reducing equivalents, inhibition of glycolysis
and release of protons in cytosol. Hence, in hypoxic con-
ditions, the administration of sodium Lac may aggravate,
rather than attenuate, intracellular acidifi cation if the
ability of Pyr oxidation is limited and gluconeogenesis is
suppressed in cells such as hepatocytes, cardiomyocytes
and macrophages, which have abundant mitochondria.
Downloaded by:
68.75.176.9 - 6/26/2014 8:37:00 AM
Pyruvate and Metabolic Acidosis
Correction
Am J Nephrol 2005;25:55–63 57
NAD
+
NADH + H
+
NADH + H
+
NAD
+
lactate/pyruvate + [H
+
]
Lactate
acetyl-CoA
oxaloacetate
citrate
malate
CO
2
cytoplasm
mitochondrial
matrix pyruvate
MCT
oxaloacetate
LDH
2-ketoglutarate
glycolysis
oxidative
phosphorylation
extracellular
space
PDC
pyruvate
phosphoenolpyruvate
MCT
PC
ME
Fructose-1,6-bisphosphate
anaplerosis
1,3 bisphosphoglycerate
glyceraldehyde 3-phosphate
glucose
NADH + [H
+
]
NADH + H
+
respiratory chain
NADH + [H
+
]
[H
+
]
[H
+
]
G-3-PD
glucose
O
2
NAD
+
+
lactate/pyruvate + [H
+
]
TCA cycle
alanine
oxidation of ketone bodies
and fatty acids
CoA-SH
CO
2
PFK-1
(2)
(1)
H
2
O + CO
2
+ ATP
pKa
Km
Fig. 1. Pyruvate metabolic pathways and their relationship with the
consumption of protons in intracellular compartments. Pyr and/or
Lac enter cytosol via the MCT system with an extracellular [H
+
]
ex in symport. The oxidation of Lac to Pyr with LDH releases an
H
+
coupled with NAD
+
reduction to NADH in cytosol, inhibiting
glycolysis at the G-3-DP step. Both MCT-imported [H
+
] and Lac-
released H
+
initially decrease pHi, whereas the Pyr reduction to Lac
consumes the [H
+
], restoring pHi and glycolysis in anaerobic con-
ditions. In aerobic conditions, the Pyr entry into mitochondria with
[H
+
] yields NADH + [H
+
] by PDC. Oxidative phosphorylation of
both NADH + [H
+
] ([NADH + H
+
]m) and transferred NADH +
H
+
([NADH + H
+
]c) generates ATP and H
2
O, eventually consum-
ing [H
+
] and H
+
, respectively. The overall oxidation of exogenous
Pyr and/or Lac consumes endogenous [H
+
] on an equimolar basis,
raising pHi and pHe. Excess Pyr simultaneously activates the PDC
activity and enhances anaplerotic pathways via PC, accelerating
the TCA cycle fl ux and the [H
+
] consumption. Pyr-based gluconeo-
genesis consumes an additional pair of [NADH + H
+
]c at the step
of G-3-DP in cytosol, compared with Lac-based gluconeogenesis.
The preservation of a near physiologic pHi and the acceleration of
TCA cycle by excess Pyr enhance the Lac oxidation in lactic acido-
sis and the oxidation of ketone bodies and fatty acids in ketoacido-
sis. The shuttle of [NADH + H
+
]c transference from cytosol to
mitochondria is not shown. See the text for details. (1) = Gluconeo-
genesis-coupled reactions in mitochondria; (2) = gluconeogenesis-
coupled reactions in cytosol; G-3-PD = glyceraldehyde-3-phos-
phate dehydrogenase; K m = Michaelis constant; LDH = lactic de-
hydrogenase; MCT = monocarboxylate transporters; ME = malic
enzyme; NAD
+
= oxidized nicotinamide adenine dinucleotide;
NADH = reduced nicotinamide adenine dinucleotide; PC = pyru-
vate carboxylase; PDC = pyruvate dehydrogenase complex;
PFK-1 = phosphofructokinase-1; pK a = dissociation constant;
TCA cycle = tricarboxylic-acid cycle.
Downloaded by:
68.75.176.9 - 6/26/2014 8:37:00 AM
Zhou
Am J Nephrol 2005;25:55–63
58
In cells with no or a few mitochondria (e.g., erythrocytes
or neutrophils, etc.), exogenous Lac would just deterio-
rate the acidosis and depress cell function [18] even in
normoxymia.
Hyperlactatemia and lactic acidosis could be corrected
with dichloroacetate (DCA) or THAM [16, 19] , but their
values in clinic settings are not clear due to the negative
survival rate and/or potential toxicity [19, 20] . The effi -
cacy of Lac therapy in patients with lactic acidosis has
not been systemically evaluated. In principle, severe aci-
demia: Type A lactic acidosis, hypercapnia with cardiac
arrest and diabetic ketoacidosis, which account for over
90% of severe acidosis in clinic settings with excess mor-
tality [19] , are contraindications of the Lac administra-
tion. The Lac infusion should be avoided in cases of el-
evated serum Lac. Although sodium Lac itself is almost
obsolete in the treatment of clinical acidosis, several re-
cent studies have demonstrated that lactated Ringer’s so-
lution (28 m M ) was benefi cial in the resuscitation of mas-
sive hemorrhagic shock accompanied with lactic acidosis.
It signifi cantly improved acidosis and survival rates [17,
21–23] . In rat working hearts after hemorrhagic shock ac-
companied with lactic acidosis and impaired PDC ac-
tivities, Lac at 8.0 m M in the presence of palmitate in
perfusates saturated with 95% oxygen improved cardiac
effi ciency [21] . Most likely the intracellular acidosis was
attenuated although pHi data were not determined. The
paradoxical effect of exogenous Lac on experimental lac-
tic acidosis above has not been fully interpreted, but may
be associated with several reasons: different species of
models, early stage of shock, over 40 mm Hg of mean ar-
terial pressure of shock and anesthesia conditions [17,
21–23] . The above fi ndings indicated that the TCA cycle
function in vital organs remained, at least partially, intact
for Lac oxidation [24] . Notably, Lac-based gluconeogen-
esis in liver and kidney may still be preserved in the ear-
ly stage of shock [25, 26] . In particular, kidney is as im-
portant a gluconeogenic organ as liver for Lac disposal. It
is estimated that one-half of glucose production within
the Lac Cori cycle takes place in the kidney cortex and
Lac accounts for approximately 50% of renal gluconeo-
genesis. The Lac uptake remains relatively stable in hem-
orrhagic shock dogs until blood loss reaches 40% of total
blood volume [26, 27] . While acidosis signifi cantly de-
presses hepatic uptake of Lac, renal gluconeogenesis is
markedly enhanced by ischemic injury or acidosis [26,
28] due to the rapid stimulation of phosphoenolpyruvate
carboxykinase [29] . Further, a recent clinical investiga-
tion found that in patients with early postoperative car-
diogenic shock accompanied with lactic acidosis, the
clearance of exogenous Lac was not signifi cantly altered,
compared with controls [30] . In contrast, if shock despite
causes in origin are severe [31] or combined with alco-
holic intoxication [23] , preexisting liver diseases, diabetes
or multiple organ injuries, the Lac infusion most likely
exacerbates lactic acidosis. Therefore, it should be taken
into account in the deduction of Lac effects on acidosis
in above-mentioned experimental results if the oxidative
phosphorylation of TCA cycle or gluconeogenesis in vital
organs was, at least in part, sustained during early shock
(hypoxia) or recovered after shock (reoxygenation). On
the other hand, because the anoxia (hypoxia) of tissues
and the dysfunction of TCA cycle commonly coexist in
lactic acidosis, exogenous Lac should be avoided in such
clinical settings even in the early stage of shock if the pa-
tient is critically ill, preferably in consideration of a new
buffer, Pyr, instead.
Pyruvate Improvement of Intracellular
Acidosis
Pyr was initially used in mid-1970 to buffer intracel-
lular hydrogen ions and improve redox status in cardio-
myocytes. The administration of exogenous Pyr promis-
ingly improved glycolysis that was depressed from isch-
emia-reperfusion injury, preserved cardiac contractile
function and prolonged the survival time of ischemic
swine hearts [32] . Also, Pyr (10 m M ), as a sole exogenous
substrate, signifi cantly attenuated the pHi decrease in the
perfused rat ischemic myocardium. It was fi rst discovered
in 1994 that animals treated with the Pyr infusion did not
develop an anesthetic agent-induced mild metabolic aci-
dosis presented in controls [33] . The fi nding confi rmed
that the intravenous Pyr would induce ‘systemic alkaliza-
tion’ that was not emphasized in its conception until 1999
[34] . The comparison of Pyr with Lac on pHi in human
cells was fi rst conducted with neutrophils in 1995 [4] . In
an acidic milieu, following a slight pHi decrease, Pyr
(40 m M ) maintained neutrophilic pHi close to a physio-
logical value, whereas an identical Lac level brought about
a prompt and substantial pHi decline. The extracellular
pH [pHe] of mixture solutions with Pyr was signifi cantly
higher than that with Lac counterparts as well. A subse-
quent report confi rmed the phenomenon [Perit Dial Int
Abstracts, 1997; 17(suppl 1):S33]. The impact of Pyr on
systemic acidosis, however, at present has not been well
appreciated [31] , and potential mechanisms whereby Pyr
preferably corrected severe acidosis have not been fully
elucidated either [6, 34] . There are several likely bio-
Downloaded by:
68.75.176.9 - 6/26/2014 8:37:00 AM
Pyruvate and Metabolic Acidosis
Correction
Am J Nephrol 2005;25:55–63 59
chemical sites on which the metabolism of exogenous Pyr
consumes intracellular protons, indicating that Pyr is of
particular signifi cance in the pHi modulation [32, 35–37]
( g. 1 ):
(1) By reducing to Lac with LDH, one exogenous Pyr
anion consumes one proton from the cytosolic hydrogen
pool, raising [NAD
+
/NADH]c and pHi, as described in
equation 3:
NAD
lactate
CHOHCOOCH H NADH
pyruvate
COCOOCH __
3
LDH
3
om (3
)
The [NADH/NAD
+
]c ratio by the law of mass action
is the major determinant of reactive direction. For any
given cellular level of Pyr, the ratio determines what
fraction of Pyr is converted to Lac. When mitochondri-
al oxidative reactions are impaired, the ratio rises, and
the conversion of Pyr to Lac increases. Pyr inhibits gly-
colysis in aerobic conditions, however, it is of pivot im-
portance that Pyr favors anaerobic glycolysis due to the
increase of [NAD
+
/NADH]c ratio and restoration of
pH-sensitive phosphofructokinase (PFK-1) activity [5,
32, 37] . This would preserve glycolytic ATP production
that is essential for maintaining sarcoplasmic mem-
brane function, modulating intracellular protons [38,
39] , and prolonging cell survival of vital organs during
hypoxia [2, 40] . In the swine model of hemorrhagic
shock manifested lactic acidosis, the intravenous infu-
sion of Pyr further raised blood Lac levels, indicating
that the Pyr reduction occurred in cytosol, but lowered
the blood Lac/Pyr ratio and effi ciently avoided or cor-
rected lactic acidosis [6, 34, 41, 42] .
Accordingly, Pyr ‘systemic alkalization’ proposed
originally referred to this cytoplasmic reduction and the
mitochondrial oxidation of exogenous Pyr [34] . In this
respect, Pyr may also consume a proton in neutrophilic
cytosol with the reduction to Lac in addition to its weak-
er buffering capacity, both raising neutrophilic pHi [4] .
(2) By oxidizing into CO
2
and H
2
O via the TCA cycle
in mitochondria, Pyr anions also consume protons on an
equimolar basis (Pyr
:H
+
= 1: 1) as equation 4 shows:
oxidation) ATP, 15 ( OH 2 CO 3 O ½ 2 H
pyruvate
COCOOCH 22
cycle TCA
23
_
o
( 4)
As mentioned above, the exogenous Pyr consumes en-
dogenous protons stoichiometrically in both anaerobic
glycolysis and aerobic oxidation, raising pHi, and Pyr
does not release a proton as Lac does, but merely con-
sumes it in the oxidation. In addition, Pyr oxidizes with
a less oxygen consumption rate, compared with Lac.
(3) Supraphysiologic doses of exogenous Pyr would
greatly stimulate the activity of PDC although there has
been no data from human beings yet. It has been demon-
strated with various animal tissues in rat, rabbit, bovine
and swine that Pyr at several millimolars simultaneously
stimulated the PDC activity by inhibiting Pyr dehydro-
genase kinase in association with the improvement of car-
diac performance and cerebral and hepatic functions [6,
41–43] .
(4) Although Lac may replenish the TCA cycle by the
malate-aspartate shuttle, an anaplerotic pathway that
transfers Lac-reduced [NADH
2
]c to mitochondria [21] ,
it is with Pyr, other than Lac or other keto acids, that
anaplerosis via carboxylation can be carried out by PC to
oxaloacetate or by malic enzyme to malate. In a near
physiological condition, Pyr cardoxylation may account
for, in vivo, only 5% of the citrate formation in swine
hearts [44] . However, it may increase as many as 7 times
in perfused rat hearts [24] . The increased availability of
Pyr as a substrate and the PDC stimulation together with
enhanced anaplerotic pathways would greatly increase
the TCA cycle fl ux and accelerate the consumption of
protons and the ATP generation. In addition, anaplerosis
pathways, per se, fi x CO
2
in mitochondria. Importantly,
during acute acidosis Pyr may increase the CO
2
xation
in the mitochondria of liver and kidney [45] , which may
be of signifi cance in improving local hypercapnia in isch-
emic tissues.
(5) Gluconeogenesis is an energy-requiring process. It
consumes equivalently 3 high-energy phosphate bonds (2
ATP and 1 GTP) for 1 mol of glucose synthesized from
1 mol of Lac. However, the experimental evidence in pHi
within the rat hepatic lobule revealed that gluconeogen-
esis from Lac is a proton-consuming process [46] . Com-
pared with one proton consumed by every Lac anion in
Lac-based gluconeogenesis (equation 1), every Pyr anion
uses two protons in Pyr-based gluconeogenesis (Pyr
:H
+
= 1: 2). In the cytosolic process of gluconeogenesis, a pair
of [NADH + H
+
]c is required at the step of G-3-PD. It is
supplied by the Lac oxidation to Pyr in Lac-based gluco-
neogenesis, but is additionally consumed in Pyr-based
gluconeogenesis. Hence, Pyr-based gluconeogenesis in
liver and kidney would be of particular signifi cance in the
preservation of a near physiologic arterial pH (pHa) in
shock. In this regard, it was affi rmed with rat hepatic in-
tralobular mapping results that protons are consumed
stoichiometrically when weak acids are converted into
neutral products (glucose, CO
2
and H
2
O) [46] . The state-
ment may be the case of Lac, other than the case of Pyr
in terms of gluconeogenesis.
Downloaded by:
68.75.176.9 - 6/26/2014 8:37:00 AM
Zhou
Am J Nephrol 2005;25:55–63
60
As mentioned earlier, the entry of Lac or Pyr into cells
is carried by a proton-linked MCT family, which includes
seven isoforms in a range of mammalian cells. They ex-
hibit distinct cellular localization and have slightly differ-
ent properties. Isoforms act as Lac
/H
+
symport with a
1: 1 stoichiometry. In contrast to the MCT in human neu-
trophils, which has a similar affi nity for both Lac and Pyr
[5] , the isoforms of hepatocytes and cardiomyocytes in
rats have much higher Michaelis constant ( K m) values for
L-Lac than those for Pyr (4.7 and 2.74 m M vs. 1.3 and
0.2 m M ), respectively [47, 48] . Thus, Lac may more read-
ily induce a pHi decline than Pyr when high extracellular
concentrations of both are comparable in human vital
organs. On the other hand, Pyr has a weaker buffering
capacity due to the dissociation constant ( pK a) of 2.49,
compared with Lac pK a of 3.9 [4] . The weaker buffering
capacity usually results in a higher pH value. Owing to
these properties, equimolar doses of external Pyr and Lac
are not expected to result in equivalent pHi reductions in
human cells as shown in neutrophils [4] . In rat cardio-
myocytes, the property of MCT K m was also considered
in the Pyr effect on pHi [49] .
Based on its biochemical and biophysical characteris-
tics, Pyr may be valuable in the treatment of severe met-
abolic acidosis such as type A lactic acidosis, hypercapnia
with cardiac arrest, diabetic ketoacidosis and alcoholic
intoxication although no clinic data have been reported
yet. In theory, Pyr with insulin might be a more effi cient
regime in the correction of diabetic ketoacidosis mainly
due to their potential synergetic effects on the oxidation
of ketone bodies. Pyr in the regime might be also valuable
to restore the sensitivity of insulin receptors inhibited by
severe acidosis and reduce the insulin dosage to avoid
hypoglycemia. Moreover, because of its better consump-
tion of intracellular protons in the absence of the direct
production of CO
2
in hypoxia, Pyr might be also benefi -
cial in respiratory acidosis. However, these deductions
require experimental and clinical validations.
Pyruvate Superiority in the Correction of
Acidosis
Pyr and Lac in the Correction of Metabolic Acidosis
The effect of intravenous Pyr on acid-base balance was
compared with equimolar Lac in anesthetized dogs in late
1970s. Lac infusion was associated with a signifi cantly
slower rate of bicarbonate level rising in comparison with
Pyr, indicating that the additional metabolic steps are
required [50] . The oxidation of Lac-reduced [NADH
2
]c
may take place either in cytoplasm-coupled reactions as-
sociated with gluconeogenesis, or a mitochondrial route
in the presence of oxygen. In addition to the difference of
the proton consumption in gluconeogenesis processes be-
tween Pyr and Lac, the mitochondrial route is an indirect
pathway, which depends on the reducing equivalent of
carrier systems. The malate-aspartate shuttle is the major
step involved in transporting [NADH + H
+
]c into the mi-
tochondria in cells of heart, liver and kidney. The shuttle
operation is driven by the electrical potential across mi-
tochondrial membrane. Thus, the oxidation of Lac to Pyr
is an energy-dependent step and operates slowly (in min-
utes) in physiological conditions [51] . In perfused rabbit
cardiomyocytes, the pHi recovery was delayed by 12 min.
with Lac-added perfusates compared with Pyr-added
counterparts in the presence of DCA [10] . This rate-lim-
ited step, which would display while the [NADH
2
]c load
is increased, may be another interpretation of the retarda-
tion of pHi recovery in addition to the elevated proton
load, per se, from the Lac oxidation and the [NADH]c
increment [10] . Although the ratio of [NAD
+
/NADH]c
determines the balance between Pyr and Lac (equation
3), the reaction is in favor of the direction from Pyr to
Lac as the lesser rise in blood Pyr concentration with Lac
infusion than in blood Lac level with equal Pyr infusion
[50] . Several recent fi ndings evidently supported that Pyr
would be superior to Lac in the treatment of clinic meta-
bolic acidemia including lactic acidosis. In the rat perito-
neal dialysis model, preliminary results revealed that ure-
mic acidosis was better corrected and the functions of
peritoneal cells were more preserved with Pyr-buffered
dialysates than with Lac counterparts [J Am Soc Nephrol
Abstracts, 1998; 9: 236A; also Chin J Nephrol 2001; 17:
365–368]. Also, in the rat model of severe hemorrhagic
shock manifested lactic acidosis, commonly seen in a
clinical setting, the intravenous infusion of Pyr improved,
whereas Lac exacerbated, metabolic acidosis. Pyr Ring-
er’s solution resulted in a higher blood base excess level
and a lower blood Lac level, compared with Lac Ringer’s
counterparts [31] . The study also substantiated that Pyr,
in vivo, signifi cantly decreased the expression of apopto-
sis markers, whereas Lac increased the markers after re-
suscitation in rat lungs [31] as observed, in vitro, in hu-
man neutrophils and endothelial cells [5, 52] . Although
mechanisms of Pyr attenuation of apoptosis in shock re-
suscitation are not fully understood, it is well recognized
that acidosis may be a triggering factor [5, 53] . Studies in
hemorrhagic shock repeatedly confi rmed that Pyr effec-
tively prevented or corrected severe lactic acidosis in
swine [6, 34, 41, 42] . A human trial demonstrated that
Downloaded by:
68.75.176.9 - 6/26/2014 8:37:00 AM
Pyruvate and Metabolic Acidosis
Correction
Am J Nephrol 2005;25:55–63 61
Pyr-fortifi ed cardioplegia solutions provided cardiopro-
tection superior to Lac-based counterparts during surgi-
cal cardiac arrest although pHi data were absent [54] .
These fi ndings strongly suggested that Pyr was preferable
in the correction of intracellular acidosis in both anaero-
bic and aerobic conditions.
In brief, because of the prevalence of LDH in mam-
malian cells, Pyr as an oxidant improves pHi and cellular
functions, which is opposite to the deleterious effects on
pHi and cytotoxic characteristics of Lac as a reductant in
molecular and cellular levels particularly in anaerobic
conditions. Meanwhile, due to the Lac oxidation to Pyr
with LDH, both of them also exhibit somewhat similar
metabolic and biological effects including the modulation
of pHi in aerobic conditions.
Pyr and Bicarbonate in the Treatment of Metabolic
Acidosis in Cardiogenic Shock and Dialysis
Bicarbonate as a natural buffer was not questioned in
the treatment of metabolic acidosis until two decades ago.
The bicarbonate corrects acidosis through chemical neu-
tralization, which depends on a fair respiratory function,
whereas Pyr exerts a metabolic titration of intracellular
acidosis without directly producing CO
2
. Bicarbonate of-
ten benefi ts patients with metabolic acidosis in the ab-
sence of tissue hypoxia (e.g., uremic, diarrheic, and renal
tubular acidosis). However, patients with cardiac arrest
or cardiogenic shock (or septic shock, hepatic failure and
diabetic ketoacidosis, etc.) usually have impaired tissue
oxygen delivery. In these circumstances, the primary
causes of acidosis result from the accumulation of lactic
acid, the proton release from ATP hydrolysis and the re-
tardation of local tissue CO
2
(‘metabolic’ hypercapnia).
And limited glycolytic ATP production may lead to a
progressive pHi decline to !
6.0 in cardiomyocytes [19,
53] . Bicarbonate administration in hypoxic states does
not correct the underlying tissue hypoxia and is generally
not successful in improving the acidosis or patients’ gen-
eral conditions. It usually results in severe adverse effects,
leading to the exacerbation of hypercapnia and lactic ac-
idosis and further decrease of tissue pHi in liver, brain,
skeletal muscle and red cells [19, 55] . In the presence of
high P
CO2
and high non-bicarbonate buffers in plasma,
bicarbonate, per se, would considerably decrease pHi al-
though pHa may increase due to the readily entrance of
CO
2
(but not -HCO
3
) into intracellular spaces [56] . Nev-
ertheless, severe acidosis (pHa ! 7.1) must be timely cor-
rected at any clinical circumstances although mild acido-
sis may be benefi cial. Without an ideal base in the clinic
setting, it is strongly recommended that the bicarbonate
infusion should be cautious in the management of cardio-
pulmonary arrest in order to maintain pHa in 7.10–7.20
after the airway is established [57] . Clinical evidence also
revealed that bicarbonate therapy provided little im-
provement of diabetic ketoacidosis and suggested that it
should not be used unless pHa is !
7.0 [19, 58] . On the
other hand, DCA, which stimulates the PDC activity by
directly inhibiting PDC kinase as one of Pyr actions and
corrects lactic acidosis as a specifi c agent, also can sig-
nifi cantly improve myocardial performance and acidosis
in hypoxia [14, 20] . In comparison, Pyr may be superior
to bicarbonate or DCA in the treatment of hypoxic aci-
dosis. Its unique metabolic and chemical characteristics
may be ideal for the correction of severe acidosis in car-
diogenic shock or hypercapnia with cardiac arrest as it
consumes protons and benefi ts in myocardium perfor-
mance without risking pHi deterioration in anaerobic
conditions. Further, Pyr protects cellular functions of
multiple cell lines and vital organs as a strong antioxidant.
It also inhibits infl ammation mediators (e.g., TNF-
, NF-
B and NO), improves Ca
2+
homeostasis, attenuates
apoptosis and preserves carbohydrates metabolic path-
ways in hyperglycemia and promotes lipid metabolism
[2, 3, 5, 6, 52] . Pyr as an intermediary metabolite of glu-
cose is also an energy-yielding substrate without any
known cytotoxicity. An intravenous Pyr loading test dem-
onstrated its safety in human subjects [59] .
In addition, Pyr might be also a promising buffer to
correct uremic acidosis in patients undergoing peritoneal
dialysis (PD), which markedly improves the biocompat-
ibility of PD solutions [5, 36] . It may effi ciently correct
chronic acidotic status in hemodialysis patients as well,
which is not satisfactorily treated at present [60] . Pyr as
one of the buffer components may decrease bicarbonate
or Lac concentrations in dialysis solutions, reduce oxida-
tive stress induced by high bicarbonate levels [61] and
Lac toxicity. It can also improve glucose and lipid me-
tabolism in dialysis patients particularly with cardiovas-
cular complications and diabetes. However, the instabil-
ity of sodium Pyr in aqueous solutions restricts its clinical
applications at the present time. It is well known that Pyr
in aqueous solutions rapidly undergoes an aldol-like con-
densation, forming parapyruvate to inhibit mitochondri-
al functions [62] . Efforts to overcome Pyr instability in
solutions creating Pyr therapeutic preparations suitable
for intravenous infusion and dialysis are urgently war-
ranted. Several clues point to a possibility.
In conclusion, Pyr as a novel generation of buffers
would be superior to conventional ones both in animal
experiments and in clinic settings, particularly in the
Downloaded by:
68.75.176.9 - 6/26/2014 8:37:00 AM
Zhou
Am J Nephrol 2005;25:55–63
62
treatment of severe acidemia, which still lacks effective
agents to deal with at present. Intravenous administra-
tion of Pyr would not only correct both pHa and pHi and
acidotic symptoms, but also improve underlying cell dys-
functions to a large extent in critically ill patients with
heart, liver, kidney or brain disorders. Further evaluation
of Pyr effects on acid-base balance disorders and their
underlying mechanisms in both animals and clinical tri-
als is required. Attempts to create stable therapeutic prep-
arations of Pyr solutions are warranted.
Acknowledgements
The author is indebted to Dr. Z.D. Zhou at St. Joseph Medical
Center, Syracuse, N.Y. for his help in the improvement of the man-
uscript. The author also thanks the reviewers for their critical com-
ments.
References
1 Hermann H-P, Pieske B, Schwarzmuller E,
Keul J, Just H, Hasenfuss G: Haemodynamic
effects of intracoronary pyruvate in patients
with congestive heart failure: An open study.
Lancet 1999; 353: 1321–1323.
2 Kristo G, Yoshimura Y, Niu J, Keith BJ, Men-
tzer RM Jr, Bunger R, Lasley RD: The inter-
mediary metabolite pyruvate attenuates stun-
ning and reduces infarct size in in vivo porcine
myocardium. Am J Physiol 2004; 286:H517–
H524.
3 Lopaschuk GD, Rebeyka IM, Allard MF: Met-
abolic modulation: A means to mend a broken
heart. Circulation 2002; 105: 140–142.
4 Ing TS, Zhou XJ, Yu AW, Zhou FQ, Vaziri
ND: Effects of pyruvate-based or lactate-based
peritoneal dialysis solutions on neutrophil in-
tracellular pH. Int J Artif Organs 1997; 20: 255–
260.
5 Zhou FQ: Advantages of pyruvate over lactate
in peritoneal dialysis solutions. Acta Pharma-
col Sin 2001; 22: 385–392.
6 Mongan PD, Karaian J, Van Der Schuur BM,
Via DK, Sharma P: Pyruvate prevents poly-
ADP ribose polymerase (PARP) activation,
oxidative damage, and pyruvate dehydroge-
nase deactivation during hemorrhagic shock in
swine. J Surg Res 2003; 112: 180–188.
7 Schwartz WB, Waters WC 3rd: Lactate versus
bicarbonate: A reconsideration of the therapy
of metabolic acidosis. Am J Med 1962; 32: 831–
834.
8 McLean AG, Davenport A, Cox D, Sweny P:
Effects of lactate-buffered and lactate- free di-
alysate in CAVHD patients with and without
liver dysfunction. Kidney Int 2000; 58: 1765–
1772.
9 Cohen RD: Roles of the liver and kidney in
acid-base regulation and its disorders. Br J An-
aesth 1991; 67: 154–164.
10 Griffi n JL, White LT, Lewandowski ED: Sub-
strate-dependent proton load and recovery of
stunned hearts during pyruvate dehydrogenase
stimulation. Am J Physiol 2000; 279:H361–
H367.
11 Hasenfuss G, Maier LS, Hermann H-P, Luers
C, Hunlich M, Zeitz O, Janssen PM, Pieske B:
Infl uence of pyruvate on contractile perfor-
mance and Ca(2+) cycling in isolated failing
human myocardium. Circulation 2002; 105:
194–199.
12 Samaja M, Allibardi S, Milano G, Neri G,
Grassi B, Gladden LB, Hogan MC: Differen-
tial depression of myocardial function and me-
tabolism by lactate and H
+
. Am J Physiol 1999;
276:H3–H8.
13 Zubay GL, Parson WW, Vance DE: The tricar-
boxylic acid cycle; in Zubay GL, Parson WW,
Vance DE (eds): Principles of Biochemistry,
Chapt 13. Dubuque, Brown Communications
Inc, 1995, pp 282–304.
14 Dodgson SJ, Forster RE 2nd: Inhibition of CA
V decreases glucose synthesis from pyruvate.
Arch Biochem Biophys 1986; 251: 198–204.
15 Filippi L, Bagnoli F, Margollicci M, Zammar-
chi E, Tronchin M, Rubaltelli FF: Pathogenic
mechanism, prophylaxis, and therapy of symp-
tomatic acidosis induced by acetazolamide. J
Investig Med 2002; 50: 125–132.
16 Stacpoole PW, Nagaraja NV, Hutson AD: Ef-
cacy of dichloroacetate as a lactate- lowering
drug. J Clin Pharmacol 2003; 43: 683–691.
17 Healey MA, Davis RE, Liu FC, Loomis WH,
Hoyt DB: Lactated Ringer’s is superior to nor-
mal saline in a model of massive hemorrhage
and resuscitation. J Trauma 1998; 45: 894–
899.
18 Yu AW, Zhou XJ, Zhou FQ, Nawab ZM, Gan-
dhi VC, Ing TS, Vaziri ND: Neutrophilic intra-
cellular acidosis induced by conventional, lac-
tate-containing peritoneal dialysis solutions.
Int J Artif Organs 1992; 15: 661–665.
19 Kraut JA, Kurtz I: Use of base in the treatment
of severe acidemic states. Am J Kidney Dis
2001; 38: 703–727.
20 Stacpoole PW, Wright EC, Baumgartner TG,
et al: A controlled clinical trial of dichloroac-
etate for treatment of lactic acidosis in adults.
N Engl J Med 1992; 327: 1564–1569.
21 Kline JA, Thornton LR, Lopaschuk GD, Bar-
bee RW, Watts JA: Lactate improves cardiac
effi ciency after hemorrhagic shock. Shock
2000; 14: 215–221.
22 Kline JA, Maiorano PC, Schroeder JD, Grat-
tan RM, Vary TC, Watts JA: Activation of py-
ruvate dehydrogenase improves heart function
and metabolism after hemorrhagic shock. J
Mol Cell Cardiol 1997; 29: 2465–2474.
23 Swafford AN Jr, Bidros D, Truxillo TM, Gi-
aimo ME, Miller HI, McDonough KH: Etha-
nol intoxication and lactated Ringer’s resusci-
tation prolong hemorrhage-induced lactic
acidosis. Shock 2003; 20: 237–244.
24 Lloyd SG, Wang P, Zeng H, Chatham JC: Im-
pact of low-fl ow ischemia on substrate oxida-
tion and glycolysis in the isolated perfused rat
heart. Am J Physiol 2004; 287:H351–H362.
25 Bailey SM, Reinke LA: Effect of low fl ow isch-
emia-reperfusion injury on liver function. Life
Sci 2000; 66: 1033–1044.
26 Bellomo R: Bench-to-bedside review: Lactate
and the kidney. Crit Care 2002; 6: 322–326.
27 Cano N: Bench-to-bedside review: Glucose
production from the kidney. Crit Care 2002; 6:
317–321.
28 Kondou I, Nakada J, Hishinuma H, Masuda
F, Machida T, Endou H: Alterations of gluco-
neogenesis by ischemic renal injury in rats.
Ren Fail 1992; 14: 479–483.
29 Hwang JJ, Curthoys NP: Effect of acute alter-
nations in acid-base balance on rat renal gluta-
minase and phosphoenolpyruvate carboxyki-
nase gene expression. J Biol Chem 1991; 266:
9392–9396.
30 Chiolero RL, Revelly JP, Leverve X, Gersbach
P, Cayeux MC, Berger MM, Tappy L: Effects
of cardiogenic shock on lactate and glucose me-
tabolism after heart surgery. Crit Care Med
2000; 28: 3784–3791.
31 Koustova E, Rhee P, Hancock T, Chen H, Ino-
cencio R, Jaskille A, Hanes W, Valeri CR,
Alam HB: Ketone and pyruvate Ringer’s solu-
tions decrease pulmonary apoptosis in a rat
model of severe hemorrhagic shock and resus-
citation. Surgery 2003; 134: 267–274.
32 Liedtke AJ, Nellis SH, Neely JR, Hughes HC:
Effects of treatment with pyruvate and tro-
methamine in experimental myocardial isch-
emia. Circ Res 1976; 39: 378–387.
Downloaded by:
68.75.176.9 - 6/26/2014 8:37:00 AM
Pyruvate and Metabolic Acidosis
Correction
Am J Nephrol 2005;25:55–63 63
33 Yanos J, Patti MJ, Stanko RT: Hemodynamic
effects of intravenous pyruvate in the intact,
anesthetized dog. Crit Care Med 1994; 22: 844–
850.
34 Mongan PD, Fontana JL, Chen R, Bunger R:
Intravenous pyruvate prolongs survival during
hemorrhagic shock in swine. Am J Physiol
1999; 277:H2253–H2263.
35 Zhou FQ: Pyruvate improvement of cellular
energetics, not simply generating ATP during
anoxia. Transplant 2002; 73: 1851.
36 Wu YT, Wu ZL, Jiang XF, Li S, Zhou FQ: Py-
ruvate preserves neutrophilic superoxide pro-
duction in acidic, high glucose-enriched peri-
toneal dialysis solutions. Artif Organs 2003; 27:
276–281.
37 Regitz V, Azumi T, Stephan H, Naujocks S,
Schaper W: Biochemical mechanism of infarct
size reduction by pyruvate. Cardiovasc Res
1981; 15: 652–658.
38 Satoh H, Sugiyama S, Nomura N, Terada H,
Hayashi H: Importance of glycolytically de-
rived ATP for Na
+
loading via Na
+
/H
+
ex-
change during metabolic inhibition in guinea
pig ventricular myocytes. Clin Sci 2001; 101:
243–251.
39 Karwatowska-Prokopczuk E, Nordberg JA, Li
HL, Engler RL, Gottlieb RA: Effect of vacuolar
proton ATPase on pH
i
, Ca
2+
, and apoptosis in
neonatal cardiomyocytes during metabolic in-
hibition/recovery. Circ Res 1998; 82: 1139–
1144.
40 Sileri P, Schena S, Morini S, Rastellini C,
Pham S, Benedetti E, Cicalese L: Pyruvate in-
hibits hepatic ischemia-reperfusion injury in
rats. Transplantation 2001; 72: 27–30.
41 Mongan PD, Capacchione J, Fontana JL, West
S, Bunger R: Pyruvate improves cerebral me-
tabolism during hemorrhagic shock. Am J
Physiol 2001; 281:H854–H864.
42 Mongan PD, Capacchione J, West S, Karaian
J, Dubois D, Keneally R, Sharma P: Pyruvate
improves redox status and decreases indicators
of hepatic apoptosis during hemorrhagic shock
in swine. Am J Physiol 2002; 283:H1634–
H1644.
43 Saiki Y, Lopaschuk GD, Dodge K, Yamaya K,
Morgan C, Rebeyka IM: Pyruvate augments
mechanical function via activation of the py-
ruvate dehydrogenase complex in reperfused
ischemic immature rabbit hearts. J Surg Res
1998; 79: 164–169.
44 Panchal AR, Comte B, Huang H, Kerwin T,
Darvish A, des Rosiers C, Brunengraber H,
Stanley WC: Partitioning of pyruvate between
oxidation and anaplerosis in swine hearts. Am
J Physiol 2000; 279:H2390–H2398.
45 Oliver FJ, Salto R, Sola MM, Vargas AM: Mi-
tochondrial pyruvate metabolism in liver and
kidney during acidosis. Cell Biochem Funct
1994; 12: 229–235.
46 Burns SP, Murphy HC, Iles RA, Cohen RD:
Lactate supply as a determinant of the distribu-
tion of intracellular pH within the hepatic lob-
ule. Biochem J 2001; 358: 569–571.
47 Jackson VN, Halestrap AP: The kinetics, sub-
strate, and inhibitor specifi city of the mono-
carboxylate (lactate) transporter of rat liver
cells determined using the fl uorescent intra-
cellular pH indicator, 2,7-bis(carboxyethyl)-
5(6)- carboxyfl uorescein. J Biol Chem 1996;
271: 861–868.
48 Wang X, Levi AJ, Halestrap AP: Kinetics of
the sarcolemmal lactate carrier in single heart
cells using BCECF to measure pHi. Am J
Physiol 1994; 267:H1759–H1769.
49 Martin BJ, Valdivia HH, Bunger R, Lasley
RD, Mentzer RM Jr: Pyruvate augments cal-
cium transients and cell shortening in rat ven-
tricular myocytes. Am J Physiol 1998; 274:H8–
H17.
50 Wathen RL, Ward RA, Harding GB, Meyer
LC: Acid-base and metabolic responses to an-
ion infusion in the anesthetized dog. Kidney
Int 1982; 21: 592–599.
51 Berry MN, Phillips JW, Gregory RB, Grivell
AR, Wallace PG: Operation and energy depen-
dence of the reducing-equivalent shuttles dur-
ing lactate metabolism by isolated hepatocytes.
Biochim Biophys Acta 1992; 1136: 223–230.
52 Lee YJ, Kang IJ, Bunger R, Kang YH: En-
hanced survival effect of pyruvate correlates
MAPK and NF-
B activation in hydrogen per-
oxide-treated human endothelial cells. J Appl
Physiol 2004; 96: 793–801.
53 Kubasiak LA, Hernandez OM, Bishopric NH,
Webster KA: Hypoxia and acidosis activate
cardiac myocyte death through the Bcl-2 fam-
ily protein BNIP3. Proc Natl Acad Sci USA
2002; 99: 12825–12830.
54 Olivencia-Yurvati AH, Blair JL, Baig M, Mal-
let RT: Pyruvate-enhanced cardioprotection
during surgery with cardiopulmonary bypass.
J Cardiothorac Vasc Anesth 2003; 17: 715–
720.
55 Mizock BA, Falk JL: Lactic acidosis in critical
illness. Crit Care Med 1992; 20: 80–93.
56 Levraut J, Giunti C, Ciebiera J-P, de Sousa G,
Ramhani R, Payan P, Grimaud D: Initial ef-
fect of sodium bicarbonate on intracellular pH
depends on the extracellular non-bicarbonate
buffering capacity. Crit Care Med 2001; 29:
1033–1039.
57 National Conference on Cardiopulmonary Re-
suscitation: Standards and guidelines for car-
diopulmonary resuscitation and emergency
cardiac care. Part III: Adult advanced cardiac
life support. JAMA 1986; 255: 2933–2954.
58 Chiasson J-L, Aris-Jilwan N, Belanger R, Ber-
trand S, Beauregard H, Ekoe J-M, Fournier H,
Havrankova, J: Diagnosis and treatment of
diabetic ketoacidosis and the hyperglycemic
hyperosmolar state. CMAJ 2003; 168: 859–
866.
59 Van Erven PMM, Gabreels FJM, Wevers RA,
Doesburg WH, Ruitenbeek W, Renier WO,
Lamers KJB: Intravenous pyruvate loading
test in Leigh syndrome. J Neurol Sci 1987; 77:
217–227.
60 Kovacic V, Roguljic L, Kovacic V: Metabolic
acidosis of chronically hemodialyzed patients.
Am J Nephrol 2003; 23: 158–164.
61 Epperlein MM, Nourooz-Zadeh J, Jayasena
SD, Hothersall JS, Noronha-Dutra A, Neild
GH: Nature and biological signifi cance of free
radicals generated during bicarbonate hemodi-
alysis. J Am Soc Nephrol 1998; 9: 457–463.
62 Fink MP: Ringer’s ethyl pyruvate solution: A
novel resuscitation fl uid. Minerva Anestesiol
2001; 67: 190–192.
Downloaded by:
68.75.176.9 - 6/26/2014 8:37:00 AM
... Notably, even the buffer capacity is equal (30 bicarbonate mmol/L) in both Pyr-ORS and WHO-ORS, neither bicarbonate-, nor citratebased WHO-ORS corrected hypoxic lactic acidosis (one of lethal complications in critical care patients), but did Pyr-ORS, robustly prolonging survival in animal studies [2,3,5]. That is because only can pyruvate consume hydrogen ([H + ]) in three metabolic pathways as a prospective alkalizer: 1) the pyruvate reduction coupled with the nicotinamide adenine dinucleotide reduced form (NADH) oxidative reaction by stereo-specific lactate dehydrogenase (LDH) to raise the oxidized form NAD + /NADH ratio throughout the body, 2) the reactivation of pyruvate dehydrogenase (PDH) activity to promote oxidative phosphorylation in the tricarboxylic acid (TCA) cycle of almost all organs and tissues and 3) pyruvatebased cytosolic gluconeogenesis mainly in liver and kidney [2,5,7], reversing the Warburg effect in glucometabolic disorders [8,9]. The post-pyruvate metabolic profile was previously illustrated [7][8][9]. ...
... That is because only can pyruvate consume hydrogen ([H + ]) in three metabolic pathways as a prospective alkalizer: 1) the pyruvate reduction coupled with the nicotinamide adenine dinucleotide reduced form (NADH) oxidative reaction by stereo-specific lactate dehydrogenase (LDH) to raise the oxidized form NAD + /NADH ratio throughout the body, 2) the reactivation of pyruvate dehydrogenase (PDH) activity to promote oxidative phosphorylation in the tricarboxylic acid (TCA) cycle of almost all organs and tissues and 3) pyruvatebased cytosolic gluconeogenesis mainly in liver and kidney [2,5,7], reversing the Warburg effect in glucometabolic disorders [8,9]. The post-pyruvate metabolic profile was previously illustrated [7][8][9]. Oral pyruvate in Pyr-ORS multifaceted protection of organ metabolism and function above was supported by many previous studies with intravenous (IV) pyruvate and further evidenced by pyruvate peritoneal resuscitation in shock resuscitation of rats [8,10,11]. It is worthy of comment that pyruvate of sodium salt in animal shock resuscitation displays superiorities to anions in current medical fluids and commercial beverages: chloride, bicarbonate, lactate, acetate, citrate, phosphate and gluconate in the correction of hypoxic lactic acidosis and improvement of glucose metabolic disorders [5,8,9]. ...
... Interestingly, recent findings showed that oral lactate favored oxidative metabolism of endurance training in mouse muscle, but a pyruvate counterpart was not compared [58]. The LDH reaction is bidirectional with preferable lactate generation; exogenous pyruvate reduction is more favorable with the LDH (A isoenzyme) than the lactate oxidation with the LDH (B isoenzyme) in cytosol, so that lactate infusion raises less blood pyruvate than blood lactate rises after equimolar pyruvate infusion [7]. Despite both pyruvate and lactate as energy substrates, they are quite opposite in the last step of glycolysis, acid-base balance, redox potential and oxidative stress by the LDH reaction and pyruvate oxidizes with a less oxygen consumption rate, compared to lactate. ...
Article
Pyruvate has been extensively and intensively studied since a half century ago. An abundance of experimental researches, in vitro and in vivo, in both animals and humans demonstrate that pyruvate is an unique anion, which is more beneficial in protection of multiple cell/organ metabolism and function than anions in commercial medical fluids and health beverages. The robust advantages of pyruvate action are mainly enhancement of anoxia/hypoxia tolerance, correction of hypoxic lactic acidosis and improvement of glucometabolic disorders in addition to anti-oxidative stress and anti-inflammation and protection of mitochondria, leading to reversal of the Warburg effect in various pathogenic attacks, including severe hypoxia/ ischemia, hypo/hyperglycemia, trauma/burn and sepsis. Many investigations in animals and humans, in vivo, reveal pyruvate protections with absence of clinical adverse effects. Innovative pyruvate-enriched fluids, both crystalloids and colloids, would be more favorable than current fluids in clinical resuscitation due to therapeutic effects in addition as a volume expander. Pyruvate-enriched oral rehydration salt (Pyr-ORS, equimolar pyruvate replacement of alkalizers in WHO-ORS) also would be more beneficial than WHO-ORS in oral rehydration, peri-operative fluid management and prehospital rescue. Alternatively, oral Pyr-ORS-based beverages may be helpful in plateau tourism, diabetes care and anti-aging. This review cited most important animal experiments and human tests with pyruvate dosages applicated, suggesting the effectiveness and clinical safety and recommending innovative Pyr-ORS-based beverages as both medical care when short of medical supply and functional drinks in endurance exercises. Pyruvate, as a novel nutritional componence, applications in clinical scenarios would be another most important medical advance this century.
... In an effort to obtain therapeutic effects with pyruvate in various diseases [11,13,14,16,26], a continuous infusion model has been developed. Mongan et al. [14] infused sodium pyruvate at a rate of 500 mg/kg/h to attain arterial pyruvate levels of 5 mM and showed improved cerebral metabolism in the swine hemorrhagic shock model. ...
... Sodium pyruvate, when used in vivo, works as a metabolic base buffer that can correct an intracellular acidosis [26]. Our data showed that pyruvate contributed to earlier correction of blood pH after CA in conjunction with elevated lactate, HCO 3 , PaCO 2 , and VCO 2 . ...
Article
Full-text available
Purine nucleotide adenosine triphosphate (ATP) is a source of intracellular energy maintained by mitochondrial oxidative phosphorylation. However, when released from ischemic cells into the extracellular space, they act as death-signaling molecules (eATP). Despite there being potential benefit in using pyruvate to enhance mitochondria by inducing a highly oxidative metabolic state, its association with eATP levels is still poorly understood. Therefore, while we hypothesized that pyruvate could beneficially increase intracellular ATP with the enhancement of mitochondrial function after cardiac arrest (CA), our main focus was whether a proportion of the raised intracellular ATP would detrimentally leak out into the extracellular space. As indicated by the increased levels in systemic oxygen consumption, intravenous administrations of bolus (500 mg/kg) and continuous infusion (1000 mg/kg/h) of pyruvate successfully increased oxygen metabolism in post 10-min CA rats. Plasma ATP levels increased significantly from 67 ± 11 nM before CA to 227 ± 103 nM 2 h after the resuscitation; however, pyruvate administration did not affect post-CA ATP levels. Notably, pyruvate improved post-CA cardiac contraction and acidemia (low pH). We also found that pyruvate increased systemic CO2 production post-CA. These data support that pyruvate has therapeutic potential for improving CA outcomes by enhancing oxygen and energy metabolism in the brain and heart and attenuating intracellular hydrogen ion disorders, but does not exacerbate the death-signaling of eATP in the blood.
... Glutamine (or less probably glutamate, see discussion here [23]) is taken up by mitochondria and metabolized ( Figure 1, steps 2-5). TCA and biochemical pathways feeding gluconeogenesis generate a series of reactions that consume and generate protons and CO 2 ( Figure 1, steps 2-7) [24]. Each biochemical reaction from glutamine to glutamate and glutamate to alpha-ketoglutarate yields one molecule of ammonium (Figure 1, steps 3 and 4) while the decarboxylation processes in parallel with oxidative phosphorylation (Figure 1, steps 4-7) [25] and the conversion of 1,3 biphosphoglycerate into glyceraldehyde 3phosphate form bicarbonate (Figure 1, step 8). ...
... Therefore, for each glutamine, two NH 4 + and two HCO 3 are formed. Gluconeogenesis from lactate generates one molecule of bicarbonate [24]. Bicarbonate is released from proximal tubule cells into blood mostly by the electrogenic sodium bicarbonate cotransporter, NBCe1 (SLC4A4, probably transporting carbonate [27]) localized at the basolateral side of these cells. ...
Article
Chronic kidney disease (CKD) is characterized by progressive reduction in kidney function and treatments aiming at stabilizing or slowing its progression may avoid or delay the necessity of kidney replacement therapy and the increased mortality associated with reduced kidney function. Metabolic acidosis, and less severe stages of the acid stress continuum, are common consequences of CKD and some interventional studies support that its correction slows the progression to end-stage kidney disease. This correction can be achieved with mineral alkali in the form of bicarbonate or citrate salts, ingestion of diets with fewer acid-producing food components or more base-producing food components, or a pharmacological approach. In this mini-review article, we summarize the potential mechanisms involved in the beneficial effects of alkali therapy. We also discuss the perspectives in the field and challenges that must be overcome to advance our understanding of such mechanisms.
... Recently, it has been suggested that activation of signal transducer and activator of transcription 3 (STAT3), malonate supplementation and monoamine oxidase inhibitors have cardioprotective effects preserving mitochondrial respiration [37,42,57]. A possible new approach might be through counteracting the increased intracellular acidification that we observed in this study [75,82]. ...
Article
Full-text available
Statins are effective drugs in reducing cardiovascular morbidity and mortality by inhibiting cholesterol synthesis. These effects are primarily beneficial for the patient’s vascular system. A significant number of statin users suffer from muscle complaints probably due to mitochondrial dysfunction, a mechanism that has recently been elucidated. This has raised our interest in exploring the effects of statins on cardiac muscle cells in an era where the elderly and patients with poorer functioning hearts and less metabolic spare capacity start dominating our patient population. Here, we investigated the effects of statins on human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-derived CMs). hiPSC-derived CMs were exposed to simvastatin, atorvastatin, rosuvastatin, and cerivastatin at increasing concentrations. Metabolic assays and fluorescent microscopy were employed to evaluate cellular viability, metabolic capacity, respiration, intracellular acidity, and mitochondrial membrane potential and morphology. Over a concentration range of 0.3–100 µM, simvastatin lactone and atorvastatin acid showed a significant reduction in cellular viability by 42–64%. Simvastatin lactone was the most potent inhibitor of basal and maximal respiration by 56% and 73%, respectively, whereas simvastatin acid and cerivastatin acid only reduced maximal respiration by 50% and 42%, respectively. Simvastatin acid and lactone and atorvastatin acid significantly decreased mitochondrial membrane potential by 20%, 6% and 3%, respectively. The more hydrophilic atorvastatin acid did not seem to affect cardiomyocyte metabolism. This calls for further research on the translatability to the clinical setting, in which a more conscientious approach to statin prescribing might be considered, especially regarding the current shift in population toward older patients with poor cardiac function.
... Both enrichments were previously identified for the differentially expressed genes in the liver between the treatment groups (36). Specifically, pyruvate was reported as a correction buffer to intracellular acidosis (63). Furthermore, an enrichment of thyroid hormone synthesis pathway was identified for the genes associated with the tissue-embedded microbes. ...
Article
Full-text available
Introduction In the dairy industry, calves are typically fed diets rich in highly fermentable carbohydrates and low in fibrous feeds to maximize ruminal papillae and tissue development. Calves on such diets are vulnerable at developing ruminal acidosis. Prevalent in cattle, liver abscess (LA) is considered a sequela to ruminal acidosis. LAs can cause significant liver function condemnation and decreased growth and production. Currently, we know little about the liver microbiome in calves with feed-induced acidosis. Methods Using our established model of ruminal acidosis, where young calves were fed an acidosis-inducing (AC) or -blunting (control) diet starting at birth until 17-week of age, we investigated microbial community changes in the liver resultant from ruminal acidosis. Eight calves were randomly assigned to each diet, with four animals per treatment. Rumen epithelium and liver tissues were collected at 17 weeks of age right after euthanasia. Total RNAs were extracted and followed by whole transcriptome sequencing. Microbial RNA reads were enriched bioinformatically and used for microbial taxonomy classification using Kraken2. Results AC Calves showed significantly less weight gain over the course of the experiment, in addition to significantly lower ruminal pH, and rumen degradation comparison to the control group (p < 0.05). In the liver, a total of 29 genera showed a significant (p < 0.05) abundance change (> 2-fold) between the treatments at 17-week of age. Among these, Fibrobacter, Treponema, Lactobacillus, and Olsenella have been reported in abscessed liver in cattle. Concurrent abundance changes in 9 of the genera were observed in both the liver and rumen tissues collected at 17-week of age, indicating potential crosstalk between the liver and rumen epithelial microbial communities. Significant association was identified between host liver gene and its embedded microbial taxa. Aside from identifying previously reported microbial taxa in cattle abscessed liver, new repertoire of actively transcribed microbial taxa was identified in this study. Discussion By employing metatranscriptome sequencing, our study painted a picture of liver microbiome in young calves with or without feed induced acidosis. Our study suggested that liver microbiome may have a critical impact on host liver physiology. Novel findings of this study emphasize the need for further in-depth analysis to uncover the functional roles of liver resident microbiome in liver metabolic acidosis resultant from feed-related ruminal acidosis.
... From the discussion above, it is clear that optimal bTBI management often requires optimal HS management, especially during the pre-hospital phase. Recent studies have suggested that pyruvate, a natural product of the reaction in the last step of the glycolytic pathway, can improve the outcome in animal models with TBI or HS through mitochondrial mechanisms (6)(7)(8)(9). In a pyruvate dose response study targeting the vital signs following hemorrhagic shock, our lab previously showed that sodium pyruvate 2.0 M was most effective in multiorgan failure and survival rate in HS (10). ...
Article
Full-text available
Background Blast injuries from improvised explosive devices (IEDs) are known to cause blast traumatic brain injuries (bTBIs), hemorrhagic shock (HS), organ damage, mitochondrial dysfunction, and subsequent free radical production. A pre-citric acid cycle reagent, pyruvate, is suggested to improve mitochondrial ATP production through the activation of the mitochondrial gatekeeper enzyme “pyruvate dehydrogenase complex (PDH).” Our study aimed to investigate the role of physiologic, metabolic, and mitochondrial effects of hypertonic sodium pyruvate resuscitation in rats with a combined blast and HS injury. Methods A pre-clinical rat model of combined injury with repetitive 20 PSI blast exposure accompanied with HS and fluid resuscitation (sodium pyruvate as metabolic adjuvant or hypertonic saline as control), followed by transfusion of shed blood was used in this study. Control sham animals (instrumental and time-matched) received anesthesia and cannulation, but neither received any injury nor treatment. The mean arterial pressure and heart rate were recorded throughout the experiment by a computerized program. Blood collected at T0 (baseline), T60 (after HS), and T180 (end) was analyzed for blood chemistry and mitochondrial PDH enzyme activity. Results Sodium pyruvate resuscitation significantly improved the mean arterial pressure (MAP), heart rate (HR), pulse pressure (PP), hemodynamic stability (Shock index), and autonomic response (Kerdo index) after the HS and/or blast injury. Compared with the baseline values, plasma lactate and lactate/pyruvate ratios were significantly increased. In contrast, base excess BE/(HCO3-) was low and the pH was also acidotic <7.3, indicating the sign of metabolic acidosis after blast and HS in all animal groups. Sodium pyruvate infusion significantly corrected these parameters at the end of the experiment. The PDH activity also improved after the sodium pyruvate infusion. Conclusion In our rat model of a combined blast and HS injury, hypertonic sodium pyruvate resuscitation was significantly effective in hemodynamic stabilization by correcting the acid–base status and mitochondrial mechanisms via its pyruvate dehydrogenase enzyme.
... Pyruvate is a potent alkalizer via the rapid metabolic consumption of hydrogen ions (proton, [H + ]) through the LDH reduction reaction, which is a systemic alkalizing enzymatic reaction, coupled with an increase in the NAD + /NADH ratio, and the gluconeogenesis pathway in the cytosol in addition to oxidative phosphorylation in the mitochondria (Figure 1). Although it is a weaker acidic anion of sodium salt with a low buffer capacity of pKa 2.49, pyruvate favors a rise in blood plasma pH accordingly (17,41). Pyruvate has the potential to effectively correct hypoxic lactic acidosis in critically ill patients, as repeatedly demonstrated with IV or oral pyruvate in small or large animal studies, which resulted in approximately doubled survival (42)(43)(44)(45). ...
Article
Full-text available
There have been ongoing debates about resuscitation fluids because each of the current fluids has its own disadvantages. The debates essentially reflect an embarrassing clinical status quo that all fluids are not quite ideal in most clinical settings. Therefore, a novel fluid that overcomes the limitations of most fluids is necessary for most patients, particularly diabetic and older patients. Pyruvate is a natural potent antioxidant/nitrosative and anti-inflammatory agent. Exogenous pyruvate as an alkalizer can increase cellular hypoxia and anoxia tolerance with the preservation of classic glycolytic pathways and the reactivation of pyruvate dehydrogenase activity to promote oxidative metabolism and reverse the Warburg effect, robustly preventing and treating hypoxic lactic acidosis, which is one of the fatal complications in critically ill patients. In animal studies and clinical reports, pyruvate has been shown to play a protective role in multi-organ functions, especially the heart, brain, kidney, and intestine, demonstrating a great potential to improve patient survival. Pyruvate-enriched fluids including crystalloids and colloids and oral rehydration solution (ORS) may be ideal due to the unique beneficial properties of pyruvate relative to anions in contemporary existing fluids, such as acetate, bicarbonate, chloride, citrate, lactate, and even malate. Preclinical studies have demonstrated that pyruvate-enriched saline is superior to 0.9% sodium chloride. Moreover, pyruvate-enriched Ringer’s solution is advantageous over lactated Ringer’s solution. Furthermore, pyruvate as a carrier in colloids, such as hydroxyethyl starch 130/0.4, is more beneficial than its commercial counterparts. Similarly, pyruvate-enriched ORS is more favorable than WHO-ORS in organ protection and shock resuscitation. It is critical that attention first be paid to improving abnormal saline with pyruvate for ICU patients. Many clinical trials with a high dose of intravenous or oral pyruvate were conducted over the past half century, and results indicated its effectiveness and safety in humans. The long-term instability of pyruvate aqueous solutions and para-pyruvate cytotoxicity is not a barrier to the pharmaceutical manufacturing of pyruvate-enriched fluids for ICU patients. Clinical trials with sodium pyruvate-enriched solutions are urgently warranted.
Article
Full-text available
Pyruvate is a three-carbon ketoacid that occurs naturally in cells. It is produced through enzymatic reactions in the glycolytic pathway and plays a crucial role in energy metabolism. Despite promising early results, later well-controlled studies of physically active people have shown that pyruvate supplementation lasting more than 1 week has no ergogenic effects. However, some data suggest that ingested pyruvate may be preferentially metabolized without accumulation in the bloodstream. Pyruvate exhibits antioxidant activity and can affect the cellular redox state, and exogenous pyruvate can influence metabolism by affecting the acid- base balance of the blood. This brief review focuses on the potential effects of pyruvate as a supplement for active people. The current state of understanding suggests that studies of the effects of pyruvate supplementation should prioritize investigating the timing of pyruvate intake.
Preprint
Full-text available
Purine nucleotide adenosine triphosphate (ATP) is a source of intracellular energy maintained by mitochondrial oxidative phosphorylation. However, when released from ischemic cells into the extracellular space, they act as death-signaling molecules (eATP). Despite there being potential benefit in using pyruvate to enhance mitochondria by inducing a highly oxidative metabolic state, its association with eATP levels is still poorly understood. Therefore, while we hypothesized that pyruvate could beneficially increase intracellular ATP with the enhancement of mitochondrial function after cardiac arrest (CA), our main focus was whether a proportion of the raised intracellular ATP would detrimentally leak out into the extracellular space. Indicated by the increased levels in systemic oxygen consumption and brain ATP levels, intravenous administrations of bolus (500 mg/kg) and continuous infusion (1000 mg/kg/hr) of pyruvate successfully increased oxygen and energy metabolism in post 10-min CA rats. The plasma ATP levels increased significantly from 67 ± 11 nM before CA to 227 ± 100 nM 2 hours after the resuscitation, while the pyruvate injection did not affect post-CA ATP levels. Notably, the pyruvate injection improved post-CA cardiac contraction and acidemia (low pH). We also found that pyruvate increased systemic CO 2 production post-CA. These data support that pyruvate has therapeutic potential for improving CA outcomes by enhancing oxygen and energy metabolism in the brain and heart, and attenuating intracellular hydrogen iron disorders, but does not exacerbate the death-signaling of eATP in the blood.
Article
Full-text available
The liver has many significant functions, such as detoxification, the urea cycle, gluconeogenesis, and protein synthesis. Systemic diseases, hypoxia, infections, drugs, and toxins can easily affect the liver, which is extremely sensitive to injury. Systemic infection of severe acute respiratory syndrome coronavirus 2 can cause liver damage. The primary regulator of intracellular pH in the liver is the Na+/H+ exchanger (NHE). Physiologically, NHE protects hepatocytes from apoptosis by making the intracellular pH alkaline. Severe acute respiratory syndrome coronavirus 2 increases local angiotensin II levels by binding to angiotensin-converting enzyme 2. In severe cases of coronavirus disease 2019, high angi-otensin II levels may cause NHE overstimulation and lipid accumulation in the liver. NHE overstimulation can lead to hepatocyte death. NHE overstimulation may trigger a cytokine storm by increasing proinflammatory cytokines in the liver. Since the release of proinflammatory cytokines such as interleukin-6 increases with NHE activation, the virus may indirectly cause an increase in fibrinogen and D-dimer levels. NHE overstimulation may cause thrombotic events and systemic damage by increasing fibrinogen levels and cytokine release. Also, NHE overstimulation causes an increase in the urea cycle while inhibiting vitamin D synthesis and gluconeogenesis in the liver. Increasing NHE3 activity leads to Na+ loading, which impairs the containment and fluidity of bile acid. NHE overstimulation can change the gut microbiota composition by disrupting the structure and fluidity of bile acid, thus triggering systemic damage. Unlike other tissues, tumor necrosis factor-alpha and angiotensin II decrease NHE3 activity in the intestine. Thus, increased luminal Na+ leads to diarrhea and cytokine release. Severe acute respiratory syndrome coronavirus 2-induced local and systemic damage can be improved by preventing virus-induced NHE overstimulation in the liver.
Article
Full-text available
We recently reported that pyruvate inhibited translocation and activation of p53 caused by DNA damage due to oxidant injury (Lee YJ, Kang IJ, Bünger R, and Kang YH. Microvasc Res 66: 91-101, 2003); this was associated with increased expression of apoptosis-related bcl-2 and decreased expression of bax gene. This study attempted to delineate possible regulatory sites and mechanisms of antiapoptotic pyruvate, focusing on reactive oxygen species-mediated signaling in a human umbilical vein endothelial cell model. We compared the effects of the cytosolic reductant l-lactate and malate-aspartate shuttle blocker aminooxyacetate, both of which increase cytosolic NADH, on the downstream signaling pathway. Hydrogen peroxide (0.5 mM H2O2) depleted intracellular total glutathione that was prevented by pyruvate but not by l-lactate or aminooxyacetate. Activation of caspase-3 and the cleavage of procaspase-6 and procaspase-7 were strongly inhibited by pyruvate but markedly enhanced by l-lactate and aminooxyacetate, implicating redox-related antiapoptotic mechanisms of pyruvate. Western blot analysis and immunochemical data revealed that H2O2-induced transactivation of nuclear factor-kappaB (NF-kappaB) was also inhibited by pyruvate but not by l-lactate or aminooxyacetate. In addition, H2O2 downregulated extracellular signal-regulated kinase (ERK1/2) and phosphorylated p38 mitogen-activated protein kinase (MAPK), effects that were fully reversed by pyruvate within 2 h. Collectively, these findings indicate that pyruvate can protect cellular glutathione, thus enhancing cellular antioxidant potential, and that enhanced antioxidant potential can desensitize NF-kappaB transactivation due to reactive oxygen species, suggesting possible metabolic redox relations to NF-kappaB. Furthermore, pyruvate blocked the p38 MAPK pathway and activated the ERK pathway in an apparently redox-sensitive manner, which may regulate expression of genes believed to prevent apoptosis and promote cell survival. Thus pyruvate may have therapeutic potential for reducing endothelial dysfunction and improving survival during oxidative stress.
Article
Full-text available
This study investigates evidence of oxidative stress during bicarbonate hemodialysis by measuring total glutathione and lipid peroxidation products in plasma, and characterizes the free radicals produced by neutrophils from healthy volunteers when incubated in vitro with increasing concentrations of bicarbonate. Blood samples were taken from nine hemodialysis patients before and after two hemodialysis sessions. Plasma hydroperoxides and total glutathione were measured. A significant increase was found in total glutathione (1.04 +/- 0.4 versus 2.11 +/- 0.9 microM, P < 0.001) and hydroperoxides by ferrous oxidation in xylenol orange version 2 method (4.6 +/- 0.53 versus 6.4 +/- 0.63 microM, P < 0.001) after hemodialysis, which indicated increased oxidative injury during hemodialysis. Normal neutrophils, activated by contact adhesion, produced a dose-dependent increase in free radical production (measured by luminol-enhanced chemiluminescence) when incubated with increasing concentrations of bicarbonate (up to 35 mM). Bicarbonate had the same effect on the chemiluminescence of a cell-free hypoxanthine/acetaldehyde system generating superoxide, but not on a glucose oxidase/myeloperoxidase system generating hydrogen peroxide and hypochlorous acid. These findings are consistent with (1) the hypothesis that superoxide generated during hemodialysis reacts with bicarbonate to form the toxic carbonate and formate radicals and (2) our previous observation that some patients undergoing bicarbonate (but not lactate) dialysis have increased plasma concentrations of formate after hemodialysis. It is suggested that the increased plasma total glutathione and hydroperoxide concentrations are a result of lipid peroxidation by these species. These reactive radicals can initiate lipid peroxidation and contribute to the cardiovascular complications of hemodialysis patients.
Article
Full-text available
This article reviews the current body of knowledge regarding lactic acidosis in critically ill patients. The classification of disordered lactate metabolism and its pathogenesis are examined. The utility of lactate as a metabolic monitor of shock is examined and current therapeutic strategies in the treatment of patients suffering from lactic acidosis are extensively reviewed. The paper is designed to integrate basic concepts with a current approach to lactate in critical illness that the clinician can use at the bedside. Comprehensive review of the available, basic science, medical, surgical, and critical care literature. The severity of lactic acidosis in critically ill patients correlates with overall oxygen debt and survival. Lactate determinations may be useful as an ongoing monitor of perfusion as resuscitation proceeds. Therapy of critically ill patients with lactic acidosis is designed to maximize oxygen delivery in order to reduce tissue hypoxia by increasing cardiac index, while maintaining hemoglobin concentration. Buffering agents have not been shown to materially affect outcome from lactic acidosis caused by shock. The benefits of other specific therapies designed to reduce the severity of lactic acidosis remain unproven.
Article
Background— Application of pyruvate was shown to improve contractile function in isolated animal myocardium and hemodynamics in patients with congestive heart failure. We assessed the influence of pyruvate on systolic and diastolic myocardial function and its subcellular mode of action in isolated myocardium from end-stage failing human hearts. Methods and Results— In muscle strip preparations, concentration-dependent effects of pyruvate on developed and diastolic force (n=6), aequorin light emission reflecting intracellular Ca²⁺ transients (n=6), and rapid cooling contractures reflecting sarcoplasmic reticulum (SR) Ca²⁺ content (n=11) were measured. Pyruvate resulted in a concentration-dependent increase in developed force and a decrease in diastolic force, with a maximum effect of 155% and 21%, respectively, at 20 mmol/L pyruvate (P<0.05). This was associated with a dose-dependent prolongation of time to peak tension and relaxation time. Pyruvate increased rapid cooling contractures by 51% and aequorin light signals by 85% (at 15 and 20 mmol/L; P<0.05). This indicates increased SR Ca²⁺ content and increased intracellular Ca²⁺ transients. The inotropic effect of pyruvate was still present after elimination of SR Ca²⁺ storage function with 10 μmol/L cyclopiazonic acid and 1 μmol/L ryanodine (n=8). Pyruvate significantly increased intracellular pH from 7.31±0.03 to 7.40±0.04 by BCECF fluorescence (n=6). Conclusions— The present findings indicate that pyruvate improves contractile performance of failing human myocardium by increasing intracellular Ca²⁺ transients as well as myofilament Ca²⁺ sensitivity. The former seem to result from increased SR Ca²⁺ accumulation and release, the latter from increased intracellular pH.
Article
Failure of glycolysis to increase sufficiently to supply optimal levels of energy production in ischemic heart muscle is due in part to the cummulative restrainst of acidosis on rate-limiting enzymes, particularly glyceraldehyde-3-phosphate dehydrogenase. In an effort to modify this inhibition and salvage jeopardized myocardium, treatment with excess levels of pyruvate and tromethamine (Tris), designed to buffer intracellular hydrogen ion accumulations and improve the oxidation-reduction ratio, NAD+/NADH, was tested in 59 swine hearts in two separate preparations of global and regional ischemia. Global ischemia, per se, caused hemodynamic deterioration and shortened survival time (44.3 +/- 3.1 minutes). Myocardial oxygen consumption, fatty acid oxidation, and glucose uptake were all significantly (P less than 0.001) reduced as were estimates of glycolysis and tissue stores of creatine phosphate and ATP (P less than 0.01). Although treatment with Tris alone was inconclusive, administrations of pyruvate (40-50 mM) buffered with Tris (added directly into the coronary perfusate) effected an improvement in mechanical function and a significant prolongation in survival time (56.9 +/- 2.6 minutes. P less than 0.01). Glycogenolysis was enhanced and levels of key glycolytic intermediates were reduced, suggesting an acceleration of glycolytic flux. Excess levels of pyruvate (1.52 +/- 0.48 mumol/ml of coronary perfusate) provided added substrate for oxidation and led to a greater than 5-fold incrase in rates of pyruvate decarboxylation as compared to untreated ischemic hearts...
Article
Mortality is very high in lactic acidosis, and there is no satisfactory treatment other than treatment of the underlying cause. Uncontrolled studies have suggested that dichloroacetate, which stimulates the oxidation of lactate to acetyl-coenzyme A and carbon dioxide, might reduce morbidity and improve survival among patients with this condition. We conducted a placebo-controlled, randomized trial of intravenous sodium dichloroacetate therapy in 252 patients with lactic acidosis; 126 were assigned to receive dichloroacetate and 126 to receive placebo. The entry criteria included an arterial-blood lactate concentration of > or = 5.0 mmol per liter and either an arterial-blood pH of < or = 7.35 or a base deficit of > or = 6 mmol per liter. The mean (+/- SD) arterial-blood lactate concentrations before treatment were 11.6 +/- 7.0 mmol per liter in the dichloroacetate-treated patients and 10.4 +/- 5.5 mmol per liter in the placebo group, and the mean initial arterial-blood pH values were 7.24 +/- 0.12 and 7.24 +/- 0.13, respectively. Eighty-six percent of the patients required mechanical ventilation, and 74 percent required pressor agents, inotropic drugs, or both because of hypotension. The arterial-blood lactate concentration decreased 20 percent or more in 83 (66 percent) of the 126 patients who received dichloroacetate and 45 (36 percent) of the 126 patients who received placebo (P = 0.001). The arterial-blood pH also increased more in the dichloroacetate-treated patients (P = 0.005). The absolute magnitude of the differences was small, however, and they were not associated with improvement in hemodynamics or survival. Only 12 percent of the dichloroacetate-treated patients and 17 percent of the placebo patients survived to be discharged from the hospital. Dichloroacetate treatment of patients with severe lactic acidosis results in statistically significant but clinically unimportant changes in arterial-blood lactate concentrations and pH and fails to alter either hemodynamics or survival.