ArticlePDF AvailableLiterature Review

The Role of Taurine in Mitochondria Health: More Than Just an Antioxidant

Authors:

Abstract and Figures

Taurine is a naturally occurring sulfur-containing amino acid that is found abundantly in excitatory tissues, such as the heart, brain, retina and skeletal muscles. Taurine was first isolated in the 1800s, but not much was known about this molecule until the 1990s. In 1985, taurine was first approved as the treatment among heart failure patients in Japan. Accumulating studies have shown that taurine supplementation also protects against pathologies associated with mitochondrial defects, such as aging, mitochondrial diseases, metabolic syndrome, cancer, cardiovascular diseases and neurological disorders. In this review, we will provide a general overview on the mitochondria biology and the consequence of mitochondrial defects in pathologies. Then, we will discuss the antioxidant action of taurine, particularly in relation to the maintenance of mitochondria function. We will also describe several reported studies on the current use of taurine supplementation in several mitochondria-associated pathologies in humans.
Content may be subject to copyright.
molecules
Review
The Role of Taurine in Mitochondria Health: More Than Just
an Antioxidant
Chian Ju Jong 1, *, Priyanka Sandal 1and Stephen W. Schaffer 2


Citation: Jong, C.J.; Sandal, P.;
Schaffer, S.W. The Role of Taurine in
Mitochondria Health: More Than Just
an Antioxidant. Molecules 2021,26,
4913. https://doi.org/10.3390/
molecules26164913
Academic Editors: Elena Grasselli,
Chiara Lambruschini, Lisa Moni and
Ilaria Demori
Received: 29 July 2021
Accepted: 10 August 2021
Published: 13 August 2021
Publisher’s Note: MDPI stays neutral
with regard to jurisdictional claims in
published maps and institutional affil-
iations.
Copyright: © 2021 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
1
Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA;
priyanka-sandal@uiowa.edu
2Department of Pharmacology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA;
sschaffe@southalabama.edu
*Correspondence: chianju-jong@uiowa.edu
Abstract:
Taurine is a naturally occurring sulfur-containing amino acid that is found abundantly in
excitatory tissues, such as the heart, brain, retina and skeletal muscles. Taurine was first isolated
in the 1800s, but not much was known about this molecule until the 1990s. In 1985, taurine was
first approved as the treatment among heart failure patients in Japan. Accumulating studies have
shown that taurine supplementation also protects against pathologies associated with mitochondrial
defects, such as aging, mitochondrial diseases, metabolic syndrome, cancer, cardiovascular diseases
and neurological disorders. In this review, we will provide a general overview on the mitochondria
biology and the consequence of mitochondrial defects in pathologies. Then, we will discuss the
antioxidant action of taurine, particularly in relation to the maintenance of mitochondria function. We
will also describe several reported studies on the current use of taurine supplementation in several
mitochondria-associated pathologies in humans.
Keywords: taurine; mitochondria; antioxidant; 5-taurinomethyluridine; oxidative stress; apoptosis
1. Introduction
Mitochondrial dysfunction, along with oxidative stress, is a key hallmark of various
pathologies, such as aging [
1
,
2
], cardiovascular diseases [
3
,
4
], mitochondrial diseases [
5
,
6
],
metabolic syndrome [
7
,
8
], cancer [
9
,
10
] and neurological disorders, such as neurodegen-
erative diseases [
11
,
12
] and neurodevelopmental disorders [
13
,
14
]. Often, antioxidant
therapy, such as coenzyme Q [
15
], mitoQ [
16
,
17
], vitamin E [
18
], gingko biloba extracts [
19
],
ebselen [
20
], creatine [
21
], lipoic acid [
22
], melatonin [
23
,
24
] and L-arginine [
25
,
26
], pro-
vide some protections, potentially by improving the mitochondrial function and reducing
oxidative stress in these diseases. Recently, taurine, a sulfur-containing amino acid, has
been approved in Japan in treating stroke-like episodes in patients with mitochondrial
myopathy, encephalopathy, lactic acidosis and stroke-like episodes (MELAS), which is a
mitochondrial disease [
27
,
28
]. Indeed, the use of taurine dates back to 1985, as taurine
was first used to treat patients with congestive heart failure in Japan [
29
,
30
]. In addition,
taurine supplementation has been shown to improve the exercise capacity of patients with
heart failure [
31
], which is likely due to improvement of the myocardial energy production.
Although taurine was first identified in the 1800s [
32
], the mitochondrial actions of taurine
still remain unclear and underappreciated. This review, therefore, will provide an overview
of the significant role of taurine in the maintenance of mitochondrial function. Clinical
studies using taurine therapy in mitochondria-targeted pathologies will also be discussed.
2. Mitochondria Biology
Mitochondria are cellular organelles that regulate various essential cellular pro-
cesses [
33
37
]. The mitochondria consist of two membranes, an ion impermeable inner
membrane and a permeable outer membrane, which envelopes a soluble matrix containing
Molecules 2021,26, 4913. https://doi.org/10.3390/molecules26164913 https://www.mdpi.com/journal/molecules
Molecules 2021,26, 4913 2 of 21
cristae [
38
]. There are hundreds of mitochondria in one cell and each mitochondrion con-
tains 2–10 copies of mitochondrial DNA (mtDNA) [39]. The mtDNA encodes 13 polypep-
tides, which are components of the electron transport chain, as well as two ribosomal
RNAs (rRNA) and 22 transfer RNAs (tRNA), which regulate the synthesis of mitochondrial
proteins [
40
]. Predominantly known as the powerhouse of the cell, mitochondria provide
cellular energy by generating ATP via oxidative phosphorylation. Reducing equivalents,
such as NADH and FADH
2
, produced via the tricarboxylic acid (TCA) cycle, deliver elec-
trons along the electron transport chain to reduce molecular oxygen to water. The influx of
electrons along the electron transport chain creates a transmembrane proton gradient that
drives ATP synthesis via the ATP synthase (F
o
F
1
complex synthase) [
41
]. A consequence of
electron transport along the electron transport chain is the generation of reactive oxygen
species (ROS), where one electron reduces molecular oxygen to produce a superoxide
anion (O
2
−·
) [
42
]. When catalyzed by the antioxidant superoxide dismutase (SOD), O
2
−·
is dismutated to hydrogen peroxide (H
2
O
2
) and molecular oxygen. H
2
O
2
can be further
partially reduced to a hydroxyl radical (OH), which is a highly reactive species [42].
The mitochondria are the main source of superoxide production, primarily via Com-
plex I and Complex III of the electron transport chain [
43
,
44
]. Under normal conditions, 2%
of electrons are diverted to reduce molecular oxygen to produce a superoxide [
42
]. Physio-
logically, ROS has been shown to regulate various crucial cellular processes, such as cellular
differentiation [
45
,
46
], autophagy [
47
,
48
], metabolic adaptation [
49
,
50
] and immune cell
activation [
50
52
]. Pathologically, ROS has often been shown to cause harm to cells, which
are described as follow. One, ROS is capable of inducing mitochondrial and nuclear DNA
damage [
53
,
54
]. Two, ROS causes irreversible protein oxidation. ROS oxidizes the side
chain of four key amino acids, lysine, arginine, proline and threonine, which adds ketone
or aldehyde groups to proteins and alters the protein structure and function [
55
,
56
]. Three,
ROS oxidizes cellular and organelle membranes, which consist of polyunsaturated fatty
acids [
57
]. Cardiolipin is a unique phospholipid localized in the inner membrane of the
mitochondria as it contains a polar head group that traps protons for oxidative phosphory-
lation [
58
], three glycerol backbones and four acyl chains [
59
]. A phospholipid generally
consists of a polar headgroup, a glycerol backbone and hydrophobic acyl chains [
60
]. There
has been increasing evidence showing cardiolipin being crucial for the functionality of the
mitochondria. Primarily, cardiolipin maintains the structural integrity of the mitochondrial
membranes [
61
,
62
], as well as stability and proper functioning of proteins and enzyme
complexes involved in oxidative phosphorylation [
63
65
]. Oxidation of cardiolipin induces
mitochondrial dysfunction, as has been shown in several
in vitro
studies. These studies
have shown impaired cellular metabolism and a sluggish activity of the electron transport
chain [
64
,
66
,
67
], as well as enhanced cell death, as evidenced by mitochondrial permeability
transport pore opening and cytochrome c release [
68
,
69
]. To counteract excessive ROS
production, the cell contains an antioxidant defense system, which encompasses enzymatic
antioxidants such as mitochondria-localized manganese superoxide dismutase (MnSOD),
cytosolic-localized zinc SOD (ZnSOD) and copper SOD (CuSOD), catalase and glutathione
peroxidase. MnSOD, ZnSOD and CuSOD catalyze the dismutation of O
2
into water and
H
2
O
2
[
42
]. Both catalase and glutathione peroxidase break down H
2
O
2
into water and
oxygen [
42
,
70
]. When ROS is excessively produced, it can overwhelm the antioxidant
defenses and this causes oxidative stress [
71
,
72
]. Often, increased oxidative stress further
exacerbates cellular damage [73,74].
3. Mitochondria in Pathologies
Impairment of mitochondrial function has been commonly reported in pathologies
such as aging, cardiovascular diseases, mitochondrial diseases, metabolic syndrome, cancer
and neurological disorders [
1
7
,
9
11
,
13
,
14
,
75
]. Often, these pathologies are character-
ized by increased ROS production [
1
,
2
], reduced ATP generation [
5
,
7
],
apoptosis [75,76]
,
impaired mitochondrial biogenesis [
4
,
76
], impaired activity of electron transport
chain [3,7,77]
and mitochondrial calcium mishandling [
4
]. Recently, increasing evidence
Molecules 2021,26, 4913 3 of 21
from
in vitro
[
78
84
] and
in vivo
[
85
89
] studies has demonstrated the beneficial effects of
taurine in maintaining mitochondrial functions.
4. Taurine Biology
Taurine or 2-aminoethane-sulfonic acid is a unique amino acid as it has a sulfonyl
group on the C-terminus and an amino group residing on the
β
-carbon (Figure 1a) rather
than
α
-carbon (Figure 1b) [
90
]. Taurine, therefore, is a
β
-sulfonic amino acid
(Figure 1)
.
Taurine was first identified by Tiedemann and Gmelin, who isolated taurine in 1827 from
the bile of the ox, Bos taurus [
32
]. As described in Figure 2, taurine (
5
) is synthesized
in the liver from methionine (
1
) or cysteine (
2
) to produce hypotaurine (
4
) by cysteine
dioxygenase and cysteine sulfonic acid decarboxylase (CSAD). Cysteine dioxygenase
converts methionine or cysteine (
1
2
) to cysteinesulfinate (
3
), while CSAD converts cysteine
sulfinate (
3
) to hypotaurine (
4
). Hypotaurine (
4
) is then readily oxidized to taurine (5),
which may be excreted directly or as a conjugate with bile salts such as taurocholate (
6
) [
90
].
Molecules 2021, 26, x FOR PEER REVIEW 3 of 22
3. Mitochondria in Pathologies
Impairment of mitochondrial function has been commonly reported in pathologies
such as aging, cardiovascular diseases, mitochondrial diseases, metabolic syndrome, can-
cer and neurological disorders [17,911,13,14,75]. Often, these pathologies are character-
ized by increased ROS production [1,2], reduced ATP generation [5,7], apoptosis [75,76],
impaired mitochondrial biogenesis [4,76], impaired activity of electron transport chain
[3,7,77] and mitochondrial calcium mishandling [4]. Recently, increasing evidence from in
vitro [7884] and in vivo [8589] studies has demonstrated the beneficial effects of taurine
in maintaining mitochondrial functions.
4. Taurine Biology
Taurine or 2-aminoethane-sulfonic acid is a unique amino acid as it has a sulfonyl
group on the C-terminus and an amino group residing on the β-carbon (Figure 1a) rather
than α-carbon (Figure 1b) [90]. Taurine, therefore, is a β-sulfonic amino acid (Figure 1).
Taurine was first identified by Tiedemann and Gmelin, who isolated taurine in 1827 from
the bile of the ox, Bos taurus [32]. As described in Figure 2, taurine (5) is synthesized in the
liver from methionine (1) or cysteine (2) to produce hypotaurine (4) by cysteine dioxygen-
ase and cysteine sulfonic acid decarboxylase (CSAD). Cysteine dioxygenase converts me-
thionine or cysteine (12) to cysteinesulfinate (3), while CSAD converts cysteine sulfinate
(3) to hypotaurine (4). Hypotaurine (4) is then readily oxidized to taurine (5), which may
be excreted directly or as a conjugate with bile salts such as taurocholate (6) [90].
(a)
(b)
Figure 1. (a) Taurine or 2-aminoethane-sulfonic acid is a β-sulfonic amino acid as it has a sulfonyl
group rather than a carboxyl group attached to the alpha carbon and an amino group on the beta
carbon; (b) a standard amino acid contains an alpha carbon, to which an both an amino group and
a carboxyl group are attached.
Figure 1.
(
a
) Taurine or 2-aminoethane-sulfonic acid is a
β
-sulfonic amino acid as it has a sulfonyl
group rather than a carboxyl group attached to the alpha carbon and an amino group on the beta
carbon; (
b
) a standard amino acid contains an alpha carbon, to which an both an amino group and a
carboxyl group are attached.
Molecules 2021, 26, x FOR PEER REVIEW 4 of 22
Figure 2. Taurine (5) is synthesized from either methionine (1) or cysteine (2). Cysteine dioxygenase catalyzes the conver-
sion of cysteine (2) to cysteinesulfinate (3), which then is converted to hypotaurine (4) by cysteine sulfinate decarboxylase.
Hypotaurine (4) is readily oxidized to form taurine (5), which can be excreted directly or as a conjugate with bile acids
such as taurocholate (6).
In most mammals including humans, rodents and some primates, taurine is consid-
ered a conditionally essential amino acid, as cysteine dioxygenase and CSAD are present
abundantly. Mammals primarily depend on taurine biosynthesis in vivo [91] and partially
from diet, such as from meat, seafood and human milk [9294]. Newborns and young
infants are unable to synthesize taurine as well as adult humans, and therefore, are de-
pendent on a taurine-supplemented diet [95]. Clinical studies investigating infants sup-
plemented with (3040 μM/dL) or without taurine showed that inadequate taurine sup-
plementation impairs lipid absorption and bile acid secretion and causes hepatic and ret-
inal dysfunction [9699]. Due to the importance of taurine in neonatal development, moth-
ers are strongly encouraged to breastfeed, due to the high concentration of taurine in
breast milk, or feed infants with taurine-supplemented formulas and taurine-supple-
mented total parenteral nutrition [97,99,100]. On the other hand, the activities of taurine
biosynthetic enzymes are low in cats, dogs and foxes, and therefore they primarily depend
on a taurine-supplemented diet [101,102]. When fed with a diet deficient in taurine, these
animals developed pathologies such as cardiomyopathy and myocardial dysfunction
[101,103,104], retinal and tapetal degeneration that leads to blindness [105107], neurolog-
ical abnormalities [108,109], weakened immune response [110], pregnancy and fetal de-
velopment complications [111,112], as well as gastrointestinal problems [113,114]. In con-
trast, when fed with taurine-supplemented diets, these animals were protected against
pathologies such as cardiomyopathy [115,116], seizure [108,117], and retinopathy [118],
and showed improved reproductive performance and neurological development [119].
Taurine has also been added to energy drinks such as Red Bull, Monster, Tab Energy and
Rockstar [120]. It was estimated that on average, there is 750 mg of taurine in an 8 oz can
of energy drink [120]. While energy drinks mainly provide an energy boost, the exact role
of taurine in energy drinks remains unclear as energy drinks also contain additional ad-
ditives such as caffeine, ginseng, vitamins, antioxidants and sugars [121]. However, two
reviews by Kurtz et al. [122] and Seidel et al. [123] have described the influence of taurine
on exercise performance.
Figure 2.
Taurine (
5
) is synthesized from either methionine (
1
) or cysteine (
2
). Cysteine dioxygenase catalyzes the conversion
of cysteine (
2
) to cysteinesulfinate (
3
), which then is converted to hypotaurine (
4
) by cysteine sulfinate decarboxylase.
Hypotaurine (
4
) is readily oxidized to form taurine (
5
), which can be excreted directly or as a conjugate with bile acids such
as taurocholate (6).
Molecules 2021,26, 4913 4 of 21
In most mammals including humans, rodents and some primates, taurine is consid-
ered a conditionally essential amino acid, as cysteine dioxygenase and CSAD are present
abundantly. Mammals primarily depend on taurine biosynthesis
in vivo
[
91
] and par-
tially from diet, such as from meat, seafood and human milk [
92
94
]. Newborns and
young infants are unable to synthesize taurine as well as adult humans, and therefore,
are dependent on a taurine-supplemented diet [
95
]. Clinical studies investigating infants
supplemented with (30–40
µ
M/dL) or without taurine showed that inadequate taurine sup-
plementation impairs lipid absorption and bile acid secretion and causes hepatic and retinal
dysfunction [
96
99
]. Due to the importance of taurine in neonatal development, mothers
are strongly encouraged to breastfeed, due to the high concentration of taurine in breast
milk, or feed infants with taurine-supplemented formulas and taurine-supplemented total
parenteral nutrition [
97
,
99
,
100
]. On the other hand, the activities of taurine biosynthetic
enzymes are low in cats, dogs and foxes, and therefore they primarily depend on a taurine-
supplemented diet [
101
,
102
]. When fed with a diet deficient in taurine, these animals
developed pathologies such as cardiomyopathy and myocardial dysfunction [
101
,
103
,
104
],
retinal and tapetal degeneration that leads to blindness [
105
107
], neurological abnor-
malities [
108
,
109
], weakened immune response [
110
], pregnancy and fetal development
complications [
111
,
112
], as well as gastrointestinal problems [
113
,
114
]. In contrast, when
fed with taurine-supplemented diets, these animals were protected against pathologies
such as cardiomyopathy [
115
,
116
], seizure [
108
,
117
], and retinopathy [
118
], and showed
improved reproductive performance and neurological development [
119
]. Taurine has also
been added to energy drinks such as Red Bull, Monster, Tab Energy and Rockstar [
120
].
It was estimated that on average, there is 750 mg of taurine in an 8 oz can of energy
drink [
120
]. While energy drinks mainly provide an energy boost, the exact role of taurine
in energy drinks remains unclear as energy drinks also contain additional additives such
as caffeine, ginseng, vitamins, antioxidants and sugars [
121
]. However, two reviews by
Kurtz et al. [
122
] and Seidel et al. [
123
] have described the influence of taurine on exercise
performance.
Taurine is ubiquitously expressed in most tissues, particularly in the excitable tissues
such as the heart, retina, brain and muscles [
32
]. The intracellular concentration of taurine
is commonly 5–50 mM and the plasma concentration of taurine is approximately 100
µ
M.
When taurine is supplemented, the plasma taurine content usually reaches its peak within
1 h to 2.5 h of taurine intake [
124
,
125
]. Ghandforoush-Sattari et al. [
124
] conducted an
analysis on the pharmacokinetics of oral taurine supplementation (4 g) in healthy adults.
These individuals, who had fasted overnight, showed a baseline taurine content in a
range of 30
µ
mol to 60
µ
mol. Then, 1.5 h after taurine intake, the plasma taurine content
increased to approximately 500
µ
mol. Plasma taurine content subsequently decreased to
baseline level 6.5 h after taurine intake. This study would be consistent with the common
notion that excess plasma taurine is mostly excreted via urine or being transported to
tissues. As taurine is only synthesized in the liver, maintenance of a high concentration
of taurine in other tissues depends on the taurine uptake from the blood via a sodium-
dependent taurine transporter (TauT). This TauT has a higher affinity for
β
-amino acids,
such as taurine, but a lower affinity for
α
-amino acids [
90
]. The importance of this taurine
transporter has been evidenced in mouse models lacking the TauT gene [
126
,
127
], as well
as in
in vivo
and
in vitro
studies utilizing taurine transporter competitive inhibitors, such
as β-alanine [128,129] or guanidinoethanesulfonate (GES) [130,131].
Two mouse models lacking the TauT (TauTKO) were generated by Ito’s group [
126
] and
Warskulat’s group [
127
]. Both TauTKO mouse models had reduced taurine concentrations
in the heart, skeletal muscles and retina, validating the requirement of the taurine transport
from the liver to these tissues [
126
,
127
]. As a consequence of taurine deficiency, these
TauTKO mice developed retinal degeneration, chronic liver disease, muscle atrophy, a
decrease in exercise capacity and increased susceptibility to streptozotocin-induced diabetic
nephropathy [
126
,
127
,
132
134
]. The TauTKO mice developed by the Ito’s group also
showed evidence of cardiomyopathy, as indicated by diminished fractional shortening;
Molecules 2021,26, 4913 5 of 21
ventricular remodeling, as shown by dilated ventricles; and reductions in ventricular
wall thickness, as well as increased expression of fetal genes that serve as heart failure
markers, such as atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP) and
β
-myosin heavy chain (MHC) [
126
]. Further examination of the TauTKO hearts revealed
mitochondrial swelling and disruption of the outer mitochondrial membrane, as well
as a reduction in the activity of succinate dehydrogenase (SDH), which is a marker of
mitochondrial enzyme [
126
]. In addition, TauTKO hearts contained defective mitochondria,
as evidenced by smaller mitochondria size, impaired activities of the electron transport
chain, oxidative stress and apoptosis [
135
]. The TauTKO hearts were also associated with
impaired autophagy, which is the cellular quality control in degrading damaged proteins
or organelles [
136
,
137
]. Defective mitochondria and impaired autophagy in TauTKO hearts,
therefore, may contribute to the underlying development of cardiomyopathy in TauTKO
mice. In addition, the same TauTKO mice showed premature aging, as characterized by
shortened lifespan and acceleration of skeletal muscle senescence [
138
]. Interestingly, while
the TauTKO mice developed by the Warskulat’s group showed normal cardiac function,
the expression of fetal genes such as ANP, BNP and CARP (cardiac ankyrin repeat protein)
increased in the TauTKO hearts, suggesting taurine depletion may predispose the mice to
the development of a heart failure [
127
]. Based on the studies from these mouse models
that lacked the TauT, it is convincing that taurine indeed has multiple physiological roles
that include maintaining mitochondrial function.
Similarly, pharmacological inhibition of taurine transport using
β
-alanine or GES
in both
in vitro
and
in vivo
studies resulted in significant pathological conditions, which
include atrophic cardiac remodeling [
126
,
139
,
140
], oxidative stress [
141
,
142
], increased
apoptosis [
143
], mitochondrial defects [
83
,
84
] and altered cardiac cell morphology [
144
],
as well as loss of retinal ganglion cells that lead to retinopathy [
128
]. All these studies
clearly demonstrated the importance of taurine as a cytoprotective agent with multiple
physiological functions. Recently, increasing studies have focused on the antioxidant role
of taurine in maintaining mitochondrial function.
5. Taurine as a Therapeutic Agent in Mitochondrial Dysfunction
Various
in vitro
and
in vivo
studies have reported that taurine supplementation pro-
tects against mitochondrial dysfunction. Homma et al. [
82
] recently showed that taurine
protects against metabolic impairment and mitochondrial dysfunction in MELAS patient-
derived undifferentiated induced pluripotent stem cells (iPSCs) and the iPSC-derived
retinal pigment epithelium (RPE). In a clinical study on oral taurine supplementation
among MELAS patients, Ohsawa et al. [
27
] reported that taurine reduces the incidence of
stroke-like episodes and increases the taurine modification of mitochondrial tRNA
Leu(UUR)
.
Shetewy et al. [
83
] showed that taurine pretreatment protects against mitochondria dam-
age and mitochondria fission in beta-alanine-treated mouse embryonic fibroblasts. Jong
et al. [
84
] also showed that taurine pretreatment protects against the effect of beta-alanine-
mediated taurine depletion on the opening of the mitochondrial permeability transition
pore and subsequently inhibited apoptosis. In rat cardiomyocytes, taurine supplementa-
tion inhibits glucose-deprivation-induced mitochondrial oxidative stress, mitochondrial
dysfunction, apoptosis and ER stress [
78
]. The aforementioned studies are some examples
describing the protective roles of taurine in maintaining mitochondria health. In the follow-
ing sections, we describe several mechanisms by which taurine may regulate mitochondrial
health. All these mechanisms are summarized in Figure 3.
Molecules 2021,26, 4913 6 of 21
Molecules 2021, 26, x FOR PEER REVIEW 6 of 22
5. Taurine as a Therapeutic Agent in Mitochondrial Dysfunction
Various in vitro and in vivo studies have reported that taurine supplementation pro-
tects against mitochondrial dysfunction. Homma et al. [82] recently showed that taurine
protects against metabolic impairment and mitochondrial dysfunction in MELAS patient-
derived undifferentiated induced pluripotent stem cells (iPSCs) and the iPSC-derived ret-
inal pigment epithelium (RPE). In a clinical study on oral taurine supplementation among
MELAS patients, Ohsawa et al. [27] reported that taurine reduces the incidence of stroke-
like episodes and increases the taurine modification of mitochondrial tRNALeu(UUR). Shet-
ewy et al. [83] showed that taurine pretreatment protects against mitochondria damage
and mitochondria fission in beta-alanine-treated mouse embryonic fibroblasts. Jong et al.
[84] also showed that taurine pretreatment protects against the effect of beta-alanine-me-
diated taurine depletion on the opening of the mitochondrial permeability transition pore
and subsequently inhibited apoptosis. In rat cardiomyocytes, taurine supplementation in-
hibits glucose-deprivation-induced mitochondrial oxidative stress, mitochondrial dys-
function, apoptosis and ER stress [78]. The aforementioned studies are some examples
describing the protective roles of taurine in maintaining mitochondria health. In the fol-
lowing sections, we describe several mechanisms by which taurine may regulate mito-
chondrial health. All these mechanisms are summarized in Figure 3.
Figure 3. Taurine is known not as a radical scavenger. Several potential mechanisms by which tau-
rine exerts its antioxidant activity in maintaining mitochondria health include: taurine conjugates
with uridine on mitochondrial tRNA to form a 5-taurinomethyluridine for proper synthesis of mi-
tochondrial proteins (mechanism 1), which regulates the stability and functionality of respiratory
chain complexes; taurine reduces superoxide generation by enhancing the activity of intracellular
antioxidants (mechanism 2); taurine prevents calcium overload and prevents reduction in energy
production and the collapse of mitochondrial membrane potential (mechanism 3); taurine directly
scavenges HOCl to form N-chlorotaurine in inhibiting a pro-inflammatory response (mechanism 4);
and taurine inhibits mitochondria-mediated apoptosis by preventing caspase activation or by re-
storing the Bax/Bcl-2 ratio and preventing Bax translocation to the mitochondria to promote apop-
tosis (mechanism 5).
5.1. Taurine Forms a Complex with Mitochondrial tRNA
Taurine is primarily a free amino acid, although it does conjugate with bile acids to
form taurocholate [90]. In 2002, a group of Japanese scientists discovered that taurine is a
Figure 3.
Taurine is known not as a radical scavenger. Several potential mechanisms by which
taurine exerts its antioxidant activity in maintaining mitochondria health include: taurine conjugates
with uridine on mitochondrial tRNA to form a 5-taurinomethyluridine for proper synthesis of
mitochondrial proteins (mechanism 1), which regulates the stability and functionality of respiratory
chain complexes; taurine reduces superoxide generation by enhancing the activity of intracellular
antioxidants (mechanism 2); taurine prevents calcium overload and prevents reduction in energy
production and the collapse of mitochondrial membrane potential (mechanism 3); taurine directly
scavenges HOCl to form N-chlorotaurine in inhibiting a pro-inflammatory response (mechanism
4); and taurine inhibits mitochondria-mediated apoptosis by preventing caspase activation or by
restoring the Bax/Bcl-2 ratio and preventing Bax translocation to the mitochondria to promote
apoptosis (mechanism 5).
5.1. Taurine Forms a Complex with Mitochondrial tRNA
Taurine is primarily a free amino acid, although it does conjugate with bile acids to
form taurocholate [
90
]. In 2002, a group of Japanese scientists discovered that taurine is
a component of the mitochondrial tRNAs [
145
]. Specifically, they identified two taurine-
containing modified uridines, namely 5-taurinomethyluridine (
τ
m
5
u) and 5-taurinomethyl-
2-thiouridine (
τ
m
5
s
2
u). These conjugates are linked to the role of taurine as an antioxi-
dant. Taurine conjugates with uridine at the anticodon wobble position of mitochondrial
tRNA
Leu(UUR)
or mitochondrial tRNA
Lys
to form
τ
m
5
u and
τ
m
5
s
2
u, respectively [
145
,
146
].
These conjugation reactions, which are catalyzed by the mitochondrial optimization 1
(Mto1) in mammals [
147
], are required for precise anticodon–codon interactions for proper
synthesis of mitochondrial-encoded proteins [148].
Based on the wobble hypothesis, the nucleoside at the first position of the anticodon
forms hydrogen bonds with the third nucleoside of the codon, forming a wobble base pair.
Normally, uridine at the anticodon wobble position base pairs with either adenine (A) or
guanine (G) at the codon position of mRNA, which translates for leucine codons (UUA
and UUG). An unmodified uridine at the anticodon wobble position can base pair with
all four bases, A, G, cytosine (C) and uracil (U) at the third position of the codon [
146
].
These pairings subsequently result in mistranslation, as it also translates for phenylalanine
codons (UUC and UUU) in addition to the usual leucine codons. However, when taurine
conjugates with uridine at the anticodon wobble position, the taurine-modified uridine base
pairs only with either adenine or guanine of the corresponding codons and translates for
Molecules 2021,26, 4913 7 of 21
leucine codons (UUA and UUG) [
145
,
148
,
149
]. This proper anticodon–codon interaction is
significantly important for proper expression of mitochondrial-encoded proteins [
149
,
150
],
implicating the significance of post-transcriptional modification of uridine through taurine
conjugation at the anticodon wobble position of mitochondrial tRNA
Leu(UUR)
. The impor-
tance of post-transcriptional modifications in tRNAs lies not only in ensuring proper codon
recognition for translation accuracy, they are also essential in improving the efficiency
of tRNA and facilitating the codon recognition by elongation factors or aminoacyl-tRNA
synthetases. Consequently, a mutation in the post-transcriptional modification of mito-
chondrial tRNA
Leu(UUR)
can affect protein synthesis as it can decrease RNase P processing,
tRNA stability and aminoacylation, resulting in an abnormal tRNA conformation [150].
A defect in taurine conjugation of mitochondrial tRNA
Leu(UUR)
, which prevents the
formation of 5-taurinomethyl-uridine, has been implicated in mitochondrial myopathy,
encephalopathy, lactic acidosis and stroke-like episodes (MELAS), as well as myoclonus
epilepsy associated with ragged-red fibers (MERRF) [
82
,
145
,
146
,
148
,
149
]. Indeed, an early
study by Kirino et al. [
149
] showed that unmodified uridine of mitochondrial tRNA
Leu(UUR)
weakens the binding affinity for the UUG codon, which could result in inefficient synthesis
of mitochondrial proteins. Among the 13 polypeptides that encode mitochondrial proteins,
ND5, ND6 and cytochrome b contain two, eight and two UUG codons, respectively. A
deficiency in taurine conjugation of mitochondrial tRNA
Leu(UUR)
, therefore, may affect the
synthesis of mitochondrial proteins. Indeed, Jong et al. [
129
] have shown a reduction in
the protein levels of ND5 and ND6 in beta-alanine-treated cells. Both ND5 and ND6 are
components of the complex I of the electron transport chain. A reduction in the expres-
sion of mitochondrial proteins ND5 and ND6 then cause the instability of the complex I,
which could cause a sluggish transport of electrons across the respiratory chain, as well as
diversion of electrons to oxygen to form a superoxide. Excessive superoxide production
then can promote oxidative stress and overwhelm the antioxidant defenses [
84
,
129
]. In-
deed, a recent study by Fakruddin et al. [
147
] showed that defective taurine-containing
mitochondrial tRNA modification causes mitochondrial dysfunction and disrupts global
protein homeostasis, thereby suggesting the significance of taurine-containing uridine
modification in regulating global protein homeostasis. When taurine was supplemented,
Homma et al. [
82
] observed an increase in the taurine modification of the mitochondrial
tRNA
Leu(UUR)
, as well as an improvement in the mitochondrial function in the iPSC gen-
erated from a MELAS patient. While many other studies [
28
,
78
,
81
84
,
88
,
89
,
93
,
99
,
117
,
145
]
have shown that taurine supplementation protects against mitochondrial dysfunction
without definite underlying mechanisms, it is likely that the antioxidant function of taurine
is associated with its role in the conjugation reaction with the uridine of the mitochondrial
tRNALeu(UUR). However, this matter will require further validation.
5.2. Taurine Reduces Superoxide Generation in the Mitochondria
There have been many studies showing taurine as an antioxidant with a role in pro-
tecting against oxidative stress in the mitochondria [
27
,
28
,
78
,
81
84
,
88
,
89
,
93
,
99
,
117
,
136
,
145
].
However, the underlying mechanism by which taurine protects against oxidative stress in
the mitochondria remains unclear, as Aruoma et al. [
151
] showed that taurine is not a radical
scavenger. It is important to note that while taurine is incapable of scavenging classical ROS,
taurine is a direct scavenger of hypochlorous acid (HOCl), which is generated from hydro-
gen peroxide (H
2
O
2
) in the presence of chloride ions, producing N-chlorotaurine [
151
,
152
].
The role of N-chlorotaurine is mainly in regulating the inflammatory response. Specifically,
N-chlorotaurine has been shown to activate nuclear factor (erythroid-derived 2)-like 2
(Nrf2), which is a transcription factor that controls the transcription of various antioxidant
genes and subsequently prevents inflammation [153155].
While recent studies have suggested that the prevention of superoxide in the mito-
chondria is linked to the taurine conjugation of the mitochondrial tRNA [
28
,
82
,
84
,
147
],
several studies have shown that taurine may exert its antioxidant function via different
mechanisms. In germ cells, taurine has been shown to protect against oxidative stress by
Molecules 2021,26, 4913 8 of 21
promoting the activity of Cu/Zn SOD [
156
]. Cu/Zn SOD is localized in the mitochondrial
intermembrane space activity [
157
,
158
]. Indeed, taurine increases the protein levels but
not mRNA levels of Cu/Zn SOD, suggesting that taurine mediates the effects of Cu/Zn
SOD at the protein level. In another study by Tabassum et al. [
159
], the antioxidant role
of taurine was attributed to the enhancement of intracellular reduced glutathione (GSH).
GSH is essential for detoxification of xenobiotics, where GSH is oxidized to GSSG (oxidized
glutathione) during oxidative stress. In a tamoxifen-treated liver, there is a reduction
in the GSH levels, which increases the susceptibility to oxidative stress due to impaired
antioxidant defense system. When tamoxifen-treated mice were co-treated with taurine,
the GSH level was restored and oxidative stress was prevented. These findings suggest the
physiological role of taurine in stabilizing the intracellular GSH levels. Other earlier studies
by Pasantes et al. [
160
162
] have also suggested that taurine protects lipid membranes
from tamoxifen-induced oxidative damage by acting as a membrane stabilizer, rather than
directly acting against the oxidants. Indeed, a review by Hansen et al. [
163
] described the
role of taurine as a buffer in the mitochondria matrix to stabilize the pH gradient across the
inner mitochondrial membranes.
5.3. Taurine Regulates Intracellular Calcium Homeostasis
Several studies from El-Idrissi’s group have shown that taurine regulates intracellu-
lar calcium homeostasis and protects against glutamate-induced mitochondrial damage
and cell death [
164
168
]. In general, glutamate increases the intracellular calcium level
and causes the collapse of the mitochondrial membrane potential and induces cell death.
However, when cultured cerebellar granule cells were pretreated with glutamate, taurine
prevented an increase in the mitochondrial calcium level, prevented mitochondrial mem-
brane depolarization and prevented an impairment in the mitochondrial function [
166
].
One of the functions of mitochondria is energy metabolism, which is regulated by cal-
cium. In glutamate-induced excitotoxicity, a collapse in the mitochondrial membrane
potential causes a depletion in the energy production, as measured by ATP levels, and
promotes neuronal death. However, when neuronal cells were pretreated with taurine,
the glutamate-induced excitotoxicity effects were inhibited, along with increased energy
metabolism [
165
]. Meanwhile, additional studies [
169
172
] have shown taurine also regu-
lates calcium homeostasis to maintain cardiac contractile function. In general, heart failure
is caused by impaired contraction due to calcium mishandling, which reduces the calcium
sensitivity of the contractile proteins, and insufficient ATP generation to drive contraction.
Studies by Steele et al. [
173
] and Galler et al. [
174
] showed that physiological concentrations
of taurine increase the calcium sensitivity of contractile proteins and modulate cardiac
contractility. As calcium is known to regulate mitochondrial oxidative phosphorylation to
produce ATP [
175
], regulation of intracellular calcium homeostasis by taurine can improve
the energy production through maintenance of mitochondrial function.
5.4. Taurine Inhibits Mitochondria-Mediated Apoptosis
It has been thought that taurine acts at the level of mitochondria to inhibit apopto-
sis. Indeed, Jong et al. [
135
] showed that TauTKO hearts pretreated with mitotempo, a
mitochondria-targeted antioxidant, were protected against oxidative stress and mitochon-
drial apoptosis. Interestingly, Takatani et al. [
79
] showed that taurine pretreatment does not
prevent the release of cytochrome c and the reduction in mitochondria membrane potential
during ischemia. However, taurine pretreatment prevents ischemia-induced cleavage
of caspase 9 and caspase 3 [
79
]. Generally in apoptosis signaling, cytochrome c, Apaf-1
and caspase 9 form a complex known as apoptosome, which activates caspase 9 and the
subsequent downstream caspase-3-mediated signaling cascade [
176
]. In another study by
Leon et al. [
177
], it was shown that taurine protects against glutamate-induced apoptosis
by inhibiting glutamate-induced membrane depolarization, potentially by acting on the
chloride channels and preventing excessive calcium influx. As a result, glutamate-induced
activation of calpain is inhibited, which then prevents Bax translocation to the mitochondria
Molecules 2021,26, 4913 9 of 21
and the subsequent cytochrome c release. Similar mechanisms have also been reported
by Wu and Prentice [
178
,
179
]. Similarly, Taranukhin et al. [
180
] showed that taurine pre-
vents ethanol-induced mitochondria-mediated apoptosis by increasing the levels of Bcl-2.
Restoration of Bax:Bcl-2 levels prevents Bax translocation to the mitochondria, and thus
protects against activation of the mitochondria-mediated apoptosis.
6. Clinical Application of Taurine in Mitochondria-Targeted Pathologies
Several clinical studies investigating the therapeutic potential of taurine have been
reported, particularly in relation to its role as an antioxidant in improving mitochondrial
function. Below, we report several known clinical studies on taurine supplementation in
various pathologies in humans.
6.1. Cardiovascular Diseases
Taurine was first used to treat heart failure patients by Azuma’s group in
Japan [
29
,
30
,
181
]. Most heart failure patients receiving taurine supplementation (2–3 g
taurine daily for a range of four to eight weeks) showed improvement in the systolic left
ventricular function, as evidenced by increased cardiac output and stroke volume, ejection
fraction and mean velocity of circumferential fiber shortening. When comparing the effects
of taurine to coenzyme Q
10
, no significant improvement in the systolic left ventricular
function was observed [
181
]. Coenzyme Q
10
is an antioxidant that mediates the transfer
of electrons from complex I and complex II to complex III [
42
]. In heart failure patients,
the levels of coenzyme Q
10
were reduced and supplementation with coenzyme Q
10
was
shown to improve the symptoms among heart failure patients [
182
184
]. The effectiveness
of taurine, but not coenzyme Q
10
, in improving myocardial energy in heart failure suggests
the significant effect of taurine as an antioxidant, potentially by improving the myocardial
energy production. In heart failure, a disturbance in ATP production affects myocardial
contraction [
185
,
186
], thereby suggesting the significance of taurine in improving mito-
chondrial function through restoration of myocardial energy production. Similarly, another
study conducted among heart failure patients in NYHA class II or III receiving 500 mg
of taurine supplementation three times a day for two weeks showed improved exercise
capacity [
31
]. Again, this study implicates the role of taurine as an antioxidant to improve
mitochondrial function, potentially through restoration of energy production.
Taurine has also been supplemented among patients with hypertension. Taurine
supplementation of 1.6 g per day for at least 12 weeks in prehypertensive patients or 6 g
per day for 7 days in hypertensive patients lowered blood pressure and improved vascular
function [
187
,
188
]. In addition, worldwide epidemiological studies among 61 different pop-
ulation groups in 25 different countries have reported that regular taurine intake through
daily consumption of seafood, nuts, soy and milk reduces the prevalence of cardiovascular
diseases that include hypertension and hypercholesterolemia [
94
,
189
192
]. While the un-
derlying mechanisms by which taurine attenuates hypertension remain unclear, several
in vitro
and
in vivo
studies have suggested the antioxidant activity of taurine in reducing
hypertension, which include reducing ROS generation [
193
,
194
], improving ATP produc-
tion [
195
] and improving mitochondrial metabolism [
196
]. Indeed, hypertension has been
largely associated with mitochondrial dysfunction, which includes mitochondrial oxidative
stress [197], alteration of mitochondrial homeostasis and impaired energy production.
6.2. Metabolic Syndrome
Taurine supplementation among patients with type II diabetes mellitus also showed
reduced oxidative stress and inflammation, as well as reduced diabetic complications,
such as nephropathy, retinopathy and neuropathy [
198
200
]. In diabetic patients, plasma
taurine concentrations have been reported to be reduced when compared to healthy control
patients [201203]. This reduction is associated with higher renal excretion of taurine and
a lower intestinal absorption of taurine, suggesting a depletion in the bioavailability of
taurine in diabetic patients [
199
,
201
203
]. Normal plasma taurine concentration has been
Molecules 2021,26, 4913 10 of 21
reported to be about 44
µ
mol/L, while the normal whole blood taurine concentration is
averaged to be 227
µ
mol/L in fasting humans [
204
]. Indeed, several
in vitro
and
in vivo
studies have also reported reduced taurine concentrations in diabetic models, which often
are associated with impaired glucose tolerance, insulin resistance and impaired glucose
and lipid metabolism [
201
,
202
,
205
207
]. It is well known that diabetic complications
arise from hyperglycemia-induced oxidative stress, which usually originates from the
mitochondrial respiratory chain as the primary source [
208
]. As a result of mitochondrial
oxidative stress, the functions of the mitochondria are impaired, which are the main
source of glucose and fatty acid metabolism. When taurine is supplemented, glucose
levels are restored, insulin secretion is enhanced and glucose and lipid metabolism in the
mitochondria are stimulated [
209
212
]. As an antioxidant, taurine potentially suppresses
excessive oxidants generation in the mitochondria and maintains the functionality of
mitochondria. While taurine is not known as a radical scavenger [
151
], the underlying
mechanism of taurine as an antioxidant could potentially occur through conjugation with
uridine of mitochondrial tRNA, equilibration of intracellular antioxidants or stabilization
of mitochondrial homeostasis.
Despite the effectiveness of taurine in reducing diabetic complications in several
clinical trials, there are a few studies contradicting the clinical effectiveness of taurine in
diabetic patients. Firstly, Franconi et al. [
202
] reported that oral taurine supplementation of
1.5 g/day for 90 days in patients with insulin-dependent diabetes mellitus did not improve
glucose metabolism, although plasma taurine concentration increased significantly in dia-
betic patients. Secondly, Chauncey et al. [
213
] reported that oral taurine supplementation
of 3 g/day for 4 months among type 2 diabetic patients increased plasma taurine content
but did not reduce the plasma glucose levels. Thirdly, Nakamura et al. [
214
] reported that
oral taurine supplementation of 3 g/day for 12 months in patients with microalbuminuria
of non-insulin-dependent diabetes mellitus increased plasma taurine content but did not
reduce microalbuminuria, which is a strong marker of diabetic nephropathy.
Taurine has also been administered in overweight or obese individuals, which has
shown promising therapeutic effects on lipid profiles. Rosa et al. [
215
] reported that obese
women supplemented with 3 g/day of taurine for eight weeks had a significant increase
in the content of both plasma taurine and adiponectin and a significant decrease in pro-
inflammatory markers and the lipid peroxidation marker. In a separate study by Mizushima
et al. [
216
], healthy individuals were given high-fat and high-cholesterol diets for three
weeks. At the same time, when these individuals were also given 6 g/day of taurine for
three weeks, the increase in the levels of total cholesterol and low density lipoprotein (LDL)-
cholesterol was attenuated. De Carvalho et al. [
217
] recently showed that a combination of
3 g taurine supplementation and exercise training for eight weeks among obese women
improved lipid metabolism and mitochondrial activity in the subcutaneous white adipose
tissue. In addition, regular taurine intake through daily consumption of seafood, nuts,
soy and milk has been shown to reduce the prevalence of metabolic syndrome, which
includes obesity [
94
,
189
192
]. This observation is based on worldwide epidemiological
studies conducted in more than 60 different populations around the world [
218
]. The
antioxidant role of taurine in modulating the lipid profiles in obesity is likely to occur
through maintenance of mitochondrial homeostasis and suppression of oxidative stress that
stimulates lipid metabolism and subsequent restoration of energy production [
215
217
].
Indeed, this is consistent with several
in vivo
and
in vitro
studies on metabolic syndrome,
showing the suppression of lipid peroxidation and restoration of mitochondrial function
with taurine treatment [219223].
6.3. Mitochondrial Diseases
Recent studies on mitochondrial diseases such as MELAS and MERRF revealed a
reduction in the formation of taurine-modified uridine of mitochondrial tRNAs, specifically
5-taurinomethyluridine (
τ
m
5
u) and 5-taurinomethyl-2-thiouridine (
τ
m
5
s
2
u) [
145
,
148
,
149
].
When taurine was supplemented, Homma et al. [
82
] observed an increase in the taurine
Molecules 2021,26, 4913 11 of 21
modification of the mitochondrial tRNA
Leu(UUR)
, as well as improvement of the mitochon-
drial function in the iPSC generated from a MELAS patient. Indeed, oral taurine supple-
mentation of either 9 g or 12 g per day for 52 weeks in MELAS patients showed prevention
of stroke-like episodes, as well as increased modification of taurine-conjugated uridine
of mitochondrial tRNA
Leu(UUR)
[
27
,
28
]. In a separate study by Fukuda and Nagao [
224
],
plasma taurine content was reduced in one Japanese man diagnosed with MELAS and ma-
ternally inherited diabetes and deafness (MIDD). While the taurine-containing modification
of mitochondrial tRNA was not examined in this study, it is plausible that the formation of
taurine-modified uridine was absent due to low plasma taurine content. Therefore, it is
likely that oral taurine supplementation in this MELAS/MIDD patient could potentially
increase the plasma taurine content, enhance taurine-containing modification of uridine
and improve his clinical symptoms. Indeed, the significance of the taurine-containing
modification of uridine has been established in several
in vitro
and
in vivo
studies that
demonstrated the efficiency of mitochondrial protein synthesis, improvement of mitochon-
drial respiratory activity, restoration of energy production, suppression of oxidative stress
and maintenance of global protein homeostasis [27,28,82,145,148,149,224].
6.4. Neurological Disorders
Currently, there are no reported clinical studies on taurine supplementation among
patients with neurodegenerative diseases. However, there have been some
in vivo
studies
assessing the therapeutic effects of taurine in several mouse models of neurodegenerative
diseases, which include Parkinson’s disease [
225
,
226
] and Alzheimer’s disease [
227
230
].
In these studies, it has been concluded that taurine suppresses pathological changes by
maintaining the mitochondrial homeostasis. A common feature in neurodegenerative
diseases is glutamate-mediated hyperexcitability that leads to calcium overload, collapse in
mitochondrial membrane potential and increased ROS production [
231
236
]. As mitochon-
drial damage is commonly observed in neurodegenerative diseases [
233
235
], it is sensible
to presume that the cellular damage is caused by the mitochondrial ROS production.
Indeed, several studies investigating the underlying mechanisms of neurodegenerative
diseases have reported a reduction in the respiratory chain activity, a decrease in the ATP
production, a collapse in the mitochondrial membrane potential and an increase in mi-
tochondrial ROS production [
11
,
234
,
235
,
237
,
238
]. As taurine is known to modulate the
mitochondrial function, it is plausible that taurine supplementation would improve the
clinical symptoms in patients with neurodegenerative diseases.
The clinical effectiveness of taurine in neurodevelopmental disorders has been re-
ported in several studies by Erickson et al. [
239
243
]. These clinical studies used acam-
prosate, which is a synthetic taurine analogue. Acamprosate is approved by the United
States Food and Drug Administration (FDA) to treat alcohol dependence [
244
]. In the
first study, oral acamprosate supplementation of 1 g/day for 21 weeks in three adults
with Fragile-X syndrome showed significant improvement in communication [
240
]. In the
second study, oral acamprosate supplementation of 1 g/day for 10 weeks in twelve young
children with Fragile-X syndrome and comorbid autism spectrum disorders showed signif-
icant improvement in social skills and inattention/hyperactivity [
243
]. Additional studies
on oral acamprosate supplementation of 1 g/day for 20 weeks in young children with
autism spectrum disorders showed significant improvement in social deficits [
239
,
241
,
242
].
Neurodevelopmental disorders are commonly characterized by excessive glutamater-
gic and deficient GABAergic neurotransmission, which is associated with social impair-
ment [
245
247
]. Glutamate excitotoxicity can lead to calcium overload, collapse in mito-
chondrial membrane potential and increased mitochondrial ROS production, which can
cause mitochondrial dysfunction [
231
236
]. Many studies have increasingly shown the
link between neurodevelopmental disorders and mitochondrial dysfunction [
248
250
]. As
acamprosate is an analogue of taurine, which is an antioxidant that maintains the mito-
chondrial homeostasis, it is plausible that acamprosate improves the clinical symptoms in
neurodevelopmental disorders by maintaining the mitochondrial homeostasis.
Molecules 2021,26, 4913 12 of 21
7. Conclusions
Taurine is a simple but unique sulfur-containing amino acid that has multiple phys-
iological functions, including the maintenance of mitochondria health. While taurine is
widely known as an antioxidant, its underlying mechanism remains unclear as taurine
is not a radical scavenger. Studies conducted by Suzuki and colleagues have shown that
taurine conjugates with mitochondrial tRNA
Leu(UUR)
or tRNA
Lys(UUU)
for proper codon–
anticodon interaction to facilitate synthesis of mitochondrial-encoded proteins. Inefficiency
of taurine modification of mitochondrial tRNA promotes inefficient translation of mito-
chondrial proteins, which are components of the electron transport chain. As a result,
the assembly and stability of the respiratory chain complexes are impaired, which causes
a sluggish response in the electron flux, yielding superoxide production. Taurine also
regulates intracellular calcium homeostasis and intracellular antioxidant activity and in-
hibits apoptosis. As evidenced in many
in vitro
,
in vivo
and clinical studies on taurine
supplementation, the occurrence of mitochondrial dysfunction, reduced energy production,
oxidative stress and apoptosis are mostly inhibited. Taurine therapy, therefore, could po-
tentially improve mitochondrial health, particularly in mitochondria-targeted pathologies,
such as cardiovascular diseases, metabolic diseases, mitochondrial diseases and neurologi-
cal disorders. Whether the protective mechanism on mitochondria primarily relies on the
taurine modification of mitochondrial tRNA requires further investigation.
Author Contributions:
Writing—original draft preparation, C.J.J.; writing—review and editing,
C.J.J. and P.S.; supervision, S.W.S. All authors have read and agreed to the published version of the
manuscript.
Funding: This research received no external funding.
Conflicts of Interest: The authors declare no conflict of interest.
References
1. Harman, D. Aging: A theory based on free radical and radiation chemistry. J. Gerontol. 1956,11, 298–300. [CrossRef]
2. Harman, D. The biologic clock: The mitochondria? J. Am. Geriatr. Soc. 1972,20, 145–147. [CrossRef]
3.
Scheubel, R.J.; Tostlebe, M.; Simm, A.; Rohrbach, S.; Prondzinsky, R.; Gellerich, F.N.; Silber, R.E.; Holtz, J. Dysfunction of
mitochondrial respiratory chain complex I in human failing myocardium is not due to disturbed mitochondrial gene expression.
J. Am. Coll. Cardiol. 2002,40, 2174–2181. [CrossRef]
4.
Marin-Garcia, J.; Goldenthal, M.J.; Moe, G.W. Mitochondrial pathology in cardiac failure. Cardiovasc. Res.
2001
,49, 17–26.
[CrossRef]
5.
Shapira, Y.; Cederbaum, S.D.; Cancilla, P.A.; Nielsen, D.; Lippe, B.M. Familial poliodystrophy, mitochondrial myopathy, and
lactate acidemia. Neurology 1975,25, 614–621. [CrossRef]
6.
Hayashi, G.; Cortopassi, G. Oxidative stress in inherited mitochondrial diseases. Free Radic. Biol. Med.
2015
,88, 10–17. [CrossRef]
[PubMed]
7.
Bournat, J.C.; Brown, C.W. Mitochondrial dysfunction in obesity. Curr. Opin. Endocrinol. Diabetes Obes.
2010
,17, 446–452.
[CrossRef] [PubMed]
8.
Prasun, P. Role of mitochondria in pathogenesis of type 2 diabetes mellitus. J. Diabetes Metab. Disord.
2020
,19, 2017–2022.
[CrossRef] [PubMed]
9. Zong, W.X.; Rabinowitz, J.D.; White, E. Mitochondria and Cancer. Mol. Cell 2016,61, 667–676. [CrossRef]
10.
Modica-Napolitano, J.S.; Singh, K.K. Mitochondrial dysfunction in cancer. Mitochondrion
2004
,4, 755–762. [CrossRef] [PubMed]
11.
Wang, W.; Zhao, F.; Ma, X.; Perry, G.; Zhu, X. Mitochondria dysfunction in the pathogenesis of Alzheimer’s disease: Recent
advances. Mol. Neurodegener. 2020,15, 30. [CrossRef]
12.
Pallardo, F.V.; Lloret, A.; Lebel, M.; D’Ischia, M.; Cogger, V.C.; Le Couteur, D.G.; Gadaleta, M.N.; Castello, G.; Pagano, G.
Mitochondrial dysfunction in some oxidative stress-related genetic diseases: Ataxia-Telangiectasia, Down Syndrome, Fanconi
Anaemia and Werner Syndrome. Biogerontology 2010,11, 401–419. [CrossRef]
13.
Griffiths, K.K.; Levy, R.J. Evidence of Mitochondrial Dysfunction in Autism: Biochemical Links, Genetic-Based Associations, and
Non-Energy-Related Mechanisms. Oxid. Med. Cell. Longev. 2017,2017, 4314025. [CrossRef]
14. Haas, R.H. Autism and mitochondrial disease. Dev. Disabil. Res. Rev. 2010,16, 144–153. [CrossRef]
15.
Negida, A.; Menshawy, A.; El Ashal, G.; Elfouly, Y.; Hani, Y.; Hegazy, Y.; El Ghonimy, S.; Fouda, S.; Rashad, Y. Coenzyme Q10 for
Patients with Parkinson’s Disease: A Systematic Review and Meta-Analysis. CNS Neurol. Disord. Drug Targets
2016
,15, 45–53.
[CrossRef]
Molecules 2021,26, 4913 13 of 21
16.
Rossman, M.J.; Santos-Parker, J.R.; Steward, C.A.C.; Bispham, N.Z.; Cuevas, L.M.; Rosenberg, H.L.; Woodward, K.A.; Chonchol,
M.; Gioscia-Ryan, R.A.; Murphy, M.P.; et al. Chronic Supplementation With a Mitochondrial Antioxidant (MitoQ) Improves
Vascular Function in Healthy Older Adults. Hypertension 2018,71, 1056–1063. [CrossRef] [PubMed]
17.
Snow, B.J.; Rolfe, F.L.; Lockhart, M.M.; Frampton, C.M.; O’Sullivan, J.D.; Fung, V.; Smith, R.A.; Murphy, M.P.; Taylor, K.M.; Protect
Study, G. A double-blind, placebo-controlled study to assess the mitochondria-targeted antioxidant MitoQ as a disease-modifying
therapy in Parkinson’s disease. Mov. Disord. 2010,25, 1670–1674. [CrossRef] [PubMed]
18.
Sozen, E.; Demirel, T.; Ozer, N.K. Vitamin E: Regulatory role in the cardiovascular system. IUBMB Life
2019
,71, 507–515.
[CrossRef] [PubMed]
19.
Le Bars, P.L.; Katz, M.M.; Berman, N.; Itil, T.M.; Freedman, A.M.; Schatzberg, A.F. A placebo-controlled, double-blind, randomized
trial of an extract of Ginkgo biloba for dementia. North American EGb Study Group. JAMA
1997
,278, 1327–1332. [CrossRef]
[PubMed]
20.
Yamaguchi, T.; Sano, K.; Takakura, K.; Saito, I.; Shinohara, Y.; Asano, T.; Yasuhara, H. Ebselen in acute ischemic stroke: A
placebo-controlled, double-blind clinical trial. Ebselen Study Group. Stroke 1998,29, 12–17. [CrossRef] [PubMed]
21.
Tarnopolsky, M.A.; Roy, B.D.; MacDonald, J.R. A randomized, controlled trial of creatine monohydrate in patients with mitochon-
drial cytopathies. Muscle Nerve 1997,20, 1502–1509. [CrossRef]
22.
Hager, K.; Kenklies, M.; McAfoose, J.; Engel, J.; Munch, G. Alpha-lipoic acid as a new treatment option for Alzheimer’s disease—A
48 months follow-up analysis. J. Neural Transm. Suppl. 2007,72, 189–193. [CrossRef]
23.
Chahbouni, M.; Escames, G.; Venegas, C.; Sevilla, B.; Garcia, J.A.; Lopez, L.C.; Munoz-Hoyos, A.; Molina-Carballo, A.; Acuna-
Castroviejo, D. Melatonin treatment normalizes plasma pro-inflammatory cytokines and nitrosative/oxidative stress in patients
suffering from Duchenne muscular dystrophy. J. Pineal Res. 2010,48, 282–289. [CrossRef]
24.
Weishaupt, J.H.; Bartels, C.; Polking, E.; Dietrich, J.; Rohde, G.; Poeggeler, B.; Mertens, N.; Sperling, S.; Bohn, M.; Huther, G.; et al.
Reduced oxidative damage in ALS by high-dose enteral melatonin treatment. J. Pineal Res. 2006,41, 313–323. [CrossRef]
25.
Koga, Y.; Akita, Y.; Nishioka, J.; Yatsuga, S.; Povalko, N.; Tanabe, Y.; Fujimoto, S.; Matsuishi, T. L-arginine improves the symptoms
of strokelike episodes in MELAS. Neurology 2005,64, 710–712. [CrossRef]
26.
Koga, Y.; Ishibashi, M.; Ueki, I.; Yatsuga, S.; Fukiyama, R.; Akita, Y.; Matsuishi, T. Effects of L-arginine on the acute phase of
strokes in three patients with MELAS. Neurology 2002,58, 827–828. [CrossRef]
27.
Ohsawa, Y.; Hagiwara, H.; Nishimatsu, S.I.; Hirakawa, A.; Kamimura, N.; Ohtsubo, H.; Fukai, Y.; Murakami, T.; Koga, Y.; Goto,
Y.I.; et al. Taurine supplementation for prevention of stroke-like episodes in MELAS: A multicentre, open-label, 52-week phase III
trial. J. Neurol. Neurosurg. Psychiatry 2019,90, 529–536. [CrossRef] [PubMed]
28.
Rikimaru, M.; Ohsawa, Y.; Wolf, A.M.; Nishimaki, K.; Ichimiya, H.; Kamimura, N.; Nishimatsu, S.; Ohta, S.; Sunada, Y. Taurine
ameliorates impaired the mitochondrial function and prevents stroke-like episodes in patients with MELAS. Intern. Med.
2012
,51,
3351–3357. [CrossRef] [PubMed]
29.
Azuma, J.; Sawamura, A.; Awata, N.; Ohta, H.; Hamaguchi, T.; Harada, H.; Takihara, K.; Hasegawa, H.; Yamagami, T.; Ishiyama,
T.; et al. Therapeutic effect of taurine in congestive heart failure: A double-blind crossover trial. Clin. Cardiol.
1985
,8, 276–282.
[CrossRef] [PubMed]
30.
Azuma, J.; Hasegawa, H.; Sawamura, A.; Awata, N.; Ogura, K.; Harada, H.; Yamamura, Y.; Kishimoto, S. Therapy of congestive
heart failure with orally administered taurine. Clin. Ther. 1983,5, 398–408.
31.
Beyranvand, M.R.; Khalafi, M.K.; Roshan, V.D.; Choobineh, S.; Parsa, S.A.; Piranfar, M.A. Effect of taurine supplementation on
exercise capacity of patients with heart failure. J. Cardiol. 2011,57, 333–337. [CrossRef] [PubMed]
32.
Jacobsen, J.G.; Smith, L.H. Biochemistry and physiology of taurine and taurine derivatives. Physiol. Rev.
1968
,48, 424–511.
[CrossRef]
33.
Detmer, S.A.; Chan, D.C. Functions and dysfunctions of mitochondrial dynamics. Nat. Rev. Mol. Cell Biol.
2007
,8, 870–879.
[CrossRef]
34.
Murphy, E.; Ardehali, H.; Balaban, R.S.; DiLisa, F.; Dorn, G.W., 2nd; Kitsis, R.N.; Otsu, K.; Ping, P.; Rizzuto, R.; Sack, M.N.; et al.
Mitochondrial Function, Biology, and Role in Disease: A Scientific Statement From the American Heart Association. Circ. Res.
2016,118, 1960–1991. [CrossRef] [PubMed]
35.
Herst, P.M.; Rowe, M.R.; Carson, G.M.; Berridge, M.V. Functional Mitochondria in Health and Disease. Front. Endocrinol.
2017
,8,
296. [CrossRef] [PubMed]
36.
Romero-Garcia, S.; Prado-Garcia, H. Mitochondrial calcium: Transport and modulation of cellular processes in homeostasis and
cancer (Review). Int. J. Oncol. 2019,54, 1155–1167. [CrossRef] [PubMed]
37. Tait, S.W.; Green, D.R. Mitochondria and cell signalling. J. Cell Sci. 2012,125, 807–815. [CrossRef]
38. Kuhlbrandt, W. Structure and function of mitochondrial membrane protein complexes. BMC Biol. 2015,13, 89. [CrossRef]
39. Alexeyev, M.F.; Ledoux, S.P.; Wilson, G.L. Mitochondrial DNA and aging. Clin. Sci. 2004,107, 355–364. [CrossRef]
40.
Xing, G.; Chen, Z.; Cao, X. Mitochondrial rRNA and tRNA and hearing function. Cell Res.
2007
,17, 227–239. [CrossRef] [PubMed]
41.
Zhao, R.Z.; Jiang, S.; Zhang, L.; Yu, Z.B. Mitochondrial electron transport chain, ROS generation and uncoupling (Review). Int. J.
Mol. Med. 2019,44, 3–15. [CrossRef]
42. Turrens, J.F. Mitochondrial formation of reactive oxygen species. J. Physiol. 2003,552, 335–344. [CrossRef] [PubMed]
43.
Chen, Q.; Vazquez, E.J.; Moghaddas, S.; Hoppel, C.L.; Lesnefsky, E.J. Production of reactive oxygen species by mitochondria:
Central role of complex III. J. Biol. Chem. 2003,278, 36027–36031. [CrossRef]
Molecules 2021,26, 4913 14 of 21
44.
Hirst, J.; King, M.S.; Pryde, K.R. The production of reactive oxygen species by complex I. Biochem. Soc. Trans.
2008
,36, 976–980.
[CrossRef]
45.
Cho, Y.M.; Kwon, S.; Pak, Y.K.; Seol, H.W.; Choi, Y.M.; Park, D.J.; Park, K.S.; Lee, H.K. Dynamic changes in mitochondrial
biogenesis and antioxidant enzymes during the spontaneous differentiation of human embryonic stem cells. Biochem. Biophys.
Res. Commun. 2006,348, 1472–1478. [CrossRef]
46.
Tormos, K.V.; Anso, E.; Hamanaka, R.B.; Eisenbart, J.; Joseph, J.; Kalyanaraman, B.; Chandel, N.S. Mitochondrial complex III ROS
regulate adipocyte differentiation. Cell Metab. 2011,14, 537–544. [CrossRef]
47.
Chen, Y.; McMillan-Ward, E.; Kong, J.; Israels, S.J.; Gibson, S.B. Mitochondrial electron-transport-chain inhibitors of complexes I
and II induce autophagic cell death mediated by reactive oxygen species. J. Cell Sci. 2007,120, 4155–4166. [CrossRef] [PubMed]
48.
Scherz-Shouval, R.; Shvets, E.; Fass, E.; Shorer, H.; Gil, L.; Elazar, Z. Reactive oxygen species are essential for autophagy and
specifically regulate the activity of Atg4. EMBO J. 2007,26, 1749–1760. [CrossRef]
49.
Nemoto, S.; Takeda, K.; Yu, Z.X.; Ferrans, V.J.; Finkel, T. Role for mitochondrial oxidants as regulators of cellular metabolism. Mol.
Cell. Biol. 2000,20, 7311–7318. [CrossRef]
50.
Liemburg-Apers, D.C.; Willems, P.H.; Koopman, W.J.; Grefte, S. Interactions between mitochondrial reactive oxygen species and
cellular glucose metabolism. Arch. Toxicol. 2015,89, 1209–1226. [CrossRef] [PubMed]
51. West, A.P.; Shadel, G.S.; Ghosh, S. Mitochondria in innate immune responses. Nat. Rev. Immunol. 2011,11, 389–402. [CrossRef]
52.
Andreyev, A.Y.; Kushnareva, Y.E.; Starkova, N.N.; Starkov, A.A. Metabolic ROS Signaling: To Immunity and Beyond. Biochemistry
2020,85, 1650–1667. [CrossRef]
53.
Cui, H.; Kong, Y.; Zhang, H. Oxidative stress, mitochondrial dysfunction, and aging. J. Signal Transduct.
2012
,2012, 646354.
[CrossRef] [PubMed]
54.
Kowalska, M.; Piekut, T.; Prendecki, M.; Sodel, A.; Kozubski, W.; Dorszewska, J. Mitochondrial and Nuclear DNA Oxidative
Damage in Physiological and Pathological Aging. DNA Cell Biol. 2020,39, 1410–1420. [CrossRef]
55.
Cai, Z.; Yan, L.J. Protein Oxidative Modifications: Beneficial Roles in Disease and Health. J. Biochem. Pharmacol. Res.
2013
,1,
15–26. [PubMed]
56. Nystrom, T. Role of oxidative carbonylation in protein quality control and senescence. EMBO J. 2005,24, 1311–1317. [CrossRef]
57.
Ramana, K.V.; Srivastava, S.; Singhal, S.S. Lipid Peroxidation Products in Human Health and Disease 2019. Oxid. Med. Cell.
Longev. 2019,2019, 7147235. [CrossRef]
58. Haines, T.H.; Dencher, N.A. Cardiolipin: A proton trap for oxidative phosphorylation. FEBS Lett. 2002,528, 35–39. [CrossRef]
59.
Houtkooper, R.H.; Vaz, F.M. Cardiolipin, the heart of mitochondrial metabolism. Cell. Mol. Life Sci.
2008
,65, 2493–2506. [CrossRef]
[PubMed]
60.
Osman, C.; Voelker, D.R.; Langer, T. Making heads or tails of phospholipids in mitochondria. J. Cell Biol.
2011
,192, 7–16.
[CrossRef]
61.
Vahaheikkila, M.; Peltomaa, T.; Rog, T.; Vazdar, M.; Poyry, S.; Vattulainen, I. How cardiolipin peroxidation alters the properties of
the inner mitochondrial membrane? Chem. Phys. Lipids 2018,214, 15–23. [CrossRef] [PubMed]
62. Wong-Ekkabut, J.; Xu, Z.; Triampo, W.; Tang, I.M.; Tieleman, D.P.; Monticelli, L. Effect of lipid peroxidation on the properties of
lipid bilayers: A molecular dynamics study. Biophys. J. 2007,93, 4225–4236. [CrossRef]
63.
Oemer, G.; Koch, J.; Wohlfarter, Y.; Alam, M.T.; Lackner, K.; Sailer, S.; Neumann, L.; Lindner, H.H.; Watschinger, K.; Haltmeier, M.;
et al. Phospholipid Acyl Chain Diversity Controls the Tissue-Specific Assembly of Mitochondrial Cardiolipins. Cell Rep.
2020
,30,
4281–4291.e4. [CrossRef] [PubMed]
64.
Paradies, G.; Petrosillo, G.; Pistolese, M.; Di Venosa, N.; Federici, A.; Ruggiero, F.M. Decrease in mitochondrial complex I activity
in ischemic/reperfused rat heart: Involvement of reactive oxygen species and cardiolipin. Circ. Res. 2004,94, 53–59. [CrossRef]
65.
Mileykovskaya, E.; Dowhan, W. Cardiolipin-dependent formation of mitochondrial respiratory supercomplexes. Chem. Phys.
Lipids 2014,179, 42–48. [CrossRef]
66.
Paradies, G.; Petrosillo, G.; Paradies, V.; Ruggiero, F.M. Role of cardiolipin peroxidation and Ca2+ in mitochondrial dysfunction
and disease. Cell Calcium 2009,45, 643–650. [CrossRef] [PubMed]
67.
Raja, V.; Greenberg, M.L. The functions of cardiolipin in cellular metabolism-potential modifiers of the Barth syndrome phenotype.
Chem. Phys. Lipids 2014,179, 49–56. [CrossRef]
68. Orrenius, S.; Zhivotovsky, B. Cardiolipin oxidation sets cytochrome c free. Nat. Chem. Biol. 2005,1, 188–189. [CrossRef]
69.
Li, X.X.; Tsoi, B.; Li, Y.F.; Kurihara, H.; He, R.R. Cardiolipin and its different properties in mitophagy and apoptosis. J. Histochem.
Cytochem. 2015,63, 301–311. [CrossRef]
70.
Manoharan, S.; Kolanjiappan, K.; Suresh, K.; Panjamurthy, K. Lipid peroxidation & antioxidants status in patients with oral
squamous cell carcinoma. Indian J. Med. Res. 2005,122, 529–534. [PubMed]
71.
Valko, M.; Leibfritz, D.; Moncol, J.; Cronin, M.T.; Mazur, M.; Telser, J. Free radicals and antioxidants in normal physiological
functions and human disease. Int. J. Biochem. Cell Biol. 2007,39, 44–84. [CrossRef] [PubMed]
72.
Lechuga-Sancho, A.M.; Gallego-Andujar, D.; Ruiz-Ocana, P.; Visiedo, F.M.; Saez-Benito, A.; Schwarz, M.; Segundo, C.; Mateos,
R.M. Obesity induced alterations in redox homeostasis and oxidative stress are present from an early age. PLoS ONE
2018
,13,
e0191547. [CrossRef] [PubMed]
73.
Scudamore, O.; Ciossek, T. Increased Oxidative Stress Exacerbates alpha-Synuclein Aggregation In Vivo. J. Neuropathol. Exp.
Neurol. 2018,77, 443–453. [CrossRef] [PubMed]
Molecules 2021,26, 4913 15 of 21
74.
Keller, J.N.; Schmitt, F.A.; Scheff, S.W.; Ding, Q.; Chen, Q.; Butterfield, D.A.; Markesbery, W.R. Evidence of increased oxidative
damage in subjects with mild cognitive impairment. Neurology 2005,64, 1152–1156. [CrossRef] [PubMed]
75.
Narula, J.; Pandey, P.; Arbustini, E.; Haider, N.; Narula, N.; Kolodgie, F.D.; Dal Bello, B.; Semigran, M.J.; Bielsa-Masdeu, A.;
Dec, G.W.; et al. Apoptosis in heart failure: Release of cytochrome c from mitochondria and activation of caspase-3 in human
cardiomyopathy. Proc. Natl. Acad. Sci. USA 1999,96, 8144–8149. [CrossRef]
76.
Chen, L.; Gong, Q.; Stice, J.P.; Knowlton, A.A. Mitochondrial OPA1, apoptosis, and heart failure. Cardiovasc. Res.
2009
,84, 91–99.
[CrossRef] [PubMed]
77.
Hanna, M.G.; Nelson, I.P.; Morgan-Hughes, J.A.; Wood, N.W. MELAS: A new disease associated mitochondrial DNA mutation
and evidence for further genetic heterogeneity. J. Neurol. Neurosurg. Psychiatry 1998,65, 512–517. [CrossRef]
78.
Yang, Y.; Zhang, Y.; Liu, X.; Zuo, J.; Wang, K.; Liu, W.; Ge, J. Exogenous taurine attenuates mitochondrial oxidative stress and
endoplasmic reticulum stress in rat cardiomyocytes. Acta Biochim. Biophys. Sin. 2013,45, 359–367. [CrossRef]
79.
Takatani, T.; Takahashi, K.; Uozumi, Y.; Shikata, E.; Yamamoto, Y.; Ito, T.; Matsuda, T.; Schaffer, S.W.; Fujio, Y.; Azuma, J. Taurine
inhibits apoptosis by preventing formation of the Apaf-1/caspase-9 apoptosome. Am. J. Physiol. Cell Physiol.
2004
,287, C949–C953.
[CrossRef]
80.
Niu, X.; Zheng, S.; Liu, H.; Li, S. Protective effects of taurine against inflammation, apoptosis, and oxidative stress in brain injury.
Mol. Med. Rep. 2018,18, 4516–4522. [CrossRef] [PubMed]
81.
Zhang, R.; Wang, X.; Gao, Q.; Jiang, H.; Zhang, S.; Lu, M.; Liu, F.; Xue, X. Taurine Supplementation Reverses Diabetes-Induced
Podocytes Injury via Modulation of the CSE/TRPC6 Axis and Improvement of Mitochondrial Function. Nephron
2020
,144, 84–95.
[CrossRef] [PubMed]
82.
Homma, K.; Toda, E.; Osada, H.; Nagai, N.; Era, T.; Tsubota, K.; Okano, H.; Ozawa, Y. Taurine rescues mitochondria-related
metabolic impairments in the patient-derived induced pluripotent stem cells and epithelial-mesenchymal transition in the retinal
pigment epithelium. Redox Biol. 2021,41, 101921. [CrossRef]
83.
Shetewy, A.; Shimada-Takaura, K.; Warner, D.; Jong, C.J.; Mehdi, A.B.; Alexeyev, M.; Takahashi, K.; Schaffer, S.W. Mitochondrial
defects associated with beta-alanine toxicity: Relevance to hyper-beta-alaninemia. Mol. Cell. Biochem.
2016
,416, 11–22. [CrossRef]
84.
Jong, C.J.; Azuma, J.; Schaffer, S. Mechanism underlying the antioxidant activity of taurine: Prevention of mitochondrial oxidant
production. Amino Acids 2012,42, 2223–2232. [CrossRef]
85.
Ommati, M.M.; Heidari, R.; Ghanbarinejad, V.; Abdoli, N.; Niknahad, H. Taurine Treatment Provides Neuroprotection in a Mouse
Model of Manganism. Biol. Trace Elem. Res. 2019,190, 384–395. [CrossRef] [PubMed]
86.
Thirupathi, A.; Freitas, S.; Sorato, H.R.; Pedroso, G.S.; Effting, P.S.; Damiani, A.P.; Andrade, V.M.; Nesi, R.T.; Gupta, R.C.; Muller,
A.P.; et al. Modulatory effects of taurine on metabolic and oxidative stress parameters in a mice model of muscle overuse.
Nutrition 2018,54, 158–164. [CrossRef]
87.
Oudit, G.Y.; Trivieri, M.G.; Khaper, N.; Husain, T.; Wilson, G.J.; Liu, P.; Sole, M.J.; Backx, P.H. Taurine supplementation reduces
oxidative stress and improves cardiovascular function in an iron-overload murine model. Circulation
2004
,109, 1877–1885.
[CrossRef] [PubMed]
88.
Wang, Q.; Fan, W.; Cai, Y.; Wu, Q.; Mo, L.; Huang, Z.; Huang, H. Protective effects of taurine in traumatic brain injury via
mitochondria and cerebral blood flow. Amino Acids 2016,48, 2169–2177. [CrossRef] [PubMed]
89.
Jamshidzadeh, A.; Heidari, R.; Abasvali, M.; Zarei, M.; Ommati, M.M.; Abdoli, N.; Khodaei, F.; Yeganeh, Y.; Jafari, F.; Zarei,
A.; et al. Taurine treatment preserves brain and liver mitochondrial function in a rat model of fulminant hepatic failure and
hyperammonemia. Biomed. Pharmacother. 2017,86, 514–520. [CrossRef] [PubMed]
90. Huxtable, R.J. Physiological actions of taurine. Physiol. Rev. 1992,72, 101–163. [CrossRef]
91.
Stipanuk, M.H. Role of the liver in regulation of body cysteine and taurine levels: A brief review. Neurochem. Res.
2004
,29,
105–110. [CrossRef]
92. Heird, W.C. Taurine in neonatal nutrition—Revisited. Arch. Dis. Child. Fetal Neonatal Ed. 2004,89, F473–F474. [CrossRef]
93.
Wojcik, O.P.; Koenig, K.L.; Zeleniuch-Jacquotte, A.; Costa, M.; Chen, Y. The potential protective effects of taurine on coronary
heart disease. Atherosclerosis 2010,208, 19–25. [CrossRef]
94.
Yamori, Y.; Taguchi, T.; Hamada, A.; Kunimasa, K.; Mori, H.; Mori, M. Taurine in health and diseases: Consistent evidence from
experimental and epidemiological studies. J. Biomed. Sci. 2010,17 (Suppl. 1), S6. [CrossRef]
95.
Galeano, N.F.; Darling, P.; Lepage, G.; Leroy, C.; Collet, S.; Giguere, R.; Roy, C.C. Taurine supplementation of a premature formula
improves fat absorption in preterm infants. Pediatr. Res. 1987,22, 67–71. [CrossRef]
96. Taurine deficiency in a child on total parenteral nutrition. Nutr. Rev. 1985,43, 81–83. [CrossRef]
97.
Chesney, R.W.; Helms, R.A.; Christensen, M.; Budreau, A.M.; Han, X.; Sturman, J.A. The role of taurine in infant nutrition. Adv.
Exp. Med. Biol. 1998,442, 463–476. [CrossRef] [PubMed]
98.
Lourenco, R.; Camilo, M.E. Taurine: A conditionally essential amino acid in humans? An overview in health and disease. Nutr.
Hosp. 2002,17, 262–270.
99.
Verner, A.; Craig, S.; McGuire, W. Effect of taurine supplementation on growth and development in preterm or low birth weight
infants. Cochrane Database Syst. Rev. 2007,4, CD006072. [CrossRef]
100. Gaull, G.E. Taurine in pediatric nutrition: Review and update. Pediatrics 1989,83, 433–442. [PubMed]
Molecules 2021,26, 4913 16 of 21
101.
Backus, R.C.; Ko, K.S.; Fascetti, A.J.; Kittleson, M.D.; Macdonald, K.A.; Maggs, D.J.; Berg, J.R.; Rogers, Q.R. Low plasma taurine
concentration in Newfoundland dogs is associated with low plasma methionine and cyst(e)ine concentrations and low taurine
synthesis. J. Nutr. 2006,136, 2525–2533. [CrossRef]
102.
Hayes, K.C.; Trautwein, E.A. Taurine deficiency syndrome in cats. Vet. Clin. N. Am. Small Anim. Pract.
1989
,19, 403–413.
[CrossRef]
103.
Novotny, M.J.; Hogan, P.M.; Flannigan, G. Echocardiographic evidence for myocardial failure induced by taurine deficiency in
domestic cats. Can. J. Vet. Res. 1994,58, 6–12.
104.
Pion, P.D.; Kittleson, M.D.; Skiles, M.L.; Rogers, Q.R.; Morris, J.G. Dilated cardiomyopathy associated with taurine deficiency in
the domestic cat: Relationship to diet and myocardial taurine content. Adv. Exp. Med. Biol. 1992,315, 63–73. [CrossRef]
105. Barnett, K.C.; Burger, I.H. Taurine deficiency retinopathy in the cat. J. Small Anim. Pract. 1980,21, 521–534. [CrossRef]
106.
Leon, A.; Levick, W.R.; Sarossy, M.G. Lesion topography and new histological features in feline taurine deficiency retinopathy.
Exp. Eye Res. 1995,61, 731–741. [CrossRef]
107.
Madl, J.E.; McIlnay, T.R.; Powell, C.C.; Gionfriddo, J.R. Depletion of taurine and glutamate from damaged photoreceptors in the
retinas of dogs with primary glaucoma. Am. J. Vet. Res. 2005,66, 791–799. [CrossRef]
108.
Fariello, R.G.; Lloyd, K.G.; Hornykiewicz, O. Cortical and subcortical projected foci in cats: Inhibitory action of taurine. Neurology
1975,25, 1077–1083. [CrossRef] [PubMed]
109.
Sturman, J.A.; Moretz, R.C.; French, J.H.; Wisniewski, H.M. Taurine deficiency in the developing cat: Persistence of the cerebellar
external granule cell layer. J. Neurosci. Res. 1985,13, 405–416. [CrossRef] [PubMed]
110.
Schuller-Levis, G.; Mehta, P.D.; Rudelli, R.; Sturman, J. Immunologic consequences of taurine deficiency in cats. J. Leukoc. Biol.
1990,47, 321–331. [CrossRef] [PubMed]
111.
Dieter, J.A.; Stewart, D.R.; Haggarty, M.A.; Stabenfeldt, G.H.; Lasley, B.L. Pregnancy failure in cats associated with long-term
dietary taurine insufficiency. J. Reprod. Fertil. Suppl. 1993,47, 457–463.
112.
Sturman, J.A.; Gargano, A.D.; Messing, J.M.; Imaki, H. Feline maternal taurine deficiency: Effect on mother and offspring. J. Nutr.
1986,116, 655–667. [CrossRef]
113.
Backus, R.C.; Rogers, Q.R.; Rosenquist, G.L.; Calam, J.; Morris, J.G. Diets causing taurine depletion in cats substantially elevate
postprandial plasma cholecystokinin concentration. J. Nutr. 1995,125, 2650–2657. [CrossRef]
114.
Rabin, B.; Nicolosi, R.J.; Hayes, K.C. Dietary influence on bile acid conjugation in the cat. J. Nutr.
1976
,106, 1241–1246. [CrossRef]
115.
Backus, R.C.; Cohen, G.; Pion, P.D.; Good, K.L.; Rogers, Q.R.; Fascetti, A.J. Taurine deficiency in Newfoundlands fed commercially
available complete and balanced diets. J. Am. Vet. Med. Assoc. 2003,223, 1130–1136. [CrossRef]
116.
Pion, P.D.; Kittleson, M.D.; Thomas, W.P.; Delellis, L.A.; Rogers, Q.R. Response of cats with dilated cardiomyopathy to taurine
supplementation. J. Am. Vet. Med. Assoc. 1992,201, 275–284. [PubMed]
117.
van Gelder, N.M.; Koyama, I.; Jasper, H.H. Taurine treatment of spontaneous chronic epilepsy in a cat. Epilepsia
1977
,18, 45–54.
[CrossRef]
118.
Berson, E.L.; Hayes, K.C.; Rabin, A.R.; Schmidt, S.Y.; Watson, G. Retinal degeneration in cats fed casein. II. Supplementation with
methionine, cysteine, or taurine. Investig. Ophthalmol. 1976,15, 52–58.
119.
Sturman, J.A.; Messing, J.M. Dietary taurine content and feline reproduction and outcome. J. Nutr.
1991
,121, 1195–1203.
[CrossRef] [PubMed]
120. Caine, J.J.; Geracioti, T.D. Taurine, energy drinks, and neuroendocrine effects. Clevel. Clin. J. Med. 2016,83, 895–904. [CrossRef]
121. Higgins, J.P.; Tuttle, T.D.; Higgins, C.L. Energy beverages: Content and safety. Mayo Clin. Proc. 2010,85, 1033–1041. [CrossRef]
122.
Kurtz, J.A.; VanDusseldorp, T.A.; Doyle, J.A.; Otis, J.S. Taurine in sports and exercise. J. Int. Soc. Sports Nutr.
2021
,18, 39.
[CrossRef]
123.
Seidel, U.; Huebbe, P.; Rimbach, G. Taurine: A Regulator of Cellular Redox Homeostasis and Skeletal Muscle Function. Mol. Nutr.
Food Res. 2019,63, e1800569. [CrossRef] [PubMed]
124.
Ghandforoush-Sattari, M.; Mashayekhi, S.; Krishna, C.V.; Thompson, J.P.; Routledge, P.A. Pharmacokinetics of oral taurine in
healthy volunteers. J. Amino Acids 2010,2010, 346237. [CrossRef]
125.
Sturman, J.A.; Hepner, G.W.; Hofmann, A.F.; Thomas, P.J. Metabolism of [35S]taurine in man. J. Nutr.
1975
,105, 1206–1214.
[CrossRef] [PubMed]
126.
Ito, T.; Oishi, S.; Takai, M.; Kimura, Y.; Uozumi, Y.; Fujio, Y.; Schaffer, S.W.; Azuma, J. Cardiac and skeletal muscle abnormality in
taurine transporter-knockout mice. J. Biomed. Sci. 2010,17 (Suppl. 1), S20. [CrossRef] [PubMed]
127.
Warskulat, U.; Flogel, U.; Jacoby, C.; Hartwig, H.G.; Thewissen, M.; Merx, M.W.; Molojavyi, A.; Heller-Stilb, B.; Schrader, J.;
Haussinger, D. Taurine transporter knockout depletes muscle taurine levels and results in severe skeletal muscle impairment but
leaves cardiac function uncompromised. FASEB J. 2004,18, 577–579. [CrossRef]
128.
Garcia-Ayuso, D.; Di Pierdomenico, J.; Valiente-Soriano, F.J.; Martinez-Vacas, A.; Agudo-Barriuso, M.; Vidal-Sanz, M.; Picaud, S.;
Villegas-Perez, M.P. beta-alanine supplementation induces taurine depletion and causes alterations of the retinal nerve fiber layer
and axonal transport by retinal ganglion cells. Exp. Eye Res. 2019,188, 107781. [CrossRef] [PubMed]
129.
Jong, C.J.; Ito, T.; Mozaffari, M.; Azuma, J.; Schaffer, S. Effect of beta-alanine treatment on mitochondrial taurine level and
5-taurinomethyluridine content. J. Biomed. Sci. 2010,17 (Suppl. 1), S25. [CrossRef] [PubMed]
130. Lake, N. Depletion of taurine in the adult rat retina. Neurochem. Res. 1982,7, 1385–1390. [CrossRef]
Molecules 2021,26, 4913 17 of 21
131.
Pasantes-Morales, H.; Quesada, O.; Carabez, A.; Huxtable, R.J. Effects of the taurine transport antagonist, guanidinoethane
sulfonate, and beta-alanine on the morphology of rat retina. J. Neurosci. Res. 1983,9, 135–143. [CrossRef]
132.
Han, X.; Patters, A.B.; Ito, T.; Azuma, J.; Schaffer, S.W.; Chesney, R.W. Knockout of the TauT gene predisposes C57BL/6 mice to
streptozotocin-induced diabetic nephropathy. PLoS ONE 2015,10, e0117718. [CrossRef]
133.
Rascher, K.; Servos, G.; Berthold, G.; Hartwig, H.G.; Warskulat, U.; Heller-Stilb, B.; Haussinger, D. Light deprivation slows but
does not prevent the loss of photoreceptors in taurine transporter knockout mice. Vision Res.
2004
,44, 2091–2100. [CrossRef]
[PubMed]
134.
Warskulat, U.; Borsch, E.; Reinehr, R.; Heller-Stilb, B.; Monnighoff, I.; Buchczyk, D.; Donner, M.; Flogel, U.; Kappert, G.; Soboll, S.;
et al. Chronic liver disease is triggered by taurine transporter knockout in the mouse. FASEB J. 2006,20, 574–576. [CrossRef]
135.
Jong, C.J.; Ito, T.; Prentice, H.; Wu, J.Y.; Schaffer, S.W. Role of Mitochondria and Endoplasmic Reticulum in Taurine-Deficiency-
Mediated Apoptosis. Nutrients 2017,9, 795. [CrossRef] [PubMed]
136.
Jong, C.J.; Ito, T.; Azuma, J.; Schaffer, S. Taurine Depletion Decreases GRP78 Expression and Downregulates Perk-Dependent
Activation of the Unfolded Protein Response. Adv. Exp. Med. Biol. 2015,803, 571–579. [CrossRef] [PubMed]
137.
Jong, C.J.; Ito, T.; Schaffer, S.W. The ubiquitin-proteasome system and autophagy are defective in the taurine-deficient heart.
Amino Acids 2015,47, 2609–2622. [CrossRef]
138.
Ito, T.; Yoshikawa, N.; Inui, T.; Miyazaki, N.; Schaffer, S.W.; Azuma, J. Tissue depletion of taurine accelerates skeletal muscle
senescence and leads to early death in mice. PLoS ONE 2014,9, e107409. [CrossRef]
139.
Azari, J.; Bahl, J.; Huxtable, R. Guanidinoethyl sulfonate and other inhibitors of the taurine transporting system in the heart. Proc.
West. Pharmacol. Soc. 1979,22, 389–393. [PubMed]
140.
Huxtable, R.J.; Laird, H.E., 2nd; Lippincott, S.E. The transport of taurine in the heart and the rapid depletion of tissue taurine
content by guanidinoethyl sulfonate. J. Pharmacol. Exp. Ther. 1979,211, 465–471.
141.
Pansani, M.C.; Azevedo, P.S.; Rafacho, B.P.; Minicucci, M.F.; Chiuso-Minicucci, F.; Zorzella-Pezavento, S.G.; Marchini, J.S.;
Padovan, G.J.; Fernandes, A.A.; Matsubara, B.B.; et al. Atrophic cardiac remodeling induced by taurine deficiency in Wistar rats.
PLoS ONE 2012,7, e41439. [CrossRef]
142. Parildar, H.; Dogru-Abbasoglu, S.; Mehmetcik, G.; Ozdemirler, G.; Kocak-Toker, N.; Uysal, M. Lipid peroxidation potential and
antioxidants in the heart tissue of beta-alanine- or taurine-treated old rats. J. Nutr. Sci. Vitaminol. 2008,54, 61–65. [CrossRef]
143.
Jong, C.J.; Azuma, J.; Schaffer, S.W. Role of mitochondrial permeability transition in taurine deficiency-induced apoptosis. Exp.
Clin. Cardiol. 2011,16, 125–128. [PubMed]
144.
Schaffer, S.W.; Ballard-Croft, C.; Azuma, J.; Takahashi, K.; Kakhniashvili, D.G.; Jenkins, T.E. Shape and size changes induced by
taurine depletion in neonatal cardiomyocytes. Amino Acids 1998,15, 135–142. [CrossRef]
145.
Suzuki, T.; Suzuki, T.; Wada, T.; Saigo, K.; Watanabe, K. Taurine as a constituent of mitochondrial tRNAs: New insights into the
functions of taurine and human mitochondrial diseases. EMBO J. 2002,21, 6581–6589. [CrossRef] [PubMed]
146.
Wada, T.; Shimazaki, T.; Nakagawa, S.; Otuki, T.; Kurata, S.; Suzuki, T.; Watanabe, K.; Saigo, K. Chemical synthesis of novel
taurine-containing uridine derivatives. Nucleic Acids Res. Suppl. 2002,2, 11–12. [CrossRef] [PubMed]
147.
Fakruddin, M.; Wei, F.Y.; Suzuki, T.; Asano, K.; Kaieda, T.; Omori, A.; Izumi, R.; Fujimura, A.; Kaitsuka, T.; Miyata, K.; et al.
Defective Mitochondrial tRNA Taurine Modification Activates Global Proteostress and Leads to Mitochondrial Disease. Cell Rep.
2018,22, 482–496. [CrossRef] [PubMed]
148.
Kirino, Y.; Goto, Y.; Campos, Y.; Arenas, J.; Suzuki, T. Specific correlation between the wobble modification deficiency in mutant
tRNAs and the clinical features of a human mitochondrial disease. Proc. Natl. Acad. Sci. USA
2005
,102, 7127–7132. [CrossRef]
[PubMed]
149.
Kirino, Y.; Yasukawa, T.; Ohta, S.; Akira, S.; Ishihara, K.; Watanabe, K.; Suzuki, T. Codon-specific translational defect caused by
a wobble modification deficiency in mutant tRNA from a human mitochondrial disease. Proc. Natl. Acad. Sci. USA
2004
,101,
15070–15075. [CrossRef]
150.
Asano, K.; Suzuki, T.; Saito, A.; Wei, F.Y.; Ikeuchi, Y.; Numata, T.; Tanaka, R.; Yamane, Y.; Yamamoto, T.; Goto, T.; et al. Metabolic
and chemical regulation of tRNA modification associated with taurine deficiency and human disease. Nucleic Acids Res.
2018
,46,
1565–1583. [CrossRef]
151.
Aruoma, O.I.; Halliwell, B.; Hoey, B.M.; Butler, J. The antioxidant action of taurine, hypotaurine and their metabolic precursors.
Biochem. J. 1988,256, 251–255. [CrossRef] [PubMed]
152.
Li, J.X.; Pang, Y.Z.; Tang, C.S.; Li, Z.Q. Protective effect of taurine on hypochlorous acid toxicity to nuclear nucleoside triphos-
phatase in isolated nuclei from rat liver. World J. Gastroenterol. 2004,10, 694–698. [CrossRef] [PubMed]
153.
Cheong, S.H.; Lee, D.S. Taurine Chloramine Prevents Neuronal HT22 Cell Damage Through Nrf2-Related Heme Oxygenase-1.
Adv. Exp. Med. Biol. 2017,975 Pt 1, 145–157. [CrossRef]
154.
Kang, I.S.; Kim, C. Taurine chloramine administered
in vivo
increases NRF2-regulated antioxidant enzyme expression in murine
peritoneal macrophages. Adv. Exp. Med. Biol. 2013,775, 259–267. [CrossRef]
155.
Kim, C.; Cha, Y.N. Taurine chloramine produced from taurine under inflammation provides anti-inflammatory and cytoprotective
effects. Amino Acids 2014,46, 89–100. [CrossRef] [PubMed]
156.
Higuchi, M.; Celino, F.T.; Shimizu-Yamaguchi, S.; Miura, C.; Miura, T. Taurine plays an important role in the protection of
spermatogonia from oxidative stress. Amino Acids 2012,43, 2359–2369. [CrossRef]
Molecules 2021,26, 4913 18 of 21
157.
Okado-Matsumoto, A.; Fridovich, I. Subcellular distribution of superoxide dismutases (SOD) in rat liver: Cu,Zn-SOD in
mitochondria. J. Biol. Chem. 2001,276, 38388–38393. [CrossRef] [PubMed]
158.
Sturtz, L.A.; Diekert, K.; Jensen, L.T.; Lill, R.; Culotta, V.C. A fraction of yeast Cu,Zn-superoxide dismutase and its metallochaper-
one, CCS, localize to the intermembrane space of mitochondria. A physiological role for SOD1 in guarding against mitochondrial
oxidative damage. J. Biol. Chem. 2001,276, 38084–38089. [CrossRef]
159.
Tabassum, H.; Rehman, H.; Banerjee, B.D.; Raisuddin, S.; Parvez, S. Attenuation of tamoxifen-induced hepatotoxicity by taurine
in mice. Clin. Chim. Acta 2006,370, 129–136. [CrossRef]
160.
Pasantes-Morales, H.; Cruz, C. Taurine and hypotaurine inhibit light-induced lipid peroxidation and protect rod outer segment
structure. Brain Res. 1985,330, 154–157. [CrossRef]
161.
Pasantes-Morales, H.; Cruz, C. Taurine: A physiological stabilizer of photoreceptor membranes. Prog. Clin. Biol. Res.
1985
,179,
371–381. [PubMed]
162.
Pasantes-Morales, H.; Wright, C.E.; Gaull, G.E. Taurine protection of lymphoblastoid cells from iron-ascorbate induced damage.
Biochem. Pharmacol. 1985,34, 2205–2207. [CrossRef]
163.
Hansen, S.H.; Andersen, M.L.; Cornett, C.; Gradinaru, R.; Grunnet, N. A role for taurine in mitochondrial function. J. Biomed. Sci.
2010,17 (Suppl. 1), S23. [CrossRef]
164.
El Idrissi, A. Taurine increases mitochondrial buffering of calcium: Role in neuroprotection. Amino Acids
2008
,34, 321–328.
[CrossRef]
165.
El Idrissi, A.; Trenkner, E. Growth factors and taurine protect against excitotoxicity by stabilizing calcium homeostasis and energy
metabolism. J. Neurosci. 1999,19, 9459–9468. [CrossRef]
166.
El Idrissi, A.; Trenkner, E. Taurine regulates mitochondrial calcium homeostasis. Adv. Exp. Med. Biol.
2003
,526, 527–536.
[CrossRef]
167.
El Idrissi, A.; Trenkner, E. Taurine as a modulator of excitatory and inhibitory neurotransmission. Neurochem. Res.
2004
,29,
189–197. [CrossRef]
168.
Trenkner, E.; el Idrissi, A.; Harris, C. Balanced interaction of growth factors and taurine regulate energy metabolism, neuronal
survival, and function of cultured mouse cerebellar cells under depolarizing conditions. Adv. Exp. Med. Biol.
1996
,403, 507–517.
[CrossRef]
169.
Bkaily, G.; Jaalouk, D.; Sader, S.; Shbaklo, H.; Pothier, P.; Jacques, D.; D’Orleans-Juste, P.; Cragoe, E.J., Jr.; Bose, R. Taurine indirectly
increases [Ca]iby inducing Ca2+ influx through the Na(+)-Ca2+ exchanger. Mol. Cell Biochem. 1998,188, 187–197. [CrossRef]
170.
Schaffer, S.; Solodushko, V.; Azuma, J. Taurine-deficient cardiomyopathy: Role of phospholipids, calcium and osmotic stress. Adv.
Exp. Med. Biol. 2000,483, 57–69. [CrossRef]
171.
Schaffer, S.W.; Punna, S.; Duan, J.; Harada, H.; Hamaguchi, T.; Azuma, J. Mechanism underlying physiological modulation of
myocardial contraction by taurine. Adv. Exp. Med. Biol. 1992,315, 193–198. [CrossRef]
172.
Takahashi, K.; Harada, H.; Schaffer, S.W.; Azuma, J. Effect of taurine on intracellular calcium dynamics of cultured myocardial
cells during the calcium paradox. Adv. Exp. Med. Biol. 1992,315, 153–161. [CrossRef] [PubMed]
173.
Steele, D.S.; Smith, G.L.; Miller, D.J. The effects of taurine on Ca2+ uptake by the sarcoplasmic reticulum and Ca2+ sensitivity of
chemically skinned rat heart. J. Physiol. 1990,422, 499–511. [CrossRef]
174.
Galler, S.; Hutzler, C.; Haller, T. Effects of taurine on Ca2(+)-dependent force development of skinned muscle fibre preparations. J.
Exp. Biol. 1990,152, 255–264. [CrossRef]
175.
Griffiths, E.J.; Rutter, G.A. Mitochondrial calcium as a key regulator of mitochondrial ATP production in mammalian cells.
Biochim. Biophys. Acta 2009,1787, 1324–1333. [CrossRef]
176.
Chen, M.; Guerrero, A.D.; Huang, L.; Shabier, Z.; Pan, M.; Tan, T.H.; Wang, J. Caspase-9-induced mitochondrial disruption
through cleavage of anti-apoptotic BCL-2 family members. J. Biol. Chem. 2007,282, 33888–33895. [CrossRef] [PubMed]
177.
Leon, R.; Wu, H.; Jin, Y.; Wei, J.; Buddhala, C.; Prentice, H.; Wu, J.Y. Protective function of taurine in glutamate-induced apoptosis
in cultured neurons. J. Neurosci. Res. 2009,87, 1185–1194. [CrossRef]
178.
Menzie, J.; Prentice, H.; Wu, J.Y. Neuroprotective Mechanisms of Taurine against Ischemic Stroke. Brain Sci.
2013
,3, 877–907.
[CrossRef]
179. Wu, J.Y.; Prentice, H. Role of taurine in the central nervous system. J. Biomed. Sci. 2010,17 (Suppl. 1), S1. [CrossRef]
180.
Taranukhin, A.G.; Taranukhina, E.Y.; Saransaari, P.; Podkletnova, I.M.; Pelto-Huikko, M.; Oja, S.S. Neuroprotection by taurine in
ethanol-induced apoptosis in the developing cerebellum. J. Biomed. Sci. 2010,17 (Suppl. 1), S12. [CrossRef]
181.
Azuma, J.; Sawamura, A.; Awata, N. Usefulness of taurine in chronic congestive heart failure and its prospective application. Jpn.
Circ. J. 1992,56, 95–99. [CrossRef]
182.
Di Lorenzo, A.; Iannuzzo, G.; Parlato, A.; Cuomo, G.; Testa, C.; Coppola, M.; D’Ambrosio, G.; Oliviero, D.A.; Sarullo, S.; Vitale, G.;
et al. Clinical Evidence for Q10 Coenzyme Supplementation in Heart Failure: From Energetics to Functional Improvement. J.
Clin. Med. 2020,9, 1266. [CrossRef]
183.
Jafari, M.; Mousavi, S.M.; Asgharzadeh, A.; Yazdani, N. Coenzyme Q10 in the treatment of heart failure: A systematic review of
systematic reviews. Indian Heart J. 2018,70 (Suppl. 1), S111–S117. [CrossRef]
184.
Sharma, A.; Fonarow, G.C.; Butler, J.; Ezekowitz, J.A.; Felker, G.M. Coenzyme Q10 and Heart Failure: A State-of-the-Art Review.
Circ. Heart Fail. 2016,9, e002639. [CrossRef]
Molecules 2021,26, 4913 19 of 21
185.
Doenst, T.; Nguyen, T.D.; Abel, E.D. Cardiac metabolism in heart failure: Implications beyond ATP production. Circ. Res.
2013
,
113, 709–724. [CrossRef] [PubMed]
186.
Sheeran, F.L.; Pepe, S. Energy deficiency in the failing heart: Linking increased reactive oxygen species and disruption of oxidative
phosphorylation rate. Biochim. Biophys. Acta 2006,1757, 543–552. [CrossRef]
187.
Militante, J.D.; Lombardini, J.B. Treatment of hypertension with oral taurine: Experimental and clinical studies. Amino Acids
2002
,
23, 381–393. [CrossRef] [PubMed]
188.
Sun, Q.; Wang, B.; Li, Y.; Sun, F.; Li, P.; Xia, W.; Zhou, X.; Li, Q.; Wang, X.; Chen, J.; et al. Taurine Supplementation Lowers
Blood Pressure and Improves Vascular Function in Prehypertension: Randomized, Double-Blind, Placebo-Controlled Study.
Hypertension 2016,67, 541–549. [CrossRef]
189.
Sagara, M.; Murakami, S.; Mizushima, S.; Liu, L.; Mori, M.; Ikeda, K.; Nara, Y.; Yamori, Y. Taurine in 24-h Urine Samples Is
Inversely Related to Cardiovascular Risks of Middle Aged Subjects in 50 Populations of the World. Adv. Exp. Med. Biol.
2015
,803,
623–636. [CrossRef] [PubMed]
190.
Yamori, Y.; Liu, L.; Mori, M.; Sagara, M.; Murakami, S.; Nara, Y.; Mizushima, S. Taurine as the nutritional factor for the longevity
of the Japanese revealed by a world-wide epidemiological survey. Adv. Exp. Med. Biol. 2009,643, 13–25. [CrossRef]
191.
Yamori, Y.; Murakami, S.; Ikeda, K.; Nara, Y. Fish and lifestyle-related disease prevention: Experimental and epidemiological
evidence for anti-atherogenic potential of taurine. Clin. Exp. Pharmacol. Physiol. 2004,31 (Suppl. 2), S20–S23. [CrossRef]
192.
Yamori, Y.; Taguchi, T.; Mori, H.; Mori, M. Low cardiovascular risks in the middle aged males and females excreting greater
24-hour urinary taurine and magnesium in 41 WHO-CARDIAC study populations in the world. J. Biomed. Sci.
2010
,17 (Suppl.
1), S21. [CrossRef] [PubMed]
193.
Adedara, I.A.; Alake, S.E.; Olajide, L.O.; Adeyemo, M.O.; Ajibade, T.O.; Farombi, E.O. Taurine Ameliorates Thyroid Hypofunction
and Renal Injury in L-NAME-Induced Hypertensive Rats. Drug Res. 2019,69, 83–92. [CrossRef]
194.
Ibrahim, M.A.; Eraqi, M.M.; Alfaiz, F.A. Therapeutic role of taurine as antioxidant in reducing hypertension risks in rats. Heliyon
2020,6, e03209. [CrossRef] [PubMed]
195.
Rahman, M.M.; Park, H.M.; Kim, S.J.; Go, H.K.; Kim, G.B.; Hong, C.U.; Lee, Y.U.; Kim, S.Z.; Kim, J.S.; Kang, H.S. Taurine prevents
hypertension and increases exercise capacity in rats with fructose-induced hypertension. Am. J. Hypertens.
2011
,24, 574–581.
[CrossRef]
196.
Zaric, B.L.; Radovanovic, J.N.; Gluvic, Z.; Stewart, A.J.; Essack, M.; Motwalli, O.; Gojobori, T.; Isenovic, E.R. Atherosclerosis
Linked to Aberrant Amino Acid Metabolism and Immunosuppressive Amino Acid Catabolizing Enzymes. Front. Immunol.
2020
,
11, 551758. [CrossRef] [PubMed]
197.
Dikalov, S.I.; Ungvari, Z. Role of mitochondrial oxidative stress in hypertension. Am. J. Physiol. Heart Circ. Physiol.
2013
,305,
H1417–H1427. [CrossRef] [PubMed]
198.
Esmaeili, F.; Maleki, V.; Kheirouri, S.; Alizadeh, M. The Effects of Taurine Supplementation on Metabolic Profiles, Pentosidine,
Soluble Receptor of Advanced Glycation End Products and Methylglyoxal in Adults With Type 2 Diabetes: A Randomized,
Double-Blind, Placebo-Controlled Trial. Can. J. Diabetes 2021,45, 39–46. [CrossRef]
199.
Maleki, V.; Alizadeh, M.; Esmaeili, F.; Mahdavi, R. The effects of taurine supplementation on glycemic control and serum lipid
profile in patients with type 2 diabetes: A randomized, double-blind, placebo-controlled trial. Amino Acids
2020
,52, 905–914.
[CrossRef]
200.
Maleki, V.; Mahdavi, R.; Hajizadeh-Sharafabad, F.; Alizadeh, M. The effects of taurine supplementation on oxidative stress indices
and inflammation biomarkers in patients with type 2 diabetes: A randomized, double-blind, placebo-controlled trial. Diabetol.
Metab. Syndr. 2020,12, 9. [CrossRef]
201.
De Luca, G.; Calpona, P.R.; Caponetti, A.; Romano, G.; Di Benedetto, A.; Cucinotta, D.; Di Giorgio, R.M. Taurine and osmoregula-
tion: Platelet taurine content, uptake, and release in type 2 diabetic patients. Metabolism 2001,50, 60–64. [CrossRef]
202.
Franconi, F.; Bennardini, F.; Mattana, A.; Miceli, M.; Ciuti, M.; Mian, M.; Gironi, A.; Anichini, R.; Seghieri, G. Plasma and platelet
taurine are reduced in subjects with insulin-dependent diabetes mellitus: Effects of taurine supplementation. Am. J. Clin. Nutr.
1995,61, 1115–1119. [CrossRef] [PubMed]
203.
Sak, D.; Erdenen, F.; Muderrisoglu, C.; Altunoglu, E.; Sozer, V.; Gungel, H.; Guler, P.A.; Sak, T.; Uzun, H. The Relationship
between Plasma Taurine Levels and Diabetic Complications in Patients with Type 2 Diabetes Mellitus. Biomolecules
2019
,9, 96.
[CrossRef]
204.
Trautwein, E.A.; Hayes, K.C. Plasma and whole blood taurine concentrations respond differently to taurine supplementation
(humans) and depletion (cats). Z. Ernahrungswiss. 1995,34, 137–142. [CrossRef] [PubMed]
205.
Haythorne, E.; Rohm, M.; van de Bunt, M.; Brereton, M.F.; Tarasov, A.I.; Blacker, T.S.; Sachse, G.; Silva Dos Santos, M.; Terron
Exposito, R.; Davis, S.; et al. Diabetes causes marked inhibition of mitochondrial metabolism in pancreatic beta-cells. Nat.
Commun. 2019,10, 2474. [CrossRef]
206.
Hyeon, J.S.; Jung, Y.; Lee, G.; Ha, H.; Hwang, G.S. Urinary Metabolomic Profiling in Streptozotocin-Induced Diabetic Mice after
Treatment with Losartan. Int. J. Mol. Sci. 2020,21, 8969. [CrossRef] [PubMed]
207.
Trachtman, H.; Futterweit, S.; Maesaka, J.; Ma, C.; Valderrama, E.; Fuchs, A.; Tarectecan, A.A.; Rao, P.S.; Sturman, J.A.; Boles,
T.H.; et al. Taurine ameliorates chronic streptozocin-induced diabetic nephropathy in rats. Am. J. Physiol.
1995
,269, F429–F438.
[CrossRef] [PubMed]
Molecules 2021,26, 4913 20 of 21
208.
Evans, J.L.; Goldfine, I.D.; Maddux, B.A.; Grodsky, G.M. Oxidative stress and stress-activated signaling pathways: A unifying
hypothesis of type 2 diabetes. Endocr. Rev. 2002,23, 599–622. [CrossRef] [PubMed]
209.
Haber, C.A.; Lam, T.K.; Yu, Z.; Gupta, N.; Goh, T.; Bogdanovic, E.; Giacca, A.; Fantus, I.G. N-acetylcysteine and taurine prevent
hyperglycemia-induced insulin resistance
in vivo
: Possible role of oxidative stress. Am. J. Physiol. Endocrinol. Metab.
2003
,285,
E744–E753. [CrossRef]
210.
Han, J.; Bae, J.H.; Kim, S.Y.; Lee, H.Y.; Jang, B.C.; Lee, I.K.; Cho, C.H.; Lim, J.G.; Suh, S.I.; Kwon, T.K.; et al. Taurine increases
glucose sensitivity of UCP2-overexpressing beta-cells by ameliorating mitochondrial metabolism. Am. J. Physiol. Endocrinol.
Metab. 2004,287, E1008–E1018. [CrossRef]
211.
Ito, T.; Schaffer, S.W.; Azuma, J. The potential usefulness of taurine on diabetes mellitus and its complications. Amino Acids
2012
,
42, 1529–1539. [CrossRef]
212.
Kim, K.S.; Oh, D.H.; Kim, J.Y.; Lee, B.G.; You, J.S.; Chang, K.J.; Chung, H.J.; Yoo, M.C.; Yang, H.I.; Kang, J.H.; et al. Taurine
ameliorates hyperglycemia and dyslipidemia by reducing insulin resistance and leptin level in Otsuka Long-Evans Tokushima
fatty (OLETF) rats with long-term diabetes. Exp. Mol. Med. 2012,44, 665–673. [CrossRef]
213.
Chauncey, K.B.; Tenner, T.E., Jr.; Lombardini, J.B.; Jones, B.G.; Brooks, M.L.; Warner, R.D.; Davis, R.L.; Ragain, R.M. The effect of
taurine supplementation on patients with type 2 diabetes mellitus. Adv. Exp. Med. Biol. 2003,526, 91–96. [CrossRef]
214.
Nakamura, T.; Ushiyama, C.; Suzuki, S.; Shimada, N.; Ohmuro, H.; Ebihara, I.; Koide, H. Effects of taurine and vitamin E on
microalbuminuria, plasma metalloproteinase-9, and serum type IV collagen concentrations in patients with diabetic nephropathy.
Nephron 1999,83, 361–362. [CrossRef]
215.
Rosa, F.T.; Freitas, E.C.; Deminice, R.; Jordao, A.A.; Marchini, J.S. Oxidative stress and inflammation in obesity after taurine
supplementation: A double-blind, placebo-controlled study. Eur. J. Nutr. 2014,53, 823–830. [CrossRef] [PubMed]
216.
Mizushima, S.; Nara, Y.; Sawamura, M.; Yamori, Y. Effects of oral taurine supplementation on lipids and sympathetic nerve tone.
Adv. Exp. Med. Biol. 1996,403, 615–622. [CrossRef] [PubMed]
217.
De Carvalho, F.G.; Brandao, C.F.C.; Batitucci, G.; Souza, A.O.; Ferrari, G.D.; Alberici, L.C.; Munoz, V.R.; Pauli, J.R.; De Moura, L.P.;
Ropelle, E.R.; et al. Taurine supplementation associated with exercise increases mitochondrial activity and fatty acid oxidation
gene expression in the subcutaneous white adipose tissue of obese women. Clin. Nutr.
2021
,40, 2180–2187. [CrossRef] [PubMed]
218.
Yamori, Y. Preliminary report of cardiac study: Cross-sectional multicenter study on dietary factors of cardiovascular diseases.
CARDIAC Study Group. Clin. Exp. Hypertens. A 1989,11, 957–972. [CrossRef]
219.
Harada, H.; Tsujino, T.; Watari, Y.; Nonaka, H.; Emoto, N.; Yokoyama, M. Oral taurine supplementation prevents fructose-induced
hypertension in rats. Heart Vessels 2004,19, 132–136. [CrossRef]
220.
Harada, N.; Ninomiya, C.; Osako, Y.; Morishima, M.; Mawatari, K.; Takahashi, A.; Nakaya, Y. Taurine alters respiratory gas
exchange and nutrient metabolism in type 2 diabetic rats. Obes. Res. 2004,12, 1077–1084. [CrossRef] [PubMed]
221.
Nandhini, A.T.; Thirunavukkarasu, V.; Ravichandran, M.K.; Anuradha, C.V. Effect of taurine on biomarkers of oxidative stress in
tissues of fructose-fed insulin-resistant rats. Singap. Med. J. 2005,46, 82–87.
222.
Nardelli, T.R.; Ribeiro, R.A.; Balbo, S.L.; Vanzela, E.C.; Carneiro, E.M.; Boschero, A.C.; Bonfleur, M.L. Taurine prevents fat
deposition and ameliorates plasma lipid profile in monosodium glutamate-obese rats. Amino Acids
2011
,41, 901–908. [CrossRef]
223.
Tsuboyama-Kasaoka, N.; Shozawa, C.; Sano, K.; Kamei, Y.; Kasaoka, S.; Hosokawa, Y.; Ezaki, O. Taurine (2-aminoethanesulfonic
acid) deficiency creates a vicious circle promoting obesity. Endocrinology 2006,147, 3276–3284. [CrossRef]
224.
Fukuda, M.; Nagao, Y. Dynamic derangement in amino acid profile during and after a stroke-like episode in adult-onset
mitochondrial disease: A case report. J. Med. Case Rep. 2019,13, 313. [CrossRef]
225.
Che, Y.; Hou, L.; Sun, F.; Zhang, C.; Liu, X.; Piao, F.; Zhang, D.; Li, H.; Wang, Q. Taurine protects dopaminergic neurons in a
mouse Parkinson’s disease model through inhibition of microglial M1 polarization. Cell Death Dis. 2018,9, 435. [CrossRef]
226.
Hou, L.; Che, Y.; Sun, F.; Wang, Q. Taurine protects noradrenergic locus coeruleus neurons in a mouse Parkinson’s disease model
by inhibiting microglial M1 polarization. Amino Acids 2018,50, 547–556. [CrossRef] [PubMed]
227.
Jang, H.; Lee, S.; Choi, S.L.; Kim, H.Y.; Baek, S.; Kim, Y. Taurine Directly Binds to Oligomeric Amyloid-beta and Recovers
Cognitive Deficits in Alzheimer Model Mice. Adv. Exp. Med. Biol. 2017,975 Pt 1, 233–241. [CrossRef]
228.
Kim, H.Y.; Kim, H.V.; Yoon, J.H.; Kang, B.R.; Cho, S.M.; Lee, S.; Kim, J.Y.; Kim, J.W.; Cho, Y.; Woo, J.; et al. Taurine in drinking
water recovers learning and memory in the adult APP/PS1 mouse model of Alzheimer’s disease. Sci. Rep.
2014
,4, 7467.
[CrossRef] [PubMed]
229.
Oh, S.J.; Lee, H.J.; Jeong, Y.J.; Nam, K.R.; Kang, K.J.; Han, S.J.; Lee, K.C.; Lee, Y.J.; Choi, J.Y. Evaluation of the neuroprotective
effect of taurine in Alzheimer’s disease using functional molecular imaging. Sci. Rep. 2020,10, 15551. [CrossRef] [PubMed]
230.
Santa-Maria, I.; Hernandez, F.; Moreno, F.J.; Avila, J. Taurine, an inducer for tau polymerization and a weak inhibitor for
amyloid-beta-peptide aggregation. Neurosci. Lett. 2007,429, 91–94. [CrossRef]
231.
Avshalumov, M.V.; Rice, M.E. NMDA receptor activation mediates hydrogen peroxide-induced pathophysiology in rat hippocam-
pal slices. J. Neurophysiol. 2002,87, 2896–2903. [CrossRef] [PubMed]
232.
Carvajal, F.J.; Mattison, H.A.; Cerpa, W. Role of NMDA Receptor-Mediated Glutamatergic Signaling in Chronic and Acute
Neuropathologies. Neural Plast. 2016,2016, 2701526. [CrossRef]
233.
Esteras, N.; Kopach, O.; Maiolino, M.; Lariccia, V.; Amoroso, S.; Qamar, S.; Wray, S.; Rusakov, D.A.; Jaganjac, M.; Abramov, A.Y.
Mitochondrial ROS control neuronal excitability and cell fate in frontotemporal dementia. Alzheimers Dement. 2021. [CrossRef]
Molecules 2021,26, 4913 21 of 21
234.
Rossi, A.; Pizzo, P.; Filadi, R. Calcium, mitochondria and cell metabolism: A functional triangle in bioenergetics. Biochim. Biophys.
Acta Mol. Cell Res. 2019,1866, 1068–1078. [CrossRef] [PubMed]
235.
Rossi, A.; Rigotto, G.; Valente, G.; Giorgio, V.; Basso, E.; Filadi, R.; Pizzo, P. Defective Mitochondrial Pyruvate Flux Affects Cell
Bioenergetics in Alzheimer’s Disease-Related Models. Cell Rep. 2020,30, 2332–2348.e10. [CrossRef]
236.
Wang, J.; Wang, F.; Mai, D.; Qu, S. Molecular Mechanisms of Glutamate Toxicity in Parkinson’s Disease. Front. Neurosci.
2020
,14,
585584. [CrossRef] [PubMed]
237.
Johri, A.; Beal, M.F. Mitochondrial dysfunction in neurodegenerative diseases. J. Pharmacol. Exp. Ther.
2012
,342, 619–630.
[CrossRef]
238.
Wu, Y.; Chen, M.; Jiang, J. Mitochondrial dysfunction in neurodegenerative diseases and drug targets via apoptotic signaling.
Mitochondrion 2019,49, 35–45. [CrossRef]
239.
Erickson, C.A.; Early, M.; Stigler, K.A.; Wink, L.K.; Mullett, J.E.; McDougle, C.J. An open-label naturalistic pilot study of
acamprosate in youth with autistic disorder. J. Child. Adolesc. Psychopharmacol. 2011,21, 565–569. [CrossRef]
240.
Erickson, C.A.; Mullett, J.E.; McDougle, C.J. Brief report: Acamprosate in fragile X syndrome. J. Autism Dev. Disord.
2010
,40,
1412–1416. [CrossRef]
241.
Erickson, C.A.; Ray, B.; Maloney, B.; Wink, L.K.; Bowers, K.; Schaefer, T.L.; McDougle, C.J.; Sokol, D.K.; Lahiri, D.K. Impact of
acamprosate on plasma amyloid-beta precursor protein in youth: A pilot analysis in fragile X syndrome-associated and idiopathic
autism spectrum disorder suggests a pharmacodynamic protein marker. J. Psychiatr. Res.
2014
,59, 220–228. [CrossRef] [PubMed]
242.
Erickson, C.A.; Wink, L.K.; Early, M.C.; Stiegelmeyer, E.; Mathieu-Frasier, L.; Patrick, V.; McDougle, C.J. Brief report: Pilot
single-blind placebo lead-in study of acamprosate in youth with autistic disorder. J. Autism Dev. Disord.
2014
,44, 981–987.
[CrossRef] [PubMed]
243.
Erickson, C.A.; Wink, L.K.; Ray, B.; Early, M.C.; Stiegelmeyer, E.; Mathieu-Frasier, L.; Patrick, V.; Lahiri, D.K.; McDougle, C.J.
Impact of acamprosate on behavior and brain-derived neurotrophic factor: An open-label study in youth with fragile X syndrome.
Psychopharmacology 2013,228, 75–84. [CrossRef] [PubMed]
244.
Wright, T.M.; Myrick, H. Acamprosate: A new tool in the battle against alcohol dependence. Neuropsychiatr. Dis. Treat.
2006
,2,
445–453. [CrossRef]
245.
McDougle, C.J.; Erickson, C.A.; Stigler, K.A.; Posey, D.J. Neurochemistry in the pathophysiology of autism. J. Clin. Psychiatry
2005,66 (Suppl. 10), 9–18.
246.
Silverman, J.L.; Tolu, S.S.; Barkan, C.L.; Crawley, J.N. Repetitive self-grooming behavior in the BTBR mouse model of autism is
blocked by the mGluR5 antagonist MPEP. Neuropsychopharmacology 2010,35, 976–989. [CrossRef] [PubMed]
247.
Yizhar, O.; Fenno, L.E.; Prigge, M.; Schneider, F.; Davidson, T.J.; O’Shea, D.J.; Sohal, V.S.; Goshen, I.; Finkelstein, J.; Paz, J.T.; et al.
Neocortical excitation/inhibition balance in information processing and social dysfunction. Nature
2011
,477, 171–178. [CrossRef]
248.
Filipek, P.A.; Juranek, J.; Smith, M.; Mays, L.Z.; Ramos, E.R.; Bocian, M.; Masser-Frye, D.; Laulhere, T.M.; Modahl, C.; Spence,
M.A.; et al. Mitochondrial dysfunction in autistic patients with 15q inverted duplication. Ann. Neurol.
2003
,53, 801–804.
[CrossRef]
249.
Giulivi, C.; Zhang, Y.F.; Omanska-Klusek, A.; Ross-Inta, C.; Wong, S.; Hertz-Picciotto, I.; Tassone, F.; Pessah, I.N. Mitochondrial
dysfunction in autism. JAMA 2010,304, 2389–2396. [CrossRef]
250.
Oliveira, G.; Diogo, L.; Grazina, M.; Garcia, P.; Ataide, A.; Marques, C.; Miguel, T.; Borges, L.; Vicente, A.M.; Oliveira, C.R.
Mitochondrial dysfunction in autism spectrum disorders: A population-based study. Dev. Med. Child. Neurol.
2005
,47, 185–189.
[CrossRef]
... Taurine has very important role in neonatal development, therefore mothers are encouraged to feed their babies as colostrum is the best source of taurine, or should feed their babies with taurine-supplemented formulas and taurine-supplemented total parenteral nutrition [44]. Taurine synthesizing enzymes are lower in concentration in cats, dogs and foxes, therefore they are primarily depend on taurine supplemented diets [44]. ...
... Taurine has very important role in neonatal development, therefore mothers are encouraged to feed their babies as colostrum is the best source of taurine, or should feed their babies with taurine-supplemented formulas and taurine-supplemented total parenteral nutrition [44]. Taurine synthesizing enzymes are lower in concentration in cats, dogs and foxes, therefore they are primarily depend on taurine supplemented diets [44]. These animals develop different pathologies like cardiomyopathy and myocardial dysfunction [45], retinal and tapetal degeneration that leads to blindness [46], neurological abnormalities [47], weakened immune response [48], gastrointestinal problems [49], pregnancy followed by foetal complications [50], when fed with taurine deficient diet. ...
... These animals were protected against these pathologies when fed with taurine supplemented diet as well as improved reproductive performance and neurological development [44], seizure [51], retinopathy [52] and cardiomyopathy [45]. Energy drinks such as Red Bull, Monster, Tab Energy and Rockstar are rich in taurine [44]. ...
Article
Full-text available
Taurine (Tau), a sulphur containing amino acid, chemically known as 2 aminoethane sulphonic acid, it's a non-proteinogenic β-amino acid, often referred to as semi essential amino acid as new born mammals have very limited ability to synthesize taurine and they have to depend on dietary sources, it is not incorporated into proteins as no aminoacyl tRNA synthetase has yet been identified and is not oxidized in mammalian cells, it attains an important place because of the antioxidant defence network. It has multiple function in the CNS, it serves as an osmoregulator, antioxidant, inhibitory neuromodulator, and regulator of intracellular Ca2flux.First time when it was discovered from ox bile by the German professors Friedrich Tiedemann and Leopold Gmelin they named it GallenAsparagin, later it was known as taurus, in latin Bos taurus means Ox, but it attains its current name (Taurine) in 1838 by von H. Demarcay. Because of presence of sulphonic acid instead of carboxylic acid it is not metabolized and not involved in gluconeogenesis and thus not envolve in direct energy sources. Taurine is produced by liver and kidney including retina, brain, heart and placenta. Taurine plays extensive role against different disorders of the body and in deadly diseases like cancer, liver cirrhosis etc. Human body contains about 0.1% of body weight as taurine. It has a number of physiological and pharmacological actions. In case of spinal cord injury elevated level of taurine has been seen, In methyl prednisolone (MP), treatment in case of SCI, elevation in level of taurine is observed, this elevated level seems to be involved in protection and regeneration of tissues following injury. In this review we try to cover every possible role of taurine which may provide enough information for future research.
... Taurine, a sulfur-containing β-amino acid, plays a crucial role in mitochondrial function, notably by modifying the first anticodon nucleotide of mitochondrial tRNA Leu(UUR) , which is a critical component for decoding codons during protein synthesis. In patients with MELAS with the common 3243A>G mutation, this taurine modification is absent, thereby resulting in defective mitochondrial protein translation and impaired respiratory chain activity [68,69]. Clinical trials have demonstrated the efficacy of taurine supplementation in reducing the frequency and severity of SLEs in patients with MELAS. ...
Article
Full-text available
Mitochondrial encephalopathy, lactic acidosis, and stroke-like episodes (MELAS) syndrome is a complex mitochondrial disorder characterized by a wide range of systemic manifestations. Key clinical features include recurrent stroke-like episodes, seizures, lactic acidosis, muscle weakness, exercise intolerance, sensorineural hearing loss, diabetes, and progressive neurological decline. MELAS is most commonly associated with mutations in mitochondrial DNA, particularly the m.3243A>G mutation in the MT-TL1 gene, which encodes tRNALeu (CUR). These mutations impair mitochondrial protein synthesis, leading to defective oxidative phosphorylation and energy failure at the cellular level. The clinical presentation and severity vary widely among patients, but the syndrome often results in significant morbidity and reduced life expectancy because of progressive neurological deterioration. Current management is largely focused on conservative care, including anti-seizure medications, arginine or citrulline supplementation, high-dose taurine, and dietary therapies. However, these therapies do not address the underlying genetic mutations, leaving many patients with substantial disease burden. Emerging experimental treatments, such as gene therapy and mitochondrial replacement techniques, aim to correct the underlying genetic defects and offer potential curative strategies. Further research is essential to understand the pathophysiology of MELAS, optimize current therapies, and develop novel treatments that may significantly improve patient outcomes and extend survival.
... Mitochondria, as the primary sites of energy production within cells, play a crucial role in cellular respiration by efficiently neutralizing ROS generated during metabolic processes. By regulating redox balance, mitochondria prevent the excessive generation and accumulation of oxygen free radicals, thereby protecting cells from oxidative stress [34]. Furthermore, previous study has confirmed that DNJ influences cardiac function through binding to the OPA, a protein necessary for mitochondrial fusion [35]. ...
Article
Full-text available
The poultry industry struggles with oxidative stress affecting gut health and productivity. This study examined using 1-Deoxynojirimycin (DNJ) extracts from mulberry leaves as an antioxidant in broilers feed to combat this issue. We divided 240 broilers, aged 16 days, into six groups, including a control and groups exposed to oxidative stress through H2O2 injections, with different supplement levels of DNJ-E (40, 80, 120, and 160 mg/kg of the basal diet) lasting until the broilers reached 42 days old. We evaluated intestinal morphology, ultrastructure, oxidative stress markers, the tight junction, and inflammatory cytokines. Adding 40 mg/kg DNJ-E improved villus height, the villus-to-crypt ratio, and cellular ultrastructure, and increased SOD levels in the jejunum and ileum, as well as CAT levels in the duodenum and jejunum (p < 0.05), compared to the H2O2 group. The addition of DNJ had differential effects on oxidative stress, the intestinal barrier, and immune-related genes. Importantly, the dosages of 40 mg/kg and 80 mg/kg resulted in an upregulation of MUC2 mRNA expression (p < 0.05). These findings suggest that DNJ-E holds potential as a beneficial feed additive for enhancing broiler health, particularly at supplementation levels below 80 mg/kg, as higher concentrations may negatively influence intestinal health. Future investigations should aim to elucidate the underlying mechanisms through which DNJ-E operates within the avian gastrointestinal system.
... To compensate for the accumulating ROS, antioxidants are needed. Our study demonstrated acute alterations in metabolites associated with sulfur-containing amino acid metabolism, including cysteine and hypotaurine, which are directly associated with the synthesis of antioxidants [33]. ...
Article
Full-text available
Background Research on traumatic brain injury (TBI) highlights the significance of counteracting its metabolic impact via exogenous fuels to support metabolism and diminish cellular damage. While ethyl pyruvate (EP) treatment shows promise in normalizing cellular metabolism and providing neuroprotection, there is a gap in understanding the precise metabolic pathways involved. Metabolomic analysis of the acute post-injury metabolic effects, with and without EP treatment, aims to deepen our knowledge by identifying and comparing the metabolite profiles, thereby illuminating the injury's effects and EP's therapeutic potential. Methods In the current study, an untargeted metabolomics approach was used to reveal brain metabolism changes in rats 24 h after a controlled cortical impact (CCI) injury, with or without EP treatment. Using principal component analysis (PCA), volcano plots, Random Forest and pathway analysis we differentiated the brain metabolomes of CCI and sham injured animals treated with saline (Veh) or EP, identifying key metabolites and pathways affected by injury. Additionally, the effect of EP on the non-injured brain was also explored. Results PCA showed a clear separation of the four study groups (sham-Veh, CCI-Veh, sham-EP, CCI-EP) based on injury. Following CCI injury (CCI-Veh), 109 metabolites belonging to the amino acid, carbohydrate, lipid, nucleotide, and xenobiotic families exhibited a twofold change at 24 h compared to the sham-Veh group, with 93 of these significantly increasing and 16 significantly decreasing (p < 0.05). CCI animals were treated with EP (CCI-EP) showed only 5 metabolites in the carbohydrate, amino acids, peptides, nucleotides, lipids, and xenobiotics super families that exhibited a twofold change, compared to the CCI-Veh group (p < 0.05). In the non-injured brain, EP treatment (sham-EP) resulted in a twofold change in 6 metabolites within the amino acid, peptide, nucleotide, and lipid super families compared to saline treated sham animals (sham-Veh, p < 0.05). Conclusions This study delineates the unique metabolic signatures resulting from a CCI injury and those related to EP treatment in both the injured and non-injured brain, underscoring the metabolic adaptations to brain injury and the effects of EP. Our analysis uncovers significant shifts in metabolites associated with inflammation, energy metabolism, and neuroprotection after injury, and demonstrates how EP intervention after injury alters metabolites associated with mitigating inflammation and oxidative damage.
... Taurine (C 2 H 7 NO 3 S) is an amino acid isolated from Calculus bovis (Niuhuang) and is also found in marine plants such as Porphyra dentata (Aung et al. 2022). Taurine can resist OS and maintain mitochondrial function Jong et al. 2021). Studies have shown that taurine deficiency may be closely related to increased DNA and mitochondrial function damage; thus, taurine deficiency may be the driving factor of ageing (Singh et al. 2023). ...
Article
Full-text available
Context Pancreatic ductal adenocarcinoma (PDAC), which is characterized by its malignant nature, presents challenges for early detection and is associated with a poor prognosis. Any strategy that can interfere with the beginning or earlier stage of PDAC greatly delays disease progression. In response to this intractable problem, the exploration of new drugs is critical to reduce the incidence of PDAC. Objective In this study, we summarize the mechanisms of pancreatitis-induced PDAC and traditional Chinese medicine (TCM) theory and review the roles and mechanisms of botanical drugs and their natural compounds that can inhibit the process of pancreatitis-induced PDAC. Methods With the keywords ‘chronic pancreatitis’, ‘TCM’, ‘Chinese medicinal formulae’, ‘natural compounds’, ‘PDAC’ and ‘pancreatic cancer’, we conducted an extensive literature search of the PubMed, Web of Science, and other databases to identify studies that effectively prevent PDAC in complex inflammatory microenvironments. Results We summarized the mechanism of pancreatitis-induced PDAC. Persistent inflammatory microenvironments cause multiple changes in the pancreas itself, including tissue damage, abnormal cell differentiation, and even gene mutation. According to TCM, pancreatitis-induced PDAC is the process of ‘dampness-heat obstructing the spleen and deficiency due to stagnation’ induced by a variety of pathological factors. A variety of botanical drugs and their natural compounds, such as Chaihu classical formulae, flavonoids, phenolics, terpenoids, etc., may be potential drugs to interfere with the development of PDAC via reshaping the inflammatory microenvironment by improving tissue injury and pancreatic fibrosis. Conclusions Botanical drugs and their natural compounds show great potential for preventing PDAC in complex inflammatory microenvironments.
... The copyright holder for this preprint this version posted October 26, 2024. ; https://doi.org/10.1101/2024.10.23.619827 doi: bioRxiv preprint activation and tumor suppression (66)(67)(68)(69)(70)(71)(72). ...
Preprint
Full-text available
Arginine metabolism in tumors is often shunted into the pathway producing pro-tumor and immune suppressive polyamines (PAs), while downmodulating the alternative nitric oxide (NO) synthesis pathway. Aiming to correct arginine metabolism in tumors, arginine deprivation therapy and inhibitors of PA synthesis have been developed. Despite some therapeutic advantages, these approaches have often yielded severe side effects, making it necessary to explore an alternative strategy. We previously reported that supplementing SEP, the endogenous precursor of BH4 (the essential NO synthase cofactor), could correct arginine metabolism in tumor cells and tumor-associated macrophages (TAMs) and induce their metabolic and phenotypic reprogramming. We saw that oral SEP treatment effectively suppressed the growth of HER2-positive mammary tumors in animals. SEP also has no reported dose-dependent toxicity in clinical trials for metabolic disorders. In the present study, we report that a long-term use of SEP in animals susceptible to HER2-positive mammary tumors effectively prevented tumor occurrence. These SEP-treated animals had undergone reprogramming of the systemic metabolism and immunity, elevating total T cell counts in the circulation and bone marrow. Given that bone marrow-resident T cells are mostly memory T cells, it is plausible that chronic SEP treatment promoted memory T cell formation, leading to a potent tumor prevention. These findings suggest the possible roles of the SEP/BH4/NO axis in promoting memory T cell formation and its potential therapeutic utility for preventing HER2-positive breast cancer.
Article
This review focuses on the roles of the two primary sulfur-containing amino acids, cysteine and methionine, in regulating reactive oxygen/nitrogen species (RONS). RONS are highly reactive oxygen/nitrogen-containing free radicals and compounds. Endogenous and exogenous antioxidants, including sulfur-containing amino acids, protect cells against the harmful effects of RONS on cellular macromolecules. This study thoroughly reviews the mechanisms by which these two sulfur-containing amino acids neutralize RONS. Additionally, a bioinformatic analysis of the percentage compositions of cysteine and methionine in metabolic proteins of humans and 12 closely related species was conducted using a “Biopython” script to assess their potential role as sinks for RONS, maintaining the structure and function of metabolic proteins. A total of 119 proteins from various metabolic pathways, including glycolysis, pyruvate to acetyl CoA conversion, tricarboxylic acid cycle, oxidative phosphorylation, pentose phosphate pathway, gluconeogenesis, glycogen metabolism, fatty acid metabolism, amino acid catabolism, nucleotide biosynthesis, and ROS scavengers were included in the bioinformatics analysis. This review shows that methionine and cysteine play crucial roles in neutralizing RONS. The bioinformatic analysis revealed that the percentage compositions of methionine and cysteine are higher in key redox enzymes like dehydrogenases, enzymes involved in oxidative phosphorylation, and those participating in the committed steps of metabolic pathways.
Preprint
Full-text available
The vaginal microbiome's role in risk, progression, and treatment of female cancers has been widely explored. Yet, there remains a need to develop methods to understand the interaction of microbiome factors with host cells and to characterize their potential therapeutic functions. To address this challenge, we developed a systems biology framework we term the Pharmacobiome for microbiome pharmacology analysis. The Pharmacobiome framework evaluates similarities between microbes and microbial byproducts and known drugs based on their impact on host transcriptomic cellular signatures. Here, we apply our framework to characterization of the Anti-Gynecologic Cancer Vaginal Pharmacobiome. Using published vaginal microbiome multi-omics data from the Partners PrEP clinical trial, we constructed vaginal epithelial gene signatures associated with each profiled vaginal microbe and metabolite. We compared these microbiome-associated host gene signatures to post-drug perturbation host gene signatures associated with 35 FDA-approved anti-cancer drugs from the Library of Integrated Network-based Cellular Signatures database to identify vaginal microbes and metabolites with high statistical and functional similarity to these drugs. We found that Lactobacilli and their metabolites can regulate host gene expression in ways similar to many anti-cancer drugs. Additionally, we experimentally tested our model prediction that taurine, a metabolite produced by L. crispatus, kills cancerous breast and endometrial cancer cells. Our study shows that the Pharmacobiome is a powerful framework for characterizing the anti-cancer therapeutic potential of vaginal microbiome factors with generalizability to other cancers, microbiomes, and diseases.
Article
Full-text available
Introduction: The second most common form of early-onset dementia-frontotemporal dementia (FTD)-is often characterized by the aggregation of the microtubule-associated protein tau. Here we studied the mechanism of tau-induced neuronal dysfunction in neurons with the FTD-related 10+16 MAPT mutation. Methods: Live imaging, electrophysiology, and redox proteomics were used in 10+16 induced pluripotent stem cell-derived neurons and a model of tau spreading in primary cultures. Results: Overproduction of mitochondrial reactive oxygen species (ROS) in 10+16 neurons alters the trafficking of specific glutamate receptor subunits via redox regulation. Increased surface expression of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and N-methyl-D-aspartate (NMDA) receptors containing GluA1 and NR2B subunits leads to impaired glutamatergic signaling, calcium overload, and excitotoxicity. Mitochondrial antioxidants restore the altered response and prevent neuronal death. Importantly, extracellular 4R tau induces the same pathological response in healthy neurons, thus proposing a mechanism for disease propagation. Discussion: These results demonstrate mitochondrial ROS modulate glutamatergic signaling in FTD, and suggest a new therapeutic strategy.
Article
Full-text available
Background Taurine has become a popular supplement among athletes attempting to improve performance. While the effectiveness of taurine as an ergogenic aid remains controversial, this paper summarizes the current evidence regarding the efficacy of taurine in aerobic and anaerobic performance, metabolic stress, muscle soreness, and recovery. Methods Google Scholar, Web of Science, and MedLine (PubMed) searches were conducted through September 2020. Peer-reviewed studies that investigated taurine as a single ingredient at dosages of
Article
Full-text available
Mitochondria participate in various metabolic pathways, and their dysregulation results in multiple disorders, including aging-related diseases. However, the metabolic changes and mechanisms of mitochondrial disorders are not fully understood. Here, we found that induced pluripotent stem cells (iPSCs) from a patient with mitochondrial myopathy, encephalopathy, lactic acidosis, and stroke-like episodes (MELAS) showed attenuated proliferation and survival when glycolysis was inhibited. These deficits were rescued by taurine administration. Metabolomic analyses showed that the ratio of the reduced (GSH) to oxidized glutathione (GSSG) was decreased; whereas the levels of cysteine, a substrate of GSH, and oxidative stress markers were upregulated in MELAS iPSCs. Taurine normalized these changes, suggesting that MELAS iPSCs were affected by the oxidative stress and taurine reduced its influence. We also analyzed the retinal pigment epithelium (RPE) differentiated from MELAS iPSCs by using a three-dimensional culture system and found that it showed epithelial mesenchymal transition (EMT), which was suppressed by taurine. Therefore, mitochondrial dysfunction caused metabolic changes, accumulation of oxidative stress that depleted GSH, and EMT in the RPE that could be involved in retinal pathogenesis. Because all these phenomena were sensitive to taurine treatment, we conclude that administration of taurine may be a potential new therapeutic approach for mitochondria-related retinal diseases.
Article
Full-text available
Parkinson’s disease (PD) is a common neurodegenerative disease, the pathological features of which include the presence of Lewy bodies and the neurodegeneration of dopaminergic neurons in the substantia nigra pars compacta. However, until recently, research on the pathogenesis and treatment of PD have progressed slowly. Glutamate and dopamine are both important central neurotransmitters in mammals. A lack of enzymatic decomposition of extracellular glutamate results in glutamate accumulating at synapses, which is mainly absorbed by excitatory amino acid transporters (EAATs). Glutamate exerts its physiological effects by binding to and activating ligand-gated ion channels [ionotropic glutamate receptors (iGluRs)] and a class of G-protein-coupled receptors [metabotropic glutamate receptors (mGluRs)]. Timely clearance of glutamate from the synaptic cleft is necessary because high levels of extracellular glutamate overactivate glutamate receptors, resulting in excitotoxic effects in the central nervous system. Additionally, increased concentrations of extracellular glutamate inhibit cystine uptake, leading to glutathione depletion and oxidative glutamate toxicity. Studies have shown that oxidative glutamate toxicity in neurons lacking functional N-methyl-D-aspartate (NMDA) receptors may represent a component of the cellular death pathway induced by excitotoxicity. The association between inflammation and excitotoxicity (i.e., immunoexcitotoxicity) has received increased attention in recent years. Glial activation induces neuroinflammation and can stimulate excessive release of glutamate, which can induce excitotoxicity and, additionally, further exacerbate neuroinflammation. Glutamate, as an important central neurotransmitter, is closely related to the occurrence and development of PD. In this review, we discuss recent progress on elucidating glutamate as a relevant neurotransmitter in PD. Additionally, we summarize the relationship and commonality among glutamate excitotoxicity, oxidative toxicity, and immunoexcitotoxicity in order to posit a holistic view and molecular mechanism of glutamate toxicity in PD.
Article
Full-text available
Diabetic kidney disease (DKD) is the leading cause of chronic kidney disease and end-stage kidney disease. Renin–angiotensin system inhibitors such as losartan are the predominant therapeutic options in clinical practice to treat DKD. Therefore, it is necessary to identify DKD-related metabolic profiles that are affected by losartan. To investigate the change in metabolism associated with the development of DKD, we performed global and targeted metabolic profiling using 800 MHz nuclear magnetic resonance spectroscopy of urine samples from streptozotocin-induced diabetic mice (DM) with or without losartan administration. A principal component analysis plot showed that the metabolic pattern in the losartan-treated diabetic mice returned from that in the DM group toward that in the control mice (CM). We found that 33 urinary metabolites were significantly changed in DM compared with CM, and the levels of 16 metabolites among them, namely, glucose, mannose, myo-inositol, pyruvate, fumarate, 2-hydroxyglutarate, isobutyrate, glycine, threonine, dimethylglycine, methyldantoin, isoleucine, leucine, acetylcarnitine, 3-hydroxy-3-methylglutarate, and taurine, shifted closer to the control level in response to losartan treatment. Pathway analysis revealed that these metabolites were associated with branched-chain amino acid degradation; taurine and hypotaurine metabolism; glycine, serine, and threonine metabolism; the tricarboxylic acid cycle; and galactose metabolism. Our results demonstrate that metabolomic analysis is a useful tool for identifying the metabolic pathways related to the development of DKD affected by losartan administration and may contribute to the discovery of new therapeutic agents for DKD.
Article
Full-text available
Purpose To evaluate the effects of taurine supplementation associated or not with chronic exercise on body composition, mitochondrial function, and expression of genes related to mitochondrial activity and lipid oxidation in the subcutaneous white adipose tissue (scWAT) of obese women. Methods A randomized and double-blind trial was developed with 24 obese women (BMI 33.1 ± 2.9 kg/m², 32.9 ± 6.3 y) randomized into three groups: Taurine supplementation group (Tau, n = 8); Exercise group (Ex, n = 8); Taurine supplementation + exercise group (TauEx, n = 8). The intervention was composed of 3 g of taurine or placebo supplementation and exercise training for eight weeks. Anthropometry, body fat composition, indirect calorimetry, scWAT biopsy for mitochondrial respiration, and gene expression related to mitochondrial activity and lipid oxidation were assessed before and after the intervention. Results No changes were observed for the anthropometric characteristics. The Ex group presented an increased resting energy expenditure rate, and the TauEx and Ex groups presented increased lipid oxidation and a decreased respiratory quotient. Both trained groups (TauEx and Ex) demonstrated improved scWAT mitochondrial respiratory capacity. Regarding mitochondrial markers, no changes were observed for the Tau group. The TauEx group had higher expression of CIDEA, PGC1a, PRDM16, UCP1, and UCP2. The genes related to fat oxidation (ACO2 and ACOX1) were increased in the Tau and Ex groups, while only the TauEx group presented increased expression of CPT1, PPARa, PPARγ, LPL, ACO1, ACO2, HSL, ACOX1, and CD36 genes. Conclusion Taurine supplementation associated with exercise improved lipid metabolism through the modulation of genes related to mitochondrial activity and fatty acid oxidation, suggesting a browning effect in the scWAT of obese women.
Article
Full-text available
Cardiovascular disease is the leading global health concern and responsible for more deaths worldwide than any other type of disorder. Atherosclerosis is a chronic inflammatory disease in the arterial wall, which underpins several types of cardiovascular disease. It has emerged that a strong relationship ex